Next Article in Journal
Neoadjuvant Approaches to Non-Melanoma Skin Cancer
Previous Article in Journal
Robotic Distal Pancreatectomy Yields Superior Outcomes Compared to Laparoscopic Technique: A Single Surgeon Experience of 123 Consecutive Cases
Previous Article in Special Issue
The Possible Role of Anti- and Protumor-Infiltrating Lymphocytes in Pathologic Complete Response in Early Breast Cancer Patients Treated with Neoadjuvant Systemic Therapy
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

The Role of the Tumor Microenvironment in Triple-Positive Breast Cancer Progression and Therapeutic Resistance

1
Department of Breast Surgery, Qilu Hospital (Qingdao), Cheeloo College of Medicine, Shandong University, Qingdao 266035, China
2
Oncology Laboratory, Qilu Hospital (Qingdao), Cheeloo College of Medicine, Shandong University, Qingdao 266035, China
*
Author to whom correspondence should be addressed.
Cancers 2023, 15(22), 5493; https://doi.org/10.3390/cancers15225493
Submission received: 15 September 2023 / Revised: 26 October 2023 / Accepted: 18 November 2023 / Published: 20 November 2023
(This article belongs to the Special Issue Tumor Microenvironment of Breast Cancer)

Abstract

:

Simple Summary

This is an important point of view that the development of breast carcinogenesis is not only related to the intrinsic characteristics of the cancer cells, but that it is also strongly dependent on surrounding microenvironmental factors. It is important to note that triple-positive breast cancer accounts for about 10–15% of all breast cancers, and research on the tumor microenvironment for this subtype is still limited. In this review, authors explored the relationship between critical cellular components and factors in the triple-positive breast cancer microenvironment and the inception, advancement, and therapeutic resistance of breast cancer to provide perspectives on the latest research on triple-positive breast cancer. The authors sufficiently described the state of the art and reported the fundamental concepts, allowing an easy comprehension of the intricate dynamics within tumor microenvironment in triple-positive breast cancer.

Abstract

Breast cancer (BRCA) is a highly heterogeneous systemic disease. It is ranked first globally in the incidence of new cancer cases and has emerged as the primary cause of cancer-related death among females. Among the distinct subtypes of BRCA, triple-positive breast cancer (TPBC) has been associated with increased metastasis and invasiveness, exhibiting greater resistance to endocrine therapy involving trastuzumab. It is now understood that invasion, metastasis, and treatment resistance associated with BRCA progression are not exclusively due to breast tumor cells but are from the intricate interplay between BRCA and its tumor microenvironment (TME). Accordingly, understanding the pathogenesis and evolution of the TPBC microenvironment demands a comprehensive approach. Moreover, addressing BRCA treatment necessitates a holistic consideration of the TME, bearing significant implications for identifying novel targets for anticancer interventions. This review expounds on the relationship between critical cellular components and factors in the TPBC microenvironment and the inception, advancement, and therapeutic resistance of breast cancer to provide perspectives on the latest research on TPBC.

1. Introduction

In recent years, breast cancer (BRCA) has become the predominant cause of mortality among women in society, surpassing the global prevalence and death toll of lung cancer [1,2]. It is essential to acknowledge that BRCA is not a single disease but rather a malignancy exhibiting significant heterogeneity. BRCA can be classified into four distinct subtypes based on the immunohistochemical identification of the estrogen receptor (ER), progesterone receptor (PR), human epidermal growth factor receptor 2 (HER2), and Ki-67 index: Luminal A, Luminal B, triple-negative, and HER2 overexpression type [3]. Substantial progress has been made in the clinical diagnosis and treatment of BRCA through molecular categorization. However, due to individual patient heterogeneity and the substantial clinical diversity of BRCA, the prognostic value of BRCA has fallen short of expectations [4,5,6,7]. Research has revealed that triple-positive breast cancer (TPBC), representing a unique hormone receptor-positive subtype within BRCA, exhibits positivity for ER, PR, and HER2 [8], with malignancy levels on par with or even exceeding triple-negative breast cancer (TNBC). There is an increasing consensus suggesting that TPBC is associated with higher rates of recurrence, particularly involving internal organs and bone tissue, thus accounting for the poor prognoses [9,10,11]. While research on BRCA has grown over the years, studies focused on TPBC remain scarce, with even fewer reports from China. Therefore, a deeper exploration and analysis of TPBC’s attributes could offer a scientific foundation for clinical treatments.
TPBC accounts for approximately 10% to 15% of all breast cancers [12,13]. Its clinicopathological characteristics are predominantly characterized by higher tumor grade, larger volume, inferior prognosis, increased invasiveness, significant vasculaturization or nerve invasion, and a higher likelihood of axillary lymph node and distant metastases, with bone metastases being mostly prevalent [14,15]. TPBC is defined by the positive expression of ER, PR, and HER2. ER and PR, intracellular receptors binding to estrogen and progesterone, respectively, regulate factor expression and cell proliferation. HER2, a cell surface receptor, stimulates cell proliferation via downstream signaling pathways. Elevated expression of these molecules suggests TPBC’s increased dependence on these receptors and hormones, often prompting anti-HER2-targeted molecular therapies. Clinical treatment typically encompasses comprehensive approaches such as surgical resection, targeted therapy combined with chemotherapy, endocrine therapy, and radiotherapy. Over the years, studies have uncovered that TPBC patients exhibit limited responses to chemotherapy and hormone therapy, possibly due to the cross-interaction between the HR and HER2 signaling pathways. When one pathway is blocked by chemotherapy or hormone therapy, the other is subsequently upregulated [16]. Mark Pegram et al. pointed out that blocking multiple HER family receptors may be more effective than blocking the HER 2 kinase alone, and using pan-HER TKI neritinib as part of the combination therapy may re-sensitize the ER pathway to endocrine therapy [17]. Consequently, treating TPBC warrants a comprehensive consideration of these dual pathways. Precisely evaluating TPBC prognosis and effectively monitoring and predicting tumor recurrence stand as pivotal topics in the era of precision medicine. Assessing patient prognosis based on clinical tumor characteristics offers valuable guidance for clinicians overseeing recurrence monitoring.
In the realm of malignant tumors, invasion and metastasis represent two crucial facets, with metastasis holding greater influence over clinical decision-making, embodying the predominant cause of death in cancer patients [18]. The metastatic course of cancer cells encompasses dissemination from the primary tumor, traversing epithelial-to-mesenchymal transition (EMT), followed by colonization at secondary sites through mesenchymal-to-epithelial transition (MET).
The tumor microenvironment (TME) refers to the evolving microecosystem accompanying normal cells and supporting them during their transformation from normal to malignant states. This localized microenvironment evolves during the tumor’s developmental course [19,20]. The TME encapsulates cellular and non-cellular constituents alongside latent malignant tumor cells [21]. It is widely thought that a bidirectional signal exchange and mutual influence is present, intricately linked to tumorigenesis, metastasis, and treatment resistance [22,23]. Cell components span mesenchymal and hematopoietic cells, the former encompassing fibroblasts, myofibroblasts, adipocytes, endothelial cells, and mesenchymal stem cells (MSCs). Hematopoietic cells involve lymphoid cells (T cells, B cells, dendritic cells, mast cells, NK cells) and bone marrow cells (tumor-associated macrophages, neutrophils, myeloid-derived suppressor cells (MDSCs)). Non-cellular components encompass tangible and intangible elements. Tangible components largely constitute critical extracellular matrix (ECM) constituents such as collagen, fibronectin, laminin, aminoglycans, and proteoglycans. Intangible components predominantly encompass soluble agents like growth factors, cytokines, and chemokines. Additionally, hormones, proteases, and exosomes, as well as features like hypoxia and metabolic imbalances, are present. The tumor microenvironment additionally encompasses pivotal metabolic conditions (pH, PO2, glucose, glutamine, lactic acid) and chemical factors (e.g., NO) [7]. Interactions among these TME components collectively promote tumor initiation, development, invasion, and metastasis, and influence responses to treatment. Following the malignant shift of mammary epithelial cells, the microenvironment composition undergoes significant change, restructuring the ECM and releasing constraints on mammary epithelial cell proliferation, consequently leading to uncontrolled tumor cell proliferation and invasion [24]. Drug resistance exhibited by TPBC toward endocrine therapy results from multifaceted mechanisms and signaling pathway coordination, which are not exclusively related to tumor cell transformations, with many TME components playing pivotal roles [25]. Although the past few years have witnessed extensive research on the TME of TNBC, research on TPBC’s TME remains limited. This paper thus reviews the roles of several crucial cellular and non-cellular constituents in TPBC’s onset, progression, and drug resistance pathways via an exploration of related cell line TMEs.

2. Tumor Mesenchymal Cell Components in TPBC

2.1. Cancer-Associated Fibroblasts (CAFs)

Within the breast cancer TME, CAFs dominate as the most abundant cellular constituents of the tumor stroma. They can originate from normal stromal fibroblasts (referred to as normal fibroblasts, NFs) [26]. Additionally, CAFs can emerge through epithelial-mesenchymal transition from bone marrow-derived mesenchymal stem cells, endothelial cells, epithelial cells, or transdifferentiated cells [27,28]. The widely utilized marker for identifying CAFs is the α-smooth muscle actin (α-SMA) [29]. CAFs secrete growth factors, cytokines, proteases, and more, which not only foster the initiation, growth, angiogenesis, invasion, and metastasis of breast cancer but also serve as biomarkers in clinical practice, guiding breast cancer diagnosis, therapy, and prognosis assessments [19,30,31,32,33].
In triple-positive breast cancer, Shekhar et al. revealed through a three-dimensional cell–cell interaction model that normal mesenchymal fibroblasts (NFs) inhibited estrogen-induced tumor cell growth, whereas CAFs produced abundant estrogens, inducing the malignant transformation of the normal breast epithelial cell line MCF10A and the precancerous breast epithelial cell line EIII8 [34]. Notably, cell surface antigens like CD44, CD24, and ESA have been effectively harnessed to isolate cancer stem cell (CSC)-like populations from breast cancer cell lines and primary tissues. CD44hi and CD44lo populations have been identified across various basic breast cancer systems, while luminal breast cancer systems exclusively consisted of CD44lo populations. ZEB1 played a crucial role in steering the transition from CD44lo to CD44hi. Importantly, CAFs’ secretion of transforming growth factor-beta (TGFβ) enhanced the conversion rate of CD44lo to CD44hi in the basal breast cancer CD44lo cell subset. This effect was contingent on the induction of ZEB1 expression, indicating the considerable role of CAFs in triple-negative breast cancer initiation, although the involvement of CAFs in TPBC initiation has received limited attention [35].
It is well-established that CAFs can release various factors that stimulate neighboring malignant cells, thereby fostering tumor progression [36]. Growth factors secreted by CAFs in the breast cancer microenvironment encompass basic fibroblast growth factor 2 (FGF2) [37], platelet-derived growth factor (PDGF) [38], insulin growth factor 1 (IGF1), and large amounts of C-X-C motif chemokine ligand 12 (CXCL12), also known as stromal cell-derived factor (SDF-1). Interestingly, heightened CXCL12 expression levels could predict diminished overall survival (OS) and relapse-free survival (RFS) in patients across all breast cancer subtypes. CXCL12 is pivotal in the initial stages of tumorigenesis, and studies have corroborated that fibroblast-derived CXCL12 consumption mitigates tumor cell proliferation while recruiting endothelial progenitor cells to the TME, thus expanding tumor vessels [39]. Furthermore, CAFs can partake in mediating inflammation-driven tumor-promoting factors by producing pro-inflammatory cytokines [40,41]. Recent findings have indicated the expression of the third estrogen receptor, the G protein-coupled estrogen receptor (GPER), in breast CAFs. In estrogen-sensitive breast cancer, GPER was found to mediate a distinct gene signature associated with cell growth, migration, and angiogenesis [42,43,44]. E2 and G-1 induced IL1β expression in CAFs through GPER activation. This process also triggered the EGFR/ERK/PKC signaling cascade, culminating in the establishment of a feedforward loop. This loop further interconnected IL1β with the expression of IL1R1 in luminal cancer cells MCF7 and SKBR3, fostering functional crosstalk between the tumor microenvironment and intracavitary breast cancer cells, consequently promoting tumor metastasis [45]. Notably, it has been shown that CAFs secrete exosome miR-500a-5p, which could bind to ubiquitin-specific peptidase 28 (USP28), thereby facilitating progression and metastasis in both the basal breast cancer cell line MDA-MB-231 and the luminal cell line MCF7 [46].
Both endocrine therapy and targeted therapy are indicated for TPBC. However, the involvement of CAFs in the emergence of resistance to anticancer drugs remains a complex area, particularly regarding the specifics of CAF-mediated tamoxifen resistance in TPBC. Investigations have unveiled that interleukin-6 (IL-6) secreted by CAFs contributes to the downregulation of ERα expression and activates downstream signaling pathways, notably JAK/STAT3 and PI3K/AKT. This orchestration results in resistance to endocrine therapy, especially in luminal cell carcinoma cells [47]. The emergence of tamoxifen (TAM) resistance stands as a significant hurdle in endocrine therapy for BRCA. The G-protein-coupled estrogen receptor (GPER) has been postulated to be a catalyst for tamoxifen resistance. Current evidence suggests that TAM, alongside E2 and GPER agonist G1, stimulates proliferation, cell-cycle progression, and E2 production through the GPER/EGFR/ERK axis within breast CAFs. These findings introduce novel perspectives into the GPER-mediated, CAF-dependent mechanism underlying TAM resistance in breast cancer [48]. Furthermore, GPER was identified as influencing β1-integrin, a target gene, wherein CAF’s release fibronectin (FN), which engages with β1 integrin, thereby activating MAPK/ERK1/2 and PI3K/AKT pathways. The resultant cascade contributes to endocrine therapy drug resistance in breast cancer [49]. Yuan et al. further found that GPER/EGFR/ERK signaling boosts β1-integrin expression and triggers downstream kinases in MCF-7R cells, fostering CAF-induced cell migration and epithelial–mesenchymal transition (EMT). GPER’s potential to promote tamoxifen resistance is linked with its interaction within the tumor microenvironment in a β1-integrin-dependent manner [50].
It is widely thought that anti-HER2-targeted therapy is pivotal in the context of triple-positive breast cancer treatment. However, the mechanism behind how cancer-associated fibroblasts induce trastuzumab resistance in HER2+ breast cancer remains elusive. Current understanding proposes that CAFs in HER2+ breast cancer confer resistance to Herceptin by orchestrating the loss of PTEN or activating the IL-6/STAT-3/NF-κB signaling pathway [51]. Furthermore, CAFs can trigger trastuzumab resistance by expanding tumor stem cells and activating multiple pathways, such as NF-κB, JAK/STAT3, and PI3K/AKT. In this light, a combined approach featuring anti-IL-6 antibodies or multi-pathway inhibitors in conjunction with trastuzumab could emerge as a novel strategy to counteract trastuzumab resistance [52].

2.2. Tumor-Associated Macrophages (TAMs)

Immune cells within the TME wield a dual influence, inhibiting tumors while also participating in processes like tumor angiogenesis, immunosuppression, and cell growth through cytokine secretion. This multifaceted interplay often fosters tumor metastasis and bolsters tolerance to drug treatments [53]. Among the immune cell populations, macrophages, mast cells, and lymphocytes are the primary inflammatory cells involved in the TME.
TAMs constitute one of the most prevalent immune cell subsets within the TME and significantly shape various aspects of solid malignancies. They catalyze tumorigenesis, promote neovascularization, remodel the TME into an immunosuppressive state, confer resistance to cancer chemotherapy, and increase the risk of recurrence and metastasis [54]. TAMs can be categorized into two functionally disparate subtypes: classically activated M1 TAMs, which possess anti-tumor traits, and activated M2 TAMs, which potentiate tumor cell formation and metastasis [55]. An increased density of M2 phenotype TAMs is closely associated with unfavorable clinical outcomes [56]. Typical markers of M1 macrophages encompass CD64, CD68, CD80, CD86, IDO, SOCS1, and CXCL10 [57], while M2 macrophages are typified by markers like MRC1, TGM2, CD23, CD163, CD204, CD206, and CCL22 [58,59,60]. In a meta-analysis involving 4541 breast cancer patients, Matikas et al. uncovered that elevated TAM density within tumor tissue correlated with poorer prognosis and hormone-receptor-negative (HR-) status [61]. In a study by Pelekanou et al. involving 113 HR+ and 37 ER- patients who underwent tumor biopsies at baseline and two cycles of neoadjuvant chemotherapy (NAC), gene mapping analysis and quantification of tumor-infiltrating lymphocytes (TILs), FOXP3+ T cells, and CD163+ TAMs, were conducted. The study unveiled that heightened CD163 expression correlates with enhanced OS in ER+ patients [62]. Additionally, Mansfield et al. were among the first to propose that reduced CD163+ TAMs may play a role in lymph node metastasis in breast cancer, with CD163 expression serving as a predictor of lymph node status [63]. Additionally, studies by FAN and He indicated that in the luminal breast cancer TME, TAM characteristics include a lower quantity, suppression, rounded morphology, and slight elevation in M2 markers [64]. Discrepancies in the relationship between TAM markers and pathologic complete response (pCR) expression and prognostic indicators could arise due to inconsistent immunohistochemical interpretation of TAMs or be associated with the molecular subtype of breast cancer. Accordingly, the predictive value of TAMs for treatment response across different BRCA subtypes, especially in TPBC, warrants further investigation [65].
While the relationship between TAMs and TNBC has become a research hotspot, studies examining TAMs and TPBC remain scarce. Interestingly, it has been reported macrophages can drive the development of triple-negative tumors by regulating succinate dehydrogenase (SDH) expression in tumor cells through mechanisms linked to the TGF-β signal transduction [66]. The overexpression of MCT-1 in triple-negative breast cancer activates MCT-1/miR-34a/IL-6/IL-6R signaling, disrupting the normal structure of breast cancer epithelial glands and inducing EMT, thus promoting the polarization of TAMs toward the M2 subtype [67]. Additionally, tumor heterogeneity significantly influences the activation of tumor-associated macrophages. Maija et al. employed full transcriptomic sequencing to study human monocytes co-cultured with ER+ or TNBC cell lines. They investigated the biological responses associated with differential gene activation in monocytes and cancer cells, revealing divergent macrophage phenotypes induced by ER+ (T47d) and TNBC (MDA-MB-231) cancer cells, exhibiting distinct functions, cytokine and chemokine secretion patterns, and morphology. Simultaneously, the presence of macrophages exerted considerable effects on ER+ and TNBC cell lines. ER+ cells demonstrated enhanced acute inflammatory responses, IL-17 signaling, and antigen-presenting pathways, while thioredoxin and vitamin D3 receptor pathways were downregulated in macrophages [68]. Derived from TAMs, the CXC motif chemokine ligand 1 (CXCL1) has been demonstrated to drive the differentiation of naive CD4+ T cells into regulatory T cells through transcriptional activation of the NF-κB/FOXP3 signaling pathway, contributing to the development of regulatory T cells (Treg) [69]. Similarly, mediating CXC motif chemokine ligand 8 (CXCL8) infiltration fosters immune escape and lung metastasis in BRCA [70].
In recent years, TAMs have been linked to drug resistance in specific solid tumor types, including BRCA. Nonetheless, investigations into the interplay between TAMs and therapeutic resistance in TPBC are in their preliminary stages. TAMs are widely believed to induce drug resistance via the IL-10/STAT3/bcl-2 signaling pathway [71]. Additionally, the secretion of chemokine ligand 2 (CCL2) by TAMs triggers the activation of the PI3K/AKT/mTOR signaling cascade in tumor cells, culminating in tamoxifen resistance within breast cancer [72]. TAMs can also activate the NF-κB/STAT3/ERK pathway within tumor cells through the release of pro-inflammatory cytokines, such as TNF-α and IL-6. This signaling cascade phosphorylates and activates ERα, leading to endocrine therapy resistance in BRCA [73]. M2-type TAMs have demonstrated the ability to activate breast cancer cells via the EGFR/PI3K/Akt pathway, resulting in the upregulation of SGLT1, a phenomenon that fosters tamoxifen resistance in cancer cells [74]. Notably, M2 macrophages play a pivotal role in instigating drug resistance in BRCA, with exosomes also assuming a crucial role in the induction process of M2 macrophages. An exosome-mediated release of miR-222 by adriamycin-resistant MCF-7 cells facilitates M2 polarization through the PTEN/Akt pathway, thereby accelerating tumor progression [75].

2.3. Cancer-Associated Adipocytes (CAAs)

Adipose tissue is an important component of the mammary gland, accounting for 56% [76] and 35% [77] of breast volume during non-lactating and lactating periods, respectively. Bioactive adipokines emanating from adipocytes have been identified as major contributors to tumorigenesis and cancer progression [78]. CAAs may promote proliferation, migration, and invasion of BRCA cells [79], drive metabolic alterations within breast cancer cells, induce heightened fatty acid storage and oxidation [80], and mediate tumor resistance to chemotherapy and radiation therapy [81]. CAAs may even be associated with an increased incidence of local events after autologous adipose tissue filling for breast cancer [82]. Additionally, CAAs are widely thought to play an important role in the development of TPBC.
Pathologically, CAAs are associated with the formation of crown-like structures (CLS) and enhanced lipolysis. These CLS—microscopic conglomerates of expiring adipocytes encircled by macrophages—are believed to reflect sites with heightened conversion of androgens to estrogens [83]. CLS constitute a histological hallmark of breast adipose tissue, marked by organized assemblies of CD68-positive macrophages enveloping deceased or perishing adipocytes. It is highly conceivable that these structures indicate chronic inflammation. Studies involving animal models and female breast tissue have suggested that CLS’ presence is correlated with obesity and pro-inflammatory and pro-carcinogenic processes, marked by augmented aromatase activity and signaling via an ER-mediated pathway [84,85,86]. It has also been reported that CLS adversely affect ER-positive breast cancer prognosis. In mice, the CLS-associated pro-inflammatory and carcinogenic processes can be ameliorated through weight loss or chemopreventive drugs [87,88]. Mullooly et al. [89] conducted a study that explored the relationship between CLS and sex steroid hormones in breast adipose tissue and serum within 83 postmenopausal breast cancer patients. They discovered that the presence of CLS in the breast adipose tissue of these patients was linked to an increased estrogen-to-androgen ratio in both breast fat and serum. Cha YJ et al. [90] compared breast adipose tissue among normal individuals, breast cancer patients, and adipose tissue adjacent to breast cancer. They observed the densest distribution of CLS in adipose tissue adjacent to cancer (p < 0.001) was correlated with age, ER expression, and HER2 expression.
In contrast to normal adipose tissue, CAAs exhibit diminished differentiation and lose their typical functions, including energy storage [91]. They also secrete a plethora of soluble cytokines, referred to as adipokines/adipocytokines [92], with ongoing research focusing on factors like leptin, adiponectin, visfatin, autotaxin, hepatocyte growth factor (HGF), tumor necrosis factor-alpha (TNF-α), and heparin-binding epidermal growth factor (HB-EGF) [93].
Regarding TPBC, CAAs could potentially activate several signaling pathways, including ER signaling. Leptin, when lacking a natural ligand, stimulates ER signaling, ultimately propelling BRCA cell proliferation [94,95]. Furthermore, leptin enhances the cellular proliferation of BRCA cells through HER2 trans-activation via EGFR and JAK2 activation [96,97]. Intriguingly, leptin’s influence even extends to increased cellular proliferation facilitated by the JAK/STAT3 [98] and PI3K/AKT pathways [99]. Notably, within luminal cell lines MCF-7 and SK-BR-3, leptin can induce EMT in BRCA cells by upregulating PKM2 expression and activating the PI3K/AKT signaling pathway. Counteracting the effects of leptin through an anti-leptin receptor antibody (anti-OBR) and PI3K/AKT signaling inhibitor LY294002 significantly suppresses leptin-induced PKM2 expression, as well as the expression of EMT-related markers [100]. Partial inhibition of leptin-induced migration, invasion, and EMT-related marker expression can be achieved through siRNA targeting PKM2 [100].
Adiponectin, a hormone predominantly synthesized in adipose tissue, is abundantly present in human plasma and has been recognized as an insulin-sensitizing adipocytokine [101,102]. Gunter et al. [103] conducted a retrospective analysis involving 875 postmenopausal breast cancer patients and 839 volunteers and found significantly reduced levels of adiponectin in the blood of breast cancer patients, concurrent with a decrease in adiponectin levels in CAAs [104,105]. AdipoR1 and AdipoR2, two transmembrane adiponectin receptor subtypes, initiate adiponectin’s biological effects by inducing the activation of protein kinases, primarily AMPK and MAPK [106]. In luminal cells, estrogen—the primary mitogen of breast cancer cells—has been reported to induce G1-S cell cycle progression by upregulating the expression of c-myc and cyclin D1 genes [107]. Dieudonne MN et al. identified functional AdipoR1 and AdipoR2 receptors in MCF-7 cells and demonstrated that adiponectin suppressed the expression of c-myc and cyclin D1 mRNA, directly impeding the cell cycle progression in the G1-S phase. Adiponectin exerted anti-proliferation and pro-apoptotic effects through the activation of AMP kinase and MAP kinase pathways, including p38 MAPK, ERK, and JNK [108]. Concurrently, adiponectin upregulated the expression of the tumor suppressor gene LKB1 in MCF7 cells. LKB1 was found to be crucial for adiponectin-mediated regulation of the AMPK-S6K axis, which mitigated adhesion, migration, and invasion of breast cancer cells [109].
Current evidence suggests that within crown-like structures, dying CAAs liberate free fatty acids, binding to toll-like receptor 4 on macrophages and adjacent adipocytes. This binding triggers the activation of nuclear transcription factor κB, resulting in the upregulation of inflammatory factors, including IL-8. Extracted from invasive breast cancer specimens, CAAs generated significantly more IL-8 compared to neighboring normal fat cells [110]. Elevated IL-8 levels correlated with earlier distant metastasis and poorer breast cancer prognosis [111]. While some scholars advocate that elevated IL-8 is mainly detected in hormone receptor-negative and HER2-positive breast cancer cases [112,113], a complex interplay exists between ER expression and IL8 in BRCA cells [114,115,116]. The underlying mechanism suggests IL-8’s involvement in the activation of intracellular BRCA pathways, such as the phosphatidylinositol 3 kinase/mitogen-activated protein kinase pathway, phospholipase C/protein kinase C pathway, and non-receptor protein tyrosine kinases, including Src family members and adhesion spot kinases [117]. Additionally, cancer cell-secreted IL-8 can convert normal fat cells into CAAs, heightening CAAs’ capacity to stimulate angiogenesis and promote tumor growth [110].
The TME assumes a pivotal role in tumor drug resistance, with CAAs playing a significant role in modulating tumor response to therapy. In ER+ breast cancer cells, CAAs secrete fibroblast growth factor 1 (FGF1), which phosphorylates the fibroblast growth factor receptor (FGFR). This process leads to resistance to endocrine therapy in BRCA. However, inhibiting the FGFR could potentially reverse this resistant phenotype [118]. In addition, leptin holds the potential to influence anti-cancer therapy by promoting the proliferation of cancer stem cells [119].

2.4. Mesenchymal Stem Cells (MSCs)

MSCs are pluripotent spindle-shaped cells obtained and cultured from diverse tissues like bone marrow, fat, umbilical cord, and placenta [120]. They exhibit surface markers CD73, CD90, and CD105 and lack the expression of CD34, CD45, CD14 or CD11b, CD19 or CD79α, and human leukocyte antigen-DR (HLA-DR). MSCs harbor trilineage differentiation potential (osteogenesis, lipogenesis, and chondrogenesis), enabling them to differentiate into bone, cartilage, muscle, fat, and nerve cells in vitro [121]. The comprehensive mechanism and impact of MSCs from varied sources (such as bone marrow, umbilical cord, fat, and placenta) on triple-positive breast cancer have yet to be systematically investigated. Ongoing research is focused on the role of MSCs from different origins in luminal cell lines and on preliminarily assessing their pro-tumor or anti-tumor effects in TPBC, along with plausible mechanisms underlying treatment resistance.
MSCs can secrete chemokine ligand 5 (CCL5) and transforming growth factor-beta (TGF-β), fostering EMT and preserving stem cell properties within tumor cells, thereby driving metastasis [122]. Breast cancer stem cells (BCSCs), with attributes resembling stem cells, including self-renewal and oncogenic maintenance, drive the proliferation, metastasis, and chemotherapy resistance of breast cancer cells. MSCs participate in the regulation of BCSCs by secreting interleukin-6 (IL-6) and cytokine CXCL7, thereby promoting BCSC self-renewal and cancer cell proliferation [123]. MSC-derived exosomes, rich in mRNA, miRNA, and soluble factors (including cytokines and chemokines), can infiltrate tumor cells, exerting biological effects [124]. These exosomes have been found to inhibit the mTOR/HIF-1α/VEGF signaling axis, thereby suppressing the expression of relevant factors, inhibiting endothelial cell migration and proliferation, and even inducing dormancy in BRCA cells, restraining the migration and proliferation of breast cancer cells [125,126]. In vitro studies focusing on adipose-derived stem cells (ASCs) have demonstrated that ASCs’ extracellular signaling exerts an inhibitory influence on BRCA progression. Under direct co-culture conditions, ASCs notably mitigated the proliferation of SKBR3 cells, an effect dependent on the SDF1α/CXCR4 signaling axis. Concurrently, ASCs induced alterations in tumor cell morphology, prompting epithelial-to-mesenchymal transition, increased the formation of breast spheres, facilitated cell fusion, and augmented the migratory capacity of SKBR3 cells. These outcomes could potentially be associated with the stimulation of cytokines and chemokines secreted by MSCs in BRCA cells [127]. In a recent study, human breast cancer luminal cell line MCF-7 and basal cell line MDA-MB-231 were exposed to a medium containing extracellular vesicles derived from human umbilical cord mesenchymal stem cells (Huc-MSC-EVs), leading to enhanced cell proliferation, migration, and invasion through activation of the ERK pathway. A simultaneous decrease in E-cadherin expression and an increase in N-cadherin expression were observed, promoting the process of epithelial–mesenchymal transition in breast cancer cells [128].
MSCs exert their influence on the breast cancer stem cell pathway by secreting IL-6 and generating factors originating from mesenchymal cells, which may underlie the mechanism of MSC-BCSC mediation in breast cancer drug resistance [129]. The secretion of IL-6 by MSCs contributes to the development of chemotherapy resistance and encourages the proliferation of ERα+ breast cancer cells [130]. Notably, IL-6 can induce resistance development in breast cancer cells against cytotoxic drugs intended for ERα+ breast cancer (like paclitaxel and doxorubicin) and anti-HER2+ breast cancer treatment, like Herceptin [51].

3. Non-Cellular Components in the TME of TPBC

3.1. Tumor-Derived Exosomes (TDEs)

Exosomes are nanoscale membrane vesicles released by a variety of cell types and significantly contribute to breast cancer pathogenesis, encompassing tumor initiation, invasion, metastasis, angiogenesis, immune modulation, and the tumor microenvironment. Additionally, they are instrumental in the emergence of drug resistance [131]. Research highlights that exosomes can transport cellular components, including proteins (like cytokines, membrane receptors, and ligands), nucleic acids (such as DNA, mRNA, long and short non-coding RNAs), and lipids, which are capable of influencing other cells or being selectively transferred to recipient cells. This, in turn, affects the behavior and function of these recipient cells [132]. Consequently, they induce alterations in the tumor microenvironment via cytokine, growth factor, chemokine, and MMP secretion, subsequently fostering tumor progression by inducing processes like extracellular matrix remodeling, tumor angiogenesis, immune evasion, and escape [133]. Notably, exosomes can activate the EMT and WNT signaling pathways, which are prevalent regulatory pathways for modifying breast cancer metastatic potential. These exosomes can facilitate breast cancer cell metastasis by inducing EMT through proteins, like MAP17, present in their cargo [134]. Similarly, exosomal miR-146a can reportedly trigger breast cancer cell metastasis by activating the WNT signaling pathway [135]. Furthermore, exosomes released by highly metastatic breast cancer cells can disrupt the blood–brain barrier, thereby facilitating brain metastasis [136]. Recent studies have underscored the potential of exosomal miRNAs as a novel avenue for breast cancer treatment [137], especially against triple-negative breast cancer [138].
In their capacity as adept carriers, exosomes transport molecules, such as miRNAs and proteins, to recipient cells and the tumor microenvironment. Consequently, they directly or indirectly influence cancer cell drug resistance via interactions with the tumor microenvironment. Exosomes enhance breast cancer cell resistance to drugs by potentiating anti-apoptotic capacities, inducing cancer cell autophagy, expediting drug efflux, inducing immunosuppression, or heightening cell stemness. A significant correlation exists between breast cancer metastasis and drug resistance. Cellular exosomes originating from chemotherapy-resistant breast cancer may foster metastasis in cancer cells through the EphA2-Ephrin A1 reverse signaling [139]. Moreover, exosomes from radiation-resistant breast cancer cells, once absorbed by radiation-sensitive cells, can augment the metastatic potential of the latter [140]. This underscores the role of exosomes in both promoting breast cancer metastasis and inducing drug resistance through pathways linked to metastasis.
Importantly, TDEs could potentially contribute to the drug resistance encountered in TPBC. In terms of endocrine therapy, cellular exosomal miRNA-205 could hinder apoptosis in breast cancer cells by blocking the Caspase signaling pathway, thereby heightening the cells’ resistance to tamoxifen [141]. In tamoxifen-resistant breast cancer cells, exosomes released by these cells might negatively regulate ADIPOQ expression in recipient cells, rendering more cancer cells tolerant to chemotherapy drugs upon interaction with surrounding drug-sensitive tumor cells [142]. Regarding targeted therapy, breast cancer serum exosome AGAP2-AS1 has been associated with altered drug resistance patterns in the context of the anti-tumor drug trastuzumab [143,144]. Autophagy, a protective cellular mechanism, is triggered by anticancer drugs in cancer cells [145]. The exosomes released by trastuzumab-resistant breast cancer cells are rich in lncRNA AGAP2-AS1, which activates the autophagy reaction of cancer cells after targeting ATG10 [143]. Importantly, exosomal transport of miR-567 into breast cancer cells can diminish their tolerance to trastuzumab by inhibiting cancer cell autophagy [146].

3.2. Extracellular Matrix (ECM)

The extracellular matrix primarily comprises proteins generated by stromal cells like epithelial cells, endothelial cells, and fibrocytes. These proteins modify tissue structure and integrity by arranging components spatially and altering their physical attributes. The ECM exerts its influence not only by virtue of its constituent proteins but also by aiding the action of soluble factors within the tumor microenvironment [49]. ECM stiffening, a form of ECM remodeling, is attributed to collagen accumulation and the cross-linking and aggregation of collagen fibers. This phenomenon has a strong correlation with the progression and clinical prognosis of breast cancer [147,148]. It is widely acknowledged that tumor cells are capable of modulating ECM composition, thereby hindering immune cells and therapeutic agents from accessing tumor cells. This orchestrated effect serves to facilitate immune evasion and confer tolerance to drug treatments. Importantly, the component proteins of the ECM are intricately linked to tamoxifen resistance in breast cancer [149].

3.3. Cytokines and Growth Factors

Cytokines and growth factors represent soluble factors within the tumor microenvironment produced by various cell types, including epithelial cells, immune cells, and stromal cells. These molecules engage in signaling interactions between tumor cells and other components within the tumor microenvironment. Consequently, they influence the growth, differentiation, and movement of tumor cells. In the context of ER-positive breast cancer cells, paracrine signaling involving the platelet-derived growth factor (PDGF-CC) stimulates CAFs through binding to the platelet-derived growth factor receptor (PDGFR). This stimulation prompts CAFs to express a hepatocyte growth factor (HGF), an insulin-like growth factor binding protein 3 (IGFBP3), and stanniocalcin1 (STC1), ultimately leading to endocrine therapy resistance. Notably, targeted inhibition of PDGF-CC has the potential to restore tumor cell sensitivity to endocrine therapy, highlighting a potential avenue for intervention [150].

4. Discussion

The TME plays a pivotal role throughout all stages of BRCA development [151]. It primarily comprises various cell types, including fibroblasts, macrophages, and endothelial cells. Among these, CAFs stand out as influential entities in the TME, actively releasing cytokines and chemokines that substantially contribute to the growth and invasiveness of cancer cells [152,153]. The administration of chemotherapy drugs triggers tumor cells to secrete diverse cytokines, a process that recruits CAFs [154,155]. In turn, CAFs can foster drug resistance in tumor cells by releasing cellular exosomes [156]. Although CAFs have been implicated in driving drug resistance in breast cancer cells, the precise mechanisms underlying their interactions with tumor cells warrant further scientific investigation. Notably, tumors rich in CAFs have been linked to inferior prognoses compared to those with fewer CAFs [30]. This correlation is particularly pronounced in TNBC, where patients with stroma-rich tumors tend to experience worse overall survival outcomes than those with stroma-poor tumors [157]. Thus, molecules secreted by CAFs, including CXCL14 and CCL5, have emerged as potential therapeutic targets for TNBC treatment [158]. Nevertheless, the role of CAFs within the tumor microenvironment of TPBC remains understudied. The molecular mechanisms underlying how CAFs influence TPBC proliferation, progression, and invasiveness remain unclear, and further exploration is needed to pinpoint potential therapeutic targets for TPBC involving CAFs.
Peritumoral adipocytes undergo regulatory changes instigated by tumor cells, transforming into cancer-associated adipocytes (CAAs). These CAAs subsequently secrete pro-inflammatory cytokines, growth factors, extracellular matrix proteins, and metabolites. These secretions collectively modulate energy metabolism, extracellular matrix remodeling, and immunomodulatory functions within tumor cells, thereby promoting the initiation and progression of breast cancer [159]. Furthermore, exosomes released by adipocytes could expedite the malignant evolution of breast cancer by triggering the Hippo signaling pathway and prompting metabolic reprogramming [160]. In light of these findings, CAAs represent a promising target for breast cancer treatment; however, their practical application remains a distant objective. Future scientific research must address several issues. Firstly, it is crucial to decipher the intricate interactions and specific mechanisms that occur between CAAs and other constituents of the tumor microenvironment, including triple-positive breast cancer cells, tumor-associated fibroblasts, and immune cells. Additionally, it is essential to ascertain whether CAAs predominantly mediate the association between obesity and triple-positive breast cancer.
Non-cellular exosomes within the tumor microenvironment exert a distinct influence on breast cancer cell drug resistance. Cellular exosomes are widely dispersed across various body fluids [161]. Possessing bilayer lipid membranes and nanoscale dimensions, exosomes are adept at evading clearance or damage by cells, such as complements or macrophages. This unique property significantly prolongs the circulating half-life of exosomal contents, enhancing their biological activity [162]. Notably, nanoparticles enveloped in cellular exosome membranes can protect internal siRNA and drugs from degradation, offering the potential for targeted delivery to tumor cells [163,164]. This underscores the potential utility of exosomes and their contents as markers for monitoring drug resistance in breast cancer. Relevant investigations have detected alterations in plasma exosome-associated components among breast tumor patients, offering predictive value for changes in breast tumor cell sensitivity to chemoradiotherapy drugs. Notably, an elevated expression of TK1, CDK9, and CD44 within plasma exosomes from drug-resistant breast cancer patients correlates with therapy resistance [165,166]. Chemoradiotherapy drugs not only prompt the release of exosomes from drug-resistant tumor cells but also elicit exosome release from other cells within the tumor microenvironment. This indirect influence subsequently impacts chemotherapy drug resistance in breast cancer cells. For instance, doxorubicin therapy triggers the upregulated expression of miR-21-5p within exosomes secreted by mesenchymal stem cells, subsequently inducing doxorubicin resistance in breast cancer cells via the microenvironment [165]. Moreover, stimulation of breast cancer cells with paclitaxel enhances their exosome-releasing capacity [167], an effect observed even when breast cancer cells are insensitive to aromatase inhibitor therapy [168]. Furthermore, tumor-released exosomes can modulate the sensitivity of breast cancer cells to drugs by dampening immune cell-mediated inflammatory responses and inducing immunosuppressive cells. Indeed, breast cancer-derived exosomes absorbed by bone marrow cells activate STAT3, leading to CXCR4 reduction. Consequently, bone marrow cells differentiate into bone marrow-derived suppressor cells (MDSCs), inhibiting T lymphocyte proliferation and considerably reducing cancer cell sensitivity to chemotherapy drugs [169]. Moreover, tumor-induced mesenchymal stem cell exosomes can drive MDSC differentiation and foster their transformation into M2-type macrophages endowed with potent immunosuppressive traits, thereby attenuating the anti-tumor effect [170]. Last but not least, radiotherapy further triggers the release of exosomes from tumor cells. As breast cancer cell damage increases, exosome secretion activity and pathways are heightened proportionally to the radiotherapy dose. Importantly, exosomes from drug-treated breast cancer cells may exert an influence on drug resistance in untreated cells within the same group [171]. In essence, exosomes emerge as a critical player in the development of breast cancer drug resistance, offering a valuable avenue for tackling clinical breast cancer treatment resistance. Although research on exosomes in TPBC is limited, further exploration of their mechanisms within the tumor microenvironment is crucial for understanding the progression of drug resistance in TPBC.

5. Conclusions and Perspectives

As of 2021, breast cancer has surpassed lung cancer to become the most prevalent cancer globally, with a significant increase in incidence both domestically and internationally [172,173]. Roughly 25% of all breast cancers exhibit HER2 overexpression, while approximately 50% of HER2-positive breast cancers display positive hormone receptor expression [174,175]. TPBC stands as a distinct subtype characterized by unique tumor cell traits, molecular functions, cellular signaling pathways, therapeutic responsiveness, and biological behavior, diverging from other molecular subtypes [176]. For TPBC patients, both targeted and endocrine therapies are options (Table 1). However, due to the intersection of the HER2 gene and ER-mediated signaling pathways, TPBC features a distinct drug response and resistance mechanism unlike other molecular subtypes [177].
Although the TME significantly influences tumor development and therapeutic responsiveness, cancer cells do not passively respond to TME regulation. Instead, they engage in intricate interactions with the tumor microenvironment, synergistically promoting cancer cell survival, proliferation, and metastasis. Although research on the tumor microenvironment in TNBC has gained momentum, TPBC remains largely underexplored. This review discussed potential TME components relevant to the development of TPBC tumors and resistance to endocrine and trastuzumab therapies (Table 2). Notably, among these components, CAAs and exosomes seem to play pivotal roles in TPBC tumor progression and treatment resistance. However, the intricate nature of TME constituents poses challenges for in vitro studies that strive to replicate the true tumor microenvironment, resulting in limited relevant research. Consequently, future investigations should prioritize the establishment of robust experimental models, utilizing as many TPBC cell lines as possible, to accurately simulate the TPBC tumor microenvironment.

Author Contributions

Conceptualization, writing—review and editing, visualization, supervision, project administration, funding acquisition: H.G. Methodology, software, investigation, resources, data curation, writing—original draft preparation: Q.P. All authors have read and agreed to the published version of the manuscript.

Funding

This research was funded by the Qilu Hospital of Shandong University (Qingdao) Flexible talent guidance project grant No. QDKY2022RX01 And The APC was funded by the Key projects of Qingdao Science and Technology Plan grant No.23–2-8-smjk-13-nsh.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Sung, H.; Ferlay, J.; Siegel, R.L.; Laversanne, M.; Soerjomataram, I.; Jemal, A.; Bray, F. Global Cancer Statistics 2020: GLOBOCAN Estimates of Incidence and Mortality Worldwide for 36 Cancers in 185 Countries. CA Cancer J. Clin. 2021, 71, 209–249. [Google Scholar] [CrossRef]
  2. Zhu, K.; Chen, Y.; Guo, R.; Dai, L.; Wang, J.; Tang, Y.; Zhou, S.; Chen, D.; Huang, S. Prognostic Factor Analysis and Model Construction of Triple-Negative Metaplastic Breast Carcinoma After Surgery. Front. Oncol. 2022, 12, 924342. [Google Scholar] [CrossRef] [PubMed]
  3. Goldhirsch, A.; Winer, E.P.; Coates, A.S.; Gelber, R.D.; Piccart-Gebhart, M.; Thurlimann, B.; Senn, H.J.; Panel, M. Personalizing the treatment of women with early breast cancer: Highlights of the St Gallen International Expert Consensus on the Primary Therapy of Early Breast Cancer 2013. Ann. Oncol. 2013, 24, 2206–2223. [Google Scholar] [CrossRef] [PubMed]
  4. Katsura, C.; Ogunmwonyi, I.; Kankam, H.K.; Saha, S. Breast cancer: Presentation, investigation and management. Br. J. Hosp. Med. 2022, 83, 1–7. [Google Scholar] [CrossRef]
  5. Jaykumar, A.B.; Jung, J.U.; Parida, P.K.; Dang, T.T.; Wichaidit, C.; Kannangara, A.R.; Earnest, S.; Goldsmith, E.J.; Pearson, G.W.; Malladi, S.; et al. WNK1 Enhances Migration and Invasion in Breast Cancer Models. Mol. Cancer Ther. 2021, 20, 1800–1808. [Google Scholar] [CrossRef]
  6. Thomas, M.; Kelly, E.D.; Abraham, J.; Kruse, M. Invasive lobular breast cancer: A review of pathogenesis, diagnosis, management, and future directions of early stage disease. Semin. Oncol. 2019, 46, 121–132. [Google Scholar] [CrossRef] [PubMed]
  7. Simmons, A.; Burrage, P.M.; Nicolau, D.V., Jr.; Lakhani, S.R.; Burrage, K. Environmental factors in breast cancer invasion: A mathematical modelling review. Pathology 2017, 49, 172–180. [Google Scholar] [CrossRef]
  8. Niu, N.; Qiu, F.; Xu, Q.; He, G.; Gu, X.; Guo, W.; Zhang, D.; Li, Z.; Zhao, Y.; Li, Y.; et al. A multicentre single arm phase 2 trial of neoadjuvant pyrotinib and letrozole plus dalpiciclib for triple-positive breast cancer. Nat. Commun. 2022, 13, 7043. [Google Scholar] [CrossRef] [PubMed]
  9. Kast, K.; Link, T.; Friedrich, K.; Petzold, A.; Niedostatek, A.; Schoffer, O.; Werner, C.; Klug, S.J.; Werner, A.; Gatzweiler, A.; et al. Impact of breast cancer subtypes and patterns of metastasis on outcome. Breast Cancer Res. Treat. 2015, 150, 621–629. [Google Scholar] [CrossRef]
  10. Yazdanpanah, P.; Alavianmehr, A.; Ghaderi, A.; Monabati, A.; Montazer, M.; Tahmasbi, K.; Farjadian, S. PD-L1 expression in tumor lesions and soluble PD-L1 serum levels in patients with breast cancer: TNBC versus TPBC. Breast Dis. 2021, 40, 43–50. [Google Scholar] [CrossRef]
  11. Geng, A.; Xiao, J.; Dong, B.; Yuan, S. Analysis of prognostic factors and construction of prognostic models for triple-positive breast cancer. Front. Oncol. 2023, 13, 1071076. [Google Scholar] [CrossRef] [PubMed]
  12. Elsers, D.A.; Masoud, E.M.; Kamel, N.; Ahmed, A.M. Immunohistochemical signaling pathways of triple negative and triple positive breast cancers: What is new? Ann. Diagn. Pathol. 2021, 55, 151831. [Google Scholar] [CrossRef] [PubMed]
  13. Hutchinson, L. Breast cancer: Challenges, controversies, breakthroughs. Nat. Rev. Clin. Oncol. 2010, 7, 669–670. [Google Scholar] [CrossRef]
  14. Negi, P.; Kingsley, P.A.; Jain, K.; Sachdeva, J.; Srivastava, H.; Marcus, S.; Pannu, A. Survival of Triple Negative versus Triple Positive Breast Cancers: Comparison and Contrast. Asian Pac. J. Cancer Prev. 2016, 17, 3911–3916. [Google Scholar]
  15. Alzahrani, W.; Althoubaity, F.; Alsobhi, D.; Mohamed, Y.; AlMutairi, A.; Sindi, D.; Alharbi, R.; Zaidi, N. Clinicopathological Features and Metastatic Pattern of Triple-positive Breast Cancer Among Female Patients at a Tertiary Care Hospital. Cureus 2019, 11, e6458. [Google Scholar] [CrossRef]
  16. Vici, P.; Pizzuti, L.; Natoli, C.; Gamucci, T.; Di Lauro, L.; Barba, M.; Sergi, D.; Botti, C.; Michelotti, A.; Moscetti, L.; et al. Triple positive breast cancer: A distinct subtype? Cancer Treat. Rev. 2015, 41, 69–76. [Google Scholar] [CrossRef]
  17. Pegram, M.; Jackisch, C.; Johnston, S.R.D. Estrogen/HER2 receptor crosstalk in breast cancer: Combination therapies to improve outcomes for patients with hormone receptor-positive/HER2-positive breast cancer. NPJ Breast Cancer 2023, 9, 45. [Google Scholar] [CrossRef]
  18. Bergers, G.; Fendt, S.M. The metabolism of cancer cells during metastasis. Nat. Rev. Cancer 2021, 21, 162–180. [Google Scholar] [CrossRef] [PubMed]
  19. Mao, Y.; Keller, E.T.; Garfield, D.H.; Shen, K.; Wang, J. Stromal cells in tumor microenvironment and breast cancer. Cancer Metastasis Rev. 2013, 32, 303–315. [Google Scholar] [CrossRef] [PubMed]
  20. Cook, J.; Hagemann, T. Tumour-associated macrophages and cancer. Curr. Opin. Pharmacol. 2013, 13, 595–601. [Google Scholar] [CrossRef] [PubMed]
  21. Pattabiraman, D.R.; Weinberg, R.A. Tackling the cancer stem cells—What challenges do they pose? Nat. Rev. Drug Discov. 2014, 13, 497–512. [Google Scholar] [CrossRef] [PubMed]
  22. Quail, D.F.; Joyce, J.A. Microenvironmental regulation of tumor progression and metastasis. Nat. Med. 2013, 19, 1423–1437. [Google Scholar] [CrossRef] [PubMed]
  23. Binnewies, M.; Roberts, E.W.; Kersten, K.; Chan, V.; Fearon, D.F.; Merad, M.; Coussens, L.M.; Gabrilovich, D.I.; Ostrand-Rosenberg, S.; Hedrick, C.C.; et al. Understanding the tumor immune microenvironment (TIME) for effective therapy. Nat. Med. 2018, 24, 541–550. [Google Scholar] [CrossRef] [PubMed]
  24. Rauner, G.; Kuperwasser, C. Microenvironmental control of cell fate decisions in mammary gland development and cancer. Dev. Cell 2021, 56, 1875–1883. [Google Scholar] [CrossRef] [PubMed]
  25. Diaz Bessone, M.I.; Gattas, M.J.; Laporte, T.; Tanaka, M.; Simian, M. The Tumor Microenvironment as a Regulator of Endocrine Resistance in Breast Cancer. Front. Endocrinol. 2019, 10, 547. [Google Scholar] [CrossRef]
  26. Yamamura, Y.; Asai, N.; Enomoto, A.; Kato, T.; Mii, S.; Kondo, Y.; Ushida, K.; Niimi, K.; Tsunoda, N.; Nagino, M.; et al. Akt-Girdin signaling in cancer-associated fibroblasts contributes to tumor progression. Cancer Res. 2015, 75, 813–823. [Google Scholar] [CrossRef] [PubMed]
  27. Aoto, K.; Ito, K.; Aoki, S. Complex formation between platelet-derived growth factor receptor beta and transforming growth factor beta receptor regulates the differentiation of mesenchymal stem cells into cancer-associated fibroblasts. Oncotarget 2018, 9, 34090–34102. [Google Scholar] [CrossRef] [PubMed]
  28. Qiao, A.; Gu, F.; Guo, X.; Zhang, X.; Fu, L. Breast cancer-associated fibroblasts: Their roles in tumor initiation, progression and clinical applications. Front. Med. 2016, 10, 33–40. [Google Scholar] [CrossRef] [PubMed]
  29. Shiga, K.; Hara, M.; Nagasaki, T.; Sato, T.; Takahashi, H.; Takeyama, H. Cancer-Associated Fibroblasts: Their Characteristics and Their Roles in Tumor Growth. Cancers 2015, 7, 2443–2458. [Google Scholar] [CrossRef]
  30. Aboussekhra, A. Role of cancer-associated fibroblasts in breast cancer development and prognosis. Int. J. Dev. Biol. 2011, 55, 841–849. [Google Scholar] [CrossRef] [PubMed]
  31. Luo, H.; Tu, G.; Liu, Z.; Liu, M. Cancer-associated fibroblasts: A multifaceted driver of breast cancer progression. Cancer Lett. 2015, 361, 155–163. [Google Scholar] [CrossRef] [PubMed]
  32. Conklin, M.W.; Keely, P.J. Why the stroma matters in breast cancer: Insights into breast cancer patient outcomes through the examination of stromal biomarkers. Cell Adh. Migr. 2012, 6, 249–260. [Google Scholar] [CrossRef] [PubMed]
  33. Feng, B.; Wu, J.; Shen, B.; Jiang, F.; Feng, J. Cancer-associated fibroblasts and resistance to anticancer therapies: Status, mechanisms, and countermeasures. Cancer Cell Int. 2022, 22, 166. [Google Scholar] [CrossRef] [PubMed]
  34. Shekhar, M.P.; Werdell, J.; Santner, S.J.; Pauley, R.J.; Tait, L. Breast stroma plays a dominant regulatory role in breast epithelial growth and differentiation: Implications for tumor development and progression. Cancer Res. 2001, 61, 1320–1326. [Google Scholar]
  35. Chaffer, C.L.; Marjanovic, N.D.; Lee, T.; Bell, G.; Kleer, C.G.; Reinhardt, F.; D’Alessio, A.C.; Young, R.A.; Weinberg, R.A. Poised chromatin at the ZEB1 promoter enables breast cancer cell plasticity and enhances tumorigenicity. Cell 2013, 154, 61–74. [Google Scholar] [CrossRef] [PubMed]
  36. Han, Y.; Zhang, Y.; Jia, T.; Sun, Y. Molecular mechanism underlying the tumor-promoting functions of carcinoma-associated fibroblasts. Tumour Biol. 2015, 36, 1385–1394. [Google Scholar] [CrossRef]
  37. Katoh, M. FGFR inhibitors: Effects on cancer cells, tumor microenvironment and whole-body homeostasis (Review). Int. J. Mol. Med. 2016, 38, 3–15. [Google Scholar] [CrossRef] [PubMed]
  38. Lisanti, M.P.; Tsirigos, A.; Pavlides, S.; Reeves, K.J.; Peiris-Pages, M.; Chadwick, A.L.; Sanchez-Alvarez, R.; Lamb, R.; Howell, A.; Martinez-Outschoorn, U.E.; et al. JNK1 stress signaling is hyper-activated in high breast density and the tumor stroma: Connecting fibrosis, inflammation, and stemness for cancer prevention. Cell Cycle 2014, 13, 580–599. [Google Scholar] [CrossRef]
  39. Ahirwar, D.K.; Nasser, M.W.; Ouseph, M.M.; Elbaz, M.; Cuitino, M.C.; Kladney, R.D.; Varikuti, S.; Kaul, K.; Satoskar, A.R.; Ramaswamy, B.; et al. Fibroblast-derived CXCL12 promotes breast cancer metastasis by facilitating tumor cell intravasation. Oncogene 2018, 37, 4428–4442. [Google Scholar] [CrossRef]
  40. Erez, N.; Glanz, S.; Raz, Y.; Avivi, C.; Barshack, I. Cancer associated fibroblasts express pro-inflammatory factors in human breast and ovarian tumors. Biochem. Biophys. Res. Commun. 2013, 437, 397–402. [Google Scholar] [CrossRef] [PubMed]
  41. Servais, C.; Erez, N. From sentinel cells to inflammatory culprits: Cancer-associated fibroblasts in tumour-related inflammation. J. Pathol. 2013, 229, 198–207. [Google Scholar] [CrossRef]
  42. Pandey, D.P.; Lappano, R.; Albanito, L.; Madeo, A.; Maggiolini, M.; Picard, D. Estrogenic GPR30 signalling induces proliferation and migration of breast cancer cells through CTGF. EMBO J. 2009, 28, 523–532. [Google Scholar] [CrossRef] [PubMed]
  43. De Francesco, E.M.; Pellegrino, M.; Santolla, M.F.; Lappano, R.; Ricchio, E.; Abonante, S.; Maggiolini, M. GPER mediates activation of HIF1alpha/VEGF signaling by estrogens. Cancer Res. 2014, 74, 4053–4064. [Google Scholar] [CrossRef] [PubMed]
  44. Lappano, R.; Santolla, M.F.; Pupo, M.; Sinicropi, M.S.; Caruso, A.; Rosano, C.; Maggiolini, M. MIBE acts as antagonist ligand of both estrogen receptor alpha and GPER in breast cancer cells. Breast Cancer Res. 2012, 14, R12. [Google Scholar] [CrossRef] [PubMed]
  45. De Marco, P.; Lappano, R.; De Francesco, E.M.; Cirillo, F.; Pupo, M.; Avino, S.; Vivacqua, A.; Abonante, S.; Picard, D.; Maggiolini, M. GPER signalling in both cancer-associated fibroblasts and breast cancer cells mediates a feedforward IL1beta/IL1R1 response. Sci. Rep. 2016, 6, 24354. [Google Scholar] [CrossRef]
  46. Chen, B.; Sang, Y.; Song, X.; Zhang, D.; Wang, L.; Zhao, W.; Liang, Y.; Zhang, N.; Yang, Q. Exosomal miR-500a-5p derived from cancer-associated fibroblasts promotes breast cancer cell proliferation and metastasis through targeting USP28. Theranostics 2021, 11, 3932–3947. [Google Scholar] [CrossRef]
  47. Sun, X.; Mao, Y.; Wang, J.; Zu, L.; Hao, M.; Cheng, G.; Qu, Q.; Cui, D.; Keller, E.T.; Chen, X.; et al. IL-6 secreted by cancer-associated fibroblasts induces tamoxifen resistance in luminal breast cancer. Oncogene 2014, 33, 4450. [Google Scholar] [CrossRef]
  48. Luo, H.; Yang, G.; Yu, T.; Luo, S.; Wu, C.; Sun, Y.; Liu, M.; Tu, G. GPER-mediated proliferation and estradiol production in breast cancer-associated fibroblasts. Endocr. Relat. Cancer 2014, 21, 355–369. [Google Scholar] [CrossRef]
  49. Pontiggia, O.; Sampayo, R.; Raffo, D.; Motter, A.; Xu, R.; Bissell, M.J.; Joffe, E.B.; Simian, M. The tumor microenvironment modulates tamoxifen resistance in breast cancer: A role for soluble stromal factors and fibronectin through beta1 integrin. Breast Cancer Res. Treat. 2012, 133, 459–471. [Google Scholar] [CrossRef]
  50. Yuan, J.; Liu, M.; Yang, L.; Tu, G.; Zhu, Q.; Chen, M.; Cheng, H.; Luo, H.; Fu, W.; Li, Z.; et al. Acquisition of epithelial-mesenchymal transition phenotype in the tamoxifen-resistant breast cancer cell: A new role for G protein-coupled estrogen receptor in mediating tamoxifen resistance through cancer-associated fibroblast-derived fibronectin and beta1-integrin signaling pathway in tumor cells. Breast Cancer Res. 2015, 17, 69. [Google Scholar]
  51. Zhong, H.; Davis, A.; Ouzounova, M.; Carrasco, R.A.; Chen, C.; Breen, S.; Chang, Y.S.; Huang, J.; Liu, Z.; Yao, Y.; et al. A Novel IL6 Antibody Sensitizes Multiple Tumor Types to Chemotherapy Including Trastuzumab-Resistant Tumors. Cancer Res. 2016, 76, 480–490. [Google Scholar] [CrossRef] [PubMed]
  52. Mao, Y.; Zhang, Y.; Qu, Q.; Zhao, M.; Lou, Y.; Liu, J.; Huang, O.; Chen, X.; Wu, J.; Shen, K. Cancer-associated fibroblasts induce trastuzumab resistance in HER2 positive breast cancer cells. Mol. Biosyst. 2015, 11, 1029–1040. [Google Scholar] [CrossRef] [PubMed]
  53. Celia-Terrassa, T.; Kang, Y. Metastatic niche functions and therapeutic opportunities. Nat. Cell Biol. 2018, 20, 868–877. [Google Scholar] [CrossRef] [PubMed]
  54. Wang, N.; Wang, S.; Wang, X.; Zheng, Y.; Yang, B.; Zhang, J.; Pan, B.; Gao, J.; Wang, Z. Research trends in pharmacological modulation of tumor-associated macrophages. Clin. Transl. Med. 2021, 11, e288. [Google Scholar] [CrossRef]
  55. Pan, Y.; Yu, Y.; Wang, X.; Zhang, T. Tumor-Associated Macrophages in Tumor Immunity. Front. Immunol. 2020, 11, 583084. [Google Scholar] [CrossRef]
  56. Yuan, A.; Hsiao, Y.J.; Chen, H.Y.; Chen, H.W.; Ho, C.C.; Chen, Y.Y.; Liu, Y.C.; Hong, T.H.; Yu, S.L.; Chen, J.J.; et al. Opposite Effects of M1 and M2 Macrophage Subtypes on Lung Cancer Progression. Sci. Rep. 2015, 5, 14273. [Google Scholar] [CrossRef]
  57. Prenen, H.; Mazzone, M. Tumor-associated macrophages: A short compendium. Cell. Mol. Life Sci. 2019, 76, 1447–1458. [Google Scholar] [CrossRef]
  58. Martinez, F.O.; Gordon, S. The M1 and M2 paradigm of macrophage activation: Time for reassessment. F1000Prime Rep. 2014, 6, 13. [Google Scholar] [CrossRef]
  59. Petty, A.J.; Yang, Y. Tumor-associated macrophages: Implications in cancer immunotherapy. Immunotherapy 2017, 9, 289–302. [Google Scholar] [CrossRef]
  60. Miyasato, Y.; Shiota, T.; Ohnishi, K.; Pan, C.; Yano, H.; Horlad, H.; Yamamoto, Y.; Yamamoto-Ibusuki, M.; Iwase, H.; Takeya, M.; et al. High density of CD204-positive macrophages predicts worse clinical prognosis in patients with breast cancer. Cancer Sci. 2017, 108, 1693–1700. [Google Scholar] [CrossRef]
  61. Zhao, X.; Qu, J.; Sun, Y.; Wang, J.; Liu, X.; Wang, F.; Zhang, H.; Wang, W.; Ma, X.; Gao, X.; et al. Prognostic significance of tumor-associated macrophages in breast cancer: A meta-analysis of the literature. Oncotarget 2017, 8, 30576–30586. [Google Scholar] [CrossRef] [PubMed]
  62. Pelekanou, V.; Villarroel-Espindola, F.; Schalper, K.A.; Pusztai, L.; Rimm, D.L. CD68, CD163, and matrix metalloproteinase 9 (MMP-9) co-localization in breast tumor microenvironment predicts survival differently in ER-positive and -negative cancers. Breast Cancer Res. 2018, 20, 154. [Google Scholar] [CrossRef]
  63. Mansfield, A.S.; Heikkila, P.; von Smitten, K.; Vakkila, J.; Leidenius, M. The presence of sinusoidal CD163(+) macrophages in lymph nodes is associated with favorable nodal status in patients with breast cancer. Virchows Arch. 2012, 461, 639–646. [Google Scholar] [CrossRef] [PubMed]
  64. Fan, Y.; He, S. The Characteristics of Tumor Microenvironment in Triple Negative Breast Cancer. Cancer Manag. Res. 2022, 14, 1–17. [Google Scholar] [CrossRef]
  65. Lu, J.; Ma, L. The role of tumor-associated macrophages in the development, metastasis and treatment of breast cancer. Pathol. Res. Pract. 2020, 216, 153085. [Google Scholar] [CrossRef] [PubMed]
  66. Gomez, V.; Eykyn, T.R.; Mustapha, R.; Flores-Borja, F.; Male, V.; Barber, P.R.; Patsialou, A.; Green, R.; Panagaki, F.; Li, C.W.; et al. Breast cancer-associated macrophages promote tumorigenesis by suppressing succinate dehydrogenase in tumor cells. Sci. Signal. 2020, 13, eaax4585. [Google Scholar] [CrossRef]
  67. Weng, Y.S.; Tseng, H.Y.; Chen, Y.A.; Shen, P.C.; Al Haq, A.T.; Chen, L.M.; Tung, Y.C.; Hsu, H.L. MCT-1/miR-34a/IL-6/IL-6R signaling axis promotes EMT progression, cancer stemness and M2 macrophage polarization in triple-negative breast cancer. Mol. Cancer 2019, 18, 42. [Google Scholar] [CrossRef]
  68. Hollmen, M.; Roudnicky, F.; Karaman, S.; Detmar, M. Characterization of macrophage--cancer cell crosstalk in estrogen receptor positive and triple-negative breast cancer. Sci. Rep. 2015, 5, 9188. [Google Scholar] [CrossRef]
  69. Li, J.; Wang, S.; Wang, N.; Zheng, Y.; Yang, B.; Wang, X.; Zhang, J.; Pan, B.; Wang, Z. Aiduqing formula inhibits breast cancer metastasis by suppressing TAM/CXCL1-induced Treg differentiation and infiltration. Cell Commun. Signal. 2021, 19, 89. [Google Scholar] [CrossRef] [PubMed]
  70. Nie, G.; Cao, X.; Mao, Y.; Lv, Z.; Lv, M.; Wang, Y.; Wang, H.; Liu, C. Tumor-associated macrophages-mediated CXCL8 infiltration enhances breast cancer metastasis: Suppression by Danirixin. Int. Immunopharmacol. 2021, 95, 107153. [Google Scholar] [CrossRef]
  71. Yang, C.; He, L.; He, P.; Liu, Y.; Wang, W.; He, Y.; Du, Y.; Gao, F. Increased drug resistance in breast cancer by tumor-associated macrophages through IL-10/STAT3/bcl-2 signaling pathway. Med. Oncol. 2015, 32, 352. [Google Scholar] [CrossRef] [PubMed]
  72. Li, D.; Ji, H.; Niu, X.; Yin, L.; Wang, Y.; Gu, Y.; Wang, J.; Zhou, X.; Zhang, H.; Zhang, Q. Tumor-associated macrophages secrete CC-chemokine ligand 2 and induce tamoxifen resistance by activating PI3K/Akt/mTOR in breast cancer. Cancer Sci. 2020, 111, 47–58. [Google Scholar] [CrossRef] [PubMed]
  73. Castellaro, A.M.; Rodriguez-Baili, M.C.; Di Tada, C.E.; Gil, G.A. Tumor-Associated Macrophages Induce Endocrine Therapy Resistance in ER+ Breast Cancer Cells. Cancers 2019, 11, 189. [Google Scholar] [CrossRef]
  74. Niu, X.; Ma, J.; Li, J.; Gu, Y.; Yin, L.; Wang, Y.; Zhou, X.; Wang, J.; Ji, H.; Zhang, Q. Sodium/glucose cotransporter 1-dependent metabolic alterations induce tamoxifen resistance in breast cancer by promoting macrophage M2 polarization. Cell Death Dis. 2021, 12, 509. [Google Scholar] [CrossRef]
  75. Chen, W.X.; Wang, D.D.; Zhu, B.; Zhu, Y.Z.; Zheng, L.; Feng, Z.Q.; Qin, X.H. Exosomal miR-222 from adriamycin-resistant MCF-7 breast cancer cells promote macrophages M2 polarization via PTEN/Akt to induce tumor progression. Aging 2021, 13, 10415–10430. [Google Scholar] [CrossRef]
  76. Vandeweyer, E.; Hertens, D. Quantification of glands and fat in breast tissue: An experimental determination. Ann. Anat. 2002, 184, 181–184. [Google Scholar] [CrossRef]
  77. Ramsay, D.T.; Kent, J.C.; Hartmann, R.A.; Hartmann, P.E. Anatomy of the lactating human breast redefined with ultrasound imaging. J. Anat. 2005, 206, 525–534. [Google Scholar] [CrossRef]
  78. Picon-Ruiz, M.; Morata-Tarifa, C.; Valle-Goffin, J.J.; Friedman, E.R.; Slingerland, J.M. Obesity and adverse breast cancer risk and outcome: Mechanistic insights and strategies for intervention. CA Cancer J. Clin. 2017, 67, 378–397. [Google Scholar] [CrossRef]
  79. Choi, J.; Cha, Y.J.; Koo, J.S. Adipocyte biology in breast cancer: From silent bystander to active facilitator. Prog. Lipid Res. 2018, 69, 11–20. [Google Scholar] [CrossRef]
  80. Balaban, S.; Shearer, R.F.; Lee, L.S.; van Geldermalsen, M.; Schreuder, M.; Shtein, H.C.; Cairns, R.; Thomas, K.C.; Fazakerley, D.J.; Grewal, T.; et al. Adipocyte lipolysis links obesity to breast cancer growth: Adipocyte-derived fatty acids drive breast cancer cell proliferation and migration. Cancer Metab. 2017, 5, 1. [Google Scholar] [CrossRef] [PubMed]
  81. Hoy, A.J.; Balaban, S.; Saunders, D.N. Adipocyte-Tumor Cell Metabolic Crosstalk in Breast Cancer. Trends Mol. Med. 2017, 23, 381–392. [Google Scholar] [CrossRef]
  82. Bertolini, F.; Lohsiriwat, V.; Petit, J.Y.; Kolonin, M.G. Adipose tissue cells, lipotransfer and cancer: A challenge for scientists, oncologists and surgeons. Biochim. Biophys. Acta 2012, 1826, 209–214. [Google Scholar] [CrossRef]
  83. Cinti, S.; Mitchell, G.; Barbatelli, G.; Murano, I.; Ceresi, E.; Faloia, E.; Wang, S.; Fortier, M.; Greenberg, A.S.; Obin, M.S. Adipocyte death defines macrophage localization and function in adipose tissue of obese mice and humans. J. Lipid Res. 2005, 46, 2347–2355. [Google Scholar] [CrossRef] [PubMed]
  84. Morris, P.G.; Hudis, C.A.; Giri, D.; Morrow, M.; Falcone, D.J.; Zhou, X.K.; Du, B.; Brogi, E.; Crawford, C.B.; Kopelovich, L.; et al. Inflammation and increased aromatase expression occur in the breast tissue of obese women with breast cancer. Cancer Prev. Res. 2011, 4, 1021–1029. [Google Scholar] [CrossRef] [PubMed]
  85. Subbaramaiah, K.; Howe, L.R.; Bhardwaj, P.; Du, B.; Gravaghi, C.; Yantiss, R.K.; Zhou, X.K.; Blaho, V.A.; Hla, T.; Yang, P.; et al. Obesity is associated with inflammation and elevated aromatase expression in the mouse mammary gland. Cancer Prev. Res. 2011, 4, 329–346. [Google Scholar] [CrossRef]
  86. Subbaramaiah, K.; Morris, P.G.; Zhou, X.K.; Morrow, M.; Du, B.; Giri, D.; Kopelovich, L.; Hudis, C.A.; Dannenberg, A.J. Increased levels of COX-2 and prostaglandin E2 contribute to elevated aromatase expression in inflamed breast tissue of obese women. Cancer Discov. 2012, 2, 356–365. [Google Scholar] [CrossRef] [PubMed]
  87. Bhardwaj, P.; Du, B.; Zhou, X.K.; Sue, E.; Giri, D.; Harbus, M.D.; Falcone, D.J.; Hudis, C.A.; Subbaramaiah, K.; Dannenberg, A.J. Estrogen Protects against Obesity-Induced Mammary Gland Inflammation in Mice. Cancer Prev. Res. 2015, 8, 751–759. [Google Scholar] [CrossRef]
  88. Bhardwaj, P.; Du, B.; Zhou, X.K.; Sue, E.; Harbus, M.D.; Falcone, D.J.; Giri, D.; Hudis, C.A.; Kopelovich, L.; Subbaramaiah, K.; et al. Caloric restriction reverses obesity-induced mammary gland inflammation in mice. Cancer Prev. Res. 2013, 6, 282–289. [Google Scholar] [CrossRef]
  89. Mullooly, M.; Yang, H.P.; Falk, R.T.; Nyante, S.J.; Cora, R.; Pfeiffer, R.M.; Radisky, D.C.; Visscher, D.W.; Hartmann, L.C.; Carter, J.M.; et al. Relationship between crown-like structures and sex-steroid hormones in breast adipose tissue and serum among postmenopausal breast cancer patients. Breast Cancer Res. 2017, 19, 8. [Google Scholar] [CrossRef]
  90. Cha, Y.J.; Kim, E.S.; Koo, J.S. Tumor-associated macrophages and crown-like structures in adipose tissue in breast cancer. Breast Cancer Res. Treat. 2018, 170, 15–25. [Google Scholar] [CrossRef]
  91. Fletcher, S.J.; Sacca, P.A.; Pistone-Creydt, M.; Colo, F.A.; Serra, M.F.; Santino, F.E.; Sasso, C.V.; Lopez-Fontana, C.M.; Caron, R.W.; Calvo, J.C.; et al. Human breast adipose tissue: Characterization of factors that change during tumor progression in human breast cancer. J. Exp. Clin. Cancer Res. 2017, 36, 26. [Google Scholar] [CrossRef]
  92. Lehr, S.; Hartwig, S.; Sell, H. Adipokines: A treasure trove for the discovery of biomarkers for metabolic disorders. Proteom. Clin. Appl. 2012, 6, 91–101. [Google Scholar] [CrossRef]
  93. Li, J.; Han, X. Adipocytokines and breast cancer. Curr. Probl. Cancer 2018, 42, 208–214. [Google Scholar] [CrossRef] [PubMed]
  94. Yin, N.; Wang, D.; Zhang, H.; Yi, X.; Sun, X.; Shi, B.; Wu, H.; Wu, G.; Wang, X.; Shang, Y. Molecular mechanisms involved in the growth stimulation of breast cancer cells by leptin. Cancer Res. 2004, 64, 5870–5875. [Google Scholar] [CrossRef] [PubMed]
  95. Catalano, S.; Mauro, L.; Marsico, S.; Giordano, C.; Rizza, P.; Rago, V.; Montanaro, D.; Maggiolini, M.; Panno, M.L.; Ando, S. Leptin induces, via ERK1/ERK2 signal, functional activation of estrogen receptor alpha in MCF-7 cells. J. Biol. Chem. 2004, 279, 19908–19915. [Google Scholar] [CrossRef]
  96. Soma, D.; Kitayama, J.; Yamashita, H.; Miyato, H.; Ishikawa, M.; Nagawa, H. Leptin augments proliferation of breast cancer cells via transactivation of HER2. J. Surg. Res. 2008, 149, 9–14. [Google Scholar] [CrossRef]
  97. Fiorio, E.; Mercanti, A.; Terrasi, M.; Micciolo, R.; Remo, A.; Auriemma, A.; Molino, A.; Parolin, V.; Di Stefano, B.; Bonetti, F.; et al. Leptin/HER2 crosstalk in breast cancer: In vitro study and preliminary in vivo analysis. BMC Cancer 2008, 8, 305. [Google Scholar] [CrossRef] [PubMed]
  98. Jiang, H.; Yu, J.; Guo, H.; Song, H.; Chen, S. Upregulation of survivin by leptin/STAT3 signaling in MCF-7 cells. Biochem. Biophys. Res. Commun. 2008, 368, 1–5. [Google Scholar] [CrossRef]
  99. Frankenberry, K.A.; Skinner, H.; Somasundar, P.; McFadden, D.W.; Vona-Davis, L.C. Leptin receptor expression and cell signaling in breast cancer. Int. J. Oncol. 2006, 28, 985–993. [Google Scholar] [CrossRef] [PubMed]
  100. Wei, L.; Li, K.; Pang, X.; Guo, B.; Su, M.; Huang, Y.; Wang, N.; Ji, F.; Zhong, C.; Yang, J.; et al. Leptin promotes epithelial-mesenchymal transition of breast cancer via the upregulation of pyruvate kinase M2. J. Exp. Clin. Cancer Res. 2016, 35, 166. [Google Scholar] [CrossRef] [PubMed]
  101. Yamauchi, T.; Kamon, J.; Waki, H.; Terauchi, Y.; Kubota, N.; Hara, K.; Mori, Y.; Ide, T.; Murakami, K.; Tsuboyama-Kasaoka, N.; et al. The fat-derived hormone adiponectin reverses insulin resistance associated with both lipoatrophy and obesity. Nat. Med. 2001, 7, 941–946. [Google Scholar] [CrossRef] [PubMed]
  102. Miyoshi, Y.; Funahashi, T.; Kihara, S.; Taguchi, T.; Tamaki, Y.; Matsuzawa, Y.; Noguchi, S. Association of serum adiponectin levels with breast cancer risk. Clin. Cancer Res. 2003, 9, 5699–5704. [Google Scholar]
  103. Sanford, D.E.; Belt, B.A.; Panni, R.Z.; Mayer, A.; Deshpande, A.D.; Carpenter, D.; Mitchem, J.B.; Plambeck-Suess, S.M.; Worley, L.A.; Goetz, B.D.; et al. Inflammatory monocyte mobilization decreases patient survival in pancreatic cancer: A role for targeting the CCL2/CCR2 axis. Clin. Cancer Res. 2013, 19, 3404–3415. [Google Scholar] [CrossRef]
  104. Dirat, B.; Bochet, L.; Dabek, M.; Daviaud, D.; Dauvillier, S.; Majed, B.; Wang, Y.Y.; Meulle, A.; Salles, B.; Le Gonidec, S.; et al. Cancer-associated adipocytes exhibit an activated phenotype and contribute to breast cancer invasion. Cancer Res. 2011, 71, 2455–2465. [Google Scholar] [CrossRef] [PubMed]
  105. Wang, F.; Gao, S.; Chen, F.; Fu, Z.; Yin, H.; Lu, X.; Yu, J.; Lu, C. Mammary fat of breast cancer: Gene expression profiling and functional characterization. PLoS ONE 2014, 9, e109742. [Google Scholar] [CrossRef]
  106. Yamauchi, T.; Kamon, J.; Ito, Y.; Tsuchida, A.; Yokomizo, T.; Kita, S.; Sugiyama, T.; Miyagishi, M.; Hara, K.; Tsunoda, M.; et al. Cloning of adiponectin receptors that mediate antidiabetic metabolic effects. Nature 2003, 423, 762–769. [Google Scholar] [CrossRef] [PubMed]
  107. Prall, O.W.; Rogan, E.M.; Musgrove, E.A.; Watts, C.K.; Sutherland, R.L. c-Myc or cyclin D1 mimics estrogen effects on cyclin E-Cdk2 activation and cell cycle reentry. Mol. Cell. Biol. 1998, 18, 4499–4508. [Google Scholar] [CrossRef] [PubMed]
  108. Dieudonne, M.N.; Bussiere, M.; Santos, E.D.; Leneveu, M.C.; Giudicelli, Y.; Pecquery, R. Adiponectin mediates antiproliferative and apoptotic responses in human MCF7 breast cancer cells. Biochem. Biophys. Res. Commun. 2006, 345, 271–279. [Google Scholar] [CrossRef]
  109. Saxena, N.K.; Sharma, D. Metastasis suppression by adiponectin: LKB1 rises up to the challenge. Cell Adh. Migr. 2010, 4, 358–362. [Google Scholar] [CrossRef]
  110. Al-Khalaf, H.H.; Al-Harbi, B.; Al-Sayed, A.; Arafah, M.; Tulbah, A.; Jarman, A.; Al-Mohanna, F.; Aboussekhra, A. Interleukin-8 Activates Breast Cancer-Associated Adipocytes and Promotes Their Angiogenesis- and Tumorigenesis-Promoting Effects. Mol. Cell. Biol. 2019, 39, e00332-18. [Google Scholar] [CrossRef]
  111. Benoy, I.H.; Salgado, R.; Van Dam, P.; Geboers, K.; Van Marck, E.; Scharpe, S.; Vermeulen, P.B.; Dirix, L.Y. Increased serum interleukin-8 in patients with early and metastatic breast cancer correlates with early dissemination and survival. Clin. Cancer Res. 2004, 10, 7157–7162. [Google Scholar] [CrossRef] [PubMed]
  112. Zuccari, C.; Leonel, C.; Castro, R.; Gelaleti, G.B.; Jardim, B.V.; Moscheta, M.G.; Regiani, V.R.; Ferreira, L.C.; Lopes, J.R.; De Neto, S.; et al. An immunohistochemical study of interleukin-8 (IL-8) in breast cancer. Acta Histochem. 2012, 114, 571–576. [Google Scholar] [CrossRef]
  113. Lin, Y.; Huang, R.; Chen, L.; Li, S.; Shi, Q.; Jordan, C.; Huang, R.P. Identification of interleukin-8 as estrogen receptor-regulated factor involved in breast cancer invasion and angiogenesis by protein arrays. Int. J. Cancer 2004, 109, 507–515. [Google Scholar] [CrossRef] [PubMed]
  114. Pantschenko, A.G.; Pushkar, I.; Anderson, K.H.; Wang, Y.; Miller, L.J.; Kurtzman, S.H.; Barrows, G.; Kreutzer, D.L. The interleukin-1 family of cytokines and receptors in human breast cancer: Implications for tumor progression. Int. J. Oncol. 2003, 23, 269–284. [Google Scholar] [CrossRef] [PubMed]
  115. Freund, A.; Chauveau, C.; Brouillet, J.P.; Lucas, A.; Lacroix, M.; Licznar, A.; Vignon, F.; Lazennec, G. IL-8 expression and its possible relationship with estrogen-receptor-negative status of breast cancer cells. Oncogene 2003, 22, 256–265. [Google Scholar] [CrossRef]
  116. Pantschenko, A.G.; Pushkar, I.; Miller, L.J.; Wang, Y.P.; Anderson, K.; Peled, Z.; Kurtzman, S.H.; Kreutzer, D.L. In vitro demonstration of breast cancer tumor cell sub-populations based on interleukin-1/tumor necrosis factor induction of interleukin-8 expression. Oncol. Rep. 2003, 10, 1011–1017. [Google Scholar] [CrossRef]
  117. Liu, Q.; Li, A.; Tian, Y.; Wu, J.D.; Liu, Y.; Li, T.; Chen, Y.; Han, X.; Wu, K. The CXCL8-CXCR1/2 pathways in cancer. Cytokine Growth Factor. Rev. 2016, 31, 61–71. [Google Scholar] [CrossRef]
  118. Wellberg, E.A.; Kabos, P.; Gillen, A.E.; Jacobsen, B.M.; Brechbuhl, H.M.; Johnson, S.J.; Rudolph, M.C.; Edgerton, S.M.; Thor, A.D.; Anderson, S.M.; et al. FGFR1 underlies obesity-associated progression of estrogen receptor-positive breast cancer after estrogen deprivation. JCI Insight 2018, 3, e120594. [Google Scholar] [CrossRef]
  119. Delort, L.; Bougaret, L.; Cholet, J.; Vermerie, M.; Billard, H.; Decombat, C.; Bourgne, C.; Berger, M.; Dumontet, C.; Caldefie-Chezet, F. Hormonal Therapy Resistance and Breast Cancer: Involvement of Adipocytes and Leptin. Nutrients 2019, 11, 2839. [Google Scholar] [CrossRef]
  120. Galipeau, J.; Sensebe, L. Mesenchymal Stromal Cells: Clinical Challenges and Therapeutic Opportunities. Cell Stem Cell 2018, 22, 824–833. [Google Scholar] [CrossRef]
  121. Dominici, M.; Le Blanc, K.; Mueller, I.; Slaper-Cortenbach, I.; Marini, F.; Krause, D.; Deans, R.; Keating, A.; Prockop, D.; Horwitz, E. Minimal criteria for defining multipotent mesenchymal stromal cells. The International Society for Cellular Therapy position statement. Cytotherapy 2006, 8, 315–317. [Google Scholar] [CrossRef]
  122. Nishimura, K.; Semba, S.; Aoyagi, K.; Sasaki, H.; Yokozaki, H. Mesenchymal stem cells provide an advantageous tumor microenvironment for the restoration of cancer stem cells. Pathobiology 2012, 79, 290–306. [Google Scholar] [CrossRef] [PubMed]
  123. Chan, T.S.; Shaked, Y.; Tsai, K.K. Targeting the Interplay Between Cancer Fibroblasts, Mesenchymal Stem Cells, and Cancer Stem Cells in Desmoplastic Cancers. Front. Oncol. 2019, 9, 688. [Google Scholar] [CrossRef]
  124. Ridge, S.M.; Sullivan, F.J.; Glynn, S.A. Mesenchymal stem cells: Key players in cancer progression. Mol. Cancer 2017, 16, 31. [Google Scholar] [CrossRef] [PubMed]
  125. Pakravan, K.; Babashah, S.; Sadeghizadeh, M.; Mowla, S.J.; Mossahebi-Mohammadi, M.; Ataei, F.; Dana, N.; Javan, M. MicroRNA-100 shuttled by mesenchymal stem cell-derived exosomes suppresses in vitro angiogenesis through modulating the mTOR/HIF-1alpha/VEGF signaling axis in breast cancer cells. Cell. Oncol. 2017, 40, 457–470. [Google Scholar] [CrossRef] [PubMed]
  126. Khazaei-Poul, Y.; Shojaei, S.; Koochaki, A.; Ghanbarian, H.; Mohammadi-Yeganeh, S. Evaluating the influence of Human Umbilical Cord Mesenchymal Stem Cells-derived exosomes loaded with miR-3182 on metastatic performance of Triple Negative Breast Cancer cells. Life Sci. 2021, 286, 120015. [Google Scholar] [CrossRef] [PubMed]
  127. Kucerova, L.; Skolekova, S.; Matuskova, M.; Bohac, M.; Kozovska, Z. Altered features and increased chemosensitivity of human breast cancer cells mediated by adipose tissue-derived mesenchymal stromal cells. BMC Cancer 2013, 13, 535. [Google Scholar] [CrossRef] [PubMed]
  128. Zhou, X.; Li, T.; Chen, Y.; Zhang, N.; Wang, P.; Liang, Y.; Long, M.; Liu, H.; Mao, J.; Liu, Q.; et al. Mesenchymal stem cell-derived extracellular vesicles promote the in vitro proliferation and migration of breast cancer cells through the activation of the ERK pathway. Int. J. Oncol. 2019, 54, 1843–1852. [Google Scholar] [CrossRef]
  129. Chan, T.S.; Hsu, C.C.; Pai, V.C.; Liao, W.Y.; Huang, S.S.; Tan, K.T.; Yen, C.J.; Hsu, S.C.; Chen, W.Y.; Shan, Y.S.; et al. Metronomic chemotherapy prevents therapy-induced stromal activation and induction of tumor-initiating cells. J. Exp. Med. 2016, 213, 2967–2988. [Google Scholar] [CrossRef]
  130. Wilcken, N.; Zdenkowski, N.; White, M.; Snyder, R.; Pittman, K.; Mainwaring, P.; Green, M.; Francis, P.; De Boer, R.; Colosimo, M.; et al. Systemic treatment of HER2-positive metastatic breast cancer: A systematic review. Asia Pac. J. Clin. Oncol. 2014, 10 (Suppl. S4), 1–14. [Google Scholar] [CrossRef]
  131. Jia, Y.; Chen, Y.; Wang, Q.; Jayasinghe, U.; Luo, X.; Wei, Q.; Wang, J.; Xiong, H.; Chen, C.; Xu, B.; et al. Exosome: Emerging biomarker in breast cancer. Oncotarget 2017, 8, 41717–41733. [Google Scholar] [CrossRef]
  132. Vader, P.; Breakefield, X.O.; Wood, M.J. Extracellular vesicles: Emerging targets for cancer therapy. Trends Mol. Med. 2014, 20, 385–393. [Google Scholar] [CrossRef] [PubMed]
  133. Kessenbrock, K.; Plaks, V.; Werb, Z. Matrix metalloproteinases: Regulators of the tumor microenvironment. Cell 2010, 141, 52–67. [Google Scholar] [CrossRef] [PubMed]
  134. Garcia-Heredia, J.M.; Otero-Albiol, D.; Perez, M.; Perez-Castejon, E.; Munoz-Galvan, S.; Carnero, A. Breast tumor cells promotes the horizontal propagation of EMT, stemness, and metastasis by transferring the MAP17 protein between subsets of neoplastic cells. Oncogenesis 2020, 9, 96. [Google Scholar] [CrossRef] [PubMed]
  135. Yang, S.S.; Ma, S.; Dou, H.; Liu, F.; Zhang, S.Y.; Jiang, C.; Xiao, M.; Huang, Y.X. Breast cancer-derived exosomes regulate cell invasion and metastasis in breast cancer via miR-146a to activate cancer associated fibroblasts in tumor microenvironment. Exp. Cell Res. 2020, 391, 111983. [Google Scholar] [CrossRef] [PubMed]
  136. Lu, Y.; Chen, L.; Li, L.; Cao, Y. Exosomes Derived from Brain Metastatic Breast Cancer Cells Destroy the Blood-Brain Barrier by Carrying lncRNA GS1-600G8. 5. Biomed. Res. Int. 2020, 2020, 7461727. [Google Scholar]
  137. Singh, T.; Kaushik, M.; Mishra, L.C.; Behl, C.; Singh, V.; Tuli, H.S. Exosomal miRNAs as novel avenues for breast cancer treatment. Front. Genet. 2023, 14, 1134779. [Google Scholar] [CrossRef] [PubMed]
  138. Weaver, J.W.; Zhang, J.; Rojas, J.; Musich, P.R.; Yao, Z.; Jiang, Y. The application of exosomes in the treatment of triple-negative breast cancer. Front. Mol. Biosci. 2022, 9, 1022725. [Google Scholar] [CrossRef]
  139. Gao, Z.; Han, X.; Zhu, Y.; Zhang, H.; Tian, R.; Wang, Z.; Cui, Y.; Wang, Z.; Niu, R.; Zhang, F. Drug-resistant cancer cell-derived exosomal EphA2 promotes breast cancer metastasis via the EphA2-Ephrin A1 reverse signaling. Cell Death Dis. 2021, 12, 414. [Google Scholar] [CrossRef]
  140. Payton, C.; Pang, L.Y.; Gray, M.; Argyle, D.J. Exosomes Derived from Radioresistant Breast Cancer Cells Promote Therapeutic Resistance in Naive Recipient Cells. J. Pers. Med. 2021, 11, 1310. [Google Scholar] [CrossRef]
  141. Zhao, Y.; Jin, L.J.; Zhang, X.Y. Exosomal miRNA-205 promotes breast cancer chemoresistance and tumorigenesis through E2F1. Aging 2021, 13, 18498–18514. [Google Scholar] [CrossRef] [PubMed]
  142. Liu, J.; Zhu, S.; Tang, W.; Huang, Q.; Mei, Y.; Yang, H. Correction to: Exosomes from tamoxifen-resistant breast cancer cells transmit drug resistance partly by delivering miR-9-5p. Cancer Cell Int. 2021, 21, 673. [Google Scholar] [CrossRef] [PubMed]
  143. Qian, X.; Qu, H.; Zhang, F.; Peng, S.; Dou, D.; Yang, Y.; Ding, Y.; Xie, M.; Dong, H.; Liao, Y.; et al. Exosomal long noncoding RNA AGAP2-AS1 regulates trastuzumab resistance via inducing autophagy in breast cancer. Am. J. Cancer Res. 2021, 11, 1962–1981. [Google Scholar] [PubMed]
  144. Li, T.; Tao, Z.; Zhu, Y.; Liu, X.; Wang, L.; Du, Y.; Cao, J.; Wang, B.; Zhang, J.; Hu, X. Exosomal annexin A6 induces gemcitabine resistance by inhibiting ubiquitination and degradation of EGFR in triple-negative breast cancer. Cell Death Dis. 2021, 12, 684. [Google Scholar] [CrossRef] [PubMed]
  145. Cocco, S.; Leone, A.; Piezzo, M.; Caputo, R.; Di Lauro, V.; Di Rella, F.; Fusco, G.; Capozzi, M.; Gioia, G.D.; Budillon, A.; et al. Targeting Autophagy in Breast Cancer. Int. J. Mol. Sci. 2020, 21, 7836. [Google Scholar] [CrossRef]
  146. Han, M.; Hu, J.; Lu, P.; Cao, H.; Yu, C.; Li, X.; Qian, X.; Yang, X.; Yang, Y.; Han, N.; et al. Exosome-transmitted miR-567 reverses trastuzumab resistance by inhibiting ATG5 in breast cancer. Cell Death Dis. 2020, 11, 43. [Google Scholar] [CrossRef] [PubMed]
  147. Schedin, P.; Keely, P.J. Mammary gland ECM remodeling, stiffness, and mechanosignaling in normal development and tumor progression. Cold Spring Harb. Perspect. Biol. 2011, 3, a003228. [Google Scholar] [CrossRef] [PubMed]
  148. Liu, C.; Li, M.; Dong, Z.X.; Jiang, D.; Li, X.; Lin, S.; Chen, D.; Zou, X.; Zhang, X.D.; Luker, G.D. Heterogeneous microenvironmental stiffness regulates pro-metastatic functions of breast cancer cells. Acta Biomater. 2021, 131, 326–340. [Google Scholar] [CrossRef]
  149. Sampayo, R.G.; Toscani, A.M.; Rubashkin, M.G.; Thi, K.; Masullo, L.A.; Violi, I.L.; Lakins, J.N.; Caceres, A.; Hines, W.C. Coluccio Leskow, et al., Fibronectin rescues estrogen receptor alpha from lysosomal degradation in breast cancer cells. J. Cell Biol. 2018, 217, 2777–2798. [Google Scholar] [CrossRef] [PubMed]
  150. Roswall, P.; Bocci, M.; Bartoschek, M.; Li, H.; Kristiansen, G.; Jansson, S.; Lehn, S.; Sjolund, J.; Reid, S.; Larsson, C.; et al. Microenvironmental control of breast cancer subtype elicited through paracrine platelet-derived growth factor-CC signaling. Nat. Med. 2018, 24, 463–473. [Google Scholar] [CrossRef]
  151. Zarrilli, G.; Businello, G.; Dieci, M.V.; Paccagnella, S.; Carraro, V.; Cappellesso, R.; Miglietta, F.; Griguolo, G.; Guarneri, V.; Mele, M.L.; et al. The Tumor Microenvironment of Primitive and Metastatic Breast Cancer: Implications for Novel Therapeutic Strategies. Int. J. Mol. Sci. 2020, 21, 8102. [Google Scholar] [CrossRef] [PubMed]
  152. Yu, Y.; Xiao, C.H.; Tan, L.D.; Wang, Q.S.; Li, X.Q.; Feng, Y.M. Cancer-associated fibroblasts induce epithelial-mesenchymal transition of breast cancer cells through paracrine TGF-beta signalling. Br. J. Cancer 2014, 110, 724–732. [Google Scholar] [CrossRef]
  153. Kawase, A.; Ishii, G.; Nagai, K.; Ito, T.; Nagano, T.; Murata, Y.; Hishida, T.; Nishimura, M.; Yoshida, J.; Suzuki, K.; et al. Podoplanin expression by cancer associated fibroblasts predicts poor prognosis of lung adenocarcinoma. Int. J. Cancer 2008, 123, 1053–1059. [Google Scholar] [CrossRef] [PubMed]
  154. Fiori, M.E.; Di Franco, S.; Villanova, L.; Bianca, P.; Stassi, G.; De Maria, R. Cancer-associated fibroblasts as abettors of tumor progression at the crossroads of EMT and therapy resistance. Mol. Cancer 2019, 18, 70. [Google Scholar] [CrossRef] [PubMed]
  155. Chen, X.; Song, E. Turning foes to friends: Targeting cancer-associated fibroblasts. Nat. Rev. Drug Discov. 2019, 18, 99–115. [Google Scholar] [CrossRef] [PubMed]
  156. Hu, J.L.; Wang, W.; Lan, X.L.; Zeng, Z.C.; Liang, Y.S.; Yan, Y.R.; Song, F.Y.; Wang, F.F.; Zhu, X.H.; Liao, W.J.; et al. CAFs secreted exosomes promote metastasis and chemotherapy resistance by enhancing cell stemness and epithelial-mesenchymal transition in colorectal cancer. Mol. Cancer 2019, 18, 91. [Google Scholar] [CrossRef]
  157. De Kruijf, E.M.; van Nes, J.G.; van de Velde, C.J.; Putter, H.; Smit, V.T.; Liefers, G.J.; Kuppen, P.J.; Tollenaar, R.A.; Mesker, W.E. Tumor-stroma ratio in the primary tumor is a prognostic factor in early breast cancer patients, especially in triple-negative carcinoma patients. Breast Cancer Res. Treat. 2011, 125, 687–696. [Google Scholar] [CrossRef] [PubMed]
  158. McCuaig, R.; Wu, F.; Dunn, J.; Rao, S.; Dahlstrom, J.E. The biological and clinical significance of stromal-epithelial interactions in breast cancer. Pathology 2017, 49, 133–140. [Google Scholar] [CrossRef]
  159. Kothari, C.; Diorio, C.; Durocher, F. The Importance of Breast Adipose Tissue in Breast Cancer. Int. J. Mol. Sci. 2020, 21, 5760. [Google Scholar] [CrossRef]
  160. Wang, S.; Su, X.; Xu, M.; Xiao, X.; Li, X.; Li, H.; Keating, A.; Zhao, R.C. Exosomes secreted by mesenchymal stromal/stem cell-derived adipocytes promote breast cancer cell growth via activation of Hippo signaling pathway. Stem Cell Res. Ther. 2019, 10, 117. [Google Scholar] [CrossRef] [PubMed]
  161. Yang, D.; Zhang, W.; Zhang, H.; Zhang, F.; Chen, L.; Ma, L.; Larcher, L.M.; Chen, S.; Liu, N.; Zhao, Q.; et al. Progress, opportunity, and perspective on exosome isolation—Efforts for efficient exosome-based theranostics. Theranostics 2020, 10, 3684–3707. [Google Scholar] [CrossRef]
  162. He, C.; Zheng, S.; Luo, Y.; Wang, B. Exosome Theranostics: Biology and Translational Medicine. Theranostics 2018, 8, 237–255. [Google Scholar] [CrossRef] [PubMed]
  163. Zhao, L.; Gu, C.; Gan, Y.; Shao, L.; Chen, H.; Zhu, H. Exosome-mediated siRNA delivery to suppress postoperative breast cancer metastasis. J. Control. Release 2020, 318, 1–15. [Google Scholar] [CrossRef]
  164. Wang, K.; Ye, H.; Zhang, X.; Wang, X.; Yang, B.; Luo, C.; Zhao, Z.; Zhao, J.; Lu, Q.; Zhang, H.; et al. An exosome-like programmable-bioactivating paclitaxel prodrug nanoplatform for enhanced breast cancer metastasis inhibition. Biomaterials 2020, 257, 120224. [Google Scholar] [CrossRef] [PubMed]
  165. Luo, T.; Liu, Q.; Tan, A.; Duan, L.; Jia, Y.; Nong, L.; Tang, J.; Zhou, W.; Xie, W.; Lu, Y.; et al. Mesenchymal Stem Cell-Secreted Exosome Promotes Chemoresistance in Breast Cancer via Enhancing miR-21-5p-Mediated S100A6 Expression. Mol. Ther. Oncolytics 2020, 19, 283–293. [Google Scholar] [CrossRef] [PubMed]
  166. Del Re, M.; Bertolini, I.; Crucitta, S.; Fontanelli, L.; Rofi, E.; De Angelis, C.; Diodati, L.; Cavallero, D.; Gianfilippo, G.; Salvadori, B.; et al. Overexpression of TK1 and CDK9 in plasma-derived exosomes is associated with clinical resistance to CDK4/6 inhibitors in metastatic breast cancer patients. Breast Cancer Res. Treat. 2019, 178, 57–62. [Google Scholar] [CrossRef] [PubMed]
  167. Wang, J.; Yeung, B.Z.; Wientjes, M.G.; Cui, M.; Peer, C.J.; Lu, Z.; Figg, W.D.; Woo, S.; Au, J.L. A Quantitative Pharmacology Model of Exosome-Mediated Drug Efflux and Perturbation-Induced Synergy. Pharmaceutics 2021, 13, 997. [Google Scholar] [CrossRef]
  168. Augimeri, G.; La Camera, G.; Gelsomino, L.; Giordano, C.; Panza, S.; Sisci, D.; Morelli, C.; Gyorffy, B.; Bonofiglio, D.; Ando, S.; et al. Evidence for Enhanced Exosome Production in Aromatase Inhibitor-Resistant Breast Cancer Cells. Int. J. Mol. Sci. 2020, 21, 5841. [Google Scholar] [CrossRef]
  169. Liu, Q.W.; Chen, Y.; Li, J.Y.; Xiao, L.; Zhang, W.J.; Zhao, J.L.; Gu, H.C.; Wu, H.Y.; Zuo, G.S.; Deng, K.Y.; et al. Bone marrow cells are differentiated into MDSCs by BCC-Ex through down-regulating the expression of CXCR4 and activating STAT3 signalling pathway. J. Cell. Mol. Med. 2021, 25, 5497–5510. [Google Scholar] [CrossRef] [PubMed]
  170. Biswas, S.; Mandal, G.; Chowdhury, S.R.; Purohit, S.; Payne, K.K.; Anadon, C.; Gupta, A.; Swanson, P.; Yu, X.; Conejo-Garcia, J.R.; et al. Exosomes Produced by Mesenchymal Stem Cells Drive Differentiation of Myeloid Cells into Immunosuppressive M2-Polarized Macrophages in Breast Cancer. J. Immunol. 2019, 203, 3447–3460. [Google Scholar] [CrossRef] [PubMed]
  171. Jabbari, N.; Nawaz, M.; Rezaie, J. Ionizing Radiation Increases the Activity of Exosomal Secretory Pathway in MCF-7 Human Breast Cancer Cells: A Possible Way to Communicate Resistance against Radiotherapy. Int. J. Mol. Sci. 2019, 20, 3649. [Google Scholar] [CrossRef] [PubMed]
  172. Laot, L.; Laas, E.; Girard, N.; Dumas, E.; Daoud, E.; Grandal, B.; Pierga, J.Y.; Coussy, F.; Kirova, Y.; El-Alam, E.; et al. The Prognostic Value of Lymph Node Involvement after Neoadjuvant Chemotherapy Is Different among Breast Cancer Subtypes. Cancers 2021, 13, 171. [Google Scholar] [CrossRef]
  173. Dalenc, F.; Sarradin, V.; Nicolai, V.; Franchet, C.; Ung, M. Recent therapeutic trends in triple-negative metastatic breast cancers: PARP inhibitors, immunotherapies and antibody-drug conjugates. Bull. Cancer 2021, 108, 67–79. [Google Scholar] [CrossRef]
  174. Waks, A.G.; Winer, E.P. Breast Cancer Treatment: A Review. JAMA 2019, 321, 288–300. [Google Scholar] [CrossRef]
  175. Lal, P.; Tan, L.K.; Chen, B. Correlation of HER-2 status with estrogen and progesterone receptors and histologic features in 3655 invasive breast carcinomas. Am. J. Clin. Pathol. 2005, 123, 541–546. [Google Scholar] [CrossRef] [PubMed]
  176. Dieci, M.V.; Guarneri, V. Should triple-positive breast cancer be recognized as a distinct subtype? Expert Rev. Anticancer Ther. 2020, 20, 1011–1014. [Google Scholar] [CrossRef] [PubMed]
  177. Alataki, A.; Dowsett, M. Human epidermal growth factor receptor-2 and endocrine resistance in hormone-dependent breast cancer. Endocr. Relat. Cancer 2022, 29, R105–R122. [Google Scholar] [CrossRef] [PubMed]
Table 1. Clinical trial targeting both HER2 and ER for TPBC.
Table 1. Clinical trial targeting both HER2 and ER for TPBC.
NCTSubjectsTreatmentPrimary Endpoint
02003209Terminal TPBCdocetaxel, carboplatin, trastuzumab, and pertuzumab (TCHP) with or without estrogen deprivation (goserelin plus an AI)N
01817452Early TPBCconjugate ado-trastuzumab emtansine (T-DM1) versus trastuzumab with ETY
02530424Early TPBCCDK4/6 inhibitor palbociclib with trastuzumab, pertuzumab, and fulvestrantY
03272477Early TPBCET versus paclitaxel 80 mg/m2 weekly plus trastuzumab and pertuzumabY
00629278Early TPBCAIs vs. tamoxifen or tamoxifenY
00878709Early TPBCneratinib plus trastuzumabY
00022672Terminal TPBCanastrozole plus trastuzumabY
00073528Terminal TPBCletrozole plus lapatinibY
01160211Terminal TPBCprior trastuzumab and ETY
Abbreviations: N, not met; Y, yes.
Table 2. Clinical significance and involved mechanisms of tumor stromal cell and non-cellular components in TPBC.
Table 2. Clinical significance and involved mechanisms of tumor stromal cell and non-cellular components in TPBC.
ItemsClinical SignificanceInvolved MechanismsReferences
CAFsproliferation and
metastasis; tamoxifen resistance; Herceptin resistance
EGFR/ERK/PKC signal; miR-500a-5p/USP28; JAK/STAT3 or PI3K/AKT pathways; GPER/EGFR/ERK signaling and E2 production; PI3K/AKT and MAPK/ERK1/2 pathways IL-6/STAT–3/NF-κB[39,46,47,48,49,50,51]
TAMsimmune escape and lung metastasis; paclitaxel resistance; tamoxifen resistanceTAM/CXCL1/NF-κB/FOXP3; IL-10/STAT3/bcl-2 signaling pathway; PI3K/AKT/mTOR; NF-κB/STAT3/ERK; EGFR/PI3K/Akt[67,71,72,73,74]
CAAscellular proliferation; promotes EMT; inhibition of adhesion, migration and invasion; endocrine therapyestrogen receptor (ER) signaling; EGFR/JAK2/HER2; JAK/STAT3 PI3K/AKT; PKM2/PI3K/AKT; LKB1-AMPK-S6K; FGF1/FGFR phosphorylation[94,95,96,97,98,99,100,109,118]
ASCsinhibit proliferation; increase breast ball formation, cell fusion, and cell migrationSDF-1α/CXCR4[127]
MSCspromote BCSCs self-renewal and cancer cell proliferation; inhibit migrationCCL5/TGF-β/EMT; IL-6 and CXCL7; mTOR/HIF-1α/VEGF[122,123,125]
TDEstamoxifen resistance; trastuzumab resistancemiRNA205-Caspase signaling pathway; lncRNA AGAP2-AS1-ATG10-autophagy reaction; miR-567-autophagy reaction[141,143,146]
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Pu, Q.; Gao, H. The Role of the Tumor Microenvironment in Triple-Positive Breast Cancer Progression and Therapeutic Resistance. Cancers 2023, 15, 5493. https://doi.org/10.3390/cancers15225493

AMA Style

Pu Q, Gao H. The Role of the Tumor Microenvironment in Triple-Positive Breast Cancer Progression and Therapeutic Resistance. Cancers. 2023; 15(22):5493. https://doi.org/10.3390/cancers15225493

Chicago/Turabian Style

Pu, Qian, and Haidong Gao. 2023. "The Role of the Tumor Microenvironment in Triple-Positive Breast Cancer Progression and Therapeutic Resistance" Cancers 15, no. 22: 5493. https://doi.org/10.3390/cancers15225493

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

Article Metrics

Back to TopTop