1
|
Normanno N, Morabito A, Rachiglio AM, Sforza V, Landi L, Bria E, Delmonte A, Cappuzzo F, De Luca A. Circulating tumour DNA in early stage and locally advanced NSCLC: ready for clinical implementation? Nat Rev Clin Oncol 2025; 22:215-231. [PMID: 39833354 DOI: 10.1038/s41571-024-00985-w] [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: 12/20/2024] [Indexed: 01/22/2025]
Abstract
Circulating tumour DNA (ctDNA) can be released by cancer cells into biological fluids through apoptosis, necrosis or active release. In patients with non-small-cell lung cancer (NSCLC), ctDNA levels correlate with clinical and pathological factors, including histology, tumour size and proliferative status. Currently, ctDNA analysis is recommended for molecular profiling in patients with advanced-stage NSCLC. In this Review, we summarize the increasing evidence suggesting that ctDNA has potential clinical applications in the management of patients with early stage and locally advanced NSCLC. In those with early stage NSCLC, detection of ctDNA before and/or after surgery is associated with a greater risk of disease recurrence. Longitudinal monitoring after surgery can further increase the prognostic value of ctDNA testing and enables detection of disease recurrence earlier than the assessment of clinical or radiological progression. In patients with locally advanced NSCLC, the detection of ctDNA after chemoradiotherapy is also associated with a greater risk of disease progression. Owing to the limited number of patients enrolled and the different technologies used for ctDNA testing in most of the clinical studies performed thus far, their results are not sufficient to currently support the routine clinical use of ctDNA monitoring in patients with early stage or locally advanced NSCLC. Therefore, we discuss the need for interventional studies to provide evidence for implementing ctDNA testing in this setting.
Collapse
Affiliation(s)
- Nicola Normanno
- Scientific Directorate, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) "Dino Amadori", Meldola, Italy.
| | - Alessandro Morabito
- Thoracic Department, Istituto Nazionale Tumori - IRCCS - Fondazione G. Pascale, Napoli, Italy
| | - Anna Maria Rachiglio
- Cell Biology and Biotherapy Unit, Istituto Nazionale Tumori - IRCCS - Fondazione G. Pascale, Napoli, Italy
| | - Vincenzo Sforza
- Thoracic Department, Istituto Nazionale Tumori - IRCCS - Fondazione G. Pascale, Napoli, Italy
| | - Lorenza Landi
- Clinical Trials Center: Phase 1 and Precision Medicine, IRCCS "Regina Elena" National Cancer Institute, Rome, Italy
| | - Emilio Bria
- Medical Oncology Unit, Università Cattolica del Sacro Cuore, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Roma, Italy
- Medical Oncology, Ospedale Isola Tiberina Gemelli Isola, Roma, Italy
| | - Angelo Delmonte
- Medical Oncology Department, IRCCS IRST "Dino Amadori", Meldola, Italy
| | - Federico Cappuzzo
- Division of Medical Oncology 2, IRCCS "Regina Elena" National Cancer Institute, Rome, Italy
| | - Antonella De Luca
- Cell Biology and Biotherapy Unit, Istituto Nazionale Tumori - IRCCS - Fondazione G. Pascale, Napoli, Italy
| |
Collapse
|
2
|
Ohara S, Suda K, Sudhaman S, Hamada A, Chiba M, Shimoji M, Takemoto T, Kalashnikova E, Cheung SK, Krainock M, Feeney J, Sethi H, Liu MC, Soh J, Tsutani Y, Mitsudomi T. Clinical Significance of Perioperative Minimal Residual Disease Detected by Circulating Tumor DNA in Patients With Lung Cancer With a Long Follow-up Data: An Exploratory Study. JTO Clin Res Rep 2025; 6:100762. [PMID: 39990137 PMCID: PMC11847260 DOI: 10.1016/j.jtocrr.2024.100762] [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: 08/14/2024] [Revised: 10/31/2024] [Accepted: 11/03/2024] [Indexed: 02/25/2025] Open
Abstract
Introduction Molecular residual disease detected by circulating tumor DNA (ctDNA) has been reported to be predictive of patients' outcomes in various types of cancers after curative intent treatment. Nevertheless, additional detailed information regarding the association of longitudinal ctDNA detection with long-term follow-up in lung cancer is needed. Here, we report on a cohort of patients with NSCLC who underwent definitive surgery and ctDNA analysis in the pre-operative, adjuvant, and surveillance settings. Method Plasma samples were collected from 46 patients with clinical stage II-III NSCLC before surgery (n = 46), after surgery (n = 45), and every six months until two years thereafter (n = 78). A clinically validated, personalized, tumor-informed 16-plex polymerase chain reaction-next-generation sequencing assay was used for the detection and quantification of ctDNA in retrospectively analyzed plasma samples. Results Circulating tumor DNA was detected in the first postoperative (within 51 days after surgery) plasma samples in 13% (6/45) of patients (landmark analysis). All of them had disease recurrence within a median of 9.1 months. These patients had shorter recurrence-free and overall survivals than those without detectable ctDNA at a landmark time point (p < 0.01) and in multivariate analyses (p < 0.03). Longitudinally (considering all postoperative follow-up time points), ctDNA was detected in 13 patients, all of whom experienced disease recurrence (positive predictive value = 100%). Three patients who had central nervous system-only metastases did not have detectable ctDNA. Conclusions The presence of ctDNA post-surgery or during surveillance identifies patients with NSCLC at high risk of recurrence. Serial testing is important to detect disease recurrence earlier (lead-time: 3.2 months).
Collapse
Affiliation(s)
- Shuta Ohara
- Division of Thoracic Surgery, Department of Surgery, Kindai University Faculty of Medicine, Osaka-Sayama, Japan
| | - Kenichi Suda
- Division of Thoracic Surgery, Department of Surgery, Kindai University Faculty of Medicine, Osaka-Sayama, Japan
| | | | - Akira Hamada
- Division of Thoracic Surgery, Department of Surgery, Kindai University Faculty of Medicine, Osaka-Sayama, Japan
| | - Masato Chiba
- Division of Thoracic Surgery, Department of Surgery, Kindai University Faculty of Medicine, Osaka-Sayama, Japan
| | - Masaki Shimoji
- Division of Thoracic Surgery, Department of Surgery, Kindai University Faculty of Medicine, Osaka-Sayama, Japan
| | - Toshiki Takemoto
- Division of Thoracic Surgery, Department of Surgery, Kindai University Faculty of Medicine, Osaka-Sayama, Japan
| | | | | | | | | | | | | | - Junichi Soh
- Division of Thoracic Surgery, Department of Surgery, Kindai University Faculty of Medicine, Osaka-Sayama, Japan
- Department of Thoracic Surgery, Osaka Metropolitan University Graduate School of Medicine, Osaka, Japan
| | - Yasuhiro Tsutani
- Division of Thoracic Surgery, Department of Surgery, Kindai University Faculty of Medicine, Osaka-Sayama, Japan
| | - Tetsuya Mitsudomi
- Division of Thoracic Surgery, Department of Surgery, Kindai University Faculty of Medicine, Osaka-Sayama, Japan
- Kindai Hospital Global Research Alliance Center, Kindai University Hospital, Osaka-Sayama, Japan
- Izumi City General Hospital, Izumi, Japan
| |
Collapse
|
3
|
Wang X, Chen Y, Ma C, Bi L, Su Z, Li W, Wang Z. Current advances and future prospects of blood-based techniques for identifying benign and malignant pulmonary nodules. Crit Rev Oncol Hematol 2025; 207:104608. [PMID: 39761937 DOI: 10.1016/j.critrevonc.2024.104608] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2024] [Revised: 12/24/2024] [Accepted: 12/27/2024] [Indexed: 01/16/2025] Open
Abstract
Lung cancer is the leading cause of cancer-related mortality worldwide, highlighting the urgent need for more accurate and minimally invasive diagnostic tools to improve early detection and patient outcomes. While low-dose computed tomography (LDCT) is effective for screening in high-risk individuals, its high false-positive rate necessitates more precise diagnostic strategies. Liquid biopsy, particularly ctDNA methylation analysis, represents a promising alternative for non-invasive classification of indeterminate pulmonary nodules (IPNs). This review highlights the progress and clinical potential of liquid biopsy technologies, including traditional proteins markers, cfDNA, exosomes, metabolomics, circulating tumor cells (CTCs) and platelets, in lung cancer diagnosis. We discuss the integration of ctDNA methylation analysis with traditional imaging and clinical data to enhance the early detection of IPNs, as well as potential solutions to address the challenges of low biomarker concentration and background noise. By advancing precision diagnostics, liquid biopsy technologies could transform lung cancer management, improve survival rates, and reduce the disease burden.
Collapse
Affiliation(s)
- Xin Wang
- Department of Respiratory and Critical Care Medicine, Institute of Respiratory Health, State Key Laboratory of Respiratory Health and Multimorbidity, West China Hospital, Sichuan University, Chengdu, Sichuan, China; Institute of Respiratory Health, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Yanmei Chen
- Health Management Center, West China Tianfu Hospital, Sichuan University, Chengdu, Sichuan, China
| | | | - Lingfeng Bi
- Department of Respiratory and Critical Care Medicine, Institute of Respiratory Health, State Key Laboratory of Respiratory Health and Multimorbidity, West China Hospital, Sichuan University, Chengdu, Sichuan, China; Institute of Respiratory Health, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Zhixi Su
- Singlera Genomics Ltd., Shanghai, China
| | - Weimin Li
- Department of Respiratory and Critical Care Medicine, Institute of Respiratory Health, State Key Laboratory of Respiratory Health and Multimorbidity, West China Hospital, Sichuan University, Chengdu, Sichuan, China; Institute of Respiratory Health, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu, Sichuan, China; Precision Medicine Center, Precision Medicine Key Laboratory of Sichuan Province, West China Hospital, Sichuan University, Chengdu, Sichuan, China; The Research Units of West China, Chinese Academy of Medical Sciences, West China Hospital, Chengdu, Sichuan, China
| | - Zhoufeng Wang
- Department of Respiratory and Critical Care Medicine, Institute of Respiratory Health, State Key Laboratory of Respiratory Health and Multimorbidity, West China Hospital, Sichuan University, Chengdu, Sichuan, China; Institute of Respiratory Health, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu, Sichuan, China; Precision Medicine Center, Precision Medicine Key Laboratory of Sichuan Province, West China Hospital, Sichuan University, Chengdu, Sichuan, China.
| |
Collapse
|
4
|
Salomon R, Razavi Bazaz S, Mutafopulos K, Gallego-Ortega D, Warkiani M, Weitz D, Jin D. Challenges in blood fractionation for cancer liquid biopsy: how can microfluidics assist? LAB ON A CHIP 2025; 25:1097-1127. [PMID: 39775440 DOI: 10.1039/d4lc00563e] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/11/2025]
Abstract
Liquid biopsy provides a minimally invasive approach to characterise the molecular and phenotypic characteristics of a patient's individual tumour by detecting evidence of cancerous change in readily available body fluids, usually the blood. When applied at multiple points during the disease journey, it can be used to monitor a patient's response to treatment and to personalise clinical management based on changes in disease burden and molecular findings. Traditional liquid biopsy approaches such as quantitative PCR, have tended to look at only a few biomarkers, and are aimed at early detection of disease or disease relapse using predefined markers. With advances in the next generation sequencing (NGS) and single-cell genomics, simultaneous analysis of both circulating tumour DNA (ctDNA) and circulating tumour cells (CTCs) is now a real possibility. To realise this, however, we need to overcome issues with current blood collection and fractionation processes. These include overcoming the need to add a preservative to the collection tube or the need to rapidly send blood tubes to a centralised processing lab with the infrastructure required to fractionate and process the blood samples. This review focuses on outlining the current state of liquid biopsy and how microfluidic blood fractionation tools can be used in cancer liquid biopsy. We describe microfluidic devices that can separate plasma for ctDNA analysis, and devices that are important in isolating the cellular component(s) in liquid biopsy, i.e., individual CTCs and CTC clusters. To facilitate a better understanding of these devices, we propose a new categorisation system based on how these devices operate. The three categories being 1) solid Interaction devices, 2) fluid Interaction devices and 3) external force/active devices. Finally, we conclude that whilst some assays and some cancers are well suited to current microfluidic techniques, new tools are necessary to support broader, clinically relevant multiomic workflows in cancer liquid biopsy.
Collapse
Affiliation(s)
- Robert Salomon
- Children's Cancer Institute, Lowy Cancer Research Centre, UNSW, Sydney, Australia.
- Institute for Biomedical Materials and Devices (IBMD)/Faculty of Science, University of Technology Sydney, Sydney, NSW, 2007 Australia
| | - Sajad Razavi Bazaz
- Children's Cancer Institute, Lowy Cancer Research Centre, UNSW, Sydney, Australia.
| | - Kirk Mutafopulos
- Department of Physics, Harvard University, Cambridge, MA, 02138, USA
| | - David Gallego-Ortega
- Institute for Biomedical Materials and Devices (IBMD)/Faculty of Science, University of Technology Sydney, Sydney, NSW, 2007 Australia
- School of Clinical Medicine, Faculty of Medicine, University of New South Wales, Sydney, NSW, 2052, Australia
- School of Biomedical Engineering, University of Technology Sydney, Sydney, New South Wales 2007, Australia
- Garvan Institute of Medical Research, Darlinghurst, Sydney, NSW 2010, Australia
| | - Majid Warkiani
- Institute for Biomedical Materials and Devices (IBMD)/Faculty of Science, University of Technology Sydney, Sydney, NSW, 2007 Australia
- School of Biomedical Engineering, University of Technology Sydney, Sydney, New South Wales 2007, Australia
| | - David Weitz
- Department of Physics, Harvard University, Cambridge, MA, 02138, USA
| | - Dayong Jin
- Institute for Biomedical Materials and Devices (IBMD)/Faculty of Science, University of Technology Sydney, Sydney, NSW, 2007 Australia
| |
Collapse
|
5
|
Venetis K, Frascarelli C, Bielo LB, Cursano G, Adorisio R, Ivanova M, Mane E, Peruzzo V, Concardi A, Negrelli M, D'Ercole M, Porta FM, Zhan Y, Marra A, Trapani D, Criscitiello C, Curigliano G, Guerini-Rocco E, Fusco N. Mismatch repair (MMR) and microsatellite instability (MSI) phenotypes across solid tumors: A comprehensive cBioPortal study on prevalence and prognostic impact. Eur J Cancer 2025; 217:115233. [PMID: 39827722 DOI: 10.1016/j.ejca.2025.115233] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2024] [Accepted: 01/08/2025] [Indexed: 01/22/2025]
Abstract
Mismatch repair deficiency (MMR-d) and microsatellite instability (MSI) are prognostic and predictive biomarkers in oncology. Current testing for MMR/MSI relies on immunohistochemistry (IHC) for MMR proteins and molecular assays for MSI detection. This combined diagnostic strategy, however, lacks tumor specificity and does not account for gene variants. This study provides an in-depth analysis of MMR mutations frequency, spectrum, and distribution in solid tumors. Data from 23,893 patients across 11 tumor types, using 66 publicly available studies, were analyzed. MMR-mutated (MMR-m) status was defined by alterations in MLH1, PMS2, MSH2, and/or MSH6; MSI was assessed by MSIsensor. Cases with indeterminate labelling were excluded. Survival was analyzed using the Kaplan-Meier method. Among 19,353 tumors, 949 MMR variants were identified, comprising 432 pathogenic and 517 variants of unknown significance (VUS), as defined by OncoKB. MSH6 mutations were the most frequent (n = 279, 29.4 %), followed by MSH2 (n = 198, 20.9 %), MLH1 (n = 187, 19.7 %), and PMS2 (n = 161, 16.9 %). MMR-m cases were more frequent in endometrial (EC, 20.5 %), colorectal (CRC, 8.2 %), bladder (BLCA, 8.7 %), and gastroesophageal cancers (GEC, 5.4 %). Pathogenic mutations were more common than non-pathogenic in EC, CRC, and GEC (p < 0.001, p = 0.01, p = 0.32, respectively). MMR-m status was not associated with MSI in 247 (48.9 %) cases, including 67 (13.2 %) with pathogenic mutations. The highest concordance between MMR-m and MSI was observed in CRC (65.7 %), EC (91.2 %), and GEC (69.6 %), while the lowest in pancreatic (0.2 %) and lung cancers (0.1 %). MMR-m GECs showed improved overall survival compared to MMR-wt (p = 0.009), a relationship not observed in other tumor types. This study demonstrates that the MMR spectrum is extremely hetoerogeneous in solid tumors, highliting the need for comprehensive and tumor-specific testing strategies.
Collapse
Affiliation(s)
| | - Chiara Frascarelli
- Division of Pathology, European Institute of Oncology IRCCS, Milan, Italy; Department of Oncology and Hemato-Oncology, University of Milan, Milan, Italy
| | - Luca Boscolo Bielo
- Division of New Drugs and Early Drug Development for Innovative Therapies, European Institute of Oncology IRCCS, Milan, Italy
| | - Giulia Cursano
- Division of Pathology, European Institute of Oncology IRCCS, Milan, Italy; Department of Oncology and Hemato-Oncology, University of Milan, Milan, Italy
| | - Riccardo Adorisio
- Division of Pathology, European Institute of Oncology IRCCS, Milan, Italy
| | - Mariia Ivanova
- Division of Pathology, European Institute of Oncology IRCCS, Milan, Italy
| | - Eltjona Mane
- Division of Pathology, European Institute of Oncology IRCCS, Milan, Italy
| | - Virginia Peruzzo
- Division of Pathology, European Institute of Oncology IRCCS, Milan, Italy
| | - Alberto Concardi
- Division of Pathology, European Institute of Oncology IRCCS, Milan, Italy
| | | | - Marianna D'Ercole
- Division of Pathology, European Institute of Oncology IRCCS, Milan, Italy
| | | | - Yinxiu Zhan
- Department of Experimental Oncology, European Institute of Oncology IRCCS, Milan, Italy
| | - Antonio Marra
- Division of New Drugs and Early Drug Development for Innovative Therapies, European Institute of Oncology IRCCS, Milan, Italy
| | - Dario Trapani
- Department of Oncology and Hemato-Oncology, University of Milan, Milan, Italy; Division of New Drugs and Early Drug Development for Innovative Therapies, European Institute of Oncology IRCCS, Milan, Italy
| | - Carmen Criscitiello
- Department of Oncology and Hemato-Oncology, University of Milan, Milan, Italy; Division of New Drugs and Early Drug Development for Innovative Therapies, European Institute of Oncology IRCCS, Milan, Italy
| | - Giuseppe Curigliano
- Department of Oncology and Hemato-Oncology, University of Milan, Milan, Italy; Division of New Drugs and Early Drug Development for Innovative Therapies, European Institute of Oncology IRCCS, Milan, Italy
| | - Elena Guerini-Rocco
- Division of Pathology, European Institute of Oncology IRCCS, Milan, Italy; Department of Oncology and Hemato-Oncology, University of Milan, Milan, Italy
| | - Nicola Fusco
- Division of Pathology, European Institute of Oncology IRCCS, Milan, Italy; Department of Oncology and Hemato-Oncology, University of Milan, Milan, Italy.
| |
Collapse
|
6
|
Al Bakir M, Reading JL, Gamble S, Rosenthal R, Uddin I, Rowan A, Przewrocka J, Rogers A, Wong YNS, Bentzen AK, Veeriah S, Ward S, Garnett AT, Kalavakur P, Martínez-Ruiz C, Puttick C, Huebner A, Cook DE, Moore DA, Abbosh C, Hiley CT, Naceur-Lombardelli C, Watkins TBK, Petkovic M, Schwarz RF, Gálvez-Cancino F, Litchfield K, Meldgaard P, Sorensen BS, Madsen LB, Jäger D, Forster MD, Arkenau T, Domingo-Vila C, Tree TIM, Kadivar M, Hadrup SR, Chain B, Quezada SA, McGranahan N, Swanton C. Clonal driver neoantigen loss under EGFR TKI and immune selection pressures. Nature 2025:10.1038/s41586-025-08586-y. [PMID: 39972134 DOI: 10.1038/s41586-025-08586-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2022] [Accepted: 01/02/2025] [Indexed: 02/21/2025]
Abstract
Neoantigen vaccines are under investigation for various cancers, including epidermal growth factor receptor (EGFR)-driven lung cancers1,2. We tracked the phylogenetic history of an EGFR mutant lung cancer treated with erlotinib, osimertinib, radiotherapy and a personalized neopeptide vaccine (NPV) targeting ten somatic mutations, including EGFR exon 19 deletion (ex19del). The ex19del mutation was clonal, but is likely to have appeared after a whole-genome doubling (WGD) event. Following osimertinib and NPV treatment, loss of the ex19del mutation was identified in a progressing small-cell-transformed liver metastasis. Circulating tumour DNA analyses tracking 467 somatic variants revealed the presence of this EGFR wild-type clone before vaccination and its expansion during osimertinib/NPV therapy. Despite systemic T cell reactivity to the vaccine-targeted ex19del neoantigen, the NPV failed to halt disease progression. The liver metastasis lost vaccine-targeted neoantigens through chromosomal instability and exhibited a hostile microenvironment, characterized by limited immune infiltration, low CXCL9 and elevated M2 macrophage levels. Neoantigens arising post-WGD were more likely to be absent in the progressing liver metastasis than those occurring pre-WGD, suggesting that prioritizing pre-WGD neoantigens may improve vaccine design. Data from the TRACERx 421 cohort3 provide evidence that pre-WGD mutations better represent clonal variants, and owing to their presence at multiple copy numbers, are less likely to be lost in metastatic transition. These data highlight the power of phylogenetic disease tracking and functional T cell profiling to understand mechanisms of immune escape during combination therapies.
Collapse
Affiliation(s)
- Maise Al Bakir
- Cancer Evolution and Genome Instability Laboratory, The Francis Crick Institute, London, UK
- Cancer Research UK Lung Cancer Centre of Excellence, University College London Cancer Institute, London, UK
| | - James L Reading
- Cancer Research UK Lung Cancer Centre of Excellence, University College London Cancer Institute, London, UK
- Pre-Cancer Immunology Laboratory, Research Department of Haematology, University College London Cancer Institute, London, UK
| | - Samuel Gamble
- Pre-Cancer Immunology Laboratory, Research Department of Haematology, University College London Cancer Institute, London, UK
| | - Rachel Rosenthal
- Cancer Evolution and Genome Instability Laboratory, The Francis Crick Institute, London, UK
| | - Imran Uddin
- Division of Infection and Immunity, University College London, London, UK
| | - Andrew Rowan
- Cancer Evolution and Genome Instability Laboratory, The Francis Crick Institute, London, UK
| | - Joanna Przewrocka
- Cancer Evolution and Genome Instability Laboratory, The Francis Crick Institute, London, UK
| | - Amber Rogers
- Pre-Cancer Immunology Laboratory, Research Department of Haematology, University College London Cancer Institute, London, UK
| | - Yien Ning Sophia Wong
- Cancer Research UK Lung Cancer Centre of Excellence, University College London Cancer Institute, London, UK
| | - Amalie K Bentzen
- Pre-Cancer Immunology Laboratory, Research Department of Haematology, University College London Cancer Institute, London, UK
| | - Selvaraju Veeriah
- Cancer Research UK Lung Cancer Centre of Excellence, University College London Cancer Institute, London, UK
| | - Sophia Ward
- Cancer Evolution and Genome Instability Laboratory, The Francis Crick Institute, London, UK
- Cancer Research UK Lung Cancer Centre of Excellence, University College London Cancer Institute, London, UK
- Genomics Science Technology Platform, The Francis Crick Institute, London, UK
| | | | | | - Carlos Martínez-Ruiz
- Cancer Genome Evolution Research Group, University College London Cancer Institute, University College London, London, UK
| | - Clare Puttick
- Cancer Evolution and Genome Instability Laboratory, The Francis Crick Institute, London, UK
- Cancer Research UK Lung Cancer Centre of Excellence, University College London Cancer Institute, London, UK
- Cancer Genome Evolution Research Group, University College London Cancer Institute, University College London, London, UK
| | - Ariana Huebner
- Cancer Evolution and Genome Instability Laboratory, The Francis Crick Institute, London, UK
- Cancer Research UK Lung Cancer Centre of Excellence, University College London Cancer Institute, London, UK
- Cancer Genome Evolution Research Group, University College London Cancer Institute, University College London, London, UK
| | - Daniel E Cook
- Cancer Evolution and Genome Instability Laboratory, The Francis Crick Institute, London, UK
| | - David A Moore
- Cancer Evolution and Genome Instability Laboratory, The Francis Crick Institute, London, UK
- Cancer Research UK Lung Cancer Centre of Excellence, University College London Cancer Institute, London, UK
- Department of Cellular Pathology, University College London Hospital NHS Foundation Trust, London, UK
| | - Chris Abbosh
- Cancer Research UK Lung Cancer Centre of Excellence, University College London Cancer Institute, London, UK
| | - Crispin T Hiley
- Cancer Evolution and Genome Instability Laboratory, The Francis Crick Institute, London, UK
- Cancer Research UK Lung Cancer Centre of Excellence, University College London Cancer Institute, London, UK
| | | | - Thomas B K Watkins
- Cancer Evolution and Genome Instability Laboratory, The Francis Crick Institute, London, UK
| | - Marina Petkovic
- Berlin Institute for Medical Systems Biology, Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany
- Department of Biology, Humboldt University of Berlin, Berlin, Germany
- Division of Oncology and Hematology, Department of Pediatrics, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt Universität zu Berlin, Berlin, Germany
| | - Roland F Schwarz
- Institute for Computational Cancer Biology (ICCB), Center for Integrated Oncology (CIO), Cancer Research Center Cologne Essen (CCCE), Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
- Berlin Institute for the Foundations of Learning and Data (BIFOLD), Berlin, Germany
| | - Felipe Gálvez-Cancino
- Immune-Regulation and Immune-Interactions Laboratory, Centre for Immuno-Oncology, Nuffield Department of Medicine, University of Oxford, Headington, UK
| | - Kevin Litchfield
- Cancer Evolution and Genome Instability Laboratory, The Francis Crick Institute, London, UK
- Cancer Research UK Lung Cancer Centre of Excellence, University College London Cancer Institute, London, UK
| | - Peter Meldgaard
- Department of Oncology, Aarhus University Hospital, Aarhus, Denmark
| | - Boe Sandahl Sorensen
- Department of Clinical Biochemistry, Aarhus University Hospital, Aarhus, Denmark
| | - Line Bille Madsen
- Department of Pathology, Aarhus University Hospital, Aarhus, Denmark
| | - Dirk Jäger
- Department of Medical Oncology, National Center for Tumor Diseases Heidelberg, Heidelberg University Hospital, Heidelberg, Germany
| | - Martin D Forster
- Cancer Research UK Lung Cancer Centre of Excellence, University College London Cancer Institute, London, UK
- Department of Oncology, UCL Cancer Institute, London, UK
| | | | - Clara Domingo-Vila
- Department of Immunobiology, Faculty of Life Sciences and Medicine, King's College London, London, UK
| | - Timothy I M Tree
- Department of Immunobiology, Faculty of Life Sciences and Medicine, King's College London, London, UK
| | - Mohammad Kadivar
- Department of Health Technology, Technical University of Denmark, Lyngby, Denmark
| | - Sine Reker Hadrup
- Department of Health Technology, Technical University of Denmark, Lyngby, Denmark
| | - Benny Chain
- Division of Infection and Immunity, University College London, London, UK
- Department of Computer Sciences, University College London, London, UK
| | - Sergio A Quezada
- Cancer Research UK Lung Cancer Centre of Excellence, University College London Cancer Institute, London, UK.
- Cancer Immunology Unit, Research Department of Haematology, University College London Cancer Institute, London, UK.
| | - Nicholas McGranahan
- Cancer Research UK Lung Cancer Centre of Excellence, University College London Cancer Institute, London, UK.
- Cancer Genome Evolution Research Group, University College London Cancer Institute, University College London, London, UK.
| | - Charles Swanton
- Cancer Evolution and Genome Instability Laboratory, The Francis Crick Institute, London, UK.
- Cancer Research UK Lung Cancer Centre of Excellence, University College London Cancer Institute, London, UK.
| |
Collapse
|
7
|
Zeng Z, Yi Z, Xu B. The biological and technical challenges facing utilizing circulating tumor DNA in non-metastatic breast cancer patients. Cancer Lett 2025; 616:217574. [PMID: 39983895 DOI: 10.1016/j.canlet.2025.217574] [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: 02/13/2025] [Accepted: 02/18/2025] [Indexed: 02/23/2025]
Abstract
Breast cancer is one of the most prevalent cancers and has emerged as a major global challenge. Circulating tumor DNA (ctDNA), a liquid biopsy method, overcomes the accessibility limitations of tissue-based testing and is widely used for monitoring minimal residual disease and molecular relapse, predicting prognosis, evaluating the response of neoadjuvant therapy, and optimizing treatment decisions in non-metastatic breast cancer. However, the application of ctDNA still faces many challenges. Here, we survey the clinical applications of ctDNA in non-metastatic breast cancer and discuss the significant biological and technical challenges of utilizing ctDNA. Importantly, we investigate potential avenues for addressing the challenges. In addition, emerging technologies, including fragmentomics detection, methylation sequencing, and long-read sequencing, have clinical potential and could be a future direction. Proper utilization of machine learning facilitates the identification of meaningful patterns from complex fragment and methylation profiles of ctDNA. There is still a lack of clinical trials focused on the subsets of ctDNA (e.g., circulating mitochondrial DNA), ctDNA-inferred drug-resistant clonal evolution, tumor heterogeneity, and ctDNA-guided clinical decision-making in non-metastatic breast cancer. Due to regional differences in the number of registered clinical trials, it is essential to enhance communication and foster global collaboration to advance the field.
Collapse
Affiliation(s)
- Zihang Zeng
- Department of Radiation and Medical Oncology, Hubei Key Laboratory of Tumor Biological Behaviors, Hubei Cancer Clinical Study Center, Zhongnan Hospital of Wuhan University, 430071, Wuhan, China
| | - Zongbi Yi
- Department of Radiation and Medical Oncology, Hubei Key Laboratory of Tumor Biological Behaviors, Hubei Cancer Clinical Study Center, Zhongnan Hospital of Wuhan University, 430071, Wuhan, China.
| | - Binghe Xu
- Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, 100021, Beijing, China.
| |
Collapse
|
8
|
Janke F, Stritzke F, Dvornikovich K, Franke H, Angeles AK, Riediger AL, Ogrodnik S, Gerhardt S, Regnery S, Schröter P, Bauer L, Weusthof K, Görtz M, Harrabi S, Herfarth K, Neelsen C, Paech D, Schlemmer H, Abdollahi A, Adeberg S, Debus J, Sültmann H, Held T. Early circulating tumor DNA changes predict outcomes in head and neck cancer patients under re-radiotherapy. Int J Cancer 2025; 156:853-864. [PMID: 39212345 PMCID: PMC11661516 DOI: 10.1002/ijc.35152] [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: 05/17/2024] [Revised: 07/17/2024] [Accepted: 07/30/2024] [Indexed: 09/04/2024]
Abstract
Local recurrence after radiotherapy is common in locally advanced head and neck cancer (HNC) patients. Re-irradiation can improve local disease control, but disease progression remains frequent. Hence, predictive biomarkers are needed to adapt treatment intensity to the patient's individual risk. We quantified circulating tumor DNA (ctDNA) in sequential plasma samples and correlated ctDNA levels with disease outcome. Ninety four longitudinal plasma samples from 16 locally advanced HNC patients and 57 healthy donors were collected at re-radiotherapy baseline, after 5 and 10 radiation fractions, at irradiation end, and at routine follow-up visits. Plasma DNA was subjected to low coverage whole genome sequencing for copy number variation (CNV) profiling to quantify ctDNA burden. CNV-based ctDNA burden was detected in 8/16 patients and 25/94 plasma samples. Ten additional ctDNA-positive samples were identified by tracking patient-specific CNVs found in earlier sequential plasma samples. ctDNA-positivity after 5 and 10 radiation fractions (both: log-rank, p = .050) as well as at the end of irradiation correlated with short progression-free survival (log-rank, p = .006). Moreover, a pronounced decrease of ctDNA toward re-radiotherapy termination was associated with worse treatment outcome (log-rank, p = .005). Dynamic ctDNA tracking in serial plasma beyond re-radiotherapy reflected treatment response and imminent disease progression. In five patients, molecular progression was detected prior to tumor progression based on clinical imaging. Our findings emphasize that quantifying ctDNA during re-radiotherapy may contribute to disease monitoring and personalization of adjuvant treatment, follow-up intervals, and dose prescription.
Collapse
Affiliation(s)
- Florian Janke
- Division of Cancer Genome Research, German Cancer Research Center (DKFZ)HeidelbergGermany
- German Center for Lung Research (DZL), TLRC HeidelbergHeidelbergGermany
- National Center for Tumor Diseases (NCT)HeidelbergGermany
| | - Florian Stritzke
- National Center for Tumor Diseases (NCT)HeidelbergGermany
- Department of Radiation OncologyHeidelberg University HospitalHeidelbergGermany
- Heidelberg Institute of Radiation Oncology (HIRO)HeidelbergGermany
- Clinical Cooperation Unit Radiation Oncology, German Cancer Research Center (DKFZ)HeidelbergGermany
| | | | - Henrik Franke
- Department of Radiation OncologyHeidelberg University HospitalHeidelbergGermany
| | - Arlou Kristina Angeles
- Division of Cancer Genome Research, German Cancer Research Center (DKFZ)HeidelbergGermany
- German Center for Lung Research (DZL), TLRC HeidelbergHeidelbergGermany
- National Center for Tumor Diseases (NCT)HeidelbergGermany
| | - Anja Lisa Riediger
- Division of Cancer Genome Research, German Cancer Research Center (DKFZ)HeidelbergGermany
- National Center for Tumor Diseases (NCT)HeidelbergGermany
- Junior Clinical Cooperation Unit, Multiparametric Methods for Early Detection of Prostate Cancer, German Cancer Research Center (DKFZ)HeidelbergGermany
- Department of UrologyUniversity Hospital HeidelbergHeidelbergGermany
- Faculty of BiosciencesHeidelberg UniversityHeidelbergGermany
| | - Simon Ogrodnik
- Division of Cancer Genome Research, German Cancer Research Center (DKFZ)HeidelbergGermany
- National Center for Tumor Diseases (NCT)HeidelbergGermany
| | - Sabrina Gerhardt
- Division of Cancer Genome Research, German Cancer Research Center (DKFZ)HeidelbergGermany
- National Center for Tumor Diseases (NCT)HeidelbergGermany
| | - Sebastian Regnery
- National Center for Tumor Diseases (NCT)HeidelbergGermany
- Department of Radiation OncologyHeidelberg University HospitalHeidelbergGermany
- Heidelberg Institute of Radiation Oncology (HIRO)HeidelbergGermany
- Heidelberg Ion Beam Therapy Center (HIT)HeidelbergGermany
| | - Philipp Schröter
- National Center for Tumor Diseases (NCT)HeidelbergGermany
- Department of Radiation OncologyHeidelberg University HospitalHeidelbergGermany
- Heidelberg Institute of Radiation Oncology (HIRO)HeidelbergGermany
| | - Lukas Bauer
- National Center for Tumor Diseases (NCT)HeidelbergGermany
- Department of Radiation OncologyHeidelberg University HospitalHeidelbergGermany
- Heidelberg Institute of Radiation Oncology (HIRO)HeidelbergGermany
| | - Katharina Weusthof
- National Center for Tumor Diseases (NCT)HeidelbergGermany
- Department of Radiation OncologyHeidelberg University HospitalHeidelbergGermany
- Heidelberg Institute of Radiation Oncology (HIRO)HeidelbergGermany
- Heidelberg Ion Beam Therapy Center (HIT)HeidelbergGermany
| | - Magdalena Görtz
- Junior Clinical Cooperation Unit, Multiparametric Methods for Early Detection of Prostate Cancer, German Cancer Research Center (DKFZ)HeidelbergGermany
- Department of UrologyUniversity Hospital HeidelbergHeidelbergGermany
| | - Semi Harrabi
- National Center for Tumor Diseases (NCT)HeidelbergGermany
- Department of Radiation OncologyHeidelberg University HospitalHeidelbergGermany
- Heidelberg Institute of Radiation Oncology (HIRO)HeidelbergGermany
- Clinical Cooperation Unit Radiation Oncology, German Cancer Research Center (DKFZ)HeidelbergGermany
- Heidelberg Ion Beam Therapy Center (HIT)HeidelbergGermany
- German Cancer Consortium (DKTK)HeidelbergGermany
| | - Klaus Herfarth
- National Center for Tumor Diseases (NCT)HeidelbergGermany
- Department of Radiation OncologyHeidelberg University HospitalHeidelbergGermany
- Heidelberg Institute of Radiation Oncology (HIRO)HeidelbergGermany
- Clinical Cooperation Unit Radiation Oncology, German Cancer Research Center (DKFZ)HeidelbergGermany
- Heidelberg Ion Beam Therapy Center (HIT)HeidelbergGermany
- German Cancer Consortium (DKTK)HeidelbergGermany
| | - Christian Neelsen
- Division of Radiology, German Cancer Research Center (DKFZ)HeidelbergGermany
| | - Daniel Paech
- Division of Radiology, German Cancer Research Center (DKFZ)HeidelbergGermany
- Department of NeuroradiologyBonn University HospitalBonnGermany
| | | | - Amir Abdollahi
- National Center for Tumor Diseases (NCT)HeidelbergGermany
- Department of Radiation OncologyHeidelberg University HospitalHeidelbergGermany
- Heidelberg Institute of Radiation Oncology (HIRO)HeidelbergGermany
- Clinical Cooperation Unit Radiation Oncology, German Cancer Research Center (DKFZ)HeidelbergGermany
- Heidelberg Ion Beam Therapy Center (HIT)HeidelbergGermany
- German Cancer Consortium (DKTK)HeidelbergGermany
| | - Sebastian Adeberg
- National Center for Tumor Diseases (NCT)HeidelbergGermany
- Department of Radiation OncologyHeidelberg University HospitalHeidelbergGermany
- Heidelberg Institute of Radiation Oncology (HIRO)HeidelbergGermany
- Clinical Cooperation Unit Radiation Oncology, German Cancer Research Center (DKFZ)HeidelbergGermany
- Heidelberg Ion Beam Therapy Center (HIT)HeidelbergGermany
- German Cancer Consortium (DKTK)HeidelbergGermany
- Department of Radiotherapy and Radiation OncologyMarburg University HospitalMarburgGermany
- Marburg Ion‐Beam Therapy Center (MIT), Department of Radiotherapy and Radiation OncologyMarburg University HospitalMarburgGermany
- Universitäres Centrum für Tumorerkrankungen (UCT) FrankfurtMarburgGermany
| | - Jürgen Debus
- National Center for Tumor Diseases (NCT)HeidelbergGermany
- Department of Radiation OncologyHeidelberg University HospitalHeidelbergGermany
- Heidelberg Institute of Radiation Oncology (HIRO)HeidelbergGermany
- Clinical Cooperation Unit Radiation Oncology, German Cancer Research Center (DKFZ)HeidelbergGermany
- Heidelberg Ion Beam Therapy Center (HIT)HeidelbergGermany
- German Cancer Consortium (DKTK)HeidelbergGermany
| | - Holger Sültmann
- Division of Cancer Genome Research, German Cancer Research Center (DKFZ)HeidelbergGermany
- German Center for Lung Research (DZL), TLRC HeidelbergHeidelbergGermany
- National Center for Tumor Diseases (NCT)HeidelbergGermany
- German Cancer Consortium (DKTK)HeidelbergGermany
| | - Thomas Held
- National Center for Tumor Diseases (NCT)HeidelbergGermany
- Department of Radiation OncologyHeidelberg University HospitalHeidelbergGermany
- Heidelberg Institute of Radiation Oncology (HIRO)HeidelbergGermany
- Clinical Cooperation Unit Radiation Oncology, German Cancer Research Center (DKFZ)HeidelbergGermany
- Heidelberg Ion Beam Therapy Center (HIT)HeidelbergGermany
- German Cancer Consortium (DKTK)HeidelbergGermany
| |
Collapse
|
9
|
Sheriff S, Saba M, Patel R, Fisher G, Schroeder T, Arnolda G, Luo D, Warburton L, Gray E, Long G, Braithwaite J, Rizos H, Ellis LA. A scoping review of factors influencing the implementation of liquid biopsy for cancer care. J Exp Clin Cancer Res 2025; 44:50. [PMID: 39934875 DOI: 10.1186/s13046-025-03322-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2024] [Accepted: 02/06/2025] [Indexed: 02/13/2025] Open
Abstract
BACKGROUND Liquid biopsy (LB) offers a promising, minimally invasive alternative to traditional tissue biopsies in cancer care, enabling real-time monitoring and personalized treatment. Despite its potential, the routine implementation of LB in clinical practice faces significant challenges. This scoping review examines the barriers and facilitators influencing the implementation of liquid biopsies into standard cancer care. METHODS Four academic databases (PubMed, Scopus, Embase, and Web of Science) were systematically searched without language restrictions. We included peer-reviewed articles that were published between January 2019 and March 2024 that focused on the implementation of LB in cancer care or described barriers and facilitators to its implementation. Data relevant to the review objective, including key article characteristics; barriers and facilitators of implementation; and recommendations for advancement or optimisation; were extracted and analysed using thematic and visual network analyses. RESULTS The majority of the included articles were narrative review articles (84%), with most from China (24.2%) and the United States (20%). Thematic analysis identified four main categories and their associated barriers and facilitators to the implementation of LB in cancer care: (1) Laboratory and personnel requirements; (2) Disease specificity; (3) Biomarker-based liquid biopsy; and (4) Policy and regulation. The majority of barriers identified were concentrated in the pre-analytical phase, highlighting the lack of standardization in LB technologies and outcomes. CONCLUSIONS Through a thematic analysis of the barriers and facilitators to LB implementation, we present an integrated tool designed to encourage the standardization of testing methods for clinical practice guidelines in the field.
Collapse
Affiliation(s)
- Samran Sheriff
- Centre for Healthcare Resilience and Implementation Science, Australian Institute of Health Innovation, Macquarie University, Level 6, 75 Talavera Road, North Ryde, Sydney, NSW, Australia.
| | - Maree Saba
- Centre for Healthcare Resilience and Implementation Science, Australian Institute of Health Innovation, Macquarie University, Level 6, 75 Talavera Road, North Ryde, Sydney, NSW, Australia
| | - Romika Patel
- Centre for Healthcare Resilience and Implementation Science, Australian Institute of Health Innovation, Macquarie University, Level 6, 75 Talavera Road, North Ryde, Sydney, NSW, Australia
| | - Georgia Fisher
- Centre for Healthcare Resilience and Implementation Science, Australian Institute of Health Innovation, Macquarie University, Level 6, 75 Talavera Road, North Ryde, Sydney, NSW, Australia
| | - Tanja Schroeder
- Centre for Healthcare Resilience and Implementation Science, Australian Institute of Health Innovation, Macquarie University, Level 6, 75 Talavera Road, North Ryde, Sydney, NSW, Australia
| | - Gaston Arnolda
- Centre for Healthcare Resilience and Implementation Science, Australian Institute of Health Innovation, Macquarie University, Level 6, 75 Talavera Road, North Ryde, Sydney, NSW, Australia
| | - Dan Luo
- The Daffodil Centre, Sydney, NSW, Australia
| | - Lydia Warburton
- Centre for Precision Health, Edith Cowan University, Joondalup, WA, Australia
- Department of Medical Oncology, Fiona Stanly Hospital, Murdoch, WA, Australia
| | - Elin Gray
- Centre for Precision Health, Edith Cowan University, Joondalup, WA, Australia
- School of Medical and Health Sciences, Edith Cowan University, Joondalup, WA, Australia
| | - Georgina Long
- Melanoma Institute Australia, The University of Sydney, Sydney, NSW, Australia
- Faculty of Medicine & Health, The University of Sydney, Sydney, NSW, Australia
- Charles Perkins Centre, The University of Sydney, Sydney, NSW, Australia
- Royal North Shore and Mater Hospitals, North Sydney, Sydney, NSW, Australia
| | - Jeffrey Braithwaite
- Centre for Healthcare Resilience and Implementation Science, Australian Institute of Health Innovation, Macquarie University, Level 6, 75 Talavera Road, North Ryde, Sydney, NSW, Australia
| | - Helen Rizos
- Melanoma Institute Australia, The University of Sydney, Sydney, NSW, Australia
- Macquarie Medical School, Faculty of Medicine Health and Human Science, Macquarie University, Sydney, NSW, Australia
| | - Louise Ann Ellis
- Centre for Healthcare Resilience and Implementation Science, Australian Institute of Health Innovation, Macquarie University, Level 6, 75 Talavera Road, North Ryde, Sydney, NSW, Australia
| |
Collapse
|
10
|
Yang H, Zhang L, Kang X, Si Y, Song P, Su X. Reaction Pathway Differentiation Enabled Fingerprinting Signal for Single Nucleotide Variant Detection. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025:e2412680. [PMID: 39903775 DOI: 10.1002/advs.202412680] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/10/2024] [Revised: 01/17/2025] [Indexed: 02/06/2025]
Abstract
Accurate identification of single-nucleotide variants (SNVs) is paramount for disease diagnosis. Despite the facile design of DNA hybridization probes, their limited specificity poses challenges in clinical applications. Here, a differential reaction pathway probe (DRPP) based on a dynamic DNA reaction network is presented. DRPP leverages differences in reaction intermediate concentrations between SNV and WT groups, directing them into distinct reaction pathways. This generates a strong pulse-like signal for SNV and a weak unidirectional increase signal for wild-type (WT). Through the application of machine learning to fluorescence kinetic data analysis, the classification of SNV and WT signals is automated with an accuracy of 99.6%, significantly exceeding the 80.7% accuracy of conventional methods. Additionally, sensitivity for variant allele frequency (VAF) is enhanced down to 0.1%, representing a ten-fold improvement over conventional approaches. DRPP accurately identified D614G and N501Y SNVs in the S gene of SARS-CoV-2 variants in patient swab samples with accuracy over 99% (n = 82). It determined the VAF of ovarian cancer-related mutations KRAS-G12R, NRAS-G12C, and BRAF-V600E in both tissue and blood samples (n = 77), discriminating cancer patients and healthy individuals with significant difference (p < 0.001). The potential integration of DRPP into clinical diagnostics, along with rapid amplification techniques, holds promise for early disease diagnostics and personalized diagnostics.
Collapse
Affiliation(s)
- Huixiao Yang
- State Key Laboratory of Organic-Inorganic Composites, Beijing Key Laboratory of Bioprocess, Beijing Advanced Innovation Center for Soft Matter Science and Engineering, College of Life Science and Technology, Beijing University of Chemical Technology, Beijing, 100029, China
| | - Linghao Zhang
- State Key Laboratory of Organic-Inorganic Composites, Beijing Key Laboratory of Bioprocess, Beijing Advanced Innovation Center for Soft Matter Science and Engineering, College of Life Science and Technology, Beijing University of Chemical Technology, Beijing, 100029, China
| | - Xinmiao Kang
- State Key Laboratory of Organic-Inorganic Composites, Beijing Key Laboratory of Bioprocess, Beijing Advanced Innovation Center for Soft Matter Science and Engineering, College of Life Science and Technology, Beijing University of Chemical Technology, Beijing, 100029, China
| | - Yunpei Si
- School of Biomedical Engineering, Zhangjiang Institute for Advanced Study and National Center for Translational Medicine, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Ping Song
- School of Biomedical Engineering, Zhangjiang Institute for Advanced Study and National Center for Translational Medicine, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Xin Su
- State Key Laboratory of Organic-Inorganic Composites, Beijing Key Laboratory of Bioprocess, Beijing Advanced Innovation Center for Soft Matter Science and Engineering, College of Life Science and Technology, Beijing University of Chemical Technology, Beijing, 100029, China
- State Key Laboratory of Natural and Biomimetic Drugs, Peking University, Beijing, 100191, China
| |
Collapse
|
11
|
Ruglioni M, Petrini I, Crucitta S, Sbrana A, Luculli GI, Sadeghi Gol L, Forte C, Chella A, Rolfo C, Danesi R, Del Re M. Clinical characteristics of EGFR-ctDNA shedders in EGFR-mutant NSCLC patients. Transl Oncol 2025; 52:102228. [PMID: 39709717 PMCID: PMC11832947 DOI: 10.1016/j.tranon.2024.102228] [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: 09/20/2024] [Revised: 11/17/2024] [Accepted: 11/28/2024] [Indexed: 12/24/2024] Open
Abstract
BACKGROUND Circulating tumor DNA (ctDNA) revolutionized the molecular diagnostics of lung cancer by enabling non-invasive, sensitive identification of actionable mutations. However, ctDNA analysis may be challenging due to tumor shedding variability, leading to false negative results. This study aims to understand the determinants for ctDNA shedding based on clinical characteristics of lung cancer patients, for a better interpretation of false negative results to be considered when ordering ctDNA analysis for clinical practice. METHODS Blood samples were collected from patients with stage IV EGFR-mutated (mEGFR) NSCLC before treatment and monitored until disease progression. EGFR was assessed on tissue by standard procedures, while EGFR status on ctDNA was tested using dPCR at baseline and at the first reassessment. NGS was used to evaluate patients mutational status at the progression of the disease. RESULTS A total of 40 mEGFR tissue samples were collected. Plasma samples were analyzed for mEGFR before starting the first line, 65 % of patients had detectable mEGFR in ctDNA ("shedders"). Higher ECOG PS (p = 0.04), bilateral localization of primary tumor (p = 0.04), and the presence of intrathoracic/extrathoracic disease (p = 0.05), were associated to mEGFR shedding. Shedders had shorter PFS compared to non-shedders (p = 0.03). Patients with detectable mEGFR in ctDNA at the first radiological assessment exhibited worse PFS compared to patients with ctDNA clearance (p = 0.05). CONCLUSION Our preliminary data demonstrate that specific clinical characteristics predict mEGFR shedding in ctDNA of NSCLC, suggesting a potential clinical applicability for understanding potential false negative results and appropriate reporting in clinical practice.
Collapse
Affiliation(s)
- Martina Ruglioni
- Unit of Clinical Pharmacology and Pharmacogenetics, Department of Clinical and Experimental Medicine, University of Pisa, Italy
| | - Iacopo Petrini
- Unit of Pneumology, Department of Translational Research and New Technologies in Medicine, University Hospital of Pisa, Pisa, Italy
| | - Stefania Crucitta
- Unit of Clinical Pharmacology and Pharmacogenetics, Department of Clinical and Experimental Medicine, University of Pisa, Italy
| | - Andrea Sbrana
- Unit of Pneumology, Department of Translational Research and New Technologies in Medicine, University Hospital of Pisa, Pisa, Italy
| | - Giovanna Irene Luculli
- Unit of Clinical Pharmacology and Pharmacogenetics, Department of Clinical and Experimental Medicine, University of Pisa, Italy
| | - Leila Sadeghi Gol
- Unit of Clinical Pharmacology and Pharmacogenetics, Department of Clinical and Experimental Medicine, University of Pisa, Italy
| | - Carola Forte
- Unit of Clinical Pharmacology and Pharmacogenetics, Department of Clinical and Experimental Medicine, University of Pisa, Italy
| | - Antonio Chella
- Unit of Pneumology, Department of Translational Research and New Technologies in Medicine, University Hospital of Pisa, Pisa, Italy
| | - Christian Rolfo
- Department of Internal Medicine, Division of Medical Oncology, The Arthur G. James Comprehensive Cancer Center, Columbus, OH, USA
| | - Romano Danesi
- Department of Oncology and Hemato-Oncology, University of Milan, Italy.
| | - Marzia Del Re
- Saint Camillus International University of Medical and Health Sciences, Rome, Italy; Direzione Scientifica Fondazione Policlinico A. Gemelli IRCCS, Rome, Italy
| |
Collapse
|
12
|
Ruiz-Vico M, Wetterskog D, Orlando F, Thakali S, Wingate A, Jayaram A, Cremaschi P, Vainauskas O, Brighi N, Castellano-Gauna D, Åström L, Matveev VB, Bracarda S, Esen A, Feyerabend S, Senkus E, López-Brea Piqueras M, Gupta S, Wenstrup R, Boysen G, Martins K, Iwata K, Chowdhury S, Gourgioti G, Serikoff A, Gonzalez-Billalabeitia E, Merseburger AS, Demichelis F, Attard G. Liquid Biopsy in Progressing Prostate Cancer Patients Starting Docetaxel with or Without Enzalutamide: A Biomarker Study of the PRESIDE Phase 3b Trial. Eur Urol Oncol 2025; 8:135-144. [PMID: 39261236 DOI: 10.1016/j.euo.2024.08.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2024] [Accepted: 08/09/2024] [Indexed: 09/13/2024]
Abstract
BACKGROUND AND OBJECTIVE The PRESIDE (NCT02288247) randomized trial demonstrated prolonged progression-free survival (PFS) with continuing enzalutamide beyond progression in metastatic castration-resistant prostate cancer (mCRPC) patients starting docetaxel. This study aims to test the associations of PFS and circulating tumor DNA (ctDNA) prior to and after one cycle (cycle 2 day 1 [C2D1]) of docetaxel and with a liquid biopsy resistance biomarker (LBRB; plasma androgen receptor [AR] gain and/or circulating tumor cells [CTCs] expressing AR splice variant 7 [CTC-AR-V7]) prior to continuation of enzalutamide/placebo. METHODS Patients consenting to the biomarker substudy and donating blood before starting docetaxel with enzalutamide/placebo (N = 157) were included. Sequential plasma DNA samples were characterized with a prostate-cancer bespoke next-generation-sequencing capture panel (PCF_SELECT), and CTCs were assessed for AR-V7 (Epic Sciences, San Diego, CA, USA). Cox models, Kaplan-Meier, and restricted mean survival time (RMST) at 18 mo were calculated. KEY FINDINGS AND LIMITATIONS There was a significant association of worse PFS with pre-docetaxel ctDNA detection (N = 86 (55%), 8.1 vs 10.8 mo hazard ratio [HR] = 1.78, p = 0.004) or persistence/rise of ctDNA at C2D1 (N = 35/134, 5.5 vs 10.9 mo, HR = 1.95, 95% confidence interval [CI] = 1.15-3.30, p = 0.019). LBRB-positive patients (N = 62) had no benefit from continuing enzalutamide with docetaxel (HR = 0.78, 95% CI = 0.41-1.48, p = 0.44; RMST: 7.9 vs 7.1 mo, p = 0.50). Conversely, resistance biomarker-negative patients (N = 87) had significantly prolonged PFS (HR = 0.49, 95% CI = 0.29-0.82, p = 0.006; RMST: 11.5 vs 8.9 mo, p = 0.005). Eight patients were unevaluable. An exploratory analysis identified increased copy-number gains (CDK6/CDK4) at progression on docetaxel. Limitations included relatively low detection of CTC-AR-V7. Validation of impact on overall survival is required. CONCLUSIONS AND CLINICAL IMPLICATIONS Liquid biopsy gives an early indication of docetaxel futility, could guide patient selection for continuing enzalutamide, and identifies cell cycle gene alterations as a potential cause of docetaxel resistance in mCRPC. PATIENT SUMMARY In the PRESIDE biomarker study, we found that detecting circulating tumor DNA in plasma after starting treatment with docetaxel (chemotherapy) for metastatic prostate cancer resistant to androgen deprivation therapy can predict early how long patients will take to respond to treatment. Patients negative for a liquid biopsy resistance biomarker (based on the status of androgen receptor (AR) gene and AR splice variant 7 in circulating tumor cells) benefit from continuing enzalutamide in combination with docetaxel, while patients positive for the resistance biomarker did not. Additionally, we identified alterations in the cell cycle genes CDK6 and CDK4 as a potential genetic cause of resistance to docetaxel, which may support testing of specific drugs targeting these alterations.
Collapse
Affiliation(s)
- Maria Ruiz-Vico
- Oncology Department, University College London Cancer Institute, London, UK; PhD Program in Biomedicine Research, Universidad Complutense de Madrid, Madrid, Spain; Medical Oncology Department, Hospital Universitario 12 de Octubre, Madrid, Spain
| | - Daniel Wetterskog
- Oncology Department, University College London Cancer Institute, London, UK
| | - Francesco Orlando
- Department of Cellular, Computational and Integrative Biology, University of Trento, Trento, Italy
| | - Suparna Thakali
- Oncology Department, University College London Cancer Institute, London, UK
| | - Anna Wingate
- Oncology Department, University College London Cancer Institute, London, UK
| | - Anuradha Jayaram
- Oncology Department, University College London Cancer Institute, London, UK
| | - Paolo Cremaschi
- Oncology Department, University College London Cancer Institute, London, UK
| | | | - Nicole Brighi
- Oncology Department, University College London Cancer Institute, London, UK
| | | | - Lennart Åström
- Department of Immunology, Genetics and Pathology, University of Uppsala, Uppsala, Sweden
| | | | - Sergio Bracarda
- Medical Oncology, Azienda Ospedaliera Santa Maria, Terni, Italy
| | - Adil Esen
- Department of Urology, Dokuz Eylul University, Konak, Turkey
| | - Susan Feyerabend
- Studienpraxis Urologie, Medius Klinik Nürtingen, Nürtingen, Germany
| | - Elżbieta Senkus
- Department of Oncology and Radiotherapy, Medical University of Gdansk, Gdansk, Poland
| | | | - Santosh Gupta
- Translational Research, Epic Sciences Inc, San Diego, CA, USA
| | - Rick Wenstrup
- Translational Research, Epic Sciences Inc, San Diego, CA, USA
| | | | | | | | - Simon Chowdhury
- Medical Oncology, Guy's and St Thomas' NHS Foundation Trust, King's College London, London, UK
| | | | | | - Enrique Gonzalez-Billalabeitia
- PhD Program in Biomedicine Research, Universidad Complutense de Madrid, Madrid, Spain; Medical Oncology Department, Hospital Universitario 12 de Octubre, Madrid, Spain
| | - Axel S Merseburger
- Department of Urology, Universitätsklinikum Schleswig-Holstein, Lübeck, Germany
| | - Francesca Demichelis
- Department of Cellular, Computational and Integrative Biology, University of Trento, Trento, Italy
| | - Gerhardt Attard
- Oncology Department, University College London Cancer Institute, London, UK.
| |
Collapse
|
13
|
Saw SPL, Zhong WZ, Fu R, Li MSC, Goto Y, Fox SB, Yatabe Y, Ong BH, Ng CSH, Lee DDW, Cam Phuong P, Park IK, Yang JCH, Tsuboi M, Tho LM, John T, Hsu HH, Tan DSW, Mok TSK, Reungwetwattana T, Singh N. Asian Thoracic Oncology Research Group expert consensus statement on the peri-operative management of non-small cell lung cancer. Lung Cancer 2025; 200:108076. [PMID: 39799810 DOI: 10.1016/j.lungcan.2024.108076] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2024] [Revised: 12/24/2024] [Accepted: 12/29/2024] [Indexed: 01/15/2025]
Abstract
The peri-operative management of non-small cell lung cancer (NSCLC) in earlier stage disease has seen significant advances in recent years with the incorporation of immune checkpoint inhibitors and targeted therapy. However, many unanswered questions and challenges remain, including the application of clinical trial data to routine clinical practice. Recognising the unique demographic profile of Asian patients with NSCLC and heterogeneous healthcare systems, the Asian Thoracic Oncology Research Group (ATORG) convened a consensus meeting in Singapore on 26 April 2024 to discuss relevant issues spanning diagnostic testing to post-neoadjuvant treatment considerations and future directions. An interdisciplinary group of 19 experts comprising medical oncologists, thoracic surgeons, radiation oncologists, pulmonologists and pathologists from Singapore, Hong Kong, Mainland China, Korea, Japan, Taiwan, India, Malaysia, Thailand, Vietnam and Australia met to discuss emerging data, identify existing gaps in clinical care and develop a multidisciplinary, multinational expert consensus statement on the peri-operative management of NSCLC tailored to the Asia-Pacific region.
Collapse
Affiliation(s)
- Stephanie P L Saw
- Division of Medical Oncology, National Cancer Centre Singapore, 30 Hospital Boulevard, Singapore 168583, Singapore.
| | - Wen-Zhao Zhong
- Guangdong Lung Cancer Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou 510080, China
| | - Rui Fu
- Guangdong Lung Cancer Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou 510080, China
| | - Molly S C Li
- Department of Clinical Oncology, The Chinese University of Hong Kong, Hong Kong China
| | - Yasushi Goto
- National Cancer Center Hospital, Department of Thoracic Oncology, 5-1-1, Tsukiji, Chuo-ku, Tokyo 104-0045, Japan
| | - Stephen B Fox
- Department of Pathology, Peter MacCallum Cancer Centre and University of Melbourne, Australia
| | - Yasushi Yatabe
- Department of Diagnostic Pathology, National Cancer Center, Tokyo, Japan
| | - Boon-Hean Ong
- Department of Cardiothoracic Surgery, National Heart Centre Singapore, 5 Hospital Drive, Singapore 169609, Singapore
| | - Calvin S H Ng
- Department of Surgery, The Chinese University of Hong Kong, Hong Kong Special Administrative Region
| | - David D W Lee
- Department of Clinical Oncology, University of Malaya, Kuala Lumpur, Malaysia
| | - Pham Cam Phuong
- The Nuclear Medicine and Oncology Center, Bach Mai Hospital, 78 Giai Phong Street, Dong Da, Hanoi, Viet Nam
| | - In Kyu Park
- Department of Thoracic and Cardiovascular Surgery, Seoul National University Hospital, Seoul, Korea
| | - James C H Yang
- Department of Oncology, National Taiwan University Hospital, Taiwan
| | - Masahiro Tsuboi
- Department of Thoracic Surgery and Oncology, National Cancer Center Hospital East, Kashiwa, Japan
| | - Lye Mun Tho
- Department of Oncology, Beacon Hospital, Petaling Jaya, Malaysia
| | - Thomas John
- Medical Oncologist, Peter MacCallum Cancer Centre and University of Melbourne, Australia
| | - Hsao-Hsun Hsu
- Department of Surgical Oncology and Surgery, National Taiwan University Cancer Center and College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Daniel S W Tan
- Division of Medical Oncology, National Cancer Centre Singapore, 30 Hospital Boulevard, Singapore 168583, Singapore
| | - Tony S K Mok
- Department of Clinical Oncology, The Chinese University of Hong Kong, Hong Kong China
| | - Thanyanan Reungwetwattana
- Division of Medical Oncology, Department of Medicine, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Bangkok, Thailand
| | - Navneet Singh
- Lung Cancer Clinic, Postgraduate Institute of Medical Education and Research (PGIMER), Chandigarh, India
| |
Collapse
|
14
|
Wu X, Yao X, Chen Z, Xu H. Re: Iver Nordentoft, Sia Viborg Lindskrog, Karin Birkenkamp-Demtröder, et al. Whole-genome Mutational Analysis for Tumor-informed Detection of Circulating Tumor DNA in Patients with Urothelial Carcinoma. Eur Urol. 2024;86:301-311. Eur Urol 2025; 87:e36-e37. [PMID: 39117526 DOI: 10.1016/j.eururo.2024.07.021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2024] [Accepted: 07/25/2024] [Indexed: 08/10/2024]
Affiliation(s)
- Xiaoliang Wu
- Department and Institute of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xiangyang Yao
- Department of Urology, Zhongnan Hospital, Wuhan University, Wuhan, China
| | - Zhong Chen
- Department and Institute of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Hua Xu
- Department of Urology, Zhongnan Hospital, Wuhan University, Wuhan, China; Hubei Engineering Research Center for Cancer Precision Diagnosis and Treatment and Translational Medicine, Wuhan, China.
| |
Collapse
|
15
|
Jacobsen CM, Matos do Canto L, Kahns S, Hansen TF, Andersen RF. What the Clinician Needs to Know About Laboratory Analyses of Circulating Tumor DNA. Clin Colorectal Cancer 2025:S1533-0028(25)00003-9. [PMID: 39956753 DOI: 10.1016/j.clcc.2025.01.003] [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: 04/25/2024] [Revised: 12/02/2024] [Accepted: 01/20/2025] [Indexed: 02/18/2025]
Abstract
Liquid biopsies offer the possibility to evaluate cancer patients using noninvasive approaches. Circulating cell-free DNA (ccfDNA) is 1 of the most used and promising sources. Detecting tumor DNA among ccfDNA (ctDNA) can be used for early cancer detection, treatment response assessment, prognosis, and predictive evaluations. Providing analyses that can increase the quality of patient treatment is very much a joint effort between laboratory scientists and clinicians. With its use approaching clinical practice, it is important for clinicians to be familiar with the basic concepts and analyses behind ctDNA results in a similar way as laboratory scientists should have knowledge of the clinical needs to provide relevant analyses. In this Perspective, we describe the whole process of ctDNA analyses, from the preanalytical standards to reporting/analyzing results, and highlight some important factors that need to be addressed in the process of implementing them to clinical practice.
Collapse
Affiliation(s)
- Cecilie Mondrup Jacobsen
- Department of Biochemistry and Immunology, Vejle Hospital, University Hospital of Southern Denmark, Vejle, Denmark; Department of Oncology, Vejle Hospital, University Hospital of Southern Denmark, Vejle, Denmark
| | - Luisa Matos do Canto
- Department of Biochemistry and Immunology, Vejle Hospital, University Hospital of Southern Denmark, Vejle, Denmark; Department of Oncology, Vejle Hospital, University Hospital of Southern Denmark, Vejle, Denmark
| | - Søren Kahns
- Department of Biochemistry and Immunology, Vejle Hospital, University Hospital of Southern Denmark, Vejle, Denmark
| | - Torben Frøstrup Hansen
- Department of Oncology, Vejle Hospital, University Hospital of Southern Denmark, Vejle, Denmark; Department of Regional Health Research, University of Southern Denmark, Odense C, Denmark
| | - Rikke Fredslund Andersen
- Department of Biochemistry and Immunology, Vejle Hospital, University Hospital of Southern Denmark, Vejle, Denmark; Department of Oncology, Vejle Hospital, University Hospital of Southern Denmark, Vejle, Denmark.
| |
Collapse
|
16
|
Deng Q, Li W, Huang Y, Wang H, Zhou X, Guan Z, Cheng B, Wang Y. Immunolipid magnetic bead-based circulating tumor cell sorting: a novel approach for pathological staging of colorectal cancer. Front Oncol 2025; 14:1531972. [PMID: 39927117 PMCID: PMC11803635 DOI: 10.3389/fonc.2024.1531972] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2024] [Accepted: 12/31/2024] [Indexed: 02/11/2025] Open
Abstract
Objective This study aimed to assess whether circulating tumor cells (CTCs) from colorectal cancer (CRC) could be used as an alternative to tissue samples for genetic mutation testing, overcoming the challenge of difficult tumor tissue acquisition. Methods We developed an immunolipid magnetic bead (IMB) system modified with antibodies against epithelial cell adhesion molecule (EpCAM) and vimentin to efficiently separate CTCs. We prepared EpCAM-modified IMBs (Ep-IMBs) and vimentin-modified IMBs (Vi-IMBs). The separation efficiency of the system was evaluated via in vitro experiments and by capturing and counting CTCs in blood samples from 23 CRC patients and 20 healthy controls. Hotspot mutations in patient tissue samples were identified via next-generation sequencing (NGS), whereas mutations in blood CTCs were detected via Sanger sequencing. The concordance between hotspot mutations in tumor tissue and blood CTCs was analyzed. Results The CTC sorting system exhibited good dispersion, stability, and low cytotoxicity, with a specificity of 90.54% and a sensitivity of 89.07%. CRC patients had an average of 8.39 CTCs per 7.5 mL of blood, whereas healthy controls had 0.09 per 7.5 mL of blood. The consistency of gene mutations was as follows: TP53 (91.31%), PIK3CA (76.00%), KRAS (85.36%), BRAF (51.00%), APC (65.67%), and EGFR (74.00%), with an overall gene mutation consistency of 85.06%. Conclusion Our CTC sorting system, which is based on Ep-IMBs and Vi-IMBs, effectively captures CTCs in the peripheral blood of CRC patients and enables clinical hotspot gene mutation testing via these enriched CTCs. This system partially solves the problem of difficult tumor tissue sample collection and provides a reference for gene mutation testing in early diagnosis, therapeutic efficacy evaluation, prognosis assessment, and minimal metastasis detection in CRC patients, showing significant potential for clinical application, especially in targeted therapy gene testing for CRC.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Yao Wang
- Department of Gastrointestinal Surgery, Zhongshan People’s Hospital, Zhongshan, Guangdong, China
| |
Collapse
|
17
|
Furlano K, Keshavarzian T, Biernath N, Fendler A, de Santis M, Weischenfeldt J, Lupien M. Epigenomics-guided precision oncology: Chromatin variants in prostate tumor evolution. Int J Cancer 2025. [PMID: 39853587 DOI: 10.1002/ijc.35327] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2024] [Revised: 12/17/2024] [Accepted: 01/02/2025] [Indexed: 01/26/2025]
Abstract
Prostate cancer is a common malignancy that in 5%-30% leads to treatment-resistant and highly aggressive disease. Metastasis-potential and treatment-resistance is thought to rely on increased plasticity of the cancer cells-a mechanism whereby cancer cells alter their identity to adapt to changing environments or therapeutic pressures to create cellular heterogeneity. To understand the molecular basis of this plasticity, genomic studies have uncovered genetic variants to capture clonal heterogeneity of primary tumors and metastases. As cellular plasticity is largely driven by non-genetic events, complementary studies in cancer epigenomics are now being conducted to identify chromatin variants. These variants, defined as genomic loci in cancer cells that show changes in chromatin state due to the loss or gain of epigenomic marks, inclusive of histone post-translational modifications, DNA methylation and histone variants, are considered the fundamental units of epigenomic heterogeneity. In prostate cancer chromatin variants hold the promise of guiding the new era of precision oncology. In this review, we explore the role of epigenomic heterogeneity in prostate cancer, focusing on how chromatin variants contribute to tumor evolution and therapy resistance. We therefore discuss their impact on cellular plasticity and stochastic events, highlighting the value of single-cell sequencing and liquid biopsy epigenomic assays to uncover new therapeutic targets and biomarkers. Ultimately, this review aims to support a new era of precision oncology, utilizing insights from epigenomics to improve prostate cancer patient outcomes.
Collapse
Affiliation(s)
- Kira Furlano
- Department of Urology, Charité- Universitätsmedizin Berlin, Berlin, Germany
| | - Tina Keshavarzian
- Princess Margaret Cancer Centre, University Health Network, Toronto, Canada
- Department of Medical Biophysics, University of Toronto, Toronto, Canada
| | - Nadine Biernath
- Department of Urology, Charité- Universitätsmedizin Berlin, Berlin, Germany
| | - Annika Fendler
- Department of Urology, Charité- Universitätsmedizin Berlin, Berlin, Germany
| | - Maria de Santis
- Department of Urology, Charité- Universitätsmedizin Berlin, Berlin, Germany
- Department of Urology, Medical University of Vienna, Vienna, Austria
| | - Joachim Weischenfeldt
- Department of Urology, Charité- Universitätsmedizin Berlin, Berlin, Germany
- Biotech Research & Innovation Centre (BRIC), The Finsen Laboratory, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
| | - Mathieu Lupien
- Princess Margaret Cancer Centre, University Health Network, Toronto, Canada
- Department of Medical Biophysics, University of Toronto, Toronto, Canada
- Ontario Institute for Cancer Research, Toronto, Ontario, Canada
| |
Collapse
|
18
|
Ryssel H, Ahlborn LB, Dejanovic D, Keller SH, Pøhl M, Østrup O, Loft A, Fischer BM, Langer SW, Kjaer A, Christensen TN. Circulating Tumor DNA and [ 18F]FDG-PET for Early Response Assessment in Patients with Advanced NSCLC. Diagnostics (Basel) 2025; 15:247. [PMID: 39941177 PMCID: PMC11817175 DOI: 10.3390/diagnostics15030247] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2024] [Revised: 01/08/2025] [Accepted: 01/18/2025] [Indexed: 02/16/2025] Open
Abstract
Background/Objectives: Identifying treatment failure at earlier time points to could spare cancer patients from ineffective treatment and side effects. In this study, circulating tumor DNA (ctDNA) and [18F]FDG-PET/CT were investigated during the first cycle of anticancer therapy in patients with advanced non-small cell lung cancer (NSCLC) to explore their potential for early response evaluation. Methods: Patients with advanced NSCLC receiving first-line therapy with immune checkpoint inhibitors and/or chemotherapy were included. CtDNA and [18F]FDG-PET/CT assessments were conducted before treatment and at weeks 1 and 3 during the first cycle of therapy. ctDNA quantification was performed using a targeted next-generation sequencing (NGS) panel, and the least favorable change in any mutated allele frequency at a given time was used for analysis. [18F]FDG-PET/CT was quantified using sumSULpeak and metabolic tumor volume (MTV4.0). Early changes in ctDNA levels and [18F]FDG-PET parameters were compared with final treatment response, measured by RECIST after 12 weeks, as well as progression-free survival and overall survival. Results: Of the sixteen included patients, eight were non-responders. ctDNA mutations were detected in baseline blood samples in eight patients. Changes in ctDNA level, MTV4.0, and sumSULpeak at week 3 indicated response in 7 out of 8 patients, 13 out of 15 patients, and 9 out of 15 patients, respectively. At week 3, no false increases were seen with ctDNA and MTV4.0. Conclusions: These results suggest that early changes in ctDNA and [18F]FDG-PET/CT at 3 weeks of treatment could be used to early assess treatment response. Increased levels of ctDNA and MTV4.0 at week 3 were only observed in patients with treatment failure.
Collapse
Affiliation(s)
- Heidi Ryssel
- Department of Clinical Physiology and Nuclear Medicine, Rigshospitalet, Copenhagen University Hospital, 9 Blegdamsvej, 2100 Copenhagen, Denmark; (H.R.); (D.D.); (S.H.K.); (A.L.); (B.M.F.); (T.N.C.)
- Department of Oncology, Rigshospitalet, Copenhagen University Hospital, 9 Blegdamsvej, 2100 Copenhagen, Denmark; (M.P.); (S.W.L.)
- Cluster of Molecular Imaging, Copenhagen University, Panum Institution, 3 Blegdamsvej, 2200 Copenhagen, Denmark
| | - Lise Barlebo Ahlborn
- Department of Genomic Medicine, Rigshospitalet, 9 Blegdamsvej, 2100 Copenhagen, Denmark; (L.B.A.); (O.Ø.)
| | - Danijela Dejanovic
- Department of Clinical Physiology and Nuclear Medicine, Rigshospitalet, Copenhagen University Hospital, 9 Blegdamsvej, 2100 Copenhagen, Denmark; (H.R.); (D.D.); (S.H.K.); (A.L.); (B.M.F.); (T.N.C.)
| | - Sune Hoegild Keller
- Department of Clinical Physiology and Nuclear Medicine, Rigshospitalet, Copenhagen University Hospital, 9 Blegdamsvej, 2100 Copenhagen, Denmark; (H.R.); (D.D.); (S.H.K.); (A.L.); (B.M.F.); (T.N.C.)
| | - Mette Pøhl
- Department of Oncology, Rigshospitalet, Copenhagen University Hospital, 9 Blegdamsvej, 2100 Copenhagen, Denmark; (M.P.); (S.W.L.)
| | - Olga Østrup
- Department of Genomic Medicine, Rigshospitalet, 9 Blegdamsvej, 2100 Copenhagen, Denmark; (L.B.A.); (O.Ø.)
| | - Annika Loft
- Department of Clinical Physiology and Nuclear Medicine, Rigshospitalet, Copenhagen University Hospital, 9 Blegdamsvej, 2100 Copenhagen, Denmark; (H.R.); (D.D.); (S.H.K.); (A.L.); (B.M.F.); (T.N.C.)
| | - Barbara Malene Fischer
- Department of Clinical Physiology and Nuclear Medicine, Rigshospitalet, Copenhagen University Hospital, 9 Blegdamsvej, 2100 Copenhagen, Denmark; (H.R.); (D.D.); (S.H.K.); (A.L.); (B.M.F.); (T.N.C.)
- Department of Clinical Medicine, University of Copenhagen, 2200 Copenhagen, Denmark
- School of Biomedical Engineering and Imaging Sciences, Kings College London, London WC2R 2LS, UK
| | - Seppo Wang Langer
- Department of Oncology, Rigshospitalet, Copenhagen University Hospital, 9 Blegdamsvej, 2100 Copenhagen, Denmark; (M.P.); (S.W.L.)
- Department of Clinical Medicine, University of Copenhagen, 2200 Copenhagen, Denmark
| | - Andreas Kjaer
- Department of Clinical Physiology and Nuclear Medicine, Rigshospitalet, Copenhagen University Hospital, 9 Blegdamsvej, 2100 Copenhagen, Denmark; (H.R.); (D.D.); (S.H.K.); (A.L.); (B.M.F.); (T.N.C.)
- Cluster of Molecular Imaging, Copenhagen University, Panum Institution, 3 Blegdamsvej, 2200 Copenhagen, Denmark
- Department of Clinical Medicine, University of Copenhagen, 2200 Copenhagen, Denmark
| | - Tine Nøhr Christensen
- Department of Clinical Physiology and Nuclear Medicine, Rigshospitalet, Copenhagen University Hospital, 9 Blegdamsvej, 2100 Copenhagen, Denmark; (H.R.); (D.D.); (S.H.K.); (A.L.); (B.M.F.); (T.N.C.)
| |
Collapse
|
19
|
Geng Y, Yin T, Li Y, He K, Zou B, Yu J, Sun X, Zhang T, Teng F. Computed Tomography-Based Radiomics and Genomics Analyses for Survival Prediction of Stage III Unresectable Non-Small Cell Lung Cancer Treated With Definitive Chemoradiotherapy and Immunotherapy. Mol Carcinog 2025. [PMID: 39835605 DOI: 10.1002/mc.23883] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2024] [Revised: 12/26/2024] [Accepted: 01/02/2025] [Indexed: 01/22/2025]
Abstract
The standard therapy for locally unresectable advanced non-small cell lung cancer (NSCLC) is comprised of chemoradiotherapy (CRT) before immunotherapy (IO) consolidation. However, how to predict treatment outcomes and recognize patients that will benefit from IO remain unclear. This study aimed to identify prognostic biomarkers by integrating computed tomography (CT)-based radiomics and genomics. Specifically, our research involved 165 patients suffering from unresectable Stage III NSCLC. Cohort 1 (IO following CRT) was divided into D1 (n = 74), D2 (n = 32), and D3 (n = 26) sets, and the remaining 33 patients treated with CRT alone were grouped in D4. According to the CT images of primary tumor regions, radiomic features were analyzed through the least absolute shrinkage and selection operator (LASSO) regression. The Rad-score was figured out to forecast the progression-free survival (PFS). According to the Rad-score, patients were divided into high and low risk groups. Next-generation sequencing was implemented on peripheral blood and tumor tissue samples in the D3 and D4 cohorts. The maximum somatic allele frequency (MSAF) about circulating tumor DNA levels was assessed. Mismatch repair and switching/sucrose non-fermenting signaling pathways were significantly enriched in the low-risk group compared to the high-risk group (p < 0.05). Moreover, patients with MSAF ≥ 1% and those showing a decrease in MSAF after treatment significantly benefited from IO. This study developed a radiomics model predicting PFS after CRT and IO in Stage III NSCLC and constructed a radio-genomic map to identify underlying biomarkers, supplying valuable insights for cancer biology.
Collapse
Affiliation(s)
- Yuxin Geng
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, China
| | - Tianwen Yin
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
- Institute of Radiation Oncology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Key Laboratory of Precision Radiation Oncology, Wuhan, China
| | - Yikun Li
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, China
| | - Kaixing He
- Department of Breast Surgery, Shengli Oilfield Central Hospital, Dongying, Shandong, China
| | - Bingwen Zou
- Department of Radiation Oncology, West China Hospital of Sichuan University, Chengdu, Sichuan, China
| | - Jinming Yu
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, China
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
- Institute of Radiation Oncology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Key Laboratory of Precision Radiation Oncology, Wuhan, China
| | - Xiao Sun
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, China
| | - Tao Zhang
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
- Institute of Radiation Oncology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Key Laboratory of Precision Radiation Oncology, Wuhan, China
| | - Feifei Teng
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, China
| |
Collapse
|
20
|
Martínez-Vila C, Teixido C, Aya F, Martín R, González-Navarro EA, Alos L, Castrejon N, Arance A. Detection of Circulating Tumor DNA in Liquid Biopsy: Current Techniques and Potential Applications in Melanoma. Int J Mol Sci 2025; 26:861. [PMID: 39859576 PMCID: PMC11766255 DOI: 10.3390/ijms26020861] [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: 11/21/2024] [Revised: 01/10/2025] [Accepted: 01/16/2025] [Indexed: 01/27/2025] Open
Abstract
The treatment landscape for advanced melanoma has transformed significantly with the advent of BRAF and MEK inhibitors (BRAF/MEKi) targeting BRAFV600 mutations, as well as immune checkpoint inhibitors (ICI) like anti-PD-1 monotherapy or its combinations with anti-CTLA-4 or anti-LAG-3. Despite that, many patients still do not benefit from these treatments at all or develop resistance mechanisms. Therefore, prognostic and predictive biomarkers are needed to identify patients who should switch or escalate their treatment strategies or initiate an intensive follow-up. In melanoma, liquid biopsy has shown promising results, with a potential role in predicting relapse in resected high-risk patients or in disease monitoring during the treatment of advanced disease. Several components in peripheral blood have been analyzed, such as circulating tumor cells (CTCs), cell-free DNA (cfDNA), and circulant tumoral DNA (ctDNA), which have turned out to be particularly promising. To analyze ctDNA in blood, different techniques have proven to be useful, including digital droplet polymerase chain reaction (ddPCR) to detect specific mutations and, more recently, next-generation sequencing (NGS) techniques, which allow analyzing a broader repertoire of the mutation landscape of each patient. In this review, our goal is to update the current understanding of liquid biopsy, focusing on the use of ctDNA as a biological material in the daily clinical management of melanoma patients, in particular those with advanced disease treated with ICI.
Collapse
Affiliation(s)
- Clara Martínez-Vila
- Department of Medical Oncology, Althaia Xarxa Assistencial Universitària de Manresa, Dr. Joan Soler, 1–3, 08243 Manresa, Spain;
- Programa de Doctorat en Medicina i Recerca Translacional, Facultat de Medicina, Universitat de Barcelona, 08036 Barcelona, Spain
- Institut de Recerca i Innovació en Ciències de la Vida i de la Salut a la Catalunya Central (IRIS-CC), Roda 70, 08500 Vic, Spain
| | - Cristina Teixido
- Department of Pathology, Hospital Clínic of Barcelona, University of Barcelona, Villarroel 170, 08036 Barcelona, Spain; (C.T.); (L.A.); (N.C.)
- August Pi i Sunyer Biomedical Research Institute (IDIBAPS), Rosselló 149, 08036 Barcelona, Spain; (F.A.); (R.M.); (E.A.G.-N.)
| | - Francisco Aya
- August Pi i Sunyer Biomedical Research Institute (IDIBAPS), Rosselló 149, 08036 Barcelona, Spain; (F.A.); (R.M.); (E.A.G.-N.)
- Department of Medical Oncology, Hospital Clínic of Barcelona, University of Barcelona, Villarroel 170, 08036 Barcelona, Spain
| | - Roberto Martín
- August Pi i Sunyer Biomedical Research Institute (IDIBAPS), Rosselló 149, 08036 Barcelona, Spain; (F.A.); (R.M.); (E.A.G.-N.)
- Department of Medical Oncology, Hospital Clínic of Barcelona, University of Barcelona, Villarroel 170, 08036 Barcelona, Spain
| | - Europa Azucena González-Navarro
- August Pi i Sunyer Biomedical Research Institute (IDIBAPS), Rosselló 149, 08036 Barcelona, Spain; (F.A.); (R.M.); (E.A.G.-N.)
- Department of Immunology, Hospital Clínic of Barcelona, University of Barcelona, Villarroel 170, 08036 Barcelona, Spain
| | - Llucia Alos
- Department of Pathology, Hospital Clínic of Barcelona, University of Barcelona, Villarroel 170, 08036 Barcelona, Spain; (C.T.); (L.A.); (N.C.)
- August Pi i Sunyer Biomedical Research Institute (IDIBAPS), Rosselló 149, 08036 Barcelona, Spain; (F.A.); (R.M.); (E.A.G.-N.)
| | - Natalia Castrejon
- Department of Pathology, Hospital Clínic of Barcelona, University of Barcelona, Villarroel 170, 08036 Barcelona, Spain; (C.T.); (L.A.); (N.C.)
| | - Ana Arance
- August Pi i Sunyer Biomedical Research Institute (IDIBAPS), Rosselló 149, 08036 Barcelona, Spain; (F.A.); (R.M.); (E.A.G.-N.)
- Department of Medical Oncology, Hospital Clínic of Barcelona, University of Barcelona, Villarroel 170, 08036 Barcelona, Spain
| |
Collapse
|
21
|
Zhang Y, Yuan X. Minimal residue disease detection in early-stage breast cancer: a review. Mol Biol Rep 2025; 52:106. [PMID: 39777588 DOI: 10.1007/s11033-024-10198-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2024] [Accepted: 12/20/2024] [Indexed: 01/11/2025]
Abstract
Over the past five years, circulating tumor DNA (ctDNA) testing has emerged as a game-changer in cancer research, serving as a less invasive and highly sensitive method to monitor tumor dynamics. CtDNA testing has a wide range of potential applications in breast cancer (BC) management, including diagnosis, monitoring treatment responses, identifying resistance mutations, predicting prognosis, and detecting future relapses. In this review, we focus on the prognostic and predictive value of ctDNA testing for BC in both neoadjuvant and adjuvant settings. We also examine the rationale behind mainstream minimal residue disease (MRD) tracking methods and highlight key considerations for successful MRD testing. Clinical evidence has shown that ctDNA-based MRD testing can accurately detect molecular relapse 8-12 months before clinical relapse in early-stage BC. Compared to advanced-stage BC, detecting ctDNA in early-stage BC is more challenging and requires ultra-sensitive testing methods due to the low levels of ctDNA released into the bloodstream, particularly in post-surgical settings, after effective neoadjuvant chemotherapy, and in late adjuvant settings that require longer follow-up. Therefore, future efforts are needed to generate additional clinical evidence in these settings to support the clinical utility and widespread adoption of ctDNA-based MRD testing.
Collapse
Affiliation(s)
- Yuan Zhang
- College of Art and Science, Northeast Agricultural University, Changjiang Road No. 600, Harbin, 150030, China
- Shuwen Biotech Co., Ltd., Moganshan National High tech Zone, Building 3, No. 333, Changhong Middle Street, Deqing, China
| | - Xiaoying Yuan
- Shuwen Biotech Co., Ltd., Moganshan National High tech Zone, Building 3, No. 333, Changhong Middle Street, Deqing, China.
| |
Collapse
|
22
|
Argento G, Rendina EA, Maurizi G. Advancing Thoracic Surgical Oncology in the Era of Precision Medicine. Cancers (Basel) 2025; 17:115. [PMID: 39796742 PMCID: PMC11720116 DOI: 10.3390/cancers17010115] [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: 11/16/2024] [Revised: 12/30/2024] [Accepted: 12/31/2024] [Indexed: 01/13/2025] Open
Abstract
The landscape of surgical oncology is rapidly evolving with the advent of precision medicine, driven by breakthroughs in genomics and proteomics. This article explores how integrating molecular data is transforming surgical decision-making and enabling personalized treatment strategies. We examine emerging technologies such as next-generation sequencing, proteomic analysis, and molecular imaging, which provide critical insights into tumor biology and guide surgical interventions. The article also highlights the application of genomic and proteomic data in preoperative planning and the development of personalized resection strategies. Additionally, we will address the current challenges and future opportunities in this rapidly evolving field, emphasizing the need for continuous education, interdisciplinary collaboration, and ongoing research to fully realize the potential of precision medicine in thoracic surgical oncology, paving the way for more effective and individualized cancer treatments.
Collapse
Affiliation(s)
| | | | - Giulio Maurizi
- Department of Thoracic Surgery, University of Rome La Sapienza, Sant’Andrea Hospital, 00189 Rome, Italy; (G.A.); (E.A.R.)
| |
Collapse
|
23
|
Black JRM, Bartha G, Abbott CW, Boyle SM, Karasaki T, Li B, Chen R, Harris J, Veeriah S, Colopi M, Bakir MA, Liu WK, Lyle J, Navarro FCP, Northcott J, Pyke RM, Hill MS, Thol K, Huebner A, Bailey C, Colliver EC, Martínez-Ruiz C, Grigoriadis K, Pawlik P, Moore DA, Marinelli D, Shutkever OG, Murphy C, Sivakumar M, Shaw JA, Hackshaw A, McGranahan N, Jamal-Hanjani M, Frankell AM, Chen RO, Swanton C. Ultrasensitive ctDNA detection for preoperative disease stratification in early-stage lung adenocarcinoma. Nat Med 2025; 31:70-76. [PMID: 39806071 PMCID: PMC11750713 DOI: 10.1038/s41591-024-03216-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2023] [Accepted: 07/29/2024] [Indexed: 01/16/2025]
Abstract
Circulating tumor DNA (ctDNA) detection can predict clinical risk in early-stage tumors. However, clinical applications are constrained by the sensitivity of clinically validated ctDNA detection approaches. NeXT Personal is a whole-genome-based, tumor-informed platform that has been analytically validated for ultrasensitive ctDNA detection at 1-3 ppm of ctDNA with 99.9% specificity. Through an analysis of 171 patients with early-stage lung cancer from the TRACERx study, we detected ctDNA pre-operatively within 81% of patients with lung adenocarcinoma (LUAD), including 53% of those with pathological TNM (pTNM) stage I disease. ctDNA predicted worse clinical outcome, and patients with LUAD with <80 ppm preoperative ctDNA levels (the 95% limit of detection of a ctDNA detection approach previously published in TRACERx) experienced reduced overall survival compared with ctDNA-negative patients with LUAD. Although prospective studies are needed to confirm the clinical utility of the assay, these data show that our approach has the potential to improve disease stratification in early-stage LUADs.
Collapse
Affiliation(s)
- James R M Black
- Cancer Research UK Lung Cancer Centre of Excellence, University College London Cancer Institute, London, UK
- Cancer Evolution and Genome Instability Laboratory., The Francis Crick Institute, London, UK
| | | | | | | | - Takahiro Karasaki
- Cancer Research UK Lung Cancer Centre of Excellence, University College London Cancer Institute, London, UK
- Cancer Evolution and Genome Instability Laboratory., The Francis Crick Institute, London, UK
- Cancer Metastasis Laboratory, University College London Cancer Institute, London, UK
- Department of Thoracic Surgery, Respiratory Center, Toranomon Hospital, Tokyo, Japan
| | | | - Rui Chen
- Personalis Inc., Fremont, CA, USA
| | | | - Selvaraju Veeriah
- Cancer Research UK Lung Cancer Centre of Excellence, University College London Cancer Institute, London, UK
| | - Martina Colopi
- Cancer Research UK Lung Cancer Centre of Excellence, University College London Cancer Institute, London, UK
| | - Maise Al Bakir
- Cancer Research UK Lung Cancer Centre of Excellence, University College London Cancer Institute, London, UK
- Cancer Evolution and Genome Instability Laboratory., The Francis Crick Institute, London, UK
| | - Wing Kin Liu
- Cancer Research UK Lung Cancer Centre of Excellence, University College London Cancer Institute, London, UK
| | | | | | | | | | - Mark S Hill
- Cancer Research UK Lung Cancer Centre of Excellence, University College London Cancer Institute, London, UK
- Cancer Evolution and Genome Instability Laboratory., The Francis Crick Institute, London, UK
| | - Kerstin Thol
- Cancer Research UK Lung Cancer Centre of Excellence, University College London Cancer Institute, London, UK
- Cancer Genome Evolution Research Group, Cancer Research UK Lung Cancer Centre of Excellence, University College London Cancer Institute, London, UK
| | - Ariana Huebner
- Cancer Research UK Lung Cancer Centre of Excellence, University College London Cancer Institute, London, UK
- Cancer Evolution and Genome Instability Laboratory., The Francis Crick Institute, London, UK
- Cancer Genome Evolution Research Group, Cancer Research UK Lung Cancer Centre of Excellence, University College London Cancer Institute, London, UK
| | - Chris Bailey
- Cancer Research UK Lung Cancer Centre of Excellence, University College London Cancer Institute, London, UK
- Cancer Evolution and Genome Instability Laboratory., The Francis Crick Institute, London, UK
| | - Emma C Colliver
- Cancer Research UK Lung Cancer Centre of Excellence, University College London Cancer Institute, London, UK
- Cancer Evolution and Genome Instability Laboratory., The Francis Crick Institute, London, UK
| | - Carlos Martínez-Ruiz
- Cancer Research UK Lung Cancer Centre of Excellence, University College London Cancer Institute, London, UK
- Cancer Genome Evolution Research Group, Cancer Research UK Lung Cancer Centre of Excellence, University College London Cancer Institute, London, UK
| | - Kristiana Grigoriadis
- Cancer Research UK Lung Cancer Centre of Excellence, University College London Cancer Institute, London, UK
- Cancer Evolution and Genome Instability Laboratory., The Francis Crick Institute, London, UK
- Cancer Genome Evolution Research Group, Cancer Research UK Lung Cancer Centre of Excellence, University College London Cancer Institute, London, UK
| | - Piotr Pawlik
- Cancer Research UK Lung Cancer Centre of Excellence, University College London Cancer Institute, London, UK
- Cancer Genome Evolution Research Group, Cancer Research UK Lung Cancer Centre of Excellence, University College London Cancer Institute, London, UK
| | - David A Moore
- Cancer Research UK Lung Cancer Centre of Excellence, University College London Cancer Institute, London, UK
- Cancer Evolution and Genome Instability Laboratory., The Francis Crick Institute, London, UK
- Department of Cellular Pathology, University College London Hospitals, London, UK
| | - Daniele Marinelli
- Cancer Research UK Lung Cancer Centre of Excellence, University College London Cancer Institute, London, UK
- Cancer Genome Evolution Research Group, Cancer Research UK Lung Cancer Centre of Excellence, University College London Cancer Institute, London, UK
- Department of Experimental Medicine, Sapienza University, Rome, Italy
| | - Oliver G Shutkever
- Cancer Research UK Lung Cancer Centre of Excellence, University College London Cancer Institute, London, UK
| | - Cian Murphy
- Cancer Research UK Lung Cancer Centre of Excellence, University College London Cancer Institute, London, UK
- Cancer Evolution and Genome Instability Laboratory., The Francis Crick Institute, London, UK
| | - Monica Sivakumar
- Cancer Research UK Lung Cancer Centre of Excellence, University College London Cancer Institute, London, UK
| | - Jacqui A Shaw
- Leicester NIHR BRC & University of Leicester, Leicester, UK
| | - Allan Hackshaw
- Cancer Research UK & UCL Cancer Trials Centre, London, UK
| | - Nicholas McGranahan
- Cancer Research UK Lung Cancer Centre of Excellence, University College London Cancer Institute, London, UK
- Cancer Genome Evolution Research Group, Cancer Research UK Lung Cancer Centre of Excellence, University College London Cancer Institute, London, UK
| | - Mariam Jamal-Hanjani
- Cancer Research UK Lung Cancer Centre of Excellence, University College London Cancer Institute, London, UK
- Cancer Metastasis Laboratory, University College London Cancer Institute, London, UK
- Department of Oncology, University College London Hospitals, London, UK
| | - Alexander M Frankell
- Cancer Research UK Lung Cancer Centre of Excellence, University College London Cancer Institute, London, UK
- Cancer Evolution and Genome Instability Laboratory., The Francis Crick Institute, London, UK
| | | | - Charles Swanton
- Cancer Research UK Lung Cancer Centre of Excellence, University College London Cancer Institute, London, UK.
- Cancer Evolution and Genome Instability Laboratory., The Francis Crick Institute, London, UK.
- Department of Oncology, University College London Hospitals, London, UK.
| |
Collapse
|
24
|
Lucas O, Ward S, Zaidi R, Bunkum A, Frankell AM, Moore DA, Hill MS, Liu WK, Marinelli D, Lim EL, Hessey S, Naceur-Lombardelli C, Rowan A, Purewal-Mann SK, Zhai H, Dietzen M, Ding B, Royle G, Aparicio S, McGranahan N, Jamal-Hanjani M, Kanu N, Swanton C, Zaccaria S. Characterizing the evolutionary dynamics of cancer proliferation in single-cell clones with SPRINTER. Nat Genet 2025; 57:103-114. [PMID: 39614124 PMCID: PMC11735394 DOI: 10.1038/s41588-024-01989-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2023] [Accepted: 10/15/2024] [Indexed: 12/01/2024]
Abstract
Proliferation is a key hallmark of cancer, but whether it differs between evolutionarily distinct clones co-existing within a tumor is unknown. We introduce the Single-cell Proliferation Rate Inference in Non-homogeneous Tumors through Evolutionary Routes (SPRINTER) algorithm that uses single-cell whole-genome DNA sequencing data to enable accurate identification and clone assignment of S- and G2-phase cells, as assessed by generating accurate ground truth data. Applied to a newly generated longitudinal, primary-metastasis-matched dataset of 14,994 non-small cell lung cancer cells, SPRINTER revealed widespread clone proliferation heterogeneity, orthogonally supported by Ki-67 staining, nuclei imaging and clinical imaging. We further demonstrated that high-proliferation clones have increased metastatic seeding potential, increased circulating tumor DNA shedding and clone-specific altered replication timing in proliferation- or metastasis-related genes associated with expression changes. Applied to previously generated datasets of 61,914 breast and ovarian cancer cells, SPRINTER revealed increased single-cell rates of different genomic variants and enrichment of proliferation-related gene amplifications in high-proliferation clones.
Collapse
Affiliation(s)
- Olivia Lucas
- Computational Cancer Genomics Research Group, University College London Cancer Institute, London, UK
- Cancer Research UK Lung Cancer Centre of Excellence, University College London Cancer Institute, London, UK
- Cancer Evolution and Genome Instability Laboratory, The Francis Crick Institute, London, UK
- University College London Hospitals, London, UK
| | - Sophia Ward
- Cancer Research UK Lung Cancer Centre of Excellence, University College London Cancer Institute, London, UK
- Cancer Evolution and Genome Instability Laboratory, The Francis Crick Institute, London, UK
- Genomics Science Technology Platform, The Francis Crick Institute, London, UK
| | - Rija Zaidi
- Computational Cancer Genomics Research Group, University College London Cancer Institute, London, UK
- Cancer Research UK Lung Cancer Centre of Excellence, University College London Cancer Institute, London, UK
| | - Abigail Bunkum
- Computational Cancer Genomics Research Group, University College London Cancer Institute, London, UK
- Cancer Research UK Lung Cancer Centre of Excellence, University College London Cancer Institute, London, UK
- Cancer Metastasis Laboratory, University College London Cancer Institute, London, UK
| | - Alexander M Frankell
- Cancer Research UK Lung Cancer Centre of Excellence, University College London Cancer Institute, London, UK
- Cancer Evolution and Genome Instability Laboratory, The Francis Crick Institute, London, UK
| | - David A Moore
- Cancer Research UK Lung Cancer Centre of Excellence, University College London Cancer Institute, London, UK
- Cancer Evolution and Genome Instability Laboratory, The Francis Crick Institute, London, UK
- Department of Cellular Pathology, University College London Hospitals, London, UK
| | - Mark S Hill
- Cancer Evolution and Genome Instability Laboratory, The Francis Crick Institute, London, UK
| | - Wing Kin Liu
- Cancer Research UK Lung Cancer Centre of Excellence, University College London Cancer Institute, London, UK
- Cancer Metastasis Laboratory, University College London Cancer Institute, London, UK
| | - Daniele Marinelli
- Cancer Metastasis Laboratory, University College London Cancer Institute, London, UK
- Cancer Genome Evolution Research Group, University College London Cancer Institute, London, UK
- Department of Experimental Medicine, Sapienza University of Rome, Rome, Italy
| | - Emilia L Lim
- Cancer Research UK Lung Cancer Centre of Excellence, University College London Cancer Institute, London, UK
- Cancer Evolution and Genome Instability Laboratory, The Francis Crick Institute, London, UK
| | - Sonya Hessey
- Computational Cancer Genomics Research Group, University College London Cancer Institute, London, UK
- Cancer Research UK Lung Cancer Centre of Excellence, University College London Cancer Institute, London, UK
- University College London Hospitals, London, UK
- Cancer Metastasis Laboratory, University College London Cancer Institute, London, UK
| | | | - Andrew Rowan
- Cancer Evolution and Genome Instability Laboratory, The Francis Crick Institute, London, UK
| | | | - Haoran Zhai
- Cancer Research UK Lung Cancer Centre of Excellence, University College London Cancer Institute, London, UK
- Cancer Evolution and Genome Instability Laboratory, The Francis Crick Institute, London, UK
| | - Michelle Dietzen
- Cancer Research UK Lung Cancer Centre of Excellence, University College London Cancer Institute, London, UK
- Cancer Evolution and Genome Instability Laboratory, The Francis Crick Institute, London, UK
- Cancer Genome Evolution Research Group, University College London Cancer Institute, London, UK
| | - Boyue Ding
- Department of Medical Physics and Biomedical Engineering, University College London, London, UK
| | - Gary Royle
- Department of Medical Physics and Biomedical Engineering, University College London, London, UK
| | - Samuel Aparicio
- Department of Molecular Oncology, British Columbia Cancer Research Centre, Vancouver, British Columbia, Canada
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, British Columbia, Canada
| | - Nicholas McGranahan
- Cancer Research UK Lung Cancer Centre of Excellence, University College London Cancer Institute, London, UK
- Cancer Genome Evolution Research Group, University College London Cancer Institute, London, UK
| | - Mariam Jamal-Hanjani
- Cancer Research UK Lung Cancer Centre of Excellence, University College London Cancer Institute, London, UK
- University College London Hospitals, London, UK
- Cancer Metastasis Laboratory, University College London Cancer Institute, London, UK
| | - Nnennaya Kanu
- Cancer Research UK Lung Cancer Centre of Excellence, University College London Cancer Institute, London, UK.
| | - Charles Swanton
- Cancer Research UK Lung Cancer Centre of Excellence, University College London Cancer Institute, London, UK.
- Cancer Evolution and Genome Instability Laboratory, The Francis Crick Institute, London, UK.
- University College London Hospitals, London, UK.
| | - Simone Zaccaria
- Computational Cancer Genomics Research Group, University College London Cancer Institute, London, UK.
- Cancer Research UK Lung Cancer Centre of Excellence, University College London Cancer Institute, London, UK.
| |
Collapse
|
25
|
Cheng L, Xu S, Wang Y, Li S, Li B, Li X. Circulating Tumor DNA Detection for Recurrence Monitoring of Stage I Non-Small Cell Lung Cancer Treated With Microwave Ablation. Thorac Cancer 2025; 16:e15534. [PMID: 39825733 PMCID: PMC11742128 DOI: 10.1111/1759-7714.15534] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2024] [Revised: 12/29/2024] [Accepted: 01/05/2025] [Indexed: 01/20/2025] Open
Abstract
PURPOSE As microwave ablation continues to be used in patients with inoperable stage I non-small cell lung cancer (NSCLC), it is particularly important to monitor efficacy. Whether plasma ctDNA detection can predict its efficacy should be illustrated. METHODS We recruited 43 patients with inoperative stage I NSCLC, all of whom underwent biopsy-synchronous microwave ablation (MWA). Peripheral blood samples were collected at baseline (n = 43), within 1 h post-MWA (n = 28), and at the landmark time point (n = 26) for MRD detection. Clinical outcomes were analyzed using Kaplan-Meier survival analysis. RESULTS Patients with undetectable ctDNA at baseline (p = 0.042) and within 1 h after MWA (p = 0.023) had better clinical outcomes. In particular, patients with undetectable ctDNA at the 1-h post-MWA time point did not experience recurrence. Detection of ctDNA at the landmark time point is considered an independent risk factor for prognosis and is strongly correlated with clinical outcomes (p = 0.001), the median time to recurrence indicated by ctDNA was 4.9 months earlier compared to imaging. The clinical outcomes of patients with ctDNA clearance were similar to those with no ctDNA (p = 0.570). Risk stratification indicated that patients with persistent ctDNA had worse clinical outcomes compared to those who never had detectable ctDNA (p = 0.004). CONCLUSION Our findings suggest that ctDNA monitoring can assist in predicting clinical outcomes in stage I NSCLC treated with microwave ablation. Patients with undetectable ctDNA within 1 h after MWA are determined to be clinically cured. Risk stratification based on ctDNA test results helps to differentiate high-risk patients.
Collapse
Affiliation(s)
- Lin Cheng
- Department of Minimally Invasive Tumor Therapies CenterBeijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine, Chinese Academy of Medical SciencesBeijingChina
- Medical SchoolUniversity of Chinese Academy of SciencesBeijingChina
| | - Sheng Xu
- Department of Minimally Invasive Tumor Therapies CenterBeijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine, Chinese Academy of Medical SciencesBeijingChina
| | - Yu‐feng Wang
- Department of Minimally Invasive Tumor Therapies CenterBeijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine, Chinese Academy of Medical SciencesBeijingChina
| | - Sheng‐wei Li
- Department of Minimally Invasive Tumor Therapies CenterBeijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine, Chinese Academy of Medical SciencesBeijingChina
| | - Bin Li
- Department of Minimally Invasive Tumor Therapies CenterBeijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine, Chinese Academy of Medical SciencesBeijingChina
| | - Xiao‐Guang Li
- Department of Minimally Invasive Tumor Therapies CenterBeijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine, Chinese Academy of Medical SciencesBeijingChina
- Medical SchoolUniversity of Chinese Academy of SciencesBeijingChina
| |
Collapse
|
26
|
Lee G, Moon SH, Kim JH, Jeong DY, Choi J, Choi JY, Lee HY. Multimodal Imaging Approach for Tumor Treatment Response Evaluation in the Era of Immunotherapy. Invest Radiol 2025; 60:11-26. [PMID: 39018248 DOI: 10.1097/rli.0000000000001096] [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: 07/19/2024]
Abstract
ABSTRACT Immunotherapy is likely the most remarkable advancement in lung cancer treatment during the past decade. Although immunotherapy provides substantial benefits, their therapeutic responses differ from those of conventional chemotherapy and targeted therapy, and some patients present unique immunotherapy response patterns that cannot be judged under the current measurement standards. Therefore, the response monitoring of immunotherapy can be challenging, such as the differentiation between real response and pseudo-response. This review outlines the various tumor response patterns to immunotherapy and discusses methods for quantifying computed tomography (CT) and 18 F-fluorodeoxyglucose positron emission tomography (PET) in the field of lung cancer. Emerging technologies in magnetic resonance imaging (MRI) and non-FDG PET tracers are also explored. With immunotherapy responses, the role for imaging is essential in both anatomical radiological responses (CT/MRI) and molecular changes (PET imaging). Multiple aspects must be considered when assessing treatment responses using CT and PET. Finally, we introduce multimodal approaches that integrate imaging and nonimaging data, and we discuss future directions for the assessment and prediction of lung cancer responses to immunotherapy.
Collapse
Affiliation(s)
- Geewon Lee
- From the Department of Radiology and Center for Imaging Science, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, South Korea (G.L., D.Y.J., J.C., H.Y.L.); Department of Radiology and Medical Research Institute, Pusan National University Hospital, Pusan National University School of Medicine, Busan, South Korea (G.L.); Department of Nuclear Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, South Korea (S.H.M., J.Y.C.); Industrial Biomaterial Research Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon, South Korea (J.H.K.); Department of Digital Health, SAIHST, Sungkyunkwan University, Seoul, South Korea (J.C.); and Department of Health Sciences and Technology, SAIHST, Sungkyunkwan University, Seoul, South Korea (H.Y.L.)
| | | | | | | | | | | | | |
Collapse
|
27
|
Wang J, Zhou W, Xu Y, Duan J, Zhou Q, Wang G, Li L, Xu C, Wang W, Cai S, Wang Z, Wang J. Antithetical impacts of deleterious LRP1B mutations in non-squamous and squamous NSCLCs on predicting benefits from immune checkpoint inhibitor alone or with chemotherapy over chemotherapy alone: retrospective analyses of the POPLAR/OAK and CHOICE-01 trials. SCIENCE CHINA. LIFE SCIENCES 2025; 68:249-262. [PMID: 39276256 DOI: 10.1007/s11427-023-2554-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/02/2024] [Accepted: 01/18/2024] [Indexed: 09/16/2024]
Abstract
In non-small cell lung cancers, the non-squamous and squamous subtypes (nsqNSCLC and sqNSCLC) exhibit disparities in pathophysiology, tumor immunology, and potential genomic correlates affecting responses to immune checkpoint inhibitor (ICI)-based treatments. In our in-house training cohort (n=85), the presence of the LRP1B deleterious mutation (LRP1B-del) was associated with longer and shorter progression-free survival (PFS) on ICIs alone in nsqNSCLCs and sqNSCLCs, respectively (Pinteraction=0.008). These results were validated using a larger public ICI cohort (n=208, Pinteraction<0.001). Multiplex immunofluorescence staining revealed an association between LRP1B-del and increased and decreased numbers of tumor-infiltrating CD8+ T cells in nsqNSCLCs (P=0.040) and sqNSCLCs (P=0.014), respectively. In the POPLAR/OAK cohort, nsqNSCLCs with LRP1B-del demonstrated improved PFS benefits from atezolizumab over docetaxel (hazard ratio (HR) =0.70, P=0.046), whereas this benefit was negligible in those without LRP1B-del (HR=1.05, P=0.64). Conversely, sqNSCLCs without LRP1B-del benefited more from atezolizumab (HR=0.60, P=0.002) than those with LRP1B-del (HR=1.30, P=0.31). Consistent results were observed in the in-house CHOICE-01 cohort, in which nsqNSCLCs with LRP1B-del and sqNSCLCs without LRP1B-del benefited more from toripalimab plus chemotherapy than from chemotherapy alone (Pinteraction=0.008). This multi-cohort study delineates the antithetical impacts of LRP1B-del in nsqNSCLCs and sqNSCLCs on predicting the benefits from ICI alone or with chemotherapy over chemotherapy alone. Our findings highlight the distinct clinical utility of LRP1B-del in guiding treatment choices for nsqNSCLCs and sqNSCLCs, emphasizing the necessity for a detailed analysis based on pathological subtypes when investigating biomarkers for cancer therapeutics.
Collapse
Affiliation(s)
- Jinliang Wang
- Department of Oncology, The Fifth Medical Center of Chinese PLA General Hospital, Beijing, 100091, China
| | - Wenyong Zhou
- Department of Thoracic Surgery, Shanghai Chest Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200025, China
| | - Yu Xu
- Burning Rock Biotech, Guangzhou, 510300, China
| | - Jianchun Duan
- CAMS Key Laboratory of Translational Research on Lung Cancer, State Key Laboratory of Molecular Oncology, Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | | | | | - Leo Li
- Burning Rock Biotech, Guangzhou, 510300, China
| | - Chunwei Xu
- Institute of Basic Medicine and Cancer (IBMC), Chinese Academy of Sciences, Hangzhou, 310000, China
| | - Wenxian Wang
- Department of Clinical Trial, The Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Hangzhou, 310022, China
| | - Shangli Cai
- Burning Rock Biotech, Guangzhou, 510300, China.
| | - Zhijie Wang
- CAMS Key Laboratory of Translational Research on Lung Cancer, State Key Laboratory of Molecular Oncology, Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China.
| | - Jie Wang
- CAMS Key Laboratory of Translational Research on Lung Cancer, State Key Laboratory of Molecular Oncology, Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China.
| |
Collapse
|
28
|
Dong S, Yan B, Liu SY, Gao X, Hong HZ, Li HJ, Gao W, Yan HH, Maggie Liu SY, Tu HY, Pan Y, Zhou Q, Yang XN, Xia XF, Yi X, Zhong WZ, Wu YL, Zhang JT. Monitoring of Circulating Tumor DNA and Indication of De-Escalation Adjuvant Targeted Therapy for EGFR-Mutated NSCLC After Complete Resection. JTO Clin Res Rep 2025; 6:100758. [PMID: 39758595 PMCID: PMC11699309 DOI: 10.1016/j.jtocrr.2024.100758] [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: 08/11/2024] [Revised: 10/14/2024] [Accepted: 10/26/2024] [Indexed: 01/07/2025] Open
Abstract
Introduction EGFR tyrosine kinase inhibitor (TKI) is the standard adjuvant treatment for patients with stages IB to IIIA EGFR-mutated NSCLC. Nevertheless, adapting this approach to include a molecular residual disease (MRD)-guided de-escalation strategy warrants further investigation. Methods From January 2019 to December 2022, 71 patients with stages I to III NSCLC and EGFR (exon 19 deletion or L858R) mutations were enrolled in this observational study. A total of 375 blood samples were analyzed using the MRD_Navigator assay. Among them, 27 patients suspended EGFR TKI treatment based on undetectable MRD and were thus included in the adaptive, de-escalation group. Results Overall, the sensitivity of longitudinal MRD was 86.2%. Only four patients (11.8%) recurred with longitudinal undetectable MRD, indicating a negative predictive value of 88.2%. Of the patients who had detectable MRD after surgery, nine subsequently received EGFR TKI treatment, with only one (11.1%) achieving persistent circulating tumor DNA clearance post-EGFR TKI. Furthermore, 22 patients with stages IB to III disease who had previously suspended their TKI treatment based on undetectable MRD were included in the adaptive group, with an average duration of TKI 3.9 (range: 0-35.0) months. The 2-year disease-free survival rate of these 22 patients was 80.2%, and the median was not reached. Five patients (n = 5 of 22, 22.7%) had disease recurrence during the period of drug cessation but were stable under EGFR TKI treatment until the latest follow-up. Two patients remained in complete remission. Conclusions Our initial findings underscore the potential of an adaptive, de-escalation approach to adjuvant EGFR TKIs based on circulating tumor DNA-MRD monitoring.
Collapse
Affiliation(s)
- Song Dong
- Guangdong Lung Cancer Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, Guangdong, People's Republic of China
| | - Bingfa Yan
- Geneplus-Beijing Institute, Beijing, People's Republic of China
- State Key Laboratory of Microbial Resources, Institute of Microbiology, Chinese Academy of Sciences, Beijing, People's Republic of China
| | - Si-Yang Liu
- Guangdong Lung Cancer Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, Guangdong, People's Republic of China
| | - Xuan Gao
- Geneplus-Beijing Institute, Beijing, People's Republic of China
| | - Hui-Zhao Hong
- Guangdong Lung Cancer Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, Guangdong, People's Republic of China
| | - Hong-Ji Li
- Guangdong Lung Cancer Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, Guangdong, People's Republic of China
| | - Wei Gao
- Geneplus-Beijing Institute, Beijing, People's Republic of China
| | - Hong-Hong Yan
- Guangdong Lung Cancer Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, Guangdong, People's Republic of China
| | - Si-Yang Maggie Liu
- Chinese Thoracic Oncology Group (CTONG), Guangzhou, Guangdong, People's Republic of China
| | - Hai-Yan Tu
- Guangdong Lung Cancer Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, Guangdong, People's Republic of China
| | - Yi Pan
- Guangdong Lung Cancer Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, Guangdong, People's Republic of China
| | - Qing Zhou
- Guangdong Lung Cancer Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, Guangdong, People's Republic of China
| | - Xue-Ning Yang
- Guangdong Lung Cancer Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, Guangdong, People's Republic of China
| | - Xue-Feng Xia
- Geneplus-Beijing Institute, Beijing, People's Republic of China
| | - Xin Yi
- Geneplus-Beijing Institute, Beijing, People's Republic of China
| | - Wen-Zhao Zhong
- Guangdong Lung Cancer Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, Guangdong, People's Republic of China
| | - Yi-Long Wu
- Guangdong Lung Cancer Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, Guangdong, People's Republic of China
| | - Jia-Tao Zhang
- Guangdong Lung Cancer Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, Guangdong, People's Republic of China
| |
Collapse
|
29
|
Lu B. Cancer phylogenetic inference using copy number alterations detected from DNA sequencing data. CANCER PATHOGENESIS AND THERAPY 2025; 3:16-29. [PMID: 39872371 PMCID: PMC11764021 DOI: 10.1016/j.cpt.2024.04.003] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 01/31/2024] [Revised: 04/05/2024] [Accepted: 04/15/2024] [Indexed: 01/30/2025]
Abstract
Cancer is an evolutionary process involving the accumulation of diverse somatic mutations and clonal evolution over time. Phylogenetic inference from samples obtained from an individual patient offers a powerful approach to unraveling the intricate evolutionary history of cancer and provides insights that can inform cancer treatment. Somatic copy number alterations (CNAs) are important in cancer evolution and are often used as markers, alone or with other somatic mutations, for phylogenetic inferences, particularly in low-coverage DNA sequencing data. Many phylogenetic inference methods using CNAs detected from bulk or single-cell DNA sequencing data have been developed over the years. However, there have been no systematic reviews on these methods. To summarize the state-of-the-art of the field and inform future development, this review presents a comprehensive survey on the major challenges in inference, different types of methods, and applications of these methods. The challenges are discussed from the aspects of input data, models of evolution, and inference algorithms. The different methods are grouped according to the markers used for inference and the types of the reconstructed trees. The applications include using phylogenetic inference to understand intra-tumor heterogeneity, metastasis, treatment resistance, and early cancer development. This review also sheds light on future directions of cancer phylogenetic inference using CNAs, including the improvement of scalability, the utilization of new types of data, and the development of more realistic models of evolution.
Collapse
Affiliation(s)
- Bingxin Lu
- School of Biosciences and Medicine, University of Surrey, Guildford GU2 7XH, UK
- Surrey Institute for People-Centred Artificial Intelligence, University of Surrey, Guildford GU2 7XH, UK
| |
Collapse
|
30
|
Nguyen THH, Vu GH, Nguyen TT, Nguyen TA, Tran VU, Vu LT, Nguyen GTH, Nguyen ND, Tran TH, Nguyen VTC, Nguyen TD, Nguyen TH, Vo DH, Van TTV, Do TT, Le MP, Huynh LAK, Nguyen DS, Tang HS, Nguyen H, Phan M, Giang H, Tu LN, Tran LS. Combination of Hotspot Mutations With Methylation and Fragmentomic Profiles to Enhance Multi-Cancer Early Detection. Cancer Med 2025; 14:e70575. [PMID: 39748775 PMCID: PMC11695824 DOI: 10.1002/cam4.70575] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2024] [Revised: 11/15/2024] [Accepted: 12/23/2024] [Indexed: 01/04/2025] Open
Abstract
BACKGROUND Multi-cancer early detection (MCED) through a single blood test significantly advances cancer diagnosis. However, most MCED tests rely on a single type of biomarkers, leading to limited sensitivity, particularly for early-stage cancers. We previously developed SPOT-MAS, a multimodal ctDNA-based assay analyzing methylation and fragmentomic profiles to detect five common cancers. Despite its potential, SPOT-MAS exhibited moderate sensitivities for early-stage cancers. This study investigated whether integrating hotspot mutations into SPOT-MAS could enhance its detection rates. METHOD A targeted amplicon sequencing approach was developed to profile 700 hotspot mutations in cell-free DNA and integrated into the SPOT-MAS assay, creating a single-blood draw workflow. This workflow, namely SPOT-MAS Plus was retrospectively validated in a cohort of 255 non-metastatic cancer patients (breast, colorectal, gastric, liver, and lung) and 304 healthy individuals. RESULTS Hotspot mutations were detected in 131 of 255 (51.4%) cancer patients, with the highest rates in liver cancer (96.5%), followed by colorectal (59.3%) and lung cancer (53.7%). Lower detection rates were found for cancers with low tumor mutational burden, such as breast (31.3%) and gastric (41.9%) cancers. In contrast, SPOT-MAS demonstrated higher sensitivities for these cancers (51.6% for breast and 62.9% for gastric). The combination of hotspot mutations with SPOT-MAS predictions improved early-stage cancer detection, achieving an overall sensitivity of 78.5% at a specificity of 97.7%. Enhanced sensitivities were observed for colorectal (81.36%) and lung cancer (82.9%). CONCLUSION The integration of genetic and epigenetic alterations into a multimodal assay significantly enhances the early detection of various cancers. Further validation in larger cohorts is necessary to support broader clinical applications.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | | | | | - Dac Ho Vo
- Medical Genetics InstituteHo Chi MinhVietnam
| | | | | | | | | | | | | | | | | | - Hoa Giang
- Medical Genetics InstituteHo Chi MinhVietnam
| | - Lan N. Tu
- Medical Genetics InstituteHo Chi MinhVietnam
| | - Le Son Tran
- Medical Genetics InstituteHo Chi MinhVietnam
| |
Collapse
|
31
|
Noji R, Tohyama K, Nakamura S, Naito T, Oikawa Y, Kuroshima T, Tomioka H, Michi Y, Ikeda S, Asakage T, Miura M, Hamamoto Y, Harada H, Kano Y. Dynamic Changes in Circulating Tumor DNA During Immunotherapy for Head and Neck Cancer: SHIZUKU-HN Study. Int J Mol Sci 2024; 26:235. [PMID: 39796090 PMCID: PMC11719933 DOI: 10.3390/ijms26010235] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2024] [Revised: 12/25/2024] [Accepted: 12/27/2024] [Indexed: 01/13/2025] Open
Abstract
Immune checkpoint inhibitors (ICIs) are effective in treating recurrent/metastatic head and neck squamous cell carcinoma (HNSCC), but only 20% of patients achieve durable responses. This study evaluated circulating tumor DNA (ctDNA) as a real-time biomarker for monitoring treatment response in HNSCC. The SHIZUKU-HN study prospectively collected and analyzed serial plasma samples (n = 27) from HNSCC patients undergoing ICIs, using Guardant360 to assess ctDNA variant allele frequency (VAF) and genetic mutations. Tumor volumes were quantified using 3D reconstruction of CT scans, and data from Japan's C-CAT database (n = 2255) provided insights into ctDNA testing in HNSCC. C-CAT data showed that ctDNA testing was underutilized, performed in only 7% of head and neck cancer cases. In SHIZUKU-HN, mean VAF significantly correlated with tumor volume (Spearman's ρ = 0.70, p = 0.001), often preceding radiographic progression. BRAF and APC mutations disappeared in partial responders, while GNAS mutations varied. EGFR and PIK3CA amplifications, detectable via ctDNA but missed in tissue biopsies, indicated emerging resistance mechanisms. The SHIZUKU-HN study demonstrates the potential of ctDNA as a dynamic biomarker in HNSCC, offering early insights into treatment efficacy and informing personalized ICI therapy.
Collapse
Affiliation(s)
- Rika Noji
- Department of Oral and Maxillofacial Surgical Oncology, Division of Health Science, Graduate School of Medical and Dental Sciences, Institute of Science Tokyo, 1-5-45 Yushima, Bunkyo-Ku, Tokyo 113-8510, Japan; (R.N.)
- Department of Medical Oncology, Institute of Science Tokyo, 1-5-45 Yushima, Bunkyo-Ku, Tokyo 113-8510, Japan
| | - Kohki Tohyama
- Department of Dental Radiology and Radiation Oncology, Graduate School of Medical and Dental Sciences, Institute of Science Tokyo, 1-5-45 Yushima, Bunkyo-Ku, Tokyo 113-8510, Japan
| | - Shin Nakamura
- Department of Dental Radiology and Radiation Oncology, Graduate School of Medical and Dental Sciences, Institute of Science Tokyo, 1-5-45 Yushima, Bunkyo-Ku, Tokyo 113-8510, Japan
| | - Takahiro Naito
- Department of Oral and Maxillofacial Surgical Oncology, Division of Health Science, Graduate School of Medical and Dental Sciences, Institute of Science Tokyo, 1-5-45 Yushima, Bunkyo-Ku, Tokyo 113-8510, Japan; (R.N.)
- Department of Medical Oncology, Institute of Science Tokyo, 1-5-45 Yushima, Bunkyo-Ku, Tokyo 113-8510, Japan
| | - Yu Oikawa
- Department of Oral and Maxillofacial Surgical Oncology, Division of Health Science, Graduate School of Medical and Dental Sciences, Institute of Science Tokyo, 1-5-45 Yushima, Bunkyo-Ku, Tokyo 113-8510, Japan; (R.N.)
| | - Takeshi Kuroshima
- Department of Oral and Maxillofacial Surgical Oncology, Division of Health Science, Graduate School of Medical and Dental Sciences, Institute of Science Tokyo, 1-5-45 Yushima, Bunkyo-Ku, Tokyo 113-8510, Japan; (R.N.)
| | - Hirofumi Tomioka
- Department of Oral and Maxillofacial Surgical Oncology, Division of Health Science, Graduate School of Medical and Dental Sciences, Institute of Science Tokyo, 1-5-45 Yushima, Bunkyo-Ku, Tokyo 113-8510, Japan; (R.N.)
| | - Yasuyuki Michi
- Department of Oral and Maxillofacial Surgical Oncology, Division of Health Science, Graduate School of Medical and Dental Sciences, Institute of Science Tokyo, 1-5-45 Yushima, Bunkyo-Ku, Tokyo 113-8510, Japan; (R.N.)
| | - Sadakatsu Ikeda
- Department of Precision Cancer Medicine, Center for Innovative Cancer Treatment, Institute of Science Tokyo, 1-5-45 Yushima, Bunkyo-Ku, Tokyo 113-8510, Japan
| | - Takahiro Asakage
- Department of Head and Neck Surgery, Institute of Science Tokyo 1-5-45 Yushima, Bunkyo-Ku, Tokyo 113-8510, Japan
| | - Masahiko Miura
- Department of Dental Radiology and Radiation Oncology, Graduate School of Medical and Dental Sciences, Institute of Science Tokyo, 1-5-45 Yushima, Bunkyo-Ku, Tokyo 113-8510, Japan
| | - Yasuo Hamamoto
- Department of Medical Oncology, Institute of Science Tokyo, 1-5-45 Yushima, Bunkyo-Ku, Tokyo 113-8510, Japan
| | - Hiroyuki Harada
- Department of Oral and Maxillofacial Surgical Oncology, Division of Health Science, Graduate School of Medical and Dental Sciences, Institute of Science Tokyo, 1-5-45 Yushima, Bunkyo-Ku, Tokyo 113-8510, Japan; (R.N.)
| | - Yoshihito Kano
- Department of Medical Oncology, Institute of Science Tokyo, 1-5-45 Yushima, Bunkyo-Ku, Tokyo 113-8510, Japan
| |
Collapse
|
32
|
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 2024:S0889-8588(24)00147-3. [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] [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
|
33
|
Swalduz A, Schiffler C, Curcio H, Ambasager B, Le Moel G, Debieuvre D, Dot JM, Duruisseaux M, Fournel P, Odier L, Demolombe S, Bizieux-Thaminy A, Peytier A, Schott R, Hominal S, Tissot C, Bombaron P, Metzger S, Donnat M, Ortiz-Cuaran S, Rosenfeld N, Pipinikas C, Saintigny P, Pérol M. LIBELULE: A Randomized Phase III Study to Evaluate the Clinical Relevance of Early Liquid Biopsy in Patients With Suspicious Metastatic Lung Cancer. J Thorac Oncol 2024:S1556-0864(24)02530-9. [PMID: 39694415 DOI: 10.1016/j.jtho.2024.12.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2024] [Revised: 10/12/2024] [Accepted: 12/08/2024] [Indexed: 12/20/2024]
Abstract
OBJECTIVES Genomic profiling is a major component for first-line treatment decisions in patients with NSCLC and the timeliness of biomarker testing is essential to improve time to treatment initiation (TTI) or avoid inappropriate treatment. METHODS The phase III LIquid Biopsy for the Early detection of LUng cancer Lesion trial (NCT03721120) included patients with radiological suspicion of advanced lung cancer. They were randomized (1:1), the control arm receiving diagnostic procedures according to each center's practice, and the liquid biopsy arm with additional testing performed at the first visit using the InVisionFirst-Lung assay. Treatment initiation and type were defined according to the European Society for Medical Oncology guidelines. Primary endpoint was the time from randomization to initiation of appropriate treatment on the basis of informative genomic and pathological results in the intention-to-treat population. RESULTS A total of 319 patients were enrolled (liquid biopsy [LB]: 161; control: 158). The median age was 68 years, 28.8% were non-smokers, 18.1% had a performance status of 2 or higher, and 56.7% had adenocarcinoma. In the LB arm, 81% of patients had circulating tumor DNA findings. The mean TTI was not significantly reduced (LB: 29.0 d; control 34 d (p = 0.26)). Sensitivity analyses found a shorter TTI in patients from the LB arm who received systemic treatment (LB: 29.1 d; control: 38.8 d, p = 0.01), in patients with advanced non-squamous NSCLC (LB: 29.5 d; control: 40.3 d, p = 0.01), and in patients with first-line targetable alterations (LB: 21d; control 37.4 d) (p = 0.004). Time to contributory genomic results was significantly reduced (LB: 17.9 d; control: 25.6 d, p < 0.001). CONCLUSION Early liquid biopsy testing did not significantly shorten the TTI in unselected patients referred for suspected advanced lung cancer. Nevertheless, it could reduce the TTI in patients eligible for systemic treatment, particularly for those with actionable alterations.
Collapse
Affiliation(s)
- Aurélie Swalduz
- Department of Medical Oncology, Centre Léon Bérard, Lyon, France; Univ Lyon, Claude Bernard Lyon 1 University, INSERM 1052, CNRS 5286, Centre Léon Bérard, Cancer Research Center of Lyon, Lyon, France.
| | - Camille Schiffler
- Department of Clinical Research and Innovation, Centre Léon Bérard, Lyon, France
| | - Hubert Curcio
- Department of Medical Oncology, Centre François Baclesse, Caen, France
| | - Bana Ambasager
- NeoGenomics, Babraham Research Campus, Cambridge, United Kingdom
| | - Gabriel Le Moel
- Department of Pneumology, Centre Hospitalier du Cotentin Louis Pasteur, Cherbourg, France
| | - Didier Debieuvre
- Department of Pneumology, Groupe Hospitalier de la Région Mulhouse Sud-Alsace, Hôpital Emile Muller, GHRMSA - Mulhouse, Mulhouse, France
| | - Jean-Marc Dot
- Department of Pneumology, Medipole, Lyon Villeurbanne, France
| | - Michael Duruisseaux
- Respiratory Department, Louis Pradel Hospital, Hospices Civils de Lyon Cancer Institute, Lyon, France
| | - Pierre Fournel
- Department of Pneumology and Thoracic Oncology, Hôpital Nord, Saint-Etienne, France
| | - Luc Odier
- Department of Pneumology, l'Hôpital Nord-Ouest Villefranche sur Saône, Villefranche-sur-Saône, France
| | - Sylvie Demolombe
- Department of Medical Oncology, Infirmerie Protestante, Caluire et Cuire, France
| | | | - Annie Peytier
- Department of Medical Oncology, Centre Hospitalier de Bayeux, Bayeux, France
| | - Roland Schott
- Department of Medical Oncology, Institut de Cancérologie Strasbourg Europe (ICANS), Strasbourg, France
| | - Stéphane Hominal
- Department of Pneumology, Centre Hospitalier Annecy-Genevois, Epagny-Metz Tessy, France
| | - Claire Tissot
- Department of Oncology, Hôpital Privé de la Loire, Saint-Etienne, France
| | - Pierre Bombaron
- Department of Medicine, Hôpital Privé Jean Mermoz, Lyon, France
| | - Séverine Metzger
- Department of Clinical Research and Innovation, Centre Léon Bérard, Lyon, France
| | - Mathilde Donnat
- Department of Clinical Research and Innovation, Centre Léon Bérard, Lyon, France
| | - Sandra Ortiz-Cuaran
- Univ Lyon, Claude Bernard Lyon 1 University, INSERM 1052, CNRS 5286, Centre Léon Bérard, Cancer Research Center of Lyon, Lyon, France
| | - Nitzan Rosenfeld
- Cancer Research UK Cambridge Institute, University of Cambridge, Cambridge, United Kingdom; Cancer Research UK Cambridge Centre, Cambridge, United Kingdom; Barts Cancer Institute, Queen Mary University of London, London, United Kingdom
| | | | - Pierre Saintigny
- Department of Medical Oncology, Centre Léon Bérard, Lyon, France; Univ Lyon, Claude Bernard Lyon 1 University, INSERM 1052, CNRS 5286, Centre Léon Bérard, Cancer Research Center of Lyon, Lyon, France
| | - Maurice Pérol
- Department of Medical Oncology, Centre Léon Bérard, Lyon, France
| |
Collapse
|
34
|
Kindt CK, Alves CL, Ehmsen S, Kragh A, Reinert T, Vogsen M, Kodahl AR, Rønlev JD, Ardik D, Sørensen AL, Evald K, Clemmensen ML, Staaf J, Ditzel HJ. Genomic alterations associated with resistance and circulating tumor DNA dynamics for early detection of progression on CDK4/6 inhibitor in advanced breast cancer. Int J Cancer 2024; 155:2211-2222. [PMID: 39128978 DOI: 10.1002/ijc.35126] [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: 01/22/2024] [Revised: 07/11/2024] [Accepted: 07/12/2024] [Indexed: 08/13/2024]
Abstract
Combined CDK4/6 inhibitor (CDK4/6i) and endocrine therapy significantly improves outcome for patients with estrogen receptor-positive (ER+) metastatic breast cancer, but drug resistance and thus disease progression inevitably occur. Herein, we aimed to identify genomic alterations associated with combined CDK4/6i and endocrine therapy resistance, and follow the levels of specific mutations in longitudinal circulating tumor DNA (ctDNA) for early detection of progression. From a cohort of 86 patients with ER+ metastatic breast cancer we performed whole exome sequencing or targeted sequencing of paired tumor (N = 8) or blood samples (N = 5) obtained before initiation of combined CDK4/6i and endocrine therapy and at disease progression. Mutations in oncogenic genes at progression were rare, while amplifications of growth-regulating genes were more frequent. The most frequently acquired alterations observed were PIK3CA and TP53 mutations and PDK1 amplification. Longitudinal ctDNA dynamics of mutant PIK3CA or private mutations revealed increased mutation levels at progression in 8 of 10 patients (80%). Impressively, rising levels of PIK3CA-mutated ctDNA were detected 4-17 months before imaging. Our data add to the growing evidence supporting longitudinal ctDNA analysis for real-time monitoring of CDK4/6i response and early detection of progression in advanced breast cancer. Further, our analysis suggests that amplification of growth-related genes may contribute to combined CDK4/6i and endocrine therapy resistance.
Collapse
Affiliation(s)
- Charlotte K Kindt
- Department of Cancer Research, Institute of Molecular Medicine, University of Southern Denmark, Odense, Denmark
| | - Carla L Alves
- Department of Cancer Research, Institute of Molecular Medicine, University of Southern Denmark, Odense, Denmark
| | - Sidse Ehmsen
- Department of Cancer Research, Institute of Molecular Medicine, University of Southern Denmark, Odense, Denmark
- Department of Oncology, Odense University Hospital; Institute of Clinical Research, University of Southern Denmark, Odense, Denmark
| | - Amalie Kragh
- Department of Oncology, Odense University Hospital; Institute of Clinical Research, University of Southern Denmark, Odense, Denmark
| | - Thomas Reinert
- Department of Molecular Medicine (MOMA), Aarhus University Hospital, Aarhus, Denmark
| | - Marianne Vogsen
- Department of Oncology, Odense University Hospital; Institute of Clinical Research, University of Southern Denmark, Odense, Denmark
| | - Annette R Kodahl
- Department of Oncology, Odense University Hospital; Institute of Clinical Research, University of Southern Denmark, Odense, Denmark
| | - Jeanette D Rønlev
- Department of Oncology, Odense University Hospital; Institute of Clinical Research, University of Southern Denmark, Odense, Denmark
| | | | | | | | | | - Johan Staaf
- Division of Translational Cancer Research, Department of Laboratory Medicine, Lund University, Medicon Village, Lund, Sweden
| | - Henrik J Ditzel
- Department of Cancer Research, Institute of Molecular Medicine, University of Southern Denmark, Odense, Denmark
- Department of Oncology, Odense University Hospital; Institute of Clinical Research, University of Southern Denmark, Odense, Denmark
| |
Collapse
|
35
|
van der Wel JWT, Jebbink M, van den Broek D, Steinbusch LC, Theelen WSME, Ruiter G, Buikhuisen W, Burgers JA, Baas P, Vermeulen M, van der Noort V, Hashemi SMS, Bosch LJW, Monkhorst K, Smit EF, Boelens MC, de Langen AJ. Combined analysis of circulating tumor DNA and tumor tissue to overcome osimertinib resistance (OSIRIS); the second line osimertinib cohort. Lung Cancer 2024; 198:107972. [PMID: 39556978 DOI: 10.1016/j.lungcan.2024.107972] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2024] [Revised: 08/15/2024] [Accepted: 09/27/2024] [Indexed: 11/20/2024]
Abstract
INTRODUCTION Osimertinib resistance inevitably occurs in EGFR mutated NSCLC. We aimed to identify resistance mechanisms (RM) using paired plasma and tumor samples in patients that progressed on 2nd/3rd line osimertinib. METHODS From 09 - 2019 to 02 - 2021, 51 patients were enrolled. Plasma sequencing used AVENIO Expanded Panel (research use only), tumor biopsies underwent DNA and RNA sequencing and histological evaluation. Sequencing was regarded successful when the driver mutation was confirmed with a variant allele frequency of ≥0.10%. Concordance between modalities was calculated for the driver mutation and RMs covered in both modalities. The Molecular Tumor Board formulated a treatment advice. RESULTS The driver mutation was detected in 42/51 plasma samples (82%) and in 50/51 tumor samples (98%), concordance rate was 80%. In 41/51 (80%) patients ≥1 RM was identified. Thirty-two RMs covered in both modalities were found in plasma (61.5%), 39 in tumor (75%), nineteen in both. RM concordance rate was 36.5%. CONCLUSION Combined analysis of plasma and tumor samples post 2nd/3rd line osimertinib identifies additional RMs regardless of the comparative approach used. Plasma sequencing identified 61.5% of RMs, tumor analysis identified 75%. Combined, they provide a superior overview of osimertinib resistance, enabling more tailored treatment options.
Collapse
Affiliation(s)
- J W T van der Wel
- Department of Thoracic Oncology, Netherlands Cancer Institute, The Netherlands
| | - M Jebbink
- Department of Thoracic Oncology, Netherlands Cancer Institute, The Netherlands
| | - D van den Broek
- Department of Laboratory Medicine, Netherlands Cancer Institute, The Netherlands
| | - L C Steinbusch
- Department of Thoracic Oncology, Netherlands Cancer Institute, The Netherlands; Department of Pulmonary Diseases, Leiden University Medical Center, The Netherlands
| | - W S M E Theelen
- Department of Thoracic Oncology, Netherlands Cancer Institute, The Netherlands
| | - G Ruiter
- Department of Thoracic Oncology, Netherlands Cancer Institute, The Netherlands
| | - W Buikhuisen
- Department of Thoracic Oncology, Netherlands Cancer Institute, The Netherlands
| | - J A Burgers
- Department of Thoracic Oncology, Netherlands Cancer Institute, The Netherlands
| | - P Baas
- Department of Thoracic Oncology, Netherlands Cancer Institute, The Netherlands
| | - M Vermeulen
- Department of Biometrics, Netherlands Cancer Institute, The Netherlands
| | - V van der Noort
- Department of Biometrics, Netherlands Cancer Institute, The Netherlands
| | - S M S Hashemi
- Department of Pulmonary Medicine, Amsterdam UMC, VU University Medical Center, The Netherlands
| | - L J W Bosch
- Department of Pathology, Netherlands Cancer Institute, The Netherlands
| | - K Monkhorst
- Department of Pathology, Netherlands Cancer Institute, The Netherlands
| | - E F Smit
- Department of Thoracic Oncology, Netherlands Cancer Institute, The Netherlands; Department of Pulmonary Diseases, Leiden University Medical Center, The Netherlands
| | - M C Boelens
- Department of Pathology, Netherlands Cancer Institute, The Netherlands
| | - A J de Langen
- Department of Thoracic Oncology, Netherlands Cancer Institute, The Netherlands.
| |
Collapse
|
36
|
Sorscher S. Circulating Tumor DNA for Solid Tumor Minimal/Molecular Residual Disease Detection in Cancer Survivors. Am J Med 2024; 137:1168-1169. [PMID: 39147086 DOI: 10.1016/j.amjmed.2024.08.014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/16/2024] [Revised: 08/08/2024] [Accepted: 08/09/2024] [Indexed: 08/17/2024]
Affiliation(s)
- Steven Sorscher
- Dr. Sorscher is a retired clinical oncologist and not currently affiliated with any institution.
| |
Collapse
|
37
|
Lim JH, Shin HT, Park S, Ryu WK, Kim L, Lee KH, Ko SM, Lee SJ, Kim JS, Ryu JS. Bronchial washing fluid sequencing is useful in the diagnosis of lung cancer with necrotic tumor. Transl Oncol 2024; 50:102134. [PMID: 39353233 PMCID: PMC11472095 DOI: 10.1016/j.tranon.2024.102134] [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: 06/28/2024] [Revised: 08/27/2024] [Accepted: 09/19/2024] [Indexed: 10/04/2024] Open
Abstract
BACKGROUND Early-stage lung cancers detected by low-dose computed tomography (CT) often require confirmation through invasive procedures due to the absence of endobronchial lesions. This study assesses the diagnostic utility of bronchial washing fluid (BW) sequencing, a less invasive alternative, aiming to identify patient characteristics most suited for this approach. METHODS From June 2017 to March 2018, we conducted a prospective cohort study by enrolling patients with incidental lung lesions suspected of early-stage lung cancer at two independent hospitals, and 114 were diagnosed with lung cancer while 50 were diagnosed with benign lesions. BW sequencing was performed using a targeted gene panel, and the clinical characteristics of patients detected with cancer through sequencing were identified. RESULTS Malignant cells were detected in 33 patients (28.9 %) through BW cytology. By applying specificity-focused mutation criteria, BW sequencing classified 42 patients (36.8 %) as having cancer. Among the cancer patients who were BW sequencing positive and BW cytology negative, 15 patients (75.0 %) showed necrosis on CT. The sensitivity of BW sequencing was particularly enhanced in patients with necrotic tumors, reaching 75 %. CONCLUSIONS BW sequencing presents a viable, non-invasive diagnostic option for early-stage lung cancer, especially valuable in patients with necrotic lesions. By potentially reducing the reliance on more invasive diagnostic procedures, this method could streamline clinical workflows, decrease patient burden, and improve overall diagnostic efficiency.
Collapse
Affiliation(s)
- Jun Hyeok Lim
- Department of Internal Medicine, Inha University Hospital, Inha University College of Medicine, Incheon, South Korea
| | - Hyun-Tae Shin
- Research Center for Controlling Intercellular Communication (RCIC), Inha University School of Medicine, Inha University, Incheon, South Korea; Department of Dermatology, Inha University Hospital, Inha University College of Medicine, Incheon, South Korea
| | - Sunmin Park
- Department of Internal Medicine, Yonsei University Wonju College of Medicine, Wonju, South Korea
| | - Woo Kyung Ryu
- Department of Internal Medicine, Inha University Hospital, Inha University College of Medicine, Incheon, South Korea
| | - Lucia Kim
- Department of Pathology, Inha University Hospital, Inha University College of Medicine, Incheon, South Korea
| | - Kyung-Hee Lee
- Department of Radiology, Inha University Hospital, Inha University College of Medicine, Incheon, South Korea
| | - Sung Min Ko
- Department of Radiology, Yonsei University Wonju College of Medicine, Wonju, South Korea
| | | | - Jung Soo Kim
- Department of Internal Medicine, Inha University Hospital, Inha University College of Medicine, Incheon, South Korea
| | - Jeong-Seon Ryu
- Department of Internal Medicine, Inha University Hospital, Inha University College of Medicine, Incheon, South Korea.
| |
Collapse
|
38
|
Kazarian AG, Bhanvadia RR, Khene ZE, Gerald T, Brooks B, Lotan Y, Tachibana I, Gaston K, Woldu S, Margulis V. Landscape of Genomic Profiling and Circulating Tumor DNA Among Rare Genitourinary Cancers. Clin Genitourin Cancer 2024; 22:102245. [PMID: 39561633 DOI: 10.1016/j.clgc.2024.102245] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2024] [Accepted: 10/13/2024] [Indexed: 11/21/2024]
Affiliation(s)
| | - Raj R Bhanvadia
- Department of Urology, University of Texas Southwestern, Dallas, TX
| | | | - Thomas Gerald
- Department of Urology, University of Texas Southwestern, Dallas, TX
| | - Bailey Brooks
- Department of Urology, University of Texas Southwestern, Dallas, TX
| | - Yair Lotan
- Department of Urology, University of Texas Southwestern, Dallas, TX
| | - Isamu Tachibana
- Department of Urology, University of Texas Southwestern, Dallas, TX
| | - Kris Gaston
- Department of Urology, University of Texas Southwestern, Dallas, TX
| | - Sol Woldu
- Department of Urology, University of Texas Southwestern, Dallas, TX
| | - Vitaly Margulis
- Department of Urology, University of Texas Southwestern, Dallas, TX
| |
Collapse
|
39
|
Wang C. Mapping the Future: A Comprehensive Bibliometric Analysis of Circulating Tumor DNA in Colorectal Cancer. J Multidiscip Healthc 2024; 17:5473-5486. [PMID: 39600714 PMCID: PMC11590644 DOI: 10.2147/jmdh.s493863] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2024] [Accepted: 11/19/2024] [Indexed: 11/29/2024] Open
Abstract
Purpose Colorectal cancer (CRC) is among the most prevalent malignancies worldwide, with rising incidence and mortality rates presenting substantial public health challenges. Traditional detection methods have inherent limitations, which has led to growing interest in liquid biopsy technologies for the identification of circulating tumor DNA (ctDNA). The aim of this study is to explore the developmental trends and future prospects of ctDNA in colorectal cancer through bibliometric analysis. Methods This bibliometric analysis examines the literature on ctDNA in CRC from 2004 to 2024, utilizing the Web of Science Core Collection database to identify research trends, key areas of interest, and potential future directions. The R package "bibliometrix" and VOSviewer software were employed for bibliometric analysis and visualization. The analysis encompassed an evaluation of publication volume, contributing authors, influential journals, country and institutional contributions, and citation metrics. Results The analysis encompassed a total of 1,054 publications, demonstrating a marked escalation in research activity since 2015. The journal "Cancers" has been identified as the most prolific publisher within this domain. Prominent researchers, including Bardelli A and Sartore-Bianchi A, have made substantial contributions to the field. The United States is the leading country in terms of both publication volume and citation frequency, followed by China and Italy. A keyword analysis identified seven conceptual clusters, with "circulating tumor DNA" and "liquid biopsy" emerging as predominant themes. Conclusion This study emphasizes the evolving emphasis on the clinical applications of ctDNA, encompassing early detection, treatment monitoring, and prognostic assessment in CRC, thereby underscoring its potential as a non-invasive biomarker in oncology.
Collapse
Affiliation(s)
- Chengzu Wang
- Clinical Laboratory, Affiliated Cixi Hospital, Wenzhou Medical University, Zhejiang, People’s Republic of China
| |
Collapse
|
40
|
Yang R, Li T, Zhang S, Shui C, Ma H, Li C. The effect of circulating tumor DNA on the prognosis of patients with head and neck squamous cell carcinoma: a systematic review and meta-analysis. BMC Cancer 2024; 24:1434. [PMID: 39574043 PMCID: PMC11580464 DOI: 10.1186/s12885-024-13116-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2024] [Accepted: 10/28/2024] [Indexed: 11/25/2024] Open
Abstract
BACKGROUND Circulating tumour DNA (ctDNA) has emerged as a valuable liquid biopsy biomarker in the field of oncology, including head and neck squamous cell carcinomas (HNSCCs), offering potential insights into cancer diagnosis, progression, and prognosis. This review aims to comprehensively evaluate the utility of ctDNA as a prognostic biomarker in HNSCC. METHODS PubMed and Ovid were searched as part of our review. Studies that investigated the relationship between ctDNA and prognosis in HNSCC patients were included. Outcomes extracted included basic characteristics, ctDNA details and survival data. Meta-analysis was performed on eligible studies to determine pooled progression-free/recurrence-free survival (RFS/PFS) and overall survival (OS). RESULTS Twenty-two studies were included, involving 5062 HNSCC patients from 11 countries. The meta-analysis demonstrated that the positive ctDNA/methylation detection was associated with worse OS (HR = 2.00, 95% CI 1.35-2.96) and worse PFS/RFS (HR = 3.54, 95% CI 1.05-11.85). Positive ctEBV DNA was associated with poorer OS (HR = 2.86, 95% CI 1.84-4.45) and poorer PFS/RFS (HR = 1.93, 95% CI 1.74-2.13). Positive ctHPV DNA was associated with poorer OS (HR = 1.38, 95% CI 1.07-1.38) but not PFS/PFS (HR = 1.33, 95% CI 0.96-1.85). CONCLUSION Meta-analysis indicates that the status of ctDNA is significantly associated with the prognosis of HNSCC patients, with ctDNA/methylation-negative patients demonstrating better PFS/RFS and OS.
Collapse
Affiliation(s)
- Ruoyi Yang
- Head and Neck Surgery Department, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Hospital and Institute, Sichuan Cancer Center, Affiliated Cancer Hospital of University of Electronic Science and Technology of China, Chengdu, 610041, China
- Department of Oral and Maxillofacial Surgery, School of Stomatology, Guizhou Medical University, Guiyang, 550004, China
| | - Teng Li
- Department of Oral and Maxillofacial Surgery, School of Stomatology, Guizhou Medical University, Guiyang, 550004, China
| | - Sicheng Zhang
- Head and Neck Surgery Department, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Hospital and Institute, Sichuan Cancer Center, Affiliated Cancer Hospital of University of Electronic Science and Technology of China, Chengdu, 610041, China
| | - Chunyan Shui
- Head and Neck Surgery Department, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Hospital and Institute, Sichuan Cancer Center, Affiliated Cancer Hospital of University of Electronic Science and Technology of China, Chengdu, 610041, China
| | - Hong Ma
- Department of Oral and Maxillofacial Surgery, School of Stomatology, Guizhou Medical University, Guiyang, 550004, China.
| | - Chao Li
- Head and Neck Surgery Department, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Hospital and Institute, Sichuan Cancer Center, Affiliated Cancer Hospital of University of Electronic Science and Technology of China, Chengdu, 610041, China.
| |
Collapse
|
41
|
Wei K, Xu Y, Nie C, Wei Q, Xie P, Chen T, Jiang J, Chu X. A Multifunctional Peptide Nucleic Acid/Peptide Copolymer-Based Dual-Mode Biosensor with Macrophage-Hitchhiking for Enhanced Tumor Imaging and Urinalysis. J Am Chem Soc 2024. [PMID: 39563630 DOI: 10.1021/jacs.4c10562] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2024]
Abstract
Biosensors are capable of diagnosing tumors through imaging in vivoor liquid biopsy, but they face the challenges of inefficient delivery into tumor sites and the lack of reliable tumor-associated biomarkers. Herein, we constructed a dual-mode biosensor based on a multifunctional peptide nucleic acid (PNA)/peptide copolymer and DNA tetrahedron for tumor imaging and urinalysis. The biosensor could enter the cancer cells to initiate a microRNA-21-specific catalytic hairpin assembly reaction after cleavage by matrix-metalloprotease (MMP) in the tumor microenvironment, and the MMP cleavage product was released into the bloodstream and then was filtered out by the kidney. As PNA was a synthetic DNA analogue that could not be degraded by nucleases and proteases, it could serve as a reliable synthetic biomarker and be easily detected by high-performance liquid chromatography in urine. Importantly, the biosensor was hitchhiked on the macrophage membrane to realize efficient delivery in the depth of tumor utilizing the macrophage ability of actively homing to the tumor site and infiltrating into the tumor. The results indicated that the signal output of the biosensor was improved remarkably and mice with a tumor volume as little as 30-40 mm3 could be reliably discriminated through urine assay. This innovative macrophage-hitchhiking dual-mode biosensor holds a great potential as a non-invasive and convenient tool for tumor diagnosis and tumor progression evaluation.
Collapse
Affiliation(s)
- Kaiji Wei
- State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, Hunan University, Changsha 410082, China
- State Key Laboratory for the Chemistry and Molecular Engineering of Medicinal Resources, School of Chemistry and Pharmaceutical Sciences, Guangxi Normal University, Guilin 541004, China
| | - Yu Xu
- State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, Hunan University, Changsha 410082, China
| | - Cunpeng Nie
- State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, Hunan University, Changsha 410082, China
| | - Qiaomei Wei
- State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, Hunan University, Changsha 410082, China
| | - Ping Xie
- State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, Hunan University, Changsha 410082, China
| | - Tingting Chen
- State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, Hunan University, Changsha 410082, China
| | - Jianhui Jiang
- State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, Hunan University, Changsha 410082, China
| | - Xia Chu
- State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, Hunan University, Changsha 410082, China
| |
Collapse
|
42
|
Li S, Yuan T, Yuan J, Zhu B, Chen D. Opportunities and challenges of using circulating tumor DNA to predict lung cancer immunotherapy efficacy. J Cancer Res Clin Oncol 2024; 150:501. [PMID: 39545998 PMCID: PMC11568038 DOI: 10.1007/s00432-024-06030-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2024] [Accepted: 11/07/2024] [Indexed: 11/17/2024]
Abstract
Immune checkpoint inhibitors (ICIs), particularly anti-programmed death 1 (PD-1)/ programmed death ligand 1 (PD-L1) antibodies, have led to significant progress in lung cancer treatment. However, only a minority of patients have responses to these therapies. Detecting peripheral blood of circulating tumor DNA (ctDNA) allows minimally invasive diagnosis, characterization, and monitoring of lung cancer. ctDNA has potential to be a prognostic biomarker and a predictor of the response to ICI therapy since it can indicate the genomic status and tumor burden. Recent studies on lung cancer have shown that pretreatment ctDNA analysis can detect residual proliferative disease in the adjuvant immunotherapy setting and evaluate tumor burden in patients with metastatic disease. Early ctDNA dynamics can not only predict the clinical outcome of ICI therapy but also help distinguish between pseudoprogression and real progression. Furthermore, in addition to quantitative assessment, ctDNA can also detect genetic predictors of response to ICI therapy. However, barriers still exist in the application of ctDNA analysis in clinical lung cancer treatment. The predictive value of ctDNA in lung cancer immunotherapy requires further identification and resolution of these challenges. This review aims to summarize the existing data of ctDNA analysis in patients receiving immunotherapy for lung cancer, understand the limitations of clinical treatment, and discuss future research directions.
Collapse
Affiliation(s)
- Shanshan Li
- School of Pharmacy and Bioengineering, Chongqing University of Technology, Chongqing, 400054, China
- Institute of Cancer, Xinqiao Hospital, Third Military Medical University, Chongqing, 400037, China
| | - Ting Yuan
- Institute of Cancer, Xinqiao Hospital, Third Military Medical University, Chongqing, 400037, China
| | - Jing Yuan
- Center for Joint Surgery, Department of Orthopedic Surgery, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, 400010, China
| | - Bo Zhu
- Institute of Cancer, Xinqiao Hospital, Third Military Medical University, Chongqing, 400037, China.
- Chongqing Key Laboratory of Immunotherapy, Xinqiao Hospital, Third Military Medical University, Chongqing, 400037, China.
| | - Degao Chen
- Institute of Cancer, Xinqiao Hospital, Third Military Medical University, Chongqing, 400037, China.
- Chongqing Key Laboratory of Immunotherapy, Xinqiao Hospital, Third Military Medical University, Chongqing, 400037, China.
| |
Collapse
|
43
|
Seyedi S, Harris VK, Kapsetaki SE, Narayanan S, Saha D, Compton Z, Yousefi R, May A, Fakir E, Boddy AM, Gerlinger M, Wu C, Mina L, Huijben S, Gouge DH, Cisneros L, Ellsworth PC, Maley CC. Resistance Management for Cancer: Lessons from Farmers. Cancer Res 2024; 84:3715-3727. [PMID: 39356625 PMCID: PMC11565176 DOI: 10.1158/0008-5472.can-23-3374] [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: 10/28/2023] [Revised: 06/29/2024] [Accepted: 09/23/2024] [Indexed: 10/04/2024]
Abstract
One of the main reasons we have not been able to cure cancers is that treatments select for drug-resistant cells. Pest managers face similar challenges with pesticides selecting for pesticide-resistant insects, resulting in similar mechanisms of resistance. Pest managers have developed 10 principles that could be translated to controlling cancers: (i) prevent onset, (ii) monitor continuously, (iii) identify thresholds below which there will be no intervention, (iv) change interventions in response to burden, (v) preferentially select nonchemical control methods, (vi) use target-specific drugs, (vii) use the lowest effective dose, (viii) reduce cross-resistance, (ix) evaluate success based on long-term management, and (x) forecast growth and response. These principles are general to all cancers and cancer drugs and so could be employed broadly to improve oncology. Here, we review the parallel difficulties in controlling drug resistance in pests and cancer cells. We show how the principles of resistance management in pests might be applied to cancer. Integrated pest management inspired the development of adaptive therapy in oncology to increase progression-free survival and quality of life in patients with cancers where cures are unlikely. These pest management principles have the potential to inform clinical trial design.
Collapse
Affiliation(s)
- Sareh Seyedi
- Arizona Cancer Evolution Center, Arizona State University, Tempe, Arizona
- Center for Biocomputing, Security and Society, Biodesign Institute, Arizona State University, Tempe, Arizona
- School of Life Sciences, Arizona State University, Tempe, Arizona
| | - Valerie K. Harris
- Arizona Cancer Evolution Center, Arizona State University, Tempe, Arizona
- Center for Biocomputing, Security and Society, Biodesign Institute, Arizona State University, Tempe, Arizona
| | - Stefania E. Kapsetaki
- Arizona Cancer Evolution Center, Arizona State University, Tempe, Arizona
- Center for Biocomputing, Security and Society, Biodesign Institute, Arizona State University, Tempe, Arizona
| | - Shrinath Narayanan
- Center for Biocomputing, Security and Society, Biodesign Institute, Arizona State University, Tempe, Arizona
- Department of Ecology and Evolution, University of Lausanne, Lausanne, Switzerland
| | - Daniel Saha
- Arizona Cancer Evolution Center, Arizona State University, Tempe, Arizona
- Center for Biocomputing, Security and Society, Biodesign Institute, Arizona State University, Tempe, Arizona
- School of Life Sciences, Arizona State University, Tempe, Arizona
| | - Zachary Compton
- Arizona Cancer Evolution Center, Arizona State University, Tempe, Arizona
- Center for Biocomputing, Security and Society, Biodesign Institute, Arizona State University, Tempe, Arizona
- School of Life Sciences, Arizona State University, Tempe, Arizona
- University of Arizona Cancer Center, University of Arizona College of Medicine, Tucson, Arizona
| | - Rezvan Yousefi
- Arizona Cancer Evolution Center, Arizona State University, Tempe, Arizona
- Center for Biocomputing, Security and Society, Biodesign Institute, Arizona State University, Tempe, Arizona
- The Polytechnic School, Ira A. Fulton Schools of Engineering, Arizona State University, Tempe, Arizona
| | - Alexander May
- Research Casting International, Quinte West, Ontario, Canada
| | - Efe Fakir
- Istanbul University Cerrahpasa School of Medicine, Istanbul, Turkey
| | - Amy M. Boddy
- Arizona Cancer Evolution Center, Arizona State University, Tempe, Arizona
- Exotic Species Cancer Research Alliance, North Carolina State University, Raleigh, North Carolina
- Department of Anthropology, University of California Santa Barbara, Santa Barbara, California
| | - Marco Gerlinger
- Translational Oncogenomics Laboratory, Centre for Evolution and Cancer, The Institute of Cancer Research, London, United Kingdom
- Gastrointestinal Cancer Unit, The Royal Marsden Hospital, London, United Kingdom
| | - Christina Wu
- Division of Hematology and Medical Oncology, Department of Medicine, Mayo Clinic, Phoenix, Arizona
| | | | - Silvie Huijben
- School of Life Sciences, Arizona State University, Tempe, Arizona
- Center for Evolution and Medicine, Arizona State University, Tempe, Arizona
| | - Dawn H. Gouge
- Department of Entomology, University of Arizona, Tucson, Arizona
| | - Luis Cisneros
- Arizona Cancer Evolution Center, Arizona State University, Tempe, Arizona
- Center for Biocomputing, Security and Society, Biodesign Institute, Arizona State University, Tempe, Arizona
- School of Life Sciences, Arizona State University, Tempe, Arizona
| | | | - Carlo C. Maley
- Arizona Cancer Evolution Center, Arizona State University, Tempe, Arizona
- Center for Biocomputing, Security and Society, Biodesign Institute, Arizona State University, Tempe, Arizona
- School of Life Sciences, Arizona State University, Tempe, Arizona
- Center for Evolution and Medicine, Arizona State University, Tempe, Arizona
| |
Collapse
|
44
|
Zhou Q, Li R, Wu S, Zhang Y, Wang W, Zhu K, Wang M, Huang Z, Duan F. Metastasis-directed ablation of hepatocellular carcinoma with pulmonary oligometastases: a long-term multicenter study. LA RADIOLOGIA MEDICA 2024:10.1007/s11547-024-01907-7. [PMID: 39531158 DOI: 10.1007/s11547-024-01907-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/11/2024] [Accepted: 10/23/2024] [Indexed: 11/16/2024]
Abstract
PURPOSE Ablation is a promising approach for eliminating intrathoracic metastases. We compared the effectiveness of a combination of metastasis-directed ablation and systemic therapy with that of systemic therapy alone for patients with hepatocellular carcinoma (HCC) having pulmonary oligometastases. MATERIALS AND METHODS We analyzed 679 patients with HCC and pulmonary oligometastases from seven tertiary hospitals. A total of 372 patients received systemic therapy (System group), whereas 307 patients received the combination therapy of pulmonary oligometastases ablation and systemic therapy (Ablation + System group). RESULTS The median progression-free survival (PFS) was 9.7 ± 0.6 and 11.5 ± 0.6 months in the System and Ablation + System groups, respectively. The Ablation + System group exhibited significantly better PFS (hazard ratio [HR], 0.71; 95% confidence interval [CI] 0.60-0.85; P < 0.001) and overall survival (OS) (HR, 0.65; 95% CI 0.52-0.81; P < 0.001) than the System group. The subgroup analysis revealed that OS (HR, 0.91; 95% CI 0.65-1.28; P = 0.590) and PFS (HR, 0.81; 95% CI 0.62-1.05; P = 0.100) did not differ between tyrosine kinase inhibitor (TKI) and TKI plus programmed cell death protein-1 (PD-1) inhibitor therapies in the Ablation + system group. In addition, PFS (HR, 0.53; 95% CI 0.38-0.74; P < 0.001) and OS (HR, 0.66; 95% CI 52-0.84; P < 0.001) showed obviously different for intrahepatic tumors with partial response (PR) status. CONCLUSION The application of a combination of ablation of pulmonary oligometastases and systemic therapy resulted in longer PFS and OS than systemic therapy alone.
Collapse
Affiliation(s)
- Qunfang Zhou
- Department of Interventional Radiology, Chinese PLA General Hospital, 28 Fuxing Road, Beijing, China
| | - Ruixia Li
- Department of Liver Surgery, The First Affiliated Hospital of Sun Yat-Sen University, 58 Zhongshan Er Road, Guangzhou, Guangdong Province, China
| | - Songsong Wu
- Department of Ultrasonography, Fujian Provincial Hospital, East Road 134, Fuzhou, Fujian Province, China
| | - Yanyang Zhang
- Department of Interventional Radiology, The Third Affiliated Hospital of Sun Yat-Sen University, Tianhe Road 600, Guangzhou, Guangdong Province, China
| | - Wei Wang
- Department of General Surgery, The First Affiliated Hospital of Jinzhou Medical University, Jinzhou, Liaoning Province, China
| | - Kangshun Zhu
- Department of Minimally Invasive Interventional Radiology, The Second Affiliated Hospital of Guangzhou Medical University, 250 Changgang East Road, Guangzhou, Guangdong Province, China
| | - Murong Wang
- Department of Minimally Invasive Interventional Radiology, Sun Yat-Sen University Cancer Center, 651 Dongfeng East Road, Guangzhou, Guangdong Province, China
| | - Zhimei Huang
- Department of Minimally Invasive Interventional Radiology, Sun Yat-Sen University Cancer Center, 651 Dongfeng East Road, Guangzhou, Guangdong Province, China.
| | - Feng Duan
- Department of Interventional Radiology, Chinese PLA General Hospital, 28 Fuxing Road, Beijing, China.
| |
Collapse
|
45
|
Øgaard N, Jensen SØ, Ørntoft MBW, Demuth C, Rasmussen MH, Henriksen TV, Nors J, Frydendahl A, Lyskjær I, Nesic M, Therkildsen C, Kleif J, Gögenur M, Jørgensen LN, Vilandt J, Seidelin JB, Gotschalck KA, Jaensch C, Andersen B, Løve US, Thorlacius-Ussing O, Andersen PV, Kolbro T, Monti A, Kildsig J, Bondeven P, Schlesinger NH, Iversen LH, Rasmussen M, Gögenur I, Bramsen JB, Andersen CL. Circulating tumour DNA and risk of recurrence in patients with asymptomatic versus symptomatic colorectal cancer. Br J Cancer 2024; 131:1707-1715. [PMID: 39390251 PMCID: PMC11555384 DOI: 10.1038/s41416-024-02867-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2024] [Revised: 09/17/2024] [Accepted: 09/23/2024] [Indexed: 10/12/2024] Open
Abstract
BACKGROUND Multiple initiatives aim to develop circulating tumour DNA (ctDNA) tests for early cancer detection in asymptomatic individuals. The few studies describing ctDNA-testing in both asymptomatic and symptomatic patients report lower ctDNA detection in the asymptomatic patients. Here, we explore if asymptomatic patients differ from symptomatic patients e.g. by including a 'low-ctDNA-shedding' and 'less-aggressive' subgroup. METHODS ctDNA assessment was performed in two independent cohorts of consecutively recruited patients with asymptomatic colorectal cancer (CRC) (Cohort#1: n = 215, Cohort#2: n = 368) and symptomatic CRC (Cohort#1: n = 117, Cohort#2: n = 722). RESULTS After adjusting for tumour stage and size, the odds of ctDNA detection was significantly lower in asymptomatic patients compared to symptomatic patients (Cohort#1: OR: 0.4, 95%CI: 0.2-0.8, Cohort#2: OR: 0.7, 95%CI: 0.5-0.9). Further, the recurrence risk was lower in asymptomatic patients (Cohort#1: sHR: 0.6, 95%CI: 0.3-1.2, Cohort#2: sHR: 0.6, 95%CI: 0.4-1.0). Notably, ctDNA-negative asymptomatic patients had the lowest recurrence risk compared to the symptomatic patients (Cohort#1: sHR: 0.2, 95%CI: 0.1-0.6, Cohort#2: sHR: 0.3, 95%CI: 0.2-0.6). CONCLUSIONS Our study suggests that asymptomatic patients are enriched for a 'low-ctDNA-shedding-low-recurrence-risk' subgroup. Such insights are needed to guide ctDNA-based early-detection initiatives and should prompt discussions about de-escalation of therapy and follow-up for ctDNA-negative asymptomatic CRC patients.
Collapse
Affiliation(s)
- Nadia Øgaard
- Department of Molecular Medicine, Aarhus University Hospital, Aarhus, Denmark
- Institute of Clinical Medicine, Faculty of Health, Aarhus University, Aarhus, Denmark
| | - Sarah Østrup Jensen
- Department of Molecular Medicine, Aarhus University Hospital, Aarhus, Denmark
- Institute of Clinical Medicine, Faculty of Health, Aarhus University, Aarhus, Denmark
| | - Mai-Britt Worm Ørntoft
- Department of Molecular Medicine, Aarhus University Hospital, Aarhus, Denmark
- Department of Surgery, Gødstrup Hospital, Herning, Denmark
| | - Christina Demuth
- Department of Molecular Medicine, Aarhus University Hospital, Aarhus, Denmark
- Institute of Clinical Medicine, Faculty of Health, Aarhus University, Aarhus, Denmark
| | - Mads Heilskov Rasmussen
- Department of Molecular Medicine, Aarhus University Hospital, Aarhus, Denmark
- Institute of Clinical Medicine, Faculty of Health, Aarhus University, Aarhus, Denmark
| | - Tenna Vesterman Henriksen
- Department of Molecular Medicine, Aarhus University Hospital, Aarhus, Denmark
- Institute of Clinical Medicine, Faculty of Health, Aarhus University, Aarhus, Denmark
| | - Jesper Nors
- Department of Molecular Medicine, Aarhus University Hospital, Aarhus, Denmark
- Institute of Clinical Medicine, Faculty of Health, Aarhus University, Aarhus, Denmark
| | - Amanda Frydendahl
- Department of Molecular Medicine, Aarhus University Hospital, Aarhus, Denmark
- Institute of Clinical Medicine, Faculty of Health, Aarhus University, Aarhus, Denmark
| | - Iben Lyskjær
- Department of Molecular Medicine, Aarhus University Hospital, Aarhus, Denmark
- Institute of Clinical Medicine, Faculty of Health, Aarhus University, Aarhus, Denmark
| | - Marijana Nesic
- Department of Molecular Medicine, Aarhus University Hospital, Aarhus, Denmark
- Institute of Clinical Medicine, Faculty of Health, Aarhus University, Aarhus, Denmark
| | - Christina Therkildsen
- Gastro Unit, Surgical Section, Copenhagen University Hospital, Amager-Hvidovre, Denmark
| | - Jakob Kleif
- Gastro Unit, Surgical Section, Copenhagen University Hospital, Amager-Hvidovre, Denmark
- Department of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark
| | - Mikail Gögenur
- Center for Surgical Science, Department of Surgery, Zealand University Hospital, Køge, Denmark
| | | | - Jesper Vilandt
- Department of Surgery, Nordsjællands Hospital, Hillerød, Denmark
| | | | - Kåre Anderson Gotschalck
- Institute of Clinical Medicine, Faculty of Health, Aarhus University, Aarhus, Denmark
- Department of Surgery, Horsens Hospital, Horsens, Denmark
| | | | - Berit Andersen
- Institute of Clinical Medicine, Faculty of Health, Aarhus University, Aarhus, Denmark
- Department of Public Health Programs and University Research Clinic for Cancer Screening, Randers Regional Hospital, Randers, Denmark
| | | | | | | | - Thomas Kolbro
- Department of Surgery, Odense University Hospital, Svendborg, Denmark
| | - Alessio Monti
- Department of Surgery, North Denmark Regional Hospital Hjørring, Hjørring, Denmark
| | - Jeppe Kildsig
- Department of Surgery, Copenhagen University Hospital, Herlev, Denmark
| | - Peter Bondeven
- Department of Surgery, Regional Hospital Randers, Randers, Denmark
| | | | - Lene Hjerrild Iversen
- Institute of Clinical Medicine, Faculty of Health, Aarhus University, Aarhus, Denmark
- Department of Surgery, Aarhus University Hospital, Aarhus, Denmark
| | - Morten Rasmussen
- Digestive Disease Centre, Bispebjerg Hospital, Copenhagen, Denmark
| | - Ismail Gögenur
- Center for Surgical Science, Department of Surgery, Zealand University Hospital, Køge, Denmark
| | - Jesper Bertram Bramsen
- Department of Molecular Medicine, Aarhus University Hospital, Aarhus, Denmark
- Institute of Clinical Medicine, Faculty of Health, Aarhus University, Aarhus, Denmark
| | - Claus Lindbjerg Andersen
- Department of Molecular Medicine, Aarhus University Hospital, Aarhus, Denmark.
- Institute of Clinical Medicine, Faculty of Health, Aarhus University, Aarhus, Denmark.
| |
Collapse
|
46
|
Hong TH, Hwang S, Dasgupta A, Abbosh C, Hung T, Bredno J, Walker J, Shi X, Milenkova T, Horn L, Choi JY, Lee HY, Cho JH, Choi YS, Shim YM, Chai S, Rhodes K, Roychowdhury-Saha M, Hodgson D, Kim HK, Ahn MJ. Clinical Utility of Tumor-Naïve Presurgical Circulating Tumor DNA Detection in Early-Stage NSCLC. J Thorac Oncol 2024; 19:1512-1524. [PMID: 38992468 DOI: 10.1016/j.jtho.2024.07.002] [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] [Received: 05/02/2024] [Revised: 06/15/2024] [Accepted: 07/06/2024] [Indexed: 07/13/2024]
Abstract
OBJECTIVES The use of tumor-informed circulating tumor DNA (ctDNA) testing in patients with early-stage disease before surgery is limited, mainly owing to restricted tissue access and extended turnaround times. This study aimed to evaluate the clinical value of a tumor-naïve, methylation-based cell-free DNA assay in a large cohort of patients with resected NSCLC. METHOD We analyzed presurgical plasma samples from 895 patients with EGFR and anaplastic lymphoma kinase-wild-type, clinical stage I or II NSCLC. The ctDNA status was evaluated for its prognostic significance in relation to tumor volume, metabolic activity, histologic diagnosis, histologic subtypes, and clinical-to-pathologic TNM upstaging. RESULTS Presurgical ctDNA detection was observed in 55 of 414 patients (13%) with clinical stage I lung adenocarcinoma (LUAD) and was associated with poor recurrence-free survival (2-year recurrence-free survival 69% versus 91%; log-rank p < 0.001), approaching that of clinical stage II LUAD. Presurgical ctDNA detection was not prognostic in patients with clinical stage II LUAD or non-LUAD. Within LUAD, tumor volume and positron emission tomography avidity interacted to predict presurgical ctDNA detection. Moreover, presurgical ctDNA detection was predictive of the postsurgical discovery of International Association for the Study of Lung Cancer grade 3 tumors (p < 0.001) and pathologic TNM upstaging (p < 0.001). Notably, presurgical ctDNA detection strongly correlated with higher programmed death-ligand 1 expression in tumors (positive rates 28% versus 55%, p < 0.001), identifying a subgroup likely to benefit from anti-programmed death-ligand 1 therapies. CONCLUSION These findings support the integration of ctDNA testing into routine diagnostic workflows in early-stage NSCLC without the need for tumor tissue profiling. Furthermore, it is clinically useful in identifying patients at high risk who might benefit from innovative treatments, including neoadjuvant immune checkpoint inhibitors.
Collapse
Affiliation(s)
- Tae Hee Hong
- Department of Thoracic Surgery, Samsung Medical Center, Seoul, Republic of Korea; Department of Thoracic and Cardiovascular Surgery, Severance Hospital, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Soohyun Hwang
- Department of Pathology and Translational Genomics, Samsung Medical Center, Seoul, Republic of Korea
| | - Abhijit Dasgupta
- Early Data Science, Oncology Data Science, Oncology R&D, AstraZeneca, Gaithersburg, Maryland
| | - Chris Abbosh
- Translational Medicine, Oncology R&D, AstraZeneca, Cambridge, United Kingdom; SAGA Diagnostics, Cambridge, United Kingdom
| | | | | | - Jill Walker
- Precision Medicine, Oncology R&D, AstraZeneca, Cambridge, United Kingdom
| | - Xiaojin Shi
- Late Development Oncology, AstraZeneca, Gaithersburg, Maryland
| | - Tsveta Milenkova
- Global Medicine Development, AstraZeneca, Cambridge, United Kingdom
| | - Leora Horn
- Late Development Oncology, AstraZeneca, Gaithersburg, Maryland
| | - Joon Young Choi
- Department of Nuclear Medicine, Samsung Medical Center, Seoul, Republic of Korea
| | - Ho Yun Lee
- Department of Radiology and Center for Imaging Science, Samsung Medical Center, Seoul, Republic of Korea
| | - Jong Ho Cho
- Department of Thoracic Surgery, Samsung Medical Center, Seoul, Republic of Korea
| | - Yong Soo Choi
- Department of Thoracic Surgery, Samsung Medical Center, Seoul, Republic of Korea
| | - Young Mog Shim
- Department of Thoracic Surgery, Samsung Medical Center, Seoul, Republic of Korea
| | | | | | | | - Darren Hodgson
- Translational Medicine, Oncology R&D, AstraZeneca, Cambridge, United Kingdom
| | - Hong Kwan Kim
- Department of Thoracic Surgery, Samsung Medical Center, Seoul, Republic of Korea
| | - Myung-Ju Ahn
- Department of Hematology-Oncology, Samsung Medical Center, Seoul, Republic of Korea.
| |
Collapse
|
47
|
Uemura S, Kabe Y, Kitago M, Matsuda S, Abe Y, Hasegawa Y, Hori S, Tanaka M, Nakano Y, Sato Y, Itonaga M, Ono M, Kawakami T, Suematsu M, Kitagawa Y. Prognosis prediction of PDAC via detection of O-glycan altered extracellular vesicles in perioperative sera. Cancer Sci 2024; 115:3718-3728. [PMID: 39285510 PMCID: PMC11531947 DOI: 10.1111/cas.16341] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2024] [Revised: 08/26/2024] [Accepted: 08/30/2024] [Indexed: 11/05/2024] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is a fatal malignancy due to the difficulty in diagnosis and poor prognosis because of the high recurrence rate, necessitating reliable biomarkers to improve the diagnosis and prognosis. However, the existing markers have limitations. We previously identified extracellular vesicles (EVs) recognized by O-glycan-binding lectins (Amaranthus caudatus agglutinin [ACA]) as a novel diagnostic biomarker for PDAC using an EV-counting system (ExoCounter). This retrospective study analyzed changes in ACA-positive EVs in perioperative PDAC serum and its association with prognosis using ExoCounter. Absolute EV levels in the pre- and postoperative sera of 44 patients who underwent curative pancreatectomy for PDAC were quantified using ExoCounter. The carbohydrate antigen 19-9 levels declined in most samples postoperatively, and presented no correlation with poor prognosis. In contrast, ACA-positive EVs increased in serum at 7 days postoperatively in 27 of 44 patients (61.4%). We therefore divided participants with ACA-positive EVs before and after surgery into elevation and decline groups. The overall survival (OS) and recurrence-free survival (RFS) of patients with higher ACA-positive EVs were significantly shorter than those with lower ACA-positive EVs (26.1 months vs. not reached, P = 0.018; 11.9 vs. 38.6 months, P = 0.013). Multivariable analysis revealed that ACA-positive EV elevation in postoperative serum was an independent prognostic factor for poor OS (hazard ratio [HR] = 3.891, P = 0.023) and RFS (HR = 2.650, P = 0.024). The detection of ACA-positive EVs in perioperative serum may be used to predict the prognosis of PDAC in the early postoperative period.
Collapse
Affiliation(s)
- Sho Uemura
- Department of SurgeryKeio University School of MedicineTokyoJapan
| | - Yasuaki Kabe
- Department of BiochemistryKeio University School of MedicineTokyoJapan
- Department of BiochemistryKochi University Medical SchoolNankokuKochiJapan
| | - Minoru Kitago
- Department of SurgeryKeio University School of MedicineTokyoJapan
| | - Sachiko Matsuda
- Department of SurgeryKeio University School of MedicineTokyoJapan
| | - Yuta Abe
- Department of SurgeryKeio University School of MedicineTokyoJapan
| | - Yasushi Hasegawa
- Department of SurgeryKeio University School of MedicineTokyoJapan
| | - Shutaro Hori
- Department of SurgeryKeio University School of MedicineTokyoJapan
| | - Masayuki Tanaka
- Department of SurgeryKeio University School of MedicineTokyoJapan
| | - Yutaka Nakano
- Department of SurgeryKeio University School of MedicineTokyoJapan
| | - Yasunori Sato
- Department of BiostatisticsKeio University School of MedicineTokyoJapan
| | | | - Masayuki Ono
- Future Creation Research LaboratoryJvckenwood CorporationYokohamaJapan
| | - Tatsuya Kawakami
- Future Creation Research LaboratoryJvckenwood CorporationYokohamaJapan
| | - Makoto Suematsu
- Department of BiochemistryKeio University School of MedicineTokyoJapan
| | - Yuko Kitagawa
- Department of SurgeryKeio University School of MedicineTokyoJapan
| |
Collapse
|
48
|
Medford AJ, Carmeli AB, Ritchie A, Wagle N, Garraway L, Lander ES, Parikh A. A standing platform for cancer drug development using ctDNA-based evidence of recurrence. Nat Rev Cancer 2024; 24:810-821. [PMID: 39349822 DOI: 10.1038/s41568-024-00742-2] [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: 08/09/2024] [Indexed: 10/26/2024]
Abstract
The time required to conduct clinical trials limits the rate at which we can evaluate and deliver new treatment options to patients with cancer. New approaches to increase trial efficiency while maintaining rigor would benefit patients, especially in oncology, in which adjuvant trials hold promise for intercepting metastatic disease, but typically require large numbers of patients and many years to complete. We envision a standing platform - an infrastructure to support ongoing identification and trial enrolment of patients with cancer with early molecular evidence of disease (MED) after curative-intent therapy for early-stage cancer, based on the presence of circulating tumour DNA. MED strongly predicts subsequent recurrence, with the vast majority of patients showing radiographic evidence of disease within 18 months. Such a platform would allow efficient testing of many treatments, from small exploratory studies to larger pivotal trials. Trials enrolling patients with MED but without radiographic evidence of disease have the potential to advance drug evaluation because they can be smaller (given high probability of recurrence) and faster (given short time to recurrence) than conventional adjuvant trials. Circulating tumour DNA may also provide a valuable early biomarker of treatment effect, which would allow small signal-finding trials. In this Perspective, we discuss how such a platform could be established.
Collapse
Affiliation(s)
- Arielle J Medford
- Department of Medicine, Division of Hematology and Oncology, Massachusetts General Hospital Cancer Center and Harvard Medical School, Boston, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | | | | | | | | | - Eric S Lander
- Broad Institute of MIT and Harvard, Cambridge, MA, USA.
- Science for America, Cambridge, MA, USA.
- Department of Biology, Massachusetts Institute of Technology, Cambridge, MA, USA.
- Department of Systems Biology, Harvard Medical School, Boston, MA, USA.
| | - Aparna Parikh
- Department of Medicine, Division of Hematology and Oncology, Massachusetts General Hospital Cancer Center and Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
49
|
Nakamura Y, Watanabe J, Akazawa N, Hirata K, Kataoka K, Yokota M, Kato K, Kotaka M, Kagawa Y, Yeh KH, Mishima S, Yukami H, Ando K, Miyo M, Misumi T, Yamazaki K, Ebi H, Okita K, Hamabe A, Sokuoka H, Kobayashi S, Laliotis G, Aushev VN, Sharma S, Jurdi A, Liu MC, Aleshin A, Rabinowitz M, Bando H, Taniguchi H, Takemasa I, Kato T, Kotani D, Mori M, Yoshino T, Oki E. ctDNA-based molecular residual disease and survival in resectable colorectal cancer. Nat Med 2024; 30:3272-3283. [PMID: 39284954 PMCID: PMC11564113 DOI: 10.1038/s41591-024-03254-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2024] [Accepted: 08/19/2024] [Indexed: 11/16/2024]
Abstract
The interim analysis of the CIRCULATE-Japan GALAXY observational study demonstrated the association of circulating tumor DNA (ctDNA)-based molecular residual disease (MRD) detection with recurrence risk and benefit from adjuvant chemotherapy (ACT) in resectable colorectal cancer (CRC). This updated analysis with a 23-month median follow-up, including 2,240 patients with stage II-III colon cancer or stage IV CRC, reinforces the prognostic value of ctDNA positivity during the MRD window with significantly inferior disease-free survival (DFS; hazard ratio (HR): 11.99, P < 0.0001) and overall survival (OS; HR: 9.68, P < 0.0001). In patients who experienced recurrence, ctDNA positivity correlated with shorter OS (HR: 2.71, P < 0.0001). The significantly shorter DFS in MRD-positive patients was consistent across actionable biomarker subsets. Sustained ctDNA clearance in response to ACT was an indicator of favorable DFS and OS compared to transient clearance (24-month DFS: 89.0% versus 3.3%; 24-month OS: 100.0% versus 82.3%). True spontaneous clearance rate with no clinical recurrence was 1.9% (2/105). Overall, our findings provide evidence for the utility of ctDNA monitoring for post-resection recurrence and mortality risk stratification that could be used for guiding adjuvant therapy.
Collapse
Affiliation(s)
- Yoshiaki Nakamura
- Department of Gastrointestinal Oncology, National Cancer Center Hospital East, Kashiwa, Japan
- Translational Research Support Office, National Cancer Center Hospital East, Kashiwa, Japan
- International Research Promotion Office, National Cancer Center Hospital East, Kashiwa, Japan
| | - Jun Watanabe
- Department of Colorectal Surgery, Kansai Medical University, Hirakata, Japan
- Department of Surgery, Gastroenterological Center, Yokohama City University Medical Center, Yokohama, Japan
| | - Naoya Akazawa
- Department of Gastroenterological Surgery, Sendai City Medical Center Sendai Open Hospital, Sendai, Japan
| | - Keiji Hirata
- Department of Surgery 1, School of Medicine, University of Occupational and Environmental Health, Kitakyushu, Japan
| | - Kozo Kataoka
- Division of Lower GI, Department of Gastroenterological Surgery, Hyogo Medical University, Kobe, Japan
| | - Mitsuru Yokota
- Department of General Surgery, Kurashiki Central Hospital, Kurashiki, Japan
| | - Kentaro Kato
- Department of Surgery, Teine-Keijinkai Hospital, Sapporo, Japan
| | | | - Yoshinori Kagawa
- Department of Gastroenterological Surgery, Osaka International Cancer Institute, Osaka, Japan
- Department of Gastroenterological Surgery, Osaka General Medical Center, Osaka, Japan
| | - Kun-Huei Yeh
- Department of Oncology, National Taiwan University Hospital, Taipei, Taiwan
| | - Saori Mishima
- Department of Gastrointestinal Oncology, National Cancer Center Hospital East, Kashiwa, Japan
| | - Hiroki Yukami
- Cancer Chemotherapy Center, Osaka Medical and Pharmaceutical University, Takatsuki, Japan
| | - Koji Ando
- Department of Surgery and Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Masaaki Miyo
- Department of Surgery, Surgical Oncology and Science, Sapporo Medical University, Sapporo, Japan
| | - Toshihiro Misumi
- Department of Data Science, National Cancer Center Hospital East, Kashiwa, Japan
| | - Kentaro Yamazaki
- Division of Gastrointestinal Oncology, Shizuoka Cancer Center, Sunto-gun, Japan
| | - Hiromichi Ebi
- Division of Molecular Therapeutics, Aichi Cancer Center Research Institute, Nagoya, Japan
| | - Kenji Okita
- Department of Surgery, Surgical Oncology and Science, Sapporo Medical University, Sapporo, Japan
| | - Atsushi Hamabe
- Department of Gastroenterological Surgery, Osaka University, Osaka, Japan
| | - Hiroki Sokuoka
- Translational Research Support Office, National Cancer Center Hospital East, Kashiwa, Japan
| | | | | | | | | | | | | | | | | | - Hideaki Bando
- Department of Gastrointestinal Oncology, National Cancer Center Hospital East, Kashiwa, Japan
- Translational Research Support Office, National Cancer Center Hospital East, Kashiwa, Japan
| | - Hiroya Taniguchi
- Department of Clinical Oncology, Aichi Cancer Center Hospital, Nagoya, Japan
| | - Ichiro Takemasa
- Department of Surgery, Surgical Oncology and Science, Sapporo Medical University, Sapporo, Japan
| | - Takeshi Kato
- Department of Surgery, NHO Osaka National Hospital, Osaka, Japan
| | - Daisuke Kotani
- Department of Gastrointestinal Oncology, National Cancer Center Hospital East, Kashiwa, Japan
| | - Masaki Mori
- Tokai University School of Medicine, Isehara, Japan
| | - Takayuki Yoshino
- Department of Gastrointestinal Oncology, National Cancer Center Hospital East, Kashiwa, Japan.
- Kindai University Faculty of Medicine, Osaka-Sayama, Japan.
| | - Eiji Oki
- Department of Surgery and Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan.
| |
Collapse
|
50
|
Payne K, Suriyanarayanan H, Brooks J, Mehanna H, Nankivell P, Gendoo D. Exploring the impact of intra-tumoural heterogeneity on liquid biopsy cell-free DNA methylation and copy number in head and neck squamous cell carcinoma. Oral Oncol 2024; 158:107011. [PMID: 39236578 DOI: 10.1016/j.oraloncology.2024.107011] [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: 04/17/2024] [Revised: 08/01/2024] [Accepted: 08/26/2024] [Indexed: 09/07/2024]
Abstract
Liquid biopsy profiling is gaining increasing promise towards biomarker-led identification and disease stratification of tumours, particularly for tumours displaying significant intra-tumoural heterogeneity (ITH). For head and neck squamous cell carcinoma (HNSCC), which display high levels of genetic ITH, identification of epigenetic modifications and methylation signatures has shown multiple uses in stratification of HNSCC for prognosis, treatment, and HPV status. In this study, we investigated the potential of liquid biopsy methylomics and genomic copy number to profile HNSCC. We conducted multi-region sampling of tumour core, tumour margin and normal adjacent mucosa, as well as plasma cell-free DNA (cfDNA) across 9 HNSCC patients. Collectively, our work highlights the prevalence of methylomic ITH in HNSCC, and demonstrates the potential of cfDNA methylation as a tool for ITH assessment and serial sampling.
Collapse
Affiliation(s)
- Karl Payne
- Institute of Head and Neck Studies and Education, University of Birmingham, Birmingham B15 2TT, United Kingdom.
| | - Harini Suriyanarayanan
- Institute of Cancer and Genomic Sciences, University of Birmingham, Birmingham B15 2TT, United Kingdom
| | - Jill Brooks
- Institute of Head and Neck Studies and Education, University of Birmingham, Birmingham B15 2TT, United Kingdom
| | - Hisham Mehanna
- Institute of Head and Neck Studies and Education, University of Birmingham, Birmingham B15 2TT, United Kingdom
| | - Paul Nankivell
- Institute of Head and Neck Studies and Education, University of Birmingham, Birmingham B15 2TT, United Kingdom
| | - Deena Gendoo
- Institute of Cancer and Genomic Sciences, University of Birmingham, Birmingham B15 2TT, United Kingdom; Institute for Interdisciplinary Data Science and AI, University of Birmingham, Birmingham B15 2TT, United Kingdom.
| |
Collapse
|