Hepatol Int DOI 10.1007/s12072-013-9482-8

SUPPLEMENT ISSUE: NAFLD

Pharmacological agents for nonalcoholic steatohepatitis Maeva Guillaume • Vlad Ratziu

Received: 8 June 2013 / Accepted: 9 October 2013 Ó Asian Pacific Association for the Study of the Liver 2013

Abstract The rationale for specific pharmacologic therapy in nonalcoholic steatohepatitis (NASH) is determined by the potential for disease progression and the difficulties, in many patients, to successfully implement diet and lifestyle changes in the long term. Because they correct insulin resistance, insulin-sensitizing agents are attractive candidates for the treatment of NASH. However, two randomized studies have shown that vitamin E, despite having no effect on insulin sensitivity, achieves interesting histological and biochemical efficacy. This review provides an insight into the therapeutic efficacy and safety issues of different pharmacological agents tested in human NASH. Keywords Steatohepatitis  Fibrosis  Steatosis  Glitazones  Vitamin E  Ursodesoxycholic acid  Insulin resistance  Oxidative stress  Insulin sensitizers

M. Guillaume Service d’He´patologie et Gastro-ente´rologie, INSERM 1048 and Universite´ Paul Sabatier, Hoˆpital Purpan, Centre Hospitalier Universitaire de Toulouse, Toulouse, France e-mail: [email protected] M. Guillaume Service d’He´patologie et Gastro-ente´rologie, Hoˆpital Purpan, Pavillon Dieulafoy, 4e`me e´tage, Place du Docteur Baylac, TSA 40031, 31059 Toulouse Cedex 9, France V. Ratziu (&) INSERM U938, Hospital Pitie´ Salpeˆtrie`re, CdR Saint-Antoine and Universite´ Pierre et Marie Curie, Paris, France e-mail: [email protected] V. Ratziu Service d’He´patologie et Gastro-ente´rologie, Hoˆpital Pitie´Salpeˆtrie`re, 47-83, Boulevard de l’hoˆpital, 75651 Paris Cedex 13, France

Introduction Nonalcoholic fatty liver disease (NAFLD) encompasses the entire spectrum of fatty liver disease in individuals without significant alcohol consumption, ranging from nonalcoholic fatty liver (NAFL) to nonalcoholic steatohepatitis (NASH) and cirrhosis. NAFL is defined as the presence of hepatic steatosis with minimal, non-specific inflammation that does not qualify for steatohepatitis; NASH is defined as the presence of lobular inflammation with hepatocyte injury (ballooning) [1]. The risk of progression to cirrhosis, liver failure and liver cancer is minimal for NAFL and real for NASH [2]. In addition, there is growing evidence to support an association between NAFLD and an increased risk of HCC [3]. Patients with NASH have increased liverrelated mortality as well as cardiovascular mortality [4]. The recommended first-line therapy for all NAFLD patients is non-pharmacological: weight reduction through diet, changes in lifestyle, physical exercise and proscribing sedentarity [5, 6]. Additionally, concurrent metabolic disorders such as type 2 diabetes (T2DM), hyperlipidemia or arterial hypertension, when present, should be well controlled, although their specific impact on improving the hepatic disease has not yet been determined. Occasionally, these measures result in a 7–10 % weight loss, a threshold that is possibly associated with histological improvement [5, 6]. Moreover, as insulin resistance, inflammation, an unbalanced adipocytokine ratio and angiogenic factors associated with NAFLD could favor the development of HCC [7], these traditional therapeutic approaches in NAFLD may theoretically reduce the risk of HCC by acting on common pathophysiological pathways shared by NAFLD and HCC. In patients who failed these measures or in those with advanced disease (NASH with significant fibrosis), pharmacological treatments specifically directed at

123

Hepatol Int

improving hepatic inflammation, fibrosis and/or clearing steatohepatitis might be necessary [8]. In some carefully selected patients with morbid obesity and without cirrhosis, bariatric surgery may be proposed [9]. Here we will provide an overview of data obtained so far with pharmacological agents in NASH mostly focusing on controlled studies with histological documentation of NASH. The therapeutic area in NASH is still in its infancy; therefore, there is an urgent need for well-conducted randomized controlled trials with relevant therapeutic endpoints [10].

Insulin sensitizers Insulin resistance is believed to be the main factor underlying liver fat buildup and possibly, through lipotoxic pathways, hepatic inflammation and fibrogenesis; improvement of insulin sensitivity is therefore a legitimate therapeutic target. Conversely, the presence of NAFLD in people with T2DM often makes it difficult to control diabetes: indeed, the intrahepatic triglyceride content is, in some studies, the major determinant of the daily requirements of insulin [11]. Moreover, NAFLD is independently associated with a twofold increased risk of T2DM. Consequently, treatment strategies decreasing intrahepatic triglyceride content and improving insulin sensitivity might contribute to better glycemic control [2]. In addition, as insulin resistance could favor the development of HCC, recent studies suggest that at least some antidiabetic drugs may reduce the risk of developing HCC [12]. This claim was made for metformin, although the data available so far are largely inconclusive from a methodological standpoint [13, 14]. Thiazolidinediones Glitazones are peroxisome proliferator-activated receptor gamma (PPARc) agonists with an insulin-sensitizing activity, currently used for treating T2DM. It has been suggested [15] but not confirmed [16] that glitazones strongly improve adipose tissue insulin resistance, which correlates with the reduction in steatosis and necroinflammation [15]. In controlled clinical trials, pioglitazone improved aminotransferase levels and histological features of liver injury (including resolution of steatohepatitis), except for fibrosis, in NASH patients with insulin resistance or T2DM [17], but also in nondiabetic NASH patients [18, 19]. The largest multicenter, randomized, controlled trial (RCT) (PIVENS) of pioglitazone (at the lower dose of 30 mg/day) in nondiabetic patients with NASH, lasting 2 years, failed to achieve statistically significant improvement of a complex composite primary endpoint that included a 2-point reduction in the NAFLD

123

activity score (NAS)[19]. This could be explained by a higher level of statistical significance used in this three-arm trial or by methodological issues ultimately restricting the statistical power [20]. Despite a significant antisteatogenic effect and maintained reduction in alanine transaminase (ALT) levels, rosiglitazone, surprisingly, failed to improve steatohepatitis and its defining histological lesions [21, 22]. A recent meta-analysis of glitazones [23] concluded a histological benefit across the spectrum of histological lesions despite significant and non-reversible weight gain [24]. The use of both glitazones has been severely restricted by black-box warnings based on an increase in cardiovascular events [20], congestive heart failure [25], bone fractures in women [26] and risk of bladder cancer for pioglitazone [27] that justified its market withdrawal in France. While rosiglitazone has been largely withdrawn from the market, current guidelines recommend the use of pioglitazone for the treatment of steatohepatitis in patients with biopsy-proven NASH. However, the long-term safety and benefit is unknown, and its use requires caution in diabetic patients and in those with impaired cardiac function [6, 8]. Metformin Metformin is a safe antidiabetic drug that reduces hepatic glucose production and can induce weight loss. Earlier studies showed encouraging results on aminotransferases [28–30]. However, this drug has no clear benefit on liver histology [30–32], probably because of its limited antisteatogenic effect and its inability to restore adiponectin levels [33]. Possibly, weight loss on therapy might account for some of the discordant findings. Based on data from trials conducted so far, metformin is not recommended specifically for the treatment of NASH [6, 8]. Preclinical studies of glucagon-like peptide-1 agonists suggested these could be promising agents for the treatment of NAFLD [34–36].

Antioxidants and hepatoprotectants Improving insulin sensitivity may not be sufficient for improving liver injury [20]; therefore, specific hepatoprotective agents with antioxidant, antiinflammatory, antiapoptotic or other cytoprotective properties may be required either alone or in addition to insulin sensitizers. Vitamin E A few early studies have suggested that vitamin E (atocopherol), a natural antioxidant, improved liver enzymes, steatosis and histological features of NASH, except for

Hepatol Int

fibrosis, when combined with pioglitazone [37] or ursodeoxycholic acid [38]. In the largest RCT to date, the PIVENS trial [19], vitamin E at 800 IU/day improved hepatocyte ballooning and inflammation and cleared steatohepatitis at a higher rate than placebo. These results were partly confirmed in a pediatric trial where vitamin E only improved hepatocyte ballooning and increased clearance of NASH but did not improve inflammation or ALT levels [39]. Interestingly, a 2-year treatment with vitamin E in combination with vitamin C did not result in histological improvement in another pediatric trial [40], thus dampening the enthusiasm for a universal benefit of this molecule in NASH. Another reason for concern is persisting doubt about the long-term safety of vitamin E supplements. Some studies suggest vitamin E might increase overall mortality [41] and the risks of hemorrhagic stroke [42] and prostate cancer [43]. While recommended as a first-line pharmacotherapy in non-diabetic adults with biopsy-proven NASH without cirrhosis [6], clearly more data on immediate efficacy and long-term safety are needed for more confident recommendations. Ursodeoxycholic acid (UDCA) A natural bile acid with hepatoprotective properties induced biochemical and histological improvement in two small RCTs at the dose of 13–15 mg/kg combined with vitamin E [38] or alone [44]. Unfortunately, UDCA alone failed to improve liver enzymes or hepatic histology in three other studies at low (13–15 mg/kg) [38, 45] or higher doses (23–28 mg/kg) [46]. These studies had, however, significant methodological shortcomings [47]. A French multicentric RCT has recently tested the effects of higher doses (28–35 mg/kg) of UDCA for 1 year in 126 NASH patients versus placebo [48]. There was an early, sustained and significant improvement of ALT levels and some suggestion of improvement of insulin resistance surrogate markers and fibrosis scores [48]. In the absence of positive studies with histological outcomes, UDCA is not universally recommended as a treatment for patients with NASH [6, 8]. Pentoxifylline It reduces TNF-a expression in animal models of NASH and has shown biochemical efficacy and histological improvement in a few uncontrolled trials of very small sample size [49]. A recent, 1-year, randomized controlled trial of 55 patients treated with 1,200 mg of pentoxifylline per day [50] has shown efficacy on liver enzymes and histological features of NASH, with even a marginal effect on fibrosis [50]. This beneficial effect could be correlated with decreased oxidized lipid products [51]. These

promising results from an early, small-size, monocentric RCT should be further validated.

Therapies with potential but yet unproven benefit in NASH Angiotensin II receptor antagonists (sartans) Experimental evidence suggests that losartan may have antifibrotic actions, possibly by inhibiting hepatic stellate cell activation [52]. In a rat model of NAFLD, losartan and telmisartan improved fatty liver [53]. While telmisartan, which also acts as a partial agonist of PPARc, improved insulin resistance, losartan could, in addition, reduce the expression of hepatic plasminogen activator inhibitor-1 (PAI-1) [53]. One pilot study in hypertensive patients with NASH has shown that 1 year of treatment with losartan reduced ALT levels, hepatic necroinflammation and fibrosis, but not steatosis [52]. The only randomized openlabel trial in NASH patients that tested 48 weeks of rosiglitazone combined with losartan, compared to rosiglitazone alone, showed that the combination therapy conferred no greater benefit with respect to histopathology [54]. Based on findings that a gene variant (A1166C) in the angiotensin II type 1 receptor predicts the therapeutic response to losartan [55], the possibility to predict response in NASH patient is appealing and should be further tested. Lipid-lowering drugs Statins There is no evidence for a beneficial effect of statins on liver histology in patients with NASH, except, possibly, for a marginal improvement in steatosis [56]. Hence, statins are not recommended for NASH treatment [6]. However, concurrent metabolic comorbidities clearly dictate the use of statins for cardiovascular endpoints in NAFLD patients. Altered liver function tests should not preclude the use of statins in these patients. Numerous studies have shown the safety of statins in regard to hepatotoxicity [57], as these drugs actually lower aminotransferases in patients with the metabolic syndrome [58]. In addition, experimental studies and preclinical observations suggest that statins might prevent portal hypertension and the development of HCC [59–62]. Indeed, by acting on both liver stem cells and endothelial cells, statins might specifically affect some of the main molecular pathways of HCC, such as inhibition of cell proliferation, induction of apoptosis and inhibition of angiogenesis [63]. One meta-analysis using a propensity score analysis has shown that statin use is associated with a

123

Hepatol Int

reduced risk of HCC, most strongly in Asian but also in Western populations [64].

at designing potent peripheral CB1R blockers, but early studies in humans are not yet available.

Fibrates

Uric acid-lowering agents

These conventional triglyceride-lowering agents had no effect on aminotransferases or liver histology in patients with NAFLD [44].

The serum uric acid level is strongly associated with individual components of the metabolic syndrome, although a causal link is controversial [71–73]. Several observational studies have demonstrated a relationship between hyperuricemia and NAFLD [74]. In a large population-based study of nondiabetic adults from the USA, elevated serum uric acid was independently associated with the presence of elevated serum liver enzymes, with ultrasound-diagnosed NAFLD [75] and with a higher risk of cirrhosis-related hospitalization or death [76, 77]. These findings warrant further studies on the role of uric acid in NAFLD, but at this point it is entirely conjectural whether uric acid-lowering drugs could carry therapeutic potential for NASH.

Probucol It is a lipid-lowering agent that also has antioxidative properties. Merat et al. [65] showed that probucol (500 mg/ day) could significantly improve liver enzymes and even hepatic histology, except for fibrosis [66], in NASH patients. Larger RCTs are now required. Omega-3 fatty acids Omega-3 fatty acids are essential polyunsaturated fatty acids (PUFA), unable to be synthesised de novo, that have antiinflammatory and antioxidant properties. Patients with NAFLD have a lower dietary intake of these omega-3 fatty acids than healthy controls, leading to an increased omega6/omega-3 ratio. Downstream products of omega-6 are proinflammatory, increase lipogenesis and hepatic uptake of circulating free fatty acids (FFAs), and reduce hepatocyte fatty acid oxidation and synthesis of fatty acid transport proteins (i.e., VLDL), thus leading to steatosis. Therefore, dietary supplementation of omega-3 PUFAs has been suggested as a treatment of NAFLD. Currently approved in the USA to treat hypertriglyceridemia, omega3 fatty acids improve aminotransferases and steatosis in patients with NAFLD [67]. Two recent meta-analyses found experimental evidence to support their use and require well-conducted RCTs to confirm these results [68, 69]. To date, it is premature to recommend their use for the specific treatment of NAFLD, but they can be considered as the first-line agents to treat hypertriglyceridemia in patients with NAFLD [6]. The results of a large multicentric US study of eicosapentaenoic acid are awaited. Weight-loss agents Orlistat, an enteric lipase inhibitor, did not enhance weight loss or improve liver enzymes and histology [70], and this drug is not recommended for the treatment of NASH [6]. Rimonabant, a non-selective cannabinoid type 1 receptor blocker, was tested in two large international phase 3 trials in NASH patients. Unfortunately, because of severe psychiatric side effects, the marketing authorization was withdrawn, and the studies were halted. Current work aims

123

Iron depletion While not a pharmacological agent in a strict sense, phlebotomy is a widely available and inexpensive therapy. Hyperferritinemia, which reflects hyperinsulinemia, is observed in about 20–30 % of patients with NAFLD [78] and is associated with the severity of liver damage [79]. Excessive body iron may play a causal role in insulin resistance through mechanisms that involve a reduced ability to oxidize carbohydrates and altered release of adipokines and beta-cell function [78]. Furthermore, it may facilitate the progression of cardiovascular disease by contributing to the recruitment and activation of macrophages within arterial lesions. Iron might also induce oxidative stress in hepatocytes, activation of HSCs and malignant transformation by promotion of cell growth and DNA damage [78]. In rats with fatty liver, iron depletion by deferoxamine upregulates glucose uptake, and increases insulin receptor activity and insulin signaling in hepatocytes [80, 81]. In humans, iron depletion by phlebotomy improved insulin resistance [81–85], liver iron content, NAS [82, 86, 87] and liver enzymes [83, 85]. It is unclear at this point whether and how iron depletion could be incorporated in the treatment strategies of patients with NASH [88]. Modulation of gut microbiota Gut microbiota is associated with obesity and the metabolic syndrome and could also play a role in the development of NASH, probably because of increased endotoxemia and activation of the Toll-like receptor-4 signaling cascade [89, 90]. Prebiotic modulation of gut microbiota in ob/ob mice

Hepatol Int

lowers intestinal permeability and increases the endogenous glucagon-like peptide 2 (GLP-2) [91]. Ampicillin and neomycin improved insulin resistance in animals [92–94]. Based on these data, treatments aiming at manipulating enteric flora by using intestinally focused antibiotics, probiotics or prebiotics are proposed. In healthy [95] and in overweight and obese adults [96], it improves glucose tolerance compared to placebo. This effect was associated with weight loss and with an increased plasma GLP-1 and peptide YY concentrations. [95, 96]. Probiotics, which exhibit immunoregulatory and antiinflammatory activity, showed beneficial effects on liver fibrosis [90] in animal models of NASH. It could also improve steatosis and insulin resistance [97, 98], and preliminary data from two pilot studies suggest that they may be well tolerated and may improve conventional liver function tests [99, 100]. Randomized controlled clinical trials are now awaited [89, 101–103].

model of NAFLD, FXR agonist decreased ALT levels and significantly reduced inflammatory cell infiltration and hepatic fibrosis without changes in steatosis [111]. A large multicentric RCT of obeticholic acid, an FXR agonist with insulin-sensitizing properties, has completed enrollment in the US. Final results are expected in 2014. Caspase inhibitors GS-9450 is a selective caspase inhibitor that improved liver injury in murine models of NASH in particular in the methionine–choline deficient diet. In human trials, it reduced aminotransferases in patients with biopsy-proven NASH in a short-term phase 2 RCT [112], but development is currently halted because of concerns about possible hepatotoxicity. GFT505

L-carnitine

This is a modulator of mitochondrial FFA transport and boxidation, with antioxidative and antiinflammatory properties [104]. In an RCT performed on 74 NASH patients, Lcarnitine (2 g/day) supplementation to the diet for 24 weeks improved ALT, triglycerides and the lipid profile, HOMA-IR, TNFa, steatosis and NAS, compared with the placebo plus diet group [105]. These results are interesting but have not been reproduced so far. Thyromimetics Hypothyroidism is associated with the risk of developing the metabolic syndrome, and high-though-normal TSH values are independently associated with NASH in euthyroid patients [106]. Thyroid hormones such as T3 might have a prosteatogenic effect by increasing plasma FFA flux from the periphery to the liver and increasing hepatic lipogenesis [107, 108]. However, a specific agonist of the b isoform of the thyroid hormone receptor induced hepatic beta-oxidation of fatty acids resulting in a reduction of triacyl and diacyl glycerol in rodent models of hepatic steatosis [109]. This reduction in hepatic fat, especially if accompanied by an improvement of insulin resistance, could prove a useful pharmacological option to test in humans [110].

It is a dual PPAR alpha-delta agonist that improves the lipid profile and glycemic control in patients with atherogenic dyslipidemia and pre-diabetes [113]. It also has potent antiinflammatory and antifibrotic activities in several models of chronic murine liver injury. Contrary to PPAR gamma agonists, which can induce weight gain and cardiovascular events, its safety profile seems unremarkable so far. Based on these preclinical results and the results of several phase 2a trials in humans, a large international study of GFT-505 has been initiated in patients with NASH. Pre-clinical studies Pre-clinical studies have tested other therapeutic agents in mouse models of NAFLD. Administration of retinoids could improve insulin sensitivity through upregulation of hepatic leptin receptor expression [114]. Pharmacological inhibition of adipocyte fatty acid binding protein, a key mediator of inflammatory response in Kupffer cells, ameliorated hepatic steatosis, macrophage infiltration and hepatocyte ballooning [115]. Carbamazepine or rapamycin, which both enhance macroautophagy, could improve steatosis and insulin sensitivity by increasing lipid degradation [116]. Conclusion and perspectives

Agents in development The farnesoid X receptor (FXR) This is a member of the nuclear hormone receptor superfamily, which plays an essential role in the regulation of enterohepatic circulation and lipid homeostasis. In a mouse

Therapeutic research and drug development in NASH has only started, but will experience considerable traction because of the clear unmet medical need. While diet and lifestyle changes should always be the first-line therapy, current practice guidelines recommend the use of hepatic pharmacological therapy in patients with NASH and

123

Hepatol Int

advanced fibrosis, without cirrhosis [6]. Despite limited results on efficacy and safety, vitamin E and pioglitazone are the recommended pharmacological agents, both of them being cost-effective in this indication [23]. Future therapeutic research in NASH should focus on exploring innovative molecules with better efficacy and a better safety profile. Because of the numerous comorbidities and heavy drug intake in patients with NASH, any new agent should not worsen, through off-target effects or drug-drug interactions, the concurrent diseases or therapeutic regimens. An individualized, dual approach of improving insulin resistance and conferring hepatoprotective, antiinflammatory and antifibrotic actions might be necessary for effective treatment of this complex condition. Compliance with Ethical Requirements This article is a review article and does not contain any studies with human or animal subjects. Conflict of interest V. R. served as a consultant for Axcan, Astellas, Enterome, GalMed, Genfit, Gilead and Roche. M. G. declared no conflict of interest.

References 1. Kleiner D, Brunt E. Nonalcoholic fatty liver disease: pathologic patterns and biopsy evaluation in clinical research. Semin Liver Dis 2012;32:3–13 2. Anstee QM, Targher G, Day CP. Progression of NAFLD to diabetes mellitus, cardiovascular disease or cirrhosis. Nat Rev Gastroenterol Hepatol 2013;10:330–344 3. White DL, Kanwal F, El-Serag HB. Association between nonalcoholic fatty liver disease and risk for hepatocellular cancer, based on systematic review. Clin Gastroenterol Hepatol 2012;10(1342–1359):e2 4. Adams LA, Lymp JF, St. Sauver J, Sanderson SO, Lindor KD, Feldstein A, et al. The natural history of nonalcoholic fatty liver disease: a population-based cohort study. Gastroenterology 2005;129:113–121 5. Promrat K, Kleiner DE, Niemeier HM, Jackvony E, Kearns M, Wands JR, et al. Randomized controlled trial testing the effects of weight loss on nonalcoholic steatohepatitis. Hepatology 2010;51:121–129 6. Chalasani N, Younossi Z, Lavine JE, Diehl AM, Brunt EM, Cusi K, 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;55:2005–2023 7. Starley BQ, Calcagno CJ, Harrison SA. Nonalcoholic fatty liver disease and hepatocellular carcinoma: a weighty connection. Hepatology 2010;51:1820–1832 8. Ratziu V, Bellentani S, Cortez-Pinto H, Day C, Marchesini G. A position statement on NAFLD/NASH based on the EASL 2009 special conference. J Hepatol 2010;53:372–384 9. Mathurin P, Hollebecque A, Arnalsteen L, Buob D, Leteurtre E, Caiazzo R, et al. Prospective study of the long-term effects of bariatric surgery on liver injury in patients without advanced disease. Gastroenterology 2009;137:532–540

123

10. Sanyal AJ, Brunt EM, Kleiner DE, Kowdley KV, Chalasani N, Lavine JE, et al. Endpoints and clinical trial design for nonalcoholic steatohepatitis. Hepatology 2011;54:344–353 11. Ryysy L, Hakkinen AM, Goto T, Vehkavaara S, Westerbacka J, Halavaara J, et al. Hepatic fat content and insulin action on free fatty acids and glucose metabolism rather than insulin absorption are associated with insulin requirements during insulin therapy in type 2 diabetic patients. Diabetes 2000;49:749–758 12. Bhalla K, Hwang BJ, Dewi RE, Twaddel W, Goloubeva OG, Wong KK, et al. Metformin prevents liver tumorigenesis by inhibiting pathways driving hepatic lipogenesis. Cancer Prev Res (Phila) 2012;5:544–552 13. Chen HP, Shieh JJ, Chang CC, Chen TT, Lin JT, Wu MS, et al. Metformin decreases hepatocellular carcinoma risk in a dosedependent manner: population-based and in vitro studies. Gut 2012;62:606–615 14. Zhang ZJ, Zheng ZJ, Shi R, Su Q, Jiang Q, Kip KE. Metformin for liver cancer prevention in patients with type 2 diabetes: a systematic review and meta-analysis. J Clin Endocrinol Metab 2012;97:2347–2353 15. Gastaldelli A, Harrison SA, Belfort-Aguilar R, Hardies LJ, Balas B, Schenker S, et al. Importance of changes in adipose tissue insulin resistance to histological response during thiazolidinedione treatment of patients with nonalcoholic steatohepatitis. Hepatology 2009;50:1087–1093 16. Bell LN, Wang J, Muralidharan S, Chalasani S, Fullenkamp AM, Wilson LA, et al. Relationship between adipose tissue insulin resistance and liver histology in nonalcoholic steatohepatitis: a pioglitazone versus vitamin E versus placebo for the treatment of nondiabetic patients with nonalcoholic steatohepatitis trial follow-up study. Hepatology 2012;56:1311–1318 17. 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– 2307 18. Aithal GP, Thomas JA, Kaye PV, Lawson A, Ryder SD, Spendlove I, et al. Randomized, placebo-controlled trial of pioglitazone in nondiabetic subjects with nonalcoholic steatohepatitis. Gastroenterology 2008;135:1176–1184 19. Sanyal AJ, Chalasani N, Kowdley KV, McCullough A, Diehl AM, Bass NM, et al. Pioglitazone, vitamin E, or placebo for nonalcoholic steatohepatitis. N Engl J Med 2010;362:1675– 1685 20. Ratziu V, Caldwell S, Neuschwander-Tetri BA. Therapeutic trials in nonalcoholic steatohepatitis: insulin sensitizers and related methodological issues. Hepatology 2010;52:2206–2215 21. Ratziu V, Giral P, Jacqueminet S, Charlotte F, HartemannHeurtier A, Serfaty L, et al. Rosiglitazone for nonalcoholic steatohepatitis: one-year results of the randomized placebocontrolled fatty liver improvement with rosiglitazone therapy (FLIRT) trial. Gastroenterology 2008;135:100–110 22. Ratziu V, Charlotte F, Bernhardt C, Giral P, Halbron M, Lenaour G, et al. Long-term efficacy of rosiglitazone in nonalcoholic steatohepatitis: results of the fatty liver improvement by rosiglitazone therapy (FLIRT 2) extension trial. Hepatology 2010;51:445–453 23. Mahady SE, Wong G, Craig JC, George J. Pioglitazone and vitamin E for nonalcoholic steatohepatitis: a cost utility analysis. Hepatology 2012;56:2172–2179 24. Balas B, Belfort R, Harrison SA, Darland C, Finch J, Schenker S, et al. Pioglitazone treatment increases whole body fat but not total body water in patients with non-alcoholic steatohepatitis. J Hepatol 2007;47:565–570 Epub 2007 May 24 25. Lincoff AM, Wolski K, Nicholls SJ, Nissen SE. Pioglitazone and risk of cardiovascular events in patients with type 2 diabetes

Hepatol Int

26.

27.

28.

29.

30.

31.

32.

33.

34.

35.

36.

37.

38.

39.

40.

41.

mellitus: a meta-analysis of randomized trials. JAMA 2007;298:1180–1188 Loke YK, Singh S, Furberg CD. Long-term use of thiazolidinediones and fractures in type 2 diabetes: a meta-analysis. CMAJ 2009;180:32–39 Neumann A, Weill A, Ricordeau P, Fagot JP, Alla F, Allemand H. Pioglitazone and risk of bladder cancer among diabetic patients in France: a population-based cohort study. Diabetologia 2012;55:1953–1962 Marchesini G, Brizi M, Bianchi G, Tomassetti S, Zoli M, Melchionda M. Metformin in nonalcoholic steatohepatitis. Lancet 2001;358:893–894 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 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 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 Haukeland JW, Konopski Z, Eggesbo HB, von Volkmann HL, Raschpichler G, Bjoro K, et al. Metformin in patients with nonalcoholic fatty liver disease: a randomized, controlled trial. Scand J Gastroenterol 2009;44:853–860 Tiikkainen M, Hakkinen AM, Korsheninnikova E, Nyman T, Makimattila S, Yki-Jarvinen H. Effects of rosiglitazone and metformin on liver fat content, hepatic insulin resistance, insulin clearance, and gene expression in adipose tissue in patients with type 2 diabetes. Diabetes 2004;53:2169–2176 Trevaskis JL, Griffin PS, Wittmer C, Neuschwander-Tetri BA, Brunt EM, Dolman CS, et al. Glucagon-like peptide-1 receptor agonism improves metabolic, biochemical, and histopathological indices of nonalcoholic steatohepatitis in mice. Am J Physiol Gastrointest Liver Physiol 2012;302:G762–772 Kim YO, Schuppan D. When GLP-1 hits the liver: a novel approach for insulin resistance and NASH. Am J Physiol Gastrointest Liver Physiol 2012;302:G759–761 Svegliati-Baroni G, Saccomanno S, Rychlicki C, Agostinelli L, De Minicis S, Candelaresi C, et al. Glucagon-like peptide-1 receptor activation stimulates hepatic lipid oxidation and restores hepatic signalling alteration induced by a high-fat diet in nonalcoholic steatohepatitis. Liver Int 2011;31:1285–1297 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 Dufour JF, Oneta CM, Gonvers JJ, Bihl F, Cerny A, Cereda JM, et al. Randomized placebo-controlled trial of ursodeoxycholic acid with vitamin e in nonalcoholic steatohepatitis. Clin Gastroenterol Hepatol 2006;4:1537–1543 Lavine JE, Schwimmer JB, Van Natta ML, Molleston JP, Murray KF, Rosenthal P, et al. Effect of vitamin E or metformin for treatment of nonalcoholic fatty liver disease in children and adolescents: the TONIC randomized controlled trial. JAMA 2011;305:1659–1668 Nobili V, Manco M, Devito R, Di Ciommo V, Comparcola D, Sartorelli MR, et al. Lifestyle intervention and antioxidant therapy in children with nonalcoholic fatty liver disease: a randomized, controlled trial. Hepatology 2008;48:119–128 Miller ER 3rd, Pastor-Barriuso R, Dalal D, Riemersma RA, Appel LJ, Guallar E. Meta-analysis: high-dosage vitamin E supplementation may increase all-cause mortality. Ann Intern Med 2005;142:37–46

42. Schurks M, Glynn RJ, Rist PM, Tzourio C, Kurth T. Effects of vitamin E on stroke subtypes: meta-analysis of randomised controlled trials. BMJ 2010;341:c5702 43. Klein EA, Thompson IM Jr, Tangen CM, Crowley JJ, Lucia MS, Goodman PJ, et al. Vitamin E and the risk of prostate cancer: the selenium and vitamin E cancer prevention trial (SELECT). JAMA 2011;306:1549–1556 44. Laurin J, Lindor KD, Crippin JS, Gossard A, Gores GJ, Ludwig J, et al. Ursodeoxycholic acid or clofibrate in the treatment of non-alcohol-induced steatohepatitis: a pilot study. Hepatology 1996;23:1464–1467 45. Lindor KD, Kowdley KV, Heathcote EJ, Harrison ME, Jorgensen R, Angulo P, et al. Ursodeoxycholic acid for treatment of nonalcoholic steatohepatitis: results of a randomized trial. Hepatology 2004;39:770–778 46. Leuschner UF, Lindenthal B, Herrmann G, Arnold JC, Rossle M, Cordes HJ, et al. High-dose ursodeoxycholic acid therapy for nonalcoholic steatohepatitis: a double-blind, randomized, placebo-controlled trial. Hepatology 2010;52:472–479 47. Ratziu V. Treatment of NASH with ursodeoxycholic acid: pro. Clin Res Hepatol Gastroenterol 2012;36(Suppl 1):S41–45 48. Ratziu V, de Ledinghen V, Oberti F, Mathurin P, WartelleBladou C, Renou C, et al. A randomized controlled trial of highdose ursodesoxycholic acid for nonalcoholic steatohepatitis. J Hepatol 2011;54:1011–1019 49. Adams LA, Zein CO, Angulo P, Lindor KD. A pilot trial of pentoxifylline in nonalcoholic steatohepatitis. Am J Gastroenterol 2004;99:2365–2368 50. Zein CO, Yerian LM, Gogate P, Lopez R, Kirwan JP, Feldstein AE, et al. Pentoxifylline improves nonalcoholic steatohepatitis: a randomized placebo-controlled trial. Hepatology 2011;54: 1610–1619 51. Zein CO, Lopez R, Fu X, Kirwan JP, Yerian LM, McCullough AJ, et al. Pentoxifylline decreases oxidized lipid products in nonalcoholic steatohepatitis: new evidence on the potential therapeutic mechanism. Hepatology 2012;56:1291–1299 52. Yokohama S, Yoneda M, Haneda M, Okamoto S, Okada M, Aso K, et al. Therapeutic efficacy of an angiotensin II receptor antagonist in patients with nonalcoholic steatohepatitis. Hepatology 2004;40:1222–1225 53. Rosselli MS, Burgueno AL, Carabelli J, Schuman M, Pirola CJ, Sookoian S. Losartan reduces liver expression of plasminogen activator inhibitor-1 (PAI-1) in a high fat-induced rat nonalcoholic fatty liver disease model. Atherosclerosis 2009;206: 119–126 54. Torres DM, Jones FJ, Shaw JC, Williams CD, Ward JA, Harrison SA. Rosiglitazone versus rosiglitazone and metformin versus rosiglitazone and losartan in the treatment of nonalcoholic steatohepatitis in humans: a 12-month randomized, prospective, open-label trial. Hepatology 2011;54:1631–1639 55. Sookoian S, Castano G, Garcia SI, Viudez P, Gonzalez C, Pirola CJ. A1166C angiotensin II type 1 receptor gene polymorphism may predict hemodynamic response to losartan in patients with cirrhosis and portal hypertension. Am J Gastroenterol 2005;100: 636–642 56. Ekstedt M, Franzen LE, Mathiesen UL, Holmqvist M, Bodemar G, Kechagias S. Statins in non-alcoholic fatty liver disease and chronically elevated liver enzymes: a histopathological followup study. J Hepatol 2007;47:135–141 57. 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

123

Hepatol Int 58. Athyros VG, Tziomalos K, Gossios TD, Griva T, Anagnostis P, Kargiotis K, et al. Safety and efficacy of long-term statin treatment for cardiovascular events in patients with coronary heart disease and abnormal liver tests in the Greek Atorvastatin and Coronary Heart Disease Evaluation (GREACE) Study: a post hoc analysis. Lancet 2010;376:1916–1922 59. Loria P, Lonardo A, Anania F. Liver and diabetes. A vicious circle. Hepatol Res 2013;43:51–64 60. Yilmaz Y, Colak Y, Kurt R, Senates E, Eren F. Linking nonalcoholic fatty liver disease to hepatocellular carcinoma: from bedside to bench and back. Tumori 2013;99:10–16 61. Lai SW, Liao KF, Lai HC, Muo CH, Sung FC, Chen PC. Statin use and risk of hepatocellular carcinoma. Eur J Epidemiol 2013;28:485–492 62. Pradelli D, Soranna D, Scotti L, Zambon A, Catapano A, Mancia G, et al. Statins and primary liver cancer: a meta-analysis of observational studies. Eur J Cancer Prev 2013;22:229–234 63. Lonardo A, Loria P. Potential for statins in the chemoprevention and management of hepatocellular carcinoma. J Gastroenterol Hepatol 2012;27:1654–1664 64. Singh S, Singh PP, Singh AG, Murad MH, Sanchez W. Statins are associated with a reduced risk of hepatocellular cancer: a systematic review and meta-analysis. Gastroenterology 2013;144:323–332 65. Merat S, Malekzadeh R, Sohrabi MR, Sotoudeh M, Rakhshani N, Sohrabpour AA, et al. Probucol in the treatment of nonalcoholic steatohepatitis: a double-blind randomized controlled study. J Hepatol 2003;38:414–418 66. Merat S, Aduli M, Kazemi R, Sotoudeh M, Sedighi N, Sohrabi M, et al. Liver histology changes in nonalcoholic steatohepatitis after one year of treatment with probucol. Dig Dis Sci 2008;53:2246–2250 67. Capanni M, Calella F, Biagini MR, Genise S, Raimondi L, Bedogni G, et al. Prolonged n-3 polyunsaturated fatty acid supplementation ameliorates hepatic steatosis in patients with non-alcoholic fatty liver disease: a pilot study. Aliment Pharmacol Ther 2006;23:1143–1151 68. Masterton GS, Plevris JN, Hayes PC. Review article: omega-3 fatty acids—a promising novel therapy for non-alcoholic fatty liver disease. Aliment Pharmacol Ther 2010;31:679–692 69. Parker HM, Johnson NA, Burdon CA, Cohn JS, O’Connor HT, George J. Omega-3 supplementation and non-alcoholic fatty liver disease: a systematic review and meta-analysis. J Hepatol 2012;56:944–951 70. Harrison SA, Fecht W, Brunt EM, Neuschwander-Tetri BA. Orlistat for overweight subjects with nonalcoholic steatohepatitis: a randomized, prospective trial. Hepatology 2009;49:80–86 71. Zoppini G, Targher G, Bonora E. The role of serum uric acid in cardiovascular disease in type 2 diabetic and non-diabetic subjects: a narrative review. J Endocrinol Invest 2011;34:881–886 72. Loria P, Lonardo A, Targher G. Is liver fat detrimental to vessels?: intersections in the pathogenesis of NAFLD and atherosclerosis. Clin Sci (Lond) 2008;115:1–12 73. Valbusa F, Bertolini L, Bonapace S, Zenari L, Zoppini G, Arcaro G, et al. Relation of elevated serum uric acid levels to incidence of atrial fibrillation in patients with type 2 diabetes mellitus. Am J Cardiol 2013. doi:10.1016/j.amjcard.2013.04.012 74. Li Y, Xu C, Yu C, Xu L, Miao M. Association of serum uric acid level with non-alcoholic fatty liver disease: a cross-sectional study. J Hepatol 2009;50:1029–1034 75. Sirota JC, McFann K, Targher G, Johnson RJ, Chonchol M, Jalal DI. Elevated serum uric acid levels are associated with nonalcoholic fatty liver disease independently of metabolic syndrome features in the United States: liver ultrasound data from

123

76.

77.

78.

79.

80.

81.

82.

83.

84.

85.

86.

87.

88.

89.

90.

91.

92.

the National Health and Nutrition Examination Survey. Metabolism 2013;62:392–399 Afzali A, Weiss NS, Boyko EJ, Ioannou GN. Association between serum uric acid level and chronic liver disease in the United States. Hepatology 2010;52:578–589 Xu CF, Yu CH, Xu L, Sa XY, Li YM. Hypouricemic therapy: a novel potential therapeutic option for nonalcoholic fatty liver disease. Hepatology 2010;52:1865–1866 Dongiovanni P, Fracanzani AL, Fargion S, Valenti L. Iron in fatty liver and in the metabolic syndrome: a promising therapeutic target. J Hepatol 2011;55:920–932 Kowdley KV, Belt P, Wilson LA, Yeh MM, NeuschwanderTetri BA, Chalasani N, et al. Serum ferritin is an independent predictor of histologic severity and advanced fibrosis in patients with nonalcoholic fatty liver disease. Hepatology 2012;55:77–85 Dongiovanni P, Valenti L, Fracanzani AL, Gatti S, Cairo G, Fargion S. Iron depletion by deferoxamine up-regulates glucose uptake and insulin signaling in hepatoma cells and in rat liver. Am J Pathol 2008;172:738–747 Fargion S, Dongiovanni P, Guzzo A, Colombo S, Valenti L, Fracanzani AL. Iron and insulin resistance. Aliment Pharmacol Ther 2005;22(Suppl 2):61–63 Equitani F, Fernandez-Real JM, Menichella G, Koch M, Calvani M, Nobili V, et al. Bloodletting ameliorates insulin sensitivity and secretion in parallel to reducing liver iron in carriers of HFE gene mutations. Diabetes Care 2008;31:3–8 Valenti L, Moscatiello S, Vanni E, Fracanzani AL, Bugianesi E, Fargion S, et al. Venesection for non-alcoholic fatty liver disease unresponsive to lifestyle counselling–a propensity scoreadjusted observational study. QJM 2011;104:141–149 Valenti L, Fracanzani AL, Dongiovanni P, Bugianesi E, Marchesini G, Manzini P, et al. Iron depletion by phlebotomy improves insulin resistance in patients with nonalcoholic fatty liver disease and hyperferritinemia: evidence from a case-control study. Am J Gastroenterol 2007;102:1251–1258 Facchini FS, Hua NW, Stoohs RA. Effect of iron depletion in carbohydrate-intolerant patients with clinical evidence of nonalcoholic fatty liver disease. Gastroenterology 2002;122:931–999 Valenti L, Dongiovanni P, Fracanzani AL, Fargion S. Bloodletting ameliorates insulin sensitivity and secretion in parallel to reducing liver iron in carriers of HFE gene mutations: response to Equitani et al. Diabetes Care 2008;31:e18 author reply e19 Riquelme A, Soza A, Nazal L, Martinez G, Kolbach M, Patillo A, et al. Histological resolution of steatohepatitis after iron depletion. Dig Dis Sci 2004;49:1012–1015 Valenti L, Dongiovanni P, Fargion S. Diagnostic and therapeutic implications of the association between ferritin level and severity of nonalcoholic fatty liver disease. World J Gastroenterol 2012;18:3782–3786 Lirussi F, Mastropasqua E, Orando S, Orlando R. Probiotics for non-alcoholic fatty liver disease and/or steatohepatitis. Cochrane Database Syst Rev 2007;21:CD005165 Velayudham A, Dolganiuc A, Ellis M, Petrasek J, Kodys K, Mandrekar P, et al. VSL#3 probiotic treatment attenuates fibrosis without changes in steatohepatitis in a diet-induced nonalcoholic steatohepatitis model in mice. Hepatology 2009;49:989–997 Cani PD, Possemiers S, Van de Wiele T, Guiot Y, Everard A, Rottier O, et al. Changes in gut microbiota control inflammation in obese mice through a mechanism involving GLP-2-driven improvement of gut permeability. Gut 2009;58:1091–1103 Cani PD, Bibiloni R, Knauf C, Waget A, Neyrinck AM, Delzenne NM, et al. Changes in gut microbiota control metabolic

Hepatol Int

93.

94.

95.

96.

97.

98.

99.

100.

101. 102.

103. 104. 105.

endotoxemia-induced inflammation in high-fat diet-induced obesity and diabetes in mice. Diabetes 2008;57:1470–1481 Membrez M, Blancher F, Jaquet M, Bibiloni R, Cani PD, Burcelin RG, et al. Gut microbiota modulation with norfloxacin and ampicillin enhances glucose tolerance in mice. FASEB J 2008;22:2416–2426 Bergheim I, Weber S, Vos M, Kramer S, Volynets V, Kaserouni S, et al. Antibiotics protect against fructose-induced hepatic lipid accumulation in mice: role of endotoxin. J Hepatol 2008;48:983–992 Cani PD, Lecourt E, Dewulf EM, Sohet FM, Pachikian BD, Naslain D, et al. Gut microbiota fermentation of prebiotics increases satietogenic and incretin gut peptide production with consequences for appetite sensation and glucose response after a meal. Am J Clin Nutr 2009;90:1236–1243 Parnell JA, Reimer RA. Weight loss during oligofructose supplementation is associated with decreased ghrelin and increased peptide YY in overweight and obese adults. Am J Clin Nutr 2009;89:1751–1759 Ma X, Hua J, Li Z. Probiotics improve high fat diet-induced hepatic steatosis and insulin resistance by increasing hepatic NKT cells. J Hepatol 2008;49:821–830 Mencarelli A, Cipriani S, Renga B, Bruno A, D’Amore C, Distrutti E, et al. VSL#3 resets insulin signaling and protects against NASH and atherosclerosis in a model of genetic dyslipidemia and intestinal inflammation. PLoS ONE 2012;7:e45425 Loguercio C, Federico A, Tuccillo C, Terracciano F, D’Auria MV, De Simone C, et al. Beneficial effects of a probiotic VSL#3 on parameters of liver dysfunction in chronic liver diseases. J Clin Gastroenterol 2005;39:540–543 Solga SF, Buckley G, Clark JM, Horska A, Diehl AM. The effect of a probiotic on hepatic steatosis. J Clin Gastroenterol 2008;42:1117–1119 Machado MV, Cortez-Pinto H. Gut microbiota and nonalcoholic fatty liver disease. Ann Hepatol 2012;11:440–449 Iacono A, Raso GM, Canani RB, Calignano A, Meli R. Probiotics as an emerging therapeutic strategy to treat NAFLD: focus on molecular and biochemical mechanisms. J Nutr Biochem 2011;22:699–711 Lam B, Younossi ZM. Treatment options for nonalcoholic fatty liver disease. Therap Adv Gastroenterol 2010;3:121–137 Walter JH. L-Carnitine. Arch Dis Child 1996;74:475–478 Malaguarnera M, Gargante MP, Russo C, Antic T, Vacante M, Avitabile T, et al. L-Carnitine supplementation to diet: a new tool in treatment of nonalcoholic steatohepatitis—a randomized

106.

107.

108.

109.

110.

111.

112.

113.

114.

115.

116.

and controlled clinical trial. Am J Gastroenterol 2010;105:1338– 1345 Carulli L, Ballestri S, Lonardo A, Lami F, Violi E, Losi L, et al. Is nonalcoholic steatohepatitis associated with a high-thoughnormal thyroid stimulating hormone level and lower cholesterol levels? Intern Emerg Med 2013;8:297–305 Cachefo A, Boucher P, Vidon C, Dusserre E, Diraison F, Beylot M. Hepatic lipogenesis and cholesterol synthesis in hyperthyroid patients. J Clin Endocrinol Metab 2001;86:5353–5357 Riis AL, Gravholt CH, Djurhuus CB, Norrelund H, Jorgensen JO, Weeke J, et al. Elevated regional lipolysis in hyperthyroidism. J Clin Endocrinol Metab 2002;87:4747–4753 Cable EE, Finn PD, Stebbins JW, Hou J, Ito BR, van Poelje PD, et al. Reduction of hepatic steatosis in rats and mice after treatment with a liver-targeted thyroid hormone receptor agonist. Hepatology 2009;49:407–417 Arrese M. Burning hepatic fat: therapeutic potential for liverspecific thyromimetics in the treatment of nonalcoholic fatty liver disease. Hepatology 2009;49:348–351 Zhang S, Wang J, Liu Q, Harnish DC. Farnesoid X receptor agonist WAY-362450 attenuates liver inflammation and fibrosis in murine model of non-alcoholic steatohepatitis. J Hepatol 2009;51:380–388 Ratziu V, Sheikh MY, Sanyal AJ, Lim JK, Conjeevaram H, Chalasani N, et al. A phase 2, randomized, double-blind, placebo-controlled study of GS-9450 in subjects with nonalcoholic steatohepatitis. Hepatology 2012;55:419–428 Cariou B, Zair Y, Staels B, Bruckert E. Effects of the new dual PPAR alpha/delta agonist GFT505 on lipid and glucose homeostasis in abdominally obese patients with combined dyslipidemia or impaired glucose metabolism. Diabetes Care 2011;34:2008–2014 Tsuchiya H, Ikeda Y, Ebata Y, Kojima C, Katsuma R, Tsuruyama T, et al. Retinoids ameliorate insulin resistance in a leptin-dependent manner in mice. Hepatology 2012;56:1319– 1330 Hoo RL, Lee IP, Zhou M, Wong JY, Hui X, Xu A, et al. Pharmacological inhibition of adipocyte fatty acid binding protein alleviates both acute liver injury and non-alcoholic steatohepatitis in mice. J Hepatol 2013;58:358–364 Lin CW, Zhang H, Li M, Xiong X, Chen X, Dong XC. Pharmacological promotion of autophagy alleviates steatosis and injury in alcoholic and non-alcoholic fatty liver conditions in mice. J Hepatol 2013. doi:10.1016/j.jhep.2013.01.011

123

Pharmacological agents for nonalcoholic steatohepatitis.

The rationale for specific pharmacologic therapy in nonalcoholic steatohepatitis (NASH) is determined by the potential for disease progression and the...
1KB Sizes 1 Downloads 18 Views