1
|
Liatsou E, Kollias I, Trapali M, Tsilimigras DI, Gavriatopoulou M, Ntanasis-Stathopoulos I. Liquid Biopsies in the Early Diagnosis, Prognosis, and Tailored Treatment of Colorectal Cancer. Cancers (Basel) 2025; 17:927. [PMID: 40149264 PMCID: PMC11940745 DOI: 10.3390/cancers17060927] [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: 10/25/2024] [Revised: 02/08/2025] [Accepted: 03/04/2025] [Indexed: 03/29/2025] Open
Abstract
INTRODUCTION Liquid biopsies provide a less-invasive option to tissue biopsies for the early diagnosis, prognosis, and tailored therapy of colorectal cancer (CRC). CRC is a major cause of cancer-related death, and early identification is essential for improving patient outcomes. REVIEW Conventional diagnostic techniques, including colonoscopy and tissue biopsy, may be enhanced by liquid biopsies that examine circulating tumor cells (CTCs), circulating tumor DNA (ctDNA), extracellular vesicles (EVs), and other indicators present in body fluids. These markers provide significant insights into tumor biology, heterogeneity, and therapeutic response. CTCs detected in early-stage CRC have prognostic significance for disease recurrence and survival, while ctDNA investigation may uncover genetic mutations, epigenetic alterations, and tumor development. The identification of ctDNA in minimal residual disease (MRD) postsurgery correlates with an elevated risk of recurrence and unfavorable prognosis, underscoring its use in assessing treatment effectiveness. Furthermore, non-coding RNAs (ncRNAs) contained inside EVs provide potential prospective biomarkers and therapeutic targets, facilitating diagnosis and treatment assessment. Notwithstanding the potential of liquid biopsies, obstacles persist in assay standardization, sensitivity enhancement, and the management of tumor heterogeneity. Additional extensive research is required to determine their function in clinical practice. CONCLUSION Overall, liquid biopsies serve as a potential instrument for real-time monitoring, evaluating therapy responses, and directing individualized therapeutic strategies in CRC patients.
Collapse
Affiliation(s)
- Efstathia Liatsou
- CAST, Center of Allogenic Transplantation and Cell Therapies, Karolinska University, 17177 Stockholm, Sweden;
| | - Ioannis Kollias
- Department of Clinical Therapeutics, National and Kapodistrian University of Athens, 11528 Athens, Greece; (I.K.); (M.G.)
| | - Maria Trapali
- Laboratory of Chemistry, Biochemistry and Cosmetic Science, Department of Biomedical Medicine, University of West Attica, 12243 Egaleo, Greece;
| | - Diamantis I. Tsilimigras
- Department of Surgery, Division of Surgical Oncology, The Ohio State University Wexner Medical Center and James Comprehensive Cancer Center, Columbus, OH 43210, USA;
| | - Maria Gavriatopoulou
- Department of Clinical Therapeutics, National and Kapodistrian University of Athens, 11528 Athens, Greece; (I.K.); (M.G.)
| | - Ioannis Ntanasis-Stathopoulos
- Department of Clinical Therapeutics, National and Kapodistrian University of Athens, 11528 Athens, Greece; (I.K.); (M.G.)
| |
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
|
Song J, Ye X, Xiao H. Liquid biopsy entering clinical practice: Past discoveries, current insights, and future innovations. Crit Rev Oncol Hematol 2025; 207:104613. [PMID: 39756526 DOI: 10.1016/j.critrevonc.2025.104613] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2024] [Revised: 12/22/2024] [Accepted: 01/02/2025] [Indexed: 01/07/2025] Open
Abstract
In recent years, liquid biopsy has gained prominence as an emerging biomarker in cancer research, providing critical insights into tumor biology and metastasis. Technological advancements have enabled its integration into clinical practice, with ongoing trials demonstrating encouraging outcomes. Key applications of liquid biopsy include early cancer detection, cancer staging, prognosis evaluation, and real-time monitoring of tumor progression to optimize treatment decisions. In this review, we present a comprehensive conceptual framework for liquid biopsy, discuss the challenges in its research and clinical application, and highlight its significant potential in identifying therapeutic targets and resistance mechanisms across various cancer types. Furthermore, we explore the emerging role of liquid biopsy-based multicancer screening, which has shown promising advancements. Looking ahead, standardization, multi-omics coanalysis, and the advancement of precision medicine and personalized treatments are expected to drive the future development and integration of liquid biopsy into routine clinical workflows, enhancing cancer diagnosis and treatment management.
Collapse
Affiliation(s)
- Jinghan Song
- Department of Respiratory and Critical Care Medicine, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xiong Ye
- School of Clinical Medicine, Shanghai University of Medicine & Health Sciences, Shanghai, China
| | - Hui Xiao
- Department of Respiratory and Critical Care Medicine, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| |
Collapse
|
4
|
Fey RM, Billo A, Clister T, Doan KL, Berry EG, Tibbitts DC, Kulkarni RP. Personalization of Cancer Treatment: Exploring the Role of Chronotherapy in Immune Checkpoint Inhibitor Efficacy. Cancers (Basel) 2025; 17:732. [PMID: 40075580 PMCID: PMC11899640 DOI: 10.3390/cancers17050732] [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: 11/04/2024] [Revised: 02/01/2025] [Accepted: 02/15/2025] [Indexed: 03/14/2025] Open
Abstract
In the era of precision medicine, mounting evidence suggests that the time of therapy administration, or chronotherapy, has a great impact on treatment outcomes. Chronotherapy involves planning treatment timing by considering circadian rhythms, which are 24 h oscillations in behavior and physiology driven by synchronized molecular clocks throughout the body. The value of chronotherapy in cancer treatment is currently under investigation, notably in the effects of treatment timing on efficacy and side effects. Immune checkpoint inhibitor (ICI) therapy is a promising cancer treatment. However, many patients still experience disease progression or need to stop the therapy early due to side effects. There is accumulating evidence that the time of day at which ICI therapy is administered can have a substantial effect on ICI efficacy. Thus, it is important to investigate the intersections of circadian rhythms, chronotherapy, and ICI efficacy. In this review, we provide a brief overview of circadian rhythms in the context of immunity and cancer. Additionally, we outline current applications of chronotherapy for cancer treatment. We synthesize the 29 studies conducted to date that examine the impact of time-of-day administration on the efficacy of ICI therapy, its associated side effects, and sex differences in both efficacy and side effects. We also discuss potential mechanisms underlying these observed results. Finally, we highlight the challenges in this area and future directions for research, including the potential for a chronotherapeutic personalized medicine approach that tailors the time of ICI administration to individual patients' circadian rhythms.
Collapse
Affiliation(s)
- Rosalyn M. Fey
- Department of Dermatology, Oregon Health & Science University, Portland, OR 97239, USA; (R.M.F.)
| | - Avery Billo
- Department of Dermatology, Oregon Health & Science University, Portland, OR 97239, USA; (R.M.F.)
| | - Terri Clister
- Department of Dermatology, Oregon Health & Science University, Portland, OR 97239, USA; (R.M.F.)
| | - Khanh L. Doan
- Department of Dermatology, Oregon Health & Science University, Portland, OR 97239, USA; (R.M.F.)
| | - Elizabeth G. Berry
- Department of Dermatology, Oregon Health & Science University, Portland, OR 97239, USA; (R.M.F.)
| | - Deanne C. Tibbitts
- Division of Oncological Sciences, Oregon Health & Science University, Portland, OR 97239, USA
- Knight Cancer Institute, Oregon Health & Science University, Portland, OR 97239, USA
| | - Rajan P. Kulkarni
- Department of Dermatology, Oregon Health & Science University, Portland, OR 97239, USA; (R.M.F.)
- Knight Cancer Institute, Oregon Health & Science University, Portland, OR 97239, USA
- Cancer Early Detection Advanced Research Center (CEDAR), Portland, OR 97239, USA
- Operative Care Division, U.S. Department of Veterans Affairs Portland Health Care System, Portland, OR 97239, USA
| |
Collapse
|
5
|
Sun D, Yu B, Xu R, Wang C, Pang K. Laser-Induced Minimization of Circulating Tumor Cells to Suppress Tumor Metastasis. JOURNAL OF BIOPHOTONICS 2025:e202400569. [PMID: 39909048 DOI: 10.1002/jbio.202400569] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/19/2024] [Revised: 01/19/2025] [Accepted: 01/24/2025] [Indexed: 02/07/2025]
Abstract
Circulating tumor cells (CTCs) are key indicators of tumor metastasis. Effective clearance of CTCs can reduce the probability of metastasis. We designed a system for the real-time dynamic monitoring and clearance of CTCs, capable of monitoring and clearing CTCs in the living circulatory system. Experimental results showed that pulsed laser treatment significantly affects the clearance of melanoma CTCs. Through in vivo imaging of small animals and survival analysis of mice, we observed that CTC clearance could reduce the size of distant metastatic lesions and prolong the lifespan of the mice. Additionally, we set up a hemolysis experiment to demonstrate that the laser energy used does not cause damage to red blood cells. This study is based on the physical and mechanical destruction of CTCs, meaning there is no issue of drug resistance. This provides a novel approach and technical means for suppressing tumor metastasis and extending the lifespan of patients in clinical settings.
Collapse
Affiliation(s)
- Da Sun
- School of Instrument Science and Opto-Electronics Engineering, Beijing Information Science and Technology University, Beijing, China
| | - Bingchen Yu
- School of Instrument Science and Opto-Electronics Engineering, Beijing Information Science and Technology University, Beijing, China
| | - Rui Xu
- School of Instrument Science and Opto-Electronics Engineering, Beijing Information Science and Technology University, Beijing, China
| | - Chenzheng Wang
- School of Instrument Science and Opto-Electronics Engineering, Beijing Information Science and Technology University, Beijing, China
| | - Kai Pang
- School of Instrument Science and Opto-Electronics Engineering, Beijing Information Science and Technology University, Beijing, China
| |
Collapse
|
6
|
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
|
7
|
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
|
8
|
Yu B, Zhu Y, Zhang F, Sun D, Xu R, Wang C, Pang K. A Miniaturized In Vivo Fluorescence Microscopy Method for Monitoring Circulating Tumor Cells in Freely Moving Animals. JOURNAL OF BIOPHOTONICS 2025; 18:e202400496. [PMID: 39716436 DOI: 10.1002/jbio.202400496] [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: 11/10/2024] [Revised: 12/08/2024] [Accepted: 12/12/2024] [Indexed: 12/25/2024]
Abstract
Metastasis is the leading cause of death in tumor patients, with circulating tumor cells (CTCs) serving as key biomarkers for tumor progression, metastasis, and recurrence. CTC quantity is closely linked to tumor dynamics, which are influenced by biological rhythms. Studying CTC distribution under various physiological conditions provides insights into metastasis mechanisms. However, due to the low abundance of CTCs, detection accuracy is limited, especially with small blood samples, making continuous data collection challenging. To address this, we developed a dual-channel miniaturized in vivo fluorescence microscopy system for real-time monitoring of CTCs in experimental animals. This system, which can be fixed to the head or back, enables dynamic, quantitative analysis of CTCs in the circulatory system. It offers a valuable tool for investigating tumor metastasis rhythms, drug evaluation, and prognostic assessment in freely moving animals, advancing research in metastasis mechanisms and cancer treatment.
Collapse
Affiliation(s)
- Bingchen Yu
- School of Instrument Science and Opto-Electronics Engineering, Beijing Information Science and Technology University, Beijing, China
| | - Yuxi Zhu
- School of Instrument Science and Opto-Electronics Engineering, Beijing Information Science and Technology University, Beijing, China
| | - Fan Zhang
- School of Instrument Science and Opto-Electronics Engineering, Beijing Information Science and Technology University, Beijing, China
| | - Da Sun
- School of Instrument Science and Opto-Electronics Engineering, Beijing Information Science and Technology University, Beijing, China
| | - Rui Xu
- School of Instrument Science and Opto-Electronics Engineering, Beijing Information Science and Technology University, Beijing, China
| | - Chenzheng Wang
- School of Instrument Science and Opto-Electronics Engineering, Beijing Information Science and Technology University, Beijing, China
| | - Kai Pang
- School of Instrument Science and Opto-Electronics Engineering, Beijing Information Science and Technology University, Beijing, China
| |
Collapse
|
9
|
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
|
10
|
Wang C, Gu H, Cai J, Zhu C, Zheng Q, Xu H, Wang L, Wan Y. Enhanced detection of circulating tumor cells using a MUC1 promoter-driven recombinant adenovirus. Front Oncol 2025; 14:1506968. [PMID: 39886667 PMCID: PMC11779711 DOI: 10.3389/fonc.2024.1506968] [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: 10/06/2024] [Accepted: 12/27/2024] [Indexed: 02/01/2025] Open
Abstract
Introduction Circulating tumor cells (CTCs) have attracted significant interest as a biomarker for cancer diagnosis. In this study, we judiciously constructed a recombinant MUC1-dependent adenovirus (rAdF35-MUC1) that can selectively replicate and overexpress copepod super green fluorescent proteins (copGFP) in MUC1-positive tumor cells to investigate its role in the detection of CTCs. Methods We conducted a comparative study between rAdF35-MUC1 and the existing hTERT-dependent adenovirus (rAdF35-hTERT). Breast cancer cell lines and healthy human peripheral blood mononuclear cells (PBMCs) were infected with both viral constructs to evaluate infection efficiency and the incidence of false-positive cells. CTC Model Samples were employed to determine detection rates, and clinical samples from breast cancer patients were analyzed to preliminarily evaluate the efficacy of CTC detection in a clinical context. Results In preclinical and clinical studies, rAdF35-MUC1 exhibited a significantly high detection efficiency for breast cancer cells, outperforming the existing hTERT-dependent adenovirus (rAdF35-hTERT), especially in detecting CTCs at low quantities. Moreover, rAdF35-MUC1 demonstrated reduced incidence of false positives in healthy PBMCs compared to rAdF35-hTERT. Conclusion In brief, rAdF35-MUC1 emerges as a potent tool for the sensitive and specific identification of CTCs derived from breast cancer patients, holding clinical translation potential for advancing cancer (early) diagnosis, treatment monitoring, and prognosis.
Collapse
Affiliation(s)
- Cheng Wang
- Department of Radiation Oncology, Nanjing Hospital Affiliated to Nanjing University of Chinese Medicine (The Second Hospital of Nanjing), Jiangsu, Nanjing, China
| | - Huihui Gu
- Department of Radiation Oncology, Nanjing Hospital Affiliated to Nanjing University of Chinese Medicine (The Second Hospital of Nanjing), Jiangsu, Nanjing, China
| | - Jia Cai
- Department of Radiation Oncology, Nanjing Hospital Affiliated to Nanjing University of Chinese Medicine (The Second Hospital of Nanjing), Jiangsu, Nanjing, China
| | - Chuandong Zhu
- Department of Oncology, Nanjing Hospital Affiliated to Nanjing University of Chinese Medicine (The Second Hospital of Nanjing), Jiangsu, Nanjing, China
| | - Qin Zheng
- Department of Oncology, Nanjing Hospital Affiliated to Nanjing University of Chinese Medicine (The Second Hospital of Nanjing), Jiangsu, Nanjing, China
| | - Hanfeng Xu
- Department of Oncology, Nanjing Hospital Affiliated to Nanjing University of Chinese Medicine (The Second Hospital of Nanjing), Jiangsu, Nanjing, China
| | - Lixue Wang
- Department of Radiation Oncology, Nanjing Hospital Affiliated to Nanjing University of Chinese Medicine (The Second Hospital of Nanjing), Jiangsu, Nanjing, China
| | - Yuan Wan
- The Pq Laboratory of BiomeDx/Rx, Department of Biomedical Engineering, Binghamton University, Binghamton, NY, United States
| |
Collapse
|
11
|
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
|
12
|
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
|
13
|
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
|
14
|
Ma C, Li Y, Zhu H, Li Z, Liu Y. Clinical applications of circulating tumor cell detection: challenges and strategies. Clin Chem Lab Med 2024:cclm-2024-0959. [PMID: 39610299 DOI: 10.1515/cclm-2024-0959] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2024] [Accepted: 11/15/2024] [Indexed: 11/30/2024]
Abstract
Circulating tumor cells (CTCs) are pivotal in the distant metastasis of tumors, serving as one of the primary materials for liquid biopsy. They hold significant clinical importance in assessing prognosis, predicting efficacy, evaluating therapeutic outcomes, and studying recurrence, metastasis, and resistance mechanisms in cancer patients. Nevertheless, the rareness and heterogeneity of CTC and the complexity of metastasis make the clinical application of CTC detection confront many challenges, which may need to be settled by some practical strategies. This article will review the content mentioned above.
Collapse
Affiliation(s)
- Chunhui Ma
- Faculty of Medical Imaging, Naval Medical University, Shanghai, China
| | - Yang Li
- Faculty of Medical Imaging, Naval Medical University, Shanghai, China
| | - Hai Zhu
- Faculty of Medical Imaging, Naval Medical University, Shanghai, China
| | - Zhiyong Li
- Faculty of Medical Imaging, Naval Medical University, Shanghai, China
| | - Yi Liu
- 26460 The Fifth Medical Center of Chinese PLA General Hospital , Beijing, China
| |
Collapse
|
15
|
Grisanti S, Sonntag SR, Tura SA. [Circulating tumor cells in uveal melanoma : "The needle in the haystack"]. DIE OPHTHALMOLOGIE 2024; 121:954-962. [PMID: 39580374 DOI: 10.1007/s00347-024-02136-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/01/2024] [Revised: 10/22/2024] [Accepted: 10/22/2024] [Indexed: 11/25/2024]
Abstract
BACKGROUND A fundamental prerequisite for the development of clinically manifested metastases in uveal melanoma (UM) is the hematogenous dissemination of melanoma cells, which circulate in the systemic circulation, deposit in organs and can subsequently colonize the organs. MATERIAL AND METHODS This article presents a review of the literature on the detection of circulating tumor cells (CTC) from UM and their relevance for the clinical and pathophysiological aspects of the malignancy. RESULTS Since the first description of CTC in UM patients 20 years ago, only 20 articles have been published on the detection of intact CTCs in UM. Besides demonstrating the presence of CTCs in the systemic circulation, these articles give insights into the pathophysiology and chronology of CTC dissemination, the diagnostics and risk stratification of UM. CONCLUSION The detection of CTC in UM has proved to be beneficial in many ways. The early time of cell dissemination and also the associated factors should lead to a rethinking in the clinical routine in the context of the fatal danger of metastases.
Collapse
Affiliation(s)
- Salvatore Grisanti
- Klinik für Augenheilkunde, Universität zu Lübeck, Universitätsklinikum Schleswig-Holstein, Ratzeburger Allee 160, 23562, Lübeck, Deutschland.
| | - S R Sonntag
- Klinik für Augenheilkunde, Universität zu Lübeck, Universitätsklinikum Schleswig-Holstein, Ratzeburger Allee 160, 23562, Lübeck, Deutschland
| | - S A Tura
- Klinik für Augenheilkunde, Universität zu Lübeck, Universitätsklinikum Schleswig-Holstein, Ratzeburger Allee 160, 23562, Lübeck, Deutschland
| |
Collapse
|
16
|
Feng T, Hu J, Wen J, Qian Z, Che G, Zhou Q, Zhu L. Personalized nanovaccines for treating solid cancer metastases. J Hematol Oncol 2024; 17:115. [PMID: 39609851 PMCID: PMC11603676 DOI: 10.1186/s13045-024-01628-4] [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: 09/11/2024] [Accepted: 10/25/2024] [Indexed: 11/30/2024] Open
Abstract
Cancer vaccines have garnered attention as a potential treatment for cancer metastases. Nevertheless, the clinical response rate to vaccines remains < 30%. Nanoparticles stabilize vaccines and improve antigen recognition and presentation, resulting in high tumor penetration or accumulation, effective co-distribution of drugs to the secondary lymphatic system, and adaptable antigen or adjuvant administration. Such vaccine-like nanomedicines have the ability to eradicate the primary tumors as well as to prevent or eliminate metastases. This review examines state-of-the-art nanocarriers developed to deliver tumor vaccines to metastases, including synthetic, semi-biogenic, and biogenic nanosystems. Moreover, it highlights the physical and pharmacological properties that enhance their anti-metastasis efficiency. This review also addresses the combination of nanovaccines with cancer immunotherapy to target various steps in the metastatic cascade, drawing insights from preclinical and clinical studies. The review concludes with a critical analysis of the challenges and frameworks linked to the clinical translation of cancer nanovaccines.
Collapse
Affiliation(s)
- Tang Feng
- Department of Medical Oncology, Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China
- Department of Biotherapy, Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Jia Hu
- Department of Medical Oncology, Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China
- Lung Cancer Center/Lung Cancer Institute, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Jirui Wen
- Deep Underground Space Medical Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Zhiyong Qian
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Guowei Che
- Lung Cancer Center/Lung Cancer Institute, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Qinghua Zhou
- Lung Cancer Center/Lung Cancer Institute, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Lingling Zhu
- Department of Medical Oncology, Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China.
- Lung Cancer Center/Lung Cancer Institute, West China Hospital, Sichuan University, Chengdu, Sichuan, China.
| |
Collapse
|
17
|
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
|
18
|
Cotan HT, Emilescu RA, Iaciu CI, Orlov-Slavu CM, Olaru MC, Popa AM, Jinga M, Nitipir C, Schreiner OD, Ciobanu RC. Prognostic and Predictive Determinants of Colorectal Cancer: A Comprehensive Review. Cancers (Basel) 2024; 16:3928. [PMID: 39682117 DOI: 10.3390/cancers16233928] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2024] [Revised: 11/20/2024] [Accepted: 11/22/2024] [Indexed: 12/18/2024] Open
Abstract
Colorectal cancer (CRC) remains a significant global health burden, necessitating a thorough understanding of prognostic and predictive factors to enhance patient outcomes. This systematic review aims to comprehensively evaluate prognostic and predictive determinants in CRC, encompassing both traditional and emerging biomarkers. A systematic search of major electronic databases was conducted to identify relevant studies published from 1995 up to 2024. Eligible articles were critically appraised, and data extraction was performed according to predefined criteria. The prognostic determinants examined included clinicopathological features such as tumor stage, grade, and lymph node involvement, as well as molecular biomarkers including RAS, BRAF, and MSI status. Predictive determinants encompassed biomarkers influencing response to targeted therapies and immunotherapy, such as HER2 and Immunoscore. The review also explores novel prognostic and predictive markers, including tumor microenvironment characteristics and liquid biopsy-based biomarkers. Synthesizing evidence from diverse studies, this review provides insights into the prognostic and predictive landscape of CRC, highlighting the potential clinical implications of identified determinants. Understanding the multifaceted nature of prognostic and predictive factors in CRC is imperative for the advancement of personalized treatment strategies and improvement of patient outcomes.
Collapse
Affiliation(s)
- Horia T Cotan
- General Medicine Faculty, Carol Davila University of Medicine and Pharmacy, 8 Sanitary Heroes Boulevard, 050474 Bucharest, Romania
| | - Radu A Emilescu
- General Medicine Faculty, Carol Davila University of Medicine and Pharmacy, 8 Sanitary Heroes Boulevard, 050474 Bucharest, Romania
| | - Cristian I Iaciu
- General Medicine Faculty, Carol Davila University of Medicine and Pharmacy, 8 Sanitary Heroes Boulevard, 050474 Bucharest, Romania
| | - Cristina M Orlov-Slavu
- General Medicine Faculty, Carol Davila University of Medicine and Pharmacy, 8 Sanitary Heroes Boulevard, 050474 Bucharest, Romania
| | - Mihaela C Olaru
- General Medicine Faculty, Carol Davila University of Medicine and Pharmacy, 8 Sanitary Heroes Boulevard, 050474 Bucharest, Romania
| | - Ana M Popa
- General Medicine Faculty, Carol Davila University of Medicine and Pharmacy, 8 Sanitary Heroes Boulevard, 050474 Bucharest, Romania
| | - Mariana Jinga
- General Medicine Faculty, Carol Davila University of Medicine and Pharmacy, 8 Sanitary Heroes Boulevard, 050474 Bucharest, Romania
| | - Cornelia Nitipir
- General Medicine Faculty, Carol Davila University of Medicine and Pharmacy, 8 Sanitary Heroes Boulevard, 050474 Bucharest, Romania
| | - Oliver Daniel Schreiner
- Regional Institute of Oncology Iasi, 2-4 General Henri Mathias Berthelot Street, 700483 Iasi, Romania
- Department 3-Medical Sciences, Grigore T. Popa University of Medicine and Pharmacy, 16 University Street, 700115 Iasi, Romania
- Department of Electrical Measurements and Materials, Gheorghe Asachi Technical University, 700050 Iasi, Romania
| | - Romeo Cristian Ciobanu
- Department of Electrical Measurements and Materials, Gheorghe Asachi Technical University, 700050 Iasi, Romania
| |
Collapse
|
19
|
Hoff CO, Manzi J, Ferreira R, Chauhan A, Housein P, Merchant N, Livingstone A, Vianna R, Abreu P. A neuroendocrine biomarker revolution from monoanalyte to multianalyte biomarkers in non-functioning gastro-entero-pancreatic neuroendocrine neoplasms. Crit Rev Oncol Hematol 2024; 203:104460. [PMID: 39153703 DOI: 10.1016/j.critrevonc.2024.104460] [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: 06/12/2023] [Revised: 07/25/2024] [Accepted: 07/26/2024] [Indexed: 08/19/2024] Open
Abstract
Neuroendocrine neoplasms (NENs) arise from neuroendocrine cells in a wide variety of organs. One of the most affected disease sites is the gastrointestinal system, which originates the gastro-entero-pancreatic NENs (GEP-NENs), a heterogenous group of malignancies that are rapidly increasing in incidence. These tumors can be functioning, with secretory activity leading to identifiable clinical syndromes, or non-functioning, with no secretory activity but with local symptoms of tumor growth and metastasis. A limitation in biomarkers is a crucial unmet need in non-secretory NEN management, as clinical decision-making is made more difficult by obstacles in tumor classification, prognostic evaluation, assessment of treatment response and surveillance. The objective of this review is to present existing and novel biomarkers for NENs that can function as prognostic factors and monitor disease progression or regression longitudinally, with a special emphasis on innovative research into novel multianalyte biomarkers.
Collapse
Affiliation(s)
- Camilla O Hoff
- University of Sao Paulo Medical School, University of Sao Paulo, Sao Paulo, Brazil; Miami Transplant Institute, Jackson Memorial Hospital, University of Miami, Miami, USA
| | - Joao Manzi
- University of Sao Paulo Medical School, University of Sao Paulo, Sao Paulo, Brazil
| | - Raphaella Ferreira
- Miami Transplant Institute, Jackson Memorial Hospital, University of Miami, Miami, USA
| | - Aman Chauhan
- Sylvester Comprehensive Cancer Center, University of Miami, Miami, USA
| | - Peter Housein
- Sylvester Comprehensive Cancer Center, University of Miami, Miami, USA
| | - Nipun Merchant
- Sylvester Comprehensive Cancer Center, University of Miami, Miami, USA
| | - Alan Livingstone
- Sylvester Comprehensive Cancer Center, University of Miami, Miami, USA
| | - Rodrigo Vianna
- Miami Transplant Institute, Jackson Memorial Hospital, University of Miami, Miami, USA
| | - Phillipe Abreu
- Division of Transplant Surgery, University of Colorado Anschutz Medical Campus, USA.
| |
Collapse
|
20
|
Reduzzi C, Nicolo' E, Singhal S, Venetis K, Ortega-Franco A, de Miguel-Perez D, Dipasquale A, Gouda MA, Saldanha EF, Kasi PM, Jantus-Lewintre E, Fusco N, Malapelle U, Gandara DR, Rolfo C, Serrano MJ, Cristofanilli M. Unveiling the impact of circulating tumor cells: Two decades of discovery and clinical advancements in solid tumors. Crit Rev Oncol Hematol 2024; 203:104483. [PMID: 39159706 DOI: 10.1016/j.critrevonc.2024.104483] [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: 05/21/2024] [Revised: 08/12/2024] [Accepted: 08/13/2024] [Indexed: 08/21/2024] Open
Abstract
Circulating tumor cells (CTCs) enumeration and molecular profiling hold promise in revolutionizing the management of solid tumors. Their understanding has evolved significantly over the past two decades, encompassing pivotal biological discoveries and clinical studies across various malignancies. While for some tumor types, such as breast, prostate, and colorectal cancer, CTCs are ready to enter clinical practice, for others, additional research is required. CTCs serve as versatile biomarkers, offering insights into tumor biology, metastatic progression, and treatment response. This review summarizes the latest advancements in CTC research and highlights future directions of investigation. Special attention is given to concurrent evaluations of CTCs and other circulating biomarkers, particularly circulating tumor DNA. Multi-analyte assessment holds the potential to unlock the full clinical capabilities of liquid biopsy. In conclusion, CTCs represent a transformative biomarker in precision oncology, offering extraordinary opportunities to translate scientific discoveries into tangible improvements in patient care.
Collapse
Affiliation(s)
- Carolina Reduzzi
- Department of Medicine, Weill Cornell Medicine, Englander Institute for Precision Medicine, New York Presbyterian Hospital, New York, NY 10021, USA.
| | - Eleonora Nicolo'
- Department of Medicine, Weill Cornell Medicine, Englander Institute for Precision Medicine, New York Presbyterian Hospital, New York, NY 10021, USA.
| | - Surbhi Singhal
- Division of Hematology and Oncology, Department of Medicine, University of California Davis Comprehensive Cancer Center, Sacramento, CA 95817, USA
| | - Konstantinos Venetis
- Division of Pathology, IEO European Institute of Oncology IRCCS, Milan 20141, Italy
| | - Ana Ortega-Franco
- Medical Oncology, The Christie NHS Foundation Trust, Manchester M20 4BX, United Kingdom
| | - Diego de Miguel-Perez
- Center for Thoracic Oncology, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY 10029, USA
| | - Angelo Dipasquale
- Medical Oncology and Hematology Unit, IRCCS Humanitas Research Hospital, Rozzano, Milan, Italy
| | - Mohamed A Gouda
- Department of Investigational Cancer Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Erick F Saldanha
- Division of Medical Oncology and Hematology, Princess Margaret Cancer Centre, University Health Network, University of Toronto, ON, Canada
| | - Pashtoon M Kasi
- Department of Medicine, Weill Cornell Medicine, Englander Institute for Precision Medicine, New York Presbyterian Hospital, New York, NY 10021, USA
| | - Eloisa Jantus-Lewintre
- Department of Biotechnology, Universitat Politècnica de València, Unidad Mixta TRIAL (Fundación para la Investigación del Hospital General Universitario de Valencia y Centro de Investigación Príncipe Felipe) and CIBERONC, Valencia, Spain
| | - Nicola Fusco
- Division of Pathology, IEO European Institute of Oncology IRCCS, Milan 20141, Italy; Department of Oncology and Hemato-oncology, University of Milan, Milan 20121, Italy
| | - Umberto Malapelle
- Department of Public Health, University of Naples Federico II, Napoli 80131, Italy
| | - David R Gandara
- Division of Hematology and Oncology, Department of Medicine, University of California Davis Comprehensive Cancer Center, Sacramento, CA 95817, USA
| | - Christian Rolfo
- Center for Thoracic Oncology, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY 10029, USA
| | - Maria Jose Serrano
- GENYO, Centre for Genomics and Oncological Research, Pfizer/University of Granada/Andalusian Regional Government, Liquid biopsy and Cancer Interception group, PTS Granada, Avenida de la Ilustración 114, Granada 18016, Spain; Pathological Anatomy Unit, Molecular Pathology Laboratory,Virgen de las Nieves. University Hospital, Av. Dr. Olóriz 16, Granada 18012, Spain
| | - Massimo Cristofanilli
- Department of Medicine, Weill Cornell Medicine, Englander Institute for Precision Medicine, New York Presbyterian Hospital, New York, NY 10021, USA
| |
Collapse
|
21
|
Dompé C, Chojnowska A, Ramlau R, Nowicki M, Alix-Panabières C, Budna-Tukan J. Unveiling the dynamics of circulating tumor cells in colorectal cancer: from biology to clinical applications. Front Cell Dev Biol 2024; 12:1498032. [PMID: 39539964 PMCID: PMC11557528 DOI: 10.3389/fcell.2024.1498032] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2024] [Accepted: 10/18/2024] [Indexed: 11/16/2024] Open
Abstract
This review delves into the pivotal role of circulating tumor cells (CTCs) in colorectal cancer (CRC) metastasis, focusing on their biological properties, interactions with the immune system, advanced detection techniques, and clinical implications. We explored how metastasis-competent CTCs evade immune surveillance and proliferate, utilizing cutting-edge detection and isolation technologies, such as microfluidic devices and immunological assays, to enhance sensitivity and specificity. The review highlights the significant impact of CTC interactions with immune cells on tumor progression and patient outcomes. It discusses the application of these findings in clinical settings, including non-invasive liquid biopsies for early diagnosis, prognosis, and treatment monitoring. Despite advancements, challenges remain, such as the need for standardized methods to consistently capture and analyze CTCs. Addressing these challenges through further molecular and cellular research on CTCs could lead to improved interventions and outcomes for CRC patients, underscoring the importance of unraveling the complex dynamics of CTCs in cancer progression.
Collapse
Affiliation(s)
- Claudia Dompé
- Department of Immunology, Poznan University of Medical Sciences, Poznan, Poland
- Doctoral School, Poznan University of Medical Sciences, Poznan, Poland
| | | | - Rodryg Ramlau
- Department of Oncology, Poznan University of Medical Sciences, Poznan, Poland
| | - Michal Nowicki
- Department of Histology and Embryology, Poznan University of Medical Sciences, Poznan, Poland
| | - Catherine Alix-Panabières
- Laboratory of Rare Human Circulating Cells and Liquid Biopsy (LCCRH), University Medical Centre of Montpellier, Montpellier, France
- Centre de Recherche en Ecologie et Evolution du Cancer, Maladies Infectieuses et Vecteurs: Ecologie, Génétique, Evolution et Contrôle, University of Montpellier, Centre National de la Recherche Scientifique, Institut de Recherche Pour le Dévelopement, Montpellier, France
- European Liquid Biopsy Society (ELBS), Hamburg, Germany
| | - Joanna Budna-Tukan
- Department of Histology and Embryology, Poznan University of Medical Sciences, Poznan, Poland
- Department of Anatomy and Histology, Collegium Medicum, University of Zielona Gora, Zielona Gora, Poland
| |
Collapse
|
22
|
Perelmuter VM, Grigoryeva ES, Alifanov VV, Kalinchuk AY, Andryuhova ES, Savelieva OE, Patskan IA, Bragina OD, Garbukov EY, Vostrikova MA, Zavyalova MV, Denisov EV, Cherdyntseva NV, Tashireva LA. Characterization of EpCAM-Positive and EpCAM-Negative Tumor Cells in Early-Stage Breast Cancer. Int J Mol Sci 2024; 25:11109. [PMID: 39456890 PMCID: PMC11508537 DOI: 10.3390/ijms252011109] [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: 09/07/2024] [Revised: 10/02/2024] [Accepted: 10/03/2024] [Indexed: 10/28/2024] Open
Abstract
Most studies on CTCs have focused on isolating cells that express EpCAM. In this study, we emphasize the presence of EpCAM-negative and EpCAMlow CTCs, in addition to EpCAMhigh CTCs, in early BC. We evaluated stem cell markers (CD44/CD24 and CD133) and EMT markers (N-cadherin) in each subpopulation. Our findings indicate that all stemness variants were present in both EpCAMhigh and EpCAM-negative CTCs, whereas only one variant of stemness (nonCD44+CD24-/CD133+) was observed among EpCAMlow CTCs. Nearly all EpCAMhigh CTCs were represented by CD133+ stem cells. Notably, the hybrid EMT phenotype was more prevalent among EpCAM-negative CTCs. scRNA-seq of isolated CTCs and primary tumor partially confirmed this pattern. Therefore, further investigation is imperative to elucidate the prognostic significance of EpCAM-negative and EpCAMlow CTCs.
Collapse
Affiliation(s)
- Vladimir M. Perelmuter
- The Department of General and Molecular Pathology, Cancer Research Institute, Tomsk National Research Medical Center, Russian Academy of Sciences, Tomsk 634009, Russia
| | - Evgeniya S. Grigoryeva
- The Laboratory of Molecular Therapy of Cancer, Cancer Research Institute, Tomsk National Research Medical Center, Russian Academy of Sciences, Tomsk 634009, Russia
- The Laboratory of Molecular Oncology and Immunology, Cancer Research Institute, Tomsk National Research Medical Center, Russian Academy of Sciences, Tomsk 634009, Russia
| | - Vladimir V. Alifanov
- The Department of General and Molecular Pathology, Cancer Research Institute, Tomsk National Research Medical Center, Russian Academy of Sciences, Tomsk 634009, Russia
| | - Anna Yu. Kalinchuk
- The Laboratory of Molecular Therapy of Cancer, Cancer Research Institute, Tomsk National Research Medical Center, Russian Academy of Sciences, Tomsk 634009, Russia
| | - Elena S. Andryuhova
- The Department of General and Molecular Pathology, Cancer Research Institute, Tomsk National Research Medical Center, Russian Academy of Sciences, Tomsk 634009, Russia
| | - Olga E. Savelieva
- The Department of General and Molecular Pathology, Cancer Research Institute, Tomsk National Research Medical Center, Russian Academy of Sciences, Tomsk 634009, Russia
| | - Ivan A. Patskan
- The Laboratory of Molecular Therapy of Cancer, Cancer Research Institute, Tomsk National Research Medical Center, Russian Academy of Sciences, Tomsk 634009, Russia
| | - Olga D. Bragina
- The Department of Nuclear Therapy and Diagnostics, Cancer Research Institute, Tomsk National Research Medical Center, Russian Academy of Sciences, Tomsk 634009, Russia
| | - Evgeniy Yu. Garbukov
- The Department of General Oncology, Cancer Research Institute, Tomsk National Research Medical Center, Russian Academy of Sciences, Tomsk 634009, Russia
| | - Mariya A. Vostrikova
- The Department of General Oncology, Cancer Research Institute, Tomsk National Research Medical Center, Russian Academy of Sciences, Tomsk 634009, Russia
| | - Marina V. Zavyalova
- The Department of General and Molecular Pathology, Cancer Research Institute, Tomsk National Research Medical Center, Russian Academy of Sciences, Tomsk 634009, Russia
| | - Evgeny V. Denisov
- The Laboratory of Cancer Progression Biology, Cancer Research Institute, Tomsk National Research Medical Center, Russian Academy of Sciences, Tomsk 634009, Russia
| | - Nadezhda V. Cherdyntseva
- The Laboratory of Molecular Oncology and Immunology, Cancer Research Institute, Tomsk National Research Medical Center, Russian Academy of Sciences, Tomsk 634009, Russia
| | - Liubov A. Tashireva
- The Department of General and Molecular Pathology, Cancer Research Institute, Tomsk National Research Medical Center, Russian Academy of Sciences, Tomsk 634009, Russia
- The Laboratory of Molecular Therapy of Cancer, Cancer Research Institute, Tomsk National Research Medical Center, Russian Academy of Sciences, Tomsk 634009, Russia
| |
Collapse
|
23
|
Liu G, Zhu J, Zhang P, Zhang T, Cui Z, Jiao F, Le W, Li X, Chen B. Exploring the diagnostic and prognostic significance of circulating tumor cells in stage II-IV colorectal cancer using a nano-based detection method. J Chin Med Assoc 2024; 87:945-952. [PMID: 39164812 DOI: 10.1097/jcma.0000000000001143] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 08/22/2024] Open
Abstract
BACKGROUND Colorectal cancer (CRC) is a leading cause of cancer mortality globally, underscoring the urgency for a noninvasive and effective biomarker to enhance patient prognosis. Circulating tumor cells (CTCs), a potential marker for real-time tumor monitoring, are limited in clinical utility due to the low sensitivity of existing detection methods. Previously, we introduced a novel nano-based CTCs detection method that relies on the electrical properties of cell surfaces, thus eliminating the need for specific molecular biomarkers. In this study, we used this technique to evaluate the diagnostic and prognostic value of CTCs in stage II-IV CRC. METHODS A total of 194 participants were included, consisting of 136 CRC patients and 58 healthy individuals. The peripheral blood of the participants was collected, and CTC enumeration was performed utilizing the nano-based detection method that we newly developed. The receiver operating characteristic (ROC) curve and multivariate Cox proportional-hazards analysis were used to assess the effectiveness of CTCs for diagnosing CRC and predicting patient prognosis. RESULTS The nano-based method demonstrated an ability to differentiate CRC patients from healthy individuals with a sensitivity of 84.6% and a specificity of 94.8%. Furthermore, baseline CTC levels were predictive of progression-free survival (PFS) in CRC patients, with lower levels associated with longer PFS compared to higher levels (4.5 vs 8.0 months at 15 CTCs/mL, p = 0.016; 4.4 vs 8.0 months at 20 CTCs/mL, p = 0.028). We also explored the dynamic changes in the number of CTCs after 1 to 5 cycles of chemotherapy. Patients with increasing CTC levels typically experienced disease progression (PD), while those with decreasing levels often achieved a partial response (PR) or maintained stable disease (SD). These findings suggest that the dynamic fluctuations in CTC counts are closely tied to the clinical course of the disease. CONCLUSION Our study indicates the potential of nano-based CTCs detection in diagnosing and predicting outcomes for patients with stage II-IV CRC.
Collapse
Affiliation(s)
- Gang Liu
- Department of Throatic Surgery, East Hospital of Tongji University School of Medicine, Shanghai, China
| | - Jinfeng Zhu
- Department of Oncology, Affiliated Quanzhou First Hospital of Fujian Medical University, Quanzhou, China
| | - Pengbo Zhang
- Zhihui Medical Technology (Shanghai) Co., Ltd., Shanghai, China
| | - Tingting Zhang
- Zhihui Medical Technology (Shanghai) Co., Ltd., Shanghai, China
| | - Zheng Cui
- Zhihui Medical Technology (Shanghai) Co., Ltd., Shanghai, China
| | - Fanglei Jiao
- Department of Throatic Surgery, East Hospital of Tongji University School of Medicine, Shanghai, China
| | - Wenjun Le
- Institute for Regenerative Medicine, Shanghai East Hospital, The Institute for Biomedical Engineering & Nano Science, Tongji University School of Medicine, Shanghai, China
| | - Xiaofeng Li
- Department of Oncology, Affiliated Quanzhou First Hospital of Fujian Medical University, Quanzhou, China
| | - Bingdi Chen
- Institute for Regenerative Medicine, Shanghai East Hospital, The Institute for Biomedical Engineering & Nano Science, Tongji University School of Medicine, Shanghai, China
| |
Collapse
|
24
|
Rapanotti MC, Cenci T, Scioli MG, Cugini E, Anzillotti S, Savino L, Coletta D, Di Raimondo C, Campione E, Roselli M, Bernardini S, Bianchi L, De Luca A, Ferlosio A, Orlandi A. Circulating Tumor Cells: Origin, Role, Current Applications, and Future Perspectives for Personalized Medicine. Biomedicines 2024; 12:2137. [PMID: 39335650 PMCID: PMC11429165 DOI: 10.3390/biomedicines12092137] [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: 07/08/2024] [Revised: 09/09/2024] [Accepted: 09/13/2024] [Indexed: 09/30/2024] Open
Abstract
Circulating tumor cells (CTCs) currently represent a revolutionary tool offering unique insights for the evaluation of cancer progression, metastasis, and response to therapies. Indeed, CTCs, upon detachment from primary tumors, enter the bloodstream and acquire a great potential for their use for personalized cancer management. In this review, we describe the current understanding of and advances in the clinical employment of CTCs. Although considered rare and fleeting, CTCs are now recognized as key players favoring the development of cancer metastasis and disease recurrence, particularly in malignant melanoma, lung, breast, and colorectal cancer patients. To date, the advancements in technology and the development of several successful approaches, also including immunomagnetic enrichment allow for a reliable and reproducible detection and characterization of CTCs. Those innovative methodologies improved the isolation, quantification, and characterization of CTCs from the blood of cancer patients, providing extremely useful evidence and new insights into the nature of the tumor, its epithelial/mesenchymal profile, and its potential resistance to therapy. In fact, in addition to their prognostic and predictive value, CTCs could serve as a valuable instrument for real-time monitoring of treatment response and disease recurrence, facilitating timely interventions and thus improving patient outcomes. However, despite their potential, several challenges hinder the widespread clinical utility of CTCs: (i) CTCs' rarity and heterogeneity pose technical limitations in isolation and characterization, as well as significant hurdles in their clinical implementation; (ii) it is mandatory to standardize CTC detection methods, optimize the sample processing techniques, and integrate them with existing diagnostic modalities; and (iii) the need for the development of new techniques, such as single-cell analysis platforms, to enhance the sensitivity and specificity of CTC detection, thereby facilitating their integration into routine clinical practice. In conclusion, CTCs represent a potential extraordinary tool in cancer diagnostics and therapeutics, offering unprecedented opportunities for personalized medicine and precision oncology. Moreover, their ability to provide real-time insights into tumor biology, treatment response, and disease progression underlines a great potential for their clinical application to improve patients' outcomes and advance our understanding of cancer biology.
Collapse
Affiliation(s)
- Maria Cristina Rapanotti
- Anatomic Pathology, Department of Integrated Care Processes, University of Rome Tor Vergata, Viale Oxford 81, 00133 Rome, Italy; (T.C.); (M.G.S.); (S.A.); (L.S.); (A.F.); (A.O.)
| | - Tonia Cenci
- Anatomic Pathology, Department of Integrated Care Processes, University of Rome Tor Vergata, Viale Oxford 81, 00133 Rome, Italy; (T.C.); (M.G.S.); (S.A.); (L.S.); (A.F.); (A.O.)
| | - Maria Giovanna Scioli
- Anatomic Pathology, Department of Integrated Care Processes, University of Rome Tor Vergata, Viale Oxford 81, 00133 Rome, Italy; (T.C.); (M.G.S.); (S.A.); (L.S.); (A.F.); (A.O.)
| | - Elisa Cugini
- Department of Experimental Medicine, University of Rome Tor Vergata, Via Montpellier 1, 00133 Rome, Italy; (E.C.)
| | - Silvia Anzillotti
- Anatomic Pathology, Department of Integrated Care Processes, University of Rome Tor Vergata, Viale Oxford 81, 00133 Rome, Italy; (T.C.); (M.G.S.); (S.A.); (L.S.); (A.F.); (A.O.)
| | - Luca Savino
- Anatomic Pathology, Department of Integrated Care Processes, University of Rome Tor Vergata, Viale Oxford 81, 00133 Rome, Italy; (T.C.); (M.G.S.); (S.A.); (L.S.); (A.F.); (A.O.)
| | - Deborah Coletta
- Oncology Unit, Department of Systems Medicine, University of Rome Tor Vergata, Viale Oxford 81, 00133 Rome, Italy; (D.C.); (M.R.)
| | - Cosimo Di Raimondo
- Dermatology Unit, Department of Systems Medicine, University of Rome Tor Vergata, Via Montpellier 1, 00133 Rome, Italy; (C.D.R.); (E.C.); (L.B.)
| | - Elena Campione
- Dermatology Unit, Department of Systems Medicine, University of Rome Tor Vergata, Via Montpellier 1, 00133 Rome, Italy; (C.D.R.); (E.C.); (L.B.)
| | - Mario Roselli
- Oncology Unit, Department of Systems Medicine, University of Rome Tor Vergata, Viale Oxford 81, 00133 Rome, Italy; (D.C.); (M.R.)
| | - Sergio Bernardini
- Department of Experimental Medicine, University of Rome Tor Vergata, Via Montpellier 1, 00133 Rome, Italy; (E.C.)
| | - Luca Bianchi
- Dermatology Unit, Department of Systems Medicine, University of Rome Tor Vergata, Via Montpellier 1, 00133 Rome, Italy; (C.D.R.); (E.C.); (L.B.)
| | - Anastasia De Luca
- Department of Biology, University of Rome Tor Vergata, Via della Ricerca Scientifica 1, 00133 Rome, Italy
| | - Amedeo Ferlosio
- Anatomic Pathology, Department of Integrated Care Processes, University of Rome Tor Vergata, Viale Oxford 81, 00133 Rome, Italy; (T.C.); (M.G.S.); (S.A.); (L.S.); (A.F.); (A.O.)
| | - Augusto Orlandi
- Anatomic Pathology, Department of Integrated Care Processes, University of Rome Tor Vergata, Viale Oxford 81, 00133 Rome, Italy; (T.C.); (M.G.S.); (S.A.); (L.S.); (A.F.); (A.O.)
| |
Collapse
|
25
|
Stefanes NM, Cunha-Silva ME, de Oliveira Silva L, Walter LO, Santos-Silva MC, Gartia MR. Circulating biomarkers for diagnosis and response to therapies in cancer patients. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2024; 391:1-41. [PMID: 39939074 DOI: 10.1016/bs.ircmb.2024.08.007] [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] [Indexed: 01/03/2025]
Abstract
Cancer presents a significant challenge to global health, driving worldwide concerted efforts to advance early detection, predict therapeutic response, and identify novel targeted therapies. Liquid biopsies emerge as promising avenues for discerning cancer biomarkers, offering less invasive approaches compared to conventional methods. Utilizing increasingly robust technologies, diverse bodily fluids can unveil genetic variants, epigenetic modifications, transcriptional alterations, and metabolomic signatures associated with cancer, thereby furnishing valuable insights for clinical management. This chapter intends to review the sources of cancer-related biomarkers found in circulation, prevalent techniques utilized for their identification, and the potential implications of different biomarker types on the management of cancer. Certain biomarkers currently used in clinical practice will be addressed, as well as potential biomarkers still in the study phase, and the inherent challenges in their practical implementation.
Collapse
Affiliation(s)
- Natália Marcéli Stefanes
- Department of Mechanical and Industrial Engineering, Louisiana State University, Baton Rouge, LA, United States
| | - Maria Eduarda Cunha-Silva
- Post-Graduation Program in Pharmacy, Health Science Center, Federal University of Santa Catarina, Florianópolis, SC, Brazil
| | - Lisandra de Oliveira Silva
- Post-Graduation Program in Pharmacy, Health Science Center, Federal University of Santa Catarina, Florianópolis, SC, Brazil
| | - Laura Otto Walter
- Post-Graduation Program in Pharmacy, Health Science Center, Federal University of Santa Catarina, Florianópolis, SC, Brazil
| | - Maria Cláudia Santos-Silva
- Post-Graduation Program in Pharmacy, Health Science Center, Federal University of Santa Catarina, Florianópolis, SC, Brazil
| | - Manas Ranjan Gartia
- Department of Mechanical and Industrial Engineering, Louisiana State University, Baton Rouge, LA, United States.
| |
Collapse
|
26
|
Saldanha EF. The role of liquid biopsy as a catalyst for sustained progress in precision oncology - Perspective of the young committee of the international society of liquid biopsy. THE JOURNAL OF LIQUID BIOPSY 2024; 5:100156. [PMID: 40027940 PMCID: PMC11863974 DOI: 10.1016/j.jlb.2024.100156] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 05/06/2024] [Revised: 05/09/2024] [Accepted: 05/13/2024] [Indexed: 03/05/2025]
Affiliation(s)
- Erick F. Saldanha
- Division of Medical Oncology and Hematology, Princess Margaret Cancer Centre, University Health Network, University of Toronto, ON, Canada
- Young Committee, International Society of Liquid Biopsy, Spain
| |
Collapse
|
27
|
Zhang Q, Cai Z, Gerratana L, Davis AA, D'Amico P, Chawla A, Jacob S, Zhang Y, Jiao J, Qin W, Reduzzi C, Flaum L, Shah A, Gradishar WJ. Early Evaluation of Risk Stratification and Clinical Outcomes for Patients with Advanced Breast Cancer through Combined Monitoring of Baseline Circulating Tumor Cells and DNA. Clin Cancer Res 2024; 30:3470-3480. [PMID: 38829582 DOI: 10.1158/1078-0432.ccr-24-0535] [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: 02/18/2024] [Revised: 04/19/2024] [Accepted: 05/31/2024] [Indexed: 06/05/2024]
Abstract
PURPOSE Early evaluation of tumor heterogeneity related to metastasis and outcomes is a major challenge in the management of advanced breast cancer (BCa) in the clinic. In this study, we introduced the value of baseline circulating tumor cells (CTC) and ctDNA for early differentiation of clinical stages, tumor heterogeneity, and prognosis in clinic. EXPERIMENTAL DESIGN A total of 292 patients with BCa were enrolled in this study, including 254 Stage IV and 38 Stage III patients, and examined the baseline levels of CTCs, CTC-clusters, and plasma ctDNA before initiating therapies. Outcomes including progression-free survival (PFS) and overall survival were evaluated using proportional hazards regression analysis. RESULTS The baseline CTCs, including HER2+ CTCs, in Stage IV patients were approximately 9.5 times higher than those detected in Stage III patients. Baseline CTC counts with a cutoff of 5 were significantly associated with the prognosis. Within each stage, patients with <5 CTCs had significantly longer PFS. Stage III patients with no CTCs exhibited the longest survival compared with patients with ≥1 CTC. CTC-clusters were only found in Stage IV patients, among whom 15 Stage IV patients with ≥5 CTC-clusters had the worst PFS compared with the 239 Stage IV patients with <5 CTC-clusters. Similar outcomes were observed in 28 out of 254 Stage IV patients who had at least one CTC-cluster detected, as these patients had shorter PFS compared with CTC-cluster negative group. The major differences in ctDNA mutations between patients with Stage III and Stage IV BCa were in PIK3CA and ESR1, which were associated with specific organ metastasis and worse outcomes. CONCLUSIONS Assessing the baseline levels of CTCs, CTC-clusters, and mutational ctDNA profile could reliably aid in differentiation of clinical stage and early prediction of metastasis and outcomes in advanced BCa.
Collapse
Affiliation(s)
- Qiang Zhang
- Division of Hematology/Oncology, Department of Medicine, Feinberg School of Medicine, Northwestern University, Chicago, Illinois
- Circulating Tumor Cell (CTC) Core Facility, Robert H. Lurie Comprehensive Cancer Center, Northwestern University, Chicago, Illinois
| | - Zheng Cai
- Biostatistics Department, University of Washington, Seattle, Washington
| | - Lorenzo Gerratana
- Department of Medicine, University of Udine, Udine, Italy
- Department of Medical Oncology, Centro di Riferimento Oncologico di Aviano (CRO), IRCCS, Aviano, Italy
| | - Andrew A Davis
- Division of Oncology, Department of Medicine, Washington University School of Medicine in St. Louis, St. Louis, Missouri
| | | | - Akhil Chawla
- Division of Surgical Oncology, Department of Surgery, Feinberg School of Medicine, Northwestern University, Chicago, Illinois
| | - Saya Jacob
- Division of Hematology/Oncology, Department of Medicine, Feinberg School of Medicine, Northwestern University, Chicago, Illinois
| | - Youbin Zhang
- Division of Hematology/Oncology, Department of Medicine, Feinberg School of Medicine, Northwestern University, Chicago, Illinois
- Circulating Tumor Cell (CTC) Core Facility, Robert H. Lurie Comprehensive Cancer Center, Northwestern University, Chicago, Illinois
| | - Jianhua Jiao
- Department of Urology, Xijing Hospital, Innovation Center for Tumor Immunocytology Therapy Technology, Xijing Innovation Research Institute, Fourth Military Medical University, Xi'an, China
| | - Weijun Qin
- Department of Urology, Xijing Hospital, Innovation Center for Tumor Immunocytology Therapy Technology, Xijing Innovation Research Institute, Fourth Military Medical University, Xi'an, China
| | - Carolina Reduzzi
- Division of Hematology and Medical Oncology, Weill Department of Medicine (WDOM), Cornell University, New York, New York
| | - Lisa Flaum
- Division of Hematology/Oncology, Department of Medicine, Feinberg School of Medicine, Northwestern University, Chicago, Illinois
| | - Ami Shah
- Division of Hematology/Oncology, Department of Medicine, Feinberg School of Medicine, Northwestern University, Chicago, Illinois
- Circulating Tumor Cell (CTC) Core Facility, Robert H. Lurie Comprehensive Cancer Center, Northwestern University, Chicago, Illinois
| | - William J Gradishar
- Division of Hematology/Oncology, Department of Medicine, Feinberg School of Medicine, Northwestern University, Chicago, Illinois
- Circulating Tumor Cell (CTC) Core Facility, Robert H. Lurie Comprehensive Cancer Center, Northwestern University, Chicago, Illinois
| |
Collapse
|
28
|
M Saini V, Oner E, Ward MP, Hurley S, Henderson BD, Lewis F, Finn SP, Fitzmaurice GJ, O'Leary JJ, O'Toole S, O'Driscoll L, Gately K. A comparative study of circulating tumor cell isolation and enumeration technologies in lung cancer. Mol Oncol 2024. [PMID: 39105395 DOI: 10.1002/1878-0261.13705] [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: 02/05/2024] [Revised: 06/17/2024] [Accepted: 07/19/2024] [Indexed: 08/07/2024] Open
Abstract
Circulating tumor cells (CTCs) have potential as diagnostic, prognostic, and predictive biomarkers in solid tumors. Despite Food and Drug Administration (FDA) approval of CTC devices in various cancers, the rarity and heterogeneity of CTCs in lung cancer make them technically challenging to isolate and analyze, hindering their clinical integration. Establishing a consensus through comparative analysis of different CTC systems is warranted. This study aimed to evaluate seven different CTC enrichment methods across five technologies using a standardized spike-in protocol: the CellMag™ (EpCAM-dependent enrichment), EasySep™ and RosetteSep™ (blood cell depletion), and the Parsortix® PR1 and the new design Parsortix® Prototype (PP) (size- and deformability-based enrichment). The Parsortix® systems were also evaluated for any differences in recovery rates between cell harvest versus in-cassette staining. Healthy donor blood (5 mL) was spiked with 100 fluorescently labeled EpCAMhigh H1975 cells, processed through each system, and the isolation efficiency was calculated. The CellMag™ had the highest recovery rate (70 ± 14%), followed by Parsortix® PR1 in-cassette staining, while the EasySep™ had the lowest recovery (18 ± 8%). Additional spike-in experiments were performed with EpCAMmoderate A549 and EpCAMlow H1299 cells using the CellMag™ and Parsortix® PR1 in-cassette staining. The recovery rate of CellMag™ significantly reduced to 35 ± 14% with A549 cells and 1 ± 1% with H1299 cells. However, the Parsortix® PR1 in-cassette staining showed cell phenotype-independent and consistent recovery rates among all lung cancer cell lines: H1975 (49 ± 2%), A549 (47 ± 10%), and H1299 (52 ± 10%). Furthermore, we demonstrated that the Parsortix® PR1 in-cassette staining method is capable of isolating heterogeneous single CTCs and cell clusters from patient samples. The Parsortix® PR1 in-cassette staining, capable of isolating different phenotypes of CTCs as either single cells or cell clusters with consistent recovery rates, is considered optimal for CTC enrichment for lung cancer, albeit needing further optimization and validation.
Collapse
Affiliation(s)
- Volga M Saini
- Thoracic Oncology Research Group, Trinity Translational Medicine Institute, St James's Hospital, Dublin, Ireland
- Department of Clinical Medicine, School of Medicine, Trinity College Dublin, Ireland
- Trinity St. James's Cancer Institute, Trinity College Dublin, Ireland
| | - Ezgi Oner
- Thoracic Oncology Research Group, Trinity Translational Medicine Institute, St James's Hospital, Dublin, Ireland
- Department of Clinical Medicine, School of Medicine, Trinity College Dublin, Ireland
- Trinity St. James's Cancer Institute, Trinity College Dublin, Ireland
| | - Mark P Ward
- Trinity St. James's Cancer Institute, Trinity College Dublin, Ireland
- Department of Obstetrics and Gynaecology, School of Medicine, Trinity College Dublin, Ireland
- Department of Histopathology and Morbid Anatomy, School of Medicine, Trinity College Dublin, Ireland
| | - Sinead Hurley
- Thoracic Oncology Research Group, Trinity Translational Medicine Institute, St James's Hospital, Dublin, Ireland
- Department of Clinical Medicine, School of Medicine, Trinity College Dublin, Ireland
- Trinity St. James's Cancer Institute, Trinity College Dublin, Ireland
- Department of Obstetrics and Gynaecology, School of Medicine, Trinity College Dublin, Ireland
| | - Brian David Henderson
- Trinity St. James's Cancer Institute, Trinity College Dublin, Ireland
- Department of Obstetrics and Gynaecology, School of Medicine, Trinity College Dublin, Ireland
- Department of Histopathology and Morbid Anatomy, School of Medicine, Trinity College Dublin, Ireland
| | - Faye Lewis
- Trinity St. James's Cancer Institute, Trinity College Dublin, Ireland
- Department of Obstetrics and Gynaecology, School of Medicine, Trinity College Dublin, Ireland
- Department of Histopathology and Morbid Anatomy, School of Medicine, Trinity College Dublin, Ireland
| | - Stephen P Finn
- Thoracic Oncology Research Group, Trinity Translational Medicine Institute, St James's Hospital, Dublin, Ireland
- Trinity St. James's Cancer Institute, Trinity College Dublin, Ireland
- Department of Histopathology and Morbid Anatomy, School of Medicine, Trinity College Dublin, Ireland
| | | | - John J O'Leary
- Trinity St. James's Cancer Institute, Trinity College Dublin, Ireland
- Department of Histopathology and Morbid Anatomy, School of Medicine, Trinity College Dublin, Ireland
| | - Sharon O'Toole
- Trinity St. James's Cancer Institute, Trinity College Dublin, Ireland
- Department of Obstetrics and Gynaecology, School of Medicine, Trinity College Dublin, Ireland
- Department of Histopathology and Morbid Anatomy, School of Medicine, Trinity College Dublin, Ireland
| | - Lorraine O'Driscoll
- Trinity St. James's Cancer Institute, Trinity College Dublin, Ireland
- School of Pharmacy and Pharmaceutical Sciences, Trinity College Dublin, Ireland
- Trinity Biomedical Sciences Institute, Trinity College Dublin, Ireland
| | - Kathy Gately
- Thoracic Oncology Research Group, Trinity Translational Medicine Institute, St James's Hospital, Dublin, Ireland
- Department of Clinical Medicine, School of Medicine, Trinity College Dublin, Ireland
- Trinity St. James's Cancer Institute, Trinity College Dublin, Ireland
| |
Collapse
|
29
|
Varol A, Klauck SM, Dantzer F, Efferth T. Enhancing cisplatin drug sensitivity through PARP3 inhibition: The influence on PDGF and G-coupled signal pathways in cancer. Chem Biol Interact 2024; 398:111094. [PMID: 38830565 DOI: 10.1016/j.cbi.2024.111094] [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: 02/16/2024] [Revised: 05/07/2024] [Accepted: 05/31/2024] [Indexed: 06/05/2024]
Abstract
Drug resistance poses a significant challenge in cancer treatment despite the clinical efficacy of cisplatin. Identifying and targeting biomarkers open new ways to improve therapeutic outcomes. In this study, comprehensive bioinformatic analyses were employed, including a comparative analysis of multiple datasets, to evaluate overall survival and mutation hotspots in 27 base excision repair (BER) genes of more than 7,500 tumors across 23 cancer types. By using various parameters influencing patient survival, revealing that the overexpression of 15 distinct BER genes, particularly PARP3, NEIL3, and TDG, consistently correlated with poorer survival across multiple factors such as race, gender, and metastasis. Single nucleotide polymorphism (SNP) analyses within protein-coding regions highlighted the potential deleterious effects of mutations on protein structure and function. The investigation of mutation hotspots in BER proteins identified PARP3 due to its high mutation frequency. Moving from bioinformatics to wet lab experiments, cytotoxic experiments demonstrated that the absence of PARP3 by CRISPR/Cas9-mediated knockdown in MDA-MB-231 breast cancer cells increased drug activity towards cisplatin, carboplatin, and doxorubicin. Pathway analyses indicated the impact of PARP3 absence on the platelet-derived growth factor (PDGF) and G-coupled signal pathways on cisplatin exposure. PDGF, a critical regulator of various cellular functions, was downregulated in the absence of PARP3, suggesting a role in cancer progression. Moreover, the influence of PARP3 knockdown on G protein-coupled receptors (GPCRs) affects their function in the presence of cisplatin. In conclusion, the study demonstrated a synthetic lethal interaction between GPCRs, PDGF signaling pathways, and PARP3 gene silencing. PARP3 emerged as a promising target.
Collapse
Affiliation(s)
- Ayşegül Varol
- Department of Pharmaceutical Biology, Institute of Pharmaceutical and Biomedical Sciences, Johannes Gutenberg University-Mainz, 55128, Mainz, Germany
| | - Sabine M Klauck
- Division of Cancer Genome Research, German Cancer Research Center (DKFZ) Heidelberg, National Center for Tumor Diseases (NCT), NCT Heidelberg, a Partnership between DKFZ and University Hospital Heidelberg, 69120, Heidelberg, Germany
| | - Françoise Dantzer
- Poly(ADP-ribosyl)ation and Genome Integrity, Laboratoire d'Excellence Medalis, UMR7242, Centre Nationale de la Recherche Scientifique/Université de Strasbourg, Institut de Recherche de l'Ecole de Biotechnologie de Strasbourg, 300 bld. S. Brant, CS10413, 67412, Illkirch, France
| | - Thomas Efferth
- Department of Pharmaceutical Biology, Institute of Pharmaceutical and Biomedical Sciences, Johannes Gutenberg University-Mainz, 55128, Mainz, Germany.
| |
Collapse
|
30
|
Løppke C, Jørgensen AM, Sand NT, Klitgaard RB, Daugaard G, Agerbæk MØ. Combined microfluidic enrichment and staining workflow for single-cell analysis of circulating tumor cells in metastatic prostate cancer patients. Sci Rep 2024; 14:17501. [PMID: 39080445 PMCID: PMC11289449 DOI: 10.1038/s41598-024-68336-4] [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: 12/13/2023] [Accepted: 07/22/2024] [Indexed: 08/02/2024] Open
Abstract
Circulating tumor cells (CTCs) are precursors of cancer in the blood and provide an attractive source for dynamic monitoring of disease progression and tumor heterogeneity. However, the scarcity of CTCs in the bloodstream has limited their use in clinical practice. In this study, we present a workflow for easy detection of CTCs by cytokeratin staining using the FDA-cleared Parsortix device for size-based microfluidic enrichment. To minimize sample handling, the isolated cells are stained inside the separation cassette and harvested for subsequent single cell isolation and whole genome copy-number analysis. We validated the workflow on a panel of four prostate cancer cell lines spiked into healthy donor blood collected in CellRescue or EDTA tubes, resulting in mean recoveries of 42% (16-69%). Furthermore, we evaluated the clinical utility in a cohort of 12 metastatic prostate cancer patients and found CTCs in 67% of patients ranging from 0 to 1172 CTCs in 10 mL blood. Additionally, we isolated single patient-derived CTCs and identified genomic aberrations associated with treatment response and clinical outcome. Thus, this workflow provides a readily scalable strategy for analysis of single CTCs, applicable for use in monitoring studies to identify genomic variations important for guiding clinical therapy decision.
Collapse
Affiliation(s)
- Caroline Løppke
- Centre for Translational Medicine and Parasitology at Department for Immunology and Microbiology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Amalie M Jørgensen
- Centre for Translational Medicine and Parasitology at Department for Immunology and Microbiology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Nicolai T Sand
- Centre for Translational Medicine and Parasitology at Department for Immunology and Microbiology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Rasmus B Klitgaard
- Flow Cytometry and Single Cell Core Facility, Department for Immunology and Microbiology, University of Copenhagen, Copenhagen, Denmark
| | - Gedske Daugaard
- Department of Oncology, Centre for Cancer and Organ Diseases, Copenhagen University Hospital Rigshospitalet, Copenhagen, Denmark
| | - Mette Ø Agerbæk
- Centre for Translational Medicine and Parasitology at Department for Immunology and Microbiology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark.
| |
Collapse
|
31
|
Tao XY, Li QQ, Zeng Y. Clinical application of liquid biopsy in colorectal cancer: detection, prediction, and treatment monitoring. Mol Cancer 2024; 23:145. [PMID: 39014366 PMCID: PMC11250976 DOI: 10.1186/s12943-024-02063-2] [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: 03/08/2024] [Accepted: 07/09/2024] [Indexed: 07/18/2024] Open
Abstract
Colorectal cancer (CRC) is one of the most prevalent malignancies affecting the gastrointestinal tract and is ranked third among cancers with the highest incidence and second-highest mortality rate worldwide. CRC exhibits a slow progression providing a wide treatment window. The currently employed CRC screening methods have shown great potential to prevent CRC and reduce CRC-related morbidity and mortality. The diagnosis of CRC is achieved by colonoscopy and tissue biopsy, with studies showing that liquid biopsy is more effective in detecting and diagnosing early CRC patients. Increasing number of studies have shown that the tumor components shed into circulating blood can be detected in liquid form, and can be applied in the clinical management of CRC. Analysis of circulating tumor cells (CTCs), circulating tumor DNA (ctDNA), or tumor-associated platelets (TEPs) in the blood can be used for early screening and diagnosis of CRC, aid tumor staging, treatment response monitoring, and prediction of CRC recurrence and metastasis in a minimally invasive manner. This chapter provides an updated review of CTCs, ctDNA, and TEPs as novel biomarkers for CRC, highlighting their strengths and limitations.
Collapse
Affiliation(s)
- Xiang-Yuan Tao
- Translational Medicine Center, Hunan Cancer Hospital, The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, China
- School of Pharmacy, University of South China, Hengyang, China
| | - Qian-Qian Li
- Translational Medicine Center, Hunan Cancer Hospital, The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, China
- School of Pharmacy, University of South China, Hengyang, China
| | - Yong Zeng
- Translational Medicine Center, Hunan Cancer Hospital, The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, China.
- School of Pharmacy, University of South China, Hengyang, China.
| |
Collapse
|
32
|
Reginacova K, Pospisilova E, Kubecova M, Svobodova P, Bobek V, Kolostova K. Circulating tumor cells in patients with cervical cancer undergoing chemoradiotherapy combined with brachytherapy. Am J Cancer Res 2024; 14:3614-3625. [PMID: 39113856 PMCID: PMC11301293 DOI: 10.62347/qixj7103] [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: 08/27/2023] [Accepted: 05/11/2024] [Indexed: 08/10/2024] Open
Abstract
Circulating tumor cells (CTCs) have significant potential to become an important tool for monitoring the effects of treatment in solid tumors. The present study reports the occurance of CTCs in cervical cancer (CC) patients during radical chemoradiotherapy (CRT), including brachytherapy (BRT), and during the follow-up period. Patients diagnosed with CC treated with radical CRT were included in the study (n=30). A total of 167 CTC-tests (MetaCell®) were provided at predefined testing time points during the study follow-up (e.g., before CRT, after CRT, every three months of follow-up). In parallel with CTC-testing, SCC-Ag were measured to compare their predictive values during treatment. CTCs were present in 96% (25/26) of patients at the time of diagnosis and in 61% (14/23) after treatment. Patients who relapsed during the 36-month follow-up (n=10) showed an elevation in pre-treatment CTC- numbers, similarly there was a significant increase in pre-treatment SCC-Ag. As next, an increased number of CTCs was observed approximately 12 weeks before relapse was diagnosed by standard imaging modalities (MRI, US, PET-CT) in 3 of 4 patients. In addition to standardized vital cytomorphology of enriched CTCs, quantitative PCR (qPCR) was used to inform the nature of CTCs before treatment. Analysis revealed increased SOX2 and POUSF expression in CTCs in the group of patients with recurrence (P < 0.02). Disease aggressiveness may be related to increased expression of stem cell markers, as found in samples from relapsed patients. CTCs may be an aid to assess tumor burden and disease aggressiveness. An increase in CTCs precedes an increase in SCC-Ag and confirmation of relapse by imaging, as shown in our study.
Collapse
Affiliation(s)
- Klaudia Reginacova
- Department of Oncology, The Third Faculty of Medicine, Charles University Prague and Faculty Hospital Kralovske VinohradySrobarova 50, 100 34 Prague 10, Czech Republic
| | - Eliska Pospisilova
- Laboratory of Personalized Medicine, Oncology Clinic, Faculty Hospital Kralovske VinohradySrobarova 50, 100 34 Prague 10, Czech Republic
| | - Martina Kubecova
- Department of Oncology, The Third Faculty of Medicine, Charles University Prague and Faculty Hospital Kralovske VinohradySrobarova 50, 100 34 Prague 10, Czech Republic
| | - Pavla Svobodova
- Department of Gynaecology, Military University Hospital and The Third Faculty of MedicineU Vojenske Nemocnice 1200, 169 02 Prague 6, Czech Republic
| | - Vladimir Bobek
- Laboratory of Personalized Medicine, Oncology Clinic, Faculty Hospital Kralovske VinohradySrobarova 50, 100 34 Prague 10, Czech Republic
- Department of Gynaecology, Military University Hospital and The Third Faculty of MedicineU Vojenske Nemocnice 1200, 169 02 Prague 6, Czech Republic
- Department of Thoracic Surgery, Krajská zdravotní a.s. Hospital41100 Ústí nad Labem, Czech Republic
- 3 Department of Surgery, 1 Faculty of Medicine Charles UniversityV Uvalu 84, 150 06 Prague 5, Czech Republic
- Department of Thoracic Surgery, Lower Silesian Oncology, Pulmonology and Hematology Center and Technical University WroclawPlac Ludwika Hirszfelda 12, 534 13 Wrocław, Poland
| | - Katarina Kolostova
- Laboratory of Personalized Medicine, Oncology Clinic, Faculty Hospital Kralovske VinohradySrobarova 50, 100 34 Prague 10, Czech Republic
- Department of Gynaecology, Military University Hospital and The Third Faculty of MedicineU Vojenske Nemocnice 1200, 169 02 Prague 6, Czech Republic
| |
Collapse
|
33
|
Frühling P, Moberg L, Ghanipour L, Birgisson H, Graf W, Ericsson C, Cashin PH. Clinical Significance of Circulating Tumor Cells in Epithelial Appendiceal Neoplasms with Peritoneal Metastases. Cancers (Basel) 2024; 16:2441. [PMID: 39001503 PMCID: PMC11240500 DOI: 10.3390/cancers16132441] [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: 06/12/2024] [Revised: 06/27/2024] [Accepted: 06/28/2024] [Indexed: 07/16/2024] Open
Abstract
Appendiceal tumors are uncommon and, at times, discovered incidentally during histological examination. The histopathological classification of the disease is complex and has generated some controversy. The analysis of circulating tumor cells can be used for the early detection of metastatic potential. The aim of the present study was to examine the prognostic value of circulating tumor cells in patients with appendiceal tumors and peritoneal metastases. To our knowledge, this is the first study to examine CTCs in appendiceal tumors. We performed a prospective cohort study of consecutive patients treated with cytoreductive surgery and hyperthermic intraperitoneal chemotherapy between 2015 and 2019 at a HIPEC referral center. In total, 31 patients were included in the analysis, and circulating tumor cells were detected in 15 patients (48%). CTC positivity was not associated with overall or recurrence-free survival, nor was it correlated with PCI score or histopathological grading. Surprisingly, however, CTCs were found in almost half the patients. The presence or quantities of these cells did not, on their own, predict systemic metastatic potential during the observed time, and they did not appear to significantly correlate with the oncological outcomes recorded.
Collapse
Affiliation(s)
- Petter Frühling
- Uppsala Sweden and Department of Surgery, Institution of Surgical Sciences, Uppsala University, Akademiska Sjukhuset, 751 85 Uppsala, Sweden
| | - Louice Moberg
- Uppsala Sweden and Department of Surgery, Institution of Surgical Sciences, Uppsala University, Akademiska Sjukhuset, 751 85 Uppsala, Sweden
| | - Lana Ghanipour
- Uppsala Sweden and Department of Surgery, Institution of Surgical Sciences, Uppsala University, Akademiska Sjukhuset, 751 85 Uppsala, Sweden
| | - Helgi Birgisson
- Uppsala Sweden and Department of Surgery, Institution of Surgical Sciences, Uppsala University, Akademiska Sjukhuset, 751 85 Uppsala, Sweden
| | - Wilhelm Graf
- Uppsala Sweden and Department of Surgery, Institution of Surgical Sciences, Uppsala University, Akademiska Sjukhuset, 751 85 Uppsala, Sweden
| | - Christer Ericsson
- iCellate Medical AB, KI Science Park, Industrivägen 1, 171 48 Solna, Sweden
- Department of Microbiology, Tumor and Cell Biology (MTC), Biomedicum 8 C, Karolinska Institute, 171 77 Stockholm, Sweden
| | - Peter H Cashin
- Uppsala Sweden and Department of Surgery, Institution of Surgical Sciences, Uppsala University, Akademiska Sjukhuset, 751 85 Uppsala, Sweden
| |
Collapse
|
34
|
Galletti G, Halima A, Gjyrezi A, Zhang J, Zimmerman B, Worroll D, Kallergi G, Barreja R, Ocean A, Saxena A, McGraw TE, Nanus DM, Elemento O, Altorki NK, Tagawa ST, Giannakakou P. Transferrin receptor-based circulating tumor cell enrichment provides a snapshot of the molecular landscape of solid tumors and correlates with clinical outcomes. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2024.06.16.24309003. [PMID: 38947080 PMCID: PMC11213041 DOI: 10.1101/2024.06.16.24309003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/02/2024]
Abstract
Circulating tumor cells (CTCs) captured from the bloodstream of patients with solid tumors have the potential to accelerate precision oncology by providing insight into tumor biology, disease progression and response to treatment. However, their potential is hampered by the lack of standardized CTC enrichment platforms across tumor types. EpCAM-based CTC enrichment, the most commonly used platform, is limited by EpCAM downregulation during metastasis and the low EpCAM expression in certain tumor types, including the highly prevalent and lethal NSCLC. In this study we demonstrate that Transferrin Receptor (TfR) is a selective, efficient biomarker for CTC identification and capture in patients with prostate, pancreatic and NSCLC. TfR identifies significantly higher CTC counts than EpCAM, and TfR + -CTC enumeration correlates with disease progression in metastatic prostate and pancreatic cancers, and overall survival and osimetrinib-resistance in non-small cell lung cancer (NSCLC). Profiling of TfR + -CTCs provides a snapshot of the molecular landscape of each respective tumor type and identifies potential mechanisms underlying treatment response to EGFR TKi and immune checkpoint inhibitors in NSCLC. One sentence summary Transferrin Receptor identifies circulating tumor cells in solid tumors.
Collapse
|
35
|
Zhang YW, Gvozdenovic A, Aceto N. A Molecular Voyage: Multiomics Insights into Circulating Tumor Cells. Cancer Discov 2024; 14:920-933. [PMID: 38581442 DOI: 10.1158/2159-8290.cd-24-0218] [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: 02/14/2024] [Revised: 03/08/2024] [Accepted: 03/13/2024] [Indexed: 04/08/2024]
Abstract
Circulating tumor cells (CTCs) play a pivotal role in metastasis, the leading cause of cancer-associated death. Recent improvements of CTC isolation tools, coupled with a steady development of multiomics technologies at single-cell resolution, have enabled an extensive exploration of CTC biology, unlocking insights into their molecular profiles. A detailed molecular portrait requires CTC interrogation across various levels encompassing genomic, epigenetic, transcriptomic, proteomic and metabolic features. Here, we review how state-of-the-art multiomics applied to CTCs are shedding light on how cancer spreads. Further, we highlight the potential implications of CTC profiling for clinical applications aimed at enhancing cancer diagnosis and treatment. SIGNIFICANCE Exploring the complexity of cancer progression through cutting-edge multiomics studies holds the promise of uncovering novel aspects of cancer biology and identifying therapeutic vulnerabilities to suppress metastasis.
Collapse
Affiliation(s)
- Yu Wei Zhang
- Department of Biology, Institute of Molecular Health Sciences, Swiss Federal Institute of Technology Zurich (ETH Zurich), Zurich, Switzerland
| | - Ana Gvozdenovic
- Department of Biology, Institute of Molecular Health Sciences, Swiss Federal Institute of Technology Zurich (ETH Zurich), Zurich, Switzerland
| | - Nicola Aceto
- Department of Biology, Institute of Molecular Health Sciences, Swiss Federal Institute of Technology Zurich (ETH Zurich), Zurich, Switzerland
| |
Collapse
|
36
|
Xiao D, Xiong M, Wang X, Lyu M, Sun H, Cui Y, Chen C, Jiang Z, Sun F. Regulation of the Function and Expression of EpCAM. Biomedicines 2024; 12:1129. [PMID: 38791091 PMCID: PMC11117676 DOI: 10.3390/biomedicines12051129] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2024] [Revised: 05/11/2024] [Accepted: 05/14/2024] [Indexed: 05/26/2024] Open
Abstract
The epithelial cell adhesion molecule (EpCAM) is a single transmembrane protein on the cell surface. Given its strong expression on epithelial cells and epithelial cell-derived tumors, EpCAM has been identified as a biomarker for circulating tumor cells (CTCs) and exosomes and a target for cancer therapy. As a cell adhesion molecule, EpCAM has a crystal structure that indicates that it forms a cis-dimer first and then probably a trans-tetramer to mediate intercellular adhesion. Through regulated intramembrane proteolysis (RIP), EpCAM and its proteolytic fragments are also able to regulate multiple signaling pathways, Wnt signaling in particular. Although great progress has been made, increasingly more findings have revealed the context-specific expression and function patterns of EpCAM and their regulation processes, which necessitates further studies to determine the structure, function, and expression of EpCAM under both physiological and pathological conditions, broadening its application in basic and translational cancer research.
Collapse
Affiliation(s)
- Di Xiao
- College of Life Sciences and Health, Wuhan University of Science and Technology, Wuhan 430081, China; (D.X.); (M.X.); (X.W.); (M.L.); (H.S.); (Y.C.)
- Institute of Biology and Medicine, Wuhan University of Science and Technology, Wuhan 430081, China
| | - Mingrui Xiong
- College of Life Sciences and Health, Wuhan University of Science and Technology, Wuhan 430081, China; (D.X.); (M.X.); (X.W.); (M.L.); (H.S.); (Y.C.)
- Institute of Biology and Medicine, Wuhan University of Science and Technology, Wuhan 430081, China
| | - Xin Wang
- College of Life Sciences and Health, Wuhan University of Science and Technology, Wuhan 430081, China; (D.X.); (M.X.); (X.W.); (M.L.); (H.S.); (Y.C.)
- Institute of Biology and Medicine, Wuhan University of Science and Technology, Wuhan 430081, China
| | - Mengqing Lyu
- College of Life Sciences and Health, Wuhan University of Science and Technology, Wuhan 430081, China; (D.X.); (M.X.); (X.W.); (M.L.); (H.S.); (Y.C.)
- Institute of Biology and Medicine, Wuhan University of Science and Technology, Wuhan 430081, China
| | - Hanxiang Sun
- College of Life Sciences and Health, Wuhan University of Science and Technology, Wuhan 430081, China; (D.X.); (M.X.); (X.W.); (M.L.); (H.S.); (Y.C.)
- Institute of Biology and Medicine, Wuhan University of Science and Technology, Wuhan 430081, China
| | - Yeting Cui
- College of Life Sciences and Health, Wuhan University of Science and Technology, Wuhan 430081, China; (D.X.); (M.X.); (X.W.); (M.L.); (H.S.); (Y.C.)
- Institute of Biology and Medicine, Wuhan University of Science and Technology, Wuhan 430081, China
| | - Chen Chen
- Tumor Precision Diagnosis and Treatment Technology and Translational Medicine, Hubei Engineering Research Center, Zhongnan Hospital of Wuhan University, Wuhan 430071, China;
| | - Ziyu Jiang
- Tumor Precision Diagnosis and Treatment Technology and Translational Medicine, Hubei Engineering Research Center, Zhongnan Hospital of Wuhan University, Wuhan 430071, China;
| | - Fan Sun
- College of Life Sciences and Health, Wuhan University of Science and Technology, Wuhan 430081, China; (D.X.); (M.X.); (X.W.); (M.L.); (H.S.); (Y.C.)
- Institute of Biology and Medicine, Wuhan University of Science and Technology, Wuhan 430081, China
| |
Collapse
|
37
|
N C, Jain A, C S, Shreevatsa B, Rajendrasozhan S, Dharmashekar C, Suresh KP, Patil SS, Singh P, Vishwanath P, Srinivasa C, Kollur SP, Shivamallu C. Progression-free survival estimation of docetaxel-based second-line treatment for advanced non-small cell lung cancer: a pooled analysis from 18 randomized control trials. Front Oncol 2024; 14:1298786. [PMID: 38807763 PMCID: PMC11130461 DOI: 10.3389/fonc.2024.1298786] [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: 09/22/2023] [Accepted: 04/02/2024] [Indexed: 05/30/2024] Open
Abstract
Background Lung cancer is the foremost cause of cancer-related death globally, with non-small cell lung cancer (NSCLC) accounting for 85-90% of cases. Targeted therapy is the most essential therapeutic option for NSCLC, other common treatments include radiation therapy, surgery, chemotherapy, and immunotherapy. Objective Our study objective was to estimate whether progression-free survival (PFS) is an outcome of NSCLC extracted from 18 randomized control trials (RCTs) with docetaxel as experimental group and antineoplastic agent, kinase inhibitor, and monoclonal antibodies as a control group. Methods We selected relevant studies published between 2011 and 2022 using Google Scholar, PubMed, Scopus, Science Direct, and Cochrane Library. Advanced NSCLC, chemotherapy, RCT, docetaxel, and second-line treatment were the terms included in the search. A total of 9738 patients were evaluated from the 18 identified studies. We used the meta package of R Studio to perform the meta-analysis. Graphical funnel plots were used to evaluate publication bias visually. Results Patients who underwent docetaxel-based therapy had a considerably longer PFS than those who got antineoplastic agents, kinase inhibitors, or monoclonal antibodies-based treatment. Patients in the standard treatment arm had a slightly longer PFS than those in the experimental therapy arm in the overall meta-analysis. Conclusion Docetaxel outperformed monoclonal antibodies, antineoplastic agents, and kinase inhibitors in the second-line therapy of advanced NSCLC since PFS was extensively utilized.
Collapse
Affiliation(s)
- Chaithra N
- Division of Medical Statistics, Life Sciences and Natural Sciences Departments, JSS Academy of Higher Education and Research, Mysuru, Karnataka, India
| | - Anisha Jain
- Department of Microbiology, JSS Academy of Higher Education and Research, Mysuru, Karnataka, India
| | - Sahana C
- Division of Medical Statistics, Life Sciences and Natural Sciences Departments, JSS Academy of Higher Education and Research, Mysuru, Karnataka, India
| | - Bhargav Shreevatsa
- Department of Biotechnology and Bioinformatics, JSS Academy of Higher Education and Research, Mysuru, Karnataka, India
- Pathology, Microbiology and Immunology Department, School of Medicine, University of South Carolina, Columbia, SC, United States
| | | | - Chandan Dharmashekar
- Department of Biotechnology and Bioinformatics, JSS Academy of Higher Education and Research, Mysuru, Karnataka, India
| | | | - Sharanagouda S. Patil
- ICAR-National Institute of Veterinary Epidemiology and Disease Informatics, Bengaluru, Karnataka, India
| | - Pranav Singh
- Department of Medicine, Kasturba Medical College, Manipal Academy of Higher Education, Udupi, Karnataka, India
| | - Prashant Vishwanath
- Center of Excellence in Molecular Biology and Regenerative Medicine, Department of Biochemistry, JSS Medical College, JSS Academy of Higher Education and Research, Mysore, India
| | | | - Shiva Prasad Kollur
- School of Physical Sciences, Amrita Vishwa Vidyapeetham, Mysuru, Karnataka, India
| | - Chandan Shivamallu
- Department of Biotechnology and Bioinformatics, JSS Academy of Higher Education and Research, Mysuru, Karnataka, India
| |
Collapse
|
38
|
Heussner RT, Whalen RM, Anderson A, Theison H, Baik J, Gibbs S, Wong MH, Chang YH. Quantitative image analysis pipeline for detecting circulating hybrid cells in immunofluorescence images with human-level accuracy. Cytometry A 2024; 105:345-355. [PMID: 38385578 PMCID: PMC11217923 DOI: 10.1002/cyto.a.24826] [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: 09/14/2023] [Revised: 01/10/2024] [Accepted: 01/24/2024] [Indexed: 02/23/2024]
Abstract
Circulating hybrid cells (CHCs) are a newly discovered, tumor-derived cell population found in the peripheral blood of cancer patients and are thought to contribute to tumor metastasis. However, identifying CHCs by immunofluorescence (IF) imaging of patient peripheral blood mononuclear cells (PBMCs) is a time-consuming and subjective process that currently relies on manual annotation by laboratory technicians. Additionally, while IF is relatively easy to apply to tissue sections, its application to PBMC smears presents challenges due to the presence of biological and technical artifacts. To address these challenges, we present a robust image analysis pipeline to automate the detection and analysis of CHCs in IF images. The pipeline incorporates quality control to optimize specimen preparation protocols and remove unwanted artifacts, leverages a β-variational autoencoder (VAE) to learn meaningful latent representations of single-cell images, and employs a support vector machine (SVM) classifier to achieve human-level CHC detection. We created a rigorously labeled IF CHC data set including nine patients and two disease sites with the assistance of 10 annotators to evaluate the pipeline. We examined annotator variation and bias in CHC detection and provided guidelines to optimize the accuracy of CHC annotation. We found that all annotators agreed on CHC identification for only 65% of the cells in the data set and had a tendency to underestimate CHC counts for regions of interest (ROIs) containing relatively large amounts of cells (>50,000) when using the conventional enumeration method. On the other hand, our proposed approach is unbiased to ROI size. The SVM classifier trained on the β-VAE embeddings achieved an F1 score of 0.80, matching the average performance of human annotators. Our pipeline enables researchers to explore the role of CHCs in cancer progression and assess their potential as a clinical biomarker for metastasis. Further, we demonstrate that the pipeline can identify discrete cellular phenotypes among PBMCs, highlighting its utility beyond CHCs.
Collapse
Affiliation(s)
- Robert T. Heussner
- Department of Biomedical Engineering, Oregon Health & Science University, Portland, Oregon, USA
| | - Riley M. Whalen
- Department of Cell, Developmental and Cancer Biology, Oregon Health & Science University, Portland, Oregon, USA
| | - Ashley Anderson
- Department of Cell, Developmental and Cancer Biology, Oregon Health & Science University, Portland, Oregon, USA
| | - Heather Theison
- Department of Cell, Developmental and Cancer Biology, Oregon Health & Science University, Portland, Oregon, USA
| | - Joseph Baik
- Department of Biomedical Engineering, Oregon Health & Science University, Portland, Oregon, USA
| | - Summer Gibbs
- Department of Cell, Developmental and Cancer Biology, Oregon Health & Science University, Portland, Oregon, USA
- Knight Cancer Institute, Oregon Health & Science University, Portland, Oregon, USA
| | - Melissa H. Wong
- Department of Cell, Developmental and Cancer Biology, Oregon Health & Science University, Portland, Oregon, USA
- Knight Cancer Institute, Oregon Health & Science University, Portland, Oregon, USA
| | - Young Hwan Chang
- Department of Biomedical Engineering, Oregon Health & Science University, Portland, Oregon, USA
- Knight Cancer Institute, Oregon Health & Science University, Portland, Oregon, USA
| |
Collapse
|
39
|
Libert DM, Zhu Y, Wang A, Allard GM, Cheng-Yi Lowe A. Detection of effusion tumor cells under different storage and processing conditions. Cancer Cytopathol 2024; 132:297-308. [PMID: 38373107 DOI: 10.1002/cncy.22803] [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: 01/12/2024] [Accepted: 01/26/2024] [Indexed: 02/21/2024]
Abstract
BACKGROUND Circulating tumor cells (CTCs) shed into blood provide prognostic and/or predictive information. Previously, the authors established an assay to detect carcinoma cells from pleural fluid, termed effusion tumor cells (ETCs), by employing an immunofluorescence-based CTC-identification platform (RareCyte) on air-dried unstained ThinPrep (TP) slides. To facilitate clinical integration, they evaluated different slide processing and storage conditions, hypothesizing that alternative comparable conditions for ETC detection exist. METHODS The authors enumerated ETCs on RareCyte, using morphology and mean fluorescence intensity (MFI) cutoffs of >100 arbitrary units (a.u.) for epithelial cellular adhesion molecule (EpCAM) and <100 a.u. for CD45. They analyzed malignant pleural fluid from three patients under seven processing and/or staining conditions, three patients after short-term storage under three conditions, and seven samples following long-term storage at -80°C. MFI values of 4',6-diamidino-2-phenylindol, cytokeratin, CD45, and EpCAM were compared. RESULTS ETCs were detected in all conditions. Among the different processing conditions tested, the ethanol-fixed, unstained TP was most similar to the previously established air-dried, unstained TP protocol. All smears and Pap-stained TPs had significantly different marker MFIs from the established condition. After short-term storage, the established condition showed comparable results, but ethanol-fixed and Pap-stained slides showed significant differences. ETCs were detectable after long-term storage at -80°C in comparable numbers to freshly prepared slides, but most marker MFIs were significantly different. CONCLUSIONS It is possible to detect ETCs under different processing and storage conditions, lending promise to the application of this method in broader settings. Because of decreased immunofluorescence-signature distinctions between cells, morphology may need to play a larger role.
Collapse
Affiliation(s)
- Diane M Libert
- Department of Pathology, Stanford University School of Medicine, Stanford, California, USA
| | - Yili Zhu
- Department of Pathology, Stanford University School of Medicine, Stanford, California, USA
| | - Aihui Wang
- Department of Pathology, Stanford University School of Medicine, Stanford, California, USA
- Department of Chemistry and Biochemistry, University of California, Los Angeles, Los Angeles, California, USA
| | - Grace M Allard
- Department of Pathology, Stanford University School of Medicine, Stanford, California, USA
| | - Alarice Cheng-Yi Lowe
- Department of Pathology, Stanford University School of Medicine, Stanford, California, USA
| |
Collapse
|
40
|
Hassanzadeh-Barforoushi A, Tukova A, Nadalini A, Inglis DW, Chang-Hao Tsao S, Wang Y. Microfluidic-SERS Technologies for CTC: A Perspective on Clinical Translation. ACS APPLIED MATERIALS & INTERFACES 2024. [PMID: 38652011 DOI: 10.1021/acsami.4c01158] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/25/2024]
Abstract
Enumeration and phenotypic profiling of circulating tumor cells (CTCs) provide critical information for clinical diagnosis and treatment monitoring in cancer. To achieve this goal, an integrated system is needed to efficiently isolate CTCs from patient samples and sensitively evaluate their phenotypes. Such integration would comprise a high-throughput single-cell processing unit for the isolation and manipulation of CTCs and a sensitive and multiplexed quantitation unit to detect clinically relevant signals from these cells. Surface-enhanced Raman scattering (SERS) has been used as an analytical method for molecular profiling and in vitro cancer diagnosis. More recently, its multiplexing capability and power to create distinct molecular signatures against their targets have garnered attention. Here, we share our insights into the combined power of microfluidics and SERS in realizing CTC isolation, enumeration, and detection from a clinical translation perspective. We highlight the key operational factors in CTC microfluidic processing and SERS detection from patient samples. We further discuss microfluidic-SERS integration and its clinical utility as a paradigm shift in clinical CTC-based cancer diagnosis and prognostication. Finally, we summarize the challenges and attempt to look forward to what lies ahead of us in potentially translating the technique into real clinical applications.
Collapse
Affiliation(s)
- Amin Hassanzadeh-Barforoushi
- School of Natural Sciences, Faculty of Science and Engineering, Macquarie University, Sydney, New South Wales 2109, Australia
| | - Anastasiia Tukova
- School of Natural Sciences, Faculty of Science and Engineering, Macquarie University, Sydney, New South Wales 2109, Australia
| | - Audrey Nadalini
- School of Natural Sciences, Faculty of Science and Engineering, Macquarie University, Sydney, New South Wales 2109, Australia
| | - David W Inglis
- School of Engineering, Faculty of Science and Engineering, Macquarie University, Sydney, New South Wales 2109, Australia
| | - Simon Chang-Hao Tsao
- School of Natural Sciences, Faculty of Science and Engineering, Macquarie University, Sydney, New South Wales 2109, Australia
- Department of Surgery, Austin Health, University of Melbourne, Heidelberg, Victoria 3084, Australia
| | - Yuling Wang
- School of Natural Sciences, Faculty of Science and Engineering, Macquarie University, Sydney, New South Wales 2109, Australia
| |
Collapse
|
41
|
Booijink R, Terstappen LWMM, Dathathri E, Isebia K, Kraan J, Martens J, Bansal R. Identification of functional and diverse circulating cancer-associated fibroblasts in metastatic castration-naïve prostate cancer patients. Mol Oncol 2024. [PMID: 38634185 DOI: 10.1002/1878-0261.13653] [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: 11/15/2023] [Revised: 03/08/2024] [Accepted: 04/03/2024] [Indexed: 04/19/2024] Open
Abstract
In prostate cancer (PCa), cancer-associated fibroblasts (CAFs) promote tumor progression, drug resistance, and metastasis. Although circulating tumor cells are studied as prognostic and diagnostic markers, little is known about other circulating cells and their association with PCa metastasis. Here, we explored the presence of circulating CAFs (cCAFs) in metastatic castration-naïve prostate cancer (mCNPC) patients. cCAFs were stained with fibroblast activation protein (FAP), epithelial cell adhesion molecule (EpCAM), and receptor-type tyrosine-protein phosphatase C (CD45), then FAP+EpCAM- cCAFs were enumerated and sorted using fluorescence-activated cell sorting. FAP+EpCAM- cCAFs ranged from 60 to 776 (389 mean ± 229 SD) per 2 × 108 mononuclear cells, whereas, in healthy donors, FAP+ EpCAM- cCAFs ranged from 0 to 71 (28 mean ± 22 SD). The mCNPC-derived cCAFs showed positivity for vimentin and intracellular collagen-I. They were viable and functional after sorting, as confirmed by single-cell collagen-I secretion after 48 h of culturing. Two cCAF subpopulations, FAP+CD45- and FAP+CD45+, were identified, both expressing collagen-I and vimentin, but with distinctly different morphologies. Collectively, this study demonstrates the presence of functional and viable circulating CAFs in mCNPC patients, suggesting the role of these cells in prostate cancer.
Collapse
Affiliation(s)
- Richell Booijink
- Personalized Diagnostics and Therapeutics, Department of Bioengineering Technologies, Technical Medical Centre, Faculty of Science and Technology, University of Twente, Enschede, The Netherlands
| | - Leon W M M Terstappen
- Department of Medical Cell BioPhysics, Technical Medical Centre, Faculty of Science and Technology, University of Twente, Enschede, The Netherlands
- Department of General, Visceral and Pediatric Surgery, University Hospital Düsseldorf, Heinrich-Heine University, Germany
| | - Eshwari Dathathri
- Department of Medical Cell BioPhysics, Technical Medical Centre, Faculty of Science and Technology, University of Twente, Enschede, The Netherlands
| | - Khrystany Isebia
- Department of Medical Oncology, Erasmus MC Cancer Institute, University Medical Center, Rotterdam, The Netherlands
| | - Jaco Kraan
- Department of Medical Oncology, Erasmus MC Cancer Institute, University Medical Center, Rotterdam, The Netherlands
| | - John Martens
- Department of Medical Oncology, Erasmus MC Cancer Institute, University Medical Center, Rotterdam, The Netherlands
| | - Ruchi Bansal
- Personalized Diagnostics and Therapeutics, Department of Bioengineering Technologies, Technical Medical Centre, Faculty of Science and Technology, University of Twente, Enschede, The Netherlands
| |
Collapse
|
42
|
Sun S, Yang Q, Jiang D, Zhang Y. Nanobiotechnology augmented cancer stem cell guided management of cancer: liquid-biopsy, imaging, and treatment. J Nanobiotechnology 2024; 22:176. [PMID: 38609981 PMCID: PMC11015566 DOI: 10.1186/s12951-024-02432-5] [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: 12/31/2023] [Accepted: 03/20/2024] [Indexed: 04/14/2024] Open
Abstract
Cancer stem cells (CSCs) represent both a key driving force and therapeutic target of tumoral carcinogenesis, tumor evolution, progression, and recurrence. CSC-guided tumor diagnosis, treatment, and surveillance are strategically significant in improving cancer patients' overall survival. Due to the heterogeneity and plasticity of CSCs, high sensitivity, specificity, and outstanding targeting are demanded for CSC detection and targeting. Nanobiotechnologies, including biosensors, nano-probes, contrast enhancers, and drug delivery systems, share identical features required. Implementing these techniques may facilitate the overall performance of CSC detection and targeting. In this review, we focus on some of the most recent advances in how nanobiotechnologies leverage the characteristics of CSC to optimize cancer diagnosis and treatment in liquid biopsy, clinical imaging, and CSC-guided nano-treatment. Specifically, how nanobiotechnologies leverage the attributes of CSC to maximize the detection of circulating tumor DNA, circulating tumor cells, and exosomes, to improve positron emission computed tomography and magnetic resonance imaging, and to enhance the therapeutic effects of cytotoxic therapy, photodynamic therapy, immunotherapy therapy, and radioimmunotherapy are reviewed.
Collapse
Affiliation(s)
- Si Sun
- Department of Obstetrics and Gynecology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Qiang Yang
- Department of Obstetrics and Gynecology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Dawei Jiang
- Department of Nuclear Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China.
- Hubei Key Laboratory of Molecular Imaging, Wuhan, 430022, China.
- Key Laboratory of Biological Targeted Therapy, the Ministry of Education, Wuhan, 430022, China.
| | - Yuan Zhang
- Department of Obstetrics and Gynecology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China.
| |
Collapse
|
43
|
Gholamzad A, Khakpour N, Khosroshahi EM, Asadi S, Koohpar ZK, Matinahmadi A, Jebali A, Rashidi M, Hashemi M, Sadi FH, Gholamzad M. Cancer stem cells: The important role of CD markers, Signaling pathways, and MicroRNAs. Pathol Res Pract 2024; 256:155227. [PMID: 38490099 DOI: 10.1016/j.prp.2024.155227] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/28/2023] [Revised: 02/23/2024] [Accepted: 02/25/2024] [Indexed: 03/17/2024]
Abstract
For the first time, a subset of small cancer cells identified in acute myeloid leukemia has been termed Cancer Stem Cells (CSCs). These cells are notorious for their robust proliferation, self-renewal abilities, significant tumor-forming potential, spread, and resistance to treatments. CSCs are a global concern, as it found in numerous types of cancer, posing a real-world challenge today. Our review encompasses research on key CSC markers, signaling pathways, and MicroRNA in three types of cancer: breast, colon, and liver. These factors play a critical role in either promoting or inhibiting cancer cell growth. The reviewed studies have shown that as cells undergo malignant transformation, there can be an increase or decrease in the expression of different Cluster of Differentiation (CD) markers on their surface. Furthermore, alterations in essential signaling pathways, such as Wnt and Notch1, may impact CSC proliferation, survival, and movement, while also providing potential targets for cancer therapies. Additionally, some research has focused on MicroRNAs due to their dual role as potential therapeutic biomarkers and their ability to enhance CSCs' response to anti-cancer drugs. MicroRNAs also regulate a wide array of cellular processes, including the self-renewal and pluripotency of CSCs, and influence gene transcription. Thus, these studies indicate that MicroRNAs play a significant role in the malignancy of various tumors. Although the gathered information suggests that specific CSC markers, signaling pathways, and MicroRNAs are influential in determining the destiny of cancer cells and could be advantageous for therapeutic strategies, their precise roles and impacts remain incompletely defined, necessitating further investigation.
Collapse
Affiliation(s)
- Amir Gholamzad
- Department of Microbiology and Immunology, Faculty of Medicine, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran; Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical sciences, Islamic Azad University, Tehran, Iran
| | - Niloofar Khakpour
- Department of Bacteriology and Virology, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Elaheh Mohandesi Khosroshahi
- Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical sciences, Islamic Azad University, Tehran, Iran
| | - Saba Asadi
- Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical sciences, Islamic Azad University, Tehran, Iran
| | - Zeinab Khazaei Koohpar
- Department of Cell and Molecular Biology, Faculty of Biological Sciences,Tonekabon Branch,Islamic Azad University, Tonekabon, Iran
| | - Arash Matinahmadi
- Department of Cellular and Molecular Biology, Nicolaus Copernicus,Torun,Poland
| | - Ali Jebali
- Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical sciences, Islamic Azad University, Tehran, Iran; Deprtment of Medical Nanotechnology,Faculty of Advanced Sciences and Technology,Tehran Medical Sciences,Islamic Azad University, Tehran, Iran
| | - Mohsen Rashidi
- Department Pharmacology, Faculty of Medicine, Mazandaran University of Medical Sciences, Sari, Iran; The Health of Plant and Livestock Products Research Center, Mazandaran University of Medical Sciences, Sari, Iran.
| | - Mehrdad Hashemi
- Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical sciences, Islamic Azad University, Tehran, Iran.
| | | | - Mehrdad Gholamzad
- Department of Microbiology and Immunology, Faculty of Medicine, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran.
| |
Collapse
|
44
|
Corrias G, Lai E, Ziranu P, Mariani S, Donisi C, Liscia N, Saba G, Pretta A, Persano M, Fanni D, Spanu D, Balconi F, Loi F, Deidda S, Restivo A, Pusceddu V, Puzzoni M, Solinas C, Massa E, Madeddu C, Gerosa C, Zorcolo L, Faa G, Saba L, Scartozzi M. Prediction of Response to Anti-Angiogenic Treatment for Advanced Colorectal Cancer Patients: From Biological Factors to Functional Imaging. Cancers (Basel) 2024; 16:1364. [PMID: 38611042 PMCID: PMC11011199 DOI: 10.3390/cancers16071364] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2024] [Revised: 03/24/2024] [Accepted: 03/27/2024] [Indexed: 04/14/2024] Open
Abstract
Colorectal cancer (CRC) is a leading tumor worldwide. In CRC, the angiogenic pathway plays a crucial role in cancer development and the process of metastasis. Thus, anti-angiogenic drugs represent a milestone for metastatic CRC (mCRC) treatment and lead to significant improvement of clinical outcomes. Nevertheless, not all patients respond to treatment and some develop resistance. Therefore, the identification of predictive factors able to predict response to angiogenesis pathway blockade is required in order to identify the best candidates to receive these agents. Unfortunately, no predictive biomarkers have been prospectively validated to date. Over the years, research has focused on biologic factors such as genetic polymorphisms, circulating biomarkers, circulating tumor cells (CTCs), circulating tumor DNA (ctDNA), and microRNA. Moreover, research efforts have evaluated the potential correlation of molecular biomarkers with imaging techniques used for tumor assessment as well as the application of imaging tools in clinical practice. In addition to functional imaging, radiomics, a relatively newer technique, shows real promise in the setting of correlating molecular medicine to radiological phenotypes.
Collapse
Affiliation(s)
- Giuseppe Corrias
- Department of Radiology, University of Cagliari, 09042 Cagliari, Italy;
| | - Eleonora Lai
- Medical Oncology Unit, University Hospital and University of Cagliari, 09042 Cagliari, Italy; (E.L.); (P.Z.); (S.M.); (C.D.); (G.S.); (A.P.); (M.P.); (D.S.); (F.B.); (F.L.); (V.P.); (M.P.); (C.S.); (E.M.); (C.M.); (M.S.)
| | - Pina Ziranu
- Medical Oncology Unit, University Hospital and University of Cagliari, 09042 Cagliari, Italy; (E.L.); (P.Z.); (S.M.); (C.D.); (G.S.); (A.P.); (M.P.); (D.S.); (F.B.); (F.L.); (V.P.); (M.P.); (C.S.); (E.M.); (C.M.); (M.S.)
| | - Stefano Mariani
- Medical Oncology Unit, University Hospital and University of Cagliari, 09042 Cagliari, Italy; (E.L.); (P.Z.); (S.M.); (C.D.); (G.S.); (A.P.); (M.P.); (D.S.); (F.B.); (F.L.); (V.P.); (M.P.); (C.S.); (E.M.); (C.M.); (M.S.)
| | - Clelia Donisi
- Medical Oncology Unit, University Hospital and University of Cagliari, 09042 Cagliari, Italy; (E.L.); (P.Z.); (S.M.); (C.D.); (G.S.); (A.P.); (M.P.); (D.S.); (F.B.); (F.L.); (V.P.); (M.P.); (C.S.); (E.M.); (C.M.); (M.S.)
| | - Nicole Liscia
- Department of Medical Oncology, IRCCS San Raffaele Scientific Institute, Vita-Salute San Raffaele University, 20132 Milan, Italy;
| | - Giorgio Saba
- Medical Oncology Unit, University Hospital and University of Cagliari, 09042 Cagliari, Italy; (E.L.); (P.Z.); (S.M.); (C.D.); (G.S.); (A.P.); (M.P.); (D.S.); (F.B.); (F.L.); (V.P.); (M.P.); (C.S.); (E.M.); (C.M.); (M.S.)
| | - Andrea Pretta
- Medical Oncology Unit, University Hospital and University of Cagliari, 09042 Cagliari, Italy; (E.L.); (P.Z.); (S.M.); (C.D.); (G.S.); (A.P.); (M.P.); (D.S.); (F.B.); (F.L.); (V.P.); (M.P.); (C.S.); (E.M.); (C.M.); (M.S.)
| | - Mara Persano
- Medical Oncology Unit, University Hospital and University of Cagliari, 09042 Cagliari, Italy; (E.L.); (P.Z.); (S.M.); (C.D.); (G.S.); (A.P.); (M.P.); (D.S.); (F.B.); (F.L.); (V.P.); (M.P.); (C.S.); (E.M.); (C.M.); (M.S.)
| | - Daniela Fanni
- Division of Pathology, Department of Medical Sciences and Public Health, AOU Cagliari, University of Cagliari, 09124 Cagliari, Italy; (D.F.); (C.G.); (G.F.)
| | - Dario Spanu
- Medical Oncology Unit, University Hospital and University of Cagliari, 09042 Cagliari, Italy; (E.L.); (P.Z.); (S.M.); (C.D.); (G.S.); (A.P.); (M.P.); (D.S.); (F.B.); (F.L.); (V.P.); (M.P.); (C.S.); (E.M.); (C.M.); (M.S.)
| | - Francesca Balconi
- Medical Oncology Unit, University Hospital and University of Cagliari, 09042 Cagliari, Italy; (E.L.); (P.Z.); (S.M.); (C.D.); (G.S.); (A.P.); (M.P.); (D.S.); (F.B.); (F.L.); (V.P.); (M.P.); (C.S.); (E.M.); (C.M.); (M.S.)
| | - Francesco Loi
- Medical Oncology Unit, University Hospital and University of Cagliari, 09042 Cagliari, Italy; (E.L.); (P.Z.); (S.M.); (C.D.); (G.S.); (A.P.); (M.P.); (D.S.); (F.B.); (F.L.); (V.P.); (M.P.); (C.S.); (E.M.); (C.M.); (M.S.)
| | - Simona Deidda
- Colorectal Surgery Unit, A.O.U. Cagliari, Department of Surgical Science, University of Cagliari, 09042 Cagliari, Italy; (S.D.); (A.R.); (L.Z.)
| | - Angelo Restivo
- Colorectal Surgery Unit, A.O.U. Cagliari, Department of Surgical Science, University of Cagliari, 09042 Cagliari, Italy; (S.D.); (A.R.); (L.Z.)
| | - Valeria Pusceddu
- Medical Oncology Unit, University Hospital and University of Cagliari, 09042 Cagliari, Italy; (E.L.); (P.Z.); (S.M.); (C.D.); (G.S.); (A.P.); (M.P.); (D.S.); (F.B.); (F.L.); (V.P.); (M.P.); (C.S.); (E.M.); (C.M.); (M.S.)
| | - Marco Puzzoni
- Medical Oncology Unit, University Hospital and University of Cagliari, 09042 Cagliari, Italy; (E.L.); (P.Z.); (S.M.); (C.D.); (G.S.); (A.P.); (M.P.); (D.S.); (F.B.); (F.L.); (V.P.); (M.P.); (C.S.); (E.M.); (C.M.); (M.S.)
| | - Cinzia Solinas
- Medical Oncology Unit, University Hospital and University of Cagliari, 09042 Cagliari, Italy; (E.L.); (P.Z.); (S.M.); (C.D.); (G.S.); (A.P.); (M.P.); (D.S.); (F.B.); (F.L.); (V.P.); (M.P.); (C.S.); (E.M.); (C.M.); (M.S.)
| | - Elena Massa
- Medical Oncology Unit, University Hospital and University of Cagliari, 09042 Cagliari, Italy; (E.L.); (P.Z.); (S.M.); (C.D.); (G.S.); (A.P.); (M.P.); (D.S.); (F.B.); (F.L.); (V.P.); (M.P.); (C.S.); (E.M.); (C.M.); (M.S.)
| | - Clelia Madeddu
- Medical Oncology Unit, University Hospital and University of Cagliari, 09042 Cagliari, Italy; (E.L.); (P.Z.); (S.M.); (C.D.); (G.S.); (A.P.); (M.P.); (D.S.); (F.B.); (F.L.); (V.P.); (M.P.); (C.S.); (E.M.); (C.M.); (M.S.)
| | - Clara Gerosa
- Division of Pathology, Department of Medical Sciences and Public Health, AOU Cagliari, University of Cagliari, 09124 Cagliari, Italy; (D.F.); (C.G.); (G.F.)
| | - Luigi Zorcolo
- Colorectal Surgery Unit, A.O.U. Cagliari, Department of Surgical Science, University of Cagliari, 09042 Cagliari, Italy; (S.D.); (A.R.); (L.Z.)
| | - Gavino Faa
- Division of Pathology, Department of Medical Sciences and Public Health, AOU Cagliari, University of Cagliari, 09124 Cagliari, Italy; (D.F.); (C.G.); (G.F.)
| | - Luca Saba
- Department of Radiology, University of Cagliari, 09042 Cagliari, Italy;
| | - Mario Scartozzi
- Medical Oncology Unit, University Hospital and University of Cagliari, 09042 Cagliari, Italy; (E.L.); (P.Z.); (S.M.); (C.D.); (G.S.); (A.P.); (M.P.); (D.S.); (F.B.); (F.L.); (V.P.); (M.P.); (C.S.); (E.M.); (C.M.); (M.S.)
| |
Collapse
|
45
|
Whalen RM, Anderson AN, Jones JA, Sims Z, Chang YH, Nederlof MA, Wong MH, Gibbs SL. Ultra high content analyses of circulating and tumor associated hybrid cells reveal phenotypic heterogeneity. Sci Rep 2024; 14:7350. [PMID: 38538742 PMCID: PMC10973471 DOI: 10.1038/s41598-024-57381-8] [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: 11/03/2023] [Accepted: 03/18/2024] [Indexed: 07/03/2024] Open
Abstract
Persistently high, worldwide mortality from cancer highlights the unresolved challenges of disease surveillance and detection that impact survival. Development of a non-invasive, blood-based biomarker would transform survival from cancer. We demonstrate the functionality of ultra-high content analyses of a newly identified population of tumor cells that are hybrids between neoplastic and immune cells in patient matched tumor and peripheral blood specimens. Using oligonucleotide conjugated antibodies (Ab-oligo) permitting cyclic immunofluorescence (cyCIF), we present analyses of phenotypes among tumor and peripheral blood hybrid cells. Interestingly, the majority of circulating hybrid cell (CHC) subpopulations were not identified in tumor-associated hybrids. These results highlight the efficacy of ultra-high content phenotypic analyses using Ab-oligo based cyCIF applied to both tumor and peripheral blood specimens. The combination of a multiplex phenotypic profiling platform that is gentle enough to analyze blood to detect and evaluate disseminated tumor cells represents a novel approach to exploring novel tumor biology and potential utility for developing the population as a blood-based biomarker in cancer.
Collapse
Affiliation(s)
- Riley M Whalen
- Department of Cell, Developmental, and Cancer Biology, Oregon Health & Science University (OHSU), Portland, OR, 97201, USA
| | - Ashley N Anderson
- Department of Cell, Developmental, and Cancer Biology, Oregon Health & Science University (OHSU), Portland, OR, 97201, USA
| | - Jocelyn A Jones
- Department of Biomedical Engineering, OHSU, Portland, OR, 97201, USA
| | - Zachary Sims
- Department of Biomedical Engineering, OHSU, Portland, OR, 97201, USA
| | - Young Hwan Chang
- Department of Biomedical Engineering, OHSU, Portland, OR, 97201, USA
- Knight Cancer Institute, 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, OHSU, Portland, OR, 97201, USA.
| | - Summer L Gibbs
- Department of Biomedical Engineering, OHSU, Portland, OR, 97201, USA.
- Knight Cancer Institute, OHSU, Portland, OR, 97201, USA.
| |
Collapse
|
46
|
Mishra A, Huang SB, Dubash T, Burr R, Edd JF, Wittner BS, Cunneely QE, Putaturo VR, Deshpande A, Antmen E, Gopinathan KA, Otani K, Miyazawa Y, Kwak JE, Guay SY, Kelly J, Walsh J, Nieman L, Galler I, Chan P, Lawrence MS, Sullivan RJ, Bardia A, Micalizzi DS, Sequist LV, Lee RJ, Franses JW, Ting DT, Brunker PAR, Maheswaran S, Miyamoto DT, Haber DA, Toner M. Tumor cell-based liquid biopsy using high-throughput microfluidic enrichment of entire leukapheresis product. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.03.13.583573. [PMID: 38559183 PMCID: PMC10980012 DOI: 10.1101/2024.03.13.583573] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/04/2024]
Abstract
Circulating Tumor Cells (CTCs), interrogated by sampling blood from patients with cancer, contain multiple analytes, including intact RNA, high molecular weight DNA, proteins, and metabolic markers. However, the clinical utility of tumor cell-based liquid biopsy has been limited since CTCs are very rare, and current technologies cannot process the blood volumes required to isolate a sufficient number of tumor cells for in-depth assays. We previously described a high-throughput microfluidic prototype utilizing high-flow channels and amplification of cell sorting forces through magnetic lenses. Here, we apply this technology to analyze patient-derived leukapheresis products, interrogating a mean blood volume of 5.83 liters from patients with metastatic cancer, with a median of 2,799 CTCs purified per patient. Isolation of many CTCs from individual patients enables characterization of their morphological and molecular heterogeneity, including cell and nuclear size and RNA expression. It also allows robust detection of gene copy number variation, a definitive cancer marker with potential diagnostic applications. High-volume microfluidic enrichment of CTCs constitutes a new dimension in liquid biopsies.
Collapse
|
47
|
Zieren RC, Zondervan PJ, Pienta KJ, Bex A, de Reijke TM, Bins AD. Diagnostic liquid biopsy biomarkers in renal cell cancer. Nat Rev Urol 2024; 21:133-157. [PMID: 37758847 DOI: 10.1038/s41585-023-00818-y] [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] [Accepted: 08/25/2023] [Indexed: 09/29/2023]
Abstract
The clinical presentation of renal cell cancer (RCC) is shifting towards incidental and early detection, creating new challenges in RCC diagnosis. Overtreatment might be reduced with the development of new diagnostic biomarkers to distinguish benign from malignant small renal masses (SRMs). Differently from tissue biopsies, liquid biopsies are obtained from a patient's blood or urine and, therefore, are minimally invasive and suitable for longitudinal monitoring. The most promising types of liquid biopsy biomarkers for RCC diagnosis are circulating tumour cells, extracellular vesicles (EVs) and cell-free DNA. Circulating tumour cell assays have the highest specificity, with low processing time and costs. However, the biological characteristics and low sensitivity limit the use of these markers in SRM diagnostics. Cell-free DNA might complement the diagnosis of high-volume RCC, but the potential for clinical application in SRMs is limited. EVs have the highest biological abundance and the highest sensitivity in identifying low-volume disease; moreover, the molecular characteristics of these markers make EVs suitable for multiple analytical applications. Thus, currently, EV assays have the greatest potential for diagnostic application in RCC (including identification of SRMs). All these liquid biomarkers have potential in clinical practice, pending validation studies. Biomarker implementation will be needed to also improve characterization of RCC subtypes. Last, diagnostic biomarkers might be extended to prognostic or predictive applications.
Collapse
Affiliation(s)
- Richard C Zieren
- Department of Urology, Amsterdam University Medical Centers, University of Amsterdam, Amsterdam, The Netherlands.
- The Brady Urological Institute, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA.
| | - Patricia J Zondervan
- Department of Urology, Amsterdam University Medical Centers, University of Amsterdam, Amsterdam, The Netherlands
| | - Kenneth J Pienta
- The Brady Urological Institute, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Axel Bex
- Specialist Centre for Kidney Cancer, Royal Free Hospital, London, United Kingdom
- Division of Surgery and Interventional Science, University College London, London, United Kingdom
- The Netherlands Cancer Institute, Antoni van Leeuwenhoek Hospital, Amsterdam, The Netherlands
| | - Theo M de Reijke
- Department of Urology, Amsterdam University Medical Centers, University of Amsterdam, Amsterdam, The Netherlands
| | - Adriaan D Bins
- Department of Medical Oncology, Amsterdam University Medical Centers, University of Amsterdam, Amsterdam, The Netherlands
| |
Collapse
|
48
|
Purcell E, Niu Z, Owen S, Grzesik M, Radomski A, Kaehr A, Onukwugha NE, Winkler HF, Ramnath N, Lawrence T, Jolly S, Nagrath S. Circulating tumor cells reveal early predictors of disease progression in patients with stage III NSCLC undergoing chemoradiation and immunotherapy. Cell Rep 2024; 43:113687. [PMID: 38261515 DOI: 10.1016/j.celrep.2024.113687] [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: 03/10/2023] [Revised: 11/02/2023] [Accepted: 01/02/2024] [Indexed: 01/25/2024] Open
Abstract
Circulating tumor cells (CTCs) are early signs of metastasis and can be used to monitor disease progression well before radiological detection by imaging. Using an ultrasensitive graphene oxide microfluidic chip nanotechnology built with graphene oxide sheets, we were able to demonstrate that CTCs can be specifically isolated and molecularly characterized to predict future progression in patients with stage III non-small cell lung cancer (NSCLC). We analyzed CTCs from 26 patients at six time points throughout the treatment course of chemoradiation followed by immune checkpoint inhibitor immunotherapy. We observed that CTCs decreased significantly during treatment, where a larger decrease in CTCs predicted a significantly longer progression-free survival time. Durvalumab-treated patients who have future progression were observed to have sustained higher programmed death ligand 1+ CTCs compared to stable patients. Gene expression profiling revealed phenotypically aggressive CTCs during chemoradiation. By using emerging innovative bioengineering approaches, we successfully show that CTCs are potential biomarkers to monitor and predict patient outcomes in patients with stage III NSCLC.
Collapse
Affiliation(s)
- Emma Purcell
- Department of Chemical Engineering, University of Michigan, Ann Arbor, MI 48109, USA
| | - Zeqi Niu
- Department of Chemical Engineering, University of Michigan, Ann Arbor, MI 48109, USA
| | - Sarah Owen
- Department of Chemical Engineering, University of Michigan, Ann Arbor, MI 48109, USA
| | - Madeline Grzesik
- Department of Chemical Engineering, University of Michigan, Ann Arbor, MI 48109, USA
| | - Abigail Radomski
- Department of Chemical Engineering, University of Michigan, Ann Arbor, MI 48109, USA
| | - Anna Kaehr
- Department of Chemical Engineering, University of Michigan, Ann Arbor, MI 48109, USA
| | - Nna-Emeka Onukwugha
- Department of Chemical Engineering, University of Michigan, Ann Arbor, MI 48109, USA
| | | | - Nithya Ramnath
- Veterans Affairs Ann Arbor Healthcare System, Ann Arbor, MI 48105, USA
| | - Theodore Lawrence
- Michigan Medicine, Department of Radiation Oncology, Ann Arbor, MI 48105, USA; Rogel Cancer Center, Ann Arbor, MI 48105, USA
| | - Shruti Jolly
- Michigan Medicine, Department of Radiation Oncology, Ann Arbor, MI 48105, USA; Rogel Cancer Center, Ann Arbor, MI 48105, USA.
| | - Sunitha Nagrath
- Department of Chemical Engineering, University of Michigan, Ann Arbor, MI 48109, USA; Rogel Cancer Center, Ann Arbor, MI 48105, USA.
| |
Collapse
|
49
|
Radhakrishnan V, Kaifi JT, Suvilesh KN. Circulating Tumor Cells: How Far Have We Come with Mining These Seeds of Metastasis? Cancers (Basel) 2024; 16:816. [PMID: 38398206 PMCID: PMC10887304 DOI: 10.3390/cancers16040816] [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: 01/10/2024] [Revised: 02/06/2024] [Accepted: 02/15/2024] [Indexed: 02/25/2024] Open
Abstract
Circulating tumor cells (CTCs) are cancer cells that slough off from the tumor and circulate in the peripheral blood and lymphatic system as micro metastases that eventually results in macro metastases. Through a simple blood draw, sensitive CTC detection from clinical samples has proven to be a useful tool for determining the prognosis of cancer. Recent technological developments now make it possible to detect CTCs reliably and repeatedly from a simple and straightforward blood test. Multicenter trials to assess the clinical value of CTCs have demonstrated the prognostic value of these cancer cells. Studies on CTCs have filled huge knowledge gap in understanding the process of metastasis since their identification in the late 19th century. However, these rare cancer cells have not been regularly used to tailor precision medicine and or identify novel druggable targets. In this review, we have attempted to summarize the milestones of CTC-based research from the time of identification to molecular characterization. Additionally, the need for a paradigm shift in dissecting these seeds of metastasis and the possible future avenues to improve CTC-based discoveries are also discussed.
Collapse
Affiliation(s)
- Vijay Radhakrishnan
- Department of Surgery, Ellis Fischel Cancer Center, Roy Blunt NextGen Precision Health Institute, University of Missouri, Columbia, MO 65212, USA; (V.R.); (J.T.K.)
| | - Jussuf T. Kaifi
- Department of Surgery, Ellis Fischel Cancer Center, Roy Blunt NextGen Precision Health Institute, University of Missouri, Columbia, MO 65212, USA; (V.R.); (J.T.K.)
- Harry S. Truman Memorial Veterans’ Hospital, Columbia, MO 65201, USA
| | - Kanve N. Suvilesh
- Department of Surgery, Ellis Fischel Cancer Center, Roy Blunt NextGen Precision Health Institute, University of Missouri, Columbia, MO 65212, USA; (V.R.); (J.T.K.)
- Harry S. Truman Memorial Veterans’ Hospital, Columbia, MO 65201, USA
| |
Collapse
|
50
|
Magri V, Marino L, De Renzi G, De Meo M, Salvatori F, Buccilli D, Bianco V, Santini D, Nicolazzo C, Gazzaniga P. Early Detection of Disease Progression in Metastatic Cancers: Could CTCs Improve RECIST Criteria? Biomedicines 2024; 12:388. [PMID: 38397990 PMCID: PMC10887063 DOI: 10.3390/biomedicines12020388] [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: 01/04/2024] [Revised: 01/29/2024] [Accepted: 02/05/2024] [Indexed: 02/25/2024] Open
Abstract
Early detection of disease progression is a crucial issue in the management of cancer patients, especially in metastatic settings. Currently, treatment selection mostly relies on criteria based on radiologic evaluations (RECIST). The aim of the present retrospective study is to evaluate the potential inclusion of circulating tumor cells (CTCs) in hybrid criteria. CTC counts from a total of 160 patients with different metastatic tumors were analyzed for this purpose. In our cohort, 73 patients were affected by breast cancer, 69 by colorectal cancer and 18 by prostate cancer. PFS and OS were evaluated according to the corresponding prediction of disease progression by CTCs and RECIST criteria. In breast cancer, CTC-I has an important impact on the progression-free survival (PFS) and overall survival (OS) values. When CTC-I predicted earlier than RECIST-I, the disease progression, the PFS and OS were shorter with respect to the opposite case. In particular, PFS was 11 (5-16) vs. 34 (23-45)-with p < 0.001-and OS was 80 (22-138) vs. 116 (43-189), p = 0.33. The results suggest a promising role of CTCs as complementary information which could significantly improve the clinical outcomes, and they encourage consideration of future trials to evaluate new hybrid criteria, particularly for patients with breast cancer.
Collapse
Affiliation(s)
- Valentina Magri
- Department of Pathology, Oncology and Radiology, Sapienza University of Rome, 00161 Rome, Italy; (F.S.); (D.B.); (V.B.); (D.S.)
| | - Luca Marino
- Department of Mechanical and Aerospace Engineering, Sapienza University of Rome, 00161 Rome, Italy;
| | - Gianluigi De Renzi
- Department of Molecular Medicine, Sapienza University of Rome, 00161 Rome, Italy; (G.D.R.); (M.D.M.); (C.N.); (P.G.)
| | - Michela De Meo
- Department of Molecular Medicine, Sapienza University of Rome, 00161 Rome, Italy; (G.D.R.); (M.D.M.); (C.N.); (P.G.)
| | - Francesca Salvatori
- Department of Pathology, Oncology and Radiology, Sapienza University of Rome, 00161 Rome, Italy; (F.S.); (D.B.); (V.B.); (D.S.)
| | - Dorelsa Buccilli
- Department of Pathology, Oncology and Radiology, Sapienza University of Rome, 00161 Rome, Italy; (F.S.); (D.B.); (V.B.); (D.S.)
| | - Vincenzo Bianco
- Department of Pathology, Oncology and Radiology, Sapienza University of Rome, 00161 Rome, Italy; (F.S.); (D.B.); (V.B.); (D.S.)
| | - Daniele Santini
- Department of Pathology, Oncology and Radiology, Sapienza University of Rome, 00161 Rome, Italy; (F.S.); (D.B.); (V.B.); (D.S.)
| | - Chiara Nicolazzo
- Department of Molecular Medicine, Sapienza University of Rome, 00161 Rome, Italy; (G.D.R.); (M.D.M.); (C.N.); (P.G.)
| | - Paola Gazzaniga
- Department of Molecular Medicine, Sapienza University of Rome, 00161 Rome, Italy; (G.D.R.); (M.D.M.); (C.N.); (P.G.)
| |
Collapse
|