Next Article in Journal
Galectin-1 Expression Is Associated with the Response and Survival Following Preoperative Chemoradiotherapy in Locally Advanced Esophageal Squamous Cell Carcinoma
Next Article in Special Issue
Hampering Stromal Cells in the Tumor Microenvironment as a Therapeutic Strategy to Destem Cancer Stem Cells
Previous Article in Journal
Reporting of Incidence and Outcome of Bone Metastases in Clinical Trials Enrolling Patients with Epidermal Growth Factor Receptor Mutated Lung Adenocarcinoma—A Systematic Review
Previous Article in Special Issue
The Cellular and Biological Impact of Extracellular Vesicles in Pancreatic Cancer
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Cancer-Associated Fibroblasts in Breast Cancer Treatment Response and Metastasis

by
Patricia Fernández-Nogueira
1,2,*,
Gemma Fuster
1,3,4,
Álvaro Gutierrez-Uzquiza
5,6,
Pere Gascón
1,
Neus Carbó
1 and
Paloma Bragado
5,6,*
1
Department of Biochemistry and Molecular Biomedicine, Institute of Biomedicine, University of Barcelona (IBUB), 08028 Barcelona, Spain
2
Department of Biomedicine, School of Medicine, University of Barcelona, 08028 Barcelona, Spain
3
Department of Biochemistry & Physiology, School of Pharmacy and Food Sciences, University of Barcelona, 08028 Barcelona, Spain
4
Department of Biosciences, Faculty of Sciences and Technology, University of Vic, 08500 Vic, Spain
5
Department of Biochemistry and Molecular Biology, Faculty of Pharmacy, Complutense University of Madrid, 28040 Madrid, Spain
6
Health Research Institute of the Hospital Clínico San Carlos, 28040 Madrid, Spain
*
Authors to whom correspondence should be addressed.
Cancers 2021, 13(13), 3146; https://doi.org/10.3390/cancers13133146
Submission received: 30 April 2021 / Revised: 15 June 2021 / Accepted: 16 June 2021 / Published: 23 June 2021

Abstract

:

Simple Summary

Breast cancer is a major public health problem with a large impact on the life of patients and their families. It is a highly curable disease when detected early, and an inevitably mortal disease when discovered too late. Therapy resistance and metastases are the most critical clinical issues faced by breast cancer oncologists nowadays. It has become evident that interactions between carcinoma cells and tumor microenvironment are an essential part of tumor growth and progression. Cells that support the function of epithelial cells, like cancer-associated fibroblasts (CAFs), contribute to therapy resistance and metastasis via the production of several secreted factors and direct interaction with cancer cells. Here we review the role of CAFs in radiotherapy, chemotherapy, endocrine and targeted therapies resistance. We also highlight the role of CAFs and fibroblasts from metastatic sites in metastasis progression. Finally, we discuss advances and potential therapeutic strategies to target CAFs for overcoming resistance and preventing metastases.

Abstract

Breast cancer (BrCa) is the leading cause of death among women worldwide, with about one million new cases diagnosed each year. In spite of the improvements in diagnosis, early detection and treatment, there is still a high incidence of mortality and failure to respond to current therapies. With the use of several well-established biomarkers, such as hormone receptors and human epidermal growth factor receptor-2 (HER2), as well as genetic analysis, BrCa patients can be categorized into multiple subgroups: Luminal A, Luminal B, HER2-enriched, and Basal-like, with specific treatment strategies. Although chemotherapy and targeted therapies have greatly improved the survival of patients with BrCa, there is still a large number of patients who relapse or who fail to respond. The role of the tumor microenvironment in BrCa progression is becoming increasingly understood. Cancer-associated fibroblasts (CAFs) are the principal population of stromal cells in breast tumors. In this review, we discuss the current understanding of CAFs’ role in altering the tumor response to therapeutic agents as well as in fostering metastasis in BrCa. In addition, we also review the available CAFs-directed molecular therapies and their potential implications for BrCa management.

1. Introduction

Breast cancer (BrCa) is the most commonly diagnosed cancer among females and the first leading cause of cancer death in women [1]. Despite the fact that the advancements in therapies and early detection have reduced the mortality rate for BrCa, advanced metastatic BrCa is still considered incurable. BrCa is a heterogeneous and complex disease. In 2000, Perou and Sorlie reported the molecular intrinsic classification of BrCa that distinguished four BrCa subtypes: Luminal A and Luminal B (both expressing the estrogen receptor (ER), Basal-like and HER2-enriched [2,3]. This classification completely altered BrCa clinical management. The main therapeutic approaches to manage BrCa are surgery, chemotherapy, radiotherapy, targeted therapy, and immunotherapy; however, all of them eventually fail, particularly in patients with advanced metastatic disease. Luminal BrCas are treated with endocrine therapy while HER2-enriched BrCas are treated with HER2-targeted therapies such as trastuzumab or pertuzumab. While chemotherapy is still the main treatment for TNBrCa patients, recently, two targeted therapies: olaparib and talazoparib, both Poly(ADP-Ribose) polymerase 1 (PARP) inhibitors, have been approved by the FDA for the management of TNBrCa with germline BRCA mutations [4,5].
Over the past few decades, it has become evident that the complexity of cancer is not only dependent on the intrinsic characteristics of tumor cells but also mainly determined by the crosstalk between tumor cells and various components of the tumor microenvironment (TME). The TME is an ecosystem composed of many cell types (e.g., cancer cells, fibroblasts, immune cells, endothelial cells) located in a complex physicochemical environment. Understanding how this complex ecosystem is organized is critical to further comprehend specific cell population roles in cancer progression from carcinoma in situ to invasive therapy responses and metastasis [6].
Among all components of the TME, fibroblasts are the most abundant cell type, playing an active role in the tumor mass. Fibroblasts are in constant communication with cancer cells, either via direct cell–cell contact or through the secretion of soluble factors able to activate multiple signaling pathways in tumor cells. Unlike normal fibroblasts, which dynamically remodel the ECM, control normal tissue homeostasis and participate in wound healing and senescence, cancer-associated fibroblasts (CAFs) can facilitate tumorigenesis. In particular, BrCa CAFs can represent up to 80% of the tumor mass and are active players in breast tumor initiation and progression [7,8,9,10]. The absence of specific CAFs’ molecular markers has complicated their identification and data comparison between studies. Recently, the analysis of six fibroblast markers: fibroblast activated protein (FAP), integrin β1/CD29, αsmooth muscle actin (αSMA), fibroblast surface protein (FSP1), platelet derived growth factor receptor β (PDGFRβ), and caveolin 1 (CAV1) has allowed the identification of four BrCa CAFs’ subsets that accumulate differentially in normal tissue and in BrCa subtypes, exerting different roles in tumor immunobiology and metastasis [11].
CAFs play multiple versatile functions in cancer, including extracellular matrix remodeling, maintenance of stemness, blood vessel formation and promotion of cancer cell proliferation, migration and invasion, all of which leads to therapy resistance and metastasis formation [12,13]. In recent years, several reports have shown that CAFs play also a critical role in reprogramming the metabolic landscape of tumors [14]. Furthermore, CAFs can also regulate the neighboring immune cells contributing to immune escape of tumors via multiple mechanisms, including secretion of multiple cytokines and chemokines as well as recruitment and modulation of tumor-infiltrating immune cells [15]. The crosstalk between CAFs and cancer cells is mediated by various intracellular and extracellular factors which could potentially be targeted for anticancer therapy.
Improving patient treatment requires a better understanding of the mechanisms leading to resistance to current therapies and metastasis, as well as a deeper comprehension of the role played by the stroma-tumor cells’ crosstalk in these processes. In this review, we focus specifically on the role of CAFs on BrCa progression from ductal carcinoma in situ (DCIS) to invasive carcinoma, therapy resistance and metastasis as well as the strategies and challenges associated with therapeutic regimens targeting the stroma.

2. Role of Fibroblasts on Ductal Carcinoma In Situ Progression

DCIS is the most common non-invasive, pre-malignant BrCa lesion and represents up to 25% of all newly diagnosed BrCa patients [16]. DCIS lesions are highly heterogeneous, and each DCIS has its own probability of progressing to invasive ductal carcinoma (IDC), and eventually undergo metastasis [17]. Deciphering the molecular and cellular processes that are occurring in DCIS transition to IDC will shed light on DCIS biology and help identify tumor and stromal biomarkers, able to predict the IDC risk.
The DCIS TME shows a high variability in composition and collagen amounts and in the topological distribution of stromal cells, including fibroblasts [18,19,20,21]. In fact, high collagen deposition and high mammographic density (HMD) are closely related to increased risk of BrCa [22]. The transcriptional profile of fibroblasts present in HMD samples indicated that they are prone to over-activate stress response, inflammation, stemness and C-Jun N-terminal kinase 1(JNK1) pathway and expression patterns similar to those of CAFs in BrCa [23]. 3D co-cultures of fibroblasts and MCF10DCIS.com, a mild aggressive BrCa cell line with low invasive characteristics, have revealed that fibroblasts promote DCIS invasiveness by increasing matrix metalloprotease 14 (MMP14) and MMP9 levels, changing collagen organization and rising BrCa cell proliferation rate [24,25,26]. Moreover, co-injection of MCF10DCIS.com with either normal fibroblasts, CAFs or rheumatoid arthritis fibroblasts resulted in an increased ability to invade the adjacent stroma, suggesting that fibroblasts can facilitate tumor cell invasion, even in the presence of the tumor-suppressing myoepithelial cells [24].
In Basal-like DCIS, the invasive potential seems to rely on the ability of fibroblasts to modify the orientation of collagen fibers [27], with an increased and perpendicular alignment of collagen near DCIS invasive lesions [28,29] compared to the normal collagen organization, parallel to the duct perimeter. HER2-enriched DCIS has also been reported to display perpendicular collagen orientation [28]. However, recurrence risk in DCIS was not associated to the collagen distribution itself, but to the fiber width and density [30]. Interestingly, collagen radial distribution, typical of IDC, is also present in the points of invasiveness in the DCIS, along with collagen perpendicular disposition to the tumor. Therefore, it can be suggested that collagen organization can be informative of the prognosis of DCIS lesions.
Regarding soluble factors, interleukin 6 (IL6) production by CAFs promotes DCIS invasion through the induction of cathepsin B expression in tumor cells [26]. Moreover, it has been demonstrated that primary tumor CAFs enhance invasion of tumor cells, via CAF chemokine (C-X-C motif) ligand 1 (CXCL1) secretion and interaction with C-X-C motif chemokine receptor 2 (CXCR2) in the tumor cells [31]. In DCIS human samples with invasive component, the presence of myofibroblasts positive for urokinase/plaminogen activator (uPA), plasminogen Activator/Urokinase Receptor (uPAR), MMP13, and/or procoagulant factors, such as tissue factor (TF), thrombin and proteinase-activated receptor 2 (PAR2), is related to progression to IDC [32,33,34]. Furthermore, the combination of high expression of Fibroblast activated protein alpha (FAP-α) in fibroblasts and Golgi phosphoprotein 3 (GOLPH3) in BrCa cells can be considered as predictive for DCIS recurrence and progression to IDC [35]. Additionally, CAV1 loss of expression in DCIS fibroblasts has been suggested to predict risk progression to IDC [36], although more studies are needed to validate these results. Therefore, CAFs can facilitate DCIS progression to IDC (Figure 1 and Table S1).

3. Role of CAFs in BrCa Therapy Resistance

3.1. Radiotherapy Resistance and CAFs

Radiation therapy is recommended for most people who have lumpectomy to remove BrCa. The goal is to destroy any residual cancer cells that may have remained in the breast after tumor removal and reduce the risk of recurrence [37]. However, radiotherapy (RT) usually results in fibrosis, which may lead to survival and RT resistance of cancer cells [38,39] (Figure 1 and Table S3).
In the BrCa context, Steer et al. have demonstrated, in 3D co-cultures and in vivo, the protective role of fibroblasts on irradiated cancer cells and tumors [40]. Transforming growth factor β (TGFβ) signaling was found upregulated in the mammary gland after RT, being the main inductor of the stromal compartment activation in in vivo models [41]. Bouquet et al. found that TGFβ inhibition sensitizes BrCa cells to RT [42]. Moreover, different studies have shown that the combination of TGFβ inhibition and RT blocks tumor growth, preventing the increase in circulating TGFβ levels induced by RT, and consequently metastasis, in preclinical studies [43].
Other growth factors have been involved in RT resistance. Hepatic growth factor (HGF) secreted by CAFs activates MET proto-oncogene, receptor tyrosine kinase (MET) receptor in BrCa cells. De Bacco et al. also found that MET inhibition promotes RT response, reducing BrCa cell invasion prompted by CAFs [44]. Furthermore, after ionizing, radiation fibroblasts upregulated β1 integrin expression, together with AKT serine/threonine kinase (AKT) pathway activation, leading to RT protection in vivo [45]. Boelens et al. further showed that the crosstalk between CAFs and BrCa cells drives chemo- and radiotherapy resistance through an exosome transfer from the stromal population. As a result, Notch receptor 3 (NOTCH3) and Signal transducer and activator of transcription-1 (STAT1)-antiviral pathways were activated, inducing a RT-resistant stem cell-like phenotype [46]. Besides, radiation induces senescence on irradiated fibroblasts, which produce IL6, CXCL8/IL8 and osteopontin (OPN), all pro-tumorigenic factors related with stromal-mediated RT resistance [47]. Senescent CAFs also improved BrCa cells’ proliferation in an AKT pathway-dependent manner, which also conferred RT resistance [48]. On the basis of these observations, it could be argued that targeting CAFs during RT could improve BrCa RT response (Figure 1 and Table S3).

3.2. CAFs and Chemotherapy Resistance in Triple Negative BrCa

TNBrCa or basal subtype represents around 15% of BrCa lesions diagnosed and is characterized by the expression of basal-related genes and the absence or low levels of ER, PR and, HER2 [2]. This BrCa subtype encompasses a wide and heterogeneous group of lesions and is generally associated with a poor prognosis due to the lack of appropriate targeted therapies [49]. Therefore, the main clinical challenge in TNBrCa is to design new therapies to reduce metastasis and improve patient’s quality of life and survival.
Concerning CAFs contribution to therapy-resistance, a 6-CAFs’ genes signature (CXCL2/MIP2 and its human homologous CXCL8/IL8, MMP1, Retinoic Acid Receptor Responder 1 (RARRES1), fibroblast growth factor 1 (FGF1), and CXCR7) has been related to taxotere treatment resistance in an in vitro model using TNBrCa cells [50]. In addition, EDALINE phase I clinical trial results suggest that blocking hedgehog (Hh) stimuli on CAFs, by smoothened inhibitors (SMOi) treatment, sensitizes TNBrCa to docetaxel and leads to survival amelioration [51]. Moreover, inhibition of TGFβ production by CAFs with the anti-fibrotic drug pirfenidone reduced BrCa tumor growth and also lung metastasis when combined with doxorubicin, acting in a synergistic way [52]. Combination of a hydrogel assembled to losartan, an inhibitor of angiotensin-II that blocks the TGFβ pathway, sensitizes TNBrCa cell lines to doxorubicin-loaded liposomes facilitating the reduction of chemotherapy resistance [53].
On the other hand, under chemotherapy pressure, interferon β1 (IFNβ1) secretion by cancer cells stimulates the transcription of pro-inflammatory cytokine genes in stromal fibroblasts, by which CAFs acquire an anti-viral state, essential for the recovery and resistance acquisition of BrCa cells after chemotherapy. Indeed, high expression of tumor IFNβ1 in TNBrCa patients correlated with an aggressiveness signature [54]. Another interesting paracrine relationship between cancer cells expressing platelet derived growth factor (PDGF) and CAFs positive for PDGF-receptors was described in basal-like BrCa patients. PDGF abrogation in the tumor produces a phenotype shift into a positive hormone-receptor subtype, sensitive to endocrine therapy, leading to a more therapeutically affordable BrCa subtype. These results also suggest that BrCa subtype control and plasticity can be exerted by fibroblasts and cancer cells interactions [55].
Furthermore, the interaction between cancer cells, immune cells, mesenchymal stem cells (MSC), and CAFs led to the release of CXCL1, CXCL2/MIP (homologous to CXCL8/IL8 in humans) (from TNBrCa cells), CXCL16 (from CAFs), and C-C motif chemokine ligand 5 (CCL5) (from CAFs and MSCs), resulting in a more reactive stroma, inducing pro-tumoral actions, including chemotherapy resistance [56,57,58,59,60,61]. This pro-resistance context attracts tumor-associated macrophages and stimulates the endothelial cell population through the release of CXCL8/IL8 and CCL5 by TNBrCa cells. All these results suggested that therapeutically targeting CXCL8/IL8 and CCL5 could be useful in TNBrCa patient’s treatment [62,63]. Expression of CXCL14 or Programmed death-ligand 1 (PD-L1) by stromal CAFs has also been associated with shorter or better survival, respectively, in TNBrCa patients [64,65], and could be considered as independent prognosis markers in TNBrCa patients. Therefore, these data emphasize that there is a strong chemotherapy-resistance dependence of tumor-stromal interactions in TNBrCa, particularly evident between fibroblasts and cancer cells (Figure 1 and Table S4), highlighting the importance of targeting stroma and tumor cell interactions as a promising clinical therapeutic strategy in preventing drug resistance in TNBrCa patients [66].

3.3. CAFs and Hormonal Therapy Resistance in Luminal BrCa

Luminal BrCa represents 70% of all BrCas and expresses hormonal receptors, hence it is considered sensitive to endocrine therapy, especially ER-targeted [67]. The prognosis for early luminal BrCa patients is particularly good, however, up to 50% of patients relapse and die from metastatic disease [67]. The treatment of patients with ER-positive BrCa is based on ER modulators such as tamoxifen and/or aromatase inhibitors. Tamoxifen has become the drug of first choice in patients with luminal BrCa. Nevertheless, it is estimated that approximately 45% of women do not respond to tamoxifen (de novo resistance) [68], whereas acquired resistance to the drug develops ultimately in all tamoxifen-receiving patients. Therefore, although tamoxifen was hailed as a major breakthrough in the management of patients with hormone-dependent BrCa, development of resistance to the treatment poses a serious clinical problem [69].
Endocrine-therapy resistance can likewise be linked to the biological action of luminal BrCa CAFs. They express a G protein-coupled estrogen receptor 1 (GPER), which suggest that they are also sensitive to estrogens. CAFs expressing GPER can activate the epidermal growth factor receptor (EGFR) and extracellular signal-regulated kinase 2 (ERK) signaling which leads to (i) estradiol production by CAFs and endocrine therapy resistance [70] and (ii) cancer cell integrin β1 activation by increased CAF-secreted fibronectin [71], which drives EMT and the tamoxifen-resistant phenotype. In turn, CAFs can also become resistant to hormonal therapy. BrCa-resistant cells secrete TGFβ1 that induces ERK activation on CAFs and promotes their resistance [72]. In addition, activation of phosphatidylinositol-4,5-bisphosphate 3-kinase (PI3K)/AKT in BrCa cells increases CAFs’ GPER translocation to the membrane and induces the activation of PKA/cAMP responsive element-binding protein (CREB) signaling, which activates the Warburg effect in CAFs. This increases pyruvate and lactate levels, metabolites used by cancer cells to fuel their mitochondrial metabolism and confer drug resistance to hormonal- and HER2-targeted therapies, as well as chemotherapy [73]. CAFs-mediated metabolic reprograming of BrCa cells to decrease sensitivity to endocrine therapy has been proposed by others. For instance, CAFs’ secretion of lactate and ketone bodies increases mitochondrial activity in BrCa cells and leads to tamoxifen resistance [74]. A recent report has shown that BrCa cells promote neighboring CAFs’ hypoxia, which leads to autophagic degradation of CAFs CAV1 and activation of the Warburg effect in them. These changes contribute to protect BrCa cells from endocrine therapy-induced apoptosis and autophagy, by providing nutrients to fuel BrCa cell metabolism [75]. In fact, BrCa patients whose stroma is negative for CAV1 expression develop resistance to tamoxifen and have worse prognosis [76].
Endocrine therapy resistance is also driven by CAFs’ secretion of growth factors. For instance, HGF secretion by CAFs leads to MET upregulation in ER-positive BrCa cell lines, which leads to increase migration, invasion, and resistance to fulvestrant through SRC proto-oncogene, non-receptor tyrosine kinase (SRC), AKT, and ERK1/2 activation [77]. FGF1 has also been shown to induce hormonal therapy resistance through activation of fibroblast growth factor receptor 3 (FGFR3) that causes PI3K/AKT and ERK1/2 induction by phospholipase C γ (PLCγ) [78]. Additionally, FGF7 can block tamoxifen effect through activation of FGFR2, which enhances ER degradation by the proteasome. FGF7/FGFR2 effect was mediated through activation of PI3K/AKT signaling and the upregulation of B-cell lymphoma 2 (Bcl-2) expression in BrCa cells [79].
Several other reports have linked CAFs-elicited resistance to tamoxifen with activation of PI3K/AKT and/or Ras/Raf/mitogen activated protein kinase kinase 1 (MEK1)/ERK1/2 [80,81,82]. Fibronectin, through interaction with Integrin β1, and soluble factors secreted by CAFs, activate EGFR and MMPs which leads to induction of PI3K/AKT and ERK1/2 in BrCa cells, protecting them from tamoxifen-induced cell death [83]. AKT and ERK1/2 hyperactivation stimulates ER phosphorylation which promotes tamoxifen resistance [80]. Moreover, fibroblasts expressing amyloid beta precursor protein binding family A, member 3 (MINT3) upregulate L1 cell adhesion molecule (L1CAM) which leads to ERK1/2 activation in BrCa cells through integrin α5β1, promoting tumor growth and resistance [81]. Stromal cells have also been shown to elicit hormonal therapy resistance through inhibition of insulin-like growth factor binding protein 5 (IGFBP5) which induces Bcl-3 upregulation and activation of NF-κB, driving resistance to fulvestrant [84].
CD146 is a cell surface marker that characterizes two different subtypes of fibroblasts in BrCa tumors. CAFs negative for CD146 foster the upregulation and activation of several tyrosine kinase receptors (TKRs), such as EGFR, HER2 and insulin-like growth factor receptor (IGF1R), and the inhibition of ER in BrCa cells which leads to tamoxifen resistance [85]. Other cell surface markers have also been linked to CAFs-mediated tamoxifen resistance. For example, a subpopulation of CAFs positive for CD63 has been shown to elicit tamoxifen resistance through secretion of exosomes enriched in miR-22, that activates ERα and phosphatase and tensin homolog (PTEN) promoting resistance. Furthermore, CD63 induces STAT3 activation to maintain the phenotype and function of CD63+ CAFs [86]. Other microRNAs have recently been associated with CAFs-mediated hormonal therapy resistance. It has recently been described that CAFs exosomes or microvesicles containing miRNA-221 can facilitate hormonal therapy resistance by promoting cancer stem cell (CSC) formation and ER downregulation, which leads to tumor growth and resistance to fulvestrant-induced apoptosis [87]. Altogether, these findings indicate that BrCa CAFs mediate resistance to endocrine therapy through several molecular mechanisms and mediators (Figure 1 and Table S4), each of them representing an intriguing target to be explored to re-sensitize BrCa cells to hormonal therapy.

3.4. CAFs- and HER2-Targeted Therapy Resistance in HER2-Enriched BrCa

Around 15% to 20% of all breast tumors belong to the HER2 positive (+) subtype, with an aggressive phenotype and worse clinical course [2,88]. HER2+ subtype is characterized by ERBB2 amplification, causing an activation of the HER2 pathway, initiating cell proliferation, promoting cell survival, and driving metastasis through different pathways such as the RAS and the PI3K–PKB(AKT)–MAPK pathways [1]. Four targeted drugs are already approved for treatment of these patients, being trastuzumab, pertuzumab, lapatinib, and ado-trastuzumab emtansine (TDM1), the commonly recommended anti-HER2 target agents with excellent therapeutic effects [89]. However, the development of resistance to these treatments has reduced their expectations in terms of overall clinical benefit in the metastatic setting, representing the principal impediment concerning better long-term patient survival [90,91,92,93].
As mentioned before, BrCa treatment is often unsuccessful due to the protection from the tumor microenvironment. Zengh and colleagues recently designed a nano-theranostic platform to battle this difficulty. HER2-DSG NPs (HER2-Doxorubicin-superparamagnetic iron oxide nanoparticles) were able to actively identify and target HER2-positive tumor cells and doxorubicin release. Moreover, NPs enhanced immunogenic cell death effect in vitro/in vivo, activating immune response and reducing CAFs’ population, increasing NPs access to cancer cells and leading to a great antineoplastic effect [94].
During the last years, different studies have highlighted the vital contribution of breast CAFs in the resistance mechanisms to anti HER2-targeted therapies. An interesting study by Nguyen et al. used recent advances in microtechnology of “organ-on-chip” to reproduce ex vivo the complexity of the tumor microenvironment ecosystems integrating the main populations of BrCa stroma: immune cells, endothelial cells and fibroblasts, together with BT-474 HER2+ BrCa cells, in the absence or presence of trastuzumab. They found a pro-invasive effect of CAFs as well as an antagonic immunomodulation effects of CAFs and trastuzumab in the HER2+ tumor ecosystem [95]. Marusyk and colleagues have defined that close physical contact between CAFs and BrCa cells protects BrCa cells from lapatinib and other chemotherapeutic agents such as taxol and doxorubicin [96]. Interestingly, this protection was not restricted to CAFs; stromal fibroblast from normal breast tissues and from brain metastases displayed similar effects [96].
Furthermore, Lin et al. found that increasing matrix stiffness was related with a reduction in lapatinib response, identifying fibronectin and type IV collagen as main contributors to therapy resistance, which was conditional on nuclear translocation of YAP transcription factor [97]. In vivo, antineoplastic effects of lapatinib were improved when treating with a recombinant human hyaluronidase (PEGPH20), which is currently being evaluated in multiple clinical trials under combined treatment settings [96,98].
On the other hand, paracrine communication between BrCa cells and CAFs has been shown to play a key role in HER2-targeted therapy resistance as well. We have recently described that soluble fibroblast growth factor 5 (FGF5) produced by the neighboring CAFs activates FGFR2 in HER2+ BrCa cells which in turn promotes c-Src-mediated HER2 transactivation. Additionally, we have demonstrated that the combination of anti-HER2 targeted therapies with FGFR2 inhibitors is able to overcome the resistance to trastuzumab and lapatinib in vitro and in vivo [99]. Additionally, in a cohort of trastuzumab-treated patients, high FGF5 stromal expression and high levels of HER2 phosphorylation define a group of patients who will not benefit from trastuzumab neoadjuvant treatment. These data led us to propose FGF5 and phospho-HER2 as potential biomarkers to predict resistance to HER2-targeted therapies [99]. In line with our results, Akhand et al., using HER2+ patient derived xenografts (PDX), obtained and characterized TDM1-resistant cells, also unable of responding to lapatinib and afatinib, but with an acquired sensitivity to a covalent FGFR kinase inhibitor, strongly supporting therapeutic combination of TDM1 with FGFR inhibitors in HER2+ BrCa treatment [100]. The role of FGFs and FGFRs in CAFs’ crosstalk with HER2+ BrCa has been also addressed by Hanker et al. [101]. By generating HER2-amplified BT-474 xenografts resistant to the combination of lapatinib and trastuzumab, they found increased copy number for FGF3, FGF4, and FGF19 genes in resistant tumors [101]. Co-treatment with the FGFR inhibitor lucitanib reversed collagen deposition in tumor stroma, as well as fibronectin, and increased αSMA staining (markers of activated stromal fibroblasts) in resistant tumors, suggesting that changes in the microenvironment are in part dependent on the FGFR pathway [101].
By using microenvironment microarrays (MEMA) [102], Watson et al. found that single growth factors like HGF or neuregulin 1 β (NRG1β), highly expressed by breast fibroblasts, were linked with lapatinib resistance in different HER2+ BrCa cell lines [103]. Independent HER2+ models also showed HGF ability to reestablish PI3K and Ras/Raf/MEK/ERK1/2 pathway activities that were initially lost under treatment with lapatinib, thereby stimulating drug resistance [104].
CAFs-derived cytokines have also been related with trastuzumab resistance. IL6 secreted by CAFs activates the STAT3 pathway and downregulates PTEN, inducing CSC population expansion [105]. Moreover, a latest report by Zervantonakis et al. has described that fibroblast-secreted factors induce survival in response to lapatinib, through mammalian target of rapamycin (mTOR) signaling activation and enhanced antiapoptotic protein levels. In accordance, mTOR, Bcl-XL or myeloid cell leukemia sequence 1 (MCL1) inhibitors restore lapatinib drug sensitivity [106]. Undoubtedly, these studies underline the idea that co-targeting the tumor and the stroma poses a great opportunity to improve current anti-HER2 therapies and to overcome treatment resistances (Figure 1 and Table S4).

4. CAFs and BrCa Metastasis

Metastasis is the principal cause of cancer death. As previously exposed, the crosstalk between stromal and tumor cells sustains cell survival, cell growth and therapeutic resistance. In this section, we will examine evidence involving fibroblast role in BrCa metastasis. We will analyze both the role of primary tumor CAFs in mediating the first steps of the metastatic cascade, such as epithelial to mesenchymal transition (EMT), migration and invasion, and the role of the secondary organs’ fibroblasts in the regulation of disseminated tumor cells’ (DTCs) colonization, dormancy and survival.

4.1. Contribution of CAFs from Primary Tumor Stroma to Metastasis Progression

Metastasis is a complex process that needs assistance from neighboring cells to be more efficient [107]. Hasabe et al. have shown that the existence of fibrotic foci correlates with nodal and distant organ metastasis in patients with IDC [108].
CAFs have been described to contribute to BrCa cells invasion by releasing growth factors (epidermal growth factor (EGF), FGF2 [109], PDGF, vascular endothelial growth factors (VEGFs), insulin-like growth factor (IGF) [110], HGF, tumor necrosis factor (TNF), CXCL12/ stromal cell-derived factor 1 (SDF1) [111,112]) as well as cytokines and chemokines CCL8 [113], CXCL16 [61], IL6 [26,114], IL4 [115], CXCL1 [116], CXCL14 [64], CCL5) [117,118], CXCL8/IL8 and CCL2 [62], IL32 [119]) which influence BrCa cell motility [120]. In a recent study Suh et al. evidenced that CAFs secretion of FGF2 was sufficient to enhance MDA-MB-231 cells growth, migration and invasion via FGFR1 signaling [109]. Dvorak et al. showed that CXCL12/SDF1 secretion induces mammalian Diaphanous-related formin-2 (MDia2) deregulation, leading to breakdown of the F-actin cytoskeleton and a subsequent increase of BrCa cell motility [112]. Moreover, αSMA+ fibroblasts produce high amounts of CXCL12/SDF1 and IGF1, which select cancer cells with high Src activity prone to colonize CXCL12/SDF1-rich bone marrow microenvironment, further suggesting a potential role of primary tumor CAFs in educating BrCa cells to metastasize to the bone microenvironment [121]. In addition, fibroblasts at tumor margins secrete higher levels of CCL8, generating a gradient between the epithelium, the stroma and the periphery that attracts BrCa cells to migrate and disseminate [113]. Besides, high stromal expression of CXCL14 is significantly associated with shorter recurrence periods [64]. A tumor necrosis factor alfa (TNFα) and IL1β enriched environment promotes metastasis in TNBrCa cells through the shift to a metastatic phenotype in both stromal and cancer cells, leading to an increased expression of pro-metastatic and immune-evasive chemokines, CXCL8/IL8, CCL2 and CCL5, among others, induction of angiogenesis and enhanced invasive and migration abilities of TNBrCa cells [62,122,123,124,125,126,127].
Several reports have also defined the importance of the release by CAFs of adipokines such as leptin in promoting the malignant aggressiveness of BrCa cells [128]. Giordano et al. described leptin as a regulator of the crosstalk between the stromal and BrCa cells. Leptin production by CAFs induce mammosphere formation, suggesting leptin can promote BrCa cells stemness [129]. Moreover, the inhibition of leptin with a synthetic farnesoid X receptor (FXR) agonist GW4064, reduces the pro-tumorigenic effect of CAFs in BrCa [130]. Other groups have also described an important role of leptin in potentiating BrCa cells migration and angiogenesis, being also involved in EMT [131]
Luga et al. also reported that CAFs produce exosomes that boost the motility and metastatic potential of BrCa cells by activating Wintless-INT family (Wnt) signaling [132]. Otherwise, BrCa exosomes such as miR-146a regulate the activation of Wnt/β-catenin signaling pathway on fibroblasts, which become CAFs contributing to invasive and metastatic abilities of BrCa cells [133]. Conversely, CAFs-derived exosomes induce proliferation and metastasis through the miR-500a-5p transference to luminal and basal-like BrCa cells. miR-500a-5p interacts with and blocks the effects of USP28, considered by the authors as a possible BrCa tumor suppressor [134]. Another work demonstrates that CAF exosomes can release miR-21, -143, and -378 that enhance important aggressive cancer hallmarks such as stemness, EMT and the ability to grow in an anchorage-independent way in TNBrCa cells [135].
One of the key steps in the metastatic process is cancer cell EMT and CAFs have been described to secrete growth factors such as transforming growth factor beta 1 (TGFβ1), EGF, PDGF and HGF that will promote EMT in primary tumor cells [107,120,136,137]. Likewise, the increased OPN secretion by TIAM rac1 associated GEF 1 (TIAM-1)-defective CAFs stimulates BrCa EMT, stemness and invasion [138]. Recently, Wen et al. have described that CAFs secrete IL32 that binds integrin β3 on BrCa cells resulting in p38 MAPK pathway activation, further enhancing the expressions of fibronectin, N-cadherin and vimentin [119]. In addition, it seems that activation of autophagy and expression of FAP-α in CAFs can activate EMT in cancer cells via Wnt pathway, resulting in an increase in cell migration, invasion and proliferation [139,140]. An interesting study describes that CAFs expression of phosphatidylinositol-4,5-bisphosphate 3-kinase catalytic subunit delta (PIK3Cδ) fosters TNBrCa cell metastasis, controlling mainly invasion through the inhibition of tumor suppressor genes such as Nuclear Receptor Subfamily 4 Group A Member 1 (NR4A1) [141]. Recently, Matsumura and colleagues have established that CAFs-secreted factors control the epithelial/mesenchymal plasticity of distinct metastatic tumor clusters, which comprise two different cell populations, one with a more epithelial phenotype and another with a mixed epithelial-mesenchymal phenotype. Both populations collectively contribute to enhanced tumor cell cluster formation and metastatic seeding in secondary organs [142]. Choi et al. have demonstrated, using three-dimensional in vitro models, that CAFs can facilitate transmigration of BrCa cells through the blood–brain barrier by increasing their expression of α5β1 and αvβ1 integrin and MET [143].
It is accepted that fibroblasts contribute to the remodeling of the surrounding stroma, which is a prerequisite for cancer cells’ invasion and metastasis. In vitro and in vivo studies have indicated that CAFs promote BrCa cell invasion and metastasis by secreting MMPs, such as MMP1, -2, -3, -7, -9, -13, and -14 [136,144,145]. Moreover, different research groups have observed that CAFs may generate tracks for tumor cells to go forward. For instance, Syndecan-1 expressing fibroblasts modify the collagen fibers in the extracellular matrix to form parallel structures that can be utilized by BrCa cells as a guide [146]. Ahirwar and colleagues showed that CAFs secreted CXCL12/SDF1, generating endothelial instability and hyper-permeable vasculature, facilitating the escape of tumor cells from the primary tumor to distant organs [147]. Other studies have additionally indicated that CAFs adopt a desmoplastic program with altered cell adhesion properties which pushes the progression of metastasis through induction of mechanical pressure on cancer tissue [148]. These studies highlight the important role of CAFs in mediating BrCa cell migration, EMT and invasion (Figure 2 and Table S5), hence suggesting stroma-targeted therapies might be an interesting approach to address metastatic disease.

4.2. Contribution of Fibroblasts from Metastatic Sites to Metastasis Formation

Once disseminated, tumor cells arrive to metastatic target organs where they will start interacting with stromal cells of the target organ. This new dialogue between DTCs and stromal cells will determine DTCs survival, activation of dormancy or the shift to a proliferative phenotype, hence regulating metastasis progression [149]. Gui et al. recently published a comparative study with BrCa fibroblasts from primary site (pCAFs), metastatic organs (mCAFs) and normal fibroblasts. They showed that mCAFs display more marked pro-tumorigenic effects, also conferring greater protection to cytotoxic drugs and improving metastatic capacity. All these differential capabilities of mCAFs were associated in part with their higher secretion of IFNβ and IGF2, which in turn activates IGF1R signaling [150]. BrCa DTCs usually colonize lungs, bone marrow and brain [151]. It has been described that communication between primary tumors and their potential metastatic sites starts even prior to the arrival of DTCs to the target organ. In the next paragraphs, we will focus on the accommodation of the stroma of the target organs and its role on DTCs fate.
During the early dissemination phase to the lungs, different primary tumor-derived mediators, such as growth factors, cytokines, extracellular matrix (ECM)-remodeling enzymes or extracellular vesicles and exosomes, have the ability to modify the pulmonary parenchyma creating a welcoming microenvironment for the tumor cells to seed, proliferate and survive [152]. Secretion of TNFα and TGFβ, along with vascular endothelial growth factor A (VEGFA), induce the expression of S100 calcium binding protein A8 (S100A8) and S100 calcium binding protein A9 (S100A9) in lung fibroblasts to prepare premetastatic niches [153]. S100A8 and S100A9 stimulate the recruitment of Mac-1+ myeloid cells to the lung, resulting in the secretion of factors able to stimulate migration like TNFα, CXCL2/MIP2 and TGFβ, as well as ECM remodeling [154]. A study by Raz et al. further demonstrated that bone marrow-derived stromal cells (BM-MSCs) are the precursors of a significant fraction of CAFs that are present in both primary breast and metastatic lung lesions [155], which are able to trigger a distinctive inflammatory profile, inducing pro-metastatic characteristics like angiogenesis and supporting metastasis to the lungs [155].
Exosomes derived from PT cancer cells play a critical role in transforming the lung microenvironment. A thorough study by Hoshino and colleagues revealed an organ specificity of exosome biodistribution that matched the organotrophic distribution of the cell line of origin [156]. Interestingly, they also found that lung-tropic exosomes, expressing α6β4 and α6β1 integrins, predominantly co-localized with S100 calcium binding protein A4 (S100A4)-positive fibroblasts and surfactant protein C-positive epithelial cells in the lungs, enhancing lung metastases [156]. Accordingly, integrin α6β4 targeting reduced lung metastasis [156]. Interestingly, Fong et al. have shown that exosomal miR-122 is able to reprogram lung niche cells to a lower glucose demand, by down-regulating pyruvate kinase activity. This metabolic reprogramming results in an increased DTCs glucose availability to effectively drive their proliferation [157].
Additionally, the contribution of BrCa-secreted exosomal long non-coding RNAs (LncRNAs) on lung pre-metastatic niche education has been described. These LncRNAs stimulated lung fibroblast proliferation and migration by targeting different genes and signaling pathways, including TGFβ, pentose phosphate, Hh, metabolic, and complement and coagulation cascade pathways [158]. Medeiros et al. found that extracellular vesicles (EVs) from TNBrCa cells were able to stimulate the expression of pre-metastatic ECM markers in lung fibroblasts [159].
During early metastatic colonization, lung DTCs reprogram resident lung fibroblasts to create a more favorable environment for proliferation and survival. For instance, DTCs decrease miR-30 family members’ expression in lung fibroblasts, which inhibit MMP9 expression, increasing vascular permeability and consequently favoring metastasis [160]. BrCa DTCs also induce the expression of αSMA in resident fibroblasts in the lung. Activated fibroblasts direct extracellular matrix remodeling by the secretion of fibronectin and LOXL2, creating a more tolerant microenvironment for DTCs to survive and outgrow [161]. Moreover, cancer cells are able to educate fibroblasts to produce periostin and Tenascin C (TNC), enhancing metastatic colonization of the lung [162]. Periostin expression in lung fibroblasts may be modulated by cancer cells through the secretion of transforming Growth Factor Beta 3 (TGFβ3). Fibroblasts in turn recruit Wnt ligands to present to cancer cells and promote DTCs stemness by inducing Wnt signaling, enhancing lung colonization [152]. Periostin also fosters the lung recruitment of MDSCs at the metastatic site, especially in ER-negative tumors, essential for achieving an immunosuppressive premetastatic niche environment. In addition, periostin can bind to TNC, anchoring it to ECM components such as fibronectin and type I collagen [163]. DTCs-derived TNC supports early metastasis outgrowth, until the lung stroma takes over as the main source of TNC. TNC can also induce the secretion of growth factors, like EGF or FGF, and interact with fibronectin, heparin-sulfate proteoglycans, fibrinogen, integrins, MMPs, and EGFR [164]. Additionally, high thrombospondin 2 (THBS2)-expressing cancer cells are also more efficient on activating lung fibroblasts, which subsequently induce the transition of tumor cells into a more epithelial phenotype [165]. In addition, Shu et al. have described a new role of the complement system C3a-C3aR on metastasis promotion by modulating CAFs activation, enhancing pro-metastatic cytokine secretion via the activation of PI3K-AKT signaling [166].
Bone is the main target organ for HER2+ BrCa metastasis, representing the major cause of pain and severe complications for these patients, including fracture, spinal cord compression or hypercalcemia [167]. Kang et al. have described a 6-gene bone metastasis signature in BrCa cell lines using mouse models. Interestingly, one of the genes was FGF5 that our group has recently described to be implicated in HER2+ targeted therapy response [99,168]. It has also been reported that BrCa cells in the bone marrow display higher expression of nanog homeobox (NANOG) OCT4 and SOX2 in the presence of MSC-derived CAFs and OPN [169], enhancing BrCa cells’ stemness. In turn, vimentin-expressing FAP+ fibroblasts produce miR-221 containing exosomes that further contribute to maintain the stem phenotype in favor of metastasis [87]. Furthermore, DTCs in the bone cavity produce abundant C-C motif chemokine ligand 4 (CCL4) as well, to attract type I collagen-expressing α-SMA+ fibroblasts expressing C-C motif chemokine receptor 5 (CCR5). Accumulated fibroblasts support cancer cells’ growth by enhancing connective tissue growth factor signals [170].
Regarding BrCa liver metastasis and the potential role of fibroblasts aiding the process, it has been defined that OPN promotes tumor progression via the transformation of MSC into CAFs in the liver as well. OPN was responsible for MSC CCL5 production. Moreover, the MSCs recovered from liver metastatic sites present CAF markers’ expression (α-SMA, TNC, CXCL12/SDF1, and fibroblast-specific protein-1) as well as MMP-2 and MMP-9 [171].
All these studies underline the essential role of resident fibroblasts in reprogramming the metastatic niche to favor DTCs seeding and survival at secondary organs (Figure 3 and Table S5). These insights could help design therapies targeting the cancer cell-stromal crosstalk of primary tumors and metastases, likely improving the clinical management and outcome of the advanced BrCa patients.

5. Targeting CAFs to Prevent BrCa Progression

As we have reviewed here, CAFs are major players in driving BrCa progression by affecting cancer cell invasion and colonization ability and its therapeutic response. Their diverse tumor-promoting functions, combined with their genetic stability and relative abundance among stromal cells, make them an appealing therapeutic target. Here, we will briefly highlight the major advances and challenges in the development of CAFs-targeted therapies in BrCa. The therapeutic approaches to target stromal cancer cells in BrCa can be categorized into three main lines: direct targeting of the ECM, targeting of CAFs and targeting of profibrotic signaling [172].
Direct targeting of the ECM has focused on the depletion of ECM molecules (collagen and hyaluronic acid (HA) mainly), and it has been successfully pursued using collagenase and/or relaxin enzymes as well as other approaches [172]. For example, PEGPH20 has been successfully used in BrCa cellular models [173], and it is known that it sensitizes HA-rich tumors to anti-PD-L1 immunotherapy in murine syngeneic breast cancer models.
The second antifibrotic strategy focuses on the elimination of CAFs as the main source of the ECM components. This therapeutic approach takes advantage of known CAFs markers such as αSMA, FSP1, and FAP-α to identify and eliminate them [172]. A BrCa phase I clinical trial testing the effects of the construct IL-2v targeting FAP (RO6874281) administered either alone or in combination with trastuzumab or cetuximab (ClinicalTrials.gov; Identifier: NCT02627274) is currently undergoing. However, this strategy still faces numerous obstacles, like unspecific markers and heterogeneity among the CAFs population. Interestingly, a specific population of BMDCs, granulin-expressing BMDCs, are recruited to metastatic niches and induce the tumor-promoting function of mammary fibroblasts. It has been suggested that targeting these granulin-expressing cells could be exploited as anticancer therapy to avoid CAFs support of tumor progression [174]. Moreover, all-trans retinoic acid (ATRA) treatment is able to reprogram activated CAFs, restoring their quiescence in pancreatic cancer [175], and decreasing malignant transformation of ductal carcinoma breast cancer cells [176].
Interfering with the TGFβ or Hh signaling pathways have also been successfully tested in BrCa. TGFβ signaling blocking, by either soluble TGFβ type II receptor or TGFβ neutralizing antibody, significantly decreased tumor growth and metastasis in BrCa [177]. Remarkably, TGFβ signaling can also be blocked through angiotensin receptor inhibitors (ARB), such as losartan, which enhanced delivery of chemotherapy and improved overall survival in BrCa models [178]. Pirfenidone (PFD), an anti-fibrotic agent as well as a TGFβ antagonist, has also been effectively examined in TNBrCa patient-derived xenograft models [66]. Several Hh pathway inhibitors have also been tested, being IPI-926, the one with the best results [179]. Furthermore, focal adhesion kinase (FAK) signaling has also been related with the formation of fibrotic tumor microenvironment [172] and appears as a druggable target, not only in tumor cells but also in the tumor microenvironment [180]. The stromal targets list in BrCa is continually growing, as do newer therapies against stromal components [181,182]. For example, in vitro and in vivo approaches against the phosphodiesterase PDE5 (Sildenafil) or the nuclear receptors FXR (GW4064) have shown promising results due to their relevant role in CAFs biology and pro-tumoral effect [130,181,183,184].
These therapeutic strategies targeting CAF/stroma-specific pathways are already being evaluated in several clinical trials on BrCa, or other solid tumors. Most of them combine classical strategies with CAFs-targeted therapies compounds to successfully improve cancer therapy. The main targets are the following: TGFβ-pathway (fresolimumab (NCT01401062) or galunisertib (NCT02304419)), Hh (vismodegib (NCT02694224) or LDE225 (NCT02027376, NCT02694224)), Notch (MK0752 (NCT00645333)), FGFR-FGFR ligand (Dovitinib (NCT01548924), AZD4547 (NCT01791985), FAP (RO6874813 (NCT02558140)), Hyaluronic acid in pancreatic cancer (PEGPH20 (NCT01453153)) and Multitargets ((Dasatinib (NCT00546104), lucitanib (NCT02053636), Famitinib (NCT04733417, NCT01653574, NCT04129996), Motesanib (NCT01349088).

6. Conclusions

In this review, we have summarized the most recent and significant findings on the role of CAFs in BrCa therapy resistance and metastasis. The data revised here emphasizes the role of CAFs as novel, promising a therapeutic target in BrCa suggesting that combination strategies aimed at targeting both BrCa cells and the surrounding microenvironment may enable more efficient therapeutic responses. Furthermore, the identification of novel and specific biomarkers of CAFs will be priceless for accelerating the translation into the clinic of CAFs-targeted therapies.

Supplementary Materials

The following are available online at https://www.mdpi.com/article/10.3390/cancers13133146/s1, Table S1: CAFs secreted molecules and function in DCIS, Table S2: Protein names abbreviations, Table S3: CAFs secreted molecules and function in radiotherapy. Table S4: CAFs secreted molecules and function in BrCa subtypes therapy resistance, Table S5: CAFs secreted molecules function in metastasis,

Author Contributions

Conceptualization, P.F.-N. and P.B.; writing—original draft preparation, P.F.-N., G.F., Á.G.-U., and P.B.; writing—review and editing, P.F.-N., G.F., Á.G.-U., P.G., N.C., P.B.; funding acquisition, P.G., N.C., P.B. All authors have read and agreed to the published version of the manuscript.

Funding

This work was supported by grants from the Spanish Ministry of Economy and Competitiveness (PID2019-104991RB-I00 to PB). All funding was cosponsored by the European FEDER Program. Á.G.-U. is supported by the Madrid Community Program for Talent Attraction (MRF 2017-T1/BMD-5468). P.F.-N. is supported by a Juan de la Cierva contract from the Spanish Ministry of science, innovation and universities. P.F.-N. and G.F. are supported by Cellex Foundation, and P.F.-N., G.F., N.C., and P.G. are funded by La Marató TV3 (201915-30-31), and by Secretaria d’Universitats i Recerca del Departament d’Economia i Coneixement (2017_SGR_1305).

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Acknowledgments

The authors thank Darya Kulyk for her help preparing the schemes that are shown in the figures and the graphical abstract.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Harbeck, N.; Penault-Llorca, F.; Cortes, J.; Gnant, M.; Houssami, N.; Poortmans, P.; Ruddy, K.; Tsang, J.; Cardoso, F. Breast cancer. Nat. Rev. Dis. Prim. 2019, 5, 1–31. [Google Scholar] [CrossRef] [PubMed]
  2. Perou, C.M.; Sørlie, T.; Eisen, M.B.; van de Rijn, M.; Jeffrey, S.S.; Rees, C.A.; Pollack, J.R.; Ross, D.T.; Johnsen, H.; Akslen, L.A.; et al. Molecular portraits of human breast tumours. Nature 2000, 406, 747–752. [Google Scholar] [CrossRef] [PubMed]
  3. Sorlie, T.; Perou, C.M.; Tibshirani, R.; Aas, T.; Geisler, S.; Johnsen, H.; Hastie, T.; Eisen, M.B.; van de Rijn, M.; Jeffrey, S.S.; et al. Gene expression patterns of breast carcinomas distinguish tumor subclasses with clinical implications. Proc. Natl. Acad. Sci. USA 2001, 98, 10869–10874. [Google Scholar] [CrossRef] [Green Version]
  4. Robson, M.; Im, S.-A.; Senkus, E.; Xu, B.; Domchek, S.M.; Masuda, N.; Delaloge, S.; Li, W.; Tung, N.; Armstrong, A.; et al. Olaparib for metastatic breast cancer in patients with a germline BRCA mutation. N. Engl. J. Med. 2017, 377, 523–533. [Google Scholar] [CrossRef]
  5. Robson, M.; Goessl, C.; Domchek, S. Olaparib for metastatic germline BRCA-mutated breast cancer. N. Engl. J. Med. 2017, 377, 1792–1793. [Google Scholar] [CrossRef] [PubMed]
  6. Klemm, F.; Joyce, J.A. Microenvironmental regulation of therapeutic response in cancer. Trends Cell Biol. 2015, 25, 198–213. [Google Scholar] [CrossRef] [Green Version]
  7. Ruocco, M.R.; Avagliano, A.; Granato, G.; Imparato, V.; Masone, S.; Masullo, M.; Nasso, R.; Montagnani, S.; Arcucci, A. Involvement of breast cancer-associated fibroblasts in tumor development, therapy resistance and evaluation of potential therapeutic strategies. Curr. Med. Chem. 2018, 25, 3414–3434. [Google Scholar] [CrossRef] [PubMed]
  8. Arcucci, A.; Ruocco, M.R.; Granato, G.; Sacco, A.M.; Montagnani, S. Cancer: An oxidative crosstalk between solid tumor cells and cancer associated fibroblasts. BioMed Res. Int. 2016, 2016, 1–7. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  9. Buchsbaum, R.J.; Oh, S.Y. Breast cancer-associated fibroblasts: Where we are and where we need to go. Cancers 2016, 8, 19. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  10. Ao, Z.; Shah, S.H.; Machlin, L.M.; Parajuli, R.; Miller, P.C.; Rawal, S.; Williams, A.J.; Cote, R.J.; Lippman, M.E.; Datar, R.H.; et al. Identification of cancer-associated fibroblasts in circulating blood from patients with metastatic breast cancer. Cancer Res. 2015, 75, 4681–4687. [Google Scholar] [CrossRef] [Green Version]
  11. Costa, A.; Kieffer, Y.; Scholer-Dahirel, A.; Pelon, F.; Bourachot, B.; Cardon, M.; Sirven, P.; Magagna, I.; Fuhrmann, L.; Bernard, C.; et al. Fibroblast heterogeneity and immunosuppressive environment in human breast cancer. Cancer Cell 2018, 33, 463–479. [Google Scholar] [CrossRef] [Green Version]
  12. Wu, F.; Yang, J.; Liu, J.; Wang, Y.; Mu, J.; Zeng, Q.; Deng, S.; Zhou, H. Signaling pathways in cancer-associated fibroblasts and targeted therapy for cancer. Signal Transduct. Target Ther. 2021, 6, 218. [Google Scholar] [CrossRef] [PubMed]
  13. Ganguly, D.; Chandra, R.; Karalis, J.; Teke, M.; Aguilera, T.; Maddipati, R.; Wachsmann, M.B.; Ghersi, D.; Siravegna, G.; Iii, H.; et al. Cancer-associated fibroblasts: Versatile players in the tumor microenvironment. Cancers 2020, 12, 2652. [Google Scholar] [CrossRef] [PubMed]
  14. Avagliano, A.; Granato, G.; Ruocco, M.R.; Romano, V.; Belviso, I.; Carfora, A.; Montagnani, S.; Arcucci, A. Metabolic reprogramming of cancer associated fibroblasts: The slavery of stromal fibroblasts. BioMed Res. Int. 2018, 2018, 1–12. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  15. Monteran, L.; Erez, N. The dark side of fibroblasts: Cancer-associated fibroblasts as mediators of immunosuppression in the tumor microenvironment. Front. Immunol. 2019, 10, 1835. [Google Scholar] [CrossRef] [Green Version]
  16. American Cancer Society. Cancer Facts & Figures 2020; American Cancer Society: Atlanta, GA, USA, 2020. [Google Scholar]
  17. Pareja, F.; Brown, D.N.; Lee, J.Y.; Paula, A.D.C.; Selenica, P.; Bi, R.; Geyer, F.C.; Gazzo, A.; da Silva, E.M.; Vahdatinia, M.; et al. Whole-exome sequencing analysis of the progression from non–low-grade ductal carcinoma in situto invasive ductal carcinoma. Clin. Cancer Res. 2020, 26, 3682–3693. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  18. Hanahan, D.; Weinberg, R.A. Hallmarks of cancer: The next generation. Cell 2011, 144, 646–674. [Google Scholar] [CrossRef] [Green Version]
  19. Nelson, A.C.; Machado, H.L.; Schwertfeger, K.L. Breaking through to the other side: Microenvironment contributions to DCIS initiation and progression. J. Mammary Gland. Biol. Neoplasia 2018, 23, 207–221. [Google Scholar] [CrossRef]
  20. Ma, X.-J.; Dahiya, S.; Richardson, E.; Erlander, M.; Sgroi, D.C. Gene expression profiling of the tumor microenvironment during breast cancer progression. Breast Cancer Res. 2009, 11, R7. [Google Scholar] [CrossRef] [Green Version]
  21. Sharma, M.; Beck, A.H.; Webster, J.A.; Espinosa, I.; Montgomery, K.; Varma, S.; van de Rijn, M.; Jensen, K.C.; West, R.B. Analysis of stromal signatures in the tumor microenvironment of ductal carcinoma in situ. Breast Cancer Res. Treat. 2010, 123, 397–404. [Google Scholar] [CrossRef] [PubMed]
  22. Fernández-Nogueira, P.; Mancino, M.; Fuster, G.; Bragado, P.; de Puig, M.P.; Gascón, P.; Casado, F.J.; Carbó, N. Breast mammographic density: Stromal implications on breast cancer detection and therapy. J. Clin. Med. 2020, 9, 776. [Google Scholar] [CrossRef] [Green Version]
  23. Lisanti, M.P.; Reeves, K.; Peiris-Pagès, M.; Chadwick, A.L.; Sanchez-Alvarez, R.; Howell, A.; Martinez-Outschoorn, U.E.; Sotgia, F.; Tsirigos, A.; Pavlides, S.; 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 2013, 13, 580–599. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  24. Hu, M.; Peluffo, G.; Chen, H.; Gelman, R.; Schnitt, S.; Polyak, K. Role of COX-2 in epithelial-stromal cell interactions and progression of ductal carcinoma in situ of the breast. Proc. Natl. Acad. Sci. USA 2009, 106, 3372–3377. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  25. Sung, K.E.; Yang, N.; Pehlke, C.; Keely, P.J.; Eliceiri, K.W.; Friedl, A.; Beebe, D.J. Transition to invasion in breast cancer: A microfluidic in vitro model enables examination of spatial and temporal effects. Integr. Biol. 2011, 3, 439–450. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  26. Osuala, K.O.; Sameni, M.; Shah, S.; Aggarwal, N.; Simonait, M.L.; Franco, O.E.; Hong, Y.; Hayward, S.W.; Behbod, F.; Mattingly, R.R.; et al. Il-6 signaling between ductal carcinoma in situ cells and carcinoma-associated fibroblasts mediates tumor cell growth and migration. BMC Cancer 2015, 15, 1–15. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  27. Dang, T.T.; Prechtl, A.M.; Pearson, G.W. Breast cancer subtype-specific interactions with the microenvironment dictate mechanisms of invasion. Cancer Res. 2011, 71, 6857–6866. [Google Scholar] [CrossRef] [Green Version]
  28. Conklin, M.W.; Gangnon, R.E.; Sprague, B.L.; van Germert, L.; Hampton, J.M.; Eliceiri, K.W.; Bredfeldt, J.S.; Liu, Y.; Surachaicharn, N.; Newcomb, P.A.; et al. Collagen alignment as a predictor of recurrence after ductal carcinoma in situ. Cancer Epidemiol. Biomark. Prev. 2017, 27, 138–145. [Google Scholar] [CrossRef] [Green Version]
  29. Provenzano, P.P.; Eliceiri, K.W.; Campbell, J.M.; Inman, D.R.; White, J.G.; Keely, P.J. Collagen reorganization at the tumor-stromal interface facilitates local invasion. BMC Med. 2006, 4, 38. [Google Scholar] [CrossRef] [Green Version]
  30. Sprague, B.L.; Vacek, P.M.; Mulrow, S.E.; Evans, M.F.; Trentham-Dietz, A.; Herschorn, S.D.; James, T.A.; Surachaicharn, N.; Keikhosravi, A.; Eliceiri, K.W.; et al. Collagen organization in relation to ductal carcinoma in situ pathology and outcomes. Cancer Epidemiol. Biomark. Prev. 2021, 30, 80–88. [Google Scholar] [CrossRef]
  31. Bernard, S.; Myers, M.; Bin Fang, W.; Zinda, B.; Smart, C.; Lambert, D.; Zou, A.; Fan, F.; Cheng, N. CXCL1 derived from mammary fibroblasts promotes progression of mammary lesions to invasive carcinoma through CXCR2 dependent mechanisms. J. Mammary Gland. Biol. Neoplasia 2018, 23, 249–267. [Google Scholar] [CrossRef]
  32. Shaker, H.; Bundred, N.; Albadry, H.; Nicholson, S.; Castle, J.; Lumsden, L.; Pritchard, S.; Landberg, G.; Kirwan, C. PO-21—Stromal fibroblasts in preinvasive breast cancer (ductal carcinoma in situ, DCIS) demonstrate a cancer-like procoagulant phenotypic switch that may facilitate invasion. Thromb. Res. 2016, 140, S184. [Google Scholar] [CrossRef]
  33. Sameni, M.; Cavallo-Medved, D.; Franco, O.E.; Chalasani, A.; Ji, K.; Aggarwal, N.; Anbalagan, A.; Chen, X.; Mattingly, R.R.; Hayward, S.W.; et al. Pathomimetic avatars reveal divergent roles of microenvironment in invasive transition of ductal carcinoma in situ. Breast Cancer Res. 2017, 19, 56. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  34. Shaker, H.; Bundred, N.J.; Landberg, G.; Pritchard, S.A.; Albadry, H.; Nicholson, S.L.; Harries, L.J.; Heah, J.Y.E.; Castle, J.; Kirwan, C.C. Breast cancer stromal clotting activation (Tissue Factor and thrombin): A pre-invasive phenomena that is prognostic in invasion. Cancer Med. 2020, 9, 1768–1778. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  35. Yu, L.-N.; Liu, Z.; Tian, Y.; Zhao, P.-P.; Hua, X. FAP-a and GOLPH3 are hallmarks of DCIS progression to invasive breast cancer. Front. Oncol. 2019, 9, 1424. [Google Scholar] [CrossRef]
  36. Witkiewicz, A.K.; Dasgupta, A.; Nguyen, K.H.; Liu, C.; Kovatich, A.J.; Schwartz, G.F.; Pestell, R.G.; Sotgia, F.; Rui, H.; Lisanti, M.P. Stromal caveolin-1 levels predict early DCIS progression to invasive breast cancer. Cancer Biol. Ther. 2009, 8, 1071–1079. [Google Scholar] [CrossRef] [Green Version]
  37. Krisnawan, V.E.; Stanley, J.A.; Schwarz, J.K.; DeNardo, D.G. Tumor microenvironment as a regulator of radiation therapy: New insights into stromal-mediated radioresistance. Cancers 2020, 12, 2916. [Google Scholar] [CrossRef] [PubMed]
  38. Wang, Z.; Tang, Y.; Tan, Y.; Wei, Q.; Yu, W. Cancer-associated fibroblasts in radiotherapy: Challenges and new opportunities. Cell Commun. Signal. 2019, 17, 47. [Google Scholar] [CrossRef] [Green Version]
  39. Barker, H.E.; Paget, J.T.E.; Khan, A.; Harrington, K. The tumour microenvironment after radiotherapy: Mechanisms of resistance and recurrence. Nat. Rev. Cancer 2015, 15, 409–425. [Google Scholar] [CrossRef]
  40. Steer, A.; Cordes, N.; Jendrossek, V.; Klein, D. Impact of cancer-associated fibroblast on the radiation-response of solid xenograft tumors. Front. Mol. Biosci. 2019, 6, 70. [Google Scholar] [CrossRef] [Green Version]
  41. Barcellos-Hoff, M.H.; Derynck, R.; Tsang, M.L.; A Weatherbee, J. Transforming growth factor-beta activation in irradiated murine mammary gland. J. Clin. Investig. 1994, 93, 892–899. [Google Scholar] [CrossRef] [PubMed]
  42. Bouquet, F.; Pal, A.; Pilones, K.A.; Demaria, S.; Hann, B.; Akhurst, R.J.; Babb, J.; Lonning, S.M.; Dewyngaert, J.K.; Formenti, S.C.; et al. TGFβ1 inhibition increases the radiosensitivity of breast cancer cells in vitro and promotes tumor control by radiation in vivo. Clin. Cancer Res. 2011, 17, 6754–6765. [Google Scholar] [CrossRef] [Green Version]
  43. Biswas, S.; Guix, M.; Rinehart, C.; Dugger, T.C.; Chytil, A.; Moses, H.L.; Freeman, M.L.; Arteaga, C.L. Inhibition of TGF-β with neutralizing antibodies prevents radiation-induced acceleration of metastatic cancer progression. J. Clin. Investig. 2007, 117, 1305–1313. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  44. De Bacco, F.; Luraghi, P.; Medico, E.; Reato, G.; Girolami, F.; Perera, T.; Gabriele, P.; Comoglio, P.; Boccaccio, C. Induction of MET by ionizing radiation and its role in radioresistance and invasive growth of cancer. J. Natl. Cancer Inst. 2011, 103, 645–661. [Google Scholar] [CrossRef] [Green Version]
  45. Park, C.C.; Zhang, H.J.; Yao, E.S.; Park, C.J.; Bissell, M.J. β1 integrin inhibition dramatically enhances radiotherapy efficacy in human breast cancer xenografts. Cancer Res. 2008, 68, 4398–4405. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  46. Boelens, M.C.; Wu, T.J.; Nabet, B.Y.; Xu, B.; Qiu, Y.; Yoon, T.; Azzam, D.J.; Victor, C.T.-S.; Wiemann, B.Z.; Ishwaran, H.; et al. Exosome transfer from stromal to breast cancer cells regulates therapy resistance pathways. Cell 2014, 159, 499–513. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  47. Pazolli, E.; Alspach, E.; Milczarek, A.; Prior, J.; Piwnica-Worms, D.; Stewart, S.A. Chromatin remodeling underlies the senescence-associated secretory phenotype of tumor stromal fibroblasts that supports cancer progression. Cancer Res. 2012, 72, 2251–2261. [Google Scholar] [CrossRef] [Green Version]
  48. Tsai, K.K.C.; Stuart, J.; Chuang, Y.-Y.E.; Little, J.B.; Yuan, Z.-M. Low-dose radiation-induced senescent stromal fibroblasts render nearby breast cancer cells radioresistant. Radiat. Res. 2009, 172, 306–313. [Google Scholar] [CrossRef]
  49. Pareja, F.; Geyer, F.C.; Marchiò, C.; Burke, K.A.; Weigelt, B.; Reis-Filho, J.S. Triple-negative breast cancer: The importance of molecular and histologic subtyping, and recognition of low-grade variants. NPJ Breast Cancer 2016, 2, 16036. [Google Scholar] [CrossRef]
  50. Rong, G.; Kang, H.; Wang, Y.; Hai, T.; Sun, H. Candidate markers that associate with chemotherapy resistance in breast cancer through the study on taxotere-induced damage to tumor microenvironment and gene expression profiling of Carcinoma-Associated Fibroblasts (CAFs). PLoS ONE 2013, 8, e70960. [Google Scholar] [CrossRef] [Green Version]
  51. Cazet, A.S.; Hui, M.N.; Elsworth, B.; Wu, S.Z.; Roden, D.; Chan, C.-L.; Skhinas, J.N.; Collot, R.; Yang, J.; Harvey, K.; et al. Targeting stromal remodeling and cancer stem cell plasticity overcomes chemoresistance in triple negative breast cancer. Nat. Commun. 2018, 9, 1–18. [Google Scholar] [CrossRef] [Green Version]
  52. Li, Q.; Li, M.; Zheng, K.; Tang, S.; Ma, S. Expression pattern analysis and drug differential sensitivity of cancer-associated fibroblasts in triple-negative breast cancer. Transl. Oncol. 2021, 14, 100891. [Google Scholar] [CrossRef] [PubMed]
  53. Hu, C.; Liu, X.; Ran, W.; Meng, J.; Zhai, Y.; Zhang, P.; Yin, Q.; Yu, H.; Zhang, Z.; Li, Y. Regulating cancer associated fibroblasts with losartan-loaded injectable peptide hydrogel to potentiate chemotherapy in inhibiting growth and lung metastasis of triple negative breast cancer. Biomaterials 2017, 144, 60–67. [Google Scholar] [CrossRef]
  54. Maia, A.; Gu, Z.; Koch, A.; Berdiel-Acer, M.; Will, R.; Schlesner, M.; Wiemann, S. IFNβ1 secreted by breast cancer cells undergoing chemotherapy reprograms stromal fibroblasts to support tumour growth after treatment. Mol. Oncol. 2021, 15, 1308–1329. [Google Scholar] [CrossRef]
  55. 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]
  56. Wee, Z.N.; Yatim, S.M.J.M.; Kohlbauer, V.K.; Feng, M.; Goh, J.Y.; Bao, Y.; Lee, P.L.; Zhang, S.; Wang, P.P.; Lim, E.; et al. IRAK1 is a therapeutic target that drives breast cancer metastasis and resistance to paclitaxel. Nat. Commun. 2015, 6, 8746. [Google Scholar] [CrossRef] [PubMed]
  57. Karagoz, K.; Sinha, R.; Arga, K.Y. Triple negative breast cancer: A multi-omics network discovery strategy for candidate targets and driving pathways. OMICS: A J. Integr. Biol. 2015, 19, 115–130. [Google Scholar] [CrossRef]
  58. Hamaguchi, T.; Wakabayashi, H.; Matsumine, A.; Sudo, A.; Uchida, A. TNF inhibitor suppresses bone metastasis in a breast cancer cell line. Biochem. Biophys. Res. Commun. 2011, 407, 525–530. [Google Scholar] [CrossRef]
  59. Jotzu, C.; Alt, E.; Welte, G.; Li, J.; Hennessy, B.T.; Devarajan, E.; Krishnappa, S.; Pinilla, S.; Droll, L.; Song, Y.-H. Adipose tissue-derived stem cells differentiate into carcinoma-associated fibroblast-like cells under the influence of tumor-derived factors. Anal. Cell. Pathol. 2010, 33, 61–79. [Google Scholar] [CrossRef]
  60. Pinilla, S.; Alt, E.; Khalek, F.A.; Jotzu, C.; Muehlberg, F.; Beckmann, C.; Song, Y.-H. Tissue resident stem cells produce CCL5 under the influence of cancer cells and thereby promote breast cancer cell invasion. Cancer Lett. 2009, 284, 80–85. [Google Scholar] [CrossRef] [PubMed]
  61. Allaoui, R.; Bergenfelz, C.; Mohlin, S.; Hagerling, C.; Salari, K.; Werb, Z.; Anderson, R.; Ethier, S.P.; Jirström, K.; Påhlman, S.; et al. Cancer-associated fibroblast-secreted CXCL16 attracts monocytes to promote stroma activation in triple-negative breast cancers. Nat. Commun. 2016, 7, 13050. [Google Scholar] [CrossRef]
  62. Liubomirski, Y.; Lerrer, S.; Meshel, T.; Morein, D.; Rubinstein-Achiasaf, L.; Sprinzak, D.; Wiemann, S.; Körner, C.; Ehrlich, M.; Ben-Baruch, A. Notch-mediated tumor-stroma-inflammation networks promote invasive properties and CXCL8 expression in triple-negative breast cancer. Front. Immunol. 2019, 10, 804. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  63. Liubomirski, Y.; Lerrer, S.; Meshel, T.; Rubinstein-Achiasaf, L.; Morein, D.; Wiemann, S.; Körner, C.; Ben-Baruch, A. Tumor-stroma-inflammation networks promote pro-metastatic chemokines and aggressiveness characteristics in triple-negative breast cancer. Front. Immunol. 2019, 10, 757. [Google Scholar] [CrossRef] [Green Version]
  64. Sjöberg, E.; Augsten, M.; Bergh, J.; Jirström, K.; Östman, A. Expression of the chemokine CXCL14 in the tumour stroma is an independent marker of survival in breast cancer. Br. J. Cancer 2016, 114, 1117–1124. [Google Scholar] [CrossRef] [Green Version]
  65. Yoshikawa, K.; Ishida, M.; Yanai, H.; Tsuta, K.; Sekimoto, M.; Sugie, T. Prognostic significance of PD-L1-positive cancer-associated fibroblasts in patients with triple-negative breast cancer. BMC Cancer 2021, 21, 1–10. [Google Scholar] [CrossRef]
  66. Takai, K.; Le, A.; Weaver, V.M.; Werb, Z. Targeting the cancer-associated fibroblasts as a treatment in triple-negative breast cancer. Oncotarget 2016, 7, 82889–82901. [Google Scholar] [CrossRef] [Green Version]
  67. Coates, A.S.; Winer, E.P.; Goldhirsch, A.; Gelber, R.D.; Gnant, M.; Piccart-Gebhart, M.; Thürlimann, B.; Senn, H.-J.; Andre, F.; Baselga, J.; et al. Tailoring therapies—improving the management of early breast cancer: St Gallen International Expert Consensus on the Primary Therapy of Early Breast Cancer 2015. Ann. Oncol. 2015, 26, 1533–1546. [Google Scholar] [CrossRef] [PubMed]
  68. Clarke, R.; Liu, M.C.; Bouker, K.B.; Gu, Z.; Lee, R.Y.; Zhu, Y.; Skaar, T.C.; Gomez, B.; O’Brien, K.; Wang, Y.; et al. Antiestrogen resistance in breast cancer and the role of estrogen receptor signaling. Oncogene 2003, 22, 7316–7339. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  69. Early Breast Cancer Trialists’ Collaborative Group (EBCTCG); Davies, C.; Godwin, J.; Gray, R.; Clarke, M.; Cutter, D.; Darby, S.; McGale, P.; Pan, H.C.; Taylor, C.; et al. Relevance of breast cancer hormone receptors and other factors to the efficacy of adjuvant tamoxifen: Patient-level meta-analysis of randomised trials. Lancet 2011, 378, 771–784. [Google Scholar] [CrossRef] [Green Version]
  70. 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] [Green Version]
  71. 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 β1-integrin signaling pathway in tumor cells. Breast Cancer Res. 2015, 17, 1–18. [Google Scholar]
  72. Jena, B.C.; Das, C.K.; Banerjee, I.; Das, S.; Bharadwaj, D.; Majumder, R.; Mandal, M. Paracrine TGF-β1 from breast cancer contributes to chemoresistance in cancer associated fibroblasts via upregulation of the p44/42 MAPK signaling pathway. Biochem. Pharmacol. 2021, 186, 114474. [Google Scholar] [CrossRef]
  73. Yu, T.; Yang, G.; Hou, Y.; Tang, X.; Wu, C.; Wu, X.-A.; Guo, L.; Zhu, Q.; Luo, H.; Du, Y.-E.; et al. Cytoplasmic GPER translocation in cancer-associated fibroblasts mediates cAMP/PKA/CREB/glycolytic axis to confer tumor cells with multidrug resistance. Oncogene 2017, 36, 2131–2145. [Google Scholar] [CrossRef] [PubMed]
  74. Martinez-Outschoorn, U.E.; Goldberg, A.F.; Lin, Z.; Ko, Y.-H.; Flomenberg, N.; Wang, C.; Pavlides, S.; Pestell, R.G.; Howell, A.; Sotgia, F.; et al. Anti-estrogen resistance in breast cancer is induced by the tumor microenvironment and can be overcome by inhibiting mitochondrial function in epithelial cancer cells. Cancer Biol. Ther. 2011, 12, 924–938. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  75. Martinez-Outschoorn, U.E.; Trimmer, C.; Lin, Z.; Whitaker-Menezes, D.; Chiavarina, B.; Zhou, J.; Wang, C.; Pavlides, S.; Cantarin, M.P.M.; Capozza, F.; et al. Autophagy in cancer associated fibroblasts promotes tumor cell survival. Cell Cycle 2010, 9, 3515–3533. [Google Scholar] [CrossRef] [PubMed]
  76. Witkiewicz, A.K.; Dasgupta, A.; Sotgia, F.; Mercier, I.; Pestell, R.G.; Sabel, M.; Kleer, C.G.; Brody, J.R.; Lisanti, M.P. An absence of Stromal Caveolin-1 expression predicts early tumor recurrence and poor clinical outcome in human breast cancers. Am. J. Pathol. 2009, 174, 2023–2034. [Google Scholar] [CrossRef] [Green Version]
  77. Hiscox, S.; Jordan, N.J.; Jiang, W.; Harper, M.; McClelland, R.; Smith, C.; I Nicholson, R. Chronic exposure to fulvestrant promotes overexpression of the c-Met receptor in breast cancer cells: Implications for tumour–stroma interactions. Endocrine-Related Cancer 2006, 13, 1085–1099. [Google Scholar] [CrossRef] [PubMed]
  78. Tomlinson, D.; Knowles, M.; Speirs, V. Mechanisms of FGFR3 actions in endocrine resistant breast cancer. Int. J. Cancer 2011, 130, 2857–2866. [Google Scholar] [CrossRef] [PubMed]
  79. Turczyk, L.; Kitowska, K.; Mieszkowska, M.; Mieczkowski, K.; Czaplinska, D.; Piasecka, D.; Kordek, R.; Skladanowski, A.C.; Potemski, P.; Romanska, H.M.; et al. FGFR2-driven signaling counteracts tamoxifen effect on ERα-positive breast cancer cells. Neoplasia 2017, 19, 791–804. [Google Scholar] [CrossRef] [PubMed]
  80. Shekhar, M.P.; Santner, S.; Carolin, K.A.; Tait, L. Direct involvement of breast tumor fibroblasts in the modulation of tamoxifen sensitivity. Am. J. Pathol. 2007, 170, 1546–1560. [Google Scholar] [CrossRef] [Green Version]
  81. Nakaoka, H.J.; Tanei, Z.; Hara, T.; Weng, J.S.; Kanamori, A.; Hayashi, T.; Sato, H.; Orimo, A.; Otsuji, K.; Tada, K.; et al. Mint3-mediated L1CAM expression in fibroblasts promotes cancer cell proliferation via integrin α5β1 and tumour growth. Oncogenesis 2017, 6, e334. [Google Scholar] [CrossRef]
  82. Busch, S.; Rydén, L.; Stål, O.; Jirstrom, K.; Landberg, G. Low ERK phosphorylation in cancer-associated fibroblasts is associated with tamoxifen resistance in pre-menopausal breast cancer. PLoS ONE 2012, 7, e45669. [Google Scholar] [CrossRef]
  83. Pontiggia, O.; Sampayo, R.; Raffo, D.; Motter, A.; Xu, R.; Bissell, M.J.; Joffé, E.B.D.K.; Simian, M. The tumor microenvironment modulates tamoxifen resistance in breast cancer: A role for soluble stromal factors and fibronectin through β1 integrin. Breast Cancer Res. Treat. 2011, 133, 459–471. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  84. Leyh, B.; Dittmer, A.; Lange, T.; Martens, J.W.M.; Dittmer, J. Stromal cells promote anti-estrogen resistance of breast cancer cells through an insulin-like growth factor binding protein 5 (IGFBP5)/B-cell leukemia/lymphoma 3 (Bcl-3) axis. Oncotarget 2015, 6, 39307–39328. [Google Scholar] [CrossRef] [Green Version]
  85. Brechbuhl, H.M.; Finlay-Schultz, J.; Yamamoto, T.M.; Gillen, A.; Cittelly, D.M.; Tan, A.C.; Sams, S.B.; Pillai, M.M.; Elias, A.D.; Robinson, W.A.; et al. Fibroblast subtypes regulate responsiveness of luminal breast cancer to estrogen. Clin. Cancer Res. 2017, 23, 1710–1721. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  86. Gao, Y.; Li, X.; Zeng, C.; Liu, C.; Hao, Q.; Li, W.; Zhang, K.; Zhang, W.; Wang, S.; Zhao, H.; et al. CD63 + Cancer-Associated Fibroblasts Confer Tamoxifen Resistance to Breast Cancer Cells through Exosomal miR-22. Adv. Sci. 2020, 7, 2002518. [Google Scholar] [CrossRef]
  87. Sansone, P.; Berishaj, M.; Rajasekhar, V.K.; Ceccarelli, C.; Chang, Q.; Strillacci, A.; Savini, C.; Shapiro, L.; Bowman, R.L.; Mastroleo, C.; et al. Evolution of cancer stem-like cells in endocrine-resistant metastatic breast cancers is mediated by stromal microvesicles. Cancer Res. 2017, 77, 1927–1941. [Google Scholar] [CrossRef] [Green Version]
  88. Carr, J.A.; Havstad, S.; Zarbo, R.J.; Divine, G.; Mackowiak, P.; Velanovich, V. The association of HER-2/neu amplification with breast cancer recurrence. Arch. Surg. 2000, 135, 1469–1474. [Google Scholar] [CrossRef] [Green Version]
  89. Wang, J.; Xu, B. Targeted therapeutic options and future perspectives for HER2-positive breast cancer. Signal Transduct. Target. Ther. 2019, 4, 1–22. [Google Scholar] [CrossRef] [Green Version]
  90. Baselga, J.; Swain, S.M. Novel anticancer targets: Revisiting ERBB2 and discovering ERBB3. Nat. Rev. Cancer 2009, 9, 463–475. [Google Scholar] [CrossRef]
  91. Campone, M.; Berton-Rigaud, D.; Bourbouloux, E.; Sophie, S.; Zanetti, A.; Frenel, J.-S. Her2 positive breast cancer: Practices. Bull. Cancer 2011, 98, 154–163. [Google Scholar] [CrossRef] [PubMed]
  92. Gomez, H.L.; Doval, D.C.; Chavez, M.A.; Ang, P.C.-S.; Aziz, Z.; Nag, S.; Ng, C.; Franco, S.X.; Chow, L.W.; Arbushites, M.C.; et al. Efficacy and safety of lapatinib as first-line therapy for ErbB2-amplified locally advanced or metastatic breast cancer. J. Clin. Oncol. 2008, 26, 2999–3005. [Google Scholar] [CrossRef] [PubMed]
  93. Pinto, A.C.; Ades, F.; de Azambuja, E.; Piccart-Gebhart, M. Trastuzumab for patients with HER2 positive breast cancer: Delivery, duration and combination therapies. Breast 2013, 22, S152–S155. [Google Scholar] [CrossRef] [PubMed]
  94. Zheng, D.; Wan, C.; Yang, H.; Xu, L.; Dong, Q.; Du, C.; Du, J.; Li, F. Her2-targeted multifunctional nano-theranostic platform mediates tumor microenvironment remodeling and immune activation for breast cancer treatment. Int. J. Nanomed. 2020, 15, 10007–10028. [Google Scholar] [CrossRef]
  95. Nguyen, M.; de Ninno, A.; Mencattini, A.; Mermet-Meillon, F.; Fornabaio, G.; Evans, S.S.; Cossutta, M.; Khira, Y.; Han, W.; Sirven, P.; et al. Dissecting effects of anti-cancer drugs and cancer-associated fibroblasts by on-chip reconstitution of immunocompetent tumor microenvironments. Cell Rep. 2018, 25, 3884–3893.e3. [Google Scholar] [CrossRef] [Green Version]
  96. Marusyk, A.; Tabassum, D.P.; Janiszewska, M.; Place, A.E.; Trinh, A.; Rozhok, A.I.; Pyne, S.; Guerriero, J.L.; Shu, S.; Ekram, M.; et al. Spatial proximity to fibroblasts impacts molecular features and therapeutic sensitivity of breast cancer cells influencing clinical outcomes. Cancer Res. 2016, 76, 6495–6506. [Google Scholar] [CrossRef] [Green Version]
  97. Lin, C.-H.; Pelissier, F.A.; Zhang, H.; Lakins, J.; Weaver, V.M.; Park, C.; LaBarge, M.A. Microenvironment rigidity modulates responses to the HER2 receptor tyrosine kinase inhibitor lapatinib via YAP and TAZ transcription factors. Mol. Biol. Cell 2015, 26, 3946–3953. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  98. Infante, J.R.; Korn, R.L.; Rosen, L.S.; Lorusso, P.; Dychter, S.S.; Zhu, J.; Maneval, D.C.; Jiang, P.; Shepard, H.M.; Frost, G.; et al. Phase 1 trials of PEGylated recombinant human hyaluronidase PH20 in patients with advanced solid tumours. Br. J. Cancer 2018, 118, 153–161. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  99. Fernandez-Nogueira, P.; Mancino, M.; Fuster, G.; Lopez-Plana, A.; Jauregui, P.; Almendro, V.; Enreig, E.; Menéndez, S.; Rojo, F.; Noguera-Castells, A.; et al. Tumor-associated fibroblasts promote HER2-targeted therapy resistance through FGFR2 Activation. Clin. Cancer Res. 2019, 26, 1432–1448. [Google Scholar] [CrossRef] [Green Version]
  100. Akhand, S.S.; Chen, H.; Purdy, S.C.; Liu, Z.; Anderson, J.C.; Willey, C.D.; Wendt, M.K. Fibroblast growth factor receptor facilitates recurrence of minimal residual disease following trastuzumab emtansine therapy. NPJ Breast Cancer 2021, 7, 1–11. [Google Scholar] [CrossRef]
  101. Hanker, A.; Garrett, J.J.; Estrada, M.V.; Moore, P.P.; Ericsson, P.G.P.; Koch, J.J.; Langley, E.E.; Singh, S.S.; Kim, P.P.; Frampton, G.G.; et al. HER2-overexpressing breast cancers amplify FGFR signaling upon acquisition of resistance to dual therapeutic blockade of HER2. Clin. Cancer Res. 2017, 23, 4323–4334. [Google Scholar] [CrossRef] [Green Version]
  102. Lin, C.-H.; Lee, J.K.; LaBarge, M.A. Fabrication and Use of MicroEnvironment microArrays (MEArrays). J. Vis. Exp. 2012, 2012, e4152. [Google Scholar] [CrossRef] [PubMed]
  103. Watson, S.S.; Dane, M.; Chin, K.; Tatarova, Z.; Liu, M.; Liby, T.; Thompson, W.; Smith, R.; Nederlof, M.; Bucher, E.; et al. Microenvironment-mediated mechanisms of resistance to HER2 inhibitors differ between HER2+ breast cancer subtypes. Cell Syst. 2018, 6, 329–342. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  104. Wilson, T.R.; Fridlyand, J.; Yan, Y.; Penuel, E.; Burton, L.; Chan, E.; Peng, J.; Lin, E.; Wang, Y.; Sosman, J.; et al. Widespread potential for growth-factor-driven resistance to anticancer kinase inhibitors. Nat. Cell Biol. 2012, 487, 505–509. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  105. Mao, Y.; Zhang, Y.; Zhao, M.; Shen, K.; Qu, Q.; Lou, Y.; Liu, J.; Huang, O.; Chen, X.; Wu, J. Cancer-associated fibroblasts induce trastuzumab resistance in HER2 positive breast cancer cells. Mol. BioSyst. 2015, 11, 1029–1040. [Google Scholar] [CrossRef]
  106. Zervantonakis, I.K.; Poskus, M.D.; Scott, A.L.; Selfors, L.M.; Lin, J.-R.; Dillon, D.A.; Pathania, S.; Sorger, P.K.; Mills, G.B.; Brugge, J.S. Fibroblast–tumor cell signaling limits HER2 kinase therapy response via activation of MTOR and antiapoptotic pathways. Proc. Natl. Acad. Sci. USA 2020, 117, 16500–16508. [Google Scholar] [CrossRef]
  107. 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]
  108. Hasebe, T. Tumor–stromal interactions in breast tumor progression—Significance of histological heterogeneity of tumor—Stromal fibroblasts. Expert Opin. Ther. Targets 2013, 17, 449–460. [Google Scholar] [CrossRef]
  109. Suh, J.; Kim, D.; Lee, Y.; Jang, J.; Surh, Y. Fibroblast growth factor-2, derived from cancer-associated fibroblasts, stimulates growth and progression of human breast cancer cells via FGFR1 signaling. Mol. Carcinog. 2020, 59, 1028–1040. [Google Scholar] [CrossRef]
  110. Matà, R.; Palladino, C.; Nicolosi, M.L.; Presti, A.R.L.; Malaguarnera, R.; Ragusa, M.; Sciortino, D.; Morrione, A.; Maggiolini, M.; Vella, V.; et al. IGF-I induces upregulation of DDR1 collagen receptor in breast cancer cells by suppressing MIR-199a-5p through the PI3K/AKT pathway. Oncotarget 2015, 7, 7683–7700. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  111. Orimo, A.; Gupta, P.B.; Sgroi, D.C.; Arenzana-Seisdedos, F.; Delaunay, T.; Naeem, R.; Carey, V.J.; Richardson, A.L.; Weinberg, R.A. Stromal fibroblasts present in invasive human breast carcinomas promote tumor growth and angiogenesis through elevated SDF-1/CXCL12 secretion. Cell 2005, 121, 335–348. [Google Scholar] [CrossRef] [PubMed]
  112. Dvorak, K.M.; Pettee, K.M.; Rubinic-Minotti, K.; Su, R.; Nestor-Kalinoski, A.; Eisenmann, K.M. Carcinoma associated fibroblasts (CAFs) promote breast cancer motility by suppressing mammalian Diaphanous-related formin-2 (mDia2). PLoS ONE 2018, 13, e0195278. [Google Scholar] [CrossRef] [PubMed]
  113. Farmaki, E.; Chatzistamou, I.; Kaza, V.; Kiaris, H. A CCL8 gradient drives breast cancer cell dissemination. Oncogene 2016, 35, 6309–6318. [Google Scholar] [CrossRef]
  114. 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]
  115. Liao, D.; Luo, Y.; Markowitz, D.; Xiang, R.; Reisfeld, R.A. Cancer associated fibroblasts promote tumor growth and metastasis by modulating the tumor immune microenvironment in a 4T1 murine breast cancer model. PLoS ONE 2009, 4, e7965. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  116. Acharyya, S.; Oskarsson, T.; Vanharanta, S.; Malladi, S.; Kim, J.; Morris, P.G.; Manova-Todorova, K.; Leversha, M.; Hogg, N.; Seshan, V.E.; et al. A CXCL1 paracrine network links cancer chemoresistance and metastasis. Cell 2012, 150, 165–178. [Google Scholar] [CrossRef] [Green Version]
  117. Öhlund, D.; Elyada, E.; Tuveson, D. Fibroblast heterogeneity in the cancer wound. J. Exp. Med. 2014, 211, 1503–1523. [Google Scholar] [CrossRef]
  118. Lee, K.W.; Cho, J.A.; Park, H.; Lim, E.H. Exosomes from breast cancer cells can convert adipose tissue-derived mesenchymal stem cells into myofibroblast-like cells. Int. J. Oncol. 2011, 40, 130–138. [Google Scholar] [CrossRef] [Green Version]
  119. Wen, S.; Hou, Y.; Fu, L.; Xi, L.; Yang, D.; Zhao, M.; Qin, Y.; Sun, K.; Teng, Y.; Liu, M. Cancer-associated fibroblast (CAF)-derived IL32 promotes breast cancer cell invasion and metastasis via integrin β3–p38 MAPK signalling. Cancer Lett. 2019, 442, 320–333. [Google Scholar] [CrossRef]
  120. Giorello, M.B.; Borzone, F.R.; Labovsky, V.; Piccioni, F.V.; Chasseing, N.A. Cancer-associated fibroblasts in the breast tumor microenvironment. J. Mammary Gland. Biol. Neoplasia 2021, 1–21. [Google Scholar] [CrossRef]
  121. Zhang, X.H.-F.; Jin, X.; Malladi, S.; Zou, Y.; Wen, Y.H.; Brogi, E.; Smid, M.; Foekens, J.A.; Massagué, J. Selection of bone metastasis seeds by mesenchymal signals in the primary tumor stroma. Cell 2013, 154, 1060–1073. [Google Scholar] [CrossRef] [Green Version]
  122. Fernando, R.I.; Castillo, M.D.; Litzinger, M.; Hamilton, D.H.; Palena, C. IL-8 signaling plays a critical role in the epithelial–mesenchymal transition of human carcinoma cells. Cancer Res. 2011, 71, 5296–5306. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  123. Nam, J.-S.; Kang, M.-J.; Suchar, A.M.; Shimamura, T.; Kohn, E.A.; Michalowska, A.M.; Jordan, V.C.; Hirohashi, S.; Wakefield, L.M. Chemokine (C-C Motif) Ligand 2 mediates the prometastatic effect of dysadherin in human breast cancer cells. Cancer Res. 2006, 66, 7176–7184. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  124. Yasuhara, R.; Irié, T.; Suzuki, K.; Sawada, T.; Miwa, N.; Sasaki, A.; Tsunoda, Y.; Nakamura, S.; Mishima, K. The β-catenin signaling pathway induces aggressive potential in breast cancer by up-regulating the chemokine CCL5. Exp. Cell Res. 2015, 338, 22–31. [Google Scholar] [CrossRef]
  125. Chen, J.-Q.; Russo, J. ERα-negative and triple negative breast cancer: Molecular features and potential therapeutic approaches. Biochim. Biophys. Acta Bioenerg. 2009, 1796, 162–175. [Google Scholar] [CrossRef] [Green Version]
  126. Hartman, Z.C.; Poage, G.M.; Den Hollander, P.; Tsimelzon, A.; Hill, J.; Panupinthu, N.; Zhang, Y.; Mazumdar, A.; Hilsenbeck, S.G.; Mills, G.B.; et al. Growth of triple-negative breast cancer cells relies upon coordinate autocrine expression of the proinflammatory Cytokines IL-6 and IL-8. Cancer Res. 2013, 73, 3470–3480. [Google Scholar] [CrossRef] [Green Version]
  127. Lv, D.; Zhang, Y.; Kim, H.-J.; Zhang, L.; Ma, X. CCL5 as a potential immunotherapeutic target in triple-negative breast cancer. Cell. Mol. Immunol. 2013, 10, 303–310. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  128. Andò, S.; Catalano, S. The multifactorial role of leptin in driving the breast cancer microenvironment. Nat. Rev. Endocrinol. 2011, 8, 263–275. [Google Scholar] [CrossRef]
  129. Giordano, C.; Chemi, F.; Panza, S.; Barone, I.; Bonofiglio, D.; Lanzino, M.; Cordella, A.; Campana, A.; Hashim, A.; Rizza, P.; et al. Leptin as a mediator of tumor-stromal interactions promotes breast cancer stem cell activity. Oncotarget 2015, 7, 1262–1275. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  130. Giordano, C.; Barone, I.; Vircillo, V.; Panza, S.; Malivindi, R.; Gelsomino, L.; Pellegrino, M.; Rago, V.; Mauro, L.; Lanzino, M.; et al. Activated FXR inhibits leptin signaling and counteracts tumor-promoting activities of cancer-associated fibroblasts in breast malignancy. Sci. Rep. 2016, 6, 21782. [Google Scholar] [CrossRef] [PubMed]
  131. 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, 1–10. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  132. Luga, V.; Wrana, J.L. Tumor–stroma interaction: Revealing fibroblast-secreted exosomes as potent regulators of wnt-planar cell polarity signaling in cancer metastasis. Cancer Res. 2013, 73, 6843–6847. [Google Scholar] [CrossRef] [Green Version]
  133. 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]
  134. 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] [PubMed]
  135. Donnarumma, E.; Fiore, D.; Nappa, M.; Roscigno, G.; Adamo, A.; Iaboni, M.; Russo, V.; Affinito, A.; Puoti, I.; Quintavalle, C.; et al. Cancer-associated fibroblasts release exosomal microRNAs that dictate an aggressive phenotype in breast cancer. Oncotarget 2017, 8, 19592–19608. [Google Scholar] [CrossRef] [Green Version]
  136. Dumont, N.; Liu, B.; DeFilippis, R.A.; Chang, H.; Rabban, J.T.; Karnezis, A.N.; Tjoe, J.A.; Marx, J.; Parvin, B.; Tlsty, T.D. Breast fibroblasts modulate early dissemination, tumorigenesis, and metastasis through alteration of extracellular matrix characteristics. Neoplasia 2013, 15, 249–262. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  137. Ren, Y.; Jia, H.-H.; Xu, Y.-Q.; Zhou, X.; Zhao, X.-H.; Wang, Y.; Song, X.; Zhu, Z.-Y.; Sun, T.; Dou, Y.; et al. Paracrine and epigenetic control of CAF-induced metastasis: The role of HOTAIR stimulated by TGF-ß1 secretion. Mol. Cancer 2018, 17, 1–14. [Google Scholar] [CrossRef] [PubMed]
  138. Xuejun, T.; Tian, X.; Mohammad, M.; Movassaghi, M.; Naber, S.P.; Kuperwasser, C.; Buchsbaum, R.J. The fibroblast Tiam1-osteopontin pathway modulates breast cancer invasion and metastasis. Breast Cancer Res. 2016, 18, 1–15. [Google Scholar]
  139. Wang, M.; Zhang, J.; Huang, Y.; Ji, S.; Shao, G.; Feng, S.; Chen, D.; Zhao, K.; Wang, Z.; Wu, A. Cancer-associated fibroblasts autophagy enhances progression of triple-negative breast cancer cells. Med Sci. Monit. 2017, 23, 3904–3912. [Google Scholar] [CrossRef] [Green Version]
  140. Huang, M.; Fu, M.; Wang, J.; Xia, C.; Zhang, H.; Xiong, Y.; He, J.; Liu, J.; Liu, B.; Pan, S.; et al. TGF-β1-activated cancer-associated fibroblasts promote breast cancer invasion, metastasis and epithelial-mesenchymal transition by autophagy or overexpression of FAP-α. Biochem. Pharmacol. 2021, 188, 114527. [Google Scholar] [CrossRef]
  141. Gagliano, T.; Shah, K.; Gargani, S.; Lao, L.; Alsaleem, M.; Chen, J.; Ntafis, V.; Huang, P.; Ditsiou, A.; Vella, V.; et al. PIK3Cδ expression by fibroblasts promotes triple-negative breast cancer progression. J. Clin. Investig. 2020, 130, 3188–3204. [Google Scholar] [CrossRef]
  142. Matsumura, Y.; Ito, Y.; Mezawa, Y.; Sulidan, K.; Daigo, Y.; Hiraga, T.; Mogushi, K.; Wali, N.; Suzuki, H.; Itoh, T.; et al. Stromal fibroblasts induce metastatic tumor cell clusters via epithelial–mesenchymal plasticity. Life Sci. Alliance 2019, 2, e201900425. [Google Scholar] [CrossRef]
  143. Choi, Y.P.; Lee, J.H.; Gao, M.-Q.; Gil Kim, B.; Kang, S.; Kim, S.H.; Cho, N.H. Cancer-associated fibroblast promote transmigration through endothelial brain cells in three-dimensionalin vitromodels. Int. J. Cancer 2014, 135, 2024–2033. [Google Scholar] [CrossRef] [PubMed]
  144. Kessenbrock, K.; Plaks, V.; Werb, Z. Matrix metalloproteinases: Regulators of the tumor microenvironment. Cell 2010, 141, 52–67. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  145. Del Casar, J.M.; Alvarez, E.; Junquera, S.; Marin, L.; Bongera, M.; Vazquez, J.; Vizoso, F.J.; González, L.O.; González, L. Comparative analysis and clinical value of the expression of metalloproteases and their inhibitors by intratumor stromal fibroblasts and those at the invasive front of breast carcinomas. Breast Cancer Res. Treat. 2009, 116, 39–52. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  146. Yang, N.; Mosher, R.; Seo, S.; Beebe, D.; Friedl, A. Syndecan-1 in breast cancer stroma fibroblasts regulates extracellular matrix fiber organization and carcinoma cell motility. Am. J. Pathol. 2011, 178, 325–335. [Google Scholar] [CrossRef] [PubMed]
  147. Ahirwar, D.K.; Nasser, M.W.; Ouseph, M.M.; Elbaz, M.; Cuitiño, 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] [PubMed]
  148. Karagiannis, G.S.; Poutahidis, T.; Erdman, S.E.; Kirsch, R.; Riddell, R.H.; Diamandis, E.P. Cancer-associated fibroblasts drive the progression of metastasis through both paracrine and mechanical pressure on cancer tissue. Mol. Cancer Res. MCR 2012, 10, 1403–1418. [Google Scholar] [CrossRef] [Green Version]
  149. Sosa, M.S.; Bragado, P.; Aguirre-Ghiso, J.A. Mechanisms of disseminated cancer cell dormancy: An awakening field. Nat. Rev. Cancer 2014, 14, 611–622. [Google Scholar] [CrossRef]
  150. Gui, Y.; Aguilar-Mahecha, A.; Krzemien, U.; Hosein, A.; Buchanan, M.; LaFleur, J.; Pollak, M.N.; Ferrario, C.; Basik, M. Metastatic breast carcinoma–Associated fibroblasts have enhanced protumorigenic properties related to increased IGF2 Expression. Clin. Cancer Res. 2019, 25, 7229–7242. [Google Scholar] [CrossRef] [Green Version]
  151. Weigelt, B.; Peterse, J.L.; van ’t Veer, L.J. Breast cancer metastasis: Markers and models. Nat. Rev. Cancer 2005, 5, 591–602. [Google Scholar] [CrossRef]
  152. Malanchi, I.; Santamaria-Martínez, A.; Susanto, E.; Peng, H.; Lehr, H.-A.; Delaloye, J.-F.; Huelsken, J. Interactions between cancer stem cells and their niche govern metastatic colonization. Nat. Cell Biol. 2011, 481, 85–89. [Google Scholar] [CrossRef] [PubMed]
  153. Peinado, H.; Zhang, H.; Matei, I.R.; Costa-Silva, B.; Hoshino, A.; Rodrigues, G.; Psaila, B.; Kaplan, R.N.; Bromberg, J.F.; Kang, Y.; et al. Pre-metastatic niches: Organ-specific homes for metastases. Nat. Rev. Cancer 2017, 17, 302–317. [Google Scholar] [CrossRef] [PubMed]
  154. Hiratsuka, S.; Watanabe, A.; Aburatani, H.; Maru, Y. Tumour-mediated upregulation of chemoattractants and recruitment of myeloid cells predetermines lung metastasis. Nat. Cell Biol. 2006, 8, 1369–1375. [Google Scholar] [CrossRef]
  155. Raz, Y.; Cohen, N.; Shani, O.; Bell, R.E.; Novitskiy, S.V.; Abramovitz, L.; Levy, C.; Milyavsky, M.; Leider-Trejo, L.; Moses, H.L.; et al. Bone marrow–derived fibroblasts are a functionally distinct stromal cell population in breast cancer. J. Exp. Med. 2018, 215, 3075–3093. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  156. Hoshino, A.; Costa-Silva, B.; Shen, T.-L.; Rodrigues, G.; Hashimoto, A.; Mark, M.T.; Molina, H.; Kohsaka, S.; Di Giannatale, A.; Ceder, S.; et al. Tumour exosome integrins determine organotropic metastasis. Nature 2015, 527, 329–335. [Google Scholar] [CrossRef] [Green Version]
  157. Fong, M.Y.; Zhou, W.; Liu, L.; Alontaga, A.Y.; Chandra, M.; Ashby, J.; Chow, A.; O’Connor, S.T.F.; Li, S.; Chin, A.R.; et al. Breast-cancer-secreted miR-122 reprograms glucose metabolism in premetastatic niche to promote metastasis. Nat. Cell Biol. 2015, 17, 183–194. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  158. Feng, T.; Zhang, P.; Sun, Y.; Wang, Y.; Tong, J.; Dai, H.; Hua, Z. High throughput sequencing identifies breast cancer-secreted exosomal LncRNAs initiating pulmonary pre-metastatic niche formation. Gene 2019, 710, 258–264. [Google Scholar] [CrossRef] [PubMed]
  159. Medeiros, B.; Allan, A.L. Molecular mechanisms of breast cancer metastasis to the lung: Clinical and experimental perspectives. Int. J. Mol. Sci. 2019, 20, 2272. [Google Scholar] [CrossRef] [Green Version]
  160. Qi, F.; He, T.; Jia, L.; Song, N.; Guo, L.; Ma, X.; Wang, C.; Xuhui, M.; Fu, Y.; Lin, J.; et al. The miR-30 family inhibits pulmonary vascular hyperpermeability in the premetastatic phase by direct targeting of Skp2. Clin. Cancer Res. 2015, 21, 3071–3080. [Google Scholar] [CrossRef] [Green Version]
  161. Canesin, G.; Cuevas, E.P.; Santos, V.; López-Menéndez, C.; Moreno-Bueno, G.; Huang, Y.; Csiszar, K.; Portillo, F.; Peinado, H.; Lyden, D.; et al. Lysyl oxidase-like 2 (LOXL2) and E47 EMT factor: Novel partners in E-cadherin repression and early metastasis colonization. Oncogene 2014, 34, 951–964. [Google Scholar] [CrossRef] [Green Version]
  162. Oskarsson, T.; Acharyya, S.; Zhang, X.H.-F.; Vanharanta, S.; Tavazoie, S.F.; Morris, P.G.; Downey, R.J.; Manova-Todorova, K.; Brogi, E.; Massague, J. Breast cancer cells produce tenascin C as a metastatic niche component to colonize the lungs. Nat. Med. 2011, 17, 867–874. [Google Scholar] [CrossRef] [Green Version]
  163. Wang, Z.; Xiong, S.; Mao, Y.; Chen, M.; Ma, X.; Zhou, X.; Ma, Z.; Liu, F.; Huang, Z.; Luo, Q.; et al. Periostin promotes immunosuppressive premetastatic niche formation to facilitate breast tumour metastasis. J. Pathol. 2016, 239, 484–495. [Google Scholar] [CrossRef]
  164. Bonnans, C.; Chou, J.; Werb, Z. Remodelling the extracellular matrix in development and disease. Nat. Rev. Mol. Cell Biol. 2014, 15, 786–801. [Google Scholar] [CrossRef] [PubMed]
  165. Martin, Y.D.P.; Park, D.; Ramachandran, A.; Ombrato, L.; Calvo, F.; Chakravarty, P.; Spencer-Dene, B.; Derzsi, S.; Hill, C.; Sahai, E.; et al. Mesenchymal cancer cell-stroma crosstalk promotes niche activation, epithelial reversion, and metastatic colonization. Cell Rep. 2015, 13, 2456–2469. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  166. Shu, C.; Zha, H.; Long, H.; Wang, X.; Yang, F.; Gao, J.; Hu, C.; Zhou, L.; Guo, B.; Zhu, B. C3a-C3aR signaling promotes breast cancer lung metastasis via modulating carcinoma associated fibroblasts. J. Exp. Clin. Cancer Res. 2020, 39, 1–14. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  167. Chen, F.; Han, Y.; Kang, Y. Bone marrow niches in the regulation of bone metastasis. Br. J. Cancer 2021, 124, 1912–1920. [Google Scholar] [CrossRef]
  168. Kang, Y.; Siegel, P.M.; Shu, W.; Drobnjak, M.; Kakonen, S.M.; Cordón-Cardo, C.; Guise, T.A.; Massagué, J. A multigenic program mediating breast cancer metastasis to bone. Cancer Cell 2003, 3, 537–549. [Google Scholar] [CrossRef] [Green Version]
  169. Kuo, M.C.; Kothari, A.N.; Kuo, P.C.; Mi, Z. Cancer stemness in bone marrow micrometastases of human breast cancer. Surgery 2018, 163, 330–335. [Google Scholar] [CrossRef]
  170. Sasaki, S.; Baba, T.; Nishimura, T.; Hayakawa, Y.; Hashimoto, S.-I.; Gotoh, N.; Mukaida, N. Essential roles of the interaction between cancer cell-derived chemokine, CCL4, and intra-bone CCR5-expressing fibroblasts in breast cancer bone metastasis. Cancer Lett. 2016, 378, 23–32. [Google Scholar] [CrossRef] [Green Version]
  171. Mi, Z.; Bhattacharya, S.D.; Kim, V.M.; Guo, H.; Talbot, L.J.; Kuo, P.C. Osteopontin promotes CCL5-mesenchymal stromal cell-mediated breast cancer metastasis. Carcinogenesis 2011, 32, 477–487. [Google Scholar] [CrossRef]
  172. Hauge, A.; Rofstad, E.K. Antifibrotic therapy to normalize the tumor microenvironment. J. Transl. Med. 2020, 18, 1–11. [Google Scholar] [CrossRef] [PubMed]
  173. Singha, N.C.; Nekoroski, T.; Zhao, C.; Symons, R.; Jiang, P.; Frost, G.I.; Huang, Z.; Shepard, H.M.; Goglia, A.G.; Delsite, R.; et al. Tumor-associated hyaluronan limits efficacy of monoclonal antibody therapy. Mol. Cancer Ther. 2015, 14, 523–532. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  174. Elkabets, M.; Gifford, A.M.; Scheel, C.; Nilsson, B.; Reinhardt, F.; Bray, M.-A.; Carpenter, A.; Jirström, K.; Magnusson, K.; Ebert, B.L.; et al. Human tumors instigate granulin-expressing hematopoietic cells that promote malignancy by activating stromal fibroblasts in mice. J. Clin. Investig. 2011, 121, 784–799. [Google Scholar] [CrossRef] [PubMed]
  175. Chronopoulos, A.; Robinson, B.; Sarper, M.; Cortes, E.; Auernheimer, V.; Lachowski, D.; Attwood, S.; García, R.; Ghassemi, S.; Fabry, B.; et al. ATRA mechanically reprograms pancreatic stellate cells to suppress matrix remodelling and inhibit cancer cell invasion. Nat. Commun. 2016, 7, 12630. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  176. Al-Qassab, Y.; Grassilli, S.; Brugnoli, F.; Vezzali, F.; Capitani, S.; Bertagnolo, V. Protective role of all-trans retinoic acid (ATRA) against hypoxia-induced malignant potential of non-invasive breast tumor derived cells. BMC Cancer 2018, 18, 1194. [Google Scholar] [CrossRef] [PubMed]
  177. Liu, J.; Liao, S.; Diop-Frimpong, B.; Chen, W.; Goel, S.; Naxerova, K.; Ancukiewicz, M.; Boucher, Y.; Jain, R.K.; Xu, L. TGF- blockade improves the distribution and efficacy of therapeutics in breast carcinoma by normalizing the tumor stroma. Proc. Natl. Acad. Sci. USA 2012, 109, 16618–16623. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  178. Chauhan, V.P.; Martin, J.D.; Liu, H.; Lacorre, D.A.; Jain, S.R.; Kozin, S.V.; Stylianopoulos, T.; Mousa, A.S.; Han, X.; Adstamongkonkul, P.; et al. Angiotensin inhibition enhances drug delivery and potentiates chemotherapy by decompressing tumour blood vessels. Nat. Commun. 2013, 4, 2516. [Google Scholar] [CrossRef] [Green Version]
  179. Olive, K.P.; Jacobetz, M.A.; Davidson, C.J.; Gopinathan, A.; McIntyre, D.; Honess, D.; Madhu, B.; Goldgraben, M.A.; Caldwell, M.E.; Allard, D.; et al. Inhibition of hedgehog signaling enhances delivery of chemotherapy in a mouse model of pancreatic cancer. Science 2009, 324, 1457–1461. [Google Scholar] [CrossRef] [Green Version]
  180. Wendt, M.K.; Schiemann, W.P. Therapeutic targeting of the focal adhesion complex prevents oncogenic TGF-β signaling and metastasis. Breast Cancer Res. 2009, 11, R68. [Google Scholar] [CrossRef] [Green Version]
  181. Van der Spek, Y.M.; Kroep, J.R.; Tollenaar, R.A.E.M.; Mesker, W.E. Chemotherapy resistance and stromal targets in breast cancer treatment: A review. Mol. Biol. Rep. 2020, 47, 8169–8177. [Google Scholar] [CrossRef]
  182. Gascard, P.; Tlsty, T.D. Carcinoma-associated fibroblasts: Orchestrating the composition of malignancy. Genes Dev. 2016, 30, 1002–1019. [Google Scholar] [CrossRef] [PubMed]
  183. Catalano, S.; Panza, S.; Augimeri, G.; Giordano, F.; Malivindi, R.; Gelsomino, L.; Marsico, S.; Győrffy, B.; Bonofiglio, D.; Andò, S.; et al. Phosphodiesterase 5 (PDE5) is Highly Expressed in Cancer-Associated Fibroblasts and Enhances Breast Tumor Progression. Cancers 2019, 11, 1740. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  184. Pantziarka, P.; Sukhatme, V.; Crispino, S.; Bouche, G.; Meheus, L.; Sukhatme, V.P. Repurposing drugs in oncology (ReDO)—Selective PDE5 inhibitors as. Ecancermedicalscience 2018, 12, 824. [Google Scholar] [CrossRef] [PubMed] [Green Version]
Figure 1. Tumor/stroma communication in ductal carcinoma in situ (DCIS) lesions (left) and tumor/stroma crosstalk in invasive breast cancer (right) favoring the resistant phenotype of breast cancer cells under radiotherapy, chemotherapy, hormonal and targeted therapy treatments. The main contributors to each situation are listed. For a list of all proteins names abbreviations please see Table S2.
Figure 1. Tumor/stroma communication in ductal carcinoma in situ (DCIS) lesions (left) and tumor/stroma crosstalk in invasive breast cancer (right) favoring the resistant phenotype of breast cancer cells under radiotherapy, chemotherapy, hormonal and targeted therapy treatments. The main contributors to each situation are listed. For a list of all proteins names abbreviations please see Table S2.
Cancers 13 03146 g001
Figure 2. Pro-metastatic CAFs-secreted factors at the primary tumor promote tumor cell migration, invasion, and dissemination. For a list of all proteins names abbreviations please see Table S2.
Figure 2. Pro-metastatic CAFs-secreted factors at the primary tumor promote tumor cell migration, invasion, and dissemination. For a list of all proteins names abbreviations please see Table S2.
Cancers 13 03146 g002
Figure 3. CAFs-secreted factors foster breast cancer cells’ dissemination to major secondary organs, such as lungs, liver and bone. For a list of all proteins names abbreviations please see Table S2.
Figure 3. CAFs-secreted factors foster breast cancer cells’ dissemination to major secondary organs, such as lungs, liver and bone. For a list of all proteins names abbreviations please see Table S2.
Cancers 13 03146 g003
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Fernández-Nogueira, P.; Fuster, G.; Gutierrez-Uzquiza, Á.; Gascón, P.; Carbó, N.; Bragado, P. Cancer-Associated Fibroblasts in Breast Cancer Treatment Response and Metastasis. Cancers 2021, 13, 3146. https://doi.org/10.3390/cancers13133146

AMA Style

Fernández-Nogueira P, Fuster G, Gutierrez-Uzquiza Á, Gascón P, Carbó N, Bragado P. Cancer-Associated Fibroblasts in Breast Cancer Treatment Response and Metastasis. Cancers. 2021; 13(13):3146. https://doi.org/10.3390/cancers13133146

Chicago/Turabian Style

Fernández-Nogueira, Patricia, Gemma Fuster, Álvaro Gutierrez-Uzquiza, Pere Gascón, Neus Carbó, and Paloma Bragado. 2021. "Cancer-Associated Fibroblasts in Breast Cancer Treatment Response and Metastasis" Cancers 13, no. 13: 3146. https://doi.org/10.3390/cancers13133146

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