1
|
Xie L, Wang Y, Wan A, Huang L, Wang Q, Tang W, Qi X, Hu X. Research trends of neoadjuvant therapy for breast cancer: A bibliometric analysis. Hum Vaccin Immunother 2025; 21:2460272. [PMID: 39904891 PMCID: PMC11801352 DOI: 10.1080/21645515.2025.2460272] [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: 09/27/2024] [Revised: 01/06/2025] [Accepted: 01/25/2025] [Indexed: 02/06/2025] Open
Abstract
The approach of neoadjuvant therapy for breast cancer, which involves administering systemic treatment prior to primary surgery, has undergone substantial advancements in recent decades. This strategy is intended to reduce tumor size, thereby enabling less invasive surgical procedures and enhancing patient outcomes. This study presents a comprehensive bibliometric analysis of research trends in neoadjuvant therapy for breast cancer from 2009 to 2024. Using data extracted from the Web of Science Core Collection, a total of 3,674 articles were analyzed to map the research landscape in this field. The analysis reveals a steady increase in publication output, peaking in 2022, with the United States and China identified as the leading contributors. Key institutions, such as the University of Texas System and MD Anderson Cancer Center, have been instrumental in advancing the research on neoadjuvant therapy. The study also highlights the contributions of influential authors like Sibylle Loibl and Gunter von Minckwitz, as well as major journals such as the Journal of Clinical Oncology. Emerging research topics, including immunotherapy, liquid biopsy, and artificial intelligence, are gaining prominence and represent potential future directions for clinical applications. This bibliometric analysis provides critical insights into global research trends, key contributors, and future developments in the field of neoadjuvant therapy for breast cancer, offering a foundation for future research and clinical practice advancements.
Collapse
Affiliation(s)
- Laiping Xie
- Department of Nuclear Medicine, Southwest Hospital, Army Medical University, Chongqing, China
| | - Yuhang Wang
- Department of Gastroenterology, Beijing Children’s Hospital, Capital Medical University, Beijing, China
| | - Andi Wan
- Department of Breast and Thyroid Surgery, Southwest Hospital, Army Medical University, Chongqing, China
- Key Laboratory of Chongqing Health Commission for Minimally Invasive and Precise Diagnosis, Chongqing, China
| | - Lin Huang
- Department of Radiology, People’s Hospital of Xingyi, Guizhou, China
| | - Qing Wang
- Institute of Medical Information, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Wanyan Tang
- Department of Oncology, Chongqing Hospital of Traditional Chinese Medicine, Chongqing, China
| | - Xiaowei Qi
- Department of Breast and Thyroid Surgery, Southwest Hospital, Army Medical University, Chongqing, China
- Key Laboratory of Chongqing Health Commission for Minimally Invasive and Precise Diagnosis, Chongqing, China
| | - Xiaofei Hu
- Department of Nuclear Medicine, Southwest Hospital, Army Medical University, Chongqing, China
| |
Collapse
|
2
|
Leiting JL, Alva-Ruiz R, Yonkus JA, Abdelrahman AM, Lynch IT, Carlson DM, Carr RM, Salomao DR, McWilliams RR, Starlinger PP, Thiels CA, Grotz TE, Warner SG, Cleary SP, Kendrick ML, Smoot RL, Kipp BR, Truty MJ. Molecular KRAS ctDNA Predicts Metastases and Survival in Pancreatic Cancer: A Prospective Cohort Study. Ann Surg Oncol 2025; 32:4453-4463. [PMID: 40067610 PMCID: PMC12049301 DOI: 10.1245/s10434-025-17036-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: 05/24/2024] [Accepted: 02/04/2025] [Indexed: 05/03/2025]
Abstract
BACKGROUND Patients with pancreatic ductal adenocarcinoma (PDAC) commonly have occult metastatic dissemination and current standard staging methods have significant limitations in identifying these patients. A clinically available assay allows for the identification of mutant KRAS (mKRAS) circulating tumor DNA (ctDNA) from patient plasma and peritoneal fluid that may identify these patients and impact treatment decision making. We investigated the patterns of diagnostic and prognostic capabilities of mKRAS ctDNA in patients with localized PDAC. METHODS Patients with non-metastatic PDAC were identified and underwent a full staging work-up during their first visit at our institution. Development of metastatic disease and long-term survival outcomes were assessed to compare between the mKRAS testing groups. RESULTS Between 2018 and 2022, 785 patients were evaluated. Among the 785 patients who underwent plasma mKRAS testing, 104 were mKRAS positive. Plasma mKRAS-positive patients were more likely to develop metastatic disease and had worse overall survival. In the 419 patients who underwent peritoneal mKRAS, 123 were mKRAS-positive and were more likely to harbor occult metastases or develop peritoneal rather than hematogenous metastases. For patients who underwent both baseline plasma and peritoneal mKRAS testing, any positive mKRAS test regardless of compartment was associated with worse outcomes. CONCLUSIONS Detection of mKRAS ctDNA in plasma and peritoneal fluid of patients with localized PDAC is not only feasible but also identifies those at high risk of metastatic progression and worse survival outcomes. It allows for better prognostication and can significantly impact subsequent treatment decisions, particularly in patients where an aggressive surgical approach is being considered.
Collapse
Affiliation(s)
- Jennifer L Leiting
- Division of Hepatobiliary and Pancreas Surgery, Department of Surgery, Mayo Clinic, Rochester, MN, USA
| | - Roberto Alva-Ruiz
- Division of Hepatobiliary and Pancreas Surgery, Department of Surgery, Mayo Clinic, Rochester, MN, USA
| | - Jennifer A Yonkus
- Division of Hepatobiliary and Pancreas Surgery, Department of Surgery, Mayo Clinic, Rochester, MN, USA
| | - Amro M Abdelrahman
- Division of Hepatobiliary and Pancreas Surgery, Department of Surgery, Mayo Clinic, Rochester, MN, USA
| | - Isaac T Lynch
- Division of Hepatobiliary and Pancreas Surgery, Department of Surgery, Mayo Clinic, Rochester, MN, USA
| | - Danielle M Carlson
- Division of Hepatobiliary and Pancreas Surgery, Department of Surgery, Mayo Clinic, Rochester, MN, USA
| | - Ryan M Carr
- Department of Medical Oncology, Mayo Clinic, Rochester, MN, USA
| | - Diva R Salomao
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN, USA
| | | | - Patrick P Starlinger
- Division of Hepatobiliary and Pancreas Surgery, Department of Surgery, Mayo Clinic, Rochester, MN, USA
| | - Cornelius A Thiels
- Division of Hepatobiliary and Pancreas Surgery, Department of Surgery, Mayo Clinic, Rochester, MN, USA
| | - Travis E Grotz
- Division of Hepatobiliary and Pancreas Surgery, Department of Surgery, Mayo Clinic, Rochester, MN, USA
| | - Susanne G Warner
- Division of Hepatobiliary and Pancreas Surgery, Department of Surgery, Mayo Clinic, Rochester, MN, USA
| | - Sean P Cleary
- Division of General Surgery, University of Toronto, Toronto, ON, Canada
| | - Michael L Kendrick
- Division of Hepatobiliary and Pancreas Surgery, Department of Surgery, Mayo Clinic, Rochester, MN, USA
| | - Rory L Smoot
- Division of Hepatobiliary and Pancreas Surgery, Department of Surgery, Mayo Clinic, Rochester, MN, USA
| | - Benjamin R Kipp
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN, USA
| | - Mark J Truty
- Division of Hepatobiliary and Pancreas Surgery, Department of Surgery, Mayo Clinic, Rochester, MN, USA.
| |
Collapse
|
3
|
Addamo-De Nard B, Geissmann M, Akhoundova D, Pistoni C, Brezina T, Zoche M, Weber A, Hussung S, Fritsch R. A novel KRAS exon 2 drop-off digital PCR assay for mutation detection in cell-free DNA of cancer patients. Diagn Pathol 2025; 20:62. [PMID: 40413426 PMCID: PMC12103757 DOI: 10.1186/s13000-025-01637-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2025] [Accepted: 03/27/2025] [Indexed: 05/27/2025] Open
Abstract
BACKGROUND KRAS exon 2 mutations are highly prevalent in human malignancies, making them attractive targets for detection and monitoring in cell-free DNA (cfDNA) of cancer patients. Drop-off assays designed for digital polymerase chain reaction (ddPCR drop-off) span entire mutational hotspots and detect any mutated allele within the covered region, overcoming a major limitation of mutation-specific ddPCR assays. We therefore set out to develop a novel KRAS codon 12/13 ddPCR drop-off assay for the robust, highly sensitive and specific detection of KRAS exon 2 hotspot mutations in cfDNA. METHODS We designed, optimized and extensively validated a KRAS codon 12/13 ddPCR drop-off assay. We compared assay performance to a commercially available KRAS multiplex assay. For clinical validation, we analyzed plasma samples collected from patients with KRAS-mutated gastrointestinal malignancies. RESULTS Limit of detection of the newly established ddPCR drop-off assay was 0.57 copies/µL, limit of blank was 0.13 copies/µ. The inter-assay precision (r2) was 0.9096. Our newly developed KRAS ddPCR drop-off assay accurately identified single nucleotide variants in 35/36 (97.2%) of circulating tumor DNA-positive samples from the patient validation cohort. Assay cross-validation showed that the newly established KRAS codon 12/13 ddPCR drop-off assay outperformed a commercially available KRAS multiplex ddPCR assay in terms of specificity. Moreover, the newly developed assay proved to be suitable for multiplexing with mutation-specific probes. CONCLUSION We developed and clinically validated a highly accurate ddPCR drop-off assay for KRAS exon 2 hot-spot detection in cfDNA with broad applicability for clinic and research.
Collapse
Affiliation(s)
- Bianca Addamo-De Nard
- Department of Medical Oncology and Hematology, University Hospital Zurich, Zurich, Switzerland
| | - Meret Geissmann
- Department of Medical Oncology and Hematology, University Hospital Zurich, Zurich, Switzerland
| | - Dilara Akhoundova
- Department of Medical Oncology, Bern University Hospital, University of Bern, Bern, Switzerland
| | - Clelia Pistoni
- Department of Medical Oncology and Hematology, University Hospital Zurich, Zurich, Switzerland
- University of Zurich, Zurich, Switzerland
| | - Tomas Brezina
- Department of Medical Oncology and Hematology, University Hospital Zurich, Zurich, Switzerland
| | - Martin Zoche
- Institute of Pathology and Molecular Pathology, University Hospital Zurich, Zurich, Switzerland
| | - Achim Weber
- Institute of Pathology and Molecular Pathology, University Hospital Zurich, Zurich, Switzerland
- Institute of Molecular Cancer Research (IMCR), University of Zurich, Zurich, Switzerland
| | - Saskia Hussung
- Department of Medical Oncology and Hematology, University Hospital Zurich, Zurich, Switzerland
- University of Zurich, Zurich, Switzerland
| | - Ralph Fritsch
- Department of Medical Oncology and Hematology, University Hospital Zurich, Zurich, Switzerland.
- University of Zurich, Zurich, Switzerland.
- Comprehensive Cancer Center Zurich, Raemistrasse 100, Zurich, 8091, Switzerland.
| |
Collapse
|
4
|
Nadeau A, Tsering T, Abdouh M, Kienzle L, Cleyle J, Taylor L, Douanne N, Dickinson K, Siegel PM, Burnier JV. Characterization of extracellular vesicle-associated DNA and proteins derived from organotropic metastatic breast cancer cells. J Exp Clin Cancer Res 2025; 44:157. [PMID: 40410902 PMCID: PMC12100931 DOI: 10.1186/s13046-025-03418-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2025] [Accepted: 05/12/2025] [Indexed: 05/25/2025] Open
Abstract
BACKGROUND While primary breast cancer (BC) is often effectively managed, metastasis remains the primary cause of BC-related fatalities. Gaps remain in our understanding of the mechanisms regulating cancer cell organotropism with predilection to specific organs. Unraveling mediators of site-specific metastasis could enhance early detection and enable more tailored interventions. Liquid biopsy represents an innovative approach in cancer involving the analysis of biological materials such as circulating tumor DNA and tumor-derived extracellular vesicles (EV) found in body fluids like blood or urine. This offers valuable insights for characterizing and monitoring tumor genomes to advance personalized medicine in metastatic cancers. METHODS We performed in-depth analyses of EV cargo associated with BC metastasis using eight murine cell line models with distinct metastatic potentials and organotropism to the lung, the bone, the liver, and the brain. We characterized the secretome of these cells to identify unique biomarkers specific to metastatic sites. RESULTS Small EVs isolated from all cell lines were quantified and validated for established EV markers. Tracking analysis and electron microscopy revealed EV secretion patterns that differed according to cell line. Cell-free (cf)DNA and EV-associated DNA (EV-DNA) were detected from all cell lines with varying concentrations. We detected a TP53 mutation in both EV-DNA and cfDNA. Mass spectrometry-based proteomics analyses identified 698 EV-associated proteins, which clustered according to metastatic site. This analysis highlighted both common EV signatures and proteins involved in cancer progression and organotropism unique to metastatic cell lines. Among these, 327 significantly differentially enriched proteins were quantified with high confidence levels across BC and metastatic BC cells. We found enrichment of specific integrin receptors in metastatic cancer EVs compared to EVs secreted from non-transformed epithelial cells and matched tumorigenic non-metastatic cells. Pathway analyses revealed that EVs derived from parental cancer cells display a cell adhesion signature and are enriched with proteins involved in cancer signaling pathways. CONCLUSION Taken together, the characterization of EV cargo in a unique model of BC organotropism demonstrated that EV-DNA and EV proteomes were informative of normal and cancer states. This work could help to identify BC biomarkers associated with site-specific metastasis and new therapeutic targets.
Collapse
Affiliation(s)
- Amélie Nadeau
- Cancer Research Program, Research Institute of the McGill University Health Centre, Montreal, QC, Canada
- Department of Pathology, McGill University, Montreal, QC, Canada
| | - Thupten Tsering
- Cancer Research Program, Research Institute of the McGill University Health Centre, Montreal, QC, Canada
- Department of Pathology, McGill University, Montreal, QC, Canada
| | - Mohamed Abdouh
- Cancer Research Program, Research Institute of the McGill University Health Centre, Montreal, QC, Canada
| | - Laura Kienzle
- Cancer Research Program, Research Institute of the McGill University Health Centre, Montreal, QC, Canada
| | - Jenna Cleyle
- Centre for Translational Biology, Research Institute of the McGill University Health Centre, Montreal, QC, Canada
| | - Lorne Taylor
- Centre for Translational Biology, Research Institute of the McGill University Health Centre, Montreal, QC, Canada
| | - Noélie Douanne
- Cancer Research Program, Research Institute of the McGill University Health Centre, Montreal, QC, Canada
- Department of Pathology, McGill University, Montreal, QC, Canada
| | - Kyle Dickinson
- Cancer Research Program, Research Institute of the McGill University Health Centre, Montreal, QC, Canada
| | - Peter M Siegel
- Rosalind & Morris Goodman Cancer Institute, McGill University, Montreal, QC, Canada
- Department of Medicine, McGill University, Montreal, QC, Canada
| | - Julia V Burnier
- Cancer Research Program, Research Institute of the McGill University Health Centre, Montreal, QC, Canada.
- Department of Pathology, McGill University, Montreal, QC, Canada.
- Rosalind & Morris Goodman Cancer Institute, McGill University, Montreal, QC, Canada.
- Gerald Bronfman Department of Oncology, McGill University, Montreal, QC, Canada.
| |
Collapse
|
5
|
Xu Y, Yang JCH, Zhao Y, Doucet L, Zhou J, Wang Y, Planchard D, Fan Y, Jin B, Han Z, Greillier L, Mazieres J, Sun M, Hu Y, Song X, Ding C, Wu L, Tang K, Liang L, Yao Y, Cheng Y, He Y, Ferreira BP, Ghiringhelli F, Felip E, Bosch-Barrera J, Liu A, Yu Y, Dong X, Gao J, Camidge DR, Nian W, Zhou C, Yang R, John T, Gao B, Bazhenova L, Nagasaka M, Wang J, Ren X, Xu F, Li W, Zhao D, Wang H, Sun S, Huang J, Zhu X, Zheng L, Jänne PA, Wang M. Genetic biomarker study of sunvozertinib for clinical prognosis and prediction in NSCLC with EGFR exon 20 insertion mutation. Cell Rep Med 2025; 6:102121. [PMID: 40334661 DOI: 10.1016/j.xcrm.2025.102121] [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: 10/09/2024] [Revised: 02/01/2025] [Accepted: 04/10/2025] [Indexed: 05/09/2025]
Abstract
This is a report of biomarker analysis for sunvozertinib, a leading epidermal growth factor receptor (EGFR) tyrosine kinase inhibitor (TKI) targeting EGFR exon 20 insertion mutation (exon20ins) non-small cell lung cancer (NSCLC). There is a positive correlation between positive EGFR exon20ins in plasma circulating tumor DNA (ctDNA) and advanced disease. Shorter progression-free survival and lower objective response rate (45.8% vs. 68.0%) were observed in patients with positive EGFR exon20ins compared to those with negative status. Droplet digital PCR analysis showed that the EGFR exon20ins allele in ctDNA decreased over time in 85.7% of patients, with the earliest clearance occurred after 1 week of sunvozertinib treatment. Acquired EGFR C797S is identified as a potential on-target resistance mutation to sunvozertinib. Finally, efforts are undertaken to investigate therapeutic approaches that aim to overcome the putative acquired resistance to sunvozertinib.
Collapse
Affiliation(s)
- Yan Xu
- Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - James Chih-Hsin Yang
- National Taiwan University Hospital and National Taiwan University Cancer Center, Taipei, Taiwan
| | - Yanqiu Zhao
- Affiliated Cancer Hospital of Zhengzhou University, Zhengzhou, China
| | - Ludovic Doucet
- Institut de Cancérologie de l'Ouest (ICO) - René Gauducheau, Saint-Herblain, France
| | - Jianying Zhou
- The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | | | - David Planchard
- Gustave Roussy, Department of Medical Oncology, Thoracic Group, Villejuif, France; Faculty of Medicine, Paris-Saclay University, Paris, France
| | - Yun Fan
- Zhejiang Cancer Hospital, Hangzhou, China
| | - Bo Jin
- The First Hospital of China Medical University, Shenyang, China
| | - Zhigang Han
- The Affiliated Cancer Hospital of Xinjiang Medical University, Urumqi, China
| | | | | | - Meili Sun
- Central Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Ying Hu
- Beijing Chest Hospital, Capital Medical University, Beijing, China
| | - Xia Song
- Shanxi Provincial Cancer Hospital, Taiyuan, China
| | - Cuimin Ding
- The Fourth Hospital of Hebei Medical University, Shijiazhuang, China
| | - Lin Wu
- Hunan Cancer Hospital, Changsha, China
| | - Kejing Tang
- The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Li Liang
- Peking University Third Hospital, Beijing, China
| | - Yu Yao
- The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | | | - Yong He
- Army Medical Center of PLA, Chongqing, China
| | | | | | | | | | - Anwen Liu
- The Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Yan Yu
- Harbin Medical University Cancer Hospital, Harbin, China
| | - Xiaorong Dong
- Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Junzhen Gao
- The Affiliated Hospital of Inner Mongolia Medical University, Hohhot, China
| | - D Ross Camidge
- University of Colorado Hospital, Anschutz Cancer Pavilion, Aurora, CO, USA
| | - Weiqi Nian
- Chongqing Cancer Hospital, Chongqing, China
| | - Chengzhi Zhou
- The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | | | - Thomas John
- Peter MacCallum Cancer Centre, Melbourne, VIC, Australia
| | - Bo Gao
- Blacktown Hospital, Sydney, NSW, Australia
| | - Lyudmila Bazhenova
- University of California, San Diego (UCSD), Moores Cancer Center, La Jolla, CA, USA
| | - Misako Nagasaka
- University of California Irvine Medical Center (UCIMC) - Chao Family Comprehensive Cancer Center, Orange, CA, USA
| | | | - Xiubao Ren
- Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
| | - Fei Xu
- The First Affiliated Hospital of Nanchang University, Nanchang, China
| | - Wen Li
- The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Dahai Zhao
- The Second Hospital of Anhui Medical University, Hefei, China
| | - Huijie Wang
- Fudan University Shanghai Cancer Center, Shanghai, China
| | - Si Sun
- Fudan University Shanghai Cancer Center, Shanghai, China
| | - Jian'an Huang
- The First Affiliated Hospital of Soochow University, Suzhou, China
| | | | - Li Zheng
- Dizal Pharmaceutical, Shanghai, China
| | - Pasi A Jänne
- Lowe Center for Thoracic Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Mengzhao Wang
- Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China.
| |
Collapse
|
6
|
Arango-Argoty G, Haghighi M, Sun GJ, Choe EY, Markovets A, Barrett JC, Lai Z, Jacob E. An artificial intelligence-based model for prediction of clonal hematopoiesis variants in cell-free DNA samples. NPJ Precis Oncol 2025; 9:147. [PMID: 40394286 DOI: 10.1038/s41698-025-00921-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2024] [Accepted: 04/23/2025] [Indexed: 05/22/2025] Open
Abstract
Circulating tumor DNA is a critical biomarker in cancer diagnostics, but its accurate interpretation requires careful consideration of clonal hematopoiesis (CH), which can contribute to variants in cell-free DNA and potentially obscure true tumor-derived signals. Accurate detection of somatic variants of CH origin in plasma samples remains challenging in the absence of matched white blood cells sequencing. Here we present an open-source machine learning framework (MetaCH) which classifies variants in cfDNA from plasma-only samples as CH or tumor origin, surpassing state-of-the-art classification rates.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Etai Jacob
- Oncology R&D, AstraZeneca, Waltham, MA, USA.
| |
Collapse
|
7
|
Zhang L, Zhang Y, Chen L, Wang X, Liu Y, Huang Y, Song Y, Zhang Y, Tai J. Research trends and hotspots of circulating tumor DNA in colorectal cancer: a bibliometric study. Front Oncol 2025; 15:1492880. [PMID: 40438683 PMCID: PMC12116327 DOI: 10.3389/fonc.2025.1492880] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2024] [Accepted: 04/23/2025] [Indexed: 06/01/2025] Open
Abstract
Introduction Colorectal cancer (CRC) is the third most common cancer worldwide and the second leading cause of cancer-related deaths. The current standard of care for patients with early-stage CRC includes surgical resection and, in selected patients, adjuvant chemotherapy. Circulating tumor DNA (ctDNA) testing is an important component of liquid biopsy, and with the development of testing technology, its value for clinical application has attracted widespread attention. The aim of this study was to help researchers review what has been achieved and better understand the direction of future research through bibliometric analysis. Methods We used the Web of Science Core Collection database to search for ctDNA in CRC-related articles published between 2014 - 2023. Bibliometric analyses of major keywords, authors, countries, institutions, literature and journals in the field were performed using CiteSpace and VOSviewer. Results The number of publications in the field has continued to increase over the last decade. The United States has the highest number of publications, and Italian research scholars have made outstanding contributions. Cancers is the journal with the highest number of publications. Conclusion This study systematically summarizes the research findings in the field of ctDNA in CRC from 2014 to 2023 and describes the research hotspots and trends worldwide that can guide future research.
Collapse
Affiliation(s)
- Lele Zhang
- Department of Colorectal & Anal Surgery, General Surgery Center, The First Hospital of Jilin University, Changchun, China
| | - Yuzhe Zhang
- The First Laboratory of Cancer Institute, The First Hospital of China Medical University, Shenyang, China
| | - Lei Chen
- Department of Colorectal & Anal Surgery, General Surgery Center, The First Hospital of Jilin University, Changchun, China
| | - Xu Wang
- Department of Colorectal & Anal Surgery, General Surgery Center, The First Hospital of Jilin University, Changchun, China
| | - Yulian Liu
- Department of Traditional Chinese Medicine, Chongqing Hospital of Jiangsu Province Hospital, Chongqing, China
| | - Yishan Huang
- Department of Colorectal & Anal Surgery, General Surgery Center, The First Hospital of Jilin University, Changchun, China
| | - Yu Song
- Department of Colorectal & Anal Surgery, General Surgery Center, The First Hospital of Jilin University, Changchun, China
| | - Ye Zhang
- The First Laboratory of Cancer Institute, The First Hospital of China Medical University, Shenyang, China
| | - Jiandong Tai
- Department of Colorectal & Anal Surgery, General Surgery Center, The First Hospital of Jilin University, Changchun, China
| |
Collapse
|
8
|
Galant N, Grenda A, Krawczyk P, Pięt M, Milanowski J. Liquid biopsy in diagnosis and monitoring of treatment efficacy in patients with small cell lung cancer. Mol Biol Rep 2025; 52:455. [PMID: 40358752 PMCID: PMC12075280 DOI: 10.1007/s11033-025-10569-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2025] [Accepted: 05/02/2025] [Indexed: 05/15/2025]
Abstract
Small-cell lung cancer (SCLC) remains one of the deadliest cancers worldwide. Patients' survival remains poor due to its rapid growth, high metastatic rate and limited possibilities of treatment. For many years, SCLC management has been based mostly on chemo and radiotherapy. However, new therapeutic approaches have been proposed in the past few years, including immunotherapy, which is currently implemented in clinical practice. Unfortunately, in many cases, response to therapy, especially chemotherapy, remains poor, or the patient becomes resistant to initially effective treatment. One of the crucial problems during SCLC patient care is a lack of appropriate predictive biomarkers for various therapeutic approaches. Another critical issue is the scarcity of collected tissue during biopsy, which may be insufficient or of too poor quality for analysis. A liquid biopsy might be the key to solving both of those problems as it is collected in a non-invasive way and enables the measurement of various biomarkers, including circulating tumor DNA (ctDNA) and circulating tumor cells (CTCs). In this review, we discuss various approaches to potentially incorporating liquid biopsy into clinical application - as a companion to imaging during SCLC diagnostics, a new approach to molecular subtyping, and a material enabling predictive or prognostic biomarkers assessment. We also summarize ongoing clinical trials encompassing SCLC patients in which liquid biopsy is collected and examined.
Collapse
Affiliation(s)
- Natalia Galant
- Department of Pneumonology, Oncology and Allergology, Medical University of Lublin, Lublin, Poland.
| | - Anna Grenda
- Department of Pneumonology, Oncology and Allergology, Medical University of Lublin, Lublin, Poland
| | - Paweł Krawczyk
- Department of Pneumonology, Oncology and Allergology, Medical University of Lublin, Lublin, Poland
| | - Mateusz Pięt
- Department of Virology and Immunology, Maria Curie-Skłodowska University, Lublin, Poland
| | - Janusz Milanowski
- Department of Pneumonology, Oncology and Allergology, Medical University of Lublin, Lublin, Poland
| |
Collapse
|
9
|
Martviset P, Chantree P, Tongsiri N, Plengsuriyakarn T, Na-Bangchang K. Targeting tumor-associated genes, immune response, and circulating tumor cells in intrahepatic cholangiocarcinoma: Therapeutic potential of Atractylodes lancea (Thunb.) DC. PLoS One 2025; 20:e0323732. [PMID: 40359186 PMCID: PMC12074528 DOI: 10.1371/journal.pone.0323732] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2024] [Accepted: 04/14/2025] [Indexed: 05/15/2025] Open
Abstract
Cholangiocarcinoma (CCA) is one of the most aggressive cancers with a poor prognosis. Current treatment strategies involve hepatobiliary surgery, chemotherapy, radiotherapy, and supportive care; however, the success of these treatments remains limited. Therefore, this study investigated the potential of Atractylodes lancea (Thunb) D.C. (AL) in limiting the progress of CCA by targeting the expression of cancer-related genes involved in immune responses and circulating tumor cells. The study was part of Phase 2A clinical trial in advanced-stage intrahepatic iCCA (iCCA) patients: Group 1 (n = 16) received low-dose AL (capsule formulation of the standardized extract of AL: CMC-AL) with standard supportive care, Group 2 (n = 16) received high-dose AL with standard supportive care, and Group 3 (n = 16) received standard supportive care alone. Venous whole blood samples (EDTA, 5 ml) were collected from each patient on Day 1 and Day 90 and the non-CCA subjects (n = 16) on Day 1. Fifty-nine samples (48 and 11 samples for Day 1 and Day 90, respectively) were processed for total RNA isolation. Gene expression was evaluated using reverse transcription followed by a PCR array. Regardless of dosage, gene expression patterns in the AL-treated groups closely resembled those of the healthy subjects. Specifically, cancer-associated genes, including VEGF-A, NR4A3, Ki-67, and EpCAM, were significantly down-regulated. Additionally, the expression levels of immune-related genes were modulated in AL-treated patients. The treatment groups exhibited lower levels of the pro-inflammatory cytokine IL-6, increased expression of the anti-inflammatory cytokine IL-10, and cell-mediated immune-related molecules such as CTLA4 and PFR1. These findings suggest the potential of AL for iCCA treatment. However, additional studies are required to confirm the correlation between gene and protein expression profiles, as well as CTCs profile.
Collapse
Affiliation(s)
- Pongsakorn Martviset
- Division of Parasitology, Department of Preclinical Science, Faculty of Medicine, Thammasat University, Pathum Thani, Thailand
- Graduate Program in Applied Biosciences, Faculty of Medicine, Thammasat University, Pathum Thani, Thailand
| | - Pathanin Chantree
- Graduate Program in Applied Biosciences, Faculty of Medicine, Thammasat University, Pathum Thani, Thailand
- Division of Anatomy, Department of Preclinical Science, Faculty of Medicine, Thammasat University, Pathum Thani, Thailand
| | - Nisit Tongsiri
- Department of Surgery, Sakon Nakhon Hospital, Sakon Nakhon, Thailand
| | - Tullayakorn Plengsuriyakarn
- Graduate Program in Bioclinical Sciences, Chulabhorn International College of Medicine, Thammasat University, Pathum Thani, Thailand
- Center of Excellence in Malaria and Cholangiocarcinoma, Thammasat University, Pathum Thani, Thailand
| | - Kesara Na-Bangchang
- Graduate Program in Bioclinical Sciences, Chulabhorn International College of Medicine, Thammasat University, Pathum Thani, Thailand
- Center of Excellence in Malaria and Cholangiocarcinoma, Thammasat University, Pathum Thani, Thailand
| |
Collapse
|
10
|
Yang W, Nguyen R, Safri F, Shiddiky MJA, Warkiani ME, George J, Qiao L. Liquid Biopsy in Hepatocellular Carcinoma: ctDNA as a Potential Biomarker for Diagnosis and Prognosis. Curr Oncol Rep 2025:10.1007/s11912-025-01681-3. [PMID: 40343687 DOI: 10.1007/s11912-025-01681-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/21/2025] [Indexed: 05/11/2025]
Abstract
PURPOSE OF REVIEW Hepatocellular carcinoma (HCC) is a leading cause of cancer-related mortality worldwide, with rising incidence and mortality. Early-stage HCC is often asymptomatic, and the lack of reliable early diagnostic markers leads to late-stage diagnosis with limited treatment options. Current treatment relies on tumour staging and patient status, but accurate staging requires invasive procedures that fail to capture tumour heterogeneity and progression. There is an urgent need for less invasive diagnostic strategies, such as liquid biopsy technologies, which allow for repeated sampling and real-time analysis of tumour dynamics. Liquid biopsies, including circulating tumour cells (CTCs) and circulating tumour DNA (ctDNA), offer the potential to monitor recurrence, metastasis, and treatment responses, potentially transforming HCC clinical management by enabling earlier intervention and personalised treatment strategies. RECENT FINDINGS Recent studies emphasise the potential of ctDNA as a non-invasive biomarker by targeting DNA methylation for early HCC detection, enabling timely intervention and personalised treatment to improve patient outcomes. Comparative analyses have shown that ctDNA mutation testing outperforms alpha-fetoprotein (AFP), with a sensitivity of 85% and a specificity of 92%, compared to 60% sensitivity and 80% specificity for AFP. Additionally, profiling the ctDNA mutation landscape of 100 HCC patients has identified recurrent mutations in genes such as TP53, CTNNB1, and AXIN1. ctDNA appears to be a promising non-invasive biomarker in the clinical management of HCC patients, with the sensitivity and specificity improving by 41.67% and 15% respectively. The ctDNA mutations, particularly those targeting DNA methylation, highlight great potential for precision medicine, critical for early diagnosis and prognosis of HCC.
Collapse
Affiliation(s)
- William Yang
- Storr Liver Centre, The Westmead Institute for Medical Research, The University of Sydney and Westmead Hospital, Westmead, NSW, 2145, Australia
| | - Romario Nguyen
- Storr Liver Centre, The Westmead Institute for Medical Research, The University of Sydney and Westmead Hospital, Westmead, NSW, 2145, Australia
| | - Fatema Safri
- Storr Liver Centre, The Westmead Institute for Medical Research, The University of Sydney and Westmead Hospital, Westmead, NSW, 2145, Australia
| | - Muhammad J A Shiddiky
- Rural Health Research Institute (RHRI), Charles Sturt University, Orange, NSW, 2800, Australia
| | - Majid E Warkiani
- School of Biomedical Engineering, The University of Technology Sydney, Ultimo, NSW, 2007, Australia
| | - Jacob George
- Storr Liver Centre, The Westmead Institute for Medical Research, The University of Sydney and Westmead Hospital, Westmead, NSW, 2145, Australia.
- Storr Liver Centre, Westmead Institute for Medical Research (WIMR), the University of Sydney, Westmead, NSW, 2145, Australia.
| | - Liang Qiao
- Storr Liver Centre, The Westmead Institute for Medical Research, The University of Sydney and Westmead Hospital, Westmead, NSW, 2145, Australia.
- Storr Liver Centre, Westmead Institute for Medical Research (WIMR), the University of Sydney, Westmead, NSW, 2145, Australia.
| |
Collapse
|
11
|
Poggiana C, Piazza AF, Catoni C, Gallingani I, Piccin L, Pellegrini S, Aneloni V, Salizzato V, Pigozzo J, Fabozzi A, Facchinetti A, Menin C, Del Fiore P, Mocellin S, Chiarion-Sileni V, Rosato A, Scaini MC. A model workflow for microfluidic enrichment and genetic analysis of circulating melanoma cells. Sci Rep 2025; 15:15329. [PMID: 40316673 PMCID: PMC12048555 DOI: 10.1038/s41598-025-99153-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2024] [Accepted: 04/17/2025] [Indexed: 05/04/2025] Open
Abstract
Circulating melanoma cells (CMCs) are responsible for the hematogenous spread of melanoma and, ultimately, metastasis. However, their study has been limited by the low abundance in patient blood and the heterogeneous expression of surface markers. The FDA-approved CellSearch platform enriches CD146-positive CMCs, whose number correlates with progression-free survival and overall survival. However, a single marker may not be sufficient to identify them all. The Parsortix system allows enrichment of CMCs based on their size and deformability, keeping them viable and suitable for downstream molecular analyses. In this study, we tested the strengths, weaknesses and potential convergences of both platforms to integrate the counting of CMCs with a protocol for their genetic analysis. Samples run on Parsortix were labeled with a customized melanoma antibody cocktail, which efficiently labeled and distinguished CMCs from endothelial cells/leukocytes. The capture rate of CellSearch and Parsortix was comparable for cell lines, but Parsortix had a higher capture rate in real-life samples. Moreover, double enrichment with both CellSearch and Parsortix succeeded in removing most of the leukocyte contamination, resulting in an almost pure CMC sample suitable for genetic analysis. In this regard, a proof-of-concept analysis of CMCs from a paradigmatic case of a metastatic uveal melanoma patient led to the identification of multiple genetic alterations. In particular, the GNAQ p.Q209L was identified as homozygous, while a deletion in BAP1 exon 9 was found hemizygous. Moreover, an isochromosome 8 and a homozygous deletion of the CDKN2A gene were detected. In conclusion, we have optimized an approach to successfully enrich and retrieve viable CMCs from metastatic melanoma patients. Moreover, this study provides proof-of-principle for the feasibility of a marker-agnostic CMC enrichment followed by CMC phenotypic identification and genetic analysis.Kindly check and confirm the processed contributed equally is correctly identify We confirm.
Collapse
Affiliation(s)
- Cristina Poggiana
- Immunology and Molecular Oncology Unit, Veneto Institute of Oncology IOV-IRCCS, Padova, Italy
| | | | - Cristina Catoni
- Immunology and Molecular Oncology Unit, Veneto Institute of Oncology IOV-IRCCS, Padova, Italy.
| | - Ilaria Gallingani
- Immunology and Molecular Oncology Unit, Veneto Institute of Oncology IOV-IRCCS, Padova, Italy.
| | - Luisa Piccin
- Medical Oncology 2, Veneto Institute of Oncology, IOV-IRCCS, Padova, Italy
| | - Stefania Pellegrini
- Immunology and Molecular Oncology Unit, Veneto Institute of Oncology IOV-IRCCS, Padova, Italy
| | - Vittorio Aneloni
- UOC Immunotrasfusionale, University-Hospital of Padova, Padova, Italy
| | | | - Jacopo Pigozzo
- Medical Oncology 2, Veneto Institute of Oncology, IOV-IRCCS, Padova, Italy
| | - Alessio Fabozzi
- Oncology Unit 3, Veneto Institute of Oncology IOV-IRCCS, Padova, 35128, Italy
| | - Antonella Facchinetti
- Immunology and Molecular Oncology Unit, Veneto Institute of Oncology IOV-IRCCS, Padova, Italy
- Department of Surgery, Oncology and Gastroenterology, University of Padova, Padova, Italy
| | - Chiara Menin
- Immunology and Molecular Oncology Unit, Veneto Institute of Oncology IOV-IRCCS, Padova, Italy
| | - Paolo Del Fiore
- Soft-Tissue, Peritoneum and Melanoma Surgical Oncology Unit, Veneto Institute of Oncology IOV-IRCCS, Padova, Italy
| | - Simone Mocellin
- Department of Surgery, Oncology and Gastroenterology, University of Padova, Padova, Italy
- Soft-Tissue, Peritoneum and Melanoma Surgical Oncology Unit, Veneto Institute of Oncology IOV-IRCCS, Padova, Italy
| | | | - Antonio Rosato
- Immunology and Molecular Oncology Unit, Veneto Institute of Oncology IOV-IRCCS, Padova, Italy
- Department of Surgery, Oncology and Gastroenterology, University of Padova, Padova, Italy
| | - Maria Chiara Scaini
- Immunology and Molecular Oncology Unit, Veneto Institute of Oncology IOV-IRCCS, Padova, Italy
| |
Collapse
|
12
|
Vajpayee K, Paida V, Shukla RK. Nanoparticle-assisted PCR: fundamentals, mechanisms, and forensic implications. Int J Legal Med 2025; 139:945-964. [PMID: 39841191 DOI: 10.1007/s00414-024-03402-0] [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: 08/31/2024] [Accepted: 12/16/2024] [Indexed: 01/23/2025]
Abstract
Polymerase Chain Reaction (PCR) has transformed forensic DNA analysis but is still limited when dealing with compromised trace or inhibitor-containing samples. Nanotechnology has been integrated into nanoPCR (nanoparticle-assisted PCR) to overcome these obstacles. Nanomaterials improve PCR sensitivity, selectivity, and efficiency. Examples of these materials are semiconductor quantum dots and metal nanoparticles. They enhance DNA binding to primers, stabilize enzymes, and function as effective heat conductors, making accurate amplification possible even with tainted samples. The developments in nanoPCR have potential uses in forensics, as they allow for the more sensitive analysis of smaller, polluted, or deteriorated samples. Nevertheless, there are methodological and ethical issues. To provide credible and legitimate forensic evidence, rigorous validation and standardization of NanoPCR techniques are vital. The article addresses the relevant ethical and methodological aspects in forensic casework while examining the integration of nanotechnology into PCR.
Collapse
Affiliation(s)
- Kamayani Vajpayee
- Biological and Life Sciences, School of Arts and Sciences, Ahmedabad University, Central Campus, Navrangpura, Ahmedabad, Gujarat, India
| | - Vidhi Paida
- Biological and Life Sciences, School of Arts and Sciences, Ahmedabad University, Central Campus, Navrangpura, Ahmedabad, Gujarat, India
| | - Ritesh K Shukla
- Biological and Life Sciences, School of Arts and Sciences, Ahmedabad University, Central Campus, Navrangpura, Ahmedabad, Gujarat, India.
| |
Collapse
|
13
|
Tan W, Zhu Y, Chen S. Innovative approach to the detection of circulating tumor biomarkers: multi-dimensional application of liposome technology. Lipids Health Dis 2025; 24:160. [PMID: 40295973 PMCID: PMC12036244 DOI: 10.1186/s12944-025-02578-7] [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: 01/28/2025] [Accepted: 04/19/2025] [Indexed: 04/30/2025] Open
Abstract
Malignant tumors represent a significant worldwide health challenge, with elevated morbidity and mortality rates necessitating enhanced early identification and individualized treatment. Liposomes, as biomimetic lipid-based nanovesicles, have developed as a multifaceted platform for detecting and treating malignant tumors due to their excellent biocompatibility, stability, and membrane fusion properties. Circulating tumor markers, such as circulating tumor cells (CTCs), extracellular vesicles (EVs), circulating tumor proteins (CTPs), and circulating tumor nucleic acids (ctNAs), play a key role in early cancer diagnosis, disease progression monitoring, and personalized therapy. Liposome-based platforms enable effective molecular recognition, targeted detection, and signal amplification by targeting circulating tumor biomarkers, significantly increasing the potential for early tumor diagnosis and treatment. This review systematically summarizes advancements in the study of liposomes concerning circulating tumor markers, including applications in targeted recognition, early detection, and disease diagnosis, while discussing present problems and prospective applications of existing technology.
Collapse
Affiliation(s)
- Weichu Tan
- Department of Laboratory Medicine, Medical Research Center of Nanfang Hospital, School of Basic Medical Sciences, Guangdong Provincial Key Laboratory of Precision Medical Diagnostics, Guangdong Provincial Key Laboratory of Single-Cell and Extracellular Vesicles, Guangdong Engineering and Technology Research Center for Rapid Diagnostic Biosensors, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, People's Republic of China
| | - Yitong Zhu
- Department of Laboratory Medicine, Medical Research Center of Nanfang Hospital, School of Basic Medical Sciences, Guangdong Provincial Key Laboratory of Precision Medical Diagnostics, Guangdong Provincial Key Laboratory of Single-Cell and Extracellular Vesicles, Guangdong Engineering and Technology Research Center for Rapid Diagnostic Biosensors, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, People's Republic of China
| | - Siting Chen
- Department of Laboratory Medicine, Medical Research Center of Nanfang Hospital, School of Basic Medical Sciences, Guangdong Provincial Key Laboratory of Precision Medical Diagnostics, Guangdong Provincial Key Laboratory of Single-Cell and Extracellular Vesicles, Guangdong Engineering and Technology Research Center for Rapid Diagnostic Biosensors, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, People's Republic of China.
- The Affiliated Qingyuan Hospital (Qingyuan People's Hospital), Guangzhou Medical University, Qingyuan, Guangdong, 511518, People's Republic of China.
| |
Collapse
|
14
|
Wang J, Xu J, Liu X, Li X, Xu Z. A microfluidic chip incorporating magnetic sorting and invasive separation for isolation, culture and telomerase analysis of circulating tumor cells. Talanta 2025; 285:127316. [PMID: 39644673 DOI: 10.1016/j.talanta.2024.127316] [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: 08/18/2024] [Revised: 11/28/2024] [Accepted: 11/30/2024] [Indexed: 12/09/2024]
Abstract
Circulating tumor cells (CTCs) are a crucial indicator of cancer metastasis, and are vital for early diagnosis, disease monitoring, and treatment response evaluation. However, their extremely low concentration and the complexities of isolation techniques pose a significant challenge in capturing and analyzing CTCs. In this study, we developed a novel microfluidic system that integrates magnetic capture and invasive screening onto a single microfluidic chip. By attaching positively charged magnetic nanoparticles to negatively charged CTCs, the magnetic separation of CTCs within the chip effectively eliminates interference from blood cells. A total of 2 mL blood sample can be processed within 3 min, achieving an impressive tumor capture efficiency of 84 %. Using the chip, we also successfully achieved long-term culture of CTCs, and identified CTCs with high activity and invasive potential in blood samples from 11 patients with colorectal cancer. Finally, we analyzed telomerase activity in cultured CTCs on the microfluidic chip. Significantly higher invasive potential and telomerase activity were observed in CTCs from the malignant tumor group compared to the benign group (P < 0.01), highlighting their increased aggressiveness. This study offers a novel approach for efficient CTCs isolation, culture, and telomerase analysis, clarifying the crucial role of telomerase in tumor metastasis and providing profound insights for future research on telomerase-targeted tumor metastasis.
Collapse
Affiliation(s)
- Jie Wang
- Research Center for Analytical Sciences, Northeastern University, Shenyang, 110819, PR China
| | - Jiali Xu
- Research Center for Analytical Sciences, Northeastern University, Shenyang, 110819, PR China
| | - Xiaopeng Liu
- Research Center for Analytical Sciences, Northeastern University, Shenyang, 110819, PR China
| | - Xin Li
- Department of Anesthesiology, Liaoning Cancer Hospital and Institute, Shenyang, 110042, PR China
| | - Zhangrun Xu
- Research Center for Analytical Sciences, Northeastern University, Shenyang, 110819, PR China.
| |
Collapse
|
15
|
Martella S, Wekking D, Lai E, Lambertini M, Pettinato A, Parrino A, Semonella F, Sanna G, Maccioni A, Scartozzi M, Addeo A, Solinas C. Liquid biopsy: An innovative tool in oncology. Where do we stand? Semin Oncol 2025; 52:152343. [PMID: 40233447 DOI: 10.1016/j.seminoncol.2025.152343] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/23/2024] [Revised: 03/12/2025] [Accepted: 03/13/2025] [Indexed: 04/17/2025]
Abstract
The Liquid Biopsy (LB) represents an ideal surrogate of tumor Tissue Biopsy (TB) when the aim is to obtain useful information on patient prognosis and personalized therapy. This technique renders it possible to isolate circulating tumor cells, circulating tumor DNA and other molecules from biological fluids. The most commonly used fluid for liquid biopsy is blood, but depending on the case it could be necessary to isolate the tumor components from other biological fluids such as urine, pleural effusion, cerebrospinal fluid, and others. The main advantages of liquid biopsy are the minimally invasive nature of the procedure and the possibility of analyzing all tumor clones. Limitations include difficulties in the isolation of tumor components and the requirement for highly sensitive analysis methods to avoid the risk of technical artifacts. In our review we will focus on describing circulating tumor biomarkers to illustrate the variety of information that can be obtained from biological fluids, particularly blood. We will then discuss the advanced biotechnological techniques suitable for the identification and analysis of Circulating Tumor DNA (ctDNA), examining both the potential and limitations of analytical methods and the clinical applicability of liquid biopsy for cancer diagnosis, monitoring, and therapeutic prediction. Additionally, we will explore strategies to enhance this valuable alternative to the more invasive tissue biopsy, with a dedicated focus on ongoing clinical studies, currently approved tests, and guideline recommendations.
Collapse
Affiliation(s)
- Serafina Martella
- University of Catania Department of Biomedical and Biotechnological Sciences, Catania, Italy
| | - Demi Wekking
- Location Academic Medical Centre, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| | - Eleonora Lai
- Medical Oncology Unit, University Hospital and University of Cagliari, Cagliari, Italy
| | - Matteo Lambertini
- Department of Internal Medicine and Medical Specialties, School of Medicine, University of Genoa, Genoa, Italy; Department of Medical Oncology, U.O. Clinica di Oncologia Medica, IRCCS Ospedale Policlinico San Martino, Genoa, Italy
| | | | - Alissa Parrino
- Medical Oncology Unit, University Hospital and University of Cagliari, Cagliari, Italy
| | | | | | | | | | - Alfredo Addeo
- Oncology Department, University Hospital Geneva (HUG), Geneva, Switzerland
| | - Cinzia Solinas
- Medical Oncology, AOU Cagliari, Policlinico Duilio Casula Monserrato (CA), Cagliari, Italy.
| |
Collapse
|
16
|
Karageorgos FF, Karakasi KE, Kofinas A, Antoniadis N, Katsanos G, Tsoulfas G. Evolving Transplant Oncology: Evolving Criteria for Better Decision-Making. Diagnostics (Basel) 2025; 15:820. [PMID: 40218170 PMCID: PMC11988714 DOI: 10.3390/diagnostics15070820] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2024] [Revised: 03/10/2025] [Accepted: 03/21/2025] [Indexed: 04/14/2025] Open
Abstract
Transplant oncology integrates a wide variety of fields, such as surgery, oncology, and transplant medicine, intending to increase the range of studies and treatments for hepatobiliary cancers and other liver-related malignant lesions. Liver transplantation (LT) has proven to be an effective treatment for hepatocellular carcinoma. While the Milan criteria are still the gold standard, several new, more inclusive criteria have been proposed, and hepatocellular carcinoma has become a major indication for liver transplantation. The continuous evolution of diagnostic technologies supported this with higher image quality and more accurate staging. This review describes the current applications of transplant oncology in hepatocellular carcinoma, cholangiocarcinoma, neuroendocrine tumors, and liver metastatic disease from colorectal cancer and discusses the path that led to the development of transplant oncology as an organized approach to managing gastrointestinal malignancies through transplantation. More importantly, the significance of a multidisciplinary approach and criteria in the selection of suitable candidates are discussed. In addition, newer aspects of transplant oncology, such as immunotherapy, circulating tumor DNA (ctDNA), novel surgical techniques, and the utilization of artificial intelligence, are presented. Finally, the opportunities and challenges involved in the field's future, as well as the evolution of the criteria used over the years and insightful thoughts for the future of the criteria, are discussed.
Collapse
Affiliation(s)
| | | | | | | | | | - Georgios Tsoulfas
- Department of Transplantation Surgery, Center for Research and Innovation in Solid Organ Transplantation, Aristotle University School of Medicine, 54642 Thessaloniki, Greece; (F.F.K.)
| |
Collapse
|
17
|
Bader KB, Padilla F, Haworth KJ, Ellens N, Dalecki D, Miller DL, Wear KA. Overview of Therapeutic Ultrasound Applications and Safety Considerations: 2024 Update. JOURNAL OF ULTRASOUND IN MEDICINE : OFFICIAL JOURNAL OF THE AMERICAN INSTITUTE OF ULTRASOUND IN MEDICINE 2025; 44:381-433. [PMID: 39526313 PMCID: PMC11796337 DOI: 10.1002/jum.16611] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/11/2024] [Revised: 10/11/2024] [Accepted: 10/19/2024] [Indexed: 11/16/2024]
Abstract
A 2012 review of therapeutic ultrasound was published to educate researchers and physicians on potential applications and concerns for unintended bioeffects (doi: 10.7863/jum.2012.31.4.623). This review serves as an update to the parent article, highlighting advances in therapeutic ultrasound over the past 12 years. In addition to general mechanisms for bioeffects produced by therapeutic ultrasound, current applications, and the pre-clinical and clinical stages are outlined. An overview is provided for image guidance methods to monitor and assess treatment progress. Finally, other topics relevant for the translation of therapeutic ultrasound are discussed, including computational modeling, tissue-mimicking phantoms, and quality assurance protocols.
Collapse
Affiliation(s)
| | - Frederic Padilla
- Gene Therapy ProgramFocused Ultrasound FoundationCharlottesvilleVirginiaUSA
- Department of RadiologyUniversity of Virginia Health SystemCharlottesvilleVirginiaUSA
| | - Kevin J. Haworth
- Department of PediatricsUniversity of CincinnatiCincinnatiOhioUnited States
- Department of Internal MedicineUniversity of CincinnatiCincinnatiOhioUSA
- Department of Biomedical EngineeringUniversity of CincinnatiCincinnatiOhioUSA
| | | | - Diane Dalecki
- Department of Biomedical EngineeringUniversity of RochesterRochesterNew YorkUSA
| | - Douglas L. Miller
- Department of RadiologyUniversity of Michigan Health SystemAnn ArborMichiganUSA
| | - Keith A. Wear
- Center for Devices and Radiological HealthU.S. Food and Drug AdministrationSilver SpringMarylandUSA
| |
Collapse
|
18
|
Francis JH. Histiocytosis Advancements Parallel Ophthalmic Innovations: The LXXXI Edward Jackson Memorial Lecture. Am J Ophthalmol 2025; 271:104-118. [PMID: 39536851 DOI: 10.1016/j.ajo.2024.10.030] [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: 10/21/2024] [Accepted: 10/25/2024] [Indexed: 11/16/2024]
Abstract
PURPOSE To highlight innovations in ophthalmic oncology through histiocytosis advancements. DESIGN Perspective and retrospective review. METHODS The literature outlining the recent advancements in histiocytosis and ocular oncology was reviewed and combined with trial data and personal recollection. Intersections between these two fields were discussed. RESULTS The understanding of genetic mutations in disease-both in which cells they occur and the timing of mutation development-has expanded in tandem for the fields of ophthalmic oncology and histiocytosis. Similarly, advancements in diagnostic and treatment technology in one field can help patients in the other. For example, in one study, cell-free DNA testing reliably detected mutations in 14 of 18 (78%) patients with suspected histiocytosis. This technique has also been used in ophthalmic oncology as an alternative to invasive biopsy to avoid the risk of tumor externalization, vision impairment, and other side effects. These and other advancements have allowed both fields to utilize targeted agents to successfully treat diseases with an actionable mutation; or deliver more targeted chemotherapy via the intraarterial technique. CONCLUSIONS The explosion of molecular genetics technology and targeted therapies has revolutionized cancer treatment, including histiocytosis and ophthalmic oncology. Recent progress in both fields has shown how these seemingly disparate areas have many intersections, and this speaks to the collaborative spirit that is inherent in clinical research.
Collapse
Affiliation(s)
- Jasmine H Francis
- From the Department of Surgery (J.H.F.), Ophthalmic Oncology Service, Memorial Sloan Kettering Cancer Center, New York, New York, USA.
| |
Collapse
|
19
|
Escalona G, Ocadiz‐Ruiz R, Ma JA, Schrack IA, Ross BC, Morrison AK, Jeruss JS, Shea LD. Design Principles of an Engineered Metastatic Niche for Monitoring of Cancer Progression. Biotechnol Bioeng 2025; 122:631-641. [PMID: 39628034 PMCID: PMC11808458 DOI: 10.1002/bit.28895] [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: 08/26/2024] [Revised: 10/29/2024] [Accepted: 11/18/2024] [Indexed: 02/11/2025]
Abstract
Across many types of cancer, metastatic disease is associated with a substantial decrease in 5-year survival rates relative to only a localized primary tumor. Many patients self-report metastatic disease due to disruption of normal organ or tissue function, and earlier detection could enable treatment with a lower burden of disease. We have previously reported a subcutaneous biomaterial implant for early detection by serving as an engineered metastatic niche, which has been reported to recruit tumor cells before colonization of solid organs. In this report, we investigated the design principles of the scaffold and defined the conditions for use in disease detection. Using the metastatic 4T1 triple-negative breast cancer model, we identified that a porous structure was essential to capture tumor and immune cells. Scaffolds of multiple diameters were investigated for their ability to serve as a metastatic niche, with a porous scaffold with a diameter as small as 2 mm identifying disease accurately. Additionally, scaffolds that had been in vivo for 1-5 weeks were able to identify disease accurately. Finally, the sensitivity of the scaffold relative to liquid biopsies was analyzed, with scaffolds accurately detecting disease at earlier time points than liquid biopsy. Collectively, these studies inform the design principles and use conditions for porous scaffolds to detect metastatic disease.
Collapse
Affiliation(s)
- Guillermo Escalona
- Department of Biomedical EngineeringUniversity of MichiganAnn ArborMichiganUSA
| | - Ramon Ocadiz‐Ruiz
- Department of Biomedical EngineeringUniversity of MichiganAnn ArborMichiganUSA
| | - Jeffrey A. Ma
- Department of Biomedical EngineeringUniversity of MichiganAnn ArborMichiganUSA
| | - Ian A. Schrack
- Department of Biomedical EngineeringUniversity of MichiganAnn ArborMichiganUSA
| | - Brian C. Ross
- Department of Biomedical EngineeringUniversity of MichiganAnn ArborMichiganUSA
| | - Alexis K. Morrison
- Department of Biomedical EngineeringUniversity of MichiganAnn ArborMichiganUSA
| | - Jacqueline S. Jeruss
- Department of Biomedical EngineeringUniversity of MichiganAnn ArborMichiganUSA
- Department of SurgeryUniversity of MichiganAnn ArborMichiganUSA
| | - Lonnie D. Shea
- Department of Biomedical EngineeringUniversity of MichiganAnn ArborMichiganUSA
- Department of SurgeryUniversity of MichiganAnn ArborMichiganUSA
- Department of Chemical EngineeringUniversity of MichiganAnn ArborMichiganUSA
| |
Collapse
|
20
|
Liu X, Zhao G, Mao W, Li Q, Liao J, He G. Clinical significance of fecal Syndecan-2 gene methylation combined with blood tumor abnormal protein detection in the diagnosis of colorectal cancer and precancerous lesions. Clin Biochem 2025; 136:110887. [PMID: 39832542 DOI: 10.1016/j.clinbiochem.2025.110887] [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: 10/23/2024] [Revised: 01/09/2025] [Accepted: 01/10/2025] [Indexed: 01/22/2025]
Abstract
OBJECTIVE To investigate the clinical significance of fecal Syndecan-2 (SDC2) gene methylation combined with blood tumor abnormal protein (TAP) detection for the diagnosis of colorectal cancer (CRC) and its precancerous lesions. METHODS A retrospective study was conducted to collect patients diagnosed with CRC or colorectal adenoma (Ade) from March 2020 to March 2023, and healthy people (Nor) without any gastrointestinal diseases during the same period as the control group. All participants underwent the fecal SDC2 gene methylation test, blood TAP test and fecal occult blood test (FOBT). The differences in the positivity rates of each index were compared, receiver operator characteristic curves were plotted and the area under the curve (AUC) was calculated to evaluate the diagnostic effects of different testing methods on CRC and its precancerous lesions. RESULTS A total of 146 individuals were included in the study, including 69 CRC patients, 47 patients with Ade and 30 healthy individuals. The results showed that, SDC2, TAP and the combined assay had high comprehensive diagnostic efficacy for the diagnosis of CRC, but there was no significant difference between the three methods in terms of AUC, sensitivity, and specificity. However, for Ade, the combined detection was statistically significant, with a high AUC (0.905), high sensitivity (95.7%), and high specificity (86.7%). CONCLUSION Fecal SDC2 gene methylation combined with blood TAP detection is an effective noninvasive screening and diagnostic method to enhance the early detection and treatment of CRC precancerous lesions, such as Ade, thereby reducing the incidence and mortality of CRC.
Collapse
Affiliation(s)
- Xuanjun Liu
- Department of Gastrointestinal Surgery, Yongchuan Hospital of Chongqing Medical University, Yongchuan, Chongqing 402160, China
| | - Guowei Zhao
- Department of Gastrointestinal Surgery, Yongchuan Hospital of Chongqing Medical University, Yongchuan, Chongqing 402160, China
| | - Weixu Mao
- Department of Respiratory Medicine, The Affiliated Yongchuan District Traditional Chinese Medicine Hospital of Chongqing Medical University, Yongchuan District, Chongqing 402160, China
| | - Qigang Li
- Department of Gastrointestinal Surgery, Yongchuan Hospital of Chongqing Medical University, Yongchuan, Chongqing 402160, China
| | - Juan Liao
- Central Laboratory, Yongchuan Hospital of Chongqing Medical University, Yongchuan, Chongqing 402160, China
| | - Gan He
- Department of Gastrointestinal Surgery, Yongchuan Hospital of Chongqing Medical University, Yongchuan, Chongqing 402160, China.
| |
Collapse
|
21
|
Gupta A, Bhardwaj S, Ghorai S, Ahmed R, Agarwal S, Mukherjee G, Desai KV. Potential applications of gene expression profiles obtained from circulating extracellular vesicles in breast cancer. THE JOURNAL OF LIQUID BIOPSY 2025; 7:100287. [PMID: 40027231 PMCID: PMC11863812 DOI: 10.1016/j.jlb.2025.100287] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/02/2024] [Revised: 01/14/2025] [Accepted: 01/14/2025] [Indexed: 03/05/2025]
Abstract
Background Liquid biopsy-based biomarkers offer several advantages since they are minimally invasive, can be useful in longitudinal monitoring of the disease and have higher patient compliance. We describe a protocol using minimal volumes of archival and prospective serum/plasma samples to define the RNA contents of EVs and discuss its benefits and limitations. Methods RNA-seq analysis of matched tumor biopsy, circulating EVs from breast cancer patients (EV-C, n = 26) and healthy donors (EV-H, n = 4) was performed and differentially expressed genes were validated by RT-PCR in a separate series of samples (EV-C, n = 32 and EV-H, n = 22). A total of 84 samples were studied. Results RNA-seq data from 500 μl serum samples yielded more than 17000 genes, of which 320 were DEGs (adjusted p value ≤ 0.05) between EV-C and EV-H samples. Pathways for Myc V1, reactive oxygen species, angiogenesis, allograft rejection and Interferon regulated genes were over-represented in EV-C samples. Computational deconvolution algorithms for cell signatures identified immune cells such as Th1 and memory T-cells, endothelial cells, and osteoblasts from the stromal compartment as significant. Top 6 genes were validated by qRT-PCR in all samples (n = 84) and they consistently and correctly classified cancer and healthy groups. An independent set of 374 and 640 DEGs could segregate ER positive/ER negative and non-metastatic versus metastatic samples, respectively. EVs from metastatic samples had higher variability in gene expression patterns whereas those from non-metastatic samples showed a better correlation. Conclusion By using low serum amounts successfully for EV transcriptomics, we demonstrate that a minimally invasive technique could be converted to a microinvasive format. These data lay the foundation for EV RNA based biomarker discovery for segregating breast cancers.
Collapse
Affiliation(s)
- Aritra Gupta
- Biotechnology Research Innovation Council-National Institute of Biomedical Genomics (BRIC-NIBMG), Kalyani, India
- Regional Centre for Biotechnology, PhD Program, India
| | - Siddharth Bhardwaj
- Biotechnology Research Innovation Council-National Institute of Biomedical Genomics (BRIC-NIBMG), Kalyani, India
| | - Sayan Ghorai
- Biotechnology Research Innovation Council-National Institute of Biomedical Genomics (BRIC-NIBMG), Kalyani, India
- Regional Centre for Biotechnology, PhD Program, India
| | - Rosina Ahmed
- Tata Medical Centre, 14 MAR (DH Block), New Town, Rajarhat, Kolkata, 700160, India
| | - Sanjit Agarwal
- Tata Medical Centre, 14 MAR (DH Block), New Town, Rajarhat, Kolkata, 700160, India
| | - Geetashree Mukherjee
- Tata Medical Centre, 14 MAR (DH Block), New Town, Rajarhat, Kolkata, 700160, India
| | - Kartiki V. Desai
- Biotechnology Research Innovation Council-National Institute of Biomedical Genomics (BRIC-NIBMG), Kalyani, India
| |
Collapse
|
22
|
Pérez-Cabello JA, Artero-Castro A, Molina-Pinelo S. Small cell lung cancer unveiled: Exploring the untapped resource of circulating tumor cells-derived organoids. Crit Rev Oncol Hematol 2025; 207:104622. [PMID: 39832682 DOI: 10.1016/j.critrevonc.2025.104622] [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: 10/01/2024] [Revised: 01/09/2025] [Accepted: 01/14/2025] [Indexed: 01/22/2025] Open
Abstract
Small cell lung cancer (SCLC) remains a challenge in oncology due to its aggressive behavior and dismal prognosis. Despite advances in treatments, novel strategies are urgently needed. Enter liquid biopsy-a game-changer in SCLC management. This revolutionary non-invasive approach allows for the analysis of circulating tumor cells (CTCs), offering insights into tumor behavior and treatment responses. Our review focuses on a groundbreaking frontier: harnessing CTCs to create three-dimensional (3D) organoid models. These models, derived from CTCs that break away from the primary tumor or metastatic locations, hold immense potential for revolutionizing cancer research, especially in SCLC. We explore the essential conditions for successfully establishing CTC-derived organoids-a transformative approach with profound implications for personalized medicine. Our evaluation spans diverse isolation techniques, shedding light on their advantages and limitations. Furthermore, we uncover the critical factors governing the cultivation of 3D organoids from CTCs, meticulously mimicking the tumor microenvironment. This review comprehensively elucidates the molecular characterization of these organoids, showcasing their potential in identifying treatment targets and predicting responses. In essence, our review amalgamates cutting-edge methodologies for isolating CTCs, establishing transformative CTC-derived organoids, and characterizing their molecular landscape. This represents a promising frontier for advancing personalized medicine in the complex realm of SCLC management and holds significant implications for translational research.
Collapse
Affiliation(s)
- Jesús A Pérez-Cabello
- Institute of Biomedicine of Seville (IBiS), HUVR, CSIC, University of Seville, Seville 41013, Spain
| | - Ana Artero-Castro
- Institute of Biomedicine of Seville (IBiS), HUVR, CSIC, University of Seville, Seville 41013, Spain
| | - Sonia Molina-Pinelo
- Institute of Biomedicine of Seville (IBiS), HUVR, CSIC, University of Seville, Seville 41013, Spain; Spanish Center for Biomedical Research Network in Oncology (CIBERONC), Madrid 28029, Spain.
| |
Collapse
|
23
|
Nguyen Phuong L, Salas S, Benzekry S. Computational Modeling for Circulating Cell-Free DNA in Clinical Oncology. JCO Clin Cancer Inform 2025; 9:e2400224. [PMID: 40020203 DOI: 10.1200/cci-24-00224] [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: 08/31/2024] [Revised: 11/23/2024] [Accepted: 01/10/2025] [Indexed: 05/12/2025] Open
Abstract
PURPOSE Liquid biopsy, specifically circulating cell-free DNA (cfDNA), has emerged as a powerful tool for cancer early diagnosis, prognosis, and treatment monitoring over a wide range of cancer types. Computational modeling (CM) of cfDNA data is essential to harness its full potential for real-time, noninvasive insights into tumor biology, enhancing clinical decision making. DESIGN This work reviews CM-cfDNA methods applied to clinical oncology, emphasizing both machine learning (ML) techniques and mechanistic approaches. The latter integrate biological principles, enabling a deeper understanding of cfDNA dynamics and its relationship with tumor evolution. RESULTS Key findings highlight the effectiveness of CM-cfDNA approaches in improving diagnostic accuracy, identifying prognostic markers, and predicting therapeutic outcomes. ML models integrating cfDNA concentration, fragmentation patterns, and mutation detection achieve high sensitivity and specificity for early cancer detection. Mechanistic models describe cfDNA kinetics, linking them to tumor growth and response to treatment, for example, immune checkpoint inhibitors. Longitudinal data and advanced statistical constructs further refine these models for quantification of interindividual and intraindividual variability. CONCLUSION CM-cfDNA represents a pivotal advancement in precision oncology. It bridges the gap between extensive cfDNA data and actionable clinical insights, supporting its integration into routine cancer care. Future efforts should focus on standardizing protocols, validating models across populations, and exploring hybrid approaches combining ML with mechanistic modeling to improve biological understanding.
Collapse
Affiliation(s)
- Linh Nguyen Phuong
- Computational Pharmacology and Clinical Oncology Department, Centre Inria d'Université Côte d'Azur, Cancer Research Centre of Marseille, Paoli Calmettes Institute, Inserm UMR1068, CNRS UMR7258, Aix Marseille University UM105, Marseille, France
| | - Sébastien Salas
- Computational Pharmacology and Clinical Oncology Department, Centre Inria d'Université Côte d'Azur, Cancer Research Centre of Marseille, Paoli Calmettes Institute, Inserm UMR1068, CNRS UMR7258, Aix Marseille University UM105, Marseille, France
- Assistance Publique-Hôpitaux de Marseille, Timone Hospital, Aix Marseille University, Marseille, France
| | - Sébastien Benzekry
- Computational Pharmacology and Clinical Oncology Department, Centre Inria d'Université Côte d'Azur, Cancer Research Centre of Marseille, Paoli Calmettes Institute, Inserm UMR1068, CNRS UMR7258, Aix Marseille University UM105, Marseille, France
| |
Collapse
|
24
|
Joshi R, Ahmadi H, Gardner K, Bright RK, Wang W, Li W. Advances in microfluidic platforms for tumor cell phenotyping: from bench to bedside. LAB ON A CHIP 2025; 25:856-883. [PMID: 39774602 PMCID: PMC11859771 DOI: 10.1039/d4lc00403e] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/11/2025]
Abstract
Heterogeneities among tumor cells significantly contribute towards cancer progression and therapeutic inefficiency. Hence, understanding the nature of cancer through liquid biopsies and isolation of circulating tumor cells (CTCs) has gained considerable interest over the years. Microfluidics has emerged as one of the most popular platforms for performing liquid biopsy applications. Various label-free and labeling techniques using microfluidic platforms have been developed, the majority of which focus on CTC isolation from normal blood cells. However, sorting and profiling of various cell phenotypes present amongst those CTCs is equally important for prognostics and development of personalized therapies. In this review, firstly, we discuss the biophysical and biochemical heterogeneities associated with tumor cells and CTCs which contribute to cancer progression. Moreover, we discuss the recently developed microfluidic platforms for sorting and profiling of tumor cells and CTCs. These techniques are broadly classified into biophysical and biochemical phenotyping methods. Biophysical methods are further classified into mechanical and electrical phenotyping. While biochemical techniques have been categorized into surface antigen expressions, metabolism, and chemotaxis-based phenotyping methods. We also shed light on clinical studies performed with these platforms over the years and conclude with an outlook for the future development in this field.
Collapse
Affiliation(s)
- Rutwik Joshi
- Department of Chemical Engineering, Texas Tech University, Lubbock, TX 79409, USA.
| | - Hesaneh Ahmadi
- Department of Chemical Engineering, Texas Tech University, Lubbock, TX 79409, USA.
| | - Karl Gardner
- Department of Chemical Engineering, Texas Tech University, Lubbock, TX 79409, USA.
| | - Robert K Bright
- Department of Immunology & Molecular Microbiology, School of Medicine & Cancer Center, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA
| | - Wenwen Wang
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, China
| | - Wei Li
- Department of Chemical Engineering, Texas Tech University, Lubbock, TX 79409, USA.
| |
Collapse
|
25
|
Yin P, Peng Z, Wang Q, Duan Y, Hu B, Lin Q. A rapid dual-mode SERS/FL cytosensor assisted via DNA Walker-based plasmonic nanostructures. SPECTROCHIMICA ACTA. PART A, MOLECULAR AND BIOMOLECULAR SPECTROSCOPY 2025; 327:125416. [PMID: 39550818 DOI: 10.1016/j.saa.2024.125416] [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: 06/20/2024] [Revised: 10/10/2024] [Accepted: 11/06/2024] [Indexed: 11/19/2024]
Abstract
Various surface-enhanced Raman scattering (SERS) biosensors offer powerful tools for the ultrasensitive detection of circulating tumor cells (CTCs) and tumor diagnosis. Despite their efficacy, the swift and precise preparation of SERS plasmonic nanostructures poses an ongoing challenge. In this study, we introduce DNA-assisted plasmonic nanostructures capable of producing dual signals and facilitating DNA Walker signal amplification, resulting in the development of a SERS/Fluorescent (FL) dual-mode cytosensor for CTCs detection. Firstly, Au@Ag nanoparticle multimers (Au@AgNMs) featuring interparticle nano-gaps were synthesized through DNA self-assembly and in-situ deposition, which provided plasmonic nanostructures. Hence, the nano-gap distance among Au@AgNMs was meticulously regulated after optimization to achieve both SERS enhancement and fluorescence quenching. Subsequently, the aptamer (Apt) of MUC1 recognized CTCs specifically for strand displacement reaction (SDR) and further triggered the DNA Walker reaction for signal amplification. The limit of detection (LOD) of proposed cytosensor can be obtained as low as 5 cells/mL in SERS mode and 21 cells/mL in FL mode. Hence, SERS mode confers highly precise information, while FL mode allow for rapid quantitative analysis. This dual-mode cytosensor based on plasmonic nanostructures facilitates the early detection and precise treatment of cancer or infectious diseases.
Collapse
Affiliation(s)
- Pengkun Yin
- Research Center of Analytical Instrumentation, School of Mechanical Engineering, Sichuan University, Chengdu 610064, China
| | - Zhengying Peng
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610061, China
| | - Qihui Wang
- College of Chemistry and Life Science, Sichuan Provincial Key Laboratory for Structural Optimization and Application of Functional Molecules, Chengdu Normal University, Chengdu 611130, China
| | - Yixiang Duan
- Research Center of Analytical Instrumentation, School of Mechanical Engineering, Sichuan University, Chengdu 610064, China
| | - Bin Hu
- Department of Thoracic Surgery, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, School of Medicine, University of Electronic Science and Technology of China, Chengdu 610041, China.
| | - Qingyu Lin
- Research Center of Analytical Instrumentation, School of Mechanical Engineering, Sichuan University, Chengdu 610064, China.
| |
Collapse
|
26
|
Yang S, Seo J, Choi J, Kim SH, Kuk Y, Park KC, Kang M, Byun S, Joo JY. Towards understanding cancer dormancy over strategic hitching up mechanisms to technologies. Mol Cancer 2025; 24:47. [PMID: 39953555 PMCID: PMC11829473 DOI: 10.1186/s12943-025-02250-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2024] [Accepted: 01/28/2025] [Indexed: 02/17/2025] Open
Abstract
Delving into cancer dormancy has been an inherent task that may drive the lethal recurrence of cancer after primary tumor relief. Cells in quiescence can survive for a short or long term in silence, may undergo genetic or epigenetic changes, and can initiate relapse through certain contextual cues. The state of dormancy can be induced by multiple conditions including cancer drug treatment, in turn, undergoes a life cycle that generally occurs through dissemination, invasion, intravasation, circulation, immune evasion, extravasation, and colonization. Throughout this cascade, a cellular machinery governs the fate of individual cells, largely affected by gene regulation. Despite its significance, a precise view of cancer dormancy is yet hampered. Revolutionizing advanced single cell and long read sequencing through analysis methodologies and artificial intelligence, the most recent stage in the research tool progress, is expected to provide a holistic view of the diverse aspects of cancer dormancy.
Collapse
Affiliation(s)
- Sumin Yang
- Department of Pharmacy, College of Pharmacy, Hanyang University, Ansan, Gyeonggi-do, 15588, Korea
| | - Jieun Seo
- Genomic Medicine Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, 34141, Korea
- Department of Functional Genomics, University of Science and Technology, Daejeon, 34113, Korea
| | - Jeonghyeon Choi
- Department of Pharmacy, College of Pharmacy, Hanyang University, Ansan, Gyeonggi-do, 15588, Korea
| | - Sung-Hyun Kim
- Department of Pharmacy, College of Pharmacy, Hanyang University, Ansan, Gyeonggi-do, 15588, Korea
| | - Yunmin Kuk
- Genomic Medicine Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, 34141, Korea
- Department of Functional Genomics, University of Science and Technology, Daejeon, 34113, Korea
| | - Kyung Chan Park
- Genomic Medicine Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, 34141, Korea
- Department of Functional Genomics, University of Science and Technology, Daejeon, 34113, Korea
| | - Mingon Kang
- Department of Computer Science, University of Nevada, Las Vegas, NV, 89154, USA
| | - Sangwon Byun
- Genomic Medicine Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, 34141, Korea.
- Department of Functional Genomics, University of Science and Technology, Daejeon, 34113, Korea.
| | - Jae-Yeol Joo
- Department of Pharmacy, College of Pharmacy, Hanyang University, Ansan, Gyeonggi-do, 15588, Korea.
- Department of Pharmacy, College of Pharmacy, Hanyang University, Rm 407, Bldg.42, 55 Hanyangdaehak-ro, Sangnok-gu Ansan, Gyeonggi-do, 15588, Republic of Korea.
| |
Collapse
|
27
|
Chiang IH, Kuo HC, Liao CT, Kuo YC, Yu SM, Wang HM, Huang YH, Nguyen Thi KA, Wu MH, Hsieh JCH. An Ex vivo cultivation model for circulating tumor cells: The success rate and correlations with cancer response to therapy. Biomed J 2025; 48:100819. [PMID: 39622435 PMCID: PMC11743102 DOI: 10.1016/j.bj.2024.100819] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2024] [Revised: 11/10/2024] [Accepted: 11/21/2024] [Indexed: 12/31/2024] Open
Abstract
BACKGROUND Cancer mortality is closely linked to recurrence and distant metastasis, posing challenges in real-time tracking due to the invasiveness of current methods. Circulating tumor cells (CTCs) show promise as potential tools; however, their scarcity remains a significant obstacle. METHOD In this prospective study, we validated a simple culture protocol and investigated the correlation between clinical response and CTC growth status. Following negative selection, the isolated cells were subjected to ex vivo cultivation in a two-dimensional environment supplemented with cytokines for up to 21 days, followed by immunofluorescence staining for analysis. RESULTS Among 37 participants with solid tumors and distant metastasis (34.8% head and neck cancer), 47 samples were collected, from which CTCs were detected. The percentages of CTCs, atypical CTCs, and white blood cells during cultivation from days 7-21 were significantly different (p < 0.001, p < 0.001, and p = 0.330, respectively). Patients were further categorized into progressive disease (PD) and non-PD groups based on disease status, revealing significant differences in CTC growth rates, which increases from Days 7-21 between groups (5.5x vs. 2.8x growth, respectively; p < 0.001). CONCLUSION With the proposed protocols, we cultured CTCs from patients with various cancers for 21 days and identified a tool for predicting cancer response. The actual cancer status (PD or non-PD) at CTC isolation correlates to CTC growth rate, guiding the required observation time and parameters for culture.
Collapse
Affiliation(s)
- I-Hsuan Chiang
- Department of Traditional Chinese Medicine, Chang Gung Memorial Hospital, Linkou, Taoyuan, Taiwan; College of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Hsuan-Chih Kuo
- College of Medicine, Chang Gung University, Taoyuan, Taiwan; Division of Hematology-Oncology, Department of Internal Medicine, New Taipei Municipal TuCheng Hospital, New Taipei City, Taiwan
| | - Chun-Ta Liao
- College of Medicine, Chang Gung University, Taoyuan, Taiwan; Department of Otorhinolaryngology, Chang Gung Memorial Hospital, Linkou, Taoyuan, Taiwan
| | - Yung-Chia Kuo
- College of Medicine, Chang Gung University, Taoyuan, Taiwan; Division of Hematology-Oncology, Department of Internal Medicine, New Taipei Municipal TuCheng Hospital, New Taipei City, Taiwan; Division of Hematology-Oncology, Department of Internal Medicine, Chang Gung Memorial Hospital, Linkou, Taoyuan, Taiwan
| | - Shao-Ming Yu
- College of Medicine, Chang Gung University, Taoyuan, Taiwan; Division of Hematology-Oncology, Department of Internal Medicine, New Taipei Municipal TuCheng Hospital, New Taipei City, Taiwan; Division of Hematology-Oncology, Department of Internal Medicine, Chang Gung Memorial Hospital, Linkou, Taoyuan, Taiwan
| | - Hung-Ming Wang
- College of Medicine, Chang Gung University, Taoyuan, Taiwan; Division of Hematology-Oncology, Department of Internal Medicine, Chang Gung Memorial Hospital, Linkou, Taoyuan, Taiwan
| | - Yi-Hui Huang
- Division of Hematology-Oncology, Department of Internal Medicine, New Taipei Municipal TuCheng Hospital, New Taipei City, Taiwan; Division of Hematology-Oncology, Department of Internal Medicine, Chang Gung Memorial Hospital, Linkou, Taoyuan, Taiwan
| | - Kim Anh Nguyen Thi
- College of Medicine, Chang Gung University, Taoyuan, Taiwan; Division of Hematology-Oncology, Department of Internal Medicine, New Taipei Municipal TuCheng Hospital, New Taipei City, Taiwan; Division of Hematology-Oncology, Department of Internal Medicine, Chang Gung Memorial Hospital, Linkou, Taoyuan, Taiwan
| | - Min-Hsien Wu
- Division of Hematology-Oncology, Department of Internal Medicine, New Taipei Municipal TuCheng Hospital, New Taipei City, Taiwan; Division of Hematology-Oncology, Department of Internal Medicine, Chang Gung Memorial Hospital, Linkou, Taoyuan, Taiwan; Graduate Institute of Biomedical Engineering, Chang Gung University, Taoyuan, Taiwan
| | - Jason Chia-Hsun Hsieh
- College of Medicine, Chang Gung University, Taoyuan, Taiwan; Division of Hematology-Oncology, Department of Internal Medicine, New Taipei Municipal TuCheng Hospital, New Taipei City, Taiwan; Division of Hematology-Oncology, Department of Internal Medicine, Chang Gung Memorial Hospital, Linkou, Taoyuan, Taiwan.
| |
Collapse
|
28
|
Solaiyappan M, Bharti SK, Sharma RK, Dbouk M, Nizam W, Brock MV, Goggins MG, Bhujwalla ZM. Artificial neural network detection of pancreatic cancer from proton (1H) magnetic resonance spectroscopy patterns of plasma metabolites. COMMUNICATIONS MEDICINE 2025; 5:24. [PMID: 39838068 PMCID: PMC11751387 DOI: 10.1038/s43856-024-00727-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2024] [Accepted: 12/20/2024] [Indexed: 01/23/2025] Open
Abstract
BACKGROUND Routine screening to detect silent but deadly cancers such as pancreatic ductal adenocarcinoma (PDAC) can significantly improve survival, creating an important need for a convenient screening test. High-resolution proton (1H) magnetic resonance spectroscopy (MRS) of plasma identifies circulating metabolites that can allow detection of cancers such as PDAC that have highly dysregulated metabolism. METHODS We first acquired 1H MR spectra of human plasma samples classified as normal, benign pancreatic disease and malignant (PDAC). We next trained a system of artificial neural networks (ANNs) to process and discriminate these three classes using the full spectrum range and resolution of the acquired spectral data. We then identified and ranked spectral regions that played a salient role in the discrimination to provide interpretability of the results. We tested the accuracy of the ANN performance using blinded plasma samples. RESULTS We show that our ANN approach yields, in a cross validation-based training of 170 samples, a sensitivity and a specificity of 100% for malignant versus non-malignant (normal and disease combined) discrimination. The trained ANNs achieve a sensitivity and specificity of 87.5% and 93.1% respectively (AUC: ROC = 0.931, P-R = 0.854), with 45 blinded plasma samples. Further, we show that the salient spectral regions of the ANN discrimination correspond to metabolites of known importance for their role in cancers. CONCLUSIONS Our results demonstrate that the ANN approach presented here can identify PDAC from 1H MR plasma spectra to provide a convenient plasma-based assay for population-level screening of PDAC. The ANN approach can be suitably expanded to detect other cancers with metabolic dysregulation.
Collapse
Affiliation(s)
- Meiyappan Solaiyappan
- Department of Radiology, The Johns Hopkins University School of Medicine, Baltimore, MD, USA.
| | - Santosh Kumar Bharti
- Department of Radiology, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Raj Kumar Sharma
- Department of Radiology, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Mohamad Dbouk
- Department of Pathology, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Wasay Nizam
- Department of Surgery, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Malcolm V Brock
- Department of Surgery, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Oncology, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Michael G Goggins
- Department of Pathology, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Oncology, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Medicine, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Zaver M Bhujwalla
- Department of Radiology, The Johns Hopkins University School of Medicine, Baltimore, MD, USA.
- Department of Pathology, The Johns Hopkins University School of Medicine, Baltimore, MD, USA.
- Department of Radiation Oncology and Molecular Radiation Sciences, The Johns Hopkins University School of Medicine, Baltimore, MD, USA.
| |
Collapse
|
29
|
Borjikhani P, Granpayeh N, Zibaii MI. High sensitivity tapered fiber refractive index biosensor using hollow gold nanoparticles. Sci Rep 2025; 15:1458. [PMID: 39789159 PMCID: PMC11717910 DOI: 10.1038/s41598-025-85739-z] [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: 09/01/2024] [Accepted: 01/06/2025] [Indexed: 01/12/2025] Open
Abstract
A localized surface plasmon resonance (LSPR) sensor based on tapered optical fiber (TOF) using hollow gold nanoparticles (HAuNPs) for measuring the refractive index (RI) is presented. This optical fiber sensor is a good candidate for a label-free RI biosensor. In practical biosensors, bioreceptors are immobilized on nanoparticles (NPs) that only absorb specific biomolecules. The binding of these biomolecules to the receptors changes the local RI around the sensor and this change is detected by the transmittance spectrum of the fiber. Fast, accurate, easy and low-cost disease diagnosis are the advantages of optical fiber biosensors. In this paper, the structure theory is reviewed and the sensor is simulated by the finite difference time domain (FDTD) method and the finite element method (FEM) and the effect of the thickness and diameter of the HAuNPs and the waist diameter of the TOF is investigated. For the structure with HAuNPs thickness (2.5 nm), diameter (50 nm), and the fiber waist diameter of 10 μm, the wavelength sensitivity of 489.8 nm/RIU and full width at half maximum (FWHM) of 50 nm are obtained, which are better than those specifications in some other LSPR fiber sensors. In addition, the sensitivity of the sensor increases about 2-3 times compared to those of sensors with the same structure. Although there are many parameters in human blood that can change its RI, in practical work, the special bioreceptors on the sensor can deactivate other markers except the specific cancer markers, which changes the effective RI. Therefore, this optical fiber sensor is used for label-free detecting the RI of cancer cells and can be used as a biosensor for the detection of early stages of cancers in a non-invasive way, just using human blood samples.
Collapse
Affiliation(s)
- Parisa Borjikhani
- Center of Excellence in Electromagnetics, Optical Communication Laboratory, Faculty of Electrical Engineering, K.N. Toosi University of Technology, Tehran, Iran
| | - Nosrat Granpayeh
- Center of Excellence in Electromagnetics, Optical Communication Laboratory, Faculty of Electrical Engineering, K.N. Toosi University of Technology, Tehran, Iran
| | | |
Collapse
|
30
|
Würth R, Donato E, Michel LL, Saini M, Becker L, Cheytan T, Doncevic D, Messmer T, Gutjahr E, Weber R, Klein C, Alborzinia H, Yildiz U, Vogel V, Hlevnjak M, Kozyulina P, Neuberth SJ, Schwerd-Kleine P, Jakab S, Pfarr N, Angeles AK, Laut AK, Karpova D, Falcone M, Hardt O, Theek B, Wagner CV, Becker M, Wagner S, Haselmayr M, Schmitt A, Müller-Tidow C, Riethdorf S, Pantel K, Zapatka M, Sültmann H, Herrmann C, Thewes V, Lichter P, Schneeweiss A, Sprick MR, Trumpp A. Circulating tumor cell plasticity determines breast cancer therapy resistance via neuregulin 1-HER3 signaling. NATURE CANCER 2025; 6:67-85. [PMID: 39753722 PMCID: PMC11779641 DOI: 10.1038/s43018-024-00882-2] [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: 09/25/2023] [Accepted: 11/18/2024] [Indexed: 01/31/2025]
Abstract
Circulating tumor cells (CTCs) drive metastasis, the leading cause of death in individuals with breast cancer. Due to their low abundance in the circulation, robust CTC expansion protocols are urgently needed to effectively study disease progression and therapy responses. Here we present the establishment of long-term CTC-derived organoids from female individuals with metastatic breast cancer. Multiomics analysis of CTC-derived organoids along with preclinical modeling with xenografts identified neuregulin 1 (NRG1)-ERBB2 receptor tyrosine kinase 3 (ERBB3/HER3) signaling as a key pathway required for CTC survival, growth and dissemination. Genome-wide CRISPR activation screens revealed that fibroblast growth factor receptor 1 (FGFR1) signaling serves a compensatory function to the NRG1-HER3 axis and rescues NRG1 deficiency in CTCs. Conversely, NRG1-HER3 activation induced resistance to FGFR1 inhibition, whereas combinatorial blockade impaired CTC growth. The dynamic interplay between NRG1-HER3 and FGFR1 signaling reveals the molecular basis of cancer cell plasticity and clinically relevant strategies to target it. Our CTC organoid platform enables the identification and validation of patient-specific vulnerabilities and represents an innovative tool for precision medicine.
Collapse
MESH Headings
- Humans
- Neuregulin-1/metabolism
- Neuregulin-1/genetics
- Receptor, ErbB-3/metabolism
- Receptor, ErbB-3/genetics
- Female
- Breast Neoplasms/pathology
- Breast Neoplasms/drug therapy
- Breast Neoplasms/metabolism
- Breast Neoplasms/genetics
- Signal Transduction
- Animals
- Neoplastic Cells, Circulating/metabolism
- Neoplastic Cells, Circulating/pathology
- Neoplastic Cells, Circulating/drug effects
- Drug Resistance, Neoplasm
- Mice
- Receptor, Fibroblast Growth Factor, Type 1/metabolism
- Receptor, Fibroblast Growth Factor, Type 1/antagonists & inhibitors
- Receptor, Fibroblast Growth Factor, Type 1/genetics
- Organoids/metabolism
- Organoids/pathology
- Cell Line, Tumor
- Xenograft Model Antitumor Assays
Collapse
Affiliation(s)
- Roberto Würth
- Division of Stem Cells and Cancer, German Cancer Research Center (DKFZ) and DKFZ-ZMBH Alliance, Heidelberg, Germany.
- Heidelberg Institute for Stem Cell Technology and Experimental Medicine (HI-STEM gGmbH)), Heidelberg, Germany.
| | - Elisa Donato
- Division of Stem Cells and Cancer, German Cancer Research Center (DKFZ) and DKFZ-ZMBH Alliance, Heidelberg, Germany
- Heidelberg Institute for Stem Cell Technology and Experimental Medicine (HI-STEM gGmbH)), Heidelberg, Germany
| | - Laura L Michel
- Gynecologic Oncology, National Center for Tumor Diseases (NCT), University of Heidelberg and German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Massimo Saini
- Division of Stem Cells and Cancer, German Cancer Research Center (DKFZ) and DKFZ-ZMBH Alliance, Heidelberg, Germany
- Heidelberg Institute for Stem Cell Technology and Experimental Medicine (HI-STEM gGmbH)), Heidelberg, Germany
| | - Lisa Becker
- Division of Stem Cells and Cancer, German Cancer Research Center (DKFZ) and DKFZ-ZMBH Alliance, Heidelberg, Germany
- Heidelberg Institute for Stem Cell Technology and Experimental Medicine (HI-STEM gGmbH)), Heidelberg, Germany
| | - Tasneem Cheytan
- Division of Stem Cells and Cancer, German Cancer Research Center (DKFZ) and DKFZ-ZMBH Alliance, Heidelberg, Germany
- Heidelberg Institute for Stem Cell Technology and Experimental Medicine (HI-STEM gGmbH)), Heidelberg, Germany
| | - Daria Doncevic
- Department of Bioinformatics, Institute of Pharmacy and Molecular Biotechnology & BioQuant, Heidelberg University, Heidelberg, Germany
| | - Tobias Messmer
- Division of Stem Cells and Cancer, German Cancer Research Center (DKFZ) and DKFZ-ZMBH Alliance, Heidelberg, Germany
- Heidelberg Institute for Stem Cell Technology and Experimental Medicine (HI-STEM gGmbH)), Heidelberg, Germany
| | - Ewgenija Gutjahr
- Division of Stem Cells and Cancer, German Cancer Research Center (DKFZ) and DKFZ-ZMBH Alliance, Heidelberg, Germany
- Heidelberg Institute for Stem Cell Technology and Experimental Medicine (HI-STEM gGmbH)), Heidelberg, Germany
- Institute of Pathology, University Hospital Heidelberg, Heidelberg, Germany
| | - Rebecca Weber
- Division of Stem Cells and Cancer, German Cancer Research Center (DKFZ) and DKFZ-ZMBH Alliance, Heidelberg, Germany
| | - Corinna Klein
- Division of Stem Cells and Cancer, German Cancer Research Center (DKFZ) and DKFZ-ZMBH Alliance, Heidelberg, Germany
- Heidelberg Institute for Stem Cell Technology and Experimental Medicine (HI-STEM gGmbH)), Heidelberg, Germany
| | - Hamed Alborzinia
- Division of Stem Cells and Cancer, German Cancer Research Center (DKFZ) and DKFZ-ZMBH Alliance, Heidelberg, Germany
- Heidelberg Institute for Stem Cell Technology and Experimental Medicine (HI-STEM gGmbH)), Heidelberg, Germany
| | - Umut Yildiz
- Division of Stem Cells and Cancer, German Cancer Research Center (DKFZ) and DKFZ-ZMBH Alliance, Heidelberg, Germany
- Heidelberg Institute for Stem Cell Technology and Experimental Medicine (HI-STEM gGmbH)), Heidelberg, Germany
| | - Vanessa Vogel
- Division of Stem Cells and Cancer, German Cancer Research Center (DKFZ) and DKFZ-ZMBH Alliance, Heidelberg, Germany
- Heidelberg Institute for Stem Cell Technology and Experimental Medicine (HI-STEM gGmbH)), Heidelberg, Germany
| | - Mario Hlevnjak
- Computational Oncology, Molecular Precision Oncology Program, German Cancer Research Center (DKFZ) and National Center for Tumor Diseases (NCT), Heidelberg, Germany
| | - Polina Kozyulina
- Computational Oncology, Molecular Precision Oncology Program, German Cancer Research Center (DKFZ) and National Center for Tumor Diseases (NCT), Heidelberg, Germany
| | - Sarah-Jane Neuberth
- Division of Stem Cells and Cancer, German Cancer Research Center (DKFZ) and DKFZ-ZMBH Alliance, Heidelberg, Germany
- Heidelberg Institute for Stem Cell Technology and Experimental Medicine (HI-STEM gGmbH)), Heidelberg, Germany
| | - Paul Schwerd-Kleine
- Division of Stem Cells and Cancer, German Cancer Research Center (DKFZ) and DKFZ-ZMBH Alliance, Heidelberg, Germany
- Heidelberg Institute for Stem Cell Technology and Experimental Medicine (HI-STEM gGmbH)), Heidelberg, Germany
| | - Sevinç Jakab
- Division of Stem Cells and Cancer, German Cancer Research Center (DKFZ) and DKFZ-ZMBH Alliance, Heidelberg, Germany
- Heidelberg Institute for Stem Cell Technology and Experimental Medicine (HI-STEM gGmbH)), Heidelberg, Germany
| | - Nicole Pfarr
- Institute of Pathology, TUM School of Medicine and Health, Technical University of Munich, Munich, Germany
| | - Arlou Kristina Angeles
- Division of Cancer Genome Research, German Cancer Research Center, National Center for Tumor Diseases, Heidelberg, Germany
| | - Astrid K Laut
- Division of Cancer Genome Research, German Cancer Research Center, National Center for Tumor Diseases, Heidelberg, Germany
| | - Darja Karpova
- Division of Stem Cells and Cancer, German Cancer Research Center (DKFZ) and DKFZ-ZMBH Alliance, Heidelberg, Germany
- Heidelberg Institute for Stem Cell Technology and Experimental Medicine (HI-STEM gGmbH)), Heidelberg, Germany
| | - Mattia Falcone
- Division of Stem Cells and Cancer, German Cancer Research Center (DKFZ) and DKFZ-ZMBH Alliance, Heidelberg, Germany
- Heidelberg Institute for Stem Cell Technology and Experimental Medicine (HI-STEM gGmbH)), Heidelberg, Germany
| | - Olaf Hardt
- Miltenyi Biotec, Bergisch Gladbach, Germany
| | | | - Celina V Wagner
- Gynecologic Oncology, National Center for Tumor Diseases (NCT), University of Heidelberg and German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Mirjam Becker
- Gynecologic Oncology, National Center for Tumor Diseases (NCT), University of Heidelberg and German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Sabine Wagner
- Gynecologic Oncology, National Center for Tumor Diseases (NCT), University of Heidelberg and German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Martina Haselmayr
- Gynecologic Oncology, National Center for Tumor Diseases (NCT), University of Heidelberg and German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Anita Schmitt
- Department of Internal Medicine V, University Hospital Heidelberg, Heidelberg, Germany
| | - Carsten Müller-Tidow
- Department of Internal Medicine V, University Hospital Heidelberg, Heidelberg, Germany
| | - Sabine Riethdorf
- Department of Tumor Biology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Klaus Pantel
- Department of Tumor Biology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Marc Zapatka
- Division of Molecular Genetics, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Holger Sültmann
- Division of Cancer Genome Research, German Cancer Research Center, National Center for Tumor Diseases, Heidelberg, Germany
- German Cancer Consortium (DKTK), Heidelberg, Germany
| | - Carl Herrmann
- Department of Bioinformatics, Institute of Pharmacy and Molecular Biotechnology & BioQuant, Heidelberg University, Heidelberg, Germany
| | - Verena Thewes
- Gynecologic Oncology, National Center for Tumor Diseases (NCT), University of Heidelberg and German Cancer Research Center (DKFZ), Heidelberg, Germany
- Division of Molecular Genetics, German Cancer Research Center (DKFZ), Heidelberg, Germany
- National Center for Tumor Diseases (NCT) Heidelberg, a partnership between DKFZ and Heidelberg University, Heidelberg, Germany
| | - Peter Lichter
- Division of Molecular Genetics, German Cancer Research Center (DKFZ), Heidelberg, Germany
- German Cancer Consortium (DKTK), Heidelberg, Germany
- National Center for Tumor Diseases (NCT) Heidelberg, a partnership between DKFZ and Heidelberg University, Heidelberg, Germany
| | - Andreas Schneeweiss
- Gynecologic Oncology, National Center for Tumor Diseases (NCT), University of Heidelberg and German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Martin R Sprick
- Division of Stem Cells and Cancer, German Cancer Research Center (DKFZ) and DKFZ-ZMBH Alliance, Heidelberg, Germany
- Heidelberg Institute for Stem Cell Technology and Experimental Medicine (HI-STEM gGmbH)), Heidelberg, Germany
| | - Andreas Trumpp
- Division of Stem Cells and Cancer, German Cancer Research Center (DKFZ) and DKFZ-ZMBH Alliance, Heidelberg, Germany.
- Heidelberg Institute for Stem Cell Technology and Experimental Medicine (HI-STEM gGmbH)), Heidelberg, Germany.
- German Cancer Consortium (DKTK), Heidelberg, Germany.
| |
Collapse
|
31
|
Zhang Y, Wang B, Cai J, Yang Y, Tang C, Zheng X, Li H, Xu F. Enrichment and separation technology for evaluation of circulating tumor cells. Talanta 2025; 282:127025. [PMID: 39406084 DOI: 10.1016/j.talanta.2024.127025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2024] [Revised: 09/29/2024] [Accepted: 10/08/2024] [Indexed: 11/20/2024]
Abstract
Circulating tumor cells (CTCs) are tumor cells that exist in human peripheral blood, which could spread to other tissues or organs via the blood circulation system and develop into metastatic foci, leading to tumor recurrence or metastasis in postoperative patients and thereby increasing the mortality of malignant tumor patients. Evaluation of CTC levels can be used for tumor metastasis prediction, prognosis evaluation, drug exploitation, individualized treatment, liquid biopsy, etc., which exhibit outstanding clinical application prospects. In recent years, accurately capturing and analyzing CTCs has become a research hotspot in the early diagnosis and precise treatment of tumors. This review summarized various enrichment and isolation technologies for evaluating CTCs based on the design principle and discussed the challenges and perspectives in this field.
Collapse
Affiliation(s)
- Yanjun Zhang
- The Key Laboratory of Laboratory Medicine, Ministry of Education, School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, 325035, China
| | - Bing Wang
- The Key Laboratory of Laboratory Medicine, Ministry of Education, School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, 325035, China
| | - Junwen Cai
- The Key Laboratory of Laboratory Medicine, Ministry of Education, School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, 325035, China
| | - Yuting Yang
- Department of Clinical Laboratory, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, 325027, China
| | - Chen Tang
- The Key Laboratory of Laboratory Medicine, Ministry of Education, School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, 325035, China
| | - Xiaoqun Zheng
- The Key Laboratory of Laboratory Medicine, Ministry of Education, School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, 325035, China; Department of Clinical Laboratory, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, 325027, China
| | - Haidong Li
- State Key Laboratory of Fine Chemicals, Frontiers Science Center for Smart Materials Oriented Chemical Engineering, School of Bioengineering, Dalian University of Technology, Dalian, 116024, China; Provincial Key Laboratory of Interdisciplinary Medical Engineering for Gastrointestinal Carcinoma, Cancer Hospital of Dalian University of Technology (Liaoning Cancer Hospital & Institute), Shenyang, 110000, China
| | - Feng Xu
- The Key Laboratory of Laboratory Medicine, Ministry of Education, School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, 325035, China.
| |
Collapse
|
32
|
Rusnáková DŠ, Aziri R, Dubovan P, Jurík M, Mego M, Pinďák D. Detection, significance and potential utility of circulating tumor cells in clinical practice in breast cancer (Review). Oncol Lett 2025; 29:10. [PMID: 39492933 PMCID: PMC11526295 DOI: 10.3892/ol.2024.14756] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2024] [Accepted: 08/15/2024] [Indexed: 11/05/2024] Open
Abstract
Although advances in diagnostic techniques, new therapeutic strategies and personalization of breast cancer (BC) care have improved the survival for a number of patients, BC remains a major cause of morbidity and mortality for women. The study of circulating tumor cells (CTCs) has significant potential in translational oncology since these cells represent promising biomarkers throughout the entire course of BC in patients. CTCs also have notable prognostic value in early BC as well as metastatic BC. Based on current knowledge, it seems that the dynamics of CTCs that change during therapy reflect therapy response, and CTCs could serve as a tool for risk stratification and real-time monitoring of treatment in patients with BC. The question of how to use this information in everyday clinical practice and how this information can guide or change therapy to affect the clinical outcome of patients with BC remains unanswered. The present review aims to discuss current completed and ongoing trials that have been designed to demonstrate the clinical significance of CTCs, offer insights into treatment efficacy and assess CTC utility, facilitating their implementation in the routine management of patients with BC.
Collapse
Affiliation(s)
- Dominika Šmičková Rusnáková
- Department of Surgical Oncology, Faculty of Medicine, Slovak Medical University and National Cancer Institute, 833 10 Bratislava, Slovak Republic
| | - Ramadan Aziri
- Department of Surgical Oncology, Faculty of Medicine, Slovak Medical University and National Cancer Institute, 833 10 Bratislava, Slovak Republic
| | - Peter Dubovan
- Department of Surgical Oncology, Faculty of Medicine, Slovak Medical University and National Cancer Institute, 833 10 Bratislava, Slovak Republic
| | - Miroslav Jurík
- Department of Surgical Oncology, Faculty of Medicine, Slovak Medical University and National Cancer Institute, 833 10 Bratislava, Slovak Republic
| | - Michal Mego
- Second Department of Oncology, Faculty of Medicine, Comenius University and National Cancer Institute, 833 10 Bratislava, Slovak Republic
| | - Daniel Pinďák
- Department of Surgical Oncology, Faculty of Medicine, Slovak Medical University and National Cancer Institute, 833 10 Bratislava, Slovak Republic
| |
Collapse
|
33
|
Wu P, He X, Fan J, Tai Y, Zheng D, Yao Y, Sun S, Luo Y, Chen J, Hu WW, Ying B, Luo F, Niu Q, Sun X, Li Y. Electrochemical cytosensors for non-invasive liquid biopsy: Detection procedures and technologies for circulating tumor cells. Biosens Bioelectron 2025; 267:116818. [PMID: 39353368 DOI: 10.1016/j.bios.2024.116818] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2024] [Revised: 09/08/2024] [Accepted: 09/27/2024] [Indexed: 10/04/2024]
Abstract
Each year, millions of new cancer cases and cancer-related deaths underscore the urgent need for effective, affordable screening methods. Circulating tumor cells (CTCs), which derived from tumors and shedding into bloodstream, are considered promising biomarkers for liquid biopsy due to their unique biological significance and the substantial volume of supporting research. Among many advanced CTCs detection methods, electrochemical sensing is rapidly developing due to their high selectivity, high sensitivity, low cost, and rapid detection capability, well meeting the growing demand for non-invasive liquid biopsy. This review focuses on the entire procedure of detecting CTCs using electrochemical cytosensors, starting from sample preparation, detailing bio-recognition elements for capturing CTCs, highlighting design strategies of cytosensor, and discussing the prospects and challenges of electrochemical cytosensor applications.
Collapse
Affiliation(s)
- Peilin Wu
- Department of Laboratory Medicine/Clinical Laboratory Medicine Research Center, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China; Sichuan Clinical Research Center for Laboratory Medicine, Chengdu, 610041, Sichuan, China
| | - Xun He
- Center for High Altitude Medicine, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China; Institute of Fundamental and Frontier Sciences, University of Electronic Science and Technology of China, Chengdu, 610054, Sichuan, China
| | - Jiwen Fan
- Department of Laboratory Medicine/Clinical Laboratory Medicine Research Center, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China; Sichuan Clinical Research Center for Laboratory Medicine, Chengdu, 610041, Sichuan, China
| | - Yunze Tai
- Department of Laboratory Medicine/Clinical Laboratory Medicine Research Center, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China; Sichuan Clinical Research Center for Laboratory Medicine, Chengdu, 610041, Sichuan, China
| | - Dongdong Zheng
- College of Chemistry, Chemical Engineering and Materials Science, Shandong Normal University, Jinan, 250014, Shandong, China
| | - Yongchao Yao
- Department of Laboratory Medicine/Clinical Laboratory Medicine Research Center, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China; Sichuan Clinical Research Center for Laboratory Medicine, Chengdu, 610041, Sichuan, China
| | - Shengjun Sun
- College of Chemistry, Chemical Engineering and Materials Science, Shandong Normal University, Jinan, 250014, Shandong, China
| | - Yao Luo
- Department of Laboratory Medicine/Clinical Laboratory Medicine Research Center, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China; Sichuan Clinical Research Center for Laboratory Medicine, Chengdu, 610041, Sichuan, China
| | - Jie Chen
- Department of Laboratory Medicine/Clinical Laboratory Medicine Research Center, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China; Sichuan Clinical Research Center for Laboratory Medicine, Chengdu, 610041, Sichuan, China
| | - Wenchuang Walter Hu
- Department of Laboratory Medicine/Clinical Laboratory Medicine Research Center, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China; Sichuan Clinical Research Center for Laboratory Medicine, Chengdu, 610041, Sichuan, China
| | - Binwu Ying
- Department of Laboratory Medicine/Clinical Laboratory Medicine Research Center, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China; Sichuan Clinical Research Center for Laboratory Medicine, Chengdu, 610041, Sichuan, China
| | - Fengming Luo
- Center for High Altitude Medicine, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China; Department of Pulmonary and Critical Care Medicine, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China
| | - Qian Niu
- Department of Laboratory Medicine/Clinical Laboratory Medicine Research Center, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China; Sichuan Clinical Research Center for Laboratory Medicine, Chengdu, 610041, Sichuan, China.
| | - Xuping Sun
- Center for High Altitude Medicine, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China; Institute of Fundamental and Frontier Sciences, University of Electronic Science and Technology of China, Chengdu, 610054, Sichuan, China; College of Chemistry, Chemical Engineering and Materials Science, Shandong Normal University, Jinan, 250014, Shandong, China.
| | - Yi Li
- Department of Laboratory Medicine/Clinical Laboratory Medicine Research Center, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China; Sichuan Clinical Research Center for Laboratory Medicine, Chengdu, 610041, Sichuan, China.
| |
Collapse
|
34
|
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
|
35
|
Francis JH, Levine S, Canestraro J, Eng J, Abramson DH. PLASMA CIRCULATING TUMOR DNA TO ASSIST IN THE DIAGNOSIS OF UNKNOWN PRIMARY MALIGNANCY UNDERLYING CHOROIDAL METASTASES. Retin Cases Brief Rep 2025; 19:46-49. [PMID: 38113252 DOI: 10.1097/icb.0000000000001518] [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: 12/21/2023]
Abstract
PURPOSE Management of suspected choroidal metastases requires diagnostic imaging and an invasive, sometimes intraocular, biopsy to determine the primary malignancy. This multistep process takes time, which may affect morbidity and mortality. METHODS This was a retrospective review of one case. RESULTS A 56-year-old woman presented with bilateral amelanotic choroidal masses suspicious for metastases of unknown origin. Plasma circulating tumor DNA revealed EGFR , PTEN , and SMAD4 , a profile consistent with non-small-cell lung cancer. Subsequent radiographic imaging and scapular biopsy revealed lung adenocarcinoma and genetic profile concordant with the liquid biopsy. The patient was started on EGFR inhibitor, osimertinib, with measurable systemic and ocular response. CONCLUSION Plasma circulating tumor DNA revealed the genetic profile of the primary malignancy underlying choroidal metastases of unknown origin, aiding in the prompt diagnosis and detecting the driver mutation that guided management with targeted therapy.
Collapse
Affiliation(s)
- Jasmine H Francis
- Ophthalmic Oncology Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, New York
- Weill-Cornell Medical Center, New York, New York; and
| | - Sara Levine
- Ophthalmic Oncology Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Julia Canestraro
- Ophthalmic Oncology Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, New York
- Weill-Cornell Medical Center, New York, New York; and
| | - Juliana Eng
- Weill-Cornell Medical Center, New York, New York; and
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York
| | - David H Abramson
- Ophthalmic Oncology Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, New York
- Weill-Cornell Medical Center, New York, New York; and
| |
Collapse
|
36
|
Hussung S, Hess ME, Haghighi EB, Wittel UA, Boerries M, Fritsch RM. Integrated Analysis of Cell-Free DNA and Novel Protein Biomarkers for Stratification and Therapy Monitoring in Stage IV Pancreatic Cancer: A Preliminary Study. Diagnostics (Basel) 2024; 15:49. [PMID: 39795577 PMCID: PMC11720586 DOI: 10.3390/diagnostics15010049] [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: 12/03/2024] [Revised: 12/25/2024] [Accepted: 12/26/2024] [Indexed: 01/13/2025] Open
Abstract
Background: Given the poor prognosis of metastatic pancreatic adenocarcinoma (mPDAC), closer disease monitoring through liquid biopsy, most frequently based on serial measurements of cell-free mutated KRAS (KRASmut cfDNA), has become a highly active research focus, aimed at improving patients' long-term outcomes. However, most of the available data show only a limited predictive and prognostic value of single-parameter-based methods. We hypothesized that a combined longitudinal analysis of KRASmut cfDNA and novel protein biomarkers could improve risk stratification and molecular monitoring of patients with mPDAC. Methods: We prospectively collected 160 plasma samples from 47 patients with mPDAC at our institution. Highly sensitive single-target ddPCR assays were employed to detect and quantify KRASmut cfDNA. Additionally, analysis of ten protein biomarkers was performed through Enzyme-linked Immunosorbent Assay (ELISA), and Carbohydrate-Antigen 19-9 (CA 19-9) dynamics were registered. Results: KRASmut cfDNA was detectable in 37/47 (78.7%) patients throughout the course of study, and CA 19-9 levels were elevated in 40 out of 47 (85.1%) patients. KRASmut cfDNA increase at the time of the first follow-up could predict inferior progression-free survival (PFS) (Hazard ratio (HR) = 3.40, p = 0.0003) and overall survival (OS) (HR = 4.91, p < 0.0001). In contrast to CA 19-9 kinetics, which were not predictive of outcome, integrated analysis of KRASmut cfDNA combined with six evaluated circulating protein biomarkers allowed basal risk stratification at the time of the first follow-up (HR = 10.2, p = 0.0014). Conclusions: A combined longitudinal analysis of KRASmut cfDNA with selected protein biomarkers offers significantly improved prognostic value for patients with mPDAC compared to single-parameter methods. This innovative approach is a step forward in the molecular monitoring of mPDAC and should be validated in further prospective studies.
Collapse
Affiliation(s)
- Saskia Hussung
- Department of Medicine I (Hematology, Oncology and Stem Cell Transplantation), Freiburg University Medical Center, 79106 Freiburg, Germany;
- Department of Medical Oncology and Hematology, Zurich University Hospital, 8091 Zurich, Switzerland
| | - Maria E. Hess
- Institute of Medical Bioinformatics and Systems Medicine, Medical Center—University of Freiburg, Faculty of Medicine, University of Freiburg, 79104 Freiburg, Germany; (M.E.H.); (E.B.H.); (M.B.)
- Faculty of Biology, University of Freiburg, 79104 Freiburg, Germany
| | - Elham Bavafaye Haghighi
- Institute of Medical Bioinformatics and Systems Medicine, Medical Center—University of Freiburg, Faculty of Medicine, University of Freiburg, 79104 Freiburg, Germany; (M.E.H.); (E.B.H.); (M.B.)
| | - Uwe A. Wittel
- Department of Surgery, Freiburg University Medical Center, 79106 Freiburg, Germany;
| | - Melanie Boerries
- Institute of Medical Bioinformatics and Systems Medicine, Medical Center—University of Freiburg, Faculty of Medicine, University of Freiburg, 79104 Freiburg, Germany; (M.E.H.); (E.B.H.); (M.B.)
- German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
- German Cancer Consortium (DKTK), Partner Site Freiburg, German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
- Comprehensive Cancer Center Freiburg (CCCF), Medical Center—University of Freiburg, Faculty of Medicine, University of Freiburg, 79104 Freiburg, Germany
| | - Ralph M. Fritsch
- Department of Medicine I (Hematology, Oncology and Stem Cell Transplantation), Freiburg University Medical Center, 79106 Freiburg, Germany;
- Department of Medical Oncology and Hematology, Zurich University Hospital, 8091 Zurich, Switzerland
- Department of Surgery, Freiburg University Medical Center, 79106 Freiburg, Germany;
- German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
- German Cancer Consortium (DKTK), Partner Site Freiburg, German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
| |
Collapse
|
37
|
Arai M, Hamad A, Almasry Y, Alamer A, Rasheed W, Aljurf M, El Fakih R. Minimal residual disease testing for classical Hodgkin lymphoma: A comprehensive review. Crit Rev Oncol Hematol 2024; 204:104503. [PMID: 39245298 DOI: 10.1016/j.critrevonc.2024.104503] [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: 05/20/2024] [Revised: 09/01/2024] [Accepted: 09/02/2024] [Indexed: 09/10/2024] Open
Abstract
Classical Hodgkin lymphoma (cHL) is a common lymphoma that affects young patients. Fortunately, the disease is highly curable as it is susceptible to the currently available treatment modalities. Disease monitoring with Positron Emission Tomography and Computed Tomography (PET/ CT) is an integral part of managing these patients. PET guided protocols are currently used to adjust treatment according to the response. The pivotal idea behind the use of response-adapted approaches is to preserve efficacy while decreasing the toxicity. It also helps to intensify therapy in patients in need because of suboptimal response. However, imaging techniques are limited by their sensitivity and specificity. Minimal Residual Disease (MRD) assessment is a newly emerging concept in many hematologic malignancies. It utilizes various molecular techniques such as polymerase chain reaction (PCR), and next-generation sequencing (NGS) as well as flow cytometry, to detect disease traces. This review looks into MRD detection techniques, its current applications, and the evidence in the literature for its use in cHL.
Collapse
Affiliation(s)
- Momo Arai
- College of Medicine, Alfaisal University, Riyadh 11533, Saudi Arabia
| | - Alaa Hamad
- College of Medicine, Alfaisal University, Riyadh 11533, Saudi Arabia.
| | - Yazan Almasry
- College of Medicine, Alfaisal University, Riyadh 11533, Saudi Arabia
| | - Abdullah Alamer
- Department of Hematology, King Faisal Specialist Hospital & Research Center, Riyadh 11564, Saudi Arabia
| | - Walid Rasheed
- Department of Hematology, King Faisal Specialist Hospital & Research Center, Riyadh 11564, Saudi Arabia
| | - Mahmoud Aljurf
- Department of Hematology, King Faisal Specialist Hospital & Research Center, Riyadh 11564, Saudi Arabia
| | - Riad El Fakih
- College of Medicine, Alfaisal University, Riyadh 11533, Saudi Arabia; Department of Hematology, King Faisal Specialist Hospital & Research Center, Riyadh 11564, Saudi Arabia
| |
Collapse
|
38
|
Wang Y, Duan Y, Guo D, Lv H, Li Q, Liu X, Qiao N, Meng H, Zhang X, Lan L, Liu X, Liu X. Value of circulating tumor cell assisting low-dose computed tomography in screening pulmonary nodules based on existing liquid biopsy techniques: a systematic review with meta-analysis and trial sequential analysis. Clin Transl Oncol 2024; 26:3252-3263. [PMID: 38869739 DOI: 10.1007/s12094-024-03556-8] [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: 04/11/2024] [Accepted: 06/04/2024] [Indexed: 06/14/2024]
Abstract
OBJECTIVE This study aims to assess the diagnostic utility of circulating tumor cells (CTCs) in conjunction with low-dose computed tomography (LDCT) for differentiating between benign and malignant pulmonary nodules and to substantiate the foundation for their integration into clinical practice. METHODS A systematic literature review was performed independently by two researchers utilizing databases including PubMed, Web of Science, The Cochrane Library, Embase, and Medline, to collate studies up to September 15, 2023, that investigated the application of CTCs in diagnosing pulmonary nodules. A meta-analysis was executed employing Stata 15.0 and Revman 5.4 to calculate the pooled sensitivity, specificity, positive and negative likelihood ratios (PLR and NLR), diagnostic odds ratio (DOR), and the area under the receiver operating characteristic curve (AUC). Additionally, trial sequential analysis was conducted using dedicated TSA software. RESULTS The selection criteria identified 16 studies, encompassing a total of 3409 patients. The meta-analysis revealed that CTCs achieved a pooled sensitivity of 0.84 (95% CI 0.80 to 0.87), specificity of 0.80 (95% CI 0.73 to 0.86), PLR of 4.23 (95% CI 3.12 to 5.72), NLR of 0.20 (95% CI 0.16 to 0.25), DOR of 20.92 (95% CI 13.52 to 32.36), and AUC of 0.89 (95% CI 0.86 to 0.93). CONCLUSIONS Circulating tumor cells demonstrate substantial diagnostic accuracy in distinguishing benign from malignant pulmonary nodules. The incorporation of CTCs into the diagnostic protocol can significantly augment the diagnostic efficacy of LDCT in screening for malignant lung diseases.
Collapse
Affiliation(s)
- Yixian Wang
- Department of Epidemiology and Statistics, School of Public Health, Jilin University, Changchun, Jilin, 130021, China
| | - Yuqing Duan
- Department of Epidemiology and Statistics, School of Public Health, Jilin University, Changchun, Jilin, 130021, China
| | - Dingjie Guo
- Department of Epidemiology and Statistics, School of Public Health, Jilin University, Changchun, Jilin, 130021, China
| | - Hongbo Lv
- Department of Epidemiology and Statistics, School of Public Health, Jilin University, Changchun, Jilin, 130021, China
| | - Qiong Li
- Department of Epidemiology and Statistics, School of Public Health, Jilin University, Changchun, Jilin, 130021, China
| | - Xuan Liu
- Department of Epidemiology and Statistics, School of Public Health, Jilin University, Changchun, Jilin, 130021, China
| | - Na Qiao
- Department of Epidemiology and Statistics, School of Public Health, Jilin University, Changchun, Jilin, 130021, China
| | - Hengyu Meng
- Department of Epidemiology and Statistics, School of Public Health, Jilin University, Changchun, Jilin, 130021, China
| | - Xin Zhang
- Department of Epidemiology and Statistics, School of Public Health, Jilin University, Changchun, Jilin, 130021, China
| | - Linwei Lan
- Department of Epidemiology and Statistics, School of Public Health, Jilin University, Changchun, Jilin, 130021, China
| | - Xiumin Liu
- Department of Clinical Laboratory, The Second Hospital of Jilin University, Changchun, Jilin, 130041, China.
| | - Xin Liu
- Department of Epidemiology and Statistics, School of Public Health, Jilin University, Changchun, Jilin, 130021, China.
| |
Collapse
|
39
|
Izhar M, Ahmad Z, Moazzam M, Jader A. Targeted liquid biopsy for brain tumors. THE JOURNAL OF LIQUID BIOPSY 2024; 6:100170. [PMID: 40027302 PMCID: PMC11863980 DOI: 10.1016/j.jlb.2024.100170] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/24/2024] [Revised: 10/08/2024] [Accepted: 10/09/2024] [Indexed: 03/05/2025]
Abstract
Cerebrospinal fluid (CSF) is a promising source of molecular biomarkers in the detection and monitoring of brain malignancies. Unlike peripheral blood, where biomarker detection is hindered by the blood-brain barrier and the complex nature of biofluids, CSF offers a more direct and enriched source of tumor-derived markers, including circulating tumor DNA (ctDNA), microRNA (miRNA), proteins, and extracellular vesicles (EVs). These biomarkers, originating from brain tumor cells, are often more concentrated in CSF than in peripheral blood due to the proximity of CSF to the central nervous system (CNS). The presence of ctDNA in CSF is notably higher than in plasma, making CSF an advantageous medium for liquid biopsy in brain tumor patients. Traditional liquid biopsy approaches relying on peripheral venous blood samples often face challenges in detecting low concentrations of tumor-derived biomarkers. The direct contact of CSF with the CNS minimizes background noise, potentially enhancing the accuracy and sensitivity of diagnostic assays. Despite the barriers posed by the blood-brain barrier and other physiological factors that limit biomarker levels in the systemic circulation, CSF's unique position within the CNS allows for more effective biomarker collection. While peripheral blood remains the standard medium for liquid biopsy in oncology, the proximity of CSF to brain tumors suggests it may offer superior diagnostic capabilities. Emerging evidence from non-CNS malignancies indicates that collecting biofluids closer to the tumor site can significantly improve biomarker detection. Although this approach has been well-documented in other solid tumors, its application to CNS malignancies remains underexplored. This study hypothesizes that targeted blood sampling from CNS tumor-draining veins could similarly enhance biomarker detection, thereby increasing the sensitivity and efficacy of liquid biopsy techniques in diagnosing and monitoring brain malignancies.
Collapse
Affiliation(s)
- Muhammad Izhar
- Department of Neurosurgery, Stanford University School of Medicine, Stanford, CA, 94304, USA
| | - Ziyad Ahmad
- Department of Orthopedics, Medical Teaching Institute, Mardan Medical Complex, Mardan, Pakistan
| | | | - Arwa Jader
- Department of Neurosurgery, Kufa University, Kufa, Iraq
| |
Collapse
|
40
|
Leyderman M, Chandrasekar T, Grivas P, Li R, Bhat S, Basnet A, Shapiro O, Jacob J, Daneshvar MA, Kord E, Bratslavsky G, Goldberg H. Metastasis development in non-muscle-invasive bladder cancer. Nat Rev Urol 2024:10.1038/s41585-024-00963-y. [PMID: 39567681 DOI: 10.1038/s41585-024-00963-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/18/2024] [Indexed: 11/22/2024]
Abstract
Non-muscle-invasive bladder cancer (NMIBC) is the most common type of bladder cancer presentation and is characterized by a varying probability of recurrence and progression. Sporadically, patients with NMIBC might also develop tumour metastases without any pathological evidence of muscle-invasive disease within the bladder, a condition known as metastatic NMIBC. In the published literature, this phenomenon is limited to several case reports and small reviews, with few data regarding the possible aetiologies. Several possible factors can be potentially associated with metastatic NMIBC, including tumour understaging, the number of transurethral resection procedures received by the patient, the presence of circulating tumour cells, the modality used for diagnostic cystoscopy and possible gender-associated differences. In this Perspective, our aim was to integrate and report currently available data on this relatively rare entity and provide some potential aetiological explanations.
Collapse
Affiliation(s)
- Michael Leyderman
- Department of Urology, SUNY Upstate Medical University, Syracuse, NY, USA
| | | | - Petros Grivas
- Department of Medicine, Division of Medical Oncology, University of Washington School of Medicine, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Roger Li
- Department of Genitourinary Oncology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, USA
| | - Seetharam Bhat
- Department of Urology, SUNY Upstate Medical University, Syracuse, NY, USA
| | - Alina Basnet
- Department of Medical Oncology, SUNY Upstate Medical University, Syracuse, NY, USA
| | - Oleg Shapiro
- Department of Urology, SUNY Upstate Medical University, Syracuse, NY, USA
| | - Joseph Jacob
- Department of Urology, SUNY Upstate Medical University, Syracuse, NY, USA
| | | | - Eyal Kord
- Department of Urology, SUNY Upstate Medical University, Syracuse, NY, USA
| | | | - Hanan Goldberg
- Department of Urology, SUNY Upstate Medical University, Syracuse, NY, USA.
- Upstate Urology at Mohawk Valley Health System (MVHS), Utica, NY, USA.
| |
Collapse
|
41
|
Zhang Y, Tian L. Advances and challenges in the use of liquid biopsy in gynaecological oncology. Heliyon 2024; 10:e39148. [PMID: 39492906 PMCID: PMC11530831 DOI: 10.1016/j.heliyon.2024.e39148] [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: 05/21/2024] [Revised: 10/08/2024] [Accepted: 10/08/2024] [Indexed: 11/05/2024] Open
Abstract
Ovarian cancer, endometrial cancer, and cervical cancer are the three primary gynaecological cancers that pose a significant threat to women's health on a global scale. Enhancing global cancer survival rates necessitates advancements in illness detection and monitoring, with the goal of improving early diagnosis and prognostication of disease recurrence. Conventional methods for identifying and tracking malignancies rely primarily on imaging techniques and, when possible, protein biomarkers found in blood, many of which lack specificity. The process of collecting tumour samples necessitates intrusive treatments that are not suitable for specific purposes, such as screening, predicting, or evaluating the effectiveness of treatment, monitoring the presence of remaining illness, and promptly detecting relapse. Advancements in treatment are being made by the detection of genetic abnormalities in tumours, both inherited and acquired. Newly designed therapeutic approaches can specifically address some of these abnormalities. Liquid biopsy is an innovative technique for collecting samples that examine specific cancer components that are discharged into the bloodstream, such as circulating tumour DNA (ctDNA), circulating tumour cells (CTCs), cell-free RNA (cfRNA), tumour-educated platelets (TEPs), and exosomes. Mounting data indicates that liquid biopsy has the potential to improve the clinical management of gynaecological cancers through enhanced early diagnosis, prognosis prediction, recurrence detection, and therapy response monitoring. Understanding the distinct genetic composition of tumours can also inform therapy choices and the identification of suitable targeted treatments. The main benefits of liquid biopsy are its non-invasive characteristics and practicality, enabling the collection of several samples and the continuous monitoring of tumour changes over time. This review aims to provide an overview of the data supporting the therapeutic usefulness of each component of liquid biopsy. Additionally, it will assess the benefits and existing constraints associated with the use of liquid biopsy in the management of gynaecological malignancies. In addition, we emphasise future prospects in light of the existing difficulties and investigate areas where further research is necessary to clarify its rising clinical capabilities.
Collapse
Affiliation(s)
- Yingfeng Zhang
- University-Town Hospital of Chongqing Medical University, Chongqing, 401331, China
| | - Libi Tian
- University-Town Hospital of Chongqing Medical University, Chongqing, 401331, China
| |
Collapse
|
42
|
Dompé C, Chojnowska A, Ramlau R, Nowicki M, Alix-Panabières C, Budna-Tukan J. Unveiling the dynamics of circulating tumor cells in colorectal cancer: from biology to clinical applications. Front Cell Dev Biol 2024; 12:1498032. [PMID: 39539964 PMCID: PMC11557528 DOI: 10.3389/fcell.2024.1498032] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2024] [Accepted: 10/18/2024] [Indexed: 11/16/2024] Open
Abstract
This review delves into the pivotal role of circulating tumor cells (CTCs) in colorectal cancer (CRC) metastasis, focusing on their biological properties, interactions with the immune system, advanced detection techniques, and clinical implications. We explored how metastasis-competent CTCs evade immune surveillance and proliferate, utilizing cutting-edge detection and isolation technologies, such as microfluidic devices and immunological assays, to enhance sensitivity and specificity. The review highlights the significant impact of CTC interactions with immune cells on tumor progression and patient outcomes. It discusses the application of these findings in clinical settings, including non-invasive liquid biopsies for early diagnosis, prognosis, and treatment monitoring. Despite advancements, challenges remain, such as the need for standardized methods to consistently capture and analyze CTCs. Addressing these challenges through further molecular and cellular research on CTCs could lead to improved interventions and outcomes for CRC patients, underscoring the importance of unraveling the complex dynamics of CTCs in cancer progression.
Collapse
Affiliation(s)
- Claudia Dompé
- Department of Immunology, Poznan University of Medical Sciences, Poznan, Poland
- Doctoral School, Poznan University of Medical Sciences, Poznan, Poland
| | | | - Rodryg Ramlau
- Department of Oncology, Poznan University of Medical Sciences, Poznan, Poland
| | - Michal Nowicki
- Department of Histology and Embryology, Poznan University of Medical Sciences, Poznan, Poland
| | - Catherine Alix-Panabières
- Laboratory of Rare Human Circulating Cells and Liquid Biopsy (LCCRH), University Medical Centre of Montpellier, Montpellier, France
- Centre de Recherche en Ecologie et Evolution du Cancer, Maladies Infectieuses et Vecteurs: Ecologie, Génétique, Evolution et Contrôle, University of Montpellier, Centre National de la Recherche Scientifique, Institut de Recherche Pour le Dévelopement, Montpellier, France
- European Liquid Biopsy Society (ELBS), Hamburg, Germany
| | - Joanna Budna-Tukan
- Department of Histology and Embryology, Poznan University of Medical Sciences, Poznan, Poland
- Department of Anatomy and Histology, Collegium Medicum, University of Zielona Gora, Zielona Gora, Poland
| |
Collapse
|
43
|
Wang B, Wang M, Lin Y, Zhao J, Gu H, Li X. Circulating tumor DNA methylation: a promising clinical tool for cancer diagnosis and management. Clin Chem Lab Med 2024; 62:2111-2127. [PMID: 38443752 DOI: 10.1515/cclm-2023-1327] [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: 11/23/2023] [Accepted: 02/19/2024] [Indexed: 03/07/2024]
Abstract
Cancer continues to pose significant challenges to the medical community. Early detection, accurate molecular profiling, and adequate assessment of treatment response are critical factors in improving the quality of life and survival of cancer patients. Accumulating evidence shows that circulating tumor DNA (ctDNA) shed by tumors into the peripheral blood preserves the genetic and epigenetic information of primary tumors. Notably, DNA methylation, an essential and stable epigenetic modification, exhibits both cancer- and tissue-specific patterns. As a result, ctDNA methylation has emerged as a promising molecular marker for noninvasive testing in cancer clinics. In this review, we summarize the existing techniques for ctDNA methylation detection, describe the current research status of ctDNA methylation, and present the potential applications of ctDNA-based assays in the clinic. The insights presented in this article could serve as a roadmap for future research and clinical applications of ctDNA methylation.
Collapse
Affiliation(s)
- Binliang Wang
- Department of Respiratory Medicine, Huangyan Hospital Affiliated to Wenzhou Medical University, Taizhou, P.R. China
| | - Meng Wang
- Institute of Health Education, Hangzhou Center for Disease Control and Prevention, Hangzhou, P.R. China
| | - Ya Lin
- Zhejiang University of Chinese Medicine, Hangzhou, P.R. China
| | - Jinlan Zhao
- Scientific Research Department, Zhejiang Shengting Medical Company, Hangzhou, P.R. China
| | - Hongcang Gu
- Institute of Health and Medical Technology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei, P.R. China
- Anhui Province Key Laboratory of Medical Physics and Technology, Institute of Health and Medical Technology, Hefei Institutes of Physical Science, Chinese Academy of Science, Hefei, P.R. China
| | - Xiangjuan Li
- Department of Gynaecology, Hangzhou Obstetrics and Gynecology Hospital, Hangzhou, P.R. China
| |
Collapse
|
44
|
Duo Y, Han L, Yang Y, Wang Z, Wang L, Chen J, Xiang Z, Yoon J, Luo G, Tang BZ. Aggregation-Induced Emission Luminogen: Role in Biopsy for Precision Medicine. Chem Rev 2024; 124:11242-11347. [PMID: 39380213 PMCID: PMC11503637 DOI: 10.1021/acs.chemrev.4c00244] [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: 04/03/2024] [Revised: 09/11/2024] [Accepted: 09/17/2024] [Indexed: 10/10/2024]
Abstract
Biopsy, including tissue and liquid biopsy, offers comprehensive and real-time physiological and pathological information for disease detection, diagnosis, and monitoring. Fluorescent probes are frequently selected to obtain adequate information on pathological processes in a rapid and minimally invasive manner based on their advantages for biopsy. However, conventional fluorescent probes have been found to show aggregation-caused quenching (ACQ) properties, impeding greater progresses in this area. Since the discovery of aggregation-induced emission luminogen (AIEgen) have promoted rapid advancements in molecular bionanomaterials owing to their unique properties, including high quantum yield (QY) and signal-to-noise ratio (SNR), etc. This review seeks to present the latest advances in AIEgen-based biofluorescent probes for biopsy in real or artificial samples, and also the key properties of these AIE probes. This review is divided into: (i) tissue biopsy based on smart AIEgens, (ii) blood sample biopsy based on smart AIEgens, (iii) urine sample biopsy based on smart AIEgens, (iv) saliva sample biopsy based on smart AIEgens, (v) biopsy of other liquid samples based on smart AIEgens, and (vi) perspectives and conclusion. This review could provide additional guidance to motivate interest and bolster more innovative ideas for further exploring the applications of various smart AIEgens in precision medicine.
Collapse
Affiliation(s)
- Yanhong Duo
- Department
of Radiation Oncology, Shenzhen People’s Hospital, The Second
Clinical Medical College, Jinan University, The First Affiliated Hospital, Southern University of Science and Technology, Shenzhen 518020, Guangdong China
- Wyss
Institute for Biologically Inspired Engineering, Harvard University, Boston, Massachusetts 02138, United States
| | - Lei Han
- College of
Chemistry and Pharmaceutical Sciences, Qingdao
Agricultural University, 700 Changcheng Road, Qingdao 266109, Shandong China
| | - Yaoqiang Yang
- Department
of Radiation Oncology, Shenzhen People’s Hospital, The Second
Clinical Medical College, Jinan University, The First Affiliated Hospital, Southern University of Science and Technology, Shenzhen 518020, Guangdong China
| | - Zhifeng Wang
- Department
of Urology, Henan Provincial People’s Hospital, Zhengzhou University
People’s Hospital, Henan University
People’s Hospital, Zhengzhou, 450003, China
| | - Lirong Wang
- State
Key Laboratory of Luminescent Materials and Devices, South China University of Technology, Guangzhou 510640, China
| | - Jingyi Chen
- Wyss
Institute for Biologically Inspired Engineering, Harvard University, Boston, Massachusetts 02138, United States
| | - Zhongyuan Xiang
- Department
of Laboratory Medicine, The Second Xiangya Hospital, Central South University, Changsha 410000, Hunan, China
| | - Juyoung Yoon
- Department
of Chemistry and Nanoscience, Ewha Womans
University, 52 Ewhayeodae-gil, Seodaemun-gu, Seoul 03760, Korea
| | - Guanghong Luo
- Department
of Radiation Oncology, Shenzhen People’s Hospital, The Second
Clinical Medical College, Jinan University, The First Affiliated Hospital, Southern University of Science and Technology, Shenzhen 518020, Guangdong China
| | - Ben Zhong Tang
- School
of Science and Engineering, Shenzhen Institute of Aggregate Science
and Technology, The Chinese University of
Hong Kong, Shenzhen 518172, Guangdong China
| |
Collapse
|
45
|
Cossu G, Ramsay DSC, Daniel RT, El Cadhi A, Kerherve L, Morlaix E, Houidi SA, Millot-Piccoli C, Chapon R, Le Van T, Cao C, Farah W, Lleu M, Baland O, Beaurain J, Petit JM, Lemogne B, Messerer M, Berhouma M. Update on Neoadjuvant and Adjuvant BRAF Inhibitors in Papillary Craniopharyngioma: A Systematic Review. Cancers (Basel) 2024; 16:3479. [PMID: 39456573 PMCID: PMC11506763 DOI: 10.3390/cancers16203479] [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: 08/31/2024] [Revised: 09/26/2024] [Accepted: 10/10/2024] [Indexed: 10/28/2024] Open
Abstract
Background/Objectives: The recent discovery of BRAF mutation in papillary craniopharyngiomas opened new avenues for targeted therapies to control tumour growth, decreasing the need for invasive treatments and relative complications. The aim of this systematic review was to summarize the recent scientific data dealing with the use of targeted therapies in papillary craniopharyngiomas, as adjuvant and neoadjuvant treatments. Methods: The PRISMA guidelines were followed with searches performed in Scopus, MEDLINE, and Embase, following a dedicated PICO approach. Results: We included 21 pertinent studies encompassing 53 patients: 26 patients received BRAF inhibitors (BRAFi) as adjuvant treatment, while 25 received them as neoadjuvant treatment. In the adjuvant setting, BRAFi were used to treat recurrent tumours after surgery or adjuvant radiation therapy. The most common regimen combined dabrafenib (BRAFi) with trametinib (MEK1 and 2 inhibitor) in 81% of cases. The mean treatment length was 8.8 months (range 1.6 to 28 months) and 32% were continuing BRAFi. A reduction of tumour volume variable from 24% to 100% was observed at cerebral MRI during treatment and volumetric reduction ≥80% was described in 64% of cases. Once the treatment was stopped, adjuvant treatments were performed to stabilize patients in remission in 11 cases (65%) or when a progression was detected in three cases (12%). In four cases no further therapies were administered (16%). Mean follow-up after the end of targeted therapy was 17.1 months. As neoadjuvant regimen, 36% of patients were treated with dabrafenib and trametinib with a near complete radiological response in all the cases with a mean treatment of 5.7 months. The neoadjuvant use of verumafenib (BRAFi) and cometinib (MEK1 inhibitor) induced a near complete response in 15 patients (94%), with a median volumetric reduction between 85% and 91%. Ten patients did not receive further treatments. Side effects varied among studies. The optimal timing, sequencing, and duration of treatment of these new therapies should be established. Moreover, questions remain about the choice of specific BRAF/MEK inhibitors, the optimal protocol of treatment, and the strategies for managing adverse events. Conclusions: Treatment is shifting to a wider multidisciplinary management, where a key role is played by targeted therapies, to improve outcomes and quality of life for patients with BRAF-mutated craniopharyngiomas. Future, larger comparative trials will optimize their protocol of use and integration into multimodal strategies of treatment.
Collapse
Affiliation(s)
- Giulia Cossu
- Department of Neurosurgery, University Hospital of Lausanne and University of Lausanne, 1011 Lausanne, Switzerland; (R.T.D.)
- Department of Neurosurgery, University Hospital of Dijon Bourgogne, 21000 Dijon, France (C.C.); (W.F.); (M.L.)
| | - Daniele S. C. Ramsay
- Imperial Brain and Spine Initiative, London W2 1NY, UK
- Imperial College School of Medicine, London W2 1PG, UK
| | - Roy T. Daniel
- Department of Neurosurgery, University Hospital of Lausanne and University of Lausanne, 1011 Lausanne, Switzerland; (R.T.D.)
| | - Ahmed El Cadhi
- Department of Neurosurgery, University Hospital of Dijon Bourgogne, 21000 Dijon, France (C.C.); (W.F.); (M.L.)
| | - Luc Kerherve
- Department of Neurosurgery, University Hospital of Dijon Bourgogne, 21000 Dijon, France (C.C.); (W.F.); (M.L.)
| | - Edouard Morlaix
- Department of Neurosurgery, University Hospital of Dijon Bourgogne, 21000 Dijon, France (C.C.); (W.F.); (M.L.)
| | - Sayda A. Houidi
- Department of Neurosurgery, University Hospital of Dijon Bourgogne, 21000 Dijon, France (C.C.); (W.F.); (M.L.)
| | - Clément Millot-Piccoli
- Department of Neurosurgery, University Hospital of Dijon Bourgogne, 21000 Dijon, France (C.C.); (W.F.); (M.L.)
| | - Renan Chapon
- Department of Neurosurgery, University Hospital of Dijon Bourgogne, 21000 Dijon, France (C.C.); (W.F.); (M.L.)
| | - Tuan Le Van
- Department of Neurosurgery, University Hospital of Dijon Bourgogne, 21000 Dijon, France (C.C.); (W.F.); (M.L.)
| | - Catherine Cao
- Department of Neurosurgery, University Hospital of Dijon Bourgogne, 21000 Dijon, France (C.C.); (W.F.); (M.L.)
| | - Walid Farah
- Department of Neurosurgery, University Hospital of Dijon Bourgogne, 21000 Dijon, France (C.C.); (W.F.); (M.L.)
| | - Maxime Lleu
- Department of Neurosurgery, University Hospital of Dijon Bourgogne, 21000 Dijon, France (C.C.); (W.F.); (M.L.)
| | - Olivier Baland
- Department of Neurosurgery, University Hospital of Dijon Bourgogne, 21000 Dijon, France (C.C.); (W.F.); (M.L.)
| | - Jacques Beaurain
- Department of Neurosurgery, University Hospital of Dijon Bourgogne, 21000 Dijon, France (C.C.); (W.F.); (M.L.)
| | - Jean Michel Petit
- Department of Endocrinology, University Hospital of Dijon Bourgogne, 21000 Dijon, France
| | - Brivaël Lemogne
- Department of Neuroradiology, University Hospital of Dijon Bourgogne, 21000 Dijon, France
| | - Mahmoud Messerer
- Department of Neurosurgery, University Hospital of Lausanne and University of Lausanne, 1011 Lausanne, Switzerland; (R.T.D.)
| | - Moncef Berhouma
- Department of Neurosurgery, University Hospital of Dijon Bourgogne, 21000 Dijon, France (C.C.); (W.F.); (M.L.)
- Functional and Molecular Imaging Team (CNRS 6302), Molecular Chemistry Institute (ICMUB), University of Burgundy, 21078 Dijon, France
| |
Collapse
|
46
|
Pessei V, Macagno M, Mariella E, Congiusta N, Battaglieri V, Battuello P, Viviani M, Gionfriddo G, Lamba S, Lorenzato A, Oddo D, Idrees F, Cavaliere A, Bartolini A, Guarrera S, Linnebacher M, Monteonofrio L, Cardone L, Milella M, Bertotti A, Soddu S, Grassi E, Crisafulli G, Bardelli A, Barault L, Di Nicolantonio F. DNA demethylation triggers cell free DNA release in colorectal cancer cells. Genome Med 2024; 16:118. [PMID: 39385243 PMCID: PMC11462661 DOI: 10.1186/s13073-024-01386-5] [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: 07/31/2023] [Accepted: 09/18/2024] [Indexed: 10/12/2024] Open
Abstract
BACKGROUND Liquid biopsy based on cell-free DNA (cfDNA) analysis holds significant promise as a minimally invasive approach for the diagnosis, genotyping, and monitoring of solid malignancies. Human tumors release cfDNA in the bloodstream through a combination of events, including cell death, active and passive release. However, the precise mechanisms leading to cfDNA shedding remain to be characterized. Addressing this question in patients is confounded by several factors, such as tumor burden extent, anatomical and vasculature barriers, and release of nucleic acids from normal cells. In this work, we exploited cancer models to dissect basic mechanisms of DNA release. METHODS We measured cell loss ratio, doubling time, and cfDNA release in the supernatant of a colorectal cancer (CRC) cell line collection (N = 76) representative of the molecular subtypes previously identified in cancer patients. Association analyses between quantitative parameters of cfDNA release, cell proliferation, and molecular features were evaluated. Functional experiments were performed to test the impact of modulating DNA methylation on cfDNA release. RESULTS Higher levels of supernatant cfDNA were significantly associated with slower cell cycling and increased cell death. In addition, a higher cfDNA shedding was found in non-CpG Island Methylator Phenotype (CIMP) models. These results indicate a positive correlation between lower methylation and increased cfDNA levels. To explore this further, we exploited methylation microarrays to identify a subset of probes significantly associated with cfDNA shedding and derive a methylation signature capable of discriminating high from low cfDNA releasers. We applied this signature to an independent set of 176 CRC cell lines and patient derived organoids to select 14 models predicted to be low or high releasers. The methylation profile successfully predicted the amount of cfDNA released in the supernatant. At the functional level, genetic ablation of DNA methyl-transferases increased chromatin accessibility and DNA fragmentation, leading to increased cfDNA release in isogenic CRC cell lines. Furthermore, in vitro treatment of five low releaser CRC cells with a demethylating agent was able to induce a significant increase in cfDNA shedding. CONCLUSIONS Methylation status of cancer cell lines contributes to the variability of cfDNA shedding in vitro. Changes in methylation pattern are associated with cfDNA release levels and might be exploited to increase sensitivity of liquid biopsy assays.
Collapse
Affiliation(s)
- Valeria Pessei
- Candiolo Cancer Institute, FPO-IRCCS, Candiolo, Turin, Italy
| | - Marco Macagno
- Candiolo Cancer Institute, FPO-IRCCS, Candiolo, Turin, Italy
| | - Elisa Mariella
- Department of Oncology, University of Torino, Turin, Italy
- IFOM, the AIRC Institute of Molecular Oncology, Milan, Italy
| | - Noemi Congiusta
- Candiolo Cancer Institute, FPO-IRCCS, Candiolo, Turin, Italy
- Department of Oncology, University of Torino, Turin, Italy
| | - Vittorio Battaglieri
- Department of Oncology, University of Torino, Turin, Italy
- IFOM, the AIRC Institute of Molecular Oncology, Milan, Italy
| | - Paolo Battuello
- Department of Oncology, University of Torino, Turin, Italy
- IFOM, the AIRC Institute of Molecular Oncology, Milan, Italy
| | - Marco Viviani
- Candiolo Cancer Institute, FPO-IRCCS, Candiolo, Turin, Italy
- Department of Oncology, University of Torino, Turin, Italy
| | - Giulia Gionfriddo
- Candiolo Cancer Institute, FPO-IRCCS, Candiolo, Turin, Italy
- Department of Oncology, University of Torino, Turin, Italy
| | - Simona Lamba
- Department of Oncology, University of Torino, Turin, Italy
| | | | - Daniele Oddo
- Candiolo Cancer Institute, FPO-IRCCS, Candiolo, Turin, Italy
- Department of Oncology, University of Torino, Turin, Italy
| | - Fariha Idrees
- Candiolo Cancer Institute, FPO-IRCCS, Candiolo, Turin, Italy
- Department of Oncology, University of Torino, Turin, Italy
| | - Alessandro Cavaliere
- Candiolo Cancer Institute, FPO-IRCCS, Candiolo, Turin, Italy
- Department of Oncology, University of Torino, Turin, Italy
| | - Alice Bartolini
- Candiolo Cancer Institute, FPO-IRCCS, Candiolo, Turin, Italy
| | - Simonetta Guarrera
- Candiolo Cancer Institute, FPO-IRCCS, Candiolo, Turin, Italy
- IIGM-Italian Institute for Genomic Medicine, c/o IRCCS, Candiolo, Turin, Italy
| | - Michael Linnebacher
- Clinic of General Surgery, Molecular Oncology and Immunotherapy, UMR, Rostock, Germany
| | - Laura Monteonofrio
- Department of Research and Advanced Technologies, Regina Elena National Cancer Institute IRCCS, Rome, Italy
| | - Luca Cardone
- Department of Research and Advanced Technologies, Regina Elena National Cancer Institute IRCCS, Rome, Italy
| | - Michele Milella
- Section of Innovation Biomedicine - Oncology Area, Department of Engineering for Innovation Medicine, University of Verona and Verona University and Hospital Trust, Verona, Italy
| | - Andrea Bertotti
- Candiolo Cancer Institute, FPO-IRCCS, Candiolo, Turin, Italy
- Department of Oncology, University of Torino, Turin, Italy
| | - Silvia Soddu
- Department of Research and Advanced Technologies, Regina Elena National Cancer Institute IRCCS, Rome, Italy
| | - Elena Grassi
- Candiolo Cancer Institute, FPO-IRCCS, Candiolo, Turin, Italy
- Department of Oncology, University of Torino, Turin, Italy
| | | | - Alberto Bardelli
- Department of Oncology, University of Torino, Turin, Italy
- IFOM, the AIRC Institute of Molecular Oncology, Milan, Italy
| | - Ludovic Barault
- Candiolo Cancer Institute, FPO-IRCCS, Candiolo, Turin, Italy.
- Department of Oncology, University of Torino, Turin, Italy.
| | - Federica Di Nicolantonio
- Candiolo Cancer Institute, FPO-IRCCS, Candiolo, Turin, Italy.
- Department of Oncology, University of Torino, Turin, Italy.
| |
Collapse
|
47
|
Fu SW, Tang C, Tan X, Srivastava S. Liquid biopsy for early cancer detection: technological revolutions and clinical dilemma. Expert Rev Mol Diagn 2024; 24:937-955. [PMID: 39360748 DOI: 10.1080/14737159.2024.2408744] [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: 05/08/2024] [Accepted: 09/22/2024] [Indexed: 10/05/2024]
Abstract
INTRODUCTION Liquid biopsy is an innovative advancement in oncology, offering a noninvasive method for early cancer detection and monitoring by analyzing circulating tumor cells, DNA, RNA, and other biomarkers in bodily fluids. This technique has the potential to revolutionize precision oncology by providing real-time analysis of tumor dynamics, enabling early detection, monitoring treatment responses, and tailoring personalized therapies based on the molecular profiles of individual patients. AREAS COVERED In this review, the authors discuss current methodologies, technological challenges, and clinical applications of liquid biopsy. This includes advancements in detecting minimal residual disease, tracking tumor evolution, and combining liquid biopsy with other diagnostic modalities for precision oncology. Key areas explored are the sensitivity, specificity, and integration of multi-omics, AI, ML, and LLM technologies. EXPERT OPINION Liquid biopsy holds great potential to revolutionize cancer care through early detection and personalized treatment strategies. However, its success depends on overcoming technological and clinical hurdles, such as ensuring high sensitivity and specificity, interpreting results amidst tumor heterogeneity, and making tests accessible and affordable. Continued innovation and collaboration are crucial to fully realize the potential of liquid biopsy in improving early cancer detection, treatment, and monitoring.
Collapse
Affiliation(s)
- Sidney W Fu
- Division of Cancer Prevention, National Cancer Institute, National Institutes of Health, Rockville, MD, USA
| | - Cong Tang
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Lisbon, Portugal
| | - Xiaohui Tan
- Division of LS Research, LSBioscience, LLC, Frederick, USA
| | - Sudhir Srivastava
- Cancer Biomarkers Research Group, Division of Cancer Prevention, National Cancer Institute, National Institutes of Health, Rockville, MD, USA
| |
Collapse
|
48
|
Goldkorn A, Tangen C, Plets M, Bsteh D, Xu T, Pinski JK, Ingles S, Triche TJ, MacVicar GR, Vaena DA, Crispino AW, McConkey DJ, Lara PN, Hussain MHA, Quinn DI, Dorff TB, Lerner SP, Thompson I, Agarwal N. Circulating Tumor Cell Count and Overall Survival in Patients With Metastatic Hormone-Sensitive Prostate Cancer. JAMA Netw Open 2024; 7:e2437871. [PMID: 39374015 PMCID: PMC11581504 DOI: 10.1001/jamanetworkopen.2024.37871] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/25/2024] [Accepted: 08/13/2024] [Indexed: 10/08/2024] Open
Abstract
Importance In metastatic hormone-sensitive prostate cancer (mHSPC), new first-line combination therapies have enhanced overall survival (OS), but clinical outcomes for individual patients vary greatly and are difficult to predict. Peripheral blood circulating tumor cell (CTC) count is the most extensively validated prognostic liquid biomarker in metastatic castration-resistant prostate cancer (mCRPC), and recent studies have suggested that it may also be informative in mHSPC. Objective To examine the prognostic value of CTC count in men with mHSPC. Design, Setting, and Participants In this prognostic study, peripheral blood was drawn at registration (baseline) and at progression to mCRPC in the S1216 study (March 1, 2013, to July 15, 2017), a phase 3, prospective, randomized clinical trial in men with mHSPC. The CTCs were enumerated using a US Food and Drug Administration-cleared isolation platform. Counts were categorized as 0, 1 to 4, or 5 or more CTCs per 7.5 mL based on the prognostic value of these cut points in prior studies. The data analysis was performed between October 28, 2022, and June 15, 2023. Exposure Metastatic hormone-sensitive prostate cancer. Main Outcomes and Measures Circulating tumor cell count was evaluated for an association with 3 prespecified trial end points: OS, progression-free survival, and 7-month prostate-specific antigen, after adjusting for other baseline covariates using proportional hazards and logistic regression models. Results Of 1313 S1216 participants (median [IQR] age, 68 [44-92] years), evaluable samples from 503 (median [IQR] age, 69 [46-90] years) with newly diagnosed mHSPC were collected at baseline, and 93 samples were collected at progression. Baseline counts were 5 or more CTCs per 7.5 mL in 60 samples (11.9%), 1 to 4 CTCs per 7.5 mL in 107 samples (21.3%), and 0 CTCs per 7.5 mL in 336 samples (66.8%). Median OS for men with 5 or more CTCs per 7.5 mL was 27.9 months (95% CI, 24.1-31.2 months) compared with 56.2 months (95% CI, 45.7-69.8 months) for men with 1 to 4 CTCs per 7.5 mL and not reached at 78.0 months follow-up for men with 0 CTCs per 7.5 mL. After adjusting for baseline clinical covariates, men with 5 or more CTCs per 7.5 mL at baseline had a significantly higher hazard of death (hazard ratio, 3.22; 95% CI, 2.22-4.68) and disease progression (hazard ratio, 2.46; 95% CI, 1.76-3.43) and a lower likelihood of prostate-specific antigen complete response (odds ratio, 0.26; 95% CI, 0.12-0.54) compared with men with 0 CTCs per 7.5 mL at baseline. Adding baseline CTC count to other known prognostic factors (covariates only: area under the curve, 0.73; 95% CI, 0.67-0.79) resulted in an increased prognostic value for 3-year survival (area under the curve, 0.79; 95% CI, 0.73-0.84). Conclusions and Relevance In this prognostic study, the findings validate CTC count as a prognostic biomarker that improved upon existing prognostic factors and estimated vastly divergent survival outcomes regardless of subsequent lines of therapy. As such, baseline CTC count in mHSPC may serve as a valuable noninvasive biomarker to identify men likely to have poor survival who may benefit from clinical trials of intensified or novel regimens.
Collapse
Affiliation(s)
- Amir Goldkorn
- Division of Medical Oncology, Department of Medicine, Keck School of Medicine of USC, Los Angeles, California
- USC Norris Comprehensive Cancer Center, Los Angeles, California
| | - Catherine Tangen
- SWOG Statistics and Data Management Center, Fred Hutchinson Cancer Center, Seattle, Washington
| | - Melissa Plets
- SWOG Statistics and Data Management Center, Fred Hutchinson Cancer Center, Seattle, Washington
| | - Daniel Bsteh
- Division of Medical Oncology, Department of Medicine, Keck School of Medicine of USC, Los Angeles, California
- USC Norris Comprehensive Cancer Center, Los Angeles, California
| | - Tong Xu
- Division of Medical Oncology, Department of Medicine, Keck School of Medicine of USC, Los Angeles, California
- USC Norris Comprehensive Cancer Center, Los Angeles, California
| | - Jacek K. Pinski
- USC Norris Comprehensive Cancer Center, Los Angeles, California
| | - Sue Ingles
- Keck School of Medicine of USC, Los Angeles, California
| | | | | | - Daniel A. Vaena
- Holden Comprehensive Cancer Center, University of Iowa Health Care, Iowa City
| | | | | | - Primo N. Lara
- UC Davis Comprehensive Cancer Center, Sacramento, California
| | | | - David I. Quinn
- USC Norris Comprehensive Cancer Center, Los Angeles, California
| | - Tanya B. Dorff
- City of Hope Comprehensive Cancer Center, Duarte, California
| | - Seth Paul Lerner
- Scott Department of Urology, Dan L Duncan Cancer Center, Baylor College of Medicine, Houston, Texas
| | | | - Neeraj Agarwal
- Huntsman Cancer Institute, University of Utah, Salt Lake City
| |
Collapse
|
49
|
Erice E, Mitxelena-Iribarren O, Arana S, Lawrie CH, Mujika M. Efficient enrichment of free target sequences in an integrated microfluidic device for point-of-care detection systems. NANOMEDICINE : NANOTECHNOLOGY, BIOLOGY, AND MEDICINE 2024; 61:102771. [PMID: 38960366 DOI: 10.1016/j.nano.2024.102771] [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: 01/18/2024] [Revised: 05/07/2024] [Accepted: 06/23/2024] [Indexed: 07/05/2024]
Abstract
Nucleic acid biomarker detection has great importance in the diagnosis of disease, the monitoring of disease progression and the classification of patients according to treatment decision making. Nucleic acid biomarkers found in the blood of patients have generated a lot of interest due to the possibility of being detected non-invasively which makes them ideal for monitoring and screening tests and particularly amenable to point-of-care (POC) or self-testing. A major challenge to POC molecular diagnostics is the need to enrich the target to optimise detection. In this work, we describe a microfabricated device for the enrichment of short dsDNA target sequences, which is especially valuable for potential detection methods, as it improves the probability of effectively detecting the target in downstream analyses. The device integrated a heating element and a temperature sensor with a microfluidic chamber to carry out the denaturation of the dsDNA combined with blocking-probes to enrich the target. This procedure was validated by fluorescence resonance energy transfer (FRET) technique, labelling DNA with a fluorophore and a quencher. As proof of concept, a 23-mer long dsDNA sequence corresponding to the L858R mutation of the EGFR gene was used. The qualitative results obtained determined that the most optimal blocking rate was obtained with the incorporation of 11/12-mer blocking-probes at a total concentration of 6 μM. This device is a powerful DNA preparation tool, which is an indispensable initial step for subsequent detection of sequences via nucleic acid hybridisation methods.
Collapse
Affiliation(s)
- Elisa Erice
- CEIT-Basque Research and Technology Alliance (BRTA), Manuel Lardizabal 15, 20018 Donostia, San Sebastián, Spain; Universidad de Navarra, Tecnun, Manuel Lardizabal 13, 20018 Donostia, San Sebastián, Spain.
| | - Oihane Mitxelena-Iribarren
- CEIT-Basque Research and Technology Alliance (BRTA), Manuel Lardizabal 15, 20018 Donostia, San Sebastián, Spain; Universidad de Navarra, Tecnun, Manuel Lardizabal 13, 20018 Donostia, San Sebastián, Spain; Group of Bioengineering in Regeneration and Cancer, Biogipuzkoa Health Research Institute, San Sebastian, Spain
| | - Sergio Arana
- CEIT-Basque Research and Technology Alliance (BRTA), Manuel Lardizabal 15, 20018 Donostia, San Sebastián, Spain; Universidad de Navarra, Tecnun, Manuel Lardizabal 13, 20018 Donostia, San Sebastián, Spain
| | - Charles H Lawrie
- Molecular Oncology Group, Biogipuzkoa Health Research Institute, San Sebastian, Spain; IKERBASQUE, Basque Foundation for Science, Bilbao, Spain; Sino-Swiss Institute of Advanced Technology (SSIAT), University of Shanghai, Shanghai, China; Radcliffe Department of Medicine, University of Oxford, Oxford, UK
| | - Maite Mujika
- CEIT-Basque Research and Technology Alliance (BRTA), Manuel Lardizabal 15, 20018 Donostia, San Sebastián, Spain; Universidad de Navarra, Tecnun, Manuel Lardizabal 13, 20018 Donostia, San Sebastián, Spain
| |
Collapse
|
50
|
Fang C, Liu S, Xia J, Wu X, Zhu J, Ke G. Clinical significance of intensity-modulated radiotherapy (IMRT) to the distant metastatic lymph nodes for metastatic cervical cancer. BMC Cancer 2024; 24:1170. [PMID: 39304814 DOI: 10.1186/s12885-024-12895-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2024] [Accepted: 09/03/2024] [Indexed: 09/22/2024] Open
Abstract
OBJECTIVE To retrospectively explore the clinical significance of radiotherapy to the distant metastatic lymph nodes (cervical/ clavicular/ mediastinal et al.) in metastatic cervical cancer. Hereinto, these cervicothoracic lymph nodes were metastasized from IB1-IVA (initial stage at first treatment), and IVB initially had metastatic disease in these areas at diagnosis. METHODS Metastatic cervical cancer only with the distant cervicothoracic metastatic lymph nodes (cervical/ clavicular/ mediastinal et al.), without distant parenchymal organs metastasis such as lung, liver, bone, and peritoneum, were enrolled in the analysis. These patients were classified into IB1-IVA and IVB based on their initial stage of first treatment. All patients received IMRT for the distant metastatic lymph nodes. The progression-free survival (PFS) and overall survival (OS) were analyzed using the Kaplan-Meier method. RESULTS Overall, the median PFS was 9 months, and the median OS was 27 months. The subgroup analysis showed that for IB1-IVA, the median PFS was 11 months, and the median OS was 30.5 months. For IVB, the median PFS was 8 months, and the median OS was 16 months. CONCLUSION Radiotherapy is beneficial to the distant metastatic lymph nodes (cervical/ clavicular/ mediastinal et al.), and could effectively bring the longer PFS and OS for metastatic cervical cancer.
Collapse
Affiliation(s)
- Chi Fang
- Department of Gynecologic Oncology, Fudan University Shanghai Cancer Center, 270 Dong-An Road, Shanghai, 200032, China
| | - Suping Liu
- Department of Gynecologic Oncology, Fudan University Shanghai Cancer Center, 270 Dong-An Road, Shanghai, 200032, China
| | - Jie Xia
- Department of Digestive Endoscopic Center, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China
| | - Xiaohua Wu
- Department of Gynecologic Oncology, Fudan University Shanghai Cancer Center, 270 Dong-An Road, Shanghai, 200032, China.
| | - Jun Zhu
- Department of Gynecologic Oncology, Fudan University Shanghai Cancer Center, 270 Dong-An Road, Shanghai, 200032, China.
| | - Guihao Ke
- Department of Gynecologic Oncology, Fudan University Shanghai Cancer Center, 270 Dong-An Road, Shanghai, 200032, China.
| |
Collapse
|