Introduction

Liver cancer, especially hepatocellular carcinoma(HCC), is the fifth most common cancer worldwide and the third leading cause of cancer mortality.1 Asia is a major region for liver cancer, and Asia reported 72.5% of the world’s cases in 2020.2 Risk factors include Hepatitis B virus (HBV), Hepatitis C virus (HCV), hepatic steatosis, and others.3,4 HCC is frequently characterized by delayed diagnosis and unfavorable prognosis, primarily attributable to its asymptomatic nature or nonspecific clinical manifestations during early stages.

HCC develops secondary to chronic hepatic disorders through complex pathogenetic mechanisms.5 The development of liver inflammation is frequently associated with chronic liver injury, subsequently leading to the damage of the epithelial or endothelial barrier, the release of inflammatory cytokines, the production of transforming growth factor-β (TGF-β) by macrophages, the overproduction of extracellular matrix, and the formation of a fibrous scar, which progresses to hepatic fibrosis (HF).6 Persistent HF injury is pathologically characterized mostly by three interconnected processes: hepatocyte apoptosis, sustained activation of inflammatory cascades, and aberrant mobilization of hematopoietic stem cells. These pathological alterations collectively drive a cascade of molecular events, synergistically promoting hepatocarcinogenesis and malignant transformation, ultimately leading to HCC.7,8

Viral infections are the primary pathogenic drivers in this process and represent the predominant factor leading to HCC. Anti-inflammatory and antiviral agents play a crucial role in disease management by simultaneously modulating inflammatory responses and suppressing viral replication, effectively attenuating the progression of HF and potentially preventing the malignant transformation to HCC.9 However, orthotopic liver transplantation remains the sole curative intervention for individuals with advanced-stage cirrhosis.10 Consequently, the ongoing pursuit of novel therapeutic strategies remains an imperative focus in contemporary clinical research.

DHA, a sesquiterpene-lactone derivative, is a bioactive phytochemical compound isolated from the medicinal plant Artemisia annua. Extensive clinical studies have demonstrated its efficacy as a potent and rapid-acting antimalarial agent.11 In addition to its antimalarial effects, DHA has a wide range of pharmacological activities. DHA demonstrates broad-spectrum antitumor activity against diverse malignancies, including but not limited to pancreatic, prostate, cervical, hepatic, and neuroblastoma cancers. The compound mediates its anticancer effects through multiple mechanisms, including cell cycle modulation, tumor angiogenesis suppression, and cancer cell apoptosis.12–15

Although extensive research has explored the therapeutic potential of DHA across various liver diseases, its therapeutic role across the disease continuum-from hepatitis and HF to HCC-remains unexplored. This review systematically addresses this critical knowledge gap by elucidating the mechanistic role of DHA in the prevention and treatment of hepatocarcinogenesis.

The Trilogy of “Hepatitis-HF-HCC”

Viral hepatitis primarily represents a major etiological factor contributing to the development and progression of HF and, ultimately, HCC.16 Notably, HBV infection has stood as a dominant oncogenic driver, significantly contributing to the initiation and progression of HCC through both direct and indirect mechanisms.17 These stress conditions activate critical signaling pathways, particularly signal transducer and activator of transcription 3 (STAT3) and NF-κB, which trigger the formation of inflammasomes and initiate inflammatory cascades. Furthermore, in chronic viral hepatitis, persistent reactive oxygen species (ROS) accumulation exacerbates hepatic injury through STAT3/NF-κB-mediated dysregulation, leading to enhanced lipid peroxidation and subsequent development of hepatic steatosis.18,19 Hepatic Stellate Cells (HSCs) are the core effector cells of HF. In a normal liver, HSCs are quiescent and store vitamin A lipid droplets (LDs). The persistent inflammatory microenvironment facilitates the activation of HSCs by modulating the TGF-β, platelet-derived growth factor-β receptor (PDGF) signaling pathways, oxidative stress, dysregulated miRNA expression, and inflammatory cytokines, including tumor necrosis factor-α (TNF-α), interleukin-6 (IL-6), and interleukin-1β (IL-1β). These inflammatory cytokines activate HSCs, transforming them into myofibroblasts that secrete large amounts of ECM components. The upregulation of tissue inhibitors of metalloproteinases inhibits extracellular matrix (ECM) degradation, resulting in fibrotic deposition.20–24 Furthermore, activated HSCs can also promote neovascularization through the secretion of angiopoietin, thereby creating a microenvironment conducive to HCC metastasis, invasion, and metastatic progression25 (Figure 1).

Figure 1 During the hepatitis stage, viral infection or metabolic damage triggers hepatocyte necrosis and inflammatory responses, activating Kupffer cells to release pro-inflammatory cytokines like TNF-α and IL-6. This process also generates ROS, leading to mitochondrial dysfunction and hepatocyte injury. In the liver fibrosis stage, inflammatory cytokines activate HSCs, transforming them into myofibroblasts that secrete large amounts of ECM components. The upregulation of tissue inhibitors of metalloproteinases inhibits ECM degradation, resulting in fibrotic deposition. During the HCC stage, the persistent inflammatory and fibrotic microenvironment promotes genetic mutations and epigenetic alterations, activating oncogenic signaling pathways such as PI3K/Akt/mTOR and Wnt/β-catenin. Additionally, HSCs can secrete factors like VEGF to promote tumor angiogenesis, driving the initiation and progression of HCC.

Phase I: DHA Prevention of Hepatitis
DHA Inhibits Inflammatory Cytokines in Alcoholic Hepatitis

The advancement of inflammatory processes in alcoholic liver disease can lead to hepatitis, HF, and ultimately HCC.26,27 In a mouse model of alcoholic fatty liver induced by Red Star wine, intraperitoneal injection of 7 mg/kg DHA significantly attenuates alcohol-induced elevations in serum levels of TNF-α, interleukin-8 (IL-8), and IL-6 while also mitigating hepatic damage. In another model of ethanol-induced alcoholic fatty liver disease, DHA restored ethanol-impaired lipin-1β function, leading to reduced expression of SREBP-1c and FAS, along with upregulation of PPARα and CPT1α in the liver. The findings suggest that DHA curbs lipin-1β nuclear import, lowers the Lpin1β/α ratio, and disrupts lipin-1-dependent lipogenesis, all while stimulating lipid breakdown. Concomitantly, DHA also inhibited the activation of endoplasmic reticulum (ER) stress.28 In vivo experiments further demonstrated that DHA attenuated the infiltration of inflammatory cells, notably those positive for CD45 and F4/80, in a model of alcoholic steatohepatitis.29 Farnesoid X Receptor (FXR) has been demonstrated to attenuate diet-induced steatohepatitis in mice via modulation of enterocyte fat absorption.30 By activating FXR, DHA reduces intracellular lipid accumulation and helps recover liver injury, thereby attenuating the alcohol-induced liver inflammatory response in mice. In a mouse model of alcohol-induced liver injury, DHA attenuated the alcohol-induced upregulation of hepatic lipogenic factors sterol regulatory element-binding protein-1c (SREBP-1c)31 and fatty acid synthase (FAS) via activation of FXR, and concurrently enhanced the expression of peroxisome proliferator-activated receptor alpha (PPAR-α) and carnitine palmitoyltransferase I (CPT1). These regulatory effects were abolished upon treatment with the FXR inhibitor Z-guggulsterone.32

DHA Regulates Lipid Metabolism

Yes-associated protein 1 (YAP1) is the key target in liver inflammation and lipid metabolism.33 An animal study showed that in a hepatocyte-specific Yap1 knockout mouse model, an arachidonic acid metabolic pathway was abnormally activated and accompanied by the infiltration of inflammatory cells. After intraperitoneal injection of 25 mg/kg DHA, it was found that DHA majorly targets 5-lipoxygenase and cyclooxygenase-2 expression to reduce the metabolism of arachidonic acid. DHA attenuated the increase in metabolites, including prostaglandin E1 and leukotrienes caused by activation of the arachidonic acid metabolic pathway, suppressed lipid vacuole accumulation, and reduced levels of triglyceride and perilipin-2. These results indicate that DHA mitigates hepatic inflammation through regulation of the YAP1-related metabolic pathway.34

HCV also constitutes a major causative factor of liver cancer globally. Existing studies have demonstrated a profound interdependency between lipoproteins and HCV replication: the virus not only exploits lipoproteins such as apolipoprotein E (apoE) for cellular attachment and entry but also coats itself in lipid-rich particles to form HCV-lipoviroparticles, while actively manipulating host lipid metabolism to facilitate its own replication. Although DHA can indeed modulate lipid metabolism, direct evidence regarding its ability to suppress HCV replication via regulation of apoE or LDLR expression remains limited. Nevertheless, this close relationship between lipid metabolism and HCV replication offers considerable scope for developing novel anti-HCV therapeutic strategies based on DHA.35,36

Phase II: Halting the Advancement of Cirrhosis by DHA
DHA Inhibits HSC Action and Cell Proliferation

HSC activation represents an essential pathological event in HF. Through HSC activation, the transformation of α-SMA-positive myofibroblasts has been increased, which increased the ECM accumulation, especially collagen I, and finally led to HSC proliferation.37 Some research showed that the platelet-derived growth factor-β receptor (PDGF-βR) is a potent proliferative cytokine, linked to cell proliferation in the pathogenesis of HF.38 In a rat model of bile duct ligation, administration of 14 mg/kg DHA markedly attenuated liver injury and HSCs activation by targeting the PDGF-βR/ERK signaling pathway. This intervention resulted in diminished hepatocyte degeneration, suppressed collagen deposition, reduced pseudolobule formation in liver tissue, and decreased activation of α-SMA-positive cells. Meanwhile, in vitro experiments, the PDGF-βR signal and the fibrosis marker protein in HSCs are observed to be changed. Furthermore, DHA also induced S-phase blockade in HSCs division to inhibit their proliferation and exert antifibrotic effects.39

DHA Inhibits HSC Cell Contraction and Fibrotic Portal Hypertension

Portal hypertension is the hallmark complication of decompensated HF.40,41 Hepatic injury triggers the activation of quiescent HSCs and HSCs contracted, contributing to portal hypertension.42 FXR is an effective regulatory factor of liver fibrosis and portal hypertension. Activation of the FXR receptor can effectively inhibit liver fibrosis-related portal hypertension.43 In vitro experiments indicated that by activating the FXR/S1PR2 signal, DHA at 20 μmol/L effectively inhibits the contraction of HSCs.44 These results were also demonstrated in the CCL4-induced rat model of liver cirrhosis and accompanied by TNF-α, IL-6, and NF-kB expression reduction.45

DHA Induces Ferroptosis in HSC Cells

Ferroptosis is defined as a non-apoptotic, iron-dependent form of regulated cell death, initiated by the accumulation of lipid peroxides. This process results in irreversible plasma membrane damage and elevated intracellular lipid ROS, distinguishing it mechanistically from other cell death modalities.46 A PDGF-BB-induced HSCs activation model study indicated that DHA exhibited antifibrotic effects by inducing ferroptosis. Specifically, 20μM DHA significantly upregulated Nuclear receptor coactivator 4 (NCOA4) expression and promoted the upregulation of iron death markers SLC11A2 and ACSL4, accompanied by several columns of iron death alterations (like lipid ROS generation or iron overload) to induce HSC cell death.47 Moreover, N6-methyladenosine (m6A) emerges as a potential therapeutic target in liver fibrosis. Interestingly, m6A exerts an antagonistic effect on DHA-induced ferroptosis in HSC and may provide a new basis for the mechanism of DHA-induced ferroptosis.48

DHA Triggers Both Autophagy and Apoptosis in HSCs

Apoptosis, a type of controlled cell suicide, inhibits HSCs’ activation and contributes to the amelioration of HF.49 In a bile duct-ligated rat model, intraperitoneal DHA (14 mg/kg) up-regulated cysteinyl aspartate specific proteinase (caspase)-3, the pro-apoptotic protein BCL2-Associated X (Bax), and down-regulated the anti-apoptotic protein B-cell lymphoma-2 (Bcl-2), exerting anti-fibrotic effects via PI3K/Akt pathway regulation.50

Autophagy is a self-degradation of damaged organelles and macromolecules within a cell, characterized by the formation of autophagosomes and lysosomes.51 20 μM DHA activates autophagy by activating the ROS-c-Jun N-terminal kinase (JNK)1/2 signal. Studies have shown that in PDGF-BB-induced HSCs, DHA increased the formation of autophagosomes and microtubule-associated protein 1A/1B-light chain 3 (LC3)-II. The detection of inflammatory cytokines in cell supernatants by ELISA revealed that phosphorylation of Autophagy Related 5 (pAtg5), like DHA, inhibited the release of interferon γ (IFN-γ), IL-4, and IL-6 and promoted the anti-inflammatory cytokines in activated HSCs to inhibit the inflammatory response.52

DHA Modulates Lipid Droplet Metabolism in HSCs

Loss of LDs is observed during activation of HSCs.53 Resting HSCs are enriched with vitamin A LDs, and retinol (vitamin A) stored in the LDs inhibits HSC activation-associated gene expression by binding to retinoic acid receptors.54 During the activation of HSCs, HSCs upregulate PPAR-α and generate adenosine triphosphate (ATP) to support their proliferation and ECM synthesis.55 IncRNA-H19 (RNA-H19) is a potential diagnostic biomarker and therapeutic target in liver fibrosis treatment.56

Reduction of LDs was found in activated HSC-LX2 cells. DHA at a concentration of 20 μM significantly inhibited the PI3K/Akt pathway activation, which inhibited the H19 expression, and ultimately inhibited the activation of the adenosine monophosphate-activated protein kinase (AMPK) signal, reducing the expression of carnitine palmitoyltransferase I A (CPT1A) and PPAR-α in activated HSC cells, reducing lipid oxidation levels in hematopoietic stem cells, restoring the content of cholesteryl esters, triglycerides, and retinyl esters in activated HSCs, thereby suppressing their activation. The same was demonstrated in the CCL4-rat model.57 Moreover, following DHA inhibits RNA-H19 expression, alcohol dehydrogenase III activity is elevated, promoting the conversion of retinol to retinaldehyde and thereby facilitating the storage of retinyl esters within lipid droplets, activating the retinoic acid signal, and restoring lipid droplet metabolism in activated HSCs.58 Recent studies have demonstrated that DHA reduces lipid droplet accumulation during HSC activation and restores HSC quiescence by targeting nuclear receptor subfamily 1 group D member.59

The regulatory effects of DHA on lipid metabolism in both hepatitis and HF involve multifaceted and interconnected mechanisms. The AMPK signaling pathway appears to serve as a central node through which DHA modulates lipid homeostasis in both pathological contexts. During hepatitis, DHA attenuates endoplasmic reticulum (ER) stress by targeting the lipin-1 signaling pathway. Considering the established crosstalk between ER stress and AMPK, it is plausible that DHA fine-tunes AMPK activity indirectly via the lipin-1–ER stress axis, thereby contributing to its lipid-modulating effects. During the HF stage, DHA activates nuclear receptor subfamily 1 group D member 1, which helps restore lipid droplet content in HSCs and inhibits their activation. In the HF stage, although not directly linked to lipid droplet metabolism in existing reports, DHA regulates key fatty acid synthases, including FAS, thereby modulating intracellular lipid droplet synthesis and metabolism and subsequently affecting HSC activation. Furthermore, in both stages, DHA regulated the expression of key proteins involved in lipid synthesis and oxidation—such as PPARα and CPT1A—thereby promoting lipid metabolic homeostasis.

Alternative Approaches Utilizing DHA for the Treatment of HF

DHA targets the VEGFA proteins via miR-29b-3 and inhibits the rate of CCL4-induced liver damage, such as hepatocellular disorders, inflammatory responses, and collagen accumulation. This was accompanied by a decrease in the expression of α-SMA and collagen type 1 alpha chain. This experiment also demonstrated that vascular endothelial growth factor receptor (VEGFR)2 regulates autophagy and degradation in HSC cells through the PI3K/Akt/mTOR/ UNC-51-like Kinase 1 (ULK1) signaling pathway. The anti-fibrotic effects of DHA may also be associated with this mechanism.60

Cellular senescence represents a stress-induced, irreversible state primarily regulated by the tumor suppressor proteins p53 and p16INK4a. DHA targeted GATA-binding factor 6 (GATA6) to promote the senescence markers p53, p16, p21, and Hmga1 expression in SA-β-Gal-positive HSC cells, and DHA-induced senescent HSC cell aggregation in rat fibrotic livers, and the senescence markers were found in an in vivo CCL4-induced animal model. In addition, this study also found that DHA at a concentration of 20 μM was effective in inhibiting p62 accumulation, inducing autophagosome formation, and disrupting the p62-GATA6 interaction, thereby promoting cellular senescence in HSC cells and inhibiting HF.61

YAP1: A Potential Therapeutic Target of DHA in HF

YAP1, a key effector of the Hippo signaling pathway, becomes rapidly activated upon hepatic stellate cell (HSC) stimulation—whether induced by CCl4 in mice or by other means in vitro. This activation is characterized by nuclear translocation and upregulated expression of YAP1 target genes, including Ankrd1 (cardiac ankyrin repeat protein) and Ctgf (connective tissue growth factor). Consistent with these findings, nuclear localization of YAP1 has also been observed in HSCs from human fibrotic livers. Moreover, both knockdown of YAP1 expression and pharmacological inhibition of YAP1 suppressed HSC activation in vitro, with pharmacological intervention also attenuating fibrotic progression in vivo.62 In a separate experiment, it was also demonstrated that modulation of YAP activity influences the phenotype of myofibroblast-like hepatic stellate cells (MF-HSCs). Specifically, YAP silencing suppressed the MF-HSC phenotype, reduced susceptibility to ferroptosis, and inhibited HSC activation.63

Phase III: DHA-Mediated Molecular Pathways in HCC Pathogenesis and Treatment
DHA Promotes Ferroptosis in HCC Cells

Activating transcription factor 4 (ATF4) was found to effectively activate the expression of solute carrier family 7a member 11, which is the small subunit of the cystine-glutamate antiporter (xCT) that has been demonstrated to increase the HCC vulnerability to ferroptosis, thereby suppressing the progression of HCC. In a mouse xenograft model of HCC, DHA at 100 mg/kg suppressed the expression of xCT, subsequently inhibiting ATF4 and promoting lipid peroxidation in HCC cells, ultimately leading to ferroptosis.64 Furthermore, the chemosensitivity of HCC to sorafenib (SRF) was enhanced through the ATF4-xCT signaling pathway.65 The synergistic use of DHA and SRF holds significant promise as a new therapeutic strategy for HCC treatment. Recently, a novel nanosheet loading DHA demonstrated multiple roles in inducing iron death, apoptosis, and immune activation driven by HCC, thereby proposing an innovative paradigm for its treatment.66 In HCC cell lines, consistent findings were observed. 15-Lipoxygenase (15-LO), a key enzyme catalyzing the peroxidation of polyunsaturated fatty acids, drives the accumulation of lipid peroxidation products in cell membranes when its activity is enhanced. DHA upregulates PEBP1 protein expression, which in turn indirectly activates 15-LO, elevating lipid peroxidation and ultimately inducing ferroptosis.67

The nuclear factor erythroid 2-related factor 2 (NRF2) signaling pathway may serve as an upstream regulator in DHA-induced ferroptosis in HSCs and HCC. NRF2 deficiency downregulates HECT and RLD domain containing E3 ubiquitin protein ligase 2 (HERC2) expression, enhances NCOA4 stability, and promotes ferritin degradation, leading to increased release of free iron. Specifically, NCOA4 recruits ferritin to autophagosomes for degradation, thereby facilitating ferritinophagy.68 Furthermore, NRF2 and ATF4 bind to the antioxidant response element (ARE) and amino acid response element (AARE) in the xCT promoter, synergistically upregulating xCT expression. This enhanced expression diminishes cellular sensitivity to ferroptosis.69 Accordingly, NRF2 likely functions as an upstream regulatory factor for both the NCOA4-dependent and ATF4-xCT signaling axes.

The JNK signaling pathway may represent an additional upstream regulatory mechanism involved in this process. In IL-1β-stimulated mouse chondrocytes, administration of the JNK inhibitor SP600125 downregulated NCOA4 expression, impeded autolysosome formation, and ultimately suppressed ferroptosis.70 β-Lapachone activates the JNK signaling pathway, resulting in upregulation of NCOA4 expression and promotion of ferritinophagy. Concurrently, it suppresses xCT transcription, leading to diminished cystine uptake and subsequent glutathione (GSH) depletion.71

DHA Triggers Both Apoptosis and Autophagy in HCC Cells

In HepG2 cell lines, DHA treatment results in elevated intracellular levels of ROS and Ca2+. Meanwhile, it upregulates DNA damage-inducible gene 153 protein expression, induces ER stress, modulates Bax and Bcl-2 expression, triggers apoptosis, and suppresses the proliferation of HepG2 cells.72 The transcription factor specificity protein 1, a ubiquitously expressed transcription factor, orchestrates fundamental biological processes ranging from cell cycle progression to survival signaling and apoptosis control.73 In SK-Hep-1 and AML12 cells, by inhibiting the transcription factor specificity protein 1 signaling pathway, DHA inhibited the X-linked inhibitor of apoptosis and increased the caspase-3, caspase-8, and caspase-9 expression to induce apoptosis and promote HCC cell destruction.74 Activator BH3-only molecules Bim trigger the activation of the BAX/BAK, leading to the induction of apoptosis.75 In Hep3B cell lines, Bim is involved in DHA-induced apoptosis in HCC cells, accompanied by activation of ROS and caspase, and found that Bcl-2 family member Bak is a dominant marker in the apoptosis process.76 In MHCC97-L cells, DHA inhibits proliferation and promotes apoptosis. Mediated by the JNK/NF-κB pathway, this effect consequently enhances the expression of TNF and executioner caspase.77 Another comprehensive in vitro and in vivo study further confirmed that DHA induces caspase and PARP activation, triggers G2/M cell cycle arrest, exerts potent cytotoxic effects, and significantly inhibits tumor growth in HCC cell lines.78

In HepG2215 cells, DHA activated the Absent in melanoma 2 inflammasome and promoted ROS generation, leading to enhanced autophagosome formation and increased conversion of LC3-II, ultimately inhibiting HCC cell proliferation.79 Meanwhile, DHA inhibited the PI3K/Akt signaling pathway, markedly upregulated LC3 expression, promoted AMPK phosphorylation, and elevated p62 expression to inhibit HCC cell migration.80

DHA Reduces Drug Resistance and Boosts Immunological Responsiveness in HCC Cells

Chemoresistance poses a significant clinical challenge in the management of HCC. P-glycoprotein represents a critical molecular target implicated in chemotherapeutic drug resistance.81 In mutant p53 (R248Q)-harbored HCC cells, DHA exerts its antitumor effects by targeting the ERK1/2-NF-κB pathway through direct binding to the p53 mutant protein, leading to suppression of P-glycoprotein expression, reduction of doxorubicin efflux, induction of apoptosis in Hep3B cell lines, and ultimately enhancing chemosensitivity to doxorubicin in HCC.82 In the N-nitroso diethylamine/1,4-Bis [2-(3,5-Dichloropyridyloxy)] benzene-induced anti-cisplatin (DDP) liver tumor mice model, intraperitoneal administration of DHA (25 mg/kg) demonstrated superior tumor growth inhibition compared to DDP treatment, with statistically significant differences observed, and the combination of DHA and DDP reduced TGF-β and enhanced the immune function of tumor-bearing mice.83 Similarly, co-treatment of HepG2 cells with 100 μM DHA and DDP upregulated the expression of cleaved caspase-3 and cleaved caspase-8 while modulating the protein levels of E-cadherin and N-cadherin. This combined treatment suppressed the proliferation and migration of HCC cells.84

SRF, a widely used therapeutic agent for liver cancer, faces persistent therapeutic obstacles, primarily attributable to prevalent drug resistance mechanisms. A TMT-based proteomic analysis demonstrated that the combination of DHA and SRF inhibits HCC cell proliferation.85 In HepG2 cells, the combined administration of DHA and SRF induced more substantial disruption of the tumor microenvironment in HepG2 hepatocellular carcinoma cells than either agent alone. DHA potentiated the suppressive effects of SRF on key energy metabolism processes—mitochondrial oxidative phosphorylation and glycolysis—and concurrently promoted ferroptosis.86 The combination of SRF with chemotherapeutic agents represents one of the earliest explored combination strategies. A Phase II clinical trial demonstrated that SRF plus capecitabine is a safe and effective conservative treatment for patients with Child–Pugh class A or B-7 cirrhosis. However, due to the small sample size, no meaningful conclusions could be drawn regarding its efficacy compared to sorafenib monotherapy. Treatment-related adverse events included neutropenia, thrombocytopenia, and abnormal liver function.87 Combining the novel immunotherapeutic agent R848 (a Toll-like receptor 7/8 agonist) with SRF elevated the infiltration of CD45⁺ immune cells and neutrophils, decreased the abundance of immunosuppressive cells, including Tregs and M2 macrophages, and inhibited the remodeling of the tumor immune microenvironment.88 Compared to the DHA–SRF combination, the primary advantage of combining SRF with immunotherapy lies in its ability to activate antitumor immune responses. However, this approach carries an increased risk of immune-related adverse events. In contrast, the DHA–SRF regimen primarily acts by directly inducing tumor cell death.

Immunotherapy has emerged as a promising therapeutic strategy and research focus in HCC treatment, improving the tumor microenvironment and helping to alleviate HCC drug resistance.89 An animal study indicated that DHA intraperitoneal injection of 50 mg/kg inhibited Cyclin-dependent kinases, increased intracellular ROS to induce immunogenic cell death, and remodeled the tumor microenvironment to inhibit tumor growth in hepatocellular carcinoma in HCC xenograft mice’s model.90

DHA Achieves Its Therapeutic Effects on HCC by Modulating the YAP1 Signaling Pathway

YAP1 regulates Plasminogen activator inhibitor-1 (PAI-1) transcription to promote HCC,91 and targets YAP1 has been shown to suppress tumor progression.92 Increased aerobic glycolysis is a critical driver of HCC progression and pathogenesis.93 DHA suppressed aerobic glycolysis in HepG2 and HEPG2215 cells by disrupting the YAP-Hypoxia-inducible factor (HIF)-1α complex formation, leading to reduced lactate production, decreased glycolytic flux, and diminished glycolytic capacity. This novel mechanism of aerobic glycolysis inhibition represents a promising therapeutic strategy for controlling HCC tumor proliferation.94 Treatment with 21.5 μM DHA in HepG2 and HepG2215 cells was shown to inhibit YAP1 activity, which subsequently suppressed the glycolytic response mediated by solute carrier family 2 member 1. This effect was achieved by down-regulating YAP1 promoter-binding proteins, GA-binding protein transcription factor subunit beta 1, and cAMP-responsive element binding protein 1.95

YAP1 knockdown in HepG2215 cells resulted in downregulation of Interleukin-18 (IL-18). Consistent with this finding, DHA treatment effectively suppressed YAP1 expression and subsequently reduced IL-18 levels in HepG2215 cells, a result that was later confirmed in in vivo models.96 The results above suggest that IL-18 represents a promising therapeutic target for DHA in HCC management. The FXR serves as a characteristic molecular marker during HCC development and tumorigenesis. DHA enhances FXR protein expression and reduces YAP1 protein expression to inhibit bile acid metabolism.97

In clinical practice, anti-programmed death 1 blockade has emerged as a cornerstone therapeutic strategy in HCC management.98 In both HepG2215 cells and their corresponding mouse xenograft models, DHA treatment significantly attenuated lipid droplet accumulation through inhibiting YAP1 expression. This was accompanied by an upregulation in the expression of perilipin-2, an adipocyte differentiation-related protein, consequently potentiating the therapeutic efficacy of anti-programmed death 1 immunotherapy.99 Moreover, DHA disrupts the YAP1-PD-L1 interaction in hepatic tumor cells while promoting CD8+ T cell infiltration, overcoming tumor immune evasion, and potentiating anti-PD-1 therapeutic efficacy in HCC.100

In summary, DHA suppresses the initiation and progression of HCC by modulating multiple pathways, including lipid metabolism, ferroptosis, and autophagy (Figure 2).

Figure 2 Mechanism of DHA in Hepatitis: DHA exerts its effects by inhibiting pro-inflammatory cytokines such as TNF-α, IL-6, and IL-8, as well as reducing the infiltration of inflammatory cells marked by CD45 positivity. Primarily, DHA targets the 5-lipoxygenase and cyclooxygenase-2, thereby reducing the metabolism of arachidonic acid and mitigating inflammatory responses. Mechanism of DHA in HF: DHA significantly alleviates liver injury and inhibits the activation of HSCs through the PDGF-βR/ERK signaling pathway. It reduces hepatocyte degeneration, suppresses collagen accumulation and pseudo-lobule formation in liver tissue, and decreases the transformation of α-SMA-positive cells. Additionally, DHA modulates the FXR to inhibit HSC contraction and downregulate the expression of TNF-α, IL-6, and NF-kB. Furthermore, DHA reverses hepatic fibrosis by inducing apoptosis, autophagy, and ferroptosis. Therapeutic Effects of DHA in HCC: DHA demonstrates anti-cancer effects by inhibiting cell proliferation, invasion, and migration. It induces apoptosis, autophagy, and ferroptosis, overcomes drug resistance in cancer cells, and enhances sensitivity to chemotherapeutic agents. (Arrows are primarily used to denote the directionality of biological processes, signal transduction and activation, transitions in cellular states, as well as drug-target interactions and their corresponding effects.).

Targeting Lipid Metabolism in the Hepatitis-to-HCC Progression: A Promising Therapeutic Strategy

During the hepatitis stage, DHA suppresses arachidonic acid metabolism, thereby reducing lipid accumulation. In the HF stage, it modulates lipid droplet metabolism in HSCs, influencing their activation and thus regulating HF progression. At the HCC stage, DHA acts through the YAP1 pathway to inhibit intracellular lipid droplet formation and disrupt the tumor microenvironment. Collectively, our findings highlight the modulation of lipid metabolism by DHA as a critical and consistent mechanism throughout the hepatitis-HF-HCC sequence, positioning it as a highly promising research avenue for curbing HCC progression.

During hepatitis infection, hepatitis viruses interact with LDs within the ER compartment. They enter the ER through lipoprotein assembly procession and secretion and produce apoE to facilitate the assembly and secretion of very low-density lipoproteins. This mechanism ultimately enhances the replication of HCV virions.101,102 It was also demonstrated that lipoprotein lipase treatment significantly reduced the apoE content within HCV particles. This treatment enhanced the binding of cell culture-derived HCV to the low-density lipoprotein receptor (LDLR). Subsequently, soluble LDLR inhibited the infectivity of such HCV, thereby suppressing its replication.103

Free cholesterol in HSCs is regulated through the sterol-regulatory element-binding protein negative feedback pathway. This results in elevated toll-like receptor 4 expression, sensitizing HSCs to TGF-β-induced activation. Consequently, cholesterol accumulation occurs, which further contributes to the activation of HSCs and accelerates liver fibrosis.104 Fatty acid binding protein 1 facilitates the upregulation of VEGFR in HCC and promotes HCC cell migration through the VEGFR2/steroid receptor coactivator signaling pathway.105 Moreover, oleate treatment activates the fatty acid-binding protein 5/HIF-1α signaling axis, driving lipid accumulation and enhancing cell proliferation in HCC cells.106 Lipid metabolism may emerge as a potential therapeutic strategy for DHA in HCC treatment. However, further research is required to elucidate the mechanisms by which DHA modulates lipid metabolism (Figure 3).

Figure 3 During the hepatitis stage, lipid metabolism dysregulation leads to excessive lipid accumulation within hepatocytes, triggering oxidative stress and inflammatory responses. This activates Kupffer cells and HSCs, promoting the release of inflammatory cytokines, which further damage hepatocytes and drive disease progression. In the liver fibrosis stage, the persistent accumulation of lipotoxic products activates HSCs, transforming them into myofibroblasts that secrete large amounts of ECM, leading to fibrosis. Concurrently, metabolic reprogramming reduces fatty acid oxidation and increases glycolysis, accelerating the fibrotic process. In the HCC stage, tumor cells meet their energy and biosynthetic demands for rapid proliferation by upregulating key enzymes involved in lipid synthesis and undergoing metabolic reprogramming. Additionally, activating lipid metabolism-related signaling pathways (like PI3K/Akt/mTOR) further promotes tumor growth and metastasis. Moreover, abnormal lipid metabolism supports HCC development by modulating the tumor microenvironment, such as promoting the polarization of tumor-associated macrophages.

Ferroptosis represents one of the key mechanisms through which DHA exerts its preventive and therapeutic effects against HCC. Previous studies have demonstrated that polyunsaturated fatty acids are critical drivers of ferroptosis. Upon peroxidation, polyunsaturated fatty acids suppress the expression of glutathione peroxidase 4 (GPX4), leading to lipid peroxide accumulation and ferroptosis induction.107–109 Emerging research findings indicate that during the process of ferroptosis, an increase in cell volume accompanied by the accumulation of lipid peroxides on the membrane has been observed, ultimately leading to cell lysis.110 DHA has been demonstrated to modulate lipid metabolism. In lung cancer cells, DHA enhances the production and intracellular accumulation of lipid peroxides, thereby inducing ferroptosis in cancer cells.111 It also promotes the accumulation of lipid ROS in glioblastoma and inhibits the expression of GPX4, accompanied by alterations in mitochondrial cristae structure, indicative of ferroptosis induction.112 A novel DHA-iron protein nanosensitizer has been developed, which significantly downregulates GPX4 expression, thereby enhancing lipid peroxidation and ultimately promoting ferroptosis.113

Novel Approaches to Enhance the Therapeutic Efficacy of DHA

DHA demonstrates superior pharmacological properties to artemisinin, including enhanced water solubility, better therapeutic efficacy, improved absorption, broader tissue distribution, faster metabolic clearance, higher efficiency, and reduced toxicity. Following intravenous administration, DHA concentrations reached peak levels within 25 minutes and were eliminated with a half-life of 30 to 60 minutes. After oral administration, the half-life of DHA averaged 0.5 to 1.5 hours, which is 2 to 3 times longer than that of artesunate.114,115

Currently, surgical resection, liver transplantation, and targeted therapy remain the primary treatment modalities for HCC. However, these approaches are inevitably associated with significant limitations, including high costs, drug resistance, limited efficacy, and substantial side effects. DHA can modulate multiple phenotypic responses, including cell proliferation and migration inhibition, reduced lipid metabolism, induced cellular autophagy, and apoptosis.

Despite its advantages over other artemisinin products in terms of pharmacological properties unique structural characteristics of DHA impose major constraints on its clinical translation for cancer treatment.116 Emerging studies have demonstrated that novel drug carriers, advanced delivery systems, or combination therapies can significantly enhance DHA sensitivity and utilization while reducing toxicity. Nanotechnology has emerged as a promising strategy for drug efficacy. SRF/DHA-loaded LDL-based lipid nanoparticles exhibit significantly enhanced affinity for LDLR overexpressed in HepG2 cells compared to DHA or SRF alone, enabling effective activation of programmed cell death in malignant cells.117 Fluorescent nanoparticles (CDs-DHA), engineered through the supramolecular assembly of carbon dots (CDs) and DHA, markedly enhanced the solubility and stability of DHA. These nanoparticles demonstrated the ability to inhibit glycolysis in HCC cells, effectively suppressing tumor growth.118 Moreover, a lipid nanoparticle featuring a lipid bilayer structure has been developed to enhance the synergistic therapeutic effects of DHA and chloroquine in the treatment of colon cancer. Nanomaterial-encapsulated DHA represents a novel and promising approach to HCC management119 (Figure 4).

Figure 4 Clinical Translation of DHA: DHA exhibits enhanced biological activities, such as superior water solubility and improved therapeutic efficacy. However, its unique structural properties result in poor water solubility and limited bioavailability. These challenges can be effectively addressed through nanotechnology, which enhances the therapeutic potential of DHA and provides a robust foundation for its future clinical translation.

Time-Dose Analysis of DHA

In the studies analyzed, demonstrating statistical significance (P < 0.05) at the experimental dosage and duration of DHA treatment were integrated into a comprehensive time-dose framework. In hepatitis treatment, intraperitoneal injections were delivered at doses spanning from 7 mg/kg to 25 mg/kg for 1 to 8 weeks. In the context of HF treatment, the dose range was similar, starting at 7 mg/kg and reaching up to 20 mg/kg for 4–8 weeks. In HCC treatment, intraperitoneal injections were administered at doses ranging from 20 to 50 mg/kg for 18 days to 4 weeks, with 25 mg/kg identified as the optimal dosage. Preclinical studies have demonstrated that DHA could attenuate the disease progression from hepatitis through fibrotic transformation to hepatocellular carcinoma. In the HF intervention phase, an intraperitoneal injection dose of 25 mg/kg over 4 weeks has been identified as optimal. For HCC treatment, doses ranging from 20 to 50 mg/kg administered over 4 to 8 weeks have shown therapeutic effectiveness. Collectively, these data demonstrate that DHA is a promising clinical candidate for HCC prevention and treatment. However, the potential of DHA in HCC prevention and treatment necessitates further validation through comprehensive clinical trials (Figure 5).

Figure 5 Scatter plot of time-dose interval analysis.

Conclusion

HCC is a multifactorial disease characterized by complex and diverse pathogenic mechanisms. Persistent inflammatory infiltrates during this progression foster a tumor-permissive microenvironment, facilitating HCC development. Throughout the progression of liver disease, DHA modulates key signaling pathways, including PI3K/Akt and interleukin cascades, which underlie its versatile role in reducing inflammation, alleviating fibrosis, and inhibiting tumor development.

During the inflammatory phase of hepatitis, DHA demonstrates anti-inflammatory properties through dual inhibition of the PI3K/Akt signaling cascade and interleukin-mediated pathways. As the disease progresses to HF, DHA exhibits multifaceted antifibrotic activity via: suppression of hepatic stellate cell activation, reduction of leukocyte infiltration, inhibition of aberrant hematopoietic stem cell expansion, induction of ferroptosis, and regulation of intestinal microbial ecology. During hepatocellular carcinoma development, DHA exerts oncostatin effects through: impairment of neoplastic cell proliferation and motility, activation of programmed cell death pathways, and reversal of chemoresistance to conventional agents, including cisplatin and chloroquine. Central to this disease continuum, lipid metabolism emerges as a critical pathological nexus, positioning DHA’s metabolic modulation as a strategic intervention point for HCC chemoprevention.

Although DHA demonstrates significant therapeutic potential in the context of HCC, current research lacks comprehensive in vitro and in vivo investigations into DHA’s dual effects on hepatitis viral suppression and lipid homeostasis modulation, which is critical for understanding its potential to halt HCC progression. In this review, we identify DHA as a therapeutic agent capable of exerting beneficial effects across the three-stage progression of hepatitis–HF–HCC. We also elaborate on the multi-target and multi-pathway mechanisms of DHA, highlighting the central role of lipid metabolism throughout its action in all three stages. By modulating lipid metabolism, DHA effectively attenuates inflammatory responses, suppresses HSC activation, promotes HCC cell death, and ultimately inhibits carcinogenesis. While existing studies have demonstrated DHA’s efficacy in suppressing HCC progression, current research remains predominantly confined to the preclinical stage. Clinical evidence regarding its therapeutic effects on HBV- and NASH-related HCC, as well as advanced HCC, remains scarce, underscoring the need for further validation. In summary, DHA represents a promising therapeutic agent for the hepatitis-to-hepatocellular carcinoma cascade, offering new avenues for treatment and hope for HCC patients.

Data Sharing Statement

The datasets used and/or analyzed during the current study are available from the corresponding author upon reasonable request.

Acknowledgment

This work was supported by the Xinglin Scholar Research Promotion Project of Chengdu University of TCM (Grant No. QJJJ2024005), the Joint Innovation Fund of Health Commission of Chengdu and Chengdu University of Traditional Chinese Medicine (grant nos. LH202402044 and WXLH202403008), Young Elite Scientists Sponsorship Program by CACM (CACM-(2024-QNRC2-A10), CACM-(2023-QNRC2-A01)), and the Science and Technology Project of Yibin City (2024SF007, 2024ZYY001, and 2024ZYY002).

Disclosure

Tingyao Wang and Wei Jiang contributed equally to this work and shared first authorship. The authors declare that they have no known competing financial interests or personal relationships that could have appeared to influence the work reported in this paper.

References

1. Yang JD, Hainaut P, Gores GJ, Amadou A, Plymoth A, Roberts LR. A global view of hepatocellular carcinoma: trends, risk, prevention and management. Nat Rev Gastroenterol Hepatol. 2019;16(10):589–604. doi:10.1038/s41575-019-0186-y

2. Zhang CH, Cheng Y, Zhang S, Fan J, Gao Q. Changing epidemiology of hepatocellular carcinoma in Asia. Liver Int. 2022;42(9):2029–2041. doi:10.1111/liv.15251

3. Center MM, Jemal A. International trends in liver cancer incidence rates. Cancer Epidemiol Biomarkers Prev. 2011;20(11):2362–2368. doi:10.1158/1055-9965.EPI-11-0643

4. Chacko S, Samanta S. “Hepatocellular carcinoma: a life-threatening disease”. Biomed Pharmacother. 2016;84:1679–1688. doi:10.1016/j.biopha.2016.10.078

5. Embade N, Millet O. Molecular determinants of chronic liver disease as studied by NMR-metabolomics. Curr Top Med Chem. 2017;17(24):2752–2766. doi:10.2174/1568026617666170707124539

6. Kisseleva T, Brenner D. Molecular and cellular mechanisms of liver fibrosis and its regression. Nat Rev Gastroenterol Hepatol. 2021;18(3):151–166. doi:10.1038/s41575-020-00372-7

7. Banerjee A, Farci P. Fibrosis and hepatocarcinogenesis: role of gene-environment interactions in liver disease progression. Int J Mol Sci. 2024;25(16):8641. doi:10.3390/ijms25168641

8. Jeng KS, Lu SJ, Wang CH, Chang CF. Liver fibrosis and inflammation under the control of ERK2. Int J Mol Sci. 2020;21(11). doi:10.3390/ijms21113796

9. Selitsky SR, Dinh TA, Toth CL, et al. Transcriptomic analysis of chronic hepatitis B and C and liver cancer reveals MicroRNA-mediated control of cholesterol synthesis programs. mBio. 2015;6(6):e01500–15. doi:10.1128/mBio.01500-15

10. Neong SF, Adebayo D, Wong F. An update on the pathogenesis and clinical management of cirrhosis with refractory ascites. Expert Rev Gastroenterol Hepatol. 2019;13(4):293–305. doi:10.1080/17474124.2018.1555469

11. Zhang S, Chen H, Gerhard GS. Heme synthesis increases artemisinin-induced radical formation and cytotoxicity that can be suppressed by superoxide scavengers. Chem Biol Interact. 2010;186(1):30–35. doi:10.1016/j.cbi.2010.03.021

12. Lu YY, Chen TS, Qu JL, Pan WL, Sun L, Wei XB. Dihydroartemisinin (DHA) induces caspase-3-dependent apoptosis in human lung adenocarcinoma ASTC-a-1 cells. J Biomed Sci. 2009;16(1):16. doi:10.1186/1423-0127-16-16

13. Li G, Ling M, Yu K, et al. Synergetic delivery of artesunate and isosorbide 5-mononitrate with reduction-sensitive polymer nanoparticles for ovarian cancer chemotherapy. J Nanobiotechnology. 2022;20(1):471. doi:10.1186/s12951-022-01676-3

14. Liu W, Zhou H, Lai W, et al. Artesunate induces melanoma cell ferroptosis and augments antitumor immunity through targeting Ido1. Cell Commun Signal. 2024;22(1):378. doi:10.1186/s12964-024-01759-8

15. Bai B, Wu F, Ying K, et al. Therapeutic effects of dihydroartemisinin in multiple stages of colitis-associated colorectal cancer. Theranostics. 2021;11(13):6225–6239. doi:10.7150/thno.55939

16. Ward EM, Sherman RL, Henley SJ, et al. Annual report to the nation on the status of cancer, featuring cancer in men and women age 20-49 years. J Natl Cancer Inst. 2019;111(12):1279–1297. doi:10.1093/jnci/djz106

17. Ringelhan M, McKeating JA, Protzer U. Correction to ‘Viral hepatitis and liver cancer’. Philos Trans R Soc Lond B Biol Sci. 2018;373(1737):20170339. doi:10.1098/rstb.2017.0339

18. Patel A, Harrison SA. Hepatitis C virus infection and nonalcoholic steatohepatitis. Gastroenterol Hepatol. 2012;8(5):305–312.

19. Mota S, Mendes M, Freitas N, Penque D, Coelho AV, Cunha C. Proteome analysis of a human liver carcinoma cell line stably expressing hepatitis delta virus ribonucleoproteins. J Proteomics. 2009;72(4):616–627. doi:10.1016/j.jprot.2008.12.003

20. Elpek GO. Cellular and molecular mechanisms in the pathogenesis of liver fibrosis: an update. World J Gastroenterol. 2014;20(23):7260–7276. doi:10.3748/wjg.v20.i23.7260

21. Roehlen N, Crouchet E, Baumert TF. Liver fibrosis: mechanistic concepts and therapeutic perspectives. Cells. 2020;9(4). doi:10.3390/cells9040875

22. Mohr R, Ozdirik B, Lambrecht J, et al. From liver cirrhosis to cancer: the role of micro-RNAs in hepatocarcinogenesis. Int J Mol Sci. 2021;22(3):1492. doi:10.3390/ijms22031492

23. Dhar D, Baglieri J, Kisseleva T, Brenner DA. Mechanisms of liver fibrosis and its role in liver cancer. Exp Biol Med. 2020;245(2):96–108. doi:10.1177/1535370219898141

24. Boulahtouf Z, Virzi A, Baumert TF, Verrier ER, Lupberger J. Signaling induced by chronic viral hepatitis: dependence and consequences. Int J Mol Sci. 2022;23(5):2787. doi:10.3390/ijms23052787

25. Matsuda M, Seki E. Hepatic stellate cell-macrophage crosstalk in Liver fibrosis and carcinogenesis. Semin Liver Dis. 2020;40(3):307–320. doi:10.1055/s-0040-1708876

26. Livero FA, Acco A. Molecular basis of alcoholic fatty liver disease: from incidence to treatment. Hepatol Res. 2016;46(1):111–123. doi:10.1111/hepr.12594

27. Gao B, Ahmad MF, Nagy LE, Tsukamoto H. Inflammatory pathways in alcoholic steatohepatitis. J Hepatol. 2019;70(2):249–259. doi:10.1016/j.jhep.2018.10.023

28. Chen X, Bian M, Jin H, et al. Dihydroartemisinin attenuates alcoholic fatty liver through regulation of lipin-1 signaling. IUBMB Life. 2019;71(11):1740–1750. doi:10.1002/iub.2113

29. Chen X, Bian M, Zhang C, et al. Dihydroartemisinin inhibits ER stress-mediated mitochondrial pathway to attenuate hepatocyte lipoapoptosis via blocking the activation of the PI3K/Akt pathway. Biomed Pharmacother. 2018;97:975–984. doi:10.1016/j.biopha.2017.11.010

30. Clifford BL, Sedgeman LR, Williams KJ, et al. FXR activation protects against NAFLD via bile-acid-dependent reductions in lipid absorption. Cell Metab. 2021;33(8):1671–1684e4. doi:10.1016/j.cmet.2021.06.012

31. Shimano H, Sato R. SREBP-regulated lipid metabolism: convergent physiology – divergent pathophysiology. Nat Rev Endocrinol. 2017;13(12):710–730. doi:10.1038/nrendo.2017.91

32. Xu W, Lu C, Yao L, Zhang F, Shao J, Zheng S. Dihydroartemisinin protects against alcoholic liver injury through alleviating hepatocyte steatosis in a farnesoid X receptor-dependent manner. Toxicol Appl Pharmacol. 2017;315:23–34. doi:10.1016/j.taap.2016.12.001

33. Balaz M, Becker AS, Balazova L, et al. Inhibition of mevalonate pathway prevents adipocyte browning in mice and men by affecting protein prenylation. Cell Metab. 2019;29(4):901–916e8. doi:10.1016/j.cmet.2018.11.017

34. Xue Y, Lu J, Liu Y, et al. Dihydroartemisinin modulated arachidonic acid metabolism and mitigated liver inflammation by inhibiting the activation of 5-LOX and COX-2. Heliyon. 2024;10(13):e33370. doi:10.1016/j.heliyon.2024.e33370

35. Andre P, Perlemuter G, Budkowska A, Brechot C, Lotteau V. Hepatitis C virus particles and lipoprotein metabolism. Semin Liver Dis. 2005;25(1):93–104. doi:10.1055/s-2005-864785

36. Bley H, Schobel A, Herker E. Whole lotta lipids-from HCV RNA replication to the mature viral particle. Int J Mol Sci. 2020;21(8):2888. doi:10.3390/ijms21082888

37. Iredale JP. Hepatic stellate cell behavior during resolution of liver injury. Semin Liver Dis. 2001;21(3):427–436. doi:10.1055/s-2001-17557

38. Chen W, Zhang Z, Yao Z, et al. Activation of autophagy is required for Oroxylin A to alleviate carbon tetrachloride-induced liver fibrosis and hepatic stellate cell activation. Int Immunopharmacol. 2018;56:148–155. doi:10.1016/j.intimp.2018.01.029

39. Chen Q, Chen L, Kong D, Shao J, Wu L, Zheng S. Dihydroartemisinin alleviates bile duct ligation-induced liver fibrosis and hepatic stellate cell activation by interfering with the PDGF-betaR/ERK signaling pathway. Int Immunopharmacol. 2016;34:250–258. doi:10.1016/j.intimp.2016.03.011

40. Iwakiri Y, Trebicka J. Portal hypertension in cirrhosis: pathophysiological mechanisms and therapy. JHEP Rep. 2021;3(4):100316. doi:10.1016/j.jhepr.2021.100316

41. Allaire M, Rudler M, Thabut D. Portal hypertension and hepatocellular carcinoma: des liaisons dangereuses. Liver Int. 2021;41(8):1734–1743. doi:10.1111/liv.14977

42. Thabut D, Shah V. Intrahepatic angiogenesis and sinusoidal remodeling in chronic liver disease: new targets for the treatment of portal hypertension? J Hepatol. 2010;53(5):976–980. doi:10.1016/j.jhep.2010.07.004

43. Verbeke L, Farre R, Trebicka J, et al. Obeticholic acid, a farnesoid X receptor agonist, improves portal hypertension by two distinct pathways in cirrhotic rats. Hepatology. 2014;59(6):2286–2298. doi:10.1002/hep.26939

44. Xu W, Lu C, Zhang F, Shao J, Zheng S. Dihydroartemisinin restricts hepatic stellate cell contraction via an FXR-S1PR2-dependent mechanism. IUBMB Life. 2016;68(5):376–387. doi:10.1002/iub.1492

45. Xu W, Lu C, Zhang F, Shao J, Yao S, Zheng S. Dihydroartemisinin counteracts fibrotic portal hypertension via farnesoid X receptor-dependent inhibition of hepatic stellate cell contraction. FEBS J. 2017;284(1):114–133. doi:10.1111/febs.13956

46. Tang D, Chen X, Kang R, Kroemer G. Ferroptosis: molecular mechanisms and health implications. Cell Res. 2021;31(2):107–125. doi:10.1038/s41422-020-00441-1

47. Zhang Z, Wang X, Wang Z, et al. Dihydroartemisinin alleviates hepatic fibrosis through inducing ferroptosis in hepatic stellate cells. Biofactors. 2021;47(5):801–818. doi:10.1002/biof.1764

48. Shen M, Guo M, Li Y, et al. m(6)A methylation is required for dihydroartemisinin to alleviate liver fibrosis by inducing ferroptosis in hepatic stellate cells. Free Radic Biol Med. 2022;182:246–259. doi:10.1016/j.freeradbiomed.2022.02.028

49. Cales P. Apoptosis and liver fibrosis: antifibrotic strategies. Biomed Pharmacother. 1998;52(6):259–263. doi:10.1016/S0753-3322(98)80011-5

50. Chen Q, Chen L, Wu X, et al. Dihydroartemisinin prevents liver fibrosis in bile duct ligated rats by inducing hepatic stellate cell apoptosis through modulating the PI3K/Akt pathway. IUBMB Life. 2016;68(3):220–231. doi:10.1002/iub.1478

51. Deng X, Li Y, Chen Y et al. Paeoniflorin protects hepatocytes from APAP-induced damage through launching via the MAPK/mTOR signaling pathway. Cell Mol Biol Lett. 2024;29(1):119. doi:10.1186/s11658-024-00631-4

52. Zhang Z, Guo M, Zhao S, Shao J, Zheng S. ROS-JNK1/2-dependent activation of autophagy is required for the induction of anti-inflammatory effect of dihydroartemisinin in liver fibrosis. Free Radic Biol Med. 2016;101:272–283. doi:10.1016/j.freeradbiomed.2016.10.498

53. Eichmann TO, Grumet L, Taschler U, et al. ATGL and CGI-58 are lipid droplet proteins of the hepatic stellate cell line HSC-T6. J Lipid Res. 2015;56(10):1972–1984. doi:10.1194/jlr.M062372

54. Blaner WS, O’Byrne SM, Wongsiriroj N, et al. Hepatic stellate cell lipid droplets: a specialized lipid droplet for retinoid storage. Biochim Biophys Acta. 2009;1791(6):467–473. doi:10.1016/j.bbalip.2008.11.001

55. Parola M, Robino G. Oxidative stress-related molecules and liver fibrosis. J Hepatol. 2001;35(2):297–306. doi:10.1016/s0168-8278(01)00142-8

56. Liu R, Li X, Zhu W, et al. Cholangiocyte-derived exosomal long noncoding RNA H19 promotes hepatic stellate cell activation and cholestatic liver fibrosis. Hepatology. 2019;70(4):1317–1335. doi:10.1002/hep.30662

57. Xia S, Wang Z, Chen L, et al. Dihydroartemisinin regulates lipid droplet metabolism in hepatic stellate cells by inhibiting lncRNA-H19-induced AMPK signal. Biochem Pharmacol. 2021;192:114730. doi:10.1016/j.bcp.2021.114730

58. Wang ZM, Xia SW, Zhang T, et al. LncRNA-H19 induces hepatic stellate cell activation via upregulating alcohol dehydrogenase III-mediated retinoic acid signals. Int Immunopharmacol. 2020;84:106470. doi:10.1016/j.intimp.2020.106470

59. Bao Z, Xu M, Kan Y, et al. Dihydroartemisinin requires NR1D1 mediated Rab7 ubiquitination to regulate hepatic stellate cells lipophagy in liver fibrosis. Int J Biol Macromol. 2025;305(Pt 1):141055. doi:10.1016/j.ijbiomac.2025.141055

60. Huan S, Sun S, Song S, et al. Dihydroartemisinin inhibits the activation and proliferation of hepatic stellate cells by regulating miR-29b-3p. Int J Mol Med. 2023;51(5). doi:10.3892/ijmm.2023.5243

61. Zhang Z, Yao Z, Zhao S, et al. Interaction between autophagy and senescence is required for dihydroartemisinin to alleviate liver fibrosis. Cell Death Dis. 2017;8(6):e2886. doi:10.1038/cddis.2017.255

62. Mannaerts I, Leite SB, Verhulst S, et al. The Hippo pathway effector YAP controls mouse hepatic stellate cell activation. J Hepatol. 2015;63(3):679–688. doi:10.1016/j.jhep.2015.04.011

63. Du K, Díaz RM, Oh SH, et al. Targeting yap-mediated hepatic stellate cell death susceptibility and senescence for treatment of liver fibrosis. Hepatology. 2022;76:S50–S51.

64. Gao R, Kalathur RKR, Coto-Llerena M, et al. YAP/TAZ and ATF4 drive resistance to sorafenib in hepatocellular carcinoma by preventing ferroptosis. EMBO Mol Med. 2021;13(12):e14351. doi:10.15252/emmm.202114351

65. Ji J, Cheng Z, Zhang J, et al. Dihydroartemisinin induces ferroptosis of hepatocellular carcinoma via inhibiting ATF4-xCT pathway. J Cell Mol Med. 2024;28(8):e18335. doi:10.1111/jcmm.18335

66. Huang D, Xu D, Chen W, et al. Fe-MnO(2) nanosheets loading dihydroartemisinin for ferroptosis and immunotherapy. Biomed Pharmacother. 2023;161:114431. doi:10.1016/j.biopha.2023.114431

67. Su Y, Zhao D, Jin C, et al. Dihydroartemisinin Induces Ferroptosis in HCC by Promoting the Formation of PEBP1/15-LO. Oxid Med Cell Longev. 2021;2021:3456725. doi:10.1155/2021/3456725

68. Anandhan A, Dodson M, Shakya A, et al. NRF2 controls iron homeostasis and ferroptosis through HERC2 and VAMP8. Sci Adv. 2023;9(5):eade9585. doi:10.1126/sciadv.ade9585

69. He F, Zhang P, Liu JL, et al. ATF4 suppresses hepatocarcinogenesis by inducing SLC7A11 (xCT) to block stress-related ferroptosis. J Hepatol. 2023;79(2):362–377. doi:10.1016/j.jhep.2023.03.016

70. Sun K, Hou LC, Guo Z, et al. JNK-JUN-NCOA4 axis contributes to chondrocyte ferroptosis and aggravates osteoarthritis via ferritinophagy. Free Radical Bio Med. 2023;200:87–101. doi:10.1016/j.freeradbiomed.2023.03.008

71. Zhao L, Miao H, Quan M, et al. beta-Lapachone induces ferroptosis of colorectal cancer cells via NCOA4-mediated ferritinophagy by activating JNK pathway. Chem Biol Interact. 2024;389:110866. doi:10.1016/j.cbi.2024.110866

72. Gao X, Luo Z, Xiang T, Wang K, Li J, Wang P. Dihydroartemisinin induces endoplasmic reticulum stress-mediated apoptosis in HepG2 human hepatoma cells. Tumori J. 2011;97(6):771–780. doi:10.1177/030089161109700615

73. Karin M, Liu Z, Zandi E. AP-1 function and regulation. Curr Opin Cell Biol. 1997;9(2):240–246. doi:10.1016/s0955-0674(97)80068-3

74. Im E, Yeo C, Lee HJ, Lee EO. Dihydroartemisinin induced caspase-dependent apoptosis through inhibiting the specificity protein 1 pathway in hepatocellular carcinoma SK-Hep-1 cells. Life Sci. 2018;192:286–292. doi:10.1016/j.lfs.2017.11.008

75. Kim H, Tu HC, Ren D, et al. Stepwise activation of BAX and BAK by tBID, BIM, and PUMA initiates mitochondrial apoptosis. Mol Cell. 2009;36(3):487–499. doi:10.1016/j.molcel.2009.09.030

76. Qin G, Zhao C, Zhang L, et al. Dihydroartemisinin induces apoptosis preferentially via a Bim-mediated intrinsic pathway in hepatocarcinoma cells. Apoptosis. 2015;20(8):1072–1086. doi:10.1007/s10495-015-1132-2

77. Wu L, Cheng Y, Deng J, Tao W, Ye J. Dihydroartemisinin Inhibits proliferation and induces apoptosis of human hepatocellular carcinoma cell by upregulating tumor necrosis factor via JNK/NF-kappaB pathways. Evid Based Complement Alternat Med. 2019;2019:9581327. doi:10.1155/2019/9581327

78. Zhang CZ, Zhang H, Yun J, Chen GG, Lai PB. Dihydroartemisinin exhibits antitumor activity toward hepatocellular carcinoma in vitro and in vivo. Biochem Pharmacol. 2012;83(9):1278–1289. doi:10.1016/j.bcp.2012.02.002

79. Shi X, Wang L, Ren L, et al. Dihydroartemisinin, an antimalarial drug, induces absent in melanoma 2 inflammasome activation and autophagy in human hepatocellular carcinoma HepG2215 cells. Phytother Res. 2019;33(5):1413–1425. doi:10.1002/ptr.6332

80. Zou J, Ma Q, Sun R, et al. Dihydroartemisinin inhibits HepG2.2.15 proliferation by inducing cellular senescence and autophagy. BMB Rep. 2019;52(8):520–524. doi:10.5483/BMBRep.2019.52.8.058

81. Yano K, Tomono T, Ogihara T. Advances in studies of P-glycoprotein and its expression regulators. Biol Pharm Bull. 2018;41(1):11–19. doi:10.1248/bpb.b17-00725

82. Yang Y, He J, Chen J, et al. Dihydroartemisinin sensitizes mutant p53 (R248Q)-expressing hepatocellular carcinoma cells to doxorubicin by inhibiting P-gp expression. Biomed Res Int. 2019;2019:8207056. doi:10.1155/2019/8207056

83. Yang Y, Gao Y, Gong Y, et al. Dihydroartemisinin breaks the immunosuppressive tumor niche during cisplatin treatment in Hepatocellular carcinoma. Acta Histochem. 2024;126(4):152171. doi:10.1016/j.acthis.2024.152171

84. Rao Q, Li R, Yu H, et al. Effects of dihydroartemisinin combined with cisplatin on proliferation, apoptosis and migration of HepG2 cells. Oncol Lett. 2022;24(2):275. doi:10.3892/ol.2022.13395

85. Hou CY, Guo DQ, Yu X, Wang SY, Liu TH. TMT-based proteomics analysis of the anti-hepatocellular carcinoma effect of combined dihydroartemisinin and sorafenib. Biomed. Pharmacother. 2020;126:109862. doi:10.1016/j.biopha.2020.109862

86. Cui Z, Wang H, Li S, et al. Dihydroartemisinin enhances the inhibitory effect of sorafenib on HepG2 cells by inducing ferroptosis and inhibiting energy metabolism. J Pharmacol Sci. 2022;148(1):73–85. doi:10.1016/j.jphs.2021.09.008

87. Patt Y, Rojas-Hernandez C, Fekrazad HM, Bansal P, Lee FC. Phase II trial of sorafenib in combination with capecitabine in patients with hepatocellular carcinoma: INST 08-20. Oncologist. 2017;22(10):1158–+. doi:10.1634/theoncologist.2017-0168

88. He YC, Zhan LL, Shi J, et al. The combination of R848 with sorafenib enhances antitumor effects by reprogramming the tumor immune microenvironment and facilitating vascular normalization in hepatocellular carcinoma. Adv Sci. 2023;10(18). doi:10.1002/advs.202207650

89. Ladd AD, Duarte S, Sahin I, Zarrinpar A. Mechanisms of drug resistance in HCC. Hepatology. 2024;79(4):926–940. doi:10.1097/HEP.0000000000000237

90. Zhou Z, Lei J, Fang J, et al. Dihydroartemisinin remodels tumor micro-environment and improves cancer immunotherapy through inhibiting cyclin-dependent kinases. Int Immunopharmacol. 2024;139:112637. doi:10.1016/j.intimp.2024.112637

91. Marquard S, Thomann S, Weiler SME, et al. Yes-associated protein (YAP) induces a secretome phenotype and transcriptionally regulates plasminogen activator Inhibitor-1 (PAI-1) expression in hepatocarcinogenesis. Cell Commun Signal. 2020;18(1):166. doi:10.1186/s12964-020-00634-6

92. Ren K, Li T, Zhang W, Ren J, Li Z, Wu G. miR-199a-3p inhibits cell proliferation and induces apoptosis by targeting YAP1, suppressing Jagged1-notch signaling in human hepatocellular carcinoma. J Biomed Sci. 2016;23(1):79. doi:10.1186/s12929-016-0295-7

93. Li S, Li J, Dai W, et al. Genistein suppresses aerobic glycolysis and induces hepatocellular carcinoma cell death. Br J Cancer. 2017;117(10):1518–1528. doi:10.1038/bjc.2017.323

94. Gao Y, Gong Y, Lu J, et al. Dihydroartemisinin breaks the positive feedback loop of YAP1 and GLUT1-mediated aerobic glycolysis to boost the CD8(+) effector T cells in hepatocellular carcinoma. Biochem Pharmacol. 2024;225:116294. doi:10.1016/j.bcp.2024.116294

95. Peng Q, Hao L, Guo Y, et al. Dihydroartemisinin inhibited the Warburg effect through YAP1/SLC2A1 pathway in hepatocellular carcinoma. J Nat Med. 2023;77(1):28–40. doi:10.1007/s11418-022-01641-2

96. Gong Y, Peng Q, Gao Y, et al. Dihydroartemisinin inhibited interleukin-18 expression by decreasing YAP1 in hepatocellular carcinoma cells. Acta Histochem. 2023;125(4):152040. doi:10.1016/j.acthis.2023.152040

97. Guo Y, Peng Q, Hao L, et al. Dihydroartemisinin promoted FXR expression independent of YAP1 in hepatocellular carcinoma. FASEB J. 2022;36(6):e22361. doi:10.1096/fj.202200171R

98. Cai J, Song L, Zhang F, et al. Targeting SRSF10 might inhibit M2 macrophage polarization and potentiate anti-PD-1 therapy in hepatocellular carcinoma. Cancer Commun. 2024;44(11):1231–1260. doi:10.1002/cac2.12607

99. Hao L, Guo Y, Peng Q, et al. Dihydroartemisinin reduced lipid droplet deposition by YAP1 to promote the anti-PD-1 effect in hepatocellular carcinoma. Phytomedicine. 2022;96:153913. doi:10.1016/j.phymed.2021.153913

100. Peng Q, Li S, Shi X, et al. Dihydroartemisinin broke the tumor immunosuppressive microenvironment by inhibiting YAP1 expression to enhance anti-PD-1 efficacy. Phytother Res. 2023;37(5):1740–1753. doi:10.1002/ptr.7695

101. Syed GH, Amako Y, Siddiqui A. Hepatitis C virus hijacks host lipid metabolism. Trends Endocrinol Metab. 2010;21(1):33–40. doi:10.1016/j.tem.2009.07.005

102. Shimotohno K. HCV assembly and egress via modifications in host lipid metabolic systems. Cold Spring Harb Perspect Med. 2021;11(1):a036814. doi:10.1101/cshperspect.a036814

103. Albecka A, Belouzard S, Op de Beeck A, et al. Role of low-density lipoprotein receptor in the hepatitis C virus life cycle. Hepatology. 2012;55(4):998–1007. doi:10.1002/hep.25501

104. Horn P, Tacke F. Metabolic reprogramming in liver fibrosis. Cell Metab. 2024;36(7):1439–1455. doi:10.1016/j.cmet.2024.05.003

105. Ohata T, Yokoo H, Kamiyama T, et al. Fatty acid-binding protein 5 function in hepatocellular carcinoma through induction of epithelial-mesenchymal transition. Cancer Med. 2017;6(5):1049–1061. doi:10.1002/cam4.1020

106. Seo J, Jeong DW, Park JW, Lee KW, Fukuda J, Chun YS. Fatty-acid-induced FABP5/HIF-1 reprograms lipid metabolism and enhances the proliferation of liver cancer cells. Commun Biol. 2020;3(1):638. doi:10.1038/s42003-020-01367-5

107. Yang WS, Kim KJ, Gaschler MM, Patel M, Shchepinov MS, Stockwell BR. Peroxidation of polyunsaturated fatty acids by lipoxygenases drives ferroptosis. Proc Natl Acad Sci U S A. 2016;113(34):E4966–75. doi:10.1073/pnas.1603244113

108. Hannun YA, Obeid LM. Principles of bioactive lipid signalling: lessons from sphingolipids. Nat Rev Mol Cell Biol. 2008;9(2):139–150. doi:10.1038/nrm2329

109. Lee J, Roh JL. Lipid metabolism in ferroptosis: unraveling key mechanisms and therapeutic potential in cancer. Biochim Biophys Acta Rev Cancer. 2025;1880(1):189258. doi:10.1016/j.bbcan.2024.189258

110. Hirata Y, Cai R, Volchuk A, et al. Lipid peroxidation increases membrane tension, Piezo1 gating, and cation permeability to execute ferroptosis. Curr Biol. 2023;33(7):1282–1294e5. doi:10.1016/j.cub.2023.02.060

111. Han N, Yang ZY, Xie ZX, et al. Dihydroartemisinin elicits immunogenic death through ferroptosis-triggered ER stress and DNA damage for lung cancer immunotherapy. Phytomedicine. 2023;112:154682. doi:10.1016/j.phymed.2023.154682

112. Yi R, Wang H, Deng C, et al. Dihydroartemisinin initiates ferroptosis in glioblastoma through GPX4 inhibition. Biosci Rep. 2020;40(6). doi:10.1042/BSR20193314

113. Zheng Y, Zheng J, Du M, et al. An iron-containing ferritin-based nanosensitizer for synergistic ferroptosis/sono-photodynamic cancer therapy. J Mater Chem B. 2023;11(22):4958–4971. doi:10.1039/d3tb00029j

114. Li QG, Peggins JO, Fleckenstein LL, Masonic K, Heiffer MH, Brewer TG. The pharmacokinetics and bioavailability of dihydroartemisinin, arteether, artemether, artesunic acid and artelinic acid in rats. J Pharm Pharmacol. 1998;50(2):173–182. doi:10.1111/j.2042-7158.1998.tb06173.x

115. Morris CA, Duparc S, Borghini-Fuhrer I, Jung D, Shin CS, Fleckenstein L. Review of the clinical pharmacokinetics of artesunate and its active metabolite dihydroartemisinin following intravenous, intramuscular, oral or rectal administration. Malar J. 2011;10:263. doi:10.1186/1475-2875-10-263

116. Galal AM, Gul W, Slade D, et al. Synthesis and evaluation of dihydroartemisinin and dihydroartemisitene acetal dimers showing anticancer and antiprotozoal activity. Bioorg Med Chem. 2009;17(2):741–751. doi:10.1016/j.bmc.2008.11.050

117. Fang Z, Gong S, Dong Z, et al. Key factors, challenges, and recommendations for science and technology innovation in traditional Chinese medicine. World Journal of Integrated Traditional and Western Medicine. 2025;11(3):103–109. doi:10.70976/j.2096-0964.WJIM-2025-0023

118. Li Y, Shi N, Zhang W, et al. Supramolecular hybrids of carbon dots and dihydroartemisinin for enhanced anticancer activity and mechanism analysis. J Mater Chem B. 2020;8(42):9777–9784. doi:10.1039/d0tb01826k

119. Peng J, Wang Q, Zhou J, et al. Targeted lipid nanoparticles encapsulating dihydroartemisinin and chloroquine phosphate for suppressing the proliferation and liver metastasis of colorectal cancer. Front Pharmacol. 2021;12:720777. doi:10.3389/fphar.2021.720777