1
|
Chen Z, Zhao J, Li Y, Feng X, Chen Y, Li Y, Nan X, Liu H, Dong B, Shen L, Zhang L. Predicting response to patients with gastric cancer via a dynamic-aware model with longitudinal liquid biopsy data. Gastric Cancer 2025:10.1007/s10120-025-01628-4. [PMID: 40526252 DOI: 10.1007/s10120-025-01628-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/20/2025] [Accepted: 05/21/2025] [Indexed: 06/19/2025]
Abstract
BACKGROUND Gastric cancer (GC) presents challenges in predicting treatment responses due to its patient-specific heterogeneity. Recently, liquid biopsies have emerged as a valuable data modality, offering essential cellular and molecular insights while facilitating the capture of time-sensitive information. This study aimed to leverage artificial intelligence (AI) technology to analyze longitudinal liquid biopsy data. METHODS We collected a dataset from longitudinal liquid biopsies of 91 patients at Peking Cancer Hospital, spanning from July 2019 to April 2022. This dataset included 1895 tumor-related cellular images and 1698 tumor marker indices. Subsequently, we introduced the Dynamic-Aware Model (DAM) to predict responses to GC treatment. DAM incorporates dynamic data through AI-engineered components, facilitating an in-depth longitudinal analysis. RESULTS Utilizing threefold cross-validation, DAM exhibited superior performance compared to traditional cell-counting methods, achieving an AUC of 0.807 in predicting GC treatment responses. In the test set, DAM maintained stable efficacy with an AUC of 0.802. Besides, DAM showed the capability to accurately predict treatment responses based on early treatment data. Moreover, DAM's visual analysis of attention mechanisms identified six dynamic visual features related to focus areas, which were strongly associated with treatment-response. CONCLUSIONS These findings represent a pioneering effort in applying AI technology to interpret longitudinal liquid biopsy data and employ visual analytics in GC. This approach provides a promising pathway toward precise response prediction and personalized treatment strategies for patients with GC.
Collapse
Affiliation(s)
- Zifan Chen
- Center for Data Science, Peking University, Haidian District, Beijing, 100080, China
| | - Jie Zhao
- National Engineering Laboratory for Big Data Analysis and Applications, Peking University, Beijing, China
- Peking University Changsha Institute for Computing and Digital Economy, Changsha, China
| | - Yanyan Li
- Department of Gastrointestinal Oncology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Peking University Cancer Hospital and Institute, Beijing, China
| | - Xujiao Feng
- Department of Gastrointestinal Oncology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Peking University Cancer Hospital and Institute, Beijing, China
| | - Yang Chen
- Department of Gastrointestinal Oncology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Peking University Cancer Hospital and Institute, Beijing, China
| | - Yilin Li
- Department of Gastrointestinal Oncology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Peking University Cancer Hospital and Institute, Beijing, China
| | - Xinyu Nan
- Center for Data Science, Peking University, Haidian District, Beijing, 100080, China
| | - Huimin Liu
- Guangzhou Medical University, Guangzhou, China
| | - Bin Dong
- Beijing International Center for Mathematical Research and the New Cornerstone Science Laboratory, Peking University, Beijing, China
- Center for Machine Learning Research, Peking University, Beijing, China
- Peking University Changsha Institute for Computing and Digital Economy, Changsha, China
| | - Lin Shen
- Department of Gastrointestinal Oncology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Peking University Cancer Hospital and Institute, Beijing, China
| | - Li Zhang
- Center for Data Science, Peking University, Haidian District, Beijing, 100080, China.
| |
Collapse
|
2
|
Boukouris AE, Michaelidou K, Joosse SA, Charpidou A, Mavroudis D, Syrigos KN, Agelaki S. A comprehensive overview of minimal residual disease in the management of early-stage and locally advanced non-small cell lung cancer. NPJ Precis Oncol 2025; 9:178. [PMID: 40514443 PMCID: PMC12166047 DOI: 10.1038/s41698-025-00984-9] [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: 03/23/2025] [Accepted: 05/30/2025] [Indexed: 06/16/2025] Open
Abstract
Novel therapies have significantly improved survival for non-metastatic non-small cell lung cancer (NSCLC), however recurrence remains a challenge. Current treatment and surveillance strategies rely on imaging and clinical assessments with limited sensitivity in early detection of disease progression. Liquid biopsy-mediated detection of minimal residual disease (MRD) allows monitoring of tumor activity at the molecular level before clinical and radiologic progression. Here, we review the current evidence for MRD in the adaptive management and surveillance of non-metastatic NSCLC, focusing on the missing links that prevent its widespread clinical adoption.
Collapse
Affiliation(s)
- Aristeidis E Boukouris
- Department of Medical Oncology, University General Hospital of Heraklion, Vassilika Vouton, Crete, Greece
- Laboratory of Translational Oncology, School of Medicine, University of Crete, Vassilika Vouton, Crete, Greece
| | - Kleita Michaelidou
- Laboratory of Translational Oncology, School of Medicine, University of Crete, Vassilika Vouton, Crete, Greece
| | - Simon A Joosse
- Department of Tumor Biology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Andriani Charpidou
- Oncology Unit, 3rd Department of Internal Medicine, National and Kapodistrian University of Athens, Athens, Greece
| | - Dimitrios Mavroudis
- Department of Medical Oncology, University General Hospital of Heraklion, Vassilika Vouton, Crete, Greece
- Laboratory of Translational Oncology, School of Medicine, University of Crete, Vassilika Vouton, Crete, Greece
| | - Konstantinos N Syrigos
- Oncology Unit, 3rd Department of Internal Medicine, National and Kapodistrian University of Athens, Athens, Greece
| | - Sofia Agelaki
- Department of Medical Oncology, University General Hospital of Heraklion, Vassilika Vouton, Crete, Greece.
- Laboratory of Translational Oncology, School of Medicine, University of Crete, Vassilika Vouton, Crete, Greece.
| |
Collapse
|
3
|
Madar S, Amor RE, Furman-Assaf S, Friedman E. Innovative Approaches to Early Detection of Cancer-Transforming Screening for Breast, Lung, and Hard-to-Screen Cancers. Cancers (Basel) 2025; 17:1867. [PMID: 40507348 PMCID: PMC12153683 DOI: 10.3390/cancers17111867] [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: 03/14/2025] [Revised: 05/25/2025] [Accepted: 05/29/2025] [Indexed: 06/16/2025] Open
Abstract
Early detection of cancer is crucial for improving patient outcomes. Traditional modalities such as mammography and low-dose computed tomography are effective but exhibit inherent limitations, including radiation exposure and accessibility challenges. This review explores innovative, non-invasive cancer screening methods, focusing on liquid biopsy and volatile organic compound (VOC)-based detection platforms. Liquid biopsy analyzes circulating tumor DNA and other biomarkers in bodily fluids, offering potential for early detection and monitoring of treatment response. VOC-based detection leverages unique metabolic signatures emitted by cancer cells, detectable in exhaled breath or other bodily emissions, providing a rapid and patient-friendly screening option. We provide a comprehensive overview of these advanced multi-cancer detection techniques to enhance diagnostic accuracy, accessibility, and patient adherence, and ultimately enhance survival rates and patient outcomes.
Collapse
Affiliation(s)
| | | | | | - Eitan Friedman
- Assuta Medical Centers, Tel-Aviv 6971028, Israel;
- Gray Faculty of Medical and Health Sciences, Tel-Aviv University, Tel-Aviv 6997801, Israel
| |
Collapse
|
4
|
Vyas A, Kumar K, Sharma A, Verma D, Bhatia D, Wahi N, Yadav AK. Advancing the frontier of artificial intelligence on emerging technologies to redefine cancer diagnosis and care. Comput Biol Med 2025; 191:110178. [PMID: 40228444 DOI: 10.1016/j.compbiomed.2025.110178] [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: 01/30/2025] [Revised: 04/04/2025] [Accepted: 04/07/2025] [Indexed: 04/16/2025]
Abstract
BACKGROUND Artificial Intelligence (AI) is capable of revolutionizing cancer therapy and advancing precision oncology via integrating genomics data and digitized health information. AI applications show promise in cancer prediction, prognosis, and treatment planning, particularly in radiomics, deep learning, and machine learning for early cancer diagnosis. However, widespread adoption requires comprehensive data and clinical validation. While AI has demonstrated advantages in treating common malignancies like lung and breast cancers, challenges remain in managing rare tumors due to limited datasets. AI's role in processing multi-omics data and supporting precision oncology decision-making is critical as genetic and health data become increasingly digitized. METHOD This review article presents current knowledge on AI and associated technologies, which are being utilized in the diagnosis and therapy of cancer. The applications of AI in radiomics, deep learning, and machine learning for cancer screening and treatment planning are examined. The study also explores the capabilities and limitations of predictive AI in diagnosis and prognosis, as well as generative AI, such as advanced chatbots, in patient and provider interactions. RESULTS AI can improve the early diagnosis and treatment of high-incidence cancers like breast and lung cancer. However, its application in rare cancers is limited by insufficient data for training and validation. AI can effectively process large-scale multi-omics data from DNA and RNA sequencing, enhancing precision oncology. Predictive AI aids in risk assessment and prognosis, while generative AI tools improve patient-provider communication. Despite these advancements, further research and technological progress are needed to overcome existing challenges. CONCLUSIONS AI holds transformative potential for cancer therapy, particularly in precision oncology, early detection, and personalized treatment planning. However, challenges such as data limitations in rare cancers, the need for clinical validation, and regulatory considerations must be addressed. Future advancements in AI could significantly improve decision-support systems in oncology, ultimately enhancing patient care and quality of life. The review highlights both the opportunities and obstacles in integrating AI into cancer diagnostics and therapeutics, calling for continued research and regulatory oversight.
Collapse
Affiliation(s)
- Akanksha Vyas
- Academy of Scientific and Innovative Research, Ghaziabad, 201002, India
| | - Krishan Kumar
- Department of Chemistry, Indian Institute of Technology Delhi, Hauz Khas, New Delhi, 110016, India
| | - Ayushi Sharma
- College of Medicine, Taipei Medical University, Taipei City, 110, Taiwan
| | - Damini Verma
- Centre for Nanotechnology, Indian Institute of Technology Roorkee, Roorkee, Uttarakhand, 247667, India
| | - Dhiraj Bhatia
- Department of Biological Sciences & Engineering, Indian Institute of Technology Gandhinagar, Near Palaj, Gandhinagar, Gujarat, 382355, India
| | - Nitin Wahi
- Department of Biotechnology, LNCT University, Kolar Road, Shirdipuram, Bhopal, Madhya Pradesh, 462042, India
| | - Amit K Yadav
- Department of Biological Sciences & Engineering, Indian Institute of Technology Gandhinagar, Near Palaj, Gandhinagar, Gujarat, 382355, India.
| |
Collapse
|
5
|
Malik S, Zaheer S. The impact of liquid biopsy in breast cancer: Redefining the landscape of non-invasive precision oncology. THE JOURNAL OF LIQUID BIOPSY 2025; 8:100299. [PMID: 40521566 PMCID: PMC12166835 DOI: 10.1016/j.jlb.2025.100299] [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: 04/15/2025] [Revised: 05/06/2025] [Accepted: 05/19/2025] [Indexed: 06/18/2025]
Abstract
Breast cancer (BC) remains a leading cause of morbidity and mortality among women worldwide, necessitating the development of innovative diagnostic and monitoring strategies. Liquid biopsy (LB), a minimally invasive approach that analyzes circulating tumor cells (CTCs), cell-free DNA (cfDNA), circulating tumor DNA (ctDNA), extracellular vesicles (EVs), and other tumor-derived biomarkers in body fluids, has emerged as a transformative tool in BC management. This review comprehensively explores the role of LB in early detection, disease monitoring, treatment stratification, and resistance surveillance in BC. We discuss the latest advancements in LB technologies, including next-generation sequencing (NGS), digital PCR, and single-cell analysis, highlighting their sensitivity and specificity. Additionally, we examine the clinical utility of LB in guiding personalized therapy, particularly in the context of hormone receptor-positive, HER2positive, and triple-negative BC subtypes. Despite its promise, several challenges, including standardization, validation, and integration into clinical practice, remain to be addressed. By summarizing current evidence and future directions, this review underscores the potential of LB to revolutionize BC diagnosis and treatment, paving the way for a more precise and dynamic approach to disease management.
Collapse
Affiliation(s)
- Shaivy Malik
- Department and Institution - Department of Pathology, Vardhman Mahavir Medical College and Safdarjung Hospital, New Delhi, India
| | - Sufian Zaheer
- Department and Institution - Department of Pathology, Vardhman Mahavir Medical College and Safdarjung Hospital, New Delhi, India
| |
Collapse
|
6
|
Zhu C, Chen L, Wei B, Yang G, Zhao L, Qu F. Investigation of Body Fluids Intervention Impact on Aptamer Screening via Capillary Electrophoresis. Anal Chem 2025; 97:10309-10318. [PMID: 40327044 DOI: 10.1021/acs.analchem.5c00369] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/07/2025]
Abstract
Aptamers have presented considerable potential for disease diagnoses but still face a great challenge from the discrepancy between their screening conditions and real application in body fluid matrices (BFMs). In this study, we investigated the impact of BFM intervention on aptamer screening based on CE efficient separation and introduced adaptive screening strategies directly under the BFM background. Five BFMs, including serum, tears, artificial cerebrospinal fluid, saliva, and embryonic fluid, demonstrated the potential differentiating effects on aptamer screening based on the characterization of their "fingerprint-like profile" and their interactions with ssDNA libraries. Besides, the matrix effects of the five BFMs were quantified by calculating kinetic parameters derived from binding investigations between disease marker protein-spiked BFMs and ssDNA libraries. Through quantitative evaluation, two tailored SELEX strategies were introduced in BFM-intervened aptamer screening: spiking foreign proteins to the BFMs, and targeting proteins naturally abundant in BFMs. As proof of concept, we completed aptamer screenings for prolactin-induced protein (serum, spiked target) and human lactoferrin (tears, natural target) via CE-SELEX that demonstrated its adaptability in "splitting" BFM interference and achieving a clear collection of target-ssDNA complex. This study highlighted the critical importance of incorporating BFMs into SELEX and provided two tailored strategies that are worthy of choice for effective aptamer discovery, advancing the practical utility of aptamers.
Collapse
Affiliation(s)
- Chao Zhu
- Key Laboratory of Molecular Medicine and Biotherapy, School of Life Science, Beijing Institute of Technology, 5 South Zhongguancun Street, Beijing 100081, China
- Institute of Quality Standard and Testing Technology for Agro-Products, Shandong Academy of Agricultural Sciences, Jinan 250100, China
| | - Li Chen
- Key Laboratory of Molecular Medicine and Biotherapy, School of Life Science, Beijing Institute of Technology, 5 South Zhongguancun Street, Beijing 100081, China
| | - Bo Wei
- Key Laboratory of Molecular Medicine and Biotherapy, School of Life Science, Beijing Institute of Technology, 5 South Zhongguancun Street, Beijing 100081, China
| | - Ge Yang
- CAMS Key Laboratory of Antiviral Drug Research, Beijing Key Laboratory of Antimicrobial Agents, NHC Key Laboratory of Biotechnology of Antibiotics, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
| | - Liping Zhao
- Precision and Intelligence Medical Imaging Lab, Beijing Friendship Hospital, Capital Medical University, Beijing 100050, China
| | - Feng Qu
- Key Laboratory of Molecular Medicine and Biotherapy, School of Life Science, Beijing Institute of Technology, 5 South Zhongguancun Street, Beijing 100081, China
| |
Collapse
|
7
|
Assunção RRS, Santos NL, Andrade LNDS. Extracellular vesicles as cancer biomarkers and drug delivery strategies in clinical settings: Advances, perspectives, and challenges. Clinics (Sao Paulo) 2025; 80:100635. [PMID: 40315797 PMCID: PMC12090321 DOI: 10.1016/j.clinsp.2025.100635] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/07/2025] [Accepted: 03/26/2025] [Indexed: 05/04/2025] Open
Abstract
Cancer is a leading cause of death worldwide, and despite the introduction of new therapeutic approaches for advanced cases aimed at improving patient survival, only a subset of patients benefits from a complete response. In this context, there is a growing need for new cancer biomarkers and therapeutic strategies, and the use of Extracellular Vesicles (EVs) has been widely explored in various approaches. As circulating lipid-bilayer particles carrying a variety of biological information from tumor cells, EVs can be employed as good biomarkers of diagnosis, prognosis, therapy evaluation, and as adjuvants in cancer treatment. In this review, we provide a brief overview of the different types of EVs and their biogenesis and discuss how tumor-derived EV cargo can serve as a potential biomarker in clinical settings through liquid biopsy. We also highlight recent advances in EV nanoengineering and their potential as adjuvants in cancer treatment. Finally, we discuss the key unknowns, gaps, and bottlenecks that must be addressed to fully integrate EVs into precision oncology.
Collapse
Affiliation(s)
- Raphaela Rebeca Silveira Assunção
- Center for Translational Research in Oncology (LIM/24), Instituto do Cancer do Estado de Sao Paulo, Hospital das Clinicas HCFMUSP, Faculdade de Medicina, Universidade de São Paulo, São Paulo, SP, Brazil; Comprehensive Center for Precision Oncology (C2PO), Universidade de Sao Paulo, São Paulo, SP, Brazil
| | - Nathalia Leal Santos
- Center for Translational Research in Oncology (LIM/24), Instituto do Cancer do Estado de Sao Paulo, Hospital das Clinicas HCFMUSP, Faculdade de Medicina, Universidade de São Paulo, São Paulo, SP, Brazil; Comprehensive Center for Precision Oncology (C2PO), Universidade de Sao Paulo, São Paulo, SP, Brazil
| | - Luciana Nogueira de Sousa Andrade
- Center for Translational Research in Oncology (LIM/24), Instituto do Cancer do Estado de Sao Paulo, Hospital das Clinicas HCFMUSP, Faculdade de Medicina, Universidade de São Paulo, São Paulo, SP, Brazil; Comprehensive Center for Precision Oncology (C2PO), Universidade de Sao Paulo, São Paulo, SP, Brazil.
| |
Collapse
|
8
|
Rahimi A, Baghernejadan Z, Hazrati A, Malekpour K, Samimi LN, Najafi A, Falak R, Khorramdelazad H. Combination therapy with immune checkpoint inhibitors in colorectal cancer: Challenges, resistance mechanisms, and the role of microbiota. Biomed Pharmacother 2025; 186:118014. [PMID: 40157004 DOI: 10.1016/j.biopha.2025.118014] [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: 01/08/2025] [Revised: 03/17/2025] [Accepted: 03/24/2025] [Indexed: 04/01/2025] Open
Abstract
Colorectal cancer (CRC) is still one of the leading causes of cancer deaths worldwide. Even though there has been progress in cancer immunotherapy, the results of applying immune checkpoint inhibitors (ICIs) have been unsatisfactory, especially in microsatellite stable (MSS) CRC. Single-agent ICIs that target programmed cell death-1 (PD-1)/ PD-L1, cytotoxic T-lymphocyte-associated protein 4 (CTLA-4), T cell Ig- and mucin-domain-containing molecule-3 (TIM-3), and lymphocyte activation gene (LAG)-3 have emerged as having specific benefits. However, many primary and secondary resistance mechanisms are available in the tumor microenvironment (TME) that prevent it from happening. Combination strategies, such as the use of anti-PD-1 and anti-CTLA-4, can be effective in overcoming these resistance pathways, but toxicities remain a significant concern. Moreover, ICIs have been integrated with various treatment modalities, including chemotherapy, radiotherapy, antibiotics, virotherapy, polyadenosine diphosphate-ribose polymerase (PARP) inhibitors, and heat shock protein 90 (HSP90) inhibitors. The outcomes observed in both preclinical and clinical settings have been encouraging. Interestingly, manipulating gut microbiota via fecal microbiota transplantation (FMT) has been identified as a new strategy to increase the efficacy of immunotherapy in CRC patients. Therefore, integrating ICIs with other treatment approaches holds promise in enhancing the prognosis of CRC patients. This review focuses on the unmet need for new biomarkers to select patients for combination therapies and the ongoing work to overcome resistance and immune checkpoint blockade.
Collapse
Affiliation(s)
- Ali Rahimi
- Department of Immunology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Zeinab Baghernejadan
- Department of Immunology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Ali Hazrati
- Department of Immunology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Kosar Malekpour
- Department of Immunology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | | | - Alireza Najafi
- Department of Immunology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Reza Falak
- Department of Immunology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran.
| | - Hossein Khorramdelazad
- Department of Immunology, School of Medicine, Rafsanjan University of Medical Sciences, Rafsanjan, Iran.
| |
Collapse
|
9
|
Zhang CY, Gu K, Chi MY, Gao XY, Gao L, Zhang NN, Liu YX, Li TZ. The application progress of PAMAM dendrimer in cancer imaging and treatment. JOURNAL OF BIOMATERIALS SCIENCE. POLYMER EDITION 2025:1-38. [PMID: 40293953 DOI: 10.1080/09205063.2025.2497623] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/07/2024] [Accepted: 12/03/2024] [Indexed: 04/30/2025]
Abstract
Polyamidoamine dendrimer (PAMAM) are effective carriers that transport diagnostic imaging reagents and drugs to the tumor site. Their excellent bio-compatibility and bio-degradability reduce damage to healthy tissues, resulting in improved treatment efficacy. Dendrimer molecules are particularly useful in targeted drug delivery within malignant cells. This article reviews recent progress of PAMAM in imaging and treating breast cancer, lung cancer, hepatocellular cancer, colorectal cancer, gastric cancer, prostate cancer, and glioblastoma. This review aims to provide new and feasible ideas for cancer diagnosis imaging and treatment while also serving as a significant reference point for personalized tumor therapy based on PAMAM materials.
Collapse
Affiliation(s)
- Cong-Ying Zhang
- Key Laboratory of Research on Human Genetic Diseases at Universities of Inner Mongolia Autonomous Region, Chifeng University, Inner Mongolia, China
- Basic Medical College, Chifeng University, Inner Mongolia, China
- Key Laboratory of Mechanism and Evaluation of Traditional Chinese & Mongolian Medicine, Chifeng University, Inner Mongolia, China
| | - Kai Gu
- Basic Medical College, Chifeng University, Inner Mongolia, China
- Key Laboratory of Mechanism and Evaluation of Traditional Chinese & Mongolian Medicine, Chifeng University, Inner Mongolia, China
| | - Meng-Yi Chi
- Key Laboratory of Mechanism and Evaluation of Traditional Chinese & Mongolian Medicine, Chifeng University, Inner Mongolia, China
| | - Xiao-Yan Gao
- Key Laboratory of Research on Human Genetic Diseases at Universities of Inner Mongolia Autonomous Region, Chifeng University, Inner Mongolia, China
- Basic Medical College, Chifeng University, Inner Mongolia, China
- Key Laboratory of Mechanism and Evaluation of Traditional Chinese & Mongolian Medicine, Chifeng University, Inner Mongolia, China
| | - Ling Gao
- Basic Medical College, Chifeng University, Inner Mongolia, China
- Key Laboratory of Mechanism and Evaluation of Traditional Chinese & Mongolian Medicine, Chifeng University, Inner Mongolia, China
| | - Nan-Nan Zhang
- Key Laboratory of Research on Human Genetic Diseases at Universities of Inner Mongolia Autonomous Region, Chifeng University, Inner Mongolia, China
- Basic Medical College, Chifeng University, Inner Mongolia, China
- Key Laboratory of Mechanism and Evaluation of Traditional Chinese & Mongolian Medicine, Chifeng University, Inner Mongolia, China
| | - Yu-Xi Liu
- Shaanxi Academy of Traditional Chinese Medicine, Shaanxi, China
| | - Tian-Zhu Li
- Key Laboratory of Research on Human Genetic Diseases at Universities of Inner Mongolia Autonomous Region, Chifeng University, Inner Mongolia, China
- Basic Medical College, Chifeng University, Inner Mongolia, China
- Key Laboratory of Mechanism and Evaluation of Traditional Chinese & Mongolian Medicine, Chifeng University, Inner Mongolia, China
| |
Collapse
|
10
|
Rafiq A, Kanavarioti A. The Potential and Limitations of the MinION/Yenos Platform for miRNA-Enabled Early Cancer Detection. Int J Mol Sci 2025; 26:3822. [PMID: 40332502 PMCID: PMC12027911 DOI: 10.3390/ijms26083822] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2025] [Revised: 04/11/2025] [Accepted: 04/15/2025] [Indexed: 05/08/2025] Open
Abstract
The 2024 Nobel Prize in Physiology or Medicine was awarded to the pioneers who reported that microRNAs (miRNAs) regulate and direct the switch between physiological and pathological pathways via their over- or underexpression. The discovery changed the medical landscape and there are many completed and on-going clinical studies based on miRNAs. MiRNAs occur at the femtomolar level in biological fluids and are typically quantified using amplification-based techniques. Experimental nanopores have illustrated potential for trace analysis including amplification-free miRNA quantification. We repurposed the MinION, the only commercially available nanopore array device, and developed unique probes and protocols to detect and measure miRNA copies in blood and urine. Here, we report that miRNA copies are proportional to the total RNA isolated from the biospecimen, and that three known miRNA cancer biomarkers, i.e., miR-21, miR-375, and miR-141, were more than 1.5-fold overexpressed in blood samples from breast, ovarian, prostate, pancreatic, lung, and colorectal cancer patients compared to healthy patients. In these cancer samples, miR-15b was not overexpressed, in agreement with earlier studies. In contrast to literature reports, sample variability was undetectable in this study. The potential and limitations of this ready-to-use MinION/Yenos platform for multiple-cancer early detection (MCED) using blood or urine are discussed.
Collapse
|
11
|
Meza-Morales W, Ayus-Martinez S, Jimenez-Osorio J, Buendia-Otero M, López L, Suleiman D, Suarez E, Freytes DO, Cunci L, Mora C. Functionalized screen-printed electrodes for non-invasive detection of vascular-endothelial cadherin in extracellular vesicles. RSC Adv 2025; 15:12609-12621. [PMID: 40264865 PMCID: PMC12012609 DOI: 10.1039/d4ra08926j] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2024] [Accepted: 04/07/2025] [Indexed: 04/24/2025] Open
Abstract
In this study, we developed a biosensor using a gold screen-printed electrode (Au-SPE) functionalized with mercaptoundecanoic acid (MUA) and an antibody for detecting the vascular-endothelial cadherin (CD144) as a endothelial biomarker protein on extracellular vesicles (EVs) isolated from saliva. The MUA functionalization provides a stable platform for immobilizing the CD144 antibody, ensuring the detection of the target protein. This biosensor combines Au-SPE technology with an immunoassay, offering a rapid, sensitive, and non-invasive method for detection of CD144 carried by EVs. Characterization of saliva-derived EVs using transmission electron microscopy (TEM) and nanoparticle tracking analysis (NTA) confirmed their morphology and size, which fell within the expected range of 80-180 nm. NTA indicated a lower concentration of particles in saliva-EVs than in serum-EVs (controls), highlighting the need for sensitive detection of EV cargos in this type of EV. Immunodetection confirmed the presence of CD144 in both saliva and serum-derived EVs, with higher concentrations in serum. Functionalization of Au-SPEs with MUA and CD144 antibodies was confirmed by significant resistance changes, and atomic force microscopy (AFM) was used to verify the preservation of EV morphology and their capturing post-immune adsorption. A calibration curve demonstrated the high sensitivity of the biosensor prototype for detecting CD144-positive EVs, with a limit of detection (LOD) of 0.111 ng mL-1 and a limit of quantification (LOQ) of 0.37 ng mL-1, requiring only 3 μL of EV-sample. This biosensor shows potential as a novel method for detecting and studying endothelial biomarkers associated with cardiovascular disease in EVs isolated from saliva, a capability not currently available with existing tools. Furthermore, it provides a key platform for expanding research to other biomarkers and diseases by monitoring protein cargos in the EVs, enhancing its utility across diverse clinical applications.
Collapse
Affiliation(s)
- William Meza-Morales
- Department of Chemical Engineering, University of Puerto Rico-Mayaguez Route 108 Mayaguez Puerto Rico USA
| | - Sahimy Ayus-Martinez
- Department of Chemical Engineering, University of Puerto Rico-Mayaguez Route 108 Mayaguez Puerto Rico USA
| | - Jesus Jimenez-Osorio
- Department of Chemical Engineering, University of Puerto Rico-Mayaguez Route 108 Mayaguez Puerto Rico USA
| | - Maria Buendia-Otero
- Department of Chemical Engineering, University of Puerto Rico-Mayaguez Route 108 Mayaguez Puerto Rico USA
| | - Luis López
- Department of Chemistry, University of Puerto Rico-Rio Piedras 601 Av. Universidad San Juan Puerto Rico USA
| | - David Suleiman
- Department of Chemical Engineering, University of Puerto Rico-Mayaguez Route 108 Mayaguez Puerto Rico USA
| | - Edu Suarez
- Department of Biology, University of Puerto Rico-Ponce Av. Santiago de los Caballeros Ponce Puerto Rico USA
| | - Donald O Freytes
- Lampe Joint Department of Biomedical Engineering, The University of North Carolina at Chapel Hill/North Carolina State University 4130 Engineering Building III, Campus Box 7115 Raleigh NC 27695 USA
| | - Lisandro Cunci
- Department of Chemistry, University of Puerto Rico-Rio Piedras 601 Av. Universidad San Juan Puerto Rico USA
| | - Camilo Mora
- Department of Chemical Engineering, University of Puerto Rico-Mayaguez Route 108 Mayaguez Puerto Rico USA
| |
Collapse
|
12
|
Figols M, Chekhun S, Fernández-Saorin M, Pérez-Criado I, Bautista A, Font A, Ruiz de Porras V. Tumor-Educated Platelets in Urological Tumors: A Novel Biosource in Liquid Biopsy. Int J Mol Sci 2025; 26:3595. [PMID: 40332071 PMCID: PMC12026913 DOI: 10.3390/ijms26083595] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2025] [Revised: 04/02/2025] [Accepted: 04/09/2025] [Indexed: 05/08/2025] Open
Abstract
Platelets, traditionally recognized for their role in hemostasis, have emerged as pivotal players in cancer biology. They actively contribute to tumor proliferation, angiogenesis, immune evasion, and metastasis and thus play a significant role in cancer progression. Tumor-educated platelets (TEPs) acquire protumorigenic phenotypes through RNA, protein, and receptor profile alterations driven by interactions with tumors and their microenvironment. These modifications enable TEPs to enhance tumor growth and dissemination and to play a critical role throughout the metastatic process. Moreover, TEPs are promising biomarkers that can easily be analyzed in liquid biopsies. Since they dynamically mirror tumor activity through transcriptomic and proteomic changes, their analysis offers a non-invasive method for determining cancer detection and diagnosis, patient prognosis, therapy monitoring, and personalization of treatment. Their demonstrated accuracy in identifying cancer types and predicting treatment responses underscores their ability to provide real-time insights into tumor biology, including in urological malignancies. Their diagnostic potential and their accessibility as blood-sourced biomarkers position TEPs as transformative tools in advancing personalized oncology. Here, we focus on the role of TEPs in urological tumors, exploring their applications in early cancer detection, disease monitoring, and the design of tailored therapeutic strategies.
Collapse
Affiliation(s)
- Mariona Figols
- Medical Oncology Department, Althaia Xarxa Assistencial Universitària de Manresa, C/ Dr. Joan Soler, 1-3, 08243 Manresa, Spain; (M.F.); (I.P.-C.); (A.B.)
- PhD Programme in Medicine and Biomedical Sciences, Doctoral School, University of Vic, Central University of Catalonia (UVic-UCC), C/ Dr. Junyent, 1, 08500 Vic, Spain
- Faculty of Medicine, University of Vic, Central University of Catalonia (UVicUCC), Can Baumann, Ctra, de Roda, 70, 08500 Vic, Spain
| | - Sviatoslav Chekhun
- CARE Program, Germans Trias i Pujol Research Institute (IGTP), Camí de les Escoles, s/n, 08916 Badalona, Spain; (S.C.); (M.F.-S.); (A.F.)
- Badalona Applied Research Group in Oncology (B⋅ARGO), Catalan Institute of Oncology, Camí de les Escoles, s/n, 08916 Badalona, Spain
- Medical Oncology Department, Catalan Institute of Oncology, Camí de les Escoles, s/n, 08916 Badalona, Spain
| | - Maria Fernández-Saorin
- CARE Program, Germans Trias i Pujol Research Institute (IGTP), Camí de les Escoles, s/n, 08916 Badalona, Spain; (S.C.); (M.F.-S.); (A.F.)
- Badalona Applied Research Group in Oncology (B⋅ARGO), Catalan Institute of Oncology, Camí de les Escoles, s/n, 08916 Badalona, Spain
| | - Ignacio Pérez-Criado
- Medical Oncology Department, Althaia Xarxa Assistencial Universitària de Manresa, C/ Dr. Joan Soler, 1-3, 08243 Manresa, Spain; (M.F.); (I.P.-C.); (A.B.)
| | - Ana Bautista
- Medical Oncology Department, Althaia Xarxa Assistencial Universitària de Manresa, C/ Dr. Joan Soler, 1-3, 08243 Manresa, Spain; (M.F.); (I.P.-C.); (A.B.)
| | - Albert Font
- CARE Program, Germans Trias i Pujol Research Institute (IGTP), Camí de les Escoles, s/n, 08916 Badalona, Spain; (S.C.); (M.F.-S.); (A.F.)
- Badalona Applied Research Group in Oncology (B⋅ARGO), Catalan Institute of Oncology, Camí de les Escoles, s/n, 08916 Badalona, Spain
- Medical Oncology Department, Catalan Institute of Oncology, Camí de les Escoles, s/n, 08916 Badalona, Spain
| | - Vicenç Ruiz de Porras
- CARE Program, Germans Trias i Pujol Research Institute (IGTP), Camí de les Escoles, s/n, 08916 Badalona, Spain; (S.C.); (M.F.-S.); (A.F.)
- Badalona Applied Research Group in Oncology (B⋅ARGO), Catalan Institute of Oncology, Camí de les Escoles, s/n, 08916 Badalona, Spain
- GRET and Toxicology Unit, Department of Pharmacology, Toxicology and Therapeutic Chemistry, Faculty of Pharmacy and Food Sciences, University of Barcelona, 08028 Barcelona, Spain
| |
Collapse
|
13
|
Villazon J, Dela Cruz N, Shi L. Cancer Cell Line Classification Using Raman Spectroscopy of Cancer-Derived Exosomes and Machine Learning. Anal Chem 2025; 97:7289-7298. [PMID: 40145503 PMCID: PMC11983372 DOI: 10.1021/acs.analchem.4c06966] [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: 12/22/2024] [Revised: 02/09/2025] [Accepted: 03/01/2025] [Indexed: 03/28/2025]
Abstract
Liquid biopsies are an emerging, noninvasive tool for cancer diagnostics, utilizing biological fluids for molecular profiling. Nevertheless, the current methods often lack the sensitivity and specificity necessary for early detection and real-time monitoring. This work explores an advanced approach to improving liquid biopsy techniques through machine learning analysis of the Raman spectra measured to classify distinct exosome solutions by their cancer origin. This was accomplished by conducting principal component analysis (PCA) of the Raman spectra of exosomes from three cancer cell lines (COLO205, A375, and LNCaP) to extract chemically significant features. This reduced set of features was then utilized to train a linear discriminant analysis (LDA) classifier to predict the source of the exosomes. Furthermore, we investigated differences in the lipid composition in these exosomes by their spectra. This spectral similarity analysis revealed differences in lipid profiles between the different cancer cell lines as well as identified the predominant lipids across all exosomes. Our PCA-LDA framework achieved 93.3% overall accuracy and F1 scores of 98.2%, 91.1%, and 91.0% for COLO205, A375, and LNCaP, respectively. Our results from spectral similarity analysis were also shown to support previous findings of lipid dynamics due to cancer pathology and pertaining to exosome function and structure. These findings underscore the benefits of enhancing Raman spectroscopy analysis with machine learning, laying the groundwork for the development of early noninvasive cancer diagnostics and personalized treatment strategies. This work potentially establishes the foundation for refining the classification model and optimizing exosome extraction and detection from clinical samples for clinical translation.
Collapse
Affiliation(s)
- Jorge Villazon
- Shu
Chien-Gene Lay Department of Bioengineering, University of California San Diego, La Jolla, California 92093, United States
| | - Nathaniel Dela Cruz
- Department
of Chemical and Biomolecular Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon 34141, Republic of Korea
| | - Lingyan Shi
- Shu
Chien-Gene Lay Department of Bioengineering, University of California San Diego, La Jolla, California 92093, United States
- Aiiso
Yufeng Li Family Department of Chemical and Nano Engineering, University of California San Diego, La Jolla, California 92093, United States
- Department
of Electrical and Computer Engineering, University of California San Diego, La Jolla, California 92093, United States
- Institute
of Engineering in Medicine, University of
California San Diego, La Jolla, California 92093, United States
- Synthetic
Biology Institute, University of California
San Diego, La Jolla, California 92093, United States
| |
Collapse
|
14
|
Oktem F, Akdeniz M, Al-Shaebi Z, Akyol G, Keklik M, Aydin O. SERS and Machine Learning-Enabled Liquid Biopsy: A Promising Tool for Early Detection and Recurrence Prediction in Acute Leukemia. ACS OMEGA 2025; 10:11887-11899. [PMID: 40191347 PMCID: PMC11966330 DOI: 10.1021/acsomega.4c08499] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/16/2024] [Revised: 02/23/2025] [Accepted: 02/27/2025] [Indexed: 04/09/2025]
Abstract
Acute leukemia (AL), classified as acute myeloid leukemia (AML) and acute lymphocytic leukemia (ALL), is a hematologic malignancy caused by the uncontrolled proliferation of leucocytes in the bone marrow. Early detection of AL is crucial for clinical treatment. Detection methods of AL are currently blood tests, bone marrow tests, imaging, and spinal fluid tests. However, these tests have drawbacks, such as high cost and time consumption. Liquid biopsy using biological fluids such as blood or serum is an emerging technique for noninvasive cancer detection and monitoring. Surface-enhanced Raman spectroscopy (SERS), which enhanced Raman signals by the interaction of plasmonic nanostructures with the analyte, is a highly sensitive and specific detection method with simple sample preparation that has been used in combination with machine learning techniques to analyze liquid biopsy. In this study, we developed a SERS-based liquid biopsy approach that enables accurate classification of AML and ALL subtypes and the prediction of disease recurrence. SERS spectra of serum samples from 24 healthy individuals, 43 AML patients, and 18 ALL patients were obtained using an Ag-based SERS substrate and clustered using hierarchical cluster analysis (HCA). The spectra were then classified using three commonly used classifiers, namely, support vector machine (SVM), random forest (RF), and k-nearest neighbor (kNN). Our findings demonstrate that the RF classifier has the highest accuracy values, with 96.1, 95.5, and 98.5% for classifying three groups and predicting the recurrence of AML and ALL, respectively. The combination of SERS-based serum analysis with machine learning algorithms represents a remarkable advancement in the realm of hematological disease diagnostics, particularly for AML and ALL. This approach not only facilitates the precise differentiation of disease subtypes but also introduces the novel capability of prognosticating disease recurrence.
Collapse
Affiliation(s)
- Fatih Oktem
- Department
of Hematology, Faculty of Medicine, Erciyes
University, 38039 Kayseri, Turkiye
| | - Munevver Akdeniz
- Department
of Biomedical Engineering, Erciyes University, 38039 Kayseri, Turkiye
- Nanothera
Lab, Drug Application and Research Center (ERFARMA), Erciyes University, 38039 Kayseri, Turkiye
| | - Zakarya Al-Shaebi
- Department
of Biomedical Engineering, Erciyes University, 38039 Kayseri, Turkiye
- Nanothera
Lab, Drug Application and Research Center (ERFARMA), Erciyes University, 38039 Kayseri, Turkiye
| | - Gulsah Akyol
- Department
of Hematology, Faculty of Medicine, Erciyes
University, 38039 Kayseri, Turkiye
| | - Muzaffer Keklik
- Department
of Hematology, Faculty of Medicine, Erciyes
University, 38039 Kayseri, Turkiye
| | - Omer Aydin
- Department
of Biomedical Engineering, Erciyes University, 38039 Kayseri, Turkiye
- Nanothera
Lab, Drug Application and Research Center (ERFARMA), Erciyes University, 38039 Kayseri, Turkiye
- Clinical
Engineering Research and Implementation Center (ERKAM), Erciyes University, 38040 Kayseri, Turkiye
- Nanotechnology
Research and Application Center (ERNAM), Erciyes University, 38040 Kayseri, Turkiye
| |
Collapse
|
15
|
Fu X, Luo Z, Deng Y, LaFramboise W, Bartlett D, Schwartz R. Marker selection strategies for circulating tumor DNA guided by phylogenetic inference. Bioinformatics 2025; 41:btaf145. [PMID: 40163695 PMCID: PMC12002908 DOI: 10.1093/bioinformatics/btaf145] [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: 06/04/2024] [Revised: 02/02/2025] [Accepted: 03/28/2025] [Indexed: 04/02/2025] Open
Abstract
MOTIVATION Blood-based profiling of tumor DNA ("liquid biopsy") offers great prospects for non-invasive early cancer diagnosis and clinical guidance, but requires further computational advances to become a robust quantitative assay of tumor clonal evolution. We propose new methods to better characterize tumor clonal dynamics from circulating tumor DNA (ctDNA), through application to two specific tasks: (i) applying longitudinal ctDNA data to refine phylogeny models of clonal evolution, and (ii) quantifying changes in clonal frequencies that may be indicative of treatment response or tumor progression. We pose these through a probabilistic framework for optimally identifying markers and using them to characterize clonal evolution. RESULTS We first estimate a density over clonal tree models using bootstrap samples over pre-treatment tissue-based sequence data. We then refine these models over successive longitudinal samples. We use the resulting framework for modeling and refining tree densities to pose a set of optimization problems for selecting ctDNA markers to maximize measures of utility for reducing uncertainty in phylogeny models and quantifying clonal frequencies given the models. We tested our methods on synthetic data and showed them to be effective at refining tree densities and inferring clonal frequencies. Application to real tumor data further demonstrated the methods' effectiveness in refining a lineage model and assessing its clonal frequencies. The work shows the power of computational methods to improve marker selection, clonal lineage reconstruction, and clonal dynamics profiling for more precise and quantitative assays of somatic evolution and tumor progression. AVAILABILITY AND IMPLEMENTATION https://github.com/CMUSchwartzLab/Mase-phi.git. (DOI: 10.5281/zenodo.14776163).
Collapse
Affiliation(s)
- Xuecong Fu
- Department of Biological Sciences, Carnegie Mellon University, Pittsburgh, PA 15217, United States
| | - Zhicheng Luo
- Department of Biological Sciences, Carnegie Mellon University, Pittsburgh, PA 15217, United States
| | - Yueqian Deng
- Ray and Stephanie Lane Computational Biology Department, Carnegie Mellon University, Pittsburgh, PA 15217, United States
| | - William LaFramboise
- Allegheny Health Network Cancer Institute, Allegheny Health Network, Pittsburgh, PA 15212, United States
| | - David Bartlett
- Allegheny Health Network Cancer Institute, Allegheny Health Network, Pittsburgh, PA 15212, United States
| | - Russell Schwartz
- Department of Biological Sciences, Carnegie Mellon University, Pittsburgh, PA 15217, United States
- Ray and Stephanie Lane Computational Biology Department, Carnegie Mellon University, Pittsburgh, PA 15217, United States
| |
Collapse
|
16
|
Yin H, Zhang M, Zhang Y, Zhang X, Zhang X, Zhang B. Liquid biopsies in cancer. MOLECULAR BIOMEDICINE 2025; 6:18. [PMID: 40108089 PMCID: PMC11923355 DOI: 10.1186/s43556-025-00257-8] [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: 07/22/2024] [Revised: 02/14/2025] [Accepted: 02/23/2025] [Indexed: 03/22/2025] Open
Abstract
Cancer ranks among the most lethal diseases worldwide. Tissue biopsy is currently the primary method for the diagnosis and biological analysis of various solid tumors. However, this method has some disadvantages related to insufficient tissue specimen collection and intratumoral heterogeneity. Liquid biopsy is a noninvasive approach for identifying cancer-related biomarkers in peripheral blood, which allows for repetitive sampling across multiple time points. In the field of liquid biopsy, representative biomarkers include circulating tumor cells (CTCs), circulating tumor DNA (ctDNA), and exosomes. Many studies have evaluated the prognostic and predictive roles of CTCs and ctDNA in various solid tumors. Although these studies have limitations, the results of most studies appear to consistently demonstrate the correlations of high CTC counts and ctDNA mutations with lower survival rates in cancer patients. Similarly, a reduction in CTC counts throughout therapy may be a potential prognostic indicator related to treatment response in advanced cancer patients. Moreover, the biochemical characteristics of CTCs and ctDNA can provide information about tumor biology as well as resistance mechanisms against targeted therapy. This review discusses the current clinical applications of liquid biopsy in cancer patients, emphasizing its possible utility in outcome prediction and treatment decision-making.
Collapse
Affiliation(s)
- Hang Yin
- The First Affiliated Hospital of Dalian Medical University, Dalian, 116000, China
| | - Manjie Zhang
- The First Affiliated Hospital of Dalian Medical University, Dalian, 116000, China
| | - Yu Zhang
- Dalian Medical University, Dalian, 116000, China
| | - Xuebing Zhang
- The First Affiliated Hospital of Dalian Medical University, Dalian, 116000, China
| | - Xia Zhang
- Dalian Fifth People's Hospital, Dalian, 116000, China.
| | - Bin Zhang
- The First Affiliated Hospital of Dalian Medical University, Dalian, 116000, China.
| |
Collapse
|
17
|
Stojchevski R, Sutanto EA, Sutanto R, Hadzi-Petrushev N, Mladenov M, Singh SR, Sinha JK, Ghosh S, Yarlagadda B, Singh KK, Verma P, Sengupta S, Bhaskar R, Avtanski D. Translational Advances in Oncogene and Tumor-Suppressor Gene Research. Cancers (Basel) 2025; 17:1008. [PMID: 40149342 PMCID: PMC11940485 DOI: 10.3390/cancers17061008] [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: 02/10/2025] [Revised: 03/10/2025] [Accepted: 03/15/2025] [Indexed: 03/29/2025] Open
Abstract
Cancer, characterized by the uncontrolled proliferation of cells, is one of the leading causes of death globally, with approximately one in five people developing the disease in their lifetime. While many driver genes were identified decades ago, and most cancers can be classified based on morphology and progression, there is still a significant gap in knowledge about genetic aberrations and nuclear DNA damage. The study of two critical groups of genes-tumor suppressors, which inhibit proliferation and promote apoptosis, and oncogenes, which regulate proliferation and survival-can help to understand the genomic causes behind tumorigenesis, leading to more personalized approaches to diagnosis and treatment. Aberration of tumor suppressors, which undergo two-hit and loss-of-function mutations, and oncogenes, activated forms of proto-oncogenes that experience one-hit and gain-of-function mutations, are responsible for the dysregulation of key signaling pathways that regulate cell division, such as p53, Rb, Ras/Raf/ERK/MAPK, PI3K/AKT, and Wnt/β-catenin. Modern breakthroughs in genomics research, like next-generation sequencing, have provided efficient strategies for mapping unique genomic changes that contribute to tumor heterogeneity. Novel therapeutic approaches have enabled personalized medicine, helping address genetic variability in tumor suppressors and oncogenes. This comprehensive review examines the molecular mechanisms behind tumor-suppressor genes and oncogenes, the key signaling pathways they regulate, epigenetic modifications, tumor heterogeneity, and the drug resistance mechanisms that drive carcinogenesis. Moreover, the review explores the clinical application of sequencing techniques, multiomics, diagnostic procedures, pharmacogenomics, and personalized treatment and prevention options, discussing future directions for emerging technologies.
Collapse
Affiliation(s)
- Radoslav Stojchevski
- Friedman Diabetes Institute, Lenox Hill Hospital, Northwell Health, New York, NY 10022, USA;
- Feinstein Institutes for Medical Research, Manhasset, NY 11030, USA
- Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Hempstead, NY 11549, USA
| | - Edward Agus Sutanto
- CUNY School of Medicine, The City College of New York, 160 Convent Avenue, New York, NY 10031, USA;
| | - Rinni Sutanto
- New York Institute of Technology College of Osteopathic Medicine, Glen Head, NY 11545, USA;
| | - Nikola Hadzi-Petrushev
- Faculty of Natural Sciences and Mathematics, Institute of Biology, Ss. Cyril and Methodius University, 1000 Skopje, North Macedonia; (N.H.-P.)
| | - Mitko Mladenov
- Faculty of Natural Sciences and Mathematics, Institute of Biology, Ss. Cyril and Methodius University, 1000 Skopje, North Macedonia; (N.H.-P.)
| | - Sajal Raj Singh
- GloNeuro, Sector 107, Vishwakarma Road, Noida 201301, Uttar Pradesh, India (J.K.S.)
| | - Jitendra Kumar Sinha
- GloNeuro, Sector 107, Vishwakarma Road, Noida 201301, Uttar Pradesh, India (J.K.S.)
| | - Shampa Ghosh
- GloNeuro, Sector 107, Vishwakarma Road, Noida 201301, Uttar Pradesh, India (J.K.S.)
| | | | - Krishna Kumar Singh
- Symbiosis Centre for Information Technology (SCIT), Rajiv Gandhi InfoTech Park, Hinjawadi, Pune 411057, Maharashtra, India;
| | - Prashant Verma
- School of Management, BML Munjal University, NH8, Sidhrawali, Gurugram 122413, Haryana, India
| | - Sonali Sengupta
- Department of Gastroenterology, All India Institute of Medical Sciences (AIIMS), New Delhi 110029, India
| | - Rakesh Bhaskar
- School of Chemical Engineering, Yeungnam University, Gyeongsan 38541, Republic of Korea
- Research Institute of Cell Culture, Yeungnam University, Gyeongsan 38541, Republic of Korea
| | - Dimiter Avtanski
- Friedman Diabetes Institute, Lenox Hill Hospital, Northwell Health, New York, NY 10022, USA;
- Feinstein Institutes for Medical Research, Manhasset, NY 11030, USA
- Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Hempstead, NY 11549, USA
| |
Collapse
|
18
|
Cox M, Vitello D, Chawla A. Translating the multifaceted use of liquid biopsy to management of early disease in pancreatic adenocarcinoma. Front Oncol 2025; 15:1520717. [PMID: 40182037 PMCID: PMC11966063 DOI: 10.3389/fonc.2025.1520717] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2024] [Accepted: 02/25/2025] [Indexed: 04/05/2025] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is a leading cause of cancer-related mortality, primarily due to late stage at diagnosis. This review examines the multifaceted applications of liquid biopsy and circulating tumor DNA (ctDNA) analysis in the diagnosis and management of PDAC. We review the current literature on the technological advancements in liquid biopsy analysis such as next generation sequencing (NGS) and digital droplet PCR (ddPCR) as well as multi-omics technologies, highlighting their potential for accurate molecular subtyping through ctDNA analysis. This review highlights the significant role of ctDNA in the assessment of tumor behavior, disease subtyping, prediction and monitoring of treatment response, and evaluation of minimal residual disease. We discuss the implications of integrating liquid biopsy techniques into clinical practice as well as its challenges and limitations. By drawing insights from recent studies, this review aims to provide a comprehensive overview of how liquid biopsy and ctDNA analysis can enhance early disease management strategies in PDAC. We underscore the need for additional prospective studies and clinical trials to validate its feasibility and accuracy in order to establish clinical utility, with the ultimate goal of routine incorporation into practice to improve patient outcomes and transform the treatment landscape for PDAC.
Collapse
Affiliation(s)
- Madison Cox
- Division of Surgical Oncology, Department of Surgery, Northwestern University Feinberg School of Medicine, Chicago, IL, United States
- Northwestern Medicine Cancer Centers, Northwestern Medicine Regional Medical Group, Winfield, IL, United States
| | - Dominic Vitello
- Division of Surgical Oncology, Department of Surgery, Northwestern University Feinberg School of Medicine, Chicago, IL, United States
- Northwestern Quality Improvement, Research and Education in Surgery, Department of Surgery, Northwestern University Feinberg School of Medicine, Chicago, IL, United States
| | - Akhil Chawla
- Division of Surgical Oncology, Department of Surgery, Northwestern University Feinberg School of Medicine, Chicago, IL, United States
- Northwestern Medicine Cancer Centers, Northwestern Medicine Regional Medical Group, Winfield, IL, United States
- Northwestern Quality Improvement, Research and Education in Surgery, Department of Surgery, Northwestern University Feinberg School of Medicine, Chicago, IL, United States
- Robert H. Lurie Comprehensive Cancer Center of Northwestern University, Chicago, IL, United States
| |
Collapse
|
19
|
Sabit H, Attia MG, Mohamed N, Taha PS, Ahmed N, Osama S, Abdel-Ghany S. Beyond traditional biopsies: the emerging role of ctDNA and MRD on breast cancer diagnosis and treatment. Discov Oncol 2025; 16:271. [PMID: 40050490 PMCID: PMC11885725 DOI: 10.1007/s12672-025-01940-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/02/2024] [Accepted: 02/05/2025] [Indexed: 03/09/2025] Open
Abstract
Breast cancer management has traditionally relied on tissue biopsies and imaging, which offer limited insights into the disease. However, the discovery of circulating tumor DNA (ctDNA) and minimal residual disease (MRD) detection has revolutionized our approach to breast cancer. ctDNA, which is fragmented tumor DNA found in the bloodstream, provides a minimally invasive way to understand the tumor's genomic landscape, revealing heterogeneity and critical mutations that biopsies may miss. MRD, which indicates cancer cells that remain after treatment, can now be detected using ctDNA and other advanced methods, improving our ability to predict disease recurrence. This allows for personalized adjuvant therapies based on individual MRD levels, avoiding unnecessary treatments for patients with low MRD. This review discusses how ctDNA and MRD represent a paradigm shift towards personalized, genomically guided cancer care, which has the potential to significantly improve patient outcomes in breast cancer.
Collapse
Affiliation(s)
- Hussein Sabit
- Department of Medical Biotechnology, College of Biotechnology, Misr University for Science and Technology, P. O. Box 77, Giza, Egypt.
| | - Manar G Attia
- Department of Pharmaceutical Biotechnology, College of Biotechnology, Misr University for Science and Technology, P. O. Box 77, Giza, Egypt
| | - Nouran Mohamed
- Department of Environmental Biotechnology, College of Biotechnology, Misr University for Science and Technology, P. O. Box 77, Giza, Egypt
| | - Pancé S Taha
- Department of Pharmaceutical Biotechnology, College of Biotechnology, Misr University for Science and Technology, P. O. Box 77, Giza, Egypt
| | - Nehal Ahmed
- Department of Agriculture Biotechnology, College of Biotechnology, Misr University for Science and Technology, P. O. Box 77, Giza, Egypt
| | - Salma Osama
- Department of Agriculture Biotechnology, College of Biotechnology, Misr University for Science and Technology, P. O. Box 77, Giza, Egypt
| | - Shaimaa Abdel-Ghany
- Department of Environmental Biotechnology, College of Biotechnology, Misr University for Science and Technology, P. O. Box 77, Giza, Egypt
| |
Collapse
|
20
|
Thomas J, Mazzara E, Guller M, Landsberger H, Tham T, Cooper D, Pereira L, Kamdar D, Frank D, Miles B, Mandal R. Methodology of cfHPV-DNA Detection in Head and Neck Cancer: A Systematic Review and Meta-analysis. Otolaryngol Head Neck Surg 2025; 172:798-810. [PMID: 39624913 DOI: 10.1002/ohn.1056] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2024] [Revised: 10/03/2024] [Accepted: 11/01/2024] [Indexed: 02/22/2025]
Abstract
OBJECTIVE We aim to compare the diagnostic accuracy of the different methodologies used in the detection of cell-free human papillomavirus (HPV) DNA in HPV-associated head and neck squamous cell carcinoma detection using bivariate analysis methods. DATA SOURCES Pubmed, Embase, and Scopus were queried using a broad search strategy to search for relevant studies. REVIEW METHODS Test characteristics were extracted from 33 studies following literature screening, and underwent analyses utilizing a bivariate approach. Summary statistics were identified for each type of methodology, and forest plots and summary receiver operating characteristic curves were constructed. Bias was estimated using Deek's Funnel Plot and the QUADAS-2 tool. RESULTS In terms of diagnostic accuracy, digital droplet polymerase chain reaction (ddPCR) based testing exhibited the highest diagnostics odds ratio at 138 (59.5, 318), followed closely by next-generation sequencing (NGS) at 120 (39.7, 362), then by polymerase chain reaction (PCR) at 31.4 (14.4, 68.6), and quantitative PCR at 8.74 (4.63, 16.5). CONCLUSION NGS and ddPCR are comparable in overall diagnostic accuracy, bringing into question their relative roles in diagnosis and screening. Cost-effective ddPCR assays may serve as useful diagnostic and screening tests in the clinic with their low false positive rates and high sensitivity. However, NGS assays also offer high sensitivity and companion metrics, suggesting they may have a more precise role in disease monitoring. Importantly, assay development and benchmarking need further standardization to improve comparison between assays. Finally, saliva-based testing needs to be further investigated using NGS and ddPCR to further understand its limitations in disease detection and monitoring.
Collapse
Affiliation(s)
- Jerin Thomas
- Institute of Molecular Medicine, The Feinstein Institutes for Medical Research, Manhasset, New York, USA
- Department of Otolaryngology-Head and Neck Surgery, Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Hempstead, New York, USA
| | - Eden Mazzara
- Institute of Molecular Medicine, The Feinstein Institutes for Medical Research, Manhasset, New York, USA
- Department of Otolaryngology-Head and Neck Surgery, Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Hempstead, New York, USA
| | - Meytal Guller
- Institute of Molecular Medicine, The Feinstein Institutes for Medical Research, Manhasset, New York, USA
- Department of Otolaryngology-Head and Neck Surgery, Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Hempstead, New York, USA
| | - Hannah Landsberger
- Institute of Molecular Medicine, The Feinstein Institutes for Medical Research, Manhasset, New York, USA
- Department of Otolaryngology-Head and Neck Surgery, Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Hempstead, New York, USA
| | - Tristan Tham
- Department of Otolaryngology-Head and Neck Surgery, Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Hempstead, New York, USA
| | - Dylan Cooper
- Institute of Molecular Medicine, The Feinstein Institutes for Medical Research, Manhasset, New York, USA
- Department of Otolaryngology-Head and Neck Surgery, Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Hempstead, New York, USA
| | - Lucio Pereira
- Department of Otolaryngology-Head and Neck Surgery, Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Hempstead, New York, USA
| | - Dev Kamdar
- Department of Otolaryngology-Head and Neck Surgery, Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Hempstead, New York, USA
| | - Douglas Frank
- Department of Otolaryngology-Head and Neck Surgery, Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Hempstead, New York, USA
| | - Brett Miles
- Department of Otolaryngology-Head and Neck Surgery, Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Hempstead, New York, USA
| | - Rajarsi Mandal
- Institute of Molecular Medicine, The Feinstein Institutes for Medical Research, Manhasset, New York, USA
- Department of Otolaryngology-Head and Neck Surgery, Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Hempstead, New York, USA
| |
Collapse
|
21
|
Pettit N, Missen MV, Noriega A, Lash R. Outcomes for Emergency Presentations of Lung Cancer: A Scoping Review. J Emerg Med 2025; 70:50-67. [PMID: 39939186 DOI: 10.1016/j.jemermed.2024.09.015] [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: 06/03/2024] [Revised: 09/18/2024] [Accepted: 09/30/2024] [Indexed: 02/14/2025]
Abstract
BACKGROUND Lung cancer is frequently detected during visits to the emergency department (ED). The ED is crucial for identifying likely cases of lung cancer and coordinating the subsequent care for these patients. OBJECTIVES This scoping review aims to explore the definitions of emergency presentations (EPs) of lung cancer, along with mortality rates, cancer stage, and treatments for patients diagnosed with lung cancer following an EP. METHODS We conducted a scoping review of the literature on EPs of lung cancer, identifying 27 relevant articles out of 1338 initially screened. RESULTS Most studies originated from the United Kingdom, collectively reporting over 270,000 EPs of lung cancer. The majority of included studies provided strong evidence. Key findings revealed higher mortality rates among patients diagnosed with lung cancer through emergency presentations, with a significant proportion presenting at advanced stages. Patients with EPs were less likely to undergo surgical removal or receive radiotherapy. Lastly, only 66.7% of the studies defined an EP, with great heterogeneity among EP definitions. Methodological differences precluded meta-analysis. CONCLUSION Despite methodological heterogeneity, our synthesis indicates that patients presenting acutely with undiagnosed lung cancer often present at advanced stages and experience high mortality rates. These findings underscore the need for further research to develop evidence-based interventions for improving outcomes among ED patients with suspected lung cancer.
Collapse
Affiliation(s)
- Nicholas Pettit
- Department of Emergency Medicine, Indiana University School of Medicine, Indianapolis Indiana.
| | - Marissa Vander Missen
- Department of Emergency Medicine, Indiana University School of Medicine, Indianapolis Indiana
| | - Andrea Noriega
- Department of Emergency Medicine, Indiana University School of Medicine, Indianapolis Indiana
| | - Rebecca Lash
- Department of Emergency Medicine, Indiana University School of Medicine, Indianapolis Indiana; Children's Hospital of Los Angeles, Institute for Nursing and Interprofessional Research
| |
Collapse
|
22
|
Qi X, Lin S, Li M. Atomic force microscopy combined with microfluidics for label-free sorting and automated nanomechanics of circulating tumor cells in liquid biopsy. NANOSCALE 2025; 17:4695-4712. [PMID: 39865849 DOI: 10.1039/d4nr04033c] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/28/2025]
Abstract
Liquid biopsies are expected to advance cancer management, and particularly physical cues are gaining attention for indicating tumorigenesis and metastasis. Atomic force microscopy (AFM) has become a standard and important tool for detecting the mechanical properties of single living cells, but studies of developing AFM-based methods to efficiently measure the mechanical properties of circulating tumor cells (CTCs) in liquid biopsy for clinical utility are still scarce. Herein, we present a proof-of-concept study based on the complementary combination of AFM and microfluidics, which allows label-free sorting of individual CTCs and subsequent automated AFM measurements of the mechanical properties of CTCs. With the use of a microfluidic system containing contraction-expansion microchannels, specific cancer cell types were separated and harvested in a marker-independent manner. Subsequently, automated AFM indentation and force spectroscopy experiments were performed on the enriched cells under the precise guidance of the label-free identification of cells using a deep learning optical image recognition model. The effectiveness of the presented method was verified on three experimental sample systems, including mixed microspheres with different sizes, a mixture of different types of cancer cells, and a mixture of cancer cells and blood cells. The study illustrates a feasible framework based on the integration of AFM and microfluidics for non-destructive and efficient nanomechanical phenotyping of CTCs in bodily fluids, which offers additional possibilities for the clinical applications of AFM-based nanomechanical analysis and will also benefit the field of mechanobiology as well as cancer liquid biopsy.
Collapse
Affiliation(s)
- Xiaoqun Qi
- School of Automation and Electrical Engineering, Shenyang Ligong University, Shenyang 110159, China
- State Key Laboratory of Robotics, Shenyang Institute of Automation, Chinese Academy of Sciences, Shenyang 110016, China.
| | - Sen Lin
- School of Automation and Electrical Engineering, Shenyang Ligong University, Shenyang 110159, China
| | - Mi Li
- State Key Laboratory of Robotics, Shenyang Institute of Automation, Chinese Academy of Sciences, Shenyang 110016, China.
| |
Collapse
|
23
|
Sheriff S, Saba M, Patel R, Fisher G, Schroeder T, Arnolda G, Luo D, Warburton L, Gray E, Long G, Braithwaite J, Rizos H, Ellis LA. A scoping review of factors influencing the implementation of liquid biopsy for cancer care. J Exp Clin Cancer Res 2025; 44:50. [PMID: 39934875 PMCID: PMC11817833 DOI: 10.1186/s13046-025-03322-w] [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: 10/21/2024] [Accepted: 02/06/2025] [Indexed: 02/13/2025] Open
Abstract
BACKGROUND Liquid biopsy (LB) offers a promising, minimally invasive alternative to traditional tissue biopsies in cancer care, enabling real-time monitoring and personalized treatment. Despite its potential, the routine implementation of LB in clinical practice faces significant challenges. This scoping review examines the barriers and facilitators influencing the implementation of liquid biopsies into standard cancer care. METHODS Four academic databases (PubMed, Scopus, Embase, and Web of Science) were systematically searched without language restrictions. We included peer-reviewed articles that were published between January 2019 and March 2024 that focused on the implementation of LB in cancer care or described barriers and facilitators to its implementation. Data relevant to the review objective, including key article characteristics; barriers and facilitators of implementation; and recommendations for advancement or optimisation; were extracted and analysed using thematic and visual network analyses. RESULTS The majority of the included articles were narrative review articles (84%), with most from China (24.2%) and the United States (20%). Thematic analysis identified four main categories and their associated barriers and facilitators to the implementation of LB in cancer care: (1) Laboratory and personnel requirements; (2) Disease specificity; (3) Biomarker-based liquid biopsy; and (4) Policy and regulation. The majority of barriers identified were concentrated in the pre-analytical phase, highlighting the lack of standardization in LB technologies and outcomes. CONCLUSIONS Through a thematic analysis of the barriers and facilitators to LB implementation, we present an integrated tool designed to encourage the standardization of testing methods for clinical practice guidelines in the field.
Collapse
Affiliation(s)
- Samran Sheriff
- Centre for Healthcare Resilience and Implementation Science, Australian Institute of Health Innovation, Macquarie University, Level 6, 75 Talavera Road, North Ryde, Sydney, NSW, Australia.
| | - Maree Saba
- Centre for Healthcare Resilience and Implementation Science, Australian Institute of Health Innovation, Macquarie University, Level 6, 75 Talavera Road, North Ryde, Sydney, NSW, Australia
| | - Romika Patel
- Centre for Healthcare Resilience and Implementation Science, Australian Institute of Health Innovation, Macquarie University, Level 6, 75 Talavera Road, North Ryde, Sydney, NSW, Australia
| | - Georgia Fisher
- Centre for Healthcare Resilience and Implementation Science, Australian Institute of Health Innovation, Macquarie University, Level 6, 75 Talavera Road, North Ryde, Sydney, NSW, Australia
| | - Tanja Schroeder
- Centre for Healthcare Resilience and Implementation Science, Australian Institute of Health Innovation, Macquarie University, Level 6, 75 Talavera Road, North Ryde, Sydney, NSW, Australia
| | - Gaston Arnolda
- Centre for Healthcare Resilience and Implementation Science, Australian Institute of Health Innovation, Macquarie University, Level 6, 75 Talavera Road, North Ryde, Sydney, NSW, Australia
| | - Dan Luo
- The Daffodil Centre, Sydney, NSW, Australia
| | - Lydia Warburton
- Centre for Precision Health, Edith Cowan University, Joondalup, WA, Australia
- Department of Medical Oncology, Fiona Stanly Hospital, Murdoch, WA, Australia
| | - Elin Gray
- Centre for Precision Health, Edith Cowan University, Joondalup, WA, Australia
- School of Medical and Health Sciences, Edith Cowan University, Joondalup, WA, Australia
| | - Georgina Long
- Melanoma Institute Australia, The University of Sydney, Sydney, NSW, Australia
- Faculty of Medicine & Health, The University of Sydney, Sydney, NSW, Australia
- Charles Perkins Centre, The University of Sydney, Sydney, NSW, Australia
- Royal North Shore and Mater Hospitals, North Sydney, Sydney, NSW, Australia
| | - Jeffrey Braithwaite
- Centre for Healthcare Resilience and Implementation Science, Australian Institute of Health Innovation, Macquarie University, Level 6, 75 Talavera Road, North Ryde, Sydney, NSW, Australia
| | - Helen Rizos
- Melanoma Institute Australia, The University of Sydney, Sydney, NSW, Australia
- Macquarie Medical School, Faculty of Medicine Health and Human Science, Macquarie University, Sydney, NSW, Australia
| | - Louise Ann Ellis
- Centre for Healthcare Resilience and Implementation Science, Australian Institute of Health Innovation, Macquarie University, Level 6, 75 Talavera Road, North Ryde, Sydney, NSW, Australia
| |
Collapse
|
24
|
Rodriguez-Nieves AL, Shah S, Taylor ML, Alle M, Huang X. Magnetic-Plasmonic Core-Shell Nanoparticles: Properties, Synthesis and Applications for Cancer Detection and Treatment. NANOMATERIALS (BASEL, SWITZERLAND) 2025; 15:264. [PMID: 39997827 PMCID: PMC11858323 DOI: 10.3390/nano15040264] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/29/2024] [Revised: 01/27/2025] [Accepted: 02/07/2025] [Indexed: 02/26/2025]
Abstract
Nanoparticles have been widely used in cancer diagnostics and treatment research due to their unique properties. Magnetic nanoparticles are popular in imaging techniques due to their ability to alter the magnetization field around them. Plasmonic nanoparticles are mainly applied in cancer treatments like photothermal therapy due to their ability to convert light into heat. While these nanoparticles are popular among their respective fields, magnetic-plasmonic core-shell nanoparticles (MPNPs) have gained popularity in recent years due to the combined magnetic and optical properties from the core and shell. MPNPs have stood out in cancer theranostics as a multimodal platform capable of serving as a contrast agent for imaging, a guidable drug carrier, and causing cellular ablation through photothermal energy conversion. In this review, we summarize the different properties of MPNPs and the most common synthesis approaches. We particularly discuss applications of MPNPs in cancer diagnosis and treatment based on different mechanisms using the magnetic and optical properties of the particles. Lastly, we look into current challenges they face for clinical applications and future perspectives using MPNPs for cancer detection and therapy.
Collapse
Affiliation(s)
| | | | | | | | - Xiaohua Huang
- Department of Chemistry, The University of Memphis, Memphis, TN 38152, USA; (A.L.R.-N.); (S.S.); (M.L.T.); (M.A.)
| |
Collapse
|
25
|
Kansara V, Patel M. Exploring the role of graphene-metal hybrid nanomaterials as Raman signal enhancers in early stage cancer detection. Talanta 2025; 283:127185. [PMID: 39532051 DOI: 10.1016/j.talanta.2024.127185] [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: 07/15/2024] [Revised: 10/24/2024] [Accepted: 11/06/2024] [Indexed: 11/16/2024]
Abstract
Molecular diagnosis plays a significant role in detection of biomolecules linked to early stage cancer since it offers greater sensitivity and reliability for identification of biomarker level changes as the disease progresses. The application of vibrational spectroscopy in biomarker detection is defined by the fingerprint spectrum of a molecule originating from single-molecule vibrations. This characteristic makes surface enhanced Raman spectroscopy (SERS) a promising tool for identification of biomarkers. The performance of the SERS technique largely depends on the material being used as the SERS substrate. Graphene, with its large surface area and abundance of aromatic regions, is considered advantageous as SERS substrate. Combining graphene with metal nanomaterials considerably increases SERS signal intensity, thereby enhancing detection sensitivity. Therefore, this review emphasizes the significance of selecting graphene-metal nanohybrids as suitable SERS substrates for signal amplification. The detail understanding of the mechanism of graphene-metal hybrid in SERS based detection of early stage cancer is also presented. Furthermore, several examples demonstrated the application of graphene-metal hybrid nanomaterials in detecting biomarkers and cancer cell differentiation using SERS imaging.
Collapse
Affiliation(s)
- Vrushti Kansara
- Maliba Pharmacy College, Uka Tarsadia University, Surat 394350, Gujarat, India
| | - Mitali Patel
- Maliba Pharmacy College, Uka Tarsadia University, Surat 394350, Gujarat, India.
| |
Collapse
|
26
|
Kattner AA. Inherently imperfect, inherently evolving - The pursuit of precision through biomarkers. Biomed J 2025; 48:100839. [PMID: 40015558 PMCID: PMC11925082 DOI: 10.1016/j.bj.2025.100839] [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: 02/22/2025] [Accepted: 02/25/2025] [Indexed: 03/01/2025] Open
Abstract
Featuring a special section on cancer biomarkers, this issue of the Biomedical Journal highlights research on cfDNA, fecal miRNA, mitochondrial pathways, Epstein-Barr virus DNA, multi-biomarker panels combined with LC-MS-based methods, and circulating tumor cell (CTC) growth status as potential tools for cancer detection and management, including colorectal cancer, ovarian cancer, nasopharyngeal carcinoma, and hepatocellular carcinoma. Additionally, it explores the social stigma surrounding mpox, and discusses the achievements in miRNA research that earned the 2024 Nobel Prize in Physiology or Medicine. Advances in deep learning for trauma imaging are examined, alongside a review of decades of enterovirus research and current vaccination strategies. Other studies investigate the combined use of anthelmintic and antibiotic therapy for roundworm infection, and the application of a well-established Traditional Chinese Medicine herbal formula, commonly prescribed for gynecological conditions, against autoimmune hepatitis. Further articles elucidate the role of the nuclear receptor HNF4 in Schistosoma, mitochondrial dysfunction in acute kidney injury, the effects of astragalus polysaccharides on prostate cancer, and the use of deep learning to predict mortality from electrocardiograms.
Collapse
|
27
|
Yu B, Shao S, Ma W. Frontiers in pancreatic cancer on biomarkers, microenvironment, and immunotherapy. Cancer Lett 2025; 610:217350. [PMID: 39581219 DOI: 10.1016/j.canlet.2024.217350] [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: 09/20/2024] [Revised: 11/06/2024] [Accepted: 11/21/2024] [Indexed: 11/26/2024]
Abstract
Pancreatic cancer remains one of the most challenging malignancies to treat due to its late-stage diagnosis, aggressive progression, and high resistance to existing therapies. This review examines the latest advancements in early detection, and therapeutic strategies, with a focus on emerging biomarkers, tumor microenvironment (TME) modulation, and the integration of artificial intelligence (AI) in data analysis. We highlight promising biomarkers, including microRNAs (miRNAs) and circulating tumor DNA (ctDNA), that offer enhanced sensitivity and specificity for early-stage diagnosis when combined with multi-omics panels. A detailed analysis of the TME reveals how components such as cancer-associated fibroblasts (CAFs), immune cells, and the extracellular matrix (ECM) contribute to therapy resistance by creating immunosuppressive barriers. We also discuss therapeutic interventions that target these TME components, aiming to improve drug delivery and overcome immune evasion. Furthermore, AI-driven analyses are explored for their potential to interpret complex multi-omics data, enabling personalized treatment strategies and real-time monitoring of treatment response. We conclude by identifying key areas for future research, including the clinical validation of biomarkers, regulatory frameworks for AI applications, and equitable access to innovative therapies. This comprehensive approach underscores the need for integrated, personalized strategies to improve outcomes in pancreatic cancer.
Collapse
Affiliation(s)
- Baofa Yu
- Taimei Baofa Cancer Hospital, Dongping, Shandong, 271500, China; Jinan Baofa Cancer Hospital, Jinan, Shandong, 250000, China; Beijing Baofa Cancer Hospital, Beijing, 100010, China; Immune Oncology Systems, Inc, San Diego, CA, 92102, USA.
| | - Shengwen Shao
- Institute of Microbiology and Immunology, Huzhou University School of Medicine, Huzhou, Zhejiang, 313000, China.
| | - Wenxue Ma
- Department of Medicine, Sanford Stem Cell Institute, and Moores Cancer Center, University of California San Diego, La Jolla, CA, 92093, USA.
| |
Collapse
|
28
|
He KJ, Wang H, Xu J, Gong G. Global, regional, and national burden of tracheal, bronchus, and lung cancer attributable to ambient particulate matter pollution among adults aged 70 and above in 1990-2021 and projected to 2044. Front Public Health 2025; 13:1524534. [PMID: 39916713 PMCID: PMC11799284 DOI: 10.3389/fpubh.2025.1524534] [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: 11/07/2024] [Accepted: 01/10/2025] [Indexed: 02/09/2025] Open
Abstract
Background Tracheal, bronchus, and lung (TBL) cancer attributable to ambient particulate matter pollution (APMP) is a growing global health concern, particularly in individuals aged 70 and above. This study aims to evaluate past trends, identify key drivers, and project future disease burden. Methods Data from the Global Burden of Disease Study 2021 was analyzed for TBL cancer-related disability-adjusted life years (DALYs) and mortality from 1990 to 2021, stratified by SDI regions. Statistical methods, including Joinpoint regression, age-period-cohort modeling, and decomposition analysis, were used to identify temporal trends and drivers of DALYs. Future projections were made using the Nordpred model. Results From 1990 to 2021, global DALYs of TBL cancer due to APMP increased steadily (AAPC 0.75%). Population growth was the main driver, accounting for 79.37% of the increase, with epidemiological factors playing a varying role across regions. The highest DALY growth was observed in middle SDI regions (AAPC 2.99%), while high SDI regions saw a decline (AAPC -1.76%). Projections up to 2044 suggest a substantial increase in DALYs across all SDI regions, with the fastest growth expected among individuals aged 70-74, but DALY rates are projected to decline steadily. Conclusion Population growth is the primary factor driving the increase in DALYs associated with TBL cancer, with significant regional disparities. Projections suggest a continued rise in disease burden, particularly in lower SDI regions, underlining the urgency for targeted public health interventions and strategies to mitigate exposure and improve healthcare outcomes for at-risk populations.
Collapse
Affiliation(s)
- Ke-Jie He
- Quzhou People’s Hospital, The Quzhou Affiliated Hospital of Wenzhou Medical University, Quzhou, China
| | - Haitao Wang
- The School of Clinical Medical Sciences, Southwest Medical University, Luzhou, Sichuan, China
| | - Jianguang Xu
- Quzhou People’s Hospital, The Quzhou Affiliated Hospital of Wenzhou Medical University, Quzhou, China
| | - Guoyu Gong
- School of Medicine, Xiamen University, Xiamen, China
| |
Collapse
|
29
|
Rafanan J, Ghani N, Kazemeini S, Nadeem-Tariq A, Shih R, Vida TA. Modernizing Neuro-Oncology: The Impact of Imaging, Liquid Biopsies, and AI on Diagnosis and Treatment. Int J Mol Sci 2025; 26:917. [PMID: 39940686 PMCID: PMC11817476 DOI: 10.3390/ijms26030917] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2024] [Revised: 01/18/2025] [Accepted: 01/20/2025] [Indexed: 02/16/2025] Open
Abstract
Advances in neuro-oncology have transformed the diagnosis and management of brain tumors, which are among the most challenging malignancies due to their high mortality rates and complex neurological effects. Despite advancements in surgery and chemoradiotherapy, the prognosis for glioblastoma multiforme (GBM) and brain metastases remains poor, underscoring the need for innovative diagnostic strategies. This review highlights recent advancements in imaging techniques, liquid biopsies, and artificial intelligence (AI) applications addressing current diagnostic challenges. Advanced imaging techniques, including diffusion tensor imaging (DTI) and magnetic resonance spectroscopy (MRS), improve the differentiation of tumor progression from treatment-related changes. Additionally, novel positron emission tomography (PET) radiotracers, such as 18F-fluoropivalate, 18F-fluoroethyltyrosine, and 18F-fluluciclovine, facilitate metabolic profiling of high-grade gliomas. Liquid biopsy, a minimally invasive technique, enables real-time monitoring of biomarkers such as circulating tumor DNA (ctDNA), extracellular vesicles (EVs), circulating tumor cells (CTCs), and tumor-educated platelets (TEPs), enhancing diagnostic precision. AI-driven algorithms, such as convolutional neural networks, integrate diagnostic tools to improve accuracy, reduce interobserver variability, and accelerate clinical decision-making. These innovations advance personalized neuro-oncological care, offering new opportunities to improve outcomes for patients with central nervous system tumors. We advocate for future research integrating these tools into clinical workflows, addressing accessibility challenges, and standardizing methodologies to ensure broad applicability in neuro-oncology.
Collapse
Affiliation(s)
| | | | | | | | | | - Thomas A. Vida
- Department of Medical Education, Kirk Kerkorian School of Medicine at UNLV, 625 Shadow Lane, Las Vegas, NV 89106, USA; (J.R.); (N.G.); (S.K.); (A.N.-T.); (R.S.)
| |
Collapse
|
30
|
Xiao J, Mukherji R, Sidarous G, Suguru S, Noel M, Weinberg BA, He A, Agarwal S. Longitudinal Circulating Tumor Cell Collection, Culture, and Characterization in Pancreatic Adenocarcinomas. Cancers (Basel) 2025; 17:355. [PMID: 39941724 PMCID: PMC11815863 DOI: 10.3390/cancers17030355] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2024] [Revised: 01/20/2025] [Accepted: 01/21/2025] [Indexed: 02/16/2025] Open
Abstract
BACKGROUND/OBJECTIVES Pancreatic adenocarcinoma (PDAC) remains one of the most lethal cancers, with limited advancements in treatment efficacy due to high rates of chemoresistance. Circulating tumor cells (CTCs) derived from liquid biopsies offer a non-invasive approach to monitoring tumor evolution and identifying molecular mechanisms of resistance. This study aims to longitudinally collect, culture, and characterize CTCs from PDAC patients to elucidate resistance mechanisms and tumor-specific gene expression profiles. METHODS Blood samples from 10 PDAC patients were collected across different treatment stages, yielding 16 CTC cultures. Differential gene expression, pathway dysregulation, and protein-protein interaction studies were utilized, highlighting patient-specific and disease progression-associated changes. Longitudinal comparisons within five patients provided further insights into dynamic molecular changes associated with therapeutic resistance. RESULTS CTC cultures exhibited the activation of key pathways implicated in PDAC progression and resistance, including TNFα/NF-kB, hedgehog signaling, and the epithelial-to-mesenchymal transition. Longitudinal samples revealed dynamic changes in signaling pathways, highlighting upregulated mechanisms of chemoresistance, including PI3K/Akt/mTOR and TGF-β pathways. Additionally, protein-protein interaction analysis emphasized the role of the immune system in PDAC progression and therapy response. Patient-specific gene expression patterns therefore suggest potential applications for precision medicine. CONCLUSIONS This proof-of-concept study demonstrates the feasibility of longitudinally capturing and analyzing CTCs from PDAC patients. The findings provide critical insights into molecular drivers of chemoresistance and highlight the potential of CTC profiling to inform personalized therapeutic strategies. Future large-scale studies are warranted to validate these findings and further explore CTC-based approaches in PDAC management.
Collapse
Affiliation(s)
- Jerry Xiao
- Department of Tumor Biology, Georgetown University, Washington, DC 20057, USA
- Department of Internal Medicine, University of California San Francisco, San Francisco, CA 94115, USA
| | - Reetu Mukherji
- Department of Hematology/Oncology, Medstar Georgetown University Hospital, Washington, DC 20007, USA
| | - George Sidarous
- Department of Internal Medicine, Medstar Georgetown University Hospital, Washington, DC 20007, USA
| | - Shravanthy Suguru
- Department of Pathology, Georgetown University, Washington, DC 20057, USA
| | - Marcus Noel
- Department of Hematology/Oncology, Medstar Georgetown University Hospital, Washington, DC 20007, USA
| | - Benjamin A. Weinberg
- Department of Hematology/Oncology, Medstar Georgetown University Hospital, Washington, DC 20007, USA
| | - Aiwu He
- Department of Hematology/Oncology, Medstar Georgetown University Hospital, Washington, DC 20007, USA
| | - Seema Agarwal
- Department of Pathology, Georgetown University, Washington, DC 20057, USA
| |
Collapse
|
31
|
Hakala S, Hämäläinen A, Sandelin S, Giannareas N, Närvä E. Detection of Cancer Stem Cells from Patient Samples. Cells 2025; 14:148. [PMID: 39851576 PMCID: PMC11764358 DOI: 10.3390/cells14020148] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2024] [Revised: 01/16/2025] [Accepted: 01/17/2025] [Indexed: 01/26/2025] Open
Abstract
The existence of cancer stem cells (CSCs) in various tumors has become increasingly clear in addition to their prominent role in therapy resistance, metastasis, and recurrence. For early diagnosis, disease progression monitoring, and targeting, there is a high demand for clinical-grade methods for quantitative measurement of CSCs from patient samples. Despite years of active research, standard measurement of CSCs has not yet reached clinical settings, especially in the case of solid tumors. This is because detecting this plastic heterogeneous population of cells is not straightforward. This review summarizes various techniques, highlighting their benefits and limitations in detecting CSCs from patient samples. In addition, methods designed to detect CSCs based on secreted and niche-associated signaling factors are reviewed. Spatial and single-cell methods for analyzing patient tumor tissues and noninvasive techniques such as liquid biopsy and in vivo imaging are discussed. Additionally, methods recently established in laboratories, preclinical studies, and clinical assays are covered. Finally, we discuss the characteristics of an ideal method as we look toward the future.
Collapse
Affiliation(s)
| | | | | | | | - Elisa Närvä
- Institute of Biomedicine and FICAN West Cancer Centre Laboratory, University of Turku and Turku University Hospital, FI-20520 Turku, Finland; (S.H.); (A.H.); (S.S.); (N.G.)
| |
Collapse
|
32
|
Khandelwal D, Bhattacharya A, Kumari V, Gupta SS, Ranjan KR, Mishra V. Leveraging nanomaterials for ultrasensitive biosensors in early cancer detection: a review. J Mater Chem B 2025; 13:802-820. [PMID: 39635753 DOI: 10.1039/d4tb02107j] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/07/2024]
Abstract
Cancer remains a major global health challenge with a high mortality rate, as evidenced by the rise in new cases every year. Conventional diagnostic methods like PET scans, MRIs, and biopsies, despite being widely used, suffer from significant drawbacks such as high radiation exposure, difficulty in distinguishing malignant from benign tumors, and invasiveness. Early detection, which is crucial for improving treatment outcomes and survival rates, is hindered by the asymptomatic nature of early-stage cancer and the limitations of current diagnostic tools. Cancer biomarkers, detectable in body fluids, offer valuable diagnostic information, and recent advances in nanotechnology have led to the development of highly sensitive nano-biosensors. This review explores recent advancements (2022-2024) in the field of ultrasensitive nano-biosensors, emphasizing the strategic integration of nanomaterials to enhance sensitivity and accuracy in cancer biomarker detection. It highlights how precise nanomaterial positioning in sensor components like electrodes and bioreceptors enables early cancer diagnosis at low biomarker concentrations. These innovations underscore the transformative potential of nanomaterials in revolutionizing early cancer diagnostics, improving patient care, and enhancing survival outcomes.
Collapse
Affiliation(s)
- Drishti Khandelwal
- Amity Institute of Click Chemistry Research and Studies, Amity University Uttar Pradesh, Noida, UP-201313, India.
| | - Aheli Bhattacharya
- Department of Chemistry, Amity Institute of Applied Sciences, Amity University Uttar Pradesh, Noida, UP-201313, India.
| | - Vanshika Kumari
- Department of Chemistry, Amity Institute of Applied Sciences, Amity University Uttar Pradesh, Noida, UP-201313, India.
| | | | - Kumar Rakesh Ranjan
- Department of Chemistry, Amity Institute of Applied Sciences, Amity University Uttar Pradesh, Noida, UP-201313, India.
| | - Vivek Mishra
- Amity Institute of Click Chemistry Research and Studies, Amity University Uttar Pradesh, Noida, UP-201313, India.
| |
Collapse
|
33
|
Cox M, Vitello DJ, Chawla A. The Current Role of Circulating Tumor DNA in the Management of Pancreatic Cancer. J Gastrointest Cancer 2025; 56:44. [PMID: 39808248 DOI: 10.1007/s12029-024-01129-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/23/2024] [Indexed: 01/16/2025]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is projected to be the second leading cause of cancer-related death by 2030. Early identification is rare, with a 5-year overall survival (OS) of less than 10%. Advances in the understanding of PDAC tumor biology are needed to improve these outcomes. Circulating tumor DNA (ctDNA) represents a promising novel biomarker in the identification and management of PDAC. Drawn from peripheral blood and analyzed using a variety of techniques, the detection of ctDNA in PDAC has been associated with shorter OS, minimal residual disease presence, and shorter recurrence-free survival. The use of ctDNA has also been examined as an indicator of therapeutic resistance, susceptibility to targeted therapy, and therapeutic response. While promising, ctDNA analysis is limited by its low rates of detection in some settings and lack of predictive ability in others. Many studies examining the utility of ctDNA for the management of PDAC have been relatively small retrospective cohort studies. The current findings will need to be validated by incorporation of ctDNA analysis into cancer registries and larger prospective studies. Given the current, rapid evolution in the field, it is possible that with time, ctDNA will be more routinely incorporated into the clinical management of PDAC.
Collapse
Affiliation(s)
- Madison Cox
- Division of Surgical Oncology, Department of Surgery, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
- Northwestern Medicine Cancer Centers, Northwestern Medicine Regional Medical Group, Winfield, IL, USA
| | - Dominic J Vitello
- Division of Surgical Oncology, Department of Surgery, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
- Northwestern Quality Improvement, Research and Education in Surgery, Department of Surgery, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Akhil Chawla
- Division of Surgical Oncology, Department of Surgery, Northwestern University Feinberg School of Medicine, Chicago, IL, USA.
- Robert H. Lurie Comprehensive Cancer Center of Northwestern University, Chicago, IL, USA.
- Northwestern Medicine Cancer Centers, Northwestern Medicine Regional Medical Group, Winfield, IL, USA.
- Northwestern Quality Improvement, Research and Education in Surgery, Department of Surgery, Northwestern University Feinberg School of Medicine, Chicago, IL, USA.
| |
Collapse
|
34
|
Vasu S, Johnson V, M A, Reddy KA, Sukumar UK. Circulating Extracellular Vesicles as Promising Biomarkers for Precession Diagnostics: A Perspective on Lung Cancer. ACS Biomater Sci Eng 2025; 11:95-134. [PMID: 39636879 DOI: 10.1021/acsbiomaterials.4c01323] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/07/2024]
Abstract
Extracellular vesicles (EVs) have emerged as promising biomarkers in liquid biopsy, owing to their ubiquitous presence in bodily fluids and their ability to carry disease-related cargo. Recognizing their significance in disease diagnosis and treatment, substantial efforts have been dedicated to developing efficient methods for EV isolation, detection, and analysis. EVs, heterogeneous membrane-encapsulated vesicles secreted by all cells, contain bioactive substances capable of modulating recipient cell biology upon internalization, including proteins, lipids, DNA, and various RNAs. Their prevalence across bodily fluids has positioned them as pivotal mediators in physiological and pathological processes, notably in cancer, where they hold potential as straightforward tumor biomarkers. This review offers a comprehensive examination of advanced nanotechnology-based techniques for detecting lung cancer through EV analysis. It begins by providing a brief overview of exosomes and their role in lung cancer progression. Furthermore, this review explores the evolving landscape of EV isolation and cargo analysis, highlighting the importance of characterizing specific biomolecular signatures within EVs for improved diagnostic accuracy in lung cancer patients. Innovative strategies for enhancing the sensitivity and specificity of EV isolation and detection, including the integration of microfluidic platforms and multiplexed biosensing technologies are summarized. The discussion then extends to key challenges associated with EV-based liquid biopsies, such as the standardization of isolation and detection protocols and the establishment of robust analytical platforms for clinical translation. This review highlights the transformative impact of EV-based liquid biopsy in lung cancer diagnosis, heralding a new era of personalized medicine and improved patient care.
Collapse
Affiliation(s)
- Sunil Vasu
- Department of Chemical Engineering, Indian Institute of Technology Tirupati, Tirupati, Andhra Pradesh, India-517 619
| | - Vinith Johnson
- Department of Chemical Engineering, Indian Institute of Technology Tirupati, Tirupati, Andhra Pradesh, India-517 619
| | - Archana M
- Department of Chemical Engineering, Indian Institute of Technology Tirupati, Tirupati, Andhra Pradesh, India-517 619
| | - K Anki Reddy
- Department of Chemical Engineering, Indian Institute of Technology Tirupati, Tirupati, Andhra Pradesh, India-517 619
| | - Uday Kumar Sukumar
- Department of Chemical Engineering, Indian Institute of Technology Tirupati, Tirupati, Andhra Pradesh, India-517 619
| |
Collapse
|
35
|
Xu Y, Yan X, Zhuang J, Hao H. The Anticancer Perspective of Tangeretin: A Small Review. Molecules 2025; 30:300. [PMID: 39860170 PMCID: PMC11767889 DOI: 10.3390/molecules30020300] [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: 11/28/2024] [Revised: 12/29/2024] [Accepted: 01/10/2025] [Indexed: 01/27/2025] Open
Abstract
Cancer is an important disease that threatens human life and health. Many natural compounds from plants have been found to have a better inhibitory effect on cancer, and flavonoids are one of them. Tangeretin, a flavonoid, is widely present in a variety of citrus plants and has been shown to have a variety of biological activities that can inhibit tumor cells. Tangeretin can inhibit the growth, proliferation, and metastasis of cancer cells by acting on JAK/STAT (Janus Kinase/signal transducer and activator of transcription) and caspase-3 signal transduction and by regulating the cell cycle of tumor cells. Tangeretin can also work with other chemotherapy drugs, such as cisplatin, to reduce the drug resistance of cancer cells and improve the therapeutic effect of chemotherapy drugs. This review summarizes the effects of tangeretin on various cancers.
Collapse
Affiliation(s)
- Yuan Xu
- Department of Organic Chemistry, College of Chemistry, Beijing University of Chemical Technology, Beijing 100029, China; (Y.X.); (J.Z.)
| | - Xi Yan
- College of Chemistry, Beijing Normal University, Beijing 100875, China
| | - Junpeng Zhuang
- Department of Organic Chemistry, College of Chemistry, Beijing University of Chemical Technology, Beijing 100029, China; (Y.X.); (J.Z.)
| | - Haijun Hao
- Department of Organic Chemistry, College of Chemistry, Beijing University of Chemical Technology, Beijing 100029, China; (Y.X.); (J.Z.)
| |
Collapse
|
36
|
Medina JE, Annapragada AV, Lof P, Short S, Bartolomucci AL, Mathios D, Koul S, Niknafs N, Noë M, Foda ZH, Bruhm DC, Hruban C, Vulpescu NA, Jung E, Dua R, Canzoniero JV, Cristiano S, Adleff V, Symecko H, van den Broek D, Sokoll LJ, Baylin SB, Press MF, Slamon DJ, Konecny GE, Therkildsen C, Carvalho B, Meijer GA, Andersen CL, Domchek SM, Drapkin R, Scharpf RB, Phallen J, Lok CA, Velculescu VE. Early Detection of Ovarian Cancer Using Cell-Free DNA Fragmentomes and Protein Biomarkers. Cancer Discov 2025; 15:105-118. [PMID: 39345137 PMCID: PMC11726017 DOI: 10.1158/2159-8290.cd-24-0393] [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: 03/18/2024] [Revised: 06/14/2024] [Accepted: 09/25/2024] [Indexed: 10/01/2024]
Abstract
SIGNIFICANCE There is an unmet need for effective ovarian cancer screening and diagnostic approaches that enable earlier-stage cancer detection and increased overall survival. We have developed a high-performing accessible approach that evaluates cfDNA fragmentomes and protein biomarkers to detect ovarian cancer.
Collapse
Affiliation(s)
- Jamie E. Medina
- The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Akshaya V. Annapragada
- The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Pien Lof
- Department of Gynecologic Oncology, Centre of Gynecologic Oncology Amsterdam, Antoni van Leeuwenhoek Hospital–The Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Sarah Short
- The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Adrianna L. Bartolomucci
- The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Dimitrios Mathios
- The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Shashikant Koul
- The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Noushin Niknafs
- The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Michaël Noë
- The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Zachariah H. Foda
- The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Daniel C. Bruhm
- The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Carolyn Hruban
- The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Nicholas A. Vulpescu
- The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Euihye Jung
- Penn Ovarian Cancer Research Center, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania
| | - Renu Dua
- The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Jenna V. Canzoniero
- The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Stephen Cristiano
- The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Vilmos Adleff
- The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Heather Symecko
- Basser Center for BRCA, Abramson Cancer Center, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Daan van den Broek
- Department of Laboratory Medicine, Antoni van Leeuwenhoek Hospital–The Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Lori J. Sokoll
- The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Stephen B. Baylin
- The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Michael F. Press
- Department of Pathology, Keck School of Medicine, University of Southern California, Los Angeles, California
| | - Dennis J. Slamon
- David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California
| | - Gottfried E. Konecny
- David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California
| | | | - Beatriz Carvalho
- Department of Pathology, Antoni van Leeuwenhoek Hospital–The Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Gerrit A. Meijer
- Department of Pathology, Antoni van Leeuwenhoek Hospital–The Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Claus Lindbjerg Andersen
- Department of Molecular Medicine, Aarhus University Hospital, Aarhus, Denmark
- Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
| | - Susan M. Domchek
- Penn Ovarian Cancer Research Center, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania
- Basser Center for BRCA, Abramson Cancer Center, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Ronny Drapkin
- Penn Ovarian Cancer Research Center, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania
- Basser Center for BRCA, Abramson Cancer Center, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Robert B. Scharpf
- The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Jillian Phallen
- The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Christine A.R. Lok
- Department of Gynecologic Oncology, Centre of Gynecologic Oncology Amsterdam, Antoni van Leeuwenhoek Hospital–The Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Victor E. Velculescu
- The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, Maryland
| |
Collapse
|
37
|
Li L, Zhao J, Wang Y, Zhang Z, Chen W, Wang J, Cai Y. Integration of machine learning and experimental validation to identify the prognostic signature related to diverse programmed cell deaths in breast cancer. Front Oncol 2025; 14:1505934. [PMID: 39834939 PMCID: PMC11744720 DOI: 10.3389/fonc.2024.1505934] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2024] [Accepted: 12/10/2024] [Indexed: 01/22/2025] Open
Abstract
Background Programmed cell death (PCD) is closely related to the occurrence, development, and treatment of breast cancer. The aim of this study was to investigate the association between various programmed cell death patterns and the prognosis of breast cancer (BRCA) patients. Methods The levels of 19 different programmed cell deaths in breast cancer were assessed by ssGSEA analysis, and these PCD scores were summed to obtain the PCDS for each sample. The relationship of PCDS with immune as well as metabolism-related pathways was explored. PCD-associated subtypes were obtained by unsupervised consensus clustering analysis, and differentially expressed genes between subtypes were analyzed. The prognostic signature (PCDRS) were constructed by the best combination of 101 machine learning algorithm combinations, and the C-index of PCDRS was compared with 30 published signatures. In addition, we analyzed PCDRS in relation to immune as well as therapeutic responses. The distribution of genes in different cells was explored by single-cell analysis and spatial transcriptome analysis. Potential drugs targeting key genes were analyzed by Cmap. Finally, the expression levels of key genes in clinical tissues were verified by RT-PCR. Results PCDS showed higher levels in cancer compared to normal. Different PCDS groups showed significant differences in immune and metabolism-related pathways. PCDRS, consisting of seven key genes, showed robust predictive ability over other signatures in different datasets. The high PCDRS group had a poorer prognosis and was strongly associated with a cancer-promoting tumor microenvironment. The low PCDRS group exhibited higher levels of anti-cancer immunity and responded better to immune checkpoint inhibitors as well as chemotherapy-related drugs. Clofibrate and imatinib could serve as potential small-molecule complexes targeting SLC7A5 and BCL2A1, respectively. The mRNA expression levels of seven genes were upregulated in clinical cancer tissues. Conclusion PCDRS can be used as a biomarker to assess the prognosis and treatment response of BRCA patients, which offers novel insights for prognostic monitoring and treatment personalization of BRCA patients.
Collapse
Affiliation(s)
- Longpeng Li
- Department of Anesthesiology, Shanxi Province Cancer Hospital/Shanxi Hospital Affiliated to Cancer Hospital, Chinese Academy of Medical Sciences/Cancer Hospital Affiliated to Shanxi Medical University, Taiyuan, China
- Institute of Physical Education and Sport, Shanxi University, Taiyuan, China
| | - Jinfeng Zhao
- Institute of Physical Education and Sport, Shanxi University, Taiyuan, China
| | - Yaxin Wang
- Institute of Physical Education and Sport, Shanxi University, Taiyuan, China
| | - Zhibin Zhang
- Institute of Physical Education and Sport, Shanxi University, Taiyuan, China
| | - Wanquan Chen
- Institute of Physical Education and Sport, Shanxi University, Taiyuan, China
| | - Jirui Wang
- Institute of Physical Education and Sport, Shanxi University, Taiyuan, China
| | - Yue Cai
- Department of Anesthesiology, Shanxi Province Cancer Hospital/Shanxi Hospital Affiliated to Cancer Hospital, Chinese Academy of Medical Sciences/Cancer Hospital Affiliated to Shanxi Medical University, Taiyuan, China
| |
Collapse
|
38
|
Kedar P, Saraf A, Maheshwari R, Sharma M. Advances in Dendritic Systems and Dendronized Nanoparticles: Paradigm Shifts in Cancer Targeted Therapy and Diagnostics. Mol Pharm 2025; 22:28-57. [PMID: 39707984 DOI: 10.1021/acs.molpharmaceut.4c00856] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2024]
Abstract
Cancer has emerged as a global health crisis, claiming millions of lives annually. Dendrimers and dendronized nanoparticles, a novel class of nanoscale molecules with highly branched three-dimensional macromolecular structures, have gained significant attention in cancer treatment and diagnosis due to their unique properties. These dendritic macromolecules offer a precisely controlled branching architecture, enabling functionalization with specific targeting molecules to enhance the selective delivery of therapeutic agents to tumor cells while minimizing systemic toxicity. Through surface modifications and the incorporation of various components, dendrimers demonstrate remarkable adaptability as nanocarriers for biomedical imaging and theranostic applications. Surface functionalization strategies, including PEGylation and ligand attachment (e.g., folic acid, RGD peptide, lactobionic acid), further enhance biocompatibility and facilitate targeted tumor cell imaging. Leveraging their improved biocompatibility and target specificity, dendritic nanosystems offer heightened sensitivity and precision in cancer diagnostics. Notably, the encapsulation of metal nanoparticles within dendrimers, such as gold nanoparticles, has shown promise in enhancing tumor imaging capabilities. Ongoing advancements in nanotechnology are poised to increase the sophistication and complexity of dendrimer-based systems, highlighting their potential as nanocarriers in drug delivery platforms, with a growing number of clinical trials on the horizon. This review provides a comprehensive overview of the potential and future prospects of dendrimers and dendrimer-based nanocarriers in targeted cancer therapy and diagnosis, exploring their ability to enhance biocompatibility, reduce toxicity, and improve therapeutic outcomes across various malignancies.
Collapse
Affiliation(s)
- Pawan Kedar
- School of Pharmacy and Technology Management, SVKM's Narsee Monjee Institute of Management Studies (NMIMS), Deemed to be University, Shirpur, Dhule, Maharashtra 425405, India
| | - Apeksha Saraf
- School of Pharmacy, Devi Ahilya Vishwavidyalaya, Takshashila Campus, Khandwa Road, Indore, Madhya Pradesh 452001, India
| | - Rahul Maheshwari
- School of Pharmacy and Technology Management, SVKM's Narsee Monjee Institute of Management Studies (NMIMS), Deemed to be University, Hyderabad 509301, India
| | - Mayank Sharma
- School of Pharmacy and Technology Management, SVKM's Narsee Monjee Institute of Management Studies (NMIMS), Deemed to be University, Shirpur, Dhule, Maharashtra 425405, India
| |
Collapse
|
39
|
Takeda K, Gereg C, Liu X, Ma W, Bearse M, Tang H, Delfino I, Huang E, Lin X, Chandler JB, Wang H. Higher sensitivity of pericardial fluid cytology than biopsy in malignant effusions with potential explanation of false-negative cytology: A multi-institutional analysis. Cytopathology 2025; 36:31-40. [PMID: 39301772 DOI: 10.1111/cyt.13447] [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: 05/22/2024] [Revised: 08/27/2024] [Accepted: 09/06/2024] [Indexed: 09/22/2024]
Abstract
OBJECTIVE Malignant pericardial effusions are associated with a poor prognosis. Pericardial fluid cytology and pericardial biopsy are the primary methods for diagnosis. This study aimed to conduct a multi-institutional analysis to compare the diagnostic sensitivity of cytology and biopsy, and to investigate potential explanations for false-negative results in cytology. METHODS A retrospective review of pericardial fluid cytology cases with concurrent biopsy was conducted across four different institutions. Results were compared using standard statistical methods with attention to sensitivity and histologic distribution. False-negative cytology cases were investigated for further exploration. RESULTS A total of 309 cases were collected, of which 99 (32.0%) were confirmed malignant through repeat sampling or clinical history. Pericardial fluid cytology and biopsy identified 84 and 64 malignant cases, respectively. Our findings confirmed significantly higher sensitivity of cytology compared to biopsy (84.8% vs 65.7%). The most common sites of origin were lung, breast, and gastrointestinal, with adenocarcinoma being the most prevalent histologic subtype. Histologic review of 12 false-negative cytology cases revealed three key explanations; lymphoma was the most common missed diagnosis (33.3%); fibrinous pericarditis obscures neoplastic cells on the pericardial surface; and pericardial involvement can be seen without extension into the pericardial space. CONCLUSION This study demonstrated diagnostic superiority of pericardial fluid cytology over biopsy in the evaluation of malignant pericardial effusions. We identified several limitations in fluid cytology causing false negatives. In the context of an underlying malignancy with pericardial effusion, pathologists should consider immunohistochemistry studies to aid on the diagnosis.
Collapse
Affiliation(s)
- Kotaro Takeda
- Department of Pathology and Laboratory Medicine, Yale New Haven Hospital, Yale University, New Haven, Connecticut, USA
| | - Catherine Gereg
- Department of Pathology and Laboratory Medicine, Yale New Haven Hospital, Yale University, New Haven, Connecticut, USA
| | - Xiaoying Liu
- Department of Pathology and Laboratory Medicine, Dartmouth-Hitchcock Medical Center, Lebanon, New Hampshire, USA
| | - Weijie Ma
- Department of Pathology and Laboratory Medicine, Dartmouth-Hitchcock Medical Center, Lebanon, New Hampshire, USA
| | - Mayara Bearse
- Department of Pathology and Laboratory Medicine, Yale New Haven Hospital, Yale University, New Haven, Connecticut, USA
| | - Haiming Tang
- Department of Pathology and Laboratory Medicine, Yale New Haven Hospital, Yale University, New Haven, Connecticut, USA
| | | | - Eric Huang
- Department of Pathology, University of Washington, Seattle, Washington, USA
| | - Xiaoqi Lin
- Department of Pathology, Northwestern Memorial Hospital, Northwestern University, Chicago, Illinois, USA
| | - Jocelyn B Chandler
- Department of Pathology and Laboratory Medicine, Yale New Haven Hospital, Yale University, New Haven, Connecticut, USA
| | - He Wang
- Department of Pathology and Laboratory Medicine, Yale New Haven Hospital, Yale University, New Haven, Connecticut, USA
| |
Collapse
|
40
|
Hu Y, Liao Y, Pan S, Zhou J, Wan C, Huang F, Bai Y, Lin C, Xia Q, Liu Z, Gong J, Nie X, Wang M, Qin R. A Triple-Mismatch Differentiating assay exploiting activation and trans cleavage of CRISPR-Cas12a for mutation detection with ultra specificity and sensitivity. Biosens Bioelectron 2025; 267:116826. [PMID: 39369517 DOI: 10.1016/j.bios.2024.116826] [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] [Revised: 09/22/2024] [Accepted: 09/30/2024] [Indexed: 10/08/2024]
Abstract
Liquid biopsy technology is non-invasive and convenient, and is currently an emerging technology for cancer screening. Among them, clustered regularly interspaced short palindromic repeats (CRISPR)-CRISPR associated protein 12a (Cas12a) based nucleic acid detection technology has the advantages of high sensitivity, rapidity, and easy operation. However, CRISPR-Cas12a does not discriminate single-base mismatches of targets well enough to meet the needs of clinical detection. Herein, we developed the Triple-Mismatch Differentiating (TMD) assay. This assay amplified the small thermodynamic difference in mismatches at one site at the level of CRISPR-Cas12a activation to a significant thermodynamic difference at three sites at both the level of CRISPR-Cas12a activation and trans-cleavage, which greatly improves the ability of CRISPR-Cas12a to discriminate between base mismatches. Our manipulation greatly improved the specificity of the CRISPR-Cas12a system while maintaining its inherent sensitivity and simplicity, increasing the detection limit to 0.0001%. When testing samples from pancreatic cancer patients, our results were highly consistent with NGS sequencing results. We believe that the TMD assay will provide a new technology for early cancer detection and will be widely used in the clinical practice.
Collapse
Affiliation(s)
- Yibo Hu
- Department of Biliary-Pancreatic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, China
| | - Yangwei Liao
- Department of Biliary-Pancreatic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, China
| | - Shutao Pan
- Department of Biliary-Pancreatic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, China
| | - Jingcong Zhou
- Department of Biliary-Pancreatic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, China
| | - Changqing Wan
- Department of Biliary-Pancreatic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, China
| | - Feiyang Huang
- Department of Biliary-Pancreatic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, China
| | - Yu Bai
- Department of Biliary-Pancreatic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, China
| | - Chen Lin
- Department of Biliary-Pancreatic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, China
| | - Qilong Xia
- Department of Biliary-Pancreatic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, China
| | - Zixi Liu
- Department of Biliary-Pancreatic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, China
| | - Jun Gong
- Department of Biliary-Pancreatic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, China.
| | - Xiaoqi Nie
- Department of Dermatology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China.
| | - Min Wang
- Department of Biliary-Pancreatic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, China.
| | - Renyi Qin
- Department of Biliary-Pancreatic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, China.
| |
Collapse
|
41
|
Fagery M, Khorshidi HA, Wong SQ, Karanfil Ö, Emery J, IJzerman MJ. Integrating Multi-Cancer Early Detection (MCED) Tests with Standard Cancer Screening: System Dynamics Model Development and Feasibility Testing. PHARMACOECONOMICS - OPEN 2025; 9:147-160. [PMID: 39424759 PMCID: PMC11717771 DOI: 10.1007/s41669-024-00533-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 09/25/2024] [Indexed: 10/21/2024]
Abstract
BACKGROUND Cancer screening plays a critical role in early disease detection and improving outcomes. In Australia, established screening protocols for colorectal, breast and cervical cancer have significantly contributed to timely cancer detection. However, the recent introduction of multi-cancer early detection (MCED) tests arguably can disrupt current screening, yet the extent to which these tests provide additional benefits remains uncertain. We present the development and initial validation of a system dynamics (SD) model that estimates the additional cancer detections and costs associated with MCED tests. AIM This article describes the development of a simulation model built to evaluate the additional patient diagnoses and the economic impact of incorporating MCED testing alongside Australia's well-established standard of care (SOC) screening programs for colorectal, breast, cervical and lung cancers. The model was designed to estimate the additional number of patients diagnosed at each cancer stage (stage I, II, III, IV, or unknown) and the associated costs. This simulation model allows for the analysis of multiple scenarios under a plausible set of assumptions regarding population-level participation rates. METHODS An SD model was developed to represent the existing SOC national cancer screening pathways and to integrate potential clinical pathways that could be introduced by MCED tests. The SD model was built to investigate three scenarios for the use of MCED testing: firstly, to explore the viability of MCED testing as a substitute among individuals who are not opting for SOC screening for any reason; secondly, to implement MCED testing exclusively for individuals ineligible for SOC screening, yet have high-risk characteristics; and thirdly, to employ MCED testing after SOC screening to serve as a triaging/confirmatory tool for individuals receiving inconclusive test results. The three primary scenarios were constructed by varying diagnostic accuracy and uptake rates of MCED tests. DISCUSSION The clinical utility and outcomes of MCED testing for screening and early detection still lack comprehensive evidence. Nonetheless, this simulation model facilitates a thorough analysis of MCED tests within the Australian healthcare context, providing insights into potential additional detections and costs to the healthcare system, which may help prioritise future evidence development. The adaptable yet novel SD model presented herein is anticipated to be of considerable interest to industry, policymakers, consumers and clinicians involved in informing clinical and economic decisions regarding integrating MCED tests as cancer screening and early detection tools. The expected results of applying this SD model will determine whether using MCED testing in conjunction with SOC screening offers any potential benefits, possibly guiding policy decisions and clinical practices towards the adoption of MCED tests.
Collapse
Affiliation(s)
- Mussab Fagery
- Cancer Health Services Research, Centre for Cancer Research, Faculty of Medicine, Dentistry and Health Sciences, The University of Melbourne, Melbourne, Australia.
- Cancer Health Services Research, Centre for Health Policy, Melbourne School of Population and Global Health, Faculty of Medicine, Dentistry and Health Sciences, The University of Melbourne, Melbourne, Australia.
| | - Hadi A Khorshidi
- Cancer Health Services Research, Centre for Cancer Research, Faculty of Medicine, Dentistry and Health Sciences, The University of Melbourne, Melbourne, Australia
- Cancer Health Services Research, Centre for Health Policy, Melbourne School of Population and Global Health, Faculty of Medicine, Dentistry and Health Sciences, The University of Melbourne, Melbourne, Australia
| | - Stephen Q Wong
- Peter MacCallum Cancer Centre, Melbourne, Australia
- Department of Oncology, Sir Peter MacCallum, University of Melbourne, Melbourne, Australia
| | - Özge Karanfil
- College of Administrative Sciences and Economics and School of Medicine, Koç University, Istanbul, Turkey
- MIT Sloan School of Management, Boston, MA, USA
| | - Jon Emery
- Department of General Practice and Primary Care, Centre for Cancer Research, University of Melbourne, Melbourne, Australia
| | - Maarten J IJzerman
- Cancer Health Services Research, Centre for Cancer Research, Faculty of Medicine, Dentistry and Health Sciences, The University of Melbourne, Melbourne, Australia
- Cancer Health Services Research, Centre for Health Policy, Melbourne School of Population and Global Health, Faculty of Medicine, Dentistry and Health Sciences, The University of Melbourne, Melbourne, Australia
- Erasmus School of Health Policy and Management, Erasmus University Rotterdam, Rotterdam, the Netherlands
| |
Collapse
|
42
|
Okunlola FO, Adetuyi TG, Olajide PA, Okunlola AR, Adetuyi BO, Adeyemo-Eleyode VO, Akomolafe AA, Yunana N, Baba F, Nwachukwu KC, Oyewole OA, Adetunji CO, Shittu OB, Ginikanwa EG. Biomedical image characterization and radio genomics using machine learning techniques. MINING BIOMEDICAL TEXT, IMAGES AND VISUAL FEATURES FOR INFORMATION RETRIEVAL 2025:397-421. [DOI: 10.1016/b978-0-443-15452-2.00019-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
|
43
|
Sharma H, Yadav V, Burchett A, Shi T, Senapati S, Datta M, Chang HC. A Mem-dELISA platform for dual color and ultrasensitive digital detection of colocalized proteins on extracellular vesicles. Biosens Bioelectron 2025; 267:116848. [PMID: 39413723 PMCID: PMC11543507 DOI: 10.1016/j.bios.2024.116848] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2024] [Revised: 07/29/2024] [Accepted: 10/09/2024] [Indexed: 10/18/2024]
Abstract
Accurate, multiplex, and ultrasensitive measurement of different colocalized protein markers on individual tumor-derived extracellular vesicles (EVs) and dimerized proteins with multiple epitopes could provide insights into cancer heterogeneity, therapy management and early diagnostics that cannot be extracted from bulk methods. However, current digital protein assays lack certain features to enable robust colocalization, including multi-color detection capability, large dynamic range, and selectivity against background proteins. Here, we report a lithography-free, inexpensive (< $0.1) and ultrasensitive dual-color Membrane Digital ELISA (Mem-dELISA) platform by using track-etched polycarbonate (PCTE) membranes to overcome these shortcomings. Their through-pores remove air bubbles through wicking before they are sealed on one side by adhesion to form microwells. Immunomagnetic bead-analyte complexes and substrate solution are then loaded into the microwells from the opposite side, with >80% loading efficiency, before sealing with oil. This enables duplex digital protein colorimetric assay with beta galactosidase and alkaline phosphatase enzymes. The platform achieves 5 logs of dynamic range with a limit of detection of 10 aM for both Biotinylated β-galactosidase (B-βG) and Biotin Alkaline Phosphatase Conjugated (B-ALP) proteins. We demonstrate its potential by showing that a higher dosage of paclitaxel suppresses EpCAM-positive EVs but not GPC-1 positive EVs from breast cancer cells, a decline in chemo-resistance that cannot be detected with Western blot analysis of cell lysate. The Mem-dELISA is poised to empower researchers to conduct ultrasensitive, high throughput protein colocalization studies for disease diagnostics, treatment monitoring and biomarker discovery.
Collapse
Affiliation(s)
- Himani Sharma
- Department of Chemical and Biomolecular Engineering, University of Notre Dame, Notre Dame, IN, 46556, USA
| | - Vivek Yadav
- Department of Chemical and Biomolecular Engineering, University of Notre Dame, Notre Dame, IN, 46556, USA
| | - Alice Burchett
- Department of Aerospace and Mechanical Engineering, University of Notre Dame, Notre Dame, IN, 46556, USA
| | - Tiger Shi
- Department of Chemical and Biomolecular Engineering, University of Notre Dame, Notre Dame, IN, 46556, USA
| | - Satyajyoti Senapati
- Department of Chemical and Biomolecular Engineering, University of Notre Dame, Notre Dame, IN, 46556, USA
| | - Meenal Datta
- Department of Aerospace and Mechanical Engineering, University of Notre Dame, Notre Dame, IN, 46556, USA
| | - Hsueh-Chia Chang
- Department of Chemical and Biomolecular Engineering, University of Notre Dame, Notre Dame, IN, 46556, USA; Department of Aerospace and Mechanical Engineering, University of Notre Dame, Notre Dame, IN, 46556, USA.
| |
Collapse
|
44
|
Thien Nguyen CV, Hanh Nguyen TH, Vo DH, Vi Van TT, Huong Nguyen GT, Tran TH, Nguyen TH, Khoa Huynh LA, Nguyen TD, Tran NH, Thi Ha TM, Quynh Le PT, Truong XL, Nguyen HDL, Tran UV, Hoang TQ, Nguyen VB, Le VC, Nguyen XC, Phuong Nguyen TM, Nguyen VH, Nhat Tran NT, Quynh Dang TN, Tran MH, Nguyen PN, Dao TH, Phuc Nguyen HT, Tran NT, Phan TV, Nguyen DS, Tang HS, Giang H, Phan MD, Nguyen HN, Tran LS. Evaluation of a multimodal ctDNA-based assay for detection of aggressive cancers lacking standard screening tests. Future Oncol 2025; 21:105-115. [PMID: 39431470 PMCID: PMC11975059 DOI: 10.1080/14796694.2024.2413266] [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: 08/10/2024] [Accepted: 10/03/2024] [Indexed: 10/22/2024] Open
Abstract
Aim: Cancers lacking standard screening (LSS) options account for approximately 70% of cancer-related deaths due to late-stage diagnosis. Circulating tumor DNA (ctDNA) is a promising biomarker for multi-cancer early detection. We previously developed SPOT-MAS, a multimodal ctDNA-based assay analyzing methylation and fragmentomic profiles, effective in detecting common cancers (breast, colorectal, liver, lung and gastric). This study extends the analysis to five LSS cancers: endometrial, esophageal, head and neck, ovarian and pancreatic.Methods: SPOT-MAS was applied to profile cfDNA methylation and fragmentomic patterns in 739 healthy individuals and 135 LSS cancer patients.Results: We identified 347 differentially methylated regions and observed genome-wide hypomethylation across all five LSS cancers. Esophageal and head and neck cancers showed an enrichment of short cfDNA fragments (<150 bp). Eleven 4-mer end motifs were consistently altered in cfDNA fragments across all LSS cancers. Many significant signatures were consistent with previous observations in common cancers. Notably, SPOT-MAS achieved 96.2% specificity and 74.8% overall sensitivity, with a lower sensitivity of 60.7% in early-stage cancers.Conclusion: This proof-of-concept study demonstrates that SPOT-MAS a non-invasive test trained on five common cancer types, could detect a number of LSS cancer cases, potentially complementing existing screening programs.
Collapse
Affiliation(s)
| | | | - Dac Ho Vo
- Medical Genetics Institute, Ho Chi Minh, Vietnam
| | | | | | | | | | | | | | | | - Thi Minh Thi Ha
- Department of Medical Genetics, University of Medicine & Pharmacy, Hue University, Vietnam
| | - Phan Tuong Quynh Le
- Department of Medical Genetics, University of Medicine & Pharmacy, Hue University, Vietnam
| | - Xuan Long Truong
- Department of Internal Medicine, University of Medicine & Pharmacy, Hue University, Vietnam
| | | | - Uyen Vu Tran
- Medical Genetics Institute, Ho Chi Minh, Vietnam
| | | | | | | | | | | | | | | | | | | | | | | | | | | | - Thi Van Phan
- Medical Genetics Institute, Ho Chi Minh, Vietnam
| | | | | | - Hoa Giang
- Medical Genetics Institute, Ho Chi Minh, Vietnam
| | | | | | - Le Son Tran
- Medical Genetics Institute, Ho Chi Minh, Vietnam
| |
Collapse
|
45
|
Ahmed NM, Eddama MMR, Beatson K, Gurung R, Patel J, Iskandar G, Abdel-Salam A, Al-Omar A, Cohen R, Abdel-Aziz T, Clapp L. Circulating large extracellular vesicles as diagnostic biomarkers of indeterminate thyroid nodules: multi-platform omics analysis. BJS Open 2024; 9:zrae139. [PMID: 39787026 PMCID: PMC11683363 DOI: 10.1093/bjsopen/zrae139] [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: 07/08/2024] [Revised: 10/02/2024] [Accepted: 10/08/2024] [Indexed: 01/12/2025] Open
Abstract
BACKGROUND While most thyroid nodules are benign, 7-15% are malignant. Patients with indeterminate thyroid nodules (specifically Bethesda IV/Thy3f) often undergo diagnostic hemithyroidectomy to reach a diagnosis on final histology. The aim of this study was to assess the feasibility of circulating large extracellular vesicles as diagnostic biomarkers in patients presenting with Thy3f thyroid nodules. METHODS This was a two-gate diagnostic accuracy study; patients with Thy3f thyroid nodules were age, sex and body mass index matched to healthy individuals. Final histology confirmed benign and malignant diagnoses. Plasma large extracellular vesicle counts were quantified using flow cytometry. Large extracellular vesicle microRNA and protein profiles were identified using next generation sequencing and mass spectrometry, respectively. RESULTS A total of 42 patients with Thy3f nodules (22 with cancer, 20 with non-cancer diagnosis) and 16 healthy controls were included. Total large extracellular vesicle concentrations and the concentrations of extracellular vesicles expressing epithelial cell adhesion molecule and the cancer markers atypical chemokine receptor type 7, extracellular matrix metalloproteinase inducer and syndecan-4 were significantly higher in patients with Thy3f nodules (cancer and non-cancer) compared with healthy individuals. In patients with cancerous versus non-cancer Thy3f nodules, one microRNA was upregulated: mir-195-3p (P < 0.001). Five were downregulated: mir-3176 (P < 0.001), mir-205-5p (P < 0.001), novel-hsa-mir-208-3p (P < 0.001), mir-3529-3p (P = 0.01) and let-7i-3p (P = 0.02). Furthermore, three large extracellular vesicle proteins (kallikrein-related peptidase11 (KLK11) (P = 0.001), alpha-1-acid glycoprotein 2 (A1AG2) (P <0.001) and small integral membrane protein 1 (SMIM1) (P = 0.04)) were significantly upregulated, while 20 large extracellular vesicle proteins were significantly downregulated (most downregulated: chemokine (C-X-C motif) ligand 7 (CXCL7), tubulin beta chain 1 (TBB1), binding immunoglobulin protein (BIP) and actinin alpha 1 (ACTN1) (P < 0.001)) in cancerous compared with non-cancer Thy3f nodules. CONCLUSION Circulating large extracellular vesicle miRNA and protein profiles have a high diagnostic value to discriminate between benign and malignant nodules for patients with Thy3f cytology. Further validation for clinical performance will be needed.
Collapse
Affiliation(s)
- Nada M Ahmed
- Institute of Cardiovascular Sciences, University College London, London, UK
- Pathology Department, Alexandria University, Alexandria, Egypt
| | - Mohammad M R Eddama
- Department of Surgical Biotechnology, Division of Surgery and Interventional Science, University College London, London, UK
| | - Kevin Beatson
- Department of Surgical Biotechnology, Division of Surgery and Interventional Science, University College London, London, UK
| | - Rijan Gurung
- Department of Surgical Biotechnology, Division of Surgery and Interventional Science, University College London, London, UK
| | - Jigisha Patel
- Department of Surgical Biotechnology, Division of Surgery and Interventional Science, University College London, London, UK
| | - Georges Iskandar
- Department of Anaesthesia and Perioperative Medicine, University College London Hospitals, London, UK
| | - Alaa Abdel-Salam
- Endocrine Surgery Unit, University College London Hospitals, London, UK
| | - Abdullah Al-Omar
- Endocrine Surgery Unit, University College London Hospitals, London, UK
| | - Richard Cohen
- Department of Surgical Biotechnology, Division of Surgery and Interventional Science, University College London, London, UK
| | - Tarek Abdel-Aziz
- Endocrine Surgery Unit, University College London Hospitals, London, UK
| | - Lucie Clapp
- Institute of Cardiovascular Sciences, University College London, London, UK
| |
Collapse
|
46
|
Al Qadire M, Abdelrahman H, Al Sabei S, Aljezawi M, Al Omari O, Al Salmi N, Al Awaisi H, Aljezawi H. Prevalence and Predictors of the Unmet Supportive Needs of Patients With Cancer in Oman. Cancer Nurs 2024:00002820-990000000-00308. [PMID: 39689231 DOI: 10.1097/ncc.0000000000001413] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2024]
Abstract
BACKGROUND Unmet supportive needs are directly correlated with more frequent psychological distress, reduced quality of life, and low patient satisfaction with healthcare and indirectly connected with low compliance with treatment and increased care-related costs. OBJECTIVE To assess the spectrum of unmet needs among patients with cancer in Oman and identify predictors of these needs and their relationship with psychological distress and symptom burden. METHOD A descriptive, correlational study design was used, involving 551 patients with cancer from 2 major healthcare facilities in Muscat, Oman. Data on patients' unmet needs were collected between January and June 2023 using the Supportive Care Needs Survey Short-Form 34. RESULTS Participants had a mean age of 45.8 (SD, 15.6) years, with female patients comprising 65.5% of the sample. Breast cancer was the most prevalent type of cancer. The mean unmet need score was 31.0/100 (SD, 20.1). Statistical analysis revealed significant predictors of unmet needs, including marital status, treatment type, performance status, symptom interference, and psychological distress, with the regression model explaining 26% of the variance in unmet needs. CONCLUSIONS Healthcare providers must regularly assess supportive needs, recognizing that they may vary across populations and are influenced by cultural factors. Additionally, individuals identified as having characteristics that predict higher levels of need should receive focused and prioritized supportive care. IMPLICATIONS FOR PRACTICE By adopting regular tailored assessments that address the comprehensive supportive needs of patients with cancer, clinicians can significantly enhance patients' quality of life and optimize treatment outcomes.
Collapse
Affiliation(s)
- Mohammad Al Qadire
- Author Affiliations: College of Nursing, Sultan Qaboos University, Muscat, Sultanate of Oman (Drs Al Qadire, Abdelrahman, Al Sabei, Al Omari, and Al Salmi); Faculty of Nursing, Al Al-Bayt University, Mafraq, Jordan (Drs Al Qadire and Aljezawi); Faculty of Nursing, Suez Canal University, Ismailia, Egypt (Dr Abdelrahman); University Medical City, Sultan Qaboos Comprehensive Cancer Center, Muscat, Sultanate of Oman (Dr Al Awaisi); and Al Mafraq Governmental Hospital, Mafraq, Jordan (Mrs Aljezawi)
| | | | | | | | | | | | | | | |
Collapse
|
47
|
Meyer ML, Hirsch FR, Bunn PA, Ujhazy P, Fredrickson D, Berg CD, Carbone DP, Halmos B, Singh H, Borghaei H, Ferris A, Langer C, Dacic S, Mok TS, Peters S, Johnson BE. Calls to action on lung cancer management and research. Oncologist 2024; 29:e1634-e1645. [PMID: 39002167 PMCID: PMC11630765 DOI: 10.1093/oncolo/oyae169] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2024] [Accepted: 05/24/2024] [Indexed: 07/15/2024] Open
Abstract
Lung cancer, the leading cause of cancer-related deaths globally, remains a pressing health issue despite significant medical advances. The New York Lung Cancer Foundation brought together experts from academia, the pharmaceutical and biotech industries as well as organizational leaders and patient advocates, to thoroughly examine the current state of lung cancer diagnosis, treatment, and research. The goal was to identify areas where our understanding is incomplete and to develop collaborative public health and scientific strategies to generate better patient outcomes, as highlighted in our "Calls to Action." The consortium prioritized 8 different calls to action. These include (1) develop strategies to cure more patients with early-stage lung cancer, (2) investigate carcinogenesis leading to lung cancers in patients without a history of smoking, (3) harness precision medicine for disease interception and prevention, (4) implement solutions to deliver prevention measures and effective therapies to individuals in under-resourced countries, (5) facilitate collaborations with industry to collect and share data and samples, (6) create and maintain open access to big data repositories, (7) develop new immunotherapeutic agents for lung cancer treatment and prevention, and (8) invest in research in both the academic and community settings. These calls to action provide guidance to representatives from academia, the pharmaceutical and biotech industries, organizational and regulatory leaders, and patient advocates to guide ongoing and planned initiatives.
Collapse
Affiliation(s)
- May-Lucie Meyer
- Hematology and Oncology Department, Tisch Cancer Institute at Mount Sinai, Icahn School of Medicine and Thoracic Oncology Center, New York, NY, United States
| | - Fred R Hirsch
- Hematology and Oncology Department, Tisch Cancer Institute at Mount Sinai, Icahn School of Medicine and Thoracic Oncology Center, New York, NY, United States
| | - Paul A Bunn
- Division of Medical Oncology, University of Colorado School of Medicine, Aurora, CO, United States
| | - Peter Ujhazy
- Translational Research Program, Division of Cancer Treatment and Diagnosis, National Cancer Institute, Rockville, MD, United States
| | | | | | - David P Carbone
- Division of Medical Oncology, The Ohio State University—James Comprehensive Cancer Center, Columbus, OH, United States
| | - Balazs Halmos
- Department of Oncology, Montefiore Medical Center, Albert Einstein College of Medicine, Bronx, NY, United States
| | - Harpreet Singh
- US Food and Drug Administration (FDA), Washington, DC, United States
| | | | | | - Corey Langer
- Abramson Cancer Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - Sanja Dacic
- Department of Pathology, Yale School of Medicine, New Haven, CT, United States
| | - Tony S Mok
- State Laboratory of Translational Oncology, The Chinese University of Hong Kong, Hong Kong, People’s Republic of China
| | - Solange Peters
- Department of Oncology, University Hospital CHUV, Lausanne, Switzerland
| | - Bruce E Johnson
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, United States
| |
Collapse
|
48
|
Smilkou S, Kaklamanis L, Balgouranidou I, Linardou H, Papatheodoridi AM, Zagouri F, Razis E, Kakolyris S, Psyrri A, Papadimitriou C, Markou A, Lianidou E. Direct comparison of an ultrasensitive real-time PCR assay with droplet digital PCR for the detection of PIK3CA hotspot mutations in primary tumors, plasma cell-free DNA and paired CTC-derived gDNAs. Front Oncol 2024; 14:1435559. [PMID: 39711963 PMCID: PMC11659196 DOI: 10.3389/fonc.2024.1435559] [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: 05/20/2024] [Accepted: 10/28/2024] [Indexed: 12/24/2024] Open
Abstract
Introduction Detection of PIK3CA mutations in primary tumors and liquid biopsy samples is of increasing importance for treatment decisions and therapy resistance in many types of cancer. The aim of the present study was to directly compare the efficacy of a relatively inexpensive ultrasensitive real-time PCR with the well-established and highly sensitive technology of ddPCR for the detection of the three most common hotspot mutations of PIK3CA, in exons 9 and 20, that are all of clinical importance in various types of cancer. Patients and methods We analyzed 42 gDNAs from primary tumors (FFPEs), 29 plasma-cfDNA samples, and 29 paired CTC-derived gDNAs, all from patients with ER+ metastatic breast cancer, and plasma from 10 healthy donors. The same blood draws were used for CTC isolation using EpCAM beads for positive immunomagnetic enrichment. All FFPEs and plasma-cfDNA samples were analyzed in parallel for PIK3CA mutations by ultrasensitive real-time PCR assay and droplet digital PCR. Results In gDNAs from FFPEs, using ultrasensitive real-time PCR, the p.E545K mutation was detected in 22/42(52.4%), and the p.E542K and p.H1047R mutations were detected in 14/42(33.3%) and 16/42(38.1%), respectively. Using ddPCR, the p.E545K mutation was detected in 22/42(52.4%), p.E542K in 17/42(40.5%), and p.H1047R in 19/42(45.2%) samples, revealing a concordance between the two methodologies of 81%, 78.6% and 78.6% for each mutation respectively. In plasma-cfDNA, using ultrasensitive real-time PCR, the p.E545K mutation was detected in 11/29(38%) and both p.E542K and p.H1047R mutations in 2/29(6.9%).In the same plasma-cfDNA samples using ddPCR, p.E545K was detected in 1/29(3.5%), p.E542K in 2/29(6.9%), and p.H1047R in 3/29(10.5%) samples, revealing a concordance of 65.5%,100% and 93.1% for each mutation respectively. In paired CTC-derived gDNAs p.E545K was detected in 11/29(38%), p.E542K in 3/29(10.3%), and p.H1047R in 7/29(24.1%) samples. Conclusions This low-cost, high-throughput and ultrasensitive real-time PCR assay provides accurate and specific detection of PIK3CA hotspot mutations in liquid biopsy samples and gives similar results to ddPCR. This assay can be performed in labs where digital PCR instrumentation is not available. In CTC-derived gDNA and paired plasma-cfDNA, PIK3CA mutations detected were not identical, revealing that CTC and plasma-cfDNA give complementary information.
Collapse
Affiliation(s)
- Stavroula Smilkou
- Analysis of Circulating Tumor Cells, Laboratory of Analytical Chemistry, Department of Chemistry, University of Athens, Athens, Greece
| | - Loukas Kaklamanis
- Department of Pathology, Onassis Cardiac Surgery Center, Athens, Greece
| | - Ioanna Balgouranidou
- Department of Medical Oncology, University General Hospital of Alexandroupolis, Alexandroupolis, Greece
| | | | - Alkistis Maria Papatheodoridi
- Department of Clinical Therapeutics, School of Medicine, Alexandra Hospital, National and Kapodistrian University of Athens, Athens, Greece
| | - Flora Zagouri
- Department of Clinical Therapeutics, School of Medicine, Alexandra Hospital, National and Kapodistrian University of Athens, Athens, Greece
| | - Evangelia Razis
- Third Department of Medical Oncology, Hygeia Hospital, Athens, Greece
| | - Stylianos Kakolyris
- Department of Medical Oncology, University General Hospital of Alexandroupolis, Alexandroupolis, Greece
| | - Amanda Psyrri
- Section of Medical Oncology, Department of Internal Medicine, Faculty of Medicine, Attikon University Hospital, Athens, Greece
| | - Christos Papadimitriou
- Oncology Unit, 2nd Department of Surgery, School of Medicine, Aretaieio Hospital, National and Kapodistrian University of Athens, Athens, Greece
| | - Athina Markou
- Analysis of Circulating Tumor Cells, Laboratory of Analytical Chemistry, Department of Chemistry, University of Athens, Athens, Greece
| | - Evi Lianidou
- Analysis of Circulating Tumor Cells, Laboratory of Analytical Chemistry, Department of Chemistry, University of Athens, Athens, Greece
| |
Collapse
|
49
|
Martínez-Espinosa I, Serrato JA, Cabello-Gutiérrez C, Carlos-Reyes Á, Ortiz-Quintero B. Exosome-Derived miRNAs in Liquid Biopsy for Lung Cancer. Life (Basel) 2024; 14:1608. [PMID: 39768316 PMCID: PMC11678223 DOI: 10.3390/life14121608] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2024] [Revised: 12/01/2024] [Accepted: 12/03/2024] [Indexed: 01/11/2025] Open
Abstract
Exosome-derived microRNAs (miRNAs) are potential biomarkers for lung cancer detection and monitoring through liquid biopsy. These small, non-coding RNA molecules are found within exosomes, which are extracellular vesicles released from cells. Their stability in biofluids, such as blood, positions them as candidates for minimally invasive diagnostics. Multiple studies have shown that lung cancer patients exhibit distinct miRNA profiles compared to healthy individuals. This finding suggests that exosome-derived miRNAs could serve as valuable biomarkers for diagnosis, prognosis, and evaluating therapeutic responses. This review summarizes recent research on exosome-derived miRNAs in liquid biopsies, including blood, pleural effusion, and pleural lavage, as biomarkers for lung cancer, focusing on publications from the last five years.
Collapse
Affiliation(s)
- Israel Martínez-Espinosa
- Department of Molecular Biomedicine and Translational Research, Instituto Nacional de Enfermedades Respiratorias Ismael Cosío Villegas, Mexico City 14080, Mexico
| | - José A. Serrato
- Department of Molecular Biomedicine and Translational Research, Instituto Nacional de Enfermedades Respiratorias Ismael Cosío Villegas, Mexico City 14080, Mexico
| | - Carlos Cabello-Gutiérrez
- Department of Research in Virology and Mycology, Instituto Nacional de Enfermedades Respiratorias Ismael Cosío Villegas, Mexico City 14080, Mexico
| | - Ángeles Carlos-Reyes
- Laboratory of Onco-Immunobiology, Instituto Nacional de Enfermedades Respiratorias Ismael Cosío Villegas, Mexico City 14080, Mexico
| | - Blanca Ortiz-Quintero
- Department of Molecular Biomedicine and Translational Research, Instituto Nacional de Enfermedades Respiratorias Ismael Cosío Villegas, Mexico City 14080, Mexico
| |
Collapse
|
50
|
Ahuja S, Zaheer S. The evolution of cancer immunotherapy: a comprehensive review of its history and current perspectives. KOREAN JOURNAL OF CLINICAL ONCOLOGY 2024; 20:51-73. [PMID: 39778508 PMCID: PMC11717579 DOI: 10.14216/kjco.24009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/19/2024] [Revised: 10/24/2024] [Accepted: 11/19/2024] [Indexed: 01/11/2025]
Abstract
Cancer immunotherapy uses the body's immune system to combat cancer, marking a significant advancement in treatment. This review traces its evolution from the late 19th century to its current status. It began with William Coley's pioneering work using bacterial toxins to stimulate the immune system against cancer cells, establishing the foundational concept of immunotherapy. In the mid-20th century, cytokine therapies like interferons and interleukins emerged, demonstrating that altering the immune response could reduce tumors and highlighting the complex interplay between cancer and the immune system. The discovery of immune checkpoints, regulatory pathways that prevent autoimmunity but are exploited by cancer cells to evade detection, was a pivotal development. Another major breakthrough is CAR-T cell therapy, which involves modifying a patient's T cells to target cancer-specific antigens. This personalized treatment has shown remarkable success in certain blood cancers. Additionally, cancer vaccines aim to trigger immune responses against tumor-specific or associated antigens, and while challenging, ongoing research is improving their efficacy. The historical progression of cancer immunotherapy, from Coley's toxins to modern innovations like checkpoint inhibitors and CAR-T cell therapy, underscores its transformative impact on cancer treatment. As research delves deeper into the immune system's complexities, immunotherapy is poised to become even more crucial in oncology, offering renewed hope to patients globally.
Collapse
Affiliation(s)
- Sana Ahuja
- Department of Pathology, Safdarjung Hospital, Vardhman Mahavir Medical College, New Delhi, India
| | - Sufian Zaheer
- Department of Pathology, Safdarjung Hospital, Vardhman Mahavir Medical College, New Delhi, India
| |
Collapse
|