1
|
Yang S, Zhan X, Yuan L, Lamy de la Chapelle M, Fu W, Yang X. Entropy driven-based catalytic biosensors for bioanalysis: From construction to application-A review. Anal Chim Acta 2025; 1338:343549. [PMID: 39832843 DOI: 10.1016/j.aca.2024.343549] [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: 08/29/2024] [Revised: 12/11/2024] [Accepted: 12/12/2024] [Indexed: 01/22/2025]
Abstract
The rapid advancement of precision medicine and the continuous emergence of novel pathogens have presented new challenges for biosensors, necessitating higher requirements. Target amplification technology serves as the core component in biosensor construction. Enzyme-based amplification methods are often sensitive and selective but involve relatively complex operational steps, whereas enzyme-free amplification methods offer simplicity but frequently fail to meet both sensitivity and selectivity simultaneously. Existing research has confirmed that entropy-driven catalyst (EDC) biosensors not only fulfills the demands for sensitivity and selectivity concurrently but also offers ease of operation and flexibility in construction. In this review, we summarize the key advantages of EDC, explore how to construct DNA nanomachines based on these advantages to achieve intracellular detection and simultaneous detection of multiple targets, as well as point-of-care testing (POCT) to address practical issues in clinical diagnosis and treatment. We also anticipate potential challenges, propose corresponding solutions, and outline future development directions for EDC-based biosensors in practical clinical applications. We firmly believe that EDC sensors will emerge as a crucial branch within the realm of biosensor development.
Collapse
Affiliation(s)
- Sha Yang
- Department of Clinical Laboratory Medicine, Southwest Hospital, Third Military Medical University, 30 Gaotanyan, Shapingba, Chongqing 400038, China; Army 953rd Hospital (Shigatse Branch, Xinqiao Hospital), Third Military Medical University, Shigatse, 857000, China
| | - Xinyu Zhan
- Department of Clinical Laboratory Medicine, Southwest Hospital, Third Military Medical University, 30 Gaotanyan, Shapingba, Chongqing 400038, China
| | - Lijia Yuan
- Emergency Department, Southwest Hospital, Third Military Medical University (Army Medical University), 30 Gaotanyan, Shapingba District, Chongqing, 400038, China
| | - Marc Lamy de la Chapelle
- Department of Clinical Laboratory Medicine, Southwest Hospital, Third Military Medical University, 30 Gaotanyan, Shapingba, Chongqing 400038, China; Institut des Molécules et Matériaux Du Mans (IMMM UMR 6283 CNRS), Le Mans Université, Avenue Olivier Messiaen, CEDEX 9, 72085 Le Mans, France; Nanobiophotonics and Laser Microspectroscopy Center, Interdisciplinary Research Institute in Bio-Nano-Sciences, Babes-Bolyai University, Cluj-Napoca, Romania
| | - Weiling Fu
- Department of Clinical Laboratory Medicine, Southwest Hospital, Third Military Medical University, 30 Gaotanyan, Shapingba, Chongqing 400038, China.
| | - Xiang Yang
- Department of Clinical Laboratory Medicine, Southwest Hospital, Third Military Medical University, 30 Gaotanyan, Shapingba, Chongqing 400038, China.
| |
Collapse
|
2
|
Badani A, Ozair A, Khasraw M, Woodworth GF, Tiwari P, Ahluwalia MS, Mansouri A. Immune checkpoint inhibitors for glioblastoma: emerging science, clinical advances, and future directions. J Neurooncol 2025; 171:531-547. [PMID: 39570554 DOI: 10.1007/s11060-024-04881-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2024] [Accepted: 11/04/2024] [Indexed: 11/22/2024]
Abstract
Glioblastoma (GBM), the most common and aggressive primary central nervous system (CNS) tumor in adults, continues to have a dismal prognosis. Across hundreds of clinical trials, few novel approaches have translated to clinical practice while survival has improved by only a few months over the past three decades. Randomized controlled trials of immune checkpoint inhibitors (ICIs), which have seen impressive success for advanced or metastatic extracranial solid tumors, have so far failed to demonstrate a clinical benefit for patients with GBM. This has been secondary to GBM heterogeneity, the unique immunosuppressive CNS microenvironment, immune-evasive strategies by cancer cells, and the rapid evolution of tumor on therapy. This review aims to summarize findings from major clinical trials of ICIs for GBM, review historic failures, and describe currently promising avenues of investigation. We explore the biological mechanisms driving ICI responses, focusing on the role of the tumor microenvironment, immune evasion, and molecular biomarkers. Beyond conventional monotherapy approaches targeting PD-1, PD-L1, CTLA-4, we describe emerging approaches for GBM, such as dual-agent ICIs, and combination of ICIs with oncolytic virotherapy, antigenic peptide vaccines, chimeric antigenic receptor (CAR) T-cell therapy, along with nanoparticle-based delivery systems to enhance ICI efficacy. We highlight potential strategies for improving patient selection and treatment personalization, along with real-time, longitudinal monitoring of therapeutic responses through advanced imaging and liquid biopsy techniques. Integrated radiomics, tissue, and plasma-based analyses, may potentially uncover immunotherapeutic response signatures, enabling early, adaptive therapeutic adjustments. By specifically targeting current therapeutic challenges, outcomes for GBM patients may potentially be improved.
Collapse
Affiliation(s)
- Aarav Badani
- Department of Neurosurgery, Penn State Health Milton S. Hershey Medical Center, Hershey, PA, USA
- Department of Neuroscience, University of California, Berkeley, CA, USA
| | - Ahmad Ozair
- Department of Neurosurgery, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Mustafa Khasraw
- Department of Neurosurgery, Preston Robert Tisch Brain Tumor Center at Duke, Duke University Medical Center, Durham, NC, USA
| | - Graeme F Woodworth
- Department of Neurosurgery, University of Maryland School of Medicine, Baltimore, MD, USA
- Brain Tumor Center, University of Maryland Marlene and Stewart Greenebaum Comprehensive Cancer Center, Baltimore, MD, USA
- University of Maryland - Medicine Institute for Neuroscience Discovery (UM-MIND), Baltimore, MD, USA
| | - Pallavi Tiwari
- Department of Radiology and Biomedical Engineering, University of Wisconsin, Madison, WI, USA
- William S. Middleton Memorial Veterans Affairs (VA) Healthcare, Madison, WI, USA
| | - Manmeet S Ahluwalia
- Miami Cancer Institute, Baptist Health South Florida, Miami, FL, USA
- Department of Translational Medicine, Herbert Wertheim College of Medicine, Florida International University, Miami, FL, USA
| | - Alireza Mansouri
- Department of Neurosurgery, Penn State Health Milton S. Hershey Medical Center, Hershey, PA, USA.
- Penn State Cancer Institute, Penn State Health Milton S. Hershey Medical Center, Hershey, PA, USA.
| |
Collapse
|
3
|
Klein-Scory S, Baraniskin A, Schmiegel W, Mika T, Schroers R, Held S, Heinrich K, Tougeron D, Modest DP, Schwaner I, Eucker J, Pihusch R, Stauch M, Kaiser F, Kahl C, Karthaus M, Müller C, Burkart C, Stintzing S, Heinemann V. Evaluation of circulating tumor DNA as a prognostic and predictive biomarker in BRAF V600E mutated colorectal cancer-results from the FIRE-4.5 study. Mol Oncol 2025; 19:344-356. [PMID: 39630848 DOI: 10.1002/1878-0261.13778] [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: 03/12/2024] [Revised: 09/05/2024] [Accepted: 11/21/2024] [Indexed: 12/07/2024] Open
Abstract
The randomized FIRE-4.5 (AIO KRK0116) trial compared first-line therapy with FOLFOXIRI (folinic acid, fluorouracil, oxaliplatin, and irinotecan) plus either cetuximab or bevacizumab in B-Raf proto-oncogene, serine/threonine kinase (BRAF) V600E-mutant metastatic colorectal cancer (mCRC) patients. This study was accompanied by a prospective translational project analyzing cell-free circulating tumor DNA (ctDNA) in plasma to test whether ctDNA analysis may help to guide clinical treatment decision making. FIRE-4.5 included mCRC patients with BRAF V600E mutation detected by tissue-based analyses. Liquid biopsies (LBs) were collected at baseline (pre-treatment) and during therapy. Digital droplet PCR (ddPCR) technology was applied for determination of BRAF mutations and the in vitro diagnostics (IVD)-certified ONCOBEAM RAS procedure for analysis of RAS mutations. The BRAF V600E variants in ctDNA were analyzable in 66 patients at start of the therapy, at baseline. No BRAF V600E mutations were detected in 26% (17/66) of patients and was associated with a significantly longer progression-free survival (PFS: 13.2 vs 6.5 months; HR 0.47; P = 0.014) and overall survival (OS: 36.8 vs 13.2 months; HR 0.35; P = 0.02) as compared to ctDNA mutant patients. Patients with detectable BRAF mutations showed a clear superiority of FOLFOXIRI plus bevacizumab with regard to PFS (10.4 vs 5.7 months; HR 0.4; P = 0.009) and OS (16.6 vs 11.6 months; HR 0.5; P = 0.15), while this was not the case for BRAF wild-type patients. Follow-up LBs were obtained from 51 patients. Patients converting from BRAF V600E mutant to a BRAF V600 wild-type status (36%, N = 18) had a superior PFS (8.6 vs 2.3 months; P = 0.0002) and OS (17.4 vs 5.1 months; P < 0.0001) compared to patients with stable or increased mutational allele frequency (12%, N = 6). Those patients also achieved a significantly greater disease control rate (89% vs 20%; P = 0.008). In conclusion, LB evaluating ctDNA is informative and may help to guide treatment in patients with BRAF V600E-mutated mCRC.
Collapse
Affiliation(s)
- Susanne Klein-Scory
- Department of Internal Medicine, Universitaetsklinikum Knappschaftskrankenhaus Bochum GmbH, Ruhr University Bochum, Germany
| | - Alexander Baraniskin
- Department of Internal Medicine, Universitaetsklinikum Knappschaftskrankenhaus Bochum GmbH, Ruhr University Bochum, Germany
- Department of Hematology, Oncology and Palliative Care, Evangelisches Krankenhaus Hamm gGmbH, Germany
| | - Wolff Schmiegel
- Department of Internal Medicine, Universitaetsklinikum Knappschaftskrankenhaus Bochum GmbH, Ruhr University Bochum, Germany
| | - Thomas Mika
- Department of Internal Medicine, Universitaetsklinikum Knappschaftskrankenhaus Bochum GmbH, Ruhr University Bochum, Germany
| | - Roland Schroers
- Department of Internal Medicine, Universitaetsklinikum Knappschaftskrankenhaus Bochum GmbH, Ruhr University Bochum, Germany
| | | | - Kathrin Heinrich
- Department of Oncology, LMU University Hospital, Munich, Germany
| | - David Tougeron
- Department of Hepato-Gastroenterology, Poitiers University Hospital and University of Poitiers, France
| | - Dominik P Modest
- Department of Hematology, Oncology, and Cancer Immunology (CCM), Charité-Universitaetsmedizin Berlin, Germany
| | - Ingo Schwaner
- Onkologische Schwerpunktpraxis Kurfürstendamm, Berlin, Germany
| | - Jan Eucker
- Department of Hematology, Oncology, and Cancer Immunology (CBF), Charité-Universitaetsmedizin Berlin, Germany
| | | | | | | | - Christoph Kahl
- Klinikum Magdeburg gGmbH, Department of Hematology, Oncology and Palliative Care Magdeburg, Germany
- Department of Internal Medicine, Clinic III - Hematology, Oncology and Palliative Care, Rostock University Medical Center, Germany
| | - Meinolf Karthaus
- Department of Hematology, Oncology and Palliative Care, München Klinik Harlaching and Neuperlach, Germany
| | | | | | - Sebastian Stintzing
- Department of Hematology, Oncology, and Cancer Immunology (CCM), Charité-Universitaetsmedizin Berlin, Germany
- German Cancer Consortium (DKTK), German Cancer Research Centre (DKFZ), Site Berlin Heidelberg, Germany
| | - Volker Heinemann
- Department of Medicine III, LMU Klinikum, Comprehensive Cancer Center Munich, Germany
- German Cancer Consortium (DKTK), German Cancer Research Centre (DKFZ), Site Munich Heidelberg, Germany
| |
Collapse
|
4
|
Kaorey N, Dickinson K, Agnihotram VR, Zeitouni A, Sadeghi N, Burnier JV. The role of ctDNA from liquid biopsy in predicting survival outcomes in HPV-negative head and neck cancer: A meta-analysis. Oral Oncol 2025; 161:107148. [PMID: 39742703 DOI: 10.1016/j.oraloncology.2024.107148] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2024] [Accepted: 12/17/2024] [Indexed: 01/04/2025]
Abstract
The incidence of head and neck cancer (HNC) is on the rise, making it a significant clinical challenge. Human papillomavirus (HPV)-related and HPV-negative HNC exhibit distinct etiopathogenesis and prognoses, requiring targeted approaches for effective management. Conventional tissue biopsies are essential for confirming the diagnosis and locating solid tumors. However, they have limitations in detecting microscopic disease, tracking treatment response, and capturing the dynamic heterogeneity of the mutational profile within the tumor. Liquid biopsy using circulating tumor DNA (ctDNA) analysis has emerged as a promising non-invasive tool to overcome the drawbacks of conventional biopsy for comprehensive molecular profiling. This meta-analysis aims to colligate available evidence on the clinical utility of ctDNA analysis in predicting survival outcomes, specifically in HPV-negative HNC. Our systematic search of six electronic databases identified eight publications (N = 886 patients) meeting the inclusion criteria. The included studies reported data from HPV-negative HNC patients, employing ctDNA analysis to report survival outcomes. Our findings reveal a significant association between mutation or methylation in ctDNA and worsened survival outcomes in HPV-negative HNC cases. The presence of ctDNA mutations in TP53 and methylation of SEPT9 and SHOX2 was linked to reduced overall survival, disease-free survival, and progression-free survival. Subgroup analyses demonstrated consistent associations across different survival outcomes, ctDNA detection methods, and blood collection tubes used. Our study underscores the need for future research endeavors prioritizing larger, well-designed prospective studies with standardized methodologies to further elucidate the role of ctDNA analysis in guiding personalized treatment approaches and optimizing patient care in this specific HNC cohort.
Collapse
Affiliation(s)
- Nivedita Kaorey
- Cancer Research Program, Research Institute of the McGill University Health Centre, Montreal, Canada; Department of Pathology, McGill University, Montreal, Canada.
| | - Kyle Dickinson
- Cancer Research Program, Research Institute of the McGill University Health Centre, Montreal, Canada.
| | | | - Anthony Zeitouni
- Department of Otolaryngology-Head and Neck Surgery, McGill University, Montreal, Canada.
| | - Nader Sadeghi
- Gerald Bronfman Department of Oncology, McGill University, Montreal, Canada; Department of Otolaryngology-Head and Neck Surgery, McGill University, Montreal, Canada.
| | - Julia V Burnier
- Cancer Research Program, Research Institute of the McGill University Health Centre, Montreal, Canada; Department of Pathology, McGill University, Montreal, Canada; Gerald Bronfman Department of Oncology, McGill University, Montreal, Canada.
| |
Collapse
|
5
|
Veronese N, Luchini C, Ciriminna S, Spinelli K, Fruscione S, Mattiolo P, Belluzzo M, Messina V, Smith L, Barbagallo M, Mazzucco W. Potentialities and critical issues of liquid biopsy in clinical practice: An umbrella review. Transl Oncol 2025; 52:102172. [PMID: 39817953 PMCID: PMC11786759 DOI: 10.1016/j.tranon.2024.102172] [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: 08/20/2024] [Revised: 10/09/2024] [Accepted: 10/29/2024] [Indexed: 01/18/2025] Open
Abstract
BACKGROUND Liquid biopsy (LB) is a laboratory test performed on a fluid sample aiming at analyzing molecular data derived from circulating cells and related entities, or from nucleic acids. This umbrella review aims to map and evaluate the evidence supporting the use of LB in medicine across different medical specialities and conditions. METHODS We searched three repositories from database inception up to October 1, 2023 and we included meta-analyses of observational studies reporting data on the use of LB, compared to gold standard, and its accuracy (area under the curve, AUC). RESULTS Among 726 articles initially screened, 42 systematic reviews were included. Most of the outcomes explored (202/211) were related to cancer. We found that 75/211 had an excellent accuracy (AUC >0.90), with one comparison with an AUC equal to 1, i.e., Cell-Free Human Papillomavirus DNA (cfHPV-DNA) for HPV-positive oropharyngeal squamous cell carcinoma. However, considering published meta-analyses, all the outcomes were graded as very low on the GRADE criteria, and the heterogeneity was never reported. DISCUSSION The literature about LB is rapidly increasing and some promising data about precision oncology are now available. However, this umbrella review on existing meta-analyses highlighted some critical issues for providing quantitative estimations on the different roles of LB.
Collapse
Affiliation(s)
- Nicola Veronese
- Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialties, University of Palermo, Palermo, Italy.
| | - Claudio Luchini
- Department of Diagnostics and Public Health, Section of Pathology, University and Hospital Trust of Verona, Verona, Italy
| | - Stefano Ciriminna
- Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialties, University of Palermo, Palermo, Italy
| | - Katia Spinelli
- Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialties, University of Palermo, Palermo, Italy
| | - Santo Fruscione
- Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialties, University of Palermo, Palermo, Italy
| | - Paola Mattiolo
- Department of Diagnostics and Public Health, Section of Pathology, University and Hospital Trust of Verona, Verona, Italy
| | - Miriam Belluzzo
- Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialties, University of Palermo, Palermo, Italy
| | - Veronica Messina
- Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialties, University of Palermo, Palermo, Italy
| | - Lee Smith
- Centre for Health Performance and Wellbeing, Anglia Ruskin University, Cambridge, UK
| | - Mario Barbagallo
- Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialties, University of Palermo, Palermo, Italy
| | - Walter Mazzucco
- Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialties, University of Palermo, Palermo, Italy; Division of Biostatistics & Epidemiology Research, Cincinnati Children's Hospital Medical Center, University of Cincinnati College of Medicine, United States
| |
Collapse
|
6
|
Koo KM, Farhana FZ, Ross AG, Shiddiky MJA. Enhancing Electrochemical Biosensing of Circulating Nucleic Acids at the Electrode-Biomolecule-Electrolyte Interfaces. Anal Chem 2025. [PMID: 39878560 DOI: 10.1021/acs.analchem.4c05771] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2025]
Abstract
The detection and analysis of circulating cell-free nucleic acid (ccfNA) biomolecules are redefining a new era of molecular targeted cancer therapies. However, the clinical translation of electrochemical ccfNA biosensing remains hindered by unresolved challenges in analytical specificity and sensitivity. In this Perspective, we present a novel electrochemical framework for improving ccfNA biosensor performance by optimizing the critical electrode-biomolecules-electrolyte interfaces. We highlight and elucidate related research works on modification-free electrode sensor surfaces, nucleic acids as biological scaffolds, and redesigning redox reporter systems. We conclude by providing an outlook into the future research developments of ccfNA electrochemical biosensing, emphasizing the potential to overcome current analytical limitations by controlling the complex interplay of target biomolecules and redox species at the electrode surface. These advances are poised to significantly impact the development of electrochemical ccfNA technologies, improving both cancer diagnostics and therapeutic monitoring.
Collapse
Affiliation(s)
- Kevin M Koo
- The University of Queensland Centre for Clinical Research (UQCCR), Brisbane, Queensland 4029, Australia
- Australian Institute for Bioengineering and Nanotechnology (AIBN), The University of Queensland, Brisbane, Queensland 4072, Australia
| | - Fatema Zerin Farhana
- Rural Health Research Institute, Charles Sturt University, Orange, New South Wales 2800, Australia
| | - Allen G Ross
- Rural Health Research Institute, Charles Sturt University, Orange, New South Wales 2800, Australia
| | - Muhammad J A Shiddiky
- Rural Health Research Institute, Charles Sturt University, Orange, New South Wales 2800, Australia
| |
Collapse
|
7
|
Tsui WHA, Ding SC, Jiang P, Lo YMD. Artificial intelligence and machine learning in cell-free-DNA-based diagnostics. Genome Res 2025; 35:1-19. [PMID: 39843210 PMCID: PMC11789496 DOI: 10.1101/gr.278413.123] [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] [Indexed: 01/24/2025]
Abstract
The discovery of circulating fetal and tumor cell-free DNA (cfDNA) molecules in plasma has opened up tremendous opportunities in noninvasive diagnostics such as the detection of fetal chromosomal aneuploidies and cancers and in posttransplantation monitoring. The advent of high-throughput sequencing technologies makes it possible to scrutinize the characteristics of cfDNA molecules, opening up the fields of cfDNA genetics, epigenetics, transcriptomics, and fragmentomics, providing a plethora of biomarkers. Machine learning (ML) and/or artificial intelligence (AI) technologies that are known for their ability to integrate high-dimensional features have recently been applied to the field of liquid biopsy. In this review, we highlight various AI and ML approaches in cfDNA-based diagnostics. We first introduce the biology of cell-free DNA and basic concepts of ML and AI technologies. We then discuss selected examples of ML- or AI-based applications in noninvasive prenatal testing and cancer liquid biopsy. These applications include the deduction of fetal DNA fraction, plasma DNA tissue mapping, and cancer detection and localization. Finally, we offer perspectives on the future direction of using ML and AI technologies to leverage cfDNA fragmentation patterns in terms of methylomic and transcriptional investigations.
Collapse
Affiliation(s)
- W H Adrian Tsui
- Center for Novostics, Hong Kong Science Park, Pak Shek Kok, New Territories, Hong Kong SAR, China
- Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Shatin, New Territories, Hong Kong SAR, China
- Department of Chemical Pathology, The Chinese University of Hong Kong, Shatin, New Territories, Hong Kong SAR, China
| | - Spencer C Ding
- Center for Novostics, Hong Kong Science Park, Pak Shek Kok, New Territories, Hong Kong SAR, China
- Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Shatin, New Territories, Hong Kong SAR, China
- Department of Chemical Pathology, The Chinese University of Hong Kong, Shatin, New Territories, Hong Kong SAR, China
| | - Peiyong Jiang
- Center for Novostics, Hong Kong Science Park, Pak Shek Kok, New Territories, Hong Kong SAR, China
- Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Shatin, New Territories, Hong Kong SAR, China
- Department of Chemical Pathology, The Chinese University of Hong Kong, Shatin, New Territories, Hong Kong SAR, China
- State Key Laboratory of Translational Oncology, The Chinese University of Hong Kong, Shatin, New Territories, Hong Kong SAR, China
| | - Y M Dennis Lo
- Center for Novostics, Hong Kong Science Park, Pak Shek Kok, New Territories, Hong Kong SAR, China;
- Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Shatin, New Territories, Hong Kong SAR, China
- Department of Chemical Pathology, The Chinese University of Hong Kong, Shatin, New Territories, Hong Kong SAR, China
- State Key Laboratory of Translational Oncology, The Chinese University of Hong Kong, Shatin, New Territories, Hong Kong SAR, China
| |
Collapse
|
8
|
Frydendahl A, Widman AJ, Øgaard N, Arora A, Halmos D, Nors J, Ahrenfeldt J, Henriksen TV, Demuth C, Raaby L, Rasmussen MH, Therkildsen C, Landau DA, Andersen CL. Whole-genome sequencing of cell-free DNA reveals DNA of tumor origin in plasma from patients with colorectal adenomas. Mol Oncol 2025. [PMID: 39831554 DOI: 10.1002/1878-0261.13803] [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: 07/15/2024] [Revised: 11/08/2024] [Accepted: 01/09/2025] [Indexed: 01/22/2025] Open
Abstract
The presence of circulating tumor DNA (ctDNA) in patients with colorectal adenomas remains uncertain. Studies using tumor-agnostic approaches report ctDNA in 10-15% of patients, though with uncertainty as to whether the signal originates from the adenoma. To obtain an accurate estimate of the proportion of patients with ctDNA, a sensitive tumor-informed strategy is preferred, as it ensures the detected signal originates from the adenoma. Here, tumor-informed whole-genome sequencing-based ctDNA analysis (MRD-EDGESNV) was applied to two independent cohorts. Cohort 1, comprising 93 patients with stage III colorectal cancer (CRC) and 40 healthy individuals, was used to establish the signal threshold at 95% specificity. This threshold was then applied to Cohort 2, consisting of 22 patients with symptomatic and 20 with asymptomatic adenomas. In stage III, MRD-EDGESNV had an area under the curve of 0.98. ctDNA was detected in 50% and 25% of patients with symptomatic and asymptomatic adenomas, respectively. The median adenoma plasma tumor fraction was 5.9 × 10-5. These finding not only demonstrate the feasibility of ctDNA detection in patients with colorectal adenomas, but also provides an estimate of the necessary sensitivity required to detect these lesions, paving the way for future ctDNA-based screening strategies.
Collapse
Affiliation(s)
- Amanda Frydendahl
- Department of Molecular Medicine, Aarhus University Hospital, Denmark
- Department of Clinical Medicine, Aarhus University, Denmark
| | - Adam J Widman
- Memorial Sloan Kettering Cancer Center, New York, NY, USA
- New York Genome Center, New York, NY, USA
| | - Nadia Øgaard
- Department of Molecular Medicine, Aarhus University Hospital, Denmark
- Department of Clinical Medicine, Aarhus University, Denmark
| | | | - Daniel Halmos
- New York Genome Center, New York, NY, USA
- Weill Cornell Medicine, New York, NY, USA
| | - Jesper Nors
- Department of Molecular Medicine, Aarhus University Hospital, Denmark
- Department of Clinical Medicine, Aarhus University, Denmark
| | - Johanne Ahrenfeldt
- Department of Molecular Medicine, Aarhus University Hospital, Denmark
- Department of Clinical Medicine, Aarhus University, Denmark
| | - Tenna V Henriksen
- Department of Molecular Medicine, Aarhus University Hospital, Denmark
- Department of Clinical Medicine, Aarhus University, Denmark
| | - Christina Demuth
- Department of Molecular Medicine, Aarhus University Hospital, Denmark
- Department of Clinical Medicine, Aarhus University, Denmark
| | - Line Raaby
- Department of Molecular Medicine, Aarhus University Hospital, Denmark
- Department of Clinical Medicine, Aarhus University, Denmark
- Department of Pathology, Aarhus University Hospital, Denmark
| | - Mads H Rasmussen
- Department of Molecular Medicine, Aarhus University Hospital, Denmark
- Department of Clinical Medicine, Aarhus University, Denmark
| | - Christina Therkildsen
- Gastro Unit, Copenhagen University Hospital, Amager - Hvidovre Hospital, Hvidovre, Denmark
| | - Dan A Landau
- New York Genome Center, New York, NY, USA
- Weill Cornell Medicine, New York, NY, USA
| | - Claus L Andersen
- Department of Molecular Medicine, Aarhus University Hospital, Denmark
- Department of Clinical Medicine, Aarhus University, Denmark
| |
Collapse
|
9
|
Normanno N, Morabito A, Rachiglio AM, Sforza V, Landi L, Bria E, Delmonte A, Cappuzzo F, De Luca A. Circulating tumour DNA in early stage and locally advanced NSCLC: ready for clinical implementation? Nat Rev Clin Oncol 2025:10.1038/s41571-024-00985-w. [PMID: 39833354 DOI: 10.1038/s41571-024-00985-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/20/2024] [Indexed: 01/22/2025]
Abstract
Circulating tumour DNA (ctDNA) can be released by cancer cells into biological fluids through apoptosis, necrosis or active release. In patients with non-small-cell lung cancer (NSCLC), ctDNA levels correlate with clinical and pathological factors, including histology, tumour size and proliferative status. Currently, ctDNA analysis is recommended for molecular profiling in patients with advanced-stage NSCLC. In this Review, we summarize the increasing evidence suggesting that ctDNA has potential clinical applications in the management of patients with early stage and locally advanced NSCLC. In those with early stage NSCLC, detection of ctDNA before and/or after surgery is associated with a greater risk of disease recurrence. Longitudinal monitoring after surgery can further increase the prognostic value of ctDNA testing and enables detection of disease recurrence earlier than the assessment of clinical or radiological progression. In patients with locally advanced NSCLC, the detection of ctDNA after chemoradiotherapy is also associated with a greater risk of disease progression. Owing to the limited number of patients enrolled and the different technologies used for ctDNA testing in most of the clinical studies performed thus far, their results are not sufficient to currently support the routine clinical use of ctDNA monitoring in patients with early stage or locally advanced NSCLC. Therefore, we discuss the need for interventional studies to provide evidence for implementing ctDNA testing in this setting.
Collapse
Affiliation(s)
- Nicola Normanno
- Scientific Directorate, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) "Dino Amadori", Meldola, Italy.
| | - Alessandro Morabito
- Thoracic Department, Istituto Nazionale Tumori - IRCCS - Fondazione G. Pascale, Napoli, Italy
| | - Anna Maria Rachiglio
- Cell Biology and Biotherapy Unit, Istituto Nazionale Tumori - IRCCS - Fondazione G. Pascale, Napoli, Italy
| | - Vincenzo Sforza
- Thoracic Department, Istituto Nazionale Tumori - IRCCS - Fondazione G. Pascale, Napoli, Italy
| | - Lorenza Landi
- Clinical Trials Center: Phase 1 and Precision Medicine, IRCCS "Regina Elena" National Cancer Institute, Rome, Italy
| | - Emilio Bria
- Medical Oncology Unit, Università Cattolica del Sacro Cuore, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Roma, Italy
- Medical Oncology, Ospedale Isola Tiberina Gemelli Isola, Roma, Italy
| | - Angelo Delmonte
- Medical Oncology Department, IRCCS IRST "Dino Amadori", Meldola, Italy
| | - Federico Cappuzzo
- Division of Medical Oncology 2, IRCCS "Regina Elena" National Cancer Institute, Rome, Italy
| | - Antonella De Luca
- Cell Biology and Biotherapy Unit, Istituto Nazionale Tumori - IRCCS - Fondazione G. Pascale, Napoli, Italy
| |
Collapse
|
10
|
Guo W, Chen W, Zhang J, Li M, Huang H, Wang Q, Fei X, Huang J, Zheng T, Fan H, Wang Y, Gu H, Ding G, Chen Y. High-throughput methylation sequencing reveals novel biomarkers for the early detection of renal cell carcinoma. BMC Cancer 2025; 25:96. [PMID: 39819319 PMCID: PMC11737265 DOI: 10.1186/s12885-024-13380-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2024] [Accepted: 12/20/2024] [Indexed: 01/19/2025] Open
Abstract
PURPOSE Renal cell carcinoma (RCC) is a common malignancy, with patients frequently diagnosed at an advanced stage due to the absence of sufficiently sensitive detection technologies, significantly compromising patient survival and quality of life. Advances in cell-free DNA (cfDNA) methylation profiling using liquid biopsies offer a promising non-invasive diagnostic option, but robust biomarkers for early detection are current not available. This study aimed to identify methylation biomarkers for RCC and establish a DNA methylation signature-based prognostic model for this disease. METHODS High-throughput methylation sequencing was performed on peripheral blood samples obtained from 49 primarily Stage I RCC patients and 44 healthy controls. Comparative analysis and Least Absolute Shrinkage and Selection Operator (LASSO) regression methods were employed to identify RCC methylation signatures.Subsequently, methylation markers-based diagnostic and prognostic models for RCC were independently trained and validated using random forest and Cox regression methodologies, respectively. RESULTS Comparative analysis revealed 864 differentially methylated CpG islands (DMCGIs), 96.3% of which were hypermethylated. Using a training set from The Cancer Genome Atlas (TCGA) dataset of 443 early-stage RCC tumors and matched normal tissues, we applied LASSO regression and identified 23 methylation signatures. We then constructed a random forest-based diagnostic model for early-stage RCC and validated the model using two independent datasets: a TCGA set of 460 RCC tumors and controls, and a blood sample set from our study of 15 RCC cases and 29 healthy controls. For Stage I RCC tissue, the model showed excellent discrimination (AUC-ROC: 0.999, sensitivity: 98.5%, specificity: 100%). Blood sample validation also yielded commendable results (AUC-ROC: 0.852, sensitivity: 73.9%, specificity: 89.7%). Further analysis using Cox regression identified 7 of the 23 DMCGIs as prognostic markers for RCC, allowing the development of a prognostic model with strong predictive power for 1-, 3-, and 5-year survival (AUC-ROC > 0.7). CONCLUSIONS Our findings highlight the critical role of hypermethylation in RCC etiology and progression, and present these identified biomarkers as promising candidates for diagnostic and prognostic applications.
Collapse
Affiliation(s)
- Wenhao Guo
- Department of Urology, Sir Run-Run Shaw Hospital, College of Medicine, Zhejiang University, Hangzhou, 310016, Zhejiang Province, China
- Department of Urology, Shaoxing Branch of Sir Run-Run Shaw Hospital, College of Medicine, Zhejiang University, Shaoxing, 312000, Zhejiang Province, China
| | - Weiwu Chen
- Department of Urology, Sir Run-Run Shaw Hospital, College of Medicine, Zhejiang University, Hangzhou, 310016, Zhejiang Province, China
- School of Medicine, Zhejiang University, Hangzhou, 310011, Zhejiang Province, China
| | - Jie Zhang
- Department of Urology, Sir Run-Run Shaw Hospital, College of Medicine, Zhejiang University, Hangzhou, 310016, Zhejiang Province, China
| | - Mingzhe Li
- Department of Urology, Sir Run-Run Shaw Hospital, College of Medicine, Zhejiang University, Hangzhou, 310016, Zhejiang Province, China
| | - Hongyuan Huang
- Department of Urology, Jinjiang Municipal Hospital, Quanzhou, 362000, Fujian Province, China
| | - Qian Wang
- Hangzhou Shengting Medical Technology Co., Ltd., Hangzhou, 310018, Zhejiang Province, China
| | - Xiaoyi Fei
- Hangzhou Shengting Medical Technology Co., Ltd., Hangzhou, 310018, Zhejiang Province, China
| | - Jian Huang
- Anhui Province Key Laboratory of Medical Physics and Technology, Institute of Health and Medical Technology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, 350 Shushanhu Road, Hefei, 230031, Anhui Province, China
| | - Tongning Zheng
- Department of Urology, Ningbo Zhenhai People's Hospital, Ningbo, 315202, Zhejiang Province, China
| | - Haobo Fan
- Department of Urology, Sir Run-Run Shaw Hospital, College of Medicine, Zhejiang University, Hangzhou, 310016, Zhejiang Province, China
- School of Medicine, Zhejiang University, Hangzhou, 310011, Zhejiang Province, China
| | - Yunfei Wang
- Hangzhou Shengting Medical Technology Co., Ltd., Hangzhou, 310018, Zhejiang Province, China
| | - Hongcang Gu
- Anhui Province Key Laboratory of Medical Physics and Technology, Institute of Health and Medical Technology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, 350 Shushanhu Road, Hefei, 230031, Anhui Province, China.
| | - Guoqing Ding
- Department of Urology, Sir Run-Run Shaw Hospital, College of Medicine, Zhejiang University, Hangzhou, 310016, Zhejiang Province, China.
| | - Yicheng Chen
- Department of Urology, Sir Run-Run Shaw Hospital, College of Medicine, Zhejiang University, Hangzhou, 310016, Zhejiang Province, China.
| |
Collapse
|
11
|
Zhu D, Li J, Zhang W, Wang Y, Wang H, Fei R, Ye Q, Peng D, Luan J, Xu C, Wu X, Huang D, Ding C, Jin S. Highly specific multiplex DNA methylation detection for liquid biopsy of colorectal cancer. Clin Chim Acta 2025; 565:120026. [PMID: 39491766 DOI: 10.1016/j.cca.2024.120026] [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: 08/05/2024] [Revised: 10/24/2024] [Accepted: 10/31/2024] [Indexed: 11/05/2024]
Abstract
BACKGROUND Circulating tumor DNA (ctDNA) has emerged as a useful biomarker for cancer detection and prognosis. In this study, we developed a strategy for developing a highly specific multiplex qPCR assay to detect methylated ctDNA in the blood of colorectal cancer (CRC) patients and investigated the potential use for the detection and prognosis of CRC. METHODS Bisulfite conversion and amplicon sequencing were used to confirm potential CRC-specific DNA methylation markers. The selected DNA methylation candidates were validated by qMSP. The six best-performing markers were used to develop a new single-tube multiplex quantitative methylation-specific PCR assay (mqMSP). The mqMSP assay was applied to analyze plasma samples from 114 CRC patients, 47 patients with advanced adenoma, 45 patients with benign polyps, and 57 healthy controls. The clinical performance of the assay and associations with clinical outcomes were assessed. RESULTS Six DNA methylation biomarkers were confirmed to be specifically hypermethylated in CRC tumor tissues. The newly developed mqMSP assay detected CRC with extremely high specificity (specificity of 98.2 %, with sensitivity of 67.5 %). The detection rate of ctDNA was significantly correlated with tumor size and clinical stage, with ctDNA methylation levels in the blood markedly increased with larger tumor size, poor differentiation, and advanced stage. Moreover, high preoperative methylated ctDNA level was associated with worse recurrence-free survival and overall survival. CONCLUSION We provided a strategy for identification of multiple highly-specific DNA methylation markers for designing multiplex DNA methylation assays for liquid biopsies of CRC. The newly developed assay has potential for CRC early detection, and prognosis.
Collapse
Affiliation(s)
- Dewen Zhu
- Key Laboratory of Laboratory Medicine, Ministry of Education of China, School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China; Department of Laboratory Medicine, Taihe Hospital, Hubei University of Medicine, Shiyan 442000, China
| | - Jinlei Li
- Wenzhou Medical University, Wenzhou, Zhejiang 325035, China; Department of Colorectal Surgery, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325000, China
| | - Wenwen Zhang
- Department of Pathology, Fudan University Shanghai Cancer Center, Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - Yishuai Wang
- Key Laboratory of Laboratory Medicine, Ministry of Education of China, School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China
| | - Huidong Wang
- Key Laboratory of Laboratory Medicine, Ministry of Education of China, School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China
| | - Ruoyan Fei
- Key Laboratory of Laboratory Medicine, Ministry of Education of China, School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China
| | - Qian Ye
- Key Laboratory of Laboratory Medicine, Ministry of Education of China, School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China
| | - Danli Peng
- Key Laboratory of Laboratory Medicine, Ministry of Education of China, School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China
| | - Ju Luan
- Zhejiang Innomed Biomedical Co., Ltd., Wenzhou 325036, China
| | - Chang Xu
- Department of Colorectal Surgery, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325000, China
| | - Xiaoli Wu
- Department of Gastroenterology, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325000, China
| | - Dan Huang
- Key Laboratory of Laboratory Medicine, Ministry of Education of China, School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China
| | - Chunming Ding
- Key Laboratory of Laboratory Medicine, Ministry of Education of China, School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China; Department of Pathology, Fudan University Shanghai Cancer Center, Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China.
| | - Shengnan Jin
- Key Laboratory of Laboratory Medicine, Ministry of Education of China, School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China.
| |
Collapse
|
12
|
Ko S, Jang J, Yi SS, Kwon C. Early detection of canine hemangiosarcoma via cfDNA fragmentation and copy number alterations in liquid biopsies using machine learning. Front Vet Sci 2025; 11:1489402. [PMID: 39872607 PMCID: PMC11769935 DOI: 10.3389/fvets.2024.1489402] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2024] [Accepted: 12/26/2024] [Indexed: 01/30/2025] Open
Abstract
Hemangiosarcoma is a highly malignant tumor commonly affecting canines, originating from endothelial cells that line blood vessels, underscoring the importance of early detection. This canine cancer is analogous to human angiosarcoma, and the development of liquid biopsies leveraging cell-free DNA (cfDNA) represents a promising step forward in early cancer diagnosis. In this study, we utilized Whole Genome Sequencing (WGS) to analyze fragment sizes and copy number alterations (CNAs) in cfDNA from 21 hemangiosarcoma-affected and 36 healthy dogs, aiming to enhance early cancer detection accuracy through machine learning models. Our findings reveal that similar to trends in human oncology, hemangiosarcoma samples exhibited shorter DNA fragment sizes compared to healthy controls, with a notable leftward shift in the primary peak. Interestingly, canine hemangiosarcoma DNA fragment sizes demonstrated eight distinct periodic patterns diverging from those typically observed in human angiosarcoma. Additionally, we identified seven novel genomic gains and nine losses in the hemangiosarcoma samples. Applying machine learning to the cfDNA fragment size distribution, we achieved an impressive average Area Under the Curve (AUC) of 0.93 in 10-fold cross-validation, underscoring the potential of this approach for precise early-stage cancer classification. This study confirms distinctive cfDNA fragment size and CNA patterns in hemangiosarcoma-affected vs. healthy dogs and demonstrates the promise of these biomarkers in canine cancer screening, early detection, and monitoring via liquid biopsies. These findings establish a foundation for broader research on cfDNA analysis in various canine cancers, integrating methodologies from human oncology to enhance early detection and diagnostic precision in veterinary medicine.
Collapse
Affiliation(s)
| | | | - Sun Shin Yi
- Department of Biomedical Laboratory Science, Soonchunhyang University, Asan, Republic of Korea
- BK21 Four Project, Department of Medical Sciences, Soonchunhyang University, Asan, Republic of Korea
| | | |
Collapse
|
13
|
Yamamoto R, Asano H, Tamaki R, Saito Y, Hosokawa A, Watari H, Umazume T. Dynamics and Half-Life of Cell-Free DNA After Exercise: Insights from a Fragment Size-Specific Measurement Approach. Diagnostics (Basel) 2025; 15:109. [PMID: 39795637 PMCID: PMC11720216 DOI: 10.3390/diagnostics15010109] [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: 11/29/2024] [Revised: 01/02/2025] [Accepted: 01/02/2025] [Indexed: 01/13/2025] Open
Abstract
Background: Cell-free DNA (cfDNA) is present in healthy individuals but is elevated in those undergoing physical exertion, trauma, sepsis, and certain cancers. Maintaining cfDNA concentrations is vital for immune homeostasis and preventing inflammatory responses. Understanding cfDNA release and clearance is essential for using cfDNA as a biomarker in clinical diagnostics. We focused on the fragment size of cfDNA and investigated cfDNA dynamics and half-life, particularly the 100-250 base pair fragments. Methods: Healthy, adult men (n = 5; age 40 ± 4.1 years) were subjected to a 30 min treadmill exercise. Blood samples were collected at 0, 5, 10, 15, 30, and 60 min post-exercise using PAXgene® Blood ccfDNA tubes to stabilize and prevent nuclease-mediated cfDNA degradation and minimize genomic DNA contamination risk. The cfDNA concentration was measured using an electrophoresis-based technique (4150 TapeStation system) to quantify the concentration based on cfDNA fragment size. Results: The results showed a cfDNA half-life of 24.2 min, with a transient increase in 100-250 base pair cfDNA fragments post-exercise, likely due to nuclease activity. These levels rapidly reverted to the baseline within an hour. Conclusions: The rapid clearance of cfDNA underscores its potential as a biomarker for real-time disease monitoring and the evaluation of treatment efficacy. This study is expected to standardize cfDNA investigations, enhancing diagnosis and treatment monitoring across various disease conditions.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Takeshi Umazume
- Department of Obstetrics, Hokkaido University Hospital, Sapporo 060-8648, Japan; (R.Y.)
| |
Collapse
|
14
|
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] [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
|
15
|
Nikai Y, Satomi K, Saito K, Gomyo M, Matsushita Y, Kato K, Nagahama K, Isomura A, Hayashi A, Yamagishi Y, Sasaki N, Kobayashi K, Tsuchiya K, Nagane M, Ichimura K, Shibahara J. Glioblastoma, IDH-wildtype manifesting as intracranial hemorrhage: A case report highlighting the clinical utility of digital polymerase chain reaction in integrated diagnoses. Neuropathology 2025. [PMID: 39748727 DOI: 10.1111/neup.13025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2024] [Revised: 12/10/2024] [Accepted: 12/14/2024] [Indexed: 01/04/2025]
Abstract
The manifestation of glioblastoma, IDH-wildtype (GB) as intracranial hemorrhage (ICH) presents diagnostic and therapeutic challenges. Molecular characteristics, including TERT promoter mutation, EGFR amplification, and chromosome 7 gain/10 loss, were incorporated to diagnose GB in the fifth edition of the World Health Organization Classification of Tumors of the Central Nervous System. When molecular analyses fail to detect low fractions of these genetic alterations, the integrated diagnosis of GB can be enigmatic. This case report describes a 58-year-old man presenting with ICH, masking an underlying GB. Initial histopathology of the evacuated hematoma revealed a small number of atypical glial cells, but a definitive diagnosis was deferred. Subsequent surgery and molecular analysis, including digital polymerase chain reaction (dPCR), confirmed the presence of a TERT C228T mutation in the promoter area, leading to an integrated diagnosis of GB. The patient experienced a favorable clinical outcome following surgery, radiation, temozolomide, and tumor-treating field therapy, without recurrence after 50 months. This case underscores the importance of meticulous histological examination of ICH and exemplifies the clinical utility of dPCR as a complementary diagnostic tool. The effectiveness of dPCR is particularly noteworthy, even in scenarios with minimal tumor cell content, reinforcing its value in the integrated diagnosis of GB.
Collapse
Affiliation(s)
- Yukino Nikai
- Department of Pathology, Kyorin University Faculty of Medicine, Tokyo, Japan
| | - Kaishi Satomi
- Department of Pathology, Kyorin University Faculty of Medicine, Tokyo, Japan
| | - Kuniaki Saito
- Department of Neurosurgery, Kyorin University Faculty of Medicine, Tokyo, Japan
| | - Miho Gomyo
- Department of Radiology, Kyorin University Faculty of Medicine, Tokyo, Japan
| | - Yuko Matsushita
- Department of Pathology, Kyorin University Faculty of Medicine, Tokyo, Japan
| | - Kenichiro Kato
- Department of Pathology, Kyorin University Faculty of Medicine, Tokyo, Japan
| | - Kiyotaka Nagahama
- Department of Pathology, Kyorin University Faculty of Medicine, Tokyo, Japan
| | - Aya Isomura
- Department of Pathology, Kyorin University Faculty of Medicine, Tokyo, Japan
| | - Akimasa Hayashi
- Department of Pathology, Kyorin University Faculty of Medicine, Tokyo, Japan
| | - Yuki Yamagishi
- Department of Neurosurgery, Kyorin University Faculty of Medicine, Tokyo, Japan
| | - Nobuyoshi Sasaki
- Department of Neurosurgery, Kyorin University Faculty of Medicine, Tokyo, Japan
| | - Keiichi Kobayashi
- Department of Neurosurgery, Kyorin University Faculty of Medicine, Tokyo, Japan
| | - Kazuhiro Tsuchiya
- Department of Radiology, Kyorin University Faculty of Medicine, Tokyo, Japan
| | - Motoo Nagane
- Department of Neurosurgery, Kyorin University Faculty of Medicine, Tokyo, Japan
| | - Koichi Ichimura
- Department of Pathology, Kyorin University Faculty of Medicine, Tokyo, Japan
| | - Junji Shibahara
- Department of Pathology, Kyorin University Faculty of Medicine, Tokyo, Japan
| |
Collapse
|
16
|
Zhou J, Huang J, Zhou Z, Fan R, Deng X, Qiu M, Wu Q, Wang Z. Value of ctDNA in surveillance of adjuvant chemosensitivity and regimen adjustment in stage III colon cancer: a protocol for phase II multicentre randomised controlled trial (REVISE trial). BMJ Open 2025; 15:e090394. [PMID: 39753246 PMCID: PMC11749494 DOI: 10.1136/bmjopen-2024-090394] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/24/2024] [Accepted: 12/16/2024] [Indexed: 01/11/2025] Open
Abstract
INTRODUCTION The standard of care for stage III colon cancer is 3 or 6 months of double-drug regimen chemotherapy following radical surgery. However, patients with positive circulating tumour DNA (ctDNA) exhibit a high risk of recurrence risk even if they receive standard adjuvant chemotherapy. The potential benefit of intensified adjuvant chemotherapy, oxaliplatin, irinotecan, leucovorin and fluoropyrimidine (FOLFOXIRI), for ctDNA-positive patients remains to be elucidated. METHODS AND ANALYSIS This multicentre phase II randomised controlled trial aims to investigate the utility of ctDNA in monitoring chemosensitivity and to preliminarily assess whether intensified chemotherapy with FOLFOXIRI can increase ctDNA clearance and improve survival outcomes. A total of 60 eligible patients with stage III colon cancer exhibiting postoperatively positive ctDNA before and after two cycles of oxaliplatin and capecitabine (XELOX) will be randomly assigned to continue five additional cycles of XELOX (control arm) or switch to eight cycles of FOLFOXIRI (experimental arm). This sequential approach is designed to escalate treatment for patients with persistent ctDNA positivity while avoiding overtreatment in those who may respond well to standard chemotherapy. The primary endpoint is the change in ctDNA concentration, defined as the difference between the ctDNA concentration measured after two cycles of XELOX and after the completion or termination of chemotherapy. Secondary endpoints include the ctDNA clearance rate, 2-year disease-free survival, distant metastasis-free survival, chemotherapy-related side effects and quality of life. ETHICS AND DISSEMINATION This trial has been approved by the Ethics Committee of the West China Hospital, Sichuan University (approval number: 20231998). The findings will be disseminated through peer-reviewed publications and presentations at scientific conferences. TRIAL REGISTRATION NUMBER ClinicalTrials.gov (NCT06242418, registered on 27 January 2024).
Collapse
Affiliation(s)
- Jiahao Zhou
- Colorectal Cancer Center, Department of General Surgery, West China Hospital of Sichuan University, Chengdu, Sichuan, China
| | - Jun Huang
- Colorectal Cancer Center, Department of General Surgery, West China Hospital of Sichuan University, Chengdu, Sichuan, China
| | - Zikai Zhou
- Colorectal Cancer Center, Department of General Surgery, West China Hospital of Sichuan University, Chengdu, Sichuan, China
| | - Rui Fan
- Genecast Biotechnology Co Ltd, Jiangsu, China
| | - Xiangbing Deng
- Colorectal Cancer Center, Department of General Surgery, West China Hospital of Sichuan University, Chengdu, Sichuan, China
| | - Meng Qiu
- Colorectal Cancer Center, Department of General Surgery, West China Hospital of Sichuan University, Chengdu, Sichuan, China
| | - Qingbin Wu
- Colorectal Cancer Center, Department of General Surgery, West China Hospital of Sichuan University, Chengdu, Sichuan, China
| | - Ziqiang Wang
- Colorectal Cancer Center, Department of General Surgery, West China Hospital of Sichuan University, Chengdu, Sichuan, China
| |
Collapse
|
17
|
Kocabey S, Cattin S, Gray I, Rüegg C. Ultrasensitive detection of cancer-associated nucleic acids and mutations by primer exchange reaction-based signal amplification and flow cytometry. Biosens Bioelectron 2025; 267:116839. [PMID: 39369516 DOI: 10.1016/j.bios.2024.116839] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2024] [Revised: 09/27/2024] [Accepted: 10/04/2024] [Indexed: 10/08/2024]
Abstract
The detection of cancer-associated nucleic acids and mutations through liquid biopsy has emerged as a highly promising non-invasive approach for early cancer detection and monitoring. In this study, we report the development of primer exchange reaction (PER) based signal amplification strategy that enables the rapid, sensitive and specific detection of nucleic acids bearing cancer specific single nucleotide mutations using flow cytometry. Using micrometer size beads as support for immobilizing oligonucleotides and programmable PER assembly for target oligonucleotide recognition and fluorescence signal amplification, we demonstrated the versatile detection of target nucleic acids including KRAS oligonucleotide, fragmented mRNAs, and miR-21. Moreover, our detection system can discriminate single base mutations frequently occurred in cancer-associated genes including KRAS, PIK3CA and P53 from cell extracts and circulating tumor DNAs (ctDNAs). The detection is highly sensitive, with a limit of detection down to 27 fM without pre-amplification. In view of a clinical application, we demonstrate the detection of single mutations after extraction and pre-amplification of ctDNAs from the plasma of breast cancer patients. Importantly, our detection strategy enabled the detection of single KRAS mutation even in the presence of 1000-fold excess of wild type (WT) DNA using multi-color flow cytometry detection approach. Overall, our strategy holds immense potential for clinical applications, offering significant improvements for early cancer detection and monitoring.
Collapse
Affiliation(s)
- Samet Kocabey
- Laboratory of Experimental and Translational Oncology, Department of Oncology, Microbiology and Immunology, Faculty of Science and Medicine, University of Fribourg, Chemin Du Musée 18, PER17, 1700, Fribourg, Switzerland; NCCR Bio-inspired Materials, University of Fribourg, 1700, Fribourg, Switzerland.
| | - Sarah Cattin
- Laboratory of Experimental and Translational Oncology, Department of Oncology, Microbiology and Immunology, Faculty of Science and Medicine, University of Fribourg, Chemin Du Musée 18, PER17, 1700, Fribourg, Switzerland; NCCR Bio-inspired Materials, University of Fribourg, 1700, Fribourg, Switzerland; Cell Analytics Facility, Faculty of Science and Medicine, University of Fribourg, Chemin Du Musée 18, PER17, 1700, Fribourg, Switzerland
| | - Isabelle Gray
- Laboratory of Experimental and Translational Oncology, Department of Oncology, Microbiology and Immunology, Faculty of Science and Medicine, University of Fribourg, Chemin Du Musée 18, PER17, 1700, Fribourg, Switzerland; NCCR Bio-inspired Materials, University of Fribourg, 1700, Fribourg, Switzerland
| | - Curzio Rüegg
- Laboratory of Experimental and Translational Oncology, Department of Oncology, Microbiology and Immunology, Faculty of Science and Medicine, University of Fribourg, Chemin Du Musée 18, PER17, 1700, Fribourg, Switzerland; NCCR Bio-inspired Materials, University of Fribourg, 1700, Fribourg, Switzerland.
| |
Collapse
|
18
|
Yoshinami T. Perspectives for the clinical application of ctDNA analysis to breast cancer drug therapy. Breast Cancer 2025; 32:1-9. [PMID: 38649655 DOI: 10.1007/s12282-024-01571-9] [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: 02/21/2024] [Accepted: 03/14/2024] [Indexed: 04/25/2024]
Abstract
Circulating tumor DNA (ctDNA) consists of DNA fragments released from cancer cells into the blood circulation with quick clearance. Analysis of ctDNA can enable real-time assessment of the presence of cancer cells and their genomic characteristics. Therefore, ctDNA is expected to be one of the most useful biomarkers for cancer. In recent years, several ultra-sensitive assays for ctDNA analysis have been developed, and many clinical trials are using these assays to investigate the efficacy of ctDNA-based therapeutic strategies. In the perioperative phase, real-time identification of minimal residual disease at the molecular level with ctDNA analysis can help evaluate the risk of recurrence to inform escalation or de-escalation of perioperative drug therapy. Many trials have examined whether therapeutic strategies using ctDNA analysis to predict treatment efficacy or resistance to molecular targeted agents can improve prognosis in metastatic breast cancer. In this review, we discuss the most recent ctDNA assays, the significance of introducing ctDNA assays to clinical practice, and the research on their application in perioperative and metastatic phases.
Collapse
Affiliation(s)
- Tetsuhiro Yoshinami
- Department of Breast and Endocrine Surgery, Graduate School of Medicine, Osaka University, Osaka, Japan.
| |
Collapse
|
19
|
Ikematsu H, Takara Y, Nishihara K, Kano Y, Owaki Y, Okamoto R, Fujiwara T, Takamatsu T, Yamada M, Tomioka Y, Takeshita N, Inaba A, Sunakawa H, Nakajo K, Murano T, Kadota T, Shinmura K, Koga Y, Yano T. Possibility of determining high quantitative fecal occult blood on stool surface using hyperspectral imaging. J Gastroenterol 2025; 60:77-85. [PMID: 39441401 PMCID: PMC11717890 DOI: 10.1007/s00535-024-02163-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/29/2024] [Accepted: 10/14/2024] [Indexed: 10/25/2024]
Abstract
BACKGROUND Fecal immunochemical tests are commonly performed for colorectal cancer screening. Instant fecal occult blood measurement in toilet bowel movements would improve convenience. Hyperspectral imaging (HSI) enables the nondestructive evaluation of materials that are difficult to assess visually. This study aimed to determine whether HSI could be used to identify fecal occult blood on stool surfaces. METHODS The study included 100 patients who underwent colonoscopy, divided into groups A and B (50 patients, each) for creating a discriminant algorithm and validating the accuracy of the algorithm, respectively. In group A, 100 areas were randomly selected from the stool surface, and the fecal occult blood quantitative values were measured and photographed using a hyperspectral camera (cutoff: > 400 ng/mL). A discriminant algorithm image was created to extract spectral feature differences obtained from HSI via machine learning. In group B, 250 random areas were evaluated and compared to fecal occult blood quantitative values, measuring sensitivity, specificity, accuracy, positive predictive value (PPV), and negative predictive value (NPV). RESULTS Groups A and B comprised 28 and 26 patients with cancer, respectively. Cancer detection sensitivity at the 400 ng/mL cutoff was 67.9% and 42.3% in groups A and B, respectively. The discriminant algorithm image exhibited high accuracy in group A (sensitivity; 77.1%, specificity; 96.9%, accuracy; 90.0%, PPV; 93.1%, NPV; 88.7%). In group B, the sensitivity, specificity, accuracy, PPV, and NPV were 83.3, 92.9, 90.8, 76.3, and 95.3%, respectively. CONCLUSION HSI can effectively discriminate high quantitative fecal occult blood, highlighting its potential for improved colorectal cancer screening.
Collapse
Affiliation(s)
- Hiroaki Ikematsu
- Division of Science and Technology for Endoscopy, Exploratory Oncology Research and Clinical Trial Center, National Cancer Center, Kashiwa, Japan.
- Department of Gastroenterology, IMSUT Hospital, The Institute of Medical Science, The University of Tokyo, 4-6-1 Shirokanedai, Minato-ku, Tokyo, 108-8639, Japan.
| | | | - Keiichiro Nishihara
- Department of Gastroenterology and Endoscopy, National Cancer Center Hospital East, Kashiwa, Japan
| | - Yuki Kano
- Department of Gastroenterology and Endoscopy, National Cancer Center Hospital East, Kashiwa, Japan
| | - Yuji Owaki
- Department of Gastroenterology and Endoscopy, National Cancer Center Hospital East, Kashiwa, Japan
| | - Ryuji Okamoto
- Department of Gastroenterology and Endoscopy, National Cancer Center Hospital East, Kashiwa, Japan
| | - Takahisa Fujiwara
- Department of Gastroenterology and Endoscopy, National Cancer Center Hospital East, Kashiwa, Japan
| | - Toshihiro Takamatsu
- Division of Science and Technology for Endoscopy, Exploratory Oncology Research and Clinical Trial Center, National Cancer Center, Kashiwa, Japan
- Health and Medical Research Institute, National Institute of Advanced Industrial Science and Technology, Tsukuba, Ibaraki, Japan
| | | | - Yutaka Tomioka
- Medical Device Innovation Project Management Office, National Cancer Center Hospital East, Kashiwa, Japan
| | - Nobuyoshi Takeshita
- Division of Medical Device Innovation Support, National Cancer Center Hospital East, Kashiwa, Japan
| | - Atsushi Inaba
- Department of Gastroenterology and Endoscopy, National Cancer Center Hospital East, Kashiwa, Japan
| | - Hironori Sunakawa
- Department of Gastroenterology and Endoscopy, National Cancer Center Hospital East, Kashiwa, Japan
| | - Keiichiro Nakajo
- Department of Gastroenterology and Endoscopy, National Cancer Center Hospital East, Kashiwa, Japan
| | - Tatsuro Murano
- Department of Gastroenterology and Endoscopy, National Cancer Center Hospital East, Kashiwa, Japan
| | - Tomohiro Kadota
- Department of Gastroenterology and Endoscopy, National Cancer Center Hospital East, Kashiwa, Japan
| | - Kensuke Shinmura
- Department of Gastroenterology and Endoscopy, National Cancer Center Hospital East, Kashiwa, Japan
| | - Yoshikatsu Koga
- Department of Strategic Programs, Exploratory Oncology Research and Clinical Trial Center, National Cancer Center, Kashiwa, Japan
| | - Tomonori Yano
- Division of Science and Technology for Endoscopy, Exploratory Oncology Research and Clinical Trial Center, National Cancer Center, Kashiwa, Japan
- Department of Gastroenterology and Endoscopy, National Cancer Center Hospital East, Kashiwa, Japan
| |
Collapse
|
20
|
Zhang W, Ye B, Song Y, Yang P, Si W, Jing H, Yang F, Yuan D, Wu Z, Lyu J, Peng K, Zhang X, Wang L, Li Y, Liu Y, Wu C, Hao X, Zhang Y, Qi W, Wang J, Dong F, Zhao Z, Jing H, Li Y. Integrating multi-omics features enables non-invasive early diagnosis and treatment response prediction of diffuse large B-cell lymphoma. Clin Transl Med 2025; 15:e70174. [PMID: 39776291 PMCID: PMC11705727 DOI: 10.1002/ctm2.70174] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2024] [Revised: 11/13/2024] [Accepted: 12/28/2024] [Indexed: 01/11/2025] Open
Abstract
BACKGROUND Multi-omics features of cell-free DNA (cfDNA) can effectively improve the performance of non-invasive early diagnosis and prognosis of cancer. However, multimodal characterization of cfDNA remains technically challenging. METHODS We developed a comprehensive multi-omics solution (COMOS) to specifically obtain an extensive fragmentomics landscape, presented by breakpoint characteristics of nucleosomes, CpG islands, DNase clusters and enhancers, besides typical methylation, copy number alteration of cfDNA. The COMOS was tested on 214 plasma samples of diffuse large B-cell lymphoma (DLBCL) and matched healthy controls. RESULTS For early diagnosis, COMOS improved the area under the curve (AUC) value to .993 compared with the individual omics model, with a sensitivity of 95% at 98% specificity. Detection sensitivity achieved 91% at 99% specificity in early-stage patients, while the AUC values of the individual omics model were 0.942, 0.968, 0.989, 0.935, 0.921, 0.781 and 0.917, respectively, with lower sensitivity and specificity. In the treatment response cohort, COMOS yielded a superior sensitivity of 88% at 86% specificity (AUC, 0.903). COMOS has achieved excellent performance in early diagnosis and treatment response prediction. CONCLUSIONS Our study provides an effectively improved approach with high accuracy for the diagnosis and prognosis of DLBCL, showing great potential for future clinical application. KEY POINTS A comprehensive multi-omics solution to specifically obtain an extensive fragmentomics landscape, presented by breakpoint characteristics of nucleosomes, CpG islands, DNase clusters and enhancers, besides typical methylation, copy number alteration of cfDNA. Integrated model of cfDNA multi-omics could be used for non-invasive early diagnosis of DLBCL. Integrated model of cfDNA multi-omics could effectively evaluate the efficacy of R-CHOP before DLBCL treatment.
Collapse
MESH Headings
- Humans
- Lymphoma, Large B-Cell, Diffuse/diagnosis
- Lymphoma, Large B-Cell, Diffuse/genetics
- Lymphoma, Large B-Cell, Diffuse/drug therapy
- Lymphoma, Large B-Cell, Diffuse/blood
- Female
- Male
- Middle Aged
- Aged
- Adult
- Early Detection of Cancer/methods
- Prognosis
- Cell-Free Nucleic Acids/blood
- Cell-Free Nucleic Acids/analysis
- Rituximab/therapeutic use
- Biomarkers, Tumor/blood
- Biomarkers, Tumor/genetics
- Doxorubicin/therapeutic use
- Early Diagnosis
- Cyclophosphamide/therapeutic use
- Multiomics
Collapse
Affiliation(s)
- Weilong Zhang
- Department of HematologyLymphoma Research CenterPeking University Third HospitalBeijingChina
| | | | - Yang Song
- BOE Technology Group Co., LtdBeijingChina
| | - Ping Yang
- Department of HematologyLymphoma Research CenterPeking University Third HospitalBeijingChina
| | - Wenzhe Si
- Department of Laboratory MedicinePeking University Third HospitalBeijingChina
| | | | - Fan Yang
- BOE Technology Group Co., LtdBeijingChina
| | - Dan Yuan
- BOE Technology Group Co., LtdBeijingChina
| | - Zhihong Wu
- BOE Technology Group Co., LtdBeijingChina
| | - Jiahao Lyu
- BOE Technology Group Co., LtdBeijingChina
| | - Kang Peng
- BOE Technology Group Co., LtdBeijingChina
| | - Xu Zhang
- Department of HematologyLymphoma Research CenterPeking University Third HospitalBeijingChina
| | - Lingli Wang
- Department of HematologyLymphoma Research CenterPeking University Third HospitalBeijingChina
| | - Yan Li
- Department of HematologyLymphoma Research CenterPeking University Third HospitalBeijingChina
| | - Yan Liu
- Department of HematologyLymphoma Research CenterPeking University Third HospitalBeijingChina
| | - Chaoling Wu
- Department of HematologyLymphoma Research CenterPeking University Third HospitalBeijingChina
| | - Xiaoyu Hao
- Department of HematologyLymphoma Research CenterPeking University Third HospitalBeijingChina
| | - Yuqi Zhang
- Department of HematologyLymphoma Research CenterPeking University Third HospitalBeijingChina
| | - Wenxin Qi
- Department of HematologyLymphoma Research CenterPeking University Third HospitalBeijingChina
| | - Jing Wang
- Department of HematologyLymphoma Research CenterPeking University Third HospitalBeijingChina
| | - Fei Dong
- Department of HematologyLymphoma Research CenterPeking University Third HospitalBeijingChina
| | | | - Hongmei Jing
- Department of HematologyLymphoma Research CenterPeking University Third HospitalBeijingChina
| | - Yanzhao Li
- BOE Technology Group Co., LtdBeijingChina
| |
Collapse
|
21
|
Ohmura H, Hanamura F, Okumura Y, Ando Y, Masuda T, Mimori K, Akashi K, Baba E. Liquid biopsy for breast cancer and other solid tumors: a review of recent advances. Breast Cancer 2025; 32:33-42. [PMID: 38492205 DOI: 10.1007/s12282-024-01556-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2023] [Accepted: 02/13/2024] [Indexed: 03/18/2024]
Abstract
Liquid biopsy using circulating tumor DNA (ctDNA) has been reported to be less invasive and effective for comprehensive genetic analysis of heterogeneous solid tumors, including decision-making for therapeutic strategies, predicting recurrence, and detecting genetic factors related to treatment resistance in various types of cancers. Breast cancer, colorectal cancer, and lung cancer are among the most prevalent malignancies worldwide, and clinical studies of liquid biopsy for these cancers are ongoing. Liquid biopsy has been used as a companion diagnostic tool in clinical settings, and research findings have accumulated, especially in cases of colorectal cancer after curative resection and non-small cell lung cancer (NSCLC) after curative chemoradiotherapy, in which ctDNA detection helps predict eligibility for adjuvant chemotherapy. Liquid biopsy using ctDNA shows promise across a wide range of cancer types, including breast cancer, and its clinical applications are expected to expand further through ongoing research. In this article, studies on liquid biopsy in breast cancer, colorectal cancer, and NSCLC are compared focusing on ctDNA.
Collapse
Affiliation(s)
- Hirofumi Ohmura
- Department of Oncology and Social Medicine, Graduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-Ku, Fukuoka, 812-8582, Japan
- Department of Internal Medicine, Kyushu University Beppu Hospital, Oita, Japan
| | - Fumiyasu Hanamura
- Department of Internal Medicine, Kyushu University Beppu Hospital, Oita, Japan
| | - Yuta Okumura
- Department of Internal Medicine, Kyushu University Beppu Hospital, Oita, Japan
- Department of Gastrointestinal and Medical Oncology, National Hospital Organization Kyushu Cancer Center, Fukuoka, Japan
| | - Yuki Ando
- Department of Surgery, Kyushu University Beppu Hospital, Oita, Japan
| | - Takaaki Masuda
- Department of Surgery, Kyushu University Beppu Hospital, Oita, Japan
| | - Koshi Mimori
- Department of Surgery, Kyushu University Beppu Hospital, Oita, Japan
| | - Koichi Akashi
- Department of Medicine and Biosystemic Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Eishi Baba
- Department of Oncology and Social Medicine, Graduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-Ku, Fukuoka, 812-8582, Japan.
| |
Collapse
|
22
|
Iden CR, Mustafa SM, Øgaard N, Henriksen T, Jensen SØ, Ahlborn LB, Egebjerg K, Baeksgaard L, Garbyal RS, Nedergaard MK, Achiam MP, Andersen CL, Mau-Sørensen M. Circulating tumor DNA predicts recurrence and survival in patients with resectable gastric and gastroesophageal junction cancer. Gastric Cancer 2025; 28:83-95. [PMID: 39369091 PMCID: PMC11706848 DOI: 10.1007/s10120-024-01556-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/27/2024] [Accepted: 09/23/2024] [Indexed: 10/07/2024]
Abstract
BACKGROUND Gastric and gastroesophageal junction (GEJ) cancer represents a significant global health challenge, with high recurrence rates and poor survival outcomes. This study investigates circulating tumor DNA (ctDNA) as a biomarker for assessing recurrence risk in patients with resectable gastric and GEJ adenocarcinomas (AC). METHODS Patients with resectable gastric and GEJ AC, undergoing perioperative chemotherapy and surgery, were prospectively enrolled. Serial plasma samples were collected at baseline, after one cycle of chemotherapy, after preoperative chemotherapy, and after surgery. ctDNA was assessed by a ddPCR test (TriMeth), which targets the gastrointestinal cancer-specific methylation patterns of the genes C9orf50, KCNQ5, and CLIP4. RESULTS ctDNA analysis was performed on 229 plasma samples from 86 patients. At baseline, ctDNA was detected in 56% of patients, which decreased to 37% following one cycle of chemotherapy, 25% after preoperative chemotherapy and 15% after surgical resection. The presence of ctDNA after one cycle of chemotherapy was associated with reduced recurrence-free survival (RFS) (HR = 2.54, 95% confidence interval (CI) 1.33-4.85, p = 0.005) and overall survival (OS) (HR = 2.23, 95% CI 1.07-4.62, p = 0.032). Similarly, ctDNA after surgery was associated with significantly shorter RFS (HR = 6.22, 95% CI 2.39-16.2, p < 0.001) and OS (HR = 6.37, 95% CI 2.10-19.3, p = 0.001). Multivariable regression analysis confirmed ctDNA after surgery as an independent prognostic factor (p < 0.001). CONCLUSION ctDNA analysis has the potential to identify patients at elevated risk of recurrence, thus providing personalized treatment strategies for patients with resectable gastric and GEJ cancer. Further validation in larger cohorts and ctDNA-guided interventions are needed for future clinical use.
Collapse
Affiliation(s)
- Cecilie Riis Iden
- Department of Oncology, Copenhagen University Hospital, Rigshospitalet, Blegdamsvej 9, 2100, Copenhagen, Denmark
| | - Salah Mohammad Mustafa
- Department of Molecular Medicine, Aarhus University Hospital, Palle Juul-Jensens, Boulevard 99, 8200, Aarhus N, Denmark
- Institute of Clinical Medicine, Faculty of Health, Aarhus University, Palle Juul-Jensens Boulevard 82, 8200, Aarhus N, Denmark
| | - Nadia Øgaard
- Department of Molecular Medicine, Aarhus University Hospital, Palle Juul-Jensens, Boulevard 99, 8200, Aarhus N, Denmark
- Institute of Clinical Medicine, Faculty of Health, Aarhus University, Palle Juul-Jensens Boulevard 82, 8200, Aarhus N, Denmark
| | - Tenna Henriksen
- Department of Molecular Medicine, Aarhus University Hospital, Palle Juul-Jensens, Boulevard 99, 8200, Aarhus N, Denmark
- Institute of Clinical Medicine, Faculty of Health, Aarhus University, Palle Juul-Jensens Boulevard 82, 8200, Aarhus N, Denmark
| | - Sarah Østrup Jensen
- Department of Molecular Medicine, Aarhus University Hospital, Palle Juul-Jensens, Boulevard 99, 8200, Aarhus N, Denmark
- Institute of Clinical Medicine, Faculty of Health, Aarhus University, Palle Juul-Jensens Boulevard 82, 8200, Aarhus N, Denmark
| | - Lise Barlebo Ahlborn
- Department of Genomic Medicine, Copenhagen University Hospital, Rigshospitalet, Blegdamsvej 9, 2100, Copenhagen, Denmark
| | - Kristian Egebjerg
- Department of Oncology, Copenhagen University Hospital, Rigshospitalet, Blegdamsvej 9, 2100, Copenhagen, Denmark
| | - Lene Baeksgaard
- Department of Oncology, Copenhagen University Hospital, Rigshospitalet, Blegdamsvej 9, 2100, Copenhagen, Denmark
| | - Rajendra Singh Garbyal
- Department of Pathology, Copenhagen University Hospital, Rigshospitalet, Blegdamsvej 9, 2100, Copenhagen, Denmark
| | - Mette Kjølhede Nedergaard
- Department of Pathology, Copenhagen University Hospital, Rigshospitalet, Blegdamsvej 9, 2100, Copenhagen, Denmark
| | - Michael Patrick Achiam
- Department of Surgery & Transplantation, Copenhagen University Hospital, Rigshospitalet, Blegdamsvej 9, 2100, Copenhagen, Denmark
| | - Claus Lindbjerg Andersen
- Department of Molecular Medicine, Aarhus University Hospital, Palle Juul-Jensens, Boulevard 99, 8200, Aarhus N, Denmark
- Institute of Clinical Medicine, Faculty of Health, Aarhus University, Palle Juul-Jensens Boulevard 82, 8200, Aarhus N, Denmark
| | - Morten Mau-Sørensen
- Department of Oncology, Copenhagen University Hospital, Rigshospitalet, Blegdamsvej 9, 2100, Copenhagen, Denmark.
| |
Collapse
|
23
|
Jee J. Circulating tumor DNA: the dawn of new, clinically scalable biomarkers for thromboembolism. J Thromb Haemost 2025; 23:39-42. [PMID: 39798969 DOI: 10.1016/j.jtha.2024.09.031] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2024] [Accepted: 09/26/2024] [Indexed: 01/15/2025]
Affiliation(s)
- Justin Jee
- Department of Medicine, Memorial Sloan Kettering Cancer Center.
| |
Collapse
|
24
|
Peng X, Zheng J, Liu T, Zhou Z, Song C, Zhang D, Zhang X, Huang Y. DNA Methylation-Based Diagnosis and Treatment of Breast Cancer. Curr Cancer Drug Targets 2025; 25:26-37. [PMID: 38441008 DOI: 10.2174/0115680096278978240204162353] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2023] [Revised: 01/01/2024] [Accepted: 01/12/2024] [Indexed: 03/06/2024]
Abstract
DNA methylation is a key epigenetic modifier involved in tumor formation, invasion, and metastasis. The development of breast cancer is a complex process, and many studies have now confirmed the involvement of DNA methylation in breast cancer. Moreover, the number of genes identified as aberrantly methylated in breast cancer is rapidly increasing, and the accumulation of epigenetic alterations becomes a chronic factor in the development of breast cancer. The combined effects of external environmental factors and the internal tumor microenvironment promote epigenetic alterations that drive tumorigenesis. This article focuses on the relevance of DNA methylation to breast cancer, describing the role of detecting DNA methylation in the early diagnosis, prediction, progression, metastasis, treatment, and prognosis of breast cancer, as well as recent advances. The reversibility of DNA methylation is utilized to target specific methylation aberrant promoters as well as related enzymes, from early prevention to late targeted therapy, to understand the journey of DNA methylation in breast cancer with a more comprehensive perspective. Meanwhile, methylation inhibitors in combination with other therapies have a wide range of prospects, providing hope to drug-resistant breast cancer patients.
Collapse
Affiliation(s)
- Xintong Peng
- School of Clinical Medicine, Affiliated Weifang Medical University, Weifang, China
| | - Jingfan Zheng
- School of Clinical Medicine, Affiliated Weifang Medical University, Weifang, China
| | - Tianzi Liu
- School of Clinical Medicine, Affiliated Weifang Medical University, Weifang, China
| | - Ziwen Zhou
- School of Clinical Medicine, Affiliated Weifang Medical University, Weifang, China
| | - Chen Song
- School of Clinical Medicine, Affiliated Weifang Medical University, Weifang, China
| | - Danyan Zhang
- School of Clinical Medicine, Affiliated Weifang Medical University, Weifang, China
| | - Xinlong Zhang
- School of Clinical Medicine, Affiliated Weifang Medical University, Weifang, China
| | - Yan Huang
- Department of Oncology, Affiliated Hospital of Weifang Medical University, Weifang, China
| |
Collapse
|
25
|
Lee SH, Yim SY, Kim JH, Lee SS, Kaseb AO, Lee JS. Correspondence to editorial 1 on "Genomic biomarkers to predict response to atezolizumab plus bevacizumab immunotherapy in hepatocellular carcinoma: insights from the IMbrave150 Trial". Clin Mol Hepatol 2025; 31:e81-e83. [PMID: 39370716 DOI: 10.3350/cmh.2024.0829] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/23/2024] [Accepted: 10/02/2024] [Indexed: 10/08/2024] Open
Affiliation(s)
- Sung Hwan Lee
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, CHA Bundang Medical Center, CHA University, Seongnam, Korea
| | - Sun Young Yim
- Department of Internal Medicine, Korea University College of Medicine, Seoul, Korea
| | - Ji Hoon Kim
- Department of Internal Medicine, Korea University College of Medicine, Seoul, Korea
| | - Sunyoung S Lee
- Department of Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Ahmed O Kaseb
- Department of Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Ju-Seog Lee
- Department of Systems Biology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| |
Collapse
|
26
|
Yazdi ZF, Roshannezhad S, Sharif S, Abbaszadegan MR. Recent progress in prompt molecular detection of liquid biopsy using Cas enzymes: innovative approaches for cancer diagnosis and analysis. J Transl Med 2024; 22:1173. [PMID: 39741289 DOI: 10.1186/s12967-024-05908-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2024] [Accepted: 11/20/2024] [Indexed: 01/02/2025] Open
Abstract
Creating fast, non-invasive, precise, and specific diagnostic tests is crucial for enhancing cancer treatment outcomes. Among diagnostic methods, those relying on nucleic acid detection are highly sensitive and specific. Recent developments in diagnostic technologies, particularly those leveraging Clustered Regularly Interspaced Short Palindromic Repeats (CRISPR), are revolutionizing cancer detection, providing accurate and timely results. In clinical oncology, liquid biopsy has become a noninvasive and early-detectable alternative to traditional biopsies over the last two decades. Analyzing the nucleic acid content of liquid biopsy samples, which include Circulating Tumor Cells (CTCs), Circulating Tumor DNA (ctDNA), Circulating Cell-Free RNA (cfRNA), and tumor extracellular vesicles, provides a noninvasive method for cancer detection and monitoring. In this review, we explore how the characteristics of various Cas (CRISPR-associated) enzymes have been utilized in diagnostic assays for cancer liquid biopsy and highlight their main applications of innovative approaches in monitoring, as well as early and rapid detection of cancers.
Collapse
Affiliation(s)
- Zahra Farshchian Yazdi
- Department of Medical Genetics, Faculty of Medicine, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| | | | - Samaneh Sharif
- Medical Genetics Research Center, Mashhad University of Medical Sciences, Mashhad, Iran.
- Department of Medical Genetics, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran.
- Mashhad University of Medical Sciences, Azadi Square, Mashhad, Iran.
| | - Mohammad Reza Abbaszadegan
- Department of Medical Genetics, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran.
- Immunology Research Center, Mashhad University of Medical Sciences, Mashhad, Iran.
- Mashhad University of Medical Sciences, Azadi Square, Mashhad, Iran.
| |
Collapse
|
27
|
Du T, Yuan Y, Sun S, Gao Z, Li X. Integrating traditional biomarkers and emerging predictors to assess neoadjuvant chemotherapy efficacy in breast cancer: a multifactorial analysis of Ki-67, CDK4, EGFR, TILs and ctDNA. BMC Womens Health 2024; 24:674. [PMID: 39736579 DOI: 10.1186/s12905-024-03486-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2024] [Accepted: 11/28/2024] [Indexed: 01/01/2025] Open
Abstract
OBJECTIVE This study aimed to analyse the correlation between the expression of cell proliferation-associated antigen (Ki-67), cell cycle protein-dependent kinase 4 (CDK4), epidermal growth factor receptor (EGFR), tumour-infiltrating lymphocytes (TILs) and circulating tumour DNA (ctDNA) with the outcome and prognosis of patients with breast cancer (BC) undergoing neoadjuvant chemotherapy (NACT). METHODS We retrospectively analysed the clinicopathological data of 231 patients with BC who underwent preoperative NACT at XX Hospital between 1 January 2018 and 31 December 2021. Logistic regression models were used to analyse factors influencing NACT efficacy. The Cox risk regression model was used to analyse prognostic factors. The TILs were assessed on pre-treatment biopsies, and ctDNA levels were monitored during NACT. Propensity score matching and subgroup analyses were performed. RESULTS After 4-6 cycles of chemotherapy, the response rate was 77.92% (180/231), with 58.87% (136/231) achieving pathological complete response (pCR). Multifactorial analysis showed that tumour, node and metastasis (TNM) stage II, EGFR positivity, low Ki-67 expression, CDK4 negativity, non-triple-negative subtypes and effective NACT results were associated with higher pCR rates. Higher TIL levels correlated with increased pCR rates (72.4% for high TILs vs 39.1% for low TILs, p < 0.001). The ctDNA levels decreased significantly in patients with pCR compared with patients without pCR during NACT (p < 0.001). After propensity score matching, the 3-year disease-free survival rate was significantly higher in the pCR group (88.9% vs 71.1%, p = 0.003). Subgroup analysis revealed varying pCR rates and predictive biomarkers across BC subtypes. CONCLUSION The TNM classification, EGFR, Ki-67, CDK4 expression, BC subtype and NACT results have predictive value for pCR in patients with BC. Lower TNM classification, lower Ki-67 expression and EGFR positivity are associated with better outcomes. High TIL levels and significant decreases in ctDNA during NACT correlate with improved response and prognosis. These findings highlight the potential for integrating traditional clinicopathological factors with novel biomarkers for personalised treatment strategies in BC.
Collapse
Affiliation(s)
- Tianzhao Du
- Central Laboratory, Cancer Hospital of China Medical University, Dalian Medical University Clinical Oncology College, Liaoning Cancer Hospital & Institute, Shenyang, Liaoning, 110042, People's Republic of China
| | - Ye Yuan
- Central Laboratory, Cancer Hospital of China Medical University, Dalian Medical University Clinical Oncology College, Liaoning Cancer Hospital & Institute, Shenyang, Liaoning, 110042, People's Republic of China
| | - Shulan Sun
- Central Laboratory, Cancer Hospital of China Medical University, Dalian Medical University Clinical Oncology College, Liaoning Cancer Hospital & Institute, Shenyang, Liaoning, 110042, People's Republic of China.
| | - Zhichao Gao
- Central Laboratory, Cancer Hospital of China Medical University, Dalian Medical University Clinical Oncology College, Liaoning Cancer Hospital & Institute, Shenyang, Liaoning, 110042, People's Republic of China
| | - Xiaoshuai Li
- Department of Blood Transfusion, Shengjing Hospital of China Medical University, Shenyang, 110004, China
| |
Collapse
|
28
|
Noji R, Tohyama K, Nakamura S, Naito T, Oikawa Y, Kuroshima T, Tomioka H, Michi Y, Ikeda S, Asakage T, Miura M, Hamamoto Y, Harada H, Kano Y. Dynamic Changes in Circulating Tumor DNA During Immunotherapy for Head and Neck Cancer: SHIZUKU-HN Study. Int J Mol Sci 2024; 26:235. [PMID: 39796090 PMCID: PMC11719933 DOI: 10.3390/ijms26010235] [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: 11/29/2024] [Revised: 12/25/2024] [Accepted: 12/27/2024] [Indexed: 01/13/2025] Open
Abstract
Immune checkpoint inhibitors (ICIs) are effective in treating recurrent/metastatic head and neck squamous cell carcinoma (HNSCC), but only 20% of patients achieve durable responses. This study evaluated circulating tumor DNA (ctDNA) as a real-time biomarker for monitoring treatment response in HNSCC. The SHIZUKU-HN study prospectively collected and analyzed serial plasma samples (n = 27) from HNSCC patients undergoing ICIs, using Guardant360 to assess ctDNA variant allele frequency (VAF) and genetic mutations. Tumor volumes were quantified using 3D reconstruction of CT scans, and data from Japan's C-CAT database (n = 2255) provided insights into ctDNA testing in HNSCC. C-CAT data showed that ctDNA testing was underutilized, performed in only 7% of head and neck cancer cases. In SHIZUKU-HN, mean VAF significantly correlated with tumor volume (Spearman's ρ = 0.70, p = 0.001), often preceding radiographic progression. BRAF and APC mutations disappeared in partial responders, while GNAS mutations varied. EGFR and PIK3CA amplifications, detectable via ctDNA but missed in tissue biopsies, indicated emerging resistance mechanisms. The SHIZUKU-HN study demonstrates the potential of ctDNA as a dynamic biomarker in HNSCC, offering early insights into treatment efficacy and informing personalized ICI therapy.
Collapse
Affiliation(s)
- Rika Noji
- Department of Oral and Maxillofacial Surgical Oncology, Division of Health Science, Graduate School of Medical and Dental Sciences, Institute of Science Tokyo, 1-5-45 Yushima, Bunkyo-Ku, Tokyo 113-8510, Japan; (R.N.)
- Department of Medical Oncology, Institute of Science Tokyo, 1-5-45 Yushima, Bunkyo-Ku, Tokyo 113-8510, Japan
| | - Kohki Tohyama
- Department of Dental Radiology and Radiation Oncology, Graduate School of Medical and Dental Sciences, Institute of Science Tokyo, 1-5-45 Yushima, Bunkyo-Ku, Tokyo 113-8510, Japan
| | - Shin Nakamura
- Department of Dental Radiology and Radiation Oncology, Graduate School of Medical and Dental Sciences, Institute of Science Tokyo, 1-5-45 Yushima, Bunkyo-Ku, Tokyo 113-8510, Japan
| | - Takahiro Naito
- Department of Oral and Maxillofacial Surgical Oncology, Division of Health Science, Graduate School of Medical and Dental Sciences, Institute of Science Tokyo, 1-5-45 Yushima, Bunkyo-Ku, Tokyo 113-8510, Japan; (R.N.)
- Department of Medical Oncology, Institute of Science Tokyo, 1-5-45 Yushima, Bunkyo-Ku, Tokyo 113-8510, Japan
| | - Yu Oikawa
- Department of Oral and Maxillofacial Surgical Oncology, Division of Health Science, Graduate School of Medical and Dental Sciences, Institute of Science Tokyo, 1-5-45 Yushima, Bunkyo-Ku, Tokyo 113-8510, Japan; (R.N.)
| | - Takeshi Kuroshima
- Department of Oral and Maxillofacial Surgical Oncology, Division of Health Science, Graduate School of Medical and Dental Sciences, Institute of Science Tokyo, 1-5-45 Yushima, Bunkyo-Ku, Tokyo 113-8510, Japan; (R.N.)
| | - Hirofumi Tomioka
- Department of Oral and Maxillofacial Surgical Oncology, Division of Health Science, Graduate School of Medical and Dental Sciences, Institute of Science Tokyo, 1-5-45 Yushima, Bunkyo-Ku, Tokyo 113-8510, Japan; (R.N.)
| | - Yasuyuki Michi
- Department of Oral and Maxillofacial Surgical Oncology, Division of Health Science, Graduate School of Medical and Dental Sciences, Institute of Science Tokyo, 1-5-45 Yushima, Bunkyo-Ku, Tokyo 113-8510, Japan; (R.N.)
| | - Sadakatsu Ikeda
- Department of Precision Cancer Medicine, Center for Innovative Cancer Treatment, Institute of Science Tokyo, 1-5-45 Yushima, Bunkyo-Ku, Tokyo 113-8510, Japan
| | - Takahiro Asakage
- Department of Head and Neck Surgery, Institute of Science Tokyo 1-5-45 Yushima, Bunkyo-Ku, Tokyo 113-8510, Japan
| | - Masahiko Miura
- Department of Dental Radiology and Radiation Oncology, Graduate School of Medical and Dental Sciences, Institute of Science Tokyo, 1-5-45 Yushima, Bunkyo-Ku, Tokyo 113-8510, Japan
| | - Yasuo Hamamoto
- Department of Medical Oncology, Institute of Science Tokyo, 1-5-45 Yushima, Bunkyo-Ku, Tokyo 113-8510, Japan
| | - Hiroyuki Harada
- Department of Oral and Maxillofacial Surgical Oncology, Division of Health Science, Graduate School of Medical and Dental Sciences, Institute of Science Tokyo, 1-5-45 Yushima, Bunkyo-Ku, Tokyo 113-8510, Japan; (R.N.)
| | - Yoshihito Kano
- Department of Medical Oncology, Institute of Science Tokyo, 1-5-45 Yushima, Bunkyo-Ku, Tokyo 113-8510, Japan
| |
Collapse
|
29
|
van Zogchel LMJ, Decarolis B, van Wezel EM, Zappeij-Kannegieter L, Gelineau NU, Schumacher-Kuckelkorn R, Simon T, Berthold F, van Noesel MM, Fiocco M, van der Schoot CE, Hero B, Stutterheim J, Tytgat GAM. Sensitive liquid biopsy monitoring correlates with outcome in the prospective international GPOH-DCOG high-risk neuroblastoma RT-qPCR validation study. J Exp Clin Cancer Res 2024; 43:331. [PMID: 39722049 DOI: 10.1186/s13046-024-03261-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] [Received: 11/06/2024] [Accepted: 12/18/2024] [Indexed: 12/28/2024] Open
Abstract
BACKGROUND Liquid biopsies offer less burdensome sensitive disease monitoring. Bone marrow (BM) metastases, common in various cancers including neuroblastoma, is associated with poor outcomes. In pediatric high-risk neuroblastoma most patients initially respond to treatment, but in the majority the disease recurs with only 40% long-term survivors, stressing the need for more sensitive detection of disseminated disease during therapy. METHODS To validate sensitive neuroblastoma mRNA RT-qPCR BM testing, we prospectively assessed serial BM samples from 345 international high-risk neuroblastoma patients, treated in trials NB2004 (GPOH) or NBL2009 (DCOG), using PHOX2B, TH, DDC, CHRNA3, and GAP43 RT-qPCR mRNA markers and BM GD2-immunocytology. Association between BM-infiltration levels and event-free survival (EFS) and overall survival (OS) was estimated by using Cox regression models and Kaplan-Meier's methodology. RESULTS BM infiltration >10% by RT-qPCR at diagnosis was prognostic for survival (adjusted hazard ratio (HR) 1.82 [95%CI 1.25-2.63] and 2.04 [1.33-3.14] for EFS and OS, respectively). Any post-induction RT-qPCR positivity correlated with poor EFS and OS, with a HR of 2.10 [1.27-3.49] and 1.76 [1.01-3.08] and 5-years EFS of 26.6% [standard error 5.2%] versus 60.4% [6.7] and OS of 43.8% [5.9] versus 65.7% [6.6] for RT-qPCR-positive patients versus RT-qPCR-negative patients. In contrast, post-induction immunocytology positivity was not associated with EFS or OS (HR 1.22 [0.68-2.19] and 1.26 [0.54-2.42]). CONCLUSION This study validates the association of not clearing of BM metastases by sensitive RT-qPCR detection with very poor outcome. We therefore propose implementation of RT-qPCR for minimal residual disease testing in neuroblastoma to guide therapy.
Collapse
Affiliation(s)
- Lieke M J van Zogchel
- Princess Máxima Center for Pediatric Oncology, Utrecht, The Netherlands
- Department of Experimental Immunohematology, Sanquin Research and Landsteiner Laboratory of the Amsterdam UMC, Amsterdam, The Netherlands
| | - Boris Decarolis
- Department of Pediatric Oncology and Hematology, University Children's Hospital of Cologne, and Medical Faculty, University of Cologne, Köln, Germany
| | - Esther M van Wezel
- Department of Experimental Immunohematology, Sanquin Research and Landsteiner Laboratory of the Amsterdam UMC, Amsterdam, The Netherlands
| | - Lily Zappeij-Kannegieter
- Department of Immunocytology, Sanquin Research and Landsteiner Laboratory of the Amsterdam UMC, Amsterdam, The Netherlands
| | - Nina U Gelineau
- Princess Máxima Center for Pediatric Oncology, Utrecht, The Netherlands
- Department of Experimental Immunohematology, Sanquin Research and Landsteiner Laboratory of the Amsterdam UMC, Amsterdam, The Netherlands
| | - Roswitha Schumacher-Kuckelkorn
- Department of Pediatric Oncology and Hematology, University Children's Hospital of Cologne, and Medical Faculty, University of Cologne, Köln, Germany
| | - Thorsten Simon
- Department of Pediatric Oncology and Hematology, University Children's Hospital of Cologne, and Medical Faculty, University of Cologne, Köln, Germany
| | - Frank Berthold
- Department of Pediatric Oncology and Hematology, University Children's Hospital of Cologne, and Medical Faculty, University of Cologne, Köln, Germany
| | - Max M van Noesel
- Princess Máxima Center for Pediatric Oncology, Utrecht, The Netherlands
- Division Imaging & Oncology, University Medical Center Utrecht, Utrecht, the Netherlands
| | - Marta Fiocco
- Princess Máxima Center for Pediatric Oncology, Utrecht, The Netherlands
- Mathematical Institute, Leiden University, Leiden, The Netherlands
- Department of Biomedical data Science, Section Medical Statistics, Leiden University Medical Center, Leiden, The Netherlands
| | - C Ellen van der Schoot
- Department of Experimental Immunohematology, Sanquin Research and Landsteiner Laboratory of the Amsterdam UMC, Amsterdam, The Netherlands
| | - Barbara Hero
- Department of Pediatric Oncology and Hematology, University Children's Hospital of Cologne, and Medical Faculty, University of Cologne, Köln, Germany
| | | | - Godelieve A M Tytgat
- Princess Máxima Center for Pediatric Oncology, Utrecht, The Netherlands.
- Department of Experimental Immunohematology, Sanquin Research and Landsteiner Laboratory of the Amsterdam UMC, Amsterdam, The Netherlands.
- Department of Genetics, Utrecht University Medical Center, Utrecht, the Netherlands.
| |
Collapse
|
30
|
Lu Y, Wang Z, Zhang D, Luo N, Yang H, Chen D, Huang H. Application of Circulating Tumor DNA in the Auxiliary Diagnosis and Prognosis Prediction of Glioma. Cell Mol Neurobiol 2024; 45:6. [PMID: 39692767 DOI: 10.1007/s10571-024-01515-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2024] [Accepted: 11/12/2024] [Indexed: 12/19/2024]
Abstract
Glioma is the most common primary malignant brain tumor. Despite significant advances in the past decade in understanding the molecular pathogenesis of this tumor and exploring therapeutic strategies, the prognosis of patients with glioma remains poor. Accurate diagnosis of glioma is very important for the treatment and prognosis. Although the gold-standard method for the diagnosis and prognosis prediction of patients with glioma is tissue biopsy, it still has many limitations. Liquid biopsy can provide information on the auxiliary diagnosis and prognosis of gliomas. In this review, we summarized the application of cell-free DNA (cfDNA) and circulating tumor DNA (ctDNA) in the auxiliary diagnosis and prognosis of glioma. The common methods used to detect ctDNA in gliomas using samples including blood and cerebrospinal fluid (CSF) and the detection techniques for ctDNA, including droplet digital PCR (ddPCR) and next-generation sequencing (NGS), were discussed. Detection of ctDNA from plasma of patients with brain tumors remains challenging because of the blood-brain barrier (BBB). CSF has been proposed as a medium for ctDNA analysis in brain tumors, and mutation detection using plasma ctDNA was less sensitive than CSF ctDNA sequencing. Moreover, ongoing relevant clinical studies were summarized. Finally, we discussed the challenges, and future directions for the studies on ctDNA in glioma.
Collapse
Affiliation(s)
- Ying Lu
- Department of Oncology, The Fourth Affiliated Hospital of Guangxi Medical University, Liuzhou, 545000, China
| | - Zhouyu Wang
- The State Key Laboratory of Neurology and Oncology Drug Development, Jiangsu Simcere Diagnostics Co., Ltd, Nanjing Simcere Medical Laboratory Science Co., Ltd, Nanjing, 210002, China
| | - Danmeng Zhang
- Department of Oncology, The Fourth Affiliated Hospital of Guangxi Medical University, Liuzhou, 545000, China
| | - Ningning Luo
- The State Key Laboratory of Neurology and Oncology Drug Development, Jiangsu Simcere Diagnostics Co., Ltd, Nanjing Simcere Medical Laboratory Science Co., Ltd, Nanjing, 210002, China
| | - Hui Yang
- Department of Oncology, The Fourth Affiliated Hospital of Guangxi Medical University, Liuzhou, 545000, China
| | - Dongsheng Chen
- The State Key Laboratory of Neurology and Oncology Drug Development, Jiangsu Simcere Diagnostics Co., Ltd, Nanjing Simcere Medical Laboratory Science Co., Ltd, Nanjing, 210002, China.
- Cancer Center, The First Affiliated Hospital of Jinzhou Medical University, Jinzhou, 121001, China.
- Center of Translational Medicine, The First Affiliated Hospital of Jinzhou Medical University, Jinzhou, 121001, China.
| | - Haixin Huang
- Department of Oncology, The Fourth Affiliated Hospital of Guangxi Medical University, Liuzhou, 545000, China.
| |
Collapse
|
31
|
Kohabir KAV, Linthorst J, Nooi LO, Brouwer R, Wolthuis RMF, Sistermans EA. Synthetic mismatches enable specific CRISPR-Cas12a-based detection of genome-wide SNVs tracked by ARTEMIS. CELL REPORTS METHODS 2024; 4:100912. [PMID: 39644903 PMCID: PMC11704620 DOI: 10.1016/j.crmeth.2024.100912] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/07/2024] [Revised: 09/13/2024] [Accepted: 11/12/2024] [Indexed: 12/09/2024]
Abstract
Detection of pathogenic DNA variants is vital in cancer diagnostics and treatment monitoring. While CRISPR-based diagnostics (CRISPRdx) offer promising avenues for cost-effective, rapid, and point-of-care testing, achieving single-nucleotide detection fidelity remains challenging. We present an in silico pipeline that scans the human genome for targeting pathogenic mutations in the seed region (ARTEMIS), the most stringent crRNA domain. ARTEMIS identified 12% of pathogenic SNVs as Cas12a recognizable, including 928 cancer-associated variants such as BRAFV600E, BRCA2E1953∗, TP53V272M, and ALDH2E504K. Cas12a exhibited remarkable tolerance to single mismatches within the seed region. Introducing deliberate synthetic mismatches within the seed region yielded on-target activity with single-nucleotide fidelity. Both positioning and nucleobase types of mismatches influenced detection accuracy. With improved specificity, Cas12a could accurately detect and semi-quantify BRAFV600E in cfDNA from cell lines and patient liquid biopsies. These results provide insights toward rationalized crRNA design for high-fidelity CRISPRdx, supporting personalized and cost-efficient healthcare solutions in oncologic diagnostics.
Collapse
Affiliation(s)
- Kavish A V Kohabir
- Department of Human Genetics, Amsterdam UMC location Vrije Universiteit Amsterdam, Amsterdam, the Netherlands; Amsterdam Reproduction & Development, Amsterdam, the Netherlands; Imaging and Biomarkers, Cancer Center Amsterdam, Amsterdam, the Netherlands
| | - Jasper Linthorst
- Department of Human Genetics, Amsterdam UMC location Vrije Universiteit Amsterdam, Amsterdam, the Netherlands; Amsterdam Reproduction & Development, Amsterdam, the Netherlands; Amsterdam Institute for Immunology and Infectious Diseases, Amsterdam, the Netherlands
| | - Lars O Nooi
- Department of Human Genetics, Amsterdam UMC location Vrije Universiteit Amsterdam, Amsterdam, the Netherlands
| | - Rick Brouwer
- Clinical Laboratory, Unilabs, Enschede, the Netherlands
| | - Rob M F Wolthuis
- Department of Human Genetics, Amsterdam UMC location Vrije Universiteit Amsterdam, Amsterdam, the Netherlands; Cancer Biology and Immunology, Cancer Center Amsterdam, Amsterdam, the Netherlands.
| | - Erik A Sistermans
- Department of Human Genetics, Amsterdam UMC location Vrije Universiteit Amsterdam, Amsterdam, the Netherlands; Amsterdam Reproduction & Development, Amsterdam, the Netherlands.
| |
Collapse
|
32
|
Huang T, Li J, Chen H, Sun H, Jang DW, Alam MM, Yeung KK, Zhang Q, Xia H, Duan L, Mao C, Gao Z. Rapid miRNA detection enhanced by exponential hybridization chain reaction in graphene field-effect transistors. Biosens Bioelectron 2024; 266:116695. [PMID: 39241340 DOI: 10.1016/j.bios.2024.116695] [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: 07/19/2024] [Accepted: 08/21/2024] [Indexed: 09/09/2024]
Abstract
Scalable electronic devices that can detect target biomarkers from clinical samples hold great promise for point-of-care nucleic acid testing, but still cannot achieve the detection of target molecules at an attomolar range within a short timeframe (<1 h). To tackle this daunting challenge, we integrate graphene field-effect transistors (GFETs) with exponential target recycling and hybridization chain reaction (TRHCR) to detect oligonucleotides (using miRNA as a model disease biomarker), achieving a detection limit of 100 aM and reducing the sensing time by 30-fold, from 15 h to 30 min. In contrast to traditional linear TRHCR, our exponential TRHCR enables the target miRNA to initiate an autocatalytic system with exponential kinetics, significantly accelerating the reaction speed. The resulting reaction products, long-necked double-stranded polymers with a negative charge, are effectively detected by the GFET through chemical gating, leading to a shift in the Dirac voltage. Therefore, by monitoring the magnitude of this voltage shift, the target miRNA is quantified with high sensitivity. Consequently, our approach successfully detects 22-mer miRNA at concentrations as low as 100 aM in human serum samples, achieving the desired short timeframe of 30 min, which is congruent with point-of-care testing, and demonstrates superior specificity against single-base mismatched interfering oligonucleotides.
Collapse
Affiliation(s)
- Ting Huang
- Department of Biomedical Engineering, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Jingwei Li
- Department of Biomedical Engineering, The Chinese University of Hong Kong, Hong Kong SAR, China; Department of Chemical and Biological Engineering, The Hong Kong University of Science and Technology, Clear Water Bay, Kowloon, Hong Kong SAR, China
| | - Haohan Chen
- Department of Biomedical Engineering, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Honglin Sun
- Department of Biomedical Engineering, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Dong Wook Jang
- Department of Biomedical Engineering, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Md Masruck Alam
- Department of Biomedical Engineering, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Kan Kan Yeung
- Department of Biomedical Engineering, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Qicheng Zhang
- Key Laboratory of 3D Micro/Nano Fabrication and Characterization of Zhejiang Province, School of Engineering, Westlake University, Hangzhou, 310030, China; Research Center for Industries of the Future, Westlake University, Hangzhou, 310030, China
| | - Han Xia
- Department of Clinical Laboratory, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, China
| | - Liting Duan
- Department of Biomedical Engineering, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Chuanbin Mao
- Department of Biomedical Engineering, The Chinese University of Hong Kong, Hong Kong SAR, China.
| | - Zhaoli Gao
- Department of Biomedical Engineering, The Chinese University of Hong Kong, Hong Kong SAR, China; Shun Hing Institute of Advanced Engineering, The Chinese University of Hong Kong, Hong Kong SAR, China.
| |
Collapse
|
33
|
Martínez-Castedo B, Camblor DG, Martín-Arana J, Carbonell-Asins JA, García-Micó B, Gambardella V, Huerta M, Roselló S, Roda D, Gimeno-Valiente F, Cervantes A, Tarazona N. Minimal residual disease in colorectal cancer. Tumor-informed versus tumor-agnostic approaches: unraveling the optimal strategy. Ann Oncol 2024:S0923-7534(24)04981-0. [PMID: 39675560 DOI: 10.1016/j.annonc.2024.12.006] [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: 07/26/2024] [Revised: 11/29/2024] [Accepted: 12/09/2024] [Indexed: 12/17/2024] Open
Abstract
BACKGROUND Circulating tumor DNA (ctDNA) analysis has emerged as a minimally invasive tool for detecting minimal residual disease (MRD) in colorectal cancer (CRC) patients. This enables dynamic risk stratification, earlier recurrence detection and optimized post-surgical treatment. Two primary methodologies have been developed for ctDNA-based MRD detection: tumor-informed strategies, which identify tumor-specific mutations through initial tissue sequencing to guide ctDNA monitoring, and tumor-agnostic approaches, which utilize predefined panels to detect common cancer-associated genomic or epigenomic alterations directly from plasma without prior tissue analysis. The debate over which is superior in terms of sensitivity, specificity, cost-effectiveness and clinical feasibility remains unsolved. DESIGN This review summarizes studies published up to November 2024, exploring the utility and performance of tumor-informed and tumor-agnostic approaches for ctDNA analysis in CRC. We evaluate the strengths and limitations of each methodology, focusing on sensitivity, specificity and clinical outcomes. RESULTS Both strategies demonstrate clinical utility in post-operative risk stratification and guiding adjuvant chemotherapy decisions in CRC patients. Tumor-informed approaches generally exhibit superior sensitivity and specificity for recurrence prediction, attributed to their personalized tumor profile designs. However, these methods are limited by the need for prior tissue sequencing and higher associated costs. In contrast, tumor-agnostic approaches offer broader applicability due to their reliance on plasma-only analysis, although with relatively lower sensitivity. Technological advancements, including fragmentomics and multi-omic integrations, are expanding the capabilities of ctDNA-based MRD detection, enhancing the performance of both approaches. CONCLUSIONS While tumor-informed strategies currently offer higher precision in MRD detection, tumor-agnostic approaches are gaining traction due to their convenience and improving performance metrics. The integration of novel technologies in ongoing clinical trials may redefine the optimal approach for MRD detection in CRC, paving the way for more personalized and adaptive patient management strategies.
Collapse
Affiliation(s)
- B Martínez-Castedo
- Department of Medical Oncology, INCLIVA Biomedical Research Institute, University of Valencia, Valencia, Spain; CIBERONC, Carlos III Health Institute, Madrid, Spain
| | - D G Camblor
- Department of Medical Oncology, INCLIVA Biomedical Research Institute, University of Valencia, Valencia, Spain
| | - J Martín-Arana
- Department of Medical Oncology, INCLIVA Biomedical Research Institute, University of Valencia, Valencia, Spain; CIBERONC, Carlos III Health Institute, Madrid, Spain
| | - J A Carbonell-Asins
- Biostatistics Unit, INCLIVA Biomedical Research Institute, University of Valencia, Valencia, Spain
| | - B García-Micó
- Department of Medical Oncology, INCLIVA Biomedical Research Institute, University of Valencia, Valencia, Spain; CIBERONC, Carlos III Health Institute, Madrid, Spain
| | - V Gambardella
- Department of Medical Oncology, INCLIVA Biomedical Research Institute, University of Valencia, Valencia, Spain
| | - M Huerta
- Department of Medical Oncology, INCLIVA Biomedical Research Institute, University of Valencia, Valencia, Spain; CIBERONC, Carlos III Health Institute, Madrid, Spain
| | - S Roselló
- Department of Medical Oncology, INCLIVA Biomedical Research Institute, University of Valencia, Valencia, Spain; CIBERONC, Carlos III Health Institute, Madrid, Spain
| | - D Roda
- Department of Medical Oncology, INCLIVA Biomedical Research Institute, University of Valencia, Valencia, Spain; CIBERONC, Carlos III Health Institute, Madrid, Spain
| | - F Gimeno-Valiente
- Cancer Evolution and Genome Instability Laboratory, University College London Cancer Institute, London, UK
| | - A Cervantes
- Department of Medical Oncology, INCLIVA Biomedical Research Institute, University of Valencia, Valencia, Spain; CIBERONC, Carlos III Health Institute, Madrid, Spain.
| | - N Tarazona
- Department of Medical Oncology, INCLIVA Biomedical Research Institute, University of Valencia, Valencia, Spain; CIBERONC, Carlos III Health Institute, Madrid, Spain.
| |
Collapse
|
34
|
Guo B, Sun X, Tao S, Tian T, Lei H. Utilizing DNA Logic Device for Precise Detection of Circulating Tumor Cells via High Catalytic Activity Au Nanoparticle Anchoring. Anal Chem 2024; 96:19430-19438. [PMID: 39576332 DOI: 10.1021/acs.analchem.4c03914] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2024]
Abstract
As medical advancements turn most cancers into manageable chronic diseases, new challenges arise in cancer recurrence monitoring. Detecting circulating tumor cells (CTCs) is crucial for monitoring cancer recurrence, but the current methods are cumbersome and costly. This study developed a new CTC detection system combining DNA aptamer recognition, hybridization chain reaction (HCR) technology, and DNA logic devices, enabling the one-step recognition of CTCs by identifying multiple membrane proteins. After catalytically active Au nanoparticles were attached through reduction synthesis in situ onto the DNA hybridization strands of the CTCs surface, a 3,3',5,5'-tetramethylbenzidine (TMB) colorimetric reaction was used to detect CTCs concentration via peroxidase-like catalysis. With this CTCs detection reporting system, we achieved an LOD of 4 cells/mL using an ultraviolet-visible (UV-vis) spectrophotometer. At certain concentrations, CTCs could even be detected visually without the need for an instrument. The development of this CTCs detection reporting system provided a convenient, reliable, and cost-effective detection strategy for widespread CTCs-based cancer recurrence monitoring.
Collapse
Affiliation(s)
- Bin Guo
- Institute of Molecular Medicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China
- College of Chemistry and Materials Science, Shanghai Normal University, Shanghai 200233, China
| | - Xiaofei Sun
- Shanghai Frontiers Science Center of TCM Chemical Biology, Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Shan Tao
- Institute of Molecular Medicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China
- School of Life Sciences, Shanghai University, Shanghai 200444, China
| | - Tian Tian
- Shanghai Frontiers Science Center of TCM Chemical Biology, Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Haozhi Lei
- Institute of Molecular Medicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China
| |
Collapse
|
35
|
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
|
36
|
Rubio-Alarcón C, Stelloo E, Vessies DCL, van 't Erve I, Mekkes NJ, Swennenhuis J, Lakbir S, van Bree EJ, Tijssen M, Delis-van Diemen P, Lanfermeijer M, Linders T, van den Broek D, Punt CJA, Heringa J, Meijer GA, Abeln S, Feitsma H, Fijneman RJA. High Prevalence of Chromosomal Rearrangements and LINE Retrotranspositions Detected in Formalin-Fixed, Paraffin-Embedded Colorectal Cancer Tissue. J Mol Diagn 2024; 26:1065-1080. [PMID: 39332629 DOI: 10.1016/j.jmoldx.2024.08.004] [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: 03/23/2024] [Revised: 07/04/2024] [Accepted: 08/23/2024] [Indexed: 09/29/2024] Open
Abstract
Structural variants (SVs) caused by chromosomal rearrangements in common fragile sites or long interspersed nuclear element (LINE) retrotranspositions are highly prevalent in colorectal cancer. However, methodology for the targeted detection of these SVs is lacking. This article reports the use of formalin-fixed, paraffin-embedded targeted-locus capture (FFPE-TLC) sequencing as a novel technology for the targeted detection of tumor-specific SVs. Analysis of 29 FFPE colorectal tumor samples and 8 matched normal samples revealed tumor-specific SVs in 24 patients (83%), with a median of 2 SVs per patient (range, 1 to 21). A total of 104 SVs were found in the common fragile site-associated genes MACROD2, PRKN, FHIT, and WWOX in 18 patients (62%), and 39 SVs caused by three LINE transposable elements were found in 15 patients (52%). Tumor specificity of SVs was independently verified by droplet digital PCR of tumor tissue DNA, and their applicability as plasma circulating tumor DNA biomarkers was demonstrated. FFPE-TLC sequencing enabled the detection of tumor-specific SVs caused by chromosomal rearrangements and LINE retrotranspositions in FFPE tissue. Therefore, FFPE-TLC sequencing facilitates the investigation of the biological and clinical effects of SVs using FFPE material from (retrospective) cohorts of cancer patients and has potential clinical applicability in the detection of SV biomarkers in the routine molecular diagnostics setting.
Collapse
Affiliation(s)
- Carmen Rubio-Alarcón
- Department of Pathology, The Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Ellen Stelloo
- Cergentis B.V., a Solvias company, Utrecht, the Netherlands
| | - Daan C L Vessies
- Department of Laboratory Medicine, The Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Iris van 't Erve
- Department of Pathology, The Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Nienke J Mekkes
- Bioinformatics Group, Department of Computer Science, Vrije Universiteit Amsterdam, Amsterdam, the Netherlands
| | | | - Soufyan Lakbir
- Department of Pathology, The Netherlands Cancer Institute, Amsterdam, the Netherlands; Bioinformatics Group, Department of Computer Science, Vrije Universiteit Amsterdam, Amsterdam, the Netherlands
| | - Elisabeth J van Bree
- Department of Pathology, The Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Marianne Tijssen
- Department of Pathology, The Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Pien Delis-van Diemen
- Department of Pathology, The Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Mirthe Lanfermeijer
- Department of Laboratory Medicine, The Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Theodora Linders
- Department of Laboratory Medicine, The Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Daan van den Broek
- Department of Laboratory Medicine, The Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Cornelis J A Punt
- Department of Epidemiology, Julius Center, University Medical Center Utrecht, Utrecht University, Utrecht, the Netherlands
| | - Jaap Heringa
- Bioinformatics Group, Department of Computer Science, Vrije Universiteit Amsterdam, Amsterdam, the Netherlands
| | - Gerrit A Meijer
- Department of Pathology, The Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Sanne Abeln
- Bioinformatics Group, Department of Computer Science, Vrije Universiteit Amsterdam, Amsterdam, the Netherlands; AI Technology for Life Group, Department of Information and Computing Sciences, Department of Biology, Utrecht University, Utrecht, the Netherlands
| | - Harma Feitsma
- Cergentis B.V., a Solvias company, Utrecht, the Netherlands
| | - Remond J A Fijneman
- Department of Pathology, The Netherlands Cancer Institute, Amsterdam, the Netherlands.
| |
Collapse
|
37
|
Cartagena J, Deshpande A, Rosenthal A, Tsang M, Hilal T, Rimsza L, Kurzrock R, Munoz J. Measurable Residual Disease in Mantle Cell Lymphoma: The Unbearable Lightness of Being Undetectable. Curr Oncol Rep 2024; 26:1664-1674. [PMID: 39641852 DOI: 10.1007/s11912-024-01620-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/23/2024] [Indexed: 12/07/2024]
Abstract
PURPOSE OF REVIEW This paper evaluates the benefits and limitations of detecting measurable residual disease (MRD) in mantle cell lymphoma (MCL) and assesses its prognostic value. It also aims to highlight the importance of detecting low MRD levels post-treatment and their application in clinical practice. RECENT FINDINGS Recent studies show that MRD levels predict relapse and survival outcomes in hematologic neoplasms, including MCL. RT-qPCR is currently the most used method due to its high reproducibility and sensitivity. Ideal MRD detection should be highly sensitive, cost-effective, and applicable to a wide demographic of patients. This paper concludes that MRD detection has prognostic value in MCL but faces limitations in sensitivity and specificity. Further research is needed to establish the significance of low MRD levels before integrating these methods into clinical practice. Improved MRD detection technologies and understanding their impact on clinical outcomes will guide better patient management in MCL.
Collapse
Affiliation(s)
- Julio Cartagena
- University of Puerto Rico School of Medicine, San Juan, PR, USA
| | | | - Allison Rosenthal
- Department of Hematology and Oncology, Mayo Clinic Arizona, Phoenix, AZ, USA
| | - Mazie Tsang
- Department of Hematology and Oncology, Mayo Clinic Arizona, Phoenix, AZ, USA
| | - Talal Hilal
- Department of Hematology and Oncology, Mayo Clinic Arizona, Phoenix, AZ, USA
| | - Lisa Rimsza
- Department of Laboratory Medicine and Pathology, Mayo Clinic Arizona, Phoenix, AZ, USA
| | - Razelle Kurzrock
- Michels Rare Cancers Research Laboratories, Froedtert and Medical College of Wisconsin, Milwaukee, WI, USA
| | - Javier Munoz
- Department of Hematology and Oncology, Mayo Clinic Arizona, Phoenix, AZ, USA
| |
Collapse
|
38
|
Kurma K, Eslami-S Z, Alix-Panabières C, Cayrefourcq L. Liquid biopsy: paving a new avenue for cancer research. Cell Adh Migr 2024; 18:1-26. [PMID: 39219215 PMCID: PMC11370957 DOI: 10.1080/19336918.2024.2395807] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2024] [Revised: 05/21/2024] [Accepted: 08/14/2024] [Indexed: 09/04/2024] Open
Abstract
The current constraints associated with cancer diagnosis and molecular profiling, which rely on invasive tissue biopsies or clinical imaging, have spurred the emergence of the liquid biopsy field. Liquid biopsy involves the extraction of circulating tumor cells (CTCs), circulating free or circulating tumor DNA (cfDNA or ctDNA), circulating cell-free RNA (cfRNA), extracellular vesicles (EVs), and tumor-educated platelets (TEPs) from bodily fluid samples. Subsequently, these components undergo molecular characterization to identify biomarkers that are critical for early cancer detection, prognosis, therapeutic assessment, and post-treatment monitoring. These innovative biosources exhibit characteristics analogous to those of the primary tumor from which they originate or interact. This review comprehensively explores the diverse technologies and methodologies employed for processing these biosources, along with their principal clinical applications.
Collapse
Affiliation(s)
- Keerthi Kurma
- Laboratory of Rare Human Circulating Cells (LCCRH), University Medical Centre of Montpellier, Montpellier, France
- CREEC/CANECEV, MIVEGEC (CREES),
University of Montpellier, CNRS, IRD, Montpellier, France
- European Liquid Biopsy Society (ELBS), Hamburg, Germany
| | - Zahra Eslami-S
- Laboratory of Rare Human Circulating Cells (LCCRH), University Medical Centre of Montpellier, Montpellier, France
- CREEC/CANECEV, MIVEGEC (CREES),
University of Montpellier, CNRS, IRD, Montpellier, France
- European Liquid Biopsy Society (ELBS), Hamburg, Germany
| | - Catherine Alix-Panabières
- Laboratory of Rare Human Circulating Cells (LCCRH), University Medical Centre of Montpellier, Montpellier, France
- CREEC/CANECEV, MIVEGEC (CREES),
University of Montpellier, CNRS, IRD, Montpellier, France
- European Liquid Biopsy Society (ELBS), Hamburg, Germany
| | - Laure Cayrefourcq
- Laboratory of Rare Human Circulating Cells (LCCRH), University Medical Centre of Montpellier, Montpellier, France
- CREEC/CANECEV, MIVEGEC (CREES),
University of Montpellier, CNRS, IRD, Montpellier, France
- European Liquid Biopsy Society (ELBS), Hamburg, Germany
| |
Collapse
|
39
|
Jin M, Fang J, Peng J, Wang X, Xing P, Jia K, Hu J, Wang D, Ding Y, Wang X, Li W, Chen Z. PD-1/PD-L1 immune checkpoint blockade in breast cancer: research insights and sensitization strategies. Mol Cancer 2024; 23:266. [PMID: 39614285 PMCID: PMC11605969 DOI: 10.1186/s12943-024-02176-8] [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: 11/13/2024] [Indexed: 12/01/2024] Open
Abstract
Immunotherapy targeting programmed cell death-1 (PD-1) and PD-L1 immune checkpoints has reshaped treatment paradigms across several cancers, including breast cancer. Combining PD-1/PD-L1 immune checkpoint blockade (ICB) with chemotherapy has shown promising efficacy in both early and metastatic triple-negative breast cancer, although only a subset of patients experiences durable responses. Identifying responders and optimizing immune drug selection are therefore critical. The effectiveness of PD-1/PD-L1 immunotherapy depends on both tumor-intrinsic factors and the extrinsic cell-cell interactions within the tumor microenvironment (TME). This review systematically summarizes the key findings from clinical trials of ICBs in breast cancer and examines the mechanisms underlying PD-L1 expression regulation. We also highlight recent advances in identifying potential biomarkers for PD-1/PD-L1 therapy and emerging evidence of TME alterations following treatment. Among these, the quantity, immunophenotype, and spatial distribution of tumor-infiltrating lymphocytes stand out as promising biomarkers. Additionally, we explore strategies to enhance the effectiveness of ICBs in breast cancer, aiming to support the development of personalized treatment approaches tailored to the unique characteristics of each patient's tumor.
Collapse
Affiliation(s)
- Menglei Jin
- Department of Breast Surgery (Surgical Oncology), Second Affiliated Hospital, Zhejiang University School of Medicine, 88 Jiefang Road, Hangzhou, Zhejiang, China
- Key Laboratory of Tumor Microenvironment and Immune Therapy of Zhejiang Province, Hangzhou, China
| | - Jun Fang
- Department of Radiation Oncology, Zhejiang Cancer Hospital, Hangzhou, Zhejiang, China
| | - Junwen Peng
- Department of General Surgery, The First People's Hospital of Jiande, Hangzhou, China
| | - Xintian Wang
- Department of General Surgery, The Second People's Hospital of Tongxiang, Jiaxing, Zhejiang, China
| | - Ping Xing
- Department of Breast Surgery (Surgical Oncology), Second Affiliated Hospital, Zhejiang University School of Medicine, 88 Jiefang Road, Hangzhou, Zhejiang, China
- Key Laboratory of Tumor Microenvironment and Immune Therapy of Zhejiang Province, Hangzhou, China
| | - Kunpeng Jia
- Department of Breast Surgery (Surgical Oncology), Second Affiliated Hospital, Zhejiang University School of Medicine, 88 Jiefang Road, Hangzhou, Zhejiang, China
- Key Laboratory of Tumor Microenvironment and Immune Therapy of Zhejiang Province, Hangzhou, China
| | - Jianming Hu
- Department of Breast Surgery (Surgical Oncology), Second Affiliated Hospital, Zhejiang University School of Medicine, 88 Jiefang Road, Hangzhou, Zhejiang, China
- Key Laboratory of Tumor Microenvironment and Immune Therapy of Zhejiang Province, Hangzhou, China
| | - Danting Wang
- Department of Breast Surgery (Surgical Oncology), Second Affiliated Hospital, Zhejiang University School of Medicine, 88 Jiefang Road, Hangzhou, Zhejiang, China
- Key Laboratory of Tumor Microenvironment and Immune Therapy of Zhejiang Province, Hangzhou, China
| | - Yuxin Ding
- Department of Breast Surgery (Surgical Oncology), Second Affiliated Hospital, Zhejiang University School of Medicine, 88 Jiefang Road, Hangzhou, Zhejiang, China
- Key Laboratory of Tumor Microenvironment and Immune Therapy of Zhejiang Province, Hangzhou, China
| | - Xinyu Wang
- Department of Radiation Oncology, Zhejiang Cancer Hospital, Hangzhou, Zhejiang, China
| | - Wenlu Li
- Key Laboratory of Neuropharmacology and Translational Medicine of Zhejiang Province, School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, 310053, China.
| | - Zhigang Chen
- Department of Breast Surgery (Surgical Oncology), Second Affiliated Hospital, Zhejiang University School of Medicine, 88 Jiefang Road, Hangzhou, Zhejiang, China.
- Key Laboratory of Tumor Microenvironment and Immune Therapy of Zhejiang Province, Hangzhou, China.
| |
Collapse
|
40
|
Dey Bhowmik A, Shaw P, Gopinatha Pillai MS, Rao G, Dwivedi SKD. Evolving landscape of detection and targeting miRNA/epigenetics for therapeutic strategies in ovarian cancer. Cancer Lett 2024; 611:217357. [PMID: 39615646 DOI: 10.1016/j.canlet.2024.217357] [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: 09/06/2024] [Revised: 11/22/2024] [Accepted: 11/25/2024] [Indexed: 12/14/2024]
Abstract
Ovarian cancer (OC) accounts for the highest mortality rates among all gynecologic malignancies. The high mortality of OC is often associated with delayed detection, prolonged latency, enhanced metastatic potential, acquired drug resistance, and frequent recurrence. This review comprehensively explores key aspects of OC, including cancer diagnosis, mechanisms of disease resistance, and the pivotal role of epigenetic regulation, particularly by microRNAs (miRs) in cancer progression. We highlight the intricate regulatory mechanisms governing miR expression within the context of OC and the current status of epigenetic advancement in the therapeutic development and clinical trial progression. Through network analysis we elucidate the regulatory interactions between dysregulated miRs in OC and their targets which are involved in different signaling pathways. By exploring these interconnected facets and critical analysis, we endeavor to provide a nuanced understanding of the molecular dynamics underlying OC, its detection and shedding light on potential avenues for miRs and epigenetics-based therapeutic intervention and management strategies.
Collapse
Affiliation(s)
- Arpan Dey Bhowmik
- Peggy and Charles Stephenson Cancer Center, University of Oklahoma Health Sciences Center, Oklahoma City, OK, 73104, USA; Department of Obstetrics and Gynecology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, 73104, USA
| | - Pallab Shaw
- Peggy and Charles Stephenson Cancer Center, University of Oklahoma Health Sciences Center, Oklahoma City, OK, 73104, USA; Department of Pathology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, 73104, USA
| | - Mohan Shankar Gopinatha Pillai
- Peggy and Charles Stephenson Cancer Center, University of Oklahoma Health Sciences Center, Oklahoma City, OK, 73104, USA; Department of Obstetrics and Gynecology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, 73104, USA
| | - Geeta Rao
- Peggy and Charles Stephenson Cancer Center, University of Oklahoma Health Sciences Center, Oklahoma City, OK, 73104, USA; Department of Pathology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, 73104, USA
| | - Shailendra Kumar Dhar Dwivedi
- Peggy and Charles Stephenson Cancer Center, University of Oklahoma Health Sciences Center, Oklahoma City, OK, 73104, USA; Department of Obstetrics and Gynecology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, 73104, USA.
| |
Collapse
|
41
|
Doculara L, Evans K, Gooding JJ, Bayat N, Lock RB. Patient-Specific Circulating Tumor DNA for Monitoring Response to Menin Inhibitor Treatment in Preclinical Models of Infant Leukemia. Cancers (Basel) 2024; 16:3990. [PMID: 39682177 DOI: 10.3390/cancers16233990] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2024] [Revised: 11/19/2024] [Accepted: 11/23/2024] [Indexed: 12/18/2024] Open
Abstract
BACKGROUND In infant KMT2A (MLL1)-rearranged (MLL-r) acute lymphoblastic leukemia (ALL), early relapse and treatment response are currently monitored through invasive repeated bone marrow (BM) biopsies. Circulating tumor DNA (ctDNA) in peripheral blood (PB) provides a minimally invasive alternative, allowing for more frequent disease monitoring. However, a poor understanding of ctDNA dynamics has hampered its clinical translation. We explored the predictive value of ctDNA for detecting minimal/measurable residual disease (MRD) and drug response in a patient-derived xenograft (PDX) model of infant MLL-r ALL. METHODS Immune-deficient mice engrafted with three MLL-r ALL PDXs were monitored for ctDNA levels before and after treatment with the menin inhibitor SNDX-50469. RESULTS The amount of ctDNA detected strongly correlated with leukemia burden during initial engraftment prior to drug treatment. However, following SNDX-50469 treatment, the leukemic burden assessed by either PB leukemia cells through flow cytometry or ctDNA levels through droplet digital polymerase chain reaction (ddPCR) was discrepant. This divergence could be attributed to the persistence of leukemia cells in the spleen and BM, highlighting the ability of ctDNA to reflect disease dynamics in key leukemia infiltration sites. CONCLUSIONS Notably, ctDNA analysis proved to be a superior predictor of MRD compared to PB assessment alone, especially in instances of low disease burden. These findings highlight the potential of ctDNA as a sensitive biomarker for monitoring treatment response and detecting MRD in infant MLL-r ALL.
Collapse
Affiliation(s)
- Louise Doculara
- Children's Cancer Institute, Lowy Cancer Research Centre, School of Clinical Medicine, UNSW Medicine & Health, UNSW Centre for Childhood Cancer Research, UNSW Sydney, Sydney, NSW 2052, Australia
| | - Kathryn Evans
- Children's Cancer Institute, Lowy Cancer Research Centre, School of Clinical Medicine, UNSW Medicine & Health, UNSW Centre for Childhood Cancer Research, UNSW Sydney, Sydney, NSW 2052, Australia
| | - J Justin Gooding
- School of Chemistry, UNSW Sydney, Sydney, NSW 2052, Australia
- Australian Centre for NanoMedicine, UNSW Sydney, Sydney, NSW 2052, Australia
| | - Narges Bayat
- Children's Cancer Institute, Lowy Cancer Research Centre, School of Clinical Medicine, UNSW Medicine & Health, UNSW Centre for Childhood Cancer Research, UNSW Sydney, Sydney, NSW 2052, Australia
| | - Richard B Lock
- Children's Cancer Institute, Lowy Cancer Research Centre, School of Clinical Medicine, UNSW Medicine & Health, UNSW Centre for Childhood Cancer Research, UNSW Sydney, Sydney, NSW 2052, Australia
| |
Collapse
|
42
|
Tiwari R, Dev D, Thalla M, Aher VD, Mundada AB, Mundada PA, Vaghela K. Nano-enabled pharmacogenomics: revolutionizing personalized drug therapy. JOURNAL OF BIOMATERIALS SCIENCE. POLYMER EDITION 2024:1-26. [PMID: 39589779 DOI: 10.1080/09205063.2024.2431426] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/12/2024] [Accepted: 11/07/2024] [Indexed: 11/27/2024]
Abstract
The combination of pharmacogenomics and nanotechnology science of pharmacogenomics into a highly advanced single entity has given birth to personalized medicine known as nano-enabled pharmacogenomics. This review article covers all aspects starting from pharmacogenomics to gene editing tools, how these have evolved or are likely to be evolved for pharmacogenomic application, and how these can be delivered using nanoparticle delivery systems. In this prior work, we explore the evolution of pharmacogenomics over the years, as well as new achievements in the field of genomic sciences, the challenges in drug creation, and application of the strategy of personalized medicine. Particular attention is paid to how nanotechnology helps avoid the problems that accompanied the development of pharmacogenomics earlier, for example, the question of drug resistance and targeted delivery. We also review the latest developments in nano-enabled pharmacogenomics, such as the coupling with other nanobio-technologies, artificial intelligence, and machine learning in pharmacogenomics, and the ethical and regulatory aspects of these developing technologies. The possible uses of nanotechnology in improving the chances of pated and treating drug-resistant cancers are exemplified by case studies together with the current clinical uses of nanotechnology. In the last section, we discuss the future trends and research prospects in this dynamically growing area, stressing the importance of further advancements and collaborations which will advance the nano-enabled pharmacogenomics to their maximum potential.
Collapse
Affiliation(s)
- Ruchi Tiwari
- Psit-Pranveer Singh Institute of Technology (Pharmacy), Kanpur-Agra-Delhi National, Kanpur, India
| | - Dhruv Dev
- Department of Pharmacy, Shivalik College of Pharmacy Nangal, Rupnagar, India
| | - Maharshi Thalla
- Department of Pharmaceutical Sciences, Irma Lerma Rangel School of Pharmacy, Texas A&M University, Kingsville, TX, USA
| | - Vaibhav Dagaji Aher
- Department of Pharmaceutical Medicine, Maharashtra University of Health Sciences, Nashik, India
| | - Anand Badrivishal Mundada
- Department of Pharmacy, R.C. Patel Institute of Pharmaceutical Education and Research, Shirpur, India
| | | | - Krishna Vaghela
- Department of Pharmacy, Saraswati Institute of Pharmaceutical Sciences, National Forensic Sciences University, Gandhinagar, India
| |
Collapse
|
43
|
Machado EDA, Land MGP, Schanaider A. PROGNOSTIC MARKERS FOR THROMBOTIC EVENTS IN PATIENTS WITH GASTRIC OR COLORECTAL ADENOCARCINOMAS. ARQUIVOS BRASILEIROS DE CIRURGIA DIGESTIVA : ABCD = BRAZILIAN ARCHIVES OF DIGESTIVE SURGERY 2024; 37:e1833. [PMID: 39699378 DOI: 10.1590/0102-6720202400039e1833] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/20/2023] [Accepted: 09/17/2024] [Indexed: 12/20/2024]
Abstract
BACKGROUND The relationship between thrombosis and cancer is based on evidence that cancer promotes prothrombotic changes in the host hemostatic system. The activation of blood coagulation is closely linked to tumor growth and dissemination. AIMS To evaluate whether quantifications of plasma circulation tumor deoxyribonucleic acid (DNA) and thrombin-antithrombin complex could act as predictors for thrombotic events and death in patients with gastric or colorectal adenocarcinomas, while also evaluating the Karnofsky Performance Status. METHODS Eighty-two patients were included in the study and divided into three groups: controls (n=20), gastric adenocarcinomas (n=21), and colorectal adenocarcinomas (n=41). In order to calculate the Karnofsky index, information was collected to measure the patient's ability to perform common daily tasks. The following serum measurements were conducted: complete blood count, platelet count, extracellular deoxyribonucleic acid, and thrombin-antithrombin complex. RESULTS Ten patients (16%) experienced thrombosis during treatment. Patients with thrombin-antithrombin complex levels greater than 0.53 had a five-times higher risk of thrombosis. Lower Karnofsky Performance Status was also a risk factor for the event in this population. Neither thrombin-antithrombin complex nor plasma circulation tumor DNA were predictors of death after multivariate adjustment. Thus, Karnofsky index signaled a better overall survival prognosis for colorectal and gastric adenocarcinoma patients. CONCLUSIONS Thrombin-antithrombin complex acts as a marker for thrombosis in patients with colorectal and gastric adenocarcinomas. We recommend prophylactic anticoagulation when the Karnofsky value is low and/or the thrombin-antithrombin complex concentration is greater than 0.53 ng/ml.
Collapse
Affiliation(s)
| | | | - Alberto Schanaider
- Universidade Federal do Rio de Janeiro, Department of Surgery - Rio de Janeiro (RJ), Brazil
| |
Collapse
|
44
|
Kędzierska M, Bańkosz M. Role of Proteins in Oncology: Advances in Cancer Diagnosis, Prognosis, and Targeted Therapy-A Narrative Review. J Clin Med 2024; 13:7131. [PMID: 39685591 DOI: 10.3390/jcm13237131] [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: 10/27/2024] [Revised: 11/19/2024] [Accepted: 11/22/2024] [Indexed: 12/18/2024] Open
Abstract
Modern oncology increasingly relies on the role of proteins as key components in cancer diagnosis, prognosis, and targeted therapy. This review examines advancements in protein biomarkers across several cancer types, including breast cancer, lung cancer, ovarian cancer, and hepatocellular carcinoma. These biomarkers have proven critical for early detection, treatment response monitoring, and tailoring personalized therapeutic strategies. The article highlights the utility of targeted therapies, such as tyrosine kinase inhibitors and monoclonal antibodies, in improving treatment efficacy while minimizing systemic toxicity. Despite these advancements, challenges like tumor resistance, variability in protein expression, and diagnostic heterogeneity persist, complicating universal application. The review underscores future directions, including the integration of artificial intelligence, advanced protein analysis technologies, and the development of combination therapies to overcome these barriers and refine personalized cancer treatment.
Collapse
Affiliation(s)
- Magdalena Kędzierska
- Department of Chemotherapy, Medical University of Lodz, Copernicus Memorial Hospital of Lodz, 90-549 Lodz, Poland
| | - Magdalena Bańkosz
- CUT Doctoral School, Faculty of Materials Engineering and Physics, Department of Material Engineering, Cracow University of Technology, 37 Jana Pawla II Av., 31-864 Krakow, Poland
| |
Collapse
|
45
|
Zhu H, Wang Y, Li L, Wang L, Zhang H, Jin X. Cell-free DNA from clinical testing as a resource of population genetic analysis. Trends Genet 2024:S0168-9525(24)00258-0. [PMID: 39578178 DOI: 10.1016/j.tig.2024.10.007] [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: 07/15/2024] [Revised: 10/23/2024] [Accepted: 10/23/2024] [Indexed: 11/24/2024]
Abstract
As a noninvasive biomarker, cell-free DNA (cfDNA) has achieved remarkable success in clinical applications. Notably, cfDNA is essentially DNA, and conducting whole-genome sequencing (WGS) can yield a wealth of genetic information. These invaluable data should not be confined to one-time use; instead, they should be leveraged for more comprehensive population genetic analysis, including genetic variation spectrum, population structure and genetic selection, and genome-wide association studies (GWASs), among others. Such research findings can, in turn, facilitate clinical practice, enabling more advanced and accurate disease predictions. This review explores the advantages, challenges, and current research areas of cfDNA in population genetics. We hope that this review can serve as a new chapter in the repurposing of cfDNA sequence data generated from clinical testing in population genetics.
Collapse
Affiliation(s)
- Huanhuan Zhu
- BGI Research, Shenzhen 518083, China; Shenzhen Key Laboratory of Transomics Biotechnologies, BGI Research, Shenzhen 518083, China
| | - Yu Wang
- BGI Research, Shenzhen 518083, China; Shenzhen Key Laboratory of Transomics Biotechnologies, BGI Research, Shenzhen 518083, China
| | - Linxuan Li
- BGI Research, Shenzhen 518083, China; Shenzhen Key Laboratory of Transomics Biotechnologies, BGI Research, Shenzhen 518083, China; College of Life Science, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Lin Wang
- BGI Research, Shenzhen 518083, China; Shenzhen Key Laboratory of Transomics Biotechnologies, BGI Research, Shenzhen 518083, China
| | - Haiqiang Zhang
- BGI Research, Shenzhen 518083, China; Shenzhen Key Laboratory of Transomics Biotechnologies, BGI Research, Shenzhen 518083, China
| | - Xin Jin
- BGI Research, Shenzhen 518083, China; Shenzhen Key Laboratory of Transomics Biotechnologies, BGI Research, Shenzhen 518083, China; School of Medicine, South China University of Technology, Guangzhou 510641, China; Shanxi Medical University-BGI Collaborative Center for Future Medicine, Shanxi Medical University, Taiyuan 030001, China.
| |
Collapse
|
46
|
Mannucci A, Goel A. Stool and blood biomarkers for colorectal cancer management: an update on screening and disease monitoring. Mol Cancer 2024; 23:259. [PMID: 39558327 PMCID: PMC11575410 DOI: 10.1186/s12943-024-02174-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2024] [Accepted: 11/07/2024] [Indexed: 11/20/2024] Open
Abstract
BACKGROUND Biomarkers have revolutionized the management of colorectal cancer (CRC), facilitating early detection, prevention, personalized treatment, and minimal residual disease (MRD) monitoring. This review explores current CRC screening strategies and emerging biomarker applications. MAIN BODY We summarize the landscape of non-invasive CRC screening and MRD detection strategies, discuss the limitations of the current approaches, and highlight the promising potential of novel biomarker solutions. The fecal immunochemical test remained the cornerstone of CRC screening, but its sensitivity has been improved by assays that combined its performance with other stool analytes. However, their sensitivity for advanced adenomas and the patient compliance both remain suboptimal. Blood-based tests promise to increase compliance but require further refinement to compete with stool-based biomarker tests. The ideal scenario involves leveraging blood tests to increase screening participation, and simultaneously promote stool- and endoscopy-based screening among those who are compliant. Once solely reliant on upfront surgery followed by stage and pathology-driven adjuvant chemotherapy, the treatment of stage II and III colon cancer has undergone a revolutionary transformation with the advent of MRD testing after surgery. A decade ago, the concept of using a post-surgical test instead of stage and pathology to determine the need for adjuvant chemotherapy was disruptive. Today, a blood test may be more informative of the need for chemotherapy than the stage at diagnosis. CONCLUSION Biomarker research is not just improving, but bringing a transformative change to CRC clinical management. Early detection is not just getting better, but improving thanks to a multi-modality approach, and personalized treatment plans are not just becoming a reality, but a promising future with MRD testing.
Collapse
Affiliation(s)
- Alessandro Mannucci
- Department of Molecular Diagnostics and Experimental Therapeutics, Beckman Research Institute at City of Hope, Monrovia, CA, USA
- Gastroenterology and Gastrointestinal Endoscopy Unit, Vita-Salute San Raffaele University, IRCCS San Raffaele Hospital, Milan, Italy
| | - Ajay Goel
- Department of Molecular Diagnostics and Experimental Therapeutics, Beckman Research Institute at City of Hope, Monrovia, CA, USA.
- City of Hope Comprehensive Cancer Center, Duarte, CA, USA.
| |
Collapse
|
47
|
Colciago RR, De Santis MC, Giandini C, Carnevale MG, Di Cosimo S. Treatment of oligometastatic breast cancer: The role of patient selection. Breast 2024; 79:103839. [PMID: 39561464 PMCID: PMC11754133 DOI: 10.1016/j.breast.2024.103839] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2024] [Revised: 10/16/2024] [Accepted: 11/13/2024] [Indexed: 11/21/2024] Open
Abstract
Up to 90 % of death from solid tumors are caused by metastases. By 2040, breast cancer (BC) is predicted to increase to over 3 million new cases. Additionally, with the personalization and intensification of BC follow-up, many patients will relapse with oligometastatic disease (OMD). Over the past decades, advances in treatment planning, image guidance, target position reproducibility, and online tracking, along with a compelling radiobiological rationale, have led to the implementation of Stereotactic Body Radiation Therapy (SBRT). This has become a valid ablative treatment option for OMD patients. However, there are still concerns about which patients benefit the most from ablative treatment. In this review, we will analyze the literature regarding SBRT for OMD in BC patients. We aim to present the current data on its effectiveness and define the optimal tailored scenarios for SBRT outcomes.
Collapse
Affiliation(s)
| | - Maria Carmen De Santis
- Department of Radiation Oncology, Fondazione IRCCS Istituto Nazionale Tumori, Milan, Italy.
| | - Carlotta Giandini
- Department of Radiation Oncology, Fondazione IRCCS Istituto Nazionale Tumori, Milan, Italy
| | - Maria Grazia Carnevale
- Department of Radiation Oncology, Fondazione IRCCS Istituto Nazionale Tumori, Milan, Italy
| | - Serena Di Cosimo
- Department of Advanced Diagnostics, Fondazione IRCCS Istituto Nazionale Dei Tumori, Milan, Italy
| |
Collapse
|
48
|
Malik MMUD, Alqahtani MM, Hadadi I, Kanbayti I, Alawaji Z, Aloufi BA. Molecular Imaging Biomarkers for Early Cancer Detection: A Systematic Review of Emerging Technologies and Clinical Applications. Diagnostics (Basel) 2024; 14:2459. [PMID: 39518426 PMCID: PMC11545511 DOI: 10.3390/diagnostics14212459] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2024] [Revised: 10/23/2024] [Accepted: 10/29/2024] [Indexed: 11/16/2024] Open
Abstract
BACKGROUND Early cancer detection is crucial for improving patient outcomes. Molecular imaging biomarkers offer the potential for non-invasive, early-stage cancer diagnosis. OBJECTIVES To evaluate the effectiveness and accuracy of molecular imaging biomarkers for early cancer detection across various imaging modalities and cancer types. METHODS A comprehensive search of PubMed/MEDLINE, Embase, Web of Science, Cochrane Library, and Scopus was performed, covering the period from January 2010 to December 2023. Eligibility criteria included original research articles published in English on molecular imaging biomarkers for early cancer detection in humans. The risk of bias for included studies was evaluated using the QUADAS-2 tool. The findings were synthesized through narrative synthesis, with quantitative analysis conducted where applicable. RESULTS In total, 50 studies were included. Positron emission tomography (PET)-based biomarkers showed the highest sensitivity (mean: 89.5%, range: 82-96%) and specificity (mean: 91.2%, range: 85-100%). Novel tracers such as [68Ga]-PSMA for prostate cancer and [18F]-FES for breast cancer demonstrated promising outcomes. Optical imaging techniques showed high specificity in intraoperative settings. CONCLUSIONS Molecular imaging biomarkers show significant potential for improving early cancer detection. Integration into clinical practice could lead to earlier interventions and improved outcomes. Further research is needed to address standardization and cost-effectiveness.
Collapse
Affiliation(s)
- Maajid Mohi Ud Din Malik
- Dr. D.Y. Patil School of Allied Health Sciences, Dr. D.Y. Patil Vidyapeeth, (Deemed to be University) Sant Tukaram Nagar, Pune 411018, MH, India;
| | - Mansour M. Alqahtani
- Department of Radiological Sciences, College of Applied Medical Sciences, Najran University, Najran 61441, Saudi Arabia;
| | - Ibrahim Hadadi
- Department of Radiological Sciences, College of Applied Medical Sciences, King Khalid University, Asir, Abha 62529, Saudi Arabia
| | - Ibrahem Kanbayti
- Radiologic Sciences Department, Faculty of Applied Medical Sciences, King Abdulaziz University, Jeddah 21589, Saudi Arabia;
| | - Zeyad Alawaji
- Department of Radiologic Technology, College of Applied Medical Sciences, Qassim University, Buraydah 51452, Saudi Arabia;
| | - Bader A. Aloufi
- Department of Diagnostic Radiology, College of Applied Medical Sciences, Taibah University, Madinah 42353, Saudi Arabia;
| |
Collapse
|
49
|
Bisig B, Lefort K, Carras S, de Leval L. Clinical use of circulating tumor DNA analysis in patients with lymphoma. Hum Pathol 2024:105679. [PMID: 39491629 DOI: 10.1016/j.humpath.2024.105679] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/25/2024] [Accepted: 10/30/2024] [Indexed: 11/05/2024]
Abstract
The analysis of circulating tumor DNA (ctDNA) in liquid biopsy specimens has an established role for the detection of predictive molecular alterations and acquired resistance mutations in several tumors. The low-invasiveness of this approach allows for repeated sampling and dynamic monitoring of disease evolution. Originating from the entire body tumor bulk, plasma-derived ctDNA reflects intra- and interlesional genetic heterogeneity. In the management of lymphoma patients, ctDNA quantification at various timepoints of the patient's clinical history is emerging as a complementary tool that may improve risk stratification, assessment of treatment response and early relapse detection during follow-up, most prominently in patients with diffuse large B-cell lymphoma or classic Hodgkin lymphoma. While liquid biopsies have not yet entered standard-of-care treatment protocols in these settings, several trials have provided evidence that at least a subset of lymphoma patients may benefit from the introduction of liquid biopsies into daily clinical care. In parallel, continuous technological developments have enabled highly sensitive ctDNA assessment methods, which span from locus-specific techniques identifying single hotspot mutations, to sequencing panels and genome-wide approaches that explore broader genetic and epigenetic alterations. Here, we provide an overview of current methods and ongoing technical developments for ctDNA evaluation. We also summarize the most important data from a selection of clinical studies that have explored the clinical use of ctDNA in several lymphoma entities.
Collapse
Affiliation(s)
- Bettina Bisig
- Institute of Pathology, Department of Laboratory Medicine and Pathology, Lausanne University Hospital and Lausanne University, Lausanne, Switzerland
| | - Karine Lefort
- Institute of Pathology, Department of Laboratory Medicine and Pathology, Lausanne University Hospital and Lausanne University, Lausanne, Switzerland
| | - Sylvain Carras
- Institute for Advanced Biosciences (INSERM U1209, CNRS UMR 5309, UGA), Department of Molecular Biology and Department of Oncohematology, University Hospital Grenoble and University Grenoble Alpes, Grenoble, France
| | - Laurence de Leval
- Institute of Pathology, Department of Laboratory Medicine and Pathology, Lausanne University Hospital and Lausanne University, Lausanne, Switzerland.
| |
Collapse
|
50
|
Luo J, Zhang C, Wu M, Yao X, Duan Y, Li Y. Excitation/emission-enhanced heterostructure photonic crystal array synergizing with "DD-A" FRET entropy-driven circuit for high-resolution and ultrasensitive analysis of ctDNA. Biosens Bioelectron 2024; 263:116615. [PMID: 39106690 DOI: 10.1016/j.bios.2024.116615] [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: 05/25/2024] [Revised: 07/21/2024] [Accepted: 07/29/2024] [Indexed: 08/09/2024]
Abstract
Circulating tumor DNA (ctDNA) is an emerging biomarker of liquid biopsy for cancer. But it remains a challenge to achieve simple, sensitive and specific detection of ctDNA because of low abundance and single-base mutation. In this work, an excitation/emission-enhanced heterostructure photonic crystal (PC) array synergizing with entropy-driven circuit (EDC) was developed for high-resolution and ultrasensitive analysis of ctDNA. The donor donor-acceptor FÖrster resonance energy transfer ("DD-A" FRET) was integrated in EDC based on the introduction of simple auxiliary strand, which exhibited higher sensitivity than that of traditional EDC. The heterostructure PC array was constructed with the bilayer periodic nanostructures of nanospheres. Because the heterostructure PC has the adjustable dual photonic band gaps (PBGs) by changing nanosphere sizes, and the "DD-A" FRET can offer the excitation and emission peak with enough distance, it helps the successful matches between the dual PBGs of heterostructure PC and the excitation/emission peaks of "DD-A" FRET; thus, the fluorescence from EDC can be enhanced effectively from both of excitation and emission processes on heterostructure PC array. Besides, high-resolution of single-base mutation was obtained through the strict recognition of EDC. Benefiting from the specific spectrum-matched and synergetic amplification of heterostructure PC and EDC with "DD-A" FRET, the proposed array obtained ultrasensitive detection of ctDNA with LOD of 12.9 fM, and achieved the analysis of mutation frequency as low as 0.01%. Therefore, the proposed strategy has the advantages of simple operation, mild conditions (enzyme-free and isothermal), high-sensitivity, high-resolution and high-throughput analysis, showing potential in bioassay and clinical application.
Collapse
Affiliation(s)
- Jie Luo
- West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, Sichuan, 610041, PR China
| | - Chuyan Zhang
- Precision Medicine Translational Research Center, Medical Equipment Innovation Research Center, Med-X Center for Manufacturing, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, PR China
| | - Mengfan Wu
- Research Center of Analytical Instrumentation, School of Mechanical Engineering, Sichuan University, Chengdu, Sichuan, 610065, PR China
| | - Xiuyuan Yao
- West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, Sichuan, 610041, PR China
| | - Yixiang Duan
- Research Center of Analytical Instrumentation, School of Mechanical Engineering, Sichuan University, Chengdu, Sichuan, 610065, PR China
| | - Yongxin Li
- West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, Sichuan, 610041, PR China.
| |
Collapse
|