Next Article in Journal
Feature-Based Molecular Networking Facilitates the Comprehensive Identification of Differential Metabolites in Diabetic Cognitive Dysfunction Rats
Previous Article in Journal
A Review of the Impact of Maternal Prenatal Stress on Offspring Microbiota and Metabolites
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Screening for NAFLD—Current Knowledge and Challenges

Liver Unit, Division of Digestive Diseases, Department of Metabolism, Digestion and Reproduction, Faculty of Medicine, Imperial College London, London W21NY, UK
*
Author to whom correspondence should be addressed.
Metabolites 2023, 13(4), 536; https://doi.org/10.3390/metabo13040536
Submission received: 13 March 2023 / Revised: 5 April 2023 / Accepted: 6 April 2023 / Published: 9 April 2023

Abstract

:
Non-alcoholic fatty liver disease (NAFLD) is the most common cause of abnormal liver function tests worldwide, with an estimated prevalence ranging between 19–46% in the general population. Of note, NAFLD is also expected to become a leading cause of end-stage liver disease in the next decades. Given the high prevalence and severity of NAFLD, especially in high-risk populations (i.e., patients with type-2 diabetes mellitus and/or obesity), there is a major interest in early detection of the disease in primary care. Nevertheless, substantial uncertainties still surround the development of a screening policy for NAFLD, such as limitations in currently used non-invasive markers of fibrosis, cost-effectiveness and the absence of a licensed treatment. In this review, we summarise current knowledge and try to identify the limitations surrounding the screening policy for NAFLD in primary care.

1. Definition and Epidemiology of NAFLD

Non-alcoholic fatty liver disease (NAFLD) is the most common cause of abnormal liver function tests worldwide, with a global estimated prevalence of 30% [1]. NAFLD encompasses a spectrum of pathological disorders characterised by macro-vesicular fat accumulation (steatosis, non-alcoholic fatty liver, NAFL) with or without hepatocellular injury and/or inflammation (non-alcoholic steato-hepatitis, NASH) and a variable degree of fibrosis up to cirrhosis [2,3].
Overall, NAFLD prevalence is particularly high in those with metabolic syndrome, i.e., a combination of central obesity, insulin resistance, type 2 diabetes mellitus (T2DM), hypertension and dyslipidaemia [4]. According to tertiary care studies, more than 50% of the patients with T2DM have NAFLD [4]. Similarly, the prevalence of NAFLD is as high as 45% among those with increased body mass index (BMI > 30 kg/m2) and up to 90% among those undergoing bariatric surgery (BMI > 35 kg/m2) [5]. Mirroring the epidemic of metabolic syndrome, the prevalence of NAFLD is constantly increasing in the general population, increasing from 33% in 2005 to 59.1% in 2010, and in similar fashion, the prevalence of NASH increasing from 15% to 25% [6]. The total NAFLD population in 2015 was estimated at 83.1 million cases, which is projected to increase by 21% to 100.9 million cases by 2030 [7]. Furthermore, NAFLD/NASH has become the fastest growing indication for liver transplantation in the USA [8]. According to data from the European Liver Transplant Registry, the proportion of liver transplants performed for NASH increased from 1.2% in 2002 to 8.4% in 2016 [9].

2. Pathogenesis of NAFLD

Far from the old concept of being a dichotomic disease, NAFLD is now considered to be a dynamic disease with a wide spectrum of disease activity within different stages of simple steatosis or NASH [10]. Insulin resistance plays a crucial role in the development and progression of liver disease, as this stimulates de novo lipogenesis and is associated with impaired lipolysis, resulting in an increased flux of fatty acid to the liver [11]. Of note, hepatic triglyceride storage is not harmful per se. Nevertheless, when the hepatic capacity of using, storing, and exporting free fatty acids becomes saturated, lipotoxicity may occur within the liver. Lipotoxicity is thought to be the crucial driver for the development and progression of hepatocellular injury, inflammation, hepatic stellate cell activation and extracellular matrix deposition, leading to fibrosis progression [12]. Overall, a status of insulin resistance also drives a dysfunctional adipose tissue, which produces metabolically active cytokines and initiates an inflammatory cascade [13] (Figure 1).
The development and progression of liver disease in NAFLD is now being explained with the multi-hit hypothesis, where a plethora of modifiable (dietary and environmental) and non-modifiable (genetic) factors contribute to the disease along with the worsening of insulin resistance (Figure 1). Among the modifiable factors, dietary elements, both in terms of overall calorie intake and specific dietary patterns, may contribute to the development of NAFLD [14,15]. Specifically, a high-fat diet and increased fructose and red meat intake have been associated with worsening hepatic steatosis and the induction of a pro-inflammatory status [16,17,18]. More recently, there has been a large body of work showing that gut microbiome plays an essential role in disease activity in patients with NAFLD [19]. The interactions between the liver and the gut—the so called “gut–liver axis”—result from a complex interplay between the gut and the immune system, which ranges from immune tolerance to immune activation. Changes in gut microbiome composition [20], gut permeability [21], and the translocation of pro-inflammatory bacterial by-products [22] are now included among the factors involved in the progression of liver disease in this population [23]. Among the non-modifiable factors, genetic factors also represent an important contribution to NAFLD progression. Few genes have been identified as conferring different levels of susceptibility to fat accumulation, hepatic inflammation and lipotoxicity, such as patatin-like phospholipase domain containing protein 3 (PNPLA3) [24,25], transmembrane 6 superfamily member 2 (TM6SF2) [25] and membrane-bound O-acyltransferase domain containing 7 (MBOAT7) [26], with many more in the pipeline to be identified.

3. Natural History

Clinical data from paired liver biopsies and an analysis of the biopsies from placebo arms of clinical trials have demonstrated that up to 25% of patients with NAFL, a condition which was previously considered “benign”, may also progress to advanced fibrosis [27]. Among those with NASH, up to 35% present fibrosis progression, while 40% remain relatively stable over time [28]. Overall, NAFLD is considered a slow progressive disease, with a one-stage fibrosis progression over 14 years for those with fibrosis stage 1 and over 7 years in patients with advanced fibrosis [29]. However, those with NASH seem to progress more rapidly than those with NAFL [30], with up to 20% of patients with NASH and fibrosis stage 3 progressing to cirrhosis [31,32]. Baseline inflammation status and uncontrolled metabolic risk factors have been suggested as the main contributors to disease progression in these patients [29]. Overall, the yearly cumulative incidence of NASH-related hepato-carcinoma (HCC) is low (1.5–2%) compared to 4% of HCC from chronic viral hepatitis [7,32]. Moreover, recent evidence also suggests that pre-cirrhotic NAFLD may confer an increased risk for HCC, independent of cirrhosis [7] (Figure 2). Interestingly, NAFLD-associated HCC in non-cirrhotics accounts for up to 25–46% of all NAFLD-associated HCC cases, and the incidence is estimated at approximately 0.1 to 1.3 per 1000 patient-years [33,34,35].
The main cause of death among NAFLD patients is cardiovascular disease (CVD) (33% of deaths), followed by extra-hepatic cancer (19% of deaths) and liver-related complications (19% of deaths) [36]. In terms of absolute risk, patients with NAFLD have been shown to have significantly increased risk of all-cause mortality (hazard ratio 2.2, 95%CI 1.2–44), mainly driven by malignancy [36]. However, CVD is still highly prevalent and represents an important cause of mortality and morbidity in these patients [37,38]. Previously published studies have explored clinical, biochemical, and histological variables that could predict mortality in patients with NAFLD, concluding that age and T2DM are strong predictors for adverse events [39]. However, it is now established that fibrosis stage represents the main prognostic factor in this population [40]. Nevertheless, obesity seems to be the main mediator of the increased risk of malignancies in this population, especially with regards to colon and breast cancer [41]. However, there is evidence suggesting that NAFLD may be associated with malignancies also in those with a lean phenotype [42].
Figure 2. Natural history of NAFLD. NAFLD affects almost 25% of the population worldwide. Up to 25% of patients with simple steatosis and up to 35% of patients with NASH may develop cirrhosis [32,43]. Overall, progression rate for one fibrosis stage is 14 years for F1 and 7 years for F3. However, a subgroup (20%) of patients with NASH and F3 may be fast progressors and develop cirrhosis in 2 years. The overall yearly incidence of HCC in those with NASH cirrhosis is 1.5–2%. The incidence of HCC among patients with non-cirrhotic NAFLD is lower, approximately 0.1 to 1.3 per 1000 patient-years [35]. Abbreviations: NAFLD: non-alcoholic fatty liver disease, NASH: non-alcoholic steatohepatitis, F3: fibrosis stage 3, F1: fibrosis stage 1.
Figure 2. Natural history of NAFLD. NAFLD affects almost 25% of the population worldwide. Up to 25% of patients with simple steatosis and up to 35% of patients with NASH may develop cirrhosis [32,43]. Overall, progression rate for one fibrosis stage is 14 years for F1 and 7 years for F3. However, a subgroup (20%) of patients with NASH and F3 may be fast progressors and develop cirrhosis in 2 years. The overall yearly incidence of HCC in those with NASH cirrhosis is 1.5–2%. The incidence of HCC among patients with non-cirrhotic NAFLD is lower, approximately 0.1 to 1.3 per 1000 patient-years [35]. Abbreviations: NAFLD: non-alcoholic fatty liver disease, NASH: non-alcoholic steatohepatitis, F3: fibrosis stage 3, F1: fibrosis stage 1.
Metabolites 13 00536 g002

4. Diagnosis and Staging of NAFLD

Most patients presenting with NAFLD are mainly asymptomatic. Hepatomegaly (liver enlarged in size) is the most common clinical finding on physical examination. As patients progress to advanced liver disease, signs and symptoms related to portal hypertension may become more evident. A diagnosis of NAFLD should be suspected in all patients with at least one component of the metabolic syndrome presenting with evidence of hepatic steatosis on imaging. Notably, NAFLD is still a diagnosis of exclusion of other common causes of liver disease, especially steatogenic medications and chronic alcohol consumption [4,44].
Histology assessed by an expert pathologist remains the gold standard for diagnosing NASH and staging liver disease in NAFLD [4,11,44]. Currently, the interpretation of liver biopsy relies on the use of semi-quantitative scores, such as the NASH clinical research network (NASH CRN) scoring system. When using such scores, disease activity is defined based on steatosis, hepatocyte ballooning and lobular inflammation, while staging is based on the assessment of fibrosis [2]. Nevertheless, obtaining a liver biopsy is expensive, invasive and associated with potential complications (i.e., bleeding, pain). More importantly, considering the high prevalence of the disease, histology cannot be considered in all patients but should be limited to selected sub-groups, such as those at high risk for progressive disease, and/or in the setting of clinical trials [10,45].

5. Non-Invasive Assessment of NAFLD

Given the drawbacks of performing a liver biopsy, there has been an explosive development and use of non-invasive tests (NITs), with significant (fibrosis stage F ≥ 2) and advanced (fibrosis stage F ≥ 3) fibrosis being the main endpoints [44,46]. A large number of blood and imaging-based NITs are now available to use in clinical practice.
Overall, serum NITs are mainly clustered into two groups: direct (or class 1) and indirect (or class 2) biomarkers (Table 1). The direct NITs measure substances that are directly associated with the process of fibrogenesis in the hepatic stellate cells. Conversely, indirect NITs consist of a combination of biochemical tests, such as LFTs, platelet (PLT) count and/or albumin, along with patient demographics, such as age, BMI and/or the presence of T2DM. There are also patented NITs, which come from a combination of class 1 and class 2 biomarkers. As histology is the gold standard to assess liver disease severity, NITs have been developed using liver biopsy as a gold standard. While direct biomarkers usually have a single cut-off with high specificity, indirect biomarkers usually provide two: a low cut-off with high sensitivity (to rule out the disease) and a high cut-off with high specificity (to rule in the disease). The use of each cut-off is mainly dictated by the clinical setting and/or the disease prevalence. If these cut-offs are combined, the number of false positive and false negative tests are usually reduced. However, when applying such a system, a significant sub-group of patients would inevitably fall in the indeterminate-risk group and, therefore, will require further investigations similar to those in the high-risk category. Among others, the Fibrosis-4 score (FIB-4) [47], NAFLD fibrosis score [48] and enhanced liver fibrosis (ELF) score [49] are the most frequently used NIT measures in clinical practice. Such non-invasive markers have now been embedded in the daily assessment of patients at high risk for NAFLD and suggested for screening for fibrosis in this population.
Imaging has also made huge advancements in the field of non-invasive assessment of liver disease in NAFLD patients. Among others, elastographic techniques have revolutionized the management of these patients, combining high sensitivity and specificity for predicting liver fibrosis. Transient elastography (TE) (Fibroscan, Echosens, Paris), acoustic radiation force imaging (ARFI), shear wave elastography and magnetic resonance elastography (MRE) are by far the most popular in the field [50,51]. Specifically, TE employs vibrations of mild amplitude and low frequency, which propagate within the liver. The subsequent pulse-echo acquisitions are reflective of the elastic properties (i.e., stiffness) of the liver, which are expressed in kilopascals (kPa). From a patient’s perspective, TE is painless and rapid (<5 min) and thus highly acceptable. From a clinical perspective, TE provides high accuracy and reproducibility for detecting advanced liver fibrosis [51]. As such, TE has now become the NIT of choice in most liver clinics around the world.
The choice of NIT used in clinical practice is based on different factors, such as the availability and cost of the test/technique as well as the “context of use”. For instance, class 2 biomarkers, which require inexpensive and widely available parameters, can be easily used to predict liver fibrosis in large populations (i.e., primary care). Conversely, sophisticated, time-consuming and expensive techniques like MRE are applied in selected groups of patients and for research purposes (i.e., tertiary care) [52,53].

6. Screening for NAFLD in Primary Care: Current Recommendations

Given the high prevalence and severity of NAFLD in those with metabolic syndrome and type-2 diabetes, there is an expected large burden of undiagnosed NAFLD with advanced fibrosis in the community, and—as such—a major interest in early detection of the disease in primary care [44]. Furthermore, as there is no licensed treatment for the disease, early detection of fibrosis is of the utmost importance in this population. For instance, in the United States, it has been estimated that up to 9 million diabetic patients have NASH, while 4 million are at risk for advanced fibrosis [54]. Similar evidence has been made available for those suffering from obesity [55].
The latest European guidelines recommend screening NAFLD in high-risk populations (i.e., patients with metabolic syndrome) following a two-tier system (Figure 3). Specifically, it is recommended that patients should be stratified using FIB-4 and/or ELF in primary care, followed in sequence by TE in a specialist setting [44]. The most recent guidelines from the America Association for the study of the liver diseases (AASLD) suggest yearly testing with FIB-4 for diabetics and those with at least two components of metabolic syndrome, whilst not recommending screening for NAFLD in the general population [46]. Interestingly, the latest UK NICE guidelines [56] recommend screening for NAFLD subjects with T2DM and metabolic syndrome, including LFTs and/or ultrasound. Similar recommendations are made in the guidelines from the Asian Pacific Association [57] and from the Latin American Association [58] for the study of the liver. However, LFTs assessment is not sufficient alone for screening NAFLD, since it is well established that NASH and significant fibrosis can occur in patients with normal range LFTs [59,60]. Furthermore, ultrasound has low reproducibility and was not designed to stage disease severity [44].
Unfortunately, there is a substantial lack of awareness among policy makers outside the hepatology community. For instance, current diabetes and obesity management guidelines do not advise for NAFLD screening in the respective populations [61]. Nevertheless, the American Association of Clinical Endocrinology has now published a clinical practice guideline on the management of NAFLD in primary care and endocrinology settings, opening the door to future joint position papers across different specialties [62]. Such guidelines highlight that patients at high risk of NAFLD do not require an abdominal ultrasound to diagnose hepatic steatosis; it is recommended to move directly to risk stratification. Finally, European guidelines on obesity care in patients with chronic GI conditions, despite recognising that obese patients should be screened for NAFLD, do not advise fibrosis risk stratification [63].

7. Screening for NAFLD in Primary Care: Limitations

Although screening for NAFLD in high-risk populations has been supported by EASL and AASLD guidelines, a consensus on the cost-effectiveness of screening has not yet been reached. Corey and colleagues performed a simulation to compare quality-adjusted life years (QALYs) between screening with liver biopsy and non-screening, including pioglitazone as therapeutical option. The authors reported that NASH screening could have been cost-effective if superior treatment had been made available at the time of the model [64]. A recent paper suggested that for a pharmacological intervention to be cost-effective in the NAFLD fibrosis population, the annual drug cost should not exceed $12,000 per patient [65]. Several studies tried to analyse the cost-effectiveness of screening by factoring in the effect of early detection in slowing disease progression rather than the therapeutical effect of a new pharmacological agent. A recent cost-utility analysis also demonstrated that screening patients with T2DM with US and LFTs, followed by non-invasive tests, was more cost-effective than not screening [66]. Interestingly, a UK-based study comparing risk stratification using TE vs. standard of care proved to be cost-effective in the general population [67]. Similarly, in a study conducted in the US health system, screening for NAFLD cirrhosis with FIB-4, followed by TE and liver biopsy, was more effective than FIB-4 followed by MRE [68]. Another study demonstrated that FIB-4 followed by share wave elastography was the most effective and least costly strategy in the community [69]. Nevertheless, the lack of licensed pharmacological treatment still represents an important limitation to establishing the cost-effectiveness of screening in this population. Furthermore, a recent metanalysis also pointed out how the currently used health economic models are associated with limitations, primarily driven by a lack of NASH-specific data [70].
Primary care clinicians play an essential role in identifying patients with NAFLD who are at risk of significant liver disease [44]. In this sense, the Lancet commission on liver disease identified the need for streamlined diagnostic pathways for screening people with NAFLD as a priority area to defeat liver disease [71]. However, an important limitation to screening pathways of NAFLD is an overall low awareness among primary care clinicians, possibly as the result of gaps in knowledge as well as lack of awareness of relevant practice guidelines. In a survey study, over 40% of general practitioners (GPs) were not familiar with clinical published guidelines for NAFLD management [72]. Moreover, GPs were more likely to screen low-risk patients while neglecting patients at high risk for liver fibrosis. Again, this phenomenon has been attributed to the misconception that LFTs may reflect disease severity. On a similar note, a UK-based qualitative study demonstrated that the diagnosis and management of NAFLD is perceived as a great challenge by GPs [73,74]. Overall, less than 3% of patients with elevated FIB-4 are currently referred to the specialist setting for further investigations [75], with GPs not perceiving NAFLD as a priority in their clinical activities [76].
Low standardisation of the screening protocols also represents an important limitation to screening NAFLD in the community. Several studies have highlighted a significant gap between guidelines and real-life clinical approaches, not only across different continents [77] but also within Europe [78]. Such inconsistency translates into a lack of clarity for primary care physicians. The use of an automated calculator for NITs as well as easier access to second-line non-invasive tests have been identified as possible strategies to overcome current barriers to screening [76]. Clear guidance on the groups to be screened and the patients to be referred for further tests also appears to be lacking. A previous study carried out in Scotland demonstrated how an algorithm for analysis of abnormal LFTs was found to correctly (in 91.3% of cases) stratify patients for referral to specialist investigation [79]. A similar approach could be potentially useful for identifying those at higher risk of fibrosis from NAFLD. Furthermore, NAFLD screening could be embedded in the routine clinical management of high-risk populations in primary care, such as those with type-2 diabetes [76].
Along with developing cost-effective screening for NAFLD in primary care, future work should also focus on education regarding high-risk stratification, providing easy-to-use tools and building awareness among primary care physicians.

8. Screening for NAFLD in Primary Care: Beware of the Spectrum Effect

Ideally, screening tests should be derived from a cohort that mirrors the target population so that spectrum biases can be minimised [80]. From an epidemiological perspective, the spectrum effect describes the variation in the diagnostic performance of predictive tests when applied to populations with different disease prevalence. Due to the spectrum effect, NITs will have lower sensitivity and higher specificity in populations with lower disease prevalence. On the other hand, in secondary/tertiary care settings (higher disease prevalence), the positive predictive value will be higher, as the probability of observing true positive cases is higher a priori. Among others, NITs based on blood tests and/or on a combination of clinical features seem to be particularly affected by the spectrum bias.
Notably, most of the NITs used for NAFLD screening were historically developed and validated in secondary or tertiary care settings. Their performance in primary care is largely unknown. Specifically, FIB-4 was developed from a cohort of patients with biopsy-proven chronic hepatitis C [81], while the NAFLD fibrosis score and the ELF were developed in a cohort of patients with biopsy-proven NAFLD [48,82]. Of note, in the pre-elastography era, the average biopsy patient was selected based on clinical parameters, mainly on LFTs. It is therefore not surprising that these cohorts were characterised by older age and elevated LFTs, despite these not necessarily being a good marker of the disease severity [60]. It is therefore expected that FIB-4, NAFLD fibrosis and ELF score captures the phenotype of patients referred to specialist clinics.
The EASL algorithm for NAFLD screening has recently been validated into a tertiary care cohort, despite primary care being the main target for the pathway [83]. Interestingly, a recent real-life study showed that up to two-thirds of the new referrals to hepatology clinics are discharged after their first assessment, suggesting that current risk stratification needs optimisation [84]. Furthermore, there is emerging evidence suggesting that FIB-4 accuracy is much lower in the community [85], especially when used to assess young patients with normal liver function tests [85,86]. Moreover, a recent metanalysis also demonstrated that ELF performance is not consistent across studies [87], suggesting that dedicated cut-offs may be needed for different populations. On this note, the most recent AASLD guidelines have highlighted the lack of evidence to support the use of some NITs in primary care, raising concerns about underestimating liver disease, especially among diabetics [46].
Overall, there is increasing evidence suggesting that FIB-4, ELF and NAFLD fibrosis scores may be affected by the spectrum effect. Offering TE to high-risk patients in primary care could represent a way forward, as it is cost-effective and is not affected by spectrum biases [86]. Future work should focus on assessing the performance of NITs in true primary care cohorts and on the optimization of current referral management strategies.

9. Novel Approaches to Diagnosing and Staging NAFLD

In the last decade, several new technologies have been developed with the aim to improve precision medicine in the field of metabolic diseases. Among others, metabolomics has been considered as a potential new approach for diagnosing and staging NAFLD. In a recent study, disease activity, assessed by hepatocellular ballooning and inflammation as per the NASH CRN scoring system, correlated with increased branched chain amino acids and aromatic amino acids. In the same study, a combination of glutamate, serine and glycine could predict fibrosis severity [88]. In another study, NASH could be diagnosed accurately by a combination of glycocholic acid, taurocholic acid, phenylalanine and branched chain amino acids [89].
Similarly, lipidomics has also been explored for the non-invasive assessment of NAFLD [90]. Previous studies demonstrated that a combination of circulating lipids may be able to accurately identify those with NASH [90,91]. Similarly, another algorithm combining genetic variants and serum lipids correlated with hepatic fat fraction, as measured by MRI-PDFF [92]. Finally, among those with biopsy-proven NASH, phosphatidylcholine levels were strongly associated with disease activity, as assessed by the NASH CRN scoring system [93].
Future studies will be required to validate the findings from lipidomic and metabolomic studies on a large scale and against histology. Despite the high cost being a potential limitation to applying these techniques on a large scale, obtaining an accurate, non-invasive diagnosis of NASH may fill a critical gap in clinical practice.

10. Conclusions

Non-alcoholic fatty liver disease poses a significant challenge to the Hepatology community due to increasing burden of the disease. As many promising drugs are in the pipeline, identifying those with advanced fibrosis or at risk of developing advanced fibrosis in the community will become a clinical priority in the near future. Engaging with primary care is crucial, as GPs are at the forefront of identifying patients with NAFLD in need for further evaluation. There is a need for a simple, pragmatic referral/management pathway that performs well in primary care and that could be easily implemented. Future work should focus on the optimisation of current algorithms for NAFLD screening.

Author Contributions

Conceptualization, R.F., writing—original draft, G.S., writing—review and editing, B.H.M., M.Y., P.M., supervision, P.M. All the authors have reviewed and approved the final draft. All authors have read and agreed to the published version of the manuscript.

Funding

The Division of Digestive Diseases receives financial support from the National Institute of Health Research (NIHR) Imperial Biomedical Research Centre (BRC).

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Younossi, Z.M.; Golabi, P.; Paik, J.M.; Henry, A.; Van Dongen, C.; Henry, L. The global epidemiology of nonalcoholic fatty liver disease (NAFLD) and nonalcoholic steatohepatitis (NASH): A systematic review. Hepatology 2023, 77, 1335–1347. [Google Scholar] [CrossRef]
  2. Kleiner, D.E.; Brunt, E.M.; Van Natta, M.; Behling, C.; Contos, M.J.; Cummings, O.W.; Ferrell, L.D.; Liu, Y.C.; Torbenson, M.S.; Unalp-Arida, A.; et al. Design and validation of a histological scoring system for nonalcoholic fatty liver disease. Hepatology 2005, 41, 1313–1321. [Google Scholar] [CrossRef] [PubMed]
  3. Brunt, E.M.; Kleiner, D.E.; Carpenter, D.H.; Rinella, M.; Harrison, S.A.; Loomba, R.; Younossi, Z.; Neuschwander-Tetri, B.A.; Sanyal, A.J.; for the American Association for the Study of Liver Diseases NASH Task Force. NAFLD: Reporting Histologic Findings in Clinical Practice. Hepatology 2020, 73, 2028–2038. [Google Scholar] [CrossRef]
  4. Glen, J.; Floros, L.; Day, C.; Pryke, R.; Guideline Development Group. Non-alcoholic fatty liver disease (NAFLD): Summary of NICE guidance. BMJ 2016, 354, i4428. [Google Scholar] [CrossRef] [PubMed]
  5. Lembo, E.; Russo, M.F.; Verrastro, O.; Anello, D.; Angelini, G.; Iaconelli, A.; Guidone, C.; Stefanizzi, G.; Ciccoritti, L.; Greco, F.; et al. Prevalence and predictors of non-alcoholic steatohepatitis in subjects with morbid obesity and with or without type 2 diabetes. Diabetes Metab. 2022, 48, 101363. [Google Scholar] [CrossRef]
  6. Younossi, Z.; Tacke, F.; Arrese, M.; Sharma, B.C.; Mostafa, I.; Bugianesi, E.; Wong, V.W.-S.; Yilmaz, Y.; George, J.; Fan, J.; et al. Global Perspectives on Nonalcoholic Fatty Liver Disease and Nonalcoholic Steatohepatitis. Hepatology 2019, 69, 2672–2682. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  7. Estes, C.; Razavi, H.; Loomba, R.; Younossi, Z.; Sanyal, A.J. Modeling the epidemic of nonalcoholic fatty liver disease demonstrates an exponential increase in burden of disease. Hepatology 2018, 67, 123–133. [Google Scholar] [CrossRef] [Green Version]
  8. Younossi, Z.; Stepanova, M.; Ong, J.P.; Jacobson, I.M.; Bugianesi, E.; Duseja, A.; Eguchi, Y.; Wong, V.W.; Negro, F.; Yilmaz, Y.; et al. Nonalcoholic Steatohepatitis Is the Fastest Growing Cause of Hepatocellular Carcinoma in Liver Transplant Candidates. Clin. Gastroenterol. Hepatol. 2018, 17, 748–755.e3. [Google Scholar] [CrossRef] [Green Version]
  9. Hardy, T.; Wonders, K.; Younes, R.; Aithal, G.P.; Aller, R.; Allison, M.; Bedossa, P.; Betsou, F.; Boursier, J.; Brosnan, M.J.; et al. The European NAFLD Registry: A real-world longitudinal cohort study of nonalcoholic fatty liver disease. Contemp. Clin. Trials 2020, 98, 106175. [Google Scholar] [CrossRef]
  10. Kleiner, D.E.; Makhlouf, H.R. Histology of Nonalcoholic Fatty Liver Disease and Nonalcoholic Steatohepatitis in Adults and Children. Clin. Liver Dis. 2016, 20, 293–312. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  11. Bugianesi, E.; Moscatiello, S.; Ciaravella, M.F.; Marchesini, G. Insulin resistance in nonalcoholic fatty liver disease. Curr. Pharm. Des. 2010, 16, 1941–1951. [Google Scholar] [CrossRef] [PubMed]
  12. Neuschwander-Tetri, B.A. Hepatic lipotoxicity and the pathogenesis of nonalcoholic steatohepatitis: The central role of nontriglyceride fatty acid metabolites. Hepatology 2010, 52, 774–788. [Google Scholar] [CrossRef] [PubMed]
  13. Guilherme, A.; Virbasius, J.V.; Puri, V.; Czech, M.P. Adipocyte dysfunctions linking obesity to insulin resistance and type 2 diabetes. Nat. Rev. Mol. Cell Biol. 2008, 9, 367–377. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  14. Salehi-Sahlabadi, A.; Sadat, S.; Beigrezaei, S.; Pourmasomi, M.; Feizi, A.; Ghiasvand, R.; Hadi, A.; Clark, C.C.T.; Miraghajani, M. Dietary patterns and risk of non-alcoholic fatty liver disease. BMC Gastroenterol. 2021, 21, 41. [Google Scholar] [CrossRef] [PubMed]
  15. Fu, J.; Shin, S. Dietary patterns and risk of non-alcoholic fatty liver disease in Korean adults: A prospective cohort study. BMJ Open 2023, 13, e065198. [Google Scholar] [CrossRef]
  16. Bergheim, I.; Weber, S.; Vos, M.; Krämer, S.; Volynets, V.; Kaserouni, S.; McClain, C.J.; Bischoff, S.C. Antibiotics protect against fructose-induced hepatic lipid accumulation in mice: Role of endotoxin. J. Hepatol. 2008, 48, 983–992. [Google Scholar] [CrossRef] [PubMed]
  17. Ma, J.; Fox, C.S.; Jacques, P.F.; Speliotes, E.K.; Hoffmann, U.; Smith, C.E.; Saltzman, E.; McKeown, N.M. Sugar-sweetened beverage, diet soda, and fatty liver disease in the Framingham Heart Study cohorts. J. Hepatol. 2015, 63, 462–469. [Google Scholar] [CrossRef] [Green Version]
  18. Zelber-Sagi, S.; Ivancovsky-Wajcman, D.; Isakov, N.F.; Webb, M.; Orenstein, D.; Shibolet, O.; Kariv, R. High red and processed meat consumption is associated with non-alcoholic fatty liver disease and insulin resistance. J. Hepatol. 2018, 68, 1239–1246. [Google Scholar] [CrossRef] [PubMed]
  19. Hu, H.; Lin, A.; Kong, M.; Yao, X.; Yin, M.; Xia, H.; Ma, J.; Liu, H. Intestinal microbiome and NAFLD: Molecular insights and therapeutic perspectives. J. Gastroenterol. 2019, 55, 142–158. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  20. Hoyles, L.; Fernández-Real, J.-M.; Federici, M.; Serino, M.; Abbott, J.; Charpentier, J.; Heymes, C.; Luque, J.L.; Anthony, E.; Barton, R.H.; et al. Molecular phenomics and metagenomics of hepatic steatosis in non-diabetic obese women. Nat. Med. 2018, 24, 1070–1080. [Google Scholar] [CrossRef]
  21. Forlano, R.; Mullish, B.H.; Roberts, L.A.; Thursz, M.R.; Manousou, P. The Intestinal Barrier and Its Dysfunction in Patients with Metabolic Diseases and Non-Alcoholic Fatty Liver Disease. Int. J. Mol. Sci. 2022, 23, 662. [Google Scholar] [CrossRef]
  22. Soppert, J.; Brandt, E.F.; Heussen, N.M.; Barzakova, E.; Blank, L.M.; Kuepfer, L.; Hornef, M.W.; Trebicka, J.; Jankowski, J.; Berres, M.-L.; et al. Blood Endotoxin Levels as Biomarker of Nonalcoholic Fatty Liver Disease: A Systematic Review and Meta-analysis. Clin. Gastroenterol. Hepatol. 2022. [Google Scholar] [CrossRef]
  23. Kirpich, I.A.; Marsano, L.S.; McClain, C.J. Gut-liver axis, nutrition, and non-alcoholic fatty liver disease. Clin. Biochem. 2015, 48, 923–930. [Google Scholar] [CrossRef] [Green Version]
  24. Chen, V.L.; Oliveri, A.; Miller, M.J.; Wijarnpreecha, K.; Du, X.; Chen, Y.; Cushing, K.C.; Lok, A.S.; Speliotes, E.K. PNPLA3 Genotype and Diabetes Identify Patients with Nonalcoholic Fatty Liver Disease at High Risk of Incident Cirrhosis. Gastroenterology 2023. [Google Scholar] [CrossRef] [PubMed]
  25. Holmer, M.; Ekstedt, M.; Nasr, P.; Zenlander, R.; Wester, A.; Tavaglione, F.; Romeo, S.; Kechagias, S.; Stål, P.; Hagström, H. Effect of common genetic variants on the risk of cirrhosis in non-alcoholic fatty liver disease during 20 years of follow-up. Liver Int. 2022, 42, 2769–2780. [Google Scholar] [CrossRef]
  26. Buzzetti, E.; Pinzani, M.; Tsochatzis, E.A. The multiple-hit pathogenesis of non-alcoholic fatty liver disease (NAFLD). Metabolism 2016, 65, 1038–1048. [Google Scholar] [CrossRef] [PubMed]
  27. McPherson, S.; Hardy, T.; Henderson, E.; Burt, A.D.; Day, C.P.; Anstee, Q.M. Evidence of NAFLD progression from steatosis to fibrosing-steatohepatitis using paired biopsies: Implications for prognosis and clinical management. J. Hepatol. 2015, 62, 1148–1155. [Google Scholar] [CrossRef]
  28. Schwabe, R.F.; Tabas, I.; Pajvani, U.B. Mechanisms of Fibrosis Development in Nonalcoholic Steatohepatitis. Gastroenterology 2020, 158, 1913–1928. [Google Scholar] [CrossRef]
  29. Pais, R.; Charlotte, F.; Fedchuk, L.; Bedossa, P.; Lebray, P.; Poynard, T.; Ratziu, V.; LIDO Study Group. A systematic review of follow-up biopsies reveals disease progression in patients with non-alcoholic fatty liver. J. Hepatol. 2013, 59, 550–556. [Google Scholar] [CrossRef]
  30. Chalasani, N.; Younossi, Z.; Lavine, J.E.; Diehl, A.M.; Brunt, E.M.; Cusi, K.; Charlton, M.; Sanyal, A.J. The diagnosis and management of non-alcoholic fatty liver disease: Practice Guideline by the American Association for the Study of Liver Diseases, American College of Gastroenterology, and the American Gastroenterological Association. Hepatology 2012, 55, 2005–2023. [Google Scholar] [CrossRef] [PubMed]
  31. Torres, D.M.; Williams, C.D.; Harrison, S.A. Features, diagnosis, and treatment of nonalcoholic fatty liver disease. Clin. Gastroenterol. Hepatol. 2012, 10, 837–858. [Google Scholar] [CrossRef]
  32. Ascha, M.S.; Hanouneh, I.A.; Lopez, R.; Tamimi, T.A.; Feldstein, A.F.; Zein, N.N. The incidence and risk factors of hepatocellular carcinoma in patients with nonalcoholic steatohepatitis. Hepatology 2010, 51, 1972–1978. [Google Scholar] [CrossRef] [PubMed]
  33. Dyson, J.; Jaques, B.; Chattopadyhay, D.; Lochan, R.; Graham, J.; Das, D.; Aslam, T.; Patanwala, I.; Gaggar, S.; Cole, M.; et al. Hepatocellular cancer: The impact of obesity, type 2 diabetes and a multidisciplinary team. J. Hepatol. 2014, 60, 110–117. [Google Scholar] [CrossRef]
  34. Piscaglia, F.; Svegliati-Baroni, G.; Barchetti, A.; Pecorelli, A.; Marinelli, S.; Tiribelli, C.; Bellentani, S.; HCC-NAFLD Italian Study Group. Clinical patterns of hepatocellular carcinoma in nonalcoholic fatty liver disease: A multicenter prospective study. Hepatology 2016, 63, 827–838. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  35. Pinyopornpanish, K.; Khoudari, G.; Saleh, M.A.; Angkurawaranon, C.; Pinyopornpanish, K.; Mansoor, E.; Dasarathy, S.; McCullough, A. Hepatocellular carcinoma in nonalcoholic fatty liver disease with or without cirrhosis: A population-based study. BMC Gastroenterol. 2021, 21, 394. [Google Scholar] [CrossRef] [PubMed]
  36. A Adams, L.; Harmsen, S.; Sauver, J.S.; Charatcharoenwitthaya, P.; Enders, F.; Therneau, T.; Angulo, P. Nonalcoholic Fatty Liver Disease Increases Risk of Death Among Patients With Diabetes: A Community-Based Cohort Study. Am. J. Gastroenterol. 2010, 105, 1567–1573. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  37. Abeles, R.D.; Mullish, B.H.; Forlano, R.; Kimhofer, T.; Adler, M.; Tzallas, A.; Giannakeas, N.; Yee, M.; Mayet, J.; Goldin, R.D.; et al. Derivation and validation of a cardiovascular risk score for prediction of major acute cardiovascular events in non-alcoholic fatty liver disease; the importance of an elevated mean platelet volume. Aliment. Pharm. Ther. 2019, 49, 1077–1085. [Google Scholar] [CrossRef] [Green Version]
  38. Targher, G.; Byrne, C.D.; Lonardo, A.; Zoppini, G.; Barbui, C. Non-alcoholic fatty liver disease and risk of incident cardiovascular disease: A meta-analysis. J. Hepatol. 2016, 65, 589–600. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  39. A Adams, L.; Roberts, S.K.; I Strasser, S.; E Mahady, S.; Powell, E.; Estes, C.; Razavi, H.; George, J. Nonalcoholic fatty liver disease burden: Australia, 2019–2030. J. Gastroenterol. Hepatol. 2020, 35, 1628–1635. [Google Scholar] [CrossRef] [Green Version]
  40. Ekstedt, M.; Hagström, H.; Nasr, P.; Fredrikson, M.; Stål, P.; Kechagias, S.; Hultcrantz, R. Fibrosis stage is the strongest predictor for disease-specific mortality in NAFLD after up to 33 years of follow-up. Hepatology 2015, 61, 1547–1554. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  41. Veracruz, N.; Hameed, B.; Saab, S.; Wong, R.J. The Association Between Nonalcoholic Fatty Liver Disease and Risk of Cardiovascular Disease, Stroke, and Extrahepatic Cancers. J. Clin. Exp. Hepatol. 2021, 11, 45–81. [Google Scholar] [CrossRef] [PubMed]
  42. Chen, N.; Zhou, J.; Wang, K.; Li, X.; Li, Z. Non-obese or lean non-alcoholic fatty liver disease was associated with increased risk of cancer in patients with type 2 diabetes mellitus. BMJ Open Diabetes Res. Care 2023, 11, e003066. [Google Scholar] [CrossRef]
  43. Starley, B.Q.; Calcagno, C.J.; Harrison, S.A. Nonalcoholic fatty liver disease and hepatocellular carcinoma: A weighty connection. Hepatology 2010, 51, 1820–1832. [Google Scholar] [CrossRef]
  44. European Association for the Study of the Liver. Electronic address: Easloffice@easloffice.eu; Clinical Practice Guideline Panel; Chair; EASL Governing Board representative; Panel members. EASL Clinical Practice Guidelines on non-invasive tests for evaluation of liver disease severity and prognosis–2021 update. J. Hepatol. 2021, 75, 659–689. [Google Scholar] [CrossRef]
  45. Nalbantoglu, I.L.; Brunt, E.M. Role of liver biopsy in nonalcoholic fatty liver disease. World J. Gastroenterol. 2014, 20, 9026–9037. [Google Scholar] [PubMed]
  46. Rinella, M.E.; Neuschwander-Tetri, B.A.; Siddiqui, M.S.; Abdelmalek, M.F.; Caldwell, S.; Barb, D.; Kleiner, D.E.; Loomba, R. AASLD Practice Guidance on the clinical assessment and management of nonalcoholic fatty liver disease. Hepatology 2023. Publish Ah. [Google Scholar] [CrossRef] [PubMed]
  47. Vallet-Pichard, A.; Mallet, V.; Nalpas, B.; Verkarre, V.; Nalpas, A.; Dhalluin-Venier, V.; Fontaine, H.; Pol, S. FIB-4: An inexpensive and accurate marker of fibrosis in HCV infection. comparison with liver biopsy and fibrotest. Hepatology 2007, 46, 32–36. [Google Scholar] [CrossRef]
  48. Angulo, P.; Hui, J.M.; Marchesini, G.; Bugianesi, E.; George, J.; Farrell, G.C.; Enders, F.; Saksena, S.; Burt, A.D.; Bida, J.P.; et al. The NAFLD fibrosis score: A noninvasive system that identifies liver fibrosis in patients with NAFLD. Hepatology 2007, 45, 846–854. [Google Scholar] [CrossRef]
  49. Rosenberg, W.M.; Voelker, M.; Thiel, R.; Becka, M.; Burt, A.; Schuppan, D.; Hubscher, S.; Roskams, T.; Pinzani, M.; Arthur, M.J.; et al. Serum markers detect the presence of liver fibrosis: A cohort study. Gastroenterology 2004, 127, 1704–1713. [Google Scholar] [CrossRef] [Green Version]
  50. Cassinotto, C.; Boursier, J.; de Lédinghen, V.; Lebigot, J.; Lapuyade, B.; Cales, P.; Hiriart, J.B.; Michalak, S.; Bail, B.L.; Cartier, V.; et al. Liver stiffness in nonalcoholic fatty liver disease: A comparison of supersonic shear imaging, FibroScan, and ARFI with liver biopsy. Hepatology 2016, 63, 1817–1827. [Google Scholar] [CrossRef] [Green Version]
  51. European Association for Study of Liver; Asociacion Latinoamericana para el Estudio del Higado. EASL-ALEH Clinical Practice Guidelines: Non-invasive tests for evaluation of liver disease severity and prognosis. J. Hepatol. 2015, 63, 237–264. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  52. Crossan, C.; A Tsochatzis, E.; Longworth, L.; Gurusamy, K.; Davidson, B.; Rodríguez-Perálvarez, M.; Mantzoukis, K.; O’Brien, J.; Thalassinos, E.; Papastergiou, V.; et al. Cost-effectiveness of non-invasive methods for assessment and monitoring of liver fibrosis and cirrhosis in patients with chronic liver disease: Systematic review and economic evaluation. Health Technol. Assess. 2015, 19, 1–410. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  53. Han, M.A.T.; Saouaf, R.; Ayoub, W.; Todo, T.; Mena, E.; Noureddin, M. Magnetic resonance imaging and transient elastography in the management of Nonalcoholic Fatty Liver Disease (NAFLD). Expert Rev. Clin. Pharmacol. 2017, 10, 379–390. [Google Scholar] [CrossRef] [PubMed]
  54. Barbosa, J.V.; Lai, M. Nonalcoholic Fatty Liver Disease Screening in Type 2 Diabetes Mellitus Patients in the Primary Care Setting. Hepatol. Commun. 2020, 5, 158–167. [Google Scholar] [CrossRef]
  55. Kim, Y.; Chang, Y.; Cho, Y.K.; Ahn, J.; Shin, H.; Ryu, S. Obesity and Weight Gain Are Associated with Progression of Fibrosis in Patients With Nonalcoholic Fatty Liver Disease. Clin. Gastroenterol. Hepatol. 2018, 17, 543–550.e2. [Google Scholar] [CrossRef] [PubMed]
  56. National Guideline Centre (UK). Non-Alcoholic Fatty Liver Disease: Assessment and Management; National Institute for Health and Care Excellence (NICE): London, UK, 2016. [Google Scholar]
  57. Eslam, M.; Sarin, S.K.; Wong, V.W.S.; Fan, J.G.; Kawaguchi, T.; Ahn, S.H.; Zheng, M.H.; Shiha, G.; Yilmaz, Y.; Gani, R.; et al. The Asian Pacific Association for the Study of the Liver clinical practice guidelines for the diagnosis and management of metabolic associated fatty liver disease. Hepatol. Int. 2020, 14, 889–919. [Google Scholar] [CrossRef] [PubMed]
  58. Arab, J.P.; Dirchwolf, M.; Álvares-da-Silva, M.R.; Barrera, F.; Benítez, C.; Castellanos-Fernandez, M.; Castro-Narro, G.; Chavez-Tapia, N.; Chiodi, D.; Cotrim, H.; et al. Latin American Association for the study of the liver (ALEH) practice guidance for the diagnosis and treatment of non-alcoholic fatty liver disease. Ann. Hepatol. 2020, 19, 674–690. [Google Scholar] [CrossRef]
  59. Ma, X.; Liu, S.; Zhang, J.; Dong, M.; Wang, Y.; Wang, M.; Xin, Y. Proportion of NAFLD patients with normal ALT value in overall NAFLD patients: A systematic review and meta-analysis. BMC Gastroenterol. 2020, 20, 10. [Google Scholar] [CrossRef] [Green Version]
  60. Forlano, R.; Mullish, B.H.; Dhar, A.; Goldin, R.D.; Thursz, M.; Manousou, P. Liver function tests and metabolic-associated fatty liver disease: Changes in upper normal limits, does it really matter? World J. Hepatol. 2021, 13, 2104–2112. [Google Scholar] [CrossRef]
  61. Davies, M.J.; D’Alessio, D.A.; Fradkin, J.; Kernan, W.N.; Mathieu, C.; Mingrone, G.; Rossing, P.; Tsapas, A.; Wexler, D.J.; Buse, J.B. Management of hyperglycaemia in type 2 diabetes, 2018. A consensus report by the American Diabetes Association (ADA) and the European Association for the Study of Diabetes (EASD). Diabetologia 2018, 61, 2461–2498. [Google Scholar] [CrossRef] [Green Version]
  62. Cusi, K.; Isaacs, S.; Barb, D.; Basu, R.; Caprio, S.; Garvey, W.T.; Kashyap, S.; Mechanick, J.I.; Mouzaki, M.; Nadolsky, K. American Association of Clinical Endocrinology Clinical Practice Guideline for the Diagnosis and Management of Nonalcoholic Fatty Liver Disease in Primary Care and Endocrinology Clinical Settings: Co-Sponsored by the American Association for the Study of Liver Diseases (AASLD). Endocr. Pract. 2022, 28, 528–562. [Google Scholar]
  63. Bischoff, S.C.; Barazzoni, R.; Busetto, L.; Campmans-Kuijpers, M.; Cardinale, V.; Chermesh, I.; Eshraghian, A.; Kani, H.T.; Khannoussi, W.; Lacaze, L.; et al. European guideline on obesity care in patients with gastrointestinal and liver diseases–Joint ESPEN/UEG guideline. Clin. Nutr. 2022, 41, 2364–2405. [Google Scholar] [CrossRef]
  64. Corey, K.E.; Kartoun, U.; Zheng, H.; Shaw, S.Y. Development and Validation of an Algorithm to Identify Nonalcoholic Fatty Liver Disease in the Electronic Medical Record. Dig. Dis. Sci. 2015, 61, 913–919. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  65. Rustgi, V.K.; Duff, S.B.; Elsaid, M.I. Cost-effectiveness and potential value of pharmaceutical treatment of nonalcoholic fatty liver disease. J. Med. Econ. 2022, 25, 347–355. [Google Scholar] [CrossRef] [PubMed]
  66. Noureddin, M.; Jones, C.; Alkhouri, N.; Gomez, E.V.; Dieterich, D.T.; Rinella, M.E.; NASHNET. Screening for Nonalcoholic Fatty Liver Disease in Persons with Type 2 Diabetes in the United States Is Cost-effective: A Comprehensive Cost-Utility Analysis. Gastroenterology 2020, 159, 1985–1987.e4. [Google Scholar] [CrossRef] [PubMed]
  67. Tanajewski, L.; Harris, R.; Harman, D.J.; Aithal, G.P.; Card, T.R.; Gkountouras, G.; Berdunov, V.; Guha, I.N.; Elliott, R.A. Economic evaluation of a community-based diagnostic pathway to stratify adults for non-alcoholic fatty liver disease: A Markov model informed by a feasibility study. BMJ Open 2017, 7, e015659. [Google Scholar] [CrossRef] [Green Version]
  68. Vilar-Gomez, E.; Lou, Z.; Kong, N.; Vuppalanchi, R.; Imperiale, T.F.; Chalasani, N. Cost Effectiveness of Different Strategies for Detecting Cirrhosis in Patients With Nonalcoholic Fatty Liver Disease Based on United States Health Care System. Clin. Gastroenterol. Hepatol. 2020, 18, 2305–2314.e12. [Google Scholar] [CrossRef] [PubMed]
  69. Congly, S.E.; Shaheen, A.A.; Swain, M.G. Modelling the cost effectiveness of non-alcoholic fatty liver disease risk stratification strategies in the community setting. PLoS ONE 2021, 16, e0251741. [Google Scholar] [CrossRef]
  70. Johansen, P.; Howard, D.; Bishop, R.; Moreno, S.I.; Buchholtz, K. Systematic Literature Review and Critical Appraisal of Health Economic Models Used in Cost-Effectiveness Analyses in Non-Alcoholic Steatohepatitis: Potential for Improvements. Pharmacoeconomics 2020, 38, 485–497. [Google Scholar] [CrossRef] [Green Version]
  71. Williams, R.; Aspinall, R.; Bellis, M.; Camps-Walsh, G.; Cramp, M.; Dhawan, A.; Ferguson, J.; Forton, D.; Foster, G.; Gilmore, I.; et al. Addressing liver disease in the UK: A blueprint for attaining excellence in health care and reducing premature mortality from lifestyle issues of excess consumption of alcohol, obesity, and viral hepatitis. Lancet 2014, 384, 1953–1997. [Google Scholar] [CrossRef]
  72. Said, A.; Gagovic, V.; Malecki, K.; Givens, M.L.; Nieto, F.J. Primary care practitioners survey of non-alcoholic fatty liver disease. Ann. Hepatol. 2013, 12, 758–765. [Google Scholar] [CrossRef]
  73. Standing, H.C.; Jarvis, H.; Orr, J.; Exley, C.; Hudson, M.; Kaner, E.; Hanratty, B. GPs’ experiences and perceptions of early detection of liver disease: A qualitative study in primary care. Br. J. Gen. Pract. 2018, 68, e743–e749. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  74. Islam, K.B.; Brandman, D.; Chu, J.N.; Goldman, M.L.; Fox, R.K. Primary Care Providers and Nonalcoholic Fatty Liver Disease: A Needs Assessment Survey. Dig. Dis. Sci. 2022, 68, 434–438. [Google Scholar] [CrossRef] [PubMed]
  75. Spann, A.; Bishop, K.M.; Weitkamp, A.O.; Stenner, S.P.; Nelson, S.D.; Izzy, M. Clinical decision support automates care gap detection among primary care patients with nonalcoholic fatty liver disease. Hepatol. Commun. 2023, 7, e0035. [Google Scholar] [CrossRef]
  76. Gracen, L.; Hayward, K.L.; Aikebuse, M.; Williams, S.; Russell, A.; O’Beirne, J.; Powell, E.E.; Valery, P.C. An exploration of barriers and facilitators to implementing a nonalcoholic fatty liver disease pathway for people with type 2 diabetes in primary care. Diabet. Med. 2022, 39, e14799. [Google Scholar] [CrossRef] [PubMed]
  77. Anstee, Q.M.; Hallsworth, K.; Lynch, N.; Hauvespre, A.; Mansour, E.; Kozma, S.; Marino, J.P.; Bottomley, J.; Piercy, J.; Higgins, V. Real-world management of non-alcoholic steatohepatitis differs from clinical practice guideline recommendations and across regions. JHEP Rep. 2022, 4, 100411. [Google Scholar] [CrossRef] [PubMed]
  78. Ratziu, V.; Anstee, Q.M.; Wong, V.W.; Schattenberg, J.M.; Bugianesi, E.; Augustin, S.; Gheorghe, L.; Zambon, V.; Reau, N. An international survey on patterns of practice in NAFLD and expectations for therapies-The POP-NEXT project. Hepatology 2022, 76, 1766–1777. [Google Scholar] [CrossRef] [PubMed]
  79. Miller, M.H.; Fraser, A.; Leggett, G.; Macgilchrist, A.; Gibson, G.; Orr, J.; Forrest, E.H.; Dow, E.; Bartlett, W.; Weatherburn, C.; et al. Development and validation of diagnostic triage criteria for liver disease from a minimum data set enabling the ‘intelligent LFT’ pathway for the automated assessment of deranged liver enzymes. Front. Gastroenterol. 2018, 9, 175–182. [Google Scholar] [CrossRef]
  80. A Usher-Smith, J.; Sharp, S.J.; Griffin, S.J. The spectrum effect in tests for risk prediction, screening, and diagnosis. BMJ 2016, 353, i3139. [Google Scholar] [CrossRef] [Green Version]
  81. Vallet-Pichard, A.; Mallet, V.; Pol, S. FIB-4: A simple, inexpensive and accurate marker of fibrosis in HCV-infected patients. Hepatology 2006, 44, 769. [Google Scholar] [CrossRef]
  82. Guha, I.N.; Parkes, J.; Roderick, P.; Chattopadhyay, D.; Cross, R.; Harris, S.; Kaye, P.; Burt, A.D.; Ryder, S.D.; Aithal, G.P.; et al. Noninvasive markers of fibrosis in nonalcoholic fatty liver disease: Validating the European Liver Fibrosis Panel and exploring simple markers. Hepatology 2008, 47, 455–460. [Google Scholar] [CrossRef]
  83. Canivet, C.M.; Costentin, C.; Irvine, K.M.; Delamarre, A.; Lannes, A.; Sturm, N.; Oberti, F.; Patel, P.J.; Decaens, T.; Irles-Depé, M.; et al. Validation of the new 2021 EASL algorithm for the noninvasive diagnosis of advanced fibrosis in NAFLD. Hepatology 2023, 77, 920–930. [Google Scholar] [CrossRef]
  84. Elangovan, H.; Rajagopaul, S.; Williams, S.M.; McKillen, B.; Britton, L.; McPhail, S.M.; Horsfall, L.U.; Valery, P.C.; Hayward, K.L.; Powell, E.E. Nonalcoholic Fatty Liver Disease: Interface Between Primary Care and Hepatology Clinics. Hepatol. Commun. 2020, 4, 518–526. [Google Scholar] [CrossRef] [Green Version]
  85. Graupera, I.; Thiele, M.; Serra-Burriel, M.; Caballeria, L.; Roulot, D.; Wong, G.L.-H.; Fabrellas, N.; Guha, I.N.; Arslanow, A.; Expósito, C.; et al. Low Accuracy of FIB-4 and NAFLD Fibrosis Scores for Screening for Liver Fibrosis in the Population. Clin. Gastroenterol. Hepatol. 2021, 20, 2567–2576.e6. [Google Scholar] [CrossRef] [PubMed]
  86. Forlano, R.; Jayawardana, S.; Mullish, B.; Yee, M.; Mossialos, E.; Goldin, R.; Petta, S.; Tsochatzis, E.; Thursz, M.; Manousou, P. Clinical and Cost-Effectiveness Analysis of Community-Based Screening Strategies for Non-Alcoholic Fatty Liver Disease in Patients with Type-2 Diabetes Mellitus. Available online: https://www.researchsquare.com/article/rs-2135338/v1 (accessed on 12 March 2023).
  87. Hinkson, A.; Lally, H.; Gibson, H.; Jones, R.; Rowe, I.A.; Shinkins, B.; Parker, R. Meta-analysis: Enhanced liver fibrosis test to identify hepatic fibrosis in chronic liver diseases. Aliment. Pharmacol. Ther. 2023, 57, 750–762. [Google Scholar] [CrossRef]
  88. Gaggini, M.; Carli, F.; Bugianesi, E.; Gastaldelli, A.; Rosso, C.; Buzzigoli, E.; Marietti, M.; Della Latta, V.; Ciociaro, D.; Abate, M.L.; et al. Altered amino acid concentrations in NAFLD: Impact of obesity and insulin resistance. Hepatology 2018, 67, 145–158. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  89. Masarone, M.; Troisi, J.; Aglitti, A.; Torre, P.; Colucci, A.; Dallio, M.; Federico, A.; Balsano, C.; Persico, M. Untargeted metabolomics as a diagnostic tool in NAFLD: Discrimination of steatosis, steatohepatitis and cirrhosis. Metabolomics 2021, 17, 1–13. [Google Scholar] [CrossRef]
  90. Erario, M.d.l.Á.; Croce, E.; Moviglia Brandolino, M.T.; Moviglia, G.; Grangeat, A.M. Ozone as Modulator of Resorption and Inflammatory Response in Extruded Nucleus Pulposus Herniation. Revising Concepts. Int. J. Mol. Sci. 2021, 22, 9946. [Google Scholar] [CrossRef]
  91. Polyzos, S.A.; Kountouras, J.; Mantzoros, C.S. Obesity and nonalcoholic fatty liver disease: From pathophysiology to therapeutics. Metabolism 2019, 92, 82–97. [Google Scholar] [CrossRef] [PubMed]
  92. Perez-Diaz-Del-Campo, N.; Riezu-Boj, J.; Marin-Alejandre, B.; Monreal, J.; Elorz, M.; Herrero, J.; Benito-Boillos, A.; Milagro, F.; Tur, J.; Abete, I.; et al. Three Different Genetic Risk Scores Based on Fatty Liver Index, Magnetic Resonance Imaging and Lipidomic for a Nutrigenetic Personalized Management of NAFLD: The Fatty Liver in Obesity Study. Diagnostics 2021, 11, 1083. [Google Scholar] [CrossRef]
  93. Ogawa, Y.; Kobayashi, T.; Honda, Y.; Kessoku, T.; Tomeno, W.; Imajo, K.; Nakahara, T.; Oeda, S.; Nagaoki, Y.; Amano, Y.; et al. Metabolomic/lipidomic-based analysis of plasma to diagnose hepatocellular ballooning in patients with non-alcoholic fatty liver disease: A multicenter study. Hepatol. Res. 2020. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Risk factors for development and progression of NAFLD. This figure illustrates the most common risk factors associated with the development and progression of NAFLD. Abbreviations: NAFLD: non-alcoholic fatty liver disease, BA: bile acid, FXR: farnesoid X receptor, LPS: lipopolysaccharide, TLR: toll-like receptor, DNL: de novo lipogenesis, T2DM: type 2 diabetes mellitus, IR: insulin resistance, PNPLA3: patatin-like phospholipase domain containing protein 3, TM6SF2: Transmembrane 6 superfamily member 2, MBOAT7: membrane-bound O-acyltransferase domain containing 7.
Figure 1. Risk factors for development and progression of NAFLD. This figure illustrates the most common risk factors associated with the development and progression of NAFLD. Abbreviations: NAFLD: non-alcoholic fatty liver disease, BA: bile acid, FXR: farnesoid X receptor, LPS: lipopolysaccharide, TLR: toll-like receptor, DNL: de novo lipogenesis, T2DM: type 2 diabetes mellitus, IR: insulin resistance, PNPLA3: patatin-like phospholipase domain containing protein 3, TM6SF2: Transmembrane 6 superfamily member 2, MBOAT7: membrane-bound O-acyltransferase domain containing 7.
Metabolites 13 00536 g001
Figure 3. Screening for NAFLD in primary care. International guidelines recommend screening for NAFLD in high-risk populations (i.e., patients with metabolic syndrome) following a two-tier system.
Figure 3. Screening for NAFLD in primary care. International guidelines recommend screening for NAFLD in high-risk populations (i.e., patients with metabolic syndrome) following a two-tier system.
Metabolites 13 00536 g003
Table 1. Overview of the main non-invasive tests for NAFLD based on blood tests and clinical parameters.
Table 1. Overview of the main non-invasive tests for NAFLD based on blood tests and clinical parameters.
Direct serum markersHyaluronate
Laminin
Chitinase-like protein 40 (YKL-40)
Procollagen type I carboxy-terminal peptide (PICP)
Procollagen type III amino-terminal peptide (PIIINP)
Metalloproteinases (MMP)-1 and MMP-2
Tissue inhibitors of the metalloproteinases (TIMPs)
Transforming growth factor β1 (TGF-β1)
Microfibril-associated glycoprotein 4 (MFAP-4)
Indirect serum markersAST/ALT ratio
Prothrombin time, γ-glutamyl transferase and apolipoprotein A1 (PGA)
APRI
Forns index
Fibrosis-4 (FIB-4)
Lok index
Fibrosis probability index (FPI)
NAFLD fibrosis score
BARD
Gamma-glutamyl transferase (GGT) to platelet (PLT) ratio
Patented serum markersFibrotest
Fibroindex
Hepascore
Fibrospect
Enhanced liver fibrosis (ELF) test
Fibrometers
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Forlano, R.; Sigon, G.; Mullish, B.H.; Yee, M.; Manousou, P. Screening for NAFLD—Current Knowledge and Challenges. Metabolites 2023, 13, 536. https://doi.org/10.3390/metabo13040536

AMA Style

Forlano R, Sigon G, Mullish BH, Yee M, Manousou P. Screening for NAFLD—Current Knowledge and Challenges. Metabolites. 2023; 13(4):536. https://doi.org/10.3390/metabo13040536

Chicago/Turabian Style

Forlano, Roberta, Giordano Sigon, Benjamin H. Mullish, Michael Yee, and Pinelopi Manousou. 2023. "Screening for NAFLD—Current Knowledge and Challenges" Metabolites 13, no. 4: 536. https://doi.org/10.3390/metabo13040536

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop