Review Open Access
Copyright ©The Author(s) 2023. Published by Baishideng Publishing Group Inc. All rights reserved.
World J Hepatol. Jun 27, 2023; 15(6): 755-774
Published online Jun 27, 2023. doi: 10.4254/wjh.v15.i6.755
Treatment of liver fibrosis: Past, current, and future
Chun-Ye Zhang, Bond Life Sciences Center, University of Missouri, Columbia, MO 65211, United States
Shuai Liu, Department of Radiology,The First Affiliated Hospital, Zhejiang University, Hangzhou 310006, Zhejiang Province, China
Ming Yang, Department of Surgery, University of Missouri, Columbia, MO 65211, United States
ORCID number: Chun-Ye Zhang (0000-0003-2567-029X); Shuai Liu (0000-0001-9695-2492); Ming Yang (0000-0002-4895-5864).
Author contributions: Zhang CY, Liu S, and Yang M designed and collected data, wrote, revised, and finalized the manuscript; all authors contributed equally and shared the first authorship.
Conflict-of-interest statement: The authors declare no conflicts of interest.
Open-Access: This article is an open-access article that was selected by an in-house editor and fully peer-reviewed by external reviewers. It is distributed in accordance with the Creative Commons Attribution NonCommercial (CC BY-NC 4.0) license, which permits others to distribute, remix, adapt, build upon this work non-commercially, and license their derivative works on different terms, provided the original work is properly cited and the use is non-commercial. See: https://creativecommons.org/Licenses/by-nc/4.0/
Corresponding author: Ming Yang, DVM, PhD, Postdoctoral Fellow, Department of Surgery, University of Missouri, Room 2203, NexGen Precision Building, 1030 Hitt Street, Columbia, MO 65211, United States. yangmin@health.missouri.edu
Received: March 12, 2023
Peer-review started: March 12, 2023
First decision: March 23, 2023
Revised: April 1, 2023
Accepted: April 18, 2023
Article in press: April 18, 2023
Published online: June 27, 2023

Abstract

Liver fibrosis accompanies the progression of chronic liver diseases independent of etiologies, such as hepatitis viral infection, alcohol consumption, and metabolic-associated fatty liver disease. It is commonly associated with liver injury, inflammation, and cell death. Liver fibrosis is characterized by abnormal accumulation of extracellular matrix components that are expressed by liver myofibroblasts such as collagens and alpha-smooth actin proteins. Activated hepatic stellate cells contribute to the major population of myofibroblasts. Many treatments for liver fibrosis have been investigated in clinical trials, including dietary supplementation (e.g., vitamin C), biological treatment (e.g., simtuzumab), drug (e.g., pegbelfermin and natural herbs), genetic regulation (e.g., non-coding RNAs), and transplantation of stem cells (e.g., hematopoietic stem cells). However, none of these treatments has been approved by Food and Drug Administration. The treatment efficacy can be evaluated by histological staining methods, imaging methods, and serum biomarkers, as well as fibrosis scoring systems, such as fibrosis-4 index, aspartate aminotransferase to platelet ratio, and non-alcoholic fatty liver disease fibrosis score. Furthermore, the reverse of liver fibrosis is slowly and frequently impossible for advanced fibrosis or cirrhosis. To avoid the life-threatening stage of liver fibrosis, anti-fibrotic treatments, especially for combined behavior prevention, biological treatment, drugs or herb medicines, and dietary regulation are needed. This review summarizes the past studies and current and future treatments for liver fibrosis.

Key Words: Liver fibrosis, Molecular mechanism, Therapeutic targets, Treatments, Clinical trials

Core Tip: Liver fibrosis accompanies the progression of chronic liver diseases independent of their etiologies. The initiation and progression of liver fibrosis are mainly driven by liver inflammation, cell death, and metabolic dysregulation, which cause the activation of hepatic stellate cells and excessive accumulation of extracellular matrix proteins. Without effective treatments, liver fibrosis can lead to cirrhosis and primary liver cancer. To date, current therapeutic options for liver fibrosis are limited to prevent the initial causing factors for liver inflammation, hepatocyte cell death, and oxidative stress. However, the reverse of liver fibrosis is slowly and frequently impossible for advanced fibrosis or cirrhosis. To avoid the life-threatening stage of liver fibrosis, anti-fibrotic treatments including biological, medicines, dietary change, and behavior prevention are needed, especially for combined therapy.



INTRODUCTION

Liver fibrosis accompanies the progression of chronic liver diseases independent of etiologies[1], such as hepatitis viral infection, alcohol abuse, and metabolic-associated fatty liver disease (MAFLD). It is commonly associated with liver injury, inflammation, and cell death. Abnormal accumulation of extracellular matrix (ECM) components expressed by liver myofibroblasts, such as collagens and alpha-smooth actin proteins, are the markers of hepatic fibrogenesis[2]. Activated hepatic stellate cells (HSCs) contribute to the major population of myofibroblasts in liver fibrosis[3]. Although many drugs have been investigated in clinical trials, there are no Food and Drug Administration (FDA)-approved treatments for liver fibrosis.

The activation of HSC is a complex pathogenesis in liver fibrosis[4]. Many factors including intrahepatic and extrahepatic factors can drive HSC activation to induce liver fibrosis. A variety of molecular signaling pathways are involved in the regulation of HSC activation[1,5], such as transforming growth factor-β (TGF-β), Toll-like receptors (TLRs), and epigenetic signals (e.g., microRNAs, or miRNAs). The activation of HSCs can be divided into two phases, the initiation and perpetuation phases. RNA sequencing results have shown that fibrogenic transcriptional programs in the initiation phase are also active in the perpetuation phase; therefore, targeting the initial activation of HSC is also critically important for live fibrosis treatment[6].

In this review, the cellular and molecular mechanisms of liver fibrosis are reviewed. Importantly, the currently available treatments for liver fibrosis are summarized and discussed. Some pros and cons of available treatments are discussed. In addition, the future direction for liver fibrosis therapy is predicted.

INITIATION OF LIVER FIBROSIS: CELLULAR AND MOLECULAR MECHANISMS
Hepatic cell death

Liver cell death and inflammation are the initial events in chronic liver disease independent of etiologies. Many factors can cause liver injury and hepatic cell death and inflammation[7,8], including hepatitis viral infection, alcohol consumption, metabolic liver disease, abnormal bile acid products, and genetic factors. These pathogenic factors cause immune cell inflammation, hepatocyte death, mitochondrial dysfunction, and endoplasmic reticulum stress (Figure 1), resulting in HSC activation and differentiation to myofibroblasts to lead to liver fibrosis[5,9].

Figure 1
Figure 1 Factors causing the activation of hepatic stellate cells and liver fibrosis. Many factors can cause liver injury and hepatic cell death and inflammation, including hepatitis viral infection, alcohol consumption, metabolic liver disease, abnormal bile acid products, and genetic factors. These pathogenic factors cause immune cell inflammation, hepatocyte death, oxidative stress, and endoplasmic reticulum stress, resulting in hepatic stellate cell activation and differentiation to myofibroblasts to lead to liver fibrosis. All cartoons in this figure were prepared using Biorender (https://biorender.com). ER: Endoplasmic reticulum; HSC: Hepatic stellate cell; ROS: Reactive oxygen species.

In non-alcoholic steatohepatitis (NASH), hepatocyte death results in the infiltration of monocyte-derived macrophages and upregulation of the expression of inflammatory cytokines[9], such as tumor necrosis factor α (TNF-α), TGF-β1, and interleukin-1β (IL-1β), Hepatocyte death can be classified into programed cell death including pyroptosis, apoptosis, necroptosis, ferroptosis, and autophagy-mediated cell death (Figure 2), as well as non-programed cell death (necrosis). In chronic liver disease, different types of cell death may be associated with the progression of liver fibrosis and end-stage of liver disease, such as hepatocellular carcinoma (HCC). Single-cell RNA sequencing coupled with spatial mapping approaches has been started to dissect the key cellular and molecular functions in liver disease[10].

Figure 2
Figure 2 Programmed cell death subtypes of hepatic cells, including apoptosis, necroptosis, pyroptosis, ferroptosis, and autophagy-mediated cell death. Apoptosis, necroptosis, pyroptosis, and ferroptosis are programmed forms of cell death, while necrosis is unprogrammed cell death. Autophagy-mediated cell death should be defined when autophagic flux is raised without the involvement of other types of programmed cell death, and pharmacological or genetic inhibition of autophagy blocks cell death. DAMPs: Danger-associated molecular patterns; FADD: Fas-associated protein with a death domain; FAS: Fas cell surface death receptor; MLKL: Mixed lineage kinase domain-like; NLRP3: Nod-like receptor family, pyrin domain containing 3; PAMPs: Pathogen-associated molecular patterns; RIPK1/3: receptor-interacting protein kinase 1/3; TNF: Tumor necrosis factor; TNFR: Tumor necrosis factor receptor. All cartoons in this figure were prepared using Biorender (https://biorender.com).

Pyroptosis is an inflammatory cell death, associated with cell membrane rupture by cleaved gasdermin D[11]. The cleave of gasdermin D is induced by the activation of caspase-1 or caspase-11/4/5[12]. For example, in mice with non-alcoholic fatty liver disease (NAFLD), feeding a high-fat diet can increase the expression of caspase-11 to cause pyroptosis of bone marrow monocyte-derived macrophages by cleave gasdermin D[13]. The expression of pyroptosis-related indicators including gasdermin D, IL-1β, and IL-18 has been shown to be increased in human patients with liver fibrosis and mice with CCl4-induced fibrosis[14]. In addition, S100 calcium-binding protein A8 plays an essential role in macrophage pyroptosis in liver fibrosis, by inducing the expression of nucleotide-binding domain leucine-rich repeat-receptor, pyrin domain-containing-3 (NLRP3) inflammasome, pro-IL-1β, and pro-IL-18 via the activation of TLR4/nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB) signaling pathway[14].

Apoptosis is a form of programmed cell death that occurs in all liver cell types. In hepatocytes, kindlin-2 deficiency can increase the apoptosis of hepatocytes, resulting in liver fibrosis and accumulation of ECM components by activating the TNF signaling pathway[15]. In NAFLD, microRNAs such as miR-22 from adipocyte-derived exosomes can cause hepatocyte apoptosis to increase hepatic inflammation, lipid accumulation, and fibrosis by regulating sirtuin 1 expression[16]. In contrast, promoting the apoptosis of HSCs can inhibit and reverse liver fibrosis. For example, treatment with gomisin D can inhibit CCl4-induced HSC proliferation and activation in mice and increase HSC apoptosis to reduce liver fibrosis by regulating the platelet-derived growth factor receptor β signaling pathway[17].

Necroptosis is a regulated cell death that has the features of apoptosis and necrosis. Necroptosis can be induced by TLRs, interferons, and death receptors[18], which is mediated by receptor-interacting protein kinase-3 and its substrate mixed lineage kinase-like[19]. In addition, receptor-interacting serine/threonine-protein kinase 1 has an important role in this process (Figure 2). It contributes to hepatocyte death in NASH. Necroptotic hepatocytes cannot be removed by liver macrophages due to the activation of "don't-eat-me" signaling pathway, the CD47/signal regulatory protein alpha axis[20].

Ferroptosis is an intracellular iron-dependent lytic cell death which is different from apoptosis and necrosis[21,22]. Excessive iron accumulation and the inhibition of glutathione peroxidase 4 trigger ferroptosis, which causes cell plasma membrane rupture[23]. Accumulating research studies have demonstrated that ferroptosis is involved in different liver diseases, including alcoholic liver disease, NASH, cirrhosis, and cancer[24,25]. Several molecular signaling pathways are involved in ferroptosis in liver diseases, such as the nuclear factor erythroid 2-related factor 2[26], heat shock protein family A member 8[27], and nuclear receptor coactivator 4[28]. Pharmacological regulation of HSC ferroptosis is a therapeutic strategy for liver fibrosis[29]. For example, curcumol can induce ferroptosis of activated HSCs to inhibit liver fibrosis by inducing autophagy[28].

Furthermore, some dying cells develop autophagosomes that trigger apoptosis and necroptosis, namely autophagy-mediated cell death (ACD). However, ACD should be defined when some criteria are met[30], including: (1) Cell death happens without the involvement of other types of programmed cell death; (2) autophagic flux is raised; and (3) pharmacological or genetic inhibition of autophagy blocks cell death. Overall, the molecules implicated in the process and signaling pathways of hepatic cell death are potential targets for liver fibrosis.

Hepatic innate and adaptive immunity

Liver resident immune cells and infiltration of myeloid cells or circulating immune cells in the liver during chronic liver injury play key roles in the activation of HSCs. For example, in NASH liver, gut microbiota-diet interplay-resulted metabolite can activate liver macrophages to produce profibrotic factors[31], such as TGF-β1. Treatments such as astaxanthin can suppress the infiltration of monocyte-derived macrophages to suppress HSC activation, liver oxidative stress response, and hepatocyte death by decreasing the expression of proinflammatory cytokines[32], such as TNF-α, TGF-β1, and IL-1β. Another study also during NAFLD progression, mast cells can increase Western diet-induced biliary and liver damage to the development of microvesicular steatosis through microRNA (miR-144-3p)-targeted signaling pathway[33].

Adaptive immune cells including T and B cells have various roles in liver fibrosis. The imbalance of liver regulatory T cells and T helper (Th) cells, such as Th1 cells and Th17 cells plays an essential role in liver fibrosis, cirrhosis, and cancer[34]. The populations of multicytokine-producing CD4 T cells were significantly increased in the livers of patients with NASH compared with patients with NAFLD[35]. The cytokines produced by these CD4 T cells include TNF-α, IFN-γ, IL-17A, and IL-10. The phenotype of T cells is also important for their functions. One study showed that the reduction of tissue-resident memory CD69+CD103CD8 T cells significantly decreased the resolution of fibrosis in NASH liver. These CD8 T cells can induce FasL/Fas-mediated apoptosis of HSCs[36]. Therefore, targeting immune cells or their secreted inflammatory cytokines is an optional treatment for liver fibrosis[37].

Furthermore, HSC activation is also associated with vascular aging[38], ischemia/reperfusion injury[39], and angiogenesis[40].

SIGNALING PATHWAYS AND MOLECULAR TARGETS FOR LIVER FIBROSIS TREATMENTS
Bile acid receptors

Farnesoid-X-receptor (FXR) and the G protein-coupled bile acid receptors are two widely studied bile acids regulating receptors, which play important roles in lipid and glucose metabolism, inflammation, fibrosis, and immune responses. Many FXR ligands have been investigated in clinical trials for NASH and liver fibrosis treatments, such as EDP-305[41,42], cilofexor[43,44], and MET409[45,46]. Hepatic concentrations of conjugated 12α-hydroxylated bile acids, such as taurodeoxycholate and glycodeoxycholate, were significantly increased in patients with NASH and mouse liver fibrosis models[47]. These bile acids contribute to HSC activation and liver fibrosis by regulating the signaling of G protein-coupled bile acid receptor 1, also known as TGR5.

Caspases

Caspases are involved in liver cell inflammation and hepatocyte cell death. Meanwhile, they play an essential role in liver fibrogenesis. For example, caspase 3-deficient mice on a methionine- and choline-deficient diet had reduced liver collagen production compared to wild-type mice[48]. Lipotoxicity can induce caspase-mediated apoptosis of hepatic cells and liver inflammation and injury in NAFLD. Treatment of caspase inhibitors such as emricasan (IDN-6556, a pan-caspase inhibitor) can reduce liver injury in patients with NAFLD[49].

Chemokine receptors

Chemokine receptors are commonly expressed by immune cells or inflammatory cells, which can be recruited during liver inflammation and fibrosis. For example, C-C chemokine receptor 2 (CCR2) and CCR5 are highly expressed by monocytes and subtypes of liver macrophages, which can be targeted to ameliorate liver fibrosis[50]. A recent study showed that the roles of CCR2 and CCR5 in liver macrophages are different during liver disease progression in mice with a hepatocyte-specific knock-out of NF-κB essential modulator. CCR2 oversees the recruitment of monocytes during liver injury, whereas CCR5 is needed to promote HSC activation[51].

In addition, many other chemokines and their receptors are involved in the pathogenesis of liver fibrosis, including C-X-C motif ligand 12 (CXCL12) /C-X-C receptor 4[52], chemokine (C-X3-C motif) ligand 1/C-X3-C receptor 1 (CX3CR1)[53], CCL19/CCR7[54], and CXCL12/atypical chemokine receptor 3[55].

Fibroblast growth factors

Fibroblast growth factor 15 (FGF15) is an important endocrine regulator for hepatic bile acid and lipid metabolism, which regulates gut-liver crosstalk in mice[56]. A combined treatment using an inhibitor of apical sodium-bile acid transporter (GSK233072) and adeno-associated virus 8-mediated hepatic FGF15 overexpression significantly can improve the therapeutic efficacy against NASH and fibrosis compared to either single treatment[57]. Bile acid nuclear receptor FXR plays an important role in the regulation of the expression of FGF15/19, bile acid homeostasis, and lipid metabolism, which is the target for NASH and liver fibrosis[58]. The expression of hepatic FXR and plasma FGF19 (the ortholog of mouse FGF15) was decreased in children with NASH compared to their expression in healthy subjects[59].

Galectins

Galectins are carbohydrate-binding proteins and play important roles in liver inflammation, immune response, and fibrosis. Galectin-1 (Gal-1) was shown to be highly expressed in the stroma of HCC by cancer-associated fibroblasts. Silencing Gal-1 in these fibroblasts can suppress inflammation and tumor progression[60]. The serum level of Gal-3 was increased in patients with advanced cirrhosis, and liver expression of Gal-3 was also correlated with liver disease severity and inflammation[61].

Lysyl oxidase family members

Lysyl oxidase (LOX) family members are extracellular copper-dependent enzymes, including LOX, lysyl oxidase-like 1 to 4 (LOXL1 to 4) members, which play important roles in the cross-linking of ECM proteins in fibrosis and carcinogenesis. Inhibition of pan-LOX family, LOX, LOXL1, or LOXL2 has been shown to prevent fibrogenesis and accelerate the reversal of liver fibrosis, as well as fibrosis in other organs. However, the roles of LOX family members as therapeutic targets for liver fibrosis need further to be evaluated[62].

NLRP3 inflammasome

Nucleotide-binding oligomerization (NLR) family pyrin domain-containing 3 (NLRP3) plays a pivotal role in liver fibrosis. Activation of NLRP3 can lead to the inflammatory response through the secretion of IL-1β and IL-18 and activation of caspase-1[63], which is involved in liver cell pyroptosis[64]. Activation of NLRP3 inflammasome can induce hepatocyte pyroptosis and liver fibrosis, while inhibiting the activation of NLRP3 inflammasome can inhibit the development of NAFLD and NASH in animal models[65,66]. Activation of NLRP3 inflammasome in pyroptosis is mediated by canonical caspase-1-mediated signaling pathway and noncanonical caspase-11-mediated signaling pathway[67].

Peroxisome proliferator-activated receptors (PPARs)

PPARs, comprised of three subtypes PPARα, β/δ, and γ, play important roles in liver lipid metabolism, inflammation, and fibrosis[68-70]. The expression of liver PPARα was shown to be negatively correlated with NASH severity, visceral fat accumulation, and insulin resistance in human patients[71]. Treatment with PPARα/γ dual agonists decreased the concentrations of total cholesterol, triglyceride (TG), and inflammatory cytokine levels in serum, reduced hepatic steatosis, infiltration of inflammatory cells, and decreased the expression of lipogenic gene and NF-κB protein[72]. Another study showed that the levels of very low-density lipoprotein receptors (VLDLR) were increased in PPARβ/δ-deficient mice. In patients with hepatic steatosis, the mRNA levels of PPARβ/δ were suppressed and associated with an increase in VLDLR levels[73]. A pre-clinical study showed that treatment with pan-PPAR agonist lanifibranor can significantly decrease portal pressure and liver inflammation and induce fibrosis regression[74].

TGF-β/Smad

TGF-β/Smad is the most well-studied signaling pathway in fibrosis. SMAD proteins are essential intracellular effectors of TGF-β and show different roles in liver fibrosis[75], including pro-fibrotic functions (e.g., SMAD3 and SMAD4) and protective functions (e.g., SMAD2 and SMAD7). In addition, many studies have demonstrated that regulating the signaling pathway of TGF-β/Smad can prevent liver fibrosis[76], as well as the protein kinase B (PKB, or AKT)/Forkhead box O3 (FOXO3) signaling pathway.

Wnt/β-catenin

Proteins-derived from human amniotic mesenchymal stem cells, including insulin-like growth factor binding protein-3, Dickkopf-1, and DKK-3, can inhibit HSC activation by suppressing Wnt/β-catenin signaling pathway in vitro[77]. In vivo study also showed that treatment of niclosamide in rats can prevent CCl4-induced liver fibrosis by inhibiting the Wnt/β-catenin pathway and glutaminolysis[78]. Another study also showed that Wnt3a can upregulate the expression of protein regulator of cytokinesis 1 to active β-catenin signaling to promote liver fibrosis[79]. The interaction of β-catenin/transcription factor 4 (TCF4) has been shown to increase during liver fibrosis in mice with bile duct ligation (BDL)[80]. Treatment with ICG-001, an inhibitor of the interaction between cyclic adenosine monophosphate response element binding protein binding protein and β-catenin, together with LF3, a small molecule antagonist that inhibits β-catenin/TCF4 transcriptional activity, can reduce liver fibrosis[80].

Yes-associated protein (YAP)

YAP plays a pivotal role in the sensitivity of HSCs to ferroptosis, apoptosis, and senescence in fibrotic livers. Selective depletion of YAP in myofibroblastic HSCs or activated HSCs can promote their senescence or apoptosis to reduce liver injury and fibrosis[81]. Taurocholic acid can induce the activation of HSCs through the sphingosine-1-phosphate receptor 2/YAP/p38 mitogen-activated protein kinase (p38 MAPK)[82].

CURRENT DIAGNOSIS FOR LIVER FIBROSIS

The golden standard method for liver fibrosis diagnosis is liver biopsy. Histological or histochemical staining can be used to stain the cells or the extracellular matrix proteins to identify liver fibrosis. Common histological staining methods for liver fibrosis evaluation are hematoxylin-eosin staining with Masson's trichrome or Sirius Red staining[83]. Due to the pain and the risk of potential complications of liver biopsy, non-invasive techniques (e.g., elastography scanning) and biomarkers (e.g., aminotransferase to platelet ratio (APRI): The aminotransferase/platelet ratio index) can be applied for diagnosing liver fibrosis[84]. Many available scoring systems can be applied for liver fibrosis diagnosis and evaluation, including fibrosis-4 index (FIB-4), APRI, and NAFLD fibrosis score (NFS)[85].

Imaging methods are commonly applied in the clinic to evaluate the progression of liver fibrosis. For example, ultrasound elastography techniques can be applied to characterize liver fibrosis and its stage in adult patients, such as vibration-controlled transient elastography, the most utilized and validated elastography method[86]. A meta-analysis study also showed that magnetic resonance elastography (MRE) and point-shear wave elastography (pSWE) can be applied for liver fibrosis diagnostic, and MRE is a more accurate imaging technique than pSWE[87]. The pooled sensitivities and specificities for MRE and pSWE were 0.94 (95% confidence level/CI: 0.89-0.97) and 0.95 (95%CI: 0.89-0.98), and 0.86 (95%CI: 0.80-0.90) and 0.88 (95%CI: 0.85-0.91), respectively. Their pooled summary receiver operating characteristic curves showed that the area under the curve (AUC) for MRE was 0.98 (95%CI: 0.96-0.99), whereas the AUC for pSWE was 0.93 (95%CI: 0.90-0.95). Another review paper has updated the conventional and molecular imaging diagnostic methods for liver fibrosis[88]. In addition, artificial intelligence models have been applied for the diagnosis of liver fibrosis[89-91]. For example, the clinical features and imaging data collected from a patient can be analyzed for liver fibrosis diagnosis using a machine learning model.

Recently, studies also have shown that miRNAs, the single-stranded, non-coding RNAs containing 21 to 23 nucleotides, are involved in the pathogenesis of liver fibrosis, which are potential biomarkers for diagnosing liver fibrosis and therapeutic targets for liver fibrosis treatment[92]. The methods for liver fibrosis diagnosis have been reviewed in some recent publications[93-96]. Here, we will not discuss more details and will focus on the treatment options for liver fibrosis.

CURRENT TREATMENT OPTIONS FOR LIVER FIBROSIS

In this section, we review some different treatment options for liver fibrosis, such as biological intervention, anti-fibrotic drugs, and other treatment strategies. These treatments either target causing factors of liver fibrosis to accelerate the recovery of liver injury, or induce the balance of liver metabolism, such as anti-hepatitis viral infection, anti-cell death treatment, and regulators of lipid metabolism.

Biological intervention

Inhibition of LOXL2 in the fibrotic tumor microenvironment can synergistically increase the efficacy of sorafenib and 5-fluorouracil for liver cancer cells[97]. However, some treatments in clinical trials did not show promising results. For example, simtuzumab is a monoclonal antibody against LOXL2. In two phase 2b clinical trials, intravenous infusions of simtuzumab (200 or 700 mg) every other day for 48 wk and 96 wk did not show promising effects to decrease liver fibrosis and the progression of cirrhosis in patients with bridging fibrosis[98]. In a pilot clinical trial, intravenous treatment of simtuzumab (700 mg) every 2 wk for 22 wk did not improve liver biopsy fibrosis score for patients with advanced liver fibrosis[99].

Drug treatment

Aramchol, a partial inhibitor of hepatic stearoyl-CoA desaturase, has been shown to improve NASH and liver fibrosis in rodents and decrease liver triglycerides and fibrosis clinical trials[100].

Anti-hepatitis viral infection drugs: Inhibition of hepatitis viral infection can suppress liver inflammation and hepatocyte death to decrease liver injury, resulting in suppression of liver fibrosis. Drugs such as faldaprevir (also known as BI 201335)[101], ribavirin (HCV treatment)[102], and peginterferon alfa-2a (HBV treatment)[103,104], have been tested in clinical trials for the treatment of liver fibrosis. In addition, many other drugs have been evaluated or are under clinical trial evaluation against hepatitis viral infection[105-107], such as simeprevir, daclatasvir, and sofosbuvir.

Cenicriviroc: C-C chemokine receptors 2 and 5 dual antagonist, has been shown to improve liver fibrosis without worsening NASH compared to the placebo in phase 2 clinical trial (Clinicaltrials.gov, NCT02217475)[108]. A phase 3 clinical trial has been designed to confirm the efficacy and safety of cenicriviroc for liver fibrosis treatment in adults with NASH[109].

Cholangitis treatment: Obeticholic acid and ursodeoxycholic acid are the only two FDA-approved medicines for the treatment of primary biliary cholangitis[110], which have the potential to cholangitis-induced liver fibrosis.

Cyclophilin inhibitors: CRV431, a pan-cyclophilin inhibitor, can decrease liver fibrosis in mice treated with CCl4 for 6 wk and mice with diet-induced NASH[111]. Another cyclophilin inhibitor NV556 also displays an antifibrotic effect in two mouse NASH models, the STAM model (streptozotocin plus a high-fat diet) and methionine- and choline-deficient diet-induced NASH model[112]. In addition, NV556 can also inhibit TGF-β1-induced activation of HSCs in vitro.

FGF regulators or analogues: Treatment of pegbelfermin (BMS-986036, 10 mg or 20 mg daily), a PEGylated human FGF21 analogue, can significantly decrease liver fat accumulation in patients with NASH without treatment-related severe adverse effects, and it can also improve liver fibrosis in patients with obesity and type 2 diabetes[113].

FXR agonists: Treatment of obeticholic acid (INT-747), a potent and orally active FXR agonist, can significantly ameliorate liver fibrosis and the histological and biological markers of NASH in patients with NASH[114].

Gal-3 inhibitors: GB1211, an inhibitor of Gal-3, can inhibit the differentiation of epithelial cells into myofibroblasts and macrophage or myofibroblast-induced fibrosis in the liver[115]. GR-MD-02 (belapectin), a galectin-3 inhibitor, has been shown to inhibit liver fibrosis and portal hypertension in rat fibrosis mode, which is safe and well-tolerated in a phase 1 clinical trial. However, a phase 2b clinical trial showed that treatment of GR-MD-02 did not significantly improve liver fibrosis and reduce portal hypertension (hepatic venous pressure gradient) in patients with NASH[116]. Further studies are required to evaluate these treatments for liver fibrosis.

Glucagon-like peptide-1 (GLP-1) receptor agonist: GLP-1 analogues have been shown to have the effects to reduce liver fat accumulation, liver injury, and insulin resistance in mice with fatty liver disease. Clinical trial (ClinicalTrials.gov, NCT01237119) showed that treatment of GLP-1 analogue liraglutide was well tolerated and suppressed liver fibrosis progression in patients with NASH[117]. Another trial also showed that treatment of liraglutide markedly reduced liver fat content and body weight in patients with uncontrolled type 2 diabetes[118].

Pan-caspase inhibitor: Emricasan (IDN-6556), a pan-caspase inhibitor, can decrease liver cell apoptosis and inflammation and improve portal pressure in rats with CCl4-induced cirrhosis[119]. However, a clinical trial (Clinicaltrials.gov, NCT03205345) did not show the efficacy of emaricasan against liver fibrosis, but it was safe and well-tolerated.

PPAR agonists: In rats with BDL-induced liver fibrosis, treatment of PPAR-γ agonist thiazolidinedione inhibited HSC activation and liver fibrosis by regulating fibrogenic factors, such as TGF-β1, platelet-derived growth factor, and connective tissue growth factor[120]. Farglitazar (GI262570), an agonist of peroxisome proliferator-activated receptor-gamma (PPARγ), can inhibit HSC activation.

Tropifexor: A non-bile acid FXR agonist, can potently inhibit cholestatic liver injury and fibrosis by enhancing the expression of FGF19 in the ileum and the expression of small heterodimer partner in the livers of piglets but inhibit cholesterol 7α-hydroxylase. In addition, tropifexor can increase the abundance of bile acid-biotransforming bacteria and later the amino acid composition in the intestine and decrease intestinal barrier injury in piglets with BLD[121]. Clinical trial (Clinicaltrials.gov, NCT02855164) also showed that treatment of tropifexor (10-90 μg) once daily for 12 weeks was safe and decreased the levels of alanine aminotransferase (ALT) and hepatic fat fraction (HFF) compared to baseline in a dose-dependent manner. The decrease of ALT and HFF can be sustained for up to 48 wk at high doses of tropifexor (140 μg and 200 μg once daily)[122].

Natural products or herbal medicines

Natural products or herbal medicines display diverse roles in the treatment of liver fibrosis. For example, a classical Traditional Chinese Medicine formula Yinchenhao decoction has been shown to ameliorate dimethylnitrosamine-induced liver fibrosis in rats and suppress liver cell apoptosis[123]. Another study showed that Xiaoyaosan decoction significantly reduced CCl4-induced liver fibrosis in rats by regulating both TGF-β1/Smad and AKT/FOXO3 signaling pathways[76]. The major components of these herbal medicines such as Tanshinone IIA extracted from the traditional herbal medicine Salvia miltiorrhiza display broad biological activities, such as anti-inflammatory, antioxidant, antiangiogenic, and anticancer functions[124]. Furthermore, clinical trials also illustrate that these traditional medicine formula such as Fuzheng Huayu display therapeutic effects against hepatitis-B-caused cirrhosis in patients[125].

Dietary regulation or supplementation

Consumption of polyunsaturated fatty acids: The endogenous metabolites of n-3 polyunsaturated fatty acids such as 19,20-epoxy docosapentaenoic acid show a protective effect against liver fibrosis in mouse NASH models[126]. G protein-coupled receptors can be regulated by polyunsaturated fatty acids to reduce liver inflammation and fibrosis[127]. For example, supplementation of docosahexaenoic acid, an omega-3 fatty acid, can reduce liver inflammation and prevent liver fibrosis in diet-induced liver fibrosis model via G protein-coupled receptor 120 (GPR120) signaling, also known as free fatty acid receptor 4[128].

Probiotics: Treatment with probiotic Lactobacillus rhamnosus GG can significantly decrease liver inflammation and fibrosis by reducing the production of hepatic bile acids in mice with BLD[129].

Vitamins: The serum levels of vitamin C have been shown to be negatively associated with the odds of liver fibrosis in patients with NAFLD in United States adults[130]. Another study showed that a decreased serum level of vitamin B12 is associated with an increased risk of liver fibrosis in patients with NAFLD[131]. Treatment of Vitamin D3 can alleviate liver injury and the expression of ECM proteins such as TGF-β and α-SMA in thioacetamide-induced hepatic fibrosis rat model[132]. The data from the National Health and Nutrition Examination Survey (2017-2018) also showed that levels of 25-Hydroxyvitamin D were inversely associated with liver fibrosis during NAFLD development and progression[133].

Antioxidant and anti-inflammatory agents: Supplementation of natural products with antioxidant and anti-inflammatory components can also ameliorate chronic liver disease to improve liver fibrosis and inhibit cancer development[9], such as β-sitosterol and silymarin.

Bariatric surgery

Studies have shown that bariatric surgery (BS) can provide long-term benefits for the resolution of liver fibrosis. The two most common procedures of BS are laparoscopic Roux-en-Y-gastric and laparoscopic sleeve gastrectomy. For example, one study showed that NASH was resolved in 84% of patients (95%CI: 73.1%-92.2%) at year 5 post-BS treatment, while fibrosis was decreased in samples from 70.2% of patients (95%CI: 56.6%-81.6%) compared with baseline and fibrosis was disappeared in samples from 56% of all patients (95%CI: 42.4%-69.3%)[134]. BS has been shown to induce NASH disappearance in nearly 85% (95%CI: 75.8%-92.2%) of patients and to decrease fibrosis in 33.8% of patients (95%CI: 23.6%-45.2%) with NASH at 1 year after surgery[135]. Another clinical study showed that excessive weight loss shown in patients with cirrhosis with 73% (33%–167%), 85% (33%–190%), and 73% (29%–107%) after 1, 2, and 3 years of BS[136], respectively. Among 27 patients with cirrhosis, 3 patients had significant improvement in liver function and did not need liver transplantation, whereas 2 out of 27 patients had deleterious liver function post-BS treatment[136].

Genetic intervention

Gene therapy is a critical tool for disease treatment, including liver fibrosis and cancer. Noncoding RNAs, such as miRNAs and long noncoding RNAs, small interference RNAs, and circular RNAs are important. For example, the treatment of siRNA silencing CCR2 can regulate liver immune to inhibit the infiltration of profibrotic macrophages and neutrophils in murine fibrotic livers[137]. Another study showed that circRNA ASPH regulated liver fibrosis by binding miR-139-5p by regulating neurogenic locus notch homolog protein 1 (Notch 1) expression[138].

Transplantation of stem cells

Transplantation of umbilical cord Wharton's Jelly-derived mesenchymal stem cells to rats with CCl4-induced hepatic fibrosis improved liver function, inflammation, and fibrosis via a paracrine mechanism possibly by targeting TGF-β1 signaling pathway[139]. Another study showed that transplantation of human umbilical cord blood mesenchymal stem cells substantially improved liver fibrosis in histopathological evaluation compared to that in the untreated group[140]. Infusions of hematopoietic stem cells into mice with methionine-choline-deficient diet- or CCl4-induced liver fibrosis can reduce hepatic collagen production and the expression of α-smooth muscle actin[141,142].

Overall, there are several potent preventive and therapeutic treatments for liver fibrosis, including physical activity (e.g., running), dietary change (e.g., avoid of high-fat and high-sugar diet), dietary supplementation (e.g., vitamin C), biological treatment (e.g., simtuzumab), bariatric surgery (e.g., Roux-en-Y-gastric procedure), drug (e.g., pegbelfermin), change of gut microbiota (e.g., probiotics), nanoparticles (e.g., BMS-986263), genetic regulation (e.g., non-coding RNAs), and transplantation of stem cells (e.g., hematopoietic stem cells) (Figure 3).

Figure 3
Figure 3 Treatment options for liver fibrosis. Currently, the preventive and therapeutic treatments for liver fibrosis include physical activity (e.g., running), dietary change (e.g., avoid of high-fat and high-sugar diet), dietary supplementation (e.g., vitamin C), biological treatment (e.g., simtuzumab), bariatric surgery (e.g., Roux-en-Y-gastric procedure), drugs and herb medicines (e.g., pegbelfermin), change of gut microbiota (e.g., probiotics), nanoparticles (e.g., BMS-986263), genetic regulation (e.g., non-coding RNAs), and transplantation of stem cells (e.g., hematopoietic stem cells). All cartoons in this figure were prepared using Biorender (https://biorender.com).
CLINICAL TRIALS

In this section, we first review some completed clinical trials (Clinicaltrials.gov, Table 1). These treatments including biological treatment, drugs, dietary supplementation, and infusion of stem cells.

Table 1 Completed clinical trials (Clinicaltrials.gov, accession date: March 1, 2023).
Method
Intervention
Trial number
Phase
Title
Condition
BiologicalPeginterferon alfa-2b (SCH 54031)NCT000498423Prevention of Disease Progress in Chronic Hepatitis C Patients with Liver Fibrosis (Study P02570AM2)Chronic HCV; Liver fibrosis
PegIntron (peginterferon alfa-2b; SCH 54031) REBETOL (ribavirin; SCH 18908)NCT000398713PEG-Intron Plus Rebetol Treatment of Chronic Hepatitis C Subjects Who Failed Response to Alpha-Interferon Plus Ribavirin (Study P02370)Hepatitis; HCV; Fibrosis; Liver cirrhosis
SimtuzumabNCT017074722Study of Simtuzumab in HIV and/or Hepatitis C- Infected Adults with Liver FibrosisLiver fibrosis; HCV infection; HIV
SimtuzumabNCT016728532Simtuzumab (GS-6624) in the Prevention of Progression of Liver Fibrosis in Adults with PSCPSC
Dietary regulationGK#10; PlaceboNCT01598064N/AProbiotics for Liver Cirrhosis with Portal HypertensionLiver cirrhosis; Portal hypertension
DrugAramcholNCT022795242A Clinical Trial to Evaluate the Efficacy and Safety of Two Aramchol Doses Versus Placebo in Patients With NASHFatty liver; NASH; Liver fibrosis
BI 201335NCT019097781Open Label Single Dose Phase I Trial of BI 201335 to Study Pharmacokinetics and Safety in Patients with Compensated Liver CirrhosisHCV; Liver cirrhosis
BMS-986036; PlaceboNCT034869122A Study of Experimental Medication BMS-986036 in Adults with NASH and Liver CirrhosisHepatic cirrhosis; Liver fibrosis; NAFLD; NASH
BMS-98603; Other: PlaceboNCT034868992A Study of Experimental Medication BMS-986036 in Adults with NASH and Stage 3 Liver FibrosisLiver fibrosis; NAFLD; NASH
BMS-986263 placeboNCT034207682A Study of Experimental Medication BMS-986263 in Adults with Advanced Hepatic Fibrosis After Cure of Hepatitis CHepatic cirrhosis; Liver fibrosis
Cenicriviroc placeboNCT022174752Efficacy and Safety Study of Cenicriviroc for the Treatment of NASH in Adult Participants with Liver FibrosisNASH
CRV431 placeboNCT044807102A Study of CRV431 Dosed Once Daily in NASH-induced F2 and F3 SubjectsNASH; Fibrosis; NAFLD
Fuzheng Huayu placeboNCT008540872Assess the Antifibrotic Activity of Fuzheng Huayu in Chronic Hepatitis C Patients with Hepatic FibrosisChronic HCV
GI262570 placeboNCT002447512Antifibrotic Activity of GI262570 In Chronic Hepatitis C SubjectsCirrhosis, liver
GR-MD-02 placeboNCT024629672Clinical Trial to Evaluation the Safety and Efficacy of GR-MD-02 for the Treatment of Liver Fibrosis and Resultant Portal Hypertension in Patients with Nash CirrhosisHypertension, portal
GR-MD-02; PlaceboNCT024210942Clinical Trial to Evaluate Efficacy of GR-MD-02 for Treatment of Liver Fibrosis in Patients with NASH With Advanced FibrosisNASH
IDN-6556; PlaceboNCT022306702A Study of IDN-6556 in Subjects with Liver CirrhosisLiver cirrhosis; Hepatic cirrhosis
IDN-6556; PlaceboNCT021382532A Trial of IDN-6556 in Post Orthotopic Liver Transplant for Chronic HCVLiver fibrosis; Liver cirrhosis
INT-747; Ursodeoxycholic acid; PlaceboNCT005508622Study of INT 747 in Combination with URSO in Patients with PBCLiver cirrhosis; Biliary injury
Nitazoxanide; BIDNCT036560682An Evaluation of the Safety and Efficacy of Nitazoxanide on Collagen Turnover in NASH Patients with FibrosisNASH; Fatty liver; Fibrosis; Compensated cirrhosis
Placebo obeticholic acidNCT005707652Study of INT-747 as Monotherapy in Participants with PBCLiver cirrhosis, biliary injury
SEL; SimtuzumabNCT024665162Safety, Tolerability, and Efficacy of GS-4997 Alone or in Combination with Simtuzumab in Adults with NASH and Fibrosis Stages F2-F3NASH
Simeprevir; Daclatasvir; SofosbuvirNCT023490482Study to Assess Efficacy, Safety, Tolerability and Pharmacokinetics of Simeprevir, Daclatasvir and Sofosbuvir in Treatment-naive Participants with Chronic HCV Genotype 1 InfectionHCV
Tropifexor (LJN452) CVCNCT035175402Study of Safety, Tolerability, and Efficacy of a Combination Treatment of LJN452 and CVC in Adult Patients with NASH and Liver FibrosisNASH
Peginterferon alfa-2a + Ribavirin; Peginterferon alfa-2aNCT000061643Long Term Interferon for Patients Who Did Not Clear HCV with Standard TreatmentChronic HCV; Cirrhosis; Fibrosis; Hepatic cirrhosis
OMACOR placebo oral capsuleNCT007605134Treatment of non-Alcoholic Fatty Liver Disease With n-3 Fatty AcidsNAFLD
Ceftriaxone normal salineNCT042186954Prophylactic Antibiotics in Admitted CirrhoticsCirrhosis, LIVER
Proton pump inhibitors placeboNCT031757314PPIs and Gastroesophageal Varices in Liver Cirrhosis (PPIs: Proton pump inhibitors)Liver cirrhosis; Hypertension, portal
OtherHuman fetal liver cell transplantationNCT010131941 or 2Human Fetal Liver Cell Transplantation in Chronic Liver FailureLiver cirrhosis
G-colony stimulating factor and infusion of the mobilized monocyte cellsNCT015037491Safety and Efficacy Study of Peripheral Blood Mononucleated Cells for Treatment of Liver CirrhosisLiver cirrhosis
Leukapheresis; Infusion of stem cells via image-guided scanNCT00147043N/AAdult Stem Cell Therapy in Liver InsufficiencyLiver cirrhosis
Future treatments

There is an unmet need for treatments for liver fibrosis due to the efficacy of available treatments. Some drugs with potent anti-fibrotic effects in pre-clinical models are now waiting to be further evaluated in clinical trials (Table 2). The promising preventive and therapeutic treatments for liver fibrosis, including treatment of hepatitis viral infection (e.g., Peginterferon Alfa 2a), transplantation of mesenchymal stem cells, bariatric surgery for patients with obesity and NAFLD, dietary modification (e.g., Mediterranean diet or Calorie-restricted diet).

Table 2 Recruiting and active clinical trials (Clinicaltrials.gov, accession date: March 1, 2023).
Method
Interventions
NCT number
Phases
Title
Conditions
DrugZED1227; PlaceboNCT053055992Different Doses of ZED1227 vs Placebo in NAFLDNAFLD; Fibrosis
Tropifexor; Licogliflozin; PlaceboNCT040658412Efficacy, Safety, and Tolerability of the Combination of Tropifexor & Licogliflozin and Each Monotherapy, Compared with Placebo in Adult Patients with NASH and Liver FibrosisNASH; Liver fibrosis
Tenofovir disoproxil Fumarate; PEG-Interferon alfa 2aNCT03957629N/AOptimized Treatment of Peginterferon Alfa 2a in Treatment Experienced Patients with HBV-Related Liver FibrosisHepatitis B; Fibrosis
SildenafilNCT049086574Sildenafil for Liver Fibrosis in Adolescents and Adults After Fontan OperationFibrosis
Saroglitazar magnesiumNCT050113052Saroglitazar Magnesium for the Treatment of NASH with FibrosisNASH; Fibrosis
Saroglitazar magnesiumNCT050454821Hepatic Impairment with Cirrhosis Due to Cholestatic Liver DiseaseHepatic impairment; Cirrhosis
Rivaroxaban apixabanNCT048744281Direct Oral Anticoagulants (Rivaroxaban and Apixaban) in Patients with Liver CirrhosisCirrhosis
Resmetirom; PlaceboNCT055002223A Phase 3 Study to Evaluate the Effect of Resmetirom on Clinical Outcomes in Patients with Well-compensated NASH Cirrhosis (MAESTRO-NASH-OUTCOMES)NASH; Cirrhosis
Rencofilstat; PlaceboNCT054023712A Study to Evaluate the Efficacy and Safety of Rencofilstat in Subjects with NASH and Advanced Liver FibrosisNASH; Fibrosis, Liver NAFLD
Placebo; EsomeprazoleNCT044480284Stop of Proton-pump Inhibitor Treatment in Patients with Liver Cirrhosis - a Double-blind, Placebo-controlled TrialLiver cirrhosis
PHIN-214NCT054908881Single Dose Escalation of PHIN-214 in Child-Pugh A and B Liver CirrhoticsCirrhosis; Fibrosis; Hepatic ascites
Placebo zibotentan + dapagliflozinNCT055164982Zibotentan and Dapagliflozin Combination, EvAluated in Liver Cirrhosis (ZEAL Study)Cirrhosis
L-ornithine; L-aspartateNCT057370304Effect of L-ornithine-L-aspertate (LOLA) on the Gut MicrobiomeCirrhosis
Hydronidone capsules; The placebo capsulesNCT051159423Hydronidone for the Treatment of Liver Fibrosis Associated with Chronic Viral Hepatitis B Phase 3 TrialLiver fibrosis
Growth hormoneNCT052532872/3Growth Hormone in Decompensated Liver CirrhosisCirrhosis; Fibrosis
Empagliflozin 10 MG; Placebo pillsNCT051470904Effects of Empagliflozin on Fibrosis and Cirrhosis in Chronic Hepatitis BNAFLD; Cirrhosis; Fibrosis
Cotadutide; PlaceboNCT053649312/3A Study to Evaluate the Safety and Efficacy of Cotadutide Given by Subcutaneous Injection in Adult Participants with Non-cirrhotic Non-alcoholic Steatohepatitis with FibrosisNon-cirrhotic NASH with Fibrosis
Candesartan; RamiprilNCT037709363Effect of Some Drugs on Liver FibrosisLiver fibrosis
Branched-chain amino acid; PlaceboNCT036332794Treatment of Sarcopenia Improves the Muscle Mass and Muscle Strength of Patients with Liver Cirrhosis-Child CLiver cirrhosis
BMS-986263; PlaceboNCT042673932Safety and Effectiveness of BMS-986263 in Adults with Compensated Cirrhosis (Liver Disease) From Nonalcoholic Steatohepatitis (NASH)NASH
AZD4831; PlaceboNCT056387372A Study in Participants with Non-cirrhotic NASH With FibrosisNon-cirrhotic NASH with fibrosis
Atorvastatin; PlaceboNCT050288292Safety and Efficacy of Atorvastatin v. Placebo on HCC RiskLiver fibrosis; Cirrhosis
Dietary supplementLeucine enriched essential amino acid; Balanced amino acid supplement (BAA)NCT03208868N/ALeucine-Enriched Essential Amino Acid Mixture to Reverse Muscle Loss in CirrhosisCirrhosis
Hydroxy methyl butyrate; Balanced Amino AcidsNCT05166499N/AHMB Enriched Amino Acids to Reverse Muscle Loss in CirrhosisCirrhosis
BiologicalUmbilical cord-derived mesenchymal stem cell comprehensive treatmentNCT039454872Mesenchymal Stem Cells Treatment for Decompensated Liver CirrhosisDecompensated liver cirrhosis
Mesenchymal stem cellNCT032547581/2A Study of ADR-001 in Patients with Liver CirrhosisDecompensated liver cirrhosis
Human umbilical cord-derived mesenchymal stem cellsNCT052278461Human Umbilical Cord-derived Mesenchymal Stem Cells for Decompensated Cirrhosis (MSC-DLC-1)Decompensated cirrhosis
Human umbilical cord-derived mesenchymal stem cell infusionNCT053318721Umbilical Cord-derived Mesenchymal Stem Cell Infusion in the Management of Adult Liver CirrhosisLiver cirrhosis
Fecal microbiota transplantation; PlaceboNCT049325772/3Fecal Microbiota Transplantation for Liver CirrhosisCirrhosis
Cellgram-LCNCT046891523Clinical Trial to Evaluate the Efficacy and Safety of Cellgram-LC Administration in Patients with Alcoholic CirrhosisAlcoholic cirrhosis
Autologous BM MSCNCT036260901/2Mesenchymal Stem Cell Therapy for Liver CirrhosisCirrhosis
Allogeneic umbilical cord mesenchymal stem cellNCT043576001/2Umbilical Cord Mesenchymal Stem Cell for Liver Cirrhosis Patient Caused by Hepatitis BCirrhosis
OtherWeight lossNCT05104541N/AImpact of Weight Loss in Cirrhosis with Obesity and MAFLDLiver cirrhosis
Lifestyle therapy bariatric surgeryNCT03472157N/AA Randomized Controlled Study Evaluating Bariatric Surgery as a Treatment for Severe NASH With Advanced Liver Fibrosis in Non-severe Obese PatientsSurgery; Obesity; NASH; Cirrhosis
Indo mediterranean diet calorie restricted dietNCT05073588N/AEffect of Indo-Mediterranean Diet on Hepatic Steatosis and Fibrosis in NAFLD ChildrenNAFLD

Furthermore, deliver system can be applied to increase the efficiency of anti-fibrotic treatments. For example, BMS-986263, a lipid nanoparticle, has been applied to deliver small interfering RNA to degrade mRNA of heat shock protein 47, a key collagen chaperone involved in the pathogenesis of fibrosis. Treatment of MS-986263 in patients with HCV infection and sustained virologic response improved the Ishak score, the histology activity index score for levels of liver fibrosis[143]. Many other types of nanoparticles have been applied to treat liver fibrosis or its causing chronic liver disease, such as Fibroblast growth factor 2 conjugated superparamagnetic iron oxide nanoparticles[144], cerium oxide nanoparticles[145], and silymarin-conjugated gold nanoparticles[146].

CONCLUSION

Liver fibrosis accompanies the progression of chronic liver diseases independent of their etiologies. The initiation and progression of liver fibrosis are mainly driven by liver inflammation and hepatocyte or cholangiocyte injury and damage, resulting in the activation of HSCs and their differentiation into ECM protein-producing myofibroblasts. Thus, current therapeutic options for liver fibrosis are to prevent the initial causing factors for liver inflammation, hepatocyte cell death and oxidative stress. Unfortunately, the reverse of liver fibrosis is slowly and frequently impossible for advanced fibrosis or cirrhosis. Liver transplantation is the only therapeutic option for the late stage of liver cirrhosis and cancer. To avoid the life-threatening stage of advanced liver fibrosis and cirrhosis, anti-fibrotic treatments including biological, medicines, dietary change, and behavior prevention are needed. Currently, promising treatments for liver fibrosis are still the preventive strategies, such as treatment of hepatitis viral infection (e.g., Peginterferon Alfa 2a), inhibition of the progression of MAFLD and obesity (e.g., bariatric surgery), dietary modification (e.g., Mediterranean diet or Calorie-restricted diet). In addition, nano-delivery systems have been applied to improve the treatment efficacy and specifically deliver the treatments. Pre-clinical and clinical evaluations for new treatments of liver fibrosis are required while we still lack currently effective strategies for liver fibrosis treatment. The treatment efficacy can be evaluated by histological staining methods, imaging methods, and serum biomarkers, as well as fibrosis scoring systems, such as FIB-4, APRI, and NFS. Although many anti-fibrotic candidate agents have shown robust effects in experimental animal models, their anti-fibrotic effects in clinical trials are less clear. The development of patient-derived organoid models for liver fibrosis may advance the development of compounds with anti-fibrotic properties in the future. In addition, new delivery systems can improve the efficacy of potent treatments and reduce the side effects of therapy. Meanwhile, additional clinical studies are required to confirm the efficacy and safety of treatments.

Footnotes

Provenance and peer review: Invited article; Externally peer reviewed.

Peer-review model: Single blind

Specialty type: Gastroenterology and hepatology

Country/Territory of origin: United States

Peer-review report’s scientific quality classification

Grade A (Excellent): 0

Grade B (Very good): B

Grade C (Good): 0

Grade D (Fair): D

Grade E (Poor): 0

P-Reviewer: Boteon YL, Brazil; Su SB, China S-Editor: Ma YJ L-Editor: A P-Editor: Yuan YY

References
1.  Higashi T, Friedman SL, Hoshida Y. Hepatic stellate cells as key target in liver fibrosis. Adv Drug Deliv Rev. 2017;121:27-42.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 530]  [Cited by in F6Publishing: 806]  [Article Influence: 115.1]  [Reference Citation Analysis (0)]
2.  Kisseleva T, Brenner D. Molecular and cellular mechanisms of liver fibrosis and its regression. Nat Rev Gastroenterol Hepatol. 2021;18:151-166.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 274]  [Cited by in F6Publishing: 663]  [Article Influence: 221.0]  [Reference Citation Analysis (0)]
3.  Mederacke I, Hsu CC, Troeger JS, Huebener P, Mu X, Dapito DH, Pradere JP, Schwabe RF. Fate tracing reveals hepatic stellate cells as dominant contributors to liver fibrosis independent of its aetiology. Nat Commun. 2013;4:2823.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 750]  [Cited by in F6Publishing: 910]  [Article Influence: 91.0]  [Reference Citation Analysis (0)]
4.  Tsuchida T, Friedman SL. Mechanisms of hepatic stellate cell activation. Nat Rev Gastroenterol Hepatol. 2017;14:397-411.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1221]  [Cited by in F6Publishing: 1570]  [Article Influence: 224.3]  [Reference Citation Analysis (0)]
5.  Hou W, Syn WK. Role of Metabolism in Hepatic Stellate Cell Activation and Fibrogenesis. Front Cell Dev Biol. 2018;6:150.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 42]  [Cited by in F6Publishing: 54]  [Article Influence: 9.0]  [Reference Citation Analysis (0)]
6.  De Smet V, Eysackers N, Merens V, Kazemzadeh Dastjerd M, Halder G, Verhulst S, Mannaerts I, van Grunsven LA. Initiation of hepatic stellate cell activation extends into chronic liver disease. Cell Death Dis. 2021;12:1110.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 4]  [Cited by in F6Publishing: 4]  [Article Influence: 1.3]  [Reference Citation Analysis (0)]
7.  Koyama Y, Brenner DA. Liver inflammation and fibrosis. J Clin Invest. 2017;127:55-64.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 548]  [Cited by in F6Publishing: 737]  [Article Influence: 105.3]  [Reference Citation Analysis (0)]
8.  Del Campo JA, Gallego P, Grande L. Role of inflammatory response in liver diseases: Therapeutic strategies. World J Hepatol. 2018;10:1-7.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 192]  [Cited by in F6Publishing: 208]  [Article Influence: 34.7]  [Reference Citation Analysis (4)]
9.  Zhang CY, Liu S, Yang M. Antioxidant and anti-inflammatory agents in chronic liver diseases: Molecular mechanisms and therapy. World J Hepatol. 2023;15:180-200.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 3]  [Cited by in F6Publishing: 12]  [Article Influence: 12.0]  [Reference Citation Analysis (0)]
10.  Ramachandran P, Matchett KP, Dobie R, Wilson-Kanamori JR, Henderson NC. Single-cell technologies in hepatology: new insights into liver biology and disease pathogenesis. Nat Rev Gastroenterol Hepatol. 2020;17:457-472.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 80]  [Cited by in F6Publishing: 124]  [Article Influence: 31.0]  [Reference Citation Analysis (0)]
11.  Yu P, Zhang X, Liu N, Tang L, Peng C, Chen X. Pyroptosis: mechanisms and diseases. Signal Transduct Target Ther. 2021;6:128.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 286]  [Cited by in F6Publishing: 760]  [Article Influence: 253.3]  [Reference Citation Analysis (0)]
12.  Kovacs SB, Miao EA. Gasdermins: Effectors of Pyroptosis. Trends Cell Biol. 2017;27:673-684.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 706]  [Cited by in F6Publishing: 754]  [Article Influence: 107.7]  [Reference Citation Analysis (0)]
13.  Drummer C 4th, Saaoud F, Jhala NC, Cueto R, Sun Y, Xu K, Shao Y, Lu Y, Shen H, Yang L, Zhou Y, Yu J, Wu S, Snyder NW, Hu W, Zhuo J', Zhong Y, Jiang X, Wang H, Yang X. Caspase-11 promotes high-fat diet-induced NAFLD by increasing glycolysis, OXPHOS, and pyroptosis in macrophages. Front Immunol. 2023;14:1113883.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 8]  [Cited by in F6Publishing: 9]  [Article Influence: 9.0]  [Reference Citation Analysis (0)]
14.  Liu Y, Kong X, You Y, Xiang L, Zhang Y, Wu R, Zhou L, Duan L. S100A8-Mediated NLRP3 Inflammasome-Dependent Pyroptosis in Macrophages Facilitates Liver Fibrosis Progression. Cells. 2022;11.  [PubMed]  [DOI]  [Cited in This Article: ]  [Reference Citation Analysis (0)]
15.  Gao H, Zhong Y, Zhou L, Lin S, Hou X, Ding Z, Li Y, Yao Q, Cao H, Zou X, Chen D, Bai X, Xiao G. Kindlin-2 inhibits TNF/NF-κB-Caspase 8 pathway in hepatocytes to maintain liver development and function. Elife. 2023;12.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in F6Publishing: 5]  [Reference Citation Analysis (0)]
16.  Chen K, Lin T, Yao W, Chen X, Xiong X, Huang Z. Adipocytes-derived exosomal miR-122 promotes non-alcoholic fat liver disease progression via targeting Sirt1. Gastroenterol Hepatol. 2022;.  [PubMed]  [DOI]  [Cited in This Article: ]  [Reference Citation Analysis (0)]
17.  Wang R, Liu F, Chen P, Li S, Gu Y, Wang L, Chen C, Yuan Y. Gomisin D alleviates liver fibrosis through targeting PDGFRβ in hepatic stellate cells. Int J Biol Macromol. 2023;235:123639.  [PubMed]  [DOI]  [Cited in This Article: ]  [Reference Citation Analysis (0)]
18.  Dhuriya YK, Sharma D. Necroptosis: a regulated inflammatory mode of cell death. J Neuroinflammation. 2018;15:199.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 314]  [Cited by in F6Publishing: 339]  [Article Influence: 56.5]  [Reference Citation Analysis (0)]
19.  Pasparakis M, Vandenabeele P. Necroptosis and its role in inflammation. Nature. 2015;517:311-320.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1257]  [Cited by in F6Publishing: 1376]  [Article Influence: 152.9]  [Reference Citation Analysis (0)]
20.  Shi H, Wang X, Li F, Gerlach BD, Yurdagul A Jr, Moore MP, Zeldin S, Zhang H, Cai B, Zheng Z, Valenti L, Tabas I. CD47-SIRPα axis blockade in NASH promotes necroptotic hepatocyte clearance by liver macrophages and decreases hepatic fibrosis. Sci Transl Med. 2022;14:eabp8309.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 19]  [Cited by in F6Publishing: 20]  [Article Influence: 10.0]  [Reference Citation Analysis (0)]
21.  Liu J, Kang R, Tang D. Signaling pathways and defense mechanisms of ferroptosis. FEBS J. 2022;289:7038-7050.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 35]  [Cited by in F6Publishing: 158]  [Article Influence: 79.0]  [Reference Citation Analysis (0)]
22.  Zhang C, Liu X, Jin S, Chen Y, Guo R. Ferroptosis in cancer therapy: a novel approach to reversing drug resistance. Mol Cancer. 2022;21:47.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 25]  [Cited by in F6Publishing: 328]  [Article Influence: 164.0]  [Reference Citation Analysis (0)]
23.  Chen X, Kang R, Kroemer G, Tang D. Ferroptosis in infection, inflammation, and immunity. J Exp Med. 2021;218.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 61]  [Cited by in F6Publishing: 249]  [Article Influence: 83.0]  [Reference Citation Analysis (0)]
24.  Zhu L, Luo S, Zhu Y, Tang S, Li C, Jin X, Wu F, Jiang H, Wu L, Xu Y. The Emerging Role of Ferroptosis in Various Chronic Liver Diseases: Opportunity or Challenge. J Inflamm Res. 2023;16:381-389.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in F6Publishing: 6]  [Reference Citation Analysis (0)]
25.  Capelletti MM, Manceau H, Puy H, Peoc'h K. Ferroptosis in Liver Diseases: An Overview. Int J Mol Sci. 2020;21.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 65]  [Cited by in F6Publishing: 167]  [Article Influence: 41.8]  [Reference Citation Analysis (0)]
26.  Abu-Halaka D, Shpaizer A, Zeigerman H, Kanner J, Tirosh O. DMF-Activated Nrf2 Ameliorates Palmitic Acid Toxicity While Potentiates Ferroptosis Mediated Cell Death: Protective Role of the NO-Donor S-Nitroso-N-Acetylcysteine. Antioxidants (Basel). 2023;12.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in F6Publishing: 3]  [Reference Citation Analysis (0)]
27.  Wang Y, Zhao M, Zhao L, Geng Y, Li G, Chen L, Yu J, Yuan H, Zhang H, Yun H, Yuan Y, Wang G, Feng J, Xu L, Wang S, Hou C, Yang G, Zhang N, Lu W, Zhang X. HBx-Induced HSPA8 Stimulates HBV Replication and Suppresses Ferroptosis to Support Liver Cancer Progression. Cancer Res. 2023;83:1048-1061.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in F6Publishing: 15]  [Reference Citation Analysis (0)]
28.  Zheng Y, Zhao T, Wang J, Jiang R, Huang J, Li W. Curcumol alleviates liver fibrosis through inducing autophagy and ferroptosis in hepatic stellate cells. FASEB J. 2022;36:e22665.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1]  [Cited by in F6Publishing: 8]  [Article Influence: 4.0]  [Reference Citation Analysis (0)]
29.  Li L, Zhu Z. Pharmacological modulation of ferroptosis as a therapeutic target for liver fibrosis. Front Pharmacol. 2022;13:1071844.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in F6Publishing: 4]  [Reference Citation Analysis (0)]
30.  Jung S, Jeong H, Yu SW. Autophagy as a decisive process for cell death. Exp Mol Med. 2020;52:921-930.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 79]  [Cited by in F6Publishing: 142]  [Article Influence: 35.5]  [Reference Citation Analysis (0)]
31.  Yang M, Qi X, Li N, Kaifi JT, Chen S, Wheeler AA, Kimchi ET, Ericsson AC, Scott Rector R, Staveley-O'Carroll KF, Li G. Western diet contributes to the pathogenesis of non-alcoholic steatohepatitis in male mice via remodeling gut microbiota and increasing production of 2-oleoylglycerol. Nat Commun. 2023;14:228.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in F6Publishing: 17]  [Reference Citation Analysis (0)]
32.  Yang M, Kimchi ET, Staveley-O'Carroll KF, Li G. Astaxanthin Prevents Diet-Induced NASH Progression by Shaping Intrahepatic Immunity. Int J Mol Sci. 2021;22.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 5]  [Cited by in F6Publishing: 5]  [Article Influence: 1.7]  [Reference Citation Analysis (0)]
33.  Kennedy L, Meadows V, Sybenga A, Demieville J, Chen L, Hargrove L, Ekser B, Dar W, Ceci L, Kundu D, Kyritsi K, Pham L, Zhou T, Glaser S, Meng F, Alpini G, Francis H. Mast Cells Promote Nonalcoholic Fatty Liver Disease Phenotypes and Microvesicular Steatosis in Mice Fed a Western Diet. Hepatology. 2021;74:164-182.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 12]  [Cited by in F6Publishing: 12]  [Article Influence: 4.0]  [Reference Citation Analysis (0)]
34.  Zhang CY, Liu S, Yang M. Regulatory T cells and their associated factors in hepatocellular carcinoma development and therapy. World J Gastroenterol. 2022;28:3346-3358.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in CrossRef: 6]  [Cited by in F6Publishing: 5]  [Article Influence: 2.5]  [Reference Citation Analysis (0)]
35.  Woestemeier A, Scognamiglio P, Zhao Y, Wagner J, Muscate F, Casar C, Siracusa F, Cortesi F, Agalioti T, Müller S, Sagebiel A, Konczalla L, Wahib R, Karstens KF, Giannou AD, Duprée A, Wolter S, Wong MN, Mühlig AK, Bielecka AA, Bansal V, Zhang T, Mann O, Puelles VG, Huber TB, Lohse AW, Izbicki JR, Palm NW, Bonn S, Huber S, Gagliani N. Multicytokine-producing CD4+ T cells characterize the livers of patients with NASH. JCI Insight. 2023;8.  [PubMed]  [DOI]  [Cited in This Article: ]  [Reference Citation Analysis (0)]
36.  Koda Y, Teratani T, Chu PS, Hagihara Y, Mikami Y, Harada Y, Tsujikawa H, Miyamoto K, Suzuki T, Taniki N, Sujino T, Sakamoto M, Kanai T, Nakamoto N. CD8(+) tissue-resident memory T cells promote liver fibrosis resolution by inducing apoptosis of hepatic stellate cells. Nat Commun. 2021;12:4474.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 22]  [Cited by in F6Publishing: 77]  [Article Influence: 25.7]  [Reference Citation Analysis (0)]
37.  He Y, Hwang S, Ahmed YA, Feng D, Li N, Ribeiro M, Lafdil F, Kisseleva T, Szabo G, Gao B. Immunopathobiology and therapeutic targets related to cytokines in liver diseases. Cell Mol Immunol. 2021;18:18-37.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 23]  [Cited by in F6Publishing: 58]  [Article Influence: 14.5]  [Reference Citation Analysis (0)]
38.  Wan Y, Li X, Slevin E, Harrison K, Li T, Zhang Y, Klaunig JE, Wu C, Shetty AK, Dong XC, Meng F. Endothelial dysfunction in pathological processes of chronic liver disease during aging. FASEB J. 2022;36:e22125.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 3]  [Cited by in F6Publishing: 11]  [Article Influence: 3.7]  [Reference Citation Analysis (0)]
39.  Cheng F, Li Y, Feng L, Li S. Hepatic stellate cell activation and hepatic fibrosis induced by ischemia/reperfusion injury. Transplant Proc. 2008;40:2167-2170.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 22]  [Cited by in F6Publishing: 23]  [Article Influence: 1.4]  [Reference Citation Analysis (0)]
40.  Elpek GÖ. Angiogenesis and liver fibrosis. World J Hepatol. 2015;7:377-391.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 101]  [Cited by in F6Publishing: 102]  [Article Influence: 11.3]  [Reference Citation Analysis (0)]
41.  Ratziu V, Rinella ME, Neuschwander-Tetri BA, Lawitz E, Denham D, Kayali Z, Sheikh A, Kowdley KV, Desta T, Elkhashab M, DeGrauw J, Goodwin B, Ahmad A, Adda N. EDP-305 in patients with NASH: A phase II double-blind placebo-controlled dose-ranging study. J Hepatol. 2022;76:506-517.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 19]  [Cited by in F6Publishing: 46]  [Article Influence: 23.0]  [Reference Citation Analysis (0)]
42.  Ahmad A, Adda N. Assessment of drug-drug interaction potential with EDP-305, a farnesoid X receptor agonist, in healthy subjects. Clin Transl Sci. 2022;15:2146-2158.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1]  [Reference Citation Analysis (0)]
43.  Patel K, Harrison SA, Elkhashab M, Trotter JF, Herring R, Rojter SE, Kayali Z, Wong VW, Greenbloom S, Jayakumar S, Shiffman ML, Freilich B, Lawitz EJ, Gane EJ, Harting E, Xu J, Billin AN, Chung C, Djedjos CS, Subramanian GM, Myers RP, Middleton MS, Rinella M, Noureddin M. Cilofexor, a Nonsteroidal FXR Agonist, in Patients With Noncirrhotic NASH: A Phase 2 Randomized Controlled Trial. Hepatology. 2020;72:58-71.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 146]  [Cited by in F6Publishing: 194]  [Article Influence: 48.5]  [Reference Citation Analysis (0)]
44.  Trauner M, Bowlus CL, Gulamhusein A, Hameed B, Caldwell SH, Shiffman ML, Landis C, Muir AJ, Billin A, Xu J, Liu X, Lu X, Chung C, Myers RP, Kowdley KV. Safety and sustained efficacy of the farnesoid X receptor (FXR) agonist cilofexor over a 96-week open-label extension in patients with PSC. Clin Gastroenterol Hepatol. 2022;.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1]  [Cited by in F6Publishing: 10]  [Article Influence: 5.0]  [Reference Citation Analysis (0)]
45.  Harrison SA, Bashir MR, Lee KJ, Shim-Lopez J, Lee J, Wagner B, Smith ND, Chen HC, Lawitz EJ. A structurally optimized FXR agonist, MET409, reduced liver fat content over 12 weeks in patients with non-alcoholic steatohepatitis. J Hepatol. 2021;75:25-33.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 40]  [Cited by in F6Publishing: 71]  [Article Influence: 23.7]  [Reference Citation Analysis (0)]
46.  Jiang H, Chen HC, Lafata KJ, Bashir MR. Week 4 Liver Fat Reduction on MRI as an Early Predictor of Treatment Response in Participants with Nonalcoholic Steatohepatitis. Radiology. 2021;300:361-368.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 8]  [Cited by in F6Publishing: 8]  [Article Influence: 2.7]  [Reference Citation Analysis (0)]
47.  Xie G, Jiang R, Wang X, Liu P, Zhao A, Wu Y, Huang F, Liu Z, Rajani C, Zheng X, Qiu J, Zhang X, Zhao S, Bian H, Gao X, Sun B, Jia W. Conjugated secondary 12α-hydroxylated bile acids promote liver fibrogenesis. EBioMedicine. 2021;66:103290.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 47]  [Cited by in F6Publishing: 42]  [Article Influence: 14.0]  [Reference Citation Analysis (0)]
48.  Thapaliya S, Wree A, Povero D, Inzaugarat ME, Berk M, Dixon L, Papouchado BG, Feldstein AE. Caspase 3 inactivation protects against hepatic cell death and ameliorates fibrogenesis in a diet-induced NASH model. Dig Dis Sci. 2014;59:1197-1206.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 70]  [Cited by in F6Publishing: 76]  [Article Influence: 7.6]  [Reference Citation Analysis (0)]
49.  Shiffman M, Freilich B, Vuppalanchi R, Watt K, Chan JL, Spada A, Hagerty DT, Schiff E. Randomised clinical trial: emricasan versus placebo significantly decreases ALT and caspase 3/7 activation in subjects with non-alcoholic fatty liver disease. Aliment Pharmacol Ther. 2019;49:64-73.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 61]  [Cited by in F6Publishing: 54]  [Article Influence: 10.8]  [Reference Citation Analysis (0)]
50.  Puengel T, Lefere S, Hundertmark J, Kohlhepp M, Penners C, Van de Velde F, Lapauw B, Hoorens A, Devisscher L, Geerts A, Boehm S, Zhao Q, Krupinski J, Charles ED, Zinker B, Tacke F. Combined Therapy with a CCR2/CCR5 Antagonist and FGF21 Analogue Synergizes in Ameliorating Steatohepatitis and Fibrosis. Int J Mol Sci. 2022;23.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 14]  [Cited by in F6Publishing: 22]  [Article Influence: 11.0]  [Reference Citation Analysis (0)]
51.  Bartneck M, Koppe C, Fech V, Warzecha KT, Kohlhepp M, Huss S, Weiskirchen R, Trautwein C, Luedde T, Tacke F. Roles of CCR2 and CCR5 for Hepatic Macrophage Polarization in Mice With Liver Parenchymal Cell-Specific NEMO Deletion. Cell Mol Gastroenterol Hepatol. 2021;11:327-347.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 8]  [Cited by in F6Publishing: 7]  [Article Influence: 1.8]  [Reference Citation Analysis (0)]
52.  Wu X, Qian L, Zhao H, Lei W, Liu Y, Xu X, Li J, Yang Z, Wang D, Zhang Y, Tang R, Yang Y, Tian Y. CXCL12/CXCR4: An amazing challenge and opportunity in the fight against fibrosis. Ageing Res Rev. 2023;83:101809.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in F6Publishing: 11]  [Reference Citation Analysis (0)]
53.  Ni Y, Zhuge F, Ni L, Nagata N, Yamashita T, Mukaida N, Kaneko S, Ota T, Nagashimada M. CX3CL1/CX3CR1 interaction protects against lipotoxicity-induced nonalcoholic steatohepatitis by regulating macrophage migration and M1/M2 status. Metabolism. 2022;136:155272.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 3]  [Cited by in F6Publishing: 17]  [Article Influence: 8.5]  [Reference Citation Analysis (0)]
54.  Liu XB, Liu H, Liu J, Cheung AKL, Zheng MZ, Cheng JL, Liu QS, Lo CM, Chen ZW, Man K. Cytomegalovirus Latency Exacerbated Small-for-size Liver Graft Injury Through Activation of CCL19/CCR7 in Hepatic Stellate Cells. Transplantation. 2022;106:519-530.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1]  [Cited by in F6Publishing: 1]  [Article Influence: 0.3]  [Reference Citation Analysis (0)]
55.  Van Loy T, De Jonghe S, Castermans K, Dheedene W, Stoop R, Verschuren L, Versele M, Chaltin P, Luttun A, Schols D. Stimulation of the atypical chemokine receptor 3 (ACKR3) by a small-molecule agonist attenuates fibrosis in a preclinical liver but not lung injury model. Cell Mol Life Sci. 2022;79:293.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1]  [Reference Citation Analysis (0)]
56.  Bhat N, Esteghamat F, Chaube BK, Gunawardhana K, Mani M, Thames C, Jain D, Ginsberg HN, Fernandes-Hernando C, Mani A. TCF7L2 transcriptionally regulates Fgf15 to maintain bile acid and lipid homeostasis through gut-liver crosstalk. FASEB J. 2022;36:e22185.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1]  [Cited by in F6Publishing: 1]  [Article Influence: 0.5]  [Reference Citation Analysis (0)]
57.  Matye DJ, Wang H, Luo W, Sharp RR, Chen C, Gu L, Jones KL, Ding WX, Friedman JE, Li T. Combined ASBT Inhibitor and FGF15 Treatment Improves Therapeutic Efficacy in Experimental Nonalcoholic Steatohepatitis. Cell Mol Gastroenterol Hepatol. 2021;12:1001-1019.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 18]  [Cited by in F6Publishing: 17]  [Article Influence: 5.7]  [Reference Citation Analysis (0)]
58.  Schumacher JD, Guo GL. Pharmacologic Modulation of Bile Acid-FXR-FGF15/FGF19 Pathway for the Treatment of Nonalcoholic Steatohepatitis. Handb Exp Pharmacol. 2019;256:325-357.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 30]  [Cited by in F6Publishing: 33]  [Article Influence: 6.6]  [Reference Citation Analysis (0)]
59.  Nobili V, Alisi A, Mosca A, Della Corte C, Veraldi S, De Vito R, De Stefanis C, D'Oria V, Jahnel J, Zohrer E, Scorletti E, Byrne CD. Hepatic farnesoid X receptor protein level and circulating fibroblast growth factor 19 concentration in children with NAFLD. Liver Int. 2018;38:342-349.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 27]  [Cited by in F6Publishing: 30]  [Article Influence: 5.0]  [Reference Citation Analysis (0)]
60.  Tsai YT, Li CY, Huang YH, Chang TS, Lin CY, Chuang CH, Wang CY, Anuraga G, Chang TH, Shih TC, Lin ZY, Chen YL, Chung I, Lee KH, Chang CC, Sung SY, Yang KH, Tsui WL, Yap CV, Wu MH. Galectin-1 orchestrates an inflammatory tumor-stroma crosstalk in hepatoma by enhancing TNFR1 protein stability and signaling in carcinoma-associated fibroblasts. Oncogene. 2022;41:3011-3023.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1]  [Cited by in F6Publishing: 13]  [Article Influence: 6.5]  [Reference Citation Analysis (0)]
61.  Cervantes-Alvarez E, Limon-de la Rosa N, Vilatoba M, Pérez-Monter C, Hurtado-Gomez S, Martinez-Cabrera C, Argemi J, Alatorre-Arenas E, Yarza-Regalado S, Tejeda-Dominguez F, Lizardo-Thiebaud MJ, Mendez-Guerrero O, Gamboa-Dominguez A, Aguilar-Salinas CA, Huang CA, Kershenobich D, Bataller R, Torre A, Navarro-Alvarez N. Galectin-3 is overexpressed in advanced cirrhosis and predicts post-liver transplant infectious complications. Liver Int. 2022;42:2260-2273.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 2]  [Cited by in F6Publishing: 2]  [Article Influence: 1.0]  [Reference Citation Analysis (0)]
62.  Chen W, Yang A, Jia J, Popov YV, Schuppan D, You H. Lysyl Oxidase (LOX) Family Members: Rationale and Their Potential as Therapeutic Targets for Liver Fibrosis. Hepatology. 2020;72:729-741.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 62]  [Cited by in F6Publishing: 94]  [Article Influence: 23.5]  [Reference Citation Analysis (0)]
63.  Jiang S, Zhang Y, Zheng JH, Li X, Yao YL, Wu YL, Song SZ, Sun P, Nan JX, Lian LH. Potentiation of hepatic stellate cell activation by extracellular ATP is dependent on P2X7R-mediated NLRP3 inflammasome activation. Pharmacol Res. 2017;117:82-93.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 66]  [Cited by in F6Publishing: 69]  [Article Influence: 8.6]  [Reference Citation Analysis (0)]
64.  Gan C, Cai Q, Tang C, Gao J. Inflammasomes and Pyroptosis of Liver Cells in Liver Fibrosis. Front Immunol. 2022;13:896473.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1]  [Cited by in F6Publishing: 37]  [Article Influence: 18.5]  [Reference Citation Analysis (0)]
65.  Wree A, Eguchi A, McGeough MD, Pena CA, Johnson CD, Canbay A, Hoffman HM, Feldstein AE. NLRP3 inflammasome activation results in hepatocyte pyroptosis, liver inflammation, and fibrosis in mice. Hepatology. 2014;59:898-910.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 680]  [Cited by in F6Publishing: 669]  [Article Influence: 66.9]  [Reference Citation Analysis (0)]
66.  Gaul S, Leszczynska A, Alegre F, Kaufmann B, Johnson CD, Adams LA, Wree A, Damm G, Seehofer D, Calvente CJ, Povero D, Kisseleva T, Eguchi A, McGeough MD, Hoffman HM, Pelegrin P, Laufs U, Feldstein AE. Hepatocyte pyroptosis and release of inflammasome particles induce stellate cell activation and liver fibrosis. J Hepatol. 2021;74:156-167.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 260]  [Cited by in F6Publishing: 250]  [Article Influence: 83.3]  [Reference Citation Analysis (0)]
67.  Zhu Y, Zhao H, Lu J, Lin K, Ni J, Wu G, Tang H. Caspase-11-Mediated Hepatocytic Pyroptosis Promotes the Progression of Nonalcoholic Steatohepatitis. Cell Mol Gastroenterol Hepatol. 2021;12:653-664.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 20]  [Cited by in F6Publishing: 10]  [Article Influence: 3.3]  [Reference Citation Analysis (0)]
68.  de Souza Basso B, Haute GV, Ortega-Ribera M, Luft C, Antunes GL, Bastos MS, Carlessi LP, Levorse VG, Cassel E, Donadio MVF, Santarém ER, Gracia-Sancho J, Rodrigues de Oliveira J. Methoxyeugenol deactivates hepatic stellate cells and attenuates liver fibrosis and inflammation through a PPAR-ɣ and NF-kB mechanism. J Ethnopharmacol. 2021;280:114433.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 20]  [Cited by in F6Publishing: 29]  [Article Influence: 9.7]  [Reference Citation Analysis (0)]
69.  Pawlak M, Lefebvre P, Staels B. Molecular mechanism of PPARα action and its impact on lipid metabolism, inflammation and fibrosis in non-alcoholic fatty liver disease. J Hepatol. 2015;62:720-733.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 793]  [Cited by in F6Publishing: 902]  [Article Influence: 100.2]  [Reference Citation Analysis (0)]
70.  Chen J, Montagner A, Tan NS, Wahli W. Insights into the Role of PPARβ/δ in NAFLD. Int J Mol Sci. 2018;19.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 28]  [Cited by in F6Publishing: 30]  [Article Influence: 5.0]  [Reference Citation Analysis (0)]
71.  Francque S, Verrijken A, Caron S, Prawitt J, Paumelle R, Derudas B, Lefebvre P, Taskinen MR, Van Hul W, Mertens I, Hubens G, Van Marck E, Michielsen P, Van Gaal L, Staels B. PPARα gene expression correlates with severity and histological treatment response in patients with non-alcoholic steatohepatitis. J Hepatol. 2015;63:164-173.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 214]  [Cited by in F6Publishing: 240]  [Article Influence: 26.7]  [Reference Citation Analysis (0)]
72.  Zhang Y, Cui Y, Wang XL, Shang X, Qi ZG, Xue J, Zhao X, Deng M, Xie ML. PPARα/γ agonists and antagonists differently affect hepatic lipid metabolism, oxidative stress and inflammatory cytokine production in steatohepatitic rats. Cytokine. 2015;75:127-135.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 29]  [Cited by in F6Publishing: 31]  [Article Influence: 3.4]  [Reference Citation Analysis (0)]
73.  Zarei M, Barroso E, Palomer X, Dai J, Rada P, Quesada-López T, Escolà-Gil JC, Cedó L, Zali MR, Molaei M, Dabiri R, Vázquez S, Pujol E, Valverde ÁM, Villarroya F, Liu Y, Wahli W, Vázquez-Carrera M. Hepatic regulation of VLDL receptor by PPARβ/δ and FGF21 modulates non-alcoholic fatty liver disease. Mol Metab. 2018;8:117-131.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 52]  [Cited by in F6Publishing: 70]  [Article Influence: 10.0]  [Reference Citation Analysis (0)]
74.  Boyer-Diaz Z, Aristu-Zabalza P, Andrés-Rozas M, Robert C, Ortega-Ribera M, Fernández-Iglesias A, Broqua P, Junien JL, Wettstein G, Bosch J, Gracia-Sancho J. Pan-PPAR agonist lanifibranor improves portal hypertension and hepatic fibrosis in experimental advanced chronic liver disease. J Hepatol. 2021;74:1188-1199.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 39]  [Cited by in F6Publishing: 56]  [Article Influence: 18.7]  [Reference Citation Analysis (0)]
75.  Xu F, Liu C, Zhou D, Zhang L. TGF-β/SMAD Pathway and Its Regulation in Hepatic Fibrosis. J Histochem Cytochem. 2016;64:157-167.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 326]  [Cited by in F6Publishing: 469]  [Article Influence: 58.6]  [Reference Citation Analysis (0)]
76.  Zhou Y, Wu R, Cai FF, Zhou WJ, Lu YY, Zhang H, Chen QL, Su SB. Xiaoyaosan decoction alleviated rat liver fibrosis via the TGFβ/Smad and Akt/FoxO3 signaling pathways based on network pharmacology analysis. J Ethnopharmacol. 2021;264:113021.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 34]  [Cited by in F6Publishing: 25]  [Article Influence: 8.3]  [Reference Citation Analysis (0)]
77.  Liu QW, Ying YM, Zhou JX, Zhang WJ, Liu ZX, Jia BB, Gu HC, Zhao CY, Guan XH, Deng KY, Xin HB. Human amniotic mesenchymal stem cells-derived IGFBP-3, DKK-3, and DKK-1 attenuate liver fibrosis through inhibiting hepatic stellate cell activation by blocking Wnt/β-catenin signaling pathway in mice. Stem Cell Res Ther. 2022;13:224.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in F6Publishing: 14]  [Reference Citation Analysis (0)]
78.  El-Ashmawy NE, Al-Ashmawy GM, Fakher HE, Khedr NF. The role of WNT/β-catenin signaling pathway and glutamine metabolism in the pathogenesis of CCl(4)-induced liver fibrosis: Repositioning of niclosamide and concerns about lithium. Cytokine. 2020;136:155250.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 8]  [Cited by in F6Publishing: 10]  [Article Influence: 2.5]  [Reference Citation Analysis (0)]
79.  Rao S, Xiang J, Huang J, Zhang S, Zhang M, Sun H, Li J. PRC1 promotes GLI1-dependent osteopontin expression in association with the Wnt/β-catenin signaling pathway and aggravates liver fibrosis. Cell Biosci. 2019;9:100.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 5]  [Cited by in F6Publishing: 7]  [Article Influence: 1.4]  [Reference Citation Analysis (0)]
80.  Gao Y, Fan S, Zhao P, Li H, Cai C, Li X, Zhou Y, Huang M, Bi H. β-catenin/TCF4 inhibitors ICG-001 and LF3 alleviate BDL-induced liver fibrosis by suppressing LECT2 signaling. Chem Biol Interact. 2023;371:110350.  [PubMed]  [DOI]  [Cited in This Article: ]  [Reference Citation Analysis (0)]
81.  Du K, Maeso-Díaz R, Oh SH, Wang E, Chen T, Pan C, Xiang K, Dutta RK, Wang XF, Chi JT, Diehl AM. Targeting YAP-mediated hepatic stellate cell death susceptibility and senescence for treatment of liver fibrosis. Hepatology. 2023;.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 10]  [Cited by in F6Publishing: 7]  [Article Influence: 7.0]  [Reference Citation Analysis (0)]
82.  Yang J, Tang X, Liang Z, Chen M, Sun L. Taurocholic Acid Promotes Hepatic Stellate Cell Activation via S1PR2/p38 MAPK/YAP Signaling under Cholestatic Conditions. Clin Mol Hepatol. 2023;.  [PubMed]  [DOI]  [Cited in This Article: ]  [Reference Citation Analysis (0)]
83.  Lo RC, Kim H. Histopathological evaluation of liver fibrosis and cirrhosis regression. Clin Mol Hepatol. 2017;23:302-307.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 61]  [Cited by in F6Publishing: 83]  [Article Influence: 11.9]  [Reference Citation Analysis (0)]
84.  Lurie Y, Webb M, Cytter-Kuint R, Shteingart S, Lederkremer GZ. Non-invasive diagnosis of liver fibrosis and cirrhosis. World J Gastroenterol. 2015;21:11567-11583.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in CrossRef: 189]  [Cited by in F6Publishing: 207]  [Article Influence: 23.0]  [Reference Citation Analysis (0)]
85.  Kumar R, Teo EK, How CH, Wong TY, Ang TL. A practical clinical approach to liver fibrosis. Singapore Med J. 2018;59:628-633.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 9]  [Cited by in F6Publishing: 9]  [Article Influence: 1.8]  [Reference Citation Analysis (0)]
86.  Taru MG, Neamti L, Taru V, Procopciuc LM, Procopet B, Lupsor-Platon M. How to Identify Advanced Fibrosis in Adult Patients with Non-Alcoholic Fatty Liver Disease (NAFLD) and Non-Alcoholic Steatohepatitis (NASH) Using Ultrasound Elastography-A Review of the Literature and Proposed Multistep Approach. Diagnostics (Basel). 2023;13.  [PubMed]  [DOI]  [Cited in This Article: ]  [Reference Citation Analysis (0)]
87.  Schambeck JPL, Forte GC, Gonçalves LM, Stuker G, Kotlinski JBF, Tramontin G, Altmayer S, Watte G, Hochhegger B. Diagnostic accuracy of magnetic resonance elastography and point-shear wave elastography for significant hepatic fibrosis screening: Systematic review and meta-analysis. PLoS One. 2023;18:e0271572.  [PubMed]  [DOI]  [Cited in This Article: ]  [Reference Citation Analysis (0)]
88.  Li S, Sun X, Chen M, Ying Z, Wan Y, Pi L, Ren B, Cao Q. Liver Fibrosis Conventional and Molecular Imaging Diagnosis Update. J Liver. 2019;8.  [PubMed]  [DOI]  [Cited in This Article: ]
89.  Decharatanachart P, Chaiteerakij R, Tiyarattanachai T, Treeprasertsuk S. Application of artificial intelligence in non-alcoholic fatty liver disease and liver fibrosis: a systematic review and meta-analysis. Therap Adv Gastroenterol. 2021;14:17562848211062807.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 5]  [Cited by in F6Publishing: 13]  [Article Influence: 4.3]  [Reference Citation Analysis (0)]
90.  Ahmed Y, Hussein RS, Basha TA, Khalifa AM, Ibrahim AS, Abdelmoaty AS, Abdella HM, Fahmy AS. Detecting liver fibrosis using a machine learning-based approach to the quantification of the heart-induced deformation in tagged MR images. NMR Biomed. 2020;33:e4215.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 10]  [Cited by in F6Publishing: 11]  [Article Influence: 2.8]  [Reference Citation Analysis (0)]
91.  He L, Li H, Dudley JA, Maloney TC, Brady SL, Somasundaram E, Trout AT, Dillman JR. Machine Learning Prediction of Liver Stiffness Using Clinical and T2-Weighted MRI Radiomic Data. AJR Am J Roentgenol. 2019;213:592-601.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 26]  [Cited by in F6Publishing: 34]  [Article Influence: 6.8]  [Reference Citation Analysis (0)]
92.  Tadokoro T, Morishita A, Masaki T. Diagnosis and Therapeutic Management of Liver Fibrosis by MicroRNA. Int J Mol Sci. 2021;22.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 8]  [Cited by in F6Publishing: 10]  [Article Influence: 3.3]  [Reference Citation Analysis (0)]
93.  Kaya A, Barutcu S, Gulsen MT. Evaluation of fibrosis with noninvasive biochemical tests in chronic viral hepatitis B. Hepatol Forum. 2023;4:25-29.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in F6Publishing: 2]  [Reference Citation Analysis (0)]
94.  Wang J, Qin T, Sun J, Li S, Cao L, Lu X. Non-invasive methods to evaluate liver fibrosis in patients with non-alcoholic fatty liver disease. Front Physiol. 2022;13:1046497.  [PubMed]  [DOI]  [Cited in This Article: ]  [Reference Citation Analysis (0)]
95.  Qi RB, Wu ZH. Association between COVID-19 and chronic liver disease: Mechanism, diagnosis, damage, and treatment. World J Virol. 2023;12:22-29.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in CrossRef: 1]  [Cited by in F6Publishing: 1]  [Article Influence: 1.0]  [Reference Citation Analysis (0)]
96.  Sang C, Yan H, Chan WK, Zhu X, Sun T, Chang X, Xia M, Sun X, Hu X, Gao X, Jia W, Bian H, Chen T, Xie G. Diagnosis of Fibrosis Using Blood Markers and Logistic Regression in Southeast Asian Patients With Non-alcoholic Fatty Liver Disease. Front Med (Lausanne). 2021;8:637652.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 5]  [Cited by in F6Publishing: 5]  [Article Influence: 1.7]  [Reference Citation Analysis (0)]
97.  Gong L, Zhang Y, Yang Y, Yan Q, Ren J, Luo J, Tiu YC, Fang X, Liu B, Lam RHW, Lam KO, Lee AW, Guan XY. Inhibition of lysyl oxidase-like 2 overcomes adhesion-dependent drug resistance in the collagen-enriched liver cancer microenvironment. Hepatol Commun. 2022;6:3194-3211.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1]  [Cited by in F6Publishing: 1]  [Article Influence: 0.5]  [Reference Citation Analysis (0)]
98.  Harrison SA, Abdelmalek MF, Caldwell S, Shiffman ML, Diehl AM, Ghalib R, Lawitz EJ, Rockey DC, Schall RA, Jia C, McColgan BJ, McHutchison JG, Subramanian GM, Myers RP, Younossi Z, Ratziu V, Muir AJ, Afdhal NH, Goodman Z, Bosch J, Sanyal AJ; GS-US-321-0105 and GS-US-321-0106 Investigators. Simtuzumab Is Ineffective for Patients With Bridging Fibrosis or Compensated Cirrhosis Caused by Nonalcoholic Steatohepatitis. Gastroenterology. 2018;155:1140-1153.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 188]  [Cited by in F6Publishing: 225]  [Article Influence: 37.5]  [Reference Citation Analysis (0)]
99.  Meissner EG, McLaughlin M, Matthews L, Gharib AM, Wood BJ, Levy E, Sinkus R, Virtaneva K, Sturdevant D, Martens C, Porcella SF, Goodman ZD, Kanwar B, Myers RP, Subramanian M, Hadigan C, Masur H, Kleiner DE, Heller T, Kottilil S, Kovacs JA, Morse CG. Simtuzumab treatment of advanced liver fibrosis in HIV and HCV-infected adults: results of a 6-month open-label safety trial. Liver Int. 2016;36:1783-1792.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 65]  [Cited by in F6Publishing: 72]  [Article Influence: 9.0]  [Reference Citation Analysis (0)]
100.  Ratziu V, de Guevara L, Safadi R, Poordad F, Fuster F, Flores-Figueroa J, Arrese M, Fracanzani AL, Ben Bashat D, Lackner K, Gorfine T, Kadosh S, Oren R, Halperin M, Hayardeny L, Loomba R, Friedman S; ARREST investigator study group, Sanyal AJ. Aramchol in patients with nonalcoholic steatohepatitis: a randomized, double-blind, placebo-controlled phase 2b trial. Nat Med. 2021;27:1825-1835.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 36]  [Cited by in F6Publishing: 79]  [Article Influence: 26.3]  [Reference Citation Analysis (0)]
101.  Wu J, Gießmann T, Lang B, Elgadi M, Huang F. Investigation of the effect of food and omeprazole on the relative bioavailability of a single oral dose of 240 mg faldaprevir, a selective inhibitor of HCV NS3/4 protease, in an open-label, randomized, three-way cross-over trial in healthy participants. J Pharm Pharmacol. 2016;68:459-466.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 2]  [Cited by in F6Publishing: 2]  [Article Influence: 0.3]  [Reference Citation Analysis (0)]
102.  Ahmed S, Ullah N, Parveen S, Javed I, Jalil NAC, Murtey MD, Sheikh IS, Khan S, Ojha SC, Chen K. Effect of Silymarin as an Adjunct Therapy in Combination with Sofosbuvir and Ribavirin in Hepatitis C Patients: A Miniature Clinical Trial. Oxid Med Cell Longev. 2022;2022:9199190.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1]  [Cited by in F6Publishing: 2]  [Article Influence: 1.0]  [Reference Citation Analysis (0)]
103.  Chen S, Zhou J, Wu X, Meng T, Wang B, Liu H, Wang T, Zhao X, Kong Y, Wu S, Ou X, Jia J, Sun Y, You H. Comparison of fibrosis regression of entecavir alone or combined with pegylated interferon alpha2a in patients with chronic hepatitis B. Hepatol Int. 2021;15:611-620.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 2]  [Cited by in F6Publishing: 5]  [Article Influence: 1.7]  [Reference Citation Analysis (0)]
104.  Mieli-Vergani G, Bansal S, Daniel JF, Kansu A, Kelly D, Martin C, Tizzard S, Wirth S, Zhou J, Vergani D. Peginterferon Alfa-2a (40KD) Plus Lamivudine or Entecavir in Children With Immune-Tolerant Chronic Hepatitis B. J Pediatr Gastroenterol Nutr. 2021;73:156-160.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 3]  [Cited by in F6Publishing: 1]  [Article Influence: 0.3]  [Reference Citation Analysis (0)]
105.  Said EM, Abdulaziz BA, El Kassas M, El Attar IH, Emadeldeen M, Abd-Elsalam SM. High success rates for the use of sofosbuvir/ombitasvir/paritaprevir/ritonavir + ribavirin and sofosbuvir/simeprevir/daclatasvir + ribavirin in retreatment of chronic hepatitis C infection after unsuccessful sofosbuvir/daclatasvir therapy: a real-life experience. Arch Virol. 2020;165:1633-1639.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 9]  [Cited by in F6Publishing: 8]  [Article Influence: 2.0]  [Reference Citation Analysis (0)]
106.  Gupta N, Manirambona L, Shumbusho F, Kabihizi J, Murangwa A, Serumondo J, Makuza JD, Nsanzimana S, Muvunyi CM, Mukabatsinda C, Musabeyezu E, Camus G, Grant PM, Kateera F. Safety and efficacy of sofosbuvir-velpatasvir-voxilaprevir for re-treatment of chronic hepatitis C virus infection in patients with previous direct-acting antiviral treatment failure in Rwanda (SHARED-3): a single-arm trial. Lancet Gastroenterol Hepatol. 2022;7:542-551.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 4]  [Cited by in F6Publishing: 3]  [Article Influence: 1.5]  [Reference Citation Analysis (0)]
107.  Ueno T, Osawa M, Shiozaki T, Green M, Garimella T. Exposure-Response Analysis for Efficacy of Daclatasvir, Asunaprevir, and Beclabuvir Combinations in HCV-Infected Patients. Clin Pharmacol Drug Dev. 2019;8:903-913.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1]  [Cited by in F6Publishing: 1]  [Article Influence: 0.2]  [Reference Citation Analysis (0)]
108.  Ratziu V, Sanyal A, Harrison SA, Wong VW, Francque S, Goodman Z, Aithal GP, Kowdley KV, Seyedkazemi S, Fischer L, Loomba R, Abdelmalek MF, Tacke F. Cenicriviroc Treatment for Adults With Nonalcoholic Steatohepatitis and Fibrosis: Final Analysis of the Phase 2b CENTAUR Study. Hepatology. 2020;72:892-905.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 149]  [Cited by in F6Publishing: 200]  [Article Influence: 50.0]  [Reference Citation Analysis (0)]
109.  Anstee QM, Neuschwander-Tetri BA, Wong VW, Abdelmalek MF, Younossi ZM, Yuan J, Pecoraro ML, Seyedkazemi S, Fischer L, Bedossa P, Goodman Z, Alkhouri N, Tacke F, Sanyal A. Cenicriviroc for the treatment of liver fibrosis in adults with nonalcoholic steatohepatitis: AURORA Phase 3 study design. Contemp Clin Trials. 2020;89:105922.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 88]  [Cited by in F6Publishing: 86]  [Article Influence: 21.5]  [Reference Citation Analysis (0)]
110.  Barba Bernal R, Ferrigno B, Medina Morales E, Castro CM, Goyes D, Trivedi H, Patwardhan VR, Bonder A. Management of Primary Biliary Cholangitis: Current Treatment and Future Perspectives. Turk J Gastroenterol. 2023;34:89-100.  [PubMed]  [DOI]  [Cited in This Article: ]  [Reference Citation Analysis (0)]
111.  Kuo J, Bobardt M, Chatterji U, Mayo PR, Trepanier DJ, Foster RT, Gallay P, Ure DR. A Pan-Cyclophilin Inhibitor, CRV431, Decreases Fibrosis and Tumor Development in Chronic Liver Disease Models. J Pharmacol Exp Ther. 2019;371:231-241.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 17]  [Cited by in F6Publishing: 22]  [Article Influence: 4.4]  [Reference Citation Analysis (0)]
112.  Simón Serrano S, Grönberg A, Longato L, Rombouts K, Kuo J, Gregory M, Moss S, Elmér E, Mazza G, Gallay P, Pinzani M, Hansson MJ, Massoumi R. Evaluation of NV556, a Novel Cyclophilin Inhibitor, as a Potential Antifibrotic Compound for Liver Fibrosis. Cells. 2019;8.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 7]  [Cited by in F6Publishing: 10]  [Article Influence: 2.0]  [Reference Citation Analysis (0)]
113.  Sanyal A, Charles ED, Neuschwander-Tetri BA, Loomba R, Harrison SA, Abdelmalek MF, Lawitz EJ, Halegoua-DeMarzio D, Kundu S, Noviello S, Luo Y, Christian R. Pegbelfermin (BMS-986036), a PEGylated fibroblast growth factor 21 analogue, in patients with non-alcoholic steatohepatitis: a randomised, double-blind, placebo-controlled, phase 2a trial. Lancet. 2019;392:2705-2717.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 282]  [Cited by in F6Publishing: 340]  [Article Influence: 68.0]  [Reference Citation Analysis (0)]
114.  Younossi ZM, Ratziu V, Loomba R, Rinella M, Anstee QM, Goodman Z, Bedossa P, Geier A, Beckebaum S, Newsome PN, Sheridan D, Sheikh MY, Trotter J, Knapple W, Lawitz E, Abdelmalek MF, Kowdley KV, Montano-Loza AJ, Boursier J, Mathurin P, Bugianesi E, Mazzella G, Olveira A, Cortez-Pinto H, Graupera I, Orr D, Gluud LL, Dufour JF, Shapiro D, Campagna J, Zaru L, MacConell L, Shringarpure R, Harrison S, Sanyal AJ; REGENERATE Study Investigators. Obeticholic acid for the treatment of non-alcoholic steatohepatitis: interim analysis from a multicentre, randomised, placebo-controlled phase 3 trial. Lancet. 2019;394:2184-2196.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 610]  [Cited by in F6Publishing: 716]  [Article Influence: 143.2]  [Reference Citation Analysis (0)]
115.  Zetterberg FR, MacKinnon A, Brimert T, Gravelle L, Johnsson RE, Kahl-Knutson B, Leffler H, Nilsson UJ, Pedersen A, Peterson K, Roper JA, Schambye H, Slack RJ, Tantawi S. Discovery and Optimization of the First Highly Effective and Orally Available Galectin-3 Inhibitors for Treatment of Fibrotic Disease. J Med Chem. 2022;65:12626-12638.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in F6Publishing: 21]  [Reference Citation Analysis (0)]
116.  Chalasani N, Abdelmalek MF, Garcia-Tsao G, Vuppalanchi R, Alkhouri N, Rinella M, Noureddin M, Pyko M, Shiffman M, Sanyal A, Allgood A, Shlevin H, Horton R, Zomer E, Irish W, Goodman Z, Harrison SA, Traber PG; Belapectin (GR-MD-02) Study Investigators. Effects of Belapectin, an Inhibitor of Galectin-3, in Patients With Nonalcoholic Steatohepatitis With Cirrhosis and Portal Hypertension. Gastroenterology. 2020;158:1334-1345.e5.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 138]  [Cited by in F6Publishing: 183]  [Article Influence: 45.8]  [Reference Citation Analysis (0)]
117.  Armstrong MJ, Gaunt P, Aithal GP, Barton D, Hull D, Parker R, Hazlehurst JM, Guo K; LEAN trial team, Abouda G, Aldersley MA, Stocken D, Gough SC, Tomlinson JW, Brown RM, Hübscher SG, Newsome PN. Liraglutide safety and efficacy in patients with non-alcoholic steatohepatitis (LEAN): a multicentre, double-blind, randomised, placebo-controlled phase 2 study. Lancet. 2016;387:679-690.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1100]  [Cited by in F6Publishing: 1175]  [Article Influence: 146.9]  [Reference Citation Analysis (1)]
118.  Petit JM, Cercueil JP, Loffroy R, Denimal D, Bouillet B, Fourmont C, Chevallier O, Duvillard L, Vergès B. Effect of Liraglutide Therapy on Liver Fat Content in Patients With Inadequately Controlled Type 2 Diabetes: The Lira-NAFLD Study. J Clin Endocrinol Metab. 2017;102:407-415.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 53]  [Cited by in F6Publishing: 71]  [Article Influence: 10.1]  [Reference Citation Analysis (0)]
119.  Frenette C, Kayali Z, Mena E, Mantry PS, Lucas KJ, Neff G, Rodriguez M, Thuluvath PJ, Weinberg E, Bhandari BR, Robinson J, Wedick N, Chan JL, Hagerty DT, Kowdley KV; IDN-6556-17 Study Investigators. Emricasan to prevent new decompensation in patients with NASH-related decompensated cirrhosis. J Hepatol. 2021;74:274-282.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 20]  [Cited by in F6Publishing: 29]  [Article Influence: 9.7]  [Reference Citation Analysis (0)]
120.  Alatas FS, Matsuura T, Pudjiadi AH, Wijaya S, Taguchi T. Peroxisome Proliferator-Activated Receptor Gamma Agonist Attenuates Liver Fibrosis by Several Fibrogenic Pathways in an Animal Model of Cholestatic Fibrosis. Pediatr Gastroenterol Hepatol Nutr. 2020;23:346-355.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 6]  [Cited by in F6Publishing: 11]  [Article Influence: 2.8]  [Reference Citation Analysis (0)]
121.  Xiao Y, Wang Y, Liu Y, Wang W, Tian X, Chen S, Lu Y, Du J, Cai W. A nonbile acid farnesoid X receptor agonist tropifexor potently inhibits cholestatic liver injury and fibrosis by modulating the gut-liver axis. Liver Int. 2021;41:2117-2131.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 7]  [Cited by in F6Publishing: 22]  [Article Influence: 7.3]  [Reference Citation Analysis (0)]
122.  Sanyal AJ, Lopez P, Lawitz EJ, Lucas KJ, Loeffler J, Kim W, Goh GBB, Huang JF, Serra C, Andreone P, Chen YC, Hsia SH, Ratziu V, Aizenberg D, Tobita H, Sheikh AM, Vierling JM, Kim YJ, Hyogo H, Tai D, Goodman Z, Schaefer F, Carbarns IRI, Lamle S, Martic M, Naoumov NV, Brass CA. Tropifexor for nonalcoholic steatohepatitis: an adaptive, randomized, placebo-controlled phase 2a/b trial. Nat Med. 2023;29:392-400.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 18]  [Cited by in F6Publishing: 30]  [Article Influence: 30.0]  [Reference Citation Analysis (0)]
123.  Cai FF, Bian YQ, Wu R, Sun Y, Chen XL, Yang MD, Zhang QR, Hu Y, Sun MY, Su SB. Yinchenhao decoction suppresses rat liver fibrosis involved in an apoptosis regulation mechanism based on network pharmacology and transcriptomic analysis. Biomed Pharmacother. 2019;114:108863.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 49]  [Cited by in F6Publishing: 64]  [Article Influence: 12.8]  [Reference Citation Analysis (0)]
124.  Shi MJ, Dong BS, Yang WN, Su SB, Zhang H. Preventive and therapeutic role of Tanshinone ⅡA in hepatology. Biomed Pharmacother. 2019;112:108676.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 21]  [Cited by in F6Publishing: 19]  [Article Influence: 3.8]  [Reference Citation Analysis (0)]
125.  Song YN, Sun JJ, Lu YY, Xu LM, Gao YQ, Zhang W, Wang XS, Xue DY, Zheng QS, Su SB. Therapeutic efficacy of fuzheng-huayu tablet based traditional chinese medicine syndrome differentiation on hepatitis-B-caused cirrhosis: a multicenter double-blind randomized controlled trail. Evid Based Complement Alternat Med. 2013;2013:709305.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 19]  [Cited by in F6Publishing: 26]  [Article Influence: 2.4]  [Reference Citation Analysis (0)]
126.  Aoki H, Isobe Y, Yoshida M, Kang JX, Maekawa M, Arita M. Enzymatically-epoxidized docosahexaenoic acid, 19,20-EpDPE, suppresses hepatic crown-like structure formation and nonalcoholic steatohepatitis fibrosis through GPR120. Biochim Biophys Acta Mol Cell Biol Lipids. 2023;1868:159275.  [PubMed]  [DOI]  [Cited in This Article: ]  [Reference Citation Analysis (0)]
127.  Yang M, Zhang CY. G protein-coupled receptors as potential targets for nonalcoholic fatty liver disease treatment. World J Gastroenterol. 2021;27:677-691.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in CrossRef: 17]  [Cited by in F6Publishing: 22]  [Article Influence: 7.3]  [Reference Citation Analysis (0)]
128.  Nakamoto K, Shimada K, Harada S, Morimoto Y, Hirasawa A, Tokuyama S. DHA supplementation prevent the progression of NASH via GPR120 signaling. Eur J Pharmacol. 2018;820:31-38.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 23]  [Cited by in F6Publishing: 23]  [Article Influence: 3.3]  [Reference Citation Analysis (0)]
129.  Liu Y, Chen K, Li F, Gu Z, Liu Q, He L, Shao T, Song Q, Zhu F, Zhang L, Jiang M, Zhou Y, Barve S, Zhang X, McClain CJ, Feng W. Probiotic Lactobacillus rhamnosus GG Prevents Liver Fibrosis Through Inhibiting Hepatic Bile Acid Synthesis and Enhancing Bile Acid Excretion in Mice. Hepatology. 2020;71:2050-2066.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 158]  [Cited by in F6Publishing: 165]  [Article Influence: 41.3]  [Reference Citation Analysis (0)]
130.  Zhao Y, Li H. Association of serum vitamin C with liver fibrosis in adults with nonalcoholic fatty liver disease. Scand J Gastroenterol. 2022;57:872-877.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1]  [Cited by in F6Publishing: 5]  [Article Influence: 2.5]  [Reference Citation Analysis (0)]
131.  Li L, Huang Q, Yang L, Zhang R, Gao L, Han X, Ji L, Zou X. The Association between Non-Alcoholic Fatty Liver Disease (NAFLD) and Advanced Fibrosis with Serological Vitamin B12 Markers: Results from the NHANES 1999-2004. Nutrients. 2022;14.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1]  [Cited by in F6Publishing: 21]  [Article Influence: 10.5]  [Reference Citation Analysis (0)]
132.  Megahed A, Gadalla H, Abdelhamid FM, Almehmadi SJ, Khan AA, Albukhari TA, Risha EF. Vitamin D Ameliorates the Hepatic Oxidative Damage and Fibrotic Effect Caused by Thioacetamide in Rats. Biomedicines. 2023;11.  [PubMed]  [DOI]  [Cited in This Article: ]  [Reference Citation Analysis (0)]
133.  Ciardullo S, Muraca E, Cannistraci R, Perra S, Lattuada G, Perseghin G. Low 25 (OH) vitamin D levels are associated with increased prevalence of nonalcoholic fatty liver disease and significant liver fibrosis. Diabetes Metab Res Rev. 2023;e3628.  [PubMed]  [DOI]  [Cited in This Article: ]  [Reference Citation Analysis (0)]
134.  Lassailly G, Caiazzo R, Ntandja-Wandji LC, Gnemmi V, Baud G, Verkindt H, Ningarhari M, Louvet A, Leteurtre E, Raverdy V, Dharancy S, Pattou F, Mathurin P. Bariatric Surgery Provides Long-term Resolution of Nonalcoholic Steatohepatitis and Regression of Fibrosis. Gastroenterology. 2020;159:1290-1301.e5.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 204]  [Cited by in F6Publishing: 271]  [Article Influence: 67.8]  [Reference Citation Analysis (0)]
135.  Lassailly G, Caiazzo R, Buob D, Pigeyre M, Verkindt H, Labreuche J, Raverdy V, Leteurtre E, Dharancy S, Louvet A, Romon M, Duhamel A, Pattou F, Mathurin P. Bariatric Surgery Reduces Features of Nonalcoholic Steatohepatitis in Morbidly Obese Patients. Gastroenterology. 2015;149:379-88; quiz e15.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 510]  [Cited by in F6Publishing: 494]  [Article Influence: 54.9]  [Reference Citation Analysis (2)]
136.  Mehdorn AS, Moulla Y, Mehdorn M, Dietrich A, Schönfels W, Becker T, Braun F, Beckmann JH, Linecker M. Bariatric surgery in liver cirrhosis. Front Surg. 2022;9:986297.  [PubMed]  [DOI]  [Cited in This Article: ]  [Reference Citation Analysis (0)]
137.  Tian T, Zhao C, Li S, Huang Z, Guo Y, Dai W, Bai R, Tang C, Lin Y, Gao J. Liver-Targeted Delivery of Small Interfering RNA of C-C Chemokine Receptor 2 with Tetrahedral Framework Nucleic Acid Attenuates Liver Cirrhosis. ACS Appl Mater Interfaces. 2023;15:10492-10505.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1]  [Cited by in F6Publishing: 5]  [Article Influence: 5.0]  [Reference Citation Analysis (0)]
138.  Meng H, Jiang L, Jia P, Niu R, Bu F, Zhu Y, Pan X, Li J, Liu J, Zhang Y, Huang C, Lv X. Inhibition of circular RNA ASPH reduces the proliferation and promotes the apoptosis of hepatic stellate cells in hepatic fibrosis. Biochem Pharmacol. 2023;210:115451.  [PubMed]  [DOI]  [Cited in This Article: ]  [Reference Citation Analysis (0)]
139.  Li M, Yan L, Yuan X, Deng M, Xi Y, Zhu D. Umbilical Cord Wharton's Jelly-Derived Mesenchymal Stem Cells Inhibit the TGF-β1 Pathway in Hepatic Fibrosis Rats Through a Paracrine Regulation Process. Clin Lab. 2023;69.  [PubMed]  [DOI]  [Cited in This Article: ]  [Reference Citation Analysis (0)]
140.  Abou Rayia DM, Ashour DS, Abo Safia HS, Abdel Ghafar MT, Amer RS, Saad AE. Human umbilical cord blood mesenchymal stem cells as a potential therapy for schistosomal hepatic fibrosis: an experimental study. Pathog Glob Health. 2023;117:190-202.  [PubMed]  [DOI]  [Cited in This Article: ]  [Reference Citation Analysis (0)]
141.  King A, Houlihan DD, Kavanagh D, Haldar D, Luu N, Owen A, Suresh S, Than NN, Reynolds G, Penny J, Sumption H, Ramachandran P, Henderson NC, Kalia N, Frampton J, Adams DH, Newsome PN. Sphingosine-1-Phosphate Prevents Egress of Hematopoietic Stem Cells From Liver to Reduce Fibrosis. Gastroenterology. 2017;153:233-248.e16.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 34]  [Cited by in F6Publishing: 39]  [Article Influence: 5.6]  [Reference Citation Analysis (0)]
142.  Berardis S, Dwisthi Sattwika P, Najimi M, Sokal EM. Use of mesenchymal stem cells to treat liver fibrosis: current situation and future prospects. World J Gastroenterol. 2015;21:742-758.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in CrossRef: 90]  [Cited by in F6Publishing: 96]  [Article Influence: 10.7]  [Reference Citation Analysis (0)]
143.  Lawitz EJ, Shevell DE, Tirucherai GS, Du S, Chen W, Kavita U, Coste A, Poordad F, Karsdal M, Nielsen M, Goodman Z, Charles ED. BMS-986263 in patients with advanced hepatic fibrosis: 36-week results from a randomized, placebo-controlled phase 2 trial. Hepatology. 2022;75:912-923.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 11]  [Cited by in F6Publishing: 31]  [Article Influence: 15.5]  [Reference Citation Analysis (0)]
144.  Kurniawan DW, Booijink R, Pater L, Wols I, Vrynas A, Storm G, Prakash J, Bansal R. Fibroblast growth factor 2 conjugated superparamagnetic iron oxide nanoparticles (FGF2-SPIONs) ameliorate hepatic stellate cells activation in vitro and acute liver injury in vivo. J Control Release. 2020;328:640-652.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 15]  [Cited by in F6Publishing: 27]  [Article Influence: 6.8]  [Reference Citation Analysis (0)]
145.  Oró D, Yudina T, Fernández-Varo G, Casals E, Reichenbach V, Casals G, González de la Presa B, Sandalinas S, Carvajal S, Puntes V, Jiménez W. Cerium oxide nanoparticles reduce steatosis, portal hypertension and display anti-inflammatory properties in rats with liver fibrosis. J Hepatol. 2016;64:691-698.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 129]  [Cited by in F6Publishing: 136]  [Article Influence: 17.0]  [Reference Citation Analysis (0)]
146.  Abdullah AS, El Sayed IET, El-Torgoman AMA, Alghamdi NA, Ullah S, Wageh S, Kamel MA. Preparation and Characterization of Silymarin-Conjugated Gold Nanoparticles with Enhanced Anti-Fibrotic Therapeutic Effects against Hepatic Fibrosis in Rats: Role of MicroRNAs as Molecular Targets. Biomedicines. 2021;9.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 5]  [Cited by in F6Publishing: 15]  [Article Influence: 5.0]  [Reference Citation Analysis (0)]