1
|
Murray K, Oldfield L, Stefanova I, Gentiluomo M, Aretini P, O'Sullivan R, Greenhalf W, Paiella S, Aoki MN, Pastore A, Birch-Ford J, Rao BH, Uysal-Onganer P, Walsh CM, Hanna GB, Narang J, Sharma P, Campa D, Rizzato C, Turtoi A, Sever EA, Felici A, Sucularli C, Peduzzi G, Öz E, Sezerman OU, Van der Meer R, Thompson N, Costello E. Biomarkers, omics and artificial intelligence for early detection of pancreatic cancer. Semin Cancer Biol 2025; 111:76-88. [PMID: 39986585 DOI: 10.1016/j.semcancer.2025.02.009] [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: 02/13/2025] [Accepted: 02/17/2025] [Indexed: 02/24/2025]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is frequently diagnosed in its late stages when treatment options are limited. Unlike other common cancers, there are no population-wide screening programmes for PDAC. Thus, early disease detection, although urgently needed, remains elusive. Individuals in certain high-risk groups are, however, offered screening or surveillance. Here we explore advances in understanding high-risk groups for PDAC and efforts to implement biomarker-driven detection of PDAC in these groups. We review current approaches to early detection biomarker development and the use of artificial intelligence as applied to electronic health records (EHRs) and social media. Finally, we address the cost-effectiveness of applying biomarker strategies for early detection of PDAC.
Collapse
Affiliation(s)
- Kate Murray
- Liverpool Experimental Cancer Medicine Centre, University of Liverpool, Liverpool, United Kingdom
| | - Lucy Oldfield
- Liverpool Experimental Cancer Medicine Centre, University of Liverpool, Liverpool, United Kingdom
| | - Irena Stefanova
- Liverpool Experimental Cancer Medicine Centre, University of Liverpool, Liverpool, United Kingdom
| | | | | | - Rachel O'Sullivan
- Liverpool Experimental Cancer Medicine Centre, University of Liverpool, Liverpool, United Kingdom
| | - William Greenhalf
- Liverpool Experimental Cancer Medicine Centre, University of Liverpool, Liverpool, United Kingdom
| | - Salvatore Paiella
- Pancreatic Surgery Unit, Department of Surgery, Dentistry, Paediatrics and Gynaecology, University of Verona, Italy
| | - Mateus N Aoki
- Laboratory for Applied Science and Technology in Health, Carlos Chagas Institute, Oswaldo Cruz Foundation (Fiocruz), Brazil
| | - Aldo Pastore
- Fondazione Pisana per la Scienza, Scuola Normale Superiore di Pisa, Italy
| | - James Birch-Ford
- Liverpool Experimental Cancer Medicine Centre, University of Liverpool, Liverpool, United Kingdom
| | - Bhavana Hemantha Rao
- Biomedical Centre, Faculty of Medicine in Pilsen, Charles University, Czech Republic
| | - Pinar Uysal-Onganer
- School of Life Sciences, Cancer Mechanisms and Biomarkers Group, The University of Westminster, United Kingdom
| | - Caoimhe M Walsh
- Department of Surgery and Cancer, Imperial College London, United Kingdom
| | - George B Hanna
- Department of Surgery and Cancer, Imperial College London, United Kingdom
| | | | | | | | | | - Andrei Turtoi
- Tumor Microenvironment and Resistance to Treatment Lab, Institut de Recherche en Cancérologie de Montpellier, INSERM U1194, Université de Montpellier, France
| | - Elif Arik Sever
- Institute of Health Sciences, Acibadem Mehmet Ali Aydinlar University, Turkiye
| | | | | | | | - Elif Öz
- Department of Biostatistics and Bioinformatics, Acibadem Mehmet Ali Aydinlar University, Turkiye
| | - Osman Uğur Sezerman
- Department of Biostatistics and Bioinformatics, Acibadem Mehmet Ali Aydinlar University, Turkiye
| | | | | | - Eithne Costello
- Liverpool Experimental Cancer Medicine Centre, University of Liverpool, Liverpool, United Kingdom.
| |
Collapse
|
2
|
Prasad V, Haslam A, Olivier T. Are Blood-Based Cancer Screening Tests Ready for Primetime? Am J Med 2025; 138:599-600. [PMID: 39522669 DOI: 10.1016/j.amjmed.2024.10.023] [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: 10/14/2024] [Revised: 10/16/2024] [Accepted: 10/16/2024] [Indexed: 11/16/2024]
Affiliation(s)
- Vinay Prasad
- Department of Epidemiology and Biostatistics, University of California San Francisco.
| | - Alyson Haslam
- Department of Epidemiology and Biostatistics, University of California San Francisco
| | | |
Collapse
|
3
|
Jopek MA, Sieczczyński M, Pastuszak K, Łapińska-Szumczyk S, Jassem J, Żaczek AJ, Rondina MT, Supernat A. Impact of clinical factors on accuracy of ovarian cancer detection via platelet RNA profiling. Blood Adv 2025; 9:979-989. [PMID: 39715465 PMCID: PMC11907454 DOI: 10.1182/bloodadvances.2024014008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2024] [Revised: 12/05/2024] [Accepted: 12/05/2024] [Indexed: 12/25/2024] Open
Abstract
ABSTRACT Ovarian cancer (OC) presents a diagnostic challenge, often resulting in poor patient outcomes. Platelet RNA sequencing, which reflects host response to disease, shows promise for earlier OC detection. This study examines the impact of sex, age, platelet count, and the training on cancer types other than OC on classification accuracy achieved in the previous platelet-alone training data set. A total of 339 samples from healthy donors and 1396 samples from patients with cancer, spanning 18 cancer types (including 135 OC cases) were analyzed. Logistic regression was applied to verify our classifiers' performance and interpretability. Models were tested at 100% specificity and 100% sensitivity levels. Incorporating patient age as an additional feature along with gene expression increased sensitivity from 68.6% to 72.6%. Models trained on data from both sexes and on female-only data achieved a sensitivity of 68.6% and 74.5%, respectively. Training solely on OC data reduced late-stage sensitivity from 69.1% to 44.1% but increased early-stage sensitivity from 66.7% to 69.7%. This study highlights the potential of platelet RNA profiling for OC detection and the importance of clinical variables in refining classification accuracy. Incorporating age with gene expression data may enhance OC diagnostic accuracy. The inclusion of male samples deteriorates classifier performance. Data from diverse cancer types improves advanced cancer detection but negatively affects early-stage diagnosis.
Collapse
Affiliation(s)
- Maksym A Jopek
- Laboratory of Translational Oncology, Intercollegiate Faculty of Biotechnology of the University of Gdańsk and the Medical University of Gdańsk, Gdańsk, Poland
- Centre of Biostatistics and Bioinformatics, Medical University of Gdańsk, Gdańsk, Poland
| | - Michał Sieczczyński
- Laboratory of Translational Oncology, Intercollegiate Faculty of Biotechnology of the University of Gdańsk and the Medical University of Gdańsk, Gdańsk, Poland
| | - Krzysztof Pastuszak
- Laboratory of Translational Oncology, Intercollegiate Faculty of Biotechnology of the University of Gdańsk and the Medical University of Gdańsk, Gdańsk, Poland
- Centre of Biostatistics and Bioinformatics, Medical University of Gdańsk, Gdańsk, Poland
- Department of Algorithms and Systems Modelling, Faculty of Electronics, Telecommunications and Informatics, Gdańsk University of Technology, Gdańsk, Poland
| | | | - Jacek Jassem
- Department of Oncology and Radiotherapy, Medical University of Gdańsk, Gdańsk, Poland
| | - Anna J Żaczek
- Laboratory of Translational Oncology, Intercollegiate Faculty of Biotechnology of the University of Gdańsk and the Medical University of Gdańsk, Gdańsk, Poland
| | - Matthew T Rondina
- Molecular Medicine Program, The University of Utah, Salt Lake City, UT
- Division of Hematology and Hematologic Malignancies, Department of Internal Medicine, The University of Utah and Huntsman Cancer Institute, Salt Lake City, UT
- George E. Wahlen Veterans Affairs Medical Center Department of Internal Medicine and the Geriatric Research Education and Clinical Center, Salt Lake City, UT
- Department of Pathology, The University of Utah, Salt Lake City, UT
| | - Anna Supernat
- Laboratory of Translational Oncology, Intercollegiate Faculty of Biotechnology of the University of Gdańsk and the Medical University of Gdańsk, Gdańsk, Poland
- Centre of Biostatistics and Bioinformatics, Medical University of Gdańsk, Gdańsk, Poland
| |
Collapse
|
4
|
Poh KC, Ren TM, Ling GL, Goh JSY, Rose S, Wong A, Mehta SS, Goh A, Chong PY, Cheng SW, Wang SSY, Saffari SE, Lim DWT, Chia NY. Development of a miRNA-Based Model for Lung Cancer Detection. Cancers (Basel) 2025; 17:942. [PMID: 40149278 DOI: 10.3390/cancers17060942] [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: 02/06/2025] [Revised: 03/02/2025] [Accepted: 03/06/2025] [Indexed: 03/29/2025] Open
Abstract
BACKGROUND Lung cancer is the leading cause of cancer-related mortality globally, with late-stage diagnoses contributing to poor survival rates. While lung cancer screening with low-dose computed tomography (LDCT) has proven effective in reducing mortality among heavy smokers, its limitations, including high false-positive rates and resource intensiveness, restrict widespread use. Liquid biopsy, particularly using microRNA (miRNA) biomarkers, offers a promising adjunct to current screening strategies. This study aimed to evaluate the predictive power of a panel of serum miRNA biomarkers for lung cancer detection. PATIENTS AND METHODS A case-control study was conducted at two tertiary hospitals, enrolling 82 lung cancer cases and 123 controls. We performed an extensive literature review to shortlist 25 candidate miRNAs, of which 16 showed a significant two-fold increase in expression compared to the controls. Machine learning techniques, including Random Forest, K-Nearest Neighbors, Neural Networks, and Support Vector Machines, were employed to identify the top six miRNAs. We then evaluated predictive models, incorporating these biomarkers with lung nodule characteristics on LDCT. RESULTS A prediction model utilising six miRNA biomarkers (mir-196a, mir-1268, mir-130b, mir-1290, mir-106b and mir-1246) alone achieved area under the curve (AUC) values ranging from 0.78 to 0.86, with sensitivities of 70-78% and specificities of 73-85%. Incorporating lung nodule size significantly improved model performance, yielding AUC values between 0.96 and 0.99, with sensitivities of 92-98% and specificities of 93-98%. CONCLUSIONS A prediction model combining serum miRNA biomarkers and nodule size showed high predictive power for lung cancer. Integration of the prediction model into current lung cancer screening protocols may improve patient outcomes.
Collapse
Affiliation(s)
- Kai Chin Poh
- Division of Respiratory Medicine, Sengkang General Hospital, Singapore 544886, Singapore
| | - Toh Ming Ren
- Division of Respiratory Medicine, Sengkang General Hospital, Singapore 544886, Singapore
| | - Goh Liuh Ling
- Molecular Diagnostic Laboratory, Tan Tock Seng Hospital, Singapore 308433, Singapore
| | - John S Y Goh
- Professional Officers Division, Singapore Institute of Technology, Singapore 828608, Singapore
| | - Sarrah Rose
- Averywell Limited, Greater Manchester OL8 4QQ, UK
| | - Alexa Wong
- Averywell Limited, Greater Manchester OL8 4QQ, UK
| | | | - Amelia Goh
- Professional Officers Division, Singapore Institute of Technology, Singapore 828608, Singapore
| | - Pei-Yu Chong
- Professional Officers Division, Singapore Institute of Technology, Singapore 828608, Singapore
| | - Sim Wey Cheng
- Molecular Diagnostic Laboratory, Tan Tock Seng Hospital, Singapore 308433, Singapore
| | | | | | | | - Na-Yu Chia
- Averywell Limited, Greater Manchester OL8 4QQ, UK
| |
Collapse
|
5
|
Ghosh AK, Stephens KR, Kandiah PA, Hurt RT, Gilman EA. Multicancer Detection (MCD) Testing in Gastrointestinal Cancers: An Evolving Tool for Early Diagnosis. Curr Gastroenterol Rep 2025; 27:19. [PMID: 40047994 PMCID: PMC11885400 DOI: 10.1007/s11894-025-00970-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/26/2025] [Indexed: 03/09/2025]
Abstract
PURPOSE OF REVIEW The current review aims to summarize the benefits and limitations of the novel multicancer detection tests (MCD) for diagnosing gastrointestinal (GI) malignancies. RECENT FINDINGS Traditional cancer screening methods can reduce deaths in malignancies involving the GI tract. For GI cancers, screening options vary by type and often involve invasive techniques with limited sensitivity. MCDs offer a promising, non-invasive (simple blood draw) alternative by analyzing biomarkers such as cell-free DNA and RNA using advanced techniques and machine learning to detect cancers across multiple organ sites. Large studies like the PATHFINDER trial and THUNDER study have demonstrated the feasibility and accuracy of MCD assays in identifying cancer signals, with high sensitivity and specificity in some GI organs that lack routine screening tests (e.g., liver, pancreas, and stomach). Despite these advancements, MCD testing faces challenges, including high costs, lack of FDA approval, false positives, and limited data on clinical utility in reducing cancer-specific mortality. MCD should not be a substitute for age-appropriate cancer screenings but may complement existing methods, particularly for cancers with no current screening tools, such as cholangiocarcinoma and pancreatic cancer. Clinicians need to discuss the limitations of MCDs, including the potential for overdiagnosis, patient anxiety, and financial burden due to insurance coverage gaps. MCD is a promising, non-invasive test that can augment traditional cancer screening. As the role of MCD in cancer detection evolves, further research is essential to establish how it will be integrated into clinical practice, ensuring informed, shared decision-making with patients.
Collapse
Affiliation(s)
- Aditya K Ghosh
- Division of General Internal Medicine, Mayo Clinic, 200 First Street SW, Rochester, MN, 55905, USA.
| | - Kyle R Stephens
- University of Louisville School of Medicine, Hiram C. Polk Jr., M.D., Department of Surgery, Louisville, KY, USA
| | - Prem A Kandiah
- Department of Neurology, Emory University School of Medicine, Atlanta, GA, USA
| | - Ryan T Hurt
- Division of General Internal Medicine, Mayo Clinic, 200 First Street SW, Rochester, MN, 55905, USA
| | - Elizabeth A Gilman
- Division of General Internal Medicine, Mayo Clinic, 200 First Street SW, Rochester, MN, 55905, USA
| |
Collapse
|
6
|
Walter N, Groth J, Zu Zwerger BVU. Evaluation of an innovative multi-cancer early detection test: high sensitivity and specificity in differentiating cancer, inflammatory conditions, and healthy individuals. Front Oncol 2025; 15:1520869. [PMID: 40115017 PMCID: PMC11922694 DOI: 10.3389/fonc.2025.1520869] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2024] [Accepted: 02/11/2025] [Indexed: 03/22/2025] Open
Abstract
Background Cancer is a leading cause of death worldwide, with early detection crucial for effective treatment. Traditional diagnostic methods, such as imaging and biopsies, are often limited by invasiveness, cost, and sensitivity. Blood-based multi-cancer early detection (MCED) tests offer a less invasive and potentially more comprehensive approach. Recently, a novel screening tool, the Carcimun® test was reported, detecting conformational changes in plasma proteins through optical extinction measurements. This study evaluates the Carcimun® test's performance, including participants with inflammatory conditions. Methods This prospective, single-blinded study included 172 participants: 80 healthy volunteers, 64 cancer patients (various types), and 28 individuals with inflammatory conditions (fibrosis, sarcoidosis, pneumonia) or benign tumors. Plasma samples were analyzed using the Carcimun® test. Sensitivity, specificity, positive predictive value (PPV), and negative predictive value (NPV) were calculated. Results Mean extinction values were significantly higher in cancer patients (315.1) compared to healthy individuals (23.9) and those with inflammatory conditions (62.7) (p<0.001). The Carcimun® test distinguished these groups with high accuracy (95.4%), sensitivity (90.6%), and specificity (98.2%). Significant differences were found between healthy participants and cancer patients (p<0.001), and between cancer patients and those with inflammation (p<0.001). Conclusion The Carcimun® test achieved high accuracy, sensitivity, and specificity, effectively identifying cancer patients while minimizing false positives and negatives. By including participants with inflammatory conditions, we addressed a significant limitation of previous studies, demonstrating the test's robustness in real-world clinical scenarios. These findings underscore the potential of the Carcimun® test as a valuable tool for early cancer detection and screening.
Collapse
Affiliation(s)
- Nike Walter
- University Hospital Regensburg, Regensburg, Germany
| | | | | |
Collapse
|
7
|
Bruhm DC, Vulpescu NA, Foda ZH, Phallen J, Scharpf RB, Velculescu VE. Genomic and fragmentomic landscapes of cell-free DNA for early cancer detection. Nat Rev Cancer 2025:10.1038/s41568-025-00795-x. [PMID: 40038442 DOI: 10.1038/s41568-025-00795-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 01/24/2025] [Indexed: 03/06/2025]
Abstract
Genomic analyses of cell-free DNA (cfDNA) in plasma are enabling noninvasive blood-based biomarker approaches to cancer detection and disease monitoring. Current approaches for identification of circulating tumour DNA typically use targeted tumour-specific mutations or methylation analyses. An emerging approach is based on the recognition of altered genome-wide cfDNA fragmentation in patients with cancer. Recent studies have revealed a multitude of characteristics that can affect the compendium of cfDNA fragments across the genome, collectively called the 'cfDNA fragmentome'. These changes result from genomic, epigenomic, transcriptomic and chromatin states of an individual and affect the size, position, coverage, mutation, structural and methylation characteristics of cfDNA. Identifying and monitoring these changes has the potential to improve early detection of cancer, especially using highly sensitive multi-feature machine learning approaches that would be amenable to broad use in populations at increased risk. This Review highlights the rapidly evolving field of genome-wide analyses of cfDNA characteristics, their comparison to existing cfDNA methods, and recent related innovations at the intersection of large-scale sequencing and artificial intelligence. As the breadth of clinical applications of cfDNA fragmentome methods have enormous public health implications for cancer screening and personalized approaches for clinical management of patients with cancer, we outline the challenges and opportunities ahead.
Collapse
Affiliation(s)
- Daniel C Bruhm
- The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Nicholas A Vulpescu
- The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Zachariah H Foda
- The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Jillian Phallen
- The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Robert B Scharpf
- The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Biostatistics, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - Victor E Velculescu
- The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA.
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA.
| |
Collapse
|
8
|
Tyson C, Li KH, Cao X, O’Brien JM, Fishman EK, O’Donnell EK, Duran C, Parthasarathy V, Rego SP, Choudhry OA, Beer TM. Tumor localization strategies of multicancer early detection tests: a quantitative assessment. JNCI Cancer Spectr 2025; 9:pkaf011. [PMID: 39854284 PMCID: PMC11897890 DOI: 10.1093/jncics/pkaf011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2024] [Revised: 12/12/2024] [Accepted: 01/17/2025] [Indexed: 01/26/2025] Open
Abstract
BACKGROUND Multicancer early detection tests may expand cancer screening. Characterizing diagnostic resolution approaches following positive multicancer early detection tests is critical. Two trials employed distinct resolution approaches: a molecular signal to predict tissue of origin and an imaging-based diagnostic strategy. This modeling study characterizes diagnostic journeys and impact in a hypothetical population of average-risk multicancer early detection-eligible patients. METHODS A mathematical expression for diagnostic burden was derived using positive predictive value (PPV), molecular tissue of origin localization accuracy, and numbers of procedures associated with each diagnostic outcome. Imaging-based and molecular tissue of origin-informed strategies were compared. Excess lifetime cancer risk due to futile radiation exposure was estimated using organ-specific diagnostic imaging radiation doses. RESULTS Across all PPVs and localization performances, a molecular tissue of origin strategy resulted in a higher diagnostic burden (mean = 3.6 [0.445] procedures vs mean = 2.6 [0.100] procedures) for the imaging strategy. Estimated diagnostic burden was higher for molecular tissue of origin in 95.5% of all PPV and tissue of origin accuracy combinations; at least 79% PPV and 90% accuracy would be required for a molecular tissue of origin-informed strategy to be less burdensome than imaging. The maximum rate of excess cancer incidence from radiation exposure for multicancer early detection false-positive results (individuals aged 50-84 years) was 64.6 of 100 000 (annual testing, 99% specificity), 48.5 of 100 000 (biennial testing, 98.5% specificity), and 64.6 of 100 000 (biennial testing, 98% specificity). CONCLUSIONS An imaging-based diagnostic strategy is more efficient than a molecular tissue of origin-informed approach across almost all PPV and tissue of origin accuracy combinations. The use of an imaging-based approach for cancer localization can be efficient and low-risk compared with a molecular-informed approach.
Collapse
Affiliation(s)
| | - Kevin H Li
- Exact Sciences Corporation, Madison, WI 53719, United States
| | - Xiting Cao
- Exact Sciences Corporation, Madison, WI 53719, United States
| | - James M O’Brien
- Visionquest Development Partners, Atlanta, GA 30240, United States
| | - Elliot K Fishman
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD 21231, United States
| | | | - Carlos Duran
- Exact Sciences Corporation, Madison, WI 53719, United States
| | | | - Seema P Rego
- Exact Sciences Corporation, Madison, WI 53719, United States
| | | | - Tomasz M Beer
- Exact Sciences Corporation, Madison, WI 53719, United States
| |
Collapse
|
9
|
Song J, Ye X, Xiao H. Liquid biopsy entering clinical practice: Past discoveries, current insights, and future innovations. Crit Rev Oncol Hematol 2025; 207:104613. [PMID: 39756526 DOI: 10.1016/j.critrevonc.2025.104613] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2024] [Revised: 12/22/2024] [Accepted: 01/02/2025] [Indexed: 01/07/2025] Open
Abstract
In recent years, liquid biopsy has gained prominence as an emerging biomarker in cancer research, providing critical insights into tumor biology and metastasis. Technological advancements have enabled its integration into clinical practice, with ongoing trials demonstrating encouraging outcomes. Key applications of liquid biopsy include early cancer detection, cancer staging, prognosis evaluation, and real-time monitoring of tumor progression to optimize treatment decisions. In this review, we present a comprehensive conceptual framework for liquid biopsy, discuss the challenges in its research and clinical application, and highlight its significant potential in identifying therapeutic targets and resistance mechanisms across various cancer types. Furthermore, we explore the emerging role of liquid biopsy-based multicancer screening, which has shown promising advancements. Looking ahead, standardization, multi-omics coanalysis, and the advancement of precision medicine and personalized treatments are expected to drive the future development and integration of liquid biopsy into routine clinical workflows, enhancing cancer diagnosis and treatment management.
Collapse
Affiliation(s)
- Jinghan Song
- Department of Respiratory and Critical Care Medicine, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xiong Ye
- School of Clinical Medicine, Shanghai University of Medicine & Health Sciences, Shanghai, China
| | - Hui Xiao
- Department of Respiratory and Critical Care Medicine, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| |
Collapse
|
10
|
Weiss NS. Reducing-a little-the high price of randomized trials of the efficacy of multicancer early detection. J Natl Cancer Inst 2025; 117:391-392. [PMID: 39699239 PMCID: PMC11884845 DOI: 10.1093/jnci/djae292] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2024] [Accepted: 11/04/2024] [Indexed: 12/20/2024] Open
Affiliation(s)
- Noel S Weiss
- Department of Epidemiology, University of Washington, Seattle, WA 98195, United States
- Fred Hutchinson Cancer Center, Seattle, WA 98109, United States
| |
Collapse
|
11
|
Chatanaka MK, Yousef GM, Diamandis EP. The Unholy Grail of cancer screening: or is it just about the Benjamins? Clin Chem Lab Med 2025; 63:499-506. [PMID: 39301604 DOI: 10.1515/cclm-2024-1013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2024] [Accepted: 09/12/2024] [Indexed: 09/22/2024]
Abstract
The biotechnology company Grail developed a non-invasive blood test (Galleri test) which is claimed to detect 50 types of cancer at early and potentially curable stages. The initially promising results from prospective studies, and the anticipated financial success of Grail led the sequencing giant Illumina to purchase Grail for $8 billion (2021). Following this event, Grail collaborated with the UK National Health System to further clarify the test's capability, in a 3-year prospective trial, along with the standard of care. The UK-NHS announced that the trial will provide a clearer understanding of the efficacy of the Galleri test within the NHS framework. If the test does not perform as expected, valuable insights will still be gained to guide future research aimed at enhancing cancer screening. We previously expressed concerns about the sensitivity and specificity of the Galleri test. In this opinion paper, we revisit the hyped technology, and we provide new suggestions on the use of this test.
Collapse
Affiliation(s)
- Miyo K Chatanaka
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, Canada
| | - George M Yousef
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, Canada
- Laboratory Medicine Program, 7989 University Health Network , Toronto, ON, Canada
| | - Eleftherios P Diamandis
- Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Sinai Health System, Toronto, Canada
| |
Collapse
|
12
|
Lippi G, Lackner KJ, Melichar B, Schlattmann P, Greaves R, Gillery P, Plebani M. Multi-cancer early detection: searching for evidence. Clin Chem Lab Med 2025; 63:459-461. [PMID: 39441817 DOI: 10.1515/cclm-2024-1201] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2024]
Affiliation(s)
- Giuseppe Lippi
- Section of Clinical Biochemistry, 19051 University of Verona , Verona, Italy
| | - Karl J Lackner
- Institute of Clinical Chemistry and Laboratory Medicine, University Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany
| | - Bohuslav Melichar
- Department of Oncology, Palacky University Medical School and University Hospital, Olomouc, Czech Republic
| | - Peter Schlattmann
- Institute for Medical Statistics, Computer and Data Sciences (IMSID), University Hospital Jena, Jena, Germany
| | - Ronda Greaves
- Biochemical Genetics, Victorian Clinical Genetics Services Ltd, Bundoora, VIC, Australia
| | - Philippe Gillery
- Laboratory of Pediatric Biology and Research, University Hospital of Reims, Reims, France
| | | |
Collapse
|
13
|
Hong TH, Hwang S, Abbosh C, Dasgupta A, Jeon YJ, Lee J, Park SY, Cho JH, Choi YS, Shim YM, Hung T, Bredno J, Hodgson D, Ahn MJ, Kim HK. Association of pre-surgical circulating tumor DNA detection, use of sublobar resection with risk of recurrence in stage I non-small cell lung cancer. Eur J Cancer 2025; 217:115237. [PMID: 39826198 DOI: 10.1016/j.ejca.2025.115237] [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: 10/19/2024] [Revised: 01/04/2025] [Accepted: 01/09/2025] [Indexed: 01/22/2025]
Abstract
BACKGROUND Sublobar resection is increasingly recognized as an effective treatment for early-stage NSCLC. However, no studies to date have investigated the potential role of preoperative ctDNA detection in guiding surgical decisions, such as opting for sublobar resection, in stage I NSCLC. METHODS Patients with solid-dominant (CTR>0.5), clinical stage I NSCLC were prospectively recruited between March 2014 and December 2020. Pre-surgical plasma samples were analyzed using a tumor-naïve, methylation-based cell-free DNA assay. The impact of sublobar resection versus lobectomy on recurrence-free survival (RFS) was assessed according to pre-surgical ctDNA status. Associations between pre-surgical ctDNA detection and clinicopathologic factors were also investigated. RESULTS The analysis included 544 patients (178 women [33 %]; median age 66 [IQR, 60-71] years). Pre-surgical ctDNA was detected in 188 (35 %) patients. In patients without presurgical ctDNA, sublobar resection did not significantly increase the risk of relapse (adjusted HR, 1.01, p = 0.98). However, among ctDNA-positive patients, sublobar resection was associated with an increased risk of relapse (adjusted HR, 2.25; 95 % CI, 1.12-4.54; p = 0.024). Patients with presurgical ctDNA had higher rates of nodal upstaging (OR, 3.58; p < 0.001) and exhibited higher pathologic grade (p = 0.021), perineural invasion (p < 0.001), and lymphovascular invasion (p < 0.001). CONCLUSIONS Pre-surgical tumor-naïve ctDNA analysis holds promise in identifying patients with aggressive tumors that may not be sufficiently managed with sublobar resection. This approach can help personalize treatment strategies, potentially improving outcomes for patients with early-stage NSCLC.
Collapse
Affiliation(s)
- Tae Hee Hong
- Department of Thoracic and Cardiovascular Surgery, Sungkyunkwan University School of Medicine, Samsung Medical Center, Seoul, South Korea; Department of Thoracic and Cardiovascular Surgery, Severance Hospital, Yonsei University College of Medicine, Seoul, South Korea
| | - Soohyun Hwang
- Department of Pathology and Translational Genomics, Sungkyunkwan University School of Medicine, Samsung Medical Center, Seoul, South Korea; Lunit Inc., Seoul, South Korea
| | - Chris Abbosh
- Translational Medicine, Oncology R&D, AstraZeneca, Cambridge, UK
| | - Abhijit Dasgupta
- Early Data Science, Oncology Data Science R&D, AstraZeneca, Gaithersburg, MD, USA
| | - Yeong Jeong Jeon
- Department of Thoracic and Cardiovascular Surgery, Sungkyunkwan University School of Medicine, Samsung Medical Center, Seoul, South Korea
| | - Junghee Lee
- Department of Thoracic and Cardiovascular Surgery, Sungkyunkwan University School of Medicine, Samsung Medical Center, Seoul, South Korea
| | - Seong Yong Park
- Department of Thoracic and Cardiovascular Surgery, Sungkyunkwan University School of Medicine, Samsung Medical Center, Seoul, South Korea
| | - Jong Ho Cho
- Department of Thoracic and Cardiovascular Surgery, Sungkyunkwan University School of Medicine, Samsung Medical Center, Seoul, South Korea
| | - Yong Soo Choi
- Department of Thoracic and Cardiovascular Surgery, Sungkyunkwan University School of Medicine, Samsung Medical Center, Seoul, South Korea
| | - Young Mog Shim
- Department of Thoracic and Cardiovascular Surgery, Sungkyunkwan University School of Medicine, Samsung Medical Center, Seoul, South Korea; Patient-Centered Outcomes Research Institute, Samsung Medical Center, Seoul, South Korea
| | | | | | - Darren Hodgson
- Translational Medicine, Oncology R&D, AstraZeneca, Cambridge, UK
| | - Myung-Ju Ahn
- Division of Hematology-Oncology, Department of Medicine, Sungkyunkwan University School of Medicine, Samsung Medical Center, Seoul, South Korea.
| | - Hong Kwan Kim
- Department of Thoracic and Cardiovascular Surgery, Sungkyunkwan University School of Medicine, Samsung Medical Center, Seoul, South Korea; Patient-Centered Outcomes Research Institute, Samsung Medical Center, Seoul, South Korea; Department of Clinical Research Design and Evaluation, SAIHST, Sungkyunkwan University, Seoul, South Korea.
| |
Collapse
|
14
|
Eisenstein M. Putting early cancer detection to the test. Nature 2025:10.1038/d41586-025-00530-4. [PMID: 39984691 DOI: 10.1038/d41586-025-00530-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/23/2025]
|
15
|
Nguyen LHD, Nguyen THH, Le VH, Bui VQ, Nguyen LH, Pham NH, Phan TH, Nguyen HT, Tran VS, Bui CV, Vo VK, Nguyen PTN, Dang HHP, Pham VD, Cao VT, Phan NM, Tieu BL, Nguyen GTH, Vo DH, Tran TH, Nguyen TD, Nguyen VTC, Nguyen TH, Tran VU, Le MP, Tran TMT, Nguyen Nguyen M, Van TTV, Nguyen AN, Nguyen TT, Doan NNT, Nguyen HT, Doan PL, Huynh LAK, Nguyen TA, Nguyen HTP, Lu YT, Cao CTT, Nguyen VT, Le Quyen Le T, Luong TLA, Doan TKP, Dao TT, Phan CD, Nguyen TX, Pham NT, Nguyen BT, Pham TTT, Le HL, Truong CT, Jasmine TX, Le MC, Phan VB, Truong QB, Tran THL, Huynh MT, Tran TQ, Nguyen ST, Tran V, Tran VK, Nguyen Nguyen H, Nguyen DS, Van Phan T, Do TTT, Truong DK, Tang HS, Giang H, Nguyen HN, Phan MD, Tran LS. Prospective validation study: a non-invasive circulating tumor DNA-based assay for simultaneous early detection of multiple cancers in asymptomatic adults. BMC Med 2025; 23:90. [PMID: 39948555 PMCID: PMC11827191 DOI: 10.1186/s12916-025-03929-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/17/2024] [Accepted: 02/06/2025] [Indexed: 02/16/2025] Open
Abstract
BACKGROUND Non-invasive multi-cancer early detection (MCED) tests have shown promise in enhancing early cancer detection. However, their clinical utility across diverse populations remains underexplored, limiting their routine implementation. This study aims to validate the clinical utility of a multimodal non-invasive circulating tumor DNA (ctDNA)-based MCED test, SPOT-MAS (Screening for the Presence Of Tumor by DNA Methylation And Size). METHODS We conducted a multicenter prospective study, K-DETEK (ClinicalTrials.gov identifier: NCT05227261), involving 9057 asymptomatic individuals aged 40 years or older across 75 major hospitals and one research institute in Vietnam. Participants were followed for 12 months. RESULTS Of the 9024 eligible participants, 43 (0.48%) tested positive for ctDNA. Among these, 17 were confirmed with malignant lesions in various primary organs through standard-of-care (SOC) imaging and biopsy, with 9 cases matching our tissue of origin (TOO) predictions. This resulted in a positive predictive value of 39.53% (95%CI 26.37-54.42) and a TOO accuracy of 52.94% (95%CI 30.96-73.83). Among the 8981 participants (99.52%) who tested negative, 8974 were confirmed cancer-free during a 12-month period after testing, yielding a negative predictive value of 99.92% (95% CI 99.84-99.96). The test demonstrated an overall sensitivity of 70.83% (95%CI 50.83-85.09) and a specificity of 99.71% (95% CI 99.58-99.80) for detecting various cancer types, including those without SOC screening options. CONCLUSIONS This study presents a prospective validation of a multi-cancer early detection (MCED) test conducted in a lower middle-income country, demonstrating the potential of SPOT-MAS for early cancer detection. Our findings indicate that MCED tests could be valuable additions to national cancer screening programs, particularly in regions where such initiatives are currently limited. TRIAL REGISTRATION ClinicalTrials.gov ID: NCT05227261. Date of registration: 07/02/2022.
Collapse
Affiliation(s)
| | | | - Van Hoi Le
- National Cancer Hospital, Hanoi, Vietnam
| | | | - Lan Hieu Nguyen
- Hanoi Medical University Hospital, Hanoi Medical University, Hanoi, Vietnam
| | | | | | | | | | | | - Van Kha Vo
- Cantho Oncology Hospital, Can Tho, Vietnam
| | | | | | - Van Dung Pham
- Thong Nhat Dongnai General Hospital, Bien Hoa, Vietnam
| | | | | | - Ba Linh Tieu
- Medical Genetics Institute, Ho Chi Minh, Vietnam
| | | | - Dac Ho Vo
- Medical Genetics Institute, Ho Chi Minh, Vietnam
| | | | | | | | | | - Vu Uyen Tran
- Medical Genetics Institute, Ho Chi Minh, Vietnam
| | | | | | | | | | | | | | | | | | | | | | | | | | - Y-Thanh Lu
- Medical Genetics Institute, Ho Chi Minh, Vietnam
| | | | | | | | - Thi Lan-Anh Luong
- Hanoi Medical University Hospital, Hanoi Medical University, Hanoi, Vietnam
| | | | - Thi Trang Dao
- Hanoi Medical University Hospital, Hanoi Medical University, Hanoi, Vietnam
| | | | | | | | | | | | | | | | | | - Minh Chi Le
- University Medical Center HCM, Ho Chi Minh, Vietnam
| | | | | | | | | | | | | | - Vu Tran
- Thong Nhat Dongnai General Hospital, Bien Hoa, Vietnam
| | | | - Huu Nguyen Nguyen
- Medical Genetics Institute, Ho Chi Minh, Vietnam
- Gene Solutions, Ho Chi Minh, Vietnam
| | - Duy Sinh Nguyen
- Medical Genetics Institute, Ho Chi Minh, Vietnam
- Gene Solutions, Ho Chi Minh, Vietnam
| | - Thi Van Phan
- Medical Genetics Institute, Ho Chi Minh, Vietnam
| | | | | | | | - Hoa Giang
- Medical Genetics Institute, Ho Chi Minh, Vietnam
- Gene Solutions, Ho Chi Minh, Vietnam
| | - Hoai-Nghia Nguyen
- Medical Genetics Institute, Ho Chi Minh, Vietnam
- Gene Solutions, Ho Chi Minh, Vietnam
| | - Minh-Duy Phan
- Medical Genetics Institute, Ho Chi Minh, Vietnam.
- Gene Solutions, Ho Chi Minh, Vietnam.
| | - Le Son Tran
- Medical Genetics Institute, Ho Chi Minh, Vietnam.
- Gene Solutions, Ho Chi Minh, Vietnam.
| |
Collapse
|
16
|
Heller M, Mann DA, Katona BW. Current Approaches of Pancreatic Cancer Surveillance in High-Risk Individuals. J Gastrointest Cancer 2025; 56:61. [PMID: 39932614 DOI: 10.1007/s12029-025-01184-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/25/2025] [Indexed: 02/14/2025]
Abstract
Currently, those recommended to undergo pancreatic cancer (PC) surveillance include appropriately aged individuals at high risk of PC due to an identifiable genetic susceptibility or those without identifiable genetic susceptibility who nonetheless have a strong family history of PC. With increases in identification of individuals at high risk for PC and increased use of PC surveillance in clinical practice, there has been increasing debate about who should undergo surveillance as well as how surveillance should be performed including use of imaging and blood-based testing. Furthermore, there is increasing interest in the outcomes of PC surveillance in high-risk individuals with some studies demonstrating that surveillance leads to downstaging of PC and improvements in survival. In this review, we summarize the current state of PC surveillance in high-risk individuals, providing an overview of the risk factors associated with PC, selection of high-risk individuals for PC surveillance, and the current, but non-uniform, recommendations for performing PC surveillance. Additionally, we review approaches to apply various imaging and blood-based tests to surveillance and the outcomes of PC surveillance.
Collapse
Affiliation(s)
- Melissa Heller
- Division of Hematology and Oncology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Derek A Mann
- Division of Hematology and Oncology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Bryson W Katona
- Division of Gastroenterology and Hepatology, University of Pennsylvania Perelman School of Medicine, 3400 Civic Center Blvd., 751 South Pavilion, Philadelphia, PA, 19104, USA.
| |
Collapse
|
17
|
Alix-Panabières C, Pantel K. Advances in liquid biopsy: From exploration to practical application. Cancer Cell 2025; 43:161-165. [PMID: 39672165 DOI: 10.1016/j.ccell.2024.11.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/24/2024] [Revised: 11/02/2024] [Accepted: 11/15/2024] [Indexed: 12/15/2024]
Abstract
Liquid biopsy has received tremendous attention as a non-invasive approach for detecting and tracking cancer. Here, we discuss the latest work on circulating tumor DNA and circulating tumor cells with respect to clinical applications, including cancer screening, early detection of relapse, real-time monitoring of therapeutic efficacy, and detection of therapeutic targets and resistance mechanisms.
Collapse
Affiliation(s)
- Catherine Alix-Panabières
- Laboratory of Rare Human Circulating Cells (LCCRH) and Liquid Biopsy, University Medical Centre of Montpellier, Montpellier, France; CREEC (CREES), Unité Mixte de Recherches, IRD 224-CNRS 5290-Université de Montpellier, Montpellier, France; European Liquid Biopsy Society (ELBS), Hamburg, Germany; Department of Tumor Biology, University Medical Center, Hamburg-Eppendorf, Hamburg, Germany.
| | - Klaus Pantel
- European Liquid Biopsy Society (ELBS), Hamburg, Germany; Department of Tumor Biology, University Medical Center, Hamburg-Eppendorf, Hamburg, Germany.
| |
Collapse
|
18
|
Lei S, Jia S, Takalkar S, Chang TC, Ma X, Szlachta K, Xu K, Cheng Z, Hui Y, Koo SC, Mead PE, Gao Q, Kumar P, Bailey CP, Sunny J, Pappo AS, Federico SM, Robinson GW, Gajjar A, Rubnitz JE, Jeha S, Pui CH, Inaba H, Wu G, Klco JM, Tatevossian RG, Mullighan CG. Genomic profiling of circulating tumor DNA for childhood cancers. Leukemia 2025; 39:420-430. [PMID: 39523434 DOI: 10.1038/s41375-024-02461-x] [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: 08/09/2024] [Revised: 10/30/2024] [Accepted: 11/01/2024] [Indexed: 11/16/2024]
Abstract
The utility of circulating tumor DNA (ctDNA) analysis has not been well-established for disease detection and monitoring of childhood cancers, especially leukemias. We developed PeCan-Seq, a deep sequencing method targeting diverse somatic genomic variants in cell-free samples in childhood cancer. Plasma samples were collected at diagnosis from 233 children with hematologic, solid and brain tumors. All children with hematologic malignancy (n = 177) had detectable ctDNA at diagnosis. The median ctDNA fraction was 0.77, and 97% of 789 expected tumor variants were identified, including sequence mutations, copy number variations, and structural variations responsible for oncogenic fusions. In contrast, ctDNA was detected in 19 of 38 solid tumor patients and 1 of 18 brain tumor patients. Somatic variants from ctDNA were correlated with minimal residual disease levels as determined by flow cytometry in serial plasma samples from patients with B-cell acute lymphoblastic leukemia (B-ALL). We showcase multi-tumor detection by ctDNA analysis for a patient with concurrent B-ALL and neuroblastoma. In conclusion, PeCan-seq sensitively identified heterogeneous ctDNA alterations from 1 mL plasma for childhood hematologic malignancies and a subset of solid tumors. PeCan-seq provides a robust, non-invasive approach to augment comprehensive genomic profiling at diagnosis and mutation-specific detection during disease monitoring.
Collapse
Affiliation(s)
- Shaohua Lei
- Department of Pathology, St. Jude Children's Research Hospital, Memphis, TN, USA
- Center of Excellence for Leukemia Studies, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Sujuan Jia
- Clinical Biomarkers Laboratory, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Sunitha Takalkar
- Clinical Biomarkers Laboratory, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Ti-Cheng Chang
- Center for Applied Bioinformatics, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Xiaotu Ma
- Department of Computational Biology, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Karol Szlachta
- Center for Applied Bioinformatics, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Ke Xu
- Center for Applied Bioinformatics, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Zhongshan Cheng
- Center for Applied Bioinformatics, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Yawei Hui
- Center for Applied Bioinformatics, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Selene C Koo
- Department of Pathology, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Paul E Mead
- Department of Pathology, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Qingsong Gao
- Department of Pathology, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Priyadarshini Kumar
- Department of Pathology, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Colin P Bailey
- Department of Pathology, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Jobin Sunny
- Department of Computational Biology, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Alberto S Pappo
- Department of Oncology, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Sara M Federico
- Department of Oncology, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Giles W Robinson
- Department of Oncology, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Amar Gajjar
- Department of Oncology, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Jeffrey E Rubnitz
- Department of Oncology, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Sima Jeha
- Department of Oncology, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Ching-Hon Pui
- Department of Oncology, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Hiroto Inaba
- Department of Oncology, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Gang Wu
- Center for Applied Bioinformatics, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Jeffery M Klco
- Department of Pathology, St. Jude Children's Research Hospital, Memphis, TN, USA.
- Center of Excellence for Leukemia Studies, St. Jude Children's Research Hospital, Memphis, TN, USA.
| | - Ruth G Tatevossian
- Clinical Biomarkers Laboratory, St. Jude Children's Research Hospital, Memphis, TN, USA.
| | - Charles G Mullighan
- Department of Pathology, St. Jude Children's Research Hospital, Memphis, TN, USA.
- Center of Excellence for Leukemia Studies, St. Jude Children's Research Hospital, Memphis, TN, USA.
| |
Collapse
|
19
|
Weiss NS. Gauging the efficacy of multicancer screening: the road ahead may be long and bumpy. J Natl Cancer Inst 2025; 117:212-213. [PMID: 39607987 PMCID: PMC11807428 DOI: 10.1093/jnci/djae267] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2024] [Accepted: 10/17/2024] [Indexed: 11/30/2024] Open
Affiliation(s)
- Noel S Weiss
- Department of Epidemiology, University of Washington, Seattle, WA 98195, United States
- Fred Hutchinson Cancer Center, Seattle, WA 98109, United States
| |
Collapse
|
20
|
Nadauld L, McDonnell CH, Dilaveri CA, Klein EA, Reid R, Marinac CR, Chung KC, Lopatin M, Fung ET, Schrag D, Patrick DL. Psychosocial impact associated with a multicancer early detection test (PATHFINDER): a prospective, multicentre, cohort study. Lancet Oncol 2025; 26:165-174. [PMID: 39818231 DOI: 10.1016/s1470-2045(24)00645-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2024] [Revised: 10/28/2024] [Accepted: 10/31/2024] [Indexed: 01/18/2025]
Abstract
BACKGROUND PATHFINDER was a prospective cohort study of multicancer early detection (MCED) testing in an outpatient ambulatory population. The aim of this study is to report the patient-reported outcomes (PROs) collected as secondary and exploratory measures in the PATHFINDER study. METHODS PATHFINDER is a prospective, multicentre, cohort study that enrolled existing healthy ambulatory outpatients at seven health networks in the USA, including hospitals, academic medical centres, and integrated health systems. Enrolled adults were aged 50 years or older without clinical suspicion of cancer, with or without additional cancer risk factors (smoking history, genetic predisposition, or previous cancer diagnosis). The primary objective was time to diagnostic resolution after an MCED cancer signal detected (CSD) result and extent of testing pursued. The objectives of the 12-month PATHFINDER study reported here were assessment of patient-reported outcomes and perceptions with MCED testing (the effect of the MCED test result disclosure, general anxiety symptoms, health-related quality of life, and satisfaction with the MCED test). PRO instruments used included an adapted Multidimensional Impact of Cancer Risk Assessment (MICRA) for distress, uncertainty, and positive experience at MCED test result disclosure; PRO Measurement Information System (PROMIS) Anxiety short-form for anxiety symptoms; and Short Form 12-Item Health Survey (SF-12v2) for health-related quality of life. Intentions towards adherence to guideline recommended screening was also assessed as an exploratory objective. This study is registered at ClinicalTrials.gov, NCT04241796, and is complete. FINDINGS Between Dec 12, 2019, and Dec 4, 2020, 6662 participants were recruited and 6621 eligible participants had analysable MCED test results (n=92 CSD and n=6529 no CSD [NCSD]). The majority of participants were women (4204 [63·5%] of 6621) and White (6071 [91·7%] of 6621). For participants who completed the MICRA at results disclosure, the mean total MICRA score was 28·4 (SD 14·9) for the 50 patients with a CSD result and 8·8 (7·2) for those with an NCSD result (n=5864 completed the full questionnaire). Mean general anxiety scores increased in true-positive and false-positive groups at results disclosure. The PROMIS anxiety true-positive group baseline score of 46·2 (SD 6·5; n=35) increased to 48·4 (7·3; n=19) and the scores in the false-positive group increased from 47·3 (7·3; n=52) to 49·7 (7·7; n=30). Mean scores in both groups returned towards baseline by end of study (true positive 46·8, SD 8·0; n=28; false positive 46·9, 8·1; n=41). Mean SF-12v2 mental component summary and scale scores were within the average general population range at all timepoints. A high proportion of participants (5749 [97·1%] of 5920) responded they were "satisfied", "very satisfied", or "extremely satisfied" with the MCED test, with this proportion highest in those with NCSD (5698 [97·2%] of 5861), followed by those with a true-positive MCED result (23 [92·0%] of 25) and those with a false-positive result (28 [82·4%] of 34). Most participants indicated they were likely or very likely to adhere to health-care providers' future cancer screening recommendations at end of study (5182 [95·6%] of 5418). INTERPRETATION The negative patient-reported effects associated with a CSD result from MCED testing were small and returned to baseline within 12 months for participants with or without a cancer diagnosis. PATHFINDER results indicate potential clinical benefit of early cancer detection and minimal patient distress associated with MCED testing. FUNDING GRAIL.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | - Deborah Schrag
- Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Donald L Patrick
- Health Systems and Population Health, University of Washington, Seattle, WA, USA.
| |
Collapse
|
21
|
Emery J. The psychological impact of screening with a multicancer early detection test. Lancet Oncol 2025; 26:148-149. [PMID: 39818232 DOI: 10.1016/s1470-2045(24)00667-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2024] [Accepted: 11/14/2024] [Indexed: 01/18/2025]
Affiliation(s)
- Jon Emery
- Faculty of Medicine, Dentistry and Health Sciences, University of Melbourne, Victorian Comprehensive Cancer Centre, Melbourne, VIC, Australia.
| |
Collapse
|
22
|
Witzig TE, Taylor WR, Mahoney DW, Bamlet WR, Foote PH, Burger KN, Doering KA, Devens ME, Arndt JR, O'Connell MC, Berger CK, Novak AJ, Cerhan JR, Hennek J, Katerov S, Allawi HT, Jevremovic D, Dao LN, Graham RP, Kisiel JB. Blood Plasma Methylated DNA Markers in the Detection of Lymphoma: Discovery, Validation, and Clinical Pilot. Am J Hematol 2025; 100:218-228. [PMID: 39564730 PMCID: PMC11705204 DOI: 10.1002/ajh.27533] [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: 10/22/2024] [Revised: 10/30/2024] [Accepted: 11/04/2024] [Indexed: 11/21/2024]
Abstract
Lymphoma is one of the leading causes of cancer and cancer deaths and yet has not been amenable to population screening. The role of methylated DNA markers (MDMs) in the detection of lymphoma has not been extensively studied. We aimed to discover, validate, and test tissue-derived MDMs of lymphoma in archival plasma specimens. Reduced representation bisulfite sequencing (RRBS) was performed on a discovery set of frozen tissues. MDMs identified were converted to methylation-specific PCR assays and validated on independent formalin-fixed, paraffin-embedded (FFPE) tissues. Target enrichment long-probe quantitative-amplified signal (TELQAS) assays were developed and assayed in plasma-extracted, bisulfite-converted DNA from independent treatment-naïve lymphoma patients and healthy controls. Prediction of cancer status was modeled using random forest model with in silico cross-validation. After discovery and validation in tissue, 16 TELQAS assays (ZNF503, VWA5B1, HOXA9, GABRG3, ITGA5, MAX.chr17.7190, BNC1, CDK20, MAX.chr4.4069, TPBG, DNAH14, SYT6, CACNG8, FAM110B, ADRA1D, and NRN1) were selected for testing in plasma. These detected 78% (95% CI, 74%-82%) of lymphoma cases at 90% specificity. Excluding marginal zone and T-cell lymphomas, sensitivity increased to 84% (80%-88%). MDMs in plasma show promise to detect lymphoma and are candidates for inclusion in multi-cancer detection studies.
Collapse
Affiliation(s)
| | - William R. Taylor
- Division of Gastroenterology and HepatologyMayo ClinicRochesterMinnesotaUSA
| | - Douglas W. Mahoney
- Department of Qualitative Health SciencesMayo ClinicRochesterMinnesotaUSA
| | - William R. Bamlet
- Department of Qualitative Health SciencesMayo ClinicRochesterMinnesotaUSA
| | - Patrick H. Foote
- Division of Gastroenterology and HepatologyMayo ClinicRochesterMinnesotaUSA
| | - Kelli N. Burger
- Department of Qualitative Health SciencesMayo ClinicRochesterMinnesotaUSA
| | - Karen A. Doering
- Division of Gastroenterology and HepatologyMayo ClinicRochesterMinnesotaUSA
| | - Mary E. Devens
- Division of Gastroenterology and HepatologyMayo ClinicRochesterMinnesotaUSA
| | - Jacquelyn R. Arndt
- Division of Gastroenterology and HepatologyMayo ClinicRochesterMinnesotaUSA
| | - Maria C. O'Connell
- Division of Gastroenterology and HepatologyMayo ClinicRochesterMinnesotaUSA
| | - Calise K. Berger
- Division of Gastroenterology and HepatologyMayo ClinicRochesterMinnesotaUSA
| | - Anne J. Novak
- Division of HematologyMayo ClinicRochesterMinnesotaUSA
| | - James R. Cerhan
- Department of Qualitative Health SciencesMayo ClinicRochesterMinnesotaUSA
| | | | - Slava Katerov
- Exact Sciences Development CompanyMadisonWisconsinUSA
| | | | | | - Linda N. Dao
- Division of Anatomic PathologyMayo ClinicRochesterMinnesotaUSA
| | | | - John B. Kisiel
- Division of Gastroenterology and HepatologyMayo ClinicRochesterMinnesotaUSA
| |
Collapse
|
23
|
Imai M, Nakamura Y, Yoshino T. Transforming cancer screening: the potential of multi-cancer early detection (MCED) technologies. Int J Clin Oncol 2025; 30:180-193. [PMID: 39799530 PMCID: PMC11785667 DOI: 10.1007/s10147-025-02694-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2024] [Accepted: 01/04/2025] [Indexed: 01/15/2025]
Abstract
Early cancer detection substantially improves the rate of patient survival; however, conventional screening methods are directed at single anatomical sites and focus primarily on a limited number of cancers, such as gastric, colorectal, lung, breast, and cervical cancer. Additionally, several cancers are inadequately screened, hindering early detection of 45.5% cases. In contrast, Multi-Cancer Early Detection (MCED) assays offer simultaneous screening of multiple cancers from a single liquid biopsy and identify molecular changes before symptom onset. These tests assess DNA mutations, abnormal DNA methylation patterns, fragmented DNA, and other tumor-derived biomarkers, indicating the presence of cancer and predicting its origin. Moreover, MCED assays concurrently detect multiple cancers without recommended screening protocols, potentially revolutionizing cancer screening and management. Large trials have reported promising results, achieving 50-95% sensitivity and 89-99% specificity for multiple cancer types. However, challenges, regarding improving accuracy, addressing ethical issues (e.g., psychosocial impact assessment), and integrating MCED into healthcare systems, must be addressed to achieve widespread adoption. Furthermore, prospective multi-institutional studies are crucial for demonstrating the clinical benefits in diverse populations. This review provides an overview of the principles, development status, and clinical significance of MCED tests, and discusses their potential and challenges.
Collapse
Affiliation(s)
- Mitsuho Imai
- Translational Research Support Section, National Cancer Center Hospital East, Chiba, Japan
- Department of Genetic Medicine and Services, National Cancer Center Hospital East, Chiba, Japan
| | - Yoshiaki Nakamura
- Translational Research Support Section, National Cancer Center Hospital East, Chiba, Japan
- Department of Gastroenterology and Gastrointestinal Oncology, National Cancer Center Hospital East, 6-5-1 Kashiwanoha, Kashiwa, Chiba, 277-8577, Japan
| | - Takayuki Yoshino
- Translational Research Support Section, National Cancer Center Hospital East, Chiba, Japan.
- Department of Gastroenterology and Gastrointestinal Oncology, National Cancer Center Hospital East, 6-5-1 Kashiwanoha, Kashiwa, Chiba, 277-8577, Japan.
| |
Collapse
|
24
|
Grady WM. Are Non-invasive Multi-cancer Early Cancer Detection Tests the Future? Dig Dis Sci 2025:10.1007/s10620-024-08839-2. [PMID: 39885052 DOI: 10.1007/s10620-024-08839-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/15/2024] [Accepted: 12/27/2024] [Indexed: 02/01/2025]
Abstract
Current cancer screening methods are effective for detecting early stage cancers and even preventing some cancers, but their effectiveness has only been demonstrated for a handful of cancers, and for many cancers, there are no screening tests clinically available. In addition, the majority of the screening methods are not ideal, resulting in suboptimal compliance and the occurrence of preventable cancers. A screening test that is convenient, safe, accurate and that can screen for multiple cancers is an ideal screening test that would address many of the shortcomings of the current tests. Multi-cancer detection tests (MCD) have the potential to meet these challenges and have engendered substantial enthusiasm in light of this. Using advances in DNA sequencing technology, cancer epigenetics and artificial intelligence, they are able to detect a large number of cancers predominantly via the patterns of methylated DNA alterations, DNA sequence alterations, and DNA fragment patterns of cell free DNA in the plasma and can accurately distinguish the cancer site of origin. Of note, some of the tests also combine circulating free DNA (cfDNA) with protein-based markers. However, for the majority of early stage cancers, the sensitivity is modest and below that of most of the current standard of care cancer screening tests. Furthermore, the clinical utility of screening for many of the cancers detectable by MCD tests remains to be proven. Here we describe the features of MCD tests, review the current data supporting their potential to be used in the clinic for cancer screening, and discuss the knowledge gaps surrounding understanding their clinical utility, with a focus on GI cancer screening.
Collapse
Affiliation(s)
- William M Grady
- Gastroenterology, University of Washington School of Medicine, Seattle, USA.
- Translational Science and Therapeutics Division, Fred Hutchinson Cancer Center, Seattle, WA, USA.
- Public Health Sciences Division, Fred Hutchinson Cancer Center, 1100 Fairview Ave N., D4-110, Seattle, WA, 98109, USA.
| |
Collapse
|
25
|
Gilman EA, Ghosh AK, Seegmiller S, Croghan IT, Hurt RT. Seminoma Diagnosed by Multicancer Detection Testing: A Case Report. SAGE Open Med Case Rep 2025; 13:2050313X251316987. [PMID: 39872676 PMCID: PMC11770699 DOI: 10.1177/2050313x251316987] [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: 10/08/2024] [Accepted: 01/14/2025] [Indexed: 01/30/2025] Open
Abstract
A 67-year-old male with positive signals for neuroendocrine and head/neck cancer on a commercially available multicancer detection test underwent an extensive negative diagnostic workup at a local hospital. Referred to our medical center for advanced imaging and endoscopic procedures ultimately led to the diagnosis of an early-stage seminoma with eventual orchiectomy. This case highlights challenges in evaluating positive multicancer detection results, emphasizing the need for a broader understanding of how to interpret these testing results to fully evaluate complex cases, as well as the need for protocolized follow-up approaches to a positive multicancer detection test.
Collapse
Affiliation(s)
- Elizabeth A. Gilman
- Division of General Internal Medicine, Department of Medicine, Mayo Clinic, Rochester, MN, USA
| | - Aditya K. Ghosh
- Division of General Internal Medicine, Department of Medicine, Mayo Clinic, Rochester, MN, USA
| | - Sara Seegmiller
- Division of General Internal Medicine, Department of Medicine, Mayo Clinic, Rochester, MN, USA
| | - Ivana T. Croghan
- Division of General Internal Medicine, Department of Medicine, Mayo Clinic, Rochester, MN, USA
| | - Ryan T. Hurt
- Division of General Internal Medicine, Department of Medicine, Mayo Clinic, Rochester, MN, USA
| |
Collapse
|
26
|
Chatanaka MK, Diamandis EP. What does cancer screening have to do with tomato growing? Clin Chem Lab Med 2025:cclm-2024-1408. [PMID: 39819368 DOI: 10.1515/cclm-2024-1408] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2024] [Accepted: 01/07/2025] [Indexed: 01/19/2025]
Abstract
Cancer screening is considered to be a major strategy for combatting cancer. The United States Preventive Services Task Force (USPSTF) recommends screening for five cancers, but the strength of evidence about the effectiveness of screening is limited. To gain insights into the efficacy of early detection requires prospective, blinded, placebo-controlled clinical trials with decades of follow-up and inclusion of millions of participants. Recently, Bretthauer et al. estimated lifetime gained with cancer screening tests by using a meta-analysis of 18 large randomized clinical trials which included more than two million subjects. They asked if cancer screening tests are saving lives and how much life is extended due to commonly used cancer screening tests. Colorectal cancer screening with sigmoidoscopy prolonged lifetime by 110 days, while fecal testing and mammography screening did not prolong life. A modest extension of 37 days was noted for prostate cancer screening with prostate-specific antigen testing and 107 days with lung cancer screening using computed tomography, but these estimates were not statistically significant. The authors concluded that current cancer screening strategies do not significantly prolong life. Based on these data, and the known biological behavior of some cancers, we hypothesized that the current strategies of treating cancer, after detection, could be modified to avoid the side effects of screening, which is a major determinant of the patient's overall survival.
Collapse
Affiliation(s)
- Miyo K Chatanaka
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, Canada
| | - Eleftherios P Diamandis
- Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Sinai Health System, Toronto, Canada
| |
Collapse
|
27
|
Vavoulis DV, Cutts A, Thota N, Brown J, Sugar R, Rueda A, Ardalan A, Howard K, Matos Santo F, Sannasiddappa T, Miller B, Ash S, Liu Y, Song CX, Nicholson BD, Dreau H, Tregidgo C, Schuh A. Multimodal cell-free DNA whole-genome TAPS is sensitive and reveals specific cancer signals. Nat Commun 2025; 16:430. [PMID: 39779727 PMCID: PMC11711490 DOI: 10.1038/s41467-024-55428-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2024] [Accepted: 12/05/2024] [Indexed: 01/11/2025] Open
Abstract
The analysis of circulating tumour DNA (ctDNA) through minimally invasive liquid biopsies is promising for early multi-cancer detection and monitoring minimal residual disease. Most existing methods focus on targeted deep sequencing, but few integrate multiple data modalities. Here, we develop a methodology for ctDNA detection using deep (80x) whole-genome TET-Assisted Pyridine Borane Sequencing (TAPS), a less destructive approach than bisulphite sequencing, which permits the simultaneous analysis of genomic and methylomic data. We conduct a diagnostic accuracy study across multiple cancer types in symptomatic patients, achieving 94.9% sensitivity and 88.8% specificity. Matched tumour biopsies are used for validation, not for guiding the analysis, imitating an early detection scenario. Furthermore, in silico validation demonstrates strong discrimination (86% AUC) at ctDNA fractions as low as 0.7%. Additionally, we successfully track tumour burden and ctDNA shedding from precancerous lesions post-treatment without requiring matched tumour biopsies. This pipeline is ready for further clinical evaluation to extend cancer screening and improve patient triage and monitoring.
Collapse
Affiliation(s)
- Dimitrios V Vavoulis
- Oxford Molecular Diagnostics Centre, Department of Oncology, University of Oxford, Oxford, UK.
- Biomedical Research Centre, Centre for Human Genetics, University of Oxford, Oxford, UK.
| | - Anthony Cutts
- Oxford Molecular Diagnostics Centre, Department of Oncology, University of Oxford, Oxford, UK
| | - Nishita Thota
- Exact Sciences Innovation LTD, The Sherard Bldg, Edmund Halley Rd, Littlemore, Oxford, UK
| | - Jordan Brown
- Exact Sciences Innovation LTD, The Sherard Bldg, Edmund Halley Rd, Littlemore, Oxford, UK
| | - Robert Sugar
- Exact Sciences Innovation LTD, The Sherard Bldg, Edmund Halley Rd, Littlemore, Oxford, UK
| | - Antonio Rueda
- Exact Sciences Innovation LTD, The Sherard Bldg, Edmund Halley Rd, Littlemore, Oxford, UK
| | - Arman Ardalan
- Oxford Molecular Diagnostics Centre, Department of Oncology, University of Oxford, Oxford, UK
| | - Kieran Howard
- Oxford Molecular Diagnostics Centre, Department of Oncology, University of Oxford, Oxford, UK
| | - Flavia Matos Santo
- Oxford Molecular Diagnostics Centre, Department of Oncology, University of Oxford, Oxford, UK
| | - Thippesh Sannasiddappa
- Exact Sciences Innovation LTD, The Sherard Bldg, Edmund Halley Rd, Littlemore, Oxford, UK
| | - Bronwen Miller
- Exact Sciences Innovation LTD, The Sherard Bldg, Edmund Halley Rd, Littlemore, Oxford, UK
| | - Stephen Ash
- Ludwig Institute for Cancer Research, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Yibin Liu
- College of Chemistry and Molecular Sciences, Wuhan University, Wuhan, China
- Taikang Centre for Life and Medical Sciences, Wuhan University, Wuhan, China
| | - Chun-Xiao Song
- Ludwig Institute for Cancer Research, Nuffield Department of Medicine, University of Oxford, Oxford, UK
- Target Discovery Institute, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Brian D Nicholson
- Nuffield Department of Primary Care Health Sciences, University of Oxford, Oxford, UK
| | - Helene Dreau
- Oxford Molecular Diagnostics Centre, Department of Oncology, University of Oxford, Oxford, UK
| | - Carolyn Tregidgo
- Exact Sciences Innovation LTD, The Sherard Bldg, Edmund Halley Rd, Littlemore, Oxford, UK
| | - Anna Schuh
- Oxford Molecular Diagnostics Centre, Department of Oncology, University of Oxford, Oxford, UK.
| |
Collapse
|
28
|
Wade R, Nevitt S, Liu Y, Harden M, Khouja C, Raine G, Churchill R, Dias S. Multi-cancer early detection tests for general population screening: a systematic literature review. Health Technol Assess 2025; 29:1-105. [PMID: 39898371 PMCID: PMC11808444 DOI: 10.3310/dlmt1294] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2025] Open
Abstract
Background General population cancer screening in the United Kingdom is limited to selected cancers. Blood-based multi-cancer early detection tests aim to detect potential cancer signals from multiple cancers in the blood. The use of a multi-cancer early detection test for population screening requires a high specificity and a reasonable sensitivity to detect early-stage disease so that the benefits of earlier diagnosis and treatment can be realised. Objective To undertake a systematic literature review of the clinical effectiveness evidence on blood-based multi-cancer early detection tests for screening. Methods Comprehensive searches of electronic databases (including MEDLINE and EMBASE) and trial registers were undertaken in September 2023 to identify published and unpublished studies of multi-cancer early detection tests. Test manufacturer websites and reference lists of included studies and pertinent reviews were checked for additional studies. The target population was individuals aged 50-79 years without clinical suspicion of cancer. Outcomes of interest included test accuracy, number and proportion of cancers detected (by site and stage), time to diagnostic resolution, mortality, potential harms, health-related quality of life, acceptability and satisfaction. The risk of bias was assessed using the quality assessment of diagnostic accuracy studies-2 checklist. Results were summarised using narrative synthesis. Stakeholders contributed to protocol development, report drafting and interpretation of review findings. Results Over 8000 records were identified. Thirty-six studies met the inclusion criteria: 1 ongoing randomised controlled trial, 13 completed cohort studies, 17 completed case-control studies and 5 ongoing cohort or case-control studies. Individual tests claimed to detect from 3 to over 50 different types of cancer. Diagnostic accuracy of currently available multi-cancer early detection tests varied substantially: Galleri® (GRAIL, Menlo Park, CA, USA) sensitivity 20.8-66.3%, specificity 98.4-99.5% (three studies); CancerSEEK (Exact Sciences, Madison, WI, USA) sensitivity 27.1-62.3%, specificity 98.9- 99.1% (two studies); SPOT-MAS™ (Gene Solutions, Ho Chi Minh City, Vietnam) sensitivity 72.4-100%, specificity 97.0-99.9% (two studies); Trucheck™ (Datar Cancer Genetics, Bayreuth, Germany) sensitivity 90.0%, specificity 96.4% (one study); Cancer Differentiation Analysis (AnPac Bio, Shanghai, China) sensitivity 40.0%, specificity 97.6% (one study). AICS® (AminoIndex Cancer Screening; Ajinomoto, Tokyo, Japan) screens for individual cancers separately, so no overall test performance statistics are available. Where reported, sensitivity was lower for detecting earlier-stage cancers (stages I-II) compared with later-stage cancers (stages III-IV). Studies of seven other multi-cancer early detection tests at an unclear stage of development were also summarised. Limitations Study selection was complex; it was often difficult to determine the stage of development of multi-cancer early detection tests. The evidence was limited; there were no completed randomised controlled trials and most included studies had a high overall risk of bias, primarily owing to limited follow-up of participants with negative test results. Only one study of Galleri recruited asymptomatic individuals aged over 50 in the United States of America; however, study results may not be representative of the United Kingdom's general screening population. No meaningful results were reported relating to patient-relevant outcomes, such as mortality, potential harms, health-related quality of life, acceptability or satisfaction. Conclusions All currently available multi-cancer early-detection tests reported high specificity (> 96%). Sensitivity was highly variable and influenced by study design, population, reference standard test used and length of follow-up. Future work Further research should report patient-relevant outcomes and consider patient and service impacts. Study registration This study is registered as PROSPERO CRD42023467901. Funding This award was funded by the National Institute for Health and Care Research (NIHR) Health Technology Assessment programme (NIHR award ref: NIHR161758) and is published in full in Health Technology Assessment; Vol. 29, No. 2. See the NIHR Funding and Awards website for further award information.
Collapse
Affiliation(s)
- Ros Wade
- Centre for Reviews and Dissemination, University of York, York, UK
| | - Sarah Nevitt
- Centre for Reviews and Dissemination, University of York, York, UK
| | - Yiwen Liu
- Centre for Reviews and Dissemination, University of York, York, UK
| | - Melissa Harden
- Centre for Reviews and Dissemination, University of York, York, UK
| | - Claire Khouja
- Centre for Reviews and Dissemination, University of York, York, UK
| | - Gary Raine
- Centre for Reviews and Dissemination, University of York, York, UK
| | - Rachel Churchill
- Centre for Reviews and Dissemination, University of York, York, UK
| | - Sofia Dias
- Centre for Reviews and Dissemination, University of York, York, UK
| |
Collapse
|
29
|
Kang SK, Gulati R, Moise N, Hur C, Elkin EB. Multi-Cancer Early Detection Tests: State of the Art and Implications for Radiologists. Radiology 2025; 314:e233448. [PMID: 39807974 PMCID: PMC11783158 DOI: 10.1148/radiol.233448] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2023] [Revised: 03/21/2024] [Accepted: 04/25/2024] [Indexed: 01/16/2025]
Abstract
Multi-cancer early detection (MCED) tests are already being marketed as noninvasive, convenient opportunities to test for multiple cancer types with a single blood sample. The technology varies-involving detection of circulating tumor DNA, fragments of DNA, RNA, or proteins unique to each targeted cancer. The priorities and tradeoffs of reaching diagnostic resolution in the setting of possible false positives and negatives remain under active study. Given the well-established role of imaging in lesion detection and characterization for most cancers, radiologists have an essential role to play in selecting diagnostic pathways, determining the validity of test results, resolving false-positive MCED test results, and evaluating tradeoffs for clinical policy. Appropriate access to and use of imaging tests will also factor into clinical guidelines. Thus, all clinicians potentially involved with MCED tests for cancer screening will need to weigh the benefits and harms of MCED testing, including consideration of how the tests will be used alongside or in place of other screening options, how diagnostic confirmation tests should be selected, and what the implications are for policy and reimbursement decisions. Further, patients will need regular support to make informed decisions about screening using MCED tests in the context of their personal cancer risks, health-related values, and access to care.
Collapse
Affiliation(s)
| | - Roman Gulati
- From the Departments of Radiology and Population Health, New York
University Langone Medical Center, New York, NY (S.K.K.); Division of Public
Health Sciences, Fred Hutchinson Cancer Center, Seattle, Wash (R.G.); Department
of Medicine, Vagelos College of Physicians and Surgeons, Columbia University,
New York, NY (N.M., C.H.); Herbert Irving Comprehensive Cancer Center, New York,
NY (C.H., E.B.E.); and Department of Health Policy and Management, Mailman
School of Public Health, Columbia University, New York, NY (E.B.E.)
| | - Nathalie Moise
- From the Departments of Radiology and Population Health, New York
University Langone Medical Center, New York, NY (S.K.K.); Division of Public
Health Sciences, Fred Hutchinson Cancer Center, Seattle, Wash (R.G.); Department
of Medicine, Vagelos College of Physicians and Surgeons, Columbia University,
New York, NY (N.M., C.H.); Herbert Irving Comprehensive Cancer Center, New York,
NY (C.H., E.B.E.); and Department of Health Policy and Management, Mailman
School of Public Health, Columbia University, New York, NY (E.B.E.)
| | - Chin Hur
- From the Departments of Radiology and Population Health, New York
University Langone Medical Center, New York, NY (S.K.K.); Division of Public
Health Sciences, Fred Hutchinson Cancer Center, Seattle, Wash (R.G.); Department
of Medicine, Vagelos College of Physicians and Surgeons, Columbia University,
New York, NY (N.M., C.H.); Herbert Irving Comprehensive Cancer Center, New York,
NY (C.H., E.B.E.); and Department of Health Policy and Management, Mailman
School of Public Health, Columbia University, New York, NY (E.B.E.)
| | - Elena B. Elkin
- From the Departments of Radiology and Population Health, New York
University Langone Medical Center, New York, NY (S.K.K.); Division of Public
Health Sciences, Fred Hutchinson Cancer Center, Seattle, Wash (R.G.); Department
of Medicine, Vagelos College of Physicians and Surgeons, Columbia University,
New York, NY (N.M., C.H.); Herbert Irving Comprehensive Cancer Center, New York,
NY (C.H., E.B.E.); and Department of Health Policy and Management, Mailman
School of Public Health, Columbia University, New York, NY (E.B.E.)
| |
Collapse
|
30
|
Pantel K, Alix-Panabières C. Minimal residual disease as a target for liquid biopsy in patients with solid tumours. Nat Rev Clin Oncol 2025; 22:65-77. [PMID: 39609625 DOI: 10.1038/s41571-024-00967-y] [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: 11/01/2024] [Indexed: 11/30/2024]
Abstract
Metastasis is the leading cause of cancer-related death in patients with solid tumours. Current imaging technologies are not sufficiently sensitive to detect minimal residual disease (MRD; also known as measurable or molecular residual disease) after initial surgery or chemotherapy, pointing to the need for more sensitive tests to detect remaining traces of cancer in the body. Liquid biopsy, or the analysis of tumour-derived or tumour-induced cells or cellular products in the blood or other body fluids, has opened a new diagnostic avenue to detect and monitor MRD. Liquid biopsy is already used in clinical decision making for patients with haematological malignancies. Here, we review current knowledge on the use of circulating tumour DNA (ctDNA) to detect and monitor MRD in patients with solid tumours. We also discuss how ctDNA-guided MRD detection and characterization could herald a new era of novel 'post-adjuvant therapies' with the potential to eliminate MRD and cure patients before terminal metastatic disease is evident on imaging.
Collapse
Affiliation(s)
- Klaus Pantel
- Department of Tumour Biology, University Medical, Center Hamburg-Eppendorf, Hamburg, Germany.
- European Liquid Biopsy Society (ELBS), Hamburg, Germany.
| | - Catherine Alix-Panabières
- European Liquid Biopsy Society (ELBS), Hamburg, Germany.
- Laboratory of Rare Human Circulating Cells (LCCRH) and Liquid Biopsy, University Medical Centre of Montpellier, Montpellier, France.
- CREEC (CREES), Unité Mixte de Recherches, IRD 224-CNRS 5290-Université de Montpellier, Montpellier, France.
| |
Collapse
|
31
|
Hurt RT, Ghosh AK, Dougan BM, Gilman EA, Salonen BR, Adusumalli J, Bonnes SL, Andersen CA, Pasha AS, Nanda S, Pagel EM, Verness CD, Crowley SD, Ressler SW, Samadder JJ, Presutti RJ, Chaudhuri AA, Sanchez W, Croghan IT, Stephenson CR, Ghosh K. Implementation of a Multicancer Detection (MCD) Test in a Tertiary Referral Center in Asymptomatic Patients: An 18-Month Prospective Cohort Study. J Prim Care Community Health 2025; 16:21501319251329290. [PMID: 40132152 PMCID: PMC11938867 DOI: 10.1177/21501319251329290] [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: 02/06/2025] [Revised: 02/24/2025] [Accepted: 03/05/2025] [Indexed: 03/27/2025] Open
Abstract
OBJECTIVE Multicancer Detection (MCD) tests, such as the GRAIL Galleri, offer a novel approach to cancer screening by detecting cancer-specific methylation patterns in cell-free DNA through a single blood sample. This study evaluated an 18-month implementation of MCD testing in a tertiary ambulatory internal medicine clinic. PATIENTS AND METHODS Between June 2022 and November 2023, 2244 asymptomatic (without symptoms attributed to cancer) patients underwent MCD testing. The study focused on operational workflows, patient and physician education, and diagnostic follow-up of positive results. Standardized materials, including electronic health record (EHR) workflows, FAQs, and diagnostic pathways, were developed to facilitate implementation. Challenges included managing false positives, patient anxiety, costs, and ethical considerations. RESULTS Of the 2244 patients tested, 17 (0.76%) had positive results, and 15 underwent further diagnostic evaluation. Cancer was confirmed in 11 (73.3%) patients, including cases of breast, colon, esophageal, lymphoma, ovarian, and pancreatic cancers. Four patients had no identifiable malignancy despite comprehensive work-up. CONCLUSIONS MCD testing is feasible in routine clinical workflows, with 73% of positive cases yielding cancer diagnoses. While promising, further research is required to assess long-term outcomes, cost-effectiveness, and optimal implementation strategies of cancer interception in broader healthcare settings.
Collapse
|
32
|
Geng S, Li S, Wu W, Chang Y, Mao M. A Cost-Effective Two-Step Approach for Multi-Cancer Early Detection in High-Risk Populations. CANCER RESEARCH COMMUNICATIONS 2025; 5:150-156. [PMID: 39740056 PMCID: PMC11758400 DOI: 10.1158/2767-9764.crc-24-0508] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/24/2024] [Revised: 12/07/2024] [Accepted: 12/26/2024] [Indexed: 01/02/2025]
Abstract
SIGNIFICANCE Large-scale screening inevitably leads to significant financial burdens on the healthcare system, which is a key factor constraining nationwide screenings. The two-step MCED approach not only maintains comparable performance but also substantially alleviates financial strains compared with the direct use of next-generation sequencing-based MCED tests for massive screenings.
Collapse
Affiliation(s)
| | - Shiyong Li
- Research & Development, SeekIn Inc, Shenzhen, China
| | - Wei Wu
- Research & Development, SeekIn Inc, Shenzhen, China
| | - Yinyin Chang
- Clinical Laboratories, Shenyou Bio, Zhengzhou, China
| | - Mao Mao
- Research & Development, SeekIn Inc, San Diego, California
- Yonsei Song-Dang Institute for Cancer Research, Yonsei University, Seoul, Republic of Korea
| |
Collapse
|
33
|
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
|
34
|
Jee J. Circulating tumor DNA: the dawn of new, clinically scalable biomarkers for thromboembolism. J Thromb Haemost 2025; 23:39-42. [PMID: 39798969 DOI: 10.1016/j.jtha.2024.09.031] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2024] [Accepted: 09/26/2024] [Indexed: 01/15/2025]
Affiliation(s)
- Justin Jee
- Department of Medicine, Memorial Sloan Kettering Cancer Center.
| |
Collapse
|
35
|
Hricak H, Mayerhoefer ME, Herrmann K, Lewis JS, Pomper MG, Hess CP, Riklund K, Scott AM, Weissleder R. Advances and challenges in precision imaging. Lancet Oncol 2025; 26:e34-e45. [PMID: 39756454 DOI: 10.1016/s1470-2045(24)00395-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2024] [Revised: 07/10/2024] [Accepted: 07/11/2024] [Indexed: 01/07/2025]
Abstract
Technological innovations in genomics and related fields have facilitated large sequencing efforts, supported new biological discoveries in cancer, and spawned an era of liquid biopsy biomarkers. Despite these advances, precision oncology has practical constraints, partly related to cancer's biological diversity and spatial and temporal complexity. Advanced imaging technologies are being developed to address some of the current limitations in early detection, treatment selection and planning, drug delivery, and therapeutic response, as well as difficulties posed by drug resistance, drug toxicity, disease monitoring, and metastatic evolution. We discuss key areas of advanced imaging for improving cancer outcomes and survival. Finally, we discuss practical challenges to the broader adoption of precision imaging in the clinic and the need for a robust translational infrastructure.
Collapse
Affiliation(s)
- Hedvig Hricak
- Department of Radiology and Molecular Pharmacology Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Marius E Mayerhoefer
- Department of Radiology, NYU Grossman School of Medicine, New York, NY, USA; Department of Biomedical Imaging and Image-guided Therapy, Medical University of Vienna, Vienna, Austria
| | - Ken Herrmann
- Department of Nuclear Medicine, University of Duisburg-Essen, Essen, Germany; German Cancer Consortium (DKTK), University Hospital Essen, Essen, Germany
| | - Jason S Lewis
- Department of Radiology and Molecular Pharmacology Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA; Department of Radiology and Department of Pharmacology, Weill Cornell Medical College, New York, NY, USA
| | - Martin G Pomper
- University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Christopher P Hess
- Department of Radiology and Biomedical Imaging, UCSF, San Francisco, CA, USA
| | - Katrine Riklund
- Department of Diagnostics and Intervention, Umeå University, Umeå, Sweden
| | - Andrew M Scott
- Department of Molecular Imaging and Therapy, Austin Health, Melbourne, VIC, Australia; Olivia Newton-John Cancer Research Institute, Melbourne, VIC, Australia; School of Cancer Medicine, La Trobe University, Melbourne, VIC, Australia; Faculty of Medicine, University of Melbourne, Melbourne, VIC, Australia
| | - Ralph Weissleder
- Department of Radiology and Center for Systems Biology, Massachusetts General Brigham, Boston, MA, USA; Department of Systems Biology, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
36
|
Dang Nguyen LH, Tieu BL, Nguyen TT, Ha NP, Huong Nguyen GT, Hanh Nguyen TH, Le VH, Bui VQ, Nguyen LH, Pham NH, Phan TH, Nguyen HT, Tran VS, Bui CV, Vo VK, Nhan Nguyen PT, Phuoc Dang HH, Pham VD, Cao VT, Phan NM, Nguyen VT, Quyen Le TL, Luong TLA, Phuong Doan TK, Phan CD, Nguyen TX, Pham NT, Nguyen BT, Thuy Pham TT, Le HL, Truong CT, Jasmine TX, Le MC, Phan VB, Truong QB, Ly Tran TH, Huynh MT, Tran TQ, Nguyen ST, Tran V, Tran VK, Nguyen HN, Phan TV, Do TTT, Truong DK, Giang H, Nguyen HN, Phan MD, Tran LS, Tang HS, Nguyen DS. A consultation and work-up diagnosis protocol for a multicancer early detection test: a case series study. Future Sci OA 2024; 10:2395244. [PMID: 39254097 PMCID: PMC11389743 DOI: 10.1080/20565623.2024.2395244] [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: 03/11/2024] [Accepted: 08/19/2024] [Indexed: 09/11/2024] Open
Abstract
The emergence of multicancer early detection (MCED) tests holds promise for improving early cancer detection and public health outcomes. However, positive MCED test results require confirmation through recommended cancer diagnostic imaging modalities. To address these challenges, we have developed a consultation and work-up protocol for definitive diagnostic results post MCED testing, named SPOT-MAS. Developed through circulating tumor DNA (ctDNA) analysis and in line with professional guidelines and advisory board consensus, this protocol standardizes information to aid general practitioners in accessing, interpreting and managing SPOT-MAS results. Clinical effectiveness is demonstrated through a series of identified cancer cases. Our research indicates that the protocol could empower healthcare professionals to confidently interpret circulating tumor DNA test results for 5 common types of cancer, thereby facilitating the clinical integration of MCED tests.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Van Hoi Le
- National Cancer Hospital, Hanoi, Vietnam
| | | | | | | | | | | | | | | | - Van Kha Vo
- Cantho Oncology Hospital, Can Tho, Vietnam
| | | | | | - Van Dung Pham
- Thong Nhat Dongnai General Hospital, Dong Nai, Vietnam
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | - Minh Chi Le
- University Medical Center HCM, Ho Chi Minh Vietnam
| | | | | | | | | | | | | | - Vu Tran
- Thong Nhat Dongnai General Hospital, Dong Nai, Vietnam
| | | | | | | | | | | | - Hoa Giang
- Gene Solutions, Ho Chi Minh, Vietnam
- Medical Genetics Institute, Ho Chi Minh, Vietnam
| | - Hoai-Nghia Nguyen
- Gene Solutions, Ho Chi Minh, Vietnam
- Medical Genetics Institute, Ho Chi Minh, Vietnam
| | - Minh-Duy Phan
- Gene Solutions, Ho Chi Minh, Vietnam
- Medical Genetics Institute, Ho Chi Minh, Vietnam
| | - Le Son Tran
- Gene Solutions, Ho Chi Minh, Vietnam
- Medical Genetics Institute, Ho Chi Minh, Vietnam
| | - Hung Sang Tang
- Gene Solutions, Ho Chi Minh, Vietnam
- Medical Genetics Institute, Ho Chi Minh, Vietnam
| | - Duy Sinh Nguyen
- Gene Solutions, Ho Chi Minh, Vietnam
- Medical Genetics Institute, Ho Chi Minh, Vietnam
| |
Collapse
|
37
|
Tabrizi S, Martin-Alonso C, Xiong K, Bhatia SN, Adalsteinsson VA, Love JC. Modulating cell-free DNA biology as the next frontier in liquid biopsies. Trends Cell Biol 2024:S0962-8924(24)00249-6. [PMID: 39730275 DOI: 10.1016/j.tcb.2024.11.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2024] [Revised: 11/05/2024] [Accepted: 11/20/2024] [Indexed: 12/29/2024]
Abstract
Technical advances over the past two decades have enabled robust detection of cell-free DNA (cfDNA) in biological samples. Yet, higher clinical sensitivity is required to realize the full potential of liquid biopsies. This opinion article argues that to overcome current limitations, the abundance of informative cfDNA molecules - such as circulating tumor DNA (ctDNA) - collected in a sample needs to increase. To accomplish this, new methods to modulate the biological processes that govern cfDNA production, trafficking, and clearance in the body are needed, informed by a deeper understanding of cfDNA biology. Successful development of such methods could enable a major leap in the performance of liquid biopsies and vastly expand their utility across the spectrum of clinical care.
Collapse
Affiliation(s)
- Shervin Tabrizi
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA; Broad Institute of MIT and Harvard, Cambridge, MA, USA; Department of Radiation Oncology, Mass General Brigham, Boston, MA, USA; Harvard Medical School, Boston, MA, USA.
| | - Carmen Martin-Alonso
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA; Harvard-MIT Division of Health Sciences and Technology, Institute for Medical Engineering and Science, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Kan Xiong
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Sangeeta N Bhatia
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA; Broad Institute of MIT and Harvard, Cambridge, MA, USA; Harvard-MIT Division of Health Sciences and Technology, Institute for Medical Engineering and Science, Massachusetts Institute of Technology, Cambridge, MA, USA; Department of Electrical Engineering and Computer Science, Massachusetts Institute of Technology, Cambridge, MA, USA; Wyss Institute at Harvard University, Boston, MA, USA; Howard Hughes Medical Institute, Cambridge, MA, USA
| | | | - J Christopher Love
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA; Broad Institute of MIT and Harvard, Cambridge, MA, USA; Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| |
Collapse
|
38
|
Gram EG, Copp T, Ransohoff DF, Plüddemann A, Kramer BS, Woloshin S, Shih P. Direct-to-consumer tests: emerging trends are cause for concern. BMJ 2024; 387:e080460. [PMID: 39631937 DOI: 10.1136/bmj-2024-080460] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/07/2024]
Affiliation(s)
- Emma Grundtvig Gram
- Center for General Practice, Department of Public Health, University of Copenhagen, Copenhagen, Denmark
- Lisa Schwartz Foundation for Truth in Medicine, Norwich, VT, USA
| | - Tessa Copp
- Lisa Schwartz Foundation for Truth in Medicine, Norwich, VT, USA
- Faculty of Medicine and Health, School of Public Health, University of Sydney, Sydney, Australia
| | - David F Ransohoff
- Division of Gastroenterology and Hepatology, Departments of Medicine and Epidemiology, University of North Carolina, Chapel Hill, USA
| | - Annette Plüddemann
- Centre for Evidence Based Medicine, Nuffield Department of Primary Care Health Sciences, University of Oxford, Oxford, UK
| | - Barnett S Kramer
- Lisa Schwartz Foundation for Truth in Medicine, Norwich, VT, USA
| | - Steven Woloshin
- Lisa Schwartz Foundation for Truth in Medicine, Norwich, VT, USA
- Center for Medicine and Media, Dartmouth Institute, Lebanon, USA
| | - Patti Shih
- Australian Centre for Health Engagement Evidence and Values, School of Health and Society, University of Wollongong, Wollongong, Australia
| |
Collapse
|
39
|
Smith LM, Mahoney DW, Bamlet WR, Yu F, Liu S, Goggins MG, Darabi S, Majumder S, Wang QL, Coté GA, Demeure MJ, Zhang Z, Srivastava S, Chawla A, Izmirlian G, Olson JE, Wolpin BM, Genkinger JM, Zaret KS, Brand R, Koay EJ, Oberg AL. Early detection of pancreatic cancer: Study design and analytical considerations in biomarker discovery and early phase validation studies. Pancreatology 2024; 24:1265-1279. [PMID: 39516175 PMCID: PMC11780679 DOI: 10.1016/j.pan.2024.10.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/15/2024] [Revised: 10/05/2024] [Accepted: 10/27/2024] [Indexed: 11/16/2024]
Abstract
OBJECTIVES Pancreatic ductal adenocarcinoma (PDAC) is a highly lethal disease that is challenging to detect at an early stage. Biomarkers are needed that can detect PDAC early in the course of disease when interventions lead to the best outcomes. We highlight study design and statistical considerations that inform pancreatic cancer early detection biomarker evaluation. METHODS We describe experimental design strategies in this setting useful for streamlining biomarker evaluation at each Early Detection Research Network (EDRN) phase of biomarker development. We break the early EDRN phases into sub-phases, proposing objectives, study design strategies, and biomarker performance benchmarks. RESULTS The goal of early detection in populations at high-risk of PDAC is described. Evaluating biomarker behavior in patients under surveillance without disease can winnow candidate biomarkers. Potential resources for biomarker validation studies are described. CONCLUSIONS Multisite and multidisciplinary collaboration can facilitate study design strategies in this lethal but low incidence disease and streamline the path from biomarker discovery to clinical use. Improvements in analytical and experimental design methods could help accelerate biomarker evaluation through the phases of biomarker development.
Collapse
Affiliation(s)
- Lynette M Smith
- Department of Biostatistics, University of Nebraska Medical Center, Omaha, NE, USA
| | - Douglas W Mahoney
- Department of Quantitative Health Sciences, Mayo Clinic, Rochester, MN, USA
| | - William R Bamlet
- Department of Quantitative Health Sciences, Mayo Clinic, Rochester, MN, USA
| | - Fang Yu
- Department of Biostatistics, University of Nebraska Medical Center, Omaha, NE, USA
| | - Suyu Liu
- Department of Biostatistics, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Michael G Goggins
- Departments of Pathology and Oncology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Sourat Darabi
- Hoag Family Cancer Institute, Newport Beach, CA, USA
| | - Shounak Majumder
- Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, MN, USA
| | - Qiao-Li Wang
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | - Gregory A Coté
- Department of Medicine, Oregon Health & Science University, Portland, OR, USA
| | | | - Zhen Zhang
- Departments of Pathology and Oncology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | | | - Akhil Chawla
- Department of Surgery, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | | | - Janet E Olson
- Department of Quantitative Health Sciences, Mayo Clinic, Rochester, MN, USA
| | - Brian M Wolpin
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | - Jeanine M Genkinger
- Department of Epidemiology, Mailman School of Public Health, Columbia University, NY, NY, USA
| | - Kenneth S Zaret
- Department of Cell and Developmental Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Randall Brand
- Department of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | - Eugene J Koay
- Department of Gastrointestinal Radiation Oncology, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Ann L Oberg
- Department of Quantitative Health Sciences, Mayo Clinic, Rochester, MN, USA.
| |
Collapse
|
40
|
Karimzadeh M, Momen-Roknabadi A, Cavazos TB, Fang Y, Chen NC, Multhaup M, Yen J, Ku J, Wang J, Zhao X, Murzynowski P, Wang K, Hanna R, Huang A, Corti D, Nguyen D, Lam T, Kilinc S, Arensdorf P, Chau KH, Hartwig A, Fish L, Li H, Behsaz B, Elemento O, Zou J, Hormozdiari F, Alipanahi B, Goodarzi H. Deep generative AI models analyzing circulating orphan non-coding RNAs enable detection of early-stage lung cancer. Nat Commun 2024; 15:10090. [PMID: 39572521 PMCID: PMC11582319 DOI: 10.1038/s41467-024-53851-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2024] [Accepted: 10/22/2024] [Indexed: 11/24/2024] Open
Abstract
Liquid biopsies have the potential to revolutionize cancer care through non-invasive early detection of tumors. Developing a robust liquid biopsy test requires collecting high-dimensional data from a large number of blood samples across heterogeneous groups of patients. We propose that the generative capability of variational auto-encoders enables learning a robust and generalizable signature of blood-based biomarkers. In this study, we analyze orphan non-coding RNAs (oncRNAs) from serum samples of 1050 individuals diagnosed with non-small cell lung cancer (NSCLC) at various stages, as well as sex-, age-, and BMI-matched controls. We demonstrate that our multi-task generative AI model, Orion, surpasses commonly used methods in both overall performance and generalizability to held-out datasets. Orion achieves an overall sensitivity of 94% (95% CI: 87%-98%) at 87% (95% CI: 81%-93%) specificity for cancer detection across all stages, outperforming the sensitivity of other methods on held-out validation datasets by more than ~ 30%.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | - Ti Lam
- Exai Bio Inc., Palo Alto, CA, US
| | | | | | | | | | | | - Helen Li
- Exai Bio Inc., Palo Alto, CA, US
| | | | | | - James Zou
- Stanford University, Stanford, CA, US
| | | | | | - Hani Goodarzi
- University of California, San Francisco, CA, US.
- Arc Institute, Palo Alto, CA, US.
| |
Collapse
|
41
|
Mannucci A, Goel A. Stool and blood biomarkers for colorectal cancer management: an update on screening and disease monitoring. Mol Cancer 2024; 23:259. [PMID: 39558327 PMCID: PMC11575410 DOI: 10.1186/s12943-024-02174-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2024] [Accepted: 11/07/2024] [Indexed: 11/20/2024] Open
Abstract
BACKGROUND Biomarkers have revolutionized the management of colorectal cancer (CRC), facilitating early detection, prevention, personalized treatment, and minimal residual disease (MRD) monitoring. This review explores current CRC screening strategies and emerging biomarker applications. MAIN BODY We summarize the landscape of non-invasive CRC screening and MRD detection strategies, discuss the limitations of the current approaches, and highlight the promising potential of novel biomarker solutions. The fecal immunochemical test remained the cornerstone of CRC screening, but its sensitivity has been improved by assays that combined its performance with other stool analytes. However, their sensitivity for advanced adenomas and the patient compliance both remain suboptimal. Blood-based tests promise to increase compliance but require further refinement to compete with stool-based biomarker tests. The ideal scenario involves leveraging blood tests to increase screening participation, and simultaneously promote stool- and endoscopy-based screening among those who are compliant. Once solely reliant on upfront surgery followed by stage and pathology-driven adjuvant chemotherapy, the treatment of stage II and III colon cancer has undergone a revolutionary transformation with the advent of MRD testing after surgery. A decade ago, the concept of using a post-surgical test instead of stage and pathology to determine the need for adjuvant chemotherapy was disruptive. Today, a blood test may be more informative of the need for chemotherapy than the stage at diagnosis. CONCLUSION Biomarker research is not just improving, but bringing a transformative change to CRC clinical management. Early detection is not just getting better, but improving thanks to a multi-modality approach, and personalized treatment plans are not just becoming a reality, but a promising future with MRD testing.
Collapse
Affiliation(s)
- Alessandro Mannucci
- Department of Molecular Diagnostics and Experimental Therapeutics, Beckman Research Institute at City of Hope, Monrovia, CA, USA
- Gastroenterology and Gastrointestinal Endoscopy Unit, Vita-Salute San Raffaele University, IRCCS San Raffaele Hospital, Milan, Italy
| | - Ajay Goel
- Department of Molecular Diagnostics and Experimental Therapeutics, Beckman Research Institute at City of Hope, Monrovia, CA, USA.
- City of Hope Comprehensive Cancer Center, Duarte, CA, USA.
| |
Collapse
|
42
|
Half E, Ovcharenko A, Shmuel R, Furman-Assaf S, Avdalimov M, Rabinowicz A, Arber N. Non-invasive multiple cancer screening using trained detection canines and artificial intelligence: a prospective double-blind study. Sci Rep 2024; 14:28204. [PMID: 39548246 PMCID: PMC11568277 DOI: 10.1038/s41598-024-79383-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2024] [Accepted: 11/08/2024] [Indexed: 11/17/2024] Open
Abstract
The specificity and sensitivity of a simple non-invasive multi-cancer screening method in detecting breast, lung, prostate, and colorectal cancer in breath samples were evaluated in a double-blind study. Breath samples of 1386 participants (59.7% males, median age 56.0 years) who underwent screening for cancer using gold-standard screening methods, or a biopsy for a suspected malignancy were collected. The samples were analyzed using a bio-hybrid platform comprising trained detection canines and artificial intelligence tools. According to cancer screening/biopsy results, 1048 (75.6%) were negative for cancer and 338 (24.4%) were positive. Among the 338 positive samples, 261 (77.2%) were positive for one of the four cancer types that the bio-hybrid platform was trained to detect, with an overall sensitivity and specificity of 93.9% (95% confidence interval [CI] 90.3-96.2%) and 94.3% (95% CI 92.7%-95.5%), respectively. The sensitivity of each cancer type was similar; breast: 95.0% (95% CI 87.8-98.0%), lung: 95.0% (95% CI 87.8-98.0%), colorectal: 90.0% (95% CI 74.4-96.5%), prostate: 93.0% (95% CI 84.6-97.0%). The sensitivity of 14 other malignant tumors that the bio-hybrid platform was not trained to detect, but identified, was 81.8% (95% CI 71.8%-88.8%). Early cancer (0-2) detection sensitivity was 94.8% (95% CI 91.0%-97.1%). This bio-hybrid multi-cancer screening platform demonstrated high sensitivity and specificity and enables early-stage cancer detection.
Collapse
Affiliation(s)
- Elizabeth Half
- Gastroenterology Unit, Rambam Health Care Campus, Haifa, Israel
| | | | - Ronit Shmuel
- Medical consultant (independent), Tel Aviv, Israel
| | | | | | | | - Nadir Arber
- Integrated Cancer Prevention Center, Tel Aviv Souraski Medical Center, Tel Aviv, Israel
- Faculty of Medical and Health Sciences, Tel Aviv University, Tel Aviv, Israel
| |
Collapse
|
43
|
Porav-Hodade D, Gherasim R, Loghin A, Lazar B, Cotoi OS, Badea MA, Ilona MOK, Todea-Moga C, Vartolomei MD, Rares G, Crisan N, Feciche OB. Bladder Adenocarcinoma in a Constellation of Multiple Site Malignancies: An Unusual Case and Systematic Review. Diagnostics (Basel) 2024; 14:2510. [PMID: 39594177 PMCID: PMC11592427 DOI: 10.3390/diagnostics14222510] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2024] [Revised: 11/05/2024] [Accepted: 11/08/2024] [Indexed: 11/28/2024] Open
Abstract
BACKGROUND AND OBJECTIVES Multiple primary malignant tumors represent a small percentage of the total number of oncological cases and can involve either metachronous or synchronous development and represent challenges in diagnosis, staging, and treatment planning. Our purpose is to present a rare case of bladder adenocarcinoma in a female patient with multiple primary malignant tumors and to provide systematic review of the available literature. MATERIALS AND METHODS A 67-year-old female patient was admitted with altered general condition and anuria. The past medical history of the patient included malignant melanoma (2014), cervical cancer (2017), colon cancer (2021), obstructive anuria (2023), and liver metastasectomy (2023). Transurethral resection of bladder tumor was performed for bladder tumors. RESULTS Contrast CT highlighted multiple pulmonary metastases, a poly nodular liver conglomerate, retroperitoneal lymph node, II/III grade left ureterohydronephrosis, and no digestive tract tumor masses. The pathological result of the bladder resection showed an infiltrative adenocarcinoma. CONCLUSIONS The difference between primary bladder adenocarcinoma tumor and metastatic colorectal adenocarcinoma is the key for the future therapeutic strategy. Identification and assessment of risk factors such as viral infection, radiotherapy, chemotherapy, smoking, and genetics are pivotal in understanding and managing multiple primary malignant tumors. Personalized prevention strategies and screening programs may facilitate the early detection of these tumors, whether synchronous or metachronous. The use of multicancer early detection (MCED) blood tests for early diagnosis appears promising. However, additional research is needed to standardize these techniques for cancer detection.
Collapse
Affiliation(s)
- Daniel Porav-Hodade
- Department of Urology, George Emil Palade University of Medicine, Pharmacy, Science, and Technology of Târgu Mureș, 540139 Târgu Mureș, Romania; (D.P.-H.); (M.O.K.I.)
- Department of Urology, Clinical County Hospital Mures, 540136 Târgu Mures, Romania;
| | - Raul Gherasim
- Department of Urology, Clinical County Hospital Mures, 540136 Târgu Mures, Romania;
| | - Andrada Loghin
- Department of Pathophysiology, George Emil Palade University of Medicine, Pharmacy, Science, and Technology of Târgu Mureș, 540139 Târgu Mureș, Romania; (A.L.); (O.S.C.)
- Department of Pathophysiology, Clinical County Hospital Mures, 540136 Târgu Mures, Romania;
| | - Bianca Lazar
- Department of Pathophysiology, Clinical County Hospital Mures, 540136 Târgu Mures, Romania;
| | - Ovidiu Simion Cotoi
- Department of Pathophysiology, George Emil Palade University of Medicine, Pharmacy, Science, and Technology of Târgu Mureș, 540139 Târgu Mureș, Romania; (A.L.); (O.S.C.)
- Department of Pathophysiology, Clinical County Hospital Mures, 540136 Târgu Mures, Romania;
| | - Mihail-Alexandru Badea
- Department of Dermatology, George Emil Palade University of Medicine, Pharmacy, Science, and Technology of Târgu Mureș, 540139 Târgu Mureș, Romania;
- Department of Dermatology, Clinical County Hospital Mures, 540136 Târgu Mures, Romania
| | - Mártha Orsolya Katalin Ilona
- Department of Urology, George Emil Palade University of Medicine, Pharmacy, Science, and Technology of Târgu Mureș, 540139 Târgu Mureș, Romania; (D.P.-H.); (M.O.K.I.)
- Department of Urology, Clinical County Hospital Mures, 540136 Târgu Mures, Romania;
| | - Ciprian Todea-Moga
- Department of Urology, George Emil Palade University of Medicine, Pharmacy, Science, and Technology of Târgu Mureș, 540139 Târgu Mureș, Romania; (D.P.-H.); (M.O.K.I.)
- Department of Urology, Clinical County Hospital Mures, 540136 Târgu Mures, Romania;
| | - Mihai Dorin Vartolomei
- Department of Cell and Molecular Biology, George Emil Palade University of Medicine, Pharmacy, Science, and Technology of Târgu Mureș, 540139 Târgu Mureș, Romania;
| | - Georgescu Rares
- Department of General Surgery, George Emil Palade University of Medicine, Pharmacy, Science, and Technology of Târgu Mureș, 540139 Târgu Mureș, Romania;
- Department of General Surgery, Clinical County Hospital Mures, 540136 Târgu Mures, Romania
| | - Nicolae Crisan
- Department of Urology, Iului Hatieganu University of Medicine and Pharmacy, 400012 Cluj-Napoca, Romania
| | - Ovidiu Bogdan Feciche
- Department of Urology, Faculty of Medicine and Pharmacy, University of Oradea, 410087 Oradea, Romania;
- Department of Urology, Emergency County Hospital Oradea, 410169 Oradea, Romania
| |
Collapse
|
44
|
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
|
45
|
Boeri M, Sabia F, Ledda RE, Balbi M, Suatoni P, Segale M, Zanghì A, Cantarutti A, Rolli L, Valsecchi C, Corrao G, Marchianò A, Pastorino U, Sozzi G. Blood microRNA testing in participants with suspicious low-dose CT findings: follow-up of the BioMILD lung cancer screening trial. THE LANCET REGIONAL HEALTH. EUROPE 2024; 46:101070. [PMID: 39319217 PMCID: PMC11421266 DOI: 10.1016/j.lanepe.2024.101070] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/19/2024] [Revised: 08/22/2024] [Accepted: 08/30/2024] [Indexed: 09/26/2024]
Abstract
Background The proper management of suspicious radiologic findings is crucial to optimize the effectiveness of low-dose computed tomography (LDCT) lung cancer screening trials. In the BioMILD study, we evaluated the utility of combining a plasma 24-microRNA signature classifier (MSC) and LDCT to define the individual risk and personalize screening strategies. Here we aim to assess the utility of repeated MSC testing during annual screening rounds in 1024 participants with suspicious LDCT findings. Methods The primary outcome was two-year lung cancer incidence in relation to MSC test results, reported as relative risk (RR) with 95% confidence interval (CI). Lung cancer incidence and mortality were estimated using extended Cox models for time-dependent covariates, yielding the respective hazard ratios (HR). Clinicaltrials.gov ID: NCT02247453. Findings With a median follow-up of 8.5 years, the full study set included 1403 indeterminate LDCT (CTind) and 584 positive LDCT (CT+) results. A lung cancer RR increase in MSC+ compared to MSC- participants was observed in both the CTind (RR: 2.5; 95% CI: 1.4-4.32) and CT+ (RR: 2.6; 95% CI: 1.81-3.74) groups and was maintained when considering stage I or resectable tumors only. A 98% negative predictive value in CTind/MSC- and a 30% positive predictive value in CT+/MSC+ lesions were recorded. At seven years' follow-up, MSC+ participants had a cumulative HR of 4.4 (95% CI: 3.0-6.4) for lung cancer incidence and of 8.1 (95% CI: 2.7-24.5) for lung cancer mortality. Interpretation Our study shows that MSC can be reliably performed during LDCT screening rounds to increase the accuracy of lung cancer risk and mortality prediction and supports its clinical utility in the management of LDCT findings of uncertain malignancy. Funding Italian Association for Cancer Research; Italian Ministry of Health; Horizon2020; National Cancer Institute (NCI); Gensignia LifeScience.
Collapse
Affiliation(s)
- Mattia Boeri
- Unit of Epigenomics & Biomarkers of Solid Tumors, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, 20133, Italy
| | - Federica Sabia
- Unit of Thoracic Surgery, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, 20133, Italy
| | - Roberta E. Ledda
- Unit of Thoracic Surgery, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, 20133, Italy
- Department of Medicine and Surgery (DiMeC), Section of Radiology, Unit of Surgical Sciences, University of Parma, Parma, 43121, Italy
| | - Maurizio Balbi
- Unit of Thoracic Surgery, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, 20133, Italy
- Department of Oncology, Radiology Unit, San Luigi Gonzaga Hospital, University of Turin, Orbassano, 10043, Italy
| | - Paola Suatoni
- Unit of Thoracic Surgery, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, 20133, Italy
| | - Miriam Segale
- Unit of Epigenomics & Biomarkers of Solid Tumors, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, 20133, Italy
| | - Anna Zanghì
- Unit of Epigenomics & Biomarkers of Solid Tumors, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, 20133, Italy
| | - Anna Cantarutti
- Division of Biostatistics, Department of Statistics and Quantitative Methods, Epidemiology and Public Health, University of Milano-Bicocca, Milan, 20126, Italy
| | - Luigi Rolli
- Unit of Thoracic Surgery, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, 20133, Italy
| | - Camilla Valsecchi
- Unit of Thoracic Surgery, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, 20133, Italy
| | - Giovanni Corrao
- Division of Biostatistics, Department of Statistics and Quantitative Methods, Epidemiology and Public Health, University of Milano-Bicocca, Milan, 20126, Italy
| | - Alfonso Marchianò
- Department of Radiology, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, 20133, Italy
| | - Ugo Pastorino
- Unit of Thoracic Surgery, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, 20133, Italy
| | - Gabriella Sozzi
- Unit of Epigenomics & Biomarkers of Solid Tumors, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, 20133, Italy
| |
Collapse
|
46
|
Faupel-Badger J, Kohaar I, Bahl M, Chan AT, Campbell JD, Ding L, De Marzo AM, Maitra A, Merrick DT, Hawk ET, Wistuba II, Ghobrial IM, Lippman SM, Lu KH, Lawler M, Kay NE, Tlsty TD, Rebbeck TR, Srivastava S. Defining precancer: a grand challenge for the cancer community. Nat Rev Cancer 2024; 24:792-809. [PMID: 39354069 DOI: 10.1038/s41568-024-00744-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 08/16/2024] [Indexed: 10/03/2024]
Abstract
The term 'precancer' typically refers to an early stage of neoplastic development that is distinguishable from normal tissue owing to molecular and phenotypic alterations, resulting in abnormal cells that are at least partially self-sustaining and function outside of normal cellular cues that constrain cell proliferation and survival. Although such cells are often histologically distinct from both the corresponding normal and invasive cancer cells of the same tissue origin, defining precancer remains a challenge for both the research and clinical communities. Once sufficient molecular and phenotypic changes have occurred in the precancer, the tissue is identified as a 'cancer' by a histopathologist. While even diagnosing cancer can at times be challenging, the determination of invasive cancer is generally less ambiguous and suggests a high likelihood of and potential for metastatic disease. The 'hallmarks of cancer' set out the fundamental organizing principles of malignant transformation but exactly how many of these hallmarks and in what configuration they define precancer has not been clearly and consistently determined. In this Expert Recommendation, we provide a starting point for a conceptual framework for defining precancer, which is based on molecular, pathological, clinical and epidemiological criteria, with the goal of advancing our understanding of the initial changes that occur and opportunities to intervene at the earliest possible time point.
Collapse
Affiliation(s)
| | - Indu Kohaar
- Division of Cancer Prevention, National Cancer Institute, NIH, Rockville, MD, USA
| | - Manisha Bahl
- Division of Breast Imaging, Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Andrew T Chan
- Clinical and Translational Epidemiology Unit, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Joshua D Campbell
- Department of Medicine, Boston University School of Medicine, Boston, MA, USA
| | - Li Ding
- Department of Medicine and Genetics, McDonnell Genome Institute, and Siteman Cancer Center, Washington University in St Louis, Saint Louis, MO, USA
| | - Angelo M De Marzo
- Department of Pathology, Urology and Oncology, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Anirban Maitra
- Department of Translational Molecular Pathology, Sheikh Ahmed Center for Pancreatic Cancer Research, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Daniel T Merrick
- Division of Pathology, School of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Ernest T Hawk
- Division of Cancer Prevention and Population Sciences, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Ignacio I Wistuba
- Department of Translational Molecular Pathology, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Irene M Ghobrial
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Scott M Lippman
- Department of Medicine, University of California, La Jolla, San Diego, CA, USA
| | - Karen H Lu
- Department of Gynecological Oncology, Moffitt Cancer Center, Tampa, FL, USA
| | - Mark Lawler
- Patrick G Johnson Centre for Cancer Research, School of Medicine, Dentistry and Biomedical Sciences, Queen's University Belfast, Belfast, UK
| | - Neil E Kay
- Division of Hematology, Mayo Clinic, Rochester, MN, USA
| | - Thea D Tlsty
- Department of Medicine and Epidemiology and Biostatistics, University of California San Francisco, San Francisco, CA, USA
| | - Timothy R Rebbeck
- Dana-Farber Cancer Institute and Harvard TH Chan School of Public Health, Boston, MA, USA
| | - Sudhir Srivastava
- Division of Cancer Prevention, National Cancer Institute, NIH, Rockville, MD, USA.
| |
Collapse
|
47
|
Linthorst J, Nivard M, Sistermans EA. GWAS shows the genetics behind cell-free DNA and highlights the importance of p.Arg206Cys in DNASE1L3 for non-invasive testing. Cell Rep 2024; 43:114799. [PMID: 39331505 DOI: 10.1016/j.celrep.2024.114799] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2024] [Revised: 07/16/2024] [Accepted: 09/11/2024] [Indexed: 09/29/2024] Open
Abstract
The properties of cell-free DNA (cfDNA) are intensely studied for their potential as non-invasive biomarkers. We explored the effect of common genetic variants on the concentration and fragmentation properties of cfDNA using a genome-wide association study (GWAS) based on low-coverage whole-genome sequencing data of 140,000 Dutch non-invasive prenatal tests (NIPTs). Our GWAS detects many genome-wide significant loci, functional enrichments for phagocytes, liver, adipose tissue, and macrophages, and genetic correlations with autoimmune and cardiovascular disease. A common (7%) missense variant in DNASE1L3 (p.Arg206Cys) strongly affects all cfDNA properties. It increases the size of fragments, lowers cfDNA concentrations, affects the distribution of cleave-site motifs, and increases the fraction of circulating fetal DNA during pregnancy. For the application of NIPT, and potentially other cfDNA-based tests, this variant has direct clinical consequences, as it increases the odds of inconclusive results and impairs the sensitivity of NIPT by causing predictors to overestimate the fetal fraction.
Collapse
Affiliation(s)
- Jasper Linthorst
- Department of Human Genetics, Amsterdam UMC Location VU, Amsterdam, the Netherlands; Amsterdam Reproduction & Development, Amsterdam, the Netherlands; Amsterdam Institute for Immunology and Infectious Diseases, Amsterdam, the Netherlands.
| | - Michel Nivard
- Department of Biological Psychology, Vrije Universiteit, Amsterdam, the Netherlands; Amsterdam Public Health Research Institute, Amsterdam, the Netherlands
| | - Erik A Sistermans
- Department of Human Genetics, Amsterdam UMC Location VU, Amsterdam, the Netherlands; Amsterdam Reproduction & Development, Amsterdam, the Netherlands.
| |
Collapse
|
48
|
Kennedy E, Durm G, Farlow JL. Multicancer Early Detection Tests: A State-of-the-Art Review for Otolaryngologists. OTO Open 2024; 8:e70040. [PMID: 39463807 PMCID: PMC11512445 DOI: 10.1002/oto2.70040] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2024] [Accepted: 10/15/2024] [Indexed: 10/29/2024] Open
Abstract
Objective To provide a review of the science and applicability of current multi-cancer early detection (MCED) tests for otolaryngologists. Data Sources PubMed, clinicaltrials.gov, company websites. Review Methods Using PRISMA methodology, primary literature regarding MCED tests was queried from April 26 to May 12, 2024 using MCED search terms. Ongoing clinical trials incorporating MCED screens were identified via the National Institutes of Health clinicaltrials.gov website. Company websites for available or upcoming MCED tests were reviewed. Conclusion Long-term robust data regarding the performance characteristics, effects on clinical outcomes, and cost-utility of MCED tests for head and neck cancer are currently lacking. Otolaryngologists should be aware of the implications of MCED tests as these assays become more widely used. Implications for Practice Although not FDA-approved or covered by insurances at the time of writing of this manuscript, MCED testing is rapidly gaining interest, and patients with positive tests are presenting to otolaryngologists for evaluation. While MCED technologies hold great promise for early detection of disease and potential reduction of morbidity and mortality, more study is needed about their utility for head and neck cancer and optimal diagnostic workflows.
Collapse
Affiliation(s)
- Elena Kennedy
- Department of Otolaryngology–Head and Neck SurgeryIndiana University School of MedicineIndianapolisIndianaUSA
| | - Greg Durm
- Department of Medicine, Division of Hematology/OncologyIndiana University School of MedicineIndianapolisIndianaUSA
| | - Janice L. Farlow
- Department of Otolaryngology–Head and Neck SurgeryIndiana University School of MedicineIndianapolisIndianaUSA
| |
Collapse
|
49
|
Fu SW, Tang C, Tan X, Srivastava S. Liquid biopsy for early cancer detection: technological revolutions and clinical dilemma. Expert Rev Mol Diagn 2024; 24:937-955. [PMID: 39360748 DOI: 10.1080/14737159.2024.2408744] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2024] [Accepted: 09/22/2024] [Indexed: 10/05/2024]
Abstract
INTRODUCTION Liquid biopsy is an innovative advancement in oncology, offering a noninvasive method for early cancer detection and monitoring by analyzing circulating tumor cells, DNA, RNA, and other biomarkers in bodily fluids. This technique has the potential to revolutionize precision oncology by providing real-time analysis of tumor dynamics, enabling early detection, monitoring treatment responses, and tailoring personalized therapies based on the molecular profiles of individual patients. AREAS COVERED In this review, the authors discuss current methodologies, technological challenges, and clinical applications of liquid biopsy. This includes advancements in detecting minimal residual disease, tracking tumor evolution, and combining liquid biopsy with other diagnostic modalities for precision oncology. Key areas explored are the sensitivity, specificity, and integration of multi-omics, AI, ML, and LLM technologies. EXPERT OPINION Liquid biopsy holds great potential to revolutionize cancer care through early detection and personalized treatment strategies. However, its success depends on overcoming technological and clinical hurdles, such as ensuring high sensitivity and specificity, interpreting results amidst tumor heterogeneity, and making tests accessible and affordable. Continued innovation and collaboration are crucial to fully realize the potential of liquid biopsy in improving early cancer detection, treatment, and monitoring.
Collapse
Affiliation(s)
- Sidney W Fu
- Division of Cancer Prevention, National Cancer Institute, National Institutes of Health, Rockville, MD, USA
| | - Cong Tang
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Lisbon, Portugal
| | - Xiaohui Tan
- Division of LS Research, LSBioscience, LLC, Frederick, USA
| | - Sudhir Srivastava
- Cancer Biomarkers Research Group, Division of Cancer Prevention, National Cancer Institute, National Institutes of Health, Rockville, MD, USA
| |
Collapse
|
50
|
Hernandez-Herrera GA, Calcano GA, Nagelschneider AA, Routman DM, Van Abel KM. Imaging Modalities for Head and Neck Cancer: Present and Future. Surg Oncol Clin N Am 2024; 33:617-649. [PMID: 39244284 DOI: 10.1016/j.soc.2024.04.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/09/2024]
Abstract
Several imaging modalities are utilized in the diagnosis, treatment, and surveillance of head and neck cancer. First-line imaging remains computed tomography (CT); however, MRI, PET with CT (PET/CT), and ultrasound are often used. In the last decade, several new imaging modalities have been developed that have the potential to improve early detection, modify treatment, decrease treatment morbidity, and augment surveillance. Among these, molecular imaging, lymph node mapping, and adjustments to endoscopic techniques are promising. The present review focuses on existing imaging, novel techniques, and the recent changes to imaging practices within the field.
Collapse
|