Next Article in Journal
Multi-Color Tunable Afterglow Materials Leveraging Energy Transfer Between Host and Guest
Previous Article in Journal
Cloud Point Behavior of Poly(trifluoroethyl methacrylate) in Supercritical CO2–Toluene Mixtures
Previous Article in Special Issue
A Comprehensive Review of the Phytochemistry, Pharmacology and Other Applications of Euphorbiae Humifusae Herba
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Natural Products as Novel Therapeutic Agents for Triple-Negative Breast Cancer: Current Evidence, Mechanisms, Challenges, and Opportunities

1
State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
2
School of Basic Medical Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
*
Authors to whom correspondence should be addressed.
These authors contributed equally to this work.
Molecules 2025, 30(6), 1201; https://doi.org/10.3390/molecules30061201
Submission received: 26 January 2025 / Revised: 23 February 2025 / Accepted: 2 March 2025 / Published: 7 March 2025
(This article belongs to the Special Issue Chemical Constituents and Biological Activities of Natural Sources)

Abstract

:
Breast cancer (BC) tops the list of causes for female fatalities globally, with the elusive triple-negative breast cancer (TNBC) constituting 10–20% of all cases. Current clinical strategies for combating TNBC encompass a multifaceted approach, including surgical intervention, radiation therapy, chemotherapy, and advanced targeted drugs and immunotherapies. While these modalities have catalyzed significant advancements in TNBC management, lingering limitations continue to pose formidable challenges. There is an acute need for novel therapeutics in the realm of TNBC treatment. Natural products (NPs) have emerged as a rich reservoir for pharmaceutical innovation, owing to their extraordinary range of structures and physicochemical properties. Scholars have reported diverse evidence of NPs’ efficacy against TNBC. This review aims to comprehensively explore the bioactive constituents, specifics and commonalities of chemical structure, and pharmacological mechanisms of NPs, specifically examining their multifaceted roles in impeding TNBC. NPs, which have recently garnered significant interest, are intriguing in terms of their capacity to combat TNBC through multifaceted mechanisms, including the suppression of tumor cell proliferation, the induction of apoptosis, and the inhibition of tumor metastasis. These natural agents primarily encompass a range of compounds, including terpenoids, glycosides, phenolic compounds, and alkaloids. An in-depth exploration has unveiled their involvement in key signaling pathways, including the transforming growth factor-beta (TGF-β), vascular endothelial growth factor A (VEGFA), phosphoinositide 3-kinase/protein kinase B (PI3K/AKT), Wingless/Int-1 (Wnt) /β-catenin, and mitogen-activated protein kinase (MAPK) pathways. Meanwhile, this review also looks at the challenges and opportunities that arise from harnessing natural compounds to influence TNBC, while outlining the prospective trajectory for future research in the field of NPs.

1. Introduction

Breast cancer (BC), which accounted for 670,000 deaths globally in 2022, has been defined as a highly life-threatening disease among the female population by the World Health Organization (WHO) [1]. The complexity of BC is strongly related to its heterogeneity, which results in diverse clinical and therapeutic outcomes. Among the various subtypes, TNBC is particularly aggressive. Unlike luminal A/B BC, human epidermal growth receptor 2 (HER2)-positive BC, and basal-like BC, TNBC lacks expression of estrogen receptors (ERs), progesterone receptors (PRs), and HER2, making it unresponsive to hormone therapy, endocrine therapy, and HER2-targeted therapy [2]. TNBC treatment currently relies primarily on chemotherapy as a therapeutic modality [3]. For instance, paclitaxel- and anthracycline-based chemotherapy drugs are currently the principal choices for treating TNBC, with platinum-based compounds demonstrating promising effects in certain treatment contexts [4,5,6,7]. However, the induction of endogenous or acquired resistance in early-stage treatment, significant adverse effects, and potential non-specific off-target effects severely limit the utility of these chemotherapy drugs [8]. Consequently, novel therapeutic strategies, such as targeted therapy and immunotherapy, are being continuously developed to improve their treatment efficacy and survival rates [9,10]. For example, poly adenosinediphosphate-ribose polymerase (PARP) inhibitors have shown therapeutic potential in patients with breast cancer gene (BRCA) mutations. Moreover, other synthetic small-molecule drugs, such as tyrosine kinase inhibitors, including Lapatinib and Afatinib, have also demonstrated commendable efficacy by targeting multiple tyrosine kinase receptors, thereby disrupting the signal transduction pathways of cancer cells [11]. In addition, immune checkpoint inhibitors, such as pembrolizumab and atezolizumab, have displayed breakthrough activity in certain TNBC patients.
Traditional Chinese medicines (TCMs) are regarded as valuable complementary and alternative therapies with significant anticancer potential [12,13]. Herbal medicines (HMs), which have served as the primary remedies in many ancient civilizations, have garnered significant interest in cancer drug discovery. Notably, numerous NPs derived from HMs have demonstrated antitumor effects, a discovery that gained considerable attention following the identification of paclitaxel’s remarkable anticancer activity. As a result, HMs-associated NPs have become a focal point in drug discovery, with research in this area experiencing substantial growth. Recently, it was found that approximately 30% of drugs approved by the U.S. Food and Drug Administration (FDA) are HMs extracts and HM-associated NPs [14]. Paclitaxel, for instance, originally extracted from the herbal plant Taxus chinensis, remains a first-line treatment for various carcinomas. However, many NPs with potential therapeutic benefits are yet to be fully explored. HMs are recognized as a rich source of medicinal compounds, providing advantages such as cost-effectiveness and a wide array of structural and functional variations. The therapeutic efficacy of HMs and associated NPs in treating TNBC has been demonstrated through their ability to target multiple signaling pathways and exhibit various bioactivities.
In this review, we present a comprehensive analysis of the chemical characteristics, bioactivities, and botanical origins of NPs identified as potential adjuvant treatment for TNBC. The primary compounds within these NPs include terpenoids, glycosides, phenolics, and alkaloids. These NPs have been shown to inhibit TNBC through multiple mechanisms, such as the suppression of angiogenesis, prevention of metastasis, induction of apoptosis, interruption of the cell cycle, and regulation of autophagy. The key pathways involved include TGF-β, VEGFA, PI3K/AKT, Wnt/β-catenin, and MAPK. Consequently, NPs offer significant potential for the development of innovative and effective therapies for TNBC.

2. Activity of Natural Products in Treatment of Triple-Negative Breast Cancer

2.1. Terpenoids

Terpenoids, which represent the largest class of compounds produced by plants, are derived from mevalonic acid, and are characterized by a molecular framework of isoprene units [15]. The compounds are connected in various ways to form polymers: monoterpenes consist of two isoprene units, sesquiterpenes consist of three, diterpenes consist of four, etc. Terpenoids hold significant importance in the fields of biology, pharmacology, and chemistry. Among their numerous biological properties, their antitumor effects are particularly noteworthy, encompassing diverse activities such as anti-proliferative, apoptotic, anti-angiogenic, and anti-metastatic actions [1,16].
Terpenoids undergo enzymatic elongation and cyclization reactions, resulting in compounds with high hydrophobicity and structural complexity. Compared to non-terpenoid NPs, terpenoids exhibit more intricate ring structures and chiral stereochemistry. In proteins, the number of hydrophobic amino acids correlates with protein folding; thus, hydrophobic residues are prevalent in protein active pockets and interfaces that are crucial for ligand binding. The hydrophobic effect is considered a significant driving force for molecular recognition in protein–ligand interactions. Consequently, hydrophobic terpenoids may demonstrate favorable properties in protein–ligand binding. Some terpenoid compounds have been found to possess inhibitory activity against protein–protein interactions, targeting various drug sites, highlighting their broad applicability in drug discovery endeavors [17,18]. Terpenoids are widely distributed in nature and exhibit diverse biological activities, including anti-TNBC effects (Figure 1, Table 1 and Table S1).

2.1.1. Anti-Metastasis

Terpenoids have been reported to induce apoptosis in TNBC cells, which is a form of programmed cell death crucial for the development, homeostasis, and defense mechanisms of multicellular organisms. Appropriate apoptosis function can prevent tumorigenesis [19]. Terpenoids such as atractylenolide-1 (AT-1), demethylzeylasteral (T-96), isotoosendanin (ITSN), toosendanin (TSN), Eupalinolide O (EO), and Jatamanvaltrate P have been documented to modulate the expression of apoptosis-related proteins, including B-cell lymphoma 2 (Bcl2), Bcl-2-associated X protein (Bax), and Caspase-3, thereby inducing death of TNBC cells [20,21,22,23,24]. Apoptosis is regulated by multiple signaling pathways. T-96 significantly reduces lysine-specific demethylase 1 (LSD1) protein levels, increases its target protein phosphatase and tensin homolog (PTEN), and enhances histone methylation. This action ultimately downregulates the PI3K/AKT signaling pathway, effectively inhibiting the growth of TNBC cells and promoting apoptosis [25]. Maackiain (MA) regulates mitochondrial apoptosis signaling in TNBC by promoting GADD45A through the inhibition of microRNA(miR)-374a [26]. EO may induce apoptosis in TNBC cells by modulating reactive oxygen species (ROS) production and the AKT/p38 MAPK pathway [23]. Terpene compounds such as Cycloart-23E-ene-3β, 25-diol, germacrone, and β-Bisabolene have also been reported to induce apoptosis in TNBC cells [27,28,29].

2.1.2. Regulation of Cell Cycle

Disruptions in the cell cycle, which is essential for regulated cell proliferation, are pivotal in tumor development and progression, and research into targeting these disruptions offers promising strategies for cancer treatment. Terpenoids, such as AT-1, Jatamanvaltrate P, Cucurbitacin B, Lineariifolianoid E, and germacrone, have been shown to inhibit the cell cycle, effectively suppressing the growth and proliferation of TNBC cells [21,24,27,29,30,31,32]. They regulate the expression of cell cycle proteins, such as cyclin-dependent kinase (CDK1) and Cyclin D1 (CCND1), leading to cell cycle arrest and inhibition of TNBC cell growth [28,32]. The progression of the cell cycle also depends on the regulation of intracellular signaling pathways. Cucurbitacin B may inhibit the proliferation of human BC cells by disrupting the microtubule network, downregulating Cellular myelocytomatosis oncogene (c-Myc) and nucleophosmin/B23, and interfering with the trafficking of nucleophosmin/B23 from the nucleolus to the nucleoplasm, leading to G2/M arrest [33,34].

2.1.3. Modulation of Autophagy

Autophagy is a self-degradative process that is crucial for regulating metabolism, maintaining energy balance, and controlling cell development and death in response to stress or starvation [35]. In cancer cells, autophagy has a dual role, both promoting tumor cell survival and growth, and potentially inhibiting tumor initiation and progression [36]. As research advances, a better understanding of the complex mechanisms of autophagy may offer new directions for cancer treatment. Terpenoid compounds such as Anomanolide C (AC), ITSN, TSN, and Jatamanvaltrate P have been reported to modulate autophagy. Treatment with these compounds increases the LC3B-II/LC3B-I ratio in TNBC cells, leading to autophagy activation and subsequent inhibition of TNBC [20,24,37,38,39].
Terpenoid compounds show significant potential in inhibiting TNBC by targeting multiple mechanisms, including metastasis suppression and apoptosis induction, demonstrating their multifaceted abilities to hinder the progression of TNBC. For instance, AC has been demonstrated to concurrently impede cell metastasis and modulate autophagy. AT-1 exhibits the potential to inhibit metastasis, arrest the cell cycle, and provoke apoptosis. Similarly, Jatamanvaltrate P has been noted for its ability to halt the cell cycle, influence autophagy, and induce apoptosis. Collectively, these active terpenoid compounds present substantial potential as therapeutic agents for TNBC. Nonetheless, a more profound understanding of their underlying mechanisms is essential, necessitating further research. Additionally, extensive preclinical studies will be pivotal in advancing their translation into clinical practice.
Table 1. Profiles of terpenoids in NPs with therapeutic effects on TNBC.
Table 1. Profiles of terpenoids in NPs with therapeutic effects on TNBC.
Natural ProductsStructureSourcesConcentration In Vitro Dosage In VivoBiological Activities/MechanismsPotential TargetsReference
Anomanolide CMolecules 30 01201 i001Tubocapsicum anomalum (S. Vidal) M. J. Pires0.25, 1, 2 μM25, 50 mg/kgInduces glutathione peroxidase 4 (GPX4) ubiquitination to trigger autophagy-dependent ferroptosis in TNBC.GPX4[38]
Atractylenolide-1Molecules 30 01201 i002Atractylodes lancea25, 50 μM20, 50 mg/kgInhibits phosphorylation of the Extracellular signal-regulated kinase (ERK) family; suppresses the expression of Triosephosphate Isomerase 1 (TPI1) and Glucose-6-phosphate isomerase GPI (GPI), thereby affecting the glycolytic/gluconeogenic pathway.TPI1 and GPI[21]
BritanninMolecules 30 01201 i003Inula japonica Thunb5, 10, 20 μM5, 10 mg/kgInduces Zinc finger e-box binding homeobox 1 (ZEB1) degradation, inhibiting invasion and metastasis of TNBC cells.ZEB1[40]
DemethylzeylasteralMolecules 30 01201 i004Tripterygium wilfordii5, 10 μM4 mg/kgReduces the expression of LSD1 protein, enhances the expression of its target protein PTEN, and suppresses the PI3K/AKT signaling pathway.LSD1[25]
IsotoosendaninMolecules 30 01201 i005Melia azedarach L. 0.1, 0.3, 1 μM0.1, 1 mg/kgDirectly engages with the kinase domain of TGF-β receptor type-1 (TGFβR1), thereby abolishing TGF-β-induced epithelial–mesenchymal transition (EMT) in the TNBC microenvironment.TGFβR1[20,37]
ToosendaninMolecules 30 01201 i006Melia azedarach L.0.1, 1, 5 μM/Inhibits autophagy by alkalizing lysosomal pH./[20,39]
Jatamanvaltrate PMolecules 30 01201 i007Valeriana jatamansi A. DC.2.5, 5 μM15 mg/kgEnhances PARP and caspase cleavage, indicating apoptosis induction, and reducing Cyclin B1, CCND1, and Cell division cycle 2 (Cdc-2) expression, which are crucial for cell cycle progression./[24]
Maackiain Molecules 30 01201 i008Spatholobus suberectus H. S. Lo1, 2, 2.5, 5 μM25, 50 mg/kgEnhances the induction of GADD45α protein and mRNA through the inhibition of miR-374a./[26]
TriptonideMolecules 30 01201 i009Tripterygium wilfordii Hook. f.0.2 μM5 mg/kgSuppresses the tumorigenesis and metastasis of TNBC via triggering the degradation of Twist1 and Notch1 oncoproteins./[41,42]
Eupalinolide OMolecules 30 01201 i010Eupatorium lindleyanum DC.5, 10 μM15, 30 mg/kgInduces apoptosis, which is orchestrated through ROS generation and the AKT/p38 MAPK signaling pathway./[23]

2.2. Glycosides

Glycosides are characterized by the presence of a sugar moiety linked to a non-sugar portion known as the aglycone. These compounds display distinct biological activities due to their varied aglycone structures, while their shared sugar moieties contribute to similar functionalities.
NPs contain numerous glycoside components, and research has documented their inhibitory effects on TNBC (Figure 2, Table 2 and Table S1).

2.2.1. Anti-Metastasis

Recent studies have highlighted that glycoside compounds, like icariin, Vanicoside B, asiaticoside (AS), Astragaloside IV (AS-IV), scutellarin (SC), Platycodin D (PD), and rutin, have the potential to inhibit the metastasis of TNBC cells through various mechanisms [43,44,45,46,47,48,49,50,51,52,53]. Icariin, for instance, has been reported to reduce TNBC cell invasion by targeting and suppressing the c-Jun N-terminal kinase (JNK)/c-Jun signaling pathway [47]. Vanicoside B has demonstrated significant anti-metastatic properties [43]. One proposed mechanism is its ability to counteract Cyclin-Dependent Kinase 8 (CDK8)-mediated signaling pathways, which are involved in tumor progression [43]. AS inhibits the P2RX7-mediated TGF-β/SMAD signaling pathway by enhancing PPARG expression, which in turn reduces EMT in TNBC [44]. AS-IV has been shown to inhibit both the proliferation and invasion of TNBC cells and to suppress tumor growth in animal models. This is achieved through mechanisms involving the downregulation of Vav Family Member 3 (Vav3) and the inactivation of the Rac1/MAPK signaling pathway [54]. SC reduces TNBC metastasis by targeting the tumor necrosis factor alpha (TNFα)/Tumor necrosis factor receptor 2 (TNFR2) pathway, which leads to the activation of runt-related transcription factor 1 (RUNX1) and increased production of granulocyte-colony stimulating factor (G-CSF) in TNBC-associated endothelial cells [52,53]. PD inhibits the migration, invasion, and adhesion of highly metastatic TNBC cells by downregulating matrix metalloproteinase-9 (MMP-9) and inhibiting epidermal growth factor receptor (EGFR), as well as the MAPK and PI3K/AKT pathways [49]. Ophiopogonin D has the potential to suppress TGF-β1-mediated metastatic behavior in TNBC cells by modulating the ITGB1/FAK/Src/AKT/β-catenin/MMP-9 signaling axis [55]. Rutin inhibits the migration and invasion of TNBC cells by reducing c-Met phosphorylation and blocking its downstream signaling pathways [51,56].

2.2.2. Pro-Apoptosis

Studies have shown that glycoside compounds, such as icariin, Vanicoside B, Ginsenoside Rg3, Saikosaponin D, DLBS1425, Saxifragifolin A, and neohesperidin, can also induce apoptosis in TNBC cells through various mechanisms [43,47,57,58,59,60,61,62,63]. Icariin has been reported to induce apoptosis by causing an excessive accumulation of ROS in cells [47]. Vanicoside B induces apoptosis in TNBC cells through activation of the Skp2-p27 signaling pathway, achieved by inhibiting CDK8 activity [43]. Rg3 induces apoptosis in TNBC cells through the classical mitochondria-dependent caspase activation pathway. Additionally, Rg3 has been reported to induce apoptosis in MDA-MB-231 cells by blocking Nuclear Factor kappa B (NF-κB) signaling, inactivating ERK and AKT, and destabilizing mutant p53 [64,65]. SSD exhibits a strong anti-proliferative effect on TNBC cells by inducing apoptosis. Its potential molecular mechanism involves inhibiting the Wnt/β-catenin signaling pathway, rather than targeting EGFR and Neurotensin receptor 1 (NTSR1) [59]. Glycoside compounds have demonstrated potential as therapeutic agents for TNBC by inducing apoptosis through various mechanisms.

2.2.3. Other Anti-TNBC Effects

In addition to inhibiting metastasis and inducing apoptosis in TNBC cells, glycoside compounds also exhibit other anti-TNBC activities, such as synergistic effects with other drugs, enhancing drug efficacy, and modulating the tumor microenvironment. Ginsenoside Rg1 enhances the cytotoxic effects of doxorubicin through a synergistic mechanism [66]. Additionally, Ginsenoside Rg1 has been shown to protect effectively against cisplatin-induced liver damage, reducing associated histological damage. This protective effect is mediated by inhibiting the Kelch-like epichlorohydrin-associated protein 1 (Keap1)-Nrf2 interaction, partly due to p62 accumulation, and primarily through increasing the production of Nrf2-related antioxidant proteins [67]. The combination of oxymatrine and AS-IV can enhance the immune system to combat TNBC by improving the tumor microenvironment and boosting the antitumor activity of T cells [45]. Anemoside A3 prevents the polarization of macrophages into the M2 type by decreasing the phosphorylation of Signal transducer and activator of transcription 3 (STAT3) protein, thereby suppressing M2 macrophage-driven tumor metastasis [68]. Glycoside compounds have shown potential in inhibiting TNBC. We can further explore additional candidate compounds with anti-TNBC activity.
Glycoside compounds inhibit the migration, invasion, and growth of TNBC cells by modulating various signaling pathways, such as MAPK, PI3K/AKT, CDK8, and ITGB1/FAK/Src/AKT. These compounds induce apoptosis in TNBC cells through multiple mechanisms: promoting excessive ROS accumulation, inhibiting CDK8 to activate the Skp2-p27 signaling pathway, triggering the mitochondria-dependent caspase activation pathway, blocking NF-κB signaling, inhibiting ERK and AKT, destabilizing mutant p53, and suppressing the Wnt/β-catenin signaling pathway. Furthermore, glycoside compounds may improve chemotherapy efficacy and reduce drug toxicity, offering better treatment options for immune-suppressed TNBC patients. However, the precise mechanisms are not yet fully understood, and need further investigation in order to enhance the use of glycoside compounds in treating TNBC.
Table 2. Profiles of glycosides in TCM with therapeutic effects on TNBC.
Table 2. Profiles of glycosides in TCM with therapeutic effects on TNBC.
Natural ProductsStructureSourcesConcentration In VitroDosage In VivoBiological Activities/MechanismsPotential TargetsReference
RutinMolecules 30 01201 i011Acacia erioloba E. Mey.150, 300 μM30 mg/kgRutin acts as an inhibitor of c-Met Kinase and effectively inhibits the proliferation of TNBC cells both in vitro and in vivo./[56]
IcariinMolecules 30 01201 i012Epimedium wushanense Maxim.5, 10, 20 μM20, 40 mg/kgInduces apoptosis and suppresses the migration of TNBC cells through modulation of the Sirtuin (Sirt) 6 /NF-κB signaling pathway./[47]
Vanicoside BMolecules 30 01201 i013Persicaria dissitiflora H. Hara5, 10, 20 μM5, 10 mg/kgThe inhibition of CDK8-mediated signaling pathways leads to downregulation of EMT proteins and instigates cell cycle arrest and apoptosis in TNBC cells./[43]
Anemoside A3Molecules 30 01201 i014Pulsatilla chinensis (Maxim.) Klob.50, 100 μg/mL10, 20 mg/kgPromotes differentiation of M0 macrophages into the M1 phenotype, which hinges upon the Toll-like receptor 4 (TLR4) /NF-κB/MAPK signaling cascade.TLR4[68]
AsiaticosideMolecules 30 01201 i015Centella asiatica (L.) Urban0.25 μM1, 2, 3 mg/kgInhibits P2RX7-mediated TGF-β/SMAD signaling through upregulation of PPARγ expression, thus attenuating EMT in TNBC.PPARγ[44]
Astragaloside IVMolecules 30 01201 i016Astragalus membranaceus Bunge20, 40 μg/mL20, 40 mg/kgReduces Vav3 expression./[54]
Ginsenoside Rg1Molecules 30 01201 i017Panax ginseng C. A. Mey.10 μM/Activates the DNA damage-response elements [Ataxia telangiectasia mutated protein kinase (ATM), H2A histone family member X (H2AX), Radiation-sensitive 51 (RAD51), X-ray repair cross-complementing protein 1(XRCC1), apoptosis-related genes (P21, TP53, Apoptotic peptidase activating factor 1 (APAF1), BAX, Caspase 3(CASP3), and Caspase 9(CASP9))] in MDA-MB-231 cells./[66,69]
Ginsenosides Rg3Molecules 30 01201 i018Panax ginseng C. A. Mey.300 mg/mL/Increases the Bax/Bcl-2 ratio, causes mitochondrial membrane potential depolarization, leads to cytochrome c release, and induces caspase-3 and PARP cleavage./[64,65]
Saikosaponin DMolecules 30 01201 i019Bupleurum rotundifolium L.10, 15, 30 μM/Suppresses the proliferation of TNBC cells through modulation of the β-catenin signaling cascade.β-catenin[70]
ScutellarinMolecules 30 01201 i020Erigeron breviscapus (Vaniot) Hand. -Mazz.20 μM10 mg/kgReduces TNBC metastasis through the targeting of TNFα/TNFR2-initiated activation of RUNX1 and subsequent production of G-CSF in TNBC-associated endothelial cells.TNFα/TNFR2[71]

2.3. Phenolics

Phenolic compounds are composed of aromatic rings and hydroxyl groups. These compounds demonstrate a variety of biological activities, such as antioxidant, anti-inflammatory, antitumor, and antimicrobial effects [72,73,74,75]. Their antioxidant properties arise from the hydroxyl groups on the aromatic rings, which can donate electrons [75]. The natural world is rich in phenolic compounds, and recent studies have shown that some of these compounds can effectively combat TNBC (Figure 3, Table 3 and Table S1).

2.3.1. Anti-Metastasis

Recent studies indicate that phenolic compounds can inhibit the metastasis of TNBC cells. For instance, (-)-sativan may regulate miR-200c, thereby affecting the EMT process and Programmed death-ligand 1 (PD-L1) expression in TNBC cells [76]. Similarly, isoliquiritigenin can also increase miR-200c levels, thereby suppressing the EMT process [77]. Propolin G inhibits the EMT in TNBC cells by degrading the cytoskeletal protein vimentin. This process is mediated by Glycogen synthase kinase 3β (GSK-3β) and regulated by Snail and Histone deacetylase 6 (HDAC6) [78]. Baicalein effectively reverses the expression of Interferon-induced protein with tetratricopeptide repeats 2 (IFIT2), which is associated with metastasis, recurrence, and poor prognosis in TNBC patients [79]. Norstictic acid significantly inhibits the proliferation, migration, and invasion of TNBC cells, while it shows minimal toxicity to non-tumorigenic MCF-10A mammary epithelial cells. Its potential mechanism involves the inhibition of c-Met phosphorylation [80]. Gigantol prevents TNBC cell migration and invasion by suppressing the Wnt/β-catenin signaling pathway [81].

2.3.2. Anti-Angiogenesis

Phenolic compounds can inhibit the progression of TNBC by suppressing angiogenesis, primarily through the inhibition of VEGFA. Emodin exhibits anti-angiogenic effects in TNBC by targeting the transcriptional regulators nuclear receptor corepressor 2 (NCOR2) and Seryl-tRNA synthetase (SerRS), thereby suppressing VEGFA transcription [82]. Research shows that the hydroxyl group at the C-3 position of Emodin is crucial for its anticancer activity [83]. Rhamnazin, a new vascular endothelial growth factor receptor 2 (VEGFR2) inhibitor, blocks tumor angiogenesis and growth. Rhamnazin significantly reduced the proliferation, migration, and tube formation of human umbilical vascular endothelial cells (HUVECs) in vitro, and also inhibited sprout formation in rat aorta ring assays [84]. Cirsimaritin reduced angiogenesis in MDA-MB-231 cells by lowering the levels of vascular endothelial growth factor (VEGF), p-AKT, and p-ERK [85].

2.3.3. Other Anti-TNBC Effects

Phenolic compounds not only inhibit angiogenesis and metastasis in TNBC, but also suppress its progression through other mechanisms. Anhydroicaritin induces apoptosis in TNBC cells [86]. Isoliquiritigenin inhibits the proliferation and migration of TNBC cells by reducing the expression of miR-374a, which negatively regulates PTEN and results in increased apoptosis and decreased invasiveness by inactivating the AKT pathway [77]. Kaempferol inhibits the expression of Sirt3 and Sirt6, which are potential therapeutic targets for TNBC due to their roles in cancer stem cell activity, chemotherapy resistance, and metastasis [87]. The combination of Morin and doxorubicin enhances the efficacy against TNBC [88]. Baicalein increases the sensitivity of TNBC cells to doxorubicin by promoting autophagy-mediated downregulation of CDK1 [89,90].
Recent studies have reported that phenolic compounds suppress metastasis and angiogenesis, induce apoptosis in TNBC cells, and enhance the sensitivity of TNBC to chemotherapy drugs. The inhibition of angiogenesis by phenolic compounds is closely linked to VEGFA, which plays a crucial role in this process by promoting the proliferation, migration, and lumen formation of endothelial cells, thereby directly influencing vascular growth and development. In the tumor microenvironment, VEGFA overexpression is strongly associated with tumor angiogenesis, creating conditions that support tumor growth [91,92]. Phenolic compounds affect TNBC cells through various mechanisms to prevent the onset and progression of the disease. For example, isoliquiritigenin has been shown to simultaneously suppress TNBC cell metastasis and induce apoptosis. Despite the promising results of phenolic compounds in anti-TNBC research, further studies are necessary to evaluate their potential for clinical application.

2.4. Alkaloids

Alkaloids are a group of naturally occurring compounds characterized by nitrogen-containing heterocyclic structures, and are frequently found in various plants. Current investigations indicate that alkaloids such as berberine, matrine, fangchinoline, and Dragmacidin D, sourced from diverse organisms, exhibit therapeutic promise against TNBC through the inhibition of proliferation [93,94,95,96,97,98] (Figure 4, Table 4 and Table S1).
Table 3. Profiles of phenolics in TCM with therapeutic effects on TNBC.
Table 3. Profiles of phenolics in TCM with therapeutic effects on TNBC.
Natural ProductsStructureSourcesConcentration In VitroDosage In VivoBiological Activities/MechanismsPotential TargetsReference
(-)-Sativan(SA)Molecules 30 01201 i021Spatholobus suberectus Dunn5, 10, 20 μM25, 50 mg/kg Upregulates miR-200c to suppress the process of EMT./[76]
AnhydroicaritinMolecules 30 01201 i022Epimedium brevicornu Maxim.10, 20, 30 μM20 mg/kgInhibits the HIF-1α/VEGFA signaling pathway./[86]
EmodinMolecules 30 01201 i023Rheum palmatum L.0.25, 10, 25 μM40 mg/kgRegulates the activity of NCOR2; inhibits SIK3; suppresses TGF-β1.NCOR2[82]
IsoliquiritigeninMolecules 30 01201 i024Spatholobus suberectus Dunn10 μM20, 40 mg/kgReduces miR-374a expression; elevates BAX protein and mRNA levels by inhibiting miR-374a; enhances miR-200c expression./[99,100]
Kaempferol Molecules 30 01201 i025Caragana frutex (L.) C. Koch12.5, 25 μg/mL,/Inhibits Sirt3 and Sirt6 proteins, inhibiting the PI3K/AKT pathway.Sirts[87]
MorinMolecules 30 01201 i026Maclura pomifera (Raf.) C. K. Schneid., Petasites formosanus Kitam.10, 50, 100, 200 μM10 mg/kgInhibits the growth and invasion of the highly metastatic BC cell line MDA-MB-231, which is partly achieved through the suppression of the AKT pathway./[100,101]
Propolin GMolecules 30 01201 i027Taiwanese propolis5, 10, 15, 20 μM/Inhibits the stabilization of vimentin protein, mediated by GSK-3β-Snail, and interferes with HDAC6, thereby impeding the suppression of cell migration and invasion in TNBC.GSK-3β, HDAC6[78]
RhamnazinMolecules 30 01201 i028Callicarpa kwangtungensis chun, Alocnemum strobilaceum10, 15, 20 μM200 mg/kgSuppresses VEGF-induced phosphorylation of VEGFR2 and its downstream signaling mediators in HUVECs.VEGFR2[84]
BaicaleinMolecules 30 01201 i029Scutellaria baicalensis Georgi50, 100 ng/ml50 mg/kgReverses the expression of IFIT2./[89,102]
Norstictic acidMolecules 30 01201 i030Dimelaena oreina, Umbilicaria virginis15, 30 μM15 mg/kgInhibits the proliferation, migration, and invasion of TNBC MDA-MB-231 cells./[80]

Anti-Proliferation

Alkaloids have been reported to inhibit the proliferation of TNBC, with the PI3K/AKT pathway playing a crucial role. Fangchinoline inhibits the proliferation of MDA-MB-231 cells by targeting the AKT/GSK-3β/CCND1 signaling pathway. Additionally, fangchinoline has been shown to induce G1 phase cell cycle arrest and promote apoptosis in these cells [94]. Matrine disrupts the PI3K/AKT signaling pathway by reducing levels of PI3K, AKT, phosphorylated AKT, and phosphoglycerate kinase 1, leading to the inhibition of TNBC cell growth. Furthermore, matrine inhibits TNBC cell proliferation and induces apoptosis by suppressing HN1 expression [97]. Harmine inhibits the proliferation, invasion, and migration of MDA-MB-231 cells, while promoting apoptosis. This effect may result from the suppression of the EMT process and the PI3K/AKT signaling pathway [103]. Berberine inhibits TNBC cell growth by targeting the GPER1/NF-κB pathway, and also influences TNBC cell migration by reducing IL-6 secretion, and LH2 expression by modulating glycolysis [104,105]. Dragmacidin D effectively induces apoptosis in spheroids of TNBC [98]. Ungeremine exhibits cytotoxic effects on MDA-MB-231 cells by inducing ferroptosis, necroptotic apoptosis, and autophagy, alongside apoptosis, via caspase activation. It also affects mitochondrial membrane potential and increases ROS production [106].
In general, alkaloids inhibit cell migration, promote apoptosis, and block the cell cycle, thereby exerting antitumor effects in TNBC. Fangchinoline has been reported to inhibit TNBC cell growth, migration, and proliferation. Research indicates that substituting benzyl units in fangchinoline is crucial for significantly enhancing its antitumor activity [107]. This insight directs our efforts to synthesize fangchinoline derivatives, with the goal of finding more effective and less toxic candidates. Dragmacidin D effectively induces apoptosis in TNBC spheroids. However, it does not show cytotoxic effects on the same cell lines after prolonged incubation under traditional 2D growth conditions [98]. This suggests that establishing suitable in vitro models is essential. Most of these alkaloids have demonstrated anticancer activity in vitro, but lack data in vivo. Further in-depth research is needed.
While this review focuses on terpenoids, glycosides, phenolics, and alkaloids as the predominant anti-TNBC agents identified in our screening, a comprehensive list of additional compounds with reported activity (including the steroid, withanolide, unsaturated fatty acids, and furanocoumarins) is systematically cataloged in Supplementary Table S1, annotated with their chemical structures, source organisms, and experimental validation data.
Table 4. Profiles of alkaloids in TCM with therapeutic effects on TNBC.
Table 4. Profiles of alkaloids in TCM with therapeutic effects on TNBC.
Natural ProductsStructureSourcesConcentration In VitroDosage In Vivo Biological Activities/MechanismsPotential TargetsReference
FangchinolineMolecules 30 01201 i031Stephania tetrandra (Radix)6, 12, 24 μM5, 10, 30 μM/Inhibits the AKT/Gsk 3β/CCND1 signaling pathway; blocks cell cycle progression at the G1 phase; induces apoptosis./[94,95]
Dragmacidin DMolecules 30 01201 i032Genus Spongosorites3.8, 7.6 μM/Induces apoptosis in TNBC spheroids./[98]
ProtopineMolecules 30 01201 i033Corydalis yanhusuo1, 10, 100 μM/Specifically inhibits the cell adhesion ability of MDA-MB-231./[108]
Subereamolline AMolecules 30 01201 i034Pseudoceratina arabica2 μM/Inhibits the migration and invasion of MDA-MB-231 cells./[109]
HarmineMolecules 30 01201 i035Peganum harmala10 μM/Inhibits the process of EMT, associated with the PI3K/AKT signaling pathway./[110]
UngeremineMolecules 30 01201 i036Crinum zeylanicum3.67 μM/Induces ferroptosis, necroptosis, and autophagy, as well as apoptosis, mediated by caspase activation, MMP alteration, and increase ROS production./[106]
BerberineMolecules 30 01201 i037Coptis chinensis1.25, 2.5, 5 μM15, 30 mg/mLReduces IL-6 secretion and LH2 expression; modulates the GPER1/NF-κB pathway; inhibits the NLRP3 inflammasome pathway.LH2[107,111]
MatrineMolecules 30 01201 i038Sophora flavescens2, 4 μM/Inhibits the PI3K/AKT signaling pathway; suppresses HN1 expression./[93,97]

3. Natural Products: Mechanisms of Inhibiting TNBC

Emerging evidence highlights the multi-target mechanisms of NPs—including terpenes, glycosides, phenolics, and alkaloids—against TNBC. These compounds suppress tumor progression through diverse pathways, such as metastasis inhibition, apoptosis induction, cell cycle arrest, and tumor microenvironment modulation. Their polypharmacological effects often arise from modulating crosstalk between key signaling nodes, particularly the TGF-β, VEGFA, PI3K/AKT, Wnt/β-catenin, and MAPK pathways. By elucidating the interplay of these critical pathways, the following section further illuminates how NPs exert their therapeutic potential in TNBC.

3.1. TGF-β Signaling Pathway

Transcriptional regulation of EMT induced by TGF-β involves a complex cascade of events. Initially, in response to TGF-β, Smad2 and Smad3 become activated and form complexes with Smad4. These complexes then regulate the transcription of target genes by interacting with other DNA-binding transcription factors. During the induction of EMT, the activated Smads play a pivotal role in mediating transcriptional regulation through three distinct families of transcription factors. This process ultimately leads to the repression of epithelial marker gene expression and the activation of mesenchymal gene expression, which are hallmarks of EMT.
Additionally, TGF-β directly activates non-Smad signaling pathways. This activation occurs through the interaction of signaling mediators either directly with the TβRII and/or TβRI receptors, or via adaptor proteins. Among these non-Smad signaling responses, the activation of Erk MAP kinases, Rho family GTPases (Rho GTPases), and the PI3K /AKT pathway in response to TGF-β has been specifically linked to TGF-β-induced EMT. These pathways regulate various cellular processes, including cytoskeleton organization and cell growth, survival, migration, and invasion, which are crucial for the EMT process. The discovery of ITSN, AS, and harmine demonstrates their capacity to inhibit the TGF-β pathway, thereby suppressing EMT. This inhibition, in turn, contributes to the suppression of TNBC. Therefore, screening NPs that modulate the TGF-β pathway represents a promising strategy for the development of anti-TNBC therapies.

3.2. VEGFA Signaling Pathway

VEGFA is a key regulator of angiogenesis, stimulating endothelial cell proliferation, migration, and new blood vessel formation by its interaction with its primary receptors, VEGFR1 and VEGFR2 [112,113]. Tumor progression is frequently linked to enhanced angiogenesis, with VEGFA playing a central role in this process. As the tumor grows, increased VEGFA expression stimulates the formation of new blood vessels that provide essential nutrients to the tumor and support the spread of cancer cells [114,115]. Research indicates that elevated VEGFA levels are associated with poor clinical outcomes [116]. Compounds such as anhydroicaritin, rhamnazin, and emodin inhibit angiogenesis and metastasis by targeting the VEGFA pathway. VEGFA serves as a key target protein for treating TNBC, and developing and screening NPs that inhibit the VEGFA pathway could provide new therapeutic approaches for TNBC.

3.3. PI3K Signaling Pathway

The PI3K/AKT pathway is one of the most critical intracellular signaling pathways, playing a key role in various biological processes, such as cell growth, proliferation, metabolism, and survival. PI3K is a lipid kinase that activates downstream signaling by phosphorylating phosphatidylinositol 4,5-bisphosphate to form phosphatidylinositol 3,4,5-trisphosphate. The generation of PIP3 promotes the recruitment and activation of AKT, which regulates various downstream effectors, like mTOR, involved in processes like cell proliferation, survival, and metabolism [117]. Overactivation of the PI3K/AKT pathway is often associated with various cancer types. Studies have shown that abnormal activation of this pathway can result from mechanisms such as PIK3CA gene mutations or PTEN deletion or inactivation [118]. These genetic alterations lead to persistent activation of the pathway, promoting tumor cell proliferation and survival. Compounds such as AT-1, rhamnazin, norstictic acid, harmine, T-96, and matrine inhibit the PI3K/AKT pathway by targeting either PI3K or the phosphorylated forms of AKT. This inhibition induces apoptosis, suppresses cell migration, and reduces cell proliferation in TNBC. AKT promotes tumor cell survival by phosphorylating pro-apoptotic proteins, such as Bcl-2-associated death promoter and Caspase-9, thereby enhancing the cells’ ability to evade apoptosis, and it also plays a crucial role in tumor invasion and metastasis [119]. Overactivation of the PI3K/AKT pathway can enhance the motility and invasiveness of tumor cells, allowing them to breach the basement membrane and invade surrounding tissues [120]. Additionally, the PI3K/AKT pathway enhances the activation of tumor-associated fibroblasts, which supports tumor metastasis. Tumor growth and metastasis depend on an effective blood supply, and the PI3K/AKT pathway plays a crucial role in tumor angiogenesis. This pathway regulates the expression of VEGF, promoting the formation of new blood vessels [121]. Therefore, developing NP-based inhibitors of the PI3K/AKT pathway could provide new therapeutic strategies for TNBC.

3.4. Wnt/β-Catenin Signaling Pathway

The Wnt/β-catenin signaling pathway is essential for various physiological processes, including cell proliferation, differentiation, migration, and tissue homeostasis [122,123]. When Wnt proteins bind to Frizzled receptors on the cell surface, this pathway is activated, leading to the activation of β-catenin’s transcriptional activity. In the absence of Wnt signals, β-catenin is degraded in the cytoplasm. However, when Wnt signals are present, β-catenin accumulates and translocates to the nucleus, where it binds to T-cell factor/lymphoid enhancer factor to initiate the transcription of target genes [122].
Inhibiting the Wnt/β-catenin signaling pathway can effectively suppress tumor stem cell renewal, reduce cell proliferation, enhance sensitivity to chemotherapy, and inhibit tumor metastasis [123]. Saikosaponin D, a compound shown to significantly repress β-catenin and its downstream target genes, induces apoptosis in TNBC cells. Thus, developing NPs that target and inhibit the Wnt/β-catenin signaling pathway represents a promising strategy for treating TNBC.

3.5. MAPK Signaling Pathway

The MAPK signaling cascade involves three key kinases: MAPKKK, MAPKK, and MAPK, collectively known as mitogen-activated protein kinases. These pathways play a crucial role in the onset and progression of cancer [124]. Abnormal activation of MAPK pathways, often due to mutations in Ras proteins and rapidly accelerated fibrosarcoma proteins, leads to the continuous activation of the ERK1/2 pathway [125]. Research has shown that compounds such as EO, AT-1, Icariin, and AS-IV can inhibit the MAPK pathway, thereby suppressing TNBC.
NPs offer potential for targeting TNBC through the inhibition of other various critical pathways. For instance, kaempferol has been shown to suppress the expression of Sirt3 and Sirt6, which are associated with cancer stem cell functions, chemotherapy resistance, and metastasis. Another important target in TNBC therapy is Methionine (MET); research indicates that norstictic acid and rutin can effectively inhibit MET activity.
AC enhances the ubiquitination of GPX4, while britannin promotes the ubiquitination of ZEB1, a key transcription factor involved in EMT, thereby reducing TNBC cell metastasis. EGFR, through its interaction with ligands like epidermal growth factor (EGF), activates downstream signaling pathways that regulate cell proliferation, migration, and survival. Protopine has been identified as an inhibitor of EGFR expression, contributing to the suppression of TNBC. In addition, compounds such as ungeremine, Jatamanvaltrate P, ITSN, and AC can modulate autophagy, and consequently inhibit TNBC. Therefore, future research should delve deeper into the mechanisms of these pathways to discover natural compounds that can target these processes, offering new therapeutic avenues for TNBC treatment.

4. Challenges and Opportunities

TNBC, characterized by the absence of ER, PR, and HER2 immunostaining, is refractory to both endocrine therapy and conventional molecularly targeted therapies, making chemotherapy the primary systemic treatment approach. NPs are hailed as a primary source for drug discovery, due to their immense structural and physicochemical diversity.
Each type of compound that inhibits TNBC exhibits unique characteristics and commonalities. The antitumor effects of terpenoids encompass a broad spectrum of activities, including anti-proliferative, apoptotic, anti-angiogenic, and anti-metastatic actions. Glycosides primarily exert their anticancer effects through inhibiting metastasis, inducing apoptosis, and synergizing with other medications to enhance therapeutic efficacy and modulate the tumor microenvironment. Phenolic compounds manifest their anticancer effects mainly by inhibiting angiogenesis and metastasis in TNBC, and by increasing the cancer cells’ sensitivity to chemotherapeutic agents. Alkaloids predominantly achieve their anticancer effects through anti-proliferation. These effects are closely linked to their structural features. For example, the substitution of benzyl units in fangchinoline is a key pharmacophore that enhances its anti-proliferative effect on cancer cells. Meanwhile, research shows that the introduction of fluorine atoms or fluorinated groups significantly enhances the antitumor efficacy of the compound and prolongs its pharmacokinetic profile in vivo [107]. By systematically categorizing and organizing these NPs, we might infer the potential anticancer properties of compounds with similar structures, such as 3-aryl isocoumarins and 8-hydroxy-3-aryl isocoumarin, which are akin to kaempferol, morin, propolin G, rhamnazin, and baicalein, and may also exhibit certain anti-TNBC activities [126,127].
To comprehensively assess the therapeutic potential of NPs in TNBC, we compared their in vitro and in vivo efficacy with current standard treatments. NPs such as AC, AT-1, Vanicoside B, SA, and emodin demonstrated promising anticancer activity in cellular and animal models. For instance, AC exhibited potent effects against MDA-MB-231 cells (IC50 = 1.02 μM) and significantly inhibited tumor growth in murine TNBC models. Rutin reduced tumor volume by 53.85% and 65.72% in mouse TNBC models. Compounds like T-96, ITSN, and AS showed remarkable efficacy at low doses in vivo. While these findings are derived from preclinical studies, they provide critical scientific rationale for further development of NPs. Paclitaxel, a microtubule inhibitor, stabilizes microtubules to block cell cycle progression and induce apoptosis. Clinical trials have demonstrated its efficacy in TNBC, particularly when combined with carboplatin, improving overall and disease-free survival rates [128]. PARP inhibitors (e.g., Olaparib, Talazoparib) target BRCA-mutated TNBC by impairing DNA repair mechanisms. These agents are approved for metastatic TNBC patients with germline BRCA1/2 mutations [129]. NPs such as AC and T-96 have shown minimal toxicity in preclinical models, with no significant hepatorenal damage or adverse effects observed during prolonged administration. In contrast, paclitaxel is associated with dose-limiting neurotoxicity, myelosuppression, and hypersensitivity reactions, while PARP inhibitors may cause hematologic and gastrointestinal toxicities [130]. Although current research on NPs remains preclinical, advances in translational medicine are bridging the gap between laboratory discoveries and clinical applications. Further studies should prioritize evaluating their therapeutic potential in specific TNBC subtypes (e.g., BRCA-mutated tumors), for which NPs may offer distinct advantages in efficacy and tolerability.
NPs possess unparalleled structural diversity, a hallmark advantage derived from the vast biochemical repertoire of plants, animals, and microorganisms. These evolutionarily refined compounds exhibit intricate and unique architectures that far surpass the structural complexity of most synthetic counterparts. Such chemical distinctiveness enables NPs to engage with diverse biological targets through multifaceted interactions, positioning them as invaluable resources for drug discovery. In the context of TNBC—a malignancy characterized by dysregulation of multiple signaling pathways (e.g., PI3K/AKT/mTOR, VEGFA) and aberrant crosstalk within oncogenic networks—monotherapies targeting single pathways often yield suboptimal outcomes, due to compensatory resistance mechanisms. The polypharmacological properties of NPs enable them to simultaneously modulate multiple therapeutic targets, offering a promising strategy to address the heterogeneity and adaptive resistance of TNBC. For instance, AT-1 has been demonstrated to inhibit phosphorylation of the ERK family and suppress the expression of TPI1 and GPI, demonstrating significant anti-TNBC activity in preclinical models [21]. This multi-targeted approach enhances therapeutic efficacy and mitigates compensatory resistance mechanisms that are commonly observed in TNBC.
However, the clinical translation of NPs for TNBC faces multiple challenges, requiring prioritized research. First, there is an intricate relationship between the structural complexity of NPs and their toxicity; for example, the pentacyclic ring structure of TSN, which contains a carbonyl group, has been associated with marked hepatotoxicity, whereas ITSN, featuring an oxygen bridge, exhibits significantly reduced toxicity [37,131,132]. Second, conventional screening paradigms overemphasizing direct cytotoxicity (e.g., oxoflavidin’s prioritization based solely on MDA-MB-231 cell inhibition) risk overlooking multimodal therapeutic mechanisms, despite murine models remaining indispensable for preclinical validation [133]. Furthermore, critical gaps persist in understanding NP-associated safety profiles and comparative advantages over existing therapies. Future investigations must integrate structural optimization, mechanism elucidation, comprehensive safety assessments, and comparative efficacy studies to advance clinically viable NP-based TNBC therapeutics.

5. Conclusions

NPs exhibit unparalleled chemical diversity, underpinning their remarkable potential as reservoirs of bioactive compounds with multifaceted pharmacological properties. Emerging evidence highlights their growing significance in addressing TNBC, with numerous NPs demonstrating antitumor efficacy through diverse mechanisms.
In this review, we have compiled information on NPs with anti-TNBC properties. These natural agents predominantly comprise a variety of compounds, such as terpenoids, glycosides, phenolic substances, and alkaloids. A profound exploration has revealed their participation in crucial signaling pathways, specifically the TGF-β, VEGFA, PI3K/AKT, Wnt/β-catenin, and MAPK signaling pathways. Although these NPs exhibit potential anticancer activities, the studies on them remain at the preclinical basic research stage, and further exploration is still required.

Supplementary Materials

The following supporting information can be downloaded at https://www.mdpi.com/article/10.3390/molecules30061201/s1. A comprehensive list of additional compounds with reported activity (including the steroid, withanolide, unsaturated fatty acids, and furanocoumarins) [134,135,136,137,138,139,140,141,142,143,144,145,146,147,148,149,150,151,152,153,154,155,156,157,158,159,160,161,162,163,164,165,166,167,168,169,170,171,172,173,174,175,176,177,178,179,180,181,182,183].

Author Contributions

Conceptualization, Q.L. and Z.Y.; methodology, Q.L.; investigation, G.W.; resources, Y.C.; data curation, J.D.; writing—original draft preparation, Q.L.; writing—review and editing, Z.Y., D.W. and Y.W.; visualization, Q.L. and Z.Y.; supervision, D.W.; project administration, Y.W.; funding acquisition, D.W. All authors have read and agreed to the published version of the manuscript.

Funding

The National Natural Science Foundation of China, grant number 82172723; the Science and Technology Department of Sichuan Province, grant number 2024YFFK0156; the Key Projects of Science and Technology Plan of Inner Mongolia Autonomous Region, grant number 201802115; and the Innovation Team and Talents Cultivation Program of the National Administration of Traditional Chinese Medicine. The APC was funded by the National Key R&D Program of China, grant number 2023YFF0720300.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

No new data were created or analyzed in this study. Data sharing is not applicable to this article.

Acknowledgments

The figures were created with BioRender.com (accessed on 4 March 2025).

Conflicts of Interest

The authors declare no conflicts of interest.

Abbreviations

The following abbreviations are used in this manuscript:
TNBCTriple-negative breast cancer
NPsNatural products
BCBreast cancer
WHOWorld Health Organization
HER2Human epidermal growth receptor 2
EREstrogen receptor
PRProgesterone receptor
TCMsTraditional Chinese medicines
HMsHerbal medicines
FDAFood and Drug Administration
AT-1Atractylenolide-1
EMTEpithelial–mesenchymal transition
ASAsiaticoside
ITSNIsotoosendanin
GPX4Glutathione peroxidase 4
T-96Demethylzeylasteral
ACAnomanolide C
TPI1Triosephosphate Isomerase 1
GPIGlucose-6-phosphate isomerase
EOEupalinolide O
LSD1Lysine-specific demethylase 1
PTENPhosphatase and tensin homolog
MAMaackiain
ROSReactive oxygen species
CDK1Cyclin-dependent kinase
CCND1Cyclin D1
PARPPoly adenosinediphosphate-ribose polymerase
BRCABreast cancer gene
TGF-βTransforming growth factor-beta
VEGFVascular endothelial growth factor
PI3KPhosphoinositide3kinase
AKTProtein kinase B
WntWingless/Int-1
AS- IVAstragaloside IV
SCScutellarin
PDPlatycodin D
TNFαTumor necrosis factor alpha
TNFR2Tumor necrosis factor receptor 2
TGFβR1TGF-β receptor type-1
ERKExtracellular signal-regulated kinase
JNKc-Jun N-terminal kinase
Rho GTPaseRho family GTPases
NF-κBNuclear Factor kappa B
Bcl2B-cell lymphoma 2
BaxBcl-2-associated X protein
c-MycCellular myelocytomatosis oncogene
ZEB1Zinc finger e-box binding homeobox 1
Cdc-2Cell division cycle 2
MAPKMitogen-activated protein kinase
PTP1BProtein tyrosine phosphatase 1B
CDK8Cyclin-Dependent Kinase 8
Vav3Vav Family Member 3
RUNX1Runt-related transcription factor 1
G-CSFGranulocyte-colony stimulating factor
MMP-9Matrix metalloproteinase-9
EGFREpidermal growth factor receptor
NTSR1Neurotensin receptor 1
Keap1Kelch-like epichlorohydrin-associated protein 1
STAT3Signal transducer and activator of transcription 3
SirtSirtuin
TLR4Toll-like receptor 4
ATMAtaxia telangiectasia mutated protein kinase
H2AXH2A histone family member X
RAD51Radiation-sensitive 51
XRCC1X-ray repair cross-complementing protein 1
APAF1Apoptotic peptidase activating factor 1
CASP3Caspase 3
CASP9Caspase 9
PD-L1Programmed death-ligand 1
miRmicroRNA
GSK-3βGlycogen synthase kinase 3β
HDAC6Histone deacetylase 6
IFIT2Interferon-induced protein with tetratricopeptide repeats 2
NCOR2Nuclear receptor corepressor 2
SerRSSeryl-tRNA synthetase
HUVECsHuman umbilical vascular endothelial cells
TSNToosendanin
METMethionine
VEGFR2Vascular endothelial growth factor receptor 2

References

  1. 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]
  2. Prat, A.; Adamo, B.; Cheang, M.C.U.; Anders, C.K.; Carey, L.A.; Perou, C.M. Molecular Characterization of Basal-Like and Non-Basal-Like Triple-Negative Breast Cancer. Oncologist 2013, 18, 123–133. [Google Scholar] [CrossRef] [PubMed]
  3. Garrido-Castro, A.C.; Lin, N.U.; Polyak, K. Insights into molecular classifications of triple-negative breast cancer: Improving patient selection for treatment. Cancer Discov. 2019, 9, 176–198. [Google Scholar] [CrossRef] [PubMed]
  4. Dawson, J.C.; Serrels, A.; Stupack, D.G.; Schlaepfer, D.D.; Frame, M.C. Targeting FAK in anticancer combination therapies. Nat. Rev. Cancer 2021, 21, 313–324. [Google Scholar] [CrossRef]
  5. Zhang, F.; Wang, Z.; Fan, Y.; Xu, Q.; Ji, W.; Tian, R.; Niu, R. Elevated STAT3 signaling-mediated upregulation of MMP-2/9 confers enhanced invasion ability in multidrug-resistant breast cancer cells. Int. J. Mol. Sci. 2015, 16, 24772–24790. [Google Scholar] [CrossRef]
  6. Koudelka, Š.; Turánek, J. Liposomal paclitaxel formulations. J. Control. Release 2012, 163, 322–334. [Google Scholar] [CrossRef]
  7. Samaan, T.M.A.; Samec, M.; Liskova, A.; Kubatka, P.; Büsselberg, D. Paclitaxel’s mechanistic and clinical effects on breast cancer. Biomolecules 2019, 9, 789. [Google Scholar] [CrossRef]
  8. Ferrari, P.; Scatena, C.; Ghilli, M.; Bargagna, I.; Lorenzini, G.; Nicolini, A. Molecular Mechanisms, Biomarkers and Emerging Therapies for Chemotherapy Resistant TNBC. Int. J. Mol. Sci. 2022, 23, 1665. [Google Scholar] [CrossRef]
  9. Li, Y.; Zhang, H.; Merkher, Y.; Chen, L.; Liu, N.; Leonov, S.; Chen, Y. Recent advances in therapeutic strategies for triple-negative breast cancer. J. Hematol. Oncol. 2022, 15, 121. [Google Scholar] [CrossRef]
  10. Keenan, T.E.; Tolaney, S.M. Role of immunotherapy in triple-negative breast cancer. J. Natl. Compr. Cancer Netw. 2020, 18, 479–489. [Google Scholar] [CrossRef]
  11. Li, H.; Liu, Z.Y.; Wu, N.; Chen, Y.C.; Cheng, Q.; Wang, J. PARP inhibitor resistance: The underlying mechanisms and clinical implications. Mol. Cancer 2020, 19, 107. [Google Scholar] [CrossRef] [PubMed]
  12. Yin, S.Y.; Wei, W.C.; Jian, F.Y.; Yang, N.S. Therapeutic applications of herbal medicines for cancer patients. Evid.-Based Complement. Altern. Med. 2013, 2013, 302426. [Google Scholar] [CrossRef] [PubMed]
  13. Pan, S.-Y.; Litscher, G.; Gao, S.-H.; Zhou, S.-F.; Yu, Z.-L.; Chen, H.-Q.; Zhang, S.-F.; Tang, M.-K.; Sun, J.-N.; Ko, K.-M. Historical perspective of traditional indigenous medical practices: The current renaissance and conservation of herbal resources. Evid.-Based Complement. Altern. Med. 2014, 2014, 525340. [Google Scholar] [CrossRef]
  14. Newman, D.J.; Cragg, G.M. Natural Products as Sources of New Drugs over the Nearly Four Decades from 01/1981 to 09/2019. J. Nat. Prod. 2020, 83, 770–803. [Google Scholar] [CrossRef]
  15. Tholl, D. Biosynthesis and biological functions of terpenoids in plants. Adv Biochem Eng Biotechnol. 2015, 148, 63–106. [Google Scholar] [CrossRef]
  16. El-Baba, C.; Baassiri, A.; Kiriako, G.; Dia, B.; Fadlallah, S.; Moodad, S.; Darwiche, N. Terpenoids’ anti-cancer effects: Focus on autophagy. Apoptosis 2021, 26, 491–511. [Google Scholar] [CrossRef]
  17. Zhao, M.; Tang, Y.; Xie, J.; Zhao, Z.; Cui, H. Meroterpenoids produced by fungi: Occurrence, structural diversity, biological activities, and their molecular targets. Eur. J. Med. Chem. 2021, 209, 112860. [Google Scholar] [CrossRef]
  18. Zeng, T.; Liu, Z.; Liu, H.; He, W.; Tang, X.; Xie, L.; Wu, R. Exploring Chemical and Biological Space of Terpenoids. J. Chem. Inf. Model. 2019, 59, 3667–3678. [Google Scholar] [CrossRef]
  19. Carneiro, B.A.; El-Deiry, W.S. Targeting apoptosis in cancer therapy. Nat. Rev. Clin. Oncol. 2020, 17, 395–417. [Google Scholar] [CrossRef]
  20. Zhang, J.; Yang, F.; Mei, X.; Yang, R.; Lu, B.; Wang, Z.; Ji, L. Isotoosendanin inhibits triple-negative breast cancer metastasis by reducing mitochondrial fission and lamellipodia formation regulated by the Smad2/3-GOT2-MYH9 signaling axis. Acta Pharmacol. Sin. 2024, 45, 2672–2683. [Google Scholar] [CrossRef]
  21. Xu, H.; Li, L.; Qu, L.; Tu, J.; Sun, X.; Liu, X.; Xu, K. Atractylenolide-1 affects glycolysis/gluconeogenesis by downregulating the expression of TPI1 and GPI to inhibit the proliferation and invasion of human triple-negative breast cancer cells. Phytother. Res. 2023, 37, 820–833. [Google Scholar] [CrossRef] [PubMed]
  22. Li, L.; Ji, Y.; Fan, J.; Li, F.; Li, Y.; Wu, M.; Cheng, H.; Xu, C. Demethylzeylasteral (T-96) inhibits triple-negative breast cancer invasion by blocking the canonical and non-canonical TGF-β signaling pathways. Naunyn-Schmiedeberg’s Arch. Pharmacol. 2019, 392, 593–603. [Google Scholar] [CrossRef] [PubMed]
  23. Zhao, Y.; Fu, L.; Chen, J.; Zhou, J.; Tian, C.; Zhou, D.; Zhu, R. Eupalinolide O Induces Apoptosis in Human Triple-Negative Breast Cancer Cells via Modulating ROS Generation and Akt/p38 MAPK Signaling Pathway. J. Oncol. 2022, 2022, 8802453. [Google Scholar] [CrossRef] [PubMed]
  24. Yang, B.; Zhu, R.; Tian, S.; Wang, Y.; Lou, S.; Zhao, H. Jatamanvaltrate P induces cell cycle arrest, apoptosis and autophagy in human breast cancer cells in vitro and in vivo. Biomed. Pharmacother. 2017, 89, 1027–1036. [Google Scholar] [CrossRef]
  25. Shen, Z.; Gu, Y.; Jiang, R.; Qian, H.; Li, S.; Xu, L.; Gu, W.; Zuo, Y. Antitumor Effect of Demethylzeylasteral (T-96) on Triple-Negative Breast Cancer via LSD1-Mediate Epigenetic Mechanisms. Anal. Cell. Pathol. 2022, 2022, 2522597. [Google Scholar] [CrossRef]
  26. Peng, F.; Wang, L.; Xiong, L.; Tang, H.; Du, J.; Peng, C. Maackiain Modulates miR-374a/GADD45A Axis to Inhibit Triple-Negative Breast Cancer Initiation and Progression. Front. Pharmacol. 2022, 13, 806869. [Google Scholar] [CrossRef]
  27. Yeo, S.K.; Ali, A.Y.; Hayward, O.A.; Turnham, D.; Jackson, T.; Bowen, I.D.; Clarkson, R. β-Bisabolene, a Sesquiterpene from the Essential Oil Extract of Opoponax (Commiphora guidottii), Exhibits Cytotoxicity in Breast Cancer Cell Lines. Phytother. Res. 2016, 30, 418–425. [Google Scholar] [CrossRef]
  28. Lin, Z.; Yang, Y.; Liu, T.; Wu, Z.; Zhang, X.; Yang, J. Germacrone alleviates breast cancer-associated osteolysis by inhibiting osteoclastogenesis via inhibition of MAPK/NF-κB signaling pathways. Phytother. Res. 2024, 38, 2860–2874. [Google Scholar] [CrossRef]
  29. Shahrestanaki, M.K.; Mirjani, A.; Ghanadian, M.; Aghaei, M. Cycloartane triterpenoid from Euphorbia macrostegia modulates ER stress signaling pathways to induce apoptosis in MDA-MB231 and MCF-7 breast cancer cell lines. Naunyn-Schmiedeberg’s Arch. Pharmacol. 2023, 396, 1749–1758. [Google Scholar] [CrossRef]
  30. Qin, J.-J.; Jin, H.-Z.; Huang, Y.; Zhang, S.-D.; Shan, L.; Voruganti, S.; Nag, S.; Wang, W.; Zhang, W.-D.; Zhang, R. Selective cytotoxicity, inhibition of cell cycle progression, and induction of apoptosis in human breast cancer cells by sesquiterpenoids from Inula lineariifolia Turcz. Eur. J. Med. Chem. 2013, 68, 473–481. [Google Scholar] [CrossRef]
  31. Chen, M.; Zhang, M.; Lu, X.; Li, Y.; Lu, C. Diselenium-linked dimeric prodrug nanomedicine breaking the intracellular redox balance for triple-negative breast cancer targeted therapy. Eur. J. Pharm. Biopharm. 2023, 193, 16–27. [Google Scholar] [CrossRef] [PubMed]
  32. Zhong, Z.; Chen, X.; Tan, W.; Xu, Z.; Zhou, K.; Wu, T.; Cui, L.; Wang, Y. Germacrone inhibits the proliferation of breast cancer cell lines by inducing cell cycle arrest and promoting apoptosis. Eur. J. Pharmacol. 2011, 667, 50–55. [Google Scholar] [CrossRef] [PubMed]
  33. Duangmano, S.; Sae-lim, P.; Suksamrarn, A.; Domann, F.E.; Patmasiriwat, P. Cucurbitacin B inhibits human breast cancer cell proliferation through disruption of microtubule polymerization and nucleophosmin/B23 translocation. BMC Complement. Altern. Med. 2012, 12, 185. [Google Scholar] [CrossRef]
  34. Luo, W.W.; Zhao, W.W.; Lu, J.J.; Wang, Y.T.; Chen, X.P. Cucurbitacin B suppresses metastasis mediated by reactive oxygen species (ROS) via focal adhesion kinase (FAK) in breast cancer MDA-MB-231 cells. Chin. J. Nat. Med. 2018, 16, 10–19. [Google Scholar] [CrossRef]
  35. Rakesh, R.; PriyaDharshini, L.C.; Sakthivel, K.M.; Rasmi, R.R. Role and regulation of autophagy in cancer. Biochim. Biophys. Acta (BBA)—Mol. Basis Dis. 2022, 1868, 166400. [Google Scholar] [CrossRef]
  36. Yun, C.W.; Jeon, J.; Go, G.; Lee, J.H.; Lee, S.H. The dual role of autophagy in cancer development and a therapeutic strategy for cancer by targeting autophagy. Int. J. Mol. Sci. 2021, 22, 179. [Google Scholar] [CrossRef]
  37. Zhang, J.-N.; Zhang, Z.; Huang, Z.-L.; Guo, Q.; Wu, Z.-Q.; Ke, C.; Lu, B.; Wang, Z.-T.; Ji, L.-L. Isotoosendanin exerts inhibition on triple-negative breast cancer through abrogating TGF-β-induced epithelial–mesenchymal transition via directly targeting TGFβR1. Acta Pharm. Sin. B 2023, 13, 2990–3007. [Google Scholar] [CrossRef]
  38. Chen, Y.-M.; Xu, W.; Liu, Y.; Zhang, J.-H.; Yang, Y.-Y.; Wang, Z.-W.; Sun, D.-J.; Li, H.; Liu, B.; Chen, L.-X. Anomanolide C suppresses tumor progression and metastasis by ubiquitinating GPX4-driven autophagy-dependent ferroptosis in triple negative breast cancer. Int. J. Biol. Sci. 2023, 19, 2531–2550. [Google Scholar] [CrossRef]
  39. Zhang, S.; Dong, Y.; Chen, X.; Tan, C.S.H.; Li, M.; Miao, K.; Lu, J.-H. Toosendanin, a late-stage autophagy inhibitor, sensitizes triple-negative breast cancer to irinotecan chemotherapy. Chin. Med. 2022, 17, 55. [Google Scholar] [CrossRef]
  40. Lu, H.; Wu, Z.; Wang, Y.; Zhao, D.; Zhang, B.; Hong, M. Study on inhibition of Britannin on triple-negative breast carcinoma through degrading ZEB1 proteins. Phytomedicine 2022, 104, 154291. [Google Scholar] [CrossRef]
  41. Zhang, M.; Meng, M.; Liu, Y.; Qi, J.; Zhao, Z.; Qiao, Y.; Hu, Y.; Lu, W.; Zhou, Z.; Xu, P.; et al. Triptonide effectively inhibits triple-negative breast cancer metastasis through concurrent degradation of Twist1 and Notch1 oncoproteins. Breast Cancer Res. 2021, 23, 116. [Google Scholar] [CrossRef] [PubMed]
  42. Gao, B.; Chen, J.; Han, B.; Zhang, X.; Hao, J.; Giuliano, A.E.; Cui, Y.; Cui, X. Identification of triptonide as a therapeutic agent for triple negative breast cancer treatment. Sci. Rep. 2021, 11, 2408. [Google Scholar] [CrossRef] [PubMed]
  43. Kim, D.; Wang, C.Y.; Hu, R.; Lee, J.Y.; Luu, T.-T.-T.; Park, H.-J.; Lee, S.K. Antitumor Activity of Vanicoside B Isolated from Persicaria dissitiflora by Targeting CDK8 in Triple-Negative Breast Cancer Cells. J. Nat. Prod. 2019, 82, 3140–3149. [Google Scholar] [CrossRef] [PubMed]
  44. Huang, X.; Jia, Z.; Li, X.; Hu, Z.; Yu, X.; Xia, J. Asiaticoside hampers epithelial–mesenchymal transition by promoting PPARG expression and suppressing P2RX7-mediated TGF-β/Smad signaling in triple-negative breast cancer. Phytother. Res. 2023, 37, 1771–1786. [Google Scholar] [CrossRef]
  45. Wang, H.; Wei, L.; Mao, D.; Che, X.; Ye, X.; Liu, Y. Combination of oxymatrine (Om) and astragaloside IV (As) enhances the infiltration and function of TILs in triple-negative breast cancer (TNBC). Int. Immunopharmacol. 2023, 125, 111026. [Google Scholar] [CrossRef]
  46. Nagai, M.; Obata, Y.; Takahashi, D.; Hase, K. Fine-tuning of the mucosal barrier and metabolic systems using the diet-microbial metabolite axis. Int. Immunopharmacol. 2016, 37, 79–86. [Google Scholar] [CrossRef]
  47. Gao, S.; Zhang, X.; Liu, J.; Ji, F.; Zhang, Z.; Meng, Q.; Zhang, Q.; Han, X.; Wu, H.; Yin, Y.; et al. Icariin Induces Triple-Negative Breast Cancer Cell Apoptosis and Suppresses Invasion by Inhibiting the JNK/c-Jun Signaling Pathway. Drug Des. Dev. Ther. 2023, 17, 821–836. [Google Scholar] [CrossRef]
  48. Tang, Z.-H.; Li, T.; Gao, H.-W.; Sun, W.; Chen, X.-P.; Wang, Y.-T.; Lu, J.-J. Platycodin D from Platycodonis Radix enhances the anti-proliferative effects of doxorubicin on breast cancer MCF-7 and MDA-MB-231 cells. Chin. Med. 2014, 9, 16. [Google Scholar] [CrossRef]
  49. Chun, J.; Kim, Y.S. Platycodin D inhibits migration, invasion, and growth of MDA-MB-231 human breast cancer cells via suppression of EGFR-mediated Akt and MAPK pathways. Chem.-Biol. Interact. 2013, 205, 212–221. [Google Scholar] [CrossRef]
  50. Son, J.-A.; Lee, S.K.; Park, J.; Jung, M.J.; An, S.-E.; Yang, H.J.; Son, S.H.; Kim, K.R.; Park, K.-K.; Chung, W.-Y. Platycodin D Inhibits Vascular Endothelial Growth Factor-Induced Angiogenesis by Blocking the Activation of Mitogen-Activated Protein Kinases and the Production of Interleukin-8. Am. J. Chin. Med. 2022, 50, 1645–1661. [Google Scholar] [CrossRef]
  51. Hajimehdipoor, H.; Tahmasvand, Z.; Nejad, F.G.; Maresca, M.; Rajabi, S. Rutin Promotes Proliferation and Orchestrates Epithelial–Mesenchymal Transition and Angiogenesis in MCF-7 and MDA-MB-231 Breast Cancer Cells. Nutrients 2023, 15, 2884. [Google Scholar] [CrossRef] [PubMed]
  52. Mei, X.; Ouyang, H.; Zhang, H.; Jia, W.; Lu, B.; Zhang, J.; Ji, L. Scutellarin suppresses the metastasis of triple-negative breast cancer via targeting TNFα/TNFR2-RUNX1-triggered G-CSF expression in endothelial cells. Biochem. Pharmacol. 2023, 217, 115808. [Google Scholar] [CrossRef] [PubMed]
  53. Mei, X.-Y.; Zhang, J.-N.; Jia, W.-Y.; Lu, B.; Wang, M.-N.; Zhang, T.-Y.; Ji, L.-L. Scutellarin suppresses triple-negative breast cancer metastasis by inhibiting TNFα-induced vascular endothelial barrier breakdown. Acta Pharmacol. Sin. 2022, 43, 2666–2677. [Google Scholar] [CrossRef]
  54. Jiang, K.; Lu, Q.; Li, Q.; Ji, Y.; Chen, W.; Xue, X. Astragaloside IV inhibits breast cancer cell invasion by suppressing Vav3 mediated Rac1/MAPK signaling. Int. Immunopharmacol. 2017, 42, 195–202. [Google Scholar] [CrossRef]
  55. Zhu, X.; Wang, K.; Chen, Y. Ophiopogonin D suppresses TGF-β1-mediated metastatic behavior of MDA-MB-231 breast carcinoma cells via regulating ITGB1/FAK/Src/AKT/β-catenin/MMP-9 signaling axis. Toxicol. Vitr. 2020, 69, 104973. [Google Scholar] [CrossRef]
  56. Elsayed, H.E.; Ebrahim, H.Y.; Mohyeldin, M.M.; Siddique, A.B.; Kamal, A.M.; Haggag, E.G.; El Sayed, K.A. Rutin as A Novel c-Met Inhibitory Lead for The Control of Triple Negative Breast Malignancies. Nutr. Cancer 2017, 69, 1256–1271. [Google Scholar] [CrossRef]
  57. Xia, J.; Zhang, S.; Zhang, R.; Wang, A.; Zhu, Y.; Dong, M.; Ma, S.; Hong, C.; Liu, S.; Wang, D.; et al. Targeting therapy and tumor microenvironment remodeling of triple-negative breast cancer by ginsenoside Rg3 based liposomes. J. Nanobiotechnol. 2022, 20, 414. [Google Scholar] [CrossRef]
  58. Nakhjavani, M.; Palethorpe, H.M.; Tomita, Y.; Smith, E.; Price, T.J.; Yool, A.J.; Pei, J.V. Stereoselective anti-cancer activities of ginsenoside rg3 on triple negative breast cancer cell models. Pharmaceuticals 2019, 12, 117. [Google Scholar] [CrossRef]
  59. Chen, J.C.; Chang, N.W.; Chung, J.G.; Chen, K.C. Saikosaponin-A induces apoptotic mechanism in human breast MDA-MB-231 and MCF-7 cancer cells. Am. J. Chin. Med. 2003, 31, 363–377. [Google Scholar] [CrossRef]
  60. Xu, F.; Zang, J.; Chen, D.; Zhang, T.; Zhan, H.; Lu, M.; Zhuge, H. Neohesperidin induces cellular apoptosis in human breast adenocarcinoma MDA-MB-231 cells via activating the Bcl-2/Bax-mediated signaling pathway. Nat. Prod. Commun. 2012, 7, 1475–1478. [Google Scholar] [CrossRef]
  61. Tandrasasmita, O.M.; Lee, J.S.; Baek, S.H.; Tjandrawinata, R.R. Induction of cellular apoptosis in human breast cancer by DLBS1425, a Phaleria macrocarpa compound extract, via downregulation of PI3-kinase/AKT pathway. Cancer Biol. Ther. 2010, 10, 814–823. [Google Scholar] [CrossRef]
  62. Wu, H.; Wei, G.; Luo, L.; Li, L.; Gao, Y.; Tan, X.; Wang, S.; Chang, H.; Liu, Y.; Wei, Y.; et al. Ginsenoside Rg3 nanoparticles with permeation enhancing based chitosan derivatives were encapsulated with doxorubicin by thermosensitive hydrogel and anti-cancer evaluation of peritumoral hydrogel injection combined with PD-L1 antibody. Biomater. Res. 2022, 26, 77. [Google Scholar] [CrossRef]
  63. Kim, K.H.; Kim, J.Y.; Kwak, J.H.; Pyo, S. Different anticancer effects of Saxifragifolin A on estrogen receptor-positive and estrogen receptor-negative breast cancer cells. Phytomedicine 2015, 22, 820–828. [Google Scholar] [CrossRef] [PubMed]
  64. Kim, B.-M.; Kim, D.-H.; Park, J.-H.; Na, H.-K.; Surh, Y.-J. Ginsenoside Rg 3 Induces Apoptosis of Human Breast Cancer (MDA-MB-231) Cells. J. Cancer Prev. 2013, 18, 177–185. [Google Scholar] [CrossRef] [PubMed]
  65. Kim, B.-M.; Kim, D.-H.; Park, J.-H.; Surh, Y.-J.; Na, H.-K. Ginsenoside Rg3 Inhibits Constitutive Activation of NF-κB Signaling in Human Breast Cancer (MDA-MB-231) Cells: ERK and Akt as Potential Upstream Targets. J. Cancer Prev. 2014, 19, 23–30. [Google Scholar] [CrossRef] [PubMed]
  66. Liu, S.; Huang, J.; Gao, F.; Yin, Z.; Zhang, R. Ginsenoside RG1 augments doxorubicin-induced apoptotic cell death in MDA-MB-231 breast cancer cell lines. J. Biochem. Mol. Toxicol. 2022, 36, e22945. [Google Scholar] [CrossRef]
  67. Gao, Y.; Chu, S.; Shao, Q.; Zhang, M.; Xia, C.; Wang, Y.; Li, Y.; Lou, Y.; Huang, H.; Chen, N. Antioxidant activities of ginsenoside Rg1 against cisplatin-induced hepatic injury through Nrf2 signaling pathway in mice. Free Radic. Res. 2017, 51, 1–13. [Google Scholar] [CrossRef]
  68. Liu, P.; Liu, Y.; Chen, L.; Fan, Z.; Luo, Y.; Cui, Y. Anemoside A3 Inhibits Macrophage M2-Like Polarization to Prevent Triple-Negative Breast Cancer Metastasis. Molecules 2023, 28, 1611. [Google Scholar] [CrossRef]
  69. Chan, R.Y.K.; Chen, W.F.; Dong, A.; Guo, D.; Wong, M.S. Estrogen-like activity of ginsenoside Rg1 derived from Panax notoginseng. J. Clin. Endocrinol. Metab. 2002, 87, 3691–3695. [Google Scholar] [CrossRef]
  70. Fu, R.; Zhang, L.; Li, Y.; Li, B.; Ming, Y.; Li, Z.; Xing, H.; Chen, J. Saikosaponin D inhibits autophagosome-lysosome fusion and induces autophagy-independent apoptosis in MDA-MB-231 breast cancer cells. Mol. Med. Rep. 2020, 22, 1026–1034. [Google Scholar] [CrossRef]
  71. Ma, H.; Yue, G.G.; Lee, J.K.-M.; Gao, S.; Yuen, K.-K.; Cheng, W.; Li, X.; Lau, C.B. Scutellarin, a flavonoid compound from Scutellaria barbata, suppresses growth of breast cancer stem cells in vitro and in tumor-bearing mice. Phytomedicine 2024, 128, 155418. [Google Scholar] [CrossRef] [PubMed]
  72. Foss, K.; Przybyłowicz, K.E.; Sawicki, T. Antioxidant Activity and Profile of Phenolic Compounds in Selected Herbal Plants. Plant Foods Hum. Nutr. 2022, 77, 383–389. [Google Scholar] [CrossRef] [PubMed]
  73. Shi, L.; Zhao, W.; Yang, Z.; Subbiah, V.; Suleria, H.A.R. Extraction and characterization of phenolic compounds and their potential antioxidant activities. Environ. Sci. Pollut. Res. 2022, 29, 81112–81129. [Google Scholar] [CrossRef]
  74. Singh, B.; Singh, J.P.; Kaur, A.; Singh, N. Phenolic composition, antioxidant potential and health benefits of citrus peel. Food Res. Int. 2020, 132, 109114. [Google Scholar] [CrossRef]
  75. Jung, K.; Everson, R.J.C.; Joshi, B.; Bulsara, P.A.; Upasani, R.; Clarke, M.J. Structure-function relationship of phenolic antioxidants in topical skin health products. Int. J. Cosmet. Sci. 2017, 39, 217–223. [Google Scholar] [CrossRef]
  76. Peng, F.; Xiong, L.; Peng, C. (-)-Sativan Inhibits Tumor Development and Regulates miR-200c/PD-L1 in Triple Negative Breast Cancer Cells. Front. Pharmacol. 2020, 11, 251. [Google Scholar] [CrossRef]
  77. Peng, F.; Tang, H.; Du, J.; Chen, J.; Peng, C. Isoliquiritigenin Suppresses EMT-Induced Metastasis in Triple-Negative Breast Cancer through miR-200c/C-JUN/β-Catenin. Am. J. Chin. Med. 2021, 49, 505–523. [Google Scholar] [CrossRef]
  78. Pai, J.T.; Chen, X.H.; Leu, Y.L.; Weng, M.S. Propolin G-Suppressed Epithelial-to-Mesenchymal Transition in Triple-Negative Breast Cancer Cells via Glycogen Synthase Kinase 3β-Mediated Snail and HDAC6-Regulated Vimentin Degradation. Int. J. Mol. Sci. 2022, 23, 1672. [Google Scholar] [CrossRef]
  79. Koh, S.Y.; Moon, J.Y.; Unno, T.; Cho, S.K. Baicalein suppresses stem cell-like characteristics in radio-and chemoresistant MDA-MB-231 human breast cancer cells through up-regulation of IFIT2. Nutrients 2019, 11, 624. [Google Scholar] [CrossRef]
  80. Ebrahim, H.Y.; Elsayed, H.E.; Mohyeldin, M.M.; Akl, M.R.; Bhattacharjee, J.; Egbert, S.; El Sayed, K.A. Norstictic Acid Inhibits Breast Cancer Cell Proliferation, Migration, Invasion, and In Vivo Invasive Growth Through Targeting C-Met. Phytother. Res. 2016, 30, 557–566. [Google Scholar] [CrossRef]
  81. Yu, S.; Wang, Z.; Su, Z.; Song, J.; Zhou, L.; Sun, Q.; Liu, S.; Li, S.; Li, Y.; Wang, M.; et al. Gigantol inhibits Wnt/β-catenin signaling and exhibits anticancer activity in breast cancer cells. BMC Complement. Med. Ther. 2018, 18, 59. [Google Scholar] [CrossRef] [PubMed]
  82. Zou, G.; Zhang, X.; Wang, L.; Li, X.; Xie, T.; Zhao, J.; Yan, J.; Wang, L.; Ye, H.; Jiao, S.; et al. Herb-sourced emodin inhibits angiogenesis of breast cancer by targeting VEGFA transcription. Theranostics 2020, 10, 6839–6853. [Google Scholar] [CrossRef] [PubMed]
  83. Abdellatef, A.A.; Fathy, M.; Mohammed, A.E.-S.I.; Bakr, M.S.A.; Ahmed, A.H.; Abbass, H.S.; El-Desoky, A.H.; Morita, H.; Nikaido, T.; Hayakawa, Y. Inhibition of cell-intrinsic NF-κB activity and metastatic abilities of breast cancer by aloe-emodin and emodic-acid isolated from Asphodelus microcarpus. J. Nat. Med. 2021, 75, 840–853. [Google Scholar] [CrossRef] [PubMed]
  84. Yu, Y.; Cai, W.; Pei, C.G.; Shao, Y. Rhamnazin, a novel inhibitor of VEGFR2 signaling with potent antiangiogenic activity and antitumor efficacy. Biochem. Biophys. Res. Commun. 2015, 458, 913–919. [Google Scholar] [CrossRef]
  85. Yeon Park, J.; Young Kim, H.; Shibamoto, T.; Su Jang, T.; Cheon Lee, S.; Suk Shim, J.; Hahm, D.-H.; Lee, H.-J.; Lee, S.; Sung Kang, K. Beneficial effects of a medicinal herb, Cirsium japonicum var. maackii, extract and its major component, cirsimaritin on breast cancer metastasis in MDA-MB-231 breast cancer cells. Bioorg. Med. Chem. Lett. 2017, 27, 3968–3973. [Google Scholar] [CrossRef]
  86. Shi, Y.; Wu, Y.; Li, F.; Zhang, Y.; Hua, C.; Yang, J.; Zheng, J.; Chen, L.; Wei, Z.; Yue, H.; et al. Identifying the anti-metastasis effect of Anhydroicaritin on breast cancer: Coupling network pharmacology with experimental validation. J. Ethnopharmacol. 2022, 293, 115326. [Google Scholar] [CrossRef]
  87. Sun, C.; Wang, T.; Wang, C.; Zhu, Z.; Wang, X.; Xu, J.; An, H. The Protective Effect of Kaempferol Against Ischemia/Reperfusion Injury Through Activating SIRT3 to Inhibit Oxidative Stress. Braz. J. Cardiovasc. Surg. 2022, 37, 335–342. [Google Scholar] [CrossRef]
  88. Maharjan, S.; Lee, M.G.; Kim, S.Y.; Lee, K.S.; Nam, K.S. Morin Sensitizes MDA-MB-231 Triple-Negative Breast Cancer Cells to Doxorubicin Cytotoxicity by Suppressing FOXM1 and Attenuating EGFR/STAT3 Signaling Pathways. Pharmaceuticals 2023, 16, 672. [Google Scholar] [CrossRef]
  89. Hua, F.; Xiao, Y.-Y.; Qu, X.-H.; Li, S.-S.; Zhang, K.; Zhou, C.; He, J.-L.; Zhu, Y.; Wan, Y.-Y.; Jiang, L.-P.; et al. Baicalein sensitizes triple negative breast cancer MDA-MB-231 cells to doxorubicin via autophagy-mediated down-regulation of CDK1. Mol. Cell. Biochem. 2023, 478, 1519–1531. [Google Scholar] [CrossRef]
  90. Liu, Q.; Li, J.; Pu, G.; Zhang, F.; Liu, H.; Zhang, Y. Co-delivery of baicalein and doxorubicin by hyaluronic acid decorated nanostructured lipid carriers for breast cancer therapy. Drug Deliv. 2016, 23, 1364–1368. [Google Scholar] [CrossRef]
  91. Pérez-Gutiérrez, L.; Ferrara, N. Biology and therapeutic targeting of vascular endothelial growth factor A. Nat. Rev. Mol. Cell Biol. 2023, 24, 816–834. [Google Scholar] [CrossRef] [PubMed]
  92. Takahashi, H.; Shibuya, M. The vascular endothelial growth factor (VEGF)/VEGF receptor system and its role under physiological and pathological conditions. Clin. Sci. 2005, 109, 227–241. [Google Scholar] [CrossRef] [PubMed]
  93. Du, Y.; Khan, M.; Fang, N.; Ma, F.; Du, H.; Tan, Z.; Wang, H.; Yin, S.; Wei, X. Berberine Attenuates Cell Motility via Inhibiting Inflammation-Mediated Lysyl Hydroxylase-2 and Glycolysis. Front. Pharmacol. 2022, 13, 856777. [Google Scholar] [CrossRef] [PubMed]
  94. Xing, Z.; Zhang, Y.; Zhang, X.; Yang, Y.; Ma, Y.; Pang, D. Fangchinoline induces g1 arrest in breast cancer cells through cell-cycle regulation. Phytother. Res. 2013, 27, 1790–1794. [Google Scholar] [CrossRef]
  95. Wang, C.-D.; Yuan, C.-F.; Bu, Y.-Q.; Wu, X.-M.; Wan, J.-Y.; Zhang, L.; Hu, N.; Liu, X.-J.; Zu, Y.; Liu, G.-L.; et al. Fangchinoline inhibits cell proliferation via Akt/GSK-3beta/cyclin D1 signaling and induces apoptosis in MDA-MB-231 breast cancer cells. Asian Pac. J. Cancer Prev. 2014, 15, 769–773. [Google Scholar] [CrossRef]
  96. Wei, S.; Zhang, Y.; Ma, X.; Yao, Y.; Zhou, Q.; Zhang, W.; Zhou, C.; Zhuang, J. MAT as a promising therapeutic strategy against triple-negative breast cancer via inhibiting PI3K/AKT pathway. Sci. Rep. 2023, 13, 12351. [Google Scholar] [CrossRef]
  97. Guo, Q.; Yu, Y.; Tang, W.; Zhou, S.; Lv, X. Matrine exerts an anti-tumor effect via regulating HN1 in triple breast cancer both in vitro and in vivo. Chem. Biol. Drug Des. 2023, 102, 1469–1477. [Google Scholar] [CrossRef]
  98. Guzmán, E.A.; Peterson, T.A.; Wright, A.E. The Marine Natural Compound Dragmacidin D Selectively Induces Apoptosis in Triple-Negative Breast Cancer Spheroids. Mar. Drugs 2023, 21, 642. [Google Scholar] [CrossRef]
  99. Peng, F.; Xiong, L.; Xie, X.; Tang, H.; Huang, R.; Peng, C. Isoliquiritigenin Derivative Regulates miR-374a/BAX Axis to Suppress Triple-Negative Breast Cancer Tumorigenesis and Development. Front. Pharmacol. 2020, 11, 378. [Google Scholar] [CrossRef]
  100. Maharjan, S.; Kwon, Y.S.; Lee, M.G.; Lee, K.S.; Nam, K.S. Cell cycle arrest-mediated cell death by morin in MDA-MB-231 triple-negative breast cancer cells. Pharmacol. Rep. 2021, 73, 1315–1327. [Google Scholar] [CrossRef]
  101. Jin, H.; Lee, W.S.; Eun, S.Y.; Jung, J.H.; Park, H.-S.; Kim, G.; Choi, Y.H.; Ryu, C.H.; Jung, J.M.; Hong, S.C.; et al. Morin, a flavonoid from Moraceae, suppresses growth and invasion of the highly metastatic breast cancer cell line MDA-MB-231 partly through suppression of the Akt pathway. Int. J. Oncol. 2014, 45, 1629–1637. [Google Scholar] [CrossRef] [PubMed]
  102. Yan, W.; Ma, X.; Zhao, X.; Zhang, S. Baicalein induces apoptosis and autophagy of breast cancer cells via inhibiting PI3K/AKT pathway in vivo and vitro. Drug Des. Dev. Ther. 2018, 12, 3961–3972. [Google Scholar] [CrossRef] [PubMed]
  103. Yao, P.; Yao, P.; Ku, X.; Yang, J. Harmine suppresses the malignant phenotypes and PI3K activity in breast cancer. Anticancer Drugs 2023, 34, 373–383. [Google Scholar] [CrossRef] [PubMed]
  104. Al-Sayed, E.; Ke, T.-Y.; Hwang, T.-L.; Chen, S.-R.; Korinek, M.; Chen, S.-L.; Cheng, Y.-B. Cytotoxic and Anti-Inflammatory Effects of Lignans and Diterpenes from Cupressus Macrocarpa. Bioorg. Med. Chem. Lett. 2020, 30, 127127. [Google Scholar] [CrossRef]
  105. Bommareddy, A.; Knapp, K.; Nemeth, A.; Steigerwalt, J.; Landis, T.; Vanwert, A.L.; Gorijavolu, H.P.; Dwivedi, C. Alpha-Santalol, a Component of Sandalwood Oil Inhibits Migration of Breast Cancer Cells by Targeting the β-Catenin Pathway. Anticancer Res. 2018, 38, 4475–4480. [Google Scholar] [CrossRef]
  106. Mbaveng, A.T.; Bitchagno, G.T.M.; Kuete, V.; Tane, P.; Efferth, T. Cytotoxicity of ungeremine towards multi-factorial drug resistant cancer cells and induction of apoptosis, ferroptosis, necroptosis and autophagy. Phytomedicine 2019, 60, 152832. [Google Scholar] [CrossRef]
  107. Zhang, Y.; Gao, X.; Liu, C.; Wang, M.; Zhang, R.; Sun, J.; Liu, Y. Design, synthesis and in vitro evaluation of fangchinoline derivatives as potential anticancer agents. Bioorg. Chem. 2020, 94, 103431. [Google Scholar] [CrossRef]
  108. He, K.; Gao, J.L. Protopine inhibits heterotypic cell adhesion in MDA-MB-231 cells through down-regulation of multi-adhesive factors. Afr. J. Tradit. Complement. Altern. Med. AJTCAM/Afr. Netw. Ethnomed. 2014, 11, 415–424. [Google Scholar] [CrossRef]
  109. Shaala, L.A.; Youssef, D.T.A.; Sulaiman, M.; Behery, F.A.; Foudah, A.I.; El Sayed, K.A. Subereamolline A as a potent breast cancer migration, invasion and proliferation inhibitor and bioactive dibrominated alkaloids from the red sea sponge Pseudoceratina arabica. Mar. Drugs 2012, 10, 2492–2508. [Google Scholar] [CrossRef]
  110. Zhang, Y.; Chen, J.; Mi, D.; Ling, J.; Li, H.; He, P.; Liu, N.; Chen, Q.; Chen, Y.; Huang, L. Discovery of YH677 as a cancer stemness inhibitor that suppresses triple-negative breast cancer growth and metastasis by regulating the TGFβ signaling pathway. Cancer Lett. 2023, 560, 216142. [Google Scholar] [CrossRef]
  111. Qi, M.; Liu, X.; Zhou, Y.; Wang, H.; Zhao, Y.; Ren, J.; Xiang, J. Berberine inhibits mda-mb-231 cells as an agonist of g protein-coupled estrogen receptor 1. Int. J. Mol. Sci. 2021, 22, 11466. [Google Scholar] [CrossRef] [PubMed]
  112. Mabeta, P.; Steenkamp, V. The VEGF/VEGFR Axis Revisited: Implications for Cancer Therapy. Int. J. Mol. Sci. 2022, 23, 15585. [Google Scholar] [CrossRef] [PubMed]
  113. Li, Y.L.; Zhao, H.; Ren, X.B. Relationship of VEGF/VEGFR with immune and cancer cells: Staggering or forward? Cancer Biol. Med. 2016, 13, 206–214. [Google Scholar] [CrossRef]
  114. Dakowicz, D.; Zajkowska, M.; Mroczko, B. Relationship between VEGF Family Members, Their Receptors and Cell Death in the Neoplastic Transformation of Colorectal Cancer. Int. J. Mol. Sci. 2022, 23, 3375. [Google Scholar] [CrossRef] [PubMed]
  115. Kang, Y.; Li, H.; Liu, Y.; Li, Z. Regulation of VEGF-A expression and VEGF-A-targeted therapy in malignant tumors. J. Cancer Res. Clin. Oncol. 2024, 150, 221. [Google Scholar] [CrossRef]
  116. Elebiyo, T.C.; Rotimi, D.; Evbuomwan, I.O.; Maimako, R.F.; Iyobhebhe, M.; Ojo, O.A.; Oluba, O.M.; Adeyemi, O.S. Reassessing vascular endothelial growth factor (VEGF) in anti-angiogenic cancer therapy. Cancer Treat. Res. Commun. 2022, 32, 100620. [Google Scholar] [CrossRef]
  117. Peng, W.; Zhang, H.; Yin, M.; Kong, D.; Kang, L.; Teng, X.; Wang, J.; Chu, Z.; Sun, Y.; Long, P.; et al. Combined Inhibition of PI3K and STAT3 signaling effectively inhibits bladder cancer growth. Oncogenesis 2024, 13, 29. [Google Scholar] [CrossRef]
  118. Glaviano, A.; Foo, A.S.C.; Lam, H.Y.; Yap, K.C.H.; Jacot, W.; Jones, R.H.; Eng, H.; Nair, M.G.; Makvan-di, P.; Geoerger, B.; et al. PI3K/AKT/mTOR signaling transduction pathway and targeted therapies in cancer. Mol. Cancer 2023, 22, 138. [Google Scholar] [CrossRef]
  119. Liu, R.; Chen, Y.; Liu, G.; Li, C.; Song, Y.; Cao, Z.; Li, W.; Hu, J.; Lu, C.; Liu, Y. PI3K/AKT pathway as a key link modulates the multidrug resistance of cancers. Cell Death Dis. 2020, 11, 797. [Google Scholar] [CrossRef]
  120. He, Y.; Sun, M.M.; Zhang, G.G.; Yang, J.; Chen, K.S.; Xu, W.W.; Li, B. Targeting PI3K/Akt signal transduction for cancer therapy. Signal Transduct. Target. Ther. 2021, 6, 425. [Google Scholar] [CrossRef]
  121. Davies, M.A. The role of the PI3K-AKT pathway in melanoma. Cancer J. 2012, 18, 142–147. [Google Scholar] [CrossRef] [PubMed]
  122. Liu, J.; Xiao, Q.; Xiao, J.; Niu, C.; Li, Y.; Zhang, X.; Zhou, Z.; Shu, G.; Yin, G. Wnt/β-catenin signalling: Function, biological mechanisms, and therapeutic opportunities. Signal Transduct. Target. Ther. 2022, 7, 3. [Google Scholar] [CrossRef] [PubMed]
  123. Yu, F.; Yu, C.; Li, F.; Zuo, Y.; Wang, Y.; Yao, L.; Wu, C.; Wang, C.; Ye, L. Wnt/β-catenin signaling in cancers and targeted therapies. Signal Transduct. Target. Ther. 2021, 6, 307. [Google Scholar] [CrossRef]
  124. Cargnello, M.; Roux, P.P. Activation and Function of the MAPKs and Their Substrates, the MAPK-Activated Protein Kinases. Microbiol. Mol. Biol. Rev. 2011, 75, 50–83. [Google Scholar] [CrossRef]
  125. Rovida, E.; Tusa, I. Targeting MAPK in Cancer 2.0. Int. J. Mol. Sci. 2022, 23, 5702. [Google Scholar] [CrossRef]
  126. Ramanan, M.; Sinha, S.; Sudarshan, K.; Aidhen, I.S.; Doble, M. Inhibition of the Enzymes in the Leukotriene and Prostaglandin Pathways in Inflammation by 3-Aryl Isocoumarins. Eur. J. Med. Chem. 2016, 124, 428–434. [Google Scholar] [CrossRef]
  127. Sudarshan, K.; Boda, A.K.; Dogra, S.; Bose, I.; Yadav, P.N.; Aidhen, I.S. Discovery of an Isocoumarin Analogue That Modulates Neuronal Functions via Neurotrophin Receptor TrkB. Bioorg. Med. Chem. Lett. 2019, 29, 585–590. [Google Scholar] [CrossRef]
  128. Han, H.S.; Vikas, P.; Costa, R.L.B.; Jahan, N.; Taye, A.; Stringer-Reasor, E.M. Early-Stage Triple-Negative Breast Cancer Journey: Beginning, End, and Everything in Between. Am. Soc. Clin. Oncol. Educ. Book 2023, 43, e390464. [Google Scholar] [CrossRef]
  129. Pusztai, L.; Yau, C.; Wolf, D.M.; Han, H.S.; Du, L.; Wallace, A.M.; String-Reasor, E.; Boughey, J.C.; Chien, A.J.; Elias, A.D.; et al. Durvalumab with Olaparib and Paclitaxel for High-Risk HER2-Negative Stage II/III Breast Cancer: Results from the Adaptively Randomized I-SPY2 Trial. Cancer Cell 2021, 39, 989–998.e5. [Google Scholar] [CrossRef]
  130. LaFargue, C.J.; Dal Molin, G.Z.; Sood, A.K.; Coleman, R.L. Exploring and Comparing Adverse Events between PARP Inhibitors. Lancet Oncol. 2019, 20, e15–e28. [Google Scholar] [CrossRef]
  131. Jin, Y.; Huang, Z.; Li, L.; Yang, Y.; Wang, C.; Wang, Z.; Ji, L. Quercetin Attenuates Toosendanin-Induced Hepatotoxicity through Inducing the Nrf2/GCL/GSH Antioxidant Signaling Pathway. Acta Pharmacol. Sin. 2019, 40, 75–85. [Google Scholar] [CrossRef]
  132. Lu, X.; Ji, C.; Tong, W.; Lian, X.; Wu, Y.; Fan, X.; Gao, Y. Integrated Analysis of microRNA and mRNA Expression Profiles Highlights the Complex and Dynamic Behavior of Toosendanin-Induced Liver Injury in Mice. Sci. Rep. 2016, 6, 34225. [Google Scholar] [CrossRef]
  133. Thant, M.T.; Bhummaphan, N.; Wuttiin, J.; Puttipanyalears, C.; Chaichompoo, W.; Rojsitthisak, P.; Punpreuk, Y.; Böttcher, C.; Likhitwitayawuid, K.; Sritularak, B. New Phenolic Glycosides from Coelogyne fuscescens Lindl. Var. brunnea and Their Cytotoxicity against Human Breast Cancer Cells. ACS Omega 2024, 9, 7679–7691. [Google Scholar] [CrossRef] [PubMed]
  134. Chakravarti, B.; Maurya, R.; Siddiqui, J.A.; Bid, H.K.; Rajendran, S.M.; Yadav, P.P.; Konwar, R. In Vitro Anti-Breast Cancer Activity of Ethanolic Extract of Wrightia Tomentosa: Role of pro-Apoptotic Effects of Oleanolic Acid and Urosolic Acid. J. Ethnopharmacol. 2012, 142, 72–79. [Google Scholar] [CrossRef]
  135. Chen, D.-L.; Ma, G.-X.; Yang, E.-L.; Yang, Y.; Wang, C.-H.; Sun, Z.-C.; Liang, H.-Q.; Xu, X.-D.; Wei, J.-H. Cadinane-Type Sesquiterpenoid Dimeric Diastereomers Hibisceusones A-C from Infected Stems of Hibiscus Tiliaceus with Cytotoxic Activity against Triple-Negative Breast Cancer Cells. Bioorganic Chem. 2022, 127, 105982. [Google Scholar] [CrossRef] [PubMed]
  136. Cheng, L.; Xia, T.-S.; Shi, L.; Xu, L.; Chen, W.; Zhu, Y.; Ding, Q. D Rhamnose β-Hederin Inhibits Migration and Invasion of Human Breast Cancer Cell Line MDA-MB-231. Biochem. Biophys. Res. 2018, 495, 775–780. [Google Scholar] [CrossRef] [PubMed]
  137. Chun, J.; Li, R.-J.; Cheng, M.-S.; Kim, Y.S. Alantolactone Selectively Suppresses STAT3 Activation and Exhibits Potent Anticancer Activity in MDA-MB-231 Cells. Cancer Lett. 2015, 357, 393–403. [Google Scholar] [CrossRef]
  138. Chun, J.; Song, K.; Kim, Y.S. Sesquiterpene Lactones-enriched Fraction of Inula helenium L. Induces Apoptosis through Inhibition of Signal Transducers and Activators of Transcription 3 Signaling Pathway in MDA-MB-231 Breast Cancer Cells. Phytother. Res. 2018, 32, 2501–2509. [Google Scholar] [CrossRef]
  139. Güney Eskiler, G.; Deveci Özkan, A.; Kaleli, S.; Bilir, C. Inhibition of TLR4/TRIF/IRF3 Signaling Pathway by Curcumin in Breast Cancer Cells. J. Pharm. Pharm. Sci. 2019, 22, 281–291. [Google Scholar] [CrossRef]
  140. Dawood, M.; Ooko, E.; Efferth, T. Collateral Sensitivity of Parthenolide via NF-κB and HIF-α Inhibition and Epigenetic Changes in Drug-Resistant Cancer Cell Lines. Front. Pharmacol. 2019, 10, 542. [Google Scholar] [CrossRef]
  141. Gou, L.; Yue, G.G.-L.; Lee, J.K.-M.; Puno, P.T.; Lau, C.B.-S. Natural Product Eriocalyxin B Suppressed Triple Negative Breast Cancer Metastasis Both in Vitro and in Vivo. Biochem. Pharmacol. 2023, 210, 115491. [Google Scholar] [CrossRef] [PubMed]
  142. Jang, H.; Ko, H.; Song, K.; Kim, Y. A Sesquiterpenoid from Farfarae Flos Induces Apoptosis of MDA-MB-231 Human Breast Cancer Cells through Inhibition of JAK–STAT3 Signaling. Biomolecules 2019, 9, 278. [Google Scholar] [CrossRef] [PubMed]
  143. Liu, L.-Y.; Yan, Z.; Kang, J.; Chen, R.-Y.; Yu, D.-Q. Three New Triterpenoids from Ganoderma theaecolum. J. Asian Nat. Prod. Res. 2017, 19, 847–853. [Google Scholar] [CrossRef]
  144. Park, J.-H.; Yoon, J.; Park, B. Pomolic Acid Suppresses HIF1α/VEGF-Mediated Angiogenesis by Targeting P38-MAPK and mTOR Signaling Cascades. Phytomedicine 2016, 23, 1716–1726. [Google Scholar] [CrossRef]
  145. Qiao, A.; Wang, Y.; Xiang, L.; Wang, C.; He, X. A Novel Triterpenoid Isolated from Apple Functions as an Anti-Mammary Tumor Agent via a Mitochondrial and Caspase-Independent Apoptosis Pathway. J. Agric. Food Chem. 2015, 63, 185–191. [Google Scholar] [CrossRef]
  146. Sathya, S.; Sudhagar, S.; Vidhya Priya, M.; Bharathi Raja, R.; Muthusamy, V.S.; Niranjali Devaraj, S.; Lakshmi, B.S. 3β-Hydroxylup-20(29)-Ene-27,28-Dioic Acid Dimethyl Ester, a Novel Natural Product from Plumbago Zeylanica Inhibits the Proliferation and Migration of MDA-MB-231 Cells. Chem. Biol. Interact. 2010, 188, 412–420. [Google Scholar] [CrossRef]
  147. Shin, M.; Lee, B.-M.; Kim, O.; Tran, H.N.K.; Lee, S.; Hwangbo, C.; Min, B.-S.; Lee, J.-H. Triterpenoids from Ziziphus jujuba Induce Apoptotic Cell Death in Human Cancer Cells through Mitochondrial Reactive Oxygen Species Production. Food Funct. 2018, 9, 3895–3905. [Google Scholar] [CrossRef]
  148. Huang, W.-C.; Yen, J.-H.; Sung, Y.-W.; Tung, S.-L.; Chen, P.-M.; Chu, P.-Y.; Shih, Y.-C.; Chi, H.-C.; Huang, Y.-C.; Huang, S.-J.; et al. Novel Function of THEMIS2 in the Enhancement of Cancer Stemness and Chemoresistance by Releasing PTP1B from MET. Oncogene 2022, 41, 997–1010. [Google Scholar] [CrossRef]
  149. Yen, J.-H.; Huang, W.-C.; Lin, S.-C.; Huang, Y.-W.; Chio, W.-T.; Tsay, G.J.; Hung, M.-C.; Huang, S.-T. Metabolic Remodeling in Tumor-Associated Macrophages Contributing to Antitumor Activity of Cryptotanshinone by Regulating TRAF6-ASK1 Axis. Mol. Ther. Oncolytics 2022, 26, 158–174. [Google Scholar] [CrossRef]
  150. Xia, Y.; Zhang, Y.; Chen, L.; Min, L.; Huang, D.; Zhang, Y.; Li, C.; Li, Z. Suppression of Migration and Invasion by Taraxerol in the Triple-Negative Breast Cancer Cell Line MDA-MB-231 via the ERK/Slug Axis. PLoS ONE 2023, 18, e0291693. [Google Scholar] [CrossRef]
  151. Kashyap, A.; Umar, S.; Dev J R, A.; Prasad, C. Dihydrotanshinone-I Modulates Epithelial Mesenchymal Transition (EMT) Thereby Impairing Migration and Clonogenicity of Triple Negative Breast Cancer Cells. Asian Pac. J. Cancer Prev. 2021, 22, 2177–2184. [Google Scholar] [CrossRef] [PubMed]
  152. Aumsuwan, P.; Khan, S.I.; Khan, I.A.; Ali, Z.; Avula, B.; Walker, L.A.; Shariat-Madar, Z.; Helferich, W.G.; Katzenellenbogen, B.S.; Dasmahapatra, A.K. The Anticancer Potential of Steroidal Saponin, Dioscin, Isolated from Wild Yam (Dioscorea villosa) Root Extract in Invasive Human Breast Cancer Cell Line MDA-MB-231 In Vitro. Arch. Biochem. Biophys. 2016, 591, 98–110. [Google Scholar] [CrossRef] [PubMed]
  153. Christodoulou, P.; Boutsikos, P.; Neophytou, C.M.; Kyriakou, T.-C.; Christodoulou, M.-I.; Papageorgis, P.; Stephanou, A.; Patrikios, I. Amygdalin as a Chemoprotective Agent in Co-Treatment with Cisplatin. Front. Pharmacol. 2022, 13, 1013692. [Google Scholar] [CrossRef] [PubMed]
  154. He, Z.; Chen, H.; Li, G.; Zhu, H.; Gao, Y.; Zhang, L.; Sun, J. Diosgenin Inhibits the Migration of Human Breast Cancer MDA-MB-231 Cells by Suppressing Vav2 Activity. Phytomedicine 2014, 21, 871–876. [Google Scholar] [CrossRef]
  155. Ho, D.V.; Hoang, H.N.T.; Vo, H.Q.; Nguyen, K.V.; Pham, T.V.; Le, A.T.; Van Phan, K.; Nguyen, H.M.; Morita, H.; Nguyen, H.T. Three New Steroidal Saponins from Aspidistra letreae Plants and Their Cytotoxic Activities. J. Nat. Med. 2020, 74, 591–598. [Google Scholar] [CrossRef]
  156. Kwak, J.H.; Park, J.Y.; Lee, D.; Kwak, J.Y.; Park, E.H.; Kim, K.H.; Park, H.-J.; Kim, H.Y.; Jang, H.J.; Ham, J.; et al. Inhibitory Effects of Ginseng sapogenins on the Proliferation of Triple Negative Breast Cancer MDA-MB-231 Cells. Bioorg. Med. Chem. Lett. 2014, 24, 5409–5412. [Google Scholar] [CrossRef]
  157. Pederson, P.J.; Cai, S.; Carver, C.; Powell, D.R.; Risinger, A.L.; Grkovic, T.; O’Keefe, B.R.; Mooberry, S.L.; Cichewicz, R.H. Triple-Negative Breast Cancer Cells Exhibit Differential Sensitivity to Cardenolides from Calotropis gigantea. J. Nat. Prod. 2020, 83, 2269–2280. [Google Scholar] [CrossRef]
  158. Xu, H.; Li, Y.; Han, B.; Li, Z.; Wang, B.; Jiang, P.; Zhang, J.; Ma, W.; Zhou, D.; Li, X.; et al. Anti-Breast-Cancer Activity Exerted by β-Sitosterol-d-Glucoside from Sweet Potato via Upregulation of MicroRNA-10a and via the PI3K–Akt Signaling Pathway. J. Agric. Food Chem. 2018, 66, 9704–9718. [Google Scholar] [CrossRef]
  159. Pei, S.; Yang, X.; Wang, H.; Zhang, H.; Zhou, B.; Zhang, D.; Lin, D. Plantamajoside, a Potential Anti-Tumor Herbal Medicine Inhibits Breast Cancer Growth and Pulmonary Metastasis by Decreasing the Activity of Matrix Metalloproteinase-9 and -2. BMC Cancer 2015, 15, 965. [Google Scholar] [CrossRef]
  160. Tan, H.; Li, M.; Han, L.; Zhao, Y.; Zhang, X. Gypensapogenin I Suppresses Cell Proliferation in Triple-Negative Breast Cancer Via Triggering the Closure of AKT/GSK3β/β-Catenin and Notch-1 Signaling Pathways. J. Agric. Food Chem. 2022, 70, 5438–5449. [Google Scholar] [CrossRef]
  161. Zhou, Y.; Fu, X.; Guan, Y.; Gong, M.; He, K.; Huang, B. 1,3-Dicaffeoylquinic Acid Targeting 14-3-3 Tau Suppresses Human Breast Cancer Cell Proliferation and Metastasis through IL6/JAK2/PI3K Pathway. Bioorg. Med. Chem. Lett. 2020, 172, 113752. [Google Scholar] [CrossRef] [PubMed]
  162. Yang, J.; Su, J.-C.; Lei, X.-P.; Huang, X.-J.; Zhang, D.-M.; Ye, W.-C.; Wang, Y. Acylphloroglucinol Derivatives from the Leaves of Syzygium samarangense and Their Cytotoxic Activities. Fitoterapia 2018, 129, 1–6. [Google Scholar] [CrossRef] [PubMed]
  163. Sunilkumar, D.; Drishya, G.; Chandrasekharan, A.; Shaji, S.K.; Bose, C.; Jossart, J.; Perry, J.J.P.; Mishra, N.; Kumar, G.B.; Nair, B.G. Oxyresveratrol Drives Caspase-Independent Apoptosis-like Cell Death in MDA-MB-231 Breast Cancer Cells through the Induction of ROS. Biochem. Pharmacol. 2020, 173, 113724. [Google Scholar] [CrossRef]
  164. Sun, L.-P.; Chen, A.-L.; Hung, H.-C.; Chien, Y.-H.; Huang, J.-S.; Huang, C.-Y.; Chen, Y.-W.; Chen, C.-N. Chrysin: A Histone Deacetylase 8 Inhibitor with Anticancer Activity and a Suitable Candidate for the Standardization of Chinese Propolis. J. Agric. Food Chem. 2012, 60, 11748–11758. [Google Scholar] [CrossRef]
  165. Nguyen, P.H.; Sharma, G.; Dao, T.T.; Uddin, M.N.; Kang, K.W.; Ndinteh, D.T.; Mbafor, J.T.; Oh, W.K. New Prenylated Isoflavonoids as Protein Tyrosine Phosphatase 1B (PTP1B) Inhibitors from Erythrina addisoniae. Bioorg. Med. Chem. 2012, 20, 6459–6464. [Google Scholar] [CrossRef]
  166. Liu, F.; Lu, W.-J.; Li, N.-P.; Liu, J.-W.; He, J.; Ye, W.-C.; Wang, L. Four New Cinnamoyl-Phloroglucinols from the Leaves of Xanthostemon chrysanthus. Fitoterapia 2018, 128, 93–96. [Google Scholar] [CrossRef]
  167. Lee, M.-F.; Pan, M.-H.; Chiou, Y.-S.; Cheng, A.-C.; Huang, H. Resveratrol Modulates MED28 (Magicin/EG-1) Expression and Inhibits Epidermal Growth Factor (EGF)-Induced Migration in MDA-MB-231 Human Breast Cancer Cells. J. Agric. Food Chem. 2011, 59, 11853–11861. [Google Scholar] [CrossRef]
  168. Lee, S.-J.; Hong, S.; Yoo, S.-H.; Kim, G.-W. Cyanidin-3-O-Sambubioside from Acanthopanax Sessiliflorus Fruit Inhibits Metastasis by Downregulating MMP-9 in Breast Cancer Cells MDA-MB-231. Planta Med. 2013, 79, 1636–1640. [Google Scholar] [CrossRef]
  169. Gao, Y.; Tollefsbol, T.O. Combinational Proanthocyanidins and Resveratrol Synergistically Inhibit Human Breast Cancer Cells and Impact Epigenetic–Mediating Machinery. Int. J. Mol. Sci. 2018, 19, 2204. [Google Scholar] [CrossRef]
  170. El Gaafary, M.; Saber, F.R.; Mahrous, E.A.; Ashour, R.M.; Okba, M.M.; Jin, L.; Lang, S.J.; Schmiech, M.; Simmet, T.; Syrovets, T. The Phloroglucinol Calcitrinone A, a Novel Mitochondria-Targeting Agent, Induces Cell Death in Breast Cancer Cells. Food Chem. Toxicol. 2022, 162, 112896. [Google Scholar] [CrossRef]
  171. Bajpai, V.K.; Alam, M.B.; Quan, K.T.; Choi, H.-J.; An, H.; Ju, M.-K.; Lee, S.-H.; Huh, Y.S.; Han, Y.-K.; Na, M. Cytotoxic Properties of the Anthraquinone Derivatives Isolated from the Roots of Rubia philippinensis. BMC Complement. Altern. Med. 2018, 18, 200. [Google Scholar] [CrossRef] [PubMed]
  172. Arunasree, K.M. Anti-Proliferative Effects of Carvacrol on a Human Metastatic Breast Cancer Cell Line, MDA-MB 231. Phytomedicine 2010, 17, 581–588. [Google Scholar] [CrossRef]
  173. Bello, I.; Smimmo, M.; d’Emmanuele Di Villa Bianca, R.; Bucci, M.; Cirino, G.; Panza, E.; Brancaleone, V. Erucin, an H2S-Releasing Isothiocyanate, Exerts Anticancer Effects in Human Triple-Negative Breast Cancer Cells Triggering Autophagy-Dependent Apoptotic Cell Death. Int. J. Mol. Sci. 2023, 24, 6764. [Google Scholar] [CrossRef] [PubMed]
  174. Chang, F.-R.; Yen, C.-T.; El-Shazly, M.; Yu, C.-Y.; Yen, M.-H.; Cheng, Y.-B.; Chen, S.-L.; Wu, Y.-C. Spirostanoids with 1,4-Dien-3-One or 3β,7α-Diol-5,6-Ene Moieties from Solanum violaceum. Bioorg. Med. Chem. Lett. 2013, 23, 2738–2742. [Google Scholar] [CrossRef] [PubMed]
  175. Li, X.; Wu, Q.; Xie, Y.; Ding, Y.; Du, W.W.; Sdiri, M.; Yang, B.B. Ergosterol Purified from Medicinal Mushroom Amauroderma rude Inhibits Cancer Growth in vitro and in vivo by up-Regulating Multiple Tumor Suppressors. Oncotarget 2015, 6, 17832–17846. [Google Scholar] [CrossRef]
  176. Yu, T.-J.; Shiau, J.-P.; Tang, J.-Y.; Farooqi, A.A.; Cheng, Y.-B.; Hou, M.-F.; Yen, C.-H.; Chang, H.-W. Physapruin A Exerts Endoplasmic Reticulum Stress to Trigger Breast Cancer Cell Apoptosis via Oxidative Stress. Int. J. Mol. Sci. 2023, 24, 8853. [Google Scholar] [CrossRef]
  177. Wang, H.-C.; Hu, H.-H.; Chang, F.-R.; Tsai, J.-Y.; Kuo, C.-Y.; Wu, Y.-C.; Wu, C.-C. Different Effects of 4β-Hydroxywithanolide E and Withaferin A, Two Withanolides from Solanaceae Plants, on the Akt Signaling Pathway in Human Breast Cancer Cells. Phytomedicine 2019, 53, 213–222. [Google Scholar] [CrossRef]
  178. Xiang, K.; Li, C.; Li, M.-X.; Song, Z.-R.; Ma, X.-X.; Sun, D.-J.; Li, H.; Chen, L.-X. Withanolides Isolated from Tubocapsicum Anomalum and Their Antiproliferative Activity. Bioorganic Chem. 2021, 110, 104809. [Google Scholar] [CrossRef]
  179. Ghosh, K.; De, S.; Das, S.; Mukherjee, S.; Sengupta Bandyopadhyay, S. Withaferin A Induces ROS-Mediated Paraptosis in Human Breast Cancer Cell-Lines MCF-7 and MDA-MB-231. PLoS ONE 2016, 11, e0168488. [Google Scholar] [CrossRef]
  180. Grossmann, M.E.; Mizuno, N.K.; Schuster, T.; Cleary, M.P. Punicic Acid Is an Omega-5 Fatty Acid Capable of Inhibiting Breast Cancer Proliferation. Int. J. Mol. Sci. 2010, 36, 421–426. [Google Scholar]
  181. Guzmán, E.A.; Pitts, T.P.; Winder, P.L.; Wright, A.E. The Marine Natural Product Furospinulosin 1 Induces Apoptosis in MDA-MB-231 Triple Negative Breast Cancer Cell Spheroids, But Not in Cells Grown Traditionally with Longer Treatment. Mar Drugs. 2021, 19, 249. [Google Scholar] [CrossRef] [PubMed]
  182. Khaw-on, P.; Pompimon, W.; Banjerdpongchai, R. Goniothalamin Induces Necroptosis and Anoikis in Human Invasive Breast Cancer MDA-MB-231 Cells. Int. J. Mol. Sci. 2019, 20, 3953. [Google Scholar] [CrossRef] [PubMed]
  183. Mi, C.; Ma, J.; Wang, K.S.; Wang, Z.; Li, M.Y.; Li, J.B.; Li, X.; Piao, L.X.; Xu, G.H.; Jin, X. Amorfrutin A Inhibits TNF-α Induced JAK/STAT Signaling, Cell Survival and Proliferation of Human Cancer Cells. Int. J. Mol. Sci. 2017, 39, 338–347. [Google Scholar] [CrossRef] [PubMed]
Figure 1. The regulation of terpenoids in TNBC.
Figure 1. The regulation of terpenoids in TNBC.
Molecules 30 01201 g001
Figure 2. The regulation of glycosides in TNBC.
Figure 2. The regulation of glycosides in TNBC.
Molecules 30 01201 g002
Figure 3. The regulation of phenolics in TNBC.
Figure 3. The regulation of phenolics in TNBC.
Molecules 30 01201 g003
Figure 4. The regulation of alkaloids in TNBC.
Figure 4. The regulation of alkaloids in TNBC.
Molecules 30 01201 g004
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

Li, Q.; Ye, Z.; Wang, G.; Chen, Y.; Deng, J.; Wang, D.; Wang, Y. Natural Products as Novel Therapeutic Agents for Triple-Negative Breast Cancer: Current Evidence, Mechanisms, Challenges, and Opportunities. Molecules 2025, 30, 1201. https://doi.org/10.3390/molecules30061201

AMA Style

Li Q, Ye Z, Wang G, Chen Y, Deng J, Wang D, Wang Y. Natural Products as Novel Therapeutic Agents for Triple-Negative Breast Cancer: Current Evidence, Mechanisms, Challenges, and Opportunities. Molecules. 2025; 30(6):1201. https://doi.org/10.3390/molecules30061201

Chicago/Turabian Style

Li, Qingzhou, Zhen Ye, Guilin Wang, Yuhui Chen, Jinghong Deng, Dong Wang, and Yumei Wang. 2025. "Natural Products as Novel Therapeutic Agents for Triple-Negative Breast Cancer: Current Evidence, Mechanisms, Challenges, and Opportunities" Molecules 30, no. 6: 1201. https://doi.org/10.3390/molecules30061201

APA Style

Li, Q., Ye, Z., Wang, G., Chen, Y., Deng, J., Wang, D., & Wang, Y. (2025). Natural Products as Novel Therapeutic Agents for Triple-Negative Breast Cancer: Current Evidence, Mechanisms, Challenges, and Opportunities. Molecules, 30(6), 1201. https://doi.org/10.3390/molecules30061201

Article Metrics

Back to TopTop