1
|
Topa J, Żaczek AJ, Markiewicz A. Isolation of Viable Epithelial and Mesenchymal Circulating Tumor Cells from Breast Cancer Patients. Methods Mol Biol 2024; 2752:43-52. [PMID: 38194026 DOI: 10.1007/978-1-0716-3621-3_3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2024]
Abstract
Circulating tumor cells (CTCs) undergoing epithelial-mesenchymal transition (EMT) may exhibit more aggressive features than epithelial CTCs and are more frequently observed during disease progression. Therefore, detection and characterization of both epithelial and mesenchymal CTCs in cancer patients are urgently needed to allow for a better understanding of the metastatic process and more effective treatment. Here we describe a method for detection and isolation of viable epithelial and mesenchymal CTCs from peripheral blood of breast cancer patients. The method is based on density gradient centrifugation, multiplex immunofluorescent staining, and negative anti-CD45 selection. Cells obtained after the procedure are suitable for genomic or transcriptomic profiling, and they can also be isolated by micromanipulation for single-cell analysis.
Collapse
Affiliation(s)
- Justyna Topa
- Laboratory of Translational Oncology, Intercollegiate Faculty of Biotechnology, University of Gdansk and Medical University of Gdansk, Gdansk, Poland
| | - Anna J Żaczek
- Laboratory of Translational Oncology, Intercollegiate Faculty of Biotechnology, University of Gdansk and Medical University of Gdansk, Gdansk, Poland
| | - Aleksandra Markiewicz
- Laboratory of Translational Oncology, Intercollegiate Faculty of Biotechnology, University of Gdansk and Medical University of Gdansk, Gdansk, Poland.
| |
Collapse
|
2
|
Qiu J, Qian D, Jiang Y, Meng L, Huang L. Circulating tumor biomarkers in early-stage breast cancer: characteristics, detection, and clinical developments. Front Oncol 2023; 13:1288077. [PMID: 37941557 PMCID: PMC10628786 DOI: 10.3389/fonc.2023.1288077] [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: 09/04/2023] [Accepted: 10/09/2023] [Indexed: 11/10/2023] Open
Abstract
Breast cancer is the most common form of cancer in women, contributing to high rates of morbidity and mortality owing to the ability of these tumors to metastasize via the vascular system even in the early stages of progression. While ultrasonography and mammography have enabled the more reliable detection of early-stage breast cancer, these approaches entail high rates of false positive and false negative results Mammograms also expose patients to radiation, raising clinical concerns. As such, there is substantial interest in the development of more accurate and efficacious approaches to diagnosing breast cancer in its early stages when patients are more likely to benefit from curative treatment efforts. Blood-based biomarkers derived from the tumor microenvironment (TME) have frequently been studied as candidate targets that can enable tumor detection when used for patient screening. Through these efforts, many promising biomarkers including tumor antigens, circulating tumor cell clusters, microRNAs, extracellular vesicles, circulating tumor DNA, metabolites, and lipids have emerged as targets that may enable the detection of breast tumors at various stages of progression. This review provides a systematic overview of the TME characteristics of early breast cancer, together with details on current approaches to detecting blood-based biomarkers in affected patients. The limitations, challenges, and prospects associated with different experimental and clinical platforms employed in this context are also discussed at length.
Collapse
Affiliation(s)
- Jie Qiu
- Department of Breast and Thyroid Surgery, Shaoxing People’s Hospital, Shaoxing, Zhejiang, China
| | - Da Qian
- Department of Burn and Plastic Surgery-Hand Surgery, Changshu Hospital Affiliated to Soochow University, Changshu No.1 People’s Hospital, Changshu, Jiangsu, China
| | - Yuancong Jiang
- Department of Breast and Thyroid Surgery, Shaoxing People’s Hospital, Shaoxing, Zhejiang, China
| | - Liwei Meng
- Department of Breast and Thyroid Surgery, Shaoxing People’s Hospital, Shaoxing, Zhejiang, China
| | - Liming Huang
- Department of Breast and Thyroid Surgery, Shaoxing People’s Hospital, Shaoxing, Zhejiang, China
| |
Collapse
|
3
|
Sun J, Huang X, Chen J, Xiang R, Ke X, Lin S, Xuan W, Liu S, Cao Z, Sun L. Recent advances in deformation-assisted microfluidic cell sorting technologies. Analyst 2023; 148:4922-4938. [PMID: 37743834 DOI: 10.1039/d3an01150j] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/26/2023]
Abstract
Cell sorting is an essential prerequisite for cell research and has great value in life science and clinical studies. Among the many microfluidic cell sorting technologies, label-free methods based on the size of different cell types have been widely studied. However, the heterogeneity in size for cells of the same type and the inevitable size overlap between different types of cells would result in performance degradation in size-based sorting. To tackle such challenges, deformation-assisted technologies are receiving more attention recently. Cell deformability is an inherent biophysical marker of cells that reflects the changes in their internal structures and physiological states. It provides additional dimensional information for cell sorting besides size. Therefore, in this review, we summarize the recent advances in deformation-assisted microfluidic cell sorting technologies. According to how the deformability is characterized and the form in which the force acts, the technologies can be divided into two categories: (1) the indirect category including transit-time-based and image-based methods, and (2) the direct category including microstructure-based and hydrodynamics-based methods. Finally, the separation performance and the application scenarios of each method, the existing challenges and future outlook are discussed. Deformation-assisted microfluidic cell sorting technologies are expected to realize greater potential in the label-free analysis of cells.
Collapse
Affiliation(s)
- Jingjing Sun
- Ministry of Education Key Lab of RF Circuits and Systems, Hangzhou Dianzi University, China.
| | - Xiwei Huang
- Ministry of Education Key Lab of RF Circuits and Systems, Hangzhou Dianzi University, China.
| | - Jin Chen
- Ministry of Education Key Lab of RF Circuits and Systems, Hangzhou Dianzi University, China.
| | - Rikui Xiang
- Ministry of Education Key Lab of RF Circuits and Systems, Hangzhou Dianzi University, China.
| | - Xiang Ke
- Ministry of Education Key Lab of RF Circuits and Systems, Hangzhou Dianzi University, China.
| | - Siru Lin
- Ministry of Education Key Lab of RF Circuits and Systems, Hangzhou Dianzi University, China.
| | - Weipeng Xuan
- Ministry of Education Key Lab of RF Circuits and Systems, Hangzhou Dianzi University, China.
| | - Shan Liu
- Sichuan Provincial Key Laboratory for Human Disease Gene Study, Sichuan Academy of Medical Sciences & Sichuan Provincial People's Hospital, China
| | - Zhen Cao
- College of Information Science and Electronic Engineering, Zhejiang University, China
| | - Lingling Sun
- Ministry of Education Key Lab of RF Circuits and Systems, Hangzhou Dianzi University, China.
| |
Collapse
|
4
|
Lin W, Fang J, Wei S, He G, Liu J, Li X, Peng X, Li D, Yang S, Li X, Yang L, Li H. Extracellular vesicle-cell adhesion molecules in tumours: biofunctions and clinical applications. Cell Commun Signal 2023; 21:246. [PMID: 37735659 PMCID: PMC10512615 DOI: 10.1186/s12964-023-01236-8] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2023] [Accepted: 07/18/2023] [Indexed: 09/23/2023] Open
Abstract
Cell adhesion molecule (CAM) is an umbrella term for several families of molecules, including the cadherin family, integrin family, selectin family, immunoglobulin superfamily, and some currently unclassified adhesion molecules. Extracellular vesicles (EVs) are important information mediators in cell-to-cell communication. Recent evidence has confirmed that CAMs transported by EVs interact with recipient cells to influence EV distribution in vivo and regulate multiple cellular processes. This review focuses on the loading of CAMs onto EVs, the roles of CAMs in regulating EV distribution, and the known and possible mechanisms of these actions. Moreover, herein, we summarize the impacts of CAMs transported by EVs to the tumour microenvironment (TME) on the malignant behaviour of tumour cells (proliferation, metastasis, immune escape, and so on). In addition, from the standpoint of clinical applications, the significance and challenges of using of EV-CAMs in the diagnosis and therapy of tumours are discussed. Finally, considering recent advances in the understanding of EV-CAMs, we outline significant challenges in this field that require urgent attention to advance research and promote the clinical applications of EV-CAMs. Video Abstract.
Collapse
Affiliation(s)
- Weikai Lin
- Department of General Surgery, The Fourth Affiliated Hospital, China Medical University, Shenyang, 110032, China
- Shenyang Clinical Medical Research Center for Diagnosis, Treatment and Health Management of Early Digestive Cancer, Shenyang, 110032, China
| | - Jianjun Fang
- Department of General Surgery, The Fourth Affiliated Hospital, China Medical University, Shenyang, 110032, China
- Shenyang Clinical Medical Research Center for Diagnosis, Treatment and Health Management of Early Digestive Cancer, Shenyang, 110032, China
| | - Shibo Wei
- Department of General Surgery, The Fourth Affiliated Hospital, China Medical University, Shenyang, 110032, China
- Shenyang Clinical Medical Research Center for Diagnosis, Treatment and Health Management of Early Digestive Cancer, Shenyang, 110032, China
| | - Guangpeng He
- Department of General Surgery, The Fourth Affiliated Hospital, China Medical University, Shenyang, 110032, China
- Shenyang Clinical Medical Research Center for Diagnosis, Treatment and Health Management of Early Digestive Cancer, Shenyang, 110032, China
| | - Jiaxing Liu
- Department of General Surgery, The Fourth Affiliated Hospital, China Medical University, Shenyang, 110032, China
- Shenyang Clinical Medical Research Center for Diagnosis, Treatment and Health Management of Early Digestive Cancer, Shenyang, 110032, China
| | - Xian Li
- Department of General Surgery, The Fourth Affiliated Hospital, China Medical University, Shenyang, 110032, China
- Shenyang Clinical Medical Research Center for Diagnosis, Treatment and Health Management of Early Digestive Cancer, Shenyang, 110032, China
| | - Xueqiang Peng
- Department of General Surgery, The Fourth Affiliated Hospital, China Medical University, Shenyang, 110032, China
- Shenyang Clinical Medical Research Center for Diagnosis, Treatment and Health Management of Early Digestive Cancer, Shenyang, 110032, China
| | - Dai Li
- Department of General Surgery, The Fourth Affiliated Hospital, China Medical University, Shenyang, 110032, China
- Shenyang Clinical Medical Research Center for Diagnosis, Treatment and Health Management of Early Digestive Cancer, Shenyang, 110032, China
| | - Shuo Yang
- Department of General Surgery, The Fourth Affiliated Hospital, China Medical University, Shenyang, 110032, China
- Shenyang Clinical Medical Research Center for Diagnosis, Treatment and Health Management of Early Digestive Cancer, Shenyang, 110032, China
| | - Xinyu Li
- Department of General Surgery, The Fourth Affiliated Hospital, China Medical University, Shenyang, 110032, China
- Shenyang Clinical Medical Research Center for Diagnosis, Treatment and Health Management of Early Digestive Cancer, Shenyang, 110032, China
| | - Liang Yang
- Department of General Surgery, The Fourth Affiliated Hospital, China Medical University, Shenyang, 110032, China.
- Shenyang Clinical Medical Research Center for Diagnosis, Treatment and Health Management of Early Digestive Cancer, Shenyang, 110032, China.
| | - Hangyu Li
- Department of General Surgery, The Fourth Affiliated Hospital, China Medical University, Shenyang, 110032, China.
- Shenyang Clinical Medical Research Center for Diagnosis, Treatment and Health Management of Early Digestive Cancer, Shenyang, 110032, China.
| |
Collapse
|
5
|
Descamps L, Garcia J, Barthelemy D, Laurenceau E, Payen L, Le Roy D, Deman AL. MagPure chip: an immunomagnetic-based microfluidic device for high purification of circulating tumor cells from liquid biopsies. LAB ON A CHIP 2022; 22:4151-4166. [PMID: 36148526 DOI: 10.1039/d2lc00443g] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/16/2023]
Abstract
The isolation of circulating tumor cells (CTCs) directly from blood, as a liquid biopsy, could lead to a paradigm shift in cancer clinical care by providing an earlier diagnosis, a more accurate prognosis, and personalized treatment. Nevertheless, CTC-specific challenges, including their rarity and heterogeneity, have hampered the wider use of CTCs in clinical studies. Microfluidic-based isolation technologies have emerged as promising tools to circumvent these limitations but still fail to meet the constraints of high purity and short processing time required to ensure compatibility with clinical follow-up. In this study, we developed an immunomagnetic-based microfluidic device, the MagPure chip, to achieve the negative selection of CTCs through the depletion of white blood cells (WBCs) and provide highly purified samples for subsequent analysis. We demonstrate that the MagPure chip depletes all magnetically labeled WBCs (85% of WBCs were successfully labeled) and ensures a CTC recovery rate of 81%. In addition, we show its compatibility with conventional biological studies, including 2D and 3D cell culture, as well as phenotypic and genotypic analyses. Finally, we successfully implemented a two-step separation workflow for whole blood processing by combining a size-based pre-enrichment system (ClearCell FX1®) with the MagPure chip as a subsequent purification step. The total workflow led to high throughput (7.5 mL blood in less than 4 h) and high purity (947 WBCs per mL remaining, 99.99% depletion rate), thus enabling us to quantify CTC heterogeneity in size and tumor marker expression level. This tumor-marker-free liquid biopsy workflow could be used in a clinical context to assess phenotype aggressiveness and the prognosis rate.
Collapse
Affiliation(s)
- Lucie Descamps
- Institut des Nanotechnologies de Lyon, INL UMR5270, Université Claude Bernard Lyon 1, Villeurbanne, France.
| | - Jessica Garcia
- Laboratoire de Biochimie et Biologie Moléculaire, CICLY UR3738, Groupe Hospitalier Sud, Hospices Civils de Lyon, Pierre Bénite, France
| | - David Barthelemy
- Laboratoire de Biochimie et Biologie Moléculaire, CICLY UR3738, Groupe Hospitalier Sud, Hospices Civils de Lyon, Pierre Bénite, France
| | - Emmanuelle Laurenceau
- Institut des Nanotechnologies de Lyon, INL UMR5270, Ecole Centrale de Lyon, Ecully, France
| | - Léa Payen
- Laboratoire de Biochimie et Biologie Moléculaire, CICLY UR3738, Groupe Hospitalier Sud, Hospices Civils de Lyon, Pierre Bénite, France
| | - Damien Le Roy
- Institut Lumière Matière, ILM UMR5306, Université Claude Bernard Lyon 1, Villeurbanne, France
| | - Anne-Laure Deman
- Institut des Nanotechnologies de Lyon, INL UMR5270, Université Claude Bernard Lyon 1, Villeurbanne, France.
| |
Collapse
|
6
|
EpCAM- and EGFR-Specific Antibody Drug Conjugates for Triple-Negative Breast Cancer Treatment. Int J Mol Sci 2022; 23:ijms23116122. [PMID: 35682800 PMCID: PMC9181111 DOI: 10.3390/ijms23116122] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2022] [Revised: 05/18/2022] [Accepted: 05/27/2022] [Indexed: 12/29/2022] Open
Abstract
Triple-negative breast cancer (TNBC) is a group of heterogeneous and refractory breast cancers with the absence of estrogen receptor (ER), progesterone receptor (PgR) and epidermal growth factor receptor 2 (HER2). Over the past decade, antibody drug conjugates (ADCs) have ushered in a new era of targeting therapy. Since the epidermal growth factor receptor (EGFR) and epithelial cell adhesion molecule (EpCAM) are over expressed on triple-negative breast cancer, we developed novel ADCs by conjugating benzylguanine (BG)-modified monomethyl auristatin E (MMAE) to EpCAM- and EGFR-specific SNAP-tagged single chain antibody fragments (scFvs). Rapid and efficient conjugation was achieved by SNAP-tag technology. The binding and internalization properties of scFv-SNAP fusion proteins were confirmed by flow cytometry and fluorescence microscopy. The dose-dependent cytotoxicity was evaluated in cell lines expressing different levels of EGFR and EpCAM. Both ADCs showed specific cytotoxicity to EGFR or EpCAM positive cell lines via inducing apoptosis at a nanomolar concentration. Our study demonstrated that EGFR specific scFv-425-SNAP-BG-MMAE and EpCAM-specific scFv-EpCAM-SNAP-BG-MMAE could be promising ADCs for the treatment of TNBC.
Collapse
|
7
|
Circulating extracellular vesicles and tumor cells: sticky partners in metastasis. Trends Cancer 2022; 8:799-805. [DOI: 10.1016/j.trecan.2022.05.002] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Revised: 04/21/2022] [Accepted: 05/05/2022] [Indexed: 12/13/2022]
|
8
|
Descamps L, Le Roy D, Deman AL. Microfluidic-Based Technologies for CTC Isolation: A Review of 10 Years of Intense Efforts towards Liquid Biopsy. Int J Mol Sci 2022; 23:ijms23041981. [PMID: 35216097 PMCID: PMC8875744 DOI: 10.3390/ijms23041981] [Citation(s) in RCA: 53] [Impact Index Per Article: 17.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2022] [Revised: 02/04/2022] [Accepted: 02/07/2022] [Indexed: 02/01/2023] Open
Abstract
The selection of circulating tumor cells (CTCs) directly from blood as a real-time liquid biopsy has received increasing attention over the past ten years, and further analysis of these cells may greatly aid in both research and clinical applications. CTC analysis could advance understandings of metastatic cascade, tumor evolution, and patient heterogeneity, as well as drug resistance. Until now, the rarity and heterogeneity of CTCs have been technical challenges to their wider use in clinical studies, but microfluidic-based isolation technologies have emerged as promising tools to address these limitations. This review provides a detailed overview of latest and leading microfluidic devices implemented for CTC isolation. In particular, this study details must-have device performances and highlights the tradeoff between recovery and purity. Finally, the review gives a report of CTC potential clinical applications that can be conducted after CTC isolation. Widespread microfluidic devices, which aim to support liquid-biopsy-based applications, will represent a paradigm shift for cancer clinical care in the near future.
Collapse
Affiliation(s)
- Lucie Descamps
- Univ Lyon, Université Claude Bernard Lyon 1, CNRS, INSA Lyon, Ecole Centrale de Lyon, CPE Lyon, INL, UMR5270, 69622 Villeurbanne, France;
| | - Damien Le Roy
- Institut Lumière Matière ILM-UMR 5306, CNRS, Université Lyon 1, 69622 Villeurbanne, France;
| | - Anne-Laure Deman
- Univ Lyon, Université Claude Bernard Lyon 1, CNRS, INSA Lyon, Ecole Centrale de Lyon, CPE Lyon, INL, UMR5270, 69622 Villeurbanne, France;
- Correspondence:
| |
Collapse
|
9
|
Frankman ZD, Jiang L, Schroeder JA, Zohar Y. Application of Microfluidic Systems for Breast Cancer Research. MICROMACHINES 2022; 13:152. [PMID: 35208277 PMCID: PMC8877872 DOI: 10.3390/mi13020152] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/17/2021] [Revised: 01/11/2022] [Accepted: 01/17/2022] [Indexed: 02/06/2023]
Abstract
Cancer is a disease in which cells in the body grow out of control; breast cancer is the most common cancer in women in the United States. Due to early screening and advancements in therapeutic interventions, deaths from breast cancer have declined over time, although breast cancer remains the second leading cause of cancer death among women. Most deaths are due to metastasis, as cancer cells from the primary tumor in the breast form secondary tumors in remote sites in distant organs. Over many years, the basic biological mechanisms of breast cancer initiation and progression, as well as the subsequent metastatic cascade, have been studied using cell cultures and animal models. These models, although extremely useful for delineating cellular mechanisms, are poor predictors of physiological responses, primarily due to lack of proper microenvironments. In the last decade, microfluidics has emerged as a technology that could lead to a paradigm shift in breast cancer research. With the introduction of the organ-on-a-chip concept, microfluidic-based systems have been developed to reconstitute the dominant functions of several organs. These systems enable the construction of 3D cellular co-cultures mimicking in vivo tissue-level microenvironments, including that of breast cancer. Several reviews have been presented focusing on breast cancer formation, growth and metastasis, including invasion, intravasation, and extravasation. In this review, realizing that breast cancer can recur decades following post-treatment disease-free survival, we expand the discussion to account for microfluidic applications in the important areas of breast cancer detection, dormancy, and therapeutic development. It appears that, in the future, the role of microfluidics will only increase in the effort to eradicate breast cancer.
Collapse
Affiliation(s)
- Zachary D. Frankman
- Department of Biomedical Engineering, University of Arizona, Tucson, AZ 85721, USA;
| | - Linan Jiang
- Department of Aerospace and Mechanical Engineering, University of Arizona, Tucson, AZ 85721, USA;
| | - Joyce A. Schroeder
- Department of Molecular and Cellular Biology, University of Arizona, Tucson, AZ 85721, USA;
| | - Yitshak Zohar
- Department of Aerospace and Mechanical Engineering, University of Arizona, Tucson, AZ 85721, USA;
| |
Collapse
|
10
|
Topa J, Grešner P, Żaczek AJ, Markiewicz A. Breast cancer circulating tumor cells with mesenchymal features-an unreachable target? Cell Mol Life Sci 2022; 79:81. [PMID: 35048186 PMCID: PMC8770434 DOI: 10.1007/s00018-021-04064-6] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2021] [Revised: 11/26/2021] [Accepted: 11/27/2021] [Indexed: 12/13/2022]
Abstract
Circulating tumor cells (CTCs) mediate dissemination of solid tumors and can be an early sign of disease progression. Moreover, they show a great potential in terms of non-invasive, longitudinal monitoring of cancer patients. CTCs have been extensively studied in breast cancer (BC) and were shown to present a significant phenotypic plasticity connected with initiation of epithelial-mesenchymal transition (EMT). Apart from conferring malignant properties, EMT affects CTCs recovery rate, making a significant portion of CTCs from patients’ samples undetected. Wider application of methods and markers designed to isolate and identify mesenchymal CTCs is required to expand our knowledge about the clinical impact of mesenchymal CTCs. Therefore, here we provide a comprehensive review of clinical significance of mesenchymal CTCs in BC together with statistical analysis of previously published data, in which we assessed the suitability of a number of methods/markers used for isolation of CTCs with different EMT phenotypes, both in in vitro spike-in tests with BC cell lines, as well as clinical samples. Results of spiked-in cell lines indicate that, in general, methods not based on epithelial enrichment only, capture mesenchymal CTCs much more efficiently that CellSearch® (golden standard in CTCs detection), but at the same time are not much inferior to Cell Search®, though large variation in recovery rates of added cells among the methods is observed. In clinical samples, where additional CTCs detection markers are needed, positive epithelial-based CTCs enrichment was the most efficient in isolating CTCs with mesenchymal features from non-metastatic BC patients. From the marker side, PI3K and VIM were contributing the most to detection of CTCs with mesenchymal features (in comparison to SNAIL) in non-metastatic and metastatic BC patients, respectively. However, additional data are needed for more robust identification of markers for efficient detection of CTCs with mesenchymal features.
Collapse
Affiliation(s)
- Justyna Topa
- Laboratory of Translational Oncology, Intercollegiate Faculty of Biotechnology, University of Gdańsk and Medical University of Gdańsk, Debinki 1, 80-211, Gdansk, Poland
| | - Peter Grešner
- Laboratory of Translational Oncology, Intercollegiate Faculty of Biotechnology, University of Gdańsk and Medical University of Gdańsk, Debinki 1, 80-211, Gdansk, Poland
| | - Anna J Żaczek
- Laboratory of Translational Oncology, Intercollegiate Faculty of Biotechnology, University of Gdańsk and Medical University of Gdańsk, Debinki 1, 80-211, Gdansk, Poland
| | - Aleksandra Markiewicz
- Laboratory of Translational Oncology, Intercollegiate Faculty of Biotechnology, University of Gdańsk and Medical University of Gdańsk, Debinki 1, 80-211, Gdansk, Poland.
| |
Collapse
|
11
|
Sharma A, Joshkon A, Ladjimi A, Traboulsi W, Bachelier R, Robert S, Foucault-Bertaud A, Leroyer AS, Bardin N, Somasundaram I, Blot-Chabaud M. Soluble CD146 as a Potential Target for Preventing Triple Negative Breast Cancer MDA-MB-231 Cell Growth and Dissemination. Int J Mol Sci 2022; 23:ijms23020974. [PMID: 35055160 PMCID: PMC8780963 DOI: 10.3390/ijms23020974] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2021] [Revised: 01/07/2022] [Accepted: 01/08/2022] [Indexed: 11/16/2022] Open
Abstract
Background: Triple Negative Breast Cancers (TNBC) are the most aggressive breast cancers and lead to poor prognoses. This is due to a high resistance to therapies, mainly because of the presence of Cancer Stem Cells (CSCs). Plasticity, a feature of CSCs, is acquired through the Epithelial to Mesenchymal Transition (EMT), a process that has been recently shown to be regulated by a key molecule, CD146. Of interest, CD146 is over-expressed in TNBC. Methods: The MDA-MB-231 TNBC cell line was used as a model to study the role of CD146 and its secreted soluble form (sCD146) in the development and dissemination of TNBC using in vitro and in vivo studies. Results: High expression of CD146 in a majority of MDA-MB-231 cells leads to an increased secretion of sCD146 that up-regulates the expression of EMT and CSC markers on the cells. These effects can be blocked with a specific anti-sCD146 antibody, M2J-1 mAb. M2J-1 mAb was able to reduce tumour development and dissemination in a model of cells xenografted in nude mice and an experimental model of metastasis, respectively, in part through its effects on CSC. Conclusion: We propose that M2J-1 mAb could be used as an additional therapeutic approach to fight TNBC.
Collapse
Affiliation(s)
- Akshita Sharma
- Department of Stem Cell and Regenerative Medicine, D.Y. Patil Universit, Kolhapur 416003, India; (A.S.); (I.S.)
| | - Ahmad Joshkon
- Faculty of Pharmacy, Aix-Marseille University, INSERM 1263, INRAE 1260, C2VN, 13005 Marseille, France; (A.J.); (A.L.); (W.T.); (R.B.); (S.R.); (A.F.-B.); (A.S.L.); (N.B.)
| | - Aymen Ladjimi
- Faculty of Pharmacy, Aix-Marseille University, INSERM 1263, INRAE 1260, C2VN, 13005 Marseille, France; (A.J.); (A.L.); (W.T.); (R.B.); (S.R.); (A.F.-B.); (A.S.L.); (N.B.)
| | - Waël Traboulsi
- Faculty of Pharmacy, Aix-Marseille University, INSERM 1263, INRAE 1260, C2VN, 13005 Marseille, France; (A.J.); (A.L.); (W.T.); (R.B.); (S.R.); (A.F.-B.); (A.S.L.); (N.B.)
| | - Richard Bachelier
- Faculty of Pharmacy, Aix-Marseille University, INSERM 1263, INRAE 1260, C2VN, 13005 Marseille, France; (A.J.); (A.L.); (W.T.); (R.B.); (S.R.); (A.F.-B.); (A.S.L.); (N.B.)
| | - Stéphane Robert
- Faculty of Pharmacy, Aix-Marseille University, INSERM 1263, INRAE 1260, C2VN, 13005 Marseille, France; (A.J.); (A.L.); (W.T.); (R.B.); (S.R.); (A.F.-B.); (A.S.L.); (N.B.)
| | - Alexandrine Foucault-Bertaud
- Faculty of Pharmacy, Aix-Marseille University, INSERM 1263, INRAE 1260, C2VN, 13005 Marseille, France; (A.J.); (A.L.); (W.T.); (R.B.); (S.R.); (A.F.-B.); (A.S.L.); (N.B.)
| | - Aurélie S. Leroyer
- Faculty of Pharmacy, Aix-Marseille University, INSERM 1263, INRAE 1260, C2VN, 13005 Marseille, France; (A.J.); (A.L.); (W.T.); (R.B.); (S.R.); (A.F.-B.); (A.S.L.); (N.B.)
| | - Nathalie Bardin
- Faculty of Pharmacy, Aix-Marseille University, INSERM 1263, INRAE 1260, C2VN, 13005 Marseille, France; (A.J.); (A.L.); (W.T.); (R.B.); (S.R.); (A.F.-B.); (A.S.L.); (N.B.)
| | - Indumathi Somasundaram
- Department of Stem Cell and Regenerative Medicine, D.Y. Patil Universit, Kolhapur 416003, India; (A.S.); (I.S.)
| | - Marcel Blot-Chabaud
- Faculty of Pharmacy, Aix-Marseille University, INSERM 1263, INRAE 1260, C2VN, 13005 Marseille, France; (A.J.); (A.L.); (W.T.); (R.B.); (S.R.); (A.F.-B.); (A.S.L.); (N.B.)
- Correspondence: ; Tel.: +33-4-91-83-56-85
| |
Collapse
|
12
|
Liquid Biopsies: Flowing Biomarkers. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2022; 1379:341-368. [DOI: 10.1007/978-3-031-04039-9_14] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
13
|
Yokota K, Hashimoto M, Kajimoto K, Tanaka M, Murayama S, Tsutsui M, Nakajima Y, Taniguchi M, Kataoka M. Effect of Electrolyte Concentration on Cell Sensing by Measuring Ionic Current Waveform through Micropores. BIOSENSORS-BASEL 2021; 11:bios11030078. [PMID: 33809382 PMCID: PMC7998150 DOI: 10.3390/bios11030078] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/18/2021] [Revised: 03/10/2021] [Accepted: 03/11/2021] [Indexed: 12/25/2022]
Abstract
Immunostaining has been widely used in cancer prognosis for the quantitative detection of cancer cells present in the bloodstream. However, conventional detection methods based on the target membrane protein expression exhibit the risk of missing cancer cells owing to variable protein expressions. In this study, the resistive pulse method (RPM) was employed to discriminate between cultured cancer cells (NCI-H1650) and T lymphoblastoid leukemia cells (CCRF-CEM) by measuring the ionic current response of cells flowing through a micro-space. The height and shape of a pulse signal were used for the simultaneous measurement of size, deformability, and surface charge of individual cells. An accurate discrimination of cancer cells could not be obtained using 1.0 × phosphate-buffered saline (PBS) as an electrolyte solution to compare the size measurements by a microscopic observation. However, an accurate discrimination of cancer cells with a discrimination error rate of 4.5 ± 0.5% was achieved using 0.5 × PBS containing 2.77% glucose as the electrolyte solution. The potential application of RPM for the accurate discrimination of cancer cells from leukocytes was demonstrated through the measurement of the individual cell size, deformability, and surface charge in a solution with a low electrolyte concentration.
Collapse
Affiliation(s)
- Kazumichi Yokota
- National Institute of Advanced Industrial Science and Technology, Takamatsu, Kagawa 761-0395, Japan; (K.Y.); (M.H.); (K.K.); (M.T.); (Y.N.)
| | - Muneaki Hashimoto
- National Institute of Advanced Industrial Science and Technology, Takamatsu, Kagawa 761-0395, Japan; (K.Y.); (M.H.); (K.K.); (M.T.); (Y.N.)
| | - Kazuaki Kajimoto
- National Institute of Advanced Industrial Science and Technology, Takamatsu, Kagawa 761-0395, Japan; (K.Y.); (M.H.); (K.K.); (M.T.); (Y.N.)
| | - Masato Tanaka
- National Institute of Advanced Industrial Science and Technology, Takamatsu, Kagawa 761-0395, Japan; (K.Y.); (M.H.); (K.K.); (M.T.); (Y.N.)
| | - Sanae Murayama
- The Institute of Scientific and Industrial Research, Osaka University, 8-1 Mihogaoka, Ibaraki, Osaka 567-0047, Japan; (S.M.); (M.T.); (M.T.)
| | - Makusu Tsutsui
- The Institute of Scientific and Industrial Research, Osaka University, 8-1 Mihogaoka, Ibaraki, Osaka 567-0047, Japan; (S.M.); (M.T.); (M.T.)
| | - Yoshihiro Nakajima
- National Institute of Advanced Industrial Science and Technology, Takamatsu, Kagawa 761-0395, Japan; (K.Y.); (M.H.); (K.K.); (M.T.); (Y.N.)
| | - Masateru Taniguchi
- The Institute of Scientific and Industrial Research, Osaka University, 8-1 Mihogaoka, Ibaraki, Osaka 567-0047, Japan; (S.M.); (M.T.); (M.T.)
| | - Masatoshi Kataoka
- National Institute of Advanced Industrial Science and Technology, Takamatsu, Kagawa 761-0395, Japan; (K.Y.); (M.H.); (K.K.); (M.T.); (Y.N.)
- Correspondence: ; Tel.: +81-87-869-3576
| |
Collapse
|
14
|
Tian C, Xu X, Wang Y, Li D, Lu H, Yang Z. Development and Clinical Prospects of Techniques to Separate Circulating Tumor Cells from Peripheral Blood. Cancer Manag Res 2020; 12:7263-7275. [PMID: 32884342 PMCID: PMC7434565 DOI: 10.2147/cmar.s248380] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2020] [Accepted: 07/15/2020] [Indexed: 12/15/2022] Open
Abstract
Detection of circulating tumor cells (CTC) is an important liquid biopsy technique that has advanced considerably in recent years. To further advance the development of technology for curing cancer, several CTC technologies have been proposed by various research groups. Despite their potential role in early cancer diagnosis and prognosis, CTC methods are currently used for research purposes only, and very few methods have been accepted for clinical applications because of difficulties, including CTC heterogeneity, CTC separation from the blood, and a lack of thorough clinical validation. Although current CTC technologies have not been truly implemented, they possess high potential as future clinical diagnostic techniques for individualized cancer. Here, we review current developments in CTC separation technology. We also explore new CTC detection methods based on telomerase and nanomaterials, such as in vivo flow cytometry. In addition, we discuss the difficulties that must be overcome before CTC can be applied in clinical settings.
Collapse
Affiliation(s)
- Cheng Tian
- Yichang Central People's Hospital, First Clinical Medical College of Three Gorges University, Yichang 443000, People's Republic of China
| | - Xinhua Xu
- Yichang Central People's Hospital, First Clinical Medical College of Three Gorges University, Yichang 443000, People's Republic of China
| | - Yuke Wang
- Yichang Central People's Hospital, First Clinical Medical College of Three Gorges University, Yichang 443000, People's Republic of China
| | - Dailong Li
- Yichang Central People's Hospital, First Clinical Medical College of Three Gorges University, Yichang 443000, People's Republic of China
| | - Haiyan Lu
- Yichang Central People's Hospital, First Clinical Medical College of Three Gorges University, Yichang 443000, People's Republic of China
| | - Ziwei Yang
- Yichang Central People's Hospital, First Clinical Medical College of Three Gorges University, Yichang 443000, People's Republic of China
| |
Collapse
|
15
|
Harigopal M, Kowalski D, Vosoughi A. Enumeration and molecular characterization of circulating tumor cells as an innovative tool for companion diagnostics in breast cancer. Expert Rev Mol Diagn 2020; 20:815-828. [PMID: 32546017 DOI: 10.1080/14737159.2020.1784009] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
INTRODUCTION Circulating tumor cells (CTC) and more recently, CTC clusters are implicated as a fundamental mechanism by which tumor cells break away from the primary site and travel to distant sites. Enumeration of CTC and CTC clusters represents a new approach to prognosis, prediction, and response to therapy in patients with early and metastatic breast cancer. Several recent studies have shown the predictive importance of monitoring CTCs levels in progression-free and overall survival in breast cancer patients. This review will focus on CTC enumeration and characterization in breast cancers. AREAS COVERED We will provide a historical perspective and clinical background of CTC detection in peripheral blood. The current methodologies for studying CTCs and newer technologies for CTC detection will be reviewed together with the current state of the art of CTCs as a biomarker in risk stratification and prognostication in breast cancers. EXPERT OPINION Currently, there is an FDA approved CTC assessment method for clinical use. While CTC enumeration, is a marker for prognostication and survival, molecular characterization of CTC, may be more accurate in monitoring response to treatment due to tumor heterogeneity rather than the tumor phenotype at the primary or metastatic sites.
Collapse
Affiliation(s)
- Malini Harigopal
- Department of Pathology, Yale School of Medicine New Haven, Connecticut ., 06520-8023, New Haven, CT, United States
| | - Diane Kowalski
- Department of Pathology, Yale School of Medicine New Haven, Connecticut ., 06520-8023, New Haven, CT, United States
| | - Aram Vosoughi
- Department of Pathology, Yale School of Medicine New Haven, Connecticut ., 06520-8023, New Haven, CT, United States
| |
Collapse
|
16
|
Angus L, Martens JWM, van den Bent MJ, Sillevis Smitt PAE, Sleijfer S, Jager A. Novel methods to diagnose leptomeningeal metastases in breast cancer. Neuro Oncol 2020; 21:428-439. [PMID: 30418595 DOI: 10.1093/neuonc/noy186] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Abstract
Leptomeningeal metastases (LM) in breast cancer patients are rare but often accompanied by devastating neurological symptoms and carry a very poor prognosis, even if treated. To date, two diagnostic methods are clinically used to diagnose LM: gadolinium MRI of the brain and/or spinal cord and cytological examination of cerebrospinal fluid (CSF). Both techniques are, however, hampered by limited sensitivities, often leading to a long diagnostic process requiring repeated lumbar punctures and MRI examinations. To improve the detection rate of LM, numerous studies have assessed new techniques. In this review, we present the current workup to diagnose LM, set out an overview of novel techniques to diagnose LM, and give recommendations for future research.
Collapse
Affiliation(s)
- Lindsay Angus
- Erasmus MC Cancer Institute, Erasmus University Medical Center, Department of Medical Oncology, Rotterdam, the Netherlands
| | - John W M Martens
- Erasmus MC Cancer Institute, Erasmus University Medical Center, Department of Medical Oncology, Rotterdam, the Netherlands
| | - Martin J van den Bent
- The Brain Tumor Center at Erasmus MC Cancer Institute, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - Peter A E Sillevis Smitt
- The Brain Tumor Center at Erasmus MC Cancer Institute, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - Stefan Sleijfer
- Erasmus MC Cancer Institute, Erasmus University Medical Center, Department of Medical Oncology, Rotterdam, the Netherlands
| | - Agnes Jager
- Erasmus MC Cancer Institute, Erasmus University Medical Center, Department of Medical Oncology, Rotterdam, the Netherlands
| |
Collapse
|
17
|
de Kruijff IE, Beije N, Martens JWM, de Wit R, Boormans JL, Sleijfer S. Liquid Biopsies to Select Patients for Perioperative Chemotherapy in Muscle-invasive Bladder Cancer: A Systematic Review. Eur Urol Oncol 2020; 4:204-214. [PMID: 32059957 DOI: 10.1016/j.euo.2020.01.003] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2019] [Revised: 01/12/2020] [Accepted: 01/23/2020] [Indexed: 02/08/2023]
Abstract
CONTEXT Neoadjuvant chemotherapy (NAC) is considered the standard treatment for muscle-invasive bladder cancer (MIBC). However, its overall survival benefit is limited and toxicity is significant; hence, NAC has not been adopted universally. OBJECTIVE To systematically evaluate whether biomarkers can guide the administration of perioperative chemotherapy in MIBC patients. EVIDENCE ACQUISITION A systematic search of the PubMed database was performed according to the Preferred Reporting Items for Systematic Reviews and Meta-analyses (PRISMA). In total, 215 papers were screened and 22 were selected to assess the potential clinical value of circulating tumor cells (CTCs) and cell-free DNA (cfDNA) in selecting MIBC patients for perioperative chemotherapy. EVIDENCE SYNTHESIS We found that the presence of one or more CTCs before radical cystectomy, as determined by the CellSearch technique, is a robust marker for poor recurrence-free and overall survival. Consequently, whether NAC can be withheld in patients without the presence of CTCs is a subject of ongoing investigation. Studies investigating various approaches to detect cfDNA showed that cfDNA is present in the blood of MIBC patients, but varying results on its prognostic value have been reported. Successful cfDNA-based approaches are likely to encompass at least a multitude of genes using next-generation sequencing, as there are generally few hotspot somatic mutations in MIBC. CONCLUSIONS Liquid biopsies hold promise in selecting MIBC patients for perioperative chemotherapy, but instead of more proof-of-principle studies, prospective studies investigating true clinical applicability for treatment decision making are urgently needed. PATIENT SUMMARY Liquid biopsies appear to be a promising tool to guide the administration of chemotherapy in patients with muscle-invasive bladder cancer; however, the optimal way to implement these remains to be determined.
Collapse
Affiliation(s)
- Ingeborg E de Kruijff
- Department of Medical Oncology and Cancer Genomics Netherlands, Erasmus MC Cancer Institute, Erasmus University Medical Center, Rotterdam, The Netherlands.
| | - Nick Beije
- Department of Medical Oncology and Cancer Genomics Netherlands, Erasmus MC Cancer Institute, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - John W M Martens
- Department of Medical Oncology and Cancer Genomics Netherlands, Erasmus MC Cancer Institute, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Ronald de Wit
- Department of Medical Oncology and Cancer Genomics Netherlands, Erasmus MC Cancer Institute, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Joost L Boormans
- Department of Urology, Erasmus MC Cancer Institute, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Stefan Sleijfer
- Department of Medical Oncology and Cancer Genomics Netherlands, Erasmus MC Cancer Institute, Erasmus University Medical Center, Rotterdam, The Netherlands
| |
Collapse
|
18
|
Agnoletto C, Corrà F, Minotti L, Baldassari F, Crudele F, Cook WJJ, Di Leva G, d'Adamo AP, Gasparini P, Volinia S. Heterogeneity in Circulating Tumor Cells: The Relevance of the Stem-Cell Subset. Cancers (Basel) 2019; 11:cancers11040483. [PMID: 30959764 PMCID: PMC6521045 DOI: 10.3390/cancers11040483] [Citation(s) in RCA: 100] [Impact Index Per Article: 16.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2019] [Revised: 03/16/2019] [Accepted: 03/30/2019] [Indexed: 12/20/2022] Open
Abstract
The release of circulating tumor cells (CTCs) into vasculature is an early event in the metastatic process. The analysis of CTCs in patients has recently received widespread attention because of its clinical implications, particularly for precision medicine. Accumulated evidence documents a large heterogeneity in CTCs across patients. Currently, the most accepted view is that tumor cells with an intermediate phenotype between epithelial and mesenchymal have the highest plasticity. Indeed, the existence of a meta-stable or partial epithelial–mesenchymal transition (EMT) cell state, with both epithelial and mesenchymal features, can be easily reconciled with the concept of a highly plastic stem-like state. A close connection between EMT and cancer stem cells (CSC) traits, with enhanced metastatic competence and drug resistance, has also been described. Accordingly, a subset of CTCs consisting of CSC, present a stemness profile, are able to survive chemotherapy, and generate metastases after xenotransplantation in immunodeficient mice. In the present review, we discuss the current evidence connecting CTCs, EMT, and stemness. An improved understanding of the CTC/EMT/CSC connections may uncover novel therapeutic targets, irrespective of the tumor type, since most cancers seem to harbor a pool of CSCs, and disclose important mechanisms underlying tumorigenicity.
Collapse
Affiliation(s)
- Chiara Agnoletto
- Department of Morphology, Surgery and Experimental Medicine, University of Ferrara, 44121 Ferrara, Italy.
| | - Fabio Corrà
- Department of Morphology, Surgery and Experimental Medicine, University of Ferrara, 44121 Ferrara, Italy.
| | - Linda Minotti
- Department of Morphology, Surgery and Experimental Medicine, University of Ferrara, 44121 Ferrara, Italy.
| | - Federica Baldassari
- Department of Morphology, Surgery and Experimental Medicine, University of Ferrara, 44121 Ferrara, Italy.
| | - Francesca Crudele
- Department of Morphology, Surgery and Experimental Medicine, University of Ferrara, 44121 Ferrara, Italy.
| | | | - Gianpiero Di Leva
- School of Environment and Life Sciences, University of Salford, Salford M5 4WT, UK.
| | - Adamo Pio d'Adamo
- Department of Medicine, Surgery and Health Sciences, University of Trieste, 34127 Trieste, Italy.
- Institute for Maternal and Child Health-IRCCS "Burlo Garofolo", 34137 Trieste, Italy.
| | - Paolo Gasparini
- Department of Medicine, Surgery and Health Sciences, University of Trieste, 34127 Trieste, Italy.
- Institute for Maternal and Child Health-IRCCS "Burlo Garofolo", 34137 Trieste, Italy.
| | - Stefano Volinia
- Department of Morphology, Surgery and Experimental Medicine, University of Ferrara, 44121 Ferrara, Italy.
| |
Collapse
|
19
|
Khetani S, Mohammadi M, Nezhad AS. Filter-based isolation, enrichment, and characterization of circulating tumor cells. Biotechnol Bioeng 2018; 115:2504-2529. [DOI: 10.1002/bit.26787] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2018] [Revised: 06/25/2018] [Accepted: 06/28/2018] [Indexed: 01/12/2023]
Affiliation(s)
- Sultan Khetani
- Department of Mechanical and Manufacturing Engineering, BioMEMS and Bioinspired Microfluidic Laboratory; University of Calgary; Calgary Canada
- Center for BioEngineering Research and Education, University of Calgary; Calgary Canada
| | - Mehdi Mohammadi
- Department of Mechanical and Manufacturing Engineering, BioMEMS and Bioinspired Microfluidic Laboratory; University of Calgary; Calgary Canada
- Center for BioEngineering Research and Education, University of Calgary; Calgary Canada
- Department of Biological Sciences; University of Calgary; Calgary Canada
| | - Amir Sanati Nezhad
- Department of Mechanical and Manufacturing Engineering, BioMEMS and Bioinspired Microfluidic Laboratory; University of Calgary; Calgary Canada
- Center for BioEngineering Research and Education, University of Calgary; Calgary Canada
| |
Collapse
|
20
|
Abstract
Metastasis contributes to poor prognosis in many types of cancer and is the leading cause of cancer-related deaths. Tumor cells metastasize to distant sites via the circulatory and lymphatic systems. In this review, we discuss the potential of circulating tumor cells for diagnosis and describe the experimental therapeutics that aim to target these disseminating cancer cells. We discuss the advantages and limitations of such strategies and how they may lead to the development of the next generation of antimetastasis treatments.
Collapse
Affiliation(s)
- Eric Lin
- Department of Chemical Engineering, University of Michigan, Ann Arbor, Michigan 48109, USA
- University of Michigan Comprehensive Cancer Center, Ann Arbor, Michigan 48109, USA
- Biointerfaces Institute, University of Michigan, Ann Arbor, Michigan 48109, USA
| | - Thong Cao
- Department of Biomedical Engineering, Vanderbilt University, Nashville, Tennessee 37235, USA
| | - Sunitha Nagrath
- Department of Chemical Engineering, University of Michigan, Ann Arbor, Michigan 48109, USA
- University of Michigan Comprehensive Cancer Center, Ann Arbor, Michigan 48109, USA
- Biointerfaces Institute, University of Michigan, Ann Arbor, Michigan 48109, USA
| | - Michael R. King
- Department of Biomedical Engineering, Vanderbilt University, Nashville, Tennessee 37235, USA
| |
Collapse
|
21
|
de Kruijff IE, Timmermans AM, den Bakker MA, Trapman-Jansen AMAC, Foekens R, Meijer-Van Gelder ME, Oomen-de Hoop E, Smid M, Hollestelle A, van Deurzen CHM, Foekens JA, Martens JWM, Sleijfer S. The Prevalence of CD146 Expression in Breast Cancer Subtypes and Its Relation to Outcome. Cancers (Basel) 2018; 10:cancers10050134. [PMID: 29734758 PMCID: PMC5977107 DOI: 10.3390/cancers10050134] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2018] [Revised: 04/30/2018] [Accepted: 05/03/2018] [Indexed: 01/18/2023] Open
Abstract
CD146, involved in epithelial-to-mesenchymal transition (EMT), might affect cancer aggressiveness. We here investigated the prevalence of CD146 expression in breast cancer subtypes, its relation to prognosis, the relation between CD146 and EMT and the outcome to tamoxifen. Primary breast cancer tissues from 1342 patients were available for this retrospective study and immunohistochemically stained for CD146. For survival analyses, pure prognosis was studied by only including lymph-node negative patients who did not receive (neo)adjuvant systemic treatment (n = 551). 11% of the tumors showed CD146 expression. CD146 expression was most prevalent in triple-negative cases (64%, p < 0.001). In univariable analysis, CD146 expression was a prognostic factor for both metastasis-free survival (MFS) (p = 0.020) and overall survival (OS) (p = 0.037), but not in multivariable analysis (including age, tumor size, grade, estrogen receptor (ER), progesterone receptor (PR), human epidermal growth factor receptor 2 (HER2) and Ki-67). No correlation between CD146 and EMT nor difference in outcome to first-line tamoxifen was seen. In this large series, our data showed that CD146 is present in primary breast cancer and is a pure prognostic factor for MFS and OS in breast cancer patients. We did not see an association between CD146 expression and EMT nor on outcome to tamoxifen.
Collapse
Affiliation(s)
- Ingeborg E de Kruijff
- Department of Medical Oncology, Erasmus MC Cancer Institute, 's-Gravendijkwal 230, 3015 CE Rotterdam, The Netherlands.
| | - Anna M Timmermans
- Department of Medical Oncology, Erasmus MC Cancer Institute, 's-Gravendijkwal 230, 3015 CE Rotterdam, The Netherlands.
| | - Michael A den Bakker
- Department of Pathology, Erasmus MC Cancer Institute, 's-Gravendijkwal 230, 3015 CE Rotterdam, The Netherlands.
| | - Anita M A C Trapman-Jansen
- Department of Medical Oncology, Erasmus MC Cancer Institute, 's-Gravendijkwal 230, 3015 CE Rotterdam, The Netherlands.
| | - Renée Foekens
- Department of Medical Oncology, Erasmus MC Cancer Institute, 's-Gravendijkwal 230, 3015 CE Rotterdam, The Netherlands.
| | - Marion E Meijer-Van Gelder
- Department of Medical Oncology, Erasmus MC Cancer Institute, 's-Gravendijkwal 230, 3015 CE Rotterdam, The Netherlands.
| | - Esther Oomen-de Hoop
- Department of Medical Oncology, Erasmus MC Cancer Institute, 's-Gravendijkwal 230, 3015 CE Rotterdam, The Netherlands.
| | - Marcel Smid
- Department of Medical Oncology, Erasmus MC Cancer Institute, 's-Gravendijkwal 230, 3015 CE Rotterdam, The Netherlands.
| | - Antoinette Hollestelle
- Department of Medical Oncology, Erasmus MC Cancer Institute, 's-Gravendijkwal 230, 3015 CE Rotterdam, The Netherlands.
| | - Carolien H M van Deurzen
- Department of Pathology, Erasmus MC Cancer Institute, 's-Gravendijkwal 230, 3015 CE Rotterdam, The Netherlands.
| | - John A Foekens
- Department of Medical Oncology, Erasmus MC Cancer Institute, 's-Gravendijkwal 230, 3015 CE Rotterdam, The Netherlands.
| | - John W M Martens
- Department of Medical Oncology, Erasmus MC Cancer Institute, 's-Gravendijkwal 230, 3015 CE Rotterdam, The Netherlands.
| | - Stefan Sleijfer
- Department of Medical Oncology, Erasmus MC Cancer Institute, 's-Gravendijkwal 230, 3015 CE Rotterdam, The Netherlands.
| |
Collapse
|
22
|
Huang Q, Wang FB, Yuan CH, He Z, Rao L, Cai B, Chen B, Jiang S, Li Z, Chen J, Liu W, Guo F, Ao Z, Chen S, Zhao XZ. Gelatin Nanoparticle-Coated Silicon Beads for Density-Selective Capture and Release of Heterogeneous Circulating Tumor Cells with High Purity. Am J Cancer Res 2018; 8:1624-1635. [PMID: 29556345 PMCID: PMC5858171 DOI: 10.7150/thno.23531] [Citation(s) in RCA: 52] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2017] [Accepted: 12/16/2017] [Indexed: 01/09/2023] Open
Abstract
Background: Circulating tumor cells (CTCs) are a burgeoning topic in cancer biomarker discovery research with minimal invasive blood draws. CTCs can be used as potential biomarkers for disease prognosis, early cancer diagnosis and pharmacodynamics. However, the extremely low abundance of CTCs limits their clinical utility because of technical challenges such as the isolation and subsequent detailed molecular and functional characterization of rare CTCs from patient blood samples. Methods: In this study, we present a novel density gradient centrifugation method employing biodegradable gelatin nanoparticles coated on silicon beads for the isolation, release, and downstream analysis of CTCs from colorectal and breast cancer patients. Results: Using clinical patient/spiked samples, we demonstrate that this method has significant CTC-capture efficiency (>80%) and purity (>85%), high CTC release efficiency (94%) and viability (92.5%). We also demonstrate the unparalleled robustness of our method in downstream CTC analyses such as the detection of PIK3CA mutations. Conclusion: The efficiency and versatility of the multifunctional density microbeads approach provides new opportunities for personalized cancer diagnostics and treatments.
Collapse
|
23
|
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
|
24
|
Praharaj PP, Bhutia SK, Nagrath S, Bitting RL, Deep G. Circulating tumor cell-derived organoids: Current challenges and promises in medical research and precision medicine. Biochim Biophys Acta Rev Cancer 2018; 1869:117-127. [PMID: 29360544 PMCID: PMC6054479 DOI: 10.1016/j.bbcan.2017.12.005] [Citation(s) in RCA: 94] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2017] [Revised: 12/22/2017] [Accepted: 12/22/2017] [Indexed: 12/11/2022]
Abstract
Traditional 2D cell cultures do not accurately recapitulate tumor heterogeneity, and insufficient human cell lines are available. Patient-derived xenograft (PDX) models more closely mimic clinical tumor heterogeneity, but are not useful for high-throughput drug screening. Recently, patient-derived organoid cultures have emerged as a novel technique to fill this critical need. Organoids maintain tumor tissue heterogeneity and drug-resistance responses, and thus are useful for high-throughput drug screening. Among various biological tissues used to produce organoid cultures, circulating tumor cells (CTCs) are promising, due to relative ease of ascertainment. CTC-derived organoids could help to acquire relevant genetic and epigenetic information about tumors in real time, and screen and test promising drugs. This could reduce the need for tissue biopsies, which are painful and may be difficult depending on the tumor location. In this review, we have focused on advances in CTC isolation and organoid culture methods, and their potential applications in disease modeling and precision medicine.
Collapse
Affiliation(s)
- Prakash P Praharaj
- Wake Forest Baptist Medical Center, Department of Cancer Biology, Winston-Salem, NC, United States; Department of Life Science, National Institute of Technology Rourkela, Odisha, India
| | - Sujit K Bhutia
- Department of Life Science, National Institute of Technology Rourkela, Odisha, India
| | - Sunitha Nagrath
- Department of Chemical Engineering, University of Michigan, Ann Arbor, MI, United States; Biointerfaces Institute, University of Michigan, Ann Arbor, MI, United States
| | - Rhonda L Bitting
- Hematology and Oncology Department, United States; Wake Forest Baptist Comprehensive Cancer Center, United States
| | - Gagan Deep
- Wake Forest Baptist Medical Center, Department of Cancer Biology, Winston-Salem, NC, United States; Wake Forest Baptist Comprehensive Cancer Center, United States; Department of Urology, Wake Forest Baptist Medical Center, Winston-Salem, NC, United States.
| |
Collapse
|
25
|
Ouhtit A, Abdraboh ME, Hollenbach AD, Zayed H, Raj MHG. CD146, a novel target of CD44-signaling, suppresses breast tumor cell invasion. Cell Commun Signal 2017; 15:45. [PMID: 29121955 PMCID: PMC5679321 DOI: 10.1186/s12964-017-0200-3] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2017] [Accepted: 10/25/2017] [Indexed: 01/27/2023] Open
Abstract
Background We have previously validated three novel CD44-downstream positively regulated transcriptional targets, including Cortactin, Survivin and TGF-β2, and further characterized the players underlying their separate signaling pathways. In the present study, we identified CD146 as a potential novel target, negatively regulated by CD44. While the exact function of CD146 in breast cancer (BC) is not completely understood, substantial evidence from our work and others support the hypothesis that CD146 is a suppressor of breast tumor progression. Methods Therefore, using molecular and pharmacological approaches both in vitro and in breast tissues of human samples, the present study validated CD146 as a novel target of CD44-signaling suppressed during BC progression. Results Our results revealed that CD44 activation could cause a substantial decrease of CD146 expression with an equally notable converse effect upon CD44-siRNA inhibition. More interestingly, activation of CD44 decreased cellular CD146 and increased soluble CD146 through CD44-dependent activation of MMP. Conclusion Here, we provide a possible mechanism by which CD146 suppresses BC progression as a target of CD44-downstream signaling, regulating neovascularization and cancer cell motility.
Collapse
Affiliation(s)
- Allal Ouhtit
- Department of Biological and Environmental Sciences, College of Arts and Sciences, Qatar University, Doha, Qatar.
| | - Mohammed E Abdraboh
- Department of Zoology, Faculty of Science, Mansoura University, Mansoura, Egypt
| | - Andrew D Hollenbach
- Department of Genetics, Louisiana State University, Health Sciences Center, New Orleans, USA
| | - Hatem Zayed
- Department of Biomedical Sciences, College of Health and Sciences, Qatar University, Doha, Qatar
| | - Madhwa H G Raj
- Department of Obstetrics and Gynecology, Louisiana State University, Health Sciences Center, New Orleans, USA
| |
Collapse
|
26
|
Witek MA, Aufforth RD, Wang H, Kamande JW, Jackson JM, Pullagurla SR, Hupert ML, Usary J, Wysham WZ, Hilliard D, Montgomery S, Bae-Jump V, Carey LA, Gehrig PA, Milowsky MI, Perou CM, Soper JT, Whang YE, Yeh JJ, Martin G, Soper SA. Discrete microfluidics for the isolation of circulating tumor cell subpopulations targeting fibroblast activation protein alpha and epithelial cell adhesion molecule. NPJ Precis Oncol 2017; 1. [PMID: 29657983 PMCID: PMC5871807 DOI: 10.1038/s41698-017-0028-8] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Circulating tumor cells consist of phenotypically distinct subpopulations that originate from the tumor microenvironment. We report a circulating tumor cell dual selection assay that uses discrete microfluidics to select circulating tumor cell subpopulations from a single blood sample; circulating tumor cells expressing the established marker epithelial cell adhesion molecule and a new marker, fibroblast activation protein alpha, were evaluated. Both circulating tumor cell subpopulations were detected in metastatic ovarian, colorectal, prostate, breast, and pancreatic cancer patients and 90% of the isolated circulating tumor cells did not co-express both antigens. Clinical sensitivities of 100% showed substantial improvement compared to epithelial cell adhesion molecule selection alone. Owing to high purity (>80%) of the selected circulating tumor cells, molecular analysis of both circulating tumor cell subpopulations was carried out in bulk, including next generation sequencing, mutation analysis, and gene expression. Results suggested fibroblast activation protein alpha and epithelial cell adhesion molecule circulating tumor cells are distinct subpopulations and the use of these in concert can provide information needed to navigate through cancer disease management challenges.
Collapse
Affiliation(s)
- Małgorzata A Witek
- Department of Chemistry, The University of Kansas, Lawrence, KS 66047, USA.,Center of Biomodular Multiscale Systems for Precision Medicine, The University of Kansas, Lawrence, KS 66047, USA.,Department of Biomedical Engineering, The University of North Carolina, Chapel Hill, NC 27599, USA
| | - Rachel D Aufforth
- Department of Surgery, The University of North Carolina, Chapel Hill, NC 27599, USA
| | - Hong Wang
- Department of Biomedical Engineering, The University of North Carolina, Chapel Hill, NC 27599, USA
| | - Joyce W Kamande
- Department of Biomedical Engineering, The University of North Carolina, Chapel Hill, NC 27599, USA
| | - Joshua M Jackson
- Department of Chemistry, The University of Kansas, Lawrence, KS 66047, USA.,Center of Biomodular Multiscale Systems for Precision Medicine, The University of Kansas, Lawrence, KS 66047, USA
| | - Swathi R Pullagurla
- Department of Chemistry, The University of Kansas, Lawrence, KS 66047, USA.,Center of Biomodular Multiscale Systems for Precision Medicine, The University of Kansas, Lawrence, KS 66047, USA
| | - Mateusz L Hupert
- Department of Biomedical Engineering, The University of North Carolina, Chapel Hill, NC 27599, USA.,BioFluidica, Inc., c/o Carolina Kick-Start, 321 Bondurant Hall, Chapel Hill NC27599, USA
| | - Jerry Usary
- Department of Genetics, The University of North Carolina, Chapel Hill, NC 27599, USA.,Lineberger Comprehensive Cancer Center, The University of North Carolina, Chapel Hill, NC 27599, USA
| | - Weiya Z Wysham
- Lineberger Comprehensive Cancer Center, The University of North Carolina, Chapel Hill, NC 27599, USA.,Division of Gynecologic Oncology, Department of Obstetrics and Gynecology, UNC-Chapel Hill, NC 27599, USA
| | - Dawud Hilliard
- Lineberger Comprehensive Cancer Center, The University of North Carolina, Chapel Hill, NC 27599, USA.,Animal Histopathology Core, The University of North Carolina, Chapel Hill, NC 27599, USA
| | - Stephanie Montgomery
- Animal Histopathology Core, The University of North Carolina, Chapel Hill, NC 27599, USA.,Department of Pathology and Laboratory Medicine, The University of North Carolina, Chapel Hill, NC 27599, USA
| | - Victoria Bae-Jump
- Lineberger Comprehensive Cancer Center, The University of North Carolina, Chapel Hill, NC 27599, USA.,Division of Gynecologic Oncology, Department of Obstetrics and Gynecology, UNC-Chapel Hill, NC 27599, USA
| | - Lisa A Carey
- Lineberger Comprehensive Cancer Center, The University of North Carolina, Chapel Hill, NC 27599, USA.,Department of Medicine, Division of Hematology and Oncology, The University of North Carolina, Chapel Hill, NC 27599, USA
| | - Paola A Gehrig
- Lineberger Comprehensive Cancer Center, The University of North Carolina, Chapel Hill, NC 27599, USA.,Division of Gynecologic Oncology, Department of Obstetrics and Gynecology, UNC-Chapel Hill, NC 27599, USA
| | - Matthew I Milowsky
- Lineberger Comprehensive Cancer Center, The University of North Carolina, Chapel Hill, NC 27599, USA
| | - Charles M Perou
- Lineberger Comprehensive Cancer Center, The University of North Carolina, Chapel Hill, NC 27599, USA
| | - John T Soper
- Lineberger Comprehensive Cancer Center, The University of North Carolina, Chapel Hill, NC 27599, USA.,Division of Gynecologic Oncology, Department of Obstetrics and Gynecology, UNC-Chapel Hill, NC 27599, USA
| | - Young E Whang
- Lineberger Comprehensive Cancer Center, The University of North Carolina, Chapel Hill, NC 27599, USA
| | - Jen Jen Yeh
- Department of Surgery, The University of North Carolina, Chapel Hill, NC 27599, USA.,Lineberger Comprehensive Cancer Center, The University of North Carolina, Chapel Hill, NC 27599, USA.,Department of Pharmacology, The University of North Carolina, Chapel Hill, NC 27599, USA
| | | | - Steven A Soper
- BioEngineering Program, The University of Kansas, Lawrence, KS 66047, USA.,Department of Mechanical Engineering, The University of Kansas, Lawrence, KS 66047, USA.,Ulsan National Institute of Science and Technology, Ulsan, Republic of Korea
| |
Collapse
|
27
|
Jackson JM, Witek MA, Kamande JW, Soper SA. Materials and microfluidics: enabling the efficient isolation and analysis of circulating tumour cells. Chem Soc Rev 2017; 46:4245-4280. [PMID: 28632258 PMCID: PMC5576189 DOI: 10.1039/c7cs00016b] [Citation(s) in RCA: 113] [Impact Index Per Article: 14.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
We present a critical review of microfluidic technologies and material effects on the analyses of circulating tumour cells (CTCs) selected from the peripheral blood of cancer patients. CTCs are a minimally invasive source of clinical information that can be used to prognose patient outcome, monitor minimal residual disease, assess tumour resistance to therapeutic agents, and potentially screen individuals for the early diagnosis of cancer. The performance of CTC isolation technologies depends on microfluidic architectures, the underlying principles of isolation, and the choice of materials. We present a critical review of the fundamental principles used in these technologies and discuss their performance. We also give context to how CTC isolation technologies enable downstream analysis of selected CTCs in terms of detecting genetic mutations and gene expression that could be used to gain information that may affect patient outcome.
Collapse
|
28
|
Beije N, Kraan J, den Bakker MA, Maat APWM, van der Leest C, Cornelissen R, Van NM, Martens JWM, Aerts JGJV, Sleijfer S. Improved diagnosis and prognostication of patients with pleural malignant mesothelioma using biomarkers in pleural effusions and peripheral blood samples - a short report. Cell Oncol (Dordr) 2017; 40:511-519. [PMID: 28577209 PMCID: PMC5608799 DOI: 10.1007/s13402-017-0327-7] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/07/2017] [Indexed: 11/27/2022] Open
Abstract
Purpose There is a lack of robust and clinically utilizable markers for the diagnosis and prognostication of malignant pleural mesothelioma (MPM). This research was aimed at optimizing and exploring novel approaches to improve the diagnosis and prognostication of MPM in pleural effusions and peripheral blood samples. Methods CellSearch-based and flow cytometry-based assays using melanoma cell adhesion molecule (MCAM) to identify circulating tumor cells (CTCs) in pleural effusions and peripheral blood samples of MPM patients were optimized, validated, explored clinically and, in case of pleural effusions, compared with cytological analyses. Additionally, tumor-associated circulating endothelial cells (CECs) were measured in peripheral blood samples. The assays were performed on a MPM cohort encompassing patients with histology-confirmed MPM (n=27) and in a control cohort of patients with alternative diagnoses (n=22). Exploratory analyses on the prognostic value of all assays were also performed. Results The malignancy of MCAM-positive cells in pleural effusions from MPM patients was confirmed. The detection of MPM CTCs in pleural effusions by CellSearch showed a poor specificity. The detection of MPM CTCs in pleural effusions by flow cytometry showed a superior sensitivity (48%) to standard cytological analysis (15%) (p = 0.03). In peripheral blood, CTCs were detected in 26% of the MPN patients, whereas in 42% of the MPM patients tumor-associated CECs were detected above the upper limit of normal (ULN). In exploratory analyses the absence of CTCs in pleural effusions, and tumor-associated CECs in peripheral blood samples above the ULN, appeared to be associated with a worse overall survival. Conclusion MCAM-based flow cytometric analysis of pleural effusions is more sensitive than routine cytological analysis. Flow cytometric analysis of pleural effusions and tumor-associated CECs in peripheral blood may serve as a promising approach for the prognostication of MPM patients and, therefore, warrants further study. Electronic supplementary material The online version of this article (doi:10.1007/s13402-017-0327-7) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Nick Beije
- Department of Medical Oncology and Cancer Genomics Netherlands, Erasmus MC Cancer Institute, Erasmus University Medical Center, Wytemaweg 80, 3015, CN, Rotterdam, The Netherlands.
| | - Jaco Kraan
- Department of Medical Oncology and Cancer Genomics Netherlands, Erasmus MC Cancer Institute, Erasmus University Medical Center, Wytemaweg 80, 3015, CN, Rotterdam, The Netherlands
| | - Michael A den Bakker
- Department of Pathology, Erasmus MC Cancer Institute, Erasmus University Medical Center, Rotterdam, The Netherlands
- Department of Pathology, Maasstad Hospital, Rotterdam, The Netherlands
| | - Alexander P W M Maat
- Department of Cardiothoracic Surgery, Erasmus MC Cancer Institute, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Cor van der Leest
- Department of Pulmonary Medicine, Amphia Hospital, Breda, The Netherlands
- Department of Pulmonary Medicine, Erasmus MC Cancer Institute, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Robin Cornelissen
- Department of Pulmonary Medicine, Erasmus MC Cancer Institute, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Ngoc M Van
- Department of Medical Oncology and Cancer Genomics Netherlands, Erasmus MC Cancer Institute, Erasmus University Medical Center, Wytemaweg 80, 3015, CN, Rotterdam, The Netherlands
| | - John W M Martens
- Department of Medical Oncology and Cancer Genomics Netherlands, Erasmus MC Cancer Institute, Erasmus University Medical Center, Wytemaweg 80, 3015, CN, Rotterdam, The Netherlands
| | - Joachim G J V Aerts
- Department of Pulmonary Medicine, Amphia Hospital, Breda, The Netherlands
- Department of Pulmonary Medicine, Erasmus MC Cancer Institute, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Stefan Sleijfer
- Department of Medical Oncology and Cancer Genomics Netherlands, Erasmus MC Cancer Institute, Erasmus University Medical Center, Wytemaweg 80, 3015, CN, Rotterdam, The Netherlands
| |
Collapse
|
29
|
Xu W, Wu B, Fu L, Chen J, Wang Z, Huang F, Chen J, Zhang M, Zhang Z, Lin J, Lan R, Chen R, Chen W, Chen L, Hong J, Zhang W, Ding Y, Okunieff P, Lin J, Zhang L. Comparison of three different methods for the detection of circulating tumor cells in mice with lung metastasis. Oncol Rep 2017; 37:3219-3226. [PMID: 28498481 PMCID: PMC5442393 DOI: 10.3892/or.2017.5613] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2016] [Accepted: 04/05/2017] [Indexed: 12/12/2022] Open
Abstract
Circulating tumor cells (CTCs) represent the key step of cancer cell dissemination. The alteration of CTCs correlates with the treatment outcome and prognosis. To enrich and identify CTCs from billions of blood cells renders a very challenging task, which triggers development of several methods, including lysis of RBC plus negative or positive enrichment using antibodies, and filter membrane or spiral microfluidics to capture CTCs. To compare the advantages of different enrichment methods for CTCs, we utilized the 4T1 breast cancer cells transfected with both green fluorescent protein (GFP) and luciferase to trace CTCs in the experimental lung metastasis model. Three methods were used to detect CTCs at the same time: bioluminescence assay, smearing method, and membrane filter method. The in vivo alive mouse imaging was used to dynamically monitor the growth of lung metastases. The sensitivity and accuracy of three detection methods were compared side-by-side. Our results showed that 1) the sensitivity of bioluminescence assay was the highest, but there was no information of CTC morphology; 2) the smearing method and membrane filter method could observe the detail of CTC morphology, such as in single or in cluster, while their sensitivity was lower than bioluminescence assay; 3) A dynamic observation at a 7-day intervals, the lung metastatic cancer grew at a log speed, while CTCs were increased at a low speed. This might be due to the activated immune cells eliminating the CTCs at a speed much faster than CTCs were generated. This comparison of three CTC detection methods in mouse model suggests that bioluminescence assay could be used in quantitative study of the effect of certain agent on the suppression of CTCs, while GFP-based morphological assays could be used to study the dissemination mechanism of CTCs. The combination of both bioluminescence assay and GFP-based assay would generate more information for quantity and quality of CTCs.
Collapse
Affiliation(s)
- Weifeng Xu
- First Affiliated Hospital of Fujian Medical University, Fuzhou, Fujian 350005, P.R. China
| | - Bing Wu
- First Affiliated Hospital of Fujian Medical University, Fuzhou, Fujian 350005, P.R. China
| | - Lengxi Fu
- First Affiliated Hospital of Fujian Medical University, Fuzhou, Fujian 350005, P.R. China
| | - Junying Chen
- First Affiliated Hospital of Fujian Medical University, Fuzhou, Fujian 350005, P.R. China
| | - Zeng Wang
- First Affiliated Hospital of Fujian Medical University, Fuzhou, Fujian 350005, P.R. China
| | - Fei Huang
- First Affiliated Hospital of Fujian Medical University, Fuzhou, Fujian 350005, P.R. China
| | - Jinrong Chen
- First Affiliated Hospital of Fujian Medical University, Fuzhou, Fujian 350005, P.R. China
| | - Mei Zhang
- Department of Radiation Oncology, University of Florida, Gainesville, FL 32610, USA
| | - Zhenhuan Zhang
- Department of Radiation Oncology, University of Florida, Gainesville, FL 32610, USA
| | - Jingan Lin
- First Affiliated Hospital of Fujian Medical University, Fuzhou, Fujian 350005, P.R. China
| | - Ruilong Lan
- First Affiliated Hospital of Fujian Medical University, Fuzhou, Fujian 350005, P.R. China
| | - Ruiqing Chen
- First Affiliated Hospital of Fujian Medical University, Fuzhou, Fujian 350005, P.R. China
| | - Wei Chen
- First Affiliated Hospital of Fujian Medical University, Fuzhou, Fujian 350005, P.R. China
| | - Long Chen
- First Affiliated Hospital of Fujian Medical University, Fuzhou, Fujian 350005, P.R. China
| | - Jinsheng Hong
- First Affiliated Hospital of Fujian Medical University, Fuzhou, Fujian 350005, P.R. China
| | - Weijian Zhang
- First Affiliated Hospital of Fujian Medical University, Fuzhou, Fujian 350005, P.R. China
| | - Yuxiong Ding
- First Affiliated Hospital of Fujian Medical University, Fuzhou, Fujian 350005, P.R. China
| | - Paul Okunieff
- Department of Radiation Oncology, University of Florida, Gainesville, FL 32610, USA
| | - Jianhua Lin
- First Affiliated Hospital of Fujian Medical University, Fuzhou, Fujian 350005, P.R. China
| | - Lurong Zhang
- First Affiliated Hospital of Fujian Medical University, Fuzhou, Fujian 350005, P.R. China
| |
Collapse
|
30
|
Tampaki EC, Tampakis A, Nonni A, Kontzoglou K, Patsouris E, Kouraklis G. Nestin and cluster of differentiation 146 expression in breast cancer: Predicting early recurrence by targeting metastasis? Tumour Biol 2017; 39:1010428317691181. [PMID: 28347241 DOI: 10.1177/1010428317691181] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
The purpose of this study was to investigate the relationship between the expression of stem-cell markers nestin and cluster of differentiation 146 with clinicopathological characteristics in breast cancer and to determine whether a prognostic impact of nestin and CD146 expression exists regarding occurrence of disease relapse in breast cancer. A total of 141 patients who were histologically diagnosed with breast cancer and underwent radical operations from November 2006 to October 2013 in Laiko General Hospital, National and Kapodistrian University of Athens, were enrolled in the study. CD146 and nestin protein expression were evaluated using immunohistochemistry. Nestin expression was observed in 18.4% (26/141) of the cases, while CD146 expression was observed in 35.5% (50/141) of the cases. Nestin expression is significantly higher in younger patients with breast cancer. Nestin and CD146 expression were not correlated with the tumor size and the presence of lymph node metastasis. On the contrary, a significantly higher expression of nestin and CD146 was observed with triple-negative cancers (p < 0.0001 for both markers), low differentiated tumors (p = 0.021 for nestin and p = 0.008 for CD146), and increased Ki-67 expression (p = 0.007 for nestin and p < 0.0001 for CD146). The nestin-positive group of patients and the CD146-positive group of patients presented significantly higher rates of disease recurrence (log-rank test, p = 0.022 for nestin and p = 0.003 for CD146) with a distant metastasis, 30 months after the primary treatment. CD146 but not nestin, however, predicted independently (p = 0.047) disease recurrence. Nestin and CD146 are expressed in breast cancer cells with highly aggressive potency. They might contribute to disease relapse in breast cancer by activating the epithelial-mesenchymal transition pathway and assist tumor neovascularization.
Collapse
Affiliation(s)
- Ekaterini Christina Tampaki
- 1 2nd Department of Propedeutic Surgery, Athens University Medical School, Laiko General Hospital, Athens, Greece
| | | | - Afroditi Nonni
- 3 1st Department of Pathology, School of Medicine, National and Kapodistrian University of Athens, Athens, Greece
| | - Konstantinos Kontzoglou
- 1 2nd Department of Propedeutic Surgery, Athens University Medical School, Laiko General Hospital, Athens, Greece
| | - Efstratios Patsouris
- 3 1st Department of Pathology, School of Medicine, National and Kapodistrian University of Athens, Athens, Greece
| | - Gregory Kouraklis
- 1 2nd Department of Propedeutic Surgery, Athens University Medical School, Laiko General Hospital, Athens, Greece
| |
Collapse
|
31
|
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
|
32
|
Swennenhuis JF, van Dalum G, Zeune LL, Terstappen LWMM. Improving the CellSearch® system. Expert Rev Mol Diagn 2016; 16:1291-1305. [PMID: 27797592 DOI: 10.1080/14737159.2016.1255144] [Citation(s) in RCA: 48] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
INTRODUCTION The CellSearch® CTC test enumerates tumor cells present in 7.5 ml blood of cancer patients. improvements, extensions and different utilities of the cellsearch system are discussed in this paper. Areas covered: This paper describes work performed with the CellSearch system, which go beyond the normal scope of the test. All results from searches with the search term 'CellSearch' from Web of Science and PubMed were categorized and discussed. Expert commentary: The CellSearch Circulating Tumor Cell test captures and identifies tumor cells in blood that are associated with poor clinical outcome. How to best use CTC in clinical practice is being explored in many clinical trials. The ability to extract information from the CTC to guide therapy will expand the potential clinical utility of CTC.
Collapse
Affiliation(s)
- J F Swennenhuis
- a Medical Cell BioPhysics , University of Twente , Enschede , The Netherlands
| | - G van Dalum
- a Medical Cell BioPhysics , University of Twente , Enschede , The Netherlands
| | - L L Zeune
- a Medical Cell BioPhysics , University of Twente , Enschede , The Netherlands
| | - L W M M Terstappen
- a Medical Cell BioPhysics , University of Twente , Enschede , The Netherlands
| |
Collapse
|
33
|
Abstract
Circulating Tumor Cells (CTC) are shed from primary or secondary tumors. Prior studies have demonstrated that enumeration of CTC is a robust independent prognostic factor of progression free and overall survival in patients with early and metastatic breast cancer. CTC, as well as other circulating tumor markers, have the appealing advantages over tissue biopsy of (1) ease of collection, (2) serial evaluation, and (3) interrogation of the entire tumor burden instead of just a limited part of the tumor. Advances have been recently made in phenotyping and genotyping of CTC, which should provide insights into the predictive role of CTC for sensitivity or resistance to therapies. In addition, CTC phenotypic marker changes during the course of treatment may serve as pharmacodynamic monitoring tools. Therefore, CTC may be considered "liquid biopsies," providing prognostic and predictive clinical information as well as additional understanding of tumor heterogeneity.
Collapse
Affiliation(s)
- Costanza Paoletti
- Department of Internal Medicine, Comprehensive Cancer Center, University of Michigan Health and Hospital System, 6312 Cancer Center University of Michigan, 1500 Medical Center Drive, 48109, Ann Arbor, MI, USA
| | - Daniel F Hayes
- Department of Internal Medicine, Comprehensive Cancer Center, University of Michigan Health and Hospital System, 6312 Cancer Center University of Michigan, 1500 Medical Center Drive, 48109, Ann Arbor, MI, USA.
| |
Collapse
|
34
|
Ody T, Panth M, Sommers AD, Eid KF. Controlling the Motion of Ferrofluid Droplets Using Surface Tension Gradients and Magnetoviscous Pinning. LANGMUIR : THE ACS JOURNAL OF SURFACES AND COLLOIDS 2016; 32:6967-6976. [PMID: 27269182 DOI: 10.1021/acs.langmuir.6b01030] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/06/2023]
Abstract
This work demonstrates the controlled motion and stopping of individual ferrofluid droplets due to a surface tension gradient and a uniform magnetic field. The surface tension gradients are created by patterning hydrophilic aluminum regions, shaped as wedges, on a hydrophobic copper surface. This pattern facilitates the spontaneous motion of water-based ferrofluid droplets down the length of the wedge toward the more hydrophilic aluminum end due to a net capillarity force created by the underlying surface wettability gradient. We observed that applying a magnetic field parallel to the surface tension gradient direction has little or no effect on the droplet's motion, while a moderate perpendicular magnetic field can stop the motion altogether effectively "pinning" the droplet. In the absence of the surface tension gradient, droplets elongate in the presence of a parallel field but do not travel. This control of the motion of individual droplets might lend itself to some biomedical and lab-on-a-chip applications. The directional dependence of the magnetoviscosity observed in this work is believed to be the consequence of the formation of nanoparticle chains in the fluid due to the existence of a minority of relatively larger magnetic particles.
Collapse
Affiliation(s)
- T Ody
- Department of Physics, Miami University , Oxford, Ohio 45056, United States
| | - M Panth
- Department of Physics, Miami University , Oxford, Ohio 45056, United States
| | - A D Sommers
- Department of Mechanical and Manufacturing Engineering, Miami University , Oxford, Ohio 45056, United States
| | - K F Eid
- Department of Physics, Miami University , Oxford, Ohio 45056, United States
| |
Collapse
|
35
|
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
|
36
|
Forte VA, Barrak DK, Elhodaky M, Tung L, Snow A, Lang JE. The potential for liquid biopsies in the precision medical treatment of breast cancer. Cancer Biol Med 2016; 13:19-40. [PMID: 27144060 PMCID: PMC4850125 DOI: 10.28092/j.issn.2095-3941.2016.0007] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Currently the clinical management of breast cancer relies on relatively few prognostic/predictive clinical markers (estrogen receptor, progesterone receptor, HER2), based on primary tumor biology. Circulating biomarkers, such as circulating tumor DNA (ctDNA) or circulating tumor cells (CTCs) may enhance our treatment options by focusing on the very cells that are the direct precursors of distant metastatic disease, and probably inherently different than the primary tumor's biology. To shift the current clinical paradigm, assessing tumor biology in real time by molecularly profiling CTCs or ctDNA may serve to discover therapeutic targets, detect minimal residual disease and predict response to treatment. This review serves to elucidate the detection, characterization, and clinical application of CTCs and ctDNA with the goal of precision treatment of breast cancer.
Collapse
Affiliation(s)
- Victoria A Forte
- Department of Medicine, Division of Medical Oncology, University of Southern California (USC), Los Angeles, CA 90033, USA; USC Norris Comprehensive Cancer Center, Los Angeles, CA 90033, USA
| | - Dany K Barrak
- USC Norris Comprehensive Cancer Center, Los Angeles, CA 90033, USA; Department of Surgery, Division of Breast, Endocrine and Soft Tissue Surgery, USC, Los Angeles, CA 90033, USA
| | - Mostafa Elhodaky
- USC Norris Comprehensive Cancer Center, Los Angeles, CA 90033, USA; Department of Stem Cell and Regenerative Medicine, USC, Los Angeles, CA 90033, USA
| | - Lily Tung
- USC Norris Comprehensive Cancer Center, Los Angeles, CA 90033, USA; Department of Surgery, Division of Breast, Endocrine and Soft Tissue Surgery, USC, Los Angeles, CA 90033, USA
| | - Anson Snow
- Department of Medicine, Division of Medical Oncology, University of Southern California (USC), Los Angeles, CA 90033, USA; USC Norris Comprehensive Cancer Center, Los Angeles, CA 90033, USA
| | - Julie E Lang
- USC Norris Comprehensive Cancer Center, Los Angeles, CA 90033, USA; Department of Surgery, Division of Breast, Endocrine and Soft Tissue Surgery, USC, Los Angeles, CA 90033, USA
| |
Collapse
|
37
|
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]
|
38
|
Bulfoni M, Gerratana L, Del Ben F, Marzinotto S, Sorrentino M, Turetta M, Scoles G, Toffoletto B, Isola M, Beltrami CA, Di Loreto C, Beltrami AP, Puglisi F, Cesselli D. In patients with metastatic breast cancer the identification of circulating tumor cells in epithelial-to-mesenchymal transition is associated with a poor prognosis. Breast Cancer Res 2016; 18:30. [PMID: 26961140 PMCID: PMC4784394 DOI: 10.1186/s13058-016-0687-3] [Citation(s) in RCA: 116] [Impact Index Per Article: 12.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2015] [Accepted: 02/12/2016] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND Although recent models suggest that the detection of Circulating Tumor Cells (CTC) in epithelial-to-mesenchymal transition (EM CTC) might be related to disease progression in metastatic breast cancer (MBC) patients, current detection methods are not efficient in identifying this subpopulation of cells. Furthermore, the possible association of EM CTC with both clinicopathological features and prognosis of MBC patients has still to be demonstrated. Aims of this study were: first, to optimize a DEPArray-based protocol meant to identify, quantify and sort single, viable EM CTC and, subsequently, to test the association of EM CTC frequency with clinical data. METHODS This prospective observational study enrolled 56 MBC patients regardless of the line of treatment. Blood samples, depleted of CD45(pos) leukocytes, were stained with an antibody cocktail recognizing both epithelial and mesenchymal markers. Four CD45(neg) cell subpopulations were identified: cells expressing only epithelial markers (E CTC), cells co-expressing epithelial and mesenchymal markers (EM CTC), cells expressing only mesenchymal markers (MES) and cells negative for every tested marker (NEG). CTC subpopulations were quantified as both absolute cell count and relative frequency. The association of CTC subpopulations with clinicopathological features, progression free survival (PFS), and overall survival (OS) was explored by Wilcoxon-Mann-Whitney test and Univariate Cox Regression Analysis, respectively. RESULTS By employing the DEPArray-based strategy, we were able to assess the presence of cells pertaining to the above-described classes in every MBC patient. We observed a significant association between specific CD45(neg) subpopulations and tumor subtypes (e.g. NEG and triple negative), proliferation (NEG and Ki67 expression) and sites of metastatic spread (e.g. E CTC and bone; NEG and brain). Importantly, the fraction of CD45(neg) cells co-expressing epithelial and mesenchymal markers (EM CTC) was significantly associated with poorer PFS and OS, computed, this latter, both from the diagnosis of a stage IV disease and from the initial CTC assessment. CONCLUSION This study suggests the importance of dissecting the heterogeneity of CTC in MBC. Precise characterization of CTC could help in estimating both metastatization pattern and outcome, driving clinical decision-making and surveillance strategies.
Collapse
Affiliation(s)
- Michela Bulfoni
- Department of Medical and Biological Sciences, University of Udine, P.le Kolbe 4, 33100, Udine, Italy
| | - Lorenzo Gerratana
- Department of Medical and Biological Sciences, University of Udine, P.le Kolbe 4, 33100, Udine, Italy
- Department of Oncology, University Hospital of Udine, P.le S.Maria della Misericordia 15, 33100, Udine, Italy
| | - Fabio Del Ben
- Department of Medical and Biological Sciences, University of Udine, P.le Kolbe 4, 33100, Udine, Italy
| | - Stefania Marzinotto
- Institute of Pathology, University Hospital of Udine, P.le S.Maria della Misericordia 15, 33100, Udine, Italy
| | - Marisa Sorrentino
- Department of Medical and Biological Sciences, University of Udine, P.le Kolbe 4, 33100, Udine, Italy
- Institute of Pathology, University Hospital of Udine, P.le S.Maria della Misericordia 15, 33100, Udine, Italy
| | - Matteo Turetta
- Department of Medical and Biological Sciences, University of Udine, P.le Kolbe 4, 33100, Udine, Italy
- Institute of Pathology, University Hospital of Udine, P.le S.Maria della Misericordia 15, 33100, Udine, Italy
| | - Giacinto Scoles
- Department of Medical and Biological Sciences, University of Udine, P.le Kolbe 4, 33100, Udine, Italy
| | - Barbara Toffoletto
- Department of Medical and Biological Sciences, University of Udine, P.le Kolbe 4, 33100, Udine, Italy
| | - Miriam Isola
- Department of Medical and Biological Sciences, University of Udine, P.le Kolbe 4, 33100, Udine, Italy
| | - Carlo Alberto Beltrami
- Department of Medical and Biological Sciences, University of Udine, P.le Kolbe 4, 33100, Udine, Italy
| | - Carla Di Loreto
- Department of Medical and Biological Sciences, University of Udine, P.le Kolbe 4, 33100, Udine, Italy
- Institute of Pathology, University Hospital of Udine, P.le S.Maria della Misericordia 15, 33100, Udine, Italy
| | - Antonio Paolo Beltrami
- Department of Medical and Biological Sciences, University of Udine, P.le Kolbe 4, 33100, Udine, Italy.
| | - Fabio Puglisi
- Department of Medical and Biological Sciences, University of Udine, P.le Kolbe 4, 33100, Udine, Italy
- Department of Oncology, University Hospital of Udine, P.le S.Maria della Misericordia 15, 33100, Udine, Italy
| | - Daniela Cesselli
- Department of Medical and Biological Sciences, University of Udine, P.le Kolbe 4, 33100, Udine, Italy.
| |
Collapse
|
39
|
Reijm EA, Sieuwerts AM, Smid M, Vries JBD, Mostert B, Onstenk W, Peeters D, Dirix LY, Seynaeve CM, Jager A, de Jongh FE, Hamberg P, van Galen A, Kraan J, Jansen MPHM, Gratama JW, Foekens JA, Martens JWM, Berns EMJJ, Sleijfer S. An 8-gene mRNA expression profile in circulating tumor cells predicts response to aromatase inhibitors in metastatic breast cancer patients. BMC Cancer 2016; 16:123. [PMID: 26892682 PMCID: PMC4759736 DOI: 10.1186/s12885-016-2155-y] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2015] [Accepted: 02/10/2016] [Indexed: 12/29/2022] Open
Abstract
Background Molecular characterization of circulating tumor cells (CTC) is promising for personalized medicine. We aimed to identify a CTC gene expression profile predicting outcome to first-line aromatase inhibitors in metastatic breast cancer (MBC) patients. Methods: CTCs were isolated from 78 MBC patients before treatment start. mRNA expression levels of 96 genes were measured by quantitative reverse transcriptase polymerase chain reaction. After applying predefined exclusion criteria based on lack of sufficient RNA quality and/or quantity, the data from 45 patients were used to construct a gene expression profile to predict poor responding patients, defined as disease progression or death <9 months, by a leave-one-out cross validation. Results Of the 45 patients, 19 were clinically classified as poor responders. To identify them, the 75 % most variable genes were used to select genes differentially expressed between good and poor responders. An 8-gene CTC predictor was significantly associated with outcome (Hazard Ratio [HR] 4.40, 95 % Confidence Interval [CI]: 2.17–8.92, P < 0.001). This predictor identified poor responding patients with a sensitivity of 63 % and a positive predictive value of 75 %, while good responding patients were correctly predicted in 85 % of the cases. In multivariate Cox regression analysis, including CTC count at baseline, the 8-gene CTC predictor was the only factor independently associated with outcome (HR 4.59 [95 % CI: 2.11–9.56], P < 0.001). This 8-gene signature was not associated with outcome in a group of 71 MBC patients treated with systemic treatments other than AI. Conclusions An 8-gene CTC predictor was identified which discriminates good and poor outcome to first-line aromatase inhibitors in MBC patients. Although results need to be validated, this study underscores the potential of molecular characterization of CTCs. Electronic supplementary material The online version of this article (doi:10.1186/s12885-016-2155-y) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Esther A Reijm
- Department of Medical Oncology and Cancer Genomics Netherlands, Erasmus MC - Cancer Institute, Erasmus University Medical Center, Room He 116, P.O. Box 2040, Rotterdam, 3000 CA, The Netherlands
| | - Anieta M Sieuwerts
- Department of Medical Oncology and Cancer Genomics Netherlands, Erasmus MC - Cancer Institute, Erasmus University Medical Center, Room He 116, P.O. Box 2040, Rotterdam, 3000 CA, The Netherlands
| | - Marcel Smid
- Department of Medical Oncology and Cancer Genomics Netherlands, Erasmus MC - Cancer Institute, Erasmus University Medical Center, Room He 116, P.O. Box 2040, Rotterdam, 3000 CA, The Netherlands
| | - Joan Bolt-de Vries
- Department of Medical Oncology and Cancer Genomics Netherlands, Erasmus MC - Cancer Institute, Erasmus University Medical Center, Room He 116, P.O. Box 2040, Rotterdam, 3000 CA, The Netherlands
| | - Bianca Mostert
- Department of Medical Oncology and Cancer Genomics Netherlands, Erasmus MC - Cancer Institute, Erasmus University Medical Center, Room He 116, P.O. Box 2040, Rotterdam, 3000 CA, The Netherlands
| | - Wendy Onstenk
- Department of Medical Oncology and Cancer Genomics Netherlands, Erasmus MC - Cancer Institute, Erasmus University Medical Center, Room He 116, P.O. Box 2040, Rotterdam, 3000 CA, The Netherlands
| | - Dieter Peeters
- Translational Cancer Research Unit, Oncology Center GZA Hospitals Sint-Augustinus and Department of Oncology, University of Antwerp, Antwerp, Belgium
| | - Luc Y Dirix
- Translational Cancer Research Unit, Oncology Center GZA Hospitals Sint-Augustinus and Department of Oncology, University of Antwerp, Antwerp, Belgium
| | - Caroline M Seynaeve
- Department of Medical Oncology and Cancer Genomics Netherlands, Erasmus MC - Cancer Institute, Erasmus University Medical Center, Room He 116, P.O. Box 2040, Rotterdam, 3000 CA, The Netherlands
| | - Agnes Jager
- Department of Medical Oncology and Cancer Genomics Netherlands, Erasmus MC - Cancer Institute, Erasmus University Medical Center, Room He 116, P.O. Box 2040, Rotterdam, 3000 CA, The Netherlands
| | - Felix E de Jongh
- Department of Internal Medicine, Ikazia Hospital, Rotterdam, The Netherlands
| | - Paul Hamberg
- Department of Internal Medicine, Sint Franciscus Gasthuis, Rotterdam, The Netherlands
| | - Anne van Galen
- Department of Medical Oncology and Cancer Genomics Netherlands, Erasmus MC - Cancer Institute, Erasmus University Medical Center, Room He 116, P.O. Box 2040, Rotterdam, 3000 CA, The Netherlands
| | - Jaco Kraan
- Department of Medical Oncology and Cancer Genomics Netherlands, Erasmus MC - Cancer Institute, Erasmus University Medical Center, Room He 116, P.O. Box 2040, Rotterdam, 3000 CA, The Netherlands
| | - Maurice P H M Jansen
- Department of Medical Oncology and Cancer Genomics Netherlands, Erasmus MC - Cancer Institute, Erasmus University Medical Center, Room He 116, P.O. Box 2040, Rotterdam, 3000 CA, The Netherlands
| | - Jan W Gratama
- Department of Medical Oncology and Cancer Genomics Netherlands, Erasmus MC - Cancer Institute, Erasmus University Medical Center, Room He 116, P.O. Box 2040, Rotterdam, 3000 CA, The Netherlands
| | - John A Foekens
- Department of Medical Oncology and Cancer Genomics Netherlands, Erasmus MC - Cancer Institute, Erasmus University Medical Center, Room He 116, P.O. Box 2040, Rotterdam, 3000 CA, The Netherlands
| | - John W M Martens
- Department of Medical Oncology and Cancer Genomics Netherlands, Erasmus MC - Cancer Institute, Erasmus University Medical Center, Room He 116, P.O. Box 2040, Rotterdam, 3000 CA, The Netherlands
| | - Els M J J Berns
- Department of Medical Oncology and Cancer Genomics Netherlands, Erasmus MC - Cancer Institute, Erasmus University Medical Center, Room He 116, P.O. Box 2040, Rotterdam, 3000 CA, The Netherlands
| | - Stefan Sleijfer
- Department of Medical Oncology and Cancer Genomics Netherlands, Erasmus MC - Cancer Institute, Erasmus University Medical Center, Room He 116, P.O. Box 2040, Rotterdam, 3000 CA, The Netherlands.
| |
Collapse
|
40
|
Peng C, Wallwiener M, Rudolph A, Ćuk K, Eilber U, Celik M, Modugno C, Trumpp A, Heil J, Marmé F, Madhavan D, Nees J, Riethdorf S, Schott S, Sohn C, Pantel K, Schneeweiss A, Chang-Claude J, Yang R, Burwinkel B. Plasma hyaluronic acid level as a prognostic and monitoring marker of metastatic breast cancer. Int J Cancer 2016; 138:2499-509. [PMID: 26686298 DOI: 10.1002/ijc.29975] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2015] [Revised: 10/30/2015] [Accepted: 11/13/2015] [Indexed: 11/12/2022]
Abstract
Conventional tumor markers have limited value for prognostication and treatment monitoring in metastatic breast cancer (MBC) patients and novel circulating tumor markers therefore need to be explored. Hyaluronic acid (HA) is a major macropolysaccharide in the extracellular matrix and is reported to be associated with tumor progression. In our study, we investigated plasma HA level with respect to progression free survival (PFS) and overall survival (OS), as well as the treatment monitoring value in MBC patients. The prognostic value of plasma HA level was investigated in a discovery cohort of 212 MBC patients with 2.5-year follow-up and validated in an independent validation cohort of 334 patients with 5-year follow-up. The treatment monitoring value of plasma HA level was investigated in 61 MBC patients from discovery cohort who had been radiographically examined after first complete cycle of chemo therapy. We found a robust association between high plasma HA level and poor prognosis of MBC patients in both discovery (pPFS = 7.92 × 10(-6) and pOS = 5.27 × 10(-5)) and validation studies (pPFS = 3.66 × 10(-4) and pOS = 1.43 × 10(-4)). In the discovery cohort, the plasma HA level displayed independent prognostic value after adjusted for age and clinicopathological factors, with respect to PFS and OS. Further, the decrease of plasma HA level displayed good concordance with treatment response evaluated by radiographic examination (AUC = 0.79). Plasma HA level displays prognostic value, as well as treatment monitoring value for MBC patients.
Collapse
Affiliation(s)
- Cike Peng
- Molecular Epidemiology, German Cancer Research Center (DKFZ), Heidelberg, Germany.,Molecular Biology of Breast Cancer, Department of Obstetrics and Gynecology, University of Heidelberg, Heidelberg, Germany
| | - Markus Wallwiener
- Department of Obstetrics and Gynecology, University of Heidelberg, Heidelberg, Germany.,National Center for Tumor Diseases, University of Heidelberg, Heidelberg, Germany
| | - Anja Rudolph
- Division of Genetic Epidemiology, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Katarina Ćuk
- Molecular Epidemiology, German Cancer Research Center (DKFZ), Heidelberg, Germany.,Molecular Biology of Breast Cancer, Department of Obstetrics and Gynecology, University of Heidelberg, Heidelberg, Germany
| | - Ursula Eilber
- Division of Genetic Epidemiology, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Muhabbet Celik
- Division of Genetic Epidemiology, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Caroline Modugno
- Department of Obstetrics and Gynecology, University of Heidelberg, Heidelberg, Germany.,National Center for Tumor Diseases, University of Heidelberg, Heidelberg, Germany
| | - Andreas Trumpp
- Hi-STEM-Heidelberg Institute for Stem Cell Technology and Experimental Medicine GmbH, Heidelberg, Germany.,Division of Stem Cells and Cancer, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Jörg Heil
- Department of Obstetrics and Gynecology, University of Heidelberg, Heidelberg, Germany
| | - Frederik Marmé
- Molecular Biology of Breast Cancer, Department of Obstetrics and Gynecology, University of Heidelberg, Heidelberg, Germany
| | - Dharanija Madhavan
- Molecular Epidemiology, German Cancer Research Center (DKFZ), Heidelberg, Germany.,Molecular Biology of Breast Cancer, Department of Obstetrics and Gynecology, University of Heidelberg, Heidelberg, Germany
| | - Juliane Nees
- Department of Obstetrics and Gynecology, University of Heidelberg, Heidelberg, Germany.,National Center for Tumor Diseases, University of Heidelberg, Heidelberg, Germany
| | - Sabine Riethdorf
- Department of Tumor Biology, University Hospital Hamburg-Eppendorf, Hamburg, Germany
| | - Sarah Schott
- Department of Obstetrics and Gynecology, University of Heidelberg, Heidelberg, Germany
| | - Christof Sohn
- Department of Obstetrics and Gynecology, University of Heidelberg, Heidelberg, Germany
| | - Klaus Pantel
- Department of Tumor Biology, University Hospital Hamburg-Eppendorf, Hamburg, Germany
| | - Andreas Schneeweiss
- Department of Obstetrics and Gynecology, University of Heidelberg, Heidelberg, Germany.,National Center for Tumor Diseases, University of Heidelberg, Heidelberg, Germany
| | - Jenny Chang-Claude
- Division of Genetic Epidemiology, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Rongxi Yang
- Molecular Epidemiology, German Cancer Research Center (DKFZ), Heidelberg, Germany.,Molecular Biology of Breast Cancer, Department of Obstetrics and Gynecology, University of Heidelberg, Heidelberg, Germany
| | - Barbara Burwinkel
- Molecular Epidemiology, German Cancer Research Center (DKFZ), Heidelberg, Germany.,Molecular Biology of Breast Cancer, Department of Obstetrics and Gynecology, University of Heidelberg, Heidelberg, Germany
| |
Collapse
|
41
|
Bidard FC, Proudhon C, Pierga JY. Circulating tumor cells in breast cancer. Mol Oncol 2016; 10:418-30. [PMID: 26809472 PMCID: PMC5528978 DOI: 10.1016/j.molonc.2016.01.001] [Citation(s) in RCA: 137] [Impact Index Per Article: 15.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2015] [Revised: 12/12/2015] [Accepted: 01/04/2016] [Indexed: 01/06/2023] Open
Abstract
Over the past decade, technically reliable circulating tumor cell (CTC) detection methods allowed the collection of large datasets of CTC counts in cancer patients. These data can be used either as a dynamic prognostic biomarker or as tumor material for “liquid biopsy”. Breast cancer appears to be the cancer type in which CTC have been the most extensively studied so far, with level‐of‐evidence‐1 studies supporting the clinical validity of CTC count in both early and metastatic stage. This review summarizes and discusses the clinical results obtained in breast cancer patients, the issues faced by the molecular characterization of CTC and the biological findings about cancer biology and metastasis that were obtained from CTC. In metastatic breast cancer, CTC count is a level‐of‐evidence 1 prognostic dynamic biomarker. Several interventional trials are ongoing to demonstrate the clinical utility of CTC detection in metastatic breast cancer. In early breast cancer, CTC count is also a prognostic biomarker, not correlated with the other usual prognostic factors. Molecular characterization of CTC is promising, trials with anti‐HER2 therapy are ongoing.
Collapse
Affiliation(s)
- Francois-Clement Bidard
- Institut Curie, PSL Research University, SiRIC, Laboratory of Circulating Tumor Biomarkers, Paris, France; Institut Curie, PSL Research University, Department of Medical Oncology, Paris, France
| | - Charlotte Proudhon
- Institut Curie, PSL Research University, SiRIC, Laboratory of Circulating Tumor Biomarkers, Paris, France
| | - Jean-Yves Pierga
- Institut Curie, PSL Research University, SiRIC, Laboratory of Circulating Tumor Biomarkers, Paris, France; Institut Curie, PSL Research University, Department of Medical Oncology, Paris, France; Université Paris Descartes, Paris, France.
| |
Collapse
|
42
|
Schneck H, Gierke B, Uppenkamp F, Behrens B, Niederacher D, Stoecklein NH, Templin MF, Pawlak M, Fehm T, Neubauer H. EpCAM-Independent Enrichment of Circulating Tumor Cells in Metastatic Breast Cancer. PLoS One 2015; 10:e0144535. [PMID: 26695635 PMCID: PMC4687932 DOI: 10.1371/journal.pone.0144535] [Citation(s) in RCA: 79] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2015] [Accepted: 11/19/2015] [Indexed: 12/18/2022] Open
Abstract
Circulating tumor cells (CTCs) are the potential precursors of metastatic disease. Most assays established for the enumeration of CTCs so far-including the gold standard CellSearch-rely on the expression of the cell surface marker epithelial cell adhesion molecule (EpCAM). But, these approaches may not detect CTCs that express no/low levels of EpCAM, e.g. by undergoing epithelial-to-mesenchymal transition (EMT). Here we present an enrichment strategy combining different antibodies specific for surface proteins and extracellular matrix (ECM) components to capture an EpCAMlow/neg cell line and EpCAMneg CTCs from blood samples of breast cancer patients depleted for EpCAM-positive cells. The expression of respective proteins (Trop2, CD49f, c-Met, CK8, CD44, ADAM8, CD146, TEM8, CD47) was verified by immunofluorescence on EpCAMpos (e.g. MCF7, SKBR3) and EpCAMlow/neg (MDA-MB-231) breast cancer cell lines. To test antibodies and ECM proteins (e.g. hyaluronic acid (HA), collagen I, laminin) for capturing EpCAMneg cells, the capture molecules were first spotted in a single- and multi-array format onto aldehyde-coated glass slides. Tumor cell adhesion of EpCAMpos/neg cell lines was then determined and visualized by Coomassie/MitoTracker staining. In consequence, marginal binding of EpCAMlow/neg MDA-MB-231 cells to EpCAM-antibodies could be observed. However, efficient adhesion/capturing of EpCAMlow/neg cells could be achieved via HA and immobilized antibodies against CD49f and Trop2. Optimal capture conditions were then applied to immunomagnetic beads to detect EpCAMneg CTCs from clinical samples. Captured CTCs were verified/quantified by immunofluorescence staining for anti-pan-Cytokeratin (CK)-FITC/anti-CD45 AF647/DAPI. In total, in 20 out of 29 EpCAM-depleted fractions (69%) from 25 metastatic breast cancer patients additional EpCAMneg CTCs could be identified [range of 1-24 CTCs per sample] applying Trop2, CD49f, c-Met, CK8 and/or HA magnetic enrichment. EpCAMneg dual-positive (CKpos/CD45pos) cells could be traced in 28 out of 29 samples [range 1-480]. By single-cell array-based comparative genomic hybridization we were able to demonstrate the malignant nature of one EpCAMneg subpopulation. In conclusion, we established a novel enhanced CTC enrichment strategy to capture EpCAMneg CTCs from clinical blood samples by targeting various cell surface antigens with antibody mixtures and ECM components.
Collapse
Affiliation(s)
- Helen Schneck
- Department of Obstetrics and Gynecology, University Hospital and Medical Faculty of the Heinrich-Heine University Duesseldorf, Duesseldorf, Germany
| | - Berthold Gierke
- NMI Natural and Medical Sciences Institute at the University of Tuebingen, Reutlingen, Germany
| | - Frauke Uppenkamp
- Department of Obstetrics and Gynecology, University Hospital and Medical Faculty of the Heinrich-Heine University Duesseldorf, Duesseldorf, Germany
| | - Bianca Behrens
- Department of General, Visceral and Pediatric Surgery, University Hospital and Medical Faculty of the Heinrich-Heine University Duesseldorf, Duesseldorf, Germany
| | - Dieter Niederacher
- Department of Obstetrics and Gynecology, University Hospital and Medical Faculty of the Heinrich-Heine University Duesseldorf, Duesseldorf, Germany
| | - Nikolas H. Stoecklein
- Department of General, Visceral and Pediatric Surgery, University Hospital and Medical Faculty of the Heinrich-Heine University Duesseldorf, Duesseldorf, Germany
| | - Markus F. Templin
- NMI Natural and Medical Sciences Institute at the University of Tuebingen, Reutlingen, Germany
| | - Michael Pawlak
- NMI Natural and Medical Sciences Institute at the University of Tuebingen, Reutlingen, Germany
| | - Tanja Fehm
- Department of Obstetrics and Gynecology, University Hospital and Medical Faculty of the Heinrich-Heine University Duesseldorf, Duesseldorf, Germany
| | - Hans Neubauer
- Department of Obstetrics and Gynecology, University Hospital and Medical Faculty of the Heinrich-Heine University Duesseldorf, Duesseldorf, Germany
| | | |
Collapse
|
43
|
Shaw Bagnall J, Byun S, Begum S, Miyamoto DT, Hecht VC, Maheswaran S, Stott SL, Toner M, Hynes RO, Manalis SR. Deformability of Tumor Cells versus Blood Cells. Sci Rep 2015; 5:18542. [PMID: 26679988 PMCID: PMC4683468 DOI: 10.1038/srep18542] [Citation(s) in RCA: 88] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2015] [Accepted: 11/19/2015] [Indexed: 01/04/2023] Open
Abstract
The potential for circulating tumor cells (CTCs) to elucidate the process of cancer metastasis and inform clinical decision-making has made their isolation of great importance. However, CTCs are rare in the blood, and universal properties with which to identify them remain elusive. As technological advancements have made single-cell deformability measurements increasingly routine, the assessment of physical distinctions between tumor cells and blood cells may provide insight into the feasibility of deformability-based methods for identifying CTCs in patient blood. To this end, we present an initial study assessing deformability differences between tumor cells and blood cells, indicated by the length of time required for them to pass through a microfluidic constriction. Here, we demonstrate that deformability changes in tumor cells that have undergone phenotypic shifts are small compared to differences between tumor cell lines and blood cells. Additionally, in a syngeneic mouse tumor model, cells that are able to exit a tumor and enter circulation are not required to be more deformable than the cells that were first injected into the mouse. However, a limited study of metastatic prostate cancer patients provides evidence that some CTCs may be more mechanically similar to blood cells than to typical tumor cell lines.
Collapse
Affiliation(s)
- Josephine Shaw Bagnall
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA
| | - Sangwon Byun
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA
| | - Shahinoor Begum
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA
- Howard Hughes Medical Institute, Massachusetts Institute of Technology, Cambridge, MA
| | - David T. Miyamoto
- Massachusetts General Hospital Cancer Center, Harvard Medical School, Boston, MA
- Department of Radiation Oncology, Massachusetts General Hospital, Harvard Medical School, Boston, MA
| | - Vivian C. Hecht
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA
| | - Shyamala Maheswaran
- Massachusetts General Hospital Cancer Center, Harvard Medical School, Boston, MA
- Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA
| | - Shannon L. Stott
- Massachusetts General Hospital Cancer Center, Harvard Medical School, Boston, MA
- Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA
- Massachusetts General Hospital Center for Engineering and Medicine, Harvard Medical School, Boston, MA
| | - Mehmet Toner
- Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA
- Massachusetts General Hospital Center for Engineering and Medicine, Harvard Medical School, Boston, MA
| | - Richard O. Hynes
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA
- Howard Hughes Medical Institute, Massachusetts Institute of Technology, Cambridge, MA
- Department of Biology, Massachusetts Institute of Technology, Cambridge, MA
| | - Scott R. Manalis
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA
- Department of Mechanical Engineering, Massachusetts Institute of Technology, Cambridge, MA
| |
Collapse
|
44
|
Cavazzini F, Campioni D, Ferrari L, Buldini B, Bardi MA, Michielotto B, Lazzari MC, Ongari M, Dabusti M, Daghia G, Sofritti O, Basso G, Lanza F, Cuneo A. Expression of the immunoglobulin superfamily cell membrane adhesion molecule Cd146 in acute leukemia. CYTOMETRY PART B-CLINICAL CYTOMETRY 2015; 90:247-56. [PMID: 26102234 DOI: 10.1002/cyto.b.21267] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/07/2014] [Revised: 06/12/2015] [Accepted: 06/17/2015] [Indexed: 01/06/2023]
Abstract
BACKGROUND The expression of the immunoglobulin superfamily cell membrane adhesion molecule CD146 has been reported on several normal and pathological cell types in human. The aim of this study was to investigate CD146 expression in acute leukemia using a multiparametric cytofluorimetric approach. METHODS Cytofluorimetric and cytogenetic studies were performed on peripheral blood and bone marrow samples from 162 patients with acute myeloid leukemia (AML, n = 121) and acute lymphoblastic leukemia (ALL, n = 41). ALL patients were subdivided in B-ALL (n = 38) and T-ALL (n = 3). Adult (n = 18) and pediatric (n = 20) B-ALL were considered as a whole group. RESULTS Four out of 121 (3.3%) AML cases, 14/38 (36.8%) B-ALL, and 2/3 (66.6%) T-ALL expressed CD146 on 12-98% of blasts (p < 0.001). CD146 expression was not observed in 10 healthy controls. Among B-ALL CD146-positive cases, 78.6% were associated with a "common"/BII-ALL and 21.4% with a pre-B/BIII-ALL immunophenotype while pro-B/BI-ALL and mature-B/BIV-ALL cases were CD146-negative. Statistical analysis showed CD146 expression strongly associated with Ph+ positivity in B-ALL with the highest percentage of CD146-positive blasts in all Ph-positive B-ALL cases (84 ± 22% Ph-positive B-ALL SD vs. 40 ± 24% SD in Ph-negative B-ALL; p < 0,001). CONCLUSION In our series, CD146 was expressed in all cases of Ph-positive B-ALL and in the vast majority of T-ALL, whereas it was rarely expressed by AML blasts. We suggest that CD146 may be considered as an additional marker for acute lymphoblastic leukemia diagnosis and monitoring of minimal residual disease in those cases which are CD146-positive at diagnosis. © 2015 International Clinical Cytometry Society.
Collapse
Affiliation(s)
| | - Diana Campioni
- Section of Hematology, University-S.Anna Hospital, Ferrara, Italy
| | - Luisa Ferrari
- Section of Hematology, University-S.Anna Hospital, Ferrara, Italy
| | - Barbara Buldini
- Onco-Hematology Division, SDB Department, University of Padova, Italy
| | | | | | | | - Manuele Ongari
- Section of Hematology, Hospital of Cremona, Cremona, Italy
| | - Melissa Dabusti
- Section of Hematology, University-S.Anna Hospital, Ferrara, Italy
| | - Giulia Daghia
- Section of Hematology, University-S.Anna Hospital, Ferrara, Italy
| | - Olga Sofritti
- Section of Hematology, University-S.Anna Hospital, Ferrara, Italy
| | - Giuseppe Basso
- Onco-Hematology Division, SDB Department, University of Padova, Italy
| | | | - Antonio Cuneo
- Section of Hematology, University-S.Anna Hospital, Ferrara, Italy
| |
Collapse
|
45
|
Mostert B, Sieuwerts A, Kraan J, Bolt-de Vries J, van der Spoel P, van Galen A, Peeters D, Dirix L, Seynaeve C, Jager A, de Jongh F, Hamberg P, Stouthard J, Kehrer D, Look M, Smid M, Gratama J, Foekens J, Martens J, Sleijfer S. Gene expression profiles in circulating tumor cells to predict prognosis in metastatic breast cancer patients. Ann Oncol 2015; 26:510-6. [DOI: 10.1093/annonc/mdu557] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
|
46
|
Hart CD, Galardi F, De Luca F, Pestrin M, Di Leo A. Circulating Tumour Cells as Liquid Biopsy in Breast Cancer—Advancing from Prognostic to Predictive Potential. CURRENT BREAST CANCER REPORTS 2015. [DOI: 10.1007/s12609-014-0177-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
|
47
|
Mostert B, Sieuwerts AM, Bolt-de Vries J, Kraan J, Lalmahomed Z, van Galen A, van der Spoel P, de Weerd V, Ramírez-Moreno R, Smid M, Verhoef C, IJzermans JNM, Gratama JW, Sleijfer S, Foekens JA, Martens JWM. mRNA expression profiles in circulating tumor cells of metastatic colorectal cancer patients. Mol Oncol 2015; 9:920-32. [PMID: 25655581 DOI: 10.1016/j.molonc.2015.01.001] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2014] [Revised: 12/05/2014] [Accepted: 01/02/2015] [Indexed: 12/17/2022] Open
Abstract
INTRODUCTION The molecular characterization of circulating tumor cells (CTCs) is a promising tool for the repeated and non-invasive evaluation of predictive and prognostic factors. Challenges associated with CTC characterization using the only FDA approved method for CTC enumeration, the CellSearch technique, include the presence of an excess of leukocytes in CTC-enriched blood fractions. Here we aimed to identify colorectal tumor-specific gene expression levels in the blood of patients with and without detectable CTCs according to CellSearch criteria. MATERIALS AND METHODS Blood of 30 healthy donors (HDs) and 142 metastatic colorectal cancer (mCRC) patients was subjected to CellSearch CTC enumeration and isolation. In all samples, 95 mRNAs were measured by reverse transcriptase quantitative PCR (RT-qPCR). HD blood samples and patient samples with three or more CTCs were compared to identify CTC-specific mRNAs. Patient samples without detectable CTCs were separately analyzed. RESULTS Thirty-four CTC-specific mRNAs were higher expressed in patients with ≥3 CTCs compared with HDs (Mann-Whitney U-test P < 0.05). Among patients without detectable CTCs, a HD-unlike subgroup was identified which could be distinguished from HDs by the expression of epithelial genes such as KRT19, KRT20 and AGR2. Also, in an independent patient set, a similar HD-unlike group could be identified among the patients without detectable CTCs according to the CellSearch system. CONCLUSION Extensive molecular characterization of colorectal CTCs is feasible and a subgroup of patients without detectable CTCs according to CellSearch criteria bears circulating tumor load, which may have clinical consequences. This CTC-specific gene panel for mCRC patients may enable the exploration of CTC characterization as a novel means to further individualize cancer treatment.
Collapse
Affiliation(s)
- Bianca Mostert
- Department of Medical Oncology, Erasmus MC Cancer Institute, Erasmus University Medical Center, Rotterdam, Netherlands
| | - Anieta M Sieuwerts
- Department of Medical Oncology and Cancer Genomics Netherlands, Erasmus MC Cancer Institute, Erasmus University Medical Center, Rotterdam, Netherlands
| | - Joan Bolt-de Vries
- Department of Medical Oncology, Erasmus MC Cancer Institute, Erasmus University Medical Center, Rotterdam, Netherlands
| | - Jaco Kraan
- Department of Medical Oncology, Erasmus MC Cancer Institute, Erasmus University Medical Center, Rotterdam, Netherlands
| | - Zarina Lalmahomed
- Department of Surgery, Erasmus University Medical Center, Rotterdam, Netherlands
| | - Anne van Galen
- Department of Medical Oncology, Erasmus MC Cancer Institute, Erasmus University Medical Center, Rotterdam, Netherlands
| | - Petra van der Spoel
- Department of Medical Oncology, Erasmus MC Cancer Institute, Erasmus University Medical Center, Rotterdam, Netherlands
| | - Vanja de Weerd
- Department of Medical Oncology, Erasmus MC Cancer Institute, Erasmus University Medical Center, Rotterdam, Netherlands
| | - Raquel Ramírez-Moreno
- Department of Medical Oncology, Erasmus MC Cancer Institute, Erasmus University Medical Center, Rotterdam, Netherlands
| | - Marcel Smid
- Department of Medical Oncology and Cancer Genomics Netherlands, Erasmus MC Cancer Institute, Erasmus University Medical Center, Rotterdam, Netherlands
| | - Cornelis Verhoef
- Department of Surgery, Erasmus University Medical Center, Rotterdam, Netherlands
| | - Jan N M IJzermans
- Department of Surgery, Erasmus University Medical Center, Rotterdam, Netherlands
| | - Jan W Gratama
- Department of Medical Oncology, Erasmus MC Cancer Institute, Erasmus University Medical Center, Rotterdam, Netherlands
| | - Stefan Sleijfer
- Department of Medical Oncology, Erasmus MC Cancer Institute, Erasmus University Medical Center, Rotterdam, Netherlands; Department of Medical Oncology and Cancer Genomics Netherlands, Erasmus MC Cancer Institute, Erasmus University Medical Center, Rotterdam, Netherlands
| | - John A Foekens
- Department of Medical Oncology, Erasmus MC Cancer Institute, Erasmus University Medical Center, Rotterdam, Netherlands
| | - John W M Martens
- Department of Medical Oncology and Cancer Genomics Netherlands, Erasmus MC Cancer Institute, Erasmus University Medical Center, Rotterdam, Netherlands.
| |
Collapse
|
48
|
Onstenk W, Kraan J, Mostert B, Timmermans MM, Charehbili A, Smit VTHBM, Kroep JR, Nortier JWR, van de Ven S, Heijns JB, Kessels LW, van Laarhoven HWM, Bos MMEM, van de Velde CJH, Gratama JW, Sieuwerts AM, Martens JWM, Foekens JA, Sleijfer S. Improved Circulating Tumor Cell Detection by a Combined EpCAM and MCAM CellSearch Enrichment Approach in Patients with Breast Cancer Undergoing Neoadjuvant Chemotherapy. Mol Cancer Ther 2014; 14:821-7. [PMID: 25552367 DOI: 10.1158/1535-7163.mct-14-0653] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2014] [Accepted: 12/11/2014] [Indexed: 11/16/2022]
Abstract
Circulating tumor cells (CTC) are detected by the CellSearch System in 20% to 25% of patients with primary breast cancer (pBC). To improve CTC detection, we investigated melanoma cell adhesion molecule (MCAM) as enrichment marker next to epithelial cell adhesion molecule (EpCAM) and tested the clinical relevance of MCAM-positive CTCs in patients with HER2-negative stage II/III pBC starting neoadjuvant chemotherapy (NAC) in the NEOZOTAC trial. Using the CellSearch System, EpCAM-positive and MCAM-positive CTCs were separately enriched from 7.5 mL blood, at baseline and after the first NAC cycle. Circulating endothelial cells (CEC) were measured using flow cytometry. Primary objective was to improve the CTC detection rate to ≥ 40% combining EpCAM/MCAM. Correlations of CTC and CEC counts and pathologic complete response (pCR) were also explored. At baseline, we detected EpCAM-positive and MCAM-positive CTCs in 12 of 68 (18%) and 8 of 68 (12%) patients, respectively. After one cycle, this was 7 of 44 (16%) and 7 of 44 (16%) patients, respectively. The detection rate improved from 18% at baseline and 16% after one cycle with EpCAM to 25% (P = 0.08) and 30% (P = 0.02), respectively, with EpCAM/MCAM. No patients with MCAM-positive CTCs versus 23% of patients without MCAM-positive CTCs at baseline achieved pCR (P = 0.13). EpCAM-positive CTCs and CEC counts were not correlated to pCR. Combined EpCAM/MCAM CellSearch enrichment thus increased the CTC detection rate in stage II/III pBC. We found no associations of CTC and CEC counts with pCR to NAC. The clinical relevance of MCAM-positive CTCs deserves further study.
Collapse
Affiliation(s)
- Wendy Onstenk
- Department of Medical Oncology and Cancer Genomics Netherlands, Erasmus MC Cancer Institute, Erasmus University Medical Center, Rotterdam, the Netherlands.
| | - Jaco Kraan
- Department of Medical Oncology and Cancer Genomics Netherlands, Erasmus MC Cancer Institute, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - Bianca Mostert
- Department of Medical Oncology and Cancer Genomics Netherlands, Erasmus MC Cancer Institute, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - Mieke M Timmermans
- Department of Medical Oncology and Cancer Genomics Netherlands, Erasmus MC Cancer Institute, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - Ayoub Charehbili
- Department of Clinical Oncology, Leiden University Medical Center, Leiden, the Netherlands. Department of Surgery, Leiden University Medical Center, Leiden, the Netherlands
| | - Vincent T H B M Smit
- Department of Pathology, Leiden University Medical Center, Leiden, the Netherlands
| | - Judith R Kroep
- Department of Clinical Oncology, Leiden University Medical Center, Leiden, the Netherlands
| | - Johan W R Nortier
- Department of Clinical Oncology, Leiden University Medical Center, Leiden, the Netherlands
| | - Saskia van de Ven
- Department of Clinical Oncology, Leiden University Medical Center, Leiden, the Netherlands
| | - Joan B Heijns
- Department of Medical Oncology, Amphia Hospital, Breda, the Netherlands
| | - Lonneke W Kessels
- Department of Internal Medicine, Deventer Hospital, Deventer, the Netherlands
| | - Hanneke W M van Laarhoven
- Department of Medical Oncology, Academic Medical Center, University of Amsterdam, Amsterdam, the Netherlands
| | - Monique M E M Bos
- Department of Internal Medicine, Reinier de Graaf Hospital, Delft, the Netherlands
| | | | - Jan W Gratama
- Department of Medical Oncology and Cancer Genomics Netherlands, Erasmus MC Cancer Institute, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - Anieta M Sieuwerts
- Department of Medical Oncology and Cancer Genomics Netherlands, Erasmus MC Cancer Institute, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - John W M Martens
- Department of Medical Oncology and Cancer Genomics Netherlands, Erasmus MC Cancer Institute, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - John A Foekens
- Department of Medical Oncology and Cancer Genomics Netherlands, Erasmus MC Cancer Institute, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - Stefan Sleijfer
- Department of Medical Oncology and Cancer Genomics Netherlands, Erasmus MC Cancer Institute, Erasmus University Medical Center, Rotterdam, the Netherlands
| |
Collapse
|
49
|
Pestrin M, Salvianti F, Galardi F, De Luca F, Turner N, Malorni L, Pazzagli M, Di Leo A, Pinzani P. Heterogeneity of PIK3CA mutational status at the single cell level in circulating tumor cells from metastatic breast cancer patients. Mol Oncol 2014; 9:749-57. [PMID: 25539732 DOI: 10.1016/j.molonc.2014.12.001] [Citation(s) in RCA: 134] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2014] [Revised: 10/30/2014] [Accepted: 12/01/2014] [Indexed: 01/18/2023] Open
Abstract
Circulating Tumor Cells (CTCs) represent a "liquid biopsy of the tumor" which might allow real-time monitoring of cancer biology and therapies in individual patients. CTCs are extremely rare in the blood stream and their analysis is technically challenging. The CellSearch(®) system provides the enumeration of CTCs with prognostic significance in patients with metastatic breast cancer (mBC), but it does not allow their molecular characterization, which might be useful to identify therapeutically relevant targets for individualized treatment. Combining the CellSearch(®) and DEPArray™ technologies allows the recovery of single CTCs as a pure sample for molecular analysis. The purpose of the study was to investigate the heterogeneity of PIK3CA mutational status within single CTCs isolated from individual mBC patients. CTCs were enriched and enumerated by CellSearch(®) in blood samples collected from 39 mBC patients. In 20 out of 39 patients enriched samples with ≥5 CTCs were sorted using DEParray™ to isolate single CTCs or pools of CTCs to be submitted to Whole Genome Amplification (WGA) before sequencing analysis. In 18 out of 20 patients, it was possible to perform PIK3CA sequencing on exons 9 and 20. Twelve subjects were wild type (wt) for the PIK3CA gene. PIK3CA status could also be assessed in pools of CTCs in seven of these patients, with consistent wt status found. Six patients (33%) had a PIK3CA mutation identified. In 2 of the six patients, molecular heterogeneity was detected when mutational analysis was performed on more than one single CTC, including one patient with loss of heterozygosity on both single and pooled CTCs, and one patient with three different PIK3CA variants on single CTCs but PIK3CA wt status on pooled CTC samples. In six out of the 18 cases PIK3CA status was also evaluable on a primary tumor sample. In one of the six cases a discordance in PIK3CA status between the primary (wild-type) and the matched CTC (exon 20 mutation) was observed. This study demonstrates the feasibility of a non-invasive approach based on the liquid biopsy in mBC patients. Moreover, our data suggest the importance of characterizing CTCs at the single cell level in order to investigate the molecular heterogeneity within cells from the same patient.
Collapse
Affiliation(s)
- Marta Pestrin
- Sandro Pitigliani Medical Oncology Department, Hospital of Prato, Istituto Toscano Tumori, Prato, Italy
| | - Francesca Salvianti
- Department of Clinical, Experimental and Biomedical Sciences, University of Florence, Florence, Italy
| | - Francesca Galardi
- Sandro Pitigliani Medical Oncology Department, Hospital of Prato, Istituto Toscano Tumori, Prato, Italy
| | - Francesca De Luca
- Sandro Pitigliani Medical Oncology Department, Hospital of Prato, Istituto Toscano Tumori, Prato, Italy
| | - Natalie Turner
- Sandro Pitigliani Medical Oncology Department, Hospital of Prato, Istituto Toscano Tumori, Prato, Italy
| | - Luca Malorni
- Sandro Pitigliani Medical Oncology Department, Hospital of Prato, Istituto Toscano Tumori, Prato, Italy
| | - Mario Pazzagli
- Department of Clinical, Experimental and Biomedical Sciences, University of Florence, Florence, Italy
| | - Angelo Di Leo
- Sandro Pitigliani Medical Oncology Department, Hospital of Prato, Istituto Toscano Tumori, Prato, Italy
| | - Pamela Pinzani
- Department of Clinical, Experimental and Biomedical Sciences, University of Florence, Florence, Italy.
| |
Collapse
|
50
|
Toss A, Mu Z, Fernandez S, Cristofanilli M. CTC enumeration and characterization: moving toward personalized medicine. ANNALS OF TRANSLATIONAL MEDICINE 2014; 2:108. [PMID: 25489582 DOI: 10.3978/j.issn.2305-5839.2014.09.06] [Citation(s) in RCA: 66] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 06/20/2014] [Accepted: 08/28/2014] [Indexed: 12/27/2022]
Abstract
The primary cause of tumor-related death in breast cancer (BC) is still represented by distant metastasization. The dissemination of tumor cells from the primary tumor to distant sites through bloodstream cannot be early detected by standard imaging methods. The enumeration of circulating tumor cells (CTCs) represents an effective prognostic and predictive biomarker, which is able to monitor efficacy of adjuvant therapies, detect early development of (micro)metastases and at last, assess therapeutic responses of advanced disease earlier than traditional imaging methods. Moreover, since repeated tissue biopsies are invasive, costly and not always feasible, the assessment of tumor characteristics on CTCs, by a peripheral blood sample as a 'liquid biopsy', represents an attractive opportunity. The implementation of molecular and genomic characterization of CTCs could contribute to improve the treatment selection and thus, to move toward more personalized treatments. This review describes the current state of the art on CTC detection strategies, the evidence to demonstrate their clinical validity, and their potential impact for both future clinical trial design and, decision-making process in our daily practice.
Collapse
Affiliation(s)
- Angela Toss
- 1 Department of Oncology, Haematology and Respiratory Diseases, University of Modena and Reggio Emilia, Modena, Italy ; 2 Department of Medical Oncology, Thomas Jefferson University & Kimmel Cancer Center, Philadelphia, PA, USA
| | - Zhaomei Mu
- 1 Department of Oncology, Haematology and Respiratory Diseases, University of Modena and Reggio Emilia, Modena, Italy ; 2 Department of Medical Oncology, Thomas Jefferson University & Kimmel Cancer Center, Philadelphia, PA, USA
| | - Sandra Fernandez
- 1 Department of Oncology, Haematology and Respiratory Diseases, University of Modena and Reggio Emilia, Modena, Italy ; 2 Department of Medical Oncology, Thomas Jefferson University & Kimmel Cancer Center, Philadelphia, PA, USA
| | - Massimo Cristofanilli
- 1 Department of Oncology, Haematology and Respiratory Diseases, University of Modena and Reggio Emilia, Modena, Italy ; 2 Department of Medical Oncology, Thomas Jefferson University & Kimmel Cancer Center, Philadelphia, PA, USA
| |
Collapse
|