You are on page 1of 11

Review

Non-invasive assessment of non-alcoholic fatty liver disease:


Clinical prediction rules and blood-based biomarkers
Eduardo Vilar-Gomez⇑, Naga Chalasani⇑

Summary Keywords: NAFLD; NASH;


The correct identification of patients at increased risk of non-alcoholic steatohepatitis (NASH) and FIB-4; APRI.
advanced fibrosis is a critical step in the assessment of non-alcoholic fatty liver disease (NAFLD). Since
Received 1 November 2017;
liver biopsy is invasive, expensive and prone to sampling error, several clinical prediction rules and
received in revised form 3
blood-based biomarkers have been developed as attractive and affordable alternatives for identification November 2017; accepted 9
of patients at high risk of NASH and advanced fibrosis. Current biomarkers constitute predictive models November 2017
(e.g. NAFLD fibrosis score, FIB-4 index and BARD score) or direct measures of inflammation (e.g. circu-
lating keratin 18 fragments), or fibrosis (e.g. FibroTestÒ, ELFTM or Pro-C3 tests). In the clinical setting,
biomarkers may discriminate between patients with NASH or advanced fibrosis, predict dynamic
changes in NASH/fibrosis over time, and provide long-term prognostic information. Although clinically
useful, current biomarker predictions may be influenced by hepatic and extrahepatic conditions (e.g.
age, patient comorbidities, and fibrosis or NASH prevalence), which may lead to inaccurate estimates
in small subsamples of patients. No highly sensitive and specific tests are available to differentiate NASH
from simple steatosis. However, diagnostic accuracy can be improved by combining blood biomarkers.
NAFLD fibrosis score and FIB-4 index are both cost-effective and highly sensitive tools to exclude
patients with advanced fibrosis. Moreover, their higher scores may identify patients at higher risk of
non-liver- and liver-related morbidity and mortality. More expensive tests such as FibroTest or ELF
are more specific for detection of patients with significant and advanced fibrosis. Recent efforts have
concentrated on ‘‘omics” approaches for developing and validating novel biomarkers. Herein, we
describe currently available clinical prediction rules and blood-based biomarkers for identifying NASH
and advanced fibrosis in patients with NAFLD, discussing their advantages and disadvantages, as well
as their potential clinical utility for predicting dynamic changes over time and identifying patients at
increased risk of adverse outcomes.
Ó 2017 European Association for the Study of the Liver. Published by Elsevier B.V. All rights reserved.

Introduction Prediction of NASH


Non-alcoholic fatty liver disease (NAFLD) is a Many biomarkers have been investigated to pre- Division of Gastroenterology and
Hepatology, Indiana University
leading cause of end-stage liver disease, hepatocel- dict NASH in patients with NAFLD. These range School of Medicine, IN, USA
lular carcinoma, and liver transplantation world- from clinical (age, gender, diabetes, body mass
wide.1–4 Non-alcoholic steatohepatitis (NASH) and index [BMI]), biochemical (aminotransferases,
most importantly fibrosis severity have been bilirubin and ferritin), metabolic (glycated hae- Key point
strongly implicated in the long-term prognosis of moglobin [HbA1c], insulin and HOMA-IR) and
Understanding the advan-
NAFLD patients.5–8 Thus, it is critical to identify lipid (triglycerides, cholesterol) parameters to tages and limitations of
patients at higher risk of NASH and advanced fibro- markers that reflect specific and complex molec- current biomarkers is
sis in order to optimise their management. Liver ular mechanisms underlying the pathogenesis imperative for selecting the
biopsy is still required to identify patients with and progression of NAFLD, including inflamma- appropriate biomarker in
the evaluation and
NASH and early fibrosis. However, its invasive nat- tion, oxidative stress, apoptosis, and glucose
management of NAFLD
ure, high cost and inter- and intra-observer vari- and lipid metabolism. As the pathogenesis of patients.
ability make it less suitable for diagnosis and NASH is complex and likely involves multiple
disease monitoring in clinical practice, and unsuit- biological aberrations, it is unlikely that a single
able for screening at a population level.9 During the biomarker could discriminate between simple
last decade, several non-invasive biomarkers have steatosis and NASH. Thus, most of current mod- Key point
been developed and validated to rule out patients els include at least two or more variables to add Because NASH and most
with NASH or advanced fibrosis. Thorough under- robustness to the non-invasive prediction importantly fibrosis sever-
standing of the advantages and limitations of cur- models. ity are critical determi-
rently available biomarkers is imperative for nants of long-term
prognosis of NAFLD, non-
selecting the appropriate biomarker for the evalua- Data from cross-sectional studies invasive biomarkers may
tion and management of patients with NAFLD. This Clinical prediction rules be useful to accurately
review article focusses on clinical prediction rules Several clinical and biochemical models have been distinguish patients at
and the blood-based biomarkers for non- evaluated to identify NASH in patients with higher risk of NASH and
invasively identifying NASH and advanced fibrosis advanced fibrosis.
NAFLD. Routinely tested blood parameters, such
in patients with NAFLD. as alanine aminotransferase (ALT) or aspartate

Journal of Hepatology 2017 vol. xxx j xxx–xxx


Please cite this article in press as: Vilar-Gomez E, Chalasani N. Non-invasive assessment of non-alcoholic fatty liver disease: Clinical prediction rules and blood-based biomarkers. J Hepa-
tol (2017), https://doi.org/10.1016/j.jhep.2017.11.013
Review

aminotransferase (AST), are associated with some before it can be incorporated into clinical practice
Key point
histological parameters such as inflammation (Fig. 1).
None of the currently and steatosis. However, correlations appear to be Adipocytokines are involved in the pathogene-
available tools differentiate influenced by severity of disease.10 Aminotrans- sis of NAFLD and have been strongly related to its
NASH from simple steato-
ferase levels do not correlate with the degree of severity. Tight links between circulating levels of
sis with high sensitivity
and specificity, however, fibrosis.11 The new normal ALT cut-off values adiponectin, leptin, ghrelin, interleukin-6 and
their diagnostic accuracy (men, 30 U/L and women, 19 U/L) were found to tumour necrosis factor-a, and insulin resistance,
can be improved by com- be associated with NASH, suggesting that their diabetes, obesity and dyslipidaemia may explain
bining blood biomarkers. adoption in clinical practice may identify patients their potential role in the progression of NAFLD.
at risk of NASH.12,13 Many clinical features such as Machado et al. found that a panel including adipo-
diabetes, hypertension, BMI, age and gender have nectin, leptin and ghrelin could differentiate
been consistently associated with NASH. However, patients with NASH and simple steatosis with an
they are inaccurate when used alone. Several clin- AUROC = 0.79.27
ical prediction models, which combine clinical and One of the more novel strategies to detect
laboratory variables, have been published in the patients at risk of NASH is to identify molecules
literature.14–17 These models perform reasonably by OMICS approaches (genomic, epigenomic,
well in predicting NASH, with an area under the metabolomic, transcriptomic, proteomic) using
receiver operating characteristic curve (AUROC) high-throughput technologies. OMIC approaches
of 0.76–0.80, and some of them yield acceptable result in the identification of a set of molecules
negative predictive values (NPV) of 80–93%, when highly related to the disease. The discovery of
using their lower cut-offs to exclude patients with new molecules via OMICS provides a very useful
NASH.14–17 framework for designing and validating highly
accurate predictive models. Genomics, epige-
Key point Blood-based models nomics, metabolomics, lipidomics and proteomics
Keratin 18 (K18) is a major intermediate filament are some of the new methods used to identify new
More recent efforts con-
centrating on ‘‘omics” protein in hepatocytes and a prominent substrate biomarkers able to distinguish between different
approaches have shown of caspase 3 cleavage that can be measured in patterns of NAFLD severity. Using a metabolomic
promising results but are serum by immunoassay.18 Higher serum concen- approach in patients with NAFLD, a model com-
not yet available for rou- bining glucose, glutamate, taurine and lactate
trations of K18 fragments have been detected in
tine clinical practice.
patients with NASH than those with simple steato- levels identified different patterns of severity of
sis.19 Data from two recent meta-analyses have NAFLD.28 Loomba et al. reported that levels of
reported a pooled AUROC of 0.82 (0.76–0.88) for polyunsaturated fatty acid metabolites were dif-
discriminating NASH patients with a median sen- ferent between patients with biopsy-proven sim-
sitivity and specificity of 66%–78% and 82%–87%, ple steatosis vs. NASH, and that combining some
respectively.18,20 For detecting NASH, a wide range metabolites such as 11,12-diHETrE, dhk PGD2,
of K18 cut-offs with their respective diagnostic and 20-COOH, enabled excellent NASH prediction
accuracy for M30 ELISA assays have been reported, (AUROC = 1), although this was a proof-of-
making it difficult to choose appropriate thresh- concept study and results require further valida-
olds for use in clinical practice. The M30 ELISA tion.29 Puri et al. also reported a significant reduc-
assay measures the caspase-cleaved K18 frag- tion in the lipogenic activity (docosahexanoic acid
ments and detects apoptosis, which is a hallmark [22:6 n3] to docosapentenoic acid [22:5n3] ratio
of steatohepatitis. The M65 ELISA assays detect was significantly decreased within phosphatidyl-
total cell death and interestingly have shown sim- choline, and phosphati-dylethanolamine pools),
ilar diagnostic accuracy for detecting NASH while the lipoxygenase metabolites were mark-
(pooled AUROC = 0.82). In one study, changes in edly increased in subjects with NASH.30 A recent
M65 were able to identify patients with NAFLD model including 28 serum lipid molecules (OWLi-
and early fibrosis stages (stages 1 and 2), as well verÒ test) yielded high predictive capability for
as fibrosis progression.21 Other combinatorial detection of subjects with NASH (AUC = 0.87 and
models which add K18 to soluble Fas,22 adipocy- 0.85 in derivation and validation sets).31 Finally,
tokines,23 clinical (diabetes, gender, BMI) and rou- Zhou and colleagues have recently reported that
tine blood-based parameters (ALT, platelets and a model ‘‘NASH ClinLipMet Score” based on lipids,
triglycerides)24,25 showed better NASH prediction metabolites, clinical markers and PNPLA genotype
in patients with NAFLD. More recently, a model identified patients with NASH, with an AUROC =
combining K18 fragments with C-terminal cleav- 0.86.32
age site of procollagen type-III N-terminal peptide
⇑ Corresponding authors.
(Pro-C3), acetyl-high mobility group box 1 and Data from longitudinal and interventional
Address: 702 Rotary Circle, patatin-like phospholipase domain-containing studies
Suite 225, Indiana University protein 3 (PNPLA3) rs738409 has significantly As a standalone measure, improvement in amino-
School of Medicine, improved the accuracy of NASH diagnosis (AURO transferases is associated with improvement in
Indianapolis, IN 46202, USA.
E-mail addresses: evilar@iu.edu
C = 0.87, sensitivity: 71% and specificity: 87%) in steatosis and inflammation over time.33 However,
(E. Vilar-Gomez), nchalasa@iu. patients with NAFLD.26 However, this needs to it fails to accurately detect patients with overall
edu (N. Chalasani). be validated and made commercially available improvement in NAFLD activity score (NAS) or

2 Journal of Hepatology 2017 vol. xxx j xxx–xxx


Please cite this article in press as: Vilar-Gomez E, Chalasani N. Non-invasive assessment of non-alcoholic fatty liver disease: Clinical prediction rules and blood-based biomarkers. J Hepa-
tol (2017), https://doi.org/10.1016/j.jhep.2017.11.013
JOURNAL OF HEPATOLOGY

Exclude
Secondary causes of steatosis
Significant alcohol consumption
Obesity
Imaging evidence of
Type 2 diabetes Risk factors ALT/GGT abnormalities
fat accumulation
Metabolic syndrome

NAFLD

Rule out advanced fibrosis (F ≥3)


NAFLD fibrosis score or FIB-4 index

NFS : < -1.455 NFS : -1.455 –0.672 NFS : >0.672 or 0.12 if >65 y
FIB -4: <1.30 FIB -4: 1.30 –3.25 FIB -4: >3.25 or 2.0 if >65 y
NPV: 88 -95% PPV: 75 -90%

55-58%* 30%* 12-15%*

Low risk Intermediate risk High risk

Exclude F ≥3 Diagnose F ≥3
- +
Consider repeating FibroTest: 0.3 FibroTest: 0.7 Consider liver biopsy
NFS/FIB-4 every 2 years FibroMeter: 0.61 FibroMeter: 0.71
Hepascore: 0.37 Hepascore: 0.7
NPV >90% PPV : 60-80%

ELF test <10.35 ELF test >10.35

Imaging methods (VCTE, MRE)

Fig. 1. Algorithm to non-invasively assess patients with NAFLD and advanced fibrosis using prediction rules and blood-based biomarkers. *Estimated
prevalence for low, intermediate and high risks groups. ALT, alanine aminotransferase; GGT, gamma-glutamyl transferase; MRE, magnetic resonance
elastography; NAFLD, non-alcoholic fatty liver disease; NFS, NAFLD fibrosis score; NPV, negative predictive value; PPV, positive predictive value; VCTE,
vibration controlled transient elastography.

resolution of NASH after therapeutic interven- pute NAS, limiting its use in real-life clinical
tions.13,34 Dynamic changes in serum K18 frag- practice.
ment levels over time and their association with
histological improvement have recently been
studied in 231 adults with NASH and 152 children Prediction of advanced fibrosis
with NAFLD, who participated in two separate Data from cross-sectional studies
prospective randomised clinical trials.34 Although Clinical prediction rules
changes in serum K18 levels over time were clo- Since fibrosis stage is a major determinant of all-
sely linked to improved liver histology, these cause and liver-related mortality,5–8,35,36 non-
changes did not perform better than changes in invasive assessment of fibrosis severity is crucial
ALT for detecting patients with overall histological in the management of patients with NAFLD. Sev-
NAS improvement or resolution of NASH. More- eral non-invasive algorithms based on clinical
over, diagnostic accuracy was not improved by and biochemical variables have been developed
adding changes in ALT to K18 levels.34 In the set- to detect individuals with NAFLD and advanced
ting of NASH and lifestyle interventions, a model fibrosis. However, major drawbacks include
including age, diabetes, baseline NAS ≥5, weight reduced accuracy for detection of earlier fibrosis
loss and normalisation of ALT (19 U/L and 30 U/L stages and the high proportion of patients with
for women and men, respectively) at end-of- undetermined results (30%). These non-invasive
intervention accurately identified patients with scoring systems, including NAFLD fibrosis score
resolution of NASH (AUROC = 0.96 and 0.95 in (NFS),37 FIB-4 index,38 AST/platelet ratio (APRI)
training and validation cohorts, respectively) after index,39 and BARD score40 yield high NPV but poor
52 weeks of diet and exercise. By using two cut- positive predictive values (PPV), suggesting that
offs to identify patients with a low and high prob- they are best applied to exclude subjects without
ability of NASH resolution, 91% of patients were advanced fibrosis, thereby avoiding unnecessary
correctly classified.13 Although clinically useful, liver biopsies. A recent meta-analysis compared
patients require a liver biopsy at baseline to com- the diagnostic efficacy of NFS, FIB-4 index and

Journal of Hepatology 2017 vol. xxx j xxx–xxx 3


Please cite this article in press as: Vilar-Gomez E, Chalasani N. Non-invasive assessment of non-alcoholic fatty liver disease: Clinical prediction rules and blood-based biomarkers. J Hepa-
tol (2017), https://doi.org/10.1016/j.jhep.2017.11.013
Review

BARD score for detecting advanced fibrosis. By fibrosis.44,48 In patients with NAFLD, Fibrometer
using their low cut-offs to rule out advanced fibro- showed higher diagnostic robustness in detecting
sis, pooled AUROC were 0.84 and 0.85 for NFS significant (AUROC = 0.94) or advanced fibrosis
(cut-off = 1.455) and FIB-4 (cut-off = 1.30), (AUROC = 0.93).
respectively. Whereas, using high thresholds to PIIINP is a serum marker of collagen turnover
diagnose advanced fibrosis, pooled AUROC were and increased levels occur as a consequence of tis-
0.65 and 0.84 for NFS (cut-off = 0.676) and FIB-4 sue repair and fibrosis. PIIINP as a single biomar-
(cut-off = 3.25). A BARD score of two showed ker has been associated with advanced fibrosis in
lower diagnostic accuracy than NFS and FIB-4 patients with NAFLD. For diagnosing advanced
score (AUROC = 0.76).41 These results indicate that fibrosis, a cut-off of 6.6 ng/ml and 11 ng/ml gave
both FIB-4 thresholds (1.30 and 3.25) have good an NPV and PPV of 95% and 100%, respectively.49
diagnostic accuracy to discriminate patients with Pro-C3 is a new serum marker derived exclusively
advanced fibrosis, whereas a 1.455 cut-off in from collagen III synthesis and deposition that has
NFS may accurately exclude patients with recently been validated as a predictor of fibrosis
advanced fibrosis. Despite NFS and FIB-4 display- progression in patients with chronic hepatitis
ing good diagnostic efficacy, many patients C.50 A recent study in 150 biopsy-proven patients
(30%) fall in-between the lower and upper with NAFLD reported that Pro-C3 was able to iden-
threshold values (indeterminate results), and tify patients with advanced fibrosis yielding an
many factors such as age, diabetes, and prevalence AUROC of 0.91, and an NPV and PPV of 97% and
of fibrosis, among others, may influence their diag- 56%, respectively.51 Pro-C3 correctly classified
nostic performance. Recently, new age-adjusted 82% of patients using a previously published cut-
cut-offs have been proposed to improve the off (>1.6738).51 Most recently, serum Pro-C3 com-
diagnostic efficacy of NFS and FIB-4 for advanced bined with clinical variables (age, BMI, diabetes
fibrosis.42 McPherson et al. found lower diagnostic and platelets) was superior to established serolog-
specificity for NFS (20%) and FIB-4 (35%) in ical fibrosis tests at identifying individuals with
patients ≥65 years old and new thresholds (FIB-4 NAFLD and advanced fibrosis, yielding AUROC of
= 2.0 and NFS = 0.12) for those patients were able 0.87 and 0.85 in training and validation cohorts.52
to improve specificity (70%) without affecting Another model incorporating serum HA, CK-18
diagnostic sensitivity. and TIMP-1 yielded an AUROC of 0.90, with a sen-
FibroTestÒ is a commercial algorithm that has sitivity of 88% and specificity of 84% for predicting
shown good predictive values for diagnosing advanced fibrosis in patients with NAFLD.53
advanced fibrosis (AUROC = 0.88) in patients with FIBROSpectÒ is a test marketed to detect fibro-
NAFLD. However, its diagnostic performance may sis in patients with chronic liver disease.54 This
be reduced in the presence of some disease- panel incorporates a2-macroglobulin, HA and
related interferences, such as acute inflammation, TIMP-1. Data from a recent study including a large
sepsis and extrahepatic cholestasis.43 cohort of histologically confirmed patients with
Key point Hepascore is a widely used algorithm to detect NAFLD showed that this panel accurately detects
significant fibrosis in many chronic liver diseases. patients with advanced fibrosis displaying AUROC
Although more expensive, It combines clinical variables of age and gender of 0.87 and 0.85 in training and validation sets.55
ELF test, FibroTest and
with blood-based parameters including bilirubin,
FibroMeter are better at
identifying patients with gamma-glutamyl transferase, hyaluronic acid, Data from longitudinal and interventional
significant and advanced and a2-macroglobulin.44 In patients with NAFLD, studies
fibrosis. High scores of a threshold of 0.37 helps to identify individuals Since fibrosis severity is strongly related to non-
non-invasive scores (NFS, with advanced fibrosis, with an AUROC of 0.81, liver and liver-related morbidity and mortality,
APRI, FIB-4) can be imple-
mented to predict long-
and NPV and PPV of 97% and 60%, respectively.45 biomarkers of fibrosis may help to identify those
term adverse outcomes in patients at high risk of developing major clinical
population-based Blood-based models outcomes and death. In population-based studies,
screening. The enhanced liver fibrosis (ELFTM) test is another high NFS, FIB-4, APRI and FORNs56 scores have
algorithm that includes HA, procollagen type III consistently been associated with increased risk
N-terminal peptide (PIIINP), and tissue inhibitor of cardiovascular- and liver-related mortality.57–59
Key point of metalloproteinase 1 (TIMP-1). This has consis- In the same context, the ELF test has been
Although certain biomark- tently demonstrated good predictive values for strongly related to liver-related complications
ers appear to predict pro- identifying patients with advanced fibrosis, with and death in a wide range of liver-related diseases,
gression, improvement or an AUROC of 0.90, sensitivity of 80% and specificity including NAFLD, reinforcing the importance of
even development of clini- of 90%, using a cut-off of 10.35.46 However, its per- these models in predicting long-term adverse clin-
cal outcomes in NAFLD
patients, future studies
formance can be notably influenced by age and ical outcomes.60,61 In terms of disease progression,
should evaluate the diag- gender in patients with chronic hepatitis C, baseline and dynamic changes in ELF scores were
nostic accuracy of direct although this caveat would require confirmation evaluated in a cohort of patients with NASH and
biomarkers of liver inflam in patients with NAFLD.47 advanced fibrosis who were enrolled in two phase
mation/fibrogenesis/fibri Ò
Fibrometer has been recognised as another IIb trials with simtuzumab, a humanised mono-
nolysis as indicators of
efficacy of therapeutic blood test of fibrosis with very good diagnostic clonal antibody designed for the treatment of
interventions. accuracy for detecting patients with advanced fibrosis. Both trials were stopped at 96 weeks

4 Journal of Hepatology 2017 vol. xxx j xxx–xxx


Please cite this article in press as: Vilar-Gomez E, Chalasani N. Non-invasive assessment of non-alcoholic fatty liver disease: Clinical prediction rules and blood-based biomarkers. J Hepa-
tol (2017), https://doi.org/10.1016/j.jhep.2017.11.013
JOURNAL OF HEPATOLOGY

Table 1. Clinical predictors and blood-based biomarkers and their accuracy in the prediction of NASH. Data from cross-sectional studies.
Markers n NAFLD phenotype AUC* Diagnostic accuracy
Serum based variables Training Validation
set set
NAFIC score:75 619 All patients 0.85 0.78 NR
Ferritin ≥200 ng/ml (female) – ≥300ng/ml (male),
fasting insulin ≥10 lU/ml and type IV collagen 7 S
≥5.0ng/ml
Exhaled breath test:76 65 Morbidly obese 0.77 NR Sensitivity: 90%, specificity: 69%
N-tridecane, 3-methyl-butanonitrile, 1-propanol PPV: 81% and NPV: 82%
y
Resistin, cleaved CK-18, adiponectin23 101 All patients 0.91 0.73 cut-off = 0.4320
Sensitivity: 72%, specificity: 91%
Adiponectin, leptin, ghrelin27 82 Morbidly obese 0.79 NR Sensitivity: 82%, specificity: 76%
PPV: 35%, NPV: 96%
Terminal peptide of procollagen III49 136 All patients 0.83 0.78 Cut-off = 6.6, NPV: 85–100%
Cut-off = 11.0, PPV: 80–100%,
CK-18, soluble Fas22 177 All patients 0.93 0.79 Cut-off = probability 36%
Sensitivity: 88%, specificity: 89%
(13) C-methionine77 118 All patients 0.87 NR Cut-off <4.20%
Sensitivity: 81%, specificity: 76%
CK-18 fragments19,78 139 All patients 0.83 NR Wide range of cut-offs provided
201 (adults) 0.93 NR Wide range of cut-offs provided
All patients
(children)
CK-18 fragments (CK-18-M30, M65, M65ED) pooled 2,415 All patients 0.82à – CK-18-M30
analysis20 Sensitivity: 68%, specificity: 74%
CK-18-M65Sensitivity: 76%, specificity: 74%
CK-18-M65EDSensitivity: 91%, specificity:
64%
NASH ClinLipMet Score: glutamate, isoleucine, 318 All patients 0.86 NR Cut-off 0.134
glycine, lysophosphatidylcholine 16:0, Sensitivity: 86%, specificity: 72% NPV: 95%,
phosphoethanolamine 40:6, AST, fasting insulin, and PPV: 45%
PNPLA3 genotype32
OWLiver test: twenty-eight serum triglycerides31 467 All patients 0.87 0.85 Cut-off 0.54
Sensitivity: 71%, specificity: 92%
NPV: 82%, PPV: 84%
Combinations with serum based and clinical variables
OxNASH: BMI, age, AST, 13-HODE79 122 All patients 0.83 0.74 Cut-off <55, sensitivity: 81–84%
Cut-off >73, specificity: 97–63%
CK-18, ALT, platelets, triglycerides24 95 All patients 0.92 NR Cut-off value = 0.361
Sensitivity: 89%, specificity: 86%
PPV: 89%, NPV: 89%
Age ≥50, female, AST ≥45 IU/L, BMI ≥30, AST/ALT 80 All patients 0.76 NR Sensitivity: 74%, specificity: 66%
ratio ≥0.80 and serum HA ≥55 mcg/l15 PPV: 68%, NPV: 71%
diabetes, gender, BMI, triglycerides, M30 (apoptosis), 79 All patients 0.81 NR Cut-off = 0.2188, NPV: 85%
and M65–M30 (necrosis)25 Cut-off = 0.5689, PPV: 81%
Age, sex, height, weight, and serum levels of 257 All patients 0.79 0.79 No cut-off provided
triglycerides, cholesterol, A2M, apolipoprotein A1, PPV: 74%, NPV: 72%
haptoglobin, GGT, aminotransferases ALT, AST, and
total bilirubin16
Hypertension, diabetes, AST ≥27 IU/L, ALT ≥27 IU/L, 200 Morbidly obese 0.80 0.75 ≥6 – PPV: 87–93%
obstructive sleep apnoea, and non-black race14 ≤2 – NPV: 93–80%
Diabetes, triglycerides >150 mg/dl, ALT, and 253 Morbidly obese 0.76 NR ≤1, NPV: 90%
obstructive sleep apnoea17 ≥4, PPV: 60%
Novel combinations
Collagen Pro-C3, CK-18 (M30) fragments, AST, ALT, 374 All patients 0.87 NR Sensitivity: 71%, specificity: 87%
procollagen III N-terminal peptide, acetyl-HMGB-1,
and PNPLA3 rs73840926
MiR34a, YKL-40, HBA1C, A2M80 238 All patients 0.82 NR Sensitivity: 73%, specificity: 78% NPV: 79%
and PPV: 72%
A2M, alpha-2-macroglobulin; ALT, alanine aminotransferase; AST, aspartate aminotransferase; AUC, area under the receiver operating characteristic curve; BMI, body mass
index; CK, cytokeratin; GGT, gamma-glutamyl transferase; HA, hyaluronic acid; HBA1C, glycated haemoglobin; HMBG, high mobility group box protein; NAFLD, non-
alcoholic fatty liver disease; NR, not reported; PNPLA, patatin-like phospholipase domain containing protein A; PPV, positive predictive value; NPV, negative predictive
value; NASH, non-alcoholic steatohepatitis; YKL-40, chitinase-3-like protein 1.
*
Studies reporting AUC over 0.75 were included.
y
This cut-off was calculated in overall cohort to improve the model performance.
à
Pooled data from two meta-analysis.

Journal of Hepatology 2017 vol. xxx j xxx–xxx 5


Please cite this article in press as: Vilar-Gomez E, Chalasani N. Non-invasive assessment of non-alcoholic fatty liver disease: Clinical prediction rules and blood-based biomarkers. J Hepa-
tol (2017), https://doi.org/10.1016/j.jhep.2017.11.013
Review

Table 2. Clinical predictors and blood-based biomarkers and their accuracy in the prediction of advanced fibrosis (F ≥3). Data from cross-sectional studies.
Markers n NAFLD phenotype AUC* Diagnostic accuracy
Serum based variables Training set Validation set
AST/ALT81–83 174 All patients 0.83 0.83–0.90 Cut-off = 0.8, NPV: 93%, PPV: 44%
Terminal peptide of procollagen III49 136 All patients 0.82 0.84 Cut-off = 6.6 ng/ml, NPV: 95%
Cut-off = 11 ng/ml, PPV = 100%
Pro-C3 levels51 150 All patients 0.91 NR Cut-off >1.6738:
82% correctly classified
FibroTest: haptoglobin, a2-macroglobulin, 1,202 All patients 0.86 0.85 AUC: 0.88 for F ≥3 (METAVIR)
apolipoprotein-A, bilirubin, and GGT84–86 Cut-off = 0.3, NPV: 98%
Cut-off = 0.7, PPV: 60%
Cut-off = 0.47, PPV:51%, NPV: 89% [45]
APRI score: AST, Platelets82,87 175 All patients 080 0.56–0.67 Cut-off = 1, NPV: 84%, PPV: 37%
ELF test: hyaluronic acid, PIIINP, and 1,329 All patients 0.87 0.90 Cut-off = 8.5–10.18, AUC: 0.82 for ≥F2
TIMP-146,48,88 Cut-off = 10.35, AUC: 0.90 for ≥F3
Combinations with serum based and clinical variables
NAFLD fibrosis score: age, BMI, albumin, AST/ 733 All patients 0.88 0.77–0.84 Cut-off = 81.45, NPV: 78–93%
ALT ratio, hyperglycaemia, and Cut-off = 0.67, PPV: 82–90%
platelets82,89,90,42 Cut-off = 0.12, >65 yrs, NPV:81–98%
Fibrometer: age, weight, glucose, AST, ALT, 1,021 All patients 0.94 0.94 F ≥2, AUC: 0.94
ferritin, and platelets48 F ≥3 AUC: 0.93
FIB-4 index: age, AST, ALT, and 686 All patients 0.80 0.86 Cut-off = 1.30, NPV: 90–95%
platelets38,82,90,42 Cut-off = 2.67, PPV: 80%
Cut-off = 3.25, PPV: 75%
Cut-off = 2.0, >65 yrs, NPV = 82–98%
BARD score: BMI, diabetes, and AST/ALT 1,513 All patients 0.81 0.77–0.78 Cut-off = 2, PPV: 27%, NPV: 95–97%
ratio40,82,90
BARDI score: BMI, diabetes, AST/ALT ratio, and 107 All patients 0.88 NR Cut-off = 3, PPV: 51%, NPV: 96%
INR91
Hepascore: age, sex, bilirubin, GGT, 242 All patients – 0.81 Cut-off = 0.37, PPV: 57%, NPV:92%
a2-macroglobulin, and hyaluronic acid45
Novel combinations
HA, CK-18 and TIMP-153 180 All patients 0.90 NR Sensitivity: 88.2%, specificity: 84.1%
NPV: 97%, PPV: 60%
FIB-C3: Pro-C3, age, BMI, diabetes, and 433 All patients 0.86 0.85
platelets52
FIBROSpect test: a2-macroglubulin, 792 All patients 0.87 0.85 Sensitivity: 84–81%, specificity: 72–74%.
hyaluronic acid, and TIMP-155
ALT, alanine aminotransferase; AST, aspartate aminotransferase; AUC, area under the receiver operating characteristic curve; BMI, body mass index; CK, cytokeratin; INR,
international normalized ratio; GGT, gamma-glutamyl transferase; HA, hyaluronic Acid; NAFLD, non-alcoholic fatty liver disease; PPV, positive predictive value; NPV,
negative predictive value; Pro-C3, C-terminal cleavage site of N-terminal type III collagen propeptide; PIIINP, N-terminal type III collagen propeptide; TIMP-1, tissue
inhibitor of metalloproteinase-1.
*
Studies reporting AUROC over 0.75 were included.

owing to a lack of efficacy and patients were com- ALT (19 U/L and 30 U/L for women and men) is
bined for analysis of disease progression. Baseline able to accurately identify patients (AUROC = 0.9
ELF values were able to predict progression to cir- 6) with one-point improvements in fibrosis after
rhosis in patients with bridging fibrosis (c- one year of diet and exercise. Changes from base-
statistic: 0.80) or development of clinical events line in other biomarkers such as NFS, APRI, FIB-4
(c-statistic: 0.69). However, changes in ELF values and AST/ALT ratio yielded low diagnostic accuracy
over time did not improve predictions when for changes in fibrosis at end-of-intervention.65
added to baseline ELF values.62 We summarise the studies reporting models
In another therapeutic trial performed in dia- for NASH and advanced fibrosis predictions and
betic patients, Pro-C3 was able to identify patients their diagnostic accuracy (Tables 1–3). Selected
at high risk of advanced fibrosis at baseline, as clinical prediction rules for non-invasive assess-
well as those who progressed over time and ment of advanced fibrosis are also described
responded to a potential anti-fibrotic therapy, con- (Table 4).
firmed by increased or attenuated liver fibrosis on
paired biopsies.63
Since lifestyle interventions have demonstrated Proteomic investigations and NAFLD
a significant impact on liver histology improve- biomarkers
ment,64 identifying biomarkers that may detect There has been significant interest in the develop-
those patients with fibrosis improvement is a ment of protein-based biomarkers that utilise
priority. A recent study has demonstrated that a mass spectrometry to identify NASH and advanced
simple panel that combines changes from baseline fibrosis in patients with NAFLD. In a systematic
in platelets and HbA1c alongside normalisation of review, Ladaru and colleagues summarised 22

6 Journal of Hepatology 2017 vol. xxx j xxx–xxx


Please cite this article in press as: Vilar-Gomez E, Chalasani N. Non-invasive assessment of non-alcoholic fatty liver disease: Clinical prediction rules and blood-based biomarkers. J Hepa-
tol (2017), https://doi.org/10.1016/j.jhep.2017.11.013
JOURNAL OF HEPATOLOGY

Table 3. Biomarkers and their potential role in predicting dynamic changes in NASH or fibrosis and providing long-term prognostic information. Data
from longitudinal and interventional studies.
Markers n Study design Outcome prediction Risk AUC Diagnostic accuracy
Longitudinal or cohort studies
FIB-4, APRI, NAFLD 14,841 Population-based Liver-related APRI (high/intermediate), HR: 9.44 – –
fibrosis score and prospective survey in mortality FIB-4 (intermediate), HR: 3.15
FORNs score59 viral hepatitis (high), HR: 25.14
negative adult from NFS (high), HR: 6.52
NHANES III. FORNs score (intermediate), HR:
3.6 (high), HR: 63.1
APRI, NAFLD fibrosis 320 Cohort study in Liver-related APRI (intermediate), HR: 8.8 – –
score, and BARD biopsy-proven NAFLD mortality (high), HR: 20.9
score58 patients BARD score (intermediate), HR: 6.2
(high), HR: 6.6
NFS (intermediate), HR: 7.7 (high),
HR: 34.2
Interventional studies
ELF test62 477 Phase IIb trials of Histological Baseline ELF test Progression to –
simtuzumab. The progression to Progression from bridging fibrosis cirrhosis: 0.80
trials were stopped at cirrhosis to cirrhosis, HR: 3.13 Development of
96 wks due to lack of Development of Development of clinical events, clinical events:
efficacy. clinical events HR: 2.37 0.69
Changes in ELF over time does not
improve prediction to baseline
values
Pro-C363 297 Interventional trial Fibrosis attenuation Robust reduction in Pro-C3 as a – –
Glitazone therapy in in high-risk groups function of therapy. This
diabetes. biomarker at baseline is useful for
identification of responders to
glitazone therapy.
CK-18 fragments34 384 Interventional trial Overall histological Relative odds for overall Overall
(PIVENS and TONIC) improvement at 96 histological improvement (1.28) histological
wks* and NASH resolution (1.37) improvement:
0.71–0.72
NASH resolution:
0.64–0.69
NASH resolution score: 261 Interventional trial Histological – 0.96 and 0.95 in Cut-off ≤46.15,
weight loss, diabetes, (lifestyle changes for resolution of NASH training and NPV: 92%
NAS ≥5 and ALT 52 wks) at 52 wks validation sets Cut-off ≥69.72,
normalisation13 PPV: 89–92%
Fibrosis improvement 261 Interventional trial 1-point fibrosis – 0.96 in training Cut-off ≥0.497,
score: changes from (lifestyle changes for improvement at 52 set PPV: 94%,
baseline in A1C and 52 wks) wks Change in NFS: NPV: 91%
platelets, and ALT 0.77, FIB-4 index:
normalisation65 0.63, AST/ALT
ratio: 0.60, and
APRI: 0.50
ALT, alanine aminotransferase; AST, aspartate aminotransferase; CK, cytokeratin: ELF, enhanced liver fibrosis; HR, hazard ratio; NAS, NAFLD activity score; NASH, non-
alcoholic steatohepatitis; NFS, NAFLD fibrosis score; NPV, negative predictive value; Pro-C3; C-terminal cleavage site of N-terminal type III collagen propeptide; PPV,
positive predictive value.
*
Histological improvement as one point or greater improvement in the hepatocellular ballooning score, no increase in the fibrosis score, and either a decrease in the NAFLD
activity score (NAS) to a score of three or less or a decrease in the NAS of two points or fewer, with at least a 1-point decrease in either the lobular inflammation or steatosis
score.

studies which reported 21 confirmed serum pro- teins changed significantly between the simple
tein biomarkers.66 Unfortunately, none of these steatosis and NASH F3/F4 group, and the NASH
candidate proteins have been translated into com- and NASH F3/F4 group, respectively. The priority
mercially available diagnostic tests. 1 proteins with >30% change between any two
Bell et al. have conducted a serum proteomics groups were subsequently considered for develop-
and biomarker discovery study in 69 patients with ing biomarker candidates. A panel consisting of six
well characterised NAFLD (simple steatosis: 24, priority 1 proteins (fibrinogen b chain, retinol
steatohepatitis without advanced fibrosis: 23, binding protein 4, serum amyloid P component,
and steatohepatitis with stage 3/4 fibrosis [F3/4]: lumican, transgelin 2, and CD5 antigen-like) differ-
22) and 16 controls without NAFLD.67 Using a entiated all four patient groups with an overall
label-free mass spectrometry-based approach, success rate of 76% (AUROC for control group =
these investigators identified over 1,700 serum 1.0, simple steatosis = 0.83, NASH = 0.86, and
proteins with a peptide ID confidence level of NASH with F3/F4 = 0.91). A group of three priority
>75%, 605 of which changed significantly between 1 proteins (complement component C7, insulin-
any two patient groups (false discovery rate <5%). like growth factor acid labile subunit, and trans-
Importantly, expression levels of 55 and 15 pro- gelin 2) correctly categorised 90% of patients as

Journal of Hepatology 2017 vol. xxx j xxx–xxx 7


Please cite this article in press as: Vilar-Gomez E, Chalasani N. Non-invasive assessment of non-alcoholic fatty liver disease: Clinical prediction rules and blood-based biomarkers. J Hepa-
tol (2017), https://doi.org/10.1016/j.jhep.2017.11.013
Review

Table 4. Selected clinical prediction rule for non-invasive assessment of advanced fibrosis in patients with NAFLD.
Model Components and regression equation Cut-offs for advanced fibrosis Clinical benefits and limitations
NAFLD fibrosis score 1.675 + 0.037  Age (yrs) + 0.094  <1.45 (low) – NPV: 88–93% Cheap and reproducible Yes
BMI (kg/m2) + 1.13  IFG/diabetes >0.67 (high) – PPV: 78–90% Sensitivity to exclude F ≥3 High
(yes = 1, no = 0) + 0.99  AST/ALT ratio Age >65 yrs Specificity to diagnose F ≥3 Modest
 0.013  Platelet (109/L) 0.66  <0.12 (low) – 81–98% Influenced by age Yes
Albumin (g/dl) Allows predict clinical outcomes Yes
FIB-4 index Age (yrs)  AST [U/L]/(Platelet [109/L] <1.30 (low) – NPV: 90–95% Cheap and reproducible Yes
 (ALT [U/L])1/2) >3.25 (high) – PPV: 75% Sensitivity to exclude F ≥3 High
Age >65 yrs Specificity to diagnose F ≥3 Modest
<2.0 (low) – 82–98% Influenced by age Yes
Allows predict clinical outcomes Yes
BARD score AST/ALT ratio ≥0.8 = 2 points BMI ≥28 Single cut-off = 2 Cheap and reproducible Yes
= 1 point NPV: 95–97%, PPV: 27% Sensitivity to exclude F ≥3 High
Presence of diabetes = 1 point Specificity to diagnose F ≥3 Low
Score ranges from 0 to 4 points Influenced by age Unknown
Allows predict clinical outcomes Yes
APRI (AST [IU/L])/(AST upper limit of normal Single cut-off = 1 Cheap and reproducible Yes
[IU/L])/(Platelet [109/L]) NPV: 84%, PPV: 37% Sensitivity to exclude F ≥3 High
Specificity to diagnose F ≥3 Low
Influenced by age Unknown
Allows predict clinical outcomes Yes
Hepascore Y = exp [4.185818  (0.0249  Age) Single cut-off = 0.37 Cheap and reproducible Yes
+ (0.7464  Sex) + (1.0039  a2- NPV: 92%, PPV: 57% Sensitivity to exclude F ≥3 High
macroglobulin) + (0.0302  Specificity to diagnose F ≥3 Modest
Hyaluronic acid) + (0.0691  Influenced by age Unknown
Bilirubin)  (0.0012  GGT)] Allows predict clinical outcomes Yes
Hepascore = Y/(1 + Y)
ALT, alanine aminotransferase; AST, aspartate aminotransferase; BMI, body mass index; IFG, impaired fasting glucose; F ≥3, advanced fibrosis (bridging fibrosis and
cirrhosis); NAFLD, non-alcoholic fatty liver disease; NPV, negative predictive value; PPV, positive predictive value.
Variables included in the prediction models are in bold.
Individual patient scores for each model can be easily obtained using free online calculator: http://gihep.com/calculators/hepatology/.

having NAFLD (simple steatosis and NASH) or cohort. This approach is being applied to differen-
NASH F3/F4 (AUROC = 0.91). Interestingly, two tiate various histological stages in patients with
proteins (prothrombin fragment and paraoxonase biopsy-proven NASH.
1) were able to totally differentiate control sub- Other promising non-invasive approaches for
jects from those with all forms of NAFLD com- detecting patients with advanced NAFLD have
bined with an AUROC = 1.0. This group has also recently been developed and these biologically
reported the results of serum proteomic analysis based strategies could improve or complement
from their Ossabaw NASH model.68 This study the success of other non-invasive markers.
identified seven priority 1 proteins which were
different between pigs which developed NASH
and pigs without NASH. Importantly, these inves- Other promising non-invasive approaches
tigators found seven priority 1 proteins In a recently published paper, Decaris et al.
(apolipoprotein C-III, apolipoprotein B, serum described a novel method to non-invasively
amyloid P component, transthyretin, paraoxonase, describe the hepatic fibrogenesis flux rates in the
protein similar to a-2-macroglobulin precursor, liver tissue and in the blood. They have shown that
and orosomucoid I) to be common between their hepatic fibrogenesis flux rates correlate signifi-
human and Ossabaw swine proteomic cantly with the degree of liver fibrosis in 24 sub-
investigations.67,68 jects who underwent diagnostic liver biopsy for
A promising new sensitive and quantitative suspected NAFLD.70 Liver collagen fractional syn-
proteomic technology (SOMAscan assay, SomaLo- thesis rate (FSR) and plasma lumican (liver extra-
gic; Boulder, CO), that is worth watching closely, cellular matrix protein) FSR were measured
has recently emerged. It uses Slow Off-rate Modi- based on 2H labelling using tandem mass spec-
fied Aptamers (SOMAmers), single-stranded apta- trometry. In this method, patients undergoing
mers with modified nucleotides that have a high clinically indicated liver biopsy drank heavy water
affinity for specific protein targets in the serum, (2H20, 50 ml two to three times daily) for three to
which are subsequently quantified as DNA. In a five weeks prior to their biopsy. Key observations
preliminary study, a proteomic classifier based from this study were (a) there was active remod-
on eight proteins had an AUROC of 0.932 for iden- elling of hepatic extracellular matrix even in
tifying steatosis in obese individuals undergoing patients with advanced fibrosis; (b) hepatic colla-
bariatric surgery.69 Interestingly, a proteomic clas- gen FSR correlated significantly with fibrosis stage
sifier has performed as well as a multicomponent as well as non-invasive fibrosis indices, such as
classifier based on PNPLA3 genotype, proteomics, FIB-4 and liver stiffness by magnetic resonance
and phenomics at identifying steatosis in this elastography (MRE); (c) plasma lumican FSR

8 Journal of Hepatology 2017 vol. xxx j xxx–xxx


Please cite this article in press as: Vilar-Gomez E, Chalasani N. Non-invasive assessment of non-alcoholic fatty liver disease: Clinical prediction rules and blood-based biomarkers. J Hepa-
tol (2017), https://doi.org/10.1016/j.jhep.2017.11.013
JOURNAL OF HEPATOLOGY

correlated significantly with hepatic collagen FSR, advanced fibrosis, are inexpensive and easy to
liver fibrosis by histology, and non-invasive fibro- obtain. Other direct markers of fibrosis such as
sis markers such as liver stiffness by MRE and FIB- the ELF test, FibroTest and FibroMeter are more
4. If this method is reproduced in follow-up publi- specific and have higher PPVs for detecting
cations, this may serve as a novel approach to test patients with advanced fibrosis. Despite tremen-
anti-fibrotic compounds in short term proof-of- dous advances in the development of liquid
concept studies. It is disappointing that this study biomarkers, an important number of patients have
did not compare the hepatic collagen FSR and undetermined results. Thus, new biologically
lumican FSR with the ELF score. based, inexpensive, easily accessible, highly sensi-
Loomba et al. recently reported the utility of a tive and specific biomarkers that permit not only Key point
metagenomics signature based on the gut micro- the identification of patients at high risk of
Since NAFLD fibrosis score
biome for non-invasively detecting advanced adverse outcomes, but also the monitoring of dis-
and FIB-4 index are highly
fibrosis in 86 patients with NAFLD (14 had stage ease progression and therapeutic response after sensitive, inexpensive and
3 or 4 fibrosis).71 Their gut microbiome composi- interventions, are urgently needed. easy to obtain tools, they
tion was characterised using whole-genome shot- should be routinely used to
gun sequencing of the stool DNA. A random forest rule out patients with
advanced fibrosis even by
classifier, consisting of 40 variables including 37 Conflict of interest non-expert physician and
bacterial species, was able to identify patients Eduardo Vilar-Gomez: Nothing to disclose. Naga patients.
with F3/4, with an AUROC of 0.936. This study Chalasani has ongoing consulting activities (or
lacked a validation cohort. had in preceding 12 months) with NuSirt, Abbvie,
There are several ongoing investigations to Eli Lilly, Afimmune (DS Biopharma), Tobira (Aller-
examine the utility of blood-based molecular gan), Madrigal, Immuron, Shire, Ardelyx, and Axo-
markers (signatures derived from circulating vant. These consulting activities are generally in
microRNA, cell-free RNA, DNA methylation, and the areas of non-alcoholic fatty liver disease and
cell-free DNA methylation).72–74 These approaches drug hepatotoxicity. Dr. Chalasani receives
hold promise but they need to be validated exter- research grant support from Intercept, Lilly,
nally using independent cohorts. Gilead, Galectin Therapeutics and Cumberland
where his institution receives the funding. Over
the last decade, Dr. Chalasani has served as a paid
Conclusion consultant to more than 30 pharmaceutical com-
In summary, no highly sensitive and specific tests panies and this outside activities have regularly
are available to differentiate NASH from simple been disclosed to his institutional authorities.
steatosis. However, diagnostic accuracy can be
Key point
improved by combining blood biomarkers. More
investigations are needed before prediction mod- Authors’ contributions Combinatorial and com-
els and blood-based biomarkers become available Eduardo Vilar-Gomez and Naga P. Chalasani plex non-invasive scoring
systems for ruling out
for routine clinical care. Combinatorial and com- equally contributed to concept, design, drafting,
NASH and advanced fibro-
plex models including clinical, routine blood- critical review for intellectual content, and final sis perform better than
based variables and markers that reflect the approval of the version to be published. simple biomarkers. Com-
dynamic nature of the fibrogenic process appear bining clinical and blood-
to have higher diagnostic accuracy and predictive based biomarkers with
innovative imaging
value in identifying advanced fibrosis, in patients Supplementary data approaches should be
with NAFLD. NFS and FIB-4 are useful screening Supplementary data associated with this article evaluated in future studies.
tools to be routinely applied in clinical practice, can be found, in the online version, at
since they can accurately exclude patients with https://doi.org/10.1016/j.jhep.2017.11.013.

References [5] Angulo P, Kleiner DE, Dam-Larsen S, Adams LA, Bjornsson ES,
Charatcharoenwitthaya P, et al. Liver fibrosis, but no other histologic
[1] Chalasani N, Younossi Z, Lavine JE, Charlton M, Cusi K, Rinella M, et al. features, is associated with long-term outcomes of patients with
The diagnosis and management of nonalcoholic fatty liver disease: nonalcoholic fatty liver disease. Gastroenterology 2015;149:389–397,
Practice guidance from the American Association for the Study of Liver e310.
Diseases. Hepatology 2017, [In press]. [6] Bhala N, Angulo P, van der Poorten D, Lee E, Hui JM, Saracco G, et al. The
[2] Younossi Z, Henry L. Contribution of alcoholic and nonalcoholic fatty natural history of nonalcoholic fatty liver disease with advanced fibrosis
liver disease to the burden of liver-related morbidity and mortality. or cirrhosis: an international collaborative study. Hepatology
Gastroenterology 2016;150:1778–1785. 2011;54:1208–1216.
[3] Younossi ZM, Blissett D, Blissett R, Henry L, Stepanova M, Younossi Y, [7] Ekstedt M, Hagstrom H, Nasr P, Fredrikson M, Stal P, Kechagias S, et al.
et al. The economic and clinical burden of nonalcoholic fatty liver Fibrosis stage is the strongest predictor for disease-specific mortality in
disease in the United States and Europe. Hepatology NAFLD after up to 33 years of follow-up. Hepatology 2015;61:1547–1554.
2016;64:1577–1586. [8] Hagstrom H, Nasr P, Ekstedt M, Hammar U, Stal P, Hultcrantz R, et al.
[4] Marchesini G, Day CP, Dufour JF, Canbay A, Nobili V, Ratziu V, et al. EASL- Fibrosis stage but not NASH predicts mortality and time to development
EASD-EASO Clinical Practice Guidelines for the management of non- of severe liver disease in biopsy-proven NAFLD. J Hepatol
alcoholic fatty liver disease. J Hepatol 2016;64:1388–1402. 2017;66:1265–1273.

Journal of Hepatology 2017 vol. xxx j xxx–xxx 9


Please cite this article in press as: Vilar-Gomez E, Chalasani N. Non-invasive assessment of non-alcoholic fatty liver disease: Clinical prediction rules and blood-based biomarkers. J Hepa-
tol (2017), https://doi.org/10.1016/j.jhep.2017.11.013
Review

[9] Rinella ME, Sanyal AJ. Management of NAFLD: a stage-based approach. [31] Barr J, Caballeria J, Martinez-Arranz I, Dominguez-Diez A, Alonso C,
Nat Rev Gastro Hepat 2016;13:196–205. Muntane J, et al. Obesity-dependent metabolic signatures associated
[10] Fracanzani AL, Valenti L, Bugianesi E, Andreoletti M, Colli A, Vanni E, with nonalcoholic fatty liver disease progression. J Proteome Res
et al. Risk of severe liver disease in nonalcoholic fatty liver disease with 2012;11:2521–2532.
normal aminotransferase levels: a role for insulin resistance and [32] Zhou Y, Oresic M, Leivonen M, Gopalacharyulu P, Hyysalo J, Arola J, et al.
diabetes. Hepatology 2008;48:792–798. Noninvasive detection of nonalcoholic steatohepatitis using clinical
[11] Mathiesen UL, Franzen LE, Fryden A, Foberg U, Bodemar G. The clinical markers and circulating levels of lipids and metabolites. Clin Gastroen-
significance of slightly to moderately increased liver transaminase terol Hepatol 2016;14:1463–1472, e1466.
values in asymptomatic patients. Scand J Gastroenterol 1999;34:85–91. [33] Adams LA, Sanderson S, Lindor KD, Angulo P. The histological course of
[12] Kunde SS, Lazenby AJ, Clements RH, Abrams GA. Spectrum of NAFLD and nonalcoholic fatty liver disease: a longitudinal study of 103 patients
diagnostic implications of the proposed new normal range for serum with sequential liver biopsies. J Hepatol 2005;42:132–138.
ALT in obese women. Hepatology 2005;42:650–656. [34] Vuppalanchi R, Jain AK, Deppe R, Yates K, Comerford M, Masuoka HC,
[13] Vilar-Gomez E, Yasells-Garcia A, Martinez-Perez Y, Calzadilla-Bertot L, et al. Relationship between changes in serum levels of keratin 18 and
Torres-Gonzalez A, Gra-Oramas B, et al. Development and validation of a changes in liver histology in children and adults with nonalcoholic fatty
noninvasive prediction model for nonalcoholic steatohepatitis resolu- liver disease. Clin Gastroenterol H 2014;12:U2121–U2416.
tion after lifestyle intervention. Hepatology 2016;63:1875–1887. [35] Rafiq N, Bai C, Fang Y, Srishord M, McCullough A, Gramlich T, et al. Long-
[14] Campos GM, Bambha K, Vittinghoff E, Rabl C, Posselt AM, Ciovica R, et al. term follow-up of patients with nonalcoholic fatty liver. Clin Gastroen-
A clinical scoring system for predicting nonalcoholic steatohepatitis in terol Hepatol 2009;7:234–238.
morbidly obese patients. Hepatology 2008;47:1916–1923. [36] Sanyal AJ, Banas C, Sargeant C, Luketic VA, Sterling RK, Stravitz RT, et al.
[15] Palekar NA, Naus R, Larson SP, Ward J, Harrison SA. Clinical model for Similarities and differences in outcomes of cirrhosis due to nonalcoholic
distinguishing nonalcoholic steatohepatitis from simple steatosis in steatohepatitis and hepatitis C. Hepatology 2006;43:682–689.
patients with nonalcoholic fatty liver disease. Liver Int [37] Angulo P, Hui JM, Marchesini G, Bugianesi E, George J, Farrell GC, et al.
2006;26:151–156. The NAFLD fibrosis score: a noninvasive system that identifies liver
[16] Poynard T, Ratziu V, Charlotte F, Messous D, Munteanu M, Imbert- fibrosis in patients with NAFLD. Hepatology 2007;45:846–854.
Bismut F, et al. Diagnostic value of biochemical markers (NashTest) for [38] Sterling RK, Lissen E, Clumeck N, Sola R, Correa MC, Montaner J, et al.
the prediction of non alcoholo steato hepatitis in patients with non- Development of a simple noninvasive index to predict significant
alcoholic fatty liver disease. BMC Gastroenterol 2006;6:34. fibrosis in patients with HIV/HCV coinfection. Hepatology
[17] Ulitsky A, Ananthakrishnan AN, Komorowski R, Wallace J, Surapaneni N, 2006;43:1317–1325.
Franco J, et al. A Noninvasive Clinical Scoring Model Predicts Risk of [39] Lin ZH, Xin YN, Dong QJ, Wang Q, Jiang XJ, Zhan SH, et al. Performance of
Nonalcoholic Steatohepatitis in Morbidly Obese Patients. Obes Surg the aspartate aminotransferase-to-platelet ratio index for the staging of
2010;20:685–691. hepatitis C-related fibrosis: an updated meta-analysis. Hepatology
[18] Kwok R, Tse YK, Wong GL, Ha Y, Lee AU, Ngu MC, et al. Systematic 2011;53:726–736.
review with meta-analysis: non-invasive assessment of non-alcoholic [40] Harrison SA, Oliver D, Arnold HL, Gogia S, Neuschwander-Tetri BA.
fatty liver disease–the role of transient elastography and plasma Development and validation of a simple NAFLD clinical scoring system
cytokeratin-18 fragments. Aliment Pharmacol Ther 2014;39:254–269. for identifying patients without advanced disease. Gut
[19] Feldstein AE, Wieckowska A, Lopez AR, Liu YC, Zein NN, McCullough AJ. 2008;57:1441–1447.
Cytokeratin-18 fragment levels as noninvasive biomarkers for nonalco- [41] Sun W, Cui H, Li N, Wei Y, Lai S, Yang Y, et al. Comparison of FIB-4 index,
holic steatohepatitis: a multicenter validation study. Hepatology NAFLD fibrosis score and BARD score for prediction of advanced fibrosis
2009;50:1072–1078. in adult patients with non-alcoholic fatty liver disease: A meta-analysis
[20] Musso G, Gambino R, Cassader M, Pagano G. Meta-analysis: Natural study. Hepatol Res 2016;46:862–870.
history of non-alcoholic fatty liver disease (NAFLD) and diagnostic [42] McPherson S, Hardy T, Dufour JF, Petta S, Romero-Gomez M, Allison M,
accuracy of non-invasive tests for liver disease severity. Ann Med et al. Age as a confounding factor for the accurate non-invasive
2011;43:617–649. diagnosis of advanced NAFLD fibrosis. Am J Gastroenterol
[21] Shen J, Chan HL, Wong GL, Chan AW, Choi PC, Chan HY, et al. Assessment 2017;112:740–751.
of non-alcoholic fatty liver disease using serum total cell death and [43] Poynard T, Morra R, Halfon P, Castera L, Ratziu V, Imbert-Bismut F, et al.
apoptosis markers. Aliment Pharmacol Ther 2012;36:1057–1066. Meta-analyses of FibroTest diagnostic value in chronic liver disease.
[22] Tamimi TIAR, Elgouhari HM, Alkhouri N, Yerian LM, Berk MP, Lopez R, BMC Gastroenterol 2007;7:40.
et al. An apoptosis panel for nonalcoholic steatohepatitis diagnosis. J [44] Leroy V, Sturm N, Faure P, Trocme C, Marlu A, Hilleret MN, et al.
Hepatol 2011;54:1224–1229. Prospective evaluation of FibroTest(R), FibroMeter(R), and HepaScore(R)
[23] Younossi ZM, Jarrar M, Nugent C, Randhawa M, Afendy M, Stepanova M, for staging liver fibrosis in chronic hepatitis B: comparison with
et al. A novel diagnostic biomarker panel for obesity-related nonalco- hepatitis C. J Hepatol 2014;61:28–34.
holic steatohepatitis (NASH). Obes Surg 2008;18:1430–1437. [45] Adams LA, George J, Bugianesi E, Rossi E, De Boer WB, van der Poorten D,
[24] Cao W, Zhao CY, Shen C, Wang YD. Cytokeratin 18, alanine aminotrans- et al. Complex non-invasive fibrosis models are more accurate than
ferase, platelets and triglycerides predict the presence of nonalcoholic simple models in non-alcoholic fatty liver disease. J Gastroen Hepatol
steatohepatitis. PLoS One 2013;8. 2011;26:1536–1543.
[25] Younossi ZM, Page S, Rafiq N, Birerdinc A, Stepanova M, Hossain N, et al. [46] Guha IN, Parkes J, Roderick P, Chattopadhyay D, Cross R, Harris S, et al.
A biomarker panel for non-alcoholic steatohepatitis (NASH) and NASH- Noninvasive markers of fibrosis in nonalcoholic fatty liver disease:
related fibrosis. Obes Surg 2011;21:431–439. Validating the European Liver Fibrosis Panel and exploring simple
[26] Chernbumroong S, Grove JI, Astbury S, Leeming DJ, Kaye P, Antoine DJ, markers. Hepatology 2008;47:455–460.
et al. Advanced machine learning techniques to identify a panel of [47] Lichtinghagen R, Pietsch D, Bantel H, Manns MP, Brand K, Bahr MJ. The
biomarkers that identify nonalcoholic steatohepatitis. Hepatology Enhanced Liver Fibrosis (ELF) score: normal values, influence factors and
2017;66:53a–54a. proposed cut-off values. J Hepatol 2013;59:236–242.
[27] Machado MV, Coutinho J, Carepa F, Costa A, Proenca H, Cortez-Pinto H. [48] Rosenberg WM, Voelker M, Thiel R, Becka M, Burt A, Schuppan D, et al.
How adiponectin, leptin, and ghrelin orchestrate together and correlate Serum markers detect the presence of liver fibrosis: a cohort study.
with the severity of nonalcoholic fatty liver disease. Eur J Gastroen Gastroenterology 2004;127:1704–1713.
Hepat 2012;24:1166–1172. [49] Tanwar S, Trembling PM, Guha IN, Parkes J, Kaye P, Burt AD, et al.
[28] Li H, Wang L, Yan X, Liu Q, Yu C, Wei H, et al. A proton nuclear magnetic Validation of terminal peptide of procollagen III for the detection and
resonance metabonomics approach for biomarker discovery in nonal- assessment of nonalcoholic steatohepatitis in patients with nonalco-
coholic fatty liver disease. J Proteome Res 2011;10:2797–2806. holic fatty liver disease. Hepatology 2013;57:103–111.
[29] Loomba R, Quehenberger O, Armando A, Dennis EA. Polyunsaturated [50] Nielsen MJ, Veidal SS, Karsdal MA, Orsnes-Leeming DJ, Vainer B, Gardner
fatty acid metabolites as novel lipidomic biomarkers for noninvasive SD, et al. Plasma Pro-C3 (N-terminal type III collagen propeptide)
diagnosis of nonalcoholic steatohepatitis. J Lipid Res 2015;56:185–192. predicts fibrosis progression in patients with chronic hepatitis C. Liver
[30] Puri P, Wiest MM, Cheung O, Mirshahi F, Sargeant C, Min HK, et al. The Int 2015;35:429–437.
plasma lipidomic signature of nonalcoholic steatohepatitis. Hepatology [51] Daniels SJ, Nielsen MJ, Krag A, Eslam M, Karsdal MA, Leeming DJ, et al.
2009;50:1827–1838. Serum Pro-C3 combined with clinical parameters is superior to estab-

10 Journal of Hepatology 2017 vol. xxx j xxx–xxx


Please cite this article in press as: Vilar-Gomez E, Chalasani N. Non-invasive assessment of non-alcoholic fatty liver disease: Clinical prediction rules and blood-based biomarkers. J Hepa-
tol (2017), https://doi.org/10.1016/j.jhep.2017.11.013
JOURNAL OF HEPATOLOGY

lished serological fibrosis tests at identifying patients with advanced advanced fibrosis in human nonalcoholic fatty liver disease. Cell Metab
fibrosis among patients with non-alcoholic fatty liver disease. J Hepatol 2017;25:1054–1062, e1055.
2017;66, S671–S671. [72] Hardy T, Zeybel M, Day CP, Dipper C, Masson S, McPherson S, et al.
[52] Boyle MP, Tiniakos DG, McPherson S, Ratziu V, Bedossa P, Hardy T, et al. Plasma DNA methylation: a potential biomarker for stratification of liver
Development and validation of the collagen neo-epitope biomarker Pro- fibrosis in non-alcoholic fatty liver disease. Gut 2017;66:1321–1328.
C3 ‘‘FIB-C3 Score” for detection and staging of advanced non-alcoholic [73] Pirola CJ, Fernandez Gianotti T, Castano GO, Mallardi P, San Martino J,
fatty liver disease in a large international multi-centre patient cohort. Ledesma Mora Gonzalez Lopez, et al. Circulating microRNA signature in
Hepatology 2017;66:54a–55a. non-alcoholic fatty liver disease: from serum non-coding RNAs to liver
[53] Pimentel CF, Otsubo T, Challies TL, Nasser I, Francescucci A, Lai M. histology and disease pathogenesis. Gut 2015;64:800–812.
Combination of serum HA, CK18 and TIMP-1 predicts advanced fibrosis [74] Sookoian S, Pirola CJ. Cell-free DNA methylation as liquid biopsy for the
in nonalcoholic fatty liver disease. Hepatology 2015;62:1260a–1261a. assessment of fibrosis in patients with nonalcoholic steatohepatitis: a
[54] Zaman A, Rosen HR, Ingram K, Corless CL, Oh E, Smith K. Assessment of gap between innovation and implementation. Hepatol Surg Nutr
FIBROSpect II to detect hepatic fibrosis in chronic hepatitis C patients. 2017;6:117–121.
Am J Med 2007;120:280, e289–e214. [75] Sumida Y, Yoneda M, Hyogo H, Yamaguchi K, Ono M, Fujii H, et al. A
[55] Abdelmalek MF, Diehl AM, Guy CD, Portenier D, Sudan R, Li XJ, et al. simple clinical scoring system using ferritin, fasting insulin, and type IV
Serum-based biomarker accurately stratifies hepatic fibrosis in patients collagen 7S for predicting steatohepatitis in nonalcoholic fatty liver
with nonalcoholic steatohepatitis. Hepatology 2017;66:55a–56a. disease. J Gastroenterol 2011;46:257–268.
[56] Forns X, Ampurdanes S, Llovet JM, Aponte J, Quinto L, Martinez-Bauer E, [76] Verdam FJ, Dalling JW, Driessen A, de Jonge C, Moonen EJC, van Berkel
et al. Identification of chronic hepatitis C patients without hepatic JBN, et al. Non-alcoholic steatohepatitis: A non-invasive diagnosis by
fibrosis by a simple predictive model. Hepatology 2002;36:986–992. analysis of exhaled breath. J Hepatol 2013;58:543–548.
[57] Kim D, Kim WR, Kim HJ, Therneau TM. Association between noninvasive [77] Banasch M, Ellrichmann M, Tannapfel A, Schmidt WE, Goetze O. The
fibrosis markers and mortality among adults with nonalcoholic fatty non-invasive (13)C-methionine breath test detects hepatic mitochon-
liver disease in the United States. Hepatology 2013;57:1357–1365. drial dysfunction as a marker of disease activity in non-alcoholic
[58] Angulo P, Bugianesi E, Bjornsson ES, Charatcharoenwitthaya P, Mills PR, steatohepatitis. Eur J Med Res 2011;16:258–264.
Barrera F, et al. Simple noninvasive systems predict long-term outcomes [78] Feldstein AE, Alkhouri N, De Vito R, Alisi A, Lopez R, Nobili V. Serum
of patients with nonalcoholic fatty liver disease. Gastroenterology cytokeratin-18 fragment levels are useful biomarkers for nonalcoholic
2013;145:782–789, e784. steatohepatitis in children. Am J Gastroenterol 2013;108:1526–1531.
[59] Unalp-Arida A, Ruhl CE. Liver fibrosis scores predict liver disease [79] Feldstein AE, Lopez R, Tamimi TAR, Yerian L, Chung YM, Berk M, et al.
mortality in the United States population. Hepatology 2017;66:84–95. Mass spectrometric profiling of oxidized lipid products in human
[60] Parkes J, Roderick P, Harris S, Day C, Mutimer D, Collier J, et al. Enhanced nonalcoholic fatty liver disease and nonalcoholic steatohepatitis. J Lipid
liver fibrosis test can predict clinical outcomes in patients with chronic Res 2010;51:3046–3054.
liver disease. Gut 2010;59:1245–1251. [80] Harrison S, Praca E, Brozek J, Cordonnier G, Sudrik FB, Roudot A, et al. A
[61] Vesterhus M, Hov JR, Holm A, Schrumpf E, Nygard S, Godang K, et al. new non-invasive diagnostic score to monitor change in disease activity
Enhanced liver fibrosis score predicts transplant-free survival in primary and predict fibrosis evolution in patients with NASH. J Hepatol 2017;66,
sclerosing cholangitis. Hepatology 2015;62:188–197. S110–S110.
[62] Harrison SA, Abdelmalek MF, Diehl AM, Caldwell S, Shiffman ML, Ghalib [81] Chowdhury SD, Ramakrishna B, Eapen CE, Goel A, Zachariah UG,
R, et al. Prospective validation of the Enhanced Liver Fibrosis (ELF) test Chandramohan A, et al. Fibrosis in non-alcoholic fatty liver disease:
for the prediction of disease progression in patients with nonalcoholic correlation with simple blood indices and association with tumor
steatohepatitis (NASH) and advanced fibrosis. Hepatology necrosis factor-alpha polymorphisms. Trop Gastroenterol
2017;66:1120a–1121a. 2013;34:31–35.
[63] Karsdal MA, Henriksen K, Nielsen MJ, Byrjalsen I, Leeming DJ, Gardner S, [82] McPherson S, Stewart SF, Henderson E, Burt AD, Day CP. Simple non-
et al. Fibrogenesis assessed by serological type III collagen formation invasive fibrosis scoring systems can reliably exclude advanced fibrosis
identifies patients with progressive liver fibrosis and responders to a in patients with non-alcoholic fatty liver disease. Gut
potential antifibrotic therapy. Am J Physiol-Gastr L 2016;311: 2010;59:1265–1269.
G1009–G1017. [83] Williams AL, Hoofnagle JH. Ratio of serum aspartate to alanine
[64] Vilar-Gomez E, Martinez-Perez Y, Calzadilla-Bertot L, Torres-Gonzalez A, aminotransferase in chronic hepatitis. Relationship to cirrhosis. Gas-
Gra-Oramas B, Gonzalez-Fabian L, et al. Weight loss through lifestyle troenterology 1988;95:734–739.
modification significantly reduces features of nonalcoholic steatohep- [84] Poynard T, Ratziu V, Naveau S, Thabut D, Charlotte F, Messous D, et al.
atitis. Gastroenterology 2015;149:367–378, [e365; quiz e314–e365]. The diagnostic value of biomarkers (SteatoTest) for the prediction of
[65] Vilar-Gomez E, Calzadilla-Bertot L, Friedman SL, Gra-Oramas B, Gonza- liver steatosis. Comp Hepatol 2005;4:10.
lez-Fabian L, Lazo-Del Vallin S, et al. Serum biomarkers can predict a [85] Lassailly G, Caiazzo R, Hollebecque A, Buob D, Leteurtre E, Arnalsteen L,
change in liver fibrosis 1 year after lifestyle intervention for biopsy- et al. Validation of noninvasive biomarkers (FibroTest, SteatoTest, and
proven NASH. Liver Int 2017;37:1887–1896. NashTest) for prediction of liver injury in patients with morbid obesity.
[66] Ladaru A, Balanescu P, Stan M, Codreanu I, Anca IA. Candidate proteomic Eur J Gastroenterol Hepatol 2011;23:499–506.
biomarkers for non-alcoholic fatty liver disease (steatosis and non- [86] Ratziu V, Massard J, Charlotte F, Messous D, Imbert-Bismut F, Bonyhay L,
alcoholic steatohepatitis) discovered with mass-spectrometry: a sys- et al. Diagnostic value of biochemical markers (FibroTest-FibroSURE) for
tematic review. Biomarkers 2016;21:102–114. the prediction of liver fibrosis in patients with non-alcoholic fatty liver
[67] Bell LN, Theodorakis JL, Vuppalanchi R, Saxena R, Bemis KG, Wang M, disease. BMC Gastroenterol 2006;6.
et al. Serum proteomics and biomarker discovery across the spectrum of [87] Loaeza-del-Castillo A, Paz-Pineda F, Oviedo-Cardenas E, Sanchez-Avila F,
nonalcoholic fatty liver disease. Hepatology 2010;51:111–120. Vargas-Vorackova F. AST to platelet ratio index (APRI) for the noninva-
[68] Bell LN, Lee L, Saxena R, Bemis KG, Wang M, Theodorakis JL, et al. Serum sive evaluation of liver fibrosis. Ann Hepatol 2008;7:350–357.
proteomic analysis of diet-induced steatohepatitis and metabolic syn- [88] Nobili V, Parkes J, Bottazzo G, Marcellini M, Cross R, Newman D, et al.
drome in the Ossabaw miniature swine. Am J Physiol Gastrointest Liver Performance of ELF serum markers in predicting fibrosis stage in
Physiol 2010;298:G746–G754. pediatric non-alcoholic fatty liver disease. Gastroenterology
[69] Wood GC, Chu X, Argyropoulos G, Benotti P, Rolston D, Mirshahi T, et al. 2009;136:160–167.
A multi-component classifier for nonalcoholic fatty liver disease [89] Angulo P, Hui JM, Marchesini G, Bugianesi E, George J, Farrell GC, et al.
(NAFLD) based on genomic, proteomic, and phenomic data domains. The NAFLD fibrosis score: A noninvasive system that identifies liver
Sci Rep 2017;7:43238. fibrosis in patients with NAFLD. Hepatology 2007;45:846–854.
[70] Decaris ML, Li KW, Emson CL, Gatmaitan M, Liu S, Wang Y, et al. [90] Shah AG, Lydecker A, Murray K, Tetri BN, Contos MJ, Sanyal AJ, et al.
Identifying nonalcoholic fatty liver disease patients with active fibrosis Comparison of noninvasive markers of fibrosis in patients with nonalco-
by measuring extracellular matrix remodeling rates in tissue and blood. holic fatty liver disease. Clin Gastroenterol Hepatol 2009;7:1104–1112.
Hepatology 2017;65:78–88. [91] Lee TH, Han SH, Yang JD, Kim D, Ahmed M. Prediction of advanced
[71] Loomba R, Seguritan V, Li W, Long T, Klitgord N, Bhatt A, et al. Gut fibrosis in nonalcoholic fatty liver disease: an enhanced model of BARD
microbiome-based metagenomic signature for non-invasive detection of score. Gut Liver 2013;7:323–328.

Journal of Hepatology 2017 vol. xxx j xxx–xxx 11


Please cite this article in press as: Vilar-Gomez E, Chalasani N. Non-invasive assessment of non-alcoholic fatty liver disease: Clinical prediction rules and blood-based biomarkers. J Hepa-
tol (2017), https://doi.org/10.1016/j.jhep.2017.11.013

You might also like