Next Article in Journal
The Legacy of COVID-19 in Breast Milk: The Association of Elevated Anti-Inflammatory and Antimicrobial Proteins with Vaccination or Infection
Next Article in Special Issue
A Comparative Study on the Mycelium and Fruiting Body of Meripilus giganteus: Chemical Composition and Biological Activity
Previous Article in Journal
Antineoplastic Activity of Methyl rosmarinate in Glioblastoma Cells
Previous Article in Special Issue
Prevention of Muscle Atrophy by Low-Molecular-Weight Fraction from Hirsutella sinensis Mycelium
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

The Therapeutic Potential of Baicalin and Baicalein in Breast Cancer: A Systematic Review of Mechanisms and Efficacy

Department of Chemistry, Institute of Food Sciences, Warsaw University of Life Sciences, Nowoursynowska 159c, 02-776 Warsaw, Poland
*
Author to whom correspondence should be addressed.
Curr. Issues Mol. Biol. 2025, 47(3), 181; https://doi.org/10.3390/cimb47030181
Submission received: 11 February 2025 / Revised: 5 March 2025 / Accepted: 8 March 2025 / Published: 11 March 2025

Abstract

:
Cancer remains a leading cause of death globally, with breast cancer being the most commonly diagnosed cancer in women. This systematic review focuses on the therapeutic potential of baicalin and baicalein, two bioactive flavonoids derived from Scutellaria baicalensis, in breast cancer treatment. These compounds exhibit anticancer properties through mechanisms such as apoptosis induction, cell cycle arrest, and inhibition of metastasis. Baicalin and baicalein modulate key signaling pathways, including NF-κB, PI3K/AKT/mTOR, and Wnt/β-catenin, and have shown efficacy in both in vitro and in vivo models. Their synergy with chemotherapy agents and incorporation into nanotechnology-based delivery systems highlight opportunities to enhance therapeutic outcomes. However, current evidence is predominantly preclinical, with limited clinical trials to validate their safety and efficacy in humans. Challenges such as poor bioavailability and rapid metabolism also underscore the need for advanced formulation strategies. This review synthesizes current evidence on the molecular mechanisms, therapeutic efficacy, and potential applications of baicalin and baicalein in breast cancer research.

1. Introduction

Cancer is currently one of the most significant public health problems worldwide and ranks high among causes of death. According to the 2018 GLOBOCAN report, which examined global cancer incidence (the number of new cases) and death rates across 185 countries for 36 types of cancer, it was estimated that there were approximately 18.1 million new cancer cases and 9.6 million cancer-related deaths globally [1]. Lung cancer, the most prevalent type, accounted for 11.6% of total cases and 18.4% of cancer deaths. When examined in terms of death, the most striking rates were colorectal cancer (9.2%), followed by stomach cancer (8.2%) and liver cancer (8.2%). Lung cancer remains the most common and deadly type among men, while breast cancer is the most frequently diagnosed in women. Furthermore, in developed countries, the most common cancers are lung, breast, and prostate cancers. In contrast, in low-income countries, there is a higher prevalence of infection-related cancers such as liver and cervical cancers [1].
According to the 2020 GLOBOCAN data, there were 19.3 million new cancer cases and 10 million cancer-related deaths worldwide. In 2020, for the first time, breast cancer surpassed lung cancer to become the most commonly diagnosed type globally [2]. The most recent data from the 2022 GLOBOCAN report indicate that lung cancer remains the most frequently diagnosed and deadliest type of cancer worldwide, with 2.48 million cases constituting 12.4% of all cancers [3]. In women, breast cancer is the most commonly diagnosed type, with an increasing trend of approximately 2.3 million new cases annually [4]. Breast cancer, with 25% of all female cancers, represents 15% of female cancer deaths [3]. Risk factors for breast cancer include age, physical inactivity, smoking and alcohol consumption, and obesity.
Breast cancer types are classified based on hormone receptors and human epidermal growth factor receptor 2 (HER2+). Estrogen receptor-positive breast cancer is the most frequently occurring subtype [4]. Endocrine therapies help to reduce the effects of estrogen that promote tumor growth for Hormone Receptor Positive (HR+) and ERBB2-Negative types. The treatment duration typically depends on regular oral medication intake over five to ten years. In cases where hormone therapy is not enough, chemotherapy may also be administered to patients exhibiting high resistance to treatment. For ERBB2-Positive (HER2/ERBB2+) cancers, treatments targeting HER2 protein (such as Trastuzumab and Pertuzumab) are combined with chemotherapy. Triple-negative Breast Cancer (TNBC) is treated with chemotherapy as well. Various new therapeutic approaches are being developed for breast cancer treatment, including PD-1 inhibitors like Pembrolizumab and Atezolizumab as alternative treatment options [5]. In HR+ breast cancer, CDK4/6 inhibitors help regulate the cell cycle and slow down disease progression when combined with chemotherapy [6]. In hormone receptor-positive breast cancers, Tamoxifen and Aromatase inhibitors support treatment by preventing estrogen from promoting tumor growth [6]. In early-stage breast cancer cases, various treatment methods such as Breast-Conserving Therapy (BCT) and Intraoperative Radiotherapy (IORT) are also observed [7]. While advances in targeted therapies and immunotherapies have improved survival rates, significant challenges persist. For instance, the initial chemotherapy response is about 40% in patients with triple-negative breast cancer. Still, at relapse, most have metastatic disease, primarily in the brain, bones, and lungs, resulting in a poor 5-year survival rate of roughly 12% [8]. Endocrine therapies often fail in metastatic hormone receptor-positive (HR+) cases due to acquired resistance [9]. Additionally, severe side effects—such as cardiotoxicity from trastuzumab [10] and hematologic complications from chemotherapy [11] limit treatment adherence and efficacy. These limitations underscore the urgent need for safer, multitargeted agents to circumvent resistance mechanisms and minimize adverse effects.
Natural polyphenols, such as flavonoids, have gained attention for their anticancer properties. Polyphenols can support cancer treatment by reducing cell proliferation, promoting apoptosis, suppressing inflammatory responses, and preventing tumor angiogenesis. Studies on polyphenols’ effects on specific types of cancers, such as melanoma, colorectal cancer, and lung cancer, indicate that they can avoid metastasis in melanoma and lung cancers while exhibiting synergistic effects when combined with chemotherapy in colorectal cancer treatments [12].
Stilbenes, like resveratrol, can inhibit tumor cell growth and reduce inflammation [13]. Phenolic acids exhibit antioxidant activity that can prevent DNA damage. Curcumin, a phenolic compound with anti-inflammatory and antioxidant properties, has shown positive results in colon cancer treatment [14]. Flavonoids can regulate cell signaling pathways and enhance tumor cell sensitivity while exhibiting effects similar to estrogen in breast cancers to inhibit the proliferation of cancer cells [15]. Resveratrol also prevents DNA damage and supports tumor cell death [16]. As seen in many examples, polyphenols were used as an adjuvant or a natural treatment method for cancer due to their antioxidant nature.
Baicalin and baicalein (Figure 1) are two prominent bioactive flavonoids widely recognized for their diverse pharmacological properties, including anti-inflammatory, antioxidant, antiviral, and anticancer activities [17]. These compounds are primarily derived from the roots of S. baicalensis, a plant extensively used in traditional Chinese medicine for centuries to treat conditions such as fever, cough, and infections [18]. While S. baicalensis is the most well-known source of baicalin and baicalein, these flavonoids can also be found in other plants but in smaller quantities. For example, other Scutellaria species, Phyllanthus amarus, Vernonia amygdalina, Tuber aestivum, Xanthium spinosum, or Centaurea mersinensis [19,20,21,22,23,24].
Baicalin has poor oral bioavailability due to low water solubility and limited membrane permeability. In contrast, its aglycone metabolite, baicalein, formed by removing glucuronic acid, exhibits superior lipophilicity and permeability for efficient absorption. Baicalin is hydrolyzed to baicalein during intestinal absorption by gut microbiota and enzymes. Conversely, UDP-glucuronosyltransferase enzymes in enterocytes and the liver can reconvert baicalein back to baicalin. This metabolic interplay enhances the compound’s pharmacodynamic profile: while baicalin is poorly absorbed, conversion to baicalein facilitates systemic uptake, with re-glucuronidation potentially prolonging therapeutic effects [25]. Notably, baicalein has shown dual antioxidant/pro-oxidant activity, inducing oxidative stress in cancer cells without harming healthy tissues [26]. Mechanistically, they selectively target pathways dysregulated in breast cancer, such as PI3K/AKT/mTOR and STAT3, while sparing normal cells, a critical advantage over conventional chemotherapies [27,28].
This systematic review aims to comprehensively evaluate the roles of baicalin and baicalein in breast cancer by synthesizing current scientific evidence on their mechanisms of action, therapeutic efficacy, and potential as adjunctive or standalone treatments. Specifically, it seeks to elucidate their molecular mechanisms in modulating key pathways such as apoptosis, autophagy, oxidative stress, and inflammation and assess their anticancer effects, including tumor growth inhibition and metastasis suppression. This review aims to highlight the potential of baicalin and baicalein as promising therapeutic agents in breast cancer treatment by providing a critical and up-to-date analysis.

2. Materials and Methods

The first methodological tool used in the current study was bibliographic analysis. Data were collected from the Scopus database, which was accessed on 1 February 2025. The search query used was TITLE-ABS-KEY (“baicalin” OR “baicalein”). CSV file was exported from the Scopus database and imported into a Microsoft Excel 2021 (Microsoft Corporation, WA, Redmond, USA) spreadsheet for additional analysis. Initially, an examination of document types, languages, journals, countries, and the number of publications over the years was conducted.
Following this, data from all selected publications (totaling 10,278) were analyzed using VOSviewer version 1.6.20 (Centre for Science and Technology Studies, Leiden University, Leiden, The Netherlands). The author’s keywords were analyzed using co-occurrence analysis, with the minimum number of occurrences set to 75. This process yielded a total of 39 distinct keywords.
A systematic review was conducted following the Preferred Reporting Items for Systematic Reviews and Meta-Analyses (PRISMA) guidelines [29]. The completed PRISMA checklist is provided as Table S1 in the Supplementary Materials. A systematic literature search was performed in the Scopus database, but the following keyword combination was employed to ensure a comprehensive retrieval of relevant studies: (TITLE-ABS-KEY (“baicalin” OR “baicalein”) AND TITLE-ABS-KEY (breast) AND TITLE-ABS-KEY (“cancer” OR “tumor” OR “carcinoma” OR “malignancy”)). For inclusion criteria, the studies were eligible if they: (a) examined the effects of baicalin or baicalein on breast cancer (in vitro, in vivo, or clinical studies), (b) were original research articles (e.g., experimental studies, preclinical trials), (c) provided full-text access, and (d) reported outcomes related to mechanisms, efficacy, or therapeutic applications. In the case of exclusion criteria, the studies were excluded if they: (a) were not published in English, (b) lacked full-text availability (e.g., conference abstracts, letters without data), (c) were non-original research (e.g., reviews, editorials, commentaries), and (d) did not directly investigate breast cancer or baicalin/baicalein.
Two authors independently screened all retrieved records in two stages: (a) title/abstract screening, i.e., irrelevant studies, non-English papers, and non-original articles were excluded, and (b) full-text review, i.e., remaining studies were assessed against inclusion/exclusion criteria. Discrepancies between the two reviewers were resolved through discussion until a consensus was reached.

3. Results

3.1. Bibliometric Analysis of Publications on Baicalin and Baicalein

The annual publication trends for baicalin and baicalein reveal a surge in research interest, particularly over the past decade (Figure 2a). After minimal output in the 20th century, publications grew steadily from 16 in 1992 to 57 in 2000, then exponentially to 709 in 2020. The early 2020s saw unprecedented growth, with annual publications exceeding 1000 papers (2022–2024), reflecting a heightened focus on their therapeutic potential in addressing global health challenges, such as the COVID-19 pandemic [30], where baicalin and baicalein were studied for their potential antiviral effects. Additionally, their applications in cancer therapy, neuroprotection, and metabolic disorders have garnered significant attention [27].
The distribution of scientific documents on baicalin and baicalein by language (Figure 2b) highlights the global reach of research on these compounds. The vast majority of publications (n = 8929; 86.9%) are in English, the universal language of scientific research. Chinese is the second most common language, with 1253 publications (12.2%). This aligns with the origins of baicalin and baicalein in traditional Chinese medicine and the significant contributions of Chinese researchers to this field. The presence of multiple languages (Japanese, Korean, Russian, etc.) underscores the worldwide interest in the research on baicalin and baicalein.
Figure 2c illustrates the distribution of scientific documents concerning baicalin and baicalein by document type. Original research papers, which include articles, make up the majority of these publications, totaling 7931 documents (77.2%). Reviews follow as the second most frequent type, comprising 1972 publications (19.2%).
Research on baicalin and baicalein spans over 100 countries. A world map (Figure 2d) visualizes the global distribution of research contributions on these topics, highlighting China as the dominant hub of activity. Significant research clusters also emerge across the Americas (United States, Brazil), Europe (United Kingdom, Italy, Germany), Asia (India, Japan, South Korea), and parts of Africa (Egypt, Nigeria) and the Middle East (Saudi Arabia, Iran).
The cluster analysis of authors’ keywords (n = 39) on baicalin and baicalein reveals five distinct clusters (Figure 3), each focusing on specific aspects of these compounds’ pharmacological properties, mechanisms, and applications.
Cluster 1 (red) highlights the broad pharmacological effects of baicalin and baicalein, including anti-inflammatory, antioxidant, and neuroprotective properties, and their potential in cancer therapy as bioactive flavonoids and polyphenols. Cluster 2 (green) focuses on phytochemistry and analytical studies, including extraction, quantification (HPLC), quality control, pharmacokinetics, and related compounds like wogonin and wogonoside. Cluster 3 (blue) emphasizes their application in COVID-19 through molecular docking and network pharmacology, alongside their significance in traditional Chinese medicine and potential synergy with other compounds. Cluster 4 (yellow) explores their impact on cell death pathways (apoptosis, autophagy, ferroptosis) and modulation of reactive oxygen species (ROS) linked to diseases like cancer. Lastly, Cluster 5 (violet) centers on their anti-inflammatory effects and modulation of the NF-κB signaling pathway. Together, these clusters showcase the versatile research landscape of baicalin and baicalein and their therapeutic potential in modern and traditional medicine, addressing global health issues like cancer and COVID-19.
Furthermore, Table 1 below is a summary of the five most frequently cited scientific publications in the Scopus database that reference baicalin and baicalein. These publications cover various aspects of polyphenols’ pharmacological properties, including antimicrobial activity, health benefits, and therapeutic applications. Cushnie and Lamb [31] lead with 3287 citations, reflecting their foundational work on the antimicrobial properties of flavonoids, including baicalin and baicalein. Del Rio et al. [32] follow with 2067 citations, emphasizing the health benefits of dietary polyphenols and their role in chronic disease prevention. Wu et al. [33], with 1712 citations, gained rapid recognition for their focus on COVID-19 therapeutic targets and the potential of baicalin, baicalein, and other bioactive compounds as antiviral agents. Moreover, this paper is the only original research paper in this set. Formica and Regelson [34], a classic review on quercetin and related bioflavonoids, has 1617 citations. Finally, Zhang et al. [35], with 1331 citations, provide a comprehensive review of extraction and isolation techniques for natural products, highlighting their importance in natural product research. The papers mentioned above demonstrate the diverse applications of polyphenol compounds, from antimicrobial and antiviral activities to chronic disease prevention and methodological advancements, and their high citation counts reflect the global scientific community’s interest in the therapeutic potential of these bioactive compounds.

3.2. Characteristics of Included Studies

In the case of systematic review, the initial search yielded 388 papers published up to 1 February 2025. After applying language filters, 376 articles were retained, as they were published in English. Subsequently, the articles were screened based on their document type, identifying 190 original research articles. Following a revision process to ensure relevance and eliminate articles with full text not available and studies not directly related to breast cancer and the application of baicalin or baicalein, 105 articles were deemed eligible for inclusion in this review. In the exclusion criteria, 12 papers were excluded for being non-English, 186 for not being original research articles, 14 due to lack of full-text availability, and 71 because they were not directly related to breast cancer and the application of baicalin/baicalein. The PRISMA flowchart is shown in Figure 4.
The 105 articles were classified into six thematic categories according to their primary focus on (a) baicalin, (b) baicalein, (c) plant extracts and herbal prescription used in traditional medicine, (d) derivatives of baicalein, (e) combination therapy, and (f) nanoparticle-based delivery systems.

3.3. The Use of Baicalin in the Treatment of Breast Cancer

Baicalin, a flavonoid glycoside, and glucuronide of baicalein, has demonstrated significant anticancer properties in various breast cancer models. Table 2 provides a summary of the research studies concerning the role of baicalin in combating breast cancer [35,36,37,38,39,40,41,42,43,44,45,46,47,48,49,50]. To date, 16 papers have been published on this topic, with the first appearing in 2001. Interestingly, Ikezoe et al. [36] demonstrated that baicalin could not inhibit the growth of the MCF-7 cell line. In other studies, the result was quite the opposite.
Baicalin has been shown to inhibit the proliferation of various breast cancer cell lines. Both Wang et al. [37] and Ge et al. [46] reported that baicalin inhibited MCF-7 cell proliferation in a dose- and time-dependent manner. At 200 µM, baicalin induced strong cytotoxicity and morphological changes indicative of cell death, arresting cells in the G0/G1 phase and upregulating p53 and the bax gene [37]. Moreover, with miRNA microarray analysis, Ge et al. [46] identified 92 upregulated and 35 downregulated miRNAs at 150 µM of baicalin.
Furthermore, baicalin induced apoptosis in different breast cancer cell lines through multiple mechanisms. It was evidenced by, e.g., chromatin condensation and upregulation of p53 and bax [37], increasing cytochrome c release, DNA fragmentation, and caspase-3, -8, and -9 activity in MDA-MB-231 and MCF-7 cells [45], as well as, by heightened ROS production in MCF-7 cells [50].
Anti-metastatic effects in breast cancer cell lines were also observed. In the vast majority of studies, baicalin inhibits breast cancer metastasis by targeting migration, invasion, and EMT, upregulating E-cadherin, and downregulating mesenchymal markers like Vimentin and N-cadherin [40,42,49].
In addition, signaling pathways involved in cancer progression were modulated by baicalin. Chung et al. [38] reported that baicalin inhibited NF-κB activation and TGF-β1-induced EMT in MCF-10A and MDA-MB-231 cells, suppressing migration and colony formation. Gao et al. [41] found that baicalin inhibited breast cancer cell proliferation, invasion, and migration by inducing G1/S arrest and suppressing the NF-κB pathway. Chen et al. [47] demonstrated that baicalin suppressed luminal breast cancer (ZR-75-1) growth by downregulating ESR1 but promoted triple-negative breast cancer (MDA-MB-231) growth by upregulating PROCR.
Finally, several in vivo studies have demonstrated the therapeutic efficacy of baicalin. Gao et al. [41] reported that baicalin reduced tumor growth in BALB/c nude mice and modulated inflammatory responses. Liu et al. [44] found that baicalin suppressed tumor growth and metastasis in BALB/c-null mice, upregulating E-cadherin and downregulating TGF-β1, vimentin, and p-Smad3 in tumors. Wang et al. [48] demonstrated that baicalin reversed bone loss in bone cancer pain rats, inhibiting osteoclast activation and reducing inflammation. Jia et al. [51] reported that baicalin suppressed epinephrine-induced migration and invasion of breast cancer cells, protecting against chronic stress-promoted metastasis in BALB/c and C57BL/6J mice.

3.4. Studies Investigating Baicalein as an Anticancer Agent

Baicalein, the aglycone of baicalin, was more commonly researched [26,52,53,54,55,56,57,58,59,60,61,62,63,64,65,66,67,68,69,70,71,72,73,74,75,76,77,78,79,80,81,82,83,84,85,86,87,88,89,90,91] (Table 3). Similarly to baicalin, baicalein has been shown to inhibit the proliferation of various breast cancer cell lines. The study of So et al. [52] was the first to demonstrate baicalein’s ability to inhibit MDA-MB-435 cell proliferation. Baicalein consistently inhibited the proliferation of breast cancer cell lines such as MDA-MB-435, MCF-7, MDA-MB-231, and 4T1. So et al. [52,53] reported that baicalein inhibited the proliferation of MDA-MB-435 and MCF-7 cells, with IC50 values of 5.9 µg/mL and 5.3 µg/mL, respectively. Interestingly, Kuntz et al. [54] found that MCF-7 cells were less sensitive to baicalein than intestinal cells. Ragazzon et al. [62] demonstrated that baicalein-induced G1-phase arrest in MCF-7 cells and caused DNA damage, highlighting its antiproliferative effects.
Mitochondrial dysfunction and caspase activation [55,60], modulation of Bcl-2 family proteins [55], and increased intracellular Ca2+ levels and ROS production [60,70] were observed as the response for baicalein-induced apoptosis.
Moreover, baicalein demonstrated potent anti-metastatic properties by inhibiting cancer cell adhesion, migration, and invasion. It suppressed the activity and expression of matrix metalloproteinases (MMPs), such as MMP-2 and MMP-9, which are critical for cancer cell invasion [64]. Baicalein also reversed epithelial-mesenchymal transition (EMT), a key process in metastasis, where Chen et al. [82] demonstrated that baicalein delayed tumor progression, reduced metastasis, and prolonged survival in a breast cancer mouse model by suppressing calpain-2 activity and modulating Ca2+ levels and ERK signaling. In another study, Gao et al. [71] found that baicalein downregulated SATB1 protein expression, inhibiting the proliferation and migration of MDA-MB-231 cells.
Baicalein targeted multiple signaling pathways, including MAPK/ERK/p38 pathway [63], PI3K/AKT/mTOR pathway [78], Wnt/β-catenin pathway [73], STAT3 signaling [84], and KDM4E/BICD1/PAR1 [88].
Regarding in vivo studies, in animal models, such as Sprague-Dawley rats, BALB/c mice, and SCID mice [69,74,88], baicalein significantly suppressed tumor growth, reduced metastasis, and prolonged survival. In detail, Wang et al. [69] reported that baicalein treatment significantly suppressed tumor growth without toxicity in an orthotopic mouse model of triple-negative breast cancer.
Baicalein exhibits potent anticancer properties in breast cancer models, including antiproliferative, pro-apoptotic, and antimetastatic effects. The compound modulates multiple signaling pathways and is efficacious in combination therapies and in targeting cancer stem cells. Baicalein showed limited effectiveness against breast cancer stem cells (CSCs) compared to other natural compounds like ginsenoside F2, tangeretin, and nobiletin [66].
Additionally, numerous recent studies have employed techniques like network pharmacology, molecular docking, and molecular dynamics simulations to minimize costs and accelerate drug discovery. As one example of such studies, He et al. [92] investigated the compound banmao capsule, a widely used antitumor mixture consisting of 11 herbs. The authors identified 128 compounds and 436 targets of this traditional medicine remedy, and moreover, VEGF (Vascular Endothelial Growth Factor) and estrogen signaling pathways seem to play a role in its antitumor activity. Similar studies, e.g., Qi et al. [93], aimed to investigate the anticancer mechanism of S. barbata, where the authors identified the active constituents of this plant, predicted their targets, and analyzed their interactions. Another study examined the potential of baicalein as an inhibitor of cyclooxygenase-2 (COX-2), a key enzyme linked to breast and ovarian cancers [94]. Finally, the study of Rathi et al. [95] explored the potential of baicalin and resveratrol as inhibitors of PIM-1 kinase, which plays a critical role in cancer progression by regulating cell proliferation, survival, and signaling pathways like PI3K/Akt/mTOR and JAK/STAT.

3.5. Anticancer Effects of Plant Extracts Containing Baicalein and Baicalin in Breast Cancer Models

As mentioned earlier, S. baicalensis is the primary source of baicalin and baicalein, but these compounds also occur in plants in other regions of the world. S. baicalensis has been utilized as a medicine in various East Asian countries for over 2000 years [18]. The studies included in the systematic review focused on extracts from Scutellaria species, P. amarus, T. aestivum, V. amygdalina, X. spinosum, and C. mersinensis, as well as Sanhuang Xiexin Decoction (SXD) and Huangqin Tang (HQT) formulations [19,20,21,22,23,24,96,97,98,99,100,101,102]. These extracts exhibited different levels of baicalein and baicalin, which influenced their distinct anticancer properties, and the data are summarized in Table 4.
Parajuli et al. [19] demonstrated the anticancer potential of thirteen Scutellaria species extracts in MDA-MB-231 cells, i.e., S. alpina, S. angulosa, S. baicalensis, S. barbata, S. costaricana, S. integrifolia, S. lateriflora, S. montana, S. ocmulgee, S. ovata, S. racemosa, S. scandens, and S. suffrutescens. The extracts contained varying levels of six flavonoids, with baicalin being the most abundant in most species. The extracts contained baicalein (0.21–2.34 μg/mg) and baicalin (0.92–28.16 μg/mg), with the highest values observed for the leaves of S. angulosa. The flavonoids showed potent antiproliferative and proapoptotic activities, with mechanisms involving cell cycle arrest (G1/G2 phase) and induction of apoptosis [19]. The extracts from S. baicalensis were also studied by Wang et al. [96], Yu et al. [97], and Park et al. [98]. Wang et al. [96] compared different fractions of the extracts with varied contents of baicalin and baicalein, and they found that a baicalin-deprived fraction, primarily containing baicalein and wogonin, demonstrated the most potent anticancer activity in MCF-7 cells and effectively induces apoptosis in MCF-7 cells. In contrast, other fractions did not exhibit similar effects, and cell cycle arrest was also observed. Yu et al. [97] reported that cellulase pretreatment was applied to enhance the antiproliferative potential of S. baicalensis extracts against MCF-7 and HCT-116 cells. The authors first optimized the deglycosylation conditions and found that 20 U/g cellulase at pH 4.8 and 50 °C for 8 h, leading to obtaining promising bioactive extracts. The final extract induced S-phase cell cycle arrest in HCT-116 cells and significantly increased early and late apoptosis rates in a dose-dependent manner [97].
Moreover, the aqueous extract of P. amarus mitigated Cr(VI)-induced oxidative toxicity in MDA-MB-435S cells [20]. Beara et al. [21] found baicalein in extracts of T. aestivum (black truffles), which notably hindered growth in both MCF-7 and HeLa cells. In contrast, T. magnatum (white truffles) lacked both baicalin and baicalein. The ethyl acetate extract of V. amygdalina leaves exhibited anticancer activity against 4T1 breast cancer cells by inducing apoptosis, enhancing cell accumulation in the G2/M phases of the cell cycle, and inhibiting the expression of PI3K and mTOR [22]. X. spinosum was another species that contained baicalin or baicalein either in methanol or hexane fractions [23]. Treatment with X. spinosum extracts significantly reduced tumor size in BALB/c mice, particularly with aqueous, methanol, and chloroform fractions. Moreover, the aqueous fraction significantly increased caspase-3 activity, suggesting a strong induction of apoptosis in cancer cells. In addition, no toxic effects on liver or kidney functions were observed in treated mice [23]. Finally, Yırtıcı et al. [24] investigated the bioactive properties of C. mersinensis, an endemic species in Turkey. The methanol extract contained significant phytochemicals, including scutellarin, chlorogenic acid, and baicalin. In vitro studies demonstrated that the methanol extract exhibited cytotoxic effects against breast cancer cells (MCF-7), which was more effective than against other breast cancer cell lines (MDA-MB-231 and SKBR-3) with IC50 values ranging from 22.17 to 46.20 µg/mL.
Traditional remedies and medicines, renowned for their extensive historical use, have also been evaluated for their potential as anticancer agents. Du et al. [99] assessed the Shuganning injection (SGNI). SGNI is a reformulation of the traditional decoction, first documented 1800 years ago. It consists of four herbal extracts: Ganoderma lucidum, Isatidis radix, Gardeniae fructus, and Artemisiae scopariae, along with baicalin. SGNI was approved by the China Food and Drug Administration (CFDA) as a traditional Chinese patent medicine in 2002 and is used to treat conditions such as hepatitis, high bilirubin levels, liver damage, fatty liver, and cholangitis. Shuganning injection selectively inhibited the proliferation of triple-negative breast cancer (TNBC) cells, and in in vivo studies confirmed that SGNI significantly suppresses the growth of TNBC xenografts and induces ferroptosis in nude mice [99].
Another so-called “heirloom recipe”, Formula X, contained Coptis chinensis Franch., S. baicalensis Georgi, Vincetoxicum atratum, Curcuma aromatica Salisb., and Atractylodes macrocephala Koidz. The authors revealed that Formula X could inhibit the growth of various cancer cells while reducing the expression of key proteins. A new formulation, BBS, containing berberine, baicalin, and saponin, effectively suppressed tumor growth in a mouse model [100].
Last but not least, Sanhuang Xiexin Decoction (SXD) was prepared by mixing Rheum palmatum L., Scutellariae Radix, and Coptis chinensis Franch in a 2:1:1 ratio. The bioactive compounds found in SXD were rhein, coptisine, berberine hydrochloride, and baicalin. The authors found that SXD demonstrated significant antitumor effects in 4T1 breast cancer mice through modulation of lipid profiles and cytokine levels, and low toxicity levels in mice were observed [101].
Plant extracts containing baicalein and baicalin exhibit significant anticancer potential in breast cancer models. According to the above-cited papers, these extracts inhibit cancer cell proliferation, induce apoptosis, and suppress tumor growth through diverse mechanisms. The variability in baicalein and baicalin content across extracts highlights the importance of standardized formulations for therapeutic applications. Despite the promising anticancer effects of plant extracts containing baicalein and baicalin, challenges in standardization remain a critical barrier to therapeutic translation. Variability in compound levels across species (e.g., S. angulosa vs. S. baicalensis), growth conditions, extraction methods (e.g., cellulase pretreatment in Yu et al. [97]), and formulation processes (e.g., SXD vs. HQT decoctions) complicates reproducibility. For instance, the baicalin-deprived fraction in Wang et al. [96] showed enhanced activity, highlighting how minor compositional differences significantly alter efficacy. Batch-to-batch inconsistencies in traditional formulations like SGNI [99] further underscore the need for rigorous quality control, phytochemical profiling, and harmonized protocols to ensure clinical reliability.

3.6. Derivatives of Baicalein in Anticancer Studies Against Breast Cancer

Baicalein demonstrates more substantial anticancer effects than baicalin because baicalein exhibits higher bioavailability compared to baicalin due to its structural characteristics, as it does not require hydrolysis for cellular uptake and its ability to interact with intracellular targets directly [103]. Furthermore, the clinical utility of baicalein is limited by its poor pharmacokinetic properties. The chemical modification of baicalein to create derivatives with improved activity and bioavailability presents a promising strategy to address these limitations. Over the last fifteen years, five papers have been published in which scientists attempted to obtain new baicalein derivatives and tested these compounds’ activity against breast cancer lines [104,105,106,107,108]. Table 5 provides a detailed overview of various baicalein derivatives and their effects on breast cancer cell lines.
One of them, 5,6-dihydroxy-7-(3-methylbut-2-enyloxy)- 2-phenyl-4H-chromen-4-one (prenylated baicalein, Figure 5), was synthesized by microwave-assisted organic synthesis from baicalein, prenyl bromide, and anhydrous potassium carbonate [104]. The obtained compound exhibited a dose-dependent inhibitory effect on MCF-7 and MDA-MB-231 cells. The observed activity was more potent in the case of MCF-7 cells and was mediated through interactions with estrogen receptors. Additionally, prenylated baicalein showed potential for synergistic effects with 4-hydroxytamoxifen and fulvestrant, suggesting its utility in combination therapies for estrogen receptor-positive breast cancer [104].
Two other sulfated derivatives, namely baicalein-7-O-sulfate and baicalein-8-sodium sulfonate, were investigated by Wang et al. [105] to evaluate their effects on the growth of MCF-7 cells and normal epithelial H184B5F5/M10 cells. The compounds showed cytotoxic effects only on MCF-7 cells, and baicalein-8-sodium sulfonate demonstrated the highest cytotoxic effect. Moreover, the obtained analogs induced apoptosis, increased intracellular ROS levels, and led to cell cycle arrest primarily at the G0/G1 phase.
Oroxylin A, a methylated metabolite of baicalein in the human body, was studied by An et al. [106]. Oroxylin A inhibited CYP1B1-mediated 4-hydroxylation of 17β-estradiol, a key process in estrogen metabolism, and was found to be more potent than baicalein [106].
Marzec et al. [107] designed a range of halogenated derivatives based on the natural flavonoids baicalein and chrysin. In the case of the former, it was found that halogenation increased the antiproliferative effects on different cell lines, such as LoVo, A549, MCF-7, and MCF-10A. Unfortunately, this indicates a lack of selectivity because they were also active against non-tumorigenic breast cells (MCF-10A). 8-Bromobaicalein was also studied by Yasuda et al. [108]. Its cytotoxic activity was evaluated, and the activity after its complexation with 2,6-di-O-methyl-β-cyclodextrin. The authors revealed that the inclusion complex of 8-bromobaicalein and cyclodextrin demonstrated significantly enhanced anticancer activity against MCF-7 human breast cancer cells compared to baicalein analog alone, with the IC50 values of 5.91 ± 0.35 and 15.53 ± 1.17 μM, respectively.
Chemical modifications, such as prenylation, sulfonation, halogenation, and complexation with cyclodextrins, have enhanced baicalein’s anticancer activity. These derivatives exhibit diverse mechanisms of action, including induction of apoptosis, cell cycle arrest, and modulation of oxidative stress. However, further research is needed to optimize their selectivity and minimize toxicity to non-cancerous cells.
Table 6 provides a detailed comparison between baicalin and baicalein, focusing on their molecular properties, pharmacokinetics [109], biological activities, and mechanisms of action. Baicalin, which has a molecular weight of 446.36 g/mol, features a sugar moiety that enhances its polarity. This is evidenced by its higher topological polar surface area (TPSA: 187.12 Å2) and hydrogen-bonding capacity (11 acceptors, 6 donors) compared to its aglycone counterpart, baicalein. The latter also exhibits greater lipophilicity (LogP 2.24 vs. 0.25). Baicalin’s high polarity results in low water solubility (−3.41 LogS) and reduced gastrointestinal (GI) absorption, further affected by its status as a P-glycoprotein (P-gp) substrate. Conversely, baicalein’s lipophilic nature enhances GI absorption despite its lower solubility. Although neither compound crosses the blood-brain barrier (BBB), baicalein has better skin penetration (Log Kp −5.70 compared to −8.23 for baicalin). Importantly, baicalein inhibits cytochrome P450 enzymes (CYP1A2, CYP2D6, CYP3A4), which raises concerns for potential drug interactions, whereas baicalin does not inhibit CYP enzymes.

3.7. Synergistic Effects of Baicalin/Baicalein in Combination Therapies for Breast Cancer

Combination therapies have emerged as a promising strategy for enhancing the efficacy of anticancer treatments while minimizing side effects. By utilizing multiple agents with complementary mechanisms of action, these therapies offer a robust approach to overcoming the limitations associated with traditional single-agent treatments, thereby improving cancer patients’ outcomes. The outcomes of combining baicalin and baicalein with other therapeutic agents focused on their synergistic and antagonistic effects in breast cancer treatment are presented in Table 7 [110,111,112,113,114,115,116,117,118,119,120,121,122,123].
Baicalin, together with Salvia miltiorrhiza extract, was the first combination documented in scientific literature and utilized in breast cancer cell lines [110]. Their combination resulted in synergistic effects, significantly enhancing the inhibition of breast cancer cell lines (MCF-7 and T-47D) as well as other cancer types (FaDu and CAL-27). Similarly, Xi et al. [114] found that the combination of baicalein and low-frequency ultrasound (5 min at 840 kHz, 0.75 W/cm2) demonstrated synergistic effects in reducing the invasive activity of triple-negative breast cancer cells (MDA-MB-231). The invasive capacity of cells in the low-frequency group, baicalein group, and combination group was reduced by 21.6%, 31.9%, and 51.6%, respectively. This was achieved by downregulating the expression of MMP-2, MMP-9, and urokinase-type plasminogen activator (u-PA), key mediators of cancer cell invasion and metastasis [114].
Baicalin and baicalein have been shown to mitigate the toxic side effects of chemotherapy agents. Chang et al. [111] found that baicalein protected against doxorubicin-induced cardiotoxicity by reducing ROS generation and preserving mitochondrial function in chick cardiomyocytes without interfering with doxorubicin’s anticancer activity in MCF-7 cells. In vivo studies have also shown promising results. Zeng et al. [116] demonstrated that baicalin enhanced the chemosensitivity of MDA-MB-231 cells to docetaxel, inhibiting tumor growth and pulmonary metastasis in female BALB/c mice. Additionally, Shehatta et al. [118] reported that the combination of baicalin and 5-fluorouracil significantly inhibited tumor growth and promoted apoptosis in Swiss albino mice. Similarly, Ibrahim et al. [120] reported that baicalein partially mitigated capecitabine-induced cardiotoxicity, oxidative stress, and inflammation in adult female albino rats.
Not all combinations of baicalin or baicalein with other agents yield synergistic effects. Zhan et al. [112] observed that the combination of baicalein and paclitaxel resulted in antagonistic effects, reducing the growth-inhibitory effect of paclitaxel in MCF-7 cells.
The mechanisms underlying the synergistic effects of baicalin and baicalein in combination therapies are diverse. An et al. [113] demonstrated that the combination of baicalein with U0126 enhanced cell proliferation inhibition, apoptosis induction, and migration suppression in MCF-7 cells, likely through modulation of the MAPK/ERK signaling pathway. Lin et al. [118] reported that baicalin enhanced doxorubicin’s anticancer effects by increasing oxidative stress, calcium levels, and mitochondrial dysfunction in breast cancer cells. Furthermore, Hua et al. [121] found that baicalein enhanced the sensitivity of MDA-MB-231 cells to doxorubicin by activating autophagy and mitophagy.
Recent studies by Sisin et al. [115,119] have explored novel combinations involving bismuth oxide nanoparticles, cisplatin, and a baicalein-rich fraction from O. indicum. The combination treatment was highly effective in breast cancer cells, demonstrating the most potent enhancement of ROS generation and radiosensitization across all types of radiation tested. However, it also showed significant radiosensitization effects in normal cells when a baicalein-rich fraction was added, which could impact safety profiles [115,119]. Awajan et al. [123] reported that the combination of baicalin, epigallocatechin gallate (EGCG), and vincristine had synergistic effects in vincristine-resistant cancer cells, significantly reducing tumor size and achieving high cure rates in female BALB/c mice.

3.8. Nanotechnology-Based Delivery Systems for Baicalin/Baicalein in Breast Cancer Research

Nanotechnology presents a promising solution to the challenges associated with traditional drug delivery systems by enabling targeted delivery. Nanoparticles can enhance therapeutic agents’ solubility, stability, and bioavailability while minimizing off-target effects [124]. This approach has been explored for various compounds, including baicalein and baicalin [125,126,127,128,129,130,131,132,133,134,135,136]. Scientists have used, e.g., iron oxide and gold nanoparticles, nanoemulsions, nanostructured lipid carriers, nanocapsules, and Metal-Organic Frameworks (MOFs) (Table 8).
Kavithaa et al. [125,129] demonstrated that baicalein-loaded iron oxide nanoparticles induced significant cytotoxicity in MDA-MB-231 cells (IC50 = 22 µg/mL) by disrupting the cell cycle and promoting apoptosis. The nanoparticles upregulated pro-apoptotic proteins (Bax, cytochrome-c, caspase-3, PARP, and p53) while downregulating the anti-apoptotic protein Bcl-2. Similar findings were reported by Srivastava et al. [133], who developed a baicalein and cinnamon essential oil nanoemulsion that exhibited 19-fold and 23-fold higher cytotoxicity compared to free baicalein after 12 and 24 h, respectively. This enhanced activity was attributed to improved drug permeability and cellular uptake via endocytosis.
Baicalin was also applied in nano-delivery systems. Lee et al. [126] developed gold nanoparticles (AuNPs) conjugated with thiolated beta-cyclodextrin for baicalin delivery, significantly enhancing its apoptotic effects in MCF-7 cells. Similarly, El-Gogary et al. [130] reported that baicalin-loaded polylactide-glycolide (PLGA) nanocapsules demonstrated superior anticancer activity, sustained drug release, and enhanced cellular uptake in MCF-7 and MDA-MB-231 cells.
Several studies have explored the synergistic effects of baicalein combined with other therapeutic agents. Meng et al. [127] developed a baicalein and paclitaxel nanoemulsion that showed enhanced anticancer efficacy in MCF-7 and MCF-7/Tax cells. The nanoemulsion achieved a tumor inhibition rate of 77.0% in an MCF-7/Tax xenograft model, with minimal weight loss in treated mice. Liu et al. [128] reported similar findings for a hyaluronic acid-decorated nanostructured lipid carrier (NLC) co-delivering baicalein and doxorubicin, which exhibited a tumor inhibition rate of 88% in Kunming mice.
Mi et al. [131] designed a methoxy poly(ethylene glycol)-folic acid-decorated zeolitic imidazolate framework (ZIF-8) for baicalin delivery, which demonstrated excellent tumor-targeting capability and pH-responsive drug release. Similarly, Meng et al. [132] and Liu et al. [135] developed folate-modified albumin nanoparticles for baicalin delivery, which showed sustained drug release, enhanced cellular uptake, and potent anticancer activity in vitro and in vivo.
Finally, green synthesis of nanoparticles using plant extracts has also been explored. Gharari et al. [136] synthesized silver nanoparticles using S. multicaulis leaf extract, demonstrating significant antioxidant, anticancer, and apoptotic activities in MDA-MB-231 cells.

4. Conclusions

This systematic review thoroughly evaluates the roles of baicalin and baicalein in breast cancer, emphasizing their therapeutic potential through various mechanisms. Both flavonoids demonstrate significant antiproliferative, pro-apoptotic, and anti-metastatic effects across different breast cancer models. Baicalin and baicalein regulate key pathways, including apoptosis (via caspase activation and Bcl-2 family regulation), autophagy, oxidative stress (ROS modulation), and inflammation (NF-κB suppression) (Figure 6).
Plant extracts from S. baicalensis and other species enhance anticancer effects through synergistic interactions, while chemical derivatives (e.g., sulfonated, halogenated, and cyclodextrin-complexed forms) increase efficacy and selectivity. Combination therapies with conventional agents like doxorubicin or docetaxel produce synergistic outcomes, reducing chemotherapy resistance and toxicity. Nanotechnology-based delivery systems, such as lipid carriers and metal-organic frameworks, tackle bioavailability challenges and improve tumor-targeted delivery.
Despite encouraging preclinical outcomes, clinical translation remains limited. Variability in responses across breast cancer subtypes and potential off-target effects in non-cancerous cells highlight the need for further optimization. Future research should focus on large-scale, randomized clinical trials to validate efficacy across breast cancer subtypes, especially in resistant or metastatic situations. Standardized formulations with defined ratios of baicalein/baicalin (e.g., enhanced through nanotechnology [125,126,127,128,129,130,131,132,133,134,135,136]) and advanced delivery systems (e.g., tumor-targeted nanoparticles, cyclodextrin complexes [108]) could improve bioavailability and minimize off-target effects. Additionally, investigating synergistic combinations with immunotherapy or targeted therapies, along with mechanistic studies on subtype-specific responses (e.g., ER+ vs. TNBC), may reveal personalized therapeutic strategies. Addressing these gaps will connect preclinical promise to clinical utility. Collectively, baicalin and baicalein emerge as promising candidates for adjuvant or standalone therapies, warranting continued exploration in the evolving landscape of breast cancer treatment.

Supplementary Materials

The following supporting information can be downloaded at https://www.mdpi.com/article/10.3390/cimb47030181/s1.

Funding

This research received no external funding.

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.

Conflicts of Interest

The authors declare no conflicts of interest.

Abbreviations

The following abbreviations are used in this manuscript:
4T1Mouse mammary carcinoma cell line
AKTProtein Kinase B
ALOX12/15Arachidonate 12/15-Lipoxygenase
ARAndrogen Receptor
AuNPsGold Nanoparticles
BaxBcl-2-Associated X Protein
BBBBlood-Brain Barrier
Bcl-2B-Cell Lymphoma 2
BCTBreast-Conserving Therapy
CCIDCell Cycle Inhibitory Dose
CDK4/6Cyclin-Dependent Kinase 4/6
COX-2Cyclooxygenase-2
CSCCancer Stem Cells
CYP1Cytochrome P450 Family 1
CYP1B1Cytochrome P450 Family 1 Subfamily B Member 1
DDIT4DNA Damage-Inducible Transcript 4
Drp1Dynamin-Related Protein 1
E2Estradiol
EGCGEpigallocatechin Gallate
EMTEpithelial-Mesenchymal Transition
EREstrogen Receptor
ERKExtracellular Signal-Regulated Kinase
ERαEstrogen Receptor Alpha
FAKFocal Adhesion Kinase
GI AbsorptionGastrointestinal Absorption
GLOBOCANGlobal Cancer Observatory
GPR30G Protein-Coupled Estrogen Receptor 30
H-BondHydrogen Bond
HCT-116Human Colon Cancer Cell Line
HER2Human Epidermal Growth Factor Receptor 2
HIF-1αHypoxia-Inducible Factor 1-Alpha
HR+Hormone Receptor-Positive
HUVECsHuman Umbilical Vein Endothelial Cells
IC50Half-Maximal Inhibitory Concentration
IFIT2Interferon-Induced Protein with Tetratricopeptide Repeats 2
IGF-IInsulin-Like Growth Factor 1
IL-10Interleukin 10
IL-6Interleukin 6
IORTIntraoperative Radiotherapy
JNKc-Jun N-Terminal Kinase
LDHLactate Dehydrogenase
Log KpLogarithm of Skin Permeation Coefficient
LogPo/wLogarithm of Octanol-Water Partition Coefficient (lipophilicity measure)
LogSLogarithm of Aqueous Solubility
LOX-12Lipoxygenase-12
MAPKMitogen-Activated Protein Kinase
MCF-10ANon-Tumorigenic Breast Epithelial Cell Line
MDA-MB-231Human Triple-Negative Breast Cancer Cell Line
MDA-MB-468Human Breast Cancer Cell Line
miRNAMicroRNA
MMPMatrix Metalloproteinase
MMTV-PyMTMouse Mammary Tumor Virus-Polyoma Middle T Antigen
MOFsMetal-Organic Frameworks
mTORMammalian Target of Rapamycin
NF-κBNuclear Factor Kappa B
NLCNanostructured Lipid Carrier
NOD/SCIDNon-Obese Diabetic/Severe Combined Immunodeficiency
P-gpP-glycoprotein (efflux transporter)
PARPPoly (ADP-Ribose) Polymerase
PD-L1Programmed Death-Ligand 1
PI3KPhosphoinositide 3-Kinase
PIM-1Proto-oncogene serine/threonine-protein kinase
PLGAPolylactic-co-Glycolic Acid
PRISMAPreferred Reporting Items for Systematic Reviews and Meta-Analyses
PROCRProtein C Receptor
ROSReactive Oxygen Species
SATB1Special AT-Rich Sequence-Binding Protein 1
SGNIShuganning Injection
SREBP1Sterol Regulatory Element-Binding Protein 1
STAT3Signal Transducer and Activator of Transcription 3
TGF-β1Transforming Growth Factor Beta 1
TNBCTriple-Negative Breast Cancer
TNF-αTumor Necrosis Factor Alpha
TPSATopological Polar Surface Area
u-PAUrokinase-Type Plasminogen Activator
VEGFVascular Endothelial Growth Factor
ZEB1Zinc Finger E-Box Binding Homeobox 1
ZIF-8Zeolitic Imidazolate Framework 8

References

  1. Bray, F.; Ferlay, J.; Soerjomataram, I.; Siegel, R.L.; Torre, L.A.; Jemal, A. Global Cancer Statistics 2018: GLOBOCAN Estimates of Incidence and Mortality Worldwide for 36 Cancers in 185 Countries. CA Cancer J. Clin. 2018, 68, 394–424. [Google Scholar] [CrossRef] [PubMed]
  2. Cao, W.; Chen, H.-D.; Yu, Y.-W.; Li, N.; Chen, W.-Q. Changing Profiles of Cancer Burden Worldwide and in China: A Secondary Analysis of the Global Cancer Statistics 2020. Chin. Med. J. 2021, 134, 783–791. [Google Scholar] [CrossRef] [PubMed]
  3. Bray, F.; Laversanne, M.; Sung, H.; Ferlay, J.; Siegel, R.L.; Soerjomataram, I.; Jemal, A. Global Cancer Statistics 2022: GLOBOCAN Estimates of Incidence and Mortality Worldwide for 36 Cancers in 185 Countries. CA Cancer J. Clin. 2024, 74, 229–263. [Google Scholar] [CrossRef] [PubMed]
  4. Guo, X.; Zuo, Z.; Wang, X.; Sun, Y.; Xu, D.; Liu, G.; Tong, Y.; Zhang, Z. Epidemiology, Risk Factors and Mechanism of Breast Cancer and Atrial Fibrillation. CardioOncology 2024, 10, 92. [Google Scholar] [CrossRef]
  5. Waks, A.G.; Winer, E.P. Breast Cancer Treatment: A Review. JAMA 2019, 321, 288. [Google Scholar] [CrossRef]
  6. Tong, C.W.S.; Wu, M.; Cho, W.C.S.; To, K.K.W. Recent Advances in the Treatment of Breast Cancer. Front. Oncol. 2018, 8, 227. [Google Scholar] [CrossRef]
  7. Murawa, P.; Murawa, D.; Adamczyk, B.; Połom, K. Breast Cancer: Actual Methods of Treatment and Future Trends. Rep. Pract. Oncol. Radiother. 2014, 19, 165–172. [Google Scholar] [CrossRef]
  8. da Silva Fernandes, T.; Gillard, B.M.; Dai, T.; Martin, J.C.; Chaudhry, K.A.; Dugas, S.M.; Fisher, A.A.; Sharma, P.; Wu, R.; Attwood, K.M.; et al. Inosine Monophosphate Dehydrogenase 2 (IMPDH2) Modulates Response to Therapy and Chemo-Resistance in Triple Negative Breast Cancer. Sci. Rep. 2025, 15, 1061. [Google Scholar] [CrossRef]
  9. Murphy, C.G.; Dickler, M.N. Endocrine Resistance in Hormone-Responsive Breast Cancer: Mechanisms and Therapeutic Strategies. Endocr. Relat. Cancer 2016, 23, R337–R352. [Google Scholar] [CrossRef]
  10. Anamika, F.; Chitkara, A.; Saharan, K.; Choudhary, T.; Soni, U.; Harmon, G.A.; Saeed, A. Trastuzumab-Induced Cardiotoxicity in Breast Cancer Patients: A Meta-Analysis and Review of the Literature (2012–2022). J. Clin. Oncol. 2023, 41, e24101. [Google Scholar] [CrossRef]
  11. Jabagi, M.J.; Goncalves, A.; Vey, N.; Le Tri, T.; Zureik, M.; Dray-Spira, R. Risk of Hematologic Malignant Neoplasms after Postoperative Treatment of Breast Cancer. Cancers 2019, 11, 1463. [Google Scholar] [CrossRef]
  12. Sharma, E.; Attri, D.C.; Sati, P.; Dhyani, P.; Szopa, A.; Sharifi-Rad, J.; Hano, C.; Calina, D.; Cho, W.C. Recent Updates on Anticancer Mechanisms of Polyphenols. Front. Cell Dev. Biol. 2022, 10, 1005910. [Google Scholar] [CrossRef] [PubMed]
  13. Asensi, M.; Ortega, A.; Mena, S.; Feddi, F.; Estrela, J.M. Natural Polyphenols in Cancer Therapy. Crit. Rev. Clin. Lab. Sci. 2011, 48, 197–216. [Google Scholar] [CrossRef]
  14. Sauter, E.R. Cancer Prevention and Treatment Using Combination Therapy with Natural Compounds. Expert Rev. Clin. Pharmacol. 2020, 13, 265–285. [Google Scholar] [CrossRef] [PubMed]
  15. Zhang, H.-W.; Hu, J.-J.; Fu, R.-Q.; Liu, X.; Zhang, Y.-H.; Li, J.; Liu, L.; Li, Y.-N.; Deng, Q.; Luo, Q.-S.; et al. Flavonoids Inhibit Cell Proliferation and Induce Apoptosis and Autophagy through Downregulation of PI3Kγ Mediated PI3K/AKT/mTOR/p70S6K/ULK Signaling Pathway in Human Breast Cancer Cells. Sci. Rep. 2018, 8, 11255. [Google Scholar] [CrossRef] [PubMed]
  16. Kursvietiene, L.; Kopustinskiene, D.M.; Staneviciene, I.; Mongirdiene, A.; Kubová, K.; Masteikova, R.; Bernatoniene, J. Anti-Cancer Properties of Resveratrol: A Focus on Its Impact on Mitochondrial Functions. Antioxidants 2023, 12, 2056. [Google Scholar] [CrossRef]
  17. Li-Weber, M. New Therapeutic Aspects of Flavones: The Anticancer Properties of Scutellaria and Its Main Active Constituents Wogonin, Baicalein and Baicalin. Cancer Treat. Rev. 2009, 35, 57–68. [Google Scholar] [CrossRef]
  18. Zhao, Q.; Chen, X.-Y.; Martin, C. Scutellaria baicalensis, the Golden Herb from the Garden of Chinese Medicinal Plants. Sci. Bull. 2016, 61, 1391–1398. [Google Scholar] [CrossRef]
  19. Parajuli, P.; Joshee, N.; Rimando, A.; Mittal, S.; Yadav, A. In Vitro Antitumor Mechanisms of Various Scutellaria Extracts and Constituent Flavonoids. Planta Med. 2009, 75, 41–48. [Google Scholar] [CrossRef]
  20. Guha, G.; Rajkumar, V.; Ashok Kumar, R.; Mathew, L. Aqueous Extract of Phyllanthus amarus Inhibits Chromium(VI)-Induced Toxicity in MDA-MB-435S Cells. Food Chem. Toxicol. 2010, 48, 396–401. [Google Scholar] [CrossRef]
  21. Beara, I.N.; Lesjak, M.M.; Cetojević-Simin, D.D.; Marjanović, Z.S.; Ristić, J.D.; Mrkonjić, Z.O.; Mimica-Dukić, N.M. Phenolic Profile, Antioxidant, Anti-Inflammatory and Cytotoxic Activities of Black (Tuber aestivum Vittad.) and White (Tuber magnatum Pico) Truffles. Food Chem. 2014, 165, 460–466. [Google Scholar] [CrossRef] [PubMed]
  22. Hasibuan, P.A.Z.; Harahap, U.; Sitorus, P.; Satria, D. The Anticancer Activities of Vernonia amygdalina Delile. Leaves on 4T1 Breast Cancer Cells through Phosphoinositide 3-Kinase (PI3K) Pathway. Heliyon 2020, 6, e04449. [Google Scholar] [CrossRef] [PubMed]
  23. Al Kury, L.T.; Taha, Z.; Mahmod, A.I.; Talib, W.H. Xanthium spinosum L. Extracts Inhibit Breast Cancer in Mice by Apoptosis Induction and Immune System Modulation. Pharmaceuticals 2022, 15, 1504. [Google Scholar] [CrossRef]
  24. Yırtıcı, Ü.; Ergene, A.; Adem, Ş.; Atalar, M.N.; Eyüpoğlu, V.; Rawat, R.; Arat, E.; Hamzaoğlu, E. Centaurea mersinensis Phytochemical Composition and Multi-Dimensional Bioactivity Properties Supported by Molecular Modeling. J. Biomol. Struct. Dyn. 2024, 42, 2341–2357. [Google Scholar] [CrossRef]
  25. Wen, Y.; Wang, Y.; Zhao, C.; Zhao, B.; Wang, J. The Pharmacological Efficacy of Baicalin in Inflammatory Diseases. Int. J. Mol. Sci. 2023, 24, 9317. [Google Scholar] [CrossRef]
  26. Liu, Z.-H.; Yang, C.-X.; Zhang, L.; Yang, C.-Y.; Xu, X.-Q. Baicalein, as a Prooxidant, Triggers Mitochondrial Apoptosis in MCF-7 Human Breast Cancer Cells through Mobilization of Intracellular Copper and Reactive Oxygen Species Generation. Onco. Targets. Ther. 2019, 12, 10749–10761. [Google Scholar] [CrossRef]
  27. Hu, Z.; Guan, Y.; Hu, W.; Xu, Z.; Ishfaq, M. An Overview of Pharmacological Activities of Baicalin and Its Aglycone Baicalein: New Insights into Molecular Mechanisms and Signaling Pathways. Iran. J. Basic Med. Sci. 2022, 25, 14–26. [Google Scholar] [CrossRef]
  28. Morshed, A.K.M.H.; Paul, S.; Hossain, A.; Basak, T.; Hossain, M.S.; Hasan, M.M.; Hasibuzzaman, M.A.; Rahaman, T.I.; Mia, M.A.R.; Shing, P.; et al. Baicalein as Promising Anticancer Agent: A Comprehensive Analysis on Molecular Mechanisms and Therapeutic Perspectives. Cancers 2023, 15, 2128. [Google Scholar] [CrossRef] [PubMed]
  29. Page, M.J.; McKenzie, J.E.; Bossuyt, P.M.; Boutron, I.; Hoffmann, T.C.; Mulrow, C.D.; Shamseer, L.; Tetzlaff, J.M.; Akl, E.A.; Brennan, S.E.; et al. The PRISMA 2020 Statement: An Updated Guideline for Reporting Systematic Reviews. BMJ 2021, 372, n71. [Google Scholar] [CrossRef]
  30. Dinda, B.; Dinda, M.; Dinda, S.; De, U.C. An Overview of Anti-SARS-CoV-2 and Anti-Inflammatory Potential of Baicalein and Its Metabolite Baicalin: Insights into Molecular Mechanisms. Eur. J. Med. Chem. 2023, 258, 115629. [Google Scholar] [CrossRef]
  31. Cushnie, T.P.T.; Lamb, A.J. Antimicrobial Activity of Flavonoids. Int. J. Antimicrob. Agents 2005, 26, 343–356. [Google Scholar] [CrossRef] [PubMed]
  32. Del Rio, D.; Rodriguez-Mateos, A.; Spencer, J.P.E.; Tognolini, M.; Borges, G.; Crozier, A. Dietary (Poly)Phenolics in Human Health: Structures, Bioavailability, and Evidence of Protective Effects against Chronic Diseases. Antioxid. Redox Signal. 2013, 18, 1818–1892. [Google Scholar] [CrossRef]
  33. Wu, C.; Liu, Y.; Yang, Y.; Zhang, P.; Zhong, W.; Wang, Y.; Wang, Q.; Xu, Y.; Li, M.; Li, X.; et al. Analysis of Therapeutic Targets for SARS-CoV-2 and Discovery of Potential Drugs by Computational Methods. Acta Pharm. Sin. B. 2020, 10, 766–788. [Google Scholar] [CrossRef] [PubMed]
  34. Formica, J.V.; Regelson, W. Review of the Biology of Quercetin and Related Bioflavonoids. Food Chem. Toxicol. 1995, 33, 1061–1080. [Google Scholar] [CrossRef]
  35. Zhang, Q.-W.; Lin, L.-G.; Ye, W.-C. Techniques for Extraction and Isolation of Natural Products: A Comprehensive Review. Chin. Med. 2018, 13, 20. [Google Scholar] [CrossRef] [PubMed]
  36. Ikezoe, T.; Chen, S.S.; Heber, D.; Taguchi, H.; Koeffler, H.P. Baicalin Is a Major Component of PC-SPES Which Inhibits the Proliferation of Human Cancer Cells via Apoptosis and Cell Cycle Arrest. Prostate 2001, 49, 285–292. [Google Scholar] [CrossRef]
  37. Wang, N.; Tang, L.-J.; Zhu, G.-Q.; Peng, D.-Y.; Wang, L.; Sun, F.-N.; Li, Q.-L. Apoptosis Induced by Baicalin Involving Up-Regulation of P53 and Bax in MCF-7 Cells. J. Asian Nat. Prod. Res. 2008, 10, 1129–1135. [Google Scholar] [CrossRef]
  38. Chung, H.; Choi, H.S.; Seo, E.-K.; Kang, D.-H.; Oh, E.-S. Baicalin and Baicalein Inhibit Transforming Growth Factor-Β1-Mediated Epithelial-Mesenchymal Transition in Human Breast Epithelial Cells. Biochem. Biophys. Res. Commun. 2015, 458, 707–713. [Google Scholar] [CrossRef]
  39. Gao, Y.; Ni, Q.; Chen, H.; Wei, N.; Jia, Q. Up-Regulation of miR-200 b by Baicalin to Inhibit Migration of MCF-7 Breast Cancer Cells. Int. J. Clin. Exp. Med. 2017, 10, 2360–2366. [Google Scholar]
  40. Zhou, T.; Zhang, A.; Kuang, G.; Gong, X.; Jiang, R.; Lin, D.; Li, J.; Li, H.; Zhang, X.; Wan, J.; et al. Baicalin Inhibits the Metastasis of Highly Aggressive Breast Cancer Cells by Reversing Epithelial-to-Mesenchymal Transition by Targeting β-Catenin Signaling. Oncol. Rep. 2017, 38, 3599–3607. [Google Scholar] [CrossRef]
  41. Gao, Y.; Liu, H.; Wang, H.; Hu, H.; He, H.; Gu, N.; Han, X.; Guo, Q.; Liu, D.; Cui, S.; et al. Baicalin Inhibits Breast Cancer Development via Inhibiting IĸB Kinase Activation in Vitro and in Vivo. Int. J. Oncol. 2018, 53, 2727–2736. [Google Scholar] [CrossRef] [PubMed]
  42. Yang, K.; Zeng, L.; Ge, A.; Chen, Z.; Bao, T.; Long, Z.; Ge, J.; Huang, L. Investigating the Regulation Mechanism of Baicalin on Triple Negative Breast Cancer’s Biological Network by a Systematic Biological Strategy. Biomed. Pharmacother. 2019, 118, 109253. [Google Scholar] [CrossRef]
  43. Duan, X.; Guo, G.; Pei, X.; Wang, X.; Li, L.; Xiong, Y.; Qiu, X. Baicalin Inhibits Cell Viability, Migration and Invasion in Breast Cancer by Regulating miR-338-3p and MORC4. Onco. Targets. Ther. 2019, 12, 11183–11193. [Google Scholar] [CrossRef]
  44. Liu, D.-K.; Dong, H.-F.; Liu, R.-F.; Xiao, X.-L. Baicalin Inhibits the TGF-Β1/p-Smad3 Pathway to Suppress Epithelial-Mesenchymal Transition-Induced Metastasis in Breast Cancer. Oncotarget 2020, 11, 2863–2872. [Google Scholar] [CrossRef] [PubMed]
  45. Wang, B.; Huang, T.; Fang, Q.; Zhang, X.; Yuan, J.; Li, M.; Ge, H. Bone-Protective and Anti-Tumor Effect of Baicalin in Osteotropic Breast Cancer via Induction of Apoptosis. Breast Cancer Res. Treat. 2020, 184, 711–721. [Google Scholar] [CrossRef]
  46. Ge, A.; Liu, L.; Deng, X.; Luo, J.; Xu, Y. Exploring the Mechanism of Baicalin Intervention in Breast Cancer Based on MicroRNA Microarrays and Bioinformatics Strategies. Evid. Based. Complement. Alternat. Med. 2021, 2021, 7624415. [Google Scholar] [CrossRef] [PubMed]
  47. Chen, W.; Wei, W.; Yu, L.; Zhang, X.; Huang, F.; Zheng, Q.; Wang, L.; Cai, C. Baicalin Promotes Mammary Gland Development via Steroid-like Activities. Front. Cell Dev. Biol. 2021, 9, 682469. [Google Scholar] [CrossRef]
  48. Wang, A.; Guo, D.; Cheng, H.; Jiang, H.; Liu, X.; Yun, Z. Transcriptome Sequencing Explores the Mechanism of Baicalin on Bone Cancer Pain. J. Inflamm. Res. 2021, 14, 5999–6010. [Google Scholar] [CrossRef]
  49. Li, J.; Liu, H.; Lin, Q.; Chen, H.; Liu, L.; Liao, H.; Cheng, Y.; Zhang, X.; Wang, Z.; Shen, A.; et al. Baicalin Suppresses the Migration and Invasion of Breast Cancer Cells via the TGF-β/lncRNA-MALAT1/miR-200c Signaling Pathway. Medicine 2022, 101, e29328. [Google Scholar] [CrossRef]
  50. Taiyab, A.; Choudhury, A.; Haidar, S.; Yousuf, M.; Rathi, A.; Koul, P.; Chakrabarty, A.; Islam, A.; Shamsi, A.; Hassan, M.I. Exploring MTH1 Inhibitory Potential of Thymoquinone and Baicalin for Therapeutic Targeting of Breast Cancer. Biomed. Pharmacother. 2024, 173, 116332. [Google Scholar] [CrossRef]
  51. Jia, Q.; Zhou, Y.; Song, L.; Shi, X.; Jiang, X.; Tao, R.; Wang, A.; Wu, Y.; Wei, Z.; Zhang, Y.; et al. Baicalin Reduces Chronic Stress-Induced Breast Cancer Metastasis via Directly Targeting Β2-Adrenergic Receptor. J. Pharm. Anal. 2024, 14, 100934. [Google Scholar] [CrossRef] [PubMed]
  52. So, F.V.; Guthrie, N.; Chambers, A.F.; Moussa, M.; Carroll, K.K. Inhibition of Human Breast Cancer Cell Proliferation and Delay of Mammary Tumorigenesis by Flavonoids and Citrus Juices. Nutr. Cancer 1996, 26, 167–181. [Google Scholar] [CrossRef] [PubMed]
  53. So, F.V.; Guthrie, N.; Chambers, A.F.; Carroll, K.K. Inhibition of Proliferation of Estrogen Receptor-Positive MCF-7 Human Breast Cancer Cells by Flavonoids in the Presence and Absence of Excess Estrogen. Cancer Lett. 1997, 112, 127–133. [Google Scholar] [CrossRef]
  54. Kuntz, S.; Wenzel, U.; Daniel, H. Comparative Analysis of the Effects of Flavonoids on Proliferation, Cytotoxicity, and Apoptosis in Human Colon Cancer Cell Lines. Eur. J. Nutr. 1999, 38, 133–142. [Google Scholar] [CrossRef]
  55. Tong, W.-G.; Ding, X.-Z.; Adrian, T.E. The Mechanisms of Lipoxygenase Inhibitor-Induced Apoptosis in Human Breast Cancer Cells. Biochem. Biophys. Res. Commun. 2002, 296, 942–948. [Google Scholar] [CrossRef]
  56. Po, L.S.; Chen, Z.-Y.; Tsang, D.S.C.; Leung, L.K. Baicalein and Genistein Display Differential Actions on Estrogen Receptor (ER) Transactivation and Apoptosis in MCF-7 Cells. Cancer Lett. 2002, 187, 33–40. [Google Scholar] [CrossRef] [PubMed]
  57. Markaverich, B.M.; Crowley, J.; Rodriquez, M.; Shoulars, K.; Thompson, T. Tetrahydrofurandiol Stimulation of Phospholipase A2, Lipoxygenase, and Cyclooxygenase Gene Expression and MCF-7 Human Breast Cancer Cell Proliferation. Environ. Health Perspect. 2007, 115, 1727–1731. [Google Scholar] [CrossRef]
  58. Lin, C.-W.; Yang, L.-Y.; Shen, S.-C.; Chen, Y.-C. IGF-I plus E2 Induces Proliferation via Activation of ROS-Dependent ERKs and JNKs in Human Breast Carcinoma Cells. J. Cell. Physiol. 2007, 212, 666–674. [Google Scholar] [CrossRef]
  59. Günther, S.; Ruhe, C.; Derikito, M.G.; Böse, G.; Sauer, H.; Wartenberg, M. Polyphenols Prevent Cell Shedding from Mouse Mammary Cancer Spheroids and Inhibit Cancer Cell Invasion in Confrontation Cultures Derived from Embryonic Stem Cells. Cancer Lett. 2007, 250, 25–35. [Google Scholar] [CrossRef]
  60. Lee, J.-H.; Li, Y.-C.; Ip, S.-W.; Hsu, S.-C.; Chang, N.-W.; Tang, N.-Y.; Yu, C.-S.; Chou, S.-T.; Lin, S.-S.; Lino, C.-C.; et al. The Role of Ca2+ in Baicalein-Induced Apoptosis in Human Breast MDA-MB-231 Cancer Cells through Mitochondria- and Caspase-3-Dependent Pathway. Anticancer Res. 2008, 28, 1701–1711. [Google Scholar]
  61. Androutsopoulos, V.P.; Ruparelia, K.; Arroo, R.R.J.; Tsatsakis, A.M.; Spandidos, D.A. CYP1-Mediated Antiproliferative Activity of Dietary Flavonoids in MDA-MB-468 Breast Cancer Cells. Toxicology 2009, 264, 162–170. [Google Scholar] [CrossRef]
  62. Ragazzon, P.A.; Bradshaw, T.; Matthews, C.; Iley, J.; Missailidis, S. The Characterisation of Flavone-DNA Isoform Interactions as a Basis for Anticancer Drug Development. Anticancer Res. 2009, 29, 2273–2283. [Google Scholar] [PubMed]
  63. Zhou, Q.-M.; Wang, S.; Zhang, H.; Lu, Y.-Y.; Wang, X.-F.; Motoo, Y.; Su, S.-B. The Combination of Baicalin and Baicalein Enhances Apoptosis via the ERK/P38 MAPK Pathway in Human Breast Cancer Cells. Acta Pharmacol. Sin. 2009, 30, 1648–1658. [Google Scholar] [CrossRef]
  64. Wang, L.; Ling, Y.; Chen, Y.; Li, C.-L.; Feng, F.; You, Q.-D.; Lu, N.; Guo, Q.-L. Flavonoid Baicalein Suppresses Adhesion, Migration and Invasion of MDA-MB-231 Human Breast Cancer Cells. Cancer Lett. 2010, 297, 42–48. [Google Scholar] [CrossRef]
  65. Gao, J.; Wang, Y.; Xing, Q.; Yan, J.; Senthil, M.; Akmal, Y.; Kowolik, C.M.; Kang, J.; Lu, D.M.; Zhao, M.; et al. Identification of a Natural Compound by Cell-Based Screening That Enhances Interferon Regulatory Factor-1 Activity and Causes Tumor Suppression. Mol. Cancer Ther. 2011, 10, 1774–1783. [Google Scholar] [CrossRef] [PubMed]
  66. Mai, T.T.; Moon, J.; Song, Y.; Viet, P.Q.; Van Phuc, P.; Lee, J.M.; Yi, T.-H.; Cho, M.; Cho, S.K. Ginsenoside F2 Induces Apoptosis Accompanied by Protective Autophagy in Breast Cancer Stem Cells. Cancer Lett. 2012, 321, 144–153. [Google Scholar] [CrossRef]
  67. Viola, K.; Kopf, S.; Huttary, N.; Vonach, C.; Kretschy, N.; Teichmann, M.; Giessrigl, B.; Raab, I.; Stary, S.; Krieger, S.; et al. Bay11-7082 Inhibits the Disintegration of the Lymphendothelial Barrier Triggered by MCF-7 Breast Cancer Spheroids; the Role of ICAM-1 and Adhesion. Br. J. Cancer 2013, 108, 564–569. [Google Scholar] [CrossRef]
  68. Aryal, P.; Kim, K.; Park, P.-H.; Ham, S.; Cho, J.; Song, K. Baicalein Induces Autophagic Cell Death through AMPK/ULK1 Activation and Downregulation of mTORC1 Complex Components in Human Cancer Cells. FEBS J. 2014, 281, 4644–4658. [Google Scholar] [CrossRef] [PubMed]
  69. Wang, Y.; Han, E.; Xing, Q.; Yan, J.; Arrington, A.; Wang, C.; Tully, D.; Kowolik, C.M.; Lu, D.M.; Frankel, P.H.; et al. Baicalein Upregulates DDIT4 Expression Which Mediates mTOR Inhibition and Growth Inhibition in Cancer Cells. Cancer Lett. 2015, 358, 170–179. [Google Scholar] [CrossRef]
  70. Chang, H.-T.; Chou, C.-T.; Kuo, D.-H.; Shieh, P.; Jan, C.-R.; Liang, W.-Z. The Mechanism of Ca2+ Movement in the Involvement of Baicalein-Induced Cytotoxicity in ZR-75-1 Human Breast Cancer Cells. J. Nat. Prod. 2015, 78, 1624–1634. [Google Scholar] [CrossRef]
  71. Gao, X.-Y.; Xue, X.-H.; Ma, Y.-N.; Zhang, S.-Q. Effect of Baicalein on the Expression of SATB1 in Human Breast Cancer Cells. Exp. Ther. Med. 2015, 9, 1665–1669. [Google Scholar] [CrossRef] [PubMed]
  72. Shang, D.; Li, Z.; Zhu, Z.; Chen, H.; Zhao, L.; Wang, X.; Chen, Y. Baicalein Suppresses 17-β-Estradiol-Induced Migration, Adhesion and Invasion of Breast Cancer Cells via the G Protein-Coupled Receptor 30 Signaling Pathway. Oncol. Rep. 2015, 33, 2077–2085. [Google Scholar] [CrossRef]
  73. Ma, X.; Yan, W.; Dai, Z.; Gao, X.; Ma, Y.; Xu, Q.; Jiang, J.; Zhang, S. Baicalein Suppresses Metastasis of Breast Cancer Cells by Inhibiting EMT via Downregulation of SATB1 and Wnt/β-Catenin Pathway. Drug Des. Devel. Ther. 2016, 10, 1419–1441. [Google Scholar] [CrossRef] [PubMed]
  74. Chen, Y.; Hong, D.-Y.; Wang, J.; Ling-Hu, J.; Zhang, Y.-Y.; Pan, D.; Xu, Y.-N.; Tao, L.; Luo, H.; Shen, X.-C. Baicalein, Unlike 4-Hydroxytamoxifen but Similar to G15, Suppresses 17β-Estradiol-Induced Cell Invasion, and Matrix Metalloproteinase-9 Expression and Activation in MCF-7 Human Breast Cancer Cells. Oncol. Lett. 2017, 14, 1823–1830. [Google Scholar] [CrossRef]
  75. Grigalius, I.; Petrikaite, V. Relationship between Antioxidant and Anticancer Activity of Trihydroxyflavones. Molecules 2017, 22, 2169. [Google Scholar] [CrossRef]
  76. Chen, Y.; Wang, J.; Hong, D.-Y.; Chen, L.; Zhang, Y.-Y.; Xu, Y.-N.; Pan, D.; Fu, L.-Y.; Tao, L.; Luo, H.; et al. Baicalein Has Protective Effects on the 17β-Estradiol-Induced Transformation of Breast Epithelial Cells. Oncotarget 2017, 8, 10470–10484. [Google Scholar] [CrossRef] [PubMed]
  77. Yu, X.; Cao, Y.; Tang, L.; Yang, Y.; Chen, F.; Xia, J. Baicalein Inhibits Breast Cancer Growth via Activating a Novel Isoform of the Long Noncoding RNA PAX8-AS1-N. J. Cell. Biochem. 2018, 119, 6842–6856. [Google Scholar] [CrossRef]
  78. 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. Devel. Ther. 2018, 12, 3961–3972. [Google Scholar] [CrossRef]
  79. Terabayashi, T.; Hanada, K.; Motani, K.; Kosako, H.; Yamaoka, M.; Kimura, T.; Ishizaki, T. Baicalein Disturbs the Morphological Plasticity and Motility of Breast Adenocarcinoma Cells Depending on the Tumor Microenvironment. Genes Cells 2018, 23, 466–479. [Google Scholar] [CrossRef]
  80. Marcial-Medina, C.; Ordoñez-Moreno, A.; Gonzalez-Reyes, C.; Cortes-Reynosa, P.; Perez Salazar, E. Oleic Acid Induces Migration through a FFAR1/4, EGFR and AKT-Dependent Pathway in Breast Cancer Cells. Endocr. Connect. 2019, 8, 252–265. [Google Scholar] [CrossRef]
  81. 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] [PubMed]
  82. Chen, Y.; Chen, L.; Hong, D.; Chen, Z.; Zhang, J.; Fu, L.; Pan, D.; Zhang, Y.; Xu, Y.; Gan, S.; et al. Baicalein Inhibits Fibronectin-Induced Epithelial-Mesenchymal Transition by Decreasing Activation and Upregulation of Calpain-2. Cell Death Dis. 2019, 10, 341. [Google Scholar] [CrossRef] [PubMed]
  83. Eichsteininger, J.; Kirisits, K.; Smöch, C.; Stadlbauer, C.; Nguyen, C.H.; Jäger, W.; Özmen, A.; Ecker, G.; Krupitza, G.; Krenn, L. Structural Insight into the in Vitro Anti-Intravasative Properties of Flavonoids. Sci. Pharm. 2019, 87, 23. [Google Scholar] [CrossRef]
  84. Susmitha, G.D.; Miyazato, K.; Ogura, K.; Yokoyama, S.; Hayakawa, Y. Anti-Metastatic Effects of Baicalein by Targeting STAT3 Activity in Breast Cancer Cells. Biol. Pharm. Bull. 2020, 43, 1899–1905. [Google Scholar] [CrossRef]
  85. An, L.; Dou, X.; Wang, M.; Luo, W.; Ma, Q.; Liu, X. Involvement of TNF-Alpha and IL-10 in Breast Cancer and Patient Survival. Trop. J. Pharm. Res. 2020, 19, 2033–2039. [Google Scholar] [CrossRef]
  86. He, S.; Wang, S.; Liu, S.; Li, Z.; Liu, X.; Wu, J. Baicalein Potentiated M1 Macrophage Polarization in Cancer through Targeting PI3Kγ/NF-κB Signaling. Front. Pharmacol. 2021, 12, 743837. [Google Scholar] [CrossRef] [PubMed]
  87. Kuo, Y.-T.; Liu, C.-H.; Wong, S.H.; Pan, Y.-C.; Lin, L.-T. Small Molecules Baicalein and Cinnamaldehyde Are Potentiators of Measles Virus-Induced Breast Cancer Oncolysis. Phytomedicine 2021, 89, 153611. [Google Scholar] [CrossRef]
  88. Dong, Y.; He, G.; Chen, K.; He, X.; Pan, M.; Huang, X.; Yu, X.; Xia, J. Baicalein Promotes KDM4E to Induce BICD1 and Inhibit Triple-Negative Breast Cancer Progression by Blocking PAR1 Signaling. Mol. Carcinog. 2024, 63, 1288–1302. [Google Scholar] [CrossRef]
  89. Liu, M.; Li, C.; Qu, J.; Sun, S.; Zhao, Z.; Wang, W.; Lv, W.; Zhang, Y.; Cai, Y.; Zhao, F.; et al. Baicalein Enhances Immune Response in TNBC by Inhibiting Leptin Expression of Adipocytes. Cancer Sci. 2023, 114, 3834–3847. [Google Scholar] [CrossRef]
  90. He, G.; Huang, X.; Dong, Y.; Chen, K.; He, X.; Pan, M.; Zeng, W.; Yu, X.; Xia, J. Preliminary Investigation on the Mechanism of Baicalein Regulating the Effects of Nischarin on Invasion and Apoptosis of Human Breast Cancer Cells MCF-7 through Wnt3α/β-Catenin Pathway. Int. Immunopharmacol. 2024, 143, 113262. [Google Scholar] [CrossRef]
  91. Syeda Abida Ejaz, M.; Sarfraz, M.; Tanveer, A.; Wani, T.; Ruby, S.; Zargar, M.; Arafat, C. Evaluation of Cytotoxic Activity and Apoptosis-Inducing Potential of 5,6,7-Trihydroxyflavone against Breast Cancer and Cervical Cancer Cell Lines. J. Biol. Regul. Homeost. Agents 2024, 38, 303–317. [Google Scholar]
  92. He, T.-M.; Liu, J.-X.; Duan, C.-C.; Li, X.-F.; Zhang, J.-Y. Effective Material Basis and Mechanism Analysis of Compound Banmao Capsule against Tumors Using Integrative Network Pharmacology and Molecular Docking. Evid. Based. Complement. Alternat. Med. 2021, 2021, 6653460. [Google Scholar] [CrossRef]
  93. Qi, X.; Xu, H.; Zhang, P.; Chen, G.; Chen, Z.; Fang, C.; Lin, L. Investigating the Mechanism of Scutellariae barbata Herba in the Treatment of Colorectal Cancer by Network Pharmacology and Molecular Docking. Evid. Based. Complement. Alternat. Med. 2021, 2021, 3905367. [Google Scholar] [CrossRef] [PubMed]
  94. Cao, X.; Xin, G.; Fang, B. Molecular Docking and Molecular Dynamics Simulation Studies on the Effect of Baicalein on Breast and Ovarian Cancers. Trop. J. Pharm. Res. 2023, 22, 1395–1402. [Google Scholar] [CrossRef]
  95. Rathi, A.; Khan, A.; Haider, S.; Roy, S.; Taiyab, A.; Mahendru, S.; Hussain, A.; Chakrabarty, A.; Islam, A.; Imtaiyaz Hassan, M.; et al. Exploring the Potential of Baicalin and Resveratrol as PIM-1 Kinase Inhibitors: Therapeutic Targeting of Prostate and Breast Cancers. J. Mol. Liq. 2024, 396, 124026. [Google Scholar] [CrossRef]
  96. Wang, C.-Z.; Li, X.-L.; Wang, Q.-F.; Mehendale, S.R.; Yuan, C.-S. Selective Fraction of Scutellaria baicalensis and Its Chemopreventive Effects on MCF-7 Human Breast Cancer Cells. Phytomedicine 2010, 17, 63–68. [Google Scholar] [CrossRef]
  97. Yu, C.; Zhang, Z.; Zhang, H.; Zhen, Z.; Calway, T.; Wang, Y.; Yuan, C.-S.; Wang, C.-Z. Pretreatment of Baicalin and Wogonoside with Glycoside Hydrolase: A Promising Approach to Enhance Anticancer Potential. Oncol. Rep. 2013, 30, 2411–2418. [Google Scholar] [CrossRef]
  98. Park, J.R.; Lee, M.C.; Moon, S.-C.; Kim, J.; Ha, K.-T.; Park, E.J.; Hong, C.; Seo, B.-D.; Kim, B.J. Scutellaria baicalensis Georgi Induces Caspase-Dependent Apoptosis via Mitogen Activated Protein Kinase Activation and the Generation of Reactive Oxygen Species Signaling Pathways in MCF-7 Breast Cancer Cells. Mol. Med. Rep. 2017, 16, 2302–2308. [Google Scholar] [CrossRef]
  99. Du, J.; Wang, L.; Huang, X.; Zhang, N.; Long, Z.; Yang, Y.; Zhong, F.; Zheng, B.; Lan, W.; Lin, W.; et al. Shuganning Injection, a Traditional Chinese Patent Medicine, Induces Ferroptosis and Suppresses Tumor Growth in Triple-Negative Breast Cancer Cells. Phytomedicine 2021, 85, 153551. [Google Scholar] [CrossRef] [PubMed]
  100. Song, Y.-C.; Lee, D.-Y.; Yeh, P.-Y. A Novel Chinese Herbal and Corresponding Chemical Formula for Cancer Treatment by Targeting Tumor Maintenance, Progression, and Metastasis. Front. Pharmacol. 2022, 13, 907826. [Google Scholar] [CrossRef] [PubMed]
  101. Qi, Y.; Wu, X.-J.; Shi, J.-B.; Shi, X.-W.; Zhao, N.; Xiong, Y.; Wang, L.-P. Sanhuang Xiexin Decoction Ameliorates TNBC by Modulating JAK2-STAT3 and Lipid Metabolism. Chin. J. Integr. Med. 2024, 30, 1080–1089. [Google Scholar] [CrossRef] [PubMed]
  102. Zhao, Z.; Zhu, Y.; Long, F.; Ma, Y.; Tang, Q.; Wang, T. Exploring the Potential of Huangqin Tang in Breast Cancer Treatment Using Network Pharmacological Analysis and Experimental Verification. BMC Complement. Med. Ther. 2024, 24, 221. [Google Scholar] [CrossRef]
  103. Chen, H.; Gao, Y.; Wu, J.; Chen, Y.; Chen, B.; Hu, J.; Zhou, J. Exploring Therapeutic Potentials of Baicalin and Its Aglycone Baicalein for Hematological Malignancies. Cancer Lett. 2014, 354, 5–11. [Google Scholar] [CrossRef] [PubMed]
  104. Neves, M.P.; Camões, A.; de São José Nascimento, M.; Cidade, H.; Sousa, M.E.; Pinto, M.M.M. Effects of a Prenyl-Baicalein Derivative on ER (+) MCF-7 and ER (−) MDA-MB-231 Breast Tumor Cell Lines. Med. Chem. Res. 2012, 21, 3154–3160. [Google Scholar] [CrossRef]
  105. Wang, N.; Ren, D.; Deng, S.; Yang, X. Differential Effects of Baicalein and Its Sulfated Derivatives in Inhibiting Proliferation of Human Breast Cancer MCF-7 Cells. Chem. Biol. Interact. 2014, 221, 99–108. [Google Scholar] [CrossRef]
  106. An, D.; Song, Z.; Yi, Y.; Zhang, Q.; Liu, J.; Zhang, Y.; Zhou, J.; Zhao, G.; Cong, D.; Li, N.; et al. Oroxylin A, a Methylated Metabolite of Baicalein, Exhibits a Stronger Inhibitory Effect than Baicalein on the CYP1B1-Mediated Carcinogenic Estradiol Metabolite Formation: Oroxylin A Has a Stronger Inhibition than Baicalein for CYP1B1. Phytother. Res. 2019, 33, 1033–1043. [Google Scholar] [CrossRef]
  107. Marzec, E.; Świtalska, M.; Winiewska-Szajewska, M.; Wójcik, J.; Wietrzyk, J.; Maciejewska, A.M.; Poznański, J.; Mieczkowski, A. The Halogenation of Natural Flavonoids, Baicalein and Chrysin, Enhances Their Affinity to Human Protein Kinase CK2. IUBMB Life 2020, 72, 1250–1261. [Google Scholar] [CrossRef]
  108. Yasuda, N.; Ali, S.; Aman, A.; Krusong, K.; Herfindo, N.; Chavasiri, W.; Choowongkomon, K.; Wolschann, P.; Mahalapbutr, P.; Rungrotmongkol, T.; et al. In Vitro and in Silico Studies of the Inclusion Complexation of 8-Bromobaicalein with β-Cyclodextrins. J. Mol. Graph. Model. 2024, 132, 108840. [Google Scholar] [CrossRef]
  109. Daina, A.; Michielin, O.; Zoete, V. SwissADME: A Free Web Tool to Evaluate Pharmacokinetics, Drug-Likeness and Medicinal Chemistry Friendliness of Small Molecules. Sci. Rep. 2017, 7, 42717. [Google Scholar] [CrossRef]
  110. Franek, K.J.; Zhou, Z.; Zhang, W.-D.; Chen, W.Y. In Vitro Studies of Baicalin Alone or in Combination with Salvia miltiorrhiza Extract as a Potential Anti-Cancer Agent. Int. J. Oncol. 2005, 26, 217–224. [Google Scholar] [CrossRef]
  111. Chang, W.-T.; Li, J.; Haung, H.-H.; Liu, H.; Han, M.; Ramachandran, S.; Li, C.-Q.; Sharp, W.W.; Hamann, K.J.; Yuan, C.-S.; et al. Baicalein Protects against Doxorubicin-Induced Cardiotoxicity by Attenuation of Mitochondrial Oxidant Injury and JNK Activation. J. Cell. Biochem. 2011, 112, 2873–2881. [Google Scholar] [CrossRef] [PubMed]
  112. Zhan, Y.; Chen, Y.; Liu, R.; Zhang, H.; Zhang, Y. Potentiation of Paclitaxel Activity by Curcumin in Human Breast Cancer Cell by Modulating Apoptosis and Inhibiting EGFR Signaling. Arch. Pharm. Res. 2014, 37, 1086–1095. [Google Scholar] [CrossRef]
  113. An, H.; Yu, X.; Xiang, C.; Zhang, Y.; Xia, J.; Wang, Y. Baicalein and U0126 Suppress Human Breast Cancer Cell Line MCF-7 through Regulating MAPK Signaling Pathway. Int. J. Clin. Exp. Pathol. 2016, 9, 10266–10273. [Google Scholar]
  114. Xi, Q.; Qi, H.; Li, Y.; Xi, Y.; Zhang, L. Low Frequency Ultrasound Combined with Baicalein Can Reduced the Invasive Capacity of Breast Cancer Cells by down Regulating the Expression of MMP-2, MMP-9, and u-PA. Transl. Cancer Res. 2016, 5, 838–844. [Google Scholar] [CrossRef]
  115. Talik Sisin, N.N.; Abdul Razak, K.; Zainal Abidin, S.; Che Mat, N.F.; Abdullah, R.; Ab Rashid, R.; Khairil Anuar, M.A.; Rahman, W.N. Synergetic Influence of Bismuth Oxide Nanoparticles, Cisplatin and Baicalein-Rich Fraction on Reactive Oxygen Species Generation and Radiosensitization Effects for Clinical Radiotherapy Beams. Int. J. Nanomed. 2020, 15, 7805–7823. [Google Scholar] [CrossRef]
  116. Zeng, A.; Liang, X.; Zhu, S.; Liu, C.; Luo, X.; Zhang, Q.; Song, L. Baicalin, a Potent Inhibitor of NF-κB Signaling Pathway, Enhances Chemosensitivity of Breast Cancer Cells to Docetaxel and Inhibits Tumor Growth and Metastasis Both in Vitro and in Vivo. Front. Pharmacol. 2020, 11, 879. [Google Scholar] [CrossRef]
  117. Lin, M.-Y.; Cheng, W.-T.; Cheng, H.-C.; Chou, W.-C.; Chen, H.-I.; Ou, H.-C.; Tsai, K.-L. Baicalin Enhances Chemosensitivity to Doxorubicin in Breast Cancer Cells via Upregulation of Oxidative Stress-Mediated Mitochondria-Dependent Apoptosis. Antioxidants 2021, 10, 1506. [Google Scholar] [CrossRef] [PubMed]
  118. Shehatta, N.H.; Okda, T.M.; Omran, G.A.; Abd-Alhaseeb, M.M. Baicalin; a Promising Chemopreventive Agent, Enhances the Antitumor Effect of 5-FU against Breast Cancer and Inhibits Tumor Growth and Angiogenesis in Ehrlich Solid Tumor. Biomed. Pharmacother. 2022, 146, 112599. [Google Scholar] [CrossRef]
  119. Sisin, N.N.T.; Mat, N.F.C.; Rashid, R.A.; Dollah, N.; Razak, K.A.; Geso, M.; Algethami, M.; Rahman, W.N. Natural Baicalein-Rich Fraction as Radiosensitizer in Combination with Bismuth Oxide Nanoparticles and Cisplatin for Clinical Radiotherapy. Int. J. Nanomed. 2022, 17, 3853–3874. [Google Scholar] [CrossRef]
  120. Ibrahim, H.A.; Abd El-Alim, A.E.-A.F.; El-Hafeez, M.A.; Metwally, M.M.M.; Khamis, T.; Galal, A.A.A. Baicalein Prevents Capecitabine-Induced Heart Damage in Female Wistar Rats and Enhances Its Anticancer Potential in MCF-7 Breast Cancer Cells. Life Sci. 2023, 319, 121523. [Google Scholar] [CrossRef]
  121. 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] [PubMed]
  122. Bernasinska-Slomczewska, J.; Hikisz, P.; Pieniazek, A.; Koceva-Chyla, A. Baicalin and Baicalein Enhance Cytotoxicity, Proapoptotic Activity, and Genotoxicity of Doxorubicin and Docetaxel in MCF-7 Breast Cancer Cells. Molecules 2024, 29, 2503. [Google Scholar] [CrossRef]
  123. Awajan, D.; Abu-Humaidan, A.H.A.; Talib, W.H. Study of the Antitumor Activity of the Combination Baicalin and Epigallocatechin Gallate in a Murine Model of Vincristine-Resistant Breast Cancer. Farmatsiia 2024, 71, 1–20. [Google Scholar] [CrossRef]
  124. Zhuo, Y.; Zhao, Y.-G.; Zhang, Y. Enhancing Drug Solubility, Bioavailability, and Targeted Therapeutic Applications through Magnetic Nanoparticles. Molecules 2024, 29, 4854. [Google Scholar] [CrossRef]
  125. Kavithaa, K.; Paulpandi, M.; Padma, P.R.; Sumathi, S. Induction of Intrinsic Apoptotic Pathway and Cell Cycle Arrest via Baicalein Loaded Iron Oxide Nanoparticles as a Competent Nano-Mediated System for Triple Negative Breast Cancer Therapy. RSC Adv. 2016, 6, 64531–64543. [Google Scholar] [CrossRef]
  126. Lee, D.; Ko, W.-K.; Hwang, D.-S.; Heo, D.N.; Lee, S.J.; Heo, M.; Lee, K.-S.; Ahn, J.-Y.; Jo, J.; Kwon, I.K. Use of Baicalin-Conjugated Gold Nanoparticles for Apoptotic Induction of Breast Cancer Cells. Nanoscale Res. Lett. 2016, 11, 381. [Google Scholar] [CrossRef] [PubMed]
  127. Meng, L.; Xia, X.; Yang, Y.; Ye, J.; Dong, W.; Ma, P.; Jin, Y.; Liu, Y. Co-Encapsulation of Paclitaxel and Baicalein in Nanoemulsions to Overcome Multidrug Resistance via Oxidative Stress Augmentation and P-Glycoprotein Inhibition. Int. J. Pharm. 2016, 513, 8–16. [Google Scholar] [CrossRef]
  128. 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]
  129. Kavithaa, K.; Sumathi, S.; Padma, P.R. Intracellular Uptake of PEG-Funtionalized Baicalein Loaded Iron Oxide Nanoparticles Regulates Apoptotic Genes in Triple Negative Breast Cancer Cells: Mitochondrial Pathway Targeted Therapy for Breast Cancer. J. Cluster Sci. 2017, 28, 2057–2073. [Google Scholar] [CrossRef]
  130. El-Gogary, R.; Gaber, S.A.A.; Nasr, M. Polymeric Nanocapsular Baicalin: Chemometric Optimization, Physicochemical Characterization and Mechanistic Anticancer Approaches on Breast Cancer Cell Lines. Sci. Rep. 2019, 9, 11064. [Google Scholar] [CrossRef]
  131. Mi, X.; Hu, M.; Dong, M.; Yang, Z.; Zhan, X.; Chang, X.; Lu, J.; Chen, X. Folic Acid Decorated Zeolitic Imidazolate Framework (ZIF-8) Loaded with Baicalin as a Nano-Drug Delivery System for Breast Cancer Therapy. Int. J. Nanomed. 2021, 16, 8337–8352. [Google Scholar] [CrossRef] [PubMed]
  132. Meng, F.; Liu, F.; Lan, M.; Zou, T.; Li, L.; Cai, T.; Cai, Y. Preparation and Evaluation of Folate-Modified Albumin Baicalin-Loaded Nanoparticles for the Targeted Treatment of Breast Cancer. J. Drug Deliv. Sci. Technol. 2021, 65, 102603. [Google Scholar] [CrossRef]
  133. Srivastava, S.; Singh, S.; Saraf, S.A.; Chourasia, M.K.; Mathew, J.; Pandey, A.C. Encapsulation of Baicalein in Cinnamon Essential Oil Nanoemulsion for Enhanced Anticancer Efficacy against MDA-MB-231 Cells. Bionanoscience 2021, 11, 1049–1060. [Google Scholar] [CrossRef]
  134. Wang, T.; Yang, J.; Kang, H.; Zhang, L.; Chen, H. Facile Preparation of a Novel Hyaluronic Acid-Modified Metal-Polyphenol Photothermal Nanoformulation for Tumor Therapy. Int. J. Biol. Macromol. 2022, 222, 3066–3076. [Google Scholar] [CrossRef] [PubMed]
  135. Liu, F.; Lan, M.; Ren, B.; Li, L.; Zou, T.; Kong, Z.; Fan, D.; Cai, T.; Cai, Y. Baicalin-Loaded Folic Acid-Modified Albumin Nanoparticles (FA-BSANPs/BA) Induce Autophagy in MCF-7 Cells via ROS-Mediated P38 MAPK and Akt/mTOR Pathway. Cancer Nanotechnol. 2022, 13, 2. [Google Scholar] [CrossRef]
  136. Gharari, Z.; Hanachi, P.; Sadeghinia, H.; Walker, T.R. Eco-Friendly Green Synthesis and Characterization of Silver Nanoparticles by Scutellaria multicaulis Leaf Extract and Its Biological Activities. Pharmaceuticals 2023, 16, 992. [Google Scholar] [CrossRef]
Figure 1. Chemical structures of (a) baicalin and (b) baicalein.
Figure 1. Chemical structures of (a) baicalin and (b) baicalein.
Cimb 47 00181 g001
Figure 2. Visualization of bibliometric analysis of baicalin and baicalein-related research: (a) annual publication trend, (b) distribution of papers by language, (c) classification of papers by type, and (d) global contributor map.
Figure 2. Visualization of bibliometric analysis of baicalin and baicalein-related research: (a) annual publication trend, (b) distribution of papers by language, (c) classification of papers by type, and (d) global contributor map.
Cimb 47 00181 g002
Figure 3. Keyword co-occurrence network visualization generated using VOSviewer.
Figure 3. Keyword co-occurrence network visualization generated using VOSviewer.
Cimb 47 00181 g003
Figure 4. The PRISMA flowchart for the selection and screening of the articles.
Figure 4. The PRISMA flowchart for the selection and screening of the articles.
Cimb 47 00181 g004
Figure 5. Chemical structures of baicalein derivatives.
Figure 5. Chemical structures of baicalein derivatives.
Cimb 47 00181 g005aCimb 47 00181 g005b
Figure 6. An overview of the key mechanisms behind the anticancer effects of baicalin and baicalein. R represents either a hydrogen atom (H) for baicalein or a glucuronide moiety for baicalin.
Figure 6. An overview of the key mechanisms behind the anticancer effects of baicalin and baicalein. R represents either a hydrogen atom (H) for baicalein or a glucuronide moiety for baicalin.
Cimb 47 00181 g006
Table 1. The five scientific publications most frequently cited in the Scopus database that reference baicalin and baicalein.
Table 1. The five scientific publications most frequently cited in the Scopus database that reference baicalin and baicalein.
RankAuthorsTitleYearJournalTotal CitationReference
1.Cushnie, T.P.T.; Lamb, A.J.Antimicrobial Activity of Flavonoids2005International Journal of Antimicrobial Agents3287[31]
2.Del Rio, D.; Rodriguez-Mateos, A.; Spencer, J.P.E.; Tognolini, M.; Borges, G.; Crozier, A.Dietary (Poly)phenolics in Human Health: Structures, Bioavailability, and Evidence of Protective Effects Against Chronic Diseases2013Antioxidants and Redox Signaling2067[32]
3.Wu, C.; Liu, Y.; Yang, Y.; Zhang, P.; Zhong, W.; Wang, Y.; Wang, Q.; Xu, Y.; Li, M.; Li, X.; et al.Analysis of Therapeutic Targets for SARS-CoV-2 and Discovery of Potential Drugs by Computational Methods.2020Acta Pharmaceutica Sinica B1712[33]
4.Formica, J.V.; Regelson, W.Review of the Biology of Quercetin and Related Bioflavonoids.1995Food and Chemical Toxicology1617[34]
5.Zhang, Q.-W.; Lin, L.-G.; Ye, W.-C.Techniques for Extraction and Isolation of Natural Products: A Comprehensive Review2018Chinese Medicine1331[35]
Table 2. Overview of studies on the anticancer potential of baicalin.
Table 2. Overview of studies on the anticancer potential of baicalin.
Cell LinesAnimal ModelsTested ConcentrationsThe Outcome of the StudyReference
PC-3, DU145, LNCaP, MCF-7, HL-60, and NB4-0.1–20 µMThe MCF-7 cell line was insensitive to growth inhibition by baicalin[36]
MCF-7-50–400 µMInhibition of MCF-7 cell proliferation in a dose- and time-dependent manner (IC50 = 206 µM after 24 h)
At 200 µM baicalin induced potent cytotoxicity and morphological changes indicative of cell death
Cells arrested in G0/G1 phase and chromatin condensation confirmed apoptosis
Upregulation of protein p53 and the bax gene
[37]
MCF-10A and MDA-MB-231-2–5 µMInhibition of NF-κB activation and TGF-β1-induced EMT in both cell lines
Suppression of TGF-β1-induced migration and colony formation in MDA-MB-231 breast cancer cells
[38]
MCF-7-10–160 µMBaicalin at 160 µM significantly reduced the survival of MCF-7 cells, and concentrations of 10–80 µM had no significant effect
Inhibition of MCF-7 cell migration by upregulating miR-200b and E-cadherin
[39]
MDA-MB-231 and 4T1-10–100 µMBaicalin (up to 100 µM) did not affect the viability of MDA-MB-231 and 4T1 cells
Baicalin suppressed breast cancer metastasis by inhibiting migration, invasion, and EMT through the β-catenin pathway
[40]
MCF-7, MDA-MB-231, and MCF-10Female BALB/c nude mice (5 weeks old)20–30 µMInhibition of breast cancer cell proliferation, invasion, and migration by inducing G1/S arrest and suppressing the NF-κB pathway
Reduction of tumor growth in vivo and modulated inflammatory responses
[41]
MDA-MB-231-10–40 µg/mLInhibition of MDA-MB-231 cell growth, proliferation, migration, and invasion, with effects increasing at higher concentrations (40 μg/mL was the most effective)
Reducing Vimentin, β-catenin, c-Myc, and MMP-7 mRNA expression while increasing E-cadherin expression
[42]
MCF-10A, MCF-7, and MDA-MB-231-25–200 µMInhibition of viability, migration, and invasion of breast cancer cells, but no cytotoxicity to normal MCF-10A cells
Inhibition of breast cancer progression by upregulating miR-338-3p
[43]
SK-BR-2 and MCF-7Female BALB/c-null and normal mice (6 weeks old)100 mg/kgBaicalin bonded directly to TGF-β1 and downregulated its expression, along with p-Smad3 and vimentin, while upregulating E-cadherin
Baicalin suppressed tumor growth and metastasis, with upregulated E-cadherin and downregulated TGF-β1, vimentin, and p-Smad3 in tumors
[44]
MDA-MB-231 and
MCF-7
-0.25–100 nMInduction of apoptosis in breast cancer cells (MDA-MB-231, MCF-7) by increasing cytochrome c release, DNA fragmentation, and caspase-3, -8, and -9 activity
Inhibition of the mTOR pathway, reducing phosphorylation of mTOR and p70 S6 kinase—Baicalin had minimal effects on osteoblasts and bone cells
[45]
MCF-7-50–200 µMInhibition of MCF-7 cell proliferation in a time- and concentration-dependent manner (0–200 µM, 24–72 h)
At 150 µM baicalin, miRNA microarray analysis identified 92 upregulated and 35 downregulated miRNAs
[46]
Eph4, MDA-MB-231, ZR-75-1, and 293TC57BL/6 and Actin-DsRed mice (3 weeks old)50–200 mg/kgBaicalin suppressed luminal breast cancer (ZR-75-1) growth by downregulating ESR1 but promoted triple-negative breast cancer (MDA-MB-231) growth by upregulating PROCR
Baicalin exhibited steroid hormone-like activity, regulating genes involved in Hippo signaling and cell cycle while suppressing breast cancer-related genes
[47]
MADB-106Sprague-Dawley (SD) rats30 mg/kgReversing bone loss in bone cancer pain rats
Inhibition of osteoclast activation and reduction of inflammation
Downregulating cancer-related genes and modulating pathways like NF-κB and TNF signaling.
[48]
MDA-MB-231-12.5–50 µMReduction of cell viability of MDA-MB-231 cells after 48–72 h of treatment
Inhibition of migration and invasion of breast cancer cells
Downregulation of the TGF-β/ZEB1 pathway, reducing TGF-β1, ZEB1, and N-cadherin (mesenchymal markers) while upregulating E-cadherin (epithelial marker) at both mRNA and protein levels
Downregulation of lncRNA-MALAT1 and upregulated miR-200c
[49]
MCF-7-0–200 µMAnti-proliferative and cytotoxic activity against breast cancer cells (IC50 = 34.8 μM)
Baicalin triggered apoptosis and heightened ROS production in MCF7 cells
[50]
MCF-7, MDA-MB-231, MCF-7 ADRB2^OEFemale BALB/c and C57BL/6J mice (6 weeks old)25–100 µMBaicalin suppressed epinephrine-induced migration and invasion of breast cancer cells in wound healing, transwell, and 3D spheroid assays
Protection against chronic stress-promoted breast cancer metastasis by binding to β2-AR and blocking its activation on tumors
[51]
Table 3. Summarized data on baicalein application in breast cancer models.
Table 3. Summarized data on baicalein application in breast cancer models.
Cell LinesAnimal ModelsTested ConcentrationsThe Outcome of the StudyReference
MDA-MB-435Female Sprague-Dawley rats0–250 µg/mLInhibition of MDA-MB-435 breast cancer cell proliferation in vitro, with an IC50 of 5.9 µg/mL[52]
MCF-7-0–2 mg/mLBaicalein inhibits the proliferation of MCF-7 breast cancer cells through a non-estrogenic mechanism (IC₅₀ = 5.3 µg/mL)[53]
Caco-2, HT-29, LLC-PK1, and MCF-7-1–1000 µMMCF-7 breast cancer cells were less sensitive to baicalein, requiring 6.6-fold higher concentrations to achieve 50% growth inhibition compared to intestinal cells[54]
MCF-7 and MDA-MB-231-1.25–20 µMInhibition of the proliferation of both MDA-MB-231 and MCF-7 breast cancer cells
Induction of apoptosis through mitochondrial pathways, caspase activation, and modulation of Bcl-2 family proteins
[55]
MCF-7-10–100 µMInduction of apoptosis in MCF-7 cells at 10 µM
Baicalein antagonized estradiol-induced ER transactivation, showing consistent antiestrogenic effects
[56]
MCF-7-5 µMInhibition of the proliferative effects of THF-diols and E2 on MCF-7 breast cancer cells by targeting LOX-12[57]
MCF-7-25–50 µMBaicalein, but not its glycoside inhibited E2/IGF-I-induced proliferation, c-Jun expression, and ROS production[58]
4T1-1–100 µMDemonstration of anti-proliferative, anti-metastatic, and antioxidant properties in 4T1 tumor spheroids[59]
MDA-MB-231-25–100 µMInduction of apoptosis in MDA-MB-231 breast cancer cells through ROS production, mitochondrial dysfunction, Ca2+ signaling, and regulation of apoptotic proteins[60]
MDA-MB-468 and MCF-10A-10 µMAntiproliferative effects in breast cancer cells (MDA-MB-468), Baicalein was not metabolized by CYP1 enzymes[61]
MCF-7 and CCRFCEM leukemia cells-1–1000 µMBaicalein (and baicalin) induced G1-phase arrest in MCF-7 and CCRFCEM cells—Moderate anticancer activity (IC50 = 69.6 μM for baicalein and 78.8 μM for baicalin against MCF-7 cells)
Baicalein-induced DNA damage in MCF-7 cells (Baicalin had no effect)
[62]
MCF-7 and MDA-MB-231-25–400 µMThe combination of baicalin and baicalein synergistically inhibited breast cancer cell growth, induced apoptosis, and caused cell cycle arrest by activating the ERK/p38 MAPK pathway[63]
MDA-MB-231-2–50 µMBaicalein demonstrates potent anti-metastatic effects by inhibiting adhesion, motility, and invasion of MDA-MB-231 breast cancer cells
Suppression of the activity and expression of MMP-2 and MMP-9, likely through the downregulation of the MAPK signaling pathway
[64]
AGS, MDA468, BT549, SKBR3, C3L5, and MDA468-C23Female SCID-Bg mice (Charles River, 6 weeks old)0.5–10 µMBaicalein enhanced IRF-1 activity, upregulated tumor-suppressive pathways, and inhibited cancer cell growth both in vitro and in vivo[65]
MCF-7 and breast cancer stem cells (CSCs)-0–120 µMBaicalein exhibits limited effectiveness against breast CSCs compared to ginsenoside F2[66]
MCF-7-100 µMInhibition of ALOX12/15 and reduction of 12(S)-HETE production[67]
PC-3, MDA-MB-231 and DU145-2.5–20 µg/mLInduction of autophagy, dysfunction of mitochondria, and cell cycle arrest[68]
MDA-MB-468, SKBR3, Hs578T, and BT549SCID-Beige mice5–80 µMBaicalein-induced DDIT4 (DNA Damage-Inducible Transcript 4) expression in multiple breast cancer cell lines
Baicalein treatment significantly suppressed tumor growth without toxicity in an orthotopic mouse model of triple-negative breast cancer
[69]
ZR-75-1-10–40 µMBaicalein increased intracellular Ca2+ concentration through Ca2+ release from the ER and Ca2+ entry via SOCCs, leading to ROS production, caspase activation, and apoptosis in ZR-75-1 cells[70]
MDA-MB-231-5–80 µMInhibition of proliferation and migration of MDA-MB-231 cells
Downregulation of SATB1 protein expression
[71]
MCF-7
and SK-BR-3
-5–50 µMInhibition of E2-induced migration, adhesion, invasion, and GPR30 signaling in breast cancer cells[72]
MCF-10A, MCF7, SKBR3, and MDA-MB-231Female Balb/c nude mice (4–8 weeks old)10–40 µMSuppression of proliferation, migration, and invasion of cancer cells
Downregulation of SATB1 and the Wnt/β-catenin pathway
Reversing EMT and reducing metastasis in vivo
[73]
MCF-7-10 µMInhibition of E2-induced invasion and MMP-9 activity in MCF-7 breast cancer cells by targeting GPR30 signaling[74]
A549, MCF-7, and U87-3.125–100 µMModerate anti-proliferative activity against the MCF-7 breast cancer cell line (EC50 = 26.1 µM)[75]
MCF-10A and MCF-12AFemale non-obese diabetic/severe combined
immunodeficiency (NOD/SCID) mice (6–9 weeks old)
2–8 µMBaicalein effectively prevents estradiol (E2)-induced neoplastic transformation in mammary epithelial cells by inhibiting cell growth, migration, invasion, and tumorigenesis, and by blocking ERα and GPR30 signaling pathways[76]
MDA-MB-231 and MCF-7Seventy-six pairs of breast cancer and adjacent normal tissues from patients after surgery50–400 µMUpregulation of PAX8-AS1-N, resulting in the inhibition of tumor growth and progression by modulating miR-17-5p and its downstream targets[77]
MCF-7 and MDA-MB-231Female BALB/c nude mice (3–6 weeks old)10–40 µMInhibition of proliferation and induction of apoptosis and autophagy
Suppression of the PI3K/AKT/mTOR signaling pathway
[78]
MDA-MB-231, MCF-10A, and HeLa-2–50 µMInhibition of breast cancer cell motility by disrupting paracrine interactions mediated by laminin-332[79]
MDA-MB-231 and MCF-7-20 µMBaicalein inhibited oleic acid-induced migration and invasion in breast cancer cells
Blocking of AKT2 and FAK activation
Reduction of 12(S)-HETE secretion through 12-LOX inhibition
Altering focal contact formation
[80]
MDA-MB-231 and MDA-MB-231/IR-20–100 µMReversion of IFIT2 expression, suppression of stem cell-like properties, and induction of apoptosis[81]
MCF-10AFemale MMTV-PyMT transgenic mice
(about 3 weeks old)
2.5–10 µMInhibition of fibronectin-induced migration, invasion, and epithelial-mesenchymal transition in breast epithelial cells by suppressing calpain-2 activity and modulating Ca2+ levels and ERK signaling
In vivo, baicalein delays tumor progression, reduces metastasis, and prolongs survival in a breast cancer mouse model
[82]
MCF-7 and MCF-10A-12.5–200 µMInhibition of MCF-7 breast cancer cell proliferation and induction of apoptosis through a copper-dependent mechanism involving ROS generation and mitochondrial dysfunction[26]
MCF-7-10–75 µMExhibition of weak activity in the CCID assay (IC50 = 130.2 µM)[83]
4T1 and MDA-MB-231-10–100 µMBaicalein suppressed STAT3 activity by inhibiting its phosphorylation and reduces IL-6 production, leading to anti-proliferative, cytotoxic, and anti-metastatic effects[84]
MCF-7-20 µMAntiproliferative effect on MCF-7 cells
Inhibition of TNF-α and IL-10
[85]
B16-F10, 4T1, THP-1-20–80 µMBaicalein exerted its anti-tumor effects by promoting M1 macrophage polarization through the NF-κB/TNF-α signaling pathway[86]
MCF-7-20–84 µMExhibition of the cytotoxicity in MCF-7 cells (CC₅₀ = 56.46 µM)—Baicalein inactivated free virus particles and modestly reduced MV infection when added concurrently or immediately post-infection[87]
MDA-MB-231, BT549, MDA-MB-468, MCF-7, ZR-75-1, T47D, and MCF-10ABALB/c nude mice (6–8 weeks old)10–100 µMAnti-tumor effects by targeting the KDM4E/BICD1/PAR1 signaling pathway[88]
MDA-MB-
231,
BT549, 4T1, and 3T3-L1
Female BALB/C mice (4 weeks old)10–40 µMBaicalein significantly slowed tumor growth and downregulated PD-L1, LEP, SREBP1, and p-STAT3 in tumors[89]
MCF-7-25–400 µMInhibition of MCF-7 cells viability, induction of apoptosis, and suppression of the migration and invasion by modulating the Wnt3α/β-catenin signaling pathway
Upregulation of Nischarin
[90]
MDA-MB-231, MCF-7, and HeLa-3.23–22.2 µMInduction of LDH release and increased caspase-3-activity, indicating cell membrane damage and apoptosis[91]
Table 4. Summary of the studies investigating plant extracts containing baicalein and baicalin for anticancer activity in breast cancer models.
Table 4. Summary of the studies investigating plant extracts containing baicalein and baicalin for anticancer activity in breast cancer models.
Cell LinesAnimal ModelsPlant/MaterialsBaicalein ContentBaicalin ContentThe Outcome of the StudyReference
U87-MG, U251, MDA-MB-231, HMEC, and PC3-thirteen Scutellaria species0.21–2.34 μg/mg extract0.92–25.34 μg/mg extractScutellaria species inhibited the proliferation of MDA-MB-231 cells and induced apoptosis[19]
MDA-MB-435S-Phyllanthus amarus (whole plant)presentnot specifiedThe application of the aqueous extract mitigated Cr(VI)-induced oxidative toxicity in MDA-MB-435S in a dosage-dependent manner[20]
MCF-7-the roots of S. baicalensis40.54% in the baicalin-deprived fraction, 0.00–14.24% in other fractions0.51–19.58%The presence of baicalin in the isolated fractions reduced their antiproliferative effects
Baicalin-deprived fraction primarily contained baicalein, and wogonin demonstrated the strongest anticancer activity, primarily through cell cycle arrest and apoptosis induction
[96]
HCT-116 and MCF-7-S. baicalensis root powder8.5–68.6%1.0–43.3%Cellulase treatment significantly enhanced the antiproliferative potential of S. baicalensis extracts against cancer cells and promoted apoptosis through S-phase cell cycle arrest[97]
MCF-7, HeLa, HT-29, and MRC-5-black (Tuber aestivum Vittad.) and white (T. magnatum Pico)
truffles
2.01–15.73 µg/g dw in T. aestivum-LC-MS/MS analysis identified 14 phenolic compounds, with T. aestivum rich in p-hydroxybenzoic acid, baicalein, and kaempferol, and T. magnatum in epicatechin and catechin
T. aestivum and T. magnatum extracts showed significant growth inhibition in MCF-7 and HeLa cells
[21]
MCF-7-S. baicalensis Georgi extractnot specifiedpresentThe extract exhibited potent anti-cancer effects on MCF-7 cells through mechanisms involving apoptosis induction via mitochondrial pathways, caspase activation, and ROS generation[98]
4T1-leaves of Vernonia amygdalina Delilepresentnot presentEthyl acetate fraction of V. amygdalina Delile demonstrated anticancer potential by inducing cell cycle arrest, apoptosis/necrosis, and inhibiting PI3K/mTOR pathways[22]
SK-BR-3, MDA-MB-231, MDA-MB-468, BT-549, MCF10A, 786-O, A2780, HCT116, HepG2, A549, Lo2, and WPMY-1, U-87 MG, and HT1080Female nude mice (4–6 weeks old)Shuganning injectionnot specifiednot specifiedIt inhibited TNBC cell proliferation more effectively than non-TNBC cells
It induces non-apoptotic, lipid peroxidation-dependent cell death (ferroptosis)
In vivo experiments with nude mice showed that SGNI significantly inhibited tumor growth without adverse effects
[99]
MCF-7, Huh7, T47D, HCC 1954, AGS, LoVo, Mia-Paca2, U2OS, and MDA-MB-231Female C57BL/6 mice (6 weeks of age)“Heirloom recipe” (Formula X)not present5.07 mg/gFormula X suppressed the growth of four breast cancer cell lines (T47D, MCF7, MDA-MB231, HCC1954) in a dose-dependent manner
Reduction of the expression of the oncogenic proteins
Berberine, baicalin, and saponin mixture reduced tumor growth in mouse models
[100]
MCF-7, T47D, Vero, and EMT6/PBALB/c female mice (4–6 weeks old)Xanthium spinosumpresent in methanol fractionpresent in hexane fraction In vivo studies showed significant tumor size reduction in mice treated with X. spinosum extracts[23]
4T1Female BALB/c mice (4–6 weeks old)Sanhuang Xiexin Decoction (SXD)not present46.04 mg/gSXD demonstrated significant antitumor effects against 4T1 breast cancer in mice through modulation of lipid profiles, cytokine levels, angiogenesis inhibition, and low toxicity[101]
MCF-7 and MDA-MB-231-Huangqin Tang (HQT)presentnot specifiedHQT inhibited proliferation, induced apoptosis, induced G2/M phase arrest, and reduced HIF-1α protein levels in breast cancer cells[102]
MCF-7, MDA-MB-231, SKBR-3, and L929-aerial parts of Centaurea mersinensis11.12 µg/g648.28 µg/gThe methanol extract showed moderate cytotoxicity against breast cancer cell lines (IC50 = 22.17–46.20 µg/mL)[24]
Table 5. Derivatives of baicalein and their anticancer effects in breast cancer research.
Table 5. Derivatives of baicalein and their anticancer effects in breast cancer research.
Derivative(s)Cell LinesAnimal ModelsThe Outcome of the StudyReference
Prenylated baicaleinMCF-7 and MDA-MB-231-The prenylated baicalein derivative demonstrated antiproliferative activity in MCF-7 cells, likely mediated through estrogen receptor interactions, and showed potential for synergistic effects with 4-hydroxytamoxifen and fulvestrant[104]
Baicalein-8-sodium sulfonate and Baicalein-7-O-sulfateMCF-7 and H184B5F5/M10-Baicalein-8-sodium sulfonate exhibited a higher anticancer effect than its precursor, induced apoptosis, cell cycle arrest, and ROS-mediated oxidative stress in MCF-7 cells[105]
Oroxylin AMCF-7-Oroxylin A inhibited CYP1B1-mediated 4-hydroxylation of 17β-estradiol
Oroxylin A was more potent than baicalein
[106]
8-Chlorobaicalein, 8-bromobaicalein, chrysin, 8-chlorochrysin, 8-bromochrysin, 6,8-dichlorochrysin, 6-bromo-8-chlorochrysin, and 6,8-dibromochrysinA549, LoVo, MV4-11, MCF-7, and MCF-10A-Baicalein and its halogenated derivatives showed increased cytotoxicity towards breast cancer cells (MCF-7) but were also toxic to non-tumorigenic breast cells (MCF-10A)[107]
8-BromobaicaleinMCF-7-8-bromobaicalein and 2,6-di-O-methyl-β-cyclodextrin complexes demonstrated enhanced cytotoxic activity against MCF-7 breast cancer cells, with IC50 values significantly lower than free 8-bromobaicalein[108]
Table 6. Comparative analysis of baicalin and baicalein in breast cancer models.
Table 6. Comparative analysis of baicalin and baicalein in breast cancer models.
Property/ActivityBaicalinBaicalein
Molecular FormulaC21H18O11C15H10O5
Molecular Weight (g/mol)446.36270.24
H-Bond Acceptors115
H-Bond Donors63
Molar Refractivity106.7273.99
TPSA (Å2)187.1290.90
Consensus LogPo/w0.252.24
Water Solubility LogS−3.41−4.03
GI AbsorptionLowHigh
BBB PermeantNoNo
P-gp SubstrateYesNo
CYP InhibitionNoneCYP1A2, CYP2D6, CYP3A4
Skin Permeation (Log Kp) (cm/s)−8.23−5.70
Breast Cancer Cell Lines Tested4T1, MADB-106, MCF-7, MCF-7 ADRB2^OE, MDA-MB-231, SK-BR-2, ZR-75-14T1, BT549, Hs578T, MCF-7, MDA-MB-231, MDA-MB-231/IR, MDA-MB-435, MDA-MB-468, MDA-MB-468-C23, SK-BR-3, T47D, ZR-75-1
Bioavailability IssuesPoor water solubility
Poor intestinal absorption
Better absorption than baicalin
Rapid glucuronidation in liver/enterocytes
DerivativesNo derivatives were examinedPrenylated, sulfonated, and halogenated derivatives improved efficacy
Anti-ProliferativeSuppressed cell growth via ESR1 downregulationInhibition of cells via mitochondrial pathways and DDIT4 induction
Apoptoticp53/Bax upregulation, caspase-3/-8/-9 activation, and ROS productionActivated mitochondrial apoptosis, caspase cascades, and Bcl-2 modulation, and increased Ca2+, ROS, and DNA damage
Anti-MetastaticInhibited EMT via β-catenin/TGF-βBlocked MMP-2/9, SATB1/Wnt pathways
Pathway TargetsNF-κB, mTOR, miRNAsSTAT3, PI3K/AKT/mTOR, Wnt/β-catenin
Table 7. Combination therapies of baicalin/baicalein with other drugs in anticancer research against breast cancer.
Table 7. Combination therapies of baicalin/baicalein with other drugs in anticancer research against breast cancer.
Combination of:Cell LinesAnimal ModelsThe Outcome of the StudyReference
Baicalin and Salvia miltiorrhiza extractMCF-7, T-47D, FaDu, and CAL-27-Combining baicalin with S. miltiorrhiza extract resulted in synergistic effects, significantly enhancing the inhibition of MCF-7, T-47D, and CAL-27 cells[110]
Baicalin and DoxorubicinMCF-7 and chick cardiomyocytes-Baicalein protected against doxorubicin-induced cardiotoxicity by reducing ROS generation, preserving mitochondrial function, and inhibiting JNK-mediated apoptosis
Baicalein did not interfere with doxorubicin anticancer activity in MCF-7 cells
[111]
Baicalein and PaclitaxelMCF-7Female Kunming mice (4–6 weeks old)When combined with paclitaxel, baicalein showed antagonistic effects, i.e., reduced the growth-inhibitory effect of paclitaxel[112]
Baicalein and U0126MCF-7-The combination of baicalein with U0126 enhanced inhibition of cell proliferation, induction of apoptosis and cell cycle arrest, and suppression of migration, likely through modulation of the MAPK/ERK signaling pathway[113]
Baicalein and ultrasoundsMDA-MB-231-Synergistic effect of the combination of baicalein and ultrasound
Both baicalein and low-frequency ultrasound effectively reduce the invasive activity of MDA-MB-231 cells by downregulating the expression of MMP-2, MMP-9, and u-PA at both the mRNA and protein levels
[114]
Bismuth Oxide Nanoparticles, Cisplatin, and Baicalein-Rich Fraction from O. indicumMCF-7, MDA-MB-231, and NIH/3T3-The combination of bismuth oxide nanoparticles and cisplatin was the most effective in enhancing ROS generation and radiosensitization in breast cancer cells
The addition of a baicalein-rich fraction had significant radiosensitization effects in normal cells
[115]
Baicalin and DocetaxelMDA-MB-231, MDA-MB-453, and 4T1Female BALB/c mice (6–8 weeks old)Baicalin inhibited cell proliferation, induced mitochondria-mediated apoptosis, suppressed migration and invasion via the NF-κB pathway, and enhanced chemosensitivity to docetaxel
Baicalin inhibited tumor growth and pulmonary metastasis
[116]
Baicalin and DoxorubicinMDA-MB-231 and MCF-7-Baicalin enhanced doxorubicin anticancer effects by increasing oxidative stress, calcium levels, and mitochondrial dysfunction in breast cancer cells[117]
Baicalin and 5-fluorouracil-Swiss albino miceThe combination of baicalin and 5-fluorouracil demonstrated the most effective antitumor activity, significantly inhibiting tumor growth, promoting apoptosis, and reducing tumor-related biomarkers[118]
Bismuth Oxide Nanoparticles, Cisplatin, and Baicalein-Rich Fraction from O. indicumMCF-7, MDA-MB-231, and NIH/3T3-The combination demonstrated the most potent radiosensitization effects across all radiation types in cancer cells[119]
Baicalein and CapecitabineMCF-7Thirty-two adult female albino ratsIn MCF-7 cells, the combination of compounds enhanced cytotoxicity, cell cycle arrest, and apoptosis compared to individual treatments
In rats, baicalein partially mitigated capecitabine-induced cardiotoxicity, oxidative stress, and inflammation
[120]
Baicalin and DoxorubicinMDA-MB-231-Baicalein enhanced the sensitivity of MDA-MB-231 cells to doxorubicin by activating autophagy and mitophagy, down-regulating CDK1, and inhibiting Drp1-mediated mitochondrial fission[121]
Baicalin/baicalein and Doxorubicin or DocetaxelMCF-7 and HUVEC-ST-Baicalin or baicalein enhanced the cytotoxicity of doxorubicin and docetaxel in MCF-7 cells
Both flavonoids increased doxorubicin uptake in MCF-7 cells, induced apoptosis, and caused DNA damage
[122]
Baicalin, Epigallocatechin Gallate (EGCG), and VincristineEMT-6/P and EMT-6/VFemale BALB/c mice (4–6 weeks old)Baicalin and EGCG demonstrate strong anti-proliferative effects, especially in vincristine-resistant cancer cells
The combination with vincristine had a synergistic effect in resistant cells
Combined therapy significantly reduced tumor size and achieved high cure rates in both models with no liver or kidney toxicity
[123]
Table 8. Overview of a nanotechnology-based delivery system for baicalin/baicalein in breast cancer research.
Table 8. Overview of a nanotechnology-based delivery system for baicalin/baicalein in breast cancer research.
FormulaCell LinesAnimal ModelsThe Outcome of the StudyReference
Baicalein-loaded iron oxide nanoparticlesMDA-MB-231 and HBL-100-Baicalein-loaded nanoparticles showed significant cytotoxicity against MDA-MB-231 cells (IC50 = 22 µg/mL)
Baicalein-loaded nanoparticles disrupted the cell cycle and induced apoptosis by downregulation of the anti-apoptotic protein Bcl-2 and upregulation of pro-apoptotic proteins (Bax, cytochrome-c, caspase-3, PARP, and p53)
[125]
Baicalin-loaded gold nanoparticles (AuNPs) conjugated with thiolated beta-cyclodextrinMCF-7-The obtained delivery system enhanced the apoptotic effects of baicalin [126]
Baicalein and paclitaxel nanoemulsionMCF-7 and MCF-7/TaxFemale BALB/c nude mice (6–8 weeks old)The obtained nanoemulsions enhanced anticancer efficacy through synergistic effects, increased cellular uptake, oxidative stress induction, and apoptosis activation
In an MCF-7/Tax xenograft model, nanoemulsion showed the highest tumor inhibition rate (77.0%) and minimal weight loss
[127]
Hyaluronic acid-decorated nanostructured lipid carriers (NLCs) as nanocarriers for co-delivery of baicalein and doxorubicinMCF-7/ADRKunming mice (4–6 weeks old)The co-delivery system exhibited enhanced targeting, sustained drug release, and synergistic anticancer effects (IC50 = 0.056 mg/mL)
The obtained formula demonstrated the highest tumor inhibition rate (88%) in mice bearing human breast cancer
[128]
Baicalein-loaded iron oxide nanoparticlesMDA-MB-231-Baicalein-loaded iron oxide nanoparticles effectively internalized into MDA-MB-231 cells, localized in critical cellular regions, and induced apoptosis through mitochondrial dysfunction, DNA damage, and cell cycle arrest.[129]
Baicalin-loaded polylactide-glycolide (PLGA) nanocapsulesMCF-7 and MDA-MB-231-Nanocapsules demonstrated superior anticancer activity and induced apoptosis, sustained drug release, and enhanced cellular uptake[130]
Methoxy poly(ethylene glycol)-folic acid-decorated zeolitic imidazolate framework (ZIF-8) loaded with baicalinMCF-7 and L929Female BALB/c miceThe nano-delivery system demonstrated excellent tumor-targeting capability, pH-responsive drug release, and potent anticancer activity both in vitro and in vivo.[131]
Folate-modified albumin baicalin-loaded nanoparticlesMCF-7Female BALB/c nude mice (6–8 weeks old)Nanoparticles demonstrated excellent tumor-targeting capability, sustained drug release, and potent anticancer activity both in vitro and in vivo
The obtained system enhanced cellular uptake, induced apoptosis, and significantly inhibited tumor growth
[132]
Baicalein and cinnamon essential oil nanoemulsionMDA-MB-231-Nanoemulsion demonstrated enhanced anticancer activity 19-fold and 23-fold higher cytotoxicity compared to free baicalein after 12 h and 24 h, respectively, against breast cancer cells
The anticancer activity is attributed to the combined effect of baicalein and cinnamon oil, along with improved drug permeability and cellular uptake via endocytosis
[133]
Hyaluronic acid-modified ferrous baicalein nanoparticleHUVECs and 4T1Female BALB/c mice (8 weeks old)In 4T1 tumor-bearing mice, nanoparticles and near-infrared significantly inhibited tumor growth (a combination of photothermal therapy and chemotherapy), showed tumor cell necrosis/apoptosis with no damage to major organs [134]
Folate-modified albumin baicalin-loaded nanoparticlesMCF-7Female BALB/c nude miceNanoparticles demonstrated sustained drug release, enhanced cellular uptake, and potent anti-cancer effects through induction of cell cycle arrest, apoptosis, and autophagy[135]
Silver nanoparticles obtained by S. multicaulis leaf extractMDA-MB-231 and HFF2-The green-synthesized silver nanoparticles demonstrated significant antioxidant, anticancer, and apoptotic activities[136]
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

Zieniuk, B.; Uğur, Ş. The Therapeutic Potential of Baicalin and Baicalein in Breast Cancer: A Systematic Review of Mechanisms and Efficacy. Curr. Issues Mol. Biol. 2025, 47, 181. https://doi.org/10.3390/cimb47030181

AMA Style

Zieniuk B, Uğur Ş. The Therapeutic Potential of Baicalin and Baicalein in Breast Cancer: A Systematic Review of Mechanisms and Efficacy. Current Issues in Molecular Biology. 2025; 47(3):181. https://doi.org/10.3390/cimb47030181

Chicago/Turabian Style

Zieniuk, Bartłomiej, and Şuheda Uğur. 2025. "The Therapeutic Potential of Baicalin and Baicalein in Breast Cancer: A Systematic Review of Mechanisms and Efficacy" Current Issues in Molecular Biology 47, no. 3: 181. https://doi.org/10.3390/cimb47030181

APA Style

Zieniuk, B., & Uğur, Ş. (2025). The Therapeutic Potential of Baicalin and Baicalein in Breast Cancer: A Systematic Review of Mechanisms and Efficacy. Current Issues in Molecular Biology, 47(3), 181. https://doi.org/10.3390/cimb47030181

Article Metrics

Back to TopTop