1
|
Panganiban J, Kehar M, Ibrahim SH, Hartmann P, Sood S, Hassan S, Ramirez CM, Kohli R, Censani M, Mauney E, Cuda S, Karjoo S. Metabolic dysfunction-associated steatotic liver disease (MASLD) in children with obesity: An Obesity Medicine Association (OMA) and expert joint perspective 2025. OBESITY PILLARS 2025; 14:100164. [PMID: 40230708 PMCID: PMC11995806 DOI: 10.1016/j.obpill.2025.100164] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/27/2024] [Revised: 01/28/2025] [Accepted: 01/29/2025] [Indexed: 04/16/2025]
Abstract
Introduction This Obesity Medicine Association (OMA) Expert Joint Perspective examines steatotic liver disease (SLD), which is composed of metabolic dysfunction-associated steatotic liver disease (MASLD), and metabolic dysfunction-associated steatohepatitis (MASH) in children with obesity. The prevalence of obesity is increasing, rates have tripled since 1963 from 5 % to now 19 % of US children affected in 2018. MASLD, is the most common liver disease seen in children, can be a precursor to the development of Type 2 Diabetes (T2DM) and is the primary reason for liver transplant listing in young adults. We must be vigilant in prevention and treatment of MASLD in childhood to prevent further progression. Methods This joint clinical perspective is based upon scientific evidence, peer and clinical expertise. The medical literature was reviewed via PubMed search and appropriate articles were included in this review. This work was formulated from the collaboration of eight hepatologists/gastroenterologists with MASLD expertise and two physicians from the OMA. Results The authors who are experts in the field, determined sentinel questions often asked by clinicians regarding MASLD in children with obesity. They created a consensus and clinical guideline for clinicians on the screening, diagnosis, and treatment of MASLD associated with obesity in children. Conclusions Obesity and the comorbidity of MASLD is increasing in children, and this is a medical problem that needs to be addressed urgently. It is well known that children with metabolic associated chronic disease often continue to have these chronic diseases as adults, which leads to reduced life expectancy, quality of life, and increasing healthcare needs and financial burden. The authors of this paper recommend healthy weight reduction not only through lifestyle modification but through obesity pharmacotherapy and bariatric surgery. Therefore, this guidance reviews available therapies to achieve healthy weight reduction and reverse MASLD to prevent progressive liver fibrosis, and metabolic disease.
Collapse
Affiliation(s)
| | - Mohit Kehar
- Division of Pediatric Gastroenterology, Hepatology and Nutrition, Children's Hospital of Eastern Ontario, Ottawa, Canada
| | - Samar H. Ibrahim
- Division of Pediatric Gastroenterology and Hepatology, Mayo Clinic, Rochester, MN, USA
| | - Phillipp Hartmann
- Department of Pediatrics, University of California San Diego, La Jolla, CA, USA
- Division of Gastroenterology, Hepatology & Nutrition, Rady Children’s Hospital San Diego, San Diego, CA, USA
| | - Shilpa Sood
- Division of Pediatric Gastroenterology, Boston Children's Health Physicians, New York Medical College, Valhalla, NY, USA
| | - Sara Hassan
- University of Texas Southwestern, Dallas, TX, United States
| | | | - Rohit Kohli
- Children's Hospital Los Angeles, CA, United States
| | - Marisa Censani
- Weill Cornell Medicine, New York Presbyterian Hospital, New York, NY, United States
| | - Erin Mauney
- Tufts Medical Center, Boston, MA, United States
| | - Suzanne Cuda
- Alamo City Healthy Kids and Families, San Antonio, TX, United States
| | - Sara Karjoo
- Johns Hopkins All Children's Hospital, St. Petersburg, FL, United States
- University of South Florida, Tampa, FL, United States
- Florida State University, Tallahassee, FL, United States
| |
Collapse
|
2
|
Piercy J, Fishman J, Higgins V, Pike J. Impact of Choice of Tariff When Calculating Clinically Meaningful EQ-5D Scores in Metabolic Dysfunction-Associated Steatohepatitis. Liver Int 2025; 45:e70043. [PMID: 40192123 PMCID: PMC11974243 DOI: 10.1111/liv.70043] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/08/2024] [Revised: 01/28/2025] [Accepted: 02/13/2025] [Indexed: 04/10/2025]
Abstract
BACKGROUND & AIMS Self-reported health varies across countries, as populations attribute different degrees of value to EQ-5D domains. Country-specific EQ-5D tariffs were developed to account for this but are not always stated in the literature. We aim to assess the reporting of EQ-5D in the literature and the impact of applying country-specific tariffs. METHODS We reviewed literature-reported EQ-5D utilities for patients with metabolic dysfunction-associated steatohepatitis versus real-world EQ-5D utilities from the Adelphi Real World Non-alcoholic steatohepatitis Disease Specific Programme (DSP), a cross-sectional survey in France, Germany, Italy, Spain, the United Kingdom and the United States. Matching-adjusted indirect comparison analysis balanced DSP data with literature studies by age, sex, comorbidities and fibrosis stage. DSP utility scores generated using national tariffs were compared with literature utilities using weighted t tests. RESULTS Ten studies with varying recruitment criteria, patient demographics and clinical characteristics were identified. Country-specific tariffs were not used or not reported. EQ-5D utilities varied, reflecting geographic, clinical and demographic characteristics. The comparison of literature and matched utilities derived using DSP data and five national tariffs revealed ≥ two comparisons for each study with a difference not exceeding the minimal clinically important difference versus the matched DSP value. CONCLUSIONS Literature-reported EQ-5D utilities vary considerably depending on study methodology and country-specific EQ-5D tariff, and even if stated may not always use the most appropriate tariff. This suggests a need for consistent use of country-specific tariffs and sensitivity analyses confirming results and conclusions that include EQ-5D-based utility measurement to inform decision-making by health authorities.
Collapse
|
3
|
Davidson JA, Brewer HR, Rice CT, Carvalho SJ, Kim Y. Estimating the clinical and healthcare burden of metabolic dysfunction-associated steatohepatitis in England: a retrospective cohort study using routinely collected healthcare data from 2011 to 2020. BMJ Open 2025; 15:e095761. [PMID: 40268491 PMCID: PMC12020757 DOI: 10.1136/bmjopen-2024-095761] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/28/2024] [Accepted: 03/31/2025] [Indexed: 04/25/2025] Open
Abstract
OBJECTIVE To characterise patients with metabolic dysfunction-associated steatohepatitis (MASH) in England and to estimate its associated healthcare resource use (HCRU) and costs, both overall and by progression status and comorbidities. DESIGN This was a retrospective observational study of adults with a MASH-coded primary and/or secondary care recorded diagnosis in England (2011-2020). The analysis used data from the Clinical Practice Research Datalink linked to the Hospital Episode Statistics and death registrations. Annualised all-cause and MASH-related (ie, coded as MASH, end-stage liver disease or major adverse cardiovascular event) HCRU and costs were calculated for patients with incident MASH. Subgroup analyses were conducted for patients with type 2 diabetes, overweight/obesity, cardiovascular disease or progression to cirrhosis. Comparative cost analysis was conducted between those with progressed MASH and those who did not progress. RESULTS A total of 2696 patients were included (mean follow-up: 4 years). Incidence of MASH was estimated at 4.7 per 100 000 person-years overall and increased among patients with key comorbidities. Patients who had type 2 diabetes had greater HCRU and costs than those who did not (eg, mean 1.8 vs 1.0 all-cause inpatient admissions and £2227 vs £1151 all-cause inpatient costs per-patient per-year). Some patients with MASH progressed to compensated (8.6%) or decompensated cirrhosis (6.5%) during the study. HCRU and costs were substantially higher among patients who progressed than among those who did not (eg, mean 2.4 vs 1.1 all-cause inpatient admissions and £3620 vs £1290 all-cause inpatient costs per-patient per-year). CONCLUSION HCRU and costs associated with MASH are higher among patients who have cardiometabolic comorbidities or who progress to advanced disease stages. Therefore, efforts to detect cases early and prevent disease progression could reduce healthcare burden.
Collapse
Affiliation(s)
| | | | | | | | - Yestle Kim
- Madrigal Pharmaceuticals Inc, West Conshohocken, Pennsylvania, USA
| |
Collapse
|
4
|
Abdel Monem MS, Adel A, Abbassi MM, Abdelaziz DH, Hassany M, Raziky ME, Sabry NA. Efficacy and safety of dapagliflozin compared to pioglitazone in diabetic and non-diabetic patients with non-alcoholic steatohepatitis: A randomized clinical trial. Clin Res Hepatol Gastroenterol 2025; 49:102543. [PMID: 39884573 DOI: 10.1016/j.clinre.2025.102543] [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: 12/08/2024] [Revised: 01/21/2025] [Accepted: 01/26/2025] [Indexed: 02/01/2025]
Abstract
BACKGROUND Non-alcoholic steatohepatitis (NASH) is a serious end-stage spectrum of non-alcoholic fatty liver disease (NAFLD) with associated high risk of hepatic and extrahepatic complications. Several studies showed the significant beneficial effect of dapagliflozin on body composition, hepatic and metabolic parameters on NAFLD/NASH patients. The study aimed to investigate the efficacy and safety of dapagliflozin in both diabetic and non-diabetic biopsy-proven NASH patients; compared to pioglitazone. METHODS This was a four-group, prospective, randomized, parallel, open label study in which 100 biopsy-proven NASH patients were selected, stratified to diabetics and non-diabetics and randomized with 1:1 allocation to either 30 mg pioglitazone or 10 mg dapagliflozin, once daily for 24 weeks. Histological evaluation, anthropometric measures, hepatic, metabolic biochemical markers, fibrosis non-invasive markers, quality of life (QOL) and medications adverse events were examined. RESULTS Dapagliflozin showed a comparable histological effect to pioglitazone in both diabetic and non-diabetic patients (P>0.05). As assessed by transient elastography, it also showed a comparable effect on liver fibrosis grade improvement from baseline in diabetics (P=0.287) versus a significant superiority in non-diabetics (P=0.018). Dapagliflozin showed a significant superiority in all anthropometric measures (P<0.001) and QOL (P<0.05) among both diabetics and non-diabetics. There was a significant interaction between interventions and diabetes status on change from baseline of hepatic and metabolic panel collectively (P=0.023) in favor to dapagliflozin among diabetics. CONCLUSION Compared to pioglitazone, dapagliflozin had a comparable effect histologically, superior effect biochemically among diabetics and superior effect on liver fibrosis, steatosis and insulin resistance among non-diabetics. TRIAL REGISTRATION The study was registered on clinicaltrials.gov, identifier number NCT05254626.
Collapse
Affiliation(s)
- Mona S Abdel Monem
- Department of Clinical Pharmacy, Faculty of Pharmacy, Cairo University, Egypt.
| | - Abdulmoneim Adel
- National Hepatology and Tropical Medicine Research Institute, Cairo, Egypt.
| | - Maggie M Abbassi
- Department of Clinical Pharmacy, Faculty of Pharmacy, Cairo University, Egypt.
| | - Doaa H Abdelaziz
- Department of Clinical Pharmacy, Faculty of Pharmacy, Al-Baha University, Al-Baha, Saudi Arabia/Department of Clinical Pharmacy, National Hepatology and Tropical Medicine Research Institute, Cairo, Egypt.
| | - Mohamed Hassany
- National Hepatology and Tropical Medicine Research Institute, Cairo, Egypt.
| | - Maissa El Raziky
- Endemic Medicine and Hepatogastroenterology, Faculty of Medicine, Cairo University, Egypt.
| | - Nirmeen A Sabry
- Department of Clinical Pharmacy, Faculty of Pharmacy, Cairo University, Egypt.
| |
Collapse
|
5
|
Hughes JH, Amin NB, Wojciechowski J, Vourvahis M. Exposure-response modeling of liver fat imaging endpoints in non-alcoholic fatty liver disease populations administered ervogastat alone and co-administered with clesacostat. CPT Pharmacometrics Syst Pharmacol 2025; 14:317-330. [PMID: 39564924 PMCID: PMC11812935 DOI: 10.1002/psp4.13275] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2024] [Revised: 10/21/2024] [Accepted: 10/25/2024] [Indexed: 11/21/2024] Open
Abstract
Non-alcoholic fatty liver disease and non-alcoholic steatohepatitis describe a collection of liver conditions characterized by the accumulation of liver fat. Despite biopsy being the reference standard for determining the severity of disease, non-invasive measures such as magnetic resonance imaging proton density fat fraction (MRI-PDFF) and FibroScan® controlled attenuation parameter (CAP™) can be used to understand longitudinal changes in steatosis. The aim of this work was to describe the exposure-response relationship of ervogastat with or without clesacostat on steatosis, through population pharmacokinetic/pharmacodynamic (PK/PD) modeling of both liver fat measurements simultaneously. Population pharmacokinetic and exposure-response models using individual predictions of average concentrations were used to describe ervogastat/clesacostat PKPD. Due to both liver fat endpoints being continuous-bounded outcomes on different scales, a dynamic transform-both-sides approach was used to link a common latent factor representing liver fat to each endpoint. Simultaneous modeling of both MRI-PDFF and CAP™ was successful with both measurements being adequately described by the model. The clinical trial simulation was able to adequately predict the results of a recent Phase 2 study, where subjects given ervogastat/clesacostat 300/10 mg BID for 6 weeks had a LS means and model-predicted median (95% confidence intervals) percent change from baseline MRI-PDFF of -45.8% and -45.6% (-61.6% to -31.8%), respectively. Simultaneous modeling of both MRI-PDFF and CAP™ was successful with both measurements being adequately described. By describing the underlying changes of steatosis with a latent variable, this model may be extended to describe biopsy results from future studies.
Collapse
Affiliation(s)
| | - Neeta B. Amin
- Pfizer Research and DevelopmentCambridgeMassachusettsUSA
| | | | | |
Collapse
|
6
|
Lu C, Ke L, Mentis AFA, Zhang Q, Wang Z, Wang Z. Tea intake and non-alcoholic fatty liver disease risk: A two-sample Mendelian randomization study. Metabol Open 2024; 24:100322. [PMID: 39399721 PMCID: PMC11470174 DOI: 10.1016/j.metop.2024.100322] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2024] [Revised: 09/23/2024] [Accepted: 09/24/2024] [Indexed: 10/15/2024] Open
Abstract
Background Non-alcoholic fatty liver disease (NAFLD) is a major global health problem due to its great disease and economic burdens. Tea is a popular beverage consumed by billions of people.globally owing to its health benefits. However, the evidence regarding the association between tea intake and NAFLD risk is inconsistent. Objective To examine the genetically predicted causal association between tea intake and NAFLD risk using the two-sample Mendelian randomization (MR) method. Methods Single-nucleotide polymorphisms (SNPs) strongly associated with tea intake were obtained from a large dataset (N = 447,485) in the UK biobank, and summary-level genetic data for NAFLD (2,275 cases and 375,002 controls) were collected from the FinnGen consortium. The two-sample MR method was used to investigate the causal association between tea intake and NAFLD risk. The random-effects inverse-variance weighted (IVW) was used as the primary approach for estimating the causal effect, and MR Egger, weighted median, simple mode, and weighted mode were used to verify the robustness of the primary results. Results Twenty-four valid SNPs were selected as the instrumental variables for tea intake. The IVW results indicated that tea intake was not causally associated with NAFLD risk (Odds ratio: 1.48; 95 % confidence interval: 0.64, 3.43; p = 0.364); moreover, the results from other methods were consistent with this finding. A leave-one-out analysis further demonstrated the robustness of our results. No evidence of heterogeneity, outliers, or horizontal pleiotropy was found. Conclusion Our results do not support tea intake being causally associated with a decreased risk of NAFLD.
Collapse
Affiliation(s)
- Cuncun Lu
- Institute of Basic Research in Clinical Medicine, China Academy of Chinese Medical Sciences, Beijing, 100700, China
| | - Lixin Ke
- Department of Pediatrics, University Medical Center Groningen, University of Groningen, Groningen, 9713GZ, Netherlands
| | | | - Qiang Zhang
- Department of Gastroenterology, The First Affiliated Hospital of Henan University of CM, Zhengzhou, 450000, China
| | - Ziyi Wang
- Evidence-Based Social Science Research Center, School of Public Health, Lanzhou University, Lanzhou, 730000, China
| | - Zhifei Wang
- Institute of Basic Research in Clinical Medicine, China Academy of Chinese Medical Sciences, Beijing, 100700, China
| |
Collapse
|
7
|
Kim Y, Medicis J, Davis M, Nunag D, Gish R. Costs associated with nonalcoholic steatohepatitis disease progression in Medicare patients: a retrospective cohort study. J Comp Eff Res 2024; 13:e240096. [PMID: 39576038 DOI: 10.57264/cer-2024-0096] [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: 11/29/2024] Open
Abstract
Aim: Non-alcoholic steatohepatitis (NASH), or metabolic dysfunction-associated steatohepatitis (MASH), is a severe form of non-alcoholic fatty liver disease (NAFLD) or metabolic dysfunction-associated liver disease (MASLD), that may progress to advanced liver disease. Costs associated with progression are not well characterized. This study sought to quantify costs and healthcare resource utilization (HRU) associated with NASH progression. Methods: Patients were included if diagnosed with NASH (ICD-10: K75.81) in 100% Medicare claims data (2015-2021) who were ≥66 years at index (diagnosis), continuously enrolled in Parts A, B and D for ≥12 months prior to and 6 months following index (unless death) and who had no evidence of other causes of liver disease. Patient-time was categorized into five severity states: non-cirrhotic NASH, compensated cirrhosis (CC), decompensated cirrhosis (DCC), hepatocellular carcinoma (HCC) and liver transplant (LT). Annualized HRU and costs were calculated during the study periods overall and stratified by occurrence and timing of progression. Results: In 14,806 unique patients (n = 12,990 non-cirrhotic NASH; 1899 CC; 997 DCC; 209 HCC; 140 LT), mean age and follow-up were 72.2 and 2.8 years, respectively. Average annualized costs increased from baseline following diagnosis, generally scaling with severity: $16,231 to $27,044; $25,122 to $57,705; $40,613 to $181,036; $36,549 to $165,121 and $35,626 to $108,918 in NASH; CC; DCC; HCC; and LT; respectively. Non-cirrhotic NASH and CC patients with progression had higher follow-up spending (1.6x for NASH; 1.7x for CC) than non-progressors (both p < 0.001), 2.8 and 6.1-times higher odds of an inpatient stay and 2.6 and 3.6-times higher odds to be in the top 20% of spenders, respectively, relative to non-progressors (both p < 0.001). Patients progressing within a year had costs 1.4, 1.6, 1.7 and 2.2-times more than year 2, 3, 4 and 5 progressors' costs, respectively, for non-cirrhotic NASH and 1.3, 1.8, 2.0 and 2.2-times more than year 2, 3, 4 and 5 progressors' costs, respectively, for CC. Conclusion: NASH progression is associated with high costs that increase in more severe disease states. Slower progression is associated with lower costs, suggesting a potential benefit of therapies that may delay or prevent progression.
Collapse
Affiliation(s)
- Yestle Kim
- Madrigal Pharmaceuticals, Inc, West Conshohocken, PA, USA
| | - Joseph Medicis
- Madrigal Pharmaceuticals, Inc, West Conshohocken, PA, USA
| | | | | | - Robert Gish
- Loma Linda University, School of Medicine, Loma Linda, CA, USA
| |
Collapse
|
8
|
Fishman J, Alexander T, Kim Y, Kindt I, Mendez P. A clinical decision support tool for metabolic dysfunction-associated steatohepatitis in real-world clinical settings: a mixed-method implementation research study protocol. J Comp Eff Res 2024; 13:e240085. [PMID: 39301878 PMCID: PMC11426282 DOI: 10.57264/cer-2024-0085] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2024] [Accepted: 08/13/2024] [Indexed: 09/22/2024] Open
Abstract
Aim: A clinical decision support (CDS) tool for metabolic dysfunction-associated steatohepatitis (MASH) was developed to align health systems with clinical guidelines detailed in the MASH Clinical Care Pathway and improve patients' proactive self-management of their disease. The tool includes a provider-facing web-based application and a mobile application (app) for patients. This protocol outlines a pilot study that will systematically evaluate the implementation of the tool in real-world clinical practice settings. Materials & methods: This implementation research study will use a simultaneous mixed-methods design and is guided by the Consolidated Framework for Implementation Research. The CDS tool for MASH will be piloted for ≥3 months at multiple US-based sites with eligible gastroenterologists and hepatologists (n = 5-10 per site) and their patients (n = 50-100 per site) with MASH or suspected MASH. Each pilot site may choose one or all focus areas within the tool (i.e., risk stratification, screening and referral, or patient care management), based on on-site capabilities. Prior to and at the end of the pilot period, providers and patients will complete quantitative surveys and partake in semi-structured interviews. Outcomes will include understanding the feasibility of implementing the tool in real-world clinical settings, its effectiveness in increasing patient screenings and risk stratification for MASH, its ability to improve provider and patient knowledge of MASH, barriers to adoption of the tool and the tool's capacity to enhance patient engagement and satisfaction with their care. Conclusion: Findings will inform the scalable implementation of the tool to ensure patients at risk for MASH are identified early, referred to specialty care when necessary and managed appropriately. Successful integration of the patient app can contribute to better health outcomes for patients by facilitating their active participation in the management of their condition.
Collapse
Affiliation(s)
- Jesse Fishman
- Madrigal Pharmaceuticals, Inc., West Conshohocken, PA 19428, USA
| | | | - Yestle Kim
- Madrigal Pharmaceuticals, Inc., West Conshohocken, PA 19428, USA
| | - Iris Kindt
- DEARhealth, Westlake Village, CA 91362, USA
| | - Patricia Mendez
- Madrigal Pharmaceuticals, Inc., West Conshohocken, PA 19428, USA
| |
Collapse
|
9
|
Kaplan A, Winters A, Klarman S, Kriss M, Hughes D, Sharma P, Asrani S, Hutchison A, Myoung P, Zaman A, Butler L, Pomposelli J, Gordon F, Duarte-Rojo A, Devuni D, Fortune B. The rising cost of liver transplantation in the United States. Liver Transpl 2024:01445473-990000000-00471. [PMID: 39315930 DOI: 10.1097/lvt.0000000000000493] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/21/2024] [Accepted: 09/04/2024] [Indexed: 09/25/2024]
Abstract
Liver transplantation (LT) is the only curative treatment for end-stage liver disease and significantly improves patient outcomes. However, LT is resource-intensive and costly, with expenditures rising dramatically in recent years. Factors contributing to this increase in cost include expanded transplant criteria, utilization of marginal organs, and broader organ distribution, resulting in significant logistical expenses. Advanced technologies like organ perfusion devices, while promising better outcomes, further inflate costs due to their high price and market monopolization. Moreover, living donor liver transplant and utilization of donation after cardiac death organs introduce higher initial expenditures yet potential long-term savings. Despite rising costs, reimbursement has remained largely stagnant, putting financial strain on transplant programs, and threatening their sustainability. This review examines the multifaceted drivers of rising costs in LT, focusing on recent policy changes, the role of organ procurement organizations, and the impact of new technologies. We also propose comprehensive solutions at national, organ procurement organization, and local levels, including optimizing resource allocation, leveraging regional collaborations, and advocating for revised reimbursement models to curb escalating costs. Addressing these challenges is critical to ensuring the continued viability of LT programs and maintaining patient access to this life-saving intervention.
Collapse
Affiliation(s)
- Alyson Kaplan
- Department of Surgery, Transplant Institute, Tufts Medical Center, Boston, Massachusetts, USA
| | - Adam Winters
- Recanati/Miller Transplantation Institute, Division of Liver Diseases, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Sharon Klarman
- Department of Surgery, Transplant Institute, Tufts Medical Center, Boston, Massachusetts, USA
| | - Michael Kriss
- Division of Gastroenterology & Hepatology, Department of Medicine, University of Colorado School of Medicine, Aurora, Colorado, USA
| | - Dempsey Hughes
- Division of Gastroenterology and Hepatology, Department of Medicine, Northwestern University, Chicago, Illinois, USA
| | - Pratima Sharma
- Division of Gastroenterology and Hepatology, University of Michigan, Ann Arbor, Michigan, USA
| | - Sumeet Asrani
- Hepatology, Baylor University Medical Center, Dallas, Texas, USA
| | - Alan Hutchison
- Section of Gastroenterology, Hepatology and Nutrition, Department of Medicine, University of Chicago Medicine, Chicago, Illinois, USA
| | - Paul Myoung
- Division of Transplant Surgery, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Asad Zaman
- Division of Gastroenterology and Hepatology, Department of Medicine, Northwestern University, Chicago, Illinois, USA
| | - Laura Butler
- Transplant Services, Montefiore Medical Center, Bronx, New York, USA
| | - James Pomposelli
- Department of Surgery, University of Colorado, Denver, Colorado, USA
| | - Fredric Gordon
- Department of Surgery, Transplant Institute, Tufts Medical Center, Boston, Massachusetts, USA
| | - Andres Duarte-Rojo
- Division of Gastroenterology and Hepatology, Department of Medicine, Northwestern University, Chicago, Illinois, USA
| | - Deepika Devuni
- Division of Gastroenterology, UMass Chan Medical School, Worcester, Massachusetts, USA
| | - Brett Fortune
- Division of Hepatology, Montefiore Einstein Medicine, Bronx, New York, USA
| |
Collapse
|
10
|
Xia M, Varmazyad M, Pla-Palacín I, Gavlock DC, DeBiasio R, LaRocca G, Reese C, Florentino RM, Faccioli LAP, Brown JA, Vernetti LA, Schurdak M, Stern AM, Gough A, Behari J, Soto-Gutierrez A, Taylor DL, Miedel MT. Comparison of wild-type and high-risk PNPLA3 variants in a human biomimetic liver microphysiology system for metabolic dysfunction-associated steatotic liver disease precision therapy. Front Cell Dev Biol 2024; 12:1423936. [PMID: 39324073 PMCID: PMC11422722 DOI: 10.3389/fcell.2024.1423936] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2024] [Accepted: 08/16/2024] [Indexed: 09/27/2024] Open
Abstract
Metabolic dysfunction-associated steatotic liver disease (MASLD) is a worldwide health epidemic with a global occurrence of approximately 30%. The pathogenesis of MASLD is a complex, multisystem disorder driven by multiple factors, including genetics, lifestyle, and the environment. Patient heterogeneity presents challenges in developing MASLD therapeutics, creating patient cohorts for clinical trials, and optimizing therapeutic strategies for specific patient cohorts. Implementing pre-clinical experimental models for drug development creates a significant challenge as simple in vitro systems and animal models do not fully recapitulate critical steps in the pathogenesis and the complexity of MASLD progression. To address this, we implemented a precision medicine strategy that couples the use of our liver acinus microphysiology system (LAMPS) constructed with patient-derived primary cells. We investigated the MASLD-associated genetic variant patatin-like phospholipase domain-containing protein 3 (PNPLA3) rs738409 (I148M variant) in primary hepatocytes as it is associated with MASLD progression. We constructed the LAMPS with genotyped wild-type and variant PNPLA3 hepatocytes, together with key non-parenchymal cells, and quantified the reproducibility of the model. We altered media components to mimic blood chemistries, including insulin, glucose, free fatty acids, and immune-activating molecules to reflect normal fasting (NF), early metabolic syndrome (EMS), and late metabolic syndrome (LMS) conditions. Finally, we investigated the response to treatment with resmetirom, an approved drug for metabolic syndrome-associated steatohepatitis (MASH), the progressive form of MASLD. This study, using primary cells, serves as a benchmark for studies using "patient biomimetic twins" constructed with patient induced pluripotent stem cell (iPSC)-derived liver cells using a panel of reproducible metrics. We observed increased steatosis, immune activation, stellate cell activation, and secretion of pro-fibrotic markers in the PNPLA3 GG variant compared to the wild-type CC LAMPS, consistent with the clinical characterization of this variant. We also observed greater resmetirom efficacy in the PNPLA3 wild-type CC LAMPS compared to the GG variant in multiple MASLD metrics, including steatosis, stellate cell activation, and the secretion of pro-fibrotic markers. In conclusion, our study demonstrates the capability of the LAMPS platform for the development of MASLD precision therapeutics, enrichment of patient cohorts for clinical trials, and optimization of therapeutic strategies for patient subgroups with different clinical traits and disease stages.
Collapse
Affiliation(s)
- Mengying Xia
- Drug Discovery Institute, University of Pittsburgh, Pittsburgh, PA, United States
| | - Mahboubeh Varmazyad
- Drug Discovery Institute, University of Pittsburgh, Pittsburgh, PA, United States
| | - Iris Pla-Palacín
- Drug Discovery Institute, University of Pittsburgh, Pittsburgh, PA, United States
| | - Dillon C. Gavlock
- Drug Discovery Institute, University of Pittsburgh, Pittsburgh, PA, United States
| | - Richard DeBiasio
- Drug Discovery Institute, University of Pittsburgh, Pittsburgh, PA, United States
| | - Gregory LaRocca
- Drug Discovery Institute, University of Pittsburgh, Pittsburgh, PA, United States
| | - Celeste Reese
- Drug Discovery Institute, University of Pittsburgh, Pittsburgh, PA, United States
| | - Rodrigo M. Florentino
- Department of Pathology, School of Medicine, University of Pittsburgh, Pittsburgh, PA, United States
- Center for Transcriptional Medicine, University of Pittsburgh, Pittsburgh, PA, United States
- Pittsburgh Liver Research Center, University of Pittsburgh, Pittsburgh, PA, United States
| | - Lanuza A. P. Faccioli
- Department of Pathology, School of Medicine, University of Pittsburgh, Pittsburgh, PA, United States
- Center for Transcriptional Medicine, University of Pittsburgh, Pittsburgh, PA, United States
- Pittsburgh Liver Research Center, University of Pittsburgh, Pittsburgh, PA, United States
| | - Jacquelyn A. Brown
- Drug Discovery Institute, University of Pittsburgh, Pittsburgh, PA, United States
- Department of Computational and System Biology, School of Medicine, University of Pittsburgh, Pittsburgh, PA, United States
| | - Lawrence A. Vernetti
- Drug Discovery Institute, University of Pittsburgh, Pittsburgh, PA, United States
- Department of Computational and System Biology, School of Medicine, University of Pittsburgh, Pittsburgh, PA, United States
| | - Mark Schurdak
- Drug Discovery Institute, University of Pittsburgh, Pittsburgh, PA, United States
- Pittsburgh Liver Research Center, University of Pittsburgh, Pittsburgh, PA, United States
- Department of Computational and System Biology, School of Medicine, University of Pittsburgh, Pittsburgh, PA, United States
| | - Andrew M. Stern
- Drug Discovery Institute, University of Pittsburgh, Pittsburgh, PA, United States
- Department of Computational and System Biology, School of Medicine, University of Pittsburgh, Pittsburgh, PA, United States
| | - Albert Gough
- Drug Discovery Institute, University of Pittsburgh, Pittsburgh, PA, United States
- Pittsburgh Liver Research Center, University of Pittsburgh, Pittsburgh, PA, United States
| | - Jaideep Behari
- Pittsburgh Liver Research Center, University of Pittsburgh, Pittsburgh, PA, United States
- Division of Gastroenterology, Hepatology and Nutrition, School of Medicine, University of Pittsburgh, Pittsburgh, PA, United States
| | - Alejandro Soto-Gutierrez
- Drug Discovery Institute, University of Pittsburgh, Pittsburgh, PA, United States
- Department of Pathology, School of Medicine, University of Pittsburgh, Pittsburgh, PA, United States
- Center for Transcriptional Medicine, University of Pittsburgh, Pittsburgh, PA, United States
- Pittsburgh Liver Research Center, University of Pittsburgh, Pittsburgh, PA, United States
| | - D. Lansing Taylor
- Drug Discovery Institute, University of Pittsburgh, Pittsburgh, PA, United States
- Pittsburgh Liver Research Center, University of Pittsburgh, Pittsburgh, PA, United States
- Department of Computational and System Biology, School of Medicine, University of Pittsburgh, Pittsburgh, PA, United States
| | - Mark T. Miedel
- Drug Discovery Institute, University of Pittsburgh, Pittsburgh, PA, United States
- Department of Pathology, School of Medicine, University of Pittsburgh, Pittsburgh, PA, United States
- Pittsburgh Liver Research Center, University of Pittsburgh, Pittsburgh, PA, United States
| |
Collapse
|
11
|
Fishman JC, Qian C, Kim Y, Rochon H, Szabo SM, Sun R, Charlton M. Cost burden of cirrhosis and liver disease progression in metabolic dysfunction-associated steatohepatitis: A US cohort study. J Manag Care Spec Pharm 2024; 30:929-941. [PMID: 38845444 PMCID: PMC11365567 DOI: 10.18553/jmcp.2024.24069] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/31/2024]
Abstract
BACKGROUND Metabolic dysfunction-associated steatohepatitis (MASH), formerly nonalcoholic steatohepatitis, is characterized by fat accumulation and inflammation of the liver and may result in progression to cirrhosis and liver-related events. OBJECTIVE To characterize the impact of cirrhosis and progression to liver-related events on costs and health care resource use (HCRU) among MASH patients in the United States. METHODS The study cohort included patients with diagnosed nonalcoholic steatohepatitis (International Classification of Diseases, Tenth Revision, Clinical Modification code K75.81) in Optum's deidentified Clinformatics Data Mart Database (October 2015 to December 2022) and were stratified by baseline cirrhosis status. Among those without cirrhosis at baseline, patients were further stratified by status of progression to cirrhosis during follow-up. Total HCRU and costs per-person per-year (PPPY) were estimated and compared descriptively between the cohorts. In addition, gamma generalized linear models were used to compare costs PPPY between those with vs without cirrhosis at baseline, as well as with vs without progression during follow-up, while adjusting for baseline patient and disease characteristics. Annual costs per person were also longitudinally modeled using gamma generalized linear mixed models to understand longitudinal changes in costs PPPY while accounting for time correlations within individual patients. Lastly, a series of sensitivity analyses were conducted to assess the impact of study design features and clinical variations of total costs PPPY. RESULTS A total of 28,576 adults were included, and 9,157 (32.0%) had baseline cirrhosis; of the 19,419 without baseline cirrhosis, a total of 4,235 (21.8%) progressed over follow-up. Mean (SD) HCRU and costs PPPY were higher among patients with cirrhosis ($110,403 [$226,037]) than without ($28,340 [$61,472]; P < 0.01) and among those with progression ($58,128 [$102,626]) than without ($20,031 [$39,740]; P < 0.01). Costs remained significantly greater when adjusted for covariates, with a risk ratio (95% CI) of 1.99 (1.89-2.09) when comparing with vs without baseline cirrhosis and 2.28 (2.15-2.42) when comparing with vs without progression over follow-up. Costs increased with each subsequent year, to 21% by year 6 among those with cirrhosis at baseline and 49% among those without baseline cirrhosis who progressed. CONCLUSIONS The financial burden of MASH is substantial and significantly greater among those with cirrhosis or disease progression. Although patients without cirrhosis incur lower burden, the increase over time is greater and associated with progression. Therapies that slow progression may help alleviate the financial burden, and strategies are needed to identify patients with MASH at risk of progressing to cirrhosis.
Collapse
Affiliation(s)
| | | | - Yestle Kim
- Madrigal Pharmaceuticals, Inc., West Conshohocken, PA
| | | | | | - Rosie Sun
- Broadstreet HEOR, Vancouver, BC, Canada
| | | |
Collapse
|
12
|
Younossi ZM, Mangla KK, Chandramouli AS, Lazarus JV. Estimating the economic impact of comorbidities in patients with MASH and defining high-cost burden in patients with noncirrhotic MASH. Hepatol Commun 2024; 8:e0488. [PMID: 39037377 PMCID: PMC11265778 DOI: 10.1097/hc9.0000000000000488] [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: 11/22/2023] [Accepted: 05/21/2024] [Indexed: 07/23/2024] Open
Abstract
BACKGROUND Metabolic dysfunction-associated steatohepatitis (MASH) is associated with high health care costs. This US study investigated the economic burden of MASH, particularly in patients without cirrhosis, and the impact of comorbidities on health care costs. METHODS This retrospective, observational study used data from patients diagnosed with MASH aged ≥18 years from October 2015 to March 2022 (IQVIA Ambulatory electronic medical record-US). Patients were stratified by the absence or presence of cirrhosis. Primary outcomes included baseline characteristics and annualized total health care cost after MASH diagnosis during follow-up. In addition, this study defined high costs for the MASH population and identified patient characteristics associated with increased health care costs among those without cirrhosis. RESULTS Overall, 16,919 patients (14,885 without cirrhosis and 2034 with cirrhosis) were included in the analysis. The prevalence of comorbidities was high in both groups; annual total health care costs were higher in patients with cirrhosis. Patients with a high-cost burden (threshold defined using the United States national estimated annual health care expenditure of $13,555) had a higher prevalence of comorbidities and were prescribed more cardiovascular medications. MASH diagnosis was associated with an increase in cost, largely driven by inpatient costs. In patients without cirrhosis, an increase in cost following MASH diagnosis was associated with the presence and burden of comorbidities and cardiovascular medication utilization. CONCLUSIONS Comorbidities, such as cardiovascular disease and type 2 diabetes, are associated with a higher cost burden and may be aggravated by MASH. Prioritization and active management may benefit patients without cirrhosis with these comorbidities. Clinical care should focus on preventing progression to cirrhosis and managing high-burden comorbidities.
Collapse
Affiliation(s)
- Zobair M. Younossi
- The Global NASH Council, Washington, District of Columbia, USA
- Beatty Liver and Obesity Research Program, Inova Health System, Falls Church, Virginia, USA
| | - Kamal Kant Mangla
- Novo Nordisk Service Center India Pvt Ltd, Bangalore, Karnataka, India
| | | | - Jeffrey V. Lazarus
- The Global NASH Council, Washington, District of Columbia, USA
- City University of New York Graduate School of Public Health and Health Policy (CUNY SPH), New York, New York, USA
- Barcelona Institute for Global Health (ISGlobal), Hospital Clínic, University of Barcelona, Barcelona, Spain
| |
Collapse
|
13
|
Sandireddy R, Sakthivel S, Gupta P, Behari J, Tripathi M, Singh BK. Systemic impacts of metabolic dysfunction-associated steatotic liver disease (MASLD) and metabolic dysfunction-associated steatohepatitis (MASH) on heart, muscle, and kidney related diseases. Front Cell Dev Biol 2024; 12:1433857. [PMID: 39086662 PMCID: PMC11289778 DOI: 10.3389/fcell.2024.1433857] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2024] [Accepted: 07/01/2024] [Indexed: 08/02/2024] Open
Abstract
Metabolic dysfunction-associated steatotic liver disease (MASLD), previously known as non-alcoholic fatty liver disease (NAFLD), is the most common liver disorder worldwide, with an estimated global prevalence of more than 31%. Metabolic dysfunction-associated steatohepatitis (MASH), formerly known as non-alcoholic steatohepatitis (NASH), is a progressive form of MASLD characterized by hepatic steatosis, inflammation, and fibrosis. This review aims to provide a comprehensive analysis of the extrahepatic manifestations of MASH, focusing on chronic diseases related to the cardiovascular, muscular, and renal systems. A systematic review of published studies and literature was conducted to summarize the findings related to the systemic impacts of MASLD and MASH. The review focused on the association of MASLD and MASH with metabolic comorbidities, cardiovascular mortality, sarcopenia, and chronic kidney disease. Mechanistic insights into the concept of lipotoxic inflammatory "spill over" from the MASH-affected liver were also explored. MASLD and MASH are highly associated (50%-80%) with other metabolic comorbidities such as impaired insulin response, type 2 diabetes, dyslipidemia, hypertriglyceridemia, and hypertension. Furthermore, more than 90% of obese patients with type 2 diabetes have MASH. Data suggest that in middle-aged individuals (especially those aged 45-54), MASLD is an independent risk factor for cardiovascular mortality, sarcopenia, and chronic kidney disease. The concept of lipotoxic inflammatory "spill over" from the MASH-affected liver plays a crucial role in mediating the systemic pathological effects observed. Understanding the multifaceted impact of MASH on the heart, muscle, and kidney is crucial for early detection and risk stratification. This knowledge is also timely for implementing comprehensive disease management strategies addressing multi-organ involvement in MASH pathogenesis.
Collapse
Affiliation(s)
| | | | | | | | - Madhulika Tripathi
- Cardiovascular and Metabolic Disorders Research Program, Duke-NUS Medical School, Singapore, Singapore
| | - Brijesh Kumar Singh
- Cardiovascular and Metabolic Disorders Research Program, Duke-NUS Medical School, Singapore, Singapore
| |
Collapse
|
14
|
Wright DR, Batista M, Wrightson T. #SharingHEOR: Developing Modern Media for Communication and Dissemination of Health Economics and Outcomes Research. APPLIED HEALTH ECONOMICS AND HEALTH POLICY 2024; 22:447-455. [PMID: 38427216 DOI: 10.1007/s40258-023-00863-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 11/27/2023] [Indexed: 03/02/2024]
Abstract
Questions regarding the effectiveness and safety of health interventions and allocation of health care resources are frequently discussed in mainstream and social media. Additionally, government and foundation funders are increasingly mandating that results be disseminated to the lay public and patients may benefit from being able to digest scientific research regarding their health conditions. Therefore, it is important to widely disseminate and clearly communication health economics and outcomes research (HEOR) findings to a range of interested parties. Digital media features such as graphical or visual abstracts, infographics and videos are informative and add value to research articles by improving reader engagement with articles, potentially increasing their impact, and allowing results to be more widely disseminated. However, use of novel digital media for research dissemination has been relatively limited to date. In this article, we discuss the rationale for developing novel media to communicate and disseminate research findings and offer practical advice for doing so. We conclude by outlining a future agenda for research regarding HEOR communication and dissemination.
Collapse
Affiliation(s)
- Davene R Wright
- Department of Population Medicine, Harvard Medical School and Harvard Pilgrim Health Care Institute, 401 Park Drive, Suite 401E, Boston, MA, 02215, USA.
| | - Mikaela Batista
- Department of Population Medicine, Harvard Medical School and Harvard Pilgrim Health Care Institute, 401 Park Drive, Suite 401E, Boston, MA, 02215, USA
| | | |
Collapse
|
15
|
Sangineto M, Ciarnelli M, Colangelo T, Moola A, Bukke VN, Duda L, Villani R, Romano A, Giandomenico S, Kanwal H, Serviddio G. Monocyte bioenergetics: An immunometabolic perspective in metabolic dysfunction-associated steatohepatitis. Cell Rep Med 2024; 5:101564. [PMID: 38733988 PMCID: PMC11148801 DOI: 10.1016/j.xcrm.2024.101564] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2023] [Revised: 02/18/2024] [Accepted: 04/18/2024] [Indexed: 05/13/2024]
Abstract
Monocytes (Mos) are crucial in the evolution of metabolic dysfunction-associated steatotic liver disease (MASLD) to metabolic dysfunction-associated steatohepatitis (MASH), and immunometabolism studies have recently suggested targeting leukocyte bioenergetics in inflammatory diseases. Here, we reveal a peculiar bioenergetic phenotype in circulating Mos of patients with MASH, characterized by high levels of glycolysis and mitochondrial (mt) respiration. The enhancement of mt respiratory chain activity, especially complex II (succinate dehydrogenase [SDH]), is unbalanced toward the production of reactive oxygen species (ROS) and is sustained at the transcriptional level with the involvement of the AMPK-mTOR-PGC-1α axis. The modulation of mt activity with dimethyl malonate (DMM), an SDH inhibitor, restores the metabolic profile and almost abrogates cytokine production. Analysis of a public single-cell RNA sequencing (scRNA-seq) dataset confirms that in murine models of MASH, liver Mo-derived macrophages exhibit an upregulation of mt and glycolytic energy pathways. Accordingly, the DMM injection in MASH mice contrasts Mo infiltration and macrophagic enrichment, suggesting immunometabolism as a potential target in MASH.
Collapse
Affiliation(s)
- Moris Sangineto
- C.U.R.E. (University Center for Liver Disease Research and Treatment), Liver Unit, Department of Medical and Surgical Sciences, University of Foggia, 71122 Foggia, Italy.
| | - Martina Ciarnelli
- C.U.R.E. (University Center for Liver Disease Research and Treatment), Liver Unit, Department of Medical and Surgical Sciences, University of Foggia, 71122 Foggia, Italy
| | - Tommaso Colangelo
- Department of Medical and Surgical Sciences, University of Foggia, 71122 Foggia, Italy; Cancer Cell Signalling Unit, Fondazione IRCCS "Casa Sollievo della Sofferenza," 71043 San Giovanni Rotondo (FG), Italy
| | - Archana Moola
- C.U.R.E. (University Center for Liver Disease Research and Treatment), Liver Unit, Department of Medical and Surgical Sciences, University of Foggia, 71122 Foggia, Italy
| | - Vidyasagar Naik Bukke
- C.U.R.E. (University Center for Liver Disease Research and Treatment), Liver Unit, Department of Medical and Surgical Sciences, University of Foggia, 71122 Foggia, Italy
| | - Loren Duda
- Pathology Unit, Department of Clinical and Experimental Medicine, University of Foggia, 71122 Foggia, Italy
| | - Rosanna Villani
- C.U.R.E. (University Center for Liver Disease Research and Treatment), Liver Unit, Department of Medical and Surgical Sciences, University of Foggia, 71122 Foggia, Italy
| | - Antonino Romano
- C.U.R.E. (University Center for Liver Disease Research and Treatment), Liver Unit, Department of Medical and Surgical Sciences, University of Foggia, 71122 Foggia, Italy
| | - Stefania Giandomenico
- C.U.R.E. (University Center for Liver Disease Research and Treatment), Liver Unit, Department of Medical and Surgical Sciences, University of Foggia, 71122 Foggia, Italy
| | - Hina Kanwal
- C.U.R.E. (University Center for Liver Disease Research and Treatment), Liver Unit, Department of Medical and Surgical Sciences, University of Foggia, 71122 Foggia, Italy
| | - Gaetano Serviddio
- C.U.R.E. (University Center for Liver Disease Research and Treatment), Liver Unit, Department of Medical and Surgical Sciences, University of Foggia, 71122 Foggia, Italy
| |
Collapse
|
16
|
Xia M, Varmazyad M, Palacin IP, Gavlock DC, Debiasio R, LaRocca G, Reese C, Florentino R, Faccioli LAP, Brown JA, Vernetti LA, Schurdak ME, Stern AM, Gough A, Behari J, Soto-Gutierrez A, Taylor DL, Miedel M. Comparison of Wild-Type and High-risk PNPLA3 variants in a Human Biomimetic Liver Microphysiology System for Metabolic Dysfunction-associated Steatotic Liver Disease Precision Therapy. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.04.22.590608. [PMID: 38712213 PMCID: PMC11071381 DOI: 10.1101/2024.04.22.590608] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/08/2024]
Abstract
Metabolic dysfunction-associated steatotic liver disease (MASLD) is a worldwide health epidemic with a global occurrence of approximately 30%. The pathogenesis of MASLD is a complex, multisystem disorder driven by multiple factors including genetics, lifestyle, and the environment. Patient heterogeneity presents challenges for developing MASLD therapeutics, creation of patient cohorts for clinical trials and optimization of therapeutic strategies for specific patient cohorts. Implementing pre-clinical experimental models for drug development creates a significant challenge as simple in vitro systems and animal models do not fully recapitulate critical steps in the pathogenesis and the complexity of MASLD progression. To address this, we implemented a precision medicine strategy that couples the use of our liver acinus microphysiology system (LAMPS) constructed with patient-derived primary cells. We investigated the MASLD-associated genetic variant PNPLA3 rs738409 (I148M variant) in primary hepatocytes, as it is associated with MASLD progression. We constructed LAMPS with genotyped wild type and variant PNPLA3 hepatocytes together with key non-parenchymal cells and quantified the reproducibility of the model. We altered media components to mimic blood chemistries, including insulin, glucose, free fatty acids, and immune activating molecules to reflect normal fasting (NF), early metabolic syndrome (EMS) and late metabolic syndrome (LMS) conditions. Finally, we investigated the response to treatment with resmetirom, an approved drug for metabolic syndrome-associated steatohepatitis (MASH), the progressive form of MASLD. This study using primary cells serves as a benchmark for studies using patient biomimetic twins constructed with patient iPSC-derived liver cells using a panel of reproducible metrics. We observed increased steatosis, immune activation, stellate cell activation and secretion of pro-fibrotic markers in the PNPLA3 GG variant compared to wild type CC LAMPS, consistent with the clinical characterization of this variant. We also observed greater resmetirom efficacy in PNPLA3 wild type CC LAMPS compared to the GG variant in multiple MASLD metrics including steatosis, stellate cell activation and the secretion of pro-fibrotic markers. In conclusion, our study demonstrates the capability of the LAMPS platform for the development of MASLD precision therapeutics, enrichment of patient cohorts for clinical trials, and optimization of therapeutic strategies for patient subgroups with different clinical traits and disease stages.
Collapse
|
17
|
Rudolfsen JH, Gluud LL, Grønbæk H, Jensen MK, Vyberg M, Olsen J, Bo Poulsen P, Hovelsø N, Gregersen NT, Thomsen AB, Jepsen P. Societal costs and survival of patients with biopsy-verified non-alcoholic steatohepatitis: Danish nationwide register-based study. Ann Hepatol 2024; 29:101285. [PMID: 38272183 DOI: 10.1016/j.aohep.2024.101285] [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: 10/11/2023] [Revised: 12/14/2023] [Accepted: 01/05/2024] [Indexed: 01/27/2024]
Abstract
INTRODUCTION AND OBJECTIVES Studies on the societal burden of patients with biopsy-confirmed non-alcoholic fatty liver disease (NAFLD) are sparse. This study examined this question, comparing NAFLD with matched reference groups. MATERIALS AND METHODS Nationwide Danish healthcare registers were used to include all patients (≥18 years) diagnosed with biopsy-verified NAFLD (1997-2021). Patients were classified as having simple steatosis or non-alcoholic steatohepatitis (NASH) with or without cirrhosis, and all matched with liver-disease free reference groups. Healthcare costs and labour market outcomes were compared from 5 years before to 11 years after diagnosis. Patients were followed for 25 years to analyse risk of disability insurance and death. RESULTS 3,712 patients with biopsy-verified NASH (n = 1,030), simple steatosis (n = 1,540) or cirrhosis (n = 1,142) were identified. The average total costs in the year leading up to diagnosis was 4.1-fold higher for NASH patients than the reference group (EUR 6,318), 6.2-fold higher for cirrhosis patients and 3.1-fold higher for simple steatosis patients. In NASH, outpatient hospital contacts were responsible for 49 % of the excess costs (EUR 3,121). NASH patients had statistically significantly lower income than their reference group as early as five years before diagnosis until nine years after diagnosis, and markedly higher risk of becoming disability insurance recipients (HR: 4.37; 95 % CI: 3.17-6.02) and of death (HR: 2.42; 95 % CI: 1.80-3.25). CONCLUSIONS NASH, simple steatosis and cirrhosis are all associated with substantial costs for the individual and the society with excess healthcare costs and poorer labour market outcomes.
Collapse
Affiliation(s)
| | - Lise Lotte Gluud
- Copenhagen University Hospital Hvidovre, Gastro Unit, DK-2650 Hvidovre, Denmark
| | - Henning Grønbæk
- Aarhus University Hospital, Department of Hepatology & Gastroenterology, DK-8200 Aarhus, Denmark; University of Aarhus, Department of Clinical Medicine, DK-8200 Aarhus, Denmark
| | - Majken K Jensen
- University of Copenhagen, Department of Public Health, Section of Epidemiology, Copenhagen, Denmark
| | - Mogens Vyberg
- Aalborg University Campus Copenhagen, Department of Clinical Medicine, DK-2450 Copenhagen, Denmark; Copenhagen University Hvidovre, Department of Pathology, DK-2650 Hvidovre, Denmark
| | | | | | - Nanna Hovelsø
- Pfizer Denmark Aps, Medical Affairs, DK-2750 Ballerup, Denmark
| | | | | | - Peter Jepsen
- Aarhus University Hospital, Department of Hepatology & Gastroenterology, DK-8200 Aarhus, Denmark; University of Aarhus, Department of Clinical Medicine, DK-8200 Aarhus, Denmark
| |
Collapse
|
18
|
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
|
19
|
Willett RA, Tryndyak VP, Beland FA, Pogribny IP. Cellular and molecular alterations in a human hepatocellular in vitro model of nonalcoholic fatty liver disease development and stratification. JOURNAL OF ENVIRONMENTAL SCIENCE AND HEALTH. PART C, TOXICOLOGY AND CARCINOGENESIS 2024; 42:74-92. [PMID: 38105681 DOI: 10.1080/26896583.2023.2293493] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/19/2023]
Abstract
The rapidly increasing incidence of nonalcoholic fatty liver disease (NAFLD) is a growing health crisis worldwide. If not detected early, NAFLD progression can lead to irreversible pathological states, including liver fibrosis and cirrhosis. Using in vitro models to understand the molecular pathogenesis has been extremely beneficial; however, most studies have utilized only short-term exposures, highlighting a limitation in current research to model extended fat-induced liver injury. We treated Hep3B cells continuously with a low dose of oleic and palmitic free fatty acids (FFAs) for 7 or 28 days. Transcriptomic analysis identified dysregulated molecular pathways and differential expression of 984 and 917 genes after FFA treatment for 7 and 28 days respectively. DNA methylation analysis of altered DNA methylated regions (DMRs) found 7 DMRs in common. Pathway analysis of differentially expressed genes (DEGs) revealed transcriptomic changes primarily involved in lipid metabolism, small molecule biochemistry, and molecular transport. Western blot analysis revealed changes in PDK4 and CPT1A protein levels, indicative of mitochondrial stress. In line with this, there was mitochondrial morphological change demonstrating breakdown of the mitochondrial network. This in vitro model of human NAFL mimics results observed in human patients and may be used as a pre-clinical model for drug intervention.
Collapse
Affiliation(s)
- Rose A Willett
- Division of Biochemical Toxicology, FDA-National Center for Toxicological Research, Jefferson, Arkansas, USA
| | - Volodymyr P Tryndyak
- Division of Biochemical Toxicology, FDA-National Center for Toxicological Research, Jefferson, Arkansas, USA
| | - Frederick A Beland
- Division of Biochemical Toxicology, FDA-National Center for Toxicological Research, Jefferson, Arkansas, USA
| | - Igor P Pogribny
- Division of Biochemical Toxicology, FDA-National Center for Toxicological Research, Jefferson, Arkansas, USA
| |
Collapse
|
20
|
Josol VJD, Salvador PBU, Cruz LLA, Ornos EDB, Tantengco OAG. Trends of nonalcoholic fatty liver research in Southeast Asia from 2004 to 2022: A bibliometric analysis. OBESITY MEDICINE 2024; 45:100527. [DOI: 10.1016/j.obmed.2023.100527] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2025]
|
21
|
Roberts R, Williams DM, Min T, Barry J, Stephens JW. Benefits in routinely measured liver function tests following bariatric surgery: a retrospective cohort study. J Diabetes Metab Disord 2023; 22:1763-1768. [PMID: 37975098 PMCID: PMC10638127 DOI: 10.1007/s40200-023-01311-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/16/2023] [Accepted: 09/12/2023] [Indexed: 11/19/2023]
Abstract
Background Bariatric surgery is well-established to support long-term metabolic health benefits associated with considerable weight loss. Here, we aim to determine the longer-term impact of bariatric surgery on liver enzymes and associations with other metabolic improvements. Methods One hundred patients who underwent bariatric surgery between 2007 and 2014 were included, and changes in liver enzymes, anthropometric measures and other parameters were observed over a mean 9.8 years. Results At the time of surgery, the mean age was 45.4 ± 9.6 years, weight 141.2 ± 31.6 kg, and body mass index (BMI) 50.2 ± 10.1 kg/m2. Most patients underwent sleeve gastrectomy [n = 71] with a mean follow-up duration 9.8 ± 2.3 years. From baseline, alanine transaminase (ALT) reduced by 41.3% within 12 months post-operatively (36.6 ± 29.2 U/L to 21.5 ± 14.9 U/L, p < 0.001), which was sustained at recent follow-up (20.2 ± 10.7 U/L, p < 0.001). There were associated reductions in body weight, BMI, HbA1c, blood pressure and triglycerides. Patients with greater baseline ALT had the greatest reduction in ALT over follow-up. Conclusions Bariatric surgery is associated with rapid and sustained improvements in routine liver enzymes at 10 years, and sustained improvements in features of the metabolic syndrome. Supplementary Information The online version contains supplementary material available at 10.1007/s40200-023-01311-4.
Collapse
Affiliation(s)
- Richard Roberts
- Diabetes Centre, Morriston Hospital, Swansea Bay University Health Board, Swansea, SA6 6NL UK
| | - David M Williams
- Diabetes Centre, Morriston Hospital, Swansea Bay University Health Board, Swansea, SA6 6NL UK
- Diabetes Centre, Singleton Hospital, Swansea Bay University Health Board, Swansea, UK
| | - Thinzar Min
- Diabetes Centre, Singleton Hospital, Swansea Bay University Health Board, Swansea, UK
- Diabetes Research Group, Swansea University Medical School, Swansea University, Swansea, UK
- Diabetes Centre, Neath Port Talbot Hospital, Baglan Way, Port Talbot, SA12 7BX UK
| | - Jonathan Barry
- Welsh Institute of Metabolic & Obesity Surgery, Morriston Hospital, Swansea Bay UHB, Swansea, SA6 6NL UK
| | - Jeffrey W Stephens
- Diabetes Centre, Morriston Hospital, Swansea Bay University Health Board, Swansea, SA6 6NL UK
- Diabetes Research Group, Swansea University Medical School, Swansea University, Swansea, UK
| |
Collapse
|
22
|
Mondal T, Smith CI, Loffredo CA, Quartey R, Moses G, Howell CD, Korba B, Kwabi-Addo B, Nunlee-Bland G, R. Rucker L, Johnson J, Ghosh S. Transcriptomics of MASLD Pathobiology in African American Patients in the Washington DC Area †. Int J Mol Sci 2023; 24:16654. [PMID: 38068980 PMCID: PMC10706626 DOI: 10.3390/ijms242316654] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2023] [Revised: 11/17/2023] [Accepted: 11/18/2023] [Indexed: 12/18/2023] Open
Abstract
Metabolic-dysfunction-associated steatotic liver disease (MASLD) is becoming the most common chronic liver disease worldwide and is of concern among African Americans (AA) in the United States. This pilot study evaluated the differential gene expressions and identified the signature genes in the disease pathways of AA individuals with MASLD. Blood samples were obtained from MASLD patients (n = 23) and non-MASLD controls (n = 24) along with their sociodemographic and medical details. Whole-blood transcriptomic analysis was carried out using Affymetrix Clarion-S Assay. A validation study was performed utilizing TaqMan Arrays coupled with Ingenuity Pathway Analysis (IPA) to identify the major disease pathways. Out of 21,448 genes in total, 535 genes (2.5%) were significantly (p < 0.05) and differentially expressed when we compared the cases and controls. A significant overlap in the predominant differentially expressed genes and pathways identified in previous studies using hepatic tissue was observed. Of note, TGFB1 and E2F1 genes were upregulated, and HMBS was downregulated significantly. Hepatic fibrosis signaling is the top canonical pathway, and its corresponding biofunction contributes to the development of hepatocellular carcinoma. The findings address the knowledge gaps regarding how signature genes and functional pathways can be detected in blood samples ('liquid biopsy') in AA MASLD patients, demonstrating the potential of the blood samples as an alternative non-invasive source of material for future studies.
Collapse
Affiliation(s)
- Tanmoy Mondal
- Department of Biology, Howard University, Washington, DC 20059, USA; (T.M.); (G.M.); (J.J.)
| | - Coleman I. Smith
- MedStar-Georgetown Transplantation Institute, Georgetown University School of Medicine, Washington, DC 20007, USA;
| | | | - Ruth Quartey
- Department of Internal Medicine, College of Medicine, Howard University, Washington, DC 20007, USA; (R.Q.); (C.D.H.)
| | - Gemeyel Moses
- Department of Biology, Howard University, Washington, DC 20059, USA; (T.M.); (G.M.); (J.J.)
| | - Charles D. Howell
- Department of Internal Medicine, College of Medicine, Howard University, Washington, DC 20007, USA; (R.Q.); (C.D.H.)
| | - Brent Korba
- Department of Microbiology & Immunology, Georgetown University, Washington, DC 20007, USA;
| | - Bernard Kwabi-Addo
- Department of Biochemistry, College of Medicine, Howard University, Washington, DC 20059, USA;
| | - Gail Nunlee-Bland
- Departments of Pediatrics and Child Health, College of Medicine, Howard University, Washington, DC 20059, USA;
| | - Leanna R. Rucker
- Department of Internal Medicine, MedStar Georgetown University Hospital, Washington, DC 20007, USA;
| | - Jheannelle Johnson
- Department of Biology, Howard University, Washington, DC 20059, USA; (T.M.); (G.M.); (J.J.)
| | - Somiranjan Ghosh
- Department of Biology, Howard University, Washington, DC 20059, USA; (T.M.); (G.M.); (J.J.)
- Departments of Pediatrics and Child Health, College of Medicine, Howard University, Washington, DC 20059, USA;
| |
Collapse
|
23
|
Fishman J, Tapper EB, Dodge S, Miller K, Lewandowski D, Bogdanov A, Bonafede M. The incremental cost of non-alcoholic steatohepatitis and type 2 diabetes in the United States using real-world data. Curr Med Res Opin 2023; 39:1425-1429. [PMID: 37740457 DOI: 10.1080/03007995.2023.2262926] [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: 08/17/2023] [Accepted: 09/21/2023] [Indexed: 09/24/2023]
Abstract
BACKGROUND Non-alcoholic steatohepatitis (NASH) and type 2 diabetes (T2D) are both linked to substantial healthcare costs and are often co-occurring. We aim to quantify the incremental cost of NASH and T2D using real-world data. METHODS Adults (≥18 years old) with ≥2 diagnosis codes for NASH and/or ≥2 diagnosis codes for T2D between 1/1/2016 and 12/31/2021 and ≥24 months of continuous claims enrollment (study period) were identified in electronic health records or claims in the Veradigm Integrated Dataset. Patients were stratified into 3 cohorts: NASH-only, T2D-only, and NASH + T2D. We calculated annualized costs for the 24-month study period and fit a generalized linear model (excluding the most expensive 1%) that controlled for disease cohort, age, sex, and modified Charlson comorbidity index to estimate the per year all-cause healthcare costs and incremental cost of adding T2D to a NASH diagnosis (or vice versa). RESULTS We identified 23,111 patients diagnosed with NASH-only, 3,548,786 patients with T2D-only, and 30,339 patients with NASH + T2D. The model-predicted mean costs per year were $7,668 for patients with NASH-only, $11,226 for patients with T2D-only, and $16,812 for patients with NASH + T2D. The incremental increase in costs per year of adding T2D to NASH was 63% (+$4,846), and the incremental increase in costs per year of adding NASH to T2D was 42% (+$4,692). CONCLUSIONS Both NASH and T2D contribute to the high healthcare costs among patients with a dual diagnosis. Results from our analysis indicate that NASH comprises a high portion of total healthcare costs among patients with NASH and T2D.
Collapse
Affiliation(s)
- Jesse Fishman
- Madrigal Pharmaceuticals, West Conshohocken, PA, USA
| | | | - Stephen Dodge
- Madrigal Pharmaceuticals, West Conshohocken, PA, USA
| | - Keith Miller
- Madrigal Pharmaceuticals, West Conshohocken, PA, USA
| | | | | | | |
Collapse
|
24
|
Poynard T, Deckmyn O, Peta V, Paradis V, Gautier JF, Brzustowski A, Bedossa P, Castera L, Pol S, Valla D. Prospective direct comparison of non-invasive liver tests in outpatients with type 2 diabetes using intention-to-diagnose analysis. Aliment Pharmacol Ther 2023; 58:888-902. [PMID: 37642160 DOI: 10.1111/apt.17688] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/05/2023] [Revised: 07/23/2023] [Accepted: 08/11/2023] [Indexed: 08/31/2023]
Abstract
BACKGROUND No prospective diagnostic studies have directly compared widespread non-invasive liver tests in patients with type 2 diabetes (T2D) using the intention-to-diagnose method for each of the three main histological features of metabolic dysfunction associated steatotic liver disease - namely fibrosis, metabolic dysfunction-associated steatohepatitis (MASH), and steatosis. AIMS To compare the performance of nine tests using the intention-to-diagnose rather than the standard method, which would exclude non-evaluable participants METHODS: Biopsy was used as the reference with predetermined cut-offs, advanced fibrosis being the main endpoint. The Nash-FibroTest panel including FibroTest-T2D, SteatoTest-T2D and MashTest-T2D was optimised for type 2 diabetes. FibroTest-T2D was compared to vibration-controlled transient elastography stiffness (VCTE), two-dimensional shear-wave elastography stiffness (TD-SWE), and Fibrosis-4 blood test. NashTest-T2D was compared to aspartate aminotransferase. SteatoTest-T2D was compared to the controlled attenuation parameter and the hepatorenal gradient. RESULTS Among 402 cases, non-evaluable tests were 6.7% for VCTE, 4.0% for hepatorenal gradient, 3.2% for controlled attenuation parameter, 1.5% for TD-SWE, 1.2% for NashTest-T2D, and 0.02% for Fibrosis-4, aspartate aminotransferase and SteatoTest-T2D. The VCTE AUROC for advanced fibrosis was over-estimated by 6% (0.83 [95% CI: 0.78-0.87]) by standard analysis compared to intention-to-diagnose (0.77 [0.72-0.81] p = 0.008). The AUROCs for advanced fibrosis did not differ significantly in intention-to-diagnose between FibroTest-T2D (0.77; 95% CI: 0.73-0.82), VCTE (0.77; 95% CI: 0.72-0.81) and TD-SWE(0.78; 0.74-0.83) but were all higher than the Fibrosis-4 score (0.70; 95% CI all differences ≥7%; p ≤ 0.03). For MASH, MashTest-T2D had a higher AUROC (0.76; 95% CI: 0.70-0.80) than aspartate aminotransferase (0.72; 95% CI: 0.66-0.77; p = 0.035). For steatosis, AUROCs did not differ significantly between SteatoTest-T2D, controlled attenuation parameter and hepatorenal gradient. CONCLUSIONS In intention-to-diagnose analysis, FibroTest-T2D, TD-SWE and VCTE performed similarly for staging fibrosis, and out-performed Fibrosis-4 in outpatients with type 2 diabetes. The standard analysis over-estimated VCTE performance. CLINICALTRIAL gov: NCT03634098.
Collapse
Affiliation(s)
- Thierry Poynard
- Centre de Recherche Saint-Antoine (CRSA), INSERM, Institute of Cardiometabolism and Nutrition (ICAN), Sorbonne Université, Paris, France
- BioPredictive, Paris, France
| | | | | | - Valérie Paradis
- Department of Pathology, AP-HP, Beaujon Hospital, Clichy, France
| | - Jean-Francois Gautier
- Department of Diabetes and Endocrinology, APHP, INSERM U1138, Hôpital Lariboisière, Paris, France
| | | | - Pierre Bedossa
- Department of Pathology, AP-HP, Beaujon Hospital, Clichy, France
| | - Laurent Castera
- Department of Hepatology, AP-HP, Beaujon Hospital, Clichy, France
| | - Stanislas Pol
- Department of Hepatology, Cochin Hospital, Université Paris Descartes, Paris, France
| | - Dominique Valla
- Department of Hepatology, AP-HP, Beaujon Hospital, Clichy, France
| |
Collapse
|
25
|
Fishman J, Higgins V, Piercy J, Pike J. Cross-walk of the Chronic Liver Disease Questionnaire for Nonalcoholic Steatohepatitis (CLDQ-NASH) and the EuroQol EQ-5D-5L in patients with NASH. Health Qual Life Outcomes 2023; 21:113. [PMID: 37838660 PMCID: PMC10576276 DOI: 10.1186/s12955-023-02195-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2023] [Accepted: 09/27/2023] [Indexed: 10/16/2023] Open
Abstract
BACKGROUND Nonalcoholic steatohepatitis (NASH) is a chronic progression of nonalcoholic fatty liver disease, which can negatively impact the health-related quality of life (HRQoL) of affected individuals. HRQoL in NASH has been assessed using the disease-specific Chronic Liver Disease Questionnaire for NASH (CLDQ-NASH) and the generic EuroQol EQ-5D-5L. As the performance of these instruments relative to each other is unknown, we performed a cross-walk analysis of CLDQ-NASH to EQ-5D-5L using data from a real-world NASH population. METHODS Data were drawn from the Adelphi Real World 2019 NASH Disease Specific Programme, a cross-sectional survey of physicians and their patients in the United States. Patients with physician-diagnosed NASH completed a questionnaire that included the CLDQ-NASH and EQ-5D-5L. Mapping from CLDQ-NASH to EQ-5D-5L was done using tenfold cross-validation; performance was assessed using root-mean squared error as accuracy measure. Subgroup analyses compared performance of the models in obese versus non-obese patients and patients with versus without type 2 diabetes (T2D). RESULTS Data from 347 patients were included in this analysis. Overall, 2172 models were tested for predicting EQ-5D-5L index score from CLDQ-NASH score. The best model for this mapping was a generalized linear model using Gaussian distribution and a power link. The best model for mapping from CLDQ-NASH domains to the EQ-5D-5L was a fractional logistic model. Models performed better at predicting upper versus lower values of EQ-5D-5L, for non-obese versus obese patients, and for patients without versus with T2D. CONCLUSION We describe a scoring algorithm for cross-walking the CLDQ-NASH to the EQ-5D-5L enabling health status comparisons of HRQoL across studies.
Collapse
|
26
|
Younossi ZM, Wong G, Anstee QM, Henry L. The Global Burden of Liver Disease. Clin Gastroenterol Hepatol 2023; 21:1978-1991. [PMID: 37121527 DOI: 10.1016/j.cgh.2023.04.015] [Citation(s) in RCA: 164] [Impact Index Per Article: 82.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/15/2023] [Revised: 04/09/2023] [Accepted: 04/23/2023] [Indexed: 05/02/2023]
Abstract
Chronic liver disease (CLD) and its associated complications (cirrhosis and liver cancer) cause significant mortality, morbidity, and economic burden. Published data from the World Health Organization and/or the Global Burden of Disease show that the burden of CLD is large and increasing, primarily owing to the increasing burden of nonalcoholic fatty liver disease and alcohol-related liver disease (ALD). Middle Eastern, Northern African, and Asian regions of the globe are most affected by hepatitis B and hepatitis C virus. Furthermore, Middle Eastern and North African regions also are affected by nonalcoholic fatty liver disease, and Eastern European, West African, and Central Asian regions are affected by ALD. In this context, the rate of increase for cirrhosis is highest in the Middle East, as well as in middle high and high sociodemographic index (SDI) regions. On the other hand, the highest SDI countries are experiencing increasing rates of hepatocellular carcinoma (HCC). Assessing HCC burden based on country and etiology shows that China, Korea, India, Japan, and Thailand have the highest hepatitis B virus-related HCC cases, while China, Japan, and the United States have the highest hepatitis C virus-related HCC cases. Additionally, the United States has the highest ALD-related HCC cases, while India, the United States, and Thailand have the highest nonalcoholic steatohepatitis-related HCC cases. Although the burden of CLD is increasing globally, regions of the world are impacted differently as a result of a number of sociodemographic factors.
Collapse
Affiliation(s)
- Zobair M Younossi
- Betty and Guy Beatty Center for Integrated Research, Inova Health System, Falls Church, Virginia; Center for Liver Disease, Department of Medicine, Inova Fairfax Medical Campus, Falls Church, Virginia; Inova Medicine, Inova Health System, Falls Church, Virginia.
| | - Grace Wong
- Department of Medicine and Therapeutics, The Chinese University of Hong Kong, Hong Kong
| | - Quentin M Anstee
- Translational and Clinical Research Institute, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne, United Kingdom; Newcastle National Institute Health and Care Research Biomedical Research Centre, Newcastle upon Tyne Hospitals National Health Service Trust, Newcastle upon Tyne, United Kingdom
| | - Linda Henry
- Betty and Guy Beatty Center for Integrated Research, Inova Health System, Falls Church, Virginia; Inova Medicine, Inova Health System, Falls Church, Virginia; Center for Outcomes Research in Liver Disease, Washington, District of Columbia
| |
Collapse
|
27
|
Clark JM, Cryer DRH, Morton M, Shubrook JH. Nonalcoholic fatty liver disease from a primary care perspective. Diabetes Obes Metab 2023; 25:1421-1433. [PMID: 36789676 DOI: 10.1111/dom.15016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/04/2022] [Revised: 01/31/2023] [Accepted: 02/07/2023] [Indexed: 02/16/2023]
Abstract
Nonalcoholic fatty liver disease (NAFLD) affects up to one-third of the US population. Approximately one-fifth of patients with NAFLD have nonalcoholic steatohepatitis (NASH), characterized by hepatocyte damage and inflammation with or without fibrosis. NASH leads to greater risk of liver-related complications and liver-related mortality, with the poorest outcomes seen in patients with advanced fibrosis. NASH is also associated with other metabolic comorbidities and conveys an increased risk of adverse cardiovascular outcomes and extrahepatic cancers. Despite its high prevalence, NAFLD is frequently underdiagnosed. This is a significant concern, given that early diagnosis of NAFLD is a key step in preventing progression to NASH. In this review, we describe the clinical impact of NASH from the perspective of both the clinician and the patient. In addition, we provide practical guidance on the diagnosis and management of NASH for primary care providers, who play a pivotal role in the frontline care of patients with NASH, and we use case studies to illustrate real-world scenarios encountered in the primary care setting.
Collapse
Affiliation(s)
- Jeanne M Clark
- Department of Medicine, Division of General Internal Medicine, The Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Donna R H Cryer
- Global Liver Institute, Washington, District of Columbia, USA
| | | | - Jay H Shubrook
- Primary Care Department, Touro University California College of Osteopathic Medicine, Vallejo, California, USA
| |
Collapse
|
28
|
Carpi RZ, Barbalho SM, Sloan KP, Laurindo LF, Gonzaga HF, Grippa PC, Zutin TLM, Girio RJS, Repetti CSF, Detregiachi CRP, Bueno PCS, Mazuqueli Pereira EDSB, Goulart RDA, Haber JFDS. The Effects of Probiotics, Prebiotics and Synbiotics in Non-Alcoholic Fat Liver Disease (NAFLD) and Non-Alcoholic Steatohepatitis (NASH): A Systematic Review. Int J Mol Sci 2022; 23:8805. [PMID: 35955942 PMCID: PMC9369010 DOI: 10.3390/ijms23158805] [Citation(s) in RCA: 74] [Impact Index Per Article: 24.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2022] [Revised: 08/01/2022] [Accepted: 08/06/2022] [Indexed: 12/11/2022] Open
Abstract
Modifications in the microbiota caused by environmental and genetic reasons can unbalance the intestinal homeostasis, deregulating the host's metabolism and immune system, intensifying the risk factors for the development and aggravation of non-alcoholic fat liver disease (NAFLD). The use of probiotics, prebiotics and synbiotics have been considered a potential and promising strategy to regulate the gut microbiota and produce beneficial effects in patients with liver conditions. For this reason, this review aimed to evaluate the effectiveness of probiotics, prebiotics, and symbiotics in patients with NAFLD and NASH. Pubmed, Embase, and Cochrane databases were consulted, and PRISMA (Preferred Reporting Items for Systematic Reviews and Meta-Analysis) guidelines were followed. The clinical trials used in this study demonstrated that gut microbiota interventions could improve a wide range of markers of inflammation, glycemia, insulin resistance, dyslipidemia, obesity, liver injury (decrease of hepatic enzymes and steatosis and fibrosis). Although microbiota modulators do not play a healing role, they can work as an important adjunct therapy in pathological processes involving NAFLD and its spectrums, either by improving the intestinal barrier or by preventing the formation of toxic metabolites for the liver or by acting on the immune system.
Collapse
Affiliation(s)
- Rodrigo Zamignan Carpi
- Department of Biochemistry and Pharmacology, School of Medicine, University of Marília (UNIMAR), Avenida Hygino Muzzy Filho, 1001, Marilia 17525-902, SP, Brazil
| | - Sandra M. Barbalho
- Department of Biochemistry and Pharmacology, School of Medicine, University of Marília (UNIMAR), Avenida Hygino Muzzy Filho, 1001, Marilia 17525-902, SP, Brazil
- Postgraduate Program in Structural and Functional Interactions in Rehabilitation, University of Marília (UNIMAR), Marilia 17525-902, SP, Brazil
- School of Food and Technology of Marilia (FATEC), Marilia 17506-000, SP, Brazil
| | | | - Lucas Fornari Laurindo
- Department of Biochemistry and Pharmacology, School of Medicine, University of Marília (UNIMAR), Avenida Hygino Muzzy Filho, 1001, Marilia 17525-902, SP, Brazil
| | - Heron Fernando Gonzaga
- Department of Biochemistry and Pharmacology, School of Medicine, University of Marília (UNIMAR), Avenida Hygino Muzzy Filho, 1001, Marilia 17525-902, SP, Brazil
- Postgraduate Program in Structural and Functional Interactions in Rehabilitation, University of Marília (UNIMAR), Marilia 17525-902, SP, Brazil
| | - Paulo Cesar Grippa
- Postgraduate Program in Structural and Functional Interactions in Rehabilitation, University of Marília (UNIMAR), Marilia 17525-902, SP, Brazil
| | - Tereza L. Menegucci Zutin
- Department of Biochemistry and Pharmacology, School of Medicine, University of Marília (UNIMAR), Avenida Hygino Muzzy Filho, 1001, Marilia 17525-902, SP, Brazil
- Postgraduate Program in Structural and Functional Interactions in Rehabilitation, University of Marília (UNIMAR), Marilia 17525-902, SP, Brazil
| | - Raul J. S. Girio
- Department of Animal Sciences, School of Veterinary Medicine, University of Marília (UNIMAR), Avenida Hygino Muzzy Filho, 1001, Marilia 17525-902, SP, Brazil
| | - Cláudia Sampaio Fonseca Repetti
- Department of Animal Sciences, School of Veterinary Medicine, University of Marília (UNIMAR), Avenida Hygino Muzzy Filho, 1001, Marilia 17525-902, SP, Brazil
| | - Cláudia Rucco Penteado Detregiachi
- Postgraduate Program in Structural and Functional Interactions in Rehabilitation, University of Marília (UNIMAR), Marilia 17525-902, SP, Brazil
| | - Patrícia C. Santos Bueno
- Department of Biochemistry and Pharmacology, School of Medicine, University of Marília (UNIMAR), Avenida Hygino Muzzy Filho, 1001, Marilia 17525-902, SP, Brazil
- Department of Animal Sciences, School of Veterinary Medicine, University of Marília (UNIMAR), Avenida Hygino Muzzy Filho, 1001, Marilia 17525-902, SP, Brazil
| | - Eliana de Souza Bastos Mazuqueli Pereira
- Postgraduate Program in Structural and Functional Interactions in Rehabilitation, University of Marília (UNIMAR), Marilia 17525-902, SP, Brazil
- Department of Biochemistry, School of Dentistry, University of Marília (UNIMAR), Avenida Hygino Muzzy Filho, 1001, Marilia 17525-902, SP, Brazil
| | - Ricardo de Alvares Goulart
- Postgraduate Program in Structural and Functional Interactions in Rehabilitation, University of Marília (UNIMAR), Marilia 17525-902, SP, Brazil
| | - Jesselina Francisco dos Santos Haber
- Department of Biochemistry and Pharmacology, School of Medicine, University of Marília (UNIMAR), Avenida Hygino Muzzy Filho, 1001, Marilia 17525-902, SP, Brazil
| |
Collapse
|
29
|
Wang X. Challenges and opportunities in nonalcoholic steatohepatitis. MEDICAL REVIEW (BERLIN, GERMANY) 2022; 2:328-330. [PMID: 37724322 PMCID: PMC10388777 DOI: 10.1515/mr-2022-0024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/29/2022] [Accepted: 08/21/2022] [Indexed: 09/20/2023]
Abstract
Nonalcoholic steatohepatitis (NASH) has emerged as the leading cause of chronic liver disease worldwide and is rapidly increasing in prevalence due to the obesity epidemic. There are currently no Food and Drug Administration (FDA) approved drugs to treat NASH, and therefore a critical need exists for novel therapies that can halt or reverse the progression to hepatic fibrosis, cirrhosis, and hepatocellular carcinoma. Clinical trials to date using single drugs to treat NASH have shown disappointing efficacy. Combination therapies to attack different targets underlying disease pathogenesis of NASH are being explored as a strategy currently. Novel RNA therapies are also being developed to target previously "undruggable" targets and are close to the maturity necessary to be viable therapeutic approaches for the treatment of NASH and fibrosis. Identifying circulating biomarkers of fibrosis could serve as a valuable, non-invasive diagnostic tool to guide clinical practice. Despite progress in translational and clinical research, one of the major reasons for the absence of effective therapeutics is our incomplete understanding of the pathophysiology that underlies the progression from steatosis to NASH and its most deadly consequence-fibrosis. Multi-omics platforms will help to drive effective precision medicine development in NASH and hepatology.
Collapse
Affiliation(s)
- Xiaobo Wang
- Department of Medicine, Columbia University Irving Medical Center, New York, NY, USA
| |
Collapse
|