Location via proxy:   [ UP ]  
[Report a bug]   [Manage cookies]                
REVIEWS Nonalcoholic Fatty Liver Disease and Nonalcoholic Steatohepatitis: Selected Practical Issues in Their Evaluation and Management Raj Vuppalanchi and Naga Chalasani Nonalcoholic fatty liver disease (NAFLD) is among the most common causes of chronic liver disease in the western world. It is now recognized that these patients have myriad of important co-morbidities (e.g., diabetes, hypothyroidism and metabolic syndrome). The workup of patients with suspected NAFLD should consist of excluding competing etiologies and systemic evaluation of metabolic comorbidities. NAFLD is histologically categorized into steatosis and steatohepatitis, two states with fairly dichotomous natural history. While significant progress has been made in terms of noninvasively predicting advanced fibrosis, insufficient progress has been made in predicting steatohepatitis. Currently, liver biopsy remains the gold standard for the histological stratification of NAFLD. While sustained weight loss can be effective to treat NASH, it is often difficult to achieve. Foregut bariatric surgery can be quite effective in improving hepatic histology in selected patients without liver failure or significant portal hypertension. Thiazolidinediones have shown promise and the results from the ongoing, large multicenter study should become available soon. Large multicenter studies of CB, receptor anatagonists are also underway but their results will not be available for several years. Several recent studies have highlighted that cardiovascular disease is the single most important cause of morbidity and mortality in this patient population. Conclusion: Health care providers should not only focus on liver disease but also concentrate on aggressively modifying and treating their cardiovascular risk factors. (HEPATOLOGY 2009;49:306-317.) N onalcoholic fatty liver disease (NAFLD) is one of the most common causes of elevated liver enzymes and chronic liver disease in Western 1-3 countries. Its incidence in adults and children is rising rapidly because of ongoing epidemics of obesity and type 2 diabetes.4,5 It is a multifaceted metabolic disorder and is en- Abbreviations: BMI, body mass index; CI, confidence interval; CK-18, cytokeratin 18; HOMA-IR, Homeostatic Model Assessment—Insulin Resistance; NAFLD, nonalcoholic fatty liver disease; NASH, nonalcoholic steatohepatitis; TZD, thiazolidinediones. From the Division of Gastroenterology and Hepatology, Department of Medicine, Indiana University School of Medicine, Indianapolis, IN. Affiliated with Clarian/IU Digestive Disease Center. Received July 25, 2008; accepted July 13, 2008. Supported in part by Public Health Service grant K24 DK072101 (to N.C.). Address reprint requests to: Naga Chalasani, M.D., Professor of Medicine, Indiana University School of Medicine, RG 4100, 1050 Wishard Boulevard, Indianapolis, IN 46202. E-mail: nchalasa@iupui.edu; Fax: 317-278-1949. Copyright © 2008 by the American Association for the Study of Liver Diseases. Published online in Wiley InterScience (www.interscience.wiley.com). DOI 10.1002/hep.22603 Potential conflict of interest: Dr. Chalasani has served as a paid consultant to Atherogenics, Pfizer, Takeda, Advanced Life Sciences, Karo-Bio, Metabasis and Eli-Lilly over the past 12-months. He has agreements to conduct clinical trials related to fatty liver disease with Debiovision, Sanofi-Aventis, Pfizer and Gilead. Dr. Vuppalanchi received grants from Sanofi-Aventis. 306 countered in clinical practice by a variety of health care specialists ranging from primary care physicians and gastroenterologists to cardiologists, radiologists, and gynecologists. It is comprised of a spectrum of liver disease ranging from simple steatosis to full-blown steatohepatitis that is characterized by steatosis, lobular inflammation, ballooning, and fibrosis.6 Over the last several years, much progress has been made in terms of our understanding of its risk factors, pathogenesis, natural history, noninvasive markers, and treatment. This review is tailored toward clinicians caring for patients with NAFLD and discusses practical issues related to selected aspects of its evaluation and management. Evaluation of Newly Suspected NAFLD Suspected NAFLD represents one of the most common reasons why patients visit gastroenterologists and hepatologists in an ambulatory setting.1-3 Although some patients may have attributable abdominal symptoms and tender hepatomegaly, most are asymptomatic, and their liver disease is identified incidentally on routine blood tests or abdominal imaging. Its initial assessment consists of excluding competing and co-existing causes and identifying clinically important comorbidities. HEPATOLOGY, Vol. 49, No. 1, 2009 Competing Causes The diagnosis of NAFLD requires that there is no history of previous or ongoing significant alcohol consumption. There is no consistent agreement regarding the definition of significant alcohol consumption; however, it is generally believed that average alcohol consumption of more than two drinks per day in women and more than three drinks per day in men is necessary to develop alcoholic fatty liver.7,8 However, in individuals with metabolic risk factors such as obesity and diabetes, it is possible that alcohol consumed at lower quantities may promote hepatic steatosis. This notion is supported by a population-based study in which Ruhl and Everhart9 have shown that one or more alcohol drinks per day cause elevated alanine aminotransferase (ALT) in obese but not in normal weight individuals.9 This study is at odds with recent epidemiological data suggesting that modest wine consumption may reduce the prevalence of NAFLD.10 When the alcohol consumption history is insufficient, it becomes difficult to distinguish alcoholic and non-alcoholic forms of fatty liver, especially in those with obesity and associated metabolic risk factors. Conventional markers such as gamma glutamyl transpeptidase, mean corpuscular volume, and aspartate aminotransferase (AST)/ALT ratio are not useful, and specific serum markers for chronic alcohol abuse are of limited utility.11 The carbohydrate-deficient transferrin is the most widely used and perhaps the most specific serum marker for detecting chronic alcohol abuse.12 This test evaluates the ratio of desialylated transferrin to total transferrin, and it has 81% sensitivity with 98% specificity in detecting chronic alcohol abuse. Unfortunately, this and other tests such as the ratio of mitochondrial AST to total AST13 are not readily available in clinical practice. More recently, investigators from the Mayo clinic have developed the “alcoholic liver disease/NAFLD index,” which consisted of five easily available variables: mean corpuscular volume, AST, and ALT values, height and weight, and sex (http://www.mayoclinic.org/gi-rst/ mayomodel10.html).14 This weighted multivariate model uses logistic regression analysis to generate the alcoholic liver disease/NAFLD index score, and a score greater than 0 incrementally favors alcoholic liver disease, whereas a value less than 0 favors NAFLD in decrement. For example, an alcoholic liver disease/NAFLD index of 8.95 would correspond to greater than 99% probability of alcoholic liver disease, whereas a value of ⫺5.04 would correspond to greater than 99% probability of NAFLD. Because most subjects with alcoholic liver disease in this study had high model for end-stage liver disease scores, this tool is more likely applicable to patients with decom- VUPPALANCHI AND CHALASANI 307 pensated cirrhosis presenting for liver transplant evaluation rather than outpatients with fatty liver. Elevated serum auto-antibodies are common in patients with NAFLD. Although low titer antinuclear antibody positivity can be seen in up to 33% of patients with NAFLD, antinuclear antibody positivity in titers greater than 1:320 is generally rare.15-17 Low titers of anti-smooth muscle and antimitochondrial antibodies also may be noted in patients with NAFLD.18 The presence of these auto-antibodies is generally thought to be an epiphenomenon, although in one study their presence correlated with more severe histological damage.19,20 In patients with suspected NAFLD and antinuclear antibody positivity at titers greater than 1:160 or anti-smooth muscle actin positivity at titers greater 1:40, a liver biopsy may be considered to exclude the presence of autoimmune hepatitis. Mildly increased serum ferritin is not uncommon in patients with NAFLD, and it does not necessarily indicate co-existing iron overload.21 Metabolic syndrome and hyperinsulinemia are known to be associated with increased serum ferritin, and this association may be mediated by the presence of NAFLD.22 Nonetheless, elevated serum ferritin in a patient with suspect NAFLD should prompt testing for hereditary hemochromatosis (HFE gene) mutations. The prevalence of HFE gene mutations in NAFLD patients has been variable, depending on the population studied, and their relevance remains largely unknown.23,24 Although the prevalence of HFE gene mutations and their clinical relevance remain unclear,22-24 homozygote or heterozygote C282Y mutations along with elevated serum ferritin may justify a liver biopsy in patients with suspected NAFLD. It is important to verify that chronic hepatitis B and hepatitis C have been convincingly excluded, and, depending on the clinical scenario, rare disorders such as alpha-1 antitrypsin deficiency and Wilson’s disease should also be excluded. Thorough medication history is important because several commonly prescribed medications (such as tamoxifen, methotrexate, and amiodarone) are noted for their ability to cause hepatic steatosis.1 Evaluation for Co-morbidities The patients with NAFLD frequently have many clinically significant co-morbidities (Table 1). Obesity, type 2 diabetes, and hyperlipidemia are well known to co-exist in patients with NAFLD, and it is important to systematically characterize them. The body mass index (BMI) and waist circumference should be measured to better characterize the degree (mild, moderate, and severe) and the nature (central versus peripheral) of obesity. Type 2 diabetes and glucose intolerance are very frequent in patients 308 VUPPALANCHI AND CHALASANI HEPATOLOGY, January 2009 Table 1. Comorbidities Commonly Associated with NAFLD Established Conditions Emerging Conditions Obesity Type 2 diabetes/glucose intolerance Dyslipidemia Metabolic syndrome Obstructive sleep apnea Hypothyroidism Obstructive sleep apnea Hypopituitarism with NAFLD, and they have prognostic significance.25 In patients without preexisting type 2 diabetes, the presence of glucose intolerance and insulin resistance should be evaluated by obtaining fasting blood glucose, insulin levels and hemoglobin A1c. In patients without diabetes, insulin resistance should be assessed by calculating HOMA-IR (homeostasis model assessment—insulin resistance) or QUICKI (quantitative insulin-sensitivity check index).26,27 Both HOMA-IR and QUICKI are mathematical transformations of fasting blood glucose and insulin levels. In our practice, we calculate HOMA-IR ([fasting insulin {␮U/mL} * fasting glucose {mmol/L}]/22.5), and as a general guideline, we consider an HOMA-IR value greater than 3 to be clinically significant.28 If not done recently, fasting lipid profile should be obtained, because dyslipidemia is nearly universal. Typically, fasting serum triglyceride levels are high, along with low high-density lipoprotein values. Recent data suggest that high-sensitivity c-reactive protein is frequently elevated in patients with NAFLD and should be measured along with fasting lipid profile to assess cardiovascular risk (vide infra).29,30 In addition, patients should be systematically explored for the presence of other comorbidities such as obstructive sleep apnea,31,32 hypopituitarism,33 hypothyroidism,34 and polycystic ovary syndrome.35-37 Obstructive sleep apnea is commonly present in patients with NAFLD and may contribute to the fatigue that these patients sometimes experience.38 Some studies have suggested a causal relationship between obstructive sleep apnea and the pathogenesis of NAFLD, but these are far from convincing.39,40 Previous studies have shown that patients with hypothyroidism and hypopituitarism have increased prevalence of NAFLD.33,34 Several studies have shown high prevalence of NAFLD in premenopausal women with polycystic ovary syndrome,35,37 and one study36 suggested that such individuals are at higher risk for disease progression. Identification of Nonalcoholic Steatohepatitis in Patients with NAFLD The NAFLD can be categorized into simple steatosis and steatohepatitis (NASH). A well-defined case of NASH histologically exhibits macrovesicular steatosis, lobular inflammation, balloon degeneration of hepatocytes, and zone 3 pericellular fibrosis.41 Undoubtedly, NASH is histologically progressive and can lead to cirrhosis and associated liver dysfunction. Simple steatosis has a relatively benign course but is not totally without histological consequences.42 For example, investigators from the Cleveland Clinic reported in their initial paper that 4% of patients with simple steatosis developed cirrhosis and 2% had liver-related mortality over a median follow-up of approximately 8 years.6 The extended follow-up of the same cohort was reported in an abstract form recently, and it confirmed that simple steatosis is not entirely benign.43 Liver biopsy is the current gold standard to identify steatohepatitis, but it is not without controversies and practical difficulties. Several recent studies have highlighted its sampling variability and interobserver discordance.44-46 The precise histological definition of NASH is not entirely known, and the experts believe that it should be based on pattern recognition rather than a composite score of individual components such as steatosis, ballooning, and fibrosis.47 It remains controversial whether ballooning or pericellular fibrosis should be considered as critical for the histological diagnosis of NASH. The recently described NAFLD Activity Score is valuable for quantifying histological changes, especially in clinical trials,47 but its generalizability and diagnostic utility are unknown. Over the past several years, there has been significant interest in noninvasively predicting liver histology in patients with NAFLD. However, it remains somewhat controversial which histological finding(s) should be targeted for noninvasive assessment. Most studies have attempted to predict advanced fibrosis (bridging fibrosis/cirrhosis),25,48,49 but it has been argued that one should attempt to predict steatohepatitis rather than advanced fibrosis.50 It is our view that future studies should attempt to predict three histological states (simple steatosis, NASH without significant fibrosis, and NASH with advanced fibrosis), rather than dichotomous descriptive states. Aminotransferase values and common imaging tests such as liver ultrasound, computed tomography, and magnetic resonance are of limited value in predicting liver histology. Numerous circulating biomarkers and prediction models have been investigated to noninvasively predict hepatic histology in patients with NAFLD (Table 2), and their full discussion is beyond the scope of this review. However, cytokeratin 18 (CK-18) fragments and serum dehydroepiandrosterone, two hypothesis-driven circulating biomarkers, deserve further discussion. Based on experimental data that hepatocyte apoptosis may play an important role in the pathogenesis of NAFLD, Wieck- HEPATOLOGY, Vol. 49, No. 1, 2009 VUPPALANCHI AND CHALASANI 309 Table 2. Noninvasive Biomarkers Previously Studied or Currently Under Evaluation Serum Biomarkers Fibrosis Oxidative stress Hepatocyte apoptosis Inflammation Imaging Studies Breath Tests 58 BARD score Fibrotest NAFLD fibrosis score56 ELF/OELF57,59 Hyaluronic acid130 TBARS119 Oxidized-LDL119 Total antioxidant response137 Total lipid peroxide levels137 CK-18 fragments51,53 TNF-␣/adiponectin ratio138,139 hsCRP29 IL-6140 CC-chemokine ligand-2141 owska et al.51 measured plasma CK-18 fragment levels in 44 consecutive individuals with suspected NAFLD undergoing liver biopsy.51 CK-18 is a major intermediate filament protein in hepatocytes, and it is cleaved by the effector caspases (mainly caspase 3) on activation of the apoptosis cascade.52,53 Compared with individuals with normal histology and those without NASH, patients with definite NASH had significantly higher CK-18 fragment levels.54 It was suggested that CK-18 fragment levels greater than 380.2 U/L can predict definite NASH in a very precise fashion.51 A puzzling and unexplained aspect of this small cross-sectional study is that nearly 25% of patients with suspected NAFLD supposedly had normal hepatic histology on liver biopsy. More recently, Charlton et al.55 have shown that serum dehydroepiandrosterone levels had a consistent and stepwise inverse relationship with the degree of hepatic fibrosis that persisted after adjusting for age.55 This observation, which was initially made on a derivation cohort consisting of 122 patients, was subsequently reproduced on a validation cohort consisting of 361 NAFLD patients recruited from two separate academic centers.55 These provocative preliminary data deserve further study, but it may be too optimistic to assume that a single biomarker can reliably predict histology in NAFLD, a condition with relatively complex phenotype and multiple comorbidities. There are numerous papers published in the literature describing noninvasive prediction models, but most of them consisted of small sample size and lacked rigorous external validation. Three recently described models with relatively large sample size and some level of validation have shown encouraging results.56-58 In a multicenter study consisting of 480 patients in the derivation and 253 in the validation cohorts, Angulo et al.56 have shown that a NAFLD fibrosis score consisting of six variables (age, BMI, AST/ALT ratio, hyperglycemia, platelet count, and 61,131 Transient elastography MR elastography132 MR spectroscopy133,134 [13C] methacetin135 [13C] ketoisocaproate135 Ethanol136 Acetone136 albumin) can reliably predict advanced fibrosis. The formula for the fibrosis score was “ ⫺1.675 ⫹ 0.037 ⫻ age (years) ⫹ 0.094 ⫻ BMI (kg/m2) ⫹ 1.13 ⫻ impaired glucose tolerance/diabetes (yes ⫽ 1, no ⫽ 0) ⫹ 0.99 ⫻ AST/ALT ratio ⫺ 0.013 ⫻ platelets (⫻109/L) ⫺0.66 ⫻ albumin (g/dL),” and a low cutoff point (score ⬍ ⫺1.455) signified the absence of advanced fibrosis, whereas a high cutoff point (score ⬎ 0.676) identified advanced fibrosis. It was concluded that NAFLD fibrosis score can be used as a triaging tool for optimizing liver biopsy yield in terms of identifying or excluding advanced fibrosis. More recently, Guha et al.57 have compared the performance of “enhanced liver fibrosis” (ELF) score with that of the “NAFLD fibrosis score” in predicting advanced fibrosis. The enhanced liver fibrosis is essentially the “original European liver fibrosis” test without “age” included in the algorithm. The original European liver fibrosis test, a model consisting of age and three serum markers of matrix turnover (tissue inhibitor of metalloproteinases, hyaluronic acid, and type III procollagen), predicted advanced fibrosis in a variety of liver disorders.59 In this report, the ELF score had excellent ability to predict different levels of fibrosis (any fibrosis, moderate fibrosis, or advanced fibrosis), and when it was combined with the “NAFLD fibrosis score,” its ability to predict different levels of fibrosis improved further.57 Harrison et al.58 have recently developed the BARD score to predict advanced fibrosis in patients with NAFLD.57 The BARD score is a weighted sum of three easily available variables (BMI ⬎ 28 kg/m2 [1 point], AST/ALT ⱖ 0.8 [2 points], and diabetes [1 point]), and the authors have shown that a score of 2 to 4 was associated with an odds ratio of 17 (95% confidence interval [CI]: 9.2-31.9) for predicting advanced fibrosis.57 These promising models will need to be validated by external investigators before they are recommended for wide clinical use. 310 VUPPALANCHI AND CHALASANI One French study tested “Fibrotest” for predicting advanced fibrosis in NAFLD and found encouraging results.60 “Fibrotest” is a popular serum test for predicting advanced fibrosis in individuals with chronic hepatitis C, but more studies are needed to test its utility in NAFLD. Similarly, liver stiffness measured by transient elastography (FibroScan) may have some role in predicting the degree of fibrosis, but the data are sparse in terms of its utility in NAFLD.61,62 In summary, there has been significant research in developing biomarkers of liver histology in patients with NAFLD, and in fact one editorialist recently opined that the future is around the corner for noninvasively diagnosing progressive NASH.63 Although this may be the case for advanced fibrosis, insufficient attention has been paid to developing markers for noninvasively identifying steatohepatitis in patients with NAFLD. Until more definite data with external validation become available, we will continue with our current practice of recommending liver biopsy to selected patients with suspected NAFLD based on the presence of certain risk factors such as older age, diabetes, severe obesity, and metabolic syndrome. Management of Patients with NAFLD Advice Regarding Weight Loss, Exercise, and Specific Diets. The NAFLD is largely a manifestation of obesity and metabolic syndrome and is characterized by excess calorie intake and lack of optimal health-related fitness or physical activity.64 It is generally believed that weight loss is beneficial for patients with NAFLD, but data are sparse in terms of specifics such as how, how much, and how rapidly to lose weight. Furthermore, precise hepatic and extrahepatic benefits of weight loss are not well defined. In a recent review, Bellentani et al.65 pointed out that there are only four human studies consisting of fewer than 40 total patients that evaluated the effect of calorie restriction alone, and change in liver enzymes was the primary end point in all but one study. There are 10 published studies consisting of 626 total patients that evaluated the effect of calorie restriction combined with exercise, but liver histology was the primary end point in only four studies (123 patients).65 This paucity of data makes it difficult to make evidence-based recommendations about dietary modification and exercise to treat NAFLD and NASH. It is generally recommended that overweight and obese patients with NAFLD lose 7% to 10% of their body weight by dietary modification and exercise over the course of 6 to 12 months. This is based on short-term studies showing that gradual weight loss of this magnitude improves insulin resistance and hepatic histology.3,66 HEPATOLOGY, January 2009 Table 3. Strategies to Enhance Patient Compliance in Lifestyle Modification ● ● ● ● ● ● ● ● Communicate with empathy Be sensitive to general stigma against obesity Discuss pros and cons of proposed changes to lifestyle Explore reasons for perpetual poor dietary and exercise choices Encourage self-efficacy Offer specific choices of food and exercise Design individualized program of eating and physical activity Explain treatment and its benefits Reproduced, with modification, from Bellentani et al., Hepatology 2008;47: 746-754. Our recommendations to enhance patient compliance with lifestyle modifications are shown in Table 3. Scientific evidence is lacking to make precise recommendations specific to modifying macronutrient composition, but it appears sensible to recommend low glycemic food with decreased saturated and trans-fat intake but increased mono and polyunsaturated fatty acid intake.67 Evolving data suggest diets consisting of high fructose should be avoided by these patients.68 Because of the lack of safety and efficacy data, popular weight-loss diets such as Atkins, Ornish, and South Beach diets should not be recommended.67 In our clinical practice, we recommend diminished portions of balanced diet (consisting of a lowglycemic and low-fat diet and increased portions of fruits and vegetables) and five to seven sessions per week of moderate aerobic exercise, with each session lasting for 30 to 45 minutes. However, such prescriptive recommendations made in a clinic setting are rarely effective both short-term and long-term.65 Orlistat, a reversible inhibitor of gastric and pancreatic lipase, may be effective in promoting limited weight loss in selected patients, but side effects such as diarrhea and bloating make it less desirable. In a randomized study, Harrison et al.69 have shown that orlistat does not cause weight loss or histological improvement above and beyond that accomplished with calorie restriction alone.69 Rimonabant (endocannabinoid receptor 1 antagonist) is approved in Europe for promoting weight loss, and it may have favorable anti-steatotic and anti-fibrotic properties.70-74 Large clinical trials are underway with this and other similar compounds in NASH, but their results will not become available for at least several years. Advice Regarding Bariatric Surgery. Because many patients with NAFLD have severe obesity, it often comes up in clinical practice whether bariatric surgery is suitable in this patient population. Jejunoileal bypass was widely popular in mid-1950s to mid-1970s for the surgical treatment of obesity, but because of disastrous hepatic and extrahepatic consequences it is now totally abandoned.75-77 Over the last decade, an increasing number of HEPATOLOGY, Vol. 49, No. 1, 2009 VUPPALANCHI AND CHALASANI 311 Table 4. Selected Studies of Metformin in Patients with NAFLD N Design Comparator Population Duration Liver Enzymes Histology Marchesini et al. 14 Open label Single arm None 4 mo Improved Not evaluated Nair et al.143 15 None 12 mo Improved Improved inflammation Uygun et al.144 36 Open label Single arm Open label Adults Mostly nondiabetic Nondiabetics Nondiabetics 6 mo Improved Improved inflammation Bugianesi et al.145 55 Randomized clinical trial Non-diabetics 12 mo Improved Schwimmer et al.146 10 None Nondiabetics 6 mo Improved Loomba et al.147 14 None Nondiabetics 48 wks Improved Nobili et al.148 57 Open label Single arm Open label Single arm Open label Antioxidant Nondiabetics 24 months No difference Author (reference) 142 Calorie-restricted diet Calorie restricted diet foregut bariatric surgery procedures are being performed to treat obesity and its complications, and its short-term and long-term benefits are becoming well established.78-80 The commonly performed foregut bariatric surgery procedures include roux-en-Y gastric bypass (most common), adjustable gastric banding, gastroplasty, and sleeve gastrectomy.81,82 There have been no studies that evaluated foregut bariatric surgery to specifically treat NAFLD, but many published papers described its favorable effect of hepatic histology when performed for other indications, thus introducing selection bias.78-80 In general, liver histology improves significantly after foregut bariatric surgery with very minimal risk of worsening.83,84 In a recent meta-analysis consisting of 15 studies and 766 paired liver biopsies, Mummadi et al.84 have shown that all components of NAFLD show significant improvement after foregut bariatric surgery. Pooled proportion of patients with improvement or resolution in steatosis was 93% (95% CI: 84%-98%), and improvement or resolution of steatohepatitis was 82% (95% CI: 64%-95%); improvement in fibrosis when assessed using needle biopsies was 73% (95% CI: 65%-81%). We speculate that liver disease is unlikely to worsen in association with rapid and profound weight loss unless there are additional risks such as bacterial overgrowth (for example, jejunoileal bypass) or nutrient depletion (such as kwashiorkor). This forms the basis for our view that very long roux limb (in other words, ⬎150 cm) should be avoided in patients with advanced fibrosis. Compensated cirrhosis is not a contraindication for foregut bariatric surgery provided it is performed by an experienced surgeon and clinically evident portal hypertension is absent (no esophageal or abdominal varices). There are reports that cirrhosis may reverse after bariatric surgery.85,86 In our practice, we recommend foregut bariatric surgery as a therapeutic possibility for the severely obese Improved steatosis, inflammation and fibrosis Not evaluated Improved steatosis and inflammation No difference NAFLD patients with advanced fibrosis who failed to lose weight despite repeated nutritional counseling. In those with cirrhosis, we exclude clinical portal hypertension by performing abdominal imaging and upper endoscopy. Bariatric surgery may be particularly attractive for carefully selected patients with Child’s A cirrhosis not only because it may stabilize or improve the liver disease but it also may enhance their future suitability for liver transplantation. Role of Insulin Sensitizers. Because insulin resistance is nearly universal in patients with NASH, it is not surprising that many studies tested insulin sensitizers as its treatment. However, a large number of them are proofof-concept studies with small numbers of patients without rigorous study design, making it difficult to make definite recommendations. Biguanides (metformin) and thiazolidinediones (pioglitazone and rosiglitazone) are the two classes of insulin sensitizers studied in humans. Metformin. Although its exact mechanism of action is not entirely clear, metformin’s therapeutic benefit as an antidiabetic agent and insulin sensitizer is well recognized. Its anti-diabetic action is likely related to decreased hepatic gluconeogenesis, decreased glucose absorption, and increased insulin sensitivity by facilitating glucose uptake and utilization.86,87 In addition, its stimulatory effect on adenosine monophosphate–activated protein kinase or modulation of hepatic tumor necrosis factor alpha (TNF-␣) expression may result in benefits.88,89 A summary of studies evaluating metformin to treat NASH is shown in Table 4. A recent meta-analysis published in Cochrane database showed that metformin leads to normalization of serum aminotransferases in a significantly greater proportion of patients compared with dietary modification (odds ratio: 2.83; 95% CI: 1.27-6.31) and improved steatosis by imaging (odds ratio: 5.25, 95% CI: 1.09-25.21).90 The total number of patients treated with 312 VUPPALANCHI AND CHALASANI HEPATOLOGY, January 2009 Table 5. Randomized Controlled Trials of TZDs Consisting of at Least 50 Randomized Patients Histological Improvement, Pre- and Post- First Author (Reference) N Study Design Duration 102 Belfort 55 6 months Ratziu103 63 Hypocaloric diet ⫹ Pioglitazone versus Hypocaloric diet ⫹ placebo *Patients with diabetes ⫹ patients without diabetes Rosiglitazone versus placebo * Patients with diabetes ⫹ patients without diabetes Guruprasad104 74 Diet ⫹ exercise ⫹ Pioglitazone versus Diet ⫹ exercise ⫹ Pioglitazone *Only patients without diabetes 12 months 12 months TZD Versus Placebo Mean Weight Gain No Yes No No No No No No No No Yes Yes Yes Borderline Yes Yes No No No No 2.5 kg TZD group had significant increase in serum adiponectin levels, and this inversely correlated with improved hepatic steatosis 1.5 kg Yes No Worse No N/A No No Yes No N/A - Primary end point was ⬎30% reduction in hepatic steatosis and predictors of response included rosiglitazone treatment, lack of diabetes at entry, and greater steatosis at baseline - No active lifestyle modification in either groups - Ranked assessment of pretreatment and posttreatment biopsies showed improved steatosis, ballooning, and fibrosis in the rosiglitazone group - Relatively lower NAS at entry may at least in part explain why no change in NAS was seen in this study NAS score reported. Placebo group had mean weight loss of 3.5 kg but TZD group had mean weight gain of 2.6 kg Histological Variable TZD Placebo Steatosis Inflammation Ballooning Fibrosis NAS Steatosis Inflammation Ballooning Fibrosis NAS Yes Yes Yes Yes Yes Yes No No No No Steatosis Inflammation Ballooning Fibrosis NAS Yes Yes Yes Yes N/A metformin in controlled studies is admittedly small, and its favorable effect on hepatic histology may not be robust, but we favor its use in patients without diabetes with NASH because of its safety profile. Because most patients without diabetes with NASH have glucose intolerance, it has the added benefit of lowering the risk of developing frank diabetes.91 Because metformin has not been studied to treat NASH in individuals with diabetes, its role in the diabetic population is not known. An ongoing, multicenter study comparing metformin with vitamin E or placebo in pediatric patients with NASH (TONIC; NCT00063635) should provide more insight into metformin’s role in treating NASH. Thiazolidinediones. Thiazolidinediones (TZDs) are a novel class of oral antidiabetic medications that improve insulin resistance by acting as selective peroxisome proliferator-activated receptor gamma agonists.92,93 Troglitazone, the first generation TZD, has been withdrawn from the market because of its hepatotoxicity,94 whereas rosiglitazone and pioglitazone are the second-generation TZDs that are currently available for clinical use.93,95 They redistribute fat from muscle and liver to adipose tissue and thereby improving peripheral (skeletal muscle) and he- 2.6 kg Comments patic insulin sensitivity.93 In addition, they increase circulating levels of adiponectin, which is produced exclusively by the adipose tissue and has insulin-sensitizing properties.96 There has been significant interest in evaluating TZDs to treat NASH, and to our knowledge, eight studies have been published either as full-length papers or solely as an abstract.97-104 Troglitazone was tested in one study,97 rosiglitazone in two,98,103 and pioglitazone in five studies.99-102,104 Four were randomized controlled studies with 213 total enrolled patients with histologically proven NASH.101-104 Selected characteristics and outcomes of three studies that randomized at least 50 patients are shown in Table 5. In general, TZDs improve hepatic histology in patients with NASH, although their favorable effect on steatosis is more striking than on other histological variables such as inflammation, ballooning, or fibrosis. Their favorable effect on liver histology and liver biochemistries disappears on their discontinuation, suggesting that long-term treatment is needed to maintain their therapeutic benefits.105 This is potentially a significant issue; recent studies have questioned the long-term HEPATOLOGY, Vol. 49, No. 1, 2009 VUPPALANCHI AND CHALASANI Table 6. Various Agents in Evaluation to Treat NASH in Humans (www. Clinicaltrials.gov) Agent Nature of the Study Metformin versus vitamin E versus placebo (TONIC) Pioglitazone versus metformin versus placebo (PIVENS) Omega-3 fatty acids Polyunsaturated fatty acids Orlistat Phase III, multicenter, RCT Silophus Silymarin Rimonabant Recombinant leptin Pentoxifylline Pentoxifylline vs. Pioglitazone Exenatide Rosi versus Rosi ⫹ metformin versus rosi ⫹ losartan SAMe Iron depletion CP 945598 TRO 19622* ASP9831† L-Alanine 313 Based on promising animal data107 and its ability to promote weight loss,108 we have initiated an open-label study of exenatide in 2006, but its recruitment has been hampered because of its injectable route of administration and potential gastrointestinal system side effects. Phase III, multicenter, RCT Phase II, pilot study Phase II, pilot study Phase III, multicenter study Phase II, pilot study Phase II, multicenter RCT Phase IIb, multicenter RCT Phase II, pilot study Phase II, pilot study Phase II, pilot study Phase II, pilot study Phase III, single center Phase II, pilot study Phase II, pilot study Phase IIb, multicenter RCT Phase II, pilot study Phase II, pilot study Phase II, pilot study *Cholesterol-like small molecule, a potent neuroprotective agent in vitro. May have mitochondrial protective effects. †Novel PDE4 inhibitor, which may improve hepatic inflammation and fibrosis. safety of TZDs (especially rosiglitazone).106 Because most of the participants in these studies did not have diabetes, it is not clear whether TZDs are equally effective in patients with diabetes with NASH. In fact, the presence of diabetes was a negative predictor of response to rosiglitazone in one study.103 Furthermore, Ratziu et al.103 recently raised the possibility that TZDs alone without lifestyle modification may not be as effective.103 Overall, there are more questions than answers about the role of TZDs in patients with NASH, and the ongoing large U.S. multicenter study (PIVENS; NCT00063622) may provide some additional insight. Promising Agents There is intense research into developing suitable treatment for NASH, and a list of compounds that are being tested to treat NASH in humans is shown in Table 6. Oral endocannabinoid receptor antagonists are of potential benefit because of multiple potential favorable effects (on body weight, fibrogenesis, and de novo lipogenesis), and large multicenter studies are underway. Neuropsychiatric side effects are of concern, but if proven effective, they may have a role at least in a select group of patients without underlying neuropsychiatric co-morbidities. The CB-1 receptor antagonist studies in humans have just begun and their results will not be available for few years. Cardiovascular Disease in NAFLD The cardiovascular morbidity and mortality is perhaps one of the most important aspects of NAFLD and NASH, and our knowledge of their association is evolving rapidly.109-112 The patients with NAFLD have very high prevalence of cardiovascular risk factors and atherosclerosis and high incidence of cardiovascular morbidity and mortality.43,113,114 Over the last decade, numerous studies have demonstrated that patients with NAFLD are enriched with classic cardiovascular risk factors such as obesity, insulin resistance, type 2 diabetes, dyslipidemia, and the metabolic syndrome.115-117 Cross-sectional studies consisting of control groups have shown increased prevalence of endothelial dysfunction,118 elevated levels of oxidized low-density lipoprotein,119 and Framingham coronary risk scores109,119 in NAFLD patients. Cross-sectional studies have also shown increased prevalence of premature atheroma formation,120 carotid artery intima media thickness (surrogate for atherosclerosis),121,122 vulnerable coronary plaques,123 increased mediastinal fat, and abnormal left ventricular energy metabolism.124 Recently it has been suggested that NAFLD poses cardiovascular risk above and beyond that conferred by the presence of the metabolic syndrome.43,113,114,125 In a nested case-control, prospective study, Targher et al. have shown that NAFLD in individuals with diabetes is associated with moderately increased risk of incident cardiovascular disease even after adjusting for classic risk factors, glycemic control and the metabolic syndrome.125 Most importantly, several longitudinal studies have shown that cardiovascular disease is much more common than liver disease as a cause of death in patients with NAFLD.43,113,114 In a longitudinal study consisting of 420 Olmsted county residents with NAFLD, Adams et al.113 have shown that ischemic heart disease accounts for 25% of deaths, compared with liver disease accounting for 13% of deaths. By linking the Third National Health and Nutrition Examination Survey to linked mortality files, Ong et al.114 have shown that cardiovascular disease is the most common cause of death, exceeding liver disease among 817 individuals with suspected NAFLD in comparison with 10,468 persons without liver disease. More recently, Rafiq et al.43 have reported extended follow-up of an expanded NAFLD cohort that has been described previously. The mortality rate over an 11.1-year median follow-up (longest follow-up, 28.5 years) was 314 VUPPALANCHI AND CHALASANI 45%, and the most common cause of death was coronary artery disease. All these data provide unequivocal evidence that coronary artery disease is a serious threat to patients with NAFLD. Therefore, it has become our practice to emphasize the significance of cardiovascular disease to patients with NAFLD and their primary care providers. Statins remain a cornerstone for managing dyslipidemia and coronary artery disease. Despite initial concerns, several recent studies have shown that statins can be safely used in patients with underlying liver disease.126-129 Studies have not been conducted to specifically show that statins diminish cardiovascular morbidity and mortality in patients with NAFLD; however, there are no suspected reasons why they would be any less effective. Minor fluctuations in aminotransferases on initiating statin therapy are not uncommon, but serious hepatotoxicity is quite rare,126 and even when happens, it is almost universally reversible on prompt recognition and withdrawal of the offending agent.127 In summary, when a patient with suspected NAFLD is seen in the clinic, it is important to carefully evaluate for competing causes and clinically important comorbidities. Many advances have been made in terms of noninvasive biomarkers for predicting advanced fibrosis, but insufficient attention has been paid to predicting steatohepatitis. Sustained weight loss can be effective to treat NASH but it is difficult to achieve. Foregut bariatric surgery can be quite effective in improving hepatic histology in selected patients without liver failure or significant portal hypertension. TZDs have shown promise; however, recent studies raised doubts about their long-term safety. Large multicenter studies of endocannabinoid receptor antagonists are underway, but their results will not be available for several years. Several recommendations made in this review are not entirely evidence-based and thus should be cautiously accepted while we await more data and practice guidelines by the consensus of the experts. There is an ongoing effort to develop a multisociety (American Association for the Study of Liver Diseases, American College of Gastroenterology, American College of Physicians, and American Gastroenterological Association) consensus practice guideline for managing patients with NAFLD, and this would clearly identify the level of evidence available for different recommendations. Note Added in Proofs In the fall 2008, Sanofi-Aventis has terminated its multinational clinical trials of rimonabant (CB1 receptor antagonist) to treat NASH due to safety concerns. Similarly, Pfizer also stopped its CB1 receptor antagonist development program. References 1. Angulo P. Nonalcoholic fatty liver disease. N Engl J Med 2002;346: 1221-1231. HEPATOLOGY, January 2009 2. Clark JM, Brancati FL, Diehl AM. The prevalence and etiology of elevated aminotransferase levels in the United States. Am J Gastroenterol 2003;98:960-967. 3. Torres DM, Harrison SA. Diagnosis and therapy of nonalcoholic steatohepatitis. Gastroenterology 2008;134:1682-1698. 4. Fraser A, Longnecker MP, Lawlor DA. Prevalence of elevated alanine aminotransferase among US adolescents and associated factors: NHANES 1999-2004. Gastroenterology 2007;133:1814-1820. 5. Charlton M. Nonalcoholic fatty liver disease: a review of current understanding and future impact. Clin Gastroenterol Hepatol 2004;2:1048-1058. 6. Matteoni CA, Younossi ZM, Gramlich T, Boparai N, Liu YC, McCullough AJ. Nonalcoholic fatty liver disease: a spectrum of clinical and pathological severity. Gastroenterology 1999;116:1413-1419. 7. McCullough AJ, O’Connor JF. Alcoholic liver disease: proposed recommendations for the American College of Gastroenterology. Am J Gastroenterol 1998;93:2022-2036. 8. Coates RA, Halliday ML, Rankin JG, Feinman SV, Fisher MM. Risk of fatty infiltration or cirrhosis of the liver in relation to ethanol consumption: a case-control study. Clin Invest Med 1986;9:26-32. 9. Ruhl CE, Everhart JE. Joint effects of body weight and alcohol on elevated serum alanine aminotransferase in the United States population. Clin Gastroenterol Hepatol 2005;3:1260-1268. 10. Dunn W, Xu R, Schwimmer JB. Modest wine drinking and decreased prevalence of suspected nonalcoholic fatty liver disease. HEPATOLOGY 2008;47:1947-1954. 11. Conigrave KM, Degenhardt LJ, Whitfield JB, Saunders JB, Helander A, Tabakoff B. CDT, GGT, and AST as markers of alcohol use: the WHO/ ISBRA collaborative project. Alcohol Clin Exp Res 2002;26:332-339. 12. Anton RF, Dominick C, Bigelow M, Westby C. Comparison of Bio-Rad %CDT TIA and CDTect as laboratory markers of heavy alcohol use and their relationships with gamma-glutamyltransferase. Clin Chem 2001; 47:1769-1775. 13. Nalpas B, Vassault A, Poupon RE, Pol S, Berthelot P. An overview of serum mitochondrial aspartate aminotransferase (mAST) activity as a marker of chronic alcohol abuse. Alcohol Alcohol Suppl 1991;1:455-457. 14. Dunn W, Angulo P, Sanderson S, Jamil LH, Stadheim L, Rosen C, et al. Utility of a new model to diagnose an alcohol basis for steatohepatitis. Gastroenterology 2006;131:1057-1063. 15. Cotler SJ, Kanji K, Keshavarzian A, Jensen DM, Jakate S. Prevalence and significance of autoantibodies in patients with non-alcoholic steatohepatitis. J Clin Gastroenterol 2004;38:801-804. 16. Cho DH, Choi MS, Kim DH, Kim DY, Shim SG, Lee JH, et al. [A prospective study on the prevalence and clinical significance of autoantibodies in patients with suspected nonalcoholic fatty liver disease]. Korean J Hepatol 2005;11:261-267. 17. Yatsuji S, Hashimoto E, Kaneda H, Taniai M, Tokushige K, Shiratori K. Diagnosing autoimmune hepatitis in nonalcoholic fatty liver disease: is the International Autoimmune Hepatitis Group scoring system useful? J Gastroenterol 2005;40:1130-1138. 18. Loria P, Lonardo A, Leonardi F, Fontana C, Carulli L, Verrone AM, et al. Non-organ-specific autoantibodies in nonalcoholic fatty liver disease: prevalence and correlates. Dig Dis Sci 2003;48:2173-2181. 19. Niwa H, Sasaki M, Haratake J, Kasai T, Katayanagi K, Kurumaya H, et al. Clinicopathological significance of antinuclear antibodies in non-alcoholic steatohepatitis. Hepatol Res 2007;37:923-931. 20. Adams LA, Lindor KD, Angulo P. The prevalence of autoantibodies and autoimmune hepatitis in patients with nonalcoholic Fatty liver disease. Am J Gastroenterol 2004;99:1316-1320. 21. Bacon BR, Farahvash MJ, Janney CG, Neuschwander-Tetri BA. Nonalcoholic steatohepatitis: an expanded clinical entity. Gastroenterology 1994;107:1103-1109. 22. Fargion S, Mattioli M, Fracanzani AL, Sampietro M, Tavazzi D, Fociani P, et al. Hyperferritinemia, iron overload, and multiple metabolic alterations identify patients at risk for nonalcoholic steatohepatitis. Am J Gastroenterol 2001;96:2448-2455. 23. Lin TJ, Lin CL, Wang CS, Liu SO, Liao LY. Prevalence of HFE mutations and relation to serum iron status in patients with chronic hepatitis C HEPATOLOGY, Vol. 49, No. 1, 2009 24. 25. 26. 27. 28. 29. 30. 31. 32. 33. 34. 35. 36. 37. 38. 39. 40. 41. 42. 43. and patients with nonalcoholic fatty liver disease in Taiwan. World J Gastroenterol 2005;11:3905-3908. Chitturi S, Weltman M, Farrell GC, McDonald D, Kench J, Liddle C, et al. HFE mutations, hepatic iron, and fibrosis: ethnic-specific association of NASH with C282Y but not with fibrotic severity. HEPATOLOGY 2002; 36:142-149. Angulo P, Keach JC, Batts KP, Lindor KD. Independent predictors of liver fibrosis in patients with nonalcoholic steatohepatitis. HEPATOLOGY 1999;30:1356-1362. Bonora E, Targher G, Alberiche M, Bonadonna RC, Saggiani F, Zenere MB, et al. Homeostasis model assessment closely mirrors the glucose clamp technique in the assessment of insulin sensitivity: studies in subjects with various degrees of glucose tolerance and insulin sensitivity. Diabetes Care 2000;23:57-63. Muniyappa R, Lee S, Chen H, Quon MJ. Current approaches for assessing insulin sensitivity and resistance in vivo: advantages, limitations, and appropriate usage. Am J Physiol Endocrinol Metab 2008;294:E15–26. Stern SE, Williams K, Ferrannini E, DeFronzo RA, Bogardus C, Stern MP. Identification of individuals with insulin resistance using routine clinical measurements. Diabetes 2005;54:333-339. Yoneda M, Mawatari H, Fujita K, Iida H, Yonemitsu K, Kato S, et al. High-sensitivity C-reactive protein is an independent clinical feature of nonalcoholic steatohepatitis (NASH) and also of the severity of fibrosis in NASH. J Gastroenterol 2007;42:573-582. Targher G, Bertolini L, Rodella S, Lippi G, Franchini M, Zoppini G, et al. NASH predicts plasma inflammatory biomarkers independently of visceral fat in men. Obesity (Silver Spring) 2008;16:1394-1399. Singh H, Pollock R, Uhanova J, Kryger M, Hawkins K, Minuk GY. Symptoms of obstructive sleep apnea in patients with nonalcoholic fatty liver disease. Dig Dis Sci 2005;50:2338-2343. Kallwitz ER, Herdegen J, Madura J, Jakate S, Cotler SJ. Liver enzymes and histology in obese patients with obstructive sleep apnea. J Clin Gastroenterol 2007;41:918-921. Adams LA, Feldstein A, Lindor KD, Angulo P. Nonalcoholic fatty liver disease among patients with hypothalamic and pituitary dysfunction. HEPATOLOGY 2004;39:909-914. Liangpunsakul S, Chalasani N. Is hypothyroidism a risk factor for nonalcoholic steatohepatitis? J Clin Gastroenterol 2003;37:340-343. Cerda C, Perez-Ayuso RM, Riquelme A, Soza A, Villaseca P, Sir-Petermann T, et al. Nonalcoholic fatty liver disease in women with polycystic ovary syndrome. J Hepatol 2007;47:412-417. Setji TL, Holland ND, Sanders LL, Pereira KC, Diehl AM, Brown AJ. Nonalcoholic steatohepatitis and nonalcoholic Fatty liver disease in young women with polycystic ovary syndrome. J Clin Endocrinol Metab 2006;91:1741-1747. Gambarin-Gelwan M, Kinkhabwala SV, Schiano TD, Bodian C, Yeh HC, Futterweit W. Prevalence of nonalcoholic fatty liver disease in women with polycystic ovary syndrome. Clin Gastroenterol Hepatol 2007;5:496-501. Newton JL, Jones DE, Henderson E, Kane L, Wilton K, Burt AD, et al. Fatigue in non-alcoholic fatty liver disease (NAFLD) is significant and associates with inactivity and excessive daytime sleepiness but not with liver disease severity or insulin resistance. Gut 2008;57:807-813. Kheirandish-Gozal L, Sans Capdevila O, Kheirandish E, Gozal D. Elevated serum aminotransferase levels in children at risk for obstructive sleep apnea. Chest 2008;133:92-99. Tanne F, Gagnadoux F, Chazouilleres O, Fleury B, Wendum D, Lasnier E, et al. Chronic liver injury during obstructive sleep apnea. HEPATOLOGY 2005;41:1290-1296. Brunt EM. Pathology of nonalcoholic steatohepatitis. Hepatol Res 2005; 33:68-71. de Alwis NM, Day CP. Non-alcoholic fatty liver disease: the mist gradually clears. J Hepatol 2008;48 Suppl 1:S104-S112. Rafiq N, Bai C, Fand Y, Srishord MK, McCullough AJ, Younossi ZM. Over twenty five years of follow up for a Non-Alcoholic Fatty Liver Disease Cohort. Gastroenterology 2008;134:A754. VUPPALANCHI AND CHALASANI 315 44. Ratziu V, Charlotte F, Heurtier A, Gombert S, Giral P, Bruckert E, et al. Sampling variability of liver biopsy in nonalcoholic fatty liver disease. Gastroenterology 2005;128:1898-906. 45. Merriman RB, Ferrell LD, Patti MG, Weston SR, Pabst MS, Aouizerat BE, et al. Correlation of paired liver biopsies in morbidly obese patients with suspected nonalcoholic fatty liver disease. HEPATOLOGY 2006;44: 874-880. 46. Vuppalanchi R, Unalp A, Natta MV, Cummings O, Wass J, Tonascia J, et al. Increased diagnostic yield from liver biopsy in suspected NAFLD using multiple cores and mutiple readings. Gastroenterology 2007;132:A809. 47. Kleiner DE, Brunt EM, Van Natta M, Behling C, Contos MJ, Cummings OW, et al. Design and validation of a histological scoring system for nonalcoholic fatty liver disease. HEPATOLOGY 2005;41:1313-1321. 48. Alazmi WM, Regev A, Molina EG, Schiff ER. Predictors of cirrhosis in Hispanic patients with nonalcoholic steatohepatitis. Dig Dis Sci 2006; 51:1725-1729. 49. Kang H, Greenson JK, Omo JT, Chao C, Peterman D, Anderson L, et al. Metabolic syndrome is associated with greater histologic severity, higher carbohydrate, and lower fat diet in patients with NAFLD. Am J Gastroenterol 2006;101:2247-2253. 50. Wilson S, Chalasani N. Noninvasive markers of advanced histology in nonalcoholic fatty liver disease: are we there yet? Gastroenterology 2007; 133:1377-1378; discussion 1378-1379. 51. Wieckowska A, Zein NN, Yerian LM, Lopez AR, McCullough AJ, Feldstein AE. In vivo assessment of liver cell apoptosis as a novel biomarker of disease severity in nonalcoholic fatty liver disease. HEPATOLOGY 2006;44:27-33. 52. Leers MP, Kolgen W, Bjorklund V, Bergman T, Tribbick G, Persson B, et al. Immunocytochemical detection and mapping of a cytokeratin 18 neo-epitope exposed during early apoptosis. J Pathol 1999;187:567-572. 53. Yilmaz Y, Dolar E, Ulukaya E, Akgoz S, Keskin M, Kiyici M, et al. Soluble forms of extracellular cytokeratin 18 may differentiate simple steatosis from nonalcoholic steatohepatitis. World J Gastroenterol 2007;13:837-844. 54. Feldstein AE, Canbay A, Angulo P, Taniai M, Burgart LJ, Lindor KD, et al. Hepatocyte apoptosis and fas expression are prominent features of human nonalcoholic steatohepatitis. Gastroenterology 2003;125:437-443. 55. Charlton M, Angulo P, Chalasani N, Merriman R, Viker K, Charatcharoenwitthaya P, et al. Low circulating levels of dehydroepiandrosterone in histologically advanced nonalcoholic fatty liver disease. HEPATOLOGY 2008;47:484-492. 56. Angulo P, Hui JM, Marchesini G, Bugianesi E, George J, Farrell GC, et al. The NAFLD fibrosis score: a noninvasive system that identifies liver fibrosis in patients with NAFLD. HEPATOLOGY 2007;45:846-854. 57. Guha IN, Parkes J, Roderick P, Chattopadhyay D, Cross R, Harris S, et al. Noninvasive markers of fibrosis in nonalcoholic fatty liver disease: Validating the European Liver Fibrosis Panel and exploring simple markers. HEPATOLOGY 2008;47:455-460. 58. Harrison SM, Oliver D, Arnold HLM, Gogia SM, Neuschwander-Tetri BAM. Development and validation of a simple NAFLD clinical scoring system for identifying patients without advanced disease. Gut 2008;57: 1441-1447. 59. Rosenberg WM, Voelker M, Thiel R, Becka M, Burt A, Schuppan D, et al. Serum markers detect the presence of liver fibrosis: a cohort study. Gastroenterology 2004;127:1704-1713. 60. Ratziu V, Massard J, Charlotte F, Messous D, Imbert-Bismut F, Bonyhay L, et al. Diagnostic value of biochemical markers (FibroTest-FibroSURE) for the prediction of liver fibrosis in patients with non-alcoholic fatty liver disease. BMC Gastroenterol 2006;6:6. 61. Yoneda M, Mawatari H, Fujita K, Endo H, Iida H, Nozaki Y, et al. Noninvasive assessment of liver fibrosis by measurement of stiffness in patients with nonalcoholic fatty liver disease (NAFLD). Dig Liver Dis 2008;40:371-378. 62. Wong GL, Wong VW, Choi PC, Chan AW, Chum RH, Chan HK, et al. Assessment of fibrosis by transient elastography compared with liver biopsy and morphometry in chronic liver diseases. Clin Gastroenterol Hepatol 2008;6:1027-1035. 316 VUPPALANCHI AND CHALASANI 63. Baranova A, Younossi ZM. The future is around the corner: noninvasive diagnosis of progressive nonalcoholic steatohepatitis. HEPATOLOGY 2008; 47:373-375. 64. Krasnoff JB, Painter PL, Wallace JP, Bass NM, Merriman RB. Healthrelated fitness and physical activity in patients with nonalcoholic fatty liver disease. HEPATOLOGY 2008;47:1158-1166. 65. Bellentani S, Dalle Grave R, Suppini A, Marchesini G. Behavior therapy for nonalcoholic fatty liver disease: the need for a multidisciplinary approach. HEPATOLOGY 2008;47:746-754. 66. Huang MA, Greenson JK, Chao C, Anderson L, Peterman D, Jacobson J, et al. One-year intense nutritional counseling results in histological improvement in patients with non-alcoholic steatohepatitis: a pilot study. Am J Gastroenterol 2005;100:1072-1081. 67. Zivkovic AM, German JB, Sanyal AJ. Comparative review of diets for the metabolic syndrome: implications for nonalcoholic fatty liver disease. Am J Clin Nutr 2007;86:285-300. 68. Ouyang X, Cirillo P, Sautin Y, McCall S, Bruchette JL, Diehl AM, et al. Fructose consumption as a risk factor for non-alcoholic fatty liver disease. J Hepatol 2008;48:993-999. 69. Harrison SA, Brunt E, Fecht WJ, Neuschwander-Tetri BA. Orlistat (Xenical) in the treatment of overweight patients with nonalcoholic steatohepatitis (NASH): a multi-centered, randomized, prospective trial [Abstract]. Gastroenterology 2007;132:A809. 70. Vemuri VK, Janero DR, Makriyannis A. Pharmacotherapeutic targeting of the endocannabinoid signaling system: drugs for obesity and the metabolic syndrome. Physiol Behav 2008;93:671-686. 71. Van Gaal LF, Rissanen AM, Scheen AJ, Ziegler O, Rossner S. Effects of the cannabinoid-1 receptor blocker rimonabant on weight reduction and cardiovascular risk factors in overweight patients: 1-year experience from the RIO-Europe study. Lancet 2005;365:1389-1397. 72. Pi-Sunyer FX, Aronne LJ, Heshmati HM, Devin J, Rosenstock J. Effect of rimonabant, a cannabinoid-1 receptor blocker, on weight and cardiometabolic risk factors in overweight or obese patients: RIO-North America: a randomized controlled trial. JAMA 2006;295:761-775. 73. Teixeira-Clerc F, Julien B, Grenard P, Tran Van Nhieu J, Deveaux V, Li L, et al. CB1 cannabinoid receptor antagonism: a new strategy for the treatment of liver fibrosis. Nat Med 2006;12:671-676. 74. Gary-Bobo M, Elachouri G, Gallas JF, Janiak P, Marini P, RavinetTrillou C, et al. Rimonabant reduces obesity-associated hepatic steatosis and features of metabolic syndrome in obese Zucker fa/fa rats. HEPATOLOGY 2007;46:122-129. 75. Lowell JA, Shenoy S, Ghalib R, Caldwell C, White FV, Peters M, et al. Liver transplantation after jejunoileal bypass for morbid obesity. J Am Coll Surg 1997;185:123-127. 76. Wills CE. Long-term follow-up of jejunoileal bypass patients with preoperative cirrhosis of the liver. Obes Surg 1994;4:37-39. 77. Hocking MP, Davis GL, Franzini DA, Woodward ER. Long-term consequences after jejunoileal bypass for morbid obesity. Dig Dis Sci 1998; 43:2493-2499. 78. Levy P, Fried M, Santini F, Finer N. The comparative effects of bariatric surgery on weight and type 2 diabetes. Obes Surg 2007;17:1248-1256. 79. Sears D, Fillmore G, Bui M, Rodriguez J. Evaluation of gastric bypass patients 1 year after surgery: changes in quality of life and obesity-related conditions. Obes Surg 2008; doi: 10.1007/s11695-008-9604-x. 80. Sjostrom L, Lindroos AK, Peltonen M, Torgerson J, Bouchard C, Carlsson B, et al. Lifestyle, diabetes, and cardiovascular risk factors 10 years after bariatric surgery. N Engl J Med 2004;351:2683-2693. 81. Tessier DJ, Eagon JC. Surgical management of morbid obesity. Curr Probl Surg 2008;45:68-137. 82. Braghetto I, Korn O, Valladares H, Gutierrez L, Csendes A, Debandi A, et al. Laparoscopic sleeve gastrectomy: surgical technique, indications and clinical results. Obes Surg 2007;17:1442-1450. 83. Furuya CK, Jr., de Oliveira CP, de Mello ES, Faintuch J, Raskovski A, Matsuda M, et al. Effects of bariatric surgery on nonalcoholic fatty liver disease: preliminary findings after 2 years. J Gastroenterol Hepatol 2007; 22:510-514. HEPATOLOGY, January 2009 84. Mummadi R, Kasturi KS, Chennareddygari S, Sood G. Effect of bariatric surgery on nonalcoholic fatty liver disease (NAFLD): systemic review and meta-analysis. Clin Gastroenterol Hepatol 2008; doi: 10.1016/ j.cgh.2008.08.012. 85. Dallal RM, Mattar SG, Lord JL, Watson AR, Cottam DR, Eid GM, et al. Results of laparoscopic gastric bypass in patients with cirrhosis. Obes Surg 2004;14:47-53. 86. Kral JG, Thung SN, Biron S, Hould FS, Lebel S, Marceau S, et al. Effects of surgical treatment of the metabolic syndrome on liver fibrosis and cirrhosis. Surgery 2004;135:48-58. 87. Kirpichnikov D, McFarlane SI, Sowers JR. Metformin: an update. Ann Intern Med 2002;137:25-33. 88. Hattori Y, Suzuki K, Hattori S, Kasai K. Metformin inhibits cytokine-induced nuclear factor kappaB activation via AMP-activated protein kinase activation in vascular endothelial cells. Hypertension 2006;47:1183-1188. 89. Lin HZ, Yang SQ, Chuckaree C, Kuhajda F, Ronnet G, Diehl AM. Metformin reverses fatty liver disease in obese, leptin-deficient mice. Nat Med 2000;6:998-1003. 90. Angelico F, Burattin M, Alessandri C, Del Ben M, Lirussi F. Drugs improving insulin resistance for non-alcoholic fatty liver disease and/or non-alcoholic steatohepatitis. Cochrane Database Syst Rev 2007: CD005166. 91. Knowler WC, Barrett-Connor E, Fowler SE, Hamman RF, Lachin JM, Walker EA, et al. Reduction in the incidence of type 2 diabetes with lifestyle intervention or metformin. N Engl J Med 2002;346:393-403. 92. Girard J. [Mechanisms of action of thiazolidinediones]. Diabetes Metab 2001;27:271-278. 93. Yki-Jarvinen H. Thiazolidinediones. N Engl J Med 2004;351:1106-1118. 94. Watkins PB. Insight into hepatotoxicity: the troglitazone experience. HEPATOLOGY 2005;41:229-230. 95. Scheen AJ. Hepatotoxicity with thiazolidinediones: is it a class effect? Drug Saf 2001;24:873-888. 96. Riera-Guardia N, Rothenbacher D. The effect of thiazolidinediones on adiponectin serum level: a meta-analysis. Diabetes Obes Metab 2008;10: 367-375. 97. Caldwell SH, Hespenheide EE, Redick JA, Iezzoni JC, Battle EH, Sheppard BL. A pilot study of a thiazolidinedione, troglitazone, in nonalcoholic steatohepatitis. Am J Gastroenterol 2001;96:519-525. 98. Neuschwander-Tetri BA, Brunt EM, Wehmeier KR, Oliver D, Bacon BR. Improved nonalcoholic steatohepatitis after 48 weeks of treatment with the PPAR-gamma ligand rosiglitazone. HEPATOLOGY 2003;38: 1008-1017. 99. Promrat K, Lutchman G, Uwaifo GI, Freedman RJ, Soza A, Heller T, et al. A pilot study of pioglitazone treatment for nonalcoholic steatohepatitis. HEPATOLOGY 2004;39:188-196. 100. Shadid S, Jensen MD. Effect of pioglitazone on biochemical indices of non-alcoholic fatty liver disease in upper body obesity. Clin Gastroenterol Hepatol 2003;1:384-387. 101. Sanyal AJ, Mofrad PS, Contos MJ, Sargeant C, Luketic VA, Sterling RK, et al. A pilot study of vitamin E versus vitamin E and pioglitazone for the treatment of nonalcoholic steatohepatitis. Clin Gastroenterol Hepatol 2004;2:1107-1115. 102. Belfort R, Harrison SA, Brown K, Darland C, Finch J, Hardies J, et al. A placebo-controlled trial of pioglitazone in subjects with nonalcoholic steatohepatitis. N Engl J Med 2006;355:2297-307. 103. Ratziu V, Giral P, Jacqueminet S, Charlotte F, Hartemann-Heurtier A, Serfaty L, et al. Rosiglitazone for nonalcoholic steatohepatitis: one-year results of the randomized placebo-controlled Fatty Liver Improvement with Rosiglitazone Therapy (FLIRT) Trial. Gastroenterology 2008;135: 100-110. 104. Aithal GP, Thomas JA, Kaye PV, Lawson A, Ryder SD, Spendlove I, et al. Randomized, placebo controlled trial of pioglitazone in non-diabetic subjects with nonalcoholic steatohepatitis (NASH). Gastroenterology 2008; doi:10.1053/j.gastro.2008.06.047. 105. Lutchman G, Modi A, Kleiner DE, Promrat K, Heller T, Ghany M, et al. The effects of discontinuing pioglitazone in patients with nonalcoholic steatohepatitis. HEPATOLOGY 2007;46:424-429. HEPATOLOGY, Vol. 49, No. 1, 2009 106. Nissen SE, Wolski K. Effect of rosiglitazone on the risk of myocardial infarction and death from cardiovascular causes. N Engl J Med 2007;356: 2457-2471. 107. Ding X, Saxena NK, Lin S, Gupta NA, Anania FA. Exendin-4, a glucagon-like protein-1 (GLP-1) receptor agonist, reverses hepatic steatosis in ob/ob mice. HEPATOLOGY 2006;43:173-181. 108. Poon T, Nelson P, Shen L, Mihm M, Taylor K, Fineman M, et al. Exenatide improves glycemic control and reduces body weight in subjects with type 2 diabetes: a dose-ranging study. Diabetes Technol Ther 2005;7:467-477. 109. Bugianesi E. Nonalcoholic fatty liver disease (NAFLD) and cardiac lipotoxicity: another piece of the puzzle. HEPATOLOGY 2008;47:2-4. 110. Ekstedt M, Franzen LE, Mathiesen UL, Thorelius L, Holmqvist M, Bodemar G, et al. Long-term follow-up of patients with NAFLD and elevated liver enzymes. HEPATOLOGY 2006;44:865-873. 111. Loria P, Lonardo A, Bellentani S, Day CP, Marchesini G, Carulli N. Non-alcoholic fatty liver disease (NAFLD) and cardiovascular disease: an open question. Nutr Metab Cardiovasc Dis 2007;17:684-698. 112. Targher G. Non-alcoholic fatty liver disease, the metabolic syndrome and the risk of cardiovascular disease: the plot thickens. Diabet Med 2007;24:1-6. 113. Adams LA, Lymp JF, St Sauver J, Sanderson SO, Lindor KD, Feldstein A, et al. The natural history of nonalcoholic fatty liver disease: a populationbased cohort study. Gastroenterology 2005;129:113-121. 114. Ong JP, Pitts A, Younossi ZM. Non-alcoholic fatty liver disease (NAFLD) is associated with higher overall mortality and liver related mortality. J Hepatol 2008;48:S5-S5. 115. Marchesini G, Bugianesi E, Forlani G, Cerrelli F, Lenzi M, Manini R, et al. Nonalcoholic fatty liver, steatohepatitis, and the metabolic syndrome. HEPATOLOGY 2003;37:917-923. 116. McCullough AJ. The clinical features, diagnosis and natural history of nonalcoholic fatty liver disease. Clin Liver Dis 2004;8:521-533, viii. 117. Mofrad P, Contos MJ, Haque M, Sargeant C, Fisher RA, Luketic VA, et al. Clinical and histologic spectrum of nonalcoholic fatty liver disease associated with normal ALT values. HEPATOLOGY 2003;37:1286-1292. 118. Villanova N, Moscatiello S, Ramilli S, Bugianesi E, Magalotti D, Vanni E, et al. Endothelial dysfunction and cardiovascular risk profile in nonalcoholic fatty liver disease. HEPATOLOGY 2005;42:473-480. 119. Chalasani N, Deeg MA, Crabb DW. Systemic levels of lipid peroxidation and its metabolic and dietary correlates in patients with nonalcoholic steatohepatitis. Am J Gastroenterol 2004;99:1497-1502. 120. Pacifico L, Cantisani V, Ricci P, Osborn JF, Schiavo E, Anania C, et al. Nonalcoholic fatty liver disease and carotid atherosclerosis in children. Pediatr Res 2008;63:423-427. 121. Fracanzani AL, Burdick L, Raselli S, Pedotti P, Grigore L, Santorelli G, et al. Carotid artery intima-media thickness in nonalcoholic fatty liver disease. Am J Med 2008;121:72-78. 122. Targher G, Bertolini L, Padovani R, Rodella S, Zoppini G, Zenari L, et al. Relations between carotid artery wall thickness and liver histology in subjects with nonalcoholic fatty liver disease. Diabetes Care 2006;29: 1325-1330. 123. Akabame S, Hamaguchi M, Tomiyasu K, Tanaka M, Kobayashi-Takenaka Y, Nakano K, et al. Evaluation of vulnerable coronary plaques and non-alcoholic fatty liver disease (NAFLD) by 64-detector multislice computed tomography (MSCT). Circ J 2008;72:618-625. 124. Perseghin G, Lattuada G, De Cobelli F, Esposito A, Belloni E, Ntali G, et al. Increased mediastinal fat and impaired left ventricular energy metabolism in young men with newly found fatty liver. HEPATOLOGY 2008;47:51-58. 125. Targher G, Bertolini L, Padovani R, Poli F, Scala L, Tessari R, et al. Increased prevalence of cardiovascular disease in type 2 diabetic patients with non-alcoholic fatty liver disease. Diabet Med 2006;23:403-409. 126. Chalasani N, Aljadhey H, Kesterson J, Murray MD, Hall SD. Patients with elevated liver enzymes are not at higher risk for statin hepatotoxicity. Gastroenterology 2004;126:1287-1292. 127. Chalasani N. Statins and hepatotoxicity: focus on patients with fatty liver. HEPATOLOGY 2005;41:690-695. 128. Vuppalanchi R, Teal E, Chalasani N. Patients with elevated baseline liver enzymes do not have higher frequency of hepatotoxicity from lovastatin than those with normal baseline liver enzymes. Am J Med Sci 2005;329:62-65. VUPPALANCHI AND CHALASANI 317 129. Lewis JH, Mortensen ME, Zweig S, Fusco MJ, Medoff JR, Belder R. Efficacy and safety of high-dose pravastatin in hypercholesterolemic patients with well-compensated chronic liver disease: results of a prospective, randomized, double-blind, placebo-controlled, multicenter trial. HEPATOLOGY 2007;46:1453-1463. 130. Kaneda H, Hashimoto E, Yatsuji S, Tokushige K, Shiratori K. Hyaluronic acid levels can predict severe fibrosis and platelet counts can predict cirrhosis in patients with nonalcoholic fatty liver disease. J Gastroenterol Hepatol 2006;21:1459-1465. 131. Yoneda M, Fujita K, Inamori M, Tamano M, Hiriishi H, Nakajima A. Transient elastography in patients with non-alcoholic fatty liver disease (NAFLD). Gut 2007;56:1330-1331. 132. Talwalkar JA, Yin M, Fidler JL, Sanderson SO, Kamath PS, Ehman RL. Magnetic resonance imaging of hepatic fibrosis: emerging clinical applications. HEPATOLOGY 2008;47:332-342. 133. Szczepaniak LS, Nurenberg P, Leonard D, Browning JD, Reingold JS, Grundy S, et al. Magnetic resonance spectroscopy to measure hepatic triglyceride content: prevalence of hepatic steatosis in the general population. Am J Physiol Endocrinol Metab 2005;288:E462-E468. 134. Tsuda N, Okada M, Murakami T. Potential of gadolinium-ethoxybenzyl-diethylenetriamine pentaacetic acid (Gd-EOB-DTPA) for differential diagnosis of nonalcoholic steatohepatitis and fatty liver in rats using magnetic resonance imaging. Invest Radiol 2007;42:242-247. 135. Portincasa P, Grattagliano I, Lauterburg BH, Palmieri VO, Palasciano G, Stellaard F. Liver breath tests non-invasively predict higher stages of nonalcoholic steatohepatitis. Clin Sci (Lond) 2006;111:135-143. 136. Solga SF, Alkhuraishe A, Cope K, Tabesh A, Clark JM, Torbenson M, et al. Breath biomarkers and non-alcoholic fatty liver disease: preliminary observations. Biomarkers 2006;11:174-183. 137. Horoz M, Bolukbas C, Bolukbas FF, Sabuncu T, Aslan M, Sarifakiogullari S, et al. Measurement of the total antioxidant response using a novel automated method in subjects with nonalcoholic steatohepatitis. BMC Gastroenterol 2005;5:35. 138. Hui JM, Hodge A, Farrell GC, Kench JG, Kriketos A, George J. Beyond insulin resistance in NASH: TNF-alpha or adiponectin? HEPATOLOGY 2004;40:46-54. 139. Musso G, Gambino R, Biroli G, Carello M, Faga E, Pacini G, et al. Hypoadiponectinemia predicts the severity of hepatic fibrosis and pancreatic Beta-cell dysfunction in nondiabetic nonobese patients with nonalcoholic steatohepatitis. Am J Gastroenterol 2005;100:2438-2446. 140. Wieckowska A, Papouchado BG, Li Z, Lopez R, Zein NN, Feldstein AE. Increased hepatic and circulating interleukin-6 levels in human nonalcoholic steatohepatitis. Am J Gastroenterol 2008;103:1372-1379. 141. Haukeland JW, Damas JK, Konopski Z, Loberg EM, Haaland T, Goverud I, et al. Systemic inflammation in nonalcoholic fatty liver disease is characterized by elevated levels of CCL2. J Hepatol 2006;44:1167-1174. 142. Marchesini G, Brizi M, Bianchi G, Tomassetti S, Zoli M, Melchionda N. Metformin in non-alcoholic steatohepatitis. Lancet 2001;358:893-894. 143. Nair S, Diehl AM, Wiseman M, Farr GH, Jr., Perrillo RP. Metformin in the treatment of non-alcoholic steatohepatitis: a pilot open label trial. Aliment Pharmacol Ther 2004;20:23-28. 144. Uygun A, Kadayifci A, Isik AT, Ozgurtas T, Deveci S, Tuzun A, et al. Metformin in the treatment of patients with non-alcoholic steatohepatitis. Aliment Pharmacol Ther 2004;19:537-544. 145. Bugianesi E, Gentilcore E, Manini R, Natale S, Vanni E, Villanova N, et al. A randomized controlled trial of metformin versus vitamin E or prescriptive diet in nonalcoholic fatty liver disease. Am J Gastroenterol 2005; 100:1082-1090. 146. Schwimmer JB, Middleton MS, Deutsch R, Lavine JE. A phase 2 clinical trial of metformin as a treatment for non-diabetic paediatric non-alcoholic steatohepatitis. Aliment Pharmacol Ther 2005;21:871-879. 147. Loomba R, Lutchman G, Kleiner D, Borg B, Feld J, Modi A, et al. Pilot study of metformin in patients with nonalcoholic steatohepatitis [Abstract]. HEPATOLOGY 2006;44:260A. 148. Nobili V, Manco M, Ciampalini P, Alisi A, Devito R, Bugianesi E, et al. Metformin use in children with nonalcoholic fatty liver disease: an openlabel, 24-month, observational pilot study. Clin Ther 2008;30:1168-1176.