1
|
Rusnáková DŠ, Aziri R, Dubovan P, Jurík M, Mego M, Pinďák D. Detection, significance and potential utility of circulating tumor cells in clinical practice in breast cancer (Review). Oncol Lett 2025; 29:10. [PMID: 39492933 PMCID: PMC11526295 DOI: 10.3892/ol.2024.14756] [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: 04/09/2024] [Accepted: 08/15/2024] [Indexed: 11/05/2024] Open
Abstract
Although advances in diagnostic techniques, new therapeutic strategies and personalization of breast cancer (BC) care have improved the survival for a number of patients, BC remains a major cause of morbidity and mortality for women. The study of circulating tumor cells (CTCs) has significant potential in translational oncology since these cells represent promising biomarkers throughout the entire course of BC in patients. CTCs also have notable prognostic value in early BC as well as metastatic BC. Based on current knowledge, it seems that the dynamics of CTCs that change during therapy reflect therapy response, and CTCs could serve as a tool for risk stratification and real-time monitoring of treatment in patients with BC. The question of how to use this information in everyday clinical practice and how this information can guide or change therapy to affect the clinical outcome of patients with BC remains unanswered. The present review aims to discuss current completed and ongoing trials that have been designed to demonstrate the clinical significance of CTCs, offer insights into treatment efficacy and assess CTC utility, facilitating their implementation in the routine management of patients with BC.
Collapse
Affiliation(s)
- Dominika Šmičková Rusnáková
- Department of Surgical Oncology, Faculty of Medicine, Slovak Medical University and National Cancer Institute, 833 10 Bratislava, Slovak Republic
| | - Ramadan Aziri
- Department of Surgical Oncology, Faculty of Medicine, Slovak Medical University and National Cancer Institute, 833 10 Bratislava, Slovak Republic
| | - Peter Dubovan
- Department of Surgical Oncology, Faculty of Medicine, Slovak Medical University and National Cancer Institute, 833 10 Bratislava, Slovak Republic
| | - Miroslav Jurík
- Department of Surgical Oncology, Faculty of Medicine, Slovak Medical University and National Cancer Institute, 833 10 Bratislava, Slovak Republic
| | - Michal Mego
- Second Department of Oncology, Faculty of Medicine, Comenius University and National Cancer Institute, 833 10 Bratislava, Slovak Republic
| | - Daniel Pinďák
- Department of Surgical Oncology, Faculty of Medicine, Slovak Medical University and National Cancer Institute, 833 10 Bratislava, Slovak Republic
| |
Collapse
|
2
|
Mazzeo R, Sears J, Palmero L, Bolzonello S, Davis AA, Gerratana L, Puglisi F. Liquid biopsy in triple-negative breast cancer: unlocking the potential of precision oncology. ESMO Open 2024; 9:103700. [PMID: 39288656 PMCID: PMC11421323 DOI: 10.1016/j.esmoop.2024.103700] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2024] [Revised: 06/29/2024] [Accepted: 08/06/2024] [Indexed: 09/19/2024] Open
Abstract
In the era of precision oncology, the management of triple-negative breast cancer (TNBC) is rapidly changing and becoming more complicated with a variety of chemotherapy, immunotherapy, and targeted treatment options. Currently, TNBC treatment is based on prognostic and predictive factors including immunohistochemical biomarkers [e.g. programmed death-ligand 1 (PD-L1)] and germline BRCA mutations. Given the current limitation of existing biomarkers, liquid biopsies may serve as clinically useful tools to determine treatment efficacy and response in both the (neo)adjuvant and metastatic settings, for detecting early relapse, and for monitoring clonal evolution during treatment. In this review, we comprehensively summarize current and future liquid biopsy applications. Specifically, we highlight the role of circulating tumor cell characterization, circulating tumor DNA, and other preclinical liquid biopsy technologies including circulating exosomes, RNA liquid biopsy, and circulating immune-based biomarkers. In the near future, these biomarkers may serve to identify early disease relapse, therapeutic targets, and disease clonality for patients with TNBC in the clinical setting.
Collapse
Affiliation(s)
- R Mazzeo
- Department of Medical Oncology, CRO Aviano, National Cancer Institute, IRCCS, Aviano; Department of Medicine, University of Udine, Udine, Italy
| | - J Sears
- Department of Medicine, Washington University in St. Louis, St. Louis
| | - L Palmero
- Department of Medical Oncology, CRO Aviano, National Cancer Institute, IRCCS, Aviano; Department of Medicine, University of Udine, Udine, Italy
| | - S Bolzonello
- Department of Medical Oncology, CRO Aviano, National Cancer Institute, IRCCS, Aviano
| | - A A Davis
- Department of Medicine, Division of Oncology, Washington University School of Medicine, St. Louis, USA
| | - L Gerratana
- Department of Medical Oncology, CRO Aviano, National Cancer Institute, IRCCS, Aviano; Department of Medicine, University of Udine, Udine, Italy.
| | - F Puglisi
- Department of Medical Oncology, CRO Aviano, National Cancer Institute, IRCCS, Aviano; Department of Medicine, University of Udine, Udine, Italy
| |
Collapse
|
3
|
Gu X, Wei S, Lv X. Circulating tumor cells: from new biological insights to clinical practice. Signal Transduct Target Ther 2024; 9:226. [PMID: 39218931 PMCID: PMC11366768 DOI: 10.1038/s41392-024-01938-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2023] [Revised: 05/31/2024] [Accepted: 07/29/2024] [Indexed: 09/04/2024] Open
Abstract
The primary reason for high mortality rates among cancer patients is metastasis, where tumor cells migrate through the bloodstream from the original site to other parts of the body. Recent advancements in technology have significantly enhanced our comprehension of the mechanisms behind the bloodborne spread of circulating tumor cells (CTCs). One critical process, DNA methylation, regulates gene expression and chromosome stability, thus maintaining dynamic equilibrium in the body. Global hypomethylation and locus-specific hypermethylation are examples of changes in DNA methylation patterns that are pivotal to carcinogenesis. This comprehensive review first provides an overview of the various processes that contribute to the formation of CTCs, including epithelial-mesenchymal transition (EMT), immune surveillance, and colonization. We then conduct an in-depth analysis of how modifications in DNA methylation within CTCs impact each of these critical stages during CTC dissemination. Furthermore, we explored potential clinical implications of changes in DNA methylation in CTCs for patients with cancer. By understanding these epigenetic modifications, we can gain insights into the metastatic process and identify new biomarkers for early detection, prognosis, and targeted therapies. This review aims to bridge the gap between basic research and clinical application, highlighting the significance of DNA methylation in the context of cancer metastasis and offering new avenues for improving patient outcomes.
Collapse
Affiliation(s)
- Xuyu Gu
- Department of Oncology, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Shiyou Wei
- Department of Anesthesiology, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Xin Lv
- Department of Anesthesiology, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, China.
| |
Collapse
|
4
|
Wang ZD, Feng YF, Wang YS, Ma Y, Liu J, Li D, Li S, Zhang GD. Peripheral arterial rather than venous blood is a better source of circulating tumor cells in early lung cancer. Thorac Cancer 2024; 15:654-660. [PMID: 38297462 DOI: 10.1111/1759-7714.15236] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2023] [Revised: 12/26/2023] [Accepted: 01/20/2024] [Indexed: 02/02/2024] Open
Abstract
BACKGROUND Circulating tumor cells (CTCs) play a crucial role in the early diagnosis and prognosis of lung cancer. Identification of a more suitable sample source could be a breakthrough towards enhancing CTC detectability in early-stage lung cancer. We investigated the differences in detectable CTCs between peripheral arterial and venous blood in early- and mid-stage lung cancer patients undergoing surgery and analyzed the association between clinicopathological factors and detectable CTCs in peripheral arterial and venous blood. METHODS Peripheral arterial and venous blood was collected in 5-mL samples from 56 patients with surgically resected and pathologically clear at early- or mid-stage lung cancer. Blood specimens were enriched for CTCs based on isolation by size of epithelial tumor cells. The CTCs were identified using Swiss Giemsa staining and immunohistochemistry for CD45/CD31. RESULTS In stage I lung cancer, CTC-positive rate was significantly higher in peripheral arterial than in venous blood (45.45% vs. 17.39%). There was no significant difference in the number of detectable CTCs between peripheral arterial and venous blood. A low degree of differentiation was associated with a high positive rate of CTCs in peripheral venous blood. The number of circulating tumor microemboli was significantly higher in patients with tumor size >3 cm compared with ≤3 cm. CONCLUSION CTC levels in peripheral arterial and venous blood differed little in lung cancer patients.Compared to peripheral venous blood, peripheral arterial blood had a higher CTC positivity rate in early-stage lung cancer.This study was favorable for early detection and monitoring of lung cancer.
Collapse
Affiliation(s)
- Zhen-Dan Wang
- Department of Thoracic Surgery, Shandong University Cancer Center, Jinan, China
- Department of Thoracic Surgery, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, China
| | - Yi-Fei Feng
- School of Clinical and Basic Medicine, Shandong First Medical University, Jinan, China
| | - Yu-Shuo Wang
- Department of Primary Care and Population Health, Institute of Epidemiology and Public Health, London's Global University, London, UK
| | - Ying Ma
- Shandong Pharmaceutical Research Institute, Jinan, China
| | - Jiyan Liu
- Shandong Pharmaceutical Research Institute, Jinan, China
| | - Dihua Li
- Shandong Pharmaceutical Research Institute, Jinan, China
| | - Sheng Li
- Shandong Pharmaceutical Research Institute, Jinan, China
- Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, China
| | - Guo-Dong Zhang
- Department of Thoracic Surgery, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, China
| |
Collapse
|
5
|
Diaz M, Chudsky S, Pentsova E, Miller AM. Clinical applications of cerebrospinal fluid liquid biopsies in central nervous system tumors. Transl Oncol 2024; 41:101881. [PMID: 38218027 PMCID: PMC10825768 DOI: 10.1016/j.tranon.2024.101881] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2022] [Revised: 11/28/2023] [Accepted: 01/03/2024] [Indexed: 01/15/2024] Open
Abstract
For patients with central nervous system (CNS) malignancies, liquid biopsies of the cerebrospinal fluid (CSF) may offer an unparalleled source of information about the tumor, with much less risk than traditional biopsies. Two techniques have been adapted to CSF in clinical settings: circulating tumor cells (CTCs) and circulating tumor DNA (ctDNA). CTCs have been employed mostly as a diagnostic tool for leptomeningeal metastases in epithelial tumors, although they may also have value in the prognostication and monitoring of this disease. The ctDNA technology has been studied in a variety of primary and metastatic brain and spinal cord tumors, where it can be used for diagnosis and molecular classification, with some work suggesting that it may also be useful for longitudinal tracking of tumor evolution or as a marker of residual disease. This review summarizes recent publications on the use of these two tests in CSF, focusing on their established and potential clinical applications.
Collapse
Affiliation(s)
- Maria Diaz
- Department of Neurology, Division of Neuro-Oncology, Columbia University Irving Medical Center, New York, NY, USA
| | - Sofia Chudsky
- Office of Professional Development, Memorial Sloan Kettering Cancer Center, New York, NY, USA; Hunter College, New York, NY, USA
| | - Elena Pentsova
- Department of Neurology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Alexandra M Miller
- Department of Neurology, Memorial Sloan Kettering Cancer Center, New York, NY, USA.
| |
Collapse
|
6
|
Goldberg M, Mondragon-Soto MG, Altawalbeh G, Meyer B, Aftahy AK. New Breakthroughs in the Diagnosis of Leptomeningeal Carcinomatosis: A Review of Liquid Biopsies of Cerebrospinal Fluid. Cureus 2024; 16:e55187. [PMID: 38558729 PMCID: PMC10980855 DOI: 10.7759/cureus.55187] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/28/2024] [Indexed: 04/04/2024] Open
Abstract
Leptomeningeal carcinomatosis represents a terminal stage and is a devastating complication of cancer. Despite its high incidence, current diagnostic methods fail to accurately detect this condition in a timely manner. This failure to diagnose leads to the refusal of treatment and the absence of clinical trials, hampering the development of new therapy strategies. The use of liquid biopsy is revolutionizing the field of diagnostic oncology. The dynamic and non-invasive detection of tumor markers has enormous potential in cancer diagnostics and treatment. Leptomeningeal carcinomatosis is a condition where invasive tissue biopsy is not part of the routine diagnostic analysis, making liquid biopsy an essential diagnostic tool. Several elements in cerebrospinal fluid (CSF) have been investigated as potential targets of liquid biopsy, including free circulating tumor cells, free circulating nucleic acids, proteins, exosomes, and even non-tumor cells as part of the dynamic tumor microenvironment. This review aims to summarize current breakthroughs in the research on liquid biopsy, including the latest breakthroughs in the identification of tumor cells and nucleic acids, and give an overview of future directions in the diagnosis of leptomeningeal carcinomatosis.
Collapse
Affiliation(s)
- Maria Goldberg
- Department of Neurosurgery, School of Medicine, Klinikum Rechts der Isar, Technical University of Munich, Munich, DEU
| | | | - Ghaith Altawalbeh
- Department of Neurosurgery, School of Medicine, Klinikum Rechts der Isar, Technical University of Munich, Munich, DEU
| | - Bernhard Meyer
- Department of Neurosurgery, School of Medicine, Klinikum Rechts der Isar, Technical University of Munich, Munich, DEU
| | - Amir Kaywan Aftahy
- Department of Neurosurgery, School of Medicine, Klinikum Rechts der Isar, Technical University of Munich, Munich, DEU
| |
Collapse
|
7
|
Wang X, Wang L, Lin H, Zhu Y, Huang D, Lai M, Xi X, Huang J, Zhang W, Zhong T. Research progress of CTC, ctDNA, and EVs in cancer liquid biopsy. Front Oncol 2024; 14:1303335. [PMID: 38333685 PMCID: PMC10850354 DOI: 10.3389/fonc.2024.1303335] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2023] [Accepted: 01/04/2024] [Indexed: 02/10/2024] Open
Abstract
Circulating tumor cells (CTCs), circulating tumor DNA (ctDNA), and extracellular vehicles (EVs) have received significant attention in recent times as emerging biomarkers and subjects of transformational studies. The three main branches of liquid biopsy have evolved from the three primary tumor liquid biopsy detection targets-CTC, ctDNA, and EVs-each with distinct benefits. CTCs are derived from circulating cancer cells from the original tumor or metastases and may display global features of the tumor. ctDNA has been extensively analyzed and has been used to aid in the diagnosis, treatment, and prognosis of neoplastic diseases. EVs contain tumor-derived material such as DNA, RNA, proteins, lipids, sugar structures, and metabolites. The three provide different detection contents but have strong complementarity to a certain extent. Even though they have already been employed in several clinical trials, the clinical utility of three biomarkers is still being studied, with promising initial findings. This review thoroughly overviews established and emerging technologies for the isolation, characterization, and content detection of CTC, ctDNA, and EVs. Also discussed were the most recent developments in the study of potential liquid biopsy biomarkers for cancer diagnosis, therapeutic monitoring, and prognosis prediction. These included CTC, ctDNA, and EVs. Finally, the potential and challenges of employing liquid biopsy based on CTC, ctDNA, and EVs for precision medicine were evaluated.
Collapse
Affiliation(s)
- Xiaoling Wang
- Laboratory Medicine, First Affiliated Hospital of Gannan Medical University, Ganzhou, China
- The First School of Clinical Medicine, Gannan Medical University, Ganzhou, China
| | - Lijuan Wang
- Laboratory Medicine, First Affiliated Hospital of Gannan Medical University, Ganzhou, China
- The First School of Clinical Medicine, Gannan Medical University, Ganzhou, China
| | - Haihong Lin
- Laboratory Medicine, First Affiliated Hospital of Gannan Medical University, Ganzhou, China
- The First School of Clinical Medicine, Gannan Medical University, Ganzhou, China
| | - Yifan Zhu
- Laboratory Medicine, First Affiliated Hospital of Gannan Medical University, Ganzhou, China
- The First School of Clinical Medicine, Gannan Medical University, Ganzhou, China
| | - Defa Huang
- Laboratory Medicine, First Affiliated Hospital of Gannan Medical University, Ganzhou, China
| | - Mi Lai
- Laboratory Medicine, First Affiliated Hospital of Gannan Medical University, Ganzhou, China
| | - Xuxiang Xi
- Laboratory Medicine, First Affiliated Hospital of Gannan Medical University, Ganzhou, China
| | - Junyun Huang
- Laboratory Medicine, First Affiliated Hospital of Gannan Medical University, Ganzhou, China
- The First School of Clinical Medicine, Gannan Medical University, Ganzhou, China
| | - Wenjuan Zhang
- Laboratory Medicine, First Affiliated Hospital of Gannan Medical University, Ganzhou, China
- The First School of Clinical Medicine, Gannan Medical University, Ganzhou, China
| | - Tianyu Zhong
- Laboratory Medicine, First Affiliated Hospital of Gannan Medical University, Ganzhou, China
- The First School of Clinical Medicine, Gannan Medical University, Ganzhou, China
| |
Collapse
|
8
|
Cani AK, Hayes DF. Breast Cancer Circulating Tumor Cells: Current Clinical Applications and Future Prospects. Clin Chem 2024; 70:68-80. [PMID: 38175590 DOI: 10.1093/clinchem/hvad191] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2023] [Accepted: 10/19/2023] [Indexed: 01/05/2024]
Abstract
BACKGROUND Identification and characterization of circulating tumor markers, designated as "liquid biopsies," have greatly impacted the care of cancer patients. Although more recently referring to circulating tumor DNA (ctDNA), the term liquid biopsy initially was coined to refer to any blood-borne biomarker related to malignancy, including circulating tumor cells (CTCs) in blood. In this manuscript, we review the specific state of the art of CTCs in breast cancer. CONTENT Liquid biopsies might play a clinical role across the entire spectrum of breast cancer, from risk assessment, prevention, screening, and treatment. CTC counts have been shown to carry clear, independent prognostic information in the latter situation. However, the clinical utility of CTCs in breast cancer remains to be determined. Nonetheless, in addition to CTC enumeration, analyses of CTCs provide tumor molecular information representing the entire, often-heterogeneous disease, relatively noninvasively and longitudinally. Technological advances have allowed the interrogation of CTC-derived information, providing renewed hope for a clinical role in disease monitoring and precision oncology. SUMMARY This narrative review examines CTCs, their clinical validity, and current prospects of clinical utility in breast cancer with the goal of improving patient outcomes.
Collapse
Affiliation(s)
- Andi K Cani
- Division of Hematology and Oncology, Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, MI, United States
- Rogel Cancer Center, University of Michigan, Ann Arbor, MI, United States
| | - Daniel F Hayes
- Division of Hematology and Oncology, Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, MI, United States
- Rogel Cancer Center, University of Michigan, Ann Arbor, MI, United States
| |
Collapse
|
9
|
Spiliopoulou P, Holanda Lopes CD, Spreafico A. Promising and Minimally Invasive Biomarkers: Targeting Melanoma. Cells 2023; 13:19. [PMID: 38201222 PMCID: PMC10777980 DOI: 10.3390/cells13010019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2023] [Revised: 11/29/2023] [Accepted: 12/17/2023] [Indexed: 01/12/2024] Open
Abstract
The therapeutic landscape of malignant melanoma has been radically reformed in recent years, with novel treatments emerging in both the field of cancer immunotherapy and signalling pathway inhibition. Large-scale tumour genomic characterization has accurately classified malignant melanoma into four different genomic subtypes so far. Despite this, only somatic mutations in BRAF oncogene, as assessed in tumour biopsies, has so far become a validated predictive biomarker of treatment with small molecule inhibitors. The biology of tumour evolution and heterogeneity has uncovered the current limitations associated with decoding genomic drivers based only on a single-site tumour biopsy. There is an urgent need to develop minimally invasive biomarkers that accurately reflect the real-time evolution of melanoma and that allow for streamlined collection, analysis, and interpretation. These will enable us to face challenges with tumour tissue attainment and process and will fulfil the vision of utilizing "liquid biopsy" to guide clinical decisions, in a manner akin to how it is used in the management of haematological malignancies. In this review, we will summarize the most recent published evidence on the role of minimally invasive biomarkers in melanoma, commenting on their future potential to lead to practice-changing discoveries.
Collapse
Affiliation(s)
- Pavlina Spiliopoulou
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON M5G 2C1, Canada;
- School of Cancer Sciences, University of Glasgow, Glasgow G61 1BD, UK
| | | | - Anna Spreafico
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON M5G 2C1, Canada;
| |
Collapse
|
10
|
Ma Y, Gan J, Bai Y, Cao D, Jiao Y. Minimal residual disease in solid tumors: an overview. Front Med 2023; 17:649-674. [PMID: 37707677 DOI: 10.1007/s11684-023-1018-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2023] [Accepted: 06/24/2023] [Indexed: 09/15/2023]
Abstract
Minimal residual disease (MRD) is termed as the small numbers of remnant tumor cells in a subset of patients with tumors. Liquid biopsy is increasingly used for the detection of MRD, illustrating the potential of MRD detection to provide more accurate management for cancer patients. As new techniques and algorithms have enhanced the performance of MRD detection, the approach is becoming more widely and routinely used to predict the prognosis and monitor the relapse of cancer patients. In fact, MRD detection has been shown to achieve better performance than imaging methods. On this basis, rigorous investigation of MRD detection as an integral method for guiding clinical treatment has made important advances. This review summarizes the development of MRD biomarkers, techniques, and strategies for the detection of cancer, and emphasizes the application of MRD detection in solid tumors, particularly for the guidance of clinical treatment.
Collapse
Affiliation(s)
- Yarui Ma
- State Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Jingbo Gan
- Genetron Health (Beijing) Co. Ltd., Beijing, 102206, China
| | - Yinlei Bai
- Genetron Health (Beijing) Co. Ltd., Beijing, 102206, China
| | - Dandan Cao
- Genetron Health (Beijing) Co. Ltd., Beijing, 102206, China
| | - Yuchen Jiao
- State Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China.
| |
Collapse
|
11
|
Kamal M, Wang YJ, Plummer S, Dickerson A, Yu M. An Image-Based Identification of Aggressive Breast Cancer Circulating Tumor Cell Subtypes. Cancers (Basel) 2023; 15:2669. [PMID: 37345005 DOI: 10.3390/cancers15102669] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2023] [Revised: 04/30/2023] [Accepted: 05/05/2023] [Indexed: 06/23/2023] Open
Abstract
Using previously established CTC lines from breast cancer patients, we identified different morphometric subgroups of CTCs with one of them having the highest tumorigenic potential in vivo despite the slowest cell proliferation in vitro. This subgroup represents 32% of all cells and contains cells with small cell volume, large nucleus to cell, dense nuclear areas to the nucleus, mitochondria to cell volume ratios and rough texture of cell membrane and termed "Small cell, Large mitochondria, Rough membrane" (SLR). RNA-seq analyses showed that the SLR group is enriched in pathways and cellular processes related to DNA replication, DNA repair and metabolism. SLR upregulated genes are associated with poor survival in patients with ER+ breast cancer based on the KM Plotter database. The high tumorigenic potential, slow proliferation, and enriched DNA replication/repair pathways suggest that the SLR subtype is associated with stemness properties. Our new findings provide a simple image-based identification of CTC subpopulations with elevated aggressiveness, which is expected to provide a more accurate prediction of patient survival and therapy response than total CTC numbers. The detection of morphometric and transcriptomic profiles related to the SLR subgroup of CTCs also opens opportunities for potential targeted cancer treatment.
Collapse
Affiliation(s)
- Mohamed Kamal
- Department of Stem Cell Biology and Regenerative Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA
- USC Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA
- Department of Zoology, Faculty of Science, University of Benha, Benha 13518, Egypt
| | - Yiru Jess Wang
- Department of Stem Cell Biology and Regenerative Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA
- USC Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA
- Department of Pharmacology, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Sarai Plummer
- Department of Stem Cell Biology and Regenerative Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA
- USC Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA
| | - Amber Dickerson
- Department of Stem Cell Biology and Regenerative Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA
- USC Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA
| | - Min Yu
- Department of Stem Cell Biology and Regenerative Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA
- USC Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA
- Department of Pharmacology, University of Maryland School of Medicine, Baltimore, MD 21201, USA
- Marlene and Stewart Greenebaum NCI Comprehensive Cancer Center, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| |
Collapse
|
12
|
Ring A, Nguyen-Sträuli BD, Wicki A, Aceto N. Biology, vulnerabilities and clinical applications of circulating tumour cells. Nat Rev Cancer 2023; 23:95-111. [PMID: 36494603 PMCID: PMC9734934 DOI: 10.1038/s41568-022-00536-4] [Citation(s) in RCA: 102] [Impact Index Per Article: 102.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 11/07/2022] [Indexed: 12/13/2022]
Abstract
In recent years, exceptional technological advances have enabled the identification and interrogation of rare circulating tumour cells (CTCs) from blood samples of patients, leading to new fields of research and fostering the promise for paradigm-changing, liquid biopsy-based clinical applications. Analysis of CTCs has revealed distinct biological phenotypes, including the presence of CTC clusters and the interaction between CTCs and immune or stromal cells, impacting metastasis formation and providing new insights into cancer vulnerabilities. Here we review the progress made in understanding biological features of CTCs and provide insight into exploiting these developments to design future clinical tools for improving the diagnosis and treatment of cancer.
Collapse
Affiliation(s)
- Alexander Ring
- Department of Biology, Institute for Molecular Health Sciences, ETH Zurich, Zurich, Switzerland
- Department of Medical Oncology and Hematology, University Hospital Zurich and University of Zurich, Zurich, Switzerland
| | - Bich Doan Nguyen-Sträuli
- Department of Biology, Institute for Molecular Health Sciences, ETH Zurich, Zurich, Switzerland
- Department of Gynecology, University Hospital Zurich and University of Zurich, Zurich, Switzerland
| | - Andreas Wicki
- Department of Medical Oncology and Hematology, University Hospital Zurich and University of Zurich, Zurich, Switzerland
| | - Nicola Aceto
- Department of Biology, Institute for Molecular Health Sciences, ETH Zurich, Zurich, Switzerland.
| |
Collapse
|
13
|
Mihalcioiu C, Li J, Badescu D, Camirand A, Kremer N, Bertos N, Omeroglu A, Sebag M, Di Battista J, Park M, Ragoussis J, Kremer R. Improved platform for breast cancer circulating tumor cell enrichment and characterization with next-generation sequencing technology. Am J Cancer Res 2023; 13:25-44. [PMID: 36777514 PMCID: PMC9906071] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2022] [Accepted: 09/29/2022] [Indexed: 02/14/2023] Open
Abstract
Circulating tumor cells (CTCs) represent cells shed from the primary tumor or metastatic sites and can be used to monitor treatment response and tumor recurrence. However, CTCs circulate in extremely low numbers making in-depth analysis beyond simple enumeration challenging when collected from peripheral blood. Furthermore, tumor heterogeneity, a hallmark of many tumors, especially breast cancer, further complicates CTC characterization. To overcome this limitation, we developed a platform based on the large-scale isolation of CTCs by apheresis, allowing us to collect CTCs in large numbers, which were preserved live in liquid nitrogen for further characterization. Flow cytometry followed by cell sorting (FACS) was performed using a combination of antibodies directed against cell surface markers of white blood cells (CD45) and epithelial tumor cells (CK8). Analysis of subpopulations CD45+/- and CK8+/- by bulk RNA sequencing (RNAseq) and the CD45-/CK8 positive population by single-cell RNAseq was performed. The CD45- population was enriched using CD45 magnetic beads separation and examined by IHC for pan-cytokeratin and immunofluorescence (IF) for specific markers, including the elusive circulating cancer stem cells (CSCs). CSC-rich mammospheres were grown in vitro for further analysis and treated to examine their response to chemotherapeutic agents. Finally, mammospheres were transplanted into the mammary fat pad and bone of immunodeficient mice to examine tumor growth in vivo. This platform enables the detection and collection of CTCs in early and late-stage breast cancer patients of every subtype. Markers including CD44/24, ALDH1 and CXCR4 were identified by IF and showed high expression following mammosphere culture, which responded predictably to chemotherapeutic agents. Mammospheres were also transplanted into nude mice and induced tumors in the mammary fat pad and bone following intra-tibial transplantation. Finally, bulk RNA analysis of the FACS isolated CD45+/- and CK8+/- cells showed a clear separation of CD45- away from CD45+ populations. Single-cell RNAseq of the FACS isolated CD45-/CK8+ cells showed the presence of 4-5 clusters, confirming the high degree of heterogeneity of CTCs. Our platform for large-scale isolation of CTCs using apheresis is suitable for an in-depth analysis of the cancer phenotype and may eventually allow evaluation in real-time of the disease process to optimize cancer regimens.
Collapse
Affiliation(s)
- Catalin Mihalcioiu
- Department of Medicine, McGill University Health CentreGlen Site, 1001 Boul. Décarie, Mail Drop EM1.3229, Montréal, Québec, H4A 3J1, Canada
| | - Jiarong Li
- Department of Medicine, McGill University Health CentreGlen Site, 1001 Boul. Décarie, Mail Drop EM1.3229, Montréal, Québec, H4A 3J1, Canada
| | - Dunarel Badescu
- McGill University Génome Centre, Department of Human GeneticsMontréal, Québec, Canada
| | - Anne Camirand
- Department of Medicine, McGill University Health CentreGlen Site, 1001 Boul. Décarie, Mail Drop EM1.3229, Montréal, Québec, H4A 3J1, Canada
| | - Nathaniel Kremer
- Department of Medicine, McGill University Health CentreGlen Site, 1001 Boul. Décarie, Mail Drop EM1.3229, Montréal, Québec, H4A 3J1, Canada
| | - Nicholas Bertos
- Department of Medicine, McGill University Health CentreGlen Site, 1001 Boul. Décarie, Mail Drop EM1.3229, Montréal, Québec, H4A 3J1, Canada
| | - Atilla Omeroglu
- Department of Medicine, McGill University Health CentreGlen Site, 1001 Boul. Décarie, Mail Drop EM1.3229, Montréal, Québec, H4A 3J1, Canada
| | - Michael Sebag
- Department of Medicine, McGill University Health CentreGlen Site, 1001 Boul. Décarie, Mail Drop EM1.3229, Montréal, Québec, H4A 3J1, Canada
| | - John Di Battista
- Department of Medicine, McGill University Health CentreGlen Site, 1001 Boul. Décarie, Mail Drop EM1.3229, Montréal, Québec, H4A 3J1, Canada
| | - Morag Park
- Breast Cancer Functional Genomics Group, Rosalind and Morris Goodman Cancer Centre, McGill UniversityMontréal, Québec, Canada
| | - Jiannis Ragoussis
- McGill University Génome Centre, Department of Human GeneticsMontréal, Québec, Canada
| | - Richard Kremer
- Department of Medicine, McGill University Health CentreGlen Site, 1001 Boul. Décarie, Mail Drop EM1.3229, Montréal, Québec, H4A 3J1, Canada
| |
Collapse
|
14
|
Cohen EN, Jayachandran G, Moore RG, Cristofanilli M, Lang JE, Khoury JD, Press MF, Kim KK, Khazan N, Zhang Q, Zhang Y, Kaur P, Guzman R, Miller MC, Reuben JM, Ueno NT. A Multi-Center Clinical Study to Harvest and Characterize Circulating Tumor Cells from Patients with Metastatic Breast Cancer Using the Parsortix ® PC1 System. Cancers (Basel) 2022; 14:5238. [PMID: 36358657 PMCID: PMC9656921 DOI: 10.3390/cancers14215238] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2022] [Revised: 10/18/2022] [Accepted: 10/20/2022] [Indexed: 08/22/2023] Open
Abstract
Circulating tumor cells (CTCs) captured from the blood of cancer patients may serve as a surrogate source of tumor material that can be obtained via a venipuncture (also known as a liquid biopsy) and used to better understand tumor characteristics. However, the only FDA-cleared CTC assay has been limited to the enumeration of surface marker-defined cells and not further characterization of the CTCs. In this study, we tested the ability of a semi-automated device capable of capturing and harvesting CTCs from peripheral blood based on cell size and deformability, agnostic of cell-surface markers (the Parsortix® PC1 System), to yield CTCs for evaluation by downstream techniques commonly available in clinical laboratories. The data generated from this study were used to support a De Novo request (DEN200062) for the classification of this device, which the FDA recently granted. As part of a multicenter clinical trial, peripheral blood samples from 216 patients with metastatic breast cancer (MBC) and 205 healthy volunteers were subjected to CTC enrichment. A board-certified pathologist enumerated the CTCs from each participant by cytologic evaluation of Wright-Giemsa-stained slides. As proof of principle, cells harvested from a concurrent parallel sample provided by each participant were evaluated using one of three additional evaluation techniques: molecular profiling by qRT-PCR, RNA sequencing, or cytogenetic analysis of HER2 amplification by FISH. The study demonstrated that the Parsortix® PC1 System can effectively capture and harvest CTCs from the peripheral blood of MBC patients and that the harvested cells can be evaluated using orthogonal methodologies such as gene expression and/or Fluorescence In Situ Hybridization (FISH).
Collapse
Affiliation(s)
- Evan N. Cohen
- Department of Hematopathology Research, Division of Pathology and Laboratory Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Gitanjali Jayachandran
- Department of Hematopathology Research, Division of Pathology and Laboratory Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Richard G. Moore
- Division of Gynecologic Oncology, Department of Obstetrics and Gynecology, Wilmot Cancer Institute, University of Rochester Medical Center, Rochester, NY 14620, USA
| | - Massimo Cristofanilli
- Department of Medicine-Hematology and Oncology, Robert H Lurie Comprehensive Cancer Center, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Julie E. Lang
- USC Breast Cancer Program, Keck School of Medicine, Norris Comprehensive Cancer Center, University of Southern California, Los Angeles, CA 90033, USA
| | - Joseph D. Khoury
- Department of Pathology, Breast Cancer Analysis Laboratory, Keck School of Medicine, Norris Comprehensive Cancer Center, University of Southern California, Los Angeles, CA 90033, USA
| | - Michael F. Press
- Department of Pathology, Breast Cancer Analysis Laboratory, Keck School of Medicine, Norris Comprehensive Cancer Center, University of Southern California, Los Angeles, CA 90033, USA
| | - Kyu Kwang Kim
- Division of Gynecologic Oncology, Department of Obstetrics and Gynecology, Wilmot Cancer Institute, University of Rochester Medical Center, Rochester, NY 14620, USA
| | - Negar Khazan
- Division of Gynecologic Oncology, Department of Obstetrics and Gynecology, Wilmot Cancer Institute, University of Rochester Medical Center, Rochester, NY 14620, USA
| | - Qiang Zhang
- Department of Medicine-Hematology and Oncology, Robert H Lurie Comprehensive Cancer Center, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Youbin Zhang
- Department of Medicine-Hematology and Oncology, Robert H Lurie Comprehensive Cancer Center, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Pushpinder Kaur
- USC Breast Cancer Program, Keck School of Medicine, Norris Comprehensive Cancer Center, University of Southern California, Los Angeles, CA 90033, USA
| | - Roberta Guzman
- Department of Pathology, Breast Cancer Analysis Laboratory, Keck School of Medicine, Norris Comprehensive Cancer Center, University of Southern California, Los Angeles, CA 90033, USA
| | - Michael C. Miller
- ANGLE Clinical Studies, ANGLE Europe Limited, Guildford, Surrey GU2 7AF, UK
| | - James M. Reuben
- Department of Hematopathology Research, Division of Pathology and Laboratory Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Naoto T. Ueno
- Department of Breast Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| |
Collapse
|
15
|
Multianalyte liquid biopsy to aid the diagnostic workup of breast cancer. NPJ Breast Cancer 2022; 8:112. [PMID: 36167819 PMCID: PMC9515081 DOI: 10.1038/s41523-022-00480-4] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2021] [Accepted: 09/16/2022] [Indexed: 11/19/2022] Open
Abstract
Breast cancer (BC) affects 1 in every 8 women in the United States and is currently the most prevalent cancer worldwide. Precise staging at diagnosis and prognosis are essential components for the clinical management of BC patients. In this study, we set out to evaluate the feasibility of the high-definition single cell (HDSCA) liquid biopsy (LBx) platform to stratify late-stage BC, early-stage BC, and normal donors using peripheral blood samples. Utilizing 5 biomarkers, we identified rare circulating events with epithelial, mesenchymal, endothelial and hematological origin. We detected a higher level of CTCs in late-stage patients, compared to the early-stage and normal donors. Additionally, we observed more tumor-associated large extracellular vesicles (LEVs) in the early-stage, compared to late-stage and the normal donor groups. Overall, we were able to detect reproducible patterns in the enumeration of rare cells and LEVs of cancer vs. normal donors and early-stage vs. late-stage BC with high accuracy, allowing for robust stratification. Our findings illustrate the feasibility of the LBx assay to provide robust detection of rare circulating events in peripheral blood draws and to stratify late-stage BC, early-stage BC, and normal donor samples.
Collapse
|
16
|
Pace J, Ivich F, Marple E, Niedre M. Near-infrared diffuse in vivo flow cytometry. JOURNAL OF BIOMEDICAL OPTICS 2022; 27:JBO-220101GR. [PMID: 36114606 PMCID: PMC9478904 DOI: 10.1117/1.jbo.27.9.097002] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/11/2022] [Accepted: 09/07/2022] [Indexed: 06/15/2023]
Abstract
Significance Diffuse in vivo flow cytometry (DiFC) is an emerging technique for enumerating rare fluorescently labeled circulating cells noninvasively in the bloodstream. Thus far, we have reported red and blue-green versions of DiFC. Use of near-infrared (NIR) fluorescent light would in principle allow use of DiFC in deeper tissues and would be compatible with emerging NIR fluorescence molecular contrast agents. Aim We describe the design of a NIR-DiFC instrument and demonstrate its use in optical flow phantoms in vitro and in mice in vivo. Approach We developed an improved optical fiber probe design for efficient collection of fluorescence from individual circulating cells and efficient rejection of instrument autofluorescence. We built a NIR-DiFC instrument. We tested this with NIR fluorescent microspheres and cell lines labeled with OTL38 fluorescence contrast agent in a flow phantom model. We also tested NIR-DiFC in nude mice injected intravenously with OTL38-labeled L1210A cells. Results NIR-DiFC allowed detection of circulating tumor cells (CTCs) in flow phantoms with mean signal-to-noise ratios (SNRs) of 19 to 32 dB. In mice, fluorescently labeled CTCs were detectable with mean SNR of 26 dB. NIR-DiFC also exhibited orders significantly lower autofluorescence and false-alarm rates than blue-green DiFC. Conclusions NIR-DiFC allows use of emerging NIR contrast agents. Our work could pave the way for future use of NIR-DiFC in humans.
Collapse
Affiliation(s)
- Joshua Pace
- Northeastern University, Department of Bioengineering, Boston, Massachusetts, United States
| | - Fernando Ivich
- Northeastern University, Department of Bioengineering, Boston, Massachusetts, United States
| | - Eric Marple
- EmVision LLC, Loxahatchee, Florida, United States
| | - Mark Niedre
- Northeastern University, Department of Bioengineering, Boston, Massachusetts, United States
| |
Collapse
|
17
|
Agnoletto C, Volinia S. Mitochondria dysfunction in circulating tumor cells. Front Oncol 2022; 12:947479. [PMID: 35992829 PMCID: PMC9386562 DOI: 10.3389/fonc.2022.947479] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2022] [Accepted: 07/11/2022] [Indexed: 12/16/2022] Open
Abstract
Circulating tumor cells (CTCs) represent a subset of heterogeneous cells, which, once released from a tumor site, have the potential to give rise to metastasis in secondary sites. Recent research focused on the attempt to detect and characterize these rare cells in the circulation, and advancements in defining their molecular profile have been reported in diverse tumor species, with potential implications for clinical applications. Of note, metabolic alterations, involving mitochondria, have been implicated in the metastatic process, as key determinants in the transition of tumor cells to a mesenchymal or stemness-like phenotype, in drug resistance, and in induction of apoptosis. This review aimed to briefly analyse the most recent knowledge relative to mitochondria dysfunction in CTCs, and to envision implications of altered mitochondria in CTCs for a potential utility in clinics.
Collapse
Affiliation(s)
- Chiara Agnoletto
- Rete Oncologica Veneta (ROV), Veneto Institute of Oncology IOV-IRCCS, Padua, Italy
| | - Stefano Volinia
- Laboratorio per le Tecnologie delle Terapie Avanzate (LTTA), Department of Translational Medicine, University of Ferrara, Ferrara, Italy
- Biological and Chemical Research Centre (CNBCh UW), University of Warsaw, Warsaw, Poland
- Center of New Technologies, University of Warsaw, Warsaw, Poland
| |
Collapse
|
18
|
Tran Thi YV, Hoang BA, Thanh HT, Nguyen TH, Ngoc TP, Thu HB, Hoang NN, Bui TT, Duc TC, Do Quang L. Design and Numerical Study on a Microfluidic System for Circulating Tumor Cells Separation From Whole Blood Using Magnetophoresis and Dielectrophoresis Techniques. Biochem Eng J 2022. [DOI: 10.1016/j.bej.2022.108551] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
|
19
|
Cardinali B, De Luca G, Tasso R, Coco S, Garuti A, Buzzatti G, Sciutto A, Arecco L, Villa F, Carli F, Reverberi D, Quarto R, Dono M, Del Mastro L. Targeting PIK3CA Actionable Mutations in the Circulome: A Proof of Concept in Metastatic Breast Cancer. Int J Mol Sci 2022; 23:ijms23116320. [PMID: 35682999 PMCID: PMC9181240 DOI: 10.3390/ijms23116320] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2022] [Revised: 05/27/2022] [Accepted: 05/30/2022] [Indexed: 11/21/2022] Open
Abstract
The study of circulating cancer-derived components (circulome) is considered the new frontier of liquid biopsy. Despite the recognized role of circulome biomarkers, their comparative molecular profiling is not yet routine. In advanced breast cancer (BC), approximately 40% of hormone-receptor-positive, HER2-negative BC cases harbor druggable PIK3CA mutations suitable for combined alpelisib/fulvestrant treatment. This pilot study investigates PIK3CA mutations in circulating tumor DNA (ctDNA), tumor cells (CTCs), and extracellular vesicles (EVs) with the aim of determining which information on molecular targetable profiling could be recollected in each of them. The in-depth molecular analysis of four BC patients demonstrated, as a proof-of-concept study, that it is possible to retrieve mutational information in the three components. Patient-specific PIK3CA mutations were found in both tissue and ctDNA and in 3/4 cases, as well as in CTCs, in the classical population (large-sized CD45−/EpCAM+/− cells), and/or in the “non-conventional” sub-population (smaller-sized CD44+/EpCAM−/CD45− cells). Consistent mutational profiles of EVs with CTCs suggest that they may have been released by CTCs. This preliminary evidence on the molecular content of the different circulating biomaterials suggests their possible function as a mirror of the intrinsic heterogeneity of BC. Moreover, this study demonstrates, through mutational assessment, the tumor origin of the different CTC sub-populations sustaining the translational value of the circulome for a more comprehensive picture of the disease.
Collapse
Affiliation(s)
- Barbara Cardinali
- Clinica di Oncologia Medica, IRCCS Ospedale Policlinico San Martino, 10-16132 Genova, Italy; (A.S.); (L.A.); (L.D.M.)
- Correspondence: (B.C.); (G.D.L.); Tel.: +39-0105558101 (B.C.); +39-0105558940 (G.D.L.)
| | - Giuseppa De Luca
- Molecular Diagnostic Unit, IRCCS Ospedale Policlinico San Martino, 10-16132 Genova, Italy;
- Correspondence: (B.C.); (G.D.L.); Tel.: +39-0105558101 (B.C.); +39-0105558940 (G.D.L.)
| | - Roberta Tasso
- Department of Experimental Medicine (DIMES), University of Genova, 5-16126 Genova, Italy; (R.T.); (R.Q.)
| | - Simona Coco
- Lung Cancer Unit, IRCCS Ospedale Policlinico San Martino, 10-16132 Genova, Italy;
| | - Anna Garuti
- Clinica Oncologia Medica ad Indirizzo Oncologico, IRCCS Ospedale Policlinico San Martino, 10-16132 Genova, Italy;
| | - Giulia Buzzatti
- Oncologia Medica 2, IRCCS Ospedale Policlinico San Martino, 10-16132 Genova, Italy;
| | - Andrea Sciutto
- Clinica di Oncologia Medica, IRCCS Ospedale Policlinico San Martino, 10-16132 Genova, Italy; (A.S.); (L.A.); (L.D.M.)
| | - Luca Arecco
- Clinica di Oncologia Medica, IRCCS Ospedale Policlinico San Martino, 10-16132 Genova, Italy; (A.S.); (L.A.); (L.D.M.)
| | - Federico Villa
- Molecular Oncology and Angiogenesis Unit, IRCCS Ospedale Policlinico San Martino, 10-16132 Genova, Italy;
| | - Franca Carli
- Surgical Pathology Unit, IRCCS Ospedale Policlinico San Martino, 10-16132 Genova, Italy;
| | - Daniele Reverberi
- Molecular Pathology Unit, IRCCS Ospedale Policlinico San Martino, 10-16132 Genova, Italy;
| | - Rodolfo Quarto
- Department of Experimental Medicine (DIMES), University of Genova, 5-16126 Genova, Italy; (R.T.); (R.Q.)
- Cellular Oncology Unit, IRCCS Ospedale Policlinico San Martino, 10-16132 Genova, Italy
| | - Mariella Dono
- Molecular Diagnostic Unit, IRCCS Ospedale Policlinico San Martino, 10-16132 Genova, Italy;
| | - Lucia Del Mastro
- Clinica di Oncologia Medica, IRCCS Ospedale Policlinico San Martino, 10-16132 Genova, Italy; (A.S.); (L.A.); (L.D.M.)
- Department of Internal Medicine and Medical Specialties (DIMI), University of Genova, 5-16126 Genova, Italy
| |
Collapse
|
20
|
Sabath DE, Perrone ME, Clein A, Tam M, Hardin M, Trimble S, Ramirez A, Duplessis M, Mojica T, Higano CS, Gadi VK, Kaldjian E, George T. Clinical Validation of a Circulating Tumor Cell Assay Using Density Centrifugation and Automated Immunofluorescence Microscopy. Am J Clin Pathol 2022; 158:270-276. [PMID: 35460401 DOI: 10.1093/ajcp/aqac040] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2021] [Accepted: 03/08/2022] [Indexed: 11/13/2022] Open
Abstract
OBJECTIVES The US Food and Drug Administration (FDA)-approved CELLSEARCH assay (Menarini Silicon Biosystems) for circulating tumor cells (CTCs) relies on expression of an epithelial cell adhesion molecule to enrich for CTCs. We sought to validate a CTC assay (RareCyte) for clinical use that instead collects a buffy coat preparation enriched for CTCs. METHODS Normal peripheral blood specimens spiked with cultured breast and prostate cancer cells and 47 clinical samples were used to validate assay performance. Specimens were enriched for buffy coat cells and applied onto 8 glass slides. The slides were immunofluorescently stained and imaged by automated microscopy and computer-aided image analysis. RESULTS The assay was 100% specific for detecting spiked tumor cells. For samples spiked with 25, 50, and 125 cells, the percentage coefficients of variation were 42%, 21%, and 3.7%, respectively. Linearity studies demonstrated a slope of 0.99, an intercept of 1.6, and R2 of 0.96. Recoveries at the 25-, 50-, and 125-cell levels were 92%, 111%, and 100%, respectively. Clinical samples run on both CELLSEARCH and RareCyte correlated with an R2 of 0.8 after log-transformation and demonstrated 87.5% concordance using the CELLSEARCH criteria for predicting adverse outcomes. CONCLUSIONS The RareCyte CTC assay has comparable performance to the FDA-cleared method and is ready for further clinical validation studies.
Collapse
Affiliation(s)
- Daniel E Sabath
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA, USA
- Department of Medicine, University of Washington, Seattle, WA, USA
| | - Marie E Perrone
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA, USA
| | - Alisa Clein
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA, USA
| | - Michael Tam
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA, USA
| | - Michael Hardin
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA, USA
| | - Sara Trimble
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA, USA
| | | | | | - Tanisha Mojica
- Department of Medicine, University of Washington, Seattle, WA, USA
| | | | - V K Gadi
- Department of Medicine, University of Washington, Seattle, WA, USA
| | | | - Tad George
- Department of RareCyte, Seattle, WA, USA
| |
Collapse
|
21
|
The Role of Circulating Tumor Cells in the Prognosis of Metastatic Triple-Negative Breast Cancers: A Systematic Review of the Literature. Biomedicines 2022; 10:biomedicines10040769. [PMID: 35453519 PMCID: PMC9025371 DOI: 10.3390/biomedicines10040769] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2022] [Revised: 03/20/2022] [Accepted: 03/22/2022] [Indexed: 12/24/2022] Open
Abstract
Breast cancer is one of the leading causes of death in women worldwide. One subtype of breast cancer is the triple-negative, which accounts for 15% of total breast cancer cases and is known for its poor prognosis. The main cause of death is due to metastasis. Circulating tumor cells (CTCs) play a key role in the metastatic process. CTCs arise either by detaching from the primary tumor or from cancer stem cells undergoing an epithelial-to-mesenchymal transition (EMT). This review aims to present up-to-date data concerning the role of CTC numbers in relation to the prognostic and treatment response in metastatic triple-negative breast cancer (mTNBC) patients, and also to discuss the methods used for CTCs’ identification. A search in the MEDLINE database was performed. A total of 234 articles were identified. The results of the 24 eligible studies showed that positive CTC status is associated with shorter overall survival (OS) and progression-free survival (PFS) in mTNBC patients. Furthermore, a decrease in number of CTCs during therapy seems to be a favorable prognostic factor, making CTCs’ detection an important prognostic tool before and during therapy in mTNBC patients. The methods used for CTC detection are still developing and need further improvement.
Collapse
|
22
|
Diaz M, Singh P, Kotchetkov IS, Skakodub A, Meng A, Tamer C, Young RJ, Reiner AS, Panageas KS, Ramanathan LV, Pentsova E. Quantitative assessment of circulating tumor cells in cerebrospinal fluid as a clinical tool to predict survival in leptomeningeal metastases. J Neurooncol 2022; 157:81-90. [PMID: 35113288 PMCID: PMC9119011 DOI: 10.1007/s11060-022-03949-1] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2021] [Accepted: 01/15/2022] [Indexed: 10/19/2022]
Abstract
PURPOSE Circulating tumor cells in cerebrospinal fluid are a quantitative diagnostic tool for leptomeningeal metastases from solid tumors, but their prognostic significance is unclear. Our objective was to evaluate CSF-CTC quantification in predicting outcomes in LM. METHODS This is a single institution retrospective study of patients with solid tumors who underwent CSF-CTC quantification using the CellSearch® platform between 04/2016 and 06/2019. Information on neuroaxis imaging, CSF results, and survival was collected. LM was diagnosed by MRI and/or CSF cytology. Survival analyses were performed using multivariable Cox proportional hazards modeling, and CSF-CTC splits associated with survival were identified through recursive partitioning analysis. RESULTS Out of 290 patients with CNS metastases, we identified a cohort of 101 patients with newly diagnosed LM. In this group, CSF-CTC count (median 200 CTCs/3 ml) predicted survival continuously (HR = 1.005, 95% CI: 1.002-1.009, p = 0.0027), and the risk of mortality doubled (HR = 2.84, 95% CI: 1.45-5.56, p = 0.0023) at the optimal cutoff of ≥ 61 CSF-CTCs/3 ml. Neuroimaging findings of LM (assessed by 3 independent neuroradiologists) were associated with a higher CSF-CTC count (median CSF-CTCs range 1.5-4 for patients without radiographic LM vs 200 for patients with radiographic LM, p < 0.001), but did not predict survival. CONCLUSION Our data shows that CSF-CTCs quantification predicts survival in newly diagnosed LM, and outperforms neuroimaging. CSF-CTC analysis can be used as a prognostic tool in patients with LM and provides quantitative assessment of disease burden in the CNS compartment.
Collapse
Affiliation(s)
- Maria Diaz
- Department of Neurology, Memorial Sloan Kettering Cancer Center, New York, NY
| | | | - Ivan S. Kotchetkov
- Department of Neurology, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Anna Skakodub
- Department of Neurology, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Alicia Meng
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Christel Tamer
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Robert J. Young
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Anne S. Reiner
- Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Katherine S. Panageas
- Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Lakshmi V. Ramanathan
- Department of Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Elena Pentsova
- Department of Neurology, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, NY, 10065, USA.
| |
Collapse
|
23
|
Diaz M, Fleisher M, Pentsova EI. Cerebrospinal fluid circulating tumor cells for diagnosis, response evaluation, and molecular profiling of leptomeningeal metastases from solid tumors. Cancer Biomark 2022. [DOI: 10.1016/b978-0-12-824302-2.00007-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/17/2022]
|
24
|
Lin D, Shen L, Luo M, Zhang K, Li J, Yang Q, Zhu F, Zhou D, Zheng S, Chen Y, Zhou J. Circulating tumor cells: biology and clinical significance. Signal Transduct Target Ther 2021; 6:404. [PMID: 34803167 PMCID: PMC8606574 DOI: 10.1038/s41392-021-00817-8] [Citation(s) in RCA: 347] [Impact Index Per Article: 115.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2021] [Revised: 10/06/2021] [Accepted: 10/27/2021] [Indexed: 02/07/2023] Open
Abstract
Circulating tumor cells (CTCs) are tumor cells that have sloughed off the primary tumor and extravasate into and circulate in the blood. Understanding of the metastatic cascade of CTCs has tremendous potential for the identification of targets against cancer metastasis. Detecting these very rare CTCs among the massive blood cells is challenging. However, emerging technologies for CTCs detection have profoundly contributed to deepening investigation into the biology of CTCs and have facilitated their clinical application. Current technologies for the detection of CTCs are summarized herein, together with their advantages and disadvantages. The detection of CTCs is usually dependent on molecular markers, with the epithelial cell adhesion molecule being the most widely used, although molecular markers vary between different types of cancer. Properties associated with epithelial-to-mesenchymal transition and stemness have been identified in CTCs, indicating their increased metastatic capacity. Only a small proportion of CTCs can survive and eventually initiate metastases, suggesting that an interaction and modulation between CTCs and the hostile blood microenvironment is essential for CTC metastasis. Single-cell sequencing of CTCs has been extensively investigated, and has enabled researchers to reveal the genome and transcriptome of CTCs. Herein, we also review the clinical applications of CTCs, especially for monitoring response to cancer treatment and in evaluating prognosis. Hence, CTCs have and will continue to contribute to providing significant insights into metastatic processes and will open new avenues for useful clinical applications.
Collapse
Affiliation(s)
- Danfeng Lin
- Department of Breast Surgery, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Cancer Institute (Key Laboratory of Cancer Prevention and Intervention, China National Ministry of Education), the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Department of Breast Surgery, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Lesang Shen
- Department of Breast Surgery, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Meng Luo
- Department of Breast Surgery, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Cancer Institute (Key Laboratory of Cancer Prevention and Intervention, China National Ministry of Education), the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Kun Zhang
- Department of Breast Surgery, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Jinfan Li
- Department of Pathology, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Qi Yang
- Department of Pathology, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Fangfang Zhu
- Department of Breast Surgery, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Dan Zhou
- Department of Surgery, Traditional Chinese Medical Hospital of Zhuji, Shaoxing, China
| | - Shu Zheng
- Department of Breast Surgery, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Cancer Institute (Key Laboratory of Cancer Prevention and Intervention, China National Ministry of Education), the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Yiding Chen
- Department of Breast Surgery, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China.
- Cancer Institute (Key Laboratory of Cancer Prevention and Intervention, China National Ministry of Education), the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China.
| | - Jiaojiao Zhou
- Department of Breast Surgery, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China.
- Cancer Institute (Key Laboratory of Cancer Prevention and Intervention, China National Ministry of Education), the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China.
| |
Collapse
|
25
|
Zhang X, Wei X, Men X, Wu CX, Bai JJ, Li WT, Yang T, Chen ML, Wang JH. Dual-Multivalent-Aptamer-Conjugated Nanoprobes for Superefficient Discerning of Single Circulating Tumor Cells in a Microfluidic Chip with Inductively Coupled Plasma Mass Spectrometry Detection. ACS APPLIED MATERIALS & INTERFACES 2021; 13:43668-43675. [PMID: 34473482 DOI: 10.1021/acsami.1c11953] [Citation(s) in RCA: 34] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/13/2023]
Abstract
The efficient recognition of circulating tumor cells (CTCs) with an aptamer probe confers numerous benefits; however, the stability and binding affinity of aptamers are significantly hampered in real biological sample matrices. Inspired by the efficient preying mechanism by multiplex tubing feet and endoskeletons of sea urchins, we engineered a superefficient biomimetic single-CTC recognition platform by conjugating dual-multivalent-aptamers (DMAs) Sgc8 and SYL3C onto AuNPs to form a sea urchin-like nanoprobe (sea urchin-DMA-AuNPs). Aptamers Sgc8 and SYL3C selectively bind with the biomarker proteins PTK7 and EpCAM expressed on the surface of CTCs. CTCs were captured with 100% efficiency, followed by sorting on a specially designed multifunctional microfluidic configuration, integrating a single-CTC separation unit and a hydrodynamic filtrating purification unit. After sorting, background-free analysis of biomarker proteins in single CTCs was undertaken with inductively coupled plasma mass spectrometry by measuring the amount of 197Au isotope in sea urchin-DMA-AuNPs. With respect to a single-aptamer nanoprobe/-interface, the dual-aptamer nanoprobe improves the binding efficiency by more than 200% (Kd < 0.35 nM). The microchip facilitates the recognition of single CTCs with a sorting separation rate of 93.6% at a flow rate of 60 μL min-1, and it exhibits 73.8 ± 5.0% measurement efficiency for single CTCs. The present strategy ensures the manipulation and detection of a single CTC in 100 μL of whole blood within 1 h.
Collapse
Affiliation(s)
- Xuan Zhang
- Research Center for Analytical Sciences, Department of Chemistry, College of Sciences, Northeastern University, Box 332, Shenyang 110819, China
| | - Xing Wei
- Research Center for Analytical Sciences, Department of Chemistry, College of Sciences, Northeastern University, Box 332, Shenyang 110819, China
| | - Xue Men
- Research Center for Analytical Sciences, Department of Chemistry, College of Sciences, Northeastern University, Box 332, Shenyang 110819, China
| | - Cheng-Xin Wu
- Research Center for Analytical Sciences, Department of Chemistry, College of Sciences, Northeastern University, Box 332, Shenyang 110819, China
| | - Jun-Jie Bai
- Research Center for Analytical Sciences, Department of Chemistry, College of Sciences, Northeastern University, Box 332, Shenyang 110819, China
| | - Wei-Tao Li
- Research Center for Analytical Sciences, Department of Chemistry, College of Sciences, Northeastern University, Box 332, Shenyang 110819, China
| | - Ting Yang
- Research Center for Analytical Sciences, Department of Chemistry, College of Sciences, Northeastern University, Box 332, Shenyang 110819, China
| | - Ming-Li Chen
- Research Center for Analytical Sciences, Department of Chemistry, College of Sciences, Northeastern University, Box 332, Shenyang 110819, China
| | - Jian-Hua Wang
- Research Center for Analytical Sciences, Department of Chemistry, College of Sciences, Northeastern University, Box 332, Shenyang 110819, China
| |
Collapse
|
26
|
Hepatic Cancer Stem Cells: Molecular Mechanisms, Therapeutic Implications, and Circulating Biomarkers. Cancers (Basel) 2021; 13:cancers13184550. [PMID: 34572776 PMCID: PMC8472624 DOI: 10.3390/cancers13184550] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2021] [Revised: 09/07/2021] [Accepted: 09/08/2021] [Indexed: 12/12/2022] Open
Abstract
Hepatocellular carcinoma (HCC) is one of the deadliest cancers. HCC is associated with multiple risk factors and is characterized by a marked tumor heterogeneity that makes its molecular classification difficult to apply in the clinics. The lack of circulating biomarkers for the diagnosis, prognosis, and prediction of response to treatments further undermines the possibility of developing personalized therapies. Accumulating evidence affirms the involvement of cancer stem cells (CSCs) in tumor heterogeneity, recurrence, and drug resistance. Owing to the contribution of CSCs to treatment failure, there is an urgent need to develop novel therapeutic strategies targeting, not only the tumor bulk, but also the CSC subpopulation. Clarification of the molecular mechanisms influencing CSC properties, and the identification of their functional roles in tumor progression, may facilitate the discovery of novel CSC-based therapeutic targets to be used alone, or in combination with current anticancer agents, for the treatment of HCC. Here, we review the driving forces behind the regulation of liver CSCs and their therapeutic implications. Additionally, we provide data on their possible exploitation as prognostic and predictive biomarkers in patients with HCC.
Collapse
|
27
|
Patil RA, Srinivasarao M, Amiji MM, Low PS, Niedre M. Fluorescence Labeling of Circulating Tumor Cells with a Folate Receptor-Targeted Molecular Probe for Diffuse In Vivo Flow Cytometry. Mol Imaging Biol 2021; 22:1280-1289. [PMID: 32519245 DOI: 10.1007/s11307-020-01505-9] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
PURPOSE We recently developed a new instrument called "diffuse in vivo flow cytometry" (DiFC) for enumeration of rare fluorescently labeled circulating tumor cells (CTCs) in small animals without drawing blood samples. Until now, we have used cell lines that express fluorescent proteins or were pre-labeled with a fluorescent dye ex vivo. In this work, we investigated the use of a folate receptor (FR)-targeted fluorescence molecular probe for in vivo labeling of FR+ CTCs for DiFC. PROCEDURES We used EC-17, a FITC-folic acid conjugate that has been used in clinical trials for fluorescence-guided surgery. We studied the affinity of EC-17 for FR+ L1210A and KB cancer cells. We also tested FR- MM.1S cells. We tested the labeling specificity in cells in culture in vitro and in whole blood. We also studied the detectability of labeled cells in mice in vivo with DiFC. RESULTS EC-17 showed a high affinity for FR+ L1210A and KB cells in vitro. In whole blood, 85.4 % of L1210A and 80.9 % of KB cells were labeled above non-specific background with EC-17, and negligible binding to FR- MM.1S cells was observed. In addition, EC-17-labeled CTCs were readily detectable in circulation in mice with DiFC. CONCLUSIONS This work demonstrates the feasibility of labeling CTCs with a cell-surface receptor-targeted probe for DiFC, greatly expanding the potential utility of the method for pre-clinical animal models. Because DiFC uses diffuse light, this method could be also used to enumerate CTCs in larger animal models and potentially even in humans.
Collapse
Affiliation(s)
- Roshani A Patil
- Department of Bioengineering, Northeastern University, Boston, MA, 02115, USA
| | | | - Mansoor M Amiji
- Department of Pharmaceutical Sciences, School of Pharmacy, Northeastern University, Boston, MA, 02115, USA
| | - Philip S Low
- Department of Chemistry, Purdue University, West Lafayette, IN, 47906, USA
| | - Mark Niedre
- Department of Bioengineering, Northeastern University, Boston, MA, 02115, USA.
| |
Collapse
|
28
|
Chantzara E, Xenidis N, Kallergi G, Georgoulias V, Kotsakis A. Circulating tumor cells as prognostic biomarkers in breast cancer: current status and future prospects. Expert Rev Mol Diagn 2021; 21:1037-1048. [PMID: 34328384 DOI: 10.1080/14737159.2021.1962710] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Introduction : Despite advances in diagnostic and therapeutic techniques breast cancer is still associated with significant morbidity and mortality. CTCs play a crucial role in the metastatic process, which is the main cause of death in BC patients.Areas covered : This review discusses the prognostic and predictive value of CTCs and their prospective in management of BC patients.Expert opinion : The analysis of CTCs through improved technologies offers a new insight into the metastatic cascade. Assessment of the number and molecular profile of CTCs holds great promises for disease monitoring and therapeutic decisions. However, more research is needed until they can be used in therapeutic decisions in clinical practice.
Collapse
Affiliation(s)
- Evagelia Chantzara
- Department of Medical Oncology, University General Hospital of Larissa, Larissa, Thessaly, Greece
| | - Nikolaos Xenidis
- Department of Medical Oncology, University General Hospital of Alexandroupolis, Alexandroupolis, Thrace, Greece
| | - Galatea Kallergi
- Division of Genetics, Cell and Developmental Biology, Department of Biology, University of Patras, Patras, Greece
| | - Vassilis Georgoulias
- Department of Medical Oncology, Hellenic Oncology Research Group (HORG), Athens, Greece
| | - Athanasios Kotsakis
- Department of Medical Oncology, University General Hospital of Larissa, Larissa, Thessaly, Greece.,Faculty of Medicine, School of Health Sciences, University of Thessaly, Larissa, Thessaly, Greece
| |
Collapse
|
29
|
Tay TKY, Tan PH. Liquid Biopsy in Breast Cancer: A Focused Review. Arch Pathol Lab Med 2021; 145:678-686. [PMID: 32045277 DOI: 10.5858/arpa.2019-0559-ra] [Citation(s) in RCA: 61] [Impact Index Per Article: 20.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/11/2019] [Indexed: 01/27/2023]
Abstract
CONTEXT.— The role of liquid biopsy in cancer management has been gaining increased prominence in the past decade, with well-defined clinical applications now being established in lung cancer. Recently, the US Food and Drug Administration also approved the Therascreen PIK3CA RGQ polymerase chain reaction assay as a companion diagnostic assay to detect PIK3CA mutations in breast cancer for both tissue and liquid biopsies, bringing the role of liquid biopsy in breast cancer management to the fore. Its utility in other aspects of breast cancer, however, is yet to be clearly defined. OBJECTIVE.— To review the studies that looked at liquid biopsies in breast cancer and examine their potential for clinical application in the areas of early diagnosis, prognostication, monitoring disease response, detecting minimal residual disease, and predicting risk of progression or relapse. We focus mainly on circulating tumor cells and circulating tumor DNA. DATA SOURCES.— Peer-reviewed articles in PubMed. CONCLUSIONS.— Liquid biopsies in breast cancers have yielded promising results, especially in the areas of monitoring treatment response and predicting disease progression or relapse. With further study, and hopefully coupled with continued improvements in technologies that isolate tumor-derived materials, liquid biopsies may go on to play a greater role in the breast cancer clinic.
Collapse
Affiliation(s)
- Timothy Kwang Yong Tay
- From the Department of Anatomical Pathology (Tay, Tan), Singapore General Hospital, Singapore
| | - Puay Hoon Tan
- From the Department of Anatomical Pathology (Tay, Tan), Singapore General Hospital, Singapore.,The Division of Pathology (Tan), Singapore General Hospital, Singapore
| |
Collapse
|
30
|
Yu Z, Song M, Chouchane L, Ma X. Functional Genomic Analysis of Breast Cancer Metastasis: Implications for Diagnosis and Therapy. Cancers (Basel) 2021; 13:cancers13133276. [PMID: 34208889 PMCID: PMC8268362 DOI: 10.3390/cancers13133276] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2021] [Revised: 06/29/2021] [Accepted: 06/29/2021] [Indexed: 12/11/2022] Open
Abstract
Simple Summary Metastasis remains the greatest cause of fatalities in breast cancer patients world-wide. The process of metastases is highly complex, and the current research efforts in this area are still rather fragmented. The revolution of genomic profiling methods to analyze samples from human and animal models dramatically improved our understanding of breast cancer metastasis. This article summarizes the recent breakthroughs in genomic analyses of breast cancer metastasis and discusses their implications for prognostic and therapeutic applications. Abstract Breast cancer (BC) is one of the most diagnosed cancers worldwide and is the second cause of cancer related death in women. The most frequent cause of BC-related deaths, like many cancers, is metastasis. However, metastasis is a complicated and poorly understood process for which there is a shortage of accurate prognostic indicators and effective treatments. With the rapid and ever-evolving development and application of genomic sequencing technologies, many novel molecules were identified that play previously unappreciated and important roles in the various stages of metastasis. In this review, we summarize current advancements in the functional genomic analysis of BC metastasis and discuss about the potential prognostic and therapeutic implications from the recent genomic findings.
Collapse
Affiliation(s)
- Ziqi Yu
- Department of Microbiology and Immunology, Weill Cornell Medicine, 1300 York Avenue, New York, NY 10065, USA;
- Correspondence: (Z.Y.); (X.M.)
| | - Mei Song
- Department of Microbiology and Immunology, Weill Cornell Medicine, 1300 York Avenue, New York, NY 10065, USA;
| | - Lotfi Chouchane
- Department of Genetic Medicine, Weill Cornell Medicine-Qatar, Qatar Foundation, Doha P.O. Box 24144, Qatar;
| | - Xiaojing Ma
- Department of Microbiology and Immunology, Weill Cornell Medicine, 1300 York Avenue, New York, NY 10065, USA;
- Correspondence: (Z.Y.); (X.M.)
| |
Collapse
|
31
|
Paoletti C, Regan MM, Niman SM, Dolce EM, Darga EP, Liu MC, Marcom PK, Hart LL, Smith JW, Tedesco KL, Amir E, Krop IE, DeMichele AM, Goodwin PJ, Block M, Aung K, Brown ME, McCormack RT, Hayes DF. Circulating tumor cell number and endocrine therapy index in ER positive metastatic breast cancer patients. NPJ Breast Cancer 2021; 7:77. [PMID: 34117261 PMCID: PMC8196036 DOI: 10.1038/s41523-021-00281-1] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2021] [Accepted: 05/12/2021] [Indexed: 12/14/2022] Open
Abstract
Circulating tumor cells (CTC) are prognostic in metastatic breast cancer (MBC). The CTC-endocrine therapy index (CTC-ETI), consisting of CTC-ER (estrogen receptor), BCL2, human epidermal growth factor receptor (HER2), and Ki67 expression, might predict resistance to endocrine therapy (ET) in patients with ER-positive MBC. One hundred twenty-one patients with ER-positive/HER2-negative MBC initiating a new ET after ≥1 lines of ET were enrolled in a prospective, multi-institutional clinical trial. CTC-ETI and clinical/imaging follow-up were performed at baseline and serial time points. Progression-free survival (PFS) and rapid progression (RP; determined at the 3-month time point) were primary endpoints. Associations with clinical outcomes used logrank and Fisher’s exact tests. At baseline, 36% (38/107) of patients had ≥5 CTC/7.5 ml whole blood (WB). Patients with ≥5 vs. <5 CTC/7.5 ml WB had significantly worse PFS (median 3.3 vs. 5.9 months, P = 0.03). Elevated CTC at 1 month was associated with even worse PFS (1.9 vs. 5.0 months from the 1-month sample, P < 0.001). Low, intermediate, and high CTC-ETI were observed in 71 (66%), 8 (8%), and 28 (26%) patients, with median PFS of 6.9, 8.5, and 2.8 months, respectively (P = 0.008). Patients with high vs. low CTC and CTC-ETI more frequently experienced RP (CTC: 66% vs. 41%; P = 0.03; CTC-ETI: 79% vs. 40%; P = 0.002). In conclusion, CTC enumeration and the CTC-ETI assay are prognostic at baseline and follow-up in patients with ER-positive/HER2-negative MBC starting new ET. CTC at first follow-up might identify a group of patients with ER-positive MBC that could forego ET, but CTC-ETI did not contribute further.
Collapse
Affiliation(s)
- Costanza Paoletti
- Department of Internal Medicine, Division of Hematology/Oncology, University of Michigan Rogel Cancer Center, Ann Arbor, MI, USA.,Currently at EISAI, Inc., Woodcliff Lake, NJ, USA
| | - Meredith M Regan
- Division of Biostatistics, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Samuel M Niman
- Division of Biostatistics, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Emily M Dolce
- Department of Internal Medicine, Division of Hematology/Oncology, University of Michigan Rogel Cancer Center, Ann Arbor, MI, USA
| | - Elizabeth P Darga
- Department of Internal Medicine, Division of Hematology/Oncology, University of Michigan Rogel Cancer Center, Ann Arbor, MI, USA
| | - Minetta C Liu
- Department of Oncology, Division of Medical Oncology, and Department of Laboratory Medicine and Pathology, Division of Anatomic Pathology, Mayo Clinic, Rochester, MN, USA
| | - P Kelly Marcom
- Department of Medicine, Division of Medical Oncology, Duke Cancer Institute, Durham, NC, USA
| | - Lowell L Hart
- Florida Cancer Specialists/Wake Forest School of Medicine, Fort Myers, FL, USA
| | - John W Smith
- US Oncology Research, Compass Oncology, Portland, OR, USA
| | - Karen L Tedesco
- New York Oncology Hematology, US Oncology Network, Albany, NY, USA
| | - Eitan Amir
- Division of Medical Oncology and Hematology, Princess Margaret Cancer Centre, Toronto, ON, Canada
| | - Ian E Krop
- Department of Medical Oncology, Division of Breast Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Angela M DeMichele
- Department of Medicine, Division of Hematology/Oncology, University of Pennsylvania, Philadelphia, PA, USA
| | - Pamela J Goodwin
- Department of Medicine, Sinai Health System, University of Toronto, Toronto, ON, Canada
| | | | - Kimberly Aung
- Department of Internal Medicine, Division of Hematology/Oncology, University of Michigan Rogel Cancer Center, Ann Arbor, MI, USA
| | - Martha E Brown
- Department of Internal Medicine, Division of Hematology/Oncology, University of Michigan Rogel Cancer Center, Ann Arbor, MI, USA
| | | | - Daniel F Hayes
- Department of Internal Medicine, Division of Hematology/Oncology, University of Michigan Rogel Cancer Center, Ann Arbor, MI, USA.
| |
Collapse
|
32
|
Wang X, Cheng S, Wang X, Wei L, Kong Q, Ye M, Luo X, Xu J, Zhang C, Xian Y. pH-Sensitive Dye-Based Nanobioplatform for Colorimetric Detection of Heterogeneous Circulating Tumor Cells. ACS Sens 2021; 6:1925-1932. [PMID: 33881313 DOI: 10.1021/acssensors.1c00314] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
The efficient capture and sensitive detection of circulating tumor cells (CTCs) play a vital role in cancer diagnosis and prognosis. However, CTCs in the peripheral blood are very rare and heterogeneous, which make them difficult to isolate and detect. Herein, a novel colorimetric nanobioplatform was successfully developed for the highly efficient capture and highly sensitive detection of heterogeneous CTCs, which consisted of two parts: the multivalent aptamer-modified gold nanoparticles as the capture unit and two kinds of aptamer-functionalized pH-sensitive allochroic dyes (thymolphthalein and curcumin) @ molybdenum disulfide nanoflakes (MoS2 NFs) acting as the visual simultaneous detection of heterogeneous CTCs. Using MCF-7 and HeLa cells as the CTC models, the capture unit can effectively isolate the CTCs due to the multivalent probe with improved affinity. The two allochroic dyes can display obvious color changes under alkaline conditions (pH 12.5) in the presence of MCF-7 and HeLa cells, which provided a rapid and sensitive strategy for visualizing simultaneous detection of heterogeneous CTCs as low as 5 cells mL-1. This nanoplatform possessed a high sensitivity toward CTC detection owing to high dye loading capacity of MoS2 NFs and allochroic dyes with excellent pH sensitivity. It can successfully distinguish and quantitatively detect the targeted heterogeneous CTCs from numerous interfering cells in diluted whole blood. It can also be used to detect CTCs from lysed blood samples from cancer patients, indicating promising application for cancer diagnosis.
Collapse
Affiliation(s)
- Xiuli Wang
- Shanghai Engineering Research Center of Molecular Therapeutics and New Drug Development, Department of Chemistry, School of Chemistry and Molecular Engineering, East China Normal University, Shanghai 200241, China
| | - Shasha Cheng
- Shanghai Engineering Research Center of Molecular Therapeutics and New Drug Development, Department of Chemistry, School of Chemistry and Molecular Engineering, East China Normal University, Shanghai 200241, China
| | - Xinjun Wang
- Shanghai Zhangjiang Institute of Medical Innovation, Shanghai 201204, China
- Translational Medical Center for Stem Cell Therapy and Institute for Regenerative Medicine, Shanghai East Hospital, School of Life Sciences and Technology, Tongji University, Shanghai 200092, China
| | - Liran Wei
- Shanghai Zhangjiang Institute of Medical Innovation, Shanghai 201204, China
| | - Qianqian Kong
- Shanghai Engineering Research Center of Molecular Therapeutics and New Drug Development, Department of Chemistry, School of Chemistry and Molecular Engineering, East China Normal University, Shanghai 200241, China
| | - Mingqiang Ye
- Shanghai Engineering Research Center of Molecular Therapeutics and New Drug Development, Department of Chemistry, School of Chemistry and Molecular Engineering, East China Normal University, Shanghai 200241, China
| | - Xianzhu Luo
- Shanghai Engineering Research Center of Molecular Therapeutics and New Drug Development, Department of Chemistry, School of Chemistry and Molecular Engineering, East China Normal University, Shanghai 200241, China
| | - Jiao Xu
- Shanghai Engineering Research Center of Molecular Therapeutics and New Drug Development, Department of Chemistry, School of Chemistry and Molecular Engineering, East China Normal University, Shanghai 200241, China
| | - Cuiling Zhang
- Shanghai Engineering Research Center of Molecular Therapeutics and New Drug Development, Department of Chemistry, School of Chemistry and Molecular Engineering, East China Normal University, Shanghai 200241, China
| | - Yuezhong Xian
- Shanghai Engineering Research Center of Molecular Therapeutics and New Drug Development, Department of Chemistry, School of Chemistry and Molecular Engineering, East China Normal University, Shanghai 200241, China
| |
Collapse
|
33
|
Jin L, Fan WH, Luan Y, Wu M, Zhao W. Evaluation of circulating tumor cells as a prognostic biomarker for early recurrence in stage II-III breast cancer patients using CytoSorter ® system: a retrospective study. PeerJ 2021; 9:e11366. [PMID: 33987029 PMCID: PMC8088762 DOI: 10.7717/peerj.11366] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2020] [Accepted: 04/07/2021] [Indexed: 12/25/2022] Open
Abstract
PURPOSE Circulating tumor cells (CTCs) are known to be associated with late recurrence and poor prognosis in breast cancer (BC). Different CTC enrichment platforms have different CTC cut-off values for poor prognosis. This study aimed to evaluate whether preoperative CTCs could be a prognostic factor for early recurrence of disease in BC patients with resectable tumors, and to ascertain the CTC cut-off value for early recurrence with CytoSorter® CTC system. METHODS Thirty-six stage II and III BC patients who had preoperative (pre-op) CTC detection and underwent a mastectomy or lumpectomy for curative intent between January and May 2018 were enrolled in this retrospective study. CTC detection was performed using CytoSorter® CTC system. Correlations of patients' demographics, clinicopathological characteristics, adjuvant therapies and CTCs with relapse and survival were evaluated. RESULTS CTCs were detected in 32 out of 36 patients before surgery. Nine patients developed relapses during follow-up, and seven of them were distant recurrence. Univariate analysis showed that CTCs were correlated with two-year recurrence free survival (RFS) and distant RFS (D-RFS) (P = 0.013 and 0.029, respectively). Two-year RFS and D-RFS were 85.2% and 88.9%, respectively, for patients with <4 CTCs, while 44.4% and 55.6%, respectively, for patients with ≧4 CTCs. In multivariate analysis, only CTC was shown to be correlated with two-year RFS (HR: 0.219, 95% CI: [0.058-0.82], P = 0.024) and D-RFS (HR: 0.218, 95% CI [0.048-0.977], P = 0.047). CONCLUSION BC patients with pre-op CTCs ≥4 per four mL of blood have significantly reduced two-year RFS and D-RFS. A pre-op CTC cut-off of four per four mL of blood was found for CytoSorter® to identify BC patients with a higher risk for early recurrence.
Collapse
Affiliation(s)
- Lidan Jin
- Department of Surgical Oncology, Sir Run Run Shaw Hospital, Zhejiang University College of Medicine, Hangzhou, China
| | | | - Yi Luan
- Department of Clinical Laboratory, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
| | | | - Wenhe Zhao
- Department of Surgical Oncology, Sir Run Run Shaw Hospital, Zhejiang University College of Medicine, Hangzhou, China
| |
Collapse
|
34
|
Magbanua MJM, Hendrix LH, Hyslop T, Barry WT, Winer EP, Hudis C, Toppmeyer D, Carey LA, Partridge AH, Pierga JY, Fehm T, Vidal-Martínez J, Mavroudis D, Garcia-Saenz JA, Stebbing J, Gazzaniga P, Manso L, Zamarchi R, Antelo ML, Mattos-Arruda LD, Generali D, Caldas C, Munzone E, Dirix L, Delson AL, Burstein HJ, Qadir M, Ma C, Scott JH, Bidard FC, Park JW, Rugo HS. Serial Analysis of Circulating Tumor Cells in Metastatic Breast Cancer Receiving First-Line Chemotherapy. J Natl Cancer Inst 2021; 113:443-452. [PMID: 32770247 PMCID: PMC8023821 DOI: 10.1093/jnci/djaa113] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2020] [Revised: 06/23/2020] [Accepted: 07/29/2020] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND We examined the prognostic significance of circulating tumor cell (CTC) dynamics during treatment in metastatic breast cancer (MBC) patients receiving first-line chemotherapy. METHODS Serial CTC data from 469 patients (2202 samples) were used to build a novel latent mixture model to identify groups with similar CTC trajectory (tCTC) patterns during the course of treatment. Cox regression was used to estimate hazard ratios for progression-free survival (PFS) and overall survival (OS) in groups based on baseline CTCs, combined CTC status at baseline to the end of cycle 1, and tCTC. Akaike information criterion was used to select the model that best predicted PFS and OS. RESULTS Latent mixture modeling revealed 4 distinct tCTC patterns: undetectable CTCs (56.9% ), low (23.7%), intermediate (14.5%), or high (4.9%). Patients with low, intermediate, and high tCTC patterns had statistically significant inferior PFS and OS compared with those with undetectable CTCs (P < .001). Akaike Information Criterion indicated that the tCTC model best predicted PFS and OS compared with baseline CTCs and combined CTC status at baseline to the end of cycle 1 models. Validation studies in an independent cohort of 1856 MBC patients confirmed these findings. Further validation using only a single pretreatment CTC measurement confirmed prognostic performance of the tCTC model. CONCLUSIONS We identified 4 novel prognostic groups in MBC based on similarities in tCTC patterns during chemotherapy. Prognostic groups included patients with very poor outcome (intermediate + high CTCs, 19.4%) who could benefit from more effective treatment. Our novel prognostic classification approach may be used for fine-tuning of CTC-based risk stratification strategies to guide future prospective clinical trials in MBC.
Collapse
Affiliation(s)
| | | | - Terry Hyslop
- Duke Cancer Institute, Duke University, Durham, NC, USA
| | - William T Barry
- Alliance Statistics and Data Center, Dana-Farber/Partners CancerCare, Boston, MA, USA
- Rho Inc., Raleigh, NC, USA
| | - Eric P Winer
- Dana-Farber/Partners CancerCare, Boston, MA, USA
| | - Clifford Hudis
- Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | | | - Lisa Anne Carey
- UNC Lineberger Comprehensive Cancer Center, Chapel Hill, NC, USA
| | | | - Jean-Yves Pierga
- Department of Medical Oncology, Institut Curie, PSL Research University, Paris, France
| | - Tanja Fehm
- Department of Gynecology and Obstetrics, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | | | - Dimitrios Mavroudis
- Laboratory of Translational Oncology, School of Medicine, University of Crete, Heraklion, Greece
- Department of Medical Oncology, University Hospital of Heraklion, Greece
| | | | - Justin Stebbing
- Division of Cancer, Department of Surgery and Cancer, Imperial College London, London, UK
| | - Paola Gazzaniga
- Department of Molecular Medicine, Sapienza University of Rome, Rome, Italy
| | | | - Rita Zamarchi
- Veneto Institute of Oncology IOV-IRCCS, Padua, Italy
| | - María Luisa Antelo
- Department of Hematology, Complejo Hospitalario de Navarra, Pamplona, Spain
| | - Leticia De Mattos-Arruda
- Val d’Hebron Institute of Oncology, Val d’Hebron University Hospital, and Universitat Autònoma de Barcelona, Barcelona, Spain
| | | | - Carlos Caldas
- Cancer Research UK Cambridge Institute and Department of Oncology Li Ka Shing Centre, University of Cambridge, Cambridge, UK
| | - Elisabetta Munzone
- Division of Medical Senology, European Institute of Oncology, IRCCS, Milano, Italy
| | - Luc Dirix
- Translational Cancer Research Unit, GZA Hospitals Sint-Augustinus, Antwerp, Belgium
- University of Antwerp, Antwerp, Belgium
| | - Amy L Delson
- Breast Science Advocacy Group, University of California San Francisco, San Francisco, CA, USA
| | | | - Misbah Qadir
- UNC Lineberger Comprehensive Cancer Center, Chapel Hill, NC, USA
| | - Cynthia Ma
- Washington University School of Medicine, St. Louis, MO, USA
| | - Janet H Scott
- Division of Hematology Oncology, University of California San Francisco, San Francisco, CA, USA
| | | | - John W Park
- Division of Hematology Oncology, University of California San Francisco, San Francisco, CA, USA
| | - Hope S Rugo
- Division of Hematology Oncology, University of California San Francisco, San Francisco, CA, USA
| |
Collapse
|
35
|
Galardi F, De Luca F, Biagioni C, Migliaccio I, Curigliano G, Minisini AM, Bonechi M, Moretti E, Risi E, McCartney A, Benelli M, Romagnoli D, Cappadona S, Gabellini S, Guarducci C, Conti V, Biganzoli L, Di Leo A, Malorni L. Circulating tumor cells and palbociclib treatment in patients with ER-positive, HER2-negative advanced breast cancer: results from a translational sub-study of the TREnd trial. Breast Cancer Res 2021; 23:38. [PMID: 33761970 PMCID: PMC7992319 DOI: 10.1186/s13058-021-01415-w] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2020] [Accepted: 03/08/2021] [Indexed: 12/24/2022] Open
Abstract
Background Circulating tumor cells (CTCs) are prognostic in patients with advanced breast cancer (ABC). However, no data exist about their use in patients treated with palbociclib. We analyzed the prognostic role of CTC counts in patients enrolled in the cTREnd study, a pre-planned translational sub-study of TREnd (NCT02549430), that randomized patients with ABC to palbociclib alone or palbociclib plus the endocrine therapy received in the prior line of treatment. Moreover, we evaluated RB1 gene expression on CTCs and explored its prognostic role within the cTREnd subpopulation. Methods Forty-six patients with ER-positive, HER2-negative ABC were analyzed. Blood samples were collected before starting palbociclib treatment (timepoint T0), after the first cycle of treatment (timepoint T1), and at disease progression (timepoint T2). CTCs were isolated and counted by CellSearch® System using the CellSearch™Epithelial Cell kit. Progression-free survival (PFS), clinical benefit (CB) during study treatment, and time to treatment failure (TTF) after study treatment were correlated with CTC counts. Samples with ≥ 5 CTCs were sorted by DEPArray system® (DA). RB1 and GAPDH gene expression levels were measured by ddPCR. Results All 46 patients were suitable for CTCs analysis. CTC count at T0 did not show significant prognostic value in terms of PFS and CB. Patients with at least one detectable CTC at T1 (n = 26) had a worse PFS than those with 0 CTCs (n = 16) (p = 0.02). At T1, patients with an increase of at least three CTCs showed reduced PFS compared to those with no increase (mPFS = 3 versus 9 months, (p = 0.004). Finally, patients with ≥ 5 CTCs at T2 (n = 6/23) who received chemotherapy as post-study treatment had a shorter TTF (p = 0.02). Gene expression data for RB1 were obtained from 19 patients. CTCs showed heterogeneous RB1 expression. Patients with detectable expression of RB1 at any timepoint showed better, but not statistically significant, outcomes than those with undetectable levels. Conclusions CTC count seems to be a promising modality in monitoring palbociclib response. Moreover, CTC count at the time of progression could predict clinical outcome post-palbociclib. RB1 expression analysis on CTCs is feasible and may provide additional prognostic information. Results should be interpreted with caution given the small studied sample size. Supplementary Information The online version contains supplementary material available at 10.1186/s13058-021-01415-w.
Collapse
Affiliation(s)
- Francesca Galardi
- "Sandro Pitigliani" Translational Research Unit, Hospital of Prato, Prato, Italy
| | - Francesca De Luca
- "Sandro Pitigliani" Translational Research Unit, Hospital of Prato, Prato, Italy
| | | | - Ilenia Migliaccio
- "Sandro Pitigliani" Translational Research Unit, Hospital of Prato, Prato, Italy
| | - Giuseppe Curigliano
- Division of Early Drug Development, Istituto Europeo di Oncologia, IRCCS, Milan, Italy.,Department of Haematology and Haemato-Oncology, University of Milan, Milan, Italy
| | - Alessandro M Minisini
- Department of Oncology, Azienda Sanitaria Universitaria del Friuli Centrale, Udine, Italy
| | - Martina Bonechi
- "Sandro Pitigliani" Translational Research Unit, Hospital of Prato, Prato, Italy
| | - Erica Moretti
- "Sandro Pitigliani" Medical Oncology Department, Hospital of Prato, Prato, Italy
| | - Emanuela Risi
- "Sandro Pitigliani" Medical Oncology Department, Hospital of Prato, Prato, Italy
| | - Amelia McCartney
- "Sandro Pitigliani" Medical Oncology Department, Hospital of Prato, Prato, Italy.,School of Clinical Sciences, Monash University, Melbourne, Australia
| | | | | | - Silvia Cappadona
- "Sandro Pitigliani" Medical Oncology Department, Hospital of Prato, Prato, Italy
| | - Stefano Gabellini
- "Sandro Pitigliani" Medical Oncology Department, Hospital of Prato, Prato, Italy
| | - Cristina Guarducci
- "Sandro Pitigliani" Translational Research Unit, Hospital of Prato, Prato, Italy.,Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, USA
| | - Valerio Conti
- Pediatric Neurology, Neurogenetics and Neurobiology Unit and Laboratories, Children's Hospital A. Meyer-University of Florence, Florence, Italy
| | - Laura Biganzoli
- "Sandro Pitigliani" Medical Oncology Department, Hospital of Prato, Prato, Italy
| | - Angelo Di Leo
- "Sandro Pitigliani" Medical Oncology Department, Hospital of Prato, Prato, Italy
| | - Luca Malorni
- "Sandro Pitigliani" Translational Research Unit, Hospital of Prato, Prato, Italy. .,"Sandro Pitigliani" Medical Oncology Department, Hospital of Prato, Prato, Italy.
| |
Collapse
|
36
|
Pinzani P, D'Argenio V, Del Re M, Pellegrini C, Cucchiara F, Salvianti F, Galbiati S. Updates on liquid biopsy: current trends and future perspectives for clinical application in solid tumors. Clin Chem Lab Med 2021; 59:1181-1200. [PMID: 33544478 DOI: 10.1515/cclm-2020-1685] [Citation(s) in RCA: 60] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2020] [Accepted: 01/20/2021] [Indexed: 01/19/2023]
Abstract
Despite advances in screening and therapeutics cancer continues to be one of the major causes of morbidity and mortality worldwide. The molecular profile of tumor is routinely assessed by surgical or bioptic samples, however, genotyping of tissue has inherent limitations: it represents a single snapshot in time and it is subjected to spatial selection bias owing to tumor heterogeneity. Liquid biopsy has emerged as a novel, non-invasive opportunity of detecting and monitoring cancer in several body fluids instead of tumor tissue. Circulating tumor cells (CTCs), circulating tumor DNA (ctDNA), RNA (mRNA and microRNA), microvesicles, including exosomes and tumor "educated platelets" were recently identified as a source of genomic information in cancer patients which could reflect all subclones present in primary and metastatic lesions allowing sequential monitoring of disease evolution. In this review, we summarize the currently available information concerning liquid biopsy in breast cancer, colon cancer, lung cancer and melanoma. These promising issues still need to be standardized and harmonized across laboratories, before fully adopting liquid biopsy approaches into clinical practice.
Collapse
Affiliation(s)
- Pamela Pinzani
- Department of Experimental and Clinical Biomedical Sciences "Mario Serio", University of Florence, Florence, Italy
| | - Valeria D'Argenio
- Department of Human Sciences and Quality of Life Promotion, San Raffaele Open University, Rome, Italy.,CEINGE-Biotecnologie Avanzate, Naples, Italy
| | - Marzia Del Re
- Clinical Pharmacology and Pharmacogenetics Unit, Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Cristina Pellegrini
- Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, L'Aquila, Italy
| | - Federico Cucchiara
- Clinical Pharmacology and Pharmacogenetics Unit, Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Francesca Salvianti
- Department of Experimental and Clinical Biomedical Sciences "Mario Serio", University of Florence, Florence, Italy
| | - Silvia Galbiati
- Diabetes Research Institute, IRCCS Ospedale San Raffaele, Milan, Italy
| |
Collapse
|
37
|
Polioudaki H, Mala A, Gkimprixi E, Papadaki MA, Chantziou A, Tzardi M, Mavroudis D, Agelaki S, Theodoropoulos PA. Epithelial/Mesenchymal Characteristics and PD-L1 Co-Expression in CTCs of Metastatic Breast Cancer Patients Treated with Eribulin: Correlation with Clinical Outcome. Cancers (Basel) 2020; 12:cancers12123735. [PMID: 33322610 PMCID: PMC7764288 DOI: 10.3390/cancers12123735] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2020] [Revised: 12/01/2020] [Accepted: 12/08/2020] [Indexed: 02/06/2023] Open
Abstract
We aimed to evaluate the co-expression of PD-L1 and epithelial-mesenchymal markers in CTCs from metastatic breast cancer (MBC) patients and to determine if there is any relationship with patients' outcome after eribulin treatment. Using cytospin preparations of peripheral blood mononuclear cells (PBMCs) from MBC patients treated with eribulin and a combination of immunocytochemistry and immunofluorescence, we quantified PD-L1, keratins and vimentin in single and cluster CTCs on days 1 and 8 of the first-treatment cycle. CTCs (n = 173) were found in 31 out of 38 patients. At baseline, the presence of cluster CTCs (p = 0.048), cluster mesenchymal CTCs (mCTCs) (p = 0.0003) or cluster PD-L1+mCTCs (p = 0.006) was associated with shorter overall survival (OS). In multivariate cox regression analysis, the detection of cluster mCTCs was the only parameter associated with increased risk of death (p = 0.024). On day 8 post-eribulin administration, PD-L1+mCTCs and especially single PD-L1+mCTCs decreased in 75% and 89% of patients, respectively. The detection of single PD-L1+mCTCs after eribulin treatment was correlated with shorter PFS (p = 0.047) and OS (p = 0.020). In conclusion, our study identified for the first time that cluster and single PD-L1+mCTCs subpopulations are of clinical significance in patients with MBC and highlighted the importance of CTC phenotyping during treatment with eribulin.
Collapse
Affiliation(s)
- Hara Polioudaki
- Laboratory of Biochemistry, School of Medicine, University of Crete, 71003 Heraklion, Crete, Greece; (H.P.); (E.G.); (A.C.)
| | - Anastasia Mala
- Department of Medical Oncology, University General Hospital of Heraklion, 71110 Heraklion, Crete, Greece; (A.M.); (D.M.); (S.A.)
| | - Eleni Gkimprixi
- Laboratory of Biochemistry, School of Medicine, University of Crete, 71003 Heraklion, Crete, Greece; (H.P.); (E.G.); (A.C.)
| | - Maria A. Papadaki
- Laboratory of Translational Oncology, School of Medicine, University of Crete, 71003 Heraklion, Crete, Greece;
| | - Amanda Chantziou
- Laboratory of Biochemistry, School of Medicine, University of Crete, 71003 Heraklion, Crete, Greece; (H.P.); (E.G.); (A.C.)
| | - Maria Tzardi
- Department of Pathology, University General Hospital of Heraklion, 71110 Heraklion, Crete, Greece;
| | - Dimitris Mavroudis
- Department of Medical Oncology, University General Hospital of Heraklion, 71110 Heraklion, Crete, Greece; (A.M.); (D.M.); (S.A.)
- Laboratory of Translational Oncology, School of Medicine, University of Crete, 71003 Heraklion, Crete, Greece;
| | - Sofia Agelaki
- Department of Medical Oncology, University General Hospital of Heraklion, 71110 Heraklion, Crete, Greece; (A.M.); (D.M.); (S.A.)
- Laboratory of Translational Oncology, School of Medicine, University of Crete, 71003 Heraklion, Crete, Greece;
| | - Panayiotis A. Theodoropoulos
- Laboratory of Biochemistry, School of Medicine, University of Crete, 71003 Heraklion, Crete, Greece; (H.P.); (E.G.); (A.C.)
- Correspondence:
| |
Collapse
|
38
|
Magbanua MJM, Savenkov O, Asmus EJ, Ballman KV, Scott JH, Park JW, Dickler M, Partridge A, Carey LA, Winer EP, Rugo HS. Clinical Significance of Circulating Tumor Cells in Hormone Receptor-positive Metastatic Breast Cancer Patients who Received Letrozole with or Without Bevacizumab. Clin Cancer Res 2020; 26:4911-4920. [PMID: 32586939 DOI: 10.1158/1078-0432.ccr-20-1329] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2020] [Revised: 06/08/2020] [Accepted: 06/19/2020] [Indexed: 11/16/2022]
Abstract
PURPOSE We evaluated the prognostic and predictive value of circulating tumor cells (CTCs) hormone receptor-positive (HR+) metastatic breast cancer (MBC) patients randomized to letrozole alone or letrozole plus bevacizumab in the first-line setting (CALGB 40503). EXPERIMENTAL DESIGN Blood samples were collected at pretreatment and three additional time points during therapy. The presence of ≥5 CTCs per 7.5 mL of blood was considered CTC positive. Association of CTCs with progression-free survival (PFS) and overall survival (OS) was assessed using Cox regression models. RESULTS Of 343 patients treated, 294 had CTC data and were included in this analysis. Median follow-up was 39 months. In multivariable analysis, CTC-positive patients at baseline (31%) had significantly reduced PFS [HR, 1.49; 95% confidence interval (CI), 1.12-1.97] and OS (HR, 2.08; 95% CI, 1.49-2.93) compared with CTC negative. Failure to clear CTCs during treatment was associated with significantly increased risk of progression (HR, 2.2; 95% CI, 1.58-3.07) and death (HR, 3.4; 95% CI, 2.36-4.88). CTC-positive patients who received only letrozole had the worse PFS (HR, 2.3; 95% CI, 1.54-3.47) and OS (HR, 2.6; 95% CI, 1.59-4.40). Median PFS in CTC-positive patients was significantly longer (18.0 vs. 7.0 months) in letrozole plus bevacizumab versus letrozole arm (P = 0.0009). Restricted mean survival time analysis further revealed that addition of bevacizumab was associated with PFS benefit in both CTC-positive and CTC-negative patients, but OS benefit was only observed in CTC-positive patients. CONCLUSIONS CTCs were highly prognostic for the addition of bevacizumab to first-line letrozole in patients with HR+ MBC in CALGB 40503. Further research to determine the potential predictive value of CTCs in this setting is warranted.
Collapse
Affiliation(s)
| | | | - Erik J Asmus
- Alliance Statistics and Data Center, Mayo Clinic, Rochester, Minnesota
| | - Karla V Ballman
- Alliance Statistics and Data Center, Weill Medical College of Cornell University, New York, New York
| | - Janet H Scott
- University of California at San Francisco, San Francisco, California
| | - John W Park
- University of California at San Francisco, San Francisco, California
| | | | - Ann Partridge
- Dana-Farber/Partners CancerCare, Boston, Massachusetts
| | - Lisa A Carey
- UNC Lineberger Comprehensive Cancer Center, Chapel Hill, North Carolina
| | - Eric P Winer
- Dana-Farber/Partners CancerCare, Boston, Massachusetts
| | - Hope S Rugo
- University of California at San Francisco, San Francisco, California
| |
Collapse
|
39
|
Cayrefourcq L, Alix-Panabières C. Clinical relevance of liquid biopsy in breast cancer: update in 2020. Expert Rev Mol Diagn 2020; 20:913-919. [DOI: 10.1080/14737159.2020.1816828] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Affiliation(s)
- Laure Cayrefourcq
- Laboratory of Rare Human Circulating Cells (LCCRH), University Medical Centre of Montpellier, Montpellier, France
| | - Catherine Alix-Panabières
- Laboratory of Rare Human Circulating Cells (LCCRH), University Medical Centre of Montpellier, Montpellier, France
| |
Collapse
|
40
|
Rasheduzzaman M, Kulasinghe A, Dolcetti R, Kenny L, Johnson NW, Kolarich D, Punyadeera C. Protein glycosylation in head and neck cancers: From diagnosis to treatment. Biochim Biophys Acta Rev Cancer 2020; 1874:188422. [PMID: 32853734 DOI: 10.1016/j.bbcan.2020.188422] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2020] [Revised: 08/20/2020] [Accepted: 08/21/2020] [Indexed: 12/24/2022]
Abstract
Glycosylation is the most common post-translational modification (PTM) of proteins. Malignant tumour cells frequently undergo an alteration in surface protein glycosylation. This phenomenon is also common in cancers of the head and neck, most of which are squamous cell carcinomas (HNSCC). It affects cell functions, including proliferation, motility and invasiveness, thus increasing the propensity to metastasise. HNSCC represents the sixth most frequent malignancy worldwide. These neoplasms, which arise from the mucous membranes of the various anatomical subsites of the upper aero-digestive tract, are heterogeneous in terms of aetiology and clinico-pathologic features. With current treatments, only about 50% of HNSCC patients survive beyond 5-years. Therefore, there is the pressing need to dissect NHSCC heterogeneity to inform treatment choices. In particular, reliable biomarkers of predictive and prognostic value are eagerly needed. This review describes the current state of the art and bio-pathological meaning of glycosylation signatures associated with HNSCC and explores the possible role of tumour specific glycoproteins as potential biomarkers and attractive therapeutic targets. We have also compiled data relating to altered glycosylation and the nature of glycoproteins as tools for the identification of circulating tumour cells (CTCs) in the new era of liquid biopsy.
Collapse
Affiliation(s)
- Mohammad Rasheduzzaman
- Saliva and Liquid Biopsy Translational Laboratory, The School of Biomedical Sciences, Institute of Health and Biomedical Innovation, Queensland University of Technology, Kelvin Grove, QLD, Australia; Translational Research Institute, Woolloongabba, QLD, Australia
| | - Arutha Kulasinghe
- Saliva and Liquid Biopsy Translational Laboratory, The School of Biomedical Sciences, Institute of Health and Biomedical Innovation, Queensland University of Technology, Kelvin Grove, QLD, Australia; Translational Research Institute, Woolloongabba, QLD, Australia
| | - Riccardo Dolcetti
- Translational Research Institute, Woolloongabba, QLD, Australia.; The University of Queensland Diamantina Institute, 37 Kent Street Woolloongabba, QLD 4102, Australia
| | - Liz Kenny
- Department of Radiation Oncology, Cancer Care Services, Royal Brisbane and Women's Hospital, Joyce Tweddell Building, Herston, QLD, 4029, Australia
| | - Newell W Johnson
- Menzies Health Institute Queensland, Griffith University, Southport, Queensland, Australia; Faculty of Dentistry, Oral and Craniofacial Sciences, King's College, London, United Kingdom
| | - Daniel Kolarich
- Institute for Glycomics, Griffith University, Gold Coast, QLD, Australia; ARC Centre of Excellence for Nanoscale BioPhotonics, Griffith University, QLD, Australia.
| | - Chamindie Punyadeera
- Saliva and Liquid Biopsy Translational Laboratory, The School of Biomedical Sciences, Institute of Health and Biomedical Innovation, Queensland University of Technology, Kelvin Grove, QLD, Australia; Translational Research Institute, Woolloongabba, QLD, Australia..
| |
Collapse
|
41
|
Narbe U, Bendahl PO, Aaltonen K, Fernö M, Forsare C, Jørgensen CLT, Larsson AM, Rydén L. The Distribution of Circulating Tumor Cells Is Different in Metastatic Lobular Compared to Ductal Carcinoma of the Breast-Long-Term Prognostic Significance. Cells 2020; 9:E1718. [PMID: 32709042 PMCID: PMC7407940 DOI: 10.3390/cells9071718] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2020] [Accepted: 07/13/2020] [Indexed: 12/18/2022] Open
Abstract
BACKGROUND Invasive lobular carcinoma (ILC) has distinguishing features when compared to invasive ductal carcinoma of no special type (NST). In this study, we explored the distributional and prognostic characteristics of circulating tumor cells (CTCs) in metastatic ILC and NST. MATERIALS AND METHODS Patients were included in an observational trial (ClinicalTrials.gov NCT01322893) with ILC (n = 28) and NST (n = 111). CTC count (number/7.5 mL blood) was evaluated with serial sampling (CellSearch). The primary endpoint was progression-free survival (PFS). RESULTS The CTC counts were higher in ILC (median 70) than in NST cases (median 2) at baseline (p < 0.001). The evidence for ≥5 CTCs as a prognostic factor for PFS in ILC was weak, but stronger with higher cut-offs (CTC ≥ 20: hazard ratio (HR) 3.0, p = 0.01) (CTC ≥ 80: HR 3.6, p = 0.004). In NST, however, the prognostic effect of CTCs ≥5 was strong. Decline in CTC count from baseline to three months was associated with improved prognosis in ILC and NST. CONCLUSIONS The number of CTCs is higher in ILC than in NST, implying that a higher CTC cut-off could be considered for ILC when applying the CellSearch technique.
Collapse
Affiliation(s)
- Ulrik Narbe
- Department of Clinical Sciences, Division of Oncology, Lund University, SE-223 81 Lund, Sweden; (U.N.); (P.-O.B.); (M.F.); (C.F.); (C.L.T.J.); (A.-M.L.)
- Department of Oncology, Växjö Central Hospital, SE-352 34 Växjö, Sweden
| | - Pär-Ola Bendahl
- Department of Clinical Sciences, Division of Oncology, Lund University, SE-223 81 Lund, Sweden; (U.N.); (P.-O.B.); (M.F.); (C.F.); (C.L.T.J.); (A.-M.L.)
| | - Kristina Aaltonen
- Department of Laboratory Medicine, Division of Translational Cancer Research, Lund University, SE-223 81 Lund, Sweden;
| | - Mårten Fernö
- Department of Clinical Sciences, Division of Oncology, Lund University, SE-223 81 Lund, Sweden; (U.N.); (P.-O.B.); (M.F.); (C.F.); (C.L.T.J.); (A.-M.L.)
| | - Carina Forsare
- Department of Clinical Sciences, Division of Oncology, Lund University, SE-223 81 Lund, Sweden; (U.N.); (P.-O.B.); (M.F.); (C.F.); (C.L.T.J.); (A.-M.L.)
| | - Charlotte Levin Tykjær Jørgensen
- Department of Clinical Sciences, Division of Oncology, Lund University, SE-223 81 Lund, Sweden; (U.N.); (P.-O.B.); (M.F.); (C.F.); (C.L.T.J.); (A.-M.L.)
| | - Anna-Maria Larsson
- Department of Clinical Sciences, Division of Oncology, Lund University, SE-223 81 Lund, Sweden; (U.N.); (P.-O.B.); (M.F.); (C.F.); (C.L.T.J.); (A.-M.L.)
- Department of Hematology, Oncology and Radiation Physics, Skåne University Hospital, SE-221 85 Lund, Sweden
| | - Lisa Rydén
- Department of Clinical Sciences, Division of Surgery, Lund University, SE-223 81 Lund, Sweden
- Department of Surgery, Skåne University Hospital, SE-221 85 Lund, Sweden
| |
Collapse
|
42
|
Cerebrospinal fluid circulating tumor cells as a quantifiable measurement of leptomeningeal metastases in patients with HER2 positive cancer. J Neurooncol 2020; 148:599-606. [PMID: 32506369 PMCID: PMC7438284 DOI: 10.1007/s11060-020-03555-z] [Citation(s) in RCA: 43] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2020] [Accepted: 06/02/2020] [Indexed: 02/05/2023]
Abstract
PURPOSE The CellSearch® system has been used to identify circulating tumor cells (CTCs) in cerebrospinal fluid (CSF) to diagnose leptomeningeal metastasis (LM) in patients with epithelial cancers. Using this system, we prospectively explored sequential CSF CTC enumeration in patients with LM from HER2+ cancers receiving intrathecal (IT) trastuzumab to capture dynamic changes in CSF CTC enumeration. METHODS CSF from patients enrolled in an IRB-approved phase I/II dose escalation trial of IT trastuzumab for LM in HER2+ cancer (NCT01325207) was obtained on day 1 of each cycle and was evaluated by the CellSearch® platform for CTC enumeration. The results were correlated with CSF cytology from the same sample, along with clinical and radiographic response. RESULTS Fifteen out of 34 patients with HER2+ LM were enrolled in CSF CTC analysis; 14 were women. Radiographic LM was documented in 14 (93%) patients; CSF cytology was positive in 6 (40%) and CSF CTCs were identified in 13 (87%). Median CSF CTC was 22 CTCs (range 0-200 +) per 3 ml. HER2/neu expression analysis of CTCs was performed in 8 patients; 75% had confirmed expression of HER2/neu positivity in CSF and HER2/neu expression was absent in 25%. Four of 10 patients received 7 or more cycles of IT trastuzumab; in 3 of these patients, increase in CSF CTCs enumeration from baseline was detected 2-3 months prior to changes seen on MRI, and while CSF cytology remained negative. CONCLUSION Our study demonstrates that enumeration of CSF CTCs may provide dynamic, quantitative assessment of tumor burden in the central nervous system compartment during treatment for LM and prior to changes on MRI or CSF cytology. TRIAL REGISTRATION Clinicaltrials.gov: NCT01325207; registered March 29th, 2011.
Collapse
|
43
|
Cui Z, Su F, Li Y, Yang D. Circulating tumour cells as prognosis predictive markers of neoadjuvant chemotherapy-treated breast cancer patients. J Chemother 2020; 32:304-309. [PMID: 32500819 DOI: 10.1080/1120009x.2020.1774207] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
In this study, we detected and measured the count of circulating tumour cells (CTCs) in breast cancer (BC) patients who were treated by neoadjuvant chemotherapy (NAC) in order to assess the clinical validity of CTCs. A total of 96 patients with locally advanced BC and who were treated by NAC were enrolled in this study. The CTC count in the peripheral blood was estimated by negative enrichment-fluorescence in situ hybridization before and after NAC. The clinicopathological data of the patients were recorded. CTCs were detected in 59 of the 96 patients with BC before NAC. Particularly, the detection rate of CTCs was significantly lower in human epidermal growth factor receptor-2 (HER-2)-negative patients than in HER-2-positive patients. CTCs were significantly fewer after NAC than before NAC. The CTC-detection sensitivity in the NAC efficacy evaluation was 75.5% (40/53), while the specificity was 72.1% (31/43). The CTC consistency analysis with clinical effects (Response Evaluation Criteria in Solid Tumors Version 1.1 Standard) was described as moderate (kappa = 0.476, P < 0.001). Thus, our findings suggest that CTC detection is a potential new approach to assess the efficacy of NAC.
Collapse
Affiliation(s)
- Zhaoqing Cui
- Department of Breast and Thyroid Surgery, Liaocheng People's Hospital, Liaocheng, Shandong, P. R. China
| | - Fengbo Su
- Department of Surgery, Shouguang Yingli Central Hospital, Shouguang, Shandong, P. R. China
| | - Yang Li
- Zhong Yuan Academy of Biological Medicine, Liaocheng People's Hospital, Liaocheng, Shandong, P. R. China
| | - Dawei Yang
- Zhong Yuan Academy of Biological Medicine, Liaocheng People's Hospital, Liaocheng, Shandong, P. R. China
| |
Collapse
|
44
|
Chelakkot C, Ryu J, Kim MY, Kim JS, Kim D, Hwang J, Park SH, Ko SB, Park JW, Jung MY, Kim RN, Song K, Kim YJ, Choi YL, Lee HS, Shin YK. An Immune-Magnetophoretic Device for the Selective and Precise Enrichment of Circulating Tumor Cells from Whole Blood. MICROMACHINES 2020; 11:mi11060560. [PMID: 32486306 PMCID: PMC7345362 DOI: 10.3390/mi11060560] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/30/2020] [Revised: 05/22/2020] [Accepted: 05/26/2020] [Indexed: 02/06/2023]
Abstract
Here, we validated the clinical utility of our previously developed microfluidic device, GenoCTC, which is based on bottom magnetophoresis, for the isolation of circulating tumor cells (CTCs) from patient whole blood. GenoCTC allowed 90% purity, 77% separation rate, and 80% recovery of circulating tumor cells at a 90 μL/min flow rate when tested on blood spiked with epithelial cell adhesion molecule (EpCAM)-positive Michigan Cancer Foundation-7 (MCF7) cells. Clinical studies were performed using blood samples from non-small cell lung cancer (NSCLC) patients. Varying numbers (2 to 114) of CTCs were found in each NSCLC patient, and serial assessment of CTCs showed that the CTC count correlated with the clinical progression of the disease. The applicability of GenoCTC to different cell surface biomarkers was also validated in a cholangiocarcinoma patient using anti-EPCAM, anti-vimentin, or anti-tyrosine protein kinase MET (c-MET) antibodies. After EPCAM-, vimentin-, or c-MET-positive cells were isolated, CTCs were identified and enumerated by immunocytochemistry using anti-cytokeratin 18 (CK18) and anti-CD45 antibodies. Furthermore, we checked the protein expression of PDL1 and c-MET in CTCs. A study in a cholangiocarcinoma patient showed that the number of CTCs varied depending on the biomarker used, indicating the importance of using multiple biomarkers for CTC isolation and enumeration.
Collapse
Affiliation(s)
- Chaithanya Chelakkot
- Technical Research Center, Genobio Corp., Seoul 08394, Korea; (C.C.); (J.R.); (D.K.); (J.H.); (S.H.P.); (S.B.K.)
| | - Jiyeon Ryu
- Technical Research Center, Genobio Corp., Seoul 08394, Korea; (C.C.); (J.R.); (D.K.); (J.H.); (S.H.P.); (S.B.K.)
| | - Mi Young Kim
- Department of Internal Medicine, Seoul National University Boramae Medical Center, Seoul 07061, Korea; (M.Y.K.); (J.-S.K.)
| | - Jin-Soo Kim
- Department of Internal Medicine, Seoul National University Boramae Medical Center, Seoul 07061, Korea; (M.Y.K.); (J.-S.K.)
| | - Dohyeong Kim
- Technical Research Center, Genobio Corp., Seoul 08394, Korea; (C.C.); (J.R.); (D.K.); (J.H.); (S.H.P.); (S.B.K.)
| | - Juhyun Hwang
- Technical Research Center, Genobio Corp., Seoul 08394, Korea; (C.C.); (J.R.); (D.K.); (J.H.); (S.H.P.); (S.B.K.)
| | - Sung Hoon Park
- Technical Research Center, Genobio Corp., Seoul 08394, Korea; (C.C.); (J.R.); (D.K.); (J.H.); (S.H.P.); (S.B.K.)
| | - Seok Bum Ko
- Technical Research Center, Genobio Corp., Seoul 08394, Korea; (C.C.); (J.R.); (D.K.); (J.H.); (S.H.P.); (S.B.K.)
| | - Jeong Won Park
- IT Convergence Technology Research Laboratory, Electronic and Telecommunications Research Institute, Daejon 34129, Korea; (J.W.P.); (M.Y.J.)
| | - Moon Youn Jung
- IT Convergence Technology Research Laboratory, Electronic and Telecommunications Research Institute, Daejon 34129, Korea; (J.W.P.); (M.Y.J.)
| | - Ryong Nam Kim
- Bio-MAX/N-Bio, Seoul National University, Seoul 08826, Korea;
| | - Kyoung Song
- The Center for Companion Diagnostics, LOGONE Bio Convergence Research Foundation, Seoul 08394, Korea; (K.S.); (Y.J.K.)
| | - Yu Jin Kim
- The Center for Companion Diagnostics, LOGONE Bio Convergence Research Foundation, Seoul 08394, Korea; (K.S.); (Y.J.K.)
| | - Yoon-La Choi
- Laboratory of Cancer Genomics and Molecular Pathology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul 08394, Korea;
- Department of Pathology and Translational Genomics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul 06351, Korea
| | - Hun Seok Lee
- Technical Research Center, Genobio Corp., Seoul 08394, Korea; (C.C.); (J.R.); (D.K.); (J.H.); (S.H.P.); (S.B.K.)
- Correspondence: (H.S.L.); (Y.K.S.)
| | - Young Kee Shin
- Laboratory of Molecular Pathology and Cancer Genomics, College of Pharmacy and Research Institute of Pharmaceutical Science, Seoul National University, Seoul 08826, Korea
- Department of Molecular Medicine and Biopharmaceutical Sciences, Graduate School of Convergence Science and Technology, Seoul National University, Seoul 08826, Korea
- The Center for Anti-Cancer Companion Diagnostics, Bio-MAX/N-Bio, Seoul National University, Seoul 08826, Korea
- Correspondence: (H.S.L.); (Y.K.S.)
| |
Collapse
|
45
|
Costa C, Muinelo-Romay L, Cebey-López V, Pereira-Veiga T, Martínez-Pena I, Abreu M, Abalo A, Lago-Lestón RM, Abuín C, Palacios P, Cueva J, Piñeiro R, López-López R. Analysis of a Real-World Cohort of Metastatic Breast Cancer Patients Shows Circulating Tumor Cell Clusters (CTC-clusters) as Predictors of Patient Outcomes. Cancers (Basel) 2020; 12:cancers12051111. [PMID: 32365530 PMCID: PMC7281711 DOI: 10.3390/cancers12051111] [Citation(s) in RCA: 47] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2020] [Revised: 04/22/2020] [Accepted: 04/27/2020] [Indexed: 01/24/2023] Open
Abstract
Circulating tumor cell (CTC) enumeration has emerged as a powerful biomarker for the assessment of prognosis and the response to treatment in metastatic breast cancer (MBC). Moreover, clinical evidences show that CTC-cluster counts add prognostic information to CTC enumeration, however, their significance is not well understood, and more clinical evidences are needed. We aim to evaluate the prognostic value of longitudinally collected single CTCs and CTC-clusters in a heterogeneous real-world cohort of 54 MBC patients. Blood samples were longitudinally collected at baseline and follow up. CTC and CTC-cluster enumeration was performed using the CellSearch® system. Associations with progression-free survival (PFS) and overall survival (OS) were evaluated using Cox proportional hazards modelling. Elevated CTC counts and CTC-clusters at baseline were significantly associated with a shorter survival time. In joint analysis, patients with high CTC counts and CTC-cluster at baseline were at a higher risk of progression and death, and longitudinal analysis showed that patients with CTC-clusters had significantly shorter survival compared to patients without clusters. Moreover, patients with CTC-cluster of a larger size were at a higher risk of death. A longitudinal analysis of a real-world cohort of MBC patients indicates that CTC-clusters analysis provides additional prognostic value to single CTC enumeration, and that CTC-cluster size correlates with patient outcome.
Collapse
Affiliation(s)
- Clotilde Costa
- Roche-Chus Joint Unit, Translational Medical Oncology Group, Oncomet, Health Research Institute of Santiago de Compostela (IDIS), Travesía da Choupana s/n, 15706 Santiago de Compostela, Spain; (C.C.); (T.P.-V.); (I.M.-P.); (C.A.); (R.L.-L.)
- CIBERONC, Centro de Investigación Biomédica en Red Cáncer, 28029 Madrid, Spain;
| | - Laura Muinelo-Romay
- CIBERONC, Centro de Investigación Biomédica en Red Cáncer, 28029 Madrid, Spain;
- Liquid Biopsy Analysis Unit, Translational Medical Oncology Group, Health Research Institute of Santiago de Santiago de Compostela (IDIS), Travesía da Choupana s/n, 15706 Santiago de Compostela, Spain; (M.A.); (A.A.); (R.M.L.-L.)
| | - Victor Cebey-López
- Department of Oncology, Complexo Hospitalario Universitario de Santiago de Compostela (SERGAS), 15706 Santiago de Compostela, Spain; (V.C.-L.); (P.P.); (J.C.)
| | - Thais Pereira-Veiga
- Roche-Chus Joint Unit, Translational Medical Oncology Group, Oncomet, Health Research Institute of Santiago de Compostela (IDIS), Travesía da Choupana s/n, 15706 Santiago de Compostela, Spain; (C.C.); (T.P.-V.); (I.M.-P.); (C.A.); (R.L.-L.)
| | - Inés Martínez-Pena
- Roche-Chus Joint Unit, Translational Medical Oncology Group, Oncomet, Health Research Institute of Santiago de Compostela (IDIS), Travesía da Choupana s/n, 15706 Santiago de Compostela, Spain; (C.C.); (T.P.-V.); (I.M.-P.); (C.A.); (R.L.-L.)
| | - Manuel Abreu
- Liquid Biopsy Analysis Unit, Translational Medical Oncology Group, Health Research Institute of Santiago de Santiago de Compostela (IDIS), Travesía da Choupana s/n, 15706 Santiago de Compostela, Spain; (M.A.); (A.A.); (R.M.L.-L.)
| | - Alicia Abalo
- Liquid Biopsy Analysis Unit, Translational Medical Oncology Group, Health Research Institute of Santiago de Santiago de Compostela (IDIS), Travesía da Choupana s/n, 15706 Santiago de Compostela, Spain; (M.A.); (A.A.); (R.M.L.-L.)
| | - Ramón M. Lago-Lestón
- Liquid Biopsy Analysis Unit, Translational Medical Oncology Group, Health Research Institute of Santiago de Santiago de Compostela (IDIS), Travesía da Choupana s/n, 15706 Santiago de Compostela, Spain; (M.A.); (A.A.); (R.M.L.-L.)
| | - Carmen Abuín
- Roche-Chus Joint Unit, Translational Medical Oncology Group, Oncomet, Health Research Institute of Santiago de Compostela (IDIS), Travesía da Choupana s/n, 15706 Santiago de Compostela, Spain; (C.C.); (T.P.-V.); (I.M.-P.); (C.A.); (R.L.-L.)
| | - Patricia Palacios
- Department of Oncology, Complexo Hospitalario Universitario de Santiago de Compostela (SERGAS), 15706 Santiago de Compostela, Spain; (V.C.-L.); (P.P.); (J.C.)
| | - Juan Cueva
- Department of Oncology, Complexo Hospitalario Universitario de Santiago de Compostela (SERGAS), 15706 Santiago de Compostela, Spain; (V.C.-L.); (P.P.); (J.C.)
| | - Roberto Piñeiro
- Roche-Chus Joint Unit, Translational Medical Oncology Group, Oncomet, Health Research Institute of Santiago de Compostela (IDIS), Travesía da Choupana s/n, 15706 Santiago de Compostela, Spain; (C.C.); (T.P.-V.); (I.M.-P.); (C.A.); (R.L.-L.)
- Correspondence: ; Tel.: +34-981-955-602
| | - Rafael López-López
- Roche-Chus Joint Unit, Translational Medical Oncology Group, Oncomet, Health Research Institute of Santiago de Compostela (IDIS), Travesía da Choupana s/n, 15706 Santiago de Compostela, Spain; (C.C.); (T.P.-V.); (I.M.-P.); (C.A.); (R.L.-L.)
- CIBERONC, Centro de Investigación Biomédica en Red Cáncer, 28029 Madrid, Spain;
- Liquid Biopsy Analysis Unit, Translational Medical Oncology Group, Health Research Institute of Santiago de Santiago de Compostela (IDIS), Travesía da Choupana s/n, 15706 Santiago de Compostela, Spain; (M.A.); (A.A.); (R.M.L.-L.)
- Department of Oncology, Complexo Hospitalario Universitario de Santiago de Compostela (SERGAS), 15706 Santiago de Compostela, Spain; (V.C.-L.); (P.P.); (J.C.)
| |
Collapse
|
46
|
Clinical Relevance and Therapeutic Application of CTCs in Advanced Breast Cancer. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2020. [PMID: 32304085 DOI: 10.1007/978-3-030-35805-1_10] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/30/2023]
Abstract
Precision medicine through liquid biopsy represents an emerging approach in the management of cancer. The CTC count in blood samples from patients with advanced breast cancer is a powerful prognostic factor for both progression free and overall survival. Moreover, high levels of CTCs at any time during the treatment can reliably predict progression before imaging studies and/or tumor markers. Furthermore, there are works on the molecular characterization of the CTCs and their potential ability to guide the treatment in a dynamic way. However, their role remains controversial. Detection and enumeration of CTCs is variable among different tumors and is subjected to biases related mainly to their methodology, which is not completely standardized. In addition, they must demonstrate their clinical value to guide the treatment and a translation on patient's survival.
Collapse
|
47
|
Dynamic changes of Receptor activator of nuclear factor-κB expression in Circulating Tumor Cells during Denosumab predict treatment effectiveness in Metastatic Breast Cancer. Sci Rep 2020; 10:1288. [PMID: 31992773 PMCID: PMC6987166 DOI: 10.1038/s41598-020-58339-2] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2019] [Accepted: 01/06/2020] [Indexed: 12/11/2022] Open
Abstract
Receptor-activator of nuclear-factor -κB-ligand (RANKL) and its receptor RANK have been recently identified as key players in breast cancer bone metastases. Since Circulating Tumor Cells (CTCs) are considered a crucial step of metastatic process, we explored RANK expression on CTCs in metastatic breast cancer (MBC), and the predictive value of RANK-positive CTCs in monitoring patients during treatment with denosumab (anti-RANKL antibody). To this purpose, we developed a novel CTC assay to quantify RANK-positive CTCs in forty-two bone MBC patients, candidates to denosumab treatment. Companion algorithms ΔAUC and Slope were developed, and correlated with time to first skeletal-related-events (SRE), time to bone metastasis progression and time to visceral metastasis progression. Twenty-seven patients had at least one CTC at baseline and, among these, nineteen (70%) had one or more RANK-positive CTCs. Notably, the baseline total CTCs, but not the RANK-positive, were associated with Time-to-first-SRE, Time-to-Bone-Metastasis-Progression and Time-to-Visceral-Metastasis-Progression. Conversely, during treatment monitoring, positive ΔAUC value, expression of RANK-positive CTCs persistence, correlated with longer Time-to-first-SRE (p = 0.0002) and Time-to-Bone-Metastasis-Progression (p = 0.0012). Furthermore, the early increase at second day, in RANK-positive CTCs (Positive-Slope) was associated with delay in time-to-first-SRE (p = 0.0038) and Time-to-Bone-Metastasis-Progression (p = 0.0024). We demonstrate, for the first time, the expression of RANK on CTCs in MBC patients and that the persistence of RANK expression determines denosumab effectiveness.
Collapse
|
48
|
Zhang Q, Wang W, Huang S, Yu S, Tan T, Zhang JR, Zhu JJ. Capture and selective release of multiple types of circulating tumor cells using smart DNAzyme probes. Chem Sci 2020; 11:1948-1956. [PMID: 34123289 PMCID: PMC8148068 DOI: 10.1039/c9sc04309h] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2019] [Accepted: 01/06/2020] [Indexed: 12/11/2022] Open
Abstract
The effective capture, release and reanalysis of circulating tumor cells (CTCs) are of great significance to acquire tumor information and promote the progress of tumor therapy. Particularly, the selective release of multiple types of CTCs is critical to further study; however, it is still a great challenge. To meet this challenge, we designed a smart DNAzyme probe-based platform. By combining multiple targeting aptamers and multiple metal ion responsive DNAzymes, efficient capture and selective release of multiple types CTCs were realized. Sgc8c aptamer integrated Cu2+-dependent DNAzyme and TD05 aptamer integrated Mg2+-dependent DNAzyme can capture CCRF-CEM cells and Ramos cells respectively on the substrate. With the addition of Cu2+ or Mg2+, CCRF-CEM cells or Ramos cells will be released from the substrate with specific selectivity. Furthermore, our platform has been successfully demonstrated in the whole blood sample. Therefore, our capture/release platform will benefit research on the molecular analysis of CTCs after release and has great potential for cancer diagnosis and individualized treatment.
Collapse
Affiliation(s)
- Qianying Zhang
- State Key Laboratory of Analytical Chemistry for Life Science, School of Chemistry and Chemical Engineering, Nanjing University Nanjing 210023 China
| | - Wenjing Wang
- State Key Laboratory of Agricultural Microbiology, College of Science, Huazhong Agricultural University Wuhan 430070 China
| | - Shan Huang
- State Key Laboratory of Analytical Chemistry for Life Science, School of Chemistry and Chemical Engineering, Nanjing University Nanjing 210023 China
| | - Sha Yu
- State Key Laboratory of Analytical Chemistry for Life Science, School of Chemistry and Chemical Engineering, Nanjing University Nanjing 210023 China
| | - Tingting Tan
- Department of Laboratory Medicine, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School Nanjing 210008 China
| | - Jian-Rong Zhang
- State Key Laboratory of Analytical Chemistry for Life Science, School of Chemistry and Chemical Engineering, Nanjing University Nanjing 210023 China
- School of Chemistry and Life Science, Nanjing University Jinling College Nanjing 210089 China
| | - Jun-Jie Zhu
- State Key Laboratory of Analytical Chemistry for Life Science, School of Chemistry and Chemical Engineering, Nanjing University Nanjing 210023 China
| |
Collapse
|
49
|
Introduction - Biology of Breast Cancer Metastasis and Importance of the Analysis of CTCs. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2020; 1220:1-10. [PMID: 32304076 DOI: 10.1007/978-3-030-35805-1_1] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Breast cancer metastasis is a complex multistep process during which tumor cells undergo structural and functional changes that allow them to move away from the primary tumor and disseminate to distant organs and tissues. Despite the inefficiency of this process, some populations of circulating tumor cells (CTCs), which are those cells responsible of metastases formation, are able to survive in blood circulation and grow into secondary tumors. Metastatic breast cancer remains an incurable disease, and the phenomenon of metastasis represents the larger cause of death in these patients. The application of liquid biopsy techniques and the advancements in the field have shown the prognostic value of CTCs, suggesting the importance that CTCs analyses may have in the clinic. However, their implementation in routine clinic has not been yet achieved due to the yet small body of evidence showing their clinical utility. This introductory chapter will revise the key aspects of breast cancer metastasis and discuss the importance of CTC analyses in the management of breast cancer patients.
Collapse
|
50
|
Cobain EF, Paoletti C, Smerage JB, Hayes DF. Clinical Applications of Circulating Tumor Cells in Breast Cancer. Recent Results Cancer Res 2020; 215:147-160. [PMID: 31605228 DOI: 10.1007/978-3-030-26439-0_8] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
The development of metastatic disease accounts for the vast majority of cancer-related deaths in solid tumor malignancies. Distant metastases primarily develop as a result of tumor cell dissemination through the circulatory system.
Collapse
Affiliation(s)
- Erin F Cobain
- Department of Internal Medicine, Breast Oncology Program of the Comprehensive Cancer Center, University of Michigan Health System, 1500 E. Medical Center Dr., Ann Arbor, MI, 48109-5942, USA
| | - Costanza Paoletti
- Department of Internal Medicine, Breast Oncology Program of the Comprehensive Cancer Center, University of Michigan Health System, 1500 E. Medical Center Dr., Ann Arbor, MI, 48109-5942, USA
| | - Jeffrey B Smerage
- Department of Internal Medicine, Breast Oncology Program of the Comprehensive Cancer Center, University of Michigan Health System, 1500 E. Medical Center Dr., Ann Arbor, MI, 48109-5942, USA
| | - Daniel F Hayes
- Department of Internal Medicine, Breast Oncology Program of the Comprehensive Cancer Center, University of Michigan Health System, 1500 E. Medical Center Dr., Ann Arbor, MI, 48109-5942, USA.
| |
Collapse
|