Next Article in Journal
Novel Application of Ion Mobility Mass Spectrometry Reveals Complex Ganglioside Landscape in Diffuse Astrocytoma Peritumoral Regions
Previous Article in Journal
Molecular and Immunomodulatory Mechanisms of Statins in Inflammation and Cancer Therapeutics with Emphasis on the NF-κB, NLRP3 Inflammasome, and Cytokine Regulatory Axes
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Artemisinin and Its Derivatives from Molecular Mechanisms to Clinical Applications: New Horizons Beyond Antimalarials

1
Institute of Interdisciplinary Integrative Medicine Research, School of Medicine, Nantong University, Nantong 226001, China
2
Department of Pharmaceutical Botany, School of Pharmacy, Naval Medical University, Shanghai 200433, China
*
Authors to whom correspondence should be addressed.
Int. J. Mol. Sci. 2025, 26(17), 8409; https://doi.org/10.3390/ijms26178409
Submission received: 21 July 2025 / Revised: 23 August 2025 / Accepted: 28 August 2025 / Published: 29 August 2025
(This article belongs to the Section Molecular Biology)

Abstract

Artemisinin and its derivatives are widely recognized for their exceptional antimalarial efficacy. Recently, accumulating evidence indicates therapeutic potential beyond malaria. Despite these advances, detailed mechanisms and pharmacological limitations remain incompletely defined. This review summarizes their pharmacological activities and molecular mechanisms associated with oncology, immunoregulation, and metabolic disorders. Mechanistically, these compounds exert potent antitumor effects by inducing oxidative stress, arresting the cell cycle, triggering apoptosis, and inhibiting angiogenesis. They likewise modulate immune responses, re-establishing immune homeostasis and enhancing the effectiveness of immunotherapeutic strategies. Preliminary evidence also suggests involvement in metabolic regulation, pointing to promising avenues for treating metabolic disorders. Given alternative mechanisms of artemisinin and its derivatives, we also discuss the trinity modulation network among antitumor activity, immunoregulation, and metabolic homeostasis. We anticipate that future research will address these knowledge gaps, thereby enhancing the clinical utility of artemisinin and its derivatives and improving patient outcomes across diverse pathologies.

1. Introduction

Artemisinin (C15H22O5), a sesquiterpene lactone characterized by a distinctive 1,2,4-trioxane ring, was first isolated through bioassay-guided fractionation in 1972 by Tu Youyou from Artemisia annua L. (Asteraceae) [1]. Atemisia annua L., a traditional Chinese medicinal herb commonly known as sweet wormwood, stores artemisinin in the glandular secretory trichomes of its leaves. With its bitter and pungent properties, this herb has long been used to relieve summer heat syndrome and treat malaria. In 1981, the World Health Organization and United Nations formally recognized artemisinin’s antimalarial efficacy at the Beijing International Symposium on Antimalarial Drug Development, marking a major milestone that led to the global adoption of artemisinin-based combination therapy (ACTs) as the first-line treatment for Plasmodium falciparum malaria [2].
Although artemisinin is the foundational antimalarial compound, it faces significant limitations, including thermal instability, poor solubility, and high production costs. To improve their clinical utility, semi-synthetic derivatives have been developed using artemisinic acid as a precursor to overcome these issues. Among these, dihydroartemisinin (C15H24O5) acts as both a key intermediate and a potent antimalarial agent itself. Furthermore, artesunate (C19H28O8) was designed specifically to provide enhanced water solubility, making it critical for treating severe malaria, while artemether (C16H26O5) and arteether (C17H28O5) utilize their lipophilicity for rapid absorption and efficacy against drug-resistant strains [3,4,5]. To combat emerging resistance, ACTs that rely on these derivatives remain the frontline strategy. Additionally, newer derivatives like SM934 and hybrid molecules not only further optimize pharmacokinetic properties but also expand potential therapeutic applications beyond malaria (Figure 1).
Beyond malaria, growing evidence suggests that artemisinin compounds have therapeutic potential in cancer treatment, immune system regulation, metabolic disorders, and kidney diseases [6,7]. Artemisinin and its derivatives have demonstrated significant antitumor activity against various tumor types (such as lung, breast, colorectal, and liver cancers), and their potential mechanisms (such as inducing ferroptosis, promoting reactive oxygen species production, cell cycle arrest, apoptosis induction, autophagy regulation, and anti-angiogenesis) have been elaborated in several reviews in recent years. However, the current literature has not yet thoroughly elucidated the molecular mechanisms by which ARTs combat tumors, particularly the regulatory network between them and immune regulation, as well as metabolic homeostasis [8,9]. Immunomodulatory effects of dihydroartemisinin have been identified, including its ability to reduce lupus-associated nephritis by inhibiting the production of anti-dsDNA antibodies, reducing TNF-α secretion, and blocking NF-κB signaling [10]. More recently, research has focused on how artemisinin regulates glucose and lipid metabolism via related enzymes and signaling pathways. In addition, clinical trials have been conducted for some diseases, and preliminary results have confirmed their clinical safety and efficacy. However, the exact mechanisms and clinical potential of artemisinin and its derivatives in oncology, immunology, and metabolism are not fully understood. Furthermore, the regulatory network among anti-tumor activity, immune regulation, and metabolic homeostasis requires more in-depth research and discussion.
In this review, we synthesize international research on the antitumor, immunoregulatory, and metabolic effects of probiotics, along with their current clinical applications. Additionally, we explored the interconnected modulation network encompassing antitumor activity, immunoregulation, and metabolic homeostasis, aiming to provide a comprehensive framework for future research and inspire novel therapeutic strategies.

2. Antitumor Activity of Artemisinin and Its Derivatives

First reported in 1995, Singh and Lai reported that dihydroartemisinin, in combination with holotransferrin, selectively killed human breast cancer cells in vitro while showing low toxicity to normal cells [11], paving the way for its development as an antitumor agent [12,13]. Subsequent studies have further confirmed its potent anticancer effects, which are mediated through mechanisms such as oxidative stress induction, cell cycle arrest, programmed cell death, and inhibition of angiogenesis (Figure 2).

2.1. Mechanisms of Antitumor Activity

2.1.1. Oxidative Stress

Oxidative stress occurs when the production of highly reactive oxygen and nitrogen species during cellular aging or in response to deleterious stimuli exceeds the body’s antioxidant defenses, resulting in tissue damage. Recently, the induction of tumor cell death through oxidative stress has garnered significant attention in cancer therapy [14].
Tumor cells exhibit a dependency on both heme and ferrous iron. Consequently, artemisinin and its derivatives interact with heme-bound Fe2+ to produce reactive oxygen species (ROS), which in turn inflict damage to tumor cell membranes, proteins, and DNA [15]. For instance, dihydroartemisinin complexes with transferrin effectively inhibit cancer cell proliferation while sparing normal cells [11,16,17]. These compounds also disrupt mitochondrial function and increase oxidative stress, primarily through peroxisomes and mitochondrial enzymes [18,19]. Specifically, oxidative stress interferes with the electron transport chain in tumor cells, causing electron leakage, reducing membrane potential and ROS production, which triggers mitochondrial dysfunction and apoptosis [20]. For instance, dihydroartemisinin damages mitochondrial DNA and promotes cancer cell death by inhibiting TOM70 [21]. Subsequently, ROS suppresses cancer cell growth, promotes apoptosis, blocks invasion and metastasis, and inhibits angiogenesis by regulating signaling pathways, including ERK1/2, RTK-independent activation, EGFR, and mTOR. This involves key proteins, such as Nrf2 and MAPK, in preventing cancer cell proliferation [22,23]; Src, NF-κB, and PI3K/Akt in promoting cancer cell apoptosis; and matrix metalloproteinases (MMP) secretion, Met overexpression, and Rho-Rac interaction in blocking cancer cell invasion and metastasis. Moreover, ROS regulates VEGF and angiopoietin release during angiogenesis inhibition [24].

2.1.2. Cell Cycle Arrest

The eukaryotic cell cycle, which spans from the end of one cell division to the completion of the next, consists of interphase (G1, S, and G2 phases) and the M phase. This cycle is regulated by genes that encode key proteins, such as cyclins and cyclin-dependent kinases (CDKs) [25].
Artemisinin upregulates p16 and downregulates ERK1/2 and cyclin D1 in gallbladder cancer cells [26]. Halofuginone and artemisinin synergistically arrest cells at G0/G1 by increasing p21 and p27 levels [27]. In hepatoma models, artesunate reduces cyclin D1 and E2F1 while increasing p21 [28]. Some artemisinin compounds induce G2/M arrest. Artesunate triggers cell cycle arrest by inducing cytokinesis defects, multinucleation, centrosome amplification, and multipolar spindle formation [29]. Additionally, it downregulates cyclin B1, enhancing cancer cell radiosensitivity [30], and induces ROS-dependent DNA damage, further blocking cell cycle progression [31].

2.1.3. Induction of Cell Death

Apoptosis
Apoptosis is an orderly and autonomous process of genetically controlled cell death that maintains internal environmental stability. Fas activation promotes the mitochondrial release of substances such as cytochrome C, which facilitates the activation of caspases, a key step in apoptosis [32].
Studies on different cell models have shown that artesunate alternatively upregulates Fas and CD95 expression, disrupts mitochondrial membrane potential, releases cytochrome C, and activates caspase-3 and caspase-9, all of which induce apoptosis [33,34]. For instance, artesunate and sorafenib (SOR) synergistically induce apoptosis by significantly upregulating the expression of cleaved caspase-3 and poly (ADP-ribose) polymerase via the STAT3 pathway [35]. Dihydroartemisinin induces the release of cytochrome C and apoptosis-inducing factor from the mitochondria, promotes Bax cleavage, increases caspase-3 levels, and impairs Bcl2 protein expression [36,37]. In addition, artesunate activates p38/MAPK and upregulates the levels of ROS, thereby inducing apoptosis [38,39].
Ferroptosis
Ferroptosis, distinct from apoptosis and autophagy, is an iron-dependent programmed cell death. Its mechanisms involve iron accumulation, which catalyzes the lipid peroxidation of polyunsaturated fatty acids (PUFAs) in cell membranes, generating ROS and triggering cell death. Additionally, ferroptosis is driven by the inactivation of GPX4, the central enzyme in the glutathione (GSH)-dependent antioxidant system [40,41].
Artemisinin and its derivatives induce ferroptosis in cancer cells through coordinated modulation of iron metabolism, redox homeostasis, and lipid peroxidation pathways. These compounds trigger ferritinophagy-mediated iron release [42,43] and differentially regulate cellular iron levels. Dihydroartemisinin increases iron accumulation and IRP-IRE binding [42], whereas artesunate decreases iron levels in KTCTL-26 cells [44]. They disrupt the antioxidant defense system by downregulating SLC7A11, GSH, and GPX4 [45,46,47], potentially via p53-dependent mechanisms [48,49]. Concurrently, artemisinin compounds elevate ROS levels via mitochondrial generation [50] and iron-catalyzed reactions [51], while promoting lipid peroxidation through ALOX5 activation [52,53] and the PEBP1/ALOX15 pathway (particularly for dihydroartemisinin) [54]. This multifaceted action leads to irreversible oxidative damage and iron-dependent cell death.
Autophagy
Autophagy is a conserved cellular process in eukaryotic organisms, in which damaged proteins and organelles are sequestered by double-membrane vesicles known as autophagosomes. These vesicles subsequently fuse with lysosomes (in animals) or vacuoles (in yeast and plants) to degrade and recycle their contents [55].
Artemisinin and its derivatives exert antitumor effects through multiple forms of autophagy, including mitophagy and ferritinophagy. For example, dihydroartemisinin impairs mitochondrial function, increases ROS production, and activates mitophagy [37,56]. It also facilitates the degradation of ferritin, elevates intracellular iron levels, and enhances ROS accumulation, ultimately driving ferritinophagy [57].
Anti-Angiogenesis
Angiogenesis, regulated by diverse factors such as HIF-1α, VEGF, and PDGF, enables endothelial cells to degrade the extracellular matrix and form new blood vessels. This process supplies tumors with oxygen and nutrients and is essential for their progression. Consequently, anti-angiogenic therapies aim to block this process, depriving tumors of critical support and representing a key strategy in cancer treatment [58].
Artemisinin and its derivatives demonstrate potent anti-angiogenic activity by modulating key angiogenic factors across various cancer cell lines [59]. For example, artesunate inhibits the expression of VEGF and PDGF, downregulates HIF-1α, and suppresses vasculogenic mimicry formation, collectively contributing to its antitumor efficacy [60]. Similarly, dihydroartemisinin markedly reduces cancer cell-induced angiogenesis by downregulating the expression of VEGF, MMP-2, and MMP-9 proteins [61].

2.2. Applications in Distinct Malignancies

2.2.1. Lung Cancer

Lung cancer remains the leading cause of cancer-related mortality worldwide, with incidence patterns varying by geography and time due to differences in tobacco exposure [62,63,64]. Conventional treatments, including surgery, radiotherapy, and chemotherapy, offer limited efficacy in advanced-stage diseases, underscoring the need for novel therapeutic strategies [65].
Artemisinin and its derivatives have demonstrated antitumor activity in lung cancer [66]. For example, artemisinin suppresses cancer cell proliferation, invasion, differentiation, and angiogenesis by inhibiting sphingosine kinase 1 [67]. Artemisinin treatment reduces CDK4 and COX-2 levels while increasing caspase-3 and IL-6 expression, resulting in apoptosis, cell cycle arrest, and inhibition of microsphere formation in lung cancer cells [68]. Dihydroartemisinin enhances cisplatin sensitivity by inhibiting PTGS1 and activating the ROS–ERK/p38/JNK signaling cascade [69]. It further disrupts tumor progression by blocking ROR1-mediated STAT3 phosphorylation, suppressing HK2/L [70], and TOM70 while activating gasdermin E-mediated pyroptosis [21]. Artesunate binds to FABP5 and inhibits the PPARγ pathway, thereby promoting apoptosis and reducing lung cancer cell proliferation and migration [71]. In non-small cell lung cancer, artesunate also downregulates BTBD7 mRNA expression in a dose-dependent manner, thereby increasing apoptosis and impairing cell migration [72].

2.2.2. Hepatocellular Carcinoma

Hepatocellular carcinoma is the sixth most common cancer worldwide, with approximately 910,000 new cases and 830,000 deaths annually [64,73]. Only about 30% of patients with early-stage disease are eligible for curative surgical resection [74].
Artemisinin and its derivatives exhibit potent antitumor effects in hepatocellular carcinoma (HCC) through diverse molecular mechanisms. Metabolic modulation is a key therapeutic strategy. For instance, these compounds disrupt the YAP1–GLUT1 feedback loop, suppressing glycolysis in tumor cells while enhancing glucose uptake in CD8+ T cells to bolster antitumor immunity [75]. Oxidative stress induction further contributes to their efficacy, as dihydroartemisinin triggers ferroptosis by accumulating lipid peroxides and inhibiting the ATF4/xCT antioxidant pathway [76]. Additionally, artemisinin targets transcriptional regulation by downregulating the oncogenic factor FoxM1 to reverse drug resistance in liver cancer cells [77]. Beyond its direct tumoricidal effects, artesunate synergizes with sorafenib (SOR) by activating the AFAP1L2–SRC–FUNDC1 axis, thereby promoting mitochondrial autophagy and amplifying therapeutic outcomes [78].

2.2.3. Breast Cancer

Breast cancer is the most commonly diagnosed malignancy among women, accounting for approximately 2.3 million new cases annually [64]. While early-stage patients benefit from over 90% five-year survival rates with surgery and radiotherapy [79], late-stage breast cancer remains challenging, with observed chemotherapy resistance [80].
Artemisinin and its derivatives have overcome breast cancer drug resistance by inducing ferroptosis and regulating autophagy–apoptosis pathways [81]. For example, artemisinin suppresses migration and chemoresistance in LCC9 cells by modulating the expression of resistance-related genes [58]. Dihydroartemisinin also promotes ferroptosis in breast cancer by downregulating hsa_circ_0001610 and enhancing radiosensitivity [82]. Furthermore, artesunate induces apoptosis by modulating Bax/Bcl-2 and G0/G1 cell cycle arrest by regulating IGF-IR levels in MCF-7 cells [83]. However, the clinical efficacy of artemisinin-based therapies is limited by their poor tumor-targeting capabilities. To address this issue, a glutathione-sensitive artemisinin ester conjugate in liposomes (TPP-SS-ATS-LS) was encapsulated to target both tumor cells and mitochondria. This delivery system significantly enhances antitumor activity in breast cancer mouse models, increasing tumor growth inhibition from 37.7% to 56.4% [84].

2.2.4. Colorectal Cancer

Colorectal cancer accounts for approximately 1.93 million new cases annually worldwide, with its incidence rising notably in developing countries [64,85]. Early-stage patients benefit from a 70–80% five-year survival rate with surgical resection, while outcomes for advanced-stage patients remain poor [86].
Emerging evidence suggests that artemisinin and its derivatives have therapeutic potential in colorectal cancer through multiple mechanisms, including the induction of apoptosis, inhibition of angiogenesis, and modulation of the tumor immune microenvironment [87]. For example, artemisinin has been shown to bind to microsomal prostaglandin E synthase-2 (mPGES-2), thereby reducing PGE2 production and suppressing colorectal cancer cell proliferation [88]. Dihydroartemisinin, a key derivative, inhibits glycogen synthase kinase 3β (GSK-3β), disrupts the TCF7/MMP9 signaling pathway, and promotes β-catenin degradation, consequently impairing tumor growth and metastasis. Moreover, it displays synergistic effects when combined with capecitabine, enhancing antitumor immunity and improving efficacy while alleviating chemotherapy-associated side effects [89].

2.2.5. Other Tumors

Artemisinin and its derivatives have also demonstrated antitumor activity against a variety of malignancies, including leukemia, ovarian cancer, glioblastoma, prostate cancer, and melanoma, as summarized in Table 1. These mechanisms include the induction of oxidative stress, cell cycle arrest, apoptosis, autophagy, and inhibition of angiogenesis.

3. Immunomodulatory Activity of Artemisinin and Its Derivatives

Artemisinin and its derivatives, initially studied for immunomodulatory effects in the early 2000s, showed promise in treating autoimmune and inflammatory diseases while ensuring safety. Recent findings have revealed their ability to modulate vital immune cells, including T cells, B cells, macrophages, and NK cells (Figure 3).

3.1. Mechanisms of Immunomodulatory Activity

3.1.1. T-Cell Regulation

T lymphocytes play a central role in maintaining immune homeostasis by coordinating cellular and humoral immune responses. They help regulate B cell–mediated antibody production through the secretion of various cytokines [121].
Artemisinin and its derivatives exhibit broad immunomodulatory effects by targeting multiple aspects of T and B cell function. Specifically, artemisinin suppresses pro-inflammatory T cell activity by inhibiting the proliferation of inflammatory subsets (e.g., Th1 and Th17) and downregulating key cytokines, such as IL-2 and IFN-γ [122,123]. These effects are mediated by the inhibition of critical signaling pathways, including Ras-Raf1-ERK1/2 [124], PI3K-AKT [125,126], and TLR [127] in T cells, which collectively impair T cell activation and differentiation. For instance, TPN10475 counteracts TCR-induced suppression of TGF-β signaling, further inhibiting effector CD4+ T cell function in vitro [128]. Additionally, artemisinin modulates regulatory T cells (Tregs) by upregulating their populations [122,123], a process driven by dihydroartemisinin’s activation of the c-Fos transcription factor, which enhances expression of proliferation-related genes (e.g., VEGF, T cell receptor, IL-2) [129]. By simultaneously suppressing inflammatory T cell responses and promoting Treg-mediated tolerance, artemisinin compounds effectively restore immune homeostasis and indirectly influence B cell-driven humoral immunity through T cell cytokine regulation [121].

3.1.2. B-Cell Regulation

B lymphocytes play a crucial role in adaptive immunity by mediating antigen-specific and humoral responses. Through the production of antibodies, they orchestrate pathogen neutralization, opsonization, and complement activation while simultaneously presenting antigens to T cells to bridge innate and adaptive immunity.
Artemisinin and its derivatives significantly modulate humoral immunity by targeting B lymphocytes’ function through multiple mechanisms. First, these compounds regulate B cell activation and plasma cell (PC) differentiation, ultimately leading to the suppression of B cell-dependent IgM antibody production [130]. Dihydroartemisinin blocks Bruton’s tyrosine kinase signaling and its downstream NF-κB and Myc pathways, directly inhibiting B cell activation and differentiation, and decreasing the number of splenic and circulatory B cells, thereby suppressing B cell response and antibody production in systemic lupus erythematosus [131,132]. Artesunate inhibits the BAFF-induced NF-κB signaling pathway by regulating the ubiquitination and degradation of TRAF6. This restricts the overactivation, proliferation, and survival of B cells and decreases the infiltration of lymphocytes and secretion of autoantibodies [133]. TRAF6 also serves as a critical adaptor in BAFF-mediated B cell signaling. Consequently, artesunate is expected to restrict excessive B-cell activation, proliferation, and survival. These compounds inhibit the proliferation of splenic marginal zone B cells while increasing the relative proportion of CD21 (low) CD23(+) follicular and B10 B cells. This shift negatively regulates IL-10 secretion, thereby weakening B cell-mediated immune responses [134]. Collectively, these results indicate potential clinical applications for attenuating antibody-mediated rejection (ABMR).

3.1.3. Macrophage Modulation

Macrophages play essential roles in both cellular and humoral immunity, acting as antigen-presenting and effector cells, and engaging in the phagocytosis of pathogens. During both acute and chronic inflammatory responses, macrophages secrete proinflammatory cytokines through pathways such as NF-κB activation [135].
Artemisinin and its derivatives modulate macrophage activity by suppressing pro-inflammatory cytokine production and enhancing macrophage autophagy. For instance, chitosan-coated artesunate inhibits TLR4/NF-κB signaling and promotes STAT6-driven M2 macrophage reprogramming in ulcerative colitis [136]. Artesunate blocks TLR4-dependent autophagy in acute pancreatitis while activating the HIF-1α/NF-κB signaling cascade to drive macrophage repolarization in rheumatoid arthritis [137,138]. Dihydroartemisinin blocks TLR4-mediated activation of NF-κB-STAT3/MAPK cascades, ameliorating the innate inflammatory response induced by muramidase-released protein [139]. SM934 inhibits the TLR4/NF-κB/NLRP3 axis in dry eye disease [140]. Artemisinin also enhances M2 polarization by downregulating the MYD88/ERK signaling [141]. Concurrently, it activates the JNK/MAPK-caspase-9 apoptotic pathway, further suppressing pro-inflammatory cytokines (IL-12, TNF-α), and demonstrates therapeutic efficacy in colitis [142]. Collectively, these mechanisms reduce pro-inflammatory cytokine expression, inhibit M1-type polarization, and promote M2-type macrophage reprogramming.

3.1.4. Natural Killer Cell Potentiation

Natural killer (NK) cells are pivotal innate immune effectors that directly eliminate virus-infected and tumor cells. They concurrently secrete cytokines, such as interferon-γ, to enhance immune surveillance and clearance and to modulate the adaptive immune response. Pretreatment with artemisinin and its derivatives sensitizes tumor cells to NK cell-mediated killing in vitro by increasing the binding of NK cells to target cells and upregulating the expression of apoptosis-related genes in tumor cells [143,144]. These mechanisms enhance susceptibility to NK cell cytotoxicity, thereby amplifying the immunological efficacy of NK cells.

3.2. Applications in Immune-Mediated Disorders

3.2.1. Systemic Lupus Erythematosus

Systemic lupus erythematosus (SLE), a multi-organ autoimmune disease, exhibits significant geographic variations in its global prevalence [145,146]. Although glucocorticoids and immunosuppressants have been shown to significantly alleviate symptoms, their therapeutic efficacy remains limited, particularly in severe cases refractory to conventional treatments [147,148].
Artemisinin and its derivatives exhibit immunomodulatory effects in SLE by restoring immune cell homeostasis and suppressing inflammatory responses. Dihydroartemisinin has been shown to inhibit T follicular helper (Tfh) and B cells, reduce abnormal antibodies [129], and restore Treg/Th17 balance by upregulating Treg cells and downregulating Th17 cells, thereby mitigating excessive immune responses and autoimmune tissue damage in SLE [149]. Artesunate reduces serum ANA and anti-dsDNA titers, urinary protein, and creatinine levels in SLE-model mice. It lowers MCP-1 levels in serum, urine, and kidneys and decreases BAFF expression in the spleen, thereby modulating the immune system and improving SLE [150]. Furthermore, SM934 demonstrates favorable efficacy in the lupus-prone female MRL/lpr mouse model, and its mechanism of action is mainly reflected in the regulation of T cell subsets. In vitro, SM934 inhibits IFN-γ and IL-17 production by activating CD4+ T cells and suppresses their differentiation into Th1 and Th17 cells. In vivo, it increases Treg levels, inhibits Th1 and Th17 development, and blocks STAT1, STAT3, and STAT5 activation in splenocytes [151].

3.2.2. Rheumatoid Arthritis

Rheumatoid arthritis (RA) is characterized by synovial inflammation and progressive bone erosion [152]. Despite progress in RA treatment, some refractory cases, particularly seronegative conditions, continue to pose a risk of irreversible joint damage, underscoring the need for optimized drug efficacy and safety [153].
Emerging evidence has demonstrated the potential of artemisinin derivatives for RA treatment. For instance, DC32, a dihydroartemisinin derivative, ameliorates footpad swelling, cartilage degradation, and inflammatory cell infiltration in a collagen-induced arthritis (CIA) model via the Nrf2-p62-Keap1 feedback loop [154]. Artemisinin shows positive effects in a CIA model in DBA/1J mice by inhibiting RA-FLS proliferation, inducing apoptosis, and reducing migration and invasion [155]. SM934 inhibits Tfh and Th17 cell development and suppresses CII-reactive T cell proliferation and inflammatory cytokine secretion in DBA/1J mice, thereby limiting pathogenic antibody production. In vitro, it also inhibits naïve CD4+ T cell polarization to Tfh, Bcl-6 expression, IL-21-producing CD4+ T cells, and STAT3 signaling, thereby regulating immune responses [156]. In addition, artesunate affects the Th17/Treg lymphocyte balance by regulating Treg cell apoptosis and Th17 cell proliferation [157].

3.2.3. Psoriasis

Psoriasis is an immune-mediated disease that affects the skin and joints. Although a multilevel therapeutic regimen has been developed (consisting of topical retinoids, photochemotherapy, bio-targeted agents, and small-molecule inhibitors), treatment resistance persists in specific subtypes, such as recalcitrant plaque lesions and generalized pustular psoriasis [158].
Artemisinin and its derivatives have demonstrated significant therapeutic potential in both animal models and preclinical studies. For example, dihydroartemisinin alleviates skin inflammation by modulating T cell subsets, suppressing cytokine levels, and inhibiting NF-κB phosphorylation and key transcription factors in psoriasis [159]. Artemisinin also modulates immune responses by reducing γδ T cell populations in lymph nodes without affecting Th17 cells, thereby lowering serum IFN-γ and IL-17 levels and suppressing keratinocyte proliferation and epidermal thickening [160]. Additionally, artesunate inhibits TNFα–induced IL-23 expression in HaCaT keratinocyte cells, a key driver of inflammatory cell recruitment and activation in psoriatic lesions [161].

3.2.4. Multiple Sclerosis

Multiple sclerosis (MS) progressively destroys myelin sheaths in the central nervous system and shows sex and latitudinal incidence variations. Despite modern treatments like β-interferon and B-cell depletion, effective strategies to prevent neurodegeneration in primary progressive MS are lacking, making neural axon repair and nerve function restoration challenging, particularly in cases of severe fatigue or cognitive dysfunction [162].
Artemisinin and its derivatives primarily treat MS by regulating immune cell activity and preserving myelin integrity. In experimental autoimmune EAE mouse models, they suppress IFN-γ production while promoting IL-4 expression, effectively shifting the immune response from Th1 to Th2 responses. This immune polarization reduces Th1-mediated inflammation and alleviates disease severity, although the precise mechanisms remain unclear [163,164]. Additionally, the artemisinin derivative ADART exhibits potent efficacy in MS models, inhibiting IFN-γ and IL-17 production in vitro, reducing the ratios of Th1 and Th17 cells, and lowering the levels of IFN-γ and IL-17A in vivo, further contributing to disease attenuation [165].

3.2.5. Other Immune Disorders

Artemisinin and its derivatives have also demonstrated efficacy on other immune diseases, including transplant rejection, type 1 diabetes, autoimmune hepatitis, experimental autoimmune encephalomyelitis, and melanoma (Table 2). These effects include promoting T and B cell proliferation, regulating the release of cytokines, and modulating signaling pathways.

4. Metabolic Regulation of Artemisinin and Its Derivatives

Artemisinin and its derivatives regulate glucose and lipid metabolism through key enzymes, altering intracellular signaling pathways, and counteracting oxidative stress (Figure 4). Recent work has extended these findings to metabolic diseases, such as diabetes, obesity, and atherosclerosis.

4.1. Glucose Metabolism

Glucose metabolism is intricately regulated by a multitude of factors, such as hormonal regulation, rate-limiting enzymes within glycolysis, allosteric modulation, and gene expression regulation. Scholarly interest in the impact of artemisinin and its derivatives on glucose metabolism has been increasing, especially in the context of diabetes, which represents a significant global health issue [176,177]. Their antidiabetic effects involve activating the insulin signaling pathway [178], enhancing glucose transport [179], and improving both intracellular glucose metabolism and inflammation [119].
Artemisinin and its derivatives activate the insulin signaling pathway, thereby promoting glucose uptake and utilization in cells [178]. They also improve insulin resistance by reducing pancreatic β-cell apoptosis, enhancing β-cell function, and increasing insulin secretion. In db/db mice, artemether significantly improves insulin sensitivity and glucose homeostasis [180]. Additionally, artemisinin promotes the transdifferentiation of pancreatic α-cells into β-cells [181]. This effect may be mediated by GABAA receptor signaling, although this mechanism remains controversial and requires further investigation [182]. Artemisinin also plays a therapeutic role in type 2 diabetes by enhancing GLUT1 expression, which further promotes glucose metabolism [183]. Additionally, novel artemisinin derivatives target GLUT1, affecting tumor cell metabolism [179,184].
Artemisinin and its derivatives inhibit glycogen synthase and improve intracellular glucose metabolism and inflammatory responses through multiple signaling pathways. AMPK, a key metabolism-regulating enzyme, regulates the metabolism of glucose, lipids, and proteins. Artemisinin promotes glucose uptake and utilization in mice with diabetes by activating the AMPK pathway while inhibiting the activity of GSK-3β, thus improving glucose tolerance and insulin sensitivity [89]. Furthermore, it also affects multiple signaling pathways, including TGF-β [185], β3-integrin [133], and RANKL [186], and influences ERK [187,188,189], PI3K, and AKT [183,190], which regulate glucose metabolism and inflammation. Additionally, they inhibit NF-κB activity, leading to the downregulation of inflammatory cytokines and indirectly disrupting glucose metabolism [191].
Regarding clinical application potential, artemisinin and its derivatives have shown promising efficacy in diabetes-related studies. They not only significantly reduce blood glucose levels but also provide protection against diabetes-related complications, such as diabetic cardiomyopathy, diabetic retinopathy, diabetic nephropathy, and diabetes-induced nerve damage [185,192,193].

4.2. Lipid Metabolism

Artemisinin and its derivatives modulate lipid metabolism by inhibiting lipogenesis, promoting lipid degradation, and restoring cholesterol metabolism homeostasis. They therefore show therapeutic potential for diseases such as obesity, atherosclerosis, and cardiovascular diseases.
Regarding the suppression of lipogenesis, artemisinin and its derivatives significantly reduced fat accumulation and improved lipid profiles in high-fat diet-induced mouse models by regulating key signaling pathways. Research has demonstrated that the PPAR signaling pathway plays a pivotal role in this process, primarily by reducing the phosphorylation levels of PPAR-γ and C/EBP-α [194,195]. Concurrently, these compounds effectively inhibit adipocyte differentiation and lipid storage by suppressing the Akt/mTOR signaling pathway [196].
Artemether exhibits remarkable efficacy in promoting lipolysis and energy metabolism. It activates the p38 MAPK/ATF2 signaling pathway to induce white adipose tissue browning and enhance brown adipose tissue thermogenesis, thereby increasing energy expenditure [196]. Furthermore, in cholesterol metabolism regulation, artemisinin upregulates Klf2 expression, facilitating the binding of TCF7L2 to the LPL promoter, which subsequently increases LPL expression and promotes lipid hydrolysis, ultimately reducing plasma cholesterol and triglyceride levels [172,182]. Artesunate has demonstrated significant lipid-lowering effects. Preclinical studies have shown that this compound ameliorates aortic root lesions and reduces body weight in obesity models without causing adverse effects, such as nausea [197]. This effect is closely associated with the ability to regulate cholesterol homeostasis.
With respect to anti-inflammatory and antioxidant effects, artemisinin effectively inhibits ROS production by downregulating NOX4 expression in adipocytes [198,199,200], thereby restricting adipocyte differentiation and lipid storage, ultimately reducing fat mass and improving glucose-lipid metabolism in obese mice [200]. Notably, in atherosclerosis models, artemisinin and its derivatives significantly attenuate plaque formation and improve vascular health by preventing lipid accumulation, alleviating oxidative stress, and exerting anti-inflammatory effects [201,202]. Specifically, artesunate reduces the expression levels of pro-inflammatory cytokines (TNF-α, IL-6) and chemokines (IL-8, MCP-1) [194], while simultaneously inhibiting M1 macrophage polarization and lipid deposition in vascular walls [203].
Taken together, these findings highlight the potential of artemisinin and its derivatives as therapeutic agents for lipid metabolism disorders, especially obesity and atherosclerosis (Table 3).

4.3. Regulation of Oxidative Stress and Metabolism Interplay

Artemisinin and its derivatives exert protective effects by simultaneously targeting the dysregulated interplay between oxidative stress and metabolic dysfunction, particularly in glucose and lipid metabolism. Unlike their pro-oxidant actions in tumors, these compounds alleviate oxidative stress in metabolic contexts via three interconnected mechanisms.
Artemisinin and its derivatives activate integrated antioxidant-metabolic defenses. They initiate protection via the Nrf2 pathway activation. This upregulates antioxidant enzymes (SOD, GSH), directly reducing oxidative damage in pathologies like diabetic nephropathy [187,211,214,215]. Significantly, this mechanism intersects with metabolic regulation by concurrently supporting lipid homeostasis [194,200,207], demonstrating intrinsic crosstalk between antioxidant and metabolic systems.
Disruption of inflammatory oxidative amplification cycles represents the second axis of intervention. Elevated ROS levels trigger NF-κB-mediated release of cytokines (TNF-α, IL-6), which further stimulates ROS production [216,217]. By suppressing NF-κB activation, artemisinin reduces cytokine levels (TNF-α, IL-6, IL-8, and MCP-1), thereby interrupting this vicious cycle. Crucially, this anti-inflammatory action translates to improved metabolic outcomes, as evidenced by restored glucose homeostasis in diabetes [218], directly linking oxidative stress control to metabolic benefits.
Targeting metabolic origins of oxidative stress completes the regulatory triad. Hyperglycemia induces oxidative stress via the polyol pathway, formation of advanced glycation end products (AGEs), and activation of protein kinase C (PKC), all of which impair insulin sensitivity and β-cell function [219,220]. Artemisinin compounds enhance insulin signaling and glucose uptake, partly by activating the MAPK and PI3K/AKT pathways [221,222]. Parallelly, it ameliorates lipid-driven oxidative stress caused by fatty acid accumulation, mitochondrial ROS, and NOX4 activation [200], through NOX4 downregulation and PPARs/Akt/mTOR modulation [194,200,207]. Moreover, artemisinin activates AMPK signaling, protecting against oxidative damage by agents like amiodarone [209,210].
In summary, artemisinin and its derivatives combat metabolic oxidative stress through multiple coordinated mechanisms, activating antioxidant pathways and improving both glucose and lipid metabolism. These multifaceted effects not only alleviate symptoms but also target upstream drivers of disease, highlighting their promising clinical potential.

5. The Trinity Modulation Network Among Antitumor Activity, Immunoregulation and Metabolic Homeostasis

5.1. Antitumor-Immunometabolic Synergy

Immunotherapy has emerged as a cornerstone of cancer treatment, mobilizing the host’s adaptive and innate immune systems against tumors [223,224]. The overlapping metabolic reprogramming of cancer and immune cells is now recognized as a pivotal determinant of antitumor immunity [225,226]. Within this framework, artemisinin compounds demonstrate dual targeting of immunometabolic crosstalk. For instance, dihydroartemisinin and artemether disrupt tumor aerobic glycolysis in non-small cell lung cancer by suppressing ERK/c-Myc signaling [227], while concurrently reprogramming immune responses—as evidenced by dihydroartemisinin delaying head and neck squamous cell carcinoma progression through reduced M2-TAM infiltration and angiogenesis inhibition [228]. Critically, this metabolic interference promotes immunostimulatory monocyte-to-dendritic cell differentiation while counteracting immunosuppressive pathways [227], establishing a mechanistic synergy. Thus, the convergence of artemisinin-induced metabolic modulation and immune activation provides a compelling rationale for combining these agents with immunotherapies targeting the immunometabolic network [226], thereby potentiating antitumor efficacy.

5.2. Oxidative Stress Regulation in Tumor and Metabolic Diseases

As mentioned in Section 3.1.1 and Section 5.3, artemisinin and its derivatives show promising therapeutic potential for regulating oxidative stress based on the microenvironment. In tumors, they promote ROS generation via the Fenton reaction in an iron-rich environment [22,26], leading to ferroptosis and mitochondrial permeability transition [21]. Although Nrf2 activation aids tumor survival under normal conditions, combining it with pro-oxidative therapies can induce synthetic lethality at elevated ROS levels. Conversely, in metabolic disorders, artemisinin and its derivatives protect against metabolic disorders by reducing NOX4-derived ROS, enhancing insulin/PI3K/AKT signaling, and blocking NF-κB/NLRP3 inflammatory pathways [183,229,230], shifting mitochondria from ROS amplifying in tumors to scavenging in metabolic disorders. Moreover, therapeutic strategies differ. Tumors focus on inhibiting angiogenesis and metastasis, while metabolic reprogramming emphasizes lipid oxidation and detoxification [231,232].
These approaches reflect the distinct microenvironments involved in tumor and metabolic disease development. Tumors rely on iron metabolism for ROS buildup, while metabolic disorders need ROS clearance [233]. Despite these differences, both strategies converge on mitochondrial regulation, underscoring the key role of organelles in redox adaptation. Given Nrf2’s dual role, tissue-targeted delivery systems are essential. Artemisinin and its derivatives offer a versatile platform for combination therapies targeting tumor cytotoxicity and metabolic balance.

5.3. Immune-Metabolic Crosstalk

Immune cell function is intrinsically linked to metabolic reprogramming, and redox metabolism plays a pivotal role in maintaining internal homeostasis. Antioxidant systems, such as thioredoxin (TRX) and glutathione (GSH), regulate the proliferation and function of T cells, B cells, and macrophages primarily through molecular hubs like Nrf2 [234,235]. Within this framework, artemisinin and its derivatives exert targeted immunomodulatory effects by reprogramming cellular metabolism. They inhibit pro-inflammatory M1 macrophage polarization while promoting anti-inflammatory M2 phenotypes via suppression of the NF-κB/NLRP3 pathway [230]. Furthermore, artemisinin critically disrupts T-cell differentiation by altering metabolic preferences. Th17 cells (pro-inflammatory) depend on glycolysis and glutamine metabolism, whereas Treg cells (anti-inflammatory) rely on oxidative phosphorylation and amino acid catabolism [236]. By activating AMPK and inhibiting mTORC1 signaling [107], artemisinin compounds shift T cell metabolism from glycolysis to fatty acid oxidation, thereby suppressing Th17 differentiation and promoting Treg expansion [237].
This crosstalk extends to metabolic pathologies in which immune dysfunction exacerbates disease progression. In atherosclerosis, inflammatory cytokines (IL-1β, TNF-α) and M1 macrophages drive plaque inflammation, necrosis, and thrombosis [238]. Similarly, Th17/Treg imbalance perpetuates chronic inflammation in obesity and in type 2 diabetes [239]. Artemisinin counteracts these processes through two mechanisms: first, by suppressing the NF-κB/NLRP3 inflammasome to reduce caspase-1, IL-1β, and TGF-β levels [96,230]; and second, by restoring Th17/Treg equilibrium through metabolic reprogramming [230]. Consequently, the simultaneous targeting of the immune and metabolic axes by artemisinin provides novel therapeutic implications for immunotherapy in metabolic disorders.

6. Clinical Safety and Efficacy

ACTs demonstrate subtype-specific clinical efficacy and safety when administered according to the WHO-recommended regimens [240]. For uncomplicated Plasmodium vivax or ovale malaria, standard ACT regimens, including dihydroartemisinin-piperaquine or artesunate-amodiaquine, achieve clinical efficacy when combined with primaquine at total doses of either 180 mg administered over 8 days or 210 mg over 14 days. These regimens show no reported severe adverse events, even in cases of mixed infections. In Plasmodium malariae or knowlesi malaria, abbreviated ACT regimens have similar efficacy and safety profiles. Effective options include dihydroartemisinin-piperaquine administered with an initial loading dose, followed by fractional dosing; artesunate-amodiaquine given as two tablets daily for three consecutive days; or a single 8-tablet dose of artemether-naphthoquine. For severe malaria cases, intravenous artesunate remains the first-line therapy due to its superior hemodynamic stability compared to intramuscular artemether. The recommended protocol for adults involves administration of 120 mg doses at 0, 12, and 24 h, followed by a daily maintenance dose. Pediatric patients weighing less than 20 kg should receive 3 mg/kg per dose. Clinicians should exercise particular caution when treating patients presenting with shock. Once patients regain consciousness and demonstrate oral tolerance, a transition to oral ACTs is clinically appropriate.
Artemisinin and its derivatives have shown preliminary anticancer efficacy with favorable safety profiles in early clinical trials across multiple malignancies. In breast cancer, Phase I/II clinical trials, including NCT00764036, have evaluated oral artemether at doses of 100, 150, or 200 mg daily, showing preliminary anticancer efficacy with good safety and tolerability, although no Phase III trials have been initiated [241]. Similarly, in colorectal cancer, a randomized, double-blind trial (n = 23) reported that oral artemisinin increased tumor apoptosis (>7%) without significant adverse events, supporting its pro-apoptotic mechanism observed in preclinical studies [242]. For cervical cancer, research has progressed to Phase I trials assessing intravaginal artesunate and oral dihydroartemisinin formulations [243]. In summary, artemisinin and its derivatives offer several potential advantages, including broad-spectrum anticancer activity, minimal adverse reactions in clinical trials, and no cross-resistance with conventional therapies. However, there are still significant limitations, as most evidence comes from small-scale early-phase trials or preclinical studies. Future research should conduct larger, well-designed Phase II/III trials to validate clinical efficacy and establish long-term safety profiles, particularly for the extended treatment durations required in oncology compared with malaria applications [244].
In immune system diseases, artemisinin and its derivatives have demonstrated promising clinical efficacy for SLE treatment. An early randomized controlled trial involving 45 SLE patients showed that artemisinin tablets administered at 50 mg twice daily combined with prednisone at 0.25 mg to 0.8 mg per kilogram per day significantly reduced disease activity and improved symptoms including fever, joint pain, erythema, rash, and hair loss compared to only prednisone at 0.8 mg to 1.25 mg per kilogram per day, with comparable safety profiles [245]. Advancing clinical development further, SM934 is approved as a Class I new drug by the China National Medical Products Administration in 2015 and is currently undergoing Phase II clinical trials. Preclinical studies have shown that it has superior efficacy in alleviating kidney and cutaneous lupus lesions, with lower toxicity than traditional immunosuppressants like cyclosporine A [246]. Although these preliminary findings are promising, larger Phase II and III trials are needed to fully establish the long-term efficacy and safety of these compounds in SLE treatment.
Current preclinical research on artemisinin and its derivatives in metabolic diseases has mainly focused on diabetes and its complications, obesity-related metabolic disorders, hyperuricemic nephropathy, and non-alcoholic fatty liver disease. However, to date, no clinical trials have been conducted. The optimal dosing parameters, administration protocol, and comprehensive safety assessments still need further investigation prior to clinical translation.
In summary, compared to the well-established short-course antimalarial treatments, artemisinin and its derivatives require relatively long treatment cycles and/or high-intensity exposure to achieve efficacy in oncology, immunomodulation, and metabolism [247]. However, this may increase the risk of mechanism-related toxicities. Artemisinin derivatives exhibit significant dose- and time-dependent differences in various therapeutic applications, inevitably triggering the inherent mechanism-based toxicity risks of the drug, including neurotoxicity, cardiotoxicity, embryotoxicity, and hematopoietic suppression, etc. [248]. Based on the above analysis, the dose-time-dependent toxicity risk of artemisinin-based drugs should be prioritized in clinical translation.

7. Conclusions and Outlook

Artemisinin and its derivatives are not only highly effective in treating malaria but also show potential in cancer therapy, immune regulation, and metabolic disorders. In cancer, they induce apoptosis, inhibit angiogenesis, and arrest the cell cycle. Importantly, they overcome drug resistance to traditional chemotherapies [244]. In immunoregulation, they enhance immune responses and macrophage activity [249]. Furthermore, in metabolic regulation, they help regulate blood sugar and lipid levels, offering new treatment options for metabolic diseases.
Although artemisinin and its derivatives share fundamental anticancer properties due to the structures of peroxide bridge-mediated free radical generation and ferroptosis induction, there are several important mechanisms in different tumor types. Firstly, the tumor selectivity of artemisinin derivatives depends on their structural modifications [250] and the intracellular levels of labile ferrous iron [42]. Artesunate C-10 succinate enhances aqueous solubility and enables intravenous administration. Its carboxyl group mediates transferrin binding, selectively targeting TfR-high cancer cells in HCC [28]. The lipophilic ether bond of artemether confers selective penetration advantages for treating breast cancer and gliomas by enhancing blood-brain barrier and tumor membrane permeability [251]. Second, the target specificity and mechanistic selectivity of artemisinin derivatives are closely associated with distinct oncogenic driver pathways across different tumor types. In HCC, artemisinin disrupts cellular bioenergetics and inactivates Hippo-YAP signaling [252], while artesunate exacerbates AFAP1L2-SRC-FUNDC1 axis-dependent mitophagy and mitigates sorafenib resistance [78]. For breast cancer, artemisinin derivatives suppress TGF-β signaling and inactivate cancer-associated fibroblasts [253]. Finally, distinct immunomodulatory effects arise from tumor microenvironment (TME) heterogeneity. For example, hepatocellular carcinoma primarily relies on innate immune regulation (macrophages) [254], while breast and lung cancers emphasize adaptive immunity (T cells/NK cells) [255,256]. Overall, artemisinin derivatives exhibit conserved peroxide-mediated cytotoxicity and tumor-specific mechanisms due to their different structural modifications, oncogenic pathways, and microenvironmental factors. Further research is needed to optimize its clinical application.
Although current studies have confirmed the therapeutic efficacy of artemisinin and its derivatives, a comprehensive elucidation of their polypharmacology demands integrating advanced technologies. AI-driven chemoproteomics, longitudinal gut microbiome analysis, and dynamic metabolomics will systematically decode their multi-target mechanisms, particularly in metabolic regulation. Building on this mechanistic foundation, broadening their therapeutic spectrum should explore emerging frontiers—including endocrine disorders (e.g., thyroid dysfunction, adrenal cortex hyperplasia) [127] and cardiovascular pathologies—where their established anti-inflammatory and immunometabolic reprogramming capacities (evidenced in obesity models) may intercept core disease drivers, suggesting untapped potential for chronic inflammatory conditions. For successful clinical translation, two pillars must be prioritized. First, mechanism-focused trials should be designed using cytokine signatures or microbial biomarkers for patient stratification. Second, developing nano-formulations (e.g., lipid-based Pheroid™ systems [257]) and solubility-enhanced delivery platforms [258] can overcome pharmacokinetic limitations, ultimately advancing both therapeutic efficacy and safety.
In summary, shifting artemisinin and its derivatives from a broad-spectrum antimalarial to precision medicine requires a multidisciplinary approach. It integrates multi-omics discoveries, disease-independent mechanism exploration, and intelligent clinical validation frameworks. By prioritizing these directions, the scientific community may fully unlock the revolutionary potential of precision medicine.

Author Contributions

Conceptualization, Y.X. and J.L.; methodology, Y.X. and Y.P.; investigation, Y.X., J.L., Z.Z. and X.H.; resources, L.Z. and A.L.; writing—original draft preparation, Y.X., J.L. and Z.Z.; writing—review and editing, Y.X., J.L., Z.Z., C.S. and A.L.; visualization, Y.X., J.L., Z.Z. and M.L.; supervision, Y.X., L.Z. and A.L.; project administration, A.L.; funding acquisition, L.Z. All authors have read and agreed to the published version of the manuscript.

Funding

This research was funded by the National Natural Science Foundation of China (No. 82225047, 82170274), the National Key Research and Development Program of China (No. 2022YFC3501703), High-level Talents Program of Nantong University, and Postgraduate Research & Practice Innovation Program of Jiangsu Province (KYCX24_3569).

Acknowledgments

Some figures were partially created using BioRender and ChemDraw 20.0.

Conflicts of Interest

The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest.

Abbreviations

The following abbreviations are used in this manuscript:
CDKCyclin-dependent kinase
p16Cyclin-dependent kinase inhibitor 2A (CDKN2A)
ERK1/2Extracellular signal-regulated protein kinase 1/2
CDK4Cyclin-dependent kinase 4
CDK2Cyclin-dependent kinase 2
RBRetinoblastoma protein
TOM70Translocase of Outer Mitochondrial Membrane 70
P450Cytochrome P450
XOXanthine Oxidase
CytcCytochrome c
ROSReactive Oxygen Species
RTK-iReceptor Tyrosine Kinase inhibitor
Nrf2Nuclear factor erythroid 2-related factor 2
MAPKMitogen-activated protein kinase
SrcProto-oncogene tyrosine-protein kinase Src
NF-κBNuclear factor kappa-light-chain-enhancer of activated B cells
PI3K/AktPhosphoinositide 3-kinase—Protein kinase B
MMPMatrix metalloproteinases
MetHepatocyte Growth Factor Receptor
RhoRas homolog
RacRas-related C3 botulinum toxin substrate
TfR1Transferrin Receptor 1
SLC3A2Solute Carrier Family 3 Member 2
SLC7A11Solute Carrier Family 7 Member 11
c-Casp9Cleaved Caspase-9
c-Casp3Cleaved Caspase-3
c-PARPCleaved Poly (ADP-ribose) Polymerase
PUFA-PLPolyunsaturated Fatty Acid-Phospholipid
ALOXsArachidonate lipoxygenases
PORCytochrome P450 oxidoreductase
PUFA-PL-OOHPolyunsaturated Fatty Acid-Phospholipid Hydroperoxide
PINK1PTEN Induced Kinase 1
PARK2Parkin RBR E3 Ubiquitin Protein Ligase
TLRToll-like Receptor
PI3KPhosphoinositide 3-Kinase
AKTProtein Kinase B (PKB)
c-FosCellular FOS Proto-Oncogene
IFN-γInterferon-Gamma
TNF-αTumor Necrosis Factor-α
BTK signalBruton’s Tyrosine Kinase signaling
MycMYC Proto-Oncogene
AMPKAMP-activated Protein Kinase
mTORMechanistic Target of Rapamycin
ULK1Unc-51 Like Autophagy Activating Kinase 1
VDRVitamin D Receptor
HIF-1αHypoxia-Inducible Factor 1-α
NLRP3NLR Family Pyrin Domain Containing 3
GLUT1Glucose Transporter 1
GSK-3βGlycogen Synthase Kinase 3 beta
β3Integrin beta-3
MCP-1Monocyte Chemoattractant Protein-1
NOX4NADPH Oxidase 4
PPARαPeroxisome Proliferator-Activated Receptor α
Klf2Krüppel-Like Factor 2
LPLLipoprotein Lipase
ABCA1ATP-Binding Cassette Subfamily A Member 1
TGTriglyceride
TCTotal Cholesterol

References

  1. Miller, L.H.; Su, X. Artemisinin: Discovery from the Chinese herbal garden. Cell 2011, 146, 855–858. [Google Scholar] [CrossRef]
  2. Ma, N.; Zhang, Z.; Liao, F.; Jiang, T.; Tu, Y. The birth of artemisinin. Pharmacol. Ther. 2020, 216, 107658. [Google Scholar] [CrossRef] [PubMed]
  3. Brown, G.D. The biosynthesis of artemisinin (Qinghaosu) and the phytochemistry of Artemisia annua L. (Qinghao). Molecules 2010, 15, 7603–7698. [Google Scholar] [CrossRef] [PubMed]
  4. Li, J.; Zhou, B. Biological Actions of Artemisinin: Insights from Medicinal Chemistry Studies. Molecules 2010, 15, 1378–1397. [Google Scholar] [CrossRef] [PubMed]
  5. Ridley, R.G.; Hudson, A.T. Chemotherapy of malaria. Curr. Opin. Infect. Dis. 1998, 11, 691–706. [Google Scholar] [CrossRef]
  6. Jin, Q.; Liu, T.; Chen, D.; Yang, L.; Mao, H.; Ma, F.; Wang, Y.; Li, P.; Zhan, Y. Therapeutic potential of artemisinin and its derivatives in managing kidney diseases. Front. Pharmacol. 2023, 14, 1097206. [Google Scholar] [CrossRef]
  7. Addissouky, T.A. Artemisinin and its derivatives throughout the therapeutic mechanisms and clinical potential. Discov. Chem. 2025, 2, 10. [Google Scholar] [CrossRef]
  8. Augustin, Y.; Staines, H.M.; Krishna, S. Artemisinins as a novel anti-cancer therapy: Targeting a global cancer pandemic through drug repurposing. Pharmacol. Ther. 2020, 216, 107706. [Google Scholar] [CrossRef]
  9. Zeng, Z.W.; Chen, D.; Chen, L.; He, B.; Li, Y. A comprehensive overview of Artemisinin and its derivatives as anticancer agents. Eur. J. Med. Chem. 2023, 247, 115000. [Google Scholar] [CrossRef]
  10. Li, W.D.; Dong, Y.J.; Tu, Y.Y.; Lin, Z.B. Dihydroarteannuin ameliorates lupus symptom of BXSB mice by inhibiting production of TNF-alpha and blocking the signaling pathway NF-kappa B translocation. Int. Immunopharmacol. 2006, 6, 1243–1250. [Google Scholar] [CrossRef]
  11. Singh, N.P.; Lai, H. Selective toxicity of dihydroartemisinin and holotransferrin toward human breast cancer cells. Life Sci. 2001, 70, 49–56. [Google Scholar] [CrossRef]
  12. Elhassanny, A.E.M.; Soliman, E.; Marie, M.; McGuire, P.; Gul, W.; ElSohly, M.; Van Dross, R. Heme-Dependent ER Stress Apoptosis: A Mechanism for the Selective Toxicity of the Dihydroartemisinin, NSC735847, in Colorectal Cancer Cells. Front. Oncol. 2020, 10, 965. [Google Scholar] [CrossRef]
  13. McCorkle, J.R.; Ahn, R.; Cao, C.D.; Hill, K.S.; Dietrich, C.S.; Kolesar, J.M. Antineoplastic Drug Synergy of Artesunate with Navitoclax in Models of High-Grade Serous Ovarian Cancer. Cancers 2024, 16, 1321. [Google Scholar] [CrossRef] [PubMed]
  14. Hayes, J.D.; Dinkova-Kostova, A.T.; Tew, K.D. Oxidative Stress in Cancer. Cancer Cell 2020, 38, 167–197. [Google Scholar] [CrossRef]
  15. Fan, J.; Liu, X.; Wang, Q.; Wang, H.; Liu, H.; Han, D.; Ren, J. A metal-organic framework-based redox homeostasis disruptor selectively potentiate the cytotoxicity of dihydroartemisinin for cancer therapy. Nano Res. 2023, 16, 7489–7495. [Google Scholar] [CrossRef]
  16. Ba, Q.; Zhou, N.; Duan, J.; Chen, T.; Hao, M.; Yang, X.; Li, J.; Yin, J.; Chu, R.; Wang, H. Dihydroartemisinin exerts its anticancer activity through depleting cellular iron via transferrin receptor-1. PLoS ONE 2012, 7, e42703. [Google Scholar] [CrossRef] [PubMed]
  17. Wang, Z.; Zhou, H.J. [Dihydroartemisinin down-regulates the expression of transferrin receptor in myeloid leukemia cells]. Yao Xue Xue Bao 2008, 43, 576–583. [Google Scholar]
  18. Xu, C.; Xiao, L.; Zhang, X.; Zhuang, T.; Mu, L.; Yang, X. Synthesis and biological activities of novel mitochondria-targeted artemisinin ester derivatives. Bioorg. Med. Chem. Lett. 2021, 39, 127912. [Google Scholar] [CrossRef]
  19. Lin, L.; Lu, W.; Dai, T.; Chen, H.; Wang, T.; Yang, L.; Yang, X.; Liu, Y.; Sun, D. Novel artemisinin derivatives with potent anticancer activities and the anti-colorectal cancer effect by the mitochondria-mediated pathway. Bioorg. Chem. 2021, 106, 104496. [Google Scholar] [CrossRef]
  20. Fang, J.; Zhao, X.; Li, S.; Xing, X.; Wang, H.; Lazarovici, P.; Zheng, W. Protective mechanism of artemisinin on rat bone marrow-derived mesenchymal stem cells against apoptosis induced by hydrogen peroxide via activation of c-Raf-Erk1/2-p90(rsk)-CREB pathway. Stem Cell Res. Ther. 2019, 10, 312. [Google Scholar] [CrossRef]
  21. Li, L.G.; Hu, J.; Han, N.; Chen, N.N.; Yu, T.T.; Ren, T.; Xu, H.Z.; Peng, X.C.; Li, X.Y.; Ma, T.Q.; et al. Dihydroartemisinin-driven TOM70 inhibition leads to mitochondrial destabilization to induce pyroptosis against lung cancer. Phytother. Res. 2024, 38, 3856–3876. [Google Scholar] [CrossRef]
  22. Matsuzawa, A.; Ichijo, H. Redox control of cell fate by MAP kinase: Physiological roles of ASK1-MAP kinase pathway in stress signaling. Biochim. Biophys. Acta 2008, 1780, 1325–1336. [Google Scholar] [CrossRef] [PubMed]
  23. Wiemer, E.A.C. Stressed tumor cell, chemosensitized cancer. Nat. Med. 2011, 17, 1552–1554. [Google Scholar] [CrossRef] [PubMed]
  24. Sosa, V.; Moliné, T.; Somoza, R.; Paciucci, R.; Kondoh, H.; Lleonart, M.E. Oxidative stress and cancer: An overview. Ageing Res. Rev. 2013, 12, 376–390. [Google Scholar] [CrossRef] [PubMed]
  25. Malumbres, M.; Barbacid, M. Cell cycle, CDKs and cancer: A changing paradigm. Nat. Rev. Cancer 2009, 9, 153–166. [Google Scholar] [CrossRef]
  26. Jia, J.; Qin, Y.; Zhang, L.; Guo, C.; Wang, Y.; Yue, X.; Qian, J. Artemisinin inhibits gallbladder cancer cell lines through triggering cell cycle arrest and apoptosis. Mol. Med. Rep. 2016, 13, 4461–4468. [Google Scholar] [CrossRef]
  27. Chen, G.; Gong, R.; Shi, X.; Yang, D.; Zhang, G.; Lu, A.; Yue, J.; Bian, Z. Halofuginone and artemisinin synergistically arrest cancer cells at the G1/G0 phase by upregulating p21Cip1 and p27Kip1. Oncotarget 2016, 7, 50302–50314. [Google Scholar] [CrossRef]
  28. Hou, J.; Wang, D.; Zhang, R.; Wang, H. Experimental therapy of hepatoma with artemisinin and its derivatives: In vitro and in vivo activity, chemosensitization, and mechanisms of action. Clin. Cancer Res. 2008, 14, 5519–5530. [Google Scholar] [CrossRef]
  29. Efferth, T.; Dunstan, H.; Sauerbrey, A.; Miyachi, H.; Chitambar, C.R. The anti-malarial artesunate is also active against cancer. Int. J. Oncol. 2001, 18, 767–773. [Google Scholar] [CrossRef]
  30. Zhao, Y.; Jiang, W.; Li, B.; Yao, Q.; Dong, J.; Cen, Y.; Pan, X.; Li, J.; Zheng, J.; Pang, X.; et al. Artesunate enhances radiosensitivity of human non-small cell lung cancer A549 cells via increasing NO production to induce cell cycle arrest at G2/M phase. Int. Immunopharmacol. 2011, 11, 2039–2046. [Google Scholar] [CrossRef]
  31. Greenshields, A.L.; Shepherd, T.G.; Hoskin, D.W. Contribution of reactive oxygen species to ovarian cancer cell growth arrest and killing by the anti-malarial drug artesunate. Mol. Carcinog. 2017, 56, 75–93. [Google Scholar] [CrossRef] [PubMed]
  32. Basu, V.; Shabnam; Murghai, Y.; Ali, M.; Sahu, S.; Verma, B.K.; Seervi, M. ONC212, alone or in synergistic conjunction with Navitoclax (ABT-263), promotes cancer cell apoptosis via unconventional mitochondrial-independent caspase-3 activation. Cell Commun. Signal. 2024, 22, 441. [Google Scholar] [CrossRef] [PubMed]
  33. Efferth, T.; Giaisi, M.; Merling, A.; Krammer, P.H.; Li-Weber, M. Artesunate induces ROS-mediated apoptosis in doxorubicin-resistant T leukemia cells. PLoS ONE 2007, 2, e693. [Google Scholar] [CrossRef] [PubMed]
  34. Sieber, S.; Gdynia, G.; Roth, W.; Bonavida, B.; Efferth, T. Combination treatment of malignant B cells using the anti-CD20 antibody rituximab and the anti-malarial artesunate. Int. J. Oncol. 2009, 35, 149–158. [Google Scholar]
  35. Chen, Y.; Tao, H.; Wang, F.; Wu, P.; Gao, J.; Zhang, X.; He, Z.; Zhou, Z.; Jia, Y. Artesunate synergistically promotes sorafenib-induced apoptosis and ferroptosis in non-Hodgkin lymphoma cells through inhibition of the STAT3 pathway. Oncol. Rep. 2023, 50, 147. [Google Scholar] [CrossRef]
  36. Nazmabadi, R.; Pooladi, M.; Amri, J.; Abbasi, Y.; Karami, H.; Darvish, M. Dihydroartemisinin Enhances the Therapeutic Efficacy of BH3 Mimetic Inhibitor in Acute Lymphoblastic Leukemia Cells via Inhibition of Mcl-1. Asian Pac. J. Cancer Prev. 2024, 25, 325–332. [Google Scholar] [CrossRef]
  37. Shi, S.; Luo, H.; Ji, Y.; Ouyang, H.; Wang, Z.; Wang, X.; Hu, R.; Wang, L.; Wang, Y.; Xia, J.; et al. Repurposing Dihydroartemisinin to Combat Oral Squamous Cell Carcinoma, Associated with Mitochondrial Dysfunction and Oxidative Stress. Oxid. Med. Cell. Longev. 2023, 2023, 9595201. [Google Scholar] [CrossRef]
  38. Beccafico, S.; Morozzi, G.; Marchetti, M.C.; Riccardi, C.; Sidoni, A.; Donato, R.; Sorci, G. Artesunate induces ROS- and p38 MAPK-mediated apoptosis and counteracts tumor growth in vivo in embryonal rhabdomyosarcoma cells. Carcinogenesis 2015, 36, 1071–1083. [Google Scholar] [CrossRef]
  39. Cheng, R.; Li, C.; Li, C.; Wei, L.; Li, L.; Zhang, Y.; Yao, Y.; Gu, X.; Cai, W.; Yang, Z.; et al. The artemisinin derivative artesunate inhibits corneal neovascularization by inducing ROS-dependent apoptosis in vascular endothelial cells. Investig. Ophthalmol. Vis. Sci. 2013, 54, 3400–3409. [Google Scholar] [CrossRef]
  40. Tang, D.; Kang, R.; Berghe, T.V.; Vandenabeele, P.; Kroemer, G. The molecular machinery of regulated cell death. Cell Res. 2019, 29, 347–364. [Google Scholar] [CrossRef]
  41. Stockwell, B.R.; Friedmann Angeli, J.P.; Bayir, H.; Bush, A.I.; Conrad, M.; Dixon, S.J.; Fulda, S.; Gascón, S.; Hatzios, S.K.; Kagan, V.E.; et al. Ferroptosis: A Regulated Cell Death Nexus Linking Metabolism, Redox Biology, and Disease. Cell 2017, 171, 273–285. [Google Scholar] [CrossRef]
  42. Chen, G.Q.; Benthani, F.A.; Wu, J.; Liang, D.; Bian, Z.X.; Jiang, X. Artemisinin compounds sensitize cancer cells to ferroptosis by regulating iron homeostasis. Cell Death Differ. 2020, 27, 242–254. [Google Scholar] [CrossRef] [PubMed]
  43. Hu, Y.; Guo, N.; Yang, T.; Yan, J.; Wang, W.; Li, X. The Potential Mechanisms by which Artemisinin and Its Derivatives Induce Ferroptosis in the Treatment of Cancer. Oxid. Med. Cell. Longev. 2022, 2022, 1458143. [Google Scholar] [CrossRef] [PubMed]
  44. Markowitsch, S.D.; Schupp, P.; Lauckner, J.; Vakhrusheva, O.; Slade, K.S.; Mager, R.; Efferth, T.; Haferkamp, A.; Juengel, E. Artesunate Inhibits Growth of Sunitinib-Resistant Renal Cell Carcinoma Cells through Cell Cycle Arrest and Induction of Ferroptosis. Cancers 2020, 12, 3150. [Google Scholar] [CrossRef] [PubMed]
  45. Kong, Y.; Li, J.; Lin, R.; Lu, S.; Rong, L.; Xue, Y.; Fang, Y. Understanding the unique mechanism of ferroptosis: A promising therapeutic target. Front. Cell Dev. Biol. 2023, 11, 1329147. [Google Scholar] [CrossRef]
  46. Soula, M.; Weber, R.A.; Zilka, O.; Alwaseem, H.; La, K.; Yen, F.; Molina, H.; Garcia-Bermudez, J.; Pratt, D.A.; Birsoy, K. Metabolic determinants of cancer cell sensitivity to canonical ferroptosis inducers. Nat. Chem. Biol. 2020, 16, 1351–1360. [Google Scholar] [CrossRef]
  47. Zhu, S.; Yu, Q.; Huo, C.; Li, Y.; He, L.; Ran, B.; Chen, J.; Li, Y.; Liu, W. Ferroptosis: A Novel Mechanism of Artemisinin and its Derivatives in Cancer Therapy. Curr. Med. Chem. 2021, 28, 329–345. [Google Scholar] [CrossRef]
  48. Estrada-Cárdenas, P.; Camacho-Jiménez, L.; Peregrino-Uriarte, A.B.; Contreras-Vergara, C.A.; Hernandez-López, J.; Yepiz-Plascencia, G. p53 knock-down and hypoxia affects glutathione peroxidase 4 antioxidant response in hepatopancreas of the white shrimp Litopenaeus vannamei. Biochimie 2022, 199, 1–11. [Google Scholar] [CrossRef]
  49. Qian, B.; Che, L.; Du, Z.B.; Guo, N.J.; Wu, X.M.; Yang, L.; Zheng, Z.X.; Gao, Y.L.; Wang, M.Z.; Chen, X.X.; et al. Protein phosphatase 2A-B55β mediated mitochondrial p-GPX4 dephosphorylation promoted sorafenib-induced ferroptosis in hepatocellular carcinoma via regulating p53 retrograde signaling. Theranostics 2023, 13, 4288–4302. [Google Scholar] [CrossRef]
  50. Du, J.; Wang, T.; Li, Y.; Zhou, Y.; Wang, X.; Yu, X.; Ren, X.; An, Y.; Wu, Y.; Sun, W.; et al. DHA inhibits proliferation and induces ferroptosis of leukemia cells through autophagy dependent degradation of ferritin. Free Radic. Biol. Med. 2019, 131, 356–369. [Google Scholar] [CrossRef]
  51. Ren, M.; Liang, S.; Lin, S.; Huang, R.; Chen, Y.; Zhang, Y.; Xu, Y. Design, synthesis and biological evaluation of artesunate-Se derivatives as anticancer agents by inducing GPX4-mediated ferroptosis. Bioorg. Chem. 2024, 152, 107733. [Google Scholar] [CrossRef]
  52. Li, C.; Zhang, Y.; Liu, J.; Kang, R.; Klionsky, D.J.; Tang, D. Mitochondrial DNA stress triggers autophagy-dependent ferroptotic death. Autophagy 2021, 17, 948–960. [Google Scholar] [CrossRef]
  53. Wang, Y.; Yuan, X.; Ren, M.; Wang, Z. Ferroptosis: A New Research Direction of Artemisinin and Its Derivatives in Anti-Cancer Treatment. Am. J. Chin. Med. 2024, 52, 161–181. [Google Scholar] [CrossRef]
  54. Su, Y.; Zhao, D.; Jin, C.; Li, Z.; Sun, S.; Xia, S.; Zhang, Y.; Zhang, Z.; Zhang, F.; Xu, X.; et al. Dihydroartemisinin Induces Ferroptosis in HCC by Promoting the Formation of PEBP1/15-LO. Oxid. Med. Cell. Longev. 2021, 2021, 3456725. [Google Scholar] [CrossRef]
  55. Norell, P.N.; Campisi, D.; Mohan, J.; Wollert, T. Biogenesis of omegasomes and autophagosomes in mammalian autophagy. Biochem. Soc. Trans. 2024, 52, 2145–2155. [Google Scholar] [CrossRef]
  56. Li, B.; Bu, S.; Sun, J.; Guo, Y.; Lai, D. Artemisinin derivatives inhibit epithelial ovarian cancer cells via autophagy-mediated cell cycle arrest. Acta Biochim. Biophys. Sin. 2018, 50, 1227–1235. [Google Scholar] [CrossRef] [PubMed]
  57. Wan, X.; Li, C.; Tan, Y.H.; Zuo, S.Q.; Deng, F.M.; Sun, J.; Liu, Y.L. Dihydroartemisinin eliminates senescent cells by promoting autophagy-dependent ferroptosis via AMPK/mTOR signaling pathway. Cell Biol. Int. 2024, 48, 726–736. [Google Scholar] [CrossRef]
  58. Zhao, C.; Zeng, Y.; Kang, N.; Liu, Y. A new perspective on antiangiogenic antibody drug resistance: Biomarkers, mechanisms, and strategies in malignancies. Drug Dev. Res. 2024, 85, e22257. [Google Scholar] [CrossRef] [PubMed]
  59. Soomro, S.; Langenberg, T.; Mahringer, A.; Konkimalla, V.B.; Horwedel, C.; Holenya, P.; Brand, A.; Cetin, C.; Fricker, G.; Dewerchin, M.; et al. Design of novel artemisinin-like derivatives with cytotoxic and anti-angiogenic properties. J. Cell. Mol. Med. 2011, 15, 1122–1135. [Google Scholar] [CrossRef] [PubMed]
  60. Ma, Q.Y.; Xu, X.Y.; Zhu, Y.Z.; Yao, N.N.; Liu, Y.C.; Gao, X.D.; Zhang, Q.; Luo, W.J. Artesunate inhibits vasculogenic mimicry in choroidal melanoma through HIF-1 α/VEGF/PDGF pathway. Acta Histochem. 2024, 126, 152174. [Google Scholar] [CrossRef]
  61. Rao, Q.; Yu, H.; Li, R.; He, B.; Wang, Y.; Guo, X.; Zhao, G.; Wu, F. Dihydroartemisinin inhibits angiogenesis in breast cancer via regulating VEGF and MMP-2/-9. Fundam. Clin. Pharmacol. 2024, 38, 113–125. [Google Scholar] [CrossRef]
  62. Barta, J.A.; Powell, C.A.; Wisnivesky, J.P. Global Epidemiology of Lung Cancer. Ann. Glob. Health 2019, 85, 8. [Google Scholar] [CrossRef]
  63. Chen, Q.; Ying, S.; Qin, J.; Zhang, L. Optimization of treatment strategies for elderly patients with advanced non-small cell lung cancer. Front. Oncol. 2024, 14, 1384906. [Google Scholar] [CrossRef]
  64. Sung, H.; Ferlay, J.; Siegel, R.L.; Laversanne, M.; Soerjomataram, I.; Jemal, A.; Bray, F. Global Cancer Statistics 2020: GLOBOCAN Estimates of Incidence and Mortality Worldwide for 36 Cancers in 185 Countries. CA Cancer J. Clin. 2021, 71, 209–249. [Google Scholar] [CrossRef] [PubMed]
  65. Li, D.; Xu, X.; Liu, J.; Liang, D.; Shi, J.; Li, S.; Jin, J.; He, Y. Small cell lung cancer (SCLC) incidence and trends vary by gender, geography, age, and subcategory based on population and hospital cancer registries in Hebei, China (2008–2017). Thorac. Cancer 2020, 11, 2087–2093. [Google Scholar] [CrossRef] [PubMed]
  66. Zhang, Y.; Xu, G.; Zhang, S.; Wang, D.; Saravana Prabha, P.; Zuo, Z. Antitumor Research on Artemisinin and Its Bioactive Derivatives. Nat. Prod. Bioprospect. 2018, 8, 303–319. [Google Scholar] [CrossRef] [PubMed]
  67. Shakeel, I.; Haider, S.; Khan, S.; Ahmed, S.; Hussain, A.; Alajmi, M.F.; Chakrabarty, A.; Afzal, M.; Imtaiyaz Hassan, M. Thymoquinone, artemisinin, and thymol attenuate proliferation of lung cancer cells as Sphingosine kinase 1 inhibitors. Biomed. Pharmacother. 2024, 177, 117123. [Google Scholar] [CrossRef]
  68. Lu, Z.; Jiang, J.; Yao, X.; Hou, G. Network pharmacological mechanism and molecular experimental validation of artemisinin in the treatment of lung adenocarcinoma. Toxicol. Appl. Pharmacol. 2025, 495, 117226. [Google Scholar] [CrossRef]
  69. Ni, L.; Zhu, X.; Zhao, Q.; Shen, Y.; Tao, L.; Zhang, J.; Lin, H.; Zhuge, W.; Cho, Y.C.; Cui, R.; et al. Dihydroartemisinin, a potential PTGS1 inhibitor, potentiated cisplatin-induced cell death in non-small cell lung cancer through activating ROS-mediated multiple signaling pathways. Neoplasia 2024, 51, 100991. [Google Scholar] [CrossRef]
  70. Li, Y.; Sun, H.; Bai, C.; Hu, Y.; Tang, J.; Zhang, Y.; Chen, J.; Zhong, Z.; He, Y.; Hu, K.; et al. Dihydroartemisinin inhibits tumor progress via blocking ROR1-induced STAT3-activation in non-small cell lung cancer. Int. Immunopharmacol. 2024, 133, 112157. [Google Scholar] [CrossRef]
  71. Wang, Q.; Zhou, J.; Cheng, A.; Liu, Y.; Guo, J.; Li, X.; Chen, M.; Hu, D.; Wu, J. Artesunate-binding FABP5 promotes apoptosis in lung cancer cells via the PPARgamma-SCD pathway. Int. Immunopharmacol. 2024, 143 Pt 1, 113381. [Google Scholar] [CrossRef]
  72. Wang, J.S.; Wang, M.J.; Lu, X.; Zhang, J.; Liu, Q.X.; Zhou, D.; Dai, J.G.; Zheng, H. Artesunate inhibits epithelial-mesenchymal transition in non-small-cell lung cancer (NSCLC) cells by down-regulating the expression of BTBD7. Bioengineered 2020, 11, 1197–1207. [Google Scholar] [CrossRef] [PubMed]
  73. Llovet, J.M.; Kelley, R.K.; Villanueva, A.; Singal, A.G.; Pikarsky, E.; Roayaie, S.; Lencioni, R.; Koike, K.; Zucman-Rossi, J.; Finn, R.S. Hepatocellular carcinoma. Nat. Rev. Dis. Primers 2021, 7, 6. [Google Scholar] [CrossRef] [PubMed]
  74. Eresen, A.; Yang, J.; Scotti, A.; Cai, K.; Yaghmai, V.; Zhang, Z. Combination of natural killer cell-based immunotherapy and irreversible electroporation for the treatment of hepatocellular carcinoma. Ann. Transl. Med. 2021, 9, 1089. [Google Scholar] [CrossRef] [PubMed]
  75. Gao, Y.; Gong, Y.; Lu, J.; Yang, Y.; Zhang, Y.; Xiong, Y.; Shi, X. 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. [Google Scholar] [CrossRef]
  76. Ji, J.; Cheng, Z.; Zhang, J.; Wu, J.; Xu, X.; Guo, C.; Feng, J. Dihydroartemisinin induces ferroptosis of hepatocellular carcinoma via inhibiting ATF4-xCT pathway. J. Cell. Mol. Med. 2024, 28, e18335. [Google Scholar] [CrossRef]
  77. Nandi, D.; Cheema, P.S.; Singal, A.; Bharti, H.; Nag, A. Artemisinin Mediates Its Tumor-Suppressive Activity in Hepatocellular Carcinoma Through Targeted Inhibition of FoxM1. Front. Oncol. 2021, 11, 751271. [Google Scholar] [CrossRef]
  78. Ma, Z.; Chen, W.; Liu, Y.; Yu, L.; Mao, X.; Guo, X.; Jiang, F.; Guo, Q.; Lin, N.; Zhang, Y. Artesunate Sensitizes human hepatocellular carcinoma to sorafenib via exacerbating AFAP1L2-SRC-FUNDC1 axis-dependent mitophagy. Autophagy 2024, 20, 541–556. [Google Scholar] [CrossRef]
  79. Wang, F.; Meszoely, I.; Pal, T.; Mayer, I.A.; Bailey, C.E.; Zheng, W.; Shu, X.O. Radiotherapy after breast-conserving surgery for elderly patients with early-stage breast cancer: A national registry-based study. Int. J. Cancer 2021, 148, 857–867. [Google Scholar] [CrossRef]
  80. Nolan, E.; Lindeman, G.J.; Visvader, J.E. Deciphering breast cancer: From biology to the clinic. Cell 2023, 186, 1708–1728. [Google Scholar] [CrossRef]
  81. Zhang, X.; Li, N.; Zhang, G.; Li, J.; Liu, Y.; Wang, M.; Ren, X. Nano Strategies for Artemisinin Derivatives to Enhance Reverse Efficiency of Multidrug Resistance in Breast Cancer. Curr. Pharm. Des. 2023, 29, 3458–3466. [Google Scholar] [CrossRef]
  82. Zhang, Y.; Cao, S.; Zeng, F.; Pan, D.; Cai, L.; Zhou, Y.; Wang, H.; Qin, G.; Zhang, C.; Chen, W. Dihydroartemisinin enhances the radiosensitivity of breast cancer by targeting ferroptosis signaling pathway through hsa_circ_0001610. Eur. J. Pharmacol. 2024, 983, 176943. [Google Scholar] [CrossRef] [PubMed]
  83. Dong, H.Y.; Wang, Z.F. Antitumor effects of artesunate on human breast carcinoma MCF-7 cells and IGF-IR expression in nude mice xenografts. Chin. J. Cancer Res. 2014, 26, 200–207. [Google Scholar] [PubMed]
  84. Gu, L.; Zhang, J.; Liu, D.; Chen, J.; Liu, S.; Peng, Q.; Tian, Y.; Du, M.; Zhang, J.; Xiao, W.; et al. Development of artesunate intelligent prodrug liposomes based on mitochondrial targeting strategy. J. Nanobiotechnol. 2022, 20, 376. [Google Scholar] [CrossRef] [PubMed]
  85. Favoriti, P.; Carbone, G.; Greco, M.; Pirozzi, F.; Pirozzi, R.E.; Corcione, F. Worldwide burden of colorectal cancer: A review. Updates Surg. 2016, 68, 7–11. [Google Scholar] [CrossRef]
  86. Brenner, H. Long-term survival rates of cancer patients achieved by the end of the 20th century: A period analysis. Lancet 2002, 360, 1131–1135. [Google Scholar] [CrossRef]
  87. Kumar, M.S.; Yadav, T.T.; Khair, R.R.; Peters, G.J.; Yergeri, M.C. Combination Therapies of Artemisinin and its Derivatives as a Viable Approach for Future Cancer Treatment. Curr. Pharm. Des. 2019, 25, 3323–3338. [Google Scholar] [CrossRef]
  88. Geng, Y.; Li, W.; Wong, N.K.; Xue, F.; Li, Q.; Zhang, Y.; Xu, J.; Deng, Z.; Zhou, Y. Discovery of Artemisinins as Microsomal Prostaglandins Synthase-2 Inhibitors for the Treatment of Colorectal Cancer via Chemoproteomics. J. Med. Chem. 2024, 67, 2083–2094. [Google Scholar] [CrossRef]
  89. Dai, X.; Chen, W.; Qiao, Y.; Chen, X.; Chen, Y.; Zhang, K.; Zhang, Q.; Duan, X.; Li, X.; Zhao, J.; et al. Dihydroartemisinin inhibits the development of colorectal cancer by GSK-3beta/TCF7/MMP9 pathway and synergies with capecitabine. Cancer Lett. 2024, 582, 216596. [Google Scholar] [CrossRef]
  90. Wei, Y.; Liu, F.; Zhu, X.; Liu, X.; Li, H.; Hou, L.; Ma, X.; Li, F.; Liu, H. Artesunate disrupts ribosome RNA biogenesis and inhibits ovarian cancer growth by targeting FANCA. Phytomedicine 2025, 136, 156333. [Google Scholar] [CrossRef]
  91. Zhang, W.; Wang, Y.; Chen, L.; Chen, H.; Qi, H.; Zheng, Y.; Du, Y.; Zhang, L.; Wang, T.; Li, Q. Dihydroartemisinin suppresses glioma growth by repressing ERRalpha-mediated mitochondrial biogenesis. Mol. Cell. Biochem. 2024, 479, 2809–2825. [Google Scholar] [CrossRef] [PubMed]
  92. Strik, H.; Efferth, T.; Kaina, B. Artesunate in glioblastoma therapy: Case reports and review of clinical studies. Phytomedicine 2024, 123, 155274. [Google Scholar] [CrossRef]
  93. Yang, J.; Xia, T.; Zhou, S.; Liu, S.; Pan, T.; Li, Y.; Luo, Z. Anticancer Effect of Dihydroartemisinin via Dual Control of ROS-induced Apoptosis and Protective Autophagy in Prostate Cancer 22Rv1 Cells. Curr. Pharm. Biotechnol. 2024, 25, 1321–1332. [Google Scholar] [CrossRef]
  94. Zhou, Z.; Farhan, M.; Xing, X.; Zhou, W.; Lin, R.; Zeng, S.; Li, M.; Zheng, W. Artemisinin Suppressed Melanoma Recurrence and Metastasis after Radical Surgery through the KIT/PI3K/AKT Pathway. Int. J. Biol. Sci. 2025, 21, 75–94. [Google Scholar] [CrossRef]
  95. Kumar, D.N.; Chaudhuri, A.; Dehari, D.; Gamper, A.M.; Kumar, D.; Agrawal, A.K. Oral delivery of dihydroartemisinin for the treatment of melanoma via bovine milk exosomes. Drug Deliv. Transl. Res. 2025, 15, 2862–2877. [Google Scholar] [CrossRef]
  96. Liu, W.; Zhou, H.; Lai, W.; Hu, C.; Wang, Q.; Yuan, C.; Luo, C.; Yang, M.; Hu, M.; Zhang, R.; et al. Artesunate induces melanoma cell ferroptosis and augments antitumor immunity through targeting Ido1. Cell Commun. Signal. 2024, 22, 378. [Google Scholar] [CrossRef]
  97. RoyMahapatra, D.; Singh, R.; Sk, U.H.; Manna, P.P. Engineered Artesunate-Naphthalimide Hybrid Dual Drug for Synergistic Multimodal Therapy against Experimental Murine Lymphoma. Mol. Pharm. 2024, 21, 1090–1107. [Google Scholar] [CrossRef]
  98. Wang, H.; Ding, Q.; Zhou, H.; Huang, C.; Liu, G.; Zhao, X.; Cheng, Z.; You, X. Dihydroartemisinin inhibited vasculogenic mimicry in gastric cancer through the FGF2/FGFR1 signaling pathway. Phytomedicine 2024, 134, 155962. [Google Scholar] [CrossRef]
  99. Xu, D.L.; Fan, K.; Zhang, H.; Tang, L.X.; Wang, Y.; Xiang, Z.; Shi, A.M.; Qu, Y.C.; Su, C.J.; Pan, J. Anti-proliferation and apoptosis-inducing effects of dihydroartemisinin on SH-SY5Y cells and metabolomic analysis. Transl. Pediatr. 2022, 11, 1346–1361. [Google Scholar] [CrossRef] [PubMed]
  100. Michaelis, M.; Kleinschmidt, M.C.; Barth, S.; Rothweiler, F.; Geiler, J.; Breitling, R.; Mayer, B.; Deubzer, H.; Witt, O.; Kreuter, J.; et al. Anti-cancer effects of artesunate in a panel of chemoresistant neuroblastoma cell lines. Biochem. Pharmacol. 2010, 79, 130–136. [Google Scholar] [CrossRef] [PubMed]
  101. Tan, W.Q.; Chen, G.; Ye, M.; Jia, B. Artemether Regulates Chemosensitivity to Doxorubicin via Regulation of B7-H3 in Human Neuroblastoma Cells. Med. Sci. Monit. 2017, 23, 4252–4259. [Google Scholar] [CrossRef]
  102. Yu, X.H.; Wu, J.B.; Fan, H.Y.; Dai, L.; Xian, H.C.; Chen, B.J.; Liao, P.; Huang, M.C.; Pang, X.; Zhang, M.; et al. Artemisinin suppressed tumour growth and induced vascular normalisation in oral squamous cell carcinoma via inhibition of macrophage migration inhibitory factor. Oral Dis. 2024, 30, 363–375. [Google Scholar] [CrossRef] [PubMed]
  103. Wu, X.; Yang, C.; Li, Z.; Lv, P.; An, X.; Peng, X.; Li, Y.; Jiang, X.; Mao, X.; Chen, D.; et al. Dihydroartemisinin inhibits HNSCC invasion and migration by controlling miR-195-5p expression. Heliyon 2024, 10, e32522. [Google Scholar] [CrossRef] [PubMed]
  104. Thongchot, S.; Vidoni, C.; Ferraresi, A.; Loilome, W.; Yongvanit, P.; Namwat, N.; Isidoro, C. Dihydroartemisinin induces apoptosis and autophagy-dependent cell death in cholangiocarcinoma through a DAPK1-BECLIN1 pathway. Mol. Carcinog. 2018, 57, 1735–1750. [Google Scholar] [CrossRef] [PubMed]
  105. Ding, X.; Zhang, Y.; Liang, J.; Li, Q.; Hu, H.; Zhou, Y.; Zhang, B. Dihydroartemisinin Potentiates VEGFR-TKIs Antitumorigenic Effect on Osteosarcoma by Regulating Loxl2/VEGFA Expression and Lipid Metabolism Pathway. J. Cancer 2023, 14, 809–820. [Google Scholar] [CrossRef]
  106. Liu, Y.; Wang, W.; Xu, J.; Li, L.; Dong, Q.; Shi, Q.; Zuo, G.; Zhou, L.; Weng, Y.; Tang, M.; et al. Dihydroartemisinin inhibits tumor growth of human osteosarcoma cells by suppressing Wnt/beta-catenin signaling. Oncol. Rep. 2013, 30, 1723–1730. [Google Scholar] [CrossRef]
  107. Luo, J.; Odaka, Y.; Huang, Z.; Cheng, B.; Liu, W.; Li, L.; Shang, C.; Zhang, C.; Wu, Y.; Luo, Y.; et al. Dihydroartemisinin Inhibits mTORC1 Signaling by Activating the AMPK Pathway in Rhabdomyosarcoma Tumor Cells. Cells 2021, 10, 1363. [Google Scholar] [CrossRef]
  108. Youns, M.; Efferth, T.; Reichling, J.; Fellenberg, K.; Bauer, A.; Hoheisel, J.D. Gene expression profiling identifies novel key players involved in the cytotoxic effect of Artesunate on pancreatic cancer cells. Biochem. Pharmacol. 2009, 78, 273–283. [Google Scholar] [CrossRef]
  109. Wang, K.; Zhang, Z.; Wang, M.; Cao, X.; Qi, J.; Wang, D.; Gong, A.; Zhu, H. Role of GRP78 inhibiting artesunate-induced ferroptosis in KRAS mutant pancreatic cancer cells. Drug Des. Dev. Ther. 2019, 13, 2135–2144. [Google Scholar] [CrossRef]
  110. Botrous, S.; Elmaghraby, A.; Achy, S.E.; Mustafa, Y.; Abdel-Rahman, S. Artemisinin pre-treatment fore cisplatin dosage enhances high grade urothelial carcinoma treatment in male albino mice via reverse gene expression modulation of FGFR3, HRAS, P53 and KDM6A. BMC Cancer 2024, 24, 971. [Google Scholar] [CrossRef]
  111. Shi, H.; Xiong, L.; Yan, G.; Du, S.; Liu, J.; Shi, Y. Susceptibility of cervical cancer to dihydroartemisinin-induced ferritinophagy-dependent ferroptosis. Front. Mol. Biosci. 2023, 10, 1156062. [Google Scholar] [CrossRef]
  112. Hu, C.J.; Zhou, L.; Cai, Y. Dihydroartemisinin induces apoptosis of cervical cancer cells via upregulation of RKIP and downregulation of bcl-2. Cancer Biol. Ther. 2014, 15, 279–288. [Google Scholar] [CrossRef]
  113. Wang, L.; Li, J.; Shi, X.; Li, S.; Tang, P.M.; Li, Z.; Li, H.; Wei, C. Antimalarial Dihydroartemisinin triggers autophagy within HeLa cells of human cervical cancer through Bcl-2 phosphorylation at Ser70. Phytomedicine 2019, 52, 147–156. [Google Scholar] [CrossRef] [PubMed]
  114. Disbrow, G.L.; Baege, A.C.; Kierpiec, K.A.; Yuan, H.; Centeno, J.A.; Thibodeaux, C.A.; Hartmann, D.; Schlegel, R. Dihydroartemisinin is cytotoxic to papillomavirus-expressing epithelial cells in vitro and in vivo. Cancer Res. 2005, 65, 10854–10861. [Google Scholar] [CrossRef] [PubMed]
  115. Zhang, L.X.; Liu, Z.N.; Ye, J.; Sha, M.; Qian, H.; Bu, X.H.; Luan, Z.Y.; Xu, X.L.; Huang, A.H.; Yuan, D.L.; et al. Artesunate exerts an anti-immunosuppressive effect on cervical cancer by inhibiting PGE2 production and Foxp3 expression. Cell Biol. Int. 2014, 38, 639–646. [Google Scholar] [CrossRef] [PubMed]
  116. Thanaketpaisarn, O.; Waiwut, P.; Sakurai, H.; Saiki, I. Artesunate enhances TRAIL-induced apoptosis in human cervical carcinoma cells through inhibition of the NF-kappaB and PI3K/Akt signaling pathways. Int. J. Oncol. 2011, 39, 279–285. [Google Scholar]
  117. Yang, C.; Wei, W.; Hu, F.; Zhao, X.; Yang, H.; Song, X.; Sun, Z. Dihydroartemisinin suppresses the tumorigenesis of esophageal carcinoma by elevating DAB2IP expression in a NFIC-dependent manner. Naunyn Schmiedebergs Arch. Pharmacol. 2024, 397, 8117–8128. [Google Scholar] [CrossRef]
  118. Xia, Y.; Tang, Y.; Huang, Z.; Ke, N.; Zheng, Y.; Zhuang, W.; Zhang, Y.; Yin, X.; Tu, M.; Chen, J.; et al. Artesunate-loaded solid lipid nanoparticles resist esophageal squamous cell carcinoma by inducing Ferroptosis through inhibiting the AKT/mTOR signaling. Cell. Signal. 2024, 117, 111108. [Google Scholar] [CrossRef]
  119. Yang, F.; Zhang, J.; Zhao, Z.; Liu, Y.; Zhao, Z.; Fu, K.; Li, B.; Jin, J. Artemisinin suppresses aerobic glycolysis in thyroid cancer cells by downregulating HIF-1a, which is increased by the XIST/miR-93/HIF-1a pathway. PLoS ONE 2023, 18, e0284242. [Google Scholar] [CrossRef]
  120. Chauhan, A.K.; Min, K.-J.; Kwon, T.K. RIP1-dependent reactive oxygen species production executes artesunate-induced cell death in renal carcinoma Caki cells. Mol. Cell. Biochem. 2017, 435, 15–24. [Google Scholar] [CrossRef]
  121. Huang, C.F.; Lin, S.S.; Liao, P.H.; Young, S.C.; Yang, C.C. The immunopharmaceutical effects and mechanisms of herb medicine. Cell. Mol. Immunol. 2008, 5, 23–31. [Google Scholar] [CrossRef] [PubMed]
  122. Langroudi, L.; Hassan, Z.M.; Ebtekar, M.; Mahdavi, M.; Pakravan, N.; Noori, S. A comparison of low-dose cyclophosphamide treatment with artemisinin treatment in reducing the number of regulatory T cells in murine breast cancer model. Int. Immunopharmacol. 2010, 10, 1055–1061. [Google Scholar] [CrossRef] [PubMed]
  123. Mancuso, R.I.; Azambuja, J.H.; Olalla Saad, S.T. Artesunate strongly modulates myeloid and regulatory T cells to prevent LPS-induced systemic inflammation. Biomed. Pharmacother. 2021, 143, 112211. [Google Scholar] [CrossRef] [PubMed]
  124. Wang, J.X.; Tang, W.; Shi, L.P.; Wan, J.; Zhou, R.; Ni, J.; Fu, Y.F.; Yang, Y.F.; Li, Y.; Zuo, J.P. Investigation of the immunosuppressive activity of artemether on T-cell activation and proliferation. Br. J. Pharmacol. 2007, 150, 652–661. [Google Scholar] [CrossRef]
  125. Hou, L.F.; He, S.J.; Wang, J.X.; Yang, Y.; Zhu, F.H.; Zhou, Y.; He, P.L.; Zhang, Y.; Yang, Y.F.; Li, Y.; et al. SM934, a water-soluble derivative of arteminisin, exerts immunosuppressive functions in vitro and in vivo. Int. Immunopharmacol. 2009, 9, 1509–1517. [Google Scholar] [CrossRef]
  126. Li, Y.; Shan, Y.; Xu, L.; Chen, W.; Li, Y. Dihydroartemisinin ameliorates experimental autoimmune myasthenia gravis by regulating CD4(+) T cells and modulating gut microbiota. Int. Immunopharmacol. 2024, 139, 112699. [Google Scholar] [CrossRef]
  127. Zhu, S.; Cui, Y.; Hu, H.; Zhang, C.; Chen, K.; Shan, Z.; Teng, W.; Li, J. Dihydroartemisinin inhibits the development of autoimmune thyroiditis by modulating oxidative stress and immune imbalance. Free Radic. Biol. Med. 2025, 231, 57–67. [Google Scholar] [CrossRef]
  128. Wang, C.; Jiang, X.; Lv, J.; Zhuang, W.; Xie, L.; Liu, G.; Saimaier, K.; Han, S.; Shi, C.; Hua, Q.; et al. TPN10475 Constrains Effector T Lymphocytes Activation and Attenuates Experimental Autoimmune Encephalomyelitis Pathogenesis by Facilitating TGF-β Signal Transduction. J. Neuroimmune Pharmacol. 2024, 19, 6. [Google Scholar] [CrossRef]
  129. Li, Q.; Jiang, N.; Zhang, Y.; Liu, Y.; Su, Z.; Yuan, Q.; Sang, X.; Chen, R.; Feng, Y.; Chen, Q. Dihydroartemisinin imposes positive and negative regulation on Treg and plasma cells via direct interaction and activation of c-Fos. Commun. Biol. 2023, 6, 52. [Google Scholar] [CrossRef]
  130. Yang, Z.; Han, F.; Liao, T.; Zheng, H.; Luo, Z.; Ma, M.; He, J.; Li, L.; Ye, Y.; Zhang, R.; et al. Artemisinin Attenuates Transplant Rejection by Inhibiting Multiple Lymphocytes and Prolongs Cardiac Allograft Survival. Front. Immunol. 2021, 12, 634368. [Google Scholar] [CrossRef]
  131. Shi, X.; Liao, T.; Chen, Y.; Chen, J.; Liu, Y.; Zhao, J.; Dang, J.; Sun, Q.; Pan, Y. Dihydroartemisinin inhibits follicular helper T and B cells: Implications for systemic lupus erythematosus treatment. Arch. Pharm. Res. 2024, 47, 632–644. [Google Scholar] [CrossRef]
  132. Zhang, T.; Zhang, Y.; Jiang, N.; Zhao, X.; Sang, X.; Yang, N.; Feng, Y.; Chen, R.; Chen, Q. Dihydroartemisinin regulates the immune system by promotion of CD8+ T lymphocytes and suppression of B cell responses. Sci. China Life Sci. 2020, 63, 737–749. [Google Scholar] [CrossRef]
  133. Zeng, X.Z.; Zhang, Y.Y.; Yang, Q.; Wang, S.; Zou, B.H.; Tan, Y.H.; Zou, M.; Liu, S.W.; Li, X.J. Artesunate attenuates LPS-induced osteoclastogenesis by suppressing TLR4/TRAF6 and PLCγ1-Ca(2+)-NFATc1 signaling pathway. Acta Pharmacol. Sin. 2020, 41, 229–236. [Google Scholar] [CrossRef] [PubMed]
  134. Liu, L.; Zhao, J.; Li, A.; Yang, X.; Sprangers, B.; Li, S. Artemisinin attenuates IgM xenoantibody production via inhibition of T cell-independent marginal zone B cell proliferation. J. Leukoc. Biol. 2021, 109, 583–591. [Google Scholar] [CrossRef] [PubMed]
  135. Fujiwara, N.; Kobayashi, K. Macrophages in inflammation. Curr. Drug Targets Inflamm. Allergy 2005, 4, 281–286. [Google Scholar] [CrossRef]
  136. Tao, Y.; Xu, L.; Liu, X.; Wang, P.; Wei, S.; Huang, Y.; Gu, W.; Bo, R.; Liu, M.; Yu, J.; et al. Chitosan-coated artesunate protects against ulcerative colitis via STAT6-mediated macrophage M2 polarization and intestinal barrier protection. Int. J. Biol. Macromol. 2024, 254 Pt 1, 127680. [Google Scholar] [CrossRef] [PubMed]
  137. Liu, D.; Liu, C.; Deng, F.; Ouyang, F.; Qin, R.; Zhai, Z.; Wang, Y.; Zhang, Y.; Liao, M.; Pan, X.; et al. Artesunate protects against a mouse model of cerulein and lipopolysaccharide-induced acute pancreatitis by inhibiting TLR4-dependent autophagy. Int. J. Mol. Med. 2025, 55, 25. [Google Scholar] [CrossRef]
  138. Li, Y.; Liang, Q.; Zhou, L.; Cao, Y.; Yang, J.; Li, J.; Liu, J.; Bi, J.; Liu, Y. An ROS-responsive artesunate prodrug nanosystem co-delivers dexamethasone for rheumatoid arthritis treatment through the HIF-1alpha/NF-kappaB cascade regulation of ROS scavenging and macrophage repolarization. Acta Biomater. 2022, 152, 406–424. [Google Scholar] [CrossRef]
  139. Ji, Y.; Sun, K.; Yang, Y.; Wu, Z. Dihydroartemisinin ameliorates innate inflammatory response induced by Streptococcussuis-derived muramidase-released protein via inactivation of TLR4-dependent NF-kappaB signaling. J. Pharm. Anal. 2023, 13, 1183–1194. [Google Scholar] [CrossRef]
  140. Yang, F.M.; Fan, D.; Yang, X.Q.; Zhu, F.H.; Shao, M.J.; Li, Q.; Liu, Y.T.; Lin, Z.M.; Cao, S.Q.; Tang, W.; et al. The artemisinin analog SM934 alleviates dry eye disease in rodent models by regulating TLR4/NF-κB/NLRP3 signaling. Acta Pharmacol. Sin. 2021, 42, 593–603. [Google Scholar] [CrossRef]
  141. Huai, M.; Zeng, J.; Ge, W. Artemisinin ameliorates intestinal inflammation by skewing macrophages to the M2 phenotype and inhibiting epithelial-mesenchymal transition. Int. Immunopharmacol. 2021, 91, 107284. [Google Scholar] [CrossRef]
  142. Sun, W.; Han, X.; Wu, S.; Wu, J.; Yang, C.; Li, X. Unexpected mechanism of colitis amelioration by artesunate, a natural product from Artemisia annua L. Inflammopharmacology 2020, 28, 851–868. [Google Scholar] [CrossRef]
  143. Houh, Y.K.; Kim, K.E.; Park, S.; Hur, D.Y.; Kim, S.; Kim, D.; Bang, S.I.; Yang, Y.; Park, H.J.; Cho, D. The Effects of Artemisinin on the Cytolytic Activity of Natural Killer (NK) Cells. Int. J. Mol. Sci. 2017, 18, 1600. [Google Scholar] [CrossRef] [PubMed]
  144. Lu, Z.; Bi, J.; Wan, X. Artemisinin sensitizes tumor cells to NK cell-mediated cytolysis. Biochem. Biophys. Res. Commun. 2020, 524, 418–423. [Google Scholar] [CrossRef] [PubMed]
  145. Izmirly, P.M.; Parton, H.; Wang, L.; McCune, W.J.; Lim, S.S.; Drenkard, C.; Ferucci, E.D.; Dall’Era, M.; Gordon, C.; Helmick, C.G.; et al. Prevalence of Systemic Lupus Erythematosus in the United States: Estimates From a Meta-Analysis of the Centers for Disease Control and Prevention National Lupus Registries. Arthritis Rheumatol. 2021, 73, 991–996. [Google Scholar] [CrossRef] [PubMed]
  146. Tian, J.; Zhang, D.; Yao, X.; Huang, Y.; Lu, Q. Global epidemiology of systemic lupus erythematosus: A comprehensive systematic analysis and modelling study. Ann. Rheum. Dis. 2023, 82, 351–356. [Google Scholar] [CrossRef]
  147. Fanouriakis, A.; Tziolos, N.; Bertsias, G.; Boumpas, D.T. Update οn the diagnosis and management of systemic lupus erythematosus. Ann. Rheum. Dis. 2021, 80, 14–25. [Google Scholar] [CrossRef]
  148. Morand, E.F.; Furie, R.; Tanaka, Y.; Bruce, I.N.; Askanase, A.D.; Richez, C.; Bae, S.C.; Brohawn, P.Z.; Pineda, L.; Berglind, A.; et al. Trial of Anifrolumab in Active Systemic Lupus Erythematosus. N. Engl. J. Med. 2020, 382, 211–221. [Google Scholar] [CrossRef]
  149. Chen, Y.; Tao, T.; Wang, W.; Yang, B.; Cha, X. Dihydroartemisinin attenuated the symptoms of mice model of systemic lupus erythematosus by restoring the Treg/Th17 balance. Clin. Exp. Pharmacol. Physiol. 2021, 48, 626–633. [Google Scholar] [CrossRef]
  150. Zhou, F.; Li, G.; Tan, R.; Wu, G.; Deng, C. Artemisinins in autoimmune diseases: Effects and mechanisms in systemic lupus erythematosus and rheumatoid arthritis. Br. J. Pharmacol. 2025, 182, 3411–3427. [Google Scholar] [CrossRef]
  151. Hou, L.F.; He, S.J.; Li, X.; Yang, Y.; He, P.L.; Zhou, Y.; Zhu, F.H.; Yang, Y.F.; Li, Y.; Tang, W.; et al. Oral administration of artemisinin analog SM934 ameliorates lupus syndromes in MRL/lpr mice by inhibiting Th1 and Th17 cell responses. Arthritis Rheum. 2011, 63, 2445–2455. [Google Scholar] [CrossRef]
  152. Nieuwenhuis, W.P.; van Steenbergen, H.W.; Stomp, W.; Stijnen, T.; Huizinga, T.W.; Bloem, J.L.; van der Heijde, D.; Reijnierse, M.; van der Helm-van Mil, A.H. The Course of Bone Marrow Edema in Early Undifferentiated Arthritis and Rheumatoid Arthritis: A Longitudinal Magnetic Resonance Imaging Study at Bone Level. Arthritis Rheumatol. 2016, 68, 1080–1088. [Google Scholar] [CrossRef]
  153. Rani, R.; Raina, N.; Sharma, A.; Kumar, P.; Tulli, H.S.; Gupta, M. Advancement in nanotechnology for treatment of rheumatoid arthritis: Scope and potential applications. Naunyn Schmiedebergs Arch. Pharmacol. 2023, 396, 2287–2310. [Google Scholar] [CrossRef] [PubMed]
  154. Fan, M.; Li, Y.; Yao, C.; Liu, X.; Liu, J.; Yu, B. DC32, a Dihydroartemisinin Derivative, Ameliorates Collagen-Induced Arthritis Through an Nrf2-p62-Keap1 Feedback Loop. Front. Immunol. 2018, 9, 2762. [Google Scholar] [CrossRef]
  155. Chen, J.; Lin, X.; He, J.; Liu, D.; He, L.; Zhang, M.; Luan, H.; Hu, Y.; Tao, C.; Wang, Q. Artemisitene suppresses rheumatoid arthritis progression via modulating METTL3-mediated N6-methyladenosine modification of ICAM2 mRNA in fibroblast-like synoviocytes. Clin. Transl. Med. 2022, 12, e1148. [Google Scholar] [CrossRef] [PubMed]
  156. Lin, Z.M.; Yang, X.Q.; Zhu, F.H.; He, S.J.; Tang, W.; Zuo, J.P. Artemisinin analogue SM934 attenuate collagen-induced arthritis by suppressing T follicular helper cells and T helper 17 cells. Sci. Rep. 2016, 6, 38115. [Google Scholar] [CrossRef]
  157. Liu, J.; Hong, X.; Lin, D.; Luo, X.; Zhu, M.; Mo, H. Artesunate influences Th17/Treg lymphocyte balance by modulating Treg apoptosis and Th17 proliferation in a murine model of rheumatoid arthritis. Exp. Ther. Med. 2017, 13, 2267–2273. [Google Scholar] [CrossRef]
  158. Griffiths, C.E.M.; Armstrong, A.W.; Gudjonsson, J.E.; Barker, J. Psoriasis. Lancet 2021, 397, 1301–1315. [Google Scholar] [CrossRef]
  159. Chen, Y.; Yan, Y.; Liu, H.; Qiu, F.; Liang, C.L.; Zhang, Q.; Huang, R.Y.; Han, L.; Lu, C.; Dai, Z. Dihydroartemisinin ameliorates psoriatic skin inflammation and its relapse by diminishing CD8+ T-cell memory in wild-type and humanized mice. Theranostics 2020, 10, 10466–10482. [Google Scholar] [CrossRef]
  160. Huang, Z.Z.; Xu, Y.; Xu, M.; Shi, Z.R.; Mai, S.Z.; Guo, Z.X.; Tang, Z.Q.; Luo, Y.J.; Guo, Q.; Xiong, H. Artesunate alleviates imiquimod-induced psoriasis-like dermatitis in BALB/c mice. Int. Immunopharmacol. 2019, 75, 105817. [Google Scholar] [CrossRef] [PubMed]
  161. Lin, L.; Huang, Z.; Jianchi, M.; Guo, Z.; Shi, Z.; Tang, Z.; Guo, Q.; Xiong, H. Artesunate alleviates psoriasis-like dermatitis by reducing interleukin-23 expression in tumor necrosis factor-alpha-induced HaCaT cells. Clin. Exp. Pharmacol. Physiol. 2023, 50, 903–913. [Google Scholar] [CrossRef]
  162. Thompson, A.J.; Baranzini, S.E.; Geurts, J.; Hemmer, B.; Ciccarelli, O. Multiple sclerosis. Lancet 2018, 391, 1622–1636. [Google Scholar] [CrossRef]
  163. Khakzad, M.R.; Ganji, A.; Ariabod, V.; Farahani, I. Artemisinin therapeutic efficacy in the experimental model of multiple sclerosis. Immunopharmacol. Immunotoxicol. 2017, 39, 348–353. [Google Scholar] [CrossRef]
  164. Thomé, R.; de Carvalho, A.C.; Alves da Costa, T.; Ishikawa, L.L.; Fraga-Silva, T.F.; Sartori, A.; de Oliveira, A.L.; Verinaud, L. Artesunate Ameliorates Experimental Autoimmune Encephalomyelitis by Inhibiting Leukocyte Migration to the Central Nervous System. CNS Neurosci. Ther. 2016, 22, 707–714. [Google Scholar] [CrossRef]
  165. Lv, J.; Zhuang, W.; Zhang, Y.; Xie, L.; Xiang, Z.; Zhao, Q.; Jiang, X.; Shen, J.; Du, C. 9,10-Anhydrodehydroartemisinin Attenuates Experimental Autoimmune Encephalomyelitis by Inhibiting Th1 and Th17 Cell Differentiation. Inflammation 2021, 44, 1793–1802. [Google Scholar] [CrossRef] [PubMed]
  166. Wang, C.; Han, M.; Li, X.; Lv, J.; Zhuang, W.; Xie, L.; Liu, G.; Saimaier, K.; Han, S.; Shi, C.; et al. TPN10475 alleviates concanavalin A-induced autoimmune hepatitis by limiting T cell development and function through inhibition of PI3K-AKT pathway. Int. Immunopharmacol. 2023, 125 Pt A, 111110. [Google Scholar] [CrossRef] [PubMed]
  167. Liu, G.; Zhang, Y.; Han, S.; Zhuang, W.; Lv, J.; Han, M.; Xie, L.; Jiang, X.; Wang, C.; Saimaier, K.; et al. TPN10466 ameliorates Concanavalin A-induced autoimmune hepatitis in mice via inhibiting ERK/JNK/p38 signaling pathway. Eur. J. Immunol. 2023, 53, e2250100. [Google Scholar] [CrossRef] [PubMed]
  168. Zhao, Y.G.; Wang, Y.; Guo, Z.; Gu, A.D.; Dan, H.C.; Baldwin, A.S.; Hao, W.; Wan, Y.Y. Dihydroartemisinin ameliorates inflammatory disease by its reciprocal effects on Th and regulatory T cell function via modulating the mammalian target of rapamycin pathway. J. Immunol. 2012, 189, 4417–4425. [Google Scholar] [CrossRef]
  169. Yu, R.; Jin, L.; Li, F.; Fujimoto, M.; Wei, Q.; Lin, Z.; Ren, X.; Jin, Q.; Li, H.; Meng, F.; et al. Dihydroartemisinin inhibits melanoma by regulating CTL/Treg anti-tumor immunity and STAT3-mediated apoptosis via IL-10 dependent manner. J. Dermatol. Sci. 2020, 99, 193–202. [Google Scholar] [CrossRef]
  170. Xiao, F.; Rui, K.; Han, M.; Zou, L.; Huang, E.; Tian, J.; Zhang, L.; Jiang, Q.; Wu, Y.; Lu, L. Artesunate suppresses Th17 response via inhibiting IRF4-mediated glycolysis and ameliorates Sjog̈ren’s syndrome. Signal Transduct. Target. Ther. 2022, 7, 274. [Google Scholar] [CrossRef]
  171. Cao, Q.; Du, H.; Fu, X.; Duan, N.; Liu, C.; Li, X. Artemisinin Attenuated Atherosclerosis in High-Fat Diet-Fed ApoE-/- Mice by Promoting Macrophage Autophagy Through the AMPK/mTOR/ULK1 Pathway. J. Cardiovasc. Pharmacol. 2020, 75, 321–332. [Google Scholar] [CrossRef]
  172. He, L.H.; Gao, J.H.; Yu, X.H.; Wen, F.J.; Luo, J.J.; Qin, Y.S.; Chen, M.X.; Zhang, D.W.; Wang, Z.B.; Tang, C.K. Artesunate inhibits atherosclerosis by upregulating vascular smooth muscle cells-derived LPL expression via the KLF2/NRF2/TCF7L2 pathway. Eur. J. Pharmacol. 2020, 884, 173408. [Google Scholar] [CrossRef]
  173. Huang, X.T.; Liu, W.; Zhou, Y.; Hao, C.X.; Zhou, Y.; Zhang, C.Y.; Sun, C.C.; Luo, Z.Q.; Tang, S.Y. Dihydroartemisinin attenuates lipopolysaccharide-induced acute lung injury in mice by suppressing NF-κB signaling in an Nrf2-dependent manner. Int. J. Mol. Med. 2019, 44, 2213–2222. [Google Scholar] [CrossRef]
  174. Jiang, M.; Zhong, G.; Zhu, Y.; Wang, L.; He, Y.; Sun, Q.; Wu, X.; You, X.; Gao, S.; Tang, D.; et al. Retardant effect of dihydroartemisinin on ulcerative colitis in a JAK2/STAT3-dependent manner. Acta Biochim. Biophys. Sin. 2021, 53, 1113–1123. [Google Scholar] [CrossRef]
  175. Hua, L.; Liang, S.; Zhou, Y.; Wu, X.; Cai, H.; Liu, Z.; Ou, Y.; Chen, Y.; Chen, X.; Yan, Y.; et al. Artemisinin-derived artemisitene blocks ROS-mediated NLRP3 inflammasome and alleviates ulcerative colitis. Int. Immunopharmacol. 2022, 113 Pt B, 109431. [Google Scholar] [CrossRef]
  176. Meng, Y.; Ma, N.; Lyu, H.; Wong, Y.K.; Zhang, X.; Zhu, Y.; Gao, P.; Sun, P.; Song, Y.; Lin, L.; et al. Recent pharmacological advances in the repurposing of artemisinin drugs. Med. Res. Rev. 2021, 41, 3156–3181. [Google Scholar] [CrossRef] [PubMed]
  177. Efferth, T. From ancient herb to modern drug: Artemisia annua and artemisinin for cancer therapy. Semin. Cancer Biol. 2017, 46, 65–83. [Google Scholar] [CrossRef] [PubMed]
  178. Chen, K.; Hua, H.; Zhu, Z.; Wu, T.; Jia, Z.; Liu, Q. Artemisinin and dihydroartemisinin promote β-cell apoptosis induced by palmitate via enhancing ER stress. Apoptosis 2020, 25, 192–204. [Google Scholar] [CrossRef] [PubMed]
  179. Shen, S.; Du, M.; Liu, Q.; Gao, P.; Wang, J.; Liu, S.; Gu, L. Development of GLUT1-targeting alkyl glucoside-modified dihydroartemisinin liposomes for cancer therapy. Nanoscale 2020, 12, 21901–21912. [Google Scholar] [CrossRef]
  180. Wang, Z.; Hao, Y.; Yu, H.; Wei, P. Dihydroartemisinin prevents palmitate-induced β-cell apoptosis. Apoptosis 2021, 26, 147–149. [Google Scholar] [CrossRef]
  181. Chen, J.; Guo, Y.; Zheng, Y.; Chen, Z.; Xu, H.; Pan, S.; Liang, X.; Zhai, L.; Guan, Y.Q. Oral glucose-responsive nanoparticles loaded with artemisinin induce pancreatic beta-cell regeneration for the treatment of type 2 diabetes. J. Colloid. Interface Sci. 2025, 684 Pt 1, 769–782. [Google Scholar] [CrossRef] [PubMed]
  182. Shen, S.; Liao, Q.; Lyu, M.; Wong, Y.K.; Zhang, X.; Zhou, J.; Ma, N.; Wang, J. The potential of artemisinins as anti-obesity agents via modulating the immune system. Pharmacol. Ther. 2020, 216, 107696. [Google Scholar] [CrossRef] [PubMed]
  183. Bai, X.; Pei, R.; Lei, W.; Zhao, M.; Zhang, J.; Tian, L.; Shang, J. Antidiabetic Effect of Artemether in Db/Db Mice Involves Regulation of AMPK and PI3K/Akt Pathways. Front. Endocrinol. 2020, 11, 568864. [Google Scholar] [CrossRef] [PubMed]
  184. Gao, P.; Shen, S.; Li, X.; Liu, D.; Meng, Y.; Liu, Y.; Zhu, Y.; Zhang, J.; Luo, P.; Gu, L. Dihydroartemisinin Inhibits the Proliferation of Leukemia Cells K562 by Suppressing PKM2 and GLUT1 Mediated Aerobic Glycolysis. Drug Des. Dev. Ther. 2020, 14, 2091–2100. [Google Scholar] [CrossRef]
  185. Zhang, H.; Qi, S.; Song, Y.; Ling, C. Artemisinin attenuates early renal damage on diabetic nephropathy rats through suppressing TGF-β1 regulator and activating the Nrf2 signaling pathway. Life Sci. 2020, 256, 117966. [Google Scholar] [CrossRef]
  186. Gao, T.; Yu, C.; Shi, X.; Hu, Y.; Chang, Y.; Zhang, J.; Wang, Y.; Zhai, Z.; Jia, X.; Mao, Y. Artemisinic acid attenuates osteoclast formation and titanium particle-induced osteolysis via inhibition of RANKL-induced ROS accumulation and MAPK and NF-κB signaling pathways. Front. Pharmacol. 2024, 15, 1345380. [Google Scholar] [CrossRef]
  187. Chen, Y.; Mi, Y.; Zhang, X.; Ma, Q.; Song, Y.; Zhang, L.; Wang, D.; Xing, J.; Hou, B.; Li, H.; et al. Dihydroartemisinin-induced unfolded protein response feedback attenuates ferroptosis via PERK/ATF4/HSPA5 pathway in glioma cells. J. Exp. Clin. Cancer Res. 2019, 38, 402. [Google Scholar] [CrossRef]
  188. Li, S.; Peng, T.; Zhao, X.; Silva, M.; Liu, L.; Zhou, W.; Chen, L.; Zheng, W. Artemether confers neuroprotection on cerebral ischemic injury through stimulation of the Erk1/2-P90rsk-CREB signaling pathway. Redox Biol. 2021, 46, 102069. [Google Scholar] [CrossRef]
  189. Peng, T.; Li, S.; Liu, L.; Yang, C.; Farhan, M.; Chen, L.; Su, Q.; Zheng, W. Artemisinin attenuated ischemic stroke induced cell apoptosis through activation of ERK1/2/CREB/BCL-2 signaling pathway in vitro and in vivo. Int. J. Biol. Sci. 2022, 18, 4578–4594. [Google Scholar] [CrossRef]
  190. Zhang, K.; Yang, Y.; Ge, H.; Wang, J.; Chen, X.; Lei, X.; Zhong, J.; Zhang, C.; Xian, J.; Lu, Y.; et al. Artesunate promotes the proliferation of neural stem/progenitor cells and alleviates Ischemia-reperfusion Injury through PI3K/Akt/FOXO-3a/p27(kip1) signaling pathway. Aging 2020, 12, 8029–8048. [Google Scholar] [CrossRef]
  191. Jiang, J.; Geng, G.; Yu, X.; Liu, H.; Gao, J.; An, H.; Cai, C.; Li, N.; Shen, D.; Wu, X.; et al. Repurposing the anti-malarial drug dihydroartemisinin suppresses metastasis of non-small-cell lung cancer via inhibiting NF-κB/GLUT1 axis. Oncotarget 2016, 7, 87271–87283. [Google Scholar] [CrossRef]
  192. Albasher, G.; Aljarba, N.; Al Sultan, N.; Alqahtani, W.S.; Alkahtani, S. Evaluation of the neuro-protective effect of Artemisia judaica extract in a murine diabetic model. J. Food Biochem. 2020, 44, e13337. [Google Scholar] [CrossRef] [PubMed]
  193. Farrag, E.A.E.; Hammad, M.O.; Safwat, S.M.; Hamed, S.; Hellal, D. Artemisinin attenuates type 2 diabetic cardiomyopathy in rats through modulation of AGE-RAGE/HMGB-1 signaling pathway. Sci. Rep. 2023, 13, 11043. [Google Scholar] [CrossRef] [PubMed]
  194. Jiang, W.; Cen, Y.; Song, Y.; Li, P.; Qin, R.; Liu, C.; Zhao, Y.; Zheng, J.; Zhou, H. Artesunate attenuated progression of atherosclerosis lesion formation alone or combined with rosuvastatin through inhibition of pro-inflammatory cytokines and pro-inflammatory chemokines. Phytomedicine 2016, 23, 1259–1266. [Google Scholar] [CrossRef] [PubMed]
  195. Jang, B.C. Artesunate inhibits adipogeneis in 3T3-L1 preadipocytes by reducing the expression and/or phosphorylation levels of C/EBP-alpha, PPAR-gamma, FAS, perilipin A, and STAT-3. Biochem. Biophys. Res. Commun. 2016, 474, 220–225. [Google Scholar] [CrossRef]
  196. Lu, P.; Zhang, F.C.; Qian, S.W.; Li, X.; Cui, Z.M.; Dang, Y.J.; Tang, Q.Q. Artemisinin derivatives prevent obesity by inducing browning of WAT and enhancing BAT function. Cell Res. 2016, 26, 1169–1172. [Google Scholar] [CrossRef]
  197. Guo, X.; Asthana, P.; Zhai, L.; Cheng, K.W.; Gurung, S.; Huang, J.; Wu, J.; Zhang, Y.; Mahato, A.K.; Saarma, M.; et al. Artesunate treats obesity in male mice and non-human primates through GDF15/GFRAL signalling axis. Nat. Commun. 2024, 15, 1034. [Google Scholar] [CrossRef]
  198. Bedard, K.; Krause, K.H. The NOX family of ROS-generating NADPH oxidases: Physiology and pathophysiology. Physiol. Rev. 2007, 87, 245–313. [Google Scholar] [CrossRef]
  199. Den Hartigh, L.J.; Omer, M.; Goodspeed, L.; Wang, S.; Wietecha, T.; O’Brien, K.D.; Han, C.Y. Adipocyte-Specific Deficiency of NADPH Oxidase 4 Delays the Onset of Insulin Resistance and Attenuates Adipose Tissue Inflammation in Obesity. Arterioscler. Thromb. Vasc. Biol. 2017, 37, 466–475. [Google Scholar] [CrossRef]
  200. Hua, H.; Wu, M.; Wu, T.; Ji, Y.; Jin, L.; Du, Y.; Zhang, Y.; Huang, S.; Zhang, A.; Ding, G.; et al. Reduction of NADPH oxidase 4 in adipocytes contributes to the anti-obesity effect of dihydroartemisinin. Heliyon 2023, 9, e14028. [Google Scholar] [CrossRef]
  201. Tsui, K.H.; Wu, M.Y.; Lin, L.T.; Wen, Z.H.; Li, Y.H.; Chu, P.Y.; Li, C.J. Disruption of mitochondrial homeostasis with artemisinin unravels anti-angiogenesis effects via auto-paracrine mechanisms. Theranostics 2019, 9, 6631–6645. [Google Scholar] [CrossRef]
  202. Zhou, H.; You, P.; Liu, H.; Fan, J.; Tong, C.; Yang, A.; Jiang, Y.; Liu, B. Artemisinin and Procyanidins loaded multifunctional nanocomplexes alleviate atherosclerosis via simultaneously modulating lipid influx and cholesterol efflux. J. Control. Release 2022, 341, 828–843. [Google Scholar] [CrossRef] [PubMed]
  203. Wang, X.; Du, H.; Li, X. Artesunate attenuates atherosclerosis by inhibiting macrophage M1-like polarization and improving metabolism. Int. Immunopharmacol. 2022, 102, 108413. [Google Scholar] [CrossRef] [PubMed]
  204. Jiang, Y.; Du, H.; Liu, X.; Fu, X.; Li, X.; Cao, Q. Artemisinin alleviates atherosclerotic lesion by reducing macrophage inflammation via regulation of AMPK/NF-κB/NLRP3 inflammasomes pathway. J. Drug Target. 2020, 28, 70–79. [Google Scholar] [CrossRef] [PubMed]
  205. Wang, Y.H.; Chen, X.; Bai, Y.Z.; Gao, P.; Yang, Z.; Guo, Q.; Lu, Y.Y.; Zheng, J.; Liu, D.; Yang, J.; et al. Palmitoylation of PKCdelta by ZDHHC5 in hypothalamic microglia presents as a therapeutic target for fatty liver disease. Theranostics 2024, 14, 988–1009. [Google Scholar] [CrossRef]
  206. Chen, X.; Xiang, J.; Gao, P.; Wang, L.; Xiang, L.; Lu, Z.; Cao, T.; Mou, A.; Zhang, X.; Jiang, X.; et al. Artemisinin alleviates obesity-related glomerulopathy by downregulating CYP24A1 expression. Diabetes Obes. Metab. 2024, 26, 767–771. [Google Scholar] [CrossRef]
  207. Li, T.; Chen, Y.; Tan, P.; Shi, H.; Huang, Z.; Cai, T.; Cheng, Y.; Du, Y.; Fu, W. Dihydroartemisinin alleviates steatosis and inflammation in nonalcoholic steatohepatitis by decreasing endoplasmic reticulum stress and oxidative stress. Bioorg. Chem. 2022, 122, 105737. [Google Scholar] [CrossRef]
  208. Guo, Y.; Cheng, Y.; Li, H.; Guan, H.; Xiao, H.; Li, Y. The Potential of Artemisinins as Novel Treatment for Thyroid Eye Disease by Inhibiting Adipogenesis in Orbital Fibroblasts. Investig. Ophthalmol. Vis. Sci. 2023, 64, 28. [Google Scholar] [CrossRef]
  209. Yang, C.; Xiong, W.; Dong, J.; Zhao, X.; Liang, G.; Zheng, W. Artemisinin protected human bronchial epithelial cells from amiodarone-induced oxidative damage via 5’-AMP-activated protein kinase (AMPK) activation. Redox Rep. 2025, 30, 2447721. [Google Scholar] [CrossRef]
  210. Yang, C.; Zhao, X.; Zhou, W.; Li, Q.; Lazarovici, P.; Zheng, W. Artemisinin conferred cytoprotection to human retinal pigment epithelial cells exposed to amiodarone-induced oxidative insult by activating the CaMKK2/AMPK/Nrf2 pathway. J. Transl. Med. 2024, 22, 844. [Google Scholar] [CrossRef]
  211. Yu, S.; Yang, N.; Li, H.; Hu, X.; Zhang, L.; Li, S. Artemether ameliorates acetaminophen-induced liver injury through Nrf2 pathway. Biomed. Pharmacother. 2024, 179, 117280. [Google Scholar] [CrossRef]
  212. Zhang, F.; Lu, L.; Ma, S.; Sun, J.; Liu, J.; Gao, N.; Gou, Z.; Zhou, Y.; Lai, C.; Li, Y.; et al. Artemisinin attenuates perinatal inflammation and consequent oxidative stress in oligodendrocyte precursor cells by inhibiting IRAK-4 and IRAK-1. Int. Immunopharmacol. 2024, 142 Pt B, 113117. [Google Scholar] [CrossRef] [PubMed]
  213. Liu, Y.; Jiang, J.J.; Du, S.Y.; Mu, L.S.; Fan, J.J.; Hu, J.C.; Ye, Y.; Ding, M.; Zhou, W.Y.; Yu, Q.H.; et al. Artemisinins ameliorate polycystic ovarian syndrome by mediating LONP1-CYP11A1 interaction. Science 2024, 384, eadk5382. [Google Scholar] [CrossRef] [PubMed]
  214. Bryan, H.K.; Olayanju, A.; Goldring, C.E.; Park, B.K. The Nrf2 cell defence pathway: Keap1-dependent and -independent mechanisms of regulation. Biochem. Pharmacol. 2013, 85, 705–717. [Google Scholar] [CrossRef] [PubMed]
  215. Zhu, M.; Wang, Y.; Han, J.; Sun, Y.; Wang, S.; Yang, B.; Wang, Q.; Kuang, H. Artesunate Exerts Organ- and Tissue-Protective Effects by Regulating Oxidative Stress, Inflammation, Autophagy, Apoptosis, and Fibrosis: A Review of Evidence and Mechanisms. Antioxidants 2024, 13, 686. [Google Scholar] [CrossRef]
  216. Masenga, S.K.; Kabwe, L.S.; Chakulya, M.; Kirabo, A. Mechanisms of Oxidative Stress in Metabolic Syndrome. Int. J. Mol. Sci. 2023, 24, 7898. [Google Scholar] [CrossRef]
  217. Rani, V.; Deep, G.; Singh, R.K.; Palle, K.; Yadav, U.C. Oxidative stress and metabolic disorders: Pathogenesis and therapeutic strategies. Life Sci. 2016, 148, 183–193. [Google Scholar] [CrossRef]
  218. Wu, T.; Feng, H.; He, M.; Yue, R.; Wu, S. Efficacy of artemisinin and its derivatives in animal models of type 2 diabetes mellitus: A systematic review and meta-analysis. Pharmacol. Res. 2022, 175, 105994. [Google Scholar] [CrossRef]
  219. Giacco, F.; Brownlee, M. Oxidative stress and diabetic complications. Circ. Res. 2010, 107, 1058–1070. [Google Scholar] [CrossRef]
  220. Gonzalez, P.; Lozano, P.; Ros, G.; Solano, F. Hyperglycemia and Oxidative Stress: An Integral, Updated and Critical Overview of Their Metabolic Interconnections. Int. J. Mol. Sci. 2023, 24, 9352. [Google Scholar] [CrossRef]
  221. Jiang, Y.Y.; Shui, J.C.; Zhang, B.X.; Chin, J.W.; Yue, R.S. The Potential Roles of Artemisinin and Its Derivatives in the Treatment of Type 2 Diabetes Mellitus. Front. Pharmacol. 2020, 11, 585487. [Google Scholar] [CrossRef]
  222. Chen, L.; Wang, J.; Ren, Y.; Ma, Y.; Liu, J.; Jiang, H.; Liu, C. Artesunate improves glucose and lipid metabolism in db/db mice by regulating the metabolic profile and the MAPK/PI3K/Akt signalling pathway. Phytomedicine 2024, 126, 155382. [Google Scholar] [CrossRef] [PubMed]
  223. Bantug, G.R.; Hess, C. The immunometabolic ecosystem in cancer. Nat. Immunol. 2023, 24, 2008–2020. [Google Scholar] [CrossRef] [PubMed]
  224. Wang, J.; Chen, Q.; Shan, Q.; Liang, T.; Forde, P.; Zheng, L. Clinical development of immuno-oncology therapeutics. Cancer Lett. 2025, 617, 217616. [Google Scholar] [CrossRef] [PubMed]
  225. Martínez-Reyes, I.; Chandel, N.S. Cancer metabolism: Looking forward. Nat. Rev. Cancer 2021, 21, 669–680. [Google Scholar] [CrossRef] [PubMed]
  226. Xia, L.; Oyang, L.; Lin, J.; Tan, S.; Han, Y.; Wu, N.; Yi, P.; Tang, L.; Pan, Q.; Rao, S.; et al. The cancer metabolic reprogramming and immune response. Mol. Cancer 2021, 20, 28. [Google Scholar] [CrossRef] [PubMed]
  227. Zhang, Y.; Wang, Y.; Li, Y.; Huang, C.; Xiao, X.; Zhong, Z.; Tang, J.; Lu, H.; Tang, Y.; Yang, J. Dihydroartemisinin and artesunate inhibit aerobic glycolysis via suppressing c-Myc signaling in non-small cell lung cancer. Biochem. Pharmacol. 2022, 198, 114941. [Google Scholar] [CrossRef]
  228. Chen, R. Dihydroartemisinin Inhibits M2-like Tumor-Associated Macrophage Polarization to Suppress the Invasion and Proliferation of Head and Neck Squamous Cell Carcinoma Cells and Its Underlying Mechanism. Ph.D. Thesis, Hebei Medical University, Shijiazhuang, China, 2020. [Google Scholar]
  229. Xia, L.; Qiu, Y.; Li, J.; Xu, M.; Dong, Z. The Potential Role of Artemisinins Against Neurodegenerative Diseases. Am. J. Chin. Med. 2024, 52, 1641–1660. [Google Scholar] [CrossRef]
  230. Liu, P.; Wang, Y.; Tian, K.; Bai, X.; Wang, Y.; Wang, Y. Artesunate inhibits macrophage-like phenotype switching of vascular smooth muscle cells and attenuates vascular inflammatory injury in atherosclerosis via NLRP3. Biomed. Pharmacother. 2024, 172, 116255. [Google Scholar] [CrossRef]
  231. Liu, T.; Zhang, L.; Joo, D.; Sun, S.-C. NF-κB signaling in inflammation. Signal Transduct. Target. Ther. 2017, 2, 17023. [Google Scholar] [CrossRef]
  232. Firestone, G.L.; Sundar, S.N. Anticancer activities of artemisinin and its bioactive derivatives. Expert Rev. Mol. Med. 2009, 11, e32. [Google Scholar] [CrossRef]
  233. Wang, X.; Xiong, X. Mitochondrial Reactive Oxygen Species (mROS) Generation and Cancer: Emerging Nanoparticle Therapeutic Approaches. Int. J. Nanomed. 2025, 20, 6085–6119. [Google Scholar] [CrossRef]
  234. Morris, G.; Gevezova, M.; Sarafian, V.; Maes, M. Redox regulation of the immune response. Cell. Mol. Immunol. 2022, 19, 1079–1101. [Google Scholar] [CrossRef]
  235. Muri, J.; Kopf, M. Redox regulation of immunometabolism. Nat. Rev. Immunol. 2021, 21, 363–381. [Google Scholar] [CrossRef]
  236. Cluxton, D.; Petrasca, A.; Moran, B.; Fletcher, J.M. Differential Regulation of Human Treg and Th17 Cells by Fatty Acid Synthesis and Glycolysis. Front. Immunol. 2019, 10, 115. [Google Scholar] [CrossRef] [PubMed]
  237. Chen, K.; Tang, L.; Nong, X. Artesunate targets cellular metabolism to regulate the Th17/Treg cell balance. Inflamm. Res. 2023, 72, 1037–1050. [Google Scholar] [CrossRef] [PubMed]
  238. Xue, S.; Su, Z.; Liu, D. Immunometabolism and immune response regulate macrophage function in atherosclerosis. Ageing Res. Rev. 2023, 90, 101993. [Google Scholar] [CrossRef]
  239. Zhang, S.; Gang, X.; Yang, S.; Cui, M.; Sun, L.; Li, Z.; Wang, G. The Alterations in and the Role of the Th17/Treg Balance in Metabolic Diseases. Front. Immunol. 2021, 12, 678355. [Google Scholar] [CrossRef] [PubMed]
  240. Poespoprodjo, J.R.; Douglas, N.M.; Ansong, D.; Kho, S.; Anstey, N.M. Malaria. Lancet 2023, 402, 2328–2345. [Google Scholar] [CrossRef]
  241. Svolacchia, F.; Brongo, S.; Catalano, A.; Ceccarini, A.; Svolacchia, L.; Santarsiere, A.; Scieuzo, C.; Salvia, R.; Finelli, F.; Milella, L.; et al. Natural Products for the Prevention, Treatment and Progression of Breast Cancer. Cancers 2023, 15, 2981. [Google Scholar] [CrossRef]
  242. Krishna, S.; Ganapathi, S.; Ster, I.C.; Saeed, M.E.; Cowan, M.; Finlayson, C.; Kovacsevics, H.; Jansen, H.; Kremsner, P.G.; Efferth, T.; et al. A Randomised, Double Blind, Placebo-Controlled Pilot Study of Oral Artesunate Therapy for Colorectal Cancer. EBioMedicine 2015, 2, 82–90. [Google Scholar] [CrossRef]
  243. Mungo, C.; Sorgi, K.; Misiko, B.; Cheserem, C.; Rahangdale, L.; Githongo, G.; Ogollah, C.; Omoto, J.; Plesa, M.; Zamboni, W. Phase I study on the pharmacokinetics of intravaginal, self-administered artesunate vaginal pessaries among women in Kenya. PLoS ONE 2025, 20, e0316334. [Google Scholar] [CrossRef]
  244. Wen, L.; Chan, B.C.; Qiu, M.H.; Leung, P.C.; Wong, C.K. Artemisinin and Its Derivatives as Potential Anticancer Agents. Molecules 2024, 29, 3886. [Google Scholar] [CrossRef]
  245. Mu, X.; Wang, C. Artemisinins—A Promising New Treatment for Systemic Lupus Erythematosus: A Descriptive Review. Curr. Rheumatol. Rep. 2018, 20, 55. [Google Scholar] [CrossRef]
  246. Tong, X.; Chen, L.; He, S.J.; Zuo, J.P. Artemisinin derivative SM934 in the treatment of autoimmune and inflammatory diseases: Therapeutic effects and molecular mechanisms. Acta Pharmacol. Sin. 2022, 43, 3055–3061. [Google Scholar] [CrossRef] [PubMed]
  247. Nyaaba, N.; Andoh, N.E.; Amoh, G.; Amuzu, D.S.Y.; Ansong, M.; Ordóñez-Mena, J.M.; Hirst, J. Comparative efficacy and safety of the artemisinin derivatives compared to quinine for treating severe malaria in children and adults: A systematic update of literature and network meta-analysis. PLoS ONE 2022, 17, e0269391. [Google Scholar] [CrossRef] [PubMed]
  248. Yin, J.Y.; Wang, H.M.; Wang, Q.J.; Dong, Y.S.; Han, G.; Guan, Y.B.; Zhao, K.Y.; Qu, W.S.; Yuan, Y.; Gao, X.X.; et al. Subchronic toxicological study of two artemisinin derivatives in dogs. PLoS ONE 2014, 9, e94034. [Google Scholar] [CrossRef] [PubMed]
  249. Shakir, L.; Hussain, M.; Javeed, A.; Ashraf, M.; Riaz, A. Artemisinins and immune system. Eur. J. Pharmacol. 2011, 668, 6–14. [Google Scholar] [CrossRef]
  250. Chaturvedi, D.; Goswami, A.; Saikia, P.P.; Barua, N.C.; Rao, P.G. Artemisinin and its derivatives: A novel class of anti-malarial and anti-cancer agents. Chem. Soc. Rev. 2010, 39, 435–454. [Google Scholar] [CrossRef]
  251. Lanna, E.G.; Siqueira, R.P.; Machado, M.G.C.; de Souza, A.; Trindade, I.C.; Branquinho, R.T.; Mosqueira, V.C.F. Lipid-based nanocarriers co-loaded with artemether and triglycerides of docosahexaenoic acid: Effects on human breast cancer cells. Biomed. Pharmacother. 2021, 134, 111114. [Google Scholar] [CrossRef]
  252. Li, Y.; Lu, J.; Chen, Q.; Han, S.; Shao, H.; Chen, P.; Jin, Q.; Yang, M.; Shangguan, F.; Fei, M.; et al. Artemisinin suppresses hepatocellular carcinoma cell growth, migration and invasion by targeting cellular bioenergetics and Hippo-YAP signaling. Arch. Toxicol. 2019, 93, 3367–3383. [Google Scholar] [CrossRef]
  253. Yao, Y.; Guo, Q.; Cao, Y.; Qiu, Y.; Tan, R.; Yu, Z.; Zhou, Y.; Lu, N. Artemisinin derivatives inactivate cancer-associated fibroblasts through suppressing TGF-β signaling in breast cancer. J. Exp. Clin. Cancer Res. 2018, 37, 282. [Google Scholar] [CrossRef] [PubMed]
  254. Shen, K.Y.; Zhu, Y.; Xie, S.Z.; Qin, L.X. Immunosuppressive tumor microenvironment and immunotherapy of hepatocellular carcinoma: Current status and prospectives. J. Hematol. Oncol. 2024, 17, 25. [Google Scholar] [CrossRef] [PubMed]
  255. Mamessier, E.; Sylvain, A.; Thibult, M.L.; Houvenaeghel, G.; Jacquemier, J.; Castellano, R.; Gonçalves, A.; André, P.; Romagné, F.; Thibault, G.; et al. Human breast cancer cells enhance self tolerance by promoting evasion from NK cell antitumor immunity. J. Clin. Investig. 2011, 121, 3609–3622. [Google Scholar] [CrossRef] [PubMed]
  256. Cheng, Y.; Zhang, X.; Wang, Z.; Wang, J. Reconstruction of Immune Microenvironment and Signaling Pathways in Endometrioid Endometrial Adenocarcinoma During Formation of Lymphovascular Space Involvement and Lymph Node Metastasis. Front. Oncol. 2020, 10, 595082. [Google Scholar] [CrossRef]
  257. Steyn, J.D.; Wiesner, L.; du Plessis, L.H.; Grobler, A.F.; Smith, P.J.; Chan, W.C.; Haynes, R.K.; Kotzé, A.F. Absorption of the novel artemisinin derivatives artemisone and artemiside: Potential application of Pheroid™ technology. Int. J. Pharm. 2011, 414, 260–266. [Google Scholar] [CrossRef]
  258. Liu, R.; Yu, X.; Su, C.; Shi, Y.; Zhao, L. Nanoparticle Delivery of Artesunate Enhances the Anti-tumor Efficiency by Activating Mitochondria-Mediated Cell Apoptosis. Nanoscale Res. Lett. 2017, 12, 403. [Google Scholar] [CrossRef]
Figure 1. Artemisinin and its derivatives. Artemisinin, biosynthetic precursor (artemisinic acid), biosynthetic intermediate (dihydroartemisinin), core derivatives (artesunate, artemether, arteether), and experimental derivatives (SM934, DC32, TPN10475, TPN10466, ADART). These compounds are drawn in ChemDraw 20.0.
Figure 1. Artemisinin and its derivatives. Artemisinin, biosynthetic precursor (artemisinic acid), biosynthetic intermediate (dihydroartemisinin), core derivatives (artesunate, artemether, arteether), and experimental derivatives (SM934, DC32, TPN10475, TPN10466, ADART). These compounds are drawn in ChemDraw 20.0.
Ijms 26 08409 g001
Figure 2. The multifaceted antitumor mechanisms of artemisinin and its derivatives. The mechanisms primarily involve three interconnected biological processes: cell cycle arrest, oxidative stress induction, and the promotion of various forms of cell death. “↓” in black and “↑” in blue denote “enhancing or increasing” while “⊥” and “↓” in red indicate “suppressing or reducing”. This figure is created in BioRender. https://BioRender.com/tea068o (accessed on 21 July 2025).
Figure 2. The multifaceted antitumor mechanisms of artemisinin and its derivatives. The mechanisms primarily involve three interconnected biological processes: cell cycle arrest, oxidative stress induction, and the promotion of various forms of cell death. “↓” in black and “↑” in blue denote “enhancing or increasing” while “⊥” and “↓” in red indicate “suppressing or reducing”. This figure is created in BioRender. https://BioRender.com/tea068o (accessed on 21 July 2025).
Ijms 26 08409 g002
Figure 3. The diverse immune regulation of artemisinin and its derivatives. The immunoregulatory actions of artemisinin and its derivatives are explored through the lens of distinct immune cell types, specifically T cells, B cells, macrophages, and natural killer (NK) cells. “↓” in black and “↑” in blue denote “enhancing or increasing” while “⊥” and “↓” in red indicate “suppressing or reducing”. This figure is created in BioRender. https://BioRender.com/9j6k8fd (accessed on 21 July 2025).
Figure 3. The diverse immune regulation of artemisinin and its derivatives. The immunoregulatory actions of artemisinin and its derivatives are explored through the lens of distinct immune cell types, specifically T cells, B cells, macrophages, and natural killer (NK) cells. “↓” in black and “↑” in blue denote “enhancing or increasing” while “⊥” and “↓” in red indicate “suppressing or reducing”. This figure is created in BioRender. https://BioRender.com/9j6k8fd (accessed on 21 July 2025).
Ijms 26 08409 g003
Figure 4. The diverse immune regulation of artemisinin and its derivatives. (a) The mechanisms of artemisinin and its derivatives on glucose metabolism; (b) The mechanisms of artemisinin and its derivatives on lipid metabolism. “↓” in black and “↑” in blue denote “enhancing or increasing” while “⊥” and “↓” in red indicate “suppressing or reducing”. This figure is created in BioRender. https://BioRender.com/m0zxmmb (accessed on 21 July 2025).
Figure 4. The diverse immune regulation of artemisinin and its derivatives. (a) The mechanisms of artemisinin and its derivatives on glucose metabolism; (b) The mechanisms of artemisinin and its derivatives on lipid metabolism. “↓” in black and “↑” in blue denote “enhancing or increasing” while “⊥” and “↓” in red indicate “suppressing or reducing”. This figure is created in BioRender. https://BioRender.com/m0zxmmb (accessed on 21 July 2025).
Ijms 26 08409 g004
Table 1. Efficacy of artemisinin and its derivatives on other tumors. “↓” denotes “suppressing or reducing” while “↑” indicates “enhancing or increasing”.
Table 1. Efficacy of artemisinin and its derivatives on other tumors. “↓” denotes “suppressing or reducing” while “↑” indicates “enhancing or increasing”.
DiseaseCompoundEffectMechanismModelReference
LeukemiaDihydroartemisinininducing cell deathMcl-1 ↓, Bcl-2 ↓, Bax ↑, P21 ↑in vitro[36]
Ovarian cancer Artesunatereducing rRNA transcription, inhibiting cell proliferation and migration, and inducing apoptosisFANCA-targeted, mTOR ↓, RPS 6 ↓in vitro and vivo[90]
GlioblastomaDihydroartemisininblocking the cell cycle, inducing apoptosisERRα ↓, Caspase—3 ↑, Cleaved Caspase—9 ↑, PARP ↑in vitro and vivo[91]
Artesunateinducing oxidative stress, DNA damage, apoptosis, and ferroptosisATM/ATR axis ↑in vitro and vivo[92]
Prostate cancerDihydroartemisinininducing ROS-dependent mitochondrial apoptosis, autophagyPI3K/AKT/mTOR pathway ↓in vitro[93]
MelanomaArtemisininsuppressing postoperative recurrence and metastasisKIT/PI3K/AKT pathway ↓, Cyclin D1 ↓, Bcl-2 ↓in vitro and vivo[94]
Dihydroartemisinininducing ROS-dependent apoptosis, inhibiting cell migrationBcl-2 ↓, Survivin ↓, Bax ↑, MMP—9 ↓in vitro and vivo[95]
Artesunateinducing ferroptosis, activating CD8+ T-cell immunityIDO1 ↓in vitro and vivo[96]
LymphomaArtesunateinducing ROS-dependent apoptosis, ferroptosis, DNA damage, inhibiting angiogenesis, and activating antitumor immunityTopo I ↓, Topo II ↓in vitro and vivo[97]
Gastric cancerDihydroartemisinininhibiting vasculogenic mimicryFGF2/FGFR1-MAPK/PI3K pathway ↓, VE-cadherin ↓, MMP-2 ↓, MMP-9 ↓in vitro and vivo[98]
NeuroblastomaDihydroartemisininblocking the cell cycle, inducing mitochondrial apoptosisPARP-1 ↑, caspase 3 ↑, γH2AX ↑in vitro[99]
Artesunateblocking the cell cycle, inducing mitochondrial apoptosisactivated caspase-3 ↑, sub-G1 fraction ↑in vitro[100]
Artemetherenhancing the caspase-dependent apoptotic effect of DoxorubicinB7-H3 ↓, NF-κB pathway ↓in vitro[101]
Head and neck squamous cell carcinoma (HNSCC)Artemisinininducing apoptosis, promoting vascular normalization, and enhancing antitumor immunityMIF ↓, VEGF ↓, IL-8 ↓in vitro and vivo[102]
Dihydroartemisininblocking EMT and invasive migrationmiR-195-5p ↑, ZEB1/MMP-9 ↓in vitro[103]
CholangiocarcinomaDihydroartemisinininducing apoptosis and autophagyDAPK1-BECLIN1 pathway ↑in vitro[104]
OsteosarcomaDihydroartemisininenhancing anti-angiogenesisLoxl2 ↓, VEGFA ↓in vitro and vivo[105]
blocking the cell cycleWnt/β-catenin pathway ↓, Cyclin D1 ↓, c-Myc ↓in vitro and vivo[106]
RhabdomyosarcomaDihydroartemisinininducing autophagy, blocking the cell cycleAMPK pathway ↑, mTORC1 ↓in vitro and vivo[107]
Artesunateinducing mitochondrial apoptosis, inhibiting angiogenesisROS ↑, p38 MAPK ↑, VEGF ↓in vitro and vivo[38]
Pancreatic cancerArtesunateInhibiting cell growth, inducing apoptosisTop2A ↓, RRM2 ↓, PCNA ↓, NAG—1 ↑in vitro[108]
inducing ferroptosisGRP78-xCT axis ↓in vitro and vivo[109]
Urothelial carcinomaArtemisinininhibiting proliferation, enhancing the DNA-damaging effects of cisplatinFGFR3 ↓, HRAS ↓, P53 ↑, KDM6A ↑in vitro and vivo[110]
Cervical cancerDihydroartemisinininducing ferroptosisGPX4 ↓, SLC7A11 ↓in vitro[111]
inducing apoptosisRKIP-NF-κB axis ↑, Bcl-2 ↓in vitro[112]
inducing autophagyBcl-2 ↓in vitro and vivo[113]
blocking the cell cycleROS ↑, caspase-9 ↑, PARP ↑in vitro and vivo[114]
Artesunatereversing immunosuppressionCOX-2 ↓, PGE2 ↓, Foxp3 ↓in vitro and vivo[115]
enhancing the TRAIL-induced DR4/DR5-Caspase ApoptosisNF-κB ↓, PI3K-Akt ↓in vitro[116]
Esophageal carcinomaDihydroartemisinininhibiting proliferation, promoting apoptosisDAB2IP ↑, RAS/ERK pathway ↓in vitro and vivo[117]
Artesunateinducing ferroptosisAKT/mTOR-xCT/GPX4 axis ↓in vitro and vivo[118]
Thyroid cancer Artemisinininhibiting proliferation, promoting apoptosisXIST/miR-93/HIF-1α axis1 ↓in vitro and vivo[119]
Renal carcinomaArtesunateinducing ROS-dependent necrotic apoptosisRIP1-MLKL pathway ↑in vitro[120]
Table 2. Efficacy of artemisinin and its derivatives on immune disorders. “↓” denotes “suppressing or reducing” while “↑” indicates “enhancing or increasing”.
Table 2. Efficacy of artemisinin and its derivatives on immune disorders. “↓” denotes “suppressing or reducing” while “↑” indicates “enhancing or increasing”.
DiseaseCompoundEffectMechanismModelReference
Autoimmune hepatitisTPN10475reducing serum levels of alanine aminotransferase and aspartate aminotransferase, and inhibiting infiltrating inflammatory T cellsPI3K—AKT pathway ↓, INF-γ ↓, TNF-α ↓In vivo and in vitro[166]
TPN10466reducing T-cell responsesERK/JNK/p38 pathway ↓, TNF-α ↓, IL-6 ↓In vivo[167]
Autoimmune EncephalomyelitisTPN10475increasing Treg, decreasing T cells, and inflammatory cell infiltrationTGF-βpathway ↑, Th1 ↓, Th17 ↓In vivo and in vitro[128]
Dihydroartemisininincreasing Treg and inhibiting Th cell differentiationIFN-γ ↓, IL-4 ↓, mTOR pathway ↓, TGF-βR:Smad pathway ↑In vivo and in vitro[168]
MelanomaDihydroartemisininincreasing CTL, inhibiting the immunosuppressive effect of Treg, and the invasion and migration of tumor cellsSTAT3 pathway ↓, Treg ↓, IL-10 ↓In vivo and in vitro[169]
Sjögren’s syndromeArtesunatereducing lymphocyte infiltration and immune inflammationIRF4 ↓, Th17 ↓In vivo and in vitro[170]
AtherosclerosisArtemisinininhibiting foam macrophage transformation and enhancing macrophage autophagyAMPK pathway ↑, mTOR ↓, ULK1 ↓, LC—3II ↑, P62 ↓In vivo[171]
Artesunatereducing the size of atheroplaque and lipid depositionKLF2/NRF2/TCF7L2 pathway ↑, LPL ↑In vivo[172]
Acute lung injuryDihydroartemisinininhibiting LPS-induced inflammatory cell infiltration, oxidative stress, and pro-inflammatory cytokine productionIL-1β ↓, TNF-α ↓, IL-6 ↓, NF-κB pathway ↓, Nrf2 pathway ↑In vivo and in vitro[173]
Ulcerative colitis (UC)Dihydroartemisininreducing inflammatory cell infiltrationJAK2/STAT3 pathway ↓, IL-6 ↓, IL-1β ↓, TNF-α ↓In vivo[174]
Artemetherinhibiting ROS production and preventing the assembly and activation of NLRP3 inflammasomesIL-1β ↓, IL-6 ↓, TNF-α ↓, mtROS ↓In vivo and in vitro[175]
Dry eye diseaseSM934reducing inflammatory cell infiltration and inflammatory factor expressionIL-1β ↓, IL-6 ↓, TNF-α ↓, TLR4/NF-κB pathway ↓, NLRP3 ↓In vivo[140]
Table 3. Efficacy of artemisinin and its derivatives on metabolic disorders. “↓” denotes “suppressing or reducing” while “↑” indicates “enhancing or increasing”.
Table 3. Efficacy of artemisinin and its derivatives on metabolic disorders. “↓” denotes “suppressing or reducing” while “↑” indicates “enhancing or increasing”.
DiseasesCompoundEffectMechanismProgressReferences
Type 2 diabetesArtemisininreducing blood glucose, oxidative stress, and inflammation levels and reversing IR; increasing regeneration of pancreatic β-cellPax4 ↑, Arx ↑in vitro and in vivo[181]
Artemetherpromoting glycogen synthesis with antidiabetic effectsAMPK ↑, PI3K/Akt ↑in vivo[183]
Diabetic cardiomyopathyArtemisininreducing oxidative stress, inflammation, and fibrosisAGE-RAGE/HMGB-1 ↓in vivo[193]
LeukemiaDihydroartemisinininhibiting leukemia cell proliferation by regulating glycolysis and metabolismPKM2 ↓, GLUT1 ↓in vitro[184]
Non-small-cell lung cancerDihydroartemisinininhibiting glucose metabolism via NF-κB pathway inhibitionNF-κB↓, GLUT1↓in vitro and in vivo[191]
ObesityArtesunatereducing food intake and promoting energy expenditure, leading to weight lossGDF15/GFRAL ↑in vivo[197]
Artesunateinhibiting lipid accumulation and TG synthesisC/EBP-a ↓, PPAR-γ ↓, FAS ↓, periilipin A ↓, STAT-3 ↓in vitro[195]
AtherosclerosisArtemisininreducing ROS and NO; suppressing lipid influx; reducing the uptake and internalization of oxLDLNF-κB/NLRP3 ↓, AMPK/mTOR ↑in vitro and in vivo[202]
Artemisininsuppressing inflammatory reactionNF-κB/NLRP3 ↓, AMPK ↑in vivo[204]
Artesunatereducing pro-inflammatory cytokine and downregulating the pro-inflammatory chemokines; attenuating the progression of atherosclerosis lesionsTNF-a ↓, IL-6 ↓, IL-8 ↓, MCP-1 ↓in vivo[194]
Fatty liver diseasesArtemetherinhibiting microglial activation in the hypothalamus; protecting the TRH neurons from inflammatory damage and promoting the release of TRHpalmitoylation of PKCδ ↓in vivo[205]
Obesity-related glomerulopathyArtemisininreducing the degradation of vitamin D;
mitigating renal tubular damage and fibrosis
CYP24A1 ↓, vitamin D ↑in vivo[206]
Nonalcoholic steatohepatitis (NASH)Dihydroartemisinindecreasing the synthesis of lipids; inhibiting OS and ERS; reducing liver steatosisNF-κB ↓in vivo[207]
Thyroid eye disease (TED)Dihydroartemisinin, Artesunateinhibiting adipogenesis; inhibiting cell proliferation and hyaluronan productionHAS2 ↓, IGF1R ↓, adipogenic markers ↓in vitro[208]
Amiodarone-induced pulmonary toxicityArtemisininregulating cellular energy homeostasis and mitigating oxidative stress; protecting against amiodarone-induced apoptosis and bronchial epithelial cell sheddingp-AMPK ↑, CaMKK2 ↑, Nrf2 ↑, SOD1 ↑in vitro and in vivo[209]
Amiodarone-induced ocular toxicityArtemisininreducing the cytotoxicity induced by amiodaroneLDH ↓, ROS ↓, MMP ↑, p-AMPK ↑, CaMKK2 ↑, Nrf2 ↑in vitro[210]
APAP-induced acute liver injury (AILI)Artemethermitigating liver histological changes, including mitochondrial damage, hepatocyte necrosis, hepatocyte apoptosis, and inflammatory infiltrationNrf2-HO-1/GPX4 ↑, ROS ↓in vivo[211]
Abnormal white matter development (periventricular leukomalacia)Artemisininreducing perinatal inflammation and inflammatory cytokine productionIRAK-4 ↓, IRAK-1 ↓, IL-1β ↓, IL-6 ↓, TNF-α ↓, ROS ↓, Nrf2 ↑in vitro[212]
Periprosthetic osteolysis (PPO)Artemisic acidreducing osteoclast formation and alleviating titanium particle-induced calvarial osteolysisROS ↓, Nrf2 ↑, MAPK ↓, NF-κB ↓, NFATc1 ↓, c-Fos ↓in vitro and in vivo[186]
Polycystic ovarian syndrome (PCOS)Artemisinininhibiting ovarian androgen synthesis; mediating LONP1-CYP11A1 interaction; regulating mitochondrial functionCYP11A1 ↓in vitro and in vivo[213]
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Xia, Y.; Shi, C.; Lu, J.; Zhu, Z.; Li, M.; Pan, Y.; Huang, X.; Zhang, L.; Liu, A. Artemisinin and Its Derivatives from Molecular Mechanisms to Clinical Applications: New Horizons Beyond Antimalarials. Int. J. Mol. Sci. 2025, 26, 8409. https://doi.org/10.3390/ijms26178409

AMA Style

Xia Y, Shi C, Lu J, Zhu Z, Li M, Pan Y, Huang X, Zhang L, Liu A. Artemisinin and Its Derivatives from Molecular Mechanisms to Clinical Applications: New Horizons Beyond Antimalarials. International Journal of Molecular Sciences. 2025; 26(17):8409. https://doi.org/10.3390/ijms26178409

Chicago/Turabian Style

Xia, Yi, Chuanjing Shi, Jingze Lu, Zeyu Zhu, Mohan Li, Yinan Pan, Xinyan Huang, Lei Zhang, and Aifen Liu. 2025. "Artemisinin and Its Derivatives from Molecular Mechanisms to Clinical Applications: New Horizons Beyond Antimalarials" International Journal of Molecular Sciences 26, no. 17: 8409. https://doi.org/10.3390/ijms26178409

APA Style

Xia, Y., Shi, C., Lu, J., Zhu, Z., Li, M., Pan, Y., Huang, X., Zhang, L., & Liu, A. (2025). Artemisinin and Its Derivatives from Molecular Mechanisms to Clinical Applications: New Horizons Beyond Antimalarials. International Journal of Molecular Sciences, 26(17), 8409. https://doi.org/10.3390/ijms26178409

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop