51
|
Iwaki M, Yoneda M, Wada N, Otani T, Kobayashi T, Nogami A, Saito S, Nakajima A. Emerging drugs for the treatment of hepatic fibrosis on nonalcoholic steatohepatitis. Expert Opin Emerg Drugs 2024; 29:127-137. [PMID: 38469871 DOI: 10.1080/14728214.2024.2328036] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2023] [Accepted: 03/05/2024] [Indexed: 03/13/2024]
Abstract
INTRODUCTION Approved drug therapies for nonalcoholic steatohepatitis (NASH) are lacking, for which various agents are currently being tested in clinical trials. Effective drugs for liver fibrosis, the factor most associated with prognosis in NASH, are important. AREAS COVERED This study reviewed the treatment of NASH with a focus on the effects of existing drugs and new drugs on liver fibrosis. EXPERT OPINION Considering the complex pathophysiology of fibrosis in NASH, drug therapy may target multiple pathways. The method of assessing fibrosis is important when considering treatment for liver fibrosis in NASH. The Food and Drug Administration considers an important fibrosis endpoint to be histological improvement in at least one fibrosis stage while preventing worsening of fatty hepatitis. To obtain approval as a drug for NASH, efficacy needs to be demonstrated on endpoints such as liver-related events and myocardial infarction. Among the current therapeutic agents for NASH, thiazolidinedione, sodium-glucose co-transporter 2, and selective peroxisome proliferator-activated receptors α modulator have been reported to be effective against fibrosis, although further evidence is required. The effects of pan-peroxisome proliferator-activated receptors, obeticholic acid, and fibroblast growth factor-21 analogs on liver fibrosis in the development stage therapeutics for NASH are of particular interest.
Collapse
Affiliation(s)
- Michihiro Iwaki
- Department of Gastroenterology and Hepatology, Yokohama City University Graduate School of Medicine, Yokohama, Japan
| | - Masato Yoneda
- Department of Gastroenterology and Hepatology, Yokohama City University Graduate School of Medicine, Yokohama, Japan
| | - Naohiro Wada
- Department of Gastroenterology and Hepatology, Yokohama City University Graduate School of Medicine, Yokohama, Japan
| | - Tomohiro Otani
- Department of Gastroenterology and Hepatology, Yokohama City University Graduate School of Medicine, Yokohama, Japan
| | - Takashi Kobayashi
- Department of Gastroenterology and Hepatology, Yokohama City University Graduate School of Medicine, Yokohama, Japan
| | - Asako Nogami
- Department of Gastroenterology and Hepatology, Yokohama City University Graduate School of Medicine, Yokohama, Japan
| | - Satoru Saito
- Department of Gastroenterology, Sanno Hospital, Minato-Ku, Tokyo, Japan
| | - Atsushi Nakajima
- Department of Gastroenterology and Hepatology, Yokohama City University Graduate School of Medicine, Yokohama, Japan
| |
Collapse
|
52
|
Tincopa MA, Anstee QM, Loomba R. New and emerging treatments for metabolic dysfunction-associated steatohepatitis. Cell Metab 2024; 36:912-926. [PMID: 38608696 DOI: 10.1016/j.cmet.2024.03.011] [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: 12/01/2023] [Revised: 02/01/2024] [Accepted: 03/18/2024] [Indexed: 04/14/2024]
Abstract
Metabolic dysfunction-associated steatohepatitis (MASH) is a leading etiology of chronic liver disease worldwide, with increasing incidence and prevalence in the setting of the obesity epidemic. MASH is also a leading indication for liver transplantation, given its associated risk of progression to end-stage liver disease. A key challenge in managing MASH is the lack of approved pharmacotherapy. In its absence, lifestyle interventions with a focus on healthy nutrition and regular physical activity have been the cornerstone of therapy. Real-world efficacy and sustainability of lifestyle interventions are low, however. Pharmacotherapy development for MASH is emerging with promising data from several agents with different mechanisms of action (MOAs) in phase 3 clinical trials. In this review, we highlight ongoing challenges and potential solutions in drug development for MASH and provide an overview of available data from emerging therapies across multiple MOAs.
Collapse
Affiliation(s)
- Monica A Tincopa
- MASLD Research Center, Division of Gastroenterology and Hepatology, University of California, San Diego, La Jolla, CA 92103, USA
| | - Quentin M Anstee
- Translational & Clinical Research Institute, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne, UK; Newcastle NIHR Biomedical Research Center, Newcastle upon Tyne Hospitals NHS Trust, Newcastle upon Tyne, UK
| | - Rohit Loomba
- MASLD Research Center, Division of Gastroenterology and Hepatology, University of California, San Diego, La Jolla, CA 92103, USA; School of Public Health, University of California, San Diego, La Jolla, CA 92103, USA.
| |
Collapse
|
53
|
Brouwers B, Rao G, Tang Y, Rodríguez Á, Glass LC, Hartman ML. Incretin-based investigational therapies for the treatment of MASLD/MASH. Diabetes Res Clin Pract 2024; 211:111675. [PMID: 38636848 DOI: 10.1016/j.diabres.2024.111675] [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: 08/30/2023] [Revised: 04/15/2024] [Accepted: 04/15/2024] [Indexed: 04/20/2024]
Abstract
Metabolic dysfunction-associated steatotic liver disease (MASLD), formerly known as non-alcoholic fatty liver disease (NAFLD), is the most common form of chronic liver disease. It exists as either simple steatosis or its more progressive form, metabolic dysfunction-associated steatohepatitis (MASH), formerly, non-alcoholic steatohepatitis (NASH). The global prevalence of MASLD is estimated to be 32% among adults and is projected to continue to rise with increasing rates of obesity, type 2 diabetes, and metabolic syndrome. While simple steatosis is often considered benign and reversible, MASH is progressive, potentially leading to the development of cirrhosis, liver failure, and hepatocellular carcinoma. Treatment of MASH is therefore directed at slowing, stopping, or reversing the progression of disease. Evidence points to improved liver histology with therapies that result in sustained body weight reduction. Incretin-based molecules, such as glucagon-like peptide-1 receptor agonists (GLP-1 RAs), alone or in combination with glucose-dependent insulinotropic polypeptide (GIP) and/or glucagon receptor agonists, have shown benefit here, and several are under investigation for MASLD/MASH treatment. In this review, we discuss current published data on GLP-1, GIP/GLP-1, GLP-1/glucagon, and GLP-1/GIP/glucagon RAs in MASLD/MASH, focusing on their efficacy on liver histology, liver fat, and MASH biomarkers.
Collapse
Affiliation(s)
| | - Girish Rao
- Eli Lilly and Company, Indianapolis, IN, USA
| | | | | | | | | |
Collapse
|
54
|
Fischer AM, Lechea N, Coxson HO. This Is What Metabolic Dysfunction-Associated Steatotic Liver Disease Looks Like: Potential of a Multiparametric MRI Protocol. Semin Liver Dis 2024; 44:226-238. [PMID: 38806158 DOI: 10.1055/a-2334-8525] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 05/30/2024]
Abstract
Metabolic dysfunction-associated steatotic liver disease (MASLD) is a prevalent condition with a broad spectrum defined by liver biopsy. This gold standard method evaluates three features: steatosis, activity (ballooning and lobular inflammation), and fibrosis, attributing them to certain grades or stages using a semiquantitative scoring system. However, liver biopsy is subject to numerous restrictions, creating an unmet need for a reliable and reproducible method for MASLD assessment, grading, and staging. Noninvasive imaging modalities, such as magnetic resonance imaging (MRI), offer the potential to assess quantitative liver parameters. This review aims to provide an overview of the available MRI techniques for the three criteria evaluated individually by liver histology. Here, we discuss the possibility of combining multiple MRI parameters to replace liver biopsy with a holistic, multiparametric MRI protocol. In conclusion, the development and implementation of such an approach could significantly improve the diagnosis and management of MASLD, reducing the need for invasive procedures and paving the way for more personalized treatment strategies.
Collapse
Affiliation(s)
- Anja M Fischer
- Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach, Germany
| | - Nazim Lechea
- Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach, Germany
| | - Harvey O Coxson
- Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach, Germany
| |
Collapse
|
55
|
Samanta A, Sen Sarma M. Metabolic dysfunction-associated steatotic liver disease: A silent pandemic. World J Hepatol 2024; 16:511-516. [PMID: 38689742 PMCID: PMC11056897 DOI: 10.4254/wjh.v16.i4.511] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/23/2024] [Revised: 03/05/2024] [Accepted: 04/07/2024] [Indexed: 04/24/2024] Open
Abstract
The worldwide epidemiology of non-alcoholic fatty liver disease (NAFLD) is showing an upward trend, parallel to the rising trend of metabolic syndrome, owing to lifestyle changes. The pathogenesis of NAFLD has not been fully understood yet. Therefore, NAFLD has emerged as a public health concern in the field of hepatology and metabolisms worldwide. Recent changes in the nomenclature from NAFLD to metabolic dysfunction-associated steatotic liver disease have brought a positive outlook changes in the understanding of the disease process and doctor-patient communication. Lifestyle changes are the main treatment modality. Recently, clinical trial using drugs that target 'insulin resistance' which is the driving force behind NAFLD, have shown promising results. Further translational research is needed to better understand the underlying pathophysiological mechanism of NAFLD which may open newer avenues of therapeutic targets. The role of gut dysbiosis in etiopathogenesis and use of fecal microbiota modification in the treatment should be studied extensively. Prevention of this silent epidemic by spreading awareness and early intervention should be our priority.
Collapse
Affiliation(s)
- Arghya Samanta
- Department of Pediatric Gastroenterology, Sanjay Gandhi Postgraduate Institute of Medical Sciences, Lucknow 226014, India
| | - Moinak Sen Sarma
- Department of Pediatric Gastroenterology, Sanjay Gandhi Postgraduate Institute of Medical Sciences, Lucknow 226014, India.
| |
Collapse
|
56
|
Yoshida K, Poon V, Dash A, Kunder R, Chinn L, Kågedal M. Simulation-based evaluation of personalized dosing approaches for anti-FGFR/KLB bispecific antibody fazpilodemab. CPT Pharmacometrics Syst Pharmacol 2024; 13:544-550. [PMID: 38343040 PMCID: PMC11015072 DOI: 10.1002/psp4.13111] [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: 11/15/2023] [Revised: 12/19/2023] [Accepted: 01/22/2024] [Indexed: 04/14/2024] Open
Abstract
Personalized dosing approaches play important roles in clinical practices to improve benefit: risk profiles. Whereas this is also important for drug development, especially in the context of drugs with narrow therapeutic windows, such approaches have not been fully evaluated during clinical development. Fazpilodemab (BFKB8488A) is an agonistic bispecific antibody which was being developed for the treatment of nonalcoholic steatohepatitis. The objective of this study was to characterize the exposure-response relationships of fazpilodemab with the purpose of guiding dose selection for a phase II study, as well as to evaluate various personalized dosing strategies to optimize the treatment benefit. Fazpilodemab exhibited clear exposure-response relationships for a pharmacodynamic (PD) biomarker and gastrointestinal adverse events (GIAEs), such as nausea and vomiting. Static exposure-response analysis, as well as longitudinal adverse event (AE) analysis using discrete-time Markov model, were performed to characterize the observations. Clinical trial simulations were performed based on the developed exposure-response models to evaluate probability of achieving target PD response and the frequency of GIAEs to inform phase II dose selection. Dynamic simulation of personalized dosing strategies demonstrated that the AE-based personalized dosing is the most effective approach for optimizing the benefit-risk profiles. The approach presented here can be a useful framework for quantifying the benefit of personalized dosing for drugs with narrow therapeutic windows.
Collapse
Affiliation(s)
- Kenta Yoshida
- Clinical PharmacologyGenentech Inc.South San FranciscoCaliforniaUSA
| | - Victor Poon
- Clinical PharmacologyGenentech Inc.South San FranciscoCaliforniaUSA
| | - Ajit Dash
- Early Development SafetyGenentech Inc.South San FranciscoCaliforniaUSA
| | - Rebecca Kunder
- Early Clinical DevelopmentGenentech Inc.South San FranciscoCaliforniaUSA
| | - Leslie Chinn
- Clinical PharmacologyGenentech Inc.South San FranciscoCaliforniaUSA
| | | |
Collapse
|
57
|
Simon TG, Wilechansky RM, Stoyanova S, Grossman A, Dichtel LE, Lauer GM, Miller KK, Hoshida Y, Corey KE, Loomba R, Chung RT, Chan AT. Aspirin for Metabolic Dysfunction-Associated Steatotic Liver Disease Without Cirrhosis: A Randomized Clinical Trial. JAMA 2024; 331:920-929. [PMID: 38502074 PMCID: PMC10951738 DOI: 10.1001/jama.2024.1215] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/09/2023] [Accepted: 01/26/2024] [Indexed: 03/20/2024]
Abstract
Importance Aspirin may reduce severity of metabolic dysfunction-associated steatotic liver disease (MASLD) and lower the incidence of end-stage liver disease and hepatocellular carcinoma, in patients with MASLD. However, the effect of aspirin on MASLD is unknown. Objective To test whether low-dose aspirin reduces liver fat content, compared with placebo, in adults with MASLD. Design, Setting, and Participants This 6-month, phase 2, randomized, double-blind, placebo-controlled clinical trial was conducted at a single hospital in Boston, Massachusetts. Participants were aged 18 to 70 years with established MASLD without cirrhosis. Enrollment occurred between August 20, 2019, and July 19, 2022, with final follow-up on February 23, 2023. Interventions Participants were randomized (1:1) to receive either once-daily aspirin, 81 mg (n = 40) or identical placebo pills (n = 40) for 6 months. Main Outcomes and Measures The primary end point was mean absolute change in hepatic fat content, measured by proton magnetic resonance spectroscopy (MRS) at 6-month follow-up. The 4 key secondary outcomes included mean percentage change in hepatic fat content by MRS, the proportion achieving at least 30% reduction in hepatic fat, and the mean absolute and relative reductions in hepatic fat content, measured by magnetic resonance imaging proton density fat fraction (MRI-PDFF). Analyses adjusted for the baseline value of the corresponding outcome. Minimal clinically important differences for study outcomes were not prespecified. Results Among 80 randomized participants (mean age, 48 years; 44 [55%] women; mean hepatic fat content, 35% [indicating moderate steatosis]), 71 (89%) completed 6-month follow-up. The mean absolute change in hepatic fat content by MRS was -6.6% with aspirin vs 3.6% with placebo (difference, -10.2% [95% CI, -27.7% to -2.6%]; P = .009). Compared with placebo, aspirin treatment significantly reduced relative hepatic fat content (-8.8 vs 30.0 percentage points; mean difference, -38.8 percentage points [95% CI, -66.7 to -10.8]; P = .007), increased the proportion of patients with 30% or greater relative reduction in hepatic fat (42.5% vs 12.5%; mean difference, 30.0% [95% CI, 11.6% to 48.4%]; P = .006), reduced absolute hepatic fat content by MRI-PDFF (-2.7% vs 0.9%; mean difference, -3.7% [95% CI, -6.1% to -1.2%]; P = .004]), and reduced relative hepatic fat content by MRI-PDFF (-11.7 vs 15.7 percentage points; mean difference, -27.3 percentage points [95% CI, -45.2 to -9.4]; P = .003). Thirteen participants (32.5%) in each group experienced an adverse event, most commonly upper respiratory tract infections (10.0% in each group) or arthralgias (5.0% for aspirin vs 7.5% for placebo). One participant randomized to aspirin (2.5%) experienced drug-related heartburn. Conclusions and Relevance In this preliminary randomized clinical trial of patients with MASLD, 6 months of daily low-dose aspirin significantly reduced hepatic fat quantity compared with placebo. Further study in a larger sample size is necessary to confirm these findings. Trial Registration ClinicalTrials.gov Identifier: NCT04031729.
Collapse
Affiliation(s)
- Tracey G. Simon
- Clinical and Translational Epidemiology Unit (CTEU), Massachusetts General Hospital, Boston
- Harvard Medical School, Boston, Massachusetts
- Division of Pharmacoepidemiology and Pharmacoeconomics, Department of Medicine, Brigham and Women’s Hospital, Boston, Massachusetts
| | - Robert M. Wilechansky
- Clinical and Translational Epidemiology Unit (CTEU), Massachusetts General Hospital, Boston
- Harvard Medical School, Boston, Massachusetts
- Division of Gastroenterology and Hepatology, Department of Medicine, Massachusetts General Hospital, Boston
| | - Stefania Stoyanova
- Division of Gastroenterology and Hepatology, Department of Medicine, Massachusetts General Hospital, Boston
| | - Alessandra Grossman
- Division of Gastroenterology and Hepatology, Department of Medicine, Massachusetts General Hospital, Boston
| | - Laura E. Dichtel
- Harvard Medical School, Boston, Massachusetts
- Neuroendocrine Unit, Department of Medicine, Massachusetts General Hospital, Boston
| | - Georg M. Lauer
- Harvard Medical School, Boston, Massachusetts
- Division of Gastroenterology and Hepatology, Department of Medicine, Massachusetts General Hospital, Boston
| | - Karen K. Miller
- Harvard Medical School, Boston, Massachusetts
- Neuroendocrine Unit, Department of Medicine, Massachusetts General Hospital, Boston
| | - Yujin Hoshida
- Division of Digestive and Liver Diseases, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas
| | - Kathleen E. Corey
- Clinical and Translational Epidemiology Unit (CTEU), Massachusetts General Hospital, Boston
- Harvard Medical School, Boston, Massachusetts
- Division of Gastroenterology and Hepatology, Department of Medicine, Massachusetts General Hospital, Boston
| | - Rohit Loomba
- NAFLD Research Center, Division of Gastroenterology and Hepatology, Department of Medicine, University of California San Diego, La Jolla
| | - Raymond T. Chung
- Clinical and Translational Epidemiology Unit (CTEU), Massachusetts General Hospital, Boston
- Harvard Medical School, Boston, Massachusetts
- Division of Gastroenterology and Hepatology, Department of Medicine, Massachusetts General Hospital, Boston
| | - Andrew T. Chan
- Clinical and Translational Epidemiology Unit (CTEU), Massachusetts General Hospital, Boston
- Harvard Medical School, Boston, Massachusetts
- Division of Gastroenterology and Hepatology, Department of Medicine, Massachusetts General Hospital, Boston
| |
Collapse
|
58
|
Björnsdottir S, Ulfsdottir H, Gudmundsson EF, Sveinsdottir K, Isberg AP, Dobies B, Akerlie Magnusdottir GE, Gunnarsdottir T, Karlsdottir T, Bjornsdottir G, Sigurdsson S, Oddsson S, Gudnason V. User Engagement, Acceptability, and Clinical Markers in a Digital Health Program for Nonalcoholic Fatty Liver Disease: Prospective, Single-Arm Feasibility Study. JMIR Cardio 2024; 8:e52576. [PMID: 38152892 PMCID: PMC10905363 DOI: 10.2196/52576] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2023] [Revised: 12/15/2023] [Accepted: 12/26/2023] [Indexed: 12/29/2023] Open
Abstract
BACKGROUND Nonalcoholic fatty liver disease (NAFLD) has become the most common chronic liver disease in the world. Common comorbidities are central obesity, type 2 diabetes mellitus, dyslipidemia, and metabolic syndrome. Cardiovascular disease is the most common cause of death among people with NAFLD, and lifestyle changes can improve health outcomes. OBJECTIVE This study aims to explore the acceptability of a digital health program in terms of engagement, retention, and user satisfaction in addition to exploring changes in clinical outcomes, such as weight, cardiometabolic risk factors, and health-related quality of life. METHODS We conducted a prospective, open-label, single-arm, 12-week study including 38 individuals with either a BMI >30, metabolic syndrome, or type 2 diabetes mellitus and NAFLD screened by FibroScan. An NAFLD-specific digital health program focused on disease education, lowering carbohydrates in the diet, food logging, increasing activity level, reducing stress, and healthy lifestyle coaching was offered to participants. The coach provided weekly feedback on food logs and other in-app activities and opportunities for participants to ask questions. The coaching was active throughout the 12-week intervention period. The primary outcome was feasibility and acceptability of the 12-week program, assessed through patient engagement, retention, and satisfaction with the program. Secondary outcomes included changes in weight, liver fat, body composition, and other cardiometabolic clinical parameters at baseline and 12 weeks. RESULTS In total, 38 individuals were included in the study (median age 59.5, IQR 46.3-68.8 years; n=23, 61% female). Overall, 34 (89%) participants completed the program and 29 (76%) were active during the 12-week program period. The median satisfaction score was 6.3 (IQR 5.8-6.7) of 7. Mean weight loss was 3.5 (SD 3.7) kg (P<.001) or 3.2% (SD 3.4%), with a 2.2 (SD 2.7) kg reduction in fat mass (P<.001). Relative liver fat reduction was 19.4% (SD 23.9%). Systolic blood pressure was reduced by 6.0 (SD 13.5) mmHg (P=.009). The median reduction was 0.14 (IQR 0-0.47) mmol/L for triglyceride levels (P=.003), 3.2 (IQR 0.0-5.4) µU/ml for serum insulin (s-insulin) levels (P=.003), and 0.5 (IQR -0.7 to 3.8) mmol/mol for hemoglobin A1c (HbA1c) levels (P=.03). Participants who were highly engaged (ie, who used the app at least 5 days per week) had greater weight loss and liver fat reduction. CONCLUSIONS The 12-week-long digital health program was feasible for individuals with NAFLD, receiving high user engagement, retention, and satisfaction. Improved liver-specific and cardiometabolic health was observed, and more engaged participants showed greater improvements. This digital health program could provide a new tool to improve health outcomes in people with NAFLD. TRIAL REGISTRATION Clinicaltrials.gov NCT05426382; https://clinicaltrials.gov/study/NCT05426382.
Collapse
Affiliation(s)
- Sigridur Björnsdottir
- Department of Endocrinology, Metabolism and Diabetes, Karolinska Institutet, Stockholm, Sweden
| | | | | | | | | | | | | | | | | | - Gudlaug Bjornsdottir
- Icelandic Heart Association, Kopavogur, Iceland
- School of Health Sciences, Faculty of Medicine, University of Iceland, Reykjavik, Iceland
| | - Sigurdur Sigurdsson
- Icelandic Heart Association, Kopavogur, Iceland
- School of Health Sciences, Faculty of Medicine, University of Iceland, Reykjavik, Iceland
| | | | - Vilmundur Gudnason
- Icelandic Heart Association, Kopavogur, Iceland
- School of Health Sciences, Faculty of Medicine, University of Iceland, Reykjavik, Iceland
| |
Collapse
|
59
|
Venkatesh SK, Idilman IS, Li J, Yin M. Shrinking fat, healing liver: unlocking the metabolic dysfunction associated steatohepatitis puzzle. Hepatobiliary Surg Nutr 2024; 13:132-135. [PMID: 38322227 PMCID: PMC10839737 DOI: 10.21037/hbsn-23-569] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/02/2023] [Accepted: 12/06/2023] [Indexed: 02/08/2024]
Affiliation(s)
| | | | - Jiahui Li
- Department of Radiology, Mayo Clinic, Rochester, MN, USA
| | - Meng Yin
- Department of Radiology, Mayo Clinic, Rochester, MN, USA
| |
Collapse
|
60
|
Harris SJ, Smith N, Hummer B, Schreibman IR, Faust AJ, Geyer NR, Chinchilli VM, Sciamanna C, Loomba R, Stine JG. Exercise training improves serum biomarkers of liver fibroinflammation in patients with metabolic dysfunction-associated steatohepatitis. Liver Int 2024; 44:532-540. [PMID: 38014619 PMCID: PMC10844956 DOI: 10.1111/liv.15769] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/05/2023] [Revised: 10/04/2023] [Accepted: 10/08/2023] [Indexed: 11/29/2023]
Abstract
BACKGROUND AND AIMS Exercise training is recommended for all patients with metabolic dysfunction-associated steatotic liver disease and may reverse liver fibrosis. Whether exercise training improves liver fibrosis without body weight loss remains controversial. We further investigated this relationship using serum biomarkers of liver fibroinflammation in a post hoc analysis of an exercise trial where patients did not lose significant body weight. METHODS In the NASHFit trial, patients with metabolic dysfunction-associated steatohepatitis were randomized to receive either moderate-intensity aerobic exercise training or standard clinical care for 20 weeks. Mediterranean-informed dietary counselling was provided to each group. Change in serum biomarkers was measured and compared between the two groups. RESULTS Exercise training led to improvement in serum biomarkers of liver fibroinflammation, including (1) ≥17 IU/L reduction in alanine aminotransferase (ALT) in 53% of individuals in the exercise training group compared to 13% in the standard clinical care group (p < 0.001; mean reduction 24% vs. 10% respectively) and (2) improvement in CK18 (-61 vs. +71 ng/mL, p = 0.040). ALT improvement ≥17 IU/L was correlated with ≥30% relative reduction in magnetic resonance imaging-measured liver fat and PNPLA3 genotype. CONCLUSION Exercise training improves multiple serum biomarkers of liver fibroinflammation at clinically significant thresholds of response without body weight loss. This study provides further evidence that exercise training should be viewed as a weight-neutral intervention for which response to intervention can be readily monitored with widely available non-invasive biomarkers that can be applied at the population level.
Collapse
Affiliation(s)
- Sara J. Harris
- College of Medicine, The Pennsylvania State University,
Hershey PA
| | - Nataliya Smith
- Division of Gastroenterology and Hepatology, Department of
Medicine, Penn State Health- Milton S. Hershey Medical Center, Hershey PA
- Fatty Liver Program, Penn State Health- Milton S. Hershey
Medical Center, Hershey PA
| | - Breianna Hummer
- Division of Gastroenterology and Hepatology, Department of
Medicine, Penn State Health- Milton S. Hershey Medical Center, Hershey PA
- Fatty Liver Program, Penn State Health- Milton S. Hershey
Medical Center, Hershey PA
| | - Ian R. Schreibman
- Division of Gastroenterology and Hepatology, Department of
Medicine, Penn State Health- Milton S. Hershey Medical Center, Hershey PA
- Fatty Liver Program, Penn State Health- Milton S. Hershey
Medical Center, Hershey PA
- Liver Center, Penn State Health- Milton S. Hershey Medical
Center, Hershey PA
| | - Alison J. Faust
- Division of Gastroenterology and Hepatology, Department of
Medicine, Penn State Health- Milton S. Hershey Medical Center, Hershey PA
- Fatty Liver Program, Penn State Health- Milton S. Hershey
Medical Center, Hershey PA
- Liver Center, Penn State Health- Milton S. Hershey Medical
Center, Hershey PA
| | - Nathaniel R. Geyer
- Department of Public Health Sciences, The Pennsylvania
State University- College of Medicine, Hershey PA
| | - Vernon M. Chinchilli
- Department of Public Health Sciences, The Pennsylvania
State University- College of Medicine, Hershey PA
| | - Chris Sciamanna
- College of Medicine, The Pennsylvania State University,
Hershey PA
| | - Rohit Loomba
- Division of Gastroenterology and Hepatology, Department of
Medicine, University of California San Diego, San Diego CA
- NAFLD Research Center, University of California San Diego,
San Diego CA
| | - Jonathan G. Stine
- Division of Gastroenterology and Hepatology, Department of
Medicine, Penn State Health- Milton S. Hershey Medical Center, Hershey PA
- Fatty Liver Program, Penn State Health- Milton S. Hershey
Medical Center, Hershey PA
- Liver Center, Penn State Health- Milton S. Hershey Medical
Center, Hershey PA
- Department of Public Health Sciences, The Pennsylvania
State University- College of Medicine, Hershey PA
- Cancer Institute, Penn State Health- Milton S. Hershey
Medical Center, Hershey PA
| |
Collapse
|
61
|
Esler WP, Cohen DE. Pharmacologic inhibition of lipogenesis for the treatment of NAFLD. J Hepatol 2024; 80:362-377. [PMID: 37977245 PMCID: PMC10842769 DOI: 10.1016/j.jhep.2023.10.042] [Citation(s) in RCA: 39] [Impact Index Per Article: 39.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/30/2023] [Revised: 10/13/2023] [Accepted: 10/23/2023] [Indexed: 11/19/2023]
Abstract
The hepatic accumulation of excess triglycerides is a seminal event in the initiation and progression of non-alcoholic fatty liver disease (NAFLD). Hepatic steatosis occurs when the hepatic accrual of fatty acids from the plasma and de novo lipogenesis (DNL) is no longer balanced by rates of fatty acid oxidation and secretion of very low-density lipoprotein-triglycerides. Accumulating data indicate that increased rates of DNL are central to the development of hepatic steatosis in NAFLD. Whereas the main drivers in NAFLD are transcriptional, owing to both hyperinsulinemia and hyperglycaemia, the effectors of DNL are a series of well-characterised enzymes. Several have proven amenable to pharmacologic inhibition or oligonucleotide-mediated knockdown, with lead compounds showing liver fat-lowering efficacy in phase II clinical trials. In humans with NAFLD, percent reductions in liver fat have closely mirrored percent inhibition of DNL, thereby affirming the critical contributions of DNL to NAFLD pathogenesis. The safety profiles of these compounds have so far been encouraging. It is anticipated that inhibitors of DNL, when administered alone or in combination with other therapeutic agents, will become important agents in the management of human NAFLD.
Collapse
Affiliation(s)
- William P Esler
- Internal Medicine Research Unit, Pfizer Worldwide Research Development and Medical, Cambridge, MA 02139 United States.
| | - David E Cohen
- Division of Gastroenterology, Hepatology and Endoscopy, Brigham and Women's Hospital and Harvard Medical School, Boston, MA 02115 United States.
| |
Collapse
|
62
|
Moore E, Patanwala I, Jafari A, Davies IG, Kirwan RP, Newson L, Mazidi M, Lane KE. A systematic review and meta-analysis of randomized controlled trials to evaluate plant-based omega-3 polyunsaturated fatty acids in nonalcoholic fatty liver disease patient biomarkers and parameters. Nutr Rev 2024; 82:143-165. [PMID: 37290426 PMCID: PMC10777680 DOI: 10.1093/nutrit/nuad054] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/10/2023] Open
Abstract
CONTEXT Nonalcoholic fatty liver disease (NAFLD) is prevalent in 25-30% of British and European populations, representing a potential global public health crisis. Marine omega-3 (n-3) polyunsaturated fatty acids offer well-evidenced benefits to NAFLD biomarkers; however, the effect of plant-based n-3 has not been evaluated with a systematic review and meta-analysis. OBJECTIVE The review aimed to systematically evaluate the effect of plant-based n-3 supplementation on NAFLD surrogate biomarkers and parameters. DATA SOURCES Medline (EBSCO), PubMed, CINAHL (EBSCO), Cochrane Central Register of Controlled Trials, the International Clinical Trials Registry Platform, and Google Scholar databases were searched to identify randomized controlled trials published between January 1970 and March 2022 evaluating the impact of plant-based n-3 interventions on diagnosed NAFLD. The review followed the PRISMA checklist and is PROSPERO registered (CRD42021251980). DATA EXTRACTION A random-effects model and generic inverse variance methods synthesized quantitative data, followed by a leave-one-out method for sensitivity analysis. We identified 986 articles; after the application of selection criteria, six studies remained with 362 patients with NAFLD. RESULTS The meta-analysis showed that plant-based n-3 fatty acid supplementation significantly reduced alanine aminotransferase (ALT) (mean difference: 8.04 IU/L; 95% confidence interval: 14.70, 1.38; I2 = 48.61%) and plasma/serum triglycerides (44.51 mg/dL; 95% confidence interval: -76.93, -12.08; I2 = 69.93%), alongside body-composition markers in patients with NAFLD (P < 0.05). CONCLUSION Plant-based n-3 fatty acid supplementation improves ALT enzyme biomarkers, triglycerides, body mass index, waist circumference, and weight loss when combined with lifestyle interventions to increase physical activity and a calorie-controlled diet. Further research is needed to identify the most effective plant-based n-3 sources in larger numbers of patients with NAFLD over longer study durations. SYSTEMATIC REVIEW REGISTRATION PROSPERO registration no. CRD42021251980.
Collapse
Affiliation(s)
- Ella Moore
- Research Institute for Sport and Exercise Sciences, Liverpool John Moores University, Liverpool, United Kingdom
| | | | - Alireza Jafari
- Physiology Research Center, Institute of Neuropharmacology, Kerman University of Medical Sciences, Kerman, Iran
| | - Ian G Davies
- Research Institute for Sport and Exercise Sciences, Liverpool John Moores University, Liverpool, United Kingdom
| | - Richard P Kirwan
- Research Institute for Sport and Exercise Sciences, Liverpool John Moores University, Liverpool, United Kingdom
| | - Lisa Newson
- School of Psychology, Liverpool John Moores University, Liverpool, United Kingdom
| | - Mohsen Mazidi
- Medical Research Council Population Health Research Unit, Clinical Trial Service Unit and Epidemiological Studies Unit (CTSU), Nuffield Department of Population Health, University of Oxford, Oxford, United Kingdom
- Department of Twin Research and Genetic Epidemiology, King’s College London, London, United Kingdom
| | - Katie E Lane
- Research Institute for Sport and Exercise Sciences, Liverpool John Moores University, Liverpool, United Kingdom
| |
Collapse
|
63
|
Sanyal AJ, Jha P, Kleiner DE. Digital pathology for nonalcoholic steatohepatitis assessment. Nat Rev Gastroenterol Hepatol 2024; 21:57-69. [PMID: 37789057 DOI: 10.1038/s41575-023-00843-7] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 08/23/2023] [Indexed: 10/05/2023]
Abstract
Histological assessment of nonalcoholic fatty liver disease (NAFLD) has anchored knowledge development about the phenotypes of the condition, their natural history and their clinical course. This fact has led to the use of histological assessment as a reference standard for the evaluation of efficacy of drug interventions for nonalcoholic steatohepatitis (NASH) - the more histologically active form of NAFLD. However, certain limitations of conventional histological assessment systems pose challenges in drug development. These limitations have spurred intense scientific and commercial development of machine learning and digital approaches towards the assessment of liver histology in patients with NAFLD. This research field remains an area in rapid evolution. In this Perspective article, we summarize the current conventional assessment of NASH and its limitations, the use of specific digital approaches for histological assessment, and their application to the study of NASH and its response to therapy. Although this is not a comprehensive review, the leading tools currently used to assess therapeutic efficacy in drug development are specifically discussed. The potential translation of these approaches to support routine clinical assessment of NAFLD and an agenda for future research are also discussed.
Collapse
Affiliation(s)
- Arun J Sanyal
- Stravitz-Sanyal Institute for Liver Disease and Metabolic Health, Virginia Commonwealth University School of Medicine, Richmond, VA, USA.
| | - Prakash Jha
- Food and Drug Administration, Silver Spring, MD, USA
| | - David E Kleiner
- Post-Mortem Section Laboratory of Pathology Center for Cancer Research National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| |
Collapse
|
64
|
Noguerol EDC, de Souza LRMF, Muglia VF, Elias J. Quantification of hepatic fat: evaluation of different magnetic resonance imaging measurement strategies in cases of homogeneous and heterogeneous distribution. Radiol Bras 2024; 57:e20240009en. [PMID: 39563789 PMCID: PMC11575847 DOI: 10.1590/0100-3984.2024.0009-en] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2024] [Revised: 07/09/2024] [Accepted: 08/04/2024] [Indexed: 11/21/2024] Open
Abstract
Objective To evaluate three different measurements strategies to quantify hepatic steatosis and to investigate the differences between homogeneous and heterogeneous forms of hepatic steatosis. Materials and Methods Retrospective study conducted by magnetic resonance imaging review. We evaluated three different strategies measures for quantification of hepatic steatosis in two matched groups: homogeneous and heterogeneous steatosis. We considered p < 0.05 significance level in all made tests. Results In heterogeneous steatosis group, the strategy with a region of interest (ROI) of 1 cm2 to measure the signal intensity in the most altered area showed significant variations in the quantification, while the average of four ROIs of 1 cm2 or representative target area in axial section did not vary significant. In diffuse hepatic steatosis, any strategy used showed no significant difference. The intraclass correlation coefficient ranged between 0.96 and 0.99, with 95% confidence interval of 0.93-0.99. Conclusion The quantification of fat liver by magnetic resonance imaging using only one ROI is less representative, especially in heterogeneous steatosis. There was no significant difference between the average of four ROIs strategy and the strategy of representative segmentation area of parenchyma.
Collapse
Affiliation(s)
- Eloa de Castro Noguerol
- Faculdade de Medicina de Ribeirão Preto da Universidade de São Paulo (FMRP-USP), Ribeirão Preto, SP, Brazil
| | | | - Valdair Francisco Muglia
- Faculdade de Medicina de Ribeirão Preto da Universidade de São Paulo (FMRP-USP), Ribeirão Preto, SP, Brazil
| | - Jorge Elias
- Faculdade de Medicina de Ribeirão Preto da Universidade de São Paulo (FMRP-USP), Ribeirão Preto, SP, Brazil
| |
Collapse
|
65
|
Kadi D, Loomba R, Bashir MR. Diagnosis and Monitoring of Nonalcoholic Steatohepatitis: Current State and Future Directions. Radiology 2024; 310:e222695. [PMID: 38226882 DOI: 10.1148/radiol.222695] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2024]
Abstract
Nonalcoholic fatty liver disease (NAFLD) is a common liver disease, with a worldwide prevalence of 25%. NAFLD is a spectrum that includes nonalcoholic fatty liver defined histologically by isolated hepatocytes steatosis without inflammation and nonalcoholic steatohepatitis (NASH) is the inflammatory subtype of NAFLD and is associated with disease progression, development of cirrhosis, and increased rates of liver-specific and overall mortality. The differentiation between NAFLD and NASH as well as staging NASH are important yet challenging clinical problems. Liver biopsy is currently the standard for disease diagnosis and fibrosis staging. However, this procedure is invasive, costly, and cannot be used for longitudinal monitoring. Therefore, several noninvasive quantitative imaging biomarkers have been proposed that can estimate the severity of hepatic steatosis and fibrosis. Despite this, noninvasive diagnosis of NASH and accurate risk stratification remain unmet needs. In this work, the most relevant available imaging biomarkers are reviewed and their application in patients with NAFLD are discussed.
Collapse
Affiliation(s)
- Diana Kadi
- From the Department of Radiology (D.K., M.R.B.), Center for Advanced Magnetic Resonance Development (M.R.B.), Department of Pathology (M.R.B.), and Division of Hepatology (M.R.B.), Duke University Medical Center, Durham, NC 27705; and Division of Gastroenterology, Department of Medicine, NAFLD Research Center, University of California at San Diego, La Jolla, Calif (R.L.)
| | - Rohit Loomba
- From the Department of Radiology (D.K., M.R.B.), Center for Advanced Magnetic Resonance Development (M.R.B.), Department of Pathology (M.R.B.), and Division of Hepatology (M.R.B.), Duke University Medical Center, Durham, NC 27705; and Division of Gastroenterology, Department of Medicine, NAFLD Research Center, University of California at San Diego, La Jolla, Calif (R.L.)
| | - Mustafa R Bashir
- From the Department of Radiology (D.K., M.R.B.), Center for Advanced Magnetic Resonance Development (M.R.B.), Department of Pathology (M.R.B.), and Division of Hepatology (M.R.B.), Duke University Medical Center, Durham, NC 27705; and Division of Gastroenterology, Department of Medicine, NAFLD Research Center, University of California at San Diego, La Jolla, Calif (R.L.)
| |
Collapse
|
66
|
Zhang X, Li G, Lin H, Wong VWS, Wong GLH. Noninvasive evaluation of liver fibrosis in MASLD—Imaging/elastography based. METABOLIC STEATOTIC LIVER DISEASE 2024:151-166. [DOI: 10.1016/b978-0-323-99649-5.00005-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/04/2025]
|
67
|
Franck M, John K, Al Aoua S, Rau M, Geier A, Schattenberg JM, Wedemeyer H, Schulze-Osthoff K, Bantel H. Hepatokine-based identification of fibrotic NASH and improved risk stratification in a multicentre cohort of NAFLD patients. Liver Int 2023; 43:2668-2679. [PMID: 37534777 DOI: 10.1111/liv.15686] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/13/2023] [Revised: 07/04/2023] [Accepted: 07/22/2023] [Indexed: 08/04/2023]
Abstract
BACKGROUND AND AIMS The presence of significant liver fibrosis associated with non-alcoholic steatohepatitis (NASH) is regarded as the major prognostic factor in non-alcoholic fatty liver disease (NAFLD). Identification of patients at risk for NASH with significant fibrosis is therefore important. Although the established fibrosis score FIB-4 is suitable to exclude advanced fibrosis, it does not allow the prediction of significant fibrosis in NAFLD patients. We therefore evaluated whether the hepatokine fibroblast growth factor 21 (FGF21), a regulator of glucose and lipid metabolism, might identify 'at-risk NASH' in NAFLD. METHODS FGF21 levels were assessed by enzyme-linked immunosorbent assay in sera from an exploration (n = 137) and a validation (n = 88) cohort of biopsy-proven NAFLD patients with different disease activity and fibrosis stages. In addition, we evaluated whether the use of FGF21 could improve risk stratification in NAFLD patients with low (<1.3) or intermediate (1.3-2.67) FIB-4. RESULTS FGF21 levels could significantly discriminate between NASH and non-alcoholic fatty liver (NAFL) patients, even in the absence of diabetes. Moreover, patients with NASH and fibrosis ≥F2 showed significantly higher FGF21 levels compared to NAFLD patients without significant fibrosis. Significantly elevated FGF21 levels could even be detected in NAFLD patients with NASH and significant fibrosis despite low or intermediate FIB-4. CONCLUSION Serological FGF21 detection might allow the identification of NAFLD patients at risk and improves patient stratification in combination with FIB-4.
Collapse
Affiliation(s)
- Martin Franck
- Department of Gastroenterology, Hepatology, Infectious Diseases and Endocrinology, Hannover Medical School, Hannover, Germany
| | - Katharina John
- Department of Gastroenterology, Hepatology, Infectious Diseases and Endocrinology, Hannover Medical School, Hannover, Germany
| | - Sherin Al Aoua
- Department of Gastroenterology, Hepatology, Infectious Diseases and Endocrinology, Hannover Medical School, Hannover, Germany
| | - Monika Rau
- Division of Hepatology, Department of Internal Medicine II, University Hospital Würzburg, Würzburg, Germany
| | - Andreas Geier
- Division of Hepatology, Department of Internal Medicine II, University Hospital Würzburg, Würzburg, Germany
| | - Jörn M Schattenberg
- Department of Internal Medicine I, University Medical Center Mainz, Mainz, Germany
| | - Heiner Wedemeyer
- Department of Gastroenterology, Hepatology, Infectious Diseases and Endocrinology, Hannover Medical School, Hannover, Germany
| | - Klaus Schulze-Osthoff
- Interfaculty Institute of Biochemistry, University of Tuebingen, Tuebingen, Germany
- German Cancer Consortium (DKTK) and German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Heike Bantel
- Department of Gastroenterology, Hepatology, Infectious Diseases and Endocrinology, Hannover Medical School, Hannover, Germany
| |
Collapse
|
68
|
Keating SE, Sabag A, Hallsworth K, Hickman IJ, Macdonald GA, Stine JG, George J, Johnson NA. Exercise in the Management of Metabolic-Associated Fatty Liver Disease (MAFLD) in Adults: A Position Statement from Exercise and Sport Science Australia. Sports Med 2023; 53:2347-2371. [PMID: 37695493 PMCID: PMC10687186 DOI: 10.1007/s40279-023-01918-w] [Citation(s) in RCA: 31] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/18/2023] [Indexed: 09/12/2023]
Abstract
Metabolic-associated fatty liver disease (MAFLD) is the most prevalent chronic liver disease worldwide, affecting 25% of people globally and up to 80% of people with obesity. MAFLD is characterised by fat accumulation in the liver (hepatic steatosis) with varying degrees of inflammation and fibrosis. MAFLD is strongly linked with cardiometabolic disease and lifestyle-related cancers, in addition to heightened liver-related morbidity and mortality. This position statement examines evidence for exercise in the management of MAFLD and describes the role of the exercise professional in the context of the multi-disciplinary care team. The purpose of these guidelines is to equip the exercise professional with a broad understanding of the pathophysiological underpinnings of MAFLD, how it is diagnosed and managed in clinical practice, and to provide evidence- and consensus-based recommendations for exercise therapy in MAFLD management. The majority of research evidence indicates that 150-240 min per week of at least moderate-intensity aerobic exercise can reduce hepatic steatosis by ~ 2-4% (absolute reduction), but as little as 135 min/week has been shown to be effective. While emerging evidence shows that high-intensity interval training (HIIT) approaches may provide comparable benefit on hepatic steatosis, there does not appear to be an intensity-dependent benefit, as long as the recommended exercise volume is achieved. This dose of exercise is likely to also reduce central adiposity, increase cardiorespiratory fitness and improve cardiometabolic health, irrespective of weight loss. Resistance training should be considered in addition to, and not instead of, aerobic exercise targets. The information in this statement is relevant and appropriate for people living with the condition historically termed non-alcoholic fatty liver disease (NAFLD), regardless of terminology.
Collapse
Affiliation(s)
- Shelley E Keating
- School of Human Movement and Nutrition Sciences, The University of Queensland, Room 534, Bd 26B, St Lucia, Brisbane, QLD, 4067, Australia.
| | - Angelo Sabag
- Faculty of Medicine and Health, Discipline of Exercise and Sport Science, University of Sydney, Sydney, NSW, Australia
- Charles Perkins Centre, University of Sydney, Camperdown, NSW, Australia
- NICM Health Research Institute, Western Sydney University, Westmead, NSW, Australia
| | - Kate Hallsworth
- NIHR Newcastle Biomedical Research Centre, Newcastle Upon Tyne Hospitals NHS Foundation Trust, Newcastle Upon Tyne, UK
- Liver Unit, Newcastle Upon Tyne Hospitals NHS Foundation Trust, Newcastle Upon Tyne, UK
- Translational and Clinical Research Institute, Faculty of Medical Sciences, Newcastle University, Newcastle Upon Tyne, UK
| | - Ingrid J Hickman
- Department of Nutrition and Dietetics, Princess Alexandra Hospital, Brisbane, QLD, Australia
- Faculty of Medicine, PA-Southside Clinical Unit, The University of Queensland, Brisbane, QLD, Australia
| | - Graeme A Macdonald
- Faculty of Medicine, PA-Southside Clinical Unit, The University of Queensland, Brisbane, QLD, Australia
- Department of Gastroenterology and Hepatology, Princess Alexandra Hospital, Brisbane, QLD, Australia
| | - Jonathan G Stine
- Division of Gastroenterology and Hepatology, Department of Medicine, The Pennsylvania State University- Milton S. Hershey Medical Center, Hershey, PA, USA
- Department of Public Health Sciences, The Pennsylvania State University- College of Medicine, Hershey, PA, USA
- Liver Center, The Pennsylvania State University- Milton S. Hershey Medical Center, Hershey, PA, USA
- Cancer Institute, The Pennsylvania State University- Milton S. Hershey Medical Center, Hershey, PA, USA
| | - Jacob George
- Storr Liver Centre, The Westmead Institute for Medical Research and Westmead Hospital, University of Sydney, Sydney, NSW, Australia
| | - Nathan A Johnson
- Faculty of Medicine and Health, Discipline of Exercise and Sport Science, University of Sydney, Sydney, NSW, Australia
- Charles Perkins Centre, University of Sydney, Camperdown, NSW, Australia
| |
Collapse
|
69
|
Noureddin M, Khan S, Portell F, Jorkasky D, Dennis J, Khan O, Johansson L, Johansson E, Sanyal AJ. Safety and efficacy of once-daily HU6 versus placebo in people with non-alcoholic fatty liver disease and high BMI: a randomised, double-blind, placebo-controlled, phase 2a trial. Lancet Gastroenterol Hepatol 2023; 8:1094-1105. [PMID: 37806314 DOI: 10.1016/s2468-1253(23)00198-x] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/20/2023] [Revised: 06/26/2023] [Accepted: 06/27/2023] [Indexed: 10/10/2023]
Abstract
BACKGROUND HU6 is a controlled metabolic accelerator that is metabolised in the liver to the mitochondrial uncoupler 2,4-dinitrophenol and increases substrate utilisation so that fat and other carbon sources are oxidised in the body rather than accumulated. We aimed to assess the safety and efficacy of HU6 compared with placebo in people with non-alcoholic fatty liver disease (NAFLD) and high BMI. METHODS This randomised, double-blind, placebo-controlled, phase 2a trial was done at a single community site in the USA. Adults (aged 28-65 years) with a BMI of 28-45 kg/m2, a FibroScan controlled attenuation parameter score of more than 270 decibels per metre, and at least 8% liver fat by MRI-proton density fat fraction (MRI-PDFF) were randomly assigned (1:1:1:1) to receive, under fasting conditions, either once-daily HU6 100 mg, HU6 300 mg, HU6 450 mg, or matching placebo by oral administration for 61 days. Randomisation was blocked (groups of four) and stratified by baseline glycated haemoglobin (<5·7% vs ≥5·7%; 39 mmol/mol). All participants and study personnel involved with outcome assessments were masked to treatment assignment. The primary endpoint was the relative change in liver fat content from baseline to day 61, as assessed by MRI-PDFF, and was analysed in the full analysis set (FAS), which comprised all participants who were randomly assigned, received at least one dose of treatment, and had less than 4·5 kg of weight gain or weight loss from the time of screening to day 1 of treatment. The safety population included all participants who were randomly assigned and received at least one dose of study drug. This study was registered at ClinicalTrials.gov, NCT04874233, and is complete. FINDINGS Between April 28, 2021, and Nov 29, 2021, 506 participants were assessed for eligibility and 80 adults (39 [49%] women and 41 [51%] men) were enrolled and randomly assigned to placebo (n=20), HU6 150 mg (n=20), HU6 300 mg (n=21), or HU6 450 mg (n=19). One participant in the HU6 450 mg group was excluded from the FAS due to weight gain. Relative mean change in liver fat content from baseline to day 61 was -26·8% (SD 17·4) for the HU6 150 mg group, -35·6% (13·8) for the HU6 300 mg group, -33·0% (18·4) for the HU6 450 mg group, and 5·4% (19·8) for the placebo group. Three people treated with HU6 (two treated with 150 mg and one treated with 300 mg) and two people treated with placebo discontinued treatment due to treatment-emergent adverse events (TEAEs). No serious TEAEs were reported. In those treated with HU6, flushing (19 [32%] participants), diarrhoea (15 [25%] participants), and palpitations (seven [12%] participants) were the most frequently reported TEAEs (in the placebo group, two [10%] participants had flushing, none had diarrhoea, and one [5%] had palpitations). There were no deaths. INTERPRETATION HU6 could be a promising pharmacological agent for treating patients with obesity and NAFLD and its metabolic complications. FUNDING Rivus Pharmaceuticals.
Collapse
Affiliation(s)
- Mazen Noureddin
- Houston Research Institute, Houston Methodist Hospital, Houston, TX, USA.
| | | | | | | | | | - Omer Khan
- Rivus Pharmaceuticals, Charlottesville, VA, USA
| | | | | | - Arun J Sanyal
- Stravitz-Sanyal Institute of Liver Disease and Metabolic Health, Department of Internal Medicine, VCU School of Medicine, Richmond, VA, USA
| |
Collapse
|
70
|
Malandris K, Papandreou S, Avgerinos I, Karagiannis T, Paschos P, Michailidis T, Liakos A, Bekiari E, Sinakos E, Tsapas A. Comparative efficacy of glucose-lowering drugs on liver steatosis as assessed by means of magnetic resonance imaging in patients with type 2 diabetes mellitus: systematic review and network meta-analysis. Hormones (Athens) 2023; 22:655-664. [PMID: 37770761 PMCID: PMC10651545 DOI: 10.1007/s42000-023-00493-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/14/2023] [Accepted: 09/18/2023] [Indexed: 09/30/2023]
Abstract
PURPOSE To assess the comparative efficacy of glucose-lowering drugs on liver steatosis as assessed by means of magnetic resonance imaging (MRI) in patients with T2D. METHODS We searched several databases and grey literature sources. Eligible trials had at least 12 weeks of intervention, included patients with T2D, and assessed the efficacy of glucose-lowering drugs as monotherapies. The primary outcome of interest was absolute reduction in liver fat content (LFC), assessed by means of MRI. Secondary efficacy outcomes were reduction in visceral and subcutaneous adipose tissue. We performed random effects frequentist network meta-analyses to estimate mean differences (MDs) with 95% confidence intervals (CIs). We ranked treatments based on P-scores. RESULTS We included 29 trials with 1906 patients. Sodium-glucose cotransporter-2 (SGLT-2) inhibitors (P-score 0.84) and glucagon-like peptide-1 receptor agonists (GLP-1 RAs) (0.71) were the most efficacious in terms of liver fat content reduction. Among individual agents, empagliflozin was the most efficacious (0.86) and superior to pioglitazone (MD -5.7, 95% CI -11.2 to -0.3) (very low confidence). GLP-1 RAs had also the most favorable effects on visceral and subcutaneous adipose tissue. CONCLUSIONS GLP-1 RAs and SGLT-2 inhibitors seem to be the most efficacious glucose-lowering drugs for liver steatosis in patients with T2D. Assessment of their efficacy on NAFLD in patients irrespective of presence of T2D is encouraged.
Collapse
Affiliation(s)
- Konstantinos Malandris
- Clinical Research and Evidence-Based Medicine Unit, Second Medical Department, Aristotle University of Thessaloniki, Thessaloniki, Greece.
| | - Stylianos Papandreou
- Clinical Research and Evidence-Based Medicine Unit, Second Medical Department, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Ioannis Avgerinos
- Clinical Research and Evidence-Based Medicine Unit, Second Medical Department, Aristotle University of Thessaloniki, Thessaloniki, Greece
- Diabetes Centre, Second Medical Department, Aristotle University of Thessaloniki, Konstantinoupoleos 49, 54642, Thessaloniki, Greece
| | - Thomas Karagiannis
- Clinical Research and Evidence-Based Medicine Unit, Second Medical Department, Aristotle University of Thessaloniki, Thessaloniki, Greece
- Diabetes Centre, Second Medical Department, Aristotle University of Thessaloniki, Konstantinoupoleos 49, 54642, Thessaloniki, Greece
| | - Paschalis Paschos
- First Medical Department, "Papageorgiou" Hospital, Thessaloniki, Greece
| | - Theodoros Michailidis
- Diabetes Centre, Second Medical Department, Aristotle University of Thessaloniki, Konstantinoupoleos 49, 54642, Thessaloniki, Greece
| | - Aris Liakos
- Clinical Research and Evidence-Based Medicine Unit, Second Medical Department, Aristotle University of Thessaloniki, Thessaloniki, Greece
- Diabetes Centre, Second Medical Department, Aristotle University of Thessaloniki, Konstantinoupoleos 49, 54642, Thessaloniki, Greece
| | - Eleni Bekiari
- Clinical Research and Evidence-Based Medicine Unit, Second Medical Department, Aristotle University of Thessaloniki, Thessaloniki, Greece
- Diabetes Centre, Second Medical Department, Aristotle University of Thessaloniki, Konstantinoupoleos 49, 54642, Thessaloniki, Greece
| | - Emmanouil Sinakos
- Fourth Medical Department, Hippokration General Hospital of Thessaloniki, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Apostolos Tsapas
- Clinical Research and Evidence-Based Medicine Unit, Second Medical Department, Aristotle University of Thessaloniki, Thessaloniki, Greece
- Diabetes Centre, Second Medical Department, Aristotle University of Thessaloniki, Konstantinoupoleos 49, 54642, Thessaloniki, Greece
- Harris Manchester College, University of Oxford, Oxford, UK
| |
Collapse
|
71
|
Mu R, Xia YC, Zhu KY, Lu JY, Luo Q, Zhang L, Lin RK, Cai XB, Qu Y, Lu LG. Diagnostic value of FibroTouch in identifying hepatic steatosis in NAFLD with MRI-PDFF as the reference standard. J Dig Dis 2023; 24:691-701. [PMID: 37994615 DOI: 10.1111/1751-2980.13242] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/21/2023] [Revised: 05/31/2023] [Accepted: 11/21/2023] [Indexed: 11/24/2023]
Abstract
OBJECTIVE To estimate the performance of the FibroTouch-based ultrasound attenuation parameter (UAP) for assessing hepatic steatosis in nonalcoholic fatty liver disease (NAFLD), with magnetic resonance imaging proton density fat fraction (MRI-PDFF) as the reference standard. METHODS This prospective, cross-sectional study included 275 individuals in the training group and 110 individuals in the validation group, all of whom completed a standardized research visit, laboratory tests, MRI-PDFF, and UAP measurements over 1 month. Pearson correlation coefficient and Bland-Altman analysis were used to assess the agreement between UAP and MRI-PDFF for the detection of hepatic steatosis. The diagnostic value of UAP was evaluated by the area under the receiver operating characteristic (ROC) curve (AUROC). Confounding factors to UAP performance were identified by ROC curves and regression analyses. RESULTS The AUROC of UAP for detecting MRI-PDFF at ≥5%, ≥10%, and ≥20% were 0.95 (95% confidence interval [CI] 0.92-0.97), 0.86 (95% CI 0.81-0.90), and 0.90 (95% CI 0.86-0.93), respectively, and their optimal thresholds were 259, 274, and 295 dB/m, respectively. The UAP measurements had higher diagnostic accuracy in participants with lower waist circumference (≤90 cm for men and ≤80 cm for women) compared to those with higher waist circumference (AUROC values: 0.97 vs 0.84, P < 0.05). Bland-Altman analysis showed good agreement between UAP and MRI-PDFF (bias 0.00021). According to established regression analyses, hepatic steatosis could be accurately diagnosed using UAP estimation. CONCLUSIONS FibroTouch-UAP has a high diagnostic potential for hepatic steatosis in NAFLD patients and helps clinical assessment and monitoring.
Collapse
Affiliation(s)
- Rong Mu
- Department of Gastroenterology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - You Chen Xia
- Department of Gastroenterology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Kou Yun Zhu
- Department of Gastroenterology, Baoshan Branch, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jing Yi Lu
- Department of Gastroenterology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Qian Luo
- Department of Radiology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Lei Zhang
- Department of Radiology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Ren Kun Lin
- Department of Gastroenterology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xiao Bo Cai
- Department of Gastroenterology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Ying Qu
- Department of Gastroenterology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Lun Gen Lu
- Department of Gastroenterology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
72
|
James M, Alazawi W. Non-invasive Tests of Fibrosis: Does One Size Fit All? Dig Dis Sci 2023; 68:4285-4286. [PMID: 37792129 DOI: 10.1007/s10620-023-08094-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 08/01/2023] [Indexed: 10/05/2023]
Affiliation(s)
- Michael James
- Centre for Immunobiology, Blizard Institute, Queen Mary University of London, 4 Newark Street, London, E1 2AT, UK
| | - William Alazawi
- Centre for Immunobiology, Blizard Institute, Queen Mary University of London, 4 Newark Street, London, E1 2AT, UK.
| |
Collapse
|
73
|
Harrison SA, Taub R, Neff GW, Lucas KJ, Labriola D, Moussa SE, Alkhouri N, Bashir MR. Resmetirom for nonalcoholic fatty liver disease: a randomized, double-blind, placebo-controlled phase 3 trial. Nat Med 2023; 29:2919-2928. [PMID: 37845512 PMCID: PMC10667098 DOI: 10.1038/s41591-023-02603-1] [Citation(s) in RCA: 184] [Impact Index Per Article: 92.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2023] [Accepted: 09/20/2023] [Indexed: 10/18/2023]
Abstract
Nonalcoholic steatohepatitis (NASH) is a progressive liver disease with no approved treatment. MAESTRO-NAFLD-1 was a 52-week randomized, double-blind, placebo-controlled phase 3 trial evaluating the safety of resmetirom in adults with nonalcoholic fatty liver disease and presumed NASH. Patients were randomized to three double-blind arms (100 mg resmetirom (n = 325), 80 mg resmetirom (n = 327) or placebo (n = 320)) or open-label 100 mg resmetirom (n = 171). The primary end point was incidence of treatment-emergent adverse events (TEAEs) over 52 weeks and key secondary end points were LDL-C, apoB, triglycerides (over 24 weeks), hepatic fat (over 16 and 52 weeks) and liver stiffness (over 52 weeks). Resmetirom was safe and well tolerated. TEAEs occurred in 86.5% (open-label 100 mg resmetirom), 86.1% (100 mg resmetirom), 88.4% (80 mg resmetirom) and 81.8% (placebo) of patients. TEAEs in excess of placebo included diarrhea and nausea at the initiation of treatment. Key secondary end points included least square means difference from placebo at 80 mg, 100 mg resmetirom: LDL-C (-11.1%, -12.6%), apoB (-15.6%, -18.0%), triglycerides (-15.4%, -20.4%), 16-week hepatic fat (-34.9%, -38.6%), (P < 0.0001) and liver stiffness (-1.02, -1.70) and 52-week hepatic fat (-28.8, -33.9). These findings demonstrate resmetirom was safe and well tolerated in adults with presumed NASH, supporting a role for further clinical development. (ClinicalTrials.gov identifier NCT04197479 ).
Collapse
Affiliation(s)
| | | | - Guy W Neff
- Covenant Metabolic Specialists, Sarasota, FL, USA
| | | | | | - Sam E Moussa
- University of Arizona for Medical Sciences, Tucson, AZ, USA
| | | | | |
Collapse
|
74
|
Dumont C, Lanthier N, Dahlqvist G. Fibrosis and steatosis of the liver graft: Are non-invasive tests useful? A short review. Clin Res Hepatol Gastroenterol 2023; 47:102194. [PMID: 37567467 DOI: 10.1016/j.clinre.2023.102194] [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: 06/05/2023] [Revised: 07/31/2023] [Accepted: 08/02/2023] [Indexed: 08/13/2023]
Abstract
As life expectancy of liver transplanted patients improves, new questions are arising to avoid progressive graft loss. The spectrum of chronic inflammation and fibrosis are known to be important triggers in the alteration of graft function. Liver biopsy remains the gold standard to better understand progressive, normal, and abnormal histological modifications of the graft. In parallel, the interest for metabolic steatosis development in post-transplantation is also growing. Long-term survival of these patients involves the management of comorbidities including metabolic syndrome and cardiovascular diseases. Early detection of altered graft parenchyma, and monitoring of its evolution are undoubtedly essential. Non-invasive methods including transient elastography and fibrosis biomarkers are attractive tools to avoid drawbacks and complications of liver biopsy. Accuracy of these methods are well-known in a pre-transplantation setting, but evidence is lacking in post-transplantation setting. We review current knowledge of progressive liver fibrosis and steatosis development after transplantation and non-invasive methods of their assessment.
Collapse
Affiliation(s)
- Colin Dumont
- Department of Gastroenterology and Hepatology, Cliniques Universitaires Saint-Luc, UCLouvain, Brussels, Belgium
| | - Nicolas Lanthier
- Department of Gastroenterology and Hepatology, Cliniques Universitaires Saint-Luc, UCLouvain, Brussels, Belgium; Laboratory of Gastroenterology and Hepatology, Institut de Recherche Expérimentale et Clinique, UCLouvain, Brussels, Belgium
| | - Géraldine Dahlqvist
- Department of Gastroenterology and Hepatology, Cliniques Universitaires Saint-Luc, UCLouvain, Brussels, Belgium.
| |
Collapse
|
75
|
Tilg H, Byrne CD, Targher G. NASH drug treatment development: challenges and lessons. Lancet Gastroenterol Hepatol 2023; 8:943-954. [PMID: 37597527 DOI: 10.1016/s2468-1253(23)00159-0] [Citation(s) in RCA: 36] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/11/2023] [Revised: 05/19/2023] [Accepted: 05/19/2023] [Indexed: 08/21/2023]
Abstract
Non-alcoholic fatty liver disease (NAFLD) has become the most common chronic liver disease worldwide. Although NAFLD is tightly linked to obesity and type 2 diabetes, this liver disease also affects individuals who do not have obesity. NAFLD increases the risk of developing cardiovascular disease, chronic kidney disease, and certain extrahepatic cancers. There is currently no licensed pharmacotherapy for NAFLD, despite numerous clinical trials in the past two decades. Currently, the reason so few drugs have been successful in the treatment of NAFLD in a trial setting is not fully understood. As cardiovascular disease is the predominant cause of mortality in people with NAFLD, future pharmacotherapies for NAFLD must consider associated cardiometabolic risk factors. The successful use of glucose-lowering drugs in the treatment of type 2 diabetes in patients with NAFLD indicates that this strategy is important, and worth developing further. Greater public awareness of NAFLD is needed because collaboration between all stakeholders is vital to enable a holistic approach to successful treatment.
Collapse
Affiliation(s)
- Herbert Tilg
- Department of Internal Medicine I, Gastroenterology, Hepatology, Endocrinology & Metabolism, Medical University Innsbruck, Innsbruck, Austria.
| | - Christopher D Byrne
- National Institute for Health and Care Research, Southampton Biomedical Research Centre, University Hospital Southampton and University of Southampton, Southampton, UK
| | - Giovanni Targher
- Endocrinology, Diabetes, and Metabolism, Department of Medicine, University of Verona, Verona, Italy; IRCCS Ospedale Sacro Cuore Don Calabria, Negrar di Valpolicella, Italy
| |
Collapse
|
76
|
Demir M, Bornstein SR, Mantzoros CS, Perakakis N. Liver fat as risk factor of hepatic and cardiometabolic diseases. Obes Rev 2023; 24:e13612. [PMID: 37553237 DOI: 10.1111/obr.13612] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/07/2022] [Revised: 06/26/2023] [Accepted: 07/10/2023] [Indexed: 08/10/2023]
Abstract
Non-alcoholic fatty liver disease (NAFLD) is a disorder characterized by excessive accumulation of fat in the liver that can progress to liver inflammation (non-alcoholic steatohepatitis [NASH]), liver fibrosis, and cirrhosis. Although most efforts for drug development are focusing on the treatment of the latest stages of NAFLD, where significant fibrosis and NASH are present, findings from studies suggest that the amount of liver fat may be an important independent risk factor and/or predictor of development and progression of NAFLD and metabolic diseases. In this review, we first describe the current tools available for quantification of liver fat in humans and then present the clinical and pathophysiological evidence that link liver fat with NAFLD progression as well as with cardiometabolic diseases. Finally, we discuss current pharmacological and non-pharmacological approaches to reduce liver fat and present open questions that have to be addressed in future studies.
Collapse
Affiliation(s)
- Münevver Demir
- Department of Hepatology and Gastroenterology, Campus Virchow Clinic and Campus Charité Mitte, Charité University Medicine, Berlin, Germany
| | - Stefan R Bornstein
- Department of Internal Medicine III, University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
- Paul Langerhans Institute Dresden (PLID), Helmholtz Center Munich, University Hospital and Faculty of Medicine, TU Dresden, Dresden, Germany
- German Center for Diabetes Research (DZD e.V.), Neuherberg, Germany
- Diabetes and Nutritional Sciences, King's College London, London, UK
| | - Christos S Mantzoros
- Division of Endocrinology, Boston VA Healthcare System and Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, 02215, USA
| | - Nikolaos Perakakis
- Department of Internal Medicine III, University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
- Paul Langerhans Institute Dresden (PLID), Helmholtz Center Munich, University Hospital and Faculty of Medicine, TU Dresden, Dresden, Germany
- German Center for Diabetes Research (DZD e.V.), Neuherberg, Germany
| |
Collapse
|
77
|
Anstee QM, Lucas KJ, Francque S, Abdelmalek MF, Sanyal AJ, Ratziu V, Gadano AC, Rinella M, Charlton M, Loomba R, Mena E, Schattenberg JM, Noureddin M, Lazas D, Goh GB, Sarin SK, Yilmaz Y, Martic M, Stringer R, Kochuparampil J, Chen L, Rodriguez-Araujo G, Chng E, Naoumov NV, Brass C, Pedrosa MC. Tropifexor plus cenicriviroc combination versus monotherapy in nonalcoholic steatohepatitis: Results from the phase 2b TANDEM study. Hepatology 2023; 78:1223-1239. [PMID: 37162151 PMCID: PMC10521801 DOI: 10.1097/hep.0000000000000439] [Citation(s) in RCA: 30] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/23/2023] [Revised: 04/18/2023] [Accepted: 04/19/2023] [Indexed: 05/11/2023]
Abstract
BACKGROUND AND AIMS With distinct mechanisms of action, the combination of tropifexor (TXR) and cenicriviroc (CVC) may provide an effective treatment for NASH. This randomized, multicenter, double-blind, phase 2b study assessed the safety and efficacy of TXR and CVC combination, compared with respective monotherapies. APPROACH AND RESULTS Patients (N = 193) were randomized 1:1:1:1 to once-daily TXR 140 μg (TXR 140 ), CVC 150 mg (CVC), TXR 140 μg + CVC 150 mg (TXR 140 + CVC), or TXR 90 μg + CVC 150 mg (TXR 90 + CVC) for 48 weeks. The primary and secondary end points were safety and histological improvement, respectively. Rates of adverse events (AEs) were similar across treatment groups. Pruritus was the most frequently experienced AE, with highest incidence in the TXR 140 group (40.0%). In TXR and combination groups, alanine aminotransferase (ALT) decreased from baseline to 48 weeks (geometric mean change: -21%, TXR 140 ; -16%, TXR 140 + CVC; -13%, TXR 90 + CVC; and +17%, CVC). Reductions in body weight observed at week 24 (mean changes from baseline: TXR 140 , -2.5 kg; TXR 140 + CVC, -1.7 kg; TXR 90 + CVC, -1.0 kg; and CVC, -0.1 kg) were sustained to week 48. At least 1-point improvement in fibrosis stage/steatohepatitis resolution without worsening of fibrosis was observed in 32.3%/25.8%, 31.6%/15.8%, 29.7%/13.5%, and 32.5%/22.5% of patients in the TXR 140 , CVC, TXR 140 + CVC, and TXR 90 + CVC groups, respectively. CONCLUSIONS The safety profile of TXR + CVC combination was similar to respective monotherapies, with no new signals. TXR monotherapy showed sustained ALT and body weight decreases. No substantial incremental efficacy was observed with TXR + CVC combination on ALT, body weight, or in histological end points compared with monotherapy.
Collapse
Affiliation(s)
- Quentin M. Anstee
- Translational & Clinical Research Institute, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne, UK
| | - Kathryn J. Lucas
- Diabetes and Endocrinology Consultants, Morehead City, North Carolina, USA
| | - Sven Francque
- Department of Gastroenterology Hepatology, Antwerp University Hospital, Antwerp, Belgium
- InflaMed Centre of Excellence, Laboratory for Experimental Medicine and Paediatrics, Translational Sciences in Inflammation and Immunology, Faculty of Medicine and Health Sciences, University of Antwerp, Antwerp, Belgium
- European Reference Network on Hepatological Diseases (ERN RARE-LIVER)
| | | | - Arun J. Sanyal
- Virginia Commonwealth University, Richmond, Virginia, USA
| | - Vlad Ratziu
- Sorbonne Université, Hôpital Pitié Salpêtrière, ICAN Paris, France
| | | | - Mary Rinella
- University of Chicago, Pritzker School of Medicine, Chicago, Illinois, USA
| | | | - Rohit Loomba
- University of California at San Diego, La Jolla, California, USA
| | - Edward Mena
- California Liver Research Institute, Pasadena, California, USA
| | - Jörn M. Schattenberg
- Metabolic Liver Research Program, I. Department of Medicine, University Medical Center Mainz, Germany
| | | | - Donald Lazas
- Digestive Health Research and ObjectiveHealth, Nashville, Tennessee, USA
| | - George B.B. Goh
- Department of Gastroenterology and Hepatology, Singapore General Hospital, Singapore
| | - Shiv K. Sarin
- Department of Hepatology, Institute of Liver and Biliary Sciences, New Delhi, India
| | - Yusuf Yilmaz
- Department of Gastroenterology, School of Medicine, Marmara University, Istanbul, Turkey
- Department of Gastroenterology, School of Medicine, Recep Tayyip Erdoğan University, Rize, Turkey
| | | | | | | | - Li Chen
- Novartis Pharmaceuticals Corporation, East Hanover, New Jersey, USA
| | | | | | | | - Clifford Brass
- Novartis Pharmaceuticals Corporation, East Hanover, New Jersey, USA
| | | |
Collapse
|
78
|
Samaha C, Chaaban H, Simsek C, Danis N, Lin JS, Gurakar A. Practice patterns and considerations in liver transplantation from living donors with high BMI: A review. HEPATOLOGY FORUM 2023; 4:145-149. [PMID: 37822307 PMCID: PMC10564250 DOI: 10.14744/hf.2023.2023.0030] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 06/27/2023] [Revised: 09/03/2023] [Accepted: 09/04/2023] [Indexed: 10/13/2023]
Abstract
Living Donor Liver Transplantation (LDLT) is a valuable solution to the shortage of donor organs for patients with end-stage liver disease. However, the eligibility of obese donors for LDLT remains a subject of debate. This literature review explores global practices and perceptions of LDLT, identifies donor eligibility criteria, and discusses special considerations and ethical caveats. The review highlights the need for standardized guidelines for donor selection, considering the global distribution of Body mass index and variations in population-specific criteria. It also emphasizes the importance of non-invasive testing and pre-operative optimization of liver steatosis for select obese donors. Furthermore, the review examines the outcomes and complications associated with obese donors in LDLT. The findings of this review contribute to the ongoing discussion on the inclusion of obese donors in LDLT and provide insights for future research and guideline development.
Collapse
Affiliation(s)
- Carl Samaha
- Division of Gastroenterology and Hepatology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Hadi Chaaban
- Division of Gastroenterology and Hepatology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Cem Simsek
- Division of Gastroenterology and Hepatology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Nilay Danis
- Division of Gastroenterology and Hepatology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Jessica S. Lin
- Division of Gastroenterology and Hepatology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Ahmet Gurakar
- Division of Gastroenterology and Hepatology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| |
Collapse
|
79
|
Ye J, Lin Y, Shao C, Sun Y, Feng S, Zhong B. Comparisons of Insulin Resistance- and Steatosis-Based Scores in Monitoring Metabolic Associated Fatty Liver Disease Treatment Response. ANNALS OF NUTRITION & METABOLISM 2023; 79:448-459. [PMID: 37678173 DOI: 10.1159/000530531] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/25/2022] [Accepted: 10/28/2022] [Indexed: 09/09/2023]
Abstract
BACKGROUND Quantitative measurements of liver fat contents (LFCs) by magnetic resonance imaging derived-proton density fat fraction (MRI-PDFF) are accurate but limited by availability, convenience, and expense in the surveillance of metabolic associated fatty liver (MAFLD). Insulin resistance (IR) and steatosis-associated serum indices are useful in screening for MAFLD, but their value in monitoring MAFLD with or without chronic hepatitis B virus (CHB) infection remains unclear and we aimed to evaluate these scores in predicting changes in LFC. METHODS We conducted a prospective study between January 2015 and December 2021 with 620 consecutive participants with MAFLD (212 participants with CHB) who received a 24-week lifestyle intervention. The homeostasis model assessment of IR (HOMA-IR), HOMA2 index, glucose-insulin ratio, quantitative insulin sensitivity check index, fasting insulin resistance index, fatty liver index (FLI), hepatic steatosis index (HSI), liver fat score (LFS), visceral adiposity index, and triglycerides * glucose were calculated. RESULTS When using endpoints such as LFS improvements of ≥5% or 10% or escalations of ≥5%, LFS had the highest area under the curve (AUC) values at all endpoints for MAFLD alone (0.756, 95% CI: 0.707-0.805; 0.761, 95% CI: 0.705-0.818; 0.807, 95% CI: 0.713-0.901, all p < 0.05, respectively). With CHB, the FLI (AUC = 0.750) and HIS (AUC = 0.770) exhibited the highest AUCs between the former two outcomes, respectively, but no score could predict LFC escalation of ≥5%. CONCLUSION Among IR and steatosis scores, changes in LFC through lifestyle interventions can be captured with LFS possessing moderate precision but not in those with CHB.
Collapse
Affiliation(s)
- Junzhao Ye
- Department of Gastroenterology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Yansong Lin
- Department of Gastroenterology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Congxian Shao
- Department of Gastroenterology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Yanhong Sun
- Department of Clinical Laboratories, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Shiting Feng
- Department of Radiology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Bihui Zhong
- Department of Gastroenterology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| |
Collapse
|
80
|
Zhang X, Yu J, Wong VWS. Vive la resistant starch: A potential treatment for metabolic dysfunction-associated steatohepatitis. Cell Metab 2023; 35:1491-1493. [PMID: 37673032 DOI: 10.1016/j.cmet.2023.07.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/03/2023] [Revised: 07/11/2023] [Accepted: 07/11/2023] [Indexed: 09/08/2023]
Abstract
Metabolic dysfunction-associated steatotic liver disease is a prevalent chronic liver disease that affects nearly a third of the global adult population. Here, we preview a randomized controlled clinical trial on the potential benefits and underlying mechanisms of using resistant starch, a prebiotic, for treating this common liver condition.
Collapse
Affiliation(s)
- Xiang Zhang
- State Key Laboratory of Digestive Disease, Institute of Digestive Disease, The Chinese University of Hong Kong, Hong Kong, China
| | - Jun Yu
- State Key Laboratory of Digestive Disease, Institute of Digestive Disease, The Chinese University of Hong Kong, Hong Kong, China.
| | - Vincent Wai-Sun Wong
- State Key Laboratory of Digestive Disease, Institute of Digestive Disease, The Chinese University of Hong Kong, Hong Kong, China; Medical Data Analytics Centre, Department of Medicine and Therapeutics, The Chinese University of Hong Kong, Hong Kong, China.
| |
Collapse
|
81
|
Wang JH, Ou HY, Yen YH, Hung CH, Lu SN. Usefulness of controlled attenuation parameter in monitoring clinically relevant decline of hepatic steatosis for non-alcoholic fatty liver disease. Liver Int 2023; 43:1901-1908. [PMID: 37249034 DOI: 10.1111/liv.15626] [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: 02/03/2023] [Revised: 03/29/2023] [Accepted: 05/16/2023] [Indexed: 05/31/2023]
Abstract
BACKGROUND AND AIMS Magnetic resonance imaging-derived proton density fat fraction (MRI-PDFF) is the reference standard of hepatic steatosis assessment. This study evaluates usefulness of controlled attenuation parameter (CAP) in monitoring the clinically relevant outcome by MRI-PDFF for non-alcoholic fatty liver disease (NAFLD) patients. METHODS NAFLD patients were enrolled prospectively. Instruction was given in lifestyle modifications with exercise and control of metabolic factors. MRI-PDFF and CAP were performed at enrollment and follow-up, with the diagnostic validity of CAP in monitoring clinically relevant outcome defined as a decline of ≥30% relative to baseline value by MRI-PDFF. RESULTS A total of 75 patients (male/female: 49/26, mean age: 53.2) were enrolled. Baseline MRI-PDFF, CAP and liver stiffness was 14.4%, 300.2 dB/m and 6.5 kPa. In a median interval of 369 days, thirteen (17.3%) patients achieved clinically relevant outcome with decline of 46.7 dB/m by CAP, compared with increase of 5.1 in the other patients. In multivariate analysis, clinically relevant outcome was associated with changes (Δ) of CAP and glucose. Assessed by area under receiver operating curve, the performances of ΔCAP in predicting clinically relevant outcome were 0.815 and 0.808, and with the specificity of >90%, the ΔCAP cutoff was -46 dB/m and -15% relative to baseline value; sensitivity was 53.8% and 46.2% with negative predictive value of 90.3% and 88.9% respectively. CONCLUSIONS For NAFLD patients, CAP exhibited good performance in monitoring clinically relevant decline of hepatic steatosis in MRI-PDFF. With the cutoffs of -46 dB/m or -15%, ΔCAP is useful in excluding clinical relevant outcome achievement.
Collapse
Affiliation(s)
- Jing-Houng Wang
- Division of Hepato-Gastroenterology, Department of Internal Medicine, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung City, Taiwan
| | - Hsin-You Ou
- Department of Radiology, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung City, Taiwan
| | - Yi-Hao Yen
- Division of Hepato-Gastroenterology, Department of Internal Medicine, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung City, Taiwan
| | - Chao-Hung Hung
- Division of Hepato-Gastroenterology, Department of Internal Medicine, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung City, Taiwan
| | - Sheng-Nan Lu
- Division of Hepato-Gastroenterology, Department of Internal Medicine, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung City, Taiwan
| |
Collapse
|
82
|
Siddiqui MS, Parmar D, Shaikh F, Forsgren M, Patel S, Bui AT, Boyett S, Patel V, Sanyal AJ. Saroglitazar improves nonalcoholic fatty liver disease and metabolic health in liver transplant recipients. Liver Transpl 2023; 29:979-986. [PMID: 36847136 DOI: 10.1097/lvt.0000000000000110] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/02/2022] [Accepted: 01/23/2023] [Indexed: 03/01/2023]
Abstract
NAFLD is common after liver transplantation (LT) and is associated with an increased metabolic burden. Currently, there is a paucity of investigations into the treatment of post-LT NAFLD. In the present study, we evaluated the safety and efficacy of saroglitazar, a novel dual peroxisome proliferator-associated receptor α/γ agonist, on the treatment of post-LT NAFLD and metabolic burden. This is a phase 2A, single-center, open-label, single-arm study in which patients with post-LT NAFLD received saroglitazar magnesium 4 mg daily for 24 weeks. NAFLD was defined by a controlled attenuation parameter ≥264 dB/m. The primary endpoint was the reduction in liver fat as measured by MRI proton density fat fraction (MRI-PDFF). Secondary MRI-based metabolic endpoints included visceral adipose tissue, abdominal subcutaneous adipose tissue volumes, muscle fat infiltration, and fat-free muscle volume. Saroglitazar treatment led to a reduction in MRI-PDFF from 10.3±10.5% at baseline to 8.1±7.6%. A relative 30% reduction from baseline MRI-PDFF value was noted in 47% of all patients and 63% of patients with baseline MRI-PDFF >5%. Reduction in serum alkaline phosphatase was an independent predictor of MRI-PDFF response. Saroglitazar did not decrease fat-free muscle volume nor increase muscle fat infiltration, but did lead to a mild increase in visceral adipose tissue and abdominal subcutaneous adipose tissue. The study drug was well tolerated and a mild nonsignificant increase in serum creatinine was noted. Saroglitazar did not affect the weight. The study provides preliminary data demonstrating the safety and metabolic benefits of saroglitazar in LT recipients and underscores the importance of future studies to establish its efficacy after LT.
Collapse
Affiliation(s)
| | | | | | - Mikael Forsgren
- AMRA Medical AB, Centre for Medical Image Science and Visualization, Linköping University, Linköping, Sweden
- Department of Health, Medicine and Caring Sciences, Linköping University, Linköping, Sweden
| | - Samarth Patel
- Leigh Valley Health Network, Allentown, Pennsylvania, USA
| | - Anh Tuan Bui
- Department of Statistical Sciences and Operations Research, VCU, Richmond, Virginia, USA
| | - Sherry Boyett
- Division of Gastroenterology, Hepatology and Nutrition, VCU, Richmond, Virginia, USA
| | - Vaishali Patel
- Division of Gastroenterology, Hepatology and Nutrition, VCU, Richmond, Virginia, USA
| | - Arun J Sanyal
- Division of Gastroenterology, Hepatology and Nutrition, VCU, Richmond, Virginia, USA
| |
Collapse
|
83
|
Huang DQ, Sharpton SR, Amangurbanova M, Tamaki N, Sirlin CB, Loomba R. Clinical Utility of Combined MRI-PDFF and ALT Response in Predicting Histologic Response in Nonalcoholic Fatty Liver Disease. Clin Gastroenterol Hepatol 2023; 21:2682-2685.e4. [PMID: 36075503 DOI: 10.1016/j.cgh.2022.08.036] [Citation(s) in RCA: 26] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/16/2022] [Revised: 08/22/2022] [Accepted: 08/24/2022] [Indexed: 02/07/2023]
Affiliation(s)
- Daniel Q Huang
- NAFLD Research Center, Division of Gastroenterology, University of California San Diego, La Jolla, California; Department of Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore; Division of Gastroenterology and Hepatology, Department of Medicine, National University Health System, Singapore
| | - Suzanne R Sharpton
- NAFLD Research Center, Division of Gastroenterology, University of California San Diego, La Jolla, California
| | - Maral Amangurbanova
- NAFLD Research Center, Division of Gastroenterology, University of California San Diego, La Jolla, California
| | - Nobuharu Tamaki
- NAFLD Research Center, Division of Gastroenterology, University of California San Diego, La Jolla, California; Division of Gastroenterology and Hepatology, Musashino Red Cross Hospital, Musashino, Japan
| | - Claude B Sirlin
- Liver Imaging Group, Department of Radiology, University of California San Diego, La Jolla, California
| | - Rohit Loomba
- NAFLD Research Center, Division of Gastroenterology, University of California San Diego, La Jolla, California; Division of Epidemiology, Department of Family Medicine and Public Health, University of California San Diego, San Diego, California.
| |
Collapse
|
84
|
Stine JG, Long MT, Corey KE, Sallis RE, Allen AM, Armstrong MJ, Conroy DE, Cuthbertson DJ, Duarte-Rojo A, Hallsworth K, Hickman IJ, Kappus MR, Keating SE, Pugh CJA, Rotman Y, Simon TL, Vilar-Gomez E, Wai-Sun Wong V, Schmitz KH. Physical Activity and Nonalcoholic Fatty Liver Disease: A Roundtable Statement from the American College of Sports Medicine. Med Sci Sports Exerc 2023; 55:1717-1726. [PMID: 37126039 PMCID: PMC10524517 DOI: 10.1249/mss.0000000000003199] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/02/2023]
Abstract
ABSTRACT Although physical activity (PA) is crucial in the prevention and clinical management of nonalcoholic fatty liver disease, most individuals with this chronic disease are inactive and do not achieve recommended amounts of PA. There is a robust and consistent body of evidence highlighting the benefit of participating in regular PA, including a reduction in liver fat and improvement in body composition, cardiorespiratory fitness, vascular biology, and health-related quality of life. Importantly, the benefits of regular PA can be seen without clinically significant weight loss. At least 150 min of moderate or 75 min of vigorous intensity PA are recommended weekly for all patients with nonalcoholic fatty liver disease, including those with compensated cirrhosis. If a formal exercise training program is prescribed, aerobic exercise with the addition of resistance training is preferred. In this roundtable document, the benefits of PA are discussed, along with recommendations for 1) PA assessment and screening; 2) how best to advise, counsel, and prescribe regular PA; and 3) when to refer to an exercise specialist.
Collapse
Affiliation(s)
- Jonathan G. Stine
- Division of Gastroenterology and Hepatology, Department of Medicine, The Pennsylvania State University- Milton S. Hershey Medical Center, Hershey PA
- Department of Public Health Sciences, The Pennsylvania State University- College of Medicine, Hershey PA
| | - Michelle T. Long
- Section of Gastroenterology, Evans Department of Medicine, Boston University School of Medicine, Boston, MA
| | - Kathleen E. Corey
- Division of Gastroenterology and Hepatology, Department of Medicine, Massachusetts General Hospital, Boston, MA
| | - Robert E. Sallis
- Department of Family Medicine and Sports Medicine, Kaiser Permanente Medical Center, Fontana, CA
| | - Alina M. Allen
- Division of Gastroenterology and Hepatology, Department of Medicine, Mayo Clinic, Rochester, MN
| | - Matthew J. Armstrong
- Liver Transplant Unit, Queen Elizabeth University Hospitals Birmingham, and NIHR Birmingham Biomedical Research Centre, Birmingham, UNITED KINGDOM
| | - David E. Conroy
- Department of Kinesiology, The Pennsylvania State University, University Park, PA
| | - Daniel J. Cuthbertson
- Department of Cardiovascular and Metabolic Medicine, Institute of Life Course and Medical Sciences, University of Liverpool, Liverpool, UNITED KINGDOM
| | - Andres Duarte-Rojo
- Division of Gastroenterology and Hepatology, Department of Medicine, Northwestern University, Chicago, IL
| | - Kate Hallsworth
- Newcastle NIHR Biomedical Research Centre and the Liver Unit, Newcastle Upon Tyne Hospitals NHS Foundation Trust, Newcastle Upon Tyne, UNITED KINGDOM
| | - Ingrid J. Hickman
- Department of Nutrition and Dietetics, Princess Alexandra Hospital, Brisbane, Queensland, AUSTRALIA
| | - Matthew R. Kappus
- Division of Gastroenterology and Hepatology, Duke University, Durham, NC
| | - Shelley E. Keating
- School of Human Movement and Nutrition Sciences, The University of Queensland, St Lucia, Queensland, AUSTRALIA
| | - Christopher J. A. Pugh
- Cardiff School of Sport & Health Sciences, Cardiff Metropolitan University, Cardiff, UNITED KINGDOM
| | - Yaron Rotman
- Liver & Energy Metabolism Section, Liver Diseases Branch, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD
| | - Tracey L. Simon
- Division of Gastroenterology and Hepatology, Department of Medicine, Massachusetts General Hospital, Boston, MA
| | - Eduardo Vilar-Gomez
- Division of Gastroenterology and Hepatology. Indiana University School of Medicine. Indianapolis
| | - Vincent Wai-Sun Wong
- Department of Medicine and Therapeutics, The Chinese University of Hong Kong, CHINA
| | | |
Collapse
|
85
|
Tincopa MA, Loomba R. Non-invasive diagnosis and monitoring of non-alcoholic fatty liver disease and non-alcoholic steatohepatitis. Lancet Gastroenterol Hepatol 2023; 8:660-670. [PMID: 37060912 DOI: 10.1016/s2468-1253(23)00066-3] [Citation(s) in RCA: 75] [Impact Index Per Article: 37.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/12/2023] [Revised: 02/19/2023] [Accepted: 02/20/2023] [Indexed: 04/17/2023]
Abstract
Non-alcoholic fatty liver disease (NAFLD) is a highly prevalent form of chronic liver disease that poses challenges in diagnosis and risk stratification. Non-alcoholic steatohepatitis (NASH), the more progressive form of NAFLD, is particularly challenging to diagnose in the absence of histology. Liver biopsy is infrequently performed due to its invasive nature, potential for sampling error, and lack of inter-rater reliability. Non-invasive tests that can accurately identify patients with at-risk NASH (ie, individuals with biopsy-proven NASH with NAFLD activity score [NAS] ≥4 and fibrosis stage ≥2) are key tools to identify candidates for pharmacologic therapy in registrational trials for the treatment of NASH-related fibrosis. With emerging pharmacotherapy, non-invasive tests are required to track treatment response. Lastly, there is an unmet need for non-invasive tests to assess risk for clinical outcomes including progression to cirrhosis, hepatic decompensation, liver-related mortality, and overall mortality. In this Review we examine advances in non-invasive tests to diagnose and monitor NAFLD and NASH.
Collapse
Affiliation(s)
- Monica A Tincopa
- NAFLD Research Center, Division of Gastroenterology and Hepatology, University of California at San Diego, La Jolla, CA, USA
| | - Rohit Loomba
- NAFLD Research Center, Division of Gastroenterology and Hepatology, University of California at San Diego, La Jolla, CA, USA; School of Public Health, University of California at San Diego, La Jolla, CA, USA.
| |
Collapse
|
86
|
Koutoukidis DA, Mozes FE, Jebb SA, Tomlinson JW, Pavlides M, Saffioti F, Huntriss R, Aveyard P, Cobbold JF. A low-energy total diet replacement program demonstrates a favorable safety profile and improves liver disease severity in nonalcoholic steatohepatitis. Obesity (Silver Spring) 2023; 31:1767-1778. [PMID: 37368513 DOI: 10.1002/oby.23793] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/19/2023] [Revised: 03/13/2023] [Accepted: 04/03/2023] [Indexed: 06/29/2023]
Abstract
OBJECTIVE Low-energy diets are used to treat obesity and diabetes, but there are fears that they may worsen liver disease in patients with nonalcoholic steatohepatitis (NASH) and significant-to-advanced fibrosis. METHODS In this 24-week single-arm trial, 16 adults with NASH, fibrosis, and obesity received one-to-one remote dietetic support to follow a low-energy (880 kcal/d) total diet replacement program for 12 weeks and stepped food reintroduction for another 12 weeks. Liver disease severity was blindly evaluated (magnetic resonance imaging proton density fat fraction [MRI-PDFF], iron-corrected T1 [cT1], liver stiffness on magnetic resonance elastography [MRE], and liver stiffness on vibration-controlled transient elastography [VCTE]). Safety signals included liver biochemical markers and adverse events. RESULTS A total of 14 participants (87.5%) completed the intervention. Weight loss was 15% (95% CI: 11.2%-18.6%) at 24 weeks. Compared with baseline, MRI-PDFF reduced by 13.1% (95% CI: 8.9%-16.7%), cT1 by 159 milliseconds (95% CI: 108-216.5), MRE liver stiffness by 0.4 kPa (95% CI: 0.1-0.8), and VCTE liver stiffness by 3.9 kPa (95% CI: 2.6-7.2) at 24 weeks. The proportions with clinically relevant reductions in MRI-PDFF (≥30%), cT1 (≥88 milliseconds), MRE liver stiffness (≥19%), and VCTE liver stiffness (≥19%) were 93%, 77%, 57%, and 93%, respectively. Liver biochemical markers improved. There were no serious intervention-related adverse events. CONCLUSIONS The intervention demonstrates high adherence, favorable safety profile, and promising efficacy as a treatment for NASH.
Collapse
Affiliation(s)
- Dimitrios A Koutoukidis
- Nuffield Department of Primary Care Health Sciences, University of Oxford, Oxford, UK
- National Institute for Health and Care Research Oxford Biomedical Research Centre, Oxford, UK
| | - Ferenc E Mozes
- Oxford Centre for Clinical Magnetic Resonance Research, Radcliffe Department of Medicine, University of Oxford, Oxford, UK
| | - Susan A Jebb
- Nuffield Department of Primary Care Health Sciences, University of Oxford, Oxford, UK
- National Institute for Health and Care Research Oxford Biomedical Research Centre, Oxford, UK
| | - Jeremy W Tomlinson
- National Institute for Health and Care Research Oxford Biomedical Research Centre, Oxford, UK
- Oxford Centre for Diabetes, Endocrinology and Metabolism, Radcliffe Department of Medicine, University of Oxford, Oxford, UK
| | - Michael Pavlides
- Oxford Centre for Clinical Magnetic Resonance Research, Radcliffe Department of Medicine, University of Oxford, Oxford, UK
- Department of Gastroenterology and Hepatology, John Radcliffe Hospital, Oxford University Hospitals National Health Service Foundation Trust, Oxford, UK
| | - Francesca Saffioti
- Department of Gastroenterology and Hepatology, John Radcliffe Hospital, Oxford University Hospitals National Health Service Foundation Trust, Oxford, UK
| | | | - Paul Aveyard
- Nuffield Department of Primary Care Health Sciences, University of Oxford, Oxford, UK
- National Institute for Health and Care Research Oxford Biomedical Research Centre, Oxford, UK
| | - Jeremy F Cobbold
- National Institute for Health and Care Research Oxford Biomedical Research Centre, Oxford, UK
- Department of Gastroenterology and Hepatology, John Radcliffe Hospital, Oxford University Hospitals National Health Service Foundation Trust, Oxford, UK
| |
Collapse
|
87
|
Harrison SA, Loomba R, Dubourg J, Ratziu V, Noureddin M. Clinical Trial Landscape in NASH. Clin Gastroenterol Hepatol 2023; 21:2001-2014. [PMID: 37059159 DOI: 10.1016/j.cgh.2023.03.041] [Citation(s) in RCA: 62] [Impact Index Per Article: 31.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/14/2023] [Revised: 03/28/2023] [Accepted: 03/29/2023] [Indexed: 04/16/2023]
Abstract
Nonalcoholic fatty liver disease consists of a spectrum starting from nonalcoholic fatty liver disease that may progress to nonalcoholic steatohepatitis (NASH), which can lead to fibrosis, cirrhosis, hepatocellular carcinoma, or even liver failure. The prevalence of NASH has increased in parallel with the rising rate of obesity and type 2 diabetes. Given the high prevalence and deadly complications of NASH, there have been significant efforts to develop effective treatments. Phase 2A studies have assessed various mechanisms of action across the spectrum of the disease, while phase 3 studies have focused mainly on NASH and fibrosis stage 2 and higher, as these patients have a higher risk of disease morbidity and mortality. The primary efficacy endpoints also vary, by using noninvasive tests in early-phase trials while relying on liver histological endpoints in phase 3 studies as required by regulatory agencies. Despite initial disappointment due to the failure of several drugs, recent phase 2 and 3 studies have shown promising results, with the first Food and Drug Administration-approved drug for NASH expected to be approved in 2023. In this review, we discuss the various drugs under development for NASH, their mechanisms of action, and the results of their clinical trials. We also highlight the potential challenges in developing pharmacological therapies for NASH.
Collapse
Affiliation(s)
- Stephen A Harrison
- Radcliffe Department of Medicine, University of Oxford, Oxford, United Kingdom; Pinnacle Clinical Research, San Antonio, Texas.
| | - Rohit Loomba
- NAFLD Liver Center, Division of Gastroenterology, University of California San Diego, San Diego California
| | | | - Vlad Ratziu
- Institute for Cardiometabolism and Nutrition, Hospital Pitié-Salpêtrière, Sorbonne Université, Paris, France
| | - Mazen Noureddin
- Houston Research Institute, Houston Methodist Hospital, Houston, Texas
| |
Collapse
|
88
|
Stine JG, DiJoseph K, Pattison Z, Harrington A, Chinchilli VM, Schmitz KH, Loomba R. Exercise Training Is Associated With Treatment Response in Liver Fat Content by Magnetic Resonance Imaging Independent of Clinically Significant Body Weight Loss in Patients With Nonalcoholic Fatty Liver Disease: A Systematic Review and Meta-Analysis. Am J Gastroenterol 2023; 118:1204-1213. [PMID: 36705333 PMCID: PMC10287833 DOI: 10.14309/ajg.0000000000002098] [Citation(s) in RCA: 31] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/08/2022] [Accepted: 11/02/2022] [Indexed: 01/28/2023]
Abstract
INTRODUCTION Exercise training is crucial in the management of nonalcoholic fatty liver disease (NAFLD); however, whether it can achieve clinically meaningful improvement in liver fat is unclear. We investigated the association between exercise training and the achievement of validated thresholds of MRI-measured treatment response. METHODS Randomized controlled trials in adults with NAFLD were identified through March 2022. Exercise training was compared with no exercise training. The primary outcome was ≥30% relative reduction in MRI-measured liver fat (threshold required for histologic improvement in nonalcoholic steatohepatitis activity, nonalcoholic steatohepatitis resolution, and liver fibrosis stage). Different exercise doses were compared. RESULTS Fourteen studies (551 subjects) met inclusion criteria (mean age 53.3 yrs; body mass index 31.1 kg/m 2 ). Exercise training subjects were more likely to achieve ≥30% relative reduction in MRI-measured liver fat (odds ratio 3.51, 95% confidence interval 1.49-8.23, P = 0.004) than those in the control condition. An exercise dose of ≥750 metabolic equivalents of task min/wk (e.g., 150 min/wk of brisk walking) resulted in significant treatment response (MRI response odds ratio 3.73, 95% confidence interval 1.34-10.41, P = 0.010), but lesser doses of exercise did not. Treatment response was independent of clinically significant body weight loss (>5%). DISCUSSION Independent of weight loss, exercise training is 3 and a half times more likely to achieve clinically meaningful treatment response in MRI-measured liver fat compared with standard clinical care. An exercise dose of at least 750 metabolic equivalents of task-min/wk seems required to achieve treatment response. These results further support the weight-neutral benefit of exercise in all patients with NAFLD.
Collapse
Affiliation(s)
- Jonathan G. Stine
- Division of Gastroenterology and Hepatology, Department of Medicine, The Pennsylvania State University-Milton S. Hershey Medical Center, Hershey, Pennsylvania, USA
- Department of Public Health Sciences, The Pennsylvania State University-College of Medicine, Hershey, Pennsylvania, USA
- Liver Center, The Pennsylvania State University-Milton S. Hershey Medical Center, Hershey, Pennsylvania, USA
- Cancer Institute, The Pennsylvania State University-Milton S. Hershey Medical Center, Hershey, Pennsylvania, USA
| | - Kara DiJoseph
- Division of Gastroenterology and Hepatology, Department of Medicine, The Pennsylvania State University-Milton S. Hershey Medical Center, Hershey, Pennsylvania, USA
| | - Zach Pattison
- Division of Gastroenterology and Hepatology, Department of Medicine, The Pennsylvania State University-Milton S. Hershey Medical Center, Hershey, Pennsylvania, USA
| | - Alex Harrington
- College of Medicine, The Pennsylvania State University-Milton S. Hershey Medical Center, Hershey, Pennsylvania, USA
| | - Vernon M. Chinchilli
- Department of Public Health Sciences, The Pennsylvania State University-College of Medicine, Hershey, Pennsylvania, USA
| | - Kathryn H. Schmitz
- Department of Public Health Sciences, The Pennsylvania State University-College of Medicine, Hershey, Pennsylvania, USA
- Cancer Institute, The Pennsylvania State University-Milton S. Hershey Medical Center, Hershey, Pennsylvania, USA
- Department of Kinesiology, The Pennsylvania State University-College of Health and Human Development, University Park, Pennsylvania, USA
- Department of Physical Medicine & Rehabilitation, The Pennsylvania State University-Milton S. Hershey Medical Center, Hershey, Pennsylvania, USA
| | - Rohit Loomba
- Division of Gastroenterology and Hepatology, Department of Medicine, University of California San Diego, La Jolla, California, USA
- Division of Epidemiology, Department of Family Medicine and Public Health, University of California San Diego, La Jolla, California, USA
- NAFLD Research Center, University of California San Diego, La Jolla, California, USA
| |
Collapse
|
89
|
Zeng KY, Bao WYG, Wang YH, Liao M, Yang J, Huang JY, Lu Q. Non-invasive evaluation of liver steatosis with imaging modalities: New techniques and applications. World J Gastroenterol 2023; 29:2534-2550. [PMID: 37213404 PMCID: PMC10198053 DOI: 10.3748/wjg.v29.i17.2534] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/23/2022] [Revised: 02/26/2023] [Accepted: 04/11/2023] [Indexed: 05/23/2023] Open
Abstract
In the world, nonalcoholic fatty liver disease (NAFLD) accounts for majority of diffuse hepatic diseases. Notably, substantial liver fat accumulation can trigger and accelerate hepatic fibrosis, thus contributing to disease progression. Moreover, the presence of NAFLD not only puts adverse influences for liver but is also associated with an increased risk of type 2 diabetes and cardiovascular diseases. Therefore, early detection and quantified measurement of hepatic fat content are of great importance. Liver biopsy is currently the most accurate method for the evaluation of hepatic steatosis. However, liver biopsy has several limitations, namely, its invasiveness, sampling error, high cost and moderate intraobserver and interobserver reproducibility. Recently, various quantitative imaging techniques have been developed for the diagnosis and quantified measurement of hepatic fat content, including ultrasound- or magnetic resonance-based methods. These quantitative imaging techniques can provide objective continuous metrics associated with liver fat content and be recorded for comparison when patients receive check-ups to evaluate changes in liver fat content, which is useful for longitudinal follow-up. In this review, we introduce several imaging techniques and describe their diagnostic performance for the diagnosis and quantified measurement of hepatic fat content.
Collapse
Affiliation(s)
- Ke-Yu Zeng
- Department of Medical Ultrasound, West China Hospital of Sichuan University, Chengdu 610041, Sichuan Province, China
| | - Wu-Yong-Ga Bao
- Department of Medical Ultrasound, West China Hospital of Sichuan University, Chengdu 610041, Sichuan Province, China
| | - Yun-Han Wang
- Department of Medical Ultrasound, West China Hospital of Sichuan University, Chengdu 610041, Sichuan Province, China
| | - Min Liao
- Department of Medical Ultrasound, West China Hospital of Sichuan University, Chengdu 610041, Sichuan Province, China
| | - Jie Yang
- Department of Medical Ultrasound, West China Hospital of Sichuan University, Chengdu 610041, Sichuan Province, China
| | - Jia-Yan Huang
- Department of Medical Ultrasound, West China Hospital of Sichuan University, Chengdu 610041, Sichuan Province, China
| | - Qiang Lu
- Department of Medical Ultrasound, West China Hospital of Sichuan University, Chengdu 610041, Sichuan Province, China
| |
Collapse
|
90
|
Dunn W, Castera L, Loomba R. Roles of Radiological Tests in Clinical Trials and the Clinical Management of Nonalcoholic Fatty Liver Disease. Clin Liver Dis 2023; 27:363-372. [PMID: 37024213 PMCID: PMC10792514 DOI: 10.1016/j.cld.2023.01.020] [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: 04/08/2023]
Abstract
Radiological testing is now routinely used for clinical trial prescreening, diagnosis, and treatment and referral. The CAP performs well in detecting fatty liver but is unable to grade and track longitudinal changes. MRI-PDFF is a better technique for evaluating longitudinal changes and is used as a primary endpoint in trials of antisteatotic agents. The probability of detecting liver fibrosis using radiological testing techniques is high when performed at referral centers, and reasonable imaging strategies include the combination of FIB-4 and VCTE, the FAST Score, MAST, and MEFIB. The strategy currently recommended is the sequential application of FIB-4 and VCTE.
Collapse
Affiliation(s)
- Winston Dunn
- Division of Gastroenterology and Hepatology, Department of Medicine, University of Kansas Medical Center, 3901 Rainbow Boulevard, Kansas City, KS 66160, USA.
| | - Laurent Castera
- Université de Paris, UMR1149 (CRI), INSERM, Paris, France; Service d'Hépatologie, Assistance Publique-Hôpitaux de Paris (AP-HP), Hôpital Beaujon, Clichy, France
| | - Rohit Loomba
- Division of Gastroenterology and Hepatology, Department of Medicine, NAFLD Research Center, University of California San Diego, La Jolla, CA, USA
| |
Collapse
|
91
|
Rinella ME, Neuschwander-Tetri BA, Siddiqui MS, Abdelmalek MF, Caldwell S, Barb D, Kleiner DE, Loomba R. AASLD Practice Guidance on the clinical assessment and management of nonalcoholic fatty liver disease. Hepatology 2023; 77:1797-1835. [PMID: 36727674 PMCID: PMC10735173 DOI: 10.1097/hep.0000000000000323] [Citation(s) in RCA: 1028] [Impact Index Per Article: 514.0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/18/2023] [Accepted: 01/18/2023] [Indexed: 02/03/2023]
Affiliation(s)
- Mary E. Rinella
- University of Chicago Pritzker School of Medicine, Chicago, Illinois, USA
| | | | | | | | - Stephen Caldwell
- School of Medicine, University of Virginia, Charlottesville, Virginia, USA
| | - Diana Barb
- University of Florida College of Medicine, Gainesville, Florida, USA
| | | | - Rohit Loomba
- University of California, San Diego, San Diego, California, USA
| |
Collapse
|
92
|
Kim GA, Cho HC, Jeong SW, Kang BK, Kim M, Jung S, Hwang J, Yoon EL, Jun DW. A Phase 2a, Randomized, Double-Blind, Placebo-Controlled Study to Assess the Efficacy and Safety of ALS-L1023 in Non-Alcoholic Fatty Liver Disease. Pharmaceuticals (Basel) 2023; 16:ph16040623. [PMID: 37111380 PMCID: PMC10142612 DOI: 10.3390/ph16040623] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2023] [Revised: 04/15/2023] [Accepted: 04/17/2023] [Indexed: 04/29/2023] Open
Abstract
Preclinical data have shown that the herbal extract, ALS-L1023, from Melissa officinalis reduces visceral fat and hepatic steatosis. We aimed to assess the safety and efficacy of ALS-L1023 as the treatment of non-alcoholic fatty liver disease (NAFLD). We conducted a 24-week randomized, double-blind, placebo-controlled 2a study in patients with NAFLD (MRI-proton density fat fraction [MRI-PDFF] ≥ 8% and liver fibrosis ≥ 2.5 kPa on MR elastography [MRE]) in Korea. Patients were randomly assigned to 1800 mg ALS-L1023 (n = 19), 1200 mg ALS-L1023 (n = 21), or placebo (n = 17) groups. Efficacy endpoints included changes in liver fat on MRI-PDFF, liver stiffness on MRE, and liver enzymes. For the full analysis set, a relative hepatic fat reduction from baseline was significant in the 1800 mg ALS-L1023 group (-15.0%, p = 0.03). There was a significant reduction in liver stiffness from baseline in the 1200 mg ALS-L1023 group (-10.7%, p = 0.03). Serum alanine aminotransferase decreased by -12.4% in the 1800 mg ALS-L1023 group, -29.8% in the 1200 mg ALS-L1023 group, and -4.9% in the placebo group. ALS-L1023 was well tolerated and there were no differences in the incidence of adverse events among the study groups. ALS-L1023 could reduce hepatic fat content in patients with NAFLD.
Collapse
Affiliation(s)
- Gi-Ae Kim
- Department of Internal Medicine, Kyung Hee University College of Medicine, Kyung Hee University Hospital, Seoul 02447, Republic of Korea
| | - Hyun Chin Cho
- Department of Internal Medicine, Gyeongsang National University School of Medicine and Gyeongsang National University Hospital, Jinju 52727, Republic of Korea
| | - Soung Won Jeong
- Department of Internal Medicine, Soonchunhyang University College of Medicine, Soonchunhyang University Seoul Hospital, Seoul 04401, Republic of Korea
| | - Bo-Kyeong Kang
- Department of Radiology, Hanyang University College of Medicine, Seoul 04763, Republic of Korea
| | - Mimi Kim
- Department of Radiology, Hanyang University College of Medicine, Seoul 04763, Republic of Korea
| | - Seungwon Jung
- Graduate School for Biomedical Science & Engineering, Hanyang University, Seoul 04763, Republic of Korea
| | - Jungwook Hwang
- Graduate School for Biomedical Science & Engineering, Hanyang University, Seoul 04763, Republic of Korea
- Hanyang Institute of Bioscience and Biotechnology, Hanyang University, Seoul 04763, Republic of Korea
| | - Eileen L Yoon
- Hanyang Institute of Bioscience and Biotechnology, Hanyang University, Seoul 04763, Republic of Korea
- Department of Internal Medicine, Hanyang University College of Medicine, Seoul 04763, Republic of Korea
| | - Dae Won Jun
- Hanyang Institute of Bioscience and Biotechnology, Hanyang University, Seoul 04763, Republic of Korea
- Department of Internal Medicine, Hanyang University College of Medicine, Seoul 04763, Republic of Korea
| |
Collapse
|
93
|
Huang Q, An Z, Xin X, Sun Q, Gao S, Lv S, Xu X, Yang S, Lu F, Yuan J, Zhao Y, Hu Y, Liu P, Feng Q. Effectiveness and safety analysis of Danggui Shaoyao Powder for the treatment of non-alcoholic fatty liver disease: study protocol for a randomized, double-blind, placebo-controlled clinical trial. BMC Complement Med Ther 2023; 23:126. [PMID: 37076843 PMCID: PMC10114303 DOI: 10.1186/s12906-023-03948-3] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2023] [Accepted: 04/06/2023] [Indexed: 04/21/2023] Open
Abstract
BACKGROUND The incidence of non-alcoholic fatty liver disease (NAFLD) has been on the rise in recent years, and there are no effective drugs to treat NAFLD; therefore, effective prevention and treatment of NAFLD have become a new challenge. Danggui Shaoyao Powder (DGSY) is a classic prescription commonly used in clinical practice and has been shown to reduce hepatic steatosis in patients with NAFLD. In addition, previous studies have shown that DGSY can alleviate hepatic steatosis and inflammation in NAFLD mice. Although clinical practice and basic studies have shown that DGSY is effective in NAFLD, high levels of clinical evidence are lacking. Therefore, a standardized RCT study protocol is required to evaluate its clinical efficacy and safety. METHODS AND ANALYSIS This study will be a randomized, double-blind, placebo-controlled, and single-center trial. According to the random number table, NAFLD participants will be randomly divided into the DGSY or placebo group for 24 weeks. The follow-up period will be 6 weeks after drug withdrawal. The primary outcome is the relative change in MRI-proton density fat fraction (MRI-PDFF) from baseline to 24 weeks. Absolute changes in serum alanine aminotransferase (ALT), liver stiffness measurement (LSM), body mass index (BMI), blood lipid, blood glucose, and insulin resistance index will be selected as secondary outcomes to comprehensively evaluate the clinical efficacy of DGSY in the treatment of NAFLD. The safety of DGSY will be evaluated by renal function, routine blood and urine tests, and electrocardiogram. DISCUSSION This study will provide evidence-based medical corroboration for the clinical application of DGSY and promote the development and application of this classic prescription. TRIAL REGISTRATION http://www.chictr.org.cn . TRIAL NUMBER ChiCTR2000029144. Registered on 15 Jan 2020.
Collapse
Affiliation(s)
- Qian Huang
- Institute of Liver DiseasesShuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Shanghai Key Laboratory of Traditional Chinese Clinical Medicine, Shanghai, China
- Key Laboratory of Liver and Kidney Diseases, Shanghai University of Traditional Chinese Medicine, Ministry of Education, Shanghai, China
| | - Ziming An
- Institute of Liver DiseasesShuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Shanghai Key Laboratory of Traditional Chinese Clinical Medicine, Shanghai, China
- Key Laboratory of Liver and Kidney Diseases, Shanghai University of Traditional Chinese Medicine, Ministry of Education, Shanghai, China
| | - Xin Xin
- Key Laboratory of Liver and Kidney Diseases, Shanghai University of Traditional Chinese Medicine, Ministry of Education, Shanghai, China
| | - Qinmei Sun
- Institute of Liver DiseasesShuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Siting Gao
- Institute of Liver DiseasesShuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Sheng Lv
- Institute of Liver DiseasesShuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Xiao Xu
- Institute of Liver DiseasesShuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Shuohui Yang
- Department of Radiology, Shanghai Municipal Hospital of Traditional Chinese MedicineShanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Fang Lu
- Department of Radiology, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Jie Yuan
- Department of Radiology, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Yu Zhao
- Institute of Liver DiseasesShuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Yiyang Hu
- Institute of Liver DiseasesShuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Shanghai Key Laboratory of Traditional Chinese Clinical Medicine, Shanghai, China
- Key Laboratory of Liver and Kidney Diseases, Shanghai University of Traditional Chinese Medicine, Ministry of Education, Shanghai, China
| | - Ping Liu
- Institute of Liver DiseasesShuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Shanghai Key Laboratory of Traditional Chinese Clinical Medicine, Shanghai, China
- Key Laboratory of Liver and Kidney Diseases, Shanghai University of Traditional Chinese Medicine, Ministry of Education, Shanghai, China
| | - Qin Feng
- Institute of Liver DiseasesShuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China.
- Shanghai Key Laboratory of Traditional Chinese Clinical Medicine, Shanghai, China.
- Key Laboratory of Liver and Kidney Diseases, Shanghai University of Traditional Chinese Medicine, Ministry of Education, Shanghai, China.
| |
Collapse
|
94
|
Yoon JH. A Placebo Effect on MRI Proton Density Fat Fraction in Nonalcoholic Steatohepatitis Trials. Radiology 2023; 306:e222295. [PMID: 36318034 DOI: 10.1148/radiol.222295] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/22/2023]
Affiliation(s)
- Jeong Hee Yoon
- From the Department of Radiology, Seoul National University Hospital and Seoul National University College of Medicine, 101 Daehak-ro, Jongro-Gu, Seoul, 03080, South Korea
| |
Collapse
|
95
|
Hepatic and renal improvements with FXR agonist vonafexor in individuals with suspected fibrotic NASH. J Hepatol 2023; 78:479-492. [PMID: 36334688 DOI: 10.1016/j.jhep.2022.10.023] [Citation(s) in RCA: 45] [Impact Index Per Article: 22.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/22/2022] [Revised: 10/04/2022] [Accepted: 10/21/2022] [Indexed: 11/11/2022]
Abstract
BACKGROUND & AIMS The LIVIFY trial investigated the safety, tolerability, and efficacy of vonafexor, a second-generation, non-bile acid farnesoid X receptor agonist in patients with suspected fibrotic non-alcoholic steatohepatitis (NASH). METHODS This double-blind phase IIa study was conducted in two parts. Patients were randomised (1:1:1:1) to receive placebo, vonafexor 100 mg twice daily (VONA-100BID), vonafexor 200 mg once daily (VONA-200QD), or 400 mg vonafexor QD (VONA-400QD) in Part A (safety run-in, pharmacokinetics/pharmacodynamics) or placebo, vonafexor 100 mg QD (VONA-100QD), or VONA-200QD (1:1:1) in Part B. The primary efficacy endpoint was a reduction in liver fat content (LFC) by MRI-proton density fat fraction, while secondary endpoints included reduced corrected T1 values and liver enzymes, from baseline to Week 12. RESULTS One hundred and twenty patients were randomised (Part A, n = 24; Part B, n = 96). In Part B, there was a significant reduction in least-square mean (SE) absolute change in LFC from baseline to Week 12 for VONA-100QD (-6.3% [0.9]) and VONA-200QD (-5.4% [0.9]), vs. placebo (-2.3% [0.9], p = 0.002 and 0.012, respectively). A >30% relative LFC reduction was achieved by 50.0% and 39.3% of patients in the VONA-100QD and VONA-200QD arms, respectively, but only in 12.5% in the placebo arm. Reductions in body weight, liver enzymes, and corrected T1 were also observed with vonafexor. Creatinine-based glomerular filtration rate improved in the active arms but not the placebo arm. Mild to moderate generalised pruritus was reported in 6.3%, 9.7%, and 18.2% of participants in the placebo, VONA-100QD, and VONA-200QD arms, respectively. CONCLUSIONS In patients with suspected fibrotic NASH, vonafexor was safe and induced potent liver fat reduction, improvement in liver enzymes, weight loss, and a possible renal benefit. CLINICAL TRIAL NUMBER (EUDRACT) 2018-003119-22. CLINICALTRIALS GOV IDENTIFIER NCT03812029. IMPACT AND IMPLICATIONS Non-alcoholic steatohepatitis (NASH) has become a leading cause of chronic liver disease worldwide. Affected patients are also at higher risk of developing chronic kidney disease. There are no approved therapies and only few options to treat this population. The phase IIa LIVIFY trial results show that single daily administration of oral vonafexor, an FXR agonist, leads in the short term to a reduction in liver fat, liver enzymes, fibrosis biomarkers, body weight and abdominal circumference, and a possible improvement in kidney function, while possible mild moderate pruritus (a peripheral FXR class effect) and an LDL-cholesterol increase are manageable with lower doses and statins. These results support exploration in longer and larger trials, with the aim of addressing the unmet medical need in NASH.
Collapse
|
96
|
Nedrud MA, Chaudhry M, Middleton MS, Moylan CA, Lerebours R, Luo S, Farjat A, Guy C, Loomba R, Abdelmalek MF, Sirlin CB, Bashir MR. MRI Quantification of Placebo Effect in Nonalcoholic Steatohepatitis Clinical Trials. Radiology 2023; 306:e220743. [PMID: 36318027 PMCID: PMC9968769 DOI: 10.1148/radiol.220743] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2022] [Revised: 07/21/2022] [Accepted: 09/09/2022] [Indexed: 02/22/2023]
Abstract
Background Several early-phase clinical trials for the treatment of nonalcoholic steatohepatitis (NASH) use liver fat content as measured with the MRI-derived proton density fat fraction (PDFF) for a primary outcome. These trials have shown relative reductions in liver fat content with placebo treatment alone, a phenomenon termed "the placebo effect." This phenomenon confounds the results and limits generalizability to future trials. Purpose To quantify the effect of placebo treatment on change in the absolute PDFF value and to identify variables associated with this observed change. Materials and Methods This is a secondary analysis of prospectively collected data from seven early phase clinical trials that included participants with a diagnosis of NASH based on MRI and/or liver biopsy who received placebo treatment. The primary outcome was a greater than or equal to 30% relative reduction in PDFF after placebo treatment. Normalization of PDFF, relative change in alanine aminotransferase (ALT) level, and normalization of ALT level were also examined. An exploratory linear mixed-effects model was used to estimate an overall change in absolute PDFF and to explore parameters associated with this response. Results A total of 187 participants (median age, 52 years [IQR, 43-60 years]; 114 women) who received placebo treatment were evaluated. A greater than or equal to 30% relative reduction in baseline PDFF was seen in 20% of participants after 12 weeks of placebo treatment (10 of 49), 9% of participants after 16 weeks (two of 22), and 28% of participants after 24 weeks (34 of 122). A repeated-measures linear mixed-effects model estimated a decrease of 2.3 units (median relative reduction of 13%) in absolute PDFF values after 24 weeks of placebo treatment (95% CI: 3.2, 1.4; P < .001). Conclusion In this analysis of 187 participants, a clinically relevant decrease in PDFF was observed with placebo treatment. Based on the study model, assuming an absolute PDFF decrease of approximately 3 units (upper limit of 95% CI) to account for this "placebo effect" in sample size calculations for future clinical trials is suggested. Clinical trial registration nos. NCT01066364, NCT01766713, NCT01963845, NCT02443116, NCT02546609, NCT02316717, and NCT02442687 © RSNA, 2022 Online supplemental material is available for this article. See also the editorial by Yoon in this issue.
Collapse
Affiliation(s)
| | | | - Michael S. Middleton
- From the Department of Radiology (M.A.N., M.R.B.), Division of
Gastroenterology, Department of Medicine (C.A.M., M.R.B.), Department of
Biostatistics & Bioinformatics (R. Lerebours, S.L., A.F.), Department of
Pathology (C.G.), and Center for Advanced Magnetic Resonance Development
(M.R.B.), Duke University Medical Center, Department of Radiology, Box 3808,
Durham, NC 27710; Rutgers University Hospital, School of Medicine, Newark, NJ
(M.C.); Liver Imaging Group, Department of Radiology (M.S.M., C.B.S.), and
Division of Gastroenterology, Department of Medicine (R. Loomba), University of
California at San Diego School of Medicine, San Diego, Calif; Department of
Medicine, Durham Veterans Affairs Medical Center, Durham, NC (C.A.M.); and
Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, Minn
(M.F.A.)
| | - Cynthia A. Moylan
- From the Department of Radiology (M.A.N., M.R.B.), Division of
Gastroenterology, Department of Medicine (C.A.M., M.R.B.), Department of
Biostatistics & Bioinformatics (R. Lerebours, S.L., A.F.), Department of
Pathology (C.G.), and Center for Advanced Magnetic Resonance Development
(M.R.B.), Duke University Medical Center, Department of Radiology, Box 3808,
Durham, NC 27710; Rutgers University Hospital, School of Medicine, Newark, NJ
(M.C.); Liver Imaging Group, Department of Radiology (M.S.M., C.B.S.), and
Division of Gastroenterology, Department of Medicine (R. Loomba), University of
California at San Diego School of Medicine, San Diego, Calif; Department of
Medicine, Durham Veterans Affairs Medical Center, Durham, NC (C.A.M.); and
Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, Minn
(M.F.A.)
| | - Reginald Lerebours
- From the Department of Radiology (M.A.N., M.R.B.), Division of
Gastroenterology, Department of Medicine (C.A.M., M.R.B.), Department of
Biostatistics & Bioinformatics (R. Lerebours, S.L., A.F.), Department of
Pathology (C.G.), and Center for Advanced Magnetic Resonance Development
(M.R.B.), Duke University Medical Center, Department of Radiology, Box 3808,
Durham, NC 27710; Rutgers University Hospital, School of Medicine, Newark, NJ
(M.C.); Liver Imaging Group, Department of Radiology (M.S.M., C.B.S.), and
Division of Gastroenterology, Department of Medicine (R. Loomba), University of
California at San Diego School of Medicine, San Diego, Calif; Department of
Medicine, Durham Veterans Affairs Medical Center, Durham, NC (C.A.M.); and
Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, Minn
(M.F.A.)
| | - Sheng Luo
- From the Department of Radiology (M.A.N., M.R.B.), Division of
Gastroenterology, Department of Medicine (C.A.M., M.R.B.), Department of
Biostatistics & Bioinformatics (R. Lerebours, S.L., A.F.), Department of
Pathology (C.G.), and Center for Advanced Magnetic Resonance Development
(M.R.B.), Duke University Medical Center, Department of Radiology, Box 3808,
Durham, NC 27710; Rutgers University Hospital, School of Medicine, Newark, NJ
(M.C.); Liver Imaging Group, Department of Radiology (M.S.M., C.B.S.), and
Division of Gastroenterology, Department of Medicine (R. Loomba), University of
California at San Diego School of Medicine, San Diego, Calif; Department of
Medicine, Durham Veterans Affairs Medical Center, Durham, NC (C.A.M.); and
Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, Minn
(M.F.A.)
| | - Alfredo Farjat
- From the Department of Radiology (M.A.N., M.R.B.), Division of
Gastroenterology, Department of Medicine (C.A.M., M.R.B.), Department of
Biostatistics & Bioinformatics (R. Lerebours, S.L., A.F.), Department of
Pathology (C.G.), and Center for Advanced Magnetic Resonance Development
(M.R.B.), Duke University Medical Center, Department of Radiology, Box 3808,
Durham, NC 27710; Rutgers University Hospital, School of Medicine, Newark, NJ
(M.C.); Liver Imaging Group, Department of Radiology (M.S.M., C.B.S.), and
Division of Gastroenterology, Department of Medicine (R. Loomba), University of
California at San Diego School of Medicine, San Diego, Calif; Department of
Medicine, Durham Veterans Affairs Medical Center, Durham, NC (C.A.M.); and
Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, Minn
(M.F.A.)
| | - Cynthia Guy
- From the Department of Radiology (M.A.N., M.R.B.), Division of
Gastroenterology, Department of Medicine (C.A.M., M.R.B.), Department of
Biostatistics & Bioinformatics (R. Lerebours, S.L., A.F.), Department of
Pathology (C.G.), and Center for Advanced Magnetic Resonance Development
(M.R.B.), Duke University Medical Center, Department of Radiology, Box 3808,
Durham, NC 27710; Rutgers University Hospital, School of Medicine, Newark, NJ
(M.C.); Liver Imaging Group, Department of Radiology (M.S.M., C.B.S.), and
Division of Gastroenterology, Department of Medicine (R. Loomba), University of
California at San Diego School of Medicine, San Diego, Calif; Department of
Medicine, Durham Veterans Affairs Medical Center, Durham, NC (C.A.M.); and
Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, Minn
(M.F.A.)
| | - Rohit Loomba
- From the Department of Radiology (M.A.N., M.R.B.), Division of
Gastroenterology, Department of Medicine (C.A.M., M.R.B.), Department of
Biostatistics & Bioinformatics (R. Lerebours, S.L., A.F.), Department of
Pathology (C.G.), and Center for Advanced Magnetic Resonance Development
(M.R.B.), Duke University Medical Center, Department of Radiology, Box 3808,
Durham, NC 27710; Rutgers University Hospital, School of Medicine, Newark, NJ
(M.C.); Liver Imaging Group, Department of Radiology (M.S.M., C.B.S.), and
Division of Gastroenterology, Department of Medicine (R. Loomba), University of
California at San Diego School of Medicine, San Diego, Calif; Department of
Medicine, Durham Veterans Affairs Medical Center, Durham, NC (C.A.M.); and
Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, Minn
(M.F.A.)
| | - Manal F. Abdelmalek
- From the Department of Radiology (M.A.N., M.R.B.), Division of
Gastroenterology, Department of Medicine (C.A.M., M.R.B.), Department of
Biostatistics & Bioinformatics (R. Lerebours, S.L., A.F.), Department of
Pathology (C.G.), and Center for Advanced Magnetic Resonance Development
(M.R.B.), Duke University Medical Center, Department of Radiology, Box 3808,
Durham, NC 27710; Rutgers University Hospital, School of Medicine, Newark, NJ
(M.C.); Liver Imaging Group, Department of Radiology (M.S.M., C.B.S.), and
Division of Gastroenterology, Department of Medicine (R. Loomba), University of
California at San Diego School of Medicine, San Diego, Calif; Department of
Medicine, Durham Veterans Affairs Medical Center, Durham, NC (C.A.M.); and
Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, Minn
(M.F.A.)
| | - Claude B. Sirlin
- From the Department of Radiology (M.A.N., M.R.B.), Division of
Gastroenterology, Department of Medicine (C.A.M., M.R.B.), Department of
Biostatistics & Bioinformatics (R. Lerebours, S.L., A.F.), Department of
Pathology (C.G.), and Center for Advanced Magnetic Resonance Development
(M.R.B.), Duke University Medical Center, Department of Radiology, Box 3808,
Durham, NC 27710; Rutgers University Hospital, School of Medicine, Newark, NJ
(M.C.); Liver Imaging Group, Department of Radiology (M.S.M., C.B.S.), and
Division of Gastroenterology, Department of Medicine (R. Loomba), University of
California at San Diego School of Medicine, San Diego, Calif; Department of
Medicine, Durham Veterans Affairs Medical Center, Durham, NC (C.A.M.); and
Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, Minn
(M.F.A.)
| | - Mustafa R. Bashir
- From the Department of Radiology (M.A.N., M.R.B.), Division of
Gastroenterology, Department of Medicine (C.A.M., M.R.B.), Department of
Biostatistics & Bioinformatics (R. Lerebours, S.L., A.F.), Department of
Pathology (C.G.), and Center for Advanced Magnetic Resonance Development
(M.R.B.), Duke University Medical Center, Department of Radiology, Box 3808,
Durham, NC 27710; Rutgers University Hospital, School of Medicine, Newark, NJ
(M.C.); Liver Imaging Group, Department of Radiology (M.S.M., C.B.S.), and
Division of Gastroenterology, Department of Medicine (R. Loomba), University of
California at San Diego School of Medicine, San Diego, Calif; Department of
Medicine, Durham Veterans Affairs Medical Center, Durham, NC (C.A.M.); and
Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, Minn
(M.F.A.)
| |
Collapse
|
97
|
Harrison SA, Allen AM, Dubourg J, Noureddin M, Alkhouri N. Challenges and opportunities in NASH drug development. Nat Med 2023; 29:562-573. [PMID: 36894650 DOI: 10.1038/s41591-023-02242-6] [Citation(s) in RCA: 175] [Impact Index Per Article: 87.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2022] [Accepted: 12/20/2022] [Indexed: 03/11/2023]
Abstract
Nonalcoholic fatty liver disease (NAFLD) and its more severe form, nonalcoholic steatohepatitis (NASH), represent a growing worldwide epidemic and a high unmet medical need, as no licensed drugs have been approved thus far. Currently, histopathological assessment of liver biopsies is mandatory as a primary endpoint for conditional drug approval. This requirement represents one of the main challenges in the field, as there is substantial variability in this invasive histopathological assessment, which leads to dramatically high screen-failure rates in clinical trials. Over the past decades, several non-invasive tests have been developed to correlate with liver histology and, eventually, outcomes to assess disease severity and longitudinal changes non-invasively. However, further data are needed to ensure their endorsement by regulatory authorities as alternatives to histological endpoints in phase 3 trials. This Review describes the challenges of drug development in NAFLD-NASH trials and potential mitigating strategies to move the field forward.
Collapse
Affiliation(s)
| | - Alina M Allen
- Division of Gastroenterology and Hepatology, Mayo Clinic Rochester, Rochester, MN, USA
| | | | | | - Naim Alkhouri
- Department of Hepatology, Arizona Liver Health, Chandler, AZ, USA
| |
Collapse
|
98
|
Zhao J, Wang Q, Zhao X, Wu L, Li J, Zhang W, Xu S, Han C, Du Y, Tong X, Duan W, Cao D, Ren H, Zhao X, Ou X, Jia J, You H. Electro-acupuncture reduced steatosis on MRI-PDFF in patients with non-alcoholic steatohepatitis: a randomized controlled pilot clinical trial. Chin Med 2023; 18:19. [PMID: 36829229 PMCID: PMC9950708 DOI: 10.1186/s13020-023-00724-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2022] [Accepted: 02/07/2023] [Indexed: 02/26/2023] Open
Abstract
BACKGROUND Non-alcoholic steatohepatitis (NASH) had not yet been approved therapy. Electro-acupuncture (EA) has been reported to have potential efficacy. However, high-quality clinical evidence was still lacking. METHODS NASH patients were randomized and allocated to either sham acupuncture (SA) or EA group in a 1:1 ratio, with the patient blinded. Each patient received 36 sessions of SA or EA treatment over 12 weeks, followed by additional 4 weeks. The primary outcome was the changes in relative liver fat content measured by magnetic resonance imaging proton density fat fraction (MRI-PDFF). RESULTS A total of 60 patients were enrolled. From baseline to week 12, the reduction of relative liver fat content measured by MRI-PDFF in the EA group (- 33.6%, quantile range: - 52.9%, - 22.7%) was significantly more significant than that in the SA group (- 15.8%, quantile range: - 36.1%, - 2.7%) (p = 0.022). Furthermore, the EA group had more patients who achieved MRI-PDFF to 30% reduction at week 12 (53.3% vs. 25.9%, p = 0.035). EA treatment also significantly reduced body weight (- 3.0 vs. + 0.1 kg, p = 0.034) and BMI (- 1.5 vs. - 0.2 kg/m2, p = 0.013) at week 16. Except for AST (- 27.4 vs. - 16.2 U/L, p = 0.015), other biochemical varieties, including ALT, fasting-glucose, cholesterol, and triglyceride, showed no statistically significant difference. Both groups measured no significant changes in liver stiffness by magnetic resonance elastography (MRE). There were no serious adverse events in either group. CONCLUSIONS Twelve weeks of EA effectively and safely reduces relative liver fat content in NASH patients. Further multicenter randomized controlled studies are needed. Trial registration Chinese Clinical Trial Registry, ChiCTR2100046617. Registered 23 May 2021, http://www.chictr.org.cn/edit.aspx?pid=127023&htm=4.
Collapse
Affiliation(s)
- Jingjie Zhao
- grid.24696.3f0000 0004 0369 153XLiver Research Center, Beijing Friendship Hospital, Capital Medical University, National Clinical Research Center of Digestive Diseases, No. 95 Yong-an Road, Xi-Cheng District, Beijing, 100050 China ,grid.24696.3f0000 0004 0369 153XClinical Center for Metabolic Associated Fatty Liver Disease, Capital Medical University, Beijing, China ,grid.411610.30000 0004 1764 2878Department of Traditional Chinese Medicine, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Qianyi Wang
- grid.24696.3f0000 0004 0369 153XLiver Research Center, Beijing Friendship Hospital, Capital Medical University, National Clinical Research Center of Digestive Diseases, No. 95 Yong-an Road, Xi-Cheng District, Beijing, 100050 China ,grid.24696.3f0000 0004 0369 153XClinical Center for Metabolic Associated Fatty Liver Disease, Capital Medical University, Beijing, China
| | - Xinyu Zhao
- grid.24696.3f0000 0004 0369 153XClinical Center for Metabolic Associated Fatty Liver Disease, Capital Medical University, Beijing, China ,grid.411610.30000 0004 1764 2878Clinical Epidemiology and EBM Unit, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Lina Wu
- grid.24696.3f0000 0004 0369 153XLiver Research Center, Beijing Friendship Hospital, Capital Medical University, National Clinical Research Center of Digestive Diseases, No. 95 Yong-an Road, Xi-Cheng District, Beijing, 100050 China
| | - Juanjuan Li
- grid.411610.30000 0004 1764 2878Department of Traditional Chinese Medicine, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Wen Zhang
- grid.24696.3f0000 0004 0369 153XLiver Research Center, Beijing Friendship Hospital, Capital Medical University, National Clinical Research Center of Digestive Diseases, No. 95 Yong-an Road, Xi-Cheng District, Beijing, 100050 China
| | - Shuai Xu
- grid.411610.30000 0004 1764 2878Department of Traditional Chinese Medicine, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Chaoru Han
- grid.411610.30000 0004 1764 2878Department of Traditional Chinese Medicine, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Yi Du
- grid.411610.30000 0004 1764 2878Department of Traditional Chinese Medicine, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Xiaofei Tong
- grid.24696.3f0000 0004 0369 153XLiver Research Center, Beijing Friendship Hospital, Capital Medical University, National Clinical Research Center of Digestive Diseases, No. 95 Yong-an Road, Xi-Cheng District, Beijing, 100050 China ,grid.24696.3f0000 0004 0369 153XClinical Center for Metabolic Associated Fatty Liver Disease, Capital Medical University, Beijing, China
| | - Weijia Duan
- grid.24696.3f0000 0004 0369 153XLiver Research Center, Beijing Friendship Hospital, Capital Medical University, National Clinical Research Center of Digestive Diseases, No. 95 Yong-an Road, Xi-Cheng District, Beijing, 100050 China
| | - Di Cao
- grid.411610.30000 0004 1764 2878Department of Radiology, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Hao Ren
- grid.411610.30000 0004 1764 2878Department of Radiology, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Xinyan Zhao
- grid.24696.3f0000 0004 0369 153XLiver Research Center, Beijing Friendship Hospital, Capital Medical University, National Clinical Research Center of Digestive Diseases, No. 95 Yong-an Road, Xi-Cheng District, Beijing, 100050 China
| | - Xiaojuan Ou
- grid.24696.3f0000 0004 0369 153XLiver Research Center, Beijing Friendship Hospital, Capital Medical University, National Clinical Research Center of Digestive Diseases, No. 95 Yong-an Road, Xi-Cheng District, Beijing, 100050 China
| | - Jidong Jia
- grid.24696.3f0000 0004 0369 153XLiver Research Center, Beijing Friendship Hospital, Capital Medical University, National Clinical Research Center of Digestive Diseases, No. 95 Yong-an Road, Xi-Cheng District, Beijing, 100050 China
| | - Hong You
- Liver Research Center, Beijing Friendship Hospital, Capital Medical University, National Clinical Research Center of Digestive Diseases, No. 95 Yong-an Road, Xi-Cheng District, Beijing, 100050, China. .,Clinical Center for Metabolic Associated Fatty Liver Disease, Capital Medical University, Beijing, China.
| |
Collapse
|
99
|
Harrison SA, Thang C, Bolze S, Dewitt S, Hallakou-Bozec S, Dubourg J, Bedossa P, Cusi K, Ratziu V, Grouin JM, Moller DE, Fouqueray P. Evaluation of PXL065 - deuterium-stabilized (R)-pioglitazone in patients with NASH: A phase II randomized placebo-controlled trial (DESTINY-1). J Hepatol 2023; 78:914-925. [PMID: 36804402 DOI: 10.1016/j.jhep.2023.02.004] [Citation(s) in RCA: 39] [Impact Index Per Article: 19.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/24/2022] [Revised: 01/19/2023] [Accepted: 02/01/2023] [Indexed: 02/23/2023]
Abstract
BACKGROUND & AIMS Pioglitazone (Pio) is efficacious in NASH, but its utility is limited by PPARγ-driven side effects. Pio is a mixture of two enantiomers (R, S). PXL065, deuterium-stabilized R-Pio, lacks PPARγ activity but retains non-genomic activity. We tested the hypothesis that PXL065 would have similar efficacy but a better safety profile than Pio in patients with NASH. METHODS Patients (≥8% liver fat, NAFLD activity score [NAS] ≥4, F1-F3) received daily doses of PXL065 (7.5, 15, 22.5 mg) or placebo 1:1:1:1 for 36 weeks. The primary endpoint was relative % change in liver fat content (LFC) on MRI-proton density fat fraction; liver histology, non-invasive tests, safety-tolerability, and pharmacokinetics were also assessed. RESULTS One hundred and seventeen patients were evaluated. All PXL065 groups met the primary endpoint (-21 to (-25% LFC, p = 0.008-0.02 vs. placebo); 40% (22.5 mg) achieved a ≥30% LFC reduction. Favorable trends in non-invasive tests including reductions in PIIINP (p = 0.02, 22.5 mg) and NAFLD fibrosis score (p = 0.04, 22.5 mg) were observed. On histology (n = 92), a ≥1 stage fibrosis improvement occurred in 40% (7.5 mg), 50% (15 mg, p = 0.06), and 35% (22.5 mg) vs. 17% for placebo; up to 50% of PXL065-treated patients achieved a ≥2 point NAS improvement without fibrosis worsening vs. 30% with placebo. Metabolic improvements included: HbA1c (-0.41% p = 0.003) and insulin sensitivity (HOMA-IR, p = 0.04; Adipo-IR, p = 0.002). Adiponectin increased (+114%, 22.5 mg, p <0.0001) vs. placebo. There was no dose-dependent effect on body weight or PXL065-related peripheral oedema signal. Overall, PXL065 was safe and well tolerated. Pharmacokinetics confirmed dose-proportional and higher steady state R- vs. S-Pio exposure. IMPACT AND IMPLICATIONS Pioglitazone (Pio) is an approved diabetes medicine with proven efficacy in non-alcoholic steatohepatitis (NASH); PXL065 is a novel related oral agent which has been shown to retain Pio's efficacy in preclinical NASH models, with reduced potential for PPARγ-driven side effects. Results of this phase II study are important as PXL065 improved several key NASH disease features with a favorable safety profile - these findings can be applied by researchers seeking to understand pathophysiology and to develop new therapies. These results also indicate that PXL065 warrants further clinical testing in a pivotal NASH trial. Other implications include the potential future availability of a distinct oral therapy for NASH that may be relevant for patients, providers and caregivers seeking to prevent the progression and complications of this disease. CONCLUSIONS PXL065 is a novel molecule which retains an efficacy profile in NASH similar to Pio with reduced potential for PPARγ-driven side effects. A pivotal clinical trial is warranted to confirm the histological benefits reported herein. IMPACT AND IMPLICATIONS Pioglitazone (Pio) is an approved diabetes medicine with proven efficacy in non-alcoholic steatohepatitis (NASH); PXL065 is a novel related oral agent which has been shown to retain Pio's efficacy in preclinical NASH models, with reduced potential for PPARγ-driven side effects. Results of this phase II study are important as PXL065 improved several key NASH disease features with a favorable safety profile - these findings can be applied by researchers seeking to understand pathophysiology and to develop new therapies. These results also indicate that PXL065 warrants further clinical testing in a pivotal NASH trial. Other implications include the potential future availability of a distinct oral therapy for NASH that may be relevant for patients, providers and caregivers seeking to prevent the progression and complications of this disease.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Kenneth Cusi
- Division of Endocrinology, Diabetes and Metabolism, University of Florida, Gainesville, FL, USA
| | - Vlad Ratziu
- Sorbonne Université, ICAN, Hospital Pitié-Salpêtrière, INSERM UMRS 1138 CRC, Paris, France
| | | | | | | |
Collapse
|
100
|
Chee D, Ng CH, Chan KE, Huang DQ, Teng M, Muthiah M. The Past, Present, and Future of Noninvasive Test in Chronic Liver Diseases. Med Clin North Am 2023; 107:397-421. [PMID: 37001944 DOI: 10.1016/j.mcna.2022.12.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/22/2023]
Abstract
Chronic liver disease is a major global health threat and is the 11th leading cause of death globally. A liver biopsy is frequently required in assessing the degree of steatosis and fibrosis, information that is important in diagnosis, management, and prognostication. However, liver biopsies have limitations and carry a considerable risk, leading to the development of various modalities of noninvasive testing tools. These tools have been developed in recent years and have improved markedly in diagnostic accuracy. Moving forward, they may change the practice of hepatology.
Collapse
Affiliation(s)
- Douglas Chee
- Division of Gastroenterology and Hepatology, Department of Medicine, National University Hospital, Tower Block Level 10, 1E Kent Ridge Road, Singapore 119228, Singapore
| | - Cheng Han Ng
- Division of Gastroenterology and Hepatology, Department of Medicine, National University Hospital, Tower Block Level 10, 1E Kent Ridge Road, Singapore 119228, Singapore
| | - Kai En Chan
- Division of Gastroenterology and Hepatology, Department of Medicine, National University Hospital, Tower Block Level 10, 1E Kent Ridge Road, Singapore 119228, Singapore
| | - Daniel Q Huang
- Division of Gastroenterology and Hepatology, Department of Medicine, National University Hospital, Tower Block Level 10, 1E Kent Ridge Road, Singapore 119228, Singapore; Yong Loo Lin School of Medicine, National University of Singapore, Tower Block Level 10, 1E Kent Ridge Road, Singapore 119228, Singapore; National University Centre for Organ Transplantation, National University Health System, Tower Block Level 10, 1E Kent Ridge Road, Singapore 119228, Singapore
| | - Margaret Teng
- Division of Gastroenterology and Hepatology, Department of Medicine, National University Hospital, Tower Block Level 10, 1E Kent Ridge Road, Singapore 119228, Singapore; Yong Loo Lin School of Medicine, National University of Singapore, Tower Block Level 10, 1E Kent Ridge Road, Singapore 119228, Singapore; National University Centre for Organ Transplantation, National University Health System, Tower Block Level 10, 1E Kent Ridge Road, Singapore 119228, Singapore
| | - Mark Muthiah
- Division of Gastroenterology and Hepatology, Department of Medicine, National University Hospital, Tower Block Level 10, 1E Kent Ridge Road, Singapore 119228, Singapore; Yong Loo Lin School of Medicine, National University of Singapore, Tower Block Level 10, 1E Kent Ridge Road, Singapore 119228, Singapore; National University Centre for Organ Transplantation, National University Health System, Tower Block Level 10, 1E Kent Ridge Road, Singapore 119228, Singapore.
| |
Collapse
|