1
|
Li W, Wang F, Li Z, Feng W, Huang H, Kwan MP, Tse LA. Lipid profile and non-alcoholic fatty liver disease detected by ultrasonography: is systemic inflammation a necessary mediator? Ann Med 2025; 57:2480250. [PMID: 40098359 PMCID: PMC11921154 DOI: 10.1080/07853890.2025.2480250] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/10/2024] [Revised: 02/11/2025] [Accepted: 03/05/2025] [Indexed: 03/19/2025] Open
Abstract
AIMS To examine the relationship between lipid profile and non-alcoholic fatty liver (NAFL), compare the predictive strengths of different lipid indicators to NAFL, and explore the possible mechanisms. METHODS Male workers from a baseline survey of a cohort of workers in southern China were included. Basic information was collected through face-to-face interviews. Plasma concentrations of fasting plasma glucose (FPG), total cholesterol (TC), triglycerides (TG), high-density lipoprotein cholesterol (HDL-C), and low-density lipoprotein cholesterol (LDL-C) were determined using a blood biochemical analyzer. Liver sonography was used to identify NAFL cases. Regression models were used to calculate ORs, and examine the association between C-reactive protein (CRP) levels and lipid profiles. Restricted cubic spline regression with four knots was used to examine the dose-response relationship, and mediation analysis was employed to examine the mediation effect. RESULTS h Among the 4016 male workers, 829 (20.64%) were diagnosed with NAFL. Compared with normal lipid profile, individuals with abnormal lipid profile had higher prevalence of NAFL (OR=2.27, 95%CI: 1.85-2.79 for TG; OR=1.45, 95%CI: 1.03-2.04 for TC; OR=1.56, 95%CI: 1.21-2.02 for HDL; OR=1.65, 95%CI: 1.25-2.18 for LDL; OR=2.28, 95%CI: 1.87-2.77 for dyslipidaemia) after adjusting for potential confounders. Dose-response relationships were observed among TG, HDL, and NAFL. In addition, no significant mediation effect of C-reactive protein (CRP) was found in the association between lipid profiles and NAFL. CONCLUSIONS Abnormal TG, TC, HDL, and LDL levels were all positively associated with NAFL, while CRP has no mediating effect, and TG tended to be a better predictor of NAFL.
Collapse
Affiliation(s)
- Wenzhen Li
- Jockey Club School of Public Health and Primary Care, the Chinese University of Hong Kong, Hong Kong SAR, China
- CUHK Centre for Public Health and Primary Care (Shenzhen), Shenzhen Municipal Key Laboratory for Health Risk Analysis, Shenzhen Research Institute of the Chinese University of Hong Kong, Shenzhen, China
| | - Feng Wang
- Jockey Club School of Public Health and Primary Care, the Chinese University of Hong Kong, Hong Kong SAR, China
| | - Zhimin Li
- Institute of Occupational Medicine, Shenzhen Prevention and Treatment Center for Occupational Diseases, Shenzhen, China
| | - Wenting Feng
- Institute of Occupational Medicine, Shenzhen Prevention and Treatment Center for Occupational Diseases, Shenzhen, China
| | - Hongying Huang
- Institute of Occupational Medicine, Shenzhen Prevention and Treatment Center for Occupational Diseases, Shenzhen, China
| | - Mei-Po Kwan
- Department of Geography and Resource Management, The Chinese University of Hong Kong, Hong Kong SAS, China
- Institute of Space and Earth Information Science, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Lap Ah Tse
- Jockey Club School of Public Health and Primary Care, the Chinese University of Hong Kong, Hong Kong SAR, China
- CUHK Centre for Public Health and Primary Care (Shenzhen), Shenzhen Municipal Key Laboratory for Health Risk Analysis, Shenzhen Research Institute of the Chinese University of Hong Kong, Shenzhen, China
- Institute of Space and Earth Information Science, The Chinese University of Hong Kong, Hong Kong SAR, China
| |
Collapse
|
2
|
Wang J, Bao S, An Q, Li C, Feng J. Roles of extracellular vesicles from different origins in metabolic-associated fatty liver disease: progress and perspectives. Front Immunol 2025; 16:1544012. [PMID: 40129979 PMCID: PMC11930831 DOI: 10.3389/fimmu.2025.1544012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2024] [Accepted: 02/19/2025] [Indexed: 03/26/2025] Open
Abstract
Metabolic-Associated Fatty Liver Disease (MAFLD) is the most common chronic liver disease worldwide, associated with systemic metabolic dysregulation. It can progress from simple hepatic steatosis (MAFL) to more severe conditions like Metabolic-Associated Steatohepatitis (MASH), fibrosis, cirrhosis, and Hepatocellular Carcinoma (HCC). There is a critical lack of reliable non-invasive diagnostic methods and effective pharmaceutical treatments for MAFLD/MASH, emphasizing the need for further research. Extracellular vesicles (EVs) are nanoscale structures that play important roles in cell signaling by delivering bioactive molecules. However, there is a significant gap in literature regarding the roles of EVs from hosts, plants, and microbiota in MAFLD. This review explores the potential of EVs from various sources-host, plants, and microbiota-as biomarkers, therapeutic agents, drug carriers, and treatment targets for MAFLD. Firstly, the roles of host-derived extracellular vesicles (EVs) in MAFLD, with a focus on cell-type specific EVs and their components-proteins, miRNAs, and lipids-for disease diagnosis and monitoring were discussed. Moreover, it highlighted the therapeutic potential of mesenchymal stem cell (MSC)-derived EVs in reducing lipid accumulation and liver injury, and immune cell-derived EVs in mitigating inflammation and fibrosis. The review also discussed the use of host-derived EVs as drug carriers and therapeutic targets due to their ability to deliver bioactive molecules that impact disease mechanisms. Additionally, it summarized research on plant-derived EVs, which help reduce liver lipid accumulation, inflammation, and enhance gut barrier function in MAFLD. Also, the review explored microbial-derived EVs as novel therapeutic targets, particularly in relation to insulin resistance, liver inflammation, and dysfunction in MAFLD. Overall, by exploring the diverse roles of EVs from host, plant, and microbiota sources in MAFLD, this review offers valuable insights into their potential as non-invasive biomarkers and novel therapeutic strategies, which could pave the way for more effective diagnostic and treatment options for this increasingly prevalent liver disease. Notably, the challenges of translating EVs into clinical practice were also thoroughly discussed, aiming to provide possible directions and strategies for future research.
Collapse
Affiliation(s)
- Jing Wang
- School of Public Health, Gansu University of Chinese Medicine, Lanzhou, China
| | - Shuoqiang Bao
- School of Public Health, Gansu University of Chinese Medicine, Lanzhou, China
| | - Qi An
- School of Public Health, Gansu University of Chinese Medicine, Lanzhou, China
| | - Caihong Li
- School of Public Health, Gansu University of Chinese Medicine, Lanzhou, China
| | - Juan Feng
- College of Health Science and Environmental Engineering, Shenzhen Technology University, Shenzhen, China
| |
Collapse
|
3
|
Ratziu V. Cirrhose métabolique : une entité en plein essor. BULLETIN DE L'ACADÉMIE NATIONALE DE MÉDECINE 2024. [DOI: 10.1016/j.banm.2024.11.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
|
4
|
Hosseini Shabanan S, Martins VF, Wolfson T, Weeks JT, Ceriani L, Behling C, Chernyak V, El Kaffas A, Borhani AA, Han A, Wang K, Fowler KJ, Sirlin CB. MASLD: What We Have Learned and Where We Need to Go-A Call to Action. Radiographics 2024; 44:e240048. [PMID: 39418184 PMCID: PMC11580021 DOI: 10.1148/rg.240048] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2024] [Revised: 04/16/2024] [Accepted: 04/24/2024] [Indexed: 10/19/2024]
Abstract
Since its introduction in 1980, fatty liver disease (now termed metabolic dysfunction-associated steatotic liver disease [MASLD]) has grown in prevalence significantly, paralleling the rise of obesity worldwide. While MASLD has been the subject of extensive research leading to significant progress in the understanding of its pathophysiology and progression factors, several gaps in knowledge remain. In this pictorial review, the authors present the latest insights into MASLD, covering its recent nomenclature change, spectrum of disease, epidemiology, morbidity, and mortality. The authors also discuss current qualitative and quantitative imaging methods for assessing and monitoring MASLD. Last, they propose six unsolved challenges in MASLD assessment, which they term the proliferation, reproducibility, reporting, needle-in-the-haystack, availability, and knowledge problems. These challenges offer opportunities for the radiology community to proactively contribute to their resolution. The authors conclude with a call to action for the entire radiology community to claim a seat at the table, collaborate with other societies, and commit to advancing the development, validation, dissemination, and accessibility of the imaging technologies required to combat the looming health care crisis of MASLD.
Collapse
Affiliation(s)
| | | | - Tanya Wolfson
- From the Department of Radiology, UC San Diego Altman Clinical and
Translational Research Institute Liver Imaging Group, University of California
San Diego, 9452 Medical Center Dr, La Jolla, CA 92037 (S.H.S., V.F.M., T.W.,
J.T.W., L.C., K.J.F., C.B.S.); Pacific Rim Pathology, San Diego, Calif (C.B.);
Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY
(V.C.); Department of Radiology, Stanford University School of Medicine,
Stanford, Calif (A.E.K.); Department of Radiology, Northwestern University
Feinberg School of Medicine, Chicago, Ill (A.A.B.); Department of Biomedical
Engineering and Mechanics, Virginia Polytechnic Institute and State University,
Blacksburg, Va (A.H.); and Department of Radiology, University of California San
Francisco, Calif (K.W.)
| | - Jake T. Weeks
- From the Department of Radiology, UC San Diego Altman Clinical and
Translational Research Institute Liver Imaging Group, University of California
San Diego, 9452 Medical Center Dr, La Jolla, CA 92037 (S.H.S., V.F.M., T.W.,
J.T.W., L.C., K.J.F., C.B.S.); Pacific Rim Pathology, San Diego, Calif (C.B.);
Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY
(V.C.); Department of Radiology, Stanford University School of Medicine,
Stanford, Calif (A.E.K.); Department of Radiology, Northwestern University
Feinberg School of Medicine, Chicago, Ill (A.A.B.); Department of Biomedical
Engineering and Mechanics, Virginia Polytechnic Institute and State University,
Blacksburg, Va (A.H.); and Department of Radiology, University of California San
Francisco, Calif (K.W.)
| | - Lael Ceriani
- From the Department of Radiology, UC San Diego Altman Clinical and
Translational Research Institute Liver Imaging Group, University of California
San Diego, 9452 Medical Center Dr, La Jolla, CA 92037 (S.H.S., V.F.M., T.W.,
J.T.W., L.C., K.J.F., C.B.S.); Pacific Rim Pathology, San Diego, Calif (C.B.);
Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY
(V.C.); Department of Radiology, Stanford University School of Medicine,
Stanford, Calif (A.E.K.); Department of Radiology, Northwestern University
Feinberg School of Medicine, Chicago, Ill (A.A.B.); Department of Biomedical
Engineering and Mechanics, Virginia Polytechnic Institute and State University,
Blacksburg, Va (A.H.); and Department of Radiology, University of California San
Francisco, Calif (K.W.)
| | - Cynthia Behling
- From the Department of Radiology, UC San Diego Altman Clinical and
Translational Research Institute Liver Imaging Group, University of California
San Diego, 9452 Medical Center Dr, La Jolla, CA 92037 (S.H.S., V.F.M., T.W.,
J.T.W., L.C., K.J.F., C.B.S.); Pacific Rim Pathology, San Diego, Calif (C.B.);
Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY
(V.C.); Department of Radiology, Stanford University School of Medicine,
Stanford, Calif (A.E.K.); Department of Radiology, Northwestern University
Feinberg School of Medicine, Chicago, Ill (A.A.B.); Department of Biomedical
Engineering and Mechanics, Virginia Polytechnic Institute and State University,
Blacksburg, Va (A.H.); and Department of Radiology, University of California San
Francisco, Calif (K.W.)
| | - Victoria Chernyak
- From the Department of Radiology, UC San Diego Altman Clinical and
Translational Research Institute Liver Imaging Group, University of California
San Diego, 9452 Medical Center Dr, La Jolla, CA 92037 (S.H.S., V.F.M., T.W.,
J.T.W., L.C., K.J.F., C.B.S.); Pacific Rim Pathology, San Diego, Calif (C.B.);
Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY
(V.C.); Department of Radiology, Stanford University School of Medicine,
Stanford, Calif (A.E.K.); Department of Radiology, Northwestern University
Feinberg School of Medicine, Chicago, Ill (A.A.B.); Department of Biomedical
Engineering and Mechanics, Virginia Polytechnic Institute and State University,
Blacksburg, Va (A.H.); and Department of Radiology, University of California San
Francisco, Calif (K.W.)
| | - Ahmed El Kaffas
- From the Department of Radiology, UC San Diego Altman Clinical and
Translational Research Institute Liver Imaging Group, University of California
San Diego, 9452 Medical Center Dr, La Jolla, CA 92037 (S.H.S., V.F.M., T.W.,
J.T.W., L.C., K.J.F., C.B.S.); Pacific Rim Pathology, San Diego, Calif (C.B.);
Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY
(V.C.); Department of Radiology, Stanford University School of Medicine,
Stanford, Calif (A.E.K.); Department of Radiology, Northwestern University
Feinberg School of Medicine, Chicago, Ill (A.A.B.); Department of Biomedical
Engineering and Mechanics, Virginia Polytechnic Institute and State University,
Blacksburg, Va (A.H.); and Department of Radiology, University of California San
Francisco, Calif (K.W.)
| | - Amir A. Borhani
- From the Department of Radiology, UC San Diego Altman Clinical and
Translational Research Institute Liver Imaging Group, University of California
San Diego, 9452 Medical Center Dr, La Jolla, CA 92037 (S.H.S., V.F.M., T.W.,
J.T.W., L.C., K.J.F., C.B.S.); Pacific Rim Pathology, San Diego, Calif (C.B.);
Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY
(V.C.); Department of Radiology, Stanford University School of Medicine,
Stanford, Calif (A.E.K.); Department of Radiology, Northwestern University
Feinberg School of Medicine, Chicago, Ill (A.A.B.); Department of Biomedical
Engineering and Mechanics, Virginia Polytechnic Institute and State University,
Blacksburg, Va (A.H.); and Department of Radiology, University of California San
Francisco, Calif (K.W.)
| | - Aiguo Han
- From the Department of Radiology, UC San Diego Altman Clinical and
Translational Research Institute Liver Imaging Group, University of California
San Diego, 9452 Medical Center Dr, La Jolla, CA 92037 (S.H.S., V.F.M., T.W.,
J.T.W., L.C., K.J.F., C.B.S.); Pacific Rim Pathology, San Diego, Calif (C.B.);
Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY
(V.C.); Department of Radiology, Stanford University School of Medicine,
Stanford, Calif (A.E.K.); Department of Radiology, Northwestern University
Feinberg School of Medicine, Chicago, Ill (A.A.B.); Department of Biomedical
Engineering and Mechanics, Virginia Polytechnic Institute and State University,
Blacksburg, Va (A.H.); and Department of Radiology, University of California San
Francisco, Calif (K.W.)
| | - Kang Wang
- From the Department of Radiology, UC San Diego Altman Clinical and
Translational Research Institute Liver Imaging Group, University of California
San Diego, 9452 Medical Center Dr, La Jolla, CA 92037 (S.H.S., V.F.M., T.W.,
J.T.W., L.C., K.J.F., C.B.S.); Pacific Rim Pathology, San Diego, Calif (C.B.);
Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY
(V.C.); Department of Radiology, Stanford University School of Medicine,
Stanford, Calif (A.E.K.); Department of Radiology, Northwestern University
Feinberg School of Medicine, Chicago, Ill (A.A.B.); Department of Biomedical
Engineering and Mechanics, Virginia Polytechnic Institute and State University,
Blacksburg, Va (A.H.); and Department of Radiology, University of California San
Francisco, Calif (K.W.)
| | - Kathryn J. Fowler
- From the Department of Radiology, UC San Diego Altman Clinical and
Translational Research Institute Liver Imaging Group, University of California
San Diego, 9452 Medical Center Dr, La Jolla, CA 92037 (S.H.S., V.F.M., T.W.,
J.T.W., L.C., K.J.F., C.B.S.); Pacific Rim Pathology, San Diego, Calif (C.B.);
Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY
(V.C.); Department of Radiology, Stanford University School of Medicine,
Stanford, Calif (A.E.K.); Department of Radiology, Northwestern University
Feinberg School of Medicine, Chicago, Ill (A.A.B.); Department of Biomedical
Engineering and Mechanics, Virginia Polytechnic Institute and State University,
Blacksburg, Va (A.H.); and Department of Radiology, University of California San
Francisco, Calif (K.W.)
| | - Claude B. Sirlin
- From the Department of Radiology, UC San Diego Altman Clinical and
Translational Research Institute Liver Imaging Group, University of California
San Diego, 9452 Medical Center Dr, La Jolla, CA 92037 (S.H.S., V.F.M., T.W.,
J.T.W., L.C., K.J.F., C.B.S.); Pacific Rim Pathology, San Diego, Calif (C.B.);
Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY
(V.C.); Department of Radiology, Stanford University School of Medicine,
Stanford, Calif (A.E.K.); Department of Radiology, Northwestern University
Feinberg School of Medicine, Chicago, Ill (A.A.B.); Department of Biomedical
Engineering and Mechanics, Virginia Polytechnic Institute and State University,
Blacksburg, Va (A.H.); and Department of Radiology, University of California San
Francisco, Calif (K.W.)
| |
Collapse
|
5
|
Rauzier C, Chartrand DJ, Alméras N, Lemieux I, Larose E, Mathieu P, Pibarot P, Lamarche B, Rhéaume C, Poirier P, Després JP, Picard F. Combination of Waist Circumference and Circulating Levels of IGFBP-2 as a Simple Screening Tool for Early Detection of Metabolic Dysfunction-Associated Steatotic Liver Disease. Diabetes Metab Syndr Obes 2024; 17:3335-3341. [PMID: 39252871 PMCID: PMC11382664 DOI: 10.2147/dmso.s466051] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/27/2024] [Accepted: 07/06/2024] [Indexed: 09/11/2024] Open
Abstract
Optimal non-invasive biomarkers for metabolic dysfunction-associated steatotic liver disease (MASLD) remain elusive, especially in the detection of early stages. This study tested in an asymptomatic cohort of 171 men (49.2 ± 8.6 years) and 131 women (51.8 ± 8.5 years) whether waist circumference (WC) and circulating levels of insulin-like growth factor-binding protein 2 (IGFBP-2) could identify individuals with liver fat >5% as assessed by magnetic resonance spectroscopy. Participants with high WC (> 85 or 90 cm for women and men, respectively) and low IGFBP-2 (< 260 or 230 ng/mL for women and men, respectively) were characterized by a higher risk of having MASLD (46.3%, p < 0.0001). Among the 68 individuals with MASLD, 73.5% fell into the subgroup with high WC and low IGFBP-2 concentrations (p < 0.0001). When combined, these markers reached a sensitivity of 73.5% and specificity of 75.2% for MASLD. Thus, WC and plasma IGFBP-2 levels might be useful as a novel, simple, and non-invasive index to support existing tools in the identification of individuals at risk of early-stage MASLD.
Collapse
Affiliation(s)
- Chloé Rauzier
- Centre de recherche de l'Institut universitaire de cardiologie et de pneumologie de Québec (IUCPQ) - Université Laval, Québec, QC, Canada
- Faculté de pharmacie, Université Laval, Québec, QC, Canada
| | - Dominic J Chartrand
- Centre de recherche de l'Institut universitaire de cardiologie et de pneumologie de Québec (IUCPQ) - Université Laval, Québec, QC, Canada
- Département de kinésiologie, Faculté de médecine, Université Laval, Québec, QC, Canada
| | - Natalie Alméras
- Centre de recherche de l'Institut universitaire de cardiologie et de pneumologie de Québec (IUCPQ) - Université Laval, Québec, QC, Canada
- Département de kinésiologie, Faculté de médecine, Université Laval, Québec, QC, Canada
| | - Isabelle Lemieux
- Centre de recherche de l'Institut universitaire de cardiologie et de pneumologie de Québec (IUCPQ) - Université Laval, Québec, QC, Canada
| | - Eric Larose
- Centre de recherche de l'Institut universitaire de cardiologie et de pneumologie de Québec (IUCPQ) - Université Laval, Québec, QC, Canada
- Département de médecine, Faculté de médecine, Université Laval, Québec, QC, Canada
| | - Patrick Mathieu
- Centre de recherche de l'Institut universitaire de cardiologie et de pneumologie de Québec (IUCPQ) - Université Laval, Québec, QC, Canada
- Département de chirurgie, Faculté de médecine, Université Laval, Québec, QC, Canada
| | - Philippe Pibarot
- Centre de recherche de l'Institut universitaire de cardiologie et de pneumologie de Québec (IUCPQ) - Université Laval, Québec, QC, Canada
- Département de médecine, Faculté de médecine, Université Laval, Québec, QC, Canada
| | - Benoît Lamarche
- Centre Nutrition, santé et société (NUTRISS), Institut sur la nutrition et les aliments fonctionnels (INAF), Université Laval, Québec, QC, Canada
- École de nutrition, Faculté des sciences de l'agriculture et de l'alimentation, Université Laval, Québec, QC, Canada
| | - Caroline Rhéaume
- Centre de recherche de l'Institut universitaire de cardiologie et de pneumologie de Québec (IUCPQ) - Université Laval, Québec, QC, Canada
- Département de médecine familiale et de médecine d'urgence, Faculté de médecine, Université Laval, Québec, QC, Canada
- VITAM - Centre de recherche en santé durable, Québec, QC, Canada
| | - Paul Poirier
- Centre de recherche de l'Institut universitaire de cardiologie et de pneumologie de Québec (IUCPQ) - Université Laval, Québec, QC, Canada
- Faculté de pharmacie, Université Laval, Québec, QC, Canada
| | - Jean-Pierre Després
- Centre de recherche de l'Institut universitaire de cardiologie et de pneumologie de Québec (IUCPQ) - Université Laval, Québec, QC, Canada
- Département de kinésiologie, Faculté de médecine, Université Laval, Québec, QC, Canada
- VITAM - Centre de recherche en santé durable, Québec, QC, Canada
| | - Frédéric Picard
- Centre de recherche de l'Institut universitaire de cardiologie et de pneumologie de Québec (IUCPQ) - Université Laval, Québec, QC, Canada
- Faculté de pharmacie, Université Laval, Québec, QC, Canada
| |
Collapse
|
6
|
Zannad F, Sanyal AJ, Butler J, Miller V, Harrison SA. Integrating liver endpoints in clinical trials of cardiovascular and kidney disease. Nat Med 2024; 30:2423-2431. [PMID: 39227442 DOI: 10.1038/s41591-024-03223-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2024] [Accepted: 07/30/2024] [Indexed: 09/05/2024]
Abstract
The intersection of cardiovascular disease, metabolic disorders and chronic kidney disease represents a complex clinical picture challenging healthcare systems worldwide. Metabolic-dysfunction-associated steatotic liver disease (MASLD) often manifests sequentially or concomitantly with these diseases, and may share underlying mechanisms and risk factors. Growing evidence suggests that new therapies could have benefits across these diseases, but trial sponsors and investigators tend to be reluctant to include patients with comorbidities-particularly liver diseases-in clinical trials. In this Perspective, we call for inclusion of patients with MASLD and measurement of liver outcomes in cardio-kidney-metabolic trials, when data suggest mechanistically plausible benefits and liver and cardiovascular safety. We discuss the implications of this new paradigm for clinical trial design and considerations for regulatory approval. Finally, we outline the challenges to implementing such an approach and provide recommendations for future clinical trial conduct.
Collapse
Affiliation(s)
- Faiez Zannad
- Université de Lorraine, Inserm Clinical Investigation Center at Institut Lorrain du Coeur et des Vaisseaux, University Hospital of Nancy, Nancy, France.
| | - Arun J Sanyal
- Department of Internal Medicine, Virginia Commonwealth University School of Medicine, Richmond, VA, USA
| | - Javed Butler
- Baylor Scott and White Research Institute, Dallas, TX, USA
- University of Mississippi, Jackson, MS, USA
| | - Veronica Miller
- Forum for Collaborative Research, Washington DC; University of California Berkeley School of Public Health, Berkeley, CA, USA
| | - Stephen A Harrison
- Visiting Professor of Hepatology Radcliffe Department of Medicine, University of Oxford, Oxford, UK
| |
Collapse
|
7
|
Pierce TT, Ozturk A, Sherlock SP, Moura Cunha G, Wang X, Li Q, Hunt D, Middleton MS, Martin M, Corey KE, Edenbaum H, Shankar SS, Heymann H, Kamphaus TN, Calle RA, Covarrubias Y, Loomba R, Obuchowski NA, Sanyal AJ, Sirlin CB, Fowler KJ, Samir AE. Reproducibility and Repeatability of US Shear-Wave and Transient Elastography in Nonalcoholic Fatty Liver Disease. Radiology 2024; 312:e233094. [PMID: 39254458 PMCID: PMC11427856 DOI: 10.1148/radiol.233094] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2023] [Revised: 05/13/2024] [Accepted: 06/03/2024] [Indexed: 09/11/2024]
Abstract
Background US shear-wave elastography (SWE) and vibration-controlled transient elastography (VCTE) enable assessment of liver stiffness, an indicator of fibrosis severity. However, limited reproducibility data restrict their use in clinical trials. Purpose To estimate SWE and VCTE measurement variability in nonalcoholic fatty liver disease (NAFLD) within and across systems to support clinical trial diagnostic enrichment and clinical interpretation of longitudinal liver stiffness. Materials and Methods This prospective, observational, cross-sectional study (March 2021 to November 2021) enrolled adults with NAFLD, stratified according to the Fibrosis-4 (FIB-4) index (≤1.3, >1.3 and <2.67, ≥2.67), at two sites to assess SWE with five US systems and VCTE with one system. Each participant underwent 12 elastography examinations over two separate days within 1 week, with each day's examinations conducted by a different operator. VCTE and SWE measurements were reported in units of meters per second. The primary end point was the different-day, different-operator reproducibility coefficient (RDCDDDO) pooled across systems for SWE and individually for VCTE. Secondary end points included system-specific RDCDDDO, same-day, same-operator repeatability coefficient (RCSDSO), and between-system same-day, same-operator reproducibility coefficient. The planned sample provided 80% power to detect a pooled RDCDDDO of less than 35%, the prespecified performance threshold. Results A total of 40 participants (mean age, 60 years ± 10 [SD]; 24 women) with low (n = 17), intermediate (n = 15), and high (n = 8) FIB-4 scores were enrolled. RDCDDDO was 30.7% (95% upper bound, 34.4%) for SWE and 35.6% (95% upper bound, 43.9%) for VCTE. SWE system-specific RDCDDDO varied from 24.2% to 34.3%. The RCSDSO was 21.0% for SWE (range, 13.9%-35.0%) and 19.6% for VCTE. The SWE between-system same-day, same-operator reproducibility coefficient was 52.7%. Conclusion SWE met the prespecified threshold, RDCDDDO less than 35%, with VCTE having a higher RDCDDDO. SWE variability was higher between different systems. These estimates advance liver US-based noninvasive test qualification by (a) defining expected variability, (b) establishing that serial examination variability is lower when performed with the same system, and (c) informing clinical trial design. ClinicalTrials.gov Identifier NCT04828551 © RSNA, 2024 Supplemental material is available for this article.
Collapse
Affiliation(s)
- Theodore T Pierce
- From the Center for Ultrasound Research and Translation, Massachusetts General Hospital, 55 Fruit St, White Bldg, Rm 270, Boston, MA 02114 (T.T.P., A.O., X.W., Q.L., D.H., M.M., H.E., A.E.S.); Harvard Medical School, Boston, Mass (T.T.P., A.O., Q.L., A.E.S.); Pfizer, Cambridge, Mass (S.P.S.); Department of Radiology, University of Washington, Seattle, Wash (G.M.C.); Department of Ultrasound, Shenzhen University General Hospital, Shenzhen, China (Q.L.); Department of Radiology, Liver Imaging Group, University of California San Diego, La Jolla, Calif (M.S.M., Y.C., C.B.S., K.J.F.); MGH Fatty Liver Program, Gastrointestinal Unit, Massachusetts General Hospital, Boston, Mass (K.E.C.); BioAge Labs, Richmond, Calif (S.S.S.); Foundation for the National Institutes of Health, North Bethesda, Md (H.H., T.N.K.); Regeneron Pharmaceuticals, Tarrytown, NY (R.A.C.); Department of Medicine, Division of Gastroenterology, NAFLD Research Center, University of California at San Diego, La Jolla, Calif (R.L.); Quantitative Health Sciences, Cleveland Clinic Foundation, Cleveland, Ohio (N.A.O.); and Department of Internal Medicine, Division of Internal Medicine, Division of Gastroenterology, Virginia Commonwealth University Medical Center, Richmond, Va (A.J.S.)
| | - Arinc Ozturk
- From the Center for Ultrasound Research and Translation, Massachusetts General Hospital, 55 Fruit St, White Bldg, Rm 270, Boston, MA 02114 (T.T.P., A.O., X.W., Q.L., D.H., M.M., H.E., A.E.S.); Harvard Medical School, Boston, Mass (T.T.P., A.O., Q.L., A.E.S.); Pfizer, Cambridge, Mass (S.P.S.); Department of Radiology, University of Washington, Seattle, Wash (G.M.C.); Department of Ultrasound, Shenzhen University General Hospital, Shenzhen, China (Q.L.); Department of Radiology, Liver Imaging Group, University of California San Diego, La Jolla, Calif (M.S.M., Y.C., C.B.S., K.J.F.); MGH Fatty Liver Program, Gastrointestinal Unit, Massachusetts General Hospital, Boston, Mass (K.E.C.); BioAge Labs, Richmond, Calif (S.S.S.); Foundation for the National Institutes of Health, North Bethesda, Md (H.H., T.N.K.); Regeneron Pharmaceuticals, Tarrytown, NY (R.A.C.); Department of Medicine, Division of Gastroenterology, NAFLD Research Center, University of California at San Diego, La Jolla, Calif (R.L.); Quantitative Health Sciences, Cleveland Clinic Foundation, Cleveland, Ohio (N.A.O.); and Department of Internal Medicine, Division of Internal Medicine, Division of Gastroenterology, Virginia Commonwealth University Medical Center, Richmond, Va (A.J.S.)
| | - Sarah P Sherlock
- From the Center for Ultrasound Research and Translation, Massachusetts General Hospital, 55 Fruit St, White Bldg, Rm 270, Boston, MA 02114 (T.T.P., A.O., X.W., Q.L., D.H., M.M., H.E., A.E.S.); Harvard Medical School, Boston, Mass (T.T.P., A.O., Q.L., A.E.S.); Pfizer, Cambridge, Mass (S.P.S.); Department of Radiology, University of Washington, Seattle, Wash (G.M.C.); Department of Ultrasound, Shenzhen University General Hospital, Shenzhen, China (Q.L.); Department of Radiology, Liver Imaging Group, University of California San Diego, La Jolla, Calif (M.S.M., Y.C., C.B.S., K.J.F.); MGH Fatty Liver Program, Gastrointestinal Unit, Massachusetts General Hospital, Boston, Mass (K.E.C.); BioAge Labs, Richmond, Calif (S.S.S.); Foundation for the National Institutes of Health, North Bethesda, Md (H.H., T.N.K.); Regeneron Pharmaceuticals, Tarrytown, NY (R.A.C.); Department of Medicine, Division of Gastroenterology, NAFLD Research Center, University of California at San Diego, La Jolla, Calif (R.L.); Quantitative Health Sciences, Cleveland Clinic Foundation, Cleveland, Ohio (N.A.O.); and Department of Internal Medicine, Division of Internal Medicine, Division of Gastroenterology, Virginia Commonwealth University Medical Center, Richmond, Va (A.J.S.)
| | - Guilherme Moura Cunha
- From the Center for Ultrasound Research and Translation, Massachusetts General Hospital, 55 Fruit St, White Bldg, Rm 270, Boston, MA 02114 (T.T.P., A.O., X.W., Q.L., D.H., M.M., H.E., A.E.S.); Harvard Medical School, Boston, Mass (T.T.P., A.O., Q.L., A.E.S.); Pfizer, Cambridge, Mass (S.P.S.); Department of Radiology, University of Washington, Seattle, Wash (G.M.C.); Department of Ultrasound, Shenzhen University General Hospital, Shenzhen, China (Q.L.); Department of Radiology, Liver Imaging Group, University of California San Diego, La Jolla, Calif (M.S.M., Y.C., C.B.S., K.J.F.); MGH Fatty Liver Program, Gastrointestinal Unit, Massachusetts General Hospital, Boston, Mass (K.E.C.); BioAge Labs, Richmond, Calif (S.S.S.); Foundation for the National Institutes of Health, North Bethesda, Md (H.H., T.N.K.); Regeneron Pharmaceuticals, Tarrytown, NY (R.A.C.); Department of Medicine, Division of Gastroenterology, NAFLD Research Center, University of California at San Diego, La Jolla, Calif (R.L.); Quantitative Health Sciences, Cleveland Clinic Foundation, Cleveland, Ohio (N.A.O.); and Department of Internal Medicine, Division of Internal Medicine, Division of Gastroenterology, Virginia Commonwealth University Medical Center, Richmond, Va (A.J.S.)
| | - Xiaohong Wang
- From the Center for Ultrasound Research and Translation, Massachusetts General Hospital, 55 Fruit St, White Bldg, Rm 270, Boston, MA 02114 (T.T.P., A.O., X.W., Q.L., D.H., M.M., H.E., A.E.S.); Harvard Medical School, Boston, Mass (T.T.P., A.O., Q.L., A.E.S.); Pfizer, Cambridge, Mass (S.P.S.); Department of Radiology, University of Washington, Seattle, Wash (G.M.C.); Department of Ultrasound, Shenzhen University General Hospital, Shenzhen, China (Q.L.); Department of Radiology, Liver Imaging Group, University of California San Diego, La Jolla, Calif (M.S.M., Y.C., C.B.S., K.J.F.); MGH Fatty Liver Program, Gastrointestinal Unit, Massachusetts General Hospital, Boston, Mass (K.E.C.); BioAge Labs, Richmond, Calif (S.S.S.); Foundation for the National Institutes of Health, North Bethesda, Md (H.H., T.N.K.); Regeneron Pharmaceuticals, Tarrytown, NY (R.A.C.); Department of Medicine, Division of Gastroenterology, NAFLD Research Center, University of California at San Diego, La Jolla, Calif (R.L.); Quantitative Health Sciences, Cleveland Clinic Foundation, Cleveland, Ohio (N.A.O.); and Department of Internal Medicine, Division of Internal Medicine, Division of Gastroenterology, Virginia Commonwealth University Medical Center, Richmond, Va (A.J.S.)
| | - Qian Li
- From the Center for Ultrasound Research and Translation, Massachusetts General Hospital, 55 Fruit St, White Bldg, Rm 270, Boston, MA 02114 (T.T.P., A.O., X.W., Q.L., D.H., M.M., H.E., A.E.S.); Harvard Medical School, Boston, Mass (T.T.P., A.O., Q.L., A.E.S.); Pfizer, Cambridge, Mass (S.P.S.); Department of Radiology, University of Washington, Seattle, Wash (G.M.C.); Department of Ultrasound, Shenzhen University General Hospital, Shenzhen, China (Q.L.); Department of Radiology, Liver Imaging Group, University of California San Diego, La Jolla, Calif (M.S.M., Y.C., C.B.S., K.J.F.); MGH Fatty Liver Program, Gastrointestinal Unit, Massachusetts General Hospital, Boston, Mass (K.E.C.); BioAge Labs, Richmond, Calif (S.S.S.); Foundation for the National Institutes of Health, North Bethesda, Md (H.H., T.N.K.); Regeneron Pharmaceuticals, Tarrytown, NY (R.A.C.); Department of Medicine, Division of Gastroenterology, NAFLD Research Center, University of California at San Diego, La Jolla, Calif (R.L.); Quantitative Health Sciences, Cleveland Clinic Foundation, Cleveland, Ohio (N.A.O.); and Department of Internal Medicine, Division of Internal Medicine, Division of Gastroenterology, Virginia Commonwealth University Medical Center, Richmond, Va (A.J.S.)
| | - David Hunt
- From the Center for Ultrasound Research and Translation, Massachusetts General Hospital, 55 Fruit St, White Bldg, Rm 270, Boston, MA 02114 (T.T.P., A.O., X.W., Q.L., D.H., M.M., H.E., A.E.S.); Harvard Medical School, Boston, Mass (T.T.P., A.O., Q.L., A.E.S.); Pfizer, Cambridge, Mass (S.P.S.); Department of Radiology, University of Washington, Seattle, Wash (G.M.C.); Department of Ultrasound, Shenzhen University General Hospital, Shenzhen, China (Q.L.); Department of Radiology, Liver Imaging Group, University of California San Diego, La Jolla, Calif (M.S.M., Y.C., C.B.S., K.J.F.); MGH Fatty Liver Program, Gastrointestinal Unit, Massachusetts General Hospital, Boston, Mass (K.E.C.); BioAge Labs, Richmond, Calif (S.S.S.); Foundation for the National Institutes of Health, North Bethesda, Md (H.H., T.N.K.); Regeneron Pharmaceuticals, Tarrytown, NY (R.A.C.); Department of Medicine, Division of Gastroenterology, NAFLD Research Center, University of California at San Diego, La Jolla, Calif (R.L.); Quantitative Health Sciences, Cleveland Clinic Foundation, Cleveland, Ohio (N.A.O.); and Department of Internal Medicine, Division of Internal Medicine, Division of Gastroenterology, Virginia Commonwealth University Medical Center, Richmond, Va (A.J.S.)
| | - Michael S Middleton
- From the Center for Ultrasound Research and Translation, Massachusetts General Hospital, 55 Fruit St, White Bldg, Rm 270, Boston, MA 02114 (T.T.P., A.O., X.W., Q.L., D.H., M.M., H.E., A.E.S.); Harvard Medical School, Boston, Mass (T.T.P., A.O., Q.L., A.E.S.); Pfizer, Cambridge, Mass (S.P.S.); Department of Radiology, University of Washington, Seattle, Wash (G.M.C.); Department of Ultrasound, Shenzhen University General Hospital, Shenzhen, China (Q.L.); Department of Radiology, Liver Imaging Group, University of California San Diego, La Jolla, Calif (M.S.M., Y.C., C.B.S., K.J.F.); MGH Fatty Liver Program, Gastrointestinal Unit, Massachusetts General Hospital, Boston, Mass (K.E.C.); BioAge Labs, Richmond, Calif (S.S.S.); Foundation for the National Institutes of Health, North Bethesda, Md (H.H., T.N.K.); Regeneron Pharmaceuticals, Tarrytown, NY (R.A.C.); Department of Medicine, Division of Gastroenterology, NAFLD Research Center, University of California at San Diego, La Jolla, Calif (R.L.); Quantitative Health Sciences, Cleveland Clinic Foundation, Cleveland, Ohio (N.A.O.); and Department of Internal Medicine, Division of Internal Medicine, Division of Gastroenterology, Virginia Commonwealth University Medical Center, Richmond, Va (A.J.S.)
| | - Marian Martin
- From the Center for Ultrasound Research and Translation, Massachusetts General Hospital, 55 Fruit St, White Bldg, Rm 270, Boston, MA 02114 (T.T.P., A.O., X.W., Q.L., D.H., M.M., H.E., A.E.S.); Harvard Medical School, Boston, Mass (T.T.P., A.O., Q.L., A.E.S.); Pfizer, Cambridge, Mass (S.P.S.); Department of Radiology, University of Washington, Seattle, Wash (G.M.C.); Department of Ultrasound, Shenzhen University General Hospital, Shenzhen, China (Q.L.); Department of Radiology, Liver Imaging Group, University of California San Diego, La Jolla, Calif (M.S.M., Y.C., C.B.S., K.J.F.); MGH Fatty Liver Program, Gastrointestinal Unit, Massachusetts General Hospital, Boston, Mass (K.E.C.); BioAge Labs, Richmond, Calif (S.S.S.); Foundation for the National Institutes of Health, North Bethesda, Md (H.H., T.N.K.); Regeneron Pharmaceuticals, Tarrytown, NY (R.A.C.); Department of Medicine, Division of Gastroenterology, NAFLD Research Center, University of California at San Diego, La Jolla, Calif (R.L.); Quantitative Health Sciences, Cleveland Clinic Foundation, Cleveland, Ohio (N.A.O.); and Department of Internal Medicine, Division of Internal Medicine, Division of Gastroenterology, Virginia Commonwealth University Medical Center, Richmond, Va (A.J.S.)
| | - Kathleen E Corey
- From the Center for Ultrasound Research and Translation, Massachusetts General Hospital, 55 Fruit St, White Bldg, Rm 270, Boston, MA 02114 (T.T.P., A.O., X.W., Q.L., D.H., M.M., H.E., A.E.S.); Harvard Medical School, Boston, Mass (T.T.P., A.O., Q.L., A.E.S.); Pfizer, Cambridge, Mass (S.P.S.); Department of Radiology, University of Washington, Seattle, Wash (G.M.C.); Department of Ultrasound, Shenzhen University General Hospital, Shenzhen, China (Q.L.); Department of Radiology, Liver Imaging Group, University of California San Diego, La Jolla, Calif (M.S.M., Y.C., C.B.S., K.J.F.); MGH Fatty Liver Program, Gastrointestinal Unit, Massachusetts General Hospital, Boston, Mass (K.E.C.); BioAge Labs, Richmond, Calif (S.S.S.); Foundation for the National Institutes of Health, North Bethesda, Md (H.H., T.N.K.); Regeneron Pharmaceuticals, Tarrytown, NY (R.A.C.); Department of Medicine, Division of Gastroenterology, NAFLD Research Center, University of California at San Diego, La Jolla, Calif (R.L.); Quantitative Health Sciences, Cleveland Clinic Foundation, Cleveland, Ohio (N.A.O.); and Department of Internal Medicine, Division of Internal Medicine, Division of Gastroenterology, Virginia Commonwealth University Medical Center, Richmond, Va (A.J.S.)
| | - Hannah Edenbaum
- From the Center for Ultrasound Research and Translation, Massachusetts General Hospital, 55 Fruit St, White Bldg, Rm 270, Boston, MA 02114 (T.T.P., A.O., X.W., Q.L., D.H., M.M., H.E., A.E.S.); Harvard Medical School, Boston, Mass (T.T.P., A.O., Q.L., A.E.S.); Pfizer, Cambridge, Mass (S.P.S.); Department of Radiology, University of Washington, Seattle, Wash (G.M.C.); Department of Ultrasound, Shenzhen University General Hospital, Shenzhen, China (Q.L.); Department of Radiology, Liver Imaging Group, University of California San Diego, La Jolla, Calif (M.S.M., Y.C., C.B.S., K.J.F.); MGH Fatty Liver Program, Gastrointestinal Unit, Massachusetts General Hospital, Boston, Mass (K.E.C.); BioAge Labs, Richmond, Calif (S.S.S.); Foundation for the National Institutes of Health, North Bethesda, Md (H.H., T.N.K.); Regeneron Pharmaceuticals, Tarrytown, NY (R.A.C.); Department of Medicine, Division of Gastroenterology, NAFLD Research Center, University of California at San Diego, La Jolla, Calif (R.L.); Quantitative Health Sciences, Cleveland Clinic Foundation, Cleveland, Ohio (N.A.O.); and Department of Internal Medicine, Division of Internal Medicine, Division of Gastroenterology, Virginia Commonwealth University Medical Center, Richmond, Va (A.J.S.)
| | - Sudha S Shankar
- From the Center for Ultrasound Research and Translation, Massachusetts General Hospital, 55 Fruit St, White Bldg, Rm 270, Boston, MA 02114 (T.T.P., A.O., X.W., Q.L., D.H., M.M., H.E., A.E.S.); Harvard Medical School, Boston, Mass (T.T.P., A.O., Q.L., A.E.S.); Pfizer, Cambridge, Mass (S.P.S.); Department of Radiology, University of Washington, Seattle, Wash (G.M.C.); Department of Ultrasound, Shenzhen University General Hospital, Shenzhen, China (Q.L.); Department of Radiology, Liver Imaging Group, University of California San Diego, La Jolla, Calif (M.S.M., Y.C., C.B.S., K.J.F.); MGH Fatty Liver Program, Gastrointestinal Unit, Massachusetts General Hospital, Boston, Mass (K.E.C.); BioAge Labs, Richmond, Calif (S.S.S.); Foundation for the National Institutes of Health, North Bethesda, Md (H.H., T.N.K.); Regeneron Pharmaceuticals, Tarrytown, NY (R.A.C.); Department of Medicine, Division of Gastroenterology, NAFLD Research Center, University of California at San Diego, La Jolla, Calif (R.L.); Quantitative Health Sciences, Cleveland Clinic Foundation, Cleveland, Ohio (N.A.O.); and Department of Internal Medicine, Division of Internal Medicine, Division of Gastroenterology, Virginia Commonwealth University Medical Center, Richmond, Va (A.J.S.)
| | - Helen Heymann
- From the Center for Ultrasound Research and Translation, Massachusetts General Hospital, 55 Fruit St, White Bldg, Rm 270, Boston, MA 02114 (T.T.P., A.O., X.W., Q.L., D.H., M.M., H.E., A.E.S.); Harvard Medical School, Boston, Mass (T.T.P., A.O., Q.L., A.E.S.); Pfizer, Cambridge, Mass (S.P.S.); Department of Radiology, University of Washington, Seattle, Wash (G.M.C.); Department of Ultrasound, Shenzhen University General Hospital, Shenzhen, China (Q.L.); Department of Radiology, Liver Imaging Group, University of California San Diego, La Jolla, Calif (M.S.M., Y.C., C.B.S., K.J.F.); MGH Fatty Liver Program, Gastrointestinal Unit, Massachusetts General Hospital, Boston, Mass (K.E.C.); BioAge Labs, Richmond, Calif (S.S.S.); Foundation for the National Institutes of Health, North Bethesda, Md (H.H., T.N.K.); Regeneron Pharmaceuticals, Tarrytown, NY (R.A.C.); Department of Medicine, Division of Gastroenterology, NAFLD Research Center, University of California at San Diego, La Jolla, Calif (R.L.); Quantitative Health Sciences, Cleveland Clinic Foundation, Cleveland, Ohio (N.A.O.); and Department of Internal Medicine, Division of Internal Medicine, Division of Gastroenterology, Virginia Commonwealth University Medical Center, Richmond, Va (A.J.S.)
| | - Tania N Kamphaus
- From the Center for Ultrasound Research and Translation, Massachusetts General Hospital, 55 Fruit St, White Bldg, Rm 270, Boston, MA 02114 (T.T.P., A.O., X.W., Q.L., D.H., M.M., H.E., A.E.S.); Harvard Medical School, Boston, Mass (T.T.P., A.O., Q.L., A.E.S.); Pfizer, Cambridge, Mass (S.P.S.); Department of Radiology, University of Washington, Seattle, Wash (G.M.C.); Department of Ultrasound, Shenzhen University General Hospital, Shenzhen, China (Q.L.); Department of Radiology, Liver Imaging Group, University of California San Diego, La Jolla, Calif (M.S.M., Y.C., C.B.S., K.J.F.); MGH Fatty Liver Program, Gastrointestinal Unit, Massachusetts General Hospital, Boston, Mass (K.E.C.); BioAge Labs, Richmond, Calif (S.S.S.); Foundation for the National Institutes of Health, North Bethesda, Md (H.H., T.N.K.); Regeneron Pharmaceuticals, Tarrytown, NY (R.A.C.); Department of Medicine, Division of Gastroenterology, NAFLD Research Center, University of California at San Diego, La Jolla, Calif (R.L.); Quantitative Health Sciences, Cleveland Clinic Foundation, Cleveland, Ohio (N.A.O.); and Department of Internal Medicine, Division of Internal Medicine, Division of Gastroenterology, Virginia Commonwealth University Medical Center, Richmond, Va (A.J.S.)
| | - Roberto A Calle
- From the Center for Ultrasound Research and Translation, Massachusetts General Hospital, 55 Fruit St, White Bldg, Rm 270, Boston, MA 02114 (T.T.P., A.O., X.W., Q.L., D.H., M.M., H.E., A.E.S.); Harvard Medical School, Boston, Mass (T.T.P., A.O., Q.L., A.E.S.); Pfizer, Cambridge, Mass (S.P.S.); Department of Radiology, University of Washington, Seattle, Wash (G.M.C.); Department of Ultrasound, Shenzhen University General Hospital, Shenzhen, China (Q.L.); Department of Radiology, Liver Imaging Group, University of California San Diego, La Jolla, Calif (M.S.M., Y.C., C.B.S., K.J.F.); MGH Fatty Liver Program, Gastrointestinal Unit, Massachusetts General Hospital, Boston, Mass (K.E.C.); BioAge Labs, Richmond, Calif (S.S.S.); Foundation for the National Institutes of Health, North Bethesda, Md (H.H., T.N.K.); Regeneron Pharmaceuticals, Tarrytown, NY (R.A.C.); Department of Medicine, Division of Gastroenterology, NAFLD Research Center, University of California at San Diego, La Jolla, Calif (R.L.); Quantitative Health Sciences, Cleveland Clinic Foundation, Cleveland, Ohio (N.A.O.); and Department of Internal Medicine, Division of Internal Medicine, Division of Gastroenterology, Virginia Commonwealth University Medical Center, Richmond, Va (A.J.S.)
| | - Yesenia Covarrubias
- From the Center for Ultrasound Research and Translation, Massachusetts General Hospital, 55 Fruit St, White Bldg, Rm 270, Boston, MA 02114 (T.T.P., A.O., X.W., Q.L., D.H., M.M., H.E., A.E.S.); Harvard Medical School, Boston, Mass (T.T.P., A.O., Q.L., A.E.S.); Pfizer, Cambridge, Mass (S.P.S.); Department of Radiology, University of Washington, Seattle, Wash (G.M.C.); Department of Ultrasound, Shenzhen University General Hospital, Shenzhen, China (Q.L.); Department of Radiology, Liver Imaging Group, University of California San Diego, La Jolla, Calif (M.S.M., Y.C., C.B.S., K.J.F.); MGH Fatty Liver Program, Gastrointestinal Unit, Massachusetts General Hospital, Boston, Mass (K.E.C.); BioAge Labs, Richmond, Calif (S.S.S.); Foundation for the National Institutes of Health, North Bethesda, Md (H.H., T.N.K.); Regeneron Pharmaceuticals, Tarrytown, NY (R.A.C.); Department of Medicine, Division of Gastroenterology, NAFLD Research Center, University of California at San Diego, La Jolla, Calif (R.L.); Quantitative Health Sciences, Cleveland Clinic Foundation, Cleveland, Ohio (N.A.O.); and Department of Internal Medicine, Division of Internal Medicine, Division of Gastroenterology, Virginia Commonwealth University Medical Center, Richmond, Va (A.J.S.)
| | - Rohit Loomba
- From the Center for Ultrasound Research and Translation, Massachusetts General Hospital, 55 Fruit St, White Bldg, Rm 270, Boston, MA 02114 (T.T.P., A.O., X.W., Q.L., D.H., M.M., H.E., A.E.S.); Harvard Medical School, Boston, Mass (T.T.P., A.O., Q.L., A.E.S.); Pfizer, Cambridge, Mass (S.P.S.); Department of Radiology, University of Washington, Seattle, Wash (G.M.C.); Department of Ultrasound, Shenzhen University General Hospital, Shenzhen, China (Q.L.); Department of Radiology, Liver Imaging Group, University of California San Diego, La Jolla, Calif (M.S.M., Y.C., C.B.S., K.J.F.); MGH Fatty Liver Program, Gastrointestinal Unit, Massachusetts General Hospital, Boston, Mass (K.E.C.); BioAge Labs, Richmond, Calif (S.S.S.); Foundation for the National Institutes of Health, North Bethesda, Md (H.H., T.N.K.); Regeneron Pharmaceuticals, Tarrytown, NY (R.A.C.); Department of Medicine, Division of Gastroenterology, NAFLD Research Center, University of California at San Diego, La Jolla, Calif (R.L.); Quantitative Health Sciences, Cleveland Clinic Foundation, Cleveland, Ohio (N.A.O.); and Department of Internal Medicine, Division of Internal Medicine, Division of Gastroenterology, Virginia Commonwealth University Medical Center, Richmond, Va (A.J.S.)
| | - Nancy A Obuchowski
- From the Center for Ultrasound Research and Translation, Massachusetts General Hospital, 55 Fruit St, White Bldg, Rm 270, Boston, MA 02114 (T.T.P., A.O., X.W., Q.L., D.H., M.M., H.E., A.E.S.); Harvard Medical School, Boston, Mass (T.T.P., A.O., Q.L., A.E.S.); Pfizer, Cambridge, Mass (S.P.S.); Department of Radiology, University of Washington, Seattle, Wash (G.M.C.); Department of Ultrasound, Shenzhen University General Hospital, Shenzhen, China (Q.L.); Department of Radiology, Liver Imaging Group, University of California San Diego, La Jolla, Calif (M.S.M., Y.C., C.B.S., K.J.F.); MGH Fatty Liver Program, Gastrointestinal Unit, Massachusetts General Hospital, Boston, Mass (K.E.C.); BioAge Labs, Richmond, Calif (S.S.S.); Foundation for the National Institutes of Health, North Bethesda, Md (H.H., T.N.K.); Regeneron Pharmaceuticals, Tarrytown, NY (R.A.C.); Department of Medicine, Division of Gastroenterology, NAFLD Research Center, University of California at San Diego, La Jolla, Calif (R.L.); Quantitative Health Sciences, Cleveland Clinic Foundation, Cleveland, Ohio (N.A.O.); and Department of Internal Medicine, Division of Internal Medicine, Division of Gastroenterology, Virginia Commonwealth University Medical Center, Richmond, Va (A.J.S.)
| | - Arun J Sanyal
- From the Center for Ultrasound Research and Translation, Massachusetts General Hospital, 55 Fruit St, White Bldg, Rm 270, Boston, MA 02114 (T.T.P., A.O., X.W., Q.L., D.H., M.M., H.E., A.E.S.); Harvard Medical School, Boston, Mass (T.T.P., A.O., Q.L., A.E.S.); Pfizer, Cambridge, Mass (S.P.S.); Department of Radiology, University of Washington, Seattle, Wash (G.M.C.); Department of Ultrasound, Shenzhen University General Hospital, Shenzhen, China (Q.L.); Department of Radiology, Liver Imaging Group, University of California San Diego, La Jolla, Calif (M.S.M., Y.C., C.B.S., K.J.F.); MGH Fatty Liver Program, Gastrointestinal Unit, Massachusetts General Hospital, Boston, Mass (K.E.C.); BioAge Labs, Richmond, Calif (S.S.S.); Foundation for the National Institutes of Health, North Bethesda, Md (H.H., T.N.K.); Regeneron Pharmaceuticals, Tarrytown, NY (R.A.C.); Department of Medicine, Division of Gastroenterology, NAFLD Research Center, University of California at San Diego, La Jolla, Calif (R.L.); Quantitative Health Sciences, Cleveland Clinic Foundation, Cleveland, Ohio (N.A.O.); and Department of Internal Medicine, Division of Internal Medicine, Division of Gastroenterology, Virginia Commonwealth University Medical Center, Richmond, Va (A.J.S.)
| | - Claude B Sirlin
- From the Center for Ultrasound Research and Translation, Massachusetts General Hospital, 55 Fruit St, White Bldg, Rm 270, Boston, MA 02114 (T.T.P., A.O., X.W., Q.L., D.H., M.M., H.E., A.E.S.); Harvard Medical School, Boston, Mass (T.T.P., A.O., Q.L., A.E.S.); Pfizer, Cambridge, Mass (S.P.S.); Department of Radiology, University of Washington, Seattle, Wash (G.M.C.); Department of Ultrasound, Shenzhen University General Hospital, Shenzhen, China (Q.L.); Department of Radiology, Liver Imaging Group, University of California San Diego, La Jolla, Calif (M.S.M., Y.C., C.B.S., K.J.F.); MGH Fatty Liver Program, Gastrointestinal Unit, Massachusetts General Hospital, Boston, Mass (K.E.C.); BioAge Labs, Richmond, Calif (S.S.S.); Foundation for the National Institutes of Health, North Bethesda, Md (H.H., T.N.K.); Regeneron Pharmaceuticals, Tarrytown, NY (R.A.C.); Department of Medicine, Division of Gastroenterology, NAFLD Research Center, University of California at San Diego, La Jolla, Calif (R.L.); Quantitative Health Sciences, Cleveland Clinic Foundation, Cleveland, Ohio (N.A.O.); and Department of Internal Medicine, Division of Internal Medicine, Division of Gastroenterology, Virginia Commonwealth University Medical Center, Richmond, Va (A.J.S.)
| | - Kathryn J Fowler
- From the Center for Ultrasound Research and Translation, Massachusetts General Hospital, 55 Fruit St, White Bldg, Rm 270, Boston, MA 02114 (T.T.P., A.O., X.W., Q.L., D.H., M.M., H.E., A.E.S.); Harvard Medical School, Boston, Mass (T.T.P., A.O., Q.L., A.E.S.); Pfizer, Cambridge, Mass (S.P.S.); Department of Radiology, University of Washington, Seattle, Wash (G.M.C.); Department of Ultrasound, Shenzhen University General Hospital, Shenzhen, China (Q.L.); Department of Radiology, Liver Imaging Group, University of California San Diego, La Jolla, Calif (M.S.M., Y.C., C.B.S., K.J.F.); MGH Fatty Liver Program, Gastrointestinal Unit, Massachusetts General Hospital, Boston, Mass (K.E.C.); BioAge Labs, Richmond, Calif (S.S.S.); Foundation for the National Institutes of Health, North Bethesda, Md (H.H., T.N.K.); Regeneron Pharmaceuticals, Tarrytown, NY (R.A.C.); Department of Medicine, Division of Gastroenterology, NAFLD Research Center, University of California at San Diego, La Jolla, Calif (R.L.); Quantitative Health Sciences, Cleveland Clinic Foundation, Cleveland, Ohio (N.A.O.); and Department of Internal Medicine, Division of Internal Medicine, Division of Gastroenterology, Virginia Commonwealth University Medical Center, Richmond, Va (A.J.S.)
| | - Anthony E Samir
- From the Center for Ultrasound Research and Translation, Massachusetts General Hospital, 55 Fruit St, White Bldg, Rm 270, Boston, MA 02114 (T.T.P., A.O., X.W., Q.L., D.H., M.M., H.E., A.E.S.); Harvard Medical School, Boston, Mass (T.T.P., A.O., Q.L., A.E.S.); Pfizer, Cambridge, Mass (S.P.S.); Department of Radiology, University of Washington, Seattle, Wash (G.M.C.); Department of Ultrasound, Shenzhen University General Hospital, Shenzhen, China (Q.L.); Department of Radiology, Liver Imaging Group, University of California San Diego, La Jolla, Calif (M.S.M., Y.C., C.B.S., K.J.F.); MGH Fatty Liver Program, Gastrointestinal Unit, Massachusetts General Hospital, Boston, Mass (K.E.C.); BioAge Labs, Richmond, Calif (S.S.S.); Foundation for the National Institutes of Health, North Bethesda, Md (H.H., T.N.K.); Regeneron Pharmaceuticals, Tarrytown, NY (R.A.C.); Department of Medicine, Division of Gastroenterology, NAFLD Research Center, University of California at San Diego, La Jolla, Calif (R.L.); Quantitative Health Sciences, Cleveland Clinic Foundation, Cleveland, Ohio (N.A.O.); and Department of Internal Medicine, Division of Internal Medicine, Division of Gastroenterology, Virginia Commonwealth University Medical Center, Richmond, Va (A.J.S.)
| |
Collapse
|
8
|
Herrera-Marcos LV, Arbones-Mainar JM, Osada J. Lipoprotein Lipidomics as a Frontier in Non-Alcoholic Fatty Liver Disease Biomarker Discovery. Int J Mol Sci 2024; 25:8285. [PMID: 39125855 PMCID: PMC11311740 DOI: 10.3390/ijms25158285] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2024] [Revised: 07/16/2024] [Accepted: 07/27/2024] [Indexed: 08/12/2024] Open
Abstract
Non-alcoholic fatty liver disease (NAFLD) is a progressive liver disease characterized by the build-up of fat in the liver of individuals in the absence of alcohol consumption. This condition has become a burden in modern societies aggravated by the lack of appropriate predictive biomarkers (other than liver biopsy). To better understand this disease and to find appropriate biomarkers, a new technology has emerged in the last two decades with the ability to explore the unmapped role of lipids in this disease: lipidomics. This technology, based on the combination of chromatography and mass spectrometry, has been extensively used to explore the lipid metabolism of NAFLD. In this review, we aim to summarize the knowledge gained through lipidomics assays exploring tissues, plasma, and lipoproteins from individuals with NAFLD. Our goal is to identify common features and active pathways that could facilitate the finding of a reliable biomarker from this field. The most frequent observation was a variable decrease (1-9%) in polyunsaturated fatty acids in phospholipids and non-esterified fatty acids in NAFLD patients, both in plasma and liver. Additionally, a reduction in phosphatidylcholines is a common feature in the liver. Due to the scarcity of studies, further research is needed to properly detect lipoprotein, plasma, and tissue lipid signatures of NAFLD etiologies, and NAFLD subtypes, and to define the relevance of this technology in disease management strategies in the push toward personalized medicine.
Collapse
Affiliation(s)
- Luis V. Herrera-Marcos
- Departamento de Bioquímica y Biología Molecular y Celular, Facultad de Veterinaria, Universidad de Zaragoza, E-50013 Zaragoza, Spain; (L.V.H.-M.); (J.O.)
- Instituto Agroalimentario de Aragón, CITA-Universidad de Zaragoza, E-50013 Zaragoza, Spain
- Instituto de Investigación Sanitaria (IIS) Aragon, E-50009 Zaragoza, Spain
- CIBER Fisiopatología Obesidad y Nutrición (CIBERObn), Instituto Salud Carlos III, E-28029 Madrid, Spain
| | - Jose M. Arbones-Mainar
- Instituto de Investigación Sanitaria (IIS) Aragon, E-50009 Zaragoza, Spain
- CIBER Fisiopatología Obesidad y Nutrición (CIBERObn), Instituto Salud Carlos III, E-28029 Madrid, Spain
- Adipocyte and Fat Biology Laboratory (AdipoFat), Translational Research Unit, University Hospital Miguel Servet, E-50013 Zaragoza, Spain
- Instituto Aragonés de Ciencias de la Salud (IACS), E-50009 Zaragoza, Spain
| | - Jesús Osada
- Departamento de Bioquímica y Biología Molecular y Celular, Facultad de Veterinaria, Universidad de Zaragoza, E-50013 Zaragoza, Spain; (L.V.H.-M.); (J.O.)
- Instituto Agroalimentario de Aragón, CITA-Universidad de Zaragoza, E-50013 Zaragoza, Spain
- Instituto de Investigación Sanitaria (IIS) Aragon, E-50009 Zaragoza, Spain
- CIBER Fisiopatología Obesidad y Nutrición (CIBERObn), Instituto Salud Carlos III, E-28029 Madrid, Spain
| |
Collapse
|
9
|
Kokkorakis M, Muzurović E, Volčanšek Š, Chakhtoura M, Hill MA, Mikhailidis DP, Mantzoros CS. Steatotic Liver Disease: Pathophysiology and Emerging Pharmacotherapies. Pharmacol Rev 2024; 76:454-499. [PMID: 38697855 DOI: 10.1124/pharmrev.123.001087] [Citation(s) in RCA: 24] [Impact Index Per Article: 24.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2023] [Revised: 12/22/2023] [Accepted: 01/25/2024] [Indexed: 05/05/2024] Open
Abstract
Steatotic liver disease (SLD) displays a dynamic and complex disease phenotype. Consequently, the metabolic dysfunction-associated steatotic liver disease (MASLD)/metabolic dysfunction-associated steatohepatitis (MASH) therapeutic pipeline is expanding rapidly and in multiple directions. In parallel, noninvasive tools for diagnosing and monitoring responses to therapeutic interventions are being studied, and clinically feasible findings are being explored as primary outcomes in interventional trials. The realization that distinct subgroups exist under the umbrella of SLD should guide more precise and personalized treatment recommendations and facilitate advancements in pharmacotherapeutics. This review summarizes recent updates of pathophysiology-based nomenclature and outlines both effective pharmacotherapeutics and those in the pipeline for MASLD/MASH, detailing their mode of action and the current status of phase 2 and 3 clinical trials. Of the extensive arsenal of pharmacotherapeutics in the MASLD/MASH pipeline, several have been rejected, whereas other, mainly monotherapy options, have shown only marginal benefits and are now being tested as part of combination therapies, yet others are still in development as monotherapies. Although the Food and Drug Administration (FDA) has recently approved resmetirom, additional therapeutic approaches in development will ideally target MASH and fibrosis while improving cardiometabolic risk factors. Due to the urgent need for the development of novel therapeutic strategies and the potential availability of safety and tolerability data, repurposing existing and approved drugs is an appealing option. Finally, it is essential to highlight that SLD and, by extension, MASLD should be recognized and approached as a systemic disease affecting multiple organs, with the vigorous implementation of interdisciplinary and coordinated action plans. SIGNIFICANCE STATEMENT: Steatotic liver disease (SLD), including metabolic dysfunction-associated steatotic liver disease and metabolic dysfunction-associated steatohepatitis, is the most prevalent chronic liver condition, affecting more than one-fourth of the global population. This review aims to provide the most recent information regarding SLD pathophysiology, diagnosis, and management according to the latest advancements in the guidelines and clinical trials. Collectively, it is hoped that the information provided furthers the understanding of the current state of SLD with direct clinical implications and stimulates research initiatives.
Collapse
Affiliation(s)
- Michail Kokkorakis
- Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts (M.K., C.S.M.); Department of Clinical Pharmacy and Pharmacology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands (M.K.); Endocrinology Section, Department of Internal Medicine, Clinical Center of Montenegro, Podgorica, Montenegro (E.M.); Faculty of Medicine, University of Montenegro, Podgorica, Montenegro (E.M.); Department of Endocrinology, Diabetes, and Metabolic Diseases, University Medical Center Ljubljana, Ljubljana, Slovenia (Š.V.); Medical Faculty Ljubljana, Ljubljana, Slovenia (Š.V.); Division of Endocrinology, Department of Internal Medicine, American University of Beirut Medical Center, Beirut, Lebanon (M.C.); Dalton Cardiovascular Research Center, University of Missouri, Columbia, Missouri (M.A.H.); Department of Medical Pharmacology and Physiology, School of Medicine, University of Missouri, Columbia, Missouri (M.A.H.); Department of Clinical Biochemistry, Royal Free Hospital Campus, University College London Medical School, University College London (UCL), London, United Kingdom (D.P.M.); Mohammed Bin Rashid University of Medicine and Health Sciences, Dubai, United Arab Emirates (D.P.M.); and Boston VA Healthcare System, Harvard Medical School, Boston, Massachusetts (C.S.M.)
| | - Emir Muzurović
- Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts (M.K., C.S.M.); Department of Clinical Pharmacy and Pharmacology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands (M.K.); Endocrinology Section, Department of Internal Medicine, Clinical Center of Montenegro, Podgorica, Montenegro (E.M.); Faculty of Medicine, University of Montenegro, Podgorica, Montenegro (E.M.); Department of Endocrinology, Diabetes, and Metabolic Diseases, University Medical Center Ljubljana, Ljubljana, Slovenia (Š.V.); Medical Faculty Ljubljana, Ljubljana, Slovenia (Š.V.); Division of Endocrinology, Department of Internal Medicine, American University of Beirut Medical Center, Beirut, Lebanon (M.C.); Dalton Cardiovascular Research Center, University of Missouri, Columbia, Missouri (M.A.H.); Department of Medical Pharmacology and Physiology, School of Medicine, University of Missouri, Columbia, Missouri (M.A.H.); Department of Clinical Biochemistry, Royal Free Hospital Campus, University College London Medical School, University College London (UCL), London, United Kingdom (D.P.M.); Mohammed Bin Rashid University of Medicine and Health Sciences, Dubai, United Arab Emirates (D.P.M.); and Boston VA Healthcare System, Harvard Medical School, Boston, Massachusetts (C.S.M.)
| | - Špela Volčanšek
- Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts (M.K., C.S.M.); Department of Clinical Pharmacy and Pharmacology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands (M.K.); Endocrinology Section, Department of Internal Medicine, Clinical Center of Montenegro, Podgorica, Montenegro (E.M.); Faculty of Medicine, University of Montenegro, Podgorica, Montenegro (E.M.); Department of Endocrinology, Diabetes, and Metabolic Diseases, University Medical Center Ljubljana, Ljubljana, Slovenia (Š.V.); Medical Faculty Ljubljana, Ljubljana, Slovenia (Š.V.); Division of Endocrinology, Department of Internal Medicine, American University of Beirut Medical Center, Beirut, Lebanon (M.C.); Dalton Cardiovascular Research Center, University of Missouri, Columbia, Missouri (M.A.H.); Department of Medical Pharmacology and Physiology, School of Medicine, University of Missouri, Columbia, Missouri (M.A.H.); Department of Clinical Biochemistry, Royal Free Hospital Campus, University College London Medical School, University College London (UCL), London, United Kingdom (D.P.M.); Mohammed Bin Rashid University of Medicine and Health Sciences, Dubai, United Arab Emirates (D.P.M.); and Boston VA Healthcare System, Harvard Medical School, Boston, Massachusetts (C.S.M.)
| | - Marlene Chakhtoura
- Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts (M.K., C.S.M.); Department of Clinical Pharmacy and Pharmacology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands (M.K.); Endocrinology Section, Department of Internal Medicine, Clinical Center of Montenegro, Podgorica, Montenegro (E.M.); Faculty of Medicine, University of Montenegro, Podgorica, Montenegro (E.M.); Department of Endocrinology, Diabetes, and Metabolic Diseases, University Medical Center Ljubljana, Ljubljana, Slovenia (Š.V.); Medical Faculty Ljubljana, Ljubljana, Slovenia (Š.V.); Division of Endocrinology, Department of Internal Medicine, American University of Beirut Medical Center, Beirut, Lebanon (M.C.); Dalton Cardiovascular Research Center, University of Missouri, Columbia, Missouri (M.A.H.); Department of Medical Pharmacology and Physiology, School of Medicine, University of Missouri, Columbia, Missouri (M.A.H.); Department of Clinical Biochemistry, Royal Free Hospital Campus, University College London Medical School, University College London (UCL), London, United Kingdom (D.P.M.); Mohammed Bin Rashid University of Medicine and Health Sciences, Dubai, United Arab Emirates (D.P.M.); and Boston VA Healthcare System, Harvard Medical School, Boston, Massachusetts (C.S.M.)
| | - Michael A Hill
- Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts (M.K., C.S.M.); Department of Clinical Pharmacy and Pharmacology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands (M.K.); Endocrinology Section, Department of Internal Medicine, Clinical Center of Montenegro, Podgorica, Montenegro (E.M.); Faculty of Medicine, University of Montenegro, Podgorica, Montenegro (E.M.); Department of Endocrinology, Diabetes, and Metabolic Diseases, University Medical Center Ljubljana, Ljubljana, Slovenia (Š.V.); Medical Faculty Ljubljana, Ljubljana, Slovenia (Š.V.); Division of Endocrinology, Department of Internal Medicine, American University of Beirut Medical Center, Beirut, Lebanon (M.C.); Dalton Cardiovascular Research Center, University of Missouri, Columbia, Missouri (M.A.H.); Department of Medical Pharmacology and Physiology, School of Medicine, University of Missouri, Columbia, Missouri (M.A.H.); Department of Clinical Biochemistry, Royal Free Hospital Campus, University College London Medical School, University College London (UCL), London, United Kingdom (D.P.M.); Mohammed Bin Rashid University of Medicine and Health Sciences, Dubai, United Arab Emirates (D.P.M.); and Boston VA Healthcare System, Harvard Medical School, Boston, Massachusetts (C.S.M.)
| | - Dimitri P Mikhailidis
- Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts (M.K., C.S.M.); Department of Clinical Pharmacy and Pharmacology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands (M.K.); Endocrinology Section, Department of Internal Medicine, Clinical Center of Montenegro, Podgorica, Montenegro (E.M.); Faculty of Medicine, University of Montenegro, Podgorica, Montenegro (E.M.); Department of Endocrinology, Diabetes, and Metabolic Diseases, University Medical Center Ljubljana, Ljubljana, Slovenia (Š.V.); Medical Faculty Ljubljana, Ljubljana, Slovenia (Š.V.); Division of Endocrinology, Department of Internal Medicine, American University of Beirut Medical Center, Beirut, Lebanon (M.C.); Dalton Cardiovascular Research Center, University of Missouri, Columbia, Missouri (M.A.H.); Department of Medical Pharmacology and Physiology, School of Medicine, University of Missouri, Columbia, Missouri (M.A.H.); Department of Clinical Biochemistry, Royal Free Hospital Campus, University College London Medical School, University College London (UCL), London, United Kingdom (D.P.M.); Mohammed Bin Rashid University of Medicine and Health Sciences, Dubai, United Arab Emirates (D.P.M.); and Boston VA Healthcare System, Harvard Medical School, Boston, Massachusetts (C.S.M.)
| | - Christos S Mantzoros
- Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts (M.K., C.S.M.); Department of Clinical Pharmacy and Pharmacology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands (M.K.); Endocrinology Section, Department of Internal Medicine, Clinical Center of Montenegro, Podgorica, Montenegro (E.M.); Faculty of Medicine, University of Montenegro, Podgorica, Montenegro (E.M.); Department of Endocrinology, Diabetes, and Metabolic Diseases, University Medical Center Ljubljana, Ljubljana, Slovenia (Š.V.); Medical Faculty Ljubljana, Ljubljana, Slovenia (Š.V.); Division of Endocrinology, Department of Internal Medicine, American University of Beirut Medical Center, Beirut, Lebanon (M.C.); Dalton Cardiovascular Research Center, University of Missouri, Columbia, Missouri (M.A.H.); Department of Medical Pharmacology and Physiology, School of Medicine, University of Missouri, Columbia, Missouri (M.A.H.); Department of Clinical Biochemistry, Royal Free Hospital Campus, University College London Medical School, University College London (UCL), London, United Kingdom (D.P.M.); Mohammed Bin Rashid University of Medicine and Health Sciences, Dubai, United Arab Emirates (D.P.M.); and Boston VA Healthcare System, Harvard Medical School, Boston, Massachusetts (C.S.M.)
| |
Collapse
|
10
|
Anstee QM, Magnanensi J, Hajji Y, Caron A, Majd Z, Rosenquist C, Hum DW, Staels B, Connelly MA, Loomba R, Harrison SA, Ratziu V, Sanyal AJ. Impact of age on NIS2+™ and other non-invasive blood tests for the evaluation of liver disease and detection of at-risk MASH. JHEP Rep 2024; 6:101011. [PMID: 38463540 PMCID: PMC10920708 DOI: 10.1016/j.jhepr.2024.101011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/29/2023] [Revised: 12/14/2023] [Accepted: 01/08/2024] [Indexed: 03/12/2024] Open
Abstract
Background & Aims Robust performance of non-invasive tests (NITs) across ages is critical to assess liver disease among patients with metabolic dysfunction-associated liver disease (MASLD). We evaluated the impact of age on the performance of NIS2+™ vs. other NITs. Methods An analysis cohort (N = 1,926) with biopsy-proven MASLD was selected among individuals screened for the phase III RESOLVE-IT clinical trial and divided into ≤45, 46-55, 56-64, and ≥65 years groups. To avoid potential confounding effects, a well-balanced cohort (n = 708; n = 177/age group) was obtained by applying a propensity score-matching algorithm to the analysis cohort. Baseline values of biomarkers and NITs were compared across age groups using one-way ANOVA, and the impact of age and histology were compared through three-way ANOVA. The impact of age on NIT performance for the detection of at-risk metabolic dysfunction-associated steatohepatitis (MASH; MASLD activity score [MAS] ≥4 and fibrosis stage [F] ≥2) was also evaluated. Results Age did not affect the distributions of NIS2+™ and APRI (aspartate aminotransferase-to-platelet ratio index), but significantly (p <0.0001) impacted those of NFS (NAFLD fibrosis score), FIB-4 (Fibrosis-4 index), and Enhanced Liver Fibrosis (ELF™) score. NIS2+™ was the only NIT on which fibrosis and MAS exerted a moderate to large effect. While the impact of fibrosis on APRI was moderate, that of MAS was low. The impact of age on FIB-4 and NFS was larger than that of fibrosis. NIS2+™ exhibited the highest AUROC values for detecting at-risk MASH across age groups, with stable performances irrespective of cut-offs. Conclusions NIS2+™ was not significantly impacted by age and was sensitive to both fibrosis and MAS grade, demonstrating a robust performance to rule in/out at-risk MASH with fixed cut-offs. Impact and Implications While metabolic dysfunction-associated steatotic liver disease (MASLD) can affect individuals of all ages, patient age could represent an important confounding factor when interpreting non-invasive test (NIT) results, highlighting the need for reliable and efficient NITs that are not impacted by age and that could be interpreted with fixed cut-offs, irrespective of patient age. We report the impact of age on different well-established NITs - among those tested, only two panels, NIS2+™ and APRI, were not impacted by age and can be used and interpreted independently of patient age. NIS2+™ was also sensitive to both fibrosis and MAS, further confirming its efficiency for the detection of the composite endpoint of at-risk MASH and its potential as a valuable candidate for large-scale implementation in clinical practice and clinical trials.
Collapse
Affiliation(s)
- 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
| | | | | | | | | | | | | | - Bart Staels
- Université de Lille, INSERM, CHU Lille, Institut Pasteur de Lille, Lille, France
| | | | - Rohit Loomba
- NAFLD Research Center, Division of Gastroenterology, University of California at San Diego, La Jolla, CA, US
| | - Stephen A. Harrison
- Summit Clinical Research, San Antonio, TX, US
- Radcliffe Department of Medicine, University of Oxford, Oxford, UK
| | - Vlad Ratziu
- Sorbonne Université, Institute for Cardiometabolism and Nutrition, Hôpital Pitié-Salpêtrière, Paris, France
| | - Arun J. Sanyal
- Division of Gastroenterology, Hepatology and Nutrition, Virginia Commonwealth University School of Medicine, Richmond, VA, US
| |
Collapse
|
11
|
Jimenez Ramos M, Kendall TJ, Drozdov I, Fallowfield JA. A data-driven approach to decode metabolic dysfunction-associated steatotic liver disease. Ann Hepatol 2024; 29:101278. [PMID: 38135251 PMCID: PMC10907333 DOI: 10.1016/j.aohep.2023.101278] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/30/2023] [Accepted: 12/04/2023] [Indexed: 12/24/2023]
Abstract
Metabolic dysfunction-associated steatotic liver disease (MASLD), defined by the presence of liver steatosis together with at least one out of five cardiometabolic factors, is the most common cause of chronic liver disease worldwide, affecting around one in three people. Yet the clinical presentation of MASLD and the risk of progression to cirrhosis and adverse clinical outcomes is highly variable. It, therefore, represents both a global public health threat and a precision medicine challenge. Artificial intelligence (AI) is being investigated in MASLD to develop reproducible, quantitative, and automated methods to enhance patient stratification and to discover new biomarkers and therapeutic targets in MASLD. This review details the different applications of AI and machine learning algorithms in MASLD, particularly in analyzing electronic health record, digital pathology, and imaging data. Additionally, it also describes how specific MASLD consortia are leveraging multimodal data sources to spark research breakthroughs in the field. Using a new national-level 'data commons' (SteatoSITE) as an exemplar, the opportunities, as well as the technical challenges of large-scale databases in MASLD research, are highlighted.
Collapse
Affiliation(s)
- Maria Jimenez Ramos
- Centre for Inflammation Research, Institute for Regeneration and Repair, University of Edinburgh, Edinburgh BioQuarter, 4-5 Little France Drive, Edinburgh EH16 4UU, UK
| | - Timothy J Kendall
- Centre for Inflammation Research, Institute for Regeneration and Repair, University of Edinburgh, Edinburgh BioQuarter, 4-5 Little France Drive, Edinburgh EH16 4UU, UK; Edinburgh Pathology, University of Edinburgh, 51 Little France Crescent, Old Dalkeith Rd, Edinburgh EH16 4SA, UK
| | - Ignat Drozdov
- Bering Limited, 54 Portland Place, London, W1B 1DY, UK
| | - Jonathan A Fallowfield
- Centre for Inflammation Research, Institute for Regeneration and Repair, University of Edinburgh, Edinburgh BioQuarter, 4-5 Little France Drive, Edinburgh EH16 4UU, UK.
| |
Collapse
|
12
|
McTeer M, Applegate D, Mesenbrink P, Ratziu V, Schattenberg JM, Bugianesi E, Geier A, Romero Gomez M, Dufour JF, Ekstedt M, Francque S, Yki-Jarvinen H, Allison M, Valenti L, Miele L, Pavlides M, Cobbold J, Papatheodoridis G, Holleboom AG, Tiniakos D, Brass C, Anstee QM, Missier P. Machine learning approaches to enhance diagnosis and staging of patients with MASLD using routinely available clinical information. PLoS One 2024; 19:e0299487. [PMID: 38421999 PMCID: PMC10903803 DOI: 10.1371/journal.pone.0299487] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2023] [Accepted: 02/09/2024] [Indexed: 03/02/2024] Open
Abstract
AIMS Metabolic dysfunction Associated Steatotic Liver Disease (MASLD) outcomes such as MASH (metabolic dysfunction associated steatohepatitis), fibrosis and cirrhosis are ordinarily determined by resource-intensive and invasive biopsies. We aim to show that routine clinical tests offer sufficient information to predict these endpoints. METHODS Using the LITMUS Metacohort derived from the European NAFLD Registry, the largest MASLD dataset in Europe, we create three combinations of features which vary in degree of procurement including a 19-variable feature set that are attained through a routine clinical appointment or blood test. This data was used to train predictive models using supervised machine learning (ML) algorithm XGBoost, alongside missing imputation technique MICE and class balancing algorithm SMOTE. Shapley Additive exPlanations (SHAP) were added to determine relative importance for each clinical variable. RESULTS Analysing nine biopsy-derived MASLD outcomes of cohort size ranging between 5385 and 6673 subjects, we were able to predict individuals at training set AUCs ranging from 0.719-0.994, including classifying individuals who are At-Risk MASH at an AUC = 0.899. Using two further feature combinations of 26-variables and 35-variables, which included composite scores known to be good indicators for MASLD endpoints and advanced specialist tests, we found predictive performance did not sufficiently improve. We are also able to present local and global explanations for each ML model, offering clinicians interpretability without the expense of worsening predictive performance. CONCLUSIONS This study developed a series of ML models of accuracy ranging from 71.9-99.4% using only easily extractable and readily available information in predicting MASLD outcomes which are usually determined through highly invasive means.
Collapse
Affiliation(s)
- Matthew McTeer
- Newcastle University, Newcastle upon Tyne, United Kingdom
| | - Douglas Applegate
- Novartis Institute for Biomedical Research, Cambridge, Massachusetts, United States of America
| | - Peter Mesenbrink
- Novartis Pharmaceuticals, East Hanover, New Jersey, United States of America
| | - Vlad Ratziu
- Institute of Cardiometabolism and Nutrition, Paris, France
| | - Jörn M. Schattenberg
- Department of Medicine II, University Medical Center Homburg and Saarland University, Homburg, Germany
| | | | | | | | | | | | | | | | | | | | - Luca Miele
- Università Cattolica del Sacro Cuore, Rome, Italy
| | | | | | | | | | - Dina Tiniakos
- Medical School of National & Kapodistrian University of Athens, Athens, Greece
- Translational & Clinical Research Institute, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne, United Kingdom
| | - Clifford Brass
- Novartis Institute for Biomedical Research, Cambridge, Massachusetts, United States of America
| | - Quentin M. Anstee
- Translational & Clinical Research Institute, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne, United Kingdom
- Newcastle NIHR Biomedical Research Centre NUTH NHS Trust, Newcastle upon Tyne, United Kingdom
| | - Paolo Missier
- Newcastle University, Newcastle upon Tyne, United Kingdom
| | | |
Collapse
|
13
|
Muzurović E, Maćešić M, Kavarić S. Liver Fibrosis and Atherosclerosis, a Consequence of Metabolic Dysfunction-Do They Share a Similar Pathophysiological Background? Angiology 2024:33197241234076. [PMID: 38358750 DOI: 10.1177/00033197241234076] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/16/2024]
Affiliation(s)
- Emir Muzurović
- Department of Internal Medicine, Endocrinology Section, Clinical Centre of Montenegro, Podgorica, Montenegro
- Faculty of Medicine, University of Montenegro, Podgorica, Montenegro
| | - Marija Maćešić
- Clinic for Endocrinology, Diabetes and Metabolic Diseases, University Clinical Center of Serbia, Belgrade, Serbia
- Faculty of Medicine, University of Belgrade, Belgrade, Serbia
| | - Sreten Kavarić
- Department of Internal Medicine, Endocrinology Section, Clinical Centre of Montenegro, Podgorica, Montenegro
- Faculty of Medicine, University of Montenegro, Podgorica, Montenegro
| |
Collapse
|
14
|
Ratziu V, Harrison SA, Hajji Y, Magnanensi J, Petit S, Majd Z, Delecroix E, Rosenquist C, Hum D, Staels B, Anstee QM, Sanyal AJ. NIS2+ TM as a screening tool to optimize patient selection in metabolic dysfunction-associated steatohepatitis clinical trials. J Hepatol 2024; 80:209-219. [PMID: 38061448 DOI: 10.1016/j.jhep.2023.10.038] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/31/2023] [Revised: 09/25/2023] [Accepted: 10/23/2023] [Indexed: 01/28/2024]
Abstract
BACKGROUND & AIMS Strategies to reduce liver biopsy (LB) screen failures through better patient selection are needed for clinical trials. Standard fibrosis biomarkers were not derived to detect "at-risk" metabolic dysfunction-associated steatohepatitis (MASH; MASH with metabolic dysfunction-associated steatotic liver disease score ≥4 and fibrosis stage ≥2). We compared the performance of screening pathways that incorporate NIS2+™, an optimized version of the blood-based NIS4® technology designed to identify at-risk MASH, with those incorporating fibrosis (FIB)-4 within the RESOLVE-IT clinical trial (NCT02704403), aiming for optimized selection of patients for LB. METHODS A retrospective simulation analysis was conducted in the RESOLVE-IT screening pathway (RSP) cohort. LB failure rate (LBFR), number of patients needed to screen, and overall cost estimations of different pathways were calculated for a range of NIS2+™ and FIB-4 cut-offs and compared with those of the RSP, which relied on investigators' local practices. An analysis of potential recruitment bias based on histology, sex, age, or comorbidities was performed. RESULTS The analysis cohort included 1,929 patients, 765 (40%) with at-risk MASH. The NIS2+™ pathway resulted in a significantly lower LBFR (39%) compared with the FIB-4 pathway (58%) or the RSP (60%) when using cost-optimized cut-offs (NIS2+™, 0.53; FIB-4, 0.58). For every 1,000 inclusions, NIS2+™ significantly reduced unnecessary LBs (632 vs. 1,522; -58%) and screening costs (US$12.7 million vs. US$15.0 million) vs. the RSP, while the number of patients needed to screen increased moderately (3,220 to 4,033). NIS2+™ alone is better than FIB-4 alone or combined with FIB-4. CONCLUSIONS This analysis demonstrated that patient selection for LB using NIS2+™ significantly reduced unnecessary biopsies and screening costs, which could greatly improve the feasibility of MASH clinical trials. IMPACT AND IMPLICATIONS Simple and accurate non-invasive strategies to optimize the selection of patients who should be referred for liver biopsy for inclusion in MASH clinical trials is critical to reduce the high liver biopsy failure rates. While the use of the Fibrosis-4 index alone did not lead to a significant improvement of the screening process, selecting patients using NIS2+™, a recently developed optimization of the NIS4® technology for the detection of at-risk MASH, showed improved performance by simultaneously reducing liver biopsy failure rates and the overall cost of the trial, while maintaining the number of patients needed to screen at a manageable level and not generating any bias in included patients' characteristics. This makes NIS2+™ an accurate and reliable screening tool that could improve the recruitment of patients in future MASH clinical trials, and would lead to increased patient comfort and security, ensuring timely and cost-efficient trial completion.
Collapse
Affiliation(s)
- Vlad Ratziu
- Sorbonne Université, Institute for Cardiometabolism and Nutrition, Hôpital Pitié-Salpêtrière, INSERM UMRS 1138 CRC, Paris, France
| | - Stephen A Harrison
- Summit Clinical Research, San Antonio, TX, USA; Radcliffe Department of Medicine, University of Oxford, Oxford, UK
| | | | | | | | | | | | | | | | - Bart Staels
- Université de Lille, INSERM, CHU Lille, Institut Pasteur de Lille, U1011-EGID, 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
|
15
|
Rinella ME, Sookoian S. From NAFLD to MASLD: updated naming and diagnosis criteria for fatty liver disease. J Lipid Res 2024; 65:100485. [PMID: 38103785 PMCID: PMC10824973 DOI: 10.1016/j.jlr.2023.100485] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2023] Open
Affiliation(s)
- Mary E Rinella
- Pritzker School of Medicine, University of Chicago, Chicago, IL, USA.
| | - Silvia Sookoian
- Clinical and Molecular Hepatology, Centro de Investigación Traslacional en Salud, Universidad Maimónides, Buenos Aires, Argentina; Facultad de Ciencias de la Salud, Universidad Maimónides, Buenos Aires, Argentina; Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires, Argentina.
| |
Collapse
|
16
|
Ozturk A, Kumar V, Pierce TT, Li Q, Baikpour M, Rosado-Mendez I, Wang M, Guo P, Schoen S, Gu Y, Dayavansha S, Grajo JR, Samir AE. The Future Is Beyond Bright: The Evolving Role of Quantitative US for Fatty Liver Disease. Radiology 2023; 309:e223146. [PMID: 37934095 PMCID: PMC10695672 DOI: 10.1148/radiol.223146] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2022] [Revised: 04/18/2023] [Accepted: 05/08/2023] [Indexed: 11/08/2023]
Abstract
Nonalcoholic fatty liver disease (NAFLD) is a common cause of morbidity and mortality. Nonfocal liver biopsy is the historical reference standard for evaluating NAFLD, but it is limited by invasiveness, high cost, and sampling error. Imaging methods are ideally situated to provide quantifiable results and rule out other anatomic diseases of the liver. MRI and US have shown great promise for the noninvasive evaluation of NAFLD. US is particularly well suited to address the population-level problem of NAFLD because it is lower-cost, more available, and more tolerable to a broader range of patients than MRI. Noninvasive US methods to evaluate liver fibrosis are widely available, and US-based tools to evaluate steatosis and inflammation are gaining traction. US techniques including shear-wave elastography, Doppler spectral imaging, attenuation coefficient, hepatorenal index, speed of sound, and backscatter-based estimation have regulatory clearance and are in clinical use. New methods based on channel and radiofrequency data analysis approaches have shown promise but are mostly experimental. This review discusses the advantages and limitations of clinically available and experimental approaches to sonographic liver tissue characterization for NAFLD diagnosis as well as future applications and strategies to overcome current limitations.
Collapse
Affiliation(s)
- Arinc Ozturk
- From the Center for Ultrasound Research & Translation,
Department of Radiology, Massachusetts General Hospital, 101 Merrimac St, 3rd
Floor, 323G, Boston, MA 02114 (A.O., V.K., T.T.P., Q.L., M.B., P.G., S.S., Y.G.,
S.D., A.E.S.); Harvard Medical School, Boston, Mass (A.O., V.K., T.T.P, Q.L.,
A.E.S.); Departments of Medical Physics and Radiology, University of Wisconsin,
Madison, Wis (I.R.M.); GE HealthCare, Milwaukee, Wis (M.W.); and Department of
Radiology, University of Florida, Gainesville, Fla (J.R.G.)
| | - Viksit Kumar
- From the Center for Ultrasound Research & Translation,
Department of Radiology, Massachusetts General Hospital, 101 Merrimac St, 3rd
Floor, 323G, Boston, MA 02114 (A.O., V.K., T.T.P., Q.L., M.B., P.G., S.S., Y.G.,
S.D., A.E.S.); Harvard Medical School, Boston, Mass (A.O., V.K., T.T.P, Q.L.,
A.E.S.); Departments of Medical Physics and Radiology, University of Wisconsin,
Madison, Wis (I.R.M.); GE HealthCare, Milwaukee, Wis (M.W.); and Department of
Radiology, University of Florida, Gainesville, Fla (J.R.G.)
| | - Theodore T. Pierce
- From the Center for Ultrasound Research & Translation,
Department of Radiology, Massachusetts General Hospital, 101 Merrimac St, 3rd
Floor, 323G, Boston, MA 02114 (A.O., V.K., T.T.P., Q.L., M.B., P.G., S.S., Y.G.,
S.D., A.E.S.); Harvard Medical School, Boston, Mass (A.O., V.K., T.T.P, Q.L.,
A.E.S.); Departments of Medical Physics and Radiology, University of Wisconsin,
Madison, Wis (I.R.M.); GE HealthCare, Milwaukee, Wis (M.W.); and Department of
Radiology, University of Florida, Gainesville, Fla (J.R.G.)
| | - Qian Li
- From the Center for Ultrasound Research & Translation,
Department of Radiology, Massachusetts General Hospital, 101 Merrimac St, 3rd
Floor, 323G, Boston, MA 02114 (A.O., V.K., T.T.P., Q.L., M.B., P.G., S.S., Y.G.,
S.D., A.E.S.); Harvard Medical School, Boston, Mass (A.O., V.K., T.T.P, Q.L.,
A.E.S.); Departments of Medical Physics and Radiology, University of Wisconsin,
Madison, Wis (I.R.M.); GE HealthCare, Milwaukee, Wis (M.W.); and Department of
Radiology, University of Florida, Gainesville, Fla (J.R.G.)
| | - Masoud Baikpour
- From the Center for Ultrasound Research & Translation,
Department of Radiology, Massachusetts General Hospital, 101 Merrimac St, 3rd
Floor, 323G, Boston, MA 02114 (A.O., V.K., T.T.P., Q.L., M.B., P.G., S.S., Y.G.,
S.D., A.E.S.); Harvard Medical School, Boston, Mass (A.O., V.K., T.T.P, Q.L.,
A.E.S.); Departments of Medical Physics and Radiology, University of Wisconsin,
Madison, Wis (I.R.M.); GE HealthCare, Milwaukee, Wis (M.W.); and Department of
Radiology, University of Florida, Gainesville, Fla (J.R.G.)
| | - Ivan Rosado-Mendez
- From the Center for Ultrasound Research & Translation,
Department of Radiology, Massachusetts General Hospital, 101 Merrimac St, 3rd
Floor, 323G, Boston, MA 02114 (A.O., V.K., T.T.P., Q.L., M.B., P.G., S.S., Y.G.,
S.D., A.E.S.); Harvard Medical School, Boston, Mass (A.O., V.K., T.T.P, Q.L.,
A.E.S.); Departments of Medical Physics and Radiology, University of Wisconsin,
Madison, Wis (I.R.M.); GE HealthCare, Milwaukee, Wis (M.W.); and Department of
Radiology, University of Florida, Gainesville, Fla (J.R.G.)
| | - Michael Wang
- From the Center for Ultrasound Research & Translation,
Department of Radiology, Massachusetts General Hospital, 101 Merrimac St, 3rd
Floor, 323G, Boston, MA 02114 (A.O., V.K., T.T.P., Q.L., M.B., P.G., S.S., Y.G.,
S.D., A.E.S.); Harvard Medical School, Boston, Mass (A.O., V.K., T.T.P, Q.L.,
A.E.S.); Departments of Medical Physics and Radiology, University of Wisconsin,
Madison, Wis (I.R.M.); GE HealthCare, Milwaukee, Wis (M.W.); and Department of
Radiology, University of Florida, Gainesville, Fla (J.R.G.)
| | - Peng Guo
- From the Center for Ultrasound Research & Translation,
Department of Radiology, Massachusetts General Hospital, 101 Merrimac St, 3rd
Floor, 323G, Boston, MA 02114 (A.O., V.K., T.T.P., Q.L., M.B., P.G., S.S., Y.G.,
S.D., A.E.S.); Harvard Medical School, Boston, Mass (A.O., V.K., T.T.P, Q.L.,
A.E.S.); Departments of Medical Physics and Radiology, University of Wisconsin,
Madison, Wis (I.R.M.); GE HealthCare, Milwaukee, Wis (M.W.); and Department of
Radiology, University of Florida, Gainesville, Fla (J.R.G.)
| | - Scott Schoen
- From the Center for Ultrasound Research & Translation,
Department of Radiology, Massachusetts General Hospital, 101 Merrimac St, 3rd
Floor, 323G, Boston, MA 02114 (A.O., V.K., T.T.P., Q.L., M.B., P.G., S.S., Y.G.,
S.D., A.E.S.); Harvard Medical School, Boston, Mass (A.O., V.K., T.T.P, Q.L.,
A.E.S.); Departments of Medical Physics and Radiology, University of Wisconsin,
Madison, Wis (I.R.M.); GE HealthCare, Milwaukee, Wis (M.W.); and Department of
Radiology, University of Florida, Gainesville, Fla (J.R.G.)
| | - Yuyang Gu
- From the Center for Ultrasound Research & Translation,
Department of Radiology, Massachusetts General Hospital, 101 Merrimac St, 3rd
Floor, 323G, Boston, MA 02114 (A.O., V.K., T.T.P., Q.L., M.B., P.G., S.S., Y.G.,
S.D., A.E.S.); Harvard Medical School, Boston, Mass (A.O., V.K., T.T.P, Q.L.,
A.E.S.); Departments of Medical Physics and Radiology, University of Wisconsin,
Madison, Wis (I.R.M.); GE HealthCare, Milwaukee, Wis (M.W.); and Department of
Radiology, University of Florida, Gainesville, Fla (J.R.G.)
| | - Sunethra Dayavansha
- From the Center for Ultrasound Research & Translation,
Department of Radiology, Massachusetts General Hospital, 101 Merrimac St, 3rd
Floor, 323G, Boston, MA 02114 (A.O., V.K., T.T.P., Q.L., M.B., P.G., S.S., Y.G.,
S.D., A.E.S.); Harvard Medical School, Boston, Mass (A.O., V.K., T.T.P, Q.L.,
A.E.S.); Departments of Medical Physics and Radiology, University of Wisconsin,
Madison, Wis (I.R.M.); GE HealthCare, Milwaukee, Wis (M.W.); and Department of
Radiology, University of Florida, Gainesville, Fla (J.R.G.)
| | - Joseph R. Grajo
- From the Center for Ultrasound Research & Translation,
Department of Radiology, Massachusetts General Hospital, 101 Merrimac St, 3rd
Floor, 323G, Boston, MA 02114 (A.O., V.K., T.T.P., Q.L., M.B., P.G., S.S., Y.G.,
S.D., A.E.S.); Harvard Medical School, Boston, Mass (A.O., V.K., T.T.P, Q.L.,
A.E.S.); Departments of Medical Physics and Radiology, University of Wisconsin,
Madison, Wis (I.R.M.); GE HealthCare, Milwaukee, Wis (M.W.); and Department of
Radiology, University of Florida, Gainesville, Fla (J.R.G.)
| | - Anthony E. Samir
- From the Center for Ultrasound Research & Translation,
Department of Radiology, Massachusetts General Hospital, 101 Merrimac St, 3rd
Floor, 323G, Boston, MA 02114 (A.O., V.K., T.T.P., Q.L., M.B., P.G., S.S., Y.G.,
S.D., A.E.S.); Harvard Medical School, Boston, Mass (A.O., V.K., T.T.P, Q.L.,
A.E.S.); Departments of Medical Physics and Radiology, University of Wisconsin,
Madison, Wis (I.R.M.); GE HealthCare, Milwaukee, Wis (M.W.); and Department of
Radiology, University of Florida, Gainesville, Fla (J.R.G.)
| |
Collapse
|
17
|
Wang S, Friedman SL. Found in translation-Fibrosis in metabolic dysfunction-associated steatohepatitis (MASH). Sci Transl Med 2023; 15:eadi0759. [PMID: 37792957 PMCID: PMC10671253 DOI: 10.1126/scitranslmed.adi0759] [Citation(s) in RCA: 49] [Impact Index Per Article: 24.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2023] [Accepted: 09/15/2023] [Indexed: 10/06/2023]
Abstract
Metabolic dysfunction-associated steatohepatitis (MASH) is a severe form of liver disease that poses a global health threat because of its potential to progress to advanced fibrosis, leading to cirrhosis and liver cancer. Recent advances in single-cell methodologies, refined disease models, and genetic and epigenetic insights have provided a nuanced understanding of MASH fibrogenesis, with substantial cellular heterogeneity in MASH livers providing potentially targetable cell-cell interactions and behavior. Unlike fibrogenesis, mechanisms underlying fibrosis regression in MASH are still inadequately understood, although antifibrotic targets have been recently identified. A refined antifibrotic treatment framework could lead to noninvasive assessment and targeted therapies that preserve hepatocellular function and restore the liver's architectural integrity.
Collapse
Affiliation(s)
- Shuang Wang
- Division of Liver Diseases, Icahn School of Medicine at Mount Sinai, New York, NY 10029
| | - Scott L. Friedman
- Division of Liver Diseases, Icahn School of Medicine at Mount Sinai, New York, NY 10029
| |
Collapse
|
18
|
Fowler KJ, Venkatesh SK, Obuchowski N, Middleton MS, Chen J, Pepin K, Magnuson J, Brown KJ, Batakis D, Henderson WC, Shankar SS, Kamphaus TN, Pasek A, Calle RA, Sanyal AJ, Loomba R, Ehman R, Samir AE, Sirlin CB, Sherlock SP. Repeatability of MRI Biomarkers in Nonalcoholic Fatty Liver Disease: The NIMBLE Consortium. Radiology 2023; 309:e231092. [PMID: 37815451 PMCID: PMC10625902 DOI: 10.1148/radiol.231092] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2023] [Revised: 07/30/2023] [Accepted: 08/29/2023] [Indexed: 10/11/2023]
Abstract
Background There is a need for reliable noninvasive methods for diagnosing and monitoring nonalcoholic fatty liver disease (NAFLD). Thus, the multidisciplinary Non-invasive Biomarkers of Metabolic Liver disease (NIMBLE) consortium was formed to identify and advance the regulatory qualification of NAFLD imaging biomarkers. Purpose To determine the different-day same-scanner repeatability coefficient of liver MRI biomarkers in patients with NAFLD at risk for steatohepatitis. Materials and Methods NIMBLE 1.2 is a prospective, observational, single-center short-term cross-sectional study (October 2021 to June 2022) in adults with NAFLD across a spectrum of low, intermediate, and high likelihood of advanced fibrosis as determined according to the fibrosis based on four factors (FIB-4) index. Participants underwent up to seven MRI examinations across two visits less than or equal to 7 days apart. Standardized imaging protocols were implemented with six MRI scanners from three vendors at both 1.5 T and 3 T, with central analysis of the data performed by an independent reading center (University of California, San Diego). Trained analysts, who were blinded to clinical data, measured the MRI proton density fat fraction (PDFF), liver stiffness at MR elastography (MRE), and visceral adipose tissue (VAT) for each participant. Point estimates and CIs were calculated using χ2 distribution and statistical modeling for pooled repeatability measures. Results A total of 17 participants (mean age, 58 years ± 8.5 [SD]; 10 female) were included, of which seven (41.2%), six (35.3%), and four (23.5%) participants had a low, intermediate, or high likelihood of advanced fibrosis, respectively. The different-day same-scanner mean measurements were 13%-14% for PDFF, 6.6 L for VAT, and 3.15 kPa for two-dimensional MRE stiffness. The different-day same-scanner repeatability coefficients were 0.22 L (95% CI: 0.17, 0.29) for VAT, 0.75 kPa (95% CI: 0.6, 0.99) for MRE stiffness, 1.19% (95% CI: 0.96, 1.61) for MRI PDFF using magnitude reconstruction, 1.56% (95% CI: 1.26, 2.07) for MRI PDFF using complex reconstruction, and 19.7% (95% CI: 15.8, 26.2) for three-dimensional MRE shear modulus. Conclusion This preliminary study suggests that thresholds of 1.2%-1.6%, 0.22 L, and 0.75 kPa for MRI PDFF, VAT, and MRE, respectively, should be used to discern measurement error from real change in patients with NAFLD. ClinicalTrials.gov registration no. NCT05081427 © RSNA, 2023 Supplemental material is available for this article. See also the editorial by Kozaka and Matsui in this issue.
Collapse
Affiliation(s)
| | | | - Nancy Obuchowski
- From the Liver Imaging Group (K.J.F., M.S.M., D.B., W.C.H., C.B.S.)
and Department of Hepatology (R.L.), University of California–San Diego,
6206 Lakewood St, San Diego, CA 92122; Department of Radiology, Mayo Clinic,
Rochester, Minn (S.K.V., J.C., K.P., J.M., K.J.B., R.E.); Department of
Quantitative Health Sciences, Cleveland Clinic, Cleveland, Ohio (N.O.); Pfizer
Research and Development, Pfizer, Inc, Sacramento, Calif (S.S.S.); Foundation
for the National Institutes of Health, North Bethesda, Md (T.N.K., A.P.);
Regeneron Pharmaceuticals, Inc, Tarrytown, NY (R.A.C.); Department of
Gastroenterology, Virginia Commonwealth University, Richmond, Va (A.J.S.);
Department of Radiology, Massachusetts General Hospital, Boston, Mass (A.E.S.);
and Department of Imaging Alliances, Pfizer, Inc, New York, NY (S.P.S.)
| | - Michael S. Middleton
- From the Liver Imaging Group (K.J.F., M.S.M., D.B., W.C.H., C.B.S.)
and Department of Hepatology (R.L.), University of California–San Diego,
6206 Lakewood St, San Diego, CA 92122; Department of Radiology, Mayo Clinic,
Rochester, Minn (S.K.V., J.C., K.P., J.M., K.J.B., R.E.); Department of
Quantitative Health Sciences, Cleveland Clinic, Cleveland, Ohio (N.O.); Pfizer
Research and Development, Pfizer, Inc, Sacramento, Calif (S.S.S.); Foundation
for the National Institutes of Health, North Bethesda, Md (T.N.K., A.P.);
Regeneron Pharmaceuticals, Inc, Tarrytown, NY (R.A.C.); Department of
Gastroenterology, Virginia Commonwealth University, Richmond, Va (A.J.S.);
Department of Radiology, Massachusetts General Hospital, Boston, Mass (A.E.S.);
and Department of Imaging Alliances, Pfizer, Inc, New York, NY (S.P.S.)
| | - Jun Chen
- From the Liver Imaging Group (K.J.F., M.S.M., D.B., W.C.H., C.B.S.)
and Department of Hepatology (R.L.), University of California–San Diego,
6206 Lakewood St, San Diego, CA 92122; Department of Radiology, Mayo Clinic,
Rochester, Minn (S.K.V., J.C., K.P., J.M., K.J.B., R.E.); Department of
Quantitative Health Sciences, Cleveland Clinic, Cleveland, Ohio (N.O.); Pfizer
Research and Development, Pfizer, Inc, Sacramento, Calif (S.S.S.); Foundation
for the National Institutes of Health, North Bethesda, Md (T.N.K., A.P.);
Regeneron Pharmaceuticals, Inc, Tarrytown, NY (R.A.C.); Department of
Gastroenterology, Virginia Commonwealth University, Richmond, Va (A.J.S.);
Department of Radiology, Massachusetts General Hospital, Boston, Mass (A.E.S.);
and Department of Imaging Alliances, Pfizer, Inc, New York, NY (S.P.S.)
| | - Kay Pepin
- From the Liver Imaging Group (K.J.F., M.S.M., D.B., W.C.H., C.B.S.)
and Department of Hepatology (R.L.), University of California–San Diego,
6206 Lakewood St, San Diego, CA 92122; Department of Radiology, Mayo Clinic,
Rochester, Minn (S.K.V., J.C., K.P., J.M., K.J.B., R.E.); Department of
Quantitative Health Sciences, Cleveland Clinic, Cleveland, Ohio (N.O.); Pfizer
Research and Development, Pfizer, Inc, Sacramento, Calif (S.S.S.); Foundation
for the National Institutes of Health, North Bethesda, Md (T.N.K., A.P.);
Regeneron Pharmaceuticals, Inc, Tarrytown, NY (R.A.C.); Department of
Gastroenterology, Virginia Commonwealth University, Richmond, Va (A.J.S.);
Department of Radiology, Massachusetts General Hospital, Boston, Mass (A.E.S.);
and Department of Imaging Alliances, Pfizer, Inc, New York, NY (S.P.S.)
| | - Jessica Magnuson
- From the Liver Imaging Group (K.J.F., M.S.M., D.B., W.C.H., C.B.S.)
and Department of Hepatology (R.L.), University of California–San Diego,
6206 Lakewood St, San Diego, CA 92122; Department of Radiology, Mayo Clinic,
Rochester, Minn (S.K.V., J.C., K.P., J.M., K.J.B., R.E.); Department of
Quantitative Health Sciences, Cleveland Clinic, Cleveland, Ohio (N.O.); Pfizer
Research and Development, Pfizer, Inc, Sacramento, Calif (S.S.S.); Foundation
for the National Institutes of Health, North Bethesda, Md (T.N.K., A.P.);
Regeneron Pharmaceuticals, Inc, Tarrytown, NY (R.A.C.); Department of
Gastroenterology, Virginia Commonwealth University, Richmond, Va (A.J.S.);
Department of Radiology, Massachusetts General Hospital, Boston, Mass (A.E.S.);
and Department of Imaging Alliances, Pfizer, Inc, New York, NY (S.P.S.)
| | - Kathy J. Brown
- From the Liver Imaging Group (K.J.F., M.S.M., D.B., W.C.H., C.B.S.)
and Department of Hepatology (R.L.), University of California–San Diego,
6206 Lakewood St, San Diego, CA 92122; Department of Radiology, Mayo Clinic,
Rochester, Minn (S.K.V., J.C., K.P., J.M., K.J.B., R.E.); Department of
Quantitative Health Sciences, Cleveland Clinic, Cleveland, Ohio (N.O.); Pfizer
Research and Development, Pfizer, Inc, Sacramento, Calif (S.S.S.); Foundation
for the National Institutes of Health, North Bethesda, Md (T.N.K., A.P.);
Regeneron Pharmaceuticals, Inc, Tarrytown, NY (R.A.C.); Department of
Gastroenterology, Virginia Commonwealth University, Richmond, Va (A.J.S.);
Department of Radiology, Massachusetts General Hospital, Boston, Mass (A.E.S.);
and Department of Imaging Alliances, Pfizer, Inc, New York, NY (S.P.S.)
| | - Danielle Batakis
- From the Liver Imaging Group (K.J.F., M.S.M., D.B., W.C.H., C.B.S.)
and Department of Hepatology (R.L.), University of California–San Diego,
6206 Lakewood St, San Diego, CA 92122; Department of Radiology, Mayo Clinic,
Rochester, Minn (S.K.V., J.C., K.P., J.M., K.J.B., R.E.); Department of
Quantitative Health Sciences, Cleveland Clinic, Cleveland, Ohio (N.O.); Pfizer
Research and Development, Pfizer, Inc, Sacramento, Calif (S.S.S.); Foundation
for the National Institutes of Health, North Bethesda, Md (T.N.K., A.P.);
Regeneron Pharmaceuticals, Inc, Tarrytown, NY (R.A.C.); Department of
Gastroenterology, Virginia Commonwealth University, Richmond, Va (A.J.S.);
Department of Radiology, Massachusetts General Hospital, Boston, Mass (A.E.S.);
and Department of Imaging Alliances, Pfizer, Inc, New York, NY (S.P.S.)
| | - Walter C. Henderson
- From the Liver Imaging Group (K.J.F., M.S.M., D.B., W.C.H., C.B.S.)
and Department of Hepatology (R.L.), University of California–San Diego,
6206 Lakewood St, San Diego, CA 92122; Department of Radiology, Mayo Clinic,
Rochester, Minn (S.K.V., J.C., K.P., J.M., K.J.B., R.E.); Department of
Quantitative Health Sciences, Cleveland Clinic, Cleveland, Ohio (N.O.); Pfizer
Research and Development, Pfizer, Inc, Sacramento, Calif (S.S.S.); Foundation
for the National Institutes of Health, North Bethesda, Md (T.N.K., A.P.);
Regeneron Pharmaceuticals, Inc, Tarrytown, NY (R.A.C.); Department of
Gastroenterology, Virginia Commonwealth University, Richmond, Va (A.J.S.);
Department of Radiology, Massachusetts General Hospital, Boston, Mass (A.E.S.);
and Department of Imaging Alliances, Pfizer, Inc, New York, NY (S.P.S.)
| | - Sudha S. Shankar
- From the Liver Imaging Group (K.J.F., M.S.M., D.B., W.C.H., C.B.S.)
and Department of Hepatology (R.L.), University of California–San Diego,
6206 Lakewood St, San Diego, CA 92122; Department of Radiology, Mayo Clinic,
Rochester, Minn (S.K.V., J.C., K.P., J.M., K.J.B., R.E.); Department of
Quantitative Health Sciences, Cleveland Clinic, Cleveland, Ohio (N.O.); Pfizer
Research and Development, Pfizer, Inc, Sacramento, Calif (S.S.S.); Foundation
for the National Institutes of Health, North Bethesda, Md (T.N.K., A.P.);
Regeneron Pharmaceuticals, Inc, Tarrytown, NY (R.A.C.); Department of
Gastroenterology, Virginia Commonwealth University, Richmond, Va (A.J.S.);
Department of Radiology, Massachusetts General Hospital, Boston, Mass (A.E.S.);
and Department of Imaging Alliances, Pfizer, Inc, New York, NY (S.P.S.)
| | - Tania N. Kamphaus
- From the Liver Imaging Group (K.J.F., M.S.M., D.B., W.C.H., C.B.S.)
and Department of Hepatology (R.L.), University of California–San Diego,
6206 Lakewood St, San Diego, CA 92122; Department of Radiology, Mayo Clinic,
Rochester, Minn (S.K.V., J.C., K.P., J.M., K.J.B., R.E.); Department of
Quantitative Health Sciences, Cleveland Clinic, Cleveland, Ohio (N.O.); Pfizer
Research and Development, Pfizer, Inc, Sacramento, Calif (S.S.S.); Foundation
for the National Institutes of Health, North Bethesda, Md (T.N.K., A.P.);
Regeneron Pharmaceuticals, Inc, Tarrytown, NY (R.A.C.); Department of
Gastroenterology, Virginia Commonwealth University, Richmond, Va (A.J.S.);
Department of Radiology, Massachusetts General Hospital, Boston, Mass (A.E.S.);
and Department of Imaging Alliances, Pfizer, Inc, New York, NY (S.P.S.)
| | - Alex Pasek
- From the Liver Imaging Group (K.J.F., M.S.M., D.B., W.C.H., C.B.S.)
and Department of Hepatology (R.L.), University of California–San Diego,
6206 Lakewood St, San Diego, CA 92122; Department of Radiology, Mayo Clinic,
Rochester, Minn (S.K.V., J.C., K.P., J.M., K.J.B., R.E.); Department of
Quantitative Health Sciences, Cleveland Clinic, Cleveland, Ohio (N.O.); Pfizer
Research and Development, Pfizer, Inc, Sacramento, Calif (S.S.S.); Foundation
for the National Institutes of Health, North Bethesda, Md (T.N.K., A.P.);
Regeneron Pharmaceuticals, Inc, Tarrytown, NY (R.A.C.); Department of
Gastroenterology, Virginia Commonwealth University, Richmond, Va (A.J.S.);
Department of Radiology, Massachusetts General Hospital, Boston, Mass (A.E.S.);
and Department of Imaging Alliances, Pfizer, Inc, New York, NY (S.P.S.)
| | - Roberto A. Calle
- From the Liver Imaging Group (K.J.F., M.S.M., D.B., W.C.H., C.B.S.)
and Department of Hepatology (R.L.), University of California–San Diego,
6206 Lakewood St, San Diego, CA 92122; Department of Radiology, Mayo Clinic,
Rochester, Minn (S.K.V., J.C., K.P., J.M., K.J.B., R.E.); Department of
Quantitative Health Sciences, Cleveland Clinic, Cleveland, Ohio (N.O.); Pfizer
Research and Development, Pfizer, Inc, Sacramento, Calif (S.S.S.); Foundation
for the National Institutes of Health, North Bethesda, Md (T.N.K., A.P.);
Regeneron Pharmaceuticals, Inc, Tarrytown, NY (R.A.C.); Department of
Gastroenterology, Virginia Commonwealth University, Richmond, Va (A.J.S.);
Department of Radiology, Massachusetts General Hospital, Boston, Mass (A.E.S.);
and Department of Imaging Alliances, Pfizer, Inc, New York, NY (S.P.S.)
| | - Arun J. Sanyal
- From the Liver Imaging Group (K.J.F., M.S.M., D.B., W.C.H., C.B.S.)
and Department of Hepatology (R.L.), University of California–San Diego,
6206 Lakewood St, San Diego, CA 92122; Department of Radiology, Mayo Clinic,
Rochester, Minn (S.K.V., J.C., K.P., J.M., K.J.B., R.E.); Department of
Quantitative Health Sciences, Cleveland Clinic, Cleveland, Ohio (N.O.); Pfizer
Research and Development, Pfizer, Inc, Sacramento, Calif (S.S.S.); Foundation
for the National Institutes of Health, North Bethesda, Md (T.N.K., A.P.);
Regeneron Pharmaceuticals, Inc, Tarrytown, NY (R.A.C.); Department of
Gastroenterology, Virginia Commonwealth University, Richmond, Va (A.J.S.);
Department of Radiology, Massachusetts General Hospital, Boston, Mass (A.E.S.);
and Department of Imaging Alliances, Pfizer, Inc, New York, NY (S.P.S.)
| | - Rohit Loomba
- From the Liver Imaging Group (K.J.F., M.S.M., D.B., W.C.H., C.B.S.)
and Department of Hepatology (R.L.), University of California–San Diego,
6206 Lakewood St, San Diego, CA 92122; Department of Radiology, Mayo Clinic,
Rochester, Minn (S.K.V., J.C., K.P., J.M., K.J.B., R.E.); Department of
Quantitative Health Sciences, Cleveland Clinic, Cleveland, Ohio (N.O.); Pfizer
Research and Development, Pfizer, Inc, Sacramento, Calif (S.S.S.); Foundation
for the National Institutes of Health, North Bethesda, Md (T.N.K., A.P.);
Regeneron Pharmaceuticals, Inc, Tarrytown, NY (R.A.C.); Department of
Gastroenterology, Virginia Commonwealth University, Richmond, Va (A.J.S.);
Department of Radiology, Massachusetts General Hospital, Boston, Mass (A.E.S.);
and Department of Imaging Alliances, Pfizer, Inc, New York, NY (S.P.S.)
| | - Richard Ehman
- From the Liver Imaging Group (K.J.F., M.S.M., D.B., W.C.H., C.B.S.)
and Department of Hepatology (R.L.), University of California–San Diego,
6206 Lakewood St, San Diego, CA 92122; Department of Radiology, Mayo Clinic,
Rochester, Minn (S.K.V., J.C., K.P., J.M., K.J.B., R.E.); Department of
Quantitative Health Sciences, Cleveland Clinic, Cleveland, Ohio (N.O.); Pfizer
Research and Development, Pfizer, Inc, Sacramento, Calif (S.S.S.); Foundation
for the National Institutes of Health, North Bethesda, Md (T.N.K., A.P.);
Regeneron Pharmaceuticals, Inc, Tarrytown, NY (R.A.C.); Department of
Gastroenterology, Virginia Commonwealth University, Richmond, Va (A.J.S.);
Department of Radiology, Massachusetts General Hospital, Boston, Mass (A.E.S.);
and Department of Imaging Alliances, Pfizer, Inc, New York, NY (S.P.S.)
| | - Anthony E. Samir
- From the Liver Imaging Group (K.J.F., M.S.M., D.B., W.C.H., C.B.S.)
and Department of Hepatology (R.L.), University of California–San Diego,
6206 Lakewood St, San Diego, CA 92122; Department of Radiology, Mayo Clinic,
Rochester, Minn (S.K.V., J.C., K.P., J.M., K.J.B., R.E.); Department of
Quantitative Health Sciences, Cleveland Clinic, Cleveland, Ohio (N.O.); Pfizer
Research and Development, Pfizer, Inc, Sacramento, Calif (S.S.S.); Foundation
for the National Institutes of Health, North Bethesda, Md (T.N.K., A.P.);
Regeneron Pharmaceuticals, Inc, Tarrytown, NY (R.A.C.); Department of
Gastroenterology, Virginia Commonwealth University, Richmond, Va (A.J.S.);
Department of Radiology, Massachusetts General Hospital, Boston, Mass (A.E.S.);
and Department of Imaging Alliances, Pfizer, Inc, New York, NY (S.P.S.)
| | | | | |
Collapse
|
19
|
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
|
20
|
Kozaka K, Matsui O. MRI Biomarkers and Their Future Impact on Nonalcoholic Fatty Liver Disease. Radiology 2023; 309:e232420. [PMID: 37815446 DOI: 10.1148/radiol.232420] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/11/2023]
Affiliation(s)
- Kazuto Kozaka
- From the Department of Radiology, Kanazawa University Graduate School of Medical Sciences, 13-1, Takaramachi, Kanazawa, Ishikawa 920-8641, Japan
| | - Osamu Matsui
- From the Department of Radiology, Kanazawa University Graduate School of Medical Sciences, 13-1, Takaramachi, Kanazawa, Ishikawa 920-8641, Japan
| |
Collapse
|
21
|
|
22
|
Ma H, Zhou IY, Chen YI, Rotile NJ, Ay I, Akam EA, Wang H, Knipe RS, Hariri LP, Zhang C, Drummond M, Pantazopoulos P, Moon BF, Boice AT, Zygmont SE, Weigand-Whittier J, Sojoodi M, Gonzalez-Villalobos RA, Hansen MK, Tanabe KK, Caravan P. Tailored Chemical Reactivity Probes for Systemic Imaging of Aldehydes in Fibroproliferative Diseases. J Am Chem Soc 2023; 145:20825-20836. [PMID: 37589185 PMCID: PMC11022681 DOI: 10.1021/jacs.3c04964] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/18/2023]
Abstract
During fibroproliferation, protein-associated extracellular aldehydes are formed by the oxidation of lysine residues on extracellular matrix proteins to form the aldehyde allysine. Here we report three Mn(II)-based, small-molecule magnetic resonance probes that contain α-effect nucleophiles to target allysine in vivo and report on tissue fibrogenesis. We used a rational design approach to develop turn-on probes with a 4-fold increase in relaxivity upon targeting. The effects of aldehyde condensation rate and hydrolysis kinetics on the performance of the probes to detect tissue fibrogenesis non-invasively in mouse models were evaluated by a systemic aldehyde tracking approach. We showed that, for highly reversible ligations, off-rate was a stronger predictor of in vivo efficiency, enabling histologically validated, three-dimensional characterization of pulmonary fibrogenesis throughout the entire lung. The exclusive renal elimination of these probes allowed for rapid imaging of liver fibrosis. Reducing the hydrolysis rate by forming an oxime bond with allysine enabled delayed phase imaging of kidney fibrogenesis. The imaging efficacy of these probes, coupled with their rapid and complete elimination from the body, makes them strong candidates for clinical translation.
Collapse
Affiliation(s)
- Hua Ma
- Athinoula A. Martinos Center for Biomedical Imaging, Institute for Innovation in Imaging (i), Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts 02129, United States
| | - Iris Y. Zhou
- Athinoula A. Martinos Center for Biomedical Imaging, Institute for Innovation in Imaging (i), Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts 02129, United States
| | - Y. Iris Chen
- Athinoula A. Martinos Center for Biomedical Imaging, Institute for Innovation in Imaging (i), Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts 02129, United States
| | - Nicholas J. Rotile
- Athinoula A. Martinos Center for Biomedical Imaging, Institute for Innovation in Imaging (i), Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts 02129, United States
| | - Ilknur Ay
- Athinoula A. Martinos Center for Biomedical Imaging, Institute for Innovation in Imaging (i), Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts 02129, United States
| | - Eman A. Akam
- Athinoula A. Martinos Center for Biomedical Imaging, Institute for Innovation in Imaging (i), Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts 02129, United States
| | - Huan Wang
- Athinoula A. Martinos Center for Biomedical Imaging, Institute for Innovation in Imaging (i), Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts 02129, United States
| | - Rachel S. Knipe
- Division of Pulmonary and Critical Care Medicine and the Center for Immunology and Inflammatory Diseases, Massachusetts General Hospital, Boston, Massachusetts 02114, United States
| | - Lida P. Hariri
- Division of Pulmonary and Critical Care Medicine and the Center for Immunology and Inflammatory Diseases, Massachusetts General Hospital, Boston, Massachusetts 02114, United States
- Department of Pathology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts 02114, United States
| | - Caiyuan Zhang
- Athinoula A. Martinos Center for Biomedical Imaging, Institute for Innovation in Imaging (i), Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts 02129, United States
| | - Matthew Drummond
- Division of Pulmonary and Critical Care Medicine and the Center for Immunology and Inflammatory Diseases, Massachusetts General Hospital, Boston, Massachusetts 02114, United States
| | - Pamela Pantazopoulos
- Athinoula A. Martinos Center for Biomedical Imaging, Institute for Innovation in Imaging (i), Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts 02129, United States
| | - Brianna F. Moon
- Athinoula A. Martinos Center for Biomedical Imaging, Institute for Innovation in Imaging (i), Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts 02129, United States
| | - Avery T. Boice
- Athinoula A. Martinos Center for Biomedical Imaging, Institute for Innovation in Imaging (i), Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts 02129, United States
| | - Samantha E. Zygmont
- Athinoula A. Martinos Center for Biomedical Imaging, Institute for Innovation in Imaging (i), Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts 02129, United States
| | - Jonah Weigand-Whittier
- Athinoula A. Martinos Center for Biomedical Imaging, Institute for Innovation in Imaging (i), Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts 02129, United States
| | - Mozhdeh Sojoodi
- Division of Gastrointestinal and Oncologic Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts 02114, United States
| | - Romer A. Gonzalez-Villalobos
- Cardiovascular and Metabolism Discovery, Janssen Research and Development LLC, Boston, Massachusetts 02115, United States
| | - Michael K. Hansen
- Cardiovascular and Metabolism Discovery, Janssen Research and Development LLC, Boston, Massachusetts 02115, United States
| | - Kenneth K. Tanabe
- Division of Gastrointestinal and Oncologic Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts 02114, United States
| | - Peter Caravan
- Athinoula A. Martinos Center for Biomedical Imaging, Institute for Innovation in Imaging (i), Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts 02129, United States
| |
Collapse
|
23
|
Abstract
Clinical trials have been a central driver of change and have provided the evidence base necessary to advance new therapies for liver diseases. This review provides a perspective on the status of trials in hepatology and a vantage point into the emerging capabilities and external forces that will shape the conduct of clinical trials in the future. The adaptations to clinical trial operations in response to the disruptions by the COVID-19 pandemic and opportunities for innovation in hepatology trials are emphasized. Future trials in hepatology will be driven by unmet therapeutic needs and fueled by technological advances incorporating digital capabilities with expanded participant-derived data collection, computing, and analytics. Their design will embrace innovative trial designs adapted to these advances and that emphasize broader and more inclusive participant engagement. Their conduct will be further shaped by evolving regulatory needs and the emergence of new stakeholders in the clinical trials ecosystem. The evolution of clinical trials will offer unique opportunities to advance new therapeutics that will ultimately improve the lives of patients with liver diseases.
Collapse
Affiliation(s)
- Paul Y Kwo
- Department of Medicine, Stanford University School of Medicine, Palo Alto, California, USA
| | - Tushar Patel
- Department of Transplantation, Mayo Clinic, Jacksonville, Florida, USA
| |
Collapse
|
24
|
Kanagalingam G, Sanyal AJ. NAFLD: Diagnostics and New Nomenclature. J Clin Exp Hepatol 2023; 13:723-724. [PMID: 37693255 PMCID: PMC10482989 DOI: 10.1016/j.jceh.2023.07.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 09/12/2023] Open
Affiliation(s)
- Gowthami Kanagalingam
- Division of Gastroenterology, Hepatology and Nutrition, Virginia Commonwealth University School of Medicine, Richmond, VA, USA
| | - Arun J Sanyal
- Stravitz-Sanyal Institute for Liver Disease and Metabolic Health, Virginia Commonwealth University School of Medicine, Richmond, VA, USA
| |
Collapse
|
25
|
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
|
26
|
Schattenberg JM, Chalasani N, Alkhouri N. Artificial Intelligence Applications in Hepatology. Clin Gastroenterol Hepatol 2023; 21:2015-2025. [PMID: 37088460 DOI: 10.1016/j.cgh.2023.04.007] [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: 10/09/2022] [Revised: 03/16/2023] [Accepted: 04/14/2023] [Indexed: 04/25/2023]
Abstract
Over the past 2 decades, the field of hepatology has witnessed major developments in diagnostic tools, prognostic models, and treatment options making it one of the most complex medical subspecialties. Through artificial intelligence (AI) and machine learning, computers are now able to learn from complex and diverse clinical datasets to solve real-world medical problems with performance that surpasses that of physicians in certain areas. AI algorithms are currently being implemented in liver imaging, interpretation of liver histopathology, noninvasive tests, prediction models, and more. In this review, we provide a summary of the state of AI in hepatology and discuss current challenges for large-scale implementation including some ethical aspects. We emphasize to the readers that most AI-based algorithms that are discussed in this review are still considered in early development and their utility and impact on patient outcomes still need to be assessed in future large-scale and inclusive studies. Our vision is that the use of AI in hepatology will enhance physician performance, decrease the burden and time spent on documentation, and reestablish the personalized patient-physician relationship that is of utmost importance for obtaining good outcomes.
Collapse
Affiliation(s)
- Jörn M Schattenberg
- Metabolic Liver Research Program, I. Department of Medicine, University Medical Center Mainz, Mainz, Germany
| | - Naga Chalasani
- Indiana University School of Medicine and Indiana University Health, Indianapolis, Indiana
| | - Naim Alkhouri
- Arizona Liver Health and University of Arizona, Tucson, Arizona.
| |
Collapse
|
27
|
Iruzubieta P, Bataller R, Arias-Loste MT, Arrese M, Calleja JL, Castro-Narro G, Cusi K, Dillon JF, Martínez-Chantar ML, Mateo M, Pérez A, Rinella ME, Romero-Gómez M, Schattenberg JM, Zelber-Sagi S, Crespo J, Lazarus JV. Research Priorities for Precision Medicine in NAFLD. Clin Liver Dis 2023; 27:535-551. [PMID: 37024222 DOI: 10.1016/j.cld.2023.01.016] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/08/2023]
Abstract
NAFLD is a multisystem condition and the leading cause of chronic liver disease globally. There are no approved NAFLD-specific dugs. To advance in the prevention and treatment of NAFLD, there is a clear need to better understand the pathophysiology and genetic and environmental risk factors, identify subphenotypes, and develop personalized and precision medicine. In this review, we discuss the main NAFLD research priorities, with a particular focus on socioeconomic factors, interindividual variations, limitations of current NAFLD clinical trials, multidisciplinary models of care, and novel approaches in the management of patients with NAFLD.
Collapse
Affiliation(s)
- Paula Iruzubieta
- Gastroenterology and Hepatology Department, Clinical and Translational Research in Digestive Diseases, Valdecilla Research Institute (IDIVAL), Marqués de Valdecilla University Hospital, Avenida Valdecilla 25, 39008, Santander, Spain
| | - Ramon Bataller
- Division of Gastroenterology, Hepatology and Nutrition, Center for Liver Diseases, University of Pittsburgh Medical Center, PA, USA
| | - María Teresa Arias-Loste
- Gastroenterology and Hepatology Department, Clinical and Translational Research in Digestive Diseases, Valdecilla Research Institute (IDIVAL), Marqués de Valdecilla University Hospital, Avenida Valdecilla 25, 39008, Santander, Spain
| | - Marco Arrese
- Department of Gastroenterology, School of Medicine, Pontificia Universidad Católica de Chile, Avenida Libertador Bernardo O'Higgins 340, 8331150, Santiago, Chile
| | - José Luis Calleja
- Department of Gastroenterology and Hepatology, Puerta de Hierro University Hospital, Puerta de Hierro Health Research Institute (IDIPHIM), CIBERehd, Universidad Autonoma de Madrid, Calle Joaquín Rodrigo 1, 28222, Majadahonda, Spain
| | - Graciela Castro-Narro
- Department of Gastroenterology, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Department of Hepatology and Transplant, Hospital Médica Sur, Asociación Latinoamericana para el Estudio del Hígado (ALEH), Mexico City, Mexico
| | - Kenneth Cusi
- Division of Endocrinology, Diabetes & Metabolism, Department of Medicine, University of Florida, Gainesville, FL, USA
| | - John F Dillon
- Division of Molecular and Clinical Medicine, University of Dundee, Ninewells Hospital and Medical School, Dundee, UK
| | - María Luz Martínez-Chantar
- Liver Disease Laboratory, Center for Cooperative Research in Biosciences (CIC BioGUNE), Basque Research and Technology Alliance (BRTA), Centro de Investigación Biomedica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Derio, Bizkaia, Spain
| | - Miguel Mateo
- Pharmacy Organisation and Inspection, Government of Cantabria, Santander, Spain
| | - Antonio Pérez
- Endocrinology and Nutrition Department, Santa Creu I Sant Pau Hospital, Universitat Autónoma de Barcelona, IIB-Sant Pau and Centro de Investigación Biomedica en Red de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Barcelona, Spain
| | - Mary E Rinella
- Department of Medicine, University of Chicago, Chicago, IL, USA
| | - Manuel Romero-Gómez
- UCM Digestive Diseases and CIBERehd, Virgen Del Rocío University Hospital, Institute of Biomedicine of Seville, University of Seville, Seville, Spain
| | - Jörn M Schattenberg
- Metabolic Liver Research Program, I. Department of Medicine, University Medical Centre Mainz, Mainz, Germany
| | - Shira Zelber-Sagi
- University of Haifa, School of Public Health, Mount Carmel, Haifa, Israel; Department of Gastroenterology, Tel- Aviv Medical Centre, Tel- Aviv, Israel
| | - Javier Crespo
- Gastroenterology and Hepatology Department, Clinical and Translational Research in Digestive Diseases, Valdecilla Research Institute (IDIVAL), Marqués de Valdecilla University Hospital, Avenida Valdecilla 25, 39008, Santander, Spain.
| | - Jeffrey V Lazarus
- Barcelona Institute for Global Health (ISGlobal), Hospital Clínic, University of Barcelona, Calle del Rossellón 171, ENT-2, Barcelona ES-08036, Spain; Faculty of Medicine and Health Sciences, University of Barcelona, Barcelona, Spain; CUNY Graduate School of Public Health and Health Policy (CUNY SPH), New York, NY, USA.
| |
Collapse
|
28
|
Ma H, Zhou IY, Chen YI, Rotile NJ, Ay I, Akam E, Wang H, Knipe R, Hariri LP, Zhang C, Drummond M, Pantazopoulos P, Moon BF, Boice AT, Zygmont SE, Weigand-Whittier J, Sojoodi M, Gonzalez-Villalobos RA, Hansen MK, Tanabe KK, Caravan P. Tailored chemical reactivity probes for systemic imaging of aldehydes in fibroproliferative diseases. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.04.20.537707. [PMID: 37131719 PMCID: PMC10153247 DOI: 10.1101/2023.04.20.537707] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/04/2023]
Abstract
During fibroproliferation, protein-associated extracellular aldehydes are formed by the oxidation of lysine residues on extracellular matrix proteins to form the aldehyde allysine. Here we report three Mn(II)-based, small molecule magnetic resonance (MR) probes that contain α-effect nucleophiles to target allysine in vivo and report on tissue fibrogenesis. We used a rational design approach to develop turn-on probes with a 4-fold increase in relaxivity upon targeting. The effects of aldehyde condensation rate and hydrolysis kinetics on the performance of the probes to detect tissue fibrogenesis noninvasively in mouse models were evaluated by a systemic aldehyde tracking approach. We showed that for highly reversible ligations, off-rate was a stronger predictor of in vivo efficiency, enabling histologically validated, three-dimensional characterization of pulmonary fibrogenesis throughout the entire lung. The exclusive renal elimination of these probes allowed for rapid imaging of liver fibrosis. Reducing the hydrolysis rate by forming an oxime bond with allysine enabled delayed phase imaging of kidney fibrogenesis. The imaging efficacy of these probes, coupled with their rapid and complete elimination from the body, make them strong candidates for clinical translation.
Collapse
|
29
|
Sanyal AJ, Williams SA, Lavine JE, Neuschwander-Tetri BA, Alexander L, Ostroff R, Biegel H, Kowdley KV, Chalasani N, Dasarathy S, Diehl AM, Loomba R, Hameed B, Behling C, Kleiner DE, Karpen SJ, Williams J, Jia Y, Yates KP, Tonascia J. Defining the serum proteomic signature of hepatic steatosis, inflammation, ballooning and fibrosis in non-alcoholic fatty liver disease. J Hepatol 2023; 78:693-703. [PMID: 36528237 PMCID: PMC10165617 DOI: 10.1016/j.jhep.2022.11.029] [Citation(s) in RCA: 24] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/17/2022] [Revised: 11/01/2022] [Accepted: 11/23/2022] [Indexed: 12/15/2022]
Abstract
BACKGROUND & AIMS Despite recent progress, non-invasive tests for the diagnostic assessment and monitoring of non-alcoholic fatty liver disease (NAFLD) remain an unmet need. Herein, we aimed to identify diagnostic signatures of the key histological features of NAFLD. METHODS Using modified-aptamer proteomics, we assayed 5,220 proteins in each of 2,852 single serum samples from 636 individuals with histologically confirmed NAFLD. We developed and validated dichotomized protein-phenotype models to identify clinically relevant severities of steatosis (grade 0 vs. 1-3), hepatocellular ballooning (0 vs. 1 or 2), lobular inflammation (0-1 vs. 2-3) and fibrosis (stages 0-1 vs. 2-4). RESULTS The AUCs of the four protein models, based on 37 analytes (18 not previously linked to NAFLD), for the diagnosis of their respective components (at a clinically relevant severity) in training/paired validation sets were: fibrosis (AUC 0.92/0.85); steatosis (AUC 0.95/0.79), inflammation (AUC 0.83/0.72), and ballooning (AUC 0.87/0.83). An additional outcome, at-risk NASH, defined as steatohepatitis with NAFLD activity score ≥4 (with a score of at least 1 for each of its components) and fibrosis stage ≥2, was predicted by multiplying the outputs of each individual component model (AUC 0.93/0.85). We further evaluated their ability to detect change in histology following treatment with placebo, pioglitazone, vitamin E or obeticholic acid. Component model scores significantly improved in the active therapies vs. placebo, and differential effects of vitamin E, pioglitazone, and obeticholic acid were identified. CONCLUSIONS Serum protein scanning identified signatures corresponding to the key components of liver biopsy in NAFLD. The models developed were sufficiently sensitive to characterize the longitudinal change for three different drug interventions. These data support continued validation of these proteomic models to enable a "liquid biopsy"-based assessment of NAFLD. CLINICAL TRIAL NUMBER Not applicable. IMPACT AND IMPLICATIONS An aptamer-based protein scan of serum proteins was performed to identify diagnostic signatures of the key histological features of non-alcoholic fatty liver disease (NAFLD), for which no approved non-invasive diagnostic tools are currently available. We also identified specific protein signatures related to the presence and severity of NAFLD and its histological components that were also sensitive to change over time. These are fundamental initial steps in establishing a serum proteome-based diagnostic signature of NASH and provide the rationale for using these signatures to test treatment response and to identify several novel targets for evaluation in the pathogenesis of NAFLD.
Collapse
Affiliation(s)
- Arun J Sanyal
- Stravitz-Sanyal Institute for Liver Disease and Metabolic Health, Virginia Commonwealth University School of Medicine, Richmond, VA, USA.
| | | | - Joel E Lavine
- Dept. of Pediatrics, Columbia University, New York, NY, USA
| | | | | | | | | | | | - Naga Chalasani
- Division of Gastroenterology and Hepatology, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Srinivasan Dasarathy
- Division of Gastroenterology and Hepatology, Cleveland Clinic, Cleveland, OH, USA
| | - Anna Mae Diehl
- Division of Gastroenterology and Hepatology, Duke University School of Medicine, Durham, NC, USA
| | - Rohit Loomba
- NAFLD Research Center, University of California San Diego School of Medicine, San Diego, CA, USA
| | - Bilal Hameed
- Division of Gastroenterology and Hepatology, University of California San Francisco School of Medicine, San Francisco, CA, USA
| | - Cynthia Behling
- NAFLD Research Center, University of California San Diego School of Medicine, San Diego, CA, USA
| | - David E Kleiner
- Laboratory of Pathology, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Saul J Karpen
- Dept. of Pediatrics, Emory University School of Medicine, Atlanta, GA, USA
| | | | - Yi Jia
- Clinical R&D, SomaLogic Inc., Boulder, CO, USA
| | - Katherine P Yates
- Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD, USA
| | - James Tonascia
- Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD, USA
| |
Collapse
|
30
|
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
|
31
|
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
|
32
|
Stine JG, Loomba R. Magnetic resonance imaging proton density fat fraction as an imaging-based biomarker of treatment response in patients with nonalcoholic steatohepatitis. Clin Liver Dis (Hoboken) 2022; 20:198-201. [PMID: 36523866 PMCID: PMC9745255 DOI: 10.1002/cld.1249] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/24/2022] [Accepted: 06/11/2022] [Indexed: 12/14/2022] Open
Abstract
Content available: Author Interview and Audio Recording.
Collapse
Affiliation(s)
- Jonathan G. Stine
- Division of Gastroenterology and HepatologyDepartment of MedicinePennsylvania State UniversityMilton S. Hershey Medical CenterHersheyPennsylvaniaUSA
- Department of Public Health SciencesPennsylvania State UniversityMilton S. Hershey Medical CenterHersheyPennsylvaniaUSA
- Liver CenterPennsylvania State UniversityMilton S. Hershey Medical CenterHersheyPennsylvaniaUSA
- Cancer InstitutePennsylvania State UniversityMilton S. Hershey Medical CenterHersheyPennsylvaniaUSA
| | - Rohit Loomba
- Division of Gastroenterology and HepatologyDepartment of MedicineUniversity of California San DiegoLa JollaCaliforniaUSA
- Division of EpidemiologyDepartment of Family Medicine and Public HealthUniversity of California San DiegoLa JollaCaliforniaUSA
- NAFLD Research CenterUniversity of California San DiegoLa JollaCaliforniaUSA
| |
Collapse
|
33
|
Xiang L, Li X, Luo Y, Zhou B, Liu Y, Li Y, Wu D, Jia L, Zhu PW, Zheng MH, Wang H, Lu Y. A multi-omic landscape of steatosis-to-NASH progression. LIFE METABOLISM 2022; 1:242-257. [PMID: 39872077 PMCID: PMC11749464 DOI: 10.1093/lifemeta/loac034] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/15/2022] [Revised: 09/20/2022] [Accepted: 11/16/2022] [Indexed: 01/29/2025]
Abstract
Nonalcoholic steatohepatitis (NASH) has emerged as a major cause of liver failure and hepatocellular carcinoma. Investigation into the molecular mechanisms that underlie steatosis-to-NASH progression is key to understanding the development of NASH pathophysiology. Here, we present comprehensive multi-omic profiles of preclinical animal models to identify genes, non-coding RNAs, proteins, and plasma metabolites involved in this progression. In particular, by transcriptomics analysis, we identified Growth Differentiation Factor 3 (GDF3) as a candidate noninvasive biomarker in NASH. Plasma GDF3 levels are associated with hepatic pathological features in patients with NASH, and differences in these levels provide a high diagnostic accuracy of NASH diagnosis (AUROC = 0.90; 95% confidence interval: 0.85-0.95) with a good sensitivity (90.7%) and specificity (86.4%). In addition, by developing integrated proteomic-metabolomic datasets and performing a subsequent pharmacological intervention in a mouse model of NASH, we show that ferroptosis may be a potential target to treat NASH. Moreover, by using competing endogenous RNAs network analysis, we found that several miRNAs, including miR-582-5p and miR-292a-3p, and lncRNAs, including XLOC-085738 and XLOC-041531, are associated with steatosis-to-NASH progression. Collectively, our data provide a valuable resource into the molecular characterization of NASH progression, leading to the novel insight that GDF3 may be a potential noninvasive diagnostic biomarker for NASH while further showing that ferroptosis is a therapeutic target for the disease.
Collapse
Affiliation(s)
- Liping Xiang
- Department of Endocrinology and Metabolism, Shanghai Clinical Center for Diabetes, Shanghai Diabetes Institute, Shanghai Key Laboratory of Diabetes Mellitus, Shanghai Sixth People’s Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Key Laboratory of Metabolism and Molecular Medicine of the Ministry of Education, Department of Endocrinology and Metabolism, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Xiaoyan Li
- Department of Oncology, the First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Yunchen Luo
- Department of Endocrinology and Metabolism, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Bing Zhou
- Department of Endocrinology and Metabolism, Shanghai Clinical Center for Diabetes, Shanghai Diabetes Institute, Shanghai Key Laboratory of Diabetes Mellitus, Shanghai Sixth People’s Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yuejun Liu
- Key Laboratory of Metabolism and Molecular Medicine of the Ministry of Education, Department of Endocrinology and Metabolism, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Yao Li
- Department of Laboratory Animal Science, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Duojiao Wu
- Zhongshan Hospital Institute of Clinical Science, Fudan University, Shanghai Institute of Clinical Bioinformatics, Shanghai, China
| | - Lijing Jia
- Department of Endocrinology, The Second Clinical Medical College, Jinan University (Shenzhen People’s Hospital), Shenzhen, China
| | - Pei-Wu Zhu
- Department of Laboratory Medicine, First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Ming-Hua Zheng
- MAFLD Research Center, Department of Hepatology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Hua Wang
- Department of Oncology, the First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Yan Lu
- Institute of Metabolism and Regenerative Medicine, Shanghai Sixth People’s Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
34
|
Role of Oxidative Stress and Lipid Peroxidation in the Pathophysiology of NAFLD. Antioxidants (Basel) 2022; 11:antiox11112217. [PMID: 36358589 PMCID: PMC9686676 DOI: 10.3390/antiox11112217] [Citation(s) in RCA: 36] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2022] [Revised: 11/02/2022] [Accepted: 11/08/2022] [Indexed: 11/11/2022] Open
Abstract
Non-alcoholic fatty liver disease (NAFLD) is characterised by an excess of hepatic fat that can progress to steatohepatitis, fibrosis, cirrhosis and hepatocarcinoma. The imbalance between lipid uptake/lipogenesis and lipid oxidation/secretion in the liver is a major feature of NAFLD. Given the lack of a non-invasive and reliable methods for the diagnosis of non-alcoholic steatohepatitis (NASH), it is important to find serum markers that are capable of discriminating or defining patients with this stage of NASH. Blood samples were obtained from 152 Caucasian subjects with biopsy-proven NAFLD due to persistently elevated liver enzyme levels. Metabolites representative of oxidative stress were assessed. The findings derived from this work revealed that NAFLD patients with a NASH score of ≥ 4 showed significantly higher levels of lipid peroxidation (LPO). Indeed, LPO levels above the optimal operating point (OOP) of 315.39 μM are an independent risk factor for presenting a NASH score of ≥ 4 (OR: 4.71; 95% CI: 1.68−13.19; p = 0.003). The area under the curve (AUC = 0.81, 95% CI = 0.73−0.89, p < 0.001) shows a good discrimination ability of the model. Therefore, understanding the molecular mechanisms underlying the basal inflammation present in these patients is postulated as a possible source of biomarkers and therapeutic targets in NASH.
Collapse
|
35
|
Cazac GD, Lăcătușu CM, Mihai C, Grigorescu ED, Onofriescu A, Mihai BM. Ultrasound-Based Hepatic Elastography in Non-Alcoholic Fatty Liver Disease: Focus on Patients with Type 2 Diabetes. Biomedicines 2022; 10:biomedicines10102375. [PMID: 36289643 PMCID: PMC9598125 DOI: 10.3390/biomedicines10102375] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2022] [Revised: 09/17/2022] [Accepted: 09/19/2022] [Indexed: 12/16/2022] Open
Abstract
Non-alcoholic fatty liver disease (NAFLD) is the most prevalent liver disease and is the hepatic expression of metabolic syndrome. The development of non-invasive methods for the diagnosis of hepatic steatosis and advanced fibrosis in high-risk patients, especially those with type 2 diabetes mellitus, is highly needed to replace the invasive method of liver biopsy. Elastographic methods can bring significant added value to screening and diagnostic procedures for NAFLD in patients with diabetes, thus contributing to improved NAFLD management. Pharmacological development and forthcoming therapeutic measures that address NAFLD should also be based on new, non-invasive, and reliable tools that assess NAFLD in at-risk patients and be able to properly guide treatment in individuals with both diabetes and NAFLD. This is the first review aiming to outline and discuss recent studies on ultrasound-based hepatic elastography, focusing on NAFLD assessment in patients with diabetes.
Collapse
Affiliation(s)
- Georgiana-Diana Cazac
- Unit of Diabetes, Nutrition and Metabolic Diseases, Faculty of Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania
- Clinical Center of Diabetes, Nutrition and Metabolic Diseases, “Sf. Spiridon” County Clinical Emergency Hospital, 700111 Iasi, Romania
| | - Cristina-Mihaela Lăcătușu
- Unit of Diabetes, Nutrition and Metabolic Diseases, Faculty of Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania
- Clinical Center of Diabetes, Nutrition and Metabolic Diseases, “Sf. Spiridon” County Clinical Emergency Hospital, 700111 Iasi, Romania
- Correspondence: (C.-M.L.); (E.-D.G.); Tel.: +40-72-321-1116 (C.-M.L.); +40-74-209-3749 (E.-D.G.)
| | - Cătălina Mihai
- Unit of Medical Semiology and Gastroenterology, Faculty of Medicine,, “Grigore T. Popa”, University of Medicine and Pharmacy, 700115 Iasi, Romania
- Institute of Gastroenterology and Hepatology, “Sf. Spiridon” Emergency Hospital, 700111 Iași, Romania
| | - Elena-Daniela Grigorescu
- Unit of Diabetes, Nutrition and Metabolic Diseases, Faculty of Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania
- Correspondence: (C.-M.L.); (E.-D.G.); Tel.: +40-72-321-1116 (C.-M.L.); +40-74-209-3749 (E.-D.G.)
| | - Alina Onofriescu
- Unit of Diabetes, Nutrition and Metabolic Diseases, Faculty of Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania
- Clinical Center of Diabetes, Nutrition and Metabolic Diseases, “Sf. Spiridon” County Clinical Emergency Hospital, 700111 Iasi, Romania
| | - Bogdan-Mircea Mihai
- Unit of Diabetes, Nutrition and Metabolic Diseases, Faculty of Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania
- Clinical Center of Diabetes, Nutrition and Metabolic Diseases, “Sf. Spiridon” County Clinical Emergency Hospital, 700111 Iasi, Romania
| |
Collapse
|
36
|
Ning Y, Zhou IY, Roberts JD, Rotile NJ, Akam E, Barrett SC, Sojoodi M, Barr MN, Punshon T, Pantazopoulos P, Drescher HK, Jackson BP, Tanabe KK, Caravan P. Molecular MRI quantification of extracellular aldehyde pairs for early detection of liver fibrogenesis and response to treatment. Sci Transl Med 2022; 14:eabq6297. [PMID: 36130015 PMCID: PMC10189657 DOI: 10.1126/scitranslmed.abq6297] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022]
Abstract
Liver fibrosis plays a critical role in the evolution of most chronic liver diseases and is characterized by a buildup of extracellular matrix, which can progress to cirrhosis, hepatocellular carcinoma, liver failure, or death. Now, there are no noninvasive methods available to accurately assess disease activity (fibrogenesis) to sensitively detect early onset of fibrosis or to detect early response to treatment. Here, we hypothesized that extracellular allysine aldehyde (LysAld) pairs formed by collagen oxidation during active fibrosis could be a target for assessing fibrogenesis with a molecular probe. We showed that molecular magnetic resonance imaging (MRI) using an extracellular probe targeting these LysAld pairs acts as a noninvasive biomarker of fibrogenesis and demonstrated its high sensitivity and specificity in detecting fibrogenesis in toxin- and dietary-induced mouse models, a cholestasis rat model of liver fibrogenesis, and in human fibrotic liver tissues. Quantitative molecular MRI was highly correlated with fibrogenesis markers and enabled noninvasive detection of early onset fibrosis and response to antifibrotic treatment, showing high potential for clinical translation.
Collapse
Affiliation(s)
- Yingying Ning
- Athinoula A. Martinos Center for Biomedical Imaging, Institute for Innovation in Imaging, Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02129, USA
| | - Iris. Y. Zhou
- Athinoula A. Martinos Center for Biomedical Imaging, Institute for Innovation in Imaging, Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02129, USA
| | - Jesse D. Roberts
- Cardiovascular Research Center of the General Medical Services, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02129, USA
| | - Nicholas J. Rotile
- Athinoula A. Martinos Center for Biomedical Imaging, Institute for Innovation in Imaging, Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02129, USA
| | - Eman Akam
- Athinoula A. Martinos Center for Biomedical Imaging, Institute for Innovation in Imaging, Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02129, USA
| | - Stephen C. Barrett
- Division of Gastrointestinal and Oncologic Surgery, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA
| | - Mozhdeh Sojoodi
- Division of Gastrointestinal and Oncologic Surgery, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA
| | - Matthew N. Barr
- Department of Earth Sciences, Dartmouth College, Hanover, NH 03766, USA
| | - Tracy Punshon
- Department of Earth Sciences, Dartmouth College, Hanover, NH 03766, USA
| | - Pamela Pantazopoulos
- Athinoula A. Martinos Center for Biomedical Imaging, Institute for Innovation in Imaging, Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02129, USA
| | - Hannah K. Drescher
- Gastrointestinal Unit, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA
| | - Brian P. Jackson
- Department of Earth Sciences, Dartmouth College, Hanover, NH 03766, USA
| | - Kenneth K. Tanabe
- Division of Gastrointestinal and Oncologic Surgery, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA
| | - Peter Caravan
- Athinoula A. Martinos Center for Biomedical Imaging, Institute for Innovation in Imaging, Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02129, USA
| |
Collapse
|
37
|
Romero-Gómez M. Non-alcoholic steatohepatitis. Med Clin (Barc) 2022; 159:388-395. [PMID: 36075749 DOI: 10.1016/j.medcli.2022.06.017] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2022] [Revised: 06/05/2022] [Accepted: 06/08/2022] [Indexed: 10/14/2022]
Abstract
Non-alcoholic steatohepatitis belongs to the spectrum of metabolic-associated fatty liver diseases characterized by steatosis linked to obesity, diabetes, metabolic syndrome, dyslipidemia and immune-mediated disorders. The main features of MAFLD include high prevalence, heterogeneity, complexity and dynamic disease. Pruritus and asthenia are the main clinical manifestation that impact on quality of life and patient-reported outcomes. Biochemical or imagen-based non-invasive test have been implemented in the diagnostic process. Liver biopsy remains as the gold standard. Therapeutic options included life-style intervention. Mediterranean hypocaloric Diet to lose weight, exercise to fight sarcopenia and alcohol abstinence. In non-responders, drug-therapy focusing on obesity, diabetes and fibrosis using sequentially or combined to promote steatosis, inflammation and fibrosis regression.
Collapse
Affiliation(s)
- Manuel Romero-Gómez
- Servicio de Aparato Digestivo, Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERHD), Departamento de Medicina, Hospital Universitario Virgen del Rocío, Instituto de Biomedicina de Sevilla (HUVR/CSIC/US), Universidad de Sevilla, Sevilla, España.
| |
Collapse
|
38
|
Abstract
Initially a condition that received limited recognition and whose clinical impact was controversial, non-alcoholic steatohepatitis (NASH) has become a leading cause of chronic liver disease. Although there are no approved therapies, major breakthroughs, which will be reviewed here, have paved the way for future therapeutic successes. The unmet medical need in NASH is no longer disputed, and progress in the understanding of its pathogenesis has resulted in the identification of many pharmacological targets. Key surrogate outcomes for therapeutic trials are now accepted by regulatory agencies, thus creating a path for drug registration. A set of non-invasive measurements enabled early-stage trials to be conducted expeditiously, thus providing early indications on the biological and possibly clinical actions of therapeutic candidates. This generated efficacy results for a number of highly promising compounds that are now in late-stage development. Intense research aimed at further improving the assessment of histological endpoints and in developing non-invasive predictive biomarkers is underway. This will help improve the design and feasibility of successful trials, ultimately providing patients with therapeutic options that can change the course of the disease.
Collapse
|
39
|
Wang L, Dong J, Xu M, Li L, Yang N, Qian G. Association Between Monocyte to High-Density Lipoprotein Cholesterol Ratio and Risk of Non-alcoholic Fatty Liver Disease: A Cross-Sectional Study. Front Med (Lausanne) 2022; 9:898931. [PMID: 35665350 PMCID: PMC9161020 DOI: 10.3389/fmed.2022.898931] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2022] [Accepted: 04/26/2022] [Indexed: 12/12/2022] Open
Abstract
Background Non-alcoholic fatty liver disease (NAFLD) is a global health problem affecting more than a quarter of the entire adult population. Both monocytes and high-density lipoprotein cholesterol (HDL-C) were found to participate in the progression of hepatic inflammation and oxidative stress. We speculated that the monocyte-to-HDL-C ratio (MHR) may be associated with the risk of NAFLD. Methods We conducted a cross-sectional study using data from the National Health and Nutrition Examination Survey (NHANES) 2017-2018. NAFLD was identified using a controlled attenuation parameter (CAP) of ≥274 dB/m. Degree of liver fibrosis were assessed by liver stiffness measurement (LSM) and LSM values≥8.0, ≥ 9.7, and ≥13.7 kPa were defined as significant fibrosis (≥F2), advanced fibrosis (≥F3) and cirrhosis (F4), respectively. The association between MHR and the risk of NAFLD and liver fibrosis was estimated using weighted multivariable logistic regression. The non-linear relationship between MHR and the risk of NAFLD was further described using smooth curve fittings and threshold effect analysis. Results Of 4,319 participants, a total of 1,703 (39.4%) participants were diagnosed with NAFLD. After complete adjustment for potential confounders, MHR was positively associated with the risk of NAFLD (OR = 2.87, 95% CI: 1.95-4.22). The risk of NAFLD increased progressively as the MHR quarter increased (P for trend < 0.001). In subgroup analysis stratified by sex, a positive association existed in both sexes; Women displayed higher risk (men: OR = 2.12, 95% CI: 1.33-3.39; women: OR = 2.64, 95%CI: 1.40-4.97). MHR was positively associated with the risk of significant liver fibrosis (OR = 1.60, 95% CI: 1.08-2.37) and cirrhosis (OR = 1.83, 95% CI: 1.08-3.13), but not with advanced liver fibrosis (OR = 1.53, 95% CI: 0.98-2.39) after full adjustment for potential confounders. In the subgroup analysis by sex, the association between MHR and different degrees of liver fibrosis was significantly positive in women. When analyzing the relationship between MHR and NAFLD risk, a reverse U-shaped curve with an inflection point of 0.36 for MHR was found in women. Conclusion Higher MHR was associated with increased odds of NAFLD among Americans of both sexes. However, an association between MHR and liver fibrosis was found mainly among women.
Collapse
Affiliation(s)
- Liping Wang
- Department of Infectious Diseases, Ningbo First Hospital, Ningbo University, Ningbo, China
- Department of Hepatology, Non-alcoholic Fatty Liver Disease (NAFLD) Research Center, Ningbo Hospital of Zhejiang University, Ningbo, China
| | - Jinzhong Dong
- Department of Intensive Care Medicine, Ningbo First Hospital, Ningbo University, Ningbo, China
| | - Miao Xu
- Department of Endocrinology and Metabolism, Ningbo First Hospital, Ningbo University, Zhejiang, China
| | - Li Li
- Department of Endocrinology and Metabolism, Ningbo First Hospital, Ningbo University, Zhejiang, China
| | - Naibin Yang
- Department of Infectious Diseases, Ningbo First Hospital, Ningbo University, Ningbo, China
- Department of Hepatology, Non-alcoholic Fatty Liver Disease (NAFLD) Research Center, Ningbo Hospital of Zhejiang University, Ningbo, China
| | - Guoqing Qian
- Department of Infectious Diseases, Ningbo First Hospital, Ningbo University, Ningbo, China
- Department of Hepatology, Non-alcoholic Fatty Liver Disease (NAFLD) Research Center, Ningbo Hospital of Zhejiang University, Ningbo, China
| |
Collapse
|
40
|
Spiers J, Brindley JH, Li W, Alazawi W. What's new in non-alcoholic fatty liver disease? Frontline Gastroenterol 2022; 13:e102-e108. [PMID: 35812024 PMCID: PMC9234732 DOI: 10.1136/flgastro-2022-102122] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/14/2022] [Accepted: 05/20/2022] [Indexed: 02/04/2023] Open
Abstract
Non-alcoholic fatty liver disease (NAFLD) is the most common cause of liver disease worldwide, with an estimated prevalence of 25% in the Western World. NAFLD is a broad spectrum of disease states and while most people with NAFLD do not have progressive disease, 10-20% of patients develop histological features of inflammation (non-alcoholic steatohepatitis), fibrosis, cirrhosis and its complications. Despite this large disease burden of significant clinical impact, most people living with NAFLD are undiagnosed, disease course prediction is imprecise and there are no treatments licensed for this condition. In this review, we discuss some of the recent developments in NAFLD, focusing on disease definition and diagnosis, risk stratification and treatments.
Collapse
Affiliation(s)
- Jessica Spiers
- Barts Liver Centre, Barts and The London School of Medicine and Dentistry, Blizard Institute, London, UK
| | - James Hallimond Brindley
- Barts Liver Centre, Barts and The London School of Medicine and Dentistry, Blizard Institute, London, UK
| | - Wenhao Li
- Barts Liver Centre, Barts and The London School of Medicine and Dentistry, Blizard Institute, London, UK
| | - William Alazawi
- Barts Liver Centre, Barts and The London School of Medicine and Dentistry, Blizard Institute, London, UK
| |
Collapse
|