1
|
Vora N, Shekar P, Hanulia T, Esmail M, Patra A, Georgakoudi I. Deep learning-enabled detection of rare circulating tumor cell clusters in whole blood using label-free, flow cytometry. LAB ON A CHIP 2024; 24:2237-2252. [PMID: 38456773 PMCID: PMC11019838 DOI: 10.1039/d3lc00694h] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/10/2023] [Accepted: 01/19/2024] [Indexed: 03/09/2024]
Abstract
Metastatic tumors have poor prognoses for progression-free and overall survival for all cancer patients. Rare circulating tumor cells (CTCs) and rarer circulating tumor cell clusters (CTCCs) are potential biomarkers of metastatic growth, with CTCCs representing an increased risk factor for metastasis. Current detection platforms are optimized for ex vivo detection of CTCs only. Microfluidic chips and size exclusion methods have been proposed for CTCC detection; however, they lack in vivo utility and real-time monitoring capability. Confocal backscatter and fluorescence flow cytometry (BSFC) has been used for label-free detection of CTCCs in whole blood based on machine learning (ML) enabled peak classification. Here, we expand to a deep-learning (DL)-based, peak detection and classification model to detect CTCCs in whole blood data. We demonstrate that DL-based BSFC has a low false alarm rate of 0.78 events per min with a high Pearson correlation coefficient of 0.943 between detected events and expected events. DL-based BSFC of whole blood maintains a detection purity of 72% and a sensitivity of 35.3% for both homotypic and heterotypic CTCCs starting at a minimum size of two cells. We also demonstrate through artificial spiking studies that DL-based BSFC is sensitive to changes in the number of CTCCs present in the samples and does not add variability in detection beyond the expected variability from Poisson statistics. The performance established by DL-based BSFC motivates its use for in vivo detection of CTCCs. Using transfer learning, we additionally validate DL-based BSFC on blood samples from different species and cancer cell types. Further developments of label-free BSFC to enhance throughput could lead to critical applications in the clinical detection of CTCCs and ex vivo isolation of CTCC from whole blood with minimal disruption and processing steps.
Collapse
Affiliation(s)
- Nilay Vora
- Department of Biomedical Engineering, Tufts University, Medford, MA, 02155, USA.
| | - Prashant Shekar
- Department of Mathematics, Embry-Riddle Aeronautical University, Daytona Beach, FL, 32114, USA
| | - Taras Hanulia
- Department of Biomedical Engineering, Tufts University, Medford, MA, 02155, USA.
- Institute of Physics, National Academy of Sciences of Ukraine, Kyiv, Ukraine
| | - Michael Esmail
- Tufts Comparative Medicine Services, Tufts University, Medford, MA, 02155, USA
| | - Abani Patra
- Data Intensive Studies Center, Tufts University, Medford, MA, 02155, USA
| | - Irene Georgakoudi
- Department of Biomedical Engineering, Tufts University, Medford, MA, 02155, USA.
| |
Collapse
|
2
|
Jiang X, Wang S, Liang Q, Liu Y, Liu L. Unraveling the multifaceted role of EpCAM in colorectal cancer: an integrated review of its function and interplay with non-coding RNAs. Med Oncol 2023; 41:35. [PMID: 38151631 DOI: 10.1007/s12032-023-02273-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2023] [Accepted: 11/27/2023] [Indexed: 12/29/2023]
Abstract
The epithelial cell adhesion molecule (EpCAM) is a critical glycoprotein involved in cell cycle progression, proliferation, differentiation, migration, and immune evasion. Its role as a target for bispecific antibodies has shown promise in annihilating cancer cells. EpCAM's potential as a biomarker for tumor-initiating cells, characterized by self-renewal and tumorigenic capabilities, underscores its value in early cancer detection, immunotherapy, and targeted drug delivery. While EpCAM monotherapies have been met with limited success, bispecific antibodies targeting both EpCAM and other proteins have exhibited encouraging results in colorectal cancer (CRC) research. The integration of EpCAM-directed nanotechnology in drug delivery systems has emerged as a pivotal innovation in CRC treatment. Moreover, developing chimeric antigen receptor (CAR) T-cell and CAR natural killer (NK) cell therapies opens promising therapeutic avenues for EpCAM-positive CRC patients. Although preliminary, this review sets the stage for future advances. Additionally, this study advances our understanding of the role of non-coding RNAs in CRC, which may be pivotal in gene regulation and could provide insights into the molecular underpinning. The findings suggest that lncRNA, miRNA, and circRNA could serve as novel therapeutic targets or biomarkers, further enriching the landscape of CRC diagnostics and therapeutics.
Collapse
Affiliation(s)
- Xingyu Jiang
- Department of Oncology, The First Affiliated Hospital of Nanjing Medical University, 300 Guangzhou Road, Nanjing, 210029, Jiangsu, People's Republic of China
| | - Sumeng Wang
- Department of Oncology, The First Affiliated Hospital of Nanjing Medical University, 300 Guangzhou Road, Nanjing, 210029, Jiangsu, People's Republic of China
| | - Qi Liang
- Department of Oncology, The First Affiliated Hospital of Nanjing Medical University, 300 Guangzhou Road, Nanjing, 210029, Jiangsu, People's Republic of China
| | - Yiqian Liu
- Department of Oncology, The First Affiliated Hospital of Nanjing Medical University, 300 Guangzhou Road, Nanjing, 210029, Jiangsu, People's Republic of China.
| | - Lingxiang Liu
- Department of Oncology, The First Affiliated Hospital of Nanjing Medical University, 300 Guangzhou Road, Nanjing, 210029, Jiangsu, People's Republic of China.
| |
Collapse
|
3
|
Vora N, Shekar P, Esmail M, Patra A, Georgakoudi I. Deep Learning-Enabled, Detection of Rare Circulating Tumor Cell Clusters in Whole Blood Using Label-free, Flow Cytometry. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.08.01.551485. [PMID: 37577660 PMCID: PMC10418242 DOI: 10.1101/2023.08.01.551485] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/15/2023]
Abstract
Metastatic tumors have poor prognoses for progression-free and overall survival for all cancer patients. Rare circulating tumor cells (CTCs) and rarer circulating tumor cell clusters (CTCCs) are potential biomarkers of metastatic growth, with CTCCs representing an increased risk factor for metastasis. Current detection platforms are optimized for ex vivo detection of CTCs only. Microfluidic chips and size exclusion methods have been proposed for CTCC detection; however, they lack in vivo utility and real-time monitoring capability. Confocal backscatter and fluorescence flow cytometry (BSFC) has been used for label-free detection of CTCCs in whole blood based on machine learning (ML) enabled peak classification. Here, we expand to a deep-learning (DL) -based, peak detection and classification model to detect CTCCs in whole blood data. We demonstrate that DL-based BSFC has a low false alarm rate of 0.78 events/min with a high Pearson correlation coefficient of 0.943 between detected events and expected events. DL-based BSFC of whole blood maintains a detection purity of 72% and a sensitivity of 35.3% for both homotypic and heterotypic CTCCs starting at a minimum size of two cells. We also demonstrate through artificial spiking studies that DL-based BSFC is sensitive to changes in the number of CTCCs present in the samples and does not add variability in detection beyond the expected variability from Poisson statistics. The performance established by DL-based BSFC motivates its use for in vivo detection of CTCCs. Further developments of label-free BSFC to enhance throughput could lead to critical applications in the clinical detection of CTCCs and ex vivo isolation of CTCC from whole blood with minimal disruption and processing steps.
Collapse
Affiliation(s)
- Nilay Vora
- Department of Biomedical Engineering, Tufts University, Medford, MA 02155, USA
| | - Prashant Shekar
- Department of Mathematics, Embry-Riddle Aeronautical University, Daytona Beach, FL, 32114, USA
| | - Michael Esmail
- Tufts Comparative Medicine Services, Tufts University, Medford, MA, 02155, USA
- # Current Affiliation: University of Massachusetts Amherst Animal Care Services, University of Massachusetts Amherst, Amherst, MA 01003, USA
| | - Abani Patra
- Data Intensive Studies Center, Tufts University, Medford, MA 02155, USA
- Department of Mathematics, Tufts University, Medford, MA 02155, USA
| | - Irene Georgakoudi
- Department of Biomedical Engineering, Tufts University, Medford, MA 02155, USA
| |
Collapse
|
4
|
Farhang Ghahremani M, Seto KKY, Cho W, Miller MC, Smith P, Englert DF. Novel method for highly multiplexed gene expression profiling of circulating tumor cells (CTCs) captured from the blood of women with metastatic breast cancer. J Transl Med 2023; 21:414. [PMID: 37365600 DOI: 10.1186/s12967-023-04242-z] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2022] [Accepted: 06/02/2023] [Indexed: 06/28/2023] Open
Abstract
BACKGROUND Enumeration of circulating tumor cells (CTCs) has proven clinical significance for monitoring patients with metastatic cancers. Multiplexed gene expression profiling of CTCs is a potential tool for assessing disease status and monitoring treatment response. The Parsortix® technology enables the capture and harvest of CTCs from blood based on cell size and deformability. The HyCEAD™ (Hybrid Capture Enrichment Amplification and Detection) assay enables simultaneous amplification of short amplicons for up to 100 mRNA targets, and the Ziplex™ instrument quantifies the amplicons for highly sensitive gene expression profiling down to single cell levels. The aim of the study was to functionally assess this system. METHODS The HyCEAD/Ziplex platform was used to quantify the expression levels for 72 genes using as little as 20 pg of total RNA or a single cultured tumor cell. Assay performance was evaluated using cells or total RNA spiked into Parsortix harvests of healthy donor blood. The assay was also evaluated using total RNA obtained from Parsortix harvests of blood from metastatic breast cancer (MBC) patients or healthy volunteers (HVs). RESULTS Using genes with low expression in WBC RNA and/or in unspiked Parsortix harvests from HVs, the assay distinguished between the different breast cancer and ovarian cancer cell lines with as little as 20 pg of total RNA (equivalent to a single cell) in the presence of 1 ng of WBC RNA. Single cultured cells spiked into Parsortix harvests from 10 mL of HV blood were also detected and distinguished from each other. CVs from repeatability experiments were less than 20%. Hierarchical clustering of clinical samples differentiated most MBC patients from HVs. CONCLUSION HyCEAD/Ziplex provided sensitive quantification of expression of 72 genes from 20 pg of total RNA from cultured tumor cell lines or from single cultured tumor cells spiked into lysates from Parsortix harvests of HV blood. The HyCEAD/Ziplex platform enables the quantification of selected genes in the presence of residual nucleated blood cells in Parsortix harvests. The HyCEAD/Ziplex platform is an effective tool for multiplexed molecular characterization of mRNA in small numbers of tumor cells harvested from blood.
Collapse
Affiliation(s)
| | | | | | - Michael Craig Miller
- Clinical Development, ANGLE North America, Inc., Plymouth Meeting, Pennsylvania, USA
| | - Paul Smith
- ANGLE Biosciences Inc., Toronto, ON, Canada
| | | |
Collapse
|
5
|
Morganti S, Parsons HA, Lin NU, Grinshpun A. Liquid biopsy for brain metastases and leptomeningeal disease in patients with breast cancer. NPJ Breast Cancer 2023; 9:43. [PMID: 37225714 DOI: 10.1038/s41523-023-00550-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2023] [Accepted: 05/12/2023] [Indexed: 05/26/2023] Open
Abstract
A significant subset of patients with metastatic breast cancer develops brain metastasis. As efficacy of systemic therapies has improved and patients live longer with metastatic breast cancer, the incidence of breast cancer brain metastases has increased. Brain metastases pose a clinical challenge in diagnosis, treatment, and monitoring across all breast cancer subtypes, and better tools are needed. Liquid biopsy, which enables minimally invasive sampling of a patient's cancer, has the potential to shed light on intra-cranial tumor biology and to improve patient care by enabling therapy tailoring. Here we review current evidence for the clinical validity of liquid biopsy in patients with breast cancer brain metastases, with a focus on circulating tumor cells and circulating tumor DNA.
Collapse
Affiliation(s)
- Stefania Morganti
- Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
- Breast Oncology Program, Dana-Farber Brigham Cancer Center, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
- Broad Institute of MIT and Harvard, Boston, MA, USA
| | - Heather A Parsons
- Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
- Breast Oncology Program, Dana-Farber Brigham Cancer Center, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
- Broad Institute of MIT and Harvard, Boston, MA, USA
| | - Nancy U Lin
- Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
- Breast Oncology Program, Dana-Farber Brigham Cancer Center, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
| | - Albert Grinshpun
- Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA.
- Breast Oncology Program, Dana-Farber Brigham Cancer Center, Boston, MA, USA.
- Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
6
|
Bates M, Mohamed BM, Ward MP, Kelly TE, O'Connor R, Malone V, Brooks R, Brooks D, Selemidis S, Martin C, O'Toole S, O'Leary JJ. Circulating tumour cells: The Good, the Bad and the Ugly. Biochim Biophys Acta Rev Cancer 2023; 1878:188863. [PMID: 36796527 DOI: 10.1016/j.bbcan.2023.188863] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2022] [Revised: 01/06/2023] [Accepted: 01/21/2023] [Indexed: 02/17/2023]
Abstract
This review is an overview of the current knowledge regarding circulating tumour cells (CTCs), which are potentially the most lethal type of cancer cell, and may be a key component of the metastatic cascade. The clinical utility of CTCs (the "Good"), includes their diagnostic, prognostic, and therapeutic potential. Conversely, their complex biology (the "Bad"), including the existence of CD45+/EpCAM+ CTCs, adds insult to injury regarding their isolation and identification, which in turn hampers their clinical translation. CTCs are capable of forming microemboli composed of both non-discrete phenotypic populations such as mesenchymal CTCs and homotypic and heterotypic clusters which are poised to interact with other cells in the circulation, including immune cells and platelets, which may increase their malignant potential. These microemboli (the "Ugly") represent a prognostically important CTC subset, however, phenotypic EMT/MET gradients bring additional complexities to an already challenging situation.
Collapse
Affiliation(s)
- Mark Bates
- Department of Histopathology, Trinity College Dublin, Dublin 2, Ireland; Emer Casey Molecular Pathology Research Laboratory, Coombe Women & Infants University Hospital, Dublin 8, Ireland; Trinity St James's Cancer Institute, Dublin 8, Ireland.
| | - Bashir M Mohamed
- Department of Histopathology, Trinity College Dublin, Dublin 2, Ireland; Emer Casey Molecular Pathology Research Laboratory, Coombe Women & Infants University Hospital, Dublin 8, Ireland; Trinity St James's Cancer Institute, Dublin 8, Ireland
| | - Mark P Ward
- Department of Histopathology, Trinity College Dublin, Dublin 2, Ireland; Emer Casey Molecular Pathology Research Laboratory, Coombe Women & Infants University Hospital, Dublin 8, Ireland; Trinity St James's Cancer Institute, Dublin 8, Ireland
| | - Tanya E Kelly
- Department of Histopathology, Trinity College Dublin, Dublin 2, Ireland; Emer Casey Molecular Pathology Research Laboratory, Coombe Women & Infants University Hospital, Dublin 8, Ireland; Trinity St James's Cancer Institute, Dublin 8, Ireland
| | - Roisin O'Connor
- Department of Histopathology, Trinity College Dublin, Dublin 2, Ireland; Emer Casey Molecular Pathology Research Laboratory, Coombe Women & Infants University Hospital, Dublin 8, Ireland; Trinity St James's Cancer Institute, Dublin 8, Ireland; Department of Pathology, Coombe Women & Infants University Hospital, Dublin 8, Ireland
| | - Victoria Malone
- Department of Histopathology, Trinity College Dublin, Dublin 2, Ireland; Emer Casey Molecular Pathology Research Laboratory, Coombe Women & Infants University Hospital, Dublin 8, Ireland; Trinity St James's Cancer Institute, Dublin 8, Ireland; Department of Pathology, Coombe Women & Infants University Hospital, Dublin 8, Ireland
| | - Robert Brooks
- Cancer Research Institute, University of South Australia, Adelaide, SA 5001, Australia
| | - Doug Brooks
- Department of Histopathology, Trinity College Dublin, Dublin 2, Ireland; Trinity St James's Cancer Institute, Dublin 8, Ireland; Cancer Research Institute, University of South Australia, Adelaide, SA 5001, Australia
| | - Stavros Selemidis
- School of Health and Biomedical Sciences, Royal Melbourne Institute of Technology, Bundoora, VIC 3083, Australia
| | - Cara Martin
- Department of Histopathology, Trinity College Dublin, Dublin 2, Ireland; Emer Casey Molecular Pathology Research Laboratory, Coombe Women & Infants University Hospital, Dublin 8, Ireland; Trinity St James's Cancer Institute, Dublin 8, Ireland; Department of Pathology, Coombe Women & Infants University Hospital, Dublin 8, Ireland
| | - Sharon O'Toole
- Department of Histopathology, Trinity College Dublin, Dublin 2, Ireland; Emer Casey Molecular Pathology Research Laboratory, Coombe Women & Infants University Hospital, Dublin 8, Ireland; Trinity St James's Cancer Institute, Dublin 8, Ireland; Department of Obstetrics and Gynaecology, Trinity College Dublin, Dublin 2, Ireland
| | - John J O'Leary
- Department of Histopathology, Trinity College Dublin, Dublin 2, Ireland; Emer Casey Molecular Pathology Research Laboratory, Coombe Women & Infants University Hospital, Dublin 8, Ireland; Trinity St James's Cancer Institute, Dublin 8, Ireland; Department of Pathology, Coombe Women & Infants University Hospital, Dublin 8, Ireland
| |
Collapse
|
7
|
Tretyakova MS, Menyailo ME, Schegoleva AA, Bokova UA, Larionova IV, Denisov EV. Technologies for Viable Circulating Tumor Cell Isolation. Int J Mol Sci 2022; 23:ijms232415979. [PMID: 36555625 PMCID: PMC9788311 DOI: 10.3390/ijms232415979] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2022] [Revised: 12/07/2022] [Accepted: 12/13/2022] [Indexed: 12/23/2022] Open
Abstract
The spread of tumor cells throughout the body by traveling through the bloodstream is a critical step in metastasis, which continues to be the main cause of cancer-related death. The detection and analysis of circulating tumor cells (CTCs) is important for understanding the biology of metastasis and the development of antimetastatic therapy. However, the isolation of CTCs is challenging due to their high heterogeneity and low representation in the bloodstream. Different isolation methods have been suggested, but most of them lead to CTC damage. However, viable CTCs are an effective source for developing preclinical models to perform drug screening and model the metastatic cascade. In this review, we summarize the available literature on methods for isolating viable CTCs based on different properties of cells. Particular attention is paid to the importance of in vitro and in vivo models obtained from CTCs. Finally, we emphasize the current limitations in CTC isolation and suggest potential solutions to overcome them.
Collapse
Affiliation(s)
- Maria S. Tretyakova
- Laboratory of Cancer Progression Biology, Cancer Research Institute, Tomsk National Research Medical Center, Russian Academy of Sciences, 634009 Tomsk, Russia
| | - Maxim E. Menyailo
- Laboratory of Cancer Progression Biology, Cancer Research Institute, Tomsk National Research Medical Center, Russian Academy of Sciences, 634009 Tomsk, Russia
- Single Cell Biology Laboratory, Research Institute of Molecular and Cellular Medicine, Peoples’ Friendship University of Russia (RUDN University), 117198 Moscow, Russia
| | - Anastasia A. Schegoleva
- Laboratory of Cancer Progression Biology, Cancer Research Institute, Tomsk National Research Medical Center, Russian Academy of Sciences, 634009 Tomsk, Russia
- Single Cell Biology Laboratory, Research Institute of Molecular and Cellular Medicine, Peoples’ Friendship University of Russia (RUDN University), 117198 Moscow, Russia
| | - Ustinia A. Bokova
- Laboratory of Cancer Progression Biology, Cancer Research Institute, Tomsk National Research Medical Center, Russian Academy of Sciences, 634009 Tomsk, Russia
| | - Irina V. Larionova
- Laboratory of Cancer Progression Biology, Cancer Research Institute, Tomsk National Research Medical Center, Russian Academy of Sciences, 634009 Tomsk, Russia
| | - Evgeny V. Denisov
- Laboratory of Cancer Progression Biology, Cancer Research Institute, Tomsk National Research Medical Center, Russian Academy of Sciences, 634009 Tomsk, Russia
- Single Cell Biology Laboratory, Research Institute of Molecular and Cellular Medicine, Peoples’ Friendship University of Russia (RUDN University), 117198 Moscow, Russia
- Correspondence: ; Tel./Fax: +7-3822-282676 (ext. 3375)
| |
Collapse
|
8
|
García-Pardo M, Makarem M, Li JJN, Kelly D, Leighl NB. Integrating circulating-free DNA (cfDNA) analysis into clinical practice: opportunities and challenges. Br J Cancer 2022; 127:592-602. [PMID: 35347327 PMCID: PMC9381753 DOI: 10.1038/s41416-022-01776-9] [Citation(s) in RCA: 72] [Impact Index Per Article: 24.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2021] [Revised: 02/16/2022] [Accepted: 02/22/2022] [Indexed: 12/12/2022] Open
Abstract
In the current era of precision medicine, the identification of genomic alterations has revolutionised the management of patients with solid tumours. Recent advances in the detection and characterisation of circulating tumour DNA (ctDNA) have enabled the integration of liquid biopsy into clinical practice for molecular profiling. ctDNA has also emerged as a promising biomarker for prognostication, monitoring disease response, detection of minimal residual disease and early diagnosis. In this Review, we discuss current and future clinical applications of ctDNA primarily in non-small cell lung cancer in addition to other solid tumours.
Collapse
Affiliation(s)
- Miguel García-Pardo
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada
| | - Maisam Makarem
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada
| | - Janice J N Li
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada
| | - Deirdre Kelly
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada
| | - Natasha B Leighl
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada.
| |
Collapse
|
9
|
Ju S, Chen C, Zhang J, Xu L, Zhang X, Li Z, Chen Y, Zhou J, Ji F, Wang L. Detection of circulating tumor cells: opportunities and challenges. Biomark Res 2022; 10:58. [PMID: 35962400 PMCID: PMC9375360 DOI: 10.1186/s40364-022-00403-2] [Citation(s) in RCA: 76] [Impact Index Per Article: 25.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2022] [Accepted: 07/26/2022] [Indexed: 11/25/2022] Open
Abstract
Circulating tumor cells (CTCs) are cells that shed from a primary tumor and travel through the bloodstream. Studying the functional and molecular characteristics of CTCs may provide in-depth knowledge regarding highly lethal tumor diseases. Researchers are working to design devices and develop analytical methods that can capture and detect CTCs in whole blood from cancer patients with improved sensitivity and specificity. Techniques using whole blood samples utilize physical prosperity, immunoaffinity or a combination of the above methods and positive and negative enrichment during separation. Further analysis of CTCs is helpful in cancer monitoring, efficacy evaluation and designing of targeted cancer treatment methods. Although many advances have been achieved in the detection and molecular characterization of CTCs, several challenges still exist that limit the current use of this burgeoning diagnostic approach. In this review, a brief summary of the biological characterization of CTCs is presented. We focus on the current existing CTC detection methods and the potential clinical implications and challenges of CTCs. We also put forward our own views regarding the future development direction of CTCs.
Collapse
Affiliation(s)
- Siwei Ju
- Department of Surgical Oncology, The Sir Run Run Shaw Hospital Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang, Hangzhou, China
- Key Laboratory of Cancer Prevention and Intervention, Ministry of Education, Zhejiang, Hangzhou, China
| | - Cong Chen
- Department of Surgical Oncology, The Sir Run Run Shaw Hospital Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang, Hangzhou, China
- Key Laboratory of Cancer Prevention and Intervention, Ministry of Education, Zhejiang, Hangzhou, China
| | - Jiahang Zhang
- Department of Surgical Oncology, The Sir Run Run Shaw Hospital Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang, Hangzhou, China
- Key Laboratory of Cancer Prevention and Intervention, Ministry of Education, Zhejiang, Hangzhou, China
| | - Lin Xu
- Department of Surgical Oncology, The Sir Run Run Shaw Hospital Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang, Hangzhou, China
- Key Laboratory of Cancer Prevention and Intervention, Ministry of Education, Zhejiang, Hangzhou, China
| | - Xun Zhang
- Department of Surgical Oncology, The Sir Run Run Shaw Hospital Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang, Hangzhou, China
- Key Laboratory of Cancer Prevention and Intervention, Ministry of Education, Zhejiang, Hangzhou, China
| | - Zhaoqing Li
- Department of Surgical Oncology, The Sir Run Run Shaw Hospital Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang, Hangzhou, China
- Key Laboratory of Cancer Prevention and Intervention, Ministry of Education, Zhejiang, Hangzhou, China
| | - Yongxia Chen
- Department of Surgical Oncology, The Sir Run Run Shaw Hospital Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang, Hangzhou, China
- Key Laboratory of Cancer Prevention and Intervention, Ministry of Education, Zhejiang, Hangzhou, China
| | - Jichun Zhou
- Department of Surgical Oncology, The Sir Run Run Shaw Hospital Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang, Hangzhou, China
- Key Laboratory of Cancer Prevention and Intervention, Ministry of Education, Zhejiang, Hangzhou, China
| | - Feiyang Ji
- Department of Surgical Oncology, The Sir Run Run Shaw Hospital Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang, Hangzhou, China.
- Key Laboratory of Cancer Prevention and Intervention, Ministry of Education, Zhejiang, Hangzhou, China.
| | - Linbo Wang
- Department of Surgical Oncology, The Sir Run Run Shaw Hospital Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang, Hangzhou, China.
- Key Laboratory of Cancer Prevention and Intervention, Ministry of Education, Zhejiang, Hangzhou, China.
| |
Collapse
|
10
|
Mitchell MI, Ma J, Carter CL, Loudig O. Circulating Exosome Cargoes Contain Functionally Diverse Cancer Biomarkers: From Biogenesis and Function to Purification and Potential Translational Utility. Cancers (Basel) 2022; 14:3350. [PMID: 35884411 PMCID: PMC9318395 DOI: 10.3390/cancers14143350] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2022] [Revised: 07/01/2022] [Accepted: 07/07/2022] [Indexed: 12/12/2022] Open
Abstract
Although diagnostic and therapeutic treatments of cancer have tremendously improved over the past two decades, the indolent nature of its symptoms has made early detection challenging. Thus, inter-disciplinary (genomic, transcriptomic, proteomic, and lipidomic) research efforts have been focused on the non-invasive identification of unique "silver bullet" cancer biomarkers for the design of ultra-sensitive molecular diagnostic assays. Circulating tumor biomarkers, such as CTCs and ctDNAs, which are released by tumors in the circulation, have already demonstrated their clinical utility for the non-invasive detection of certain solid tumors. Considering that exosomes are actively produced by all cells, including tumor cells, and can be found in the circulation, they have been extensively assessed for their potential as a source of circulating cell-specific biomarkers. Exosomes are particularly appealing because they represent a stable and encapsulated reservoir of active biological compounds that may be useful for the non-invasive detection of cancer. T biogenesis of these extracellular vesicles is profoundly altered during carcinogenesis, but because they harbor unique or uniquely combined surface proteins, cancer biomarker studies have been focused on their purification from biofluids, for the analysis of their RNA, DNA, protein, and lipid cargoes. In this review, we evaluate the biogenesis of normal and cancer exosomes, provide extensive information on the state of the art, the current purification methods, and the technologies employed for genomic, transcriptomic, proteomic, and lipidomic evaluation of their cargoes. Our thorough examination of the literature highlights the current limitations and promising future of exosomes as a liquid biopsy for the identification of circulating tumor biomarkers.
Collapse
Affiliation(s)
- Megan I Mitchell
- Center for Discovery and Innovation, Hackensack Meridian Health, Nutley, NJ 07110, USA
| | - Junfeng Ma
- Department of Oncology, Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington, DC 20007, USA
| | - Claire L Carter
- Center for Discovery and Innovation, Hackensack Meridian Health, Nutley, NJ 07110, USA
| | - Olivier Loudig
- Center for Discovery and Innovation, Hackensack Meridian Health, Nutley, NJ 07110, USA
| |
Collapse
|
11
|
Vora N, Shekhar P, Esmail M, Patra A, Georgakoudi I. Label-free flow cytometry of rare circulating tumor cell clusters in whole blood. Sci Rep 2022; 12:10721. [PMID: 35750889 PMCID: PMC9232518 DOI: 10.1038/s41598-022-14003-5] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2021] [Accepted: 05/31/2022] [Indexed: 11/09/2022] Open
Abstract
Circulating tumor cell clusters (CTCCs) are rare cellular events found in the blood stream of metastatic tumor patients. Despite their scarcity, they represent an increased risk for metastasis. Label-free detection methods of these events remain primarily limited to in vitro microfluidic platforms. Here, we expand on the use of confocal backscatter and fluorescence flow cytometry (BSFC) for label-free detection of CTCCs in whole blood using machine learning for peak detection/classification. BSFC uses a custom-built flow cytometer with three excitation wavelengths (405 nm, 488 nm, and 633 nm) and five detectors to detect CTCCs in whole blood based on corresponding scattering and fluorescence signals. In this study, detection of CTCC-associated GFP fluorescence is used as the ground truth to assess the accuracy of endogenous back-scattered light-based CTCC detection in whole blood. Using a machine learning model for peak detection/classification, we demonstrated that the combined use of backscattered signals at the three wavelengths enable detection of ~ 93% of all CTCCs larger than two cells with a purity of > 82% and an overall accuracy of > 95%. The high level of performance established through BSFC and machine learning demonstrates the potential for label-free detection and monitoring of CTCCs in whole blood. Further developments of label-free BSFC to enhance throughput could lead to important applications in the isolation of CTCCs in whole blood with minimal disruption and ultimately their detection in vivo.
Collapse
Affiliation(s)
- Nilay Vora
- Department of Biomedical Engineering, Tufts University, Medford, MA, 02155, USA
| | - Prashant Shekhar
- Department of Mathematics, Embry-Riddle Aeronautical University, Daytona Beach, FL, 32114, USA
| | - Michael Esmail
- Tufts Comparative Medicine Services, Tufts University, Medford, MA, 02155, USA
| | - Abani Patra
- Department of Computer Science, Tufts University, Medford, MA, 02155, USA
| | - Irene Georgakoudi
- Department of Biomedical Engineering, Tufts University, Medford, MA, 02155, USA.
| |
Collapse
|
12
|
Kalita-de Croft P, Joshi V, Saunus JM, Lakhani SR. Emerging Biomarkers for Diagnosis, Prevention and Treatment of Brain Metastases-From Biology to Clinical Utility. Diseases 2022; 10:11. [PMID: 35225863 PMCID: PMC8884016 DOI: 10.3390/diseases10010011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2021] [Revised: 01/18/2022] [Accepted: 01/27/2022] [Indexed: 11/17/2022] Open
Abstract
Primary malignancies of the lung, skin (melanoma), and breast have higher propensity for metastatic spread to the brain. Advances in molecular tumour profiling have aided the development of targeted therapies, stereotactic radiotherapy, and immunotherapy, which have led to some improvement in patient outcomes; however, the overall prognosis remains poor. Continued research to identify new prognostic and predictive biomarkers is necessary to further impact patient outcomes, as this will enable better risk stratification at the point of primary cancer diagnosis, earlier detection of metastatic deposits (for example, through surveillance), and more effective systemic treatments. Brain metastases exhibit considerable inter- and intratumoural heterogeneity-apart from distinct histology, treatment history and other clinical factors, the metastatic brain tumour microenvironment is incredibly variable both in terms of subclonal diversity and cellular composition. This review discusses emerging biomarkers; specifically, the biological context and potential clinical utility of tumour tissue biomarkers, circulating tumour cells, extracellular vesicles, and circulating tumour DNA.
Collapse
Affiliation(s)
- Priyakshi Kalita-de Croft
- UQ Centre for Clinical Research, The University of Queensland Faculty of Medicine, Herston, QLD 4029, Australia; (V.J.); (J.M.S.)
| | - Vaibhavi Joshi
- UQ Centre for Clinical Research, The University of Queensland Faculty of Medicine, Herston, QLD 4029, Australia; (V.J.); (J.M.S.)
| | - Jodi M. Saunus
- UQ Centre for Clinical Research, The University of Queensland Faculty of Medicine, Herston, QLD 4029, Australia; (V.J.); (J.M.S.)
| | - Sunil R. Lakhani
- UQ Centre for Clinical Research, The University of Queensland Faculty of Medicine, Herston, QLD 4029, Australia; (V.J.); (J.M.S.)
- Pathology Queensland, The Royal Brisbane and Women’s Hospital Herston, Herston, QLD 4029, Australia
| |
Collapse
|
13
|
Shliakhtunou YA, Siamionau VM, Pobyarzhin VV. Transcription phenotype of circulating tumor cells in non-metastatic breast cancer. Carcinogenesis 2021; 43:21-27. [PMID: 34919653 DOI: 10.1093/carcin/bgab112] [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: 09/15/2021] [Revised: 11/16/2021] [Accepted: 11/25/2021] [Indexed: 11/14/2022] Open
Abstract
The presented research is relevant, as breast cancer is the most commonly diagnosed cancer in the female population worldwide, with the exception of skin cancer. The aim of this article is to study the transcription phenotype of circulating tumor cells in non-metastatic breast cancer. The transcriptional phenotype of circulating tumor cells (CTCs) was studied using real-time polymerase chain reaction (PCR). Three-year OS was 79.2, and 90.8 without the expression with p Log-Rank=0.04. Independent prognostic factors for the recurrence of disease include the presence of CTCs expressing BIRC5 genes and ABC transporter genes in the peripheral blood before the start of special treatment for resectable breast cancer, as well as the preservation of CTCs per se after completion of special anticancer therapy. In patients with breast cancer stage I-IIIC, circulating tumor cells before special treatment have significant heterogeneity, manifested by a different transcriptional phenotype, including both actively growing and stem tumor cells, and cells at the epithelial-to-mesenchymal transition.
Collapse
Affiliation(s)
- Yauheni A Shliakhtunou
- Department of Oncology and Courses of Radiation Diagnostics and Radiation Therapy, Vitebsk State Order of Peoples' Friendship Medical University, Vitebsk, Republic of Belarus
| | - Valery M Siamionau
- Department of Infectious Diseases, Vitebsk State Order of Peoples' Friendship Medical University, Vitebsk, Republic of Belarus
| | - Vyacheslau V Pobyarzhin
- Department of Medical Biology and General Genetics, Vitebsk State Order of Peoples' Friendship Medical University, Vitebsk, Republic of Belarus
| |
Collapse
|
14
|
Sanches SM, Braun AC, Calsavara VF, Barbosa PNVP, Chinen LTD. Comparison of hormonal receptor expression and HER2 status between circulating tumor cells and breast cancer metastases. Clinics (Sao Paulo) 2021; 76:e2971. [PMID: 34644733 PMCID: PMC8478133 DOI: 10.6061/clinics/2021/e2971] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Accepted: 08/19/2021] [Indexed: 02/04/2023] Open
Abstract
OBJECTIVES Breast cancer (BC) is the most common neoplasm in women. Biopsy of metastatic lesions is recommended to confirm estrogen receptor (ER), progesterone receptor (PR), and human epidermal growth factor receptor 2 (HER2) status as there are discrepancies in these patterns between primary tumors and metastases in up to 40% of the cases. Circulating tumor cells (CTCs) are related to BC outcomes and could potentially be an alternative to the invasive procedures of metastasis rebiopsy. ISET® technology is not currently employed to detect CTCs in patients with BC. Emerging data support that the characterization of CTC protein expression can refine its prognostic value. Transforming growth factor (TGF)-β plays a role in BC progression and invasiveness. Thus, in this study, we aimed to compare ER, PR, and HER2 expression in primary tumors, CTCs, and metastases and evaluate TGF-β type 1 receptor (TGF-β RI) expression in CTCs as prognostic factor for progression free survival (PFS) and overall survival (OS). METHODS This prospective study was conducted at the A.C. Camargo Cancer Center, Brazil. Blood samples were processed in ISET® (Isolation by SizE of Tumors, Rarecells, France) before computed tomography-guided biopsy of suspected metastatic lesions. Protein expression levels in CTCs were compared to those in primary tumors/metastases (medical records). RESULTS Of the 39 patients initially included, 27 underwent both biopsies of metastases and blood collection and were considered for analysis. The concordance rates for ER, PR, and HER2 expression between primary tumors and metastases were high. No loss of HER2 expression at any metastasis site and retention of the same pattern of protein expression in all triple-negative (TN) tumors (92.5%, 81.5% and 96.2% respectively) (p<0.0001) was observed. When metastases/CTCs were classified as TN/non-TN, CTCs showed high specificity (93%), accuracy (84.2%), and negative predictive value (88%). The median OS of patients without TGF-β RI expression in CTCs was 42.6 versus 20.8 months for TGF-β RI expression-positive ones (p>0.05). CONCLUSION The role of CTCs detected by ISET has not yet been established in BC. Here, we suggest that this methodology may be useful to evaluate metastasis in non-TN cases as well as TGF-β RI expression in CTCs, which may impact patient survival. Due to sample limitations, future studies must focus on specific BC subtypes and an expansion of the cohort.
Collapse
|
15
|
Katopodis P, Anikin V, Kishore U, Carter T, Hall M, Asadi N, Polychronis A, Karteris E. Circulating tumour cells and circulating cell-free DNA in patients with lung cancer: a comparison between thoracotomy and video-assisted thoracoscopic surgery. BMJ Open Respir Res 2021; 8:8/1/e000917. [PMID: 34493540 PMCID: PMC8424856 DOI: 10.1136/bmjresp-2021-000917] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2021] [Accepted: 08/07/2021] [Indexed: 11/03/2022] Open
Abstract
INTRODUCTION The type of lung cancer surgery impacts on tumour manipulation during surgery and may drive dissemination of cancer cells into the vasculature, thus facilitating metastatic spread. The aim of this study was to investigate the impact of surgically induced trauma using peripheral blood from preoperative and postoperative patients with non-small cell lung cancer (NSCLC) undergoing thoracotomy or video-assisted thoracoscopic surgery (VATS) resection. METHODS Imaging flow cytometry was used to measure circulating cancer-associated cells (CCs). Circulating cell-free DNA (ccfDNA) isolation was performed using Promega dsDNA HS Assay Kit. DNA integrity measurements were calculated by the ALU247 to ALU115 ratio and cytokine levels measured using the Luminex screening assay. RESULTS CCs were increased in postoperative blood samples in 54 patients with NSCLC. Patients who underwent thoracotomy instead of VATS had higher numbers of EpCAM (p=0.004) and PanCK-labelled (p=0.03) CCs postoperatively. ccfDNA and DNA integrity index were also significantly increased in postoperative samples (p=0.0009 and p=0.04), with concomitant increase in interleukin 6 and interleukin 10 levels in the same cohorts (p=0.0004 and p=0.034, respectively). CONCLUSIONS In this study we have shown the potential clinical utility of several biomarkers from liquid biopsies to guide perioperative management, as well as provide a snapshot of the type of surgical resection in terms of circulating tumour cell release. Obtaining reliable readouts from blood can provide crucial information for disease progression, as well as being of prognostic value monitoring patients' response to treatment.
Collapse
Affiliation(s)
- Periklis Katopodis
- Biosciences, College of Health, Medicine and Life Sciences, Brunel University London, Uxbridge, UK.,Thoracic Surgery, Royal Brompton & Harefield NHS Foundation Trust, Harefield, UK
| | - Vladimir Anikin
- Biosciences, College of Health, Medicine and Life Sciences, Brunel University London, Uxbridge, UK.,Thoracic Surgery, Royal Brompton & Harefield NHS Foundation Trust, Harefield, UK.,Department of Oncology and Reconstructive Surgery, Sechenov First Moscow State Medical University, Moscow, Russia
| | - Uday Kishore
- Biosciences, College of Health, Medicine and Life Sciences, Brunel University London, Uxbridge, UK
| | | | - Marcia Hall
- Biosciences, College of Health, Medicine and Life Sciences, Brunel University London, Uxbridge, UK.,Mount Vernon Cancer Centre, Northwood, UK
| | - Nizar Asadi
- Royal Brompton and Harefield NHS Trust, London, UK
| | | | - Emmanouil Karteris
- Biosciences, College of Health, Medicine and Life Sciences, Brunel University London, Uxbridge, UK .,Thoracic Surgery, Royal Brompton & Harefield NHS Foundation Trust, Harefield, UK
| |
Collapse
|
16
|
Deregulation of protein phosphatase 2A inhibitor SET is associated with malignant progression in breast cancer. Sci Rep 2021; 11:14238. [PMID: 34244560 PMCID: PMC8270961 DOI: 10.1038/s41598-021-93620-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2021] [Accepted: 06/22/2021] [Indexed: 11/18/2022] Open
Abstract
To understand the mechanism underlying metastasis, identification of a mechanism-based and common biomarker for circulating tumour cells (CTCs) in heterogenous breast cancer is needed. SET, an endogenous inhibitor of protein phosphatase 2A, was overexpressed in all subtypes of invasive breast carcinoma tissues. Treatment with SET-targeted siRNAs reduced the motility of MCF-7 and MDA-MB-231 cells in transwell assay. SET knockdown reduced the number of mammospheres by 60–70% in MCF-7 and MDA-MB-231 cells, which was associated with the downregulation of OCT4 and SLUG. Hence, we analysed the presence of SET-expressing CTCs (SET-CTCs) in 24 breast cancer patients. CTCs were enriched using a size-based method and then immunocytochemically analysed using an anti-SET antibody. SET-CTCs were detected in 6/6 (100%) patients with recurrent breast cancer with a median value of 12 (12 cells/3 mL blood), and in 13/18 (72.2%) patients with stage I–III breast cancer with a median value of 2.5, while the median value of healthy controls was 0. Importantly, high numbers of SET-CTCs were correlated with lymph node metastasis in patients with stage I–III disease. Our results indicate that SET contributes to breast cancer progression and can act as a potential biomarker of CTCs for the detection of metastasis.
Collapse
|
17
|
Guedes de Castro D, Braun AC, Calsavara VF, Melo Gondim GR, Gobo Silva ML, Chen MJ, Fogaroli RC, Ramos H, Coelho TM, Scintini Herbst AC, Abdallah EA, Yu LS, Fidarova E, Zubizarreta E, Assis Pellizzon AC, Domingos Chinen LT. Prospective Assessment of the Association Between Circulating Tumor Cells and Control of Brain Disease After Focal Radiation Therapy of Breast Cancer Brain Metastases. Adv Radiat Oncol 2021; 6:100673. [PMID: 33912738 PMCID: PMC8071730 DOI: 10.1016/j.adro.2021.100673] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2020] [Revised: 01/20/2021] [Accepted: 01/27/2021] [Indexed: 11/28/2022] Open
Abstract
Purpose Predicting the risk of early distant brain failure (DBF) is in demand for management decisions in patients who are candidates for local treatment of brain metastases. This study aimed to analyze the association between circulating tumor cells (CTCs) and brain disease control after stereotactic radiation therapy/radiosurgery (SRT) for breast cancer brain metastasis (BCBM). Methods and Materials We prospectively assessed CTCs before (CTC1) and 4 to 5 weeks after (CTC2) SRT and their relationship with the number of new lesions (NL) suggestive of BCBM before SRT. CTC were quantified and analyzed by immunocytochemistry to evaluate the expression of the proteins COX2, EGFR, ST6GALNAC5, NOTCH1, and HER2. Distant brain failure-free survival (DBFFS), the primary endpoint, diffuse DBFFS (D-DBFFS), and overall survival were estimated. Analysis for DBF within 6 months, with death as competing risk, was performed. Results Patients were included between 2016 and 2018. CTCs were detected in all 39 patients before and in 34 of 35 patients after SRT. After median follow-up of 16.6 months, median DBFFS, D-DBFFS, and overall survival were 15.3, 14.1, and 19.5 months, respectively. DBF at 6 months was 40% with CTC1 ≤0.5 and 8.82% with CTC1 >0.5 CTC/mL (P = .007), and D-DBF at 6 months was 40% with CTC1 ≤0.5 and 0 with CTC1 >0.5 CTC/mL (P = .005) and 25% with NL/CTC1 >6.8 and 2.65% with NL/CTC1 ≤6.8 (P = .063). On multivariate analysis, DBFFS was inferior with CTC1 ≤0.5 (hazard ratio, 8.27; 95% confidence interval, 2.12-32.3; P = .002), and D-DBFFS was inferior with CTC1 ≤0.5 (hazard ratio, 10.22; 95% confidence interval, 1.99-52.41; P = .005). Protein expression was not associated with outcomes. Conclusions These data suggest that CTC1 and NL/CTC1 may have a role as a biomarker of early diffuse DBF and as a subsequent guide between focal or whole-brain radiation therapy in patients with BCBM.
Collapse
|
18
|
Mehdipour P, Javan F, Jouibari MF, Khaleghi M, Mehrazin M. Evolutionary model of brain tumor circulating cells: Cellular galaxy. World J Clin Oncol 2021; 12:13-30. [PMID: 33552936 PMCID: PMC7829626 DOI: 10.5306/wjco.v12.i1.13] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/29/2020] [Revised: 11/05/2020] [Accepted: 11/28/2020] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Although circulating tumor cells (CTCs) have been the focus of consideration for a decade, a categorized cell-based diagnostic strategy is unavailable. The personalized management and complementary/analytical-strategy of data require an alphabetic guide. Therefore, we aimed to determine the behavior of CTCs in tumor and blood in order to provide the hypothetical-based agenda in the brain neoplasms. Exploring the protein expression (PE) using a single cell-based method would clarify the heterogeneity and diversity in tumor and blood, which are key events in the evolution in brain tumors. In fact, heterogeneity, diversity, and evolution are required for cancer initiation and progression. AIM To explore CTCs in brain tumors and blood cells and to assay intensity of PE through personalized insight. METHODS The focal population included 14 patients with meningioma, and four patients with metastatic brain tumors (T). PE was assayed by immunofluorescence in tumors cells and CTCs in 18 patients with brain tumors. Ratio test was applied between the T cells and CTCs in tumor tissue and in vascular system. T/CTC ratio-based classification of PE in macrophage chemoattractant chemokine ligand 2 (CCL2), vascular endothelial growth factor (VEGF), epidermal growth factor (EGF), CD133, cyclin E, neurofilament marker, cytokeratin 19, and leukocyte common antigen (CD45) were investigated. RESULTS Total analyzed cells ranged between 10794-92283 for tumor cells and between 117-2870 for CTCs. Characteristics of histopathologic and status of an ataxia-telangiectasia mutated polymorphism (D1853N) in 18 patients affected with brain tumors were also provided. The course of evolution and metastatic event relied on the elevated protein expression in CTCs, which could be considered as a prognostic value. Diverse protein expression of the migrated cells into the blood stream and the tumor was indicative of the occurrence of evolution. Besides, the harmonic co-expression between CCL2/EGF and CCL2/VEGF could facilitate the tumor progression including the metastatic event. Expression of these proteins in the migrated vasculature and into the buccal tissue offered a non-invasive follow-up detection in neoplastic disorders. PE-exploration of neurofilament marker/CD133/VEGF of the CTCs in meningioma and cytokeratin 19/CD45/ cyclin E in the patients with metastatic brain tumor would clarify the tumor biology of the brain neoplastic disorders. CONCLUSION The alphabetical base of the evolutionary mechanisms relies on dual-, triple-, and multi-models with diverse intensity of expression. In fact, cross-talk between initiative and the complementary channels defines the evolutionary insight in cancer. A diverse-model of protein expression, including low, medium, and high intensity, is the key requirement for the completed model. The cluster of cells with diverse expression and remarkable co-expression between CCL2/EGF/VEGF and NM/CD133/VEGF in CTCs may be indicative of probable invasiveness of the tumor. Furthermore, the mode of cytokeratin-19+/CD45- can be traced in the metastatic patients.
Collapse
Affiliation(s)
- Parvin Mehdipour
- Department of Medical Genetics, Tehran University of Medical Sciences, School of Medicine, Tehran 1417613151, Tehran, Iran
| | - Firoozeh Javan
- Department of Medical Genetics, Tehran University of Medical Sciences, School of Medicine, Tehran 1417613151, Tehran, Iran
| | | | - Mehdi Khaleghi
- Shariati Hospital, Tehran University of Medical Sciences, Tehran 1417613151, Tehran, Iran
| | - Masoud Mehrazin
- Tehran University of Medical Science, Tehran 1417613151, Tehran, Iran
| |
Collapse
|
19
|
Hu L, Chen X, Chen M, Fang J, Nie J, Dai H. Enrichment and detection of circulating tumor cells by immunomagnetic beads and flow cytometry. Biotechnol Lett 2020; 43:25-34. [PMID: 32959190 DOI: 10.1007/s10529-020-03007-8] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2020] [Accepted: 09/14/2020] [Indexed: 01/07/2023]
Abstract
OBJECTIVE The purpose of the article is to establish a quick enrichment and detection method using immunomagnetic beads and flow cytometry to analyze circulating tumor cells (CTCs) in the peripheral blood. RESULTS After incubation with CD326-PE and CD45-APC antibodies, more than 60% MCF7 cells in M-Buffer could be detected while less than 10% of the same cells could be detected by flow cytometry (FCM) if spiked into blood. However, in combination with CD326 and CD45 immunomagnetic beads, detection rate of MCF7 cells in blood reached 57%. For circulating tumor cells, enrichment by CD326 and CD45 immunomagnetic beads improve the detection rate from nearly undetectable to more than 24.14%. CONCLUSIONS Live CTCs in peripheral blood can be effectively and sensitively detected by using a combination of immunomagnetic beads (CD45 and CD326) and flow cytometry.
Collapse
Affiliation(s)
- Lei Hu
- Anhui Province Key Laboratory of Medical Physics and Technology, Institute of Health & Medical Technology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei, 230031, China.,Hefei Cancer Hospital, Chinese Academy of Sciences, 350 Shushanhu Road, Hefei, 230031, Anhui, China
| | - Xueran Chen
- Anhui Province Key Laboratory of Medical Physics and Technology, Institute of Health & Medical Technology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei, 230031, China.,Hefei Cancer Hospital, Chinese Academy of Sciences, 350 Shushanhu Road, Hefei, 230031, Anhui, China
| | - Meng Chen
- Anhui Province Key Laboratory of Medical Physics and Technology, Institute of Health & Medical Technology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei, 230031, China.,University of Science and Technology of China, Hefei, 230026, China.,Hefei Cancer Hospital, Chinese Academy of Sciences, 350 Shushanhu Road, Hefei, 230031, Anhui, China
| | - Jinman Fang
- Anhui Province Key Laboratory of Medical Physics and Technology, Institute of Health & Medical Technology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei, 230031, China.,Hefei Cancer Hospital, Chinese Academy of Sciences, 350 Shushanhu Road, Hefei, 230031, Anhui, China
| | - Jinfu Nie
- Anhui Province Key Laboratory of Medical Physics and Technology, Institute of Health & Medical Technology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei, 230031, China. .,Hefei Cancer Hospital, Chinese Academy of Sciences, 350 Shushanhu Road, Hefei, 230031, Anhui, China. .,Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, 510530, China.
| | - Haiming Dai
- Anhui Province Key Laboratory of Medical Physics and Technology, Institute of Health & Medical Technology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei, 230031, China. .,Hefei Cancer Hospital, Chinese Academy of Sciences, 350 Shushanhu Road, Hefei, 230031, Anhui, China.
| |
Collapse
|
20
|
Risk factors for breast cancer brain metastases: a systematic review. Oncotarget 2020; 11:650-669. [PMID: 32110283 PMCID: PMC7021234 DOI: 10.18632/oncotarget.27453] [Citation(s) in RCA: 50] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2019] [Accepted: 01/04/2020] [Indexed: 11/25/2022] Open
Abstract
Background: Brain metastasis (BM) is an increasingly common and devastating complication of breast cancer (BC). Methods: A systematic literature search of EMBASE and MEDLINE was conducted to elucidate the current state of knowledge on known and novel prognostic factors associated with 1) the risk for BCBM and 2) the time to brain metastases (TTBM). Results: A total of 96 studies involving institutional records from 28 countries were identified. Of these, 69 studies reported risk factors of BCBM, 46 factors associated with the TTBM and twenty studies examined variables for both outcomes. Young age, estrogen receptor negativity (ER-), overexpression of human epidermal factor (HER2+), and higher presenting stage, histological grade, tumor size, Ki67 labeling index and nodal involvement were consistently found to be independent risk factors of BCBM. Of these, triple-negative BC (TNBC) subtype, ER-, higher presenting histological grade, tumor size, and nodal involvement were also reported to associate with shorter TTBM. In contrast, young age, hormone receptor negative (HR-) status, higher presenting stage, nodal involvement and development of liver metastasis were the most important risk factors for BM in HER2-positive patients. Conclusions: The study provides a comprehensive and individual evaluation of the risk factors that could support the design of screening tools and interventional trials for early detection of BCBM.
Collapse
|
21
|
CTCs Expression Profiling for Advanced Breast Cancer Monitoring. Cancers (Basel) 2019; 11:cancers11121941. [PMID: 31817194 PMCID: PMC6966538 DOI: 10.3390/cancers11121941] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2019] [Revised: 11/18/2019] [Accepted: 12/02/2019] [Indexed: 12/29/2022] Open
Abstract
The study of circulating tumor cells (CTCs) has a huge clinical interest in advance and metastatic breast cancer patients. However, many approaches are biased by the use of epithelial markers, which underestimate non-epithelial CTCs phenotypes. CTCs enumeration provides valuable prognostic information; however, molecular characterization could be the best option to monitor patients throughout the disease since it may provide more relevant clinical information to the physicians. In this work, we aimed at enumerating and performing a molecular characterization of CTCs from a cohort of 20 patients with metastatic breast cancer (MBC), monitoring the disease at different time points of the therapy, and at progression when it occurred. To this end, we used a CTC negative enrichment protocol that allowed us to recover a higher variety of CTCs phenotypes. With this strategy, we were able to obtain gene expression data from CTCs from all the patients. In addition, we found that high expression levels of PALB2 and MYC were associated with a worse outcome. Interestingly, we identified that CTCs with an EpCAMhighVIMlowALDH1A1high signature showed both shorter overall survival (OS) and progression-free survival (PFS), suggesting that CTCs with epithelial-stem features had the most aggressive phenotype.
Collapse
|
22
|
An In Vitro Model of Triple-Negative Breast Cancer. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2019; 1164:35-46. [PMID: 31576538 DOI: 10.1007/978-3-030-22254-3_3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/22/2023]
Abstract
We have characterized two highly tumorigenic and metastatic basal B TNBC cell lines, XtMCF and LmMCF, with the additional values of having the normal and early-stage counterparts of them. This model allows the study of the evolution of TNBC, and investigates molecular pathways at different stages of transformation and progression in a relatively constant genetic background. This constitutes an ideal model for developing targeted therapy in two important fields in cancer biology which are the epithelial mesenchymal transition (EMT) and cancer stem cells (CSC).
Collapse
|
23
|
Abstract
As an alternative target to surgically resected tissue specimens, liquid biopsy has gained much attention over the past decade. Of the various circulating biomarkers, circulating tumor cells (CTCs) have particularly opened new windows into the metastatic cascade, with their functional, biochemical, and biophysical properties. Given the extreme rarity of intact CTCs and the associated technical challenges, however, analyses have been limited to bulk-cell strategies, missing out on clinically significant sources of information from cellular heterogeneity. With recent technological developments, it is now possible to probe genetic material of CTCs at the single-cell resolution to study spatial and temporal dynamics in circulation. Here, we discuss recent transcriptomic profiling efforts that enabled single-cell characterization of patient-derived CTCs spanning diverse cancer types. We further highlight how expression data of these putative biomarkers have advanced our understanding of metastatic spectrum and provided a basis for the development of CTC-based liquid biopsies to track, monitor, and predict the efficacy of therapy and any emergent resistance.
Collapse
|
24
|
EpCAM low Circulating Tumor Cells: Gold in the Waste. DISEASE MARKERS 2019; 2019:1718920. [PMID: 31636732 PMCID: PMC6766153 DOI: 10.1155/2019/1718920] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 05/03/2019] [Revised: 09/06/2019] [Accepted: 09/11/2019] [Indexed: 12/14/2022]
Abstract
The CellSearch® system which is still considered the gold standard for the enumeration of circulating tumor cells (CTC) utilizes antibodies against the epithelial cell adhesion molecule (EpCAM) for CTC enrichment. Recently, CTC discarded by the CellSearch® system due to their low EpCAM expression have been isolated and analyzed. We here sought to discuss technical and biological issues concerning the isolation and characterization of EpCAMlow CTC, highlighting the enormous potential of this subpopulation discarded by CellSearch®, which might instead reveal an unexpected clinical significance in tumor types where CTC enumeration has never been validated for prognostic and predictive purpose.
Collapse
|
25
|
Riebensahm C, Joosse SA, Mohme M, Hanssen A, Matschke J, Goy Y, Witzel I, Lamszus K, Kropidlowski J, Petersen C, Kolb-Kokocinski A, Sauer S, Borgmann K, Glatzel M, Müller V, Westphal M, Riethdorf S, Pantel K, Wikman H. Clonality of circulating tumor cells in breast cancer brain metastasis patients. Breast Cancer Res 2019; 21:101. [PMID: 31481116 PMCID: PMC6720990 DOI: 10.1186/s13058-019-1184-2] [Citation(s) in RCA: 48] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2019] [Accepted: 08/09/2019] [Indexed: 12/17/2022] Open
Abstract
Background The incidence of brain metastases in breast cancer (BCBM) patients is increasing. These patients have a very poor prognosis, and therefore, identification of blood-based biomarkers, such as circulating tumor cells (CTCs), and understanding the genomic heterogeneity could help to personalize treatment options. Methods Both EpCAM-dependent (CellSearch® System) and EpCAM-independent Ficoll-based density centrifugation methods were used to detect CTCs from 57 BCBM patients. DNA from individual CTCs and corresponding primary tumors and brain metastases were analyzed by next-generation sequencing (NGS) in order to evaluate copy number aberrations and single nucleotide variations (SNVs). Results CTCs were detected after EpCAM-dependent enrichment in 47.7% of the patients (≥ 5 CTCs/7.5 ml blood in 20.5%). The CTC count was associated with ERBB2 status (p = 0.029) of the primary tumor as well as with the prevalence of bone metastases (p = 0.021). EpCAM-independent enrichment revealed CTCs in 32.6% of the patients, especially among triple-negative breast cancer (TNBC) patients (70.0%). A positive CTC status after enrichment of either method was significantly associated with decreased overall survival time (p < 0.05). Combining the results of both enrichment methods, 63.6% of the patients were classified as CTC positive. In three patients, the matched tumor tissue and single CTCs were analyzed by NGS showing chromosomal aberrations with a high genomic clonality and mutations in pathways potentially important in brain metastasis formation. Conclusion The detection of CTCs, regardless of the enrichment method, is of prognostic relevance in BCBM patients and in combination with molecular analysis of CTCs can help defining patients with higher risk of early relapse and suitability for targeted treatment. Electronic supplementary material The online version of this article (10.1186/s13058-019-1184-2) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Carlotta Riebensahm
- Department of Tumor Biology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Simon A Joosse
- Department of Tumor Biology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Malte Mohme
- Department of Neurosurgery, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Annkathrin Hanssen
- Department of Tumor Biology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Jakob Matschke
- Department of Neuropathology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Yvonne Goy
- Department of Radiotherapy and Radiooncology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany.,Laboratory of Radiobiology and Experimental Radiooncology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Isabell Witzel
- Department of Gynecology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Katrin Lamszus
- Department of Neurosurgery, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Jolanthe Kropidlowski
- Department of Tumor Biology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Cordula Petersen
- Department of Radiotherapy and Radiooncology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | | | - Sascha Sauer
- Max Delbrück Center for Molecular Medicine (BIMSB and BIH), Berlin, Germany.,Max Planck Institute for Molecular Genetics, Otto Warburg Laboratory, Berlin, Germany
| | - Kerstin Borgmann
- Laboratory of Radiobiology and Experimental Radiooncology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Markus Glatzel
- Department of Neuropathology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Volkmar Müller
- Department of Gynecology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Manfred Westphal
- Department of Neurosurgery, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Sabine Riethdorf
- Department of Tumor Biology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Klaus Pantel
- Department of Tumor Biology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Harriet Wikman
- Department of Tumor Biology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany.
| |
Collapse
|
26
|
Lozar T, Gersak K, Cemazar M, Kuhar CG, Jesenko T. The biology and clinical potential of circulating tumor cells. Radiol Oncol 2019; 53:131-147. [PMID: 31104002 PMCID: PMC6572494 DOI: 10.2478/raon-2019-0024] [Citation(s) in RCA: 44] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2019] [Accepted: 05/03/2019] [Indexed: 02/06/2023] Open
Abstract
Background Tumor cells can shed from the tumor, enter the circulation and travel to distant organs, where they can seed metastases. These cells are called circulating tumor cells (CTCs). The ability of CTCs to populate distant tissues and organs has led us to believe they are the primary cause of cancer metastasis. The biological properties and interaction of CTCs with other cell types during intravasation, circulation in the bloodstream, extravasation and colonization are multifaceted and include changes of CTC phenotypes that are regulated by many signaling molecules, including cytokines and chemokines. Considering a sample is readily accessible by a simple blood draw, monitoring CTC levels in the blood has exceptional implications in oncology field. A method called the liquid biopsy allows the extraction of not only CTC, but also CTC products, such as cell free DNA (cfDNA), cell free RNA (cfRNA), microRNA (miRNA) and exosomes. Conclusions The clinical utility of CTCs and their products is increasing with advances in liquid biopsy technology. Clinical applications of liquid biopsy to detect CTCs and their products are numerous and could be used for screening of the presence of the cancer in the general population, as well as for prognostic and predictive biomarkers in cancer patients. With the development of better CTC isolation technologies and clinical testing in large prospective trials, increasing clinical utility of CTCs can be expected. The understanding of their biology and interactions with other cell types, particularly with those of the immune system and the rise of immunotherapy also hold great promise for novel therapeutic possibilities.
Collapse
Affiliation(s)
- Taja Lozar
- Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia
| | - Klara Gersak
- Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia
- Institute of Oncology Ljubljana, Ljubljana, Slovenia
- General Hospital Izola, Izola, Slovenia
| | - Maja Cemazar
- Institute of Oncology Ljubljana, Ljubljana, Slovenia
- Faculty of Health Sciences, University of Primorska, Izola, Slovenia
| | | | - Tanja Jesenko
- Institute of Oncology Ljubljana, Ljubljana, Slovenia
| |
Collapse
|
27
|
Hanssen A, Riebensahm C, Mohme M, Joosse SA, Velthaus JL, Berger LA, Bernreuther C, Glatzel M, Loges S, Lamszus K, Westphal M, Riethdorf S, Pantel K, Wikman H. Frequency of Circulating Tumor Cells (CTC) in Patients with Brain Metastases: Implications as a Risk Assessment Marker in Oligo-Metastatic Disease. Cancers (Basel) 2018; 10:E527. [PMID: 30572662 PMCID: PMC6315958 DOI: 10.3390/cancers10120527] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2018] [Revised: 12/09/2018] [Accepted: 12/17/2018] [Indexed: 12/12/2022] Open
Abstract
Forty percent of non-small cell lung cancer (NSCLC) patients develop brain metastases, resulting in a dismal prognosis. However, patients in an oligo-metastatic brain disease setting seem to have better outcomes. Here, we investigate the possibility of using circulating tumor cells (CTCs) as biomarkers to differentiate oligo-metastatic patients for better risk assessment. Using the CellSearch® system, few CTCs were detected among NSCLC patients with brain metastases (n = 52, 12.5% ≥ two and 8.9% ≥ five CTC/7.5 mL blood) and especially oligo-metastatic brain patients (n = 34, 5.9%, and 2.9%). Still, thresholds of both ≥ two and ≥ five CTCs were independent prognostic indicators for shorter overall survival time among all of the NSCLC patients (n = 90, two CTC ≥ HR: 1.629, p = 0.024, 95% CI: 1.137⁻6.465 and five CTC ≥ HR: 2.846, p = 0.0304, CI: 1.104⁻7.339), as well as among patients with brain metastases (two CTC ≥ HR: 4.694, p = 0.004, CI: 1.650⁻13.354, and five CTC ≥ HR: 4.963, p = 0.003, CI: 1.752⁻14.061). Also, oligo-brain NSCLC metastatic patients with CTCs had a very poor prognosis (p = 0.019). Similarly, in other tumor entities, only 9.6% of patients with brain metastases (n = 52) had detectable CTCs. Our data indicate that although patients with brain metastases more seldom harbor CTCs, they are still predictive for overall survival, and CTCs might be a useful biomarker to identify oligo-metastatic NSCLC patients who might benefit from a more intense therapy.
Collapse
Affiliation(s)
- Annkathrin Hanssen
- Department of Tumor Biology, University Medical Centre Hamburg-Eppendorf, 20246 Hamburg, Germany.
| | - Carlotta Riebensahm
- Department of Tumor Biology, University Medical Centre Hamburg-Eppendorf, 20246 Hamburg, Germany.
| | - Malte Mohme
- Department of Neurosurgery University Medical Centre Hamburg-Eppendorf, 20246 Hamburg, Germany.
| | - Simon A Joosse
- Department of Tumor Biology, University Medical Centre Hamburg-Eppendorf, 20246 Hamburg, Germany.
| | - Janna-Lisa Velthaus
- Department of Internal Medicine II and Clinic (Oncology Centre) University Medical Centre Hamburg-Eppendorf, 20246 Hamburg, Germany.
| | - Lars Arne Berger
- Department of Internal Medicine II and Clinic (Oncology Centre) University Medical Centre Hamburg-Eppendorf, 20246 Hamburg, Germany.
| | - Christian Bernreuther
- Institute of Neuropathology University Medical Centre Hamburg-Eppendorf, 20246 Hamburg, Germany.
| | - Markus Glatzel
- Institute of Neuropathology University Medical Centre Hamburg-Eppendorf, 20246 Hamburg, Germany.
| | - Sonja Loges
- Department of Tumor Biology, University Medical Centre Hamburg-Eppendorf, 20246 Hamburg, Germany.
- Department of Internal Medicine II and Clinic (Oncology Centre) University Medical Centre Hamburg-Eppendorf, 20246 Hamburg, Germany.
| | - Katrin Lamszus
- Department of Neurosurgery University Medical Centre Hamburg-Eppendorf, 20246 Hamburg, Germany.
| | - Manfred Westphal
- Department of Neurosurgery University Medical Centre Hamburg-Eppendorf, 20246 Hamburg, Germany.
| | - Sabine Riethdorf
- Department of Tumor Biology, University Medical Centre Hamburg-Eppendorf, 20246 Hamburg, Germany.
| | - Klaus Pantel
- Department of Tumor Biology, University Medical Centre Hamburg-Eppendorf, 20246 Hamburg, Germany.
| | - Harriet Wikman
- Department of Tumor Biology, University Medical Centre Hamburg-Eppendorf, 20246 Hamburg, Germany.
| |
Collapse
|
28
|
Poudineh M, Sargent EH, Pantel K, Kelley SO. Profiling circulating tumour cells and other biomarkers of invasive cancers. Nat Biomed Eng 2018; 2:72-84. [DOI: 10.1038/s41551-018-0190-5] [Citation(s) in RCA: 144] [Impact Index Per Article: 20.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2017] [Accepted: 01/09/2018] [Indexed: 02/07/2023]
|
29
|
Riethdorf S, O'Flaherty L, Hille C, Pantel K. Clinical applications of the CellSearch platform in cancer patients. Adv Drug Deliv Rev 2018; 125:102-121. [PMID: 29355669 DOI: 10.1016/j.addr.2018.01.011] [Citation(s) in RCA: 179] [Impact Index Per Article: 25.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2017] [Revised: 01/12/2018] [Accepted: 01/15/2018] [Indexed: 12/29/2022]
Abstract
The CellSearch® system (CS) enables standardized enrichment and enumeration of circulating tumor cells (CTCs) that are repeatedly assessable via non-invasive "liquid biopsy". While the association of CTCs with poor clinical outcome for cancer patients has clearly been demonstrated in numerous clinical studies, utilizing CTCs for the identification of therapeutic targets, stratification of patients for targeted therapies and uncovering mechanisms of resistance is still under investigation. Here, we comprehensively review the current benefits and drawbacks of clinical CTC analyses for patients with metastatic and non-metastatic tumors. Furthermore, the review focuses on approaches beyond CTC enumeration that aim to uncover therapeutically relevant antigens, genomic aberrations, transcriptional profiles and epigenetic alterations of CTCs at a single cell level. This characterization of CTCs may shed light on the heterogeneity and genomic landscapes of malignant tumors, an understanding of which is highly important for the development of new therapeutic strategies.
Collapse
|
30
|
Lu YJ, Wang P, Wang X, Peng J, Zhu YW, Shen N. The significant prognostic value of circulating tumor cells in triple-negative breast cancer: a meta-analysis. Oncotarget 2018; 7:37361-37369. [PMID: 27008698 PMCID: PMC5095082 DOI: 10.18632/oncotarget.8156] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2015] [Accepted: 02/29/2016] [Indexed: 01/10/2023] Open
Abstract
Background The clinical validity of circulating tumor cells (CTCs) is still controversial in patients with triple-negative breast cancer (TNBC). Methods A comprehensive literature search was performed to identify relevant articles in the PubMed, Web of Science, MEDLINE, and Embase databases through September 2015. The outcomes of interest were disease progression and overall survival. The hazard ratio (HR) and 95% confidence interval (95% CI) were considered the effect indicators and were pooled in meta-analyses under a fixed- or random-effect model according to heterogeneity. Results Ten of the eligible studies were included for a total of 642 enrolled TNBC patients. Overall analyses revealed that the presence of CTCs predicted aggressive disease progression (HR = 2.18, 95% CI = 1.59-2.99, Pheterogeneity = 0.010, I2 = 52.2%) and reduced overall survival (HR = 2.02, 95% CI = 1.59-2.57, Pheterogeneity = 0.169, I2 = 26.6%). Further subgroup analyses demonstrated that CTC-positive patients also had poor disease progression and overall survival in different subsets, including cancer stage. Conclusion Our meta-analysis provides strong evidence that detection of CTC in the peripheral blood is an independent prognosticator of poor survival outcomes for TNBC patients.
Collapse
Affiliation(s)
- Yan-Jun Lu
- Department of Laboratory Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Peng Wang
- Institute and Department of Infectious Disease, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xiong Wang
- Department of Laboratory Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jing Peng
- Department of Laboratory Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yao-Wu Zhu
- Department of Laboratory Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Na Shen
- Department of Laboratory Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
31
|
Li YR, Tseng CP, Hsu HL, Lin HC, Chen YA, Chen ST, Liou MJ, Lin JD. Circulating epithelial cells as potential biomarkers for detection of recurrence in patients of papillary thyroid carcinoma with positive serum anti-thyroglobulin antibody. Clin Chim Acta 2017; 477:74-80. [PMID: 29229463 DOI: 10.1016/j.cca.2017.12.011] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2017] [Revised: 11/28/2017] [Accepted: 12/07/2017] [Indexed: 12/16/2022]
Abstract
BACKGROUND Serum thyroglobulin (Tg) is not a reliable tumor marker for monitoring disease status after treatment in patients with papillary thyroid carcinoma (PTC) with positive anti-thyroglobulin antibody (TgAb). The aim of this study was to evaluate the clinical role of circulating epithelial cells (CECs) in PTC patients with positive serum TgAb and undetectable serum Tg. METHODS A pilot study was performed to evaluate CECs in 25 PTC patients with positive serum TgAb and undetectable serum Tg. CECs were isolated and enriched from peripheral blood with a negative selection system PowerMag. Immunofluorescence staining with anti-epithelial cell adhesion molecule (anti-EpCAM) and anti-thyroid stimulating hormone receptor (anti-TSHR) antibodies were used to define EpCAM+-CECs and TSHR+-CECs. After CECs testing, 25 patients were classified into two groups: recurrence group (n=7) and remission group (n=18) based on biopsy or imaging studies. The diagnostic accuracy and cutoff points of EpCAM+-CECs and TSHR+-CECs were evaluated using receiver operating characteristic (ROC) curves. The optimal cut-off values of CECs were determined by the Youden index (sensitivity+specificity-1). RESULTS The median numbers of EpCAM+-CECs (72.5 vs. 10.75) and TSHR+-CECs (54 vs. 5.25) were significantly increased in recurrence group compared to remission group. The area under the curve (AUC) showed good performance of EpCAM+-CECs (0.937) and TSHR+-CECs (0.825) to discriminate between recurrence and remission. The cut-off value for EpCAM+-CECs and TSHR+-CECs were set at 48cells/ml and 10cells/ml, respectively and showed a sensitivity (EpCAM+-CECs: 85.7%; TSHR+-CECs: 85.7%) and a specificity (EpCAM+-CECs: 100%; TSHR+-CECs: 77.8%) in predicting the recurrence. CONCLUSIONS Our study suggests CECs testing could be a potential biomarker to identify recurrence in PTC patients with positive serum TgAb and undetectable serum Tg.
Collapse
Affiliation(s)
- Yan-Rong Li
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Chang Gung Memorial Hospital, Taiwan
| | - Ching-Ping Tseng
- Department of Medical Biotechnology and Laboratory Science, College of Medicine, Chang Gung University, Taiwan; Graduate Institute of Biomedical Science, College of Medicine, Chang Gung University, Taiwan; Molecular Medicine Research Center, Chang Gung University, Taiwan; Department of Laboratory Medicine, Chang Gung Memorial Hospital, Taiwan
| | - Hsueh-Ling Hsu
- Department of Medical Biotechnology and Laboratory Science, College of Medicine, Chang Gung University, Taiwan
| | - Hung-Chih Lin
- Graduate Institute of Biomedical Science, College of Medicine, Chang Gung University, Taiwan
| | - Yi-An Chen
- Department of Medical Biotechnology and Laboratory Science, College of Medicine, Chang Gung University, Taiwan
| | - Szu-Tah Chen
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Chang Gung Memorial Hospital, Taiwan
| | - Miaw-Jene Liou
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Chang Gung Memorial Hospital, Taiwan
| | - Jen-Der Lin
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Chang Gung Memorial Hospital, Taiwan.
| |
Collapse
|
32
|
Jakabova A, Bielcikova Z, Pospisilova E, Matkowski R, Szynglarewicz B, Staszek-Szewczyk U, Zemanova M, Petruzelka L, Eliasova P, Kolostova K, Bobek V. Molecular characterization and heterogeneity of circulating tumor cells in breast cancer. Breast Cancer Res Treat 2017; 166:695-700. [PMID: 28815327 PMCID: PMC5680374 DOI: 10.1007/s10549-017-4452-9] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2017] [Accepted: 08/07/2017] [Indexed: 12/19/2022]
Abstract
Introduction This study analyzes peripheral blood samples from breast cancer (BC) patients. CTCs from peripheral blood were enriched by size-based separation and were then cultivated in vitro. The primary aim of this study was to demonstrate the antigen independent CTC separation method with high CTC recovery. Subsequently, CTCs enriched several times during the treatment were characterized molecularly. Methods Patients with different stages of BC (N = 167) were included into the study. All patients were candidates for surgery, surgical diagnostics, or were undergoing chemotherapy. In parallel, 20 patients were monitored regularly and in addition to CTC presence, also CTC character was examined by qPCR, with special focus on HER2 and ESR status. Results CTC positivity in the cohort was 76%. There was no significant difference between the tested groups, but the highest CTC occurrence was identified in the group undergoing surgery and similarly in the group before the start of neoadjuvant treatment. On the other hand, the lowest CTC frequencies were observed in the menopausal patient group (56%), ESR+ patient group (60%), and DCIS group (44.4%). It is worth noting that after completion of neoadjuvant therapy (NACT) CTCs were present in 77.7% of cases. On the other hand, patients under hormonal treatment were CTC positive only in 52% of cases. Discussions Interestingly, HER2 and ESR status of CTCs differs from the status of primary tumor. In 50% of patients HER2 status on CTCs changed not only from HER2+ to HER2−, but also from HER2– to HER2+ (33%). ESR status in CTCs changed only in one direction from ESR+ to ESR−. Conclusions Data obtained from the present study suggest that BC is a heterogeneous disease but CTCs may be detected independently of the disease characteristics in 76% of patients at any time point during the course of the disease. This relatively high CTC occurrence in BC should be considered when planning the long-term patient monitoring.
Collapse
Affiliation(s)
- Anna Jakabova
- Department of Laboratory Genetics, University Hospital Kralovske Vinohrady, Srobarova 50, 100 34, Prague, Czech Republic
| | - Zuzana Bielcikova
- Department of Oncology, First Faculty of Medicine, Charles University and General University Hospital in Prague, Prague, Czech Republic
| | - Eliska Pospisilova
- Department of Laboratory Genetics, University Hospital Kralovske Vinohrady, Srobarova 50, 100 34, Prague, Czech Republic
| | - Rafal Matkowski
- Division of Surgical Oncology and Department of Oncology, Wroclaw Medical University, Wybrzeże Ludwika Pasteura 1, 50-367, Wrocław, Poland.,Breast Unit, Lower Silesian Cancer Center, Wroclaw, Plac Hirszfelda 12, 53-413, Wrocław, Poland
| | - Bartlomiej Szynglarewicz
- Division of Surgical Oncology and Department of Oncology, Wroclaw Medical University, Wybrzeże Ludwika Pasteura 1, 50-367, Wrocław, Poland.,Breast Unit, Lower Silesian Cancer Center, Wroclaw, Plac Hirszfelda 12, 53-413, Wrocław, Poland
| | - Urszula Staszek-Szewczyk
- Division of Surgical Oncology and Department of Oncology, Wroclaw Medical University, Wybrzeże Ludwika Pasteura 1, 50-367, Wrocław, Poland.,Breast Unit, Lower Silesian Cancer Center, Wroclaw, Plac Hirszfelda 12, 53-413, Wrocław, Poland
| | - Milada Zemanova
- Department of Oncology, First Faculty of Medicine, Charles University and General University Hospital in Prague, Prague, Czech Republic
| | - Lubos Petruzelka
- Department of Oncology, First Faculty of Medicine, Charles University and General University Hospital in Prague, Prague, Czech Republic
| | - Petra Eliasova
- Department of Laboratory Genetics, University Hospital Kralovske Vinohrady, Srobarova 50, 100 34, Prague, Czech Republic
| | - Katarina Kolostova
- Department of Laboratory Genetics, University Hospital Kralovske Vinohrady, Srobarova 50, 100 34, Prague, Czech Republic
| | - Vladimir Bobek
- Department of Laboratory Genetics, University Hospital Kralovske Vinohrady, Srobarova 50, 100 34, Prague, Czech Republic. .,Department of Surgery, University Hospital Motol, V Uvalu 84, 150 06, Prague, Czech Republic. .,Faculty of Medicine, Charles University, V Uvalu 84, 150 06, Prague, Czech Republic. .,Department of Thoracic Surgery, Masaryk´s Hospital, Krajska zdravotni a.s., Socialni pece 3316/12A, 40113, Usti Nad Labem, Czech Republic. .,Department of Histology and Embryology, Wroclaw Medical University, Wybrzeże Ludwika Pasteura 1, 50-367, Wrocław, Poland.
| |
Collapse
|
33
|
Su DW, Nieva J. Biophysical technologies for understanding circulating tumor cell biology and metastasis. Transl Lung Cancer Res 2017; 6:473-485. [PMID: 28904890 DOI: 10.21037/tlcr.2017.05.08] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
An understanding of cancer evolution in lung cancer with its associated resistance to therapy can only be achieved with repeated sampling and analysis of the cancer. Given the high risks and costs associated with repeat physical biopsy, alternative technologies must be applied. Several modalities exist for analysis and re-analysis of cancer biology. Among them are the CellSearch platform, the CTC chip, and the high-definition CTC platform. While the former is primarily able to provide prognosticating information in the form of CTC enumeration, the latter two have the advantage of serving as a platform to study tumor biology. Techniques for analysis of single cell genomics, as well as protein expression on a single cell basis provide scientists with the capacity to understand cancer cell populations as a collection of individual cells, rather than as an average of all cells. A multimodal combination of circulating tumor DNAs (ctDNAs), CTCs, proteomics, and CTC-derived xenografts (CDXs) to create computational models useful in diagnosis, prognostication, and predictiveness to treatment is likely the future of tailoring individualized cancer care.
Collapse
Affiliation(s)
- Derrick W Su
- Norris Cancer Center, University of Southern California, Los Angeles, USA
| | - Jorge Nieva
- Norris Cancer Center, University of Southern California, Los Angeles, USA
| |
Collapse
|
34
|
Lampignano R, Schneck H, Neumann M, Fehm T, Neubauer H. Enrichment, Isolation and Molecular Characterization of EpCAM-Negative Circulating Tumor Cells. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2017; 994:181-203. [PMID: 28560675 DOI: 10.1007/978-3-319-55947-6_10] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
The presence of EpCAM-positive circulating tumor cells (CTCs) in the peripheral blood is associated with poor clinical outcomes in breast, colorectal and prostate cancer, as well as the prognosis of other tumor types. In addition, recent studies have suggested that the presence of CTCs undergoing epithelial-to-mesenchymal transition and, as such, may exhibit reduced or no expression of epithelial proteins e.g. EpCAM, might be related to disease progression in metastatic breast cancer (MBC) patients. Analyzing the neoplastic nature of this EpCAM-low/negative (EpCAM-neg) subpopulation remains an open issue as the current standard detection methods for CTCs are not efficient at identifying this subpopulation of cells. The possible association of EpCAM-neg CTCs with EpCAM-positive (EpCAM-pos) CTCs and role in the clinicopathological features and prognosis of MBC patients has still to be demonstrated. Several technologies have been developed and are currently being tested for the identification and the downstream analyses of EpCAM-pos CTCs. These technologies can be adapted and implemented into workflows to isolate and investigate EpCAM-neg cells to understand their biology and clinical relevance. This chapter will endeavour to explain the rationale behind the identification and analyses of all CTC subgroups, as well as to review the current strategies employed to enrich, isolate and characterize EpCAM-negative CTCs. Finally, the latest findings in the field will briefly be discussed with regard to their clinical relevance.
Collapse
Affiliation(s)
- Rita Lampignano
- Department of Obstetrics and Gynecology, University Hospital and Medical Faculty of the Heinrich-Heine University Duesseldorf, Duesseldorf, Germany
| | - Helen Schneck
- Department of Obstetrics and Gynecology, University Hospital and Medical Faculty of the Heinrich-Heine University Duesseldorf, Duesseldorf, Germany
| | - Martin Neumann
- Department of Obstetrics and Gynecology, University Hospital and Medical Faculty of the Heinrich-Heine University Duesseldorf, Duesseldorf, Germany
| | - Tanja Fehm
- Department of Obstetrics and Gynecology, University Hospital and Medical Faculty of the Heinrich-Heine University Duesseldorf, Duesseldorf, Germany
| | - Hans Neubauer
- Forschungslabore der Frauenklinik des, Universitätsklinikums Düsseldorf, Life Science Center, Merowingerplatz 1A, 40225, Düsseldorf, Germany.
| |
Collapse
|
35
|
Bulfoni M, Turetta M, Del Ben F, Di Loreto C, Beltrami AP, Cesselli D. Dissecting the Heterogeneity of Circulating Tumor Cells in Metastatic Breast Cancer: Going Far Beyond the Needle in the Haystack. Int J Mol Sci 2016; 17:ijms17101775. [PMID: 27783057 PMCID: PMC5085799 DOI: 10.3390/ijms17101775] [Citation(s) in RCA: 48] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2016] [Revised: 10/08/2016] [Accepted: 10/17/2016] [Indexed: 02/06/2023] Open
Abstract
Although the enumeration of circulating tumor cells (CTC) defined as expressing both epithelial cell adhesion molecule and cytokeratins (EpCAM+/CK+) can predict prognosis and response to therapy in metastatic breast, colon and prostate cancer, its clinical utility (i.e., the ability to improve patient outcome by guiding therapy) has not yet been proven in clinical trials. Therefore, scientists are now focusing on the molecular characterization of CTC as a way to explore its possible use as a “surrogate” of tumor tissues to non-invasively assess the genomic landscape of the cancer and its evolution during treatment. Additionally, evidences confirm the existence of CTC in epithelial-to-mesenchymal transition (EMT) characterized by a variable loss of epithelial markers. Since the EMT process can originate cells with enhanced invasiveness, stemness and drug-resistance, the enumeration and characterization of this population, perhaps the one truly responsible of tumor recurrence and progression, could be more clinically useful. For these reasons, several devices able to capture CTC independently from the expression of epithelial markers have been developed. In this review, we will describe the types of heterogeneity so far identified and the key role played by the epithelial-to-mesenchymal transition in driving CTC heterogeneity. The clinical relevance of detecting CTC-heterogeneity will be discussed as well.
Collapse
Affiliation(s)
- Michela Bulfoni
- Department of Medical and Biological Sciences, University of Udine, Piazzale M. Kolbe 4, 33100 Udine, Italy.
| | - Matteo Turetta
- Department of Medical and Biological Sciences, University of Udine, Piazzale M. Kolbe 4, 33100 Udine, Italy.
| | - Fabio Del Ben
- Department of Clinical Pathology, CRO Aviano National Cancer Institute, via F. Gallini 2, 33081 Aviano, Italy.
| | - Carla Di Loreto
- Department of Medical and Biological Sciences, University of Udine, Piazzale M. Kolbe 4, 33100 Udine, Italy.
- Institute of Pathology, University Hospital of Udine-ASUIUD, Piazzale Santa Maria della Misericordia 15, 33100 Udine, Italy.
| | - Antonio Paolo Beltrami
- Department of Medical and Biological Sciences, University of Udine, Piazzale M. Kolbe 4, 33100 Udine, Italy.
| | - Daniela Cesselli
- Department of Medical and Biological Sciences, University of Udine, Piazzale M. Kolbe 4, 33100 Udine, Italy.
| |
Collapse
|
36
|
Lee JS, Magbanua MJM, Park JW. Circulating tumor cells in breast cancer: applications in personalized medicine. Breast Cancer Res Treat 2016; 160:411-424. [DOI: 10.1007/s10549-016-4014-6] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2016] [Accepted: 10/08/2016] [Indexed: 12/11/2022]
|
37
|
Aktas B, Kasimir-Bauer S, Müller V, Janni W, Fehm T, Wallwiener D, Pantel K, Tewes M. Comparison of the HER2, estrogen and progesterone receptor expression profile of primary tumor, metastases and circulating tumor cells in metastatic breast cancer patients. BMC Cancer 2016; 16:522. [PMID: 27456970 PMCID: PMC4960681 DOI: 10.1186/s12885-016-2587-4] [Citation(s) in RCA: 64] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2015] [Accepted: 07/20/2016] [Indexed: 11/18/2022] Open
Abstract
Background The expression of HER2, estrogen (ER) and progesterone (PR) receptor can change during the course of the disease in breast cancer (BC). Therefore, reassessment of these markers at the time of disease progression might help to optimize treatment decisions. In this context, characterization of circulating tumor cells (CTCs) could be of relevance since metastatic tissue may be difficult to obtain for repeated analysis. Here we compared HER2/ER/PR expression profiles of primary tumors, metastases and CTCs. Methods Ninety-six patients with metastatic BC from seven University BC Centers in Germany were enrolled in this study. Blood was obtained at the time of first diagnosis of metastatic disease or disease progression and analyzed for CTCs using the AdnaTest BreastCancer (QIAGEN Hannover GmbH, Germany) for the expression of EpCAM, MUC-1, HER2, ER and PR. HER2 expression on CTCs was additionally assessed by immunocytochemistry using the CellSearch® assay. Results The detection rate for CTCs using the AdnaTest was 43 % (36/84 patients) with the expression rates of 50 % for HER2 (18/36 patients), 19 % for ER (7/36 patients) and 8 % for PR (3/36 patients), respectively. Primary tumors and CTCs displayed a concordant HER2, ER and PR status in 59 % (p = 0.262), 39 % (p = 0.51) and 44 % (p = 0.62) of cases, respectively. For metastases and CTCs, the concordance values were 67 % for HER2 (p = 0.04), 43 % for ER (p = 0.16) and 46 % for PR (p = 0.6). Using the CellSearch® assay, the CTC-positivity rate was 53 % (42/79 patients) with HER2 expressed in 29 % (12/42) of the patients. No significant concordance (58 % and 53 %) was found when HER2 on CTCs was compared with HER2 on primary tumors (p = 0.24) and metastases (p = 0.34). Interestingly, primary tumors and metastases were highly concordant for HER2 (84 %, p = 1.13E-08), ER (90 %, p = 3.26E-10) and PR (83 %, p = 2.09E-09) and ER-and PR-positive metastases were significantly found to be of visceral origin (p = 0.03, p = 0.02). Conclusion Here we demonstrate that the molecular detection of HER2 overexpression in CTC is predictive of the HER2 status on metastases. Detailed analysis of ER and PR expression rates in tissue samples and CTCs may provide useful information for making treatment decisions.
Collapse
Affiliation(s)
- Bahriye Aktas
- Department of Gynecology and Obstetrics, University of Duisburg-Essen, Essen, Germany.
| | - Sabine Kasimir-Bauer
- Department of Gynecology and Obstetrics, University of Duisburg-Essen, Essen, Germany
| | - Volkmar Müller
- Department of Gynecology and Obstetrics, University Hospital Hamburg-Eppendorf, Hamburg, Germany
| | - Wolfgang Janni
- Department of Gynecology and Obstetrics, University Hospital Ulm, Ulm, Germany
| | - Tanja Fehm
- Department of Gynecology and Obstetrics, University Hospital Duesseldorf, Duesseldorf, Germany
| | - Diethelm Wallwiener
- Department of Gynecology and Obstetrics, University Hospital Tuebingen, Tuebingen, Germany
| | - Klaus Pantel
- Institut of Tumor Biology, Center of Experimental Medicine, University Hospital Hamburg-Eppendorf, Hamburg, Germany
| | - Mitra Tewes
- Department of Internal Medicine (Cancer Research), University Hospital Essen, Essen, Germany
| | | |
Collapse
|
38
|
Tachtsidis A, McInnes LM, Jacobsen N, Thompson EW, Saunders CM. Minimal residual disease in breast cancer: an overview of circulating and disseminated tumour cells. Clin Exp Metastasis 2016; 33:521-50. [PMID: 27189371 PMCID: PMC4947105 DOI: 10.1007/s10585-016-9796-8] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2015] [Accepted: 04/22/2016] [Indexed: 12/11/2022]
Abstract
Within the field of cancer research, focus on the study of minimal residual disease (MRD) in the context of carcinoma has grown exponentially over the past several years. MRD encompasses circulating tumour cells (CTCs)—cancer cells on the move via the circulatory or lymphatic system, disseminated tumour cells (DTCs)—cancer cells which have escaped into a distant site (most studies have focused on bone marrow), and resistant cancer cells surviving therapy—be they local or distant, all of which may ultimately give rise to local relapse or overt metastasis. Initial studies simply recorded the presence and number of CTCs and DTCs; however recent advances are allowing assessment of the relationship between their persistence, patient prognosis and the biological properties of MRD, leading to a better understanding of the metastatic process. Technological developments for the isolation and analysis of circulating and disseminated tumour cells continue to emerge, creating new opportunities to monitor disease progression and perhaps alter disease outcome. This review outlines our knowledge to date on both measurement and categorisation of MRD in the form of CTCs and DTCs with respect to how this relates to cancer outcomes, and the hurdles and future of research into both CTCs and DTCs.
Collapse
Affiliation(s)
- A Tachtsidis
- St. Vincent's Institute, Melbourne, VIC, Australia
- University of Melbourne, Department of Surgery, St. Vincent's Hospital, Melbourne, VIC, Australia
| | - L M McInnes
- School of Surgery, The University of Western Australia, Perth, WA, Australia
| | - N Jacobsen
- School of Surgery, The University of Western Australia, Perth, WA, Australia
| | - E W Thompson
- University of Melbourne, Department of Surgery, St. Vincent's Hospital, Melbourne, VIC, Australia
- Institute of Health and Biomedical Innovation and School of Biomedical Sciences, Queensland University of Technology, Brisbane, QLD, Australia
- Translational Research Institute, Woolloongabba, QLD, Australia
| | - C M Saunders
- School of Surgery, The University of Western Australia, Perth, WA, Australia.
| |
Collapse
|
39
|
Assi HI, Assi RE, El Saghir NS. Emerging Biomarkers of the Future: Changing Clinical Practice for 2020. CURRENT BREAST CANCER REPORTS 2016. [DOI: 10.1007/s12609-016-0214-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
|
40
|
Wang S, Thomas A, Lee E, Yang S, Cheng X, Liu Y. Highly efficient and selective isolation of rare tumor cells using a microfluidic chip with wavy-herringbone micro-patterned surfaces. Analyst 2016; 141:2228-37. [PMID: 26907962 PMCID: PMC5051543 DOI: 10.1039/c6an00236f] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Circulating tumor cells (CTCs) in peripheral blood have been recognized as a general biomarker for diagnosing cancer and providing guidance for personalized treatments. Yet due to their rarity, the challenge for their clinical utility lies in the efficient isolation while avoiding the capture of other non-targeted white blood cells (WBCs). In this paper, a wavy-herringbone (HB) microfluidic chip coated with antibody directly against epithelial cell adhesion molecule (anti-EpCAM) was developed for highly efficient and selective isolation of tumor cells from tumor cell-spiked whole blood samples. By extending the concept of the hallmark HB-Chip in the literature, the wavy-HB chip not only achieves high capture efficiency (up to 85.0%) by micro-vortexes induced by HB structures, but also achieves high purity (up to 39.4%) due to the smooth wavy microstructures. These smooth wavy-HB structures eliminate the ultra-low shear rate regions in the traditional grooved-HB structures that lead to non-specific trapping of cells. Compared with the grooved-HB chip with sharp corners, the wavy-HB chip shows significantly higher purity while maintaining similarly high capture efficiency. Furthermore, the wavy-HB chip has up to 11% higher captured cell viability over the grooved-HB chip. The distributions of tumor cells and WBCs along the grooves and waves are investigated to help understand the mechanisms behind the better performance of the wavy-HB chip. The wavy-HB chip may serve as a promising platform for CTC capture and cancer diagnosis.
Collapse
Affiliation(s)
- Shunqiang Wang
- Department of Mechanical Engineering and Mechanics, Lehigh University, Bethlehem, PA 18015, USA.
| | - Antony Thomas
- Bioengineering Program, Lehigh University, Bethlehem, PA 18015, USA
| | - Elaine Lee
- Lawrence Livermore National Laboratory, Livermore, California 94550, USA and Department of Materials Science and Engineering, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Shu Yang
- Department of Materials Science and Engineering, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Xuanhong Cheng
- Bioengineering Program, Lehigh University, Bethlehem, PA 18015, USA and Department of Materials Science and Engineering, Lehigh University, Bethlehem, PA 18015, USA
| | - Yaling Liu
- Department of Mechanical Engineering and Mechanics, Lehigh University, Bethlehem, PA 18015, USA. and Bioengineering Program, Lehigh University, Bethlehem, PA 18015, USA
| |
Collapse
|
41
|
Kaifi JT, Li G, Clawson G, Kimchi ET, Staveley-O'Carroll KF. Perioperative circulating tumor cell detection: Current perspectives. Cancer Biol Ther 2016; 17:859-69. [PMID: 27045201 DOI: 10.1080/15384047.2016.1167296] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Primary cancer resections and in selected cases surgical metastasectomies significantly improve survival, however many patients develop recurrences. Circulating tumor cells (CTCs) function as an independent marker that could be used in the prognostication of different cancers. Sampling of blood and bone marrow compartments during cancer resections is a unique opportunity to increase individual tumor cell capture efficiency. This review will address the diagnostic and therapeutic potentials of perioperative tumor isolation and highlight the focus of future studies on characterization of single disseminated cancer cells to identify targets for molecular therapy and immune escape mechanisms.
Collapse
Affiliation(s)
- Jussuf T Kaifi
- a Hugh E. Stephenson Jr., M.D. , Department of Surgery , University of Missouri , Columbia , MO , USA.,b Ellis Fischel Cancer Center , University of Missouri , Columbia , MO , USA
| | - Guangfu Li
- a Hugh E. Stephenson Jr., M.D. , Department of Surgery , University of Missouri , Columbia , MO , USA.,c Department of Molecular Microbiology and Immunology , University of Missouri , Columbia , MO , USA
| | - Gary Clawson
- d Gittlen Cancer Research Foundation and Department of Pathology , Materials Research Institute, Penn State College of Medicine, Pennsylvania State University , Hershey , PA , USA
| | - Eric T Kimchi
- a Hugh E. Stephenson Jr., M.D. , Department of Surgery , University of Missouri , Columbia , MO , USA.,b Ellis Fischel Cancer Center , University of Missouri , Columbia , MO , USA.,c Department of Molecular Microbiology and Immunology , University of Missouri , Columbia , MO , USA
| | - Kevin F Staveley-O'Carroll
- a Hugh E. Stephenson Jr., M.D. , Department of Surgery , University of Missouri , Columbia , MO , USA.,b Ellis Fischel Cancer Center , University of Missouri , Columbia , MO , USA.,c Department of Molecular Microbiology and Immunology , University of Missouri , Columbia , MO , USA
| |
Collapse
|
42
|
Caceres G, Puskas JA, Magliocco AM. Circulating Tumor Cells: A Window Into Tumor Development and Therapeutic Effectiveness. Cancer Control 2016; 22:167-76. [PMID: 26068761 DOI: 10.1177/107327481502200207] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
BACKGROUND Circulating tumor cells (CTCs) are an important diagnostic tool for understanding the metastatic process and the development of cancer. METHODS This review covers the background, relevance, and potential limitations of CTCs as a measurement of cancer progression and how information derived from CTCs may affect treatment efficacy. It also highlights the difficulties of characterizing these rare cells due to the limited cell surface molecules unique to CTCs and each particular type of cancer. RESULTS The analysis of cancer in real time, through the measure of the number of CTCs in a " liquid" biopsy specimen, gives us the ability to monitor the therapeutic efficacy of treatments and possibly the metastatic potential of a tumor. CONCLUSIONS Through novel and innovative techniques yielding encouraging results, including microfluidic techniques, isolating and molecularly analyzing CTCs are becoming a reality. CTCs hold promise for understanding how tumors work and potentially aiding in their demise.
Collapse
Affiliation(s)
- Gisela Caceres
- Department of Anatomic Pathology, Moffitt Cancer Center, Tampa, FL 33612, USA.
| | | | | |
Collapse
|
43
|
Kaifi JT, Kunkel M, Das A, Harouaka RA, Dicker DT, Li G, Zhu J, Clawson GA, Yang Z, Reed MF, Gusani NJ, Kimchi ET, Staveley-O'Carroll KF, Zheng SY, El-Deiry WS. Circulating tumor cell isolation during resection of colorectal cancer lung and liver metastases: a prospective trial with different detection techniques. Cancer Biol Ther 2016; 16:699-708. [PMID: 25807199 DOI: 10.1080/15384047.2015.1030556] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND Colorectal cancer (CRC) metastasectomy improves survival, however most patient develop recurrences. Circulating tumor cells (CTCs) are an independent prognostic marker in stage IV CRC. We hypothesized that CTCs can be enriched during metastasectomy applying different isolation techniques. METHODS 25 CRC patients undergoing liver (16 (64%)) or lung (9 (36%)) metastasectomy were prospectively enrolled (clinicaltrial.gov identifier: NCT01722903). Central venous (liver) or radial artery (lung) tumor outflow blood (7.5 ml) was collected at incision, during resection, 30 min after resection, and on postoperative day (POD) 1. CTCs were quantified with 1. EpCAM-based CellSearch® system and 2. size-based isolation with a novel filter device (FMSA). CTCs were immunohistochemically identified using CellSearch®'s criteria (cytokeratin 8/18/19+, CD45- cells containing a nucleus (DAPI+)). CTCs were also enriched with a centrifugation technique (OncoQuick®). RESULTS CTC numbers peaked during the resection with the FMSA in contrast to CellSearch® (mean CTC number during resection: FMSA: 22.56 (SEM 7.48) (p = 0.0281), CellSearch®: 0.87 (SEM ± 0.44) (p = 0.3018)). Comparing the 2 techniques, CTC quantity was significantly higher with the FMSA device (range 0-101) than CellSearch® (range 0-9) at each of the 4 time points examined (P < 0.05). Immunofluorescence staining of cultured CTCs revealed that CTCs have a combined epithelial (CK8/18/19) and macrophage (CD45/CD14) phenotype. CONCLUSIONS Blood sampling during CRC metastasis resection is an opportunity to increase CTC capture efficiency. CTC isolation with the FMSA yields more CTCs than the CellSearch® system. Future studies should focus on characterization of single CTCs to identify targets for molecular therapy and immune escape mechanisms of cancer cells.
Collapse
Affiliation(s)
- Jussuf T Kaifi
- a Program for Liver, Pancreas and Foregut (Lung & Esophageal) Tumors; Department of Surgery (Surgical Oncology)
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
44
|
Kaifi JT, Kunkel M, Dicker DT, Joude J, Allen JE, Das A, Zhu J, Yang Z, Sarwani NE, Li G, Staveley-O'Carroll KF, El-Deiry WS. Circulating tumor cell levels are elevated in colorectal cancer patients with high tumor burden in the liver. Cancer Biol Ther 2016; 16:690-8. [PMID: 25785486 DOI: 10.1080/15384047.2015.1026508] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND Metastatic spread is the most common cause of cancer-related death in colorectal cancer (CRC) patients, with the liver being the mostly affected organ. Circulating tumor cells (CTCs) are a prognostic marker in stage IV CRC. We hypothesized that tumor burden in the liver correlates with CTC quantity. METHODS Blood (7.5 ml) was prospectively collected from 24 patients with novel stage IV CRC diagnosis. Baseline EpCAM+ CTCs were analyzed with the FDA-approved CellSearch® system. Clinicopathological data were collected, and hepatic tumor burden was determined by radiographic liver volumetry with contrast-enhanced CT scans. CRC primary tumors were immunohistochemically stained for EpCAM expression with BerEP4 monoclonal antibody. Statistical analyses were performed using 2-sample T-test, non-parametric Wilcoxon Rank-Sum test, and Fisher's exact test. RESULTS CTCs were detected n 17 (71%) of 24 patients. The overall mean CTC number as determined by EpCAM-based CellSearch® detection was 6.3 (SEM 2.9). High baseline CTC numbers (≥3) correlated significantly with a high tumor/liver ratio (≥30%), and with high serum CEA levels, as determined by two-sample T-test on log-transformed data and by Fisher's Exact test on categorical data analysis (P < 0.05). The CRC primary tumors were consistently expressing EpCAM by immunostaining. CONCLUSIONS High tumor burden in the liver and high baseline serum CEA levels are associated with high number of baseline CTCs in stage IV CRC patients. Future studies should further investigate the biological role and expression patterns of single CTCs in cancer patients to further improve personalized treatment strategies.
Collapse
|
45
|
Diagnostic and Prognostic Value of Circulating Tumor Cells in Head and Neck Squamous Cell Carcinoma: a systematic review and meta-analysis. Sci Rep 2016; 6:20210. [PMID: 26831813 PMCID: PMC4735798 DOI: 10.1038/srep20210] [Citation(s) in RCA: 62] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2015] [Accepted: 12/23/2015] [Indexed: 12/13/2022] Open
Abstract
Several techniques have been developed to detect circulating tumor cells (CTC) in patients with head and neck squamous cell carcinoma (HNSCC), but their diagnostic and prognostic value are not yet fully established. A computerized retrieval of literatures was conducted without time restrictions using the electronic database in December 2014. Diagnostic accuracy variables were pooled and analyzed by the Meta-DiSc software. Engauge Digitizer and Stata software were used for pooled survival analysis. Twenty-two retrieved studies were eligible for systematic review, of which 9 conformed for the diagnostic test meta-analysis and 5 for the prognostic analysis. Subgroup analysis showed 24.6% pooled sensitivity and 100% pooled specificity of detections by using positive selection strategy, which moreover presented low heterogeneity. The presence of CTC was significantly associated with shorter disease free survival (DFS, HR 4.62, 95% CI 2.51-8.52). In conclusion, current evidence identifies the CTC detection assay as an extremely specific, but low sensitive test in HNSCC. Also, the presence of CTC indicates a worse DFS.
Collapse
|
46
|
Witzel I, Oliveira-Ferrer L, Pantel K, Müller V, Wikman H. Breast cancer brain metastases: biology and new clinical perspectives. Breast Cancer Res 2016; 18:8. [PMID: 26781299 PMCID: PMC4717619 DOI: 10.1186/s13058-015-0665-1] [Citation(s) in RCA: 216] [Impact Index Per Article: 24.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Because of improvements in the treatment of patients with metastatic breast cancer, the development of brain metastases (BM) has become a major limitation of life expectancy and quality of life for many breast cancer patients. The improvement of management strategies for BM is thus an important clinical challenge, especially among high-risk patients such as human epidermal growth factor receptor 2-positive and triple-negative patients. However, the formation of BM as a multistep process is thus far poorly understood. To grow in the brain, single tumor cells must pass through the tight blood–brain barrier (BBB). The BBB represents an obstacle for circulating tumor cells entering the brain, but it also plays a protective role against immune cell and toxic agents once metastatic cells have colonized the cerebral compartment. Furthermore, animal studies have shown that, after passing the BBB, the tumor cells not only require close contact with endothelial cells but also interact closely with many different brain residential cells. Thus, in addition to a genetic predisposition of the tumor cells, cellular adaptation processes within the new microenvironment may also determine the ability of a tumor cell to metastasize. In this review, we summarize the biology of breast cancer that has spread into the brain and discuss the implications for current and potential future treatment strategies.
Collapse
Affiliation(s)
- Isabell Witzel
- Department of Gynecology, University Medical Center Hamburg-Eppendorf, Martinistraße 52, 20246, Hamburg, Germany.
| | - Leticia Oliveira-Ferrer
- Department of Gynecology, University Medical Center Hamburg-Eppendorf, Martinistraße 52, 20246, Hamburg, Germany.
| | - Klaus Pantel
- Institute of Tumour Biology, University Medical Center Hamburg-Eppendorf, Center of Experimental Medicine, Martinistraße 52, 20246, Hamburg, Germany.
| | - Volkmar Müller
- Department of Gynecology, University Medical Center Hamburg-Eppendorf, Martinistraße 52, 20246, Hamburg, Germany.
| | - Harriet Wikman
- Institute of Tumour Biology, University Medical Center Hamburg-Eppendorf, Center of Experimental Medicine, Martinistraße 52, 20246, Hamburg, Germany.
| |
Collapse
|
47
|
Su Y, Pogash TJ, Nguyen TD, Russo J. Development and characterization of two human triple-negative breast cancer cell lines with highly tumorigenic and metastatic capabilities. Cancer Med 2016; 5:558-73. [PMID: 26775583 PMCID: PMC4799943 DOI: 10.1002/cam4.616] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2015] [Revised: 11/20/2015] [Accepted: 11/23/2015] [Indexed: 12/12/2022] Open
Abstract
Triple-negative breast cancer (TNBC) is a group of cancer with high diversity, limited therapies, and poor prognosis. TNBC cell lines and animal models provide effective tools for studies and drug discovery. Here, we report the development of two TNBC cell lines (XtMCF and LmMCF) based on our existing cell model that consists of normal breast epithelial cell line MCF10F, estradiol-transformed cells trMCF, and Boyden chamber-selected tumorigenic cells bsMCF. The XtMCF and LmMCF cell line were derived from xenograft and lung metastasis of bsMCF cells, respectively. The bsMCF, XtMCF, and LmMCF cells have undergone epithelial-mesenchymal transition (EMT), exhibiting a mesenchymal-like feature. In vivo studies showed XtMCF and LmMCF cells were highly tumorigenic and metastatic. The injection of 5 × 10(4) cells to CB17/SCID mice mammary fat pad produced xenografts in 9/9 mice and tumors reached 10 millimeters in diameter in 5 weeks. The injection of 1 × 10(6) XtMCF or 8 × 10(4) LmMCF cells into the mice tail vein was sufficient to form extensive lung metastases in 4 weeks. The two new cell lines exhibited CD44(+) /CD49f(+) and CD44(+) /EpCAM(+) cancer stem cell (CSC) characteristics, and the EGF-like domain of EpCAM was cleaved off. Together with the normal and early transformed counterparts, herein we provide a complete cancer model for the study of initiation, evolution, and identification of new therapeutics for TNBC. The finding that EGF-like domain of EpCAM was cleaved off in cells which have undergone EMT suggests this cleavage may be involved in the EMT process and the cancer stem cell properties of these cells.
Collapse
Affiliation(s)
- Yanrong Su
- The Irma H. Russo, MD Breast Cancer Research Laboratory, Fox Chase Cancer Center-Temple University Health System, Philadelphia, PA, 19111, USA
| | - Thomas J Pogash
- The Irma H. Russo, MD Breast Cancer Research Laboratory, Fox Chase Cancer Center-Temple University Health System, Philadelphia, PA, 19111, USA
| | - Theresa D Nguyen
- The Irma H. Russo, MD Breast Cancer Research Laboratory, Fox Chase Cancer Center-Temple University Health System, Philadelphia, PA, 19111, USA
| | - Jose Russo
- The Irma H. Russo, MD Breast Cancer Research Laboratory, Fox Chase Cancer Center-Temple University Health System, Philadelphia, PA, 19111, USA
| |
Collapse
|
48
|
Moon PG, Lee JE, Cho YE, Lee SJ, Jung JH, Chae YS, Bae HI, Kim YB, Kim IS, Park HY, Baek MC. Identification of Developmental Endothelial Locus-1 on Circulating Extracellular Vesicles as a Novel Biomarker for Early Breast Cancer Detection. Clin Cancer Res 2015; 22:1757-66. [DOI: 10.1158/1078-0432.ccr-15-0654] [Citation(s) in RCA: 124] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2015] [Accepted: 10/21/2015] [Indexed: 11/16/2022]
|
49
|
Welinder C, Jansson B, Lindell G, Wenner J. Cytokeratin 20 improves the detection of circulating tumor cells in patients with colorectal cancer. Cancer Lett 2015; 358:43-6. [DOI: 10.1016/j.canlet.2014.12.024] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2014] [Revised: 12/11/2014] [Accepted: 12/11/2014] [Indexed: 12/26/2022]
|
50
|
Clinical phenotypes associated with circulating tumor cell enumeration in metastatic castration–resistant prostate cancer. Urol Oncol 2015; 33:110.e1-9. [DOI: 10.1016/j.urolonc.2014.09.002] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2014] [Revised: 09/04/2014] [Accepted: 09/04/2014] [Indexed: 12/29/2022]
|