v
Search
Advanced

Publications > Journals > Journal of Clinical and Translational Hepatology> Article Full Text

  • OPEN ACCESS

Exploring the Therapeutic Potential of TGF-β Inhibitors for Liver Fibrosis: Targeting Multiple Signaling Pathways

  • Wanchun Zhu1,#,
  • Yu Cui1,#,
  • Jiahao Qiu1,
  • Xin Zhang1,2,
  • Yueqiu Gao3,
  • Zhi Shang1,*  and
  • Lingying Huang2,* 
 Author information 

Abstract

Liver fibrosis is a pathological process resulting from various chronic liver injuries that lead to the formation of liver fibrous scars. It can further progress to cirrhosis and even liver cancer. Currently, there are no effective drugs specifically approved for the treatment of liver fibrosis; etiological therapy remains the main treatment strategy. Therefore, it is necessary to develop anti-fibrotic drugs targeting different pathways involved in liver fibrosis. Transforming growth factor-beta (TGF-β) is a key driver of fibrosis, and targeting TGF-β can effectively reduce liver fibrosis. In this review, we discussed the anti-liver fibrosis effects of TGF-β inhibitors through different signaling pathways, including the application of certain active ingredients from Traditional Chinese Medicine.

Keywords

Transforming growth factor-beta, Fibrosis, Liver cirrhosis, Antifibrotic agents, Enzyme inhibitors, Molecular targeted therapy

Introduction

Liver fibrosis is usually caused by various chronic liver injuries,1 including viral hepatitis, alcoholic liver disease, non-alcoholic fatty liver disease, and autoimmune liver disease.2 The pathological features of liver fibrosis include fibroblast proliferation and differentiation, inflammatory cell infiltration, abnormal extracellular matrix (ECM), and fibrous scar formation in the liver. Hepatic stellate cell (HSC) activation is the main cause of fibrous scarring in liver fibrosis.3

The central feature of liver fibrosis is the accumulation of excess collagen and ECM components in liver tissue.2 This abnormal ECM deposition leads to severe disruption of liver structure and function.4 Although potentially reversible, uncontrolled fibrosis can progress to end-stage cirrhosis and hepatocellular carcinoma (HCC).5 Due to the complex progression of liver fibrosis, no satisfactory anti-fibrotic drugs have been developed to date.

The transforming growth factor-beta (TGF-β) superfamily includes TGF-βs and bone morphogenetic proteins (BMPs).3,6 These proteins are involved in cell growth, differentiation, immunomodulation, and tissue repair.4 There are three TGF-β homologs: TGF-β1, TGF-β2, and TGF-β3, all of which bind to TGF-β receptor II (TGF-βRII) as dimers. TGF-βRII then recruits and activates the type I receptor kinase (TGF-βRI, also known as activin receptor-like kinase (ALK) 5). After activation, TGF-βRI phosphorylates its substrates, thereby activating downstream signal transduction pathways and regulating various cellular activities.6–8

The liver comprises parenchymal cells (hepatocytes) and diverse non-parenchymal cells, including liver sinusoidal endothelial cells, HSCs, Kupffer cells, intrahepatic lymphocytes, and liver-resident dendritic cells. All non-parenchymal cell populations constitutively synthesize TGF-β, whereas hepatocytes primarily absorb and store this cytokine.9 In hepatocytes, TGF-β primarily inhibits proliferation, induces apoptosis, promotes epithelial-mesenchymal transition (EMT), and contributes to fibrogenesis.10,11 Research indicates that even small amounts of TGF-β can strongly suppress hepatocyte proliferation.12 TGF-β further promotes liver fibrosis by increasing ECM component synthesis and inhibiting ECM degradation. It regulates the balance of matrix metalloproteinases (MMPs) and their inhibitors.

TGF-β can not only induce hepatocyte apoptosis but also regulate fibroblast proliferation. Collectively, these factors promote the formation and progression of fibrosis.13 TGF-β can stimulate the activation of HSCs, which are normally quiescent. Activated HSCs transform into myofibroblasts and begin secreting large amounts of collagen and other ECM components.14 Therefore, activated HSCs are mainly responsible for the excessive synthesis and deposition of ECM in the liver interstitium, leading to fibrosis.15 TGF-β also regulates HSC autophagy, senescence, metabolism, reactive oxygen species production, epigenetic modifications, and circadian genes, all of which amplify the fibrogenic response.16

TGF-β promotes fibrosis by regulating immune cells through complex signaling pathways that recruit and activate multiple immune cell types, particularly macrophages, T cells, and neutrophils. These immune cells are critical for initiating and sustaining the fibrotic response. TGF-β induces macrophages to secrete cytokines such as PDGF and TGF-β itself, which, in turn, activate HSCs and enhance collagen deposition.11 TGF-β can also polarize macrophages to an M2 phenotype, which, while suppressing excessive inflammation, secretes fibrogenic mediators (including TGF-β itself) that further activate HSCs.11 Concurrently, TGF-β reprograms effector T cells (Th1/Th17) into regulatory T cells, establishing a positive feedback loop that perpetuates fibrosis.17 Therefore, TGF-β is considered the main factor accelerating liver fibrosis and even regulating liver carcinogenesis.18

Despite its pro-fibrotic actions, TGF-β is indispensable for peripheral immune tolerance: Tgfb1 knockout mice succumb to multifocal inflammation within three weeks.19 Deficiency of TGF-β1 in humans manifests as severe inflammatory bowel disease and encephalopathic syndromes.20 As a pivotal orchestrator of hepatic immunity, TGF-β fine-tunes immune cell responses to sustain equilibrium between tolerance and effector activation. It is crucial for immune homeostasis through its dual regulation of immunosuppressive regulatory T cells and pro-inflammatory Th17 cell differentiation.17 Accordingly, pan-ALK5 inhibitors such as LY2157299 produced dose-limiting aortic-valve thickening and anemia in first-in-human studies.21 Therefore, systemic blockade therapy may trigger autoimmunity, colitis, and impaired tumor surveillance.

TGF-β is a central regulator in chronic liver disease, involved in all stages of progression from initial liver injury through inflammation and fibrosis to cirrhosis and HCC. Upon liver injury, active TGF-β ligands appear in the liver and induce downstream signaling across all cell types. During hepatocarcinogenesis, TGF-β acts as a tumor suppressor in the early stages but later promotes tumor progression.22

Due to the key role of TGF-β in liver fibrosis, it has become a potential therapeutic target. Researchers are working to develop strategies targeting the TGF-β signaling pathway, including TGF-β receptor antagonists and TGF-β neutralizing antibodies. These strategies aim to slow or reverse the fibrotic process, thereby improving the prognosis for patients with liver disease.23 This review describes the current literature on the use of TGF-β inhibitors for treating liver fibrosis.

TGF-β signaling pathways

TGF-β itself (TGF-β1, -β2, and -β3) signals through specific receptor complexes composed of two different proteins, TGFβRII and TGFβRI, which are expressed in all cell types.8,24 These receptors are transmembrane glycoproteins with kinase activity and can transmit signals across the cell membrane.25 TGF-β exists initially in an inactive precursor form and binds to TGFβRII after enzymatic activation. TGF-βRII acts as a tyrosine kinase receptor; upon binding to TGF-β, it recruits and promotes the phosphorylation of TGF-βRI.8 Once activated, TGFβ-RI phosphorylates its substrates, namely the SMAD family proteins. Specifically, TGF-βRI phosphorylates SMAD2 and SMAD3 in the case of TGF-β, or SMAD1, SMAD5, and SMAD8 in the case of BMPs.26 Phosphorylation of TGFβRI further activates downstream signaling pathways, including both SMAD-dependent and non-SMAD pathways. The SMAD family comprises signal transduction molecules activated by ligands of the TGF-β superfamily (e.g., TGF-β, BMPs), mainly divided into three categories: receptor-regulated SMADs (R-SMADs), common-mediator SMADs, and inhibitory SMADs.27

The SMAD-dependent TGF-β signaling pathway in hepatic fibrosis

The SMAD signaling pathway is a classic TGF-β signaling pathway. TGF-β binds to the TGFβRII on the cell membrane in its precursor form, causing a conformational change in TGFβRII and recruitment of TGFβRI (ALK5), which is then phosphorylated. This leads to the phosphorylation of intracellular SMAD2 and SMAD3.28 These R-SMADs form a heteromeric complex with common-mediator SMAD (SMAD4), which accumulates in the nucleus.27 In the nucleus, the SMAD complex associates with other transcription factors and regulatory proteins to regulate the transcription of target genes. Additionally, phosphorylation of SMAD2/3 enhances production of MMP1, α-SMA, and type I collagen, promoting liver fibrosis (Fig. 1).14

TGF-β signaling pathway in hepatic fibrosis.
Fig. 1  TGF-β signaling pathway in hepatic fibrosis.

TGF-β binds to TGF-β receptors and activates both SMAD-dependent and SMAD-independent pathways. SMAD-independent pathways include p38 MAPK, JNK, NF-κB, PI3K/Akt, and ERK1/2 signaling pathways. Created with BioRender.

SMAD2/3 signaling plays a vital role in liver fibrosis. Deletion of SMAD3 was reported to prevent liver fibrosis induced by dimethylnitrosamine. SMAD3 acts as a potent stimulator of ECM accumulation and may be a novel target for treating chronic hepatitis complicated by fibrosis.29 Hepatocyte-specific deletion of SMAD4 inhibited hepatocarcinogenesis in mice by attenuating fibrosis and reducing myeloid-derived suppressor cell infiltration.30 Moreover, inhibitory SMADs, including SMAD6 and SMAD7, are important antagonists of TGF-β signaling. Studies show that SMAD6 competes with SMAD4 to bind activated SMAD1, thereby inhibiting SMAD1 signaling and SMAD complex formation.31

Therefore, SMAD6 and SMAD7 act as negative regulators of TGF-β-mediated liver fibrosis.32 SMAD6 primarily inhibits BMP signaling, whereas SMAD7 is involved in both BMP and TGF-β signaling (Fig. 1).31 SMAD7 binds directly to activated type I TGF-β receptors, occupying the same kinase surface that would normally dock SMAD2/3. This competitive binding prevents further phosphorylation and activation of R-SMADs. SMAD7 also competes with SMAD4, preventing the formation of a functional transcriptional complex.33

In liver fibrosis, increased expression of SMAD2 and SMAD3 positively regulates fibrogenesis, whereas decreased expression of SMAD7 negatively regulates it.34 Overexpression of SMAD7 inhibits SMAD3 and significantly reduces the fibrotic response in animal liver fibrosis models, demonstrating SMAD7’s anti-fibrotic effects by antagonizing the TGF-β/SMAD3 signaling pathway.35 Lemonine reduces hepatocyte EMT and HSC activation by upregulating SMAD7, reducing C-terminal phosphorylation (p-SMAD2/3c) and nuclear translocation of SMAD2/3, and inhibiting the TGF-β/SMAD signaling pathway, thereby inhibiting carbon tetrachloride (CCl4)-induced liver fibrosis in mice.36

The SMAD signaling pathway has been extensively studied in liver fibrosis. In human keratinocytes, TGF-β induces phosphorylation of SMAD2 and SMAD3, as well as SMAD1 and SMAD5, both dependent on type I and type II kinase activity of the TGF-β receptor.37 Beyond the canonical SMAD pathway, TGF-β also promotes HSC activation via the ALK1/SMAD1/5 pathway and non-SMAD pathways, including MAPK and PI3K/AKT.18,38 These pathways can act directly or synergistically with SMAD proteins to regulate gene expression, leading to increased ECM, decreased MMP expression, and promotion of fibrosis.23,28,39 For example, activated TGF-β1 increases kindlin-2 expression through p38 and MAPK signaling, while kindlin-2 overexpression enhances TGF-β signaling by upregulating SMAD2/3 phosphorylation (Fig. 1).40

In HSCs, TGF-β promotes expression of differentiation-inducing factor-1 through the ALK1/SMAD1/5 pathway. Differentiation-inducing factor-1 is a key mediator of liver fibrosis, promoting transdifferentiation of HSCs into myofibroblasts and leading to fibrosis.41 ALK1 is a transmembrane serine/threonine receptor kinase belonging to the TGF-β receptor family, mainly expressed in endothelial cells.42 One study showed that ALK1 regulates SMAD1/5 phosphorylation via TGF-β.43 Recently, Anassuya Ramachandran and colleagues found that TGF-β-induced SMAD1/5 phosphorylation requires two type I receptors: the typical TGF-β receptor TGF-βRI and the classic BMP type I receptor ACVR1. The kinase activity of TGF-βRI is necessary to activate ACVR1, and ACVR1 kinase activity is essential for SMAD1/5 phosphorylation (Fig. 1).44

The SMAD-independent TGF-β signaling pathway in liver fibrosis

PI3K/Akt pathway

The PI3K/AKT pathway plays an important regulatory role in liver fibrosis progression by modulating cell proliferation, differentiation, autophagy, apoptosis, and other functions, including promoting and inhibiting fibrosis.45 TGF-β induces PI3K activation, possibly via interaction of the p85 subunit of PI3K with the TGF-β receptor. Activated AKT controls translational responses through mTOR1.46 EMT is a crucial step in tumor progression and fibrosis, with TGF-β signaling playing a key role. Activation of the PI3K/AKT pathway contributes to TGF-β-induced EMT as well as fibroblast proliferation.47,48 In an in vitro study, Mi et al. investigated the involvement of the PI3K/AKT pathway in HSC activation, a driver of liver fibrosis progression. Their study revealed that maltol attenuated liver fibrosis by inducing apoptosis in activated HSCs via regulation of the TGF-β1-mediated PI3K/Akt pathway.49 Similarly, salvianolic acid A was reported to prevent HSC stimulation by inhibiting the PI3K/AKT/mTOR signaling pathway (Fig. 1).50

MAPK pathway

MAPKs are a family of serine/threonine kinases that regulate cellular processes by transmitting extracellular signals into intracellular responses. These include extracellular signal-regulated kinase (ERK), p38 MAPK, and c-Jun N-terminal kinase (JNK), which regulate apoptosis and proliferation.51–54 MAPK signaling plays a critical role in regulating liver inflammation.55 TGF-β1 activates HSCs by inducing autophagy through ERK and JNK activation in the MAPK pathway (Fig. 1).55

ERK is the most widely studied MAPK signaling pathway, consisting mainly of ERK1 (p44 MAPK) and ERK2 (p42 MAPK). The activation of ERK1/2 is closely associated with HSC activation. Studies have found that incubation of HSCs with cytokines induces interleukin (IL)-11 production, leading to the activation (phosphorylation) of ERK and increases fibrosis markers. Mice injected with IL-11 developed liver damage and fibrosis, whereas blocking IL-11 signaling reduced liver fibrosis in mice. Lee et al. found that TGF-β directly phosphorylates ShcA to activate ERK MAPK signaling.56 ShcA is essential for TGF-β-induced ERK activation (Fig. 1).57

TGF-β ligand binding also activates JNK and p38 MAPK signaling pathways via TGF-β receptors.58 TGF-β-activated kinase 1 (TAK1), a MAPK family member with anti-inflammatory effects,51 can be activated by multiple stimuli, including TGF-β, tumor necrosis factor-α, IL-1, and lipopolysaccharide.55 TGF-β1-induced TAK1 activation triggers downstream signaling through MKK4-JNK, MKK3/6-p38 cascades, and the NF-κB-inducing IκB kinase pathway, regulating apoptosis, proliferation, collagen synthesis, and inflammatory cytokines.55 Hepatocyte-specific TAK1 deletion causes liver injury, fibrosis, and even HCC.59 The critical role of this pathway was further demonstrated as mice deficient in both TAK1 and Tgfbr2 exhibited fewer activated HSCs and reduced fibrogenic gene expression.60 TGF-β1-induced TAK1 activation regulates collagen and inflammatory cytokine expression via the MKK4-JNK and MKK3-p38 cascades.55 Within cells, JNK activates fibrotic genes and stimulates collagen production. Proinflammatory cytokines activate JNK, which in turn promotes TGF-β expression, amplifying fibrosis. Fabre et al. found that IL-17A enhances the response of HSCs to TGF-β by activating the JNK pathway, resulting in increased profibrotic gene expression and collagen production (Fig. 1).61

p38 MAPK plays a significant role in promoting liver inflammation progression. Both JNK and p38 MAPK participate in HSC activation, which leads to ECM production and liver fibrosis.62 A previous study showed that the RING finger protein PNF2, aberrantly expressed in tumors, is highly upregulated in human fibrotic liver tissue. Knockdown of PNF2 inhibited ERK/p38 signaling, thereby reducing liver fibrosis.63 Additionally, HSCs treated with IL-6 differentiate into myofibroblast-like cells; inhibition of the MAPK pathway suppresses HSC activation (Fig. 1).64

Targeting TGF-β to treat liver fibrosis

Current therapeutic strategies for hepatic fibrosis still focus primarily on etiological control. However, etiological treatment alone cannot meet the needs of all patients, making the development of specific anti-fibrotic agents urgently necessary. Table 1 summarizes drugs currently in use for treating hepatic fibrosis that are undergoing clinical trials.65–70

Table 1

Targeted therapies against liver fibrosis

Mechanistic classGeneric nameGov numberRef
FXR agonistObeticholic AcidNCT0254835166
CCR2/CCR5 dual antagonistCenicrivirocNCT0221747567
Pan-PPAR agonistLanifibranorNCT0300807068
THR-β selective agonistResmetiromNCT0390042969
LOXL2 monoclonal antibodySimtuzumabNCT01672866, NCT0167287970
ALK5 inhibitorGalunisertibNCT0124698665

Given the vital role of TGF-β in liver fibrosis, inhibition of the TGF-β signaling pathway is considered a promising therapeutic strategy.14,71 Most anti-TGF-β treatments target various steps of the canonical TGF-β/SMAD signaling pathway to inhibit TGF-β signaling. These approaches can be broadly divided into three categories: (1) monoclonal antibodies that prevent TGF-β receptor binding to ligands, (2) TGF-β receptor kinase small-molecule inhibitors, and (3) small molecules or antibodies that selectively interfere with TGF-β1 activation.72 These inhibitors include both natural and synthetic compounds. Several TGF-β signaling inhibitors, such as neutralizing antibodies and receptor kinase inhibitors, have been tested in clinical trials.73 The latest TGF-β inhibitors and related pathway blockers as potential therapies for liver fibrosis are summarized in Table 2.74–86 Additionally, we screened TGF-β inhibitors for fibrosis treatment, and clinical trial results are presented in Table 3.87–89

Table 2

The latest TGF-β inhibitors and related pathway blockers as potential therapies for liver fibrosis

DrugTargetsDiseaseRef
GC1008 (Fresolimumab)Pan-TGF-βAdvanced malignant melanoma and renal cell carcinoma74,75
CAT-152 (Genzyme)TGF-β2/3Reduction of scar after glaucoma surgery76
CAT192 (Metilimumab)TGF-β1Diffuse Systemic Sclerosis77
AP-12009 (Trabedersen)TGF-β2 mRNAOncology83
LY2157299 (Galunisertib)TGFβRI kinaseLiver fibrosis79
TP-0427736ALK5 inhibitorLiver fibrosis80
LY2109761TGFβRI & RII KinaseLiver fibrosis81,82
P11 P12 P144, P54 P106TGF-β1Liver fibrosis78
PirfenidoneTGF-β/SMAD signalingLiver fibrosis84
HydronidoneTGF-β/SMAD signalingLiver fibrosis85
PentoxifyllineTGF-β/SMAD signalingLiver fibrosis86
Table 3

Clinical trial landscape of TGF-β inhibitors

DrugTargetDiseaseGov numberSafetyEffectivenessCompletion rateRef
GalunisertibTGFβRI kinaseLiver fibrosisNCT01246986well toleratedImproved overall survival93%87
VactosertibALK5 inhibitorDesmoid TumorsNCT03802084well toleratedProlonged survival100%88
PirfenidoneTGF-β/SMAD signalingPulmonary fibrosisNCT01366209well tolerateddelayed disease progression/89

The effects of TGF-β and its targeted interventions may produce either beneficial or detrimental outcomes for the organ, largely depending on the disease stage. Therefore, it is critical to strategically select the precise therapeutic window, target the correct cell types, and interfere with detrimental signaling branches downstream of the pathway while preserving its protective functions.71

Direct blockade of TGF-β

TGF-β antibodies are monoclonal antibodies that specifically target TGF-β. By directly binding to the TGF-β protein, they prevent it from binding to its receptor, thereby inhibiting its biological effects.90 Monoclonal antibodies targeting TGF-β include CAT-152, CAT-192, and GC1008, which have been tested for systemic inhibition in fibrotic diseases and cancer.91

Fresolimumab (GC1008) is a fully human monoclonal antibody targeting all three TGF-β isoforms (TGF-β1, -β2, and -β3). It is currently used in clinical trials for idiopathic pulmonary fibrosis, focal segmental glomerulosclerosis, and cancer. To date, no clinical trial data on liver fibrosis have been reported.74,75 A clinical trial in chemotherapy-refractory metastatic breast cancer patients found that high-dose fresolimumab improved overall survival compared with low-dose.75

The human monoclonal antibody CAT-152 (Cambridge Antibody Technology, Genzyme) primarily inhibits TGF-β2 activation. Two Phase III clinical studies have suggested that CAT-152 may treat fibrosis after trabeculectomy and prevent postoperative scar formation.76,92 Another study showed that CAT-152 significantly inhibited streptozotocin-induced renal fibrosis in diabetic rats compared to controls.93

Metelimumab (CAT-192) is a human IgG4 monoclonal antibody developed as a TGF-β1-specific antagonist. It was tested in clinical trials for early diffuse cutaneous systemic sclerosis. However, Phase I/II trials showed no evidence of therapeutic effect, and the study was terminated due to lack of efficacy.77

Polypeptides derived from TGF-β1 (P11 and P12) and from its type III receptor (P54, P144, and P106) have shown potential in reducing liver fibrosis in chronic liver injury. Studies demonstrated that these peptides block TGF-β1 binding to its cellular receptors and antagonize its activity in vitro. In a rat CCl4-induced liver fibrosis model, low-dose intraperitoneal injection of P144 improved fibrosis (Fig. 2).78

Therapeutic approaches to inhibit TGF-β/SMAD-induced liver fibrosis.
Fig. 2  Therapeutic approaches to inhibit TGF-β/SMAD-induced liver fibrosis.

Schematic representation of different strategies targeting TGF-β signaling for liver fibrosis therapy. Created with BioRender.

As noted, the effects of TGF-β and its targeted interventions may yield either beneficial or detrimental outcomes for the organ, largely contingent on the disease stage. It is therefore essential to select the optimal therapeutic window, target appropriate cell types, and inhibit harmful downstream signaling branches while preserving protective functions.71

TGF-β1 is the predominant profibrotic and immunosuppressive isoform in liver disease, while TGF-β2/3 maintain regenerative capacity and immune homeostasis. SRK-181, a selective TGF-β1 inhibitor, prevented valvulopathy in rats and exhibited no measurable effect on human platelet aggregation, activation, or binding. This agent is currently in a Phase 1 clinical trial.33 SRK-181, combined with anti-PD-(L)1 antibodies, is also under evaluation in a Phase I trial for solid tumors.33

The critical role of integrin-mediated TGF-β activation in hepatic fibrosis is well established. Conditional knockout of αv integrins in HSCs profoundly suppresses fibrogenesis in experimental models.94,95

PLN-1474, a selective αvβ1 integrin inhibitor, targets TGF-β1 activation in the liver to reduce fibrosis. It has completed a Phase I clinical trial for NASH-associated liver fibrosis, demonstrating favorable safety and pharmacokinetic profiles.96

TGF-β receptor inhibitor

LY2157299 (Galunisertib) is an oral small-molecule inhibitor of TGF-β receptor I kinase with antitumor activity and potential use in treating glioblastoma, pancreatic cancer, and HCC.65,79 Galunisertib blocks activation of the ALK5 pathway and inhibits SMAD2 phosphorylation, while SMAD1 phosphorylation remains unchanged. Studies have shown that galunisertib inhibits TGF-β-induced collagen production by blocking TGF-β receptors. The treatment has a manageable safety profile.87 A clinical study involving patients with advanced liver cancer showed that LY2157299 exhibits favorable pharmacokinetics and antitumor efficacy, with no cardiovascular toxicity detected.79 Luangmonkong et al. used precise liver sections from healthy adults and cirrhotic patients to evaluate galunisertib’s anti-fibrotic properties, finding significant anti-fibrotic effects likely mediated by inhibition of SMAD2 phosphorylation.97 Galunisertib was administered on an intermittent dosing regimen (14 days on/14 days off). In HCC patients exhibiting a TGF-β1 response, treatment significantly prolonged median overall survival (22.8 months vs. 12.0 months) while maintaining an acceptable safety profile.65

TP-0427736, a novel ALK5 inhibitor first reported in 2013, inhibits TGF-β1-induced SMAD2/3 phosphorylation in A549 cells in a concentration-dependent manner and shows potential as a therapy for Androgenic alopecia.80 Employing delivery vehicles to selectively transport ALK5 inhibitors to HSCs significantly enhances therapeutic efficacy while reducing systemic exposure risks.98

Another TGF-β receptor I kinase inhibitor, LY2109761, has been evaluated in HCC models and found to block HCC cell migration and invasion (Fig. 2).81,82

The αvβ8 integrin binds latent TGF-β1 and induces ligand activation in situ without releasing the complex, providing a structural basis for compartment-specific blockade of pathogenic TGF-β signaling.99 Bexotegrast exerts dual-target inhibition of αvβ6/αvβ1-mediated latent TGF-β activation, simultaneously blocking disease-driving TGF-β signaling from epithelial and fibroblast sources while avoiding systemic TGF-β-related toxicities. Phase I-II data indicate favorable safety and dose-dependent anti-fibrotic signaling responses, demonstrating promising therapeutic potential in pulmonary fibrosis.100

Antisense oligonucleotides (ASOs)

The key step in activating the TGF-β signaling pathway is ligand binding to its receptor. Classical TGF-β signal transduction involves ligand binding to the TGF-β type II receptor, phosphorylation of SMAD2/3, and activation of SMAD2/3/4 complexes that regulate TGF-β target gene transcription.27 Therefore, targeting the transcriptional effects of TGF-β is critical.91 ASOs are single-stranded, modified oligonucleotides that regulate gene expression by binding specifically to target mRNAs. One study showed that using ASOs to antagonize TGF-β1 and SMAD3 inhibited TGF-β signaling, thereby reducing postoperative scar formation.101

Trabedersen (AP-12009) is an 18-mer phosphorothioate ASO designed to bind specifically to TGF-β2 mRNA, preventing its translation.83 It inhibits TGF-β2 production in advanced glioma, melanoma, and colorectal cancer cells. AP15012 and AP11014 have been tested in preclinical trials for prostate, non-small-cell lung, and colorectal cancers. Currently, no clinical trials for HCC have been reported.102 A clinical study of trabedersen (AP-12009) in glioma treatment showed significantly improved survival rates compared with controls, with 10 µM identified as the optimal dose (Fig. 2).83

Inhibition of TGF-β/SMAD signaling

Seniutkin found that pirfenidone exhibited a strong anti-fibrotic effect in early liver fibrosis, but was less effective in late-stage fibrosis and showed no protective effect against liver cancer.84 Hydronidone, a derivative of pirfenidone, demonstrated in a Phase II clinical trial that when combined with entecavir, it significantly improved liver fibrosis in patients with chronic hepatitis B compared with entecavir alone after 52 weeks.85 Another study showed that hydroxynidone inhibited phosphorylation of proteins in the TGF-β signaling pathway and alleviated CCl4-induced liver fibrosis in rats (Fig. 2).103

As a pirfenidone derivative, hydroxynidone can inhibit phosphorylation within the TGF-β signaling pathway. Additionally, a clinical study showed that fenofibrate combined with pentoxifylline significantly reduced liver fibrosis markers and liver stiffness in patients with non-alcoholic fatty liver disease compared to fenofibrate alone (Fig. 2).104

Pentoxifylline is considered an effective antifibrotic agent that inhibits HSC activation in vitro. Pentoxifylline exhibits anti-hepatic fibrosis effects both in vitro and in vivo.105 In addition, it has protective effects on the lungs and kidneys. Evidence shows that pentoxifylline treatment benefits patients with chronic hepatitis C, improving inflammation and fibrosis, with effects becoming more pronounced after two years of treatment (Fig. 2).86,106

Traditional Chinese medicine (TCM)

Natural products contribute to drug discovery, especially for cancer and infectious diseases.107 TCM, characterized by multi-component, multi-target, and multi-pathway actions, offers unique advantages in treating liver fibrosis.108,109 Many natural products derived from TCM, including flavonoids, alkaloids, and terpenoids, have demonstrated significant anti-fibrotic activity.109 TCM-derived compounds with anti-fibrotic potential are summarized in Table 4.14,36,110–130

Table 4

TCM-derived compounds with anti-fibrotic potential

CompoundsSourceMolecular formulaMechanismsRefAdditional targetsRef
PiperinePiper nigrumC17H19NO3Inhibiting TGF-β expression110NF-κB120
SinomenineSinomenium acutumC19H23NO4Inhibiting TGF-β/SMAD path-way111Nrf2-HO-1121
CaffeineCoffea arabica LC8H10N4O2Inhibiting SMAD2/3 phosphorylation112Adenosine A2A receptors122
CapsaicinCapsicum annuum L.C18H27NO3Inhibiting TGF-β1/SMAD pathway113TRPV1123
EvodiamineEuodia rutaecarpa (Juss.) BenthC19H17N3OInhibiting TGF-β1/SMAD pathway14Gut microbiota124
MatrineSophora flavescens Alt.C15H24N2OInhibiting TGF-β1 expression114PI3K-Akt125
ChrysinOroxylum indicum (Linn.) KurzC15H10O4Inhyibiting TGF-β1/SMAD path-way115AMPK126
LigustroflavoneLigustrum lucidum AitC33H40OInhibiting TGF-β/SMAD path-way116//
QuercetinSophora flavescens Ait.C15H10O7Regulating p38 MAPK and TGF-β1/SMADs pathway117,127PI3K/Akt127
LimoninCitrus aurantium LC26H30O8Upregulating SMAD736AMPK128
AndrographolideAndrographis paniculateC20H30O5Inhibiting the TGF-β1/SMAD2 pathway119NF-κB129
PaeoniflorinPaeonia lactiflora Pall.C23H28O1Inhibiting TGF-β1/SMADs signaling118PPAR-γ130

Alkaloids are highly bioactive nitrogen-containing organic compounds widely found in TCM. Numerous studies have shown that alkaloids possess anti-hepatic fibrosis effects.131 For example, piperine inhibits TGF-β/SMAD signaling by restoring SMAD7, thereby further inhibiting HSC activation (Fig. 2).110 Chen et al. reported that sinomenine suppressed activation of the TGF-β/SMAD pathway both in vitro and in vivo, alleviating acute liver injury.111 In addition, caffeine, capsaicin, evodiamine, and matrine can inhibit liver fibrosis by modulating the TGF-β/SMAD pathway (Fig. 2).112–114,132

Flavonoids, abundant in plants and berries, exhibit various biological effects. They reduce or reverse liver fibrosis via multiple pathways and targets.133 Chrysin inhibits HSC activation through the TGF-β/SMAD.115 Ligustroflavone alleviates CCl4-induced liver fibrosis by downregulating the TGF-β/SMAD pathway.116 Quercetin prevents liver fibrosis by reducing TGF-β levels and inhibiting the p38 MAPK signaling pathway.117

Terpenoids exhibit antioxidant, metabolic, immunomodulatory, and anti-inflammatory activities. Recognized as anticancer agents, they show good prospects for treating chronic liver diseases.133 For example, Euphorbesulin A (10), extracted from Euphorbia sieboldiana, inhibits the TGF-β/SMAD signaling pathway and represents a novel anti-liver fibrosis drug; its target may be the TGF-β type I receptor.134 Limonin alleviates CCl4-induced liver fibrosis by upregulating SMAD7, which suppresses the TGF-β/SMAD cascade.36 Furthermore, andrographolide and paeoniflorin can treat liver fibrosis by regulating the TGF-β1/SMAD pathway (Fig. 2).118,119

Discussion

Hepatic fibrosis is a complex pathological process involving multiple factors, targets, and pathways that can seriously harm human health. Numerous studies have demonstrated that TGF-β plays a critical role in liver fibrosis through multiple signaling pathways, and TGF-β inhibitors hold significant promise for its treatment. Notably, the TGF-βRI kinase inhibitor galunisertib has been clinically used to improve overall survival in patients with HCC. However, in recent years, few clinical trials have targeted TGF-β for treating HCC and liver fibrosis. This may be because dysregulation of the TGF-β cascade is not the primary driver of HCC development, and currently, no TGF-β inhibitors for liver fibrosis have reached the market.

Although the TGF-β signaling pathway is a core driver of fibrosis, current inhibitors face challenges related to efficacy and safety. Therefore, developing next-generation TGF-β inhibitors with improved selectivity, tissue-targeting capabilities, or mechanisms to overcome existing limitations, such as isoform-specific inhibitors, prodrugs, or targeted delivery systems, should be prioritized in the anti-fibrotic drug development pipeline.

Due to the diverse structures, wide availability, multiple targets, and pathways of natural products, exploration of these compounds for liver fibrosis treatment is expanding rapidly. In recent years, many TCMs and their active ingredients have been found to effectively treat hepatic fibrosis through multiple mechanisms with significant effects. However, most of these studies remain at the in vitro and in vivo experimental stages and lack detailed preclinical and clinical evaluation. Furthermore, research on targets and mechanisms is not yet comprehensive, and pharmacokinetic and pharmacological profiles require further investigation. Therefore, thorough mechanistic studies are essential to define precise targets, pathway modulation, and metabolic behavior in vivo, facilitating the rapid translation of these agents into clinical-grade therapeutics.

Given the complex pathological mechanisms underlying fibrosis, combination therapies targeting distinct pathways or cell types, such as combining anti-inflammatory and anti-fibrotic agents or co-administering drugs against different fibrogenic drivers, demonstrate significant potential to overcome drug resistance and enhance therapeutic efficacy, warranting active progression to clinical evaluation.

The TGF-β pathway regulates multiorgan homeostasis, and systemic inhibition may cause immunosuppression, impaired wound healing, and increased tumorigenic risk. Traditional small-molecule inhibitors exhibit off-target effects, and monotherapy struggles to modulate multiple pathological processes simultaneously, including fibrogenesis, inflammation, and metabolic dysregulation. Future research should focus on enhancing localized targeting through organ-restricted inhibitors or novel drug delivery systems (e.g., nanoparticles) to improve selectivity, as well as developing synergistic combination therapies for multi-pathway intervention in hepatic fibrosis.

Conclusions

TGF-β signaling inhibitors hold important therapeutic potential for hepatic fibrosis. Although many challenges remain, detailed exploration of these inhibitors, including certain natural products, is necessary to clarify their anti-fibrotic effects. In the future, effective anti-fibrotic drugs may be developed to alleviate or even reverse the progression of liver fibrosis, ushering in a new era in its pharmacological treatment.

Declarations

Funding

This research was funded by the National Natural Science Foundation of China (No. 82074372,82474231), the Shanghai Science and Technology Innovation Action Plan (No. 23Y21920200), and the Three-year Action Plan for Strengthening Public Health System Construction in Shanghai Municipality (No. GWVI-2.1.5).

Conflict of interest

The authors have no conflict of interests related to this publication.

Authors’ contributions

Conceptualization (WZ, YC), writing - original draft preparation (WZ, JQ), writing - review and editing (XZ, YG), and supervision (ZS, LH). All authors have read and agreed to the published version of the manuscript.

References

  1. Kisseleva T, Brenner D. Molecular and cellular mechanisms of liver fibrosis and its regression. Nat Rev Gastroenterol Hepatol 2021;18(3):151-166 View Article PubMed/NCBI
  2. Tanwar S, Rhodes F, Srivastava A, Trembling PM, Rosenberg WM. Inflammation and fibrosis in chronic liver diseases including non-alcoholic fatty liver disease and hepatitis C. World J Gastroenterol 2020;26(2):109-133 View Article PubMed/NCBI
  3. Henderson NC, Rieder F, Wynn TA. Fibrosis: from mechanisms to medicines. Nature 2020;587(7835):555-566 View Article PubMed/NCBI
  4. Sanjabi S, Oh SA, Li MO. Regulation of the Immune Response by TGF-β: From Conception to Autoimmunity and Infection. Cold Spring Harb Perspect Biol 2017;9(6):a022236 View Article PubMed/NCBI
  5. Arriazu E, Ruiz de Galarreta M, Cubero FJ, Varela-Rey M, Pérez de Obanos MP, Leung TM, et al. Extracellular matrix and liver disease. Antioxid Redox Signal 2014;21(7):1078-1097 View Article PubMed/NCBI
  6. Meng XM, Nikolic-Paterson DJ, Lan HY. TGF-β: the master regulator of fibrosis. Nat Rev Nephrol 2016;12(6):325-338 View Article PubMed/NCBI
  7. Tirado-Rodriguez B, Ortega E, Segura-Medina P, Huerta-Yepez S. TGF- β: an important mediator of allergic disease and a molecule with dual activity in cancer development. J Immunol Res 2014;2014:318481 View Article PubMed/NCBI
  8. Tzavlaki K, Moustakas A. TGF-β Signaling. Biomolecules 2020;10(3):487 View Article PubMed/NCBI
  9. Schon HT, Weiskirchen R. Immunomodulatory effects of transforming growth factor-β in the liver. Hepatobiliary Surg Nutr 2014;3(6):386-406 View Article PubMed/NCBI
  10. Braczkowski MJ, Kufel KM, Kulińska J, Czyż DŁ, Dittmann A, Wiertelak M, et al. Pleiotropic Action of TGF-Beta in Physiological and Pathological Liver Conditions. Biomedicines 2024;12(4):925 View Article PubMed/NCBI
  11. Fabregat I, Caballero-Díaz D. Transforming Growth Factor-β-Induced Cell Plasticity in Liver Fibrosis and Hepatocarcinogenesis. Front Oncol 2018;8:357 View Article PubMed/NCBI
  12. Kimura M, Moteki H, Ogihara M. Role of Hepatocyte Growth Regulators in Liver Regeneration. Cells 2023;12(2):208 View Article PubMed/NCBI
  13. Xu F, Liu C, Zhou D, Zhang L. TGF-β/SMAD Pathway and Its Regulation in Hepatic Fibrosis. J Histochem Cytochem 2016;64(3):157-167 View Article PubMed/NCBI
  14. Dewidar B, Meyer C, Dooley S, Meindl-Beinker AN. TGF-β in Hepatic Stellate Cell Activation and Liver Fibrogenesis-Updated 2019. Cells 2019;8(11):E1419 View Article PubMed/NCBI
  15. Zhang CY, Yuan WG, He P, Lei JH, Wang CX. Liver fibrosis and hepatic stellate cells: Etiology, pathological hallmarks and therapeutic targets. World J Gastroenterol 2016;22(48):10512-10522 View Article PubMed/NCBI
  16. Zhang J, Jiang N, Ping J, Xu L. TGF-β1-induced autophagy activates hepatic stellate cells via the ERK and JNK signaling pathways. Int J Mol Med 2021;47(1):256-266 View Article PubMed/NCBI
  17. Wang J, Zhao X, Wan YY. Intricacies of TGF-β signaling in Treg and Th17 cell biology. Cell Mol Immunol 2023;20(9):1002-1022 View Article PubMed/NCBI
  18. Yoshida K, Murata M, Yamaguchi T, Matsuzaki K. TGF-β/Smad signaling during hepatic fibro-carcinogenesis (review). Int J Oncol 2014;45(4):1363-1371 View Article PubMed/NCBI
  19. Shull MM, Ormsby I, Kier AB, Pawlowski S, Diebold RJ, Yin M, et al. Targeted disruption of the mouse transforming growth factor-beta 1 gene results in multifocal inflammatory disease. Nature 1992;359(6397):693-699 View Article PubMed/NCBI
  20. Kotlarz D, Marquardt B, Barøy T, Lee WS, Konnikova L, Hollizeck S, et al. Human TGF-β1 deficiency causes severe inflammatory bowel disease and encephalopathy. Nat Genet 2018;50(3):344-348 View Article PubMed/NCBI
  21. Kovacs RJ, Maldonado G, Azaro A, Fernández MS, Romero FL, Sepulveda-Sánchez JM, et al. Cardiac Safety of TGF-β Receptor I Kinase Inhibitor LY2157299 Monohydrate in Cancer Patients in a First-in-Human Dose Study. Cardiovasc Toxicol 2015;15(4):309-323 View Article PubMed/NCBI
  22. Gonzalez-Sanchez E, Vaquero J, Férnandez-Barrena MG, Lasarte JJ, Avila MA, Sarobe P, et al. The TGF-β Pathway: A Pharmacological Target in Hepatocellular Carcinoma?. Cancers (Basel) 2021;13(13):3248 View Article PubMed/NCBI
  23. Peng D, Fu M, Wang M, Wei Y, Wei X. Targeting TGF-β signal transduction for fibrosis and cancer therapy. Mol Cancer 2022;21(1):104 View Article PubMed/NCBI
  24. Heldin CH, Moustakas A. Signaling Receptors for TGF-β Family Members. Cold Spring Harb Perspect Biol 2016;8(8):a022053 View Article PubMed/NCBI
  25. Paul MK, Mukhopadhyay AK. Tyrosine kinase - Role and significance in Cancer. Int J Med Sci 2004;1(2):101-115 View Article PubMed/NCBI
  26. Derynck R, Budi EH. Specificity, versatility, and control of TGF-β family signaling. Sci Signal 2019;12(570):eaav5183 View Article PubMed/NCBI
  27. Hata A, Chen YG. TGF-β Signaling from Receptors to Smads. Cold Spring Harb Perspect Biol 2016;8(9):a022061 View Article PubMed/NCBI
  28. Finnson KW, Almadani Y, Philip A. Non-canonical (non-SMAD2/3) TGF-β signaling in fibrosis: Mechanisms and targets. Semin Cell Dev Biol 2020;101:115-122 View Article PubMed/NCBI
  29. Latella G, Vetuschi A, Sferra R, Catitti V, D’Angelo A, Zanninelli G, et al. Targeted disruption of Smad3 confers resistance to the development of dimethylnitrosamine-induced hepatic fibrosis in mice. Liver Int 2009;29(7):997-1009 View Article PubMed/NCBI
  30. Xin X, Li Z, Yan X, Liu T, Li Z, Chen Z, et al. Hepatocyte-specific Smad4 deficiency inhibits hepatocarcinogenesis by promoting CXCL10/CXCR3-dependent CD8(+)- T cell-mediated anti-tumor immunity. Theranostics 2024;14(15):5853-5868 View Article PubMed/NCBI
  31. Miyazawa K, Miyazono K. Regulation of TGF-β Family Signaling by Inhibitory Smads. Cold Spring Harb Perspect Biol 2017;9(3):a022095 View Article PubMed/NCBI
  32. Deng L, Huang L, Guo Q, Shi X, Xu K. CREB1 and Smad3 mediate TGF-β3-induced Smad7 expression in rat hepatic stellate cells. Mol Med Rep 2017;16(6):8455-8462 View Article PubMed/NCBI
  33. Deng Z, Fan T, Xiao C, Tian H, Zheng Y, Li C, et al. TGF-β signaling in health, disease, and therapeutics. Signal Transduct Target Ther 2024;9(1):61 View Article PubMed/NCBI
  34. Walton KL, Johnson KE, Harrison CA. Targeting TGF-β Mediated SMAD Signaling for the Prevention of Fibrosis. Front Pharmacol 2017;8:461 View Article PubMed/NCBI
  35. Yan X, Liu Z, Chen Y. Regulation of TGF-beta signaling by Smad7. Acta Biochim Biophys Sin (Shanghai) 2009;41(4):263-272 View Article PubMed/NCBI
  36. Shu G, Dai C, Yusuf A, Sun H, Deng X. Limonin relieves TGF-β-induced hepatocyte EMT and hepatic stellate cell activation in vitro and CCl(4)-induced liver fibrosis in mice via upregulating Smad7 and subsequent suppression of TGF-β/Smad cascade. J Nutr Biochem 2022;107:109039 View Article PubMed/NCBI
  37. Bharathy S, Xie W, Yingling JM, Reiss M. Cancer-associated transforming growth factor beta type II receptor gene mutant causes activation of bone morphogenic protein-Smads and invasive phenotype. Cancer Res 2008;68(6):1656-1666 View Article PubMed/NCBI
  38. Luo K. Signaling Cross Talk between TGF-β/Smad and Other Signaling Pathways. Cold Spring Harb Perspect Biol 2017;9(1):a022137 View Article PubMed/NCBI
  39. Muñoz-Félix JM, González-Núñez M, López-Novoa JM. ALK1-Smad1/5 signaling pathway in fibrosis development: friend or foe?. Cytokine Growth Factor Rev 2013;24(6):523-537 View Article PubMed/NCBI
  40. Yu J, Hu Y, Gao Y, Li Q, Zeng Z, Li Y, et al. Kindlin-2 regulates hepatic stellate cells activation and liver fibrogenesis. Cell Death Discov 2018;4:34 View Article PubMed/NCBI
  41. Wiercinska E, Wickert L, Denecke B, Said HM, Hamzavi J, Gressner AM, et al. Id1 is a critical mediator in TGF-beta-induced transdifferentiation of rat hepatic stellate cells. Hepatology 2006;43(5):1032-1041 View Article PubMed/NCBI
  42. Roman BL, Hinck AP. ALK1 signaling in development and disease: new paradigms. Cell Mol Life Sci 2017;74(24):4539-4560 View Article PubMed/NCBI
  43. Zhang H, Du L, Zhong Y, Flanders KC, Roberts JD. Transforming growth factor-β stimulates Smad1/5 signaling in pulmonary artery smooth muscle cells and fibroblasts of the newborn mouse through ALK1. Am J Physiol Lung Cell Mol Physiol 2017;313(3):L615-L627 View Article PubMed/NCBI
  44. Ramachandran A, Vizán P, Das D, Chakravarty P, Vogt J, Rogers KW, et al. TGF-β uses a novel mode of receptor activation to phosphorylate SMAD1/5 and induce epithelial-to-mesenchymal transition. Elife 2018;7:e31756 View Article PubMed/NCBI
  45. Shamsan E, Almezgagi M, Gamah M, Khan N, Qasem A, Chuanchuan L, et al. The role of PI3k/AKT signaling pathway in attenuating liver fibrosis: a comprehensive review. Front Med (Lausanne) 2024;11:1389329 View Article PubMed/NCBI
  46. Zhang YE. Non-Smad Signaling Pathways of the TGF-β Family. Cold Spring Harb Perspect Biol 2017;9(2):a022129 View Article PubMed/NCBI
  47. Wilkes MC, Mitchell H, Penheiter SG, Doré JJ, Suzuki K, Edens M, et al. Transforming growth factor-beta activation of phosphatidylinositol 3-kinase is independent of Smad2 and Smad3 and regulates fibroblast responses via p21-activated kinase-2. Cancer Res 2005;65(22):10431-10440 View Article PubMed/NCBI
  48. Xue G, Restuccia DF, Lan Q, Hynx D, Dirnhofer S, Hess D, et al. Akt/PKB-mediated phosphorylation of Twist1 promotes tumor metastasis via mediating cross-talk between PI3K/Akt and TGF-β signaling axes. Cancer Discov 2012;2(3):248-259 View Article PubMed/NCBI
  49. Mi XJ, Hou JG, Jiang S, Liu Z, Tang S, Liu XX, et al. Maltol Mitigates Thioacetamide-induced Liver Fibrosis through TGF-β1-mediated Activation of PI3K/Akt Signaling Pathway. J Agric Food Chem 2019;67(5):1392-1401 View Article PubMed/NCBI
  50. Wang R, Song F, Li S, Wu B, Gu Y, Yuan Y. Salvianolic acid A attenuates CCl(4)-induced liver fibrosis by regulating the PI3K/AKT/mTOR, Bcl-2/Bax and caspase-3/cleaved caspase-3 signaling pathways. Drug Des Devel Ther 2019;13:1889-1900 View Article PubMed/NCBI
  51. Shim JH, Xiao C, Paschal AE, Bailey ST, Rao P, Hayden MS, et al. TAK1, but not TAB1 or TAB2, plays an essential role in multiple signaling pathways in vivo. Genes Dev 2005;19(22):2668-2681 View Article PubMed/NCBI
  52. Lawan A, Bennett AM. Mitogen-Activated Protein Kinase Regulation in Hepatic Metabolism. Trends Endocrinol Metab 2017;28(12):868-878 View Article PubMed/NCBI
  53. Kyriakis JM, Avruch J. Mammalian MAPK signal transduction pathways activated by stress and inflammation: a 10-year update. Physiol Rev 2012;92(2):689-737 View Article PubMed/NCBI
  54. Manieri E, Sabio G. Stress kinases in the modulation of metabolism and energy balance. J Mol Endocrinol 2015;55(2):R11-R22 View Article PubMed/NCBI
  55. Choi ME, Ding Y, Kim SI. TGF-β signaling via TAK1 pathway: role in kidney fibrosis. Semin Nephrol 2012;32(3):244-252 View Article PubMed/NCBI
  56. Lee MK, Pardoux C, Hall MC, Lee PS, Warburton D, Qing J, et al. TGF-beta activates Erk MAP kinase signalling through direct phosphorylation of ShcA. EMBO J 2007;26(17):3957-3967 View Article PubMed/NCBI
  57. Muthusamy BP, Budi EH, Katsuno Y, Lee MK, Smith SM, Mirza AM, et al. ShcA Protects against Epithelial-Mesenchymal Transition through Compartmentalized Inhibition of TGF-β-Induced Smad Activation. PLoS Biol 2015;13(12):e1002325 View Article PubMed/NCBI
  58. Weston CR, Davis RJ. The JNK signal transduction pathway. Curr Opin Cell Biol 2007;19(2):142-149 View Article PubMed/NCBI
  59. Song IJ, Yang YM, Inokuchi-Shimizu S, Roh YS, Yang L, Seki E. The contribution of toll-like receptor signaling to the development of liver fibrosis and cancer in hepatocyte-specific TAK1-deleted mice. Int J Cancer 2018;142(1):81-91 View Article PubMed/NCBI
  60. Yang L, Inokuchi S, Roh YS, Song J, Loomba R, Park EJ, et al. Transforming growth factor-β signaling in hepatocytes promotes hepatic fibrosis and carcinogenesis in mice with hepatocyte-specific deletion of TAK1. Gastroenterology 2013;144(5):1042-1054.e4 View Article PubMed/NCBI
  61. Fabre T, Kared H, Friedman SL, Shoukry NH. IL-17A enhances the expression of profibrotic genes through upregulation of the TGF-β receptor on hepatic stellate cells in a JNK-dependent manner. J Immunol 2014;193(8):3925-3933 View Article PubMed/NCBI
  62. Westenberger G, Sellers J, Fernando S, Junkins S, Han SM, Min K, et al. Function of Mitogen-Activated Protein Kinases in Hepatic Inflammation. J Cell Signal 2021;2(3):172-180 PubMed/NCBI
  63. Yan Q, Pan L, Qi S, Liu F, Wang Z, Qian C, et al. RNF2 Mediates Hepatic Stellate Cells Activation by Regulating ERK/p38 Signaling Pathway in LX-2 Cells. Front Cell Dev Biol 2021;9:634902 View Article PubMed/NCBI
  64. Kagan P, Sultan M, Tachlytski I, Safran M, Ben-Ari Z. Both MAPK and STAT3 signal transduction pathways are necessary for IL-6-dependent hepatic stellate cells activation. PLoS One 2017;12(5):e0176173 View Article PubMed/NCBI
  65. Kelley RK, Gane E, Assenat E, Siebler J, Galle PR, Merle P, et al. A Phase 2 Study of Galunisertib (TGF-β1 Receptor Type I Inhibitor) and Sorafenib in Patients With Advanced Hepatocellular Carcinoma. Clin Transl Gastroenterol 2019;10(7):e00056 View Article PubMed/NCBI
  66. Chang Y, Jeong SW, Jang JY. Recent updates on pharmacologic therapy in non-alcoholic fatty liver disease. Clin Mol Hepatol 2024;30(1):129-133 View Article PubMed/NCBI
  67. Friedman S, Sanyal A, Goodman Z, Lefebvre E, Gottwald M, Fischer L, et al. Efficacy and safety study of cenicriviroc for the treatment of non-alcoholic steatohepatitis in adult subjects with liver fibrosis: CENTAUR Phase 2b study design. Contemp Clin Trials 2016;47:356-365 View Article PubMed/NCBI
  68. Gastaldelli A. Is it necessary to target lipid metabolism in different organs for effective treatment of NASH?-the results of the Pan-PPAR Lanifibranor trial. Hepatobiliary Surg Nutr 2022;11(3):481-484 View Article PubMed/NCBI
  69. Harrison SA, Bedossa P, Guy CD, Schattenberg JM, Loomba R, Taub R, et al. A Phase 3, Randomized, Controlled Trial of Resmetirom in NASH with Liver Fibrosis. N Engl J Med 2024;390(6):497-509 View Article PubMed/NCBI
  70. Harrison SA, Abdelmalek MF, Caldwell S, Shiffman ML, Diehl AM, Ghalib R, et al. Simtuzumab Is Ineffective for Patients With Bridging Fibrosis or Compensated Cirrhosis Caused by Nonalcoholic Steatohepatitis. Gastroenterology 2018;155(4):1140-1153 View Article PubMed/NCBI
  71. Dooley S, ten Dijke P. TGF-β in progression of liver disease. Cell Tissue Res 2012;347(1):245-256 View Article PubMed/NCBI
  72. Derynck R, Turley SJ, Akhurst RJ. TGFβ biology in cancer progression and immunotherapy. Nat Rev Clin Oncol 2021;18(1):9-34 View Article PubMed/NCBI
  73. Akhurst RJ. Targeting TGF-β Signaling for Therapeutic Gain. Cold Spring Harb Perspect Biol 2017;9(10):a022301 View Article PubMed/NCBI
  74. Hodson EM, Sinha A, Cooper TE. Interventions for focal segmental glomerulosclerosis in adults. Cochrane Database Syst Rev 2022;2(2):CD003233 View Article PubMed/NCBI
  75. Formenti SC, Hawtin RE, Dixit N, Evensen E, Lee P, Goldberg JD, et al. Baseline T cell dysfunction by single cell network profiling in metastatic breast cancer patients. J Immunother Cancer 2019;7(1):177 View Article PubMed/NCBI
  76. Grehn F, Holló G, Khaw P, Overton B, Wilson R, Vogel R, et al. Factors affecting the outcome of trabeculectomy: an analysis based on combined data from two phase III studies of an antibody to transforming growth factor beta2, CAT-152. Ophthalmology 2007;114(10):1831-1838 View Article PubMed/NCBI
  77. Denton CP, Merkel PA, Furst DE, Khanna D, Emery P, Hsu VM, et al. Recombinant human anti-transforming growth factor beta1 antibody therapy in systemic sclerosis: a multicenter, randomized, placebo-controlled phase I/II trial of CAT-192. Arthritis Rheum 2007;56(1):323-333 View Article PubMed/NCBI
  78. Ezquerro IJ, Lasarte JJ, Dotor J, Castilla-Cortázar I, Bustos M, Peñuelas I, et al. A synthetic peptide from transforming growth factor beta type III receptor inhibits liver fibrogenesis in rats with carbon tetrachloride liver injury. Cytokine 2003;22(1-2):12-20 View Article PubMed/NCBI
  79. Herbertz S, Sawyer JS, Stauber AJ, Gueorguieva I, Driscoll KE, Estrem ST, et al. Clinical development of galunisertib (LY2157299 monohydrate), a small molecule inhibitor of transforming growth factor-beta signaling pathway. Drug Des Devel Ther 2015;9:4479-4499 View Article PubMed/NCBI
  80. Naruse T, Aoki M, Fujimoto N, Arase S, Oura H, Ueda Y, et al. Novel ALK5 inhibitor TP0427736 reduces TGF-β induced growth inhibition in human outer root sheath cells and elongates anagen phase in mouse hair follicles. Pharmacol Rep 2017;69(3):485-491 View Article PubMed/NCBI
  81. Fransvea E, Angelotti U, Antonaci S, Giannelli G. Blocking transforming growth factor-beta up-regulates E-cadherin and reduces migration and invasion of hepatocellular carcinoma cells. Hepatology 2008;47(5):1557-1566 View Article PubMed/NCBI
  82. He X, Guo X, Zhang H, Kong X, Yang F, Zheng C. Mechanism of action and efficacy of LY2109761, a TGF-β receptor inhibitor, targeting tumor microenvironment in liver cancer after TACE. Oncotarget 2018;9(1):1130-1142 View Article PubMed/NCBI
  83. Desgraves JF, Mendez Valdez MJ, Chandar J, Gurses ME, Henderson L, Castro JR, et al. Antisense Oligonucleotides for Rapid Translation of Gene Therapy in Glioblastoma. Cancers (Basel) 2024;16(10):1944 View Article PubMed/NCBI
  84. Seniutkin O, Furuya S, Luo YS, Cichocki JA, Fukushima H, Kato Y, et al. Effects of pirfenidone in acute and sub-chronic liver fibrosis, and an initiation-promotion cancer model in the mouse. Toxicol Appl Pharmacol 2018;339:1-9 View Article PubMed/NCBI
  85. Cai X, Liu X, Xie W, Ma A, Tan Y, Shang J, et al. Hydronidone for the Treatment of Liver Fibrosis Related to Chronic Hepatitis B: A Phase 2 Randomized Controlled Trial. Clin Gastroenterol Hepatol 2023;21(7):1893-1901.e7 View Article PubMed/NCBI
  86. Armendáriz-Borunda J, Islas-Carbajal MC, Meza-García E, Rincón AR, Lucano S, Sandoval AS, et al. A pilot study in patients with established advanced liver fibrosis using pirfenidone. Gut 2006;55(11):1663-1665 View Article PubMed/NCBI
  87. Faivre S, Santoro A, Kelley RK, Gane E, Costentin CE, Gueorguieva I, et al. Novel transforming growth factor beta receptor I kinase inhibitor galunisertib (LY2157299) in advanced hepatocellular carcinoma. Liver Int 2019;39(8):1468-1477 View Article PubMed/NCBI
  88. Ahn JH, Lee J, Park C, Beom SH, Kim SH, Lee YH, et al. Clinical Activity of TGF-β Inhibitor Vactosertib in Combination with Imatinib in Desmoid Tumors: A Multicenter Phase Ib/II Study. Clin Cancer Res 2024;30(8):1457-1465 View Article PubMed/NCBI
  89. King TE, Bradford WZ, Castro-Bernardini S, Fagan EA, Glaspole I, Glassberg MK, et al. A phase 3 trial of pirfenidone in patients with idiopathic pulmonary fibrosis. N Engl J Med 2014;370(22):2083-2092 View Article PubMed/NCBI
  90. Nagaraj NS, Datta PK. Targeting the transforming growth factor-beta signaling pathway in human cancer. Expert Opin Investig Drugs 2010;19(1):77-91 View Article PubMed/NCBI
  91. Zhang S, Sun WY, Wu JJ, Wei W. TGF-β signaling pathway as a pharmacological target in liver diseases. Pharmacol Res 2014;85:15-22 View Article PubMed/NCBI
  92. Khaw P, Grehn F, Holló G, Overton B, Wilson R, Vogel R, et al. A phase III study of subconjunctival human anti-transforming growth factor beta(2) monoclonal antibody (CAT-152) to prevent scarring after first-time trabeculectomy. Ophthalmology 2007;114(10):1822-1830 View Article PubMed/NCBI
  93. Hill C, Flyvbjerg A, Rasch R, Bak M, Logan A. Transforming growth factor-beta2 antibody attenuates fibrosis in the experimental diabetic rat kidney. J Endocrinol 2001;170(3):647-651 View Article PubMed/NCBI
  94. Henderson NC, Arnold TD, Katamura Y, Giacomini MM, Rodriguez JD, McCarty JH, et al. Targeting of αv integrin identifies a core molecular pathway that regulates fibrosis in several organs. Nat Med 2013;19(12):1617-1624 View Article PubMed/NCBI
  95. Chia ZJ, Cao YN, Little PJ, Kamato D. Transforming growth factor-β receptors: versatile mechanisms of ligand activation. Acta Pharmacol Sin 2024;45(7):1337-1348 View Article PubMed/NCBI
  96. Sharip A, Kunz J. Mechanosignaling via Integrins: Pivotal Players in Liver Fibrosis Progression and Therapy. Cells 2025;14(4):266 View Article PubMed/NCBI
  97. Luangmonkong T, Suriguga S, Bigaeva E, Boersema M, Oosterhuis D, de Jong KP, et al. Evaluating the antifibrotic potency of galunisertib in a human ex vivo model of liver fibrosis. Br J Pharmacol 2017;174(18):3107-3117 View Article PubMed/NCBI
  98. van Beuge MM, Prakash J, Lacombe M, Post E, Reker-Smit C, Beljaars L, et al. Enhanced effectivity of an ALK5-inhibitor after cell-specific delivery to hepatic stellate cells in mice with liver injury. PLoS One 2013;8(2):e56442 View Article PubMed/NCBI
  99. Jin M, Seed RI, Cai G, Shing T, Wang L, Ito S, et al. Dynamic allostery drives autocrine and paracrine TGF-β signaling. Cell 2024;187(22):6200-6219.e23 View Article PubMed/NCBI
  100. Bellani S, Molyneaux PL, Maher TM, Spagnolo P. Potential of αvβ6 and αvβ1 integrin inhibition for treatment of idiopathic pulmonary fibrosis. Expert Opin Ther Targets 2024;28(7):575-585 View Article PubMed/NCBI
  101. Loiselle AE, Yukata K, Geary MB, Kondabolu S, Shi S, Jonason JH, et al. Development of antisense oligonucleotide (ASO) technology against Tgf-β signaling to prevent scarring during flexor tendon repair. J Orthop Res 2015;33(6):859-866 View Article PubMed/NCBI
  102. Ali S, Rehman MU, Yatoo AM, Arafah A, Khan A, Rashid S, et al. TGF-β signaling pathway: Therapeutic targeting and potential for anti-cancer immunity. Eur J Pharmacol 2023;947:175678 View Article PubMed/NCBI
  103. Zhao Z, Dong H, Li B, Shen B, Guo Y, Gu T, et al. [Hydroxynitone suppresses hepatic stellate cell activation by inhibiting TGF-β1 phosphorylation to alleviate CCl(4)-induced liver fibrosis in rats]. Nan Fang Yi Ke Da Xue Xue Bao 2022;42(10):1511-1516 View Article PubMed/NCBI
  104. El-Haggar SM, Mostafa TM. Comparative clinical study between the effect of fenofibrate alone and its combination with pentoxifylline on biochemical parameters and liver stiffness in patients with non-alcoholic fatty liver disease. Hepatol Int 2015;9(3):471-479 View Article PubMed/NCBI
  105. Desmoulière A, Xu G, Costa AM, Yousef IM, Gabbiani G, Tuchweber B. Effect of pentoxifylline on early proliferation and phenotypic modulation of fibrogenic cells in two rat models of liver fibrosis and on cultured hepatic stellate cells. J Hepatol 1999;30(4):621-631 View Article PubMed/NCBI
  106. Lopez-de la Mora DA, Sanchez-Roque C, Montoya-Buelna M, Sanchez-Enriquez S, Lucano-Landeros S, Macias-Barragan J, et al. Role and New Insights of Pirfenidone in Fibrotic Diseases. Int J Med Sci 2015;12(11):840-847 View Article PubMed/NCBI
  107. Atanasov AG, Zotchev SB, Dirsch VM, Supuran CT, International Natural Product Sciences Taskforce. Natural products in drug discovery: advances and opportunities. Nat Rev Drug Discov 2021;20(3):200-216 View Article PubMed/NCBI
  108. Hamza AA, Lashin FM, Gamel M, Hassanin SO, Abdalla Y, Amin A. Hawthorn Herbal Preparation from Crataegus oxyacantha Attenuates In Vivo Carbon Tetrachloride -Induced Hepatic Fibrosis via Modulating Oxidative Stress and Inflammation. Antioxidants (Basel) 2020;9(12):1173 View Article PubMed/NCBI
  109. Li WQ, Liu WH, Qian D, Liu J, Zhou SQ, Zhang L, et al. Traditional Chinese medicine: An important source for discovering candidate agents against hepatic fibrosis. Front Pharmacol 2022;13:962525 View Article PubMed/NCBI
  110. Abdelhamid AM, Selim A, Zaafan MA. The Hepatoprotective Effect of Piperine Against Thioacetamide-Induced Liver Fibrosis in Mice: The Involvement of miR-17 and TGF-β/Smads Pathways. Front Mol Biosci 2021;8:754098 View Article PubMed/NCBI
  111. Chen H, Wang Y, Jiao FZ, Yang F, Li X, Wang LW. Sinomenine Attenuates Acetaminophen-Induced Acute Liver Injury by Decreasing Oxidative Stress and Inflammatory Response via Regulating TGF-β/Smad Pathway in vitro and in vivo. Drug Des Devel Ther 2020;14:2393-2403 View Article PubMed/NCBI
  112. Gressner OA, Lahme B, Rehbein K, Siluschek M, Weiskirchen R, Gressner AM. Pharmacological application of caffeine inhibits TGF-beta-stimulated connective tissue growth factor expression in hepatocytes via PPARgamma and SMAD2/3-dependent pathways. J Hepatol 2008;49(5):758-767 View Article PubMed/NCBI
  113. Choi JH, Jin SW, Choi CY, Kim HG, Lee GH, Kim YA, et al. Capsaicin Inhibits Dimethylnitrosamine-Induced Hepatic Fibrosis by Inhibiting the TGF-β1/Smad Pathway via Peroxisome Proliferator-Activated Receptor Gamma Activation. J Agric Food Chem 2017;65(2):317-326 View Article PubMed/NCBI
  114. Yu JL, Li JH, Chengz RG, Ma YM, Wang XJ, Liu JC. Effect of matrine on transforming growth factor β1 and hepatocyte growth factor in rat liver fibrosis model. Asian Pac J Trop Med 2014;7(5):390-393 View Article PubMed/NCBI
  115. Balta C, Herman H, Boldura OM, Gasca I, Rosu M, Ardelean A, et al. Chrysin attenuates liver fibrosis and hepatic stellate cell activation through TGF-β/Smad signaling pathway. Chem Biol Interact 2015;240:94-101 View Article PubMed/NCBI
  116. Kang R, Tian W, Cao W, Sun Y, Zhang HN, Feng YD, et al. Ligustroflavone ameliorates CCl(4)-induced liver fibrosis through down-regulating the TGF-β/Smad signaling pathway. Chin J Nat Med 2021;19(3):170-180 View Article PubMed/NCBI
  117. Wang R, Zhang H, Wang Y, Song F, Yuan Y. Inhibitory effects of quercetin on the progression of liver fibrosis through the regulation of NF-кB/IкBα, p38 MAPK, and Bcl-2/Bax signaling. Int Immunopharmacol 2017;47:126-133 View Article PubMed/NCBI
  118. Hu Z, Qin F, Gao S, Zhen Y, Huang D, Dong L. Paeoniflorin exerts protective effect on radiation-induced hepatic fibrosis in rats via TGF-β1/Smads signaling pathway. Am J Transl Res 2018;10(3):1012-1021 PubMed/NCBI
  119. Lin L, Li R, Cai M, Huang J, Huang W, Guo Y, et al. Andrographolide Ameliorates Liver Fibrosis in Mice: Involvement of TLR4/NF-κB and TGF-β1/Smad2 Signaling Pathways. Oxid Med Cell Longev 2018;2018:7808656 View Article PubMed/NCBI
  120. Ran S, Song L, Yang H, Yu J, Zhen Y, Liu Q. Piperine alleviates nonalcoholic steatohepatitis by inhibiting NF-κB-mediated hepatocyte pyroptosis. PLoS One 2024;19(3):e0301133 View Article PubMed/NCBI
  121. Fan H, Tu T, Zhang X, Yang Q, Liu G, Zhang T, et al. Sinomenine attenuates alcohol-induced acute liver injury via inhibiting oxidative stress, inflammation and apoptosis in mice. Food Chem Toxicol 2022;159:112759 View Article PubMed/NCBI
  122. Wang H, Guan W, Yang W, Wang Q, Zhao H, Yang F, et al. Caffeine inhibits the activation of hepatic stellate cells induced by acetaldehyde via adenosine A2A receptor mediated by the cAMP/PKA/SRC/ERK1/2/P38 MAPK signal pathway. PLoS One 2014;9(3):e92482 View Article PubMed/NCBI
  123. Tao L, Yang G, Sun T, Jie Tao, Zhu C, Yu H, et al. Capsaicin receptor TRPV1 maintains quiescence of hepatic stellate cells in the liver via recruitment of SARM1. J Hepatol 2023;78(4):805-819 View Article PubMed/NCBI
  124. Miao J, Cui HT, Wang L, Guo LY, Wang J, Li P, et al. [Effects of evodiamine on carbon tetrachloride-induced liver fibrosis mice based on modulating gut microbiota]. Zhonghua Lao Dong Wei Sheng Zhi Ye Bing Za Zhi 2021;39(6):401-406 View Article PubMed/NCBI
  125. Lin Y, He F, Wu L, Xu Y, Du Q. Matrine Exerts Pharmacological Effects Through Multiple Signaling Pathways: A Comprehensive Review. Drug Des Devel Ther 2022;16:533-569 View Article PubMed/NCBI
  126. Gao C, Zhang H, Nie L, He K, Li P, Wang X, et al. Chrysin prevents inflammation-coinciding liver steatosis via AMPK signalling. J Pharm Pharmacol 2023;75(8):1086-1099 View Article PubMed/NCBI
  127. Wu L, Zhang Q, Mo W, Feng J, Li S, Li J, et al. Quercetin prevents hepatic fibrosis by inhibiting hepatic stellate cell activation and reducing autophagy via the TGF-β1/Smads and PI3K/Akt pathways. Sci Rep 2017;7(1):9289 View Article PubMed/NCBI
  128. Wang SW, Lan T, Chen HF, Sheng H, Xu CY, Xu LF, et al. Limonin, an AMPK Activator, Inhibits Hepatic Lipid Accumulation in High Fat Diet Fed Mice. Front Pharmacol 2022;13:833705 View Article PubMed/NCBI
  129. Cabrera D, Wree A, Povero D, Solís N, Hernandez A, Pizarro M, et al. Andrographolide Ameliorates Inflammation and Fibrogenesis and Attenuates Inflammasome Activation in Experimental Non-Alcoholic Steatohepatitis. Sci Rep 2017;7(1):3491 View Article PubMed/NCBI
  130. Lan T, Li P, Zhang SJ, Liu SY, Zeng XX, Chai F, et al. Paeoniflorin promotes PPARγ expression to suppress HSCs activation by inhibiting EZH2-mediated histone H3K27 trimethylation. Phytomedicine 2024;128:155477 View Article PubMed/NCBI
  131. Mondal A, Gandhi A, Fimognari C, Atanasov AG, Bishayee A. Alkaloids for cancer prevention and therapy: Current progress and future perspectives. Eur J Pharmacol 2019;858:172472 View Article PubMed/NCBI
  132. Yang D, Li L, Qian S, Liu L. Evodiamine ameliorates liver fibrosis in rats via TGF-β1/Smad signaling pathway. J Nat Med 2018;72(1):145-154 View Article PubMed/NCBI
  133. Sánchez-Crisóstomo I, Fernández-Martínez E, Cariño-Cortés R, Betanzos-Cabrera G, Bobadilla-Lugo RA. Phytosterols and Triterpenoids for Prevention and Treatment of Metabolic-related Liver Diseases and Hepatocellular Carcinoma. Curr Pharm Biotechnol 2019;20(3):197-214 View Article PubMed/NCBI
  134. Li S, Gan L, Tian YJ, Tian Y, Fan RZ, Huang D, et al. Presegetane diterpenoids from Euphorbia sieboldiana as a new type of anti-liver fibrosis agents that inhibit TGF-β/Smad signaling pathway. Bioorg Chem 2021;114:105222 View Article PubMed/NCBI

About this Article

Cite this article
Zhu W, Cui Y, Qiu J, Zhang X, Gao Y, Shang Z, et al. Exploring the Therapeutic Potential of TGF-β Inhibitors for Liver Fibrosis: Targeting Multiple Signaling Pathways. J Clin Transl Hepatol. 2025;13(7):588-598. doi: 10.14218/JCTH.2025.00029.
Copy        Export to RIS        Export to EndNote
Article History
Received Revised Accepted Published
January 16, 2025 June 21, 2025 July 3, 2025 July 15, 2025
DOI http://dx.doi.org/10.14218/JCTH.2025.00029
  • Journal of Clinical and Translational Hepatology
  • pISSN 2225-0719
  • eISSN 2310-8819
Back to Top

Exploring the Therapeutic Potential of TGF-β Inhibitors for Liver Fibrosis: Targeting Multiple Signaling Pathways

Wanchun Zhu, Yu Cui, Jiahao Qiu, Xin Zhang, Yueqiu Gao, Zhi Shang, Lingying Huang
  • Reset Zoom
  • Download TIFF