201
|
Horn P, Newsome PN. Emerging therapeutic targets for NASH: key innovations at the preclinical level. Expert Opin Ther Targets 2020; 24:175-186. [PMID: 32053033 DOI: 10.1080/14728222.2020.1728742] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Introduction: nonalcoholic steatohepatitis (NASH) is a globally emerging health problem, mainly caused by increasing trends in the prevalence of obesity and metabolic syndrome. Patients with NASH are mainly affected by cardiovascular risk and extrahepatic cancer, but a significant proportion of patients will develop advanced liver disease, eventually resulting in liver failure or hepatocellular carcinoma. Recent research has yielded a better understanding of the underlying mechanisms and potential targetability for drug development.Areas covered: This review focuses on the role of fructose metabolism, de novo lipogenesis (DNL), endoplasmic reticulum (ER) stress, NLRP3 inflammasome, bone morphogenetic protein (BMP) signaling and platelets in the pathophysiology of NASH. We discuss the suitability of these substrates for targeting liver disease as well as cardiovascular health in patients with NASH. A non-systematic literature search was performed on PubMed and ClinicalTrials.gov.Expert opinion: Targeting fructose metabolism, DNL, ER stress, NLRP3 inflammasome, BMP signaling and platelets are promising therapeutic strategies, warranting further preclinical and clinical investigation. The discussed approaches might not only benefit liver-related outcomes but improve cardiovascular disease as well. Amidst the euphoria of advances in drug development for NASH, parallel endeavors need to address the underlying causes of obesity and metabolic syndrome to prevent NASH.
Collapse
Affiliation(s)
- Paul Horn
- National Institute for Health Research Birmingham Biomedical Research Centre at University Hospitals Birmingham NHS Foundation Trust and the University of Birmingham, Birmingham, UK.,Centre for Liver and Gastrointestinal Research, Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham, UK.,Liver Unit, University Hospitals Birmingham NHS Foundation Trust, Birmingham, UK
| | - Phlip N Newsome
- National Institute for Health Research Birmingham Biomedical Research Centre at University Hospitals Birmingham NHS Foundation Trust and the University of Birmingham, Birmingham, UK.,Centre for Liver and Gastrointestinal Research, Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham, UK.,Liver Unit, University Hospitals Birmingham NHS Foundation Trust, Birmingham, UK
| |
Collapse
|
202
|
Recent development in acetyl-CoA carboxylase inhibitors and their potential as novel drugs. Future Med Chem 2020; 12:533-561. [PMID: 32048880 DOI: 10.4155/fmc-2019-0312] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
Acetyl-CoA carboxylase (ACC), a critical enzyme in the regulation of fatty acid synthesis and metabolism, has emerged as an attractive target for a plethora of emerging diseases, such as diabetes mellitus, nonalcoholic fatty liver disease, cancer, bacterial infections and so on. With decades of efforts in medicinal chemistry, significant progress has been made toward the design and discovery of a considerable number of inhibitors of this enzyme. In this review, we not only clarify the role of ACC in emerging diseases, but also summarize recent developments of potent ACC inhibitors and discuss their molecular mechanisms of action and potentials as novel drugs as well as future perspectives toward the design and discovery of novel ACC inhibitors.
Collapse
|
203
|
Romero FA, Jones CT, Xu Y, Fenaux M, Halcomb RL. The Race to Bash NASH: Emerging Targets and Drug Development in a Complex Liver Disease. J Med Chem 2020; 63:5031-5073. [PMID: 31930920 DOI: 10.1021/acs.jmedchem.9b01701] [Citation(s) in RCA: 63] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Nonalcoholic steatohepatitis (NASH) is a severe form of nonalcoholic fatty liver disease (NAFLD) characterized by liver steatosis, inflammation, and hepatocellular damage. NASH is a serious condition that can progress to cirrhosis, liver failure, and hepatocellular carcinoma. The association of NASH with obesity, type 2 diabetes mellitus, and dyslipidemia has led to an emerging picture of NASH as the liver manifestation of metabolic syndrome. Although diet and exercise can dramatically improve NASH outcomes, significant lifestyle changes can be challenging to sustain. Pharmaceutical therapies could be an important addition to care, but currently none are approved for NASH. Here, we review the most promising targets for NASH treatment, along with the most advanced therapeutics in development. These include targets involved in metabolism (e.g., sugar, lipid, and cholesterol metabolism), inflammation, and fibrosis. Ultimately, combination therapies addressing multiple aspects of NASH pathogenesis are expected to provide benefit for patients.
Collapse
Affiliation(s)
- F Anthony Romero
- Terns Pharmaceuticals, 1065 E. Hillsdale Blvd., Suite 100, Foster City, California 94404, United States
| | - Christopher T Jones
- Terns Pharmaceuticals, 1065 E. Hillsdale Blvd., Suite 100, Foster City, California 94404, United States
| | - Yingzi Xu
- Terns Pharmaceuticals, 1065 E. Hillsdale Blvd., Suite 100, Foster City, California 94404, United States
| | - Martijn Fenaux
- Terns Pharmaceuticals, 1065 E. Hillsdale Blvd., Suite 100, Foster City, California 94404, United States
| | - Randall L Halcomb
- Terns Pharmaceuticals, 1065 E. Hillsdale Blvd., Suite 100, Foster City, California 94404, United States
| |
Collapse
|
204
|
Matsumoto M, Yashiro H, Ogino H, Aoyama K, Nambu T, Nakamura S, Nishida M, Wang X, Erion DM, Kaneko M. Acetyl-CoA carboxylase 1 and 2 inhibition ameliorates steatosis and hepatic fibrosis in a MC4R knockout murine model of nonalcoholic steatohepatitis. PLoS One 2020; 15:e0228212. [PMID: 31990961 PMCID: PMC6986730 DOI: 10.1371/journal.pone.0228212] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2019] [Accepted: 01/10/2020] [Indexed: 12/16/2022] Open
Abstract
Acetyl-CoA carboxylase (ACC) catalyzes the rate-limiting step in de novo lipogenesis, which is increased in the livers of patients with nonalcoholic steatohepatitis. GS-0976 (firsocostat), an inhibitor of isoforms ACC1 and ACC2, reduced hepatic steatosis and serum fibrosis biomarkers such as tissue inhibitor of metalloproteinase 1 in patients with nonalcoholic steatohepatitis in a randomized controlled trial, although the impact of this improvement on fibrosis has not fully been evaluated in preclinical models. Here, we used Western diet-fed melanocortin 4 receptor-deficient mice that have similar phenotypes to nonalcoholic steatohepatitis patients including progressively developed hepatic steatosis as well as fibrosis. We evaluated the effects of ACC1/2 inhibition on hepatic fibrosis. After the confirmation of significant hepatic fibrosis with a 13-week pre-feeding, GS-0976 (4 and 16 mg/kg/day) treatment for 9 weeks lowered malonyl-CoA and triglyceride content in the liver and improved steatosis, histologically. Furthermore, GS-0976 reduced the histological area of hepatic fibrosis, hydroxyproline content, mRNA expression level of type I collagen in the liver, and plasma tissue metalloproteinase inhibitor 1, suggesting an improvement of hepatic fibrosis. The treatment with GS-0976 was also accompanied by reductions of plasma ALT and AST levels. These data demonstrate that improvement of hepatic lipid metabolism by ACC1/2 inhibition could be a new option to suppress fibrosis progression as well as to improve hepatic steatosis in nonalcoholic steatohepatitis.
Collapse
Affiliation(s)
- Mitsuharu Matsumoto
- Department of Integrated Biology, Axcelead Drug Discovery Partners, Inc., Fujisawa, Kanagawa, Japan
- * E-mail:
| | - Hiroaki Yashiro
- Gastroenterology Drug Discovery Unit, Takeda Pharmaceutical Company Limited, Cambridge, Massachusetts, United States of America
| | - Hitomi Ogino
- Department of Integrated Biology, Axcelead Drug Discovery Partners, Inc., Fujisawa, Kanagawa, Japan
| | - Kazunobu Aoyama
- Department of Drug Disposition & Analysis, Axcelead Drug Discovery Partners, Inc., Fujisawa, Kanagawa, Japan
| | - Tadahiro Nambu
- Department of Nonclinical Safety Research, Axcelead Drug Discovery Partners, Inc., Fujisawa, Kanagawa, Japan
| | - Sayuri Nakamura
- Department of Nonclinical Safety Research, Axcelead Drug Discovery Partners, Inc., Fujisawa, Kanagawa, Japan
| | - Mayumi Nishida
- Department of Integrated Biology, Axcelead Drug Discovery Partners, Inc., Fujisawa, Kanagawa, Japan
| | - Xiaolun Wang
- Gastroenterology Drug Discovery Unit, Takeda Pharmaceutical Company Limited, Cambridge, Massachusetts, United States of America
| | - Derek M. Erion
- Gastroenterology Drug Discovery Unit, Takeda Pharmaceutical Company Limited, Cambridge, Massachusetts, United States of America
| | - Manami Kaneko
- Department of Integrated Biology, Axcelead Drug Discovery Partners, Inc., Fujisawa, Kanagawa, Japan
| |
Collapse
|
205
|
Marjot T, Moolla A, Cobbold JF, Hodson L, Tomlinson JW. Nonalcoholic Fatty Liver Disease in Adults: Current Concepts in Etiology, Outcomes, and Management. Endocr Rev 2020; 41:5601173. [PMID: 31629366 DOI: 10.1210/endrev/bnz009] [Citation(s) in RCA: 123] [Impact Index Per Article: 30.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/21/2019] [Accepted: 10/14/2019] [Indexed: 02/06/2023]
Abstract
Nonalcoholic fatty liver disease (NAFLD) is a spectrum of disease, extending from simple steatosis to inflammation and fibrosis with a significant risk for the development of cirrhosis. It is highly prevalent and is associated with significant adverse outcomes both through liver-specific morbidity and mortality but, perhaps more important, through adverse cardiovascular and metabolic outcomes. It is closely associated with type 2 diabetes and obesity, and both of these conditions drive progressive disease toward the more advanced stages. The mechanisms that govern hepatic lipid accumulation and the predisposition to inflammation and fibrosis are still not fully understood but reflect a complex interplay between metabolic target tissues including adipose and skeletal muscle, and immune and inflammatory cells. The ability to make an accurate assessment of disease stage (that relates to clinical outcome) can also be challenging. While liver biopsy is still regarded as the gold-standard investigative tool, there is an extensive literature on the search for novel noninvasive biomarkers and imaging modalities that aim to accurately reflect the stage of underlying disease. Finally, although no therapies are currently licensed for the treatment of NAFLD, there are interventions that appear to have proven efficacy in randomized controlled trials as well as an extensive emerging therapeutic landscape of new agents that target many of the fundamental pathophysiological processes that drive NAFLD. It is highly likely that over the next few years, new treatments with a specific license for the treatment of NAFLD will become available.
Collapse
Affiliation(s)
- Thomas Marjot
- Translational Gastroenterology Unit, NIHR Oxford Biomedical Research Centre, University of Oxford, John Radcliffe Hospital, Oxford, UK.,Oxford Centre for Diabetes, Endocrinology and Metabolism, NIHR Oxford Biomedical Research Centre, University of Oxford, Churchill Hospital, Oxford, UK
| | - Ahmad Moolla
- Oxford Centre for Diabetes, Endocrinology and Metabolism, NIHR Oxford Biomedical Research Centre, University of Oxford, Churchill Hospital, Oxford, UK
| | - Jeremy F Cobbold
- Translational Gastroenterology Unit, NIHR Oxford Biomedical Research Centre, University of Oxford, John Radcliffe Hospital, Oxford, UK
| | - Leanne Hodson
- Oxford Centre for Diabetes, Endocrinology and Metabolism, NIHR Oxford Biomedical Research Centre, University of Oxford, Churchill Hospital, Oxford, UK
| | - Jeremy W Tomlinson
- Oxford Centre for Diabetes, Endocrinology and Metabolism, NIHR Oxford Biomedical Research Centre, University of Oxford, Churchill Hospital, Oxford, UK
| |
Collapse
|
206
|
Wong VWS, Adams LA. Fibroblast growth factor 21 for non-alcoholic steatohepatitis. Lancet 2019; 392:2658-2660. [PMID: 30554786 DOI: 10.1016/s0140-6736(18)32165-2] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/03/2018] [Accepted: 08/30/2018] [Indexed: 12/11/2022]
Affiliation(s)
- Vincent W S Wong
- Department of Medicine and Therapeutics 9/F, Clinical Sciences Building, Chinese University of Hong Kong, Prince of Wales Hospital, Shatin, Hong Kong, China.
| | - Leon A Adams
- University of Western Australia, Perth, WA, Australia
| |
Collapse
|
207
|
Lu Y, Su X, Zhao M, Zhang Q, Liu C, Lai Q, Wu S, Fang A, Yang J, Chen X, Yao Y. Comparative RNA-sequencing profiled the differential gene expression of liver in response to acetyl-CoA carboxylase inhibitor GS-0976 in a mouse model of NASH. PeerJ 2019; 7:e8115. [PMID: 31879571 PMCID: PMC6927352 DOI: 10.7717/peerj.8115] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2019] [Accepted: 10/28/2019] [Indexed: 02/05/2023] Open
Abstract
Background Non-alcoholic steatohepatitis (NASH) is a progressive liver disease characterized by hepatic steatosis, lobular inflammation and fibrosis. Acetyl-CoA carboxylase (ACC) isoform 1 and 2 involved in de novo lipogenesis (DNL) and fatty acid oxidation have been identified as a therapeutic target in NASH. GS-0976, the inhibitor of ACC1 and ACC2, has achieved favorable therapeutic effects in clinical trials with NASH. The purpose of this study was to explore the transcriptional alterations regulated by GS-0976 in NASH. Methods C57BL/6 mice were fed on a choline-deficient, L-amino acid-defined, high-fat diet (CDAHFD) or normal diet for 12 weeks. Mice were treated with or without GS-0976 (3 mg/kg per day) in the last 8 weeks. Oil Red O, Haematoxylin-eosin (H & E), and Sirius Red were used to evaluate hepatic steatosis, inflammation and fibrosis. The comparative RNA-sequencing was conducted to analyse the hepatic gene expression profiles in mice. Reverse transcription–polymerase chain reaction analysis was performed to validate the differential expression of representative genes. Results GS-0976 attenuated the steatosis, inflammation, and fibrosis of NASH in CDAHFD mouse model. High-throughput sequencing and differential gene expression analysis showed that there were 516 up-regulated genes and 525 down-regulated genes after GS-0976 treatment. Genes involved in the metabolic process, extracellular matrix formation, immune response, and angiogenesis were significantly enriched. The “Metabolic pathways” and “ECM-receptor interaction” pathways were the most significantly enriched KEGG pathways in the up-regulated and down-regulated differentially expressed genes (DEGs), respectively. Conclusions Transcriptome analysis showed that GS-0976 could regulate the expression of genes related to metabolism, inflammation and fibrosis in NASH. The global transcriptomic changes in gene expression promote the further understanding for the inhibition mechanisms of GS-0976 in NASH.
Collapse
Affiliation(s)
- Ying Lu
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital and Healthy Food Evaluation Research Center, Sichuan University, Chengdu, China.,West China School of Public Health and West China Fourth Hospital, Healthy Food Evaluation Research Center, Sichuan University, Chengdu, China
| | - Xiaolan Su
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital and Healthy Food Evaluation Research Center, Sichuan University, Chengdu, China.,West China School of Public Health and West China Fourth Hospital, Healthy Food Evaluation Research Center, Sichuan University, Chengdu, China
| | - Manyu Zhao
- West China School of Public Health and West China Fourth Hospital, Healthy Food Evaluation Research Center, Sichuan University, Chengdu, China
| | - Qianru Zhang
- West China School of Public Health and West China Fourth Hospital, Healthy Food Evaluation Research Center, Sichuan University, Chengdu, China.,Guangdong Zhongsheng Pharmaceutical Co., Ltd., Dongguan, China
| | - Chuang Liu
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital and Healthy Food Evaluation Research Center, Sichuan University, Chengdu, China.,West China School of Public Health and West China Fourth Hospital, Healthy Food Evaluation Research Center, Sichuan University, Chengdu, China
| | - Qinhuai Lai
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital and Healthy Food Evaluation Research Center, Sichuan University, Chengdu, China.,West China School of Public Health and West China Fourth Hospital, Healthy Food Evaluation Research Center, Sichuan University, Chengdu, China
| | - Sijia Wu
- West China School of Public Health and West China Fourth Hospital, Healthy Food Evaluation Research Center, Sichuan University, Chengdu, China
| | - Aiping Fang
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital and Healthy Food Evaluation Research Center, Sichuan University, Chengdu, China.,West China School of Public Health and West China Fourth Hospital, Healthy Food Evaluation Research Center, Sichuan University, Chengdu, China
| | - Jinliang Yang
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital and Healthy Food Evaluation Research Center, Sichuan University, Chengdu, China.,West China School of Public Health and West China Fourth Hospital, Healthy Food Evaluation Research Center, Sichuan University, Chengdu, China.,Guangdong Zhongsheng Pharmaceutical Co., Ltd., Dongguan, China
| | - Xiaoxin Chen
- Guangdong Zhongsheng Pharmaceutical Co., Ltd., Dongguan, China
| | - Yuqin Yao
- West China School of Public Health and West China Fourth Hospital, Healthy Food Evaluation Research Center, Sichuan University, Chengdu, China
| |
Collapse
|
208
|
Qureshi K, Neuschwander-Tetri BA. The molecular basis for current targets of NASH therapies. Expert Opin Investig Drugs 2019; 29:151-161. [PMID: 31847612 DOI: 10.1080/13543784.2020.1703949] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Introduction: Nonalcoholic steatohepatitis (NASH) is a leading cause of liver disease in children and adults, a major contributor to health-care expenditures, and now a leading reason for liver transplantation. Adopting lifestyle modifications with regular exercise and a focus on healthy eating habits is the primary recommendation. However, patients are often unable to achieve and sustain such changes for a variety of social, physical, psychological and genetic reasons. Thus, treatments that can prevent and reverse NASH and its associated fibrosis are a major focus of current drug development.Areas covered: This review covers the current understanding of lipotoxic liver injury in the pathogenesis of NASH and how lifestyle modification and the spectrum of drugs currently in clinical trials address the many pathways leading to the phenotype of NASH.Expert opinion: Contrary to the frequently expressed nihilistic view of our understanding of NASH and disappointment with clinical trial results, much is known about the pathogenesis of NASH and there is much reason to be optimistic that effective therapies will be identified in the next 5-10 years. Achieving this will require continued refinement of clinical trial endpoints, continued engagement of trial sponsors and regulatory authorities, and continued participation of dedicated patients in clinical trials.
Collapse
Affiliation(s)
- Kamran Qureshi
- Division of Gastroenterology and Hepatology, Saint Louis University, St. Louis, MO, USA
| | | |
Collapse
|
209
|
Takada I, Makishima M. Peroxisome proliferator-activated receptor agonists and antagonists: a patent review (2014-present). Expert Opin Ther Pat 2019; 30:1-13. [PMID: 31825687 DOI: 10.1080/13543776.2020.1703952] [Citation(s) in RCA: 72] [Impact Index Per Article: 14.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Introduction: Peroxisome proliferator-activated receptors (PPARs), PPARα, PPARδ, and PPARγ, play an important role in the regulation of various physiological processes, specifically lipid and energy metabolism and immunity. PPARα agonists (fibrates) and PPARγ agonists (thiazolidinediones) are used for the treatment of hypertriglyceridemia and type 2 diabetes, respectively. PPARδ activation enhances mitochondrial and energy metabolism but PPARδ-acting drugs are not yet available. Many synthetic ligands for PPARs have been developed to expand their therapeutic applications.Areas covered: The authors searched recent patent activity regarding PPAR ligands. Novel PPARα agonists, PPARδ agonists, PPARγ agonists, PPARα/γ dual agonists, and PPARγ antagonists have been claimed for the treatment of metabolic disease and inflammatory disease. Methods for the combination of PPAR ligands with other drugs and expanded application of PPAR agonists for bone and neurological disease have been also claimed.Expert opinion: Novel PPAR ligands and the combination of PPAR ligands with other drugs have been claimed for the treatment of mitochondrial disease, inflammatory/autoimmune disease, neurological disease, and cancer in addition to metabolic diseases including dyslipidemia and type 2 diabetes. Selective therapeutic actions of PPAR ligands should be exploited to avoid adverse effects. More basic studies are needed to elucidate the molecular mechanisms of selective actions.
Collapse
Affiliation(s)
- Ichiro Takada
- Division of Biochemistry, Department of Biomedical Sciences, Nihon University School of Medicine, Tokyo, Japan
| | - Makoto Makishima
- Division of Biochemistry, Department of Biomedical Sciences, Nihon University School of Medicine, Tokyo, Japan
| |
Collapse
|
210
|
Reimer KC, Wree A, Roderburg C, Tacke F. New drugs for NAFLD: lessons from basic models to the clinic. Hepatol Int 2019; 14:8-23. [PMID: 31802390 DOI: 10.1007/s12072-019-10001-4] [Citation(s) in RCA: 52] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/05/2019] [Accepted: 11/01/2019] [Indexed: 12/14/2022]
Abstract
The term nonalcoholic fatty liver disease (NAFLD) comprises a spectrum of increasingly harmful conditions ranging from nonalcoholic fatty liver (NAFL) to nonalcoholic steatohepatitis (NASH) to liver fibrosis and end-stage cirrhosis. NAFLD is the currently most common form of chronic liver disease in both adults and children worldwide. As NAFLD evolves as a global pandemic alongside the still growing prevalence of metabolic syndrome, obesity, and diabetes, it is inevitable to develop effective counterstrategies. Over the last decades, great effort has been dedicated to the understanding of the pathogenesis of NAFLD. This includes the development of an array of models for NAFLD, ranging from advanced in vitro (primary cells, 3D cultures, biochip, spheroids, organoids) to in vivo rodent models (particularly in mice). Based on these approaches novel therapies have been proposed and subsequently evaluated for patients with advanced forms of NAFLD, in particular those with NASH and liver fibrosis or cirrhosis. In this review, we delineate the current understanding of disease pathophysiology and depict how novel therapeutic strategies aim to exploit these different mechanisms to ameliorate, treat, or stop progression of NASH. We also discuss obstacles and chances along the way from basic models to promising clinical treatment options.
Collapse
Affiliation(s)
- Katharina C Reimer
- Department of Medicine II, Nephrology/Rheumatology/Clinical Immunology, University Hospital RWTH Aachen, 52074, Aachen, Germany
| | - Alexander Wree
- Department of Hepatology and Gastroenterology, Charité University Medical Center, Augustenburger Platz 1, 13353, Berlin, Germany
| | - Christoph Roderburg
- Department of Hepatology and Gastroenterology, Charité University Medical Center, Augustenburger Platz 1, 13353, Berlin, Germany
| | - Frank Tacke
- Department of Hepatology and Gastroenterology, Charité University Medical Center, Augustenburger Platz 1, 13353, Berlin, Germany.
| |
Collapse
|
211
|
Treatments of nonalcoholic fatty liver disease in adults who have no other illness: A Review article. Arab J Gastroenterol 2019; 20:189-197. [DOI: 10.1016/j.ajg.2019.11.004] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/09/2019] [Accepted: 11/26/2019] [Indexed: 12/28/2022]
|
212
|
Chen Z, Yu Y, Cai J, Li H. Emerging Molecular Targets for Treatment of Nonalcoholic Fatty Liver Disease. Trends Endocrinol Metab 2019; 30:903-914. [PMID: 31597607 DOI: 10.1016/j.tem.2019.08.006] [Citation(s) in RCA: 78] [Impact Index Per Article: 15.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/21/2019] [Revised: 08/14/2019] [Accepted: 08/16/2019] [Indexed: 12/12/2022]
Abstract
In parallel with the obesity epidemic, nonalcoholic fatty liver disease (NAFLD) has emerged as the most common chronic liver disease worldwide. Disequilibrium of lipid metabolism and the subsequent metabolic-stress-induced inflammation are believed to be central in the pathogenesis of NAFLD. Of note, metabolic inflammation is primarily mediated by innate immune signaling, which is increasingly recognized as a driving force in NAFLD progression. Currently, a series of agents targeting one or more of these pathomechanisms have shown encouraging results in preclinical models and clinical trials. This review summarizes the emerging molecular targets involved in signaling in the lipid metabolism and innate immunity aspects of NAFLD, focusing on their mechanistic roles and translational potentials.
Collapse
Affiliation(s)
- Ze Chen
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan 430060, China; Institute of Model Animals of Wuhan University, Wuhan 430072, China
| | - Yao Yu
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan 430060, China; Institute of Model Animals of Wuhan University, Wuhan 430072, China
| | - Jingjing Cai
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan 430060, China; Institute of Model Animals of Wuhan University, Wuhan 430072, China; Department of Cardiology, The Third Xiangya Hospital, Central South University, Changsha 410013, China.
| | - Hongliang Li
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan 430060, China; Institute of Model Animals of Wuhan University, Wuhan 430072, China; Basic Medical School, Wuhan University, Wuhan 430071, China.
| |
Collapse
|
213
|
Hodson L, Gunn PJ. The regulation of hepatic fatty acid synthesis and partitioning: the effect of nutritional state. Nat Rev Endocrinol 2019; 15:689-700. [PMID: 31554932 DOI: 10.1038/s41574-019-0256-9] [Citation(s) in RCA: 147] [Impact Index Per Article: 29.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Nonalcoholic fatty liver disease (NAFLD) is an increasing global public health burden. NAFLD is strongly associated with type 2 diabetes mellitus, obesity and cardiovascular disease and begins with intrahepatic triacylglycerol accumulation. Under healthy conditions, the liver regulates lipid metabolism to meet systemic energy needs in the fed and fasted states. The processes of fatty acid uptake, fatty acid synthesis and the intracellular partitioning of fatty acids into storage, oxidation and secretion pathways are tightly regulated. When one or more of these processes becomes dysregulated, excess lipid accumulation can occur. Although genetic and environmental factors have been implicated in the development of NAFLD, it remains unclear why an imbalance in these pathways begins. The regulation of fatty acid partitioning occurs at several points, including during triacylglycerol synthesis, lipid droplet formation and lipolysis. These processes are influenced by enzyme function, intake of dietary fats and sugars and whole-body metabolism, and are further affected by the presence of obesity or insulin resistance. Insight into how the liver controls fatty acid metabolism in health and how these processes might be affected in disease would offer the potential for new therapeutic treatments for NAFLD to be developed.
Collapse
Affiliation(s)
- Leanne Hodson
- Oxford Centre for Diabetes, Endocrinology and Metabolism, Radcliffe Department of Medicine, University of Oxford, Headington, Oxford, UK.
- Oxford NIHR Biomedical Research Centre, Churchill Hospital, Headington, Oxford, UK.
| | - Pippa J Gunn
- Oxford Centre for Diabetes, Endocrinology and Metabolism, Radcliffe Department of Medicine, University of Oxford, Headington, Oxford, UK
- Oxford NIHR Biomedical Research Centre, Churchill Hospital, Headington, Oxford, UK
| |
Collapse
|
214
|
Harrison SA, Bashir MR, Guy CD, Zhou R, Moylan CA, Frias JP, Alkhouri N, Bansal MB, Baum S, Neuschwander-Tetri BA, Taub R, Moussa SE. 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. [PMID: 31727409 DOI: 10.1016/s0140-6736(19)32517-6] [Citation(s) in RCA: 400] [Impact Index Per Article: 80.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/12/2019] [Revised: 09/26/2019] [Accepted: 10/01/2019] [Indexed: 12/19/2022]
Abstract
BACKGROUND Non-alcoholic steatohepatitis (NASH) is characterised by hepatic steatosis, inflammation, hepatocellular injury, and progressive liver fibrosis. Resmetirom (MGL-3196) is a liver-directed, orally active, selective thyroid hormone receptor-β agonist designed to improve NASH by increasing hepatic fat metabolism and reducing lipotoxicity. We aimed to assess the safety and efficacy of resmetirom in patients with NASH. METHODS MGL-3196-05 was a 36-week randomised, double-blind, placebo-controlled study at 25 centres in the USA. Adults with biopsy confirmed NASH (fibrosis stages 1-3) and hepatic fat fraction of at least 10% at baseline when assessed by MRI-proton density fat fraction (MRI-PDFF) were eligible. Patients were randomly assigned 2:1 by a computer-based system to receive resmetirom 80 mg or matching placebo, orally once a day. Serial hepatic fat measurements were obtained at weeks 12 and 36, and a second liver biopsy was obtained at week 36. The primary endpoint was relative change in MRI-PDFF assessed hepatic fat compared with placebo at week 12 in patients who had both a baseline and week 12 MRI-PDFF. This trial is registered with ClinicalTrials.gov, number NCT02912260. FINDINGS 348 patients were screened and 84 were randomly assigned to resmetirom and 41 to placebo at 18 sites in the USA. Resmetirom-treated patients (n=78) showed a relative reduction of hepatic fat compared with placebo (n=38) at week 12 (-32·9% resmetirom vs -10·4% placebo; least squares mean difference -22·5%, 95% CI -32·9 to -12·2; p<0·0001) and week 36 (-37·3% resmetirom [n=74] vs -8·5 placebo [n=34]; -28·8%, -42·0 to -15·7; p<0·0001). Adverse events were mostly mild or moderate and were balanced between groups, except for a higher incidence of transient mild diarrhoea and nausea with resmetirom. INTERPRETATION Resmetirom treatment resulted in significant reduction in hepatic fat after 12 weeks and 36 weeks of treatment in patients with NASH. Further studies of resmetirom will allow assessment of safety and effectiveness of resmetirom in a larger number of patients with NASH with the possibility of documenting associations between histological effects and changes in non-invasive markers and imaging. FUNDING Madrigal Pharmaceuticals.
Collapse
Affiliation(s)
- Stephen A Harrison
- Pinnacle Clinical Research, San Antonio, TX, USA; Radcliffe Department of Medicine, University of Oxford, Oxford, UK.
| | - Mustafa R Bashir
- Department of Radiology, Center for Advanced Magnetic Resonance Development, Department of Pathology, and Division of Hepatology, Duke University Medical Center, Durham, NC, USA
| | - Cynthia D Guy
- Department of Radiology, Center for Advanced Magnetic Resonance Development, Department of Pathology, and Division of Hepatology, Duke University Medical Center, Durham, NC, USA
| | | | - Cynthia A Moylan
- Department of Radiology, Center for Advanced Magnetic Resonance Development, Department of Pathology, and Division of Hepatology, Duke University Medical Center, Durham, NC, USA
| | - Juan P Frias
- Department of Medicine, University of California, San Diego, CA, USA
| | - Naim Alkhouri
- Division of Gastroenterology, Department of Medicine, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
| | - Meena B Bansal
- Division of Hepatology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Seth Baum
- Department of Integrated Medicine, Florida Atlantic University, Miami, FL, USA
| | | | | | - Sam E Moussa
- Department of Medicine, University of Arizona College of Medicine, Tucson, AZ, USA
| |
Collapse
|
215
|
Ganguli S, DeLeeuw P, Satapathy SK. A Review Of Current And Upcoming Treatment Modalities In Non-Alcoholic Fatty Liver Disease And Non-Alcoholic Steatohepatitis. Hepat Med 2019; 11:159-178. [PMID: 31814783 PMCID: PMC6863115 DOI: 10.2147/hmer.s188991] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/17/2019] [Accepted: 10/03/2019] [Indexed: 12/15/2022] Open
Abstract
Non-alcoholic fatty liver disease (NAFLD) is the most common cause of chronic liver disease in the West. Non-alcoholic steatohepatitis (NASH) is the progressive form of NAFLD and can lead to cirrhosis, hepatocellular carcinoma, and is associated with increased cardiovascular risks. Multiple components and risk factors are thought to be involved in the pathogenesis of NAFLD and NASH. Optimal therapy has not yet been found, but many advances have been made with the discovery of potential therapeutic options. In this paper, we aim to provide a comprehensive review of approved, studied, and upcoming treatment options for NAFLD and NASH. Non-pharmacologic therapy (lifestyle modifications and bariatric surgery) and pharmacologic therapy are both reviewed. Pharmacologic therapy target components thought to be involved in the pathogenesis of this disease process including insulin resistance, oxidative stress, inflammation, lipid metabolism, and fibrosis are reviewed in this paper. Results of the emerging treatment targets in phase 2 and 3 clinical trials are also included.
Collapse
Affiliation(s)
- Surosree Ganguli
- Division of Internal Medicine, University of Louisville, Louisville, KY 40202, USA
| | - Peter DeLeeuw
- Division of Internal Medicine, University of Tennessee Health Science Center, Memphis, TN 38163, USA
| | - Sanjaya K Satapathy
- Division of Hepatology and Sandra Atlas Bass Center for Liver Diseases, Northwell Health, Manhasset, NY 11030, USA
| |
Collapse
|
216
|
Gapp B, Jourdain M, Bringer P, Kueng B, Weber D, Osmont A, Zurbruegg S, Knehr J, Falchetto R, Roma G, Dietrich W, Valdez R, Beckmann N, Nigsch F, Sanyal AJ, Ksiazek I. Farnesoid X Receptor Agonism, Acetyl-Coenzyme A Carboxylase Inhibition, and Back Translation of Clinically Observed Endpoints of De Novo Lipogenesis in a Murine NASH Model. Hepatol Commun 2019; 4:109-125. [PMID: 31909359 PMCID: PMC6939503 DOI: 10.1002/hep4.1443] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/24/2019] [Accepted: 10/03/2019] [Indexed: 12/16/2022] Open
Abstract
A promising approach for the treatment of nonalcoholic steatohepatitis (NASH) is the inhibition of enhanced hepatic de novo lipogenesis (DNL), which is the synthesis of fatty acids from nonlipid sources. This study assesses three approaches to DNL suppression in a newly developed dietary NASH mouse model: i) dietary intervention (switch from NASH‐inducing diet to normal diet); ii) inhibition of acetyl‐coenzyme A carboxylase (ACC), the enzyme catalyzing the rate‐limiting step in DNL; and iii) activation of farnesoid X receptor (FXR), a major transcriptional regulator of DNL. C57BL/6J mice on a high‐fat diet combined with ad libitum consumption of a fructose–sucrose solution developed several of the liver histologic features seen in human disease, including steatosis, inflammation, and fibrosis, accompanied by elevated fibrosis biomarkers and liver injury enzymes. Obesity and metabolic impairments were associated with increased intestinal permeability and progression to adenoma and hepatocellular carcinoma. All three approaches led to resolution of established NASH with fibrosis in mice; however, some differences were noted, e.g., with respect to the degree of hepatic steatosis attenuation. While ACC inhibition resulted in elevated blood triglycerides and peripheral obesity, FXR activation prevented peripheral obesity in NASH mice. Comparative transcriptome analysis underlined the translatability of the mouse model to human NASH and revealed novel mechanistic insights into differential regulation of lipid, inflammatory, and extracellular matrix pathways by FXR agonism and ACC inhibition. Conclusion: Novel insights are provided on back translation of clinically observed endpoints of DNL inhibition by targeting ACC or FXR, which are promising therapeutic options for the treatment of NASH, in a newly developed diet‐induced NASH mouse model.
Collapse
Affiliation(s)
- Berangere Gapp
- Disease Area X Novartis Institutes for BioMedical Research Basel Switzerland
| | - Marie Jourdain
- Disease Area X Novartis Institutes for BioMedical Research Basel Switzerland
| | - Pauline Bringer
- Disease Area X Novartis Institutes for BioMedical Research Basel Switzerland
| | - Benjamin Kueng
- Disease Area X Novartis Institutes for BioMedical Research Basel Switzerland
| | - Delphine Weber
- Disease Area X Novartis Institutes for BioMedical Research Basel Switzerland
| | - Arnaud Osmont
- Biotherapeutic and Analytical Technologies Novartis Institutes for BioMedical Research Basel Switzerland
| | - Stefan Zurbruegg
- Neurosciences Novartis Institutes for BioMedical Research Basel Switzerland
| | - Judith Knehr
- Chemical Biology and Therapeutics Novartis Institutes for BioMedical Research Basel Switzerland
| | - Rocco Falchetto
- Biotherapeutic and Analytical Technologies Novartis Institutes for BioMedical Research Basel Switzerland
| | - Guglielmo Roma
- Chemical Biology and Therapeutics Novartis Institutes for BioMedical Research Basel Switzerland
| | - William Dietrich
- Disease Area X Novartis Institutes for BioMedical Research Cambridge MA
| | - Reginald Valdez
- Disease Area X Novartis Institutes for BioMedical Research Cambridge MA
| | - Nicolau Beckmann
- Musculoskeletal Diseases Novartis Institutes for BioMedical Research Basel Switzerland
| | - Florian Nigsch
- Chemical Biology and Therapeutics Novartis Institutes for BioMedical Research Basel Switzerland
| | - Arun J Sanyal
- Department of Internal Medicine Division of Gastroenterology Virginia Commonwealth University Richmond VA
| | - Iwona Ksiazek
- Disease Area X Novartis Institutes for BioMedical Research Basel Switzerland
| |
Collapse
|
217
|
Pharmacological Therapy of Non-Alcoholic Fatty Liver Disease: What Drugs Are Available Now and Future Perspectives. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2019; 16:ijerph16224334. [PMID: 31703268 PMCID: PMC6888162 DOI: 10.3390/ijerph16224334] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/16/2019] [Revised: 11/01/2019] [Accepted: 11/03/2019] [Indexed: 12/25/2022]
Abstract
The non-alcoholic fatty liver disease (NAFLD) is rapidly becoming the most common cause of chronic liver disease as well as the first cause of liver transplantation. NAFLD is commonly associated with metabolic syndrome (MetS), and this is the most important reason why it is extremely difficult to treat this disease bearing in mind the enormous amount of interrelationships between the liver and other systems in maintaining the metabolic health. The treatment of NAFLD is a key point to prevent NASH progression to advanced fibrosis, to prevent cirrhosis and to prevent the development of its hepatic complications (such as liver decompensation and HCC) and even extrahepatic one. A part of the well-known healthy effect of diet and physical exercise in this setting it is important to design the correct pharmaceutical strategy in order to antagonize the progression of the disease. In this regard, the current review has the scope to give a panoramic view on the possible pharmacological treatment strategy in NAFLD patients.
Collapse
|
218
|
Ajmera VH, Cachay E, Ramers C, Vodkin I, Bassirian S, Singh S, Mangla N, Bettencourt R, Aldous JL, Park D, Lee D, Blanchard J, Mamidipalli A, Boehringer A, Aslam S, Leinhard OD, Richards L, Sirlin C, Loomba R. 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. [PMID: 31013363 PMCID: PMC7164416 DOI: 10.1002/hep.30674] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/04/2019] [Accepted: 04/01/2019] [Indexed: 12/12/2022]
Abstract
Aramchol, an oral stearoyl-coenzyme-A-desaturase-1 inhibitor, has been shown to reduce hepatic fat content in patients with primary nonalcoholic fatty liver disease (NAFLD); however, its effect in patients with human immunodeficiency virus (HIV)-associated NAFLD is unknown. The aramchol for HIV-associated NAFLD and lipodystrophy (ARRIVE) trial was a double-blind, randomized, investigator-initiated, placebo-controlled trial to test the efficacy of 12 weeks of treatment with aramchol versus placebo in HIV-associated NAFLD. Fifty patients with HIV-associated NAFLD, defined by magnetic resonance imaging (MRI)-proton density fat fraction (PDFF) ≥5%, were randomized to receive either aramchol 600 mg daily (n = 25) or placebo (n = 25) for 12 weeks. The primary endpoint was a change in hepatic fat as measured by MRI-PDFF in colocalized regions of interest. Secondary endpoints included changes in liver stiffness using magnetic resonance elastography (MRE) and vibration-controlled transient elastography (VCTE), and exploratory endpoints included changes in total-body fat and muscle depots on dual-energy X-ray absorptiometry (DXA), whole-body MRI, and cardiac MRI. The mean (± standard deviation) of age and body mass index were 48.2 ± 10.3 years and 30.7 ± 4.6 kg/m2 , respectively. There was no difference in the reduction in mean MRI-PDFF between the aramchol group at -1.3% (baseline MRI-PDFF 15.6% versus end-of-treatment MRI-PDFF 14.4%, P = 0.24) and the placebo group at -1.4% (baseline MRI-PDFF 13.3% versus end-of-treatment MRI-PDFF 11.9%, P = 0.26). There was no difference in the relative decline in mean MRI-PDFF between the aramchol and placebo groups (6.8% versus 1.1%, P = 0.68). There were no differences in MRE-derived and VCTE-derived liver stiffness and whole-body (fat and muscle) composition analysis by MRI or DXA. Compared to baseline, end-of-treatment aminotransferases were lower in the aramchol group but not in the placebo arm. There were no significant adverse events. Conclusion: Aramchol, over a 12-week period, did not reduce hepatic fat or change body fat and muscle composition by using MRI-based assessment in patients with HIV-associated NAFLD (clinicaltrials.gov ID:NCT02684591).
Collapse
Affiliation(s)
- Veeral H. Ajmera
- NAFLD Research Center, Department of Medicine, La Jolla, California,Division of Gastroenterology, Department of Medicine, La Jolla, California
| | - Edward Cachay
- Division of Infectious Diseases, Owen Clinic, University of California San Diego, San Diego, California
| | | | - Irine Vodkin
- Division of Gastroenterology, Department of Medicine, La Jolla, California
| | - Shirin Bassirian
- NAFLD Research Center, Department of Medicine, La Jolla, California
| | - Seema Singh
- NAFLD Research Center, Department of Medicine, La Jolla, California
| | - Neeraj Mangla
- NAFLD Research Center, Department of Medicine, La Jolla, California
| | | | | | | | - Daniel Lee
- Division of Infectious Diseases, Owen Clinic, University of California San Diego, San Diego, California
| | - Jennifer Blanchard
- Division of Infectious Diseases, Owen Clinic, University of California San Diego, San Diego, California
| | - Adrija Mamidipalli
- Liver Imaging Group, University of California, San Diego, La Jolla, California
| | - Andrew Boehringer
- Liver Imaging Group, University of California, San Diego, La Jolla, California
| | - Saima Aslam
- Division of Infectious Diseases and Global Public Health, University of California, San Diego, San Diego California
| | - Olof Dahlqvist Leinhard
- AMRA Medical AB, Linkoping Sweden,Department of Medical and Health Sciences, Linköping University, Linköping, Sweden
| | - Lisa Richards
- NAFLD Research Center, Department of Medicine, La Jolla, California
| | - Claude Sirlin
- Liver Imaging Group, University of California, San Diego, La Jolla, California
| | - Rohit Loomba
- NAFLD Research Center, Department of Medicine, La Jolla, California,Division of Gastroenterology, Department of Medicine, La Jolla, California,Division of Epidemiology, Department of Family and Preventive Medicine, University of California at San Diego, La Jolla, California
| |
Collapse
|
219
|
Lemoinne S, Friedman SL. New and emerging anti-fibrotic therapeutics entering or already in clinical trials in chronic liver diseases. Curr Opin Pharmacol 2019; 49:60-70. [PMID: 31590120 DOI: 10.1016/j.coph.2019.09.006] [Citation(s) in RCA: 49] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2019] [Accepted: 09/13/2019] [Indexed: 02/07/2023]
Abstract
Chronic liver diseases with different etiologies represent a major public health issue worldwide. Liver fibrosis is the common feature of almost all chronic liver diseases and remains a key determinant of clinical prognosis. Over the last two decades, basic science studies have uncovered molecular mechanisms underlying the pathophysiology of chronic liver diseases, leading to the recent development of new anti-fibrotic drugs. These new drugs target different steps in the pathophysiology of chronic liver injury: metabolism of glucose, lipids and bile acids, apoptosis, inflammation and fibrosis. Many targets are shared between non-alcoholic steatohepatitis (NASH) and cholestatic diseases, explaining why some drugs have been assessed concurrently in both conditions. This review reports the most recent clinical trials designed to treat liver fibrosis, with a special focus on NASH and cholestatic diseases.
Collapse
Affiliation(s)
- Sara Lemoinne
- Division of Liver Diseases, Icahn Medical School at Mount Sinai, New York, NY, USA
| | - Scott L Friedman
- Division of Liver Diseases, Icahn Medical School at Mount Sinai, New York, NY, USA.
| |
Collapse
|
220
|
Alkhouri N, Lawitz E, Noureddin M, DeFronzo R, Shulman GI. GS-0976 (Firsocostat): an investigational liver-directed acetyl-CoA carboxylase (ACC) inhibitor for the treatment of non-alcoholic steatohepatitis (NASH). Expert Opin Investig Drugs 2019; 29:135-141. [PMID: 31519114 DOI: 10.1080/13543784.2020.1668374] [Citation(s) in RCA: 46] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Introduction: De novo lipogenesis (DNL) plays a major role in fatty acid metabolism and contributes significantly to triglyceride accumulation within the hepatocytes in patients with nonalcoholic steatohepatitis (NASH). Acetyl-CoA carboxylase (ACC) converts acetyl-CoA to malonyl CoA and is a rate-controlling step in DNL. Furthermore, malonyl-CoA is an important regulator of hepatic mitochondrial fat oxidation through its ability to inhibit carnitine palmitoyltransferase I. Therefore, inhibiting ACC pharmacologically represents an attractive approach to treating NASH.Areas covered: This article summarizes preclinical and clinical data on the efficacy and safety of the liver-targeted ACC inhibitor GS-0976 (Firsocostat) for the treatment of NASH. In a phase 2 trial that included 126 patients with NASH and fibrosis, GS-0976 20 mg daily for 12 weeks showed significant relative reduction in liver fat by 29%; however, treatment was associated with an increase in plasma triglycerides with 16 patients having levels >500 mg/dL.Expert opinion: Preclinical and preliminary clinical data support the development of GS-0976 as treatment for NASH. ACC-induced hypertriglyceridemia can be mitigated by fish oil or fibrates, but the long-term cardiovascular effects require further investigations.
Collapse
Affiliation(s)
- Naim Alkhouri
- University of Texas (UT) Health San Antonio, San Antonio, TX, USA.,Texas Liver Institute, San Antonio, TX, USA
| | - Eric Lawitz
- University of Texas (UT) Health San Antonio, San Antonio, TX, USA.,Texas Liver Institute, San Antonio, TX, USA
| | | | - Ralph DeFronzo
- University of Texas (UT) Health San Antonio, San Antonio, TX, USA.,Texas Diabetes Institute (TDI), San Antonio, TX, USA
| | - Gerald I Shulman
- Departments of Internal Medicine and Cellular & Molecular Physiology, Yale School of Medicine, New Haven, CT, USA
| |
Collapse
|
221
|
Wong VWS, Singal AK. Emerging medical therapies for non-alcoholic fatty liver disease and for alcoholic hepatitis. Transl Gastroenterol Hepatol 2019; 4:53. [PMID: 31463412 PMCID: PMC6691078 DOI: 10.21037/tgh.2019.06.06] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/14/2019] [Accepted: 05/29/2019] [Indexed: 12/12/2022] Open
Abstract
Non-alcoholic fatty liver disease (NAFLD) and alcoholic liver disease (ALD) are currently the two most common liver diseases in the world. Alcoholic hepatitis (AH), a unique clinical syndrome among ALD patients has high short-term mortality. Apart from controlling the risk factor for individual respective disease, there are no Food and Drug Administration (FDA) approved medical therapies for these diseases. Over the last 5-10 years, the field has extensively grown with many new targets being studied in randomized clinical trials for these diseases, with many of these drugs being tested in both the conditions. In this chapter, we will describe the novel therapeutic agents and current status of ongoing clinical trials with these agents for the treatment of NAFLD and/or AH.
Collapse
Affiliation(s)
- Vincent Wai-Sun Wong
- Department of Medicine and Therapeutics, the Chinese University of Hong Kong, Hong Kong, China
- State Key Laboratory of Digestive Disease, the Chinese University of Hong Kong, Hong Kong, China
| | - Ashwani K. Singal
- Division of Gastroenterology and Hepatology, Avera Transplant Institute, University of South Dakota Sanford School of Medicine, Sioux Falls, SD, USA
| |
Collapse
|
222
|
Manka P, Zeller A, Syn WK. Fibrosis in Chronic Liver Disease: An Update on Diagnostic and Treatment Modalities. Drugs 2019; 79:903-927. [DOI: 10.1007/s40265-019-01126-9] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
|
223
|
Esler WP, Bence KK. Metabolic Targets in Nonalcoholic Fatty Liver Disease. Cell Mol Gastroenterol Hepatol 2019; 8:247-267. [PMID: 31004828 PMCID: PMC6698700 DOI: 10.1016/j.jcmgh.2019.04.007] [Citation(s) in RCA: 90] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/25/2019] [Revised: 04/09/2019] [Accepted: 04/11/2019] [Indexed: 12/18/2022]
Abstract
The prevalence and diagnosis of nonalcoholic fatty liver disease (NAFLD) is on the rise worldwide and currently has no FDA-approved pharmacotherapy. The increase in disease burden of NAFLD and a more severe form of this progressive liver disease, nonalcoholic steatohepatitis (NASH), largely mirrors the increase in obesity and type 2 diabetes (T2D) and reflects the hepatic manifestation of an altered metabolic state. Indeed, metabolic syndrome, defined as a constellation of obesity, insulin resistance, hyperglycemia, dyslipidemia and hypertension, is the major risk factor predisposing the NAFLD and NASH. There are multiple potential pharmacologic strategies to rebalance aspects of disordered metabolism in NAFLD. These include therapies aimed at reducing hepatic steatosis by directly modulating lipid metabolism within the liver, inhibiting fructose metabolism, altering delivery of free fatty acids from the adipose to the liver by targeting insulin resistance and/or adipose metabolism, modulating glycemia, and altering pleiotropic metabolic pathways simultaneously. Emerging data from human genetics also supports a role for metabolic drivers in NAFLD and risk for progression to NASH. In this review, we highlight the prominent metabolic drivers of NAFLD pathogenesis and discuss the major metabolic targets of NASH pharmacotherapy.
Collapse
Key Words
- acc, acetyl-coa carboxylase
- alt, alanine aminotransferase
- aso, anti-sense oligonucleotide
- ast, aspartate aminotransferase
- chrebp, carbohydrate response element binding protein
- ci, confidence interval
- dgat, diacylglycerol o-acyltransferase
- dnl, de novo lipogenesis
- fas, fatty acid synthase
- ffa, free fatty acid
- fgf, fibroblast growth factor
- fxr, farnesoid x receptor
- glp-1, glucagon-like peptide-1
- hdl, high-density lipoprotein
- homa-ir, homeostatic model assessment of insulin resistance
- ldl, low-density lipoprotein
- nafld, nonalcoholic fatty liver disease
- nas, nonalcoholic fatty liver disease activity score
- nash, nonalcoholic steatohepatitis
- or, odds ratio
- pdff, proton density fat fraction
- ppar, peroxisome proliferator-activated receptor
- sglt2, sodium glucose co-transporter 2
- srebp-1c, sterol regulatory element binding protein-1c
- t2d, type 2 diabetes
- t2dm, type 2 diabetes mellitus
- tg, triglyceride
- th, thyroid hormone
- thr, thyroid hormone receptor
- treg, regulatory t cells
- tzd, thiazolidinedione
- vldl, very low-density lipoprotein
Collapse
Affiliation(s)
- William P Esler
- Internal Medicine Research Unit, Pfizer Worldwide Research, Development, and Medical, Cambridge, Massachusetts
| | - Kendra K Bence
- Internal Medicine Research Unit, Pfizer Worldwide Research, Development, and Medical, Cambridge, Massachusetts.
| |
Collapse
|
224
|
Naini A, Sasse F, Brönstrup M. The intriguing chemistry and biology of soraphens. Nat Prod Rep 2019; 36:1394-1411. [PMID: 30950477 DOI: 10.1039/c9np00008a] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Covering: up to the end of 2018Soraphens are a class of polyketide natural products discovered from the myxobacterial strain Sorangium cellulosum. The review is intended to provide an overview on the biosynthesis, chemistry and biological properties of soraphens, that represent a prime example to showcase the value of natural products as tools to decipher cell biology, but also to open novel therapeutic options. The prototype soraphen A is an inhibitor of acetyl coenzyme A carboxylase (ACC1/2), an enzyme that converts acetyl-CoA to malonyl-CoA and thereby controls essential cellular metabolic processes like lipogenesis and fatty acid oxidation. Soraphens illustrate how the inhibition of a single target (ACC1/2) may be explored to treat various pathological conditions: initially developed as a fungicide, efforts in the past decade were directed towards human diseases, including diabetes/obesity, cancer, hepatitis C, HIV, and autoimmune disease - and led to a synthetic molecule, discovered by virtual screening of the allosteric binding site of soraphen in ACC, that is currently in phase 2 clinical trials. We will summarize how structural analogs of soraphen A have been generated through extensive isolation efforts, genetic engineering of the biosynthetic gene cluster, semisynthesis as well as partial and total synthesis.
Collapse
Affiliation(s)
- Arun Naini
- Department of Chemical Biology, Helmholtz Centre for Infection Research, 38124 Braunschweig, Germany. and Center of Biomolecular Drug Research (BMWZ), Leibniz University, 30159 Hannover, Germany
| | - Florenz Sasse
- Center of Biomolecular Drug Research (BMWZ), Leibniz University, 30159 Hannover, Germany
| | - Mark Brönstrup
- Department of Chemical Biology, Helmholtz Centre for Infection Research, 38124 Braunschweig, Germany. and Center of Biomolecular Drug Research (BMWZ), Leibniz University, 30159 Hannover, Germany and German Center for Infection Research (DZIF), Partner Site Hannover-Braunschweig, Germany
| |
Collapse
|
225
|
Diagnostic value of MRI-PDFF for hepatic steatosis in patients with non-alcoholic fatty liver disease: a meta-analysis. Eur Radiol 2019; 29:3564-3573. [DOI: 10.1007/s00330-019-06072-4] [Citation(s) in RCA: 135] [Impact Index Per Article: 27.0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2018] [Revised: 01/21/2019] [Accepted: 02/05/2019] [Indexed: 02/07/2023]
|
226
|
Orlicky DJ, Libby AE, Bales ES, McMahan RH, Monks J, La Rosa FG, McManaman JL. Perilipin-2 promotes obesity and progressive fatty liver disease in mice through mechanistically distinct hepatocyte and extra-hepatocyte actions. J Physiol 2019; 597:1565-1584. [PMID: 30536914 PMCID: PMC6418763 DOI: 10.1113/jp277140] [Citation(s) in RCA: 55] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2018] [Accepted: 12/05/2018] [Indexed: 12/22/2022] Open
Abstract
KEY POINTS Wild-type mice and mice with hepatocyte-specific or whole-body deletions of perilipin-2 (Plin2) were used to define hepatocyte and extra-hepatocyte effects of altered cellular lipid storage on obesity and non-alcoholic fatty liver disease (NAFLD) pathophysiology in a Western-diet (WD) model of these disorders. Extra-hepatocyte actions of Plin2 are responsible for obesity, adipose inflammation and glucose clearance abnormalities in WD-fed mice. Hepatocyte and extra-hepatic actions of Plin2 mediate fatty liver formation in WD-fed mice through distinct mechanisms. Hepatocyte-specific actions of Plin2 are primary mediators of immune cell infiltration and fibrotic injury in livers of obese mice. ABSTRACT Non-alcoholic fatty liver disease (NAFLD) is an obesity- and insulin resistance-related metabolic disorder with progressive pathology. Perilipin-2 (Plin2), a ubiquitously expressed cytoplasmic lipid droplet scaffolding protein, is hypothesized to contribute to NAFLD in humans and rodent models through effects on cellular lipid metabolism. In this study, we delineate hepatocyte-specific and extra-hepatocyte Plin2 mechanisms regulating the effects of obesity and insulin resistance on NAFLD pathophysiology in mice fed an obesogenic Western-style diet (WD). Total Plin2 deletion (Plin2-Null) fully protected WD-fed mice from obesity, insulin resistance, adipose inflammation, steatohepatitis (NASH) and liver fibrosis found in WT animals. Hepatocyte-specific Plin2 deletion (Plin2-HepKO) largely protected against NASH and fibrosis and partially protected against steatosis in WD-fed animals, but it did not protect against obesity, insulin resistance, or adipose inflammation. Significantly, total or hepatocyte-specific Plin2 deletion impaired WD-induced monocyte recruitment and pro-inflammatory macrophage polarization found in livers of WT mice. Analyses of the molecular and cellular processes mediating steatosis, inflammation and fibrosis identified differences in total and hepatocyte-specific actions of Plin2 on the mechanisms promoting NAFLD pathophysiology. Our results demonstrate that hepatocyte-specific actions of Plin2 are central to the initiation and pathological progression of NAFLD in obese and insulin-resistant mice through effects on immune cell recruitment and fibrogenesis. Conversely, extra-hepatocyte Plin2 actions promote NAFLD pathophysiology through effects on obesity, inflammation and insulin resistance. Our findings provide new insight into hepatocyte and extra-hepatocyte mechanisms underlying NAFLD development and progression.
Collapse
Affiliation(s)
- David J. Orlicky
- Department of PathologyUniversity of Colorado School of MedicineAuroraCOUSA
| | - Andrew E. Libby
- Graduate Program in Integrated PhysiologyUniversity of Colorado School of MedicineAuroraCOUSA
- Division of Reproductive SciencesUniversity of Colorado School of MedicineAuroraCOUSA
| | - Elise S. Bales
- Division of Reproductive SciencesUniversity of Colorado School of MedicineAuroraCOUSA
| | - Rachel H. McMahan
- Division of Gastroenterology and HepatologyUniversity of Colorado School of MedicineAuroraCOUSA
| | - Jenifer Monks
- Division of Reproductive SciencesUniversity of Colorado School of MedicineAuroraCOUSA
| | | | - James L. McManaman
- Graduate Program in Integrated PhysiologyUniversity of Colorado School of MedicineAuroraCOUSA
- Division of Reproductive SciencesUniversity of Colorado School of MedicineAuroraCOUSA
- Center for Human NutritionUniversity of Colorado School of MedicineAuroraCOUSA
| |
Collapse
|
227
|
Yu Y, Cai J, She Z, Li H. Insights into the Epidemiology, Pathogenesis, and Therapeutics of Nonalcoholic Fatty Liver Diseases. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2019; 6:1801585. [PMID: 30828530 PMCID: PMC6382298 DOI: 10.1002/advs.201801585] [Citation(s) in RCA: 98] [Impact Index Per Article: 19.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/12/2018] [Revised: 11/14/2018] [Indexed: 05/05/2023]
Abstract
Nonalcoholic fatty liver disease (NAFLD) is the most common liver disease which affects ≈25% of the adult population worldwide, placing a tremendous burden on human health. The disease spectrum ranges from simple steatosis to steatohepatitis, fibrosis, and ultimately, cirrhosis and carcinoma, which are becoming leading reasons for liver transplantation. NAFLD is a complex multifactorial disease involving myriad genetic, metabolic, and environmental factors; it is closely associated with insulin resistance, metabolic syndrome, obesity, diabetes, and many other diseases. Over the past few decades, countless studies focusing on the investigation of noninvasive diagnosis, pathogenesis, and therapeutics have revealed different aspects of the mechanism and progression of NAFLD. However, effective pharmaceuticals are still in development. Here, the current epidemiology, diagnosis, animal models, pathogenesis, and treatment strategies for NAFLD are comprehensively reviewed, emphasizing the outstanding breakthroughs in the above fields and promising medications in and beyond phase II.
Collapse
Affiliation(s)
- Yao Yu
- Department of CardiologyRenmin Hospital of Wuhan UniversityJiefang Road 238Wuhan430060P. R. China
- Institute of Model AnimalWuhan UniversityDonghu Road 115Wuhan430071P. R. China
| | - Jingjing Cai
- Department of CardiologyRenmin Hospital of Wuhan UniversityJiefang Road 238Wuhan430060P. R. China
- Institute of Model AnimalWuhan UniversityDonghu Road 115Wuhan430071P. R. China
| | - Zhigang She
- Department of CardiologyRenmin Hospital of Wuhan UniversityJiefang Road 238Wuhan430060P. R. China
- Institute of Model AnimalWuhan UniversityDonghu Road 115Wuhan430071P. R. China
| | - Hongliang Li
- Department of CardiologyRenmin Hospital of Wuhan UniversityJiefang Road 238Wuhan430060P. R. China
- Institute of Model AnimalWuhan UniversityDonghu Road 115Wuhan430071P. R. China
| |
Collapse
|
228
|
|
229
|
Goedeke L, Bates J, Vatner DF, Perry RJ, Wang T, Ramirez R, Li L, Ellis MW, Zhang D, Wong KE, Beysen C, Cline GW, Ray AS, Shulman GI. Acetyl-CoA Carboxylase Inhibition Reverses NAFLD and Hepatic Insulin Resistance but Promotes Hypertriglyceridemia in Rodents. Hepatology 2018; 68:2197-2211. [PMID: 29790582 PMCID: PMC6251774 DOI: 10.1002/hep.30097] [Citation(s) in RCA: 168] [Impact Index Per Article: 28.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/23/2017] [Accepted: 04/30/2018] [Indexed: 12/13/2022]
Abstract
Pharmacologic inhibition of acetyl-CoA carboxylase (ACC) enzymes, ACC1 and ACC2, offers an attractive therapeutic strategy for nonalcoholic fatty liver disease (NAFLD) through simultaneous inhibition of fatty acid synthesis and stimulation of fatty acid oxidation. However, the effects of ACC inhibition on hepatic mitochondrial oxidation, anaplerosis, and ketogenesis in vivo are unknown. Here, we evaluated the effect of a liver-directed allosteric inhibitor of ACC1 and ACC2 (Compound 1) on these parameters, as well as glucose and lipid metabolism, in control and diet-induced rodent models of NAFLD. Oral administration of Compound 1 preferentially inhibited ACC enzymatic activity in the liver, reduced hepatic malonyl-CoA levels, and enhanced hepatic ketogenesis by 50%. Furthermore, administration for 6 days to high-fructose-fed rats resulted in a 20% reduction in hepatic de novo lipogenesis. Importantly, long-term treatment (21 days) significantly reduced high-fat sucrose diet-induced hepatic steatosis, protein kinase C epsilon activation, and hepatic insulin resistance. ACCi treatment was associated with a significant increase in plasma triglycerides (approximately 30% to 130%, depending on the length of fasting). ACCi-mediated hypertriglyceridemia could be attributed to approximately a 15% increase in hepatic very low-density lipoprotein production and approximately a 20% reduction in triglyceride clearance by lipoprotein lipase (P ≤ 0.05). At the molecular level, these changes were associated with increases in liver X receptor/sterol response element-binding protein-1 and decreases in peroxisome proliferator-activated receptor-α target activation and could be reversed with fenofibrate co-treatment in a high-fat diet mouse model. Conclusion: Collectively, these studies warrant further investigation into the therapeutic utility of liver-directed ACC inhibition for the treatment of NAFLD and hepatic insulin resistance.
Collapse
Affiliation(s)
- Leigh Goedeke
- Department of Internal Medicine, Yale University School of Medicine, New Haven CT, 06520
| | | | - Daniel F. Vatner
- Department of Internal Medicine, Yale University School of Medicine, New Haven CT, 06520
| | - Rachel J. Perry
- Department of Internal Medicine, Yale University School of Medicine, New Haven CT, 06520
| | - Ting Wang
- Gilead Sciences Inc., Foster City CA 94404
| | | | - Li Li
- Gilead Sciences Inc., Foster City CA 94404
| | - Matthew W. Ellis
- Department of Cellular & Molecular Physiology, Yale University School of Medicine, New Haven CT 06520
| | - Dongyan Zhang
- Department of Internal Medicine, Yale University School of Medicine, New Haven CT, 06520
| | | | | | - Gary W. Cline
- Department of Internal Medicine, Yale University School of Medicine, New Haven CT, 06520
| | | | - Gerald I. Shulman
- Department of Internal Medicine, Yale University School of Medicine, New Haven CT, 06520,Department of Cellular & Molecular Physiology, Yale University School of Medicine, New Haven CT 06520,Howard Hughes Medical Institute, Yale University School of Medicine, New Haven CT 06520
| |
Collapse
|
230
|
Sayiner M, Lam B, Golabi P, Younossi ZM. Advances and challenges in the management of advanced fibrosis in nonalcoholic steatohepatitis. Therap Adv Gastroenterol 2018; 11:1756284818811508. [PMID: 30479664 PMCID: PMC6243399 DOI: 10.1177/1756284818811508] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/20/2018] [Accepted: 10/16/2018] [Indexed: 02/04/2023] Open
Abstract
Nonalcoholic fatty liver disease (NAFLD) is rapidly becoming the most common type of chronic liver disease worldwide. From the spectrum of NAFLD, it is nonalcoholic steatohepatitis (NASH) that predominantly predisposes patients to higher risk for development of cirrhosis and hepatocellular carcinoma. There is growing evidence that the risk of progression to cirrhosis and hepatocellular carcinoma is not uniform among all patients with NASH. In fact, NASH patients with increasing numbers of metabolic diseases such as diabetes, hypertension, visceral obesity and dyslipidemia are at a higher risk of mortality. Additionally, patients with higher stage of liver fibrosis are also at increased risk of mortality. In this context, NASH patients with fibrosis are in the most urgent need of treatment. Also, the first line of treatment for NASH is lifestyle modification with diet and exercise. Nevertheless, the efficacy of lifestyle modification is quite limited. Additionally, vitamin E and pioglitazone may be considered for subset of patients with NASH. There are various medications targeting one or more steps in the pathogenesis of NASH being developed. These drug regimens either alone or in combination, may provide potential treatment option for patients with NASH.
Collapse
Affiliation(s)
- Mehmet Sayiner
- Department of Medicine, Inova Fairfax Hospital, Falls Church, VA,Betty and Guy Beatty Center for Integrated Research, Inova Health System, Falls Church, VA
| | - Brian Lam
- Betty and Guy Beatty Center for Integrated Research, Inova Health System, Falls Church, VA
| | - Pegah Golabi
- Betty and Guy Beatty Center for Integrated Research, Inova Health System, Falls Church, VA
| | | |
Collapse
|
231
|
Nakagawa H, Hayata Y, Kawamura S, Yamada T, Fujiwara N, Koike K. Lipid Metabolic Reprogramming in Hepatocellular Carcinoma. Cancers (Basel) 2018; 10:cancers10110447. [PMID: 30445800 PMCID: PMC6265967 DOI: 10.3390/cancers10110447] [Citation(s) in RCA: 101] [Impact Index Per Article: 16.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2018] [Revised: 11/10/2018] [Accepted: 11/13/2018] [Indexed: 12/23/2022] Open
Abstract
Metabolic reprogramming for adaptation to the local environment has been recognized as a hallmark of cancer. Although alterations in fatty acid (FA) metabolism in cancer cells have received less attention compared to other metabolic alterations such as glucose or glutamine metabolism, recent studies have uncovered the importance of lipid metabolic reprogramming in carcinogenesis. Obesity and nonalcoholic steatohepatitis (NASH) are well-known risk factors of hepatocellular carcinoma (HCC), and individuals with these conditions exhibit an increased intake of dietary FAs accompanied by enhanced lipolysis of visceral adipose tissue due to insulin resistance, resulting in enormous exogenous FA supplies to hepatocytes via the portal vein and lymph vessels. This “lipid-rich condition” is highly characteristic of obesity- and NASH-driven HCC. Although the way in which HCC cells adapt to such a condition and exploit it to aid their progression is not understood, we recently obtained new insights into this mechanism through lipid metabolic reprogramming. In addition, accumulating evidence supports the importance of lipid metabolic reprogramming in various situations of hepatocarcinogenesis. Thus, in this review, we discuss the latest findings regarding the role of FA metabolism pathways in hepatocarcinogenesis, focusing on obesity- and NASH-driven lipid metabolic reprogramming.
Collapse
Affiliation(s)
- Hayato Nakagawa
- Department of Gastroenterology, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8655, Japan.
| | - Yuki Hayata
- Department of Gastroenterology, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8655, Japan.
| | - Satoshi Kawamura
- Department of Gastroenterology, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8655, Japan.
| | - Tomoharu Yamada
- Department of Gastroenterology, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8655, Japan.
| | - Naoto Fujiwara
- Department of Gastroenterology, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8655, Japan.
| | - Kazuhiko Koike
- Department of Gastroenterology, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8655, Japan.
| |
Collapse
|
232
|
Mehal WZ. Acetyl-Coenzyme A carboxylase inhibition Delivers, as Anticipated, for Patients With Nonalcoholic Steatohepatitis. Gastroenterology 2018; 155:1304-1306. [PMID: 30308191 DOI: 10.1053/j.gastro.2018.10.013] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Affiliation(s)
- Wajahat Z Mehal
- Yale Non-Alcoholic Fatty Liver Disease Program, Yale University School of Medicine, New Haven, Connecticut.
| |
Collapse
|
233
|
Neuschwander-Tetri BA. Pharmacologic Management of Nonalcoholic Steatohepatitis. Gastroenterol Hepatol (N Y) 2018; 14:582-589. [PMID: 30846912 PMCID: PMC6384397] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/09/2023]
Abstract
Nonalcoholic steatohepatitis (NASH) is increasingly recognized as a major form of chronic liver disease in adults and children. Although improved dietary habits and regular exercise remain the primary recommendations for patients with or at risk of having NASH, implementing and sustaining these lifestyle changes have proven to be challenging. Pharmacologic approaches are now being evaluated to prevent the development of cirrhosis and its complications in the approximately 1% of the population of countries consuming a Western diet at risk for NASH. Although some therapies are available for the treatment of NASH, none is currently approved by the US Food and Drug Administration. Approval of new drugs for NASH is expected within the next several years. Thus, a rational approach to understanding how these drugs work is needed. This article explains how the many new therapies that are currently in clinical trials address the varied mechanisms by which patients develop NASH and NASH-induced cirrhosis.
Collapse
Affiliation(s)
- Brent A Neuschwander-Tetri
- Dr Neuschwander-Tetri is a professor of internal medicine in the Division of Gastroenterology and Hepatology at Saint Louis University in St. Louis, Missouri
| |
Collapse
|