Abstract Breast cancer is the second leading cancer type in women, accounting for 11.6% of all cancers. Recently, its incidence has increased in younger individuals. Estrogen receptors (ERs) play important roles in the development and progression of breast cancer by controlling hormone signaling. Therefore, targeting ERs is one of the most promising therapeutic strategies for treating ER-positive breast cancer. High heterogeneity contributes to the tumorigenicity, metastatic ability, and therapeutic resistance of breast cancer. However, drug discovery studies at the single-cell level are lacking. In the present study, we used single-cell sequencing data analysis, together with a network pharmacology approach, to determine the targets and molecular mechanisms of formononetin in ER-positive breast cancer. Comparative single-cell sequencing analysis identified 3899, 3395, 333, 398, 398, and 17 differentially expressed genes in stromal cells, epithelial cells, fibroblasts, neutrophils, eosinophils, and macrophages, respectively, of ER-positive breast cancer compared with normal breast tissues. Further network pharmacology analysis highlighted the importance of formononetin targets in biological functions and signaling pathways related to immune and inflammatory responses, metastatic ability, metabolism, cell proliferation, and gland development in different ER-positive breast cancer cell types. For the first time, we used a systems biology approach to investigate the targets of formononetin and its anti-ER-positive breast cancer mechanisms at the single-cell level. Keywords: Network pharmacology, Formononetin, Estrogen receptor, ER-positive breast cancer, Single cell Subject terms: Medical research, Molecular medicine, Cancer Introduction According to the 2022 Global Cancer Statistics, female breast cancer is the second leading disease in terms of incidence rate, accounting for 11.6% of all cancers^[32]1. Breast cancer has become a major global public health challenge and is gradually affecting younger individuals. Its incidence among young people in Japan has increased annually^[33]2. Breast cancer can be pathologically classified into ductal and lobular carcinomas with cancerous lesions occurring in the ducts and lobules, respectively^[34]3. Breast cancer can be caused by multiple factors, such as heredity, environmental exposure, individual differences, and lifestyle^[35]4. Among these, hormone receptors, including estrogen receptor (ER), progesterone receptor (PR), and human epidermal growth factor receptor 2 (HER2), are routine indicators of breast cancer treatment and prognostic markers because they are expressed in the vast majority of breast cancers^[36]5. ER and PR positivity (ER +/PR +) accounts for two-thirds of all breast cancers^[37]6, and these tumors express either ERs or PRs^[38]7. Because ER expression is closely related to the proliferation of breast cancer cells, the use of hormone therapy to target ERs has become one of the major treatments for ER-positive breast cancers^[39]6. Clinically, breast cancer heterogeneity is associated with the expression of different hormone receptors, which provides different options for breast cancer treatment^[40]8. Conventional treatments for breast cancer are not limited to surgical excision, radiotherapy, chemotherapy, and endocrine therapy^[41]9. Although these methods have improved the effectiveness of treatment to a certain extent, they have several limitations. For example, endocrine therapy for breast cancer is associated with a risk of drug resistance and recurrence^[42]10. Radiotherapy may cause adverse effects in normal tissues and organs. Furthermore, radiotherapy for breast cancer increases the risk of heart disease and myocardial infarction^[43]11. Therefore, an in-depth understanding of the role of ER in breast cancer tumorigenicity is crucial for the development of effective treatment strategies. With the development of biomedical technology, numerous studies have revealed the complex mechanisms underlying breast cancer development, including gene mutations, epigenetic regulation, and signaling pathway abnormalities. Therefore, breast cancer treatment needs to be constantly innovated and explored to develop new therapeutic approaches that improve treatment efficacy and reduce side effects. Network pharmacology is a network-based research approach used to analyze systems biology that can reveal complex network relationships between drugs and organisms. Traditional single-target approaches tend to focus only on the effects of drugs on a single target and ignore the complex effects of drugs on an organism. Network pharmacology can be used to understand multi-pathway and multi-target mechanisms of action in organisms by constructing drug–target–disease interaction networks for studying many complex diseases^[44]12–[45]15. In addition, tumor development is no longer considered a single-factor-induced disease^[46]16. Drug resistance, which is a major challenge in breast cancer treatment, is associated with tumor heterogeneity. The development of single-cell RNA sequencing has recently played a great advantage in the analysis of tumor heterogeneity and has allowed scientists to analyze the genetic profiles of drug-resistant cells to determine the mechanism of cellular resistance acquisition^[47]17. Various studies have indicated that breast cancer is highly heterogeneous and has different molecular subtypes^[48]18,[49]19. The molecular drivers and heterogeneity of breast cancer can be precisely studied using single-cell sequencing technology. Formononetin, a natural phytoestrogen, can be extracted from many plants, such as soybeans and red clover^[50]20. It has a promising potential for clinical applications, especially in cancer treatment^[51]21. Its chemical structure is similar to that of estrogen, making it a potential drug for treating ER-positive breast cancer. Moreover, formononetin has anti-inflammatory functions, such as the modulation of mast cells and basophils through the activation of the JAK/STAT/PI3-Akt pathways^[52]22, suggesting a possible immune activation response. In the present study, we applied network pharmacology together with single-cell sequencing data of ER-positive breast cancer obtained from a previous report to determine the targets of formononetin for treating breast cancer at the single-cell level^[53]23. Following bioinformatics analysis, we further highlighted the mechanisms underlying the anti-breast cancer effects of formononetin. Materials and methods Source and analysis of single cell sequencing data of ER-positive breast cancer To determine the differential gene expression in ER-positive breast cancer at the single-cell level, the keywords “ER-positive breast cancer” and “single cell” were used to search in the Gene Expression Omnibus (GEO) database ([54]https://www.ncbi.nlm.nih.gov/geo/). The single-cell sequencing dataset [55]GSE245601 was obtained^[56]23. The sequencing data were analyzed using the tools “Seurat,” “cowplot,” “BiocManager,” “SingleR,” “dplyr,” “tidyverse,” and “patchwork” of R packages. The “Seurat” tool was used for QC, the included data criteria including the number of genes in a cell should be > 200, and the genes should be expressed in at least 10 cells. The PercentageFeatureSet tool was used to calculate the percentage of mitochondria-related genes in each sample. The cell with 500 < gene < 3000 and with a percentage of mitochondria-related genes < 15% were used for further analysis. A total of 2000 genes with highly variable coefficients between cells were extracted, and the data were analyzed using canonical correlation analysis to eliminate batch effects between different groups of data. After principal component analysis and uniform manifold approximation and projection (Umap) cluster analysis, the cells were divided into different sub-clusters. Cell types for each cluster were annotated using the SingleR automated annotation package. After grouping the data, the data from two normal breast tissues (named “N”) and the data from 10 ER-positive breast cancerous tissues (named “T”) was subjected to the FindMarkers function to determine differentially expressed genes (DEGs) between cancerous tissues and normal tissues of the same cell type, and genes with p < 0.05 and |log2 FoldChange|> 1 were defined as DEGs. Harvesting formononetin’s targets against ER-positive breast cancer at the single-cell level The targeted genes of formononetin were identified by scanning online databases, including the Swiss Target Prediction^[57]24, PharmMapper, and SuperPred. The targets obtained were corrected using the UniProtKB database. The harvested genes were compared with DEGs obtained from single-cell sequencing data of ER-positive breast cancer cells to determine the targets of formononetin in ER-positive cancer. Gene ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway enrichment analysis The “ClusterProfiler” package of the R language was used for biological-term classification and the enrichment analysis of gene clusters, and the R package “GOplot” was used to visualize the expression data with functional analysis. GO terms and pathways were considered statistically significant at p < 0.05. Results DEGs in ER-positive breast cancer at the single-cell level To determine differential gene expression in ER-positive breast cancer at the single-cell level, we downloaded single-cell sequencing data containing two normal breast tissues and 10 ER-positive breast cancerous tissues from the GEO database^[58]23. We then determined the number of expressed genes, sum of gene expression, and percentage of mitochondria-related genes per cell (Supplementary Fig. [59]1). Using Umap cluster analysis and SingleR automatic annotation package, we identified 24 cell clusters in normal and ER-positive breast cancer tissues (Fig. [60]1A) belonging to six distinct cell types, including stromal cells, epithelial cells, fibroblasts, neutrophils, eosinophils, and macrophages (Fig. [61]1B). When we compared the gene expression profiles of normal breast tissues and ER-positive breast cancer tissues, we identified 3899 DEGs in stromal cells, 3395 DEGs in epithelial cells, 333 DEGs in fibroblasts, 398 DEGs in neutrophils, 398 DEGs in eosinophils, and 17 DEGs in macrophages. Fig. 1. [62]Fig. 1 [63]Open in a new tab Comparative single cell sequencing analysis of estrogen receptor (ER)-positive breast cancer. (A) Uniform manifold approximation and projection (Umap) classified the single cell of ER-positive breast cancer tissues and normal breast tissues into 24 cell clusters. (B) The clusters were classified into stromal cells, epithelial cells, neutrophils, fibroblasts, eosinophils, and macrophages using the SingleR automatic annotation package. Formononetin’s targets against ER-positive breast cancer tissues at the single-cell level By searching online databases, followed by correcting using the UniProtKB database, 402 formononetin-related genes were identified. When we overlaid the formononetin-associated genes and DEGs in each ER-positive breast cancer cell type, we identified the target genes of formononetin in ER-positive breast cancer at the single-cell level (Fig. [64]2). Fig. 2. [65]Fig. 2 [66]Open in a new tab Network pharmacology analysis identified formononetin’s targets against estrogen receptor-positive breast cancer in single-cell level. Venn diagram showed the number of overlapped genes in stromal cells, epithelial cells, neutrophils, fibroblasts, eosinophils, and macrophages. Formononetin targeted the genes related to immune and inflammatory responses in immune cells of ER-positive breast cancer GO enrichment analysis of the biological processes of immune cells revealed that formononetin targeted genes in response to external stimuli and defense responses, including the response to wounds and blood coagulation (Fig. [67]3A). In addition, formononetin targeted genes that contribute to different stresses, including oxidative and chemical stresses (Fig. [68]3A). This triggered immune and inflammatory responses, such as endocytosis and phagocytosis (Fig. [69]3B). Additionally, the targets of formononetin were involved in the regulation of cell signal transduction (Fig. [70]3B). In the GO enrichment analysis of molecular functions, formononetin regulated many enzyme activities, such as protein kinase and phosphatase activities (Fig. [71]3C). Moreover, the KEGG pathway analysis suggested the involvement of formononetin in mitogen activated protein kinase (MAPK), 5′ adenosine monophosphate-activated protein kinase (AMPK), and Rap1 signaling (Fig. [72]3D). More importantly, formononetin targets contributed to transcriptional dysregulation in cancer cells (Fig. [73]3D). Fig. 3. [74]Fig. 3 [75]Fig. 3 [76]Open in a new tab Functions of formononetin’s targets in immune cells of estrogen receptor-positive breast cancer. Gene ontology (GO) enrichment analysis highlighted the biological roles and molecular functions of formononetin’s targets in (A) in response to wound healing and blood coagulation, (B) immune and inflammatory responses and cell signaling, (C) protein kinase and phosphatase activities. (D) Kyoto Encyclopedia of Genes and Genomes pathway enrichment analysis further highlighted the roles of formononetin’s targets in MAPK signaling, AMPK signaling, and Rap1 signaling; and transcriptional dysregulation in cancer. The bubble’s size represents the number of involved gene. The bubble’s color represents the significance of the processes and pathways. Formononetin mediated stress response, metastasis ability, and proliferation of the epithelial cells in ER-positive breast cancer The GO analysis of the epithelial cells of ER-positive breast cancer revealed that formononetin targeted genes involved in stress responses, such as cell death in response to oxidative stress and cellular response to chemical stress, through activated MAPK cascade and protein kinase signaling cascade (Fig. [77]4A). In addition, formononetin targets contributed to apoptosis by regulating the intrinsic apoptotic signaling pathway (Fig. [78]4A). More importantly, formononetin regulated the expression of genes involved in cancer metastasis, including those involved in wound healing and cell migration (Fig. [79]4B). Formononetin targets also played an important role in cell proliferation and regulation of tumor necrosis factor production (Fig. [80]4C). In the KEGG pathway analysis, formononetin could target the genes involved in breast cancer development including epidermal growth factor receptor (EGFR), ESR1, KIT, FGFR1, CDK6, insulin-like growth factor 1 (IGF1), and HRas proto-oncogene (HRAS). In addition, formononetin’s targets controlled numerous cell signaling pathways contributed to the breast carcinogenesis, such as Ras, MAPK, Relaxin, IL-17, TNF, Rap1, estrogen, phospholipase D, PI3K–Akt, HIF-1, FoxO, GnRH, NF-kappa B, and JAK–STAT signaling pathways (Fig. [81]4D). Fig. 4. [82]Fig. 4 [83]Fig. 4 [84]Fig. 4 [85]Fig. 4 [86]Open in a new tab Functions of formononetin’s targets in epithelial cells of estrogen receptor-positive breast cancer. Gene ontology enrichment analysis showed the biological roles of formononetin’s targets in (A) stress responses and cell apoptosis, (B) cancer metastasis, such as wound healing and cell migration, and (C) cell proliferation and regulation of tumor necrosis factor production. (D) Kyoto Encyclopedia of Genes and Genomes pathway enrichment analysis further highlighted the roles of formononetin’s targets in cell signaling of breast cancer development. The bubble’s size represents the number of involved genes. The bubble’s color represents the significance of the processes and pathways. Formononetin mediated cellular response to estrogen stimulus and gland development through targeting the stromal cells of ER-positive breast cancer We focused on the effects of formononetin on stromal cells in ER-positive breast cancer. The results of the GO enrichment analysis highlighted the importance of formononetin targets in biological processes related to stress-associated cell death, especially apoptosis (Fig. [87]5A). In addition, formononetin targeted the stromal cell genes involved in cell proliferation by regulating the cell cycle (Fig. [88]5B). More importantly, formononetin targets played important roles in response to estrogen, mammary gland development, and morphogenesis (Fig. [89]5C). In addition, the targets of formononetin contributed to the regulation of angiogenesis, cell migration, and wound healing, which are closely associated with cancer metastasis (Fig. [90]5D). In KEGG pathway enrichment analysis, our results further highlighted the importance of formononetin in breast carcinogenesis by targeting EGFR, ESR1, phosphoinositide-3-kinase regulatory subunit 1 (PIK3R1), PGR, CDK6, and IGF1. In addition, formononetin targets were involved in many important signaling pathways closely associated with breast cancer tumorigenicity, especially estrogen, PI3K–Akt, insulin, MAPK, JAK–STAT, FoxO, VEGF, TNF, and p53 signaling (Fig. [91]5E). Fig. 5. [92]Fig. 5 [93]Fig. 5 [94]Fig. 5 [95]Fig. 5 [96]Open in a new tab Functions of formononetin’s targets in stromal cells of estrogen receptor-positive breast cancer. Gene ontology enrichment analysis showed the biological roles of formononetin’s targets in (A) stress-associated cell death, (B) cell proliferation, (C) in response to estrogen and mammary gland development and morphogenesis, and (D) regulation of angiogenesis, cell migration ability, and wound healing. (E) Kyoto Encyclopedia of Genes and Genomes pathway enrichment analysis further highlighted the roles of formononetin’s targets in breast carcinogenicity and cell signaling. The bubble’s size represents the number of involved gene. The bubble’s color represents the significance of the processes and pathways. Formononetin targeted NFKB1 and monoamine oxidase B (MAOB) to mediate metabolisms in fibroblasts of ER-positive breast cancer Formononetin only targeted limited gene candidates, including NFKB1 and MAOB, to regulate metabolic processes, such as polysaccharide, carbohydrate, steroid, and hydrogen peroxide biosynthesis (Fig. [97]6). Furthermore, formononetin targeted genes involved in mammary gland morphogenesis (Fig. [98]6). Fig. 6. [99]Fig. 6 [100]Open in a new tab Functions of formononetin’s targets in fibroblasts of estrogen receptor-positive breast cancer. Gene ontology enrichment analysis revealed the biological roles of formononetin targets in metabolism and mammary gland morphogenesis. The bubble’s size represents the number of genes involved. The bubble’s color represents the significance of the processes. Discussion Breast cancer is one of the most common malignant tumors in women worldwide and poses a serious threat to their physical and mental health^[101]25. ER-positive breast cancer, a major subtype of breast cancer, is closely related to ER expression^[102]26. In the present study, we used single-cell sequencing analysis and network pharmacology to identify the targets of formononetin in ER-positive breast cancer at the single-cell level, including immune, epithelial, and stromal cells. Role of immune cells in ER-positive breast cancer With the increase in immunotherapy in recent years, the role of immune cells in ER-positive breast cancer has gradually received attention. Immune cells recognize and kill ER-positive breast cancer cells. For example, T-cells specifically recognize antigens on the surface of breast cancer cells and directly kill them by releasing cytotoxins. A significant increase in CD(4)(+) and CD(25)(+) regulatory T cells has been reported in the peripheral blood of patients with breast cancer^[103]27. Natural killer (NK) cells can kill tumor cells by exerting a cytotoxic effect^[104]28. Moreover, immune cells play an important regulatory role in the breast cancer tumor microenvironment (TME), and can be classified into lymphocytes, tumor-associated macrophages, and myeloid-derived suppressor cells^[105]29. Tumor antagonist cells, such as effector T cells and NK cells, inhibit tumor growth and metastasis^[106]30. Meanwhile, tumor-promoting cells, such as M2-like macrophages, stimulated by cytokines secreted by type 2 T-helper cells, such as IL-4, IL-10 and IL-13 as well as transforming growth factor-beta, exhibit strong tumor-promoting characteristics^[107]29,[108]31 Therefore, formononetin targeting the immune cells in ER-positive breast cancer cell could be a promising approach for treating breast cancer. Role of epithelial cells in ER-positive breast cancer In addition, formononetin could target epithelial cells in ER-positive breast cancer, which not only participates in the proliferation and invasion of tumor cells but also influences the prognosis and treatment response of patients with breast cancer. In breast tissue, epithelial cells are the main cell type that make up the ducts and alveoli of the breast, and mammary epithelial cells are mainly classified as either basal or luminal cells, which are essential for maintaining the normal physiological function of the breast^[109]32. Recent studies have shown that changes in the polarity of breast epithelial cells play an important role in breast cancer development. During epithelial–mesenchymal transition (EMT), the loss of cell apex–base polarity, accompanied by the loss of epithelial phenotype and the formation of a mesenchymal phenotype, enhances epithelial cell motility and acquires the ability to migrate and infiltrate^[110]33. In addition, epithelial cells in ER-positive breast cancer provide a favorable environment for tumor cell growth through the secretion of growth factors and cytokines, thereby promoting the proliferation of epithelial and tumor cells. For example, sustained expression of CCL2 in mammary epithelial cells induces low levels of chronic inflammation, thereby increasing the risk of cancer^[111]34. Moreover, the abnormal proliferation and differentiation of epithelial cells in breast cancer can lead to increased invasiveness of tumor cells. In addition, the layer of myoepithelial cells surrounding the ducts of the breast can capture cancer cells that intend to escape, preventing them from spreading throughout the body^[112]35. However, when the myoepithelial cell layer is damaged, cancer cells may break through the defects and form invasive ductal carcinoma^[113]36. Moreover, the state of epithelial cells may also affect the patient’s sensitivity to systemic therapies, such as chemotherapy and targeted therapies, thereby affecting the patient’s prognosis. Role of stromal cells in ER-positive breast cancer Stromal cells, an important part of the TME, play a key role in the carcinogenesis, progression, and metastasis of ER-positive breast cancer^[114]37. In addition to providing support during tumorigenesis, stromal cells and their products can alter neighboring tissues by inducing phenotypic and genomic changes in epithelial cells^[115]38. Moreover, stromal cells may provide supportive functions during breast cancer development and promote angiogenesis to facilitate tumor metastasis^[116]37. Fibroblasts, as a type of stromal cell, break down to mimic traumatic damage to the tissue in invasive breast cancer. These fibroblasts become tumor-associated fibroblasts, which provide essential nutritional support and growth signals to ER-positive breast cancer cells^[117]37. Stromal cells can promote angiogenesis by releasing high levels of stromal-derived factor-1, which attracts endothelial progenitor cells in the tumor mass and directly promotes cancer cell growth by interacting with their receptor CXCR4^[118]39. In addition, stromal cells release growth factors, such as epidermal growth factor (EGF) and transforming growth factor β (TGF), to create an environment conducive to tumor growth^[119]40. Role of formononetin’s targets EGFR, ESR1, KIT, FGFR1, CDK6, IGF1, HRAS, PIK3R1, PGR, NFKB1, and MAOB in ER-positive breast cancer Network pharmacology analysis identified several formononetin targets that have been reported to play important roles in the tumorigenesis of ER-positive breast cancer. EGFR, also known as HER1 or ErbB-1, is a member of the epidermal growth factor receptor (HER) family. EGFR, with its tyrosine kinase activity, is a transmembrane glycoprotein that is widely distributed across the cell membranes of various human tissues and is particularly highly expressed in various tumor cells^[120]41. Moreover, the overexpression of EGFR in primary tumors indicates poor prognosis and is inversely correlated with ER in breast cancer^[121]42. In addition, the expression level of transforming growth factor alpha, a ligand of EGFR, is significantly lower in ER-positive than in ER-negative breast cancer^[122]43. ESR1 is a hallmark of ER-positive breast cancer. This gene encodes the ER, and its corresponding protein product is ERα, whose positive expression implies that the tumor cells are sensitive to estrogen, one of the main targets for endocrine therapy^[123]44. Increased activity of ERα is positively correlated with breast cancer progression, and overactivation of ERα makes ER-positive breast cancers more aggressive and refractory^[124]45. The proto-oncogene tyrosine–protein kinase kit (KIT), also known as proto-oncogene c-Kit, encodes a receptor tyrosine kinase that can activate various signaling pathways. KIT phosphorylates PIK3R1, PLCG1, SH2B2/APS, and CBL; stimulates major cellular functions, such as cell survival, proliferation, differentiation, adhesion, and chemotaxis; and induces apoptosis and cancer cell invasiveness^[125]46, which is used to differentiate between malignant and benign breast lesions^[126]47. Fibroblast growth factor receptor 1 (FGFR1) is a growth factor receptor tyrosine kinase that contains extracellular, transmembrane, and cytoplasmic structural domains^[127]48. Amplification of the chromosomal region of FGFR1 has been detected in approximately 10% of human breast cancers and is predominantly present in ER-positive breast cancers, where it negatively influences the overall survival of the patients with breast cancer^[128]49. In addition, FGFR1 amplification contributes to breast carcinogenesis and promotes resistance to endocrine therapy^[129]50. Cyclin-dependent kinase 6 (CDK6), also known as cell cycle protein-dependent kinase 6, plays an important role in cell-cycle progression and regulation. CDK6 binds to D-type cell cycle proteins to form an active kinase complex that regulates the progression of G1 phase of the cell cycle^[130]51. Early preclinical data suggest that breast cancer, particularly ER-positive breast cancer, is very sensitive to CDK6 inhibition, and the median progression-free survival of patients with breast cancer can be significantly prolonged by the administration of CDK4/6 inhibitors such as Palbociclib^[131]52. IGF1 promotes cell growth and differentiation. The binding of IGF-1 to its cognate receptor IGF-1R triggers a signaling cascade, leading to cell proliferation and anti-apoptotic effects. Although IGF-1 receptor signaling is critical for cancer cell growth, IRS-1 can be overexpressed in ER-positive breast cancer^[132]53, and IGF-1 levels positively correlate with poor cancer prognosis^[133]54. Moreover, IGF-1 receptor inhibitors may accelerate tumor growth through aberrant feedback loops in resistant tumors^[134]54. Abnormal feedback loops accelerate tumor growth^[135]55. HRAS is an important signal transduction protein involved in intracellular signal transduction. HRAS mutation results in tumor development^[136]56. Moreover, the RAS oncogene family can regulate p63 to induce EMT, leading to an increased invasive capacity of breast cancer epithelial cells^[137]57. PIK3R1 is a tumor suppressor that is downregulated in many tumor types^[138]58. Upregulation of PIK3R1, particularly its p50α isoform, greatly activates the PI3K/Akt pathway, which may attenuate resistance to chemotherapy in breast cancer^[139]59. PRs are important regulators of breast cancer cell growth. PR mutations may lead to abnormal progesterone receptor function, thereby increasing the risk of breast cancer. Since PRs are an estrogen-responsive gene, plasma estradiol levels are associated with the expression levels of PR in ER-positive breast cancer^[140]60. In breast cancer, PRs synergistically interact with ERs to regulate tumor cell growth and differentiation, and PRs are commonly reduced in recurrent breast cancer^[141]61. Nuclear factor kappa B subunit 1, an important nuclear transcription factor in the cell, is involved in the body’s inflammatory and immune responses, and it can regulate apoptosis and stress response. NF-kB hyperactivation is associated with many human diseases^[142]62. In addition, the typical pathway of NF-κB is associated with adriamycin resistance in breast cancer cells^[143]63. Additionally, NFKB1 upregulates ETS proto-oncogene 1 expression to promote breast cancer cell invasiveness^[144]64. MAOB is involved in monoamine neurotransmitters^[145]65. Studies on breast cancer cell lines have shown that estrogen-related receptors increase MAOB expression, whereas ER decreases MAOB expression, and its expression is generally reduced in ER-positive breast cancers^[146]66 Thus, formononetin targeting all these important candidates involved in tumorigenesis of ER-positive breast cancer may provide us opportunities to identify novel approaches for treating breast cancer. More importantly, most of these targets are involved in the estrogen response. Role of estrogen response in ER-positive breast cancer The estrogen signaling pathway is one of the most prominent signaling pathways in ER-positive breast cancer^[147]67. When estrogen binds to ER, it undergoes a conformational change to form an ER/estrogen complex, which regulates gene transcription and expression, thereby promoting breast cancer development and progression^[148]68. Moreover, the regulation of breast cancer tumor growth by estrogen may be an overmodification of the normal regulatory mechanisms of breast epithelial cell proliferation and differentiation. Systematic studies using mouse models have demonstrated that estrogen can control the growth of ER-positive breast cancers by inducing pituitary synthesis and prolactin secretion^[149]69. In addition, estrogen induces invasiveness and rearrangement of cytoskeletal and adhesion structures in breast cancer cells^[150]70,[151]71. Therefore, endocrine therapy is the mainstay of treatment for patients with ER-positive breast cancer, aiming to inhibit or block the effects of estrogen^[152]72and delay tumor growth. Commonly used drugs for endocrine therapy include tamoxifen, toremifene, letrozole, and anastrozole^[153]73. These drugs inhibit tumor cell growth by binding to or degrading ER and blocking estrogen signaling. Estrogen also plays a crucial role in the recurrence, development, and treatment of ER-positive breast cancer. An in-depth study of the relationship between estrogen and ER-positive breast cancer and its mechanism of action can provide new ideas and methods for clinical diagnosis and treatment. Owing to the study limitations, the results were mainly obtained using systems biology and network pharmacology analyses. These findings need to be validated through experimental studies. Supplementary Information [154]Supplementary Information.^ (202.3KB, pdf) Acknowledgements