1
|
Senore C, Doubeni C, Guittet L. FIT as a Comparator for Evaluating the Effectiveness of New Non-invasive CRC Screening Test. Dig Dis Sci 2025; 70:1625-1636. [PMID: 39560807 DOI: 10.1007/s10620-024-08718-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/06/2024] [Accepted: 10/24/2024] [Indexed: 11/20/2024]
Abstract
Randomized Controlled Trials (RCT) demonstrated that guaiac-based fecal occult blood test (gFOBT), sigmoidoscopy, or colonoscopy are effective at reducing colorectal cancer (CRC) risk and mortality. Even if the impact of fecal immunochemical test (FIT) has not been evaluated within population-based RCT with mortality as the outcome, the results of comparative analyses with gFOBT provide strong indirect evidence of its effectiveness. Extensive information is also available on sensitivity and specificity of FIT, compared with gFOBT. FIT has almost universally replaced gFOBT in organized screening programs worldwide. Using FIT as a comparator is an efficient way to evaluate the effectiveness of new tests, with respect to test performance and relevant intermediate outcomes such as rates of interval cancer and late-stage cancer incidence. Direct comparison with FIT in the pre-screening evaluation of the accuracy of the new test will guide selection of the cut-off of the new test, and document the potential gain in sensitivity. Comparison in cross-sectional single-round screening evaluation can either use paired or parallel designs. Only parallel designs allow direct comparisons of participation rates. Relative accuracy can be derived in both designs with an assumption of similar colorectal cancer and precursor prevalence between groups in parallel designs. Finally, multiple-rounds prospective comparison will document potential effect on risk of CRC and precancerous lesions, and on absolute reductions in late-stage incidence as a proxy of mortality. This paper provides an overview of evidence and rationale for using FIT as a comparator for evaluating new non-invasive tests with repeated testing at short intervals.
Collapse
Affiliation(s)
- Carlo Senore
- Epidemiology and Screening Unit, CPO, University hospital Città della Salute e della Scienza, Turin, Italy
| | - Chyke Doubeni
- Department of Family and Community Medicine, Comprehensive Cancer Center-The James Cancer Hospital, The Ohio State University Wexner Medical Center, Columbus, OH, USA
| | - Lydia Guittet
- CHU Caen Normandie, INSERM ANTICIPE U1086, Université Caen Normandie, 14000, Caen, France.
| |
Collapse
|
2
|
Zhang Z, Liu X, Peng C, Du R, Hong X, Xu J, Chen J, Li X, Tang Y, Li Y, Liu Y, Xu C, Liu D. Machine Learning-Aided Identification of Fecal Extracellular Vesicle microRNA Signatures for Noninvasive Detection of Colorectal Cancer. ACS NANO 2025; 19:10013-10025. [PMID: 40033785 DOI: 10.1021/acsnano.4c16698] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/05/2025]
Abstract
Colorectal cancer (CRC) remains a formidable threat to human health, with considerable challenges persisting in its diagnosis, particularly during the early stages of the malignancy. In this study, we elucidated that fecal extracellular vesicle microRNA signatures (FEVOR) could serve as potent noninvasive CRC biomarkers. FEVOR was first revealed by miRNA sequencing, followed by the construction of a CRISPR/Cas13a-based detection platform to interrogate FEVOR expression across a diverse spectrum of clinical cohorts. Machine learning-driven models were subsequently developed within the realms of CRC diagnostics, prognostics, and early warning systems. In a cohort of 38 CRC patients, our diagnostic model achieved an outstanding accuracy of 97.4% (37/38), successfully identifying 37 of 38 CRC cases. This performance significantly outpaced the diagnostic efficacy of two clinically established biomarkers, CEA and CA19-9, which showed accuracies of mere 26.3% (10/38) and 7.9% (3/38), respectively. We also examined the expression levels of FEVOR in several CRC patients both before and after surgery, as well as in patients with colorectal adenomas (CA). Impressively, the results showed that FEVOR could serve as a robust prognostic indicator for CRC and a potential predictor for CA. This endeavor aimed to harness the predictive power of FEVOR for enhancing the precision and efficacy of CRC management paradigms. We envision that these findings will propel both foundational and preclinical research on CRC, as well as clinical studies.
Collapse
Affiliation(s)
- Zhaowei Zhang
- State Key Laboratory of Medicinal Chemical Biology, Tianjin Key Laboratory of Molecular Recognition and Biosensing, Frontiers Science Center for New Organic Matter, College of Chemistry, Nankai University, Tianjin 300071, China
| | - Xuyang Liu
- State Key Laboratory of Medicinal Chemical Biology, Tianjin Key Laboratory of Molecular Recognition and Biosensing, Frontiers Science Center for New Organic Matter, College of Chemistry, Nankai University, Tianjin 300071, China
| | - Chuanyue Peng
- Department of Colorectal Surgery, Tianjin Union Medical Center, Tianjin Institute of Coloproctology, School of Medicine, Nankai University, Tianjin 300071, China
| | - Rui Du
- State Key Laboratory of Medicinal Chemical Biology, Tianjin Key Laboratory of Molecular Recognition and Biosensing, Frontiers Science Center for New Organic Matter, College of Chemistry, Nankai University, Tianjin 300071, China
| | - Xiaoqin Hong
- State Key Laboratory of Medicinal Chemical Biology, Tianjin Key Laboratory of Molecular Recognition and Biosensing, Frontiers Science Center for New Organic Matter, College of Chemistry, Nankai University, Tianjin 300071, China
| | - Jia Xu
- State Key Laboratory of Medicinal Chemical Biology, Tianjin Key Laboratory of Molecular Recognition and Biosensing, Frontiers Science Center for New Organic Matter, College of Chemistry, Nankai University, Tianjin 300071, China
| | - Jiaming Chen
- State Key Laboratory of Medicinal Chemical Biology, Tianjin Key Laboratory of Molecular Recognition and Biosensing, Frontiers Science Center for New Organic Matter, College of Chemistry, Nankai University, Tianjin 300071, China
| | - Xiaomin Li
- Medical and Hygienic Materials Research Institute, SINOPEC (Beijing) Research Institute of Chemical Industry Co., Ltd., Beijing 100013, China
| | - Yujing Tang
- Medical and Hygienic Materials Research Institute, SINOPEC (Beijing) Research Institute of Chemical Industry Co., Ltd., Beijing 100013, China
| | - Yuwei Li
- Department of Colorectal Surgery, Tianjin Union Medical Center, Tianjin Institute of Coloproctology, School of Medicine, Nankai University, Tianjin 300071, China
| | - Yang Liu
- Department of Hepatobiliary Cancer, Liver Cancer Center, Tianjin Medical University Cancer Institute & Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, Tianjin 300060, China
- Department of Hepatobiliary and Pancreatic Oncology, Tianjin Cancer Hospital Airport Hospital, National Clinical Research Center for Cancer, Tianjin 300308, China
| | - Chen Xu
- Department of Colorectal Surgery, Tianjin Union Medical Center, Tianjin Institute of Coloproctology, School of Medicine, Nankai University, Tianjin 300071, China
| | - Dingbin Liu
- State Key Laboratory of Medicinal Chemical Biology, Tianjin Key Laboratory of Molecular Recognition and Biosensing, Frontiers Science Center for New Organic Matter, College of Chemistry, Nankai University, Tianjin 300071, China
| |
Collapse
|
3
|
Zheng H, Liu Z, Chen Y, Ji P, Fang Z, He Y, Guo C, Xiao P, Wang C, Yin W, Li F, Chen X, Liu M, Pan Y, Liu F, Liu Y, He Z, Ke Y. Development and external validation of a quantitative diagnostic model for malignant gastric lesions in clinical opportunistic screening: A multicenter real-world study. Chin Med J (Engl) 2024; 137:2343-2350. [PMID: 38403900 PMCID: PMC11441920 DOI: 10.1097/cm9.0000000000002903] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2023] [Indexed: 02/27/2024] Open
Abstract
BACKGROUND Clinical opportunistic screening is a cost-effective cancer screening modality. This study aimed to establish an easy-to-use diagnostic model serving as a risk stratification tool for identification of individuals with malignant gastric lesions for opportunistic screening. METHODS We developed a questionnaire-based diagnostic model using a joint dataset including two clinical cohorts from northern and southern China. The cohorts consisted of 17,360 outpatients who had undergone upper gastrointestinal endoscopic examination in endoscopic clinics. The final model was derived based on unconditional logistic regression, and predictors were selected according to the Akaike information criterion. External validation was carried out with 32,614 participants from a community-based randomized controlled trial. RESULTS This questionnaire-based diagnostic model for malignant gastric lesions had eight predictors, including advanced age, male gender, family history of gastric cancer, low body mass index, unexplained weight loss, consumption of leftover food, consumption of preserved food, and epigastric pain. This model showed high discriminative power in the development set with an area under the receiver operating characteristic curve (AUC) of 0.791 (95% confidence interval [CI]: 0.750-0.831). External validation of the model in the general population generated an AUC of 0.696 (95% CI: 0.570-0.822). This model showed an ideal ability for enriching prevalent malignant gastric lesions when applied to various scenarios. CONCLUSION This easy-to-use questionnaire-based model for diagnosis of prevalent malignant gastric lesions may serve as an effective prescreening tool in clinical opportunistic screening for gastric cancer.
Collapse
Affiliation(s)
- Hongchen Zheng
- State Key Laboratory of Molecular Oncology, Beijing Key Laboratory of Carcinogenesis and Translational Research, Department of Genetics, Peking University Cancer Hospital & Institute, Beijing 100142, China
| | - Zhen Liu
- State Key Laboratory of Molecular Oncology, Beijing Key Laboratory of Carcinogenesis and Translational Research, Department of Genetics, Peking University Cancer Hospital & Institute, Beijing 100142, China
| | - Yun Chen
- Department of Ultrasound, Peking University Shenzhen Hospital, Shenzhen, Guangdong 516473, China
- Shenzhen Key Laboratory for Drug Addiction and Medication Safety, Shenzhen Peking University-Hong Kong University of Science and Technology Medical Center, Shenzhen, Guangdong 516473, China
| | - Ping Ji
- Clinical Research Institute, Shenzhen Peking University-Hong Kong University of Science and Technology Medical Center, Shenzhen, Guangdong 516473, China
| | - Zhengyu Fang
- Clinical Research Institute, Shenzhen Peking University-Hong Kong University of Science and Technology Medical Center, Shenzhen, Guangdong 516473, China
| | - Yujie He
- Endoscopy Center, Hua County People's Hospital, Anyang, Henan 456483, China
| | - Chuanhai Guo
- State Key Laboratory of Molecular Oncology, Beijing Key Laboratory of Carcinogenesis and Translational Research, Department of Genetics, Peking University Cancer Hospital & Institute, Beijing 100142, China
| | - Ping Xiao
- Clinical Research Institute, Shenzhen Peking University-Hong Kong University of Science and Technology Medical Center, Shenzhen, Guangdong 516473, China
| | - Chengwen Wang
- Endoscope Group, Department of Gastroenterology, Peking University Shenzhen Hospital, Shenzhen, Guangdong 516473, China
| | - Weihua Yin
- Department of Pathology, Peking University Shenzhen Hospital, Shenzhen, Guangdong 516473, China
| | - Fenglei Li
- Hua County People's Hospital, Anyang, Henan 456483, China
| | - Xiujian Chen
- Department of Pathology, Hua County People's Hospital, Anyang, Henan 456483, China
| | - Mengfei Liu
- State Key Laboratory of Molecular Oncology, Beijing Key Laboratory of Carcinogenesis and Translational Research, Department of Genetics, Peking University Cancer Hospital & Institute, Beijing 100142, China
| | - Yaqi Pan
- State Key Laboratory of Molecular Oncology, Beijing Key Laboratory of Carcinogenesis and Translational Research, Department of Genetics, Peking University Cancer Hospital & Institute, Beijing 100142, China
| | - Fangfang Liu
- State Key Laboratory of Molecular Oncology, Beijing Key Laboratory of Carcinogenesis and Translational Research, Department of Genetics, Peking University Cancer Hospital & Institute, Beijing 100142, China
| | - Ying Liu
- State Key Laboratory of Molecular Oncology, Beijing Key Laboratory of Carcinogenesis and Translational Research, Department of Genetics, Peking University Cancer Hospital & Institute, Beijing 100142, China
| | - Zhonghu He
- State Key Laboratory of Molecular Oncology, Beijing Key Laboratory of Carcinogenesis and Translational Research, Department of Genetics, Peking University Cancer Hospital & Institute, Beijing 100142, China
| | - Yang Ke
- State Key Laboratory of Molecular Oncology, Beijing Key Laboratory of Carcinogenesis and Translational Research, Department of Genetics, Peking University Cancer Hospital & Institute, Beijing 100142, China
| |
Collapse
|
4
|
Fu SW, Tang C, Tan X, Srivastava S. Liquid biopsy for early cancer detection: technological revolutions and clinical dilemma. Expert Rev Mol Diagn 2024; 24:937-955. [PMID: 39360748 DOI: 10.1080/14737159.2024.2408744] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2024] [Accepted: 09/22/2024] [Indexed: 10/05/2024]
Abstract
INTRODUCTION Liquid biopsy is an innovative advancement in oncology, offering a noninvasive method for early cancer detection and monitoring by analyzing circulating tumor cells, DNA, RNA, and other biomarkers in bodily fluids. This technique has the potential to revolutionize precision oncology by providing real-time analysis of tumor dynamics, enabling early detection, monitoring treatment responses, and tailoring personalized therapies based on the molecular profiles of individual patients. AREAS COVERED In this review, the authors discuss current methodologies, technological challenges, and clinical applications of liquid biopsy. This includes advancements in detecting minimal residual disease, tracking tumor evolution, and combining liquid biopsy with other diagnostic modalities for precision oncology. Key areas explored are the sensitivity, specificity, and integration of multi-omics, AI, ML, and LLM technologies. EXPERT OPINION Liquid biopsy holds great potential to revolutionize cancer care through early detection and personalized treatment strategies. However, its success depends on overcoming technological and clinical hurdles, such as ensuring high sensitivity and specificity, interpreting results amidst tumor heterogeneity, and making tests accessible and affordable. Continued innovation and collaboration are crucial to fully realize the potential of liquid biopsy in improving early cancer detection, treatment, and monitoring.
Collapse
Affiliation(s)
- Sidney W Fu
- Division of Cancer Prevention, National Cancer Institute, National Institutes of Health, Rockville, MD, USA
| | - Cong Tang
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Lisbon, Portugal
| | - Xiaohui Tan
- Division of LS Research, LSBioscience, LLC, Frederick, USA
| | - Sudhir Srivastava
- Cancer Biomarkers Research Group, Division of Cancer Prevention, National Cancer Institute, National Institutes of Health, Rockville, MD, USA
| |
Collapse
|
5
|
Eng C, Yoshino T, Ruíz-García E, Mostafa N, Cann CG, O'Brian B, Benny A, Perez RO, Cremolini C. Colorectal cancer. Lancet 2024; 404:294-310. [PMID: 38909621 DOI: 10.1016/s0140-6736(24)00360-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/09/2023] [Revised: 02/15/2024] [Accepted: 02/21/2024] [Indexed: 06/25/2024]
Abstract
Despite decreased incidence rates in average-age onset patients in high-income economies, colorectal cancer is the third most diagnosed cancer in the world, with increasing rates in emerging economies. Furthermore, early onset colorectal cancer (age ≤50 years) is of increasing concern globally. Over the past decade, research advances have increased biological knowledge, treatment options, and overall survival rates. The increase in life expectancy is attributed to an increase in effective systemic therapy, improved treatment selection, and expanded locoregional surgical options. Ongoing developments are focused on the role of sphincter preservation, precision oncology for molecular alterations, use of circulating tumour DNA, analysis of the gut microbiome, as well as the role of locoregional strategies for colorectal cancer liver metastases. This overview is to provide a general multidisciplinary perspective of clinical advances in colorectal cancer.
Collapse
Affiliation(s)
- Cathy Eng
- Division of Hematology and Oncology, Vanderbilt University Medical Center, Vanderbilt-Ingram Cancer Center, Nashville, TN, USA.
| | - Takayuki Yoshino
- Department of Gastroenterology and Gastrointestinal Oncology, Cancer Center Hospital East, Kashiwa, Japan
| | - Erika Ruíz-García
- Department of Gastrointestinal Tumors and Translational Medicine Laboratory, Instituto Nacional de Cancerologia, Mexico City, Mexico
| | | | - Christopher G Cann
- Department of Hematology/Oncology, Fox Chase Cancer Center, Philadelphia, PA, USA
| | - Brittany O'Brian
- Division of Hematology and Oncology, Vanderbilt University Medical Center, Vanderbilt-Ingram Cancer Center, Nashville, TN, USA
| | - Amala Benny
- Division of Hematology and Oncology, Vanderbilt University Medical Center, Vanderbilt-Ingram Cancer Center, Nashville, TN, USA
| | | | - Chiara Cremolini
- Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Pisa, Italy
| |
Collapse
|
6
|
Zhang K, Fu R, Liu R, Su Z. Circulating cell-free DNA-based multi-cancer early detection. Trends Cancer 2024; 10:161-174. [PMID: 37709615 DOI: 10.1016/j.trecan.2023.08.010] [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: 07/03/2023] [Revised: 08/03/2023] [Accepted: 08/21/2023] [Indexed: 09/16/2023]
Abstract
Patients benefit considerably from early detection of cancer. Existing single-cancer tests have various limitations, which could be effectively addressed by circulating cell-free DNA (cfDNA)-based multi-cancer early detection (MCED). With sensitive detection and accurate localization of multiple cancer types at a very low and fixed false-positive rate (FPR), MCED has great potential to revolutionize early cancer detection. Herein, we review state-of-the-art approaches for cfDNA-based MCED and their limitations and discuss both technical and clinical challenges in the development and application of MCED tests. Given the constant improvements in technology and understanding of cancer biology, we propose that a cfDNA-based targeted sequencing assay that integrates multimodal features should be optimized for MCED.
Collapse
Affiliation(s)
- Kai Zhang
- Department of Cancer Prevention, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, 17 South Panjiayuan Lane, Chaoyang District, Beijing 100021, China
| | - Ruiqing Fu
- Singlera Genomics Ltd, Shanghai 201203, China
| | - Rui Liu
- Singlera Genomics Ltd, Shanghai 201203, China
| | - Zhixi Su
- Singlera Genomics Ltd, Shanghai 201203, China.
| |
Collapse
|
7
|
Parisi C, Tagliamento M, Belcaid L, Aldea M, Bayle A, Remon-Masip J, Italiano A, Planchard D, Besse B, Barlesi F. Circulating tumor DNA in clinical trials for solid tumors: Challenges and current applications. THE JOURNAL OF LIQUID BIOPSY 2023; 1:100007. [PMID: 40027283 PMCID: PMC11863815 DOI: 10.1016/j.jlb.2023.100007] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/24/2023] [Revised: 08/27/2023] [Accepted: 08/27/2023] [Indexed: 03/05/2025]
Abstract
Tumor derived biomarkers including circulating tumor DNA (ctDNA) and/or circulating tumors cells (CTCs) may be detected and quantified through liquid biopsy (LB). ctDNA analysis through LB is a validated tool for monitoring response to systemic treatment and detecting molecular mechanisms of resistance at the time of progression of advanced stage malignancies. Several applications of ctDNA have been investigated in the diagnostic phase of cancer or in the post-curative treatment surveillance phase (e.g., minimal residual disease assessment after neoadjuvant or adjuvant therapy). Recently, the improvement of ctDNA technology and its implementation have affected early phase trials design, with significant changes in the inclusion and randomization phases. Implementation of LB has resulted in large-scale development of academic programs aimed at exploiting all the potential applications of ctDNA, such as patients extended molecular screening, molecular oriented treatment decision making, monitoring of anti-cancer treatments response. In this rapid evolving field, the challenge is no longer the technique, but the evaluation of the results and the interpretation of their impact on diagnosis, prognosis, or therapeutic decision. Leading research cancer centers may favor education for scientific community, by capturing data on this evolving technology and sharing knowledge. In this review we summarize the main applications and challenges of ctDNA genotyping in clinical trials, with special focus on ongoing studies. We finally describe the most important next generation academic and industry-sponsored programs addressing early cancer detection and prevention in high-risk populations through ctDNA genotyping.
Collapse
Affiliation(s)
- Claudia Parisi
- Drug Development Department (DITEP), Gustave Roussy, Villejuif, France
- Department of Medical and Surgical Sciences and Translational Medicine, St Andrea University Hospital, Sapienza University, Rome, Italy
| | - Marco Tagliamento
- Department of Cancer Medicine, Gustave Roussy, Villejuif, France
- Department of Internal Medicine and Medical Specialties, University of Genova, Genova, Italy
| | - Laila Belcaid
- Department of Oncology, Copenaghen University Hospital, Rigshospitalet, Denmark
| | - Mihaela Aldea
- Department of Cancer Medicine, Gustave Roussy, Villejuif, France
- Paris-Saclay University, Orsay, France
| | - Arnaud Bayle
- Drug Development Department (DITEP), Gustave Roussy, Villejuif, France
| | | | - Antoine Italiano
- Drug Development Department (DITEP), Gustave Roussy, Villejuif, France
- Medical Oncology Department, Institute Bergonié, Bordeaux, France
| | - David Planchard
- Department of Cancer Medicine, Gustave Roussy, Villejuif, France
- Paris-Saclay University, Orsay, France
| | - Benjamin Besse
- Department of Cancer Medicine, Gustave Roussy, Villejuif, France
- Paris-Saclay University, Orsay, France
| | - Fabrice Barlesi
- Department of Cancer Medicine, Gustave Roussy, Villejuif, France
- Paris-Saclay University, Orsay, France
| |
Collapse
|
8
|
Wisse PHA, de Klaver W, van Wifferen F, Meiqari L, Bierkens M, Greuter MJE, Carvalho B, van Leerdam ME, Spaander MCW, Dekker E, Coupé VMH, de Wit M, Meijer GA. The multitarget fecal immunochemical test versus the fecal immunochemical test for programmatic colorectal cancer screening: a cross-sectional intervention study with paired design. BMC Cancer 2022; 22:1299. [PMID: 36503495 PMCID: PMC9743627 DOI: 10.1186/s12885-022-10372-2] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2022] [Accepted: 11/24/2022] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND Many screening programs for colorectal cancer (CRC) use the fecal immunochemical test (FIT) to triage individuals for colonoscopy. Although these programs reduce CRC incidence and CRC-related mortality, the detection of advanced precursor lesions (advanced adenomas and advanced serrated polyps) by FIT could be improved. As an alternative for FIT, the antibody-based multitargetFIT (mtFIT) has been proposed. The mtFIT measures three protein markers: hemoglobin, calprotectin, and serpin family F member 2. In a retrospective diagnostic accuracy study in a large colonoscopy-controlled series (n = 1284), mtFIT showed increased sensitivity for advanced neoplasia (AN), at equal specificity, compared to FIT (42.9% versus 37.3%; p = 0.025). This increase was mainly due to a higher sensitivity of mtFIT for advanced adenomas (37.8% versus 28.1% for FIT; p = 0.006). The present mtFIT study aims to prospectively validate these findings in the context of the Dutch national CRC screening program. METHOD The mtFIT study is a cross-sectional intervention study with a paired design. Eligible subjects for the Dutch FIT-based national CRC screening program are invited to perform mtFIT in addition to FIT. Samples are collected at home, from the same bowel movement, and are shipped to a central laboratory by postal mail. If either one or both tests are positive, participants are referred for colonoscopy. Detailed colonoscopy and pathology data are centrally stored in a national screening database (ScreenIT; Topicus, Deventer, the Netherlands) that is managed by the screening organization, and will be retrieved for this study. We aim to determine the relative sensitivity for AN, comprising of CRC, advanced adenomas and advanced serrated polyps, of mtFIT compared to FIT at an equal positivity rate. Additionally, we will use the Adenoma and Serrated Pathway to Colorectal CAncer model to predict lifetime health effects and costs for programmatic mtFIT- versus FIT-based screening. The target sample size is 13,131 participants. DISCUSSION The outcome of this study will inform on the comparative clinical utility of mtFIT versus FIT in the Dutch national CRC screening program and is an important step forward in the development of a new non-invasive stool test for CRC screening. TRIAL REGISTRATION Clinicaltrials.gov ; NCT05314309, registered April 6th 2022, first inclusions March 25th 2022 https://clinicaltrials.gov/ct2/results?cond=&term=NCT05314309&cntry=&state=&city=&dist =.
Collapse
Affiliation(s)
- P. H. A. Wisse
- grid.430814.a0000 0001 0674 1393Department of Pathology, Netherlands Cancer Institute, Plesmanlaan 121, Amsterdam, CX the Netherlands ,grid.5645.2000000040459992XDepartment of Gastroenterology and Hepatology, Erasmus University Medical Center, Doctor Molewaterplein 40, Rotterdam, GD 3015 the Netherlands
| | - W. de Klaver
- grid.5645.2000000040459992XDepartment of Gastroenterology and Hepatology, Erasmus University Medical Center, Doctor Molewaterplein 40, Rotterdam, GD 3015 the Netherlands ,grid.7177.60000000084992262Department of Gastroenterology and Hepatology, Amsterdam University Medical Centers, Location University of Amsterdam, Meibergdreef 9, Amsterdam, AZ 1105 the Netherlands
| | - F. van Wifferen
- grid.509540.d0000 0004 6880 3010Department of Epidemiology and Data Science, Amsterdam University Medical Centers, Location Vrije Universiteit, De Boelelaan 1117, Amsterdam, HV 1081 the Netherlands
| | - L. Meiqari
- grid.430814.a0000 0001 0674 1393Department of Pathology, Netherlands Cancer Institute, Plesmanlaan 121, Amsterdam, CX the Netherlands
| | - M. Bierkens
- grid.430814.a0000 0001 0674 1393Department of Pathology, Netherlands Cancer Institute, Plesmanlaan 121, Amsterdam, CX the Netherlands
| | - M. J. E. Greuter
- grid.509540.d0000 0004 6880 3010Department of Epidemiology and Data Science, Amsterdam University Medical Centers, Location Vrije Universiteit, De Boelelaan 1117, Amsterdam, HV 1081 the Netherlands
| | - B. Carvalho
- grid.430814.a0000 0001 0674 1393Department of Pathology, Netherlands Cancer Institute, Plesmanlaan 121, Amsterdam, CX the Netherlands
| | - M. E. van Leerdam
- grid.430814.a0000 0001 0674 1393Department of Gastro-intestinal Oncology, Netherlands Cancer Institute, Plesmanlaan 121, Amsterdam, CX 1066 the Netherlands
| | - M. C. W. Spaander
- grid.5645.2000000040459992XDepartment of Gastroenterology and Hepatology, Erasmus University Medical Center, Doctor Molewaterplein 40, Rotterdam, GD 3015 the Netherlands
| | - E. Dekker
- grid.7177.60000000084992262Department of Gastroenterology and Hepatology, Amsterdam University Medical Centers, Location University of Amsterdam, Meibergdreef 9, Amsterdam, AZ 1105 the Netherlands
| | - V. M. H. Coupé
- grid.509540.d0000 0004 6880 3010Department of Epidemiology and Data Science, Amsterdam University Medical Centers, Location Vrije Universiteit, De Boelelaan 1117, Amsterdam, HV 1081 the Netherlands
| | - M. de Wit
- grid.430814.a0000 0001 0674 1393Department of Pathology, Netherlands Cancer Institute, Plesmanlaan 121, Amsterdam, CX the Netherlands
| | - G. A. Meijer
- grid.430814.a0000 0001 0674 1393Department of Pathology, Netherlands Cancer Institute, Plesmanlaan 121, Amsterdam, CX the Netherlands
| |
Collapse
|
9
|
Peralta P, Hall MP, Singh Bhan S, Brown K, Parton MA, Yeshwant K, Finucane S, Keeling P, Ofman JJ. Industry engagement: Accelerating discovery, application, and adoption through industry partnerships. Cancer 2022; 128 Suppl 4:918-926. [PMID: 35133660 DOI: 10.1002/cncr.34041] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2021] [Revised: 10/08/2021] [Accepted: 10/12/2021] [Indexed: 11/11/2022]
Affiliation(s)
| | - Megan P Hall
- GRAIL, LLC, a subsidiary of Illumina, Inc, Menlo Park, California
| | | | - Kim Brown
- Thrive, an Exact Sciences Company, Cambridge, Massachusetts
| | | | | | | | | | - Joshua J Ofman
- GRAIL, LLC, a subsidiary of Illumina, Inc, Menlo Park, California
| |
Collapse
|