You are on page 1of 15

Expert Review of Gastroenterology & Hepatology

ISSN: 1747-4124 (Print) 1747-4132 (Online) Journal homepage: https://www.tandfonline.com/loi/ierh20

Non-invasive assessment of liver fibrosis and


prognosis: an update on serum and elastography
markers

Uchenna Agbim & Sumeet K. Asrani

To cite this article: Uchenna Agbim & Sumeet K. Asrani (2019): Non-invasive assessment of
liver fibrosis and prognosis: an update on serum and elastography markers, Expert Review of
Gastroenterology & Hepatology, DOI: 10.1080/17474124.2019.1579641

To link to this article: https://doi.org/10.1080/17474124.2019.1579641

Accepted author version posted online: 06


Feb 2019.
Published online: 20 Feb 2019.

Submit your article to this journal

Article views: 2

View Crossmark data

Full Terms & Conditions of access and use can be found at


https://www.tandfonline.com/action/journalInformation?journalCode=ierh20
EXPERT REVIEW OF GASTROENTEROLOGY & HEPATOLOGY
https://doi.org/10.1080/17474124.2019.1579641

REVIEW

Non-invasive assessment of liver fibrosis and prognosis: an update on serum and


elastography markers
Uchenna Agbima and Sumeet K. Asranib
a
Division of Transplant Surgery, Department of Surgery, University of Tennessee Health Science Center, Memphis, TN, USA; bBaylor University
Medical Center, Dallas, TX, USA

ABSTRACT ARTICLE HISTORY


Introduction: Non-invasive assessment of fibrosis is increasingly utilized in clinical practice to diagnose Received 10 August 2018
hepatic fibrosis. Non-invasive assessment of liver fibrosis relies on biologic and/or physical properties to Accepted 4 February 2019
assess tissue fibrosis. Serum markers estimate fibrosis by incorporating markers reflecting hepatic KEYWORDS
function (indirect markers) and/or markers measuring extracellular matrix degradation/fibrogenesis Elastography; hepatitis C;
(direct markers). Radiology based techniques relay the mechanical properties and stiffness of a tissue, hepatitis B; liver fibrosis;
with increased stiffness associated with more advanced fibrosis. liver stiffness; magnetic
Areas covered: In this comprehensive review, the recent literature discussing serum markers and resonance elastography;
elastography-based techniques will be covered. These modalities are also explored in the setting of nonalcoholic fatty liver
various liver diseases. disease; shear wave
Expert opinion: The etiology of liver disease and clinical context should be taken into consideration elastography; vibration-
when non-invasive markers are incorporated in clinical practice. Non-invasive assessment of fibrosis has controlled transient
elastography
been most extensively utilized in hepatitis C, followed by hepatitis B and nonalcoholic fatty liver
disease, but its role remains less developed in other etiologies of liver disease such as alcohol-
associated liver disease and autoimmune liver disease. The role of non-invasive markers in predicting
progression or regression of fibrosis, development of liver-related events and survival needs to be
further explored.

1. Introduction specimen size, sampling error, inter- and intra-observer reliability


Progressive fibrosis of the liver can lead to cirrhosis and compli- with histologic assessment [2,3], and decreased level of perfor-
cations of portal hypertension including ascites, variceal hemor- mance for intermediate stages of fibrosis [4]. Thus, there exists
rhage, portosystemic encephalopathy, as well as hepatocellular a need for a less-invasive, reproducible, and accurate method to
carcinoma (HCC). Evaluation or staging of fibrosis is critical to characterize fibrosis in subjects with chronic liver disease.
monitor the progression of chronic liver disease, determine
prognosis, establish optimal timing for treatment, monitor 2.1. Overview of non-invasive assessment
response to treatment, and assess the evolution of disease to
mitigate morbidity and mortality associated with the sequelae of Non-invasive assessment of liver fibrosis relies on biologic and/or
cirrhosis [1]. Long heralded as the gold standard for assessing physical properties to assess tissue fibrosis. Specifically, serum
fibrosis, liver biopsy is fraught with many limitations [2]. markers represent biologic properties [5], while the most com-
Over the last two decades, there has been significant interest mon clinically applied radiologic markers utilize physical proper-
in the development of non-invasive surrogates of liver fibrosis. In ties, that is principles of elastography to indirectly determine
contrast to liver biopsy, a majority of these tests are simple to tissue stiffness [5,6].
perform, are more accessible and repeatable, rendering them
easier to use at a population level to evaluate fibrosis. The 2.2. Serum tests
purpose of this review is to summarize the basic principles of
non-invasive assessment of liver fibrosis and inform the reader In recent years, serum tests have gained widespread use, as they
on updates in this field over the last few years. are readily available, simple to utilize, objective, and cost less as
compared to a liver biopsy. A majority of these tests were devel-
oped in viral hepatitis or nonalcoholic fatty liver disease (NAFLD)
2. Assessment of liver fibrosis but have been adapted for use in various other liver diseases.
Liver biopsy is the putative gold standard for diagnosing fibrosis, A unifying theme among most serum markers is they have
but carries notable risk including the potential complications of reasonable negative predictive value (NPV), rendering the mar-
pain, bleeding, infection, perforation, and even death [2]. Further, kers useful tests at excluding disease, but fair positive predictive
the quality of liver biopsy may often be limited by small value (PPV) with many false positives. In general, these tests

CONTACT Sumeet K. Asrani Sumeet.Asrani@baylorhealth.edu Baylor University Medical Center, Dallas, TX, USA
© 2019 Informa UK Limited, trading as Taylor & Francis Group
2 U. AGBIM AND S. K. ASRANI

estimate fibrosis by incorporating markers reflecting hepatic The FIB-4 test was originally developed to estimate fibrosis
injury and/or complications of portal hypertension (indirect mar- in subjects co-infected with human immunodeficiency virus
kers) and/or markers measuring extracellular matrix (ECM) degra- and HCV with two cut-off scores similar to NFS. For the low
dation/fibrogenesis (direct markers) [7]. Some tests are cut-off of <1.45, the AUROC value to detect Ishak stage 4–6
proprietary and available as a send-out test. Others can be (equivalent to Metavir F3–4) was 0.765, the NPV was 90%, and
calculated using routine blood work in combination with patient sensitivity was 70%. For the high cut-off value of >3.25, the
characteristics. A few examples of serum tests include the NAFLD PPV was 65% and specificity was 97% [17]. The pitfall with the
Fibrosis Score (NFS), BARD score, FIB-4, aspartate aminotransfer- FIB-4 test lies with scores 1.45–3.25, where a test with better
ase (AST) to platelet ratio index (APRI), and Enhanced Liver discrimination is needed.
Fibrosis Score (ELF). The NFS and BARD score are utilized speci- Another serum test is the AST to platelet ratio (APRI) score,
fically for NAFLD, while the remaining tests were designed for which was also developed in HCV subjects to predict both F ≥
hepatitis C virus (HCV) but can assess any chronic liver disease. 3 and cirrhosis. This score has two cut-off values each for F ≥ 3
These, among other tests, are briefly described in Table 1. and cirrhosis and can identify significant fibrosis in 51% of
Using six common clinical parameters (age, hyperglycemia, subjects and even more accurately identify cirrhosis in 81% of
BMI, platelet count, albumin, AST/alanine aspartate [ALT] subjects [18]. Additionally, the use of two cut-off values in the
ratio), the NFS was developed to detect fibrosis in subjects APRI, FIB-4, and NFS results in an intermediate range where
with NAFLD with two cut-off scores. The low cut-off score of further testing, including a biopsy, may be needed [19]. Hence,
<-1.455 can identify stage 3–4 fibrosis (F ≥ 3) using Brunt- studies have used a wide range of cut-off values for significant
Kleiner staging system with an NPV of 93% and the high cut- fibrosis which limits its practical use [20].
off of >0.696 has a PPV of 90% for detecting F ≥ 3 [15]. Many of the direct markers can be used singularly or incorpo-
In addition to the NFS, another scoring system developed rated into panels to assess fibrosis. These direct markers include
to stage fibrosis in subjects with NAFLD is the BARD score, amino-terminal propeptide of type III collagen (Pro-C3), hyaluronic
which combines BMI, AST/ALT ratio, and presence of diabetes acid, and tissue inhibitor of metalloproteinase-1 (TIMP-1). Pro-C3
to produce a weighted score assessing fibrosis risk. A BARD measures collagen formation [21] while hyaluronic acid,
score ≥2 indicates advanced fibrosis with an area under the a glycosaminoglycan, is highly prevalent in the liver ECM [22].
receiver operating characteristic (AUROC) value of 0.81 and TIMP-1 is a pivotal regulator in the synthesis and degradation of
NPV of 96% [16]. the ECM, with its level increasing in parallel with severity of liver
fibrosis [23]. Wisteria floribunda agglutinin-positive Mac-2-binding
protein (WFA(+)-M2BP) is a novel marker involved in ECM adhe-
Table 1. Selected serum tests for assessment of fibrosis. sion and has shown promising results as a surrogate for fibrosis in
Non-invasive Serum Test Formula/Components many chronic liver diseases [24,25]. ELF is a proprietary test com-
APRI[18] AST/ULN of laboratory AST bining age with direct markers of fibrosis including hyaluronic
Platelets (109/L) X 100
BARD[16] Weighted Sum of: BMI ≥ 28 = 1 point, AST/ALT ≥ acid, Pro-C3, and TIMP-1 [26]. It has three cut-offs scores, but
0.8 = 2 points, Presence of DM = 1 point there are fluctuations in the scores by age and gender raising
FIB-4[17] Age ([yr] X AST [U/L]) questions about the accuracy of the test itself [27]. Other commer-
((Platelets [109/L]) X (ALT [U/L]))1/2
NFS[15] −1.675 + 0.037 X age (yr) +0.094 X BMI (kg/m2) cially available tests incorporating direct markers are characterized
+ 1.13 X IFG/DM in Table 1.
(yes = 1, no = 0) + 0.99 X AST/ALT ratio –
0.013 X platelet (109/L) -
0.66 X albumin (g/dl) 2.3. Radiology tests
Forns Index[8] 7.811–3.131 X ln(platelet[109/L])
+ 0.781 X ln(GGT[IU/L]) Over the past two decades, tissue elastography has been used to
+ 3.467 X ln(age[yr]) – 0.014.(cholesterol[mg/
dL]) diagnose fibrosis, assess response to treatment, and guide man-
GPR[9] GGT agement of patients with chronic liver disease. Elastography
Platelets reflects the mechanical properties and stiffness of a tissue, with
Lok Index[10] −5.56–0.0089 X platelet (103/mm3) + 1.26 X AST/
ALT ratio + 5.27 X INR increased stiffness correlating with more advanced fibrosis [28].
ELF[26] Age, hyaluronic acid, amino-terminal propeptide The current modalities are classified into ultrasound-based elas-
of type III collagen, and tissue inhibitor of tography or magnetic resonance imaging (MRI)-based elastogra-
metalloproteinase 1
FibroTest-ActiTest[11] Age, gender, α2-macroglobulin, haptoglobin, phy (MRE). US-based elastography consists of strain-based or
(also referred to as GGT, ALT, apolipoprotein A1 shear-wave based elastography, with the vast majority of com-
FibroSure in the US) monly used techniques using the latter. Strain elastography
FibroMeter[12] Age, platelets, PT, AST, α2-macroglobulin,
hyaluronic acid, urea utilizes mechanical force (manual compression of tissue) and
FibroMeter NAFLD[13] Age, body weight, platelets, AST, ALT, glucose, has limited applications in assessing liver fibrosis [6,28,29]. Shear-
ferritin wave elastography evaluates stiffness by assessing the velocity of
Hepascore[14] Age, gender, bilirubin, GGT, hyaluronic acid,,
α2-macroglobulin shear waves through tissue; shear waves travel more quickly
ALT: alanine aminotransferase; APRI: aspartate aminotransferase to platelet ratio through stiff or more fibrotic tissue and slower in soft tissue
index; AST: aspartate aminotransferase; BMI: body mass index; DM: diabetes [28]. Shear waves are generated either through mechanical vibra-
mellitus; ELF: Enhanced Liver Fibrosis; GGT: gamma-glutamyl transpeptidase; tion or via acoustic radiation force impulses (ARFI) [28]. MRE
GPR: gamma-glutamyl transpeptidase to platelet ratio; IFG: impaired fasting
glucose; NAFLD: nonalcoholic fatty liver disease; NFS: nonalcoholic fatty liver utilizes mechanical vibration to produce waves in the liver,
disease fibrosis score; PT: prothrombin time; ULN: upper limit of normal. which subsequently undergo processing using modified phase
EXPERT REVIEW OF GASTROENTEROLOGY & HEPATOLOGY 3

contrast MRI and are converted to tissue stiffness maps using an kPa and ≥11–14 kPa, respectively, although this depends on the
inversion algorithm [28]. Examples of the most common elasto- disease process [7]. Probes are selected by the operator based on
graphy modalities include vibration controlled transient elasto- the patient’s habitus; the standard 3.5 MHz M-probe can measure
graphy (VCTE), point-shear wave elastography (pSWE), 2D shear depths between 25 and 65 mm [28,31], while the 2.5 MHz XL-
wave elastography (2D-SWE), and MRE. probe measures tissue depths between 35 and 75 mm [28]. The
In all methods of elastography, subjects should be in the smaller 5 MHz S1 and S2 probes can measure tissue depths
fasting state [7,28]. Directly comparing values across the tech- between 15–40 mm and 20–50 mm, respectively [28,32]. The
niques is not valid as the shear waves generated by each software determines when measurements are successful and
technique vary in frequency, thereby influencing shear-wave does not record unsuccessful measurements. Successful read-
speed. Furthermore, techniques also differ in wave duration, ings have at least 10 valid measurements, a success rate (ratio
as waves are transient in ultrasound-based elastography ver- of valid measurements/total measurements) ≥60%, and have an
sus continuous in MRE [28]. A comparison of the different interquartile range (value assesses variability of measurements)
elastography techniques is summarized in Table 2. of ≤30% of the median value [5,29,31]. Limitations of VCTE
include low reliability in subjects of large body habitus, ascites,
2.3.1. VCTE subjects with severe hepatic inflammation, and hepatic conges-
VCTE, often known as its proprietary name, FibroScan (Echosens) tion, with failure rates ranging from 6% to 23%. However, VCTE’s
is the most widely used modality to measure elastography utility lies in the ability to serve as a quick, point-of-care test such
[7,28,29]. It is a one-dimensional approach and does not evaluate as in office settings, its relatively inexpensive cost, and portable
morphological features of the liver [7,28]. The transducer probe is nature [7,28,29].
placed between the intercostal space over the right lobe of the
liver, delivering short, vibrations, which, in turn, indirectly gen- 2.3.2. Point shear wave elastography
erate a shear wave propagating through the liver [28,29]. The In contrast to VCTE which generates shear waves utilizing low-
shear waves cause tissue displacement, which is tracked and frequency mechanical vibrations, pSWE (Virtual Touch
measured by the ultrasound transducer probe. Since VCTE does Quantification or Acuson S2000 and S3000 systems-Siemens
not display a real-time anatomic or grayscale image, it cannot Healthcare; ElastPQ-Philips Healthcare; Shear Wave
show the exact location where stiffness is measured [7,28,29]. Elastography-GE Healthcare) generates shear waves with high-
Data are displayed in A mode, M mode, and as a shear-wave frequency ultrasound ‘push pulses’ or ARFI in a region of interest
propagation graph on the machine [28]. Results are expressed as (ROI) chosen by the operator. Additionally, unlike VCTE, this ROI
Young’s modulus (E) and recorded in kilopascals (kPa), ranging can be saved and used at a subsequent time-point for monitor-
from 2.5 to 75 kPa. A normal value is approximately 5 kPa [5] and ing. After the ultrasound probe is positioned in an appropriate
those indicating stage 2–4 fibrosis (≥F2) and cirrhosis (F4) are >7 ROI measuring 10 × 6 mm in the right lobe, acoustic ‘push pulses’

Table 2. Comparison of biopsy and elastography techniques.


Biopsy VCTE pSWE SWE MRE
Example Reference FibroScan Siemens S2000, Virtual Aixplorer, Supersonic Magnetic Resonance
Standard Touch Quantification, Shear Imaging Elastography
ElastPQ
Wave Generation N/A Mechanical ARFI Multiple points using
vibration at 50 Hz ARFI in real time
Mechanical
vibration
at 60 Hz
Dimension N/A 1D 2D 2D 2D or 3D
Associated N/A No associated anatomic image Anatomic imaging via Anatomic imaging via Anatomic imaging via
Imaging ultrasound ultrasound and magnetic resonance
elastograms imaging and
superimposed
elastograms
Limitations Invasive; Technical failure in subjects with ascites, Not as well validated Not as well validated Technical failures in iron
Variability increased abdominal girth, narrow compared to VCTE; compared to VCTE; overload;
with inter- intercostal spaces, congestion, cholestasis, Difficulty controlling Difficulty controlling Claustrophobia;
and intra- hyperemia with food consumption frequency of shear frequency of shear Cost;
observer wave; waves; Availability
reliability Availability Availability
Ease No Yes, Point-of-Care Yes, ultrasound facility Yes, ultrasound facility Advanced radiology
center
Region of 1–2 cm length 10 X 40 mm 10X 6 mm, Operator 20 X 20 mm, Operator Operator dependent
Interest/ dependent location dependent location location of any size
Size or shape
Assessment Yes, with quantification Yes, quantification with Yes, but no quantification Yes, but no quantification
of CAP
steatosis
Yes, quantification with PDFF
ARFI: acoustic radiation force impulse; CAP: controlled attenuation parameter; MRE: magnetic resonance elastography; N/A: not applicable PDFF: proton density fat
fraction; pSWE: point-shear wave elastography; SWE: shear wave elastography; VCTE: vibration controlled transient elastography.
Adapted from Hagan et al. [30].
4 U. AGBIM AND S. K. ASRANI

are transmitted through the liver creating shear waves that travel results. Additionally, the inability to use this technique in subjects
perpendicular to the ultrasound beam [5,7,28,33]. Results are with an aversion to confined spaces, availability, and cost has
expressed in meters per second at a range of 0.5–5 m/s, with largely limited MRE to research purposes though this pattern
a variable range between normal and pathologic values reported may be changing [7,28,29,31,36].
in the literature. Unlike VCTE where the vibrations are diminished Currently, MRE is the best studied MRI-based imaging.
by fat, pSWE can detect stiffness in the presence of obesity and However, other MRI-imaging technology currently under inves-
ascites since shear waves are produced inside the liver [7,28]. tigation for the detection of fibrosis includes MRI with diffusion-
However, a recent study noted decreased reliability with Virtual weighted imaging (DWI) [7], metabolomic analysis with proton
Touch Quantification with abdominal adiposity as measured by nuclear magnetic resonance spectroscopy [37], and 3D-MRE [38].
a skin-liver capsule distance of ≥30 mm [34]. An added benefit of
pSWE is that it can be coupled to conventional ultrasound-based
3. Applications of markers to various liver diseases
imaging providing further characterization of the liver parench-
and complications
yma as well as other abdominal structures, ultimately decreasing
costs [5,7,28,31]. The diagnostic accuracy of non-invasive tests is crucial for man-
agement of chronic liver disease. Since the diagnostic para-
2.3.3. 2D-SWE meters are often dependent upon the clinical context in which
In 2D-SWE (Aixplorer or Supersonic Shear Imaging-Supersonic they are employed, a growing amount of literature has evaluated
Imagine), ARFI is used at several depths to create multiple shear the utility of the various modalities in specific disease states. In
waves in multiple ROIs measuring 20 × 20 mm which are cap- the remainder of this evaluation, we review the recent literature
tured by an ultrasound imaging with a high frame rate (up to of non-invasive techniques as it pertains to specific liver diseases,
15,000 images per second) [28], allowing for real-time imaging their sequelae, and prognosis. A comparison of clinical utility and
[5–7,28,31]. This allows for the creation of quantitative maps of a summary of meta-analysis assessing diagnostic accuracy of
tissue stiffness or elastograms [6,28]. Results are expressed as selected noninvasive markers in assessment of fibrosis is shown
meters/second or kilopascals [28]. Compared to point-shear in Tables 3 and 4, respectively.
wave elastography, multiple ROIs can be used with SWE, thereby
reducing sampling variability [7,28]. Limitations include
3.1. Nonalcoholic fatty liver disease
decreased availability and a lack of studies evaluating its validity
compared to VCTE [5,7,28,31]. An emerging technology, deep Ultrasound alone has a pooled sensitivity of 84.8% and specificity
learning Radionomics of elastography (DLRE) utilizes neural net- of 93.8% for detecting ≥20–30% steatosis compared to liver
works whereby large amounts of images are analyzed to provide biopsy [44]. However, fibrosis rather than steatosis is the stron-
finer detail and may supplement 2D-SWE, thereby increasing its gest predictor of mortality [45]. Given the increasing prevalence
diagnostic performance [35]. of NAFLD (global prevalence 25%) [46], it is impractical to biopsy
everyone. Thus, the use of non-invasive tests to detect fibrosis in
2.3.4. MRI-based imaging subjects with NAFLD may play a valuable role in long-term
MRE is the most current, accurate non-invasive mode at measur- management.
ing fibrosis [6,7,28,29,31,36]. Similar to VCTE, MRE uses mechanical In a study of 452 biopsy-proven NAFLD subjects evaluating
vibrations generated in an active driver to create shear waves, fibrosis by nine serum markers of fibrosis and VCTE, FibroMeter
which are assessed via specialized software producing elasto- and VCTE were the most accurate at predicting fibrosis with
grams. More specifically the active driver, housed in an area out- AUROC values of 0.79 and 0.84 for detecting F ≥ 2, 0.82 and
side the scanner, activates a passive driver by way of a long 0.83 for detecting F ≥ 3, and 0.82, and 0.86 for detecting F4,
connecting tube. The passive driver is physically positioned over respectively. Further, both of these tests showed good discrimi-
the largest portion of the patient’s liver and transmits continuous native ability for predicting mortality from liver-related complica-
mechanical acoustic vibrations of 60 Hz [28,36] during four breath tions in the longitudinal cohort of the study [19]. Petta et al. [47]
holds. During each breath hold, a sagittal slice of the liver is found that FIB-4, NFS, and VCTE had the best diagnostic perfor-
assessed. The shear waves are tracked by motion-sensitized MRI mance at diagnosing fibrosis in a study of 761 subjects with
sequences such as gradient recalled echo and processed to create biopsy-proven NAFLD. When VCTE was combined with either
wave images and elastograms. A confidence map can be created NFS or FIB-4, there was a decreased rate of incorrectly classified
that shows areas with low reliability. ROI can be placed on the subjects compared to any of these tests alone, but the percen-
elastograms avoiding areas with low reliability as well as major tage of discordant results was high, at over 50%. A stepwise
vessels and fissures [7,28,29,31,36]. These techniques allow for approach (fibrosis assessed initially with either NFS or FIB-4,
a large area of liver to be sampled (≥250 cm3), which is more followed by VCTE) improved performance particularly in subjects
than 5,000 times that of a liver biopsy and 250 times that of VCTE with intermediate NFS and FIB-4 scores compared to the com-
[6]. Results are expressed as the magnitude of the complex shear bined approach. This suggests that a multi-staged or stepwise
modulus (µ) reported as average stiffness in kPa from 0 to 8 kPa. approach may be more useful than using a single test to diag-
An added benefit of MRE is the ability to define lesions such as nose fibrosis non-invasively [47].
HCC with contrast-enhanced MRI, which is often combined with When comparing the available ultrasound-based imaging
the test. Limitations of MRI-based techniques include decreased tests (VCTE, pSWE, and 2D-SWE), all performed well in diagnosing
reliability in iron-overload states, passive congestion, and inflam- fibrosis, but 2D-SWE was superior to both (AUROC 0.86–0.89 for
mation as these can increase liver stiffness, thereby confounding 2D-SWE, 0.82–0.87 for VCTE, and 0.77–0.84 for pSWE).
Table 3. Comparison of selected non-invasive markers.
NFS APRI FibroTest FIB-4 VCTE pSWE SWE MRE
Application Developed specifically Developed in context of Developed in Developed in context of Can be applied to any CLD Can be applied Can be applied Can be applied to any CLD
for NAFLD HCV, but can be context of HCV, HIV/HCV, but can be to any CLD to any CLD
applied to any CLD but can be applied to any CLD
applied to any
CLD
Point-of-Care Yes Yes No Yes Yes Yes Yes No
Most + ++ ++ + ++ + + +
clinically
used/
validated
Strength of +++ + + ++ + + + +
validation (NPV >90% for ≥F3 (NPV >90% for ≥F3
in NAFLD and F4) and F4)
Strength of N/A +++ +++ ++ +++ ++ ++ +
validation (HBV<HCV) (HBV<HCV) (HBV<HCV) (HBV = HCV) (HBV<HCV) (HBV<HCV)
in viral overlap of values between
hepatitis adjacent stages of fibrosis; most
accurate non-invasive marker
for detecting cirrhosis
Strength of N/A + ++ + + (elevated LSM in alcoholic + + +
validation hepatitis)
in
alcoholic
liver
disease
Other Two cutoffs result in an Two cutoffs result in an Patented test- not Two cutoffs result in an May need to use XL probe for Difficulty differentiating intermediate
comments intermediate range intermediate range readily available; intermediate range patients with increased stages of fibrosis
where further where further testing Outperforms non- where further testing abdominal girth;
testing is required is required; patented tests for is required; May need to consider
May need to ≥F2 May need to aminotransferase elevation,
consider False positives consider when interpreting results;
aminotransferase occur in setting of aminotransferase NPV > 90% for F4
elevation, when hemolysis or elevation, when
interpreting results Gilbert syndrome interpreting results
Difficulty differentiating Needs more validation
intermediate stages but appears to be
of fibrosis most accurate for
detecting
intermediate stages
of fibrosis in NAFLD
APRI: aspartate aminotransferase to platelet ratio index; HBV: hepatitis B virus; HCV: hepatitis C virus; HIV: human immunodeficiency virus; CLD: chronic liver disease; MRE: magnetic resonance elastography; NAFLD: non-
alcoholic fatty liver disease; NFS: non-alcoholic fatty liver disease fibrosis score; NPV: negative predictive value; pSWE: point-shear wave elastography; SWE: shear wave elastography.
EXPERT REVIEW OF GASTROENTEROLOGY & HEPATOLOGY
5
6
U. AGBIM AND S. K. ASRANI

Table 4. Summary of meta-analysis assessing diagnostic accuracy of noninvasive markers in fibrosis.


F≥3 F4
Patients Summary AUROC Patients Summary AUROC
Meta-analysis Disease (n of studies), % F ≥ 3 Cutoff (range) Sn (%) Sp (%) NPV (%) PPV (%) value LR+ LR- (n of studies), % F4 Cutoff (range) Sn (%) Sp (%) NPV (%) PPV (%) value LR+ LR-
APRI[56] NAFLD 682 (4) 1.5 32.9 90.5 79.1 55.5 0.77* N/A N/A 790 (5) 0.54–2.00 56.2 83.6 91.7 37.8 0.76* N/A N/A
BARD[56] NAFLD 3057 (14) 2 75.2 61.6 88.7 38.3 0.76* N/A N/A 242 (1) 3 52.2 83.8 94.3 25.3 N/A N/A N/A
FIB-4[56] NAFLD 1910 (6) 2.67 31.9 95.7 85.0 66.0 0.84* N/A N/A 439 (2) 1.92–2.48 76.4 82.4 96.2 39.0 N/A N/A N/A
NFS[56] NAFLD 3896 (14) 0.67–0.676 43.1 88.4 88.5 66.9 0.84* N/A N/A 197 (1) −0.014 80.0 80.8 95.7 42.8 N/A N/A N/A
FibroScan M probe[56] NAFLD 1540 (9) 7.6–8 88.9 77.2 95.5 43.4 0.88* N/A N/A 1362 (9) 10.3–11.3 87.7 86.3 98.0 46.8 0.94* N/A N/A
FibroScan XL probe[56] NAFLD 579 (3) 5.7–9.3 75.3 74.0 88.7 58.7 0.85* N/A N/A 654 (4) 7.2–16 87.8 82.0 97.8 39.8 0.91* N/A N/A
SWE[56] NAFLD 429 (3) 3.02–10.6 89.9 91.8 93.4 88.2 0.95* N/A N/A 181 (1) 3.36 100 85.6 100 55.2 N/A N/A N/A
MRE[56] NAFLD 628 (5) 3.62–4.8 85.7 90.8 93.4 71.0 0.96* N/A N/A 384 (3) 4.15–6.7 86.6 93.4 98.8 53.4 0.97* 13.1 N/A
FibroScan[79] Alcohol 564 (8) 9.5 92 70 N/A N/A 3.1 0.11 330 (7) 12.5 95 71 N/A N/A N/A 3.3 0.07
FIB-4[39] HBV 1473 (6), 33.5 1.45–1.65 68 75 N/A N/A 0.77 2.75 0.42 2105 (10), 20.8 2.9–3.6 42 96 N/A N/A 0.96 13.38 0.3
FibroScan[77] HBV 2437 (9) 9.15 79.0 84.6 N/A N/A 0.893 5.23 0.25 2522 (11) 12.17 80 86.6 N/A N/A 0.905 5.81 0.25
FibroScan[76] HBV (19) 9.4 81.9 86.6 N/A N/A 0.91 N/A N/A (24) 9.0–16.9 86.3 87.5 N/A N/A 0.93 N/A N/A
MRE[77] HBV 1470 (9) 3.62 89.6 93.2 N/A N/A 0.972 10.87 0.09 1470 (9) 4.63 89.5 92 N/A N/A 0.972 10.74 0.10
APRI[40] HCV 441 (13), 28 1.0 61 64 81 40 0.80 N/A N/A 4548 (18), 19 1.0 76 72 69 55 0.83 N/A N/A
2.0 46 91 63 82
FibroScan XL probe [41] All CLD (7) 9.5 66 82 0.83 N/A N/A (6) 12.5 84 78 0.88 N/A N/A
ARFI[42] All CLD 518 (8), 42.6 1.55 86 86 89 82 0.91 N/A N/A 518 (8), 26.6 1.80 92 86 97 71 0.93 N/A N/A
ARFI[43] All CLD (10) 1.43–1.81 81 84 N/A N/A 0.93 5.75 0.13 (13) 1.55–2.25 88 83 N/A N/A 0.94 6.67 0.17
MRE[43] All CLD (11) 3.1–6.47 87 92 N/A N/A 0.96 10.22 0.15 (8) 4–6.47 93 91 N/A N/A 0.97 8.85 0.06

APRI: aspartate aminotransferase to platelet ratio index; ARFI: acoustic radiation force impulse; AUROC: area under the receiver operator characteristic; CLD: chronic liver disease; HBV: hepatitis B virus; HCV: hepatitis C virus; kPa:
kilopascal; LR: likelihood ratio; MRE: magnetic resonance elastography; NAFLD: non-alcoholic fatty liver disease; NFS: non-alcoholic fatty liver disease fibrosis score; NPV: negative predictive value; PPV: positive predictive
value; Sn: Sensitivity; Sp: Specificity; SWE: shear wave elastography.
* Values for all cutoffs.
EXPERT REVIEW OF GASTROENTEROLOGY & HEPATOLOGY 7

Interestingly, 2D-SWE and VCTE had very similar cut-off scores at poor ability to discern NASH from normal liver, although the
the various stages of fibrosis, which may facilitate ease of use and AUROC value for MRE at discerning NASH is significantly higher
comparability [48]. than VCTE (0.70 vs 0.35; p = 0.0011) [53]. Although these tests
Comorbidities may attenuate the diagnostic accuracy of non- perform well in detecting fibrosis, more investigation is needed
invasive tests. Bertot et al. [49] noted decreased ability of to discern their ability in detecting NASH.
Hepascore, FIB-4, and APRI to differentiate cirrhosis as well as Currently, there is a lack of knowledge regarding cost-
predict decompensation in NAFLD subjects with diabetes. The effectiveness of the various modalities for diagnosing NAFLD
NFS had no difference in differentiating between outcomes as well as markers to distinguish simple steatosis from steato-
based on the presence of diabetes, likely due to the inclusion hepatitis. One study evaluating cost-effectiveness found that
of diabetes in the formula. Additionally, steatosis or obesity NFS, followed by the combination of NFS and VCTE is the most
affects the diagnostic performance of serum and imaging tech- cost-effective strategy to evaluate subjects with NAFLD in the
niques to detect fibrosis, as pSWE may be unreliable and 2D-SWE primary care setting [58].
may have a high degree of failure in obesity [48]. Further, VCTE Non-invasive markers have the ability to predict clinical out-
overestimates the amount of fibrosis present in the setting of comes in those with chronic liver disease. Using data from the
increased steatosis [48,50,51]. Controlled attenuation parameter National Health and Nutrition Examination Survey, Kim et al. [59]
(CAP) is a tool that can be coupled to VCTE to quantify steatosis, . were the first to demonstrate increasing mortality with
with increasing CAP values signifying more steatosis. However, advanced fibrosis scores. For instance, the adjusted hazard ratio
a recent secondary analysis of over 2000 subjects noted the use (HR) for the NFS, APRI, and FIB-4 for subjects with advanced
of CAP only slightly improves the NPV in select situations [51]. fibrosis compared to those without any fibrosis was 1.69 (95%
Other comorbidities associated with unreliable or failed liver confidence interval [CI] 1.09–2.63), 1.85 (95% CI 1.02–3.37), and
stiffness measurements (LSM) of VCTE, pSWE, and 2D-SWE 1.44 (95% CI 0.98–2.82), respectively, with most of the mortality
include increased waist circumference, BMI, intercostal wall thick- attributed to cardiovascular disease [59]. Similarly, in those with
ness, and the presence of metabolic syndrome [48]. NAFLD, a recent meta-analysis demonstrated an NFS > 0.676 is
Analogous to CAP, MRI-based proton density fat fraction associated with fourfold increased risk of mortality compared to
(PDFF) can also quantitatively assess hepatic fat content. MRE lower values, with a dose-response effect in mortality with
with PDFF performed better than VCTE with CAP using increases in NFS scores [60]. The FIB-4 and APRI were less effec-
the M probe in detecting F ≥ 2 (AUROC value of 0.91 vs tive at predicting mortality in NAFLD patients compared to NFS
0.82) and steatosis grade ≥2 (AUROC value 0.90 vs 0.73) [52]. [60]. Further, compared to its performance in other liver disease
Most of the studies addressing VCTE’s inadequate reliability in etiologies, FibroTest had the highest 10-year prognostic value in
the setting of obesity utilized the M probe without inclusion of predicting liver-related death in a cohort from France [61]. Non-
the XL probe. To address this limitation, a study in the United invasive tests may also assist in monitoring response to therapy,
States with both the M and XL probe found MRE with PDFF but presently, the data are limited. Post-hoc analysis from a trial
was significantly better than VCTE with CAP at diagnosing evaluating the use of obeticholic acid in NAFLD demonstrated
fibrosis as well as steatosis [53]. For instance, MRE was sig- improvement in histologic fibrosis was associated with improve-
nificantly able to detect fibrosis at earlier stages than VCTE, ment in APRI, FIB-4, and NFS compared to baseline scores sug-
whereas there were no differences in performance for ≥F2, gesting serum markers may be useful in monitoring evolution of
≥F3 and F4 [53]. Another study using both the M and XL probe treatment [62].
noted superior MRE performance for ≥F2 and ≥F3 when com- In summary, imaging tests are superior to serum tests in
pared to VCTE on an intention to diagnosis basis (for ≥F3, the non-invasive assessment of fibrosis in NAFLD subjects. The
AUROC value of 0.92 vs 0.84; p = 0.046), but MRE and VCTE evaluation of fibrosis by elastography can be challenging in
performed equally in these intermediate stages when exclud- NAFLD, but the available evidence reveals MRE is the most
ing unreliable VCTE examinations (for ≥F3, AUROC value of accurate modality, particularly in light of failed or unreliable
0.92 vs. 0.87; p = 0.150) [54]. LSM in the context of obesity with VCTE. Unfortunately, cost
A growing body of research demonstrates MRE’s superior and availability limit the use of MRE. As such, ultrasound-
ability to detect fibrosis in NAFLD. MRE outperformed pSWE in based modalities are viable contenders, with VCTE being the
determining all stages of fibrosis, particularly in the obese in most accessible modality.
a prospective cohort study of 125 biopsy-proven NAFLD patients
[55]. A recent meta-analysis of 64 studies with a total of 13,046
3.2. Viral hepatitis
NAFLD subjects demonstrated MRE and SWE had a significantly
greater AUROC value for detecting ≥F3 (0.96 and 0.95, respec- 3.2.1. Hepatitis C
tively) compared to FibroScan M probe (AUROC value 0.88), Non-invasive staging of fibrosis is incorporated in the manage-
FibroScan XL probe (AUROC value 0.85), NFS (AUROC value ment of chronic HCV in current society guidelines [63,64]. VCTE is
0.84), FIB-4 (AUROC value 0.84), APRI (AUROC value 0.77), and one of the most utilized non-invasive tool evaluating liver fibrosis
BARD score (AUROC value 0.76) (p < 0.01). The NPV for excluding in subjects with HCV with better diagnostic accuracy as com-
cirrhosis for all the tests was >90% [56]. Despite the evidence of pared to serum tests [63]. Overall VCTE cut-off values are well-
MRE’s superior discriminative ability in adults, it has not been established for the spectrum of fibrosis stages. However, in
well validated in children, as cut-off scores from adults may not a large US multicenter cohort study of subjects with HCV and
be applicable and further refinement is needed [57]. In terms of hepatitis B virus (HBV), VCTE could not validate cut-off values for
differentiating NASH from normal liver, both MRE and VCTE have ≥F2 as robustly as ≥F3 and F4 in the validation cohort,
8 U. AGBIM AND S. K. ASRANI

suggesting difficulty in differentiating intermediate stages of sites, 2D-SWE was superior to VCTE at discriminating among
fibrosis [65]. Use of both VCTE and serum biomarkers sequen- ≥F2, ≥F3, and F4 in HBV subjects [67]. In fact, using the new
tially, followed by a coupled test if the initial individual tests are DLRE with 2D-SWE, the AUROC value for ≥F3 was 0.98 and F4 was
discordant may help with these uncertainties [66]. Additionally, 0.97, which was significantly better than 2D-SWE, APRI, or FIB-4
2D-SWE is equivalent to VCTE in diagnosing fibrosis with similar alone [35]. Further, a meta-analysis has shown MRE is superior to
diagnostic performance [67]. VCTE for detecting ≥F2 (AUROC value 0.981 vs. 0.796; p < 0.001),
Evidence suggests longitudinal changes in non-invasive ≥F3 (AUROC value 0.972 vs. 0.893; p < 0.001), and F4 (AUROC
measures predict prognosis. When evaluating the evolution value 0.972 vs. 0.905; p < 0.001) [77]. In addition, LSM as mea-
of disease in subjects with HCV, those with a baseline LSM sured by VCTE and FibroTest had the best 5-year prognostic
< 7 kPa as measured by VCTE or with an increase in LSM value for survival compared to APRI, FIB-4, and liver biopsy in
of ≥1 kPa/year or with ≤0 kPa/year in those with a baseline a European cohort of Hepatitis B subjects, even after adjusting
LSM ≥ 14 kPa had increased risk of liver-related mortality for age, inflammatory activity, viral load, and treatment [78].
[68]. Newer direct antiviral agents have high rates of viral
clearance, improve subjects’ symptoms, and decrease their
3.3. Alcoholic liver disease
need for liver transplantation [69]. While using non-
invasive methods to stage disease in HCV is commonplace, In comparison to other chronic liver diseases, there is lack of
the role of non-invasive assessment in monitoring fibrosis literature examining diagnostic and prognostic management in
regression after achieving a sustained viral response (SVR) alcoholic liver disease [79,80], which is needed as a diagnosis of
is unclear. HCV viral clearance decreases LSM evaluated by fibrosis may compel subjects to abstain from alcohol use.
VCTE [70], FIB-4, APRI, and pSWE [71]. A recent meta- Although VCTE has been the most validated non-invasive test
analysis of 24 studies examining paired LSM prior to and used in alcoholic liver disease, a recent meta-analysis could not
after achieving an SVR, revealed the decrease in liver stiff- identify optimal cut-off values for VCTE in alcoholic liver disease,
ness (LS) magnified over time, which they postulate initi- as there was a paucity of studies included were the sole etiology
ally was attributed to the improvement in inflammation of disease was due to alcohol [79]. Nevertheless, studies have
with later declines in LS attributed to fibrosis regression suggested cut-off scores, and VCTE can be used to rule out ≥F3
[70]. However, decrease in liver stiffness is not equated and cirrhosis with negative likelihood ratios of 0.11 and 0.07,
with resolution of cirrhosis. Hence, despite decline in liver respectively [79].
stiffness, routine management of cirrhosis (e.g. HCC sur- In a multicenter prospective study of 217 subjects with heavy
veillance) after effective HCV therapy should continue. alcohol use, Voican et al. [80] evaluated liver biopsy, VCTE alone,
and VCTE paired with other non-invasive serum markers. At
3.2.2. Hepatitis B a cut-off score of ≥12 kPa for ≥F3 and 15 kPa for F4, VCTE had
The role of non-invasive testing in patients with HBV is less clear, a sensitivity of 92.2%, specificity of 75.6%, NPV of 84.8%, and
though recent society guidelines endorse the use of elastogra- AUROC value of 0.90 for ≥F3, and sensitivity of 85.4%, specificity
phy as the preferred test and FIB-4 or FibroTest as alternative of 93.1%, NPV of 98.6%, and AUROC value of 0.93 for F4. The
tests [72]. Additionally, the World Health Organization advises addition of FibroTest did not add further diagnostic value to
using APRI to stage disease among HBV subjects in resource- VCTE. The authors attributed the lower specificity of VCTE for
limited settings, whereas VCTE or FibroTest is the preferred test diagnosing ≥F3 and F4 to a high frequency of subjects with
of choice where cost and/or availability are not barriers [73]. alcoholic hepatitis seen on biopsy. Interestingly, after one
However, APRI has only modest accuracy in detecting fibrosis month of alcohol cessation, there was a significant decrease in
among all stages with a summary AUROC value of 0.74 for ≥F2, LS as measured by VCTE [80]. A similar finding of lower specificity
0.73 for ≥F3, and 0.73 for F4 [74]. Similarly, FIB-4 had moderate for pSWE was noted in a prospective study evaluating the diag-
accuracy in the same study, with a summary AUROC value of nostic performance of pSWE and paired liver biopsies in 82
0.78 for ≥F2, 0.82 for ≥F3, and 0.85 for F4 [74]. A new model, the alcoholics attending rehabilitation in France [81]. Kiani et al.
gamma-glutamyl transpeptidase to platelet ratio (GPR), was [81] noted at a cut-off value of 1.84 m/s for ≥F3 and 1.94 m/s
developed in France and Western Africa to evaluate fibrosis in for F4, pSWE had a sensitivity of 82.4%, specificity of 78.5%, NPV
subjects with HBV, particularly in low-resource settings. The of 94.4%, and AUROC value of 0.86 for ≥F3, and sensitivity of
AUROC value was significantly superior to APRI and FIB-4 at 92.3%, specificity of 81.6%, NPV of 98.2%, and AUROC value of
identifying ≥F2 and ≥F3 in the African training and validation 0.89 for F4. Evidence is mixed regarding the specificity of non-
cohorts [75]. In fact, using the WHO criteria’s APRI cut-off value invasive assessment of fibrosis in the context of alcoholic liver
of 2 to diagnosis cirrhosis [73], the sensitivity for diagnosis of F4 disease, as other studies demonstrate non-invasive tests have
was 25%, while the sensitivity of GPR was 85% at a cut-off value high specificities. In the largest study to date of subjects with
of 0.56 [75]. Further studies investigating the role of GPR are heavy alcohol use, Thiele et al. [82] showed VCTE and 2D-SWE
needed. both have very reliable diagnostic accuracy for evaluating
Similar to other liver diseases, VCTE has been recognized as advanced fibrosis (defined as Ishak ≥3) and cirrhosis (defined as
a very reliable and accurate tool for detecting fibrosis in subjects Ishak ≥5) with sensitivities ranging from 82% to 83%, specificities
with HBV, with summary AUROC values ranging from 0.80 to 0.88 ranging from 91% to 93%, NPVs of 88%, and AUROC values of
for ≥F2, 0.89–0.91 for ≥F3, and 0.91–0.93 for F4 [76,77]. 0.94–0.96 for advanced fibrosis, and sensitivities of 94–97%,
Notwithstanding, 2D-SWE and MRE may perform better than specificities of 90–91%, NPVs of 99% and AUROC valued of
VCTE. In a large-scale analysis of subjects from 13 different 0.95–0.96 for cirrhosis. They noted cut-off values for alcoholic
EXPERT REVIEW OF GASTROENTEROLOGY & HEPATOLOGY 9

liver disease are higher compared to viral liver disease due to the When evaluating the role of non-invasive markers in asses-
different patterns of fibrosis distribution in alcoholic liver disease sing fibrosis in primary sclerosing cholangitis (PSC), Corpechot
[82]. Overall, diagnostic cut-off values for elastography remain to et al. [90] noted VCTE had superior diagnostic accuracy to FIB-
be defined in the setting of alcoholic liver disease. 4, but similar to APRI and hyaluronic acid. Baseline LSM was
correlated with the risk of clinically significant outcomes such
as hepatic decompensation, liver transplantation, or death.
3.4. Autoimmune liver diseases Additionally, the progression rate of LSM exponentially
increased at ≥F2 and the authors postulate this may be used
Similar to alcoholic liver disease, non-invasive assessment of to stratify subjects who are at risk for liver-related complica-
fibrosis has not been rigorously studied in autoimmune liver tions [90]. Given that cholestatic diseases may have an uneven
disease, likely due to the rare nature of the disease. Studies geographical distribution throughout the liver, spleen dia-
reveal serum tests inferior to imaging tests such as VCTE meter has also been used to assess fibrosis in patients with
[83,84] or MRE when evaluating fibrosis in autoimmune hepa- PSC [91].
titis (AIH) [85] or in overlap diseases [86]. After defining opti-
mal cut-off values for VCTE with good diagnostic accuracy for
≥F2, ≥F3, and F4 in AIH, Hartl et al. [87] sought to determine
the impact of immunosuppressive therapy had on perfor- 3.5. Post-transplant care
mance of VCTE. They found a correlation between LSM and Non-invasive measures of fibrosis have been extensively stu-
histologic grade (Spearman’s ρ coefficient 0.558; p = 0.001), died and validated in native livers, but there is growing work
not stage (Spearman’s ρ coefficient 0.339; p = 0.19), in patients to explore their role in the post-transplant setting, where the
who had less than three months of immunosuppressive ther- allograft is often more susceptible to recurrent or de novo
apy. Conversely, LSM among subjects with longer treatment disease. Again, much of the available literature in the post-
intervals correlated more with stage (Spearman’s ρ coefficient transplant setting focuses on recipients with recurrent HCV
0.809; p < 0.0001 at 6–18 months after immunosuppression [92], but there are some data on all etiologies of liver disease
initiation) as opposed to histologic grade (Spearman’s ρ coef- [20] and even in children with biliary atresia [93]. In a meta-
ficient 0.404; p < 0.062 at 6–18 months after immunosuppres- analysis of 24 studies examining FIB-4, APRI, and VCTE in the
sion initiation). Further, the diagnostic accuracy of VCTE for post-transplant setting, Bhat et al. [20], noted they all had
staging fibrosis improved with increasing treatment time. As good diagnostic accuracy, but the diagnostic accuracy of
such, care is advised when interpreting LSM by VCTE in the VCTE was significantly superior to FIB-4 and APRI. In
months following treatment initiation, as in this study, inflam- a separate study of liver transplant recipients with recurrent
mation had a major impact on LS [87], although this was not HCV who had achieved a sustained viral response after ther-
confirmed in another study [84]. Additionally, VCTE may be apy, VCTE was accurate to diagnose the presence of ≥F3 at
used to monitor disease evolution to therapy. When evaluat- a cut-off value of 14 kPa and exclude ≥F3 at a cut-off value of
ing the impact of biochemical remission had on LSM in sub- 10.6 kPa with an AUROC value of 0.902, which outperformed
jects who had not achieved biochemical remission, LSM ELF (0.764; p = 0.002) [92]. Further, baseline and change in
strongly correlated with stage (Spearman’s ρ coefficient fibrosis scores with serial measurements may be able to pre-
0.801; p < 0.001) and weakly correlated with grade dict death and graft loss [94], as well as response to therapy
(Spearman’s ρ coefficient 0.312; p = 0.049). The same trend [92]. In a unique study in which liver biopsies, hepatic venous
held true for those who had achieved biochemical remission pressure gradient (HVPG), ELF score, and VCTE were measured
[87]. Angiotensin-converting enzyme, a responsible agent in at baseline and 12 months after achieving an SVR in liver
stellate cell proliferation and inflammation, may predict fibro- transplant recipients with recurrent HCV, Mauro and collea-
sis stages in AIH with 85.2% sensitivity, 94.8% specificity, and gues [92] noted LS was significantly lower in patients who
an AUROC value of 0.91 at a cut-off score of 64 U/l for ≥F3 and achieved fibrosis regression (12.6 kPa) than those who did not
100% sensitivity, 84.4% specificity and an AUROC value of 0.95 achieve regression after antiviral therapy (26 kPa).
at a cut-off value of 68 U/l for F4 [88]. Intermediate stages of fibrosis may be challenging to
detect by non-invasive tests. For example, Singh et al. [95]
3.4.1. Cholestatic liver disease demonstrated MRE is suitable for detecting cirrhosis in the
Few studies exist evaluating non-invasive assessment in primary post-transplant setting but inferior for detecting ≥F2 or ≥F3.
biliary cholangitis (PBC), but Corpechot et al. demonstrated VCTE In a pooled study of 141 liver transplant recipients from six
outperformed APRI, FIB-4, AST/ALT ratio, hyaluronic acid and the cohorts, the authors noted the summary AUROC value for
Mayo risk score in predicting ≥F2, ≥F3, and F4. In fact, they stage 1–4 fibrosis (≥F1), ≥F2, ≥F3, and F4 was 0.73, 0.69,
reported an increase in LSM of 2.1 kPa per year by VCTE was 0.83, and 0.96, respectively [95], which was notably different
associated with a 58% rate of morbidity/mortality including liver from a previous study evaluating fibrosis in native livers where
decompensation, liver transplantation, or death [89]. Wisteria the AUROC value for ≥F1, ≥F2, ≥F3, and F4 was 0.84, 0.88, 0.93,
floribunda agglutinin-positive Mac-2-binding protein (WFA and 0.92, respectively [96].
(+)-M2BP) was recently introduced as a fibrotic marker for PBC More data are needed to validate the modalities in the
with an AUROC value ≥0.93 for ≥F2, ≥F3, and F4 [25]. More work post-transplant population. However, its use may prompt clin-
is needed to assess various markers’ role in predicting fibrosis icians to perform more investigative analysis when uncertain-
in PBC. ties arise in the post-transplant setting. With more information
10 U. AGBIM AND S. K. ASRANI

regarding the validity, serial measurements could be utilized whether elevated values actually represent passive conges-
in lieu of routine protocol biopsy employed at many centers. tion, increased LS, or a combination of long-standing conges-
tion and ischemia from decreased cardiac output leading to
fibrosis [104].
3.6. Portal hypertension
Non-invasive measurements may help identify subjects that
need interventions to prevent portal hypertensive complica- 4. Expert opinion
tions. According to the Baveno VI proceedings [1], two sepa-
Given the limitations of liver biopsy, alternative strategies are
rate VCTE measurements of ≥20–25 kPa on different days for
needed to stage liver disease. Non-invasive serum and imaging
viral-related chronic liver disease is sufficient to rule-in clini-
tests are growing in utility, helping to assess fibrosis stage, assist
cally significant portal hypertension as defined by an HVPG
in prognosis of liver disease, and ultimately guide clinical deci-
≥10. In fact, the proceedings recommend avoiding screening
sions. Serum markers fill this unmet need, as they are simple,
endoscopy in any patient with VCTE kPa <20 and platelets
accessible, reproducible tests with good performance character-
over 150 X 109/L [1]. This has been validated in a subsequent
istics. In general, elastography outperforms the serum markers
study of 310 subjects to identify cirrhotics with high-risk
for detecting cirrhosis. VCTE, the most studied imaging modality,
varices, classifying 80% correctly with a sensitivity of 87%,
has good diagnostic accuracy for fibrosis, although its superior
specificity of 34%, NPV of 96% and AUROC value of 0.76 [97].
performance diminishes relative to serum markers for advanced
Further, in liver transplant recipients with recurrent HCV, VCTE
stages of fibrosis. Nevertheless, the utility of VCTE lies in its ability
and ELF measurements were accurate at diagnosing clinically
to provide point-of-care, accurate testing. MRE may outperform
significant portal hypertension [92]. VCTE and the combination
VCTE in intermediate stages, but its accessibility hinders its com-
of liver stiffness-spleen diameter to platelet ratio score may
monplace use. When utilizing non-invasive tests – both serum
also predict clinically significant portal hypertension [98].
and imaging tests, it is imperative to consider the underlying
disease process in the context of the selected tests and results.
Established cut-off values are available for VCTE to stage HCV,
3.7. Other disease processes and complications
which often guide treatment and surveillance decisions, particu-
Non-invasive measurements can be used to predict risk of HCC. larly in advanced disease.
In a retrospective study of 986 Korean HBV surface antigen Despite the advances in development of non-invasive tests,
positive subjects, Suh et al. [99] demonstrated high FIB-4 scores several questions remain. First, it is unclear how to best define
were more predictive of HCC than traditional ultrasonography intermediate stages of fibrosis, particularly concerning serum
(C-index: 0.775 vs. 0.701; p = 0.0040). This can be attributed to the tests with intermediate ranges, as this is where much uncer-
use of FIB-4 as marker of fibrosis whereas traditional ultrasono- tainty exists. Second, the appropriate combination of tests,
graphic methods are not an accurate reflection of fibrosis. whether in a deliberate, stepwise or simultaneous fashion
Further, after adjusting for age, sex, BMI, smoking, alcohol use, has yet to be defined. For example, using a combination
and HBV antiviral therapy, compared to subjects with a FIB-4 approach may afford the opportunity to classify cases that
score, those with a FIB-4 score 1.7–2.4 had adjusted HR of 4.57 otherwise would have been considered to lie in an intermedi-
(95% CI 1.50–13.92) of incident HCC, while those with a FIB-4 ate range when used with a singular test. Again, this will need
score of ≥2.4 had an HR of 21.34 (95% CI 7.73–58.92) compared to be tailored to individual diseases as the approaches and
to those with a FIB-4 score <1.25. Similarly, FIB-4 has a higher tests use may change in the setting of different diseases. Third,
predictive value of incident HCC in subjects with moderate to the utilization of these tests in routine management of liver
heavy alcohol use compared to traditional ultrasonography diseases other than deciding appropriate treatment time for
[100]. Conversely, LS as measured by VCTE was not associated HCV has not been determined. For instance, the role of non-
with HCC development in a cohort of Asian cirrhotics (>50% HBV invasive tests in longitudinal disease regression, particularly
prevalence), but was associated with the progression of portal with treatment of the underlying disease will need to be
hypertensive complications [101]. explored. Although there is some idea regarding changes in
The Fontan procedure is an operation to connect a single VCTE with disease regression, it is uncertain how to interpret
ventricle to the systemic circulation in addition to the ventricle improvement in many of the non-invasive serum tests when
receiving blood flow from the pulmonary circulation and can disease regression occurs. Similarly, how to utilize serial mea-
lead to a spectrum of liver complications. A common hepatic surements of non-invasive tests in the clinical monitoring of
consequence of the Fontan procedure is elevated hepatic progression of liver disease is not entirely clear. Additionally,
pressures from congestion, which over time can proceed to relatively little information exists on how to interpret the
fibrosis. Wu et al. [102] report LSM as indicated by VCTE over- magnitude of LSM above the cut-off value of cirrhosis or if
estimated the actual degree of fibrosis when using established this is significant at all. For example, does an elevated LSM
cut-off values in subjects with a Fontan. Serial LSM by MRE in beyond the cut-off value of cirrhosis for all the non-invasive
Fontan subjects correlates with liver disease severity as mea- modalities translate into elevated HVPG and thereby worsen-
sured by the Model for End Stage Liver Disease (MELD) score ing portal hypertension? Outside the Baveno VI proceedings,
and model for end stage liver disease excluding international there is little formal direction on how to interpret LSM in
normalized ratio (MELD-XI) [103]. Although the use of elasto- context of portal hypertension. Understanding how changes
graphy in Fontan related liver disease is evolving, it is unclear in LSM translate into clinically significant outcomes such as
EXPERT REVIEW OF GASTROENTEROLOGY & HEPATOLOGY 11

progression in portal hypertension will guide management non-invasive testing, however, should be avoided. Akin to
and improve care of patients with liver disease. blood work and other tools, context matters and novel tech-
With the revolutionary HCV treatment and eradication of niques should not replace clinical judgment, physical exam
the disease, a shift to investigate NAFLD has occurred, with and the art of medicine.
more emphasis placed on identifying subjects at risk for
advanced disease. These warrants further refinement to vali-
Funding
date imaging tests in NAFLD since obesity and steatosis can
decrease accuracy. As understanding and refinement of tech- This paper was funded by a Baylor Foundation Grant.
niques improve so will the need to identify and risk-stratify
patients. This means more investigation is warranted to
Declaration of interest
develop clinical prediction tools and screening algorithms
directing how to employ these tests in the general population The authors have no relevant affiliations or financial involvement with any
organization or entity with a financial interest in or financial conflict with
and/or in select patient populations. Ideally, some simple
the subject matter or materials discussed in the manuscript. This includes
algorithms could be implemented outside of the gastroenter- employment, consultancies, honoraria, stock ownership or options, expert
ologists’ or hepatologists’ office and implemented in the pri- testimony, grants or patents received or pending, or royalties.
mary care setting with subsequent referral to subspecialty
care.
In general, the goal of these tests is to minimize the need Reviewer disclosures
for routine biopsy. Certainly, this represents an exciting time Peer reviewers on this manuscript have no relevant financial or other
for novel approaches exploring the role of non-invasive fibro- relationships to disclose.
sis staging in liver disease.
References
Papers of special note have been highlighted as either of interest (•) or of
5. Five-year view considerable interest (••) to readers.
1. de Franchis R, Baveno VIF. Expanding consensus in portal hyper-
Non-invasive tests, particularly imaging, will become more rou-
tension: report of the Baveno VI consensus workshop: stratifying
tine in clinical practice with systemic implementation occurring risk and individualizing care for portal hypertension. J Hepatol.
to guide management of patients. Serum markers such as GTP, 2015 Sep;63(3):743–752. PubMed PMID: 26047908; eng.
which are simple and easily accessible, will be optimized for use 2. Rockey DC, Caldwell SH, Goodman ZD, et al. Liver biopsy. Hepatology.
in low-resource populations and a combination of serial serum 2009 Mar;49(3):1017–1044. PubMed PMID: 19243014; eng.
tests and/or imaging tests will be further refined and used to 3. Regev A, Berho M, Jeffers LJ, et al. Sampling error and intraobserver
variation in liver biopsy in patients with chronic HCV infection. Am
stage disease. The – omics (e.g. metabolomics, genomics, and J Gastroenterol. 2002 Oct;97(10):2614–2618. PubMed PMID:
proteomics) are an emerging and simulating area, which will 12385448; eng.
push the field forward and will continue to be added to our 4. Poynard T, Lenaour G, Vaillant JC, et al. Liver biopsy analysis has
research and clinical armamentarium. Novel imaging methods a low level of performance for diagnosis of intermediate stages of
fibrosis. Clin Gastroenterol Hepatol. 2012 Jun;10(6):657–63 e7.
including DWI, gadolinium-based imaging, and 3D-MRI and
PubMed PMID: 22343514; eng.
other dynamic techniques may be utilized and incorporated 5. Castera L. Noninvasive assessment of liver fibrosis. Dig Dis. 2015;33
into clinical prediction tools for fibrosis in the future. These (4):498–503. PubMed PMID: 26159265; eng.
novel imaging methods combined with the -omics should be 6. Yin M, Venkatesh SK. Ultrasound or MR elastography of liver: which
particularly salient tools to optimize detection of NAFLD-related one shall I use? Abdom Radiol (NY). 2018 Jul;43(7):1546–1551.
PubMed PMID: 28988274; eng.
fibrosis.
7. Horowitz JM, Venkatesh SK, Ehman RL, et al. Evaluation of hepatic
Future studies will assess accuracy of non-invasive modalities fibrosis: a review from the society of abdominal radiology disease
in children, as this is not a population in which non-invasive tests focus panel. Abdom Radiol (NY). 2017 Aug;42(8):2037–2053.
have been well characterized or validated. More research will PubMed PMID: 28624924; PubMed Central PMCID:
delve into how to account for the confounding effect of severe PMCPMC5994771. eng.
inflammation on LSM (e.g. as in flares of autoimmune disease or 8. Forns X, Ampurdanes S, Llovet JM, et al. Identification of chronic
hepatitis C patients without hepatic fibrosis by a simple predictive
alcoholic hepatitis), and the effect of hepatic congestion from model. Hepatology. 2002 Oct;36(4 Pt 1):986–992. PubMed PMID:
cardiac etiologies on LSM, as these factors may complicate fibro- 12297848; eng.
sis assessment. The role of non-invasive markers may evolve to 9. Lemoine M, Shimakawa Y, Nayagam S, et al. The gamma-glutamyl
include prognosis given the advent of effective therapy for liver transpeptidase to platelet ratio (GPR) predicts significant liver fibro-
sis and cirrhosis in patients with chronic HBV infection in West
disease especially in patients with HCV. In the years to come, liver
Africa. Gut. 2016 Aug;65(8):1369-76. doi: 10.1136/gutjnl-2015-
biopsy as the reference standard will continue to be challenged 309260. PubMed PMID: 26109530; PubMed Central PMCID:
by refinement in these exciting non-invasive tools. Clinical trials PMCPMC4975834. eng.
will incorporate non-invasive markers and tests to select partici- 10. Lok AS, Ghany MG, Goodman ZD, et al. Predicting cirrhosis in
pants, monitor the evolution of disease, and evaluate treatment patients with hepatitis C based on standard laboratory tests: results
of the HALT-C cohort. Hepatology. 2005 Aug;42(2):282–292.
response to drugs and interventions, which will make it an
PubMed PMID: 15986415; eng.
attractive option compared to serial liver biopsies. 11. Poynard T, Imbert-Bismut F, Munteanu M, et al. Overview of the
An array of highly sensitive and specific diagnostic tests diagnostic value of biochemical markers of liver fibrosis (FibroTest,
exist for clinicians to use for fibrosis staging. Overreliance on HCV FibroSure) and necrosis (ActiTest) in patients with chronic
12 U. AGBIM AND S. K. ASRANI

hepatitis C. Comparative hepatology. 2004 Sep 23;3(1):8. doi: 30. Hagan M, Asrani SK, Talwalkar J. Non-invasive assessment of liver
10.1186/1476-5926-3-8. PubMed PMID: 15387887; PubMed fibrosis and prognosis. Expert Rev Gastroenterol Hepatol. 2015;9
Central PMCID: PMCPMC522750. eng. (10):1251–1260. PubMed PMID: 26377444; eng.
12. Cales P, Oberti F, Michalak S, et al. A novel panel of blood markers 31. Friedrich-Rust M, Poynard T, Castera L. Critical comparison of elas-
to assess the degree of liver fibrosis. Hepatology. 2005 Dec;42 tography methods to assess chronic liver disease. Nat Clin Pract
(6):1373–1381. PubMed PMID: 16317693; eng. Gastroenterol Hepatol. 2016 Jul;13(7):402–411. PubMed PMID:
13. Cales P, Laine F, Boursier J, et al. Comparison of blood tests for liver 27273167; eng.
fibrosis specific or not to NAFLD. J Hepatol. 2009 Jan;50(1):165–173. 32. Kim S, Kang Y, Lee MJ, et al. Points to be considered when applying
PubMed PMID: 18977552; eng. FibroScan S probe in children with biliary atresia. J Pediatr
14. Adams LA, Bulsara M, Rossi E, et al. Hepascore: an accurate vali- Gastroenterol Nutr. 2014 Nov;59(5):624–628. PubMed PMID:
dated predictor of liver fibrosis in chronic hepatitis C infection. Clin 25003372; PubMed Central PMCID: PMCPMC4222704. eng.
Chem. 2005 Oct;51(10):1867–1873. PubMed PMID: 16055434; eng. 33. Friedrich-Rust M, Ong MF, Martens S, et al. Performance of transi-
15. Angulo P, Hui JM, Marchesini G, et al. The NAFLD fibrosis score: ent elastography for the staging of liver fibrosis: a meta-analysis.
a noninvasive system that identifies liver fibrosis in patients with Gastroenterology. 2008 Apr;134(4):960–974. PubMed PMID:
NAFLD. Hepatology. 2007 Apr;45(4):846–854. PubMed PMID: 18395077; eng.
17393509; eng. 34. Boursier J, Cassinotto C, Hunault G, et al. Criteria to determine
16. Harrison SA, Oliver D, Arnold HL, et al. Development and validation reliability of noninvasive assessment of liver fibrosis with virtual
of a simple NAFLD clinical scoring system for identifying patients touch quantification. Clin Gastroenterol Hepatol. 2018 May 9.
without advanced disease. Gut. 2008 Oct;57(10):1441–1447. PubMed PMID: 29753082; eng. DOI:10.1016/j.cgh.2018.04.062
PubMed PMID: 18390575; eng. 35. Wang K, Lu X, Zhou H, et al. Deep learning radiomics of shear wave
17. Sterling RK, Lissen E, Clumeck N, et al. Development of a simple elastography significantly improved diagnostic performance for
noninvasive index to predict significant fibrosis in patients with assessing liver fibrosis in chronic hepatitis B: a prospective multi-
HIV/HCV coinfection. Hepatology. 2006 Jun;43(6):1317–1325. centre study. Gut. 2018 May 5. PubMed PMID: 29730602; eng.
PubMed PMID: 16729309; eng. DOI:10.1136/gutjnl-2018-316204
18. Wai CT, Greenson JK, Fontana RJ, et al. A simple noninvasive index 36. Venkatesh SK, Talwalkar JA. When and how to use magnetic reso-
can predict both significant fibrosis and cirrhosis in patients with nance elastography for patients with liver disease in clinical
chronic hepatitis C. Hepatology. 2003 Aug;38(2):518–526. PubMed practice. Am J Gastroenterol. 2018 Jul;113(7):923–926. PubMed
PMID: 12883497; . PMID: 29487410; eng.
19. Boursier J, Vergniol J, Guillet A, et al. Diagnostic accuracy and 37. Gabbani T, Marsico M, Bernini P, et al. Metabolomic analysis with
prognostic significance of blood fibrosis tests and liver stiffness (1)H-NMR for non-invasive diagnosis of hepatic fibrosis degree in
measurement by FibroScan in non-alcoholic fatty liver disease. patients with chronic hepatitis C. Dig Liver Dis. 2017 Dec;49
J Hepatol. 2016 Sep;65(3):570–578. PubMed PMID: 27151181; eng. (12):1338–1344. PubMed PMID: 28625405; eng.
20. Bhat M, Tazari M, Sebastiani G. Performance of transient elastogra- 38. Loomba R, Cui J, Wolfson T, et al. Novel 3D magnetic resonance
phy and serum fibrosis biomarkers for non-invasive evaluation of elastography for the noninvasive diagnosis of advanced fibrosis in
recurrent fibrosis after liver transplantation: A meta-analysis. PloS NAFLD: a prospective study. Am J Gastroenterol. 2016 Jul;111
one. 2017;12(9):e0185192. PubMed PMID: 28953939; PubMed (7):986–994. PubMed PMID: 27002798; PubMed Central PMCID:
Central PMCID: PMCPMC5617176. Eng. PMCPMC5001170. eng.
21. Hansen JF, Juul Nielsen M, Nystrom K, et al. PRO-C3: a new and 39. Li Y, Chen Y, Zhao Y. The diagnostic value of the FIB-4 index for
more precise collagen marker for liver fibrosis in patients with staging hepatitis B-related fibrosis: a meta-analysis. PloS one.
chronic hepatitis C. Scand J Gastroenterol. 2018 Jan;53(1):83–87. 2014;9(8):e105728. PubMed PMID: 25165830; PubMed Central
PubMed PMID: 29069995; eng. PMCID: PMCPMC4148327. eng.
22. Neuman MG, Cohen LB, Nanau RM. Hyaluronic acid as a 40. Lin ZH, Xin YN, Dong QJ, et al. Performance of the aspartate
non-invasive biomarker of liver fibrosis. Clin Biochem. 2016 aminotransferase-to-platelet ratio index for the staging of hepatitis
Feb;49(3):302–315. PubMed PMID: 26188920; eng. C-related fibrosis: an updated meta-analysis. Hepatology. 2011
23. Murawaki Y, Ikuta Y, Idobe Y, et al. Tissue inhibitor of Mar;53(3):726–736. PubMed PMID: 21319189; eng.
metalloproteinase-1 in the liver of patients with chronic liver disease. 41. Xia B, Wang F, Friedrich-Rust M, et al. Feasibility and efficacy of
J Hepatol. 1997 Jun;26(6):1213–1219. PubMed PMID: 9210606; eng. transient elastography using the XL probe to diagnose liver fibrosis
24. Ito K, Murotani K, Nakade Y, et al. Serum Wisteria floribunda and cirrhosis: a meta-analysis. Medicine (Baltimore). 2018 Sep;97
agglutinin-positive Mac-2-binding protein levels and liver fibrosis: (39):e11816. PubMed PMID: 30278481; PubMed Central PMCID:
a meta-analysis. J Gastroenterol Hepatol. 2017 Dec;32 PMCPMC6181481. eng.
(12):1922–1930. PubMed PMID: 28406534; eng. 42. Friedrich-Rust M, Nierhoff J, Lupsor M, et al. Performance of acous-
25. Umemura T, Joshita S, Sekiguchi T, et al. Serum Wisteria floribunda tic radiation force impulse imaging for the staging of liver fibrosis:
agglutinin-positive mac-2-binding protein level predicts liver fibro- a pooled meta-analysis. J Viral Hepat. 2012 Feb;19(2):e212–9.
sis and prognosis in primary biliary cirrhosis. Am J Gastroenterol. PubMed PMID: 22239521; eng.
2015 Jun;110(6):857–864. PubMed PMID: 25916223; eng. 43. Guo Y, Parthasarathy S, Goyal P, et al. Magnetic resonance elasto-
26. Rosenberg WM, Voelker M, Thiel R, et al. Serum markers detect the graphy and acoustic radiation force impulse for staging hepatic
presence of liver fibrosis: a cohort study. Gastroenterology. 2004 fibrosis: a meta-analysis. Abdominal Imaging. 2015 Apr;40
Dec;127(6):1704–1713. PubMed PMID: 15578508; eng. (4):818–834. PubMed PMID: 24711064; eng.
27. Lichtinghagen R, Pietsch D, Bantel H, et al. The enhanced liver 44. Hernaez R, Lazo M, Bonekamp S, et al. Diagnostic accuracy and
fibrosis (ELF) score: normal values, influence factors and proposed reliability of ultrasonography for the detection of fatty liver: a
cut-off values. J Hepatol. 2013 Aug;59(2):236–242. PubMed PMID: meta-analysis. Hepatology. 2011 Sep 2;54(3):1082–1090. PubMed
23523583; eng. PMID: 21618575; PubMed Central PMCID: PMCPMC4197002. eng.
28. Tang A, Cloutier G, Szeverenyi NM, et al. Ultrasound elastography 45. Ekstedt M, Hagstrom H, Nasr P, et al. Fibrosis stage is the strongest
and MR elastography for assessing liver fibrosis: part 1, principles predictor for disease-specific mortality in NAFLD after up to 33
and techniques. AJR. 2015 Jul;205(1):22–32. PubMed PMID: years of follow-up. Hepatology. 2015 May;61(5):1547–1554.
25905647; PubMed Central PMCID: PMCPMC4819982. eng. PubMed PMID: 25125077; eng.
29. Barr RG, Ferraioli G, Palmeri ML, et al. Elastography assessment of 46. Younossi ZM, Koenig AB, Abdelatif D, et al. Global epidemiology of
liver fibrosis: society of radiologists in ultrasound consensus con- nonalcoholic fatty liver disease-meta-analytic assessment of preva-
ference statement. Radiology. 2015 Sep;276(3):845–861. PubMed lence, incidence, and outcomes. Hepatology. 2016 Jul;64(1):73–84.
PMID: 26079489; eng. PubMed PMID: 26707365.
EXPERT REVIEW OF GASTROENTEROLOGY & HEPATOLOGY 13

47. Petta S, Wong VW, Camma C, et al. Serial combination of Aliment Pharmacol Ther. 2018 Oct;17(48):1117–1127. PubMed
non-invasive tools improves the diagnostic accuracy of severe PMID: 30334263; eng.
liver fibrosis in patients with NAFLD. Aliment Pharmacol Ther. 62. Chalasani N, Abdelmalek MF, Loomba R, et al. Relationship between
2017 Sep;46(6):617–627. PubMed PMID: 28752524; eng. three commonly used non-invasive fibrosis biomarkers and improve-
48. Cassinotto C, Boursier J, de Ledinghen V, et al. Liver stiffness in ment in fibrosis stage in patients with NASH. Liver Int. 2018 Sep 25.
nonalcoholic fatty liver disease: a comparison of supersonic shear PubMed PMID: 30253043; eng. DOI:10.1111/liv.13974
imaging, fibroscan, and ARFI with liver biopsy. Hepatology. 2016 63. European Association for Study of L, Asociacion Latinoamericana para
Jun;63(6):1817–1827. PubMed PMID: 26659452; eng. el Estudio del H. EASL-ALEH clinical practice guidelines: non-invasive
49. Bertot LC, Jeffrey GP, de Boer B, et al. Diabetes impacts prediction tests for evaluation of liver disease severity and prognosis. J Hepatol.
of cirrhosis and prognosis by non-invasive fibrosis models in 2015 Jul;63(1):237–264. PubMed PMID: 25911335; eng.
non-alcoholic fatty liver disease. Liver Int. 2018 Mar 25. PubMed 64. AASLD-IDSA. HCV testing and linkage to care; [cited 2018 May 18].
PMID: 29575516; eng. DOI:10.1111/liv.13739 Available from: https://www.hcvguidelines.org/evaluate/testing-
50. Joo SK, Kim W, Kim D, et al. Steatosis severity affects the diagnostic and-linkage
performances of noninvasive fibrosis tests in nonalcoholic fatty 65. Afdhal NH, Bacon BR, Patel K, et al. Accuracy of fibroscan, com-
liver disease. Liver Int. 2018 Feb;38(2):331–341. PubMed PMID: pared with histology, in analysis of liver fibrosis in patients with
28796410; eng. hepatitis B or C: a United States multicenter study. Clin
51. Karlas T, Petroff D, Sasso M, et al. Impact of controlled attenuation Gastroenterol Hepatol. 2015 Apr;13(4):772–9 e1–3. PubMed PMID:
parameter on detecting fibrosis using liver stiffness measurement. 25528010; eng.
Aliment Pharmacol Ther. 2018 Apr;47(7):989–1000. PubMed PMID: 66. Cales P, Boursier J, Lebigot J, et al. Liver fibrosis diagnosis by blood
29446106; eng. test and elastography in chronic hepatitis C: agreement or
52. Imajo K, Kessoku T, Honda Y, et al. Magnetic resonance imaging combination? Aliment Pharmacol Ther. 2017 Apr;45(7):991–1003.
more accurately classifies steatosis and fibrosis in patients with PubMed PMID: 28164327; eng.
nonalcoholic fatty liver disease than transient elastography. 67. Herrmann E, de Ledinghen V, Cassinotto C, et al. Assessment of
Gastroenterology. 2016 Mar;150(3):626–637 e7. PubMed PMID: biopsy-proven liver fibrosis by two-dimensional shear wave elasto-
26677985; eng. graphy: an individual patient data-based meta-analysis.
53. Park CC, Nguyen P, Hernandez C, et al. Magnetic resonance elasto- Hepatology. 2018 Jan;67(1):260–272. PubMed PMID: 28370257;
graphy vs transient elastography in detection of fibrosis and non- PubMed Central PMCID: PMCPMC5765493. eng.
invasive measurement of steatosis in patients with biopsy-proven 68. Vergniol J, Boursier J, Coutzac C, et al. Evolution of noninvasive
nonalcoholic fatty liver disease. Gastroenterology. 2017 Feb;152 tests of liver fibrosis is associated with prognosis in patients with
(3):598–607 e2. PubMed PMID: 27911262; PubMed Central PMCID: chronic hepatitis C. Hepatology. 2014 Jul;60(1):65–76. PubMed
PMCPMC5285304. eng. PMID: 24519328; eng.
54. Chen J, Yin M, Talwalkar JA, et al. Diagnostic performance of mr 69. Pascasio JM, Vinaixa C, Ferrer MT, et al. Clinical outcomes of
elastography and vibration-controlled transient elastography in the patients undergoing antiviral therapy while awaiting liver
detection of hepatic fibrosis in patients with severe to morbid transplantation. J Hepatol. 2017 Dec;67(6):1168–1176. PubMed
obesity. Radiology. 2017 May;283(2):418–428. PubMed PMID: PMID: 28842296; eng.
27861111; PubMed Central PMCID: PMCPMC5395333. eng. 70. Singh S, Facciorusso A, Loomba R, et al. Magnitude and kinetics of
55. Cui J, Heba E, Hernandez C, et al. Magnetic resonance elastography decrease in liver stiffness after antiviral therapy in patients with
is superior to acoustic radiation force impulse for the diagnosis of chronic hepatitis C: a systematic review and meta-analysis. Clin
fibrosis in patients with biopsy-proven nonalcoholic fatty liver dis- Gastroenterol Hepatol. 2018 Jan;16(1):27–38 e4. PubMed PMID:
ease: a prospective study. Hepatology. 2016 Feb;63(2):453–461. 28479504; PubMed Central PMCID: PMCPMC5671365. eng.
PubMed PMID: 26560734; PubMed Central PMCID: 71. Chen SH, Lai HC, Chiang IP, et al. Changes in liver stiffness mea-
PMCPMC4818645. eng. surement using acoustic radiation force impulse elastography after
56. Xiao G, Zhu S, Xiao X, et al. Comparison of laboratory tests, ultra- antiviral therapy in patients with chronic hepatitis C. PloS one.
sound, or magnetic resonance elastography to detect fibrosis in 2018;13(1):e0190455. PubMed PMID: 29293628; PubMed Central
patients with nonalcoholic fatty liver disease: a meta-analysis. PMCID: PMCPMC5749809. eng.
Hepatology. 2017 Nov;66(5):1486–1501. PubMed PMID: 28586172; 72. Terrault NA, Lok ASF, McMahon BJ, et al. Update on prevention,
eng. diagnosis, and treatment of chronic hepatitis B: AASLD 2018 hepa-
57. Schwimmer JB, Behling C, Angeles JE, et al. Magnetic resonance titis B guidance. Hepatology. 2018 Apr;67(4):1560–1599. PubMed
elastography measured shear stiffness as a biomarker of fibrosis in PMID: 29405329; eng.
pediatric nonalcoholic fatty liver disease. Hepatology. 2017 Nov;66 73. WHO Guidelines Approved by the Guidelines Review Committee.
(5):1474–1485. PubMed PMID: 28493388; PubMed Central PMCID: Guidelines for the prevention, care and treatment of persons with
PMCPMC5650504. eng. chronic hepatitis B infection. WHO Guidelines Approved by the
58. Tapper EB. Building effective quality improvement programs for Guidelines Review Committee. Geneva: World Health
liver disease: a systematic review of quality improvement Organization; 2015. Copyright (c) World Health Organization 2015.
initiatives. Clin Gastroenterol Hepatol. 2016 Sep;14(9):1256–1265 74. Xiao G, Yang J, Yan L. Comparison of diagnostic accuracy of aspartate
e3. PubMed PMID: 27103114; eng. aminotransferase to platelet ratio index and fibrosis-4 index for
59. Kim D, Kim WR, Kim HJ, et al. Association between noninvasive detecting liver fibrosis in adult patients with chronic hepatitis
fibrosis markers and mortality among adults with nonalcoholic B virus infection: a systemic review and meta-analysis. Hepatology.
fatty liver disease in the United States. Hepatology. 2013 Apr;57 2015 Jan;61(1):292–302. PubMed PMID: 25132233; eng.
(4):1357–1365. PubMed PMID: 23175136; PubMed Central PMCID: 75. Lemoine M, Shimakawa Y, Nayagam S, et al. The gamma-glutamyl
PMCPMC3622816. eng. transpeptidase to platelet ratio (GPR) predicts significant liver fibro-
60. Salomone F, Micek A, Godos J. simple scores of fibrosis and mor- sis and cirrhosis in patients with chronic HBV infection in West
tality in patients with NAFLD: a systematic review with Africa. Gut. 2016 Aug;65(8):1369–1376. PubMed PMID: 26109530;
meta-analysis. J Clin Med. 2018 Aug 15;7(8). PubMed PMID: PubMed Central PMCID: PMCPMC4975834. eng.
30111756; PubMed Central PMCID: PMCPMC6111765. eng. 76. Li Y, Huang YS, Wang ZZ, et al. Systematic review with
DOI:10.3390/jcm7080219 meta-analysis: the diagnostic accuracy of transient elastography
61. Munteanu M, Pais R, Peta V, et al. Long-term prognostic value of for the staging of liver fibrosis in patients with chronic hepatitis
the FibroTest in patients with non-alcoholic fatty liver disease, B. Aliment Pharmacol Ther. 2016 Feb;43(4):458–469. PubMed PMID:
compared to chronic hepatitis C, B, and alcoholic liver disease. 26669632; eng.
14 U. AGBIM AND S. K. ASRANI

77. Xiao H, Shi M, Xie Y, et al. Comparison of diagnostic accuracy of 91. Ehlken H, Wroblewski R, Corpechot C, et al. Validation of transient
magnetic resonance elastography and Fibroscan for detecting liver elastography and comparison with spleen length measurement for
fibrosis in chronic hepatitis B patients: a systematic review and staging of fibrosis and clinical prognosis in primary sclerosing
meta-analysis. PloS one. 2017;12(11):e0186660. PubMed PMID: cholangitis. PloS one. 2016;11(10):e0164224. PubMed PMID:
29107943; PubMed Central PMCID: PMCPMC5673175. eng. 27723798; PubMed Central PMCID: PMCPMC5056739. eng.
78. de Ledinghen V, Vergniol J, Barthe C, et al. Non-invasive tests for 92. Mauro E, Crespo G, Montironi C, et al. Portal pressure and liver
fibrosis and liver stiffness predict 5-year survival of patients chroni- stiffness measurements in the prediction of fibrosis regression after
cally infected with hepatitis B virus. Aliment Pharmacol Ther. 2013 sustained virological response in recurrent hepatitis C. Hepatology.
May;37(10):979–988. PubMed PMID: 23557139; eng. 2018 May;67(5):1683–1694. PubMed PMID: 28960366; eng.
79. Pavlov CS, Casazza G, Nikolova D, et al. Systematic review with 93. Vinciguerra T, Brunati A, David E, et al. Transient elastography for
meta-analysis: diagnostic accuracy of transient elastography for staging non-invasive evaluation of post-transplant liver graft fibrosis in
of fibrosis in people with alcoholic liver disease. Aliment Pharmacol children. Pediatr Transplant. 2018 Mar;22(2). PubMed PMID:
Ther. 2016 Mar;43(5):575–585. PubMed PMID: 26791825; eng. 29369488; eng. DOI:10.1111/petr.13125
80. Voican CS, Louvet A, Trabut JB, et al. Transient elastography alone 94. Bhat M, Ghali P, Rollet-Kurhajec KC, et al. Serum fibrosis biomarkers
and in combination with FibroTest((R)) for the diagnosis of hepatic predict death and graft loss in liver transplantation recipients. Liver
fibrosis in alcoholic liver disease. Liver Int. 2017 Nov;37 Transplant. 2015 Nov;21(11):1383–1394. PubMed PMID: 26194602;
(11):1697–1705. PubMed PMID: 28387018; eng. eng.
81. Kiani A, Brun V, Laine F, et al. Acoustic radiation force impulse 95. Singh S, Venkatesh SK, Keaveny A, et al. Diagnostic accuracy of
imaging for assessing liver fibrosis in alcoholic liver disease. World magnetic resonance elastography in liver transplant recipients:
J Gastroenterol. 2016 May 28;22(20):4926–4935. PubMed PMID: a pooled analysis. Ann Hepatol. 2016 Jun;15(3):363–376. PubMed
27239119; PubMed Central PMCID: PMCPMC4873885. eng. PMID: 27049490; PubMed Central PMCID: PMCPMC5032623. eng.
82. Thiele M, Detlefsen S, Sevelsted Moller L, et al. Transient and 96. Singh S, Venkatesh SK, Wang Z, et al. Diagnostic performance of
2-dimensional shear-wave elastography provide comparable magnetic resonance elastography in staging liver fibrosis:
assessment of alcoholic liver fibrosis and cirrhosis. a systematic review and meta-analysis of individual participant
Gastroenterology. 2016 Jan;150(1):123–133. PubMed PMID: data. Clin Gastroenterol Hepatol. 2015 Mar;13(3):440–451 e6.
26435270; eng. PubMed PMID: 25305349; PubMed Central PMCID:
83. Guo L, Zheng L, Hu L, et al. Transient elastography (fibroscan) PMCPMC4333001. eng.
performs better than non-invasive markers in assessing liver fibro- 97. Maurice JB, Brodkin E, Arnold F, et al. Validation of the Baveno VI
sis and cirrhosis in autoimmune hepatitis patients. Med Sci Monit. criteria to identify low risk cirrhotic patients not requiring endo-
2017 Oct 26;23:5106–5112. PubMed PMID: 29073121; PubMed scopic surveillance for varices. J Hepatol. 2016 Nov;65(5):899–905.
Central PMCID: PMCPMC5669534. eng. PubMed PMID: 27388923; eng.
84. Xu Q, Sheng L, Bao H, et al. Evaluation of transient elastography 98. Cho EJ, Kim MY, Lee JH, et al. Diagnostic and prognostic values of
in assessing liver fibrosis in patients with autoimmune hepatitis. noninvasive predictors of portal hypertension in patients with
J Gastroenterol Hepatol. 2017 Mar;32(3):639–644. PubMed PMID: alcoholic cirrhosis. PloS one. 2015;10(7):e0133935. PubMed PMID:
27505153; eng. 26196942; PubMed Central PMCID: PMCPMC4511411. eng.
85. Wang J, Malik N, Yin M, et al. Magnetic resonance elastography is 99. Suh B, Park S, Shin DW, et al. High liver fibrosis index FIB-4 is highly
accurate in detecting advanced fibrosis in autoimmune hepatitis. predictive of hepatocellular carcinoma in chronic hepatitis B
World J Gastroenterol. 2017 Feb 7;23(5):859–868. PubMed PMID: carriers. Hepatology. 2015 Apr;61(4):1261–1268. PubMed PMID:
28223730; PubMed Central PMCID: PMCPMC5296202. eng. 25502481; eng.
86. Wu HM, Sheng L, Wang Q, et al. Performance of transient elasto- 100. Suh B, Yun JM, Park S, et al. Prediction of future hepatocellular
graphy in assessing liver fibrosis in patients with autoimmune carcinoma incidence in moderate to heavy alcohol drinkers with the
hepatitis-primary biliary cholangitis overlap syndrome. World FIB-4 liver fibrosis index. Cancer. 2015 Nov 1;121(21):3818–3825.
J Gastroenterol. 2018 Feb 14;24(6):737–743. PubMed PMID: PubMed PMID: 26178294; eng.
29456412; PubMed Central PMCID: PMCPMC5807676. eng. 101. Wang JH, Chuah SK, Lu SN, et al. Baseline and serial liver stiffness
87. Hartl J, Denzer U, Ehlken H, et al. Transient elastography in autoimmune measurement in prediction of portal hypertension progression for
hepatitis: timing determines the impact of inflammation and fibrosis. patients with compensated cirrhosis. Liver Int. 2014 Oct;34
J Hepatol. 2016 Oct;65(4):769–775. PubMed PMID: 27238753; eng. (9):1340–1348. PubMed PMID: 24620731; eng.
88. Efe C, Cengiz M, Kahramanoglu-Aksoy E, et al. Angiotensin- 102. Wu FM, Opotowsky AR, Raza R, et al. Transient elastography may
converting enzyme for noninvasive assessment of liver fibrosis in identify Fontan patients with unfavorable hemodynamics and
autoimmune hepatitis. Eur J Gastroenterol Hepatol. 2015 Jun;27 advanced hepatic fibrosis. Congenit Heart Dis. 2014 Sep;9
(6):649–654. PubMed PMID: 25860719; eng. (5):438–447. PubMed PMID: 24418160; eng.
89. Corpechot C, Carrat F, Poujol-Robert A, et al. Noninvasive 103. Egbe A, Miranda WR, Connolly HM, et al. Temporal changes in
elastography-based assessment of liver fibrosis progression and liver stiffness after Fontan operation: results of serial magnetic
prognosis in primary biliary cirrhosis. Hepatology. 2012 Jul;56 resonance elastography. Int J Cardiol. 2018 May;1(258):299–304.
(1):198–208. PubMed PMID: 22271046; eng. PubMed PMID: 29433966; eng.
90. Corpechot C, Gaouar F, El Naggar A, et al. Baseline values and 104. Poterucha JT, Johnson JN, Qureshi MY, et al. magnetic resonance
changes in liver stiffness measured by transient elastography are elastography: a novel technique for the detection of hepatic
associated with severity of fibrosis and outcomes of patients with fibrosis and hepatocellular carcinoma after the fontan
primary sclerosing cholangitis. Gastroenterology. 2014 Apr;146 operation. Mayo Clin Proc. 2015 Jul;90(7):882–894. PubMed
(4):970–9; quiz e15–6. PubMed PMID: 24389304; eng. PMID: 26059757; PubMed Central PMCID: PMCPMC4500048. eng.

You might also like