1
|
Aredo JV, Jamali A, Zhu J, Heater N, Wakelee HA, Vaklavas C, Anagnostou V, Lu J. Liquid Biopsy Approaches for Cancer Characterization, Residual Disease Detection, and Therapy Monitoring. Am Soc Clin Oncol Educ Book 2025; 45:e481114. [PMID: 40305739 DOI: 10.1200/edbk-25-481114] [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: 05/02/2025]
Abstract
Liquid biopsy encompasses a variety of molecular approaches to detect circulating tumor DNA (ctDNA) and has become a powerful tool in the diagnosis and treatment of solid tumors. Current applications include comprehensive genomic profiling for identifying targetable mutations and therapeutic resistance mechanisms, with emerging applications in minimal residual disease detection and treatment response monitoring. Increasingly, the potential for liquid biopsy in guiding treatment decisions is under active investigation through prospective clinical trials using ctDNA-adaptive interventions in patients with early-stage and metastatic cancers. Limitations arise on the basis of the sensitivity and feasibility of individual liquid biopsy assays; nonetheless, emerging technologies set the stage for improving these shortcomings. As the global oncology community continues to ascertain the clinical value of liquid biopsy across the continuum of patient care, this minimally invasive approach heralds a significant advancement in the promise of precision oncology.
Collapse
Affiliation(s)
- Jacqueline V Aredo
- Division of Oncology, Department of Medicine, Stanford Cancer Institute, Stanford University School of Medicine, Stanford, CA
| | - Amna Jamali
- The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD
- The Johns Hopkins Molecular Tumor Board, Johns Hopkins School of Medicine, Baltimore, MD
| | - Jessica Zhu
- Huntsman Cancer Institute, University of Utah, Salt Lake City, UT
| | - Natalie Heater
- Division of Hematology and Oncology, Department of Medicine, Robert H. Lurie Comprehensive Cancer Center, Northwestern University Feinberg School of Medicine, Chicago, IL
| | - Heather A Wakelee
- Division of Oncology, Department of Medicine, Stanford Cancer Institute, Stanford University School of Medicine, Stanford, CA
| | | | - Valsamo Anagnostou
- The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD
- The Johns Hopkins Molecular Tumor Board, Johns Hopkins School of Medicine, Baltimore, MD
- Lung Cancer Precision Medicine Center of Excellence, Johns Hopkins University School of Medicine, Baltimore, MD
- The Bloomberg∼Kimmel Institute for Cancer Immunotherapy, Johns Hopkins University School of Medicine, Baltimore, MD
| | - Janice Lu
- Division of Hematology and Oncology, Department of Medicine, Robert H. Lurie Comprehensive Cancer Center, Northwestern University Feinberg School of Medicine, Chicago, IL
- Circulating Tumor Cell (CTC) Core Facility, Robert H. Lurie Comprehensive Cancer Center, Northwestern University, Chicago, IL
| |
Collapse
|
2
|
Borea R, Reduzzi C. The growing field of liquid biopsy and its Snowball effect on reshaping cancer management. THE JOURNAL OF LIQUID BIOPSY 2025; 8:100293. [PMID: 40255897 PMCID: PMC12008596 DOI: 10.1016/j.jlb.2025.100293] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 03/13/2025] [Revised: 03/24/2025] [Accepted: 03/24/2025] [Indexed: 04/22/2025]
Abstract
Liquid biopsy (LB) has emerged as a transformative tool in oncology, providing a minimally invasive approach for tumor detection, molecular characterization, and real-time treatment monitoring. By analyzing circulating tumor DNA (ctDNA), circulating tumor cells (CTCs), extracellular vesicles (EVs), and microRNA (miRNA), LB enables comprehensive tumor profiling without the need for traditional tissue biopsies. Over the past decade, research in this field has expanded exponentially, leading to the integration of LB into clinical practice for specific cancer types, including lung and breast cancer. In 2024, the Journal of Liquid Biopsy (JLB) published innovative studies exploring the latest advancements in LB technologies, biomarkers, and their applications for cancer detection, minimal residual disease (MRD) monitoring, and therapy response assessment. This review synthesizes recent findings on the role of LB in cancer treatment and monitoring across different biomarkers, with a particular focus on newly published studies and their context within translational research. Additionally, it highlights emerging techniques such as fragmentomics, artificial intelligence, and multiomics, paving the way for more precise, personalized treatment decisions. Despite these advancements, challenges remain in standardizing methodologies, optimizing clinical validation, and integrating LB into routine oncological workflows. This mini-review highlights the evolving landscape of LB research and its potential to revolutionize cancer diagnosis, treatment monitoring, and therapeutic decision-making, ushering in a new era of precision oncology.
Collapse
Affiliation(s)
- Roberto Borea
- Department of Public Health, University Federico II of Naples, Naples, Italy
- Department of Internal Medicine and Medical Sciences (DiMI), School of Medicine, University of Genova, Genova, Italy
| | - Carolina Reduzzi
- Department of Medicine, Weill Cornell Medicine, Englander Institute for Precision Medicine, New York Presbyterian Hospital, New York, NY, 10021, USA
| |
Collapse
|
3
|
Hruban C, Bruhm DC, Chen IM, Koul S, Annapragada AV, Vulpescu NA, Short S, Theile S, Boyapati K, Alipanahi B, Skidmore ZL, Leal A, Cristiano S, Adleff V, Johannsen JS, Scharpf RB, Foda ZH, Phallen J, Velculescu VE. Genome-wide analyses of cell-free DNA for therapeutic monitoring of patients with pancreatic cancer. SCIENCE ADVANCES 2025; 11:eads5002. [PMID: 40397745 DOI: 10.1126/sciadv.ads5002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/15/2024] [Accepted: 04/07/2025] [Indexed: 05/23/2025]
Abstract
Determining response to therapy for patients with pancreatic cancer can be challenging. We evaluated methods for assessing therapeutic response using cell-free DNA (cfDNA) in plasma from patients with metastatic pancreatic cancer in the CheckPAC trial (NCT02866383). Patients were evaluated before and after initiation of therapy using tumor-informed plasma whole-genome sequencing (WGMAF) and tumor-independent genome-wide cfDNA fragmentation profiles and repeat landscapes (ARTEMIS-DELFI). Using WGMAF, molecular responders had a median overall survival (OS) of 319 days compared to 126 days for nonresponders [hazard ratio (HR) = 0.29, 95% confidence interval (CI) = 0.11-0.79, P = 0.011]. For ARTEMIS-DELFI, patients with low scores after therapy initiation had longer median OS than patients with high scores (233 versus 172 days, HR = 0.12, 95% CI = 0.046-0.31, P < 0.0001). We validated ARTEMIS-DELFI in patients with pancreatic cancer in the PACTO trial (NCT02767557). These analyses suggest that noninvasive mutation and fragmentation-based cfDNA approaches can identify therapeutic response of individuals with pancreatic cancer.
Collapse
Affiliation(s)
- Carolyn Hruban
- The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Daniel C Bruhm
- The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Inna M Chen
- Department of Oncology, Copenhagen University Hospital-Herlev and Gentofte, Herlev, Denmark
| | - Shashikant Koul
- The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Akshaya V Annapragada
- The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Nicholas A Vulpescu
- The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Sarah Short
- The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Susann Theile
- Department of Oncology, Copenhagen University Hospital-Herlev and Gentofte, Herlev, Denmark
| | - Kavya Boyapati
- The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | | | | | - Alessandro Leal
- The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Stephen Cristiano
- The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Vilmos Adleff
- The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Julia S Johannsen
- Department of Oncology, Copenhagen University Hospital-Herlev and Gentofte, Herlev, Denmark
- Department of Medicine, Copenhagen University Hospital-Herlev and Gentofte, Herlev, Denmark
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Robert B Scharpf
- The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Zachariah H Foda
- The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Jillian Phallen
- The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Victor E Velculescu
- The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| |
Collapse
|
4
|
Bao P, Wang T, Liu X, Xing S, Ruan H, Ma H, Tao Y, Zhan Q, Belmonte-Reche E, Qin L, Han Z, Mao M, Li M, Lu ZJ. Peak analysis of cell-free RNA finds recurrently protected narrow regions with clinical potential. Genome Biol 2025; 26:119. [PMID: 40340952 PMCID: PMC12060323 DOI: 10.1186/s13059-025-03590-x] [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: 08/18/2023] [Accepted: 04/25/2025] [Indexed: 05/10/2025] Open
Abstract
BACKGROUND Cell-free RNAs (cfRNAs) can be detected in biofluids and have emerged as valuable disease biomarkers. Accurate identification of the fragmented cfRNA signals, especially those originating from pathological cells, is crucial for understanding their biological functions and clinical value. However, many challenges still need to be addressed for their application, including developing specific analysis methods and translating cfRNA fragments with biological support into clinical applications. RESULTS We present cfPeak, a novel method combining statistics and machine learning models to detect the fragmented cfRNA signals effectively. When test in real and artificial cfRNA sequencing (cfRNA-seq) data, cfPeak shows an improved performance compared with other applicable methods. We reveal that narrow cfRNA peaks preferentially overlap with protein binding sites, vesicle-sorting sites, structural sites, and novel small non-coding RNAs (sncRNAs). When applied in clinical cohorts, cfPeak identified cfRNA peaks in patients' plasma that enable cancer detection and are informative of cancer types and metastasis. CONCLUSIONS Our study fills the gap in the current small cfRNA-seq analysis at fragment-scale and builds a bridge to the scientific discovery in cfRNA fragmentomics. We demonstrate the significance of finding low abundant tissue-derived signals in small cfRNA and prove the feasibility for application in liquid biopsy.
Collapse
Affiliation(s)
- Pengfei Bao
- MOE Key Laboratory of Bioinformatics, State Key Lab of Green Biomanufacturing, Center for Synthetic and Systems Biology, School of Life Sciences, Tsinghua University, Beijing, 100084, China
- Institute for Precision Medicine, Tsinghua University, Beijing, 100084, China
- Peking University-Tsinghua University-National Institute of Biological Sciences Joint Graduate Program, School of Life Sciences, Tsinghua University, Beijing, China
| | - Taiwei Wang
- MOE Key Laboratory of Bioinformatics, State Key Lab of Green Biomanufacturing, Center for Synthetic and Systems Biology, School of Life Sciences, Tsinghua University, Beijing, 100084, China
- Department of Rheumatology and Clinical Immunology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100730, China
- National Clinical Research Center for Dermatologic and Immunologic Diseases (Ministry of Science & Technology), MOE Key Laboratory of Rheumatology and Clinical Immunology, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Beijing, 100730, China
- Academy for Advanced Interdisciplinary Studies (AAIS)and, Sciences Joint Graduate Program (PTN) , Peking University, Beijing, China
| | - Xiaofan Liu
- MOE Key Laboratory of Bioinformatics, State Key Lab of Green Biomanufacturing, Center for Synthetic and Systems Biology, School of Life Sciences, Tsinghua University, Beijing, 100084, China
- Institute for Precision Medicine, Tsinghua University, Beijing, 100084, China
| | - Shaozhen Xing
- MOE Key Laboratory of Bioinformatics, State Key Lab of Green Biomanufacturing, Center for Synthetic and Systems Biology, School of Life Sciences, Tsinghua University, Beijing, 100084, China
- Institute for Precision Medicine, Tsinghua University, Beijing, 100084, China
| | - Hanjin Ruan
- Department of Oral and Maxillofacial & Head and Neck Oncology, Beijing Stomatological Hospital, Capital Medical University, Beijing, China
| | - Hongli Ma
- MOE Key Laboratory of Bioinformatics, State Key Lab of Green Biomanufacturing, Center for Synthetic and Systems Biology, School of Life Sciences, Tsinghua University, Beijing, 100084, China
| | - Yuhuan Tao
- MOE Key Laboratory of Bioinformatics, State Key Lab of Green Biomanufacturing, Center for Synthetic and Systems Biology, School of Life Sciences, Tsinghua University, Beijing, 100084, China
- Institute for Precision Medicine, Tsinghua University, Beijing, 100084, China
| | - Qing Zhan
- MOE Key Laboratory of Bioinformatics, State Key Lab of Green Biomanufacturing, Center for Synthetic and Systems Biology, School of Life Sciences, Tsinghua University, Beijing, 100084, China
- Institute for Precision Medicine, Tsinghua University, Beijing, 100084, China
| | - Efres Belmonte-Reche
- Centre for Genomics and Oncological Research (GENYO), Avenida de La Ilustración 114, Granada, 18016, Spain
- Department of Biochemistry and Molecular Biology II, Faculty of Pharmacy, University of Granada, Granada, Spain
- Instituto de Investigación Biosanitaria Ibs.GRANADA, Hospital Virgen de Las Nieves, Granada, Spain
| | - Lizheng Qin
- Department of Oral and Maxillofacial & Head and Neck Oncology, Beijing Stomatological Hospital, Capital Medical University, Beijing, China
| | - Zhengxue Han
- Department of Oral and Maxillofacial & Head and Neck Oncology, Beijing Stomatological Hospital, Capital Medical University, Beijing, China
| | - Minghui Mao
- Department of Oral and Maxillofacial & Head and Neck Oncology, Beijing Stomatological Hospital, Capital Medical University, Beijing, China.
| | - Mengtao Li
- Department of Rheumatology and Clinical Immunology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100730, China.
- National Clinical Research Center for Dermatologic and Immunologic Diseases (Ministry of Science & Technology), MOE Key Laboratory of Rheumatology and Clinical Immunology, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Beijing, 100730, China.
| | - Zhi John Lu
- MOE Key Laboratory of Bioinformatics, State Key Lab of Green Biomanufacturing, Center for Synthetic and Systems Biology, School of Life Sciences, Tsinghua University, Beijing, 100084, China.
- Institute for Precision Medicine, Tsinghua University, Beijing, 100084, China.
- Peking University-Tsinghua University-National Institute of Biological Sciences Joint Graduate Program, School of Life Sciences, Tsinghua University, Beijing, China.
- Academy for Advanced Interdisciplinary Studies (AAIS)and, Sciences Joint Graduate Program (PTN) , Peking University, Beijing, China.
- The Center for Regeneration Aging and Chronic Diseases, School of Basic Medical Sciences, Tsinghua University, Beijing, China.
| |
Collapse
|
5
|
Lee TR, Ahn JM, Lee J, Kim D, Park J, Jeong BH, Oh D, Kim SM, Jung GC, Choi BH, Kwon MJ, Wang M, Salmans M, Carson A, Leatham B, Fathe K, Lee BI, Jung B, Ki CS, Park YS, Cho EH. Integrating Plasma Cell-Free DNA Fragment End Motif and Size with Genomic Features Enables Lung Cancer Detection. Cancer Res 2025; 85:1696-1707. [PMID: 40136052 DOI: 10.1158/0008-5472.can-24-1517] [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: 05/10/2024] [Revised: 07/28/2024] [Accepted: 11/08/2024] [Indexed: 03/27/2025]
Abstract
Early detection of lung cancer is important for improving patient survival rates. Liquid biopsy using whole-genome sequencing of cell-free DNA (cfDNA) offers a promising avenue for lung cancer screening, providing a potential alternative or complementary approach to current screening modalities. Here, we aimed to develop and validate an approach by integrating fragment and genomic features of cfDNA to enhance lung cancer detection accuracy across diverse populations. Deep learning-based classifiers were trained using comprehensive cfDNA fragmentomic features from participants in multi-institutional studies, including a Korean discovery dataset (218 patients with lung cancer and 2,559 controls), a Korean validation dataset (111 patients with lung cancer and 1,136 controls), and an independent Caucasian validation cohort (50 patients with lung cancer and 50 controls). In the discovery dataset, classifiers using fragment end motif by size, a feature that captures both fragment end motif and size profiles, outperformed standalone fragment end motif and fragment size classifiers, achieving an area under the curve (AUC) of 0.917. The ensemble classifier integrating fragment end motif by size and genomic coverage achieved an improved performance, with an AUC of 0.937. This performance extended to the Korean validation dataset and demonstrated ethnic generalizability in the Caucasian validation cohort. Overall, the development of a deep learning-based classifier integrating cfDNA fragmentomic and genomic features in this study highlights the potential for accurate lung cancer detection across diverse populations. Significance: Evaluating fragment-based features and genomic coverage in cell-free DNA offers an accurate lung cancer screening method, promising improvements in early cancer detection and addressing challenges associated with current screening methods.
Collapse
Affiliation(s)
- Tae-Rim Lee
- Genome Research Center, GC Genome, Yongin-si, South Korea
| | - Jin Mo Ahn
- Genome Research Center, GC Genome, Yongin-si, South Korea
| | - Junnam Lee
- Genome Research Center, GC Genome, Yongin-si, South Korea
| | - Dasom Kim
- Genome Research Center, GC Genome, Yongin-si, South Korea
| | - Juntae Park
- Genome Research Center, GC Genome, Yongin-si, South Korea
| | - Byeong-Ho Jeong
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, South Korea
| | - Dongryul Oh
- Department of Radiation Oncology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, South Korea
| | | | | | | | - Min-Jung Kwon
- Department of Laboratory Medicine, Kangbuk Samsung Hospital, Sungkyunkwan University School of Medicine, Seoul, South Korea
| | | | | | | | | | | | | | - Byoungsok Jung
- Genome Research Center, GC Genome, Yongin-si, South Korea
| | - Chang-Seok Ki
- Genome Research Center, GC Genome, Yongin-si, South Korea
| | - Young Sik Park
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, Seoul National University Hospital, Seoul, South Korea
| | - Eun-Hae Cho
- Genome Research Center, GC Genome, Yongin-si, South Korea
| |
Collapse
|
6
|
Nguyen VTC, Vo DH, Tran TT, Tran TT, Nguyen THH, Vo TDH, Van TTV, Vu TL, Lam MQ, Nguyen GTH, Tran TH, Pham NT, Trac QT, Nguyen TH, Phan TV, Dao TH, Nguyen HTP, Nguyen LHD, Nguyen DS, Tang HS, Giang H, Phan MD, Nguyen HN, Tran LS. Cost-effective shallow genome-wide sequencing for profiling plasma cfDNA signatures to enhance lung cancer detection. Future Oncol 2025; 21:1391-1402. [PMID: 40133038 PMCID: PMC12051589 DOI: 10.1080/14796694.2025.2483154] [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: 01/11/2025] [Accepted: 03/19/2025] [Indexed: 03/27/2025] Open
Abstract
BACKGROUND Lung cancer (LC) screening via low-dose computed tomography (LDCT) faces challenges including high false-positive rates and low patient compliance. Circulating tumor DNA (ctDNA)-based tests offer a minimally invasive alternative but are limited by high costs and low sensitivity, particularly in early-stage detection. This study introduces a cost-effective, shallow genome-wide sequencing approach for LC detection by profiling multiple cell-free DNA (cfDNA) signatures. METHODS We developed a multimodal cfDNA assay with shallow sequencing coverage (0.5×) that integrates fragmentomic, nucleosome, end-motif, and copy number alteration analyses. A machine-learning model trained on a discovery cohort (99 LC patients, 168 healthy controls) and validated on an independent cohort (58 LC patients, 71 controls) demonstrated robust performance. RESULTS The ensemble model exhibited outstanding performance, achieving an AUC of 0.97 and a specificity of 92% in both the discovery and validation cohorts, with sensitivities of 94% and 90%, respectively. Notably, it outperformed hotspot mutation-based assays and the multi-cancer SPOT-MAS assay in sensitivity across all LC stages. CONCLUSIONS This assay provides a cost-effective, accurate, and minimally invasive method for LC detection, addressing the limitations of current screening methods. It represents a promising complementary tool to improve early detection and patient outcomes in LC.
Collapse
Affiliation(s)
- Van Thien Chi Nguyen
- Research and Development Department, Medical Genetics Institute, Ho Chi Minh, Vietnam
| | - Dac Ho Vo
- Research and Development Department, Medical Genetics Institute, Ho Chi Minh, Vietnam
| | - Thi Trang Tran
- Research and Development Department, Medical Genetics Institute, Ho Chi Minh, Vietnam
| | - Thanh Truong Tran
- Research and Development Department, Medical Genetics Institute, Ho Chi Minh, Vietnam
| | - Thi Hue Hanh Nguyen
- Research and Development Department, Medical Genetics Institute, Ho Chi Minh, Vietnam
| | - Truong Dang Huy Vo
- Research and Development Department, Medical Genetics Institute, Ho Chi Minh, Vietnam
| | - Thi Tuong Vi Van
- Research and Development Department, Medical Genetics Institute, Ho Chi Minh, Vietnam
| | - Thi Luyen Vu
- Research and Development Department, Medical Genetics Institute, Ho Chi Minh, Vietnam
| | - Minh Quang Lam
- Research and Development Department, Medical Genetics Institute, Ho Chi Minh, Vietnam
| | | | - Trung Hieu Tran
- Research and Development Department, Medical Genetics Institute, Ho Chi Minh, Vietnam
| | - Ngoc Tan Pham
- Research and Development Department, Medical Genetics Institute, Ho Chi Minh, Vietnam
| | - Quang Thinh Trac
- Research and Development Department, Medical Genetics Institute, Ho Chi Minh, Vietnam
| | - Trong Hieu Nguyen
- Research and Development Department, Medical Genetics Institute, Ho Chi Minh, Vietnam
| | - Thi Van Phan
- Research and Development Department, Medical Genetics Institute, Ho Chi Minh, Vietnam
| | - Thi Huyen Dao
- Research and Development Department, Medical Genetics Institute, Ho Chi Minh, Vietnam
| | - Huu Tam Phuc Nguyen
- Research and Development Department, Medical Genetics Institute, Ho Chi Minh, Vietnam
| | - Luu Hong Dang Nguyen
- Research and Development Department, Medical Genetics Institute, Ho Chi Minh, Vietnam
| | - Duy Sinh Nguyen
- Research and Development Department, Medical Genetics Institute, Ho Chi Minh, Vietnam
| | - Hung Sang Tang
- Research and Development Department, Medical Genetics Institute, Ho Chi Minh, Vietnam
| | - Hoa Giang
- Research and Development Department, Medical Genetics Institute, Ho Chi Minh, Vietnam
| | - Minh Duy Phan
- Research and Development Department, Medical Genetics Institute, Ho Chi Minh, Vietnam
| | - Hoai-Nghia Nguyen
- Research and Development Department, Medical Genetics Institute, Ho Chi Minh, Vietnam
| | - Le Son Tran
- Research and Development Department, Medical Genetics Institute, Ho Chi Minh, Vietnam
| |
Collapse
|
7
|
Mazzilli SA, Rahal Z, Rouhani MJ, Janes SM, Kadara H, Dubinett SM, Spira AE. Translating premalignant biology to accelerate non-small-cell lung cancer interception. Nat Rev Cancer 2025; 25:379-392. [PMID: 39994467 DOI: 10.1038/s41568-025-00791-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 01/02/2025] [Indexed: 02/26/2025]
Abstract
Over the past decade, substantial progress has been made in the development of targeted and immune-based therapies for patients with advanced non-small-cell lung cancer. To further improve outcomes for patients with lung cancer, identifying and intercepting disease at the earliest and most curable stages are crucial next steps. With the recent implementation of low-dose computed tomography scan screening in populations at high risk, there is an emerging unmet need for new diagnostic, prognostic and therapeutic tools to help treat patients suspected of harbouring premalignant lesions and minimally invasive non-small-cell lung cancer. Continued advances in the identification of the earliest drivers of lung carcinogenesis are poised to address these unmet needs. Employing multimodal approaches to chart the temporal and spatial maps of the molecular events driving lung premalignant lesion progression will refine our understanding of early carcinogenesis. Elucidating the molecular drivers of premalignancy is critical to the development of biomarkers to detect those incubating a premalignant lesion, to stratify risk for progression to invasive cancer and to identify novel therapeutic targets to intercept that process. In this Review, we summarize emerging insights into the earliest cellular and molecular events associated with lung squamous and adenocarcinoma carcinogenesis and highlight the growing opportunity for translating these insights into clinical tools for early detection and disease interception to transform the outcomes for those at risk for lung cancer.
Collapse
Affiliation(s)
- Sarah A Mazzilli
- Sectional Computational Biomedicine, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA.
| | - Zahraa Rahal
- Division of Pathology-Lab Medicine, Department of Translational Molecular Pathology, MD Anderson Cancer Center, Houston, TX, USA
| | - Maral J Rouhani
- Lungs for Living Research Centre, UCL Respiratory, University College London, London, UK
| | - Sam M Janes
- Lungs for Living Research Centre, UCL Respiratory, University College London, London, UK
| | - Humam Kadara
- Division of Pathology-Lab Medicine, Department of Translational Molecular Pathology, MD Anderson Cancer Center, Houston, TX, USA
| | - Steven M Dubinett
- Division of Pulmonary and Critical Care, Department of Medicine, David Geffen School of Medicine at University of California, Los Angeles, and Greater Los Angeles Healthcare System, Los Angeles, CA, USA
| | - Avrum E Spira
- Sectional Computational Biomedicine, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA.
- Johnson & Johnson Innovative Medicine, Boston, MA, USA.
| |
Collapse
|
8
|
Bruhm DC, Vulpescu NA, Foda ZH, Phallen J, Scharpf RB, Velculescu VE. Genomic and fragmentomic landscapes of cell-free DNA for early cancer detection. Nat Rev Cancer 2025; 25:341-358. [PMID: 40038442 DOI: 10.1038/s41568-025-00795-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 01/24/2025] [Indexed: 03/06/2025]
Abstract
Genomic analyses of cell-free DNA (cfDNA) in plasma are enabling noninvasive blood-based biomarker approaches to cancer detection and disease monitoring. Current approaches for identification of circulating tumour DNA typically use targeted tumour-specific mutations or methylation analyses. An emerging approach is based on the recognition of altered genome-wide cfDNA fragmentation in patients with cancer. Recent studies have revealed a multitude of characteristics that can affect the compendium of cfDNA fragments across the genome, collectively called the 'cfDNA fragmentome'. These changes result from genomic, epigenomic, transcriptomic and chromatin states of an individual and affect the size, position, coverage, mutation, structural and methylation characteristics of cfDNA. Identifying and monitoring these changes has the potential to improve early detection of cancer, especially using highly sensitive multi-feature machine learning approaches that would be amenable to broad use in populations at increased risk. This Review highlights the rapidly evolving field of genome-wide analyses of cfDNA characteristics, their comparison to existing cfDNA methods, and recent related innovations at the intersection of large-scale sequencing and artificial intelligence. As the breadth of clinical applications of cfDNA fragmentome methods have enormous public health implications for cancer screening and personalized approaches for clinical management of patients with cancer, we outline the challenges and opportunities ahead.
Collapse
Affiliation(s)
- Daniel C Bruhm
- The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Nicholas A Vulpescu
- The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Zachariah H Foda
- The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Jillian Phallen
- The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Robert B Scharpf
- The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Biostatistics, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - Victor E Velculescu
- The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA.
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA.
| |
Collapse
|
9
|
Li S, Geng S, Chen Y, Ren Q, Luan Y, Liang W, Chang Y, Zhang L, Zhu D, Wu W, Zhang Y, Zhang L, Wang Y, Zhong G, Wei B, Ma J, Chang Y, Wang X, Li Z, Duan C, Long G, Mao M. Clinical Validation of a Noninvasive Multi-Omics Method for Multicancer Early Detection in Retrospective and Prospective Cohorts. J Mol Diagn 2025:S1525-1578(25)00106-0. [PMID: 40311780 DOI: 10.1016/j.jmoldx.2025.04.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2024] [Revised: 03/04/2025] [Accepted: 04/04/2025] [Indexed: 05/03/2025] Open
Abstract
Recent studies highlight the promise of blood-based multicancer early detection (MCED) tests for identifying asymptomatic patients with cancer. However, most focus on a single cancer hallmark, thus limiting effectiveness because of cancer's heterogeneity. Here, a blood-based multi-omics test named SeekInCare for MCED is reported. SeekInCare incorporates multiple genomic and epigenetic hallmarks, including copy number aberration, fragment size, end motif, and oncogenic virus, via shallow whole-genome sequencing from cell-free DNA, alongside seven protein tumor markers in one tube of blood. Artificial intelligence algorithms were developed to distinguish patients with cancer from individuals without cancer and to predict the likely affected organ. The retrospective study included 617 patients with cancer and 580 individuals without cancer, covering 27 cancer types. SeekInCare achieved 60.0% sensitivity at 98.3% specificity, resulting in an area under the curve of 0.899. Sensitivities were 37.7%, 50.4%, 66.7%, and 78.1% in patients with stage I, II, III, and IV disease, respectively. Additionally, SeekInCare was evaluated in a prospective cohort consisting of 1203 individuals who received the test as a laboratory-developed test (median follow-up time, 753 days) in which it achieved 70.0% sensitivity at 95.2% specificity. The performances of SeekInCare in both retrospective and prospective studies demonstrate that SeekInCare is a blood-based MCED test, showing comparable performance to the other tests currently in development. These findings support its potential clinical utility as a cancer screening test in high-risk populations.
Collapse
Affiliation(s)
- Shiyong Li
- Research and Development, SeekIn Inc., Shenzhen, China
| | | | - Yan Chen
- Research and Development, SeekIn Inc., Shenzhen, China
| | - Qingqi Ren
- Department of Oncology, Peking University Shenzhen Hospital, Shenzhen, China
| | - Yi Luan
- Department of Oncology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Weijie Liang
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Yinyin Chang
- Clinical Laboratories, Shenyou Bio, Zhengzhou, China
| | - Lijuan Zhang
- Clinical Laboratories, Shenyou Bio, Zhengzhou, China
| | - Dandan Zhu
- Clinical Laboratories, Shenyou Bio, Zhengzhou, China
| | - Wei Wu
- Research and Development, SeekIn Inc., Shenzhen, China
| | - Yingying Zhang
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Linfeng Zhang
- Department of Breast Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Yan Wang
- Department of Gastroenterology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Guolin Zhong
- Research and Development, SeekIn Inc., Shenzhen, China
| | - Bing Wei
- Department of Molecular Pathology, Henan Cancer Hospital, The Affiliated Cancer Hospital of Zhengzhou University, Zhengzhou, China
| | - Jie Ma
- Department of Molecular Pathology, Henan Cancer Hospital, The Affiliated Cancer Hospital of Zhengzhou University, Zhengzhou, China
| | - Yu Chang
- Department of Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Xinhua Wang
- Department of Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Zhiming Li
- Department of Internal Medicine, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Chaohui Duan
- Department of Oncology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Guanghui Long
- Department of Oncology, Peking University Shenzhen Hospital, Shenzhen, China
| | - Mao Mao
- Research and Development, SeekIn Inc., San Diego, California; Yonsei Song-Dang Institute for Cancer Research, Yonsei University, Seoul, Republic of Korea.
| |
Collapse
|
10
|
Zhang S, Tang J, Cui P, He W, Lin X, Wang S, Liu Y, Tan X, Xu S, Feng M, Lai H. Accurate and Efficient Detection of Nasopharyngeal Carcinoma Using Multi-Dimensional Features of Plasma Cell-Free DNA. Head Neck 2025. [PMID: 40256837 DOI: 10.1002/hed.28154] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2024] [Revised: 03/08/2025] [Accepted: 03/23/2025] [Indexed: 04/22/2025] Open
Abstract
BACKGROUND The incidence of Nasopharyngeal carcinoma (NPC) is rising in recent years, especially in some non-developed parts of the world. Hence, cost-efficient means for sensitive detection of NPC are vital. METHODS We recruited 646 participants, including healthy individuals, patients with benign nasopharyngeal diseases, and NPC patients for plasma cell-free DNA(cfDNA), which underwent low-depth whole-genome sequencing (WGS) to extract multi-dimensional molecular features, including fragmentation pattern, end motif, copy number variation(CNV), and transcription factors(TF). Based on these features, we employed a machine learning algorithm to build prediction models for NPC detection. RESULTS We achieved a sensitivity of 95.8% and a specificity of 99.4% to discriminate NPC patients from healthy individuals. CONCLUSIONS This study can be a proof-of-concept for these multi-dimensional molecular features to be implemented as a noninvasive approach for the detection and even early detection of NPC.
Collapse
Affiliation(s)
- Song Zhang
- Department of Otolaryngology, Shenzhen Guangming District People's Hospital, Shenzhen, China
| | - Jiahui Tang
- Department of Otolaryngology, Shenzhen Guangming District People's Hospital, Shenzhen, China
| | - Pin Cui
- Shenzhen Rapha Biotechnology Incorporate, Shenzhen, China
| | - Weihuang He
- Shenzhen Rapha Biotechnology Incorporate, Shenzhen, China
| | - Xiaohui Lin
- Department of Oncology, People's Hospital of Shenzhen Baoan District, The Second Affiliated Hospital of Shenzhen University, Shenzhen, China
| | - Shubing Wang
- Department of Oncology, Shenzhen Key Laboratory of Gastrointestinal Cancer Translational Research, Cancer Institute, Peking University Shenzhen Hospital, Shenzhen-Peking University-Hong Kong University of Science and Technology Medical Center, Shenzhen, China
| | - Yuanxian Liu
- Department of Otolaryngology, Shenzhen Traditional Chinese Medicine Hospital, Shenzhen, China
| | - Xiaohua Tan
- Department of Oncology, Shenzhen Third People's Hospital, Shenzhen, China
| | - Shu Xu
- Department of Oncology, Shenzhen Guangming District People's Hospital, Shenzhen, China
| | - Mingji Feng
- Department of Oncology, Shenzhen Key Laboratory of Gastrointestinal Cancer Translational Research, Cancer Institute, Peking University Shenzhen Hospital, Shenzhen-Peking University-Hong Kong University of Science and Technology Medical Center, Shenzhen, China
| | - Hanming Lai
- Department of Oncology, Shenzhen Key Laboratory of Gastrointestinal Cancer Translational Research, Cancer Institute, Peking University Shenzhen Hospital, Shenzhen-Peking University-Hong Kong University of Science and Technology Medical Center, Shenzhen, China
| |
Collapse
|
11
|
Liu X, Liang C, Ding L, Zhang Q, Liu Y, Wang W. Analysis of the clinical application value of cfDNA methylation and fragmentation in early diagnosis of esophageal cancer. Genomics 2025; 117:111034. [PMID: 40188889 DOI: 10.1016/j.ygeno.2025.111034] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2024] [Revised: 03/02/2025] [Accepted: 03/19/2025] [Indexed: 04/11/2025]
Abstract
BACKGROUND This study explores the clinical value of cfDNA methylation and fragmentation for the early diagnosis of esophageal cancer using liquid biopsy. METHODS Whole genome bisulfite sequencing and low-pass whole genome sequencing were utilized to detect cfDNA biomarkers, comparing 30 esophageal cancer patients with 10 healthy controls. RESULTS Significant differences in cfDNA methylation and fragmentation were observed between cancerous and non-cancerous samples (p < 0.05). A volcano plot identified 822 differentially methylated markers (817 upregulated, 5 downregulated), with SOX17, SOX1, ZNF382, ZNF667-AS1, and TFPI2 highly associated with esophageal cancer. Fragmentation markers (EDM, FSD, FSR, TFBS, CNV) showed 95 % specificity and sensitivity, with EDM demonstrating the best performance. CONCLUSION Our study highlights the clinical potential of cfDNA methylation and fragmentation biomarkers for the early diagnosis of esophageal cancer.
Collapse
Affiliation(s)
- Xin Liu
- Department of Cardiothoracic Surgery, Xiangyang Central Hospital, Hospital Affiliated to Hubei University of Arts and Science, Xiangyang, Hubei, China; Department of Thoracic Surgery, First Hospital of Jilin University, Changchun, China
| | - Chen Liang
- Department of Thoracic Surgery, First Hospital of Jilin University, Changchun, China
| | - Lingwen Ding
- Department of Vaccination clinic, Xiangyang Center for Disease Control and Prevention, Xiangyang, Hubei, China
| | - Qian Zhang
- Department of Cardiothoracic Surgery, Xiangyang Central Hospital, Hospital Affiliated to Hubei University of Arts and Science, Xiangyang, Hubei, China
| | - Yi Liu
- Department of Cardiothoracic Surgery, Xiangyang Central Hospital, Hospital Affiliated to Hubei University of Arts and Science, Xiangyang, Hubei, China
| | - Wei Wang
- Department of Cardiothoracic Surgery, Xiangyang Central Hospital, Hospital Affiliated to Hubei University of Arts and Science, Xiangyang, Hubei, China.
| |
Collapse
|
12
|
Meshkovska Y, Dzhuraeva B, Godugu C, Pooladanda V, Thatikonda S. Deciphering the interplay: circulating cell-free DNA, signaling pathways, and disease progression in idiopathic pulmonary fibrosis. 3 Biotech 2025; 15:102. [PMID: 40165930 PMCID: PMC11954786 DOI: 10.1007/s13205-025-04272-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2024] [Accepted: 03/10/2025] [Indexed: 04/02/2025] Open
Abstract
Idiopathic pulmonary fibrosis (IPF) is a lung disease with an unknown etiology and a short survival rate. There is no accurate method of early diagnosis, and it involves computed tomography (CT) or lung biopsy. Since diagnostic methods are not accurate due to their similarity to other lung pathologies, discovering new biomarkers is a key issue for diagnosticians. Currently, the use of ccf-DNA (circulating cell-free deoxyribonucleic acid) is an important focus due to its association with IPF-induced alterations in metabolic pathways, such as amino acid metabolism, energy metabolism, and lipid metabolism pathways. Other biomarkers associated with metabolic changes have been found, and they are related to changes in type II/type I alveolar epithelial cells (AECs I/II), changes in extracellular matrix (ECM), and inflammatory processes. Currently, IPF pathogenetic treatment remains unknown, and the mortality rates are increasing, and the patients are diagnosed at a late stage. Signaling pathways and metabolic dysfunction have a significant role in the disease occurrence, particularly the transforming growth factor-β (TGF-β) signaling pathway, which plays an essential role. TGF-β, Wnt, Hedgehog (Hh), and integrin signaling are the main drivers of fibrosis. These pathways activate the transformation of fibroblasts into myofibroblasts, extracellular matrix (ECM) deposition, and tissue remodeling fibrosis. Therapy targeting diverse signaling pathways to slow disease progression is crucial in the treatment of IPF. Two antifibrotic medications, including pirfenidone and nintedanib, are Food and Drug Administration (FDA)-approved for treatment. ccf-DNA could become a new biomarker for IPF diagnosis to detect the disease at the early stage, while FDA-approved therapies could help to prevent late conditions from forming and decrease mortality rates.
Collapse
Affiliation(s)
- Yeva Meshkovska
- Department of Head and Neck-Endocrine Oncology, Moffitt Cancer Center, Tampa, FL 33612 USA
| | - Barchinai Dzhuraeva
- Department of Hospital Pediatrics, Moffitt Cancer Center, Tampa, FL 33612 USA
- Department of Hospital Pediatrics with a Course of Neonatology, National Center of Maternal and Child Health, Bishkek, 720017 Kyrgyzstan
| | - Chandraiah Godugu
- Department of Regulatory Toxicology, Biological Sciences, National Institute of Pharmaceutical Education and Research (NIPER), Balanagar, Hyderabad, Telangana 500037 India
| | - Venkatesh Pooladanda
- Vincent Center for Reproductive Biology, Department of Obstetrics and Gynecology, Massachusetts General Hospital, 60 Blossom Street, Thier 9, Boston, MA 02114 USA
- Obstetrics, Gynecology and Reproductive Biology, Harvard Medical School, Boston, MA 02115 USA
| | - Sowjanya Thatikonda
- Department of Head and Neck-Endocrine Oncology, Moffitt Cancer Center, Tampa, FL 33612 USA
| |
Collapse
|
13
|
Hashmi A, Greiner LJ, Chauhan PS, Szymanski JJ, Park S, Olivier K, Owen D, Chaudhuri AA. Emergence of Circulating Tumor DNA as a Precision Biomarker in Lung Cancer Radiation Oncology and Beyond. Hematol Oncol Clin North Am 2025; 39:257-268. [PMID: 39732580 DOI: 10.1016/j.hoc.2024.11.002] [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: 12/30/2024]
Abstract
Circulating tumor DNA (ctDNA) is emerging as a transformative biomarker in the management of non-small cell lung cancer (NSCLC). This review focuses on its role in detecting minimal residual disease (MRD), predicting treatment response, and guiding therapeutic decision-making in radiation oncology and immunotherapy. Key studies demonstrate ctDNA's prognostic value, particularly in identifying relapse risk and refining patient stratification for curative-intent and consolidative treatments. Future research is essential to standardize ctDNA assays, optimize integration into clinical workflows, and expand its clinical utility. This biomarker holds substantial promise by enabling non-invasive, real-time monitoring and improving outcomes for patients with NSCLC and beyond.
Collapse
Affiliation(s)
- Ayesha Hashmi
- Department of Radiation Oncology, Mayo Clinic, 200 First Street SW, Rochester, MN 55905, USA
| | - Lilli J Greiner
- Department of Radiation Oncology, Mayo Clinic, 200 First Street SW, Rochester, MN 55905, USA
| | - Pradeep S Chauhan
- Department of Radiation Oncology, Mayo Clinic, 200 First Street SW, Rochester, MN 55905, USA
| | - Jeffrey J Szymanski
- Department of Radiation Oncology, Mayo Clinic, 200 First Street SW, Rochester, MN 55905, USA
| | - Sean Park
- Department of Radiation Oncology, Mayo Clinic, 200 First Street SW, Rochester, MN 55905, USA
| | - Kenneth Olivier
- Department of Radiation Oncology, Mayo Clinic, 200 First Street SW, Rochester, MN 55905, USA
| | - Dawn Owen
- Department of Radiation Oncology, Mayo Clinic, 200 First Street SW, Rochester, MN 55905, USA
| | - Aadel A Chaudhuri
- Department of Radiation Oncology, Mayo Clinic, 200 First Street SW, Rochester, MN 55905, USA.
| |
Collapse
|
14
|
Parisi FM, Lentini M, Chiesa-Estomba CM, Mayo-Yanez M, Leichen JR, White M, Giurdanella G, Cocuzza S, Bianco MR, Fakhry N, Maniaci A. Liquid Biopsy in HPV-Associated Head and Neck Cancer: A Comprehensive Review. Cancers (Basel) 2025; 17:977. [PMID: 40149311 PMCID: PMC11940600 DOI: 10.3390/cancers17060977] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2025] [Revised: 02/25/2025] [Accepted: 03/11/2025] [Indexed: 03/29/2025] Open
Abstract
Objectives: Head and neck squamous cell carcinoma (HNSCC) is the sixth most common cancer globally, with HPV-positive cases emerging as a distinct subtype with unique clinical and molecular characteristics. Current diagnostic methods, including tissue biopsy and imaging, face limitations in terms of invasiveness, static disease assessment, and difficulty in distinguishing recurrence from treatment-related changes. This review aimed to assess the potential of liquid biopsy as a minimally invasive tool for the diagnosis, treatment monitoring, and surveillance of HPV-associated HNSCC. Methods: This systematic review analyzed literature from PubMed/MEDLINE, Embase, and Web of Science, focusing on original research and reviews related to liquid biopsy applications in HPV-positive HNSCC. Included studies were evaluated based on the robustness of the study design, clinical relevance, and analytical performance of liquid biopsy technologies. Biomarker types, detection methods, and implementation strategies were assessed to identify advancements and challenges in this field. Results: Liquid biopsy technologies, including circulating HPV DNA, ctDNA, and extracellular vesicles, demonstrated high sensitivity (90-95%) and specificity (>98%) in detecting HPV-positive HNSCC. These methods enabled real-time monitoring of tumor dynamics, early detection of recurrence, and insights into treatment resistance. Longitudinal analysis revealed that biomarker clearance during treatment correlates strongly with patient outcomes. Conclusions: Liquid biopsy is a transformative diagnostic and monitoring tool for HPV-associated HNSCC, offering minimally invasive, real-time insights into tumor biology. While challenges remain in standardization and clinical implementation, ongoing research and technological innovations hold promise for integrating liquid biopsy into personalized cancer care, ultimately improving patient outcomes.
Collapse
Affiliation(s)
- Federica Maria Parisi
- Department of Medical and Surgical Sciences and Advanced Technologies “G.F. Ingrassia”, ENT Section, University of Catania, 95125 Catania, Italy; (F.M.P.); (S.C.)
| | - Mario Lentini
- Department of Otolaryngology, ASP 7, Ragusa Hospital, 97100 Ragusa, Italy
| | - Carlos M. Chiesa-Estomba
- Department of Otorhinolaryngology-Head and Neck Surgery, Hospital Universitario Donostia, 20001 San Sebastian, Spain
| | - Miguel Mayo-Yanez
- Otorhinolaryngology-Head and Neck Surgery Department, Complexo Hospitalario Universitario A Coruña (CHUAC), 15006 La Coruña, Spain;
- Otorhinolaryngology-Head and Neck Surgery Department, Hospital San Rafael (HSR) de A Coruña, 15006 La Coruña, Spain
- Otorhinolaryngology-Head and Neck Surgery Research Group, Institute of Biomedical Research of A Coruña, (INIBIC), Complexo Hospitalario Universitario de A Corñna (CHUAC), Universidade da Corñna (UDC), 15494 La Coruña, Spain
| | - Jerome R. Leichen
- Department of Human Anatomy and Experimental Oncology, UMONS Research Institute for Health Sciences and Technology, University of Mons (UMons), 7011 Mons, Belgium;
| | - Matthew White
- Division of Otorhinolaryngology, Head and Neck Surgery, University of Cape Town, Cape Town 8001, South Africa;
| | - Giovanni Giurdanella
- Department of Medicine and Surgery, University of Enna “Kore”, 94100 Enna, Italy;
| | - Salvatore Cocuzza
- Department of Medical and Surgical Sciences and Advanced Technologies “G.F. Ingrassia”, ENT Section, University of Catania, 95125 Catania, Italy; (F.M.P.); (S.C.)
| | - Maria Rita Bianco
- Otolaryngology-Department of Health Science, University of Catanzaro, 88100 Catanzaro, Italy;
| | - Nicolas Fakhry
- Department of Oto-Rhino-Laryngology Head and Neck Surgery, La Conception University Hospital, AP-HM, Aix Marseille Université, 13006 Marseille, France;
| | - Antonino Maniaci
- Department of Medicine and Surgery, University of Enna “Kore”, 94100 Enna, Italy;
| |
Collapse
|
15
|
D'Amiano AJ, Cheunkarndee T, Azoba C, Chen KY, Mak RH, Perni S. Transparency and Representation in Clinical Research Utilizing Artificial Intelligence in Oncology: A Scoping Review. Cancer Med 2025; 14:e70728. [PMID: 40059400 PMCID: PMC11891267 DOI: 10.1002/cam4.70728] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2023] [Revised: 02/12/2025] [Accepted: 02/13/2025] [Indexed: 05/13/2025] Open
Abstract
INTRODUCTION Artificial intelligence (AI) has significant potential to improve health outcomes in oncology. However, as AI utility increases, it is imperative to ensure that these models do not systematize racial and ethnic bias and further perpetuate disparities in health. This scoping review evaluates the transparency of demographic data reporting and diversity of participants included in published clinical studies utilizing AI in oncology. METHODS We utilized PubMed to search for peer-reviewed research articles published between 2016 and 2021 with the query type "("deep learning" or "machine learning" or "neural network" or "artificial intelligence") and ("neoplas$" or "cancer$" or "tumor$" or "tumour$")." We included clinical trials and original research studies and excluded reviews and meta-analyses. Oncology-related studies that described data sets used in training or validation of the AI models were eligible. Data regarding public reporting of patient demographics were collected, including age, sex at birth, and race. We used descriptive statistics to analyze these data across studies. RESULTS Out of 220 total studies, 118 were eligible and 47 (40%) had at least one described training or validation data set publicly available. 69 studies (58%) reported age data for patients included in training or validation sets, 60 studies (51%) reported sex, and six studies (5%) reported race. Of the studies that reported race, a range of 70.7%-93.4% of individuals were White. Only three studies reported racial demographic data with greater than two categories (i.e. "White" vs. "non-White" or "White" vs. "Black"). CONCLUSIONS We found that a minority of studies (5%) analyzed reported racial and ethnic demographic data. Furthermore, studies that did report racial demographic data had few non-White patients. Increased transparency regarding reporting of demographics and greater representation in data sets is essential to ensure fair and unbiased clinical integration of AI in oncology.
Collapse
Affiliation(s)
| | | | - Chinenye Azoba
- Johns Hopkins University School of MedicineBaltimoreMarylandUSA
| | - Krista Y. Chen
- Johns Hopkins University School of MedicineBaltimoreMarylandUSA
| | - Raymond H. Mak
- Brigham and Women's Hospital/Dana‐Farber Cancer InstituteHarvard Medical SchoolBostonMassachusettsUSA
| | - Subha Perni
- Brigham and Women's Hospital/Dana‐Farber Cancer InstituteHarvard Medical SchoolBostonMassachusettsUSA
- The University of Texas MD Anderson Cancer CenterHoustonTexasUSA
| |
Collapse
|
16
|
Zhu G, Rahman CR, Getty V, Odinokov D, Baruah P, Carrié H, Lim AJ, Guo YA, Poh ZW, Sim NL, Abdelmoneim A, Cai Y, Lakshmanan LN, Ho D, Thangaraju S, Poon P, Lau YT, Gan A, Ng S, Koo SL, Chong DQ, Tay B, Tan TJ, Yap YS, Chok AY, Ng MCH, Tan P, Tan D, Wong L, Wong PM, Tan IB, Skanderup AJ. A deep-learning model for quantifying circulating tumour DNA from the density distribution of DNA-fragment lengths. Nat Biomed Eng 2025; 9:307-319. [PMID: 40055581 DOI: 10.1038/s41551-025-01370-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2023] [Accepted: 02/12/2025] [Indexed: 03/21/2025]
Abstract
The quantification of circulating tumour DNA (ctDNA) in blood enables non-invasive surveillance of cancer progression. Here we show that a deep-learning model can accurately quantify ctDNA from the density distribution of cell-free DNA-fragment lengths. We validated the model, which we named 'Fragle', by using low-pass whole-genome-sequencing data from multiple cancer types and healthy control cohorts. In independent cohorts, Fragle outperformed tumour-naive methods, achieving higher accuracy and lower detection limits. We also show that Fragle is compatible with targeted sequencing data. In plasma samples from patients with colorectal cancer, longitudinal analysis with Fragle revealed strong concordance between ctDNA dynamics and treatment responses. In patients with resected lung cancer, Fragle outperformed a tumour-naive gene panel in the prediction of minimal residual disease for risk stratification. The method's versatility, speed and accuracy for ctDNA quantification suggest that it may have broad clinical utility.
Collapse
Affiliation(s)
- Guanhua Zhu
- Genome Institute of Singapore (GIS), Agency for Science, Technology and Research (A*STAR), Singapore, Singapore
- Centre for Novostics, Hong Kong SAR, China
- Department of Chemical Pathology, The Chinese University of Hong Kong, Hong Kong SAR, China
- Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Chowdhury Rafeed Rahman
- Genome Institute of Singapore (GIS), Agency for Science, Technology and Research (A*STAR), Singapore, Singapore
- School of Computing, National University of Singapore, Singapore, Singapore
| | - Victor Getty
- Genome Institute of Singapore (GIS), Agency for Science, Technology and Research (A*STAR), Singapore, Singapore
| | - Denis Odinokov
- Genome Institute of Singapore (GIS), Agency for Science, Technology and Research (A*STAR), Singapore, Singapore
| | - Probhonjon Baruah
- Genome Institute of Singapore (GIS), Agency for Science, Technology and Research (A*STAR), Singapore, Singapore
| | - Hanaé Carrié
- Genome Institute of Singapore (GIS), Agency for Science, Technology and Research (A*STAR), Singapore, Singapore
- School of Computing, National University of Singapore, Singapore, Singapore
- Institute of Data Science, National University of Singapore, Singapore, Singapore
- Integrative Sciences and Engineering Programme, Graduate School, National University of Singapore, Singapore, Singapore
| | - Avril Joy Lim
- Genome Institute of Singapore (GIS), Agency for Science, Technology and Research (A*STAR), Singapore, Singapore
- School of Computing, National University of Singapore, Singapore, Singapore
| | - Yu Amanda Guo
- Genome Institute of Singapore (GIS), Agency for Science, Technology and Research (A*STAR), Singapore, Singapore
| | - Zhong Wee Poh
- Genome Institute of Singapore (GIS), Agency for Science, Technology and Research (A*STAR), Singapore, Singapore
- Duke-NUS Medical School, National University of Singapore, Singapore, Singapore
| | - Ngak Leng Sim
- Genome Institute of Singapore (GIS), Agency for Science, Technology and Research (A*STAR), Singapore, Singapore
| | - Ahmed Abdelmoneim
- Genome Institute of Singapore (GIS), Agency for Science, Technology and Research (A*STAR), Singapore, Singapore
| | - Yutong Cai
- Genome Institute of Singapore (GIS), Agency for Science, Technology and Research (A*STAR), Singapore, Singapore
| | | | - Danliang Ho
- Genome Institute of Singapore (GIS), Agency for Science, Technology and Research (A*STAR), Singapore, Singapore
| | - Saranya Thangaraju
- Genome Institute of Singapore (GIS), Agency for Science, Technology and Research (A*STAR), Singapore, Singapore
| | - Polly Poon
- Genome Institute of Singapore (GIS), Agency for Science, Technology and Research (A*STAR), Singapore, Singapore
| | - Yi Ting Lau
- Genome Institute of Singapore (GIS), Agency for Science, Technology and Research (A*STAR), Singapore, Singapore
| | - Anna Gan
- Genome Institute of Singapore (GIS), Agency for Science, Technology and Research (A*STAR), Singapore, Singapore
| | - Sarah Ng
- Genome Institute of Singapore (GIS), Agency for Science, Technology and Research (A*STAR), Singapore, Singapore
| | - Si-Lin Koo
- National Cancer Center Singapore, Singapore, Singapore
| | - Dawn Q Chong
- Duke-NUS Medical School, National University of Singapore, Singapore, Singapore
- National Cancer Center Singapore, Singapore, Singapore
| | - Brenda Tay
- National Cancer Center Singapore, Singapore, Singapore
| | - Tira J Tan
- National Cancer Center Singapore, Singapore, Singapore
| | - Yoon Sim Yap
- Duke-NUS Medical School, National University of Singapore, Singapore, Singapore
- National Cancer Center Singapore, Singapore, Singapore
| | | | - Matthew Chau Hsien Ng
- Duke-NUS Medical School, National University of Singapore, Singapore, Singapore
- National Cancer Center Singapore, Singapore, Singapore
| | - Patrick Tan
- Genome Institute of Singapore (GIS), Agency for Science, Technology and Research (A*STAR), Singapore, Singapore
- Duke-NUS Medical School, National University of Singapore, Singapore, Singapore
| | - Daniel Tan
- Genome Institute of Singapore (GIS), Agency for Science, Technology and Research (A*STAR), Singapore, Singapore
- Duke-NUS Medical School, National University of Singapore, Singapore, Singapore
- National Cancer Center Singapore, Singapore, Singapore
| | - Limsoon Wong
- School of Computing, National University of Singapore, Singapore, Singapore
| | - Pui Mun Wong
- Genome Institute of Singapore (GIS), Agency for Science, Technology and Research (A*STAR), Singapore, Singapore
| | - Iain Beehuat Tan
- Genome Institute of Singapore (GIS), Agency for Science, Technology and Research (A*STAR), Singapore, Singapore
- Duke-NUS Medical School, National University of Singapore, Singapore, Singapore
- National Cancer Center Singapore, Singapore, Singapore
| | - Anders Jacobsen Skanderup
- Genome Institute of Singapore (GIS), Agency for Science, Technology and Research (A*STAR), Singapore, Singapore.
- School of Computing, National University of Singapore, Singapore, Singapore.
- National Cancer Center Singapore, Singapore, Singapore.
| |
Collapse
|
17
|
Nguyen Phuong L, Salas S, Benzekry S. Computational Modeling for Circulating Cell-Free DNA in Clinical Oncology. JCO Clin Cancer Inform 2025; 9:e2400224. [PMID: 40020203 DOI: 10.1200/cci-24-00224] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2024] [Revised: 11/23/2024] [Accepted: 01/10/2025] [Indexed: 05/12/2025] Open
Abstract
PURPOSE Liquid biopsy, specifically circulating cell-free DNA (cfDNA), has emerged as a powerful tool for cancer early diagnosis, prognosis, and treatment monitoring over a wide range of cancer types. Computational modeling (CM) of cfDNA data is essential to harness its full potential for real-time, noninvasive insights into tumor biology, enhancing clinical decision making. DESIGN This work reviews CM-cfDNA methods applied to clinical oncology, emphasizing both machine learning (ML) techniques and mechanistic approaches. The latter integrate biological principles, enabling a deeper understanding of cfDNA dynamics and its relationship with tumor evolution. RESULTS Key findings highlight the effectiveness of CM-cfDNA approaches in improving diagnostic accuracy, identifying prognostic markers, and predicting therapeutic outcomes. ML models integrating cfDNA concentration, fragmentation patterns, and mutation detection achieve high sensitivity and specificity for early cancer detection. Mechanistic models describe cfDNA kinetics, linking them to tumor growth and response to treatment, for example, immune checkpoint inhibitors. Longitudinal data and advanced statistical constructs further refine these models for quantification of interindividual and intraindividual variability. CONCLUSION CM-cfDNA represents a pivotal advancement in precision oncology. It bridges the gap between extensive cfDNA data and actionable clinical insights, supporting its integration into routine cancer care. Future efforts should focus on standardizing protocols, validating models across populations, and exploring hybrid approaches combining ML with mechanistic modeling to improve biological understanding.
Collapse
Affiliation(s)
- Linh Nguyen Phuong
- Computational Pharmacology and Clinical Oncology Department, Centre Inria d'Université Côte d'Azur, Cancer Research Centre of Marseille, Paoli Calmettes Institute, Inserm UMR1068, CNRS UMR7258, Aix Marseille University UM105, Marseille, France
| | - Sébastien Salas
- Computational Pharmacology and Clinical Oncology Department, Centre Inria d'Université Côte d'Azur, Cancer Research Centre of Marseille, Paoli Calmettes Institute, Inserm UMR1068, CNRS UMR7258, Aix Marseille University UM105, Marseille, France
- Assistance Publique-Hôpitaux de Marseille, Timone Hospital, Aix Marseille University, Marseille, France
| | - Sébastien Benzekry
- Computational Pharmacology and Clinical Oncology Department, Centre Inria d'Université Côte d'Azur, Cancer Research Centre of Marseille, Paoli Calmettes Institute, Inserm UMR1068, CNRS UMR7258, Aix Marseille University UM105, Marseille, France
| |
Collapse
|
18
|
Kiavue N, Cabel L. [Benefits and limitations of circulating tumor DNA in breast cancer]. Bull Cancer 2025:S0007-4551(25)00033-5. [PMID: 40011140 DOI: 10.1016/j.bulcan.2024.12.016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2024] [Revised: 12/13/2024] [Accepted: 12/22/2024] [Indexed: 02/28/2025]
Abstract
The detection of circulating tumor DNA (ctDNA) has made significant advances in oncology in recent years. ctDNA offers a range of applications, including the identification of theranostic mutations, monitoring of tumor recurrence, and assessing treatment efficacy. In breast cancer, several ctDNA-based tests for detecting relapse during follow-up are currently under validation, with some already available in countries like the United States. In metastatic breast cancer, ctDNA levels and their dynamics during treatment have prognostic value. The PADA-1 trial demonstrated that a therapeutic adaptation based on the detection of a circulating subclone via circulating tumor DNA (ctDNA) was feasible and potentially beneficial for patients. This review will explore the methods for ctDNA detection and discuss the potential benefits of incorporating this technology into breast cancer monitoring and management across various clinical scenarios.
Collapse
Affiliation(s)
- Nicolas Kiavue
- Département d'oncologie médicale, Institut Curie, Paris, France
| | - Luc Cabel
- Département d'oncologie médicale, Institut Curie, Paris, France; Circulating Tumor Biomarkers Laboratory, Inserm CIC BT-1428, Institut Curie, Paris, France.
| |
Collapse
|
19
|
Fu H, Huang K, Zhu W, Zhang L, Bandaru R, Wang L, Liu Y, Xia Z. Circulating cell-free DNA methylation profiles as noninvasive multiple sclerosis biomarkers: A proof-of-concept study. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2025:2025.02.14.25322180. [PMID: 40034794 PMCID: PMC11875267 DOI: 10.1101/2025.02.14.25322180] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/05/2025]
Abstract
In multiple sclerosis (MS), there is a critical need for non-invasive biomarkers to concurrently classify disease subtypes, evaluate disability severity, and predict long-term progression. In this proof-of-concept study, we performed low-coverage whole-genome bisulfite sequencing (WGBS) on 75 plasma cell-free DNA (cfDNA) samples and assessed the clinical utility of cfDNA methylation as a single assay for distinguishing MS patients from non-MS controls, identifying MS subtypes, estimating disability severity, and predicting disease trajectories. We identified thousands of differentially methylated CpGs and hundreds of differentially methylated regions (DMRs) that significantly distinguished MS from controls, separated MS subtypes, and stratified disability severity levels. These DMRs were highly enriched in immunologically and neurologically relevant regulatory elements (e.g., active promoters and enhancers) and contained motifs associated with neuronal function and T-cell differentiation. To distinguish MS subtypes and severity groups, we achieved area-under-the-curve (AUC) values ranging from 0.67 to 0.81 using DMRs and 0.70 to 0.82 using inferred tissue-of-origin patterns from cfDNA methylation, significantly outperforming benchmark neurofilament light chain (NfL) and glial fibrillary acidic protein (GFAP) in the same cohort. Finally, a linear mixed-effects model identified "prognostic regions" where baseline cfDNA methylation levels were associated with disease progression and predicted future disability severity (AUC=0.81) within a 4-year evaluation window. As we plan to generate higher-depth WGBS data and validation in independent cohorts, the present findings suggest the potential clinical utility of circulating cfDNA methylation profiles as promising noninvasive biomarkers in MS diagnosis and prognosis.
Collapse
Affiliation(s)
- Hailu Fu
- Department of Biochemistry and Molecular Genetics, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611
- Robert H. Lurie Comprehensive Cancer Center of Northwestern University, Chicago, IL 60611
| | - Kevin Huang
- Computational Sciences, gRED, Genentech Inc. South San Francisco, CA 94080
| | - Wen Zhu
- University of Pittsburgh, Pittsburgh, PA 15260
| | - Lili Zhang
- University of Pittsburgh, Pittsburgh, PA 15260
| | - Ravi Bandaru
- Department of Biochemistry and Molecular Genetics, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611
- Robert H. Lurie Comprehensive Cancer Center of Northwestern University, Chicago, IL 60611
| | - Li Wang
- Department of Biochemistry and Molecular Genetics, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611
- Robert H. Lurie Comprehensive Cancer Center of Northwestern University, Chicago, IL 60611
| | - Yaping Liu
- Department of Biochemistry and Molecular Genetics, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611
- Robert H. Lurie Comprehensive Cancer Center of Northwestern University, Chicago, IL 60611
| | - Zongqi Xia
- University of Pittsburgh, Pittsburgh, PA 15260
| |
Collapse
|
20
|
Dai Y, He J, Zhou Y, Yu Y, Hui H, Guo L, Yin H. Constructing a highly sensitive duplex immunoassay using AuNPs and AgNPs as nanolabels for investigating the epithelial-mesenchymal transition occurring on circulating tumor cells with lung cancer patients. Biosens Bioelectron 2025; 270:116947. [PMID: 39561553 DOI: 10.1016/j.bios.2024.116947] [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: 09/06/2024] [Revised: 11/01/2024] [Accepted: 11/14/2024] [Indexed: 11/21/2024]
Abstract
Transformation of epithelial to mesenchymal (EMT) is an important event in the process of tumor initiation, invasion and metastasis. Circulating tumor cells (CTCs) are one kind of important markers in the field of liquid tumor biopsy, whose number and phenotype represent the occurrence and progression of tumors. Therefore, it is our interest to investigate the epithelial mesenchymal transition process occurring on the surface of CTCs. Herein in this work, two proteins of E-cadherin (E-cad) and N-cadherin (N-cad) were selected as representative proteins of EMT process. To achieve simultaneous analysis of E-cad and N-cad on the surface of rare CTCs, we designed a duplex and portable immunosensor using AuNPs and AgNPs as nanolabels to amplify the immunoreaction signals. The dual channel immunosensor not only exhibited good electrochemical responses for recombinant E-cad and N-cad as low as 0.1 ng/mL and 0.05 ng/mL, respectively, but also showed good linear correlations with different numbers of phenotypic CTCs (10-500 cells/10 μL). The above strategy was further employed to inspect the occurrence of EMT on CTCs surface, which displayed a high consistence with other molecular biological characterizations. Finally, this immunoassay was successfully applied to inspect the correlations of numbers, phenotype of CTCs, as well as E-cad and N-cad expressions on these CTCs in bloods of NSCLC patients with disease stage.
Collapse
Affiliation(s)
- Yunuo Dai
- Department of Radiotherapy Central Hospital, Affiliated Xuzhou Clinical College of Xuzhou Medical University, 199 Jiefang South Road, Xuzhou, 221009, Jiangsu, PR China
| | - Jie He
- Department of Radiotherapy Central Hospital, Affiliated Xuzhou Clinical College of Xuzhou Medical University, 199 Jiefang South Road, Xuzhou, 221009, Jiangsu, PR China
| | - Yun Zhou
- Department of Radiotherapy Central Hospital, Affiliated Xuzhou Clinical College of Xuzhou Medical University, 199 Jiefang South Road, Xuzhou, 221009, Jiangsu, PR China
| | - Yanyan Yu
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, 209 Tongshan Road, Xuzhou, 221004, Jiangsu, PR China
| | - Hui Hui
- Department of Radiotherapy Central Hospital, Affiliated Xuzhou Clinical College of Xuzhou Medical University, 199 Jiefang South Road, Xuzhou, 221009, Jiangsu, PR China
| | - Lin Guo
- Department of Radiotherapy Central Hospital, Affiliated Xuzhou Clinical College of Xuzhou Medical University, 199 Jiefang South Road, Xuzhou, 221009, Jiangsu, PR China
| | - Haitao Yin
- The Affiliated Xuzhou Municipal Hospital of Xuzhou Medical University, 269 University Road, Xuzhou, 221002, Jiangsu, PR China.
| |
Collapse
|
21
|
Wong J, Muralidhar R, Wang L, Huang CC. Epigenetic modifications of cfDNA in liquid biopsy for the cancer care continuum. Biomed J 2025; 48:100718. [PMID: 38522508 PMCID: PMC11745953 DOI: 10.1016/j.bj.2024.100718] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2024] [Revised: 02/28/2024] [Accepted: 03/14/2024] [Indexed: 03/26/2024] Open
Abstract
This review provides a comprehensive overview of the latest advancements in the clinical utility of liquid biopsy, with a particular focus on epigenetic approaches aimed at overcoming challenges in cancer diagnosis and treatment. It begins by elucidating key epigenetic terms, including methylomics, fragmentomics, and nucleosomics. The review progresses to discuss methods for analyzing circulating cell-free DNA (cfDNA) and highlights recent studies showcasing the clinical relevance of epigenetic modifications in areas such as diagnosis, drug treatment response, minimal residual disease (MRD) detection, and prognosis prediction. While acknowledging hurdles like the complexity of interpreting epigenetic data and the absence of standardization, the review charts a path forward. It advocates for the integration of multi-omic data through machine learning algorithms to refine predictive models and stresses the importance of collaboration among clinicians, researchers, and data scientists. Such cooperative efforts are essential to fully leverage the potential of epigenetic features in clinical practice.
Collapse
Affiliation(s)
- Jodie Wong
- Department of Tumor Microenvironment and Metastasis, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, USA
| | - Rohit Muralidhar
- Nova Southeastern University, Kiran C. Patel College of Osteopathic Medicine, Davie, FL, USA
| | - Liang Wang
- Department of Tumor Microenvironment and Metastasis, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, USA.
| | - Chiang-Ching Huang
- Zilber College of Public Health, University of Wisconsin, Milwaukee, WI, USA.
| |
Collapse
|
22
|
Wang Y, Guo Q, Huang Z, Song L, Zhao F, Gu T, Feng Z, Wang H, Li B, Wang D, Zhou B, Guo C, Xu Y, Song Y, Zheng Z, Bing Z, Li H, Yu X, Fung KL, Xu H, Shi J, Chen M, Hong S, Jin H, Tong S, Zhu S, Zhu C, Song J, Liu J, Li S, Li H, Sun X, Liang N. Cell-free epigenomes enhanced fragmentomics-based model for early detection of lung cancer. Clin Transl Med 2025; 15:e70225. [PMID: 39909829 PMCID: PMC11798665 DOI: 10.1002/ctm2.70225] [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: 11/08/2024] [Revised: 12/24/2024] [Accepted: 01/27/2025] [Indexed: 02/07/2025] Open
Abstract
BACKGROUND Lung cancer is a leading cause of cancer mortality, highlighting the need for innovative non-invasive early detection methods. Although cell-free DNA (cfDNA) analysis shows promise, its sensitivity in early-stage lung cancer patients remains a challenge. This study aimed to integrate insights from epigenetic modifications and fragmentomic features of cfDNA using machine learning to develop a more accurate lung cancer detection model. METHODS To address this issue, a multi-centre prospective cohort study was conducted, with participants harbouring suspicious malignant lung nodules and healthy volunteers recruited from two clinical centres. Plasma cfDNA was analysed for its epigenetic and fragmentomic profiles using chromatin immunoprecipitation sequencing, reduced representation bisulphite sequencing and low-pass whole-genome sequencing. Machine learning algorithms were then employed to integrate the multi-omics data, aiding in the development of a precise lung cancer detection model. RESULTS Cancer-related changes in cfDNA fragmentomics were significantly enriched in specific genes marked by cell-free epigenomes. A total of 609 genes were identified, and the corresponding cfDNA fragmentomic features were utilised to construct the ensemble model. This model achieved a sensitivity of 90.4% and a specificity of 83.1%, with an AUC of 0.94 in the independent validation set. Notably, the model demonstrated exceptional sensitivity for stage I lung cancer cases, achieving 95.1%. It also showed remarkable performance in detecting minimally invasive adenocarcinoma, with a sensitivity of 96.2%, highlighting its potential for early detection in clinical settings. CONCLUSIONS With feature selection guided by multiple epigenetic sequencing approaches, the cfDNA fragmentomics-based machine learning model demonstrated outstanding performance in the independent validation cohort. These findings highlight its potential as an effective non-invasive strategy for the early detection of lung cancer. KEYPOINTS Our study elucidated the regulatory relationships between epigenetic modifications and their effects on fragmentomic features. Identifying epigenetically regulated genes provided a critical foundation for developing the cfDNA fragmentomics-based machine learning model. The model demonstrated exceptional clinical performance, highlighting its substantial potential for translational application in clinical practice.
Collapse
Affiliation(s)
- Yadong Wang
- Department of Thoracic SurgeryPeking Union Medical College HospitalChinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
| | - Qiang Guo
- Department of Thoracic SurgeryAffiliated Hospital of Hebei UniversityBaodingChina
| | - Zhicheng Huang
- Department of Thoracic SurgeryPeking Union Medical College HospitalChinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
| | - Liyang Song
- Shanghai Weihe Medical Laboratory Co., LtdShanghaiChina
| | - Fei Zhao
- Shanghai Weihe Medical Laboratory Co., LtdShanghaiChina
| | - Tiantian Gu
- Shanghai Weihe Medical Laboratory Co., LtdShanghaiChina
| | - Zhe Feng
- Department of Cardiothoracic Surgerythe Sixth Hospital of BeijingBeijingChina
| | - Haibo Wang
- Department of Thoracic SurgeryAffiliated Hospital of Hebei UniversityBaodingChina
| | - Bowen Li
- Department of Thoracic SurgeryPeking Union Medical College HospitalChinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
| | - Daoyun Wang
- Department of Thoracic SurgeryPeking Union Medical College HospitalChinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
| | - Bin Zhou
- Department of Thoracic SurgeryAffiliated Hospital of Hebei UniversityBaodingChina
| | - Chao Guo
- Department of Thoracic SurgeryPeking Union Medical College HospitalChinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
| | - Yuan Xu
- Department of Thoracic SurgeryPeking Union Medical College HospitalChinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
| | - Yang Song
- Department of Thoracic SurgeryPeking Union Medical College HospitalChinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
| | - Zhibo Zheng
- Department of Thoracic SurgeryPeking Union Medical College HospitalChinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
| | - Zhongxing Bing
- Department of Thoracic SurgeryPeking Union Medical College HospitalChinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
| | - Haochen Li
- Department of Thoracic SurgeryPeking Union Medical College HospitalChinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
| | - Xiaoqing Yu
- Department of Thoracic SurgeryPeking Union Medical College HospitalChinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
| | - Ka Luk Fung
- Department of Thoracic SurgeryPeking Union Medical College HospitalChinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
| | - Heqing Xu
- Department of Thoracic SurgeryPeking Union Medical College HospitalChinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
| | - Jianhong Shi
- Department of Scientific ResearchAffiliated Hospital of Hebei UniversityBaodingChina
| | - Meng Chen
- Department of Scientific ResearchAffiliated Hospital of Hebei UniversityBaodingChina
| | - Shuai Hong
- Shanghai Weihe Medical Laboratory Co., LtdShanghaiChina
| | - Haoxuan Jin
- Shanghai Weihe Medical Laboratory Co., LtdShanghaiChina
| | - Shiyuan Tong
- Shanghai Weihe Medical Laboratory Co., LtdShanghaiChina
| | - Sibo Zhu
- Shanghai Weihe Medical Laboratory Co., LtdShanghaiChina
| | - Chen Zhu
- Shanghai Weihe Medical Laboratory Co., LtdShanghaiChina
| | - Jinlei Song
- Shanghai Weihe Medical Laboratory Co., LtdShanghaiChina
| | - Jing Liu
- Shanghai Weihe Medical Laboratory Co., LtdShanghaiChina
| | - Shanqing Li
- Department of Thoracic SurgeryPeking Union Medical College HospitalChinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
| | - Hefei Li
- Department of Thoracic SurgeryAffiliated Hospital of Hebei UniversityBaodingChina
| | - Xueguang Sun
- Shanghai Weihe Medical Laboratory Co., LtdShanghaiChina
| | - Naixin Liang
- Department of Thoracic SurgeryPeking Union Medical College HospitalChinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
| |
Collapse
|
23
|
Afridi WA, Picos SH, Bark JM, Stamoudis DAF, Vasani S, Irwin D, Fielding D, Punyadeera C. Minimally invasive biomarkers for triaging lung nodules-challenges and future perspectives. Cancer Metastasis Rev 2025; 44:29. [PMID: 39888565 PMCID: PMC11785609 DOI: 10.1007/s10555-025-10247-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/05/2024] [Accepted: 01/23/2025] [Indexed: 02/01/2025]
Abstract
CT chest scans are commonly performed worldwide, either in routine clinical practice for a wide range of indications or as part of lung cancer screening programs. Many of these scans detect lung nodules, which are small, rounded opacities measuring 8-30 mm. While the concern about nodules is that they may represent early lung cancer, in screening programs, only 1% of such nodules turn out to be cancer. This leads to a series of complex decisions and, at times, unnecessary biopsies for nodules that are ultimately determined to be benign. Additionally, patients may be anxious about the status of detected lung nodules. The high rate of false positive lung nodule detections has driven advancements in biomarker-based research aimed at triaging lung nodules (benign versus malignant) to identify truly malignant nodules better. Biomarkers found in biofluids and breath hold promise owing to their minimally invasive sampling methods, ease of use, and cost-effectiveness. Although several biomarkers have demonstrated clinical utility, their sensitivity and specificity are still relatively low. Combining multiple biomarkers could enhance the characterisation of small pulmonary nodules by addressing the limitations of individual biomarkers. This approach may help reduce unnecessary invasive procedures and accelerate diagnosis in the future. This review offers a thorough overview of emerging minimally invasive biomarkers for triaging lung nodules, emphasising key challenges and proposing potential solutions for biomarker-based nodule differentiation. It focuses on diagnosis rather than screening, analysing research published primarily in the past five years with some exceptions. The incorporation of biomarkers into clinical practice will facilitate the early detection of malignant nodules, leading to timely interventions and improved outcomes. Further efforts are needed to increase the cost-effectiveness and practicality of many of these applications in clinical settings. However, the range of technologies is advancing rapidly, and they may soon be implemented in clinics in the near future.
Collapse
Affiliation(s)
- Waqar Ahmed Afridi
- Saliva and Liquid Biopsy Translational Laboratory, Institute for Biomedicine and Glycomics (IBG), Griffith University, Brisbane, 4111, Australia
- Virtual University of Pakistan, Islamabad, 44000, Pakistan
| | - Samandra Hernandez Picos
- Saliva and Liquid Biopsy Translational Laboratory, Institute for Biomedicine and Glycomics (IBG), Griffith University, Brisbane, 4111, Australia
| | - Juliana Muller Bark
- Saliva and Liquid Biopsy Translational Laboratory, Institute for Biomedicine and Glycomics (IBG), Griffith University, Brisbane, 4111, Australia
| | - Danyelle Assis Ferreira Stamoudis
- Saliva and Liquid Biopsy Translational Laboratory, Institute for Biomedicine and Glycomics (IBG), Griffith University, Brisbane, 4111, Australia
| | - Sarju Vasani
- Department of Otolaryngology, Royal Brisbane and Women's Hospital, Herston, 4006, Australia
| | - Darryl Irwin
- The Agena Biosciences, Bowen Hills, Brisbane, 4006, Australia
| | - David Fielding
- The Royal Brisbane and Women's Hospital, Herston, Brisbane, 4006, Australia
| | - Chamindie Punyadeera
- Saliva and Liquid Biopsy Translational Laboratory, Institute for Biomedicine and Glycomics (IBG), Griffith University, Brisbane, 4111, Australia.
| |
Collapse
|
24
|
Tsui WHA, Ding SC, Jiang P, Lo YMD. Artificial intelligence and machine learning in cell-free-DNA-based diagnostics. Genome Res 2025; 35:1-19. [PMID: 39843210 PMCID: PMC11789496 DOI: 10.1101/gr.278413.123] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2025]
Abstract
The discovery of circulating fetal and tumor cell-free DNA (cfDNA) molecules in plasma has opened up tremendous opportunities in noninvasive diagnostics such as the detection of fetal chromosomal aneuploidies and cancers and in posttransplantation monitoring. The advent of high-throughput sequencing technologies makes it possible to scrutinize the characteristics of cfDNA molecules, opening up the fields of cfDNA genetics, epigenetics, transcriptomics, and fragmentomics, providing a plethora of biomarkers. Machine learning (ML) and/or artificial intelligence (AI) technologies that are known for their ability to integrate high-dimensional features have recently been applied to the field of liquid biopsy. In this review, we highlight various AI and ML approaches in cfDNA-based diagnostics. We first introduce the biology of cell-free DNA and basic concepts of ML and AI technologies. We then discuss selected examples of ML- or AI-based applications in noninvasive prenatal testing and cancer liquid biopsy. These applications include the deduction of fetal DNA fraction, plasma DNA tissue mapping, and cancer detection and localization. Finally, we offer perspectives on the future direction of using ML and AI technologies to leverage cfDNA fragmentation patterns in terms of methylomic and transcriptional investigations.
Collapse
Affiliation(s)
- W H Adrian Tsui
- Center for Novostics, Hong Kong Science Park, Pak Shek Kok, New Territories, Hong Kong SAR, China
- Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Shatin, New Territories, Hong Kong SAR, China
- Department of Chemical Pathology, The Chinese University of Hong Kong, Shatin, New Territories, Hong Kong SAR, China
| | - Spencer C Ding
- Center for Novostics, Hong Kong Science Park, Pak Shek Kok, New Territories, Hong Kong SAR, China
- Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Shatin, New Territories, Hong Kong SAR, China
- Department of Chemical Pathology, The Chinese University of Hong Kong, Shatin, New Territories, Hong Kong SAR, China
| | - Peiyong Jiang
- Center for Novostics, Hong Kong Science Park, Pak Shek Kok, New Territories, Hong Kong SAR, China
- Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Shatin, New Territories, Hong Kong SAR, China
- Department of Chemical Pathology, The Chinese University of Hong Kong, Shatin, New Territories, Hong Kong SAR, China
- State Key Laboratory of Translational Oncology, The Chinese University of Hong Kong, Shatin, New Territories, Hong Kong SAR, China
| | - Y M Dennis Lo
- Center for Novostics, Hong Kong Science Park, Pak Shek Kok, New Territories, Hong Kong SAR, China;
- Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Shatin, New Territories, Hong Kong SAR, China
- Department of Chemical Pathology, The Chinese University of Hong Kong, Shatin, New Territories, Hong Kong SAR, China
- State Key Laboratory of Translational Oncology, The Chinese University of Hong Kong, Shatin, New Territories, Hong Kong SAR, China
| |
Collapse
|
25
|
Rech GE, Lau AC, Goldfeder RL, Maurya R, Danilov AV, Wei CL. Global DNA methylomes reveal oncogenic-associated 5-hydroxylmethylated cytosine (5hmC) signatures in the cell-free DNA of cancer patients. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2025:2025.01.09.25320283. [PMID: 39867387 PMCID: PMC11759829 DOI: 10.1101/2025.01.09.25320283] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 01/28/2025]
Abstract
Characterization of tumor epigenetic aberrations is integral to understanding the mechanisms of tumorigenesis and provide diagnostic, prognostic, and predictive information of high clinical relevance. Among the different tumor-associated epigenetic signatures, 5 methyl-cytosine (5mC) and 5-hydroxymethylcytosine (5hmC) are the two most well-characterized DNA methylation alterations linked to cancer pathogenesis. 5hmC has a tissue-specific distribution and its abundance is subjected to changes in tumor DNA, making it a promising biomarker. Detecting tumor-related DNA methylation alterations in tissues is highly invasive, while the analysis of the cell-free DNA (cfDNA) is poised to supplement, if not replace, surgical biopsies. Despite many studies attempted to identify new epigenetic targets for liquid biopsy assays, little is known about the regulatory roles of 5hmC, its impacts on the molecular phenotypes in tumors. Most importantly, whether the oncogenic-associated 5hmC signatures found in tumor tissues can be recapitulated in patients' cfDNA. In this study, we performed the unbiased and simultaneous detection of 5mC and 5hmC whole-genome DNA modifications at base-resolution from two distinct cancer cohorts, from patients with bladder cancer or B-Cell lymphoma, their corresponding normal tissues, and cfDNAs from plasma. We analyzed tissue-specific methylation patters and searched for signatures in gene coding and regulatory regions linked to cancerous states. We then looked for methylation signatures in patients' cfDNA to determine if they were consistent with the tumor-specific patterns. We determined the functional significance of 5hmC in tissue specific transcription and uncovered hundreds of tumor-associated 5hmC signatures. These tumor-associated 5hmC changes, particularly in genes and enhancers, were functionally significant in tumorigenesis pathways and correlated with tumor specific gene expression. To investigate if cfDNA is a faithful surrogate for tumor-associated 5hmC, we devised a targeted capture strategy to examine the alterations of 5hmC in cfDNA from patients with bladder cancer and lymphoma with sufficient sensitivity and specificity and confirmed that they recapitulated the patterns we observed in tumor tissues. Our results provide analytic validation of 5hmC as a cancer-specific biomarker. The methods described here for systematic characterization of 5hmC at functional elements open new avenues to discover epigenetic markers for non-invasive diagnosis, monitoring, and stratifying cancer.
Collapse
Affiliation(s)
- Gabriel E Rech
- The Jackson Laboratory for Genomic Medicine, Farmington, CT, 06032, USA
| | - Alyssa C Lau
- The Jackson Laboratory for Genomic Medicine, Farmington, CT, 06032, USA
| | | | - Rahul Maurya
- The Jackson Laboratory for Genomic Medicine, Farmington, CT, 06032, USA
| | | | - Chia-Lin Wei
- Department of Genome Sciences, University of Washington, Seattle, WA 98195, USA
| |
Collapse
|
26
|
Zhu D, Li J, Zhang W, Wang Y, Wang H, Fei R, Ye Q, Peng D, Luan J, Xu C, Wu X, Huang D, Ding C, Jin S. Highly specific multiplex DNA methylation detection for liquid biopsy of colorectal cancer. Clin Chim Acta 2025; 565:120026. [PMID: 39491766 DOI: 10.1016/j.cca.2024.120026] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2024] [Revised: 10/24/2024] [Accepted: 10/31/2024] [Indexed: 11/05/2024]
Abstract
BACKGROUND Circulating tumor DNA (ctDNA) has emerged as a useful biomarker for cancer detection and prognosis. In this study, we developed a strategy for developing a highly specific multiplex qPCR assay to detect methylated ctDNA in the blood of colorectal cancer (CRC) patients and investigated the potential use for the detection and prognosis of CRC. METHODS Bisulfite conversion and amplicon sequencing were used to confirm potential CRC-specific DNA methylation markers. The selected DNA methylation candidates were validated by qMSP. The six best-performing markers were used to develop a new single-tube multiplex quantitative methylation-specific PCR assay (mqMSP). The mqMSP assay was applied to analyze plasma samples from 114 CRC patients, 47 patients with advanced adenoma, 45 patients with benign polyps, and 57 healthy controls. The clinical performance of the assay and associations with clinical outcomes were assessed. RESULTS Six DNA methylation biomarkers were confirmed to be specifically hypermethylated in CRC tumor tissues. The newly developed mqMSP assay detected CRC with extremely high specificity (specificity of 98.2 %, with sensitivity of 67.5 %). The detection rate of ctDNA was significantly correlated with tumor size and clinical stage, with ctDNA methylation levels in the blood markedly increased with larger tumor size, poor differentiation, and advanced stage. Moreover, high preoperative methylated ctDNA level was associated with worse recurrence-free survival and overall survival. CONCLUSION We provided a strategy for identification of multiple highly-specific DNA methylation markers for designing multiplex DNA methylation assays for liquid biopsies of CRC. The newly developed assay has potential for CRC early detection, and prognosis.
Collapse
Affiliation(s)
- Dewen Zhu
- Key Laboratory of Laboratory Medicine, Ministry of Education of China, School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China; Department of Laboratory Medicine, Taihe Hospital, Hubei University of Medicine, Shiyan 442000, China
| | - Jinlei Li
- Wenzhou Medical University, Wenzhou, Zhejiang 325035, China; Department of Colorectal Surgery, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325000, China
| | - Wenwen Zhang
- Department of Pathology, Fudan University Shanghai Cancer Center, Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - Yishuai Wang
- Key Laboratory of Laboratory Medicine, Ministry of Education of China, School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China
| | - Huidong Wang
- Key Laboratory of Laboratory Medicine, Ministry of Education of China, School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China
| | - Ruoyan Fei
- Key Laboratory of Laboratory Medicine, Ministry of Education of China, School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China
| | - Qian Ye
- Key Laboratory of Laboratory Medicine, Ministry of Education of China, School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China
| | - Danli Peng
- Key Laboratory of Laboratory Medicine, Ministry of Education of China, School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China
| | - Ju Luan
- Zhejiang Innomed Biomedical Co., Ltd., Wenzhou 325036, China
| | - Chang Xu
- Department of Colorectal Surgery, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325000, China
| | - Xiaoli Wu
- Department of Gastroenterology, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325000, China
| | - Dan Huang
- Key Laboratory of Laboratory Medicine, Ministry of Education of China, School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China
| | - Chunming Ding
- Key Laboratory of Laboratory Medicine, Ministry of Education of China, School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China; Department of Pathology, Fudan University Shanghai Cancer Center, Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China.
| | - Shengnan Jin
- Key Laboratory of Laboratory Medicine, Ministry of Education of China, School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China.
| |
Collapse
|
27
|
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
|
28
|
Vavoulis DV, Cutts A, Thota N, Brown J, Sugar R, Rueda A, Ardalan A, Howard K, Matos Santo F, Sannasiddappa T, Miller B, Ash S, Liu Y, Song CX, Nicholson BD, Dreau H, Tregidgo C, Schuh A. Multimodal cell-free DNA whole-genome TAPS is sensitive and reveals specific cancer signals. Nat Commun 2025; 16:430. [PMID: 39779727 PMCID: PMC11711490 DOI: 10.1038/s41467-024-55428-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2024] [Accepted: 12/05/2024] [Indexed: 01/11/2025] Open
Abstract
The analysis of circulating tumour DNA (ctDNA) through minimally invasive liquid biopsies is promising for early multi-cancer detection and monitoring minimal residual disease. Most existing methods focus on targeted deep sequencing, but few integrate multiple data modalities. Here, we develop a methodology for ctDNA detection using deep (80x) whole-genome TET-Assisted Pyridine Borane Sequencing (TAPS), a less destructive approach than bisulphite sequencing, which permits the simultaneous analysis of genomic and methylomic data. We conduct a diagnostic accuracy study across multiple cancer types in symptomatic patients, achieving 94.9% sensitivity and 88.8% specificity. Matched tumour biopsies are used for validation, not for guiding the analysis, imitating an early detection scenario. Furthermore, in silico validation demonstrates strong discrimination (86% AUC) at ctDNA fractions as low as 0.7%. Additionally, we successfully track tumour burden and ctDNA shedding from precancerous lesions post-treatment without requiring matched tumour biopsies. This pipeline is ready for further clinical evaluation to extend cancer screening and improve patient triage and monitoring.
Collapse
Affiliation(s)
- Dimitrios V Vavoulis
- Oxford Molecular Diagnostics Centre, Department of Oncology, University of Oxford, Oxford, UK.
- Biomedical Research Centre, Centre for Human Genetics, University of Oxford, Oxford, UK.
| | - Anthony Cutts
- Oxford Molecular Diagnostics Centre, Department of Oncology, University of Oxford, Oxford, UK
| | - Nishita Thota
- Exact Sciences Innovation LTD, The Sherard Bldg, Edmund Halley Rd, Littlemore, Oxford, UK
| | - Jordan Brown
- Exact Sciences Innovation LTD, The Sherard Bldg, Edmund Halley Rd, Littlemore, Oxford, UK
| | - Robert Sugar
- Exact Sciences Innovation LTD, The Sherard Bldg, Edmund Halley Rd, Littlemore, Oxford, UK
| | - Antonio Rueda
- Exact Sciences Innovation LTD, The Sherard Bldg, Edmund Halley Rd, Littlemore, Oxford, UK
| | - Arman Ardalan
- Oxford Molecular Diagnostics Centre, Department of Oncology, University of Oxford, Oxford, UK
| | - Kieran Howard
- Oxford Molecular Diagnostics Centre, Department of Oncology, University of Oxford, Oxford, UK
| | - Flavia Matos Santo
- Oxford Molecular Diagnostics Centre, Department of Oncology, University of Oxford, Oxford, UK
| | - Thippesh Sannasiddappa
- Exact Sciences Innovation LTD, The Sherard Bldg, Edmund Halley Rd, Littlemore, Oxford, UK
| | - Bronwen Miller
- Exact Sciences Innovation LTD, The Sherard Bldg, Edmund Halley Rd, Littlemore, Oxford, UK
| | - Stephen Ash
- Ludwig Institute for Cancer Research, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Yibin Liu
- College of Chemistry and Molecular Sciences, Wuhan University, Wuhan, China
- Taikang Centre for Life and Medical Sciences, Wuhan University, Wuhan, China
| | - Chun-Xiao Song
- Ludwig Institute for Cancer Research, Nuffield Department of Medicine, University of Oxford, Oxford, UK
- Target Discovery Institute, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Brian D Nicholson
- Nuffield Department of Primary Care Health Sciences, University of Oxford, Oxford, UK
| | - Helene Dreau
- Oxford Molecular Diagnostics Centre, Department of Oncology, University of Oxford, Oxford, UK
| | - Carolyn Tregidgo
- Exact Sciences Innovation LTD, The Sherard Bldg, Edmund Halley Rd, Littlemore, Oxford, UK
| | - Anna Schuh
- Oxford Molecular Diagnostics Centre, Department of Oncology, University of Oxford, Oxford, UK.
| |
Collapse
|
29
|
Kang SK, Gulati R, Moise N, Hur C, Elkin EB. Multi-Cancer Early Detection Tests: State of the Art and Implications for Radiologists. Radiology 2025; 314:e233448. [PMID: 39807974 PMCID: PMC11783158 DOI: 10.1148/radiol.233448] [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: 12/19/2023] [Revised: 03/21/2024] [Accepted: 04/25/2024] [Indexed: 01/16/2025]
Abstract
Multi-cancer early detection (MCED) tests are already being marketed as noninvasive, convenient opportunities to test for multiple cancer types with a single blood sample. The technology varies-involving detection of circulating tumor DNA, fragments of DNA, RNA, or proteins unique to each targeted cancer. The priorities and tradeoffs of reaching diagnostic resolution in the setting of possible false positives and negatives remain under active study. Given the well-established role of imaging in lesion detection and characterization for most cancers, radiologists have an essential role to play in selecting diagnostic pathways, determining the validity of test results, resolving false-positive MCED test results, and evaluating tradeoffs for clinical policy. Appropriate access to and use of imaging tests will also factor into clinical guidelines. Thus, all clinicians potentially involved with MCED tests for cancer screening will need to weigh the benefits and harms of MCED testing, including consideration of how the tests will be used alongside or in place of other screening options, how diagnostic confirmation tests should be selected, and what the implications are for policy and reimbursement decisions. Further, patients will need regular support to make informed decisions about screening using MCED tests in the context of their personal cancer risks, health-related values, and access to care.
Collapse
Affiliation(s)
| | - Roman Gulati
- From the Departments of Radiology and Population Health, New York
University Langone Medical Center, New York, NY (S.K.K.); Division of Public
Health Sciences, Fred Hutchinson Cancer Center, Seattle, Wash (R.G.); Department
of Medicine, Vagelos College of Physicians and Surgeons, Columbia University,
New York, NY (N.M., C.H.); Herbert Irving Comprehensive Cancer Center, New York,
NY (C.H., E.B.E.); and Department of Health Policy and Management, Mailman
School of Public Health, Columbia University, New York, NY (E.B.E.)
| | - Nathalie Moise
- From the Departments of Radiology and Population Health, New York
University Langone Medical Center, New York, NY (S.K.K.); Division of Public
Health Sciences, Fred Hutchinson Cancer Center, Seattle, Wash (R.G.); Department
of Medicine, Vagelos College of Physicians and Surgeons, Columbia University,
New York, NY (N.M., C.H.); Herbert Irving Comprehensive Cancer Center, New York,
NY (C.H., E.B.E.); and Department of Health Policy and Management, Mailman
School of Public Health, Columbia University, New York, NY (E.B.E.)
| | - Chin Hur
- From the Departments of Radiology and Population Health, New York
University Langone Medical Center, New York, NY (S.K.K.); Division of Public
Health Sciences, Fred Hutchinson Cancer Center, Seattle, Wash (R.G.); Department
of Medicine, Vagelos College of Physicians and Surgeons, Columbia University,
New York, NY (N.M., C.H.); Herbert Irving Comprehensive Cancer Center, New York,
NY (C.H., E.B.E.); and Department of Health Policy and Management, Mailman
School of Public Health, Columbia University, New York, NY (E.B.E.)
| | - Elena B. Elkin
- From the Departments of Radiology and Population Health, New York
University Langone Medical Center, New York, NY (S.K.K.); Division of Public
Health Sciences, Fred Hutchinson Cancer Center, Seattle, Wash (R.G.); Department
of Medicine, Vagelos College of Physicians and Surgeons, Columbia University,
New York, NY (N.M., C.H.); Herbert Irving Comprehensive Cancer Center, New York,
NY (C.H., E.B.E.); and Department of Health Policy and Management, Mailman
School of Public Health, Columbia University, New York, NY (E.B.E.)
| |
Collapse
|
30
|
Huo B, Kontouli KM, Manos D, Xu Z, Chun S, Fris J, Wallace AMR, French DG. Screening Criteria Evaluation for Expansion in Pulmonary Neoplasias (SCREEN) II. Can J Surg 2025; 68:E1-E9. [PMID: 39753323 PMCID: PMC11684922 DOI: 10.1503/cjs.015223] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/15/2024] [Indexed: 01/23/2025] Open
Abstract
BACKGROUND There is a need to expand eligibility criteria for lung cancer screening beyond age and smoking history. In this study, we sought to assess whether light-or-never-smokers and heavy smokers differ in molecular and immunologic markers based on conventional lung cancer screening criteria. METHODS We conducted a retrospective review of lung cancer cases from 2005 to 2018 at a tertiary Canadian institution. We used multivariable logistic regression to compare the rate of molecular mutations (KRAS, EGFR, BRAF, PIK3CA, ALK, and PD-L1 [< 1%, 1%-49%, ≥ 50%]) and survival between light-or-never-smokers and heavy smokers. RESULTS We included 1156 patients with lung cancer. Overall, 46.4% (National Lung Screening Trial [NLST], n = 536) and 63.3% (Nederlands-Leuvens Long-kanker Screenings Onderzoek [NELSON], n = 732) of the patients were heavy smokers. Using NELSON criteria, screen-ineligible light-or-never-smokers were more frequently from areas at high risk for radon exposure (n = 175 [41.3%]) than screen-eligible heavy smokers (n = 285 [38.9%]). Light-or-never-smokers were more likely to be EGFR-positive in both NLST (odds ratio [OR] 0.79, 95% confidence interval [CI] 0.21-1.37; p = 0.008] and NELSON (OR 0.79, 95% CI 0.28-1.31; p = 0.002) models. Female light-or-never-smokers were more likely than male light-or-never-smokers to be EGFR-positive in NELSON (OR 0.59, 95% CI 0.06-1.12; p = 0.03] but not NLST (OR 0.51, 95% CI 0.02-1.05; p = 0.06) models. Light-or-never-smokers were more often PIK3CA-positive using NLST (OR 1.33, 95% CI 0.54-2.13; p = 0.001) and NELSON (OR 1.19, 95% CI 0.49-1.90; p = 0.001) models. Light-or-never-smokers in the NELSON model were at higher risk of death. CONCLUSION Screen-ineligible light-or-never-smokers had a higher rate of EGFR-and PIK3CA-positive lung cancers than screen-eligible heavy smokers when defined using trial-based lung cancer screening eligibility criteria. Molecular profiling, particularly where targeted therapy is available, should be considered in future studies establishing criteria for lung cancer screening. CONTEXTE Il faut élargir les critères d'admissibilité au dépistage du cancer du poumon au-delà de l'âge et des antécédents tabagiques. Dans cette étude, nous avons voulu vérifier s'il y a des différences entre les personnes dont le tabagisme est léger, voire nul (groupe 1) et celles qui fument beaucoup (groupe 2) au plan des marqueurs moléculaires et immunologiques selon les critères classiques de dépistage du cancer du poumon. MÉTHODES: Nous avons procédé à une revue rétrospective des cas de cancer du poumon de 2005 à 2018 dans un établissement de soins tertiaires canadien. Nous avons utilisé la régression logistique multivariée pour comparer les taux de mutations moléculaires (KRAS, EGFR, BRAF, PIK3CA, ALK et PD-L1 [< 1 %, 1 %-49 %, ≥ 50 %]) et la survie entre les 2 groupes. RÉSULTATS: Nous avons inclus 1156 cas de cancer du poumon. En tout, 46,4 % (étude NLST [National Lung Screening Trial], n = 536) et 63,3 % (étude NELSON [ Nederlands-Leuvens Longkanker Screenings Onderzoek], n = 732) des malades étaient de gros fumeurs. À partir des critères de l'étude NELSON, le groupe 1, non admissible au dépistage, venait de secteurs à risque élevé d'exposition au radon (n = 175 [41,3 %]) comparativement au groupe 2, admissible au dépistage (n = 285 [38,9 %]). Le groupe 1 était plus susceptible d'être EGFR-positif, tant selon le modèle NLST (rapport des cotes [RC] 0,79, intervalle de confiance [IC] de 95 % 0,21-1,37; p = 0,008), que le modèle NELSON (RC 0,79, IC de 95 % 0,28-1,31; p = 0,002). Dans le groupe 1, les femmes étaient plus susceptibles que les hommes d'être EGFR-positives selon le modèle NELSON (RC 0,59, IC de 95 % 0,06-1,12; p = 0,03), mais non selon le modèle NLST (RC 0,51, IC de 95 % 0,02-1,05; p = 0,06). Le groupe 1 avait plus tendance à être PIK3CA-positif selon les modèles NLST (RC 1,33, IC de 95 % 0,54-2,13; p = 0,001) et NELSON (RC 1,19, IC de 95 % 0,49-1,90; p = 0,001). Selon le modèle NELSON, le groupe 1 était exposé à un risque de mortalité plus élevé. CONCLUSION Les personnes dont le tabagisme est léger voire nul qui ne sont pas admissibles au dépistage ont présenté un taux plus élevé de cancer du poumon EGFRet PIK3CA-positifs comparativement aux gros fumeurs, lorsqu'on appliquait les critères d'admissibilité au dépistage du cancer du poumon des 2 essais cités. Il faudrait envisager un profilage moléculaire lors des prochaines études qui porteront sur les critères d'admissibilité au dépistage du cancer du poumon, surtout lorsqu'il existe des modalités thérapeutiques ciblées.
Collapse
Affiliation(s)
- Bright Huo
- From the Faculty of Medicine, Dalhousie University, Halifax, N.S. (Huo); the Department of Primary Education, School of Education, University of Ioannina, Ioannina, Greece (Kontouli); the Department of Diagnostic Radiology, Dalhousie University, Halifax, N.S. (Manos); the Department of Pathology, Dalhousie University, Halifax, N.S. (Xu, Fris); the Department of Urology, Dalhousie University, Halifax, N.S. (Chun); the Division of Thoracic Surgery, Department of Surgery, Dalhousie University, Halifax, N.S. (Wallace, French)
| | - Katerina-Maria Kontouli
- From the Faculty of Medicine, Dalhousie University, Halifax, N.S. (Huo); the Department of Primary Education, School of Education, University of Ioannina, Ioannina, Greece (Kontouli); the Department of Diagnostic Radiology, Dalhousie University, Halifax, N.S. (Manos); the Department of Pathology, Dalhousie University, Halifax, N.S. (Xu, Fris); the Department of Urology, Dalhousie University, Halifax, N.S. (Chun); the Division of Thoracic Surgery, Department of Surgery, Dalhousie University, Halifax, N.S. (Wallace, French)
| | - Daria Manos
- From the Faculty of Medicine, Dalhousie University, Halifax, N.S. (Huo); the Department of Primary Education, School of Education, University of Ioannina, Ioannina, Greece (Kontouli); the Department of Diagnostic Radiology, Dalhousie University, Halifax, N.S. (Manos); the Department of Pathology, Dalhousie University, Halifax, N.S. (Xu, Fris); the Department of Urology, Dalhousie University, Halifax, N.S. (Chun); the Division of Thoracic Surgery, Department of Surgery, Dalhousie University, Halifax, N.S. (Wallace, French)
| | - Zhaolin Xu
- From the Faculty of Medicine, Dalhousie University, Halifax, N.S. (Huo); the Department of Primary Education, School of Education, University of Ioannina, Ioannina, Greece (Kontouli); the Department of Diagnostic Radiology, Dalhousie University, Halifax, N.S. (Manos); the Department of Pathology, Dalhousie University, Halifax, N.S. (Xu, Fris); the Department of Urology, Dalhousie University, Halifax, N.S. (Chun); the Division of Thoracic Surgery, Department of Surgery, Dalhousie University, Halifax, N.S. (Wallace, French)
| | - Samuel Chun
- From the Faculty of Medicine, Dalhousie University, Halifax, N.S. (Huo); the Department of Primary Education, School of Education, University of Ioannina, Ioannina, Greece (Kontouli); the Department of Diagnostic Radiology, Dalhousie University, Halifax, N.S. (Manos); the Department of Pathology, Dalhousie University, Halifax, N.S. (Xu, Fris); the Department of Urology, Dalhousie University, Halifax, N.S. (Chun); the Division of Thoracic Surgery, Department of Surgery, Dalhousie University, Halifax, N.S. (Wallace, French)
| | - John Fris
- From the Faculty of Medicine, Dalhousie University, Halifax, N.S. (Huo); the Department of Primary Education, School of Education, University of Ioannina, Ioannina, Greece (Kontouli); the Department of Diagnostic Radiology, Dalhousie University, Halifax, N.S. (Manos); the Department of Pathology, Dalhousie University, Halifax, N.S. (Xu, Fris); the Department of Urology, Dalhousie University, Halifax, N.S. (Chun); the Division of Thoracic Surgery, Department of Surgery, Dalhousie University, Halifax, N.S. (Wallace, French)
| | - Alison M R Wallace
- From the Faculty of Medicine, Dalhousie University, Halifax, N.S. (Huo); the Department of Primary Education, School of Education, University of Ioannina, Ioannina, Greece (Kontouli); the Department of Diagnostic Radiology, Dalhousie University, Halifax, N.S. (Manos); the Department of Pathology, Dalhousie University, Halifax, N.S. (Xu, Fris); the Department of Urology, Dalhousie University, Halifax, N.S. (Chun); the Division of Thoracic Surgery, Department of Surgery, Dalhousie University, Halifax, N.S. (Wallace, French)
| | - Daniel G French
- From the Faculty of Medicine, Dalhousie University, Halifax, N.S. (Huo); the Department of Primary Education, School of Education, University of Ioannina, Ioannina, Greece (Kontouli); the Department of Diagnostic Radiology, Dalhousie University, Halifax, N.S. (Manos); the Department of Pathology, Dalhousie University, Halifax, N.S. (Xu, Fris); the Department of Urology, Dalhousie University, Halifax, N.S. (Chun); the Division of Thoracic Surgery, Department of Surgery, Dalhousie University, Halifax, N.S. (Wallace, French)
| |
Collapse
|
31
|
Behrouzi R, Clipson A, Simpson KL, Blackhall F, Rothwell DG, Dive C, Mouliere F. Cell-free and extrachromosomal DNA profiling of small cell lung cancer. Trends Mol Med 2025; 31:64-78. [PMID: 39232927 DOI: 10.1016/j.molmed.2024.08.004] [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: 05/30/2024] [Revised: 08/09/2024] [Accepted: 08/12/2024] [Indexed: 09/06/2024]
Abstract
Small cell lung cancer (SCLC) is highly aggressive with poor prognosis. Despite a relative prevalence of circulating tumour DNA (ctDNA) in SCLC, liquid biopsies are not currently implemented, unlike non-SCLC where cell-free DNA (cfDNA) mutation profiling in the blood has utility for guiding targeted therapies and assessing minimal residual disease. cfDNA methylation profiling is highly sensitive for SCLC detection and holds promise for disease monitoring and molecular subtyping; cfDNA fragmentation profiling has also demonstrated clinical potential. Extrachromosomal DNA (ecDNA), that is often observed in SCLC, promotes tumour heterogeneity and chemotherapy resistance and can be detected in blood. We discuss how these cfDNA profiling modalities can be harnessed to expand the clinical applications of liquid biopsy in SCLC.
Collapse
Affiliation(s)
- Roya Behrouzi
- Department of Medical Oncology, The Christie NHS Foundation Trust, Manchester, UK; Faculty of Biology, Medicine, and Health, University of Manchester, Manchester, UK; Cancer Research UK National Biomarker Centre, University of Manchester, Manchester, UK; Cancer Research UK Manchester Institute, University of Manchester, Manchester, UK
| | - Alexandra Clipson
- Cancer Research UK National Biomarker Centre, University of Manchester, Manchester, UK
| | - Kathryn L Simpson
- Cancer Research UK National Biomarker Centre, University of Manchester, Manchester, UK; Cancer Research UK Manchester Institute, University of Manchester, Manchester, UK
| | - Fiona Blackhall
- Department of Medical Oncology, The Christie NHS Foundation Trust, Manchester, UK; Faculty of Biology, Medicine, and Health, University of Manchester, Manchester, UK
| | - Dominic G Rothwell
- Cancer Research UK National Biomarker Centre, University of Manchester, Manchester, UK
| | - Caroline Dive
- Cancer Research UK National Biomarker Centre, University of Manchester, Manchester, UK; Cancer Research UK Manchester Institute, University of Manchester, Manchester, UK
| | - Florent Mouliere
- Cancer Research UK National Biomarker Centre, University of Manchester, Manchester, UK.
| |
Collapse
|
32
|
Zhang W, Ye B, Song Y, Yang P, Si W, Jing H, Yang F, Yuan D, Wu Z, Lyu J, Peng K, Zhang X, Wang L, Li Y, Liu Y, Wu C, Hao X, Zhang Y, Qi W, Wang J, Dong F, Zhao Z, Jing H, Li Y. Integrating multi-omics features enables non-invasive early diagnosis and treatment response prediction of diffuse large B-cell lymphoma. Clin Transl Med 2025; 15:e70174. [PMID: 39776291 PMCID: PMC11705727 DOI: 10.1002/ctm2.70174] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2024] [Revised: 11/13/2024] [Accepted: 12/28/2024] [Indexed: 01/11/2025] Open
Abstract
BACKGROUND Multi-omics features of cell-free DNA (cfDNA) can effectively improve the performance of non-invasive early diagnosis and prognosis of cancer. However, multimodal characterization of cfDNA remains technically challenging. METHODS We developed a comprehensive multi-omics solution (COMOS) to specifically obtain an extensive fragmentomics landscape, presented by breakpoint characteristics of nucleosomes, CpG islands, DNase clusters and enhancers, besides typical methylation, copy number alteration of cfDNA. The COMOS was tested on 214 plasma samples of diffuse large B-cell lymphoma (DLBCL) and matched healthy controls. RESULTS For early diagnosis, COMOS improved the area under the curve (AUC) value to .993 compared with the individual omics model, with a sensitivity of 95% at 98% specificity. Detection sensitivity achieved 91% at 99% specificity in early-stage patients, while the AUC values of the individual omics model were 0.942, 0.968, 0.989, 0.935, 0.921, 0.781 and 0.917, respectively, with lower sensitivity and specificity. In the treatment response cohort, COMOS yielded a superior sensitivity of 88% at 86% specificity (AUC, 0.903). COMOS has achieved excellent performance in early diagnosis and treatment response prediction. CONCLUSIONS Our study provides an effectively improved approach with high accuracy for the diagnosis and prognosis of DLBCL, showing great potential for future clinical application. KEY POINTS A comprehensive multi-omics solution to specifically obtain an extensive fragmentomics landscape, presented by breakpoint characteristics of nucleosomes, CpG islands, DNase clusters and enhancers, besides typical methylation, copy number alteration of cfDNA. Integrated model of cfDNA multi-omics could be used for non-invasive early diagnosis of DLBCL. Integrated model of cfDNA multi-omics could effectively evaluate the efficacy of R-CHOP before DLBCL treatment.
Collapse
MESH Headings
- Humans
- Lymphoma, Large B-Cell, Diffuse/diagnosis
- Lymphoma, Large B-Cell, Diffuse/genetics
- Lymphoma, Large B-Cell, Diffuse/drug therapy
- Lymphoma, Large B-Cell, Diffuse/blood
- Female
- Male
- Middle Aged
- Aged
- Adult
- Early Detection of Cancer/methods
- Prognosis
- Cell-Free Nucleic Acids/blood
- Cell-Free Nucleic Acids/analysis
- Rituximab/therapeutic use
- Biomarkers, Tumor/blood
- Biomarkers, Tumor/genetics
- Doxorubicin/therapeutic use
- Early Diagnosis
- Cyclophosphamide/therapeutic use
- Multiomics
Collapse
Affiliation(s)
- Weilong Zhang
- Department of HematologyLymphoma Research CenterPeking University Third HospitalBeijingChina
| | | | - Yang Song
- BOE Technology Group Co., LtdBeijingChina
| | - Ping Yang
- Department of HematologyLymphoma Research CenterPeking University Third HospitalBeijingChina
| | - Wenzhe Si
- Department of Laboratory MedicinePeking University Third HospitalBeijingChina
| | | | - Fan Yang
- BOE Technology Group Co., LtdBeijingChina
| | - Dan Yuan
- BOE Technology Group Co., LtdBeijingChina
| | - Zhihong Wu
- BOE Technology Group Co., LtdBeijingChina
| | - Jiahao Lyu
- BOE Technology Group Co., LtdBeijingChina
| | - Kang Peng
- BOE Technology Group Co., LtdBeijingChina
| | - Xu Zhang
- Department of HematologyLymphoma Research CenterPeking University Third HospitalBeijingChina
| | - Lingli Wang
- Department of HematologyLymphoma Research CenterPeking University Third HospitalBeijingChina
| | - Yan Li
- Department of HematologyLymphoma Research CenterPeking University Third HospitalBeijingChina
| | - Yan Liu
- Department of HematologyLymphoma Research CenterPeking University Third HospitalBeijingChina
| | - Chaoling Wu
- Department of HematologyLymphoma Research CenterPeking University Third HospitalBeijingChina
| | - Xiaoyu Hao
- Department of HematologyLymphoma Research CenterPeking University Third HospitalBeijingChina
| | - Yuqi Zhang
- Department of HematologyLymphoma Research CenterPeking University Third HospitalBeijingChina
| | - Wenxin Qi
- Department of HematologyLymphoma Research CenterPeking University Third HospitalBeijingChina
| | - Jing Wang
- Department of HematologyLymphoma Research CenterPeking University Third HospitalBeijingChina
| | - Fei Dong
- Department of HematologyLymphoma Research CenterPeking University Third HospitalBeijingChina
| | | | - Hongmei Jing
- Department of HematologyLymphoma Research CenterPeking University Third HospitalBeijingChina
| | - Yanzhao Li
- BOE Technology Group Co., LtdBeijingChina
| |
Collapse
|
33
|
Wang D, Liu L, Chi W, Liu Z, Wu J, Liang Y, He F, Zhang R, Huang P, Li Y, Qiu G. Interfacial cfDNA Enrichment and Amplification with On-Chip Thermoplasmonics for Highly Sensitive Cancerous Liquid Biopsy. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2409708. [PMID: 39630008 PMCID: PMC11789577 DOI: 10.1002/advs.202409708] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/15/2024] [Revised: 11/14/2024] [Indexed: 01/30/2025]
Abstract
Tumor-derived cell-free DNA (cfDNA) has been exploited as an effective liquid biopsy biomarker for early cancer diagnosis. However, the fragmented and low-abundance nature in circulating blood pose challenges for highly sensitive cfDNA quantification. Herein, a multifunctional plasmonic biosensor termed Interfacial cfDNA Enrichment, Amplification and Sensing with on-chip Thermoplasmonics (INEAST) is developed for cfDNA-based liquid biopsy and lung cancer diagnosis. The INEAST biosensor achieved in situ thermoregulation and label-free cfDNA biosensing by simultaneously harnessing interfacial thermoplasmonics and localized surface plasmon resonance. Typical cfDNA biomarkers, including epidermal growth factor receptor (EGFR), tumor protein 53 (TP53), phosphatase and tensin homologue deleted on chromosome 10 (PTEN), and cyclin-dependent kinase inhibitor (CDKN2A), are quantified with detection limits down to femtomolar-level. Through further validation using blood samples from lung cancer patients, the proposed INEAST bioassays demonstrated superior reliability for lung cancer screening, particularly when combined with clinically available tumor-protein metrics. This study demonstrated that the INEAST biosensor enables rapid and sensitive cfDNA quantification, yielding a promising and compatible liquid biopsy for early-stage lung cancer diagnosis.
Collapse
Affiliation(s)
- Danhua Wang
- Institute of Medical Robotics, School of Biomedical EngineeringShanghai Jiao Tong UniversityShanghai200240China
| | - Linlin Liu
- Institute of Medical Robotics, School of Biomedical EngineeringShanghai Jiao Tong UniversityShanghai200240China
| | - Wenjing Chi
- Department of Laboratory MedicineHuadong Hospital Affiliated to Fudan UniversityShanghai200031China
| | - Zhenping Liu
- The First People's Hospital of Linping DistrictHangzhouZhejiang Province311100China
| | - Jiayun Wu
- Institute of Medical Robotics, School of Biomedical EngineeringShanghai Jiao Tong UniversityShanghai200240China
| | - Yirou Liang
- Institute of Medical Robotics, School of Biomedical EngineeringShanghai Jiao Tong UniversityShanghai200240China
| | - Fei He
- Institute of Medical Robotics, School of Biomedical EngineeringShanghai Jiao Tong UniversityShanghai200240China
| | - Ruixiang Zhang
- Institute of Medical Robotics, School of Biomedical EngineeringShanghai Jiao Tong UniversityShanghai200240China
| | - Pengxin Huang
- Institute of Medical Robotics, School of Biomedical EngineeringShanghai Jiao Tong UniversityShanghai200240China
| | - Yunbo Li
- Institute of Medical Robotics, School of Biomedical EngineeringShanghai Jiao Tong UniversityShanghai200240China
| | - Guangyu Qiu
- Institute of Medical Robotics, School of Biomedical EngineeringShanghai Jiao Tong UniversityShanghai200240China
| |
Collapse
|
34
|
Xu F, Wang C, Li H, Yu B, Chang L, Wang F, Long C, Bai L, Zhao H, Che N. Evaluation of cfDNA fragmentation characteristics in plasma for the diagnosis of lung cancer: A prospective cohort study. Cancer Sci 2025; 116:248-256. [PMID: 39466000 PMCID: PMC11711045 DOI: 10.1111/cas.16360] [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: 06/19/2024] [Revised: 09/12/2024] [Accepted: 09/15/2024] [Indexed: 10/29/2024] Open
Abstract
Lung cancer is one of the most prevalent cancers worldwide, yet only approximately 16% of patients are diagnosed in early stage, highlighting the urgent need for novel, highly accurate detection models. In our study, patients with suspected lung cancer or lung disease, as identified through radiographic imaging, along with healthy individuals, were consecutively recruited from Beijing Chest Hospital. Circulating free DNA (cfDNA) was extracted from plasma samples, and low-depth whole-genome sequencing was performed to identify fragmentomic features for model construction. A total of 265 participants were prospectively enrolled, comprising 124 lung cancer patients and 141 noncancer individuals. The model we developed was based on four cfDNA fragmentation characteristics, including 20-bp breakpoint nucleotides motif, fragmentation size coverage, fragmentation size distribution, and copy number variation. In an independent test cohort, the model achieved an area under the curve (AUC) of 0.861 (95% CI: 0.781-0.942) and demonstrated a sensitivity of 70% (95% CI: 53.5%-83.4%) at a specificity of 89.4% (95% CI: 76.9%-96.5%). Notably, the model was also effective in detecting early-stage cancer, with an AUC of 0.808 (95% CI: 0.69-0.925). In summary, our lung cancer detection model shows strong screening capabilities by leveraging four cfDNA fragmentation characteristics, exhibiting robust performance in early cancer diagnosis.
Collapse
Affiliation(s)
- Fudong Xu
- Department of Pathology, Beijing Chest HospitalCapital Medical UniversityBeijingChina
- Beijing Tuberculosis & Thoracic Tumor Research InstituteBeijingChina
| | - Chong Wang
- Thoracic Minimally Invasive Treatment Center, Beijing Chest HospitalCapital Medical UniversityBeijingChina
| | - Hongxia Li
- Department of Medical Oncology, Beijing Chest HospitalCapital Medical UniversityBeijingChina
| | - Bo Yu
- Department of MedicineBeijing USCI Medical LaboratoryBeijingChina
| | - Luyuan Chang
- Department of MedicineBeijing USCI Medical LaboratoryBeijingChina
| | - Feng Wang
- Thoracic Minimally Invasive Treatment Center, Beijing Chest HospitalCapital Medical UniversityBeijingChina
| | - Chaolian Long
- Department of Pathology, Beijing Chest HospitalCapital Medical UniversityBeijingChina
- Beijing Tuberculosis & Thoracic Tumor Research InstituteBeijingChina
| | - Ling Bai
- Department of MedicineBeijing USCI Medical LaboratoryBeijingChina
| | - Hanqing Zhao
- Department of MedicineBeijing USCI Medical LaboratoryBeijingChina
| | - Nanying Che
- Department of Pathology, Beijing Chest HospitalCapital Medical UniversityBeijingChina
- Beijing Tuberculosis & Thoracic Tumor Research InstituteBeijingChina
| |
Collapse
|
35
|
Liu D, Yehia L, Dhawan A, Ni Y, Eng C. Protocol for analyzing plasma cell-free DNA fragment end motifs from ultra-low-pass whole-genome sequencing. STAR Protoc 2024; 5:103357. [PMID: 39368093 PMCID: PMC11489062 DOI: 10.1016/j.xpro.2024.103357] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2024] [Revised: 08/15/2024] [Accepted: 09/11/2024] [Indexed: 10/07/2024] Open
Abstract
Circulating cell-free DNA (cfDNA) fragment end motif profiles are a promising biomarker in precision oncology. Here, we present a protocol for analyzing plasma cfDNA fragment end motifs from ultra-low-pass whole-genome sequencing (WGS) data. We detail a pipeline composed of sequential bash scripts for processing post-alignment BAM files. Subsequently, we outline the procedure for downstream analysis and visualization of 4-mer as well as other n-mer cfDNA end motifs in R. For complete details on the use and execution of this protocol, please refer to Liu et al.1.
Collapse
Affiliation(s)
- Darren Liu
- Genomic Medicine Institute, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195, USA; Cleveland Clinic Lerner College of Medicine of Case Western Reserve University, Cleveland, OH 44195, USA; Department of Internal Medicine, Cleveland Clinic, Cleveland, OH 44195, USA.
| | - Lamis Yehia
- Genomic Medicine Institute, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195, USA
| | - Andrew Dhawan
- Genomic Medicine Institute, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195, USA; Rose Ella Burkhardt Brain Tumor and Neuro-Oncology Center, Cleveland Clinic, Cleveland, OH 44195, USA; Center for Personalized Genetic Healthcare, Medical Specialties Institute, Cleveland Clinic, Cleveland, OH 44195, USA
| | - Ying Ni
- Cleveland Clinic Lerner College of Medicine of Case Western Reserve University, Cleveland, OH 44195, USA; Center for Immunotherapy and Precision Immuno-oncology, Cleveland Clinic, Cleveland, OH 44195, USA
| | - Charis Eng
- Genomic Medicine Institute, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195, USA; Cleveland Clinic Lerner College of Medicine of Case Western Reserve University, Cleveland, OH 44195, USA; Center for Personalized Genetic Healthcare, Medical Specialties Institute, Cleveland Clinic, Cleveland, OH 44195, USA; Taussig Cancer Institute, Cleveland Clinic, Cleveland, OH 44195, USA; Department of Genetics and Genome Sciences, Case Western Reserve University School of Medicine, Cleveland, OH 44106, USA; Germline High Risk Cancer Focus Group, Case Comprehensive Cancer Center, Case Western Reserve University, Cleveland, OH 44106, USA
| |
Collapse
|
36
|
Costa J, Membrino A, Zanchetta C, Rizzato S, Cortiula F, Rossetto C, Pelizzari G, Aprile G, Macerelli M. The Role of ctDNA in the Management of Non-Small-Cell Lung Cancer in the AI and NGS Era. Int J Mol Sci 2024; 25:13669. [PMID: 39769431 PMCID: PMC11727717 DOI: 10.3390/ijms252413669] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2024] [Revised: 12/18/2024] [Accepted: 12/19/2024] [Indexed: 01/11/2025] Open
Abstract
Liquid biopsy (LB) involves the analysis of circulating tumour-derived DNA (ctDNA), providing a minimally invasive method for gathering both quantitative and qualitative information. Genomic analysis of ctDNA through next-generation sequencing (NGS) enables comprehensive genetic profiling of tumours, including non-driver alterations that offer prognostic insights. LB can be applied in both early-stage disease settings, for the diagnosis and monitoring of minimal residual disease (MRD), and advanced disease settings, for monitoring treatment response and understanding the mechanisms behind disease progression and tumour heterogeneity. Currently, LB has limited use in clinical practice, primarily due to its significant costs, limited diagnostic yield, and uncertain prognostic role. The application of artificial intelligence (AI) in the medical field is a promising approach to processing extensive information and applying it to individual cases to enhance therapeutic decision-making and refine risk assessment.
Collapse
Affiliation(s)
- Jacopo Costa
- Department of Medicine (DAME), University of Udine, 33100 Udine, Italy; (A.M.); (C.Z.)
- Department of Oncology, University Hospital of Udine, 33100 Udine, Italy; (S.R.); (F.C.); (C.R.); (G.P.); (G.A.); (M.M.)
| | - Alexandro Membrino
- Department of Medicine (DAME), University of Udine, 33100 Udine, Italy; (A.M.); (C.Z.)
- Department of Oncology, University Hospital of Udine, 33100 Udine, Italy; (S.R.); (F.C.); (C.R.); (G.P.); (G.A.); (M.M.)
| | - Carol Zanchetta
- Department of Medicine (DAME), University of Udine, 33100 Udine, Italy; (A.M.); (C.Z.)
- Department of Oncology, University Hospital of Udine, 33100 Udine, Italy; (S.R.); (F.C.); (C.R.); (G.P.); (G.A.); (M.M.)
| | - Simona Rizzato
- Department of Oncology, University Hospital of Udine, 33100 Udine, Italy; (S.R.); (F.C.); (C.R.); (G.P.); (G.A.); (M.M.)
| | - Francesco Cortiula
- Department of Oncology, University Hospital of Udine, 33100 Udine, Italy; (S.R.); (F.C.); (C.R.); (G.P.); (G.A.); (M.M.)
- Department of Respiratory Medicine, Maastricht University Medical Centre, GROW School for Oncology and Reproduction, 6229 ER Maastricht, The Netherlands
| | - Ciro Rossetto
- Department of Oncology, University Hospital of Udine, 33100 Udine, Italy; (S.R.); (F.C.); (C.R.); (G.P.); (G.A.); (M.M.)
| | - Giacomo Pelizzari
- Department of Oncology, University Hospital of Udine, 33100 Udine, Italy; (S.R.); (F.C.); (C.R.); (G.P.); (G.A.); (M.M.)
| | - Giuseppe Aprile
- Department of Oncology, University Hospital of Udine, 33100 Udine, Italy; (S.R.); (F.C.); (C.R.); (G.P.); (G.A.); (M.M.)
| | - Marianna Macerelli
- Department of Oncology, University Hospital of Udine, 33100 Udine, Italy; (S.R.); (F.C.); (C.R.); (G.P.); (G.A.); (M.M.)
| |
Collapse
|
37
|
Lazzeri I, Spiegl BG, Hasenleithner SO, Speicher MR, Kircher M. LBFextract: Unveiling transcription factor dynamics from liquid biopsy data. Comput Struct Biotechnol J 2024; 23:3163-3174. [PMID: 39660220 PMCID: PMC11630664 DOI: 10.1016/j.csbj.2024.08.007] [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: 06/06/2024] [Revised: 08/09/2024] [Accepted: 08/09/2024] [Indexed: 12/12/2024] Open
Abstract
Motivation The analysis of circulating cell-free DNA (cfDNA) holds immense promise as a non-invasive diagnostic tool across various human conditions. However, extracting biological insights from cfDNA fragments entails navigating complex and diverse bioinformatics methods, encompassing not only DNA sequence variation, but also epigenetic characteristics like nucleosome footprints, fragment length, and methylation patterns. Results We introduce Liquid Biopsy Feature extract (LBFextract), a comprehensive package designed to streamline feature extraction from cfDNA sequencing data, with the aim of enhancing the reproducibility and comparability of liquid biopsy studies. LBFextract facilitates the integration of preprocessing and postprocessing steps through alignment fragment tags and a hook mechanism. It incorporates various methods, including coverage-based and fragment length-based approaches, alongside two novel feature extraction methods: an entropy-based method to infer TF activity from fragmentomics data and a technique to amplify signals from nucleosome dyads. Additionally, it implements a method to extract condition-specific differentially active TFs based on these features for biomarker discovery. We demonstrate the use of LBFextract for the subtype classification of advanced prostate cancer patients using coverage signals at transcription factor binding sites from cfDNA. We show that LBFextract can generate robust and interpretable features that can discriminate between different clinical groups. LBFextract is a versatile and user-friendly package that can facilitate the analysis and interpretation of liquid biopsy data. Data and Code Availability and Implementation LBFextract is freely accessible at https://github.com/Isy89/LBF. It is implemented in Python and compatible with Linux and Mac operating systems. Code and data to reproduce these analyses have been uploaded to 10.5281/zenodo.10964406.
Collapse
Affiliation(s)
- Isaac Lazzeri
- Institute of Human Genetics, Diagnostic and Research Center for Molecular BioMedicine, Medical University of Graz, Neue Stiftingtalstr. 6, Graz 8010, Austria
| | - Benjamin Gernot Spiegl
- Institute of Human Genetics, Diagnostic and Research Center for Molecular BioMedicine, Medical University of Graz, Neue Stiftingtalstr. 6, Graz 8010, Austria
| | - Samantha O. Hasenleithner
- Division of Oncology, Department of Internal Medicine, Medical University of Graz, 8010 Graz, Austria
| | - Michael R. Speicher
- Institute of Human Genetics, Diagnostic and Research Center for Molecular BioMedicine, Medical University of Graz, Neue Stiftingtalstr. 6, Graz 8010, Austria
- BioTechMed-Graz, Graz, Austria
| | - Martin Kircher
- Berlin Institute of Health (BIH) at Charité – Universitätsmedizin Berlin, Berlin 10178, Germany
- Institute of Human Genetics, University Medical Center Schleswig-Holstein, University of Lübeck, Lübeck, Germany
| |
Collapse
|
38
|
Nafar S, Hosseini K, Shokrgozar N, Farahmandi AY, Alamdari-Palangi V, Saber Sichani A, Fallahi J. An Investigation into Cell-Free DNA in Different Common Cancers. Mol Biotechnol 2024; 66:3462-3474. [PMID: 38071680 DOI: 10.1007/s12033-023-00976-9] [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: 12/20/2022] [Accepted: 10/23/2023] [Indexed: 11/15/2024]
Abstract
Diagnosis is the most important step in different diseases, especially in cancers and blood malignancies. There are different methods in order to better diagnose of cancer, but many of them are invasive and also, some of them are not useful for immediate diagnosis. Cell-free DNA (cfDNA) or liquid biopsy easily accessible in peripheral blood is one of the non-invasive prognostic biomarkers in various areas of cancer management. In fact, amounts of cfDNA in serum or plasma can be used for diagnosis. In this review, we have considered some cancers such as hepatocellular carcinoma, lung cancer, breast cancer, and hematologic malignancies to compare the various methods of cfDNA diagnosis.
Collapse
Affiliation(s)
- Samira Nafar
- Medical Genetic Department, Shiraz University of Medical Science, Shiraz, Iran
| | - Kamran Hosseini
- Department of Molecular Medicine, School of Advanced Medical Sciences and Technologies, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Negin Shokrgozar
- Hematology Research Center, Shiraz University of Medical Science, Shiraz, Iran
| | | | - Vahab Alamdari-Palangi
- Department of Molecular Medicine, School of Advanced Medical Sciences and Technologies, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Ali Saber Sichani
- Department of Biology, Texas A&M University, College Station, TX, 77843, USA
| | - Jafar Fallahi
- Department of Molecular Medicine, School of Advanced Medical Sciences and Technologies, Shiraz University of Medical Sciences, Shiraz, Iran.
| |
Collapse
|
39
|
Gristina V, Russo G, Bazan Russo TD, Busuito G, Iannì G, Pisapia P, Scimone C, Palumbo L, Incorvaia L, Badalamenti G, Galvano A, Bazan V, Russo A, Troncone G, Malapelle U, Pepe F. Recent advances in the use of liquid biopsy for the diagnosis and treatment of lung cancer. Expert Rev Respir Med 2024; 18:991-1001. [PMID: 39491533 DOI: 10.1080/17476348.2024.2423824] [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: 09/03/2024] [Accepted: 10/28/2024] [Indexed: 11/05/2024]
Abstract
INTRODUCTION In the era of precision medicine, liquid biopsy rapidly emerges as an integrative diagnostic tool to successfully stratify solid tumor patients in accordance with molecular fingerprinting. As the matter of fact, a plethora of analytes may be isolated from liquid biosources supporting the potential application of liquid biopsy in several clinical scenarios. Despite this promising role, liquid biopsy is drastically affected by low abundance of analytes in biological matrix requiring highly sensitive technologies, trained personnel, and optimized diagnostic procedures to successfully administrate this revolutionary diagnostic tool in clinical practice. AREAS COVERED This review aims to investigate the recent advancements in technical approaches available to manage liquid biopsy samples, particularly focusing on their application in LC diagnosis and treatment. EXPERT OPINION The rapidly evolving scenario of liquid biopsy-based approaches is revolutionizing clinical administration of lung cancer patients. Of note, the integration of genomic, epigenomic, and transcriptomic markers lays the basis for 'comprehensive' molecular fingerprinting of lung cancer patients. Here, the next-generation technologies are fundamental in molecular profiling in diagnostic routine biofluids.
Collapse
Affiliation(s)
- Valerio Gristina
- Department of Precision Medicine in Medical, Surgical and Critical Care (Me.Pre.C.C.), University of Palermo, Palermo, Italy
| | - Gianluca Russo
- Department of Public Health, University Federico II of Naples, Naples, Italy
| | - Tancredi Didier Bazan Russo
- Department of Precision Medicine in Medical, Surgical and Critical Care (Me.Pre.C.C.), University of Palermo, Palermo, Italy
| | - Giulia Busuito
- Department of Precision Medicine in Medical, Surgical and Critical Care (Me.Pre.C.C.), University of Palermo, Palermo, Italy
| | - Giuliana Iannì
- Department of Precision Medicine in Medical, Surgical and Critical Care (Me.Pre.C.C.), University of Palermo, Palermo, Italy
| | - Pasquale Pisapia
- Department of Public Health, University Federico II of Naples, Naples, Italy
| | - Claudia Scimone
- Department of Public Health, University Federico II of Naples, Naples, Italy
| | - Lucia Palumbo
- Department of Public Health, University Federico II of Naples, Naples, Italy
| | - Lorena Incorvaia
- Department of Precision Medicine in Medical, Surgical and Critical Care (Me.Pre.C.C.), University of Palermo, Palermo, Italy
| | - Giuseppe Badalamenti
- Department of Precision Medicine in Medical, Surgical and Critical Care (Me.Pre.C.C.), University of Palermo, Palermo, Italy
| | - Antonio Galvano
- Department of Precision Medicine in Medical, Surgical and Critical Care (Me.Pre.C.C.), University of Palermo, Palermo, Italy
| | - Viviana Bazan
- Department of Biomedicine, Neuroscience and Advanced Diagnostic (Bi.N.D.), University of Palermo, Palermo, Italy
| | - Antonio Russo
- Department of Precision Medicine in Medical, Surgical and Critical Care (Me.Pre.C.C.), University of Palermo, Palermo, Italy
| | - Giancarlo Troncone
- Department of Public Health, University Federico II of Naples, Naples, Italy
| | - Umberto Malapelle
- Department of Public Health, University Federico II of Naples, Naples, Italy
| | - Francesco Pepe
- Department of Public Health, University Federico II of Naples, Naples, Italy
| |
Collapse
|
40
|
Liu Y, Peng F, Wang S, Jiao H, Dang M, Zhou K, Guo W, Guo S, Zhang H, Song W, Xing J. Aberrant fragmentomic features of circulating cell-free mitochondrial DNA as novel biomarkers for multi-cancer detection. EMBO Mol Med 2024; 16:3169-3183. [PMID: 39478151 PMCID: PMC11628560 DOI: 10.1038/s44321-024-00163-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2024] [Revised: 09/27/2024] [Accepted: 10/18/2024] [Indexed: 12/11/2024] Open
Abstract
Fragmentomic features of circulating cell free mitochondrial DNA (ccf-mtDNA) including fragmentation profile, 5' end base preference and motif diversity are poorly understood. Here, we generated ccf-mtDNA sequencing data of 1607 plasma samples using capture-based next generation sequencing. We firstly found that fragmentomic features of ccf-mtDNA were remarkably different from those of circulating cell free nuclear DNA. Furthermore, region-specific fragmentomic features of ccf-mtDNA were observed, which was associated with protein binding, base composition and special structure of mitochondrial DNA. When comparing to non-cancer controls, six types of cancer patients exhibited aberrant fragmentomic features. Then, cancer detection models were built based on the fragmentomic features. Both internal and external validation cohorts demonstrated the excellent capacity of our model in distinguishing cancer patients from non-cancer control, with all area under curve higher than 0.9322. The overall accuracy of tissue-of-origin was 89.24% and 87.92% for six cancer types in two validation cohort, respectively. Altogether, our study comprehensively describes cancer-specific fragmentomic features of ccf-mtDNA and provides a proof-of-principle for the ccf-mtDNA fragmentomics-based multi-cancer detection and tissue-of-origin classification.
Collapse
Affiliation(s)
- Yang Liu
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers and Department of Physiology and Pathophysiology, Fourth Military Medical University, Xi'an, China
- Department of Clinical Diagnosis, Tangdu Hospital, Fourth Military Medical University, Xi'an, China
| | - Fan Peng
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers and Department of Physiology and Pathophysiology, Fourth Military Medical University, Xi'an, China
| | - Siyuan Wang
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers and Department of Physiology and Pathophysiology, Fourth Military Medical University, Xi'an, China
| | - Huanmin Jiao
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers and Department of Physiology and Pathophysiology, Fourth Military Medical University, Xi'an, China
| | - Miao Dang
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers and Department of Physiology and Pathophysiology, Fourth Military Medical University, Xi'an, China
| | - Kaixiang Zhou
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers and Department of Physiology and Pathophysiology, Fourth Military Medical University, Xi'an, China
| | - Wenjie Guo
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers and Department of Physiology and Pathophysiology, Fourth Military Medical University, Xi'an, China
| | - Shanshan Guo
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers and Department of Physiology and Pathophysiology, Fourth Military Medical University, Xi'an, China
| | - Huanqin Zhang
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers and Department of Physiology and Pathophysiology, Fourth Military Medical University, Xi'an, China
| | - Wenjie Song
- Department of Hepatobiliary Surgery, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Jinliang Xing
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers and Department of Physiology and Pathophysiology, Fourth Military Medical University, Xi'an, China.
| |
Collapse
|
41
|
Jia H, Meng W, Gao R, Wang Y, Zhan C, Yu Y, Cong H, Yu L. Integrated SERS-Microfluidic Sensor Based on Nano-Micro Hierarchical Cactus-like Array Substrates for the Early Diagnosis of Prostate Cancer. BIOSENSORS 2024; 14:579. [PMID: 39727845 DOI: 10.3390/bios14120579] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/24/2024] [Revised: 11/19/2024] [Accepted: 11/26/2024] [Indexed: 12/28/2024]
Abstract
The detection and analysis of cancer cell exosomes with high sensitivity and precision are pivotal for the early diagnosis and treatment strategies of prostate cancer. To this end, a microfluidic chip, equipped with a cactus-like array substrate (CAS) based on surface-enhanced Raman spectroscopy (SERS) was designed and fabricated for the detection of exosome concentrations in Lymph Node Carcinoma of the Prostate (LNCaP). Double layers of polystyrene (PS) microspheres were self-assembled onto a polyethylene terephthalate (PET) film to form an ordered cactus-like nanoarray for detection and analysis. By combining EpCAM aptamer-labeled SERS nanoprobes and a CD63 aptamer-labeled CAS, a 'sandwich' structure was formed and applied to the microfluidic chips, further enhancing the Raman scattering signal of Raman reporter molecules. The results indicate that the integrated microfluidic sensor exhibits a good linear response within the detection concentration range of 105 particles μL-1 to 1 particle μL-1. The detection limit of exosomes in cancer cells can reach 1 particle μL-1. Therefore, we believed that the CAS integrated microfluidic sensor offers a superior solution for the early diagnosis and therapeutic intervention of prostate cancer.
Collapse
Affiliation(s)
- Huakun Jia
- State Key Laboratory of Chemical Safety, College of Control Science and Engineering, China University of Petroleum (East China), Qingdao 266580, China
| | - Weiyang Meng
- State Key Laboratory of Chemical Safety, College of Control Science and Engineering, China University of Petroleum (East China), Qingdao 266580, China
| | - Rongke Gao
- State Key Laboratory of Chemical Safety, College of Control Science and Engineering, China University of Petroleum (East China), Qingdao 266580, China
| | - Yeru Wang
- State Key Laboratory of Chemical Safety, College of Control Science and Engineering, China University of Petroleum (East China), Qingdao 266580, China
| | - Changbiao Zhan
- State Key Laboratory of Chemical Safety, College of Control Science and Engineering, China University of Petroleum (East China), Qingdao 266580, China
| | - Yiyue Yu
- State Key Laboratory of Chemical Safety, College of Control Science and Engineering, China University of Petroleum (East China), Qingdao 266580, China
| | - Haojie Cong
- State Key Laboratory of Chemical Safety, College of Control Science and Engineering, China University of Petroleum (East China), Qingdao 266580, China
| | - Liandong Yu
- State Key Laboratory of Chemical Safety, College of Control Science and Engineering, China University of Petroleum (East China), Qingdao 266580, China
| |
Collapse
|
42
|
Smyth R, Billatos E. Novel Strategies for Lung Cancer Interventional Diagnostics. J Clin Med 2024; 13:7207. [PMID: 39685665 DOI: 10.3390/jcm13237207] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2024] [Revised: 11/05/2024] [Accepted: 11/20/2024] [Indexed: 12/18/2024] Open
Abstract
Lung cancer is a major global health issue, with 2.21 million cases and 1.80 million deaths reported in 2020. It is the leading cause of cancer death worldwide. Most lung cancers have been linked to tobacco use, with changes in cigarette composition over the years contributing to shifts in cancer types and tumor locations within the lungs. Additionally, there is a growing incidence of lung cancer among never-smokers, particularly in East Asia, which is expected to increase the global burden of the disease. The classification of non-small cell lung cancer (NSCLC) into distinct subtypes is crucial for treatment efficacy and patient safety, especially as different subtypes respond differently to chemotherapy. For instance, certain chemotherapeutic agents are more effective for adenocarcinoma than for squamous carcinoma, which has led to the exclusion of squamous carcinoma from treatments like Bevacizumab due to safety concerns. This necessitates accurate histological diagnosis, which requires sufficient tissue samples from biopsies. However, acquiring adequate tissue is challenging due to the complex nature of lung tumors, patient comorbidities, and potential complications from biopsy procedures, such as bleeding, pneumothorax, and the purported risk of local recurrence. The need for improved diagnostic techniques has led to the development of advanced technologies like electromagnetic navigation bronchoscopy (ENB), radial endobronchial ultrasound (rEBUS), and robotic bronchoscopy. ENB and rEBUS have enhanced the accuracy and safety of lung biopsies, particularly for peripheral lesions, but both have limitations, such as the dependency on the presence of a bronchus sign. Robotic bronchoscopy, which builds on ENB, offers greater maneuverability and stability, improving diagnostic yields. Additionally, new imaging adjuncts, such as Cone Beam Computed Tomography (CBCT) and augmented fluoroscopy, further enhance the precision of these procedures by providing real-time, high-resolution imaging. These advancements are crucial as lung cancer is increasingly being detected at earlier stages due to screening programs, which require minimally invasive, accurate diagnostic methods to improve patient outcomes. This review aims to provide a comprehensive overview of the current challenges in lung cancer diagnostics and the innovative technological advancements in this rapidly evolving field, which represents an increasingly exciting career path for aspiring pulmonologists.
Collapse
Affiliation(s)
- Robert Smyth
- Department of Medicine, Section of Pulmonary, Critical Care and Occupational Medicine University of Iowa, Iowa City, IA 52242, USA
| | - Ehab Billatos
- Department of Medicine, Section of Pulmonary and Critical Care Medicine, Boston University, Boston, MA 02215, USA
- Department of Medicine, Section of Computational Biomedicine, Boston University, Boston, MA 02215, USA
| |
Collapse
|
43
|
Karimzadeh M, Momen-Roknabadi A, Cavazos TB, Fang Y, Chen NC, Multhaup M, Yen J, Ku J, Wang J, Zhao X, Murzynowski P, Wang K, Hanna R, Huang A, Corti D, Nguyen D, Lam T, Kilinc S, Arensdorf P, Chau KH, Hartwig A, Fish L, Li H, Behsaz B, Elemento O, Zou J, Hormozdiari F, Alipanahi B, Goodarzi H. Deep generative AI models analyzing circulating orphan non-coding RNAs enable detection of early-stage lung cancer. Nat Commun 2024; 15:10090. [PMID: 39572521 PMCID: PMC11582319 DOI: 10.1038/s41467-024-53851-9] [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: 05/21/2024] [Accepted: 10/22/2024] [Indexed: 11/24/2024] Open
Abstract
Liquid biopsies have the potential to revolutionize cancer care through non-invasive early detection of tumors. Developing a robust liquid biopsy test requires collecting high-dimensional data from a large number of blood samples across heterogeneous groups of patients. We propose that the generative capability of variational auto-encoders enables learning a robust and generalizable signature of blood-based biomarkers. In this study, we analyze orphan non-coding RNAs (oncRNAs) from serum samples of 1050 individuals diagnosed with non-small cell lung cancer (NSCLC) at various stages, as well as sex-, age-, and BMI-matched controls. We demonstrate that our multi-task generative AI model, Orion, surpasses commonly used methods in both overall performance and generalizability to held-out datasets. Orion achieves an overall sensitivity of 94% (95% CI: 87%-98%) at 87% (95% CI: 81%-93%) specificity for cancer detection across all stages, outperforming the sensitivity of other methods on held-out validation datasets by more than ~ 30%.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | - Ti Lam
- Exai Bio Inc., Palo Alto, CA, US
| | | | | | | | | | | | - Helen Li
- Exai Bio Inc., Palo Alto, CA, US
| | | | | | - James Zou
- Stanford University, Stanford, CA, US
| | | | | | - Hani Goodarzi
- University of California, San Francisco, CA, US.
- Arc Institute, Palo Alto, CA, US.
| |
Collapse
|
44
|
Xue R, Li X, Yang L, Yang M, Zhang B, Zhang X, Li L, Duan X, Yan R, He X, Cui F, Wang L, Wang X, Wu M, Zhang C, Zhao J. Evaluation and integration of cell-free DNA signatures for detection of lung cancer. Cancer Lett 2024; 604:217216. [PMID: 39233043 DOI: 10.1016/j.canlet.2024.217216] [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: 05/29/2024] [Revised: 08/29/2024] [Accepted: 08/30/2024] [Indexed: 09/06/2024]
Abstract
Cell-free DNA (cfDNA) analysis has shown potential in detecting early-stage lung cancer based on non-genetic features. To distinguish patients with lung cancer from healthy individuals, peripheral blood were collected from 926 lung cancer patients and 611 healthy individuals followed by cfDNA extraction. Low-pass whole genome sequencing and targeted methylation sequencing were conducted and various features of cfDNA were evaluated. With our customized algorithm using the most optimal features, the ensemble stacked model was constructed, called ESim-seq (Early Screening tech with Integrated Model). In the independent validation cohort, the ESim-seq model achieved an area under the curve (AUC) of 0.948 (95 % CI: 0.915-0.981), with a sensitivity of 79.3 % (95 % CI: 71.5-87.0 %) across all stages at a specificity of 96.0 % (95 % CI: 90.6-100.0 %). Specifically, the sensitivity of the ESim-seq model was 76.5 % (95 % CI: 67.3-85.8 %) in stage I patients, 100 % (95 % CI: 100.0-100.0 %) in stage II patients, 100 % (95 % CI: 100.0-100.0 %) in stage III patients and 87.5 % (95 % CI: 64.6%-100.0 %) in stage IV patients in the independent validation cohort. Besides, we constructed LCSC model (Lung Cancer Subtype multiple Classification), which was able to accurately distinguish patients with small cell lung cancer from those with non-small cell lung cancer, achieving an AUC of 0.961 (95 % CI: 0.949-0.957). The present study has established a framework for assessing cfDNA features and demonstrated the benefits of integrating multiple features for early detection of lung cancer.
Collapse
Affiliation(s)
- Ruyue Xue
- Internet Medical and System Applications of National Engineering Laboratory, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Xiaomin Li
- Jiangsu Simcere Diagnostics Co., Ltd., Nanjing Simcere Medical Laboratory Science Co., Ltd., The State Key Laboratory of Neurology and Oncology Drug Development, Nanjing, China; Cancer Center, The First Affiliated Hospital of Jinzhou Medical University, Jinzhou, China
| | - Lu Yang
- Jiangsu Simcere Diagnostics Co., Ltd., Nanjing Simcere Medical Laboratory Science Co., Ltd., The State Key Laboratory of Neurology and Oncology Drug Development, Nanjing, China; Cancer Center, The First Affiliated Hospital of Jinzhou Medical University, Jinzhou, China
| | - Meijia Yang
- Internet Medical and System Applications of National Engineering Laboratory, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Bei Zhang
- Jiangsu Simcere Diagnostics Co., Ltd., Nanjing Simcere Medical Laboratory Science Co., Ltd., The State Key Laboratory of Neurology and Oncology Drug Development, Nanjing, China
| | - Xu Zhang
- Internet Medical and System Applications of National Engineering Laboratory, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Lifeng Li
- Internet Medical and System Applications of National Engineering Laboratory, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Xiaoran Duan
- Internet Medical and System Applications of National Engineering Laboratory, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Rui Yan
- Internet Medical and System Applications of National Engineering Laboratory, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Xianying He
- Internet Medical and System Applications of National Engineering Laboratory, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Fangfang Cui
- Internet Medical and System Applications of National Engineering Laboratory, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Linlin Wang
- Internet Medical and System Applications of National Engineering Laboratory, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Xiaoqiang Wang
- Jiangsu Simcere Diagnostics Co., Ltd., Nanjing Simcere Medical Laboratory Science Co., Ltd., The State Key Laboratory of Neurology and Oncology Drug Development, Nanjing, China
| | - Mengsi Wu
- Jiangsu Simcere Diagnostics Co., Ltd., Nanjing Simcere Medical Laboratory Science Co., Ltd., The State Key Laboratory of Neurology and Oncology Drug Development, Nanjing, China
| | - Chao Zhang
- Jiangsu Simcere Diagnostics Co., Ltd., Nanjing Simcere Medical Laboratory Science Co., Ltd., The State Key Laboratory of Neurology and Oncology Drug Development, Nanjing, China
| | - Jie Zhao
- Internet Medical and System Applications of National Engineering Laboratory, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China.
| |
Collapse
|
45
|
Wang S, Meng F, Chen P, Lv Y, Wu M, Tang H, Bao H, Wu X, Shao Y, Wang J, Dai J, Xu L, Wang X, Yin R. Cell-free DNA assay for malignancy classification of high-risk lung nodules. J Thorac Cardiovasc Surg 2024; 168:e140-e175. [PMID: 38670484 DOI: 10.1016/j.jtcvs.2024.04.026] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/08/2023] [Revised: 03/18/2024] [Accepted: 04/15/2024] [Indexed: 04/28/2024]
Abstract
OBJECTIVE Although low-dose computed tomography has been proven effective to reduce lung cancer-specific mortality, a considerable proportion of surgically resected high-risk lung nodules were still confirmed pathologically benign. There is an unmet need of a novel method for malignancy classification in lung nodules. METHODS We recruited 307 patients with high-risk lung nodules who underwent curative surgery, and 247 and 60 cases were pathologically confirmed malignant and benign lung lesions, respectively. Plasma samples from each patient were collected before surgery and performed low-depth (5×) whole-genome sequencing. We extracted cell-free DNA characteristics and determined radiomic features. We built models to classify the malignancy using our data and further validated models with 2 independent lung nodule cohorts. RESULTS Our models using one type of profile were able to distinguish lung cancer and benign lung nodules at an area under the curve metrics of 0.69 to 0.91 in the study cohort. Integrating all the 5 base models using cell-free DNA profiles, the cell-free DNA-based ensemble model achieved an area under the curve of 0.95 (95% CI, 0.92-0.97) in the study cohort and 0.98 (95% CI, 0.96-1.00) in the validation cohort. At a specificity of 95.0%, the sensitivity reached 80.0% in the study cohort. With the same threshold, the specificity and sensitivity had similar performances in both validation cohorts. Furthermore, the performance of area under the curve reached 0.97 in both the study and validation cohorts when considering the radiomic profile. CONCLUSIONS The cell-free DNA profiles-based method is an efficient noninvasive tool to distinguish malignancies and high-risk but pathologically benign lung nodules.
Collapse
Affiliation(s)
- Siwei Wang
- Department of Thoracic Surgery, Jiangsu Key Laboratory of Molecular and Translational Cancer Research, Nanjing Medical University Affiliated Cancer Hospital & Jiangsu Cancer Hospital & Jiangsu Institute of Cancer Research, Nanjing, Jiangsu, China; Clinical Research Institute of Traditional Chinese Medicine, Jiangsu Province Hospital of Chinese Medicine, The Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, Jiangsu, China
| | - Fanchen Meng
- Department of Thoracic Surgery, Jiangsu Key Laboratory of Molecular and Translational Cancer Research, Nanjing Medical University Affiliated Cancer Hospital & Jiangsu Cancer Hospital & Jiangsu Institute of Cancer Research, Nanjing, Jiangsu, China
| | - Peng Chen
- Department of Thoracic Surgery, Jiangsu Key Laboratory of Molecular and Translational Cancer Research, Nanjing Medical University Affiliated Cancer Hospital & Jiangsu Cancer Hospital & Jiangsu Institute of Cancer Research, Nanjing, Jiangsu, China
| | - Yang Lv
- Department of Information Center, The Affiliated Jiangning Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Min Wu
- Geneseeq Research Institute, Nanjing Geneseeq Technology Inc, Nanjing, Jiangsu, China
| | - Haimeng Tang
- Geneseeq Research Institute, Nanjing Geneseeq Technology Inc, Nanjing, Jiangsu, China
| | - Hua Bao
- Geneseeq Research Institute, Nanjing Geneseeq Technology Inc, Nanjing, Jiangsu, China
| | - Xue Wu
- Geneseeq Research Institute, Nanjing Geneseeq Technology Inc, Nanjing, Jiangsu, China
| | - Yang Shao
- Geneseeq Research Institute, Nanjing Geneseeq Technology Inc, Nanjing, Jiangsu, China; School of Public Health, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Jie Wang
- Department of Science and Technology, Nanjing Medical University Affiliated Cancer Hospital & Jiangsu Cancer Hospital & Jiangsu Institute of Cancer Research, Nanjing, Jiangsu, China; Biobank of Lung Cancer, Jiangsu Biobank of Clinical Resources, Nanjing, Jiangsu, China
| | - Juncheng Dai
- School of Public Health, Nanjing Medical University, Nanjing, Jiangsu, China; Collaborative Innovation Center for Cancer Personalized Medicine, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Lin Xu
- Department of Thoracic Surgery, Jiangsu Key Laboratory of Molecular and Translational Cancer Research, Nanjing Medical University Affiliated Cancer Hospital & Jiangsu Cancer Hospital & Jiangsu Institute of Cancer Research, Nanjing, Jiangsu, China; Collaborative Innovation Center for Cancer Personalized Medicine, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Xiaoxiao Wang
- Clinical Research Institute of Traditional Chinese Medicine, Jiangsu Province Hospital of Chinese Medicine, The Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, Jiangsu, China.
| | - Rong Yin
- Department of Thoracic Surgery, Jiangsu Key Laboratory of Molecular and Translational Cancer Research, Nanjing Medical University Affiliated Cancer Hospital & Jiangsu Cancer Hospital & Jiangsu Institute of Cancer Research, Nanjing, Jiangsu, China; Department of Science and Technology, Nanjing Medical University Affiliated Cancer Hospital & Jiangsu Cancer Hospital & Jiangsu Institute of Cancer Research, Nanjing, Jiangsu, China; Biobank of Lung Cancer, Jiangsu Biobank of Clinical Resources, Nanjing, Jiangsu, China; Collaborative Innovation Center for Cancer Personalized Medicine, Nanjing Medical University, Nanjing, Jiangsu, China.
| |
Collapse
|
46
|
Peng T, Zhang H, Li L, Cao C, Xu M, Liu X, Lin S, Wu P, Chu T, Liu B, Xu Y, Zhang Y, Wang Y, Yu J, Ding W, Jin X, Wu P. Plasma Cell-Free DNA Concentration and Fragmentomes Predict Neoadjuvant Chemotherapy Response in Cervical Cancer Patients. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2309422. [PMID: 39319610 DOI: 10.1002/advs.202309422] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/05/2023] [Revised: 05/18/2024] [Indexed: 09/26/2024]
Abstract
Cervical cancer remains one of the most lethal gynecological malignancies. However, biomarkers for more precise patient care are an unmet need. Herein, the concentration of 285 plasma cell-free DNA (cfDNA) samples are analyzed from 84 cervical patients and the clinical significance of cfDNA fragmentomic characteristics across the neoadjuvant chemotherapy (NACT) treatment. Patients with poor NACT response exhibit a significantly greater escalation in cfDNA levels following the initial cycle of treatment, in comparison to patients with a favorable response. Distinctive end motif profiles and promoter coverages of cfDNA in initial plasma are observed between patients with differing NACT responses. Notably, the DNASE1L3 analysis further demonstrates the intrinsic association between cfDNA characteristics and chemotherapy resistance. The cfDNA and motif ratios show a good discriminative capacity for predicting non-responders from responders (area under the curve (AUC) > 0.8). In addition, transcriptional start sites (TSS) coverages around promoters discern the alteration of biological processes associated with chemotherapy resistance and reflect the potential value in predicting chemotherapy response. These findings in predictive biomarkers may optimize treatment selection, minimize unnecessary treatment, and assist in establishing personalized treatment strategies for cervical cancer patients.
Collapse
Affiliation(s)
- Ting Peng
- Department of Obstetrics and Gynecology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430000, China
- National Clinical Research Center for Gynecology and Obstetrics, Cancer Biology Research Center (Key Laboratory of the Ministry of Education), Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430000, China
| | | | - Lingguo Li
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Canhui Cao
- National Clinical Research Center for Gynecology and Obstetrics, Cancer Biology Research Center (Key Laboratory of the Ministry of Education), Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430000, China
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430000, China
| | - Miaochun Xu
- Department of Obstetrics and Gynecology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430000, China
- National Clinical Research Center for Gynecology and Obstetrics, Cancer Biology Research Center (Key Laboratory of the Ministry of Education), Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430000, China
| | - Xiaojie Liu
- Department of Obstetrics and Gynecology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430000, China
- National Clinical Research Center for Gynecology and Obstetrics, Cancer Biology Research Center (Key Laboratory of the Ministry of Education), Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430000, China
| | - Shitong Lin
- Department of Obstetrics and Gynecology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430000, China
- National Clinical Research Center for Gynecology and Obstetrics, Cancer Biology Research Center (Key Laboratory of the Ministry of Education), Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430000, China
| | - Ping Wu
- National Clinical Research Center for Gynecology and Obstetrics, Cancer Biology Research Center (Key Laboratory of the Ministry of Education), Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430000, China
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430000, China
| | - Tian Chu
- National Clinical Research Center for Gynecology and Obstetrics, Cancer Biology Research Center (Key Laboratory of the Ministry of Education), Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430000, China
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430000, China
| | - Binghan Liu
- Department of Obstetrics and Gynecology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430000, China
- National Clinical Research Center for Gynecology and Obstetrics, Cancer Biology Research Center (Key Laboratory of the Ministry of Education), Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430000, China
| | - Yashi Xu
- Department of Obstetrics and Gynecology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430000, China
- National Clinical Research Center for Gynecology and Obstetrics, Cancer Biology Research Center (Key Laboratory of the Ministry of Education), Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430000, China
| | - Yan Zhang
- BGI Research, Shenzhen, 518083, China
| | | | - Jinjin Yu
- Department of Obstetrics and Gynecology, Affiliated Hospital of Jiangnan University, Wuxi Medical College, Jiangnan University, Wuxi, 214000, China
| | - Wencheng Ding
- National Clinical Research Center for Gynecology and Obstetrics, Cancer Biology Research Center (Key Laboratory of the Ministry of Education), Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430000, China
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430000, China
| | - Xin Jin
- BGI Research, Shenzhen, 518083, China
| | - Peng Wu
- Department of Obstetrics and Gynecology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430000, China
- National Clinical Research Center for Gynecology and Obstetrics, Cancer Biology Research Center (Key Laboratory of the Ministry of Education), Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430000, China
| |
Collapse
|
47
|
Li L, Sun Y. Circulating tumor DNA methylation detection as biomarker and its application in tumor liquid biopsy: advances and challenges. MedComm (Beijing) 2024; 5:e766. [PMID: 39525954 PMCID: PMC11550092 DOI: 10.1002/mco2.766] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2024] [Revised: 09/02/2024] [Accepted: 09/03/2024] [Indexed: 11/16/2024] Open
Abstract
Circulating tumor DNA (ctDNA) methylation, an innovative liquid biopsy biomarker, has emerged as a promising tool in early cancer diagnosis, monitoring, and prognosis prediction. As a noninvasive approach, liquid biopsy overcomes the limitations of traditional tissue biopsy. Among various biomarkers, ctDNA methylation has garnered significant attention due to its high specificity and early detection capability across diverse cancer types. Despite its immense potential, the clinical application of ctDNA methylation faces substantial challenges pertaining to sensitivity, specificity, and standardization. In this review, we begin by introducing the basic biology and common detection techniques of ctDNA methylation. We then explore recent advancements and the challenges faced in the clinical application of ctDNA methylation in liquid biopsies. This includes progress in early screening and diagnosis, identification of clinical molecular subtypes, monitoring of recurrence and minimal residual disease (MRD), prediction of treatment response and prognosis, assessment of tumor burden, and determination of tissue origin. Finally, we discuss the future perspectives and challenges of ctDNA methylation detection in clinical applications. This comprehensive overview underscores the vital role of ctDNA methylation in enhancing cancer diagnostic accuracy, personalizing treatments, and effectively monitoring disease progression, providing valuable insights for future research and clinical practice.
Collapse
Affiliation(s)
- Lingyu Li
- Central Laboratory & Shenzhen Key Laboratory of Epigenetics and Precision Medicine for CancersNational Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital & Shenzhen HospitalChinese Academy of Medical Sciences and Peking Union Medical CollegeShenzhenChina
| | - Yingli Sun
- Central Laboratory & Shenzhen Key Laboratory of Epigenetics and Precision Medicine for CancersNational Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital & Shenzhen HospitalChinese Academy of Medical Sciences and Peking Union Medical CollegeShenzhenChina
| |
Collapse
|
48
|
Boeri M, Sabia F, Ledda RE, Balbi M, Suatoni P, Segale M, Zanghì A, Cantarutti A, Rolli L, Valsecchi C, Corrao G, Marchianò A, Pastorino U, Sozzi G. Blood microRNA testing in participants with suspicious low-dose CT findings: follow-up of the BioMILD lung cancer screening trial. THE LANCET REGIONAL HEALTH. EUROPE 2024; 46:101070. [PMID: 39319217 PMCID: PMC11421266 DOI: 10.1016/j.lanepe.2024.101070] [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: 06/19/2024] [Revised: 08/22/2024] [Accepted: 08/30/2024] [Indexed: 09/26/2024]
Abstract
Background The proper management of suspicious radiologic findings is crucial to optimize the effectiveness of low-dose computed tomography (LDCT) lung cancer screening trials. In the BioMILD study, we evaluated the utility of combining a plasma 24-microRNA signature classifier (MSC) and LDCT to define the individual risk and personalize screening strategies. Here we aim to assess the utility of repeated MSC testing during annual screening rounds in 1024 participants with suspicious LDCT findings. Methods The primary outcome was two-year lung cancer incidence in relation to MSC test results, reported as relative risk (RR) with 95% confidence interval (CI). Lung cancer incidence and mortality were estimated using extended Cox models for time-dependent covariates, yielding the respective hazard ratios (HR). Clinicaltrials.gov ID: NCT02247453. Findings With a median follow-up of 8.5 years, the full study set included 1403 indeterminate LDCT (CTind) and 584 positive LDCT (CT+) results. A lung cancer RR increase in MSC+ compared to MSC- participants was observed in both the CTind (RR: 2.5; 95% CI: 1.4-4.32) and CT+ (RR: 2.6; 95% CI: 1.81-3.74) groups and was maintained when considering stage I or resectable tumors only. A 98% negative predictive value in CTind/MSC- and a 30% positive predictive value in CT+/MSC+ lesions were recorded. At seven years' follow-up, MSC+ participants had a cumulative HR of 4.4 (95% CI: 3.0-6.4) for lung cancer incidence and of 8.1 (95% CI: 2.7-24.5) for lung cancer mortality. Interpretation Our study shows that MSC can be reliably performed during LDCT screening rounds to increase the accuracy of lung cancer risk and mortality prediction and supports its clinical utility in the management of LDCT findings of uncertain malignancy. Funding Italian Association for Cancer Research; Italian Ministry of Health; Horizon2020; National Cancer Institute (NCI); Gensignia LifeScience.
Collapse
Affiliation(s)
- Mattia Boeri
- Unit of Epigenomics & Biomarkers of Solid Tumors, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, 20133, Italy
| | - Federica Sabia
- Unit of Thoracic Surgery, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, 20133, Italy
| | - Roberta E. Ledda
- Unit of Thoracic Surgery, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, 20133, Italy
- Department of Medicine and Surgery (DiMeC), Section of Radiology, Unit of Surgical Sciences, University of Parma, Parma, 43121, Italy
| | - Maurizio Balbi
- Unit of Thoracic Surgery, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, 20133, Italy
- Department of Oncology, Radiology Unit, San Luigi Gonzaga Hospital, University of Turin, Orbassano, 10043, Italy
| | - Paola Suatoni
- Unit of Thoracic Surgery, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, 20133, Italy
| | - Miriam Segale
- Unit of Epigenomics & Biomarkers of Solid Tumors, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, 20133, Italy
| | - Anna Zanghì
- Unit of Epigenomics & Biomarkers of Solid Tumors, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, 20133, Italy
| | - Anna Cantarutti
- Division of Biostatistics, Department of Statistics and Quantitative Methods, Epidemiology and Public Health, University of Milano-Bicocca, Milan, 20126, Italy
| | - Luigi Rolli
- Unit of Thoracic Surgery, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, 20133, Italy
| | - Camilla Valsecchi
- Unit of Thoracic Surgery, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, 20133, Italy
| | - Giovanni Corrao
- Division of Biostatistics, Department of Statistics and Quantitative Methods, Epidemiology and Public Health, University of Milano-Bicocca, Milan, 20126, Italy
| | - Alfonso Marchianò
- Department of Radiology, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, 20133, Italy
| | - Ugo Pastorino
- Unit of Thoracic Surgery, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, 20133, Italy
| | - Gabriella Sozzi
- Unit of Epigenomics & Biomarkers of Solid Tumors, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, 20133, Italy
| |
Collapse
|
49
|
Yan B, Chen Y, Wang Z, Li J, Wang R, Pan X, Li B, Li R. Analysis and identification of mRNAsi‑related expression signatures via RNA sequencing in lung cancer. Oncol Lett 2024; 28:549. [PMID: 39319211 PMCID: PMC11420643 DOI: 10.3892/ol.2024.14682] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2024] [Accepted: 08/15/2024] [Indexed: 09/26/2024] Open
Abstract
High stemness index scores are associated with poor survival in patients with lung cancer. Studies on the mRNA expression-based stemness index (mRNAsi) are typically conducted using tumor tissues; however, mRNAsi-related expression signatures based on cell-free RNA (cfRNA) are yet to be comprehensively investigated. The present study aimed to elucidate the gene expression profiles of tumor stemness in lung cancer tissues and corresponding cfRNAs in blood, and to assess their links with immune infiltration. Tumor tissue, paracancerous tissue, peripheral blood and lymph node samples were collected from patients with stage I-III non-small cell lung cancer and RNA sequencing was performed. The TCGAbiolinks package was used to calculate the mRNAsi for each of these four types of sample. Weighted gene co-expression network analysis and differentially expressed gene analyses were performed to investigate mRNAsi-related genes, and pathway enrichment analysis was performed using the Kyoto Encyclopedia of Genes and Genomes (KEGG) orthology-based annotation system. In addition, the STAR-Fusion tool was used to detect fusion variants, and CIBERSORT was used to analyze the correlations of stemness signatures in tissues and blood with immune cell infiltration. The mRNAsi values in peripheral blood and lymph nodes were found to be higher than those in cancer tissues. 'Hematopoietic cell lineage' was the only KEGG pathway enriched in mRNAsi-related genes in both lung cancer tissues and peripheral blood. In addition, the protein tyrosine phosphatase receptor type C associated protein gene was the only gene commonly associated with the mRNAsi in these two types of sample. The expression of mRNAsi-related genes was increased in the dendritic and Treg cells in tumor tissues, but was elevated in Treg and CD8 cells in the blood. In conclusion, cfRNAs in the blood exhibit unique stemness signatures that have potential for use in the diagnosis of lung cancer.
Collapse
Affiliation(s)
- Bo Yan
- Clinical Research Unit, Shanghai Chest Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200050, P.R. China
| | - Yong Chen
- Department of Thoracic Surgery, Shanghai Chest Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200050, P.R. China
| | - Zhouyu Wang
- Berry Oncology Corporation, Beijing 100102, P.R. China
| | - Jing Li
- Berry Oncology Corporation, Beijing 100102, P.R. China
| | - Ruiru Wang
- Berry Oncology Corporation, Beijing 100102, P.R. China
| | - Xufeng Pan
- Department of Thoracic Surgery, Shanghai Chest Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200050, P.R. China
| | - Boyi Li
- Kanghui Biotechnology Corporation, Shenyang, Liaoning 110042, P.R. China
| | - Rong Li
- Clinical Research Unit, Shanghai Chest Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200050, P.R. China
| |
Collapse
|
50
|
Bates M, Mohamed BM, Lewis F, O'Toole S, O'Leary JJ. Biomarkers in high grade serous ovarian cancer. Biochim Biophys Acta Rev Cancer 2024; 1879:189224. [PMID: 39581234 DOI: 10.1016/j.bbcan.2024.189224] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2024] [Revised: 11/15/2024] [Accepted: 11/15/2024] [Indexed: 11/26/2024]
Abstract
High-grade serous ovarian cancer (HGSC) is the most common subtype of ovarian cancer. HGSC patients typically present with advanced disease, which is often resistant to chemotherapy and recurs despite initial responses to therapy, resulting in the poor prognosis associated with this disease. There is a need to utilise biomarkers to manage the various aspects of HGSC patient care. In this review we discuss the current state of biomarkers in HGSC, focusing on the various available immunohistochemical (IHC) and blood-based biomarkers, which have been examined for their diagnostic, prognostic and theranostic potential in HGSC. These include various routine clinical IHC biomarkers such as p53, WT1, keratins, PAX8, Ki67 and p16 and clinical blood-borne markers and algorithms such as CA125, HE4, ROMA, RMI, ROCA, and others. We also discuss various components of the liquid biopsy as well as a number of novel IHC biomarkers and non-routine blood-borne biomarkers, which have been examined in various ovarian cancer studies. We also discuss the future of ovarian cancer biomarker research and highlight some of the challenges currently facing the field.
Collapse
Affiliation(s)
- Mark Bates
- Department of Histopathology, Trinity College Dublin, Dublin, Ireland; Emer Casey Molecular Pathology Research Laboratory, Coombe Women & Infants University Hospital, Dublin, Ireland; Trinity St James's Cancer Institute, Dublin, Ireland.
| | - Bashir M Mohamed
- Department of Histopathology, Trinity College Dublin, Dublin, Ireland; Emer Casey Molecular Pathology Research Laboratory, Coombe Women & Infants University Hospital, Dublin, Ireland; Trinity St James's Cancer Institute, Dublin, Ireland
| | - Faye Lewis
- Department of Histopathology, Trinity College Dublin, Dublin, Ireland; Emer Casey Molecular Pathology Research Laboratory, Coombe Women & Infants University Hospital, Dublin, Ireland; Trinity St James's Cancer Institute, Dublin, Ireland
| | - Sharon O'Toole
- Department of Histopathology, Trinity College Dublin, Dublin, Ireland; Emer Casey Molecular Pathology Research Laboratory, Coombe Women & Infants University Hospital, Dublin, Ireland; Trinity St James's Cancer Institute, Dublin, Ireland; Department of Obstetrics and Gynaecology, Trinity College Dublin, Dublin, Ireland
| | - John J O'Leary
- Department of Histopathology, Trinity College Dublin, Dublin, Ireland; Emer Casey Molecular Pathology Research Laboratory, Coombe Women & Infants University Hospital, Dublin, Ireland; Trinity St James's Cancer Institute, Dublin, Ireland; Department of Pathology, Coombe Women & Infants University Hospital, Dublin, Ireland
| |
Collapse
|