1
|
Zhang Y, Scholten D, Qiang W, Platanias LC, Gradishar WJ, Kelley SO, Liu H. Circulation tumor cell isolation and enrichment technologies. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2025; 392:119-149. [PMID: 40287218 DOI: 10.1016/bs.ircmb.2025.01.009] [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: 04/29/2025]
Abstract
During cancer metastasis, tumor cells migrate from the primary tumor site and spread to distant tissue or organs through the circulatory system of the body. While it is challenging to track metastatic tumor cells, circulating tumor cells (CTCs) via liquid biopsy provide a unique and important opportunity for longitudinal monitoring of residual cancer diseases and progression, showing great potential to facilitate precision medicine in cancer patients. The enumeration and characterization of CTCs represent prognostic and predictive biomarkers, which can be used to monitor the response to and efficacy of various therapies. Along with molecular and cellular features of CTCs, this data can inform the detection of early micro-metastases and assess progression of advanced disease in a more sensitive manner than traditional imaging modalities, serving as a complementary approach with added value. Nevertheless, comprehensive multiomic analyses of CTCs at inter-cellular (cluster), single-cell, and subcellular levels to elucidate relevant CTC cancer biology, tumor immune ecosystem biology, and clinical outcomes have yet to be achieved, demanding multidisciplinary collaboration to advance the field. Complementary to the published chapter on multiomic analyses and functional properties of CTCs, this chapter summarizes key methods and integrated strategies in CTC isolation, highlighting an accelerated evolution in high-throughput analysis of CTCs.
Collapse
Affiliation(s)
- Youbin Zhang
- Circulating Tumor Cell Core, Lurie Comprehensive Cancer Center, Northwestern University, Chicago, IL, United States; Hematology & Oncology Division, Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL, United States
| | - David Scholten
- Driskill Graduate Program (DGP) in Life Sciences, Northwestern University Feinberg School of Medicine, Chicago, IL, United States; Department of Pharmacology, Northwestern University Feinberg School of Medicine, Chicago, IL, United States
| | - Wenan Qiang
- Chemistry of Life Processes, Chicago, IL, United States
| | - Leonidas C Platanias
- Circulating Tumor Cell Core, Lurie Comprehensive Cancer Center, Northwestern University, Chicago, IL, United States; Hematology & Oncology Division, Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL, United States
| | - William J Gradishar
- Circulating Tumor Cell Core, Lurie Comprehensive Cancer Center, Northwestern University, Chicago, IL, United States; Hematology & Oncology Division, Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL, United States
| | - Shana O Kelley
- Chemistry of Life Processes, Chicago, IL, United States; Department of Chemistry, Northwestern University, Chicago, IL, United States; Department of Biomedical Engineering, Northwestern University, Chicago, IL, United States; Chan Zuckerberg Biohub Chicago, Chicago, IL, United States
| | - Huiping Liu
- Circulating Tumor Cell Core, Lurie Comprehensive Cancer Center, Northwestern University, Chicago, IL, United States; Hematology & Oncology Division, Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL, United States; Department of Pharmacology, Northwestern University Feinberg School of Medicine, Chicago, IL, United States; Chan Zuckerberg Biohub Chicago, Chicago, IL, United States.
| |
Collapse
|
2
|
Ho CR, Tsai HJ, Wang JR, Wang CT, Chiou CC, Cheng JC, Chiang SF, Tseng CP. Development of PowerMag System II for Isolation of Circulating Tumor Cells with Improved Purity. Biomedicines 2025; 13:431. [PMID: 40002844 PMCID: PMC11852382 DOI: 10.3390/biomedicines13020431] [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: 12/20/2024] [Revised: 01/30/2025] [Accepted: 02/08/2025] [Indexed: 02/27/2025] Open
Abstract
Background/Objectives: The PowerMag system (PM) is a platform for the isolation of circulating tumor cells (CTCs) by the depletion of CD45+-leukocytes. However, an EpCAM-CD45- cell population is present in large numbers in the cell filtrates collected by PM. This lowers the purity of the CTCs and negatively impacts their molecular characterization. The aims of this study are to characterize the cellular properties of the EpCAM-CD45- cells and to upgrade the system to improve CTC purity. Methods: A real-time RT-PCR assay, Liu's stain analysis, and Annexin V (AnxV) binding assay were used to define the cellular properties of the EpCAM-CD45- cells. An upgraded system was developed to remove the EpCAM-CD45- cells and improve the CTC purity. Clinical blood samples were used to evaluate the performance of the system. Results: The EpCAM-CD45- cells were defined as apoptotic cells, which displayed apoptotic body-like morphology and elicited AnxV binding activity. AnxV beads developed in-house can effectively bind and remove EpCAM-CD45- cells from the cell filtrates. An improved generation of a CTCs isolation platform, designated as PM II, was developed by integration of AnxV beads into the workflow to remove the apoptotic cells. PM II recovered CTCs with improved CTC purity by effective removal of the background apoptotic cells. The improved performance of PM II allowed for direct profiling of cancer-related gene mutations by next-generation sequencing without cell picking and further purification. Conclusions: PM II holds great promise as a platform for isolating CTCs with improved purity and for exploring its application in cancer diagnosis and monitoring in a clinical setting.
Collapse
Affiliation(s)
- Cheng-Rou Ho
- Department of Medical Biotechnology and Laboratory Science, College of Medicine, Chang Gung University, Taoyuan 333, Taiwan
| | - Hui-Ju Tsai
- Department of Medical Biotechnology and Laboratory Science, College of Medicine, Chang Gung University, Taoyuan 333, Taiwan
| | - Jin-Ru Wang
- Department of Medical Biotechnology and Laboratory Science, College of Medicine, Chang Gung University, Taoyuan 333, Taiwan
| | - Chia-Te Wang
- International Master Degree Program for Molecular Medicine in Emerging Viral Infections, College of Medicine, Chang Gung University, Taoyuan 333, Taiwan
| | - Chiuan-Chian Chiou
- Department of Medical Biotechnology and Laboratory Science, College of Medicine, Chang Gung University, Taoyuan 333, Taiwan
- Master and PhD Program in Biotechnology Industry, College of Medicine, Chang Gung University, Taoyuan 333, Taiwan
- Department of Thoracic Medicine, Chang Gung Memorial Hospital, Linkou Branch, Taoyuan 333, Taiwan
| | - Ju-Chien Cheng
- Department of Medical Laboratory Science and Biotechnology, China Medical University, Taichung 404, Taiwan
| | - Sum-Fu Chiang
- Division of Colon and Rectal Surgery, Chang Gung Memorial Hospital, Linkou Branch, Taoyuan 333, Taiwan
- School of Traditional Chinese Medicine, Chang Gung University, Taoyuan 333, Taiwan
| | - Ching-Ping Tseng
- Department of Medical Biotechnology and Laboratory Science, College of Medicine, Chang Gung University, Taoyuan 333, Taiwan
- Graduate Institute of Biomedical Science, College of Medicine, Chang Gung University, Taoyuan 333, Taiwan
- Department of Laboratory Medicine, Chang Gung Memorial Hospital, Linkou Branch, Taoyuan 333, Taiwan
| |
Collapse
|
3
|
Liu C, Cai Y, Mou S. Liquid biopsy in lung cancer: The role of circulating tumor cells in diagnosis, treatment, and prognosis. Biomed Pharmacother 2024; 181:117726. [PMID: 39612860 DOI: 10.1016/j.biopha.2024.117726] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/01/2024] Open
Abstract
Despite numerous therapeutic advancements, such as immune checkpoint inhibitors, lung cancer continues to be the leading cause of cancer-related mortality. Therefore, the identification of cancer at an early stage is becoming a significant subject in contemporary oncology. Despite significant advancements in early detection tactics in recent decades, they continue to provide challenges because of the inconspicuous symptoms observed during the early stages of the primary tumor. Presently, tumor biomarkers and imaging techniques are extensively employed across different forms of cancer. Nevertheless, every approach has its own set of constraints. In certain instances, the detriments outweigh the advantages. Hence, there is an urgent need to enhance early detection methods. Currently, liquid biopsy is considered more flexible and not intrusive method in comparison to conventional test for early detection. Circulating tumor cells (CTCs) are crucial components of liquid biopsy and have a pivotal function in the spread and formation of secondary tumors. These indicators show great promise in the early identification of cancer. This study presents a comprehensive examination of the methodologies employed for the isolation and enrichment of circulating tumor cells (CTCs) in lung cancer. Additionally, it explores the formation of clusters of CTCs, which have a pivotal function in facilitating the effective dissemination of cancer to distant organs. In addition, we discuss the importance of CTCs in the detection, treatment, and prognosis of lung cancer.
Collapse
Affiliation(s)
- Chibo Liu
- Department of Clinical Laboratory, Taizhou Municipal Hospital, Taizhou, Zhejiang, China.
| | - Yanqun Cai
- Department of Clinical Laboratory, Taizhou Municipal Hospital, Taizhou, Zhejiang, China
| | - Sihua Mou
- Department of Clinical Laboratory, Taizhou Municipal Hospital, Taizhou, Zhejiang, China.
| |
Collapse
|
4
|
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
|
5
|
Chauhan A, Pal A, Sachdeva M, Boora GS, Parsana M, Bakshi J, Verma RK, Srinivasan R, Chatterjee D, Maitra A, Ghoshal S. A FACS-based novel isolation technique identifies heterogeneous CTCs in oral squamous cell carcinoma. Front Oncol 2024; 14:1269211. [PMID: 38469233 PMCID: PMC10925612 DOI: 10.3389/fonc.2024.1269211] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2023] [Accepted: 02/02/2024] [Indexed: 03/13/2024] Open
Abstract
Purpose Isolating circulating tumour cells (CTCs) from the blood is challenging due to their low abundance and heterogeneity. Limitations of conventional CTC detection methods highlight the need for improved strategies to detect and isolate CTCs. Currently, the Food and Drug Administration (FDA)-approved CellSearch™ and other RUO techniques are not available in India. Therefore, we wanted to develop a flexible CTC detection/isolation technique that addresses the limitation(s) of currently available techniques and is suitable for various downstream applications. Methods We developed a novel, efficient, user-friendly CTC isolation strategy combining density gradient centrifugation and immuno-magnetic hematogenous cell depletion with fluorescence-activated cell sorting (FACS)-based positive selection using multiple CTC-specific cell-surface markers. For FACS, a stringent gating strategy was optimised to exclude debris and doublets by side scatter/forward scatter (SSC/FSC) discriminator, remove dead cells by 4',6-diamidino-2-phenylindole (DAPI) staining, and eliminate non-specific fluorescence using a "dump" channel. APC-labelled anti-CD45mAB was used to gate remaining hematogenous cells, while multiple epithelial markers (EpCAM, EGFR, and Pan-Cytokeratin) and an epithelial-mesenchymal transition (EMT) marker (Vimentin) labelled with fluorescein isothiocyanate (FITC) were used to sort cancer cells. The technique was initially developed by spiking Cal 27 cancer cells into the blood of healthy donors and then validated in 95 biopsy-proven oral squamous cell carcinoma (OSCC) patients. CTCs isolated from patients were reconfirmed by Giemsa staining, immuno-staining, and whole transcriptome amplification (WTA), followed by qRT-PCR. In vitro culture and RNA sequencing (RNA-Seq) were also performed to confirm their suitability for various downstream applications. Results The mean detection efficiency for the Cal 27 tongue cancer cells spiked in the whole blood of healthy donors was 32.82% ± 12.71%. While ~75% of our patients (71/95) had detectable CTCs, the CTC positivity was independent of the TNM staging. The isolated potential cancer cells from OSCC patients were heterogeneous in size. They expressed different CTC-specific markers in various combinations as identified by qRT-PCR after WTA in different patients. Isolated CTCs were also found to be suitable for downstream applications like short-term CTC culture and RNA-Seq. Conclusion We developed a sensitive, specific, flexible, and affordable CTC detection/isolation technique, which is scalable to larger patient cohorts, provides a snapshot of CTC heterogeneity, isolates live CTCs ready for downstream molecular analysis, and, most importantly, is suitable for developing countries.
Collapse
Affiliation(s)
- Anshika Chauhan
- Department of Biochemistry, Post Graduate Institute of Medical Education and Research, Chandigarh, India
| | - Arnab Pal
- Department of Biochemistry, Post Graduate Institute of Medical Education and Research, Chandigarh, India
| | - Meenakshi Sachdeva
- Department of Regenerative Medicine, Post Graduate Institute of Medical Education and Research, Chandigarh, India
| | - Geeta S. Boora
- Department of Biochemistry, Post Graduate Institute of Medical Education and Research, Chandigarh, India
| | - Monil Parsana
- Department of Otolaryngology, Post Graduate Institute of Medical Education and Research, Chandigarh, India
| | - Jaimanti Bakshi
- Department of Otolaryngology, Post Graduate Institute of Medical Education and Research, Chandigarh, India
| | - Roshan Kumar Verma
- Department of Otolaryngology, Post Graduate Institute of Medical Education and Research, Chandigarh, India
| | - Radhika Srinivasan
- Department of Cytology and Gynecological Pathology, Post Graduate Institute of Medical Education and Research, Chandigarh, India
| | - Debajyoti Chatterjee
- Department of Histopathology, Post Graduate Institute of Medical Education and Research, Chandigarh, India
| | - Arindam Maitra
- National Institute of Biomedical Genomics, Kalyani, West Bengal, India
| | - Sushmita Ghoshal
- Department of Radiotherapy, Post Graduate Institute of Medical Education and Research, Chandigarh, India
| |
Collapse
|
6
|
Huang Z, Fu Y, Yang H, Zhou Y, Shi M, Li Q, Liu W, Liang J, Zhu L, Qin S, Hong H, Liu Y. Liquid biopsy in T-cell lymphoma: biomarker detection techniques and clinical application. Mol Cancer 2024; 23:36. [PMID: 38365716 PMCID: PMC10874034 DOI: 10.1186/s12943-024-01947-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2023] [Accepted: 01/25/2024] [Indexed: 02/18/2024] Open
Abstract
T-cell lymphoma is a highly invasive tumor with significant heterogeneity. Invasive tissue biopsy is the gold standard for acquiring molecular data and categorizing lymphoma patients into genetic subtypes. However, surgical intervention is unfeasible for patients who are critically ill, have unresectable tumors, or demonstrate low compliance, making tissue biopsies inaccessible to these patients. A critical need for a minimally invasive approach in T-cell lymphoma is evident, particularly in the areas of early diagnosis, prognostic monitoring, treatment response, and drug resistance. Therefore, the clinical application of liquid biopsy techniques has gained significant attention in T-cell lymphoma. Moreover, liquid biopsy requires fewer samples, exhibits good reproducibility, and enables real-time monitoring at molecular levels, thereby facilitating personalized health care. In this review, we provide a comprehensive overview of the current liquid biopsy biomarkers used for T-cell lymphoma, focusing on circulating cell-free DNA (cfDNA), circulating tumor DNA (ctDNA), circulating tumor cells (CTCs), Epstein-Barr virus (EBV) DNA, antibodies, and cytokines. Additionally, we discuss their clinical application, detection methodologies, ongoing clinical trials, and the challenges faced in the field of liquid biopsy.
Collapse
Affiliation(s)
- Zongyao Huang
- Department of Pathology, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, Affiliated Cancer Hospital of University of Electronic Science and Technology of China, Chengdu, China
| | - Yao Fu
- Department of Pathology, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, Affiliated Cancer Hospital of University of Electronic Science and Technology of China, Chengdu, China
| | - Hong Yang
- Department of Pathology, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, Affiliated Cancer Hospital of University of Electronic Science and Technology of China, Chengdu, China
| | - Yehan Zhou
- Department of Pathology, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, Affiliated Cancer Hospital of University of Electronic Science and Technology of China, Chengdu, China
| | - Min Shi
- Department of Pathology, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, Affiliated Cancer Hospital of University of Electronic Science and Technology of China, Chengdu, China
| | - Qingyun Li
- Genecast Biotechnology Co., Ltd, Wuxi, 214104, China
| | - Weiping Liu
- Department of Pathology, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China
| | - Junheng Liang
- Nanjing Geneseeq Technology Inc., Nanjing, 210032, Jiangsu, China
| | - Liuqing Zhu
- Nanjing Geneseeq Technology Inc., Nanjing, 210032, Jiangsu, China
| | - Sheng Qin
- Department of Pathology, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, Affiliated Cancer Hospital of University of Electronic Science and Technology of China, Chengdu, China.
| | - Huangming Hong
- Department of Medical Oncology, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, Affiliated Cancer Hospital of University of Electronic Science and Technology of China, Chengdu, China.
| | - Yang Liu
- Department of Pathology, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, Affiliated Cancer Hospital of University of Electronic Science and Technology of China, Chengdu, China.
| |
Collapse
|
7
|
Jiang Z, He J, Zhang B, Wang L, Long C, Zhao B, Yang Y, Du L, Luo W, Hu J, Hong X. A Potential "Anti-Warburg Effect" in Circulating Tumor Cell-mediated Metastatic Progression? Aging Dis 2024; 16:AD.2023.1227. [PMID: 38300633 PMCID: PMC11745448 DOI: 10.14336/ad.2023.1227] [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: 09/16/2023] [Accepted: 12/27/2023] [Indexed: 02/02/2024] Open
Abstract
Metabolic reprogramming is a defining hallmark of cancer metastasis, warranting thorough exploration. The tumor-promoting function of the "Warburg Effect", marked by escalated glycolysis and restrained mitochondrial activity, is widely acknowledged. Yet, the functional significance of mitochondria-mediated oxidative phosphorylation (OXPHOS) during metastasis remains controversial. Circulating tumor cells (CTCs) are considered metastatic precursors that detach from primary or secondary sites and harbor the potential to seed distant metastases through hematogenous dissemination. A comprehensive metabolic characterization of CTCs faces formidable obstacles, including the isolation of these rare cells from billions of blood cells, coupled with the complexities of ex vivo-culturing of CTC lines or the establishment of CTC-derived xenograft models (CDX). This review summarized the role of the "Warburg Effect" in both tumorigenesis and CTC-mediated metastasis. Intriguingly, bioinformatic analysis of single-CTC transcriptomic studies unveils a potential OXPHOS dominance over Glycolysis signature genes across several important cancer types. From these observations, we postulate a potential "Anti-Warburg Effect" (AWE) in CTCs-a metabolic shift bridging primary tumors and metastases. The observed AWE could be clinically important as they are significantly correlated with therapeutic response in melanoma and prostate patients. Thus, unraveling dynamic metabolic regulations within CTC populations might reveal an additional layer of regulatory complexities of cancer metastasis, providing an avenue for innovative anti-metastasis therapies.
Collapse
Affiliation(s)
- Zhuofeng Jiang
- Department of Biochemistry, School of Medicine, Southern University of Science and Technology, Shenzhen, Guangdong, China.
| | - Jiapeng He
- Department of Biochemistry, School of Medicine, Southern University of Science and Technology, Shenzhen, Guangdong, China.
| | - Binyu Zhang
- Department of Biochemistry, School of Medicine, Southern University of Science and Technology, Shenzhen, Guangdong, China.
| | - Liping Wang
- Department of Oncology, Southern University of Science and Technology Hospital, Shenzhen, Guangdong, China.
| | - Chunhao Long
- Department of Biochemistry, School of Medicine, Southern University of Science and Technology, Shenzhen, Guangdong, China.
| | - Boxi Zhao
- Department of Biochemistry, School of Medicine, Southern University of Science and Technology, Shenzhen, Guangdong, China.
| | - Yufan Yang
- Department of Biochemistry, School of Medicine, Southern University of Science and Technology, Shenzhen, Guangdong, China.
| | - Longxiang Du
- Department of Biochemistry, School of Medicine, Southern University of Science and Technology, Shenzhen, Guangdong, China.
| | - Weiren Luo
- Cancer Research Institute, The Second Affiliated Hospital of Southern University of Science and Technology, Shenzhen Third People's Hospital, National Clinical Research Center for Infectious Diseases, Shenzhen, China.
| | - Jianyang Hu
- Department of Biochemistry, School of Medicine, Southern University of Science and Technology, Shenzhen, Guangdong, China.
- Key University Laboratory of Metabolism and Health of Guangdong, Southern University of Science and Technology, Shenzhen, Guangdong, China.
| | - Xin Hong
- Department of Biochemistry, School of Medicine, Southern University of Science and Technology, Shenzhen, Guangdong, China.
- Key University Laboratory of Metabolism and Health of Guangdong, Southern University of Science and Technology, Shenzhen, Guangdong, China.
- Guangdong Provincial Key Laboratory of Cell Microenvironment and Disease Research, Southern University of Science and Technology, Shenzhen, Guangdong, China.
| |
Collapse
|
8
|
Shuai Y, Ma Z, Ju J, Wei T, Gao S, Kang Y, Yang Z, Wang X, Yue J, Yuan P. Liquid-based biomarkers in breast cancer: looking beyond the blood. J Transl Med 2023; 21:809. [PMID: 37957623 PMCID: PMC10644618 DOI: 10.1186/s12967-023-04660-z] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2023] [Accepted: 10/26/2023] [Indexed: 11/15/2023] Open
Abstract
In recent decades, using circulating tumor cell (CTC), circulating tumor DNA (ctDNA), circulating tumor RNA (ctRNA), exosomes and etc. as liquid biomarkers has received enormous attention in various tumors, including breast cancer (BC). To date, efforts in the area of liquid biopsy predominantly focus on the analysis of blood-based markers. It is worth noting that the identifications of markers from non-blood sources provide unique advantages beyond the blood and these alternative sources may be of great significance in offering supplementary information in certain settings. Here, we outline the latest advances in the analysis of non-blood biomarkers, predominantly including urine, saliva, cerebrospinal fluid, pleural fluid, stool and etc. The unique advantages of such testings, their current limitations and the appropriate use of non-blood assays and blood assays in different settings are further discussed. Finally, we propose to highlight the challenges of these alternative assays from basic to clinical implementation and explore the areas where more investigations are warranted to elucidate its potential utility.
Collapse
Affiliation(s)
- You Shuai
- Department of VIP Medical Services, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Zhonghua Ma
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Endoscopy, Peking University Cancer Hospital & Institute, Beijing, 100142, China
| | - Jie Ju
- Department of VIP Medical Services, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Tong Wei
- Department of VIP Medical Services, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Songlin Gao
- Department of VIP Medical Services, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Yikun Kang
- Department of VIP Medical Services, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Zixuan Yang
- Department of VIP Medical Services, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Xue Wang
- Department of VIP Medical Services, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Jian Yue
- Department of VIP Medical Services, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Peng Yuan
- Department of VIP Medical Services, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China.
| |
Collapse
|
9
|
Surappa S, Multani P, Parlatan U, Sinawang PD, Kaifi J, Akin D, Demirci U. Integrated "lab-on-a-chip" microfluidic systems for isolation, enrichment, and analysis of cancer biomarkers. LAB ON A CHIP 2023; 23:2942-2958. [PMID: 37314731 PMCID: PMC10834032 DOI: 10.1039/d2lc01076c] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
The liquid biopsy has garnered considerable attention as a complementary clinical tool for the early detection, molecular characterization and monitoring of cancer over the past decade. In contrast to traditional solid biopsy techniques, liquid biopsy offers a less invasive and safer alternative for routine cancer screening. Recent advances in microfluidic technologies have enabled handling of liquid biopsy-derived biomarkers with high sensitivity, throughput, and convenience. The integration of these multi-functional microfluidic technologies into a 'lab-on-a-chip' offers a powerful solution for processing and analyzing samples on a single platform, thereby reducing the complexity, bio-analyte loss and cross-contamination associated with multiple handling and transfer steps in more conventional benchtop workflows. This review critically addresses recent developments in integrated microfluidic technologies for cancer detection, highlighting isolation, enrichment, and analysis strategies for three important sub-types of cancer biomarkers: circulating tumor cells, circulating tumor DNA and exosomes. We first discuss the unique characteristics and advantages of the various lab-on-a-chip technologies developed to operate on each biomarker subtype. This is then followed by a discussion on the challenges and opportunities in the field of integrated systems for cancer detection. Ultimately, integrated microfluidic platforms form the core of a new class of point-of-care diagnostic tools by virtue of their ease-of-operation, portability and high sensitivity. Widespread availability of such tools could potentially result in more frequent and convenient screening for early signs of cancer at clinical labs or primary care offices.
Collapse
Affiliation(s)
- Sushruta Surappa
- Canary Center at Stanford for Cancer Early Detection, Bio-Acoustic MEMS in Medicine (BAMM) Lab, Department of Radiology, School of Medicine, Stanford University, Palo Alto, CA 94304, USA.
| | - Priyanka Multani
- Canary Center at Stanford for Cancer Early Detection, Bio-Acoustic MEMS in Medicine (BAMM) Lab, Department of Radiology, School of Medicine, Stanford University, Palo Alto, CA 94304, USA.
| | - Ugur Parlatan
- Canary Center at Stanford for Cancer Early Detection, Bio-Acoustic MEMS in Medicine (BAMM) Lab, Department of Radiology, School of Medicine, Stanford University, Palo Alto, CA 94304, USA.
| | - Prima Dewi Sinawang
- Canary Center at Stanford for Cancer Early Detection, Bio-Acoustic MEMS in Medicine (BAMM) Lab, Department of Radiology, School of Medicine, Stanford University, Palo Alto, CA 94304, USA.
- Department of Chemical Engineering, Stanford University, Stanford, CA 94305, USA
| | - Jussuf Kaifi
- Department of Surgery, School of Medicine, University of Missouri, Columbia, MO 65212, USA
- Harry S. Truman Memorial Veterans' Hospital, Columbia, MO 65201, USA
| | - Demir Akin
- Canary Center at Stanford for Cancer Early Detection, Bio-Acoustic MEMS in Medicine (BAMM) Lab, Department of Radiology, School of Medicine, Stanford University, Palo Alto, CA 94304, USA.
- Center for Cancer Nanotechnology Excellence for Translational Diagnostics (CCNE-TD), School of Medicine, Stanford University, Stanford, CA 94305, USA
| | - Utkan Demirci
- Canary Center at Stanford for Cancer Early Detection, Bio-Acoustic MEMS in Medicine (BAMM) Lab, Department of Radiology, School of Medicine, Stanford University, Palo Alto, CA 94304, USA.
| |
Collapse
|
10
|
Jiang B, Xie D, Wang S, Li X, Wu G. Advances in early detection methods for solid tumors. Front Genet 2023; 14:1091223. [PMID: 36911396 PMCID: PMC9998680 DOI: 10.3389/fgene.2023.1091223] [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: 11/06/2022] [Accepted: 02/13/2023] [Indexed: 02/26/2023] Open
Abstract
During the last decade, non-invasive methods such as liquid biopsy have slowly replaced traditional imaging and invasive pathological methods used to diagnose and monitor cancer. Improvements in the available detection methods have enabled the early screening and diagnosis of solid tumors. In addition, advances in early detection methods have made the continuous monitoring of tumor progression using repeat sampling possible. Previously, the focus of liquid biopsy techniques included the following: 1) the isolation of circulating tumor cells, circulating tumor DNA, and extracellular tumor vesicles from solid tumor cells in the patient's blood; in addition to 2) analyzing genomic and proteomic data contained within the isolates. Recently, there has been a rapid devolvement in the techniques used to isolate and analyze molecular markers. This rapid evolvement in detection techniques improves their accuracy, especially when few samples are available. In addition, there is a tremendous expansion in the acquisition of samples and targets for testing; solid tumors can be detected from blood and other body fluids. Test objects have also expanded from samples taken directly from cancer to include indirect objects affected in cancer development. Liquid biopsy technology has limitations. Even so, this detection technique is the key to a new phase of oncogenetics. This review aims to provide an overview of the current advances in liquid biopsy marker selection, isolation, and detection methods for solid tumors. The advantages and disadvantages of liquid biopsy technology will also be explored.
Collapse
Affiliation(s)
| | | | | | - Xiunan Li
- Department of Urology, The First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Guangzhen Wu
- Department of Urology, The First Affiliated Hospital of Dalian Medical University, Dalian, China
| |
Collapse
|
11
|
Vasantharajan SS, Barnett E, Gray ES, Rodger EJ, Eccles MR, Pattison S, Munro F, Chatterjee A. Size-Based Method for Enrichment of Circulating Tumor Cells from Blood of Colorectal Cancer Patients. Methods Mol Biol 2023; 2588:231-248. [PMID: 36418692 DOI: 10.1007/978-1-0716-2780-8_15] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/16/2023]
Abstract
Circulating tumor cells (CTCs) are precursors of the metastatic cascade, which is responsible for 90% of all cancer-related deaths. CTCs arise from solid tumors and travel through the bloodstream and lymphatic system. Developments in the isolation and analysis of CTCs promise potential biomarker assays for detection and monitoring of cancer through a minimally invasive procedure. Despite this, the precise role of CTCs in metastasis remains poorly characterized, mainly due to the low density of CTCs (1-10 CTCs per 10 mL of blood) present in patient blood and the lack of robust methods for their isolation in a standard laboratory setting. CellSearch is currently the only FDA-approved CTC enrichment protocol, but limitations of this EpCAM-based method include cost, availability, and the use of a single surface marker for capture. To address these limitations, we have optimized an existing method, MetaCell, which exploits the differences in size of CTCs compared to other blood cells for CTC enrichment from blood. MetaCell contains a membrane with 8 μm pores, and blood is filtered through this kit by capillary action and CTCs, which are typically larger than the pores and remain on top of the membrane, while most leukocytes pass through the pores. The membrane along with these CTCs can be detached and transferred to 6-well plates for culturing or directly used for characterization. Here, we provide a detailed protocol for enrichment of CTCs using the filtration device MetaCell and a procedure for characterization of CTCs by immunohistochemical staining.
Collapse
Affiliation(s)
- Sai Shyam Vasantharajan
- Department of Pathology, Dunedin School of Medicine, University of Otago, Dunedin, New Zealand.
| | - Edward Barnett
- Department of Pathology, Dunedin School of Medicine, University of Otago, Dunedin, New Zealand
| | - Elin S Gray
- Centre for Precision Health, Edith Cowan University, Joondalup, Australia
| | - Euan J Rodger
- Department of Pathology, Dunedin School of Medicine, University of Otago, Dunedin, New Zealand
| | - Michael R Eccles
- Department of Pathology, Dunedin School of Medicine, University of Otago, Dunedin, New Zealand
| | - Sharon Pattison
- Department of Medicine, Dunedin School of Medicine, University of Otago, Dunedin, New Zealand
| | - Fran Munro
- Department of Surgical Sciences, Dunedin School of Medicine, University of Otago, Dunedin, New Zealand
| | - Aniruddha Chatterjee
- Department of Pathology, Dunedin School of Medicine, University of Otago, Dunedin, New Zealand.
- UPES University, School of Health Sciences, Dehradun, India.
| |
Collapse
|
12
|
Wu M, Huang Y, Zhou Y, Zhao H, Lan Y, Yu Z, Jia C, Cong H, Zhao J. The Discovery of Novel Circulating Cancer-Related Cells in Circulation Poses New Challenges to Microfluidic Devices for Enrichment and Detection. SMALL METHODS 2022; 6:e2200226. [PMID: 35595707 DOI: 10.1002/smtd.202200226] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/21/2022] [Revised: 04/19/2022] [Indexed: 06/15/2023]
Abstract
Circulating tumor cells (CTCs) enumeration has been widely used as a surrogate predictive marker for early diagnoses, the evaluation of chemotherapy efficacy, and cancer prognosis. Microfluidic technologies for CTCs enrichment and detection have been developed and commercialized as automation platforms. Currently, in addition to CTCs, some new types of circulating cancer-related cells (e.g., CCSCs, CTECs, CAMLs, and heterotypic CTC clusters) in circulation are also reported to be correlated to cancer diagnosis, metastasis, or prognosis. And they widely differ from the conventional CTCs in positive markers, cellular morphology, or size, which presents a new technological challenge to microfluidic devices that use affinity-based capture methods or size-based filtration methods for CTCs detection. This review focuses on the biological and physical properties as well as clinical significance of the novel circulating cancer-related cells, and discusses the challenges of their discovery to microfluidic chip for enrichment. Finally, the current challenges of CTCs detection in clinical application and future opportunities are also discussed.
Collapse
Affiliation(s)
- Man Wu
- State Key Laboratory of Transducer Technology, Shanghai Institute of Microsystem and Information Technology, Chinese Academy of Sciences, Shanghai, 200050, China
- Center of Materials Science and Optoelectronics Engineering, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Yuhang Huang
- Shanghai Normal University, Shanghai, 200030, China
| | - Yang Zhou
- State Key Laboratory of Transducer Technology, Shanghai Institute of Microsystem and Information Technology, Chinese Academy of Sciences, Shanghai, 200050, China
- Center of Materials Science and Optoelectronics Engineering, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Hui Zhao
- State Key Laboratory of Transducer Technology, Shanghai Institute of Microsystem and Information Technology, Chinese Academy of Sciences, Shanghai, 200050, China
- Center of Materials Science and Optoelectronics Engineering, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Yuwei Lan
- State Key Laboratory of Transducer Technology, Shanghai Institute of Microsystem and Information Technology, Chinese Academy of Sciences, Shanghai, 200050, China
- Center of Materials Science and Optoelectronics Engineering, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Zhibin Yu
- State Key Laboratory of Transducer Technology, Shanghai Institute of Microsystem and Information Technology, Chinese Academy of Sciences, Shanghai, 200050, China
- Center of Materials Science and Optoelectronics Engineering, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Chunping Jia
- State Key Laboratory of Transducer Technology, Shanghai Institute of Microsystem and Information Technology, Chinese Academy of Sciences, Shanghai, 200050, China
- Center of Materials Science and Optoelectronics Engineering, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Hui Cong
- Department of Laboratory Medicine, Affiliated Hospital of Nantong University, Nantong, 226001, China
| | - Jianlong Zhao
- State Key Laboratory of Transducer Technology, Shanghai Institute of Microsystem and Information Technology, Chinese Academy of Sciences, Shanghai, 200050, China
- Center of Materials Science and Optoelectronics Engineering, University of Chinese Academy of Sciences, Beijing, 100049, China
| |
Collapse
|
13
|
Feng Z, Wu J, Lu Y, Chan YT, Zhang C, Wang D, Luo D, Huang Y, Feng Y, Wang N. Circulating tumor cells in the early detection of human cancers. Int J Biol Sci 2022; 18:3251-3265. [PMID: 35637960 PMCID: PMC9134923 DOI: 10.7150/ijbs.71768] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2022] [Accepted: 04/14/2022] [Indexed: 11/25/2022] Open
Abstract
Cancer is a severe disease with high morbidity and mortality globally. Thus, early detection is emerging as an important topic in modern oncology. Although the strategies for early detection have developed rapidly in recent decades, they remain challenging due to the subtle symptoms in the initial stage of the primary tumor. Currently, tumor biomarkers, imaging, and specific screening tests are widely used in various cancer types; however, each method has limitations. The harms are even overweight against the benefits in some cases. Therefore, early detection approaches should be improved urgently. Liquid biopsy, for now, is a convenient and non-invasive way compared to the traditional tissue biopsy in screening and early diagnosis. Circulating tumor cells (CTCs) are vital in liquid biopsy and play a central role in tumor dissemination and metastases. They have promising potential as cancer biomarkers in early detection. This review updates the knowledge of the biology of CTC; it also highlights the CTC enrichment technologies and their applications in the early detection of several human cancers.
Collapse
Affiliation(s)
- Zixin Feng
- School of Chinese Medicine, The University of Hong Kong, Hong Kong S.A.R, P.R. China
| | - Junyu Wu
- School of Chinese Medicine, The University of Hong Kong, Hong Kong S.A.R, P.R. China
| | - Yuanjun Lu
- School of Chinese Medicine, The University of Hong Kong, Hong Kong S.A.R, P.R. China
| | - Yau-Tuen Chan
- School of Chinese Medicine, The University of Hong Kong, Hong Kong S.A.R, P.R. China
| | - Cheng Zhang
- School of Chinese Medicine, The University of Hong Kong, Hong Kong S.A.R, P.R. China
| | - Di Wang
- School of Life Science, Jilin University, Jilin, P.R. China
| | - Dan Luo
- Cellomics International Limited, Hong Kong S.A.R., P.R. China
| | - Yuan Huang
- Cellomics International Limited, Hong Kong S.A.R., P.R. China
| | - Yibin Feng
- School of Chinese Medicine, The University of Hong Kong, Hong Kong S.A.R, P.R. China
| | - Ning Wang
- School of Chinese Medicine, The University of Hong Kong, Hong Kong S.A.R, P.R. China
| |
Collapse
|
14
|
Lone SN, Nisar S, Masoodi T, Singh M, Rizwan A, Hashem S, El-Rifai W, Bedognetti D, Batra SK, Haris M, Bhat AA, Macha MA. Liquid biopsy: a step closer to transform diagnosis, prognosis and future of cancer treatments. Mol Cancer 2022; 21:79. [PMID: 35303879 PMCID: PMC8932066 DOI: 10.1186/s12943-022-01543-7] [Citation(s) in RCA: 384] [Impact Index Per Article: 128.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2021] [Accepted: 02/21/2022] [Indexed: 02/07/2023] Open
Abstract
Over the past decade, invasive techniques for diagnosing and monitoring cancers are slowly being replaced by non-invasive methods such as liquid biopsy. Liquid biopsies have drastically revolutionized the field of clinical oncology, offering ease in tumor sampling, continuous monitoring by repeated sampling, devising personalized therapeutic regimens, and screening for therapeutic resistance. Liquid biopsies consist of isolating tumor-derived entities like circulating tumor cells, circulating tumor DNA, tumor extracellular vesicles, etc., present in the body fluids of patients with cancer, followed by an analysis of genomic and proteomic data contained within them. Methods for isolation and analysis of liquid biopsies have rapidly evolved over the past few years as described in the review, thus providing greater details about tumor characteristics such as tumor progression, tumor staging, heterogeneity, gene mutations, and clonal evolution, etc. Liquid biopsies from cancer patients have opened up newer avenues in detection and continuous monitoring, treatment based on precision medicine, and screening of markers for therapeutic resistance. Though the technology of liquid biopsies is still evolving, its non-invasive nature promises to open new eras in clinical oncology. The purpose of this review is to provide an overview of the current methodologies involved in liquid biopsies and their application in isolating tumor markers for detection, prognosis, and monitoring cancer treatment outcomes.
Collapse
Affiliation(s)
- Saife N Lone
- Department of Biotechnology, School of Life Sciences, Central University of Kashmir, Ganderbal, Jammu & Kashmir, India
| | - Sabah Nisar
- Laboratory of Molecular and Metabolic Imaging, Cancer Research Department, Sidra Medicine, PO BOX 26999, Doha, Qatar
| | - Tariq Masoodi
- Laboratory of Molecular and Metabolic Imaging, Cancer Research Department, Sidra Medicine, PO BOX 26999, Doha, Qatar
| | - Mayank Singh
- Department of Medical Oncology, Dr. B. R. Ambedkar Institute Rotary Cancer Hospital, All India Institute of Medical Sciences (AIIMS), New Delhi, India
| | - Arshi Rizwan
- Department of Nephrology, All India Institute of Medical Sciences, New Delhi, India
| | - Sheema Hashem
- Laboratory of Molecular and Metabolic Imaging, Cancer Research Department, Sidra Medicine, PO BOX 26999, Doha, Qatar
| | - Wael El-Rifai
- Department of Surgery, University of Miami Miller School of Medicine, Miami, FL, USA
- Sylvester Comprehensive Cancer Center, Miller School of Medicine, University of Miami, Miami, FL, USA
- Department of Veterans Affairs, Miami Healthcare System, Miami, FL, USA
| | - Davide Bedognetti
- Cancer Research Department, Research Branch, Sidra Medicince, Doha, Qatar
- Department of Internal Medicine and Medical Specialities, University of Genova, Genova, Italy
- College of Health and Life Sciences, Hamad Bin Khalifa University, Doha, Qatar
| | - Surinder K Batra
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, NE 68198, Omaha, USA
- Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center , Omaha, NE 68198, USA
- Fred and Pamela Buffett Cancer Center, University of Nebraska Medical Center, University of Nebraska Medical Center, NE 68198, Omaha, USA
| | - Mohammad Haris
- Laboratory of Molecular and Metabolic Imaging, Cancer Research Department, Sidra Medicine, PO BOX 26999, Doha, Qatar
- Laboratory Animal Research Center, Qatar University, Doha, Qatar
- Center for Advanced Metabolic Imaging in Precision Medicine, Department of Radiology, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, USA
| | - Ajaz A Bhat
- Laboratory of Molecular and Metabolic Imaging, Cancer Research Department, Sidra Medicine, PO BOX 26999, Doha, Qatar.
| | - Muzafar A Macha
- Watson-Crick Centre for Molecular Medicine, Islamic University of Science and Technology, (IUST), 192122, Awantipora, Jammu & Kashmir, India.
| |
Collapse
|
15
|
Negative enrichment of circulating tumor cells from unmanipulated whole blood with a 3D printed device. Sci Rep 2021; 11:20583. [PMID: 34663896 PMCID: PMC8523721 DOI: 10.1038/s41598-021-99951-0] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2021] [Accepted: 09/23/2021] [Indexed: 01/07/2023] Open
Abstract
Reliable and routine isolation of circulating tumor cells (CTCs) from peripheral blood would allow effective monitoring of the disease and guide the development of personalized treatments. Negative enrichment of CTCs by depleting normal blood cells ensures against a biased selection of a subpopulation and allows the assay to be applied on different tumor types. Here, we report an additively manufactured microfluidic device that can negatively enrich viable CTCs from clinically-relevant volumes of unmanipulated whole blood samples. Our device depletes nucleated blood cells based on their surface antigens and the smaller anucleated cells based on their size. Enriched CTCs are made available off the device in suspension making our technique compatible with standard immunocytochemical, molecular and functional assays. Our device could achieve a ~ 2.34-log depletion by capturing > 99.5% of white blood cells from 10 mL of whole blood while recovering > 90% of spiked tumor cells. Furthermore, we demonstrated the capability of the device to isolate CTCs from blood samples collected from patients (n = 15) with prostate and pancreatic cancers in a pilot study. A universal CTC assay that can differentiate tumor cells from normal blood cells with the specificity of clinically established membrane antigens yet require no label has the potential to enable routine blood-based tumor biopsies at the point-of-care.
Collapse
|
16
|
Curtin J, Choi SW, Thomson PJ, Lam AKY. Characterization and clinicopathological significance of circulating tumour cells in patients with oral squamous cell carcinoma. Int J Oral Maxillofac Surg 2021; 51:289-299. [PMID: 34154876 DOI: 10.1016/j.ijom.2021.05.020] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2020] [Revised: 04/06/2021] [Accepted: 05/24/2021] [Indexed: 02/07/2023]
Abstract
Circulating tumour cells (CTCs) are cancer cells released by cancer into the peripheral circulation. Haematogenous tumour spread is a hallmark of metastatic malignancy and a key factor in cancer recurrence and prognosis. CTCs have diagnostic and prognostic significance for a number of adenocarcinomas and melanoma. A review of the published peer-reviewed literature was performed to determine the clinical relevance of CTCs as a biomarker in the management of oral squamous cell carcinoma (OSCC). Fourteen studies met the eligibility criteria. With regard to patients with OSCC, this review found the following: (1) CTCs have been detected using multiple techniques; (2) the presence of CTCs does not appear to be related to tumour differentiation or size; (3) CTCs may be detected without lymph node involvement; (4) the detection of CTCs may be prognostic for both disease-free survival and overall survival; (5) quantification of CTCs may reflect the efficacy of therapy; (6) CTCs may be of value for ongoing patient monitoring. Preliminary evidence suggests that CTCs have diagnostic and prognostic potential as a biomarker for oral cancer management and warrant further investigation to determine their appropriate place in the management of OSCC patients.
Collapse
Affiliation(s)
- J Curtin
- School of Medicine, Griffith University, Gold Coast, Queensland, Australia.
| | - S-W Choi
- Department of Oral and Maxillofacial Surgery, Faculty of Dentistry, University of Hong Kong, Hong Kong
| | - P J Thomson
- Department of Oral and Maxillofacial Surgery, Faculty of Dentistry, University of Hong Kong, Hong Kong
| | - A K-Y Lam
- School of Medicine, Griffith University, Gold Coast, Queensland, Australia
| |
Collapse
|
17
|
Verkhovskii RA, Kozlova AA, Sindeeva OA, Kozhevnikov IO, Prikhozhdenko ES, Mayorova OA, Grishin OV, Makarkin MA, Ermakov AV, Abdurashitov AS, Tuchin VV, Bratashov DN. Lightsheet-based flow cytometer for whole blood with the ability for the magnetic retrieval of objects from the blood flow. BIOMEDICAL OPTICS EXPRESS 2021; 12:380-394. [PMID: 33659080 PMCID: PMC7899519 DOI: 10.1364/boe.413845] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/02/2020] [Revised: 11/29/2020] [Accepted: 12/06/2020] [Indexed: 05/04/2023]
Abstract
Detection and extraction of circulating tumor cells and other rare objects in the bloodstream are of great interest for modern diagnostics, but devices that can solve this problem for the whole blood volume of laboratory animals are still rare. Here we have developed SPIM-based lightsheet flow cytometer for the detection of fluorescently-labeled objects in whole blood. The bypass channel between two blood vessels connected with the external flow cell was used to visualize, detect, and magnetically separate fluorescently-labeled objects without hydrodynamic focusing. Carriers for targeted drug delivery were used as model objects to test the device performance. They were injected into the bloodstream of the rat, detected fluorescently, and then captured from the bloodstream by a magnetic separator prior to filtration in organs. Carriers extracted from the whole blood were studied by a number of in vitro methods.
Collapse
Affiliation(s)
| | | | - Olga A. Sindeeva
- Saratov State University, 83 Astrakhanskaya str., Saratov 410012, Russia
- Skolkovo Innovation Center, 3 Nobel str., Moscow 121205, Russia
| | | | | | - Oksana A. Mayorova
- Saratov State University, 83 Astrakhanskaya str., Saratov 410012, Russia
| | - Oleg V. Grishin
- Saratov State University, 83 Astrakhanskaya str., Saratov 410012, Russia
| | | | - Alexey V. Ermakov
- Saratov State University, 83 Astrakhanskaya str., Saratov 410012, Russia
| | | | - Valery V. Tuchin
- Saratov State University, 83 Astrakhanskaya str., Saratov 410012, Russia
- National Research Tomsk State University, 36 Lenin Avenue, Tomsk 634050, Russia
- Institute of Precision Mechanics and Control of the RAS, 24 Rabochaya str., Saratov 410028, Russia
| | | |
Collapse
|
18
|
Minimal Residual Disease in Melanoma:molecular characterization of in transit cutaneous metastases and Circulating Melanoma Cells recognizes an expression panel potentially related to disease progression. Cancer Treat Res Commun 2020; 25:100262. [PMID: 33338742 DOI: 10.1016/j.ctarc.2020.100262] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2020] [Accepted: 12/01/2020] [Indexed: 12/12/2022]
Abstract
Isolating circulating melanoma cells (CMCs) represents a powerful method to monitor minimal residual disease. We documented that MCAM/MUC18/CD146 expression is strongly associated with disease progression. ABCB5 is melanoma-stem antigen with self-renewal, proliferation, differentiation, tumorigenicity capabilities. These findings supported us to improve CMC detection, investigating MCAM/MUC18/CD146 and ABCB5 as enrichment targets in MM progression. Moreover, we decided to compare possible molecular diversity of these CMC fractions with metastatic tissue expression, collecting concomitantly cutaneous in transit metastases (CTM). We enriched CMCs from eight melanoma patients staged ≥pT1b AJCC, who developed CTMs at baseline or during follow up. We assessed a gene expression panel comprising ABCB5, the differentiation markers (Tyrosinase, MART1), angiogenic factors (VEGF, bFGF), the cell-cell adhesion molecules (MCAM/MUC18/CD146 5'-portion, Long, and Short isoforms, E-Cadherin, N-Cadherin, VE-Cadherin) and matrix-metallo-proteinases (MMP2 and MMP9) via high-sensitive RT-PCR. Preliminary findings defined three distinct sub-populations: "endothelial" CD45-CD146+CMCs, "stem" CD45-ABCB5+CMCs and a "hybrid- stem-endothelial"- CD45-MCAM+ABCB5+CMCs. The expression panel documented that - almost high expression found in CTMs - like in 73.5% of CMCs resulted positive for at least one transcript at baseline, showing gene-expression variability. Longitudinal monitoring documented shut-down of all gene-expressions in "endothelial"- and "hybrid stem-endothelial"-subsets, whilst persistency or acquisition of MCAM/MUC18/CD146, VE-CADH and MMPs was documented in disease-progression status.Conversely, a drastic expression shut-down was documented when patients achieved clinical remission. The "stem"- CMCs fraction" showed quite lower gene expression frequencies. MCAM/MUC18/CD146 and ABCB5 as melanoma-specific-targets are effective in the selection of highly primitive CMCs and highlights those putative genes associated with disease spreading progression.
Collapse
|
19
|
Arechederra M, Ávila MA, Berasain C. Liquid biopsy for cancer management: a revolutionary but still limited new tool for precision medicine. ADVANCES IN LABORATORY MEDICINE 2020; 1:20200009. [PMID: 37361495 PMCID: PMC10197281 DOI: 10.1515/almed-2020-0009] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 01/15/2020] [Accepted: 03/19/2020] [Indexed: 06/28/2023]
Abstract
The term liquid biopsy is used in contraposition to the traditional "solid" tissue biopsy. In the oncology field it has opened a new plethora of clinical opportunities as tumor-derived material is shedded into the different biofluids from where it can be isolated and analyzed. Common biofluids include blood, urine, saliva, cerebrospinal fluid (CSF), pleural effusion or bile. Starting from these biological specimens several analytes can be isolated, among which we will review the most widely used: circulating tumor cells (CTCs), circulating tumor DNA (ctDNA), circulating tumor RNA (ctRNA), proteins, metabolites, and exosomes. Regarding the nature of the biomarkers it will depend on the analyte, the type of tumor and the clinical application of the liquid biopsy and it includes, somatic point mutations, deletions, amplifications, gene-fusions, DNA-methylated marks, tumor-specific miRNAs, proteins or metabolites. Here we review the characteristics of the analytes and the methodologies used for their isolation. We also describe the applications of the liquid biopsy in the management of patients with cancer, from the early detection of cancers to treatment guidance in patients with advanced tumors. Finally, we also discuss some current limitations and still open questions.
Collapse
Affiliation(s)
- María Arechederra
- Instituto de Investigaciones Sanitarias de Navarra-IdiSNA, Pamplona, Spain
- Hepatology Program, CIMA, University of Navarra, Avda. Pio XII, n55, 31008, Pamplona, Spain
| | - Matías A. Ávila
- Hepatology Program, CIMA, University of Navarra, Pamplona, Spain
- Instituto de Investigaciones Sanitarias de Navarra-IdiSNA, Pamplona, Spain
- CIBERehd, Instituto de Salud Carlos III, Madrid, Spain
| | - Carmen Berasain
- Instituto de Investigaciones Sanitarias de Navarra-IdiSNA, Pamplona, Spain
- CIBERehd, Instituto de Salud Carlos III, Madrid, Spain
- Hepatology Program, CIMA, University of Navarra, Avda. Pio XII, n55, 31008, Pamplona, Spain
| |
Collapse
|
20
|
Tian C, Xu X, Wang Y, Li D, Lu H, Yang Z. Development and Clinical Prospects of Techniques to Separate Circulating Tumor Cells from Peripheral Blood. Cancer Manag Res 2020; 12:7263-7275. [PMID: 32884342 PMCID: PMC7434565 DOI: 10.2147/cmar.s248380] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2020] [Accepted: 07/15/2020] [Indexed: 12/15/2022] Open
Abstract
Detection of circulating tumor cells (CTC) is an important liquid biopsy technique that has advanced considerably in recent years. To further advance the development of technology for curing cancer, several CTC technologies have been proposed by various research groups. Despite their potential role in early cancer diagnosis and prognosis, CTC methods are currently used for research purposes only, and very few methods have been accepted for clinical applications because of difficulties, including CTC heterogeneity, CTC separation from the blood, and a lack of thorough clinical validation. Although current CTC technologies have not been truly implemented, they possess high potential as future clinical diagnostic techniques for individualized cancer. Here, we review current developments in CTC separation technology. We also explore new CTC detection methods based on telomerase and nanomaterials, such as in vivo flow cytometry. In addition, we discuss the difficulties that must be overcome before CTC can be applied in clinical settings.
Collapse
Affiliation(s)
- Cheng Tian
- Yichang Central People's Hospital, First Clinical Medical College of Three Gorges University, Yichang 443000, People's Republic of China
| | - Xinhua Xu
- Yichang Central People's Hospital, First Clinical Medical College of Three Gorges University, Yichang 443000, People's Republic of China
| | - Yuke Wang
- Yichang Central People's Hospital, First Clinical Medical College of Three Gorges University, Yichang 443000, People's Republic of China
| | - Dailong Li
- Yichang Central People's Hospital, First Clinical Medical College of Three Gorges University, Yichang 443000, People's Republic of China
| | - Haiyan Lu
- Yichang Central People's Hospital, First Clinical Medical College of Three Gorges University, Yichang 443000, People's Republic of China
| | - Ziwei Yang
- Yichang Central People's Hospital, First Clinical Medical College of Three Gorges University, Yichang 443000, People's Republic of China
| |
Collapse
|
21
|
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: 3.2] [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: 08/14/2020] [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
|
22
|
Wang J, Sun N, Lee YT, Ni Y, Koochekpour R, Zhu Y, Tseng HR, Wang S, Jiang L, Zhu H. A circulating tumor cell-based digital assay for the detection of EGFR T790M mutation in advanced non-small cell lung cancer. J Mater Chem B 2020; 8:5636-5644. [PMID: 32525199 PMCID: PMC8136811 DOI: 10.1039/d0tb00589d] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Determining the status of epidermal growth factor receptor (EGFR) T790M mutation is crucial for guiding further treatment intervention in advanced non-small cell lung cancer (NSCLC) patients who develop acquired resistance to initial EGFR tyrosine kinase inhibitor (TKI) treatment. Circulating tumor cells (CTCs) which contain plentiful copies of well-preserved RNA offer an ideal source for noninvasive detection of T790M mutation in NSCLC. We developed a CTC-based digital assay which synergistically integrates NanoVelcro Chips for enriching NSCLC CTCs and reverse-transcription droplet digital PCR (RT-ddPCR) for quantifying T790M transcripts in the enriched CTCs. We collected 46 peripheral arterial and venous blood samples from 27 advanced NSCLC patients for testing this CTC-based digital assay. The results showed that the T790M mutational status observed by the CTC-based digital assay matched with those observed by tissue-based diagnostic methods. Furthermore, higher copy numbers of T790M transcripts were observed in peripheral arterial blood than those detected in the matched peripheral venous blood. In short, our results demonstrated the potential of the NanoVelcro CTC-digital assay for noninvasive detection of the T790M mutation in NSCLC, and suggested that peripheral arterial blood sampling may offer a more abundant CTC source than peripheral venous blood in advanced NSCLC patients.
Collapse
Affiliation(s)
- Jing Wang
- Department of Pathology, School of Basic Medical Sciences, Fudan University, Shanghai, 200032, P. R. China. and California NanoSystems Institute, Crump Institute for Molecular Imaging, Department of Molecular and Medical Pharmacology, University of California, Los Angeles, CA 90095, USA
| | - Na Sun
- California NanoSystems Institute, Crump Institute for Molecular Imaging, Department of Molecular and Medical Pharmacology, University of California, Los Angeles, CA 90095, USA and Key Laboratory for Nano-Bio Interface, Suzhou Institute of Nano-Tech and Nano-Bionics, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Suzhou 215123, P. R. China
| | - Yi-Te Lee
- California NanoSystems Institute, Crump Institute for Molecular Imaging, Department of Molecular and Medical Pharmacology, University of California, Los Angeles, CA 90095, USA
| | - Yiqian Ni
- Department of Respiratory Medicine, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai, 200030, P. R. China.
| | - Rose Koochekpour
- California NanoSystems Institute, Crump Institute for Molecular Imaging, Department of Molecular and Medical Pharmacology, University of California, Los Angeles, CA 90095, USA
| | - Yazhen Zhu
- California NanoSystems Institute, Crump Institute for Molecular Imaging, Department of Molecular and Medical Pharmacology, University of California, Los Angeles, CA 90095, USA
| | - Hsian-Rong Tseng
- California NanoSystems Institute, Crump Institute for Molecular Imaging, Department of Molecular and Medical Pharmacology, University of California, Los Angeles, CA 90095, USA
| | - Shuyang Wang
- Department of Pathology, School of Basic Medical Sciences, Fudan University, Shanghai, 200032, P. R. China.
| | - Liyan Jiang
- Department of Respiratory Medicine, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai, 200030, P. R. China.
| | - Hongguang Zhu
- Department of Pathology, School of Basic Medical Sciences, Fudan University, Shanghai, 200032, P. R. China.
| |
Collapse
|
23
|
Ooi C, Earhart CM, Hughes CE, Lee JR, Wong DJ, Wilson RJ, Rohatgi R, Wang SX. Flow Homogenization Enables a Massively Parallel Fluidic Design for High-throughput and Multiplexed Cell Isolation. ADVANCED MATERIALS TECHNOLOGIES 2020; 5:1900960. [PMID: 33072854 PMCID: PMC7567302 DOI: 10.1002/admt.201900960] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/24/2019] [Accepted: 03/04/2020] [Indexed: 06/11/2023]
Abstract
Microfluidic devices are widely used for applications such as cell isolation. Currently, the most common method to improve throughput for microfluidic devices involves fabrication of multiple, identical channels in parallel. However, this 'numbering up' only occurs in one dimension, thereby limiting gains in volumetric throughput. In contrast, macro-fluidic devices permit high volumetric flow-rates but lack the finer control of microfluidics. Here, we demonstrate how a micro-pore array design enables flow homogenization across a magnetic cell capture device, thus creating a massively parallel series of micro-scale flow channels with consistent fluidic and magnetic properties, regardless of spatial location. This design enables scaling in 2-dimensions, allowing flow-rates exceeding 100 mL/hr while maintaining >90% capture efficiencies of spiked lung cancer cells from blood in a simulated circulating tumor cell system. Additionally, this design facilitates modularity in operation, which we demonstrate by combining two different devices in tandem for multiplexed cell separation in a single pass with no additional cell losses from processing.
Collapse
Affiliation(s)
- Chinchun Ooi
- Department of Chemical Engineering, Stanford University, Stanford, California, USA; Department of Fluid Dynamics, Institute of High Performance Computing, Singapore
| | - Christopher M. Earhart
- Department of Materials Science and Engineering, Stanford University, Stanford, California, USA
| | - Casey E. Hughes
- Division of Oncology, Department of Medicine, Stanford University School of Medicine, Stanford, California, USA; Department of Biochemistry, University of Utah School of Medicine, Salt Lake City, Utah, USA
| | - Jung-Rok Lee
- Division of Mechanical and Biomechanical Engineering, Ewha Womans University, Seoul, South Korea
| | - Dawson J. Wong
- Department of Electrical Engineering, Stanford University, Stanford, California, USA
| | - Robert J. Wilson
- Department of Materials Science and Engineering, Stanford University, Stanford, California, USA
| | - Rajat Rohatgi
- Department of Biochemistry, Stanford University School of Medicine, Stanford, California, USA
| | - Shan X. Wang
- Department of Materials Science and Engineering, Stanford University, Stanford, California, USA; Department of Electrical Engineering, Stanford University, Stanford, California, USA
| |
Collapse
|
24
|
Gopinathan P, Chiang N, Bandaru A, Sinha A, Huang W, Hung S, Shan Y, Lee G. Exploring Circulating Tumor Cells in Cholangiocarcinoma Using a Novel Glycosaminoglycan Probe on a Microfluidic Platform. Adv Healthc Mater 2020; 9:e1901875. [PMID: 32329247 DOI: 10.1002/adhm.201901875] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2019] [Revised: 02/26/2020] [Accepted: 03/12/2020] [Indexed: 12/14/2022]
Abstract
The search of alternative approaches to epithelial cell adhesion molecule (EpCAM), for the isolation of circulating tumor cells (CTC), is on the rise. This work attempts at evaluating the feasibility of using a new glycosaminoglycan, SCH45, as a probe to isolate CTCs from the peripheral blood of 65 advanced/metastatic cholangiocarcinoma (CCA) patients. The positive enrichment of CTCs from 1 mL of blood using SCH45-bound magnetic beads and subsequent staining on an integrated microfluidic platform is demonstrated. Results detailing CTC concentrations averaging ≥1 CTCs mL-1 of blood are shown, and a conventional protein biomarker, EpCAM, has been used to corroborate the finding that 100% of the patients possess CTCs in their blood. Studies detailing the use of CTCs in the prognostic monitoring and treatment effectiveness of advanced/metastatic CCA are scarce, and the isolation of CTCs from all CCA patients tested has not been reported yet. A strong correlation between CTC counts and disease progression at the time of and/or in advance of radiographic imaging in patients receiving chemotherapy is also reported. This study is one of its kind with the new probe and reduced sample volume and has potential for use in CCA diagnosis and prognosis in the near future.
Collapse
Affiliation(s)
- Priya Gopinathan
- Institute of Nanoengineering and MicrosystemsNational Tsing Hua University Hsinchu 30013 Taiwan
| | - Nai‐Jung Chiang
- Institute of Clinical MedicineCollege of MedicineNational Cheng Kung University Tainan 70457 Taiwan
- National Institute of Cancer ResearchNational Health Research Institutes Miaoli 35053 Taiwan
- Department of Internal MedicineNational Cheng Kung University HospitalCollege of MedicineNational Cheng Kung University Tainan 70403 Taiwan
| | - Anandaraju Bandaru
- Genomics Research CentreAcademia Sinica Taipei Taiwan 11529 Republic of China
| | - Anirban Sinha
- Institute of Nanoengineering and MicrosystemsNational Tsing Hua University Hsinchu 30013 Taiwan
| | - Wen‐Yen Huang
- Department of Power Mechanical EngineeringNational Tsing Hua University Hsinchu City 30013 Taiwan
| | - Shang‐Cheng Hung
- Genomics Research CentreAcademia Sinica Taipei Taiwan 11529 Republic of China
- Department of Applied ScienceNational Taitung University Taitung 95053 Taiwan
| | - Yan‐Shen Shan
- Institute of Clinical MedicineCollege of MedicineNational Cheng Kung University Tainan 70457 Taiwan
- Department of SurgeryNational Cheng Kung University HospitalCollege of MedicineNational Cheng Kung University Tainan 70403 Taiwan
| | - Gwo‐Bin Lee
- Institute of Nanoengineering and MicrosystemsNational Tsing Hua University Hsinchu 30013 Taiwan
- Department of Power Mechanical EngineeringNational Tsing Hua University Hsinchu City 30013 Taiwan
- Institute of Biomedical EngineeringNational Tsing Hua University Hsinchu 30013 Taiwan
| |
Collapse
|
25
|
Künzel J, Gribko A, Lu Q, Stauber RH, Wünsch D. Nanomedical detection and downstream analysis of circulating tumor cells in head and neck patients. Biol Chem 2020; 400:1465-1479. [PMID: 30903749 DOI: 10.1515/hsz-2019-0141] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2019] [Accepted: 03/14/2019] [Indexed: 12/27/2022]
Abstract
The establishment of novel biomarkers in liquid biopsies of cancer patients has come more into focus in prognostic and diagnostic research efforts. Due to their prognostic relevance disseminated tumor cells or circulating tumor cells are the subject of intensive research and are discussed as early diagnostic indicators for treatment failure and the formation of micrometastases. A potential association of this early-systemic tumor component with poor prognosis of cancer patients could be already demonstrated for various entities including breast, colon, lung, melanoma, ovarian and prostate cancers. Thus, the detection of circulating tumor cells seems to be also applicable for minimal-invasive monitoring of therapy progress in head and neck cancer patients. A major problem of the use in clinical routine is that circulating tumor cells could not be detected by modern imaging techniques. To overcome these limitations highly sensitive detection methods and techniques for their molecular characterization are urgently needed allowing mechanistic understanding and targeting of circulating tumor cells. Especially the medical application of nanotechnology (nanomedical methods) has made valuable contributions to the field. Here, we want to provide a comprehensive overview on (nanomedical) detection methods for circulating tumor cells and discuss their merits, pitfalls and future perspectives especially for head and neck solid squamous cell carcinoma (HNSCC) patients.
Collapse
Affiliation(s)
- Julian Künzel
- Nanobiomedicine Department/Department of Otorhinolaryngology-Head and Neck Surgery/ENT, University Medical Center Mainz, Langenbeckstrasse 1, D-55131 Mainz, Germany
| | - Alena Gribko
- Nanobiomedicine Department/Department of Otorhinolaryngology-Head and Neck Surgery/ENT, University Medical Center Mainz, Langenbeckstrasse 1, D-55131 Mainz, Germany
| | - Qiang Lu
- Nanobiomedicine Department/Department of Otorhinolaryngology-Head and Neck Surgery/ENT, University Medical Center Mainz, Langenbeckstrasse 1, D-55131 Mainz, Germany
| | - Roland H Stauber
- Nanobiomedicine Department/Department of Otorhinolaryngology-Head and Neck Surgery/ENT, University Medical Center Mainz, Langenbeckstrasse 1, D-55131 Mainz, Germany
| | - Désirée Wünsch
- Nanobiomedicine Department/Department of Otorhinolaryngology-Head and Neck Surgery/ENT, University Medical Center Mainz, Langenbeckstrasse 1, D-55131 Mainz, Germany
| |
Collapse
|
26
|
Rizzo MI, Ralli M, Nicolazzo C, Gradilone A, Carletti R, Di Gioia C, De Vincentiis M, Greco A. Detection of circulating tumor cells in patients with laryngeal cancer using ScreenCell: Comparative pre- and post-operative analysis and association with prognosis. Oncol Lett 2020; 19:4183-4188. [PMID: 32391112 DOI: 10.3892/ol.2020.11528] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2019] [Accepted: 02/20/2020] [Indexed: 12/20/2022] Open
Abstract
The presence of circulating tumor cells (CTCs) in the blood of patients with metastatic breast, colorectal and prostate cancer have been widely investigated; however, few studies have examined CTCs in patients with laryngeal cancer. The present pilot study aimed to detect pre- and postoperative CTCs in the blood of patients with laryngeal cancer and evaluate the association with prognosis. Eight patients with laryngeal squamous cell carcinoma (LSCC) at stage III were included in the present study and underwent total or subtotal laryngectomy and radical bilateral neck lymph node dissection. Blood samples were collected from all patients before and after surgery at different time-points. The following processing steps were followed; preoperative blood sampling, surgery, postoperative blood sampling at 3, 6 and 12 month follow-ups, and prognostic association analysis. CTCs were retained on ScreenCell filters for cytological characterization. The presence of CTCs was associated with a less favorable prognosis, whereas a decrease of CTCs in the postoperative sampling was observed in patients who exhibited an improved therapeutic response. The results of the present pilot study revealed a possible association between the presence of CTCs and a less favorable prognosis in patients with LSCC; therefore, these preliminary findings may encourage further research into the incorporation of a liquid biopsy in the management of LSCC, as this may help identify patients with occult metastatic disease earlier and in a non-invasive manner. In addition, this approach may represent novel independent prognostic factor for use in the clinical evaluation of patients with LSCC.
Collapse
Affiliation(s)
- Maria Ida Rizzo
- Department of Surgical Science, Sapienza University of Rome, Rome 00186, Italy.,Craniofacial Center, Plastic and Maxillofacial Surgery Unit, Bambino Gesù Children Hospital, Rome 00165, Italy
| | - Massimo Ralli
- Department of Sense Organs, Sapienza University of Rome, Rome 00186, Italy
| | - Chiara Nicolazzo
- Department of Molecular Medicine-Circulating Tumor Cells Unit, Sapienza University of Rome, Rome 00186, Italy
| | - Angela Gradilone
- Department of Molecular Medicine-Circulating Tumor Cells Unit, Sapienza University of Rome, Rome 00186, Italy
| | - Raffaella Carletti
- Department of Radiological, Oncological and Pathological Sciences, Sapienza University of Rome, Rome 00186, Italy
| | - Cira Di Gioia
- Department of Radiological, Oncological and Pathological Sciences, Sapienza University of Rome, Rome 00186, Italy
| | - Marco De Vincentiis
- Department of Oral and Maxillofacial Sciences, Sapienza University of Rome, Rome 00186, Italy
| | - Antonio Greco
- Department of Sense Organs, Sapienza University of Rome, Rome 00186, Italy
| |
Collapse
|
27
|
Dong J, Chen JF, Smalley M, Zhao M, Ke Z, Zhu Y, Tseng HR. Nanostructured Substrates for Detection and Characterization of Circulating Rare Cells: From Materials Research to Clinical Applications. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2020; 32:e1903663. [PMID: 31566837 PMCID: PMC6946854 DOI: 10.1002/adma.201903663] [Citation(s) in RCA: 59] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/10/2019] [Revised: 08/02/2019] [Indexed: 05/03/2023]
Abstract
Circulating rare cells in the blood are of great significance for both materials research and clinical applications. For example, circulating tumor cells (CTCs) have been demonstrated as useful biomarkers for "liquid biopsy" of the tumor. Circulating fetal nucleated cells (CFNCs) have shown potential in noninvasive prenatal diagnostics. However, it is technically challenging to detect and isolate circulating rare cells due to their extremely low abundance compared to hematologic cells. Nanostructured substrates offer a unique solution to address these challenges by providing local topographic interactions to strengthen cell adhesion and large surface areas for grafting capture agents, resulting in improved cell capture efficiency, purity, sensitivity, and reproducibility. In addition, rare-cell retrieval strategies, including stimulus-responsiveness and additive reagent-triggered release on different nanostructured substrates, allow for on-demand retrieval of the captured CTCs/CFNCs with high cell viability and molecular integrity. Several nanostructured substrate-enabled CTC/CFNC assays are observed maturing from enumeration and subclassification to molecular analyses. These can one day become powerful tools in disease diagnosis, prognostic prediction, and dynamic monitoring of therapeutic response-paving the way for personalized medical care.
Collapse
Affiliation(s)
- Jiantong Dong
- California NanoSystems Institute, Crump Institute for Molecular Imaging, Department of Molecular and Medical Pharmacology, University of California, Los Angeles, Los Angeles, CA, 90095, USA
- Beijing National Laboratory for Molecular Sciences, MOE Key Laboratory of Bioorganic Chemistry and Molecular Engineering, College of Chemistry and Molecular Engineering, Peking University, Beijing, 100871, P. R. China
| | - Jie-Fu Chen
- Department of Pathology and Immunology, School of Medicine, Washington University in St. Louis, St. Louis, MO, 63110, USA
| | - Matthew Smalley
- California NanoSystems Institute, Crump Institute for Molecular Imaging, Department of Molecular and Medical Pharmacology, University of California, Los Angeles, Los Angeles, CA, 90095, USA
| | - Meiping Zhao
- Beijing National Laboratory for Molecular Sciences, MOE Key Laboratory of Bioorganic Chemistry and Molecular Engineering, College of Chemistry and Molecular Engineering, Peking University, Beijing, 100871, P. R. China
| | - Zunfu Ke
- Department of Pathology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, 510080, P. R. China
| | - Yazhen Zhu
- California NanoSystems Institute, Crump Institute for Molecular Imaging, Department of Molecular and Medical Pharmacology, University of California, Los Angeles, Los Angeles, CA, 90095, USA
| | - Hsian-Rong Tseng
- California NanoSystems Institute, Crump Institute for Molecular Imaging, Department of Molecular and Medical Pharmacology, University of California, Los Angeles, Los Angeles, CA, 90095, USA
| |
Collapse
|
28
|
Abstract
Circulating tumor cells (CTCs) are responsible for the metastatic spread of cancer and therefore are extremely valuable not only for basic research on cancer metastasis but also as potential biomarkers in diagnosing and managing cancer in the clinic. While relatively non-invasive access to the blood tissue presents an opportunity, CTCs are mixed with approximately billion-times more-populated blood cells in circulation. Therefore, the accuracy of technologies for reliable enrichment of the rare CTC population from blood samples is critical to the success of downstream analyses. The focus of this chapter is to provide the reader an overview of significant advances made in the development of diverse CTC enrichment technologies by presenting the strengths of individual techniques in addition to specific challenges remaining to be addressed.
Collapse
|
29
|
Lix K, Tran MV, Massey M, Rees K, Sauvé ER, Hudson ZM, Algar WR. Dextran Functionalization of Semiconducting Polymer Dots and Conjugation with Tetrameric Antibody Complexes for Bioanalysis and Imaging. ACS APPLIED BIO MATERIALS 2019; 3:432-440. [DOI: 10.1021/acsabm.9b00899] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Affiliation(s)
- Kelsi Lix
- Department of Chemistry, University of British Columbia, 2036 Main Mall, Vancouver, British Columbia, Canada V6T 1Z1
| | - Michael V. Tran
- Department of Chemistry, University of British Columbia, 2036 Main Mall, Vancouver, British Columbia, Canada V6T 1Z1
| | - Melissa Massey
- Department of Chemistry, University of British Columbia, 2036 Main Mall, Vancouver, British Columbia, Canada V6T 1Z1
| | - Kelly Rees
- Department of Chemistry, University of British Columbia, 2036 Main Mall, Vancouver, British Columbia, Canada V6T 1Z1
| | - Ethan R. Sauvé
- Department of Chemistry, University of British Columbia, 2036 Main Mall, Vancouver, British Columbia, Canada V6T 1Z1
| | - Zachary M. Hudson
- Department of Chemistry, University of British Columbia, 2036 Main Mall, Vancouver, British Columbia, Canada V6T 1Z1
| | - W. Russ Algar
- Department of Chemistry, University of British Columbia, 2036 Main Mall, Vancouver, British Columbia, Canada V6T 1Z1
| |
Collapse
|
30
|
Abstract
Nanotechnology has made remarkable contributions to clinical oncology. Nanotherapeutics and diagnostic tools have distinctive characteristics which allow them superior abilities to deliver therapeutics and imaging agents for radiation oncology. Compared to solid biopsies and imaging, the analysis of circulating tumor cells (CTCs) offers a more rapid, real-time, and less invasive method to monitor the dynamic molecular profiles of tumors. The potential of CTCs to be translated as a novel cancer biomarker has been demonstrated in numerous clinical studies. This review will discuss clinical applications of nanomaterials in radiation oncology and the implication of CTCs in cancer detection and monitoring.
Collapse
Affiliation(s)
- Bo Sun
- Radiation Oncology, The University of North Carolina at Chapel Hill, 125 Mason Farm Road, Marsico 2236, Chapel Hill, NC 27599, USA
| | - C Tilden Hagan
- UNC/NCSU Joint Department of Biomedical Engineering, 125 Mason Farm Road, Marsico 2120, Chapel Hill, NC 27599, USA
| | - Joseph Caster
- Radiation Oncology, University of Iowa Carver College of Medicine, 200 Hawkins Drive, Iowa City, IA 52242, USA
| | - Andrew Z Wang
- Radiation Oncology, The University of North Carolina at Chapel Hill, 101 Manning Drive, Chapel Hill, NC 27599, USA.
| |
Collapse
|
31
|
Merugu S, Chen L, Gavens E, Gabra H, Brougham M, Makin G, Ng A, Murphy D, Gabriel AS, Robinson ML, Wright JH, Burchill SA, Humphreys A, Bown N, Jamieson D, Tweddle DA. Detection of Circulating and Disseminated Neuroblastoma Cells Using the ImageStream Flow Cytometer for Use as Predictive and Pharmacodynamic Biomarkers. Clin Cancer Res 2019; 26:122-134. [DOI: 10.1158/1078-0432.ccr-19-0656] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2019] [Revised: 08/13/2019] [Accepted: 10/18/2019] [Indexed: 11/16/2022]
|
32
|
|
33
|
Lee SJ, Sim TS, Shin HY, Lee J, Kim MY, Sunoo J, Lee JG, Yea K, Kim YZ, van Noort D, Park SK, Kim WH, Park KW, Kim MS. Microslit on a chip: A simplified filter to capture circulating tumor cells enlarged with microbeads. PLoS One 2019; 14:e0223193. [PMID: 31647823 PMCID: PMC6812780 DOI: 10.1371/journal.pone.0223193] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2019] [Accepted: 09/15/2019] [Indexed: 12/11/2022] Open
Abstract
Microchips are widely used to separate circulating tumor cells (CTCs) from whole blood by virtues of sophisticated manipulation for microparticles. Here, we present a chip with an 8 μm high and 27.9 mm wide slit to capture cancer cells bound to 3 μm beads. Apart from a higher purity and recovery rate, the slit design allows for simplified fabrication, easy cell imaging, less clogging, lower chamber pressure and, therefore, higher throughput. The beads were conjugated with anti-epithelial cell adhesion molecules (anti-EpCAM) to selectively bind to breast cancer cells (MCF-7) used to spike the whole blood. The diameter of the cell-bead construct was in average 23.1 μm, making them separable from other cells in the blood. As a result, the cancer cells were separated from 5 mL of whole blood with a purity of 52.0% and a recovery rate of 91.1%, and also we confirmed that the device can be applicable to clinical samples of human breast cancer patients. The simple design with microslit, by eliminating any high-aspect ratio features, is expected to reduce possible defects on the chip and, therefore, more suitable for mass production without false separation outputs.
Collapse
Affiliation(s)
- Seung Joon Lee
- Department of New Biology, DGIST, Daegu, Republic of Korea
- CytoDx, Pangyo-ro, Beon-gil, Bundang-gu, Seongnam-si, Gyeonggi-do, Republic of Korea
| | - Tae Seok Sim
- Samsung Electronics, Ltd., Maetan3-dong, Youngtong-gu, Suwon-si, Gyeonggi-do, Republic of Korea
| | | | - Jungmin Lee
- Department of New Biology, DGIST, Daegu, Republic of Korea
| | - Min Young Kim
- Department of New Biology, DGIST, Daegu, Republic of Korea
| | - Joseph Sunoo
- CytoDx, Pangyo-ro, Beon-gil, Bundang-gu, Seongnam-si, Gyeonggi-do, Republic of Korea
| | - Jeong-Gun Lee
- Samsung Electronics, Ltd., Maetan3-dong, Youngtong-gu, Suwon-si, Gyeonggi-do, Republic of Korea
| | - Kyungmoo Yea
- Department of New Biology, DGIST, Daegu, Republic of Korea
| | - Young Zoon Kim
- Division of Neurooncology and Department of Neurosurgery, Samsung Changwon Hospital, Sungkyunkwan University School of Medicine, Changwon, Republic of Korea
| | - Danny van Noort
- Department of New Biology, DGIST, Daegu, Republic of Korea
- Division of Biotechnology, IFM, Linköping University, Linköping, Sweden
| | - Soo Kyung Park
- CytoDx, Pangyo-ro, Beon-gil, Bundang-gu, Seongnam-si, Gyeonggi-do, Republic of Korea
| | - Woon-Hae Kim
- Department of New Biology, DGIST, Daegu, Republic of Korea
| | - Kyun Woo Park
- Daejeon Wellness Hospital, Beon-gil, Dongseo-daero, Daedeok-gu, Daejeon, Republic of Korea
| | - Minseok S. Kim
- Department of New Biology, DGIST, Daegu, Republic of Korea
- Translational Responsive Medicine Center, DGIST, Daegu, Republic of Korea
- * E-mail:
| |
Collapse
|
34
|
Li W, Li R, Huang B, Wang Z, Sun Y, Wei X, Heng C, Liu W, Yu M, Guo SS, Zhao XZ. TiO 2 nanopillar arrays coated with gelatin film for efficient capture and undamaged release of circulating tumor cells. NANOTECHNOLOGY 2019; 30:335101. [PMID: 30965310 DOI: 10.1088/1361-6528/ab176c] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/18/2023]
Abstract
Circulating tumor cells (CTCs) are important for the detection and treatment of cancer. Nevertheless, a low density of circulating tumor cells makes the capture and release of CTCs an obstacle. In this work, TiO2 nanopillar arrays coated with gelatin film were synthesized for efficient capture and undamaged release of circulating tumor cells. The scanning electron microscope and atomic force microscope images demonstrate that the substrate has a certain roughness. The interaction between the cell membrane and the nanostructure substrate contributes to the efficient capture of CTC (capture efficiency up to 94.98%). The gelatin layer has excellent biocompatibility and can be rapidly digested by matrix metalloproteinase (MMP9), which realizes the non-destructive release of CTCs (0.1 mg ml-1, 5 min, nearly 100% release efficiency, activity 100%). Therefore, by our strategy, the CTCs can be efficiently captured and released undamaged, which is important for subsequent analysis.
Collapse
Affiliation(s)
- Wei Li
- Key Laboratory of Artificial Micro- and Nano-Structures of Ministry of Education, School of Physics and Technology, Wuhan University, Wuhan, Hubei, 430072, People's Republic of China
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
35
|
Yue WQ, Tan Z, Li XP, Liu FF, Wang C. Micro/nanofluidic technologies for efficient isolation and detection of circulating tumor cells. Trends Analyt Chem 2019. [DOI: 10.1016/j.trac.2019.06.009] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
|
36
|
Iliescu FS, Poenar DP, Yu F, Ni M, Chan KH, Cima I, Taylor HK, Cima I, Iliescu C. Recent advances in microfluidic methods in cancer liquid biopsy. BIOMICROFLUIDICS 2019; 13:041503. [PMID: 31431816 PMCID: PMC6697033 DOI: 10.1063/1.5087690] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/03/2019] [Accepted: 06/24/2019] [Indexed: 05/04/2023]
Abstract
Early cancer detection, its monitoring, and therapeutical prediction are highly valuable, though extremely challenging targets in oncology. Significant progress has been made recently, resulting in a group of devices and techniques that are now capable of successfully detecting, interpreting, and monitoring cancer biomarkers in body fluids. Precise information about malignancies can be obtained from liquid biopsies by isolating and analyzing circulating tumor cells (CTCs) or nucleic acids, tumor-derived vesicles or proteins, and metabolites. The current work provides a general overview of the latest on-chip technological developments for cancer liquid biopsy. Current challenges for their translation and their application in various clinical settings are discussed. Microfluidic solutions for each set of biomarkers are compared, and a global overview of the major trends and ongoing research challenges is given. A detailed analysis of the microfluidic isolation of CTCs with recent efforts that aimed at increasing purity and capture efficiency is provided as well. Although CTCs have been the focus of a vast microfluidic research effort as the key element for obtaining relevant information, important clinical insights can also be achieved from alternative biomarkers, such as classical protein biomarkers, exosomes, or circulating-free nucleic acids. Finally, while most work has been devoted to the analysis of blood-based biomarkers, we highlight the less explored potential of urine as an ideal source of molecular cancer biomarkers for point-of-care lab-on-chip devices.
Collapse
Affiliation(s)
- Florina S. Iliescu
- School of Applied Science, Republic Polytechnic, Singapore 738964, Singapore
| | - Daniel P. Poenar
- VALENS-Centre for Bio Devices and Signal Analysis, School of EEE, Nanyang Technological University, Singapore 639798, Singapore
| | - Fang Yu
- Singapore Institute of Manufacturing Technology, A*STAR, Singapore 138634, Singapore
| | - Ming Ni
- School of Biological Sciences and Engineering, Yachay Technological University, San Miguel de Urcuquí 100105, Ecuador
| | - Kiat Hwa Chan
- Division of Science, Yale-NUS College, Singapore 138527, Singapore
| | | | - Hayden K. Taylor
- Department of Mechanical Engineering, University of California, Berkeley, California 94720, USA
| | - Igor Cima
- DKFZ-Division of Translational Oncology/Neurooncology, German Cancer Consortium (DKTK), Heidelberg and University Hospital Essen, Essen 45147, Germany
| | | |
Collapse
|
37
|
Insights on CTC Biology and Clinical Impact Emerging from Advances in Capture Technology. Cells 2019; 8:cells8060553. [PMID: 31174404 PMCID: PMC6627072 DOI: 10.3390/cells8060553] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2019] [Revised: 05/31/2019] [Accepted: 06/03/2019] [Indexed: 01/01/2023] Open
Abstract
Circulating tumor cells (CTCs) and circulating tumor microemboli (CTM) have been shown to correlate negatively with patient survival. Actual CTC counts before and after treatment can be used to aid in the prognosis of patient outcomes. The presence of circulating tumor materials (CTMat) can advertise the presence of metastasis before clinical presentation, enabling the early detection of relapse. Importantly, emerging evidence is indicating that cancer treatments can actually increase the incidence of CTCs and metastasis in pre-clinical models. Subsequently, the study of CTCs, their biology and function are of vital importance. Emerging technologies for the capture of CTC/CTMs and CTMat are elucidating vitally important biological and functional information that can lead to important alterations in how therapies are administered. This paves the way for the development of a "liquid biopsy" where treatment decisions can be informed by information gleaned from tumor cells and tumor cell debris in the blood.
Collapse
|
38
|
Gribko A, Künzel J, Wünsch D, Lu Q, Nagel SM, Knauer SK, Stauber RH, Ding GB. Is small smarter? Nanomaterial-based detection and elimination of circulating tumor cells: current knowledge and perspectives. Int J Nanomedicine 2019; 14:4187-4209. [PMID: 31289440 PMCID: PMC6560927 DOI: 10.2147/ijn.s198319] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Circulating tumor cells (CTCs) are disseminated cancer cells. The occurrence and circulation of CTCs seem key for metastasis, still the major cause of cancer-associated deaths. As such, CTCs are investigated as predictive biomarkers. However, due to their rarity and heterogeneous biology, CTCs’ practical use has not made it into the clinical routine. Clearly, methods for the effective isolation and reliable detection of CTCs are urgently needed. With the development of nanotechnology, various nanosystems for CTC isolation and enrichment and CTC-targeted cancer therapy have been designed. Here, we summarize the relationship between CTCs and tumor metastasis, and describe CTCs’ unique properties hampering their effective enrichment. We comment on nanotechnology-based systems for CTC isolation and recent achievements in microfluidics and lab-on-a-chip technologies. We discuss recent advances in CTC-targeted cancer therapy exploiting the unique properties of nanomaterials. We conclude by introducing developments in CTC-directed nanosystems and other advanced technologies currently in (pre)clinical research.
Collapse
Affiliation(s)
- Alena Gribko
- Nanobiomedicine Department/ENT, University Medical Center Mainz, Mainz 55131, Germany, ;
| | - Julian Künzel
- Nanobiomedicine Department/ENT, University Medical Center Mainz, Mainz 55131, Germany, ;
| | - Désirée Wünsch
- Nanobiomedicine Department/ENT, University Medical Center Mainz, Mainz 55131, Germany, ;
| | - Qiang Lu
- Nanobiomedicine Department/ENT, University Medical Center Mainz, Mainz 55131, Germany, ;
| | - Sophie Madeleine Nagel
- Nanobiomedicine Department/ENT, University Medical Center Mainz, Mainz 55131, Germany, ;
| | - Shirley K Knauer
- Department of Molecular Biology II, Center for Medical Biotechnology (ZMB)/Center for Nanointegration (CENIDE), University Duisburg-Essen, Essen 45117, Germany
| | - Roland H Stauber
- Nanobiomedicine Department/ENT, University Medical Center Mainz, Mainz 55131, Germany, ;
| | - Guo-Bin Ding
- Nanobiomedicine Department/ENT, University Medical Center Mainz, Mainz 55131, Germany, ; .,Institute of Biotechnology, Key Laboratory of Chemical Biology and Molecular Engineering of Ministry of Education, Shanxi University, Taiyuan 030006, People's Republic of China,
| |
Collapse
|
39
|
Bankó P, Lee SY, Nagygyörgy V, Zrínyi M, Chae CH, Cho DH, Telekes A. Technologies for circulating tumor cell separation from whole blood. J Hematol Oncol 2019; 12:48. [PMID: 31088479 PMCID: PMC6518774 DOI: 10.1186/s13045-019-0735-4] [Citation(s) in RCA: 220] [Impact Index Per Article: 36.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2019] [Accepted: 04/18/2019] [Indexed: 12/13/2022] Open
Abstract
The importance of early cancer diagnosis and improved cancer therapy has been clear for years and has initiated worldwide research towards new possibilities in the care strategy of patients with cancer using technological innovations. One of the key research fields involves the separation and detection of circulating tumor cells (CTC) because of their suggested important role in early cancer diagnosis and prognosis, namely, providing easy access by a liquid biopsy from blood to identify metastatic cells before clinically detectable metastasis occurs and to study the molecular and genetic profile of these metastatic cells. Provided the opportunity to further progress the development of technology for treating cancer, several CTC technologies have been proposed in recent years by various research groups and companies. Despite their potential role in cancer healthcare, CTC methods are currently mainly used for research purposes, and only a few methods have been accepted for clinical application because of the difficulties caused by CTC heterogeneity, CTC separation from the blood, and a lack of thorough clinical validation. Therefore, the standardization and clinical application of various developed CTC technologies remain important subsequent necessary steps. Because of their suggested future clinical benefits, we focus on describing technologies using whole blood samples without any pretreatment and discuss their advantages, use, and significance. Technologies using whole blood samples utilize size-based, immunoaffinity-based, and density-based methods or combinations of these methods as well as positive and negative enrichment during separation. Although current CTC technologies have not been truly implemented yet, they possess high potential as future clinical diagnostic techniques for the individualized therapy of patients with cancer. Thus, a detailed discussion of the clinical suitability of these new advanced technologies could help prepare clinicians for the future and can be a foundation for technologies that would be used to eliminate CTCs in vivo.
Collapse
Affiliation(s)
- Petra Bankó
- Department of Biochemical Engineering, Budapest University of Technology and Economics, Budapest, Hungary
| | - Sun Young Lee
- Department of Radiation Oncology, Chonbuk National University Hospital, Jeonju, Republic of Korea
- Research Institute of Clinical Medicine of Chonbuk National University-Biomedical, Research Institute of Chonbuk National University Hospital, Jeonju, Republic of Korea
| | | | - Miklós Zrínyi
- Laboratory of Nanochemistry, Department of Biophysics and Radiation Biology, Semmelweis University, Budapest, Hungary
| | - Chang Hoon Chae
- Laboratory of Nanochemistry, Department of Biophysics and Radiation Biology, Semmelweis University, Budapest, Hungary
| | - Dong Hyu Cho
- Research Institute of Clinical Medicine of Chonbuk National University-Biomedical, Research Institute of Chonbuk National University Hospital, Jeonju, Republic of Korea
- Department of Obstetrics and Gynecology, Chonbuk National University Hospital, Jeonju, Republic of Korea
| | - András Telekes
- Department of Oncology, St. Lazarus Hospital, Salgótarján, Hungary
| |
Collapse
|
40
|
Agnoletto C, Corrà F, Minotti L, Baldassari F, Crudele F, Cook WJJ, Di Leva G, d'Adamo AP, Gasparini P, Volinia S. Heterogeneity in Circulating Tumor Cells: The Relevance of the Stem-Cell Subset. Cancers (Basel) 2019; 11:cancers11040483. [PMID: 30959764 PMCID: PMC6521045 DOI: 10.3390/cancers11040483] [Citation(s) in RCA: 100] [Impact Index Per Article: 16.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2019] [Revised: 03/16/2019] [Accepted: 03/30/2019] [Indexed: 12/20/2022] Open
Abstract
The release of circulating tumor cells (CTCs) into vasculature is an early event in the metastatic process. The analysis of CTCs in patients has recently received widespread attention because of its clinical implications, particularly for precision medicine. Accumulated evidence documents a large heterogeneity in CTCs across patients. Currently, the most accepted view is that tumor cells with an intermediate phenotype between epithelial and mesenchymal have the highest plasticity. Indeed, the existence of a meta-stable or partial epithelial–mesenchymal transition (EMT) cell state, with both epithelial and mesenchymal features, can be easily reconciled with the concept of a highly plastic stem-like state. A close connection between EMT and cancer stem cells (CSC) traits, with enhanced metastatic competence and drug resistance, has also been described. Accordingly, a subset of CTCs consisting of CSC, present a stemness profile, are able to survive chemotherapy, and generate metastases after xenotransplantation in immunodeficient mice. In the present review, we discuss the current evidence connecting CTCs, EMT, and stemness. An improved understanding of the CTC/EMT/CSC connections may uncover novel therapeutic targets, irrespective of the tumor type, since most cancers seem to harbor a pool of CSCs, and disclose important mechanisms underlying tumorigenicity.
Collapse
Affiliation(s)
- Chiara Agnoletto
- Department of Morphology, Surgery and Experimental Medicine, University of Ferrara, 44121 Ferrara, Italy.
| | - Fabio Corrà
- Department of Morphology, Surgery and Experimental Medicine, University of Ferrara, 44121 Ferrara, Italy.
| | - Linda Minotti
- Department of Morphology, Surgery and Experimental Medicine, University of Ferrara, 44121 Ferrara, Italy.
| | - Federica Baldassari
- Department of Morphology, Surgery and Experimental Medicine, University of Ferrara, 44121 Ferrara, Italy.
| | - Francesca Crudele
- Department of Morphology, Surgery and Experimental Medicine, University of Ferrara, 44121 Ferrara, Italy.
| | | | - Gianpiero Di Leva
- School of Environment and Life Sciences, University of Salford, Salford M5 4WT, UK.
| | - Adamo Pio d'Adamo
- Department of Medicine, Surgery and Health Sciences, University of Trieste, 34127 Trieste, Italy.
- Institute for Maternal and Child Health-IRCCS "Burlo Garofolo", 34137 Trieste, Italy.
| | - Paolo Gasparini
- Department of Medicine, Surgery and Health Sciences, University of Trieste, 34127 Trieste, Italy.
- Institute for Maternal and Child Health-IRCCS "Burlo Garofolo", 34137 Trieste, Italy.
| | - Stefano Volinia
- Department of Morphology, Surgery and Experimental Medicine, University of Ferrara, 44121 Ferrara, Italy.
| |
Collapse
|
41
|
Lustberg MB, Stover DG, Chalmers JJ. Implementing Liquid Biopsies in Clinical Trials: State of Affairs, Opportunities, and Challenges. Cancer J 2019; 24:61-64. [PMID: 29601331 PMCID: PMC5880324 DOI: 10.1097/ppo.0000000000000309] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
A primary goal of personalized medicine is to develop tumor-specific biomarkers to aid in treatment selection and to better evaluate response to targeted therapies. The assessment of circulating blood markers as surrogate real-time biopsies of disease status, termed liquid biopsies, has been under investigation. There are many different types of liquid biopsies each with different functionalities and limitations. These include tumor markers, circulating tumor cells, cell-free DNA, and extracellular vesicles including exosomes. Multiple clinical trials have evaluated liquid biopsies as prognostic biomarkers with positive results. Additional studies are underway to evaluate liquid biopsies as predictive biomarkers, pharmacodynamic biomarkers, and surrogate efficacy endpoints for treatment response evaluation. There are several challenges in and barriers to implementation of liquid biopsies into clinical trials and subsequently into routine clinical practice, which are addressed in this review.
Collapse
Affiliation(s)
- Maryam B. Lustberg
- Stefanie Spielman Comprehensive Breast Center, The Ohio State University, Columbus, OH, USA
| | - Daniel G Stover
- Stefanie Spielman Comprehensive Breast Center, The Ohio State University, Columbus, OH, USA
| | - Jeff J Chalmers
- William G. Lowrie Department of Chemical and Biomolecular Engineering, Ohio State University, 151 W. Woodruff Ave., Columbus, OH, 43210, USA
| |
Collapse
|
42
|
Wang HM, Wu MH, Chang PH, Lin HC, Liao CD, Wu SM, Hung TM, Lin CY, Chang TC, Tzu-Tsen Y, Hsieh JCH. The change in circulating tumor cells before and during concurrent chemoradiotherapy is associated with survival in patients with locally advanced head and neck cancer. Head Neck 2019; 41:2676-2687. [PMID: 30903634 DOI: 10.1002/hed.25744] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2018] [Accepted: 03/07/2019] [Indexed: 12/26/2022] Open
Abstract
BACKGROUND This study aimed to evaluate the role of baseline circulating tumor cells (CTCs) before and during concurrent chemoradiotherapy and attempted to determine the impacts of CTCs on the outcomes in patients with head and neck squamous cell carcinoma. METHODS CTCs were detected using a negative selection strategy and flow cytometry protocol. RESULTS We observed a significant correlation between baseline CTCs and staging (P = 0.001). The CTC counts were significantly reduced within 2-4 weeks in 47 concurrent chemoradiotherapy responders (P < 0.001). Change of CTC counts correlates with progression-free survival (PFS, P = 0.01) and overall survival (OS, P = 0.01). CTC decline status was an independent prognostic factor in PFS (P = 0.03) and OS (P = 0.05) in multivariate analyses. CONCLUSION In chemoradiotherapy responders, CTCs are significantly reduced. CTC decline within the first month indicates a longer PFS and OS, suggesting that the dynamics of CTCs could be more important than CTC number alone.
Collapse
Affiliation(s)
- Hung-Ming Wang
- Circulating Tumor Cell Lab, Division of Hematology-Oncology, Department of Internal Medicine, Chang Gung Memorial Hospital, Linkou and Chang Gung University, Taoyuan, Taiwan
| | - Min-Hsien Wu
- Circulating Tumor Cell Lab, Division of Hematology-Oncology, Department of Internal Medicine, Chang Gung Memorial Hospital, Linkou and Chang Gung University, Taoyuan, Taiwan.,Graduate Institute of Biomedical Engineering, Chang Gung University, Taoyuan, Taiwan.,Department of Chemical Engineering, Ming Chi University of Technology, New Taipei City, Taiwan
| | - Pei-Hung Chang
- Division of Hematology-Oncology, Department of Internal Medicine, Chang Gung Memorial Hospital, Keelung, Taiwan and Chang Gung University, Taoyuan, Taiwan.,Cancer Center, Chang Gung Memorial Hospital, Keelung, and Chang Gung University, Taoyuan, Taiwan
| | - Hung-Chi Lin
- Circulating Tumor Cell Lab, Division of Hematology-Oncology, Department of Internal Medicine, Chang Gung Memorial Hospital, Linkou and Chang Gung University, Taoyuan, Taiwan
| | - Chun-Da Liao
- Department of Otorhinolaryngology, Head and Neck Surgery, Chang Gung Memorial Hospital, Linkou and Chang Gung University, Taoyuan, Taiwan
| | - Sheng-Min Wu
- Circulating Tumor Cell Lab, Division of Hematology-Oncology, Department of Internal Medicine, Chang Gung Memorial Hospital, Linkou and Chang Gung University, Taoyuan, Taiwan
| | - Tsung-Min Hung
- Department of Radiation Oncology, Chang Gung Memorial Hospital, Linkou, College of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Chine-Yu Lin
- Department of Radiation Oncology, Chang Gung Memorial Hospital, Linkou, College of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Tung-Chieh Chang
- Department of Radiation Oncology, Chang Gung Memorial Hospital, Linkou, College of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Yen Tzu-Tsen
- Molecular Imaging Center, Linkou Chang Gung Memorial Hospital, Linkou and Chang Gung University, Taoyuan, Taiwan.,Department of Nuclear Medicine, Linkou Chang Gung Memorial Hospital, Linkou and Chang Gung University, Taoyuan, Taiwan
| | - Jason Chia-Hsun Hsieh
- Circulating Tumor Cell Lab, Division of Hematology-Oncology, Department of Internal Medicine, Chang Gung Memorial Hospital, Linkou and Chang Gung University, Taoyuan, Taiwan.,Graduate Institute of Biomedical Engineering, Chang Gung University, Taoyuan, Taiwan
| |
Collapse
|
43
|
Perumal V, Corica T, Dharmarajan AM, Sun Z, Dhaliwal SS, Dass CR, Dass J. Circulating Tumour Cells (CTC), Head and Neck Cancer and Radiotherapy; Future Perspectives. Cancers (Basel) 2019; 11:E367. [PMID: 30875950 PMCID: PMC6468366 DOI: 10.3390/cancers11030367] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2019] [Revised: 03/10/2019] [Accepted: 03/11/2019] [Indexed: 02/07/2023] Open
Abstract
Head and neck cancer is the seventh most common cancer in Australia and globally. Despite the current improved treatment modalities, there is still up to 50⁻60% local regional recurrence and or distant metastasis. High-resolution medical imaging technologies such as PET/CT and MRI do not currently detect the early spread of tumour cells, thus limiting the potential for effective minimal residual detection and early diagnosis. Circulating tumour cells (CTCs) are a rare subset of cells that escape from the primary tumour and enter into the bloodstream to form metastatic deposits or even re-establish themselves in the primary site of the cancer. These cells are more aggressive and accumulate gene alterations by somatic mutations that are the same or even greater than the primary tumour because of additional features acquired in the circulation. The potential application of CTC in clinical use is to acquire a liquid biopsy, by taking a reliable minimally invasive venous blood sample, for cell genotyping during radiotherapy treatment to monitor the decline in CTC detectability, and mutational changes in response to radiation resistance and radiation sensitivity. Currently, very little has been published on radiation therapy, CTC, and circulating cancer stem cells (CCSCs). The prognostic value of CTC in cancer management and personalised medicine for head and neck cancer radiotherapy patients requires a deeper understanding at the cellular level, along with other advanced technologies. With this goal, this review summarises the current research of head and neck cancer CTC, CCSC and the molecular targets for personalised radiotherapy response.
Collapse
Affiliation(s)
- Vanathi Perumal
- School of Pharmacy and Biomedical Sciences, Curtin University, Perth, WA 6102, Australia.
- Radiation Oncology, Sir Charles Gairdner Hospital, Cancer Centre, Nedlands, Perth, WA 6009, Australia.
- Stem Cell and Cancer Biology Laboratory, School of Pharmacy and Biomedical Sciences, Curtin Health Innovation Research Institute, Curtin University, Perth, WA 6102, Australia.
- Curtin Health Innovation Research Institute, Curtin University, Perth, WA 6102, Australia.
| | - Tammy Corica
- Radiation Oncology, Sir Charles Gairdner Hospital, Cancer Centre, Nedlands, Perth, WA 6009, Australia.
| | - Arun M Dharmarajan
- School of Pharmacy and Biomedical Sciences, Curtin University, Perth, WA 6102, Australia.
- Stem Cell and Cancer Biology Laboratory, School of Pharmacy and Biomedical Sciences, Curtin Health Innovation Research Institute, Curtin University, Perth, WA 6102, Australia.
- Curtin Health Innovation Research Institute, Curtin University, Perth, WA 6102, Australia.
| | - Zhonghua Sun
- Discipline of Medical Radiation Sciences, School of Molecular and Life Sciences, Faculty of Science and Engineering, Curtin University, Perth, WA 6102, Australia.
| | - Satvinder S Dhaliwal
- School of Public Health, Faculty of Health Sciences, Curtin University, Perth, WA 6102, Australia.
| | - Crispin R Dass
- School of Pharmacy and Biomedical Sciences, Curtin University, Perth, WA 6102, Australia.
- Curtin Health Innovation Research Institute, Curtin University, Perth, WA 6102, Australia.
| | - Joshua Dass
- Radiation Oncology, Sir Charles Gairdner Hospital, Cancer Centre, Nedlands, Perth, WA 6009, Australia.
| |
Collapse
|
44
|
S Iliescu F, Sim WJ, Heidari H, P Poenar D, Miao J, Taylor HK, Iliescu C. Highlighting the uniqueness in dielectrophoretic enrichment of circulating tumor cells. Electrophoresis 2019; 40:1457-1477. [PMID: 30676660 DOI: 10.1002/elps.201800446] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2018] [Revised: 01/19/2019] [Accepted: 01/20/2019] [Indexed: 12/14/2022]
Abstract
Circulating tumor cells (CTCs) play an essential role in the metastasis of tumors, and thus can serve as a valuable prognostic factor for malignant diseases. As a result, the ability to isolate and characterize CTCs is essential. This review underlines the potential of dielectrophoresis for CTCs enrichment. It begins by summarizing the key performance parameters and challenges of CTCs isolation using microfluidics. The two main categories of CTCs enrichment-affinity-based and label-free methods-are analysed, emphasising the advantages and disadvantages of each as well as their clinical potential. While the main argument in favour of affinity-based methods is the strong specificity of CTCs isolation, the major advantage of the label-free technologies is in preserving the integrity of the cellular membrane, an essential requirement for downstream characterization. Moving forward, we try to answer the main question: "What makes dielectrophoresis a method of choice in CTCs isolation?" The uniqueness of dielectrophoretic CTCs enrichment resides in coupling the specificity of the isolation process with the conservation of the membrane surface. The specificity of the dielectrophoretic method stems from the differences in the dielectric properties between CTCs and other cells in the blood: the capacitances of the malignantly transformed cellular membranes of CTCs differ from those of other cells. Examples of dielectrophoretic devices are described and their performance evaluated. Critical requirements for using dielectrophoresis to isolate CTCs are highlighted. Finally, we consider that DEP has the potential of becoming a cytometric method for large-scale sorting and characterization of cells.
Collapse
Affiliation(s)
| | - Wen Jing Sim
- Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, Singapore.,NUS Synthetic Biology for Clinical and Technological Innovation (SynCTI), National University of Singapore, Singapore
| | - Hossein Heidari
- Department of Mechanical Engineering, University of California, Berkeley, CA, USA
| | - Daniel P Poenar
- VALENS-Centre for Bio Devices and Signal Analysis, Nanyang Technological University, Singapore
| | - Jianmin Miao
- School of Mechanical and Aerospace Engineering, Nanyang Technological University, Singapore
| | - Hayden K Taylor
- Department of Mechanical Engineering, University of California, Berkeley, CA, USA
| | - Ciprian Iliescu
- Biomedical Institute for Global Health Research & Technology (BIGHEART), National University of Singapore, Singapore
| |
Collapse
|
45
|
Aljohani HM, Aittaleb M, Furgason JM, Amaya P, Deeb A, Chalmers JJ, Bahassi EM. Genetic mutations associated with lung cancer metastasis to the brain. Mutagenesis 2019; 33:137-145. [PMID: 29474635 DOI: 10.1093/mutage/gey003] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2017] [Accepted: 01/28/2018] [Indexed: 12/30/2022] Open
Abstract
Approximately 90% of all cancer deaths arise from the metastatic spread of primary tumours. Of all the processes involved in carcinogenesis, local invasion and the formation of metastases are clinically the most relevant, but they are the least well understood at the molecular level. As a barrier to metastasis, cells normally undergo an apoptotic process known as 'anoikis', in circulation. The recent technological advances in the isolation and characterisation of rare circulating tumour cells (CTCs) will allow a better understanding of anoikis resistance. Detailed molecular and functional analyses of anoikis-resistant cells may provide insight into the biology of cancer metastasis and help identify novel targets for prevention of cancer dissemination. To uncover the molecular changes that govern the transition from a primary lung tumour to a secondary metastasis and specifically the mechanisms by which CTCs survive in circulation, we carried out whole genome sequencing (WGS) of normal lung, primary tumours and the corresponding brain metastases from five patients with progressive metastatic non-small-cell lung carcinoma. We also isolated CTCs from patients with metastatic cancer and subjected them to whole genome amplification and Sanger sequencing of genes of interest. While the primary tumours showed mutations in genes associated with cell adhesion and motility, brain metastases acquired mutations in adaptive, cytoprotective genes involved in response to cellular stress such as Keap-1, Nrf2 and P300, which are key players of the Keap1-Nrf2-ARE survival pathway. Nrf2 is a transcriptional factor that upon stress translocates into the nucleus, binds to the anti-oxidant response elements (ARE) and drives the expression of anti-oxidant genes. The identified mutations affect regulatory domains in all three proteins, suggesting a functional role in providing a survival advantage to CTCs in the peripheral blood allowing their dissemination to distant organs.
Collapse
Affiliation(s)
- Hashim M Aljohani
- Department of Internal Medicine, Division of Hematology and Oncology and UC Brain Tumor Center, Cincinnati, OH, USA.,Department of Molecular Genetics and Biochemistry, Cincinnati, OH, USA
| | - Mohamed Aittaleb
- Department of Internal Medicine, Division of Hematology and Oncology and UC Brain Tumor Center, Cincinnati, OH, USA
| | - John M Furgason
- Department of Internal Medicine, Division of Hematology and Oncology and UC Brain Tumor Center, Cincinnati, OH, USA.,Cincinnati Children's Hospital Medical Center, University of Cincinnati, Cincinnati, OH, USA
| | - Peter Amaya
- William G. Lowrie Department of Chemical and Biomolecular Engineering, The Ohio State University, Koffolt Lab, CBEC, Columbus, OH, USA
| | - Ayham Deeb
- Department of Internal Medicine, Division of Hematology and Oncology and UC Brain Tumor Center, Cincinnati, OH, USA
| | - Jeffery J Chalmers
- William G. Lowrie Department of Chemical and Biomolecular Engineering, The Ohio State University, Koffolt Lab, CBEC, Columbus, OH, USA
| | - El Mustapha Bahassi
- Department of Internal Medicine, Division of Hematology and Oncology and UC Brain Tumor Center, Cincinnati, OH, USA
| |
Collapse
|
46
|
Opoku-Damoah Y, Assanhou AG, Sooro MA, Baduweh CA, Sun C, Ding Y. Functional Diagnostic and Therapeutic Nanoconstructs for Efficient Probing of Circulating Tumor Cells. ACS APPLIED MATERIALS & INTERFACES 2018; 10:14231-14247. [PMID: 29557165 DOI: 10.1021/acsami.7b17896] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/08/2023]
Abstract
The circulation of tumor cells in peripheral blood is mostly recognized as a prerequisite for cancer progression or systemic invasion, and it correlates with the pivotal hallmark of malignancies known as metastasis. Multiple detection schemes for circulating tumor cells (CTCs) have emerged as the most discerning criteria for monitoring the outcome of anticancer therapy. Therefore, there has been a tremendous increase in the use of robust nanostructured platforms for observation of these mobile tumor cells through various simultaneous diagnosis and treatment regimens developed from conventional techniques. This review seeks to give detailed information about the nature of CTCs as well as techniques for exploiting specific biomarkers to help monitor cancer via detection, capturing, and analysis of unstable tumor cells. We will further discuss nanobased diagnostic interventions and novel platforms which have recently been developed from versatile nanomaterials such as polymer nanocomposites, metal organic frameworks, bioderived nanomaterials and other physically responsive particles with desirable intrinsic and external properties. Herein, we will also include in vivo nanotheranostic platforms which have received a lot of attention because of their enormous clinical potential. In all, this review sums up the general potential of key promising nanoinspired systems as well as other advanced strategies under research and those in clinical use.
Collapse
Affiliation(s)
- Yaw Opoku-Damoah
- Australian Institute for Bioengineering & Nanotechnology , The University of Queensland , St. Lucia , Brisbane, QLD 4072
| | - Assogba G Assanhou
- UFR Pharmacie, Falculté des Sciences de la Santé , Université d'Abomey-Calavi , 01BP188 Cotonou , Benin
| | | | | | | | | |
Collapse
|
47
|
Kulasinghe A, Kenny L, Perry C, Thiery JP, Jovanovic L, Vela I, Nelson C, Punyadeera C. Impact of label-free technologies in head and neck cancer circulating tumour cells. Oncotarget 2018; 7:71223-71234. [PMID: 27655722 PMCID: PMC5342074 DOI: 10.18632/oncotarget.12086] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2016] [Accepted: 08/31/2016] [Indexed: 12/19/2022] Open
Abstract
Background The ability to identify high risk head and neck cancer (HNC) patients with disseminated disease prior to presenting with clinically detectable metastases holds remarkable potential. A fraction of circulating tumour cells (CTCs) are invasive cancer cells which mediate metastasis by intravasation, survival and extravasation from the blood stream to metastatic sites. CTCs have been cleared by the FDA for use as surrogate markers of overall survival and progression free survival for breast, prostate and colorectal cancers using the CellSearch® system. However, the clinical significance of CTCs in head and neck cancer patients has yet to be determined. There has been a significant shift in CTC enrichment platforms, away from exclusively single marker selection, to epitope-independent systems. Methods The aim of this study was to screen advanced stage HNC patients by the CellSearch® platform and utilise two other epitope-independent approaches, ScreenCell® (microfiltration device) and RosetteSep™ (negative enrichment), to determine how a shift to such methodologies would enable CTC enrichment and detection. Results In advanced stage HNC patients, single CTCs were detected in 8/43 (18.6%) on CellSearch®, 13/28 (46.4%) on ScreenCell® and 16/25 (64.0%) by RosetteSep™ (the latter could also detect CTC clusters). Notably, in patients with suspicious lung nodules, too small to biopsy, CTCs were found upon presentation. Moreover, CTCs were readily detected in advanced stage HNC patients. Conclusion The epitope-independent platforms detected higher CTC numbers and clusters. Further studies are needed to ascertain whether CTCs can be used as independent prognostic markers for HNCs.
Collapse
Affiliation(s)
- Arutha Kulasinghe
- The School of Biomedical Sciences, Institute of Health and Biomedical Innovation, Queensland University of Technology, Kelvin Grove, Queensland, Australia
| | - Liz Kenny
- School of Medicine, University of Queensland, Royal Brisbane and Women’s Hospital, Herston, Queensland, Australia
| | - Chris Perry
- Department of Otolaryngology, Princess Alexandra Hospital, Woolloongabba, Queensland, Australia
| | - Jean-Paul Thiery
- Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Lidija Jovanovic
- Australian Prostate Cancer Research Centre-Queensland, Institute of Health and Biomedical Innovation, Queensland University of Technology, Brisbane, Australia
| | - Ian Vela
- Australian Prostate Cancer Research Centre-Queensland, Institute of Health and Biomedical Innovation, Queensland University of Technology, Brisbane, Australia.,Department of Urology, Princess Alexandra Hospital, Wolloongabba, Queensland, Australia
| | - Colleen Nelson
- Australian Prostate Cancer Research Centre-Queensland, Institute of Health and Biomedical Innovation, Queensland University of Technology, Brisbane, Australia
| | - Chamindie Punyadeera
- The School of Biomedical Sciences, Institute of Health and Biomedical Innovation, Queensland University of Technology, Kelvin Grove, Queensland, Australia
| |
Collapse
|
48
|
Austin RG, Huang TJ, Wu M, Armstrong AJ, Zhang T. Clinical utility of non-EpCAM based circulating tumor cell assays. Adv Drug Deliv Rev 2018; 125:132-142. [PMID: 29366804 DOI: 10.1016/j.addr.2018.01.013] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2017] [Revised: 01/14/2018] [Accepted: 01/17/2018] [Indexed: 01/06/2023]
Abstract
Methods enabling the isolation, detection, and characterization of circulating tumor cells (CTCs) in blood have clear potential to facilitate precision medicine approaches in patients with cancer, not only for prognostic purposes but also for prediction of the benefits of specific therapies in oncology. However, current CTC assays, which capture CTCs based on expression of epithelial cell adhesion molecule (EpCAM), fail to capture cells from de-differentiated tumors and carcinomas undergoing loss of the epithelial phenotype during the invasion/metastatic process. To address this limitation, many groups are developing non-EpCAM based CTC assays that incorporate nanotechnology to improve test sensitivity for rare but important cells that may otherwise go undetected, and therefore may improve upon clinical utility. In this review, we outline emerging non-EpCAM based CTC assays utilizing nanotechnology approaches for CTC capture or characterization, including dendrimers, magnetic nanoparticles, gold nanoparticles, negative selection chip or software-based on-slide methods, and nano-scale substrates. In addition, we address challenges that remain for the clinical translation of these platforms.
Collapse
|
49
|
Jan YJ, Chen JF, Zhu Y, Lu YT, Chen SH, Chung H, Smalley M, Huang YW, Dong J, Chen LC, Yu HH, Tomlinson JS, Hou S, Agopian VG, Posadas EM, Tseng HR. NanoVelcro rare-cell assays for detection and characterization of circulating tumor cells. Adv Drug Deliv Rev 2018; 125:78-93. [PMID: 29551650 PMCID: PMC5993593 DOI: 10.1016/j.addr.2018.03.006] [Citation(s) in RCA: 74] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2017] [Revised: 03/08/2018] [Accepted: 03/13/2018] [Indexed: 12/12/2022]
Abstract
Circulating tumor cells (CTCs) are cancer cells shredded from either a primary tumor or a metastatic site and circulate in the blood as the potential cellular origin of metastasis. By detecting and analyzing CTCs, we will be able to noninvasively monitor disease progression in individual cancer patients and obtain insightful information for assessing disease status, thus realizing the concept of "tumor liquid biopsy". However, it is technically challenging to identify CTCs in patient blood samples because of the extremely low abundance of CTCs among a large number of hematologic cells. In order to address this challenge, our research team at UCLA pioneered a unique concept of "NanoVelcro" cell-affinity substrates, in which CTC capture agent-coated nanostructured substrates were utilized to immobilize CTCs with remarkable efficiency. Four generations of NanoVelcro CTC assays have been developed over the past decade for a variety of clinical utilities. The 1st-gen NanoVelcro Chips, composed of a silicon nanowire substrate (SiNS) and an overlaid microfluidic chaotic mixer, were created for CTC enumeration. The 2nd-gen NanoVelcro Chips (i.e., NanoVelcro-LMD), based on polymer nanosubstrates, were developed for single-CTC isolation in conjunction with the use of the laser microdissection (LMD) technique. By grafting thermoresponsive polymer brushes onto SiNS, the 3rd-gen Thermoresponsive NanoVelcro Chips have demonstrated the capture and release of CTCs at 37 and 4 °C respectively, thereby allowing for rapid CTC purification while maintaining cell viability and molecular integrity. Fabricated with boronic acid-grafted conducting polymer-based nanomaterial on chip surface, the 4th-gen NanoVelcro Chips (Sweet chip) were able to purify CTCs with well-preserved RNA transcripts, which could be used for downstream analysis of several cancer specific RNA biomarkers. In this review article, we will summarize the development of the four generations of NanoVelcro CTC assays, and the clinical applications of each generation of devices.
Collapse
Affiliation(s)
- Yu Jen Jan
- Urologic Oncology Program and Uro-Oncology Research Laboratories, Samuel Oschin Comprehensive Cancer Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA; Department of Molecular and Medical Pharmacology, California NanoSystems Institute, Crump Institute for Molecular Imaging, University of California, Los Angeles, Los Angeles, CA, USA
| | - Jie-Fu Chen
- Urologic Oncology Program and Uro-Oncology Research Laboratories, Samuel Oschin Comprehensive Cancer Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Yazhen Zhu
- Department of Molecular and Medical Pharmacology, California NanoSystems Institute, Crump Institute for Molecular Imaging, University of California, Los Angeles, Los Angeles, CA, USA
| | - Yi-Tsung Lu
- Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Szu Hao Chen
- Department of Molecular and Medical Pharmacology, California NanoSystems Institute, Crump Institute for Molecular Imaging, University of California, Los Angeles, Los Angeles, CA, USA
| | - Howard Chung
- Department of Molecular and Medical Pharmacology, California NanoSystems Institute, Crump Institute for Molecular Imaging, University of California, Los Angeles, Los Angeles, CA, USA
| | - Matthew Smalley
- Department of Molecular and Medical Pharmacology, California NanoSystems Institute, Crump Institute for Molecular Imaging, University of California, Los Angeles, Los Angeles, CA, USA; CytoLumina Technologies Corp., Los Angeles, CA, USA
| | - Yen-Wen Huang
- Department of Molecular and Medical Pharmacology, California NanoSystems Institute, Crump Institute for Molecular Imaging, University of California, Los Angeles, Los Angeles, CA, USA; CytoLumina Technologies Corp., Los Angeles, CA, USA
| | - Jiantong Dong
- Department of Molecular and Medical Pharmacology, California NanoSystems Institute, Crump Institute for Molecular Imaging, University of California, Los Angeles, Los Angeles, CA, USA
| | - Li-Ching Chen
- Department of Obstetrics and Gynecology, Cathay General Hospital, Taipei, Taiwan
| | - Hsiao-Hua Yu
- Institute of Chemistry, Academia Sinica, Taipei, Taiwan
| | - James S Tomlinson
- Department of Surgery, University of California, Los Angeles, Los Angeles, CA, USA; Center for Pancreatic Disease, University of California, Los Angeles, Los Angeles, CA, USA; Department of Surgery, Greater Los Angeles Veteran's Affairs Administration, Los Angeles, CA, USA
| | - Shuang Hou
- Department of Surgery, University of California, Los Angeles, Los Angeles, CA, USA
| | - Vatche G Agopian
- Department of Surgery, University of California, Los Angeles, Los Angeles, CA, USA; Liver Transplantation and Hepatobiliary Surgery, University of California, Los Angeles, Los Angeles, CA, USA
| | - Edwin M Posadas
- Urologic Oncology Program and Uro-Oncology Research Laboratories, Samuel Oschin Comprehensive Cancer Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Hsian-Rong Tseng
- Department of Molecular and Medical Pharmacology, California NanoSystems Institute, Crump Institute for Molecular Imaging, University of California, Los Angeles, Los Angeles, CA, USA.
| |
Collapse
|
50
|
Bozekowski JD, Graham AJ, Daugherty PS. High-titer antibody depletion enhances discovery of diverse serum antibody specificities. J Immunol Methods 2018; 455:1-9. [PMID: 29360471 DOI: 10.1016/j.jim.2018.01.003] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2017] [Revised: 12/26/2017] [Accepted: 01/16/2018] [Indexed: 12/15/2022]
Abstract
The human antibody repertoire is a unique repository of information regarding infection, inflammation, and autoimmunity of the past, present, and future. However, antibodies can span vast ranges of concentrations with varying affinities and the repertoire is often heavily polarized by a few species. These complexities lead to difficulties detecting and characterizing low abundance antibody species that may be relevant to disease. We therefore developed a method to selectively remove antibodies from a sample in proportion to the titer of the species prior to analysis, referred to as high-titer depletion (HTD). Peptides from a large random peptide display library were enriched towards binding high-titer antibody species and utilized as binding reagents to deplete the corresponding species from the specimen. HTD enabled the discovery of antibody binding specificities using random peptide library screening with reduced cross-reactivity and background signal and improved coverage of low abundance species. With HTD, three monoclonal antibody species were detected at concentrations at least an order of magnitude lower than without HTD. Additionally, 92 serum antibody specificities were readily discovered from an individual specimen using HTD compared to only 25 specificities without HTD. Parameters affecting the extent of depletion such as the concentration of depleted serum were also adjusted to reproducibly improve the coverage of antibody specificities. These results demonstrate that HTD could be employed for the discovery of rare specificities related to disease and enable extensive characterization of the antibody repertoire. Moreover, the strategy of depletion in proportion to titer could be extended to other applications with complex biological samples to improve discovery.
Collapse
Affiliation(s)
- Joel D Bozekowski
- Department of Chemical Engineering, University of California Santa Barbara, Santa Barbara, CA 93106, USA
| | - Austin J Graham
- Department of Chemical Engineering, University of California Santa Barbara, Santa Barbara, CA 93106, USA
| | - Patrick S Daugherty
- Department of Chemical Engineering, University of California Santa Barbara, Santa Barbara, CA 93106, USA..
| |
Collapse
|