1
|
Yin H, Xin Y, Yang J, Luo Q, Yang M, Sun J, Wang Y, Wang Q, Kalvakolanu DV, Guo B, Jiang W, Zhang L. Multifunctional nanozymes: Promising applications in clinical diagnosis and cancer treatment. Biosens Bioelectron 2025; 279:117383. [PMID: 40121930 DOI: 10.1016/j.bios.2025.117383] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2024] [Revised: 02/09/2025] [Accepted: 03/14/2025] [Indexed: 03/25/2025]
Abstract
Cancer remains one of the greatest challenges in modern medicine. Traditional chemotherapy drugs often cause severe side effects, including nausea, vomiting, diarrhea, neurotoxicity, liver damage, and nephrotoxicity. In addition to these adverse effects, high recurrence and metastasis rates following treatment pose significant challenges for clinicians. There is an urgent need for novel therapeutic strategies to improve cancer treatment outcomes. In this context, nanozymes-artificial enzyme mimetics-have attracted considerable attention due to their unique advantages, including potent tumor-killing effects, enhanced biocompatibility, and reduced toxicity. Notably, nanozymes can dynamically monitor tumors through imaging and tracing. The multifunctional nanozyme (MN) is a promising research focus, integrating multiple catalytic activities, signal enhancement, sensing capabilities, and diverse modifications within a single nanozyme system. MNs can selectively target tumor regions, facilitating synergistic effects with other cancer therapies while enabling real-time imaging and tumor tracking. In this review, we first categorize MNs based on their composition and structural characteristics. We then discuss the primary mechanisms by which MNs exert their anticancer effects. Additionally, we review three types of MN biosensors and four MN-based therapeutic approaches applied in cancer treatment. Finally, we highlight the current challenges in MN research and provide an outlook on future developments in this field.
Collapse
Affiliation(s)
- Hailin Yin
- College of Basic Medical Sciences, The Medical Basic Research Innovation Center of Airway Disease in North China, Key Laboratory of Pathobiology, Ministry of Education, Jilin University, Changchun, 130021, China
| | - Yang Xin
- College of Basic Medical Sciences, The Medical Basic Research Innovation Center of Airway Disease in North China, Key Laboratory of Pathobiology, Ministry of Education, Jilin University, Changchun, 130021, China
| | - Jiaying Yang
- College of Basic Medical Sciences, The Medical Basic Research Innovation Center of Airway Disease in North China, Key Laboratory of Pathobiology, Ministry of Education, Jilin University, Changchun, 130021, China
| | - Qian Luo
- College of Basic Medical Sciences, The Medical Basic Research Innovation Center of Airway Disease in North China, Key Laboratory of Pathobiology, Ministry of Education, Jilin University, Changchun, 130021, China
| | - Mei Yang
- College of Basic Medical Sciences, The Medical Basic Research Innovation Center of Airway Disease in North China, Key Laboratory of Pathobiology, Ministry of Education, Jilin University, Changchun, 130021, China
| | - Jicheng Sun
- Department of Surgery, China-Japan Union Hospital, Jilin University, Changchun, 130033, China
| | - Yingtong Wang
- The Undergraduate Center of Hospital of Stomatology, Jilin University, Changchun, 130021, China
| | - Qi Wang
- College of Basic Medical Sciences, The Medical Basic Research Innovation Center of Airway Disease in North China, Key Laboratory of Pathobiology, Ministry of Education, Jilin University, Changchun, 130021, China
| | - Dhan V Kalvakolanu
- Greenebaum NCI Comprehensive Cancer Center, Department of Microbiology and Immunology, University of Maryland School Medicine, Baltimore, MD, USA
| | - Baofeng Guo
- Department of Surgery, China-Japan Union Hospital, Jilin University, Changchun, 130033, China
| | - Wei Jiang
- Academy of Medical Sciences, Tianjian Laboratory of Advanced Biomedical Sciences, Zhengzhou University, Zhengzhou, 450052, China.
| | - Ling Zhang
- College of Basic Medical Sciences, The Medical Basic Research Innovation Center of Airway Disease in North China, Key Laboratory of Pathobiology, Ministry of Education, Jilin University, Changchun, 130021, China.
| |
Collapse
|
2
|
Liu P, He S, Mentink A, Hart P, Wu Y, Terstappen LWMM, Jonkheijm P, Stevens M. Silica-coated magnetic nanobeads in a flow enrichment target capture Halbach (FETCH) magnetic separation system for circulating tumor cell enrichment. FEBS Lett 2025; 599:724-738. [PMID: 39743435 DOI: 10.1002/1873-3468.15094] [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: 09/12/2024] [Revised: 10/28/2024] [Accepted: 10/31/2024] [Indexed: 01/04/2025]
Abstract
Detecting circulating tumor cells (CTCs) is challenging due to their low presence and heterogeneity. Traditional methods using EpCAM-based separation struggle with CTCs that have undergone epithelial-mesenchymal transition, as this results in lower EpCAM expression. This study presents the use of silica-coated magnetic nanobeads functionalized with streptavidin for CTC capture. Using the FETCH magnetic separation system, we validated the capture efficiency of our beads on tumor cells with varying EpCAM expression. Our beads showed superior capture rates for LNCaP (97%), PC3-9 (91%), PC3 (23%), A549 (22%), and T24 (8%) cells compared to commercial MojoSort™ beads. Despite slightly higher nonspecific binding than CellSearch, our beads demonstrated improved sensitivity for EpCAMlow cells, suggesting they have promise for enhanced CTC capture.
Collapse
Affiliation(s)
- Peng Liu
- Department of Medical Cell Biophysics, TechMed Center, Faculty of Science and Technology, University of Twente, Enschede, The Netherlands
- Laboratory of Biointerface Chemistry, Department of Molecules and Materials, TechMed Centre, University of Twente, Enschede, The Netherlands
| | - Sitian He
- Department of Medical Cell Biophysics, TechMed Center, Faculty of Science and Technology, University of Twente, Enschede, The Netherlands
- College of Public Health, Zhengzhou University, China
| | - Anouk Mentink
- Department of Medical Cell Biophysics, TechMed Center, Faculty of Science and Technology, University of Twente, Enschede, The Netherlands
| | - Pieter Hart
- Department of Medical Cell Biophysics, TechMed Center, Faculty of Science and Technology, University of Twente, Enschede, The Netherlands
| | - Yongjun Wu
- College of Public Health, Zhengzhou University, China
| | - Leon W M M Terstappen
- Department of Medical Cell Biophysics, TechMed Center, Faculty of Science and Technology, University of Twente, Enschede, The Netherlands
- Department of General, Visceral and Pediatric Surgery, Heinrich-Heine University, University Hospital Düsseldorf, Germany
| | - Pascal Jonkheijm
- Laboratory of Biointerface Chemistry, Department of Molecules and Materials, TechMed Centre, University of Twente, Enschede, The Netherlands
| | - Michiel Stevens
- Department of Medical Cell Biophysics, TechMed Center, Faculty of Science and Technology, University of Twente, Enschede, The Netherlands
- FETCH BV, Deventer, The Netherlands
| |
Collapse
|
3
|
Ahmadi S, Yazdi F, Khastar S, Kaur I, Ahmed MH, Kumar A, Rathore G, Kaur P, Shahsavan M, Dehghani-Ghorbi M, Akhavan-Sigari R. Molecular Mechanism of lncRNAs in Regulation of Breast Cancer Metastasis; a Comprehensive Review. Cell Biochem Biophys 2025; 83:229-245. [PMID: 39367197 DOI: 10.1007/s12013-024-01535-y] [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] [Accepted: 09/07/2024] [Indexed: 10/06/2024]
Abstract
Although the number of breast cancer deaths has decreased, and there have been developments in targeted therapies and combination treatments for the management of metastatic illness, metastatic breast cancer is still the second most common cause of cancer-related deaths in U.S. women. Numerous phases and a vast number of proteins and signaling molecules are involved in the invasion-metastasis cascade. The tumor cells penetrate and enter the blood or lymphatic vessels, and travel to distant organs via the lymphatic or blood vessels. Tumor cells enter cell cycle arrest, adhere to capillary beds in the target organ, and then disseminate throughout the organ's parenchyma, proliferating and enhancing angiogenesis. Each of these processes is regulated by changes in the expression of different genes, in which lncRNAs play a role in this regulation. Transcripts that are longer than 200 nucleotides and do not translate into proteins are called RNAs. LncRNA molecules, whose function depends on their unique molecular structure, play significant roles in controlling the expression of genes at various epigenetic levels, transcription, and so on. LncRNAs have essential functions in regulating the expression of genes linked to cell development in healthy and pathological processes, specialization, programmed cell death, cell division, invasion, DNA damage, and spread to other parts of the body. A number of cancer types have been shown to exhibit aberrant expression of lncRNAs. In this review, we describe the general characteristics, potential molecular mechanisms and targeted therapy of lncRNAs and discuss the emerging functions of lncRNAs in breast cancer.
Collapse
Affiliation(s)
- Shokoufeh Ahmadi
- Department of Microbiology, Rabe'Rashidi University, Tabriz, Iran
| | - Farzaneh Yazdi
- Neuroscience Research Center, Institute of Neuropharmacology, Kerman University of Medical Sciences, Kerman, Iran
| | - Sahar Khastar
- Department of Immunology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Irwanjot Kaur
- Department of Biotechnology and Genetics, Jain (Deemed-to-be) University, Bengaluru, Karnataka-560069, India
- Department of Allied Healthcare and Sciences, Vivekananda Global University, Jaipur, Rajasthan-303012, India
| | | | - Abhishek Kumar
- School of Pharmacy-Adarsh Vijendra Institute of Pharmaceutical Sciences, Shobhit University, Gangoh, Uttar Pradesh-247341, India
- Department of Pharmacy, Arka Jain University, Jamshedpur, Jharkhand-831001, India
| | - Gulshan Rathore
- Department of Pharmaceutics, NIMS Institute of Pharmacy, NIMS University Rajasthan, Jaipur, India
| | - Parjinder Kaur
- Chandigarh Pharmacy College, Chandigarh Group of Colleges-Jhanjeri, Mohali 140307, Punjab, India
| | - Mohammad Shahsavan
- Department of Orthopedic Surgery, Isfahan University of Medical Sciences, Isfahan, Iran.
| | - Mahmoud Dehghani-Ghorbi
- Hematology-Oncology Department, Imam Hossein Educational Hospital, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Reza Akhavan-Sigari
- Department of Neurosurgery, University Medical Center, Tuebingen, Germany
- Department of Health Care Management and Clinical Research, Collegium Humanum Warsaw Management University Warsaw, Warsaw, Poland
| |
Collapse
|
4
|
Salomon R, Razavi Bazaz S, Mutafopulos K, Gallego-Ortega D, Warkiani M, Weitz D, Jin D. Challenges in blood fractionation for cancer liquid biopsy: how can microfluidics assist? LAB ON A CHIP 2025; 25:1097-1127. [PMID: 39775440 DOI: 10.1039/d4lc00563e] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/11/2025]
Abstract
Liquid biopsy provides a minimally invasive approach to characterise the molecular and phenotypic characteristics of a patient's individual tumour by detecting evidence of cancerous change in readily available body fluids, usually the blood. When applied at multiple points during the disease journey, it can be used to monitor a patient's response to treatment and to personalise clinical management based on changes in disease burden and molecular findings. Traditional liquid biopsy approaches such as quantitative PCR, have tended to look at only a few biomarkers, and are aimed at early detection of disease or disease relapse using predefined markers. With advances in the next generation sequencing (NGS) and single-cell genomics, simultaneous analysis of both circulating tumour DNA (ctDNA) and circulating tumour cells (CTCs) is now a real possibility. To realise this, however, we need to overcome issues with current blood collection and fractionation processes. These include overcoming the need to add a preservative to the collection tube or the need to rapidly send blood tubes to a centralised processing lab with the infrastructure required to fractionate and process the blood samples. This review focuses on outlining the current state of liquid biopsy and how microfluidic blood fractionation tools can be used in cancer liquid biopsy. We describe microfluidic devices that can separate plasma for ctDNA analysis, and devices that are important in isolating the cellular component(s) in liquid biopsy, i.e., individual CTCs and CTC clusters. To facilitate a better understanding of these devices, we propose a new categorisation system based on how these devices operate. The three categories being 1) solid Interaction devices, 2) fluid Interaction devices and 3) external force/active devices. Finally, we conclude that whilst some assays and some cancers are well suited to current microfluidic techniques, new tools are necessary to support broader, clinically relevant multiomic workflows in cancer liquid biopsy.
Collapse
Affiliation(s)
- Robert Salomon
- Children's Cancer Institute, Lowy Cancer Research Centre, UNSW, Sydney, Australia.
- Institute for Biomedical Materials and Devices (IBMD)/Faculty of Science, University of Technology Sydney, Sydney, NSW, 2007 Australia
| | - Sajad Razavi Bazaz
- Children's Cancer Institute, Lowy Cancer Research Centre, UNSW, Sydney, Australia.
| | - Kirk Mutafopulos
- Department of Physics, Harvard University, Cambridge, MA, 02138, USA
| | - David Gallego-Ortega
- Institute for Biomedical Materials and Devices (IBMD)/Faculty of Science, University of Technology Sydney, Sydney, NSW, 2007 Australia
- School of Clinical Medicine, Faculty of Medicine, University of New South Wales, Sydney, NSW, 2052, Australia
- School of Biomedical Engineering, University of Technology Sydney, Sydney, New South Wales 2007, Australia
- Garvan Institute of Medical Research, Darlinghurst, Sydney, NSW 2010, Australia
| | - Majid Warkiani
- Institute for Biomedical Materials and Devices (IBMD)/Faculty of Science, University of Technology Sydney, Sydney, NSW, 2007 Australia
- School of Biomedical Engineering, University of Technology Sydney, Sydney, New South Wales 2007, Australia
| | - David Weitz
- Department of Physics, Harvard University, Cambridge, MA, 02138, USA
| | - Dayong Jin
- Institute for Biomedical Materials and Devices (IBMD)/Faculty of Science, University of Technology Sydney, Sydney, NSW, 2007 Australia
| |
Collapse
|
5
|
Szerenyi D, Jarvas G, Guttman A. Multifaceted Approaches in Epithelial Cell Adhesion Molecule-Mediated Circulating Tumor Cell Isolation. Molecules 2025; 30:976. [PMID: 40076201 PMCID: PMC11901967 DOI: 10.3390/molecules30050976] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2024] [Revised: 02/11/2025] [Accepted: 02/12/2025] [Indexed: 03/14/2025] Open
Abstract
Circulating tumor cells (CTCs) are pivotal in cancer metastasis and serve as valuable biomarkers for diagnosis, prognosis, and treatment monitoring. Traditional CTC capture methods predominantly utilize the epithelial cell adhesion molecule (EpCAM) as a marker for isolation. However, the heterogeneity of these circulating cells and the epithelial-to-mesenchymal transition process (wherein epithelial cells acquire mesenchymal characteristics) limit the efficacy of EpCAM-based capture techniques. In this paper, we critically review the role of the EpCAM in CTC capture, explore the impact of epithelial-to-mesenchymal transition on EpCAM expression, and discuss alternative biomarkers and strategies to enhance CTC isolation. By evaluating the limitations of EpCAM-mediated capture and the challenges posed by epithelial-to-mesenchymal transition, we aim to provide insights into the development of more comprehensive liquid biopsy approaches for cancer management.
Collapse
Affiliation(s)
- Dora Szerenyi
- Research Institute of Biomolecular and Chemical Engineering, Faculty of Engineering, University of Pannonia, 8200 Veszprem, Hungary;
| | - Gabor Jarvas
- Research Institute of Biomolecular and Chemical Engineering, Faculty of Engineering, University of Pannonia, 8200 Veszprem, Hungary;
- CAPTEC Medical Ltd., 8200 Veszprem, Hungary
| | - Andras Guttman
- Research Institute of Biomolecular and Chemical Engineering, Faculty of Engineering, University of Pannonia, 8200 Veszprem, Hungary;
- CAPTEC Medical Ltd., 8200 Veszprem, Hungary
- Horváth Csaba Memorial Laboratory of Bioseparation Sciences, Research Center for Molecular Medicine, Faculty of Medicine, University of Debrecen, 4032 Debrecen, Hungary
| |
Collapse
|
6
|
Yadav M, Sharma A, Patne K, Tabasum S, Suryavanshi J, Rawat L, Machaalani M, Eid M, Singh RP, Choueiri TK, Pal S, Sabarwal A. AXL signaling in cancer: from molecular insights to targeted therapies. Signal Transduct Target Ther 2025; 10:37. [PMID: 39924521 PMCID: PMC11808115 DOI: 10.1038/s41392-024-02121-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2024] [Revised: 11/02/2024] [Accepted: 12/19/2024] [Indexed: 02/11/2025] Open
Abstract
AXL, a member of the TAM receptor family, has emerged as a potential target for advanced-stage human malignancies. It is frequently overexpressed in different cancers and plays a significant role in various tumor-promoting pathways, including cancer cell proliferation, invasion, metastasis, epithelial-mesenchymal transition (EMT), angiogenesis, stemness, DNA damage response, acquired therapeutic resistance, immunosuppression, and inflammatory responses. Beyond oncology, AXL also facilitates viral infections, including SARS-CoV-2 and Zika highlighting its importance in both cancer and virology. In preclinical models, small-molecule kinase inhibitors targeting AXL have shown promising anti-tumorigenic potential. This review primarily focuses on the induction, regulation and biological functions of AXL in mediating these tumor-promoting pathways. We discuss a range of therapeutic strategies, including recently developed small-molecule tyrosine kinase inhibitors (TKIs), monoclonal antibodies, and antibody-drug conjugates (ADCs), anti-AXL-CAR, and combination therapies. These interventions are being examined in both preclinical and clinical studies, offering the potential for improved drug sensitivity and therapeutic efficacy. We further discuss the mechanisms of acquired therapeutic resistance, particularly the crosstalk between AXL and other critical receptor tyrosine kinases (RTKs) such as c-MET, EGFR, HER2/HER3, VEGFR, PDGFR, and FLT3. Finally, we highlight key research areas that require further exploration to enhance AXL-mediated therapeutic approaches for improved clinical outcomes.
Collapse
Affiliation(s)
- Monika Yadav
- Cancer Biology Laboratory, School of Life Sciences, Jawaharlal Nehru University, New Delhi, Delhi, India
- Laboratory of Nanotechnology and Chemical Biology, Regional Center for Biotechnology, Faridabad, Haryana, India
| | - Akansha Sharma
- Department of Microbiology and Immunology, Thomas Jefferson University, Philadelphia, PA, USA
| | - Ketki Patne
- Chromatin Remodeling Laboratory, School of Life Sciences, Jawaharlal Nehru University, New Delhi, India
| | - Saba Tabasum
- Dana-Farber Cancer Institute, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
| | - Jyoti Suryavanshi
- Department of Cell Biology and Anatomy, New York Medical College, Valhalla, NY, USA
| | - Laxminarayan Rawat
- Harvard Medical School, Boston, MA, USA
- Division of Nephrology, Boston Children's Hospital, Boston, MA, USA
| | - Marc Machaalani
- Dana-Farber Cancer Institute, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
| | - Marc Eid
- Dana-Farber Cancer Institute, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
| | - Rana P Singh
- Cancer Biology Laboratory, School of Life Sciences, Jawaharlal Nehru University, New Delhi, Delhi, India
| | - Toni K Choueiri
- Dana-Farber Cancer Institute, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
| | - Soumitro Pal
- Harvard Medical School, Boston, MA, USA.
- Division of Nephrology, Boston Children's Hospital, Boston, MA, USA.
| | - Akash Sabarwal
- Harvard Medical School, Boston, MA, USA.
- Division of Nephrology, Boston Children's Hospital, Boston, MA, USA.
| |
Collapse
|
7
|
Magnusson C, Rezayati Charan M, Augustsson P. Two-Step Acoustic Cell Separation Based on Cell Size and Acoustic Impedance─toward Isolation of Viable Circulating Tumor Cells. Anal Chem 2025; 97:2120-2126. [PMID: 39818757 PMCID: PMC11800186 DOI: 10.1021/acs.analchem.4c04911] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2024] [Revised: 12/20/2024] [Accepted: 12/24/2024] [Indexed: 01/19/2025]
Abstract
Isolation and characterization of circulating tumor cells (CTCs) present a noninvasive alternative to monitor disease progression in individual patients. However, the heterogeneous lineage specificity of CTCs makes it difficult to isolate and identify possible CTCs by a liquid biopsy. Better label-free methods for the isolation of viable CTCs are needed. Our solution is a combined approach that is inherently epitope independent. Cells are separated by size-sensitive acoustophoresis using an ultrasonic standing wave field, followed by size-insensitive, acoustic barrier-medium focusing, which enables the enrichment of viable cancer cells in blood. With standard acoustophoresis in homogeneous medium, lymphocytes and monocytes were efficiently removed, while removal of granulocytes from the target MCF7 breast cancer cells was not possible due to overlapping acoustic migration velocities for viable cells. Remaining granulocytes were removed by a second separation step with an acoustic impedance barrier-medium selectively blocking the transport of MCF7 cells to generate a clean cancer cell fraction. For two series of 500 mL samples containing 5 × 105 white blood cells, spiked with 2 × 104 or 1 × 103 MCF7 cells, the recovery of MCF7 cells was 77.3% with a 99.9% depletion of white blood cells in the final cancer cell fraction. The most abundant contaminating cell type was granulocytes (85.9% of remaining cells). Nearly all lymphocytes (99.996%) and monocytes (99.995%) were depleted. A two-step acoustic cell separation based on cell size and acoustic impedance is well suited to generate a purified cancer cell fraction as a preparatory step for downstream single-cell analysis.
Collapse
Affiliation(s)
- Cecilia Magnusson
- Department
of Translational Medicine, Lund University, Lund SE-22100, Sweden
| | | | - Per Augustsson
- Department
of Biomedical Engineering, Lund University, Lund SE-223 63, Sweden
| |
Collapse
|
8
|
Meunier A, Hernández-Castro JA, Chahley N, Communal L, Kheireddine S, Koushki N, Davoudvandi N, Al Habyan S, Péant B, Lazaris A, Ng A, Veres T, McCaffrey L, Provencher D, Metrakos P, Mes-Masson AM, Juncker D. Gravity-based microfiltration reveals unexpected prevalence of circulating tumor cell clusters in ovarian and colorectal cancer. COMMUNICATIONS MEDICINE 2025; 5:33. [PMID: 39900650 PMCID: PMC11790846 DOI: 10.1038/s43856-024-00702-9] [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: 07/23/2022] [Accepted: 12/10/2024] [Indexed: 02/05/2025] Open
Abstract
BACKGROUND Circulating tumor cells (CTCs) are rare (a few cells per milliliter of blood) and mostly isolated as single-cell CTCs (scCTCs). CTC clusters (cCTCs), even rarer, are of growing interest, notably because of their higher metastatic potential, but very difficult to isolate. METHOD We introduce gravity-based microfiltration (GµF) for facile isolation of cCTCs using in-house fabricated microfilters and 3D printed cartridges. Optimal flow rate and pore size for cCTC isolation are determined by GµF of cultured ovarian single cells and cell clusters spiked in healthy blood. We perform GµF of blood from orthotopic ovarian cancer mouse models and characterize the morphological features of scCTCs and cCTCs, and the expression of molecular markers for aggressiveness. Finally, we analyze blood from 17 epithelial ovarian cancer patients with either localized or metastatic disease, and from 13 colorectal cancer liver metastasis patients. RESULTS Here, we show that GµF optimized for cell cluster isolation captures cCTCs from blood while minimizing unwanted cluster disaggregation, with ~85% capture efficiency. We detect cCTCs in every patient, with between 2-100+ cells. We find cCTCs represent between 5-30% of all CTC capture events, and 10-80% of the CTCs are clustered; remarkably, in 10 patients, most CTCs are circulating not as scCTCs, but as cCTCs. CONCLUSIONS GµF uncovers the unexpected prevalence and frequency of cCTCs including sometimes very large ones in epithelial ovarian cancer patients, and motivates additional studies to uncover their properties and role in disease progression.
Collapse
Affiliation(s)
- Anne Meunier
- Biomedical Engineering Department, McGill University, Montreal, QC, H3A 2B4, Canada
- Victor Phillip Dahdaleh Institute of Genomic Medicine, McGill University, Montreal, QC, H3A 0G1, Canada
| | - Javier Alejandro Hernández-Castro
- Biomedical Engineering Department, McGill University, Montreal, QC, H3A 2B4, Canada
- Victor Phillip Dahdaleh Institute of Genomic Medicine, McGill University, Montreal, QC, H3A 0G1, Canada
- National Research Council of Canada, Boucherville, QC, J4B 6Y4, Canada
| | - Nicholas Chahley
- Biomedical Engineering Department, McGill University, Montreal, QC, H3A 2B4, Canada
- Victor Phillip Dahdaleh Institute of Genomic Medicine, McGill University, Montreal, QC, H3A 0G1, Canada
| | - Laudine Communal
- Institut du cancer de Montréal and Centre de recherche du Centre hospitalier de l'Université de Montréal (CRCHUM), Montréal, QC, H2X 0A9, Canada
| | - Sara Kheireddine
- Biomedical Engineering Department, McGill University, Montreal, QC, H3A 2B4, Canada
- Victor Phillip Dahdaleh Institute of Genomic Medicine, McGill University, Montreal, QC, H3A 0G1, Canada
| | - Newsha Koushki
- Biomedical Engineering Department, McGill University, Montreal, QC, H3A 2B4, Canada
- Victor Phillip Dahdaleh Institute of Genomic Medicine, McGill University, Montreal, QC, H3A 0G1, Canada
| | - Nadia Davoudvandi
- Biomedical Engineering Department, McGill University, Montreal, QC, H3A 2B4, Canada
- Victor Phillip Dahdaleh Institute of Genomic Medicine, McGill University, Montreal, QC, H3A 0G1, Canada
| | - Sara Al Habyan
- Rosalind and Morris Goodman Cancer Institute, McGill University, Montreal, QC, H3A 1A3, Canada
- Division of Experimental Medicine, McGill University, Montreal, QC, H4A 3J1, Canada
| | - Benjamin Péant
- Institut du cancer de Montréal and Centre de recherche du Centre hospitalier de l'Université de Montréal (CRCHUM), Montréal, QC, H2X 0A9, Canada
| | - Anthoula Lazaris
- Cancer Research Program, The Research Institute of McGill University Health Center, Montreal, QC, H4A 3J1, Canada
| | - Andy Ng
- Biomedical Engineering Department, McGill University, Montreal, QC, H3A 2B4, Canada
- Victor Phillip Dahdaleh Institute of Genomic Medicine, McGill University, Montreal, QC, H3A 0G1, Canada
| | - Teodor Veres
- Biomedical Engineering Department, McGill University, Montreal, QC, H3A 2B4, Canada
- National Research Council of Canada, Boucherville, QC, J4B 6Y4, Canada
| | - Luke McCaffrey
- Rosalind and Morris Goodman Cancer Institute, McGill University, Montreal, QC, H3A 1A3, Canada
- Division of Experimental Medicine, McGill University, Montreal, QC, H4A 3J1, Canada
- Gerald Bronfman Department of Oncology, McGill University, Montreal, QC, H4A 3T2, Canada
| | - Diane Provencher
- Institut du cancer de Montréal and Centre de recherche du Centre hospitalier de l'Université de Montréal (CRCHUM), Montréal, QC, H2X 0A9, Canada
- Division of Gynecologic Oncology, Department of Obstetrics-Gynecology, Université de Montréal, Montreal, QC, H3T 1J4, Canada
| | - Peter Metrakos
- Cancer Research Program, The Research Institute of McGill University Health Center, Montreal, QC, H4A 3J1, Canada
| | - Anne-Marie Mes-Masson
- Institut du cancer de Montréal and Centre de recherche du Centre hospitalier de l'Université de Montréal (CRCHUM), Montréal, QC, H2X 0A9, Canada
- Department of Medicine, Faculty of Medicine, Université de Montréal, Montréal, QC, H3T 1J4, Canada
| | - David Juncker
- Biomedical Engineering Department, McGill University, Montreal, QC, H3A 2B4, Canada.
- Victor Phillip Dahdaleh Institute of Genomic Medicine, McGill University, Montreal, QC, H3A 0G1, Canada.
- Rosalind and Morris Goodman Cancer Institute, McGill University, Montreal, QC, H3A 1A3, Canada.
| |
Collapse
|
9
|
Bergmann L, Afflerbach AK, Yuan T, Pantel K, Smit DJ. Lessons (to be) learned from liquid biopsies: assessment of circulating cells and cell-free DNA in cancer and pregnancy-acquired microchimerism. Semin Immunopathol 2025; 47:14. [PMID: 39893314 PMCID: PMC11787191 DOI: 10.1007/s00281-025-01042-z] [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: 08/05/2024] [Accepted: 01/20/2025] [Indexed: 02/04/2025]
Abstract
Tumors constantly shed cancer cells that are considered the mediators of metastasis via the blood stream. Analysis of circulating cells and circulating cell-free DNA (cfDNA) in liquid biopsies, mostly taken from peripheral blood, have emerged as powerful biomarkers in oncology, as they enable the detection of genomic aberrations. Similarly, liquid biopsies taken from pregnant women serve as prenatal screening test for an abnormal number of chromosomes in the fetus, e.g., via the analysis of microchimeric fetal cells and cfDNA circulating in maternal blood. Liquid biopsies are minimally invasive and, consequently, associated with reduced risks for the patients. However, different challenges arise in oncology and pregnancy-acquired liquid biopsies with regard to the analyte concentration and biological (background) noise among other factors. In this review, we highlight the unique biological properties of circulating tumor cells (CTC), summarize the various techniques that have been developed for the enrichment, detection and analysis of CTCs as well as for analysis of genetic and epigenetic aberrations in cfDNA and highlight the range of possible clinical applications. Lastly, the potential, but also the challenges of liquid biopsies in oncology as well as their translational value for the analysis of pregnancy-acquired microchimerism are discussed.
Collapse
Affiliation(s)
- Lina Bergmann
- Institute of Tumor Biology, University Medical Center Hamburg-Eppendorf, Martinistraße 52, Hamburg, 20246, Germany
| | - Ann-Kristin Afflerbach
- Institute of Tumor Biology, University Medical Center Hamburg-Eppendorf, Martinistraße 52, Hamburg, 20246, Germany
| | - Tingjie Yuan
- Institute of Tumor Biology, University Medical Center Hamburg-Eppendorf, Martinistraße 52, Hamburg, 20246, Germany
| | - Klaus Pantel
- Institute of Tumor Biology, University Medical Center Hamburg-Eppendorf, Martinistraße 52, Hamburg, 20246, Germany.
| | - Daniel J Smit
- Institute of Tumor Biology, University Medical Center Hamburg-Eppendorf, Martinistraße 52, Hamburg, 20246, Germany.
| |
Collapse
|
10
|
Rzhevskiy AS, Sagitova GR, Karashaeva TA, Morozov AO, Fatyanova AS, Kazantseva VV, Joosse SA, Zvyagin AV, Warkini ME. A comprehensive review and meta-analysis of CTC isolation methods in breast cancer. Crit Rev Oncol Hematol 2025; 206:104579. [PMID: 39615710 DOI: 10.1016/j.critrevonc.2024.104579] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2024] [Revised: 11/22/2024] [Accepted: 11/24/2024] [Indexed: 12/08/2024] Open
Abstract
The application of circulating tumor cells (CTCs) as diagnostic and prognostic markers in oncology is gaining increasing importance in clinical practice. Currently, various methods exist for detecting CTCs in patients' biological fluids. This systematic review aimed to compare the efficacy of different techniques for isolating and detecting CTCs from blood, against the FDA-cleared CellSearch® technology, in breast cancer patients. We performed a systematic literature search using two databases (PubMed and the Cochrane Library) with the following terms: ("Circulating tumor cells" OR CTC) AND "breast cancer", covering the period from 2004 to April 2023. The primary outcome measured was the sensitivity, specificity, and overall accuracy of various CTC enrichment methods in comparison with the CellSearch® System. Secondary outcomes included the prognostic value of CTCs in evaluating response to treatment based on survival rates. Generally, a high level of agreement between CellSearch and other methods was observed in isolating CTCs from patients' blood with both metastatic and early-stage disease. Studies asserting the superiority of new methods over CellSearch frequently used clinically unvalidated cut-off thresholds for their patient cohorts. Additionally, these studies sometimes included different nonoverlapping patient cohorts and lacked a standardized chemotherapy treatment protocol, which could affect the quantitative changes in CTC. It is evident that methods simultaneously composed of physical and immunomagnetic approaches for CTC isolation significantly surpass CellSearch, which relies solely on the expression of specific markers on the CTCs' surface. The count of CTCs has been established as a predictive marker in terms of clinically important parameters namely progression-free survival (PFS) and overall survival (OS). The CTC-count value was significantly correlated with PFS and OS rates.
Collapse
Affiliation(s)
- Alexey S Rzhevskiy
- Institute of Molecular Theranostics, Sechenov First Moscow State Medical University, Moscow 119991, Russia; Faculty of Computer Science, National Research University Higher School of Economics, Moscow 101000, Russia
| | - Guzel R Sagitova
- Institute of Molecular Theranostics, Sechenov First Moscow State Medical University, Moscow 119991, Russia
| | - Tamilla A Karashaeva
- Institute of Molecular Theranostics, Sechenov First Moscow State Medical University, Moscow 119991, Russia
| | - Andrey O Morozov
- Institute for Urology and Reproductive Health, Sechenov University, Moscow 119991, Russia
| | - Anastasia S Fatyanova
- Department of Oncology, Radiotherapy and Reconstructive Surgery, Institution of Clinical Medicine, Sechenov First Moscow State Medical University, Moscow 119991, Russia
| | - Vlada V Kazantseva
- Institute of Molecular Theranostics, Sechenov First Moscow State Medical University, Moscow 119991, Russia
| | - Simon A Joosse
- Department of Tumor Biology, University Medical Center Hamburg-Eppendorf, Martinistr. 52, Hamburg 20246, Germany; Mildred Scheel Cancer Career Center HaTriCS4, University Medical Center Hamburg-Eppendorf, Martinistr. 52, Hamburg 20246, Germany
| | - Andrei V Zvyagin
- Institute of Molecular Theranostics, Sechenov First Moscow State Medical University, Moscow 119991, Russia; Australian Research Council Centre of Excellence for Nanoscale Biophotonics, Macquarie University, Sydney, NSW 2109, Australia; Research Center for Translational Medicine, Sirius University of Science and Technology, Sochi 354340, Russia.
| | - Majid Ebrahimi Warkini
- Institute of Molecular Theranostics, Sechenov First Moscow State Medical University, Moscow 119991, Russia; School of Biomedical Engineering, University of Technology Sydney, Sydney, NSW 2007, Australia
| |
Collapse
|
11
|
Ah-Thiane L, Campion L, Allouache N, Meyer E, Pommier P, Mesgouez-Nebout N, Serre AA, Créhange G, Guimas V, Rio E, Sargos P, Ladoire S, Mahier Ait Oukhatar C, Supiot S. Combination of Abiraterone Acetate, Prostate Bed Radiotherapy, and Luteinizing Hormone-releasing Hormone Agonists in Biochemically Relapsing Patients After Prostatectomy (CARLHA): A Phase 2 Clinical Trial. Eur Urol Oncol 2025; 8:38-46. [PMID: 38734543 DOI: 10.1016/j.euo.2024.04.014] [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: 02/12/2024] [Revised: 03/26/2024] [Accepted: 04/23/2024] [Indexed: 05/13/2024]
Abstract
BACKGROUND The relevance of next-generation hormone therapies and circulating tumor cells (CTCs) are not elucidated in biochemical recurrence after prostatectomy. OBJECTIVE To evaluate the combination of abiraterone acetate plus prednisone (AAP), prostate bed radiotherapy (PBRT), and goserelin in biochemically relapsing men after prostatectomy, and to investigate the utility of CTCs. DESIGN, SETTING, AND PARTICIPANTS In this single-arm multicenter phase 2 trial, 46 biochemically relapsing men were enrolled between December 2012 and January 2019. The median follow-up was 47 mo. INTERVENTION All patients received AAP 1000 mg daily (but 750 mg during PBRT), salvage PBRT, and goserelin. OUTCOME MEASUREMENTS AND STATISTICAL ANALYSIS The primary outcome was 3-yr biochemical recurrence-free survival (bRFS) when prostate-specific antigen (PSA) levels were ≥0.2 ng/ml. The secondary outcomes included alternative bRFS (alt-bRFS) when PSA levels were ≥0.5 ng/ml and safety assessment. CTC count was assessed. RESULTS AND LIMITATIONS The 3-yr bRFS and alt-bRFS were 81.5% (95% confidence interval or CI [66.4-90.3%]) and 95.6% (95% CI [83.5-98.9%]), respectively. The most common acute radiotherapy-related adverse effect (AE; all grades was pollakiuria (41.3%). The most common late AE (all grades) was urinary incontinence (15.2%). Grade 3-4 acute or late radiotherapy-related AEs were scarce. Most frequent AEs nonrelated to radiotherapy were hot flashes (76%), hypertension (63%), and hepatic cytolysis (50%, of which 20% were of grades 3-4). Of the patients, 11% had a CTC count of ≥5, which was correlated with poorer bRFS (p = 0.042) and alt-bRFS (p = 0.008). The association between CTC count and higher rates of relapse was independent of the baseline PSA level and PSA doubling time (p = 0.42 and p = 0.09, respectively). This study was nonrandomized with a limited number of patients, and few clinical events were reported. CONCLUSIONS Adding AAP to salvage radiation therapy and goserelin resulted in high bRFS and alt-bRFS. AEs remained manageable, although a close liver surveillance is advised. CTC count appears as a promising biomarker for prognosis and predicting response to treatment. PATIENT SUMMARY Our study was a phase 2 clinical trial that exhibited the efficacy and tolerance of a novel androgen-receptor targeting agent (abiraterone acetate plus prednisone) in patients with prostate cancer who experienced rising prostate-specific antigen after radical prostatectomy, in combination with prostate bed radiotherapy. The results also indicated the feasibility and potential value of circulating tumor cell detection, which constitutes a possible advance in managing prostate cancers.
Collapse
Affiliation(s)
- Loic Ah-Thiane
- Department of Radiotherapy, ICO Rene Gauducheau, St-Herblain, France
| | - Loic Campion
- Department of Biostatistics, ICO Rene Gauducheau, St-Herblain, France
| | - Nedjla Allouache
- Department of Radiotherapy, Francois Baclesse Center, Caen, France
| | - Emmanuel Meyer
- Department of Radiotherapy, Francois Baclesse Center, Caen, France
| | - Pascal Pommier
- Department of Radiotherapy, Leon Berard Center, Lyon, France
| | | | | | - Gilles Créhange
- Department of Radiotherapy, Georges Francois Leclerc Center, Dijon, France
| | - Valentine Guimas
- Department of Radiotherapy, ICO Rene Gauducheau, St-Herblain, France
| | - Emmanuel Rio
- Department of Radiotherapy, ICO Rene Gauducheau, St-Herblain, France
| | - Paul Sargos
- Department of Radiotherapy, Bergonie Institute, Bordeaux, France
| | - Sylvain Ladoire
- Department of Radiotherapy, Georges Francois Leclerc Center, Dijon, France
| | | | - Stéphane Supiot
- Department of Radiotherapy, ICO Rene Gauducheau, St-Herblain, France; Inserm UMR1232, CNRS ERL 6001, Nantes University, Nantes, France.
| |
Collapse
|
12
|
Saadh MJ, Bishoyi AK, Ballal S, Singh A, Kareem RA, Devi A, Sharma GC, Naidu KS, Sead FF. MicroRNAs as behind-the-scenes molecules in breast cancer metastasis and their therapeutic role through novel microRNA-based delivery strategies. Gene 2025; 944:149272. [PMID: 39894085 DOI: 10.1016/j.gene.2025.149272] [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: 12/18/2024] [Accepted: 01/20/2025] [Indexed: 02/04/2025]
Abstract
Breast cancer is the primary cause of cancer-related death and the most frequent malignancy among women in Western countries. Although there have been advancements in combination treatments and targeted therapies for the metastatic diseases management, metastatic breast cancer is still the second most common cause of cancer-related deaths among U.S. women. The routes of metastasis encompass invasion, intravasation, circulation, extravasation, infiltration into a remote location to establish a metastatic niche, and the formation of micro-metastases in a new environment. Each of these processes is regulated by changes in gene expression. MicroRNAs (miRNAs) are widely expressed by a variety of organisms and have a key role in cell activities including suppressing or promoting cancer through regulating various pathways. Target gene expression is post-transcriptionally regulated by miRNAs, which contribute to the development, spread, and metastasis of breast cancer. In this study, we comprehensively discussed the role of miRNAs as predictors of breast cancer metastasis, their correlation with the spread of the disease to certain organs, and their potential application as targets for breast cancer treatment. We also provided molecular mechanisms of miRNAs in the progression of breast cancer, as well as current challenges in miRNA-based therapeutic approaches. Furthermore, as one of the primary issues with the treatment of solid malignancies is the efficient delivery of miRNAs, we examined a number of cutting-edge carriers for miRNA-based therapies and CRISPR/Cas9 as a targeted therapy for breast cancer.
Collapse
Affiliation(s)
- Mohamed J Saadh
- Faculty of Pharmacy, Middle East University, Amman 11831, Jordan.
| | - Ashok Kumar Bishoyi
- Marwadi University Research Center, Department of Microbiology, Faculty of Science, Marwadi University, Rajkot 360003, Gujarat, India
| | - Suhas Ballal
- Department of Chemistry and Biochemistry, School of Sciences, JAIN (Deemed to be University), Bangalore, Karnataka, India
| | - Abhayveer Singh
- Centre for Research Impact & Outcome, Chitkara University Institute of Engineering and Technology, Chitkara University, Rajpura 140401, Punjab, India
| | | | - Anita Devi
- Department of Chemistry Chandigarh Engineering College, Chandigarh Group of Colleges-Jhanjeri, Mohali 140307, Punjab, India
| | - Girish Chandra Sharma
- Department of Applied Sciences-Chemistry, NIMS Institute of Engineering & Technology, NIMS University Rajasthan, Jaipur, India
| | - K Satyam Naidu
- Department of Chemistry, Raghu Engineering College, Visakhapatnam, Andhra Pradesh 531162, India
| | - Fadhil Faez Sead
- Department of Dentistry, College of Dentistry, The Islamic University, Najaf, Iraq; Department of Medical Analysis, Medical Laboratory Technique College, the Islamic University of Al Diwaniyah, Al Diwaniyah, Iraq
| |
Collapse
|
13
|
Murgoitio-Esandi J, Tessone D, Naghdloo A, Shishido SN, Zhang B, Xu H, Dasgupta A, Mason J, Nagaraju RM, Hicks J, Kuhn P, Oberai A. Unsupervised Detection of Rare Events in Liquid Biopsy Assays. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.01.29.635501. [PMID: 39975209 PMCID: PMC11838382 DOI: 10.1101/2025.01.29.635501] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 02/21/2025]
Abstract
The use of liquid biopsies in the detection, diagnosis and treatment monitoring of different types of cancers and other diseases often requires identifying and enumerating instances of analytes that are rare. Most current techniques that aim to computationally isolate these rare instances or events first learn the signature of the event, and then scan the appropriate biological assay for this signature. While such techniques have proven to be very useful, they are limited because they must first establish what signature to look for, and only then identify events that are consistent with this signature. In contrast to this, in this study, we present an automated approach that does not require the knowledge of the signature of the rare event. It works by breaking the assay into a sequence of components, learning the probability distribution of these components, and then isolating those that are rare. This is done with the help of deep generative algorithms in an unsupervised manner, meaning without a-priori knowledge of the rare event associated with an analyte. In this study, this approach is applied to immunofluorescence microscopy images of peripheral blood, where it is shown that it successfully isolates biologically relevant events in blood from normal donors spiked with cancer-related cells and in blood from patients with late-stage breast cancer.
Collapse
Affiliation(s)
- Javier Murgoitio-Esandi
- Department of Aerospace and Mechanical Engineering, Viterbi School of Engineering, University of Southern California, Los Angeles, 90089, CA, USA
| | - Dean Tessone
- Convergent Science Institute for Cancer, Michelson Center, University of Southern California, Los Angeles, 90089, CA, USA
- Department of Biological Sciences, Dornsife College of Letters, Arts and Sciences, University of Southern California, Los Angeles, 90089, CA, USA
| | - Amin Naghdloo
- Department of Aerospace and Mechanical Engineering, Viterbi School of Engineering, University of Southern California, Los Angeles, 90089, CA, USA
- Convergent Science Institute for Cancer, Michelson Center, University of Southern California, Los Angeles, 90089, CA, USA
| | - Stephanie N Shishido
- Convergent Science Institute for Cancer, Michelson Center, University of Southern California, Los Angeles, 90089, CA, USA
| | - Brian Zhang
- Convergent Science Institute for Cancer, Michelson Center, University of Southern California, Los Angeles, 90089, CA, USA
| | - Haofeng Xu
- Department of Computer Science, Viterbi School of Engineering, University of Southern California, Los Angeles, 90089, CA, USA
| | - Agnimitra Dasgupta
- Department of Aerospace and Mechanical Engineering, Viterbi School of Engineering, University of Southern California, Los Angeles, 90089, CA, USA
| | - Jeremy Mason
- Convergent Science Institute for Cancer, Michelson Center, University of Southern California, Los Angeles, 90089, CA, USA
- Institute of Urology, Catherine & Joseph Aresty Department of Urology, Keck School of Medicine, University of Southern California, Los Angeles, 90033, CA, USA
- Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, Los Angeles, 90033, CA, USA
| | - Rajiv M Nagaraju
- Convergent Science Institute for Cancer, Michelson Center, University of Southern California, Los Angeles, 90089, CA, USA
| | - James Hicks
- Convergent Science Institute for Cancer, Michelson Center, University of Southern California, Los Angeles, 90089, CA, USA
| | - Peter Kuhn
- Department of Aerospace and Mechanical Engineering, Viterbi School of Engineering, University of Southern California, Los Angeles, 90089, CA, USA
- Convergent Science Institute for Cancer, Michelson Center, University of Southern California, Los Angeles, 90089, CA, USA
- Department of Biological Sciences, Dornsife College of Letters, Arts and Sciences, University of Southern California, Los Angeles, 90089, CA, USA
- Institute of Urology, Catherine & Joseph Aresty Department of Urology, Keck School of Medicine, University of Southern California, Los Angeles, 90033, CA, USA
- Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, Los Angeles, 90033, CA, USA
- Department of Biomedical Engineering, Viterbi School of Engineering, University of Southern California, Los Angeles, 90089, CA, USA
| | - Assad Oberai
- Department of Aerospace and Mechanical Engineering, Viterbi School of Engineering, University of Southern California, Los Angeles, 90089, CA, USA
| |
Collapse
|
14
|
Moon GY, Dalkiran B, Park HS, Shin D, Son C, Choi JH, Bang S, Lee H, Doh I, Kim DH, Jeong WJ, Bu J. Dual Biomarker Strategies for Liquid Biopsy: Integrating Circulating Tumor Cells and Circulating Tumor DNA for Enhanced Tumor Monitoring. BIOSENSORS 2025; 15:74. [PMID: 39996976 PMCID: PMC11852634 DOI: 10.3390/bios15020074] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/28/2024] [Revised: 01/21/2025] [Accepted: 01/26/2025] [Indexed: 02/26/2025]
Abstract
The liquid biopsy has gained significant attention in cancer diagnostics, with circulating tumor cells (CTCs) and circulating tumor DNA (ctDNA) being recognized as key biomarkers for tumor detection and monitoring. However, each biomarker possesses inherent limitations that restrict its standalone clinical utility, such as the rarity and heterogeneity of CTCs and the variable sensitivity and specificity of ctDNA assays. This highlights the necessity of integrating both biomarkers to maximize diagnostic and prognostic potential, offering a more comprehensive understanding of the tumor biology and therapeutic response. In this review, we summarize clinical studies that have explored the combined analysis of CTCs and ctDNA as biomarkers, providing insights into their synergistic value in diverse tumor types. Specifically, this paper examines the individual advantages and limitations of CTCs and ctDNA, details the findings of combined biomarker studies across various cancers, highlights the benefits of dual biomarker approaches over single-biomarker strategies, and discusses future prospects for advancing personalized oncology through liquid biopsies. By offering a comprehensive overview of clinical studies combining CTCs and ctDNA, this review serves as a guideline for researchers and clinicians aiming to enhance biomarker-based strategies in oncology and informs biosensor design for improved biomarker detection.
Collapse
Affiliation(s)
- Ga Young Moon
- Department of Biological Sciences and Bioengineering, Inha University, 100 Inha-ro, Michuhol-gu, Incheon 22212, Republic of Korea; (G.Y.M.); (B.D.); (H.S.P.); (D.S.); (C.S.); (J.H.C.); (S.B.); (H.L.)
| | - Basak Dalkiran
- Department of Biological Sciences and Bioengineering, Inha University, 100 Inha-ro, Michuhol-gu, Incheon 22212, Republic of Korea; (G.Y.M.); (B.D.); (H.S.P.); (D.S.); (C.S.); (J.H.C.); (S.B.); (H.L.)
| | - Hyun Sung Park
- Department of Biological Sciences and Bioengineering, Inha University, 100 Inha-ro, Michuhol-gu, Incheon 22212, Republic of Korea; (G.Y.M.); (B.D.); (H.S.P.); (D.S.); (C.S.); (J.H.C.); (S.B.); (H.L.)
| | - Dongjun Shin
- Department of Biological Sciences and Bioengineering, Inha University, 100 Inha-ro, Michuhol-gu, Incheon 22212, Republic of Korea; (G.Y.M.); (B.D.); (H.S.P.); (D.S.); (C.S.); (J.H.C.); (S.B.); (H.L.)
| | - Chaeyeon Son
- Department of Biological Sciences and Bioengineering, Inha University, 100 Inha-ro, Michuhol-gu, Incheon 22212, Republic of Korea; (G.Y.M.); (B.D.); (H.S.P.); (D.S.); (C.S.); (J.H.C.); (S.B.); (H.L.)
| | - Jung Hyun Choi
- Department of Biological Sciences and Bioengineering, Inha University, 100 Inha-ro, Michuhol-gu, Incheon 22212, Republic of Korea; (G.Y.M.); (B.D.); (H.S.P.); (D.S.); (C.S.); (J.H.C.); (S.B.); (H.L.)
- Division of Biomedical Metrology, Korea Research Institute of Standards and Science, 267 Gajeongno, Yuseong-gu, Daejeon 34113, Republic of Korea; (I.D.); (D.H.K.)
| | - Seha Bang
- Department of Biological Sciences and Bioengineering, Inha University, 100 Inha-ro, Michuhol-gu, Incheon 22212, Republic of Korea; (G.Y.M.); (B.D.); (H.S.P.); (D.S.); (C.S.); (J.H.C.); (S.B.); (H.L.)
| | - Hosu Lee
- Department of Biological Sciences and Bioengineering, Inha University, 100 Inha-ro, Michuhol-gu, Incheon 22212, Republic of Korea; (G.Y.M.); (B.D.); (H.S.P.); (D.S.); (C.S.); (J.H.C.); (S.B.); (H.L.)
| | - Il Doh
- Division of Biomedical Metrology, Korea Research Institute of Standards and Science, 267 Gajeongno, Yuseong-gu, Daejeon 34113, Republic of Korea; (I.D.); (D.H.K.)
| | - Dong Hyung Kim
- Division of Biomedical Metrology, Korea Research Institute of Standards and Science, 267 Gajeongno, Yuseong-gu, Daejeon 34113, Republic of Korea; (I.D.); (D.H.K.)
| | - Woo-jin Jeong
- Department of Biological Sciences and Bioengineering, Inha University, 100 Inha-ro, Michuhol-gu, Incheon 22212, Republic of Korea; (G.Y.M.); (B.D.); (H.S.P.); (D.S.); (C.S.); (J.H.C.); (S.B.); (H.L.)
- Department of Biological Engineering, Inha University, 100 Inha-ro, Michuhol-gu, Incheon 22212, Republic of Korea
| | - Jiyoon Bu
- Department of Biological Sciences and Bioengineering, Inha University, 100 Inha-ro, Michuhol-gu, Incheon 22212, Republic of Korea; (G.Y.M.); (B.D.); (H.S.P.); (D.S.); (C.S.); (J.H.C.); (S.B.); (H.L.)
- Department of Biological Engineering, Inha University, 100 Inha-ro, Michuhol-gu, Incheon 22212, Republic of Korea
- Biohybrid Systems Research Center, Inha University, 100 Inha-ro, Michuhol-gu, Incheon 22212, Republic of Korea
| |
Collapse
|
15
|
Hakala S, Hämäläinen A, Sandelin S, Giannareas N, Närvä E. Detection of Cancer Stem Cells from Patient Samples. Cells 2025; 14:148. [PMID: 39851576 PMCID: PMC11764358 DOI: 10.3390/cells14020148] [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: 12/30/2024] [Revised: 01/16/2025] [Accepted: 01/17/2025] [Indexed: 01/26/2025] Open
Abstract
The existence of cancer stem cells (CSCs) in various tumors has become increasingly clear in addition to their prominent role in therapy resistance, metastasis, and recurrence. For early diagnosis, disease progression monitoring, and targeting, there is a high demand for clinical-grade methods for quantitative measurement of CSCs from patient samples. Despite years of active research, standard measurement of CSCs has not yet reached clinical settings, especially in the case of solid tumors. This is because detecting this plastic heterogeneous population of cells is not straightforward. This review summarizes various techniques, highlighting their benefits and limitations in detecting CSCs from patient samples. In addition, methods designed to detect CSCs based on secreted and niche-associated signaling factors are reviewed. Spatial and single-cell methods for analyzing patient tumor tissues and noninvasive techniques such as liquid biopsy and in vivo imaging are discussed. Additionally, methods recently established in laboratories, preclinical studies, and clinical assays are covered. Finally, we discuss the characteristics of an ideal method as we look toward the future.
Collapse
Affiliation(s)
| | | | | | | | - Elisa Närvä
- Institute of Biomedicine and FICAN West Cancer Centre Laboratory, University of Turku and Turku University Hospital, FI-20520 Turku, Finland; (S.H.); (A.H.); (S.S.); (N.G.)
| |
Collapse
|
16
|
Takahashi Y, Ijiri Y, Fujino S, Elnaz N, Kishimoto A, Shirai K, Iwanaga S, Yanagida M, Bhagat AAS, Miyoshi N. Detection and Characterization of Circulating Tumor Cells in Colorectal Cancer Patients via Epithelial-Mesenchymal Transition Markers. Cancers (Basel) 2025; 17:303. [PMID: 39858085 PMCID: PMC11763958 DOI: 10.3390/cancers17020303] [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: 11/16/2024] [Revised: 01/12/2025] [Accepted: 01/14/2025] [Indexed: 01/27/2025] Open
Abstract
Background/Objectives: Liquid biopsy methods have gained prominence as minimally invasive tools to improve cancer treatment outcomes. Circulating tumor cells (CTCs) offer valuable insights into both primary and metastatic lesions. However, validating the CTC test results requires confirmation that the detected cells originate from cancer tissue. While studies have identified CTCs in colorectal cancer (CRC) patients using molecular markers, simultaneous validation of their cancer tissue origin remains unexplored. Methods: This study introduces a simple approach to detect adenomatous polyposis coli (APC) gene abnormalities alongside established CTC markers using a molecular imaging flow cytometer (MI-FCM). Given that APC gene abnormalities occur in 60-70% of CRC patients, their detection serves as strong evidence of cancer origin. Results: Our method achieved 92% concordance with DNA sequence analysis of tumor-derived cells. In a proof-of-concept study using 5 mL of whole blood from CRC patients, we observed a high frequency of cells exhibiting APC abnormalities, cytokeratin (CK), and vimentin (Vim) expression. Extending the study to 80 CRC patients across pathological stages I-IV confirmed CK and Vim as valid CTC markers. Three distinct cell populations were identified in blood: CK+/Vim-, CK+/Vim+, and CK-/Vim+. CTC number and frequency increased progressively with cancer stage. Conclusions: This is the first report demonstrating CK and Vim as effective markers for direct CTC detection in CRC patients. Our findings provide evidence-based validation of CTC markers, offering new insights and advancing approaches for patient care.
Collapse
Affiliation(s)
- Yusuke Takahashi
- Department of Central Research Laboratories, Sysmex Corporation, Kobe 651-2271, Japan; (Y.T.); (Y.I.); (N.E.); (A.K.); (K.S.); (S.I.); (M.Y.)
| | - Yuichi Ijiri
- Department of Central Research Laboratories, Sysmex Corporation, Kobe 651-2271, Japan; (Y.T.); (Y.I.); (N.E.); (A.K.); (K.S.); (S.I.); (M.Y.)
| | - Shiki Fujino
- Department of Gastroenterology, Central Clinical School, Monash University, Melbourne 3004, Australia;
- Innovative Oncology Research and Regenerative Medicine, Osaka International Cancer Institute, Osaka 541-8567, Japan
- Department of Gastroenterological Surgery, Graduate School of Medicine, Osaka University, Suita 565-0871, Japan
| | - Nakhaei Elnaz
- Department of Central Research Laboratories, Sysmex Corporation, Kobe 651-2271, Japan; (Y.T.); (Y.I.); (N.E.); (A.K.); (K.S.); (S.I.); (M.Y.)
| | - Ayuko Kishimoto
- Department of Central Research Laboratories, Sysmex Corporation, Kobe 651-2271, Japan; (Y.T.); (Y.I.); (N.E.); (A.K.); (K.S.); (S.I.); (M.Y.)
| | - Kentaro Shirai
- Department of Central Research Laboratories, Sysmex Corporation, Kobe 651-2271, Japan; (Y.T.); (Y.I.); (N.E.); (A.K.); (K.S.); (S.I.); (M.Y.)
| | - Shigeki Iwanaga
- Department of Central Research Laboratories, Sysmex Corporation, Kobe 651-2271, Japan; (Y.T.); (Y.I.); (N.E.); (A.K.); (K.S.); (S.I.); (M.Y.)
| | - Masatoshi Yanagida
- Department of Central Research Laboratories, Sysmex Corporation, Kobe 651-2271, Japan; (Y.T.); (Y.I.); (N.E.); (A.K.); (K.S.); (S.I.); (M.Y.)
| | - Ali Asgar S. Bhagat
- Biolidics Limited, Singapore 577177, Singapore;
- Institute for Health Innovation and Technology (iHealthtech), National University of Singapore, Singapore 119276, Singapore
- Department of Biomedical Engineering, College of Design and Engineering, National University of Singapore, Singapore 117575, Singapore
| | - Norikatsu Miyoshi
- Innovative Oncology Research and Regenerative Medicine, Osaka International Cancer Institute, Osaka 541-8567, Japan
- Department of Gastroenterological Surgery, Graduate School of Medicine, Osaka University, Suita 565-0871, Japan
| |
Collapse
|
17
|
Zhang C, Xu L, Miao X, Zhang D, Xie Y, Hu Y, Zhang Z, Wang X, Wu X, Liu Z, Zang W, He C, Li Z, Ren W, Chen T, Xu C, Zhang Y, Wu A, Lin J. Machine learning assisted dual-modal SERS detection for circulating tumor cells. Biosens Bioelectron 2025; 268:116897. [PMID: 39488132 DOI: 10.1016/j.bios.2024.116897] [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: 02/29/2024] [Revised: 10/05/2024] [Accepted: 10/29/2024] [Indexed: 11/04/2024]
Abstract
Detecting circulating tumor cells (CTCs) from blood has become a promising approach for cancer diagnosis. Surface-enhanced Raman Spectroscopy (SERS) has rapidly developed as a significant detection technology for CTCs, offering high sensitivity and selectivity. Encoded SERS bioprobes have gained attention due to their excellent specificity and ability to identify tumor cells using Raman signals. Machine learning has also made significant contributions to biomedical applications, especially in medical diagnosis. In this study, we developed a detection strategy combining encoded SERS bioprobes and machine learning models to identify CTCs. Dual-modal SERS bioprobes were designed and co-incubated with tumor cells by the "cocktail" method. An identification model for CTCs was constructed using principal component analysis (PCA) and the Random Forest classification algorithm. This innovative strategy endows SERS bioprobes with both effective magnetic separation and highly sensitive identification of CTCs, even at low concentrations of 2 cells/mL. It achieved a high detection rate of 98% for CTCs and effectively eliminated interference from peripheral WBCs. This simple and efficient strategy provides a new approach for CTCs detection and holds important significance for cancer diagnosis.
Collapse
Affiliation(s)
- Chenguang Zhang
- Ningbo Key Laboratory of Biomedical Imaging Probe Materials and Technology, Zhejiang International Cooperation Base of Biomedical Materials Technology and Application, Chinese Academy of Sciences (CAS) Key Laboratory of Magnetic Materials and Devices, Ningbo Cixi Institute of Biomedical Engineering, Zhejiang Engineering Research Center for Biomedical Materials, Ningbo Institute of Materials Technology and Engineering, Chinese Academy of Sciences, Ningbo, 315201, PR China; Cixi Biomedical Research Institute, Wenzhou Medical University, Zhejiang, PR China
| | - Lei Xu
- Ningbo Key Laboratory of Biomedical Imaging Probe Materials and Technology, Zhejiang International Cooperation Base of Biomedical Materials Technology and Application, Chinese Academy of Sciences (CAS) Key Laboratory of Magnetic Materials and Devices, Ningbo Cixi Institute of Biomedical Engineering, Zhejiang Engineering Research Center for Biomedical Materials, Ningbo Institute of Materials Technology and Engineering, Chinese Academy of Sciences, Ningbo, 315201, PR China
| | - Xinyu Miao
- Ningbo Key Laboratory of Biomedical Imaging Probe Materials and Technology, Zhejiang International Cooperation Base of Biomedical Materials Technology and Application, Chinese Academy of Sciences (CAS) Key Laboratory of Magnetic Materials and Devices, Ningbo Cixi Institute of Biomedical Engineering, Zhejiang Engineering Research Center for Biomedical Materials, Ningbo Institute of Materials Technology and Engineering, Chinese Academy of Sciences, Ningbo, 315201, PR China
| | - Dinghu Zhang
- Zhejiang Cancer Hospital, Hangzhou, 310022, PR China
| | - Yujiao Xie
- Ningbo Key Laboratory of Biomedical Imaging Probe Materials and Technology, Zhejiang International Cooperation Base of Biomedical Materials Technology and Application, Chinese Academy of Sciences (CAS) Key Laboratory of Magnetic Materials and Devices, Ningbo Cixi Institute of Biomedical Engineering, Zhejiang Engineering Research Center for Biomedical Materials, Ningbo Institute of Materials Technology and Engineering, Chinese Academy of Sciences, Ningbo, 315201, PR China.
| | - Yue Hu
- Ningbo Key Laboratory of Biomedical Imaging Probe Materials and Technology, Zhejiang International Cooperation Base of Biomedical Materials Technology and Application, Chinese Academy of Sciences (CAS) Key Laboratory of Magnetic Materials and Devices, Ningbo Cixi Institute of Biomedical Engineering, Zhejiang Engineering Research Center for Biomedical Materials, Ningbo Institute of Materials Technology and Engineering, Chinese Academy of Sciences, Ningbo, 315201, PR China; Cixi Biomedical Research Institute, Wenzhou Medical University, Zhejiang, PR China
| | - Zhouxu Zhang
- Ningbo Key Laboratory of Biomedical Imaging Probe Materials and Technology, Zhejiang International Cooperation Base of Biomedical Materials Technology and Application, Chinese Academy of Sciences (CAS) Key Laboratory of Magnetic Materials and Devices, Ningbo Cixi Institute of Biomedical Engineering, Zhejiang Engineering Research Center for Biomedical Materials, Ningbo Institute of Materials Technology and Engineering, Chinese Academy of Sciences, Ningbo, 315201, PR China; Cixi Biomedical Research Institute, Wenzhou Medical University, Zhejiang, PR China
| | - Xinfangzi Wang
- Ningbo Key Laboratory of Biomedical Imaging Probe Materials and Technology, Zhejiang International Cooperation Base of Biomedical Materials Technology and Application, Chinese Academy of Sciences (CAS) Key Laboratory of Magnetic Materials and Devices, Ningbo Cixi Institute of Biomedical Engineering, Zhejiang Engineering Research Center for Biomedical Materials, Ningbo Institute of Materials Technology and Engineering, Chinese Academy of Sciences, Ningbo, 315201, PR China; Cixi Biomedical Research Institute, Wenzhou Medical University, Zhejiang, PR China
| | - Xiaoxia Wu
- Zhejiang Cancer Hospital, Hangzhou, 310022, PR China
| | - Zhusheng Liu
- Ningbo Key Laboratory of Biomedical Imaging Probe Materials and Technology, Zhejiang International Cooperation Base of Biomedical Materials Technology and Application, Chinese Academy of Sciences (CAS) Key Laboratory of Magnetic Materials and Devices, Ningbo Cixi Institute of Biomedical Engineering, Zhejiang Engineering Research Center for Biomedical Materials, Ningbo Institute of Materials Technology and Engineering, Chinese Academy of Sciences, Ningbo, 315201, PR China; Cixi Biomedical Research Institute, Wenzhou Medical University, Zhejiang, PR China
| | - Wen Zang
- Ningbo Key Laboratory of Biomedical Imaging Probe Materials and Technology, Zhejiang International Cooperation Base of Biomedical Materials Technology and Application, Chinese Academy of Sciences (CAS) Key Laboratory of Magnetic Materials and Devices, Ningbo Cixi Institute of Biomedical Engineering, Zhejiang Engineering Research Center for Biomedical Materials, Ningbo Institute of Materials Technology and Engineering, Chinese Academy of Sciences, Ningbo, 315201, PR China; Cixi Biomedical Research Institute, Wenzhou Medical University, Zhejiang, PR China
| | - Chenglong He
- Ningbo Key Laboratory of Biomedical Imaging Probe Materials and Technology, Zhejiang International Cooperation Base of Biomedical Materials Technology and Application, Chinese Academy of Sciences (CAS) Key Laboratory of Magnetic Materials and Devices, Ningbo Cixi Institute of Biomedical Engineering, Zhejiang Engineering Research Center for Biomedical Materials, Ningbo Institute of Materials Technology and Engineering, Chinese Academy of Sciences, Ningbo, 315201, PR China; Cixi Biomedical Research Institute, Wenzhou Medical University, Zhejiang, PR China
| | - Zihou Li
- Ningbo Key Laboratory of Biomedical Imaging Probe Materials and Technology, Zhejiang International Cooperation Base of Biomedical Materials Technology and Application, Chinese Academy of Sciences (CAS) Key Laboratory of Magnetic Materials and Devices, Ningbo Cixi Institute of Biomedical Engineering, Zhejiang Engineering Research Center for Biomedical Materials, Ningbo Institute of Materials Technology and Engineering, Chinese Academy of Sciences, Ningbo, 315201, PR China; University of Chinese Academy of Sciences, Beijing, 100049, PR China
| | - Wenzhi Ren
- Ningbo Key Laboratory of Biomedical Imaging Probe Materials and Technology, Zhejiang International Cooperation Base of Biomedical Materials Technology and Application, Chinese Academy of Sciences (CAS) Key Laboratory of Magnetic Materials and Devices, Ningbo Cixi Institute of Biomedical Engineering, Zhejiang Engineering Research Center for Biomedical Materials, Ningbo Institute of Materials Technology and Engineering, Chinese Academy of Sciences, Ningbo, 315201, PR China; Cixi Biomedical Research Institute, Wenzhou Medical University, Zhejiang, PR China; University of Chinese Academy of Sciences, Beijing, 100049, PR China
| | - Tianxiang Chen
- Ningbo Key Laboratory of Biomedical Imaging Probe Materials and Technology, Zhejiang International Cooperation Base of Biomedical Materials Technology and Application, Chinese Academy of Sciences (CAS) Key Laboratory of Magnetic Materials and Devices, Ningbo Cixi Institute of Biomedical Engineering, Zhejiang Engineering Research Center for Biomedical Materials, Ningbo Institute of Materials Technology and Engineering, Chinese Academy of Sciences, Ningbo, 315201, PR China; Cixi Biomedical Research Institute, Wenzhou Medical University, Zhejiang, PR China; University of Chinese Academy of Sciences, Beijing, 100049, PR China
| | - Chen Xu
- Ningbo Key Laboratory of Biomedical Imaging Probe Materials and Technology, Zhejiang International Cooperation Base of Biomedical Materials Technology and Application, Chinese Academy of Sciences (CAS) Key Laboratory of Magnetic Materials and Devices, Ningbo Cixi Institute of Biomedical Engineering, Zhejiang Engineering Research Center for Biomedical Materials, Ningbo Institute of Materials Technology and Engineering, Chinese Academy of Sciences, Ningbo, 315201, PR China; Cixi Biomedical Research Institute, Wenzhou Medical University, Zhejiang, PR China; University of Chinese Academy of Sciences, Beijing, 100049, PR China
| | - Yujie Zhang
- Ningbo Key Laboratory of Biomedical Imaging Probe Materials and Technology, Zhejiang International Cooperation Base of Biomedical Materials Technology and Application, Chinese Academy of Sciences (CAS) Key Laboratory of Magnetic Materials and Devices, Ningbo Cixi Institute of Biomedical Engineering, Zhejiang Engineering Research Center for Biomedical Materials, Ningbo Institute of Materials Technology and Engineering, Chinese Academy of Sciences, Ningbo, 315201, PR China; Cixi Biomedical Research Institute, Wenzhou Medical University, Zhejiang, PR China; University of Chinese Academy of Sciences, Beijing, 100049, PR China.
| | - Aiguo Wu
- Ningbo Key Laboratory of Biomedical Imaging Probe Materials and Technology, Zhejiang International Cooperation Base of Biomedical Materials Technology and Application, Chinese Academy of Sciences (CAS) Key Laboratory of Magnetic Materials and Devices, Ningbo Cixi Institute of Biomedical Engineering, Zhejiang Engineering Research Center for Biomedical Materials, Ningbo Institute of Materials Technology and Engineering, Chinese Academy of Sciences, Ningbo, 315201, PR China; Cixi Biomedical Research Institute, Wenzhou Medical University, Zhejiang, PR China; University of Chinese Academy of Sciences, Beijing, 100049, PR China.
| | - Jie Lin
- Ningbo Key Laboratory of Biomedical Imaging Probe Materials and Technology, Zhejiang International Cooperation Base of Biomedical Materials Technology and Application, Chinese Academy of Sciences (CAS) Key Laboratory of Magnetic Materials and Devices, Ningbo Cixi Institute of Biomedical Engineering, Zhejiang Engineering Research Center for Biomedical Materials, Ningbo Institute of Materials Technology and Engineering, Chinese Academy of Sciences, Ningbo, 315201, PR China; Cixi Biomedical Research Institute, Wenzhou Medical University, Zhejiang, PR China; University of Chinese Academy of Sciences, Beijing, 100049, PR China.
| |
Collapse
|
18
|
Schreier S, Budchart P, Borwornpinyo S, Adireklarpwong L, Chirappapha P, Triampo W, Lertsithichai P. Rare Cell Population Analysis in Early-Stage Breast Cancer Patients. Breast Cancer (Auckl) 2025; 19:11782234241310596. [PMID: 39803593 PMCID: PMC11724413 DOI: 10.1177/11782234241310596] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2024] [Accepted: 12/09/2024] [Indexed: 01/16/2025] Open
Abstract
Background Circulating rare cells participate in breast cancer evolution as systemic components of the disease and thus, are a source of theranostic information. Exploration of cancer-associated rare cells is in its infancy. Objectives We aimed to investigate and classify abnormalities in the circulating rare cell population among early-stage breast cancer patients using fluorescence marker identification and cytomorphology. In addition, we sought to determine the dependency of these markers on the presence of tumors. Design We evaluated the validity of a multi-rare-cell detection platform and demonstrated the utility of a specific rare cell subset as a novel approach to characterize the breast cancer system. Sampling was conducted both before and after tumor resection. Methods Linearity of the Rarmax platform was established using a spike-in approach. The platform includes red blood cell lysis, leukocyte depletion and high-resolution fluorescence image recording. Rare cell analysis was conducted on 28 samples (before and after surgery) from 14 patients with breast cancer, 20 healthy volunteers and 9 noncancer control volunteers. In-depth identification of rare cells, including circulating tumor cells, endothelial-like cells, erythroblasts, and inflammation-associated cells, was performed using a phenotype and morphology-based classification system. Results The platform performed linearly over a range of 5 to 950 spiked cells, with an average recovery of 84.6%. Circulating epithelial and endothelial-like cell subsets have been demonstrated to be associated with or independent of cancer with tumor presence. Furthermore, certain cell profile patterns may be associated with treatment-related adverse effects. The sensitivity in detecting tumor-presence and cancer-associated abnormality before surgery was 43% and 85.7%, respectively, and the specificity was 100% and 96.6%, respectively. Conclusion This study supports the idea of a cancer-associated rare cell abnormality to represent tumor entities as well as systemic cancer. The latter is independent of the apparent clinical cancer.
Collapse
Affiliation(s)
- Stefan Schreier
- School of Bioinnovation and Bio-based Product Intelligence, Faculty of Science, Mahidol University, Bangkok, Thailand
- MUSC Centre of Excellence in STEM Education, School of Bioinnovation and Bio-based Product Intelligence, Faculty of Science, Mahidol University, Bangkok, Thailand
- Premise Biosystems Co., Ltd. Bangkok, Thailand
| | | | - Suparerk Borwornpinyo
- Premise Biosystems Co., Ltd. Bangkok, Thailand
- Excellent Center for Drug Discovery, Faculty of Science, Mahidol University, Bangkok, Thailand
| | - Lakkana Adireklarpwong
- Department of Surgery, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Bangkok, Thailand
| | - Prakasit Chirappapha
- Department of Surgery, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Bangkok, Thailand
| | - Wannapong Triampo
- MUSC Centre of Excellence in STEM Education, School of Bioinnovation and Bio-based Product Intelligence, Faculty of Science, Mahidol University, Bangkok, Thailand
- Biophysics Lab, Department of Physics, Faculty of Science, Mahidol University, Bangkok, Thailand
| | - Panuwat Lertsithichai
- Department of Surgery, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Bangkok, Thailand
| |
Collapse
|
19
|
Hoshi Y, Matsuda S, Takeuchi M, Kawakubo H, Kitagawa Y. Liquid Biopsy and Multidisciplinary Treatment for Esophageal Cancer. Cancers (Basel) 2025; 17:196. [PMID: 39857978 PMCID: PMC11763614 DOI: 10.3390/cancers17020196] [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: 12/02/2024] [Revised: 01/02/2025] [Accepted: 01/07/2025] [Indexed: 01/27/2025] Open
Abstract
Esophageal cancer (EC) is one of the leading causes of cancer-related deaths globally. Surgery is the standard treatment for resectable EC after preoperative chemoradiotherapy or chemotherapy, followed by postoperative adjuvant chemotherapy in certain cases. Upper gastrointestinal endoscopy and computed tomography (CT) are predominantly performed to evaluate the efficacy of these treatments, but their sensitivity and accuracy for evaluating minimal residual disease remain unsatisfactory, thereby requiring the development of alternative methods. In recent years, interest has been increasing in using liquid biopsy to assess treatment responses. Liquid biopsy is a noninvasive technology for detecting cell components in the blood and other body fluids. It involves collecting a small sample of body fluid, which is then analyzed for the presence of components, including circulating tumor DNA (ctDNA), microRNA (miRNA), or circulating tumor cells (CTCs). Further, ctDNA and miRNA are analyzed with various techniques, including digital polymerase chain reaction (dPCR) and next-generation sequencing (NGS). CTCs are isolated by determining surface antigens using immunomagnetic techniques or by filtering the blood according to cell size and rigidity. Several studies indicate that investigating these materials helps predict EC prognosis and recurrence and possibly stratifies high-risk groups. Liquid biopsy may also apply to the selection of cases that have achieved a complete response through preoperative treatment to prevent surgery and preserve the esophagus, as well as identifying the suitability of postoperative chemotherapy and the timing of conversion surgery for unresectable EC. The potential of liquid biopsy to enhance treatment decisions will further advance EC treatment.
Collapse
Affiliation(s)
| | - Satoru Matsuda
- Department of Surgery, Keio University School of Medicine, Tokyo 160-8582, Japan
| | | | | | | |
Collapse
|
20
|
Cieslik SA, Zafra AG, Driemel C, Sudarsanam M, Cieslik JP, Flügen G, Dizdar L, Krieg A, Vaghiri S, Ashmawy H, Fung S, Wilms M, Terstappen LWMM, Nanou A, Neubauer H, Rahbari NN, Knoefel WT, Stoecklein NH, Neves RPL. Phenotypic diversity of CTCs and tdEVs in liquid biopsies of tumour-draining veins is linked to poor prognosis in colorectal cancer. J Exp Clin Cancer Res 2025; 44:9. [PMID: 39773651 PMCID: PMC11708080 DOI: 10.1186/s13046-024-03259-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2024] [Accepted: 12/18/2024] [Indexed: 01/11/2025] Open
Abstract
BACKGROUND Circulating tumour cells (CTCs) and tumour-derived extracellular vesicles (tdEVs) have great potential for monitoring therapy response and early detection of tumour relapse, facilitating personalized adjuvant therapeutic strategies. However, their low abundance in peripheral blood limits their informative value. In this study, we explored the presence of CTCs and tdEVs collected intraoperatively from a tumour-draining vein (DV) and via a central venous catheter (CVC) prior to tumour resection. METHODS CellSearch analyses of 395 blood samples from 306 patients with gastrointestinal tumours and 93 blood samples from healthy donors were used to establish and validate gates for the automated detection of CTCs and tdEVs with ACCEPT software and R scripts. The selected gate settings were applied to 227 samples of 142 patients with colorectal cancer (CRC) from two independent collectives. Phenotypic features were obtained via numeric analysis of their fluorescence signals (e.g. size, shape, and intensity) and were used for calculating diversity using Shannon index (SI) of clusters generated via the k-means algorithm after Uniform Manifold Approximation and Projection (UMAP) pre-processing, and standard deviation (SD). RESULTS CTCs and tdEVs were more abundant in the DV samples compared to CVC samples (p < 0.05). tdEVs were detected in higher numbers than CTCs in both compartments. Importantly, tdEVs in CVCs were associated with tumor spread, whereas CTCs in DVs were linked to tumor size. In both compartments, the prognostic value of tdEVs for overall survival (OS) surpassed that of CTCs, as demonstrated by univariate, multivariate, and Kaplan-Meier analyses. CTCs and tdEVs in DVs were phenotypically distinct, being larger, more eccentric, and displaying stronger cytokeratin intensities (p < 0.05) compared to those in CVC samples. Furthermore, increased diversity in CTC and tdEV phenotypes was significantly associated with shorter survival, validating the prognostic relevance of the SD-diversity metric. CONCLUSION Our study demonstrates that DV sampling significantly enhances the detection of prognostically relevant CTCs and tdEVs in CRC patients, underscoring the superior prognostic significance of tdEVs compared to CTCs. Importantly, the combined phenotypic diversity of both markers emerges as a more powerful biomarker than their enumeration alone. These findings suggest that comprehensive, automated analysis of CTCs and tdEVs in DVs may open new avenues for tailoring individualized therapies in CRC patients.
Collapse
Affiliation(s)
- Stefan A Cieslik
- Department of General, Visceral and Pediatric Surgery, University Hospital and Medical Faculty of Heinrich-Heine University Düsseldorf, Moorenstr. 5, 40225, Düsseldorf, Germany
| | - Andrés G Zafra
- Department of General, Visceral and Pediatric Surgery, University Hospital and Medical Faculty of Heinrich-Heine University Düsseldorf, Moorenstr. 5, 40225, Düsseldorf, Germany
| | - Christiane Driemel
- Department of General, Visceral and Pediatric Surgery, University Hospital and Medical Faculty of Heinrich-Heine University Düsseldorf, Moorenstr. 5, 40225, Düsseldorf, Germany
| | - Monica Sudarsanam
- Department of General, Visceral and Pediatric Surgery, University Hospital and Medical Faculty of Heinrich-Heine University Düsseldorf, Moorenstr. 5, 40225, Düsseldorf, Germany
| | - Jan-Philipp Cieslik
- Department of Obstetrics and Gynecology, University Hospital and Medical Faculty of the Heinrich-Heine University Düsseldorf, Moorenstr. 5, 40225, Düsseldorf, Germany
| | - Georg Flügen
- Department of General, Visceral and Pediatric Surgery, University Hospital and Medical Faculty of Heinrich-Heine University Düsseldorf, Moorenstr. 5, 40225, Düsseldorf, Germany
| | - Levent Dizdar
- Department of General, Visceral and Pediatric Surgery, University Hospital and Medical Faculty of Heinrich-Heine University Düsseldorf, Moorenstr. 5, 40225, Düsseldorf, Germany
| | - Andreas Krieg
- Department of General, Visceral and Pediatric Surgery, University Hospital and Medical Faculty of Heinrich-Heine University Düsseldorf, Moorenstr. 5, 40225, Düsseldorf, Germany
- Department of General and Visceral Surgery, Thoracic Surgery and Proctology, Medical Campus OWL, University Hospital Herford, Ruhr University Bochum, 32049, Herford, Germany
| | - Sascha Vaghiri
- Department of General, Visceral and Pediatric Surgery, University Hospital and Medical Faculty of Heinrich-Heine University Düsseldorf, Moorenstr. 5, 40225, Düsseldorf, Germany
| | - Hany Ashmawy
- Department of General, Visceral and Pediatric Surgery, University Hospital and Medical Faculty of Heinrich-Heine University Düsseldorf, Moorenstr. 5, 40225, Düsseldorf, Germany
| | - Stephen Fung
- Department of General, Visceral and Pediatric Surgery, University Hospital and Medical Faculty of Heinrich-Heine University Düsseldorf, Moorenstr. 5, 40225, Düsseldorf, Germany
| | - Miriam Wilms
- Department of General, Visceral and Pediatric Surgery, University Hospital and Medical Faculty of Heinrich-Heine University Düsseldorf, Moorenstr. 5, 40225, Düsseldorf, Germany
| | - Leon W M M Terstappen
- Department of General, Visceral and Pediatric Surgery, University Hospital and Medical Faculty of Heinrich-Heine University Düsseldorf, Moorenstr. 5, 40225, Düsseldorf, Germany
- Department of Medical Cell BioPhysics, Faculty of Science and Technology, University of Twente, Enschede, 7522 NH, The Netherlands
- Decisive Science, Amsterdam, The Netherlands
| | - Afroditi Nanou
- Department of Medical Cell BioPhysics, Faculty of Science and Technology, University of Twente, Enschede, 7522 NH, The Netherlands
| | - Hans Neubauer
- Department of Obstetrics and Gynecology, University Hospital and Medical Faculty of the Heinrich-Heine University Düsseldorf, Moorenstr. 5, 40225, Düsseldorf, Germany
| | - Nuh N Rahbari
- Department of General and Visceral Surgery, University Hospital Ulm, Albert-Einstein-Allee 23, 89081, Ulm, Germany
| | - Wolfram T Knoefel
- Department of General, Visceral and Pediatric Surgery, University Hospital and Medical Faculty of Heinrich-Heine University Düsseldorf, Moorenstr. 5, 40225, Düsseldorf, Germany
| | - Nikolas H Stoecklein
- Department of General, Visceral and Pediatric Surgery, University Hospital and Medical Faculty of Heinrich-Heine University Düsseldorf, Moorenstr. 5, 40225, Düsseldorf, Germany.
| | - Rui P L Neves
- Department of General, Visceral and Pediatric Surgery, University Hospital and Medical Faculty of Heinrich-Heine University Düsseldorf, Moorenstr. 5, 40225, Düsseldorf, Germany
| |
Collapse
|
21
|
Hattori M. Role of circulating tumor cells in breast cancer. Breast Cancer 2025; 32:26-32. [PMID: 39656381 DOI: 10.1007/s12282-024-01651-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2024] [Accepted: 11/17/2024] [Indexed: 01/11/2025]
Abstract
Circulating tumor cells (CTCs) are tumor cells that shed from the primary tumor or metastatic loci, intravasate, and circulate in the bloodstream. CTCs have been suggested to play a major role in the metastatic spread of cancer, constantly shedding from tumors during proliferation or as a result of mechanical insults. Breast cancer (BC) is one of the most representative tumors in CTC research, with several studies conducted on its clinical validity and utility in both early and advanced BC (EBC and ABC, respectively). The assessment of the number and molecular profiles of CTCs is expected to provide a more tailored therapy for patients with BC. The detection of CTCs is usually dependent on molecular markers, and epithelial cell adhesion molecules are widely used. Although the CellSearch® technology has been widely utilized for CTC detection, recent advances have led to the development of novel detection methods, facilitating further molecular analysis. In this review, we discuss the clinical applications of CTCs, current status of research, and efforts to incorporate CTC analysis into clinical practice. Additionally, we discuss potential challenges and future directions for integrating CTC analysis into clinical practice.
Collapse
Affiliation(s)
- Masaya Hattori
- Department of Breast Oncology, Aichi Cancer Center, 1-1 Kanokoden Chikusa-ku, Nagoya, 464-8681, Japan.
| |
Collapse
|
22
|
Eboshida N, Hamada A, Higaki M, Obayashi F, Ito N, Yamasaki S, Tani R, Shintani T, Koizumi K, Yanamoto S. Potential role of circulating tumor cells and cell-free DNA as biomarkers in oral squamous cell carcinoma: A prospective single-center study. PLoS One 2024; 19:e0309178. [PMID: 39729421 DOI: 10.1371/journal.pone.0309178] [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: 05/30/2024] [Accepted: 08/06/2024] [Indexed: 12/29/2024] Open
Abstract
Metastasis in patients with oral squamous cell carcinoma has been associated with a poor prognosis. However, sensitive and reliable tests for monitoring their occurrence are unavailable, with the exception of PET-CT. Circulating tumor cells and cell-free DNA have emerged as promising biomarkers for determining treatment efficacy and as prognostic predictors in solid tumors such as breast cancer and colorectal cancer. Hence, this study aimed to determine the potential role of liquid biopsy, circulating tumor cells, and cell-free DNA as biomarkers of oral squamous cell carcinoma. Thirteen patients with primary oral squamous cell carcinoma who visited our hospital between 2022 and 2023 were recruited, and plasma samples were collected from each patient preoperatively and postoperatively. We examined the relationship between the prognosis, the number of circulating tumor cells per four milliliters of peripheral blood, and the amount of cell-free DNA per milliliter of serum or the gene mutation in cell-free DNA. We observed no correlation between the number of preoperative circulating tumor cells and metastatic events. However, the number of circulating tumor cell clusters or the amount of preoperative cell-free DNA in metastatic cases was higher than that in non-metastatic cases. In oral squamous cell carcinoma, circulating tumor cell clusters or cell-free DNA levels may help inform management decisions regarding metastasis. However, further studies are required to provide a possible window for therapeutic interventions.
Collapse
Affiliation(s)
- Natsuki Eboshida
- Department of Oral Oncology, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Atsuko Hamada
- Department of Oral Oncology, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Mirai Higaki
- Department of Oral Oncology, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Fumitaka Obayashi
- Department of Oral Oncology, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Nanako Ito
- Department of Oral Oncology, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Sachiko Yamasaki
- Department of Oral Oncology, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Ryouji Tani
- Department of Oral Oncology, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Tomoaki Shintani
- Center of Oral Clinical Examination, Hiroshima University Hospital, Hiroshima, Japan
| | - Koichi Koizumi
- Department of Oral Oncology, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Souichi Yanamoto
- Department of Oral Oncology, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| |
Collapse
|
23
|
Zhou Z, Cai S, Zhou X, Zhao W, Sun J, Zhou Z, Yang Z, Li W, Wang Z, Zou H, Fu H, Wang X, Khoo BL, Yang M. Circulating Tumor Cells Culture: Methods, Challenges, and Clinical Applications. SMALL METHODS 2024:e2401026. [PMID: 39726345 DOI: 10.1002/smtd.202401026] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/06/2024] [Revised: 11/10/2024] [Indexed: 12/28/2024]
Abstract
Circulating tumor cells (CTCs) play a pivotal role in cancer metastasis and hold considerable potential for clinical diagnosis, therapeutic monitoring, and prognostic evaluation. Nevertheless, the limited quantity of CTCs in liquid biopsy samples poses challenges for comprehensive downstream analysis. In vitro culture of CTCs can effectively address the issue of insufficient CTC numbers. Furthermore, research based on CTC cell lines serves as a valuable complement to traditional cancer cell line-based research. While numerous reports exist on CTC in vitro culture and even the establishment of CTC cell lines, the methods used vary, leading to disparate culture outcomes. This review presents the developmental history and current status of CTC in vitro culture research. Additionally, the culture strategies applied in different methods and analyzed the impact of various steps on culture outcomes are compared. Overall, the review indicates that while the short-term culture of CTCs is relatively straightforward, long-term culture success has been achieved for various specific cancer types but still faces challenges. Further optimization of efficient and widely applicable culture strategies is needed. Additionally, ongoing applications of CTC in vitro culture are summarized, highlighting the potential of expanded CTCs for drug susceptibility testing and as therapeutic tools in personalized treatment.
Collapse
Affiliation(s)
- Zhengdong Zhou
- Department of Precision Diagnostic and Therapeutic Technology, City University of Hong Kong Shenzhen Futian Research Institute, Shenzhen, 518057, China
- Department of Biomedical Sciences, Tung Biomedical Sciences Centre, City University of Hong Kong, Hong Kong SAR, 999077, China
- Key Laboratory of Biochip Technology, Biotech and Health Centre, Shenzhen Research Institute of City University of Hong Kong, Shenzhen, 518057, China
| | - Songhua Cai
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital & Shenzhen Hospital Chinese Academy of Medical Sciences and Peking Union Medical College, Shenzhen, 518116, China
| | - Xiaoyu Zhou
- Department of Precision Diagnostic and Therapeutic Technology, City University of Hong Kong Shenzhen Futian Research Institute, Shenzhen, 518057, China
- Department of Biomedical Sciences, Tung Biomedical Sciences Centre, City University of Hong Kong, Hong Kong SAR, 999077, China
- Key Laboratory of Biochip Technology, Biotech and Health Centre, Shenzhen Research Institute of City University of Hong Kong, Shenzhen, 518057, China
| | - Wei Zhao
- Department of Biomedical Sciences, Tung Biomedical Sciences Centre, City University of Hong Kong, Hong Kong SAR, 999077, China
| | - Jiayu Sun
- Department of Precision Diagnostic and Therapeutic Technology, City University of Hong Kong Shenzhen Futian Research Institute, Shenzhen, 518057, China
- Department of Biomedical Sciences, Tung Biomedical Sciences Centre, City University of Hong Kong, Hong Kong SAR, 999077, China
| | - Zhihang Zhou
- Department of Biomedical Sciences, Tung Biomedical Sciences Centre, City University of Hong Kong, Hong Kong SAR, 999077, China
| | - Zihan Yang
- Department of Precision Diagnostic and Therapeutic Technology, City University of Hong Kong Shenzhen Futian Research Institute, Shenzhen, 518057, China
- Department of Biomedical Sciences, Tung Biomedical Sciences Centre, City University of Hong Kong, Hong Kong SAR, 999077, China
| | - Wenxiu Li
- Department of Precision Diagnostic and Therapeutic Technology, City University of Hong Kong Shenzhen Futian Research Institute, Shenzhen, 518057, China
- Department of Biomedical Sciences, Tung Biomedical Sciences Centre, City University of Hong Kong, Hong Kong SAR, 999077, China
| | - Zhe Wang
- The First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou, 510080, China
| | - Heng Zou
- Cellomics (Shenzhen) Limited, Shenzhen, 518118, China
| | - Huayang Fu
- Cellomics (Shenzhen) Limited, Shenzhen, 518118, China
| | - Xicheng Wang
- The First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou, 510080, China
| | - Bee Luan Khoo
- Department of Precision Diagnostic and Therapeutic Technology, City University of Hong Kong Shenzhen Futian Research Institute, Shenzhen, 518057, China
- Department of Biomedical Engineering, City University of Hong Kong, Hong Kong SAR, 999077, China
| | - Mengsu Yang
- Department of Precision Diagnostic and Therapeutic Technology, City University of Hong Kong Shenzhen Futian Research Institute, Shenzhen, 518057, China
- Department of Biomedical Sciences, Tung Biomedical Sciences Centre, City University of Hong Kong, Hong Kong SAR, 999077, China
- Key Laboratory of Biochip Technology, Biotech and Health Centre, Shenzhen Research Institute of City University of Hong Kong, Shenzhen, 518057, China
| |
Collapse
|
24
|
Kang S, Skapek S, Krishnan S, Gambhir SS, Zeng Y, Zhou Q, Zaman R. A Novel Approach to Harnessing Acoustic A-Lines to Detect Circulating Tumor Cells in Flowing Blood. NANO LETTERS 2024; 24:15615-15622. [PMID: 39556103 DOI: 10.1021/acs.nanolett.4c03982] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2024]
Abstract
Circulating tumor cells (CTCs) are associated with tumor burden and treatment response and, as hallmarks of the initiation of tumor dissemination, can predict the likelihood of metastatic progression before widespread tumors can be detected by standard anatomic imaging. However, early diagnosis of recurrence through the detection of CTCs is limited by their low prevalence in blood and the limited sensitivity of existing technologies. To address these challenges, we investigated the use of ultrasound and targeted microbubbles (MBs) for early CTC detection. While MBs have been used in cardiovascular/molecular tumor imaging, there is limited research on their acoustic properties when bound to CTCs. We developed a hydrophone system for detecting characteristic A-lines from CTCs encapsulating MBs. Our study is the first to identify distinctive characteristics in the acoustic frequency response of MBs bound to different cancer CTCs using in vitro suspensions and in vivo mice that will benefit metastatic cancer detection and management.
Collapse
Affiliation(s)
- Shu Kang
- Department of Biomedical Engineering, University of Texas Southwestern Medical Center, Dallas, Texas 75235, United States
| | - Stephen Skapek
- Department of Pediatrics, Division of Hematology/Oncology, Duke University School of Medicine, Durham, North Carolina 27710, United States
| | - Sunil Krishnan
- Lilian L. Smith Department of Neurosurgery, UT Health Science Center, Houston, Texas 77054, United States
| | - Sanjiv S Gambhir
- Department of Radiology, Stanford University School of Medicine, Stanford, California 94304, United States
| | - Yushun Zeng
- Alfred E. Mann Department of Biomedical Engineering, Viterbi School of Engineering, University of Southern California, Los Angeles, California 90089, United States
| | - Qifa Zhou
- Alfred E. Mann Department of Biomedical Engineering, Viterbi School of Engineering, University of Southern California, Los Angeles, California 90089, United States
| | - Raiyan Zaman
- Department of Biomedical Engineering, University of Texas Southwestern Medical Center, Dallas, Texas 75235, United States
| |
Collapse
|
25
|
Ma L, Guo H, Zhao Y, Liu Z, Wang C, Bu J, Sun T, Wei J. Liquid biopsy in cancer current: status, challenges and future prospects. Signal Transduct Target Ther 2024; 9:336. [PMID: 39617822 PMCID: PMC11609310 DOI: 10.1038/s41392-024-02021-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2024] [Revised: 09/10/2024] [Accepted: 10/14/2024] [Indexed: 12/06/2024] Open
Abstract
Cancer has a high mortality rate across the globe, and tissue biopsy remains the gold standard for tumor diagnosis due to its high level of laboratory standardization, good consistency of results, relatively stable samples, and high accuracy of results. However, there are still many limitations and drawbacks in the application of tissue biopsy in tumor. The emergence of liquid biopsy provides new ideas for early diagnosis and prognosis of tumor. Compared with tissue biopsy, liquid biopsy has many advantages in the diagnosis and treatment of various types of cancer, including non-invasive, quickly and so on. Currently, the application of liquid biopsy in tumor detection has received widely attention. It is now undergoing rapid progress, and it holds significant potential for future applications. Around now, liquid biopsies encompass several components such as circulating tumor cells, circulating tumor DNA, exosomes, microRNA, circulating RNA, tumor platelets, and tumor endothelial cells. In addition, advances in the identification of liquid biopsy indicators have significantly enhanced the possibility of utilizing liquid biopsies in clinical settings. In this review, we will discuss the application, advantages and challenges of liquid biopsy in some common tumors from the perspective of diverse systems of tumors, and look forward to its future development prospects in the field of cancer diagnosis and treatment.
Collapse
Affiliation(s)
- Liwei Ma
- Department of Clinical Laboratory, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China.
- Key Clinical Laboratory of Henan province, Zhengzhou, Henan, China.
| | - Huiling Guo
- Department of Clinical Laboratory, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
- Key Clinical Laboratory of Henan province, Zhengzhou, Henan, China
| | - Yunxiang Zhao
- Department of Neurosurgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Zhibo Liu
- Department of Clinical Laboratory, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
- Key Clinical Laboratory of Henan province, Zhengzhou, Henan, China
| | - Chenran Wang
- Department of Clinical Laboratory, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
- Key Clinical Laboratory of Henan province, Zhengzhou, Henan, China
| | - Jiahao Bu
- Department of Neurosurgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Ting Sun
- Department of Clinical Laboratory, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China.
- Key Clinical Laboratory of Henan province, Zhengzhou, Henan, China.
| | - Jianwei Wei
- Department of Neurosurgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China.
| |
Collapse
|
26
|
Liu C, Cai Y, Mou S. Liquid biopsy in lung cancer: The role of circulating tumor cells in diagnosis, treatment, and prognosis. Biomed Pharmacother 2024; 181:117726. [PMID: 39612860 DOI: 10.1016/j.biopha.2024.117726] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/01/2024] Open
Abstract
Despite numerous therapeutic advancements, such as immune checkpoint inhibitors, lung cancer continues to be the leading cause of cancer-related mortality. Therefore, the identification of cancer at an early stage is becoming a significant subject in contemporary oncology. Despite significant advancements in early detection tactics in recent decades, they continue to provide challenges because of the inconspicuous symptoms observed during the early stages of the primary tumor. Presently, tumor biomarkers and imaging techniques are extensively employed across different forms of cancer. Nevertheless, every approach has its own set of constraints. In certain instances, the detriments outweigh the advantages. Hence, there is an urgent need to enhance early detection methods. Currently, liquid biopsy is considered more flexible and not intrusive method in comparison to conventional test for early detection. Circulating tumor cells (CTCs) are crucial components of liquid biopsy and have a pivotal function in the spread and formation of secondary tumors. These indicators show great promise in the early identification of cancer. This study presents a comprehensive examination of the methodologies employed for the isolation and enrichment of circulating tumor cells (CTCs) in lung cancer. Additionally, it explores the formation of clusters of CTCs, which have a pivotal function in facilitating the effective dissemination of cancer to distant organs. In addition, we discuss the importance of CTCs in the detection, treatment, and prognosis of lung cancer.
Collapse
Affiliation(s)
- Chibo Liu
- Department of Clinical Laboratory, Taizhou Municipal Hospital, Taizhou, Zhejiang, China.
| | - Yanqun Cai
- Department of Clinical Laboratory, Taizhou Municipal Hospital, Taizhou, Zhejiang, China
| | - Sihua Mou
- Department of Clinical Laboratory, Taizhou Municipal Hospital, Taizhou, Zhejiang, China.
| |
Collapse
|
27
|
Wenta R, Richert J, Muchlińska A, Senkus E, Suchodolska G, Łapińska-Szumczyk S, Domżalski P, Miszewski K, Matuszewski M, Dziadziuszko R, Supernat A, Żaczek A, Bednarz-Knoll N. Measurable morphological features of single circulating tumor cells in selected solid tumors-A pilot study. Cytometry A 2024; 105:883-892. [PMID: 39498617 DOI: 10.1002/cyto.a.24906] [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: 05/15/2024] [Revised: 10/16/2024] [Accepted: 10/22/2024] [Indexed: 12/15/2024]
Abstract
Liquid biopsies developed into a range of sensitive technologies aiming to analyze for example, circulating tumor cells (CTCs) in peripheral blood, which significantly deepens understanding of the metastatic process. Nevertheless, examination of CTCs is mostly limited to their enumeration and usually only 2-3 markers-based phenotyping, not offering yet sufficient insight into their biology. In contrast, quantitative analysis of their morphological details might extend our knowledge about dissemination and even improve CTC isolation or label-free identification methods dependent on their physical features such as size, and deformability. Current study was conducted to describe CTCs' and their size, shape, presence of protrusions, and micronuclei across various types of cancers (lung, n = 29; ovarian, n = 24, breast, n = 54; and prostate, n = 33). Epithelial (pan-keratins), mesenchymal (vimentin), and two exclusion markers were used to identify CTCs and classify them into four epithelial and epithelial-mesenchymal transition-related phenotypes using standardized and throughput method, imaging flow cytometry. The morphological characteristics of CTCs, including their nuclei, such as circularity, the maximum, and minimum diagonal values were determined using an open-source software QuPath. On average, detected CTCs (n = 1156) were larger, and more irregular in shape compared to leukocytes/endothelial cells (n = 400). Epithelial and mesenchymal CTCs had the largest (median = 18.2 μm) and the smallest diameter (median = 10.4 μm), respectively. In terms of cancer-specific variations, the largest CTCs were identified in lung cancer, whereas the smallest-in prostate and breast cancers. Epithelial CTCs and those negative for both epithelial and mesenchymal markers exhibited the highest degree of elongation, whereas mesenchymal CTCs were the most irregular in shape. Protrusions and micronuclei were observed extremely rarely within CTCs of breast and prostate cancer (0.6%-0.8% of CTCs). Micronuclei were observed only in epithelial and epithelial-mesenchymal CTCs. This study underscores the significant variability in the morphological features of CTCs in relation to their phenotypic classification or even the particular organ of origin, potentially influencing for example, size-dependent CTC isolation methods. It demonstrates for the first time the morphological measurements of CTCs undergoing epithelial-mesenchymal transition, and some specific morphological details (i.e., protrusions, micronuclei) within CTCs in general.
Collapse
Affiliation(s)
- Robert Wenta
- Laboratory of Translational Oncology, Intercollegiate Faculty of Biotechnology, Medical University of Gdańsk, Gdańsk, Poland
| | - Julia Richert
- Laboratory of Translational Oncology, Intercollegiate Faculty of Biotechnology, Medical University of Gdańsk, Gdańsk, Poland
| | - Anna Muchlińska
- Laboratory of Translational Oncology, Intercollegiate Faculty of Biotechnology, Medical University of Gdańsk, Gdańsk, Poland
| | - Elżbieta Senkus
- Department of Oncology and Radiotherapy, Medical University of Gdańsk, Gdańsk, Poland
| | - Grażyna Suchodolska
- Department of Oncology and Radiotherapy, Medical University of Gdańsk, Gdańsk, Poland
| | - Sylwia Łapińska-Szumczyk
- Department of Gynaecology, Gynaecological Oncology and Gynaecological Endocrinology, Medical University of Gdańsk, Gdańsk, Poland
| | - Paweł Domżalski
- Early Phase Clinical Trials Centre, Medical University of Gdańsk, Gdańsk, Poland
- Department of Histology, Medical University of Gdańsk, Gdańsk, Poland
| | - Kevin Miszewski
- Department of Urology, Medical University of Gdańsk, Gdańsk, Poland
| | | | - Rafał Dziadziuszko
- Department of Oncology and Radiotherapy, Medical University of Gdańsk, Gdańsk, Poland
| | - Anna Supernat
- Laboratory of Translational Oncology, Intercollegiate Faculty of Biotechnology, Medical University of Gdańsk, Gdańsk, Poland
| | - Anna Żaczek
- Laboratory of Translational Oncology, Intercollegiate Faculty of Biotechnology, Medical University of Gdańsk, Gdańsk, Poland
| | - Natalia Bednarz-Knoll
- Laboratory of Translational Oncology, Intercollegiate Faculty of Biotechnology, Medical University of Gdańsk, Gdańsk, Poland
| |
Collapse
|
28
|
Thomas-Bonafos T, Pierga JY, Bidard FC, Cabel L, Kiavue N. Circulating tumor cells in breast cancer: clinical validity and utility. NPJ Breast Cancer 2024; 10:103. [PMID: 39613809 DOI: 10.1038/s41523-024-00706-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2024] [Accepted: 10/23/2024] [Indexed: 12/01/2024] Open
Abstract
Circulating tumor cells (CTCs) have been extensively studied in breast cancer (BC), with large studies establishing CTCs as a robust prognostic biomarker in early and metastatic breast cancer (MBC). Several phase II and phase III trials have investigated the clinical utility of CTCs in BC. Here, we outline the current landscape for the use of CTCs in the clinic at different stages of BC, focusing first on early BC, then on MBC, with a particular focus on interventional clinical trials based on CTCs.
Collapse
Affiliation(s)
- Thibault Thomas-Bonafos
- Institut Curie, Department of Medical Oncology, Paris, France
- Circulating Tumor Biomarkers laboratory, Inserm CIC 1428, Department of Translational Research, Institut Curie, Paris, France
| | - Jean Yves Pierga
- Institut Curie, Department of Medical Oncology, Paris, France
- Circulating Tumor Biomarkers laboratory, Inserm CIC 1428, Department of Translational Research, Institut Curie, Paris, France
- Université Paris Cité, Paris, France
| | - François-Clément Bidard
- Institut Curie, Department of Medical Oncology, Paris, France
- Circulating Tumor Biomarkers laboratory, Inserm CIC 1428, Department of Translational Research, Institut Curie, Paris, France
- Université de Versailles Saint-Quentin, Université Paris-Saclay, Saint-Cloud, France
| | - Luc Cabel
- Institut Curie, Department of Medical Oncology, Paris, France
- Circulating Tumor Biomarkers laboratory, Inserm CIC 1428, Department of Translational Research, Institut Curie, Paris, France
| | - Nicolas Kiavue
- Institut Curie, Department of Medical Oncology, Paris, France.
- Circulating Tumor Biomarkers laboratory, Inserm CIC 1428, Department of Translational Research, Institut Curie, Paris, France.
| |
Collapse
|
29
|
Lewis F, Beirne J, Henderson B, Norris L, Cadoo K, Kelly T, Martin C, Hurley S, Kanjuga M, O'Driscoll L, Gately K, Oner E, Saini VM, Brooks D, Selemidis S, Kamran W, Haughey N, Maguire P, O'Gorman C, Saadeh FA, Ward MP, O'Leary JJ, O'Toole SA. Unravelling the biological and clinical challenges of circulating tumour cells in epithelial ovarian carcinoma. Cancer Lett 2024; 605:217279. [PMID: 39341451 DOI: 10.1016/j.canlet.2024.217279] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2024] [Revised: 09/22/2024] [Accepted: 09/24/2024] [Indexed: 10/01/2024]
Abstract
Epithelial ovarian carcinoma (EOC) is the eighth most common cancer in women and the leading cause of gynaecological cancer death, predominantly due to the absence of effective screening tools, advanced stage at diagnosis, and high rates of recurrence. Circulating tumour cells (CTCs), a rare subset of tumour cells that disseminate from a tumour and migrate into the circulation, play a pivotal role in the metastatic cascade, and therefore hold promise as biomarkers for disease monitoring and prognostication. Exploring CTCs from liquid biopsies is an appealing approach for research and clinical practice, given it is minimally invasive, facilitates serial sampling and enables the capture of the entire spectrum of cancer cells circulating in the blood. The prognostic utility of CTC enumeration has been FDA-approved for clinical use in metastatic breast, prostate, and colorectal cancers. However, the unique biology of EOC, discussed herein, compounds the detection and characterisation complexities already inherent in CTC research, consequently hindering progress towards clinical applications. The aim of this review is to provide an overview of both the biological and clinical challenges encountered in harnessing the power of CTCs in EOC.
Collapse
Affiliation(s)
- Faye Lewis
- Department of Histopathology, School of Medicine, Trinity College Dublin, Dublin, Ireland; Department of Obstetrics and Gynaecology, School of Medicine, Trinity College Dublin, Dublin, Ireland; Trinity St. James's Cancer Institute, Trinity College Dublin, Dublin, Ireland
| | - James Beirne
- Blackrock Health Hermitage Clinic, Old Lucan Road, Dublin, Ireland
| | - Brian Henderson
- Department of Histopathology, School of Medicine, Trinity College Dublin, Dublin, Ireland; Department of Obstetrics and Gynaecology, School of Medicine, Trinity College Dublin, Dublin, Ireland; Trinity St. James's Cancer Institute, Trinity College Dublin, Dublin, Ireland
| | - Lucy Norris
- Department of Obstetrics and Gynaecology, School of Medicine, Trinity College Dublin, Dublin, Ireland; Trinity St. James's Cancer Institute, Trinity College Dublin, Dublin, Ireland
| | - Karen Cadoo
- Trinity St. James's Cancer Institute, Trinity College Dublin, Dublin, Ireland; The Haematology, Oncology and Palliative Care (HOPe) Directorate, St James's Hospital, Dublin, Ireland
| | - Tanya Kelly
- Department of Histopathology, School of Medicine, Trinity College Dublin, Dublin, Ireland; Department of Obstetrics and Gynaecology, School of Medicine, Trinity College Dublin, Dublin, Ireland; Trinity St. James's Cancer Institute, Trinity College Dublin, Dublin, Ireland
| | - Cara Martin
- Department of Histopathology, School of Medicine, Trinity College Dublin, Dublin, Ireland; Trinity St. James's Cancer Institute, Trinity College Dublin, Dublin, Ireland
| | - Sinéad Hurley
- Department of Histopathology, School of Medicine, Trinity College Dublin, Dublin, Ireland; Trinity St. James's Cancer Institute, Trinity College Dublin, Dublin, Ireland; Department of Clinical Medicine, School of Medicine, Trinity College Dublin, Ireland; Thoracic Oncology Research Group, Trinity Translational Medicine Institute, St James's Hospital, Dublin, Ireland
| | - Marika Kanjuga
- Department of Histopathology, School of Medicine, Trinity College Dublin, Dublin, Ireland; Department of Obstetrics and Gynaecology, School of Medicine, Trinity College Dublin, Dublin, Ireland; Trinity St. James's Cancer Institute, Trinity College Dublin, Dublin, Ireland
| | - Lorraine O'Driscoll
- Trinity St. James's Cancer Institute, Trinity College Dublin, Dublin, Ireland; School of Pharmacy and Pharmaceutical Sciences, Trinity College Dublin, Ireland; Trinity Biomedical Sciences Institute, Trinity College Dublin, Ireland
| | - Kathy Gately
- Trinity St. James's Cancer Institute, Trinity College Dublin, Dublin, Ireland; Department of Clinical Medicine, School of Medicine, Trinity College Dublin, Ireland; Thoracic Oncology Research Group, Trinity Translational Medicine Institute, St James's Hospital, Dublin, Ireland
| | - Ezgi Oner
- Trinity St. James's Cancer Institute, Trinity College Dublin, Dublin, Ireland; Department of Clinical Medicine, School of Medicine, Trinity College Dublin, Ireland; Thoracic Oncology Research Group, Trinity Translational Medicine Institute, St James's Hospital, Dublin, Ireland
| | - Volga M Saini
- Trinity St. James's Cancer Institute, Trinity College Dublin, Dublin, Ireland; Department of Clinical Medicine, School of Medicine, Trinity College Dublin, Ireland; Thoracic Oncology Research Group, Trinity Translational Medicine Institute, St James's Hospital, Dublin, Ireland
| | - Doug Brooks
- Cancer Research Institute, University of South Australia, 5001, Adelaide, Australia
| | - Stavros Selemidis
- School of Health and Biomedical Sciences, RMIT University, Victoria, 3083, Bundoora, Australia
| | - Waseem Kamran
- Department of Obstetrics and Gynaecology, School of Medicine, Trinity College Dublin, Dublin, Ireland; Trinity St. James's Cancer Institute, Trinity College Dublin, Dublin, Ireland; Division of Gynaecological Oncology, St James's Hospital, Dublin, Ireland
| | - Niamh Haughey
- Department of Obstetrics and Gynaecology, School of Medicine, Trinity College Dublin, Dublin, Ireland; Trinity St. James's Cancer Institute, Trinity College Dublin, Dublin, Ireland; Division of Gynaecological Oncology, St James's Hospital, Dublin, Ireland
| | - Patrick Maguire
- Department of Obstetrics and Gynaecology, School of Medicine, Trinity College Dublin, Dublin, Ireland; Trinity St. James's Cancer Institute, Trinity College Dublin, Dublin, Ireland; Division of Gynaecological Oncology, St James's Hospital, Dublin, Ireland
| | - Catherine O'Gorman
- Department of Obstetrics and Gynaecology, School of Medicine, Trinity College Dublin, Dublin, Ireland; Trinity St. James's Cancer Institute, Trinity College Dublin, Dublin, Ireland; Division of Gynaecological Oncology, St James's Hospital, Dublin, Ireland
| | - Feras Abu Saadeh
- Department of Obstetrics and Gynaecology, School of Medicine, Trinity College Dublin, Dublin, Ireland; Trinity St. James's Cancer Institute, Trinity College Dublin, Dublin, Ireland; Division of Gynaecological Oncology, St James's Hospital, Dublin, Ireland
| | - Mark P Ward
- Department of Histopathology, School of Medicine, Trinity College Dublin, Dublin, Ireland; Department of Obstetrics and Gynaecology, School of Medicine, Trinity College Dublin, Dublin, Ireland; Trinity St. James's Cancer Institute, Trinity College Dublin, Dublin, Ireland.
| | - John J O'Leary
- Department of Histopathology, School of Medicine, Trinity College Dublin, Dublin, Ireland; Trinity St. James's Cancer Institute, Trinity College Dublin, Dublin, Ireland.
| | - Sharon A O'Toole
- Department of Histopathology, School of Medicine, Trinity College Dublin, Dublin, Ireland; Department of Obstetrics and Gynaecology, School of Medicine, Trinity College Dublin, Dublin, Ireland; Trinity St. James's Cancer Institute, Trinity College Dublin, Dublin, Ireland.
| |
Collapse
|
30
|
Ebrahimi A, Ak G, Özel C, İzgördü H, Ghorbanpoor H, Hassan S, Avci H, Metintaş M. Clinical Perspectives and Novel Preclinical Models of Malignant Pleural Mesothelioma: A Critical Review. ACS Pharmacol Transl Sci 2024; 7:3299-3333. [PMID: 39539262 PMCID: PMC11555512 DOI: 10.1021/acsptsci.4c00324] [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: 05/31/2024] [Revised: 09/24/2024] [Accepted: 09/30/2024] [Indexed: 11/16/2024]
Abstract
Pleural mesothelioma (PM), a rare malignant tumor explicitly associated with asbestos and erionite exposures, has become a global health problem due to limited treatment options and a poor prognosis, in which the median life expectancy varies depending on the method of treatment. However, the importance of early diagnosis is emphasized, and the practical methods have not matured yet. This study provides a critical overview of PM, addressing various aspects like epidemiology, etiology, diagnosis, treatment options, and the potential use of advanced technologies like microfluidic chip-based models for research and diagnosis. It initially begins with fundamentals of clinical aspects and then discusses the identification of disease-specific biomarkers in patients' serum or plasma samples, which could potentially be used for early diagnosis. A detailed investigation of the sophisticated preclinical models is highlighted. Recent three-dimensional (3D) model accomplishments, including microarchitecture modeling by transwell coculture, spheroids, organoids, 3D bioprinting constructs, and ex vivo tumor slices, are discussed comprehensively. On-chip models that imitate physiological processes, such as detection chips and therapeutic screening chips, are assessed as potential techniques. The review concludes with a critical and constructive discussion of the growing interest in the topic and its limitations and suggestions.
Collapse
Affiliation(s)
- Aliakbar Ebrahimi
- Cellular
Therapy and Stem Cell Production Application and Research Center (ESTEM), Eskişehir Osmangazi University, Eskişehir 26040, Turkey
| | - Güntülü Ak
- Eskisehir
Osmangazi University, Faculty of Medicine, Department of Pulmonary
Diseases, Lung and Pleural Cancers Research
and Clinical Center, Eskisehir 26040, Turkey
| | - Ceren Özel
- Cellular
Therapy and Stem Cell Production Application and Research Center (ESTEM), Eskişehir Osmangazi University, Eskişehir 26040, Turkey
- Department
of Stem Cell, Institute of Health Sciences, Eskişehir Osmangazi University, Eskişehir 26040, Turkey
| | - Hüseyin İzgördü
- Eskisehir
Osmangazi University, Faculty of Medicine, Department of Pulmonary
Diseases, Lung and Pleural Cancers Research
and Clinical Center, Eskisehir 26040, Turkey
| | - Hamed Ghorbanpoor
- Cellular
Therapy and Stem Cell Production Application and Research Center (ESTEM), Eskişehir Osmangazi University, Eskişehir 26040, Turkey
- Department
of Biomedical Engineering, Eskişehir
Osmangazi University, Eskişehir 26040, Turkey
| | - Shabir Hassan
- Department
of Biological Sciences, Khalifa University
of Science and Technology, Abu Dhabi 127788, United Arab Emirates
| | - Huseyin Avci
- Cellular
Therapy and Stem Cell Production Application and Research Center (ESTEM), Eskişehir Osmangazi University, Eskişehir 26040, Turkey
- Department
of Stem Cell, Institute of Health Sciences, Eskişehir Osmangazi University, Eskişehir 26040, Turkey
- Department
of Metallurgical and Materials Engineering, Eskişehir Osmangazi University, Eskişehir 26040, Turkey
- Translational
Medicine Research and Clinical Center (TATUM), Eskişehir Osmangazi University, Eskişehir 26040, Turkey
| | - Muzaffer Metintaş
- Eskisehir
Osmangazi University, Faculty of Medicine, Department of Pulmonary
Diseases, Lung and Pleural Cancers Research
and Clinical Center, Eskisehir 26040, Turkey
- Translational
Medicine Research and Clinical Center (TATUM), Eskişehir Osmangazi University, Eskişehir 26040, Turkey
| |
Collapse
|
31
|
Haghighi AH, Ghaderian A, Mirzaei E. Isolation of B Cells Using Silane-Coated Magnetic Nanoparticles. Int J Biomater 2024; 2024:8286525. [PMID: 39512856 PMCID: PMC11540882 DOI: 10.1155/2024/8286525] [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: 06/05/2024] [Accepted: 09/26/2024] [Indexed: 11/15/2024] Open
Abstract
One of the most important advantages and applications of coated nanoparticles in biological applications is their use in isolating different types of cells to diagnose and treat all types of diseases. Therefore, in this research work, the possibility of isolation and enrichment of B cells using magnetic iron oxide nanoparticles have been investigated. In this regard, magnetic nanoparticles are first coated with (3-aminopropyl)triethoxysilane to make them hydrophilic and prevent their clumping, then reacted with and rendered biocompatible by FITC anti-human CD20 antibody. These nanoparticles containing antibodies have been used to isolate B cells from the lymphatic cells. Transmission electron microscopy (TEM) and vibrating-sample magnetometry (VSM) tests were used to check the magnetic properties and coating of nanoparticles. The flow cytometry and fluorescent microscopy tests are used to check antibody binding to nanoparticles. Moreover, flow cytometry tests were used to check the extent of cell separation. Results show that nanoparticles reacted with 450 μL of antibody (T450) performed better than other nanoparticles in isolating B cells.
Collapse
Affiliation(s)
- Amir Hossein Haghighi
- Department of Polymer Engineering, Islamic Azad University, Shiraz Branch, Shiraz, Iran
| | - Abolfazl Ghaderian
- Young Researchers and Elite Club, Islamic Azad University, Shiraz Branch, Shiraz, Iran
| | - Esmaeil Mirzaei
- Department of Medical Nanotechnology, School of Advanced Medical Sciences and Technologies, Shiraz University of Medical Sciences, Shiraz, Iran
| |
Collapse
|
32
|
Lișcu HD, Verga N, Atasiei DI, Badiu DC, Dumitru AV, Ultimescu F, Pavel C, Stefan RE, Manole DC, Ionescu AI. Biomarkers in Colorectal Cancer: Actual and Future Perspectives. Int J Mol Sci 2024; 25:11535. [PMID: 39519088 PMCID: PMC11546354 DOI: 10.3390/ijms252111535] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2024] [Revised: 10/23/2024] [Accepted: 10/24/2024] [Indexed: 11/16/2024] Open
Abstract
Biomarkers in colorectal cancer (CRC) are of great interest in the current literature due to improvements in techniques such as liquid biopsy and next-generation sequencing (NGS). However, screening methods vary globally, with multi-target stool DNA (mt-sDNA) predominantly used in the USA and, more recently, the Cologuard Plus; biomarkers such as the Galectins family and septins show promise in early detection. Gut microbiome assessments, such as Fusobacterium nucleatum, are under intense exploration. Diagnostic tests, such as circulating DNA analysis via NGS, exhibit effectiveness and are being increasingly adopted. Circulating tumor cells emerge as potential alternatives to traditional methods in terms of diagnosis and prognosis. Predictive biomarkers are well established in guidelines; nonetheless, with the aid of machine learning and artificial intelligence, these biomarkers may be improved. This review critically explores the actual dynamic landscape of CRC biomarkers and future, promising biomarkers involved in screening, diagnosis, and prognosis.
Collapse
Affiliation(s)
- Horia-Dan Lișcu
- Discipline of Oncological Radiotherapy and Medical Imaging, Carol Davila University of Medicine and Pharmacy, 050474 Bucharest, Romania; (H.-D.L.); (A.-I.I.)
- Radiotherapy Department, Colțea Clinical Hospital, 030167 Bucharest, Romania;
| | - Nicolae Verga
- Radiotherapy Department, Colțea Clinical Hospital, 030167 Bucharest, Romania;
| | - Dimitrie-Ionuț Atasiei
- Discipline of Oncological Radiotherapy and Medical Imaging, Carol Davila University of Medicine and Pharmacy, 050474 Bucharest, Romania; (H.-D.L.); (A.-I.I.)
| | - Dumitru-Cristinel Badiu
- Department of Surgery, Bagdasar Arseni Clinical Emergency Hospital, Carol Davila University of Medicine and Pharmacy, 8 Eroii Sanitari Bd., 050474 Bucharest, Romania;
| | - Adrian Vasile Dumitru
- Department of Pathology, University Emergency Hospital Bucharest, Carol Davila University of Medicine and Pharmacy, 8 Eroii Sanitari Bd., 050474 Bucharest, Romania;
| | - Flavia Ultimescu
- Department of Pathology, Institute of Oncology Alexandru Trestioreanu, Carol Davila University of Medicine and Pharmacy, 050474 Bucharest, Romania;
| | - Christopher Pavel
- Department of Gastroenterology, Floreasca Emergency Hospital Bucharest, Carol Davila University of Medicine and Pharmacy, 050474 Bucharest, Romania;
| | - Roxana-Elena Stefan
- General Surgery Department, Clinic of General and Esophageal Surgery, Sf. Maria Clinical Hospital, Carol Davila University of Medicine and Pharmacy, 050474 Bucharest, Romania;
| | - Diandra-Carmen Manole
- Department of Endocrinology, Faculty of General Medicine, Carol Davila University of Medicine and Pharmacy, 8 Eroii Sanitari Bd., 050474 Bucharest, Romania;
| | - Andreea-Iuliana Ionescu
- Discipline of Oncological Radiotherapy and Medical Imaging, Carol Davila University of Medicine and Pharmacy, 050474 Bucharest, Romania; (H.-D.L.); (A.-I.I.)
- Radiotherapy Department, Colțea Clinical Hospital, 030167 Bucharest, Romania;
| |
Collapse
|
33
|
Leitão TP, Corredeira P, Rodrigues C, Piairo P, Miranda M, Cavaco A, Kucharczak S, Antunes M, Peixoto S, dos Reis JP, Lopes T, Diéguez L, Costa L. A Randomized Controlled Trial Assessing the Release of Circulating Tumor and Mesenchymal Cells in No-Touch Radical Nephrectomy. Cancers (Basel) 2024; 16:3601. [PMID: 39518041 PMCID: PMC11545310 DOI: 10.3390/cancers16213601] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2024] [Revised: 10/17/2024] [Accepted: 10/23/2024] [Indexed: 11/16/2024] Open
Abstract
BACKGROUND Circulating tumor cells (CTCs) may be the missing renal cell carcinoma (RCC) biomarker. No-touch (NT) resection has shown benefit in several tumors. METHODS A randomized controlled trial comparing CTC and circulating mesenchymal cell (CMC) release in no-touch (NT) vs. conventional (C) laparoscopic RN. Blood samples were collected at operation room arrival (S0), specimen extraction (S1), postoperative D1, and D30. CTCs were isolated and analyzed using RUBYchip™. RESULTS Thirty-four patients were included. No significant differences were found between groups in CTC and CMC counts, count variations between time points, complications, and survival. The total circulating cell detection rates in the NT, C, and overall RCC groups were 58.3%, 80.0%, and 70.4% at S0; 41.6%, 86.7%, and 66.7% at S1; 50.0%, 64.3%, and 60.0% at D1; and 54.5%, 42.9%, and 44.0% at D30, respectively. A progressive decrease in CMCs was observed in the C group after surgery, especially at D1 (4.78 to 1.64 CMCs/7.5 mL blood, p = 0.035). Healthy controls had no circulating cells; however, high CMC counts were found in chronic inflammation controls and oncocytoma patients, with no significant difference from RCC patients (p = 0.460). CONCLUSIONS NT RN did not reduce circulating cell release nor improve survival compared to C RN.
Collapse
Affiliation(s)
- Tito Palmela Leitão
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, 1649-028 Lisboa, Portugal; (P.C.); (C.R.); (A.C.); (S.K.); (L.C.)
- Faculdade de Medicina, Universidade de Lisboa, 1649-028 Lisboa, Portugal
- Urology Department, Hospital de Santa Maria, Centro Hospitalar Universitário Lisboa Norte, ULS Santa Maria, 1649-028 Lisboa, Portugal;
| | - Patrícia Corredeira
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, 1649-028 Lisboa, Portugal; (P.C.); (C.R.); (A.C.); (S.K.); (L.C.)
| | - Carolina Rodrigues
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, 1649-028 Lisboa, Portugal; (P.C.); (C.R.); (A.C.); (S.K.); (L.C.)
- International Iberian Nanotechnology Laboratory, 4715-330 Braga, Portugal; (P.P.); (L.D.)
| | - Paulina Piairo
- International Iberian Nanotechnology Laboratory, 4715-330 Braga, Portugal; (P.P.); (L.D.)
- RUBYnanomed Lda, 4700-314 Braga, Portugal
| | - Miguel Miranda
- Urology Department, Hospital de Santa Maria, Centro Hospitalar Universitário Lisboa Norte, ULS Santa Maria, 1649-028 Lisboa, Portugal;
| | - Ana Cavaco
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, 1649-028 Lisboa, Portugal; (P.C.); (C.R.); (A.C.); (S.K.); (L.C.)
| | - Sandra Kucharczak
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, 1649-028 Lisboa, Portugal; (P.C.); (C.R.); (A.C.); (S.K.); (L.C.)
- Department of Clinical and Molecular Medicine, Faculty of Medicine and Health Sciences, Norwegian University of Science and Technology, P.O. Box 8905, 7491 Trondheim, Norway
| | - Marília Antunes
- CEAUL—Centro de Estatística e Aplicações, Faculdade de Ciências, Universidade de Lisboa, 1749-028 Lisboa, Portugal;
| | - Sara Peixoto
- Radiology Department, Hospital de Santa Maria, Centro Hospitalar Universitário Lisboa Norte, ULS Santa Maria, 1649-028 Lisboa, Portugal;
| | - José Palma dos Reis
- Faculdade de Medicina, Universidade de Lisboa, 1649-028 Lisboa, Portugal
- Urology Department, Hospital de Santa Maria, Centro Hospitalar Universitário Lisboa Norte, ULS Santa Maria, 1649-028 Lisboa, Portugal;
| | - Tomé Lopes
- Faculdade de Medicina, Universidade de Lisboa, 1649-028 Lisboa, Portugal
| | - Lorena Diéguez
- International Iberian Nanotechnology Laboratory, 4715-330 Braga, Portugal; (P.P.); (L.D.)
- RUBYnanomed Lda, 4700-314 Braga, Portugal
| | - Luís Costa
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, 1649-028 Lisboa, Portugal; (P.C.); (C.R.); (A.C.); (S.K.); (L.C.)
- Faculdade de Medicina, Universidade de Lisboa, 1649-028 Lisboa, Portugal
- Oncology Department, Hospital de Santa Maria, Centro Hospitalar Universitário Lisboa Norte, ULS Santa Maria, 1649-028 Lisboa, Portugal
| |
Collapse
|
34
|
Maqueda JJ, De Feo A, Scotlandi K. Evaluating Circulating Biomarkers for Diagnosis, Prognosis, and Tumor Monitoring in Pediatric Sarcomas: Recent Advances and Future Directions. Biomolecules 2024; 14:1306. [PMID: 39456239 PMCID: PMC11506719 DOI: 10.3390/biom14101306] [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: 08/30/2024] [Revised: 10/10/2024] [Accepted: 10/15/2024] [Indexed: 10/28/2024] Open
Abstract
Pediatric sarcomas present a significant challenge in oncology. There is an urgent need for improved therapeutic strategies for high-risk patients and better management of long-term side effects for those who survive the disease. Liquid biopsy is emerging as a promising tool to optimize treatment in these patients by offering non-invasive, repeatable assessments of disease status. Circulating biomarkers can provide valuable insights into tumor genetics and treatment response, potentially facilitating early diagnosis and dynamic disease monitoring. This review examines the potential of liquid biopsies, focusing on circulating biomarkers in the most common pediatric sarcomas, i.e., osteosarcoma, Ewing sarcoma, and rhabdomyosarcoma. We also highlight the current research efforts and the necessary advancements required before these technologies can be widely adopted in clinical practice.
Collapse
Affiliation(s)
- Joaquín J. Maqueda
- Laboratory of Experimental Oncology, IRCCS Istituto Ortopedico Rizzoli, 40136 Bologna, Italy; (A.D.F.); (K.S.)
| | | | | |
Collapse
|
35
|
Witek MA, Larkey NE, Bartakova A, Hupert ML, Mog S, Cronin JK, Vun J, August KJ, Soper SA. Microfluidic Affinity Selection of B-Lineage Cells from Peripheral Blood for Minimal Residual Disease Monitoring in Pediatric B-Type Acute Lymphoblastic Leukemia Patients. Int J Mol Sci 2024; 25:10619. [PMID: 39408948 PMCID: PMC11477226 DOI: 10.3390/ijms251910619] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2024] [Revised: 09/28/2024] [Accepted: 09/29/2024] [Indexed: 10/20/2024] Open
Abstract
Assessment of minimal residual disease (MRD) is the most powerful predictor of outcome in B-type acute lymphoblastic leukemia (B-ALL). MRD, defined as the presence of leukemic cells in the blood or bone marrow, is used for the evaluation of therapy efficacy. We report on a microfluidic-based MRD (MF-MRD) assay that allows for frequent evaluation of blood for the presence of circulating leukemia cells (CLCs). The microfluidic chip affinity selects B-lineage cells, including CLCs using anti-CD19 antibodies poised on the wall of the microfluidic chip. Affinity-selected cells are released from the capture surface and can be subjected to immunophenotyping to enumerate the CLCs, perform fluorescence in situ hybridization (FISH), and/or molecular analysis of the CLCs' mRNA/gDNA. During longitudinal testing of 20 patients throughout induction and consolidation therapy, the MF-MRD performed 116 tests, while only 41 were completed with multiparameter flow cytometry (MFC-MRD) using a bone marrow aspirate, as standard-of-care. Overall, 57% MF-MRD tests were MRD(+) as defined by CLC numbers exceeding a threshold of 5 × 10-4%, which was determined to be the limit of quantitation. Above a threshold of 0.01%, MFC-MRD was positive in 34% of patients. The MF offered the advantage of the opportunity for efficiently processing small volumes of blood (2 mL), which is important in the care of pediatric patients, especially infants. The minimally invasive means of blood collection are of high value when treating patients whose MRD is typically tested using an invasive bone marrow biopsy. MF-MRD detection can be useful for stratification of patients into risk groups and monitoring of patient well-being after completion of treatment for early recognition of potential impending disease recurrence.
Collapse
Affiliation(s)
- Malgorzata A. Witek
- Department of Chemistry, The University of Kansas, Lawrence, KS 66047, USA;
- Center of BioModular Multiscale Systems for Precision Medicine, Lawrence, KS 66045, USA; (N.E.L.); (S.M.)
| | - Nicholas E. Larkey
- Center of BioModular Multiscale Systems for Precision Medicine, Lawrence, KS 66045, USA; (N.E.L.); (S.M.)
- Department of Cancer Biology, The University of Kansas Medical Center, Kansas City, KS 66160, USA
| | - Alena Bartakova
- Biofluidica Inc., San Diego, CA 92121, USA; (A.B.); (M.L.H.)
| | | | - Shalee Mog
- Center of BioModular Multiscale Systems for Precision Medicine, Lawrence, KS 66045, USA; (N.E.L.); (S.M.)
| | - Jami K. Cronin
- Division of Hematology/Oncology/Bone Marrow Transplant, Children’s Mercy Kansas City, Kansas City, MO 64108, USA; (J.K.C.); (J.V.)
| | - Judy Vun
- Division of Hematology/Oncology/Bone Marrow Transplant, Children’s Mercy Kansas City, Kansas City, MO 64108, USA; (J.K.C.); (J.V.)
| | - Keith J. August
- Division of Hematology/Oncology/Bone Marrow Transplant, Children’s Mercy Kansas City, Kansas City, MO 64108, USA; (J.K.C.); (J.V.)
| | - Steven A. Soper
- Department of Chemistry, The University of Kansas, Lawrence, KS 66047, USA;
- Center of BioModular Multiscale Systems for Precision Medicine, Lawrence, KS 66045, USA; (N.E.L.); (S.M.)
- Department of Cancer Biology, The University of Kansas Medical Center, Kansas City, KS 66160, USA
- Biofluidica Inc., San Diego, CA 92121, USA; (A.B.); (M.L.H.)
- Bioengineering Program, The University of Kansas, Lawrence, KS 66045, USA
- Department of Mechanical Engineering, The University of Kansas, Lawrence, KS 66045, USA
| |
Collapse
|
36
|
Liu W, Wang Y, Jiang P, Huang K, Zhang H, Chen J, Chen P. DNAzyme and controllable cholesterol stacking DNA machine integrates dual-target recognition CTCs enable homogeneous liquid biopsy of breast cancer. Biosens Bioelectron 2024; 261:116493. [PMID: 38901393 DOI: 10.1016/j.bios.2024.116493] [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: 02/26/2024] [Revised: 05/22/2024] [Accepted: 06/09/2024] [Indexed: 06/22/2024]
Abstract
Although circulating tumor cells (CTCs) have demonstrated considerable importance in liquid biopsy, their detection is limited by low concentrations and complex sample components. Herein, we developed a homogeneous, simple, and high-sensitivity strategy targeting breast cancer cells. This method was based on a non-immunological stepwise centrifugation preprocessing approach to isolate CTCs from whole blood. Precise quantification is achieved through the specific binding of aptamers to the overexpressed mucin 1 (MUC1) and human epidermal growth factor receptor 2 (HER2) proteins of breast cancer cells. Subsequently, DNAzyme cleavage and parallel catalytic hairpin assembly (CHA) reactions on the cholesterol-stacking DNA machine were initiated, which opened the hairpin structures T-Hg2+-T and C-Ag+-C, enabling multiple amplifications. This leads to the fluorescence signal reduction from Hg2+-specific carbon dots (CDs) and CdTe quantum dots (QDs) by released ions. This strategy demonstrated a detection performance with a limit of detection (LOD) of 3 cells/mL and a linear range of 5-100 cells/mL. 42 clinical samples have been validated, confirming their consistency with clinical imaging, pathology findings and the folate receptor (FR)-PCR kit results, exhibiting desirable specificity of 100% and sensitivity of 80.6%. These results highlight the promising applicability of our method for diagnosing and monitoring breast cancer.
Collapse
Affiliation(s)
- Weijing Liu
- Department of Laboratory Medicine, Med+X Center for Manufacturing, Department of General Surgery, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, China; Breast Center, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Yue Wang
- Department of Laboratory Medicine, Med+X Center for Manufacturing, Department of General Surgery, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Pengjun Jiang
- Department of Laboratory Medicine, Med+X Center for Manufacturing, Department of General Surgery, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Ke Huang
- College of Chemistry and Material Science, Sichuan Normal University, Chengdu, Sichuan, 610068, China
| | - He Zhang
- Department of Laboratory Medicine, Med+X Center for Manufacturing, Department of General Surgery, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Jie Chen
- Department of Laboratory Medicine, Med+X Center for Manufacturing, Department of General Surgery, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, China; Breast Center, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, China.
| | - Piaopiao Chen
- Department of Laboratory Medicine, Med+X Center for Manufacturing, Department of General Surgery, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, China.
| |
Collapse
|
37
|
Szewczyk K, Jiang L, Khawaja H, Miranti CK, Zohar Y. Microfluidic Applications in Prostate Cancer Research. MICROMACHINES 2024; 15:1195. [PMID: 39459070 PMCID: PMC11509716 DOI: 10.3390/mi15101195] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/19/2024] [Revised: 09/13/2024] [Accepted: 09/23/2024] [Indexed: 10/28/2024]
Abstract
Prostate cancer is a disease in which cells in the prostate, a gland in the male reproductive system below the bladder, grow out of control and, among men, it is the second-most frequently diagnosed cancer (other than skin cancer). In recent years, prostate cancer death rate has stabilized and, currently, it is the second-most frequent cause of cancer death in men (after lung cancer). Most deaths occur due to metastasis, as cancer cells from the original tumor establish secondary tumors in distant organs. For a long time, classical cell cultures and animal models have been utilized in basic and applied scientific research, including clinical applications for many diseases, such as prostate cancer, since no better alternatives were available. Although helpful in dissecting cellular mechanisms, these models are poor predictors of physiological behavior mainly because of the lack of appropriate microenvironments. Microfluidics has emerged in the last two decades as a technology that could lead to a paradigm shift in life sciences and, in particular, controlling cancer. Microfluidic systems, such as organ-on-chips, have been assembled to mimic the critical functions of human organs. These microphysiological systems enable the long-term maintenance of cellular co-cultures in vitro to reconstitute in vivo tissue-level microenvironments, bridging the gap between traditional cell cultures and animal models. Several reviews on microfluidics for prostate cancer studies have been published focusing on technology advancement and disease progression. As metastatic castration-resistant prostate cancer remains a clinically challenging late-stage cancer, with no curative treatments, we expanded this review to cover recent microfluidic applications related to prostate cancer research. The review includes discussions of the roles of microfluidics in modeling the human prostate, prostate cancer initiation and development, as well as prostate cancer detection and therapy, highlighting potentially major contributions of microfluidics in the continuous march toward eradicating prostate cancer.
Collapse
Affiliation(s)
- Kailie Szewczyk
- Department of Aerospace and Mechanical Engineering, University of Arizona, Tucson, AZ 85721, USA; (K.S.); (L.J.)
| | - Linan Jiang
- Department of Aerospace and Mechanical Engineering, University of Arizona, Tucson, AZ 85721, USA; (K.S.); (L.J.)
| | - Hunain Khawaja
- Cancer Biology Graduate Interdisciplinary Program, University of Arizona, Tucson, AZ 85724, USA;
| | - Cindy K. Miranti
- Department of Molecular and Cellular Biology, University of Arizona, Tucson, AZ 85721, USA;
- University of Arizona Cancer Center, University of Arizona, Tucson, AZ 85724, USA
| | - Yitshak Zohar
- Department of Aerospace and Mechanical Engineering, University of Arizona, Tucson, AZ 85721, USA; (K.S.); (L.J.)
- University of Arizona Cancer Center, University of Arizona, Tucson, AZ 85724, USA
| |
Collapse
|
38
|
Xie Z, Wang Y, Chen T, Fan W, Wei L, Liu B, Situ X, Zhan Q, Fu T, Tian T, Li S, He Q, Zhou J, Wang H, Du J, Tseng HR, Lei Y, Tang KJ, Ke Z. Circulating tumor cells with increasing aneuploidy predict inferior prognosis and therapeutic resistance in small cell lung cancer. Drug Resist Updat 2024; 76:101117. [PMID: 38996549 DOI: 10.1016/j.drup.2024.101117] [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: 04/26/2024] [Revised: 06/23/2024] [Accepted: 06/28/2024] [Indexed: 07/14/2024]
Abstract
AIMS Treatment resistance commonly emerges in small cell lung cancer (SCLC), necessitating the development of novel and effective biomarkers to dynamically assess therapeutic efficacy. This study aims to evaluate the clinical utility of aneuploid circulating tumor cells (CTCs) for risk stratification and treatment response monitoring. METHODS A total of 126 SCLC patients (two cohorts) from two independent cancer centers were recruited as the study subjects. Blood samples were collected from these patients and aneuploid CTCs were detected. Aneuploid CTC count (ACC) and aneuploid CTC score (ACS), were used to predict progression-free survival (PFS) and overall survival (OS). The performance of the ACC and the ACS was evaluated by calculating the area under the receiver operating characteristic (ROC) curve (AUC). RESULTS Compared to ACC, ACS exhibited superior predictive power for PFS and OS in these 126 patients. Moreover, both univariate and multivariate analyses revealed that ACS was an independent prognostic factor. Dynamic ACS changes reflected treatment response, which is more precise than ACC changes. ACS can be used to assess chemotherapy resistance and is more sensitive than radiological examination (with a median lead time of 2.8 months; P < 0.001). When patients had high ACS levels (> 1.115) at baseline, the combination of immunotherapy and chemotherapy resulted in longer PFS (median PFS, 7.7 months; P = 0.007) and OS (median OS, 16.3 months; P = 0.033) than chemotherapy alone (median PFS, 4.9 months; median OS, 13.6 months). CONCLUSIONS ACS could be used as a biomarker for risk stratification, treatment response monitoring, and individualized therapeutic intervention in SCLC patients.
Collapse
Affiliation(s)
- Zhongpeng Xie
- Department of Pathology, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou 510080, China; Molecular Diagnosis and Gene Test Centre, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou 510080, China; Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou 510080, China
| | - Yanxia Wang
- Department of Pathology, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou 510080, China; Molecular Diagnosis and Gene Test Centre, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou 510080, China; Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou 510080, China
| | - Tingfei Chen
- Department of Thoracic Surgery, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou 510080, China
| | - Wei Fan
- Cyttel Biomedical Technology Co., Ltd, Taizhou 225300, China
| | - Lihong Wei
- Department of Pathology, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou 510080, China; Molecular Diagnosis and Gene Test Centre, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou 510080, China
| | - Bixia Liu
- Molecular Diagnosis and Gene Test Centre, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou 510080, China
| | - Xiaohua Situ
- Department of Pathology, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou 510080, China; Molecular Diagnosis and Gene Test Centre, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou 510080, China
| | - Qinru Zhan
- Department of Pathology, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou 510080, China; Molecular Diagnosis and Gene Test Centre, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou 510080, China
| | - Tongze Fu
- Department of Pathology, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou 510080, China; Molecular Diagnosis and Gene Test Centre, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou 510080, China
| | - Tian Tian
- Department of Pathology, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou 510080, China
| | - Shuhua Li
- Department of Pathology, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou 510080, China; Molecular Diagnosis and Gene Test Centre, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou 510080, China; Precision Medicine Institute, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou 510080, China
| | - Qiong He
- Department of Pathology, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou 510080, China; Molecular Diagnosis and Gene Test Centre, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou 510080, China; Precision Medicine Institute, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou 510080, China
| | - Jianwen Zhou
- Department of Pathology, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou 510080, China; Molecular Diagnosis and Gene Test Centre, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou 510080, China; Precision Medicine Institute, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou 510080, China
| | - Huipin Wang
- Molecular Diagnostic Center, Zhongshan City People's Hospital, Zhongshan 528403, China
| | - Juan Du
- Molecular Diagnostic Center, Zhongshan City People's Hospital, Zhongshan 528403, China
| | - Hsian-Rong Tseng
- California NanoSystems Institute, Crump Institute for Molecular Imaging, Department of Molecular and Medical Pharmacology, University of California, Los Angeles, CA, USA.
| | - Yiyan Lei
- Department of Thoracic Surgery, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou 510080, China.
| | - Ke-Jing Tang
- Division of Pulmonary and Critical Care Medicine, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou 510080, China; Department of Pharmacy, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510080, China.
| | - Zunfu Ke
- Department of Pathology, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou 510080, China; Molecular Diagnosis and Gene Test Centre, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou 510080, China; Precision Medicine Institute, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou 510080, China.
| |
Collapse
|
39
|
Patel RK, Parappilly MS, Walker BS, Heussner RT, Fung A, Chang YH, Kardosh A, Lopez CD, Mayo SC, Wong MH. Exploratory Analyses of Circulating Neoplastic-Immune Hybrid Cells as Prognostic Biomarkers in Advanced Intrahepatic Cholangiocarcinoma. Int J Mol Sci 2024; 25:9198. [PMID: 39273147 PMCID: PMC11395231 DOI: 10.3390/ijms25179198] [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: 07/29/2024] [Revised: 08/16/2024] [Accepted: 08/19/2024] [Indexed: 09/15/2024] Open
Abstract
Existing clinical biomarkers do not reliably predict treatment response or disease progression in patients with advanced intrahepatic cholangiocarcinoma (ICC). Circulating neoplastic-immune hybrid cells (CHCs) have great promise as a blood-based biomarker for patients with advanced ICC. Peripheral blood specimens were longitudinally collected from patients with advanced ICC enrolled in the HELIX-1 phase II clinical trial (NCT04251715). CHCs were identified by co-expression of pan-cytokeratin (CK) and CD45, and levels were correlated to patient clinical disease course. Unsupervised machine learning was then performed to extract their morphological features to compare them across disease courses. Five patients were included in this study, with a median of nine specimens collected per patient. A median of 13.5 CHCs per 50,000 peripheral blood mononuclear cells were identified at baseline, and levels decreased to zero following the initiation of treatment in all patients. Counts remained undetectable in three patients who demonstrated end-of-trial clinical treatment response and conversely increased in two patients with evidence of therapeutic resistance. In the post-trial surveillance period, interval counts increased prior to or at the time of clinical progression in three patients and remain undetectable in one patient with continued long-term disease stability. Using our machine learning platform, treatment-resistant CHCs exhibited upregulation of CK and downregulation of CD45 relative to treatment-responsive CHCs. CHCs represent a promising blood-based biomarker to supplement traditional radiographic and biochemical measures.
Collapse
Affiliation(s)
- Ranish K. Patel
- Department of Surgery, Division of Surgical Oncology, Oregon Health & Science University (OHSU), Portland, OR 97239, USA; (R.K.P.)
| | - Michael S. Parappilly
- Department of Cell, Developmental, and Cancer Biology, Oregon Health & Science University (OHSU), Portland, OR 97201, USA
| | - Brett S. Walker
- Department of Surgery, Division of Surgical Oncology, Oregon Health & Science University (OHSU), Portland, OR 97239, USA; (R.K.P.)
| | - Robert T. Heussner
- Department of Biomedical Engineering, Oregon Health & Science University (OHSU), Portland, OR 97201, USA
| | - Alice Fung
- Department of Diagnostic Radiology, Oregon Health & Science University (OHSU), Portland, OR 97239, USA
| | - Young Hwan Chang
- Department of Biomedical Engineering, Oregon Health & Science University (OHSU), Portland, OR 97201, USA
- Knight Cancer Institute, Oregon Health & Science University (OHSU), Portland, OR 97201, USA
| | - Adel Kardosh
- Knight Cancer Institute, Oregon Health & Science University (OHSU), Portland, OR 97201, USA
- Department of Medicine, Division of Medical Oncology, Oregon Health & Science University (OHSU), Portland, OR 97239, USA
| | - Charles D. Lopez
- Knight Cancer Institute, Oregon Health & Science University (OHSU), Portland, OR 97201, USA
- Department of Medicine, Division of Medical Oncology, Oregon Health & Science University (OHSU), Portland, OR 97239, USA
| | - Skye C. Mayo
- Department of Surgery, Division of Surgical Oncology, Oregon Health & Science University (OHSU), Portland, OR 97239, USA; (R.K.P.)
- Knight Cancer Institute, Oregon Health & Science University (OHSU), Portland, OR 97201, USA
| | - Melissa H. Wong
- Department of Cell, Developmental, and Cancer Biology, Oregon Health & Science University (OHSU), Portland, OR 97201, USA
- Knight Cancer Institute, Oregon Health & Science University (OHSU), Portland, OR 97201, USA
| |
Collapse
|
40
|
Rodriguez-Tirado C, Sosa MS. How much do we know about the metastatic process? Clin Exp Metastasis 2024; 41:275-299. [PMID: 38520475 PMCID: PMC11374507 DOI: 10.1007/s10585-023-10248-0] [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: 08/13/2023] [Accepted: 11/17/2023] [Indexed: 03/25/2024]
Abstract
Cancer cells can leave their primary sites and travel through the circulation to distant sites, where they lodge as disseminated cancer cells (DCCs), even during the early and asymptomatic stages of tumor progression. In experimental models and clinical samples, DCCs can be detected in a non-proliferative state, defined as cellular dormancy. This state can persist for extended periods until DCCs reawaken, usually in response to niche-derived reactivation signals. Therefore, their clinical detection in sites like lymph nodes and bone marrow is linked to poor survival. Current cancer therapy designs are based on the biology of the primary tumor and do not target the biology of the dormant DCC population and thus fail to eradicate the initial or subsequent waves of metastasis. In this brief review, we discuss the current methods for detecting DCCs and highlight new strategies that aim to target DCCs that constitute minimal residual disease to reduce or prevent metastasis formation. Furthermore, we present current evidence on the relevance of DCCs derived from early stages of tumor progression in metastatic disease and describe the animal models available for their study. We also discuss our current understanding of the dissemination mechanisms utilized by genetically less- and more-advanced cancer cells, which include the functional analysis of intermediate or hybrid states of epithelial-mesenchymal transition (EMT). Finally, we raise some intriguing questions regarding the clinical impact of studying the crosstalk between evolutionary waves of DCCs and the initiation of metastatic disease.
Collapse
Affiliation(s)
- Carolina Rodriguez-Tirado
- Department of Microbiology and Immunology, Albert Einstein College of Medicine/Montefiore Medical Center, Bronx, NY, 10461, USA.
- Department of Oncology, Albert Einstein College of Medicine/Montefiore Medical Center, Bronx, NY, 10461, USA.
- Montefiore Einstein Comprehensive Cancer Center, Albert Einstein College of Medicine/Montefiore Medical Center, Bronx, NY, 10461, USA.
- Cancer Dormancy and Tumor Microenvironment Institute/Montefiore Medical Center, Albert Einstein College of Medicine, Bronx, NY, 10461, USA.
- Ruth L. and David S. Gottesman Institute for Stem Cell Research and Regenerative Medicine, Albert Einstein College of Medicine/Montefiore Medical Center, Bronx, NY, 10461, USA.
| | - Maria Soledad Sosa
- Department of Microbiology and Immunology, Albert Einstein College of Medicine/Montefiore Medical Center, Bronx, NY, 10461, USA.
- Department of Oncology, Albert Einstein College of Medicine/Montefiore Medical Center, Bronx, NY, 10461, USA.
- Montefiore Einstein Comprehensive Cancer Center, Albert Einstein College of Medicine/Montefiore Medical Center, Bronx, NY, 10461, USA.
- Cancer Dormancy and Tumor Microenvironment Institute/Montefiore Medical Center, Albert Einstein College of Medicine, Bronx, NY, 10461, USA.
- Ruth L. and David S. Gottesman Institute for Stem Cell Research and Regenerative Medicine, Albert Einstein College of Medicine/Montefiore Medical Center, Bronx, NY, 10461, USA.
| |
Collapse
|
41
|
Pace J, Lee JJ, Srinivasarao M, Kallepu S, Low PS, Niedre M. In Vivo Labeling and Detection of Circulating Tumor Cells in Mice Using OTL38. Mol Imaging Biol 2024; 26:603-615. [PMID: 38594545 PMCID: PMC11281960 DOI: 10.1007/s11307-024-01914-0] [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: 12/12/2023] [Revised: 03/04/2024] [Accepted: 03/30/2024] [Indexed: 04/11/2024]
Abstract
PURPOSE We recently developed an optical instrument to non-invasively detect fluorescently labeled circulating tumor cells (CTCs) in mice called 'Diffuse in vivo Flow Cytometry' (DiFC). OTL38 is a folate receptor (FR) targeted near-infrared (NIR) contrast agent that is FDA approved for use in fluorescence guided surgery of ovarian and lung cancer. In this work, we investigated the use OTL38 for in vivo labeling and detection of FR + CTCs with DiFC. PROCEDURES We tested OTL38 labeling of FR + cancer cell lines (IGROV-1 and L1210A) as well as FR- MM.1S cells in suspensions of Human Peripheral Blood Mononuclear cells (PBMCs) in vitro. We also tested OTL38 labeling and NIR-DIFC detection of FR + L1210A cells in blood circulation in nude mice in vivo. RESULTS 62% of IGROV-1 and 83% of L1210A were labeled above non-specific background levels in suspensions of PBMCs in vitro compared to only 2% of FR- MM.1S cells. L1210A cells could be labeled with OTL38 directly in circulation in vivo and externally detected using NIR-DiFC in mice with low false positive detection rates. CONCLUSIONS This work shows the feasibility of labeling CTCs in vivo with OTL38 and detection with DiFC. Although further refinement of the DiFC instrument and signal processing algorithms and testing with other animal models is needed, this work may eventually pave the way for human use of DiFC.
Collapse
Affiliation(s)
- Joshua Pace
- Department of Bioengineering, Northeastern University, Boston, MA, 02115, USA
| | - Jane J Lee
- Department of Bioengineering, Northeastern University, Boston, MA, 02115, USA
| | | | | | - Philip S Low
- Department of Chemistry, Purdue University, West Lafayette, IN, 047906, USA
| | - Mark Niedre
- Department of Bioengineering, Northeastern University, Boston, MA, 02115, USA.
| |
Collapse
|
42
|
Seyhan AA. Circulating Liquid Biopsy Biomarkers in Glioblastoma: Advances and Challenges. Int J Mol Sci 2024; 25:7974. [PMID: 39063215 PMCID: PMC11277426 DOI: 10.3390/ijms25147974] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2024] [Revised: 07/16/2024] [Accepted: 07/18/2024] [Indexed: 07/28/2024] Open
Abstract
Gliomas, particularly glioblastoma (GBM), represent the most prevalent and aggressive tumors of the central nervous system (CNS). Despite recent treatment advancements, patient survival rates remain low. The diagnosis of GBM traditionally relies on neuroimaging methods such as magnetic resonance imaging (MRI) or computed tomography (CT) scans and postoperative confirmation via histopathological and molecular analysis. Imaging techniques struggle to differentiate between tumor progression and treatment-related changes, leading to potential misinterpretation and treatment delays. Similarly, tissue biopsies, while informative, are invasive and not suitable for monitoring ongoing treatments. These challenges have led to the emergence of liquid biopsy, particularly through blood samples, as a promising alternative for GBM diagnosis and monitoring. Presently, blood and cerebrospinal fluid (CSF) sampling offers a minimally invasive means of obtaining tumor-related information to guide therapy. The idea that blood or any biofluid tests can be used to screen many cancer types has huge potential. Tumors release various components into the bloodstream or other biofluids, including cell-free nucleic acids such as microRNAs (miRNAs), circulating tumor DNA (ctDNA), circulating tumor cells (CTCs), proteins, extracellular vesicles (EVs) or exosomes, metabolites, and other factors. These factors have been shown to cross the blood-brain barrier (BBB), presenting an opportunity for the minimally invasive monitoring of GBM as well as for the real-time assessment of distinct genetic, epigenetic, transcriptomic, proteomic, and metabolomic changes associated with brain tumors. Despite their potential, the clinical utility of liquid biopsy-based circulating biomarkers is somewhat constrained by limitations such as the absence of standardized methodologies for blood or CSF collection, analyte extraction, analysis methods, and small cohort sizes. Additionally, tissue biopsies offer more precise insights into tumor morphology and the microenvironment. Therefore, the objective of a liquid biopsy should be to complement and enhance the diagnostic accuracy and monitoring of GBM patients by providing additional information alongside traditional tissue biopsies. Moreover, utilizing a combination of diverse biomarker types may enhance clinical effectiveness compared to solely relying on one biomarker category, potentially improving diagnostic sensitivity and specificity and addressing some of the existing limitations associated with liquid biomarkers for GBM. This review presents an overview of the latest research on circulating biomarkers found in GBM blood or CSF samples, discusses their potential as diagnostic, predictive, and prognostic indicators, and discusses associated challenges and future perspectives.
Collapse
Affiliation(s)
- Attila A. Seyhan
- Laboratory of Translational Oncology and Experimental Cancer Therapeutics, Warren Alpert Medical School, Brown University, Providence, RI 02912, USA;
- Department of Pathology and Laboratory Medicine, Warren Alpert Medical School, Brown University, Providence, RI 02912, USA
- Joint Program in Cancer Biology, Lifespan Health System and Brown University, Providence, RI 02912, USA
- Legorreta Cancer Center, Brown University, Providence, RI 02912, USA
| |
Collapse
|
43
|
Cai Q, He Y, Zhou Y, Zheng J, Deng J. Nanomaterial-Based Strategies for Preventing Tumor Metastasis by Interrupting the Metastatic Biological Processes. Adv Healthc Mater 2024; 13:e2303543. [PMID: 38411537 DOI: 10.1002/adhm.202303543] [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: 10/17/2023] [Revised: 02/01/2024] [Indexed: 02/28/2024]
Abstract
Tumor metastasis is the primary cause of cancer-related deaths. The prevention of tumor metastasis has garnered notable interest and interrupting metastatic biological processes is considered a potential strategy for preventing tumor metastasis. The tumor microenvironment (TME), circulating tumor cells (CTCs), and premetastatic niche (PMN) play crucial roles in metastatic biological processes. These processes can be interrupted using nanomaterials due to their excellent physicochemical properties. However, most studies have focused on only one aspect of tumor metastasis. Here, the hypothesis that nanomaterials can be used to target metastatic biological processes and explore strategies to prevent tumor metastasis is highlighted. First, the metastatic biological processes and strategies involving nanomaterials acting on the TME, CTCs, and PMN to prevent tumor metastasis are briefly summarized. Further, the current challenges and prospects of nanomaterials in preventing tumor metastasis by interrupting metastatic biological processes are discussed. Nanomaterial-and multifunctional nanomaterial-based strategies for preventing tumor metastasis are advantageous for the long-term fight against tumor metastasis and their continued exploration will facilitate rapid progress in the prevention, diagnosis, and treatment of tumor metastasis. Novel perspectives are outlined for developing more effective strategies to prevent tumor metastasis, thereby improving the outcomes of patients with cancer.
Collapse
Affiliation(s)
- Qingjin Cai
- Department of Urology, Urologic Surgery Center, Xinqiao Hospital, Third Military Medical University (Army Medical University), Chongqing, 400037, China
| | - Yijia He
- School of Basic Medicine, Third Military Medical University (Army Medical University), Chongqing, 400038, China
| | - Yang Zhou
- Department of Urology, Urologic Surgery Center, Xinqiao Hospital, Third Military Medical University (Army Medical University), Chongqing, 400037, China
| | - Ji Zheng
- Department of Urology, Urologic Surgery Center, Xinqiao Hospital, Third Military Medical University (Army Medical University), Chongqing, 400037, China
| | - Jun Deng
- Institute of Burn Research, Southwest Hospital, State Key Lab of Trauma, Burn and Combined Injury, Chongqing Key Laboratory for Disease Proteomics, Third Military Medical University (Army Medical University), Chongqing, 400038, China
| |
Collapse
|
44
|
Tsai KY, Huang PS, Chu PY, Nguyen TNA, Hung HY, Hsieh CH, Wu MH. Current Applications and Future Directions of Circulating Tumor Cells in Colorectal Cancer Recurrence. Cancers (Basel) 2024; 16:2316. [PMID: 39001379 PMCID: PMC11240518 DOI: 10.3390/cancers16132316] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2024] [Revised: 06/19/2024] [Accepted: 06/21/2024] [Indexed: 07/16/2024] Open
Abstract
The ability to predict or detect colorectal cancer (CRC) recurrence early after surgery enables physicians to apply appropriate treatment plans and different follow-up strategies to improve patient survival. Overall, 30-50% of CRC patients experience cancer recurrence after radical surgery, but current surveillance tools have limitations in the precise and early detection of cancer recurrence. Circulating tumor cells (CTCs) are cancer cells that detach from the primary tumor and enter the bloodstream. These can provide real-time information on disease status. CTCs might become novel markers for predicting CRC recurrence and, more importantly, for making decisions about additional adjuvant chemotherapy. In this review, the clinical application of CTCs as a therapeutic marker for stage II CRC is described. It then discusses the utility of CTCs for monitoring cancer recurrence in advanced rectal cancer patients who undergo neoadjuvant chemoradiotherapy. Finally, it discusses the roles of CTC subtypes and CTCs combined with clinicopathological factors in establishing a multimarker model for predicting CRC recurrence.
Collapse
Affiliation(s)
- Kun-Yu Tsai
- Division of Colon and Rectal Surgery, New Taipei Municipal TuCheng Hospital, New Taipei City 23652, Taiwan
| | - Po-Shuan Huang
- Graduate Institute of Biomedical Engineering, Chang Gung University, Taoyuan City 33302, Taiwan
| | - Po-Yu Chu
- Graduate Institute of Biomedical Engineering, Chang Gung University, Taoyuan City 33302, Taiwan
| | - Thi Ngoc Anh Nguyen
- Graduate Institute of Biomedical Engineering, Chang Gung University, Taoyuan City 33302, Taiwan
| | - Hsin-Yuan Hung
- Division of Colon and Rectal Surgery, New Taipei Municipal TuCheng Hospital, New Taipei City 23652, Taiwan
- College of Medicine, Chang Gung University, Taoyuan City 33302, Taiwan
| | - Chia-Hsun Hsieh
- College of Medicine, Chang Gung University, Taoyuan City 33302, Taiwan
- Division of Hematology and Oncology, Department of Internal Medicine, Chang Gung Memorial Hospital at Linkou, Taoyuan City 33302, Taiwan
- Division of Hematology and Oncology, Department of Internal Medicine, New Taipei Municipal Hospital, New Taipei City 23652, Taiwan
| | - Min-Hsien Wu
- Graduate Institute of Biomedical Engineering, Chang Gung University, Taoyuan City 33302, Taiwan
- Division of Hematology and Oncology, Department of Internal Medicine, Chang Gung Memorial Hospital at Linkou, Taoyuan City 33302, Taiwan
- Division of Hematology and Oncology, Department of Internal Medicine, New Taipei Municipal Hospital, New Taipei City 23652, Taiwan
- Department of Biomedical Engineering, Chang Gung University, Taoyuan City 33302, Taiwan
| |
Collapse
|
45
|
Chen K, Mao M, Huo L, Wang G, Pu Z, Zhang Y. Flexible DNA Nanoclaws Offer Multivalent and Powerful Spatial Pattern-Recognition for Tumor Cells. ACS APPLIED MATERIALS & INTERFACES 2024; 16:29760-29769. [PMID: 38813974 DOI: 10.1021/acsami.4c03382] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/31/2024]
Abstract
Multivalent receptor-ligand interactions (RLIs) exhibit excellent affinity for binding when targeting cell membrane receptors with low expression. However, existing strategies only allow for limited control of the valency and spacing of ligands for a certain receptor, lacking recognition patterns for multiple interested receptors with complex spatial distributions. Here, we developed flexible DNA nanoclaws with multivalent aptamers to achieve powerful cell recognition by controlling the spacing of aptamers to match the spatial patterns of receptors. The DNA nanoclaw with spacing-controllable binding sites was constructed via hybrid chain reaction (HCR), enabling dual targeting of HER2 and EpCAM molecules. The results demonstrate that the binding affinity of multivalent DNA nanoclaws to tumor cells is enhanced. We speculate that the flexible structure may conform better to irregularly shaped membrane surfaces, increasing the probability of intermolecular contact. The capture efficiency of circulating tumor cells successfully verified the high affinity and selectivity of this spatial pattern. This strategy will further promote the potential application of DNA frameworks in future disease diagnosis and treatment.
Collapse
Affiliation(s)
- Kang Chen
- Department of Laboratory Medicine, Zhongshan City People's Hospital, 528403 Zhongshan, Guangdong, China
| | - Miao Mao
- School of Pharmaceutical Sciences, Sun Yat-Sen University, 510006 Guangzhou, Guangdong, China
| | - Lian Huo
- School of Pharmaceutical Sciences, Sun Yat-Sen University, 510006 Guangzhou, Guangdong, China
| | - Guanzhao Wang
- School of Pharmaceutical Sciences, Sun Yat-Sen University, 510006 Guangzhou, Guangdong, China
| | - Zhe Pu
- School of Pharmaceutical Sciences, Sun Yat-Sen University, 510006 Guangzhou, Guangdong, China
| | - Yuanqing Zhang
- Department of Laboratory Medicine, Zhongshan City People's Hospital, 528403 Zhongshan, Guangdong, China
- School of Pharmaceutical Sciences, Sun Yat-Sen University, 510006 Guangzhou, Guangdong, China
| |
Collapse
|
46
|
Espinoza AF, Kureti P, Patel RH, Do SL, Govindu SR, Armbruster BW, Urbicain M, Patel KR, Lopez-Terrada D, Vasudevan SA, Woodfield SE. An indocyanine green-based liquid biopsy test for circulating tumor cells for pediatric liver cancer. Hepatol Commun 2024; 8:e0435. [PMID: 38727682 PMCID: PMC11093570 DOI: 10.1097/hc9.0000000000000435] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/19/2023] [Accepted: 02/05/2024] [Indexed: 05/16/2024] Open
Abstract
BACKGROUND Hepatoblastoma and HCC are the most common malignant hepatocellular tumors seen in children. The aim of this study was to develop a liquid biopsy test for circulating tumor cells (CTCs) for these tumors that would be less invasive and provide real-time information about tumor response to therapy. METHODS For this test, we utilized indocyanine green (ICG), a far-red fluorescent dye used clinically to identify malignant liver cells during surgery. We assessed ICG accumulation in cell lines using fluorescence microscopy and flow cytometry. For our CTC test, we developed a panel of liver tumor-specific markers, including ICG, Glypican-3, and DAPI, and tested it with cell lines and noncancer control blood samples. We then used this panel to analyze whole-blood samples for CTC burden with a cohort of 15 patients with hepatoblastoma and HCC and correlated with patient characteristics and outcomes. RESULTS We showed that ICG accumulation is specific to liver cancer cells, compared to nonmalignant liver cells, non-liver solid tumor cells, and other nonmalignant cells, and can be used to identify liver tumor cells in a mixed population of cells. Experiments with the ICG/Glypican-3/DAPI panel showed that it specifically tagged malignant liver cells. Using patient samples, we found that CTC burden from sequential blood samples from the same patients mirrored the patients' responses to therapy. CONCLUSIONS Our novel ICG-based liquid biopsy test for CTCs can be used to specifically detect and quantify CTCs in the blood of pediatric patients with liver cancer.
Collapse
Affiliation(s)
- Andres F. Espinoza
- Pediatric Surgical Oncology Laboratory, Michael E. DeBakey Department of Surgery, Divisions of Pediatric Surgery and Surgical Research, Texas Children’s Surgical Oncology Program, Texas Children’s Liver Tumor Program, Dan L. Duncan Cancer Center, Baylor College of Medicine, Houston, Texas, USA
| | - Pavan Kureti
- Pediatric Surgical Oncology Laboratory, Michael E. DeBakey Department of Surgery, Divisions of Pediatric Surgery and Surgical Research, Texas Children’s Surgical Oncology Program, Texas Children’s Liver Tumor Program, Dan L. Duncan Cancer Center, Baylor College of Medicine, Houston, Texas, USA
| | - Roma H. Patel
- Pediatric Surgical Oncology Laboratory, Michael E. DeBakey Department of Surgery, Divisions of Pediatric Surgery and Surgical Research, Texas Children’s Surgical Oncology Program, Texas Children’s Liver Tumor Program, Dan L. Duncan Cancer Center, Baylor College of Medicine, Houston, Texas, USA
| | - Susan L. Do
- Pediatric Surgical Oncology Laboratory, Michael E. DeBakey Department of Surgery, Divisions of Pediatric Surgery and Surgical Research, Texas Children’s Surgical Oncology Program, Texas Children’s Liver Tumor Program, Dan L. Duncan Cancer Center, Baylor College of Medicine, Houston, Texas, USA
| | - Saiabhiroop R. Govindu
- Pediatric Surgical Oncology Laboratory, Michael E. DeBakey Department of Surgery, Divisions of Pediatric Surgery and Surgical Research, Texas Children’s Surgical Oncology Program, Texas Children’s Liver Tumor Program, Dan L. Duncan Cancer Center, Baylor College of Medicine, Houston, Texas, USA
| | - Bryan W. Armbruster
- Pediatric Surgical Oncology Laboratory, Michael E. DeBakey Department of Surgery, Divisions of Pediatric Surgery and Surgical Research, Texas Children’s Surgical Oncology Program, Texas Children’s Liver Tumor Program, Dan L. Duncan Cancer Center, Baylor College of Medicine, Houston, Texas, USA
| | - Martin Urbicain
- Department of Pathology and Immunology, Baylor College of Medicine, Texas Children’s Department of Pathology, Houston, Texas, USA
| | - Kalyani R. Patel
- Department of Pathology and Immunology, Baylor College of Medicine, Texas Children’s Department of Pathology, Houston, Texas, USA
| | - Dolores Lopez-Terrada
- Department of Pathology and Immunology, Baylor College of Medicine, Texas Children’s Department of Pathology, Houston, Texas, USA
| | - Sanjeev A. Vasudevan
- Pediatric Surgical Oncology Laboratory, Michael E. DeBakey Department of Surgery, Divisions of Pediatric Surgery and Surgical Research, Texas Children’s Surgical Oncology Program, Texas Children’s Liver Tumor Program, Dan L. Duncan Cancer Center, Baylor College of Medicine, Houston, Texas, USA
| | - Sarah E. Woodfield
- Pediatric Surgical Oncology Laboratory, Michael E. DeBakey Department of Surgery, Divisions of Pediatric Surgery and Surgical Research, Texas Children’s Surgical Oncology Program, Texas Children’s Liver Tumor Program, Dan L. Duncan Cancer Center, Baylor College of Medicine, Houston, Texas, USA
| |
Collapse
|
47
|
Lyu N, Hassanzadeh-Barforoushi A, Rey Gomez LM, Zhang W, Wang Y. SERS biosensors for liquid biopsy towards cancer diagnosis by detection of various circulating biomarkers: current progress and perspectives. NANO CONVERGENCE 2024; 11:22. [PMID: 38811455 PMCID: PMC11136937 DOI: 10.1186/s40580-024-00428-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/31/2024] [Accepted: 05/09/2024] [Indexed: 05/31/2024]
Abstract
Liquid biopsy has emerged as a promising non-invasive strategy for cancer diagnosis, enabling the detection of various circulating biomarkers, including circulating tumor cells (CTCs), circulating tumor nucleic acids (ctNAs), circulating tumor-derived small extracellular vesicles (sEVs), and circulating proteins. Surface-enhanced Raman scattering (SERS) biosensors have revolutionized liquid biopsy by offering sensitive and specific detection methodologies for these biomarkers. This review comprehensively examines the application of SERS-based biosensors for identification and analysis of various circulating biomarkers including CTCs, ctNAs, sEVs and proteins in liquid biopsy for cancer diagnosis. The discussion encompasses a diverse range of SERS biosensor platforms, including label-free SERS assay, magnetic bead-based SERS assay, microfluidic device-based SERS system, and paper-based SERS assay, each demonstrating unique capabilities in enhancing the sensitivity and specificity for detection of liquid biopsy cancer biomarkers. This review critically assesses the strengths, limitations, and future directions of SERS biosensors in liquid biopsy for cancer diagnosis.
Collapse
Affiliation(s)
- Nana Lyu
- School of Natural Sciences, Macquarie University, Sydney, NSW, 2109, Australia
| | | | - Laura M Rey Gomez
- School of Natural Sciences, Macquarie University, Sydney, NSW, 2109, Australia
| | - Wei Zhang
- School of Natural Sciences, Macquarie University, Sydney, NSW, 2109, Australia
| | - Yuling Wang
- School of Natural Sciences, Macquarie University, Sydney, NSW, 2109, Australia.
| |
Collapse
|
48
|
Le MCN, Smith KA, Dopico PJ, Greer B, Alipanah M, Zhang Y, Siemann DW, Lagmay JP, Fan ZH. Investigating surface proteins and antibody combinations for detecting circulating tumor cells of various sarcomas. Sci Rep 2024; 14:12374. [PMID: 38811642 PMCID: PMC11137101 DOI: 10.1038/s41598-024-61651-w] [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: 01/21/2024] [Accepted: 05/08/2024] [Indexed: 05/31/2024] Open
Abstract
Circulating tumor cells (CTCs) have gathered attention as a biomarker for carcinomas. However, CTCs in sarcomas have received little attention. In this work, we investigated cell surface proteins and antibody combinations for immunofluorescence detection of sarcoma CTCs. A microfluidic device that combines filtration and immunoaffinity using gangliosides 2 and cell surface vimentin (CSV) antibodies was employed to capture CTCs. For CTC detection, antibodies against cytokeratins 7 and 8 (CK), pan-cytokeratin (panCK), or a combination of panCK and CSV were used. Thirty-nine blood samples were collected from 21 patients of various sarcoma subtypes. In the independent samples study, samples were subjected to one of three antibody combination choices. Significant difference in CTC enumeration was found between CK and panCK + CSV, and between panCK and panCK + CSV. Upon stratification of CK+ samples, those of metastatic disease had a higher CTC number than those of localized disease. In the paired samples study involving cytokeratin-positive sarcoma subtypes, using panCK antibody detected more CTCs than CK. Similarly, for osteosarcoma, using panCK + CSV combination resulted in a higher CTC count than panCK. This study emphasized deliberate selection of cell surface proteins for sarcoma CTC detection and subtype stratification for studying cancers as heterogeneous as sarcomas.
Collapse
Affiliation(s)
- Minh-Chau N Le
- Interdisciplinary Microsystems Group, Department of Mechanical and Aerospace Engineering, University of Florida, PO Box 116250, Gainesville, FL, 32611, USA
| | - Kierstin A Smith
- Interdisciplinary Microsystems Group, Department of Mechanical and Aerospace Engineering, University of Florida, PO Box 116250, Gainesville, FL, 32611, USA
| | - Pablo J Dopico
- Interdisciplinary Microsystems Group, Department of Mechanical and Aerospace Engineering, University of Florida, PO Box 116250, Gainesville, FL, 32611, USA
| | - Beate Greer
- Department of Pediatrics, Division of Hematology-Oncology, University of Florida, Gainesville, FL, 32610, USA
| | - Morteza Alipanah
- Interdisciplinary Microsystems Group, Department of Mechanical and Aerospace Engineering, University of Florida, PO Box 116250, Gainesville, FL, 32611, USA
| | - Yang Zhang
- Interdisciplinary Microsystems Group, Department of Mechanical and Aerospace Engineering, University of Florida, PO Box 116250, Gainesville, FL, 32611, USA
| | - Dietmar W Siemann
- Department of Radiation Oncology, University of Florida, Gainesville, FL, 32610, USA
| | - Joanne P Lagmay
- Department of Pediatrics, Division of Hematology-Oncology, University of Florida, Gainesville, FL, 32610, USA.
| | - Z Hugh Fan
- Interdisciplinary Microsystems Group, Department of Mechanical and Aerospace Engineering, University of Florida, PO Box 116250, Gainesville, FL, 32611, USA.
- J. Crayton Pruitt Family Department of Biomedical Engineering, University of Florida, Gainesville, FL, 32611, USA.
| |
Collapse
|
49
|
Magnusson C, Augustsson P, Undvall Anand E, Lenshof A, Josefsson A, Welén K, Bjartell A, Ceder Y, Lilja H, Laurell T. Acoustic Enrichment of Heterogeneous Circulating Tumor Cells and Clusters from Metastatic Prostate Cancer Patients. Anal Chem 2024; 96:6914-6921. [PMID: 38655666 PMCID: PMC11079855 DOI: 10.1021/acs.analchem.3c05371] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2023] [Revised: 04/16/2024] [Accepted: 04/17/2024] [Indexed: 04/26/2024]
Abstract
BACKGROUND There are important unmet clinical needs to develop cell enrichment technologies to enable unbiased label-free isolation of both single cell and clusters of circulating tumor cells (CTCs) manifesting heterogeneous lineage specificity. Here, we report a pilot study based on the microfluidic acoustophoresis enrichment of CTCs using the CellSearch CTC assay as a reference modality. METHODS Acoustophoresis uses an ultrasonic standing wave field to separate cells based on biomechanical properties (size, density, and compressibility), resulting in inherently label-free and epitope-independent cell enrichment. Following red blood cell lysis and paraformaldehyde fixation, 6 mL of whole blood from 12 patients with metastatic prostate cancer and 20 healthy controls were processed with acoustophoresis and subsequent image cytometry. RESULTS Acoustophoresis enabled enrichment and characterization of phenotypic CTCs (EpCAM+, Cytokeratin+, DAPI+, CD45-/CD66b-) in all patients with metastatic prostate cancer and detected CTC-clusters composed of only CTCs or heterogeneous aggregates of CTCs clustered with various types of white blood cells in 9 out of 12 patients. By contrast, CellSearch did not detect any CTC clusters, but detected comparable numbers of phenotypic CTCs as acoustophoresis, with trends of finding a higher number of CTCs using acoustophoresis. CONCLUSION Our preliminary data indicate that acoustophoresis provides excellent possibilities to detect and characterize CTC clusters as a putative marker of metastatic disease and outcomes. Moreover, acoustophoresis enables the sensitive label-free enrichment of cells with epithelial phenotypes in blood and offers opportunities to detect and characterize CTCs undergoing epithelial-to-mesenchymal transitioning and lineage plasticity.
Collapse
Affiliation(s)
- Cecilia Magnusson
- Department of Translational Medicine, Lund University, Lund SE-22100, Sweden
| | - Per Augustsson
- Department of Biomedical Engineering, Lund University, Lund SE-22100, Sweden
| | - Eva Undvall Anand
- Department of Biomedical Engineering, Lund University, Lund SE-22100, Sweden
| | - Andreas Lenshof
- Department of Biomedical Engineering, Lund University, Lund SE-22100, Sweden
| | - Andreas Josefsson
- Institute of Clinical Sciences, Department of Urology, Gothenburg University, Gothenburg SE-41345, Sweden
- Wallenberg Center for Molecular Medicine, Umeå University, Umeå SE-90187, Sweden
- Department of Urology and Andrology, Institute of Surgery and Perioperative Sciences, Umeå University, Umeå SE-90185, Sweden
| | - Karin Welén
- Institute of Clinical Sciences, Department of Urology, Gothenburg University, Gothenburg SE-41345, Sweden
| | - Anders Bjartell
- Department of Translational Cancer Research, Lund University, Lund SE-22100, Sweden
| | - Yvonne Ceder
- Department of Laboratory Medicine, Lund University, Lund SE-22100, Sweden
| | - Hans Lilja
- Department of Translational Medicine, Lund University, Lund SE-22100, Sweden
- Department of Pathology and Laboratory Medicine, Surgery (Urology), and Medicine (GU Oncology), Memorial Sloan-Kettering Cancer Center, New York, New York 10065, United States
| | - Thomas Laurell
- Department of Biomedical Engineering, Lund University, Lund SE-22100, Sweden
| |
Collapse
|
50
|
Wang J, Liu X, Li J, Chen W. Digital Circulating Tumor Cells Quantification. Anal Chem 2024; 96:6881-6888. [PMID: 38659346 DOI: 10.1021/acs.analchem.3c02769] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/26/2024]
Abstract
Circulating tumor cells (CTCs) are an emerging but vital biomarker for cancer management. An efficient methodology for accurately quantifying CTCs remains challenging due to their rareness. Here, we develop a digital CTC detection strategy using partitioning instead of enrichment to quantify CTCs. By utilizing the characteristics of droplet microfluidics that can rapidly generate a large number of parallel independent reactors, combined with Poisson distribution, we realize the quantification of CTCs in the blood directly. The limit of detection of our digital CTCs quantification assay is five cells per 5 mL of whole blood. By simultaneously detecting multiple genetic mutations, our approach achieves highly sensitive and specific detection of CTCs in peripheral blood from NSCLC patients (AUC = 1). Our digital platform offers a potential approach and strategy for the quantification of CTCs, which could contribute to the advancement of cancer medical management.
Collapse
Affiliation(s)
- Jidong Wang
- Medical Research Center, Huazhong University of Science and Technology Union Shenzhen Hospital, the Sixth Affiliated Hospital, Shenzhen University Medical School, Shenzhen University, Shenzhen 518052, People's Republic of China
| | - Xiaolei Liu
- School of Biomedical Engineering, Shenzhen University Medical School, Shenzhen University, Shenzhen 518055, People's Republic of China
| | - Jiang Li
- Gynecology Department, Huazhong University of Science and Technology Union Shenzhen Hospital, the Sixth Affiliated Hospital, Shenzhen University Medical School, Shenzhen University, Shenzhen 518052, People's Republic of China
| | - Wenwen Chen
- School of Biomedical Engineering, Shenzhen University Medical School, Shenzhen University, Shenzhen 518055, People's Republic of China
| |
Collapse
|