Clin Mol Hepatol > Volume 29(Suppl); 2023 > Article
An and Sohn: Pharmacological advances in the treatment of nonalcoholic fatty liver diseases : focused on global results of randomized controlled trials

ABSTRACT

Nonalcoholic fatty liver disease (NAFLD) is the most common cause of liver disease globally, and its prevalence is rapidly increasing. Nonalcoholic steatohepatitis (NASH), a progressive form of NAFLD, is characterized by hepatocellular injury, inflammation, and fibrosis. Patients with NASH or severe fibrosis should be treated according to international NAFLD guidelines. Currently, regulatory agencies have not approved any pharmaceutical treatment for NAFLD. Vitamin E and pioglitazone are efficacious for NASH resolution; however, their benefits must be weighed against the reported risks. In a phase 2 trial, a glucagon-like peptide-1 agonist commonly used for diabetes and obesity was found to improve liver histology in patients with NASH. Furthermore, therapeutic agents targeting NASH pathogenesis, including bile acid signaling, insulin resistance, and lipid metabolism, are in various phases of clinical development. In this article, we review the benefits and drawbacks of current pharmacotherapy and the efficacy of upcoming treatments for NASH.

INTRODUCTION

Nonalcoholic fatty liver disease (NAFLD) affects approximately one-quarter of the adult population worldwide, making it the most common liver disease [1]. Nonalcoholic steatohepatitis (NASH), the progressive form of NAFLD, is characterized by hepatic triglyceride accumulation, hepatocyte injury, and lobular inflammation [2]. NASH is associated with accelerated fibrosis progression to cirrhosis and increased morbidity and mortality from liver disease [3]. More than 20% of patients with NASH will develop cirrhosis during their lifespan [4]. NASH is the leading indication for liver transplant in the United States [5], and it is expected to become the most common cause of hepatocellular carcinoma in developed countries [6].
Patients with NAFLD should be encouraged to lose weight by following a hypocaloric diet and engaging in physical activity [2,7]. In patients with NASH who are overweight or obese, more than 10% of weight loss due to lifestyle modification is associated with NASH resolution and fibrosis regression [8,9]. Weight loss also leads to a reduction of liver fat content in non-obese patients with NAFLD [10]. However, only a small percentage of patients achieve substantial weight loss, and long-term lifestyle changes are difficult to implement [8,11]. Therefore, patients with NASH require a practical therapeutic approach.
Currently, there are no licensed drugs specifically approved for the treatment of NASH. In clinical practice, vitamin E and pioglitazone are efficacious for biopsy-proven NASH [12]. Furthermore, glucagon-like peptide 1 (GLP-1) agonists, which are commonly prescribed medications for diabetes and obesity, have the potential to ameliorate NASH [13]. The field of NASH treatment is rapidly evolving owing to the rising disease incidence and scarcity of current treatment options. Because the underlying mechanism of NASH is complex, NASH treatments are being developed for a wide range of targets, including oxidative stress, insulin resistance, apoptosis, bile acids, lipid metabolism, and hepatic inflammation and fibrosis. In this article, we review and summarize the efficacy and safety of current treatment options, based primarily on representative data from randomized controlled trials (RCTs), as well as emerging therapies that may enter clinical practice in the future.

CURRENT PHARMACOLOGIC THERAPIES

Vitamin E (alpha-tocopherol)

The imbalance between the reactive oxygen species’ production and scavenging capacity causes oxidative stress [14]. Excess hepatic lipid causes reactive oxygen species over-production, accelerating the transition from NAFLD to NASH [14]. Vitamin E shows antioxidant properties by increasing specific enzymes and anti-fibrotic actions by regulating the inflammatory response [15]. In phase 3 PIVENS trial, patients with NASH without diabetes who received high dose vitamin E (800 IU/day; n=84) for 96 weeks showed a more statistically significant histological improvement, defined as ≥2 point reduction in the NAFLD activity score, than the placebo group (n=83) (43% vs. 19%) [12]. The proportion of NASH resolution in the vitamin E group was also higher (36% vs. 21%). Recent prospective trials involving patients with NASH and diabetes, found that a combination treatment of vitamin E (800 IU/day) and pioglitazone is more efficacious than a placebo in terms of NASH resolution and steatosis improvement [16]. No prospective randomized studies have reported improved liver fibrosis and reduced liver-related death [16]. The international NAFLD guidelines suggest vitamin E supplementation for patients with NASH without diabetes (Table 1) [2,7,17]. Unfortunately, although controversial, long-term administration of vitamin E is likely to raise the incidence of prostate cancer and hemorrhagic stroke [18].

Pioglitazone

Pioglitazone, a peroxisome proliferator-activated receptor (PPAR)-γ agonist, reduces insulin resistance in the adipose tissue, muscle, and liver. Several prospective trials reported that patients with or without diabetes who received pioglitazone (30 or 45 mg/day) showed more histological improvement in NASH than those who received placebo [12,19,20]. Cusi et al. [21] conducted a single-center study in which patients with prediabetes/diabetes and histologically confirmed NASH were randomly administered either pioglitazone (45 mg/day; n=50) or placebo (n=51). Pioglitazone treatment reduced NAFLD activity score by at least 2 points (58% vs. 17%) and resolved NASH (51% vs. 19%). A meta-analysis of eight RCTs found pioglitazone is efficacious for NASH resolution (odds ratio [OR] 3.22), improvement of advanced fibrosis (OR 3.15), and reversal of fibrosis (OR 1.66) [22]. Thus, regardless of the diabetes status, pioglitazone is indicated for biopsy-proven patients with NASH (Table 1) [2,7,17]. It is important to note that weight gain, fluid retention, and increased risk of fracture and bladder cancer are side effects of pioglitazone.

GLP-1 agonists

GLP-1 agonists affect glucose regulation by enhancing glucose-dependent insulin release, suppressing postprandial glucagon levels, and slowing gastric emptying. GLP-1 agonist is the mainstay treatment of obesity and diabetes because of their significant therapeutic benefits in weight loss, glycemic control, and improvements in the cardiometabolic system [23]. Although the underlying mechanisms of GLP-1 agonists on NASH have not been fully explained, considerable weight loss induced by GLP-1 agonists may lead to subsequent disease improvement. A phase 2 RCT with 320 biopsy-confirmed patients with NASH found that the semaglutide group (0.4 mg once daily for 72 weeks) had a higher proportion of disease resolution than the placebo group (59% vs. 17%) [24]. Even though the treatment group had a lower rate of liver fibrosis progression (4.9% vs. 18.8%), there were no significant differences in the proportion of patients whose fibrosis stage improved. The American Association of Clinical Endocrinology guidelines recommend the use of GLP-1 agonist in patients with histology-proven NASH and diabetes [17]. A phase 3 ESSENCE trial involving 1,200 patients with NASH and F2-F3 fibrosis is currently investigating the efficacy of semaglutide at a dose of 2.4 mg once-weekly for NASH resolution and fibrosis improvement (NCT04822181; Table 2). The most common side effects among patients that receive GLP-1 agonist are gastrointestinal symptoms, such as nausea, vomiting, and diarrhea. GLP-1 agonists may increase the risk of acute pancreatitis, gallbladder disease, and biliary disease. Although GLP-1 agonists are currently used as subcutaneous injections in clinical protocols, oral formulations with improved tolerability are being developed.
Recently, advances have been made in developing glucagon-containing co-agonists to enhance the efficacy of GLP-1 agonists. A glucagon-stimulated increase in energy expenditure augments the effect of GLP-1-induced weight loss [25]. Cotadutide is a dual-receptor agonist with balanced GLP-1 and glucagon action. In phase 2 PROXYMO trial, 74 obese patients with biopsy-proven NASH and F1-F3 fibrosis were randomized to receive once-daily subcutaneous injections of cotadutide (300 μg or 600 μg) or placebo [26]. Cotadutide was associated with dose-dependent reductions in hepatic fat compared to the placebo. In the ongoing phase 3 PROXYMOADV trial, cotadutide is expected to show efficacy in treating NASH (Table 2).

FUTURE PHARMACOLOGIC THERAPIES

Obeticholic acid

The farnesoid X receptor is a nuclear receptor activated by bile acids that is abundant in the liver and intestines. It regulates bile synthesis, conjugation, and transport [27,28], and plays a role in lipid and glucose metabolism [28]. The farnesoid X receptor activation can help reduce hepatic inflammation and fibrosis [29,30].
Obeticholic acid is a potent and selective farnesoid X receptor agonist. In the interim analysis of phase 3 REGENERATE trial, 931 biopsy-proven patients with NASH and fibrosis stages F2-F3 were randomly assigned to receive obeticholic acid 25 mg daily (n=308), obeticholic acid 10 mg daily (n=312), or placebo (n=311) (Table 2) [31]. At 18 months, the obeticholic acid group improved liver fibrosis by at least one stage with no worsening of NASH in a dose-dependent manner (23% vs. 18% vs. 12%, respectively), with no difference in the proportion of NASH resolution (12% vs. 11% vs. 8%, respectively). Indeed, in NASH phase 3 trials, obeticholic acid was the first agent to show a significant improvement in fibrosis. Mild to moderate pruritus was the most common adverse event, affecting up to 51% of patients treated with obeticholic acid 25 mg. Furthermore, nearly 17% of the obeticholic acid group experienced an early increase in low-density lipoprotein cholesterol, which returned to baseline levels at the end of the study. In contrast, in the recent REVERSE trials of 919 randomized patients with compensated NASH cirrhosis, obeticholic acid did not improve fibrosis (11.1% vs. 11.9% vs. 9.9% in obeticholic acid 10 mg vs. obeticholic acid 10 mg titrated to 25 mg vs. placebo, respectively) [32]. The US Food and Drug Administration has not yet approved obeticholic acid as a NASH treatment due to its uncertain long-term benefit and safety risks.

pan-PPAR agonist

PPARs are a nuclear receptor family with three isotypes that regulate glucose and lipid metabolism, inflammatory cell activation, and fibrotic processes [33]. Three PPAR isotypes have been identified: PPAR-α, PPAR-β/δ, and PPAR-γ. PPAR-α is an essential regulator of fatty acid oxidation that suppresses inflammation by reducing reactive oxygen species formation. PPAR-β/δ stimulates hepatic glucose utilization and de novo lipogenesis. PPAR-γ regulates adipocyte differentiation and insulin sensitization.
Lanifibranor (IVA337), a pan-PPAR agonist, demonstrated higher efficacy in terms of improvement of insulin sensitivity, macrophage activation, and reduction of liver fibrosis than single or dual PPAR agonists [34,35]. In 2021, the results of phase 2b trials comparing lanifibranor 1,200 mg (n=83), lanifibranor 800 mg (n=83), or placebo (n=81) for 24 weeks in patients with biopsy-proven NASH were published [36]. The proportion of patients who met the primary endpoint, a decrease of at least 2 points in the SAF-activity score (the activity component of the Steatosis, Activity, Fibrosis [SAF] scoring system that includes hepatocytes ballooning and inflammation), was higher among those who received lanifibranor 1,200 mg than the placebo group (55% vs. 33%). The outcomes favored lanifibranor 1,200 mg over placebo for improvement in the fibrosis stage of at least one without worsening of NASH (48% vs. 29%). Fewer than 10% of patients in the lanifibranor group reported diarrhea, weight gain, and peripheral edema as common adverse effects. An ongoing phase 3 study of lanifibranor for NASH and F2-F3 fibrosis (NATiV3) is also expected to reveal similar results (Table 2).
In contrast, a phase 3 RCT of the dual PPARα–PPARδ agonist elafibranor (RESOLVE- IT) was halted because it failed to meet the predefined primary surrogate efficacy endpoint, NASH resolution without fibrosis worsening the interim analysis [37].

Thyroid hormone receptor β-agonist

The thyroid hormone regulates glucose and lipid metabolism, in addition to fatty acids oxidation [38,39]. A selective thyroid hormone receptor beta (THR-β) agonist has been developed to improve liver-specific action while minimizing negative effects on the cardiac and skeletal systems, which are predominantly mediated by THR alpha. Resmetirom, an oral THR-β agonist, was studied in a phase 2 RCT involving 125 overweight or obese adults with biopsy-confirmed NASH and stages 1–3 fibrosis [40]. Resmetirom treatment for 36 weeks resulted in a significant reduction in hepatic fat measured using magnetic resonance imaging (MRI)-proton density fat fraction compared with placebo (-37% vs. -9%). An ongoing phase 3 MAESTRO-NAFLD1 trial is evaluating the impact of resmetirom on liver histology in patients with NASH and stage 2–3 fibrosis (Table 2). The preliminary results showed that resmetirom was efficacious for hepatic fat assessed using MRI-proton density fat fraction [41]. The most prevalent side effects were mild gastrointestinal symptoms, including diarrhea and nausea.

Selonsertib

Apoptosis-signal regulating kinase 1 (ASK1) is a member of the mitogen-activated protein kinase (MAPK) family [42]. ASK1 is activated in response to oxidative stress and promotes hepatic inflammation and apoptosis, leading to liver fibrogenesis via MAPK downstream signaling. Hence, ASK1 is considered a treatment target for NASH [43]. Selonsertib is a first-in-class small-molecule ASK1 inhibitor with antifibrotic and anti-inflammatory effects. Based on the success in phase 2 trials of selonsertib in patients with NASH and F2-F3 fibrosis [44], phase 3 RCTs comparing selonsertib 18 mg, selonsertib 6 mg, and placebo were subsequently conducted in patients with NASH and bridging fibrosis (F3, STELLAR-3; n=802) or compensated cirrhosis (F4, STELLAR-4; n=877) (Table 2) [45]. The STELLAR-3 trial did not reveal significantly different fibrosis improvement without worsening of NASH between groups (10% vs. 12% vs. 13%, respectively). Moreover, fibrosis improvement was not observed in STELLAR-4 patients with cirrhosis (14% vs. 13% vs. 13%, respectively). In phase 2b ATLAS trial with 392 patients with NASH and F3-F4 fibrosis, selonsertib combination therapy revealed unfavorable outcomes in reversing fibrosis [46]. Although selonsertib is no longer being investigated, ASK1 may still be a viable candidate if more effective inhibitors are discovered.

Other NASH therapies in clinical trials

The novel medications that have entered phase 3 development stage include armachol (a bile acid and fatty acid analog) [47] and cenicriviroc (inhibitor of CC chemokine receptors 2 and 5) (Table 2) [48]. Moreover, a large number of additional agents with diverse mechanisms for targeting the pathogenesis of NASH are in phase 2 development [49].

CONCLUSIONS

Since the PIVENS study with vitamin E and pioglitazone on NASH resolution was successful in 2010, NASH has been extensively investigated to identify optimal medications. Large-scale RCTs have yielded promising results for farnesoid X receptor, GLP-1, and pan-PPAR agonists in improving hepatic inflammation and fibrosis. However, several obstacles must be overcome before they are approved by the US Food and Drug Administration for NASH treatment: 1) while liver biopsy remains the gold standard for diagnosis in clinical trials, further studies are needed to develop easy-to-use panels of serum and imaging-based biomarkers for noninvasive patient selection and treatment response; 2) given the complex pathophysiology of NASH and modest treatment response rates to individual drugs, it is highly likely that a combination treatment will also be required; and 3) the external validity of the RCT results should be confirmed, especially for real-world patients with NASH with more significant comorbidities. We believe that numerous drugs added to the pipeline of novel therapies could increase the chances of successful treatment of NASH and more completely reverse disease progression in affected patients in the future.

ACKNOWLEDGMENTS

This study was supported by grant from the National Research Foundation of Korea funded by the Ministry of Science and ICT (RS-2022-00166674).

FOOTNOTES

Authors’ contribution
Study concept and design: Jihyun An and Joo Hyun Sohn; Data analysis and interpretation: Jihyun An and Joo Hyun Sohn; Wrote the paper: Jihyun An and Joo Hyun Sohn; All authors have read and approved the final version of the manuscript.
Conflicts of Interest
The authors have no conflicts to disclose.

Table 1.
Summary of current NASH medications recommended by international guidelines
Drugs Mechanism Population Guidelines (level of recommendation)
Vitamin E Anti-oxidant Non-diabetic patients with biopsy-proven NASH AASLD 2018*
EASL 2016 (B2)
KASL 2021 (B1)
AACE 2022 (Grade B, high strength of evidence)
Pioglitazone PPAR-γ agonist Diabetic patients with biopsy- proven NASH AASLD 2018*
EASL 2016 (B2)
KASL 2021 (B1)
AACE 2022 (Grade A, high strength of evidence)

NASH, nonalcoholic steatohepatitis; PPAR, peroxisome proliferator-activated receptor; AASLD, American Association for the Study of Liver Diseases; EASL, European Association for the Study of the Liver; KASL, Korean Association for the Study of the Liver; AACE, American Association of Clinical Endocrinology.

* The level and length of recommendations were not presented in the AASLD guidance 2018.

Table 2.
Current status of emerging drugs from phase 3 clinical trials of nonalcoholic steatohepatitis
Drug Target Population Study name Status
Obeticholic acid Farnesoid X receptor agonist NASH with F2-F3 fibrosis REGENERATE Ongoing
Lanifibranor Pan-PPAR agonist NASH with F2-F3 fibrosis NATiV3 Ongoing
Resmetirom Thyroid hormone receptor-beta agonist NASH with F1-F3 fibrosis MAESTRO-NASH Ongoing
Semaglutide Glucagon-like peptide-1 (GLP-1) agonist NASH with F2-F3 fibrosis ESSENCE Ongoing
Cotadutide dual GLP-1 and glucagon receptor agonist NASH with F2-F3 fibrosis PROXYMO-ADV Ongoing
Obeticholic acid Farnesoid X receptor agonist NASH with compensated LC REVERSE Halted
Elafibranor PPAR-alpha and -delta agonist NASH with F1-F3 fibrosis RESOLVE-IT Halted
Selonsertib Apoptosis signal-regulating kinase inhibitor NASH with F3 fibrosis STELLAR-3 Halted
Selonsertib Apoptosis signal-regulating kinase inhibitor NASH with compensated LC STELLAR-4 Halted
Cenicriviroc Inhibitor of CC chemokine receptors 2 and 5 NASH with F2-F3 fibrosis AURORA Halted
Aramchol Fatty acid bile acid conjugate NASH with F1-F3 fibrosis ARMOR Suspended*

PPAR, peroxisome proliferator-activated receptor; NASH, nonalcoholic steatohepatitis; LC, liver cirrhosis.

* Starting the double-blind part of phase 3 trial is delayed due to the formulation of Aramchol Meglumine.

Abbreviations

NAFLD
nonalcoholic fatty liver disease
NASH
nonalcoholic steatohepatitis
GLP-1
glucagon-like peptide 1
RCTs
randomized controlled trials
PPAR
peroxisome proliferator-activated receptor
SAF
steatosis
THR-β
thyroid hormone receptor beta
MRI
magnetic resonance imaging
ASK1
apoptosis-signal regulating kinase 1
MAPK
mitogen-activated protein kinase

REFERENCES

1. Younossi Z, Tacke F, Arrese M, Chander Sharma B, Mostafa I, et al. Global perspectives on nonalcoholic fatty liver disease and nonalcoholic steatohepatitis. Hepatology 2019;69:2672-2682.
crossref pmid pdf
2. Kang SH, Lee HW, Yoo JJ, Cho Y, Kim SU, Lee TH, et al.; Korean Association for the Study of the Liver (KASL). KASL clinical practice guidelines: Management of nonalcoholic fatty liver disease. Clin Mol Hepatol 2021;27:363-401.
crossref pmid pmc pdf
3. Dulai PS, Singh S, Patel J, Soni M, Prokop LJ, Younossi Z, et al. Increased risk of mortality by fibrosis stage in nonalcoholic fatty liver disease: systematic review and meta-analysis. Hepatology 2017;65:1557-1565.
crossref pmid pdf
4. 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.
crossref pmid
5. Noureddin M, Vipani A, Bresee C, Todo T, Kim IK, Alkhouri N, et al. NASH leading cause of liver transplant in women: updated analysis of indications for liver transplant and ethnic and gender variances. Am J Gastroenterol 2018;113:1649-1659.
crossref pmid pmc pdf
6. Anstee QM, Reeves HL, Kotsiliti E, Govaere O, Heikenwalder M. From NASH to HCC: current concepts and future challenges. Nat Rev Gastroenterol Hepatol 2019;16:411-428.
crossref pmid pdf
7. Chalasani N, Younossi Z, Lavine JE, Charlton M, Cusi K, Rinella M, et al. The diagnosis and management of nonalcoholic fatty liver disease: practice guidance from the American Association for the Study of Liver Diseases. Hepatology 2018;67:328-357.
crossref pmid pdf
8. Vilar-Gomez E, Martinez-Perez Y, Calzadilla-Bertot L, TorresGonzalez A, Gra-Oramas B, Gonzalez-Fabian L, et al. Weight loss through lifestyle modification significantly reduces features of nonalcoholic steatohepatitis. Gastroenterology 2015;149:367-378.e5 quiz e14-5.
crossref pmid
9. Hydes TJ, Ravi S, Loomba R, E Gray M. Evidence-based clinical advice for nutrition and dietary weight loss strategies for the management of NAFLD and NASH. Clin Mol Hepatol 2020;26:383-400.
crossref pmid pmc pdf
10. Wong VW, Wong GL, Chan RS, Shu SS, Cheung BH, Li LS, et al. Beneficial effects of lifestyle intervention in non-obese patients with non-alcoholic fatty liver disease. J Hepatol 2018;69:1349-1356.
crossref pmid
11. Franz MJ, VanWormer JJ, Crain AL, Boucher JL, Histon T, Caplan W, et al. Weight-loss outcomes: a systematic review and metaanalysis of weight-loss clinical trials with a minimum 1-year follow-up. J Am Diet Assoc 2007;107:1755-1767.
crossref pmid
12. Sanyal AJ, Chalasani N, Kowdley KV, McCullough A, Diehl AM, Bass NM, et al.; NASH CRN. Pioglitazone, vitamin E, or placebo for nonalcoholic steatohepatitis. N Engl J Med 2010;362:1675-1685.
crossref pmid pmc
13. Patel Chavez C, Cusi K, Kadiyala S. The emerging role of glucagon-like peptide-1 receptor agonists for the management of NAFLD. J Clin Endocrinol Metab 2022;107:29-38.
crossref pmid pdf
14. Arroyave-Ospina JC, Wu Z, Geng Y, Moshage H. Role of oxidative stress in the pathogenesis of non-alcoholic fatty liver disease: implications for prevention and therapy. Antioxidants (Basel) 2021;10:174.
crossref pmid pmc
15. Jiang Q. Natural forms of vitamin E: metabolism, antioxidant, and anti-inflammatory activities and their role in disease prevention and therapy. Free Radic Biol Med 2014;72:76-90.
crossref pmid pmc
16. Bril F, Biernacki DM, Kalavalapalli S, Lomonaco R, Subbarayan SK, Lai J, et al. Role of vitamin E for nonalcoholic steatohepatitis in patients with type 2 diabetes: a randomized controlled trial. Diabetes Care 2019;42:1481-1488.
crossref pmid pdf
17. Cusi K, Isaacs S, Barb D, Basu R, Caprio S, Garvey WT, et al. American association of clinical endocrinology clinical practice guideline for the diagnosis and management of nonalcoholic fatty liver disease in primary care and endocrinology clinical settings: co-sponsored by the American Association for the Study of Liver Diseases (AASLD). Endocr Pract 2022;28:528-562.
pmid
18. 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.
crossref pmid pmc
19. 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.
crossref pmid
20. 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.
crossref pmid
21. Cusi K, Orsak B, Bril F, Lomonaco R, Hecht J, Ortiz-Lopez C, et al. Long-term pioglitazone treatment for patients with nonalcoholic steatohepatitis and prediabetes or type 2 diabetes mellitus: a randomized trial. Ann Intern Med 2016;165:305-315.
crossref pmid
22. Musso G, Cassader M, Paschetta E, Gambino R. Thiazolidinediones and Advanced liver fibrosis in nonalcoholic steatohepatitis: a meta-analysis. JAMA Intern Med 2017;177:633-640 Erratum in: JAMA Intern Med 2017;177:747.
crossref pmid pmc
23. Kim KS, Lee BW. Beneficial effect of anti-diabetic drugs for nonalcoholic fatty liver disease. Clin Mol Hepatol 2020;26:430-443.
crossref pmid pmc pdf
24. Newsome PN, Buchholtz K, Cusi K, Linder M, Okanoue T, Ratziu V, et al.; NN9931-4296 Investigators. A placebo-controlled trial of subcutaneous semaglutide in nonalcoholic steatohepatitis. N Engl J Med 2021;384:1113-1124.
crossref pmid
25. Day JW, Ottaway N, Patterson JT, Gelfanov V, Smiley D, Gidda J, et al. A new glucagon and GLP-1 co-agonist eliminates obesity in rodents. Nat Chem Biol 2009;5:749-757.
crossref pmid pdf
26. Robertson D, Challis B, Daniels JS, Sarv J, Sanchez J, Schumi J, et al. PROXYMO demonstrates safety and efficacy of cotadutide, a novel incretin co-agonist in biopsy-proven non-cirrhotic NASH with fibrosis. Oral abstract. Hepatology 2021;74:1383A.
crossref pmid pdf
27. Modica S, Gadaleta RM, Moschetta A. Deciphering the nuclear bile acid receptor FXR paradigm. Nucl Recept Signal 2010;8:e005.
crossref pmid pmc pdf
28. Claudel T, Staels B, Kuipers F. The Farnesoid X receptor: a molecular link between bile acid and lipid and glucose metabolism. Arterioscler Thromb Vasc Biol 2005;25:2020-2030.
pmid
29. Fiorucci S, Antonelli E, Rizzo G, Renga B, Mencarelli A, Riccardi L, et al. The nuclear receptor SHP mediates inhibition of hepatic stellate cells by FXR and protects against liver fibrosis. Gastroenterology 2004;127:1497-1512.
crossref pmid
30. Wang YD, Chen WD, Wang M, Yu D, Forman BM, Huang W. Farnesoid X receptor antagonizes nuclear factor kappaB in hepatic inflammatory response. Hepatology 2008;48:1632-1643.
crossref pmid
31. Younossi ZM, Ratziu V, Loomba R, Rinella M, Anstee QM, Goodman Z, et al.; REGENERATE Study Investigators. Obeticholic acid for the treatment of non-alcoholic steatohepatitis: interim analysis from a multicentre, randomised, placebo-controlled phase 3 trial. Lancet 2020;394:2184-2196 Erratum in: Lancet 2020;396:312. Erratum in: Lancet 2021;397:2336.

32. Intercept Pharmaceuticals. Intercept pharmaceuticals announces REVERSE phase 3 study of obeticholic acid (OCA) in compensated cirrhosis due to NASH did not meet its primary endpoint. Intercept web site, <https://ir.interceptpharma.com/newsreleases/news-release-details/intercept-pharmaceuticalsannounces-reverse-phase-3-study>. Press release; 30 Sep 2006. Accessed 1 Dec 2022.

33. Francque S, Szabo G, Abdelmalek MF, Byrne CD, Cusi K, Dufour JF, et al. Nonalcoholic steatohepatitis: the role of peroxisome proliferator-activated receptors. Nat Rev Gastroenterol Hepatol 2021;18:24-39.
crossref pmid pdf
34. Wettstein G, Luccarini JM, Poekes L, Faye P, Kupkowski F, Adarbes V, et al. The new-generation pan-peroxisome proliferator-activated receptor agonist IVA337 protects the liver from metabolic disorders and fibrosis. Hepatol Commun 2017;1:524-537.
crossref pmid pmc pdf
35. Lefere S, Puengel T, Hundertmark J, Penners C, Frank AK, Guillot A, et al. Differential effects of selective- and pan-PPAR agonists on experimental steatohepatitis and hepatic macrophages. J Hepatol 2020;73:757-770.
crossref pmid
36. Francque SM, Bedossa P, Ratziu V, Anstee QM, Bugianesi E, Sanyal AJ, et al.; NATIVE Study Group. A randomized, controlled trial of the pan-PPAR agonist lanifibranor in NASH. N Engl J Med 2021;385:1547-1558.
crossref pmid
37. National Library of Medicine. Phase 3 study to evaluate the efficacy and safety of elafibranor versus placebo in patients with nonalcoholic steatohepatitis (NASH) (RESOLVE-IT). National Library of Medicine web site, <https://clinicaltrials.gov/ct2/show/NCT02704403>. Accessed 1 Dec 2022.

38. Reinehr T. Obesity and thyroid function. Mol Cell Endocrinol 2010;316:165-171.
crossref pmid
39. Sinha RA, Bruinstroop E, Singh BK, Yen PM. Nonalcoholic fatty liver disease and hypercholesterolemia: roles of thyroid hormones, metabolites, and agonists. Thyroid 2019;29:1173-1191.
crossref pmid
40. Harrison SA, Bashir MR, Guy CD, Zhou R, Moylan CA, Frias JP, et al. Resmetirom (MGL-3196) for the treatment of non-alcoholic steatohepatitis: a multicentre, randomised, double-blind, placebo-controlled, phase 2 trial. Lancet 2019;394:2012-2024.
crossref pmid
41. Harrison S, Taub R, Neff G, Moussa S, Alkhouri N, Bashir MJJoH. Primary data analyses of MAESTRO-NAFLD-1 a 52 week doubleblind placebo-controlled phase 3 clinical trial of resmetirom in patients with NAFLD. J Hepatol 2022;77:S14.
crossref
42. Ichijo H, Nishida E, Irie K, ten Dijke P, Saitoh M, Moriguchi T, et al. Induction of apoptosis by ASK1, a mammalian MAPKKK that activates SAPK/JNK and p38 signaling pathways. Science 1997;275:90-94.
crossref pmid
43. Odagiri N, Matsubara T, Sato-Matsubara M, Fujii H, Enomoto M, Kawada N. Anti-fibrotic treatments for chronic liver diseases: the present and the future. Clin Mol Hepatol 2021;27:413-424.
crossref pmid pdf
44. Loomba R, Lawitz E, Mantry PS, Jayakumar S, Caldwell SH, Arnold H, et al.; GS-US-384-1497 Investigators. The ASK1 inhibitor selonsertib in patients with nonalcoholic steatohepatitis: a randomized, phase 2 trial. Hepatology 2018;67:549-559 Erratum in: Hepatology 2018;67:2063.
crossref pmid pdf
45. Harrison SA, Wong VW, Okanoue T, Bzowej N, Vuppalanchi R, Younes Z, et al.; STELLAR-3 and STELLAR-4 Investigators. Selonsertib for patients with bridging fibrosis or compensated cirrhosis due to NASH: Results from randomized phase III STELLAR trials. J Hepatol 2020;73:26-39.
crossref pmid
46. Loomba R, Noureddin M, Kowdley KV, Kohli A, Sheikh A, Neff G, et al.; ATLAS Investigators. Combination therapies including cilofexor and firsocostat for bridging fibrosis and cirrhosis attributable to NASH. Hepatology 2021;73:625-643.
crossref pmid pdf
47. Ajmera VH, Cachay E, Ramers C, Vodkin I, Bassirian S, Singh S, et al. MRI assessment of treatment response in HIV-associated NAFLD: a randomized trial of a Stearoyl-Coenzyme-A-Desaturase-1 inhibitor (ARRIVE trial). Hepatology 2019;70:1531-1545.
crossref pmid pdf
48. Friedman SL, Ratziu V, Harrison SA, Abdelmalek MF, Aithal GP, Caballeria J, et al. A randomized, placebo-controlled trial of cenicriviroc for treatment of nonalcoholic steatohepatitis with fibrosis. Hepatology 2018;67:1754-1767.
crossref pmid pdf
49. Attia SL, Softic S, Mouzaki M. Evolving role for pharmacotherapy in NAFLD/NASH. Clin Transl Sci 2021;14:11-19.
crossref pmid pdf

Editorial Office
The Korean Association for the Study of the Liver
Room A1210, 53 Mapo-daero(MapoTrapalace, Dowha-dong), Mapo-gu, Seoul, 04158, Korea
TEL: +82-2-703-0051   FAX: +82-2-703-0071    E-mail: kasl@kams.or.kr
Copyright © The Korean Association for the Study of the Liver.         
COUNTER
TODAY : 1485
TOTAL : 1735380
Close layer