Review

Expert Review of Gastroenterology & Hepatology Downloaded from informahealthcare.com by Nanyang Technological University on 04/26/15 For personal use only.

Management and diagnosis of fatty liver disease Expert Rev. Gastroenterol. Hepatol. 9(5), 671–683 (2015)

Amanda Tamar Schneier*, Caitlin Colleen Citti and Douglas T Dieterich Department of Liver Diseases, Icahn School of Medicine at Mount Sinai,1 Gustave Levy Place, New York, NY 10029, USA *Author for correspondence: [email protected]

Nonalcoholic fatty liver disease is a common cause of chronic liver disease and has been an increasingly studied topic of research as the obesity epidemic has been growing. There is a significant morbidity and mortality with uncontrolled steatohepatitis, which can progress to fibrosis, cirrhosis and hepatocellular carcinoma. The prevalence of this disease has been estimated to be roughly one-third of the western population, thought to be largely due to diet and sedentary lifestyle. Several treatments have been studied including vitamin E, insulin-sensitizing agents and ursodeoxycholic acid; however, the only treatment shown to improve the histologic changes of nonalcoholic fatty liver disease is weight loss. Given the proven benefit of weight loss, there may be reason to screen at-risk populations; however, limited availability of other disease-modifying treatments may limit the cost–benefit ratios. A better understanding of the diagnosis and management of this condition is required to alter the course of this modifiable disease. KEYWORDS: fatty liver disease . NAFLD . NASH . review . steatohepatitis

Definitions

Nonalcoholic fatty liver disease (NAFLD) refers to a spectrum of liver diseases whose hallmark, fat accumulation (or steatosis), cannot be attributed to secondary causes such as alcohol or certain drugs. NAFLD includes steatosis, steatohepatitis and cirrhosis. Nonalcoholic fatty liver (NAFL) refers to evidence of macrovesicular hepatic steatosis without signs hepatocellular injury such as hepatocyte ballooning or fibrosis. The term nonalcoholic steatohepatitis (NASH) denotes the presence hepatic inflammation and hepatocyte injury (seen as ballooning) with or without the presence of fibrosis. It is worthy of note that this pattern of inflammation can be histologically indistinguishable from alcoholic steatohepatitis. Cryptogenic cirrhosis (CC) refers to cirrhosis without evidence of etiology. In many of these patients, the evidence of NASH such as ballooning and inflammation, while at one time present, can no longer be seen; thus, the term ‘burned-out steatohepatitis’ is also sometimes used [1–3]. TABLE 1 summarizes the above terminology. Epidemiology

First described in 1980 [4], NAFLD has become the leading cause of chronic liver informahealthcare.com

10.1586/17474124.2015.1003209

disease in western developed countries, and is a growing cause of liver disease worldwide [5,6]. The numbers are likely even higher, as many patients with NAFLD are clinically silent and often have normal liver tests [7,8]. Prevalence

In recent studies looking at developed countries in Europe, the USA and Australia, the prevalence of NAFLD in the general population has been reported to be approximately 30% [8–10]. One study by Younossi et al. reported that the prevalence of chronic liver disease increased from 11.78 to 14.78% between 1988 and 2008 [5]. The study revealed that while viral and alcoholic etiologies remained relatively stable, the prevalence of NAFLD (defined in this case by elevated liver transaminases without any other explanation) doubled from 5.51 to 11.01%. Thus, the proportion of chronic liver disease due to NAFLD also increased from 46.8 to 75.1%, as shown in FIGURE 1. Another prospective study conducted in 2010 found that on ultrasound, the prevalence of NAFLD in adults aged 18– 70 was 46% [11]. The discrepancy between these two studies may be in the fact that imaging is a much more sensitive modality for diagnosing fatty liver disease that would otherwise be clinically silent.

 2015 Informa UK Ltd

ISSN 1747-4124

671

Review

Schneier, Citti & Dieterich

Expert Review of Gastroenterology & Hepatology Downloaded from informahealthcare.com by Nanyang Technological University on 04/26/15 For personal use only.

Table 1. Defining conditions within the spectrum of non-alcoholic liver disease. NAFLD

Encompasses all forms of liver disease in patients without significant alcohol consumption, including simple steatosis, steatohepatitis and cirrhosis

NAFL

Evidence of hepatic steatosis without signs of hepatocellular injury such as hepatocyte ballooning or fibrosis

NASH

The presence of hepatic inflammation which may be histologically indistinguishable from alcoholic steatohepatitis with inflammation and hepatocyte injury (ballooning) ± fibrosis

deemed to be good surgical candidates. There are also studies in high-risk groups that can shed light on the prevalence of NASH. One such study looked at tissue obtained from morbidly obese patients undergoing bariatric surgery and found 35% prevalence of biopsy-proven NASH in this population [17]. Demographics

There is inconsistent data regarding gender and NAFLD. Most data have pointed to male predominance [7,15,18,19]; however, there is evidence that women make up a NAFL: Nonalcoholic fatty liver; NAFLD: Nonalcoholic fatty liver disease; NASH: Nonalcoholic steatohepatitis. larger proportion of NAFLD patients who are classified as lean [20]. NAFLD The prevalence of NAFLD in Asia appears to be as high as has been reported in all age groups, but most patients tend to in the west, that is, up to 30% [12–14]; however, it is more fre- be middle-aged [21]. quently seen in patients of lesser weight than in the west. But In the USA, there have been several studies examining the the definition of obesity in Asia has been changed to a BMI role that ethnicity plays in developing NAFLD. They have >25 instead of 30, based on anthropomorphic standards. There shown that Hispanic groups have the highest prevalence of is little data on NAFLD in South American, African and Mid- NAFLD at 21.2–58.3% [7,11–24] and also have the highest rates dle Eastern populations, but as the developing nations adopt of NASH, followed by non-Hispanic whites having an incidence western eating habits and have more access to diagnostic test- of 12.5–44.4% and African Americans having the lowest rates at ing, the documented prevalence will likely increase. 11.6–35.1% [20], as shown in FIGURE 2. As these were populationbased studies, the diagnostic criteria were based on imaging and Nonalcoholic steatohepatitis liver enzymes, and not on biopsy. There are several theories to Because NASH can only be diagnosed accurately on biopsy, explain why these differences occur, including the variation in there are few population-based studies that report the preva- traditional diet [25] and distribution of abdominal fat [18,26–28], lence of this more severe form of NAFLD. The prospective but the most likely determinants are genetic factors, such as carcohort study by Williams et al. did take biopsies of patients in rying the rs738409 G-allele of the patatin-like phospholipase a general population who had evidence of fatty liver on ultra- domain-containing protein 3 gene, or the PNPLA3 gene (also sound, and found NASH in 12.2% of the overall cohort and known as adiponutrin), which appears to modify the accumulain 29.9% of those with positive imaging [11]. Two other studies tion of hepatic triacylglycerol [29–31]. The link between NAFLD looked at biopsies from orthotopic liver donation candidates and insulin resistance has been extensively studied; however, in and found the prevalence to be 1.1–2.2% [15,16], although this terms of ethnicity, the data are inconsistent: while African Amermay be a low estimate as this population consisted of patients icans and Hispanics both have high rates of insulin resistance, African Americans actually have much lower prevalence of NAFLD [32,33], again supporting a genetic hypothesis. 16.00% Cryptogenic cirrhosis

14.00%

Cirrhosis without evidence of etiology. Many patients with ‘burnt-out’ steatohepatitis fall into this category as they no longer have signs of ballooning and inflammation

Chronic Liver disease NAFLD

Association with the metabolic syndrome

12.00% 10.00% 8.00% 6.00% 4.00% 2.00% 0.00%

1988

2008

Figure 1. Proportion of NAFLD as the etiology of chronic liver disease in the USA based on National Health and Nutrition Examination Survey data comparing 1988 and 2008 [5]. NAFLD: Nonalcoholic fatty liver disease.

672

Risk factors for NAFLD include age, ethnicity, BMI, diabetes or insulin resistance, hypertension, dyslipidemia and visceral fat [4,34–37]. These risk factors are nearly identical to those of the metabolic syndrome, and many consider NAFLD to be a surrogate for the syndrome. For instance, Marchesini et al. reported that among patients with NAFLD but without diabetes, metabolic syndrome was diagnosed in 67% of NAFL patients and in 88% of patients with biopsy-proven NASH [34]. The link between NAFLD and metabolic syndrome has implications toward both mortality and treatment. Pathogenesis

The pathological processes in NAFLD can be thought of in the multi-hit hypothesis framework. A susceptible individual Expert Rev. Gastroenterol. Hepatol. 9(5), (2015)

Expert Review of Gastroenterology & Hepatology Downloaded from informahealthcare.com by Nanyang Technological University on 04/26/15 For personal use only.

Management & diagnosis of fatty liver disease

may be faced with environmental factors (diet, activity, gut flora) that then lead to a combination of pathologic processes. When discussing the pathogenesis of NAFLD, two processes have to be considered: the accumulation of intrahepatic fat, or steatosis, and the hepatotoxic process that leads to inflammation and fibrosis.

70

Steatosis

30

The accumulation of triglycerides in the liver causes hepatic steatosis. This can occur with increased supply of free fatty acids (FFAs) from the diet, de novo synthesis in the liver or increased delivery from the peripheral tissues. A link also exists between insulin resistance and the development of steatosis: insulin resistance in the periphery leads to lipolysis by adipose tissue, releasing FFAs to be delivered to the liver [38]. Also, insulin resistance can increase de novo synthesis of FFAs in the liver by upregulating the enzymes that catalyze lipogenesis. Normally, insulin also inhibits gluconeogenesis simultaneously [39]. With hepatic insulin resistance, there is a paradoxical effect in which the stimulatory effect of insulin on lipogenesis is retained but the inhibitory effect of insulin on glucose production in the liver is diminished and FFA accumulates, a phenomenon called selective insulin resistance [40]. Studies have shown that even without overt diabetes, NAFLD patients have decreased hepatic response to insulin [41]. Thus, in response to normal insulin signaling, these patients produce less glucose and accumulate more FFAs, and can acquire hepatic steatosis at a faster rate than their peers. In addition to increased production, decreased export of FFAs (via the transport lipid VLDL) can lead to steatosis. When synthesis of VLDL or its component protein apolipoprotein B is impaired (seen in protein deficiency), FFAs remain in the hepatocytes, leading to steatosis. There is evidence that patients with NASH may also have defects in the production of postprandial apolipoprotein B [42]. This may help explain how some lean individuals develop steatosis without increased dietary intake or insulin resistance. Inflammation & damage

There are several mechanisms that have been proposed to explain the inflammation seen in more advanced forms of NAFLD. First, FFAs can themselves cause hepatotoxic free oxygen radical species by inducing microsomal lipoxygenases, or during beta-oxidation [43,44]. Defects in mitochondria have been proposed as a cause of increased free oxygen radicals during beta-oxidation, and electron microscopy has shown mitochondrial defects in patients with NASH but not in NAFL controls [45]. Reactive oxidative species lead to cell injury, apoptosis and necrosis, and are an important driver of inflammation and fibrosis in liver disease. A second mechanism that likely leads to liver inflammation in NAFLD patients is the interplay between the gut flora and the liver. There is evidence of increased gut permeability in patients with NASH and NASH risk factors [46–48]. The theoretical mechanism suggests that endotoxin may enter the portal informahealthcare.com

Review

60 50 40

20 10 0 White

Black

Hispanic

Figure 2. Comparing the prevalence of NAFLD across ethnicities as documented by percentage in population-based studies [7,11,18,20,22–24]. NAFLD: Nonalcoholic fatty liver disease.

blood, and induce hepatic injury via TNF-a [49,50]. Another area of research looks at the role of adiponectin, an adipokine that blocks the release of TNF-a, thus having antiinflammatory and anti-fibrotic effects on adipose and hepatic tissue [51–54]. An inverse relationship between NASH severity and adiponectin has been demonstrated [51,55], and its suppressive role in inflammation may be a key link in the progression from steatosis to steatohepatitis. Finally, recent studies have suggested that just as the case with other complications of the metabolic syndrome, endothelial damage within the liver and vascular dysfunction precedes the inflammation and fibrosis seen in NASH [56,57]. The inflammatory process in NAFLD and other liver diseases appears to be mediated mostly by NF-kB [58], natural killer T cells [59], the hedgehog pathway [60,61] and the activation of cytokines such as TNF-a [62]. Other players, such as myeloperoxidase [63], and activation of the complement system [64] have also been identified. A full discussion of the immune response from hepatic inflammation to liver fibrosis and cirrhosis is out of the scope of this review and can be found elsewhere [65–67]. Natural history

Simple steatosis (NAFL) and NASH behave differently. While there is evidence that without intervention simple steatosis can be progressive [68], NASH appears to have a more aggressive course. One prospective hospital cohort study looked at paired biopsies taken 36 months apart [69]. Of the 13 patients with steatosis at baseline, 23% continued to have simple steatosis while 15% regained normal histology; 39% progressed to borderline NASH and 23% developed clear NASH. Of the 17 patients with baseline NASH, however, only 6% returned to simple steatosis, indicating there may be a physiological ‘point of no return’. The risk of progression to liver fibrosis and cirrhosis appears to be relatively low in those with simple steatosis, compared to 673

Expert Review of Gastroenterology & Hepatology Downloaded from informahealthcare.com by Nanyang Technological University on 04/26/15 For personal use only.

Review

Schneier, Citti & Dieterich

those with steatohepatitis [70,71]. A systematic review of 10 studies found that of 187 patients with NAFLD on biopsy, 17% of those with no evidence of inflammation went on to develop severe fibrosis on repeat biopsy, compared to 49% of those with initial inflammation [72]. Moreover, the median of time to the development of advanced fibrosis was 13.4 years for those with initial simple steatosis, compared to 4.2 years for those with inflammation. Additional paired biopsy studies have shown that up to 53% of patients with proven NASH will show significant progression within 7 years [71,73,74]. Finally, a recent systematic review by Singh et al. was published looking at 411 patients with biopsy-proven NAFLD from 11 cohort paired-biopsy studies [75]. It found that the annual fibrosis progression rate in NASH patients was double that of patients with an initial fibrosis level of 0 (0.14 stages/year compared to 0.07). Put another way, it would take an NAFL patient 14.3 years to progress to the next stage of fibrosis, compared to 7.1 years for patients with NASH. The mutation of the PNPLA3 gene mentioned above has been shown to affect not only hepatic fat content but also the rate of progression to fibrosis. A study by Valenti et al. found that the mutated allele was not only disproportionately expressed in patients who developed NALFD but also was strongly associated with NASH (odds ratio [OR]: 1.5) and fibrosis stage >1 (OR: 1.5), independent of other known risk factors [76]. Such other risk factors for progression appear to be age, elevated liver transaminases, diabetes, BMI and visceral fat [72,77–80]. Mortality

Though simple steatosis has been shown to progress to fibrosis, the mortality remains roughly the same as in the general population [81–83]. NASH, however, has been shown to have an increased liver-related mortality rate [84,85]. One study following 132 NAFLD patients found that after 8 years of follow-up, 2% of the non-inflammatory NAFLD developed liver-related mortality, compared to 11% of the NASH cohort [81]. After 18.5 years of follow-up in the same cohort, liver-related mortality had risen to 18% for NASH and 3% for non-NASH patients [86]. Predictably, the degree of liver damage has the most effect on mortality. An international, multicenter prospective study showed that among patients with Childs–Pugh Class A disease followed over a mean period of 85.6 months, 12.3% of NASH patients required transplant or died, which, when corrected for age and gender, was statistically similar to patients with untreated Class A hepatitis C [87]. It is important to note that as the metabolic syndrome commonly affects NAFLD patients, the leading cause of death in these patients is cardiovascular disease [84,86,88]. The study by Ekstedt et al. found that mortality from cardiovascular events was double that of a matched reference population, at 15.5% compared to 7.5%, which could be explained by the shared risk factors as the metabolic syndrome [84]. There is also evidence that NAFLD is associated with an increased risk of incident cardiovascular disease even after controlling for 674

components of the metabolic syndrome and other risk factors [87,89,90]. It appears that even following liver transplantation, NASH patients remained at increased risk for cardiovascular events independent of other traditional risk factors [91]. NAFLD & hepatocellular carcinoma

Hepatocellular carcinoma (HCC) is the most common primary liver cancer and the fifth most common malignancy worldwide [92]. Cirrhosis is present in approximately 80% of patients with HCC, being the largest risk factor for the malignancy regardless of its etiology [93]. An estimated 30–40% of HCC cases are not classified under hepatitis C virus (HCV), hepatitis B virus (HBV) or heavy ethanol intake [92]. Of these, NAFLD is assumed to be a large component, though it is often missed as the steatosis and necro-inflammation of NASH ‘burn out’ or fade with the scarring of cirrhosis by the time HCC is diagnosed [35]. Because NAFLD often has an indolent course, there are few prospective studies directly examining the progression from NAFLD to HCC, as they require long periods of follow-up. Case reports of progression from NAFL to HCC exist [94], and there are numerous case reports [95–100] describing the development of HCC several years after the NASH is diagnosed. One cohort study at the higher end of reported prevalence showed that among 420 NAFLD patients followed over a mean of 7.6 years, only 2 progressed to HCC, making the prevalence 0.5% (however, this represented 10% of the 21 patients with cirrhosis) [21]. A second, larger study showed that of 817 NAFLD patients followed over 8.4 years, the incidence and prevalence of HCC was 0% [101]. The above studies included mostly patients with NAFL and few cases of biopsyproven NASH. In regards to NASH, the risk of HCC is higher. Two studies, by Ekstedt et al. and Rafiq et al. [84,86], also specifically looked at patients with documented NASH, showing the prevalence of HCC in these patients to be 0–2.8%. Another prospective study followed known cirrhotics with various etiologies including NASH (either on histology or by lack of any other etiology + 3 known risk factors) over a mean period of 6.8 years and found the annual rate of development of HCC to be 0.46%, compared to 1.5% for HBV and 3.6% for HCV [102]. As fibrosis advances, so does the risk for HCC. A fourth study, looking at NASH patients with evident cirrhosis, found that over a median follow-up of 3.2 years, 12.8% developed HCC with a yearly cumulative incidence of 2.6% (compared to 4.0% in HCV cirrhotics) [103]. Interestingly, the PNPLA3 gene may also be implicated in the risk of progression to HCC. In a recent study by Liu et al., the mutated allele was identified in 100 patients with NAFLDrelated HCC and 275 controls with histology-proven NAFLD [31]. They found that when controlled for age, gender, diabetes, BMI and cirrhosis, carriage of each copy of the mutated allele conferred an additive risk for HCC (adjusted OR: 2.26; 95% CI: 1.23–4.14; p = 0.0082), with mutation homozygotes exhibiting a fivefold increased risk over the wild type. Expert Rev. Gastroenterol. Hepatol. 9(5), (2015)

Management & diagnosis of fatty liver disease

Review

Table 2. Secondary causes of hepatic steatosis characterized by size of cytoplasmic vacuoles. Macrovesicular steatosis

Environmental

Hereditary

.

Excessive alcohol consumption Hepatitis C Starvation Parenteral nutrition Medications (amiodarone, methotrexate, tamoxifen, corticosteroids)

.

Reye’s syndrome Medications (valproate, anti-retroviral medications)

.

. .

Expert Review of Gastroenterology & Hepatology Downloaded from informahealthcare.com by Nanyang Technological University on 04/26/15 For personal use only.

. .

Microvesicular steatosis

. .

. .

. .

Often the physical evidence of NAFLD disappears by the time of HCC diagnosis, and many such cases have thus fallen under the term cryptogenic cirrhosis (CC). Because of this, the most robust evidence for an association between NASH and HCC is indirect, inferred from either retrospective studies connecting HCC risk with obesity and diabetes, the two conditions most strongly associated with NASH [104–107], or by evaluating the connection between HCC and CC, then selecting a subgroup of patients with NAFLD based on patient demographics [108–111]. One retrospective cohort study showed that CC accounted for the second-most common etiology of HCC after HCV, at 29% [108]. Of these CC patients, half had a prior histologic diagnosis of NASH or clinical diagnosis of NAFLD before the diagnosis of HCC, implying NAFLD was the underlying etiology in up to 13% of the patients with HCC evaluated. A systematic review has tried to pool data to give a clear sense of the incidence of HCC in the NAFLD population [112]. It looked at 17 cohort studies, 18 case–control and crosssectional studies and 26 case series, with the inclusion criteria of patients with NAFLD, NASH and CC suspected to be NASH. It found that cohorts with NASH and defined cirrhosis had a significant but inconsistent risk, with cumulative incidence ranging from 2.4% over 7 years to 12.8% over 3 years. Cohorts defined as NAFL or NASH without cirrhosis, however, had a minimal risk for HCC, with cumulative HCC mortality 0–3% for up to 20 years of follow-up. Screening

Despite the prevalence of NAFLD being close to 30% in the US population, the role of screening is controversial. Given the usually benign course of the condition, it would not be costeffective to screen even those deemed to be at high risk, such as patients with obesity, Type 2 diabetes or metabolic syndrome. Also, how to screen is also debatable: imaging is more sensitive, but is expensive and less specific, while liver enzymes may not reveal enough cases as often transaminases can be normal. Currently, the American Association for the Study of Liver Diseases (AASLD) does not recommend screening, even in families with high rates of the disease [113]. informahealthcare.com

Wilson’s disease Congenital lipodystrophy Abetalipoproteinemia

Inborn errors of metabolism (lecithin cholesterol acyltransferase (LCAT) deficiency, cholesterol ester storage disease, Wolman disease) Acute fatty liver of pregnancy HELLP syndrome

Diagnosis

Identification of at-risk individuals has been an area of great interest in the recent past. As the presence of metabolic syndrome is highly correlated with NAFLD, it should prompt further noninvasive diagnostic evaluation in patients with abnormal liver chemistries. Laboratory abnormalities such as elevated transaminases, though frequently seen with liver disease, are insufficient to make the diagnosis for NAFLD alone. Of importance, one must exclude alternative causes of liver disease or steatosis, including alcoholic liver disease [113]. Differentiated by the size of cytoplasmic vacuoles, examples of secondary causes of steatosis can be seen in TABLE 2. Alternative etiologies of liver disease include significant alcohol use, starvation, medication use, pregnancy, autoimmune hepatitis, hemochromatosis, and infections such as hepatitis A, B or C. If the clinical scenario lends itself to alternative diagnosis, further testing can be pursued to rule out Wilson’s disease, thyroid disorders, celiac disease, a-1 antitrypsin deficiency, HELLP Syndrome and Budd–Chiari syndrome. Imaging

Most often, the diagnosis of NAFLD is made radiographically with ultrasound, computed tomography (CT) or MRI. Findings consistent with fatty infiltration of the liver, without signs and symptoms of cirrhosis, are consistent with NAFL. Ultrasonographic findings include hyperechoic texture, or brightness of the hepatic parenchyma. Although biopsy remains the gold standard for diagnosis, a meta-analysis conducted in 2011 of 49 studies reported that ultrasound has a sensitivity of 85% and a specificity of 94% for the detection of NAFLD [114]. As ultrasound is a low-cost and reliable diagnostic tool, it has become the modality of choice. An important caveat is in the morbidly obese population (BMI 35–40 kg/m2), wherein one study revealed a 95.4% prevalence confirmed by liver biopsy at the time of bariatric surgery, but the sensitivity of pre-surgical detection by ultrasound was reduced to 49.1% [115]. There have been limited studies detailing the sensitivity and specificity of CT and MRI scanning, given the need for comparative histologic examination. One study including 131 patients with 675

Expert Review of Gastroenterology & Hepatology Downloaded from informahealthcare.com by Nanyang Technological University on 04/26/15 For personal use only.

Review

Normal liver

Schneier, Citti & Dieterich

Steatosis

NASH

Figure 3. Histologic appearance of normal liver biopsy, simple steatosis and NASH, with hepatocyte ballooning and fibrosis seen on trichrome stain. NASH: Nonalcoholic steatohepatitis.

radiographic evidence of NAFLD on non-contrast CT, contrast-enhanced CT or MRI prior to partial hepatectomy showed sensitivities of 33, 50 and 88%, respectively, and specificities of 100, 83 and 63%, respectively [116]. In recent years, newer imaging modalities have been developed for the identification and risk stratification of NAFLD, including controlled attenuation parameter (CAP) fibroscan as well as magnetic resonance (MR)-proton density fat fraction. CAP is an ultrasound-based transient elastography technique approved in April 2014 by the US FDA for screening of liver disease. CAP has been shown in a recent meta-analysis to have good sensitivity (ranging from 79 to 92%) and adequate specificity (ranging from 75 to 92%) for the detection of hepatic steatosis, and detects advanced fibrosis in patients effectively [117,118]. Given the reliance on ultrasound, it is notable that this test is limited in those with greater depth of subcutaneous tissue. Similarly, MR-proton density fat fraction, which uses MR imaging to quantify hepatic fat content, has shown promise in the detection and grading of hepatic steatosis. However, it has limited accuracy when compared to liver biopsy in the setting of concomitant fibrosis, as it is only accurate with presence of >30% steatosis [119,120]. Biopsy

Radiographic studies are all limited in that one cannot differentiate fat content in the liver due to alcoholic disease and NAFLD [121]. In patients in whom radiographic studies and laboratory testing are inconclusive, liver biopsy remains the gold standard for diagnosis. Biopsy is the only method to demonstrate the severity of NAFLD [122]. Although limited by cost, sampling error and procedure-related complications, the information from a liver biopsy may facilitate education of patients about their disease and may serve to motivate appropriate treatment and or screening protocols in the case of cirrhosis. Risk stratification scores have been developed, including the NAFLD fibrosis score (including age, BMI, hyperglycemia, platelet count, albumin and aspartate transaminase/alanine transaminase ratio), which has been validated in a 2011 meta-analysis [123]. 676

The NAFLD fibrosis score uses clinical and biochemical parameters to reliably identify patients with advanced fibrosis. It is limited, however, by the prevalence of intermediate scores which compel further testing. The enhanced liver fibrosis panel has also been studied in the above meta-analysis [124], with a sensitivity of 80% and a specificity of 90%; this test, however, requires measurement of three matrix turnover proteins (hyaluronic acid, TIMP-1 and PIIINP). Also, circulating cytokeratin-18 has been studied [117], and found to have a sensitivity of 78% and specificity of 87% for steatohepatitis. This test, however, is not commercially available and, therefore, not of practical use presently. The AASLD consensus guidelines recommend biopsy for patients who are at risk of steatohepatitis or advanced fibrosis by NAFLD fibrosis score, enhanced liver fibrosis, or transient elastography, or those in whom a competing diagnosis cannot be excluded without tissue diagnosis [116]. Histologic findings of NAFLD require observation of ballooning, macrovesicular steatosis, intra-acinar and portal inflammation, and may include fibrosis. Examples of the histologic findings of steatosis and NASH can be found in FIGURE 3. A disease grading system, the NAFLD activity score (NAS), has been designed to further characterize NAFLD particularly for clinical research [124]. Based on the histologic scores for steatosis (0–3), lobular inflammation (0–3) and hepatocyte ballooning (0–2), an unweighted sum of NAS ‡5 is consistent with NASH and those less than 3 are consistent with ‘not NASH’. Fibrosis on liver biopsies is staged separately (0–4). The composite NAS and fibrosis stage together comprise the NASH Clinical Research Network scoring system. As fibrosis progresses to cirrhosis, biopsies may lose their distinct pattern of inflammation and may be labeled as CC. Management

Given that patients with NAFLD without steatohepatitis have an excellent prognosis, the goal for those patients remains control of comorbid disease processes: obesity, hyperlipidemia and insulin resistance. Treatment of patients with NASH will be discussed further here and the treatment trials are summarized in TABLE 3. Many therapies have been tried for treatment or prevention of progression of NASH; weight loss, however, remains the only therapy with good evidence to support safety and efficacy. A randomized control trial evaluating a combination of diet, exercise and behavioral modification, with a goal of 7–10% weight loss, found an improvement in 48-week NASH histologic activity score (NAS) of at least 3 points, particularly in those who achieved the weight loss goal [125]. Therefore, weight loss achieved by diet and exercise, supervised by a medical practitioner, is recommended for patients with NASH. A reasonable goal for weight loss is between 0.5 and 1 kg/ week, as more rapid weight loss and poor nutrition have been associated with worsening fibrosis [126]. Medication

Aimed at reducing oxidative stress and hepatocellular injury, vitamin E was seen in observational studies to improve surrogate end points such as transaminase levels [127]. Randomized Expert Rev. Gastroenterol. Hepatol. 9(5), (2015)

informahealthcare.com

Weight loss

Vitamin E

Pioglitazone

Metformin

UDCA

Vitamin E and UDCA

Orlistat

Probucol

Betaine

Atorvastatin

Pentoxifylline

Resveratrol

Ethyl-eicosapentanoic acid

Omega-3 fatty acid

Promrat et al. (2010)

Sanyal et al. (2010)

Boettcher et al. (2012)

Rakoski et al. (2010)

Leuschner et al. (2010)

Pietu et al. (2012)

Zelber-Sagi et al. (2006)

Merat et al. (2003)

Abdelmalek et al. (2009)

Foster et al. (2011)

Van Wagner et al. (2011)

Chachay et al. (2014)

Sanyal et al. (2014)

Scorletti et al. (2014)

Randomized controlled trial, n = 103

Randomized controlled trial, n = 243

Randomized controlled trial, n = 20

Randomized controlled trial, n = 30

Randomized controlled trial, n = 80

Randomized controlled trial, n = 35

Randomized controlled trial, n = 27

Randomized controlled trial, n = 52

Pending results

No significant difference in histologic findings

No significant difference in insulin resistance, steatosis or abdominal fat distribution. Elevations in transaminases noted in treatment group

Statistically significant improvements in AST and ALT, steatosis and cellular ballooning. No overall statistically significant improvement in histology

Significant improvement in CT correlates of steatosis using atorvastatin in combination with vitamins C and E versus placebo (34.4 vs 70.4%, p = 0.001)

No significant difference in histologic findings

Statistically improved ALT. No significant difference in histologic findings

Statistically improved ALT, serum insulin and greater degree of reversibility on ultrasound. No significant difference in histologic findings

Normalization of AST, ALT and gamma-glutamyl transferase in 80, 70 and 65% of the patients, respectively

Retrospective cohort, n = 101

[134]

Metformin failed to improve any pooled outcome: steatosis, hepatocyte ballooning or ALT

[147]

[146]

[145]

[144]

[143]

[142]

[141]

[140]

[139]

[138]

[132]

Improvement in ballooning degeneration, lobular inflammation and steatosis, when compared to placebo, combined ORs of 2.11 (95% CI: 1.33–3.36), 2.58 (95% CI: 1.68–3.97) and 3.39 (95% CI: 2.19–5.25), respectively

No significant differences in overall histology at 18 months, significant improvement in lobular inflammation (modified Brunt score p = 0.011, NAS p = 0.005)

[128]

[125]

Improvement in 48-week NAS of at least 3 points (72 vs 30%, p = 0.03) Higher rate of improved NASH by histology versus placebo (43 vs 19%, p = 0.001)

Ref.

Results

Randomized controlled trial, n = 186

Meta-analysis of nine trials: five with thiazolidinedione, three with metformin and one with combination therapy

Meta-analysis of four randomized control trials, n = 334

Randomized controlled trial, n = 247

Randomized controlled trial, n = 31

Study design, n

CT: Computed tomogram; NAS: NASH histologic activity score; NASH: Nonalcoholic steatohepatitis; OR: Odds ratio; UDCA: Ursodeoxycholic acid.

Intervention

Study (year)

Table 3. Summary of treatment trials for nonalcoholic fatty liver disease.

Expert Review of Gastroenterology & Hepatology Downloaded from informahealthcare.com by Nanyang Technological University on 04/26/15 For personal use only.

Management & diagnosis of fatty liver disease

Review

677

Expert Review of Gastroenterology & Hepatology Downloaded from informahealthcare.com by Nanyang Technological University on 04/26/15 For personal use only.

Review

Schneier, Citti & Dieterich

controlled trials of 800 international units (IU) daily conducted later showed improved histologic findings when compared with pioglitazone, particularly in patients without diabetes [128]. The AASLD consensus guidelines now recommend 800 IU daily as first-line treatment for non-diabetic patients with biopsy-proven NASH [113]. Further studies are required to understand the long-term benefits of vitamin E therapy, as well as the utility in diabetic or cirrhotic patients. Insulin-sensitizing agents are thought to improve hepatic histology and biochemical markers by improving insulin resistance and, therefore, fatty deposition within the liver [128–131]. In a meta-analysis of four randomized trials of thiazolidinediones, such as pioglitazone, versus placebo, there was improved histology marked by reduced ballooning degeneration, lobular inflammation and steatosis, with a combined OR being 2.11 (95% CI: 1.33–3.36) [132]. Given the frequency of side effects from this medication class, however, including the risk of bladder cancer [133], they have been removed from French and German markets. Recommendations for use of these medications are limited to the diabetic patients who would otherwise be candidates for this drug class [113]. Metformin has similarly been evaluated for its utility in NAFLD [134], but was found to have an insignificant effect on liver histology. Recently, the glucagon-like peptide-1 receptor agonist, exenatide, has been shown to improve hepatic steatosis in murine models through the antiinflammatory mammalian sirtuin 1 pathway, thus suggesting exenatide may have a role in the treatment of NAFLD [135]. Another agent, ursodeoxycholic acid (UDCA), has also been studied. UDCA has been found to be anti-inflammatory, cytoprotective and anti-apoptotic due to activation of glucocorticoid and other survival pathways [136]; however, a large multicenter, randomized control trial in 2004 demonstrated no improvement in laboratory or histologic parameters [137]. Studies conducted later comparing high-dose UDCA (25–28 mg/kg/day) to placebo showed no statistically significant improvement in laboratory parameters or histology at 18 months [138]. Notably, however, a single-center long-term follow-up study has shown improved laboratory parameters after 10 years of dual therapy with vitamin E and low-dose UDCA [139]. The clinical relevance of these findings is unknown as the rates of progression to fibrosis or cirrhosis were not examined. Several other drugs have been studied for treatment of NASH, including orlistat [140], probucol [141], betaine [142], atorvastatin [143], pentoxifylline [144], resveratrol [145] and ethyleicosapentanoic acid [146]. None of the drugs studied has shown a definitive and/or reproducible benefit to be recommended as a primary treatment for NASH. A randomized, double-blind, placebo-controlled trial is underway to evaluate the long-term efficacy of w-3 fatty acid on hepatic fat composition [147], and investigation continues to identify and target new mechanisms to mitigate hepatic steatosis and inflammation.

procedures (laparoscopic adjustable gastric banding), malabsorptive procedures, Roux-en-Y gastric bypass and biliointesinal diversion have been shown to improve aspects of the metabolic syndrome, such as Type 2 diabetes [148]. Histologic improvement has also been observed following bariatric surgery in obese patients with NASH. An initial study of 18 consecutive patients undergoing Roux-en-Y gastric bypass showed that with a mean excess weight loss of 60%, there was resolution of steatosis in 84%, hepatocyte ballooning in 50%, and findings of fibrosis in 75%, with a persistence of lobular inflammatory infiltrate in 81%, on 2-year interval liver biopsies [149]. A subsequent larger case series of 381 patients undergoing biliointestinal bypass, laparoscopic gastric banding or Roux-en-Y gastric bypass examined 5-year interval liver biopsies after bariatric surgery [150]. Mathurin et al. showed that fibrosis remained the same or worsened in >90% of patients after surgery, but there was sustained improvement in steatosis and ballooning. The study also showed no significant difference in the type of weight loss surgery on NAS score. In 2010, a Cochrane review concluded that there is limited evidence for bariatric surgery in the management of NAFLD due to the paucity of wellstructured, well-powered trials [151]. At this time, it is unclear whether bariatric surgery should be performed for primary treatment of NASH, or which type of bariatric surgery may confer benefit. Given the risk for worsening fibrosis, postoperative management of bariatric surgery patients should include close monitoring. Surveillance

Because survival after HCC becomes symptomatic is so low (0–10%, 5-year survival) [152], early detection is essential. Currently, there is little-to-no definitive data showing survival advantage with screening for HCC [153]. There are only two randomized controlled trials showing any survival benefit from screening, both looking exclusively at an HBV cohort in China, each with internal validity concerns [154,155]. Because of this, the AASLD has tried to explore a more pragmatic approach to screening. The goal of screening is to strike a balance between early detection, when intervention can prolong life, and the cost of screening the population in question. As a framework, it defines an effective intervention as one being able to prolong life by 100 days and a cost-effective intervention as one that costs US$50,000/year or less [156]. It has found that in patients with non-viral cirrhosis, screening becomes cost-effective when the annual incidence exceeds 1.5–2%. As reviewed above, the incidence of HCC in NASH cirrhotics is ill-defined, ranging anywhere from 2.4% over 7 years to 12.8% over 3 years [112]. Thus, while there are no definitive guidelines for NAFLD/ NASH per se, many practitioners choose to screen only once the diagnosis of cirrhosis is made. Expert opinion

Surgery

Surgical treatment of obesity has been proposed as the means to sustained weight loss in patients with NASH. Restrictive 678

There is no consensus regarding appropriate risk stratification for screening. Given the morbidity and mortality associated with cirrhosis and HCC, there is a clear role for such a tool. Expert Rev. Gastroenterol. Hepatol. 9(5), (2015)

Expert Review of Gastroenterology & Hepatology Downloaded from informahealthcare.com by Nanyang Technological University on 04/26/15 For personal use only.

Management & diagnosis of fatty liver disease

Certainly, the prevalence of subclinical disease in patients with metabolic syndrome, those of Hispanic ethnicity, and those with a strong family history and may be even with PNPLA3 may benefit from earlier detection and optimization of modifiable risk factors. More robust prospective studies are required to predict both the natural history and the potential benefits of treatment. It is clear that liver biopsy has limited the enrollment of patients in such vital studies. With the recent development of MR-proton density fat fraction protocols, and the measurement of CAP, non-invasive diagnosis and serial examinations can be standardized for research. Such approaches will be more attractive to potential study participants, and may aid in a better understanding of this complex and common disease. Lastly, given the proven benefits of weight loss for NAFLD patients, bariatric surgery should be studied further as a potential curative strategy. It is not known whether there are sustained benefits from weight loss in patients with liver disease; however, both the increased incidence of obesity and weight loss surgery lend themselves to further study.

Review

Five-year view

With the recent development of tolerable and effective HCV treatments, and the spread of HBV vaccination, it is likely that non-viral etiologies of cirrhosis will become the norm, not the exception. As the obesity epidemic continues, there are models predicting that by 2030, in the USA, there will be 65 million more obese adults and an additional 6–8.5 million more cases of diabetes [157]. With this data in mind, it is projected that NAFLD will be the leading cause of liver transplantation by 2020 [158]. Thus, better understanding of the fateful step from NAFLD to NASH, and exploring new and effective therapeutic opportunities will be critical. Financial & competing interests disclosure

DT Dieterich has had research support and has acted as a consultant for Gilead, BMS, Merck and Abbvie. The authors have no other relevant affiliations or financial involvement with any organization or entity with a financial interest in or financial conflict with the subject matter or materials discussed in the manuscript apart from those disclosed. No writing assistance was utilized in the production of this manuscript.

Key issues .

Reviewing the pathogenesis and epidemiology of nonalcoholic fatty liver disease.

.

Understanding the transformation from nonalcoholic fatty liver to nonalcoholic steatohepatitis.

.

Interventions which may help stifle the transformation.

.

Prevention of complications such as cirrhosis and malignancy.

steatohepatitis. Clin Liver Dis 2009;13: 511-31

References 1.

2.

3.

4.

5.

6.

Abdelmalek M, Ludwig J, Lindor KD. Two cases from the spectrum of nonalcoholic steatohepatitis. J Clin Gastroenterol 1995;20:127-30 Brunt EM. Nonalcoholic steatohepatitis: pathologic features and differential diagnosis. Semin Diagn Pathol 2005;22: 300-38 Brunt EM, Tiniakos DG. Alcoholic and nonalcoholic fatty liver disease. In: Odze RD, Goldblum JR, editors. Surgical Pathology of the GI Tract, Liver, Biliary Tract and Pancreas. 2nd edition. Elsevier; Philadelphis, PA: 2009. p. 1007-14 Ludwig J, Viggiano TR, McGill DB, et al. Nonalcoholic steatohepatitis: mayo Clinic experiences with a hitherto unnamed disease. Mayo Clin Proc 1980;55:434-8 Younossi ZM, Stepanova M, Afendy M, et al. Changes in the prevalence of the most common causes of chronic liver diseases in the United States from 1988 to 2008. Clin Gastroenterol Hepatol 2011;9:524-30 Argo CK, Caldwell SH. Epidemiology and natural history of non-alcoholic

informahealthcare.com

7.

8.

9.

Lazo M, Hernandez R, Eberhardt MS, et al. Prevalence of nonalcoholic fatty liver disease in the United States: the Third National Health and Nutrition Examination Survey, 1998-1994. Am J Epidemiol 2013;178(1): 38-45 Bedogni G, Miglioli L, Masutti F, et al. Prevalence of and risk factors for nonalcoholic fatty liver disease: the Dionysos nutrition and liver study. Hepatology 2005;42:44-52 Caballerı´a L, Pera G, Auladell MA, et al. Prevalence and factors associated with the presence of nonalcoholic fatty liver disease in an adult population in Spain. Eur J Gastroenterol Hepatol 2010;22:24-32

10.

Bedogni G, Bellentani S. Fatty liver: how frequent is it and why? Ann Hepatol 2004;3:63-5

11.

Williams CD, Stengel J, Asike MI, et al. Prevalence of nonalcoholic fatty liver disease and nonalcoholic steatohepatitis among a largely middle-aged population utilizing ultrasound and liver biopsy: a prospective study. Gastroenterology 2011;140(1):124-31

12.

Kojima SI, Watanabe N, Numata M, et al. Increase in the prevalence of fatty liver in Japan over the past 12 years: analysis of clinical background. J Gastroenterol 2003;38:954-61

13.

Amarapurkar D, Kamani P, Patel N, et al. Prevalence of non-alcoholic fatty liver disease: population based study. Ann Hepatol 2007;6:161-3

14.

Fan JG, Farrell GC. Epidemiology of non-alcoholic fatty liver disease in China. J Hepatol 2009;50:204-10

15.

Yamamoto K, Takada Y, Fujimoto Y, et al. Nonalcoholic steatohepatitis in donors for living donor liver transplantation. Transplantation 2007;83:257-62

16.

Lee JY, Kim KM, Lee SG, et al. Prevalence and risk factors of non-alcoholic fatty liver disease in potential living liver donors in Korea: a review of 589 consecutive liver biopsies in a single center. J Hepatol 2007;47:239-44

17.

Reha JL, Lee S, Hofmann LJ. Prevalence and predictors of nonalcoholic steatohepatitis in obese patients undergoing bariatric surgery: a department of defense experience. Am Surg 2014;80(6):595-9

679

Review 18.

Expert Review of Gastroenterology & Hepatology Downloaded from informahealthcare.com by Nanyang Technological University on 04/26/15 For personal use only.

19.

Schneier, Citti & Dieterich

Browning JD, Szczepaniak LS, Dobbins R, et al. Prevalence of hepatic steatosis in an urban population in the United States: impact of ethnicity. Hepatology 2004;40: 1387-95 Weston SR, Leyden W, Murphy R, et al. Racial and ethnic distribution of nonalcoholic fatty liver in persons with newly diagnosed chronic liver disease. Hepatology 2005;41:372-9

20.

Younossi ZM, Stepanova M, Negro F, et al. Nonalcoholic fatty liver disease in lean individuals in the United States. Medicine (Baltimore) 2012;91:319-27

21.

Adams LA, Lymp JF, St Sauver J, et al. The natural history of nonalcoholic fatty liver disease: a population-based cohort study. Gastroenterology 2005;129:113-21

22.

23.

24.

25.

26.

27.

28.

29.

30.

Kallwitz ER, Kumar M, Aggarwal R, et al. Ethnicity and nonalcoholic fatty liver disease in an obesity clinic: the impact of triglycerides. Dig Dis Sci 2008;53:1358-63 Schneider AL, Lazo M, Selvin E, Clark JM. Racial differences in nonalcoholic fatty liver disease in the U.S. population. Obesity (Silver Spring) 2014;22:292-9 Smits MM, Ioannou GN, Boyko EJ, et al. Non-alcoholic fatty liver disease as an independent manifestation of the metabolic syndrome: results of a US national survey in three ethnic groups. J Gastroenterol Hepatol 2013;28:664-7 Fallon MB, Pan JJ. Gender and racial differences in nonalcoholic fatty liver disease. World J Hepatol 2014;6(5):274-83 Casas YG, Schiller BC, DeSouza CA, et al. Total and regional body composition across age in healthy hispanic and white women of similar socioeconomic status. Am J Clin Nutr 2001;73:13-18 Miyazaki Y, Glass L, Triplitt C, et al. Abdominal fat distribution and peripheral and hepatic insulin resistance in type 2 diabetes mellitus. Am J Physiol Endocrinol Metab 2002;283:E1135-43

31.

32.

Liu YL, Patman GL, Leathart JB, et al. Carriage of the PNPLA3 rs738409 C >G polymorphism confers an increased risk of non-alcoholic fatty liver disease associated hepatocellular carcinoma. J Hepatol 2014; 61(1):75-81 Guerrero R, Vega GL, Grundy SM, et al. Ethnic differences in hepatic steatosis: an insulin resistance paradox. Hepatology 2009;49:791-01

nonalcoholic steatohepatitis: the central role of nontriglyceride fatty acid metabolites. Hepatology 2010;52(2):774-88 45.

Sanyal AJ, Campbell-Sargent C, Mirshahi F, et al. Nonacloholic steatohepatitis: association of insulin resistance and mitochondrial abnormalities. Gastroenterology 2001;120(5):1183

46.

Cani PD, Possemiers S, Van de Wiele T, et al. Changes in gut microbiota control inflammation in obese mice through a mechanism involving GLP-2-driven improvement of gut permeability. Gut 2009;58:1091-03

33.

Wagenknecht LE, Scherzinger AL, Stamm ER, et al. Correlates and heritability of nonalcoholic fatty liver disease in a minority cohort. Obesity (Silver Spring) 2009;17:1240-6

47.

34.

Marchesini G, Bugianesi E, Forlani G, et al. Nonalcoholic fatty liver, steatohepatitis, and the metabolic syndrome. Hepatology 2003;37:917-23

Miele L, Valenza V, La Torre G, et al. Increased intestinal permeability and tight junction alterations in nonalcoholic fatty liver disease. Hepatology 2009;49:1877-87

48.

35.

Powell EE, Cooksley WG, Hanson R, et al. The natural history of nonalcoholic steatohepatitis: a follow-up study of forty-two patients for up to 21 years. Hepatology 1990;11:74-80

Brun P, Castagliuolo I, Di Leo V, et al. Increased intestinal permeability in obese mice: new evidence in the pathogenesis of nonalcoholic steatohepatitis. Am J Physiol Gastrointest Liver Physiol 2007;292: G518-25

36.

Marchesini G, Brizi M, Bianchi G, et al. Nonalcoholic fatty liver disease: a feature of the metabolic syndrome. Diabetes 2001; 50(8):1844-50

49.

37.

Angelico F, Del Ben M, Conti R, et al. Non-alcoholic fatty liver syndrome: a hepatic consequence of common metabolic diseases. J Gastroenterol Hepatol 2003;18:1115-17

Wigg AJ, Roberts-Thomson IC, Dymock RB, et al. The role of small intestinal bacterial overgrowth, intestinal permeability, endotoxaemia, and tumour necrosis factor alpha in the pathogenesis of non-alcoholic steatohepatitis. Gut 2001;48: 206-11

50.

Schnabl B, Brenner DA. Interactions between the intestinal microbiome and liver diseases. Gastroenterology 2014;146(6): 1513-24

51.

Targher G, Bertolini L, Rodella S, et al. Associations between plasma adiponectin concentrations and liver histology in patients with nonalcoholic fatty liver disease. Clin Endocrinol 2006;64:679-83

52.

Bianchi G, Bugianesi E, Frystyk J, et al. Adiponectin isoforms, insulin resistance and liver histology in nonalcoholic fatty liver disease. Dig Liver Dis 2011;43:73-7

38.

39.

Bugianesi E, Gastaldelli A, Vanni E, et al. Insulin resistance in non-diabetic patients with non-alcoholic fatty liver disease: sites and mechanisms. Diabetologia 2005;48(4): 634-42 Cohen JC, Horton JD, Hobbs HH. Human fatty liver disease: old questions and new insights. Science 2011;332(6037): 1519-23

40.

Brown MS, Goldstein JL. Selective versus total insulin resistance: a pathogenic paradox. Cell Metab 2008;7(2):95-6

53.

Van der Poorten D, Milner KL, Hui J, et al. Visceral fat: a key mediator of steatohepatitis in metabolic liver disease. Hepatology 2008;48:449-57

41.

Marchesini G, Brizi M, Bianchi G, et al. Nonalcoholic fatty liver disease: a feature of the metabolic syndrome. Diabetes 2001; 50(8):1844-50

Hui JM, Hodge A, Farrell GC, et al. Beyond insulin resistance in NASH: TNF-alpha or adiponectin? Hepatology 2004;40:46-54

54.

Romeo S, Kozlitina J, Xing C, et al. Genetic variation in PNPLA3 confers susceptibility to nonalcoholic fatty liver disease. Nat Genet 2008;40:1461-5

42.

Larter CZ, Farrell GC. Insulin resistance, adiponectin, cytokines in NASH: which is the best target to treat? J Hepatol 2006; 44(2):253-61

55.

Polyzos SA, Toulis KA, Goulis DG, et al. Serum total adiponectin in nonalcoholic fatty liver disease: a systematic review and meta-analysis. Metabolism 2011;60(3): 313-26

56.

Pasarin M, La Mura V, Gracia-Sancho J, et al. Sinusoidal endothelial dysfunction

Sookoian S, Pirola CJ. Meta-analysis of the influence of I148M variant of patatin-like phospholipase domain containing 3 gene (PNPLA3) on the susceptibility and histological severity of nonalcoholic fatty liver disease. Hepatology 2011;53:1883-94

680

Musso G, Gambino R, De Michieli F, et al. Dietary habits and their relations to insulin resistance and postprandial lipemia in nonalcoholic steatohepatitis. Hepatology 2003;37(4):909-16

43.

Angulo P. Nonalcoholic fatty liver disease. N Engl J Med 2002;346(16):1221

44.

Neuschwander-Tetri BA. Hepatic lipotoxicity and the pathogenesis of

Expert Rev. Gastroenterol. Hepatol. 9(5), (2015)

Management & diagnosis of fatty liver disease

precedes inflammation and fibrosis in a model of NAFLD. PLoS One 2012;7(4): e32785

Expert Review of Gastroenterology & Hepatology Downloaded from informahealthcare.com by Nanyang Technological University on 04/26/15 For personal use only.

57.

58.

59.

60.

61.

62.

Francque S, Lalwman W, Verbeke L, et al. Increased intrahepatic resistance in severe steatosis: endothelial dysfunction, vasoconstrictor overprodution and altered microvascular architecture. Lab Invest 2012; 92(10):1428-39 Li L, Chen L, Hu L, et al. Nuclear factor high-mobility group box1 mediating the activation of Toll-like receptor 4 signaling in hepatocytes in the early stage of nonalcoholic fatty liver disease in mice. Hepatology 2011;54(5):1620-30 Tajiri K, Shimizu Y, Tsuneyama K, et al. Role of liver-infiltrating CD3+CD56+ natural killer T cells in the pathogenesis of nonalcoholic fatty liver disease. Eur J Gatroenterol Hepatol 2009;21(6):673 Guy CD, Suzuki A, Zdanowicz M, et al. Hedgehog pathway activation parallels histologic severity of injury and fibrosis in human nonalcoholic fatty liver disease. Hepatology 2012;55(6):1711-21 Syn WK, Choi SS, Liaskou E, et al. Osteopontin is induced by hedgehog pathway activation and promotes fibrosis progression in nonalcoholic steatohepatitis. Hepatology 2011;53(1):106-15 Diehl AM. Tumor necrosis factor and its potential role in insulin resistance and nonalcoholic fatty liver disease. Clin Liver Dis 2004;8:619-38

69.

70.

71.

72.

73.

74.

75.

Wong VW, Wong GL, Choi PC, et al. Disease progression of non-alcoholic fatty liver disease: a prospective study with paired liver biopsies at 3 years. Gut 2010;59(7): 969-74 Torres DM, Williams CD, Harrison SA. Features, diagnosis, and treatment of nonalcoholic fatty liver disease. Clin Gastroent and Hepatol 2012;10(8):837-58 Sorrentino P, Tarantino G, Conca P, et al. Silent non-alcoholic fatty liver disease-a clinical-histological study. J Hepatol 2004;41:751-7 Argo CK, Northup PG, Al-Osaimi AM, et al. Systematic review of risk factors for fibrosis progression in non-alcoholic steatohepatitis. J Hepatol 2009;51(2):371-9 Hui AY, Wong VW, Chan HL, et al. Histological progression of non-alcoholic fatty liver disease in Chinese patients. Aliment Pharmacol Ther 2005;21:407-13 Fassio E, Alvarez E, Domı´nguez N, et al. Natural history of nonalcoholic steatohepatitis: a longitudinal study of repeat liver biopsies. Hepatology 2004;40: 820-6 Singh S, Allen AM, Wang Z, et al. Fibrosis progression in nonalcoholic fatty liver vs nonalcoholic steatohepatitis: a systematic review and meta-analysis of paired-biopsy studies. Clin Gastroenterol Hepatol 2014. [Epub ahead of print]

76.

Rensen SS, Slaats Y, Nijhuis J, et al. Increased hepatic myeloperoxidase activity in obese subjects with nonalcoholic steatohepatitis. Am J Pathol 2009;175(4): 1473-82

Valenti L, Al-Serri A, Daly AK, et al. Homozygosity for the patatin-like phospholipase-3/adiponutrin I148M polymorphism influences liver fibrosis in patients with nonalcoholic fatty liver disease. Hepatology 2010;51(4):1209-17

77.

64.

Rensen SS, Slaats Y, Driessen A, et al. Activation of the complement system in human nonalcoholic fatty liver disease. Hepatology 2009;50(6):1809-17

Angulo P, Keach AC, Batts KB, et al. Independent predictors of liver fibrosis in patients with nonalcoholic steatohepatitis. Hepatology 1999;30(6):1356

78.

65.

Hernandez-Gea V, Friedman SL. Pathogenesis of liver fibrosis. Annu Rev Pathol 2011;6:425-56

66.

Poynard T, Mathurin P, Lai CL, et al. A comparison of fibrosis progression in chronic liver diseases. J Hepatol 2003;38:257

Noureddin M, Yates KP, Vaughn IA, et al. Clinical and histological determinants of nonalcoholic steatohepatitis and advanced fibrosis in elderly patients. Hepatology 2013;58(5):1644-54

79.

Hossain N, Afendy A, Stepanova M, et al. Independent predictors of fibrosis in patients with nonalcoholic fatty liver disease. Clin Gastroenterol Hepatol 2009;7(11): 1224-9

80.

Petta S, Amato MC, Di Marco V, et al. Visceral adiposity index is associated with significant fibrosis in patients with non-alcoholic fatty liver disease. Aliment Pharmacol Ther 2012;35(2):238-47

63.

67.

Schuppan D, Afdhal NH. Liver Cirrhosis. Lancet 2008;371(9615):838-51

68.

Pais R, Charlotte F, Fedchuk L, et al. A systematic review of follow-up biopsies reveals disease progression in patients with non-alcoholic fatty liver. J Hepatol 2013; 59(3):550-6

81.

informahealthcare.com

Matteoni CA, Younossi ZM, Gramlich T, et al. Non alcoholic fatty liver disease (a

Review

spectrum of clinical and pathological severity). Gastroenterology 1999;116: 1413-19 82.

Harrison SA, Torgerson S, Hayashi PH. The natural history of non alcoholic fatty liver disease (a clinical histopathological study). Am J Gastroenterol 2003;98:2042-7

83.

Dam-Larsen S, Franzmann M, Andersen IB, et al. Long term prognosis of fatty liver (risk of chronic liver disease and death). Gut 2004;53:750-5

84.

Ekstedt M, Franzen LE, Mathiesen UL, et al. Long-term follow-up of patients with NAFLD and elevated liver enzymes. Hepatology 2006;44:865-73

85.

So¨derberg C, Sta˚l P, Askling J, et al. Decreased survival of subjects with elevated liver function tests during a 28- year follow-up. Hepatology 2010;51:595-02

86.

Rafiq N, Bai C, Fang Y, et al. Longterm follow-up of patients with nonalcoholic fatty liver. Clin Gastroenterol Hepatol 2009;7: 234-8

87.

Hamaguchi M, Kojima T, Takeda N, et al. Nonalcoholic fatty liver disease is a novel predictor of cardiovascular disease. World J Gastroenterol 2007;13:1579-84

88.

Bhala N, Angulo P, van der Poorten D, et al. The natural history of nonalcoholic fatty liver disease with advanced fibrosis or cirrhosis: an international collaborative study. Hepatology 2011;54:1208-16

89.

Targher G, Bertolini L, Padovani R, et al. Prevalence of nonalcoholic fatty liver disease and its association with cardiovascular disease among type 2 diabetic patients. Diabetes Care 2007;30:1212-18

90.

Haring R, Wallaschofski H, Nauck M, et al. Ultrasonographic hepatic steatosis increases prediction of mortality risk from elevated serum gamma-glutamyl-transpeptidase levels. Hepatology 2009;50:1403-11

91.

Vanwagner LB, Bhave M, Te HS, et al. Patients transplanted for nonalcoholic steatohepatitis are at increased risk for postoperative cardiovascular events. Hepatology 2012;56(5):1741-50

92.

El-Serag HB, Rudolph KL. Hepatocellular carcinoma: epidemiology and molecular carcinogenesis. Gastroenterology 2007;132: 2557-76

93.

Hashimoto E, Yatsuji S, Tobari M, et al. Hepatocellular carcinoma in patients with nonalcoholic steatohepatitis. J Gastroenterol 2009;44:89-95

94.

Paredis V, Zalinski S, Chelbi E, et al. Hepatocellular carcinomas in patients with metabolic syndrome often develop without

681

Review

Schneier, Citti & Dieterich

Expert Review of Gastroenterology & Hepatology Downloaded from informahealthcare.com by Nanyang Technological University on 04/26/15 For personal use only.

significant liver fibrosis: a pathological analysis. Hepatology 2009;49(3):851-9 95.

Cotrim HP, Parana R, Braga E, et al. Nonalcoholic steatohepatitis and hepatocellular carcinoma: natural history? Am J Gastroenterol 2000;44:3018-19

96.

Zen Y, Katayanagi K, Tsuneyama K, et al. Hepatocellular carcinoma arising in non-alcoholic steatohepatitis. Pathol Int 2001;51:127-31

97.

98.

99.

Mori S, Yamasaki T, Sakaida I, et al. Hepatocellular carcinoma with nonalcoholic steatohepatitis. J Gastroenterol 2004;39: 391-6 Yoshioka Y, Hashimoto E, Yatsuji S, et al. Nonalcoholic steatohepatitis: cirrhosis, hepatocellular carcinoma and burn-out NASH. J Gastroenterol 2004;39:1215-18 Bullock RE, Zaitoun AM, Aithal GP, et al. Association of non-alcoholic steatohepatitis without significant fibrosis with hepatocellular carcinoma. J Hepatol 2004;41:685-90

100.

Shimada M, Hashimoto E, Taniai M, et al. Hepatocellular carcinoma in patients with nonalcoholic steatohepatitis. J Hepatol 2002;37:154-60

101.

Ong JP, Pitts A, Younossi ZM. Increased overall mortality and liver-related mortality in non-alcoholic fatty liver disease. J Hepatol 2008;9:608-12

102.

Amarapurkar DN, Dharod M, Gautam S, et al. Risk of development of hepatocellular carcinoma in patients with NASH-related cirrhosis. Trop Gastroenterol 2013;34(3): 159-63

103.

104.

105.

106.

107.

Ascha MS, Hanouneh IA, Lopez R, et al. The incidence and risk factors of hepatocellular carcinoma in patients with nonalcoholic steatohepatitis. Hepatology 2010;51(6):1972-8 Ratziu VD, Bonyhay L, Di Martino V, et al. Survival, liver failure, and hepatocellular carcinoma in obesity-related cryptogenic cirrhosis. Hepatology 2002;35: 1485-93 Calle EE, Rodriguez C, Walker-Thurmond K, et al. Overweight, obesity, and mortality from cancer in a prospectively studied cohort of U.S. adults. N Engl J Med 2003;348:1625-38 Moller H, Mellemgaard A, Lindvig K, et al. Obesity and cancer risk: a Danish record-linkage study. Eur J Cancer 1994;30A:344-50 El-Serag HB, Hampel H, Javadi F. The association between diabetes and hepatocellular carcinoma: a systematic

682

review of epidemiologic evidence. Clin Gastroenterol Hepatol 2006;4:369-80 108.

Marrero JA, Fontana RJ, Su GL, et al. NAFLD may be a common underlying liver disease in patients with hepatocellular carcinoma in the United States. Hepatology 2002;36:1349-54

109.

Bugianesi E, Leone N, Vanni E, et al. Expanding the natural history of nonalcoholic steatohepatitis: from cryptogenic cirrhosis to hepatocellular carcinoma. Gastroenterology 2002;123: 134-40

110.

Regimbeau JM, Colombat M, Mognol P, et al. Obesity and diabetes as a risk factor for hepatocellular carcinoma. Liver Transpl 2004;10(2):S69-73

111.

Hui JM, Kench JG, Chitturi S, et al. Long-term outcomes of cirrhosis in nonalcoholic steatohepatitis compared with hepatitis C. Hepatology 2003;38(2):420-7

112.

White DL, Kanwal F, El-Sarag H. Association between nonalcoholic fatty liver disease and risk for hepatocellular cancer, based on systematic review. Clin Gantroenterol Hepatol 2012;10(12):1342

113.

Chalasani N, Younossi Z, Lavine JE, et al. The diagnosis and management of non-alcoholic fatty liver disease: practice Guideline by the American Association for the Study of Liver Diseases, American College of Gastroenterology, and the American Gastroenterological Association. Hepatology 2012;55:2005-23

114.

Hernaez R, Lazo M, Bonekamp S, et al. Diagnostic accuracy and reliability of ultrasonography for the detection of fatty liver: a meta-analysis. Hepatology 2011;54:1082

115.

Mottin CC, Moretto M, Padoin AV, et al. The role of ultrasound in the diagnosis of hepatic steatosis in morbidly obese patients. Obes Surg 2004;14(5):635-7

116.

117.

118.

Cho CS, Curran S, Schwartz LH, et al. Preoperative radiographic assessment of hepatic steatosis with histologic correlation. J Am Coll Surg 2008;206(3):480 Kwok R, Tse YK, Wong GL, et al. Systematic review with meta-analysis: non-invasive assessment of non-alcoholic fatty liver disease – the role of transient elastography and plasma cytokeratin-18 fragments. Aliment Pharmacol Ther 2014;39(3):254-69 Shi KQ, Tang JZ, Zhu XL, et al. Controlled attenuation parameter for the detection of steatosis severity in chronic liver disease: a meta-analysis of diagnostic

accuracy. J Gastroenterol Hepatol 2014; 29(6):1149-58 119.

Tang A, Tan J, Sun M, et al. Nonalcoholic fatty liver disease: MR imaging of liver proton density fat fraction to assess hepatic steatosis. Radiology 2013;267(2):422-31

120.

Idilman IS, Aniktar H, Idilman R, et al. Hepatic steatosis: quantification by proton density fat fraction with MR imaging versus liver biopsy. Radiology 2013;267(3):767-75

121.

Schwenzer NF, Springer F, Schraml C, et al. Non-invasive assessment and quantification of liver steatosis by ultrasound, computed tomography and magnetic resonance. J Hepatol 2009;51:433

122.

Neuschwander-Tetri BA, Clark JM, Bass NM, et al. Clinical, laboratory and histological associations in adults with nonalcoholic fatty liver disease. Hepatology 2010;52:913

123.

Musso G, Gambino R, Cassader M, et al. 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):617-49

124.

Kleiner DE, Brunt EM, Van Natta M, et al. Design and validation of a histological scoring system for nonalcoholic fatty liver disease. Hepatology 2005;41:1313

125.

Promrat K, Kleiner DE, Niemeier HM, et al. Randomized controlled trial testing the effects of weight loss on nonalcoholic steatohepatitis. Hepatology 2010;51:121

126.

Andersen T, Gluud C, Franzmann MB, et al. Hepatic effects of dietary weight loss in morbidly obese subjects. J Hepatol 1991;12:224

127.

Lavine JE. Vitamin E treatment of nonalcoholic steatohepatitis in children: a pilot study. J Pediatr 2000;136:734

128.

Sanyal AJ, Chalasani N, Kowdley KV, et al. Pioglitazone, vitamin E, or placebo for nonalcoholic steatohepatitis. N Engl J Med 2010;362:1675-85

129.

Belfort R, Harrison SA, Brown K, et al. A placebo-controlled trial of pioglitazone in subjects with nonalcoholic steatohepatitis. N Engl J Med 2006;355:2297

130.

Aithal GP, Thomas JA, Kaye PV, et al. Randomized, placebo-controlled trial of pioglitazone in nondiabetic subjects with nonalcoholic steatohepatitis. Gastroenterology 2008;135:1176

131.

Ratziu V, Giral P, Jacqueminet S, et al. Rosiglitazone for nonalcoholic steatohepatitis: one-year results of the randomized placebo-controlled Fatty Liver

Expert Rev. Gastroenterol. Hepatol. 9(5), (2015)

Management & diagnosis of fatty liver disease

Improvement with Rosiglitazone Therapy (FLIRT) Trial. Gastroenterology 2008;135:100

Expert Review of Gastroenterology & Hepatology Downloaded from informahealthcare.com by Nanyang Technological University on 04/26/15 For personal use only.

132.

133.

Boettcher E, Csako G, Pucino F, et al. Meta-analysis: pioglitazone improves liver histology and fibrosis in patients with non-alcoholic steatohepatitis. Aliment Pharmacol Ther 2012;35:66 Neumann A, Weill A, Ricordeau P, et al. Pioglitazone and risk of bladder cancer among diabetic patients in France: a population-based cohort study. Diabetologia 2012;55:1953

treatment of nonalcoholic fatty liver disease. Clin Gastroenterol Hepatol 2006;4(5):639 141.

142.

143.

134.

Rakoski MO, Singal AG, Rogers MA, et al. Meta-analysis: insulin sensitizers for the treatment of non-alcoholic steatohepatitis. Aliment Pharmacol Ther 2010;32:1211

135.

Xu F, Li Z, Zheng X, et al. SIRT1 Mediates the Effect of GLP-1 receptor agonist exenatide on ameliorating hepatic steatosis. Diabetes 2014;63(11):3637-46

144.

Bellentani S. Immunomodulating and anti-apoptotic action of ursodeoxycholic acid: where are we and where should we go? Eur J Gastroenterol Hepatol 2005;17:137

145.

136.

137.

138.

Lindor KD, Kowdley KV, Heathcote EJ, et al. Ursodeoxycholic acid for treatment of nonalcoholic steatohepatitis: results of a randomized trial. Hepatology 2004;39:770 Leuschner UF, Lindenthal B, Herrmann G, et al. High-dose ursodeoxycholic acid therapy for nonalcoholic steatohepatitis: a double-blind, randomized, placebo-controlled trial. Hepatology 2010; 52(2):472-9

139.

Pietu F, Guillaud O, Walter T, et al. Ursodeoxycholic acid with vitamin E in patients with nonalcoholic steatohepatitis: long-term results. Clin Res Hepatol Gastroenterol 2012;36:146

140.

Zelber-Sagi S, Kessler A, Brazowsky E, et al. A double-blind randomized placebo-controlled trial of Orlistat for the

informahealthcare.com

146.

147.

Merat S, Malekzadeh R, Sohrabi MR, et al. Probucol in the treatment of non-alcoholic steatohepatitis: a double-blind randomized controlled study. J Hepatol 2003;38(4):414 Abdelmalek MF, Sanderson SO, Angulo P, et al. Betaine for nonalcoholic fatty liver disease: results of a randomized placebo-controlled trial. Hepatology 2009; 50(6):1818 Foster T, Budoff MJ, Saab S, et al. Atorvastatin and antioxidants for the treatment of nonalcoholic fatty liver disease: the St Francis Heart Study randomized clinical trial. Am J Gastroenterol 2011; 106(1):71 Van Wagner LB, Koppe SW, Brunt EM, et al. Pentoxifylline for the treatment of non-alcoholic steatohepatitis: a randomized controlled trial. Ann Hepatol 2011;10(3): 277-86 Chachay VS, Macdonald GA. Martin, JH, et al. Resveratrol does not benefit patientswith non-alcoholic fatty liver disease. Clin Gastroenterol Hepatol 2014;12(12): 2092-103 Sanyal A, Abdelmalek M, Suzuki A, et al. No significant Effects of Ethyl-Eicosapentoic Acid on Histologic Features of Nonalcoholic Steatohepatitis in a Phase 2 Trial. Gastroenterology 2014; 147(2):377-84 Scorletti E, Bhatia L, McCormick KG, et al. Design and rationale of the WELCOME trial: a randomised, placebo controlled study to test the efficacy of purified long chain omega-3 fatty treatment in non-alcoholic fatty liver disease. Contemp Clin Trials 2014;37(2):301-11

148.

Angulo P. NAFLD, obesity, and bariatric surgery. Gastroenterology 2006;130:1848-52

149.

Furuya CK Jr, de Oliveira CP, de Mello ES, et al. Effects of bariatric surgery on

Review

nonalcoholic fatty liver disease: preliminary findings after 2 years. J Gastroenterol Hepatol 2007;22(4):510 150.

Mathurin P, Hollebecque A, Arnalsteen L, et al. Prospective study of the long-term effects of bariatric surgery on liver injury in patients without advanced disease. Gastroenterology 2009;137(2):532-40

151.

Chavez-Tapia NC, Tellez-Avili FI, Barrientos-Gutierrez T, et al. Bariatric surgery for nonalcoholic steatohepatitis in obese patients. Cochrane Database Syst Rev 2010(1):CD007340

152.

Llovet JM, Burroughs A, Bruix J. Hepatocellular carcinoma. Lancet 2003;362: 1907-17

153.

Kansagara D, Papak J, Pasha AS, et al. Screening for hepatocellular carcinoma in chronic liver disease: a systematic review. Ann Intern Med 2014;161(4):261-9

154.

Zhang BH, Yang BH, Tang ZY. Randomized controlled trial of screening for hepatocellular carcinoma. J Cancer Res Clin Oncol 2004;130:417-22

155.

Chen JG, Parkin DM, Chen QG, et al. Screening for liver cancer: results of a randomised controlled trial in Qidong. China. J Med Screen 2003;10:204-9

156.

Bruix J, Sherman M. Management of hepatocellular carcinoma: an update. AASLD Practise Guideline 2011(3):1020-2

157.

Wang YC, McPherson K, Marsh T, et al. Health and economic burden of the projected obesity trends in the USA and the UK. Lancet 2011;378(9793):815-25

158.

McCullough AJ, Dasarathy S. Nonalcoholic Fatty Liver Disease. In: Hawkey CJ, Bosch J, Richter JE, et al. editors. Textbook of Clinical Gastroenterology and Hepatology. John Wiley and Sons, Ltd; Chichester, West Sussex: 2012

683

Management and diagnosis of fatty liver disease.

Nonalcoholic fatty liver disease is a common cause of chronic liver disease and has been an increasingly studied topic of research as the obesity epid...
884KB Sizes 0 Downloads 12 Views