51
|
Marti-Aguado D, Arnouk J, Liang JX, Lara-Romero C, Behari J, Furlan A, Jimenez-Pastor A, Ten-Esteve A, Alfaro-Cervello C, Bauza M, Gallen-Peris A, Gimeno-Torres M, Merino-Murgui V, Perez-Girbes A, Benlloch S, Pérez-Rojas J, Puglia V, Ferrández-Izquierdo A, Aguilera V, Giesteira B, França M, Monton C, Escudero-García D, Alberich-Bayarri Á, Serra MA, Bataller R, Romero-Gomez M, Marti-Bonmati L. Development and validation of an image biomarker to identify metabolic dysfunction associated steatohepatitis: MR-MASH score. Liver Int 2024; 44:202-213. [PMID: 37904633 DOI: 10.1111/liv.15766] [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: 07/07/2023] [Revised: 09/11/2023] [Accepted: 10/08/2023] [Indexed: 11/01/2023]
Abstract
BACKGROUND AND AIMS Diagnosis of metabolic dysfunction-associated steatohepatitis (MASH) requires histology. In this study, a magnetic resonance imaging (MRI) score was developed and validated to identify MASH in patients with metabolic dysfunction-associated steatotic liver disease (MASLD). Secondarily, a screening strategy for MASH diagnosis was investigated. METHODS This prospective multicentre study included 317 patients with biopsy-proven MASLD and contemporaneous MRI. The discovery cohort (Spain, Portugal) included 194 patients. NAFLD activity score (NAS) and fibrosis were assessed with the NASH-CRN histologic system. MASH was defined by the presence of steatosis, lobular inflammation, and ballooning, with NAS ≥4 with or without fibrosis. An MRI-based composite biomarker of Proton Density Fat Fraction and waist circumference (MR-MASH score) was developed. Findings were afterwards validated in an independent cohort (United States, Spain) with different MRI protocols. RESULTS In the derivation cohort, 51% (n = 99) had MASH. The MR-MASH score identified MASH with an AUC = .88 (95% CI .83-.93) and strongly correlated with NAS (r = .69). The MRI score lower cut-off corresponded to 88% sensitivity with 86% NPV, while the upper cut-off corresponded to 92% specificity with 87% PPV. MR-MASH was validated with an AUC = .86 (95% CI .77-.92), 91% sensitivity (lower cut-off) and 87% specificity (upper cut-off). A two-step screening strategy with sequential MR-MASH examination performed in patients with indeterminate-high FIB-4 or transient elastography showed an 83-84% PPV to identify MASH. The AUC of MR-MASH was significantly higher than that of the FAST score (p < .001). CONCLUSIONS The MR-MASH score has clinical utility in the identification and management of patients with MASH at risk of progression.
Collapse
Affiliation(s)
- David Marti-Aguado
- Digestive Disease Department, Clinic University Hospital, INCLIVA Health Research Institute, Valencia, Spain
- Biomedical Imaging Research Group (GIBI230), La Fe Health Research Institute, and Imaging La Fe node at Distributed Network for Biomedical Imaging (ReDIB) Unique Scientific and Technical Infrastructures (ICTS), Valencia, Spain
| | - Joud Arnouk
- Division of Gastroenterology, Hepatology and Nutrition, Center for Liver Diseases, Pittsburgh Liver Research Center, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania, USA
| | - Jia-Xu Liang
- Digestive Diseases Department, CIBERehd, Virgen del Rocio University Hospital, Seville, Spain
- Institute of Biomedicine of Seville (HUVR/CSIC/US), Department of Medicine, University of Seville, Seville, Spain
| | - Carmen Lara-Romero
- Digestive Diseases Department, CIBERehd, Virgen del Rocio University Hospital, Seville, Spain
- Institute of Biomedicine of Seville (HUVR/CSIC/US), Department of Medicine, University of Seville, Seville, Spain
| | - Jaideep Behari
- Division of Gastroenterology, Hepatology and Nutrition, Center for Liver Diseases, Pittsburgh Liver Research Center, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania, USA
| | - Alessandro Furlan
- Division of Abdominal Imaging, Department of Radiology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Ana Jimenez-Pastor
- Biomedical Imaging Research Group (GIBI230), La Fe Health Research Institute, and Imaging La Fe node at Distributed Network for Biomedical Imaging (ReDIB) Unique Scientific and Technical Infrastructures (ICTS), Valencia, Spain
- Quantitative Imaging Biomarkers in Medicine, QUIBIM SL, Valencia, Spain
| | - Amadeo Ten-Esteve
- Biomedical Imaging Research Group (GIBI230), La Fe Health Research Institute, and Imaging La Fe node at Distributed Network for Biomedical Imaging (ReDIB) Unique Scientific and Technical Infrastructures (ICTS), Valencia, Spain
| | - Clara Alfaro-Cervello
- Pathology Department, Clinic University Hospital, INCLIVA Health Research Institute, Valencia, Spain
- Faculty of Medicine, University of Valencia, Valencia, Spain
| | - Mónica Bauza
- Pathology Department, La Fe University and Polytechnic Hospital, Valencia, Spain
| | - Ana Gallen-Peris
- Digestive Disease Department, Hospital Arnau de Vilanova, Valencia, Spain
| | - Marta Gimeno-Torres
- Digestive Disease Department, La Fe University and Polytechnic Hospital, Valencia, Spain
| | - Víctor Merino-Murgui
- Digestive Disease Department, Clinic University Hospital, INCLIVA Health Research Institute, Valencia, Spain
| | - Alexandre Perez-Girbes
- Biomedical Imaging Research Group (GIBI230), La Fe Health Research Institute, and Imaging La Fe node at Distributed Network for Biomedical Imaging (ReDIB) Unique Scientific and Technical Infrastructures (ICTS), Valencia, Spain
- Radiology Department, La Fe University and Polytechnic Hospital, Valencia, Spain
| | - Salvador Benlloch
- Digestive Disease Department, Hospital Arnau de Vilanova, Valencia, Spain
- CIBERehd, Centro de Investigación Biomédica en Red en Enfermedades Hepáticas y Digestivas, Instituto de Salud Carlos III, Madrid, Spain
| | - Judith Pérez-Rojas
- Pathology Department, La Fe University and Polytechnic Hospital, Valencia, Spain
| | - Víctor Puglia
- Pathology Department, Hospital Arnau de Vilanova, Valencia, Spain
| | - Antonio Ferrández-Izquierdo
- Pathology Department, Clinic University Hospital, INCLIVA Health Research Institute, Valencia, Spain
- Faculty of Medicine, University of Valencia, Valencia, Spain
| | - Victoria Aguilera
- CIBERehd, Centro de Investigación Biomédica en Red en Enfermedades Hepáticas y Digestivas, Instituto de Salud Carlos III, Madrid, Spain
- Hepatology and Liver Transplantation Unit, La Fe University and Polytechnic Hospital, Valencia, Spain
| | - Bruno Giesteira
- Radiology Department, Centro Hospitalar Universitário do Porto, Instituto de Ciências Biomédicas de Abel Salazar, University of Porto, Porto, Portugal
| | - Manuela França
- Radiology Department, Centro Hospitalar Universitário do Porto, Instituto de Ciências Biomédicas de Abel Salazar, University of Porto, Porto, Portugal
| | - Cristina Monton
- Digestive Disease Department, Clinic University Hospital, INCLIVA Health Research Institute, Valencia, Spain
| | - Desamparados Escudero-García
- Digestive Disease Department, Clinic University Hospital, INCLIVA Health Research Institute, Valencia, Spain
- Faculty of Medicine, University of Valencia, Valencia, Spain
| | - Ángel Alberich-Bayarri
- Biomedical Imaging Research Group (GIBI230), La Fe Health Research Institute, and Imaging La Fe node at Distributed Network for Biomedical Imaging (ReDIB) Unique Scientific and Technical Infrastructures (ICTS), Valencia, Spain
- Quantitative Imaging Biomarkers in Medicine, QUIBIM SL, Valencia, Spain
| | - Miguel A Serra
- Digestive Disease Department, Clinic University Hospital, INCLIVA Health Research Institute, Valencia, Spain
- Faculty of Medicine, University of Valencia, Valencia, Spain
| | - Ramon Bataller
- Division of Gastroenterology, Hepatology and Nutrition, Center for Liver Diseases, Pittsburgh Liver Research Center, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania, USA
| | - Manuel Romero-Gomez
- Digestive Diseases Department, CIBERehd, Virgen del Rocio University Hospital, Seville, Spain
- Institute of Biomedicine of Seville (HUVR/CSIC/US), Department of Medicine, University of Seville, Seville, Spain
- University of Seville, Seville, Spain
| | - Luis Marti-Bonmati
- Biomedical Imaging Research Group (GIBI230), La Fe Health Research Institute, and Imaging La Fe node at Distributed Network for Biomedical Imaging (ReDIB) Unique Scientific and Technical Infrastructures (ICTS), Valencia, Spain
- Radiology Department, La Fe University and Polytechnic Hospital, Valencia, Spain
| |
Collapse
|
52
|
Noureddin M, Truong E, Mayo R, Martínez-Arranz I, Mincholé I, Banales JM, Arrese M, Cusi K, Arias-Loste MT, Bruha R, Romero-Gómez M, Iruzubieta P, Aller R, Ampuero J, Calleja JL, Ibañez-Samaniego L, Aspichueta P, Martín-Duce A, Kushner T, Ortiz P, Harrison SA, Anstee QM, Crespo J, Mato JM, Sanyal AJ. Serum identification of at-risk MASH: The metabolomics-advanced steatohepatitis fibrosis score (MASEF). Hepatology 2024; 79:135-148. [PMID: 37505221 PMCID: PMC10718221 DOI: 10.1097/hep.0000000000000542] [Citation(s) in RCA: 32] [Impact Index Per Article: 32.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/08/2022] [Accepted: 07/08/2023] [Indexed: 07/29/2023]
Abstract
BACKGROUND Early identification of those with NAFLD activity score ≥ 4 and significant fibrosis (≥F2) or at-risk metabolic dysfunction-associated steatohepatitis (MASH) is a priority as these patients are at increased risk for disease progression and may benefit from therapies. We developed and validated a highly specific metabolomics-driven score to identify at-risk MASH. METHODS We included derivation (n = 790) and validation (n = 565) cohorts from international tertiary centers. Patients underwent laboratory assessment and liver biopsy for metabolic dysfunction-associated steatotic liver disease. Based on 12 lipids, body mass index, aspartate aminotransferase, and alanine aminotransferase, the MASEF score was developed to identify at-risk MASH and compared to the FibroScan-AST (FAST) score. We further compared the performance of a FIB-4 + MASEF algorithm to that of FIB-4 + liver stiffness measurements (LSM) by vibration-controlled transient elastography (VCTE). RESULTS The diagnostic performance of the MASEF score showed an area under the receiver-operating characteristic curve, sensitivity, specificity, and positive and negative predictive values of 0.76 (95% CI 0.72-0.79), 0.69, 0.74, 0.53, and 0.85 in the derivation cohort, and 0.79 (95% CI 0.75-0.83), 0.78, 0.65, 0.48, and 0.88 in the validation cohort, while FibroScan-AST performance in the validation cohort was 0.74 (95% CI 0.68-0.79; p = 0.064), 0.58, 0.79, 0.67, and 0.73, respectively. FIB-4+MASEF showed similar overall performance compared with FIB-4 + LSM by VCTE ( p = 0.69) to identify at-risk MASH. CONCLUSION MASEF is a promising diagnostic tool for the assessment of at-risk MASH. It could be used alternatively to LSM by VCTE in the algorithm that is currently recommended by several guidance publications.
Collapse
Affiliation(s)
- Mazen Noureddin
- Houston Methodist Hospital, Houston Research Institute Houston, Texas, USA
- Houston Research Institute, Houston, Texas, USA
| | - Emily Truong
- Karsh Division of Gastroenterology and Hepatology, Cedars-Sinai Medical Center, Los Angeles, CA
| | | | | | | | - Jesus M. Banales
- Biodonostia Research Institute, Donostia University Hospital, University of the Basque Country (UPV-EHU), CIBERehd, IKERBASQUE, Donostia, Spain
- Department of Biochemistry and Genetics, School of Sciences, University of Navarra, Pamplona, Spain
| | - Marco Arrese
- Department of Gastroenterology, School of Medicine, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Kenneth Cusi
- University of Florida, Gainesville, Florida, USA
| | | | - Radan Bruha
- General University Hospital and the First Faculty of Medicine, Charles University, Prague, Czech Republic
| | | | - Paula Iruzubieta
- Marqués de Valdecilla University Hospital, Cantabria University, IDIVAL, Santander, Spain
| | - Rocio Aller
- Clinic University Hospital, University of Valladolid, Valladolid, Spain
| | | | | | | | - Patricia Aspichueta
- Department of Physiology, Faculty of Medicine and Nursing, University of the Basque Country UPV/EHU, Leioa, Spain
- Biocruces Bizkaia Health Research Institute, Barakaldo, Spain
- National Institute for the Study of Liver and Gastrointestinal Diseases (CIBERehd, Instituto de Salud Carlos III), Madrid, Spain
| | | | - Tatyana Kushner
- Division of Liver Diseases, Icahn School of Medicine at Mount Sinai, New York City, New York, USA
| | | | | | - Quentin M. Anstee
- Translational & Clinical Research Institute, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne, United Kingdom
| | - Javier Crespo
- Marqués de Valdecilla University Hospital, Cantabria University, IDIVAL, Santander, Spain
| | - José M. Mato
- National Institute for the Study of Liver and Gastrointestinal Diseases (CIBERehd, Instituto de Salud Carlos III), Madrid, Spain
- CIC bioGUNE, Basque Research and Technology Alliance (BRTA), Derio, Spain
| | - Arun J. Sanyal
- Virginia Commonwealth University Medical Center, Richmond, Virginia, USA
| |
Collapse
|
53
|
Chouik Y, Levrero M. Monocyte phenotypic liquid biopsy for NASH and liver fibrosis diagnosis: a new kid on the block. Gut 2023; 73:10-11. [PMID: 37328260 DOI: 10.1136/gutjnl-2022-328189] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/24/2022] [Accepted: 11/23/2022] [Indexed: 06/18/2023]
Affiliation(s)
- Yasmina Chouik
- Cancer Research Center of Lyon (CRCL), U1052, INSERM, Lyon, France
- Department of Hepatology, Hopital de la Croix Rousse, Hospices Civils de Lyon, Lyon, France
- Institute of Hepatology of Lyon, Lyon, France
| | - Massimo Levrero
- Cancer Research Center of Lyon (CRCL), U1052, INSERM, Lyon, France
- Department of Hepatology, Hopital de la Croix Rousse, Hospices Civils de Lyon, Lyon, France
- Institute of Hepatology of Lyon, Lyon, France
| |
Collapse
|
54
|
Lin H, Qiu S, Yang Y, Yang C, Shen Z, Chen Y, Zhang Z, Feng Y, Yan F. Three-dimensional magnetic resonance elastography combining proton-density fat fraction precisely identifies metabolic dysfunction-associated steatohepatitis with significant fibrosis. Magn Reson Imaging 2023; 104:1-8. [PMID: 37553044 DOI: 10.1016/j.mri.2023.07.017] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2023] [Revised: 07/30/2023] [Accepted: 07/31/2023] [Indexed: 08/10/2023]
Abstract
PURPOSE Patients with metabolic dysfunction-associated steatohepatitis (MASH) and significant fibrosis (fibrosis stage≥2), known as Fibro-MASH, are at increased risk of liver-related outcomes and lower rates of spontaneous disease regression. The aim was to investigate three-dimensional MR elastography (3D-MRE) combining proton-density fat fraction (PDFF) as a means of identifying Fibro-MASH. METHODS Forty-eight New Zealand rabbits were fed a high-fat/cholesterol or standard diet to obtain different disease activity and fibrosis stages. Shear stiffness (SS) and Damping Ratio (DR) were derived from 3D-MRE, whereas PDFF was from a volumetric 3D imaging sequence. Steatosis grade, metabolic dysfunction-associated steatotic liver disease activity score (MAS), and fibrosis stage were diagnosed histologically. Serum markers of fibrosis and inflammation were also measured. Correlation and comparison analysis, Receiver operating characteristic curves (ROC), Delong test, logistic regression analysis, and Net reclassification improvement (NRI) were performed. RESULTS PDFF correlated with steatosis grade (rho = 0.853). SS increased with developed liver fibrosis (rho = 0.837). DR correlated with MAS grade (rho = 0.678). The areas under the ROC (AUROCs) of SS for fibrosis grading were 0.961 and 0.953 for ≥F2, and ≥ F3, respectively. All the biochemical parameters were considered but excluded from the logistic regression analysis to identify Fibro-MASH. FF, SS, and DR were finally included in the further analysis. The three-parameter model combining PDFF, SS, and DR showed significant improvement in NRI over the model combining SS and PDFF (AUROC 0.973 vs. 0.906, P = 0.081; NRI 0.28, P < 0.05). CONCLUSION 3D-MRE combining PDFF may characterize the state of fat content, disease activity and fibrosis, thus precisely identify Fibro-MASH.
Collapse
Affiliation(s)
- Huimin Lin
- Department of Radiology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| | - Suhao Qiu
- Department of Radiology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China; School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, China.
| | - Yanzhao Yang
- Department of Radiology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| | - Chunxue Yang
- Department of Pathology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| | - Zhehan Shen
- Department of Radiology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| | - Yu Chen
- School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, China.
| | - Zhihan Zhang
- Department of Pathology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| | - Yuan Feng
- School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, China.
| | - Fuhua Yan
- Department of Radiology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China; School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, China.
| |
Collapse
|
55
|
Wai-Sun Wong V, Anstee QM, Nitze LM, Geerts A, George J, Nolasco V, Kjær MS, Ladelund S, Newsome PN, Ratziu V. FibroScan-aspartate aminotransferase (FAST) score for monitoring histological improvement in non-alcoholic steatohepatitis activity during semaglutide treatment: post-hoc analysis of a randomised, double-blind, placebo-controlled, phase 2b trial. EClinicalMedicine 2023; 66:102310. [PMID: 38058795 PMCID: PMC10696384 DOI: 10.1016/j.eclinm.2023.102310] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/15/2023] [Revised: 10/24/2023] [Accepted: 10/26/2023] [Indexed: 12/08/2023] Open
Abstract
Background Currently, assessment of candidate pharmacotherapies in patients with non-alcoholic steatohepatitis (NASH) involves invasive liver biopsy. Non-invasive scores, such as the FibroScan-aspartate aminotransferase (FAST) score, are used to identify candidates for therapy, but their ability to assess disease progression or treatment effect is unknown. We aimed to assess the association between FAST score and histological endpoints. Methods We conducted a post-hoc analysis using data from a prior randomised, double-blind, placebo-controlled, phase 2b trial at 143 sites across 16 countries. Patients (aged 18-75 years) with biopsy-confirmed NASH, fibrosis stage 1-3, and a Non-alcoholic fatty liver disease Activity Score (NAS) ≥4 were enrolled between January 2017 and September 2018, and randomly assigned to receive once-daily subcutaneous semaglutide 0.1, 0.2, or 0.4 mg or placebo for 72 weeks. A subgroup analysis of patients with FAST score and histological data in the pooled semaglutide treatment and placebo arms at baseline and week 72 was performed. The original trial is registered at ClinicalTrials.gov, NCT02970942. Findings A total of 122 patients were included in this post-hoc analysis (93 received semaglutide and 29 received placebo). FAST score reduction was associated with achieving the primary endpoint of NASH resolution without worsening of fibrosis in the pooled semaglutide group (area under the receiver operating curve 0.69; 95% confidence interval [CI] 0.58, 0.81). Mean FAST score reduction from baseline to week 72 was greatest in patients who met the primary endpoint vs those who did not in both the semaglutide (-0.40 [95% CI -0.84, 0.04] vs -0.22 [95% CI -0.74, 0.30] points; p = 0.002) and placebo groups (-0.25 [95% CI -0.72, 0.23] vs 0.00 [95% CI -0.50, 0.50] points; p = 0.047). Similarly, mean reductions in FAST score at week 72 were greater in those with NAS improvement vs those without in the semaglutide and placebo groups (≥1 point, -0.36 [95% CI -0.82, 0.11] vs -0.08 [95% CI -0.53, 0.38] points [p < 0.001] and -0.25 [95% CI -0.64, 0.14] vs -0.06 [95% CI -0.40, 0.53] points [p = 0.001]; ≥2 points, -0.40 [95% CI -0.86, 0.06] vs -0.14 [95% CI -0.56, 0.28] points [p < 0.001] and -0.29 [95% CI -0.67, 0.09] vs -0.05 [95% CI -0.40, 0.50] points; [p < 0.001]). A FAST score reduction of more than 0.22 points after semaglutide treatment was associated with meeting the primary endpoint (sensitivity 78%; specificity 60%; positive likelihood ratio 1.26; negative likelihood ratio 0.25; odds ratio 4.93). Interpretation The potential of the FAST score as a non-invasive monitoring tool to identify histological changes following treatment requires further evaluation and validation. Funding Novo Nordisk A/S.
Collapse
Affiliation(s)
- Vincent Wai-Sun Wong
- Department of Medicine and Therapeutics, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Quentin M. Anstee
- Translational & Clinical Research Institute, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne, United Kingdom
- NIHR Newcastle Biomedical Research Centre, Newcastle upon Tyne Hospitals NHS Foundation Trust, Newcastle upon Tyne, United Kingdom
| | | | - Anja Geerts
- Department of Gastroenterology and Hepatology, Hepatology Research Unit, Ghent University, Ghent, Belgium
| | - Jacob George
- Storr Liver Centre, The Westmead Institute for Medical Research, Westmead Hospital and University of Sydney, Sydney, NSW, Australia
| | | | | | | | - Philip N. Newsome
- NIHR Birmingham Biomedical Research Centre at University Hospitals Birmingham NHS Foundation Trust and the University of Birmingham, Birmingham, United Kingdom
| | - Vlad Ratziu
- Institute for Cardiometabolism and Nutrition, Sorbonne Université, Hôpital Pitié-Salpêtrière, APHP, INSERM UMRS 1138 CRC, Paris, France
| |
Collapse
|
56
|
Boeriu A, Dobru D, Fofiu C. Non-Invasive Diagnostic of NAFLD in Type 2 Diabetes Mellitus and Risk Stratification: Strengths and Limitations. Life (Basel) 2023; 13:2262. [PMID: 38137863 PMCID: PMC10744403 DOI: 10.3390/life13122262] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2023] [Revised: 10/26/2023] [Accepted: 11/25/2023] [Indexed: 12/24/2023] Open
Abstract
The progressive potential of liver damage in type 2 diabetes mellitus (T2DM) towards advanced fibrosis, end-stage liver disease, and hepatocarcinoma has led to increased concern for quantifying liver injury and individual risk assessment. The combination of blood-based markers and imaging techniques is recommended for the initial evaluation in NAFLD and for regular monitoring to evaluate disease progression. Continued development of ultrasonographic and magnetic resonance imaging methods for accurate quantification of liver steatosis and fibrosis, as well as promising tools for the detection of high-risk NASH, have been noted. In this review, we aim to summarize available evidence regarding the usefulness of non-invasive methods for the assessment of NAFLD in T2DM. We focus on the power and limitations of various methods for diagnosis, risk stratification, and patient monitoring that support their implementation in clinical setting or in research field.
Collapse
Affiliation(s)
- Alina Boeriu
- Gastroenterology Department, University of Medicine Pharmacy, Sciences, and Technology “George Emil Palade” Targu Mures, 540142 Targu Mures, Romania;
- Gastroenterology Department, Mures County Clinical Hospital, 540103 Targu Mures, Romania
| | - Daniela Dobru
- Gastroenterology Department, University of Medicine Pharmacy, Sciences, and Technology “George Emil Palade” Targu Mures, 540142 Targu Mures, Romania;
- Gastroenterology Department, Mures County Clinical Hospital, 540103 Targu Mures, Romania
| | - Crina Fofiu
- Gastroenterology Department, University of Medicine Pharmacy, Sciences, and Technology “George Emil Palade” Targu Mures, 540142 Targu Mures, Romania;
- Internal Medicine Department, Bistrita County Clinical Hospital, 420094 Bistrita, Romania
| |
Collapse
|
57
|
Jia X, Song E, Liu Y, Chen J, Wan P, Hu Y, Ye D, Chakrabarti S, Mahajan H, George J, Yan S, Yu Y, Zhang G, Wang Y, Yang W, Wu L, Hua S, Lee CH, Li H, Jiang X, Lam KSL, Wang C, Xu A. Identification and multicentric validation of soluble CDCP1 as a robust serological biomarker for risk stratification of NASH in obese Chinese. Cell Rep Med 2023; 4:101257. [PMID: 37918406 PMCID: PMC10694619 DOI: 10.1016/j.xcrm.2023.101257] [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: 05/18/2023] [Revised: 08/15/2023] [Accepted: 10/03/2023] [Indexed: 11/04/2023]
Abstract
The definitive diagnosis of non-alcoholic steatohepatitis (NASH) currently relies on invasive and labor-intensive liver biopsy. Here, we identified soluble CUB domain-containing protein 1 (sCDCP1) as a top-ranked non-invasive biomarker for NASH using Olink-based proteomics in 238 obese individuals with liver biopsies. Both the circulating concentration and hepatic mRNA abundance of sCDCP1 were significantly elevated in patients with NASH and correlated closely with each histological feature of NASH. In the pooled multicenter validation cohort, sCDCP1 as a standalone biomarker achieved an area under the receiver operating characteristic (AUROC) of 0.838 (95% confidence interval [CI] 0.789-0.887) for diagnosing NASH, which is better than those achieved with cytokeratin-18 and other non-invasive tests. Furthermore, the C-DAG model established by the combination of sCDCP1 with diabetes, aspartate aminotransferase (AST), and gender accurately rules in and rules out both NASH and fibrotic NASH (gray zones <20%). Thus, sCDCP1-based non-invasive tests can be potentially implemented for screening and early diagnosis of NASH and for ruling out low-risk individuals to avoid unnecessary liver biopsies.
Collapse
Affiliation(s)
- Xi Jia
- State Key Laboratory of Pharmaceutical Biotechnology, The University of Hong Kong, Hong Kong SAR, China; Department of Medicine, School of Clinical Medicine, The University of Hong Kong, Hong Kong SAR, China
| | - Erfei Song
- State Key Laboratory of Pharmaceutical Biotechnology, The University of Hong Kong, Hong Kong SAR, China; Department of Medicine, School of Clinical Medicine, The University of Hong Kong, Hong Kong SAR, China; Department of Metabolic and Bariatric Surgery, The First Affiliated Hospital of Jinan University, Guangzhou, China
| | - Yan Liu
- State Key Laboratory of Pharmaceutical Biotechnology, The University of Hong Kong, Hong Kong SAR, China
| | - Jiarui Chen
- State Key Laboratory of Pharmaceutical Biotechnology, The University of Hong Kong, Hong Kong SAR, China; Department of Medicine, School of Clinical Medicine, The University of Hong Kong, Hong Kong SAR, China
| | - Pei Wan
- State Key Laboratory of Pharmaceutical Biotechnology, The University of Hong Kong, Hong Kong SAR, China; Department of Medicine, School of Clinical Medicine, The University of Hong Kong, Hong Kong SAR, China
| | - Yue Hu
- State Key Laboratory of Pharmaceutical Biotechnology, The University of Hong Kong, Hong Kong SAR, China; Department of Medicine, School of Clinical Medicine, The University of Hong Kong, Hong Kong SAR, China
| | - Dewei Ye
- Key Laboratory of Glucolipid Metabolic Diseases of the Ministry of Education, Guangdong Pharmaceutical University, Guangzhou, China; Key Laboratory of Metabolic Phenotyping in Model Animals, Guangdong Pharmaceutical University, Guangzhou, China
| | - Subrata Chakrabarti
- Department of Pathology and Laboratory Medicine, Western University, London, ON, Canada; Department of Pathology and Laboratory Medicine, London Health Sciences Centre, London, ON, Canada
| | - Hema Mahajan
- Institute of Clinical Pathology and Medical Research, Pathology West, NSW Health Pathology, Sydney, NSW 2145, Australia; University of Western Sydney, Sydney, NSW, Australia
| | - Jacob George
- Storr Liver Centre, The Westmead Institute for Medical Research, Westmead Hospital and University of Sydney, Sydney, NSW, Australia
| | - Sen Yan
- Dr. Everett Chalmers Hospital, Fredericton, NB, Canada
| | - Yongtao Yu
- Department of Gastrointestinal Surgery, General Hospital of Ningxia Medical University, Yinchuan, China
| | - Guanghui Zhang
- Department of Gastrointestinal Surgery, Zhengzhou Second Hospital, Zhengzhou, China
| | - Yong Wang
- Department of General Surgery, The Second Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Wah Yang
- State Key Laboratory of Pharmaceutical Biotechnology, The University of Hong Kong, Hong Kong SAR, China; Department of Medicine, School of Clinical Medicine, The University of Hong Kong, Hong Kong SAR, China; Department of Metabolic and Bariatric Surgery, The First Affiliated Hospital of Jinan University, Guangzhou, China
| | - Lihong Wu
- Shenzhen Institute of Research and Innovation, The University of Hong Kong, Shenzhen, China
| | - Shuang Hua
- Key Laboratory of Glucolipid Metabolic Diseases of the Ministry of Education, Guangdong Pharmaceutical University, Guangzhou, China
| | - Chi Ho Lee
- State Key Laboratory of Pharmaceutical Biotechnology, The University of Hong Kong, Hong Kong SAR, China; Department of Medicine, School of Clinical Medicine, The University of Hong Kong, Hong Kong SAR, China
| | - Huixin Li
- State Key Laboratory of Pharmaceutical Biotechnology, The University of Hong Kong, Hong Kong SAR, China; Department of Medicine, School of Clinical Medicine, The University of Hong Kong, Hong Kong SAR, China
| | - Xue Jiang
- State Key Laboratory of Pharmaceutical Biotechnology, The University of Hong Kong, Hong Kong SAR, China; Department of Medicine, School of Clinical Medicine, The University of Hong Kong, Hong Kong SAR, China
| | - Karen S L Lam
- State Key Laboratory of Pharmaceutical Biotechnology, The University of Hong Kong, Hong Kong SAR, China; Department of Medicine, School of Clinical Medicine, The University of Hong Kong, Hong Kong SAR, China
| | - Cunchuan Wang
- Department of Metabolic and Bariatric Surgery, The First Affiliated Hospital of Jinan University, Guangzhou, China.
| | - Aimin Xu
- State Key Laboratory of Pharmaceutical Biotechnology, The University of Hong Kong, Hong Kong SAR, China; Department of Medicine, School of Clinical Medicine, The University of Hong Kong, Hong Kong SAR, China; Shenzhen Institute of Research and Innovation, The University of Hong Kong, Shenzhen, China; Department of Pharmacology and Pharmacy, The University of Hong Kong, Hong Kong SAR, China.
| |
Collapse
|
58
|
Canivet CM, Zheng MH, Qadri S, Vonghia L, Chuah KH, Costentin C, George J, Armandi A, Adams LA, Lange NF, Blanchet O, Moal V, Younes R, Roux M, Chan WK, Sturm N, Eslam M, Bugianesi E, Wang Z, Dufour JF, Francque S, Yki-Järvinen H, Zheng KI, Boursier J. Validation of the Blood Test MACK-3 for the Noninvasive Diagnosis of Fibrotic Nonalcoholic Steatohepatitis: An International Study With 1924 Patients. Clin Gastroenterol Hepatol 2023; 21:3097-3106.e10. [PMID: 37031715 DOI: 10.1016/j.cgh.2023.03.032] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/21/2022] [Revised: 02/03/2023] [Accepted: 03/24/2023] [Indexed: 04/11/2023]
Abstract
BACKGROUND & AIMS Drug development in nonalcoholic steatohepatitis (NASH) is hampered by a high screening failure rate that reaches 60% to 80% in therapeutic trials, mainly because of the absence of fibrotic NASH on baseline liver histology. MACK-3, a blood test including 3 biomarkers (aspartate aminotransferase, homeostasis model assessment, and cytokeratin 18), recently was developed for the noninvasive diagnosis of fibrotic NASH. We aimed to validate the diagnostic accuracy of this noninvasive test in an international multicenter study. METHODS A total of 1924 patients with biopsy-proven nonalcoholic fatty liver disease from 10 centers in Asia, Australia, and Europe were included. The blood test MACK-3 was calculated for all patients. FibroScan-aspartate aminotransferase score (FAST), an elastography-based test for fibrotic NASH, also was available in a subset of 655 patients. Fibrotic NASH was defined as the presence of NASH on liver biopsy with a Nonalcoholic Fatty Liver Disease Activity Score of 4 or higher and fibrosis stage of F2 or higher according to the NASH Clinical Research Network scoring system. RESULTS The area under the receiver operating characteristic of MACK-3 for fibrotic NASH was 0.791 (95% CI 0.768-0.814). Sensitivity at the previously published MACK-3 threshold of less than 0.135 was 91% and specificity at a greater than 0.549 threshold was 85%. The MACK-3 area under the receiver operating characteristic was not affected by age, sex, diabetes, or body mass index. MACK-3 and FAST results were well correlated (Spearman correlation coefficient, 0.781; P < .001). Except for an 8% higher rate of patients included in the grey zone, MACK-3 provided similar accuracy to that of FAST. Both tests included 27% of patients in their rule-in zone, with 85% specificity and 35% false positives (screen failure rate). CONCLUSIONS The blood test MACK-3 is an accurate tool to improve patient selection in NASH therapeutic trials.
Collapse
Affiliation(s)
- Clémence M Canivet
- Service d'Hépato-Gastroentérologie et Oncologie Digestive, Hôpital Universitaire d'Angers, Angers, France; Laboratoire HIFIH UPRES EA3859, SFR ICAT 4208, Université d'Angers, Angers, France
| | - Ming-Hua Zheng
- Nonalcoholic Fatty Liver Disease Research Center, Department of Hepatology, First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Sami Qadri
- Department of Medicine, University of Helsinki, Helsinki University Hospital, Helsinki, Finland; Minerva Foundation Institute for Medical Research, University Hospital of Helsinki, Helsinki, Finland
| | - Luisa Vonghia
- Department of Gastroenterology and Hepatology, Antwerp University Hospital, Antwerp, Belgium; Laboratory of Experimental Medicine and Pediatrics, Division of Gastroenterology-Hepatology, Faculty of Medicine and Health Sciences, University of Antwerp, Antwerp, Belgium
| | - Kee-Huat Chuah
- Gastroenterology and Hepatology Unit, Department of Medicine, Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia
| | - Charlotte Costentin
- Grenoble Institute for Advanced Biosciences, Research Center UGA/Inserm U 1209/CNRS 5309, Université Grenoble Alpes, Service d'Hépato-Gastroentérologie, Centre Hospitalier Universitaire Grenoble-Alpes, Grenoble, France
| | - Jacob George
- Storr Liver Centre, Westmead Hospital, University of Sydney, New South Wales, Australia
| | - Angelo Armandi
- Dipartimento di Scienze Mediche, Università di Torino, Turin, Italy
| | - Leon A Adams
- Medical School, University of Western Australia, Perth, Australia
| | - Naomi F Lange
- Department of Visceral Surgery and Medicine, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland; Graduate School for Health Sciences, University of Bern, Bern, Switzerland
| | - Odile Blanchet
- CRB-BB-0033-00038, Angers University Hospital, Angers, France
| | - Valérie Moal
- Biochemistry Department, Angers University Hospital, Angers, France
| | - Ramy Younes
- Dipartimento di Scienze Mediche, Università di Torino, Turin, Italy
| | - Marine Roux
- Laboratoire HIFIH UPRES EA3859, SFR ICAT 4208, Université d'Angers, Angers, France
| | - Wah-Kheong Chan
- Gastroenterology and Hepatology Unit, Department of Medicine, Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia
| | - Nathalie Sturm
- Service d'Anatomie et de Cytologie Pathologique, Centre Hospitalier Universitaire Grenoble-Alpes, La Tronche, France
| | - Mohammed Eslam
- Storr Liver Centre, Westmead Hospital, University of Sydney, New South Wales, Australia
| | | | - Zhengyi Wang
- Medical School, University of Western Australia, Perth, Australia
| | - Jean-François Dufour
- Centre des Maladies Digestives, Lausanne, Switzerland; Swiss Nonalcoholic Steatohepatitis Foundation, Bern, Switzerland
| | - Sven Francque
- Department of Gastroenterology and Hepatology, Antwerp University Hospital, Antwerp, Belgium; Laboratory of Experimental Medicine and Pediatrics, Division of Gastroenterology-Hepatology, Faculty of Medicine and Health Sciences, University of Antwerp, Antwerp, Belgium
| | - Hannele Yki-Järvinen
- Department of Medicine, University of Helsinki, Helsinki University Hospital, Helsinki, Finland; Minerva Foundation Institute for Medical Research, University Hospital of Helsinki, Helsinki, Finland
| | - Kenneth I Zheng
- Nonalcoholic Fatty Liver Disease Research Center, Department of Hepatology, First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Jérôme Boursier
- Service d'Hépato-Gastroentérologie et Oncologie Digestive, Hôpital Universitaire d'Angers, Angers, France; Laboratoire HIFIH UPRES EA3859, SFR ICAT 4208, Université d'Angers, Angers, France.
| |
Collapse
|
59
|
Ramezani M, Zobeiry M, Abdolahi S, Hatami B, Zali MR, Baghaei K. A crosstalk between epigenetic modulations and non-alcoholic fatty liver disease progression. Pathol Res Pract 2023; 251:154809. [PMID: 37797383 DOI: 10.1016/j.prp.2023.154809] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/30/2023] [Revised: 09/08/2023] [Accepted: 09/08/2023] [Indexed: 10/07/2023]
Abstract
Non-alcoholic fatty liver disease (NAFLD) has recently emerged as a major public health concern worldwide due to its rapidly rising prevalence and its potential to progress into end-stage liver disease. While the precise pathophysiology underlying NAFLD remains incompletely understood, it is strongly associated with various environmental triggers and other metabolic disorders. Epigenetics examines changes in gene expression that are not caused by alterations in the DNA sequence itself. There is accumulating evidence that epigenetics plays a key role in linking environmental cues to the onset and progression of NAFLD. Our understanding of how epigenetic mechanisms contribute to NAFLD pathophysiology has expanded considerably in recent years as research on the epigenetics of NAFLD has developed. This review summarizes recent insights into major epigenetic processes that have been implicated in NAFLD pathogenesis including DNA methylation, histone acetylation, and microRNAs that have emerged as promising targets for further investigation. Elucidating epigenetic mechanisms in NAFLD may uncover novel diagnostic biomarkers and therapeutic targets for this disease. However, many questions have remained unanswered regarding how epigenetics promotes NAFLD onset and progression. Additional studies are needed to further characterize the epigenetic landscape of NAFLD and validate the potential of epigenetic markers as clinical tools. Nevertheless, an enhanced understanding of the epigenetic underpinnings of NAFLD promises to provide key insights into disease mechanisms and pave the way for novel prognostic and therapeutic approaches.
Collapse
Affiliation(s)
- Meysam Ramezani
- Basic and Molecular Epidemiology of Gastrointestinal Disorders Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | | | - Shahrokh Abdolahi
- Basic and Molecular Epidemiology of Gastrointestinal Disorders Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| | - Behzad Hatami
- Basic and Molecular Epidemiology of Gastrointestinal Disorders Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mohammad Reza Zali
- Basic and Molecular Epidemiology of Gastrointestinal Disorders Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Kaveh Baghaei
- Basic and Molecular Epidemiology of Gastrointestinal Disorders Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran; Gastroenterology and Liver Diseases Research center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
60
|
Terracciani F, Falcomatà A, Gallo P, Picardi A, Vespasiani-Gentilucci U. Prognostication in NAFLD: physiological bases, clinical indicators, and newer biomarkers. J Physiol Biochem 2023; 79:851-868. [PMID: 36472795 DOI: 10.1007/s13105-022-00934-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2022] [Accepted: 11/23/2022] [Indexed: 12/12/2022]
Abstract
Non-alcoholic fatty liver disease (NAFLD) is becoming an epidemic in Western countries. Notably, while the majority of NAFLD patients will not evolve until advanced liver disease, a minority of them will progress towards liver-related events. Therefore, risk stratification and prognostication are emerging as fundamental in order to optimize human and economic resources for the care of these patients.Liver fibrosis has been clearly recognized as the main predictor of poor hepatic and extrahepatic outcomes. However, a prediction based only on the stage of fibrosis is near-sighted and static, as it does not capture the propensity of disease to further progress, the speed of progression and their changes over time. These determinants, which result from the interaction between genetic predisposition and acquired risk factors (obesity, diabetes, etc.), express themselves in disease activity, and can be synthesized by biomarkers of hepatic inflammation and fibrogenesis.In this review, we present the currently available clinical tools for risk stratification and prognostication in NAFLD specifically with respect to the risk of progression towards hard hepatic outcomes, i.e., liver-related events and death. We also discuss about the genetic and acquired drivers of disease progression, together with the physiopathological bases of their come into action. Finally, we introduce the most promising biomarkers in the direction of repeatedly assessing disease activity over time, mainly in response to future therapeutic interventions.
Collapse
Affiliation(s)
- Francesca Terracciani
- Hepatology and Clinical Medicine Unit, University Campus Bio-Medico of Rome, Rome, Italy
| | - Andrea Falcomatà
- Hepatology and Clinical Medicine Unit, University Campus Bio-Medico of Rome, Rome, Italy
| | - Paolo Gallo
- Hepatology and Clinical Medicine Unit, University Campus Bio-Medico of Rome, Rome, Italy.
| | - Antonio Picardi
- Hepatology and Clinical Medicine Unit, University Campus Bio-Medico of Rome, Rome, Italy
| | | |
Collapse
|
61
|
Hu Y, Li H, Zhang H, Chen X, Chen J, Xu Z, You H, Dong R, Peng Y, Li J, Li X, Wu D, Zhang L, Cao D, Jin H, Qiu D, Yang A, Lou J, Zhu X, Niu J, Ding Y. ZSP1601, a novel pan-phosphodiesterase inhibitor for the treatment of NAFLD, A randomized, placebo-controlled phase Ib/IIa trial. Nat Commun 2023; 14:6409. [PMID: 37828034 PMCID: PMC10570369 DOI: 10.1038/s41467-023-42162-0] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Accepted: 10/02/2023] [Indexed: 10/14/2023] Open
Abstract
Non-alcoholic fatty liver disease is a growing health burden with limited treatment options worldwide. Herein we report a randomized, double-blind, placebo-controlled, multiple-dose trial of a first-in-class pan-phosphodiesterase inhibitor ZSP1601 in 36 NAFLD patients (NCT04140123). There were three cohorts. Each cohort included twelve patients, nine of whom received ZSP1601 50 mg once daily, 50 mg twice daily, or 100 mg twice daily, and three of whom received matching placebos for 28 days. The primary outcomes were the safety and tolerability of ZSP1601. A total of 27 (27/36, 75%) patients experienced at least one treatment-emergent adverse event (TEAE). Most TEAEs were mild to moderate. There was no Serious Adverse Event. Diarrhea, transiently elevated creatinine and adaptive headache were frequently reported adverse drug reaction. We conclude that ZSP1601 is well-tolerated and safe, showing effective improvement in liver chemistries, liver fat content and fibrosis in patients with NAFLD.
Collapse
Affiliation(s)
- Yue Hu
- Phase I Clinical Research Center, First Hospital of Jilin University, Changchun, China
| | - Haijun Li
- Guangdong Raynovent Biotech Co., Ltd, Guangzhou, China
- Department of Anatomy and Neurobiology, School of Basic Medical Science, Central South University, Changsha, China
| | - Hong Zhang
- Phase I Clinical Research Center, First Hospital of Jilin University, Changchun, China
| | - Xiaoxin Chen
- Guangdong Raynovent Biotech Co., Ltd, Guangzhou, China
| | - Jinjun Chen
- Nafang Hospital, Nanfang Medical University, Guangzhou, China
| | - Zhongyuan Xu
- Nafang Hospital, Nanfang Medical University, Guangzhou, China
| | - Hong You
- Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Ruihua Dong
- Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Yun Peng
- Guangdong Raynovent Biotech Co., Ltd, Guangzhou, China
| | - Jing Li
- Guangdong Raynovent Biotech Co., Ltd, Guangzhou, China
| | - Xiaojiao Li
- Phase I Clinical Research Center, First Hospital of Jilin University, Changchun, China
| | - Dandan Wu
- Phase I Clinical Research Center, First Hospital of Jilin University, Changchun, China
| | - Lei Zhang
- Department of Radiology, First Hospital of Jilin University, Changchun, China
| | - Di Cao
- Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - He Jin
- Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Dongdong Qiu
- Department of Radiology, First Hospital of Jilin University, Changchun, China
| | - Aruhan Yang
- Phase I Clinical Research Center, First Hospital of Jilin University, Changchun, China
| | - Jinfeng Lou
- Phase I Clinical Research Center, First Hospital of Jilin University, Changchun, China
| | - Xiaoxue Zhu
- Phase I Clinical Research Center, First Hospital of Jilin University, Changchun, China.
| | - Junqi Niu
- Department of Hepatology, First Hospital of Jilin University, Changchun, China.
| | - Yanhua Ding
- Phase I Clinical Research Center, First Hospital of Jilin University, Changchun, China.
| |
Collapse
|
62
|
Kouvari M, Valenzuela-Vallejo L, Guatibonza-Garcia V, Polyzos SA, Deng Y, Kokkorakis M, Agraz M, Mylonakis SC, Katsarou A, Verrastro O, Markakis G, Eslam M, Papatheodoridis G, George J, Mingrone G, Mantzoros CS. Liver biopsy-based validation, confirmation and comparison of the diagnostic performance of established and novel non-invasive steatotic liver disease indexes: Results from a large multi-center study. Metabolism 2023; 147:155666. [PMID: 37527759 DOI: 10.1016/j.metabol.2023.155666] [Citation(s) in RCA: 41] [Impact Index Per Article: 20.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/31/2023] [Revised: 07/25/2023] [Accepted: 07/25/2023] [Indexed: 08/03/2023]
Abstract
BACKGROUND Non-invasive tools (NIT) for metabolic-dysfunction associated liver disease (MASLD) screening or diagnosis need to be thoroughly validated using liver biopsies. PURPOSE To externally validate NITs designed to differentiate the presence or absence of liver steatosis as well as more advanced disease stages, to confirm fully validated indexes (n = 7 NITs), to fully validate partially validated indexes (n = 5 NITs), and to validate for the first time one new index (n = 1 NIT). METHODS This is a multi-center study from two Gastroenterology-Hepatology Departments (Greece and Australia) and one Bariatric-Metabolic Surgery Department (Italy). Overall, n = 455 serum samples of patients with biopsy-proven MASLD (n = 374, including 237 patients with metabolic-dysfunction associated steatohepatitis (MASH)) and Controls (n = 81) were recruited. A complete validation analysis was performed to differentiate the presence of MASLD vs. Controls, MASH vs. metabolic-dysfunction associated steatotic liver (MASL), histological features of MASH, and fibrosis stages. RESULTS The index of NASH (ION) demonstrated the highest differentiation ability for the presence of MASLD vs. Controls, with the area under the curve (AUC) being 0.894. For specific histological characterization of MASH, no NIT demonstrated adequate performance, while in the case of specific features of MASH, such as hepatocellular ballooning and lobular inflammation, ION demonstrated the best performance with AUC being close to or above 0.850. For fibrosis (F) classification, the highest AUC was reached by the aspartate aminotransferase to platelet ratio index (APRI) being ~0.850 yet only with the potential to differentiate the severe fibrosis stages (F3, F4) vs. mild or moderate fibrosis (F0-2) with an AUC > 0.900 in patients without T2DM. When we excluded patients with morbid obesity, the differentiation ability of APRI was improved, reaching AUC = 0.802 for differentiating the presence of fibrosis F2-4 vs. F0-1. The recommended by current guidelines index FIB-4 seemed to differentiate adequately between severe (i.e., F3-4) and mild or moderate fibrosis (F0-2) with an AUC = 0.820, yet this was not the case when FIB-4 was used to classify patients with fibrosis F2-4 vs. F0-1. Trying to improve the predictive value of all NITs, using Youden's methodology, to optimize the suggested cut-off points did not materially improve the results. CONCLUSIONS The validation of currently available NITs using biopsy-proven samples provides new evidence for their ability to differentiate between specific disease stages, histological features, and, most importantly, fibrosis grading. The overall performance of the examined NITs needs to be further improved for applications in the clinic.
Collapse
Affiliation(s)
- Matina Kouvari
- Department of Medicine, Beth-Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, United States of America
| | - Laura Valenzuela-Vallejo
- Department of Medicine, Beth-Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, United States of America
| | - Valentina Guatibonza-Garcia
- Department of Medicine, Beth-Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, United States of America
| | - Stergios A Polyzos
- First Laboratory of Pharmacology, School of Medicine, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Yixiang Deng
- Department of Medicine, Beth-Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, United States of America
| | - Michail Kokkorakis
- Department of Medicine, Beth-Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, United States of America; Department of Clinical Pharmacy and Pharmacology, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| | - Melih Agraz
- Department of Medicine, Beth-Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, United States of America
| | - Sophia C Mylonakis
- Department of Medicine, Beth-Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, United States of America
| | - Angeliki Katsarou
- Department of Medicine, Beth-Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, United States of America
| | - Ornella Verrastro
- Department of Gastroenterology, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Georgios Markakis
- Department of Gastroenterology, Medical School of National and Kapodistrian University of Athens, General Hospital of Athens "Laiko", Athens, Greece
| | - Mohammed Eslam
- Storr Liver Centre, Westmead Institute for Medical Research, Westmead Hospital and University of Sydney, Sydney, New South Wales, Australia
| | - Georgios Papatheodoridis
- Department of Gastroenterology, Medical School of National and Kapodistrian University of Athens, General Hospital of Athens "Laiko", Athens, Greece
| | - Jacob George
- Storr Liver Centre, Westmead Institute for Medical Research, Westmead Hospital and University of Sydney, Sydney, New South Wales, Australia.
| | - Geltrude Mingrone
- Department of Gastroenterology, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Christos S Mantzoros
- Department of Medicine, Beth-Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, United States of America; Department of Medicine, Boston VA Healthcare System, Boston, MA 02130, United States of America
| |
Collapse
|
63
|
Li J, Lu X, Zhu Z, Kalutkiewicz KJ, Mounajjed T, Therneau TM, Venkatesh SK, Sui Y, Glaser KJ, Hoodeshenas S, Manduca A, Shah VH, Ehman RL, Allen AM, Yin M. Head-to-head comparison of magnetic resonance elastography-based liver stiffness, fat fraction, and T1 relaxation time in identifying at-risk NASH. Hepatology 2023; 78:1200-1208. [PMID: 37080558 PMCID: PMC10521779 DOI: 10.1097/hep.0000000000000417] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/28/2022] [Revised: 03/14/2023] [Accepted: 03/24/2023] [Indexed: 04/22/2023]
Abstract
BACKGROUND AND AIMS The presence of at-risk NASH is associated with an increased risk of cirrhosis and complications. Therefore, noninvasive identification of at-risk NASH with an accurate biomarker is a critical need for pharmacologic therapy. We aim to explore the performance of several magnetic resonance (MR)-based imaging parameters in diagnosing at-risk NASH. APPROACH AND RESULTS This prospective clinical trial (NCT02565446) includes 104 paired MR examinations and liver biopsies performed in patients with suspected or diagnosed NAFLD. Magnetic resonance elastography-assessed liver stiffness (LS), 6-point Dixon-derived proton density fat fraction (PDFF), and single-point saturation-recovery acquisition-calculated T1 relaxation time were explored. Among all predictors, LS showed the significantly highest accuracy in diagnosing at-risk NASH [AUC LS : 0.89 (0.82, 0.95), AUC PDFF : 0.70 (0.58, 0.81), AUC T1 : 0.72 (0.61, 0.82), z -score test z >1.96 for LS vs any of others]. The optimal cutoff value of LS to identify at-risk NASH patients was 3.3 kPa (sensitivity: 79%, specificity: 82%, negative predictive value: 91%), whereas the optimal cutoff value of T1 was 850 ms (sensitivity: 75%, specificity: 63%, and negative predictive value: 87%). PDFF had the highest performance in diagnosing NASH with any fibrosis stage [AUC PDFF : 0.82 (0.72, 0.91), AUC LS : 0.73 (0.63, 0.84), AUC T1 : 0.72 (0.61, 0.83), |z| <1.96 for all]. CONCLUSION Magnetic resonance elastography-assessed LS alone outperformed PDFF, and T1 in identifying patients with at-risk NASH for therapeutic trials.
Collapse
Affiliation(s)
- Jiahui Li
- Department of Radiology, Mayo Clinic, Rochester, Minnesota, USA
| | - Xin Lu
- Department of Radiology, Mayo Clinic, Rochester, Minnesota, USA
- Department of Radiology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Zheng Zhu
- Department of Radiology, Mayo Clinic, Rochester, Minnesota, USA
| | | | - Taofic Mounajjed
- Division of Anatomic Pathology, Mayo Clinic, Rochester, Minnesota, USA
| | - Terry M. Therneau
- Department of Biomedical Statistics and Informatics, Mayo Clinic, Rochester, Minnesota, USA
| | | | - Yi Sui
- Department of Radiology, Mayo Clinic, Rochester, Minnesota, USA
| | - Kevin J. Glaser
- Department of Radiology, Mayo Clinic, Rochester, Minnesota, USA
| | | | - Armando Manduca
- Department of Radiology, Mayo Clinic, Rochester, Minnesota, USA
| | - Vijay H. Shah
- Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, Minnesota, USA
| | | | - Alina M. Allen
- Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, Minnesota, USA
| | - Meng Yin
- Department of Radiology, Mayo Clinic, Rochester, Minnesota, USA
| |
Collapse
|
64
|
Sanyal AJ, Shankar SS, Yates KP, Bolognese J, Daly E, Dehn CA, Neuschwander-Tetri B, Kowdley K, Vuppalanchi R, Behling C, Tonascia J, Samir A, Sirlin C, Sherlock SP, Fowler K, Heymann H, Kamphaus TN, Loomba R, Calle RA. Diagnostic performance of circulating biomarkers for non-alcoholic steatohepatitis. Nat Med 2023; 29:2656-2664. [PMID: 37679433 PMCID: PMC10579051 DOI: 10.1038/s41591-023-02539-6] [Citation(s) in RCA: 49] [Impact Index Per Article: 24.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2023] [Accepted: 08/08/2023] [Indexed: 09/09/2023]
Abstract
There are no approved diagnostic biomarkers for at-risk non-alcoholic steatohepatitis (NASH), defined by the presence of NASH, high histological activity and fibrosis stage ≥2, which is associated with higher incidence of liver-related events and mortality. FNIH-NIMBLE is a multi-stakeholder project to support regulatory approval of NASH-related biomarkers. The diagnostic performance of five blood-based panels was evaluated in an observational (NASH CRN DB2) cohort (n = 1,073) with full spectrum of non-alcoholic fatty liver disease (NAFLD). The panels were intended to diagnose at-risk NASH (NIS4), presence of NASH (OWLiver) or fibrosis stages >2, >3 or 4 (enhanced liver fibrosis (ELF) test, PROC3 and FibroMeter VCTE). The prespecified performance metric was an area under the receiver operating characteristic curve (AUROC) ≥0.7 and superiority over alanine aminotransferase for disease activity and the FIB-4 test for fibrosis severity. Multiple biomarkers met these metrics. NIS4 had an AUROC of 0.81 (95% confidence interval: 0.78-0.84) for at-risk NASH. The AUROCs of the ELF test, PROC3 and FibroMeterVCTE for clinically significant fibrosis (≥stage 2), advanced fibrosis (≥stage 3) or cirrhosis (stage 4), respectively, were all ≥0.8. ELF and FibroMeter VCTE outperformed FIB-4 for all fibrosis endpoints. These data represent a milestone toward qualification of several biomarker panels for at-risk NASH and also fibrosis severity in individuals with NAFLD.
Collapse
Affiliation(s)
- Arun J Sanyal
- Virginia Commonwealth University School of Medicine, Richmond, VA, USA.
| | | | - Katherine P Yates
- Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD, USA
| | | | | | | | | | | | - Raj Vuppalanchi
- Indiana University School of Medicine, Indianapolis, IN, USA
| | - Cynthia Behling
- Department of Pathology, University of California San Diego School of Medicine, San Diego, CA, USA
| | - James Tonascia
- Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD, USA
| | - Anthony Samir
- Center for Ultrasound Research & Translation, Department of Radiology, Massachusetts General Hospital,Harvard Medical School, Boston, MA, USA
| | - Claude Sirlin
- Deptartment of Radiology, University of California San Diego School of Medicine, San Diego, CA, USA
| | | | - Kathryn Fowler
- Deptartment of Radiology, University of California San Diego School of Medicine, San Diego, CA, USA
| | - Helen Heymann
- US Food and Drug Administration, Silver Springs, MD, USA
| | | | - Rohit Loomba
- NAFLD Research Center, Division of Gastroenterology, Department of Medicine, University of California, San Diego, La Jolla, CA, USA
| | | |
Collapse
|
65
|
Xu Q, Feng M, Ren Y, Liu X, Gao H, Li Z, Su X, Wang Q, Wang Y. From NAFLD to HCC: Advances in noninvasive diagnosis. Biomed Pharmacother 2023; 165:115028. [PMID: 37331252 DOI: 10.1016/j.biopha.2023.115028] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2023] [Revised: 06/10/2023] [Accepted: 06/14/2023] [Indexed: 06/20/2023] Open
Abstract
Non-alcoholic fatty liver disease (NAFLD) has gradually become one of the major liver health problems in the world. The dynamic course of the disease goes through steatosis, inflammation, fibrosis, and carcinoma. Before progressing to carcinoma, timely and effective intervention will make the condition better, which highlights the importance of early diagnosis. With the further study of the biological mechanism in the pathogenesis and progression of NAFLD, some potential biomarkers have been discovered, and the possibility of their clinical application is gradually being discussed. At the same time, the progress of imaging technology and the emergence of new materials and methods also provide more possibilities for the diagnosis of NAFLD. This article reviews the diagnostic markers and advanced diagnostic methods of NAFLD in recent years.
Collapse
Affiliation(s)
- Qinchen Xu
- Department of Clinical Laboratory, The Second Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250033, China
| | - Maoxiao Feng
- Department of Clinical Laboratory, Shandong Provincial Hospital Affiliated to Shandong First Medical University, 250021, Jinan, Shandong Province, China
| | - Yidan Ren
- Department of Clinical Laboratory, The Second Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250033, China
| | - Xiaoyan Liu
- Department of Clinical Laboratory, Shandong Provincial Hospital Affiliated to Shandong First Medical University, 250021, Jinan, Shandong Province, China
| | - Huiru Gao
- Department of Clinical Laboratory, The Second Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250033, China
| | - Zigan Li
- Department of Clinical Laboratory, The Second Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250033, China
| | - Xin Su
- Department of Clinical Laboratory, The Second Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250033, China
| | - Qin Wang
- Department of Anesthesiology, Qilu Hospital, Shandong University, 107 Wenhua Xi Road, Jinan 250012, China.
| | - Yunshan Wang
- Department of Clinical Laboratory, Shandong Provincial Hospital Affiliated to Shandong First Medical University, 250021, Jinan, Shandong Province, China.
| |
Collapse
|
66
|
Truong E, Gornbein JA, Yang JD, Noureddin N, Harrison SA, Alkhouri N, Noureddin M. MRI-AST (MAST) Score Accurately Predicts Major Adverse Liver Outcome, Hepatocellular Carcinoma, Liver Transplant, and Liver-Related Death. Clin Gastroenterol Hepatol 2023; 21:2570-2577.e1. [PMID: 36813013 DOI: 10.1016/j.cgh.2023.02.003] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/22/2022] [Revised: 01/17/2023] [Accepted: 02/03/2023] [Indexed: 02/24/2023]
Abstract
BACKGROUND & AIMS The MRI-AST (MAST) score accurately identifies patients with at-risk nonalcoholic steatohepatitis, defined as nonalcoholic steatohepatitis with nonalcoholic fatty liver disease activity score ≥4 and fibrosis stage ≥2 at highest risk for disease progression. It is important to determine the robustness of the MAST score in predicting major adverse liver outcomes (MALO), hepatocellular carcinoma (HCC), liver transplant, and death. METHODS This retrospective analysis included patients with nonalcoholic fatty liver disease from a tertiary care center who underwent magnetic resonance imaging proton density fat fraction, magnetic resonance elastography, and laboratory testing within 6 months from 2013 to 2022. Other causes of chronic liver disease were excluded. Hazard ratios between logit MAST and MALO (ascites, hepatic encephalopathy, or bleeding esophageal varices), liver transplant, HCC, or liver-related death were computed using a Cox proportional hazards regression model. We computed the hazard ratio of MALO or death associated with MAST scores 0.165-0.242 and 0.242-1.000, using MAST scores 0.000-0.165 as the reference group. RESULTS Among 346 total patients, average age was 58.8 years with 52.9% females and 34.4% with type 2 diabetes. Average alanine aminotransferase was 50.7 IU/L (24.3-60.0 IU/L), aspartate aminotransferase was 38.05 IU/L (22.00-41.00 IU/L), platelets were 242.9 × 109/L (193.8-290.0 × 109/L), proton density fat fraction was 12.90% (5.90%-18.22%), and liver stiffness on magnetic resonance elastography was 2.75 kPa (2.07-2.90 kPa). Median follow-up was 29.5 months. Fourteen had adverse outcomes, including 10 MALO, 1 HCC, 1 liver transplant, and 2 liver-related deaths. The Cox regression of MAST versus adverse event rate had a hazard ratio of 2.01 (95% confidence interval, 1.59-2.54; P < .0001) for each 1 logit unit increases in MAST. The corresponding Harrell concordance statistic (C statistic) was 0.919 (95% confidence interval, 0.865-0.953). The MAST score ranges of 0.165-0.242 and 0.242-1.0, respectively, had adverse event rate hazard ratio of 7.75 (1.40-42.9; P = .0189) and 22.11 (6.59-74.2; P < .0000) relative to MAST 0-0.165. CONCLUSIONS The MAST score noninvasively identifies at-risk nonalcoholic steatohepatitis and accurately predicts MALO, HCC, liver transplant, and liver-related death.
Collapse
Affiliation(s)
- Emily Truong
- Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, California
| | - Jeffrey A Gornbein
- Department of Medicine, University of California Los Angeles, Los Angeles, California
| | - Ju Dong Yang
- Karsh Division of Gastroenterology and Hepatology, Cedars-Sinai Medical Center, Los Angeles, California; Comprehensive Transplant Center, Cedars Sinai Medical Center, Los Angeles, California
| | - Nabil Noureddin
- Division of Gasteroenterology, University of California San Diego, San Diego, California
| | | | | | - Mazen Noureddin
- Houston Methodist Hospital, Houston Research Institute, Houston, Texas.
| |
Collapse
|
67
|
Tun KM, Noureddin N, Noureddin M. Noninvasive tests in the evaluation of nonalcoholic fatty liver disease: A review. Clin Liver Dis (Hoboken) 2023; 22:103-112. [PMID: 37799634 PMCID: PMC10550044 DOI: 10.1097/cld.0000000000000066] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/27/2023] [Accepted: 04/08/2023] [Indexed: 10/07/2023] Open
Abstract
1_kpnc0f20Kaltura.
Collapse
Affiliation(s)
- Kyaw Min Tun
- Department of Internal Medicine, Kirk Kerkorian School of Medicine, University of Nevada, Las Vegas, Nevada, USA
| | - Nabil Noureddin
- Division of Gastroenterology, Department of Medicine, University of California, San Diego, School of Medicine, California, USA
| | - Mazen Noureddin
- Houston Research Institute and Houston Liver Institute, Texas, USA
| |
Collapse
|
68
|
Sanyal AJ, Magnanensi J, Majd Z, Rosenquist C, Vera DM, Almas JP, Connelly MA. NIS2+™, an effective blood-based test for the diagnosis of at-risk nonalcoholic steatohepatitis in adults 65 years and older. Hepatol Commun 2023; 7:e0223. [PMID: 37556372 PMCID: PMC10412428 DOI: 10.1097/hc9.0000000000000223] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/19/2023] [Accepted: 06/13/2023] [Indexed: 08/11/2023] Open
Abstract
BACKGROUND Older patients are at increased risk for at-risk NASH, defined as NASH with NAFLD activity scores (NAS) ≥4 and significant fibrosis (F ≥ 2). The aim of this study was to compare the performance of 2 new blood tests, NIS4® and NIS2+™, with FIB-4, NFS, ELF™, and alanine aminotransferase (ALT) for the diagnosis of at-risk NASH in a cohort of patients aged ≥65 years. METHODS The clinical performance of multiple blood-based tests was assessed for their ability to detect at-risk NASH using the RESOLVE-IT diag cohort, a large population of patients with metabolic risk who were screened for potential inclusion in the RESOLVE-IT phase 3 trial. RESULTS The study cohort (n = 2053) included patients with the full histological spectrum of NAFLD, with patients having liver fibrosis stages F0-4 and NAS scores 0-8. NIS4® and NIS2+™ showed similar assay performance in patients who were <65 versus ≥65 years of age (AUROC = 0.80 vs. 0.78, p = 0.47; 0.81 vs. 0.83 p = 0.45, respectively) for the identification of at-risk NASH. In patients ≥65 (n = 410), NIS2+™ exhibited the highest AUROC compared to NIS4®, FIB-4, NFS, ELF™, and ALT (AUROC = 0.83 vs. 0.78, 0.68, 0.58, 0.69, 0.74, respectively; all p ≤ 0.0009). For NIS2+™, the sensitivity and NPV for ruling-out at-risk NASH at the 0.46 cutoff were 90.2% and 86.0%, and the specificity and PPV for ruling-in at-risk NASH at the 0.68 cutoff were81.1% and 76.3%, respectively. CONCLUSIONS The clinical performance of NIS2+™ was superior for the diagnosis of at-risk NASH in patients ≥65 years of age. These data support the clinical value of this blood-based test for the diagnosis of at-risk NASH in older adults.
Collapse
Affiliation(s)
- Arun J. Sanyal
- Stravitz-Sanyal Institute for Liver Disease and Metabolic Health, Virginia Commonwealth University School of Medicine, Richmond, Virginia, USA
| | | | | | | | | | - James P. Almas
- Digital Innovation Group, Labcorp, Burlington, North Carolina, USA
| | | |
Collapse
|
69
|
Harrison SA, Ratziu V, Magnanensi J, Hajji Y, Deledicque S, Majd Z, Rosenquist C, Hum DW, Staels B, Anstee QM, Sanyal AJ. NIS2+™, an optimisation of the blood-based biomarker NIS4® technology for the detection of at-risk NASH: A prospective derivation and validation study. J Hepatol 2023; 79:758-767. [PMID: 37224923 DOI: 10.1016/j.jhep.2023.04.031] [Citation(s) in RCA: 26] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/14/2022] [Revised: 03/17/2023] [Accepted: 04/06/2023] [Indexed: 05/26/2023]
Abstract
BACKGROUND & AIMS NIS4® is a blood-based non-invasive test designed to effectively rule in/rule out at-risk non-alcoholic steatohepatitis (NASH), defined as non-alcoholic fatty liver disease activity score ≥4 and significant fibrosis (stage ≥2), among patients with metabolic risk factors. Robustness of non-invasive test scores across characteristics of interest including age, type 2 diabetes mellitus, and sex, and optimised analytical aspects are critical for large-scale implementation in clinical practice. We developed and validated NIS2+™, an optimisation of NIS4®, specifically designed to improve score robustness. METHODS A well-balanced training cohort (n = 198) included patients from the GOLDEN-505 trial. The validation (n = 684) and test (n = 2,035) cohorts included patients from the RESOLVE-IT trial. Well-matched subgroups were created to avoid potential confounding effects during modelling and analysis of score robustness. Models were trained using logistic regressions for at-risk NASH detection and compared using Bayesian information criteria. Performance of NIS2+™ was compared with that of NIS4®, Fibrosis-4, and alanine aminotransferase using area under the receiver operating characteristic curve, and robustness was analysed through score distribution. RESULTS Using the training cohort to compare all combinations of NIS4® biomarkers, NIS2 (miR-34a-5p, YKL-40) was identified as the best combination of parameters. To correct for the sex effect on miR-34a-5p (validation cohort), sex and sex ∗ miR-34a-5p parameters were added, creating NIS2+™. In the test cohort, NIS2+™ exhibited a statistically higher area under the receiver operating characteristic curve (0.813) vs. NIS4® (0.792; p = 0.0002), Fibrosis-4 (0.653; p <0.0001), and alanine aminotransferase (0.699; p <0.0001). NIS2+™ scores were not affected by age, sex, BMI, or type 2 diabetes mellitus status, providing robust clinical performances irrespective of patient characteristics. CONCLUSION NIS2+™ constitutes a robust optimisation of NIS4® technology for the detection of at-risk NASH. IMPACT AND IMPLICATIONS The development of non-invasive tests for accurate, large-scale detection of patients with at-risk non-alcoholic steatohepatitis (NASH; defined as NASH with non-alcoholic fatty liver disease activity score ≥4 and fibrosis stage ≥2) - who are at higher risk for disease progression and for developing liver-related life-threatening outcomes - is critical for identifying this patient population in the clinical setting and improving the screening process of NASH clinical trials. We report the development and validation of NIS2+™, a diagnostic test designed as an optimisation of NIS4® technology, a blood-based panel currently used to detect at-risk NASH in patients with metabolic risk factors. NIS2+™ showed improved performance for the detection of at-risk NASH compared with NIS4® and other non-invasive liver tests that was not impacted by patients' characteristics of interest, such as age, sex, type 2 diabetes mellitus, BMI, dyslipidaemia, and hypertension. This makes NIS2+™ a robust and reliable tool for the diagnosis of at-risk NASH among patients with metabolic risk factors, and an effective candidate for large-scale implementation in clinical practice and clinical trials.
Collapse
Affiliation(s)
- Stephen A Harrison
- Summit Clinical Research, San Antonio, TX, USA; Radcliffe Department of Medicine, University of Oxford, Oxford, UK
| | - Vlad Ratziu
- Sorbonne Université, Institute for Cardiometabolism and Nutrition, Hôpital Pitié-Salpêtrière, Paris, France
| | | | | | | | | | | | | | - Bart Staels
- Université de Lille, INSERM, CHU Lille, Institut Pasteur de Lille, Lille, France
| | - Quentin M Anstee
- Translational and Clinical Research Institute, Faculty of Medical Sciences, Newcastle University, Newcastle Upon Tyne, UK; Newcastle NIHR Biomedical Research Centre, Newcastle Upon Tyne Hospitals NHS Foundation Trust, Freeman Hospital, Newcastle Upon Tyne, UK
| | - Arun J Sanyal
- Division of Gastroenterology, Hepatology and Nutrition, Virginia Commonwealth University School of Medicine, Richmond, VA, USA
| |
Collapse
|
70
|
Vali Y, Lee J, Boursier J, Petta S, Wonders K, Tiniakos D, Bedossa P, Geier A, Francque S, Allison M, Papatheodoridis G, Cortez-Pinto H, Pais R, Dufour JF, Leeming DJ, Harrison SA, Chen Y, Cobbold JF, Pavlides M, Holleboom AG, Yki-Jarvinen H, Crespo J, Karsdal M, Ostroff R, Zafarmand MH, Torstenson R, Duffin K, Yunis C, Brass C, Ekstedt M, Aithal GP, Schattenberg JM, Bugianesi E, Romero-Gomez M, Ratziu V, Anstee QM, Bossuyt PM. Biomarkers for staging fibrosis and non-alcoholic steatohepatitis in non-alcoholic fatty liver disease (the LITMUS project): a comparative diagnostic accuracy study. Lancet Gastroenterol Hepatol 2023; 8:714-725. [PMID: 36958367 DOI: 10.1016/s2468-1253(23)00017-1] [Citation(s) in RCA: 82] [Impact Index Per Article: 41.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/21/2022] [Revised: 01/20/2023] [Accepted: 01/23/2023] [Indexed: 03/25/2023]
Abstract
BACKGROUND The reference standard for detecting non-alcoholic steatohepatitis (NASH) and staging fibrosis-liver biopsy-is invasive and resource intensive. Non-invasive biomarkers are urgently needed, but few studies have compared these biomarkers in a single cohort. As part of the Liver Investigation: Testing Marker Utility in Steatohepatitis (LITMUS) project, we aimed to evaluate the diagnostic accuracy of 17 biomarkers and multimarker scores in detecting NASH and clinically significant fibrosis in patients with non-alcoholic fatty liver disease (NAFLD) and identify their optimal cutoffs as screening tests in clinical trial recruitment. METHODS This was a comparative diagnostic accuracy study in people with biopsy-confirmed NAFLD from 13 countries across Europe, recruited between Jan 6, 2010, and Dec 29, 2017, from the LITMUS metacohort of the prospective European NAFLD Registry. Adults (aged ≥18 years) with paired liver biopsy and serum samples were eligible; those with excessive alcohol consumption or evidence of other chronic liver diseases were excluded. The diagnostic accuracy of the biomarkers was expressed as the area under the receiver operating characteristic curve (AUC) with liver histology as the reference standard and compared with the Fibrosis-4 index for liver fibrosis (FIB-4) in the same subgroup. Target conditions were the presence of NASH with clinically significant fibrosis (ie, at-risk NASH; NAFLD Activity Score ≥4 and F≥2) or the presence of advanced fibrosis (F≥3), analysed in all participants with complete data. We identified thres holds for each biomarker for reducing the number of biopsy-based screen failures when recruiting people with both NASH and clinically significant fibrosis for future trials. FINDINGS Of 1430 participants with NAFLD in the LITMUS metacohort with serum samples, 966 (403 women and 563 men) were included after all exclusion criteria had been applied. 335 (35%) of 966 participants had biopsy-confirmed NASH and clinically significant fibrosis and 271 (28%) had advanced fibrosis. For people with NASH and clinically significant fibrosis, no single biomarker or multimarker score significantly reached the predefined AUC 0·80 acceptability threshold (AUCs ranging from 0·61 [95% CI 0·54-0·67] for FibroScan controlled attenuation parameter to 0·81 [0·75-0·86] for SomaSignal), with accuracy mostly similar to FIB-4. Regarding detection of advanced fibrosis, SomaSignal (AUC 0·90 [95% CI 0·86-0·94]), ADAPT (0·85 [0·81-0·89]), and FibroScan liver stiffness measurement (0·83 [0·80-0·86]) reached acceptable accuracy. With 11 of 17 markers, histological screen failure rates could be reduced to 33% in trials if only people who were marker positive had a biopsy for evaluating eligibility. The best screening performance for NASH and clinically significant fibrosis was observed for SomaSignal (number needed to test [NNT] to find one true positive was four [95% CI 4-5]), then ADAPT (six [5-7]), MACK-3 (seven [6-8]), and PRO-C3 (nine [7-11]). INTERPRETATION None of the single markers or multimarker scores achieved the predefined acceptable AUC for replacing biopsy in detecting people with both NASH and clinically significant fibrosis. However, several biomarkers could be applied in a prescreening strategy in clinical trial recruitment. The performance of promising markers will be further evaluated in the ongoing prospective LITMUS study cohort. FUNDING The Innovative Medicines Initiative 2 Joint Undertaking.
Collapse
Affiliation(s)
- Yasaman Vali
- Epidemiology and Data Science, Amsterdam Public Health, University of Amsterdam, Amsterdam, Netherlands
| | - Jenny Lee
- Epidemiology and Data Science, Amsterdam Public Health, University of Amsterdam, Amsterdam, Netherlands
| | - Jerome Boursier
- Laboratoire Hémodynamique, Interaction Fibrose et Invasivité Tumorales Hépatiques, University Paris Research, Structure Fédérative de Recherche, Interactions Cellulaires et Applications Thérapeutiques 4208, University of Angers, Angers, France; Department of Hepato-Gastroenterology and Digestive Oncology, University Hospital of Angers, Angers, France
| | - Salvatore Petta
- Section of Gastroenterology and Hepatology, Promozione della Salute, Materno-Infantile, di Medicina Interna e Specialistica di Eccellenza, Department, University of Palermo, Palermo, Italy
| | - Kristy Wonders
- Translational and Clinical Research Institute, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne, UK
| | - Dina Tiniakos
- Translational and Clinical Research Institute, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne, UK; Department of Pathology, Aretaieion Hospital, National and Kapodistrian University of Athens, Athens, Greece
| | - Pierre Bedossa
- Translational and Clinical Research Institute, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne, UK
| | - Andreas Geier
- Division of Hepatology, Department of Medicine II, Wurzburg University Hospital, Wurzburg, Germany
| | - Sven Francque
- Department of Gastroenterology Hepatology, Antwerp University Hospital, Laboratory of Experimental Medicine and Paediatrics, University of Antwerp, Antwerp, Belgium
| | - Mike Allison
- Liver Unit, Department of Medicine, Cambridge National Institute for Health and Care Research Biomedical Research Centre, Cambridge University National Health Service Foundation Trust, Cambridge, UK
| | | | - Helena Cortez-Pinto
- University Clinic of Gastroenterology, Faculty of Medicine, University of Lisbon, Lisbon, Portugal
| | - Raluca Pais
- Public Assistance Hospital of Paris, Pitié Salpêtrière Hospital, Institute of Cardiometabolism and Nutrition, Sorbonne University, Paris, France
| | - Jean-Francois Dufour
- Hepatology, Department of Biomedical Research, University of Bern, Bern, Switzerland
| | | | | | - Yu Chen
- Lilly Research Laboratories, Eli Lilly, Indianapolis, IN, USA
| | - Jeremy F Cobbold
- Department of Gastroenterology and Hepatology, Oxford National Institute for Health and Care Research Biomedical Research Centre, Oxford University Hospitals, Oxford, UK
| | - Michael Pavlides
- Department of Medicine, Oxford National Institute for Health and Care Research Biomedical Research Centre, Oxford, UK
| | - Adriaan G Holleboom
- Department of Internal and Vascular Medicine, Amsterdam University Medical Centers, University of Amsterdam, Amsterdam, Netherlands
| | - Hannele Yki-Jarvinen
- Department of Medicine, Helsinki University Hospital, University of Helsinki, Helsinki, Finland; Minerva Foundation Institute for Medical Research, Helsinki, Finland
| | - Javier Crespo
- Gastroenterology and Hepatology Department, Valdecilla Health Research Institute, Marqués de Valdecilla University Hospital, Santander, Spain
| | | | | | - Mohammad Hadi Zafarmand
- Epidemiology and Data Science, Amsterdam Public Health, University of Amsterdam, Amsterdam, Netherlands
| | - Richard Torstenson
- Cardiovascular, Renal or Metabolism Regulatory Affairs, AstraZeneca, Mölndal, Sweden
| | - Kevin Duffin
- Lilly Research Laboratories, Eli Lilly, Indianapolis, IN, USA
| | - Carla Yunis
- Clinical Development and Operations, Pfizer, Lake Mary, FL, USA
| | | | - Mattias Ekstedt
- Department of Health, Medicine and Caring Sciences, Linköping University, Linköping, Sweden
| | - Guruprasad P Aithal
- Nottingham Digestive Diseases Centre, School of Medicine, National Institute for Health and Care Research Nottingham Biomedical Research Centre, Nottingham University Hospitals NHS Trust, The University of Nottingham, Nottingham, UK
| | - Jörn M Schattenberg
- Metabolic Liver Research Program, Department of Medicine, University Medical Center Mainz, Mainz, Germany
| | - Elisabetta Bugianesi
- Department of Medical Sciences, Division of Gastro-Hepatology, City of Health and Science of Turin, University of Turin, Turin, Italy
| | - Manuel Romero-Gomez
- Digestive Diseases, Virgen of Rocio University Hospital, Institute of Biomedicine of Seville, Department of Medicine, University of Seville, Seville, Spain
| | - Vlad Ratziu
- Public Assistance Hospital of Paris, Pitié Salpêtrière Hospital, Institute of Cardiometabolism and Nutrition, Sorbonne University, Paris, France
| | - Quentin M Anstee
- Translational and Clinical Research Institute, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne, UK; Newcastle National Institute for Health and Care Research Biomedical Research Centre, Newcastle upon Tyne Hospitals National Health Service Trust, Newcastle upon Tyne, UK.
| | - Patrick M Bossuyt
- Epidemiology and Data Science, Amsterdam Public Health, University of Amsterdam, Amsterdam, Netherlands
| |
Collapse
|
71
|
Shen Y, Cheng L, Xu M, Wang W, Wan Z, Xiong H, Guo W, Cai M, Xu F. SGLT2 inhibitor empagliflozin downregulates miRNA-34a-5p and targets GREM2 to inactivate hepatic stellate cells and ameliorate non-alcoholic fatty liver disease-associated fibrosis. Metabolism 2023:155657. [PMID: 37422021 DOI: 10.1016/j.metabol.2023.155657] [Citation(s) in RCA: 53] [Impact Index Per Article: 26.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/18/2023] [Revised: 06/30/2023] [Accepted: 07/05/2023] [Indexed: 07/10/2023]
Abstract
BACKGROUND AND RATIONALE Activation of hepatic stellate cells (HSCs), the central event of fibrosis, indicates the severe stage of non-alcoholic fatty liver disease (NAFLD). MicroRNAs (miRNAs) participate in this process. Treatment with a sodium-glucose cotransporter 2 inhibitor (SGLT2i) alleviates liver fibrosis in patients with type 2 diabetes and NAFLD; however, the role of SGLT2i in ameliorating liver fibrosis in NAFLD by regulating miRNAs remains unclear. APPROACH AND RESULTS We monitored the expression of NAFLD-associated miRNAs in the livers of two NAFLD models and observed high expression of miR-34a-5p. miR-34a-5p was highly expressed in mouse primary liver non-parenchymal cells and LX-2 HSCs, and this miRNA was positively correlated with alanine transaminase levels in NAFLD models. Overexpression of miR-34a-5p enhanced LX-2 activation, whereas its inhibition prevented HSCs activation by regulating the TGFβ signaling pathway. The SGLT2i empagliflozin significantly downregulated miR-34a-5p, inhibited the TGFβ signaling pathway, and ameliorated hepatic fibrosis in NAFLD models. Subsequently, GREM2 was identified as a direct target of miR-34a-5p through database prediction and a dual-luciferase reporter assay. In LX-2 HSCs, the miR-34a-5p mimic and inhibitor directly downregulated and upregulated GREM2, respectively. Overexpressing GREM2 inactivated the TGFβ pathway whereas GREM2 knockdown activated it. Additionally, empagliflozin upregulated Grem2 expression in NAFLD models. In methionine- and choline-deficient diet-fed ob/ob mice, a fibrosis model, empagliflozin downregulated miR-34a-5p and upregulated Grem2 to improve liver fibrosis. CONCLUSIONS Empagliflozin ameliorates NAFLD-associated fibrosis by downregulating miR-34a-5p and targeting GREM2 to inhibit the TGFβ pathway in HSCs.
Collapse
Affiliation(s)
- Yunfeng Shen
- Department of Endocrinology and Metabolism, the Second Affiliated Hospital of Nanchang University, Nanchang 330006, China; Branch of National Clinical Research Center for Metabolic Diseases, Nanchang 330006, China; Institute for the Study of Endocrinology and Metabolism in Jiangxi Province, Nanchang 330006, China
| | - Lidan Cheng
- Department of Endocrinology and Metabolism, the Second Affiliated Hospital of Nanchang University, Nanchang 330006, China; Guangdong Provincial Key Laboratory of Diabetology, Guangzhou 510630, China; Department of Endocrinology and Metabolism, Jiujiang University Affiliated Hospital, Jiujiang 330300, China
| | - Minxuan Xu
- Department of Endocrinology and Metabolism, the Second Affiliated Hospital of Nanchang University, Nanchang 330006, China; Guangdong Provincial Key Laboratory of Diabetology, Guangzhou 510630, China
| | - Wei Wang
- Department of Endocrinology and Metabolism, the Second Affiliated Hospital of Nanchang University, Nanchang 330006, China; Guangdong Provincial Key Laboratory of Diabetology, Guangzhou 510630, China; Department of Gastroenterology, the First Affiliated Hospital of Yangtze University, Jingzhou 434000, China
| | - Zhiping Wan
- Guangdong Provincial Key Laboratory of Diabetology, Guangzhou 510630, China; Department of Infectious Diseases, the Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou 510630, China
| | - Haixia Xiong
- Department of Endocrinology and Metabolism, the Second Affiliated Hospital of Nanchang University, Nanchang 330006, China; Branch of National Clinical Research Center for Metabolic Diseases, Nanchang 330006, China; Institute for the Study of Endocrinology and Metabolism in Jiangxi Province, Nanchang 330006, China
| | - Wanrong Guo
- Department of Endocrinology and Metabolism, the Third Affiliated Hospital of Sun Yat-sen University, Guangzhou 510630, China; Guangdong Provincial Key Laboratory of Diabetology, Guangzhou 510630, China
| | - Mengyin Cai
- Department of Endocrinology and Metabolism, the Third Affiliated Hospital of Sun Yat-sen University, Guangzhou 510630, China; Guangdong Provincial Key Laboratory of Diabetology, Guangzhou 510630, China.
| | - Fen Xu
- Department of Endocrinology and Metabolism, the Third Affiliated Hospital of Sun Yat-sen University, Guangzhou 510630, China; Guangdong Provincial Key Laboratory of Diabetology, Guangzhou 510630, China.
| |
Collapse
|
72
|
Pennisi G, Enea M, Falco V, Aithal GP, Palaniyappan N, Yilmaz Y, Boursier J, Cassinotto C, de Lédinghen V, Chan WK, Mahadeva S, Eddowes P, Newsome P, Karlas T, Wiegand J, Wong VWS, Schattenberg JM, Labenz C, Kim W, Lee MS, Lupsor-Platon M, Cobbold JFL, Fan JG, Shen F, Staufer K, Trauner M, Stauber R, Nakajima A, Yoneda M, Bugianesi E, Younes R, Gaia S, Zheng MH, Cammà C, Anstee QM, Mózes FE, Pavlides M, Petta S. Noninvasive assessment of liver disease severity in patients with nonalcoholic fatty liver disease (NAFLD) and type 2 diabetes. Hepatology 2023; 78:195-211. [PMID: 36924031 DOI: 10.1097/hep.0000000000000351] [Citation(s) in RCA: 26] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/25/2022] [Accepted: 01/04/2023] [Indexed: 03/18/2023]
Abstract
BACKGROUND AND AIMS We evaluated the diagnostic accuracy of simple, noninvasive tests (NITs) in NAFLD patients with type 2 diabetes (T2D). METHODS AND RESULTS This was an individual patient data meta-analysis of 1780 patients with biopsy-proven NAFLD and T2D. The index tests of interest were FIB-4, NAFLD Fibrosis Score (NFS), aspartate aminotransferase-to-platelet ratio index, liver stiffness measurement (LSM) by vibration-controlled transient elastography, and AGILE 3+. The target conditions were advanced fibrosis, NASH, and fibrotic NASH(NASH plus F2-F4 fibrosis). The diagnostic performance of noninvasive tests. individually or in sequential combination, was assessed by area under the receiver operating characteristic curve and by decision curve analysis. Comparison with 2278 NAFLD patients without T2D was also made. In NAFLD with T2D LSM and AGILE 3+ outperformed, both NFS and FIB-4 for advanced fibrosis (area under the receiver operating characteristic curve:LSM 0.82, AGILE 3+ 0.82, NFS 0.72, FIB-4 0.75, aspartate aminotransferase-to-platelet ratio index 0.68; p < 0.001 of LSM-based versus simple serum tests), with an uncertainty area of 12%-20%. The combination of serum-based with LSM-based tests for advanced fibrosis led to a reduction of 40%-60% in necessary LSM tests. Decision curve analysis showed that all scores had a modest net benefit for ruling out advanced fibrosis at the risk threshold of 5%-10% of missing advanced fibrosis. LSM and AGILE 3+ outperformed both NFS and FIB-4 for fibrotic NASH (area under the receiver operating characteristic curve:LSM 0.79, AGILE 3+ 0.77, NFS 0.71, FIB-4 0.71; p < 0.001 of LSM-based versus simple serum tests). All noninvasive scores were suboptimal for diagnosing NASH. CONCLUSIONS LSM and AGILE 3+ individually or in low availability settings in sequential combination after FIB-4 or NFS have a similar good diagnostic accuracy for advanced fibrosis and an acceptable diagnostic accuracy for fibrotic NASH in NAFLD patients with T2D.
Collapse
Affiliation(s)
- Grazia Pennisi
- Sezione di Gastroenterologia, PROMISE, University of Palermo, Italy
| | - Marco Enea
- Sezione di Gastroenterologia, PROMISE, University of Palermo, Italy
| | - Vincenzo Falco
- Department of Economics and Statistics, University of Palermo, Palermo, Italy
| | - Guruprasad P Aithal
- NIHR Nottingham Biomedical Research Centre (BRC), Nottingham University Hospitals NHS Trust and the University of Nottingham, Nottingham, UK
- Nottingham Digestive Diseases Centre, Translational Medical Sciences, School of Medicine, University of Nottingham, Nottingham, UK
| | - Naaventhan Palaniyappan
- NIHR Nottingham Biomedical Research Centre (BRC), Nottingham University Hospitals NHS Trust and the University of Nottingham, Nottingham, UK
- Nottingham Digestive Diseases Centre, Translational Medical Sciences, School of Medicine, University of Nottingham, Nottingham, UK
| | - Yusuf Yilmaz
- Department of Gastroenterology, School of Medicine, Recep Tayyip Erdogan University, Rize, Turkey
| | - Jerome Boursier
- Hepato-Gastroenterology Department, Angers University Hospital, Angers, France
- HIFIH Laboratory, UPRES EA3859, Angers University, Angers, France
| | - Christophe Cassinotto
- Department of Diagnostic and Interventional Radiology, Saint-Eloi Hospital, University Hospital of Montpellier, Montpellier, France
| | - Victor de Lédinghen
- Hepatology Unit, University Hospital Bordeaux and INSERM U-1053, Bordeaux University, Pessac, France
| | - Wah Kheong Chan
- Department of Medicine, Faculty of Medicine, University of Malaya, Malaysia
| | - Sanjiv Mahadeva
- Department of Medicine, Faculty of Medicine, University of Malaya, Malaysia
| | - Peter Eddowes
- NIHR Nottingham Biomedical Research Centre (BRC), Nottingham University Hospitals NHS Trust and the University of Nottingham, Nottingham, UK
- Nottingham Digestive Diseases Centre, Translational Medical Sciences, School of Medicine, University of Nottingham, Nottingham, UK
| | - Philip Newsome
- NIHR Nottingham Biomedical Research Centre (BRC), Nottingham University Hospitals NHS Trust and the University of Nottingham, Nottingham, UK
- Nottingham Digestive Diseases Centre, Translational Medical Sciences, School of Medicine, University of Nottingham, Nottingham, UK
| | - Thomas Karlas
- Department of Oncology, Gastroenterology, Hepatology, Pulmonology and Infectious Diseases, University Hospital Leipzig, Leipzig, Germany
| | - Johannes Wiegand
- Department of Oncology, Gastroenterology, Hepatology, Pulmonology and Infectious Diseases, University Hospital Leipzig, Leipzig, Germany
| | - Vincent Wai-Sun Wong
- Department of Medicine and Therapeutics, Faculty of Medicine, The Chinese University of Hong Kong
| | - Jörn M Schattenberg
- Department of Internal Medicine I, University Medical Center of the Johannes Gutenberg-University, Mainz, Germany
- Cirrhosis Center Mainz (CCM), University Medical Center of the Johannes Gutenberg-University, Mainz, Germany
| | - Christian Labenz
- Department of Internal Medicine I, University Medical Center of the Johannes Gutenberg-University, Mainz, Germany
- Cirrhosis Center Mainz (CCM), University Medical Center of the Johannes Gutenberg-University, Mainz, Germany
| | - Won Kim
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Seoul National University College of Medicine, Seoul Metropolitan Government Boramae Medical Center, Seoul, Korea
| | - Myoung Seok Lee
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Seoul National University College of Medicine, Seoul Metropolitan Government Boramae Medical Center, Seoul, Korea
| | - Monica Lupsor-Platon
- Department of Medical Imaging, Iuliu Hatieganu, University of Medicine and Pharmacy, Regional Institute of Gastroenterology and Hepatology "Prof. Dr. Octavian Fodor", Cluj-Napoca, Romania
| | | | - Jian-Gao Fan
- Department of Gastroenterology, Xinhua Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Feng Shen
- Department of Gastroenterology, Xinhua Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Katharina Staufer
- Division of Gastroenterology & Hepatology, Department of Internal Medicine III, Medical University of Vienna, Austria
- Division of Transplantation, Department of General Surgery, Medical University of Vienna, Austria
| | - Michael Trauner
- Division of Gastroenterology & Hepatology, Department of Internal Medicine III, Medical University of Vienna, Austria
| | - Rudolf Stauber
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Medical University of Graz, Austria
| | - Atsushi Nakajima
- Department of Gastroenterology and Hepatology, Yokohama City University School of Medicine, Yokohama, Japan
| | - Masato Yoneda
- Department of Gastroenterology and Hepatology, Yokohama City University School of Medicine, Yokohama, Japan
| | - Elisabetta Bugianesi
- Division of Gastroenterology and Hepatology, Department of Medical Sciences, University of Turin, Italy
| | - Ramy Younes
- Division of Gastroenterology and Hepatology, Department of Medical Sciences, University of Turin, Italy
| | - Silvia Gaia
- Division of Gastroenterology and Hepatology, Department of Medical Sciences, University of Turin, Italy
| | - Ming-Hua Zheng
- Department of Hepatology, MAFLD Research Center, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
- Key Laboratory of Diagnosis and Treatment for The Development of Chronic Liver Disease in Zhejiang Province, Wenzhou, China
| | - Calogero Cammà
- Sezione di Gastroenterologia, PROMISE, University of Palermo, Italy
- Department of Economics and Statistics, University of Palermo, Palermo, Italy
| | - Quentin M Anstee
- Translational and Clinical Research Institute, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne, Tyne and Wear, UK
- Newcastle NIHR Biomedical Research Center, Newcastle upon Tyne Hospitals NHS Trust, Newcastle upon Tyne, Tyne and Wear, UK
| | - Ferenc E Mózes
- Radcliffe Department of Medical Sciences, Cardiovascular Medicine, University of Oxford, Oxford, UK
| | - Michael Pavlides
- Translational Gastroenterology Unit, University of Oxford, Oxford, UK
- Radcliffe Department of Medical Sciences, Cardiovascular Medicine, University of Oxford, Oxford, UK
| | - Salvatore Petta
- Sezione di Gastroenterologia, PROMISE, University of Palermo, Italy
- Department of Economics and Statistics, University of Palermo, Palermo, Italy
| |
Collapse
|
73
|
Armandi A, Michel M, Gjini K, Emrich T, Bugianesi E, Schattenberg JM. Emerging concepts in the detection of liver fibrosis in non-alcoholic fatty liver disease. Expert Rev Mol Diagn 2023; 23:771-782. [PMID: 37505901 DOI: 10.1080/14737159.2023.2242779] [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: 04/06/2023] [Revised: 06/24/2023] [Accepted: 07/27/2023] [Indexed: 07/30/2023]
Abstract
INTRODUCTION The non-invasive identification of liver fibrosis related to Non-Alcoholic Fatty Liver Disease is crucial for risk-stratification of patients. Currently, the reference standard to stage hepatic fibrosis relies on liver biopsy, but multiple approaches are developed to allow for non-invasive diagnosis and risk stratification. Non-invasive tests, including blood-based scores and vibration-controlled transient elastography, have been widely validated and represent a good surrogate for risk stratification according to recent European and American guidelines. AREAS COVERED Novel approaches are based on 'liquid' biomarkers of liver fibrogenesis, including collagen-derived markers (PRO-C3 or PRO-C6), or 'multi-omics' technologies (e.g. proteomic-based molecules or miRNA testing), bearing the advantage of tailoring the intrahepatic disease activity. Alternative approaches are based on 'dry' biomarkers, including magnetic resonance-based tools (including proton density fat fraction, magnetic resonance elastography, or corrected T1), which reach similar accuracy of liver histology and will potentially help identify the best candidates for pharmacological treatment of fibrosing non-alcoholic steatohepatitis. EXPERT OPINION In the near future, the sequential use of non-invasive tests, as well as the complimentary use of liquid and dry biomarkers according to the clinical need (diagnosis, risk stratification, and prognosis, or treatment response) will guide and improve the management of this liver disease.
Collapse
Affiliation(s)
- Angelo Armandi
- Division of Gastroenterology and Hepatology, Department of Medical Sciences, University of Turin, Corso Dogliotti 14, 10126 Turin, Italy
- Metabolic Liver Research Program, I. Department of Medicine, University Medical Centre Mainz, Langenbeckstrasse 1, 55131 Mainz, Germany
| | - Maurice Michel
- Metabolic Liver Research Program, I. Department of Medicine, University Medical Centre Mainz, Langenbeckstrasse 1, 55131 Mainz, Germany
- I. Department of Medicine, University Medical Centre Mainz, Langenbeckstrasse 1, 55131 Mainz, Germany
| | - Kamela Gjini
- Division of Gastroenterology and Hepatology, Department of Medical Sciences, University of Turin, Corso Dogliotti 14, 10126 Turin, Italy
| | - Tilman Emrich
- Department of Radiology, University Medical Centre Mainz, Langenbeckstrasse 1, 55131 Mainz, Germany
| | - Elisabetta Bugianesi
- Division of Gastroenterology and Hepatology, Department of Medical Sciences, University of Turin, Corso Dogliotti 14, 10126 Turin, Italy
| | - Jorn M Schattenberg
- Metabolic Liver Research Program, I. Department of Medicine, University Medical Centre Mainz, Langenbeckstrasse 1, 55131 Mainz, Germany
- I. Department of Medicine, University Medical Centre Mainz, Langenbeckstrasse 1, 55131 Mainz, Germany
| |
Collapse
|
74
|
Tincopa MA, Loomba R. Non-invasive diagnosis and monitoring of non-alcoholic fatty liver disease and non-alcoholic steatohepatitis. Lancet Gastroenterol Hepatol 2023; 8:660-670. [PMID: 37060912 DOI: 10.1016/s2468-1253(23)00066-3] [Citation(s) in RCA: 75] [Impact Index Per Article: 37.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/12/2023] [Revised: 02/19/2023] [Accepted: 02/20/2023] [Indexed: 04/17/2023]
Abstract
Non-alcoholic fatty liver disease (NAFLD) is a highly prevalent form of chronic liver disease that poses challenges in diagnosis and risk stratification. Non-alcoholic steatohepatitis (NASH), the more progressive form of NAFLD, is particularly challenging to diagnose in the absence of histology. Liver biopsy is infrequently performed due to its invasive nature, potential for sampling error, and lack of inter-rater reliability. Non-invasive tests that can accurately identify patients with at-risk NASH (ie, individuals with biopsy-proven NASH with NAFLD activity score [NAS] ≥4 and fibrosis stage ≥2) are key tools to identify candidates for pharmacologic therapy in registrational trials for the treatment of NASH-related fibrosis. With emerging pharmacotherapy, non-invasive tests are required to track treatment response. Lastly, there is an unmet need for non-invasive tests to assess risk for clinical outcomes including progression to cirrhosis, hepatic decompensation, liver-related mortality, and overall mortality. In this Review we examine advances in non-invasive tests to diagnose and monitor NAFLD and NASH.
Collapse
Affiliation(s)
- Monica A Tincopa
- NAFLD Research Center, Division of Gastroenterology and Hepatology, University of California at San Diego, La Jolla, CA, USA
| | - Rohit Loomba
- NAFLD Research Center, Division of Gastroenterology and Hepatology, University of California at San Diego, La Jolla, CA, USA; School of Public Health, University of California at San Diego, La Jolla, CA, USA.
| |
Collapse
|
75
|
Lee J, Westphal M, Vali Y, Boursier J, Petta S, Ostroff R, Alexander L, Chen Y, Fournier C, Geier A, Francque S, Wonders K, Tiniakos D, Bedossa P, Allison M, Papatheodoridis G, Cortez-Pinto H, Pais R, Dufour JF, Leeming DJ, Harrison S, Cobbold J, Holleboom AG, Yki-Järvinen H, Crespo J, Ekstedt M, Aithal GP, Bugianesi E, Romero-Gomez M, Torstenson R, Karsdal M, Yunis C, Schattenberg JM, Schuppan D, Ratziu V, Brass C, Duffin K, Zwinderman K, Pavlides M, Anstee QM, Bossuyt PM. Machine learning algorithm improves the detection of NASH (NAS-based) and at-risk NASH: A development and validation study. Hepatology 2023; 78:258-271. [PMID: 36994719 DOI: 10.1097/hep.0000000000000364] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/16/2022] [Accepted: 12/22/2022] [Indexed: 03/31/2023]
Abstract
BACKGROUND AND AIMS Detecting NASH remains challenging, while at-risk NASH (steatohepatitis and F≥ 2) tends to progress and is of interest for drug development and clinical application. We developed prediction models by supervised machine learning techniques, with clinical data and biomarkers to stage and grade patients with NAFLD. APPROACH AND RESULTS Learning data were collected in the Liver Investigation: Testing Marker Utility in Steatohepatitis metacohort (966 biopsy-proven NAFLD adults), staged and graded according to NASH CRN. Conditions of interest were the clinical trial definition of NASH (NAS ≥ 4;53%), at-risk NASH (NASH with F ≥ 2;35%), significant (F ≥ 2;47%), and advanced fibrosis (F ≥ 3;28%). Thirty-five predictors were included. Missing data were handled by multiple imputations. Data were randomly split into training/validation (75/25) sets. A gradient boosting machine was applied to develop 2 models for each condition: clinical versus extended (clinical and biomarkers). Two variants of the NASH and at-risk NASH models were constructed: direct and composite models.Clinical gradient boosting machine models for steatosis/inflammation/ballooning had AUCs of 0.94/0.79/0.72. There were no improvements when biomarkers were included. The direct NASH model produced AUCs (clinical/extended) of 0.61/0.65. The composite NASH model performed significantly better (0.71) for both variants. The composite at-risk NASH model had an AUC of 0.83 (clinical and extended), an improvement over the direct model. Significant fibrosis models had AUCs (clinical/extended) of 0.76/0.78. The extended advanced fibrosis model (0.86) performed significantly better than the clinical version (0.82). CONCLUSIONS Detection of NASH and at-risk NASH can be improved by constructing independent machine learning models for each component, using only clinical predictors. Adding biomarkers only improved the accuracy of fibrosis.
Collapse
Affiliation(s)
- Jenny Lee
- Department of Epidemiology and Data Science, Amsterdam UMC, Amsterdam, the Netherlands
| | - Max Westphal
- Fraunhofer Institute for Digital Medicine MEVIS, Bremen, Germany
| | - Yasaman Vali
- Department of Epidemiology and Data Science, Amsterdam UMC, Amsterdam, the Netherlands
| | - Jerome Boursier
- Department of Hepatology, Angers University Hospital, Angers, France
| | - Salvatorre Petta
- Section of Gastroenterology and Hepatology, Promozione della Salute, Materno-Infantile, di Medicina Interna e Specialistica di Eccellenza, Department, University of Palermo, Palermo, Italy
| | | | | | - Yu Chen
- Lilly Research Laboratories, Eli Lilly and Company Ltd (LLY), Indianapolis, Indiana, USA
| | | | - Andreas Geier
- Division of Hepatology, Department of Medicine II, Wurzburg University Hospital, Wurzburg, Germany
| | - Sven Francque
- Department of Gastroenterology Hepatology, Antwerp University Hospital, and Laboratory of Experimental Medicine and Paediatrics, University of Antwerp, Belgium
| | - Kristy Wonders
- Translational and Clinical Research Institute, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne, UK
| | - Dina Tiniakos
- Translational and Clinical Research Institute, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne, UK
- Department of Pathology, Aretaieion Hospital, national and Kapodistrian University of Athens, Athens, Greece
| | - Pierre Bedossa
- Translational and Clinical Research Institute, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne, UK
| | - Mike Allison
- Liver Unit, Department of Medicine, Cambridge NIHR Biomedical Research Centre, Cambridge University NHS Foundation Trust, CB2 0QQ, Cambridge, UK
| | - Georgios Papatheodoridis
- Gastroenterology Department, National and Kapodistrian University of Athens, General Hospital of Athens "Laiko", Athens, Greece
| | - Helena Cortez-Pinto
- Clínica Universitária de Gastrenterologia, Faculdade de Medicina, Universidade de Lisboa, Portugal
| | - Raluca Pais
- Assistance Publique-Hôpitaux de Paris, hôpital Pitié Salpêtrière, Sorbonne University, ICAN (Institute of Cardiometabolism and Nutrition), Paris, France
| | - Jean-Francois Dufour
- Hepatology, Department of Biomedical Research, University of Bern, Bern, Switzerland
| | | | - Stephen Harrison
- Department of Gastroenterology and Hepatology, Oxford NIHR Biomedical Research Centre, John Radcliffe Hospital, Oxford, UK
| | - Jeremy Cobbold
- Department of Gastroenterology and Hepatology, Oxford NIHR Biomedical Research Centre, John Radcliffe Hospital, Oxford, UK
| | - Adriaan G Holleboom
- Department of Internal and Vascular Medicine, Amsterdam University Medical Centres, location AMC, Amsterdam, the Netherlands
| | - Hannele Yki-Järvinen
- Department of Medicine, University of Helsinki and Helsinki University Hospital, Finland; Minerva Foundation Institute for Medical Research, Helsinki, Finland
| | - Javier Crespo
- Department of Gastroenterology and Hepatology, University Hospital Marques de Valdecilla. Research Institute Valdecilla-IDIVAL, Santander, Spain
| | - Mattias Ekstedt
- Department of Health, Medicine and Caring Sciences, Linköping University, Linköping, Sweden
| | - Guruprasad P Aithal
- Nottingham Digestive Diseases Centre, School of Medicine, NIHR Nottingham Biomedical Research Centre, Nottingham University Hospitals NHS Trust and The University of Nottingham, Nottingham, UK
| | - Elisabetta Bugianesi
- Department of Medical Sciences, Division of Gastro-Hepatology, A.O. Città della Salute e della Scienza di Torino, University of Turin, Turin, Italy
| | - Manuel Romero-Gomez
- UCM Digestive Diseases, ciberehd, Virgen del Rocio University Hospital. Institute of Biomedicine of Seville (CSIC/HUVR/US), Department of Medicine, University of Seville, Seville, Spain
| | - Richard Torstenson
- Cardiovascular, Renal and Metabolism Regulatory Affairs, AstraZeneca, Mölndal, Sweden
| | | | - Carla Yunis
- Internal Medicine and Hospital, Global Product Development, Pfizer, Inc, New York, New York, USA
| | - Jörn M Schattenberg
- Metabolic Liver Research Program, I. Department of Medicine, University Medical Center Mainz, Mainz, Germany
| | - Detlef Schuppan
- Institute of Translational Immunology and Research Center for Immune Therapy, University Medical Center Mainz, Mainz, Germany
- Division of Gastroenterology, Beth Israel Medical Center, Harvard Medical School, Boston, Massachusetts, USA
| | - Vlad Ratziu
- Assistance Publique-Hôpitaux de Paris, hôpital Pitié Salpêtrière, Sorbonne University, ICAN (Institute of Cardiometabolism and Nutrition), Paris, France
| | - Clifford Brass
- Novartis Pharmaceuticals Corporation, East Hanover, New Jersey
| | - Kevin Duffin
- Lilly Research Laboratories, Eli Lilly and Company Ltd (LLY), Indianapolis, Indiana, USA
| | - Koos Zwinderman
- Department of Epidemiology and Data Science, Amsterdam UMC, Amsterdam, the Netherlands
| | | | - Quentin M Anstee
- Department of Gastroenterology Hepatology, Antwerp University Hospital, and Laboratory of Experimental Medicine and Paediatrics, University of Antwerp, Belgium
- Newcastle NIHR Biomedical Research Centre, Newcastle upon Tyne Hospitals NHS Trust, Newcastle upon Tyne, UK
| | - Patrick M Bossuyt
- Department of Epidemiology and Data Science, Amsterdam UMC, Amsterdam, the Netherlands
| |
Collapse
|
76
|
Zeng M, Chen L, Li Y, Mi Y, Xu L. Problems and Challenges Associated with Renaming Non-alcoholic Fatty Liver Disease to Metabolic Associated Fatty Liver Disease. Medicine (Baltimore) 2023; 3:105-113. [PMCID: PMC10368226 DOI: 10.1097/id9.0000000000000085] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/09/2022] [Indexed: 10/08/2024] Open
Abstract
Non-alcoholic fatty liver disease (NAFLD) has become the world’s largest chronic liver disease in the 21st century, affecting 20%–30% of the world’s population. As the epidemiology, etiology, and pathogenesis of NAFLD have been studied in-depth, it has been gradually recognized that most patients with NAFLD have one or more combined metabolic abnormalities known as metabolic syndrome. In 2020, the international expert group changed the name of NAFLD to metabolic-associated fatty liver disease (MAFLD) and proposed new diagnostic criteria for MAFLD and MAFLD-related liver cirrhosis, as well as the conceptual framework of other cause-related fatty liver diseases to avoid diagnosis based on the exclusion of other causes and better reflect its pathogenesis. However, there are still many ambiguities in the term, and changing the name does not address the unmet key needs in the field. The change from NAFLD to MAFLD was not just a change of definition. The problems and challenges are summarized as follows: epidemiology, children, rationality of “metabolism,” diagnostic criteria, double/multiple causes, drug discovery, clinical trials, and awareness raising. Metabolic-associated fatty liver disease has complex disease characteristics, and there are still some problems that need to be solved.
Collapse
Affiliation(s)
- Minghui Zeng
- Clinical School of the Second People’s Hospital, Tianjin Medical University, Tianjin 300192, China
- Department of Hepatology, Tianjin Second People’s Hospital, Tianjin 300192, China
| | - Lin Chen
- Clinical School of the Second People’s Hospital, Tianjin Medical University, Tianjin 300192, China
- Department of Hepatology, Tianjin Second People’s Hospital, Tianjin 300192, China
| | - Yuqin Li
- Clinical School of the Second People’s Hospital, Tianjin Medical University, Tianjin 300192, China
- Department of Hepatology, Tianjin Second People’s Hospital, Tianjin 300192, China
| | - Yuqiang Mi
- Department of Hepatology, Tianjin Second People’s Hospital, Tianjin 300192, China
- Tianjin Research Institute of Liver Diseases, Tianjin 300192, China
| | - Liang Xu
- Department of Hepatology, Tianjin Second People’s Hospital, Tianjin 300192, China
- Tianjin Research Institute of Liver Diseases, Tianjin 300192, China
| |
Collapse
|
77
|
Tavaglione F, Jamialahmadi O, De Vincentis A, Qadri S, Mowlaei ME, Mancina RM, Ciociola E, Carotti S, Perrone G, Bruni V, Gallo IF, Tuccinardi D, Bianco C, Prati D, Manfrini S, Pozzilli P, Picardi A, Caricato M, Yki-Järvinen H, Valenti L, Vespasiani-Gentilucci U, Romeo S. Development and Validation of a Score for Fibrotic Nonalcoholic Steatohepatitis. Clin Gastroenterol Hepatol 2023; 21:1523-1532.e1. [PMID: 35421583 DOI: 10.1016/j.cgh.2022.03.044] [Citation(s) in RCA: 51] [Impact Index Per Article: 25.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/15/2022] [Revised: 03/25/2022] [Accepted: 03/28/2022] [Indexed: 02/07/2023]
Abstract
BACKGROUND & AIMS Noninvasive assessment of histological features of nonalcoholic fatty liver disease (NAFLD) has been an intensive research area over the last decade. Herein, we aimed to develop a simple noninvasive score using routine laboratory tests to identify, among individuals at high risk for NAFLD, those with fibrotic nonalcoholic steatohepatitis (NASH) defined as NASH, NAFLD activity score ≥4, and fibrosis stage ≥2. METHODS The derivation cohort included 264 morbidly obese individuals undergoing intraoperative liver biopsy in Rome, Italy. The best predictive model was developed and internally validated using a bootstrapping stepwise logistic regression analysis (2000 bootstrap samples). Performance was estimated by the area under the receiver operating characteristic curve (AUROC). External validation was assessed in 3 independent European cohorts (Finland, n = 370; Italy, n = 947; England, n = 5368) of individuals at high risk for NAFLD. RESULTS The final predictive model, designated as Fibrotic NASH Index (FNI), combined aspartate aminotransferase, high-density lipoprotein cholesterol, and hemoglobin A1c. The performance of FNI for fibrotic NASH was satisfactory in both derivation and external validation cohorts (AUROC = 0.78 and AUROC = 0.80-0.95, respectively). In the derivation cohort, rule-out and rule-in cutoffs were 0.10 for sensitivity ≥0.89 (negative predictive value, 0.93) and 0.33 for specificity ≥0.90 (positive predictive value, 0.57), respectively. In the external validation cohorts, sensitivity ranged from 0.87 to 1 (negative predictive value, 0.99-1) and specificity from 0.73 to 0.94 (positive predictive value, 0.12-0.49) for rule-out and rule-in cutoff, respectively. CONCLUSION FNI is an accurate, simple, and affordable noninvasive score which can be used to screen for fibrotic NASH in individuals with dysmetabolism in primary health care.
Collapse
Affiliation(s)
- Federica Tavaglione
- Clinical Medicine and Hepatology Unit, Department of Internal Medicine and Geriatrics, Campus Bio-Medico University, Rome, Italy; Department of Molecular and Clinical Medicine, Institute of Medicine, Sahlgrenska Academy, Wallenberg Laboratory, University of Gothenburg, Gothenburg, Sweden.
| | - Oveis Jamialahmadi
- Department of Molecular and Clinical Medicine, Institute of Medicine, Sahlgrenska Academy, Wallenberg Laboratory, University of Gothenburg, Gothenburg, Sweden
| | - Antonio De Vincentis
- Internal Medicine Unit, Department of Internal Medicine and Geriatrics, Campus Bio-Medico University, Rome, Italy; Clinical Lecturer of Internal Medicine, Saint Camillus International University of Health and Medical Sciences, Rome, Italy
| | - Sami Qadri
- Department of Medicine, University of Helsinki and Helsinki University Hospital, Helsinki, Finland; Minerva Foundation Institute for Medical Research, Helsinki, Finland
| | - Mohammad Erfan Mowlaei
- Department of Computer & Information Sciences, College of Science and Technology, Temple University, Philadelphia, Pennsylvania
| | - Rosellina Margherita Mancina
- Department of Molecular and Clinical Medicine, Institute of Medicine, Sahlgrenska Academy, Wallenberg Laboratory, University of Gothenburg, Gothenburg, Sweden
| | - Ester Ciociola
- Department of Molecular and Clinical Medicine, Institute of Medicine, Sahlgrenska Academy, Wallenberg Laboratory, University of Gothenburg, Gothenburg, Sweden
| | - Simone Carotti
- Research Unit of Microscopic and Ultrastructural Anatomy, Department of Medicine, Campus Bio-Medico University, Rome, Italy; Predictive Molecular Diagnostic Unit, Department of Pathology, Campus Bio-Medico University Hospital, Rome, Italy
| | - Giuseppe Perrone
- Predictive Molecular Diagnostic Unit, Department of Pathology, Campus Bio-Medico University Hospital, Rome, Italy; Research Unit of Pathology, Campus Bio-Medico University, Rome, Italy
| | - Vincenzo Bruni
- Bariatric Surgery Unit, Campus Bio-Medico University, Rome, Italy
| | | | - Dario Tuccinardi
- Department of Endocrinology and Diabetes, University Campus Bio-Medico, Rome, Italy
| | - Cristiana Bianco
- Translational Medicine, Department of Transfusion Medicine and Hematology, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milano, Italy
| | - Daniele Prati
- Translational Medicine, Department of Transfusion Medicine and Hematology, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milano, Italy
| | - Silvia Manfrini
- Department of Endocrinology and Diabetes, University Campus Bio-Medico, Rome, Italy
| | - Paolo Pozzilli
- Department of Endocrinology and Diabetes, University Campus Bio-Medico, Rome, Italy
| | - Antonio Picardi
- Clinical Medicine and Hepatology Unit, Department of Internal Medicine and Geriatrics, Campus Bio-Medico University, Rome, Italy
| | - Marco Caricato
- Unit of Colon and Rectal Surgery, Department of General Surgery, Campus Bio-Medico University, Rome, Italy
| | - Hannele Yki-Järvinen
- Department of Medicine, University of Helsinki and Helsinki University Hospital, Helsinki, Finland; Minerva Foundation Institute for Medical Research, Helsinki, Finland
| | - Luca Valenti
- Translational Medicine, Department of Transfusion Medicine and Hematology, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milano, Italy; Department of Pathophysiology and Transplantation, Università degli Studi di Milano, Milano, Italy
| | - Umberto Vespasiani-Gentilucci
- Clinical Medicine and Hepatology Unit, Department of Internal Medicine and Geriatrics, Campus Bio-Medico University, Rome, Italy.
| | - Stefano Romeo
- Department of Molecular and Clinical Medicine, Institute of Medicine, Sahlgrenska Academy, Wallenberg Laboratory, University of Gothenburg, Gothenburg, Sweden; Department of Cardiology, Sahlgrenska University Hospital, Gothenburg, Sweden; Clinical Nutrition Unit, Department of Medical and Surgical Sciences, University Magna Graecia, Catanzaro, Italy.
| |
Collapse
|
78
|
Henry L, Eberly KE, Shah D, Kumar A, Younossi ZM. Noninvasive Tests Used in Risk Stratification of Patients with Nonalcoholic Fatty Liver Disease. Clin Liver Dis 2023; 27:373-395. [PMID: 37024214 DOI: 10.1016/j.cld.2023.01.022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/08/2023]
Abstract
As the prevalence of obesity and type 2 diabetes increases around the world, the prevalence of nonalcoholic fatty liver disease (NAFLD) has grown proportionately. Although most patients with NAFLD do not experience progressive liver disease, about 15% to 20% of those with nonalcoholic steatohepatitis can and do progress. Because liver biopsy's role in NAFLD has become increasingly limited, efforts have been undertaken to develop non-invasive tests (NITs) to help identify patients at high risk of progression. The following article discusses the NITs that are available to determine the presence of NAFLD as well as high-risk NAFLD.
Collapse
Affiliation(s)
- Linda Henry
- Inova Medicine, Inova Health System, 3300 Gallows Road, Falls Church, VA 22042, USA; Liver and Obesity Research Program, Inova Health System, 3300 Gallows Road, Falls Church, VA 22042, USA; Department of Medicine, Center for Liver Diseases, Inova Fairfax Medical Campus, 3300 Gallows Road, Falls Church, VA 22042, USA; Center for Outcomes Research in Liver Diseases, 2411 I Street, Northwest Washington, DC 20037, USA
| | - Katherine Elizabeth Eberly
- Inova Medicine, Inova Health System, 3300 Gallows Road, Falls Church, VA 22042, USA; Department of Medicine, Center for Liver Diseases, Inova Fairfax Medical Campus, 3300 Gallows Road, Falls Church, VA 22042, USA
| | - Dipam Shah
- Inova Medicine, Inova Health System, 3300 Gallows Road, Falls Church, VA 22042, USA; Department of Medicine, Center for Liver Diseases, Inova Fairfax Medical Campus, 3300 Gallows Road, Falls Church, VA 22042, USA
| | - Ameeta Kumar
- Inova Medicine, Inova Health System, 3300 Gallows Road, Falls Church, VA 22042, USA; Department of Medicine, Center for Liver Diseases, Inova Fairfax Medical Campus, 3300 Gallows Road, Falls Church, VA 22042, USA
| | - Zobair M Younossi
- Inova Medicine, Inova Health System, 3300 Gallows Road, Falls Church, VA 22042, USA; Liver and Obesity Research Program, Inova Health System, 3300 Gallows Road, Falls Church, VA 22042, USA; Department of Medicine, Center for Liver Diseases, Inova Fairfax Medical Campus, 3300 Gallows Road, Falls Church, VA 22042, USA.
| |
Collapse
|
79
|
Golabi P, Shah D, Younossi ZM. How to Identify Advanced Nonalcoholic Fatty Liver Disease in the Primary Care Setting. Semin Liver Dis 2023; 43:142-148. [PMID: 37414024 DOI: 10.1055/s-0043-1770984] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 07/08/2023]
Abstract
Nonalcoholic fatty liver disease (NAFLD) affects 30 to 40% of the population globally and is increasingly considered the most common liver disease. Patients with type 2 diabetes, obesity, and cardiovascular diseases are at especially increased risk for NAFLD. Although most patients with NAFLD do not have progressive liver disease, some patients progress to cirrhosis, liver cancer, and liver mortality. Given the sheer number of patients with NAFLD, the burden of disease is enormous. Despite this large and increasing burden, identification of NAFLD patients at risk for progressive liver disease in the primary care and diabetology practice settings remains highly suboptimal. In this review, our aim is to summarize a stepwise approach to risk stratify patients with NAFLD which should help practitioners in their management of patients with NAFLD.
Collapse
Affiliation(s)
- Pegah Golabi
- Betty and Guy Beatty Center for Integrated Research, Inova Health System, Falls Church, Virginia
- Department of Medicine, Center for Liver Disease, Inova Fairfax Medical Campus, Falls Church, Virginia
- Inova Medicine, Inova Health System, Falls Church, Virginia
| | - Dipam Shah
- Department of Medicine, Center for Liver Disease, Inova Fairfax Medical Campus, Falls Church, Virginia
| | - Zobair M Younossi
- Betty and Guy Beatty Center for Integrated Research, Inova Health System, Falls Church, Virginia
- Department of Medicine, Center for Liver Disease, Inova Fairfax Medical Campus, Falls Church, Virginia
- Inova Medicine, Inova Health System, Falls Church, Virginia
| |
Collapse
|
80
|
Rinella ME, Neuschwander-Tetri BA, Siddiqui MS, Abdelmalek MF, Caldwell S, Barb D, Kleiner DE, Loomba R. AASLD Practice Guidance on the clinical assessment and management of nonalcoholic fatty liver disease. Hepatology 2023; 77:1797-1835. [PMID: 36727674 PMCID: PMC10735173 DOI: 10.1097/hep.0000000000000323] [Citation(s) in RCA: 1047] [Impact Index Per Article: 523.5] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/18/2023] [Accepted: 01/18/2023] [Indexed: 02/03/2023]
Affiliation(s)
- Mary E. Rinella
- University of Chicago Pritzker School of Medicine, Chicago, Illinois, USA
| | | | | | | | - Stephen Caldwell
- School of Medicine, University of Virginia, Charlottesville, Virginia, USA
| | - Diana Barb
- University of Florida College of Medicine, Gainesville, Florida, USA
| | | | - Rohit Loomba
- University of California, San Diego, San Diego, California, USA
| |
Collapse
|
81
|
Li QY, Gong T, Huang YK, Kang L, Warner CA, Xie H, Chen LM, Duan XQ. Role of noncoding RNAs in liver fibrosis. World J Gastroenterol 2023; 29:1446-1459. [PMID: 36998425 PMCID: PMC10044853 DOI: 10.3748/wjg.v29.i9.1446] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/04/2022] [Revised: 10/27/2022] [Accepted: 02/27/2023] [Indexed: 03/07/2023] Open
Abstract
Liver fibrosis is a wound-healing response following chronic liver injury caused by hepatitis virus infection, obesity, or excessive alcohol. It is a dynamic and reversible process characterized by the activation of hepatic stellate cells and excess accumulation of extracellular matrix. Advanced fibrosis could lead to cirrhosis and even liver cancer, which has become a significant health burden worldwide. Many studies have revealed that noncoding RNAs (ncRNAs), including microRNAs, long noncoding RNAs and circular RNAs, are involved in the pathogenesis and development of liver fibrosis by regulating signaling pathways including transforming growth factor-β pathway, phosphatidylinositol 3-kinase/protein kinase B pathway, and Wnt/β-catenin pathway. NcRNAs in serum or exosomes have been reported to tentatively applied in the diagnosis and staging of liver fibrosis and combined with elastography to improve the accuracy of diagnosis. NcRNAs mimics, ncRNAs in mesenchymal stem cell-derived exosomes, and lipid nanoparticles-encapsulated ncRNAs have become promising therapeutic approaches for the treatment of liver fibrosis. In this review, we update the latest knowledge on ncRNAs in the pathogenesis and progression of liver fibrosis, and discuss the potentials and challenges to use these ncRNAs for diagnosis, staging and treatment of liver fibrosis. All these will help us to develop a comprehensive understanding of the role of ncRNAs in liver fibrosis.
Collapse
Affiliation(s)
- Qing-Yuan Li
- Department of Clinical Medicine, North Sichuan Medical College, Nanchong 637000, Sichuan Province, China
| | - Tao Gong
- Department of Clinical Medicine, North Sichuan Medical College, Nanchong 637000, Sichuan Province, China
| | - Yi-Ke Huang
- Center for Transfusion-transmitted Infectious Diseases, Institute of Blood Transfusion, Chinese Academy of Medical Sciences and Peking Union Medical College, Chengdu 610052, Sichuan Province, China
| | - Lan Kang
- Center for Transfusion-transmitted Infectious Diseases, Institute of Blood Transfusion, Chinese Academy of Medical Sciences and Peking Union Medical College, Chengdu 610052, Sichuan Province, China
| | - Charlotte A Warner
- Liver Center and Gastrointestinal Division, Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, United States
| | - He Xie
- Department of Clinical Laboratory, The Hospital of Xidian Group, Xi’an 710077, Shaanxi Province, China
| | - Li-Min Chen
- Center for Transfusion-transmitted Infectious Diseases, Institute of Blood Transfusion, Chinese Academy of Medical Sciences and Peking Union Medical College, Chengdu 610052, Sichuan Province, China
- Department of Clinical Laboratory, The Hospital of Xidian Group, Xi’an 710077, Shaanxi Province, China
| | - Xiao-Qiong Duan
- Center for Transfusion-transmitted Infectious Diseases, Institute of Blood Transfusion, Chinese Academy of Medical Sciences and Peking Union Medical College, Chengdu 610052, Sichuan Province, China
| |
Collapse
|
82
|
Harrison SA, Allen AM, Dubourg J, Noureddin M, Alkhouri N. Challenges and opportunities in NASH drug development. Nat Med 2023; 29:562-573. [PMID: 36894650 DOI: 10.1038/s41591-023-02242-6] [Citation(s) in RCA: 175] [Impact Index Per Article: 87.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2022] [Accepted: 12/20/2022] [Indexed: 03/11/2023]
Abstract
Nonalcoholic fatty liver disease (NAFLD) and its more severe form, nonalcoholic steatohepatitis (NASH), represent a growing worldwide epidemic and a high unmet medical need, as no licensed drugs have been approved thus far. Currently, histopathological assessment of liver biopsies is mandatory as a primary endpoint for conditional drug approval. This requirement represents one of the main challenges in the field, as there is substantial variability in this invasive histopathological assessment, which leads to dramatically high screen-failure rates in clinical trials. Over the past decades, several non-invasive tests have been developed to correlate with liver histology and, eventually, outcomes to assess disease severity and longitudinal changes non-invasively. However, further data are needed to ensure their endorsement by regulatory authorities as alternatives to histological endpoints in phase 3 trials. This Review describes the challenges of drug development in NAFLD-NASH trials and potential mitigating strategies to move the field forward.
Collapse
Affiliation(s)
| | - Alina M Allen
- Division of Gastroenterology and Hepatology, Mayo Clinic Rochester, Rochester, MN, USA
| | | | | | - Naim Alkhouri
- Department of Hepatology, Arizona Liver Health, Chandler, AZ, USA
| |
Collapse
|
83
|
Jacobson IM, Wong VWS, Castera L, Anstee QM, Noureddin M, Cusi K, Harrison SA, Bugianesi E, Younossi ZM. Expert Panel Consensus on Clinical Assertion Statements Describing Noninvasive Tools for Diagnosing Nonalcoholic Steatohepatitis. J Clin Gastroenterol 2023; 57:253-264. [PMID: 36251413 PMCID: PMC9911115 DOI: 10.1097/mcg.0000000000001780] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/10/2022]
Abstract
GOALS AND BACKGROUND A panel of 9 experts in nonalcoholic steatohepatitis gathered to assess multiple components of the diagnostic process. MATERIALS AND METHODS The Clinical Assertion Statements covered screening of patients with type 2 diabetes for high-risk nonalcoholic fatty liver disease, which-if any-noninvasive tests could determine whether to delay or defer biopsy, whether primary care providers and endocrinologists should routinely calculate Fibrosis-4 (FIB-4) scores in patients with nonalcoholic fatty liver disease or those at risk for it, optimal noninvasive tests to stage fibrosis, the need to consider fibrosis in patients with normal transaminase levels, periodic monitoring for progressive fibrosis, whether patients should undergo biopsy before pharmacotherapy, and the clinical utility of genetic testing. RESULTS AND CONCLUSIONS Evidence was presented to support or refute each Clinical Assertion Statement; the panel voted on the nature of the evidence, level of support, and level of agreement with each Statement. Panel level of agreement and rationale of each Clinical Assertion Statement are reported here.
Collapse
Affiliation(s)
- Ira M. Jacobson
- Department of Medicine and Therapeutics, NYU Langone Health, New York, NY
| | - Vincent Wai-Sun Wong
- Department of Medicine and Therapeutics
- State Key Laboratory of Digestive Disease, The Chinese University of Hong Kong, Hong Kong, Hong Kong
| | - Laurent Castera
- Department of Hepatology, Hôpital Beaujon (Beaujon Hospital), Assistance Publique-Hôpitaux de Paris, Clichy
- Department of Hepatology, University of Paris, Paris, France
| | - Quentin M. Anstee
- Translational and Clinical Research Institute, Faculty of Medical Sciences, Newcastle University
- Newcastle NIHR Biomedical Research Centre, Newcastle upon Tyne Hospitals NHS Foundation Trust, Newcastle upon Tyne
| | - Mazen Noureddin
- Fatty Liver Program, Karsh Division of Gastroenterology and Hepatology, Cedars Sinai Medical Center, Los Angeles, CA
| | - Kenneth Cusi
- Division of Endocrinology, Diabetes and Metabolism, University of Florida
- Malcom Randall VAMC, Gainesville, FL
| | | | - Elisabetta Bugianesi
- Department of Medical Sciences, Division of Gastroenterology, University of Torino, Torino, Italy
| | - Zobair M. Younossi
- Inova Medicine, Inova Health System
- Department of Medicine, Center for Liver Diseases, Inova Fairfax Medical Campus, Falls Church, VA
| |
Collapse
|
84
|
Sun Y, Shen Y, Liang X, Zheng H, Zhang Y. MicroRNAs as Biomarkers and Therapeutic Targets for Nonalcoholic Fatty Liver Disease: A Narrative Review. Clin Ther 2023; 45:234-247. [PMID: 36841739 DOI: 10.1016/j.clinthera.2023.02.001] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2022] [Revised: 02/03/2023] [Accepted: 02/04/2023] [Indexed: 02/27/2023]
Abstract
PURPOSE Nonalcoholic fatty liver disease (NAFLD) is the most common chronic liver disease in the world. However, biomarkers for NAFLD diagnosis and liver-specific drugs for treatment are lacking. This article reviews the possibility of circulating miRNAs in the diagnosis and treatment of NAFLD diseases and focuses on several well-studied miRNAs to provide preclinical data for subsequent related studies. METHODS Related articles were identified through searches of the PubMed database for literature published from 2010 to December 2022. Search terms included NAFLD, microRNA, biomarker, diagnosis, and therapy. FINDINGS Current research data indicate that some key circulating miRNAs may be used as diagnostic biomarkers of NAFLD and the combination of several miRNAs improves diagnostic performance. In addition, some preclinical trials using cell and mouse models provide a basis for some miRNAs as potential therapeutic targets. IMPLICATIONS Current evidence suggests that circulating miRNAs are potential noninvasive biomarkers for clinical diagnosis of NAFLD, which needs to be validated in more heterogeneous and larger cohorts. In addition, several miRNAs regulate multiple downstream pathways related to the pathophysiology of NAFLD in a cell- and tissue-specific manner, making them attractive drug therapeutic targets for NAFLD. However, more preclinical and clinical trials are needed for these miRNAs to become therapeutic targets of NAFLD.
Collapse
Affiliation(s)
- Yu Sun
- Department of Clinical Laboratory, Tianjin Children's Hospital/Tianjin University Children's Hospital, 238 Longyan Road, Beichen District, 300134 Tianjin, China.
| | - Yongming Shen
- Department of Clinical Laboratory, Tianjin Children's Hospital/Tianjin University Children's Hospital, 238 Longyan Road, Beichen District, 300134 Tianjin, China
| | - Xiurui Liang
- Department of Cardiology, The First Affiliated Hospital of Liaoning University of Traditional Chinese Medicine, Shenyang, China
| | - Huilin Zheng
- School of Biological & Chemical Engineering, Zhejiang University of Science and Technology, Zhejiang, China
| | - Yitong Zhang
- Department of Clinical Laboratory, Tianjin Children's Hospital/Tianjin University Children's Hospital, 238 Longyan Road, Beichen District, 300134 Tianjin, China
| |
Collapse
|
85
|
Stefan N, Schick F, Birkenfeld AL, Häring HU, White MF. The role of hepatokines in NAFLD. Cell Metab 2023; 35:236-252. [PMID: 36754018 PMCID: PMC10157895 DOI: 10.1016/j.cmet.2023.01.006] [Citation(s) in RCA: 139] [Impact Index Per Article: 69.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/09/2022] [Revised: 11/18/2022] [Accepted: 01/13/2023] [Indexed: 02/09/2023]
Abstract
Non-alcoholic fatty liver disease (NAFLD) is not only a consequence of insulin resistance, but it is also an important cause of insulin resistance and major non-communicable diseases (NCDs). The close relationship of NAFLD with visceral obesity obscures the role of fatty liver from visceral adiposity as the main pathomechanism of insulin resistance and NCDs. To overcome this limitation, in analogy to the concept of adipokines, in 2008 we introduced the term hepatokines to describe the role of fetuin-A in metabolism. Since then, several other hepatokines were tested for their effects on metabolism. Here we address the dysregulation of hepatokines in people with NAFLD. Then, we discuss pathophysiological mechanisms of cardiometabolic diseases specifically related to NAFLD by focusing on hepatokine-related organ crosstalk. Finally, we propose how the determination of major hepatokines and adipokines can be used for pathomechanism-based clustering of insulin resistance in NAFLD and visceral obesity to better implement precision medicine in clinical practice.
Collapse
Affiliation(s)
- Norbert Stefan
- Department of Internal Medicine IV, Division of Endocrinology, Diabetology and Nephrology, University Hospital of Tübingen, Otfried-Müller Str. 10, 72076 Tübingen, Germany; Institute of Diabetes Research and Metabolic Diseases (IDM) of the Helmholtz Center Munich, Tübingen, Germany; German Center for Diabetes Research (DZD), Neuherberg, Germany.
| | - Fritz Schick
- Institute of Diabetes Research and Metabolic Diseases (IDM) of the Helmholtz Center Munich, Tübingen, Germany; German Center for Diabetes Research (DZD), Neuherberg, Germany; Section of Experimental Radiology, Department of Radiology, University Hospital of Tübingen, Tübingen, Germany
| | - Andreas L Birkenfeld
- Department of Internal Medicine IV, Division of Endocrinology, Diabetology and Nephrology, University Hospital of Tübingen, Otfried-Müller Str. 10, 72076 Tübingen, Germany; Institute of Diabetes Research and Metabolic Diseases (IDM) of the Helmholtz Center Munich, Tübingen, Germany; German Center for Diabetes Research (DZD), Neuherberg, Germany
| | - Hans-Ulrich Häring
- Department of Internal Medicine IV, Division of Endocrinology, Diabetology and Nephrology, University Hospital of Tübingen, Otfried-Müller Str. 10, 72076 Tübingen, Germany; Institute of Diabetes Research and Metabolic Diseases (IDM) of the Helmholtz Center Munich, Tübingen, Germany; German Center for Diabetes Research (DZD), Neuherberg, Germany
| | - Morris F White
- Division of Endocrinology, Boston Children's Hospital, Harvard Medical School, 300 Longwood Avenue, Boston, MA 02115, USA.
| |
Collapse
|
86
|
Indira Chandran V, Wernberg CW, Lauridsen MM, Skytthe MK, Bendixen SM, Larsen FT, Hansen CD, Grønkjær LL, Siersbæk MS, Caterino TD, Detlefsen S, Møller HJ, Grøntved L, Ravnskjaer K, Moestrup SK, Thiele MS, Krag A, Graversen JH. Circulating TREM2 as a noninvasive diagnostic biomarker for NASH in patients with elevated liver stiffness. Hepatology 2023; 77:558-572. [PMID: 35712786 PMCID: PMC9869959 DOI: 10.1002/hep.32620] [Citation(s) in RCA: 34] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/18/2022] [Revised: 05/24/2022] [Accepted: 06/12/2022] [Indexed: 02/01/2023]
Abstract
BACKGROUND AND AIMS Reliable noninvasive biomarkers are an unmet clinical need for the diagnosis of NASH. This study investigates the diagnostic accuracy of the circulating triggering receptor expressed on myeloid cells 2 (plasma TREM2) as a biomarker for NASH in patients with NAFLD and elevated liver stiffness. APPROACH AND RESULTS We collected cross-sectional, clinical data including liver biopsies from a derivation ( n = 48) and a validation cohort ( n = 170) of patients with elevated liver stiffness measurement (LSM ≥ 8.0 kPa). Patients with NAFLD activity scores (NAS) ≥4 were defined as having NASH. Plasma TREM2 levels were significantly elevated in patients with NASH of the derivation cohort, with an area under the receiver operating characteristics curve (AUROC) of 0.92 (95% confidence interval [CI], 0.84-0.99). In the validation cohort, plasma TREM2 level increased approximately two-fold in patients with NASH, and a strong diagnostic accuracy was confirmed (AUROC, 0.83; 95% CI, 0.77-0.89; p < 0.0001). Plasma TREM2 levels were associated with the individual histologic features of NAS: steatosis, lobular inflammation, and ballooning ( p < 0.0001), but only weakly with fibrosis stages. Dual cutoffs for rule-in and rule-out were explored: a plasma TREM2 level of ≤38 ng/ml was found to be an optimal NASH rule-out cutoff (sensitivity 90%; specificity 52%), whereas a plasma TREM2 level of ≥65 ng/ml was an optimal NASH rule-in cutoff (specificity 89%; sensitivity 54%). CONCLUSIONS Plasma TREM2 is a plausible individual biomarker that can rule-in or rule-out the presence of NASH with high accuracy and thus has the potential to reduce the need for liver biopsies and to identify patients who are eligible for clinical trials in NASH.
Collapse
Affiliation(s)
| | - Charlotte Wilhelmina Wernberg
- Liver Research Group, Department of Gastroenterology and Hepatology, University Hospital of South Denmark, Esbjerg, Denmark,Centre for Liver Research, Department of Gastroenterology and Hepatology, Odense University Hospital, Odense, Denmark
| | - Mette Munk Lauridsen
- Liver Research Group, Department of Gastroenterology and Hepatology, University Hospital of South Denmark, Esbjerg, Denmark
| | | | | | - Frederik Tibert Larsen
- Department of Biochemistry and Molecular Biology, University of Southern Denmark, Odense, Denmark
| | - Camilla Dalby Hansen
- Centre for Liver Research, Department of Gastroenterology and Hepatology, Odense University Hospital, Odense, Denmark
| | - Lea Ladegaard Grønkjær
- Liver Research Group, Department of Gastroenterology and Hepatology, University Hospital of South Denmark, Esbjerg, Denmark
| | - Majken Storm Siersbæk
- Department of Biochemistry and Molecular Biology, University of Southern Denmark, Odense, Denmark
| | - Tina Di Caterino
- Department of Pathology, Odense University Hospital, Odense, Denmark
| | - Sönke Detlefsen
- Department of Pathology, Odense University Hospital, Odense, Denmark
| | - Holger Jon Møller
- Department of Clinical Biochemistry, Aarhus University Hospital, Aarhus, Denmark,Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
| | - Lars Grøntved
- Department of Biochemistry and Molecular Biology, University of Southern Denmark, Odense, Denmark
| | - Kim Ravnskjaer
- Department of Biochemistry and Molecular Biology, University of Southern Denmark, Odense, Denmark
| | - Søren Kragh Moestrup
- Department of Molecular Medicine, University of Southern Denmark, Odense, Denmark
| | - Maja Sofie Thiele
- Centre for Liver Research, Department of Gastroenterology and Hepatology, Odense University Hospital, Odense, Denmark
| | - Aleksander Krag
- Centre for Liver Research, Department of Gastroenterology and Hepatology, Odense University Hospital, Odense, Denmark
| | | |
Collapse
|
87
|
Toyoda H, Kariyama K, Hiraoka A. Letter: rising incidence and poor survival in patients with non-viral HCC - better HCC surveillance and treatment for alcohol-associated and non-alcohol fatty liver diseases are needed. Authors' reply. Aliment Pharmacol Ther 2023; 57:363-364. [PMID: 36641796 DOI: 10.1111/apt.17361] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/01/2022] [Revised: 12/03/2022] [Accepted: 12/06/2022] [Indexed: 01/16/2023]
Affiliation(s)
- Hidenori Toyoda
- Department of Gastroenterology, Ogaki Municipal Hospital, Ogaki, Japan
| | - Kazuya Kariyama
- Department of Hepatology, Okayama City Hospital, Okayama, Japan
| | - Atsushi Hiraoka
- Gastroenterology Center, Ehime Prefectural Central Hospital, Matsuyama, Japan
| |
Collapse
|
88
|
Chang D, Truong E, Mena EA, Pacheco F, Wong M, Guindi M, Todo TT, Noureddin N, Ayoub W, Yang JD, Kim IK, Kohli A, Alkhouri N, Harrison S, Noureddin M. Machine learning models are superior to noninvasive tests in identifying clinically significant stages of NAFLD and NAFLD-related cirrhosis. Hepatology 2023; 77:546-557. [PMID: 35809234 DOI: 10.1002/hep.32655] [Citation(s) in RCA: 37] [Impact Index Per Article: 18.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/11/2021] [Revised: 06/28/2022] [Accepted: 07/05/2022] [Indexed: 01/28/2023]
Abstract
BACKGROUND AND AIMS We assessed the performance of machine learning (ML) models in identifying clinically significant NAFLD-associated liver fibrosis and cirrhosis. APPROACH AND RESULTS We implemented ML models including logistic regression (LR), random forest (RF), and artificial neural network to predict histological stages of fibrosis using 17 demographic/clinical features in 1370 patients with NAFLD who underwent liver biopsy, FibroScan, and labs within a 6-month period at multiple U.S. centers. Histological stages of fibrosis (≥F2, ≥F3, and F4) were predicted using ML, FibroScan liver stiffness measurements, and Fibrosis-4 index (FIB-4). NASH with significant fibrosis (NAS ≥ 4 + ≥F2) was assessed using ML, FibroScan-AST (FAST) score, FIB-4, and NAFLD fibrosis score (NFS). We used 80% of the cohort to train and 20% to test the ML models. For ≥F2, ≥F3, F4, and NASH + NAS ≥ 4 + ≥F2, all ML models, especially RF, had primarily higher accuracy and AUC compared with FibroScan, FIB-4, FAST, and NFS. AUC for RF versus FibroScan and FIB-4 for ≥F2, ≥F3, and F4 were (0.86 vs. 0.81, 0.78), (0.89 vs. 0.83, 0.82), and (0.89 vs. 0.86, 0.85), respectively. AUC for RF versus FAST, FIB-4, and NFS for NASH + NAS ≥ 4 + ≥F2 were (0.80 vs. 0.77, 0.66, 0.63). For NASH + NAS ≥ 4 + ≥F2, all ML models had lower/similar percentages within the indeterminate zone compared with FIB-4 and NFS. Overall, ML models performed better in sensitivity, specificity, positive predictive value, and negative predictive value compared with traditional noninvasive tests. CONCLUSIONS ML models performed better overall than FibroScan, FIB-4, FAST, and NFS. ML could be an effective tool for identifying clinically significant liver fibrosis and cirrhosis in patients with NAFLD.
Collapse
Affiliation(s)
- Devon Chang
- Arnold O. Beckman High School , Irvine , California , USA
| | - Emily Truong
- Department of Medicine , Cedars Sinai Medical Center , Los Angeles , California , USA
| | - Edward A Mena
- California Liver Institute , Pasadena , California , USA
| | | | - Micaela Wong
- California Liver Institute , Pasadena , California , USA
| | - Maha Guindi
- Department of Pathology , Cedars-Sinai Medical Center , Los Angeles , California , USA
| | - Tsuyoshi T Todo
- Comprehensive Transplant Center , Cedars-Sinai Medical Center , Los Angeles , California , USA
| | - Nabil Noureddin
- Division of Gastroenterology , University of California at San Diego , La Jolla , California , USA
| | - Walid Ayoub
- Department of Medicine , Cedars Sinai Medical Center , Los Angeles , California , USA.,Comprehensive Transplant Center , Cedars-Sinai Medical Center , Los Angeles , California , USA.,Karsh Division of Gastroenterology and Hepatology , Cedars-Sinai Medical Center , Los Angeles , California , USA
| | - Ju Dong Yang
- Department of Medicine , Cedars Sinai Medical Center , Los Angeles , California , USA.,Comprehensive Transplant Center , Cedars-Sinai Medical Center , Los Angeles , California , USA.,Karsh Division of Gastroenterology and Hepatology , Cedars-Sinai Medical Center , Los Angeles , California , USA
| | - Irene K Kim
- Comprehensive Transplant Center , Cedars-Sinai Medical Center , Los Angeles , California , USA
| | - Anita Kohli
- Arizona Liver Health , Phoenix , Arizona , USA
| | | | - Stephen Harrison
- Oxford University, Pinnacle Research Center , Live Oak , Texas , USA
| | - Mazen Noureddin
- Department of Medicine , Cedars Sinai Medical Center , Los Angeles , California , USA.,Comprehensive Transplant Center , Cedars-Sinai Medical Center , Los Angeles , California , USA.,Karsh Division of Gastroenterology and Hepatology , Cedars-Sinai Medical Center , Los Angeles , California , USA
| |
Collapse
|
89
|
Shi YW, Fan JG. Surveillance of the progression and assessment of treatment endpoints for nonalcoholic steatohepatitis. Clin Mol Hepatol 2023; 29:S228-S243. [PMID: 36521452 PMCID: PMC10029951 DOI: 10.3350/cmh.2022.0401] [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: 11/15/2022] [Revised: 12/08/2022] [Accepted: 12/10/2022] [Indexed: 12/23/2022] Open
Abstract
Nonalcoholic steatohepatitis (NASH) is an aggressive form of nonalcoholic fatty liver disease (NAFLD) characterized by steatosis-associated inflammation and liver injury. Without effective treatment or management, NASH can have life-threatening outcomes. Evaluation and identification of NASH patients at risk for adverse outcomes are therefore important. Key issues in screening NASH patients are the assessment of advanced fibrosis, differentiation of NASH from simple steatosis, and monitoring of dynamic changes during follow-up and treatment. Currently, NASH staging and evaluation of the effectiveness for drugs still rely on pathological diagnosis, despite sample error issues and the subjectivity associated with liver biopsy. Optimizing the pathological assessment of liver biopsy samples and developing noninvasive surrogate methods for accessible, accurate, and safe evaluation are therefore critical. Although noninvasive methods including elastography, serum soluble biomarkers, and combined models have been implemented in the last decade, noninvasive diagnostic measurements are not widely applied in clinical practice. More work remains to be done in establishing cost-effective strategies both for screening for at-risk NASH patients and identifying changes in disease severity. In this review, we summarize the current state of noninvasive methods for detecting steatosis, steatohepatitis, and fibrosis in patients with NASH, and discuss noninvasive assessments for screening at-risk patients with a focus on the characteristics that should be monitored at follow-up.
Collapse
Affiliation(s)
- Yi-wen Shi
- Center for Fatty Liver, Department of Gastroenterology, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai Key Lab of Pediatric Gastroenterology and Nutrition, Shanghai, China
| | - Jian-Gao Fan
- Center for Fatty Liver, Department of Gastroenterology, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai Key Lab of Pediatric Gastroenterology and Nutrition, Shanghai, China
| |
Collapse
|
90
|
Another arrow in the NASH quiver? Lancet Gastroenterol Hepatol 2023; 8:96-97. [PMID: 36521502 DOI: 10.1016/s2468-1253(22)00383-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/01/2022] [Accepted: 11/02/2022] [Indexed: 12/15/2022]
|
91
|
Atic AI, Thiele M, Munk A, Dalgaard LT. Circulating miRNAs associated with nonalcoholic fatty liver disease. Am J Physiol Cell Physiol 2023; 324:C588-C602. [PMID: 36645666 DOI: 10.1152/ajpcell.00253.2022] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
MicroRNAs (miRNAs) are secreted from cells as either protein-bound or enclosed in extracellular vesicles. Circulating liver-derived miRNAs are modifiable by weight-loss or insulin-sensitizing treatments, indicating that they could be important biomarker candidates for diagnosis, monitoring, and prognosis in nonalcoholic liver disease (NAFLD) and nonalcoholic steatohepatitis (NASH). Unfortunately, the noninvasive diagnosis of NASH and fibrosis remains a key challenge, which limits case finding. Current diagnostic guidelines, therefore, recommend liver biopsies, with risks of pain and bleeding for the patient and substantial healthcare costs. Here, we summarize mechanisms of RNA secretion and review circulating RNAs associated with NAFLD and NASH for their biomarker potential. Few circulating miRNAs are consistently associated with NAFLD/NASH: miR-122, miR-21, miR-34a, miR-192, miR-193, and the miR-17-92 miRNA-cluster. The hepatocyte-enriched miRNA-122 is consistently increased in NAFLD and NASH but decreased in liver cirrhosis. Circulating miR-34a, part of an existing diagnostic algorithm for NAFLD, and miR-21 are consistently increased in NAFLD and NASH. MiR-192 appears to be prominently upregulated in NASH compared with NAFDL, whereas miR-193 was reported to distinguish NASH from fibrosis. Various members of miRNA cluster miR-17-92 are reported to be associated with NAFLD and NASH, although with less consistency. Several other circulating miRNAs have been reported to be associated with fatty liver in a few studies, indicating the existence of more circulating miRNAs with relevant as diagnostic markers for NAFLD or NASH. Thus, circulating miRNAs show potential as biomarkers of fatty liver disease, but more information about phenotype specificity and longitudinal regulation is needed.
Collapse
Affiliation(s)
- Amila Iriskic Atic
- Department of Science and Environment, Roskilde University, Roskilde, Denmark.,Novo Nordisk A/S, Obesity Research, Måløv, Denmark
| | - Maja Thiele
- Department of Gastroenterology and Hepatology, Center for Liver Research, Odense University Hospital, Odense, Denmark.,Department of Clinical Research, Faculty of Health Sciences, University of Southern Denmark, Odense, Denmark
| | | | | |
Collapse
|
92
|
Huang Y, Dong S, Wang C, Dong Z, Chen W. Significant fibrosis assessed by liver biopsy among Chinese bariatric surgery patients: A prospective cross-sectional study. Front Endocrinol (Lausanne) 2023; 14:1090598. [PMID: 36793287 PMCID: PMC9922831 DOI: 10.3389/fendo.2023.1090598] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/10/2022] [Accepted: 01/04/2023] [Indexed: 01/31/2023] Open
Abstract
BACKGROUND Fibrosis stages affect clinical prognoses related to nonalcoholic fatty liver disease (NAFLD). However, data on the prevalence and clinical features of significant fibrosis are scarce in Chinese bariatric surgery patients. We aimed to investigate the prevalence of significant fibrosis in bariatric surgery patients and to identify its predictors. METHODS We prospectively enrolled the patients performing intra-operative liver biopsies during bariatric surgery from a bariatric surgery center in a university hospital between May 2020 and January 2022. Anthropometric characteristics, co-morbidities, laboratory data and pathology reports were collected and analyzed. The performance of non-invasive models was evaluated. RESULTS Of 373 patients, 68.9%% had non-alcoholic steatohepatitis (NASH) and 60.9% exhibited fibrosis. Significant fibrosis was present in 9.1% of patients, advanced fibrosis in 4.0%, and cirrhosis in 1.6%. Multivariate logistic regression showed that increasing age (odds ratio [OR], 1.06; p=0.003), presence of diabetes (OR, 2.62; p=0.019), elevated c- peptide (OR, 1.26; p=0.025) and elevated aspartate aminotransferase (AST) (OR, 1.02; p=0.004) were independent predictors of significant fibrosis. The non-invasive models, AST to Platelet ratio (APRI), Fibrosis-4 (FIB-4), and Hepamet fibrosis scores (HFS) provided greater accuracy for predicting significant fibrosis, compared to the NAFLD Fibrosis Score (NFS) and BARD score. CONCLUSION More than two-thirds of bariatric surgery patients had NASH and the prevalence of significant fibrosis was high. Elevated levels of AST and c- peptide, advanced age and diabetes indicated a higher risk of significant fibrosis. Non-invasive models, APRI, FIB-4 and HFS can be used to identify significant liver fibrosis in bariatric surgery patients.
Collapse
Affiliation(s)
- Yongsheng Huang
- Department of Gastrointestinal Surgery, The First People’s Hospital of Zunyi and Third Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou, China
| | - Shiliang Dong
- Department of Metabolic and Bariatric Surgery, The First Affiliated Hospital of Jinan University, Guangzhou, China
| | - Cunchuan Wang
- Department of Metabolic and Bariatric Surgery, The First Affiliated Hospital of Jinan University, Guangzhou, China
| | - Zhiyong Dong
- Department of Metabolic and Bariatric Surgery, The First Affiliated Hospital of Jinan University, Guangzhou, China
| | - Wenhui Chen
- Department of Metabolic and Bariatric Surgery, The First Affiliated Hospital of Jinan University, Guangzhou, China
| |
Collapse
|
93
|
Understanding NAFLD: From Case Identification to Interventions, Outcomes, and Future Perspectives. Nutrients 2023; 15:nu15030687. [PMID: 36771394 PMCID: PMC9921401 DOI: 10.3390/nu15030687] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2023] [Revised: 01/25/2023] [Accepted: 01/28/2023] [Indexed: 02/01/2023] Open
Abstract
While non-alcoholic fatty liver disease (NAFLD) is a prevalent and frequent cause of liver-related morbidity and mortality, it is also strongly associated with cardiovascular disease-related morbidity and mortality, likely driven by its associations with insulin resistance and other manifestations of metabolic dysregulation. However, few satisfactory pharmacological treatments are available for NAFLD due in part to its complex pathophysiology, and challenges remain in stratifying individual patient's risk for liver and cardiovascular disease related outcomes. In this review, we describe the development and progression of NAFLD, including its pathophysiology and outcomes. We also describe different tools for identifying patients with NAFLD who are most at risk of liver-related and cardiovascular-related complications, as well as current and emerging treatment options, and future directions for research.
Collapse
|
94
|
Sanyal A, Shankar S, Yates K, Bolognese J, Daly E, Dehn C, Neuschwander-Tetri B, Kowdley K, Vuppalanchi R, Behling CA, Tonascia J, Samir A, Sirlin C, Sherlock S, Fowler K, Heymann H, Kamphaus T, Loomba R, Calle R. The Nimble Stage 1 Study Validates Diagnostic Circulating Biomarkers for Nonalcoholic Steatohepatitis. RESEARCH SQUARE 2023:rs.3.rs-2492725. [PMID: 36711803 PMCID: PMC9882658 DOI: 10.21203/rs.3.rs-2492725/v1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
Background There are no approved noninvasive tests (NIT) for the diagnosis of nonalcoholic steatohepatitis (NASH) and its histological phenotypes. Methods The FNIH-NIMBLE consortium tested 5 serum-based NIT panels for the following intended uses: NIS4: At-risk NASH, a composite of NASH with NAFLD activity score (NAS) ≥ 4 and fibrosis stage ≥ 2, OWLiver: NASH and NAS ≥ 4, enhanced liver fibrosis (ELF), PROC3 and Fibrometer VCTE: fibrosis stages ≥ 2, ≥ 3 or 4. Aliquots from a single blood sample obtained within 90 days of histological confirmation of NAFLD were tested. The prespecified performance metric tested for was a diagnostic AUROC greater than 0.7 and superiority to ALT for diagnosis of NASH or NAS ≥ 4 and to FIB-4 for fibrosis. Results A total of 1073 adults including NASH (n = 848), at-risk NASH (n = 539) and fibrosis stages 0-4 (n = 222, 114, 262, 277 and 198 respectively) were studied. The AUROC of NIS4 for at-risk NASH was 0.81 and superior to ALT and FIB4 (p < 0.001 for both). OWliver diagnosed NASH with sensitivity and specificity of 77.3% and 66.8% respectively. The AUROCs (95% CI) of ELF, PROC3 and Fibrometer VCTE respectively for fibrosis were as follows: ≥ stage 2 fibrosis [0.82 (0.8-0.85), 0.8 (0.77-0.83), and 0.84 (0.79-0.88)], ≥ stage 3 [0.83 (0.8-0.86), 0.76 (0.73-0.79), 0.85 (0.81-0.9), stage 4 [0.85 (0.81-0.89), 0.81 (0.77-0.85), 0.89 (0.84-0.95)]. ELF and Fibrometer VCTE were significantly superior to FIB-4 for all fibrosis endpoints (p < 0.01 for all). Conclusions These data support the further development of NIS4, ELF and Fibrometer VCTE for their intended uses.
Collapse
Affiliation(s)
- Arun Sanyal
- Virginia Commonwealth University School of Medicine
| | | | | | | | | | | | | | | | | | | | - James Tonascia
- Bloomberg School of Public Health, Johns Hopkins University
| | | | | | | | | | | | | | | | | |
Collapse
|
95
|
Dinesen S, El-Faitarouni A, Dalgaard LT. Circulating microRNAs associated with gestational diabetes mellitus: useful biomarkers? J Endocrinol 2023; 256:JOE-22-0170. [PMID: 36346274 DOI: 10.1530/joe-22-0170] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/28/2022] [Accepted: 11/08/2022] [Indexed: 11/09/2022]
Abstract
Different types of small non-coding RNAs, especially miRNAs, may be found in the circulation, either protein-bound or enclosed in extracellular vesicles. During gestation, and particularly during gestational diabetes mellitus (GDM), the levels of several miRNAs are altered. Worldwide the incidence of GDM is increasing, in part driven by the current obesity epidemic. This is a point of public health concern because offspring of women with GDM frequently suffer from short- and long-term complications of maternal GDM. This has prompted the investigation of whether levels of specific miRNA species, detected early in gestation, may be used as diagnostic or prognostic markers for the development of GDM. Here, we summarize the mechanisms of RNA secretion and review circulating miRNAs associated with GDM. Several miRNAs are associated with GDM: miR-29a-3p and miR-29b-3p are generally upregulated in GDM pregnancies, also when measured prior to the development of GDM, while miR-16-5p is consistently upregulated in GDM pregnancies, especially in late gestation. miR-330-3p in circulation is increased in late gestation GDM women, especially in those with poor insulin secretion. miR-17-5p, miR-19a/b-3p, miR-223-3p, miR-155-5p, miR-125-a/b-5p, miR-210-3p and miR-132 are also associated with GDM, but less so and with more contradictory results reported. There could be a publication bias as miRNAs identified early are investigated the most, suggesting that it is likely that additional, more recently detected miRNAs could also be associated with GDM. Thus, circulating miRNAs show potential as biomarkers of GDM diagnosis or prognosis, especially multiple miRNAs containing prediction algorithms show promise, but further studies are needed.
Collapse
Affiliation(s)
- Sofie Dinesen
- Department of Science and Environment, Roskilde University, Universitetsvej 1, Roskilde, Denmark
| | - Alisar El-Faitarouni
- Department of Science and Environment, Roskilde University, Universitetsvej 1, Roskilde, Denmark
| | - Louise T Dalgaard
- Department of Science and Environment, Roskilde University, Universitetsvej 1, Roskilde, Denmark
| |
Collapse
|
96
|
Lu F, Du L, Chen W, Jiang H, Yang C, Pu Y, Wu J, Zhu J, Chen T, Zhang X, Wu C. T 1- T 2 dual-modal magnetic resonance contrast-enhanced imaging for rat liver fibrosis stage. RSC Adv 2022; 12:35809-35819. [PMID: 36545112 PMCID: PMC9749127 DOI: 10.1039/d2ra05913d] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2022] [Accepted: 12/02/2022] [Indexed: 12/16/2022] Open
Abstract
The development of an effective method for staging liver fibrosis has always been a hot topic of research in the field of liver fibrosis. In this paper, PEGylated ultrafine superparamagnetic iron oxide nanocrystals (SPIO@PEG) were developed for T 1-T 2 dual-modal contrast-enhanced magnetic resonance imaging (MRI) and combined with Matrix Laboratory (MATLAB)-based image fusion for staging liver fibrosis in the rat model. Firstly, SPIO@PEG was synthesized and characterized with physical and biological properties as a T 1-T 2 dual-mode MRI contrast agent. Secondly, in the subsequent MR imaging of liver fibrosis in rats in vivo, conventional T 1 and T 2-weighted imaging, and T 1 and T 2 mapping of the liver pre- and post-intravenous administration of SPIO@PEG were systematically collected and analyzed. Thirdly, by creative design, we fused the T 1 and T 2 mapping images by MATLAB and quantitively measured each rat's hepatic fibrosis positive pixel ratio (PPR). SPIO@PEG was proved to have an ultrafine core size (4.01 ± 0.16 nm), satisfactory biosafety and T 1-T 2 dual-mode contrast effects under a 3.0 T MR scanner (r 2/r 1 = 3.51). According to the image fusion results, the SPIO@PEG contrast-enhanced PPR shows significant differences among different stages of liver fibrosis (P < 0.05). The combination of T 1-T 2 dual-modal SPIO@PEG and MATLAB-based image fusion technology could be a promising method for diagnosing and staging liver fibrosis in the rat model. PPR could also be used as a non-invasive biomarker to diagnose and discriminate the stages of liver fibrosis.
Collapse
Affiliation(s)
- Fulin Lu
- Medical Imaging Key Laboratory of Sichuan Province, School of Medical Imaging, Affiliated Hospital of North Sichuan Medical College Nanchong 637000 China
- Department of Radiology, Sichuan Academy of Medical Sciences, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China Chengdu 610072 China
| | - Liang Du
- Medical Imaging Key Laboratory of Sichuan Province, School of Medical Imaging, Affiliated Hospital of North Sichuan Medical College Nanchong 637000 China
| | - Wei Chen
- Medical Imaging Key Laboratory of Sichuan Province, School of Medical Imaging, Affiliated Hospital of North Sichuan Medical College Nanchong 637000 China
| | - Hai Jiang
- Medical Imaging Key Laboratory of Sichuan Province, School of Medical Imaging, Affiliated Hospital of North Sichuan Medical College Nanchong 637000 China
| | - Chenwu Yang
- Medical Imaging Key Laboratory of Sichuan Province, School of Medical Imaging, Affiliated Hospital of North Sichuan Medical College Nanchong 637000 China
| | - Yu Pu
- Medical Imaging Key Laboratory of Sichuan Province, School of Medical Imaging, Affiliated Hospital of North Sichuan Medical College Nanchong 637000 China
| | - Jun Wu
- Medical Imaging Key Laboratory of Sichuan Province, School of Medical Imaging, Affiliated Hospital of North Sichuan Medical College Nanchong 637000 China
| | - Jiang Zhu
- Medical Imaging Key Laboratory of Sichuan Province, School of Medical Imaging, Affiliated Hospital of North Sichuan Medical College Nanchong 637000 China
| | - Tianwu Chen
- Medical Imaging Key Laboratory of Sichuan Province, School of Medical Imaging, Affiliated Hospital of North Sichuan Medical College Nanchong 637000 China
| | - Xiaoming Zhang
- Medical Imaging Key Laboratory of Sichuan Province, School of Medical Imaging, Affiliated Hospital of North Sichuan Medical College Nanchong 637000 China
| | - Changqiang Wu
- Medical Imaging Key Laboratory of Sichuan Province, School of Medical Imaging, Affiliated Hospital of North Sichuan Medical College Nanchong 637000 China
| |
Collapse
|
97
|
Baldelli E, Lonardo A. Digging the metabolic roots of NASH up. LIFE METABOLISM 2022; 1:203-204. [PMID: 39872078 PMCID: PMC11749814 DOI: 10.1093/lifemeta/loac036] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/20/2022] [Accepted: 11/29/2022] [Indexed: 01/03/2025]
Affiliation(s)
- Enrica Baldelli
- Department of Biomedical, Metabolic and Neural Sciences, University of Modena and Reggio Emilia, Modena, Italy
| | - Amedeo Lonardo
- Azienda Ospedaliero-Universitaria di Modena—Department of Internal Medicine—Ospedale Civile di Baggiovara, Modena, Italy
| |
Collapse
|
98
|
Ratziu V, Anstee QM, Wong VWS, Schattenberg JM, Bugianesi E, Augustin S, Gheorghe L, Zambon V, Reau N. An international survey on patterns of practice in NAFLD and expectations for therapies-The POP-NEXT project. Hepatology 2022; 76:1766-1777. [PMID: 35363906 DOI: 10.1002/hep.32500] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/08/2022] [Revised: 03/27/2022] [Accepted: 03/28/2022] [Indexed: 12/08/2022]
Abstract
BACKGROUND AND AIMS Differences between countries in NAFLD patient care pathways and management need to be understood prior to defining supranational guidelines. APPROACH AND RESULTS We conducted an anonymous survey in France, Germany, Hong Kong, Italy, Romania, Spain, the United Kingdom, and the United States among physicians providing specialist care for patients with NAFLD. Modalities of patient referral, patterns of practice (diagnosis, staging, monitoring, and indications for liver biopsy), therapeutic management, and expectations for future NASH pharmacotherapies were assessed, with 664 physicians completing the survey. Referral to surveyed physicians (SPs) mostly came from primary care. Prior to referral, NAFLD was rarely diagnosed, and noninvasive tests were not performed. Screening for comorbidities by SPs was incomplete and cardiovascular risk not calculated. Elastometry in combination with a serum biomarker was the most common first-line method for fibrosis staging. Liver biopsy, when performed, was often delayed by at least 1 year after diagnosis. It was, however, recommended even if noninvasive methods indicated advanced fibrosis. Frequent, biannual monitoring was conducted, including HCC surveillance in Stage 3 fibrosis. SPs rarely implemented and followed dietary and lifestyle changes themselves, and local availability of such programs was highly heterogenous. SPs favored pharmacotherapy based on mechanism of action adapted to the stage of the disease, including for early stages such as steatohepatitis with mild fibrosis. CONCLUSIONS This international survey revealed major deficiencies and delays in referral pathways, suboptimal screening for comorbidities or managing of lifestyle modifications by SPs, and limited local availability for nonpharmacological interventions. Monitoring practices are not aligned with current guidelines.
Collapse
Affiliation(s)
- Vlad Ratziu
- Sorbonne Université, Hospital Pitié-SalpêtrièreParisFrance.,Institute for Cardiometabolism and Nutrition (ICAN)ParisFrance
| | - Quentin M Anstee
- 5994Translational and Clinical Research InstituteFaculty of Medical SciencesNewcastle UniversityNewcastle upon TyneUK.,Newcastle NIHR Biomedical Research CentreNewcastle upon Tyne Hospitals NHS Foundation TrustNewcastle upon TyneUK
| | | | - Jörn M Schattenberg
- Metabolic Liver Research Program, I. Department of MedicineUniversity Medical CenterMainzGermany
| | - Elisabetta Bugianesi
- Division of GastroenterologyDepartment of Medical SciencesUniversity of TurinTurinItaly
| | - Salvador Augustin
- Liver UnitDepartment of Internal MedicineVall d'Hebron Hospital UniversitariVall d'Hebron Institut de RecercaVall d'Hebron Barcelona HospitalUniversitat Autònoma de BarcelonaCIBERehdBarcelonaSpain
| | - Liana Gheorghe
- Center for Digestive Diseases and Liver TransplantationFundeni Clinical InstituteCarol Davila University of Medicine and PharmacyBucharestRomania
| | - Vittoria Zambon
- Institute for Cardiometabolism and Nutrition (ICAN)ParisFrance
| | - Nancy Reau
- Section of HepatologyRush University Medical CenterChicagoIllinoisUSA
| |
Collapse
|
99
|
Konings MCJM, Baumgartner S, Mensink RP, Plat J. Investigating microRNAs to Explain the Link between Cholesterol Metabolism and NAFLD in Humans: A Systematic Review. Nutrients 2022; 14:nu14234946. [PMID: 36500981 PMCID: PMC9738374 DOI: 10.3390/nu14234946] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2022] [Revised: 11/17/2022] [Accepted: 11/18/2022] [Indexed: 11/24/2022] Open
Abstract
Non-Alcoholic Fatty Liver Disease (NAFLD) is characterized by hepatic free cholesterol accumulation. In addition, microRNAs (miRNAs) might be involved in NAFLD development. Therefore, we systematically reviewed the literature to examine the link between miRNAs and cholesterol metabolism in NAFLD. Nineteen studies were retrieved by a systematic search in September 2022. From these papers, we evaluated associations between 13 miRNAs with NAFLD and cholesterol metabolism. Additionally, their diagnostic potential was examined. Four miRNAs (miR122, 34a, 132 and 21) were associated with cholesterol metabolism and markers for NAFLD. MiR122 was upregulated in serum of NAFLD patients, increased with disease severity and correlated with HDL-C, TAG, VLDL-C, AST, ALT, ALP, lobular inflammation, hepatocellular ballooning and NAFLD score. Serum and hepatic levels also correlated. Serum and hepatic miR34a levels were increased in NAFLD, and correlated with VLDL-C and TAG. Serum miR379 was also higher in NAFLD, especially in early stages, while miR21 gave ambiguous results. The diagnostic properties of these miRNAs were comparable to those of existing biomarkers. However, serum miR122 levels appeared to be elevated before increases in ALT and AST were evident. In conclusion, miR122, miR34a, miR21 and miR132 may play a role in the development of NAFLD via effects on cholesterol metabolism. Furthermore, it needs to be explored if miRNAs 122, 34a and 379 could be used as part of a panel in addition to established biomarkers in early detection of NAFLD.
Collapse
|
100
|
Buzova D, Braghini MR, Bianco SD, Lo Re O, Raffaele M, Frohlich J, Kisheva A, Crudele A, Mosca A, Sartorelli MR, Balsano C, Cerveny J, Mazza T, Alisi A, Vinciguerra M. Profiling of cell-free DNA methylation and histone signatures in pediatric NAFLD: A pilot study. Hepatol Commun 2022; 6:3311-3323. [PMID: 36264206 PMCID: PMC9701487 DOI: 10.1002/hep4.2082] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/05/2022] [Revised: 07/19/2022] [Accepted: 08/10/2022] [Indexed: 01/21/2023] Open
Abstract
Nonalcoholic fatty liver disease (NAFLD) has become the most common chronic liver disease in children and adolescents, increasing the risk of its progression toward nonalcoholic steatohepatitis (NASH), cirrhosis, and cancer. There is an urgent need for noninvasive early diagnostic and prognostic tools such as epigenetic marks (epimarks), which would replace liver biopsy in the future. We used plasma samples from 67 children with biopsy-proven NAFLD, and as controls we used samples from 20 children negative for steatosis by ultrasound. All patients were genotyped for patatin-like phospholipase domain containing 3 (PNPLA3), transmembrane 6 superfamily member 2 (TM6SF2), membrane bound O-acyltransferase domain containing 7 (MBOAT7), and klotho-β (KLB) gene variants, and data on anthropometric and biochemical parameters were collected. Furthermore, plasma cell-free DNA (cfDNA) methylation was quantified using a commercially available kit, and ImageStream(X) was used for the detection of free circulating histone complexes and variants. We found a significant enrichment of the levels of histone macroH2A1.2 in the plasma of children with NAFLD compared to controls, and a strong correlation between cfDNA methylation levels and NASH. Receiver operating characteristic curve analysis demonstrated that combination of cfDNA methylation, PNPLA3 rs738409 variant, coupled with either high-density lipoprotein cholesterol or alanine aminotransferase levels can strongly predict the progression of pediatric NAFLD to NASH with area under the curve >0.87. Conclusion: Our pilot study combined epimarks and genetic and metabolic markers for a robust risk assessment of NAFLD development and progression in children, offering a promising noninvasive tool for the consistent diagnosis and prognosis of pediatric NAFLD. Further studies are necessary to identify their pathogenic origin and function.
Collapse
Affiliation(s)
- Diana Buzova
- Department of Adaptive BiotechnologiesGlobal Change Research Institute CASBrnoCzech Republic
| | - Maria Rita Braghini
- Unit of Molecular Genetics of Complex PhenotypesBambino Gesù Children's Hospital, IRCCSRomeItaly
| | - Salvatore Daniele Bianco
- Laboratory of BioinformaticsFondazione IRCCS Casa Sollievo della SofferenzaSan Giovanni Rotondo (FG)Italy
| | - Oriana Lo Re
- International Clinical Research CenterSt. Anne's University HospitalBrnoCzech Republic,Department of Translational Stem Cell BiologyResearch Institute of the Medical University of VarnaVarnaBulgaria
| | - Marco Raffaele
- International Clinical Research CenterSt. Anne's University HospitalBrnoCzech Republic
| | - Jan Frohlich
- International Clinical Research CenterSt. Anne's University HospitalBrnoCzech Republic
| | - Antoniya Kisheva
- Department of Internal Diseases IMedical University of VarnaVarnaBulgaria
| | - Annalisa Crudele
- Unit of Molecular Genetics of Complex PhenotypesBambino Gesù Children's Hospital, IRCCSRomeItaly
| | - Antonella Mosca
- Hepatology, Gastroenterology and Nutrition UnitBambino Gesù Children's Hospital, IRCCSRomeItaly
| | - Maria Rita Sartorelli
- Hepatology, Gastroenterology and Nutrition UnitBambino Gesù Children's Hospital, IRCCSRomeItaly
| | - Clara Balsano
- Department of LifeHealth & Environmental Sciences‐ MESVA‐School of Emergency and Urgency Medicine, University of L'AquilaL'AquilaItaly
| | - Jan Cerveny
- Department of Adaptive BiotechnologiesGlobal Change Research Institute CASBrnoCzech Republic
| | - Tommaso Mazza
- Laboratory of BioinformaticsFondazione IRCCS Casa Sollievo della SofferenzaSan Giovanni Rotondo (FG)Italy
| | - Anna Alisi
- Unit of Molecular Genetics of Complex PhenotypesBambino Gesù Children's Hospital, IRCCSRomeItaly
| | - Manlio Vinciguerra
- International Clinical Research CenterSt. Anne's University HospitalBrnoCzech Republic,Department of Translational Stem Cell BiologyResearch Institute of the Medical University of VarnaVarnaBulgaria,Liverpool Center for Cardiovascular ScienceLiverpool Johns Moore UniversityLiverpoolUK
| |
Collapse
|