1
|
Rous B, Clarke CA, Hubbell E, Sasieni P. Assessment of the impact of multi-cancer early detection test screening intervals on late-stage cancer at diagnosis and mortality using a state-transition model. BMJ Open 2025; 15:e086648. [PMID: 40341158 DOI: 10.1136/bmjopen-2024-086648] [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] [Indexed: 05/10/2025] Open
Abstract
OBJECTIVE Multi-cancer early detection (MCED) tests are novel technologies that detect cancer signals from a broad set of cancer types using a single blood sample. The objective of this study was to estimate the effect of screening with an MCED test at different intervals on cancer stage at diagnosis and mortality endpoints. DESIGN The current model is based on a previously published state-transition model that estimated the outcomes of a screening programme using an MCED test when added to usual care for persons aged 50-79. Herein, we expand this analysis to model the time of cancer diagnosis and patient mortality with MCED screening undertaken using different screening schedules. Screening intervals between 6 months and 3 years, with emphasis on annual and biennial screening, were investigated for two sets of tumour growth rate scenarios: 'fast (dwell time=2-4 years in stage I) and 'fast aggressive' (dwell time=1-2 years in stage I), with decreasing dwell times for successive stages. SETTING Inputs for the model include (1) published MCED performance measures from a large case-control study by cancer type and stage at diagnosis and (2) Surveillance, Epidemiology and End Results (SEER) data describing stage-specific incidence and cancer-specific survival for persons aged 50-79 in the US for all cancer incidence. OUTCOME MEASURES We used the following outcome measures: diagnostic yield, stage shift, and mortality. RESULTS Annual screening under the fast tumour growth scenario was associated with more favourable diagnostic yield. There were 370 more cancer signals detected/year/100,000 people screened, 49% fewer late-stage diagnoses, and 21% fewer deaths within 5 years than usual care. Biennial screening had a similar, but less substantial, impact (292 more cancer signals detected/year/100,000 people screened; 39% fewer late-stage diagnoses, and 17% fewer deaths within 5 years than usual care). Annual screening prevented more deaths within 5 years than biennial screening for the fast tumour growth scenario. However, biennial screening had a higher positive predictive value (54% vs 43%); it was also more efficient per 100,000 tests in preventing deaths within 5 years (132 vs 84), but prevented fewer deaths per year. CONCLUSION Adding MCED test screening to usual care at any interval could improve patient outcomes. Annual MCED test screening provided more overall benefit than biennial screening. Modelling the sensitivity of outcomes to different MCED screening intervals can inform timescales for investigation in trials.
Collapse
Affiliation(s)
- Brian Rous
- Cambridge University Hospitals NHS Foundation Trust, Cambridge, UK
| | | | | | | |
Collapse
|
2
|
Margoni A, Papavassiliou AG. The Potential of a Cell-Free DNA Methylation-Based Blood Test in Colorectal Cancer Screening. Mol Diagn Ther 2025:10.1007/s40291-025-00783-9. [PMID: 40327299 DOI: 10.1007/s40291-025-00783-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/13/2025] [Indexed: 05/07/2025]
Affiliation(s)
- Angeliki Margoni
- Department of Biological Chemistry, Medical School, National and Kapodistrian University of Athens, 11527, Athens, Greece
| | - Athanasios G Papavassiliou
- Department of Biological Chemistry, Medical School, National and Kapodistrian University of Athens, 11527, Athens, Greece.
| |
Collapse
|
3
|
Guigal-Stephan N, Lockhart B, Moser T, Heitzer E. A perspective review on the systematic implementation of ctDNA in phase I clinical trial drug development. J Exp Clin Cancer Res 2025; 44:79. [PMID: 40022112 PMCID: PMC11871688 DOI: 10.1186/s13046-025-03328-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2024] [Accepted: 02/13/2025] [Indexed: 03/03/2025] Open
Abstract
Circulating tumour DNA (ctDNA) represents an increasingly important biomarker for the screening, diagnosis and management of patients in clinical practice in advanced/metastatic disease across multiple cancer types. In this context, ctDNA-based comprehensive genomic profiling is now available for patient management decisions, and several ctDNA-based companion diagnostic assays have been approved by regulatory agencies. However, although the assessment of ctDNA levels in Phase II-III drug development is now gathering momentum, it remains somewhat surprisingly limited in the early Phase I phases in light of the potential opportunities provided by such analysis. In this perspective review, we investigate the potential and hurdles of applying ctDNA testing for the inclusion and monitoring of patients in phase 1 clinical trials. This will enable more informed decisions regarding patient inclusion, dose optimization, and proof-of-mechanism of drug biological activity and molecular response, thereby supporting the evolving oncology drug development paradigm. Furthermore, we will highlight the use of cost-efficient, agnostic genome-wide techniques (such as low-pass whole genome sequencing and fragmentomics) and methylation-based methods to facilitate a more systematic integration of ctDNA in early clinical trial settings.
Collapse
Affiliation(s)
- Nolwen Guigal-Stephan
- Translational Medicine, Institut de Recherches Servier, 22 route 128, Gif-sur-Yvette, Saclay, 91190, France.
| | - Brian Lockhart
- Translational Medicine, Institut de Recherches Servier, 22 route 128, Gif-sur-Yvette, Saclay, 91190, France
| | - Tina Moser
- Institute of Human Genetics, Diagnostic & Research Center for Molecular BioMedicine, Medical University of Graz, Neue Stiftingtalstrasse 6, Graz, 8010, Austria
| | - Ellen Heitzer
- Institute of Human Genetics, Diagnostic & Research Center for Molecular BioMedicine, Medical University of Graz, Neue Stiftingtalstrasse 6, Graz, 8010, Austria.
| |
Collapse
|
4
|
Driussi A, Lamaze FC, Kordahi M, Armero VS, Gaudreault N, Orain M, Enlow W, Abbosh C, Hodgson D, Dasgupta A, Gagné A, Bossé Y, Joubert P. Clinicopathological Predictors of the Presence of Blood Circulating Tumor DNA in Early-Stage Non-Small Cell Lung Cancers. Mod Pathol 2025; 38:100744. [PMID: 40020968 DOI: 10.1016/j.modpat.2025.100744] [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: 10/25/2024] [Revised: 02/12/2025] [Accepted: 02/12/2025] [Indexed: 03/03/2025]
Abstract
The implementation of lung cancer screening programs across the world has drawn considerable attention to improving early-stage lung cancer detection and prognostication. Several blood-based assays detecting circulating tumor DNA (ctDNA) recently emerged as noninvasive methods to detect malignancies. However, their limited sensitivity and predictive value remain a hurdle to their clinical use. We aimed to evaluate the association between clinicopathological parameters and presurgical ctDNA detection in clinical stage I non-small cell lung cancer patients to further understand ctDNA shedding biology. The cohort included 180 adenocarcinomas (LUAD) and 80 squamous cell carcinomas (LUSC) stage I patients who underwent lung cancer resection. Patients' clinical and pathological features were collected. A multicancer early-detection test (GRAIL LLC) was used to detect ctDNA using targeted methylation patterns. The association between the cell-free DNA tumor methylated fraction (TMeF) and the clinicopathological predictors was evaluated using univariate and multivariate modeling. LUSC was associated with a higher TMeF than LUAD. Pathological stage, tumor grade, and tumor volume were key determinants of ctDNA detection in both LUSC and LUAD. In LUAD, ctDNA detection also correlated with histologic pattern composition, necrosis, acute inflammation, and, to a lesser degree, spread through alveolar spaces and lymphovascular invasion. Based on our results, we propose classification methods for both LUAD (using histologic pattern composition) and LUSC (using tumor grade and pathological stage) to identify patients likely to have high ctDNA levels. These results confirm previous findings and suggest that previously unidentified factors, including histologic pattern composition and acute inflammation, influence ctDNA levels. These results will help in understanding the ctDNA shedding process and may allow identification of patients eligible for ctDNA detection-based follow-up.
Collapse
Affiliation(s)
- Arnaud Driussi
- Institut universitaire de cardiologie et de pneumologie de Québec - Université Laval, Quebec City, Canada
| | - Fabien C Lamaze
- Institut universitaire de cardiologie et de pneumologie de Québec - Université Laval, Quebec City, Canada
| | - Manal Kordahi
- Institut universitaire de cardiologie et de pneumologie de Québec - Université Laval, Quebec City, Canada
| | - Victoria Saavedra Armero
- Institut universitaire de cardiologie et de pneumologie de Québec - Université Laval, Quebec City, Canada
| | - Nathalie Gaudreault
- Institut universitaire de cardiologie et de pneumologie de Québec - Université Laval, Quebec City, Canada
| | - Michèle Orain
- Institut universitaire de cardiologie et de pneumologie de Québec - Université Laval, Quebec City, Canada
| | - William Enlow
- Institut universitaire de cardiologie et de pneumologie de Québec - Université Laval, Quebec City, Canada
| | - Chris Abbosh
- Translational Medicine Early Oncology, AstraZeneca, Cambridge, United Kingdom
| | - Darren Hodgson
- Translational Medicine Early Oncology, AstraZeneca, Cambridge, United Kingdom
| | - Abhijit Dasgupta
- Oncology Data Science, Oncology R&D, AstraZeneca, Gaithersburg, Maryland
| | - Andréanne Gagné
- Institut universitaire de cardiologie et de pneumologie de Québec - Université Laval, Quebec City, Canada
| | - Yohan Bossé
- Institut universitaire de cardiologie et de pneumologie de Québec - Université Laval, Quebec City, Canada; Department of Molecular Medicine, Université Laval, Quebec City, Canada
| | - Philippe Joubert
- Institut universitaire de cardiologie et de pneumologie de Québec - Université Laval, Quebec City, Canada; Department of Molecular Biology, Pathology and Medical Biochemistry, Université Laval, Quebec City, Canada.
| |
Collapse
|
5
|
Hu P, Prorok PC, Katki HA. Design of randomized controlled trials to estimate cancer-mortality reductions from multicancer detection screening. J Natl Cancer Inst 2025; 117:303-311. [PMID: 39383209 PMCID: PMC11807432 DOI: 10.1093/jnci/djae247] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2024] [Revised: 09/04/2024] [Accepted: 09/30/2024] [Indexed: 10/11/2024] Open
Abstract
BACKGROUND Determining whether screening with multicancer detection (MCD) tests saves lives requires randomized controlled trials (RCTs). To inform RCT design, we estimated cancer-mortality outcomes from hypothetical MCD RCTs. METHODS We used the Hu-Zelen model, previously used to plan the National Lung Screening Trial (NLST), to estimate mortality reductions, sample size, and power for 9 cancers for different RCT design parameters and MCD test parameters. RESULTS Our base-case RCT with 5 yearly screens and 100 000 people ages 60-74 in each arm, who also undergo standard-of-care screens, has 87%-89% power to detect a 9%-10% mortality reduction (Number Needed to Screen [NNS] = 578-724) over 7-9 years. The majority of prevented deaths were from lung cancers (base-case [64%-66%] and all sensitivity analyses), 8%-10% from colorectal cancer, and 26% from the other 7 cancers, mostly from stomach or ovary or esophagus (due to excellent stage 1 survival) and less from liver or pancreas (poor stage 1 survival) or head and neck or lymphoma (excellent stage 4 survival). There was limited power for mortality reductions at most individual cancer sites. Base-case findings were sensitive to test sensitivity, stage shifts, and mean sojourn times in the preclinical state (especially for lung cancer), but 90% power could be recovered by recruiting a substantially higher risk population. CONCLUSIONS Large-scale MCD RCTs would have 89% power to detect an approximate 10% cancer mortality reduction over a relatively short 7-9 year timeframe from trial entry. The estimated NNS for MCD RCTs compares favorably with mammographic screening. Most prevented cancer deaths in a well-powered MCD RCT would likely be from lung cancer. Non-lung and non-colorectal cancer sites could be a meaningful proportion of prevented cancer deaths, but power is insufficient to isolate non-lung-cancer mortality reductions.
Collapse
Affiliation(s)
- Ping Hu
- Division of Cancer Prevention, Department of Health and Human Services, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Philip C Prorok
- Division of Cancer Prevention, Department of Health and Human Services, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Hormuzd A Katki
- Division of Cancer Epidemiology and Genetics, Department of Health and Human Services, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| |
Collapse
|
6
|
Øgaard N, Jensen SØ, Ørntoft MBW, Demuth C, Rasmussen MH, Henriksen TV, Nors J, Frydendahl A, Lyskjær I, Nesic M, Therkildsen C, Kleif J, Gögenur M, Jørgensen LN, Vilandt J, Seidelin JB, Gotschalck KA, Jaensch C, Andersen B, Løve US, Thorlacius-Ussing O, Andersen PV, Kolbro T, Monti A, Kildsig J, Bondeven P, Schlesinger NH, Iversen LH, Rasmussen M, Gögenur I, Bramsen JB, Andersen CL. Circulating tumour DNA and risk of recurrence in patients with asymptomatic versus symptomatic colorectal cancer. Br J Cancer 2024; 131:1707-1715. [PMID: 39390251 PMCID: PMC11555384 DOI: 10.1038/s41416-024-02867-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2024] [Revised: 09/17/2024] [Accepted: 09/23/2024] [Indexed: 10/12/2024] Open
Abstract
BACKGROUND Multiple initiatives aim to develop circulating tumour DNA (ctDNA) tests for early cancer detection in asymptomatic individuals. The few studies describing ctDNA-testing in both asymptomatic and symptomatic patients report lower ctDNA detection in the asymptomatic patients. Here, we explore if asymptomatic patients differ from symptomatic patients e.g. by including a 'low-ctDNA-shedding' and 'less-aggressive' subgroup. METHODS ctDNA assessment was performed in two independent cohorts of consecutively recruited patients with asymptomatic colorectal cancer (CRC) (Cohort#1: n = 215, Cohort#2: n = 368) and symptomatic CRC (Cohort#1: n = 117, Cohort#2: n = 722). RESULTS After adjusting for tumour stage and size, the odds of ctDNA detection was significantly lower in asymptomatic patients compared to symptomatic patients (Cohort#1: OR: 0.4, 95%CI: 0.2-0.8, Cohort#2: OR: 0.7, 95%CI: 0.5-0.9). Further, the recurrence risk was lower in asymptomatic patients (Cohort#1: sHR: 0.6, 95%CI: 0.3-1.2, Cohort#2: sHR: 0.6, 95%CI: 0.4-1.0). Notably, ctDNA-negative asymptomatic patients had the lowest recurrence risk compared to the symptomatic patients (Cohort#1: sHR: 0.2, 95%CI: 0.1-0.6, Cohort#2: sHR: 0.3, 95%CI: 0.2-0.6). CONCLUSIONS Our study suggests that asymptomatic patients are enriched for a 'low-ctDNA-shedding-low-recurrence-risk' subgroup. Such insights are needed to guide ctDNA-based early-detection initiatives and should prompt discussions about de-escalation of therapy and follow-up for ctDNA-negative asymptomatic CRC patients.
Collapse
Affiliation(s)
- Nadia Øgaard
- Department of Molecular Medicine, Aarhus University Hospital, Aarhus, Denmark
- Institute of Clinical Medicine, Faculty of Health, Aarhus University, Aarhus, Denmark
| | - Sarah Østrup Jensen
- Department of Molecular Medicine, Aarhus University Hospital, Aarhus, Denmark
- Institute of Clinical Medicine, Faculty of Health, Aarhus University, Aarhus, Denmark
| | - Mai-Britt Worm Ørntoft
- Department of Molecular Medicine, Aarhus University Hospital, Aarhus, Denmark
- Department of Surgery, Gødstrup Hospital, Herning, Denmark
| | - Christina Demuth
- Department of Molecular Medicine, Aarhus University Hospital, Aarhus, Denmark
- Institute of Clinical Medicine, Faculty of Health, Aarhus University, Aarhus, Denmark
| | - Mads Heilskov Rasmussen
- Department of Molecular Medicine, Aarhus University Hospital, Aarhus, Denmark
- Institute of Clinical Medicine, Faculty of Health, Aarhus University, Aarhus, Denmark
| | - Tenna Vesterman Henriksen
- Department of Molecular Medicine, Aarhus University Hospital, Aarhus, Denmark
- Institute of Clinical Medicine, Faculty of Health, Aarhus University, Aarhus, Denmark
| | - Jesper Nors
- Department of Molecular Medicine, Aarhus University Hospital, Aarhus, Denmark
- Institute of Clinical Medicine, Faculty of Health, Aarhus University, Aarhus, Denmark
| | - Amanda Frydendahl
- Department of Molecular Medicine, Aarhus University Hospital, Aarhus, Denmark
- Institute of Clinical Medicine, Faculty of Health, Aarhus University, Aarhus, Denmark
| | - Iben Lyskjær
- Department of Molecular Medicine, Aarhus University Hospital, Aarhus, Denmark
- Institute of Clinical Medicine, Faculty of Health, Aarhus University, Aarhus, Denmark
| | - Marijana Nesic
- Department of Molecular Medicine, Aarhus University Hospital, Aarhus, Denmark
- Institute of Clinical Medicine, Faculty of Health, Aarhus University, Aarhus, Denmark
| | - Christina Therkildsen
- Gastro Unit, Surgical Section, Copenhagen University Hospital, Amager-Hvidovre, Denmark
| | - Jakob Kleif
- Gastro Unit, Surgical Section, Copenhagen University Hospital, Amager-Hvidovre, Denmark
- Department of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark
| | - Mikail Gögenur
- Center for Surgical Science, Department of Surgery, Zealand University Hospital, Køge, Denmark
| | | | - Jesper Vilandt
- Department of Surgery, Nordsjællands Hospital, Hillerød, Denmark
| | | | - Kåre Anderson Gotschalck
- Institute of Clinical Medicine, Faculty of Health, Aarhus University, Aarhus, Denmark
- Department of Surgery, Horsens Hospital, Horsens, Denmark
| | | | - Berit Andersen
- Institute of Clinical Medicine, Faculty of Health, Aarhus University, Aarhus, Denmark
- Department of Public Health Programs and University Research Clinic for Cancer Screening, Randers Regional Hospital, Randers, Denmark
| | | | | | | | - Thomas Kolbro
- Department of Surgery, Odense University Hospital, Svendborg, Denmark
| | - Alessio Monti
- Department of Surgery, North Denmark Regional Hospital Hjørring, Hjørring, Denmark
| | - Jeppe Kildsig
- Department of Surgery, Copenhagen University Hospital, Herlev, Denmark
| | - Peter Bondeven
- Department of Surgery, Regional Hospital Randers, Randers, Denmark
| | | | - Lene Hjerrild Iversen
- Institute of Clinical Medicine, Faculty of Health, Aarhus University, Aarhus, Denmark
- Department of Surgery, Aarhus University Hospital, Aarhus, Denmark
| | - Morten Rasmussen
- Digestive Disease Centre, Bispebjerg Hospital, Copenhagen, Denmark
| | - Ismail Gögenur
- Center for Surgical Science, Department of Surgery, Zealand University Hospital, Køge, Denmark
| | - Jesper Bertram Bramsen
- Department of Molecular Medicine, Aarhus University Hospital, Aarhus, Denmark
- Institute of Clinical Medicine, Faculty of Health, Aarhus University, Aarhus, Denmark
| | - Claus Lindbjerg Andersen
- Department of Molecular Medicine, Aarhus University Hospital, Aarhus, Denmark.
- Institute of Clinical Medicine, Faculty of Health, Aarhus University, Aarhus, Denmark.
| |
Collapse
|
7
|
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
|
8
|
Du H, Liu W, Li Y, Zhang L, Jiang F, Zhu D, Li J, Hu P, Yan N, Mao M, Li S. Genomic profiling of cell-free DNA from dogs with benign and malignant tumors. BMC Res Notes 2024; 17:264. [PMID: 39272211 PMCID: PMC11401444 DOI: 10.1186/s13104-024-06932-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2024] [Accepted: 09/03/2024] [Indexed: 09/15/2024] Open
Abstract
OBJECTIVE Cancer is currently the most common cause of death in adult dogs. Like humans, dogs have a one-third chance of developing cancer in their lifetime. We used shallow whole-genome sequencing (sWGS) to analyze blood cell-free DNA (cfDNA) from four tumor-bearing dogs (one with benign and three with malignant tumors) and 38 healthy dogs. RESULTS Similar to the results observed in the healthy dogs, no copy number aberration (CNA) was detected in the dog with benign lipomas, and the distribution of cfDNA fragment size (FS) closely resembled that of the healthy dogs. However, among the three dogs diagnosed with malignant tumors, two dogs exhibited varying degrees and quantities of CNAs. Compared to the distribution of FS in the healthy dogs, the cancer dogs exhibited a noticeable shift towards shorter lengths. These findings indicated that CNA and FS profiles derived from sWGS data can be used for non-invasive cancer detection in dogs.
Collapse
Affiliation(s)
- Hongchao Du
- New Ruipeng Pet Healthcare Group Co, Ltd, Shenzhen, China
| | - Wenfeng Liu
- Shanghai Companion Animal Hospital, Shanghai, China
| | - Yunfei Li
- Research & Development, TwixBio, Shenzhen, China
| | - Lijuan Zhang
- Clinical Laboratories, Shenyou Bio, Zhengzhou, China
| | | | - Dandan Zhu
- Clinical Laboratories, Shenyou Bio, Zhengzhou, China
| | - Jingshuai Li
- Clinical Laboratories, Shenyou Bio, Zhengzhou, China
| | - Pan Hu
- Research & Development, TwixBio, Shenzhen, China
| | - Ningning Yan
- Clinical Laboratories, Shenyou Bio, Zhengzhou, China
| | - Mao Mao
- Research & Development, TwixBio, Shenzhen, China.
- Yonsei Song-Dang Institute for Cancer Research, Yonsei University, Seoul, Korea.
- DBH Life Sciences & Health Industrial Park, 11F, Building 2, 2028 Shenyan Road, Yantian, Shenzhen, 518000, Guangdong, China.
| | - Shiyong Li
- Research & Development, TwixBio, Shenzhen, China.
| |
Collapse
|
9
|
Dai JY, Zhang J, Braun JV, Simon N, Hubbell E, Zhang N. Clinical performance and utility: A microsimulation model to inform the design of screening trials for a multi-cancer early detection test. J Med Screen 2024; 31:140-149. [PMID: 38304990 PMCID: PMC11330083 DOI: 10.1177/09691413241228041] [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: 07/19/2023] [Revised: 12/14/2023] [Accepted: 12/29/2023] [Indexed: 02/03/2024]
Abstract
OBJECTIVES Designing cancer screening trials for multi-cancer early detection (MCED) tests presents a significant methodology challenge, as natural histories of cell-free DNA-shedding cancers are not yet known. A microsimulation model was developed to project the performance and utility of an MCED test in cancer screening trials. METHODS Individual natural history of preclinical progression through cancer stages for 23 cancer classes was simulated by a stage-transition model under a broad range of cancer latency parameters. Cancer incidences and stage distributions at clinical presentation in simulated trials were set to match the data from Surveillance, Epidemiology, and End Results program. One or multiple rounds of annual screening using a targeted methylation-based MCED test (GalleriⓇ) was conducted to detect preclinical cancers. Mortality benefit of early detection was simulated by a stage-shift model. RESULTS In simulated trials, accounting for healthy volunteer effect and varying test sensitivity, positive predictive value in the prevalence screening round reached 48% to 61% in 6 natural history scenarios. After 3 rounds of annual screening, the cumulative proportions of stage I/II cancers increased by approximately 9% to 14%, the incidence of stage IV cancers was reduced by 37% to 46%, the reduction of stages III and IV cancer incidences was 9% to 24%, and the reduction of mortality reached 13% to 16%. Greater reductions of late-stage cancers and cancer mortality were achieved by five rounds of MCED screening. CONCLUSIONS Simulation results guide trial design and suggest that adding this MCED test to routine screening in the United States may shift cancer detection to earlier stages, and potentially save lives.
Collapse
Affiliation(s)
| | | | | | - Noah Simon
- Department of Biostatistics, University of Washington, Seattle, WA, USA
| | | | | |
Collapse
|
10
|
Mahal BA, Margolis M, Hubbell E, Chen C, Venstrom JM, Abran J, Kartlitz JJ, Wyatt AW, Klein EA. A Targeted Methylation-Based Multicancer Early Detection Blood Test Preferentially Detects High-Grade Prostate Cancer While Minimizing Overdiagnosis of Indolent Disease. JCO Precis Oncol 2024; 8:e2400269. [PMID: 39208374 PMCID: PMC11371104 DOI: 10.1200/po.24.00269] [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: 04/23/2024] [Revised: 06/12/2024] [Accepted: 06/28/2024] [Indexed: 09/04/2024] Open
Abstract
PURPOSE Indolent prostate cancer (PCa) is prevalent in the intended use population (adults age 50-79 years) for blood-based multicancer early detection (MCED) tests. We examined the detectability of PCa by a clinically validated, targeted methylation-based MCED test. METHODS Detectability by Gleason grade group (GG), clinical stage, association of detection status with tumor methylated fraction (TMeF), and overall survival (OS) were assessed in substudy 3 of Circulating Cell-Free Genome Atlas (CCGA; ClinicalTrials.gov identifier: NCT02889978) and PATHFINDER (ClinicalTrials.gov identifier: NCT04241796) studies. RESULTS Test sensitivity for PCa in substudy 3 of CCGA was 11.2% (47/420). The test detected 0 (0%) of 58 low-grade (GG1), 3 (1.9%) of 157 favorable intermediate-grade (GG2), 4 (5.1%) of 78 unfavorable intermediate-grade (GG3), and 36 (31.9%) of 113 high-grade (GG4 and 5) cancers and 3 (3.2%) of 95 stage I, 11 (4.7%) of 235 stage II, 7 (14.9%) of 47 stage III, and 22 (81.5%) of 27 stage IV cases. The median TMeF was higher for detected than nondetected cases (2,106.0 parts per million [PPM]; IQR, 349.8-24,376.3 v 24.4 PPM; IQR, 17.8-38.5; P < .05). Nondetected cases had better OS (P < .05; hazard ratio [HR], 0.263 [95% CI, 0.104 to 0.533]) and detected cases had similar survival (P = .2; HR, 0.672 [95% CI, 0.323 to 1.21]) compared with SEER adjusted for age, GG, and stage. Performance was similar in PATHFINDER, with no detected GG1/2 (0/13) or stage I/II (0/16) cases. CONCLUSION This MCED test preferentially detects high-grade, clinically significant PCa. Use in population-based screening programs in addition to standard-of-care screening is unlikely to exacerbate overdiagnosis of indolent PCa.
Collapse
Affiliation(s)
- Brandon A. Mahal
- Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Miami, FL
| | | | | | | | | | | | | | - Alexander W. Wyatt
- Department of Urologic Sciences, Vancouver Prostate Centre, University of British Columbia, Vancouver, BC, Canada
| | | |
Collapse
|
11
|
Scimia M, Pepe F, Russo G, Palumbo L, Malapelle U, Chuang R, Scimia S, Sha M, Tanaka H, Shen S, Chen D, Troncone G, Bianco MA. The Impact of potential 'confounders' on the diagnostic sensitivity of circulating free DNA in management of FIT+ patients: a pilot study. J Clin Pathol 2024; 77:557-560. [PMID: 38649261 PMCID: PMC11287558 DOI: 10.1136/jcp-2024-209527] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2024] [Accepted: 03/29/2024] [Indexed: 04/25/2024]
Abstract
Cell-free DNA (cfDNA) has long been established as a useful diagnostic and prognostic tool in a variety of clinical settings, ranging from infectious to cardiovascular and neoplastic diseases. However, non-neoplastic diseases can act as confounders impacting on the amount of cfDNA shed in bloodstream and on technical feasibility of tumour derived free circulating nucleic acids selecting patients with cancer. Here, we investigated the potential impact of other pathological processes in the clinical stratification of 637 FIT+ patients. A single and multiple logistic regression yielded similar results. Crude sensitivity was 75.9% versus adjusted sensitivity of 74.1%, relative risk 0.9761 (0.8516 to 1.1188), risk difference 0.0181 (-0.0835 to 0.1199) and OR 0.9079 (0.5264 to 1.5658). Potential confounding effect from other source of cfDNA plays a pivotal role in the clinical stratification of FIT+ patients.
Collapse
Affiliation(s)
| | - Francesco Pepe
- Public Health, University of Naples Federico II, Naples, Italy
| | - Gianluca Russo
- Public Health, University of Naples Federico II, Naples, Italy
| | - Lucia Palumbo
- Public Health, University of Naples Federico II, Naples, Italy
| | | | | | | | | | | | - Shuo Shen
- DiaCarta Inc, Richmond, Virginia, USA
| | | | | | | |
Collapse
|
12
|
Lukacova E, Hanzlikova Z, Podlesnyi P, Sedlackova T, Szemes T, Grendar M, Samec M, Hurtova T, Malicherova B, Leskova K, Budis J, Burjanivova T. Novel liquid biopsy CNV biomarkers in malignant melanoma. Sci Rep 2024; 14:15786. [PMID: 38982214 PMCID: PMC11233564 DOI: 10.1038/s41598-024-65928-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2024] [Accepted: 06/25/2024] [Indexed: 07/11/2024] Open
Abstract
Malignant melanoma (MM) is known for its abundance of genetic alterations and a tendency for rapid metastasizing. Identification of novel plasma biomarkers may enhance non-invasive diagnostics and disease monitoring. Initially, we examined copy number variations (CNV) in CDK genes (CDKN2A, CDKN2B, CDK4) using MLPA (gDNA) and ddPCR (ctDNA) analysis. Subsequently, low-coverage whole genome sequencing (lcWGS) was used to identify the most common CNV in plasma samples, followed by ddPCR verification of chosen biomarkers. CNV alterations in CDK genes were identified in 33.3% of FFPE samples (Clark IV, V only). Detection of the same genes in MM plasma showed no significance, neither compared to healthy plasmas nor between pre- versus post-surgery plasma. Sequencing data showed the most common CNV occurring in 6q27, 4p16.1, 10p15.3, 10q22.3, 13q34, 18q23, 20q11.21-q13.12 and 22q13.33. CNV in four chosen genes (KIF25, E2F1, DIP2C and TFG) were verified by ddPCR using 2 models of interpretation. Model 1 was concordant with lcWGS results in 54% of samples, for model 2 it was 46%. Although CDK genes have not been proven to be suitable CNV liquid biopsy biomarkers, lcWGS defined the most frequently affected chromosomal regions by CNV. Among chosen genes, DIP2C demonstrated a potential for further analysis.
Collapse
Affiliation(s)
- E Lukacova
- Department of Molecular Biology and Genomics, Comenius University in Bratislava, Jessenius Faculty of Medicine in Martin (JFM CU), Martin, Slovakia
| | | | - P Podlesnyi
- Instituto de Investigaciones Biomedicas de Barcelona (IIBB), CSIC /Centro Investigacion Biomedica en Red Enfermedades Neurodegenerativas (CiberNed), Barcelona, Spain
| | - T Sedlackova
- Geneton Ltd., Bratislava, Slovakia
- Science Park, Comenius University in Bratislava, Bratislava, Slovakia
| | - T Szemes
- Geneton Ltd., Bratislava, Slovakia
- Science Park, Comenius University in Bratislava, Bratislava, Slovakia
| | - M Grendar
- Laboratory of Bioinformatics and Biostatistics, Biomedical Center Martin, Comenius University in Bratislava, Jessenius Faculty of Medicine in Martin (JFM CU), Martin, Slovakia
| | - M Samec
- Department of Medical Biology, Jessenius Faculty of Medicine in Martin, Comenius University in Bratislava, Martin, Slovakia
| | - T Hurtova
- Department of Dermatovenereology, Jessenius Faculty of Medicine in Martin, Comenius University in Bratislava, Martin, Slovakia
| | - B Malicherova
- Department of Clinical Biochemistry, University Hospital in Martin and Jessenius Faculty of Medicine, Comenius University, Martin, Slovakia
| | - K Leskova
- Department of Pathological Anatomy, Jessenius Faculty of Medicine and University Hospital in Martin, Comenius University, Martin, Slovakia
| | - J Budis
- Geneton Ltd., Bratislava, Slovakia
- Science Park, Comenius University in Bratislava, Bratislava, Slovakia
| | - T Burjanivova
- Department of Molecular Biology and Genomics, Comenius University in Bratislava, Jessenius Faculty of Medicine in Martin (JFM CU), Martin, Slovakia.
| |
Collapse
|
13
|
Rickles-Young M, Tinoco G, Tsuji J, Pollock S, Haynam M, Lefebvre H, Glover K, Owen DH, Collier KA, Ha G, Adalsteinsson VA, Cibulskis C, Lennon NJ, Stover DG. Assay Validation of Cell-Free DNA Shallow Whole-Genome Sequencing to Determine Tumor Fraction in Advanced Cancers. J Mol Diagn 2024; 26:413-422. [PMID: 38490303 PMCID: PMC11090203 DOI: 10.1016/j.jmoldx.2024.01.014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2023] [Revised: 09/21/2023] [Accepted: 01/18/2024] [Indexed: 03/17/2024] Open
Abstract
Blood-based liquid biopsy is increasingly used in clinical care of patients with cancer, and fraction of tumor-derived DNA in circulation (tumor fraction; TFx) has demonstrated clinical validity across multiple cancer types. To determine TFx, shallow whole-genome sequencing of cell-free DNA (cfDNA) can be performed from a single blood sample, using an established computational pipeline (ichorCNA), without prior knowledge of tumor mutations, in a highly cost-effective manner. We describe assay validation of this approach to facilitate broad clinical application, including evaluation of assay sensitivity, precision, repeatability, reproducibility, pre-analytic factors, and DNA quality/quantity. Sensitivity to detect TFx of 3% (lower limit of detection) was 97.2% to 100% at 1× and 0.1× mean sequencing depth, respectively. Precision was demonstrated on distinct sequencing instruments (HiSeqX and NovaSeq) with no observable differences. The assay achieved prespecified 95% agreement of TFx across replicates of the same specimen (repeatability) and duplicate samples in different batches (reproducibility). Comparison of samples collected in EDTA and Streck tubes from single venipuncture in 23 patients demonstrated that EDTA or Streck tubes were comparable if processed within 8 hours. On the basis of a range of DNA inputs (1 to 50 ng), 20 ng cfDNA is the preferred input, with 5 ng minimum acceptable. Overall, this shallow whole-genome sequencing of cfDNA and ichorCNA approach offers sensitive, precise, and reproducible quantitation of TFx, facilitating assay application in clinical cancer care.
Collapse
Affiliation(s)
- Micah Rickles-Young
- Genomics Platform, Broad Institute of Harvard and Massachusetts Institute of Technology, Cambridge, Massachusetts
| | - Gabriel Tinoco
- Division of Medical Oncology, The Ohio State University College of Medicine, Columbus, Ohio; Ohio State University Comprehensive Cancer Center, Columbus, Ohio
| | - Junko Tsuji
- Genomics Platform, Broad Institute of Harvard and Massachusetts Institute of Technology, Cambridge, Massachusetts
| | - Sam Pollock
- Genomics Platform, Broad Institute of Harvard and Massachusetts Institute of Technology, Cambridge, Massachusetts
| | - Marcy Haynam
- Ohio State University Comprehensive Cancer Center, Columbus, Ohio; Stefanie Spielman Comprehensive Breast Center, Columbus, Ohio
| | - Heather Lefebvre
- Ohio State University Comprehensive Cancer Center, Columbus, Ohio; Stefanie Spielman Comprehensive Breast Center, Columbus, Ohio
| | - Kristyn Glover
- Ohio State University Comprehensive Cancer Center, Columbus, Ohio; Stefanie Spielman Comprehensive Breast Center, Columbus, Ohio
| | - Dwight H Owen
- Division of Medical Oncology, The Ohio State University College of Medicine, Columbus, Ohio; Ohio State University Comprehensive Cancer Center, Columbus, Ohio
| | - Katharine A Collier
- Division of Medical Oncology, The Ohio State University College of Medicine, Columbus, Ohio; Ohio State University Comprehensive Cancer Center, Columbus, Ohio
| | - Gavin Ha
- Public Health Sciences Division, Fred Hutchinson Cancer Research Center, Seattle, Washington
| | - Viktor A Adalsteinsson
- Genomics Platform, Broad Institute of Harvard and Massachusetts Institute of Technology, Cambridge, Massachusetts
| | - Carrie Cibulskis
- Genomics Platform, Broad Institute of Harvard and Massachusetts Institute of Technology, Cambridge, Massachusetts
| | - Niall J Lennon
- Genomics Platform, Broad Institute of Harvard and Massachusetts Institute of Technology, Cambridge, Massachusetts.
| | - Daniel G Stover
- Division of Medical Oncology, The Ohio State University College of Medicine, Columbus, Ohio; Ohio State University Comprehensive Cancer Center, Columbus, Ohio; Stefanie Spielman Comprehensive Breast Center, Columbus, Ohio.
| |
Collapse
|
14
|
Frydendahl A, Rasmussen MH, Jensen SØ, Henriksen TV, Demuth C, Diekema M, Ditzel HJ, Wen SWC, Pedersen JS, Dyrskjøt L, Andersen CL. Error-Corrected Deep Targeted Sequencing of Circulating Cell-Free DNA from Colorectal Cancer Patients for Sensitive Detection of Circulating Tumor DNA. Int J Mol Sci 2024; 25:4252. [PMID: 38673836 PMCID: PMC11049993 DOI: 10.3390/ijms25084252] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2024] [Revised: 04/09/2024] [Accepted: 04/10/2024] [Indexed: 04/28/2024] Open
Abstract
Circulating tumor DNA (ctDNA) is a promising biomarker, reflecting the presence of tumor cells. Sequencing-based detection of ctDNA at low tumor fractions is challenging due to the crude error rate of sequencing. To mitigate this challenge, we developed ultra-deep mutation-integrated sequencing (UMIseq), a fixed-panel deep targeted sequencing approach, which is universally applicable to all colorectal cancer (CRC) patients. UMIseq features UMI-mediated error correction, the exclusion of mutations related to clonal hematopoiesis, a panel of normal samples for error modeling, and signal integration from single-nucleotide variations, insertions, deletions, and phased mutations. UMIseq was trained and independently validated on pre-operative (pre-OP) plasma from CRC patients (n = 364) and healthy individuals (n = 61). UMIseq displayed an area under the curve surpassing 0.95 for allele frequencies (AFs) down to 0.05%. In the training cohort, the pre-OP detection rate reached 80% at 95% specificity, while it was 70% in the validation cohort. UMIseq enabled the detection of AFs down to 0.004%. To assess the potential for detection of residual disease, 26 post-operative plasma samples from stage III CRC patients were analyzed. From this we found that the detection of ctDNA was associated with recurrence. In conclusion, UMIseq demonstrated robust performance with high sensitivity and specificity, enabling the detection of ctDNA at low allele frequencies.
Collapse
Affiliation(s)
- Amanda Frydendahl
- Department of Molecular Medicine, Aarhus University Hospital, 8200 Aarhus, Denmark; (A.F.); (S.Ø.J.); (T.V.H.); (C.D.); (M.D.); (J.S.P.); (L.D.)
- Department of Clinical Medicine, Aarhus University, 8000 Aarhus, Denmark
| | - Mads Heilskov Rasmussen
- Department of Molecular Medicine, Aarhus University Hospital, 8200 Aarhus, Denmark; (A.F.); (S.Ø.J.); (T.V.H.); (C.D.); (M.D.); (J.S.P.); (L.D.)
- Department of Clinical Medicine, Aarhus University, 8000 Aarhus, Denmark
| | - Sarah Østrup Jensen
- Department of Molecular Medicine, Aarhus University Hospital, 8200 Aarhus, Denmark; (A.F.); (S.Ø.J.); (T.V.H.); (C.D.); (M.D.); (J.S.P.); (L.D.)
- Department of Clinical Medicine, Aarhus University, 8000 Aarhus, Denmark
| | - Tenna Vesterman Henriksen
- Department of Molecular Medicine, Aarhus University Hospital, 8200 Aarhus, Denmark; (A.F.); (S.Ø.J.); (T.V.H.); (C.D.); (M.D.); (J.S.P.); (L.D.)
- Department of Clinical Medicine, Aarhus University, 8000 Aarhus, Denmark
| | - Christina Demuth
- Department of Molecular Medicine, Aarhus University Hospital, 8200 Aarhus, Denmark; (A.F.); (S.Ø.J.); (T.V.H.); (C.D.); (M.D.); (J.S.P.); (L.D.)
- Department of Clinical Medicine, Aarhus University, 8000 Aarhus, Denmark
| | - Mathilde Diekema
- Department of Molecular Medicine, Aarhus University Hospital, 8200 Aarhus, Denmark; (A.F.); (S.Ø.J.); (T.V.H.); (C.D.); (M.D.); (J.S.P.); (L.D.)
- Department of Clinical Medicine, Aarhus University, 8000 Aarhus, Denmark
| | - Henrik Jørn Ditzel
- Institute of Molecular Medicine, University of Southern Denmark, 5000 Odense, Denmark;
- Department of Oncology, Odense University Hospital, 5000 Odense, Denmark
| | | | - Jakob Skou Pedersen
- Department of Molecular Medicine, Aarhus University Hospital, 8200 Aarhus, Denmark; (A.F.); (S.Ø.J.); (T.V.H.); (C.D.); (M.D.); (J.S.P.); (L.D.)
- Department of Clinical Medicine, Aarhus University, 8000 Aarhus, Denmark
- Bioinformatics Research Center, Faculty of Science, Aarhus University, 8000 Aarhus, Denmark
| | - Lars Dyrskjøt
- Department of Molecular Medicine, Aarhus University Hospital, 8200 Aarhus, Denmark; (A.F.); (S.Ø.J.); (T.V.H.); (C.D.); (M.D.); (J.S.P.); (L.D.)
- Department of Clinical Medicine, Aarhus University, 8000 Aarhus, Denmark
| | - Claus Lindbjerg Andersen
- Department of Molecular Medicine, Aarhus University Hospital, 8200 Aarhus, Denmark; (A.F.); (S.Ø.J.); (T.V.H.); (C.D.); (M.D.); (J.S.P.); (L.D.)
- Department of Clinical Medicine, Aarhus University, 8000 Aarhus, Denmark
| |
Collapse
|
15
|
Yasui K, Toshima T, Inada R, Umeda Y, Yano S, Tanioka H, Nyuya A, Fujiwara T, Yamada T, Naomoto Y, Goel A, Nagasaka T. Circulating cell-free DNA methylation patterns as non-invasive biomarkers to monitor colorectal cancer treatment efficacy without referencing primary site mutation profiles. Mol Cancer 2024; 23:1. [PMID: 38172877 PMCID: PMC10762960 DOI: 10.1186/s12943-023-01910-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2023] [Accepted: 11/30/2023] [Indexed: 01/05/2024] Open
Abstract
This study investigates methylation patterns in circulating cell-free DNA (ccfDNA) for their potential role in colorectal cancer (CRC) detection and the monitoring of treatment response. Through methylation microarrays and quantitative PCR assays, we analyzed 440 samples from The Cancer Genome Atlas (TCGA) and an additional 949 CRC samples. We detected partial or extensive methylation in over 85% of cases within three biomarkers: EFEMP1, SFRP2, and UNC5C. A methylation score for at least one of the six candidate regions within these genes' promoters was present in over 95% of CRC cases, suggesting a viable detection method. In evaluating ccfDNA from 97 CRC patients and 62 control subjects, a difference in methylation and recovery signatures was observed. The combined score, integrating both methylation and recovery metrics, showed high diagnostic accuracy, evidenced by an area under the ROC curve of 0.90 (95% CI = 0.86 to 0.94). While correlating with tumor burden, this score gave early insight into disease progression in a small patient cohort. Our results suggest that DNA methylation in ccfDNA could serve as a sensitive biomarker for CRC, offering a less invasive and potentially more cost-effective approach to augment existing cancer detection and monitoring modalities, possibly supporting comprehensive genetic mutation profiling.
Collapse
Affiliation(s)
- Kazuya Yasui
- Department of Gastroenterological Surgery, Dentistry and Pharmaceutical Sciences, Okayama University Graduate School of Medicine, Okayama, 700-8558, Japan
| | - Toshiaki Toshima
- Department of Gastroenterological Surgery, Dentistry and Pharmaceutical Sciences, Okayama University Graduate School of Medicine, Okayama, 700-8558, Japan
- Department of Gastroenterological Surgery, Kochi Health Sciences Centre, Kochi, 781-0111, Japan
| | - Ryo Inada
- Department of Gastroenterological Surgery, Kochi Health Sciences Centre, Kochi, 781-0111, Japan
| | - Yuzo Umeda
- Department of Gastroenterological Surgery, Dentistry and Pharmaceutical Sciences, Okayama University Graduate School of Medicine, Okayama, 700-8558, Japan
| | - Shuya Yano
- Department of Clinical Oncology, Kawasaki Medical School, 577 Matsushim, Kurashiki, 701-0912, Japan
| | - Hiroaki Tanioka
- Department of Clinical Oncology, Kawasaki Medical School, 577 Matsushim, Kurashiki, 701-0912, Japan
| | - Akihiro Nyuya
- Department of Clinical Oncology, Kawasaki Medical School, 577 Matsushim, Kurashiki, 701-0912, Japan
| | - Toshiyoshi Fujiwara
- Department of Gastroenterological Surgery, Dentistry and Pharmaceutical Sciences, Okayama University Graduate School of Medicine, Okayama, 700-8558, Japan
| | - Takeshi Yamada
- Department of Gastrointestinal and Hepato-Biliary-Pancreatic Surgery, Nippon Medical School, Tokyo, 113-8602, Japan
| | - Yoshio Naomoto
- Department of General Surgery, Kawasaki Medical School, Okayama, 700-8505, Japan
| | - Ajay Goel
- Department of Molecular Diagnostics and Experimental Therapeutics, Beckman Research Institute of City of Hope, Biomedical Research Center, Monrovia, CA, 91016, USA.
| | - Takeshi Nagasaka
- Department of Clinical Oncology, Kawasaki Medical School, 577 Matsushim, Kurashiki, 701-0912, Japan.
| |
Collapse
|
16
|
Melton CA, Freese P, Zhou Y, Shenoy A, Bagaria S, Chang C, Kuo CC, Scott E, Srinivasan S, Cann G, Roychowdhury-Saha M, Chang PY, Singh AH. A Novel Tissue-Free Method to Estimate Tumor-Derived Cell-Free DNA Quantity Using Tumor Methylation Patterns. Cancers (Basel) 2023; 16:82. [PMID: 38201510 PMCID: PMC10777919 DOI: 10.3390/cancers16010082] [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: 11/07/2023] [Revised: 12/07/2023] [Accepted: 12/20/2023] [Indexed: 01/12/2024] Open
Abstract
Estimating the abundance of cell-free DNA (cfDNA) fragments shed from a tumor (i.e., circulating tumor DNA (ctDNA)) can approximate tumor burden, which has numerous clinical applications. We derived a novel, broadly applicable statistical method to quantify cancer-indicative methylation patterns within cfDNA to estimate ctDNA abundance, even at low levels. Our algorithm identified differentially methylated regions (DMRs) between a reference database of cancer tissue biopsy samples and cfDNA from individuals without cancer. Then, without utilizing matched tissue biopsy, counts of fragments matching the cancer-indicative hyper/hypo-methylated patterns within DMRs were used to determine a tumor methylated fraction (TMeF; a methylation-based quantification of the circulating tumor allele fraction and estimate of ctDNA abundance) for plasma samples. TMeF and small variant allele fraction (SVAF) estimates of the same cancer plasma samples were correlated (Spearman's correlation coefficient: 0.73), and synthetic dilutions to expected TMeF of 10-3 and 10-4 had estimated TMeF within two-fold for 95% and 77% of samples, respectively. TMeF increased with cancer stage and tumor size and inversely correlated with survival probability. Therefore, tumor-derived fragments in the cfDNA of patients with cancer can be leveraged to estimate ctDNA abundance without the need for a tumor biopsy, which may provide non-invasive clinical approximations of tumor burden.
Collapse
|
17
|
Helal C, Pobel C, Bayle A, Vasseur D, Nicotra C, Blanc-Durand F, Naoun N, Bernard-Tessier A, Patrikidou A, Colomba E, Flippot R, Fuerea A, Auger N, Ngo Camus M, Besse B, Lacroix L, Rouleau E, Ponce S, Italiano A, Loriot Y. Clinical utility of plasma ctDNA sequencing in metastatic urothelial cancer. Eur J Cancer 2023; 195:113368. [PMID: 37897866 DOI: 10.1016/j.ejca.2023.113368] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2023] [Revised: 09/22/2023] [Accepted: 09/26/2023] [Indexed: 10/30/2023]
Abstract
BACKGROUND Genomic stratification may help improve the management of patients with metastatic urothelial cancer (mUC), given the recent identification of targetable alterations. However, the collection of tissue samples remains challenging. Here, we assessed the clinical utility of plasma circulating tumour DNA (ctDNA) sequencing in these patients. METHODS Patients with mUC were prospectively enroled in the STING trial (NCT04932525), in which ctDNA was profiled using the Foundation One Liquid CDx Assay (324 genes, blood tumour mutational burden [bTMB], microsatellite instability status). Each genomic report was reviewed by a multidisciplinary tumor board (MTB). RESULTS Between January 2021 and June 2022, 140 mUC patients underwent molecular profiling. The median time to obtain the assay results was 20 days ((confidence interval) CI95%: [20,21]). The ctDNA analysis reproduced the somatic genomic landscape of previous tissue-based cohorts. Concordance for serial ctDNA samples was strong (r = 0.843 CI95%: [0.631-0.938], p < 0.001). At least one actionable target was detected in 63 patients (45%) with a total of 35 actionable alterations, including bTMB high (≥10 mutations/Mb) (N = 39, 21.1%), FGFR3 (N = 20, 10.8%), and Homologous recombination deficiency (HRD) alterations (N = 14, 7.6%). MTB recommended matched therapy in 63 patients (45.0%). Eight patients (5.7%) were treated, with an overall response rate of 50% (CI95%: 15.70-84.30) and a median progression-free survival (PFS) of 5.2 months (CI95%: 4.1 - NR). FGFR3 alterations were associated with a shorter PFS in patients treated with immunotherapy. CONCLUSION Overall, we demonstrated that genomic profiling with ctDNAs in mUC is a reliable and feasible approach for the timely initiation of genotype-matched therapies.
Collapse
Affiliation(s)
- Clara Helal
- Sorbonne University, Paris, France; Département de médecine oncologique, Gustave Roussy, université Paris-Saclay, Villejuif, France
| | | | - Arnaud Bayle
- INSERM U981, Gustave Roussy, Villejuif, France; Drug Development Department (DITEP), Gustave Roussy, Université Paris-Saclay, Villejuif, France
| | - Damien Vasseur
- Department of Pathology and Laboratory Medicine, Translational Research Laboratory and Biobank, Gustave Roussy, Université Paris-Saclay, Villejuif, France; AMMICA, INSERM US23/CNRS UMS3655,Gustave Roussy, Université Paris-Saclay, Villejuif, France
| | - Claudio Nicotra
- Drug Development Department (DITEP), Gustave Roussy, Université Paris-Saclay, Villejuif, France
| | - Félix Blanc-Durand
- Drug Development Department (DITEP), Gustave Roussy, Université Paris-Saclay, Villejuif, France
| | - Natacha Naoun
- Département de médecine oncologique, Gustave Roussy, université Paris-Saclay, Villejuif, France
| | - Alice Bernard-Tessier
- Département de médecine oncologique, Gustave Roussy, université Paris-Saclay, Villejuif, France; Drug Development Department (DITEP), Gustave Roussy, Université Paris-Saclay, Villejuif, France
| | - Anna Patrikidou
- Département de médecine oncologique, Gustave Roussy, université Paris-Saclay, Villejuif, France
| | - Emeline Colomba
- Département de médecine oncologique, Gustave Roussy, université Paris-Saclay, Villejuif, France
| | - Ronan Flippot
- Département de médecine oncologique, Gustave Roussy, université Paris-Saclay, Villejuif, France
| | - Alina Fuerea
- Département de médecine oncologique, Gustave Roussy, université Paris-Saclay, Villejuif, France
| | - Nathalie Auger
- Département de médecine oncologique, Gustave Roussy, université Paris-Saclay, Villejuif, France
| | - Maud Ngo Camus
- Drug Development Department (DITEP), Gustave Roussy, Université Paris-Saclay, Villejuif, France
| | - Benjamin Besse
- Département de médecine oncologique, Gustave Roussy, université Paris-Saclay, Villejuif, France
| | - Ludovic Lacroix
- Département de médecine oncologique, Gustave Roussy, université Paris-Saclay, Villejuif, France
| | - Etienne Rouleau
- Département de médecine oncologique, Gustave Roussy, université Paris-Saclay, Villejuif, France
| | - Santiago Ponce
- Drug Development Department (DITEP), Gustave Roussy, Université Paris-Saclay, Villejuif, France
| | - Antoine Italiano
- Drug Development Department (DITEP), Gustave Roussy, Université Paris-Saclay, Villejuif, France
| | - Yohann Loriot
- Département de médecine oncologique, Gustave Roussy, université Paris-Saclay, Villejuif, France; INSERM U981, Gustave Roussy, Villejuif, France; Drug Development Department (DITEP), Gustave Roussy, Université Paris-Saclay, Villejuif, France.
| |
Collapse
|
18
|
Keup C, Kimmig R, Kasimir-Bauer S. The Diversity of Liquid Biopsies and Their Potential in Breast Cancer Management. Cancers (Basel) 2023; 15:5463. [PMID: 38001722 PMCID: PMC10670968 DOI: 10.3390/cancers15225463] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2023] [Revised: 11/10/2023] [Accepted: 11/14/2023] [Indexed: 11/26/2023] Open
Abstract
Analyzing blood as a so-called liquid biopsy in breast cancer (BC) patients has the potential to adapt therapy management. Circulating tumor cells (CTCs), extracellular vesicles (EVs), cell-free DNA (cfDNA) and other blood components mirror the tumoral heterogeneity and could support a range of clinical decisions. Multi-cancer early detection tests utilizing blood are advancing but are not part of any clinical routine yet. Liquid biopsy analysis in the course of neoadjuvant therapy has potential for therapy (de)escalation.Minimal residual disease detection via serial cfDNA analysis is currently on its way. The prognostic value of blood analytes in early and metastatic BC is undisputable, but the value of these prognostic biomarkers for clinical management is controversial. An interventional trial confirmed a significant outcome benefit when therapy was changed in case of newly emerging cfDNA mutations under treatment and thus showed the clinical utility of cfDNA analysis for therapy monitoring. The analysis of PIK3CA or ESR1 variants in plasma of metastatic BC patients to prescribe targeted therapy with alpesilib or elacestrant has already arrived in clinical practice with FDA-approved tests available and is recommended by ASCO. The translation of more liquid biopsy applications into clinical practice is still pending due to a lack of knowledge of the analytes' biology, lack of standards and difficulties in proving clinical utility.
Collapse
Affiliation(s)
- Corinna Keup
- Department of Gynecology and Obstetrics, University Hospital of Essen, 45147 Essen, Germany
| | | | | |
Collapse
|
19
|
Schrag D, Beer TM, McDonnell CH, Nadauld L, Dilaveri CA, Reid R, Marinac CR, Chung KC, Lopatin M, Fung ET, Klein EA. Blood-based tests for multicancer early detection (PATHFINDER): a prospective cohort study. Lancet 2023; 402:1251-1260. [PMID: 37805216 PMCID: PMC11027492 DOI: 10.1016/s0140-6736(23)01700-2] [Citation(s) in RCA: 129] [Impact Index Per Article: 64.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/04/2023] [Revised: 07/29/2023] [Accepted: 08/11/2023] [Indexed: 10/09/2023]
Abstract
BACKGROUND Multicancer early detection (MCED) blood tests can detect a cancer signal from circulating cell-free DNA (cfDNA). PATHFINDER was a prospective cohort study investigating the feasibility of MCED testing for cancer screening. METHODS In this prospective cohort study done in oncology and primary care outpatient clinics at seven US health networks, a convenience sample of adults aged 50 years or older without signs or symptoms of cancer consented to MCED testing. We collected blood, analysed cfDNA, and returned results to participants' doctors. If a methylation signature indicative of cancer was detected, predicted cancer signal origin(s) informed diagnostic assessment. The primary outcome was time to, and extent of, diagnostic testing required to confirm the presence or absence of cancer. This trial is registered at ClinicalTrials.gov, NCT04241796, and is completed. FINDINGS Between Dec 12, 2019, and Dec 4, 2020, we recruited 6662 participants. 4204 (63·5%) of 6621 participants with analysable results were women, 2417 (36·5%) were men, and 6071 (91·7%) were White. A cancer signal was detected in 92 (1·4%) of 6621 participants with analysable results. 35 (38%) participants were diagnosed with cancer (true positives) and 57 (62%) had no cancer diagnosis (false positives). Excluding two participants whose diagnostic assessments began before MCED test results were reported, median time to diagnostic resolution was 79 days (IQR 37-219): 57 days (33-143) in true-positive and 162 days (44-248) in false-positive participants. Most participants had both laboratory tests (26 [79%] of 33 with true-positive results and 50 [88%] of 57 with false-positive results) and imaging (30 [91%] of 33 with true-positive results and 53 [93%] of 57 with false-positive results). Fewer procedures were done in participants with false-positive results (17 [30%] of 57) than true-positive results (27 [82%] of 33) and few had surgery (one with a false-positive result and three with a true-positive result). INTERPRETATION This study supports the feasibility of MCED screening for cancer and underscores the need for further research investigating the test's clinical utility. FUNDING GRAIL.
Collapse
Affiliation(s)
- Deb Schrag
- Memorial Sloan Kettering Cancer Center, New York, NY, USA.
| | | | | | | | | | - Robert Reid
- US Oncology Research, VA Cancer Specialists, Fairfax, VA, USA
| | | | | | | | | | | |
Collapse
|
20
|
Chen TWW, Hsiao W, Dai MS, Lin CH, Chang DY, Chen IC, Wang MY, Chang SH, Huang SM, Cheng AL, Wu KW, Tan KT, Lu YS. Plasma cell-free tumor DNA, PIK3CA and TP53 mutations predicted inferior endocrine-based treatment outcome in endocrine receptor-positive metastatic breast cancer. Breast Cancer Res Treat 2023; 201:377-385. [PMID: 37344660 PMCID: PMC10460702 DOI: 10.1007/s10549-023-06967-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2023] [Accepted: 05/03/2023] [Indexed: 06/23/2023]
Abstract
PURPOSE How to factor both tumor burden and oncogenic genomic mutations as variables to predict the outcome of endocrine-based therapy (ET) in ER-positive/HER2-negative metastatic breast cancer patients (MBC) remains to be explored. METHOD Blood samples prospectively collected from 163 ER-positive/HER2-negative female MBC patients, before ET, were used for cell-free tumor DNA (cfDNA) analysis. cfDNA was subjected to next-generation sequencing (NGS) to interrogate oncogenic PIK3CA hotspot and TP53 DNA-binding domain (DBD) mutations, including single nucleotide variants (SNVs) or small insertions and deletions (InDels). The variant calling threshold was set at 0.5%. Progression-free survival (PFS) was measured from the start of the ET treatment to the time of disease progression of the same treatment regimen. RESULTS Overall, the median PFS was 8.3 months (95% CI 5.7-11.1 months). The median cfDNA was 38.5 ng (range 4.4-1935 ng). The proportion of patients with PIK3CA and TP53 alterations were 25.1 and 15.3%, respectively. Patients with high total cfDNA (HR 1.74, p = 0.003), PIK3CA mutation (HR 1.74, p = 0.007), and TP53 mutation (HR 1.64, p = 0.047) in liquid biopsy conferred worse outcome after ET. Even for patients with low tumor burden, the detrimental effect of PIK3CA or TP53 mutation remained significant (p < 0.001). For patients with either PIK3CA (p < 0.001) or TP53 mutation (p = 0.004), there was significant positive correlation between allele frequency (AF) and total cfDNA. CONCLUSION After adjustment of cfDNA level, PIK3CA and TP53 mutations observed in liquid biopsy exerted detrimental effects on the outcome of ET-based regimens. The AF of PIK3CA or TP53 may be a surrogate marker for PFS.
Collapse
Affiliation(s)
- Tom Wei-Wu Chen
- Department of Oncology, National Taiwan University Hospital, Taipei, Taiwan
- Graduate Institute of Oncology, National Taiwan University College of Medicine, Taipei, Taiwan
- Department of Medical Oncology, National Taiwan University Cancer Center, Taipei, Taiwan
| | - Wen Hsiao
- ACT Genomics Co., Ltd, Taipei, Taiwan
| | - Ming-Shen Dai
- Division of Hematology and Oncology, Department of Medicine, Tri-Service General Hospital, Taipei, Taiwan
| | - Ching-Hung Lin
- Department of Oncology, National Taiwan University Hospital, Taipei, Taiwan
- Department of Medical Oncology, National Taiwan University Cancer Center, Taipei, Taiwan
| | - Dwan-Ying Chang
- Department of Oncology, National Taiwan University Hospital, Taipei, Taiwan
- Department of Medical Oncology, National Taiwan University Cancer Center, Taipei, Taiwan
| | - I-Chun Chen
- Department of Oncology, National Taiwan University Hospital, Taipei, Taiwan
- Graduate Institute of Oncology, National Taiwan University College of Medicine, Taipei, Taiwan
- Department of Medical Oncology, National Taiwan University Cancer Center, Taipei, Taiwan
| | - Ming-Yang Wang
- Department of Surgery, National Taiwan University Cancer Center, Taipei, Taiwan
| | - Shu-Han Chang
- Department of Oncology, National Taiwan University Hospital, Taipei, Taiwan
| | - Shu-Min Huang
- Department of Oncology, National Taiwan University Hospital, Taipei, Taiwan
| | - Ann-Lii Cheng
- Department of Oncology, National Taiwan University Hospital, Taipei, Taiwan
- Department of Medical Oncology, National Taiwan University Cancer Center, Taipei, Taiwan
| | - Ko-Wen Wu
- ACT Genomics Co., Ltd, Taipei, Taiwan
| | | | - Yen-Shen Lu
- Department of Oncology, National Taiwan University Hospital, Taipei, Taiwan.
- Department of Medical Oncology, National Taiwan University Cancer Center, Taipei, Taiwan.
| |
Collapse
|
21
|
Funderburk K, Bang-Christensen SR, Miller BF, Tan H, Margolin G, Petrykowska HM, Baugher C, Farney SK, Grimm SA, Jameel N, Holland DO, Altman NS, Elnitski L. Evaluating Stacked Methylation Markers for Blood-Based Multicancer Detection. Cancers (Basel) 2023; 15:4826. [PMID: 37835520 PMCID: PMC10571530 DOI: 10.3390/cancers15194826] [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: 08/02/2023] [Revised: 09/16/2023] [Accepted: 09/27/2023] [Indexed: 10/15/2023] Open
Abstract
The ability to detect several types of cancer using a non-invasive, blood-based test holds the potential to revolutionize oncology screening. We mined tumor methylation array data from the Cancer Genome Atlas (TCGA) covering 14 cancer types and identified two novel, broadly-occurring methylation markers at TLX1 and GALR1. To evaluate their performance as a generalized blood-based screening approach, along with our previously reported methylation biomarker, ZNF154, we rigorously assessed each marker individually or combined. Utilizing TCGA methylation data and applying logistic regression models within each individual cancer type, we found that the three-marker combination significantly increased the average area under the ROC curve (AUC) across the 14 tumor types compared to single markers (p = 1.158 × 10-10; Friedman test). Furthermore, we simulated dilutions of tumor DNA into healthy blood cell DNA and demonstrated increased AUC of combined markers across all dilution levels. Finally, we evaluated assay performance in bisulfite sequenced DNA from patient tumors and plasma, including early-stage samples. When combining all three markers, the assay correctly identified nine out of nine lung cancer plasma samples. In patient plasma from hepatocellular carcinoma, ZNF154 alone yielded the highest combined sensitivity and specificity values averaging 68% and 72%, whereas multiple markers could achieve higher sensitivity or specificity, but not both. Altogether, this study presents a comprehensive pipeline for the identification, testing, and validation of multi-cancer methylation biomarkers with a considerable potential for detecting a broad range of cancer types in patient blood samples.
Collapse
Affiliation(s)
- Karen Funderburk
- Translational and Functional Genomics Branch, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Sara R. Bang-Christensen
- Translational and Functional Genomics Branch, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Brendan F. Miller
- Translational and Functional Genomics Branch, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Hua Tan
- Translational and Functional Genomics Branch, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Gennady Margolin
- Translational and Functional Genomics Branch, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Hanna M. Petrykowska
- Translational and Functional Genomics Branch, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Catherine Baugher
- Translational and Functional Genomics Branch, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - S. Katie Farney
- Translational and Functional Genomics Branch, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Sara A. Grimm
- Integrative Bioinformatics Support Group, Biostatistics and Computational Biology Branch, National Institute of Environmental Health Sciences (NIEHS), National Institutes of Health, Research Triangle Park, Durham, NC 27709, USA
| | - Nader Jameel
- Translational and Functional Genomics Branch, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - David O. Holland
- Translational and Functional Genomics Branch, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Naomi S. Altman
- Department of Statistics, Pennsylvania State University, University Park, PA 16802, USA
| | - Laura Elnitski
- Translational and Functional Genomics Branch, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD 20892, USA
| |
Collapse
|
22
|
Ong WL, Lunca S, Morarasu S, Musina AM, Puscasu A, Iacob S, Iftincai I, Marinca A, Ivanov I, Roata CE, Velenciuc N, Dimofte G. Evaluation of Changes in Circulating Cell-Free DNA as an Early Predictor of Response to Chemoradiation in Rectal Cancer-A Pilot Study. MEDICINA (KAUNAS, LITHUANIA) 2023; 59:1742. [PMID: 37893461 PMCID: PMC10608193 DOI: 10.3390/medicina59101742] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/02/2023] [Revised: 09/22/2023] [Accepted: 09/26/2023] [Indexed: 10/29/2023]
Abstract
Background and Objectives: The objective of this study was to investigate quantitative changes in cell-free DNA (cfDNA) found in the bloodstream of patients with locally advanced rectal cancer who received neoadjuvant long-course chemoradiation, assuming a change in DNA fragments release during therapeutic stress. Materials and Methods: This was a prospective observational study that involved 49 patients who had three distinct pathologies requiring neoadjuvant chemoradiation: 18 patients with breast cancer, 18 patients with cervical cancer, and 13 patients with rectal cancer. Both breast cancer and cervical cancer patients were used as a control groups. Breast cancer patients were used as a control group as irradiation targeted healthy tissue after the tumor resection (R0), while cervical cancer patients were used as a control group to evaluate the effect of chemoradiation regarding cfDNA in a different setting (squamous cell carcinomas) and a different tumor burden. Rectal cancer patients were the study group, and were prospectively evaluated for a correlation between fragmentation of cfDNA and late response to chemoradiation. Blood samples were collected before the initiation of treatment and after the fifth radiation dose delivery. cfDNA was quantified in peripheral blood and compared with the patients' clinicopathological characteristics and tumor volume. Conclusion: Thirteen patients with locally advanced rectal cancer (T3/T4/N+/M0) were included in the study, and all of them had their samples analyzed. Eight were male (61.54%) and five were female (38.46%), with an average age of 70.85 years. Most of the patients had cT3 (53.85%) or cT4 (46.15%) tumors, and 92.31% had positive lymph nodes (N2-3). Of the thirteen patients, only six underwent surgery, and one of them achieved a pathological complete response (pCR). The mean size of the tumor was 122.60 mm3 [35.33-662.60 mm3]. No significant correlation was found between cfDNA, tumor volume, and tumor regression grade. cfDNA does not seem to predict response to neoadjuvant chemoradiotherapy and it is not correlated to tumor volume or tumor regression grade.
Collapse
Affiliation(s)
- Wee Liam Ong
- 2nd Department of Surgical Oncology, Regional Institute of Oncology (IRO), 700483 Iasi, Romania; (W.L.O.); (A.-M.M.); (A.P.); (S.I.); (C.E.R.); (N.V.); (G.D.)
| | - Sorinel Lunca
- 2nd Department of Surgical Oncology, Regional Institute of Oncology (IRO), 700483 Iasi, Romania; (W.L.O.); (A.-M.M.); (A.P.); (S.I.); (C.E.R.); (N.V.); (G.D.)
- Department of Surgery, University of Medicine and Pharmacy “Grigore T. Popa”, 700115 Iasi, Romania
| | - Stefan Morarasu
- 2nd Department of Surgical Oncology, Regional Institute of Oncology (IRO), 700483 Iasi, Romania; (W.L.O.); (A.-M.M.); (A.P.); (S.I.); (C.E.R.); (N.V.); (G.D.)
- Department of Surgery, University of Medicine and Pharmacy “Grigore T. Popa”, 700115 Iasi, Romania
| | - Ana-Maria Musina
- 2nd Department of Surgical Oncology, Regional Institute of Oncology (IRO), 700483 Iasi, Romania; (W.L.O.); (A.-M.M.); (A.P.); (S.I.); (C.E.R.); (N.V.); (G.D.)
- Department of Surgery, University of Medicine and Pharmacy “Grigore T. Popa”, 700115 Iasi, Romania
| | - Alina Puscasu
- 2nd Department of Surgical Oncology, Regional Institute of Oncology (IRO), 700483 Iasi, Romania; (W.L.O.); (A.-M.M.); (A.P.); (S.I.); (C.E.R.); (N.V.); (G.D.)
- Department of Surgery, University of Medicine and Pharmacy “Grigore T. Popa”, 700115 Iasi, Romania
| | - Stefan Iacob
- 2nd Department of Surgical Oncology, Regional Institute of Oncology (IRO), 700483 Iasi, Romania; (W.L.O.); (A.-M.M.); (A.P.); (S.I.); (C.E.R.); (N.V.); (G.D.)
- Department of Surgery, University of Medicine and Pharmacy “Grigore T. Popa”, 700115 Iasi, Romania
| | - Irina Iftincai
- Radiotherapy Department, Regional Institute of Oncology (IRO), 700483 Iasi, Romania; (I.I.); (A.M.)
| | - Andreea Marinca
- Radiotherapy Department, Regional Institute of Oncology (IRO), 700483 Iasi, Romania; (I.I.); (A.M.)
| | - Iuliu Ivanov
- “TRANSCEND” Centre for Fundamental Research and Experimental Development in Translational Medicine, Regional Institute of Oncology (IRO), 700483 Iasi, Romania;
| | - Cristian Ena Roata
- 2nd Department of Surgical Oncology, Regional Institute of Oncology (IRO), 700483 Iasi, Romania; (W.L.O.); (A.-M.M.); (A.P.); (S.I.); (C.E.R.); (N.V.); (G.D.)
- Department of Surgery, University of Medicine and Pharmacy “Grigore T. Popa”, 700115 Iasi, Romania
| | - Natalia Velenciuc
- 2nd Department of Surgical Oncology, Regional Institute of Oncology (IRO), 700483 Iasi, Romania; (W.L.O.); (A.-M.M.); (A.P.); (S.I.); (C.E.R.); (N.V.); (G.D.)
- Department of Surgery, University of Medicine and Pharmacy “Grigore T. Popa”, 700115 Iasi, Romania
| | - Gabriel Dimofte
- 2nd Department of Surgical Oncology, Regional Institute of Oncology (IRO), 700483 Iasi, Romania; (W.L.O.); (A.-M.M.); (A.P.); (S.I.); (C.E.R.); (N.V.); (G.D.)
- Department of Surgery, University of Medicine and Pharmacy “Grigore T. Popa”, 700115 Iasi, Romania
| |
Collapse
|
23
|
Medina JE, Dracopoli NC, Bach PB, Lau A, Scharpf RB, Meijer GA, Andersen CL, Velculescu VE. Cell-free DNA approaches for cancer early detection and interception. J Immunother Cancer 2023; 11:e006013. [PMID: 37696619 PMCID: PMC10496721 DOI: 10.1136/jitc-2022-006013] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/03/2023] [Indexed: 09/13/2023] Open
Abstract
Rapid advancements in the area of early cancer detection have brought us closer to achieving the goals of finding cancer early enough to treat or cure it, while avoiding harms of overdiagnosis. We evaluate progress in the development of early cancer detection tests in the context of the current principles for cancer screening. We review cell-free DNA (cfDNA)-based approaches using mutations, methylation, or fragmentomes for early cancer detection. Lastly, we discuss the challenges in demonstrating clinical utility of these tests before integration into routine clinical care.
Collapse
Affiliation(s)
- Jamie E Medina
- Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | | | | | - Anna Lau
- Delfi Diagnostics Inc, Baltimore, Maryland, USA
| | - Robert B Scharpf
- Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | | | | | | |
Collapse
|
24
|
Farhana A, Alsrhani A, Khan YS, Rasheed Z. Cancer Bioenergetics and Tumor Microenvironments-Enhancing Chemotherapeutics and Targeting Resistant Niches through Nanosystems. Cancers (Basel) 2023; 15:3836. [PMID: 37568652 PMCID: PMC10416858 DOI: 10.3390/cancers15153836] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2023] [Accepted: 07/16/2023] [Indexed: 08/13/2023] Open
Abstract
Cancer is an impending bottleneck in the advanced scientific workflow to achieve diagnostic, prognostic, and therapeutic success. Most cancers are refractory to conventional diagnostic and chemotherapeutics due to their limited targetability, specificity, solubility, and side effects. The inherent ability of each cancer to evolve through various genetic and epigenetic transformations and metabolic reprogramming underlies therapeutic limitations. Though tumor microenvironments (TMEs) are quite well understood in some cancers, each microenvironment differs from the other in internal perturbations and metabolic skew thereby impeding the development of appropriate diagnostics, drugs, vaccines, and therapies. Cancer associated bioenergetics modulations regulate TME, angiogenesis, immune evasion, generation of resistant niches and tumor progression, and a thorough understanding is crucial to the development of metabolic therapies. However, this remains a missing element in cancer theranostics, necessitating the development of modalities that can be adapted for targetability, diagnostics and therapeutics. In this challenging scenario, nanomaterials are modular platforms for understanding TME and achieving successful theranostics. Several nanoscale particles have been successfully researched in animal models, quite a few have reached clinical trials, and some have achieved clinical success. Nanoparticles exhibit an intrinsic capability to interact with diverse biomolecules and modulate their functions. Furthermore, nanoparticles can be functionalized with receptors, modulators, and drugs to facilitate specific targeting with reduced toxicity. This review discusses the current understanding of different theranostic nanosystems, their synthesis, functionalization, and targetability for therapeutic modulation of bioenergetics, and metabolic reprogramming of the cancer microenvironment. We highlight the potential of nanosystems for enhanced chemotherapeutic success emphasizing the questions that remain unanswered.
Collapse
Affiliation(s)
- Aisha Farhana
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, Jouf University, Sakaka 72388, Aljouf, Saudi Arabia
| | - Abdullah Alsrhani
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, Jouf University, Sakaka 72388, Aljouf, Saudi Arabia
| | - Yusuf Saleem Khan
- Department of Anatomy, College of Medicine, Jouf University, Sakaka 72388, Aljouf, Saudi Arabia
| | - Zafar Rasheed
- Department of Pathology, College of Medicine, Qassim University, P.O. Box 6655, Buraidah 51452, Qassim, Saudi Arabia
| |
Collapse
|
25
|
Cohen SA, Liu MC, Aleshin A. Practical recommendations for using ctDNA in clinical decision making. Nature 2023; 619:259-268. [PMID: 37438589 DOI: 10.1038/s41586-023-06225-y] [Citation(s) in RCA: 67] [Impact Index Per Article: 33.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2022] [Accepted: 05/16/2023] [Indexed: 07/14/2023]
Abstract
The continuous improvement in cancer care over the past decade has led to a gradual decrease in cancer-related deaths. This is largely attributed to improved treatment and disease management strategies. Early detection of recurrence using blood-based biomarkers such as circulating tumour DNA (ctDNA) is being increasingly used in clinical practice. Emerging real-world data shows the utility of ctDNA in detecting molecular residual disease and in treatment-response monitoring, helping clinicians to optimize treatment and surveillance strategies. Many studies have indicated ctDNA to be a sensitive and specific biomarker for recurrence. However, most of these studies are largely observational or anecdotal in nature, and peer-reviewed data regarding the use of ctDNA are mainly indication-specific. Here we provide general recommendations on the clinical utility of ctDNA and how to interpret ctDNA analysis in different treatment settings, especially in patients with solid tumours. Specifically, we provide an understanding around the implications, strengths and limitations of this novel biomarker and how to best apply the results in clinical practice.
Collapse
Affiliation(s)
- Stacey A Cohen
- Fred Hutchinson Cancer Center, Seattle, WA, USA.
- University of Washington, Seattle, WA, USA.
| | | | | |
Collapse
|
26
|
Sasieni P, Smittenaar R, Hubbell E, Broggio J, Neal RD, Swanton C. Modelled mortality benefits of multi-cancer early detection screening in England. Br J Cancer 2023; 129:72-80. [PMID: 37185463 PMCID: PMC10307803 DOI: 10.1038/s41416-023-02243-9] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2023] [Revised: 03/14/2023] [Accepted: 03/17/2023] [Indexed: 05/17/2023] Open
Abstract
BACKGROUND Screening programmes utilising blood-based multi-cancer early detection (MCED) tests, which can detect a shared cancer signal from any site in the body with a single, low false-positive rate, could reduce cancer burden through early diagnosis. METHODS A natural history ('interception') model of cancer was previously used to characterise potential benefits of MCED screening (based on published performance of an MCED test). We built upon this using a two-population survival model to account for an increased risk of death from cfDNA-detectable cancers relative to cfDNA-non-detectable cancers. We developed another model allowing some cancers to metastasise directly from stage I, bypassing intermediate tumour stages. We used incidence and survival-by-stage data from the National Cancer Registration and Analysis Service in England to estimate longer-term benefits to a cohort screened between ages 50-79 years. RESULTS Estimated late-stage and mortality reductions were robust to a range of assumptions. With the least favourable dwell (sojourn) time and cfDNA status hazard ratio assumptions, we estimated, among 100,000 screened individuals, 67 (17%) fewer cancer deaths per year corresponding to 2029 fewer deaths in those screened between ages 50-79 years. CONCLUSION Realising the potential benefits of MCED tests could substantially reduce late-stage cancer diagnoses and mortality.
Collapse
Affiliation(s)
- Peter Sasieni
- Comprehensive Cancer Centre, King's College London, Guy's Campus, Great Maze Pond, London, SE1 1UL, UK.
| | | | | | - John Broggio
- NHS Digital, 7 and 8 Wellington Place, Leeds, West Yorkshire, LS1 4AP, UK
| | - Richard D Neal
- Department of Health and Community Sciences, Faculty of Health and Life Sciences, University of Exeter, Exeter, UK
| | - Charles Swanton
- Cancer Research UK Lung Cancer Centre of Excellence, University College London Cancer Institute, London, WC1E 6DD, UK
- Cancer Evolution and Genome Instability Laboratory, Francis Crick Institute, London, NW1 1AT, UK
| |
Collapse
|
27
|
Hudnut AG, Hubbell E, Venn O, Church TR. Modeled residual current cancer risk after clinical investigation of a positive multicancer early detection test result. Cancer 2023; 129:2056-2063. [PMID: 36943898 DOI: 10.1002/cncr.34747] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2022] [Revised: 11/10/2022] [Accepted: 11/14/2022] [Indexed: 03/23/2023]
Abstract
BACKGROUND Positive results of a multi-cancer early detection (MCED) test require confirmatory diagnostic workup. Here, residual current cancer risk (RR) during the process of diagnostic resolution, including situations where the initial confirmatory test does not provide resolution, was modeled. METHODS A decision-tree framework was used to model conditional risk in a patient's journey through confirmatory diagnostic options and outcomes. The diagnostic journey assumed that cancer signal detection (a positive MCED test result) had already led to a transition from screening to diagnosis and began with an initial positive predictive value (PPV) from the positive result. Evaluation of a most probable (top) predicted cancer signal origin (CSO) and then a second-most probable predicted CSO followed. Under the assumption that the top- and second-predicted CSOs were each followed by a targeted confirmatory test, the RR was estimated for each subsequent scenario. RESULTS For an initial MCED test result with typical performance characteristics modeled (PPV, 40%; top-predicted CSO accuracy, 90%), after a negative initial confirmatory test (sensitivity, 70%, 90%, or 100%) the RR ranged from 6% to 20%. A second-predicted CSO (accuracy, 50%), after a negative second confirmatory test, still provided a significant RR (3%-18%) in comparison with the National Institute for Health and Care Excellence-recommended cancer risk threshold warranting investigation in symptomatic individuals (3%). With a 40% PPV for an MCED test and 90% specificity for a confirmatory test, the risk of incidental findings after one or two confirmatory tests was 6% and 12%, respectively. CONCLUSIONS These results may illustrate the impact of a positive MCED test on follow-up decision-making.
Collapse
Affiliation(s)
- Andrew G Hudnut
- Family Medicine, Sutter Medical Group, Elk Grove, California, USA
| | - Earl Hubbell
- GRAIL, LLC, a subsidiary of Illumina, Inc., Menlo Park, California, USA
| | - Oliver Venn
- GRAIL, LLC, a subsidiary of Illumina, Inc., Menlo Park, California, USA
| | - Timothy R Church
- Division of Environmental Health Sciences, University of Minnesota, Minneapolis, Minnesota, USA
| |
Collapse
|
28
|
Shao SH, Allen B, Clement J, Chung G, Gao J, Hubbell E, Liu MC, Swanton C, Tang WW, Yimer H, Tummala M. Multi-cancer early detection test sensitivity for cancers with and without current population-level screening options. TUMORI JOURNAL 2023; 109:335-341. [PMID: 36316952 PMCID: PMC10248281 DOI: 10.1177/03008916221133136] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2022] [Revised: 09/23/2022] [Accepted: 09/26/2022] [Indexed: 11/18/2022]
Abstract
There are four solid tumors with common screening options in the average-risk population aged 21 to 75 years (breast, cervical, colorectal, and, based on personalized risk assessment, prostate), but many cancers lack recommended population screening and are often detected at advanced stages when mortality is high. Blood-based multi-cancer early detection tests have the potential to improve cancer mortality through additional population screening. Reported here is a post-hoc analysis from the third Circulating Cell-free Genome Atlas substudy to examine multi-cancer early detection test performance in solid tumors with and without population screening recommendations and in hematologic malignancies. Participants with cancer in the third Circulating Cell-free Genome Atlas substudy analysis were split into three subgroups: solid screened tumors (breast, cervical, colorectal, prostate), solid unscreened tumors, and hematologic malignancies. In this post hoc analysis, sensitivity is reported for each subgroup across all ages and those aged ⩾50 years overall, by cancer, and by clinical cancer stage. Aggregate sensitivity in the solid screened, solid unscreened, and hematologic malignancy subgroups was 34%, 66%, and 55% across all cancer stages, respectively; restricting to participants aged ⩾50 years showed similar aggregate sensitivity. Aggregate sensitivity was 27%, 53%, and 60% across stages I to III, respectively. Within the solid unscreened subgroup, aggregate sensitivity was >75% in 8/18 cancers (44%) and >50% in 13/18 (72%). This multi-cancer early detection test detected cancer signals at high (>75%) sensitivity for multiple cancers without existing population screening recommendations, suggesting its potential to complement recommended screening programs.Clinical trial identifier: NCT02889978.
Collapse
Affiliation(s)
| | - Brian Allen
- GRAIL, LLC, a subsidiary of Illumina Inc.,
Menlo Park, CA, USA†
| | | | - Gina Chung
- The Christ Hospital Health Network,
Cincinnati, OH, USA
| | - Jingjing Gao
- GRAIL, LLC, a subsidiary of Illumina Inc.,
Menlo Park, CA, USA†
| | - Earl Hubbell
- GRAIL, LLC, a subsidiary of Illumina Inc.,
Menlo Park, CA, USA†
| | | | - Charles Swanton
- The Francis Crick Institute, London, UK
- University College London Cancer Institute,
London, UK
| | | | | | | |
Collapse
|
29
|
Moser T, Kühberger S, Lazzeri I, Vlachos G, Heitzer E. Bridging biological cfDNA features and machine learning approaches. Trends Genet 2023; 39:285-307. [PMID: 36792446 DOI: 10.1016/j.tig.2023.01.004] [Citation(s) in RCA: 36] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2022] [Revised: 01/10/2023] [Accepted: 01/19/2023] [Indexed: 02/15/2023]
Abstract
Liquid biopsies (LBs), particularly using circulating tumor DNA (ctDNA), are expected to revolutionize precision oncology and blood-based cancer screening. Recent technological improvements, in combination with the ever-growing understanding of cell-free DNA (cfDNA) biology, are enabling the detection of tumor-specific changes with extremely high resolution and new analysis concepts beyond genetic alterations, including methylomics, fragmentomics, and nucleosomics. The interrogation of a large number of markers and the high complexity of data render traditional correlation methods insufficient. In this regard, machine learning (ML) algorithms are increasingly being used to decipher disease- and tissue-specific signals from cfDNA. Here, we review recent insights into biological ctDNA features and how these are incorporated into sophisticated ML applications.
Collapse
Affiliation(s)
- Tina Moser
- Institute of Human Genetics, Diagnostic & Research Center for Molecular BioMedicine, Medical University of Graz, Neue Stiftingtalstrasse 6, 8010 Graz, Austria; Christian Doppler Laboratory for Liquid Biopsies for Early Detection of Cancer, Medical University of Graz, Graz, Austria
| | - Stefan Kühberger
- Institute of Human Genetics, Diagnostic & Research Center for Molecular BioMedicine, Medical University of Graz, Neue Stiftingtalstrasse 6, 8010 Graz, Austria; Christian Doppler Laboratory for Liquid Biopsies for Early Detection of Cancer, Medical University of Graz, Graz, Austria
| | - Isaac Lazzeri
- Institute of Human Genetics, Diagnostic & Research Center for Molecular BioMedicine, Medical University of Graz, Neue Stiftingtalstrasse 6, 8010 Graz, Austria; Christian Doppler Laboratory for Liquid Biopsies for Early Detection of Cancer, Medical University of Graz, Graz, Austria
| | - Georgios Vlachos
- Institute of Human Genetics, Diagnostic & Research Center for Molecular BioMedicine, Medical University of Graz, Neue Stiftingtalstrasse 6, 8010 Graz, Austria; Christian Doppler Laboratory for Liquid Biopsies for Early Detection of Cancer, Medical University of Graz, Graz, Austria
| | - Ellen Heitzer
- Institute of Human Genetics, Diagnostic & Research Center for Molecular BioMedicine, Medical University of Graz, Neue Stiftingtalstrasse 6, 8010 Graz, Austria; Christian Doppler Laboratory for Liquid Biopsies for Early Detection of Cancer, Medical University of Graz, Graz, Austria.
| |
Collapse
|
30
|
Műzes G, Bohusné Barta B, Szabó O, Horgas V, Sipos F. Cell-Free DNA in the Pathogenesis and Therapy of Non-Infectious Inflammations and Tumors. Biomedicines 2022; 10:2853. [PMID: 36359370 PMCID: PMC9687442 DOI: 10.3390/biomedicines10112853] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2022] [Revised: 10/31/2022] [Accepted: 11/07/2022] [Indexed: 11/09/2022] Open
Abstract
The basic function of the immune system is the protection of the host against infections, along with the preservation of the individual antigenic identity. The process of self-tolerance covers the discrimination between self and foreign antigens, including proteins, nucleic acids, and larger molecules. Consequently, a broken immunological self-tolerance results in the development of autoimmune or autoinflammatory disorders. Immunocompetent cells express pattern-recognition receptors on their cell membrane and cytoplasm. The majority of endogenous DNA is located intracellularly within nuclei and mitochondria. However, extracellular, cell-free DNA (cfDNA) can also be detected in a variety of diseases, such as autoimmune disorders and malignancies, which has sparked interest in using cfDNA as a possible biomarker. In recent years, the widespread use of liquid biopsies and the increasing demand for screening, as well as monitoring disease activity and therapy response, have enabled the revival of cfDNA research. The majority of studies have mainly focused on the function of cfDNA as a biomarker. However, research regarding the immunological consequences of cfDNA, such as its potential immunomodulatory or therapeutic benefits, is still in its infancy. This article discusses the involvement of various DNA-sensing receptors (e.g., absent in melanoma-2; Toll-like receptor 9; cyclic GMP-AMP synthase/activator of interferon genes) in identifying host cfDNA as a potent danger-associated molecular pattern. Furthermore, we aim to summarize the results of the experimental studies that we recently performed and highlight the immunomodulatory capacity of cfDNA, and thus, the potential for possible therapeutic consideration.
Collapse
Affiliation(s)
| | | | | | | | - Ferenc Sipos
- Department of Internal Medicine and Hematology, Semmelweis University, Szentkirályi Street 46, 1088 Budapest, Hungary
| |
Collapse
|
31
|
Bredno J, Venn O, Chen X, Freese P, Ofman JJ. Circulating Tumor DNA Allele Fraction: A Candidate Biological Signal for Multicancer Early Detection Tests to Assess the Clinical Significance of Cancers. THE AMERICAN JOURNAL OF PATHOLOGY 2022; 192:1368-1378. [PMID: 35948080 DOI: 10.1016/j.ajpath.2022.07.007] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/24/2022] [Revised: 06/02/2022] [Accepted: 07/07/2022] [Indexed: 05/28/2023]
Abstract
Current imaging-based cancer screening approaches provide useful but limited prognostic information. Complementary to existing screening tests, cell-free DNA-based multicancer early detection (MCED) tests account for cancer biology [manifested through circulating tumor allele fraction (cTAF)], which could inform prognosis and help assess the cancer's clinical significance. This review discusses the factors affecting circulating tumor DNA (ctDNA) levels and cTAF, and their correlation with the cancer's clinical significance. Furthermore, it discusses the influence of cTAF on MCED test performance, which could help inform prognosis. Clinically significant cancers show higher ctDNA levels quantified by cTAF than indolent phenotype cancers within each stage. This is because more frequent mitosis and cell death combined with increased trafficking of cell-free DNA into circulation leads to greater vascularization and depth of tumor invasion. cTAF has been correlated with biomarkers for cancer aggressiveness and overall survival; cancers with lower cTAF had better survival when compared with cancers as determined by the higher cTAF and Surveillance, Epidemiology, and End Results-based survival for that cancer type at each stage. MCED-detected cancers in case-control studies had comparable survival to Surveillance, Epidemiology, and End Results-based survival at each stage. Because many MCED tests use ctDNA as an analyte, cTAF could provide a common metric to compare performance. The prognostic value of cTAF may allow MCED tests to preferentially detect clinically significant cancers at early stages when outcomes are favorable and this may avoid overdiagnosis.
Collapse
Affiliation(s)
- Joerg Bredno
- GRAIL, LLC, a subsidiary of Illumina, Inc., Menlo Park, California
| | - Oliver Venn
- GRAIL, LLC, a subsidiary of Illumina, Inc., Menlo Park, California.
| | - Xiaoji Chen
- GRAIL, LLC, a subsidiary of Illumina, Inc., Menlo Park, California
| | - Peter Freese
- GRAIL, LLC, a subsidiary of Illumina, Inc., Menlo Park, California
| | - Joshua J Ofman
- GRAIL, LLC, a subsidiary of Illumina, Inc., Menlo Park, California
| |
Collapse
|
32
|
Güttlein L, Luca MR, Esteso F, Fresno C, Mariani J, Otero Pizarro M, Brest E, Starapoli S, Kreimberg K, Teves P, Mendoza Bertelli A, R Girotti M, Salanova R, O'Connor JM. Liquid biopsy for KRAS, NRAS and BRAF mutation testing in advanced colorectal cancer patients: the Argentinean experience. Future Oncol 2022; 18:3277-3287. [PMID: 36004810 DOI: 10.2217/fon-2022-0329] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Objective: To analyze the frequency of KRAS, NRAS and BRAF hotspot mutations in circulating tumor DNA (ctDNA) from patients with metastatic colorectal cancer (mCRC). Methods: Observational, descriptive and retrospective study in mCRC patients with available ctDNA-based genotype of KRAS, NRAS and BRAF. Results: The frequencies of plasma mutations for KRAS, NRAS and BRAF were 34% (± 7), 4% (± 3) and 4% (± 3), respectively. Median overall survival of plasma-tested RAS/BRAF-mutated patients was 26.6 months (95% CI: 14.4-not estimable [NE]), while RAS/BRAF wild-type patients did not reach the median survival during follow-up. Median progression-free survival for RAS/BRAF wild-type and RAS/BRAF-mutated patients was 12 (95% CI: 7-NE) and 4 months (95% CI: 4-NE), respectively. Conclusion: Our work supports the utility of KRAS, NRAS and BRAF analysis in liquid biopsy from mCRC patients.
Collapse
Affiliation(s)
- Leandro Güttlein
- Molecular Biology Laboratory. Biomakers S.A. Av. Pueyrredón 1777. 9 Floor. Buenos Aires, C1119ACA, Argentina
| | - María R Luca
- Clinical Oncology, Instituto Alexander Fleming, Av Cramer 1180, Buenos Aires, C1426ANZ, Argentina
| | - Federico Esteso
- Clinical Oncology, Instituto Alexander Fleming, Av Cramer 1180, Buenos Aires, C1426ANZ, Argentina
| | | | - Javier Mariani
- Clinical Oncology, Instituto Alexander Fleming, Av Cramer 1180, Buenos Aires, C1426ANZ, Argentina
| | - Mercedes Otero Pizarro
- Molecular Biology Laboratory. Biomakers S.A. Av. Pueyrredón 1777. 9 Floor. Buenos Aires, C1119ACA, Argentina
| | - Esteban Brest
- Molecular Biology Laboratory. Biomakers S.A. Av. Pueyrredón 1777. 9 Floor. Buenos Aires, C1119ACA, Argentina
| | - Solange Starapoli
- Molecular Biology Laboratory. Biomakers S.A. Av. Pueyrredón 1777. 9 Floor. Buenos Aires, C1119ACA, Argentina
| | - Kevin Kreimberg
- Molecular Biology Laboratory. Biomakers S.A. Av. Pueyrredón 1777. 9 Floor. Buenos Aires, C1119ACA, Argentina
| | - Paula Teves
- Molecular Biology Laboratory. Biomakers S.A. Av. Pueyrredón 1777. 9 Floor. Buenos Aires, C1119ACA, Argentina
| | - Andrea Mendoza Bertelli
- Molecular Biology Laboratory. Biomakers S.A. Av. Pueyrredón 1777. 9 Floor. Buenos Aires, C1119ACA, Argentina
| | - María R Girotti
- Molecular Biology Laboratory. Biomakers S.A. Av. Pueyrredón 1777. 9 Floor. Buenos Aires, C1119ACA, Argentina
| | - Ruben Salanova
- Molecular Biology Laboratory. Biomakers S.A. Av. Pueyrredón 1777. 9 Floor. Buenos Aires, C1119ACA, Argentina
| | - Juan M O'Connor
- Clinical Oncology, Instituto Alexander Fleming, Av Cramer 1180, Buenos Aires, C1426ANZ, Argentina
| |
Collapse
|
33
|
Bodei L, Herrmann K, Schöder H, Scott AM, Lewis JS. Radiotheranostics in oncology: current challenges and emerging opportunities. Nat Rev Clin Oncol 2022; 19:534-550. [PMID: 35725926 PMCID: PMC10585450 DOI: 10.1038/s41571-022-00652-y] [Citation(s) in RCA: 164] [Impact Index Per Article: 54.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/20/2022] [Indexed: 12/20/2022]
Abstract
Structural imaging remains an essential component of diagnosis, staging and response assessment in patients with cancer; however, as clinicians increasingly seek to noninvasively investigate tumour phenotypes and evaluate functional and molecular responses to therapy, theranostics - the combination of diagnostic imaging with targeted therapy - is becoming more widely implemented. The field of radiotheranostics, which is the focus of this Review, combines molecular imaging (primarily PET and SPECT) with targeted radionuclide therapy, which involves the use of small molecules, peptides and/or antibodies as carriers for therapeutic radionuclides, typically those emitting α-, β- or auger-radiation. The exponential, global expansion of radiotheranostics in oncology stems from its potential to target and eliminate tumour cells with minimal adverse effects, owing to a mechanism of action that differs distinctly from that of most other systemic therapies. Currently, an enormous opportunity exists to expand the number of patients who can benefit from this technology, to address the urgent needs of many thousands of patients across the world. In this Review, we describe the clinical experience with established radiotheranostics as well as novel areas of research and various barriers to progress.
Collapse
Affiliation(s)
- Lisa Bodei
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Department of Radiology, Weill Cornell Medical School, New York, NY, USA
| | - Ken Herrmann
- German Cancer Consortium, University Hospital Essen, Essen, Germany
- Department of Nuclear Medicine, University of Duisburg-Essen, University Hospital Essen, Essen, Germany
| | - Heiko Schöder
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Department of Radiology, Weill Cornell Medical School, New York, NY, USA
| | - Andrew M Scott
- Tumour Targeting Laboratory, Olivia Newton-John Cancer Research Institute, Melbourne, Victoria, Australia
- Department of Molecular Imaging and Therapy, Austin Health, Melbourne, Victoria, Australia
- School of Cancer Medicine, La Trobe University, Melbourne, Victoria, Australia
- Department of Medicine, University of Melbourne, Melbourne, Victoria, Australia
| | - Jason S Lewis
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY, USA.
- Department of Radiology, Weill Cornell Medical School, New York, NY, USA.
- Molecular Pharmacology Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA.
- Department of Pharmacology, Weill Cornell Medical School, New York, NY, USA.
| |
Collapse
|
34
|
The Early Detection of Breast Cancer Using Liquid Biopsies: Model Estimates of the Benefits, Harms, and Costs. Cancers (Basel) 2022; 14:cancers14122951. [PMID: 35740615 PMCID: PMC9220983 DOI: 10.3390/cancers14122951] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2022] [Revised: 05/24/2022] [Accepted: 06/07/2022] [Indexed: 01/20/2023] Open
Abstract
Simple Summary Breast cancer screening is associated with benefits, such as mortality reduction and improved quality of life, and harms, such as false-positive results, overdiagnoses, and costs. Novel screen tests could be considered to reduce the harms and increase the benefits of screening. Liquid biopsies have been proposed as a novel method for the early detection of breast cancer. However, studies show that liquid biopsies based on cell-free DNA have a low sensitivity for early-stage breast cancer. Using the microsimulation model MISCAN-Fadia, we model the benefits, harms, and costs of the early detection of breast cancer using liquid biopsies for varying levels of liquid biopsy sensitivity and specificity. We found that liquid biopsies are unlikely to be an alternative to digital mammography, given the test performance based on a CCGA substudy. When liquid biopsies are unable to detect the precursor lesion of breast cancer—ductal carcinoma in situ (DCIS)—they need to be able to detect small, early-stage tumors, with high specificity, at low costs in order to be an alternative to digital mammography. We estimated a maximum liquid biopsy price of USD 187, which is substantially lower than currently listed prices. Abstract Breast cancer screening is associated with harms, such as false-positives and overdiagnoses, and, thus, novel screen tests can be considered. Liquid biopsies have been proposed as a novel method for the early detection of cancer, but low cell-free DNA tumor fraction might pose a problem for the use in population screening. Using breast cancer microsimulation model MISCAN-Fadia, we estimated the outcomes of using liquid biopsies in breast cancer screening in women aged 50 to 74 in the United States. For varying combinations of test sensitivity and specificity, we quantify the impact of the use of liquid biopsies on the harms and benefits of screening, and we estimate the maximum liquid biopsy price for cost-effective implementation in breast cancer screening at a cost-effectiveness threshold of USD 50,000. We investigate under what conditions liquid biopsies could be a suitable alternative to digital mammography and compare these conditions to a CCGA substudy. Outcomes were compared to digital mammography screening, and include mortality reduction, overdiagnoses, quality-adjusted life-years (QALYs), and the maximum price of a liquid biopsy for cost-effective implementation. When liquid biopsies are unable to detect DCIS, a large proportion of overdiagnosed cases is prevented but overall breast cancer mortality reduction and quality of life are lower, and costs are higher compared to digital mammography screening. Liquid biopsies prices should be restricted to USD 187 per liquid biopsy depending on test performance. Overall, liquid biopsies that are unable to detect ductal carcinoma in situ (DCIS) need to be able to detect small, early-stage tumors, with high specificity, at low costs in order to be an alternative to digital mammography. Liquid biopsies might be more suitable as an addition to digital mammography than as an alternative.
Collapse
|
35
|
Fernandez-Uriarte A, Pons-Belda OD, Diamandis EP. Cancer Screening Companies Are Rapidly Proliferating: Are They Ready for Business? Cancer Epidemiol Biomarkers Prev 2022; 31:1146-1150. [PMID: 35642390 DOI: 10.1158/1055-9965.epi-22-0102] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2022] [Revised: 02/16/2022] [Accepted: 03/14/2022] [Indexed: 11/16/2022] Open
Abstract
Cancer screening has been a major research front for decades. The classical circulating biomarkers for cancer (such as PSA, CEA, CA125, AFP, etc.) are neither sensitive nor specific and are not recommended for population screening. Recently, circulating tumor DNA (ctDNA) emerged as a new pan-cancer tumor marker, with much promise for clinical applicability. ctDNA released by tumor cells can be used as a proxy of the tumor burden and molecular composition. It has been hypothesized that if ctDNA is extracted from plasma and analyzed for genetic changes, it may form the basis for a non-invasive cancer detection test. Lately, there has been a proliferation of "for-profit" companies that will soon offer cancer screening services. Here, we comment on Grail, Thrive, Guardant, Delfi, and Freenome. Previously, we identified some fundamental difficulties associated with this new technology. In addition, clinical trials are exclusively case-control studies. The sensitivities/specificities/predictive values of the new screening tests have not been well-defined or, the literature-reported values are rather poor. Despite these deficiencies some of the aforementioned companies are already testing patients. We predict that the premature use of ctDNA as a cancer screening tool may add another disappointment in the long history of this field.
Collapse
Affiliation(s)
| | - Oscar D Pons-Belda
- Department of Pathology and Laboratory Medicine, Mount Sinai Hospital, Toronto, Canada
| | - Eleftherios P Diamandis
- Department of Pathology and Laboratory Medicine, Mount Sinai Hospital, Toronto, Canada.,Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Canada.,Department of Clinical Biochemistry, University Health Network, Toronto, Canada
| |
Collapse
|
36
|
Multi Cancer Early Detection by Using Circulating Tumor DNA—The Galleri Test. Reply to Klein et al. The Promise of Multicancer Early Detection. Comment on “Pons-Belda et al. Can Circulating Tumor DNA Support a Successful Screening Test for Early Cancer Detection? The Grail Paradigm. Diagnostics 2021, 11, 2171”. Diagnostics (Basel) 2022; 12:diagnostics12051244. [PMID: 35626399 PMCID: PMC9141547 DOI: 10.3390/diagnostics12051244] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2022] [Revised: 05/09/2022] [Accepted: 05/11/2022] [Indexed: 02/04/2023] Open
Abstract
We recently published some concerns with new technologies which are based on circulating tumor DNA (ctDNA) for early cancer detection. Most of our published criticism, including a commentary in this journal, has focused on tests developed by the biotechnology company GRAIL (their commercial product is also known as The Galleri Test). Scientists from GRAIL provided explanations and rebuttals to our criticism. They also posed some questions. Here, we reiterate our position and provide rebuttals, explanations and answers to these questions. We believe that constructive scientific debates, like this one, can profoundly contribute to advancements in scientific fields such as early cancer detection.
Collapse
|
37
|
Klein EA, Beer TM, Seiden M. The Promise of Multicancer Early Detection. Comment on Pons-Belda et al. Can Circulating Tumor DNA Support a Successful Screening Test for Early Cancer Detection? The Grail Paradigm. Diagnostics 2021, 11, 2171. Diagnostics (Basel) 2022; 12:diagnostics12051243. [PMID: 35626398 PMCID: PMC9141107 DOI: 10.3390/diagnostics12051243] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2022] [Revised: 04/16/2022] [Accepted: 05/09/2022] [Indexed: 12/20/2022] Open
Abstract
Multicancer Early Detection (MCED) represents a new and exciting paradigm for the early detection of cancer, which is the leading cause of death worldwide. Current screening tests, recommended for only five cancer types (breast, lung, colon, cervical, and prostate), are limited by a lack of complete adherence to guideline-based use and by the fact that they have cumulative high false positive rates. MCED tests agnostically detect cancer signals in the blood with good sensitivity and low false positive rates, can predict the cancer site of origin with high accuracy, can detect highly lethal cancers that have no current screening tests, and promise to improve cancer screening by improving efficiency and reducing the overall number needed to screen. Herein we outline this promise and clarify several published misconceptions about this field.
Collapse
Affiliation(s)
- Eric A. Klein
- Glickman Urological and Kidney Institute, Cleveland Clinic Lerner College of Medicine, 9500 Euclid Ave, Q10-1, Cleveland, OH 44195, USA
- Correspondence:
| | - Tomasz M. Beer
- Knight Cancer Institute, Oregon Health & Science University, CH-14R, 3303 SW Bond Ave, Portland, OR 97239, USA;
| | | |
Collapse
|
38
|
Pich O, Bailey C, Watkins TBK, Zaccaria S, Jamal-Hanjani M, Swanton C. The translational challenges of precision oncology. Cancer Cell 2022; 40:458-478. [PMID: 35487215 DOI: 10.1016/j.ccell.2022.04.002] [Citation(s) in RCA: 45] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/12/2022] [Revised: 03/16/2022] [Accepted: 04/05/2022] [Indexed: 12/11/2022]
Abstract
The translational challenges in the field of precision oncology are in part related to the biological complexity and diversity of this disease. Technological advances in genomics have facilitated large sequencing efforts and discoveries that have further supported this notion. In this review, we reflect on the impact of these discoveries on our understanding of several concepts: cancer initiation, cancer prevention, early detection, adjuvant therapy and minimal residual disease monitoring, cancer drug resistance, and cancer evolution in metastasis. We discuss key areas of focus for improving cancer outcomes, from biological insights to clinical application, and suggest where the development of these technologies will lead us. Finally, we discuss practical challenges to the wider adoption of molecular profiling in the clinic and the need for robust translational infrastructure.
Collapse
Affiliation(s)
- Oriol Pich
- Cancer Evolution and Genome Instability Laboratory, The Francis Crick Institute, London, UK
| | - Chris Bailey
- Cancer Evolution and Genome Instability Laboratory, The Francis Crick Institute, London, UK
| | - Thomas B K Watkins
- Cancer Evolution and Genome Instability Laboratory, The Francis Crick Institute, London, UK
| | - Simone Zaccaria
- Cancer Research UK Lung Cancer Centre of Excellence, University College London Cancer Institute, London, UK; Computational Cancer Genomics Research Group, University College London Cancer Institute, London, UK
| | - Mariam Jamal-Hanjani
- Cancer Research UK Lung Cancer Centre of Excellence, University College London Cancer Institute, London, UK; Cancer Metastasis Laboratory, University College London Cancer Institute, London, UK; Department of Medical Oncology, University College London Hospitals, London, UK
| | - Charles Swanton
- Cancer Evolution and Genome Instability Laboratory, The Francis Crick Institute, London, UK.
| |
Collapse
|
39
|
Walsh K, Raghavachari N, Kerr C, Bick AG, Cummings SR, Druley T, Dunbar CE, Genovese G, Goodell MA, Jaiswal S, Maciejewski J, Natarajan P, Shindyapina AV, Shuldiner AR, Van Den Akker EB, Vijg J. Clonal Hematopoiesis Analyses in Clinical, Epidemiologic, and Genetic Aging Studies to Unravel Underlying Mechanisms of Age-Related Dysfunction in Humans. FRONTIERS IN AGING 2022; 3:841796. [PMID: 35821803 PMCID: PMC9261374 DOI: 10.3389/fragi.2022.841796] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/22/2021] [Accepted: 02/07/2022] [Indexed: 11/13/2022]
Abstract
Aging is characterized by increased mortality, functional decline, and exponential increases in the incidence of diseases such as cancer, stroke, cardiovascular disease, neurological disease, respiratory disease, etc. Though the role of aging in these diseases is widely accepted and considered to be a common denominator, the underlying mechanisms are largely unknown. A significant age-related feature observed in many population cohorts is somatic mosaicism, the detectable accumulation of somatic mutations in multiple cell types and tissues, particularly those with high rates of cell turnover (e.g., skin, liver, and hematopoietic cells). Somatic mosaicism can lead to the development of cellular clones that expand with age in otherwise normal tissues. In the hematopoietic system, this phenomenon has generally been referred to as "clonal hematopoiesis of indeterminate potential" (CHIP) when it applies to a subset of clones in which mutations in driver genes of hematologic malignancies are found. Other mechanisms of clonal hematopoiesis, including large chromosomal alterations, can also give rise to clonal expansion in the absence of conventional CHIP driver gene mutations. Both types of clonal hematopoiesis (CH) have been observed in studies of animal models and humans in association with altered immune responses, increased mortality, and disease risk. Studies in murine models have found that some of these clonal events are involved in abnormal inflammatory and metabolic changes, altered DNA damage repair and epigenetic changes. Studies in long-lived individuals also show the accumulation of somatic mutations, yet at this advanced age, carriership of somatic mutations is no longer associated with an increased risk of mortality. While it remains to be elucidated what factors modify this genotype-phenotype association, i.e., compensatory germline genetics, cellular context of the mutations, protective effects to diseases at exceptional age, it points out that the exceptionally long-lived are key to understand the phenotypic consequences of CHIP mutations. Assessment of the clinical significance of somatic mutations occurring in blood cell types for age-related outcomes in human populations of varied life and health span, environmental exposures, and germline genetic risk factors will be valuable in the development of personalized strategies tailored to specific somatic mutations for healthy aging.
Collapse
Affiliation(s)
- Kenneth Walsh
- University of Virginia, Charlottesville, VA, United States
| | - Nalini Raghavachari
- National Institute on Aging, NIH, Bethesda, MD, United States,*Correspondence: Nalini Raghavachari,
| | - Candace Kerr
- National Institute on Aging, NIH, Bethesda, MD, United States
| | | | - Steven R. Cummings
- University of California, San Francisco, San Francisco, CA, United States
| | - Todd Druley
- Angle Biosciences, St. Louis, MO, United States
| | - Cynthia E. Dunbar
- National Heart, Lung and Blood Institute, NIH, Bethesda, MD, United States
| | | | | | | | | | | | | | | | | | - Jan Vijg
- Department of Biomedical Data Sciences, Leiden University Medical Center, Leiden, Netherlands
| |
Collapse
|
40
|
Rich NE, Singal AG. Overdiagnosis of hepatocellular carcinoma: Prevented by guidelines? Hepatology 2022; 75:740-753. [PMID: 34923659 PMCID: PMC8844206 DOI: 10.1002/hep.32284] [Citation(s) in RCA: 36] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/10/2021] [Revised: 11/27/2021] [Accepted: 12/04/2021] [Indexed: 12/13/2022]
Abstract
Overdiagnosis refers to detection of disease that would not otherwise become clinically apparent during a patient's lifetime. Overdiagnosis is common and has been reported for several cancer types, although there are few studies describing its prevalence in HCC surveillance programs. Overdiagnosis can have serious negative consequences including overtreatment and associated complications, financial toxicity, and psychological harms related to being labeled with a cancer diagnosis. Overdiagnosis can occur for several different reasons including inaccurate diagnostic criteria, detection of premalignant or very early malignant lesions, detection of indolent tumors, and competing risks of mortality. The risk of overdiagnosis is partly mitigated, albeit not eliminated, by several guideline recommendations, including definitions for the at-risk population in whom surveillance should be performed, surveillance modalities, surveillance interval, recall procedures, and HCC diagnostic criteria. Continued research is needed to further characterize the burden and trends of overdiagnosis as well as identify strategies to reduce overdiagnosis in the future.
Collapse
Affiliation(s)
- Nicole E Rich
- Division of Digestive and Liver Diseases, UT Southwestern Medical Center, Dallas, Texas, USA
| | - Amit G Singal
- Division of Digestive and Liver Diseases, UT Southwestern Medical Center, Dallas, Texas, USA
| |
Collapse
|
41
|
Abouali H, Hosseini SA, Purcell E, Nagrath S, Poudineh M. Recent Advances in Device Engineering and Computational Analysis for Characterization of Cell-Released Cancer Biomarkers. Cancers (Basel) 2022; 14:288. [PMID: 35053452 PMCID: PMC8774172 DOI: 10.3390/cancers14020288] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2021] [Revised: 12/21/2021] [Accepted: 01/04/2022] [Indexed: 02/04/2023] Open
Abstract
During cancer progression, tumors shed different biomarkers into the bloodstream, including circulating tumor cells (CTCs), extracellular vesicles (EVs), circulating cell-free DNA (cfDNA), and circulating tumor DNA (ctDNA). The analysis of these biomarkers in the blood, known as 'liquid biopsy' (LB), is a promising approach for early cancer detection and treatment monitoring, and more recently, as a means for cancer therapy. Previous reviews have discussed the role of CTCs and ctDNA in cancer progression; however, ctDNA and EVs are rapidly evolving with technological advancements and computational analysis and are the subject of enormous recent studies in cancer biomarkers. In this review, first, we introduce these cell-released cancer biomarkers and briefly discuss their clinical significance in cancer diagnosis and treatment monitoring. Second, we present conventional and novel approaches for the isolation, profiling, and characterization of these markers. We then investigate the mathematical and in silico models that are developed to investigate the function of ctDNA and EVs in cancer progression. We convey our views on what is needed to pave the way to translate the emerging technologies and models into the clinic and make the case that optimized next-generation techniques and models are needed to precisely evaluate the clinical relevance of these LB markers.
Collapse
Affiliation(s)
- Hesam Abouali
- Department of Electrical and Computer Engineering, University of Waterloo, Waterloo, ON N2L 3G1, Canada; (H.A.); (S.A.H.)
| | - Seied Ali Hosseini
- Department of Electrical and Computer Engineering, University of Waterloo, Waterloo, ON N2L 3G1, Canada; (H.A.); (S.A.H.)
| | - Emma Purcell
- Department of Chemical Engineering, University of Michigan, Ann Arbor, MI 48109-2800, USA; (E.P.); (S.N.)
| | - Sunitha Nagrath
- Department of Chemical Engineering, University of Michigan, Ann Arbor, MI 48109-2800, USA; (E.P.); (S.N.)
| | - Mahla Poudineh
- Department of Electrical and Computer Engineering, University of Waterloo, Waterloo, ON N2L 3G1, Canada; (H.A.); (S.A.H.)
| |
Collapse
|
42
|
Can Circulating Tumor DNA Support a Successful Screening Test for Early Cancer Detection? The Grail Paradigm. Diagnostics (Basel) 2021; 11:diagnostics11122171. [PMID: 34943407 PMCID: PMC8700281 DOI: 10.3390/diagnostics11122171] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2021] [Revised: 11/05/2021] [Accepted: 11/16/2021] [Indexed: 01/02/2023] Open
Abstract
Circulating tumor DNA (ctDNA) is a new pan-cancer tumor marker with important applications for patient prognosis, monitoring progression, and assessing the success of the therapeutic response. Another important goal is an early cancer diagnosis. There is currently a debate if ctDNA can be used for early cancer detection due to the small tumor burden and low mutant allele fraction (MAF). We compare our previous calculations on the size of detectable cancers by ctDNA analysis with the latest experimental data from Grail’s clinical trial. Current ctDNA-based diagnostic methods could predictably detect tumors of sizes greater than 10–15 mm in diameter. When tumors are of this size or smaller, their MAF is about 0.01% (one tumor DNA molecule admixed with 10,000 normal DNA molecules). The use of 10 mL of blood (4 mL of plasma) will likely contain less than a complete cancer genome, thus rendering the diagnosis of cancer impossible. Grail’s new data confirm the low sensitivity for early cancer detection (<30% for Stage I–II tumors, <20% for Stage I tumors), but specificity was high at 99.5%. According to these latest data, the sensitivity of the Grail test is less than 20% in Stage I disease, casting doubt if this test could become a viable pan-cancer clinical screening tool.
Collapse
|