You are on page 1of 9

Contemporary Clinical Trials 97 (2020) 106174

Contents lists available at ScienceDirect

Contemporary Clinical Trials


journal homepage: www.elsevier.com/locate/conclintrial

Semaglutide for the treatment of non-alcoholic steatohepatitis: Trial design T


and comparison of non-invasive biomarkers
Stephen A. Harrisona, , Salvatore Calannab, Kenneth Cusic,d, Martin Linderb, Takeshi Okanouee,

Vlad Ratziuf, Arun Sanyalg, Anne-Sophie Sejlingb, Philip N. Newsomeh,i


a
Radcliffe Department of Medicine, University of Oxford, Oxford, OX3 9DU, UK
b
Novo Nordisk A/S, 2860 Søborg, Denmark
c
Division of Endocrinology, Diabetes & Metabolism, University of Florida, Gainesville, FL 32608, USA
d
Endocrinology, Diabetes and Metabolism, Malcom Randall VA Medical Center, Gainesville, FL 32608, USA
e
Department of Gastroenterology & Hepatology, Saiseikai Suita Hospital, Suita, Japan
f
Sorbonne University, ICAN – Institute for Cardiometabolism and Nutrition, Hôpital Pitié Salpêtrière, 75013 Paris, France
g
Division of Gastroenterology, Hepatology and Nutrition, Department of Internal Medicine, Virginia Commonwealth University, Richmond, VA 23284, USA
h
National Institute for Health Research Birmingham Biomedical Research Centre and Liver Unit at University Hospitals Birmingham NHS Foundation Trust, Birmingham,
UK
i
Centre for Liver & Gastrointestinal Research, Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham B15 2TT, UK

ARTICLE INFO ABSTRACT

Keywords: Non-alcoholic steatohepatitis (NASH) is a chronic liver disease. There is a clear need to develop pharmacological
Biomarkers treatment for patients with NASH as well as biomarkers that can diagnose the disease. We describe a trial of
Clinical trial protocol semaglutide treatment for NASH, identify key patient characteristics and compare the relationship of patient
General diabetes characteristics and non-invasive biomarkers/scores.
Hepatology
NCT02970942 is a randomised, double-blind, placebo-controlled, multi-national Phase 2 trial of daily sub­
Non-alcoholic fatty liver disease
cutaneous semaglutide (0.1 mg, 0.2 mg, 0.4 mg) in patients with biopsy-confirmed NASH, F1–F3 fibrosis, NAFLD
Semaglutide
Activity Score ≥ 4, and body mass index (BMI) > 25 kg/m2. Exploratory analyses were performed to evaluate
correlations between baseline parameters and biomarkers in NASH.
Mean (standard deviation [SD]) age of 320 randomised patients was 55 (11) years, mean BMI was
36 (6) kg/m2, and 199 (62%) had type 2 diabetes. Of the total patients, 28% had F1 fibrosis, 23% had F2 fibrosis
and 49% had F3 fibrosis. The highest area under the receiver operating characteristic curve (0.69) for accuracy
in classifying fibrosis stage, F2–3 versus F1, was observed for Fib-4 and Enhanced Liver Fibrosis (ELF). No
substantial correlation between BMI or other clinical or biochemical parameters and fibrosis stage was observed.
In this large Phase 2 trial of semaglutide treatment for NASH, the clinical profile of enrolled patients was
typical for patients with NASH. Of the investigated biomarkers/scores, ELF and Fib-4 showed the most apparent
correlation in classifying fibrosis stage, but had only moderate predictive value.

1. Introduction mortality [3]. However, up to 30% of patients with NAFLD progress


from steatosis to NASH [3]. Hence, the estimated global prevalence of
Non-alcoholic steatohepatitis (NASH) is a severe form of non-alco­ NASH is 1.5–6.5% and an increasing number of patients are developing
holic fatty liver disease (NAFLD) [1,2]. NAFLD encompasses a number NASH-related end-stage liver disease [2,4]. NASH is defined as the
of pathological conditions characterised by excessive hepatic fat de­ presence of hepatic steatosis and inflammation with hepatocyte injury
position and is a leading cause of liver disease in Western countries [1]. (ballooning) with or without fibrosis [2]. While NASH is often asymp­
The majority of patients with NAFLD have isolated steatosis, which tomatic, it is a complex progressive condition that can develop over
carries a relatively benign prognosis, with no overall increase in time into end-stage liver diseases and hepatocellular carcinoma [1,2];

Corresponding author.

E-mail addresses: stephenharrison87@gmail.com (S.A. Harrison), sca@novonordisk.com (S. Calanna), Kenneth.Cusi@medicine.ufl.edu (K. Cusi),
MLIX@novonordisk.com (M. Linder), okanoue@suita.saiseikai.or.jp (T. Okanoue), vlad.ratziu@inserm.fr (V. Ratziu), arun.sanyal@vcuhealth.org (A. Sanyal),
asji@novonordisk.com (A.-S. Sejling), P.N.Newsome@bham.ac.uk (P.N. Newsome).

https://doi.org/10.1016/j.cct.2020.106174
Received 29 May 2020; Received in revised form 6 October 2020; Accepted 6 October 2020
Available online 08 October 2020
1551-7144/ © 2020 The Authors. Published by Elsevier Inc. This is an open access article under the CC BY license
(http://creativecommons.org/licenses/by/4.0/).
S.A. Harrison, et al. Contemporary Clinical Trials 97 (2020) 106174

therefore, resolution of NASH is a major endpoint in current pharma­ (ClinicalTrials.gov identifier NCT02970942) and identify key char­
cological studies. acteristics of patients with NASH from analysis of the enrolled popu­
NASH has consistently been identified as one of the leading causes lation. Moreover, we analysed how to potentially diagnose F2–F3 fi­
of liver transplantation across a range of patients globally [5–7]. Type 2 brosis by assessing correlations between the key patient characteristics
diabetes mellitus (T2D) and metabolic syndrome are well-known risk and specific biomarkers, including FibroScan®, Fib-4 and ELF, with fi­
factors for advanced liver disease and have been shown to exacerbate brosis stage.
NASH, and, in turn, the presence of NASH has been shown to have a
greater cardiovascular risk than in patients with isolated steatosis
2. Methods
[1,2,8]. The development of cirrhosis due to NASH is also associated
with poor long-term prognosis [3]. Approximately 20–40% of patients
2.1. Trial design
with NASH will develop significant liver fibrosis, resulting in cirrhosis
in 10–30% of those patients [3,9].
The NCT02970942 trial was a randomised, double-blind, placebo-
Subjects with NASH are often asymptomatic and there is low
controlled, six-armed, parallel group, multi-national dose-finding trial
awareness among both subjects and care providers [3,10–12]. Liver
comparing semaglutide with placebo in patients with NASH. A planned
biopsy is currently the only accepted and definitive method of NASH
total of 823 patients were to be screened and, on the basis of an as­
diagnosis [1,2]. The presence of hepatocellular injury, inflammation
sumed 65% screening failure rate, 288 patients were to be randomised.
and fibrosis are key histological features that distinguish NASH from
isolated steatosis [3]. However, a liver biopsy can be impractical, prone
to sampling error and complications, and costly [2]. The enrolment of 2.2. Randomisation and treatment
study subjects into NASH clinical trials is challenging as subjects can be
expected to undertake multiple liver biopsies to confirm the presence of Randomisation for all patients was controlled by the interactive web
NASH [12]. Moreover, high screen failure rates have been reported in response system. Patients were randomised 3:3:3:1:1:1 to receive either
NASH studies with up to 50% of screened subjects not meeting the once-daily subcutaneous semaglutide (0.1 mg, 0.2 mg, 0.4 mg) or cor­
required eligibility criteria [13,14]. Thus, there is a pressing need to responding injection volumes of placebo for 72 weeks of treatment
identify a non-invasive test or NASH biomarkers with predictive cap­ (Fig. 1). Dose escalation occurred until the target dose was reached; the
ability for the diagnosis of NASH and for selection of patients for starting dose for all treatment arms was 0.05 mg per day with doses
treatment and monitoring [4,15]. increasing step-wise every fourth week, first to 0.1, then 0.2, 0.3 and
Non-invasive laboratory biomarker models, to indicate less ad­ 0.4 mg. Patients did not change the dose once the randomised target
vanced liver fibrosis, have previously been investigated and include dose was reached. The trial was double-blinded within dose levels;
serum aspartate aminotransferase (AST)-to-platelet ratio index (APRI), however, the dose levels were not blinded between each other due to
fibrosis-4 (Fib-4), NAFLD Fibrosis Score (NFS) and Enhanced Liver the different dose escalations, target doses, and volumes required.
Fibrosis (ELF) score [16]. Of these markers, Fib-4 and ELF scores have Randomisation was stratified in five strata based on region
shown diagnostic accuracy (area under the receiver operating char­ (Japanese or non-Japanese) and, within the non-Japanese group, based
acteristic curve [AUROC] > 0.8) for the assessment of fibrosis [16,17]. on diabetes status at screening (with or without T2D) and fibrosis stage
NFS, Fib-4 and ELF score predict overall mortality, cardiovascular for baseline liver biopsy (F1/F2 or F3). Patients in all treatment groups,
mortality and liver-related mortality; additionally, NFS predicts in­ including placebo, received nutritional and physical activity counsel­
cident diabetes, and changes in NFS are associated with mortality [1]. ling by a member of the trial team according to local practice.
The tests perform best at distinguishing advanced fibrosis from other
fibrosis stages with negative predictive values higher than positive
2.3. Assessments
predictive values, thus allowing first-line risk stratification to exclude
severe disease [1,18,19]. Additionally, cytokeratin-18 (CK-18) frag­
Assessments of randomised patients were carried out throughout the
ments have also been investigated extensively as biomarkers for stea­
trial and included body weight, waist circumference, vital signs, liver
tohepatitis [2].
enzymes (ALT, AST, gamma-glutamyl transferase [GGT]), hsCRP, lipids
There are currently no FDA-approved pharmacological treatments
(total cholesterol, free fatty acids, high-density lipoprotein cholesterol,
for patients with NASH, and therefore there is a substantial unmet
low-density lipoprotein cholesterol, triglycerides, very low-density li­
medical need. A growing cohort of clinical development programmes
poprotein cholesterol) and glucose metabolism (fasting plasma glucose,
evaluating novel pharmacotherapies for NASH is emerging, primarily
HbA1c, fasting insulin, fasting glucagon, and homeostatic model as­
focused on demonstrating improvement in histological characteristics
sessment of insulin resistance). FibroScan® measurements of liver
of NASH, including steatosis, steatohepatitis and fibrosis [20,21]. In
stiffness and liver steatosis (with controlled attenuation parameter)
particular, the use of glucagon-like peptide-1 (GLP-1) analogues is
were taken if FibroScan® equipment was available.
currently being studied as a possible treatment option in NASH. GLP-1
receptor agonists have pleiotropic effects on several aspects of meta­
bolic syndrome relevant to treatment of NASH, including body weight, 2.4. Outcome measures
inflammation, and glucose and lipid metabolism [22–24]. Liraglutide, a
GLP-1 analogue indicated for treatment of T2D and weight manage­ The primary endpoint was NASH resolution without worsening of
ment, has been shown to promote NASH resolution without worsening fibrosis (defined by an increase of ≥ 1 stage of the Kleiner fibrosis
of fibrosis [25]. Semaglutide, also a GLP-1 analogue indicated for T2D, classification) after 72 weeks. The resolution of NASH was defined per
is under evaluation for both weight management and NASH treatment the NASH Clinical Research Network (CRN) criteria as “no more than
[26]. Semaglutide has been shown to significantly reduce alanine mild residual inflammatory cells (0–1) and no ballooning (0)” based on
aminotransferase (ALT) and high sensitivity C-reactive protein (hsCRP) comprehensive interpretation by two independent pathologists (central
in a dose-dependent manner in patients with obesity or T2D [26]. reading) blinded to treatment allocation. The secondary endpoints in­
With an increasing number of patients developing NASH-related cluded safety endpoints as well as changes in: liver-related histological
end-stage liver disease, there is a need for effective pharmacological parameters (including at least one stage of liver fibrosis improvement
treatments, and a need for non-invasive biomarkers for those at high with no worsening of NASH), biomarkers of NASH disease, weight-re­
risk of liver-related complications. Here we describe the study design of lated and glucose-metabolism-related parameters, cardiovascular risk
a Phase 2 trial investigating semaglutide for the treatment of NASH factors and patient-reported outcomes.

2
S.A. Harrison, et al. Contemporary Clinical Trials 97 (2020) 106174

Placebo*

• Biopsy-confirmed
NASH
• F1–F3 Placebo*
• NAS ≥4
• With or without
T2D

Placebo*
7-week FU

4 8 12 18 72
Liver biopsy Time (weeks) Liver biopsy
(recent or new)

from 0.05 mg
every 4 weeks

Fig. 1. Trial design.


a
Matching the active arm with corresponding injection volumes once daily.
F1–F3 fibrosis stage 1 to 3, FU follow-up, NAFLD non-alcoholic fatty liver disease, NAS NAFLD Activity Score, NASH non-alcoholic steatohepatitis, s.c. subcutaneous,
T2D type 2 diabetes

2.5. Statistical analysis NASH trials, for which most have included patients with fibrosis stage
0–3 [28], or 1–3 [13]. The trial protocol is available as part of the
The target sample size of 288 patients was based on an aim to detect electronic supplementary material.
a difference in the primary endpoint between the highest semaglutide The trial was conducted in accordance with Good Clinical Practice
dose and placebo with 90% power. After accounting for trial with­ guidelines and principles of the Declaration of Helsinki. An independent
drawals who would be imputed as non-responders, the proportion of ethics committee or institutional review board at each site approved the
primary endpoint responders was assumed to be 38% with the highest trial protocol and any subsequent amendments. Informed consent was
semaglutide dose versus 14% with placebo. obtained from every participant before any trial-related activities took
The primary analysis was based on the Cochran–Mantel–Haenszel place, including activities to determine suitability for the trial.
test [27], which was performed separately for the comparisons between
each of the semaglutide and placebo arms. The three different placebo
2.7. Biomarker assessments
arms were pooled into one placebo arm in the statistical analyses. The
test adjusted for baseline diabetes status and baseline fibrosis stage. To
One liver biopsy was performed during the screening period (on the
account for multiple testing, the comparisons were evaluated hier­
condition that no biopsy within 21 weeks before screening was avail­
archically according to descending semaglutide dose using a global
able) and one biopsy is scheduled at 72 weeks after randomisation.
significance level of 5%.
Exploratory biomarkers (including CK-18 fragments, and monocyte
chemoattractant protein 1 [MCP-1]) and algorithms (such as Fib-4
score and NFS) were assessed. The ELF panel includes measures of
2.6. Trial population
hyaluronic acid, tissue inhibitor of matrix metalloproteinase-1 and
amino terminal propeptide of procollagen type III and was calculated
Patients were eligible if they were 18–75 years of age (20–75 years
using the original algorithm by Guha et al. 2008 [18,29]. The Fib-4
of age in Japan), and had histological evidence of NASH based on
score algorithm incorporates measures of platelets, ALT, AST and age
central pathologist evaluation of a liver biopsy obtained up to 21 weeks
[29]. The calculation of the NFS requires values for age, hypergly­
before screening. Other inclusion criteria included an NAFLD Activity
caemia, BMI, platelet count, albumin, and AST/ALT ratio [29]. In­
Score (NAS) of ≥ 4 with a score of 1 or more in each sub-component
formation on adverse events and hypoglycaemic episodes was collected
(steatosis, hepatocyte ballooning, and lobular inflammation) of the
throughout the trial period.
score; a NASH fibrosis stage of 1, 2 or 3 according to the NASH CRN
fibrosis staging system; HbA1c ≤ 10%; and a stable weight with a body
mass index (BMI) > 25 kg/m2. Patients were excluded if they had 2.8. Exploratory correlative and diagnostic analyses
causes of chronic liver disease other than NASH; positive hepatitis B
surface antigen, anti-HIV or hepatitis C virus-RNA; ALT or AST le­ The full analysis set was used in the exploratory correlative and
vels > 5 × the upper limit of normal; significant alcohol consumption; diagnostic analyses and included all randomised participants.
type 1 diabetes or current or history of pancreatitis (chronic or acute); Exploratory analyses were performed to evaluate the diagnostic accu­
or treatment with GLP-1 receptor agonists or sodium glucose trans­ racy of baseline parameters and biomarkers to classify the fibrosis at
porter-2 inhibitors within 90 days prior to screening or the liver biopsy. baseline. The classes of fibrosis stage were defined by a dichotomous
In the original protocol, only patients with a NASH fibrosis stage of split (e.g. F3 vs F1–2). A positive correlation between biomarker and
2 or 3 were eligible. However, a protocol amendment was made on fibrosis stage was assumed, i.e. a high value of the biomarker was as­
1 March 2018 to include patients with F1 to reflect the view that some sumed to indicate a high fibrosis stage and vice versa. Corresponding
patients may be “fast progressors” who could benefit from pharmaco­ analyses were also performed to evaluate the accuracy in classifying
logical treatment. This amendment was also in line with other Phase 2 NAS and its components. Geometric mean and coefficient of variation

3
S.A. Harrison, et al. Contemporary Clinical Trials 97 (2020) 106174

were presented for baseline characteristics and biomarker parameters, compared with F1 or F2. Patients aged ≥ 65 years were more likely to
unless otherwise specified in the results. have F3 fibrosis (n = 36, 62% of the 58 patients aged ≥ 65 years) ra­
The receiver operating characteristic (ROC) curve was calculated to ther than F1 (n = 6, 10%) or F2 (n = 16, 28%), and a slightly greater
describe the relationship of the true positive rate (sensitivity) versus the proportion of patients with F3 fibrosis had T2D compared with those
false positive rate (1 - specificity) as the cut-point on the biomarker was with F1 or F2. No obvious relationship between BMI and fibrosis stage
moved. The AUROC with the associated approximative 95% confidence was observed.
interval (CI) was estimated using the Mann–Whitney statistic. If the The demographics and baseline characteristics of the enrolled pa­
AUROC was greater than 0.5 (supporting a positive correlation), an tients were also assessed in relation to the NAS (Table S1 in the elec­
optimal cut-point was determined by finding the point which max­ tronic supplementary material). In patients with a NAS of 7–8, serum
imised Youden's index (sensitivity + specificity - 1). The results were levels of ALT and AST appeared to be higher and T2D was less prevalent
summarised by presenting the AUROC with 95% CI and, where ap­ compared with those with a NAS of 4–6. Serum levels of free fatty acids,
plicable, sensitivity, specificity, positive predictive value (PPV) and triglycerides and fasting plasma glucose did not differ substantially as a
negative predictive value (NPV) at the optimal cut-point. function of NAS.
In the total population, plasma CK-18 fragment M30 (cleaved CK-
2.9. Patient and public involvement 18) levels were 537 U/L and CK-18 fragment M65 (intact CK-18) levels
were 1030 U/L (Table 2). Plasma concentrations of CK-18 fragment
No patients or the public were involved in the study trial design or M30 and M65 were higher in patients with F3 fibrosis compared with
dissemination of results. F1 and F2. Circulating MCP-1 levels were similar in patients with dif­
ferent fibrosis stages. Mean NFS for the enrolled population was −0.6
3. Results (Table 2) and did not greatly differ between the different fibrosis stages.
FibroScan® values were 9.2 kPa for patients with F1 fibrosis, 10.3 kPa
3.1. Patient characteristics for patients with F2 fibrosis and 11.8 kPa for patients with F3 fibrosis
(Table 2). ELF score values ranged from −0.4 in patients with F1 fi­
The study was completed in March 2020 and met its primary end brosis to 0.2 in patients with F3 fibrosis. The Fib-4 score appeared to
point; these results will be reported separately. A total of 320 patients increase with the severity of fibrosis stage (Table 2).
with NASH were randomised in the trial; 28% had F1 fibrosis, 23% had
F2 fibrosis and 49% had F3 fibrosis (Table 1). At baseline, the mean
(standard deviation [SD]) NAS for the total study population was 4.9 3.2. Accuracy of baseline characteristics and biomarkers in classifying
(0.89). The mean age was 55 years and the mean BMI was 36 kg/m2; fibrosis stage
62% of the patients had T2D (Table 2). Serum levels of ALT and AST
were 54 U/L and 43 U/L, respectively. Patients had serum levels of free The accuracy of classifying fibrosis stage (F2–3 vs F1 and F3 vs
fatty acids of 0.5 mmol/L and triglycerides of 1.9 mmol/L. FibroScan® F1–2) based on baseline characteristics and biomarkers was assessed
score for the 212 patients with available data was 11 kPa (SD 57.4). (Table 3). The cut-points were determined by maximising Youden
Serum levels of free fatty acids, triglycerides and fasting plasma glucose index, and the sensitivity and specificity were evaluated at the specified
did not differ substantially between the fibrosis stages. There appeared cut-point. The highest AUROC of 0.69 was observed for Fib-4 for ac­
to be a slight trend for increased ALT and AST levels with F3 fibrosis, curacy in classifying fibrosis stage, F2–3 versus F1, with a sensitivity of
64% and specificity of 70% at a cut-point of 1.3. An AUROC of 0.69 was
also observed for ELF for accuracy in classifying F2–3 versus F1 fibrosis
Table 1 with a sensitivity of 62% and specificity of 68% at a cut-point of −0.2.
Histopathological characteristics. The next highest AUROC of 0.63 was reported for liver stiffness by
Characteristic Total FAS, n (%) FibroScan®, F2–3 versus F1, with a sensitivity of 89% and specificity of
33% at a cut-point of 7.3. For AST alone, the AUROC was 0.63 for F2–3
N = 320 versus F1 (sensitivity 57%; specificity 68% at a cut-point of 42 U/L).
The third highest AUROC was reported as 0.60 for NFS F2–3 versus F1
Fibrosis stage
F1 90 (28%) with a sensitivity of 66% and specificity of 52% at a cut-point of −0.9.
F2 72 (23%) Accuracy of Fib-4 and ELF for classifying fibrosis stage (F2–3 vs F1)
F3 158 (49%) was slightly higher for patients with T2D (Fib-4 AUROC 0.73, ELF
Overall NAFLD Activity Score, 0–8 AUROC 0.71) than those without (Fib-4 AUROC 0.63, ELF AUROC
Mean, SD 5 (0.9)
0.67). Accuracy of both ALT and AST for fibrosis stage (F2–3 vs F1 and
NAFLD Activity Score
4 133 (42%) F3 vs F1–2) was also slightly higher for patients with T2D when com­
5 114 (36%) pared with patients without T2D (data not shown). Limited accuracy for
6 59 (18%) classifying fibrosis stage (AUROC < 0.5) or any other histopathological
7 12 (4%)
characteristic (NAS, steatosis score, hepatocyte ballooning score, lob­
8 2 (0.6%)
Steatosis score
ular inflammation score) was observed for BMI (data not shown).
1 90 (28%)
2 162 (51%)
3 68 (21%) 3.3. Boxplots of patient baseline characteristics and biomarkers
Ballooning score
1 218 (68%)
2 102 (32%) Boxplots of biomarkers are provided by fibrosis stage in Fig. 2 and
Lobular inflammation score additional boxplots of baseline characteristics by fibrosis stage are
1 135 (42%) available in Figs. S1–S4 in the electronic supplementary material. The
2 174 (54%)
box plots show the median, and 1st and 3rd quartiles. The whiskers
3 11 (3%)
extend to minimum and maximum values (including possible outliers).
FAS full analysis set, N number of patients, NAFLD non-al­ In agreement with the ROC analyses, the most apparent correlation
coholic fatty liver disease, SD standard deviation. with fibrosis stage was seen for ELF and Fib-4.

4
S.A. Harrison, et al. Contemporary Clinical Trials 97 (2020) 106174

Table 2
Demographics and baseline characteristics by fibrosis stage.
Characteristic Fibrosis stage, n (%) Total FAS, n (%)

1 2 3 N = 320

n = 90 n = 72 n = 158

Female 51 (57%) 47 (65%) 96 (61%) 194 (61%)


Age group, years
18–64 84 (93%) 56 (78%) 122 (77%) 262 (82%)
≥ 65–74 6 (7%) 16 (22%) 33 (21%) 55 (17%)
≥ 75 0 (0%) 0 (0%) 3 (2%) 3 (1%)
Mean (SD) 52 (9) 56 (11) 56 (11) 55 (11)
Ethnicity
Hispanic or Latino 10 (11%) 13 (18%) 17 (11%) 40 (13%)
NA 2 (2%) 4 (6%) 11 (7%) 17 (5%)
Not Hispanic or Latino 78 (87%) 55 (76%) 130 (82%) 263 (82%)
BMI, kg/m2
< 25 0 (0%) 1 (1%) 3 (2%) 4 (1%)
≥ 25 to < 30 14 (16%) 12 (17%) 38 (24%) 64 (20%)
≥ 30 to < 35 19 (21%) 22 (31%) 46 (29%) 87 (27%)
≥ 35 to < 40 32 (36%) 19 (26%) 35 (22%) 86 (27%)
≥ 40 25 (28%) 18 (25%) 36 (23%) 79 (25%)
Mean (SD) 37 (6) 36 (7) 35 (6) 36 (6)
ALT, U/L 51 (64) 49 (58) 59 (58) 54 (60)
AST, U/L 37 (50) 39 (50) 49 (49) 43 (51)
GGT, U/L 57 (72) 54 (78) 72 (79) 63 (79)
HbA1c, mmol/mol
Mean (SD) 51 (14) 50 (14) 50 (13) 50 (13)
Total bilirubin, umol/L 11 (50) 10 (49) 11 (47) 11 (48)
Alkaline phosphatase, U/L 81 (27) 82 (32) 83 (33) 82 (31)
Total chol., mmol/La 5 (23) 5 (29) 5 (23) 5 (25)
High-density lipoprotein chol., mmol/Lb 1 (23) 1 (25) 1 (24) 1 (24)
Very low-density lipoprotein chol., mmol/Lb 0.9 (48) 0.9 (45) 0.8 (45) 0.8 (47)
Triglycerides, mmol/Lb 2.1 (55.7) 2.0 (48.4) 1.7 (47.3) 1.9 (50.9)
Free fatty acids, mmol/Lc 0.5 (46.8) 0.5 (56.6) 0.6 (50.3) 0.5 (50.6)
Type 2 diabetes mellitus, n (%) 49 (54%) 40 (56%) 110 (70%) 199 (62%)
Fasting plasma glucose, mmol/L
Mean (SD)d 7.4 (2.5) 7.6 (3.0) 7.4 (2.2) 7.4 (2.5)
CK-18 fragments M30, U/Lf 482 (86) 463 (87) 609 (73) 537 (81)
CK-18 fragments M65, U/Lf 906 (79) 934 (70) 1158 (60) 1030 (69)
MCP-1, pg/mLf 361 (35) 374 (39) 375 (36) 371 (36)
High sensitivity C-reactive protein, mg/L 4 (165) 4 (183) 4 (157) 4 (164)
Fibrosis-4 scoreg 1.1 (52.2) 1.3 (58.1) 1.6 (58.1) 1.4 (59.3)
NAFLD Fibrosis Scoreg
Mean (SD) −0.9 (2) −0.6 (2) −0.4 (1) −0.6 (2)
ELFa
Mean (SD) −0.4 (0.7) −0.1 (0.9) 0.2 (0.8) −0.0 (0.8)
Liver stiffness by FibroScan®, kPae 9.2 (51) 10.3 (45) 11.8 (64) 10.6 (57)

N (%) or geometric mean (CV) unless otherwise indicated.


ALT alanine aminotransferase, AST aspartate aminotransferase, BMI body mass index, CK-18 cytokeratin-18, CV coefficient of variation, ELF enhanced
liver fibrosis, FAS full analysis set, GGT gamma-glutamyl transferase, MCP-1, monocyte chemotactic protein 1, N number of patients, NA not available,
NAFLD non-alcoholic fatty liver disease, SD standard deviation.
a
n = 316.
b
n = 315.
c
n = 308.
d
n = 319.
e
n = 212.
f
n = 318.
g
n = 307.

3.4. Strengths and limitations the biomarker would be used in practice. For example, the accuracy for
classifying F3 versus F1–F2 fibrosis will be lower compared with clas­
The strengths of the study are its large trial population and a diverse sifying F3–F4 versus F0–F2. There may also be other less obvious
clinical profile of enrolled patients, typical for patients with NASH. sources of spectrum bias such as a non-representative prevalence of
The presented ROC analyses are subject to limitations due to the comorbidities, such as T2D. The measures of accuracy such as AUROC
trial being primarily designed to evaluate the effect of semaglutide on should not therefore be compared with studies using different inclusion
NASH rather than to qualify diagnostic biomarkers. Most importantly, criteria. However, the spectrum bias does not necessarily impact the
there is a spectrum bias present since the analyses were conducted on a internal validity, which means the comparisons of accuracy between
population that excluded patients with NAFLD or NASH at the least different biomarkers within the same trial would be valid.
severe end of the spectrum as well as those at the most severe end. The Another limitation is the potential presence of a partial verification
consequences are that both the sensitivity and the specificity would bias due to the investigator selecting patients for screening based on
likely be lower than would be the case in a broader population in which some of the parameters that were evaluated as biomarkers in this paper.

5
S.A. Harrison, et al. Contemporary Clinical Trials 97 (2020) 106174

Table 3
Accuracy of baseline characteristics and biomarkers in classifying fibrosis stage.
Biomarker Fibrosis stage Cut-point Sensitivity Specificity PPV NPV AUROC (95% CI)

Fibrosis-4 score 2–3 vs 1 1.3 64% (139/218) 70% (62/89) 84% (139/166) 44% (62/141) 0.69 (0.62;0.75)
3 vs 1–2 1.3 69% (103/150) 64% (101/157) 65% (103/159) 68% (101/148) 0.67 (0.61;0.73)
NAFLD Fibrosis Score 2–3 vs 1 −0.9 66% (144/218) 52% (46/89) 77% (144/187) 38% (46/120) 0.60 (0.53;0.67)
3 vs 1–2 −0.9 71% (106/150) 48% (76/157) 57% (106/187) 63% (76/120) 0.58 (0.51;0.64)
ALT, U/L 2–3 vs 1 54.0 53% (121/230) 60% (54/90) 77% (121/157) 33% (54/163) 0.55 (0.48;0.62)
3 vs 1–2 68.0 41% (65/158) 74% (120/162) 61% (65/107) 56% (120/213) 0.58 (0.52;0.64)
AST, U/L 2–3 vs 1 42.0 57% (132/230) 68% (61/90) 82% (132/161) 38% (61/159) 0.63 (0.56;0.70)
3 vs 1–2 37.0 73% (116/158) 52% (84/162) 60% (116/194) 67% (84/126) 0.66 (0.60;0.72)
GGT, U/L 2–3 vs 1 70.0 40% (93/230) 72% (65/90) 79% (93/118) 32% (65/202) 0.56 (0.49;0.63)
3 vs 1–2 68.0 49% (77/158) 72% (117/162) 63% (77/122) 59% (117/198) 0.62 (0.56;0.68)
ELF score 2–3 vs 1 −0.2 62% (141/226) 68% (61/90) 83% (141/170) 42% (61/146) 0.69 (0.63;0.76)
3 vs 1–2 −0.1 67% (104/155) 63% (101/161) 63% (104/164) 66% (101/152) 0.68 (0.62;0.74)
CK-18 fragments M30, U/L 2–3 vs 1 260.0 90% (206/229) 26% (23/89) 76% (206/272) 50% (23/46) 0.56 (0.48;0.63)
3 vs 1–2 260.0 94% (149/158) 23% (37/160) 55% (149/272) 80% (37/46) 0.59 (0.53;0.65)
CK-18 fragments M65, U/L 2–3 vs 1 545.0 90% (207/229) 29% (26/89) 77% (207/270) 54% (26/48) 0.58 (0.50;0.65)
3 vs 1–2 545.0 95% (150/158) 25% (40/160) 56% (150/270) 83% (40/48) 0.60 (0.54;0.66)
MCP-1, pg/mL 2–3 vs 1 497.2 21% (48/229) 87% (77/89) 80% (48/60) 30% (77/258) 0.52 (0.45;0.59)
3 vs 1–2 245.1 93% (147/158) 14% (22/160) 52% (147/285) 67% (22/33) 0.51 (0.44;0.57)
Liver stiffness by FibroScan®, kPa 2–3 vs 1 7.3 89% (135/151) 33% (20/61) 77% (135/176) 56% (20/36) 0.63 (0.54;0.71)
3 vs 1–2 11.5 56% (58/104) 71% (77/108) 65% (58/89) 63% (77/123) 0.65 (0.58;0.73)

The cut-points were determined by maximising Youden index. Sensitivity, specificity, PPV and NPV were evaluated at the specified cut-point.
ALT alanine aminotransferase, AST aspartate aminotransferase, AUROC area under the receiver operating characteristic curve, CI confidence interval, CK-18 cy­
tokeratin-18, ELF enhanced liver fibrosis, GGT gamma-glutamyl transferase, MCP-1 monocyte chemotactic protein 1, NAFLD non-alcoholic fatty liver disease, NPV
negative predictive value, PPV positive predictive value.

Potentially affected biomarkers include standard assessments such as reference standard (i.e. the histological score) is itself just a proxy of the
the liver enzymes (ALT, AST and GGT), indices that can be derived from underlying severity of the disease and is known to be imperfect. The
such assessments (Fib-4 and NAS) as well as FibroScan® where avail­ true accuracy of a biomarker that is as good as, or better than, the
able. If patients with low values on these biomarkers tended to be ex­ reference standard may consequently be underestimated.
cluded from screening (and consequently from randomisation), it
would lead to an artificial increase in sensitivity but primarily a de­ 4. Conclusions
crease in specificity. There may also be operator-related variation in
liver stiffness measurements made by transient elastometry that were In this large Phase 2 trial of semaglutide treatment for NASH, the
conducted locally by a number of different operators. A general lim­ baseline demographics of the enrolled participants appeared to be
itation common for most studies of biomarkers for NASH is that the generally consistent with patients with NASH described in other trials

8
2
NAFLD Fibrosis score
Fibrosis-4 score

6
0
4
-2
2
-4
0
1 2 3 1 2 3
Fibrosis stage Fibrosis stage

50
2
Serum ELF-derived

40
1
30
0
20
-1
10
-2 0
1 2 3 1 2 3
Fibrosis stage Fibrosis stage

Fig. 2. Biomarkers by fibrosis stage. Boxplots of Fibrosis-4 score, NAFLD Fibrosis Score, enhanced liver fibrosis score and liver stiffness by FibroScan® by fibrosis
stage.
NAFLD non-alcoholic fatty liver disease.

6
S.A. Harrison, et al. Contemporary Clinical Trials 97 (2020) 106174

[13,28,30]. Patients with F3 fibrosis tended to be older and have higher As part of this trial, patients were stratified by presence of T2D.
ALT and AST levels, compared with patients with F1 or F2 fibrosis. Overall, accuracy of Fib-4, ELF, AST and ALT for classifying fibrosis
However, serum levels of free fatty acids, triglycerides, and fasting stage appeared to be fractionally higher for patients with T2D than
plasma glucose did not differ substantially between the fibrosis stages. those without T2D. Circulating MCP-1 levels, CK-18 fragments M30 and
Similarly, BMI did not appear to be related to fibrosis stage. More pa­ M65, BMI, HbA1c and fasting plasma glucose showed poor accuracy in
tients with NAS 4–5 had a BMI ≥ 40 kg/m2, were aged ≥ 65–74 years classifying fibrosis stage in patients with or without T2D. Overall, the
and were of Hispanic/Latino ethnicity than patients with NAS 6–8. diagnostic accuracy of baseline characteristics and biomarkers was not
Overall, there was a higher percentage of patients with T2D with NAS conclusive for the full population, patients with T2D or patients without
4–6 than NAS 7–8. AST and ALT levels appeared to be higher in patients T2D.
with NAS 7–8. Similar to the fibrosis stages, serum levels of free fatty In conclusion, the clinical profile of enrolled patients in the
acids, triglycerides, and fasting plasma glucose did not differ sub­ NCT02970942 study was typical for patients with NASH. Fib-4 and ELF
stantially between NAS. appeared to be better at classifying fibrosis stage than other biomarkers
Additionally, we performed correlation analyses to determine if in the trial. To our knowledge, these data represent one of the largest
there were any clinical or biochemical data that could identify histo­ analyses of baseline characteristics and biomarkers in patients with
logical disease severity among patients with NASH. Both invasive and NASH and highlight that there is a need for further investigation of non-
non-invasive methods are currently used to detect and monitor NAFLD invasive biomarkers for the diagnosis and monitoring of NASH.
and NASH. Non-invasive imaging techniques have improved over time
and may be used to determine the progression of steatosis, fibrosis and Funding
liver stiffness; however, liver biopsies are still considered to be the gold
standard for confirmatory diagnosis of NASH [1,2]. There is a need for This trial was supported by Novo Nordisk.
less invasive methodologies that can provide an accurate and reliable
diagnosis of NASH. Authorship
FibroScan® is a validated non-invasive method for the assessment of
liver fibrosis and has been used for screening and detection of com­ SAH, SC, KC, ML, TO, VR, AS and PNN were involved in the study
plications in patients with NAFLD [3,31]. In this trial, FibroScan® did design. SAH, KC, TO, VR, AS and PNN collected the data. ML performed
not appear to have substantial predictive value for screening for ad­ the statistical data analysis. All authors participated in interpretation of
vanced fibrosis stage. However, liver fibrosis assessments using Fi­ the data and drafting and revision of the manuscript. All authors re­
broScan® have been shown in some literature to significantly correlate viewed and approved the final submitted version.
with Fib-4 values, APRI scores, and AST/ALT ratios for patients with PNN was supported by the National Institute of Health Research
NAFLD [19,31]. (NIHR) Birmingham Biomedical Research Centre (BRC). The views
In the present study, Fib-4 and ELF appeared to be better at clas­ expressed are those of the authors and not necessarily those of the NHS,
sifying fibrosis stage in patients with NASH than other biomarkers. the NIHR or the Department of Health.
However, the performance/results were modest and further investiga­
tion of these biomarkers is warranted. The findings are in line with Medical writing, editorial, and other assistance
previous studies in which Fib-4 has been found to have more promising
clinical utility for prediction of fibrosis against some other non-invasive Editorial assistance in the preparation of this article was provided
scoring systems, including NFS, in studies of patients with NAFLD by Anna Bacon of Articulate Science. Support for this assistance was
[19,32–34]. In one study, the AUROC for Fib-4 for identifying patients funded by Novo Nordisk.
with F3–F4 fibrosis was 0.8, which was similar to the NFS (0.77) but
higher than the other scoring systems it was compared against (all < Disclosures
0.75) in a cohort of 541 patients with NAFLD from the NASH CRN
network [34]. A Fib-4 score of ≥ 2.67 has been shown to have an 80% SAH reports research grants from Gilead, Intercept, Genfit, Cirius,
PPV and a Fib-4 score of ≤ 1.30 had a 90% NPV for advanced fibrosis NGM Biopharmaceuticals, Novo Nordisk, Novartis, Galmed, Immuron,
[34]. For ELF, Guha et al. 2008 reported an AUROC of 0.90 for dis­ Galectin, Madrigal, Conatus, Pfizer, Tobira/Allergan and CymaBay.
tinguishing advanced fibrosis (≥ F3; sensitivity 80%, specificity 90% SAH has served as an advisor or consultant for Echosens, Allergan,
with a threshold of 0.4) in a study of 196 patients with NAFLD [18]. Metacrine, Perspectum, Prometheus, Galmed, CiVi Biopharma, Corcept,
The ELF score has been endorsed by the National Institute of Health and Madrigal, Pfizer, NGM Biopharmaceuticals, Bristol-Myers Squibb,
Care Excellence (United Kingdom) in their guidelines for management Gilead, Intercept, Histoindex, Cirius, Axcella, Genfit, Novo Nordisk,
of NAFLD as a promising predictor of advanced fibrosis stage [35]. It Novartis, Pharmaceutical Product Development, Medpace, IQVIA,
should be noted that the results from the present study cannot be di­ CymaBay, Chronic Liver Disease Foundation, Innovate, Albireo,
rectly compared with those obtained from studies due to the different Hightide, Terns, ConSynance, Galectin, Second Genome and Akero, and
eligibility criteria for the different studies. he has served as a speaker or a member of a speaker's bureau for AbbVie
In this study, AST and ALT showed some correlation with fibrosis and Alexion. KC reports grants and non-financial support for research
stage. Results from the literature are mixed, with some studies finding support as a Principal Investigator (PI) from Cirius, Inventiva, Janssen,
typically higher ALT levels in the presence of NASH or advanced fi­ Novartis, and Zydus; grants, personal fees and non-financial support for
brosis, but others finding no such relationship [36–39]. Moreover, up to research support as a PI/consultant from Novo Nordisk; and personal
80% of patients with NAFLD [40] and 30–60% of patients with biopsy- fees for consulting from Bristol-Myers Squibb, Deuterex, Eli Lilly and
confirmed NASH have normal ALT levels [41]. Overall, ALT and/or AST Company, Janssen Research and Development LLC, Poxel, and Amgen.
may not have high diagnostic capability on their own, but could con­ VR reports consultancy from Allergan, Genfit, Intercept, Novartis,
tribute to the accurate prediction of NASH or fibrosis stage with other Boehringer Ingelheim, Novo Nordisk, Galmed and Pfizer; he reports
biomarkers. Recently, combination of type IV collagen 7S and AST have grants from Gilead and Intercept. AS reports grants and consultancy
shown a promising ability to predict both NASH and NASH-related fi­ from Bristol-Myers Squibb, Gilead, Intercept, and Novartis; grants from
brosis in patients with NAFLD [42]. ALT and/or AST are currently used Echosens, Merck, and Tobira; consultancy from Galectin, Nitto Denko,
in different diagnostic panels, such as the FibroTest [43], NashTest Nimbus, Ardelyx, Vivelix, Teva Pharmaceutical Industries Ltd., Can-
[44], NFS [45], FibroMeter™ [46], Fib-4 index [47] and other different Fite, Boehringer Ingelheim, Pfizer, Salix, and Enyo; royalties from
models [48,49]. UpToDate; stocks in Akarna, Durect, Exalenz, HemoShear and Natural

7
S.A. Harrison, et al. Contemporary Clinical Trials 97 (2020) 106174

Shield; board membership from Sanyal Bio; and board membership and J. Gastrointest. Pathophysiol. 8 (2017) 51–58.
stocks from Genfit and Tiziana. PN reports grants, consultancy and non- [9] G. Sebastiani, R. Alshaalan, P. Wong, et al., Prognostic value of non-invasive fibrosis
and steatosis tools, hepatic venous pressure gradient (HVPG) and histology in
financial support from Novo Nordisk; grants and consultancy from nonalcoholic steatohepatitis, PLoS One 10 (2015) e0128774.
Boehringer Ingelheim; consultancy from Afimmune, Gilead Sciences, [10] M. Tesfay, W.J. Goldkamp, B.A. Neuschwander-Tetri, NASH: the emerging most
Intercept Pharmaceuticals, Pfizer and Shire; and speaker fees from common form of chronic liver disease, Mo. Med. 115 (2018) 225–229.
[11] S. Polanco-Briceno, D. Glass, M. Stuntz, A. Caze, Awareness of nonalcoholic stea­
Norgine. SC, ML and A-SS are employees of Novo Nordisk A/S. TO tohepatitis and associated practice patterns of primary care physicians and spe­
declares no competing interests. cialists, BMC Res. Notes 9 (2016) 157.
[12] M.A. Konerman, J.C. Jones, S.A. Harrison, et al., Pharmacotherapy for NASH:
current and emerging, J. Hepatol. 68 (2018) 362–375.
Compliance with ethics guidelines [13] S.L. Friedman, V. Ratziu, S.A. Harrison, et al., A randomized, placebo-controlled
trial of cenicriviroc for treatment of nonalcoholic steatohepatitis with fibrosis,
An independent ethics committee or institutional review board at Hepatology. 67 (2018) 1754–1767.
[14] N.P. Chalasani, A.J. Sanyal, K.V. Kowdley, et al., Pioglitazone versus vitamin E
each site approved the trial protocol and any subsequent amendments.
versus placebo for the treatment of non-diabetic patients with non-alcoholic stea­
Informed consent was obtained from every participant before any trial- tohepatitis: PIVENS trial design, Contemp. Clin. Trials 30 (2009) 88–96.
related activities took place, including activities to determine suitability [15] V. Wong, L. Adams, V. de Lédinghen, G. Wong, S. Sookoian, Noninvasive bio­
for the trial. markers in NAFLD and NASH - current progress and future promise, Nat. Rev.
Gastroenterol. Hepatol. 15 (2018) 461–478.
[16] M.S. Siddiqui, S.A. Harrison, M.F. Abdelmalek, et al., Case definitions for inclusion
Data availability and analysis of endpoints in clinical trials for nonalcoholic steatohepatitis through
the lens of regulatory science, Hepatology. 67 (2018) 2001–2012.
[17] K. Staufer, E. Halilbasic, W. Spindelboeck, et al., Evaluation and comparison of six
Deidentified participant data will be made available for this Article noninvasive tests for prediction of significant or advanced fibrosis in nonalcoholic
on a specialised SAS data platform. Datasets from Novo Nordisk will be fatty liver disease, United European Gastroenterol J 7 (2019) 1113–1123.
available permanently after completion of the study and completion of [18] I.N. Guha, J. Parkes, P. Roderick, et al., Noninvasive markers of fibrosis in non­
alcoholic fatty liver disease: validating the European liver fibrosis panel and ex­
data analysis. Access to data can be made through a request proposal ploring simple markers, Hepatology. 47 (2008) 455–460.
form and the access criteria can be found online (novonordisk-trials. [19] S. McPherson, S.F. Stewart, E. Henderson, A.D. Burt, C.P. Day, Simple non-invasive
com). Data will be shared with bona fide researchers submitting a re­ fibrosis scoring systems can reliably exclude advanced fibrosis in patients with non-
alcoholic fatty liver disease, Gut. 59 (2010) 1265–1269.
search proposal requesting access to data. Data use is subject to ap­ [20] J.J. Connolly, K. Ooka, J.K. Lim, Future pharmacotherapy for non-alcoholic stea­
proval by the Independent Review Board. tohepatitis (NASH): review of phase 2 and 3 trials, J. Clin. Transl. Hepatol. 6 (2018)
264–275.
[21] Y. Sumida, M. Yoneda, Current and future pharmacological therapies for NAFLD/
Thanking patient participants
NASH, J. Gastroenterol. 53 (2018) 362–376.
[22] Y.S. Lee, H.S. Jun, Anti-inflammatory effects of GLP-1-based therapies beyond
We gratefully acknowledge the trial participants and all personnel glucose control, Mediat. Inflamm. 2016 (2016) 3094642.
involved in the trial. [23] F. Sun, S. Wu, J. Wang, et al., Effect of glucagon-like peptide-1 receptor agonists on
lipid profiles among type 2 diabetes: a systematic review and network meta-ana­
lysis, Clin. Ther. 37 (2015) 225–41.e8.
Patient involvement [24] M. Katout, H. Zhu, J. Rutsky, et al., Effect of GLP-1 mimetics on blood pressure and
relationship to weight loss and glycemia lowering: results of a systematic meta-
analysis and meta-regression, Am. J. Hypertens. 27 (2014) 130–139.
No patients were involved in the study trial design or dissemination [25] M.J. Armstrong, P. Gaunt, G.P. Aithal, et al., Liraglutide safety and efficacy in pa­
of results. tients with non-alcoholic steatohepatitis (LEAN): a multicentre, double-blind, ran­
domised, placebo-controlled phase 2 study, Lancet. 387 (2016) 679–690.
[26] P. Newsome, S. Francque, S. Harrison, et al., Effect of semaglutide on liver enzymes
Prior presentation and markers of inflammation in subjects with type 2 diabetes and/or obesity,
Aliment. Pharmacol. Ther. 50 (2019) 193–203.
No part of the manuscript has been previously shared nor is based [27] Alan Agresti, Categorical Data Analysis, 3rd edn, John Wiley & Sons, Inc., Hoboken,
2013.
on work that has been previously presented.
[28] V. Ratziu, S.A. Harrison, S. Francque, et al., Elafibranor, an agonist of the peroxi­
some proliferator-activated receptor-alpha and -delta, induces resolution of non­
Appendix A. Supplementary data alcoholic steatohepatitis without fibrosis worsening, Gastroenterology. 150 (2016)
1147–59.e5.
[29] M.G. Sanal, Biomarkers in nonalcoholic fatty liver disease-the emperor has no
Supplementary data to this article can be found online at https:// clothes? World J. Gastroenterol. 21 (2015) 3223–3231.
doi.org/10.1016/j.cct.2020.106174. [30] S. Dasarathy, J. Dasarathy, A. Khiyami, et al., Double-blind randomized placebo-
controlled clinical trial of omega 3 fatty acids for the treatment of diabetic patients
with nonalcoholic steatohepatitis, J. Clin. Gastroenterol. 49 (2015) 137–144.
References [31] H. Fallatah, H.O. Akbar, A.M. Fallatah, Fibroscan compared to FIB-4, APRI, and
AST/ALT ratio for assessment of liver fibrosis in Saudi patients with nonalcoholic
[1] European Association for the Study of the Liver, European Association for the Study fatty liver disease, Hepat. Mon. 16 (2016) e38346.
of Diabetes, European Association for the Study of Obesity. EASL-EASD-EASO [32] W. Sun, H. Cui, N. Li, et al., Comparison of FIB-4 index, NAFLD fibrosis score and
clinical practice guidelines for the management of non-alcoholic fatty liver disease, BARD score for prediction of advanced fibrosis in adult patients with non-alcoholic
J. Hepatol. 64 (2016) 1388–1402. fatty liver disease: a meta-analysis study, Hepatol. Res. 46 (2016) 862–870.
[2] N. Chalasani, Z. Younossi, J.E. Lavine, et al., The diagnosis and management of [33] L.A. Adams, J. George, E. Bugianesi, et al., Complex non-invasive fibrosis models
nonalcoholic fatty liver disease: practice guidance from the American Association are more accurate than simple models in non-alcoholic fatty liver disease, J.
for the Study of Liver Diseases, Hepatology. 67 (2018) 328–357. Gastroenterol. Hepatol. 26 (2011) 1536–1543.
[3] J.K. Dyson, Q.M. Anstee, S. McPherson, Non-alcoholic fatty liver disease: a practical [34] A.G. Shah, A. Lydecker, K. Murray, et al., Comparison of noninvasive markers of
approach to diagnosis and staging, Frontl. Gastroenterol. 5 (2014) 211–218. fibrosis in patients with nonalcoholic fatty liver disease, Clin. Gastroenterol.
[4] T. Kennedy-Martin, J.P. Bae, R. Paczkowski, E. Freeman, Health-related quality of Hepatol. 7 (2009) 1104–1112.
life burden of nonalcoholic steatohepatitis: a robust pragmatic literature review, J. [35] National Institute for Health and Care Excellence (NICE), Non-alcoholic Fatty Liver
Patient Rep. Outcomes 2 (2018) 28. Disease. Assessment and Management, https://www.nice.org.uk/guidance/ng49/
[5] D. Haldar, B. Kern, J. Hodson, et al., Outcomes of liver transplantation for non- evidence/full-guideline-pdf-2548213310, (2016) (Accessed October 4, 2019).
alcoholic steatohepatitis: a European liver transplant registry study, J. Hepatol. 71 [36] A.L. Fracanzani, L. Valenti, E. Bugianesi, et al., Risk of severe liver disease in
(2019) 313–322. nonalcoholic fatty liver disease with normal aminotransferase levels: a role for in­
[6] R.J. Wong, M. Aguilar, R. Cheung, et al., Nonalcoholic steatohepatitis is the second sulin resistance and diabetes, Hepatology. 48 (2008) 792–798.
leading etiology of liver disease among adults awaiting liver transplantation in the [37] P. Mofrad, M.J. Contos, M. Haque, et al., Clinical and histologic spectrum of non­
United States, Gastroenterology. 148 (2015) 547–555. alcoholic fatty liver disease associated with normal ALT values, Hepatology. 37
[7] M. Noureddin, A. Vipani, C. Bresee, et al., NASH leading cause of liver transplant in (2003) 1286–1292.
women: updated analysis of indications for liver transplant and ethnic and gender [38] N. Hossain, A. Afendy, M. Stepanova, et al., Independent predictors of fibrosis in
variances, Am. J. Gastroenterol. 113 (2018) 1649–1659. patients with nonalcoholic fatty liver disease, Clin. Gastroenterol. Hepatol. 7 (2009)
[8] R. Patil, G.K. Sood, Non-alcoholic fatty liver disease and cardiovascular risk, World 1224–1229.

8
S.A. Harrison, et al. Contemporary Clinical Trials 97 (2020) 106174

[39] S. Verma, D. Jensen, J. Hart, S.R. Mohanty, Predictive value of ALT levels for non- (NashTest) for the prediction of non alcoholo steato hepatitis in patients with non-
alcoholic steatohepatitis (NASH) and advanced fibrosis in non-alcoholic fatty liver alcoholic fatty liver disease, BMC Gastroenterol. 6 (2006) 34.
disease (NAFLD), Liver Int. 33 (2013) 1398–1405. [45] P. Angulo, J.M. Hui, G. Marchesini, et al., The NAFLD fibrosis score: a noninvasive
[40] A.E. Junker, The role of incretin hormones and glucagon in patients with liver system that identifies liver fibrosis in patients with NAFLD, Hepatology. 45 (2007)
disease, Dan. Med. J. 64 (2017) (pii: B5363). 846–854.
[41] M.E. Rinella, Nonalcoholic fatty liver disease: a systematic review, JAMA. 313 [46] P. Calès, F. Lainé, J. Boursier, et al., Comparison of blood tests for liver fibrosis
(2015) 2263–2273. specific or not to NAFLD, J. Hepatol. 50 (2009) 165–173.
[42] T. Okanoue, H. Ebise, T. Kai, et al., A simple scoring system using type IV collagen [47] Y. Sumida, M. Yoneda, H. Hyogo, et al., Validation of the FIB4 index in a Japanese
7S and aspartate aminotransferase for diagnosing nonalcoholic steatohepatitis and 73on-alcoholic fatty liver disease population, BMC Gastroenterol. 12 (2012) 2.
related fibrosis, J. Gastroenterol. 53 (2018) 129–139. [48] N.A. Palekar, R. Naus, S.P. Larson, J. Ward, S.A. Harrison, Clinical model for dis­
[43] V. Ratziu, P. Giral, M. Munteanu, et al., Screening for liver disease using non-in­ tinguishing nonalcoholic steatohepatitis from simple steatosis in patients with
vasive biomarkers (FibroTest, SteatoTest and NashTest) in patients with hyperli­ nonalcoholic fatty liver disease, Liver Int. 26 (2006) 151–156.
pidaemia, Aliment. Pharmacol. Ther. 25 (2007) 207–218. [49] Z.M. Younossi, S. Page, N. Rafiq, et al., A biomarker panel for non-alcoholic stea­
[44] T. Poynard, V. Ratziu, F. Charlotte, et al., Diagnostic value of biochemical markers tohepatitis (NASH) and NASH-related fibrosis, Obes. Surg. 21 (2011) 431–439.

You might also like