Accurate liquid biopsy for the diagnosis of non-alcoholic steatohepatitis and liver fibrosis

Giulia Angelini, Simona Panunzi, Lidia Castagneto-Gissey, Francesca Pellicanò, Andrea De Gaetano, Maurizio Pompili, Laura Riccardi, Matteo Garcovich, Marco Raffaelli, Luigi Ciccoritti, Ornella Verrastro, Maria Francesca Russo, Fabio Maria Vecchio, Giovanni Casella, James Casella-Mariolo, Luigi Papa, Pier Luigi Marini, Francesco Rubino, Carel W le Roux, Stefan Bornstein, Geltrude Mingrone, Giulia Angelini, Simona Panunzi, Lidia Castagneto-Gissey, Francesca Pellicanò, Andrea De Gaetano, Maurizio Pompili, Laura Riccardi, Matteo Garcovich, Marco Raffaelli, Luigi Ciccoritti, Ornella Verrastro, Maria Francesca Russo, Fabio Maria Vecchio, Giovanni Casella, James Casella-Mariolo, Luigi Papa, Pier Luigi Marini, Francesco Rubino, Carel W le Roux, Stefan Bornstein, Geltrude Mingrone

Abstract

Objective: Clinical diagnosis and approval of new medications for non-alcoholic steatohepatitis (NASH) require invasive liver biopsies. The aim of our study was to identify non-invasive biomarkers of NASH and/or liver fibrosis.

Design: This multicentre study includes 250 patients (discovery cohort, n=100 subjects (Bariatric Surgery Versus Non-alcoholic Steato-hepatitis - BRAVES trial); validation cohort, n=150 (Liquid Biopsy for NASH and Liver Fibrosis - LIBRA trial)) with histologically proven non-alcoholic fatty liver (NAFL) or NASH with or without fibrosis. Proteomics was performed in monocytes and hepatic stellate cells (HSCs) with iTRAQ-nano- Liquid Chromatography - Mass Spectrometry/Mass Spectrometry (LC-MS/MS), while flow cytometry measured perilipin-2 (PLIN2) and RAB14 in peripheral blood CD14+CD16- monocytes. Neural network classifiers were used to predict presence/absence of NASH and NASH stages. Logistic bootstrap-based regression was used to measure the accuracy of predicting liver fibrosis.

Results: The algorithm for NASH using PLIN2 mean florescence intensity (MFI) combined with waist circumference, triglyceride, alanine aminotransferase (ALT) and presence/absence of diabetes as covariates had an accuracy of 93% in the discovery cohort and of 92% in the validation cohort. Sensitivity and specificity were 95% and 90% in the discovery cohort and 88% and 100% in the validation cohort, respectively.The area under the receiver operating characteristic (AUROC) for NAS level prediction ranged from 83.7% (CI 75.6% to 91.8%) in the discovery cohort to 97.8% (CI 95.8% to 99.8%) in the validation cohort.The algorithm including RAB14 MFI, age, waist circumference, high-density lipoprotein cholesterol, plasma glucose and ALT levels as covariates to predict the presence of liver fibrosis yielded an AUROC of 95.9% (CI 87.9% to 100%) in the discovery cohort and 99.3% (CI 98.1% to 100%) in the validation cohort, respectively. Accuracy was 99.25%, sensitivity 100% and specificity 95.8% in the discovery cohort and 97.6%, 99% and 89.6% in the validation cohort. This novel biomarker was superior to currently used FIB4, non-alcoholic fatty liver disease fibrosis score and aspartate aminotransferase (AST)-to-platelet ratio and was comparable to ultrasound two-dimensional shear wave elastography.

Conclusions: The proposed novel liquid biopsy is accurate, sensitive and specific in diagnosing the presence and severity of NASH or liver fibrosis and is more reliable than currently used biomarkers.

Clinical trials: Discovery multicentre cohort: Bariatric Surgery versus Non-Alcoholic Steatohepatitis, BRAVES, ClinicalTrials.gov identifier: NCT03524365.Validation multicentre cohort: Liquid Biopsy for NASH and Fibrosis, LIBRA, ClinicalTrials.gov identifier: NCT04677101.

Keywords: HEPATIC FIBROSIS; NONALCOHOLIC STEATOHEPATITIS.

Conflict of interest statement

Competing interests: GM reports consulting fees from Novo Nordisk,_Fractyl Inc and Recor Inc; she is also scientific current advisor and consultant of Metadeq Limited, and current advisor and consultant of Keyron Limited, GHP Scientific Limited, and Jemyll Limited. FR reports receiving research grants from Ethicon and Medtronic; consulting fees from Novo Nordisk, Ethicon and Medtronic; serving on scientific advisory boards for GI Dynamics; and is former director and current stock option holder of Metadeq Limited and former director and current advisor of Keyron Limited and GHP Scientific Limited. CWlR reports grants from the Irish Research Council, Science Foundation Ireland, Anabio and the Health Research Board; serves on advisory boards of Novo Nordisk, Herbalife, GI Dynamics, Eli Lilly, Johnson & Johnson, Sanofi Aventis, AstraZeneca, Janssen, Bristol-Myers Squibb, Glia and Boehringer Ingelheim. ClR is a member of the Irish Society for Nutrition and Metabolism outside the area of work commented on here, and is the chief medical officer and director of the Medical Device Division of Keyron since January 2011; both of these are unremunerated positions. CWlR is also current director, shareholder and stock option holder of Metadeq Limited, current director of GHP Scientific Limited, was a previous investor in Keyron, which develops endoscopically implantable medical devices intended to mimic the surgical procedures of sleeve gastrectomy and gastric bypass. The product has only been tested in rodents and none of Keyron’s products are currently licensed. They do not have any contracts with other companies to put their products into clinical practice. No patients have been included in any of Keyron’s studies and they are not listed on the stock market. He continues to provide scientific advice to Keyron for no remuneration. All other authors declare no competing interests.

© Author(s) (or their employer(s)) 2023. Re-use permitted under CC BY-NC. No commercial re-use. See rights and permissions. Published by BMJ.

Figures

Figure 1
Figure 1
Laser scanning confocal immunofluorescence of monocytes, HSCs and hepatocytes from a representative subject with NASH/fibrosis (upper panels) and a representative subjects with negative histology for NASH/fibrosis (lower panels). (A–D) RAB14 staining in monocytes (A, B) and in HSCs (C, D). (E) Quantification of RAB14 protein expression by flow cytometry, in monocytes and HSCs of 20 subjects with NASH/fibrosis and 20 subjects with negative histology for NASH/fibrosis. (F) Spearman correlation analysis and linear regression line fitting of RAB14 expression in monocytes and HSCs. (G–J) PLIN2 staining in monocytes (G, H) and in hepatocytes (I, J). (K) Quantification of PLIN2 protein expression by flow cytometry in monocytes and hepatocytes of 20 subjects with NASH/fibrosis and 20 subjects with negative histology for NASH/fibrosis. (L) Spearman correlation analysis and linear regression line fitting of PLIN2 expression in monocytes and hepatocytes. (M–P) LDs staining with Nile Red in monocytes (M, N) and in hepatocytes (O, P). The mean NAS was 4.05±0.15 in the 20 subjects with NASH and liver fibrosis and 0.65±0.11 in the 20 subjects study without NASH and liver fibrosis from LIBRA. SAF-F mean was 1.95±0.17. Data are expressed as mean±SEM for linear regression analysis; Spearman rank correlation coefficients (R2) and p values are shown. Magnification ×60. Scale bar: 50 µm. DAPI, 4', 6-Diamidino-2-Phenylindole; HSC, hepatic stellate cell; LD, lipid droplet; MFI, mean florescence intensity; NAS, Non-alcoholic Fatty Liver Disease Activity Score; NASH, non-alcoholic steatohepatitis; PLIN2, perilipin-2.
Figure 2
Figure 2
PLIN2 diagnostic performance for predicting presence/absence of NASH. (A) Architecture of the NN used. Each input node represents a biomarker, while edges represent the weights between layers. The thickness of the edge is proportional to the magnitude of each weight. Positive weights are plotted as black lines; negative weights as grey lines. In the NN, there are five input nodes: PLIN2, presence of diabetes, plasma triglycerides and ALT levels and waist circumference. There are three hidden nodes and one output node for presence of NASH. Two biases nodes are included. The bias node covers the same function of the intercept in a regression model. (B) ROC curves for predicting the presence of NASH (NAS ≥3) in the discovery and in the validation cohorts. FPR=1–specificity. AUC, area under the curve; B, bias weight; FPR, false-positive rate; H, hidden node; I, input node; MFI, mean florescence intensity; NAS, Non-alcoholic Fatty Liver Disease Activity Score; NASH, non-alcoholic steatohepatitis; NN, neural network; O, output node; PLIN2, perilipin-2; ROC, receiver operating characteristic; TPR, true-positive rate.
Figure 3
Figure 3
PLIN2 diagnostic performance for predicting NASH severity. (A) PLIN2 biomarker network for diagnosing of NASH severity through NAS levels. Architecture of the NN used. Each input node represents a biomarker, while edges represent the weights between layers. The thickness of the edge is proportional to the magnitude of each weight. Positive weights are plotted as black lines; negative weights as grey lines. In the NN, there are five input nodes: PLIN2, presence of diabetes, plasma triglycerides and ALT levels and waist circumference. There are three hidden nodes and three output nodes for NAS level=0 for total NAS score

Figure 4

Whisker plots of monocyte PLIN2…

Figure 4

Whisker plots of monocyte PLIN2 levels (MFI) measured by flow cytometry in the…

Figure 4
Whisker plots of monocyte PLIN2 levels (MFI) measured by flow cytometry in the training and validation cohorts in different NASH stages: histological NAS

Figure 5

RAB14 (A) and elastography (C)…

Figure 5

RAB14 (A) and elastography (C) diagnostic performance for presence/absence of NASH. The model…

Figure 5
RAB14 (A) and elastography (C) diagnostic performance for presence/absence of NASH. The model estimates ±SE (A) (RAB14) are intercept −62.25±22.15, RAB14 −0.87±0.81, waist circumference 12.03±4.28, plasma glucose 1.09±3.66, age 3.33±2.40, HDL cholesterol −1.80±4.64 and ALT −0.23±1.25. The model estimates ±SE (C) (elastography) are intercept −61.69±22.84, elastography 2.49±2.48, waist circumference 10.1±4.84, plasma glucose 1.64±3.74, age 2.97±2.49, HDL cholesterol −1.43±1.70 and ALT −0.16±1.22. Monocyte RAB14 levels (MFI) measured by flow cytometry at Steatosis, Activity, Fibrosis-Fibrosis (SAF-F) scores 0 (NO Fibrosis), ≥1 (YES Fibrosis) in the training and validation cohorts (B). Ultrasound transient elastography (kPa) at SAF-F scores 0 (NO Fibrosis), ≥1 (YES Fibrosis) in the training and validation cohorts (D). AUC, area under the curve; FPR, false-positive rate; HDL, high-density lipoprotein; MFI, mean fluorescence intensity; ROC, receiver operating characteristic; TPR, true-positive rate.
Similar articles
Cited by
References
    1. Wong VW-S, Chitturi S, Wong GL-H, et al. . Pathogenesis and novel treatment options for non-alcoholic steatohepatitis. Lancet Gastroenterol Hepatol 2016;1:56–67. 10.1016/S2468-1253(16)30011-5 - DOI - PubMed
    1. Pioglitazone vs vitamin E vs placebo for treatment of non-diabetic patients with nonalcoholic steatohepatitis (PIVENS). Available: https://clinicaltrials.gov/ct2/show/NCT00063622
    1. Friedman SL, Ratziu V, Harrison SA, et al. . A randomized, placebo-controlled trial of cenicriviroc for treatment of nonalcoholic steatohepatitis with fibrosis. Hepatology 2018;67:1754–67. 10.1002/hep.29477 - DOI - PMC - PubMed
    1. Haukeland JW, Damås JK, Konopski Z, et al. . Systemic inflammation in nonalcoholic fatty liver disease is characterized by elevated levels of CCL2. J Hepatol 2006;44:1167–74. 10.1016/j.jhep.2006.02.011 - DOI - PubMed
    1. Smits MM, van Geenen EJM. The clinical significance of pancreatic steatosis. Nat Rev Gastroenterol Hepatol 2011;8:169–77. 10.1038/nrgastro.2011.4 - DOI - PubMed
Show all 43 references
Publication types
Associated data
Full text links [x]
[x]
Cite
Copy Download .nbib
Format: AMA APA MLA NLM
Figure 4
Figure 4
Whisker plots of monocyte PLIN2 levels (MFI) measured by flow cytometry in the training and validation cohorts in different NASH stages: histological NAS

Figure 5

RAB14 (A) and elastography (C)…

Figure 5

RAB14 (A) and elastography (C) diagnostic performance for presence/absence of NASH. The model…

Figure 5
RAB14 (A) and elastography (C) diagnostic performance for presence/absence of NASH. The model estimates ±SE (A) (RAB14) are intercept −62.25±22.15, RAB14 −0.87±0.81, waist circumference 12.03±4.28, plasma glucose 1.09±3.66, age 3.33±2.40, HDL cholesterol −1.80±4.64 and ALT −0.23±1.25. The model estimates ±SE (C) (elastography) are intercept −61.69±22.84, elastography 2.49±2.48, waist circumference 10.1±4.84, plasma glucose 1.64±3.74, age 2.97±2.49, HDL cholesterol −1.43±1.70 and ALT −0.16±1.22. Monocyte RAB14 levels (MFI) measured by flow cytometry at Steatosis, Activity, Fibrosis-Fibrosis (SAF-F) scores 0 (NO Fibrosis), ≥1 (YES Fibrosis) in the training and validation cohorts (B). Ultrasound transient elastography (kPa) at SAF-F scores 0 (NO Fibrosis), ≥1 (YES Fibrosis) in the training and validation cohorts (D). AUC, area under the curve; FPR, false-positive rate; HDL, high-density lipoprotein; MFI, mean fluorescence intensity; ROC, receiver operating characteristic; TPR, true-positive rate.
Figure 5
Figure 5
RAB14 (A) and elastography (C) diagnostic performance for presence/absence of NASH. The model estimates ±SE (A) (RAB14) are intercept −62.25±22.15, RAB14 −0.87±0.81, waist circumference 12.03±4.28, plasma glucose 1.09±3.66, age 3.33±2.40, HDL cholesterol −1.80±4.64 and ALT −0.23±1.25. The model estimates ±SE (C) (elastography) are intercept −61.69±22.84, elastography 2.49±2.48, waist circumference 10.1±4.84, plasma glucose 1.64±3.74, age 2.97±2.49, HDL cholesterol −1.43±1.70 and ALT −0.16±1.22. Monocyte RAB14 levels (MFI) measured by flow cytometry at Steatosis, Activity, Fibrosis-Fibrosis (SAF-F) scores 0 (NO Fibrosis), ≥1 (YES Fibrosis) in the training and validation cohorts (B). Ultrasound transient elastography (kPa) at SAF-F scores 0 (NO Fibrosis), ≥1 (YES Fibrosis) in the training and validation cohorts (D). AUC, area under the curve; FPR, false-positive rate; HDL, high-density lipoprotein; MFI, mean fluorescence intensity; ROC, receiver operating characteristic; TPR, true-positive rate.

References

    1. Wong VW-S, Chitturi S, Wong GL-H, et al. . Pathogenesis and novel treatment options for non-alcoholic steatohepatitis. Lancet Gastroenterol Hepatol 2016;1:56–67. 10.1016/S2468-1253(16)30011-5
    1. Pioglitazone vs vitamin E vs placebo for treatment of non-diabetic patients with nonalcoholic steatohepatitis (PIVENS). Available:
    1. Friedman SL, Ratziu V, Harrison SA, et al. . A randomized, placebo-controlled trial of cenicriviroc for treatment of nonalcoholic steatohepatitis with fibrosis. Hepatology 2018;67:1754–67. 10.1002/hep.29477
    1. Haukeland JW, Damås JK, Konopski Z, et al. . Systemic inflammation in nonalcoholic fatty liver disease is characterized by elevated levels of CCL2. J Hepatol 2006;44:1167–74. 10.1016/j.jhep.2006.02.011
    1. Smits MM, van Geenen EJM. The clinical significance of pancreatic steatosis. Nat Rev Gastroenterol Hepatol 2011;8:169–77. 10.1038/nrgastro.2011.4
    1. Guijas C, Meana C, Astudillo AM. Foamy monocytes are enriched in cis-7-Hexadecenoic fatty acid (16:1n-9), a possible biomarker for early detection of cardiovascular disease. Cell Chem Biol 2016;2368:9–99.
    1. Sztalryd C, Brasaemle DL. The perilipin family of lipid droplet proteins: Gatekeepers of intracellular lipolysis. Biochim Biophys Acta Mol Cell Biol Lipids 2017;1862:1221–32. 10.1016/j.bbalip.2017.07.009
    1. Diehl AM, Day C, Cause DC. Cause, pathogenesis, and treatment of nonalcoholic steatohepatitis. N Engl J Med 2017;377:2063–72. 10.1056/NEJMra1503519
    1. Angulo P, Kleiner DE, Dam-Larsen S, et al. . Liver fibrosis, but no other histologic features, is associated with long-term outcomes of patients with nonalcoholic fatty liver disease. Gastroenterology 2015;149:389–97. 10.1053/j.gastro.2015.04.043
    1. Karlmark KR, Wasmuth HE, Trautwein C, et al. . Chemokine-directedimmune cell infiltration in acute and chronic liver disease. Expert Rev Gastroenterol Hepatol 2008;2:233–42. 10.1586/17474124.2.2.233
    1. Kwok R, Tse Y-K, Wong GL-H, et al. . Systematic review with metaanalysis: non-invasive assessment of non-alcoholic fatty liver disease--the role of transient elastography and plasma cytokeratin-18 fragments. Aliment Pharmacol Ther 2014;39:254–69. 10.1111/apt.12569
    1. Feldstein AE, Lopez R, Tamimi TA-R, et al. . Mass spectrometric profiling of oxidized lipid products in human nonalcoholic fatty liver disease and nonalcoholic steatohepatitis. J Lipid Res 2010;51:3046–54. 10.1194/jlr.M007096
    1. He L, Deng L, Zhang Q, et al. . Diagnostic value of CK-18, FGF-21, and related biomarker panel in nonalcoholic fatty liver disease: a systematic review and meta-analysis. Biomed Res Int 2017;2017:1–12. 10.1155/2017/9729107
    1. Vilar-Gomez E, Chalasani N. Non-invasive assessment of non-alcoholic fatty liver disease: clinical prediction rules and blood-based biomarkers. J Hepatol 2018;68:305–15. 10.1016/j.jhep.2017.11.013
    1. Cabré N, Luciano-Mateo F, Baiges-Gayà G, et al. . Plasma metabolic alterations in patients with severe obesity and non-alcoholic steatohepatitis. Aliment Pharmacol Ther 2020;51:374–87. 10.1111/apt.15606
    1. Bril F, Millán L, Kalavalapalli S, et al. . Use of a metabolomic approach to non-invasively diagnose non-alcoholic fatty liver disease in patients with type 2 diabetes mellitus. Diabetes Obes Metab 2018;20:1702–9. 10.1111/dom.13285
    1. Mayo R, Crespo J, Martínez-Arranz I, et al. . Metabolomic-based noninvasive serum test to diagnose nonalcoholic steatohepatitis: results from discovery and validation cohorts. Hepatol Commun 2018;2:807–20. 10.1002/hep4.1188
    1. Qadri S, Ahlholm N, Lønsmann I, et al. . Obesity modifies the performance of fibrosis biomarkers in nonalcoholic fatty liver disease. J Clin Endocrinol Metab 2022;107:e2008–20. 10.1210/clinem/dgab933
    1. Angelini G, Salinari S, Bertuzzi A, et al. . Metabolic surgery improves insulin resistance through the reduction of gut-secreted heat shock proteins. Commun Biol 2018;1:69. 10.1038/s42003-018-0069-8
    1. Angelini G, Castagneto Gissey L, Corpo GD, et al. . Publisher correction: new insight into the mechanisms of ectopic fat deposition improvement after bariatric surgery. Sci Rep 2020;10:2786. 10.1038/s41598-020-58961-0
    1. Wang T, Ma C. The hepatic macrophage pool in NASH. Cell Mol Immunol 2021;18:2059–60. 10.1038/s41423-021-00690-z
    1. Davies LC, Jenkins SJ, Allen JE, et al. . Tissue-resident macrophages. Nat Immunol 2013;14:986–95. 10.1038/ni.2705
    1. Klein I, Cornejo JC, Polakos NK, et al. . Kupffer cell heterogeneity: functional properties of bone marrow derived and sessile hepatic macrophages. Blood 2007;110:4077–85. 10.1182/blood-2007-02-073841
    1. Hinz B, Phan SH, Thannickal VJ, et al. . Recent developments in myofibroblast biology: paradigms for connective tissue remodeling. Am J Pathol 2012;180:1340–55. 10.1016/j.ajpath.2012.02.004
    1. Itoh Y. Membrane-type matrix metalloproteinases: their functions and regulations. Matrix Biol 2015;44-46:207–23. 10.1016/j.matbio.2015.03.004
    1. Jin H, Tang Y, Yang L, et al. . Rab GTPases: central coordinators of membrane trafficking in cancer. Front. Cell Dev. Biol. 2021;9:648384. 10.3389/fcell.2021.648384
    1. Bedossa P. Diagnosis of non-alcoholic fatty liver disease/non-alcoholic steatohepatitis: why liver biopsy is essential. Liver Int 2018;38 Suppl 1:64–6. 10.1111/liv.13653
    1. Bedossa P, Poynard T. An algorithm for the grading of activity in chronic hepatitis C. the METAVIR cooperative Study Group. Hepatology 1996;24:289–93. 10.1002/hep.510240201
    1. Kleiner DE, Brunt EM, Van Natta M. Nonalcoholic steatohepatitis clinical research network. design and validation of a histological scoring system for nonalcoholic fatty liver disease. Hepatology 2005;41:1313–21.
    1. Bedossa P, Poitou C, Veyrie N, et al. . Histopathological algorithm and scoring system for evaluation of liver lesions in morbidly obese patients. Hepatology 2012;56:1751–9. 10.1002/hep.25889
    1. Brunt EM, Kleiner DE, Wilson LA, et al. . Nonalcoholic fatty liver disease (NAFLD) activity score and the histopathologic diagnosis in NAFLD: distinct clinicopathologic meanings. Hepatology 2011;53:810–20. 10.1002/hep.24127
    1. Garcovich M, Faccia M, Di Stasio E, et al. . Correlation between QElaXto techniques and supersonic Imagine for liver stiffness quantification in chronic liver disease. J Ultrasound Med 2022;41:877–86. 10.1002/jum.15767
    1. Goksuluk D, Korkmaz S, Zararsiz G, et al. . easyROC: an interactive web-tool for ROC curve analysis using R language environment. R J 2016;8:213–30. 10.32614/RJ-2016-042
    1. Olden JD, Joy MK, Death RG. An accurate comparison of methods for quantifying variable importance in artificial neural networks using simulated data. Ecol Modell 2004;178:389–97. 10.1016/j.ecolmodel.2004.03.013
    1. Perkins NJ, Schisterman EF. The inconsistency of "optimal" cutpoints obtained using two criteria based on the receiver operating characteristic curve. Am J Epidemiol 2006;163:670–5. 10.1093/aje/kwj063
    1. DeLong ER, DeLong DM, Clarke-Pearson DL. Comparing the areas under two or more correlated receiver operating characteristic curves: a nonparametric approach. Biometrics 1988;44:837–45. 10.2307/2531595
    1. R Core Team . R: a language and environment for statistical computing. R foundation for statistical computing, Vienna, Austria, 2013. Available:
    1. Najt CP, Senthivinayagam S, Aljazi MB, et al. . Liver-specific loss of perilipin 2 alleviates diet-induced hepatic steatosis, inflammation, and fibrosis. Am J Physiol Gastrointest Liver Physiol 2016;310:G726–38. 10.1152/ajpgi.00436.2015
    1. Francque SM, Bedossa P, Ratziu V, et al. . A randomized, controlled trial of the pan-PPAR agonist Lanifibranor in NASH. N Engl J Med 2021;385:1547–58. 10.1056/NEJMoa2036205
    1. Tan EX-X, Lee JW-J, Jumat NH, et al. . Non-obese non-alcoholic fatty liver disease (NAFLD) in Asia: an international registry study. Metabolism 2022;126:154911. 10.1016/j.metabol.2021.154911
    1. Ye Q, Zou B, Yeo YH, et al. . Global prevalence, incidence, and outcomes of non-obese or lean non-alcoholic fatty liver disease: a systematic review and meta-analysis. Lancet Gastroenterol Hepatol 2020;5:739–52. 10.1016/S2468-1253(20)30077-7
    1. Younossi ZM, Golabi P, de Avila L, et al. . The global epidemiology of NAFLD and NASH in patients with type 2 diabetes: a systematic review and meta-analysis. J Hepatol 2019;71:793–801. 10.1016/j.jhep.2019.06.021
    1. Vuppalanchi R, Siddiqui MS, Van Natta ML, et al. . Performance characteristics of vibration-controlled transient elastography for evaluation of nonalcoholic fatty liver disease. Hepatology 2018;67:134–44. 10.1002/hep.29489

Source: PubMed

3
구독하다