166

Diagnosis of Liver Fibrosis: Present and Future Keyur Patel, MD1

Pierre Bedossa, MD, PhD2

Laurent Castera, MD, PhD3

1 Division of Gastroenterology, Duke University Medical Center, Duke

Clinical Research Institute, Durham, North Carolina 2 Department of Pathology, Physiology and Imaging, Hôpital Beaujon, Clichy, France 3 Department of Hepatology, Hôpital Beaujon, Clichy, France

Address for correspondence Laurent Castera, MD, PhD, Department of Hepatology, Hôpital Beaujon, Clichy, France (e-mail: [email protected]).

Abstract

Keywords

► ► ► ► ► ►

noninvasive liver fibrosis biopsy serum biomarkers imaging liver stiffness

The diagnostic assessment of liver fibrosis, a major determinant of disease severity, is an important step in the management of patients with chronic liver diseases. Liver biopsy is still considered the gold standard for the assessment of necroinflammation and fibrosis; however, recent technical advances have resulted in the development of numerous serum biomarkers and imaging tools as noninvasive alternatives to biopsy. These tests include biological (serum biomarker algorithms), physical (imaging assessment of tissue stiffness), and physiological (breath test) methods. Accumulating evidence shows that noninvasive tests provide prognostic information of clinical relevance, which has led to their incorporation into clinical guidelines and everyday practice. Here, the authors review and compare invasive and noninvasive tests for the diagnosis of liver fibrosis. They discuss emerging functional genomic, microparticle, protein-profiling, and bioimaging tools, focusing on novel sensitive tools that are able to assess the dynamic nature of fibrogenesis, a key requirement for the assessment of the efficacy of antifibrotic compounds in the future.

Histologic assessment of liver disease severity has been a cornerstone for therapeutic decisions and predicting prognosis in chronic liver disease (CLD), and been a foundation for much of modern hepatology. Although liver biopsy is still regarded as the gold standard for assessment of injury, inflammation, and fibrosis stage, its clinical application has significantly decreased over the past two decades. At the same time, noninvasive approaches that avoid the risks and possibly overcome limitations of liver biopsy have been under development for the past two decades. Noninvasive assessment of liver fibrosis has been increasingly incorporated into clinical studies and into everyday clinical practice. As such, semiquantitative categorical grading systems for histopathologic analysis, such as METAVIR, were developed to standardize and improve observer variability to determine thresholds for therapy in viral hepatitis, but do not quantify linearity of fibrosis deposition or actual matrix content. Moreover, the

Issue Theme Liver Fibrosis; Guest Editors, Robert Schwabe, MD, and Ramon Bataller, MD, PhD

rapid evolution of simplified noninterferon (IFN) direct acting antiviral therapeutic regimens, with high efficacy and tolerability amongst all hepatitis C virus (HCV) genotypes, suggest that the need for a liver biopsy for staging disease prior to antiviral therapy may be limited. Many noninvasive approaches, initially developed for disease staging in chronic hepatitis C (CHC), continue to be refined for diagnostic use in other CLD such as nonalcoholic fatty liver disease (NAFLD). One of the main goals in developing noninvasive measures of fibrosis has been to overcome the diagnostic limitations of a liver biopsy—which include the only semiquantitative assessment of matrix content as well as sampling error. Many of the existing serum biomarker panels and imaging tools were developed in relation to a cross-sectional, binary assessment of categorical histopathologic scores, and not to specifically reflect the dynamic nature of liver fibrogenesis. As we consider how to best monitor efficacy measures for

Copyright © 2015 by Thieme Medical Publishers, Inc., 333 Seventh Avenue, New York, NY 10001, USA. Tel: +1(212) 584-4662.

DOI http://dx.doi.org/ 10.1055/s-0035-1550059. ISSN 0272-8087.

Downloaded by: University of Connecticut. Copyrighted material.

Semin Liver Dis 2015;35:166–183.

Diagnosis of Liver Fibrosis: Present and Future

Liver Biopsy The Reference Tool for Evaluation of Liver Fibrosis Hepatic fibrosis consists of the deposition of extracellular matrix (ECM) components in highly stable and optically visible structures (usually fibers) within the liver parenchyma.3 Because histological examination allows for direct visualization of the liver parenchyma, it is still considered as the reference tool for evaluation of fibrosis. However, liver biopsy has some disadvantages.4,5 Liver biopsy is an invasive procedure associated with morbidity (such as pain in 84%, and bleeding risk in 1 of 2,500 to 1 of 10,000 of patients), and mortality rates of 1 of 10,000. Moreover, the cost of a liver biopsy is high with conservative estimates at $1,032 and $2,745 for biopsies without and with complications, respectively.6 Actual costs may be higher depending on the type of complication, the need for further imaging, patient admission, and hospital facility fee. Because the liver specimen contained in a biopsy only represents a very small portion of the whole organ, there is a risk of sampling error due to heterogeneity in fibrosis distribution.7–9 Increasing the length of liver biopsy decreases the risk of such sampling error. Except for cirrhosis, for which microfragments may be sufficient, a 25-mm-long biopsy is considered an optimal specimen for an accurate evaluation, although 15 mm is considered sufficient in most studies.10–13 These thresholds have been set up in viral chronic hepatitis. Because of the variable fibrosis pattern depending on the etiology, it is possible that the recommended lengths may vary from disease to disease. Interobserver variation is another potential limit related to the discordance between pathologists in the interpretation of the biopsy.14 The use of histopathological semiquantitative scoring systems for evaluation of liver fibrosis (staging) may increase reproducibility because several studies have shown that different scoring systems provide more-consistent fibrosis evaluation especially when used by experienced liver pathologists.15–19 In this regard, specific fibrous tissue stains such as Masson’s trichrome or Sirius red are of great value for pathologists and may explain why fibrosis staging is one of the most reliable feature with the lowest interobserver variation. 18,20,21

167

Thus, although liver biopsy has limitations, appropriate precautions may reduce the flaws inherent to this technique.

Liver Biopsy and Patterns of Fibrosis Although fibrosis is the mainstay in CLD, the histological pattern of fibrosis is not unique. The extent and distribution of hepatic fibrosis have different patterns depending on the underlying disease, and can be useful in the identification of the cause of the liver disease. Excess fibrous tissue often develops within the portal tracts and expands into the adjacent parenchyma. This pattern is seen in conditions associated with persistent portal inflammation, including chronic viral hepatitis, inflammatory cholangiopathy, etc. Periportal fibrosis thus represent the first stage in the evolution to bridging fibrosis.22,23 Bridging fibrosis, also known as septal fibrosis, designates the presence of fibrous septa that extend across lobules and connect mesenchymal structures (portal tracts and central veins) in various arrangements. It is an important indicator of prognosis and marks the shift from minimal fibrosis to significant fibrosis.24 Based on the vascular structures involved, bridging fibrosis can be separated into portal-portal, portal-central, and central-central types. Although this classification is conceptually relevant, the nature of a fibrous bridge can be difficult to determine in biopsy specimens, especially in the precirrhosis stage. Cirrhosis is histologically defined as a diffuse process characterized by annular fibrosis and a conversion of normal architecture into structurally abnormal hepatocyte-containing nodules.25 Cirrhosis is associated with the development of intrahepatic portohepatic vascular shunts. Regeneration of hepatocytes may occur, but is impaired in patients with advanced cirrhosis. Currently, histopathologists do not consider cirrhosis as a unique entity, but a spectrum of disease with different degrees of severity.26,27 Pericellular fibrosis is characterized by collagen fibers that extend along the sinusoids and surround single or small groups of hepatocytes. On special stains, pericellular fibrosis has a chicken-wire appearance and because of its distribution, it is also referred as perisinusoidal fibrosis. The most prominent causes of pericellular fibrosis are alcoholic and nonalcoholic fatty liver diseases.28,29 In diabetes, pericellular fibrosis spreads across entire lobules, producing a pattern of diffuse intralobular fibrosis (diabetic hepatopathy).30

Liver Biopsy and Scoring Fibrosis (Staging) The development of scoring systems to quantify the degree of fibrosis has increased the reliability of histological assessment. Indeed, using accurate guidelines that include the presence and association of elementary lesions, most pathologists are able to accurately assess the degree of fibrosis. By categorizing patients into homogeneous groups with the same extent and severity of a disease, histological scores have helped to perform large, multicentric studies as well as clinical trials. These reasons explain why histological scores of fibrosis have been widely accepted for more than 40 years. Scoring systems of fibrosis exist not only for viral chronic hepatitis, but also for other CLD such as NAFLD and biliary Seminars in Liver Disease

Vol. 35

No. 2/2015

Downloaded by: University of Connecticut. Copyrighted material.

antifibrotic therapeutic development (see “Antifibrotic Therapies in the Liver” by Mehal and Schuppan in this issue), current noninvasive tests do not yet provide sufficiently reliable and sensitive assessment of quantitative changes in fibrosis. Although clinical practice guidelines for CHC and NAFLD now include noninvasive tests for assessing the presence or absence of advanced fibrosis, liver biopsy is still recommended for prognostic and therapeutic decisions.1,2 However, this is likely to change with increased availability, validation, and greater acceptance of noninvasive tests as an alternative to biopsy for diagnosis of advanced stage disease and determination of prognosis (►Table 1). Moreover, further developments will undoubtedly lead to noninvasive tests that will allow for reliable and sensitive assessment of quantitative changes, even in early stages of liver fibrosis.

Patel et al.

Seminars in Liver Disease

Vol. 35

No. 2/2015

•Unable to discriminate between intermediate stages of fibrosis •Units (m/s) different from that of TE (kPa) •Narrow range of values (0.5–4.4 m/s) •Quality criteria not well defined •Prognostic value in cirrhosis?

•Can be implemented on a regular US machine •ROI smaller than TE but chosen by the operator •Higher applicability than TE (ascites and obesity) •Performance equivalent to that of TE

ARFI

•Further validation warranted •Unable to discriminate between intermediate stages of fibrosis •Quality criteria not well defined •Learning curve? •Influence of inflammation?

•Can be implemented on a regular US machine •ROI can be adjusted in size and location and chosen by the operator •Measures liver stiffness in real-time •High range of values (2–150 kPa) •Good applicability •High performance for cirrhosis

SWE

•Further validation warranted especially in comparison with TE •Not applicable in case of iron overload •Requires a MRI facility •Time-consuming •Costly

•Can be implemented on a regular MRI machine •Examination of the whole liver •High applicability: overcomes the limitations of TE (ascites and obesity) •High performance for cirrhosis

MR elastography

Downloaded by: University of Connecticut. Copyrighted material.

Abbreviations: ARFI, acoustic radiation force impulse imaging; AUROC, area under ROC curve; MR, magnetic resonance; MRI, magnetic resonance imaging; ROI, region of interest; SWE, ShearWave elastography; TE, transient elastography; US, ultrasound.

•Requires a dedicated device •ROI cannot be chosen •Unable to discriminate between intermediate stages of fibrosis •Applicability (80%) lower than serum biomarker: (obesity, ascites, operator experi ence) •False-positive in case of acute hepatitis, extrahepatic cholestasis and congestion

•Most widely used and validated technique: standard to be beaten •User-friendly (performed at bedside; rapid, easy to learn) •High range of values (2–75 kPa) •Quality criteria well defined •Good reproducibility •High performance for cirrhosis (AUROC > 0.9) •Prognostic value in cirrhosis

Transient elastography

Measurement of liver stiffness

Diagnosis of Liver Fibrosis: Present and Future

•Nonspecific of the liver •Unable to discriminate between intermediate stages of fibrosis •Performance not as good as TE for cirrhosis •Cost and limited availability (proprietary) •Limitations (hemolysis, Gilbert’s syndrome, inflammation…)

Disadvantages

•Good reproducibility •High applicability (95%) •No cost and wide availability (nonpatented) •Well validated •Can be performed in the outpatient clinic

Advantages

Serum biomarkers

Table 1 Respective advantages and disadvantages of currently available noninvasive methods in patients with chronic liver disease

168 Patel et al.

Diagnosis of Liver Fibrosis: Present and Future

Liver Biopsy and Fibrosis Regression It is now well accepted that fibrosis and even cirrhosis may regress after successful treatment of the underlying disease.37,38 The landmark article by Wanless et al showed histological evidence from serial biopsies of patients who showed apparent shift from fully developed cirrhosis to incomplete septal cirrhosis.39 The most fascinating demonstration of the reversibility of fibrosis/cirrhosis came from studies analyzing large cohorts of patients with HCV or hepatitis B virus (HBV) effectively treated with new antiviral drugs and sequential biopsies with sufficient time interval.40–44 After viral suppression or eradication, most patients show regression of fibrosis, and regression of cirrhosis is observed in 50 to 60% of patients with a baseline diagnosis of cirrhosis. Cirrhosis regression is associated with a marked decrease in the amount of fibrosis that can easily be demonstrated by morphometric measurement of the area of fibrosis.45,46 One characteristic feature is the major thinning of fibrous septa, which may be reduced to a few collagen fibers. Associated with septa thinning is the disappearance of neovascular shunting. Indeed, thin septa in regressing cirrhosis usually lack vessels, or contain only few residual capillaries that may not function as shunting vessels anymore. Thereafter, septa become incomplete or may totally disappear.39 Regression of fibrosis is associated with a partial or full restoration of the lobular organization. However, nodular organization may partially persist despite disappearance of fibrous septa leading to a pattern that resembles vaguely to nodular regenerative hyperplasia. Whether perisinusoidal fibrosis and sinusoidal capillarization regress with the acqui-

169

sition of a normal sinusoidal phenotype remains unclear. Indeed, recent studies suggest that this pattern may regress more inconsistently. However, ductular proliferation, a feature associated with fibrosis progression, quickly disappears in regressing cirrhosis. In addition, though the metabolic zonation is usually lost in cirrhosis, reversion is associated with the normalization of the lobular enzymatic zonation.46

New Technologies for Assessing Fibrosis in Liver Biopsy In the field of fibrosis, liver biopsy can provide information that is more than a semiquantitative score. Indeed, to get more accuracy and objectivity in the quantification of fibrosis, pathologists have developed a more sophisticated approach (i.e., morphometry), which is based on image analysis. The principles are obtaining a digitized image from a histological slide through high-definition digital cameras, thresholding the digitized image according to greyscale values, segment the image into the region that contains objects of interest for analysis (e.g., fibrous tissue) and background tissue, and measuring the structure of interest using image analysis software. This approach provides a very high level of accuracy for fibrous tissue quantitation (collagen percentage area [CPA]), which has been shown to be clinically relevant.47–50 More sophisticated approaches include measurement of form factors (size, shape, or other more complex measurements). For this approach, fractal geometry is the technique of choice.51 The development of easy-to-use systems and high-performance algorithms has made morphometry more accessible and useful to pathologists. It should be noted that, even when using fully automated techniques, morphometry remains time consuming and is not recommended for routine practice. A major advantage of morphometry is to provide a finite quantitative scale, which is linear and much more accurate than those determined by semiquantitative scoring methods.52 This approach has been used in clinical trials in the context of chronic viral hepatitis and NAFLD.52–54 Interestingly, these studies have shown the nonlinearity relationship between CPA and semiquantitative stages of fibrosis, a further demonstration that a fibrosis score does not equate to amount of fibrosis.11,48,55 Immunohistochemistry, although not a new tool, is a potential complementary approach that may allow a more dynamic assessment of fibrosis. It can be used for the labeling of activated hepatic stellate cells (mainly with α-smooth muscle antibody) or capillarization and/or neoangiogenesis (with CD34) or ductular proliferation (with cytokeratin 7), which are markers potentially associated with the progression of fibrosis in CHC or NAFLD.46,56,57 These tools are simple, relatively cheap, and reproducible. Biopsy can also provide useful information on the texture of fibrosis, which might have clinical relevance. Indeed, nonlinear optical microscopy enables the observation of endogenous signals such as two-photon excitation fluorescence (TPEF) and second harmonic generation (SHG) in unstained samples and deep within the tissue.58 In short, although TPEF allows for the visualization of the liver background and lobular organization, the SHG displays the threedimensional (3D) architecture of fibrillar collagen, a Seminars in Liver Disease

Vol. 35

No. 2/2015

Downloaded by: University of Connecticut. Copyrighted material.

diseases.20,21,31 Most of these scoring systems are clinically relevant because they are able to predict prognosis in most CLD.32,33 Recently, and in addition to score of fibrosis, several histological subscoring systems of cirrhosis have been proposed. They are all based on the size of parenchymal nodules and the width of fibrous septa such as the newly developed Laennec scoring system dividing cirrhosis into three groups (4a, 4b, 4c), a subclassification that correlates well both with hepatic venous pressure gradient (HVPG) and clinical outcome.34–36 Histological scores have also some drawbacks that have to be considered by both clinicians and pathologists. Histological staging of fibrosis is a reductive approach to the evaluation of liver damage. Fibrosis is a complex biological process with different patterns, which are not always reflected in the staging systems. In addition, fibrosis is usually present in combination with other histopathological features (inflammation, liver cell necrosis, steatosis, iron overload) that can influence the rate of progression, and the evolution to cirrhosis. Therefore, scoring systems should complement, but not replace the complete descriptive report of the pathologist. An important issue is that scores are semiquantitative values. The consequence is that scores should not be handled statistically as linear values, but as categories with adapted statistical tests. Unfortunately, many studies including clinical trials have missed this point, which limits the value of their scientific conclusions.

Patel et al.

Diagnosis of Liver Fibrosis: Present and Future

Patel et al.

Table 2 Diagnostic performance of serum biomarkers of fibrosis for significant fibrosis (F  2) and cirrhosis (F4) in patients with chronic liver diseases Biomarkers

Year

Etiologies

Patients (n)

F2 (%)

FibroTest81

2001

HCV

339

77

F4 (%)

Cutoffs

AUROC

Se (%)

Sp (%)

CC (%)

80

>0.48

0.87

75

85

46

2002

HCV

476

26

6.9

0.81

30–94

51–95

45

APRI64

2003

HCV

270

50

0.5 >1.5 0.36

0.83

77

73

75

2004

HCV

194

45

0.4

0.82

35–65

85–96

NA

2004

Mixed

1021

40

0.102 NA

0.78 0.89

87 NA

51 NA

NA NA

Forns Index

17 MP3 ELF

65

83

12 66

2005

HCV

302

48

0.2 0.8

0.77

42–85

48–98

40–49

Hepascore84

2005

HCV

211

57

0.5 >0.84

0.82 0.89

63 71

89 89

92 NA

Fibrometer85

2005

Mixed

598

56

NA

0.89

80

84

82

0.1

0.85

80

70

NA

Hui Score69

2005

HBV

235

25

0.15 >0.5

0.79

37–88

50–88

49

Zeng Score70

GUCI

ViraHep-C

38 12

2005

HBV

372

58

8.7

0.77

40–98

28–90

35

71

2006

HCV

398

37

0.22 >0.55

0.83

51–90

54–90

52

72

50

2007

HCV

360

1.25  2.25

0.83

30–40

97–97

35

FIB-478

2007

HCV

847

17a

3.25

0.85

38–74

81–98

68

NFS75

2007

NAFLD

733

30a

0.676

0.82

43–77

97–97

68

2008

NAFLD

669

38

2

0.81





NA

2008

HCV

512

38

69,000 differentially methylated CpG sites between mild and advanced NAFLD disease. Pathway analysis implicated differential methylation profiles in genes regulating several processes such as fatty acid metabolism, stellate cell activation, tissue repair, and carcinogenesis.122 An important caveat is that miRNA or epigenetic biomarkers may provide differing results within the same individual depending on methods or sample type such as urine, circulating white cells, within microvesicles, or circulating free in serum or plasma. At this stage, the identification of biomarkers based on miRNA transcripts that are detectable in blood or urine, and provide viable clinical diagnostic or prognostic utility, are still awaited. Long noncoding RNA transcripts (> 200 nucleotides), genomic and proteomic profiling of circulating extracellular vesicles (such as microvesicles, membrane particles, exosomes or apoptotic bodies) implicated in disease pathogenesis and intercellular signaling, represent novel approaches that hold promise for refined “liquid biopsy” biomarker development in the future.115,123,124

Imaging Assessment of Fibrosis Currently Available Methods Imaging techniques, particularly ultrasound (US), have been used for many years for the workup of patients with CLD because of their ease of use and wide availability.125 A major advance has been the introduction for liver fibrosis staging of liver stiffness measurement by using a variety of US or magnetic resonance- (MR-) based techniques. Monodimensional ultrasound transient elastography (TE; FibroScan), was the first introduced technique in the early 2000s.126 Its principle relies on the measurement of the velocity of a low-frequency (50 Hz) elastic shear wave propagating through the liver, that is directly related to tissue stiffness, called the elastic modulus (expressed as E ¼ 3ρv2, where v is the shear velocity and ρ is the density of tissue, assumed to be Seminars in Liver Disease

Vol. 35

No. 2/2015

173

Downloaded by: University of Connecticut. Copyrighted material.

Diagnosis of Liver Fibrosis: Present and Future

Diagnosis of Liver Fibrosis: Present and Future

Patel et al.

Table 3 Diagnostic performance of transient elastography for significant fibrosis (F  2) and cirrhosis (F4) in chronic liver diseases Authors

Year

Etiologies

Patient

F  2 (n)

Castera et al132

2005

HCV

183

74

Ziol et al133

2005

HCV

251

65

F4 (%)

25 19 134

AUROC (kPa)

Se (%)

Sp (%)

7.1

0.83

67

89

12.5

0.95

87

91

8.6

0.79

56

91

14.6

0.87

86

96

7.3

0.92

84

87

2006

PBC-PSC

95

16

17.3

0.96

93

95

Ganne-Carrie et al135

2006

Mixed

775

15

14.6

0.95

79

95

Foucher et al136

2007

Mixed

354

13

17.6

0.96

77

97

137

2007

Mixed

200

7.9

0.86

72

84

11.9

0.90

91

89

8.3

0.93

85

91

14.0

0.96

78

98

7.8

0.91

83

82

14.8

0.98

94

92

6.6

0.86

88

74

Corpechot et al

Fraquelli et al

60

Cutoffs (%)

50 12

Coco et al

138

2007

HCV/HBV

228

62

2008

HCV

150

56

50 Arena et al139

19 140

2008

NAFLD

97

50

17.0

0.99

100

97

Nobili et al141

2008

NAFLD

50

24

7.4

0.99

100

92

Nguyen-Khac et al142

2008

ALD

103

75

7.8

0.91

80

91

Nahon et al143

2008

ALD

147

Marcellin et al144

2009

HBV

173

Yoneda et al

9

32

19.5

0.92

86

84

54

22.7

0.87

84

83

7.2

0.81

70

83

0.93

93

87

50 8

Chan et al

145

Wong et al146 Degos et al

147

2009

HBV

161

2010

NAFLD

246

2010

HCV/HBV

25

1307

Petta et al Gaia et al

148

149

0.93

98

75

0.84

79

76

10

10.3

0.95

92

88

5.2

0.75

90

32

14

12.9

0.90

72

89

7.25

0.79

69

70

7.0

0.80

76

80

10.5

0.94

78

96

5.2

0.82

97

35

14

12.9

0.93

77

90

7.2

0.87

74

88

8

11.0

0.93

75

90

7.8

0.86

84

79

57

2011

NAFLD

169

47

2011

NAFLD

72

46 12.5

Zarski et al

150

Cardoso et al

2012

151

2012

HCV HBV

382

47

202

42

152

2012

NAFLD

75

NA

Wong et al153

2012

NAFLD

193

45

Myers et al

NA

Afdhal et al

154

2015

HCV/HBV

560

a

12–13.4

a

7.0

41

HCV-HIV

11.0

22.3

0.88

80

91

7.0

0.83

79

64

13

10.3

0.89

81

83

8.4

0.73

58

75

14.8

12.8

0.90

76

85

67

Abbreviations: AUROC, area under ROC curve; HBV, chronic hepatitis B; HCV, chronic hepatitis C; NA, not available; NAFLD, nonalcoholic fatty liver disease; Se, sensitivity; Sp, specificity. a Validation cohort.

Seminars in Liver Disease

Vol. 35

No. 2/2015

Downloaded by: University of Connecticut. Copyrighted material.

174

Diagnosis of Liver Fibrosis: Present and Future

175

respective advantages and disadvantages are shown in ►Table 1. Finally, liver stiffness can be measured by means of MR elastography using a modified phase-contrast method to image the propagation characteristics of the shear wave in the liver.131 Elasticity is quantified by MR elastography (expressed in kPa) using a formula that determines the shear modulus, which is equivalent to one-third the Young’s modulus used with TE. The theoretical advantages of MR elastography include its ability to analyze almost the entire liver, and its applicability to patients with obesity or ascites. However, MR elastography is so far too costly and time-consuming to be used in routine practice (►Table 1).

Diagnostic Algorithms for Staging Liver Fibrosis Viral Hepatitis The high efficacy of new HCV treatments (with SVR > 90%) as well as the increased tolerability of IFN-free regimens has made exact staging of disease less important. The most important knowledge derived from staging fibrosis in HCV is identifying those patients with bridging fibrosis and especially cirrhosis who should be screened for esophageal varices and hepatocellular carcinoma (HCC). The results of TE in the diagnosis of significant fibrosis and cirrhosis are summarized in ►Table 3.132–154 The value of TE in the assessment of liver fibrosis has been initially suggested in patients with chronic hepatitis C.132,133 These results have since been confirmed by many other groups both in hepatitis C139,147,150,154,155 and B.138,144,145,147,156 Transient elastography more accurately detects cirrhosis (AUROC values, 0.87– 0.99) than significant fibrosis (AUROC values, 0.73–0.93) (►Table 3). Several meta-analyses have confirmed the better diagnostic performance of TE for cirrhosis than for fibrosis, with mean AUROC values of 0.94 and 0.84, respectively.157 In

Fig. 1 Confounders of liver stiffness measurement with transient elastography. (Adapted from Tapper et al130). ALT, alanine transaminase; BMI, Body Mass Index. Seminars in Liver Disease

Vol. 35

No. 2/2015

Downloaded by: University of Connecticut. Copyrighted material.

constant). The stiffer the tissue, the faster the shear wave propagates. Transient elastography measures liver stiffness in a volume that approximates a cylinder (10 mm wide, 40 mm long), 25 to 65 mm below skin surface. Its results are expressed in kilopascals (kPa), and range from 2.5 to 75 kPa, with normal value around 5 kPa.127 Accurate results require careful interpretation of data, based on at least 10 validated measurements, a success rate (the ratio of valid measurements to the total number of measurement) > 60%, and an interquartile range (IQR; reflects variations among measurements) < 30% of the median value (IQR/LSM  30%).128 Transient elastography is a fast (5 minutes), safe, and reproducible procedure that can be performed at the bedside with immediate results (point of care). For these reasons, TE is currently the most widely used and best validated technique for noninvasive assessment of liver fibrosis worldwide. However, its main limitation in clinical practice is its limited applicability (80%) mostly due to obesity, ascites, or limited operator experience.129 Apart from obesity, important liver stiffness measurement confounders include inflammation, cholestasis, congestion, and food intake. Thus, TE should be performed by an experienced operator (> 100 examinations) in fasting patients (for at least 6 hours) and taking into account ALT levels (< 100 IU) (►Fig. 1).130 More recently, 2D-elastography techniques incorporated into conventional US machines (sonoelastography), based on the measurement of the velocity of shear waves generated by mechanically exciting liver tissue by ultrasound pushes have been introduced. The first described was acoustic radiation force impulse imaging (ARFI; Acuson 2000 Virtual Touch Tissue Quantification, Siemens Healthcare) followed by ShearWave elastography (SWE; Aixplorer, Supersonic Imagine).125 ShearWave elastography results are measured in kPa, whereas ARFI results are measured in meters/second. Their

Patel et al.

Diagnosis of Liver Fibrosis: Present and Future

Patel et al.

a meta-analysis of 40 studies (32 papers and 8 abstracts), sensitivity and specificity values were 0.83 and 0.89 for patients with cirrhosis and 0.79 and 0.78 for patients with significant fibrosis.158 However, only nine studies (comprising 1,364 patients) had acceptable standards for liver biopsy and TE, which limit the conclusions. In a recent meta-analysis of 18 studies including 2,772 HBV patients,159 mean AUROC values for diagnosing cirrhosis and significant fibrosis were 0.93 and 0.86, respectively. However, we are still lacking a meta-analysis of data from individual patients. When compared, the performances of TE have been shown to be similar between patients with HBV and HCV.151 However, serum levels of aminotransferases should be taken into account in interpreting results from TE in patients with hepatitis B.160 To avoid the risk of false-positive results, some authors have proposed to adapt TE cutoffs based on levels of ALT,145 a strategy that might not apply to patients with fluctuating levels of ALT or hepatitis flares. Conversely, in hepatitis e antigen- (HBeAg-) negative patients with normal levels of ALT, TE could be used as an adjunct tool in the measurement of HBV DNA, to follow inactive carriers or better identify patients who require liver biopsy (those with ongoing disease activity or significant fibrosis, despite normal levels of ALT).156,161,162 Although performances of TE and serum biomarkers have been shown to be equivalent for diagnosing significant fibrosis in patients with chronic viral hepatitis,132,147 TE has been shown to outperform serum biomarkers for diagnosing cirrhosis.147 Also a strategy combining TE with serum biomarkers has been shown to increase diagnostic accuracy, particularly for significant fibrosis in chronic hepatitis C.132 In HCV treatment-naïve patients without comorbidities, TE combined with a serum biomarker can be used as first-line assessment of liver fibrosis, with liver biopsy performed only in case of unexplained discordance between TE and the biomarker.127 In HBV treatment-naïve patients, TE could be used as first-line for patients with levels of ALT < twofold the upper limit of normal and levels of HBV DNA > 20,000 IU/mL (for HBeAg-positive patients) or > 2,000 IU/mL (for HBeAgnegative patients). The benefit of combining TE with a serum biomarker remains to be demonstrated in hepatitis B. Similar to TE, ARFI more accurately detects cirrhosis (mean AUROC value: 0.91) than significant fibrosis (mean AUROC value: 0.84).163 In a meta-analysis164 including 13 studies (n ¼ 1,163 patients) comparing ARFI with TE, no significant difference were found between ARFI and TE: summary sensitivities and specificities for the diagnosis of significant fibrosis were 0.74 and 0.83 for ARFI and 0.78 and 0.84 for TE, respectively, and 0.87 and 0.87 for ARFI and 0.89 and 0.87 for TE for the diagnosis of cirrhosis, respectively. Preliminary studies comparing SWE to TE, suggest that it may perform better than TE for significant fibrosis.165–167 These results need to be further validated.

Nonalcoholic Fatty Liver Disease Transient elastography has also been investigated in NAFLD patients, but in a smaller number of studies.140,141,146,148,149,152,153,168 Like in viral hepatitis, Seminars in Liver Disease

Vol. 35

No. 2/2015

TE performances are better for cirrhosis than for significant fibrosis with AUROCs ranging from 0.94 to 0.99 and from 0.79 to 0.99, respectively (►Table 3). However, these results deserve several comments. First, these studies have been conducted in heterogeneous or peculiar populations such as Asian patients or children with low Body Mass Index (BMI; < 28 kg/m2); second, most of them are underpowered with small sample size (< 100 patients) and very few patients with cirrhosis; third, the scoring systems (Brunt or Kleiner) and endpoints (significant fibrosis or severe fibrosis) are heterogeneous. These differences among the studies are likely the explanation for the observed differences among proposed cutoffs for a given endpoint, known as the spectrum bias.169,170 Finally, it should be stressed that all these studies have been conducted in tertiary referral centers where the proportion of patients with severe fibrosis is higher than that in the general population, making it difficult to extrapolate the performance of TE in detecting cirrhosis in large populations. The major challenge for the use of TE in patients with NAFLD in clinical practice is the high rate of failure (no valid shot) or unreliable results (valid shots < 10, success rate < 60%, or IQR/LSM > 30%) using the regular (M) probe, ranging from 3.8 to 50%.93 Such a wide range could be explained by the differences in the definitions used (failure vs. unreliable results) as well as in the BMI of the studied populations (higher rates in the populations with higher BMI). The so-called XL probe has been proposed to overcome these limitations for overweight and obese patients. Myers et al152 have shown, in 276 patients with chronic liver disease (42% viral hepatitis, 46% NAFLD) and a BMI > 28 kg/m2, that failure was significantly less frequent with the XL probe than the M probe (1.1% vs. 16%; P < 0.00005). However, unreliable results were still observed with the XL probe in 25% of cases compared with 50% of cases with the M probe (P < 0.00005). In another series of 193 patients with NAFLD,153 the failure rate of XL probe was significantly lower than that of M probe (2% vs. 10%, respectively; p ¼ 0.002), but the unreliable result rate, although lower, did not differ significantly (25% vs. 33%, respectively; p ¼ 0.093). The overall diagnostic accuracy of M probe and XL probe was similar for significant fibrosis. However, by intention-to-diagnose the performance of M probe was unsatisfactory due to the high rate of unreliable results (AUROCs) compared with the XL probe. Among 155 patients with 10 valid measurements by both M probe and XL probe, 124 (80%) patients had lower liver stiffness values by XL probe than M probe. Pairwise examination showed that LSM by XL probe was 1.7  2.3 kPa lower than that by M probe. Altogether, these results indicate that the use of TE could be of interest to exclude severe fibrosis and cirrhosis in NAFLD patients with confidence, but the high rate of unreliable results remains the main challenge. The XL probe could be used as second line in the subset of patients (30%) in whom the regular M probe fails. However, appropriate cutoff values for the XL probe remain to be defined and validated. Algorithms combining TE with a serum biomarker remain to be validated.

Downloaded by: University of Connecticut. Copyrighted material.

176

Diagnosis of Liver Fibrosis: Present and Future As for ARFI, SWE, and MRE, results are too preliminary to draw any firm conclusions.

Other Liver Diseases Transient elastography has also been evaluated in cholestatic liver diseases,134,171 in a variety of chronic liver diseases135–137,172 as well as in ALD (►Table 3).142,143 For this latter condition, cutoffs for cirrhosis diagnosis have been reported to be higher than in viral hepatitis (> 20 kPa). However, it has been suggested by several groups that the presence of alcoholic hepatitis may lead to overestimation of liver stiffness results173–175 thus, TE should be performed after alcohol withdrawal to improve accuracy.

Patel et al.

177

1,457 HCV patients, liver stiffness values and FibroTest had the highest 5-year predictive values for predicting survival and liver-related death, which did not change after adjustment for treatment response, patient age, and estimates of necroinflammatory grade.108 Interestingly, Corpechot et al192 have shown in 168 patients with PSC that, not only those with high baseline but also those with increase in liver stiffness values (> 1.5 kPa/y) were at very high risk (10 times the risk estimated in the other group) of death, liver transplantation, or hepatic complications within a 4-year period. Thus, the potential of liver stiffness values for predicting clinical outcomes seems to be greater than that of liver biopsy, probably liver stiffness measures ongoing pathophysiological processes and functions that a biopsy cannot.

Prognostic Role Future Imaging Methods Among US-based elastography challenging TE, ARFI, and SWE are the most advanced and promising with performances at least similar or even better than TE.167 Their use is becoming more common in practice although their exact place remains to be defined. Compared with US-based elastography techniques, MR elastography has the advantage of analyzing a substantially larger liver volume, allowing a precise analysis of the viscoelastic properties of the liver via a full 3D assessment of the displacement of the wave. However, studies comparing MR elastography with TE have been limited so far and have shown conflicting results.194,195 In addition, MRelastography procedure remains too cumbersome and not standardized enough for widespread use in routine practice. Several novel imaging technologies, including perfusion, MR diffusion-weighted imaging, intravoxel incoherent motion or acoustic structure quantification, are being developed, but their use remains limited so far to research settings.196

Abbreviations 3D APRI ARFI AST AUROC BMI CHB CHC CPA CRS ECM ELF GMCSF GWAS HBV HCC HCV HVPG IL MBT MELD miRNA

three-dimensional AST to Platelet Ratio Index acoustic radiation force impulse imaging aspartate aminotransferase area under ROC curve Body Mass Index chronic hepatitis B chronic hepatitis C collagen percentage area Cirrhosis Risk Score extracellular matrix Enhanced Liver Fibrosis granulocyte monocyte colony stimulating factor genome-wide association studies hepatitis B virus hepatocellular carcinoma hepatitis C virus hepatic venous pressure gradient interleukin 13 C-Methacetin Breath Test Model for End-Stage Liver Disease microRNA Seminars in Liver Disease

Vol. 35

No. 2/2015

Downloaded by: University of Connecticut. Copyrighted material.

Liver stiffness can be used to identify patients with cirrhosis who are at risk of disease progression because it correlates with the severity of liver disease,136 portal pressure,176,177 which accurately predicts clinical events, as well as with the presence of esophageal varices. A recent meta-analysis found pooled positive and negative predictive values of 88% for the ability of TE to predict the presence of significant portal hypertension (HVPG > 10 mm Hg).178 However, the diagnostic accuracy of TE (specificity below 60%) is too low for identification of patients with esophageal varices in clinical practice.179 More recently, spleen stiffness has received much attention as a potential better predictor of portal hypertension than liver stiffness.180–183 However, results remain conflicting and ARFI or SWE might be more suited for the measurement of spleen stiffness with better applicability then TE.184–186 Finally, it has been recently suggested that spleen stiffness could predict the occurrence of complications.187 Further studies to better define the most appropriate technique to measure spleen stiffness as well as its limitations are needed before spleen stiffness can be used in clinical practice. Liver stiffness, measured using TE, can also be used in determining prognosis. Large, prospective cohort studies in Asia of patients with hepatitis B or C correlated liver stiffness values with HCC occurrence.188–190 Among 866 Japanese patients with HCV infection, the cumulative incidence of HCC within 3 years was as high as 38.5%, among patients with baseline liver stiffness values > 25 kPa, compared with 0.4% among patients with values  10 kPa.188 Although measurements of liver stiffness could be used to identify patients at risk of developing HCC, more data are needed before they could be integrated into an HCC surveillance program. Several recent studies have shown that in patients with chronic liver disease, liver stiffness could also predict clinical decompensation as well as survival.108,191–193 For instance, Robic et al191 found that TE was as effective as HVPG in predicting clinical decompensations in 100 patients with chronic liver disease with a 2-year follow-up. Both HVPG  10 mm Hg and liver stiffness  21.1 kPa had 100% NPV for portal-hypertensive complications. Similarly, in a cohort of 128 Korean patients with active HBV cirrhosis, liver stiffness at a cutoff of 19 kPa, had a hazard ratio of 7 for development of clinical decompensation.193 In a cohort of

Diagnosis of Liver Fibrosis: Present and Future MMP MR MRI NAFLD SHG SNP SWE TE TPEF US

Patel et al.

matrix metalloproteinase magnetic resonance magnetic resonance imaging nonalcoholic fatty liver disease second harmonic generation single-nucleotide polymorphism ShearWave elastography transient elastography two-photon excitation fluorescence ultrasound

16 Altamirano J, Miquel R, Katoonizadeh A, et al. A histologic scoring

17

18

19

20

References

21

1 European Association for Study of Liver. EASL Clinical Practice

2

3 4 5 6

7

8

9 10

11

12

13

14

15

Guidelines: management of hepatitis C virus infection. J Hepatol 2014;60(2):392–420 Chalasani N, Younossi Z, Lavine JE, et al; American Gastroenterological Association; American Association for the Study of Liver Diseases; American College of Gastroenterologyh. The diagnosis and management of non-alcoholic fatty liver disease: practice guideline by the American Gastroenterological Association, American Association for the Study of Liver Diseases, and American College of Gastroenterology. Gastroenterology 2012;142(7): 1592–1609 Bedossa P, Paradis V. Liver extracellular matrix in health and disease. J Pathol 2003;200(4):504–515 Mehta SH, Lau B, Afdhal NH, Thomas DL. Exceeding the limits of liver histology markers. J Hepatol 2009;50(1):36–41 Manning DS, Afdhal NH. Diagnosis and quantitation of fibrosis. Gastroenterology 2008;134(6):1670–1681 Wong JB, Koff RS. Watchful waiting with periodic liver biopsy versus immediate empirical therapy for histologically mild chronic hepatitis C. A cost-effectiveness analysis. Ann Intern Med 2000;133(9):665–675 Merriman RB, Ferrell LD, Patti MG, et al. Correlation of paired liver biopsies in morbidly obese patients with suspected nonalcoholic fatty liver disease. Hepatology 2006;44(4):874–880 Ratziu V, Charlotte F, Heurtier A, et al; LIDO Study Group. Sampling variability of liver biopsy in nonalcoholic fatty liver disease. Gastroenterology 2005;128(7):1898–1906 Bedossa P, Dargère D, Paradis V. Sampling variability of liver fibrosis in chronic hepatitis C. Hepatology 2003;38(6):1449–1457 Cholongitas E, Senzolo M, Standish R, et al. A systematic review of the quality of liver biopsy specimens. Am J Clin Pathol 2006; 125(5):710–721 Standish RA, Cholongitas E, Dhillon A, Burroughs AK, Dhillon AP. An appraisal of the histopathological assessment of liver fibrosis. Gut 2006;55(4):569–578 Schiano TD, Azeem S, Bodian CA, et al. Importance of specimen size in accurate needle liver biopsy evaluation of patients with chronic hepatitis C. Clin Gastroenterol Hepatol 2005;3(9):930–935 Colloredo G, Guido M, Sonzogni A, Leandro G. Impact of liver biopsy size on histological evaluation of chronic viral hepatitis: the smaller the sample, the milder the disease. J Hepatol 2003; 39(2):239–244 Regev A, Berho M, Jeffers LJ, et al. Sampling error and intraobserver variation in liver biopsy in patients with chronic HCV infection. Am J Gastroenterol 2002;97(10):2614–2618 Bedossa P; FLIP Pathology Consortium. Utility and appropriateness of the fatty liver inhibition of progression (FLIP) algorithm and steatosis, activity, and fibrosis (SAF) score in the evaluation of biopsies of nonalcoholic fatty liver disease. Hepatology 2014; 60(2):565–575

Seminars in Liver Disease

Vol. 35

No. 2/2015

22

23

24

25

26

27

28

29 30

31

32 33

34

35

36

37

system for prognosis of patients with alcoholic hepatitis. Gastroenterology 2014;146(5):1231–9.e1, 6 Venturi C, Sempoux C, Bueno J, et al. Novel histologic scoring system for long-term allograft fibrosis after liver transplantation in children. Am J Transplant 2012;12(11):2986–2996 Westin J, Lagging LM, Wejstål R, Norkrans G, Dhillon AP. Interobserver study of liver histopathology using the Ishak score in patients with chronic hepatitis C virus infection. Liver 1999;19(3):183–187 Younossi ZM, Gramlich T, Liu YC, et al. Nonalcoholic fatty liver disease: assessment of variability in pathologic interpretations. Mod Pathol 1998;11(6):560–565 Kleiner DE, Brunt EM, Van Natta M, et al; Nonalcoholic Steatohepatitis Clinical Research Network. Design and validation of a histological scoring system for nonalcoholic fatty liver disease. Hepatology 2005;41(6):1313–1321 Bedossa P, Poynard T; The METAVIR Cooperative Study Group. An algorithm for the grading of activity in chronic hepatitis C. Hepatology 1996;24(2):289–293 Baptista A, Bianchi L, De Groote J, et al. The diagnostic significance of periportal hepatic necrosis and inflammation. Histopathology 1988;12(6):569–579 Mariño Z, Mensa L, Crespo G, et al. Early periportal sinusoidal fibrosis is an accurate marker of accelerated HCV recurrence after liver transplantation. J Hepatol 2014;61(2):270–277 Cooksley WG, Bradbear RA, Robinson W, et al. The prognosis of chronic active hepatitis without cirrhosis in relation to bridging necrosis. Hepatology 1986;6(3):345–348 Anthony PP, Ishak KG, Nayak NC, Poulsen HE, Scheuer PJ, Sobin LH. The morphology of cirrhosis: definition, nomenclature, and classification. Bull World Health Organ 1977;55(4):521–540 Hytiroglou P, Snover DC, Alves V, et al. Beyond “cirrhosis”: a proposal from the International Liver Pathology Study Group. Am J Clin Pathol 2012;137(1):5–9 Garcia-Tsao G, Friedman S, Iredale J, Pinzani M. Now there are many (stages) where before there was one: in search of a pathophysiological classification of cirrhosis. Hepatology 2010; 51(4):1445–1449 Nakano M, Worner TM, Lieber CS. Perivenular fibrosis in alcoholic liver injury: ultrastructure and histologic progression. Gastroenterology 1982;83(4):777–785 Tiniakos DG. Liver biopsy in alcoholic and non-alcoholic steatohepatitis patients. Gastroenterol Clin Biol 2009;33(10-11):930–939 Harrison SA, Brunt EM, Goodman ZD, Di Bisceglie AM. Diabetic hepatosclerosis: diabetic microangiopathy of the liver. Arch Pathol Lab Med 2006;130(1):27–32 Knodell RG, Ishak KG, Black WC, et al. Formulation and application of a numerical scoring system for assessing histological activity in asymptomatic chronic active hepatitis. Hepatology 1981;1(5):431–435 Yano M, Kumada H, Kage M, et al. The long-term pathological evolution of chronic hepatitis C. Hepatology 1996;23(6):1334–1340 Younossi ZM, Stepanova M, Rafiq N, et al. Pathologic criteria for nonalcoholic steatohepatitis: interprotocol agreement and ability to predict liver-related mortality. Hepatology 2011;53(6):1874–1882 Rastogi A, Maiwall R, Bihari C, et al. Cirrhosis histology and Laennec staging system correlate with high portal pressure. Histopathology 2013;62(5):731–741 Kim MY, Cho MY, Baik SK, et al. Histological subclassification of cirrhosis using the Laennec fibrosis scoring system correlates with clinical stage and grade of portal hypertension. J Hepatol 2011;55(5):1004–1009 Nagula S, Jain D, Groszmann RJ, Garcia-Tsao G. Histologicalhemodynamic correlation in cirrhosis-a histological classification of the severity of cirrhosis. J Hepatol 2006;44(1):111–117 Friedman SL, Bansal MB. Reversal of hepatic fibrosis —fact or fantasy? Hepatology 2006;43(2, Suppl 1):S82–S88

Downloaded by: University of Connecticut. Copyrighted material.

178

Patel et al.

38 Pérez-Tamayo R. Cirrhosis of the liver: a reversible disease?

58 Tai DC, Tan N, Xu S, et al. Fibro-C-Index: comprehensive, mor-

Pathol Annu 1979;14(Pt 2):183–213 Wanless IR, Nakashima E, Sherman M. Regression of human cirrhosis. Morphologic features and the genesis of incomplete septal cirrhosis. Arch Pathol Lab Med 2000;124(11):1599–1607 Poynard T, McHutchison J, Manns M, et al. Impact of pegylated interferon alfa-2b and ribavirin on liver fibrosis in patients with chronic hepatitis C. Gastroenterology 2002;122(5):1303–1313 Chang TT, Liaw YF, Wu SS, et al. Long-term entecavir therapy results in the reversal of fibrosis/cirrhosis and continued histological improvement in patients with chronic hepatitis B. Hepatology 2010;52(3):886–893 Shiratori Y, Imazeki F, Moriyama M, et al. Histologic improvement of fibrosis in patients with hepatitis C who have sustained response to interferon therapy. Ann Intern Med 2000;132(7): 517–524 Asselah T, Marcellin P. Long-term results of treatment with nucleoside and nucleotide analogues (entecavir and tenofovir) for chronic hepatitis B. Clin Liver Dis 2013;17(3):445–450 Marcellin P, Chang TT, Lim SG, et al. Long-term efficacy and safety of adefovir dipivoxil for the treatment of hepatitis B e antigenpositive chronic hepatitis B. Hepatology 2008;48(3):750–758 Serpaggi J, Carnot F, Nalpas B, et al. Direct and indirect evidence for the reversibility of cirrhosis. Hum Pathol 2006;37(12): 1519–1526 D’Ambrosio R, Aghemo A, Rumi MG, et al. A morphometric and immunohistochemical study to assess the benefit of a sustained virological response in hepatitis C virus patients with cirrhosis. Hepatology 2012;56(2):532–543 Tsochatzis E, Bruno S, Isgro G, et al. Collagen proportionate area is superior to other histological methods for sub-classifying cirrhosis and determining prognosis. J Hepatol 2014;60(5):948–954 Calvaruso V, Burroughs AK, Standish R, et al. Computer-assisted image analysis of liver collagen: relationship to Ishak scoring and hepatic venous pressure gradient. Hepatology 2009;49(4): 1236–1244 Calvaruso V, Dhillon AP, Tsochatzis E, et al. Liver collagen proportionate area predicts decompensation in patients with recurrent hepatitis C virus cirrhosis after liver transplantation. J Gastroenterol Hepatol 2012;27(7):1227–1232 Manousou P, Burroughs AK, Tsochatzis E, et al. Digital image analysis of collagen assessment of progression of fibrosis in recurrent HCV after liver transplantation. J Hepatol 2013;58(5):962–968 Sandrini J, Boursier J, Chaigneau J, et al. Quantification of portalbridging fibrosis area more accurately reflects fibrosis stage and liver stiffness than whole fibrosis or perisinusoidal fibrosis areas in chronic hepatitis C. Mod Pathol 2014;27(7):1035–1045 Goodman ZD, Becker RL Jr, Pockros PJ, Afdhal NH. Progression of fibrosis in advanced chronic hepatitis C: evaluation by morphometric image analysis. Hepatology 2007;45(4):886–894 Goodman ZD, Stoddard AM, Bonkovsky HL, et al; HALT-C Trial Group. Fibrosis progression in chronic hepatitis C: morphometric image analysis in the HALT-C trial. Hepatology 2009;50(6):1738–1749 McHutchison J, Goodman Z, Patel K, et al; Farglitizar Study Investigators. Farglitazar lacks antifibrotic activity in patients with chronic hepatitis C infection. Gastroenterology 2010; 138(4):1365–1373, 1373.e1–1373.e2 Pilette C, Rousselet MC, Bedossa P, et al. Histopathological evaluation of liver fibrosis: quantitative image analysis vs semiquantitative scores. Comparison with serum markers. J Hepatol 1998;28(3):439–446 Gadd VL, Skoien R, Powell EE, et al. The portal inflammatory infiltrate and ductular reaction in human nonalcoholic fatty liver disease. Hepatology 2014;59(4):1393–1405 Hu DD, Habib S, Li XM, Wang TL, Wang BE, Zhao XY. Angiogenesis: a new surrogate histopathological marker is capable of differentiating between mild and significant portal hypertension. Histol Histopathol 2015;30(2):205–212

phology-based quantification of liver fibrosis using second harmonic generation and two-photon microscopy. J Biomed Opt 2009;14(4):044013 Chen AC, McNeilly C, Liu AP, et al. Second harmonic generation and multiphoton microscopic detection of collagen without the need for species specific antibodies. Burns 2011;37(6): 1001–1009 Gailhouste L, Le Grand Y, Odin C, et al. Fibrillar collagen scoring by second harmonic microscopy: a new tool in the assessment of liver fibrosis. J Hepatol 2010;52(3):398–406 Xu S, Wang Y, Tai DC, et al. qFibrosis: a fully-quantitative innovative method incorporating histological features to facilitate accurate fibrosis scoring in animal model and chronic hepatitis B patients. J Hepatol 2014;61(2):260–269 Friedman SL. Evolving challenges in hepatic fibrosis. Nat Rev Gastroenterol Hepatol 2010;7(8):425–436 Guha IN, Myers RP, Patel K, Talwalkar JA. Biomarkers of liver fibrosis: what lies beneath the receiver operating characteristic curve? Hepatology 2011;54(4):1454–1462 Wai CT, Greenson JK, Fontana RJ, et al. A simple noninvasive index can predict both significant fibrosis and cirrhosis in patients with chronic hepatitis C. Hepatology 2003;38(2):518–526 Leroy V, Monier F, Bottari S, et al. Circulating matrix metalloproteinases 1, 2, 9 and their inhibitors TIMP-1 and TIMP-2 as serum markers of liver fibrosis in patients with chronic hepatitis C: comparison with PIIINP and hyaluronic acid. Am J Gastroenterol 2004;99(2):271–279 Sud A, Hui JM, Farrell GC, et al. Improved prediction of fibrosis in chronic hepatitis C using measures of insulin resistance in a probability index. Hepatology 2004;39(5):1239–1247 Lok AS, Ghany MG, Goodman ZD, et al. Predicting cirrhosis in patients with hepatitis C based on standard laboratory tests: results of the HALT-C cohort. Hepatology 2005;42(2):282–292 Islam S, Antonsson L, Westin J, Lagging M. Cirrhosis in hepatitis C virus-infected patients can be excluded using an index of standard biochemical serum markers. Scand J Gastroenterol 2005; 40(7):867–872 Hui AY, Chan HL, Wong VW, et al. Identification of chronic hepatitis B patients without significant liver fibrosis by a simple noninvasive predictive model. Am J Gastroenterol 2005;100(3): 616–623 Zeng MD, Lu LG, Mao YM, et al. Prediction of significant fibrosis in HBeAg-positive patients with chronic hepatitis B by a noninvasive model. Hepatology 2005;42(6):1437–1445 Fontana RJ, Kleiner DE, Bilonick R, et al; Virahep-C Study Group. Modeling hepatic fibrosis in African American and Caucasian American patients with chronic hepatitis C virus infection. Hepatology 2006;44(4):925–935 Koda M, Matunaga Y, Kawakami M, Kishimoto Y, Suou T, Murawaki Y. FibroIndex, a practical index for predicting significant fibrosis in patients with chronic hepatitis C. Hepatology 2007; 45(2):297–306 Sterling RK, Lissen E, Clumeck N, et al; APRICOT Clinical Investigators. Development of a simple noninvasive index to predict significant fibrosis in patients with HIV/HCV coinfection. Hepatology 2006;43(6):1317–1325 Fontana RJ, Goodman ZD, Dienstag JL, et al; HALT-C Trial Group. Relationship of serum fibrosis markers with liver fibrosis stage and collagen content in patients with advanced chronic hepatitis C. Hepatology 2008;47(3):789–798 Angulo P, Hui JM, Marchesini G, et al. The NAFLD fibrosis score: a noninvasive system that identifies liver fibrosis in patients with NAFLD. Hepatology 2007;45(4):846–854 Harrison SA, Oliver D, Arnold HL, Gogia S, Neuschwander-Tetri BA. Development and validation of a simple NAFLD clinical scoring system for identifying patients without advanced disease. Gut 2008;57(10):1441–1447

39

40

41

42

43

44

45

46

47

48

49

50

51

52

53

54

55

56

57

59

60

61

62 63

64

65

66

67

68

69

70

71

72

73

74

75

76

Seminars in Liver Disease

Vol. 35

No. 2/2015

179

Downloaded by: University of Connecticut. Copyrighted material.

Diagnosis of Liver Fibrosis: Present and Future

Diagnosis of Liver Fibrosis: Present and Future

Patel et al.

77 Forns X, Ampurdanès S, Llovet JM, et al. Identification of chronic

97 Salkic NN, Jovanovic P, Hauser G, Brcic M. FibroTest/Fibrosure for

hepatitis C patients without hepatic fibrosis by a simple predictive model. Hepatology 2002;36(4 Pt 1):986–992 Vallet-Pichard A, Mallet V, Nalpas B, et al. FIB-4: an inexpensive and accurate marker of fibrosis in HCV infection. comparison with liver biopsy and fibrotest. Hepatology 2007;46(1):32–36 Lin ZH, Xin YN, Dong QJ, et al. Performance of the aspartate aminotransferase-to-platelet ratio index for the staging of hepatitis C-related fibrosis: an updated meta-analysis. Hepatology 2011;53(3):726–736 Amorim TG, Staub GJ, Lazzarotto C, et al. Validation and comparison of simple noninvasive models for the prediction of liver fibrosis in chronic hepatitis C. Ann Hepatol 2012;11(6): 855–861 Imbert-Bismut F, Ratziu V, Pieroni L, Charlotte F, Benhamou Y, Poynard T; MULTIVIRC Group. Biochemical markers of liver fibrosis in patients with hepatitis C virus infection: a prospective study. Lancet 2001;357(9262):1069–1075 Patel K, Gordon SC, Jacobson I, et al. Evaluation of a panel of noninvasive serum markers to differentiate mild from moderate-toadvanced liver fibrosis in chronic hepatitis C patients. J Hepatol 2004;41(6):935–942 Rosenberg WM, Voelker M, Thiel R, et al; European Liver Fibrosis Group. Serum markers detect the presence of liver fibrosis: a cohort study. Gastroenterology 2004;127(6):1704–1713 Adams LA, Bulsara M, Rossi E, et al. Hepascore: an accurate validated predictor of liver fibrosis in chronic hepatitis C infection. Clin Chem 2005;51(10):1867–1873 Calès P, Oberti F, Michalak S, et al. A novel panel of blood markers to assess the degree of liver fibrosis. Hepatology 2005;42(6): 1373–1381 Chou R, Wasson N. Blood tests to diagnose fibrosis or cirrhosis in patients with chronic hepatitis C virus infection. Ann Intern Med 2013;159(5):372 Sebastiani G, Halfon P, Castera L, et al. Comparison of three algorithms of non-invasive markers of fibrosis in chronic hepatitis C. Aliment Pharmacol Ther 2012;35(1):92–104 Boursier J, de Ledinghen V, Zarski JP, et al; multicentric groups from SNIFF 32, VINDIAG 7, and ANRS/HC/EP23 FIBROSTAR studies. Comparison of eight diagnostic algorithms for liver fibrosis in hepatitis C: new algorithms are more precise and entirely noninvasive. Hepatology 2012;55(1):58–67 Sebastiani G, Castera L, Halfon P, et al. The impact of liver disease aetiology and the stages of hepatic fibrosis on the performance of non-invasive fibrosis biomarkers: an international study of 2411 cases. Aliment Pharmacol Ther 2011;34(10):1202–1216 Musso G, Gambino R, Cassader M, Pagano G. Meta-analysis: natural history of non-alcoholic fatty liver disease (NAFLD) and diagnostic accuracy of non-invasive tests for liver disease severity. Ann Med 2011;43(8):617–649 Cusi K, Chang Z, Harrison S, et al. Limited value of plasma cytokeratin-18 as a biomarker for NASH and fibrosis in patients with non-alcoholic fatty liver disease. J Hepatol 2014;60(1): 167–174 Festi D, Schiumerini R, Marzi L, et al. Review article: the diagnosis of non-alcoholic fatty liver disease — availability and accuracy of noninvasive methods. Aliment Pharmacol Ther 2013;37(4):392–400 Castera L, Vilgrain V, Angulo P. Noninvasive evaluation of NAFLD. Nat Rev Gastroenterol Hepatol 2013;10(11):666–675 Shaheen AA, Myers RP. Systematic review and meta-analysis of the diagnostic accuracy of fibrosis marker panels in patients with HIV/hepatitis C coinfection. HIV Clin Trials 2008;9(1):43–51 Naveau S, Gaudé G, Asnacios A, et al. Diagnostic and prognostic values of noninvasive biomarkers of fibrosis in patients with alcoholic liver disease. Hepatology 2009;49(1):97–105 Castera L. Hepatitis B: are non-invasive markers of liver fibrosis reliable? Liver Int 2014;34(Suppl 1):91–96

significant liver fibrosis and cirrhosis in chronic hepatitis B: a meta-analysis. Am J Gastroenterol 2014;109(6):796–809 Mayo MJ, Parkes J, Adams-Huet B, et al. Prediction of clinical outcomes in primary biliary cirrhosis by serum enhanced liver fibrosis assay. Hepatology 2008;48(5):1549–1557 Dinesen L, Caspary WF, Chapman RW, Dietrich CF, Sarrazin C, Braden B. 13C-methacetin-breath test compared to also noninvasive biochemical blood tests in predicting hepatic fibrosis and cirrhosis in chronic hepatitis C. Dig Liver Dis 2008;40(9):743–748 Everson GT, Shiffman ML, Hoefs JC, et al; HALT-C Trial Group. Quantitative liver function tests improve the prediction of clinical outcomes in chronic hepatitis C: results from the Hepatitis C Antiviral Long-term Treatment Against Cirrhosis Trial. Hepatology 2012;55(4):1019–1029 Karsdal MA, Krarup H, Sand JM, et al. Review article: the efficacy of biomarkers in chronic fibroproliferative diseases - early diagnosis and prognosis, with liver fibrosis as an exemplar. Aliment Pharmacol Ther 2014;40(3):233–249 Poynard T, Lenaour G, Vaillant JC, et al. Liver biopsy analysis has a low level of performance for diagnosis of intermediate stages of fibrosis. Clin Gastroenterol Hepatol 2012;10(6):657–63.e7 Calès P, de Ledinghen V, Halfon P, et al. Evaluating the accuracy and increasing the reliable diagnosis rate of blood tests for liver fibrosis in chronic hepatitis C. Liver Int 2008;28(10):1352–1362 Patel K, Remlinger KS, Walker TG, et al. Multiplex protein analysis to determine fibrosis stage and progression in patients with chronic hepatitis C. Clin Gastroenterol Hepatol 2014;12(12): 2113–20.e1, 3 Nielsen MJ, Veidal SS, Karsdal MA, et al. Plasma Pro-C3 (N-terminal type III collagen propeptide) predicts fibrosis progression in patients with chronic hepatitis C. Liver Int 2015;35(2):429–437 Poynard T, Moussalli J, Munteanu M, et al; FibroFrance-GHPS group. Slow regression of liver fibrosis presumed by repeated biomarkers after virological cure in patients with chronic hepatitis C. J Hepatol 2013;59(4):675–683 Ngo Y, Munteanu M, Messous D, et al. A prospective analysis of the prognostic value of biomarkers (FibroTest) in patients with chronic hepatitis C. Clin Chem 2006;52(10):1887–1896 Vergniol J, Foucher J, Terrebonne E, et al. Noninvasive tests for fibrosis and liver stiffness predict 5-year outcomes of patients with chronic hepatitis C. Gastroenterology 2011;140(7): 1970–1979, 1979.e1–1979.e3 Poynard T, Vergniol J, Ngo Y, et al; FibroFrance Study Group; Epic3 Study Group; Bordeaux HCV Study Group. Staging chronic hepatitis C in seven categories using fibrosis biomarker (FibroTest™) and transient elastography (FibroScan®). J Hepatol 2014;60(4): 706–714 Parkes J, Roderick P, Harris S, et al. Enhanced liver fibrosis test can predict clinical outcomes in patients with chronic liver disease. Gut 2010;59(9):1245–1251 Vergniol J, Boursier J, Coutzac C, et al. Evolution of noninvasive tests of liver fibrosis is associated with prognosis in patients with chronic hepatitis C. Hepatology 2014;60(1):65–76 Ghany MG, Lok AS, Everhart JE, et al; HALT-C Trial Group. Predicting clinical and histologic outcomes based on standard laboratory tests in advanced chronic hepatitis C. Gastroenterology 2010;138(1):136–146 Kim D, Kim WR, Kim HJ, Therneau TM. Association between noninvasive fibrosis markers and mortality among adults with nonalcoholic fatty liver disease in the United States. Hepatology 2013;57(4):1357–1365 Angulo P, Bugianesi E, Bjornsson ES, et al. Simple noninvasive systems predict long-term outcomes of patients with nonalcoholic fatty liver disease. Gastroenterology 2013;145(4):782–9.e4 Schuppan D, Pinzani M. Anti-fibrotic therapy: lost in translation? J Hepatol 2012;56(Suppl 1):S66–S74

78

79

80

81

82

83

84

85

86

87

88

89

90

91

92

93 94

95

96

Seminars in Liver Disease

Vol. 35

No. 2/2015

98

99

100

101

102

103

104

105

106

107

108

109

110

111

112

113

114

115

Downloaded by: University of Connecticut. Copyrighted material.

180

Patel et al.

116 Trépo E, Potthoff A, Pradat P, et al. Role of a cirrhosis risk score for

139 Arena U, Vizzutti F, Abraldes JG, et al. Reliability of transient

the early prediction of fibrosis progression in hepatitis C patients with minimal liver disease. J Hepatol 2011;55(1):38–44 Estrabaud E, Vidaud M, Marcellin P, Asselah T. Genomics and HCV infection: progression of fibrosis and treatment response. J Hepatol 2012;57(5):1110–1125 Dongiovanni P, Donati B, Fares R, et al. PNPLA3 I148M polymorphism and progressive liver disease. World J Gastroenterol 2013; 19(41):6969–6978 Hannivoort RA, Hernandez-Gea V, Friedman SL. Genomics and proteomics in liver fibrosis and cirrhosis. Fibrogenesis Tissue Repair 2012;5(1):1 Szabo G, Bala S. MicroRNAs in liver disease. Nat Rev Gastroenterol Hepatol 2013;10(9):542–552 Murakami Y, Toyoda H, Tanahashi T, et al. Comprehensive miRNA expression analysis in peripheral blood can diagnose liver disease. PLoS ONE 2012;7(10):e48366 Murphy SK, Yang H, Moylan CA, et al. Relationship between methylome and transcriptome in patients with nonalcoholic fatty liver disease. Gastroenterology 2013;145(5):1076–1087 Lemoinne S, Thabut D, Housset C, et al. The emerging roles of microvesicles in liver diseases. Nat Rev Gastroenterol Hepatol 2014;11(6):350–361 Takahashi K, Yan I, Haga H, Patel T. Long noncoding RNA in liver diseases. Hepatology 2014;60(2):744–753 Berzigotti A, Castera L. Update on ultrasound imaging of liver fibrosis. J Hepatol 2013;59(1):180–182 Sandrin L, Fourquet B, Hasquenoph JM, et al. Transient elastography: a new noninvasive method for assessment of hepatic fibrosis. Ultrasound Med Biol 2003;29(12):1705–1713 Castera L. Noninvasive methods to assess liver disease in patients with hepatitis B or C. Gastroenterology 2012;142(6):1293–1302.e4 Castera L, Forns X, Alberti A. Non-invasive evaluation of liver fibrosis using transient elastography. J Hepatol 2008;48(5):835–847 Castéra L, Foucher J, Bernard PH, et al. Pitfalls of liver stiffness measurement: a 5-year prospective study of 13,369 examinations. Hepatology 2010;51(3):828–835 Tapper EB, Castera L, Afdhal NH. FibroScan (vibration-controlled transient elastography): where does it stand in the United States practice. Clin Gastroenterol Hepatol 2015;13(1):27–36 Muthupillai R, Lomas DJ, Rossman PJ, Greenleaf JF, Manduca A, Ehman RL. Magnetic resonance elastography by direct visualization of propagating acoustic strain waves. Science 1995; 269(5232):1854–1857 Castéra L, Vergniol J, Foucher J, et al. Prospective comparison of transient elastography, Fibrotest, APRI, and liver biopsy for the assessment of fibrosis in chronic hepatitis C. Gastroenterology 2005;128(2):343–350 Ziol M, Handra-Luca A, Kettaneh A, et al. Noninvasive assessment of liver fibrosis by measurement of stiffness in patients with chronic hepatitis C. Hepatology 2005;41(1):48–54 Corpechot C, El Naggar A, Poujol-Robert A, et al. Assessment of biliary fibrosis by transient elastography in patients with PBC and PSC. Hepatology 2006;43(5):1118–1124 Ganne-Carrié N, Ziol M, de Ledinghen V, et al. Accuracy of liver stiffness measurement for the diagnosis of cirrhosis in patients with chronic liver diseases. Hepatology 2006;44(6):1511–1517 Foucher J, Chanteloup E, Vergniol J, et al. Diagnosis of cirrhosis by transient elastography (FibroScan): a prospective study. Gut 2006;55(3):403–408 Fraquelli M, Rigamonti C, Casazza G, et al. Reproducibility of transient elastography in the evaluation of liver fibrosis in patients with chronic liver disease. Gut 2007;56(7):968–973 Coco B, Oliveri F, Maina AM, et al. Transient elastography: a new surrogate marker of liver fibrosis influenced by major changes of transaminases. J Viral Hepat 2007;14(5):360–369

elastography for the diagnosis of advanced fibrosis in chronic hepatitis C. Gut 2008;57(9):1288–1293 Yoneda M, Yoneda M, Mawatari H, et al. Noninvasive assessment of liver fibrosis by measurement of stiffness in patients with nonalcoholic fatty liver disease (NAFLD). Dig Liver Dis 2008; 40(5):371–378 Nobili V, Vizzutti F, Arena U, et al. Accuracy and reproducibility of transient elastography for the diagnosis of fibrosis in pediatric nonalcoholic steatohepatitis. Hepatology 2008;48(2):442–448 Nguyen-Khac E, Chatelain D, Tramier B, et al. Assessment of asymptomatic liver fibrosis in alcoholic patients using fibroscan: prospective comparison with seven non-invasive laboratory tests. Aliment Pharmacol Ther 2008;28(10):1188–1198 Nahon P, Kettaneh A, Tengher-Barna I, et al. Assessment of liver fibrosis using transient elastography in patients with alcoholic liver disease. J Hepatol 2008;49(6):1062–1068 Marcellin P, Ziol M, Bedossa P, et al. Non-invasive assessment of liver fibrosis by stiffness measurement in patients with chronic hepatitis B. Liver Int 2009;29(2):242–247 Chan HL, Wong GL, Choi PC, et al. Alanine aminotransferasebased algorithms of liver stiffness measurement by transient elastography (Fibroscan) for liver fibrosis in chronic hepatitis B. J Viral Hepat 2009;16(1):36–44 Wong VW, Vergniol J, Wong GL, et al. Diagnosis of fibrosis and cirrhosis using liver stiffness measurement in nonalcoholic fatty liver disease. Hepatology 2010;51(2):454–462 Degos F, Perez P, Roche B, et al; FIBROSTIC study group. Diagnostic accuracy of FibroScan and comparison to liver fibrosis biomarkers in chronic viral hepatitis: a multicenter prospective study (the FIBROSTIC study). J Hepatol 2010;53(6):1013–1021 Petta S, Di Marco V, Cammà C, Butera G, Cabibi D, Craxì A. Reliability of liver stiffness measurement in non-alcoholic fatty liver disease: the effects of body mass index. Aliment Pharmacol Ther 2011;33(12):1350–1360 Gaia S, Carenzi S, Barilli AL, et al. Reliability of transient elastography for the detection of fibrosis in non-alcoholic fatty liver disease and chronic viral hepatitis. J Hepatol 2011;54(1): 64–71 Zarski JP, Sturm N, Guechot J, et al; ANRS HCEP 23 Fibrostar Group. Comparison of nine blood tests and transient elastography for liver fibrosis in chronic hepatitis C: the ANRS HCEP-23 study. J Hepatol 2012;56(1):55–62 Cardoso AC, Carvalho-Filho RJ, Stern C, et al. Direct comparison of diagnostic performance of transient elastography in patients with chronic hepatitis B and chronic hepatitis C. Liver Int 2012; 32(4):612–621 Myers RP, Pomier-Layrargues G, Kirsch R, et al. Feasibility and diagnostic performance of the FibroScan XL probe for liver stiffness measurement in overweight and obese patients. Hepatology 2012;55(1):199–208 Wong VW, Vergniol J, Wong GL, et al. Liver stiffness measurement using XL probe in patients with nonalcoholic fatty liver disease. Am J Gastroenterol 2012;107(12):1862–1871 Afdhal NH, Bacon BR, Patel K, et al. Accuracy of Fibroscan, Compared with Histology, in Analysis of Liver Fibrosis in Patients With Hepatitis B or C: A United States Multicenter Study. Clin Gastroenterol Hepatol 2014 Lupşor M, Badea R, Stefănescu H, et al. Analysis of histopathological changes that influence liver stiffness in chronic hepatitis C. Results from a cohort of 324 patients. J Gastrointestin Liver Dis 2008;17(2):155–163 Oliveri F, Coco B, Ciccorossi P, et al. Liver stiffness in the hepatitis B virus carrier: a non-invasive marker of liver disease influenced by the pattern of transaminases. World J Gastroenterol 2008;14(40): 6154–6162

117

118

119

120 121

122

123

124 125 126

127 128 129

130

131

132

133

134

135

136

137

138

140

141

142

143

144

145

146

147

148

149

150

151

152

153

154

155

156

Seminars in Liver Disease

Vol. 35

No. 2/2015

181

Downloaded by: University of Connecticut. Copyrighted material.

Diagnosis of Liver Fibrosis: Present and Future

Diagnosis of Liver Fibrosis: Present and Future

Patel et al.

157 Friedrich-Rust M, Ong MF, Martens S, et al. Performance of

175 Trabut JB, Thépot V, Nalpas B, et al. Rapid decline of liver stiffness

transient elastography for the staging of liver fibrosis: a metaanalysis. Gastroenterology 2008;134(4):960–974 Tsochatzis EA, Gurusamy KS, Ntaoula S, Cholongitas E, Davidson BR, Burroughs AK. Elastography for the diagnosis of severity of fibrosis in chronic liver disease: a meta-analysis of diagnostic accuracy. J Hepatol 2011;54(4):650–659 Chon YE, Choi EH, Song KJ, et al. Performance of transient elastography for the staging of liver fibrosis in patients with chronic hepatitis B: a meta-analysis. PLoS ONE 2012;7(9): e44930 Fraquelli M, Rigamonti C, Casazza G, et al. Etiology-related determinants of liver stiffness values in chronic viral hepatitis B or C. J Hepatol 2011;54(4):621–628 Maimone S, Calvaruso V, Pleguezuelo M, et al. An evaluation of transient elastography in the discrimination of HBeAg-negative disease from inactive hepatitis B carriers. J Viral Hepat 2009; 16(11):769–774 Castéra L, Bernard PH, Le Bail B, et al. Transient elastography and biomarkers for liver fibrosis assessment and follow-up of inactive hepatitis B carriers. Aliment Pharmacol Ther 2011;33(4): 455–465 Nierhoff J, Chávez Ortiz AA, Herrmann E, Zeuzem S, FriedrichRust M. The efficiency of acoustic radiation force impulse imaging for the staging of liver fibrosis: a meta-analysis. Eur Radiol 2013; 23(11):3040–3053 Bota S, Herkner H, Sporea I, et al. Meta-analysis: ARFI elastography versus transient elastography for the evaluation of liver fibrosis. Liver Int 2013;33(8):1138–1147 Ferraioli G, Tinelli C, Dal Bello B, Zicchetti M, Filice G, Filice C; Liver Fibrosis Study Group. Accuracy of real-time shear wave elastography for assessing liver fibrosis in chronic hepatitis C: a pilot study. Hepatology 2012;56(6):2125–2133 Leung VY, Shen J, Wong VW, et al. Quantitative elastography of liver fibrosis and spleen stiffness in chronic hepatitis B carriers: comparison of shear-wave elastography and transient elastography with liver biopsy correlation. Radiology 2013;269(3): 910–918 Cassinotto C, Lapuyade B, Mouries A, et al. Non-invasive assessment of liver fibrosis with impulse elastography: comparison of Supersonic Shear Imaging with ARFI and FibroScan®. J Hepatol 2014;61(3):550–557 Lupsor M, Badea R, Stefanescu H, et al. Performance of unidimensional transient elastography in staging non-alcoholic steatohepatitis. J Gastrointestin Liver Dis 2010;19(1):53–60 Ransohoff DF, Feinstein AR. Problems of spectrum and bias in evaluating the efficacy of diagnostic tests. N Engl J Med 1978; 299(17):926–930 Poynard T, Halfon P, Castera L, et al; FibroPaca Group. Standardization of ROC curve areas for diagnostic evaluation of liver fibrosis markers based on prevalences of fibrosis stages. Clin Chem 2007;53(9):1615–1622 Corpechot C, Carrat F, Poujol-Robert A, et al. Noninvasive elastography-based assessment of liver fibrosis progression and prognosis in primary biliary cirrhosis. Hepatology 2012;56(1): 198–208 Gómez-Domínguez E, Mendoza J, Rubio S, Moreno-Monteagudo JA, García-Buey L, Moreno-Otero R. Transient elastography: a valid alternative to biopsy in patients with chronic liver disease. Aliment Pharmacol Ther 2006;24(3):513–518 Bardou-Jacquet E, Legros L, Soro D, et al. Effect of alcohol consumption on liver stiffness measured by transient elastography. World J Gastroenterol 2013;19(4):516–522 Mueller S, Millonig G, Sarovska L, et al. Increased liver stiffness in alcoholic liver disease: differentiating fibrosis from steatohepatitis. World J Gastroenterol 2010;16(8):966–972

following alcohol withdrawal in heavy drinkers. Alcohol Clin Exp Res 2012;36(8):1407–1411 Carrión JA, Navasa M, Bosch J, Bruguera M, Gilabert R, Forns X. Transient elastography for diagnosis of advanced fibrosis and portal hypertension in patients with hepatitis C recurrence after liver transplantation. Liver Transpl 2006;12(12): 1791–1798 Vizzutti F, Arena U, Romanelli RG, et al. Liver stiffness measurement predicts severe portal hypertension in patients with HCVrelated cirrhosis. Hepatology 2007;45(5):1290–1297 Shi KQ, Fan YC, Pan ZZ, et al. Transient elastography: a metaanalysis of diagnostic accuracy in evaluation of portal hypertension in chronic liver disease. Liver Int 2013;33(1):62–71 Castera L, Pinzani M, Bosch J. Non invasive evaluation of portal hypertension using transient elastography. J Hepatol 2012;56(3): 696–703 Calvaruso V, Bronte F, Conte E, Simone F, Craxì A, Di Marco V. Modified spleen stiffness measurement by transient elastography is associated with presence of large oesophageal varices in patients with compensated hepatitis C virus cirrhosis. J Viral Hepat 2013;20(12):867–874 Colecchia A, Montrone L, Scaioli E, et al. Measurement of spleen stiffness to evaluate portal hypertension and the presence of esophageal varices in patients with HCV-related cirrhosis. Gastroenterology 2012;143(3):646–654 Sharma P, Kirnake V, Tyagi P, et al. Spleen stiffness in patients with cirrhosis in predicting esophageal varices. Am J Gastroenterol 2013;108(7):1101–1107 Stefanescu H, Grigorescu M, Lupsor M, Procopet B, Maniu A, Badea R. Spleen stiffness measurement using Fibroscan for the noninvasive assessment of esophageal varices in liver cirrhosis patients. J Gastroenterol Hepatol 2011;26(1):164–170 Elkrief L, Rautou PE, Ronot M, et al. Prospective Comparison of Spleen and Liver Stiffness by Using Shear-Wave and Transient Elastography for Detection of Portal Hypertension in Cirrhosis. Radiology 2014:141210 Procopet B, Berzigotti A, Abraldes JG, et al. Real-time shear-wave elastography: Applicability, reliability and accuracy for clinically significant portal hypertension. J Hepatol 2014 Takuma Y, Nouso K, Morimoto Y, et al. Measurement of spleen stiffness by acoustic radiation force impulse imaging identifies cirrhotic patients with esophageal varices. Gastroenterology 2013;144(1):92–101.e2 Colecchia A, Colli A, Casazza G, et al. Spleen stiffness measurement can predict clinical complications in compensated HCV-related cirrhosis: a prospective study. J Hepatol 2014;60(6):1158–1164 Masuzaki R, Tateishi R, Yoshida H, et al. Prospective risk assessment for hepatocellular carcinoma development in patients with chronic hepatitis C by transient elastography. Hepatology 2009; 49(6):1954–1961 Fung J, Lai CL, Seto WK, Wong DK, Yuen MF. Prognostic significance of liver stiffness for hepatocellular carcinoma and mortality in HBeAgnegative chronic hepatitis B. J Viral Hepat 2011;18(10):738–744 Jung KS, Kim SU, Ahn SH, et al. Risk assessment of hepatitis B virusrelated hepatocellular carcinoma development using liver stiffness measurement (FibroScan). Hepatology 2011;53(3):885–894 Robic MA, Procopet B, Métivier S, et al. Liver stiffness accurately predicts portal hypertension related complications in patients with chronic liver disease: a prospective study. J Hepatol 2011; 55(5):1017–1024 Corpechot C, Gaouar F, El Naggar A, et al. Baseline values and changes in liver stiffness measured by transient elastography are associated with severity of fibrosis and outcomes of patients with primary sclerosing cholangitis. Gastroenterology 2014;146(4): 970–979, quiz e15–e16

158

159

160

161

162

163

164

165

166

167

168

169

170

171

172

173

174

Seminars in Liver Disease

Vol. 35

No. 2/2015

176

177

178

179

180

181

182

183

184

185

186

187

188

189

190

191

192

Downloaded by: University of Connecticut. Copyrighted material.

182

Diagnosis of Liver Fibrosis: Present and Future

Patel et al.

195 Huwart L, Sempoux C, Vicaut E, et al. Magnetic resonance

using fibroscan in chronic hepatitis B patients showing advanced liver fibrosis. PLoS ONE 2012;7(5):e36676 194 Bohte AE, de Niet A, Jansen L, et al. Non-invasive evaluation of liver fibrosis: a comparison of ultrasound-based transient elastography and MR elastography in patients with viral hepatitis B and C. Eur Radiol 2014;24(3):638–648

elastography for the noninvasive staging of liver fibrosis. Gastroenterology 2008;135(1):32–40 196 Van Beers BE, Daire JL, Garteiser P. New imaging techniques for liver diseases. J Hepatol 2015;62(3):690–700

Downloaded by: University of Connecticut. Copyrighted material.

193 Kim SU, Lee JH, Kim Y, et al. Prediction of liver-related events

183

Seminars in Liver Disease

Vol. 35

No. 2/2015

Diagnosis of liver fibrosis: present and future.

The diagnostic assessment of liver fibrosis, a major determinant of disease severity, is an important step in the management of patients with chronic ...
321KB Sizes 5 Downloads 9 Views