Open Access

Effects of bisphenol A and its chemical analogues on the microenvironment and female breast cancer development (Review)

  • Authors:
    • Shreeja Tripathi
    • Katharina Kusserow
    • Kerri Palmer
    • Abigail Dodson
    • Dylan Peter Mcclurg
    • Valerie Speirs
  • View Affiliations

  • Published online on: May 26, 2025     https://doi.org/10.3892/wasj.2025.354
  • Article Number: 66
  • Copyright : © Tripathi et al. This is an open access article distributed under the terms of Creative Commons Attribution License [CC BY 4.0].

Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Female breast cancer (BC) is the fourth most commonly diagnosed malignancy globally and its incidence is rising. Endocrine‑disrupting chemicals can bind to and activate oestrogen receptors, disrupt the microenvironment and promote oncogenic pathways. Consequently, bisphenols have been associated with the potential onset of breast carcinogenesis. The present narrative review discusses bisphenol A (BPA) and three of its chemical analogues, bisphenol AF (BPAF), bisphenol S (BPS) and bisphenol S (BPF), as well as their potential role in altering the breast microenvironment. In doing so, the present review highlights the effects that BPA, and its analogues may have in driving carcinogenesis in the microenvironment through endocrine disruption, epigenetic changes, and the modulation of immune and inflammatory pathways. Further studies are required to better understand these interactions in order to develop preventative strategies and regulatory policies to mitigate the potential risk of BC development through environmental exposure to BPA, BPAF, BPS and BPF.

1. Introduction

Breast cancer (BC) is the most prevalent type of cancer among women worldwide, with 2.3 million new cases diagnosed each year (1). The aetiology of BC is multifactorial and involves genetic, hormonal, environmental and lifestyle factors. While the precise mechanisms remain poorly understood, research has identified various risk factors, including non-modifiable factors such as age, genetic predisposition (BRCA1/2 mutations) and hormone levels. Modifiable factors include lifestyle choices, the use of oral contraceptives or hormone replacement therapy (2) and parity (3). In addition, a high mammographic density has been found to be associated with an increased risk of BC development (4).

At a cellular level, the majority of BC cases originate in the epithelial lining of breast ducts and lobules, termed ductal or lobular carcinoma in situ. Once malignant cells spread to nearby structures or tissues, the cancer becomes invasive carcinoma. BC can be classified into four main molecular subtypes based on the hormone receptor (HR; i.e., oestrogen and progesterone) and the human epidermal growth factor receptor 2 (HER2) status and include luminal A (HR+ and HER2-), luminal B (HR+ and HER2+), triple-negative (HR- and HER2-), and HER2-positive (5). The current standard of care depends on curative intent or palliative approaches, and can include a combination of chemotherapy, radiotherapy and surgery. Targeted approaches and hormonal therapies are also available depending on the subtype, apart from triple-negative BC (TNBC) which is associated with the poorest prognosis (6). While epithelial cells predominantly drive BC pathogenesis, its progression is widely accepted to be influenced by components of the breast microenvironment, a rich milieu, which includes fibroblasts, adipocytes, and immune cells such as macrophages, mast cells and T-cells (7-10).

Endocrine-disrupting chemicals (EDCs) are environmental substances known for interfering with the normal functioning of the endocrine system (11). These chemicals can mimic, block, or disrupt various processes related to the production, release, transport, metabolism, binding, action, or elimination of natural hormones in the body (12). Hence, there is potential to impact cellular function and increase the risk of carcinogenesis. Exposure to EDCs can occur through ingestion, inhalation or dermal contact, for example with contaminated air, water, food, consumer products, or contaminated surfaces (11,13-15). EDCs interfere with and modulate the endocrine system, and can lead to various adverse effects, such as alterations of the hypothalamic-pituitary-adrenal axis and urogenital tract malformations, and may also contribute to the development of BC (16). By disrupting the cellular microenvironment, EDCs may affect cell signalling, communication and the extracellular matrix, contributing to a pro-carcinogenic environment.

Bisphenols are some of the most commonly recognised EDCs (11), with bisphenol A (BPA) being the most well-known. Alternatives to BPA, include bisphenol AF (BPAF), bisphenol S (BPS) and bisphenol F (BPF), which share structural similarities with BPA (17). In response to regulatory measures, use of BPA has been increasingly restricted (11) while alternatives, particularly BPS and BPF, have gained favour as replacements (18). However, despite being postulated as safer substitutes, BPAF, BPS and BPF exhibit comparable structural characteristics and demonstrate similar oestrogenic, androgenic, anti-oestrogenic and anti-androgenic activities both in vivo and in vitro (19). These properties suggest that these compounds may influence cellular processes in ways that could elevate the risk of cancer development. Therefore, a comprehensive evaluation of BPA and its alternatives is essential to fully assess their potential implications in BC development, and to determine whether these BPA substitutes are safer alternatives.

The present narrative review aimed to provide a comprehensive summary of the available literature to determine the potential role of BPA and its chemical analogues (BPAF, S and F) on the cell types that reside within the breast microenvironment (Fig. 1) and to determine how this may influence BC development.

2. Bisphenols

BPA. The structure of BPA is illustrated in Fig. 2, a synthetic xenoestrogen used in the manufacturing of polycarbonate plastics and epoxy resins (20). BPA has been extensively investigated for its effects on nuclear receptor signalling pathways and has been found to be disruptive (21). For example, high-dose BPA predominantly functions as an oestrogen receptor (ER) antagonist through modulating genomic transcription (22). Conversely, at nanomolar doses, BPA is considered to interfere with biological processes through non-genomic mechanisms facilitated by membrane signalling (23). Materials containing BPA are various and include plastic containers, tin cans, water bottles, toys, healthcare equipment and up to 2020, thermal paper (including till receipts) (24). Consumer exposure primarily occurs through food coming into contact with BPA-containing materials, such as polycarbonate baby bottles, food containers, and epoxy resin-lined food and beverage cans. The impact of BPA on human health has led to the implementation of various regulatory measures aimed at limiting lifetime exposure. For instance, the European Food Safety Authority (EFSA) has reduced the recommended tolerable daily intake (TDI) of BPA from 4 µg/kg bodyweight/day (set in 2015) to 0.2 ng/kg bodyweight/day (set in 2023) (25), a 20,000-fold decrease. Additionally, the use of BPA in baby products was completely banned in the EU from 2011 onwards (26). In 2024, the EFSA issued a statement for phasing out BPA and its analogues in consumer products (27). By contrast, in the USA, the TDI has not been changed since it was first introduced in the 1980s and still stands at 50 µg/kg bodyweight/day (28). Furthermore, numerous petitions with new evidence indicating the potential adverse effects of BPA have failed to initiate a review or reduction of the current TDI (29).

BPA chemical analogues. The growing public awareness surrounding the potential adverse effects of BPA on human health has placed manufacturers under significant pressure to eliminate their use, resulting in the emergence of ‘BPA-free’ products. However, many of these contain BPA analogues, including BPAF, BPS and BPF (Fig. 2), whose use currently remains unregulated (30). Despite research efforts into the potential oestrogenic effects of these alternatives, the available literature remains limited. A single systematic review of 32 studies highlighted the significant knowledge gaps on their impact (19). Nonetheless, findings from preclinical studies have shown considerable endocrine-disrupting effects of BPA, BPS and BPF at micromolar values (31,32). Phenotypic and proteomic changes were observed within a human-derived normal breast organoid model exposed to 15 nM BPA, BPS and BPF. This included the disruption of tissue architecture and abnormal branching (33). Another study on human breast cancer cell lines demonstrated that BPAF was a more potent activator of ERα than BPA (34). Moreover, animal studies have shown that BPS and BPF have direct oestrogenic activity; for instance, BPS and BPF can induce uterine growth in rodents (35,36) and disrupt reproduction in zebrafish (37).

3. Bisphenols and the breast microenvironment

Investigations into the potential oestrogenic effects of BPA and its analogues have been extensive in normal and cancerous breast epithelial cells, as reviewed extensively elsewhere (38-40). The present review provides insight into the potential effects of BPA on other cells found within the breast microenvironment, including fibroblasts, adipocytes and immune cells.

Fibroblasts. Fibroblasts are the primary cellular constituents of the breast stroma and undergo activation and proliferation in response to various stimuli, including inflammation, wound repair and malignancy (41,42). In addition, fibroblasts can regulate mammary epithelial cell morphogenesis, including expansion, differentiation, ductal elongation and invasion into adipose tissue (43). The development of fibrous connective tissue, typically in response to injury or damage, results in fibrosis, a process known to be associated with an increased risk of malignancy (44). Fibroblasts and myofibroblasts undergo genetic alterations, predisposing them to transform into cancer-associated fibroblasts (CAFs), which play a pivotal role in tumour development, stromal remodelling, and intercellular communication pathways (45). Collectively, these actions create a tumour-supportive microenvironment and contribute to resistance to anticancer therapies (46).

Previous studies on the immune regulation of mammary fibroblasts have highlighted the role of immune cells in regulating fibroblast function within the mammary gland, which impacts mammographic density, a known risk factor for BC development (47-50). Mammographic density is dependent on the proportions of epithelial and fibrous stromal tissues. Increased mammographic density is analogous with a higher content of fibrous tissue within the stroma and is associated with a 4-6-fold increased risk of developing BC (50). Studies have shown that exposure to BPA promotes collagen production by cardiac fibroblasts via the ERK1/2 pathway (51), and is associated with an elevated mammographic density (52) and increased mammary gland rigidity, a property which is known to enhance proliferation and tumour progression (53). Fibroblast-driven paracrine signalling and extracellular matrix remodelling, as mediated by the EGFR pathway; this further influences epithelial morphogenesis and may play a role in tumour initiation (54).

A previous study investigating the effects of BPA on co-cultures of CAFs and breast cancer cells, demonstrated that BPA mediated CAF proliferation and SkBr3 migration through the activation of the G-protein coupled ER pathway (55). Primary mammary fibroblasts from mouse mammary glands exposed to BPA exhibit the differential expression of genes commonly associated with an increased risk of developing BC (56).

BPA exposure significantly alters the gene expression profiles of mesenchymal cells, including fibroblasts, derived from murine mammary glands (57), disrupting their function and behaviour by influencing pathways associated with extracellular matrix production. These disruptions could potentially alter fibroblast-epithelial interactions, affecting mammary gland development and potentially predisposing the tissue to developing malignancy.

There are a limited number of studies investigating the effects of BPS and BPF on fibroblasts. Potential cytotoxic effects of these compounds on mammalian fibroblasts, at 50-500 mM doses has been reported; however, the use of such high concentrations is questionable compared to real-world exposure levels (58). In MRC5 human lung fibroblasts exposed to up to 100 µM BPA, no effects were observed on proliferation, cell cycle progression or apoptosis (59).

Adipocytes. Adipocytes are abundant in the breast tumour microenvironment (TME). White adipocytes are a major component and primarily store energy as triacylglycerol, releasing this as fatty acids and glycerol under high metabolic demand (60). Breast adipocytes are increasingly associated with BC development and progression (61). In a previous study using mice exposed to oral BPA (5, 50, 500 and 5,000 µg/kg/day) alongside normal chow, bodyweight and adiposity increased, indicating the potential interaction of BPA with diet (62). Furthermore, BPA increased circulating inflammatory factors leptin and tumour necrosis factor (TNF)-α in lean female, but not male mice (62). At an endocrine level, obesity poses a significant risk factor for ER+ BC due to the aromatisation of androgens to oestrogens in adipocytes, which elevates the overall oestrogen levels (63). This also extends to BC in males, where research has demonstrated that the increasing number of males receiving a BC diagnosis parallels with an increase in obesity (64).

Some endocrine disruptors can modify inflammatory responses in adipocytes (62). Cancer-associated adipocytes (CAAs) are adipocytes located close to the tumour leading edge and are characterised by loss of lipid content, loss of mature adipocyte markers, such as peroxisome proliferator-activated receptor γ (PPARγ), and an increase in adipokines, primarily leptin and resistin, which can promote BC development (65-67). Additionally, CAAs display an increased secretion of inflammatory mediators, including interleukin (IL)-1β, IL-6(68), vascular endothelial growth factor (VEGF) and TNF-α, potentially in response to cancer cells (69). Moreover, CAAs release adipokines that promote cancer cell migration, such as IL-6 and monocyte chemoattractant protein-1 (MCP-1) (70), at higher levels compared to non-cancer adipocytes (70). BPA increases the expression of these adipokines to help facilitate their role in breast carcinogenesis. For example, in a previous study, when mature human adipocytes were treated with 0.1 nM BPA, the G protein-coupled oestrogen receptor pathway was activated, leading to the release of IL-6, IL-8 and MCP1α (23). Both IL-6 and MCP-1α can promote cancer cell proliferation and migration, and IL-8 enhances the invasiveness and angiogenesis of BC cells, particularly ER+ subtypes (68,71). The implication of these findings is that BPA may cause adipocytes in the breast microenvironment to release pro-inflammatory cytokines, potentially accelerating tumour development and growth.

BPA has also been implicated in altering hormone levels in adipose tissue by modulating the activity of key regulators, such as lipoprotein lipase and aromatase (72,73). Although limited, studies on the effects of BPA on breast adipocytes have shown that nanomolar concentrations of BPA do not stimulate the proliferation of adipocytes in human subcutaneous adipose tissue (74). However, several studies have demonstrated that BPA accelerates adipocyte differentiation into mature cells in vitro (75-80). For instance, murine 3T3-L1 cells and adipocyte stem cells undergo accelerated differentiation into mature adipocytes upon exposure to low concentrations of BPA (10-80 µM) (75-80). Mature adipocytes are crucial for secreting adipokines, such as adiponectin, which have anti-inflammatory and antioxidative properties, and can inhibit cell proliferation and angiogenesis (81). In addition, adipokines promoted apoptosis in diabetes (82). If these effects are mirrored in BC, this may provide potential protective effects. Conversely, exposure to 0.1 and 1 nM of BPA has been shown to decrease adiponectin release from human breast adipose tissue (83). In metabolic disorders, adiponectin functions as a protective factor, promoting insulin sensitivity and exhibiting anti-inflammatory and anti-tumorigenic properties (84).

Both preadipocytes and mature adipocytes significantly influence mammary gland development, with preadipocytes promoting epithelial branching and elongation, as demonstrated in mouse models (85). In vivo, murine studies have indicated that prolonged exposure to both 1 and 10 µg/ml BPA, classified as low and high concentrations, respectively, influences obesity and hyperlipidaemia development during perinatal and postnatal periods (86). In another study, the in utero exposure of mice to BPA at 250 ng/kg/day increased the expression of PPARγ and other adipogenesis-related genes (57). In addition, BPA exposure has been shown to increase the concentration of mature adipocytes, negatively influencing lumen formation in the foetal mammary gland epithelium (57,87). Additionally, oestradiol sensitivity is increased (88), potentially elevating risk of carcinogenesis. Collectively, these results suggest that, at least in mouse models, in utero BPA exposure may be a critical window for abnormal changes in epithelial, stromal and adipose cells, potentially enhancing carcinogenic potential in adulthood.

BPA alternatives, BPAF, BPF and BPS have been shown to alter leptin hormone levels and increase lipid accumulation in the murine 3T3L1 cell line (89). Leptin is secreted by adipocytes and plays a critical role in energy balance and appetite regulation. It also exerts pro-inflammatory effects and may affect cancer progression (90). In a previous study, in 3T3-L1 cells, exposure to 32 µM BPS and BPF increased the number of differentiated adipocytes and their ability to store more lipids compared to BPA, indicating a higher obesogenic potential and endocrine toxicity (91). Furthermore, in adult mice, doses of both 10 nM and 1 µM BPS resulted in the upregulation of the expression of PPARγ and perilipin 1, both involved in preadipocyte differentiation, much earlier in the differentiation process than BPA and BPF. Hence, BPS exposure may contribute to increased adiposity and an increased risk of developing BC. However, the effects of BPF on adiposity are reduced (92). These contrasting findings suggest uncertainty over the potential influence of BPS and BPF on the risk of developing BC. On the other hand, a previous study demonstrated that BPAF treatment (5 µM) increased lipid accumulation and sensitivity to inflammatory cytokines, including interferon-γ, in differentiating adipocytes (93). This increased sensitivity resulted in a reduction in mitochondrial and cellular respiratory capacity in adipocytes through the suppression of UCP1 by interferon-γ, which may also influence BC progression (93). Collectively, these findings demonstrate the potential impact of BPA and its analogues on adipocyte function and the putative link of bisphenols in increasing breast carcinogenesis.

Immune cells. Different immune cell populations often infiltrate breast cancers. This varies between different BC subtypes. For example, TNBC often has high numbers of tumour-infiltrating lymphocytes, which is associated with an improved survival (94). Several studies have identified the importance of lymphoid and myeloid cells, which are primarily located within the epithelium of breast lobules, in breast carcinogenesis, progression and treatment (95-97).

Lymphoid cells. Lymphoid cells, including lymphocytes, such as T-cells, B-cells and natural killer (NK) cells are key components of the immune system. In a previous study, human T-cells exposed to nanomolar concentrations of BPA exhibited a reduced telomerase activity in CD8+ but not CD4+ T-cells, accompanied by telomere shortening and human telomerase reverse transcriptase suppression, suggesting adverse effects of BPA on T-cell responses (98). In another study, a 30-day chronic exposure of the human BC cell lines, MCF-7, SkBr3 and MDA-MB-231 to physiologically relevant BPA concentrations (10 nM) upregulated the expression of genes associated with NK cell and T-cell activation, suggesting the potential of BPA to modulate immune function (99). Notably, each cell line exhibited distinct gene expression changes related to immune responses. In MCF-7 cells, IL-19 expression was upregulated, while in SkBr3 cells, the upregulation of CXCL5 was linked to BC progression (99). Further research has highlighted an abundance of NK cells in HER2+ and TNBC subtypes, with their presence being associated with increased lymphocyte infiltration and a higher grade (100), suggesting that NK cell activation is associated with more advanced BC. These findings underscore the complex immune-related gene expression alterations induced by BPA, implicating its potential role in BC progression through immune modulation.

Myeloid cells. Myeloid cells, including monocytes, macrophages and granulocytes contribute to TMEs by modulating immune responses, promoting inflammation, and supporting tumour growth and metastasis. The immune subtyping of myeloid cells in murine breast cancer models and in clinical datasets has identified neutrophil- and macrophage-enriched subtypes (101).

Neutrophils circulate in the bone marrow and peripheral blood, recognising pathogens through host protein interaction, aiding in phagocytosis, pathogen clearance and tissue regeneration. To the best of our knowledge, no studies to date have examined the effect of BPA on neutrophils in BC. However, an examination of the effects of BPA on human T-cells in vitro indicated increased reactive oxygen species (ROS) production, which is associated with breast carcinogenesis (102).

Macrophages are classified into two main types: M1, which is associated with anti-tumour immunity, and M2, which is linked to pro-tumorigenic properties (103). Evidence from animal models suggests that BPA may alter tumour-associated macrophage (TAM) phenotypes (104). Another study assessed the effects of BPA on macrophage polarisation, demonstrating a significant increase in M2 markers (Arg-1 and CD206) and a reduction in TAMs with M1 markers in ductal carcinomas exposed to 10 nM BPA, which is typically considered an environmentally relevant concentration (105).

BPA alternatives may also influence immune cell activation and differentiation (106). While there are no BC studies, at least to the best of our knowledge, studies using zebrafish and carp have demonstrated that BPS and BPF exert immunotoxic effects by modulating immunoregulatory genes in a concentration-dependent manner during early development. Furthermore, exposure to BPF increases the levels of immunomodulating chemokines and cytokines and is associated with elevated ROS levels, oxidative stress and the upregulation of inflammatory cytokine genes (107,108). The effects of BPA on human myeloid cells in BC macrophages have yet to be determined.

4. Discussion and future directions

The present narrative review demonstrates that BPA and its chemical analogues; BPAF, BPS and BPF, exert diverse effects on the breast microenvironment. Some of these effects may promote BC development, for example, through structural and epigenetic changes to epithelial cells, the disruption of fibroblast functions and stromal modifications, altered adipocyte differentiation, and influences on lymphoid immune activity. Previous studies on the effects of BPA on the female breast microenvironment have mainly focused on BPA, overlooking its analogues and the critical role of non-epithelial cells in the breast milieu. To the best of our knowledge, the present review is the first to systematically compare the differential effects of BPA and its analogues on adipocytes, providing new insight into how these compounds may alter breast tissue composition and function. In addition, the present review also highlights safety concerns regarding BPA analogues and the urgent need for regulatory guidelines.

As demonstrated herein, one of the most critical issues surrounding research on BPA is the reliance on animal models and in vitro cell lines, which take a reductionist approach and fail to fully capture the complexity of the breast microenvironment. While these models have provided valuable insight, their translational relevance remains limited. For instance, current animal model limitations include murine mammary tissue, which contains terminal end buds (TEBs) vs. terminal ductal lobular units (TDLUs) found in humans. TDLUs are specialised structures containing collagen, hyaluronan and other matrix proteins, and give rise to primary sites where ductal carcinomas can occur (109). By contrast, mouse TEBs lack such specialised stroma, primarily comprising adipose tissue with few fibroblasts (109). Furthermore, research has highlighted that murine mammary cytokines may not functionally match human receptor interactions, limiting the translational relevance of these models (110). To address these shortcomings, recent advancements have introduced humanised models including organoid cultures, 3D co-culture systems and clinical sample analyses, which better replicate the breast microenvironment by preserving multicellular contacts and immune interactions. Incorporating these models could improve the clinical applicability of BPA research in future. Additionally, the majority of available studies have examined single compounds and have overlooked the combined effects of multiple chemicals or ‘cocktail effects’, which may lead to underestimating the real-world risks. Future research is thus warranted to incorporate experimental designs that consider mixed bisphenol exposures to provide a closer representation of the environmental risks. Moreover, the heavy reliance on cell lines does not address the potential role of BPA in initiating breast carcinogenesis.

To overcome these limitations, future studies are required to focus on the effects of BPA and its analogues on all cells within the breast microenvironment, particularly in the context of cancer-stromal interactions, using multicellular 3D models (111). A critical aspect of BPA-induced breast carcinogenesis may lie in its ability to remodel the TME. CAFs, key components of the TME, play an essential role in metabolic symbiosis with tumour cells and contribute to immune escape mechanisms by modulating cytokine signalling. Bisphenols may enhance CAF activation, leading to extracellular matrix remodelling, increased tumour invasiveness and altered immune responses. Thus, further studies using tumour niche models and stroma-epithelium interaction assays are required to elucidate the mechanisms through which BPA and its analogues promote breast cancer progression. Studies should also aim to use more reflective models and novel technological advances to better understand BPA and its alternatives, and their impact on the TME, as well as to determine safe cut-off levels to guide regulatory recommendations to prevent toxic harm. The use of organoids derived from cancerous and non-cancerous human breast tissue is recommended (112), that can recapitulate their primary tissue both phenotypically and molecularly (113). Although definitive evidence is currently lacking, an observational study of male BC across various regions of Scotland found higher incidence rates in areas with significant agricultural activity, where the use of pesticides and EDCs may be more prevalent (114). As such, tissue collection efforts should include both males and females (114). Multi-cellular 3D models of cell lines (115), co-culture approaches (116) and the use of novel bioprinting techniques (117), would enable a more insightful representation of microenvironmental complexity. In addition, the use of breast-on-a-chip models would enable microfluidic techniques to create controlled, miniaturised environments for studying dynamic tissue interactions with greater precision (118). To permit the comprehensive mapping of cellular interactions and molecular changes within breast tissue exposed to BPA and its analogues, the use of emerging spatial multi-omics technologies should also be utilised (119). Drawing insights from the Human Breast Cell Atlas (120), a comprehensive map of cell types found within the normal human breast, may inform these studies and enhance our understanding of the tissue-specific impacts of bisphenols. Lastly, a thorough investigation into both the short- and long-term effects of BPA on various mammary cells, alongside sensitivity testing across low, medium, and high BPA exposure levels, is crucial for making any meaningful study comparisons and guiding regulatory decisions on safe exposure limits.

Another observation was the wide variation in doses of bisphenols used experimentally, ranging from 500 mM (58) to 10 nM (105). Optimum concentrations should be defined to allow their biological effects to be defined more rigorously. It is also recognised that these chemicals are pervasive in the environment which may be confounding, although this is difficult to control.

Finally, from a regulatory and public health perspective, there is a lack of consensus regarding the threshold for BPA exposure, making it challenging to define what constitutes ‘safe’ exposure levels. This inconsistency is particularly evident when comparing international guidelines; for instance, the US TDI currently stands at 50 µg/kg body weight/day, which is 250,000-fold higher than the European TDI of 0.2 ng/kg bodyweight/day (28,121). In the UK, recent estimates indicate that average daily exposure to BPA is ~2.5 ng/kg body weight/day, depending on dietary habits and environmental factors (122); however, precise numbers are difficult to assess. Moreover, the term ‘chronic BPA exposure’ is inconsistently defined among studies, impeding cross-study comparability. Consequently, the potential impact of bioaccumulation on breast carcinogenesis remains largely unexplored. It is also important to highlight that BPAF, BPS and BPF currently lack regulatory oversight, a concern underscored by the findings presented herein, as these unregulated compounds may be driving the same pheno- and genotypic changes as found in BPA.

Acknowledgements

Not applicable.

Funding

Funding: The present study received funding from Breast Cancer UK (RG/2017-VS; 2122029), Animal Free Research UK and the Cyril Margaret Gates Charitable Trust.

Availability of data and materials

Not applicable.

Authors' contributions

ST was involved in the writing, reviewing and editing of the manuscript and in the writing of the original draft, as well as in visualization (figure preparation) and project administration. KK was involved in the writing, reviewing and editing of the manuscript. KP and AD were involved in the writing, reviewing and editing of the manuscript, as well as in data curation (assessing all the relevant research articles for inclusion in the review). DPMC was involved in the writing, reviewing and editing of the manuscript, in visualization and study supervision, as well as in project administration, methodology, investigation, formal analysis, data curation and in the conceptualisation of the study. VS was involved in the writing, reviewing and editing of the manuscript, as well as in visualization, study supervision and project administration, resources (funding for the study and the tissue image), and in the conceptualisation of the study.

Ethics approval and consent to participate

The tissue image included in Fig. 1 was generated from an anonymised breast tissue sample donated with ethical approval from the Leeds Breast Tissue Bank (REC 15/YH/0025), whose samples now reside in the Breast Cancer Now Biobank (REC 23/EE/0229). As the sample was provided with full anonymisation, the donor cannot be identified meaning that written informed patient consent was not required.

Patient consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

References

1 

Sung H, Ferlay J, Siegel RL, Laversanne M, Soerjomataram I, Jemal A and Bray F: Global cancer statistics 2020: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J Clin. 71:209–249. 2021.PubMed/NCBI View Article : Google Scholar

2 

Lippman ME, Krueger KA, Eckert S, Sashegyi A, Walls EL, Jamal S, Cauley JA and Cummings SR: Indicators of lifetime estrogen exposure: effect on breast cancer incidence and interaction with raloxifene therapy in the multiple outcomes of raloxifene evaluation study participants. J Clin Oncol. 19:3111–3116. 2001.PubMed/NCBI View Article : Google Scholar

3 

Nguyen B, Venet D, Lambertini M, Desmedt C, Salgado R, Horlings HM, Rothé F and Sotiriou C: Imprint of parity and age at first pregnancy on the genomic landscape of subsequent breast cancer. Breast Cancer Res. 21(25)2019.PubMed/NCBI View Article : Google Scholar

4 

Boyd NF, Rommens JM, Vogt K, Lee V, Hopper JL, Yaffe MJ and Paterson AD: Mammographic breast density as an intermediate phenotype for breast cancer. Lancet Oncol. 6:798–808. 2005.PubMed/NCBI View Article : Google Scholar

5 

Perou CM, Sørile T, Eisen MB, van de Rijn M, Jeffrey SS, Rees CA, Pollack JR, Ross DT, Johnsen H, Akslen LA, et al: Molecular portraits of human breast tumours. Nature. 406:747–752. 2000.PubMed/NCBI View Article : Google Scholar

6 

Pareja F, Geyer FC, Marchiò C, Burke KA, Weigelt B and Reis-Filho JS: Triple-negative breast cancer: The importance of molecular and histologic subtyping, and recognition of low-grade variants. NPJ Breast Cancer. 2(16036)2016.PubMed/NCBI View Article : Google Scholar

7 

Soysal SD, Tzankov A and Muenst SE: Role of the tumor microenvironment in breast cancer. Pathobiology. 82:142–152. 2015.PubMed/NCBI View Article : Google Scholar

8 

Terceiro LEL, Edechi CA, Ikeogu NM, Nickel BE, Hombach-Klonisch S, Sharif T, Leygue E and Myal Y: The breast tumor microenvironment: A key player in metastatic spread. Cancers (Basel). 13(4798)2021.PubMed/NCBI View Article : Google Scholar

9 

Weigelt B and Bissell MJ: Unraveling the microenvironmental influences on the normal mammary gland and breast cancer. Semin Cancer Biol. 18:311–321. 2008.PubMed/NCBI View Article : Google Scholar

10 

Mittal S, Brown NJ and Holen I: The breast tumor microenvironment: Role in cancer development, progression and response to therapy. Expert Rev Mol Diagn. 18:227–243. 2018.PubMed/NCBI View Article : Google Scholar

11 

Diamanti-Kandarakis E, Bourguignon JP, Giudice LC, Hauser R, Prins GS, Soto AM, Zoeller RT and Gore AC: Endocrine-disrupting chemicals: An endocrine society scientific statement. Endocr Rev. 30:293–342. 2009.PubMed/NCBI View Article : Google Scholar

12 

Yilmaz B, Terekeci H, Sandal S and Kelestimur F: Endocrine disrupting chemicals: Exposure, effects on human health, mechanism of action, models for testing and strategies for prevention. Rev Endocr Metab Disord. 21:127–147. 2020.PubMed/NCBI View Article : Google Scholar

13 

Gore AC, Chappell VA, Fenton SE, Flaws JA, Nadal A, Prins GS, Toppari J and Zoeller RT: Executive summary to EDC-2: The endocrine society's second scientific statement on endocrine-disrupting chemicals. Endocr Rev. 36:593–602. 2015.PubMed/NCBI View Article : Google Scholar

14 

Zoeller RT, Brown TR, Doan LL, Gore AC, Skakkebaek NE, Soto AM, Woodruff TJ and Vom Saal FS: Endocrine-disrupting chemicals and public health protection: A statement of principles from the endocrine society. Endocrinology. 153:4097–4110. 2012.PubMed/NCBI View Article : Google Scholar

15 

Cwiek-Ludwicka K and Ludwicki JK: Endocrine disruptors in food contact materials; is there a health threat? Rocz Panstw Zakl Hig. 65:169–177. 2014.PubMed/NCBI

16 

La Merrill MA, Vandenberg LN, Smith MT, Goodson W, Browne P, Patisaul HB, Guyton KZ, Kortenkamp A, Cogliano VJ, Woodruff TJ, et al: Consensus on the key characteristics of endocrine-disrupting chemicals as a basis for hazard identification. Nat Rev Endocrinol. 16:45–57. 2020.PubMed/NCBI View Article : Google Scholar

17 

Brody JG and Rudel RA: Environmental pollutants and breast cancer. Environ Health Perspect. 111:1007–1019. 2003.PubMed/NCBI View Article : Google Scholar

18 

Hawke E: Human biomonitoring results reveal widespread exposure of general public to harmful chemicals. https://chemtrust.org/hbm4eu_conference/. Accessed on December 4, 2024.

19 

Rochester JR and Bolden AL: Bisphenol S and F: A systematic review and comparison of the hormonal activity of bisphenol A substitutes. Environ Health Perspect. 123:643–650. 2015.PubMed/NCBI View Article : Google Scholar

20 

European Food Safety Authority: Bisphenol A | EFSA, 2023. https://www.efsa.europa.eu/en/topics/topic/bisphenol. Accessed on December 11, 2024.

21 

Liu X, Sakai H, Nishigori M, Suyama K, Nawaji T, Ikeda S, Nishigouchi M, Okada H, Matsushima A, Nose T, et al: Receptor-binding affinities of bisphenol A and its next-generation analogs for human nuclear receptors. Toxicol Appl Pharmacol. 377(114610)2019.PubMed/NCBI View Article : Google Scholar

22 

Nadal A, Fuentes E, Ripoll C, Villar-Pazos S, Castellano-Muñoz M, Soriano S, Martinez-Pinna J, Quesada I and Alonso-Magdalena P: Extranuclear-initiated estrogenic actions of endocrine disrupting chemicals: Is there toxicology beyond paracelsus? J Steroid Biochem Mol Biol. 176:16–22. 2018.PubMed/NCBI View Article : Google Scholar

23 

Cimmino I, Oriente F, D'Esposito V, Liguoro D, Liguoro P, Ambrosio MR, Cabaro S, D'Andrea F, Beguinot F, Formisano P and Valentino R: Low-dose bisphenol-A regulates inflammatory cytokines through GPR30 in mammary adipose cells. J Mol Endocrinol. 63:273–283. 2019.PubMed/NCBI View Article : Google Scholar

24 

European Union: Comission Regulation (EU) 2016/2235, 2016. https://eur-lex.europa.eu/eli/reg/2016/2235/oj/eng. Accessed on December 13, 2024.

25 

EFSA Panel on Food Contact Materials, Enzymes and Processing Aids (CEP). Lambré C, Barat Baviera JM, Bolognesi C, Chesson A, Cocconcelli PS, Crebelli R, Gott DM, Grob K, Lampi E, et al: Re-evaluation of the risks to public health related to the presence of bisphenol A (BPA) in foodstuffs. EFSA J. 21(e06857)2023.PubMed/NCBI View Article : Google Scholar

26 

European Commision: Internal Market, Industry, Entrepreneurship and Smes-Bisphenol A: EU ban on use in baby bottles. https://ec.europa.eu/newsroom/growth/items/44933/en. Accessed on December 4, 2024.

27 

European Commission: European Commission-Daily News, 2024. https://ec.europa.eu/commission/presscorner/api/files/document/print/en/mex_24_3243/MEX_24_3243_EN.pdf. Accessed on December 11, 2024.

28 

U.S. Environmental Protection Agency: Bisphenol A. (CASRN 80-05-7) | IRIS | US EPA. https://cfpub.epa.gov/ncea/iris/iris_documents/documents/subst/0356_summary.pdf. Accessed on December 4, 2024.

29 

U.S. Food and Drug Administration (FDA): Bisphenol A (BPA) | FDA. FDA, Silver Spring, MD, 2023. https://www.fda.gov/food/food-packaging-other-substances-come-contact-food-information-consumers/bisphenol-bpa. Accessed on December 4, 2024.

30 

European Chemicals Agency (ECHA): Assessment of regulatory needs. ECHA, Helsinki, 2021. https://echa.europa.eu/documents/10162/7de6871f-30db-9cdc-0a13-20942f511e00. Accessed on December 11, 2024.

31 

Atlas E and Dimitrova V: Bisphenol S and Bisphenol A disrupt morphogenesis of MCF-12A human mammary epithelial cells. Sci Rep. 9(16005)2019.PubMed/NCBI View Article : Google Scholar

32 

Kim JY, Choi HG, Lee HM, Lee GA, Hwang KA and Choi KC: Effects of bisphenol compounds on the growth and epithelial mesenchymal transition of MCF-7 CV human breast cancer cells. J Biomed Res. 31(358)2017.PubMed/NCBI View Article : Google Scholar

33 

Winkler J, Liu P, Phong K, Hinrichs JH, Ataii N, Williams K, Hadler-Olsen E, Samson S, Gartner ZJ, Fisher S and Werb Z: Bisphenol A replacement chemicals, BPF and BPS, induce protumorigenic changes in human mammary gland organoid morphology and proteome. Proc Natl Acad Sci USA. 119(e2115308119)2022.PubMed/NCBI View Article : Google Scholar

34 

Mesnage R, Phedonos A, Arno M, Balu S, Corton JC and Antoniou MN: Editor's highlight: Transcriptome profiling reveals bisphenol A alternatives activate estrogen receptor alpha in human breast cancer cells. Toxicol Sci. 158:431–443. 2017.PubMed/NCBI View Article : Google Scholar

35 

Yamasaki K, Noda S, Imatanaka N and Yakabe Y: Comparative study of the uterotrophic potency of 14 chemicals in a uterotrophic assay and their receptor-binding affinity. Toxicol Lett. 146:111–120. 2004.PubMed/NCBI View Article : Google Scholar

36 

Stroheker T, Chagnon MC, Pinnert MF, Berges R and Canivenc-Lavier MC: Estrogenic effects of food wrap packaging xenoestrogens and flavonoids in female Wistar rats: A comparative study. Reprod Toxicol. 17:421–432. 2003.PubMed/NCBI View Article : Google Scholar

37 

Ji K, Hong S, Kho Y and Choi K: Effects of bisphenol S exposure on endocrine functions and reproduction of zebrafish. Environ Sci Technol. 47:8793–8800. 2013.PubMed/NCBI View Article : Google Scholar

38 

Darbre PD: Endocrine disrupting chemicals and breast cancer cells. Adv Pharmacol. 92:485–520. 2021.PubMed/NCBI View Article : Google Scholar

39 

Wan MLY, Co VA and El-Nezami H: Endocrine disrupting chemicals and breast cancer: A systematic review of epidemiological studies. Crit Rev Food Sci Nutr. 62:6549–6576. 2022.PubMed/NCBI View Article : Google Scholar

40 

Vandenberg LN: Endocrine disrupting chemicals and the mammary gland. Adv Pharmacol. 92:237–277. 2021.PubMed/NCBI View Article : Google Scholar

41 

Avagliano A, Granato G, Ruocco MR, Romano V, Belviso I, Carfora A, Montagnani S and Arcucci A: Metabolic reprogramming of cancer associated fibroblasts: The slavery of stromal fibroblasts. Biomed Res Int. 2018(6075403)2018.PubMed/NCBI View Article : Google Scholar

42 

Camps JL, Chang SM, Hsu TC, Freeman MR, Hong SJ, Zhau HE, von Eschenbach AC and Chung LW: Fibroblast-mediated acceleration of human epithelial tumor growth in vivo. Proc Natl Acad Sci USA. 87:75–79. 1990.PubMed/NCBI View Article : Google Scholar

43 

Bissell MJ, Rizki A and Mian IS: Tissue architecture: The ultimate regulator of breast epithelial function. Curr Opin Cell Biol. 15:753–762. 2003.PubMed/NCBI View Article : Google Scholar

44 

Kuziel G, Moore BN and Arendt LM: Obesity and fibrosis: Setting the stage for breast cancer. Cancers (Basel). 15(2929)2023.PubMed/NCBI View Article : Google Scholar

45 

Ye F, Liang Y, Wang Y, Le Yang R, Luo D, Li Y, Jin Y, Han D, Chen B, Zhao W, et al: Cancer-associated fibroblasts facilitate breast cancer progression through exosomal circTBPL1-mediated intercellular communication. Cell Death Dis. 14(471)2023.PubMed/NCBI View Article : Google Scholar

46 

Hu D, Li Z, Zheng B, Lin X, Pan Y, Gong P, Zhuo W, Hu Y, Chen C, Chen L, et al: Cancer-associated fibroblasts in breast cancer: Challenges and opportunities. Cancer Commun (Lond). 42:401–434. 2022.PubMed/NCBI View Article : Google Scholar

47 

Archer M, Dasari P, Walsh D, Britt KL, Evdokiou A and Ingman WV: Immune regulation of mammary fibroblasts and the impact of mammographic density. J Clin Med. 11(799)2022.PubMed/NCBI View Article : Google Scholar

48 

Unsworth A, Anderson R and Britt K: Stromal fibroblasts and the immune microenvironment: Partners in mammary gland biology and pathology? J Mammary Gland Biol Neoplasia. 19:169–182. 2014.PubMed/NCBI View Article : Google Scholar

49 

Thurfjell E: Breast density and the risk of breast cancer. N Engl J Med. 347(866)2002.PubMed/NCBI View Article : Google Scholar

50 

McCormack VA and dos Santos Silva I: Breast density and parenchymal patterns as markers of breast cancer risk: A meta-analysis. Cancer Epidemiol Biomarkers Prev. 15:1159–1169. 2006.PubMed/NCBI View Article : Google Scholar

51 

Hu Y, Zhang L, Wu X, Hou L, Li Z, Ju J, Li Q, Qin W, Li J, Zhang Q, et al: Bisphenol A, an environmental estrogen-like toxic chemical, induces cardiac fibrosis by activating the ERK1/2 pathway. Toxicol Lett. 250-251:1–9. 2016.PubMed/NCBI View Article : Google Scholar

52 

Sprague BL, Trentham-Dietz A, Hedman CJ, Wang J, Hemming JD, Hampton JM, Buist DS, Aiello Bowles EJ, Sisney GS and Burnside ES: Circulating serum xenoestrogens and mammographic breast density. Breast Cancer Res. 15(R45)2013.PubMed/NCBI View Article : Google Scholar

53 

Schedin P and Keely PJ: Mammary gland ECM remodeling, stiffness, and mechanosignaling in normal development and tumor progression. Cold Spring Harb Perspect Biol. 3(a003228)2011.PubMed/NCBI View Article : Google Scholar

54 

Koledova Z, Zhang X, Streuli C, Clarke RB, Klein OD, Werb Z and Lu P: SPRY1 regulates mammary epithelial morphogenesis by modulating EGFR-dependent stromal paracrine signaling and ECM remodeling. Proc Natl Acad Sci USA. 113:E5731–E5740. 2016.PubMed/NCBI View Article : Google Scholar

55 

Pupo M, Pisano A, Lappano R, Santolla MF, De Francesco EM, Abonante S, Rosano C and Maggiolini M: Bisphenol A induces gene expression changes and proliferative effects through GPER in breast cancer cells and cancer-associated fibroblasts. Environ Health Perspect. 120:1177–1182. 2012.PubMed/NCBI View Article : Google Scholar

56 

Wormsbaecher C, Hindman AR, Avendano A, Cortes-Medina M, Jones CE, Bushman A, Onua L, Kovalchin CE, Murphy AR, Helber HL, et al: In utero estrogenic endocrine disruption alters the stroma to increase extracellular matrix density and mammary gland stiffness. Breast Cancer Res. 22(41)2020.PubMed/NCBI View Article : Google Scholar

57 

Wadia PR, Cabaton NJ, Borrero MD, Rubin BS, Sonnenschein C, Shioda T and Soto AM: Low-dose BPA exposure alters the mesenchymal and epithelial transcriptomes of the mouse fetal mammary gland. PLoS One. 8(e63902)2013.PubMed/NCBI View Article : Google Scholar

58 

Hyun M, Rathor L, Kim HJ, McElroy T, Hwang KH, Wohlgemuth S, Curry S, Xiao R, Leeuwenburgh C, Heo JD and Han SM: Comparative toxicities of BPA, BPS, BPF, and TMBPF in the nematode Caenorhabditis elegans and mammalian fibroblast cells. Toxicology. 461(152924)2021.PubMed/NCBI View Article : Google Scholar

59 

Kim JY, Shin GS, Kim CH, Kim MJ, An MJ, Lee HM and Kim JW: The cytotoxic effects of bisphenol A alternatives in human lung fibroblast MRC5 cells. Mol Cell Toxicol. 17:267–276. 2021.

60 

Li Q and Spalding KL: The regulation of adipocyte growth in white adipose tissue. Front Cell Dev Biol. 10(1003219)2022.PubMed/NCBI View Article : Google Scholar

61 

Vaysse C, Lømo J, Garred Ø, Fjeldheim F, Lofteroed T, Schlichting E, McTiernan A, Frydenberg H, Husøy A, Lundgren S, et al: Inflammation of mammary adipose tissue occurs in overweight and obese patients exhibiting early-stage breast cancer. NPJ Breast Cancer. 3(19)2017.PubMed/NCBI View Article : Google Scholar

62 

Yang M, Chen M, Wang J, Xu M, Sun J, Ding L, Lv X, Ma Q, Bi Y, Liu R, et al: Bisphenol A promotes adiposity and inflammation in a nonmonotonic dose-response way in 5-week-old male and female C57BL/6J mice fed a low-calorie diet. Endocrinology. 157:2333–2345. 2016.PubMed/NCBI View Article : Google Scholar

63 

Chetrite GS, Cortes-Prieto J, Philippe JC, Wright F and Pasqualini JR: Comparison of estrogen concentrations, estrone sulfatase and aromatase activities in normal, and in cancerous, human breast tissues. J Steroid Biochem Mol Biol. 72:23–27. 2000.PubMed/NCBI View Article : Google Scholar

64 

Humphries MP, Jordan VC and Speirs V: Obesity and male breast cancer: Provocative parallels? BMC Med. 13(134)2015.PubMed/NCBI View Article : Google Scholar

65 

Hu Y, Liu L, Chen Y, Zhang X, Zhou H, Hu S, Li X, Li M, Li J, Cheng S, et al: Cancer-cell-secreted miR-204-5p induces leptin signalling pathway in white adipose tissue to promote cancer-associated cachexia. Nat Commun. 14(5179)2023.PubMed/NCBI View Article : Google Scholar

66 

Gao Y, Chen X, He Q, Gimple RC, Liao Y, Wang L, Wu R, Xie Q, Rich JN, Shen K and Yuan Z: Adipocytes promote breast tumorigenesis through TAZ-dependent secretion of Resistin. Proc Natl Acad Sci USA. 117:33295–33304. 2020.PubMed/NCBI View Article : Google Scholar

67 

Zhu Q, Zhu Y, Hepler C, Zhang Q, Park J, Gliniak C, Henry GH, Crewe C, Bu D, Zhang Z, et al: Adipocyte mesenchymal transition contributes to mammary tumor progression. Cell Rep. 40(111362)2022.PubMed/NCBI View Article : Google Scholar

68 

Dirat B, Bochet L, Dabek M, Daviaud D, Dauvillier S, Majed B, Wang YY, Meulle A, Salles B, Le Gonidec S, et al: Cancer-associated adipocytes exhibit an activated phenotype and contribute to breast cancer invasion. Cancer Res. 71:2455–2465. 2011.PubMed/NCBI View Article : Google Scholar

69 

De Palma M, Biziato D and Petrova TV: Microenvironmental regulation of tumour angiogenesis. Nat Rev Cancer. 17:457–474. 2017.PubMed/NCBI View Article : Google Scholar

70 

Fujisaki K, Fujimoto H, Sangai T, Nagashima T, Sakakibara M, Shiina N, Kuroda M, Aoyagi Y and Miyazaki M: Cancer-mediated adipose reversion promotes cancer cell migration via IL-6 and MCP-1. Breast Cancer Res Treat. 150:255–263. 2015.PubMed/NCBI View Article : Google Scholar

71 

Todorović-Raković N and Milovanović J: Interleukin-8 in breast cancer progression. J Interferon Cytokine Res. 33:563–570. 2013.PubMed/NCBI View Article : Google Scholar

72 

Williams GP and Darbre PD: Low-dose environmental endocrine disruptors, increase aromatase activity, estradiol biosynthesis and cell proliferation in human breast cells. Mol Cell Endocrinol. 486:55–64. 2019.PubMed/NCBI View Article : Google Scholar

73 

Linehan C, Gupta S, Samali A and O'Connor L: Bisphenol A-mediated suppression of LPL gene expression inhibits triglyceride accumulation during adipogenic differentiation of human adult stem cells. PLoS One. 7(e36109)2012.PubMed/NCBI View Article : Google Scholar

74 

Ahmed F, Sarsenbayeva A, Katsogiannos P, Aguer C and Pereira MJ: The effects of bisphenol A and bisphenol S on adipokine expression and glucose metabolism in human adipose tissue. Toxicology. 445(152600)2020.PubMed/NCBI View Article : Google Scholar

75 

Masuno H, Iwanami J, Kidani T, Sakayama K and Honda K: Bisphenol a accelerates terminal differentiation of 3T3-L1 cells into adipocytes through the phosphatidylinositol 3-kinase pathway. Toxicol Sci. 84:319–327. 2005.PubMed/NCBI View Article : Google Scholar

76 

Sargis RM, Johnson DN, Choudhury RA and Brady MJ: Environmental endocrine disruptors promote adipogenesis in the 3T3-L1 cell line through glucocorticoid receptor activation. Obesity (Silver Spring). 18:1283–1288. 2010.PubMed/NCBI View Article : Google Scholar

77 

Ohlstein JF, Strong AL, McLachlan JA, Gimble JM, Burow ME and Bunnell BA: Bisphenol A enhances adipogenic differentiation of human adipose stromal/stem cells. J Mol Endocrinol. 53:345–353. 2014.PubMed/NCBI View Article : Google Scholar

78 

Ahmed S and Atlas E: Bisphenol S- and bisphenol A-induced adipogenesis of murine preadipocytes occurs through direct peroxisome proliferator-activated receptor gamma activation. Int J Obes (Lond). 40:1566–1573. 2016.PubMed/NCBI View Article : Google Scholar

79 

Salehpour A, Shidfar F, Hedayati M, Neshatbini Tehrani A, Farshad AA and Mohammadi S: Bisphenol A enhances adipogenic signaling pathways in human mesenchymal stem cells. Genes Environ. 42(13)2020.PubMed/NCBI View Article : Google Scholar

80 

Cohen IC, Cohenour ER, Harnett KG and Schuh SM: BPA, BPAF and TMBPF alter adipogenesis and fat accumulation in human mesenchymal stem cells, with implications for obesity. Int J Mol Sci. 22(5363)2021.PubMed/NCBI View Article : Google Scholar

81 

Pham DV and Park PH: Adiponectin triggers breast cancer cell death via fatty acid metabolic reprogramming. J Exp Clin Cancer Res. 41(9)2022.PubMed/NCBI View Article : Google Scholar

82 

Kim Y and Park CW: Mechanisms of adiponectin action: Implication of adiponectin receptor agonism in diabetic kidney disease. Int J Mol Sci. 20(1782)2019.PubMed/NCBI View Article : Google Scholar

83 

Hugo ER, Brandebourg TD, Woo JG, Loftus J, Alexander JW and Ben-Jonathan N: Bisphenol A at environmentally relevant doses inhibits adiponectin release from human adipose tissue explants and adipocytes. Environ Health Perspect. 116:1642–1647. 2008.PubMed/NCBI View Article : Google Scholar

84 

Lim S, Bae JH, Chun EJ, Kim H, Kim SY, Kim KM, Choi SH, Park KS, Florez JC and Jang HC: Differences in pancreatic volume, fat content, and fat density measured by multidetector-row computed tomography according to the duration of diabetes. Acta Diabetol. 51:739–748. 2014.PubMed/NCBI View Article : Google Scholar

85 

Kimata K, Sakakura T, Inaguma Y, Kato M and Nishizuka Y: Participation of two different mesenchymes in the developing mouse mammary gland: Synthesis of basement membrane components by fat pad precursor cells. J Embryol Exp Morphol. 89:243–257. 1985.PubMed/NCBI

86 

Miyawaki J, Sakayama K, Kato H, Yamamoto H and Masuno H: Perinatal and postnatal exposure to bisphenol a increases adipose tissue mass and serum cholesterol level in mice. J Atheroscler Thromb. 14:245–252. 2007.PubMed/NCBI View Article : Google Scholar

87 

Vandenberg LN, Maffini MV, Wadia PR, Sonnenschein C, Rubin BS and Soto AM: Exposure to environmentally relevant doses of the xenoestrogen bisphenol-A alters development of the fetal mouse mammary gland. Endocrinology. 148:116–127. 2007.PubMed/NCBI View Article : Google Scholar

88 

Munoz-de-Toro M, Markey C, Wadia PR, Luque EH, Rubin BS, Sonnenschein C and Soto AM: Perinatal exposure to bisphenol-A alters peripubertal mammary gland development in mice. Endocrinology. 146:4138–4147. 2005.PubMed/NCBI View Article : Google Scholar

89 

Ramskov Tetzlaff CN, Svingen T, Vinggaard AM, Rosenmai AK and Taxvig C: Bisphenols B, E, F, and S and 4-cumylphenol induce lipid accumulation in mouse adipocytes similarly to bisphenol A. Environ Toxicol. 35:543–552. 2020.PubMed/NCBI View Article : Google Scholar

90 

Frühbeck G, Méndez-Giménez L, Fernández-Formoso JA, Fernández S and Rodríguez A: Regulation of adipocyte lipolysis. Nutr Res Rev. 27:63–93. 2014.PubMed/NCBI View Article : Google Scholar

91 

Martínez M, Blanco J, Rovira J, Kumar V, Domingo JL and Schuhmacher M: Bisphenol A analogues (BPS and BPF) present a greater obesogenic capacity in 3T3-L1 cell line. Food Chem Toxicol. 140(111298)2020.PubMed/NCBI View Article : Google Scholar

92 

Drobna Z, Talarovicova A, Schrader HE, Fennell TR, Snyder RW and Rissman EF: Bisphenol F has different effects on preadipocytes differentiation and weight gain in adult mice as compared with Bisphenol A and S. Toxicology. 420:66–72. 2019.PubMed/NCBI View Article : Google Scholar

93 

Chernis N, Masschelin P, Cox AR and Hartig SM: Bisphenol AF promotes inflammation in human white adipocytes. Am J Physiol Cell Physiol. 318:C63–C72. 2020.PubMed/NCBI View Article : Google Scholar

94 

Leon-Ferre RA, Jonas SF, Salgado R, Loi S, de Jong V, Carter JM, Nielsen TO, Leung S, Riaz N, Chia S, et al: Tumor-infiltrating lymphocytes in triple-negative breast cancer. JAMA. 331:1135–1144. 2024.PubMed/NCBI View Article : Google Scholar

95 

Valenza C, Taurelli Salimbeni B, Santoro C, Trapani D, Antonarelli G and Curigliano G: Tumor infiltrating lymphocytes across breast cancer subtypes: Current Issues for biomarker assessment. Cancers (Basel). 15(767)2023.PubMed/NCBI View Article : Google Scholar

96 

Qiu SQ, Waaijer SJH, Zwager MC, de Vries EGE, van der Vegt B and Schröder CP: Tumor-associated macrophages in breast cancer: Innocent bystander or important player? Cancer Treat Rev. 70:178–189. 2018.PubMed/NCBI View Article : Google Scholar

97 

No authors listed. Estrogen receptor-positive breast cancer subtypes show differential macrophage functions. Nat Cancer. 4:450–451. 2023.PubMed/NCBI View Article : Google Scholar

98 

Tran HTT, Herz C and Lamy E: Long-term exposure to ‘low-dose’ bisphenol A decreases mitochondrial DNA copy number, and accelerates telomere shortening in human CD8 + T cells. Sci Rep. 10(15786)2020.PubMed/NCBI View Article : Google Scholar

99 

Kim H, Kim HS and Moon WK: Comparison of transcriptome expression alterations by chronic exposure to low-dose bisphenol A in different subtypes of breast cancer cells. Toxicol Appl Pharmacol. 385(114814)2019.PubMed/NCBI View Article : Google Scholar

100 

Bouzidi L, Triki H, Charfi S, Kridis WB, Derbel M, Ayadi L, Sellami-Boudawara T and Cherif B: Prognostic value of natural killer cells besides tumor-infiltrating lymphocytes in breast cancer tissues. Clin Breast Cancer. 21:e738–e747. 2021.PubMed/NCBI View Article : Google Scholar

101 

Kim IS, Gao Y, Welte T, Wang H, Liu J, Janghorban M, Sheng K, Niu Y, Goldstein A, Zhao N, et al: Immuno-subtyping of breast cancer reveals distinct myeloid cell profiles and immunotherapy resistance mechanisms. Nat Cell Biol. 21:1113–1126. 2019.PubMed/NCBI View Article : Google Scholar

102 

Balistrieri A, Hobohm L, Srivastava T, Meier A and Corriden R: Alterations in human neutrophil function caused by bisphenol A. Am J Physiol Cell Physiol. 315:C636–C642. 2018.PubMed/NCBI View Article : Google Scholar

103 

Chanmee T, Ontong P, Konno K and Itano N: Tumor-associated macrophages as major players in the tumor microenvironment. Cancers (Basel). 6:1670–1690. 2014.PubMed/NCBI View Article : Google Scholar

104 

Palacios-Arreola MI, Nava-Castro KE, Río-Araiza VHD, Pérez-Sánchez NY and Morales-Montor J: A single neonatal administration of Bisphenol A induces higher tumour weight associated to changes in tumour microenvironment in the adulthood. Sci Rep. 7(10573)2017.PubMed/NCBI View Article : Google Scholar

105 

Kim H, Kim HS, Piao YJ and Moon WK: Bisphenol A promotes the invasive and metastatic potential of ductal carcinoma in situ and protumorigenic polarization of macrophages. Toxicol Sci. 170:283–295. 2019.PubMed/NCBI View Article : Google Scholar

106 

Pahović PŠ, Iulini M, Maddalon A, Galbiati V, Buoso E, Dolenc MS and Corsini E: In vitro effects of bisphenol analogs on immune cells activation and Th differentiation. Endocr Metab Immune Disord Drug Targets. 23:1750–1761. 2023.PubMed/NCBI View Article : Google Scholar

107 

Oshi M, Gandhi S, Yan L, Tokumaru Y, Wu R, Yamada A, Matsuyama R, Endo I and Takabe K: Abundance of reactive oxygen species (ROS) is associated with tumor aggressiveness, immune response, and worse survival in breast cancer. Breast Cancer Res Treat. 194:231–241. 2022.PubMed/NCBI View Article : Google Scholar

108 

Qiu W, Shao H, Lei P, Zheng C, Qiu C, Yang M and Zheng Y: Immunotoxicity of bisphenol S and F are similar to that of bisphenol A during zebrafish early development. Chemosphere. 194:1–8. 2018.PubMed/NCBI View Article : Google Scholar

109 

Dontu G and Ince TA: Of mice and women: A comparative tissue biology perspective of breast stem cells and differentiation. J Mammary Gland Biol Neoplasia. 20:51–62. 2015.PubMed/NCBI View Article : Google Scholar

110 

Cardiff RD and Wellings SR: The comparative pathology of human and mouse mammary glands. J Mammary Gland Biol Neoplasia. 4:105–122. 1999.PubMed/NCBI View Article : Google Scholar

111 

Roberts GC, Morris PG, Moss MA, Maltby SL, Palmer CA, Nash CE, Smart E, Holliday DL and Speirs V: An evaluation of matrix-containing and humanised matrix-free 3-dimensional cell culture systems for studying breast cancer. PLoS One. 11(e0157004)2016.PubMed/NCBI View Article : Google Scholar

112 

Russo J and Russo IH: Genotoxicity of steroidal estrogens. Trends Endocrinol Metab. 15:211–214. 2004.PubMed/NCBI View Article : Google Scholar

113 

Sachs N and Clevers H: Organoid cultures for the analysis of cancer phenotypes. Curr Opin Genet Dev. 24:68–73. 2014.PubMed/NCBI View Article : Google Scholar

114 

Reddington R, Galer M, Hagedorn A, Liu P, Barrack S, Husain E, Sharma R, Speirs V and Masannat Y: Incidence of male breast cancer in Scotland over a twenty-five-year period (1992-2017). Eur J Surg Oncol. 46:1546–1550. 2020.PubMed/NCBI View Article : Google Scholar

115 

Roberts S, Peyman S and Speirs V: Current and emerging 3D models to study breast cancer. Adv Exp Med Biol. 1152:413–427. 2019.PubMed/NCBI View Article : Google Scholar

116 

Weigelt B, Ghajar CM and Bissell MJ: The need for complex 3D culture models to unravel novel pathways and identify accurate biomarkers in breast cancer. Adv Drug Deliv Rev. 69-70:42–51. 2014.PubMed/NCBI View Article : Google Scholar

117 

Tang M, Jiang S, Huang X, Ji C, Gu Y, Qi Y, Xiang Y, Yao E, Zhang N, Berman E, et al: Integration of 3D bioprinting and multi-algorithm machine learning identified glioma susceptibilities and microenvironment characteristics. Cell Discov. 10(39)2024.PubMed/NCBI View Article : Google Scholar

118 

Huh D, Hamilton GA and Ingber DE: From 3D cell culture to organs-on-chips. Trends Cell Biol. 21:745–754. 2011.PubMed/NCBI View Article : Google Scholar

119 

Ståhl PL, Salmén F, Vickovic S, Lundmark A, Navarro JF, Magnusson J, Giacomello S, Asp M, Westholm JO, Huss M, et al: Visualization and analysis of gene expression in tissue sections by spatial transcriptomics. Science. 353:78–82. 2016.PubMed/NCBI View Article : Google Scholar

120 

Reed AD, Pensa S, Steif A, Stenning J, Kunz DJ, Porter LJ, Hua K, He P, Twigger AJ, Siu AJQ, et al: A single-cell atlas enables mapping of homeostatic cellular shifts in the adult human breast. Nat Genet. 56:652–662. 2024.PubMed/NCBI View Article : Google Scholar

121 

EFSA Panel on Food Contact Materials, Enzymes Flavourings and Processing Aids (CEF). Scientific opinion on the risks to public health related to the presence of bisphenol A (BPA) in foodstuffs. EFSA J. 13(3978)2015.

122 

Dueñas-Moreno J, Mora A, Kumar M, Meng XZ and Mahlknecht J: Worldwide risk assessment of phthalates and bisphenol A in humans: The need for updating guidelines. Environ Int. 181(108294)2023.PubMed/NCBI View Article : Google Scholar

Related Articles

Journal Cover

July-August 2025
Volume 7 Issue 4

Print ISSN: 2632-2900
Online ISSN:2632-2919

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Tripathi S, Kusserow K, Palmer K, Dodson A, Mcclurg DP and Speirs V: Effects of bisphenol A and its chemical analogues on the microenvironment and female breast cancer development (Review). World Acad Sci J 7: 66, 2025.
APA
Tripathi, S., Kusserow, K., Palmer, K., Dodson, A., Mcclurg, D.P., & Speirs, V. (2025). Effects of bisphenol A and its chemical analogues on the microenvironment and female breast cancer development (Review). World Academy of Sciences Journal, 7, 66. https://doi.org/10.3892/wasj.2025.354
MLA
Tripathi, S., Kusserow, K., Palmer, K., Dodson, A., Mcclurg, D. P., Speirs, V."Effects of bisphenol A and its chemical analogues on the microenvironment and female breast cancer development (Review)". World Academy of Sciences Journal 7.4 (2025): 66.
Chicago
Tripathi, S., Kusserow, K., Palmer, K., Dodson, A., Mcclurg, D. P., Speirs, V."Effects of bisphenol A and its chemical analogues on the microenvironment and female breast cancer development (Review)". World Academy of Sciences Journal 7, no. 4 (2025): 66. https://doi.org/10.3892/wasj.2025.354