1
|
Jaber F, El-Serag HB. HES V2.0 surpasses GALAD for HCC detection: a review of multi-dimensional biomarker scores and studies. Hepat Oncol 2025; 12:2494446. [PMID: 40308043 PMCID: PMC12051611 DOI: 10.1080/20450923.2025.2494446] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2024] [Accepted: 04/14/2025] [Indexed: 05/02/2025] Open
Abstract
This was a narrative review of select studies published through September of 2024. We review the shift toward multi-dimensional scores such as HCC early detection screening (HES), GALAD, ASAP, and mt-HBT represents a significant advancement in biomarker research for hepatocellular carcinoma (HCC) detection. Unlike single biomarker approaches, these scores integrate various clinical and biochemical factors to enhance predictive accuracy by reflecting different complementary aspects of disease progression and HCC oncogenesis. Proper testing and validation of biomarker scores in phase 3 biomarker studies is essential before wide use can be recommended. We also review the comparative performance of biomarker scores in phase 3 studies. The new version of HES (HES V2.0) which includes AFP, AFP L3, DCP, and changes in their levels the past one year, if available, in addition to age, platelets, albumin, ALT and underlying liver disease etiology outperforms GALAD in detecting early-stage HCC with overall 6.7% higher sensitivity, and ASAP with 13.4%-18.0% higher sensitivity, both at fixed 90% specificity. HES V2.0 is a leading candidate biomarker score for prospective testing in clinical studies of early HCC detection.
Collapse
Affiliation(s)
- Fouad Jaber
- Department of Internal Medicine, Section of Gastroenterology and Hepatology, Baylor College of Medicine, Houston, Texas, USA
| | - Hashem B. El-Serag
- Department of Internal Medicine, Section of Gastroenterology and Hepatology, Baylor College of Medicine, Houston, Texas, USA
| |
Collapse
|
2
|
Liou WL, Tan SY, Yamada H, Krishnamoorthy T, Chang JPE, Yeo CP, Tan CK. Performance of the GALAD Model in an Asian Cohort Undergoing Hepatocellular Carcinoma Surveillance: A Prospective Cohort Study. J Gastroenterol Hepatol 2025. [PMID: 40346978 DOI: 10.1111/jgh.16997] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/18/2025] [Revised: 04/05/2025] [Accepted: 04/28/2025] [Indexed: 05/12/2025]
Abstract
BACKGROUND AND AIM Current hepatocellular carcinoma (HCC) surveillance strategy has its limitations, consequently delaying early detection. The GALAD model has been validated in retrospective studies, with two published cut-off values yielding different sensitivities for HCCs of different etiologies. We evaluated the performance of GALAD model in HCC surveillance and determined the ideal cut-off value for our cohort. METHODS Patients undergoing 6-monthly HCC surveillance in Singapore General Hospital were recruited between December 2017-October 2018. Study serum specimens were prospectively collected and retrospectively tested using the μTASWako alpha-fetoprotein (AFP), AFP-L3, and protein induced by vitamin K antagonism-II (PIVKA-II) kits. GALAD score was calculated and compared with individual biomarkers using area under the curve (AUC) analysis. Published GALAD cut-offs of -0.63 and -1.95 were compared for their performance in HCC detection. RESULTS There were 207 patients (median age 59 years, 55.1% males). Hepatitis B was the commonest etiology (72.9%). By February 2023, with a median follow-up of 48.9 months, 20 patients had developed HCC. Eight patients developed HCC within 1 year from specimen collection. For HCC developing within 1 year, GALAD model detected HCC with an AUC of 0.84, greater than AFP (AUC 0.77), AFP-L3 (AUC 0.60), and PIVKA-II (AUC 0.67). GALAD at cut-off -1.95 achieved sensitivity and specificity of 75% and 92.5% for HCCs detected within 1 year, superior to cut-off -0.63 (sensitivity 12.5%, specificity 100%). CONCLUSION In this prospective study of HCC surveillance, the GALAD model performed better than individual biomarkers. The cut-off of -1.95 was more useful in our predominantly chronic hepatitis B cohort.
Collapse
Affiliation(s)
- Wei-Lun Liou
- Department of Gastroenterology and Hepatology, Singapore General Hospital, Singapore
| | - Si-Yu Tan
- Department of Clinical Pathology, Singapore General Hospital, Singapore
| | - Hiroyuki Yamada
- Medical Systems Business Division, FUJIFILM Corporation, Osaka, Japan
| | | | - Jason Pik-Eu Chang
- Department of Gastroenterology and Hepatology, Singapore General Hospital, Singapore
| | - Chin-Pin Yeo
- Department of Clinical Pathology, Singapore General Hospital, Singapore
| | - Chee-Kiat Tan
- Department of Gastroenterology and Hepatology, Singapore General Hospital, Singapore
| |
Collapse
|
3
|
Tirkes T, Yadav D, Conwell DL, Zhao X, Dasyam AK, Halappa VG, Patel A, Shah ZK, Swensson J, Takahashi N, Venkatesh S, Wachsman A, Li L, Jennings K, Yang Y, Hart PA, Pandol SJ, Park WG, Vege SS, Topazian M, Territo PR, Persohn SA, Andersen DK, Fogel EL. Multiparametric MRI Scoring System of the Pancreas for the Diagnosis of Chronic Pancreatitis. J Magn Reson Imaging 2025; 61:2183-2194. [PMID: 39225586 PMCID: PMC11873175 DOI: 10.1002/jmri.29594] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2024] [Revised: 08/16/2024] [Accepted: 08/16/2024] [Indexed: 09/04/2024] Open
Abstract
BACKGROUND Ductal features alone may not offer high diagnostic sensitivity or most accurate disease severity of chronic pancreatitis (CP). PURPOSE Diagnose CP based on multiparametric MRI and MRCP features. STUDY TYPE Prospective. POPULATION Between February 2019 and May 2021, 46 control (23 males, 49.3 ± 14.1 years), 45 suspected (20 males, 48.7 ± 12.5 years), and 46 definite (20 males, 53.7 ± 14.6 years) CP patients were enrolled at seven hospitals enrolled in the MINIMAP study. CP classification was based on imaging findings and clinical presentation. FIELD STRENGTH AND SEQUENCES 1.5 T. T1-weighted (T1W) spoiled gradient echo, T1 map with variable flip angle, dual-echo Dixon, secretin-enhanced MRCP before and after secretin infusion. ASSESSMENT Dual-echo fat fraction (FF), T1 relaxation time, extracellular volume (ECV), T1 signal intensity ratio of the pancreas to the spleen (T1 score), arterial-to-venous enhancement ratio (AVR), pancreatic tail diameter (PTD), pancreas volume, late gadolinium enhancement, pancreatic ductal elasticity (PDE), and duodenal filling grade of secretin-enhanced MRCP were measured. STATISTICAL TESTS Logistic regression analysis generated CP-MRI and secretin-enhanced CP-SMRI scores. Receiver operating characteristics analysis was used to differentiate definite CP from control. Interobserver agreement was assessed using Lin's concordance correlation coefficient. RESULTS Compared to control, definite CP cohort showed significantly higher dual-echo FF (7% vs. 11%), lower AVR (1.35 vs. 0.85), smaller PTD (2.5 cm vs. 1.95 cm), higher ECV (28% vs. 38%), and higher incidence of PDE loss (6.5% vs. 50%). With the cut-off of >2.5 CP-MRI score (dual-echo FF, AVR, and PTD) and CP-SMRI score (dual-echo FF, AVR, PTD, and PDE) had cross-validated area under the curves of 0.84 (sensitivity 87%, specificity 68%) and 0.86 (sensitivity 89%, specificity 67%), respectively. Interobserver agreement for both CP-MRI and CP-SMRI scores was 0.74. CONCLUSION The CP-MRI and CP-SMRI scores yielded acceptable performance and interobserver agreement for the diagnosis of CP. EVIDENCE LEVEL 1 TECHNICAL EFFICACY: Stage 2.
Collapse
Grants
- U01DK108323 The Consortium for the Study of Chronic Pancreatitis, Diabetes, and Pancreatic Cancer
- U01 DK108288 NIDDK NIH HHS
- U01 DK108323 NIDDK NIH HHS
- U01DK108306 The Consortium for the Study of Chronic Pancreatitis, Diabetes, and Pancreatic Cancer
- U01DK108328 The Consortium for the Study of Chronic Pancreatitis, Diabetes, and Pancreatic Cancer
- U01DK108300 The Consortium for the Study of Chronic Pancreatitis, Diabetes, and Pancreatic Cancer
- U01DK108327 The Consortium for the Study of Chronic Pancreatitis, Diabetes, and Pancreatic Cancer
- U01 DK108327 NIDDK NIH HHS
- U01DK108288 The Consortium for the Study of Chronic Pancreatitis, Diabetes, and Pancreatic Cancer
- DKP3041301 The Consortium for the Study of Chronic Pancreatitis, Diabetes, and Pancreatic Cancer
- U01 DK108300 NIDDK NIH HHS
- R01 DK116963 NIDDK NIH HHS
- U01 DK108306 NIDDK NIH HHS
- U01 DK108328 NIDDK NIH HHS
- R01DK116963 NIDDK NIH HHS
- R01DK116963 NIDDK NIH HHS
- National Institute of Diabetes and Digestive and Kidney Diseases
Collapse
Affiliation(s)
- Temel Tirkes
- Department of Radiology and Imaging SciencesIndiana University School of MedicineIndianapolisIndianaUSA
| | - Dhiraj Yadav
- Division of Gastroenterology, Hepatology & Nutrition, Department of MedicineUniversity of Pittsburgh School of MedicinePittsburghPennsylvaniaUSA
| | - Darwin L. Conwell
- Department of Internal MedicineUniversity of Kentucky College of MedicineLexingtonKentuckyUSA
| | - Xuandong Zhao
- Department of Radiology and Imaging SciencesIndiana University School of MedicineIndianapolisIndianaUSA
| | - Anil K. Dasyam
- Department of RadiologyUniversity of Pittsburgh Medical CenterPittsburghPennsylvaniaUSA
| | - Vivek Gowdra Halappa
- Department of Radiology and Imaging SciencesIndiana University School of MedicineIndianapolisIndianaUSA
| | - Aashish Patel
- Department of Radiology and Imaging SciencesIndiana University School of MedicineIndianapolisIndianaUSA
| | - Zarine K. Shah
- Department of RadiologyThe Ohio State University Wexner Medical CenterColumbusOhioUSA
| | - Jordan Swensson
- Department of Radiology and Imaging SciencesIndiana University School of MedicineIndianapolisIndianaUSA
| | | | | | - Ashley Wachsman
- Department of Imaging, Cedars‐Sinai Medical CenterUniversity of California in Los AngelesLos AngelesCaliforniaUSA
| | - Liang Li
- Department of BiostatisticsThe University of Texas MD Anderson Cancer CenterHoustonTexasUSA
| | - Kristofer Jennings
- Department of BiostatisticsThe University of Texas MD Anderson Cancer CenterHoustonTexasUSA
| | - Yunlong Yang
- Department of BiostatisticsThe University of Texas MD Anderson Cancer CenterHoustonTexasUSA
| | - Phil A. Hart
- Division of Gastroenterology, Hepatology & NutritionThe Ohio State University Wexner Medical CenterColumbusOhioUSA
| | - Stephen J. Pandol
- Division of Digestive and Liver DiseasesCedars‐Sinai Medical CenterLos AngelesCaliforniaUSA
| | - Walter G. Park
- Division of Gastroenterology and Hepatology, Department of MedicineStanford University Medical CenterStanfordCaliforniaUSA
| | | | | | - Paul R. Territo
- Division of Clinical PharmacologyStark Neurosciences Research Institute, Indiana University School of MedicineIndianapolisIndianaUSA
| | - Scott A. Persohn
- Stark Neurosciences Research Institute, Indiana University School of MedicineIndianapolisIndianaUSA
| | - Dana K. Andersen
- Division of Digestive Diseases and NutritionNational Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of HealthBethesdaMarylandUSA
| | - Evan L. Fogel
- Lehman, Bucksot and Sherman Section of Pancreatobiliary EndoscopyIndiana University School of MedicineIndianapolisIndianaUSA
| | | |
Collapse
|
4
|
Seif El Dahan K, Yokoo T, Daher D, Davenport MS, Fetzer DT, Mendiratta-Lala M, Rich NE, Yang E, Parikh ND, Singal AG. Multicenter evaluation of abbreviated MRI and ultrasound for detecting early-stage hepatocellular carcinoma. JHEP Rep 2025; 7:101357. [PMID: 40321196 PMCID: PMC12048809 DOI: 10.1016/j.jhepr.2025.101357] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/06/2024] [Revised: 02/06/2025] [Accepted: 02/10/2025] [Indexed: 05/08/2025] Open
Abstract
Background & aims Abbreviated MRI (AMRI) has been proposed as an alternative to ultrasound for hepatocellular carcinoma (HCC) surveillance; however, comparative data for AMRI and ultrasound are needed. Thus, we evaluated the sensitivity and specificity of dynamic contrast-enhanced (DCE)-AMRI and ultrasound for early-stage HCC detection in patients with cirrhosis. Methods We conducted a multicenter retrospective case-control study among patients with cirrhosis (cases with early-stage HCC as per Milan Criteria; controls without HCC) who underwent an ultrasound and a DCE-MRI within a 6-month period between 2012 and 2019. HCC diagnosis was confirmed by imaging alone in 85% and by histopathology in 15% of patients. Dynamic AMRI examinations were simulated from the full MRI by selecting relevant sequences. Independent, blinded interpretations of ultrasounds and AMRI results were performed using Liver Imaging Reporting and Data System algorithms. Ultrasounds were considered positive if US-3 observations were detected. AMRI was considered positive if LR-4, LR-5, or LR-M were detected. Per-patient sensitivity and specificity for early-stage HCC detection were estimated, and cross-modality differences were tested. Results We included 216 cases and 432 controls. Patient-level sensitivity and specificity of AMRI were significantly higher compared with ultrasound: 80.1% (95% CI 76.1-83.6) vs. 71.1% (95% CI 66.6-75.2), p <0.001, and 91.9% (95% CI 89.9-93.5) vs. 72.3% (95% CI 69.3-75.2), p <0.001, respectively. AMRI sensitivity was significantly higher compared with ultrasound among patients with Child-Pugh B cirrhosis (80.8% vs. 57.4%, p <0.001) but not among those with Child-Pugh A (84.7% vs. 78.6%, p = 0.07) or Child-Pugh C cirrhosis (52.6% vs. 68.4%, p = 0.18). Conclusions Dynamic AMRI may be more sensitive and specific for early-stage HCC detection in patients with cirrhosis compared with ultrasound, although its relative benefit might be smaller in patients with Child-Pugh A cirrhosis. Larger direct comparative data sets are needed, particularly among patients with Child-Pugh C cirrhosis who may benefit from alternative surveillance strategies. Impact and implications Abbreviated MRI (AMRI) is increasingly recognized as an alternative to ultrasound for hepatocellular carcinoma (HCC) surveillance. However, existing data are limited by single-center samples, spectrum bias, and lack of comparative data for AMRI vs. ultrasound. We found that AMRI had significantly higher per-patient sensitivity and specificity compared with ultrasound for the detection of early-stage HCC, although its relative benefit might be smaller in patients with Child-Pugh A cirrhosis, and both modalities underperformed in patients with Child-Pugh C cirrhosis. If sufficiently validated, AMRI could be adopted into practice guidelines for HCC surveillance and serve as a preferred alternative in select subgroups of patients.
Collapse
Affiliation(s)
- Karim Seif El Dahan
- Department of Internal Medicine, UT Southwestern Medical Center, Dallas, TX, USA
| | - Takeshi Yokoo
- Department of Radiology, UT Southwestern Medical Center, Dallas, TX, USA
| | - Darine Daher
- Department of Internal Medicine, UT Southwestern Medical Center, Dallas, TX, USA
| | - Matthew S. Davenport
- Department of Radiology, University of Michigan, Ann Arbor, MI, USA
- Department of Urology, University of Michigan, Ann Arbor, MI, USA
| | - David T. Fetzer
- Department of Radiology, UT Southwestern Medical Center, Dallas, TX, USA
| | | | - Nicole E. Rich
- Department of Internal Medicine, UT Southwestern Medical Center, Dallas, TX, USA
| | - Edward Yang
- Division of Gastroenterology, Kaiser Permanente Medical Group, Riverside, CA, USA
| | - Neehar D. Parikh
- Department of Internal Medicine, University of Michigan, Ann Arbor, MI, USA
| | - Amit G. Singal
- Department of Internal Medicine, UT Southwestern Medical Center, Dallas, TX, USA
| |
Collapse
|
5
|
Nakamura S, Hatakeyama H, Yoshida S, Ungkulpasvich U, Hirotsu T, di Luccio E, Abe M. Detection of Hematological Malignancies Using N-NOSE (Nematode-NOSE). Hematol Oncol 2025; 43:e70062. [PMID: 40156876 PMCID: PMC11954616 DOI: 10.1002/hon.70062] [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: 01/22/2025] [Revised: 03/11/2025] [Accepted: 03/14/2025] [Indexed: 04/01/2025]
Abstract
Hematological malignancies often lack defined risk factors and present with non-specific symptoms, underscoring the urgent need for simple and reliable detection methods. To address this challenge, Hirotsu et al. innovated N-NOSE, a novel, non-invasive cancer screening test that utilizes the chemotaxis response of the nematode Caenorhabditis elegans to detect tumor-related odors in urine. In this clinical study, we assessed the performance of N-NOSE in patients with various hematological malignancies at diagnosis and during treatment. Urine samples were collected from 30 healthy individuals and 89 patients, including those with leukemia (n = 13), malignant lymphoma (n = 53), multiple myeloma (n = 15), primary AL amyloidosis (n = 3), Waldenström's macroglobulinemia (n = 2), myelodysplastic syndrome (n = 2), and blastic plasmacytoid dendritic cell neoplasm (n = 1). Based on the optimal cut-off values in detecting hematological malignancies, N-NOSE demonstrated high positivity rates in treatment-naïve patients: leukemia and multiple myeloma were very high (over 90%), whereas malignant lymphoma was slightly lower than 80%. In the small subset of malignant lymphoma patients who tested N-NOSE-negative, confounding factors included steroid administration and hemodialysis. Importantly, no significant correlation emerged between N-NOSE index values and baseline characteristics or comorbidities other than the presence of cancer. Moreover, in all 32 patients who achieved clinical response following chemotherapy, the N-NOSE index declined, reflecting disease status. These findings highlight N-NOSE's strong potential as a sensitive, non-invasive screening tool for hematological malignancies-particularly multiple myeloma-and support its use in initial detection and monitoring of therapeutic response.
Collapse
Affiliation(s)
- Shingen Nakamura
- Department of Community Medicine and Medical ScienceTokushima University Graduate School of Biomedical SciencesTokushimaJapan
| | | | - Sumiko Yoshida
- Department of Hematology, Endocrinology and MetabolismTokushima University Graduate School of Biomedical SciencesTokushimaJapan
- Department of Clinical ResearchNational Hospital Organization Shikoku Medical Center for Children and AdultsKagawaJapan
| | | | | | | | - Masahiro Abe
- Department of HematologyKawashima HospitalTokushimaJapan
| |
Collapse
|
6
|
Schulze K, Rose TD, Adlung L, Peschka M, Pagani F, Gorgulho J, Fründt TW, Labgaa I, Haber PK, Zimpel C, Castven D, Weinmann A, Garzia-Lezana T, Waldmann M, Renné T, Voß H, Moritz M, Orlikowski D, Schlüter H, Baumbach J, Schwartz M, Lohse AW, Huber S, Sangro B, Macias RI, Izquierdo-Sanchez L, Banales JM, Wege H, Marquardt JU, Villanueva A, Pauling JK, von Felden J. Metabolomic liquid biopsy dynamics predict early-stage HCC and actionable candidates of human hepatocarcinogenesis. JHEP Rep 2025; 7:101340. [PMID: 40290517 PMCID: PMC12023797 DOI: 10.1016/j.jhepr.2025.101340] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/14/2024] [Revised: 01/16/2025] [Accepted: 01/22/2025] [Indexed: 04/30/2025] Open
Abstract
Background & Aims Actionable candidates of hepatocarcinogenesis remain elusive, and tools for early detection are suboptimal. Our aim was to demonstrate that serum metabolome profiles reflect the initiation of hepatocellular carcinoma (HCC) and enable the identification of biomarkers for early HCC detection and actionable candidates for chemoprevention. Methods This global cohort study included 654 patients and 801 biospecimens. Following serum metabolome profiling across the spectrum of hepatocarcinogenesis, we conducted a phase II biomarker case-control study for early HCC detection. Findings were independently validated through in silico analysis, mRNA sequencing, and proteome profiling of primary HCC and non-tumoral tissue, and in vitro experiments. Results Aspartic acid, glutamic acid, taurine, and hypoxanthine were differentially abundant in the serum across chronic liver disease, cirrhosis, initial HCC, and progressed HCC, independent of sex, age, and etiology. In a phase II biomarker case-control study, a blood-based metabolite signature yielded an AUC of 94% to discriminate between patients with early-stage HCC and controls with cirrhosis, including independent validation. Unsupervised biclustering (MoSBi), lipid network analysis (LINEX2), and pathway enrichment analysis confirmed alterations in amino acid-, lipid-, and nucleotide-related pathways. In tumor tissue, these pathways were significantly deregulated regarding gene and protein expression in two independent datasets, including actionable targets RRM2, GMPS, BCAT1, PYCR2, and NEU1. In vitro knockdown confirmed a functional role in proliferation and migration, as exemplified for PYCR2. Conclusions These findings demonstrate that serum metabolome profiling indicates deregulated metabolites and pathways during hepatocarcinogenesis. Our liquid biopsy approach accurately detects early-stage HCC outperforming currently recommended surveillance tools and facilitates identification of actionable candidates for chemoprevention. Impact and implications Deregulated cellular metabolism is a hallmark of cancer. In smaller studies, circulating metabolite profiles have been associated with HCC, although mainly in the context of fatty liver disease. Translation strategies for primary prevention or early detection are lacking. In this global study, we present an unsupervised landscape of the altered serum metabolome profile during hepatocarcinogenesis, independent of age, sex, and etiology. We provide a blood-based metabolite signature that accurately identifies early-stage HCC in a phase II biomarker study including independent validation. Further RRM2, GMPS, BCAT1, PYCR2, and NEU1 are identified in tumor tissue as actionable candidates for prevention. Our data provide the rationale for clinical trials testing liquid biopsy metabolome-based signatures for early HCC detection and the development of chemoprevention strategies.
Collapse
Affiliation(s)
- Kornelius Schulze
- I. Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- ERN-RARE-LIVER, Hamburg, Germany
| | - Tim Daniel Rose
- LipiTUM, Chair of Experimental Bioinformatics, TUM School of Life Sciences, Technical University of Munich, Munich, Germany
- Structural and Computational Biology Unit, European Molecular Biology Laboratory, Heidelberg, Germany
| | - Lorenz Adlung
- I. Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Hamburg Center for Translational Immunology (HCTI), and Center for Biomedical AI (bAIome), Hamburg, Germany
- Mildred Scheel Cancer Career Center HaTriCS4, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Manuela Peschka
- Institute of Clinical Chemistry and Laboratory Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Newborn Screening and Metabolic Laboratory, Department of Pediatrics, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Francesca Pagani
- I. Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Joao Gorgulho
- Department of Oncology, Hematology and Bone Marrow Transplantation with Section of Pneumology, University Medical Centre Hamburg-Eppendorf, Hamburg, Germany
- University Cancer Center Hamburg–Hubertus Wald Tumorzentrum, University Medical Centre Hamburg-Eppendorf, Hamburg, Germany
| | - Thorben W. Fründt
- I. Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- ERN-RARE-LIVER, Hamburg, Germany
| | - Ismail Labgaa
- Division of Liver Diseases, Liver Cancer Program, Tisch Cancer Institute, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Visceral Surgery, Lausanne University Hospital (CHUV), University of Lausanne (UNIL), Lausanne, Switzerland
| | - Philipp K. Haber
- Division of Liver Diseases, Liver Cancer Program, Tisch Cancer Institute, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Carolin Zimpel
- Department of Medicine I, University Medical Center Schleswig-Holstein-Campus Lübeck, Germany
| | - Darko Castven
- Department of Medicine I, University Medical Center Schleswig-Holstein-Campus Lübeck, Germany
| | - Arndt Weinmann
- I. Department of Medicine, University Medical Center Mainz, Germany
| | - Teresa Garzia-Lezana
- Division of Liver Diseases, Liver Cancer Program, Tisch Cancer Institute, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Moritz Waldmann
- Institute of Clinical Chemistry and Laboratory Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Thomas Renné
- Institute of Clinical Chemistry and Laboratory Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Irish Centre for Vascular Biology, School of Pharmacy and Biomolecular Sciences, Royal College of Surgeons in Ireland, Dublin, Ireland
- Center for Thrombosis and Hemostasis (CTH), Johannes Gutenberg University Medical Center, Mainz, Germany
| | - Hannah Voß
- Institute of Clinical Chemistry and Laboratory Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Manuela Moritz
- Institute of Clinical Chemistry and Laboratory Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Dorian Orlikowski
- Institute of Clinical Chemistry and Laboratory Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Hartmut Schlüter
- Institute of Clinical Chemistry and Laboratory Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Jan Baumbach
- Chair of Computational Systems Biology, University of Hamburg, 22607 Hamburg, Germany
| | - Myron Schwartz
- Recanati Miller Transplant Institute, The Icahn School of Medicine at Mount Sinai Hospital, New York, NY, USA
| | - Ansgar W. Lohse
- I. Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- ERN-RARE-LIVER, Hamburg, Germany
| | - Samuel Huber
- I. Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- ERN-RARE-LIVER, Hamburg, Germany
| | - Bruno Sangro
- Liver Unit, Clinica Universidad de Navarra-IDISNA and CIBEREHD, Pamplona, Spain
- Center for the Study of Liver and Gastrointestinal Diseases (CIBEREHD), Carlos III National Institute of Health, Madrid, Spain
| | - Rocio I.R. Macias
- Center for the Study of Liver and Gastrointestinal Diseases (CIBEREHD), Carlos III National Institute of Health, Madrid, Spain
- Experimental Hepatology and Drug Targeting (HEVEPHARM), University of Salamanca, IBSAL, Salamanca, Spain
| | - Laura Izquierdo-Sanchez
- Department of Liver and Gastrointestinal Diseases, Biogipuzkoa Health Research Institute–Donostia University Hospital, University of the Basque Country (UPV/EHU), CIBEREHD, Donostia-San Sebastian, Spain
| | - Jesus M. Banales
- Department of Liver and Gastrointestinal Diseases, Biogipuzkoa Health Research Institute–Donostia University Hospital, University of the Basque Country (UPV/EHU), CIBEREHD, Donostia-San Sebastian, Spain
- IKERBASQUE, Basque Foundation for Science, Bilbao, Spain
- Department of Biochemistry and Genetics, School of Sciences, University of Navarra, Pamplona, Spain
| | - Henning Wege
- I. Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- ERN-RARE-LIVER, Hamburg, Germany
| | - Jens U. Marquardt
- Department of Medicine I, University Medical Center Schleswig-Holstein-Campus Lübeck, Germany
- I. Department of Medicine, University Medical Center Mainz, Germany
| | - Augusto Villanueva
- Division of Liver Diseases, Liver Cancer Program, Tisch Cancer Institute, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Division of Hematology and Medical Oncology, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Josch Konstantin Pauling
- LipiTUM, Chair of Experimental Bioinformatics, TUM School of Life Sciences, Technical University of Munich, Munich, Germany
- Institute for Clinical Chemistry and Laboratory Medicine, University Hospital and Faculty of Medicine Carl Gustav Carus, Dresden University of Technology, Dresden, Germany
| | - Johann von Felden
- I. Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- ERN-RARE-LIVER, Hamburg, Germany
| |
Collapse
|
7
|
Seif El Dahan K, Yokoo T, Mendiratta-Lala M, Fetzer D, Davenport M, Daher D, Rich NE, Yang E, Parikh ND, Singal AG. Exam quality of ultrasound and dynamic contrast-enhanced abbreviated MRI and impact on early-stage HCC detection. Abdom Radiol (NY) 2025; 50:2097-2109. [PMID: 39542949 DOI: 10.1007/s00261-024-04674-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2024] [Revised: 10/31/2024] [Accepted: 11/02/2024] [Indexed: 11/17/2024]
Abstract
PURPOSE MRI is a potential alternative to ultrasound for hepatocellular carcinoma (HCC) detection. We evaluated the impact of ultrasound and dynamic abbreviated MRI (AMRI) exam quality on early-stage HCC detection. METHODS We conducted a multicenter case-control study among patients with cirrhosis (cases with early-stage HCC per Milan Criteria; controls without HCC) who underwent both a liver ultrasound and dynamic contrast-enhanced (DCE) AMRI within 6 months in 2012-2019. Two radiologists performed independent, blinded interpretations of both exams for HCC detection and scored exam quality as no/mild, moderate, or severe limitations. Associations between exam quality, patient characteristics, and HCC detection were assessed by odds ratios (OR). RESULTS Of 216 cases and 432 controls, severe limitations were reported in 7% and 8% of ultrasounds and DCE-AMRIs, respectively. Severe limitations at ultrasound were associated with obesity (OR 2.08, 95%CI [1.32-3.32]) and metabolic dysfunction-associated steatotic liver disease (MASLD) (OR 1.98 [1.12-3.44]) but not for DCE-AMRI. Decompensated cirrhosis (Child-Pugh C) was associated with severe limitations for both ultrasound (OR 2.54 [1.37-4.58]) and DCE-AMRI (OR 3.96 [2.36-6.58]). Compared to exams with no/mild limitations, exams with severe limitations had lower sensitivity for ultrasound (79.6% vs. 21.7%, P < 0.001) and AMRI (86.1% vs. 50.0%, P = 0.001). In patients in whom ultrasound was severely limited, DCE-AMRI had significantly higher odds of early-stage HCC detection than ultrasound (OR 8.23 [1.25-54.02]). CONCLUSIONS HCC detection by DCE-AMRI may be preferred in patients with severe limitations at ultrasound due to obesity and MASLD. Both modalities remain limited for patients with decompensated cirrhosis, for whom alternative strategies may be needed.
Collapse
Affiliation(s)
| | - Takeshi Yokoo
- The University of Texas Southwestern Medical Center, Dallas, USA
| | | | - David Fetzer
- The University of Texas Southwestern Medical Center, Dallas, USA
| | | | - Darine Daher
- The University of Texas Southwestern Medical Center, Dallas, USA
| | - Nicole E Rich
- The University of Texas Southwestern Medical Center, Dallas, USA
| | - Edward Yang
- The University of Texas Southwestern Medical Center, Dallas, USA
| | | | - Amit G Singal
- The University of Texas Southwestern Medical Center, Dallas, USA.
| |
Collapse
|
8
|
He H, Wu Y, Jia Z, Zhang Y, Pan Y, Zhang Y, Su K, Cui Y, Sun Y, Li D, Lv H, Yi J, Wang Y, Kou C, Sun X, Jiang J. A stratified precision screening strategy for enhancing hepatitis B- and C-associated liver cancer detection: a prospective study. Sci Rep 2025; 15:11396. [PMID: 40181083 PMCID: PMC11968812 DOI: 10.1038/s41598-025-95795-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2024] [Accepted: 03/24/2025] [Indexed: 04/05/2025] Open
Abstract
This study explores new screening strategies to enhance liver cancer screening effectiveness. In a prospective study, 2605 participants underwent baseline, 6-months self-reported, and 1-year follow-up screenings using abdominal ultrasonography, AFP, AFP-L3%, and DCP. The results demonstrated the GALADUS protocol exhibited superior performance with higher AUC (0.935 vs. 0.836; DeLong P < 0.001), sensitivity (91.0% vs. 70.8%; P < 0.001), detection (3.1% vs. 2.4%; P < 0.001), and early diagnosis rates (64.2% vs. 58.7%) compared to the AFP/US protocol. Notably, among individuals with an aMAP score ≥ 60, GALADUS had significantly outperformed AFP/US in AUC (0.923 vs. 0.826; DeLong P < 0.001), sensitivity (94.2% vs. 69.6%; P < 0.001), detection (9.7% vs. 7.2%; P < 0.001), and early diagnosis rates (63.1% vs. 54.2%). However, for those with an aMAP score < 60, GALADUS offered no significant advantages. Introducing the "aMAP triage" protocol, combining GALADUS for aMAP ≥ 60 and AFP/US for aMAP < 60, further enhanced AUC to 0.925 (DeLong P < 0.001), improved sensitivity by 19.1% (89.9% vs. 70.8%; P < 0.001), and increased detection (3.1% vs. 2.4%; P < 0.001) and early diagnosis rates (65.0% vs. 58.7%), being cost-effective compared to GALADUS. In conclusion, this study highlights the potential of a stratified precision screening strategy in identifying high-risk individuals, applying tailored early detection protocols to improve liver cancer screening efficacy.
Collapse
Affiliation(s)
- Hua He
- Department of Clinical Epidemiology, the First Hospital of Jilin University, No. 1, Xinmin Street, Changchun, 130021, Jilin Province, China
- Cancer Center, the First Hospital of Jilin University, Changchun, 130021, Jilin Province, China
| | - Yanhua Wu
- Department of Clinical Epidemiology, the First Hospital of Jilin University, No. 1, Xinmin Street, Changchun, 130021, Jilin Province, China
| | - Zhifang Jia
- Department of Clinical Epidemiology, the First Hospital of Jilin University, No. 1, Xinmin Street, Changchun, 130021, Jilin Province, China
| | - Yangyu Zhang
- Department of Clinical Epidemiology, the First Hospital of Jilin University, No. 1, Xinmin Street, Changchun, 130021, Jilin Province, China
- Department of Epidemiology and Biostatistics, School of Public Health, Jilin University, Changchun, 130021, Jilin Province, China
| | - Yuchen Pan
- Department of Clinical Epidemiology, the First Hospital of Jilin University, No. 1, Xinmin Street, Changchun, 130021, Jilin Province, China
- Center of Infectious Diseases and Pathogen Biology, the First Hospital of Jilin University, Changchun, 130021, Jilin Province, China
| | - Yuzheng Zhang
- Department of Clinical Epidemiology, the First Hospital of Jilin University, No. 1, Xinmin Street, Changchun, 130021, Jilin Province, China
| | - Kaisheng Su
- Department of Clinical Epidemiology, the First Hospital of Jilin University, No. 1, Xinmin Street, Changchun, 130021, Jilin Province, China
| | - Yingnan Cui
- Department of Clinical Epidemiology, the First Hospital of Jilin University, No. 1, Xinmin Street, Changchun, 130021, Jilin Province, China
| | - Yuanlin Sun
- Department of Clinical Epidemiology, the First Hospital of Jilin University, No. 1, Xinmin Street, Changchun, 130021, Jilin Province, China
| | - Dongming Li
- Department of Clinical Epidemiology, the First Hospital of Jilin University, No. 1, Xinmin Street, Changchun, 130021, Jilin Province, China
| | - Haiyong Lv
- Department of Clinical Epidemiology, the First Hospital of Jilin University, No. 1, Xinmin Street, Changchun, 130021, Jilin Province, China
| | - Jiaxin Yi
- Department of Clinical Epidemiology, the First Hospital of Jilin University, No. 1, Xinmin Street, Changchun, 130021, Jilin Province, China
| | - Yuehui Wang
- Department of Geriatrics, the First Hospital of Jilin University, Changchun, 130021, Jilin Province, China
| | - Changgui Kou
- Department of Epidemiology and Biostatistics, School of Public Health, Jilin University, Changchun, 130021, Jilin Province, China
| | - Xiaofeng Sun
- Department of Cadre's Wards Ultrasound Diagnostics, Ultrasound Diagnostic Center, the First Hospital of Jilin University, Changchun, 130021, Jilin Province, China
| | - Jing Jiang
- Department of Clinical Epidemiology, the First Hospital of Jilin University, No. 1, Xinmin Street, Changchun, 130021, Jilin Province, China.
- Department of Epidemiology and Biostatistics, School of Public Health, Jilin University, Changchun, 130021, Jilin Province, China.
- Center of Infectious Diseases and Pathogen Biology, the First Hospital of Jilin University, Changchun, 130021, Jilin Province, China.
| |
Collapse
|
9
|
Al-Hasan M, Mehta N, Yang JD, Singal AG. Role of biomarkers in the diagnosis and management of HCC. Liver Transpl 2025; 31:384-394. [PMID: 38738964 DOI: 10.1097/lvt.0000000000000398] [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: 03/01/2024] [Accepted: 05/06/2024] [Indexed: 05/14/2024]
Abstract
For many cancers, biomarkers have served as an important tool across the cancer care continuum from risk stratification and early detection to diagnosis and treatment. Alpha-fetoprotein (AFP) remains one of the few validated biomarkers for patients with HCC. Although AFP has shown potential for each of these steps, its performance, when used alone, has often been suboptimal. There continue to be discordant recommendations about AFP's value when combined with ultrasound for surveillance, as well as its role in diagnostic algorithms. Conversely, high AFP levels are associated with aggressive tumor biology and survival, so it remains a key factor for the selection of candidates for liver transplant. There have been immense efforts to identify and validate additional biomarkers for each of these steps in the HCC care continuum. Indeed, biomarker panels have shown promising data for HCC risk stratification and surveillance among patients with cirrhosis, as well as prognostication and detection of minimal residual disease in patients undergoing HCC treatment. Several large prospective studies are currently ongoing to evaluate the role of these emerging biomarkers in clinical practice.
Collapse
Affiliation(s)
- Mohammed Al-Hasan
- Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Neil Mehta
- Department of Internal Medicine, University of California San Francisco, San Francisco, California, USA
| | - Ju Dong Yang
- Department of Internal Medicine, Cedars Sinai, Los Angeles, California, USA
| | - Amit G Singal
- Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| |
Collapse
|
10
|
Huy DQ, Khai NX, Thinh TH, Linh BT, Minh NN, Thuy VTB. Application of Indirect ELISA and PCR Techniques for Detecting of Hepatocellular Carcinoma using Des-gamma Carboxyprothrombin, Alpha-fetoprotein, and Thioredoxin Biomarkers. Mol Biotechnol 2025:10.1007/s12033-025-01401-z. [PMID: 39998774 DOI: 10.1007/s12033-025-01401-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2023] [Accepted: 02/03/2025] [Indexed: 02/27/2025]
Abstract
Hepatocellular carcinoma (HCC) is one of the five most common cancers and the second leading cause of cancer-related death worldwide. In this study, three monoclonal antibodies were developed for the early detection of HCC. The enzyme-linked immunosorbent assay (ELISA) method is used to detect antigens causing HCC. The final working dilutions of the coated antigen, monoclonal antibody, and enzyme-labeled secondary antibody were determined to be 1:5, 1:100, and 1:15,000, respectively. The optimal dilution of blocking buffer was 1.5% BSA phosphate buffer. The cutoff values were determined to be 0.1989, 0.2539, and 0.3059 for the Des-gamma carboxyprothrombin (DCP), Alpha-fetoprotein (AFP) and Thioredoxin (TXN) antigens, respectively. There is no cross-reaction between antigens and antibodies of different types. The coincidence rates between the indirect ELISA and commercial kits for detecting DCP, AFP, and TXN antigens were 95.24%, 95.24%, and 96.83%, respectively. In addition, a procedure to detect genes encoding TXN, DCP, and AFP via PCR has been developed. The results of the indirect ELISA and PCR methods are similar. In summary, we successfully constructed an indirect ELISA method to detect HCC-causing antigens via three monoclonal antibodies and designed primers to amplify HCC-causing gene fragments, which can be used for diagnosis and screening in clinical medicine.
Collapse
Affiliation(s)
- Duong Quang Huy
- 103 Military Hospital, Vietnam Military Medical University, Hanoi, Vietnam
| | - Nguyen Xuan Khai
- 103 Military Hospital, Vietnam Military Medical University, Hanoi, Vietnam
| | - Tran Hong Thinh
- Institute of Genome Research, Vietnam Academy of Science and Technology, 18 Hoang Quoc Viet, Cau Giay, Hanoi, Vietnam
| | - Bui Thuy Linh
- Institute of Genome Research, Vietnam Academy of Science and Technology, 18 Hoang Quoc Viet, Cau Giay, Hanoi, Vietnam
| | - Nghiem Ngoc Minh
- Institute of Genome Research, Vietnam Academy of Science and Technology, 18 Hoang Quoc Viet, Cau Giay, Hanoi, Vietnam
| | - Vo Thi Bich Thuy
- Institute of Genome Research, Vietnam Academy of Science and Technology, 18 Hoang Quoc Viet, Cau Giay, Hanoi, Vietnam.
| |
Collapse
|
11
|
El-Serag HB, Jin Q, Tayob N, Salem E, Luster M, Alsarraj A, Khaderi S, Singal AG, Marrero JA, Asrani SK, Kanwal F. HES V2.0 outperforms GALAD for detection of HCC: A phase 3 biomarker study in the United States. Hepatology 2025; 81:465-475. [PMID: 38899967 PMCID: PMC11655698 DOI: 10.1097/hep.0000000000000953] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/19/2024] [Accepted: 05/20/2024] [Indexed: 06/21/2024]
Abstract
BACKGROUND AND AIMS The original hepatocellular carcinoma early detection screening (HES) score, which combines alpha-fetoprotein (AFP) with age, alanine aminotransferase, and platelets, has better performance than AFP alone for early HCC detection. We have developed HES V2.0 by adding AFP-L3 and des-gamma-carboxy prothrombin to the score and compared its performance to GALAD and ASAP scores among patients with cirrhosis. APPROACH AND RESULTS We conducted a prospective-specimen collection, retrospective-blinded-evaluation phase 3 biomarker cohort study in patients with cirrhosis enrolled in imaging and AFP surveillance. True-positive rate (TPR)/sensitivity and false-positive rate for any or early HCC were calculated for GALAD, ASAP, and HES V2.0 scores within 6, 12, and 24 months of HCC diagnosis. We calculated the AUROC curve and estimated TPR based on an optimal threshold at a fixed false-positive rate of 10%. We analyzed 2331 patients, of whom 125 developed HCC (71% in the early stages). For any HCC, HES V2.0 had higher TPR than GALAD overall (+7.2%), at 6 months (+3.6%), at 12 months (+7.2%), and 24 months (+13.0%) before HCC diagnosis. HES V2.0 had higher TPR than ASAP for all time points (+5.9% to +12.0%). For early HCC, HES V2.0 had higher sensitivity/TPR than GALAD overall (+6.7%), at 12 months (+6.3%), and 24 months (+14.6%) but not at 6 months (+0.0%) and higher than ASAP for all time points (+13.4% to +18.0%). CONCLUSIONS In a prospective cohort study, HES V2.0 had a significantly higher performance for identifying new HCC, including early stage, than GALAD or ASAP.
Collapse
Affiliation(s)
- Hashem B. El-Serag
- Section of Gastroenterology and Hepatology, Department of Medicine, Michael E. DeBakey VA Medical Center and Baylor College of Medicine, Houston, Texas, USA
- Section of Health Services Research, Michael E. DeBakey VA Medical Center and Baylor College of Medicine, Houston, Texas, USA
| | - Qingchun Jin
- Department of Data Science, Dana-Farber Cancer Institute, Boston, Massachusetts, USA
| | - Nabihah Tayob
- Department of Data Science, Dana-Farber Cancer Institute, Boston, Massachusetts, USA
| | - Emad Salem
- Section of Gastroenterology and Hepatology, Department of Medicine, Michael E. DeBakey VA Medical Center and Baylor College of Medicine, Houston, Texas, USA
| | - Michelle Luster
- Section of Gastroenterology and Hepatology, Department of Medicine, Michael E. DeBakey VA Medical Center and Baylor College of Medicine, Houston, Texas, USA
| | - Abeer Alsarraj
- Section of Health Services Research, Michael E. DeBakey VA Medical Center and Baylor College of Medicine, Houston, Texas, USA
| | - Saira Khaderi
- Section of Gastroenterology and Hepatology, Department of Medicine, Michael E. DeBakey VA Medical Center and Baylor College of Medicine, Houston, Texas, USA
| | - Amit G. Singal
- Department of Medicine, UT Southwestern Medical Center, Dallas, Texas, USA
| | - Jorge A. Marrero
- Department of Medicine, Division of Gastroenterology and Hepatology, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Sumeet K. Asrani
- Department of Medicine, Baylor University Medical Center, Dallas, Texas, USA
| | - Fasiha Kanwal
- Section of Gastroenterology and Hepatology, Department of Medicine, Michael E. DeBakey VA Medical Center and Baylor College of Medicine, Houston, Texas, USA
- Section of Health Services Research, Michael E. DeBakey VA Medical Center and Baylor College of Medicine, Houston, Texas, USA
| |
Collapse
|
12
|
Cooley MA, Schneider AR, Fritcher EGB, Milosevic D, Levy MJ, Bridgeman AR, Martin JA, Petersen BT, Dayyeh BKA, Storm AC, Law RJ, Vargas EJ, Garimella V, Zemla T, Jenkins SM, Yin J, Gores GJ, Roberts LR, Kipp BR, Chandrasekhara V. Utility of methylated DNA markers for the diagnosis of malignant biliary strictures. Hepatology 2025; 81:453-464. [PMID: 38905442 PMCID: PMC11827039 DOI: 10.1097/hep.0000000000000970] [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: 10/31/2023] [Accepted: 05/24/2024] [Indexed: 06/23/2024]
Abstract
BACKGROUND AND AIMS Early identification of malignant biliary strictures (MBSs) is challenging, with up to 20% classified as indeterminants after preliminary testing and tissue sampling with endoscopic retrograde cholangiopancreatography. We aimed to evaluate the use of methylated DNA markers (MDMs) from biliary brushings to enhance MBS detection in a prospective cohort. APPROACH Candidate MDMs were evaluated for their utility in MBS diagnosis through a series of discovery and validation phases. DNA was extracted from biliary brushing samples, quantified, bisulfite-converted, and then subjected to methylation-specific droplet digital polymerase chain reaction. Patients were considered to have no malignancy if the sampling was negative and there was no evidence of malignancy after 1 year or definitive negative surgical histopathology. RESULTS Fourteen candidate MDMs were evaluated in the discovery phase, with top-performing and new markers evaluated in the technical validation phase. The top 4 MDMs were TWIST1, HOXA1, VSTM2B, and CLEC11A, which individually achieved AUC values of 0.82, 0.81, 0.83, and 0.78, respectively, with sensitivities of 59.4%, 53.1%, 62.5%, and 50.0%, respectively, at high specificities for malignancy of 95.2%-95.3% for the final biologic validation phase. When combined as a panel, the AUC was 0.86, achieving 73.4% sensitivity and 92.9% specificity, which outperformed cytology and fluorescence in situ hybridization (FISH). CONCLUSIONS The selected MDMs demonstrated improved performance characteristics for the detection of MBS compared to cytology and FISH. Therefore, MDMs should be considered viable candidates for inclusion in diagnostic testing algorithms.
Collapse
Affiliation(s)
- Matthew A. Cooley
- Mayo Clinic Graduate School of Biomedical Sciences, Rochester, Minnesota
| | - Amber R. Schneider
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, Minnesota
| | | | - Dragana Milosevic
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, Minnesota
| | - Michael J. Levy
- Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, Minnesota
| | - Amber R. Bridgeman
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, Minnesota
| | - John A. Martin
- Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, Minnesota
| | - Bret T. Petersen
- Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, Minnesota
| | | | - Andrew C. Storm
- Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, Minnesota
| | - Ryan J. Law
- Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, Minnesota
| | - Eric J. Vargas
- Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, Minnesota
| | - Vishal Garimella
- Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, Minnesota
| | - Tyler Zemla
- Health Science Research Biomedical Statistics and Informatics, Mayo Clinic, Rochester, Minnesota
| | - Sarah M. Jenkins
- Health Science Research Biomedical Statistics and Informatics, Mayo Clinic, Rochester, Minnesota
| | - Jun Yin
- Health Science Research Biomedical Statistics and Informatics, Mayo Clinic, Rochester, Minnesota
| | - Gregory J. Gores
- Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, Minnesota
| | - Lewis R. Roberts
- Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, Minnesota
| | - Benjamin R. Kipp
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, Minnesota
| | | |
Collapse
|
13
|
Goodman MT, Lombardi C, Torrens A, Bresee C, Saloman JL, Li L, Yang Y, Fisher WE, Fogel EL, Forsmark CE, Conwell DL, Hart PA, Park WG, Topazian M, Vege SS, Van Den Eeden SK, Bellin MD, Andersen DK, Serrano J, Yadav D, Pandol SJ, Piomelli D. Association of Serum Endocannabinoid Levels with Pancreatitis and Pancreatitis-Related Pain. Cannabis Cannabinoid Res 2025; 10:60-70. [PMID: 39291350 PMCID: PMC11947650 DOI: 10.1089/can.2024.0079] [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] [Indexed: 09/19/2024] Open
Abstract
Background and Aims: This investigation examined the association of pancreatitis and pancreatitis-related pain with serum levels of two endocannabinoid molecules such as anandamide (AEA) and 2-arachidonoylglycerol (2-AG) and two paracannabinoid molecules such as oleoylethanolamide (OEA) and palmitoylethanolamide (PEA). Methods: A case-control study was conducted within the Prospective Evaluation of Chronic Pancreatitis for Epidemiological and Translational Studies, including participants with no pancreas disease (N = 56), chronic abdominal pain of suspected pancreatic origin or indeterminate chronic pancreatitis (CP) (N = 22), acute pancreatitis (N = 33), recurrent acute pancreatitis (N = 57), and definite CP (N = 63). Results: Circulating AEA concentrations were higher in women than in men (p = 0.0499), and PEA concentrations were higher in obese participants than those who were underweight/normal or overweight (p = 0.003). Asymptomatic controls with no pancreatic disease had significantly (p = 0.03) lower concentrations of AEA compared with all disease groups combined. The highest concentrations of AEA were observed in participants with acute pancreatitis, followed by those with recurrent acute pancreatitis, chronic abdominal pain/indeterminant CP, and definite CP. Participants with pancreatitis reporting abdominal pain in the past year had significantly (p = 0.04) higher concentrations of AEA compared with asymptomatic controls. Levels of 2-AG were significantly lower (p = 0.02) among participants reporting abdominal pain in the past week, and pain intensity was inversely associated with concentrations of 2-AG and OEA. Conclusions: Endocannabinoid levels may be associated with stage of pancreatitis, perhaps through activation of the CB1 receptor. Validation of our findings would support the investigation of novel therapeutics, including cannabinoid receptor-1 antagonists, in this patient population.
Collapse
Affiliation(s)
- Marc T. Goodman
- Prevention and Control Program, Cancer Center, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - Christina Lombardi
- Prevention and Control Program, Cancer Center, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - Alexa Torrens
- Department and Anatomy and Neurobiology, University of California, Irvine, California, USA
| | - Catherine Bresee
- Department of Biostatistics and Bioinformatics Research Center, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - Jami L. Saloman
- Center for Pain Research, Division of Gastroenterology, Hepatology and Nutrition, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Liang Li
- Department of Biostatistics, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Yunlong Yang
- Department of Biostatistics, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - William E. Fisher
- Michael E. DeBakey Department of Surgery, Baylor College of Medicine, Houston, Texas, USA
| | - Evan L. Fogel
- Division of Gastroenterology and Hepatology, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Christopher E. Forsmark
- Division of Gastroenterology, Hepatology, and Nutrition, University of Florida, Gainesville, Florida, USA
| | - Darwin L. Conwell
- Department of Internal Medicine, University of Kentucky College of Medicine, Lexington, Kentucky, USA
| | - Phil A. Hart
- Division of Gastroenterology, Hepatology, & Nutrition, The Ohio State University Wexner Medical Center, Columbus, Ohio, USA
| | - Walter G. Park
- Department of Medicine, Division of Gastroenterology and Hepatology, Stanford University Medical Center, Stanford, California, USA
| | | | - Santhi S. Vege
- Department of Internal Medicine, Mayo Clinic, Rochester, Minnesota, USA
| | | | - Melena D. Bellin
- Division of Pediatric Endocrinology, Department of Pediatrics, University of Minnesota Medical School, Minneapolis, Minnesota, USA
| | - Dana K. Andersen
- Division of Digestive Diseases and Nutrition, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, Maryland, USA
| | - Jose Serrano
- Division of Digestive Diseases and Nutrition, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, Maryland, USA
| | - Dhiraj Yadav
- Department of Medicine Division of Gastroenterology, Hepatology & Nutrition University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Stephen J. Pandol
- Division of Gastroenterology and Hepatology, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - Daniele Piomelli
- Department and Anatomy and Neurobiology, University of California, Irvine, California, USA
- Department of Biological Chemistry, University of California, Irvine, California, USA
- Department of Pharmaceutical Sciences, University of California, Irvine, California, USA
| | - on behalf of the Consortium for the Study of Chronic Pancreatitis, Diabetes, and Pancreatic Cancer (CPDPC)
- Prevention and Control Program, Cancer Center, Cedars-Sinai Medical Center, Los Angeles, California, USA
- Department and Anatomy and Neurobiology, University of California, Irvine, California, USA
- Department of Biostatistics and Bioinformatics Research Center, Cedars-Sinai Medical Center, Los Angeles, California, USA
- Center for Pain Research, Division of Gastroenterology, Hepatology and Nutrition, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
- Department of Biostatistics, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
- Michael E. DeBakey Department of Surgery, Baylor College of Medicine, Houston, Texas, USA
- Division of Gastroenterology and Hepatology, Indiana University School of Medicine, Indianapolis, Indiana, USA
- Division of Gastroenterology, Hepatology, and Nutrition, University of Florida, Gainesville, Florida, USA
- Department of Internal Medicine, University of Kentucky College of Medicine, Lexington, Kentucky, USA
- Division of Gastroenterology, Hepatology, & Nutrition, The Ohio State University Wexner Medical Center, Columbus, Ohio, USA
- Department of Medicine, Division of Gastroenterology and Hepatology, Stanford University Medical Center, Stanford, California, USA
- Mayo Clinic, Rochester, Minnesota, USA
- Department of Internal Medicine, Mayo Clinic, Rochester, Minnesota, USA
- Division of Research, Kaiser Permanente Northern California, Oakland, California, USA
- Division of Pediatric Endocrinology, Department of Pediatrics, University of Minnesota Medical School, Minneapolis, Minnesota, USA
- Division of Digestive Diseases and Nutrition, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, Maryland, USA
- Department of Medicine Division of Gastroenterology, Hepatology & Nutrition University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
- Division of Gastroenterology and Hepatology, Cedars-Sinai Medical Center, Los Angeles, California, USA
- Department of Biological Chemistry, University of California, Irvine, California, USA
- Department of Pharmaceutical Sciences, University of California, Irvine, California, USA
| |
Collapse
|
14
|
Witzig TE, Taylor WR, Mahoney DW, Bamlet WR, Foote PH, Burger KN, Doering KA, Devens ME, Arndt JR, O'Connell MC, Berger CK, Novak AJ, Cerhan JR, Hennek J, Katerov S, Allawi HT, Jevremovic D, Dao LN, Graham RP, Kisiel JB. Blood Plasma Methylated DNA Markers in the Detection of Lymphoma: Discovery, Validation, and Clinical Pilot. Am J Hematol 2025; 100:218-228. [PMID: 39564730 PMCID: PMC11705204 DOI: 10.1002/ajh.27533] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2024] [Revised: 10/30/2024] [Accepted: 11/04/2024] [Indexed: 11/21/2024]
Abstract
Lymphoma is one of the leading causes of cancer and cancer deaths and yet has not been amenable to population screening. The role of methylated DNA markers (MDMs) in the detection of lymphoma has not been extensively studied. We aimed to discover, validate, and test tissue-derived MDMs of lymphoma in archival plasma specimens. Reduced representation bisulfite sequencing (RRBS) was performed on a discovery set of frozen tissues. MDMs identified were converted to methylation-specific PCR assays and validated on independent formalin-fixed, paraffin-embedded (FFPE) tissues. Target enrichment long-probe quantitative-amplified signal (TELQAS) assays were developed and assayed in plasma-extracted, bisulfite-converted DNA from independent treatment-naïve lymphoma patients and healthy controls. Prediction of cancer status was modeled using random forest model with in silico cross-validation. After discovery and validation in tissue, 16 TELQAS assays (ZNF503, VWA5B1, HOXA9, GABRG3, ITGA5, MAX.chr17.7190, BNC1, CDK20, MAX.chr4.4069, TPBG, DNAH14, SYT6, CACNG8, FAM110B, ADRA1D, and NRN1) were selected for testing in plasma. These detected 78% (95% CI, 74%-82%) of lymphoma cases at 90% specificity. Excluding marginal zone and T-cell lymphomas, sensitivity increased to 84% (80%-88%). MDMs in plasma show promise to detect lymphoma and are candidates for inclusion in multi-cancer detection studies.
Collapse
Affiliation(s)
| | - William R. Taylor
- Division of Gastroenterology and HepatologyMayo ClinicRochesterMinnesotaUSA
| | - Douglas W. Mahoney
- Department of Qualitative Health SciencesMayo ClinicRochesterMinnesotaUSA
| | - William R. Bamlet
- Department of Qualitative Health SciencesMayo ClinicRochesterMinnesotaUSA
| | - Patrick H. Foote
- Division of Gastroenterology and HepatologyMayo ClinicRochesterMinnesotaUSA
| | - Kelli N. Burger
- Department of Qualitative Health SciencesMayo ClinicRochesterMinnesotaUSA
| | - Karen A. Doering
- Division of Gastroenterology and HepatologyMayo ClinicRochesterMinnesotaUSA
| | - Mary E. Devens
- Division of Gastroenterology and HepatologyMayo ClinicRochesterMinnesotaUSA
| | - Jacquelyn R. Arndt
- Division of Gastroenterology and HepatologyMayo ClinicRochesterMinnesotaUSA
| | - Maria C. O'Connell
- Division of Gastroenterology and HepatologyMayo ClinicRochesterMinnesotaUSA
| | - Calise K. Berger
- Division of Gastroenterology and HepatologyMayo ClinicRochesterMinnesotaUSA
| | - Anne J. Novak
- Division of HematologyMayo ClinicRochesterMinnesotaUSA
| | - James R. Cerhan
- Department of Qualitative Health SciencesMayo ClinicRochesterMinnesotaUSA
| | | | - Slava Katerov
- Exact Sciences Development CompanyMadisonWisconsinUSA
| | | | | | - Linda N. Dao
- Division of Anatomic PathologyMayo ClinicRochesterMinnesotaUSA
| | | | - John B. Kisiel
- Division of Gastroenterology and HepatologyMayo ClinicRochesterMinnesotaUSA
| |
Collapse
|
15
|
Marsh TL, Parikh ND, Roberts LR, Schwartz ME, Nguyen MH, Befeler A, Page-Lester S, Tayob N, Srivastava S, Rinaudo JA, Singal AG, Reddy KR, Marrero JA. A Phase 3 Biomarker Validation of GALAD for the Detection of Hepatocellular Carcinoma in Cirrhosis. Gastroenterology 2025; 168:316-326.e6. [PMID: 39293548 DOI: 10.1053/j.gastro.2024.09.008] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/12/2024] [Revised: 08/12/2024] [Accepted: 09/02/2024] [Indexed: 09/20/2024]
Abstract
BACKGROUND & AIMS Better surveillance tests for hepatocellular carcinoma (HCC) are needed. The GALAD score (gender, age, α-fetoprotein [AFP] L3, AFP, and des-γ carboxyprothrombin) has been shown to have excellent sensitivity and specificity for HCC in phase 2 studies. We performed a phase 3 biomarker validation study to compare GALAD with AFP in detecting HCC. METHODS This is a prospective study of patients with cirrhosis enrolled at 7 centers. Surveillance for HCC was performed every 6 months at each site, and HCC diagnosis was confirmed per American Association for the Study of Liver Diseases guidelines. Blood for biomarker research was obtained at each follow-up visit and stored in a biorepository. Measurements of AFP, AFP-L3, and des-γ carboxyprothrombin) were performed in a FujiFilm laboratory by staff blinded to clinical data. The performance of GALAD in detecting HCC was retrospectively evaluated within 12 months before the clinical diagnosis. All analyses were conducted by an unblinded statistician in the Early Detection Research Network data management and coordinating center. RESULTS A total of 1,558 patients with cirrhosis were enrolled and followed for a median of 2.2 years. A total of 109 patients developed HCC (76 very early or early stage), with an annual incident rate of 2.4%. The areas under the curve for AFP and GALAD within 12 months before HCC were 0.66 and 0.78 (P < .001), respectively. Using a cutoff for GALAD of -1.36, the specificity was 82%, and the sensitivity at 12 months before HCC diagnosis was 62%. For comparison, performance of AFP at 82% specificity showed 41% sensitivity at 12 months before HCC diagnosis (P = .001). CONCLUSIONS GALAD score, compared to AFP, improves the detection of HCC within 12 months before the actual diagnosis.
Collapse
Affiliation(s)
- Tracey L Marsh
- Biostatistics Program, Public Health Sciences Division, Fred Hutchinson Cancer Research Center, Seattle, Washington
| | - Neehar D Parikh
- Division of Gastroenterology and Hepatology, University of Michigan, Ann Arbor, Michigan
| | - Lewis R Roberts
- Division of Gastroenterology and Hepatology, Mayo Clinic College of Medicine and Science, Rochester, Minnesota
| | - Myron E Schwartz
- Recanati/Miller Transplant Institute, Mount Sinai Medical Center, New York, New York
| | - Mindie H Nguyen
- Division of Gastroenterology and Hepatology and Department of Epidemiology and Population Health, Stanford University, Palo Alto, California
| | - Alex Befeler
- Division of Gastroenterology, Saint Louis University, St. Louis, Missouri
| | - Stephanie Page-Lester
- Biostatistics Program, Public Health Sciences Division, Fred Hutchinson Cancer Research Center, Seattle, Washington
| | - Nabihah Tayob
- Department of Data Science, Dana Farber Cancer Institute, Boston, Massachusetts
| | - Sudhir Srivastava
- Cancer Biomarkers Research Group, Division of Cancer Prevention, National Cancer Institute, Bethesda, Maryland
| | - Jo Ann Rinaudo
- Cancer Biomarkers Research Group, Division of Cancer Prevention, National Cancer Institute, Bethesda, Maryland
| | - Amit G Singal
- Division of Digestive and Liver Disease, University of Texas Southwestern, Dallas, Texas
| | - K Rajender Reddy
- Division of Gastroenterology and Hepatology, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Jorge A Marrero
- Division of Gastroenterology and Hepatology, University of Pennsylvania, Philadelphia, Pennsylvania.
| |
Collapse
|
16
|
Olislagers M, de Jong FC, Rutten VC, Boormans JL, Mahmoudi T, Zuiverloon TCM. Molecular biomarkers of progression in non-muscle-invasive bladder cancer - beyond conventional risk stratification. Nat Rev Urol 2025; 22:75-91. [PMID: 39095581 DOI: 10.1038/s41585-024-00914-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/21/2024] [Indexed: 08/04/2024]
Abstract
The global incidence of bladder cancer is more than half a million diagnoses each year. Bladder cancer can be categorized into non-muscle-invasive bladder cancer (NMIBC), which accounts for ~75% of diagnoses, and muscle-invasive bladder cancer (MIBC). Up to 45% of patients with NMIBC develop disease progression to MIBC, which is associated with a poor outcome, highlighting a clinical need to identify these patients. Current risk stratification has a prognostic value, but relies solely on clinicopathological parameters that might not fully capture the complexity of disease progression. Molecular research has led to identification of multiple crucial players involved in NMIBC progression. Identified biomarkers of progression are related to cell cycle, MAPK pathways, apoptosis, tumour microenvironment, chromatin stability and DNA-damage response. However, none of these biomarkers has been prospectively validated. Reported gene signatures of progression do not improve NMIBC risk stratification. Molecular subtypes of NMIBC have improved our understanding of NMIBC progression, but these subtypes are currently unsuitable for clinical implementation owing to a lack of prospective validation, limited predictive value as a result of intratumour subtype heterogeneity, technical challenges, costs and turnaround time. Future steps include the development of consensus molecular NMIBC subtypes that might improve conventional clinicopathological risk stratification. Prospective implementation studies of biomarkers and the design of biomarker-guided clinical trials are required for the integration of molecular biomarkers into clinical practice.
Collapse
Affiliation(s)
- Mitchell Olislagers
- Department of Urology, Erasmus University Medical Center, Erasmus MC Cancer Institute, Rotterdam, the Netherlands
| | - Florus C de Jong
- Department of Urology, Erasmus University Medical Center, Erasmus MC Cancer Institute, Rotterdam, the Netherlands
| | - Vera C Rutten
- Department of Urology, Erasmus University Medical Center, Erasmus MC Cancer Institute, Rotterdam, the Netherlands
| | - Joost L Boormans
- Department of Urology, Erasmus University Medical Center, Erasmus MC Cancer Institute, Rotterdam, the Netherlands
| | - Tokameh Mahmoudi
- Department of Urology, Erasmus University Medical Center, Erasmus MC Cancer Institute, Rotterdam, the Netherlands
- Department of Pathology, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - Tahlita C M Zuiverloon
- Department of Urology, Erasmus University Medical Center, Erasmus MC Cancer Institute, Rotterdam, the Netherlands.
| |
Collapse
|
17
|
Iyer PG. EMERALD in the making? A promising blood-based microRNA panel to detect oesophageal adenocarcinoma and Barrett's oesophagus. Gut 2025:gutjnl-2025-334795. [PMID: 39875185 DOI: 10.1136/gutjnl-2025-334795] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/13/2025] [Accepted: 01/16/2025] [Indexed: 01/30/2025]
Affiliation(s)
- Prasad G Iyer
- Gastroenterology and Hepatology, Mayo Clinic, Scottsdale, Arizona, USA
| |
Collapse
|
18
|
Mishra UK, Srivastava S, Singh KR, Kumar A, Singh V, Mishra DP, Chandel VS, Singh J, Pandey SS, Srivastava S. A bio-nano-engineered platform fabricated from cerium oxide-carbon nanoparticles stabilized with chitosan for label-free sensing of a lung cancer biomarker. ANALYTICAL METHODS : ADVANCING METHODS AND APPLICATIONS 2025; 17:349-359. [PMID: 39633582 DOI: 10.1039/d4ay01535e] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/07/2024]
Abstract
Herein, we report a label-free cancer biosensor designed for carcinoembryonic antigen (CEA) detection using a nanohybrid comprising CeO2 nanoparticles, carbon nanoparticles (CNPs), and chitosan (Ch). CeO2 nanoparticles were prepared using a simple green synthesis process. A thin film of the CeO2-CNPs-Ch nanohybrid was formed on indium tin oxide (ITO)-coated glass plates that endowed a high surface area, excellent stability, and good adsorption for the efficient loading of CEA antibodies. Quantitative and selective determination of CEA antigen was achieved by immobilizing monoclonal CEA antibodies (anti-CEA) on the CeO2-CNPs-Ch/ITO platform. The electrochemical response of the anti-CEA/CeO2-CNPs-Ch/ITO immunoelectrode was evaluated in a label-free immunoassay format using differential pulse voltammetry (DPV). The response studies of immunoelectrodes indicated wider linearity with respect to the CEA concentration in the range of 0.05-100 ng mL-1. The electrochemical cancer biosensor exhibited a higher sensitivity of 22.40 μA cm-2 per decade change in concentration along with storage stability for up to 35 days. The limit of detection (LOD) was 0.037 ng mL-1. Furthermore, this cancer biosensor exhibited good specificity and reproducibility. Thus, the proposed CeO2-CNPs-Ch nanocomposite-based platform provides an efficient method for the analysis of other antigen-antibody interactions and biomolecule detection. The efficacy of the anti-CEA/CeO2-CNPs-Ch/ITO immunoelectrode was further examined by measuring CEA levels in human serum.
Collapse
Affiliation(s)
- Upendra Kumar Mishra
- Department of Applied Science and Humanities, Rajkiya Engineering College Ambedkar Nagar (Dr A.P.J. Abdul Kalam Technical University, Lucknow), Uttar Pradesh 224122, India.
| | - Saurabh Srivastava
- Department of Applied Science and Humanities, Rajkiya Engineering College Ambedkar Nagar (Dr A.P.J. Abdul Kalam Technical University, Lucknow), Uttar Pradesh 224122, India.
| | - Kshitij Rb Singh
- Graduate School of Life Science and Systems Engineering, Kyushu Institute of Technology, 2-4 Hibikino, Wakamatsu, Kitakyushu 808-0196, Japan
| | - Atul Kumar
- Department of Applied Science and Humanities, Rajkiya Engineering College Ambedkar Nagar (Dr A.P.J. Abdul Kalam Technical University, Lucknow), Uttar Pradesh 224122, India.
| | - Vivekanand Singh
- Department of Applied Science and Humanities, Rajkiya Engineering College Ambedkar Nagar (Dr A.P.J. Abdul Kalam Technical University, Lucknow), Uttar Pradesh 224122, India.
| | - Devendra P Mishra
- Department of Applied Science and Humanities, Rajkiya Engineering College Ambedkar Nagar (Dr A.P.J. Abdul Kalam Technical University, Lucknow), Uttar Pradesh 224122, India.
| | - Vishal Singh Chandel
- Department of Applied Science and Humanities, Rajkiya Engineering College Ambedkar Nagar (Dr A.P.J. Abdul Kalam Technical University, Lucknow), Uttar Pradesh 224122, India.
| | - Jay Singh
- Department of Chemistry, Institute of Science, Banaras Hindu University, Varanasi, Uttar Pradesh 221005, India
| | - Shyam S Pandey
- Graduate School of Life Science and Systems Engineering, Kyushu Institute of Technology, 2-4 Hibikino, Wakamatsu, Kitakyushu 808-0196, Japan
| | - Saurabh Srivastava
- Department of Applied Science and Humanities, Rajkiya Engineering College Ambedkar Nagar (Dr A.P.J. Abdul Kalam Technical University, Lucknow), Uttar Pradesh 224122, India.
| |
Collapse
|
19
|
Kang SK, Gulati R, Moise N, Hur C, Elkin EB. Multi-Cancer Early Detection Tests: State of the Art and Implications for Radiologists. Radiology 2025; 314:e233448. [PMID: 39807974 PMCID: PMC11783158 DOI: 10.1148/radiol.233448] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2023] [Revised: 03/21/2024] [Accepted: 04/25/2024] [Indexed: 01/16/2025]
Abstract
Multi-cancer early detection (MCED) tests are already being marketed as noninvasive, convenient opportunities to test for multiple cancer types with a single blood sample. The technology varies-involving detection of circulating tumor DNA, fragments of DNA, RNA, or proteins unique to each targeted cancer. The priorities and tradeoffs of reaching diagnostic resolution in the setting of possible false positives and negatives remain under active study. Given the well-established role of imaging in lesion detection and characterization for most cancers, radiologists have an essential role to play in selecting diagnostic pathways, determining the validity of test results, resolving false-positive MCED test results, and evaluating tradeoffs for clinical policy. Appropriate access to and use of imaging tests will also factor into clinical guidelines. Thus, all clinicians potentially involved with MCED tests for cancer screening will need to weigh the benefits and harms of MCED testing, including consideration of how the tests will be used alongside or in place of other screening options, how diagnostic confirmation tests should be selected, and what the implications are for policy and reimbursement decisions. Further, patients will need regular support to make informed decisions about screening using MCED tests in the context of their personal cancer risks, health-related values, and access to care.
Collapse
Affiliation(s)
| | - Roman Gulati
- From the Departments of Radiology and Population Health, New York
University Langone Medical Center, New York, NY (S.K.K.); Division of Public
Health Sciences, Fred Hutchinson Cancer Center, Seattle, Wash (R.G.); Department
of Medicine, Vagelos College of Physicians and Surgeons, Columbia University,
New York, NY (N.M., C.H.); Herbert Irving Comprehensive Cancer Center, New York,
NY (C.H., E.B.E.); and Department of Health Policy and Management, Mailman
School of Public Health, Columbia University, New York, NY (E.B.E.)
| | - Nathalie Moise
- From the Departments of Radiology and Population Health, New York
University Langone Medical Center, New York, NY (S.K.K.); Division of Public
Health Sciences, Fred Hutchinson Cancer Center, Seattle, Wash (R.G.); Department
of Medicine, Vagelos College of Physicians and Surgeons, Columbia University,
New York, NY (N.M., C.H.); Herbert Irving Comprehensive Cancer Center, New York,
NY (C.H., E.B.E.); and Department of Health Policy and Management, Mailman
School of Public Health, Columbia University, New York, NY (E.B.E.)
| | - Chin Hur
- From the Departments of Radiology and Population Health, New York
University Langone Medical Center, New York, NY (S.K.K.); Division of Public
Health Sciences, Fred Hutchinson Cancer Center, Seattle, Wash (R.G.); Department
of Medicine, Vagelos College of Physicians and Surgeons, Columbia University,
New York, NY (N.M., C.H.); Herbert Irving Comprehensive Cancer Center, New York,
NY (C.H., E.B.E.); and Department of Health Policy and Management, Mailman
School of Public Health, Columbia University, New York, NY (E.B.E.)
| | - Elena B. Elkin
- From the Departments of Radiology and Population Health, New York
University Langone Medical Center, New York, NY (S.K.K.); Division of Public
Health Sciences, Fred Hutchinson Cancer Center, Seattle, Wash (R.G.); Department
of Medicine, Vagelos College of Physicians and Surgeons, Columbia University,
New York, NY (N.M., C.H.); Herbert Irving Comprehensive Cancer Center, New York,
NY (C.H., E.B.E.); and Department of Health Policy and Management, Mailman
School of Public Health, Columbia University, New York, NY (E.B.E.)
| |
Collapse
|
20
|
Wisman GBA, Wojdacz TK, Altucci L, Rots MG, DeMeo DL, Snieder H. Clinical promise and applications of epigenetic biomarkers. Clin Epigenetics 2024; 16:192. [PMID: 39732727 DOI: 10.1186/s13148-024-01806-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2024] [Accepted: 12/19/2024] [Indexed: 12/30/2024] Open
Affiliation(s)
- G Bea A Wisman
- Department of Gynecologic Oncology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands.
| | - Tomasz K Wojdacz
- Independent Clinical Epigenetics Laboratory, Pomeranian Medical University, Szczecin, Poland
| | - Lucia Altucci
- Dipartimento di Medicina di Precisione, Università degli Studi della Campania "Luigi Vanvitelli" Napoli, Napoli, Italy
- Biogem, Molecular Biology and Genetics Research Institute, Ariano Irpino, Italy
- IEOS CNR, Naples, Italy
- Medical Epigenetics Program, Azienda Ospedaliera Universitaria "Luigi Vanvitelli", Naples, Italy
| | - Marianne G Rots
- Department of Pathology and Medical Biology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Dawn L DeMeo
- Channing Division of Network Medicine and Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, USA
| | - Harold Snieder
- Department of Epidemiology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands.
| |
Collapse
|
21
|
Smith LM, Mahoney DW, Bamlet WR, Yu F, Liu S, Goggins MG, Darabi S, Majumder S, Wang QL, Coté GA, Demeure MJ, Zhang Z, Srivastava S, Chawla A, Izmirlian G, Olson JE, Wolpin BM, Genkinger JM, Zaret KS, Brand R, Koay EJ, Oberg AL. Early detection of pancreatic cancer: Study design and analytical considerations in biomarker discovery and early phase validation studies. Pancreatology 2024; 24:1265-1279. [PMID: 39516175 PMCID: PMC11780679 DOI: 10.1016/j.pan.2024.10.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/15/2024] [Revised: 10/05/2024] [Accepted: 10/27/2024] [Indexed: 11/16/2024]
Abstract
OBJECTIVES Pancreatic ductal adenocarcinoma (PDAC) is a highly lethal disease that is challenging to detect at an early stage. Biomarkers are needed that can detect PDAC early in the course of disease when interventions lead to the best outcomes. We highlight study design and statistical considerations that inform pancreatic cancer early detection biomarker evaluation. METHODS We describe experimental design strategies in this setting useful for streamlining biomarker evaluation at each Early Detection Research Network (EDRN) phase of biomarker development. We break the early EDRN phases into sub-phases, proposing objectives, study design strategies, and biomarker performance benchmarks. RESULTS The goal of early detection in populations at high-risk of PDAC is described. Evaluating biomarker behavior in patients under surveillance without disease can winnow candidate biomarkers. Potential resources for biomarker validation studies are described. CONCLUSIONS Multisite and multidisciplinary collaboration can facilitate study design strategies in this lethal but low incidence disease and streamline the path from biomarker discovery to clinical use. Improvements in analytical and experimental design methods could help accelerate biomarker evaluation through the phases of biomarker development.
Collapse
Affiliation(s)
- Lynette M Smith
- Department of Biostatistics, University of Nebraska Medical Center, Omaha, NE, USA
| | - Douglas W Mahoney
- Department of Quantitative Health Sciences, Mayo Clinic, Rochester, MN, USA
| | - William R Bamlet
- Department of Quantitative Health Sciences, Mayo Clinic, Rochester, MN, USA
| | - Fang Yu
- Department of Biostatistics, University of Nebraska Medical Center, Omaha, NE, USA
| | - Suyu Liu
- Department of Biostatistics, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Michael G Goggins
- Departments of Pathology and Oncology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Sourat Darabi
- Hoag Family Cancer Institute, Newport Beach, CA, USA
| | - Shounak Majumder
- Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, MN, USA
| | - Qiao-Li Wang
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | - Gregory A Coté
- Department of Medicine, Oregon Health & Science University, Portland, OR, USA
| | | | - Zhen Zhang
- Departments of Pathology and Oncology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | | | - Akhil Chawla
- Department of Surgery, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | | | - Janet E Olson
- Department of Quantitative Health Sciences, Mayo Clinic, Rochester, MN, USA
| | - Brian M Wolpin
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | - Jeanine M Genkinger
- Department of Epidemiology, Mailman School of Public Health, Columbia University, NY, NY, USA
| | - Kenneth S Zaret
- Department of Cell and Developmental Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Randall Brand
- Department of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | - Eugene J Koay
- Department of Gastrointestinal Radiation Oncology, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Ann L Oberg
- Department of Quantitative Health Sciences, Mayo Clinic, Rochester, MN, USA.
| |
Collapse
|
22
|
Zeng X, Ma Q, Huang CX, Xiao JJ, Fu X, Ren YF, Qu YL, Xiang HX, Lei M, Zheng RY, Zhong Y, Xiao P, Zhuang X, You FM, He JW. Diagnostic potential of salivary microbiota in persistent pulmonary nodules: identifying biomarkers and functional pathways using 16S rRNA sequencing and machine learning. J Transl Med 2024; 22:1079. [PMID: 39609902 PMCID: PMC11603953 DOI: 10.1186/s12967-024-05802-7] [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: 07/25/2024] [Accepted: 10/23/2024] [Indexed: 11/30/2024] Open
Abstract
BACKGROUND The aim of this study was to explore the microbial variations and biomarkers in the oral environment of patients with persistent pulmonary nodules (pPNs) and to reveal the potential biological functions of the salivary microbiota in pPNs. MATERIALS AND METHODS This study included a total of 483 participants (141 healthy controls and 342 patients with pPNs) from June 2022 and January 2024. Saliva samples were subjected to sequencing of the V3-V4 region of the 16S rRNA gene to assess microbial diversity and differential abundance. Seven advanced machine learning algorithms (logistic regression, support vector machine, multi-layer perceptron, naïve Bayes, random forest, gradient boosting decision tree, and LightGBM) were utilized to evaluate performance and identify key microorganisms, with fivefold cross-validation employed to ensure robustness. The Shapley Additive exPlanations (SHAP) algorithm was employed to explain the contribution of these core microbiotas to the predictive model. Additionally, the PICRUSt2 algorithm was used to predict the microbial functions. RESULTS The salivary microbial composition in pPNs group showed significantly lower α- and β-diversity compared to healthy controls. A high-accuracy LightGBM model was developed, identifying six core genera-Fusobacterium, Solobacterium, Actinomyces, Porphyromonas, Atopobium, and Peptostreptococcus-as pPNs biomarkers. Additionally, a visualization pPNs risk prediction system was developed. The immune responses and metabolic activities differences in salivary microbiota between the patients with pPNs and healthy controls were revealed. CONCLUSIONS This study highlights the potential clinical applications of the salivary microbiota for enable earlier detection and targeted interventions, offering significant promise for advancing clinical management and improving patient outcomes in pPNs.
Collapse
Affiliation(s)
- Xiao Zeng
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, 610072, Sichuan Province, China
| | - Qiong Ma
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, 610072, Sichuan Province, China
| | - Chun-Xia Huang
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, 610072, Sichuan Province, China
| | - Jun-Jie Xiao
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, 610072, Sichuan Province, China
| | - Xi Fu
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, 610072, Sichuan Province, China
- TCM Regulating Metabolic Diseases Key Laboratory of Sichuan Province, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, 610072, Sichuan Province, China
| | - Yi-Feng Ren
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, 610072, Sichuan Province, China
- TCM Regulating Metabolic Diseases Key Laboratory of Sichuan Province, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, 610072, Sichuan Province, China
| | - Yu-Li Qu
- College of Artificial Intelligence, Xi'an Jiaotong University, Xian, 710061, Shanxi Province, China
| | - Hong-Xia Xiang
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, 610072, Sichuan Province, China
| | - Mao Lei
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, 610072, Sichuan Province, China
| | - Ru-Yi Zheng
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, 610072, Sichuan Province, China
| | - Yang Zhong
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, 610072, Sichuan Province, China
| | - Ping Xiao
- Department of Thoracic Surgery, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, School of Medicine, University of Electronic Science and Technology of China, Chengdu, 610042, Sichuan Province, China
| | - Xiang Zhuang
- Department of Thoracic Surgery, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, School of Medicine, University of Electronic Science and Technology of China, Chengdu, 610042, Sichuan Province, China
| | - Feng-Ming You
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, 610072, Sichuan Province, China.
- TCM Regulating Metabolic Diseases Key Laboratory of Sichuan Province, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, 610072, Sichuan Province, China.
| | - Jia-Wei He
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, 610072, Sichuan Province, China.
| |
Collapse
|
23
|
El Abiad E, Al-Kuwari A, Al-Aani U, Al Jaidah Y, Chaari A. Navigating the Alzheimer's Biomarker Landscape: A Comprehensive Analysis of Fluid-Based Diagnostics. Cells 2024; 13:1901. [PMID: 39594648 PMCID: PMC11593284 DOI: 10.3390/cells13221901] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2024] [Revised: 11/12/2024] [Accepted: 11/14/2024] [Indexed: 11/28/2024] Open
Abstract
BACKGROUND Alzheimer's disease (AD) affects a significant portion of the aging population, presenting a serious challenge due to the limited availability of effective therapies during its progression. The disease advances rapidly, underscoring the need for early diagnosis and the application of preventative measures. Current diagnostic methods for AD are often expensive and invasive, restricting access for the general public. One potential solution is the use of biomarkers, which can facilitate early detection and treatment through objective, non-invasive, and cost-effective evaluations of AD. This review critically investigates the function and role of biofluid biomarkers in detecting AD, with a specific focus on cerebrospinal fluid (CSF), blood-based, and saliva biomarkers. RESULTS CSF biomarkers have demonstrated potential for accurate diagnosis and valuable prognostic insights, while blood biomarkers offer a minimally invasive and cost-effective approach for diagnosing cognitive issues. However, while current biomarkers for AD show significant potential, none have yet achieved the precision needed to replace expensive PET scans and CSF assays. The lack of a single accurate biomarker underscores the need for further research to identify novel or combined biomarkers to enhance the clinical efficacy of existing diagnostic tests. In this context, artificial intelligence (AI) and deep-learning (DL) tools present promising avenues for improving biomarker analysis and interpretation, enabling more precise and timely diagnoses. CONCLUSIONS Further research is essential to confirm the utility of all AD biomarkers in clinical settings. Combining biomarker data with AI tools offers a promising path toward revolutionizing the personalized characterization and early diagnosis of AD symptoms.
Collapse
Affiliation(s)
| | | | | | | | - Ali Chaari
- Weill Cornell Medicine–Qatar, Qatar Foundation, Education City, Doha P.O. Box 24144, Qatar; (E.E.A.); (A.A.-K.); (U.A.-A.); (Y.A.J.)
| |
Collapse
|
24
|
Park SH, Han K, Lee JG. Conceptual review of outcome metrics and measures used in clinical evaluation of artificial intelligence in radiology. LA RADIOLOGIA MEDICA 2024; 129:1644-1655. [PMID: 39225919 DOI: 10.1007/s11547-024-01886-9] [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: 06/01/2024] [Accepted: 08/21/2024] [Indexed: 09/04/2024]
Abstract
Artificial intelligence (AI) has numerous applications in radiology. Clinical research studies to evaluate the AI models are also diverse. Consequently, diverse outcome metrics and measures are employed in the clinical evaluation of AI, presenting a challenge for clinical radiologists. This review aims to provide conceptually intuitive explanations of the outcome metrics and measures that are most frequently used in clinical research, specifically tailored for clinicians. While we briefly discuss performance metrics for AI models in binary classification, detection, or segmentation tasks, our primary focus is on less frequently addressed topics in published literature. These include metrics and measures for evaluating multiclass classification; those for evaluating generative AI models, such as models used in image generation or modification and large language models; and outcome measures beyond performance metrics, including patient-centered outcome measures. Our explanations aim to guide clinicians in the appropriate use of these metrics and measures.
Collapse
Affiliation(s)
- Seong Ho Park
- Department of Radiology and Research Institute of Radiology, Asan Medical Center, University of Ulsan College of Medicine, 88, Olympic-Ro 43-Gil, Songpa-Gu, Seoul, 05505, South Korea.
| | - Kyunghwa Han
- Department of Radiology, Research Institute of Radiological Science and Center for Clinical Imaging Data Science, Yonsei University College of Medicine, Seoul, South Korea
| | - June-Goo Lee
- Biomedical Engineering Research Center, Asan Institute for Life Sciences, University of Ulsan College of Medicine, Seoul, South Korea
| |
Collapse
|
25
|
Singal AG, Parikh ND, Kanwal F, Marrero JA, Deodhar S, Page-Lester S, Lopez C, Feng Z, Tayob N. National Liver Cancer Screening Trial (TRACER) study protocol. Hepatol Commun 2024; 8:e0565. [PMID: 39495136 PMCID: PMC11537583 DOI: 10.1097/hc9.0000000000000565] [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: 08/03/2024] [Accepted: 09/11/2024] [Indexed: 11/05/2024] Open
Abstract
BACKGROUND Professional guidelines recommend HCC screening in at-risk patients using semi-annual ultrasound with or without alpha-fetoprotein (AFP); however, this strategy has limited effectiveness due to low adherence and sensitivity. Increasing data support the potential role of blood-based biomarker panels, which could improve both aspects. The biomarker panel GALAD, comprised of sex, age, and 3 blood biomarkers (AFP, AFP-L3, and des-carboxy prothrombin des-carboxy prothrombin), has shown high sensitivity and specificity in biomarker phase II (case-control) and phase III (retrospective cohort) validation studies. However, prospective validation in a large phase IV biomarker clinical utility trial is necessary before its adoption in practice. METHODS The National Liver Cancer Screening Trial is an adaptive pragmatic randomized phase IV trial, which began enrollment in January 2024, comparing ultrasound-based versus biomarker-based screening in 5500 patients with chronic hepatitis B infection or cirrhosis from any etiology. Eligible patients are randomly assigned in a 1:1 ratio to semi-annual screening with ultrasound ± alpha-fetoprotein (arm A) or semi-annual screening with GALAD (arm B). Randomization is stratified by enrollment site, liver disease severity (per Child-Pugh class), liver disease etiology (viral, nonviral, and noncirrhotic HBV), and sex. Patients are being recruited from 15 sites (a mix of tertiary care academic referral centers, safety-net health systems, and large community health systems) over a 3-year period, and the primary endpoint, reduction in late-stage HCC, will be assessed at the end of year 5.5. DISCUSSION The results of this trial will inform the best strategy for HCC screening and early-stage detection in patients with chronic liver diseases. If GALAD shows superiority, HCC screening would primarily shift from an ultrasound-based strategy to the adoption of the biomarker panel. TRIAL REGISTRATION NCT06084234. TRIAL STATUS The TRACER Study is actively enrolling.
Collapse
Affiliation(s)
- Amit G. Singal
- Department of Internal Medicine, UT Southwestern Medical Center, Dallas, Texas, USA
| | - Neehar D. Parikh
- Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | - Fasiha Kanwal
- Department of Medicine, Baylor College of Medicine, Houston, Texas, USA
| | - Jorge A. Marrero
- Department of Internal Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Sneha Deodhar
- Department of Internal Medicine, UT Southwestern Medical Center, Dallas, Texas, USA
| | - Stephanie Page-Lester
- Biostatistics Program, Public Health Science Division, Fred Hutchinson Cancer Research Center, Seattle, Washington, USA
| | - Camden Lopez
- Biostatistics Program, Public Health Science Division, Fred Hutchinson Cancer Research Center, Seattle, Washington, USA
| | - Ziding Feng
- Biostatistics Program, Public Health Science Division, Fred Hutchinson Cancer Research Center, Seattle, Washington, USA
| | - Nabihah Tayob
- Department of Data Science, Dana Farber Cancer Institute, Boston, Massachusetts, USA
| |
Collapse
|
26
|
Li P, Huang D. NSUN2-mediated RNA methylation: Molecular mechanisms and clinical relevance in cancer. Cell Signal 2024; 123:111375. [PMID: 39218271 DOI: 10.1016/j.cellsig.2024.111375] [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: 07/06/2024] [Revised: 08/26/2024] [Accepted: 08/29/2024] [Indexed: 09/04/2024]
Abstract
Cancer remains a leading cause of morbidity and mortality worldwide, necessitating the ongoing investigation of molecular targets for improved diagnosis, prognosis, and therapy. Among these targets, RNA modifications, particularly N5-methylcytosine (m5C) in RNA, have emerged as critical regulators of gene expression and cellular functions. NOP2/Sun RNA methyltransferase family member 2 (NSUN2) is a key enzyme in m5C modification, significantly influencing various biological processes and tumorigenesis. NSUN2 methylates multiple RNA species, including transfer RNAs (tRNAs), messenger RNAs (mRNAs), and non-coding RNAs, impacting RNA stability, translation efficiency, and cellular stress responses. These modifications, in turn, affect cell proliferation, differentiation, and survival. In cancer, NSUN2 is frequently upregulated, associated with aggressive tumor phenotypes, poor prognosis, and therapy resistance. Its role in oncogenic signaling pathways further underscores its importance in cancer biology. This review offers a comprehensive overview of NSUN2's role in cancer, focusing on its involvement in RNA methylation and its implications for tumor initiation and progression. Additionally, we explore the potential of NSUN2 as a biomarker for cancer diagnosis and prognosis, and its promise as a therapeutic target.
Collapse
Affiliation(s)
- Penghui Li
- Department of gastrointestinal surgery, The First Affiliated Hospital, College of Clinical Medicine, Henan University of Science and Technology, Luoyang 471000, Henan, China.
| | - Di Huang
- Department of Child Health Care, The Third Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, Henan, China
| |
Collapse
|
27
|
Singal AG, Ng M, Kulkarni A. Advancing Surveillance Strategies for Hepatocellular Carcinoma: A New Era of Efficacy and Precision. J Clin Exp Hepatol 2024; 14:101448. [PMID: 38946864 PMCID: PMC11214318 DOI: 10.1016/j.jceh.2024.101448] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/01/2024] [Accepted: 05/13/2024] [Indexed: 07/02/2024] Open
Abstract
Hepatocellular carcinoma (HCC) is one of the few cancers with a 5-year survival that has remained below 20%; however, prognosis differs by tumor stage at diagnosis. Curative treatment options among patients with early-stage HCC afford a median survival of 5-10 years. Accordingly, international society guidelines recommend semi-annual HCC surveillance in at-risk patients, including those with cirrhosis or high-risk chronic hepatitis B infection. Surveillance is associated with increased early-stage HCC detection and curative treatments, leading to reduced HCC-related mortality. Abdominal ultrasound has been the cornerstone for HCC surveillance for the past two decades, but recent data have highlighted its suboptimal sensitivity for early-stage HCC detection, particularly in patients with obesity and those with non-viral etiologies of liver disease. The combination of ultrasound plus alpha fetoprotein (AFP) has higher sensitivity for early-stage HCC detection than ultrasound alone, although the combination still misses over one-third of HCC at an early stage. Emerging imaging and blood-based biomarker strategies have promising data in biomarker phase 2 (case-control) and phase 3 (cohort) studies. Beyond ultrasound, Magnetic resonance imaging (MRI) is the best-studied imaging strategy, with superior sensitivity and specificity compared to ultrasound in a cohort study. Abbreviated MRI protocols have been proposed to address concerns about MRI radiological capacity, costs, and patient acceptance. Of biomarker strategies, GALAD (a panel including gender, age, AFP, AFP-L3, and DCP) is the best validated, with promising sensitivity for early-stage HCC detection in a national multi-center cohort study. Liquid biopsy biomarkers, including methylated DNA markers, have also shown promising accuracy in case-control studies. Abbreviated MRI and GALAD are now entering prospective trials that examine clinical outcomes such as early-stage HCC detection and screening-related harms, which are essential data to understand for adoption in clinical practice. As additional surveillance strategies become available, it will allow an era of precision surveillance in which optimal surveillance modalities are tailored to individual patient risk and expected test performance.
Collapse
Affiliation(s)
- Amit G. Singal
- Department of Internal Medicine, UT Southwestern Medical Center, Dallas, TX, USA
| | - Michelle Ng
- Department of Internal Medicine, UT Southwestern Medical Center, Dallas, TX, USA
| | - Anand Kulkarni
- Department of Hepatology, AIG Hospitals, Hyderabad, India
| |
Collapse
|
28
|
Liss MA, Zeltser N, Zheng Y, Lopez C, Liu M, Patel Y, Yamaguchi TN, Eng SE, Tian M, Semmes OJ, Lin DW, Brooks JD, Wei JT, Klein EA, Tewari AK, Mosquera JM, Khani F, Robinson BD, Aasad M, Troyer DA, Kagan J, Sanda MG, Thompson IM, Boutros PC, Leach RJ. Upgrading of Grade Group 1 Prostate Cancer at Prostatectomy: Germline Risk Factors in a Prospective Cohort. Cancer Epidemiol Biomarkers Prev 2024; 33:1500-1511. [PMID: 39158404 PMCID: PMC11528207 DOI: 10.1158/1055-9965.epi-24-0326] [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: 02/29/2024] [Revised: 05/21/2024] [Accepted: 08/14/2024] [Indexed: 08/20/2024] Open
Abstract
BACKGROUND Localized prostate tumors show significant spatial heterogeneity, with regions of high-grade disease adjacent to lower grade disease. Consequently, prostate cancer biopsies are prone to sampling bias, potentially leading to underestimation of tumor grade. To study the clinical, epidemiologic, and molecular hallmarks of this phenomenon, we conducted a prospective study of grade upgrading: differences in detected prostate cancer grade between biopsy and surgery. METHODS We established a prospective, multi-institutional cohort of men with grade group 1 (GG1) prostate cancer on biopsy who underwent radical prostatectomy. Upgrading was defined as detection of GG2+ in the resected tumor. Germline DNA from 192 subjects was subjected to whole-genome sequencing to quantify ancestry, pathogenic variants in DNA damage response genes, and polygenic risk. RESULTS Of 285 men, 67% upgraded at surgery. PSA density and percent of cancer in pre-prostatectomy positive biopsy cores were significantly associated with upgrading. No assessed genetic risk factor was predictive of upgrading, including polygenic risk scores for prostate cancer diagnosis. CONCLUSIONS In a cohort of patients with low-grade prostate cancer, a majority upgraded at radical prostatectomy. PSA density and percent of cancer in pre-prostatectomy positive biopsy cores portended the presence of higher-grade disease, while germline genetics was not informative in this setting. Patients with low-risk prostate cancer, but elevated PSA density or percent cancer in positive biopsy cores, may benefit from repeat biopsy, additional imaging or other approaches to complement active surveillance. IMPACT Further risk stratification of patients with low-risk prostate cancer may provide useful context for active surveillance decision-making.
Collapse
Affiliation(s)
- Michael A. Liss
- Department of Urology, University of Texas Health San Antonio, San Antonio, Texas
| | - Nicole Zeltser
- Department of Human Genetics, University of California Los Angeles, Los Angeles, California
- Jonsson Comprehensive Cancer Center, University of California Los Angeles, Los Angeles, California
| | - Yingye Zheng
- Department of Biostatistics, Fred Hutchinson Cancer Center, Seattle, Washington
| | - Camden Lopez
- Department of Biostatistics, Fred Hutchinson Cancer Center, Seattle, Washington
| | - Menghan Liu
- Department of Biostatistics, Fred Hutchinson Cancer Center, Seattle, Washington
| | - Yash Patel
- Jonsson Comprehensive Cancer Center, University of California Los Angeles, Los Angeles, California
- Institute of Precision Health, University of California Los Angeles, Los Angeles, California
| | - Takafumi N. Yamaguchi
- Department of Human Genetics, University of California Los Angeles, Los Angeles, California
- Jonsson Comprehensive Cancer Center, University of California Los Angeles, Los Angeles, California
- Institute of Precision Health, University of California Los Angeles, Los Angeles, California
| | - Stefan E. Eng
- Jonsson Comprehensive Cancer Center, University of California Los Angeles, Los Angeles, California
| | - Mao Tian
- Department of Human Genetics, University of California Los Angeles, Los Angeles, California
- Jonsson Comprehensive Cancer Center, University of California Los Angeles, Los Angeles, California
- Institute of Precision Health, University of California Los Angeles, Los Angeles, California
| | - Oliver J. Semmes
- Department of Microbiology and Molecular Cell Biology, Leroy T. Canoles Jr. Cancer Research Center, Eastern Virginia Medical School, Norfolk, Virginia
| | - Daniel W. Lin
- Division of Public Health Sciences, Department of Urology, Fred Hutchinson Cancer Center, University of Washington, Seattle, Washington
| | - James D. Brooks
- Department of Urology, Stanford University, Palo Alto, California
| | - John T. Wei
- Department of Urology, University of Michigan, Ann Arbor, Michigan
| | - Eric A. Klein
- Glickman Urological and Kidney Institute, Cleveland Clinic Lerner College of Medicine, Cleveland, Ohio
| | - Ashutosh K. Tewari
- Department of Urology, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Juan Miguel Mosquera
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, New York
| | - Francesca Khani
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, New York
| | - Brian D. Robinson
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, New York
| | - Muhammad Aasad
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, New York
| | - Dean A. Troyer
- Department of Microbiology and Molecular Cell Biology, Leroy T. Canoles Jr. Cancer Research Center, Eastern Virginia Medical School, Norfolk, Virginia
- Department of Pathology, University of Texas Health San Antonio, San Antonio, Texas
| | - Jacob Kagan
- Division of Cancer Prevention, National Cancer Institute, Bethesda, Maryland
| | | | - Ian M. Thompson
- The Children’s Hospital of San Antonio Foundation and Christus Health, San Antonio, Texas
| | - Paul C. Boutros
- Department of Human Genetics, University of California Los Angeles, Los Angeles, California
- Jonsson Comprehensive Cancer Center, University of California Los Angeles, Los Angeles, California
- Institute of Precision Health, University of California Los Angeles, Los Angeles, California
- Department of Urology, University of California Los Angeles, Los Angeles, California
| | - Robin J. Leach
- Department of Cell Systems and Anatomy, University of Texas Health San Antonio, San Antonio, Texas
- Department of Pediatrics, University of Texas Health San Antonio, San Antonio, Texas
| |
Collapse
|
29
|
Gell M, Noble S, Laumann TO, Nelson SM, Tervo-Clemmens B. Psychiatric neuroimaging designs for individualised, cohort, and population studies. Neuropsychopharmacology 2024; 50:29-36. [PMID: 39143320 PMCID: PMC11525483 DOI: 10.1038/s41386-024-01918-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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/01/2024] [Revised: 05/30/2024] [Accepted: 06/11/2024] [Indexed: 08/16/2024]
Abstract
Psychiatric neuroimaging faces challenges to rigour and reproducibility that prompt reconsideration of the relative strengths and limitations of study designs. Owing to high resource demands and varying inferential goals, current designs differentially emphasise sample size, measurement breadth, and longitudinal assessments. In this overview and perspective, we provide a guide to the current landscape of psychiatric neuroimaging study designs with respect to this balance of scientific goals and resource constraints. Through a heuristic data cube contrasting key design features, we discuss a resulting trade-off among small sample, precision longitudinal studies (e.g., individualised studies and cohorts) and large sample, minimally longitudinal, population studies. Precision studies support tests of within-person mechanisms, via intervention and tracking of longitudinal course. Population studies support tests of generalisation across multifaceted individual differences. A proposed reciprocal validation model (RVM) aims to recursively leverage these complementary designs in sequence to accumulate evidence, optimise relative strengths, and build towards improved long-term clinical utility.
Collapse
Affiliation(s)
- Martin Gell
- Department of Psychiatry, Psychotherapy and Psychosomatics, Faculty of Medicine, RWTH Aachen University, Aachen, Germany.
- Institute of Neuroscience and Medicine (INM-7: Brain & Behaviour), Research Centre Jülich, Jülich, Germany.
- Masonic Institute for the Developing Brain, University of Minnesota, Minneapolis, MN, USA.
| | - Stephanie Noble
- Psychology Department, Northeastern University, Boston, MA, USA
- Bioengineering Department, Northeastern University, Boston, MA, USA
- Center for Cognitive and Brain Health, Northeastern University, Boston, MA, USA
| | - Timothy O Laumann
- Department of Psychiatry, Washington University School of Medicine, St. Louis, MO, USA
| | - Steven M Nelson
- Masonic Institute for the Developing Brain, University of Minnesota, Minneapolis, MN, USA
- Department of Pediatrics, University of Minnesota, Minneapolis, MN, USA
| | - Brenden Tervo-Clemmens
- Masonic Institute for the Developing Brain, University of Minnesota, Minneapolis, MN, USA.
- Department of Psychiatry and Behavioral Sciences, University of Minnesota, Minneapolis, MN, USA.
- Institute for Translational Neuroscience, University of Minnesota, Minneapolis, MN, USA.
| |
Collapse
|
30
|
Hamashima C, Fukao A. Failure of evaluation on ABC classification for gastric cancer screening. Gastric Cancer 2024; 27:1346-1347. [PMID: 39245690 DOI: 10.1007/s10120-024-01550-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/03/2024] [Accepted: 08/24/2024] [Indexed: 09/10/2024]
Affiliation(s)
- Chisato Hamashima
- Health Policy Section, Department of Nursing, Faculty of Medical Technology, Teikyo University, 2-11-1 Kaga, Itabashi-Ku, Tokyo, 173-1211, Japan.
| | | |
Collapse
|
31
|
Tylden ES, Delgado AB, Lukic M, Moi L, Busund LTR, Pedersen MI, Lombardi AP, Olsen KS. Roles of miR-20a-5p in breast cancer based on the clinical and multi-omic (CAMO) cohort and in vitro studies. Sci Rep 2024; 14:25022. [PMID: 39443510 PMCID: PMC11499649 DOI: 10.1038/s41598-024-75557-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2024] [Accepted: 10/07/2024] [Indexed: 10/25/2024] Open
Abstract
MicroRNAs are involved in breast cancer development and progression, holding potential as biomarkers and therapeutic targets or tools. The roles of miR-20a-5p, a member of the oncogenic miR-17-92 cluster, remain poorly understood in the context of breast cancer. In this study, we elucidate the role of miR-20a-5p in breast cancer by examining its associations with breast cancer risk factors and clinicopathological features, and its functional roles in vitro. Tissue microarrays from 313 CAMO cohort breast cancer surgical specimens were constructed, in situ hybridization was performed and miR-20a-5p expression was semiquantitatively scored in tumor stromal fibroblasts, and in the cytoplasm and nuclei of cancer cells. In vitro analysis of the effect of miR-20a-5p transfection on proliferation, migration and invasion was performed in three breast cancer cell lines. High stromal miR-20a-5p was associated with higher Ki67 expression, and higher odds of relapse, compared to low expression. Compared to postmenopausal women, women who were premenopausal at diagnosis had higher odds of high stromal and cytoplasmic miR-20a-5p expression. Cytoplasmic miR-20a-5p was significantly associated with tumor grade. In tumors with high cytoplasmic miR-20a-5p expression compared to low expression, there was a tendency towards having a basal-like subtype and high Ki67. In contrast, high nuclear miR-20a-5p in cancer cells was associated with smaller tumor size and lower odds of lymph node metastasis, compared to low nuclear expression. Transfection with miR-20a-5p in breast cancer cell lines led to increased migration and invasion in vitro. While the majority of our results point towards an oncogenic role, some of our findings indicate that the associations of miR-20a-5p with breast cancer related risk factors and outcomes may vary based on tissue- and subcellular location. Larger studies are needed to validate our findings and further investigate the clinical utility of miR-20a-5p.
Collapse
Affiliation(s)
- Eline Sol Tylden
- Department of Medical Biology, Faculty of Health Sciences, UiT The Arctic University of Norway, Tromso, Norway
| | - André Berli Delgado
- Department of Medical Biology, Faculty of Health Sciences, UiT The Arctic University of Norway, Tromso, Norway
| | - Marko Lukic
- Department of Community Medicine, Faculty of Health Sciences, UiT The Arctic University of Norway, Tromso, Norway
| | - Line Moi
- Department of Clinical Pathology, University Hospital of North Norway, Tromso, Norway
| | - Lill-Tove Rasmussen Busund
- Department of Medical Biology, Faculty of Health Sciences, UiT The Arctic University of Norway, Tromso, Norway
- Department of Clinical Pathology, University Hospital of North Norway, Tromso, Norway
| | - Mona Irene Pedersen
- Department of Clinical Medicine, Faculty of Health Sciences, UiT The Arctic University of Norway, Tromso, Norway
| | - Ana Paola Lombardi
- Department of Medical Biology, Faculty of Health Sciences, UiT The Arctic University of Norway, Tromso, Norway
| | - Karina Standahl Olsen
- Department of Community Medicine, Faculty of Health Sciences, UiT The Arctic University of Norway, Tromso, Norway.
| |
Collapse
|
32
|
Attia AM, Rezaee-Zavareh MS, Hwang SY, Kim N, Adetyan H, Yalda T, Chen PJ, Koltsova EK, Yang JD. Novel Biomarkers for Early Detection of Hepatocellular Carcinoma. Diagnostics (Basel) 2024; 14:2278. [PMID: 39451600 PMCID: PMC11507329 DOI: 10.3390/diagnostics14202278] [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: 08/31/2024] [Revised: 10/08/2024] [Accepted: 10/12/2024] [Indexed: 10/26/2024] Open
Abstract
Hepatocellular carcinoma (HCC) is a leading cause of cancer mortality globally. Most patients present with late diagnosis, leading to poor prognosis. This narrative review explores novel biomarkers for early HCC detection. We conducted a comprehensive literature review analyzing protein, circulating nucleic acid, metabolite, and quantitative proteomics-based biomarkers, evaluating the advantages and limitations of each approach. While established markers like alpha-fetoprotein (AFP), des-gamma-carboxy prothrombin, and AFP-L3 remain relevant, promising candidates include circulating tumor DNA, microRNAs, long noncoding RNAs, extracellular vesicle, and metabolomic biomarkers. Multi-biomarker panels like the GALAD score, Oncoguard, and Helio liver test show promise for improved diagnostic accuracy. Non-invasive approaches like urine and gut microbiome analysis are also emerging possibilities. Integrating these novel biomarkers with current screening protocols holds significant potential for earlier HCC detection and improved patient outcomes. Future research should explore multi-biomarker panels, omics technologies, and artificial intelligence to further enhance early HCC diagnosis and management.
Collapse
Affiliation(s)
- Abdelrahman M. Attia
- Karsh Division of Gastroenterology and Hepatology, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA; (A.M.A.); (N.K.); (H.A.); (T.Y.)
| | | | - Soo Young Hwang
- Department of Internal Medicine, University of Maryland Medical Center, Midtown Campus, Baltimore, MD 21201, USA;
| | - Naomy Kim
- Karsh Division of Gastroenterology and Hepatology, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA; (A.M.A.); (N.K.); (H.A.); (T.Y.)
| | - Hasmik Adetyan
- Karsh Division of Gastroenterology and Hepatology, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA; (A.M.A.); (N.K.); (H.A.); (T.Y.)
| | - Tamar Yalda
- Karsh Division of Gastroenterology and Hepatology, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA; (A.M.A.); (N.K.); (H.A.); (T.Y.)
| | - Pin-Jung Chen
- Department of Hematology and Oncology, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA;
| | - Ekaterina K. Koltsova
- Cedars-Sinai Cancer, Smidt Heart Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA;
| | - Ju Dong Yang
- Karsh Division of Gastroenterology and Hepatology, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA; (A.M.A.); (N.K.); (H.A.); (T.Y.)
- Comprehensive Transplant Center, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
- Samuel Oschin Comprehensive Cancer Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| |
Collapse
|
33
|
Neagoe CXR, Ionică M, Neagoe OC, Trifa AP. The Influence of Microbiota on Breast Cancer: A Review. Cancers (Basel) 2024; 16:3468. [PMID: 39456562 PMCID: PMC11506631 DOI: 10.3390/cancers16203468] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2024] [Revised: 10/05/2024] [Accepted: 10/11/2024] [Indexed: 10/28/2024] Open
Abstract
Breast cancer remains one of the leading causes of death among women worldwide, and recent research highlights its growing connection to alterations in the microbiota. This review delves into the intricate relationship between microbiotas and breast cancer, exploring its presence in healthy breast tissue, its changes during cancer progression, and its considerable impact on both the tumor microenvironment (TME) and the tumor immune microenvironment (TIME). We extensively analyze how the microbiota influences cancer growth, invasion, metastasis, resistance to drugs, and the evasion of the immune system, with a special focus on its effects on the TIME. Furthermore, we investigate distinct microbial profiles associated with the four primary molecular subtypes of breast cancer, examining how the microbiota in tumor tissues compares with that in adjacent normal tissues. Emerging studies suggest that microbiotas could serve as valuable diagnostic and prognostic biomarkers, as well as targets for therapy. This review emphasizes the urgent need for further research to improve strategies for breast cancer prevention, diagnosis, and treatment. By offering a detailed examination of the microbiota's critical role in breast cancer, this review aims to foster the development of novel microbiota-based approaches for managing the disease.
Collapse
Affiliation(s)
- Cara-Xenia-Rafaela Neagoe
- Doctoral School, “Victor Babes” University of Medicine and Pharmacy, Eftimie Murgu Square No. 2, 300041 Timisoara, Romania;
| | - Mihaela Ionică
- Second Clinic of General Surgery and Surgical Oncology, Emergency Clinical Municipal Hospital, 300079 Timișoara, Romania;
- Second Discipline of Surgical Semiology, First Department of Surgery, “Victor Babeș” University of Medicine and Pharmacy, 300041 Timișoara, Romania
- Breast Surgery Research Center, “Victor Babeș” University of Medicine and Pharmacy, 300079 Timișoara, Romania
| | - Octavian Constantin Neagoe
- Second Clinic of General Surgery and Surgical Oncology, Emergency Clinical Municipal Hospital, 300079 Timișoara, Romania;
- Second Discipline of Surgical Semiology, First Department of Surgery, “Victor Babeș” University of Medicine and Pharmacy, 300041 Timișoara, Romania
- Breast Surgery Research Center, “Victor Babeș” University of Medicine and Pharmacy, 300079 Timișoara, Romania
| | - Adrian Pavel Trifa
- The Discipline of Genetics, “Victor Babes” University of Medicine and Pharmacy, 300041 Timisoara, Romania;
- Department of Genetics, Clinical Hospital of Infectious Diseases and Pneumophthisiology “Dr. Victor Babes” Timisoara, 300041 Timisoara, Romania
- Center for Research and Innovation in Personalized Medicine of Respiratory Diseases, “Victor Babeș” University of Medicine and Pharmacy, 300041 Timișoara, Romania
| |
Collapse
|
34
|
Chen B, Wang D, Xu Y, Guo Q, Pan J, Yu S, Fang Y, Xiao S, Ruan Y, Yang S, Lin M, Hong J, Zhan Z, Lin S. 5-Hydroxymethylcytosines in circulating cell-free DNA as a diagnostic biomarker for nasopharyngeal carcinoma. Eur J Cancer 2024; 210:114294. [PMID: 39213787 DOI: 10.1016/j.ejca.2024.114294] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2024] [Revised: 07/07/2024] [Accepted: 08/12/2024] [Indexed: 09/04/2024]
Abstract
OBJECTIVE To evaluate the diagnostic value of 5-hydroxymethylcytosines (5hmC) in circulating cell-free DNA (cfDNA) for nasopharyngeal carcinoma (NPC) and to develop a diagnostic model. METHODS Genome-wide 5hmC profiles in cfDNA from 174 NPC patients and 146 non-cancer individuals were analyzed using the 5hmC-Seal technique. A cfDNA 5hmC-based diagnostic model to identify NPC patients was developed using least absolute shrinkage and selection operator (LASSO) logistic regression, and performance was evaluated with receiver operating characteristic (ROC) curves and confusion matrices. RESULTS The 5hmC-Seal data from patients with NPC showed a different genome-wide distribution than non-tumor samples. Our initial analysis revealed a 12-gene-based 5hmC marker panel to be an accurate diagnostic model effectively distinguishing between NPC samples and non-cancerous samples (training set: area under curve (AUC)= 0.97 [95 % CI: 0.94-0.99]; and test set: AUC= 0.93 [95 % CI: 0.88-0.98]) superior to EBV DNA testing. The diagnostic score performed well in differentiating the non-cancer subjects from early-stage NPC (training set: AUC=0.99 [95 % CI: 0.98-1]; test set: AUC=0.98 [95 % CI: 0.95-1]), and advanced-stage NPC (training set: AUC=0.96 [95 % CI: 0.93-0.99]; test set: AUC=0.93 [95 % CI: 0.88-0.98]). Notably, in EBV-negative patients, the diagnostic scores showed excellent capacity for distinguishing EBV-negative patients with NPC from non-cancer subjects in both the training set (AUC= 0.94 [95 % CI: 0.88-1]) and test set (AUC=0.91 [95 % CI: 0.81-1]). CONCLUSION 5hmC modifications in cfDNA are promising noninvasive biomarkers for NPC, offering high sensitivity and specificity, particularly for early-stage and EBV-negative NPC.
Collapse
Affiliation(s)
- Bijuan Chen
- Department of Radiation Oncology, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fuzhou, Fujian 350014, China
| | - Di Wang
- Department of Molecular Pathology, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fuzhou, Fujian 350014, China
| | - Yun Xu
- Department of Radiation Oncology, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fuzhou, Fujian 350014, China
| | - Qiaojuan Guo
- Department of Radiation Oncology, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fuzhou, Fujian 350014, China
| | - Jianji Pan
- Department of Radiation Oncology, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fuzhou, Fujian 350014, China
| | - Sisi Yu
- Department of Radiation Oncology, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fuzhou, Fujian 350014, China
| | - Yunxiang Fang
- Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fuzhou, Fujian 350014, China
| | - Shuxiang Xiao
- Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fuzhou, Fujian 350014, China
| | - Yuanyuan Ruan
- Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fuzhou, Fujian 350014, China
| | - Shanshan Yang
- Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fuzhou, Fujian 350014, China
| | - Mingan Lin
- Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fuzhou, Fujian 350014, China
| | - Jinsheng Hong
- Department of Radiotherapy, Cancer Center, The First Affiliated Hospital of Fujian Medical University, Fuzhou 350005, China; Department of Radiotherapy, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou 350212, China; Key Laboratory of Radiation Biology of Fujian Higher Education Institutions, The First Affiliated Hospital, Fujian Medical University, Fuzhou 350005, China.
| | - Zhouwei Zhan
- Department of Medical Oncology, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fuzhou, Fujian 350014, China.
| | - Shaojun Lin
- Department of Radiation Oncology, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fuzhou, Fujian 350014, China.
| |
Collapse
|
35
|
Orive D, Echepare M, Bernasconi-Bisio F, Sanmamed MF, Pineda-Lucena A, de la Calle-Arroyo C, Detterbeck F, Hung RJ, Johansson M, Robbins HA, Seijo LM, Montuenga LM, Valencia K. Protein Biomarkers in Lung Cancer Screening: Technical Considerations and Feasibility Assessment. Arch Bronconeumol 2024; 60 Suppl 2:S67-S76. [PMID: 39079848 DOI: 10.1016/j.arbres.2024.07.007] [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: 04/16/2024] [Revised: 06/28/2024] [Accepted: 07/12/2024] [Indexed: 08/25/2024]
Abstract
Lung cancer remains the leading cause of cancer-related deaths worldwide, mainly due to late diagnosis and the presence of metastases. Several countries around the world have adopted nation-wide LDCT-based lung cancer screening that will benefit patients, shifting the stage at diagnosis to earlier stages with more therapeutic options. Biomarkers can help to optimize the screening process, as well as refine the TNM stratification of lung cancer patients, providing information regarding prognostics and recommending management strategies. Moreover, novel adjuvant strategies will clearly benefit from previous knowledge of the potential aggressiveness and biological traits of a given early-stage surgically resected tumor. This review focuses on proteins as promising biomarkers in the context of lung cancer screening. Despite great efforts, there are still no successful examples of biomarkers in lung cancer that have reached the clinics to be used in early detection and early management. Thus, the field of biomarkers in early lung cancer remains an evident unmet need. A more specific objective of this review is to present an up-to-date technical assessment of the potential use of protein biomarkers in early lung cancer detection and management. We provide an overview regarding the benefits, challenges, pitfalls and constraints in the development process of protein-based biomarkers. Additionally, we examine how a number of emerging protein analytical technologies may contribute to the optimization of novel robust biomarkers for screening and effective management of lung cancer.
Collapse
Affiliation(s)
- Daniel Orive
- Solid Tumors Program, CIMA-University of Navarra, Pamplona, Spain; Department of Pathology, Anatomy and Physiology, School of Medicine, University of Navarra, Pamplona, Spain; Consorcio de Investigación Biomédica en Red de Cáncer (CIBERONC), Madrid, Spain
| | - Mirari Echepare
- Solid Tumors Program, CIMA-University of Navarra, Pamplona, Spain; Department of Pathology, Anatomy and Physiology, School of Medicine, University of Navarra, Pamplona, Spain; Consorcio de Investigación Biomédica en Red de Cáncer (CIBERONC), Madrid, Spain; Navarra Health Research Institute (IDISNA), Pamplona, Spain
| | - Franco Bernasconi-Bisio
- Molecular Therapeutics Program, CIMA-University of Navarra, Pamplona, Spain; Department of Biochemistry and Genetics, School of Sciences, University of Navarra, Pamplona, Spain
| | - Miguel Fernández Sanmamed
- Consorcio de Investigación Biomédica en Red de Cáncer (CIBERONC), Madrid, Spain; Program of Immunology and Immunotherapy, CIMA-University of Navarra, Pamplona, Spain; Department of Oncology, Clínica Universidad de Navarra, Pamplona, Spain
| | - Antonio Pineda-Lucena
- Navarra Health Research Institute (IDISNA), Pamplona, Spain; Molecular Therapeutics Program, CIMA-University of Navarra, Pamplona, Spain; Department of Biochemistry and Genetics, School of Sciences, University of Navarra, Pamplona, Spain
| | - Carlos de la Calle-Arroyo
- Instituto de Ciencia de los Datos e Inteligencia Artificial (DATAI), Universidad de Navarra, Pamplona, Spain
| | - Frank Detterbeck
- Division of Thoracic Surgery, Department of Surgery, Yale School of Medicine, New Haven, CT, USA
| | - Rayjean J Hung
- Prosserman Centre for Population Health Research, Lunenfeld-Tanenbaum Research Institute, Sinai Health, Toronto, Canada; Dalla Lana School of Public Health, University of Toronto, Toronto, Canada
| | | | | | - Luis M Seijo
- CIBER of Respiratory Diseases (CIBERES), Institute of Health Carlos III, Madrid, Spain; Pulmonary Department, Clínica Universidad de Navarra, Madrid, Spain
| | - Luis M Montuenga
- Solid Tumors Program, CIMA-University of Navarra, Pamplona, Spain; Department of Pathology, Anatomy and Physiology, School of Medicine, University of Navarra, Pamplona, Spain; Consorcio de Investigación Biomédica en Red de Cáncer (CIBERONC), Madrid, Spain; Navarra Health Research Institute (IDISNA), Pamplona, Spain.
| | - Karmele Valencia
- Solid Tumors Program, CIMA-University of Navarra, Pamplona, Spain; Consorcio de Investigación Biomédica en Red de Cáncer (CIBERONC), Madrid, Spain; Navarra Health Research Institute (IDISNA), Pamplona, Spain; Department of Biochemistry and Genetics, School of Sciences, University of Navarra, Pamplona, Spain.
| |
Collapse
|
36
|
Jensen SØ, Moore DA, Surani AA, Crosbie PAJ, Rosenfeld N, Rintoul RC. Second Primary Lung Cancer - An Emerging Issue in Lung Cancer Survivors. J Thorac Oncol 2024; 19:1415-1426. [PMID: 39059487 DOI: 10.1016/j.jtho.2024.07.014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2024] [Revised: 06/22/2024] [Accepted: 07/18/2024] [Indexed: 07/28/2024]
Abstract
As a result of an increased focus on early detection including lung cancer screening, combined with more curative treatment options, the 5-year survival rates for lung cancer are improving. Welcome though this is, it brings new, hitherto unseen challenges. As more patients are cured and survive longer, they are at risk of developing second primary cancers, particularly lung cancer. In this review, we examine the challenges that surveillance, diagnosis, and management of second primary lung cancer (SPLC) bring and how these can be addressed. Recent data from prospective follow-up studies suggests that the incidence of SPLC may be higher than previously appreciated, partly due to an increase in multi-focal adenocarcinoma spectrum disease. Over 5 years, up to 1 in 6 long-term lung cancer survivors may develop a SPLC. Although not routinely used in clinical practice at present, genomic approaches for differentiating SPLC from intrapulmonary metastases of the first primary are emerging, and we highlight how this could be used to help differentiate lesions. An accurate distinction between SPLC and the recurrence of the first primary is of paramount importance due to the very different management strategies that may be required. Wrongly classifying an SPLC as a recurrence of the first primary may have significant consequences for patient management and overall survival. Updated approaches to the classification of SPLC combining clinical history, histopathological assessment, and genomic profiling are needed. Finally, we review the potential role of early detection biomarkers in the identification of SPLC, focusing in particular on blood-based biomarkers that are being examined in a multi-center prospective study recruiting lung cancer survivors.
Collapse
Affiliation(s)
- Sarah Østrup Jensen
- Cancer Research UK Cambridge Institute, University of Cambridge, Cambridge, United Kingdom; Cancer Research UK Cambridge Centre, University of Cambridge, Cambridge, United Kingdom
| | - David A Moore
- Department of Cellular Pathology, University College Hospital, London United Kingdom; Cancer Research UK Lung Cancer Centre of Excellence, University College London Cancer Institute, London, United Kingdom
| | - Arif A Surani
- Cancer Research UK Cambridge Institute, University of Cambridge, Cambridge, United Kingdom; Cancer Research UK Cambridge Centre, University of Cambridge, Cambridge, United Kingdom
| | - Philip A J Crosbie
- Division of Immunology, Immunity and Infection and Respiratory Medicine, University of Manchester, Manchester, United Kingdom
| | - Nitzan Rosenfeld
- Cancer Research UK Cambridge Institute, University of Cambridge, Cambridge, United Kingdom; Barts Cancer Institute, Queen Mary University of London, London, United Kingdom
| | - Robert C Rintoul
- Cancer Research UK Cambridge Institute, University of Cambridge, Cambridge, United Kingdom; Cancer Research UK Cambridge Centre, University of Cambridge, Cambridge, United Kingdom; Department of Oncology, University of Cambridge, Cambridge, United Kingdom; Department of Thoracic Oncology, Royal Papworth Hospital, Cambridge, United Kingdom.
| |
Collapse
|
37
|
Gasparri R, Papale M, Sabalic A, Catalano V, Deleonardis A, De Luca F, Ranieri E, Spaggiari L. Circulating RKIP and pRKIP in Early-Stage Lung Cancer: Results from a Pilot Study. J Clin Med 2024; 13:5830. [PMID: 39407890 PMCID: PMC11476948 DOI: 10.3390/jcm13195830] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2024] [Revised: 09/15/2024] [Accepted: 09/24/2024] [Indexed: 10/20/2024] Open
Abstract
Background: Lung cancer (LC) is the leading cause of cancer-related deaths. Although low-dose computed tomography (LD-CT) reduces mortality, its clinical use is limited by cost, radiation, and false positives. Therefore, there is an urgent need for non-invasive and cost-effective biomarkers. The Raf Kinase Inhibitor Protein (RKIP) plays a crucial role in cancer development and progression and may also contribute to regulating the tumor-immune system axis. This protein has recently been described in biological fluids. Therefore, we conducted a pilot case-control study to assess RKIP and phosphorylated RKIP (pRKIP) levels in the urine and blood of LC patients. Methods: A novel enzyme linked immunosorbent assay (ELISA) assay was used to measure RKIP and pRKIP levels in urine and blood samples of two cohorts of LC patients and healthy controls (HSs). Furthermore, the biomarkers levels were correlated with tumor characteristics. Results: Serum, but not urine, levels of RKIP were significantly elevated in LC patients, distinguishing them from low- and high-risk healthy subjects with 93% and 74% accuracy, respectively. The RKIP/pRKIP ratio (RpR score) showed an accuracy of 90% and 79% in distinguishing LC patients from HS and HR-HS, respectively. Additionally, the RpR score correlated better with dimension, stage, and lymph node involvement in the tumor group. Conclusions: The serum RKIP and pRKIP profile may be a promising novel biomarker for early-stage LC.
Collapse
Affiliation(s)
- Roberto Gasparri
- Department of Thoracic Surgery, European Institute of Oncology (IEO), IRCCS, 20141 Milan, Italy; (R.G.); (L.S.)
| | - Massimo Papale
- Unit of Clinical Pathology, Department of Laboratory Diagnostics, University Hospital “Policlinico Foggia”, 71122 Foggia, Italy
| | - Angela Sabalic
- Department of Thoracic Surgery, European Institute of Oncology (IEO), IRCCS, 20141 Milan, Italy; (R.G.); (L.S.)
| | - Valeria Catalano
- Unit of Clinical Pathology, Advanced Research Center on Kidney Aging (A.R.K.A.), Department of Medical and Surgical Sciences, University of Foggia, Viale Luigi Pinto, 71122 Foggia, Italy; (V.C.); (F.D.L.); (E.R.)
| | - Annamaria Deleonardis
- Nephrology, Dialysis and Transplantation Unit, Department of Precision and Regenerative Medicine and Ionian Area (DiMePRe-J), University of Bari “Aldo Moro”, 70121 Bari, Italy;
- R&D Unit, Fluidia s.r.l., 71122 Foggia, Italy
| | - Federica De Luca
- Unit of Clinical Pathology, Advanced Research Center on Kidney Aging (A.R.K.A.), Department of Medical and Surgical Sciences, University of Foggia, Viale Luigi Pinto, 71122 Foggia, Italy; (V.C.); (F.D.L.); (E.R.)
| | - Elena Ranieri
- Unit of Clinical Pathology, Advanced Research Center on Kidney Aging (A.R.K.A.), Department of Medical and Surgical Sciences, University of Foggia, Viale Luigi Pinto, 71122 Foggia, Italy; (V.C.); (F.D.L.); (E.R.)
| | - Lorenzo Spaggiari
- Department of Thoracic Surgery, European Institute of Oncology (IEO), IRCCS, 20141 Milan, Italy; (R.G.); (L.S.)
- Department of Oncology and Hemato-Oncology, University of Milan, 20141 Milan, Italy
| |
Collapse
|
38
|
Zheng Y, Wagner PD, Singal AG, Hanash SM, Srivastava S, Huang Y, Zhao YQ, Chari ST, Marquez G, Etizioni R, Marsh TL, Feng Z. Designing Rigorous and Efficient Clinical Utility Studies for Early Detection Biomarkers. Cancer Epidemiol Biomarkers Prev 2024; 33:1150-1157. [PMID: 39223980 PMCID: PMC11534000 DOI: 10.1158/1055-9965.epi-23-1594] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2023] [Revised: 03/11/2024] [Accepted: 06/07/2024] [Indexed: 09/04/2024] Open
Abstract
Before implementing a biomarker in routine clinical care, it must demonstrate clinical utility by leading to clinical actions that positively affect patient-relevant outcomes. Randomly controlled early detection utility trials, especially those targeting mortality endpoint, are challenging due to their high costs and prolonged duration. Special design considerations are required to determine the clinical utility of early detection assays. This commentary reports on discussions among the National Cancer Institute's Early Detection Research Network investigators, outlining the recommended process for carrying out single-organ biomarker-driven clinical utility studies. We present the early detection utility studies in the context of phased biomarker development. We describe aspects of the studies related to the features of biomarker tests, the clinical context of endpoints, the performance criteria for later phase evaluation, and study size. We discuss novel adaptive design approaches for improving the efficiency and practicality of clinical utility trials. We recommend using multiple strategies, including adopting real-world evidence, emulated trials, and mathematical modeling to circumvent the challenges in conducting early detection utility trials.
Collapse
Affiliation(s)
- Yingye Zheng
- Biostatistics Program, Public Health Sciences Division, Fred Hutchinson Cancer Research Center, Seattle, WA
| | - Paul D. Wagner
- Division of Cancer Prevention, National Cancer Institute, Rockville, MD
| | - Amit G. Singal
- Division of Digestive and Liver Diseases, UT Southwestern Medical Center, Dallas TX
| | - Samir M Hanash
- Department of Clinical Cancer Prevention, McCombs Institute for Cancer Detection and Treatment at MD Anderson Cancer Center, Houston, TX
| | - Sudhir Srivastava
- Division of Cancer Prevention, National Cancer Institute, Rockville, MD
| | - Ying Huang
- Biostatstics, Bioinformatics and Epidemiology Program, Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, WA
| | - Ying-Qi Zhao
- Biostatistics Program, Public Health Sciences Division, Fred Hutchinson Cancer Research Center, Seattle, WA
| | - Suresh T Chari
- Department of Gastroenterology, Hepatology & Nutrition, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Guillermo Marquez
- Division of Cancer Prevention, National Cancer Institute, Rockville, MD
| | - Ruth Etizioni
- Biostatistics Program, Public Health Sciences Division, Fred Hutchinson Cancer Research Center, Seattle, WA
| | - Tracey L Marsh
- Biostatistics Program, Public Health Sciences Division, Fred Hutchinson Cancer Research Center, Seattle, WA
| | - Ziding Feng
- Biostatistics Program, Public Health Sciences Division, Fred Hutchinson Cancer Research Center, Seattle, WA
| |
Collapse
|
39
|
Barnhart KT, Bollig KJ, Senapati S, Takacs P, Robins JC, Haisenleder DJ, Beer LA, Savaris RF, Koelper NC, Speicher DW, Chittams J, Bao J, Wen Z, Feng Y, Kim M, Mumford S, Shen L, Gimotty P. Multiplexed serum biomarkers to discriminate nonviable and ectopic pregnancy. Fertil Steril 2024; 122:482-493. [PMID: 38677710 DOI: 10.1016/j.fertnstert.2024.04.028] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2024] [Revised: 04/18/2024] [Accepted: 04/18/2024] [Indexed: 04/29/2024]
Abstract
OBJECTIVE To evaluate combinations of candidate biomarkers to develop a multiplexed prediction model for identifying the viability and location of an early pregnancy. In this study, we assessed 24 biomarkers with multiple machine learning-based methodologies to assess if multiplexed biomarkers may improve the diagnosis of normal and abnormal early pregnancies. DESIGN A nested case-control design evaluated the predictive ability and discrimination of biomarkers in patients at risk of early pregnancy failure in the first trimester to classify viability and location. SETTING Three university hospitals. PATIENTS A total of 218 individuals with pain and/or bleeding in early pregnancy: 75 had an ongoing intrauterine gestation; 68 had ectopic pregnancies (EPs); and 75 had miscarriages. INTERVENTIONS Serum levels of 24 biomarkers were assessed in the same patients. Multiple machine learning-based methodologies to evaluate combinations of these top candidates to develop a multiplexed prediction model for the identification of a nonviable pregnancy (ongoing intrauterine pregnancy vs. miscarriage or EP) and an EP (EP vs. ongoing intrauterine pregnancy or miscarriage). MAIN OUTCOME MEASURES The predicted classification using each model was compared with the actual diagnosis, and sensitivity, specificity, positive predictive value, negative predictive value, conclusive classification, and accuracy were calculated. RESULTS Models using classification regression tree analysis using 3 (pregnancy-specific beta-1-glycoprotein 3 [PSG3], chorionic gonadotropin-alpha subunit, and pregnancy-associated plasma protein-A) biomarkers were able to predict a maximum sensitivity of 93.3% and a maximum specificity of 98.6%. The model with the highest accuracy was 97.4% (with 70.2% receiving classification). Models using an overlapping group of 3 (soluble fms-like tyrosine kinase-1, PSG3, and tissue factor pathway inhibitor 2) biomarkers achieved a maximum sensitivity of 98.5% and a maximum specificity of 95.3%. The model with the highest accuracy was 94.4% (with 65.6% receiving classification). When the models were used simultaneously, the conclusive classification increased to 72.7% with an accuracy of 95.9%. The predictive ability of the biomarkers in the random forest produced similar test characteristics when using 11 predictive biomarkers. CONCLUSION We have demonstrated a pool of biomarkers from divergent biological pathways that can be used to classify individuals with potential early pregnancy loss. The biomarkers choriogonadotropin alpha, pregnancy-associated plasma protein-A, and PSG3 can be used to predict viability, and soluble fms-like tyrosine kinase-1, tissue factor pathway inhibitor 2, and PSG3 can be used to predict pregnancy location.
Collapse
Affiliation(s)
- Kurt T Barnhart
- Department of Obstetrics and Gynecology, University of Pennsylvania, Philadelphia, Pennsylvania.
| | - Kassie J Bollig
- Department of Obstetrics and Gynecology, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Suneeta Senapati
- Department of Obstetrics and Gynecology, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Peter Takacs
- Department of Obstetrics and Gynecology, Eastern Virginia Medical School, Norfolk, Virginia
| | - Jared C Robins
- Department of Obstetrics and Gynecology, Northwestern University, Chicago, Illinois
| | - Daniel J Haisenleder
- Department of Internal Medicine and the Center for Research in Reproduction, University of Virginia, Charlottesville, Virginia
| | - Lynn A Beer
- Center for Systems & Computational Biology, The Wistar Institute, Philadelphia, Pennsylvania
| | - Ricardo F Savaris
- Department of Gynecology and Obstetrics, School of Medicine, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
| | - Nathanael C Koelper
- Department of Obstetrics and Gynecology, University of Pennsylvania, Philadelphia, Pennsylvania
| | - David W Speicher
- Center for Systems & Computational Biology, The Wistar Institute, Philadelphia, Pennsylvania
| | - Jesse Chittams
- Biostatistics Consulting Unit, University of Pennsylvania School of Nursing, Philadelphia, Pennsylvania
| | - Jingxuan Bao
- Department of Biostatistics, Epidemiology and Informatics, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania
| | - Zixuan Wen
- Department of Biostatistics, Epidemiology and Informatics, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania
| | - Yanbo Feng
- Department of Biostatistics, Epidemiology and Informatics, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania
| | - Mansu Kim
- Department of Biostatistics, Epidemiology and Informatics, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania
| | - Sunni Mumford
- Department of Obstetrics and Gynecology, University of Pennsylvania, Philadelphia, Pennsylvania; Department of Biostatistics, Epidemiology and Informatics, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania
| | - Li Shen
- Department of Biostatistics, Epidemiology and Informatics, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania
| | - Phyllis Gimotty
- Department of Biostatistics, Epidemiology and Informatics, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania
| |
Collapse
|
40
|
Wang B, Zhang L, Liang G, Meng L, Xu Y, Li H, Song Z, Zhang X, Li Z, Guo C, Guan T, He Y. Realization of high-performance biosensor through sandwich analysis utilizing weak value amplification. Talanta 2024; 277:126302. [PMID: 38830277 DOI: 10.1016/j.talanta.2024.126302] [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: 02/15/2024] [Revised: 05/07/2024] [Accepted: 05/20/2024] [Indexed: 06/05/2024]
Abstract
A label-free optical sandwich immunoassay sensor, utilizing weak value amplification and total internal reflection, was devised for real-time, high-sensitivity analysis and detection of low-concentration targets. 3D printed channels and sodium chloride solution were employed to ensure reproducibility, reliability, and stability of the measurements for calibration. The sandwich structure demonstrated enhanced responsiveness in the proposed optical biosensor through a comparative analysis of the direct assay and sandwich assay for detecting alpha-fetoprotein (AFP) at the same concentration. By optimizing the binding sequences of the coating antibody, target, and detection antibody in the sandwich method, a more suitable sandwich sensing approach based on weak value amplification was achieved. With this approach, the limit of detection (LOD) of 6.29 ng/mL (pM level) for AFP in PBS solution was achieved. AFP testing and regeneration experiments in human serum have proved the feasibility of our methods in detecting complex samples and the reusability of sensing chips. Additionally, the method demonstrated excellent selectivity for unpaired antigens. The efficacy of this methodology was evaluated by simultaneously detecting AFP, carcinoembryonic antigen (CEA), and CA15-3 on a singular sensor chip. In conclusion, the label-free sandwich immunoassay sensing scheme holds promise for advancing the proposed optical sensors based on weak value amplification in early diagnosis and prevention applications. Compared to other biomarker detection methods, it will be easier to promote in practical applications.
Collapse
Affiliation(s)
- Bei Wang
- Institute of Optical Imaging and Sensing, Shenzhen Key Laboratory for Minimal Invasive Medical Technologies, Tsinghua Shenzhen International Graduate School, Tsinghua University, Shenzhen, 518055, China; Institute of Biopharmaceutical and Health Engineering, Tsinghua Shenzhen International Graduate School, Tsinghua University, Shenzhen, 518055, China
| | - Lizhong Zhang
- Institute of Optical Imaging and Sensing, Shenzhen Key Laboratory for Minimal Invasive Medical Technologies, Tsinghua Shenzhen International Graduate School, Tsinghua University, Shenzhen, 518055, China; Institute of Biopharmaceutical and Health Engineering, Tsinghua Shenzhen International Graduate School, Tsinghua University, Shenzhen, 518055, China
| | - Gengyu Liang
- Institute of Optical Imaging and Sensing, Shenzhen Key Laboratory for Minimal Invasive Medical Technologies, Tsinghua Shenzhen International Graduate School, Tsinghua University, Shenzhen, 518055, China; Institute of Biopharmaceutical and Health Engineering, Tsinghua Shenzhen International Graduate School, Tsinghua University, Shenzhen, 518055, China
| | - Lingqin Meng
- Institute of Optical Imaging and Sensing, Shenzhen Key Laboratory for Minimal Invasive Medical Technologies, Tsinghua Shenzhen International Graduate School, Tsinghua University, Shenzhen, 518055, China; Institute of Biopharmaceutical and Health Engineering, Tsinghua Shenzhen International Graduate School, Tsinghua University, Shenzhen, 518055, China
| | - Yang Xu
- Institute of Optical Imaging and Sensing, Shenzhen Key Laboratory for Minimal Invasive Medical Technologies, Tsinghua Shenzhen International Graduate School, Tsinghua University, Shenzhen, 518055, China; Institute of Biopharmaceutical and Health Engineering, Tsinghua Shenzhen International Graduate School, Tsinghua University, Shenzhen, 518055, China
| | - Han Li
- Institute of Optical Imaging and Sensing, Shenzhen Key Laboratory for Minimal Invasive Medical Technologies, Tsinghua Shenzhen International Graduate School, Tsinghua University, Shenzhen, 518055, China; Institute of Biopharmaceutical and Health Engineering, Tsinghua Shenzhen International Graduate School, Tsinghua University, Shenzhen, 518055, China
| | - Zishuo Song
- Institute of Optical Imaging and Sensing, Shenzhen Key Laboratory for Minimal Invasive Medical Technologies, Tsinghua Shenzhen International Graduate School, Tsinghua University, Shenzhen, 518055, China; Institute of Biopharmaceutical and Health Engineering, Tsinghua Shenzhen International Graduate School, Tsinghua University, Shenzhen, 518055, China
| | - Xiaonan Zhang
- Institute of Optical Imaging and Sensing, Shenzhen Key Laboratory for Minimal Invasive Medical Technologies, Tsinghua Shenzhen International Graduate School, Tsinghua University, Shenzhen, 518055, China; Institute of Biopharmaceutical and Health Engineering, Tsinghua Shenzhen International Graduate School, Tsinghua University, Shenzhen, 518055, China
| | - Zhangyan Li
- College of Physics and Information Engineering, Zhaotong University, Zhaotong, 657000, China
| | - Cuixia Guo
- School of Mechanical Engineering and Automation, Fuzhou University, Fuzhou, 350108, China.
| | - Tian Guan
- Institute of Biopharmaceutical and Health Engineering, Tsinghua Shenzhen International Graduate School, Tsinghua University, Shenzhen, 518055, China
| | - Yonghong He
- Institute of Optical Imaging and Sensing, Shenzhen Key Laboratory for Minimal Invasive Medical Technologies, Tsinghua Shenzhen International Graduate School, Tsinghua University, Shenzhen, 518055, China; Institute of Biopharmaceutical and Health Engineering, Tsinghua Shenzhen International Graduate School, Tsinghua University, Shenzhen, 518055, China.
| |
Collapse
|
41
|
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
|
42
|
Jiang X, Li W, Wang K, Li R, Ning J. Analyzing heterogeneity in biomarker discriminative performance through partial time-dependent receiver operating characteristic curve modeling. Stat Methods Med Res 2024; 33:1424-1436. [PMID: 39053568 PMCID: PMC11449645 DOI: 10.1177/09622802241262521] [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] [Indexed: 07/27/2024]
Abstract
This study investigates the heterogeneity of a biomarker's discriminative performance for predicting subsequent time-to-event outcomes across different patient subgroups. While the area under the curve (AUC) for the time-dependent receiver operating characteristic curve is commonly used to assess biomarker performance, the partial time-dependent AUC (PAUC) provides insights that are often more pertinent for population screening and diagnostic testing. To achieve this objective, we propose a regression model tailored for PAUC and develop two distinct estimation procedures for discrete and continuous covariates, employing a pseudo-partial likelihood method. Simulation studies are conducted to assess the performance of these procedures across various scenarios. We apply our model and inference procedure to the Alzheimer's Disease Neuroimaging Initiative data set to evaluate potential heterogeneities in the discriminative performance of biomarkers for early Alzheimer's disease diagnosis based on patients' characteristics.
Collapse
Affiliation(s)
- Xinyang Jiang
- Department of Biostatistics and Data Science, The University of Texas Health Science Center at Houston, Houston, USA
| | - Wen Li
- Department of Internal Medicine, The University of Texas Health Science Center at Houston McGovern Medical School, Houston, USA
| | - Kang Wang
- Department of Biostatistics, The University of Texas MD Anderson Cancer Center, Houston, USA
| | - Ruosha Li
- Department of Biostatistics and Data Science, The University of Texas Health Science Center at Houston, Houston, USA
| | - Jing Ning
- Department of Biostatistics, The University of Texas MD Anderson Cancer Center, Houston, USA
| | | |
Collapse
|
43
|
Zamuner FT, Ramos-López A, García-Negrón A, Purcell-Wiltz A, Cortés-Ortiz A, Cuevas AR, Gosala K, Winkler E, Sidransky D, Guerrero-Preston R. Evaluation of silica spin‑column and magnetic bead formats for rapid DNA methylation analysis in clinical and point‑of‑care settings. Biomed Rep 2024; 21:112. [PMID: 38912171 PMCID: PMC11190640 DOI: 10.3892/br.2024.1800] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2024] [Accepted: 05/20/2024] [Indexed: 06/25/2024] Open
Abstract
Late-stage cancers lack effective treatment, underscoring the need for early diagnosis to improve prognosis and decrease mortality rates. Molecular markers, such as DNA methylation, offer promise in early cancer detection. The present study compared commercial kits for analyzing DNA from cervical liquid cytology samples in cancer screening. Rapid bisulfite conversion kits using silica spin-columns and magnetic beads were assessed against standard DNA extraction and bisulfite conversion methods for profiling DNA methylation using quantitative methylation-specific PCR. β-actin amplification indicated the suitability of small sample volumes for methylation studies using either the pellet or supernatant (cell-free DNA) parts. Comparison of Bisulfite Conversion Kit-Whole Cell (Abcam), Methylamp Bisulfite Modification (Epigentek), EpiTect Fast LyseAll Bisulfite Kit (Qiagen GmbH) and EZ DNA Methylation-Direct Kit (Zymo Research Corp.) showed no significant differences in β-actin cycle threshold values. EZ-96 DNA Methylation-Lightning MagPrep (Zymo Research Corp.), a hybrid kit in a 96-well plate format, exhibited swift turnaround time and similar amplification efficiency. Automation with magnetic bead kits increased throughput without compromising amplification efficiency in open PCR systems. Cost analysis favored direct kits over the gold standard manual protocol. This comparison aids in selecting cost-effective DNA methylation diagnostic tests. The present study confirmed comparable kit performance in methylation-based analysis, highlighting the adequacy of cytology samples and the potential of bodily fluids as alternatives for liquid biopsy.
Collapse
Affiliation(s)
- Fernando T. Zamuner
- Department of Otolaryngology and Head and Neck Surgery, Head and Neck Cancer Research Laboratory, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
- Department of Oncology, The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA
| | - Ashley Ramos-López
- LifeGene-Biomarks, Research and Development Unit, Toa Baja 00949, Puerto Rico
| | | | - Ana Purcell-Wiltz
- LifeGene-Biomarks, Research and Development Unit, Toa Baja 00949, Puerto Rico
- Department of Medicine, San Juan Bautista School of Medicine, Caguas 00725, Puerto Rico
| | - Andrea Cortés-Ortiz
- LifeGene-Biomarks, Research and Development Unit, Toa Baja 00949, Puerto Rico
- Department of Medicine, San Juan Bautista School of Medicine, Caguas 00725, Puerto Rico
| | - Aniris Román Cuevas
- LifeGene-Biomarks, Research and Development Unit, Toa Baja 00949, Puerto Rico
- Department of Biology, University of Puerto Rico, Río Piedras 00931, Puerto Rico
| | - Keerthana Gosala
- Department of Otolaryngology and Head and Neck Surgery, Head and Neck Cancer Research Laboratory, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Eli Winkler
- Department of Otolaryngology and Head and Neck Surgery, Head and Neck Cancer Research Laboratory, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
- New York University Langone Health, New York, NY 10016, USA
| | - David Sidransky
- Department of Otolaryngology and Head and Neck Surgery, Head and Neck Cancer Research Laboratory, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
- Department of Oncology, The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA
| | - Rafael Guerrero-Preston
- LifeGene-Biomarks, Research and Development Unit, Toa Baja 00949, Puerto Rico
- LifeGene-Biomarks, FastForward Innovation Hub, Baltimore, MD 21205, USA
| |
Collapse
|
44
|
Kumar SH. Alternative endpoints to mortality in cancer screening trials. Mol Oncol 2024; 18:1817-1820. [PMID: 38970563 PMCID: PMC11306507 DOI: 10.1002/1878-0261.13697] [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: 05/28/2024] [Accepted: 06/25/2024] [Indexed: 07/08/2024] Open
Abstract
The reliance on mortality endpoints in cancer screening trials is not always compatible with the need to accelerate progress in outcomes for patient and public benefit. Evaluation of novel cancer screening technologies, such as multi-cancer early detection (MCED) tests, could be expedited by using alternative metrics that are measurable earlier than mortality. These include endpoints based on cancer stage at diagnosis, such as reduction in late-stage cancer incidence, and endpoints following cancer diagnosis, such as eligibility for curative therapy. Innovative trial designs with earlier measures that complement cancer mortality are needed to realise the potential benefits of novel screening technologies such as MCEDs more rapidly.
Collapse
|
45
|
Kohaar I, Hodges NA, Srivastava S. Biomarkers in Cancer Screening: Promises and Challenges in Cancer Early Detection. Hematol Oncol Clin North Am 2024; 38:869-888. [PMID: 38782647 PMCID: PMC11222039 DOI: 10.1016/j.hoc.2024.04.004] [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] [Indexed: 05/25/2024]
Abstract
Cancer continues to be one the leading causes of death worldwide, primarily due to the late detection of the disease. Cancers detected at early stages may enable more effective intervention of the disease. However, most cancers lack well-established screening procedures except for cancers with an established early asymptomatic phase and clinically validated screening tests. There is a critical need to identify and develop assays/tools in conjunction with imaging approaches for precise screening and detection of the aggressive disease at an early stage. New developments in molecular cancer screening and early detection include germline testing, synthetic biomarkers, and liquid biopsy approaches.
Collapse
Affiliation(s)
- Indu Kohaar
- Cancer Biomarkers Research Group, Division of Cancer Prevention, National Cancer Institute, NIH, 9609 Medical Center Drive, NCI Shady Grove Building, Rockville, MD 20850, USA
| | - Nicholas A Hodges
- Cancer Biomarkers Research Group, Division of Cancer Prevention, National Cancer Institute, NIH, 9609 Medical Center Drive, NCI Shady Grove Building, Rockville, MD 20850, USA
| | - Sudhir Srivastava
- Cancer Biomarkers Research Group, Division of Cancer Prevention, National Cancer Institute, NIH, 9609 Medical Center Drive, NCI Shady Grove Building, Rockville, MD 20850, USA.
| |
Collapse
|
46
|
Goggins M. The role of biomarkers in the early detection of pancreatic cancer. Fam Cancer 2024; 23:309-322. [PMID: 38662265 PMCID: PMC11309746 DOI: 10.1007/s10689-024-00381-4] [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: 03/09/2024] [Accepted: 03/19/2024] [Indexed: 04/26/2024]
Abstract
Pancreatic surveillance can detect early-stage pancreatic cancer and achieve long-term survival, but currently involves annual endoscopic ultrasound and MRI/MRCP, and is recommended only for individuals who meet familial/genetic risk criteria. To improve upon current approaches to pancreatic cancer early detection and to expand access, more accurate, inexpensive, and safe biomarkers are needed, but finding them has remained elusive. Newer approaches to early detection, such as using gene tests to personalize biomarker interpretation, and the increasing application of artificial intelligence approaches to integrate complex biomarker data, offer promise that clinically useful biomarkers for early pancreatic cancer detection are on the horizon.
Collapse
Affiliation(s)
- Michael Goggins
- Department of Pathology, The Sol Goldman Pancreatic Cancer Research Center, The Johns Hopkins University School of Medicine, 1550 Orleans Street, Baltimore, MD, 21231, USA.
- Department of Medicine, The Sol Goldman Pancreatic Cancer Research Center, The Johns Hopkins University School of Medicine, Baltimore, MD, USA.
- Department of Oncology, The Sol Goldman Pancreatic Cancer Research Center, The Johns Hopkins University School of Medicine, Baltimore, MD, USA.
| |
Collapse
|
47
|
Ibnidris A, Liaskos N, Eldem E, Gunn A, Streffer J, Gold M, Rea M, Teipel S, Gardiol A, Boccardi M. Facilitating the use of the target product profile in academic research: a systematic review. J Transl Med 2024; 22:693. [PMID: 39075460 PMCID: PMC11288132 DOI: 10.1186/s12967-024-05476-1] [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] [Accepted: 07/03/2024] [Indexed: 07/31/2024] Open
Abstract
BACKGROUND The Target Product Profile (TPP) is a tool used in industry to guide development strategies by addressing user needs and fostering effective communication among stakeholders. However, they are not frequently used in academic research, where they may be equally useful. This systematic review aims to extract the features of accessible TPPs, to identify commonalities and facilitate their integration in academic research methodology. METHODS We searched peer-reviewed papers published in English developing TPPs for different products and health conditions in four biomedical databases. Interrater agreement, computed on random abstract and paper sets (Cohen's Kappa; percentage agreement with zero tolerance) was > 0.91. We interviewed experts from industry contexts to gain insight on the process of TPP development, and extracted general and specific features on TPP use and structure. RESULTS 138 papers were eligible for data extraction. Of them, 92% (n = 128) developed a new TPP, with 41.3% (n = 57) focusing on therapeutics. The addressed disease categories were diverse; the largest (47.1%, n = 65) was infectious diseases. Only one TPP was identified for several fields, including global priorities like dementia. Our analyses found that 56.5% of papers (n = 78) was authored by academics, and 57.8% of TPPs (n = 80) featured one threshold level of product performance. The number of TPP features varied widely across and within product types (n = 3-44). Common features included purpose/context of use, shelf life for drug stability and validation aspects. Most papers did not describe the methods used to develop the TPP. We identified aspects to be taken into account to build and report TPPs, as a starting point for more focused initiatives guiding use by academics. DISCUSSION TPPs are used in academic research mostly for infectious diseases and have heterogeneous features. Our extraction of key features and common structures helps to understand the tool and widen its use in academia. This is of particular relevance for areas of notable unmet needs, like dementia. Collaboration between stakeholders is key for innovation. Tools to streamline communication such as TPPs would support the development of products and services in academia as well as industry.
Collapse
Affiliation(s)
- Aliaa Ibnidris
- German Center for Neurodegenerative Diseases (DZNE), Rostock-Greifswald site, Gehlsheimer Str. 20, 18147, Rostock, Germany
- Neuroscience Institute, Department of Psychiatry and Mental Health, University of Cape Town, Cape Town, South Africa
| | - Nektarios Liaskos
- German Center for Neurodegenerative Diseases (DZNE), Rostock-Greifswald site, Gehlsheimer Str. 20, 18147, Rostock, Germany
- European Infrastructure for Translational Medicine (EATRIS), Amsterdam, The Netherlands
| | - Ece Eldem
- German Center for Neurodegenerative Diseases (DZNE), Rostock-Greifswald site, Gehlsheimer Str. 20, 18147, Rostock, Germany
| | | | - Johannes Streffer
- Reference Center for Biological Markers of Dementia (BIODEM), Department of Biomedical Sciences, University of Antwerp, Antwerp, Belgium
| | - Michael Gold
- AriLex Life Sciences LLC, 780 Elysian Way, Deerfield, IL, 60015, USA
| | | | - Stefan Teipel
- German Center for Neurodegenerative Diseases (DZNE), Rostock-Greifswald site, Gehlsheimer Str. 20, 18147, Rostock, Germany
- Department of Psychosomatic Medicine and Psychotherapy, University of Medicine Rostock, Rostock, Germany
| | - Alejandra Gardiol
- European Infrastructure for Translational Medicine (EATRIS), Amsterdam, The Netherlands
- Queen Mary University of London, London, UK
| | - Marina Boccardi
- German Center for Neurodegenerative Diseases (DZNE), Rostock-Greifswald site, Gehlsheimer Str. 20, 18147, Rostock, Germany.
- Department of Psychosomatic Medicine and Psychotherapy, University of Medicine Rostock, Rostock, Germany.
| |
Collapse
|
48
|
Rubinstein WS, Patriotis C, Dickherber A, Han PKJ, Katki HA, LeeVan E, Pinsky PF, Prorok PC, Skarlupka AL, Temkin SM, Castle PE, Minasian LM. Cancer screening with multicancer detection tests: A translational science review. CA Cancer J Clin 2024; 74:368-382. [PMID: 38517462 PMCID: PMC11226362 DOI: 10.3322/caac.21833] [Citation(s) in RCA: 17] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/21/2023] [Revised: 02/06/2024] [Accepted: 02/12/2024] [Indexed: 03/23/2024] Open
Abstract
Multicancer detection (MCD) tests use a single, easily obtainable biospecimen, such as blood, to screen for more than one cancer concurrently. MCD tests can potentially be used to improve early cancer detection, including cancers that currently lack effective screening methods. However, these tests have unknown and unquantified benefits and harms. MCD tests differ from conventional cancer screening tests in that the organ responsible for a positive test is unknown, and a broad diagnostic workup may be necessary to confirm the location and type of underlying cancer. Among two prospective studies involving greater than 16,000 individuals, MCD tests identified those who had some cancers without currently recommended screening tests, including pancreas, ovary, liver, uterus, small intestine, oropharyngeal, bone, thyroid, and hematologic malignancies, at early stages. Reported MCD test sensitivities range from 27% to 95% but differ by organ and are lower for early stage cancers, for which treatment toxicity would be lowest and the potential for cure might be highest. False reassurance from a negative MCD result may reduce screening adherence, risking a loss in proven public health benefits from standard-of-care screening. Prospective clinical trials are needed to address uncertainties about MCD accuracy to detect different cancers in asymptomatic individuals, whether these tests can detect cancer sufficiently early for effective treatment and mortality reduction, the degree to which these tests may contribute to cancer overdiagnosis and overtreatment, whether MCD tests work equally well across all populations, and the appropriate diagnostic evaluation and follow-up for patients with a positive test.
Collapse
Affiliation(s)
- Wendy S. Rubinstein
- Division of Cancer Prevention, US National Cancer Institute, Rockville, Maryland, USA
| | - Christos Patriotis
- Division of Cancer Prevention, US National Cancer Institute, Rockville, Maryland, USA
| | - Anthony Dickherber
- Center for Strategic Scientific Initiatives, US National Cancer Institute, Rockville, Maryland, USA
| | - Paul K. J. Han
- Division of Cancer Control and Population Sciences, US National Cancer Institute, Rockville, Maryland, USA
| | - Hormuzd A. Katki
- Division of Cancer Epidemiology and Genetics, US National Cancer Institute, Rockville, Maryland, USA
| | - Elyse LeeVan
- Division of Cancer Prevention, US National Cancer Institute, Rockville, Maryland, USA
| | - Paul F. Pinsky
- Division of Cancer Prevention, US National Cancer Institute, Rockville, Maryland, USA
| | - Philip C. Prorok
- Division of Cancer Prevention, US National Cancer Institute, Rockville, Maryland, USA
| | - Amanda L. Skarlupka
- Division of Cancer Prevention, US National Cancer Institute, Rockville, Maryland, USA
| | - Sarah M. Temkin
- National Institutes of Health Office of Research on Women’s Health, Bethesda, Maryland, USA
| | - Philip E. Castle
- Division of Cancer Prevention, US National Cancer Institute, Rockville, Maryland, USA
- Division of Cancer Epidemiology and Genetics, US National Cancer Institute, Rockville, Maryland, USA
| | - Lori M. Minasian
- Division of Cancer Prevention, US National Cancer Institute, Rockville, Maryland, USA
| |
Collapse
|
49
|
Zhu K, Zhao YQ, Zheng Y. Designing cancer screening trials for reduction in late-stage cancer incidence. Biometrics 2024; 80:ujae097. [PMID: 39302139 DOI: 10.1093/biomtc/ujae097] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2023] [Revised: 08/08/2024] [Accepted: 09/03/2024] [Indexed: 09/22/2024]
Abstract
Before implementing a biomarker test for early cancer detection into routine clinical care, the test must demonstrate clinical utility, that is, the test results should lead to clinical actions that positively affect patient-relevant outcomes. Unlike therapeutical trials for patients diagnosed with cancer, designing a randomized controlled trial (RCT) to demonstrate the clinical utility of an early detection biomarker with mortality and related endpoints poses unique challenges. The hurdles stem from the prolonged natural progression of the disease and the lack of information regarding the time-varying screening effect on the target asymptomatic population. To facilitate the study design of screening trials, we propose using a generic multistate disease history model and derive model-based effect sizes. The model links key performance metrics of the test, such as sensitivity, to primary endpoints like the incidence of late-stage cancer. It also incorporates the practical implementation of the biomarker-testing program in real-world scenarios. Based on the chronological time scale aligned with RCT, our method allows the assessment of study powers based on key features of the new program, including the test sensitivity, the length of follow-up, and the number and frequency of repeated tests. The calculation tool from the proposed method will enable practitioners to perform realistic and quick evaluations when strategizing screening trials for specific diseases. We use numerical examples based on the National Lung Screening Trial to demonstrate the method.
Collapse
Affiliation(s)
- Kehao Zhu
- Department of Biostatistics, University of Washington, Seattle, WA 98109, USA
| | - Ying-Qi Zhao
- Public Health Sciences Division, Fred Hutchinson Cancer Center, Seattle, WA 98109, USA
| | - Yingye Zheng
- Public Health Sciences Division, Fred Hutchinson Cancer Center, Seattle, WA 98109, USA
| |
Collapse
|
50
|
Wan F, Zhu Y, Wu F, Huang X, Chen Y, Zhou Y, Li H, Liang L, Qin L, Wang Q, He M. Retinol-binding protein 4 as a promising serum biomarker for the diagnosis and prognosis of hepatocellular Carcinoma. Transl Oncol 2024; 45:101979. [PMID: 38728873 PMCID: PMC11107351 DOI: 10.1016/j.tranon.2024.101979] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2024] [Revised: 03/20/2024] [Accepted: 04/26/2024] [Indexed: 05/12/2024] Open
Abstract
BACKGROUND The prognosis of hepatocellular carcinoma (HCC) is universally poor. Early diagnosis plays a pivotal role in determining the outcome of HCC. METHODS We employed a comparative proteomics approach to identify potential biomarkers and validated the application of retinol-binding protein 4 (RBP4) as a biomarker for HCC. RBP4 protein expression was examined in liver tissues from 80 HCC patients through immunohistochemical analysis. Serum RBP4 concentrations were measured by ELISA in a cohort comprising 290 HCC patients, matched 202 chronic hepatitis B patients and 269 healthy controls. Survival data were collected from HCC patients. The diagnostic and prognostic values of RBP4 were evaluated using receiver operating curve (ROC) analysis. RESULTS The validation results demonstrated a significant reduction in RBP4 levels in both liver tissues and serum samples from HCC patients. ROC analysis of the diagnostic value of RBP4 revealed an AUC of 0.879 (95 % CI: 0.854∼0.903) for HCC. When combined with AFP, the AUC increased to 0.919, with a sensitivity of 87.9 % and specificity of 80 %. Survival analysis revealed significantly reduced overall survival time in individuals with low-expression of RBP4 compared to those with high-expression. The joint prognostic model exhibited an AUC of 0.926 (95 % CI: 0.888∼0.964), which was significantly higher than that of AFP alone (AUC=0.809; P <0.0001). CONCLUSIONS RBP4 shows a great potential as a biomarker with appreciable diagnostic value, complementing the AFP in HCC diagnosis. Additionally, it holds promise as a prognostic biomarker that, when integrated into a combined prognostic model, could greatly improve HCC prognosis efficiency.
Collapse
Affiliation(s)
- Fengjie Wan
- Guangxi Medical University School of Public Health, Nanning, Guangxi 530021, PR China; Guangxi Colleges and Universities Key Laboratory of Prevention and Control of Highly Prevalent Diseases, Guangxi Medical University, Nanning, Guangxi 530021, PR China
| | - Yujia Zhu
- Guigang Dermatosis Prevention and Treatment Hospital, Guigang, Guangxi 537100, PR China
| | - Feixiang Wu
- Guangxi Medical University Cancer Hospital, Nanning, Guangxi 530021, PR China
| | - Xuejing Huang
- Animal Center of Guangxi Medical University, Nanning, Guangxi 530021, PR China
| | - Ying Chen
- Department of Environmental Health Sciences, Yale School of Public Health, Yale University, New Haven, CT 06520, USA
| | - Yi Zhou
- Guangxi Medical University Life Sciences Institute, Nanning, Guangxi 530021, PR China
| | - Hongtao Li
- Guilin Medical University, Guilin, Guangxi 541001, PR China
| | - Lifang Liang
- Guangxi Medical University School of Public Health, Nanning, Guangxi 530021, PR China
| | - Lirong Qin
- The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi 530021, PR China
| | - Qi Wang
- Guangxi Medical University Cancer Hospital, Nanning, Guangxi 530021, PR China.
| | - Min He
- Guangxi Medical University School of Public Health, Nanning, Guangxi 530021, PR China; Animal Center of Guangxi Medical University, Nanning, Guangxi 530021, PR China; Key Laboratory of High-Incidence-Tumor Prevention & Treatment, Guangxi Medical University, Ministry of Education, Nanning 530021, PR China.
| |
Collapse
|