Clin Mol Hepatol > Volume 29(Suppl); 2023 > Article
Shi and Fan: Surveillance of the progression and assessment of treatment endpoints for nonalcoholic steatohepatitis

ABSTRACT

Nonalcoholic steatohepatitis (NASH) is an aggressive form of nonalcoholic fatty liver disease (NAFLD) characterized by steatosis-associated inflammation and liver injury. Without effective treatment or management, NASH can have life-threatening outcomes. Evaluation and identification of NASH patients at risk for adverse outcomes are therefore important. Key issues in screening NASH patients are the assessment of advanced fibrosis, differentiation of NASH from simple steatosis, and monitoring of dynamic changes during follow-up and treatment. Currently, NASH staging and evaluation of the effectiveness for drugs still rely on pathological diagnosis, despite sample error issues and the subjectivity associated with liver biopsy. Optimizing the pathological assessment of liver biopsy samples and developing noninvasive surrogate methods for accessible, accurate, and safe evaluation are therefore critical. Although noninvasive methods including elastography, serum soluble biomarkers, and combined models have been implemented in the last decade, noninvasive diagnostic measurements are not widely applied in clinical practice. More work remains to be done in establishing cost-effective strategies both for screening for at-risk NASH patients and identifying changes in disease severity. In this review, we summarize the current state of noninvasive methods for detecting steatosis, steatohepatitis, and fibrosis in patients with NASH, and discuss noninvasive assessments for screening at-risk patients with a focus on the characteristics that should be monitored at follow-up.

INTRODUCTION

Nonalcoholic fatty liver disease (NAFLD) is a heterogeneous and silently progressive disease that affects roughly one-third (32%) of the global population [1,2]. With an alarming increase in both worldwide prevalence and incidence, NAFLD has become one of the most common causes of chronic liver diseases in the majority of industrialized areas [3,4]. Compared with nonalcoholic fatty liver (NAFL), which is characterized by bland steatosis, nonalcoholic steatohepatitis (NASH) is a more progressive phenotype of NAFLD characterized by hepatocyte injury, inflammation, and scarring. It has been estimated that around 25% of NAFLD patients will develop NASH, and 20% of patients with NASH will develop cirrhosis and hepatocellular carcinoma (HCC) in 20 to 30 years from disease onset [5]. In the past decade, liver-specific and overall mortality rates of NASH have been increasing rapidly, especially in the patients with obesity, type 2 diabetes mellitus (T2DM), and metabolic syndrome [6]. Early identification and targeted treatment for NASH are urgently needed to improve patient outcomes.
Currently, diagnosis and evaluation of the severity of NASH is still based on liver biopsy-proven histopathological assessment and scoring, and is therefore reliant on invasive liver biopsy. The main scoring systems for NASH consider liver fibrosis, inflammation, and steatosis [7,8]. Although a number of noninvasive tests and predictive models have been developed to characterize fibrosis in NASH patients, their diagnostic performance and clinical application can be improved. Since there are still no NASH-specific drugs that have been approved by major drug administration agencies worldwide, lifestyle interventions including dietary changes and exercise, with the purpose of 10% weight loss, are the most effective approaches for the management of fibrotic NASH and underlying cardiometabolic comorbidities [9]. Liver biopsy, the current “gold” standard for the diagnosis of NASH, is essential for both patient enrollment and efficacy assessment of phase 2b trials of drugs currently under development in addition to all phase 3 trials [10,11].
Accurate evaluation of the severity of NASH and the risk of progression to liver cirrhosis and HCC is essential for screening at-risk NASH patients and determining treatment responses (including NASH remission and cirrhosis prevention) to novel NASH drugs in clinical trials. In the present review, we discuss approaches used for the surveillance of the progression of NASH and assessment of treatment endpoints.

RISK OF NASH PROGRESSION

Fibrosis

Liver fibrosis is recognized as a determinant of liver-related morbidity and mortality in patients with NAFLD/NASH [12]. Previous studies have shown that significant fibrosis (≥F2) and advanced fibrosis (≥F3) are independently associated with overall mortality, liver transplantation, and liver-specific mortality in patients with NAFLD [13]. In one study, patients with fibrotic NAFLD had a lower survival rate after liver transplantation than those with non-fibrotic NAFLD, regardless of the presence of NASH [14]. A recent meta-analysis demonstrated that the risk of liver-related mortality, all-cause mortality, and requirement for a liver transplant increased with poorer biopsy-confirmed fibrosis stage [15]. According to the Finnish population-based FINRISK and Health 2000 studies with a median follow-up of 12.1 years, the crude incidence of liverrelated outcomes in NAFLD was 0.97/1,000 person-years, and outcomes were associated with noninvasive fibrosis stage [16]. Moreover, HCC risk was highest with cirrhosis, followed by noncirrhotic fibrosis and comorbid T2DM in a biopsy-proven NAFLD cohort [17]. Correspondingly, NASH patients with compensated cirrhosis may have fewer liver-related complications if fibrosis regression is evident, which presents as a decrease in NAFLD fibrosis score (NFS), liver stiffness measurements, and hepatic collagen and alpha-smooth muscle actin expression [18]. In addition, most of novel drugs in phase 3 clinical trials targeting NASH also target fibrosis with stage ≥F2 to prevent fibrosis progression and liver-related events. Therefore, identifying NASH patients with significant fibrosis or advanced fibrosis can be used to identify populations at high risk for progression to liver cirrhosis and HCC [19].

Inflammation: a trigger of fibrosis and carcinogenesis

Patients with simple steatosis are often considered to have a similar life expectancy to that of the general population, while patients with NASH are generally considered to have a lower life expectancy. In the presence of chronic inflammation, adipose tissue releases free fatty acids and toxic lipids, followed by fat accumulation, lipotoxicity, oxidative stress, and mitochondrial dysfunction in hepatocytes [20], leading to liver fibrogenesis and carcinogenesis. It has been reported that up to one-third of NASH patients without effective intervention will develop advanced liver fibrosis or cirrhosis, and potentially HCC [21]. Although a previous study investigated the impact of fibrosis on the prognosis of NAFLD patients, persistent hepatocyte injury or chronic inflammation in the liver is one of the driving forces of disease progression and carcinogenesis [22]. A further study confirmed that fibrosis progression is faster in NASH than NAFL and that NASH patients are at higher risk for HCC than NAFL patients; NAFL patients progress one fibrosis stage per 14.3 years, while patients with NASH progress one fibrosis stage per 7.1 years [23].

Metabolic dysfunction: cause or consequence?

Obesity is the most common cause of metabolic dysfunction, and is considered related to the epidemic of NAFLD. Overall obesity increases de novo lipogenesis and decreases β oxidation of free fatty acids and very low-density lipoprotein secretion, resulting in hepatocyte lipidosis and lipotoxicity. However, it should be noted that a large proportion of patients with NAFLD are lean or non-obese based on body mass index [24,25]. Approximately 8–19% of Asians with a body mass index (BMI) less than 25 kg/m2 also have NAFLD [26], and the prevalence of NAFLD in non-obese subjects has been found to be as high as 16% [24]. However, obesity as defined by BMI is only a crude measurement of obese status. Other anthropometric parameters might be useful for diagnosis of central obesity, occult obesity, and sarcopenic obesity. Central adiposity, sarcopenia, dyslipidemia, and insulin resistance are strongly associated with NASH and related fibrosis in a dose-dependent manner [27]. The progressive course of NASH is closely linked to an increasing number of metabolic comorbidities. T2DM has the strongest association with incident HCC in patients with NAFLD [28-30]. Metabolic syndrome is an independent predictor of all-cause, liver-specific, and cardiovascular mortality in patients with NAFLD [31,32]. In contrast, mortality of metabolically normal NAFLD patients is similar to that of patients without liver disease [33-35]. Thus, assessing metabolic dysfunction, including insulin resistance, may help define high-risk NASH patients [36]. In addition, accumulating evidence suggests that NAFLD has complex links with metabolic dysfunction; for example, NAFLD, especially NASH, is also associated with an increased risk of incident T2DM and atherosclerotic cardiovascular disease events [37].

HISTOPATHOLOGICAL SURVEILLANCE FOR NASH

Liver biopsy is imperfect

Screening of high-risk patients and surveillance for the development of liver-related complications are urgently needed for the management of NASH given the chronic progressive nature of this disease. Several novel NASH pharmacological agents are currently under development, and monitoring the treatment response relies on accurate assessment in clinical trials. Histopathological assessment is considered the “gold” standard for the diagnosis and evaluating of NASH severity and fibrosis stage. However, liver biopsy is not feasible for repeated assessment due to its invasive nature. Furthermore, histological evidence from liver biopsies is only moderately accurate and requires additional validation, therefore more reliable techniques for accurate quantification of the severity of NASH and fibrotic stage are required.
Histological classification of NASH is currently performed using semiquantitative scoring systems. NAFLD activity score (NAS) which was developed by the NASH clinical research network, and the steatosis, activity, fibrosis scoring system developed by fatty liver inhibition of progression Pathology Consortium, are the two most widely used scoring systems [7,38]. Both systems identify the location and features of fibrosis, number of inflammatory foci, number of balloon cells, and percentage of parenchymal involvement of the steatosis. Assessment depends on manual and subjective judgment, resulting in intra- and inter-observer variability. Although liver biopsy is generally considered safe and is widely available, histological scoring is limited by sampling error and ordinal classification. Developing innovative methods based on machine learning (ML), artificial intelligence (AI), and whole-slide images (WSI) may be a key to improve histopathological assessment.

Novel liver biopsy-based assessment tools

Second-harmonic generation (SHG) microscopy is highly sensitive to the collagen fibril/fiber structure, and has enabled the imaging of fibrillar collagen in various tissues. SHG-based novel technology has also been applied to assess hepatic fibrosis in chronic liver diseases [39]. HistoIndex as one of the SHG-based novel technologies for the assessment of hepatic steatosis has shown a good correlation with histopathologist scores [40], and was applied in a phase 2 clinical trial (MGL-3196, Resmetirom) to evaluate dynamic changes in steatosis during treatment [41]. A model to quantify fibrosis-related parameters (q-FPs) was developed by Wang et al. to assess the characteristics of liver fibrosis in NAFLD. A model containing four q-FPs (number of collagen strands, strand length, strand eccentricity, and strand solidity) was established based on findings in 50 test subjects and validated in 42 validation subjects to facilitate continuous and quantitative evaluation of fibrosis [42]. Furthermore, a combination of qFibrosis, qInflammation, qBallooning, and qSteatosis (qFIBS index) was developed to allow quantitative assessment of the characteristics of NAS (lobular inflammation, ballooning, and steatosis) by using SHG and two-photon excitation fluorescence imaging technology. qFIBS was developed and then validated in a cohort of 219 patients with biopsy-proven NAFLD/NASH and showed a robust correlation with NAS and fibrosis stages [43]. Recently, qFIBS was applied in a phase 2 trial of tropifexor (NCT02855164), to assess the resolution of NASH and fibrosis. qFIBS was found to have sufficient sensitivity to evaluate regressive changes in septa morphology and a reduction in septa parameters in F3 patients, which cannot be captured by traditional scoring systems [44].
Advances in machine-learning-based approaches are enabling histopathological monitoring of the progression and regression of NASH [45]. Digital WSI comprises scanning of hematoxylin-eosin -stained slides to quantify steatosis by assessing the steatosis proportionate area. Elastica van Gieson-stained slides can be scanned to quantify fibrosis by assessing the number of collagen and elastin fibers [46-48], and is regarded as an automated, precise, objective and quantitative method to assess NASH. Assessment of ballooning cells, one of the most important features of NASH, is highly subjective. AI-based technology can be trained to reproducibly quantify ballooned hepatocytes and standardize the evaluation [49]. ML-based models have been used to assess NASH histological characteristics accurately in addition to treatment response. PathAI showed concordance with ordinal grades from pathologists in terms of three NAS components. In addition, PathAI detected improvements in the DELTA Liver Fibrosis score in fibrosis responders in the combination group (cilofexor+firsocostat) in the ATLAS study [50]. AI- and ML-based technologies are advancing rapidly and can potentially address the inadequacies of pathological assessment of fibrotic NASH.

NONINVASIVE MARKERS ARE MORE PRACTICAL THAN LIVER BIOPSY FOR MONITORING OF NASH

Given the increasing prevalence of NASH, the base of at-risk patients who need screening is large. Liver biopsy is a critical bottleneck in the diagnosis and monitoring of these patients. Thus, it is critical to develop accurate noninvasive tests, markers, and models to evaluate NASH severity and monitor drug efficacy. Based on these needs, researchers have developed several noninvasive assessment methods including serum biomarkers, elastography-based markers, imaging studies, genetic tests, and omics profiling.
Noninvasive tests are more acceptable for evaluation of steatosis degree and fibrosis stage than liver biopsy, and also improve screening compliance and monitoring of NAFLD. As histological assessment from liver biopsy is still imperfect, an ideal solution is to link clinical outcomes such as cirrhosis, HCC, and liver-related complications with novel noninvasive markers. Correlating the histological severity of NASH and fibrosis stages with quantified noninvasive markers is a feasible approach (Table 1) [51-57].

Serum biomarkers for assessment of steatosis

Currently, the most promising noninvasive diagnostic tools for hepatic steatosis are the fatty liver index (FLI), the hepatic steatosis index (HSI), the NAFLD-liver fat score, the visceral adiposity index, the lipid accumulation product (LAP), and the triglyceride×glucose index [58]. Most of these indexes have been validated in biopsy-proven cohorts or magnetic resonance spectroscopy (MRS) results have been used as a reference. The accuracy of FLI, HSI, LAP, and the Zhejiang University index (ZJU) was evaluated in a general population by ultrasonography. Although FLI showed the highest C-statistic (0.85), the relatively low sensitivity of ultrasonography in detecting mild steatosis is of concern [59]. Although assessing steatosis grade is simpler than assessing inflammation or fibrosis, detecting >5% hepatic steatosis by circulating biomarkers alone is insufficient. Combinations of biomarkers would likely increase the accuracy of detecting steatosis. Dallas Steatosis Index (DSI), which consists of age, sex, diabetes, hypertension, race, BMI, serum triglycerides and alanine amiotransferase, was developed in the Dallas Heart Study of 737 patients with MRS-diagnosed liver fat. The C-statistic of DSI was found to be 0.82, but its diagnostic performance still needs external validation [60]. It should be noted that ultrasound tests are more widely available than blood-based tests. Serum proteins measured in these models are associated with metabolic disorders or insulin resistance and are not strictly specific to hepatic fat content, which may explain why these models have insufficient accuracy, especially in non-obese or lean subjects. The current serum-based noninvasive markers therefore have limited utility for surveillance.

Serum biomarkers for the assessment of liver fibrosis

Given that fibrosis is the major driver of liver-related outcomes in NAFLD, assessing fibrosis stage is essential for screening at-risk patients. The simple serum biomarker panel used in the fibrosis-4 index (FIB-4) and the aspartate aminotransferase (AST) to Platelet Ratio Index (APRI), originally developed for chronic viral hepatitis, could be applied in NASH patients. The cut-off value for FIB-4 is 2.67 and 1.30 to rule in and rule out advanced fibrosis in patients with NAFLD, respectively. NFS, developed from a liver biopsy-proven NAFLD cohort, has cut-off values of -1.455 and 0.676 to rule out or rule in advanced fibrosis [61]. BARD score (a scoring system include body mass index, AST/ALT ratio, and diabetes) was developed to diagnose advanced fibrosis by combining BMI, AST/ALT levels, and diabetic status [62]. Both FIB-4 and NFS are relatively easy to perform and are recommended for identification of NAFLD patients at low or high risk of advanced fibrosis. These tests have been widely used, and are available in primary health care units. However, due to the various etiologies of the cohorts who these makers were validated in, the accuracy of these tests needed to be improved when applied to NAFLD cohorts. In addition, models developed from biopsy-proven NAFLD cohorts often use higher cut-off values than those used for the general population. This leads to inferior diagnostic performance of NFS, FIB-4, and APRI in general population [63,64].
Many biomarker tests, including those with patented markers, involve direct biomarkers of fibrogenesis or fibrinolysis from the extracellular matrix. Type III collagen and hyaluronic acid (HA) are common biomarkers. The amino-terminal propeptide of type III procollagen (PIIINP) can discriminate between regular and advanced fibrosis with a C-statistic of 0.82-0.84 [65]. Enhanced liver fibrosis (ELF) test, a commercial panel of markers comprising serum HA, the PIIINP, and the tissue inhibitor of metalloproteinase-1 (TIMP-1), was first developed in children with NAFLD and validated in larger cohorts [66,67]. Recently, the ELF test was used to assess fibrosis improvement during aldafermin (NGM282) treatment [68]. Another type III collagen-based fibrosis algorithm including age, presence of diabetes, PRO-C3 (a marker of type III collagen formation), and platelet count (called ADAPT) showed better diagnostic performance than APRI, FIB-4 and NFS in predicting advanced fibrosis [69]. FibroMeter consists of age, weight, glucose, AST, ALT, ferritin, and platelets, and has been directly compared with ELF. ELF and FibroMeter had significantly higher C-statistics than NFS and FIB-4 in diagnosing advanced fibrosis, while the C-statistic did not differ significantly between ELF and FibroMeter [70]. FibroTest is a commercial panel with a C-statistic of 0.75–0.86 for significant fibrosis and 0.81–0.92 for advanced fibrosis [71]. FIBROSpect, which comprises alpha 2 macroglobulin, HA, and TIMP-1, is highly sensitive for advanced fibrosis (positive predictive value, PPV 92.5–94.7%), with a C-statistic of 0.86 [72]. Hepamet was developed in 2,452 biopsy-proven NAFLD patients, and had a higher C-statistic than FIB-4 and NFS. Hepamet is unaffected by age, BMI or diabetes [73]. These tests, although more accurate at predicting advanced fibrosis, are expensive, and there is still a dearth of direct comparisons in the same cohorts. In general, biomarkers or models detecting advanced fibrosis have a relatively high negative predictive value (NPV) while the positive predictive value (PPV) requires improvement.

Serum biomarkers to assess steatohepatitis

Hepatocyte ballooning and inflammation are the most important features of steatohepatitis, but current biochemical and imaging measures cannot effectively distinguish NASH from NAFL. Serum ALT is not a sufficiently sensitive predictive marker for diagnosis of steatohepatitis as less than 30% of NASH patients have elevated ALT levels (>35 U/L). Use of ALT >2 times the upper limit of normal to diagnose NASH only has 50% sensitivity and 61% specificity [74]. Cytokeratin 18 (CK18) is released into the serum on initiation of apoptosis in the form of CK18-M30 and CK18-M65 fragments. Serum CK18 has been the most widely investigated in the diagnosis of NASH. In one study, CK18 was thought to have potential predictive value for fibrosis, but showed a better correlation with ALT rather than with steatosis or fibrosis [75]. Another study involving repeated liver biopsy found that serum CK18 level was associated with NAS ≥5 (definite NASH) in patients with NAFLD [76]. Meta-analyses have confirmed that CK18 can predict steatohepatitis with a C-statistic around CK18 0.80 and sensitivity of 66–78% [77,78]. Index of NASH, which consists of waist-to-hip ratio, triglyceride, ALT, homeostatic model assessment for insulin resistance (HOMA) and gender, was developed to diagnose steatosis [79] but showed low sensitivity in an external cohort, especially in non-obese subjects [80]. Although serum level of hypersensitive C-reactive-protein (hsCRP) is included in the diagnosis of metabolic-dysfunction associated fatty liver disease [81] its diagnostic value in NASH requires further investigation. A recent study of 100 subjects observed an independent relationship between hs-CRP and NAFLD [82]. More direct evidence is required for use of hs-CRP as a diagnostic marker for NASH. Both single nucleotide polymorphisms and noncoding RNAs have been used to predict NASH. NASH Score (PNPLA3 genotype, AST, and fasting insulin) and circulating miR-122 have shown potential prognostic significance in NASH [83,84]. Unlike NASH-related fibrosis, there are currently no direct biomarkers for steatohepatitis. The available evidence indicates that use of a single biomarker to discriminate bland steatosis from NASH is unlikely to be successful.

Advances in imaging-based approaches

Ultrasonography is the most widely used imaging tool for identifying liver disease but lacks sensitivity. In patients with mild to moderate steatosis, the accuracy of ultrasonography is only around 50% [85]. Thus, quantitative ultrasound-based techniques are being developed to improve the diagnosis of hepatic steatosis. Attenuation coefficient (AC) and back scatter coefficient (BSC) have been shown to be correlated with the severity of hepatic steatosis. In a biopsy-proven study, AC and BSC achieved an accuracy of 61.7% and 68.3% in predicting steatosis grade, respectively, which are significantly higher accuracies that achieved with traditional ultrasonography [85]. Ultrasound-guided attenuation parameter has also showed excellent ability to distinguish steatosis grades (0.92, 95% confidence interval: 0.87–0.97) in non-B non-C chronic hepatitis subjects [86]. Transient elastography (TE) devices can be used to assess the controlled attenuation parameter (CAP) for liver fat quantification. CAP showed good sensitivity for detecting mild steatosis (S1) and excellent diagnostic accuracy in distinguishing S1, S2, and S3 in a study that used liver biopsy as the reference [87,88]. In terms of incidence and resolution of steatosis, CAP can also be used to assess dynamic changes [89]. Although the sampling error of CAP can be reduced by increasing the detection volume (3 cm3), its accuracy is reduced by increasing amounts of subcutaneous adipose.
Among magnetic resonance imaging (MRI)-based biomarkers, MRS is sensitive to small amount of hepatic adipose and is recognized as the most accurate noninvasive method to quantify steatosis. MRS is often used as the reference when assessing other noninvasive markers [59]. However, advanced training is required to measure MRS, which has limited its widespread application. MRI-proton density fat fraction (PDFF) is more accessible than MRS in most tertiary health centers. MRI-PDFF can assess the fat content in the whole liver and also allow for the assessment of regions of interest. Multiple studies have proven a close agreement between fat content as assessed by MRI-PDFF and histological steatosis grade [90,91]. Liver fat content measured by MRS or MRI-PDFF changes over time, which could reflect dynamic changes in hepatic steatosis. MRI-PDFF can be used to determine absolute and relative liver fat content. MRI-PDFF was shown to have better diagnostic accuracy than CAP in a head-to-head comparison [88].
Computed tomography (CT) can be used to assess liver fat content through the absolute attenuation of liver parenchyma value [92]. CT is more sensitive to moderate-to-severe steatosis than mild steatosis. The sensitivity for detecting grade ≥2 steatosis is more than 90%. Although CT is not routinely used to identify steatosis, it can be important in detecting incidental steatosis.
TE is the simplest and most commonly used noninvasive imaging tool for screening for fibrosis in clinics. The cut-off values of liver stiffness measurement (LSM) by TE for identifying advanced fibrosis varies with liver disease etiology. For NAFLD, a recent study determined a cut-off of 6.5 kPa to rule out advanced fibrosis and a cut-off of 12.1 kPa to rule in advanced fibrosis [93]. In a study of Asian NAFLD patients, the cut-off value to rule out advanced fibrosis was 7.9 kPa and the cut-off to rule in advanced fibrosis was 9.6 kPa [94]. LSM is sensitive to advanced fibrosis and cirrhosis, while its specificity for ruling out F1 and F2 fibrosis requires improvement. In addition, LSM can be affected by various factors including obesity, subcutaneous fat thickness, high ALT levels, and cholestasis [95]. Agile 3+ and Agile 4 are models that combine LSM with routine clinical parameters to identify advanced fibrosis and cirrhosis, respectively. Both Agile 4 and Agile 3+ showed better diagnostic performance, especially positive predictive value, than FIB-4 and LSM [96]. Acoustic radiation force imaging (ARFI) was developed from a chronic hepatitis C patient cohort to diagnose advanced fibrosis. The efficacy of ARFI, supersonic shear imaging (SSI), and TE was compared in a head-to-head study. Similar to TE, the application of ARFI and SSI in obese subjects is limited, and SSI showed higher accuracy than ARFI for diagnoses of F2 fibrosis [97].
MRI machines can be equipped with magnetic resonance elastography (MRE) to assess liver stiffness. Both MRE and TE showed excellent diagnostic accuracy for diagnosing stage F2-F4 fibrosis with a C-statistic of greater than 0.90 [98]. Several studies have reported that MRE is more accurate than TE [88,98,99]. MRE also has a higher success rate than TE at detecting fibrosis in obese patients (95.8% vs. 88.5%). In a recent meta-analysis, MRE had a higher C-statistic for detecting F≥2 and F≥3 but a similar performance to TE and shear wave elastography at detecting cirrhosis [100]. The combination of MRI with other imaging tests and biomarkers could increase diagnostic performance. MEFIB is the combination of MRE and FIB-4, and showed a relatively high PPV of 97.1% in diagnosing ≥stage F2 fibrosis [101]. The MRI-aspartate aminotransferase (MAST) score refers to the combination of MRI and NFS, FIB4, and FibroScan-aspartate aminotransferase (FAST). MAST had a higher C-statistic than that of the components of this index, reducing the number of the patients in the “gray zone” [102].

DYNAMIC MONITORING AND PROGNOSIS RISK ASSESSMENT

Definition and biomarkers of at-risk NASH patients

Given the progressive nature of NASH, there are numerous efforts underway to develop novel drugs. Emerging treatments mostly target hepatic fibrosis and steatohepatitis-associated inflammatory activity. Patients who are at risk of disease progression should therefore be included in clinical trials and effective tests should be used to repeatedly assess the drug response. The Liver Forum defined the following NAFLD subgroups: NAFL, indeterminate NASH, NASH without fibrosis, NASH with early fibrosis, NASH with bridging fibrosis, compensated cirrhosis, and decompensated cirrhosis [103]. A number of biopsy-proven studies have showed that both fibrosis stage and NAS at baseline are correlated with a higher risk of increased fibrosis stage during follow-up. Recently, Harrison et al. [104] defined “at-risk NASH” patients as NAFLD patients with NAS ≥4 and fibrosis stage ≥2. Following this definition, several studies have offered noninvasive solutions to distinguish these patients from others.
MACK-3 is the combination of AST, HOMA, and CK18, and has shown high accuracy in at-risk NASH patients (NAS ≥4 and F ≥2) [105]. Cut-off MACK-3 values of ≤0.134 and ≥0.550 can be used to rule out and rule in these patients who need more aggressive drug intervention, respectively [106]. The algorithm ADAPT mentioned previously is also effective at detecting at-risk patients [107]. A recent study compared the diagnostic performance of MEFIB, MAST, and FAST at detecting at-risk NASH patients. All three models provided utility in NAFLD risk stratification, while MEFIB showed better performance at detecting at-risk NASH than MAST and FAST [108]. Direct correlation with the severity of inflammation was previously regarded as the bottleneck of imaging tests, but currently corrected T1 (cT1) showed potential in predicting NASH. cT1 had better diagnostic accuracy (0.78 vs. 0.69) in identifying high-risk NASH than MRI-PDFF [109]. Furthermore, a protein-based signature of fibrosis could also serve as a diagnostic tool. A disintegrin, a metalloproteinase with thrombospondin motif like 2 (ADAMTSL2), and an 8-protein panel showed predictive value for at-risk NASH [110].

Biomarkers of treatment response and clinical outcomes

The best clinical outcome to evaluate the efficacy of NASH treatment is liver-related morbidity and mortality, while the surrogate endpoint is histologic outcome. Current guidelines recommend histological NASH resolution without worsening of fibrosis or regression of fibrosis without worsening of NASH as the treatment endpoint in phase 3 trials of NASH [11]. The reliance on histologic outcomes for primary trial endpoints is a barrier to patient enrollment. There is an urgent need to develop accurate noninvasive markers that reflect drug-induced changes. Markers or algorithms that reflect disease severity or long-term prognosis could be utilized as surrogate endpoints for clinical trials of drugs targeting NASH (Fig. 1).
Some noninvasive markers reflect dynamic changes associated with histological changes. Imaging-based tests have the best potential to be surrogates of histological assessment of steatosis grade and fibrosis stage [111]. As early as in the FLINT trial of obeticholic acid (OCA), MRI-PDFF was used as a surrogate marker of steatosis. Taking a 30% relative reduction in MRI-PDFF as an endpoint, OCA was better than the placebo in achieving the goal. In addition, non-responders also showed less histological improvement than responders (19% vs. 50%, respectively) [112]. Patented ELF and PIIINP were also used as serum markers of treatment efficacy in the PIVENS Trial. ELF showed a significant correlation with advanced fibrosis in patients with NASH, but not with longitudinal changes in fibrosis [113]. As mentioned above, ML-based methods can be used to translate histological characteristics into continuous variables. For instance, collagen proportionate area (CPA) as assessed by digital image analysis may offer a more granular assessment of fibrosis than routine histological analysis. Small changes detected by CPA might be missed when comparing fibrosis stages [114,115]. Furthermore, ML-based histological assessment is worth evaluation as a surrogate endpoint in clinical trials.

MEASUREMENT OF HEALTH-RELATED QUALITY OF LIFE AND EXTRAHEPATIC OUTCOMES

NASH patients often have concomitant extrahepatic diseases, such as obesity, dyslipidemia, hypertension, T2DM, cardiovascular disease, and chronic kidney disease. In obese NASH patients, the diagnostic accuracy of noninvasive markers needs to be improved [116]. Our research group investigated the diagnostic value of metabolic disorders in NASH fibrosis [36]. Insulin resistance has been proven to play an essential role in the development of steatohepatitis and fibrosis. Although treatment may benefit comorbidities in NASH patients, there is insufficient evidence to use an improvement in metabolic comorbidities as a trial endpoint. Compared with cirrhotic patients, non-cirrhotic NASH patients are likely to have a higher incidence of cardiovascular disease [117]. In this case, metabolic-related events should be closely monitored, while longer follow-up periods are required to observe liver-related outcomes.
NAFLD not only increases the risk for development of hepatic and extrahepatic outcomes, but impairs health-related quality of life (HRQoL). In comparison with healthy controls, patients with NAFLD have decreased HRQoL scores and impaired patient-reported outcomes (PRO) that are worse than those of patients with other chronic liver diseases [118]. Changes in HRQoL and PRO scores in NAFLD are associated with hepatic disease severity and its improvement after effective treatment. The HRQoL score declines in order from NAFL to NASH, then advanced fibrosis, and cirrhosis in patients with NAFLD. Histological improvement such as reduction of steatosis degree, remission of NASH, decreased NAS, and regression of fibrosis stage after multiple new drugs trial for NASH can improve PRO and HRQoL scores. Therefore, evaluation and monitoring of HRQoL and PRO in NAFLD patients should be encouraged in routine diagnosis and treatment. PRO and HRQoL should be regarded as primary endpoints for the management of NASH and NASH-related cirrhosis.

SUMMARY

The increasing prevalence of NASH is associated with a large health economic burden globally that is characterized by excess mortality, adverse clinical outcomes, and poor patient-reported outcomes (PROs). Since there are still no effective drugs for NASH treatment, clinical trials of novel drugs have been ongoing over the past decade. NASH encompasses a heterogeneous collection of metabolic disorders and slowly progressing features of liver diseases. The challenge in monitoring NASH lies in developing techniques that allow dynamic assessment. Many noninvasive markers and algorithms to evaluate NASH severity and the efficacy of treatment have been developed. A number of serum markers, imaging modalities, and noninvasive algorithms are currently under investigation. Nevertheless, the diagnostic performance, accessibility, and cost-effectiveness of most of these modalities require improvement. Furthermore, the monitoring of NASH should also include PROs and extrahepatic diseases, especially metabolic disorders. Comprehensive but individualized surveillance should be available for each patient. We are convinced that given more efforts and cooperation among healthcare systems, researchers, pharmaceutical companies and NASH patients, advances can be made in monitoring and evaluation systems that will improve the management and prognosis of NASH patients.

ACKNOWLEDGMENTS

The study was funded by the National Key Research and Development Program, No. 2017YFC0908903; National Natural Science Foundation of China, No. 81900507.

FOOTNOTES

Authors’ contribution
Shi YW and Fan JG contributed to the study concept and design; Shi YW and Fan JG contributed to drafting the manuscript; Fan JG contributed to critical revision of the manuscript for important intellectual content; both authors confirmed critical revision of the manuscript for important intellectual content.
Conflicts of Interest
The authors have no conflicts to disclose.

Figure 1.
Evaluation approaches for different trial phases and different stages of NASH. Specific sets of evaluation tools should be used for different phases of NASH. Different assessments are also required for patients with different stages of NASH. MRE, magnetic resonance elastography; MRI-PDFF, magnetic resonance imaging-proton density fat fraction; NASH, nonalcoholic steatohepatitis; NAS, NASH activity score; AEs, adverse events; CAP, controlled attenuation parameter; US, ultrasound; CK18, Cytokeratin 18; cT1, corrected T1; FLI, fatty liver index; HSI, hepatic steatosis index; FIB-4, fibrosis-4 index; LAP, lipid accumulation product; NFS, NAFLD fibrosis score; ELF test, enhanced liver fibrosis test; LSM, liver stiffness measurement; MELD score, model for end-stage liver disease score; HVPG, hepatic venous pressure gradient.

cmh-2022-0401f1.jpg
Table 1.
Surveillance markers for steatosis, steatohepatitis, and fibrosis in patients with NASH
Characteristics Assessment C-statistic
Steatosis Controlled attenuation parameter (CAP) [51] 0.82
Dallas steatosis index (DSI) [60] 0.82
MRI-proton density fat fraction (PDFF)52 0.99
Steatohepatitis Cytokeratin 18 (CK18) [53] 0.83–0.93
NAFIC score [54],* 0.85
Corrected T1 (cT1) [109] 0.78
Fibrosis Fibrosis-4 index (FIB-4) [55] 0.75 for SF
0.80 for AF
0.85 for cirrhosis
Liver stiffness measurement (LSM) [88] 0.86 for SF
0.80 for AF
0.69 for cirrhosis
NAFLD fibrosis score (NFS) [55] 0.83 for cirrhosis
0.73 for AF
0.72 for SF
Aspartate aminotransferase (AST) to Platelet Ratio Index (APRI) [55] 0.70 for SF
0.75 for AF
0.75 for cirrhosis
BARD score [56], 0.64 for SF
0.73 for AF
0.70 for cirrhosis
Enhanced liver fibrosis (ELF) score [70] 0.79 for AF
FiberMeter [70] 0.80 for AF
Shear wave elastography (SWE) [57] 0.86 for AF
0.89 for SF
0.88 for cirrhosis
Acoustic radiation force imaging (ARFI) [57] 0.77 for AF
0.84 for SF
0.84 for cirrhosis
Magnetic resonance elastography (MRE) [88] 0.89 for SF
0.87 for AF
0.87 for cirrhosis

NASH, nonalcoholic steatohepatitis; AF, advanced fibrosis; SF, significant fibrosis.

* A scoring system using ferritin, fasting insulin, and type IV collagen 7S.

A scoring system including body mass index, AST/ALT ratio, and diabetes.

Abbreviations

NASH
nonalcoholic steatohepatitis
NAFLD
nonalcoholic fatty liver disease
NAFL
nonalcoholic fatty liver
HCC
hepatocellular carcinoma
T2DM
type 2 diabetes mellitus
NFS
NAFLD fibrosis score
α-SMA
alpha-smooth muscle actin
BMI
body mass index
NAS
NAFLD activity score
ML
machine learning
AI
artificial intelligence
WSI
whole-slide images
SHG
second-harmonic generation
q-FPs
quantify fibrosis-related parameters
qFIBS
qFibrosis
FLI
fatty liver index
HSI
hepatic steatosis index
LAP
lipid accumulation product
MRS
magnetic resonance spectroscopy
DSI
Dallas Steatosis Index
FIB-4
fibrosis-4
AST
aspartate aminotransferase
APRI
AST to Platelet Ratio Index
HA
hyaluronic acid
PIIINP
amino-terminal propeptide of type III procollagen
TIMP-1
tissue inhibitor of metalloproteinase-1
NPV
negative predictive value
PPV
positive predictive value
CK18
cytokeratin 18
hs-CRP
hypersensitive C-reactive-protein
AC
attenuation coefficient
BSC
back scatter coefficient
TE
transient elastography
CAP
controlled attenuation parameter
MRI
magnetic resonance imaging
PDFF
proton density fat fraction
CT
computed tomography
LSM
liver stiffness measurement
ARFI
acoustic radiation force imaging
SSI
supersonic shear imaging
MRE
magnetic resonance elastography
MAST
MRI-aspartate aminotransferase
FAST
FibroScan-aspartate aminotransferase
HOMA
homeostasis model assessment
ADAMTSL2
A disintegrin
OCA
obeticholic acid
CPA
collagen proportionate area
HRQoL
health-related quality of life
PRO
patientreported outcomes
PROs
patient-reported outcomes

REFERENCES

1. Riazi K, Azhari H, Charette JH, Underwood FE, King JA, Afshar EE, et al. The prevalence and incidence of NAFLD worldwide: A systematic review and meta-analysis. Lancet Gastroenterol Hepatol 2022;7:851-861.
crossref pmid
2. Lee J, Kim T, Yang H, Bae SH. Prevalence trends of non-alcoholic fatty liver disease among young men in Korea: A Korean military population-based cross-sectional study. Clin Mol Hepatol 2022;28:196-206.
crossref pmid pmc pdf
3. Cotter TG, Rinella M. Nonalcoholic fatty liver disease 2020: The state of the disease. Gastroenterology 2020;158:1851-1864.
crossref pmid
4. Le MH, Yeo YH, Zou B, Barnet S, Henry L, Cheung R, et al. Forecasted 2040 global prevalence of nonalcoholic fatty liver disease using hierarchical bayesian approach. Clin Mol Hepatol 2022;28:841-850.
crossref pmid pmc pdf
5. Sheka AC, Adeyi O, Thompson J, Hameed B, Crawford PA, Ikramuddin S. Nonalcoholic steatohepatitis: A review. JAMA 2020;323:1175-1183 Erratum in: JAMA 2020;323:1619.
crossref pmid
6. Tan DJH, Setiawan VW, Ng CH, Lim WH, Muthiah MD, Tan EX, et al. Global burden of liver cancer in males and females: Changing etiological basis and the growing contribution of NASH. Hepatology 2022 Aug 29;doi: 10.1002/hep.32758.
crossref pmid pdf
7. Kleiner DE, Brunt EM, Van Natta M, Behling C, Contos MJ, Cummings OW, et al.; Nonalcoholic Steatohepatitis Clinical Research Network. Design and validation of a histological scoring system for nonalcoholic fatty liver disease. Hepatology 2005;41:1313-1321.
crossref pmid
8. Bedossa P, Poitou C, Veyrie N, Bouillot JL, Basdevant A, Paradis V, et al. Histopathological algorithm and scoring system for evaluation of liver lesions in morbidly obese patients. Hepatology 2012;56:1751-1759.
crossref pmid
9. Younossi ZM, Corey KE, Lim JK. AGA clinical practice update on lifestyle modification using diet and exercise to achieve weight loss in the management of nonalcoholic fatty liver disease: Expert review. Gastroenterology 2021;160:912-918.
crossref pmid
10. European Medicines Agency. Draft reflection paper on regulatory requirements for the development of medicinal products for chronic non-infectious liver diseases (PBC, PSC, NASH) - Scientific guideline. <https://www.ema.europa.eu/en/draftreflection-paper-regulatory-requirements-developmentmedicinal-products-chronic-non-infectious>. Accessed Nov 2022.

11. U.S. Food and Drug Administration (FDA) Guidance for industry. Noncirrhotic nonalcoholic steatohepatitis with liver fibrosis: developing drugs for treatment; draft guidance for industry; availability. Department of Health and Human Services, Center for Drug Evaluation and Research (CDER). Federal Register web site, <https://www.federalregister.gov/documents/2018/12/04/2018-26333/noncirrhotic-nonalcoholicsteatohepatitis-with-liver-fibrosis-developing-drugs-fortreatment-draft>. Accessed Apr 2021.

12. Dulai PS, Singh S, Patel J, Soni M, Prokop LJ, Younossi Z, et al. Increased risk of mortality by fibrosis stage in nonalcoholic fatty liver disease: systematic review and meta-analysis. Hepatology 2017;65:1557-1565.
crossref pmid pmc pdf
13. Kang SH, Lee HW, Yoo JJ, Cho Y, Kim SU, Lee TH, et al.; Korean Association for the Study of the Liver (KASL). KASL clinical practice guidelines: Management of nonalcoholic fatty liver disease. Clin Mol Hepatol 2021;27:363-401.
crossref pmid pmc pdf
14. Angulo P, Kleiner DE, Dam-Larsen S, Adams LA, Bjornsson ES, Charatcharoenwitthaya P, et al. Liver fibrosis, but no other histologic features, is associated with long-term outcomes of patients with nonalcoholic fatty liver disease. Gastroenterology 2015;149:389-397.e10.
crossref pmid pmc
15. Taylor RS, Taylor RJ, Bayliss S, Hagström H, Nasr P, Schattenberg JM, et al. Association between fibrosis stage and outcomes of patients with nonalcoholic fatty liver disease: A systematic review and meta-analysis. Gastroenterology 2020;158:1611-1625.e12.
crossref pmid
16. Männistö VT, Salomaa V, Färkkilä M, Jula A, Männistö S, Erlund I, et al. Incidence of liver-related morbidity and mortality in a population cohort of non-alcoholic fatty liver disease. Liver Int 2021;41:2590-2600.
crossref pmid pdf
17. Simon TG, Roelstraete B, Sharma R, Khalili H, Hagström H, Ludvigsson JF. Cancer risk in patients with biopsy-confirmed nonalcoholic fatty liver disease: A population-based cohort study. Hepatology 2021;74:2410-2423.
crossref pmid pdf
18. Sanyal AJ, Anstee QM, Trauner M, Lawitz EJ, Abdelmalek MF, Ding D, et al. Cirrhosis regression is associated with improved clinical outcomes in patients with nonalcoholic steatohepatitis. Hepatology 2022;75:1235-1246.
crossref pmid pmc pdf
19. Shin HS, Jun BG, Yi SW. Impact of diabetes, obesity, and dyslipidemia on the risk of hepatocellular carcinoma in patients with chronic liver diseases. Clin Mol Hepatol 2022;28:773-789.
crossref pmid pmc pdf
20. Marra F, Svegliati-Baroni G. Lipotoxicity and the gut-liver axis in NASH pathogenesis. J Hepatol 2018;68:280-295.
crossref pmid
21. Marengo A, Jouness RI, Bugianesi E. Progression and natural history of nonalcoholic fatty liver disease in adults. Clin Liver Dis 2016;20:313-324.
crossref pmid
22. Koyama Y, Brenner DA. Liver inflammation and fibrosis. J Clin Invest 2017;127:55-64.
crossref pmid pmc
23. Singh S, Allen AM, Wang Z, Prokop LJ, Murad MH, Loomba R. Fibrosis progression in nonalcoholic fatty liver vs nonalcoholic steatohepatitis: A systematic review and meta-analysis of paired-biopsy studies. Clin Gastroenterol Hepatol 2015;13:643-54.e1-9 quiz e39-40.
crossref pmid pmc
24. Shi Y, Wang Q, Sun Y, Zhao X, Kong Y, Ou X, et al. The prevalence of lean/nonobese nonalcoholic fatty liver disease: A systematic review and meta-analysis. J Clin Gastroenterol 2020;54:378-387.
pmid
25. Zeng J, Yang RX, Sun C, Pan Q, Zhang RN, Chen GY, et al. Prevalence, clinical characteristics, risk factors, and indicators for lean Chinese adults with nonalcoholic fatty liver disease. World J Gastroenterol 2020;26:1792-1804.
crossref pmid pmc
26. Fan J, Zeng J. NAFLD in Chinese: Growing concern and management strategy. Hong Kong Medical Diary 2020;25:16-17.

27. Fan JG, Kim SU, Wong VW. New trends on obesity and NAFLD in Asia. J Hepatol 2017;67:862-873.
crossref pmid
28. Younossi ZM, Henry L, Bush H, Mishra A. Clinical and economic burden of nonalcoholic fatty liver disease and nonalcoholic steatohepatitis. Clin Liver Dis 2018;22:1-10.
crossref pmid
29. Chalasani N, Younossi Z, Lavine JE, Charlton M, Cusi K, Rinella M, et al. The diagnosis and management of nonalcoholic fatty liver disease: Practice guidance from the American Association for the Study of Liver Diseases. Hepatology 2018;67:328-357.
crossref pmid pdf
30. Kanwal F, Kramer JR, Li L, Dai J, Natarajan Y, Yu X, et al. Effect of metabolic traits on the risk of cirrhosis and hepatocellular cancer in nonalcoholic fatty liver disease. Hepatology 2020;71:808-819.
crossref pmid pdf
31. Kim HY. Recent advances in nonalcoholic fatty liver disease metabolomics. Clin Mol Hepatol 2021;27:553-559.
crossref pmid pmc pdf
32. Sohn W, Lee HW, Lee S, Lim JH, Lee MW, Park CH, et al. Obesity and the risk of primary liver cancer: A systematic review and meta-analysis. Clin Mol Hepatol 2021;27:157-174.
crossref pmid pmc pdf
33. Younossi ZM, Otgonsuren M, Venkatesan C, Mishra A. In patients with non-alcoholic fatty liver disease, metabolically abnormal individuals are at a higher risk for mortality while metabolically normal individuals are not. Metabolism 2013;62:352-360.
crossref pmid
34. Mantovani A, Petracca G, Beatrice G, Tilg H, Byrne CD, Targher G. Non-alcoholic fatty liver disease and risk of incident diabetes mellitus: An updated meta-analysis of 501 022 adult individuals. Gut 2021;70:962-969.
crossref pmid
35. Zaharia OP, Strassburger K, Strom A, Bönhof GJ, Karusheva Y, Antoniou S, et al.; German Diabetes Study Group. Risk of diabetes-associated diseases in subgroups of patients with recent-onset diabetes: A 5-year follow-up study. Lancet Diabetes Endocrinol 2019;7:684-694.
crossref pmid
36. Shi YW, He FP, Chen JJ, Deng H, Shi JP, Zhao CY, et al. Metabolic disorders combined with noninvasive tests to screen advanced fibrosis in nonalcoholic fatty liver disease. J Clin Transl Hepatol 2021;9:607-614.
crossref pmid pmc
37. Lonardo A, Nascimbeni F, Mantovani A, Targher G. Hypertension, diabetes, atherosclerosis and NASH: Cause or consequence? J Hepatol 2018;68:335-352.
crossref pmid
38. Bedossa P; FLIP Pathology Consortium. Utility and appropriateness of the fatty liver inhibition of progression (FLIP) algorithm and steatosis, activity, and fibrosis (SAF) score in the evaluation of biopsies of nonalcoholic fatty liver disease. Hepatology 2014;60:565-575.
crossref pmid
39. Gailhouste L, Le Grand Y, Odin C, Guyader D, Turlin B, Ezan F, et al. Fibrillar collagen scoring by second harmonic microscopy: A new tool in the assessment of liver fibrosis. J Hepatol 2010;52:398-406.
crossref pmid
40. Goh GB, Leow WQ, Liang S, Wan WK, Lim TKH, Tan CK, et al. Quantification of hepatic steatosis in chronic liver disease using novel automated method of second harmonic generation and two-photon excited fluorescence. Sci Rep 2019;9:2975.
crossref pmid pmc pdf
41. Harrison SA, Guy CD, Bashir M, Frias JP, Alkhouri N, Baum S, et al. In a placebo-controlled 36-week phase 2 trial, treatment with MGL-3196 compared to placebo results in significant reductions in hepatic fat (MRI-PDFF), liver enzymes, fibrosis biomarkers, atherogenic lipids, and improvement in NASH on serial liver biopsy. Hepatology 2018;68 Suppl:9A-10A Abstract no. 14.

42. Wang Y, Vincent R, Yang J, Asgharpour A, Liang X, Idowu MO, et al. Dual-photon microscopy-based quantitation of fibrosisrelated parameters (q-FP) to model disease progression in steatohepatitis. Hepatology 2017;65:1891-1903.
crossref pmid pmc pdf
43. Liu F, Goh GB, Tiniakos D, Wee A, Leow WQ, Zhao JM, et al. qFIBS: An automated technique for quantitative evaluation of fibrosis, inflammation, ballooning, and steatosis in patients with nonalcoholic steatohepatitis. Hepatology 2020;71:1953-1966.
crossref pmid pdf
44. Naoumov NV, Brees D, Loeffler J, Chng E, Ren Y, Lopez P, et al. Digital pathology with artificial intelligence analyses provides greater insights into treatment-induced fibrosis regression in NASH. J Hepatol 2022;77:1399-1409.
crossref pmid
45. Marti-Aguado D, Rodríguez-Ortega A, Mestre-Alagarda C, Bauza M, Valero-Pérez E, Alfaro-Cervello C, et al. Digital pathology: Accurate technique for quantitative assessment of histological features in metabolic-associated fatty liver disease. Aliment Pharmacol Ther 2021;53:160-171.
crossref pmid pdf
46. Masugi Y, Abe T, Tsujikawa H, Effendi K, Hashiguchi A, Abe M, et al. Quantitative assessment of liver fibrosis reveals a nonlinear association with fibrosis stage in nonalcoholic fatty liver disease. Hepatol Commun 2017;2:58-68.
crossref pmid pmc pdf
47. Munsterman ID, van Erp M, Weijers G, Bronkhorst C, de Korte CL, Drenth JPH, et al. A novel automatic digital algorithm that accurately quantifies steatosis in NAFLD on histopathological whole-slide images. Cytometry B Clin Cytom 2019;96:521-528.
crossref pmid pmc pdf
48. Melo RCN, Raas MWD, Palazzi C, Neves VH, Malta KK, Silva TP. Whole slide imaging and its applications to histopathological studies of liver disorders. Front Med (Lausanne) 2020;6:310.
crossref pmid pmc
49. Brunt EM, Clouston AD, Goodman Z, Guy C, Kleiner DE, Lackner C, et al. Complexity of ballooned hepatocyte feature recognition: Defining a training atlas for artificial intelligence-based imaging in NAFLD. J Hepatol 2022;76:1030-1041.
crossref pmid
50. Taylor-Weiner A, Pokkalla H, Han L, Jia C, Huss R, Chung C, et al. A machine learning approach enables quantitative measurement of liver histology and disease monitoring in NASH. Hepatology 2021;74:133-147.
crossref pmid pmc pdf
51. Zhu J, He M, Zhang Y, Li T, Liu Y, Xu Z, et al. Validation of simple indexes for nonalcoholic fatty liver disease in western China: A retrospective cross-sectional study. Endocr J 2018;65:373-381.
crossref pmid
52. Xiao G, Zhu S, Xiao X, Yan L, Yang J, Wu G. Comparison of laboratory tests, ultrasound, or magnetic resonance elastography to detect fibrosis in patients with nonalcoholic fatty liver disease: A meta-analysis. Hepatology 2017;66:1486-1501.
crossref pmid pdf
53. Feldstein AE, Wieckowska A, Lopez AR, Liu YC, Zein NN, McCullough AJ. Cytokeratin-18 fragment levels as noninvasive biomarkers for nonalcoholic steatohepatitis: A multicenter validation study. Hepatology 2009;50:1072-1078.
crossref pmid pmc
54. Sumida Y, Yoneda M, Hyogo H, Yamaguchi K, Ono M, Fujii H, et al.; Japan Study Group of Nonalcoholic Fatty Liver Disease (JSG-NAFLD). A simple clinical scoring system using ferritin, fasting insulin, and type IV collagen 7S for predicting steatohepatitis in nonalcoholic fatty liver disease. J Gastroenterol 2011;46:257-268.
crossref pmid pdf
55. Ekstedt M, Franzén LE, Mathiesen UL, Thorelius L, Holmqvist M, Bodemar G, et al. Long-term follow-up of patients with NAFLD and elevated liver enzymes. Hepatology 2006;44:865-873.
crossref pmid
56. Harrison SA, Oliver D, Arnold HL, Gogia S, Neuschwander-Tetri BA. Development and validation of a simple NAFLD clinical scoring system for identifying patients without advanced disease. Gut 2008;57:1441-1447.
crossref pmid
57. Duarte SMB, Stefano JT, Miele L, Ponziani FR, Souza-Basqueira M, Okada LSRR, et al. Gut microbiome composition in lean patients with NASH is associated with liver damage independent of caloric intake: A prospective pilot study. Nutr Metab Cardiovasc Dis 2018;28:369-384.
crossref pmid
58. Fedchuk L, Nascimbeni F, Pais R, Charlotte F, Housset C, Ratziu V; LIDO Study Group. Performance and limitations of steatosis biomarkers in patients with nonalcoholic fatty liver disease. Aliment Pharmacol Ther 2014;40:1209-1222.
crossref pmid
59. Foschi FG, Conti F, Domenicali M, Giacomoni P, Borghi A, Bevilacqua V, et al. External validation of surrogate indices of fatty liver in the general population: The bagnacavallo study. J Clin Med 2021;10:520.
crossref pmid pmc
60. McHenry S, Park Y, Browning JD, Sayuk G, Davidson NO. Dallas steatosis index identifies patients with nonalcoholic fatty liver disease. Clin Gastroenterol Hepatol 2020;18:2073-2080.e7.
crossref pmid pmc
61. Angulo P, Hui JM, Marchesini G, Bugianesi E, George J, Farrell GC, et al. The NAFLD fibrosis score: A noninvasive system that identifies liver fibrosis in patients with NAFLD. Hepatology 2007;45:846-854.
crossref pmid
62. Lee TH, Han SH, Yang JD, Kim D, Ahmed M. Prediction of advanced fibrosis in nonalcoholic fatty liver disease: An enhanced model of BARD score. Gut Liver 2013;7:323-328.
crossref pmid pmc
63. Labenz C, Huber Y, Kalliga E, Nagel M, Ruckes C, Straub BK, et al. Predictors of advanced fibrosis in non-cirrhotic non-alcoholic fatty liver disease in Germany. Aliment Pharmacol Ther 2018;48:1109-1116.
crossref pmid pdf
64. Nielsen MJ, Leeming DJ, Goodman Z, Friedman S, Frederiksen P, Rasmussen DGK, et al. Comparison of ADAPT, FIB-4 and APRI as non-invasive predictors of liver fibrosis and NASH within the CENTAUR screening population. J Hepatol 2021;75:1292-1300.
crossref pmid
65. Tanwar S, Trembling PM, Guha IN, Parkes J, Kaye P, Burt AD, et al. Validation of terminal peptide of procollagen III for the detection and assessment of nonalcoholic steatohepatitis in patients with nonalcoholic fatty liver disease. Hepatology 2013;57:103-111.
crossref pmid
66. Nobili V, Parkes J, Bottazzo G, Marcellini M, Cross R, Newman D, et al. Performance of ELF serum markers in predicting fibrosis stage in pediatric non-alcoholic fatty liver disease. Gastroenterology 2009;136:160-167.
crossref pmid
67. Vali Y, Lee J, Boursier J, Spijker R, Löffler J, Verheij J, et al.; LITMUS systematic review team(†). Enhanced liver fibrosis test for the non-invasive diagnosis of fibrosis in patients with NAFLD: A systematic review and meta-analysis. J Hepatol 2020;73:252-262.
crossref pmid
68. Harrison SA, Rossi SJ, Paredes AH, Trotter JF, Bashir MR, Guy CD, et al. NGM282 improves liver fibrosis and histology in 12 weeks in patients with nonalcoholic steatohepatitis. Hepatology 2020;71:1198-1212.
crossref pmid pmc pdf
69. Daniels SJ, Leeming DJ, Eslam M, Hashem AM, Nielsen MJ, Krag A, et al. ADAPT: An algorithm incorporating PRO-C3 accurately identifies patients with NAFLD and advanced fibrosis. Hepatology 2019;69:1075-1086.
crossref pmid pdf
70. Guillaume M, Moal V, Delabaudiere C, Zuberbuhler F, Robic MA, Lannes A, et al. Direct comparison of the specialised blood fibrosis tests FibroMeterV2G and enhanced liver fibrosis score in patients with non-alcoholic fatty liver disease from tertiary care centres. Aliment Pharmacol Ther 2019;50:1214-1222.
crossref pmid pdf
71. Guha IN, Parkes J, Roderick P, Chattopadhyay D, Cross R, Harris S, 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.
crossref pmid
72. Loomba R, Jain A, Diehl AM, Guy CD, Portenier D, Sudan R, et al. Validation of serum test for advanced liver fibrosis in patients with nonalcoholic steatohepatitis. Clin Gastroenterol Hepatol 2019;17:1867-1876.e3.
crossref pmid
73. Ampuero J, Pais R, Aller R, Gallego-Durán R, Crespo J, García- Monzón C, et al.; HEPAmet Registry. Development and validation of hepamet fibrosis scoring system-a simple, noninvasive test to identify patients with nonalcoholic fatty liver disease with advanced fibrosis. Clin Gastroenterol Hepatol 2020;18:216-225.e5.
crossref pmid
74. Verma S, Jensen D, Hart J, Mohanty SR. Predictive value of ALT levels for non-alcoholic steatohepatitis (NASH) and advanced fibrosis in non-alcoholic fatty liver disease (NAFLD). Liver Int 2013;33:1398-1405.
crossref pmid pdf
75. Cusi K, Chang Z, Harrison S, Lomonaco R, Bril F, Orsak B, et al. Limited value of plasma cytokeratin-18 as a biomarker for NASH and fibrosis in patients with non-alcoholic fatty liver disease. J Hepatol 2014;60:167-174.
crossref pmid
76. Kawanaka M, Nishino K, Nakamura J, Urata N, Oka T, Goto D, et al. Correlation between serum cytokeratin-18 and the progression or regression of non-alcoholic fatty liver disease. Ann Hepatol 2015;14:837-844.
crossref pmid
77. Musso G, Gambino R, Cassader M, Pagano G. Meta-analysis: Natural history of non-alcoholic fatty liver disease (NAFLD) and diagnostic accuracy of non-invasive tests for liver disease severity. Ann Med 2011;43:617-649.
crossref pmid
78. Kwok R, Tse YK, Wong GL, Ha Y, Lee AU, Ngu MC, et al. Systematic review with meta-analysis: Non-invasive assessment of non-alcoholic fatty liver disease--the role of transient elastography and plasma cytokeratin-18 fragments. Aliment Pharmacol Ther 2014;39:254-269.
crossref pmid
79. Otgonsuren M, Estep MJ, Hossain N, Younossi E, Frost S, Henry L, et al. Single non-invasive model to diagnose non-alcoholic fatty liver disease (NAFLD) and non-alcoholic steatohepatitis (NASH). J Gastroenterol Hepatol 2014;29:2006-2013.
crossref pmid pdf
80. Younes R, Rosso C, Petta S, Cucco M, Marietti M, Caviglia GP, et al. Usefulness of the index of NASH - ION for the diagnosis of steatohepatitis in patients with non-alcoholic fatty liver: An external validation study. Liver Int 2018;38:715-723.
crossref pmid pdf
81. Eslam M, Newsome PN, Sarin SK, Anstee QM, Targher G, Romero-Gomez M, et al. A new definition for metabolic dysfunction-associated fatty liver disease: An international expert consensus statement. J Hepatol 2020;73:202-209.
crossref pmid
82. Kumar R, Porwal YC, Dev N, Kumar P, Chakravarthy S, Kumawat A. Association of high-sensitivity C-reactive protein (hs-CRP) with non-alcoholic fatty liver disease (NAFLD) in Asian Indians: A cross-sectional study. J Family Med Prim Care 2020;9:390-394.
crossref pmid pmc
83. Pirola CJ, Fernández Gianotti T, Castaño GO, Mallardi P, San Martino J, Mora Gonzalez Lopez Ledesma M, et al. Circulating microRNA signature in non-alcoholic fatty liver disease: From serum non-coding RNAs to liver histology and disease pathogenesis. Gut 2015;64:800-812.
crossref pmid pmc
84. Hyysalo J, Männistö VT, Zhou Y, Arola J, Kärjä V, Leivonen M, et al. A population-based study on the prevalence of NASH using scores validated against liver histology. J Hepatol 2014;60:839-846.
crossref pmid
85. Paige JS, Bernstein GS, Heba E, Costa EAC, Fereirra M, Wolfson T, et al. A pilot comparative study of quantitative ultrasound, conventional ultrasound, and MRI for predicting histologydetermined steatosis grade in adult nonalcoholic fatty liver disease. AJR Am J Roentgenol 2017;208:W168-W177.
crossref pmid pmc
86. Tada T, Kumada T, Toyoda H, Kobayashi N, Sone Y, Oguri T, et al. Utility of attenuation coefficient measurement using an ultrasound-guided attenuation parameter for evaluation of hepatic steatosis: Comparison with MRI-determined proton density fat fraction. AJR Am J Roentgenol 2019;212:332-341.
crossref pmid
87. de Lédinghen V, Wong GL, Vergniol J, Chan HL, Hiriart JB, Chan AW, et al. Controlled attenuation parameter for the diagnosis of steatosis in non-alcoholic fatty liver disease. J Gastroenterol Hepatol 2016;31:848-855.
crossref pmid pdf
88. Park CC, Nguyen P, Hernandez C, Bettencourt R, Ramirez K, Fortney L, et al. Magnetic resonance elastography vs transient elastography in detection of fibrosis and noninvasive measurement of steatosis in patients with biopsy-proven nonalcoholic fatty liver disease. Gastroenterology 2017;152:598-607.e2.
crossref pmid pmc
89. Garg H, Aggarwal S, Shalimar , Yadav R, Datta Gupta S, Agarwal L, et al. Utility of transient elastography (fibroscan) and impact of bariatric surgery on nonalcoholic fatty liver disease (NAFLD) in morbidly obese patients. Surg Obes Relat Dis 2018;14:81-91.
crossref pmid
90. Middleton MS, Heba ER, Hooker CA, Bashir MR, Fowler KJ, Sandrasegaran K, et al.; NASH Clinical Research Network. Agreement between magnetic resonance imaging proton density fat fraction measurements and pathologist-assigned steatosis grades of liver biopsies from adults with nonalcoholic steatohepatitis. Gastroenterology 2017;153:753-761.
crossref pmid pmc
91. Noureddin M, Lam J, Peterson MR, Middleton M, Hamilton G, Le TA, et al. Utility of magnetic resonance imaging versus histology for quantifying changes in liver fat in nonalcoholic fatty liver disease trials. Hepatology 2013;58:1930-1940.
crossref pmid pmc
92. Wells MM, Li Z, Addeman B, McKenzie CA, Mujoomdar A, Beaton M, et al. Computed tomography measurement of hepatic steatosis: Prevalence of hepatic steatosis in a Canadian population. Can J Gastroenterol Hepatol 2016;2016:4930987.
crossref pmid pmc pdf
93. Siddiqui MS, Vuppalanchi R, Van Natta ML, Hallinan E, Kowdley KV, Abdelmalek M, et al.; NASH Clinical Research Network. Vibration-controlled transient elastography to assess fibrosis and steatosis in patients with nonalcoholic fatty liver disease. Clin Gastroenterol Hepatol 2019;17:156-163.e2.
crossref pmid pmc
94. Wong VW, Vergniol J, Wong GL, Foucher J, Chan HL, Le Bail B, et al. Diagnosis of fibrosis and cirrhosis using liver stiffness measurement in nonalcoholic fatty liver disease. Hepatology 2010;51:454-462.
crossref pmid
95. Tapper EB, Castera L, Afdhal NH. FibroScan (vibrationcontrolled transient elastography): Where does it stand in the United States practice. Clin Gastroenterol Hepatol 2015;13:27-36.
crossref pmid
96. Sanyal AJ, Foucquier J, Younossi ZM, Harrison SA, Newsome PN, Chan WK, et al. Enhanced diagnosis of advanced fibrosis and cirrhosis in individuals with NAFLD using FibroScanbased Agile scores. J Hepatol 2022 Nov 12;doi: 10.1016/j.jhep.2022.10.034.
crossref pmid
97. Cassinotto C, Boursier J, de Lédinghen V, Lebigot J, Lapuyade B, Cales P, 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.
crossref pmid
98. Chen J, Yin M, Talwalkar JA, Oudry J, Glaser KJ, Smyrk TC, et al. Diagnostic performance of MR elastography and vibrationcontrolled transient elastography in the detection of hepatic fibrosis in patients with severe to morbid obesity. Radiology 2017;283:418-428.
crossref pmid
99. Imajo K, Kessoku T, Honda Y, Tomeno W, Ogawa Y, Mawatari H, et al. Magnetic resonance imaging more accurately classifies steatosis and fibrosis in patients with nonalcoholic fatty liver disease than transient elastography. Gastroenterology 2016;150:626-637.e7.
crossref pmid
100. Selvaraj EA, Mózes FE, Jayaswal ANA, Zafarmand MH, Vali Y, Lee JA, et al.; LITMUS Investigators. Diagnostic accuracy of elastography and magnetic resonance imaging in patients with NAFLD: A systematic review and meta-analysis. J Hepatol 2021;75:770-785.
pmid
101. Jung J, Loomba RR, Imajo K, Madamba E, Gandhi S, Bettencourt R, et al. MRE combined with FIB-4 (MEFIB) index in detection of candidates for pharmacological treatment of NASHrelated fibrosis. Gut 2021;70:1946-1953.
crossref pmid pmc
102. Noureddin M, Truong E, Gornbein JA, Saouaf R, Guindi M, Todo T, et al. MRI-based (MAST) score accurately identifies patients with NASH and significant fibrosis. J Hepatol 2022;76:781-787.
crossref pmid
103. Siddiqui MS, Harrison SA, Abdelmalek MF, Anstee QM, Bedossa P, Castera L, et al.; Liver Forum Case Definitions Working Group. Case definitions for inclusion and analysis of endpoints in clinical trials for nonalcoholic steatohepatitis through the lens of regulatory science. Hepatology 2018;67:2001-2012.
crossref pmid pmc pdf
104. Harrison SA, Ratziu V, Boursier J, Francque S, Bedossa P, Majd Z, et al. A blood-based biomarker panel (NIS4) for non-invasive diagnosis of non-alcoholic steatohepatitis and liver fibrosis: A prospective derivation and global validation study. Lancet Gastroenterol Hepatol 2020;5:970-985.
crossref pmid
105. Boursier J, Anty R, Vonghia L, Moal V, Vanwolleghem T, Canivet CM, et al. Screening for therapeutic trials and treatment indication in clinical practice: MACK-3, a new blood test for the diagnosis of fibrotic NASH. Aliment Pharmacol Ther 2018;47:1387-1396.
crossref pmid pdf
106. Chuah KH, Wan Yusoff WNI, Sthaneshwar P, Nik Mustapha NR, Mahadeva S, Chan WK. MACK-3 (combination of hoMa, Ast and CK18): A promising novel biomarker for fibrotic non-alcoholic steatohepatitis. Liver Int 2019;39:1315-1324.
crossref pmid pdf
107. Tang LJ, Ma HL, Eslam M, Wong GL, Zhu PW, Chen SD, et al. Among simple non-invasive scores, Pro-C3 and ADAPT best exclude advanced fibrosis in Asian patients with MAFLD. Metabolism 2022;128:154958.
crossref pmid
108. Kim BK, Tamaki N, Imajo K, Yoneda M, Sutter N, Jung J, et al. Head-to-head comparison between MEFIB, MAST, and FAST for detecting stage 2 fibrosis or higher among patients with NAFLD. J Hepatol 2022;77:1482-1490.
crossref pmid
109. Andersson A, Kelly M, Imajo K, Nakajima A, Fallowfield JA, Hirschfield G, et al. Clinical utility of magnetic resonance imaging biomarkers for identifying nonalcoholic steatohepatitis patients at high risk of progression: A multicenter pooled data and meta-analysis. Clin Gastroenterol Hepatol 2022;20:2451-2461.e3.
crossref pmid
110. Corey KE, Pitts R, Lai M, Loureiro J, Masia R, Osganian SA, et al. ADAMTSL2 protein and a soluble biomarker signature identify at-risk non-alcoholic steatohepatitis and fibrosis in adults with NAFLD. J Hepatol 2022;76:25-33.
crossref pmid pmc
111. Starekova J, Hernando D, Pickhardt PJ, Reeder SB. Quantification of liver fat content with CT and MRI: State of the art. Radiology 2021;301:250-262.
crossref pmid pmc
112. Loomba R, Neuschwander-Tetri BA, Sanyal A, Chalasani N, Diehl SAMB, Terrault N, et al.; NASH Clinical Research Network. Multicenter validation of association between decline in MRI-PDFF and histologic response in NASH. Hepatology 2020;72:1219-1229.
crossref pmid pmc pdf
113. Gawrieh S, Wilson LA, Yates KP, Cummings OW, Vilar-Gomez E, Ajmera V, et al. Relationship of ELF and PIIINP with liver histology and response to Vitamin E or pioglitazone in the PIVENS trial. Hepatol Commun 2021;5:786-797.
crossref pmc pdf
114. Forlano R, Mullish BH, Maurice JB, Thursz MR, Goldin RD, Manousou P. NAFLD: Time to apply quantitation in liver biopsies as endpoints in clinical trials. J Hepatol 2021;74:241-242.
crossref pmid
115. Buzzetti E, Hall A, Ekstedt M, Manuguerra R, Guerrero Misas M, Covelli C, et al. Collagen proportionate area is an independent predictor of long-term outcome in patients with non-alcoholic fatty liver disease. Aliment Pharmacol Ther 2019;49:1214-1222.
crossref pmid pdf
116. Qadri S, Ahlholm N, Lønsmann I, Pellegrini P, Poikola A, Luukkonen PK, et al. Obesity modifies the performance of fibrosis biomarkers in nonalcoholic fatty liver disease. J Clin Endocrinol Metab 2022;107:e2008-e2020.
crossref pmid pmc pdf
117. Vilar-Gomez E, Calzadilla-Bertot L, Wai-Sun Wong V, Castellanos M, Aller-de la Fuente R, Metwally M, et al. Fibrosis severity as a determinant of cause-specific mortality in patients with advanced nonalcoholic fatty liver disease: A multi-national cohort study. Gastroenterology 2018;155:443-457.e17.
crossref pmid
118. Sun C, Fan JG. Editorial: changes of health-related quality of life associated with liver disease severity and its improvement after treatment in NAFLD. Aliment Pharmacol Ther 2023;57:257-258.
crossref pmid pdf

Editorial Office
The Korean Association for the Study of the Liver
Room A1210, 53 Mapo-daero(MapoTrapalace, Dowha-dong), Mapo-gu, Seoul, 04158, Korea
TEL: +82-2-703-0051   FAX: +82-2-703-0071    E-mail: kasl@kams.or.kr
Copyright © The Korean Association for the Study of the Liver.         
COUNTER
TODAY : 995
TOTAL : 1778141
Close layer