You are viewing the site in preview mode

Skip to main content

Histone acetylation modulators in breast cancer

Abstract

Breast cancer is the most prevalent cancer in women worldwide. Aberrant epigenetic reprogramming such as dysregulation of histone acetylation has been associated with the development of breast cancer. Histone acetylation modulators have been targeted as potential treatments for breast cancer. This review comprehensively discusses the roles of these modulators and the effects of their inhibitors on breast cancer. In addition, epigenetic reprogramming not only affects breast cancer cells but also the immunosuppressive myeloid cells, which can facilitate breast cancer progression. Therefore, the review also highlights the roles of these immunosuppressive myeloid cells and summarizes how histone acetylation modulators affect their functions and phenotypes. This review provides insights into histone acetylation modulators as potential therapeutic targets for breast cancer.

Background

Breast cancer is the most common cancer among women globally [1]. Breast cancer can be classified by the expression of estrogen receptor (ER) and progesterone receptor (PR) and amplification of human epidermal growth factor receptor 2 (HER2) [2]. Breast cancer that lacks those biomarkers is categorized as triple-negative breast cancer (TNBC) [3, 4].

Epigenetic changes contribute to tumorigenesis, progression, and metastasis of breast cancer [5]. They can also affect the tumor-associated immune cells, which play important roles in tumor growth and treatment response [5, 6]. Therefore, many therapeutics have been developed to target epigenetic factors in breast cancer [7]. Histone acetylation is one of the most important epigenetic modifications. Histones are critical components of nucleosomes. Each nucleosome contains two subunits made of H3, H4, H2A, and H2B histones. Each histone contains a tail enriched with lysine (K) residues, which can be acetylation sites. Acetylation of histone tails can increase chromatin accessibility at the enhancer, promoter, and transcribed regions and thus promote gene transcription [8] (Table 1). The “writers” of histone acetylation are histone acetyltransferases (HATs) categorized into four major families Gcn5-related N-acetyltransferases (GNATs), MYST, CREB-binding protein (CBP)/E1A-associated protein p300 (EP300) and steroid receptor coactivators (SRCs) [9, 10]. Histone acetylation is removed by histone deacetylases (HDACs), which include classical HDACs and sirtuins with different cellular localization (Table 2) [11]. Histone acetylation also acts as a signal recognized by “readers” bromodomains (BRDs), and many chromatin-modulating proteins including HATs can contain BRDs [12]. In addition, both HDACs and HATs can have non-histone targets such as transcription factors (Tables 1 and 2).

Inhibitors of both writers and erasers of histone acetylation have been investigated as potential therapeutics for breast cancer [13]. However, very few published reviews have an in-depth focus on these modulators. Therefore, here we provide a comprehensive review of current findings on histone acetylation modulators and their inhibitors in breast cancer. In addition, we also summarize the roles of tumor-promoting myeloid cells in breast cancer and discuss how they can also be regulated by the histone acetylation modulators.

HATs in breast cancer

HATs have been reported as both oncogenes and tumor suppressors in many cancer types including breast cancer [14, 15]. Histone H4K8 acetylation by KAT2B, a GNAT family HAT, reduced replication fork stability in breast cancer cells in vitro, and reduced levels of KAT2B may predict PARP inhibitor resistance [16]. KAT2B also inhibits proliferation of p53 mutant breast cancer cells in vitro by acetylating p53 and histones [17]. KAT5, a HAT of the MYST family, has been identified as a haploinsufficient tumor suppressor, and loss or low expression of KAT5 was observed in a fraction of breast cancer cases, correlating with poor prognosis [18, 19]. Another study showed that low expression of KAT5 led to decreased H3K4 acetylation and knockdown of KAT5 promoted the progression of MDA-MB-231 xenografts, a TNBC model, but not MCF-7 xenografts, an ER-positive breast cancer model [20]. This indicates that the role of KAT5 in breast cancer is complex and context dependent.

Compared to the tumor-suppressing role, more evidence has been found regarding the tumor-promoting roles of various HATs. Acetyltransferase activity of KAT7 (MYST family) was found to facilitate radiotherapy resistance in breast cancer cells in vitro through activation of the PI3K/AKT pathway [21]. KAT2B, EP300, KAT6A (MYST family), and KAT2A (GNAT family) are recruited to ER-responsive promoters and are critical for estrogen-dependent proliferation of ER-positive breast cancer cells [22,23,24]. KAT6A was found to be frequently amplified and/or overexpressed in breast cancer and has been correlated with worse prognosis in ER-positive breast cancer patients [24, 25]. Moreover, the silencing of ATF2 (GNAT family) reduced the expression of genes associated with endocrine therapy resistance in ER-positive breast cancer cells in vitro [26]. The SRC family HATs are transcription coactivators for steroid hormone receptors including ER and PR and can acetylate steroid hormone receptor-responsive promoters [27,28,29,30]. SRC-1 and SRC-3 also facilitate endocrine therapy resistance and activate breast cancer-promoting genes in an ER-independent manner [27, 31]. In addition, CBP/EP300 also activates transcription of the androgen receptor (AR) and thus promotes AR signaling in AR-positive breast cancer model MDA-MB-453 in vitro and in vivo [32].

Besides activating the transcription of hormone receptor-responsive genes, HATs can also promote epithelial-mesenchymal transition (EMT). EMT describes a process in which epithelial cells lose their polarity and junctions to gain mesenchymal traits [33]. It is associated with breast cancer invasion, migration, metastasis, and stem-cell-like phenotypes [34]. In TNBC, enrichment of the EMT gene signature was found in residual tumors after neoadjuvant chemotherapy [35]. Multiple HATs were found to be involved in EMT activation. KAT2A induced EMT in breast cancer cells by activating the transforming growth factor-β (TGF-β)/Smad pathway, and inhibition of KAT2A reduced the survival, migration, and invasion of MDA-MB-231 cells in vitro [36]. KAT5 was shown to acetylate the key EMT-inducing transcription factor (EMT-TF) Twist to promote transcription of EMT genes in vitro in basal-like breast cancer cells HEK293 and SUM1315 [37]. In addition, EP300 induces the expression of key EMT regulators in non-tumorigenic breast epithelial cells MCF10A by histone H3 acetylation and interacting with other transcription factors such as c-Myc [38].

In summary, the functions of HATs have been primarily studied in hormone receptor-positive breast cancer, as HATs were known to regulate the transcription of hormone receptor-dependent genes (Fig. 1). In contrast, studies investigating other roles of HATs were mostly conducted in breast cancer cell lines in vitro. More in vivo studies will be needed to further elucidate the roles of HATs in breast cancer, especially the hormone receptor-independent subtypes.

Table 1 The targets and functions of histone acetyltransferases (HATs)
Fig. 1
figure 1

Protumor and antitumor functions of HATs and HDACs in breast cancer

HAT inhibitors in breast cancer

HAT inhibitors have been developed and shown to have antitumor efficacy in many cancer types including breast cancer. Most current studies focus on inhibitors of CBP/EP300 and KAT6A/KAT6B, MYST family HATs [48].

CBP/EP300 HAT inhibitors CPI-1612 and A-485 were shown to inhibit the growth of ER-positive breast cancer in vitro and in vivo by reducing ER-dependent gene expression [49, 50]. However, inhibitors of the CBP/EP300 HAT domain were not selective and not successful in the clinic. In contrast, inhibitors of their BRD have recently shown promising results [51]. BRD inhibition can reduce acetylation by the HAT domain, but compared to HAT inhibition, BRD inhibition led to an attenuated effect and decreased acetylation at some unique sites [52]. In ER-positive breast cancer, the CBP/EP300 BRD inhibitor GNE-049 had similar effects as A-485 in downregulating the expression of ER-dependent genes and inhibiting cancer cell proliferation [50]. In TNBC cells, another CBP/EP300 BRD inhibitor I-CBP112 reduced drug efflux by repressing ATP-binding cassette transporters and sensitized the cells to chemotherapies in vitro [53]. The CBP/EP300 BRD inhibitor FT-6876 reduced AR signaling and inhibited the growth of AR-dependent breast cancer models in vitro and in vivo [32]. Our recent publication demonstrated that CBP/EP300 BRD inhibitor IACS-70,654 reduced the proliferation and inhibited the metastasis of neutrophil-enriched TNBC in vivo [54]. However, currently Inobrodib is the only CBP/EP300 BRD inhibitor in early clinical trials for treating solid tumors, including breast cancer [48]. It showed promising results in early clinical trials for hematological malignancies and prostate cancer, but its effects on breast cancer models/patients have not been published [55, 56].

In addition to CBP/EP300 inhibitors, the KAT6A/KAT6B inhibitor CTx-648 demonstrated antitumor activity in vivo in ER-positive breast cancer models with resistance to endocrine therapy [57]. PF-07248144 is the KAT6A/KAT6B inhibitor currently in clinical trials, and the results from the phase 1 clinical trial were recently published and showed durable antitumor effects in metastatic ER-positive HER2-negative breast cancer [58].

Taken together, HAT inhibitors have been tested in breast cancer, and two inhibitors have entered early phase clinical trials. Most studies of HAT inhibitors focused on hormone receptor-positive breast cancer, most likely because the roles of HATs in hormone receptor signaling have been more extensively studied. HAT inhibitors may also be suitable for treating hormone receptor-independent breast cancer, but more preclinical studies might be needed before more inhibitors can enter clinical trials.

HDACs in breast cancer

HDACs have been targeted for the treatment of many cancer types including breast cancer because of their role in many biological functions associated with tumor progression [59,60,61]. In the clinic, metaplastic breast cancer, an aggressive and treatment-resistant subtype, was found to have elevated HDAC activity [62]. Among all HDACS, Class I HDACs were most extensively studied. High expression of HDAC1 has been associated with high ER and PR expression in multiple studies [63,64,65,66]. In contrast, HDAC2 expression was found to be significantly higher in hormone receptor-negative breast tumors [64]. In ER-negative breast cancer, studies suggested that HDAC1 can suppress the expression of ER and its associated genes to promote their growth, indicating its complex functions [67, 68]. HDAC1 was also shown to induce proliferation and migration of breast cancer cells by upregulating Interleukin (IL)-8 signaling [69]. Induced cytoplasmic expression of HDAC3 has been associated with brain metastasis in breast cancer patients [70]. HDAC1, HDAC2, and HDAC8 were found to form a complex with EMT-TF Snail and induce EMT in breast cancer cells to promote migration [71,72,73]. However, HDAC1 was also demonstrated to downregulate Wnt signaling to reduce migration and invasion in breast cancer cells [74]. In addition, HDAC2 and HDAC3 were shown to facilitate the inhibition of vascular endothelial growth factor (VEGF) signaling, which promotes angiogenesis to support tumor progression, in breast cancer cells in vitro [75]. These seemingly contradictory results can be explained by the study illustrating that HDAC1 has distinct substrates in different breast cancer cell lines, highlighting the effects of tumor heterogeneity on HDAC functions [76].

Compared to Class I HDACs, the functions of Class II and IV classic HDACs are not as well characterized, but have also been studied in the breast cancer setting [11]. The loss of HDAC5 induced the expression of cell-cycle genes and thus led to cyclin-dependent kinase (CDK) 4/6 inhibitor resistance in breast cancer [77]. In addition, HDAC5 was shown to deacetylate SOX9 to promote c-Myc expression and help drive endocrine therapy resistance in ER-positive breast cancer in vitro [78]. HDAC6 expression has been correlated with reduced cell motility and better response to endocrine therapy in ER-positive breast cancer cells in vitro [79, 80]. In inflammatory breast cancer, however, HDAC6 was found to have aberrantly high activity and was essential for cell viability [81]. A recent study found induced HDAC6 activity in about 30% of breast cancer patients analyzed and suggested that HDAC6 deacetylates c-Myc to reduce its degradation, contributing to tumor cell viability [82]. Another recent study by Lu et al. demonstrated that phosphorylated HDAC6 induces aberrant chromatin architecture, which supports the tumor growth of TNBC [83]. HDAC11 expression, in contrast, was correlated with better overall survival of breast cancer patients, and HDAC11 knockdown led to enhanced proliferation, migration, and invasion of breast cancer cells in vitro [84]. Nevertheless, another study showed using mouse models that HDAC11 facilitates the growth of breast cancer lymph node metastases while inhibiting the migration from lymph node to distant organs [85].

Besides classic HDACs, sirtuins (SIRT) have also been extensively studied in breast cancer. SIRT1 is overexpressed in ER-positive breast cancer and was shown to promote tumor progression by facilitating ER and estrogen-related receptor signaling [86,87,88]. SIRT1 was also demonstrated to promote breast cancer formation by interacting with and promoting the activity of AKT [89]. In TNBC, however, SIRT1 is downregulated, and the loss of SIRT1 may promote tumor invasion and survival by impairing lysosomal integrity [90]. Elevated expression of SIRT1 has also been associated with higher rates of metastasis in TNBC but lower rates in all other types of breast cancer [91]. High protein expression of SIRT2 was correlated with poor prognosis in high-grade breast cancer, but the correlation was reversed in intermediate-grade breast cancer [92]. In basal-like breast cancer, SIRT2 can be overexpressed and stabilize EMT-TF Slug to promote tumor invasion and stem-like phenotypes [93]. However, SIRT2 expression was shown to sensitize breast cancer cells to oxidant stress-inducing agents by modulating peroxidase activity [94]. It also was demonstrated to inhibit tumor growth by deacetylating M2 isoform of pyruvate kinase, thus altering glucose metabolism [95]. SIRT3 was correlated with poor prognosis in breast cancer patients, but decreased mitochondrial expression of SIRT3 was associated with poor prognosis [96, 97]. Multiple studies have demonstrated the tumor-suppressing role of SIRT3 in reprogramming cancer cell metabolism in the mitochondria [98, 99]. Distinct from other SIRTs, multiple studies of SIRT4 agreed that it is tumor-suppressive. Decreased SIRT4 has been associated with poor prognosis and induced stemness in breast cancer [100, 101]. In addition, SIRT4 can inhibit IL-6/STAT3 signaling to improve the response of ER-positive breast cancer to endocrine therapy [102]. SIRT5 is another SIRT that plays an important role in cancer metabolism to promote breast cancer progression. SIRT5 can induce the expression of glutaminase and promote aerobic glycolysis in breast cancer [103, 104]. SIRT6 facilitates mammary tumorigenesis by increasing oxidative phosphorylation and has been associated with poor prognosis in HER2-positive breast cancer [105, 106]. SIRT7 can inhibit metastasis of breast cancer by inhibiting TGF-β signaling, and HDAC8 can suppress the expression of SIRT7 to promote cancer cell survival and migration [107, 108].

In summary, HDACs are much more extensively studied in breast cancer than HATs, but most HDACs were found to both inhibit and promote breast cancer depending on the cell context, and some studies reported seemingly contradictory results (Fig. 1). These findings indicate that the functions and targets of HDACs are not the same across all breast cancers and can be dependent on the subcellular location of the HDAC, breast cancer subtype, metastatic status, hormone receptor expression, and tumor grade. Therefore, all those factors will need to be considered when targeting HDACs in breast cancer.

Table 2 Family, class, localization, and functions of histone deacetylases (HDACs)

HDAC inhibitors in breast cancer

Although the roles of HDACs in breast cancer are complicated and heterogeneous, many HDAC inhibitors have exhibited antitumor effects in preclinical models of breast cancer [123]. However, to date, no HDAC inhibitor has been approved for the treatment of breast cancer. HDAC inhibitors currently in active clinical trials are vorinostat, belinostat, romidepsin, entinostat, and tucidinostat (Table 3). Vorinostat is a pan HDAC inhibitor and the first HDAC inhibitor approved by the Food and Drug Administration (FDA) [124]. In preclinical models of breast cancer, it was shown to induce apoptosis and autophagy while inhibiting proliferation, EMT, and migration [125]. In addition, vorinostat was found to induce ER degradation and improve the response of ER-positive breast cancer cells to endocrine therapy [126]. Breast cancer patients treated with vorinostat as a single agent failed to show an adequate response in the clinical trial [127]. The published clinical study of vorinostat in combination with endocrine therapy or chemotherapy showed encouraging results, but it never entered late-phase clinical trials for breast cancer (Table 3) [128,129,130]. Similar to vorinostat, belinostat is also an FDA-approved pan HDAC inhibitor still in early-phase clinical trials for breast cancer [131] (Table 3). In TNBC cells in vitro, belinostat induced cell apoptosis and showed possible synergy with chemotherapy [132]. Belinostat also exhibited synergistic effects with the PARP inhibitor olaparib in BRCA1-mutated TNBC cells and xenografts [133]. Currently, no clinical trial results have been published for belinostat.

Romidepsin and entinostat are Class I HDAC inhibitors, different from vorinostat and belinostat. Romidepsin inhibits HDAC1 and HDAC2 specifically and is FDA-approved [134]. In a preclinical model of inflammatory breast cancer, romidepsin treatment led to the destruction of tumor emboli and lymphatic vascular structure, inhibiting the growth of primary tumors and metastases in combination with paclitaxel [135]. In TNBC preclinical models, romidepsin in combination with gemcitabine and cisplatin inhibited tumor growth, EMT, invasion, and metastasis [136]. In contrast, entinostat inhibits HDAC1 and HDAC3 but not HDAC2. The effects of entinostat have been studied across all subtypes of breast cancer. Entinostat was shown to induce the expression of ER in ER-negative breast cancer and sensitize it to endocrine therapy in vitro and in vivo [137]. Entinostat also inhibited tumor-initiating cells in TNBC [138]. In preclinical models of HER2-positive breast cancer, entinostat exhibited combinational synergistic effects with the HER2/epidermal growth factor receptor (EGFR) dual tyrosine kinase inhibitor to inhibit tumor progression, sensitizing tumor cells to anti-HER2 treatments [139]. For ER-positive breast cancer, entinostat reversed endocrine therapy resistance in a xenograft model by reducing HER2 expression [140]. However, in the phase 3 clinical trial, entinostat did not improve the overall survival of ER-positive breast cancer patients resistant to endocrine therapy [141]. Moreover, entinostat in combination with azacitidine, a DNA methyltransferase inhibitor, showed limited benefits to breast cancer patients in a phase 2 clinical trial [142]. Recent early-phase clinical trials are investigating the effects of entinostat in combination with immune checkpoint blockade in advanced breast cancer [143] (Table 3).

Tucidinostat is distinct from other HDAC inhibitors because it inhibits HDAC1-3 (Class I) and HDAC10 (Class II). It is approved by the Chinese and Japanese FDAs but not the United States FDA and is currently in many more clinical trials than all other HDAC inhibitors (Table 3). Tucidinostat was shown to promote autophagy and apoptosis in breast cancer cells in vitro and improve the response to doxorubicin in vivo [144]. In addition, tucidinostat was demonstrated to improve the response of AR-positive TNBC to AR antagonists [145]. Extensive clinical trial results have demonstrated that tucidinostat in combination with endocrine therapy provided therapeutic benefits to patients with advanced ER-positive breast cancer, but adverse events from the treatment were a potential concern [146,147,148,149].

Besides those in active clinical trials, panobinostat is another pan HDAC inhibitor approved by the FDA and tested in breast cancer. Preclinical studies indicated that panobinostat induces autophagy in breast cancer cells and inhibits TNBC in vitro and in vivo [150, 151]. Panobinostat was also shown to reduce aromatase expression in ER-positive breast cancer and synergize with endocrine therapy [152]. In the published phase 1 clinical trial of panobinostat in combination with endocrine therapy, a partial response was observed with the highest dose [153]. Other clinical trials of panobinostat in breast cancer were terminated, withdrawn, or completed, but with no published results.

In addition to the ones mentioned, HDAC inhibitors such as mocetinostat and abexinostat have also been tested in the preclinical models of breast cancer. Mocetinostat, an inhibitor of HDAC1-3 and HDAC11, induced the expression of tumor suppressor Fyn-related kinase in basal-like breast cancer and showed antitumor effects in those overexpressing HDAC2 [154, 155]. Our previous study demonstrated that mocetinostat in combination with azacitidine reduced the growth of mesenchymal TNBC in vivo [156]. Abexinostat, a pan HDAC inhibitor, was shown to reduce cancer stem cells in breast cancer with low Xist expression [157]. Our previous findings demonstrated that mocetinostat and abexinostat can reverse EMT in in vitro models of mesenchymal breast cancer [156].

Compared to inhibitors of classic HDACs, SIRT inhibitors have not been as extensively studied in breast cancer. SIRT inhibitors MHY2256, Sirtinol, and Salermide were shown to inhibit the growth of breast cancer cells in vitro and in vivo by increasing p53 acetylation to induce cell death [158, 159]. TM, a SIRT2 inhibitor, induced the degradation of c-Myc and thus inhibited the growth of breast cancer cells and xenograft models [160]. Studies also suggested that sirtuin inhibitors might be able to overcome chemotherapy resistance in breast cancer, but those were not recent studies and were limited to in vitro treatments [91]. A more recent study showed that SIRT5 inhibitors have antitumor activity in breast cancer models [161]. However, to date, SIRT inhibitors have not entered any clinical studies.

In summary, despite the positive results seen in the preclinical setting, most HDAC inhibitors did not show impressive results in late-phase clinical studies for breast cancer. Moreover, a recent study suggested that HDAC inhibitors might promote breast cancer metastasis [162]. This again indicates that the roles of HDACs are complex. To improve their efficacy, especially in the clinic, biomarkers and more in-depth mechanistic studies will be needed to further elucidate the effects of HDAC inhibition. In addition, toxicity and selection of the combination therapy should also be considered and addressed in future studies of HDAC inhibitors.

Table 3 HDAC inhibitors in currently active clinical trials for breast cancer. All trial information was obtained from clinicaltrials.gov. PD1: programmed cell death protein 1; CTLA4: cytotoxic T-lymphocyte associated protein 4

Immunosuppressive myeloid cells in breast cancer

The tumor immune microenvironment (TIME) of breast cancer can elicit both antitumor and protumor effects [163]. Immunosuppressive cells in TIME can support tumor progression by promoting tumor growth, facilitating immune escape, contributing to metastasis, and affecting treatment response [164]. Myeloid cells are the most abundant infiltrated immune cells in many cancer types, including breast cancer [165, 166]. Immunosuppressive myeloid cells include polymorphonuclear myeloid-derived suppressor cells (PMN-MDSCs), monocytic MDSCs (mMDSCs), and immunosuppressive subsets of tumor-associated macrophages, monocytes, and dendritic cells (DCs) [167,168,169].

PMN-MDSC

PMN-MDSCs resemble many features of classical neutrophils, and most studies indicate that they originate from granulocytic lineage but are immature and pathologically activated [167, 169]. One study by Mastio et al. suggested that monocytic precursors can also differentiate into PMN-MDSCs [170]. The nomenclature of PMN-MDSCs has been controversial and evolved over time. In humans, PMN-MDSCs are defined as CD11b+CD33+HLADR−/lowCD14CD15+(or CD66b+), and in mice, they are defined as CD11b+Ly6G+Ly6Cmid/low [171, 172]. There were no markers to distinguish PMN-MDSCs and neutrophils in mice, and therefore PMN-MDSCs can only be defined based on functional studies that assess the immunosuppression activity [171]. Our previous study indicated that TANs of neutrophil-enriched breast cancer suppress T cells and should be considered PMN-MDSCs [173]. Recently, CD84 was identified as an emerging marker to identify MDSCs in breast cancer [174]. Multiple studies showed that tumor-secreted cytokines such as granulocyte and granulocyte-macrophage colony-stimulating factors (G-CSF and GM-CSF) skewed the differentiation of hematopoietic cells towards myelopoiesis in the bone marrow (BM) [175,176,177]. The overproduction of neutrophils in BM leads to neutrophil accumulation in the blood and spleen [173, 174]. The recent study by Alshetaiwi et al. demonstrated that neutrophils become PMN-MDSCs through an abnormal maturation trajectory in the spleen [174]. In contrast, our previous study showed that BM neutrophils in mammary tumor-bearing mice are immunosuppressive [173]. The study by Patel et al. suggested that BM neutrophils become immunosuppressive only in mice bearing late-stage tumors, providing a possible explanation for different findings in the two studies [178]. How BM neutrophils acquire immunosuppressive activity still needs to be further elucidated. PMN-MDSCs are recruited to the mammary tumor by chemokines such as C-X-C motif chemokine ligand 2 (CXCL2) [179, 180]. In tumors, PMN-MDSCs inhibit antitumor immune cells such as cytotoxic T lymphocytes (CTL) by producing reactive oxygen species and arginase 1 (Arg1) [172]. They also promote the activation and expansion of immunosuppressive regulatory T cells (Tregs) [181]. Breast cancer highly infiltrated with PMN-MDSCs is resistant to immune checkpoint blockade (ICB) [173]. Besides affecting immune cells, PMN-MDSCs also promote breast cancer initiation and support metastatic outgrowth by reverting the EMT phenotype [180, 182,183,184].

Monocytes and mMDSCs

Monocytes can give rise to macrophages and DCs, but some tumor-associated monocytes can be immunosuppressive without differentiation [169]. Monocytes can be categorized into classical and non-classical monocytes. Classical monocytes are defined as CD14highCD16 (human) or CD11b+Ly6GLy6C+ (mouse), whereas non-classical are defined as CD14lowCD16+ (human) or CD11b+Ly6GLy6Clow (mouse) [171, 185]. Classical monocytes exhibiting an inflammatory phenotype were shown to suppress CTLs and facilitate tumor metastasis in breast cancer models [186,187,188]. Those monocytes are recruited to the tumor by C-C motif ligand (CCL) 2 - C-C motif chemokine receptor 2 (CCR2) signaling [187]. In contrast, non-classical monocytes were demonstrated to inhibit breast cancer metastasis [189].

Tumor-associated mMDSCs are very similar to classical monocytes in marker expression. Human mMDSCs are defined as CD11b+CD33+HLA-DR−/low CD14+CD15 [172]. Mouse mMDSCs were defined as CD11b+Ly6GLy6C+, and those classic markers cannot distinguish mMDSCs from monocytes as both originate from monocytic precursors [172]. Similar to PMN-MDSCs, mMDSCs are immature myeloid cells and are the result of tumor-dependent abnormal cell activation. The study by Alshetaiwi et al. indicated that CD84 can also be used to identify mMDSCs from monocytes in breast cancer, in addition to distinguishing PMN-MDSCs from neutrophils [174]. Similar to that of PMN-MDSCs, the first step of mMDSC production is the abnormal expansion of BM myeloid cells driven by tumor-derived cytokines such as G-CSF, TGF-β, and IL-34 [190, 191]. Those cytokines also promote the immunosuppressive activity of mMDSCs. The release of mMDSCs from BM was shown to be regulated by tumor-derived factor PTH1R in a breast cancer model [192]. However, studies demonstrated BM and spleen mMDSCs of mammary tumor-bearing mice are not immunosuppressive, suggesting that they gain immunosuppressive activity when they reach the tumor [192, 193]. In addition, the study by Calvert et al. indicated that tumor mMDSCs have a limited ability to differentiate, while the study by Biswas et al. suggested that exosomes secreted by mesenchymal stem cells can promote differentiation of mMDSCs to protumor TAMs in breast cancer [193, 194]. The recruitment of mMDSCs and monocytes is facilitated by CCL2 and CCL5 in breast cancer [195]. T-cell suppression by mMDSCs is driven by the production of nitric oxide and Arg1 [195]. Moreover, recently, a study by Sarkar et al. showed that mMDSCs suppress CTLs by releasing adenosine in mouse models of multiple cancer types including breast cancer [196]. This study also showed that increased adenosine levels were a result of CD73 expression, which was induced by tumor-derived prostaglandin E2. Furthermore, mMDSCs also induce EMT in tumor cells to support the dissemination and accordingly metastasis in mouse models of breast cancer [184]. Elevated levels of mMDSCs have been correlated with poor clinical outcomes in patients with metastatic breast cancer [197].

Tumor-associated macrophages

Tumor-associated macrophages (TAMs) are defined as CD11b+Gr-1F4/80+ in mice and CD14+CD68+ in humans. Many early studies categorized TAMs into M1-like (antitumor) and M2-like (protumor) TAMs, but the field has realized that this binary system is oversimplified [198, 199]. Recently, the development of single-cell omics has further revealed the heterogeneity in TAM phenotypes and complexity in TAM biology [200]. In breast cancer, TAMs can arise from both tissue-resident macrophages and monocytes recruited to the tumor by tumor-derived cytokines [201, 202]. High TAM infiltration is associated with the more aggressive Claudin-low subtype of breast cancer, an EMT signature expression, and worse prognosis [173, 185]. The immunosuppressive activity of TAMs in breast cancer was reported in many early studies dating from more than 15 years ago. TAMs inhibit T cell response in breast cancer TIME by downregulating nitric oxide synthase gene expression and upregulating the production of Arg1 and hypoxia-inducible factor (HIF)-1α [203,204,205,206]. Recent studies, however, have mostly focused on other tumor-promoting roles of TAMs. A subset of breast cancer TAMs has been found to accumulate in hypoxic regions of mammary tumors and display a proangiogenic phenotype by activating the HIF-2α pathway and VEGF expression [207,208,209,210]. Many studies have demonstrated that TAMs induce breast cancer metastasis by promoting cancer cell migration, intravasation, and seeding at the metastasis as reviewed by Williams et al. [201]. In addition, TAMs can induce stem cell-like phenotypes in breast cancer cells through both paracrine and juxtracrine signaling [211, 212]. Because of their roles in tumor progression, inhibitors that deplete macrophages such as colony stimulating factor 1 receptor (CSF1R) antibodies are currently being tested in clinics for cancer treatment [201, 213]. However, besides promoting tumor progression, TAMs have the potential to exhibit tumor-inhibitory phenotypes [200, 213]. Therefore, as reviewed by Rannikko and Hollmen, therapeutics have been developed to reprogram TAMs by targeting various regulatory receptors and metabolic enzymes [214]. HDACs were mentioned as a potential target for TAM reprogramming.

Dendritic cells

DCs consist of three different subtypes, plasmacytoid DC (pDCs), conventional DC (cDCs), and monocytic DC (moDCs) [215]. While most DC arise from myeloid progenitors in BM, some pDC can differentiate from lymphoid progenitors [215, 216]. Although pCs can produce interferon and are involved in anti-viral immunity, they have been shown to facilitate immune tolerance in cancer settings [216, 217]. In breast cancer, tumor-derived factors such as tumor necrosis factor-α (TNF-α) reprogram pDCs leading to impaired interferon (IFN)-α production [218, 219]. The reprogrammed pDCs then promote Treg expansion by expressing forkhead box O3 (FOXO3) and inducible costimulatory molecule ligands, and therefore pDCs infiltration is correlated with poor prognosis in breast cancer patients [220,221,222,223]. In contrast, cDCs arise from DC progenitors in BM and can be divided into two subtypes cDC1 and cDC2 [215]. Because of their antigen presentation and T cell priming abilities, the infiltration of cDCs, especially cDC1, has been associated with better prognosis in breast cancer patients [224]. However, the normal functions of cDCs can also be impaired by breast cancer and reprogrammed to promote immunosuppression [217]. In the PyMT breast cancer model, cDC1 was shown to highly express the immune inhibitory receptor TIM-3, inhibiting T cell recruitment [225]. Distinct from other DCs, moDCs differentiate from monocytes during inflammation and cancer [215, 217]. In breast cancer, the functions of moDCs have not been widely studied, but one study demonstrated that moDCs from breast cancer patients induced the proliferation of Tregs but not immunostimulatory T cells [226]. Although some DCs can gain immunosuppressive activity, DCs are important in stimulating antitumor immune response. Therefore, various therapies including those targeting epigenetic modulators have been developed and tested to activate DC-dependent immune response or promote DC infiltration [217].

HATs and HDACs in immunosuppressive myeloid cells

Histone acetylation can regulate the accumulation and phenotypes of myeloid cells in TIME. The study by Sasidharan Nair et al. indicated that the expression of HAT-associated genes increased in PMN-MDSCs while that of HDAC-associated genes decreased [227]. In addition, HDAC2 was shown to facilitate the conversion of monocytes to PMN-MDSCs by reducing the transcription of the retinoblastoma gene [228]. Several studies investigated the effects of HAT inhibitors in MDSCs. The CBP/EP300 BRD inhibitor GNE-781 reprogrammed both mMDSCs and PMN-MDSCs from an immunosuppressive phenotype to a more inflammatory phenotype by inhibiting the expression of STAT-related genes, Arg1, and inducible nitric oxide synthase [229]. Our recent study also showed that CBP/EP300 BRD inhibition can reduce PMN-MDSCs and inhibit abnormal production of granulocytic progenitors in BM [54]. Moreover, the KAT6A (MYST family HAT) was found to acetylate SMAD3 and H3K23 to induce SMAD3 activation resulting in MDSC (Gr-1+) recruitment in TNBC [230]. This study also showed that KAT6A inhibitor WM-1119 decreased MDSC recruitment and activated T-cell response when combined with ICB treatment. Many HDAC inhibitors have been demonstrated to reduce MDSC accumulation and/or functions in various cancer types, as reviewed by Adeshakin et al. [231]. For example, the low-dose HDAC inhibitor entinostat in combination with azacytidine inhibited the migration of mMDSCs and reprogrammed the mMDSCs to be more macrophage-like [232]. The HDAC inhibitor vorinostat reduced MDSC infiltration and activated T-cell response in 4T1 tumors by inducing MDSC apoptosis [233]. This study also indicated that the epigenetic therapy combination inhibited the formation of lung metastases by disrupting the premetastatic niches supported by mMDSCs. Another study by Kim et al. found that entinostat in combination with ICB reduced PMN-MDSCs in the breast cancer model 4T1 and suppressed their function [234]. These findings suggest that HDAC or HAT inhibitors can reduce immunosuppression by MDSCs and be combined with ICB to improve T cell activation.

As mentioned previously, targeting the epigenetic modulators is a potential strategy for TAM reprogramming. The class IIA HDAC inhibitor TMP195 was shown to induce phagocytic and immunostimulatory activities of TAMs in a breast cancer mouse model [235]. The low-dose HDAC inhibitor trichostatin-A promoted antitumor phenotypes in TAMs and showed synergistic effects with ICB [236]. HDACs were found to mediate the downregulation of major histocompatibility complex II (MHCII) expression in TAMs, and HDAC inhibitor treatment restored the expression [237]. High expression of HDAC6 was found to promote protumor phenotypes in TAMs, and HDAC6 inhibition improved the response of breast cancer to ICB in part by stimulating an antitumor immune response [238, 239]. Compared to those of HDACs, very few studies investigated HATs in TAMs. One study from Wang et al. showed that EP300 can facilitate the expression of IL-6, a metastasis-promoting cytokine, in TAMs by increasing the acetylation of histone H3 [240].

Compared to TAMs and MDSCs, how HATs or HDACs affect the DC phenotype has not been extensively studied, especially in the cancer setting. HDAC1 was found to be critical for the development of pDCs and cDC2, and HDAC inhibition led to altered differentiation in bone marrow resulting in no pDC production [241, 242]. In tumor-bearing mice, HDAC1 deletion promoted the activation of cDC1 and CTL in TIME [242]. In contrast, HDAC9 deficiency leads to reduced CD8 + DC infiltration and impaired antigen presentation [243]. In addition, inhibition of HDAC6 was shown to inhibit the production of immunosuppressive cytokine IL-10 in both TAMs and DCs [244].

Overall, these studies demonstrated that HDAC and HAT inhibition can reprogram the phenotypes of tumor-infiltrated myeloid cells. However, the effects of HATs have not been as extensively studied compared to those of HDACs. These findings again emphasize the importance of having more specific inhibitors and highlight the importance of examining the effects on TIME while testing those inhibitors for potential cancer treatment. Tumor-associated immune cells such as immunosuppressive myeloid cells might contribute to whether tumors respond to HAT and HDAC inhibitors. Furthermore, HAT or HDAC inhibitors may facilitate T cell activation, and therefore combination with ICB should be considered in the future studies of those inhibitors.

Conclusion

In summary, although HATs and HDACs have opposite functions in modulating histone acetylation, inhibitors of both have been investigated as potential treatments for breast cancer. The functions of HATs and their inhibitors were mostly studied in hormone receptor-positive breast cancer. Currently, no HAT inhibitors are being tested in clinics for breast cancer specifically, but novel inhibitors such as those targeting CBP/EP300 BRD recently entered early clinical trials for solid tumor treatment. The novel inhibitors have been reported to be effective in inhibiting breast cancer and immunosuppression, but their effects will need to be further elucidated. Compared to HATs, HDACs have been more extensively investigated across different breast cancer subtypes and in tumor-infiltrated myeloid cells. The functions and targets of different HDACs were demonstrated to be complex and context dependent. Many HDAC inhibitors have been developed but have not succeeded in the clinic, especially as single agents. Current clinical trials mostly focus on testing HDAC inhibitors in combination with standard-of-care therapies. The effects of HAT and HDAC inhibitors on breast cancer alone and in combination with standard-of-care therapies should be more carefully investigated in future studies. Biomarkers may be needed to better identify breast cancer patients that might benefit from those inhibitors. The frequency of tumor-associated myeloid cells may potentially serve as biomarkers.

Data availability

No datasets were generated or analysed during the current study.

Abbreviations

ER:

Estrogen receptor

PR:

Progesterone receptor

HER2:

Human epidermal growth factor receptor 2

TNBC:

Triple-negative breast cancer

K:

lysine

HAT:

Histone acetyltransferase

GNAT:

Gcn5-related N-acetyltransferase

CBP:

CREB-binding protein

EP300:

E1A-associated protein p300

SRC:

Steroid receptor coactivator

HDAC:

Histone deacetylase

BRD:

BromodomainPARP: poly (ADP-ribose) polymerase

AR:

Androgen receptor

EMT:

Epithelial-mesenchymal transition

TGF-β:

transforming growth factorβ

EMT-TF:

EMT-inducing transcription factor

VEGF:

Vascular endothelial growth factor

IL:

Interleukin

CDK:

Cyclin-dependent kinase

SIRT:

Sirtuin

EGFR:

Epidermal growth factor receptor

PD1:

Programmed cell death protein 1

CTLA4:

Cytotoxic T-lymphocyte associated protein 4

TIME:

Tumor immune microenvironment

PMN:

Polymorphonuclear

MDSCs:

Myeloid-derived suppressor cells

mMDSCs:

monocytic MDSCs

DCs:

Dendritic cells

G-CSF:

Granulocyte colony-stimulating factor

GM-CSF:

Granulocyte-macrophage colony-stimulating factor

BM:

Bone marrow

CXCL2:

C-X-C motif chemokine ligand 2

CTL:

Cytotoxic T lymphocytes

Arg1:

Arginase 1

Tregs:

regulatory T cells

ICB:

Immune checkpoint blockade

CCL:

C-C motif ligand

CCR:

C-C motif chemokine receptor

TAMs:

Tumor-associated macrophages

HIF:

Hypoxia-inducible factor

CSF1R:

Colony stimulating factor 1 receptor

pDCs:

plasmacytoid DCs

cDCs:

conventional DCs

moDCs:

monocytic DCs

TNF-α:

Tumor necrosis factor-α

IFN:

Interferon

FOXO3:

Forehead box O3

MHCII:

Major histocompatibility complex II

References

  1. Arnold M, Morgan E, Rumgay H, Mafra A, Singh D, Laversanne M, et al. Current and future burden of breast cancer: global statistics for 2020 and 2040. Breast. 2022;66:15–23.

    Article  PubMed  PubMed Central  Google Scholar 

  2. Lukasiewicz S, Czeczelewski M, Forma A, Baj J, Sitarz R, Stanislawek A. Breast Cancer-Epidemiology, risk factors, classification, prognostic markers, and current treatment Strategies-An updated review. Cancers (Basel). 2021;13(17).

  3. Bianchini G, Balko JM, Mayer IA, Sanders ME, Gianni L. Triple-negative breast cancer: challenges and opportunities of a heterogeneous disease. Nat Rev Clin Oncol. 2016;13(11):674–90.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  4. Dent R, Trudeau M, Pritchard KI, Hanna WM, Kahn HK, Sawka CA, et al. Triple-negative breast cancer: clinical features and patterns of recurrence. Clin Cancer Res. 2007;13(15 Pt 1):4429–34.

    Article  PubMed  Google Scholar 

  5. Thakur C, Qiu Y, Fu Y, Bi Z, Zhang W, Ji H, et al. Epigenetics and environment in breast cancer: new paradigms for anti-cancer therapies. Front Oncol. 2022;12:971288.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  6. Yang J, Xu J, Wang W, Zhang B, Yu X, Shi S. Epigenetic regulation in the tumor microenvironment: molecular mechanisms and therapeutic targets. Signal Transduct Target Ther. 2023;8(1):210.

    Article  PubMed  PubMed Central  Google Scholar 

  7. Li W, Wu H, Sui S, Wang Q, Xu S, Pang D. Targeting histone modifications in breast cancer: A precise weapon on the way. Front Cell Dev Biol. 2021;9:736935.

    Article  PubMed  PubMed Central  Google Scholar 

  8. Grunstein M. Histone acetylation in chromatin structure and transcription. Nature. 1997;389(6649):349–52.

    Article  CAS  PubMed  Google Scholar 

  9. Marmorstein R, Zhou MM. Writers and readers of histone acetylation: structure, mechanism, and Inhibition. Cold Spring Harb Perspect Biol. 2014;6(7):a018762.

    Article  PubMed  PubMed Central  Google Scholar 

  10. Lee KK, Workman JL. Histone acetyltransferase complexes: one size doesn’t fit all. Nat Rev Mol Cell Biol. 2007;8(4):284–95.

    Article  CAS  PubMed  Google Scholar 

  11. Park SY, Kim JS. A short guide to histone deacetylases including recent progress on class II enzymes. Exp Mol Med. 2020;52(2):204–12.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  12. Fujisawa T, Filippakopoulos P. Functions of bromodomain-containing proteins and their roles in homeostasis and cancer. Nat Rev Mol Cell Biol. 2017;18(4):246–62.

    Article  CAS  PubMed  Google Scholar 

  13. Wu D, Qiu Y, Jiao Y, Qiu Z, Liu D. Small molecules targeting hats, HDACs, and BRDs in cancer therapy. Front Oncol. 2020;10:560487.

    Article  PubMed  PubMed Central  Google Scholar 

  14. Koutelou E, Farria AT, Dent SYR. Complex functions of Gcn5 and Pcaf in development and disease. Biochim Biophys Acta Gene Regul Mech. 2021;1864(2):194609.

    Article  CAS  PubMed  Google Scholar 

  15. Iyer NG, Ozdag H, Caldas C. p300/CBP and cancer. Oncogene. 2004;23(24):4225–31.

    Article  CAS  PubMed  Google Scholar 

  16. Kim JJ, Lee SY, Choi JH, Woo HG, Xhemalce B, Miller KM. PCAF-Mediated histone acetylation promotes replication fork degradation by MRE11 and EXO1 in BRCA-Deficient cells. Mol Cell. 2020;80(2):327–44. e8.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  17. Watts GS, Oshiro MM, Junk DJ, Wozniak RJ, Watterson S, Domann FE, et al. The acetyltransferase p300/CBP-associated factor is a p53 target gene in breast tumor cells. Neoplasia. 2004;6(3):187–94.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  18. Gorrini C, Squatrito M, Luise C, Syed N, Perna D, Wark L, et al. Tip60 is a haplo-insufficient tumour suppressor required for an oncogene-induced DNA damage response. Nature. 2007;448(7157):1063–7.

    Article  CAS  PubMed  Google Scholar 

  19. McGuire A, Casey MC, Shalaby A, Kalinina O, Curran C, Webber M, et al. Quantifying Tip60 (Kat5) stratifies breast cancer. Sci Rep. 2019;9(1):3819.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  20. Judes G, Dubois L, Rifai K, Idrissou M, Mishellany F, Pajon A, et al. TIP60: an actor in acetylation of H3K4 and tumor development in breast cancer. Epigenomics. 2018;10(11):1415–30.

    Article  CAS  PubMed  Google Scholar 

  21. Ma Y, Chen X, Ding T, Zhang H, Zhang Q, Dai H, et al. KAT7 promotes radioresistance through upregulating PI3K/AKT signaling in breast cancer. J Radiat Res. 2023;64(2):448–56.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  22. Germaniuk-Kurowska A, Nag A, Zhao X, Dimri M, Band H, Band V. Ada3 requirement for HAT recruitment to Estrogen receptors and Estrogen-dependent breast cancer cell proliferation. Cancer Res. 2007;67(24):11789–97.

    Article  CAS  PubMed  Google Scholar 

  23. Benecke A, Gaudon C, Garnier JM, vom Baur E, Chambon P, Losson R. ADA3-containing complexes associate with Estrogen receptor alpha. Nucleic Acids Res. 2002;30(11):2508–14.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  24. Yu L, Liang Y, Cao X, Wang X, Gao H, Lin SY, et al. Identification of MYST3 as a novel epigenetic activator of ERalpha frequently amplified in breast cancer. Oncogene. 2017;36(20):2910–8.

    Article  CAS  PubMed  Google Scholar 

  25. Turner-Ivey B, Guest ST, Irish JC, Kappler CS, Garrett-Mayer E, Wilson RC, et al. KAT6A, a chromatin modifier from the 8p11-p12 amplicon is a candidate oncogene in luminal breast cancer. Neoplasia. 2014;16(8):644–55.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  26. Giannoudis A, Malki MI, Rudraraju B, Mohhamed H, Menon S, Liloglou T, et al. Activating transcription factor-2 (ATF2) is a key determinant of resistance to endocrine treatment in an in vitro model of breast cancer. Breast Cancer Res. 2020;22(1):126.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  27. Kiliti AJ, Sharif GM, Martin MB, Wellstein A, Riegel AT. AIB1/SRC-3/NCOA3 function in Estrogen receptor alpha positive breast cancer. Front Endocrinol (Lausanne). 2023;14:1250218.

    Article  PubMed  Google Scholar 

  28. Liu Z, Wong J, Tsai SY, Tsai MJ, O’Malley BW. Steroid receptor coactivator-1 (SRC-1) enhances ligand-dependent and receptor-dependent cell-free transcription of chromatin. Proc Natl Acad Sci U S A. 1999;96(17):9485–90.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  29. Spencer TE, Jenster G, Burcin MM, Allis CD, Zhou J, Mizzen CA, et al. Steroid receptor coactivator-1 is a histone acetyltransferase. Nature. 1997;389(6647):194–8.

    Article  CAS  PubMed  Google Scholar 

  30. Gao X, Loggie BW, Nawaz Z. The roles of sex steroid receptor coregulators in cancer. Mol Cancer. 2002;1:7.

    Article  PubMed  PubMed Central  Google Scholar 

  31. Browne AL, Charmsaz S, Vareslija D, Fagan A, Cosgrove N, Cocchiglia S, et al. Network analysis of SRC-1 reveals a novel transcription factor hub which regulates endocrine resistant breast cancer. Oncogene. 2018;37(15):2008–21.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  32. Caligiuri M, Williams GL, Castro J, Battalagine L, Wilker E, Yao L, et al. FT-6876, a potent and selective inhibitor of CBP/p300, is active in preclinical models of androgen Receptor-Positive breast cancer. Target Oncol. 2023;18(2):269–85.

    Article  PubMed  PubMed Central  Google Scholar 

  33. Dongre A, Weinberg RA. New insights into the mechanisms of epithelial-mesenchymal transition and implications for cancer. Nat Rev Mol Cell Biol. 2019;20(2):69–84.

    Article  CAS  PubMed  Google Scholar 

  34. Felipe Lima J, Nofech-Mozes S, Bayani J, Bartlett JM. EMT in breast Carcinoma-A review. J Clin Med. 2016;5(7).

  35. Creighton CJ, Li X, Landis M, Dixon JM, Neumeister VM, Sjolund A, et al. Residual breast cancers after conventional therapy display mesenchymal as well as tumor-initiating features. Proc Natl Acad Sci U S A. 2009;106(33):13820–5.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  36. Zhao L, Pang A, Li Y. Function of GCN5 in the TGF-beta1-induced epithelial-to-mesenchymal transition in breast cancer. Oncol Lett. 2018;16(3):3955–63.

    PubMed  PubMed Central  Google Scholar 

  37. Shi J, Wang Y, Zeng L, Wu Y, Deng J, Zhang Q, et al. Disrupting the interaction of BRD4 with diacetylated twist suppresses tumorigenesis in basal-like breast cancer. Cancer Cell. 2014;25(2):210–25.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  38. Cho MH, Park JH, Choi HJ, Park MK, Won HY, Park YJ, et al. DOT1L cooperates with the c-Myc-p300 complex to epigenetically derepress CDH1 transcription factors in breast cancer progression. Nat Commun. 2015;6:7821.

    Article  CAS  PubMed  Google Scholar 

  39. Fang Z, Wang X, Sun X, Hu W, Miao QR. The role of histone protein acetylation in regulating endothelial function. Front Cell Dev Biol. 2021;9:672447.

    Article  PubMed  PubMed Central  Google Scholar 

  40. Poziello A, Nebbioso A, Stunnenberg HG, Martens JHA, Carafa V, Altucci L. Recent insights into histone Acetyltransferase-1: biological function and involvement in pathogenesis. Epigenetics. 2021;16(8):838–50.

    Article  PubMed  Google Scholar 

  41. Glozak MA, Sengupta N, Zhang X, Seto E. Acetylation and deacetylation of non-histone proteins. Gene. 2005;363:15–23.

    Article  CAS  PubMed  Google Scholar 

  42. Yuan ZL, Guan YJ, Chatterjee D, Chin YE. Stat3 dimerization regulated by reversible acetylation of a single lysine residue. Science. 2005;307(5707):269–73.

    Article  CAS  PubMed  Google Scholar 

  43. Fu M, Wang C, Reutens AT, Wang J, Angeletti RH, Siconolfi-Baez L, et al. p300 and p300/cAMP-response element-binding protein-associated factor acetylate the androgen receptor at sites governing hormone-dependent transactivation. J Biol Chem. 2000;275(27):20853–60.

    Article  CAS  PubMed  Google Scholar 

  44. Gaughan L, Logan IR, Cook S, Neal DE, Robson CN. Tip60 and histone deacetylase 1 regulate androgen receptor activity through changes to the acetylation status of the receptor. J Biol Chem. 2002;277(29):25904–13.

    Article  CAS  PubMed  Google Scholar 

  45. Wang C, Fu M, Angeletti RH, Siconolfi-Baez L, Reutens AT, Albanese C, et al. Direct acetylation of the Estrogen receptor alpha hinge region by p300 regulates transactivation and hormone sensitivity. J Biol Chem. 2001;276(21):18375–83.

    Article  CAS  PubMed  Google Scholar 

  46. Sun Y, Du F. Functional characterization of ATM kinase using Acetylation-Specific antibodies. Methods Mol Biol. 2017;1599:157–62.

    Article  CAS  PubMed  Google Scholar 

  47. Cohen HY, Lavu S, Bitterman KJ, Hekking B, Imahiyerobo TA, Miller C, et al. Acetylation of the C terminus of Ku70 by CBP and PCAF controls Bax-mediated apoptosis. Mol Cell. 2004;13(5):627–38.

    Article  CAS  PubMed  Google Scholar 

  48. White J, Derheimer FA, Jensen-Pergakes K, O’Connell S, Sharma S, Spiegel N, et al. Histone lysine acetyltransferase inhibitors: an emerging class of drugs for cancer therapy. Trends Pharmacol Sci. 2024;45(3):243–54.

    Article  CAS  PubMed  Google Scholar 

  49. Bommi-Reddy A, Park-Chouinard S, Mayhew DN, Terzo E, Hingway A, Steinbaugh MJ, et al. CREBBP/EP300 acetyltransferase Inhibition disrupts FOXA1-bound enhancers to inhibit the proliferation of ER + breast cancer cells. PLoS ONE. 2022;17(3):e0262378.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  50. Waddell A, Mahmud I, Ding H, Huo Z, Liao D. Pharmacological Inhibition of CBP/p300 blocks Estrogen receptor alpha (ERalpha) function through suppressing enhancer H3K27 acetylation in luminal breast cancer. Cancers (Basel). 2021;13(11).

  51. Breen ME, Mapp AK. Modulating the masters: chemical tools to dissect CBP and p300 function. Curr Opin Chem Biol. 2018;45:195–203.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  52. Weinert BT, Narita T, Satpathy S, Srinivasan B, Hansen BK, Scholz C, et al. Time-Resolved analysis reveals rapid dynamics and broad scope of the CBP/p300 acetylome. Cell. 2018;174(1):231–44. e12.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  53. Strachowska M, Gronkowska K, Michlewska S, Robaszkiewicz A. CBP/p300 bromodomain Inhibitor-I-CBP112 declines transcription of the key ABC transporters and sensitizes cancer cells to chemotherapy drugs. Cancers (Basel). 2021;13:18.

    Article  Google Scholar 

  54. Yuan X, Hao X, Chan HL, Zhao N, Pedroza DA, Liu F et al. CREB-binding protein/P300 bromodomain Inhibition reduces neutrophil accumulation and activates antitumor immunity in triple-negative breast cancer. JCI Insight. 2024;9(20).

  55. Welti J, Sharp A, Brooks N, Yuan W, McNair C, Chand SN, et al. Targeting the p300/CBP axis in lethal prostate cancer. Cancer Discov. 2021;11(5):1118–37.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  56. Nicosia L, Spencer GJ, Brooks N, Amaral FMR, Basma NJ, Chadwick JA, et al. Therapeutic targeting of EP300/CBP by bromodomain Inhibition in hematologic malignancies. Cancer Cell. 2023;41(12):2136–53. e13.

    Article  CAS  PubMed  Google Scholar 

  57. Sharma S, Chung CY, Uryu S, Petrovic J, Cao J, Rickard A, et al. Discovery of a highly potent, selective, orally bioavailable inhibitor of KAT6A/B histone acetyltransferases with efficacy against KAT6A-high ER + breast cancer. Cell Chem Biol. 2023;30(10):1191–210. e20.

    Article  CAS  PubMed  Google Scholar 

  58. Mukohara T, Park YH, Sommerhalder D, Yonemori K, Hamilton E, Kim SB et al. Inhibition of lysine acetyltransferase KAT6 in ER(+)HER2(-) metastatic breast cancer: a phase 1 trial. Nat Med. 2024.

  59. Mehmood SA, Sahu KK, Sengupta S, Partap S, Karpoormath R, Kumar B, et al. Recent advancement of HDAC inhibitors against breast cancer. Med Oncol. 2023;40(7):201.

    Article  CAS  PubMed  Google Scholar 

  60. Li Y, Seto E. HDACs and HDAC inhibitors in cancer development and therapy. Cold Spring Harb Perspect Med. 2016;6(10).

  61. Li G, Tian Y, Zhu WG. The roles of histone deacetylases and their inhibitors in cancer therapy. Front Cell Dev Biol. 2020;8:576946.

    Article  PubMed  PubMed Central  Google Scholar 

  62. Yam C, Abuhadra N, Sun R, Adrada BE, Ding QQ, White JB, et al. Molecular characterization and prospective evaluation of pathologic response and outcomes with neoadjuvant therapy in metaplastic Triple-Negative breast cancer. Clin Cancer Res. 2022;28(13):2878–89.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  63. Krusche CA, Wulfing P, Kersting C, Vloet A, Bocker W, Kiesel L, et al. Histone deacetylase-1 and– 3 protein expression in human breast cancer: a tissue microarray analysis. Breast Cancer Res Treat. 2005;90(1):15–23.

    Article  CAS  PubMed  Google Scholar 

  64. Muller BM, Jana L, Kasajima A, Lehmann A, Prinzler J, Budczies J, et al. Differential expression of histone deacetylases HDAC1, 2 and 3 in human breast cancer–overexpression of HDAC2 and HDAC3 is associated with clinicopathological indicators of disease progression. BMC Cancer. 2013;13:215.

    Article  PubMed  PubMed Central  Google Scholar 

  65. Zhang Z, Yamashita H, Toyama T, Sugiura H, Ando Y, Mita K, et al. Quantitation of HDAC1 mRNA expression in invasive carcinoma of the breast*. Breast Cancer Res Treat. 2005;94(1):11–6.

    Article  CAS  PubMed  Google Scholar 

  66. Weichert W. HDAC expression and clinical prognosis in human malignancies. Cancer Lett. 2009;280(2):168–76.

    Article  CAS  PubMed  Google Scholar 

  67. Kawai H, Li H, Avraham S, Jiang S, Avraham HK. Overexpression of histone deacetylase HDAC1 modulates breast cancer progression by negative regulation of Estrogen receptor alpha. Int J Cancer. 2003;107(3):353–8.

    Article  CAS  PubMed  Google Scholar 

  68. Citro S, Miccolo C, Meloni L, Chiocca S. PI3K/mTOR mediate mitogen-dependent HDAC1 phosphorylation in breast cancer: a novel regulation of Estrogen receptor expression. J Mol Cell Biol. 2015;7(2):132–42.

    Article  CAS  PubMed  Google Scholar 

  69. Tang Z, Ding S, Huang H, Luo P, Qing B, Zhang S, et al. HDAC1 triggers the proliferation and migration of breast cancer cells via upregulation of interleukin-8. Biol Chem. 2017;398(12):1347–56.

    Article  CAS  PubMed  Google Scholar 

  70. Ma L, Qi L, Li S, Yin Q, Liu J, Wang J, et al. Aberrant HDAC3 expression correlates with brain metastasis in breast cancer patients. Thorac Cancer. 2020;11(9):2493–505.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  71. Huang W, Chen J, Liu X, Liu X, Duan S, Chen L, et al. MIER3 induces epithelial-mesenchymal transition and promotes breast cancer cell aggressiveness via forming a co-repressor complex with HDAC1/HDAC2/Snail. Exp Cell Res. 2021;406(1):112722.

    Article  CAS  PubMed  Google Scholar 

  72. Pantelaiou-Prokaki G, Mieczkowska I, Schmidt GE, Fritzsche S, Prokakis E, Gallwas J, et al. HDAC8 suppresses the epithelial phenotype and promotes EMT in chemotherapy-treated basal-like breast cancer. Clin Epigenetics. 2022;14(1):7.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  73. An P, Chen F, Li Z, Ling Y, Peng Y, Zhang H, et al. HDAC8 promotes the dissemination of breast cancer cells via AKT/GSK-3beta/Snail signals. Oncogene. 2020;39(26):4956–69.

    Article  CAS  PubMed  Google Scholar 

  74. Gong C, Qu S, Lv XB, Liu B, Tan W, Nie Y, et al. BRMS1L suppresses breast cancer metastasis by inducing epigenetic silence of FZD10. Nat Commun. 2014;5:5406.

    Article  PubMed  Google Scholar 

  75. Ray A, Alalem M, Ray BK. Loss of epigenetic Kruppel-like factor 4 histone deacetylase (KLF-4-HDAC)-mediated transcriptional suppression is crucial in increasing vascular endothelial growth factor (VEGF) expression in breast cancer. J Biol Chem. 2013;288(38):27232–42.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  76. Zhang Y, Nalawansha DA, Herath KE, Andrade R, Pflum MKH. Differential profiles of HDAC1 substrates and associated proteins in breast cancer cells revealed by trapping. Mol Omics. 2021;17(4):544–53.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  77. Zhou Y, Jin X, Ma J, Ding D, Huang Z, Sheng H, et al. HDAC5 loss impairs RB repression of Pro-Oncogenic genes and confers CDK4/6 inhibitor resistance in cancer. Cancer Res. 2021;81(6):1486–99.

    Article  CAS  PubMed  Google Scholar 

  78. Xue Y, Lian W, Zhi J, Yang W, Li Q, Guo X, et al. HDAC5-mediated deacetylation and nuclear localisation of SOX9 is critical for Tamoxifen resistance in breast cancer. Br J Cancer. 2019;121(12):1039–49.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  79. Saji S, Kawakami M, Hayashi S, Yoshida N, Hirose M, Horiguchi S, et al. Significance of HDAC6 regulation via Estrogen signaling for cell motility and prognosis in Estrogen receptor-positive breast cancer. Oncogene. 2005;24(28):4531–9.

    Article  CAS  PubMed  Google Scholar 

  80. Zhang Z, Yamashita H, Toyama T, Sugiura H, Omoto Y, Ando Y, et al. HDAC6 expression is correlated with better survival in breast cancer. Clin Cancer Res. 2004;10(20):6962–8.

    Article  CAS  PubMed  Google Scholar 

  81. Putcha P, Yu J, Rodriguez-Barrueco R, Saucedo-Cuevas L, Villagrasa P, Murga-Penas E, et al. HDAC6 activity is a non-oncogene addiction hub for inflammatory breast cancers. Breast Cancer Res. 2015;17(1):149.

    Article  PubMed  PubMed Central  Google Scholar 

  82. Zeleke TZ, Pan Q, Chiuzan C, Onishi M, Li Y, Tan H, et al. Network-based assessment of HDAC6 activity predicts preclinical and clinical responses to the HDAC6 inhibitor Ricolinostat in breast cancer. Nat Cancer. 2023;4(2):257–75.

    Article  CAS  PubMed  Google Scholar 

  83. Lu B, Qiu R, Wei J, Wang L, Zhang Q, Li M et al. Phase separation of phospho-HDAC6 drives aberrant chromatin architecture in triple-negative breast cancer. Nat Cancer. 2024.

  84. Zhao H, Zhang XM, Xiao S, Wu ZR, Shi YJ, Xie MJ. HDAC11 is related to breast cancer prognosis and inhibits invasion and proliferation of breast cancer cells. Int J Clin Exp Pathol. 2023;16(7):172–83.

    CAS  PubMed  PubMed Central  Google Scholar 

  85. Leslie PL, Chao YL, Tsai YH, Ghosh SK, Porrello A, Van Swearingen AED, et al. Histone deacetylase 11 Inhibition promotes breast cancer metastasis from lymph nodes. Nat Commun. 2019;10(1):4192.

    Article  PubMed  PubMed Central  Google Scholar 

  86. Elangovan S, Ramachandran S, Venkatesan N, Ananth S, Gnana-Prakasam JP, Martin PM, et al. SIRT1 is essential for oncogenic signaling by Estrogen/estrogen receptor alpha in breast cancer. Cancer Res. 2011;71(21):6654–64.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  87. Santolla MF, Avino S, Pellegrino M, De Francesco EM, De Marco P, Lappano R, et al. SIRT1 is involved in oncogenic signaling mediated by GPER in breast cancer. Cell Death Dis. 2015;6(7):e1834.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  88. Parija M, Prakash S, Krishna BM, Dash S, Mishra SK. SIRT1 mediates breast cancer development and tumorigenesis controlled by estrogen-related receptor beta. Breast Cancer. 2024;31(3):440–55.

    Article  PubMed  Google Scholar 

  89. Jin X, Wei Y, Xu F, Zhao M, Dai K, Shen R, et al. SIRT1 promotes formation of breast cancer through modulating Akt activity. J Cancer. 2018;9(11):2012–23.

    Article  PubMed  PubMed Central  Google Scholar 

  90. Latifkar A, Ling L, Hingorani A, Johansen E, Clement A, Zhang X, et al. Loss of Sirtuin 1 alters the secretome of breast cancer cells by impairing lysosomal integrity. Dev Cell. 2019;49(3):393–408. e7.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  91. Sinha S, Sharma S, Vora J, Shrivastava N. Emerging role of sirtuins in breast cancer metastasis and multidrug resistance: implication for novel therapeutic strategies targeting sirtuins. Pharmacol Res. 2020;158:104880.

    Article  CAS  PubMed  Google Scholar 

  92. McGlynn LM, Zino S, MacDonald AI, Curle J, Reilly JE, Mohammed ZM, et al. SIRT2: tumour suppressor or tumour promoter in operable breast cancer? Eur J Cancer. 2014;50(2):290–301.

    Article  CAS  PubMed  Google Scholar 

  93. Zhou W, Ni TK, Wronski A, Glass B, Skibinski A, Beck A, et al. The SIRT2 deacetylase stabilizes slug to control malignancy of Basal-like breast cancer. Cell Rep. 2016;17(5):1302–17.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  94. Fiskus W, Coothankandaswamy V, Chen J, Ma H, Ha K, Saenz DT, et al. SIRT2 deacetylates and inhibits the peroxidase activity of Peroxiredoxin-1 to sensitize breast cancer cells to oxidant Stress-Inducing agents. Cancer Res. 2016;76(18):5467–78.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  95. Park SH, Ozden O, Liu G, Song HY, Zhu Y, Yan Y, et al. SIRT2-Mediated deacetylation and tetramerization of pyruvate kinase directs Glycolysis and tumor growth. Cancer Res. 2016;76(13):3802–12.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  96. He S, He C, Yuan H, Xiong S, Xiao Z, Chen L. The SIRT 3 expression profile is associated with pathological and clinical outcomes in human breast cancer patients. Cell Physiol Biochem. 2014;34(6):2061–9.

    Article  CAS  PubMed  Google Scholar 

  97. Desouki MM, Doubinskaia I, Gius D, Abdulkadir SA. Decreased mitochondrial SIRT3 expression is a potential molecular biomarker associated with poor outcome in breast cancer. Hum Pathol. 2014;45(5):1071–7.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  98. Finley LW, Carracedo A, Lee J, Souza A, Egia A, Zhang J, et al. SIRT3 opposes reprogramming of cancer cell metabolism through HIF1alpha destabilization. Cancer Cell. 2011;19(3):416–28.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  99. Zou X, Zhu Y, Park SH, Liu G, O’Brien J, Jiang H, et al. SIRT3-Mediated dimerization of IDH2 directs cancer cell metabolism and tumor growth. Cancer Res. 2017;77(15):3990–9.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  100. Du L, Liu X, Ren Y, Li J, Li P, Jiao Q, et al. Loss of SIRT4 promotes the self-renewal of breast cancer stem cells. Theranostics. 2020;10(21):9458–76.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  101. Shi Q, Liu T, Zhang X, Geng J, He X, Nu M, et al. Decreased Sirtuin 4 expression is associated with poor prognosis in patients with invasive breast cancer. Oncol Lett. 2016;12(4):2606–12.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  102. Xing J, Li J, Fu L, Gai J, Guan J, Li Q. SIRT4 enhances the sensitivity of ER-positive breast cancer to Tamoxifen by inhibiting the IL-6/STAT3 signal pathway. Cancer Med. 2019;8(16):7086–97.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  103. Greene KS, Lukey MJ, Wang X, Blank B, Druso JE, Lin MJ, et al. SIRT5 stabilizes mitochondrial glutaminase and supports breast cancer tumorigenesis. Proc Natl Acad Sci U S A. 2019;116(52):26625–32.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  104. He S, Jia Q, Zhou L, Wang Z, Li M. SIRT5 is involved in the proliferation and metastasis of breast cancer by promoting aerobic Glycolysis. Pathol Res Pract. 2022;235:153943.

    Article  CAS  PubMed  Google Scholar 

  105. Andreani C, Bartolacci C, Persico G, Casciaro F, Amatori S, Fanelli M, et al. SIRT6 promotes metastasis and relapse in HER2-positive breast cancer. Sci Rep. 2023;13(1):22000.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  106. Becherini P, Caffa I, Piacente F, Damonte P, Vellone VG, Passalacqua M, et al. SIRT6 enhances oxidative phosphorylation in breast cancer and promotes mammary tumorigenesis in mice. Cancer Metab. 2021;9(1):6.

    Article  PubMed  PubMed Central  Google Scholar 

  107. Tang X, Li G, Su F, Cai Y, Shi L, Meng Y, et al. HDAC8 cooperates with SMAD3/4 complex to suppress SIRT7 and promote cell survival and migration. Nucleic Acids Res. 2020;48(6):2912–23.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  108. Tang X, Shi L, Xie N, Liu Z, Qian M, Meng F, et al. SIRT7 antagonizes TGF-beta signaling and inhibits breast cancer metastasis. Nat Commun. 2017;8(1):318.

    Article  PubMed  PubMed Central  Google Scholar 

  109. Grozinger CM, Hassig CA, Schreiber SL. Three proteins define a class of human histone deacetylases related to yeast Hda1p. Proc Natl Acad Sci U S A. 1999;96(9):4868–73.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  110. Dancy BM, Cole PA. Protein lysine acetylation by p300/CBP. Chem Rev. 2015;115(6):2419–52.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  111. Wang F, Marshall CB, Ikura M. Transcriptional/epigenetic regulator CBP/p300 in tumorigenesis: structural and functional versatility in target recognition. Cell Mol Life Sci. 2013;70(21):3989–4008.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  112. Lahm A, Paolini C, Pallaoro M, Nardi MC, Jones P, Neddermann P, et al. Unraveling the hidden catalytic activity of vertebrate class IIa histone deacetylases. Proc Natl Acad Sci U S A. 2007;104(44):17335–40.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  113. Zhao X, Sternsdorf T, Bolger TA, Evans RM, Yao TP. Regulation of MEF2 by histone deacetylase 4- and SIRT1 deacetylase-mediated lysine modifications. Mol Cell Biol. 2005;25(19):8456–64.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  114. Zhang Y, Li N, Caron C, Matthias G, Hess D, Khochbin S, et al. HDAC-6 interacts with and deacetylates tubulin and microtubules in vivo. EMBO J. 2003;22(5):1168–79.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  115. Hai Y, Shinsky SA, Porter NJ, Christianson DW. Histone deacetylase 10 structure and molecular function as a polyamine deacetylase. Nat Commun. 2017;8:15368.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  116. Vaquero A, Scher M, Erdjument-Bromage H, Tempst P, Serrano L, Reinberg D. SIRT1 regulates the histone methyl-transferase SUV39H1 during heterochromatin formation. Nature. 2007;450(7168):440–4.

    Article  CAS  PubMed  Google Scholar 

  117. Tennen RI, Chua KF. Chromatin regulation and genome maintenance by mammalian SIRT6. Trends Biochem Sci. 2011;36(1):39–46.

    Article  CAS  PubMed  Google Scholar 

  118. Feige JN, Auwerx J. Transcriptional targets of sirtuins in the coordination of mammalian physiology. Curr Opin Cell Biol. 2008;20(3):303–9.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  119. North BJ, Marshall BL, Borra MT, Denu JM, Verdin E. The human Sir2 ortholog, SIRT2, is an NAD+-dependent tubulin deacetylase. Mol Cell. 2003;11(2):437–44.

    Article  CAS  PubMed  Google Scholar 

  120. Mao Z, Hine C, Tian X, Van Meter M, Au M, Vaidya A, et al. SIRT6 promotes DNA repair under stress by activating PARP1. Science. 2011;332(6036):1443–6.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  121. Villagra A, Cheng F, Wang HW, Suarez I, Glozak M, Maurin M, et al. The histone deacetylase HDAC11 regulates the expression of Interleukin 10 and immune tolerance. Nat Immunol. 2009;10(1):92–100.

    Article  CAS  PubMed  Google Scholar 

  122. Cao J, Sun L, Aramsangtienchai P, Spiegelman NA, Zhang X, Huang W, et al. HDAC11 regulates type I interferon signaling through defatty-acylation of SHMT2. Proc Natl Acad Sci U S A. 2019;116(12):5487–92.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  123. Damaskos C, Garmpis N, Valsami S, Kontos M, Spartalis E, Kalampokas T, et al. Histone deacetylase inhibitors: an attractive therapeutic strategy against breast cancer. Anticancer Res. 2017;37(1):35–46.

    Article  CAS  PubMed  Google Scholar 

  124. Mann BS, Johnson JR, Cohen MH, Justice R, Pazdur R. FDA approval summary: Vorinostat for treatment of advanced primary cutaneous T-cell lymphoma. Oncologist. 2007;12(10):1247–52.

    Article  CAS  PubMed  Google Scholar 

  125. Wawruszak A, Borkiewicz L, Okon E, Kukula-Koch W, Afshan S, Halasa M. Vorinostat (SAHA) and breast cancer: an overview. Cancers (Basel). 2021;13:18.

    Article  Google Scholar 

  126. Yi X, Wei W, Wang SY, Du ZY, Xu YJ, Yu XD. Histone deacetylase inhibitor SAHA induces ERalpha degradation in breast cancer MCF-7 cells by CHIP-mediated ubiquitin pathway and inhibits survival signaling. Biochem Pharmacol. 2008;75(9):1697–705.

    Article  CAS  PubMed  Google Scholar 

  127. Luu TH, Morgan RJ, Leong L, Lim D, McNamara M, Portnow J, et al. A phase II trial of Vorinostat (suberoylanilide hydroxamic acid) in metastatic breast cancer: a California cancer consortium study. Clin Cancer Res. 2008;14(21):7138–42.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  128. Munster PN, Thurn KT, Thomas S, Raha P, Lacevic M, Miller A, et al. A phase II study of the histone deacetylase inhibitor Vorinostat combined with Tamoxifen for the treatment of patients with hormone therapy-resistant breast cancer. Br J Cancer. 2011;104(12):1828–35.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  129. Tu Y, Hershman DL, Bhalla K, Fiskus W, Pellegrino CM, Andreopoulou E, et al. A phase I-II study of the histone deacetylase inhibitor Vorinostat plus sequential weekly Paclitaxel and doxorubicin-cyclophosphamide in locally advanced breast cancer. Breast Cancer Res Treat. 2014;146(1):145–52.

    Article  CAS  PubMed  Google Scholar 

  130. Munster PN, Marchion D, Thomas S, Egorin M, Minton S, Springett G, et al. Phase I trial of Vorinostat and doxorubicin in solid tumours: histone deacetylase 2 expression as a predictive marker. Br J Cancer. 2009;101(7):1044–50.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  131. Lee HZ, Kwitkowski VE, Del Valle PL, Ricci MS, Saber H, Habtemariam BA, et al. FDA approval: Belinostat for the treatment of patients with relapsed or refractory peripheral T-cell lymphoma. Clin Cancer Res. 2015;21(12):2666–70.

    Article  CAS  PubMed  Google Scholar 

  132. Hsu KW, Huang CY, Tam KW, Lin CY, Huang LC, Lin CL et al. The application of Non-Invasive apoptosis detection sensor (NIADS) on histone deacetylation inhibitor (HDACi)-Induced breast cancer cell death. Int J Mol Sci. 2018;19(2).

  133. Marijon H, Lee DH, Ding L, Sun H, Gery S, de Gramont A, et al. Co-targeting poly(ADP-ribose) polymerase (PARP) and histone deacetylase (HDAC) in triple-negative breast cancer: higher synergism in BRCA mutated cells. Biomed Pharmacother. 2018;99:543–51.

    Article  CAS  PubMed  Google Scholar 

  134. Frye R, Myers M, Axelrod KC, Ness EA, Piekarz RL, Bates SE, et al. Romidepsin: a new drug for the treatment of cutaneous T-cell lymphoma. Clin J Oncol Nurs. 2012;16(2):195–204.

    Article  PubMed  PubMed Central  Google Scholar 

  135. Robertson FM, Chu K, Boley KM, Ye Z, Liu H, Wright MC, et al. The class I HDAC inhibitor Romidepsin targets inflammatory breast cancer tumor emboli and synergizes with Paclitaxel to inhibit metastasis. J Exp Ther Oncol. 2013;10(3):219–33.

    CAS  PubMed  Google Scholar 

  136. Pattarawat P, Hunt JT, Poloway J, Archibald CJ, Wang HR. A triple combination gemcitabine + romidepsin + cisplatin to effectively control triple-negative breast cancer tumor development, recurrence, and metastasis. Cancer Chemother Pharmacol. 2021;88(3):415–25.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  137. Sabnis GJ, Goloubeva O, Chumsri S, Nguyen N, Sukumar S, Brodie AM. Functional activation of the Estrogen receptor-alpha and aromatase by the HDAC inhibitor entinostat sensitizes ER-negative tumors to letrozole. Cancer Res. 2011;71(5):1893–903.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  138. Schech A, Kazi A, Yu S, Shah P, Sabnis G. Histone deacetylase inhibitor entinostat inhibits Tumor-Initiating cells in Triple-Negative breast cancer cells. Mol Cancer Ther. 2015;14(8):1848–57.

    Article  CAS  PubMed  Google Scholar 

  139. Lee J, Bartholomeusz C, Mansour O, Humphries J, Hortobagyi GN, Ordentlich P, et al. A class I histone deacetylase inhibitor, Entinostat, enhances lapatinib efficacy in HER2-overexpressing breast cancer cells through FOXO3-mediated Bim1 expression. Breast Cancer Res Treat. 2014;146(2):259–72.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  140. Sabnis GJ, Goloubeva OG, Kazi AA, Shah P, Brodie AH. HDAC inhibitor entinostat restores responsiveness of letrozole-resistant MCF-7Ca xenografts to aromatase inhibitors through modulation of Her-2. Mol Cancer Ther. 2013;12(12):2804–16.

    Article  CAS  PubMed  Google Scholar 

  141. Connolly RM, Zhao F, Miller KD, Lee MJ, Piekarz RL, Smith KL, et al. E2112: randomized phase III trial of endocrine therapy plus entinostat or placebo in hormone Receptor-Positive advanced breast cancer. A trial of the ECOG-ACRIN cancer.Research group. J Clin Oncol. 2021;39(28):3171–81.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  142. Connolly RM, Li H, Jankowitz RC, Zhang Z, Rudek MA, Jeter SC, et al. Combination epigenetic therapy in advanced breast cancer with 5-Azacitidine and Entinostat: A phase II National cancer institute/stand up to cancer study. Clin Cancer Res. 2017;23(11):2691–701.

    Article  CAS  PubMed  Google Scholar 

  143. Roussos Torres ET, Rafie C, Wang C, Lim D, Brufsky A, LoRusso P, et al. Phase I study of entinostat and nivolumab with or without ipilimumab in advanced solid tumors (ETCTN-9844). Clin Cancer Res. 2021;27(21):5828–37.

    Article  CAS  PubMed  Google Scholar 

  144. Li J. Chidamide enhances cytotoxicity of doxorubicin by promoting autophagy and apoptosis in breast cancer. BMC Cancer. 2023;23(1):353.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  145. Zhao YX, Wang H, Zhang SW, Zhang WX, Jiang YZ, Shao ZM. Enhancing therapeutic efficacy in luminal androgen receptor triple-negative breast cancer: exploring Chidamide and enzalutamide as a promising combination strategy. Cancer Cell Int. 2024;24(1):131.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  146. Li D, Jin Y, Lin M, Zeng C, Guo Q, Liu Y, et al. Treatment patterns and clinical outcomes of Chidamide combined with endocrine therapy in hormone receptor-positive, HER2-negative metastatic breast cancer: A real-world multicenter study. Cancer Med. 2024;13(4):e6762.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  147. Suo J, Zhu K, Zhuang C, Zhong X, Bravaccini S, Maltoni R, et al. Efficacy and safety of tucidinostat in patients with advanced hormone receptor-positive human epidermal growth factor receptor 2-negative breast cancer: real-world insights. Ann Transl Med. 2023;11(12):409.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  148. Zhou J, Wu X, Zhang H, Wang X, Yuan Y, Zhang S, et al. Clinical outcomes of tucidinostat-based therapy after prior CDK4/6 inhibitor progression in hormone receptor-positive heavily pretreated metastatic breast cancer. Breast. 2022;66:255–61.

    Article  PubMed  PubMed Central  Google Scholar 

  149. Jiang Z, Li W, Hu X, Zhang Q, Sun T, Cui S, et al. Tucidinostat plus exemestane for postmenopausal patients with advanced, hormone receptor-positive breast cancer (ACE): a randomised, double-blind, placebo-controlled, phase 3 trial. Lancet Oncol. 2019;20(6):806–15.

    Article  CAS  PubMed  Google Scholar 

  150. Tate CR, Rhodes LV, Segar HC, Driver JL, Pounder FN, Burow ME, et al. Targeting triple-negative breast cancer cells with the histone deacetylase inhibitor Panobinostat. Breast Cancer Res. 2012;14(3):R79.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  151. Wang X, Yin X. Panobinostat inhibits breast cancer progression via Vps34-mediated Exosomal pathway. Hum Cell. 2023;36(1):366–76.

    Article  PubMed  Google Scholar 

  152. Chen S, Ye J, Kijima I, Evans D. The HDAC inhibitor LBH589 (panobinostat) is an inhibitory modulator of aromatase gene expression. Proc Natl Acad Sci U S A. 2010;107(24):11032–7.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  153. Tan WW, Allred JB, Moreno-Aspitia A, Northfelt DW, Ingle JN, Goetz MP, et al. Phase I study of Panobinostat (LBH589) and letrozole in postmenopausal metastatic breast cancer patients. Clin Breast Cancer. 2016;16(2):82–6.

    Article  CAS  PubMed  Google Scholar 

  154. Shan W, Jiang Y, Yu H, Huang Q, Liu L, Guo X, et al. HDAC2 overexpression correlates with aggressive clinicopathological features and DNA-damage response pathway of breast cancer. Am J Cancer Res. 2017;7(5):1213–26.

    CAS  PubMed  PubMed Central  Google Scholar 

  155. Bagu ET, Miah S, Dai C, Spriggs T, Ogunbolude Y, Beaton E, et al. Repression of Fyn-related kinase in breast cancer cells is associated with promoter site-specific CpG methylation. Oncotarget. 2017;8(7):11442–59.

    Article  PubMed  PubMed Central  Google Scholar 

  156. Zhao N, Powell RT, Yuan X, Bae G, Roarty KP, Stossi F, et al. Morphological screening of mesenchymal mammary tumor organoids to identify drugs that reverse epithelial-mesenchymal transition. Nat Commun. 2021;12(1):4262.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  157. Salvador MA, Wicinski J, Cabaud O, Toiron Y, Finetti P, Josselin E, et al. The histone deacetylase inhibitor abexinostat induces cancer stem cells differentiation in breast cancer with low Xist expression. Clin Cancer Res. 2013;19(23):6520–31.

    Article  CAS  PubMed  Google Scholar 

  158. Peck B, Chen CY, Ho KK, Di Fruscia P, Myatt SS, Coombes RC, et al. SIRT inhibitors induce cell death and p53 acetylation through targeting both SIRT1 and SIRT2. Mol Cancer Ther. 2010;9(4):844–55.

    Article  CAS  PubMed  Google Scholar 

  159. Park EY, Woo Y, Kim SJ, Kim DH, Lee EK, De U, et al. Anticancer effects of a new SIRT inhibitor, MHY2256, against human breast cancer MCF-7 cells via regulation of MDM2-p53 binding. Int J Biol Sci. 2016;12(12):1555–67.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  160. Jing H, Hu J, He B, Negron Abril YL, Stupinski J, Weiser K, et al. A SIRT2-Selective inhibitor promotes c-Myc oncoprotein degradation and exhibits broad anticancer activity. Cancer Cell. 2016;29(3):297–310.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  161. Abril YLN, Fernandez IR, Hong JY, Chiang YL, Kutateladze DA, Zhao Q, et al. Pharmacological and genetic perturbation Establish SIRT5 as a promising target in breast cancer. Oncogene. 2021;40(9):1644–58.

    Article  CAS  PubMed  Google Scholar 

  162. Hu Z, Wei F, Su Y, Wang Y, Shen Y, Fang Y, et al. Histone deacetylase inhibitors promote breast cancer metastasis by elevating NEDD9 expression. Signal Transduct Target Ther. 2023;8(1):11.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  163. Harris MA, Savas P, Virassamy B, O’Malley MMR, Kay J, Mueller SN, et al. Towards targeting the breast cancer immune microenvironment. Nat Rev Cancer. 2024;24(8):554–77.

    Article  CAS  PubMed  Google Scholar 

  164. Salemme V, Centonze G, Cavallo F, Defilippi P, Conti L. The crosstalk between tumor cells and the immune microenvironment in breast cancer: implications for immunotherapy. Front Oncol. 2021;11:610303.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  165. Gentles AJ, Newman AM, Liu CL, Bratman SV, Feng W, Kim D, et al. The prognostic landscape of genes and infiltrating immune cells across human cancers. Nat Med. 2015;21(8):938–45.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  166. Danenberg E, Bardwell H, Zanotelli VRT, Provenzano E, Chin SF, Rueda OM, et al. Breast tumor microenvironment structures are associated with genomic features and clinical outcome. Nat Genet. 2022;54(5):660–9.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  167. van Vlerken-Ysla L, Tyurina YY, Kagan VE, Gabrilovich DI. Functional States of myeloid cells in cancer. Cancer Cell. 2023;41(3):490–504.

    Article  PubMed  PubMed Central  Google Scholar 

  168. Wu L, Zhang XH. Tumor-Associated neutrophils and Macrophages-Heterogenous but not chaotic. Front Immunol. 2020;11:553967.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  169. Cha YJ, Koo JS. Role of Tumor-Associated myeloid cells in breast cancer. Cells. 2020;9(8).

  170. Mastio J, Condamine T, Dominguez G, Kossenkov AV, Donthireddy L, Veglia F, et al. Identification of monocyte-like precursors of granulocytes in cancer as a mechanism for accumulation of PMN-MDSCs. J Exp Med. 2019;216(9):2150–69.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  171. Veglia F, Sanseviero E, Gabrilovich DI. Myeloid-derived suppressor cells in the era of increasing myeloid cell diversity. Nat Rev Immunol. 2021;21(8):485–98.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  172. Liu H, Wang Z, Zhou Y, Yang Y. MDSCs in breast cancer: an important enabler of tumor progression and an emerging therapeutic target. Front Immunol. 2023;14:1199273.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  173. Kim IS, Gao Y, Welte T, Wang H, Liu J, Janghorban M, et al. Immuno-subtyping of breast cancer reveals distinct myeloid cell profiles and immunotherapy resistance mechanisms. Nat Cell Biol. 2019;21(9):1113–26.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  174. Alshetaiwi H, Pervolarakis N, McIntyre LL, Ma D, Nguyen Q, Rath JA et al. Defining the emergence of myeloid-derived suppressor cells in breast cancer using single-cell transcriptomics. Sci Immunol. 2020;5(44).

  175. Casbon AJ, Reynaud D, Park C, Khuc E, Gan DD, Schepers K, et al. Invasive breast cancer reprograms early myeloid differentiation in the bone marrow to generate immunosuppressive neutrophils. Proc Natl Acad Sci U S A. 2015;112(6):E566–75.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  176. Hao X, Shen Y, Chen N, Zhang W, Valverde E, Wu L, et al. Osteoprogenitor-GMP crosstalk underpins solid tumor-induced systemic immunosuppression and persists after tumor removal. Cell Stem Cell. 2023;30(5):648–64. e8.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  177. Condamine T, Mastio J, Gabrilovich DI. Transcriptional regulation of myeloid-derived suppressor cells. J Leukoc Biol. 2015;98(6):913–22.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  178. Patel S, Fu S, Mastio J, Dominguez GA, Purohit A, Kossenkov A, et al. Unique pattern of neutrophil migration and function during tumor progression. Nat Immunol. 2018;19(11):1236–47.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  179. Ozbay Kurt FG, Lasser S, Arkhypov I, Utikal J, Umansky V. Enhancing immunotherapy response in melanoma: myeloid-derived suppressor cells as a therapeutic target. J Clin Invest. 2023;133(13).

  180. Kumar S, Wilkes DW, Samuel N, Blanco MA, Nayak A, Alicea-Torres K, et al. DeltaNp63-driven recruitment of myeloid-derived suppressor cells promotes metastasis in triple-negative breast cancer. J Clin Invest. 2018;128(11):5095–109.

    Article  PubMed  PubMed Central  Google Scholar 

  181. Gabrilovich DI, Ostrand-Rosenberg S, Bronte V. Coordinated regulation of myeloid cells by tumours. Nat Rev Immunol. 2012;12(4):253–68.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  182. Li P, Lu M, Shi J, Gong Z, Hua L, Li Q, et al. Lung mesenchymal cells elicit lipid storage in neutrophils that fuel breast cancer lung metastasis. Nat Immunol. 2020;21(11):1444–55.

    Article  PubMed  PubMed Central  Google Scholar 

  183. Welte T, Kim IS, Tian L, Gao X, Wang H, Li J, et al. Oncogenic mTOR signalling recruits myeloid-derived suppressor cells to promote tumour initiation. Nat Cell Biol. 2016;18(6):632–44.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  184. Ouzounova M, Lee E, Piranlioglu R, El Andaloussi A, Kolhe R, Demirci MF, et al. Monocytic and granulocytic myeloid derived suppressor cells differentially regulate Spatiotemporal tumour plasticity during metastatic cascade. Nat Commun. 2017;8:14979.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  185. Cassetta L, Fragkogianni S, Sims AH, Swierczak A, Forrester LM, Zhang H, et al. Human Tumor-Associated macrophage and monocyte transcriptional landscapes reveal Cancer-Specific reprogramming, biomarkers, and therapeutic targets. Cancer Cell. 2019;35(4):588–e60210.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  186. Gallina G, Dolcetti L, Serafini P, De Santo C, Marigo I, Colombo MP, et al. Tumors induce a subset of inflammatory monocytes with immunosuppressive activity on CD8 + T cells. J Clin Invest. 2006;116(10):2777–90.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  187. Qian BZ, Li J, Zhang H, Kitamura T, Zhang J, Campion LR, et al. CCL2 recruits inflammatory monocytes to facilitate breast-tumour metastasis. Nature. 2011;475(7355):222–5.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  188. Hagerling C, Gonzalez H, Salari K, Wang CY, Lin C, Robles I, et al. Immune effector monocyte-neutrophil Cooperation induced by the primary tumor prevents metastatic progression of breast cancer. Proc Natl Acad Sci U S A. 2019;116(43):21704–14.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  189. Hanna RN, Cekic C, Sag D, Tacke R, Thomas GD, Nowyhed H, et al. Patrolling monocytes control tumor metastasis to the lung. Science. 2015;350(6263):985–90.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  190. Lee CR, Lee W, Cho SK, Park SG. Characterization of multiple cytokine combinations and TGF-beta on differentiation and functions of Myeloid-Derived suppressor cells. Int J Mol Sci. 2018;19(3).

  191. Kajihara N, Kobayashi T, Otsuka R, Nio-Kobayashi J, Oshino T, Takahashi M, et al. Tumor-derived interleukin-34 creates an immunosuppressive and chemoresistant tumor microenvironment by modulating myeloid-derived suppressor cells in triple-negative breast cancer. Cancer Immunol Immunother. 2023;72(4):851–64.

    Article  CAS  PubMed  Google Scholar 

  192. Lee EJ, Lee KJ, Jung S, Park KH, Park SI. Mobilization of monocytic myeloid-derived suppressor cells is regulated by PTH1R activation in bone marrow stromal cells. Bone Res. 2023;11(1):22.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  193. Calvert RD, Fleet JC, Fournier PGJ, Juarez P, Burcham GN, Haverkamp JM, et al. Monocytic Myeloid-Derived suppressor cells from tumor tissue are a differentiated cell with limited fate plasticity. Immunohorizons. 2022;6(12):790–806.

    Article  CAS  PubMed  Google Scholar 

  194. Biswas S, Mandal G, Roy Chowdhury S, Purohit S, Payne KK, Anadon C, et al. Exosomes produced by mesenchymal stem cells drive differentiation of myeloid cells into immunosuppressive M2-Polarized macrophages in breast cancer. J Immunol. 2019;203(12):3447–60.

    Article  CAS  PubMed  Google Scholar 

  195. Kumar V, Patel S, Tcyganov E, Gabrilovich DI. The nature of Myeloid-Derived suppressor cells in the tumor microenvironment. Trends Immunol. 2016;37(3):208–20.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  196. Sarkar OS, Donninger H, Al Rayyan N, Chew LC, Stamp B, Zhang X, et al. Monocytic MDSCs exhibit superior immune suppression via adenosine and depletion of adenosine improves efficacy of immunotherapy. Sci Adv. 2023;9(26):eadg3736.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  197. Bergenfelz C, Roxa A, Mehmeti M, Leandersson K, Larsson AM. Clinical relevance of systemic monocytic-MDSCs in patients with metastatic breast cancer. Cancer Immunol Immunother. 2020;69(3):435–48.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  198. DeNardo DG, Ruffell B. Macrophages as regulators of tumour immunity and immunotherapy. Nat Rev Immunol. 2019;19(6):369–82.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  199. Pan Y, Yu Y, Wang X, Zhang T. Tumor-Associated macrophages in tumor immunity. Front Immunol. 2020;11:583084.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  200. Ma RY, Black A, Qian BZ. Macrophage diversity in cancer revisited in the era of single-cell omics. Trends Immunol. 2022;43(7):546–63.

    Article  CAS  PubMed  Google Scholar 

  201. Williams CB, Yeh ES, Soloff AC. Tumor-associated macrophages: unwitting accomplices in breast cancer malignancy. NPJ Breast Cancer. 2016;2:15025.

    Article  PubMed  PubMed Central  Google Scholar 

  202. Hirano R, Okamoto K, Shinke M, Sato M, Watanabe S, Watanabe H, et al. Tissue-resident macrophages are major tumor-associated macrophage resources, contributing to early TNBC development, recurrence, and metastases. Commun Biol. 2023;6(1):144.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  203. Ojalvo LS, King W, Cox D, Pollard JW. High-density gene expression analysis of tumor-associated macrophages from mouse mammary tumors. Am J Pathol. 2009;174(3):1048–64.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  204. Rodriguez PC, Quiceno DG, Zabaleta J, Ortiz B, Zea AH, Piazuelo MB, et al. Arginase I production in the tumor microenvironment by mature myeloid cells inhibits T-cell receptor expression and antigen-specific T-cell responses. Cancer Res. 2004;64(16):5839–49.

    Article  CAS  PubMed  Google Scholar 

  205. Dinapoli MR, Calderon CL, Lopez DM. The altered tumoricidal capacity of macrophages isolated from tumor-bearing mice is related to reduce expression of the inducible nitric oxide synthase gene. J Exp Med. 1996;183(4):1323–9.

    Article  CAS  PubMed  Google Scholar 

  206. Doedens AL, Stockmann C, Rubinstein MP, Liao D, Zhang N, DeNardo DG, et al. Macrophage expression of hypoxia-inducible factor-1 alpha suppresses T-cell function and promotes tumor progression. Cancer Res. 2010;70(19):7465–75.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  207. Leek RD, Talks KL, Pezzella F, Turley H, Campo L, Brown NS, et al. Relation of hypoxia-inducible factor-2 alpha (HIF-2 alpha) expression in tumor-infiltrative macrophages to tumor angiogenesis and the oxidative thymidine phosphorylase pathway in human breast cancer. Cancer Res. 2002;62(5):1326–9.

    CAS  PubMed  Google Scholar 

  208. Leek RD, Lewis CE, Whitehouse R, Greenall M, Clarke J, Harris AL. Association of macrophage infiltration with angiogenesis and prognosis in invasive breast carcinoma. Cancer Res. 1996;56(20):4625–9.

    CAS  PubMed  Google Scholar 

  209. Murdoch C, Muthana M, Coffelt SB, Lewis CE. The role of myeloid cells in the promotion of tumour angiogenesis. Nat Rev Cancer. 2008;8(8):618–31.

    Article  CAS  PubMed  Google Scholar 

  210. Cheng S, Li Z, Gao R, Xing B, Gao Y, Yang Y, et al. A pan-cancer single-cell transcriptional atlas of tumor infiltrating myeloid cells. Cell. 2021;184(3):792–e80923.

    Article  CAS  PubMed  Google Scholar 

  211. Yang J, Liao D, Chen C, Liu Y, Chuang TH, Xiang R, et al. Tumor-associated macrophages regulate murine breast cancer stem cells through a novel paracrine EGFR/Stat3/Sox-2 signaling pathway. Stem Cells. 2013;31(2):248–58.

    Article  CAS  PubMed  Google Scholar 

  212. Lu H, Clauser KR, Tam WL, Frose J, Ye X, Eaton EN, et al. A breast cancer stem cell niche supported by juxtacrine signalling from monocytes and macrophages. Nat Cell Biol. 2014;16(11):1105–17.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  213. Mantovani A, Allavena P, Marchesi F, Garlanda C. Macrophages as tools and targets in cancer therapy. Nat Rev Drug Discov. 2022;21(11):799–820.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  214. Rannikko JH, Hollmen M. Clinical landscape of macrophage-reprogramming cancer immunotherapies. Br J Cancer. 2024;131(4):627–40.

    Article  PubMed  PubMed Central  Google Scholar 

  215. Nutt SL, Chopin M. Transcriptional networks driving dendritic cell differentiation and function. Immunity. 2020;52(6):942–56.

    Article  CAS  PubMed  Google Scholar 

  216. Wculek SK, Cueto FJ, Mujal AM, Melero I, Krummel MF, Sancho D. Dendritic cells in cancer immunology and immunotherapy. Nat Rev Immunol. 2020;20(1):7–24.

    Article  CAS  PubMed  Google Scholar 

  217. Wylie B, Macri C, Mintern JD, Waithman J. Dendritic cells and cancer: from biology to therapeutic intervention. Cancers (Basel). 2019;11(4).

  218. Ghirelli C, Reyal F, Jeanmougin M, Zollinger R, Sirven P, Michea P, et al. Breast cancer Cell-Derived GM-CSF licenses regulatory Th2 induction by plasmacytoid predendritic cells in aggressive disease subtypes. Cancer Res. 2015;75(14):2775–87.

    Article  CAS  PubMed  Google Scholar 

  219. Sisirak V, Vey N, Goutagny N, Renaudineau S, Malfroy M, Thys S, et al. Breast cancer-derived transforming growth factor-beta and tumor necrosis factor-alpha compromise interferon-alpha production by tumor-associated plasmacytoid dendritic cells. Int J Cancer. 2013;133(3):771–8.

    Article  CAS  PubMed  Google Scholar 

  220. Sisirak V, Faget J, Gobert M, Goutagny N, Vey N, Treilleux I, et al. Impaired IFN-alpha production by plasmacytoid dendritic cells favors regulatory T-cell expansion that May contribute to breast cancer progression. Cancer Res. 2012;72(20):5188–97.

    Article  CAS  PubMed  Google Scholar 

  221. Faget J, Bendriss-Vermare N, Gobert M, Durand I, Olive D, Biota C, et al. ICOS-ligand expression on plasmacytoid dendritic cells supports breast cancer progression by promoting the accumulation of immunosuppressive CD4 + T cells. Cancer Res. 2012;72(23):6130–41.

    Article  CAS  PubMed  Google Scholar 

  222. Hurwitz AA, Watkins SK. Immune suppression in the tumor microenvironment: a role for dendritic cell-mediated tolerization of T cells. Cancer Immunol Immunother. 2012;61(2):289–93.

    Article  PubMed  PubMed Central  Google Scholar 

  223. Sisirak V, Faget J, Vey N, Blay JY, Menetrier-Caux C, Caux C, et al. Plasmacytoid dendritic cells deficient in IFNalpha production promote the amplification of FOXP3(+) regulatory T cells and are associated with poor prognosis in breast cancer patients. Oncoimmunology. 2013;2(1):e22338.

    Article  PubMed  PubMed Central  Google Scholar 

  224. Michea P, Noel F, Zakine E, Czerwinska U, Sirven P, Abouzid O, et al. Adjustment of dendritic cells to the breast-cancer microenvironment is subset specific. Nat Immunol. 2018;19(8):885–97.

    Article  CAS  PubMed  Google Scholar 

  225. de Mingo Pulido A, Gardner A, Hiebler S, Soliman H, Rugo HS, Krummel MF, et al. TIM-3 regulates CD103(+) dendritic cell function and response to chemotherapy in breast cancer. Cancer Cell. 2018;33(1):60–74. e6.

    Article  PubMed  PubMed Central  Google Scholar 

  226. Ramos RN, Chin LS, Dos Santos AP, Bergami-Santos PC, Laginha F, Barbuto JA. Monocyte-derived dendritic cells from breast cancer patients are biased to induce CD4 + CD25 + Foxp3 + regulatory T cells. J Leukoc Biol. 2012;92(3):673–82.

    Article  CAS  PubMed  Google Scholar 

  227. Sasidharan Nair V, Saleh R, Toor SM, Taha RZ, Ahmed AA, Kurer MA, et al. Transcriptomic profiling disclosed the role of DNA methylation and histone modifications in tumor-infiltrating myeloid-derived suppressor cell subsets in colorectal cancer. Clin Epigenetics. 2020;12(1):13.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  228. Youn JI, Kumar V, Collazo M, Nefedova Y, Condamine T, Cheng P, et al. Epigenetic Silencing of retinoblastoma gene regulates pathologic differentiation of myeloid cells in cancer. Nat Immunol. 2013;14(3):211–20.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  229. de Almeida Nagata DE, Chiang EY, Jhunjhunwala S, Caplazi P, Arumugam V, Modrusan Z, et al. Regulation of Tumor-Associated myeloid cell activity by CBP/EP300 bromodomain modulation of H3K27 acetylation. Cell Rep. 2019;27(1):269–81. e4.

    Article  PubMed  Google Scholar 

  230. Yu B, Luo F, Sun B, Liu W, Shi Q, Cheng SY, et al. KAT6A acetylation of SMAD3 regulates Myeloid-Derived suppressor cell recruitment, metastasis, and immunotherapy in Triple-Negative breast cancer. Adv Sci (Weinh). 2021;8(20):e2100014.

    Article  PubMed  Google Scholar 

  231. Adeshakin AO, Adeshakin FO, Yan D, Wan X. Regulating histone deacetylase signaling pathways of Myeloid-Derived suppressor cells enhanced T Cell-Based immunotherapy. Front Immunol. 2022;13:781660.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  232. Lu Z, Zou J, Li S, Topper MJ, Tao Y, Zhang H, et al. Epigenetic therapy inhibits metastases by disrupting premetastatic niches. Nature. 2020;579(7798):284–90.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  233. Wang HF, Ning F, Liu ZC, Wu L, Li ZQ, Qi YF, et al. Histone deacetylase inhibitors deplete myeloid-derived suppressor cells induced by 4T1 mammary tumors in vivo and in vitro. Cancer Immunol Immunother. 2017;66(3):355–66.

    Article  CAS  PubMed  Google Scholar 

  234. Kim K, Skora AD, Li Z, Liu Q, Tam AJ, Blosser RL, et al. Eradication of metastatic mouse cancers resistant to immune checkpoint Blockade by suppression of myeloid-derived cells. Proc Natl Acad Sci U S A. 2014;111(32):11774–9.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  235. Guerriero JL, Sotayo A, Ponichtera HE, Castrillon JA, Pourzia AL, Schad S, et al. Class IIa HDAC Inhibition reduces breast tumours and metastases through anti-tumour macrophages. Nature. 2017;543(7645):428–32.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  236. Li X, Su X, Liu R, Pan Y, Fang J, Cao L, et al. HDAC Inhibition potentiates anti-tumor activity of macrophages and enhances anti-PD-L1-mediated tumor suppression. Oncogene. 2021;40(10):1836–50.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  237. Chang YC, Chen TC, Lee CT, Yang CY, Wang HW, Wang CC, et al. Epigenetic control of MHC class II expression in tumor-associated macrophages by decoy receptor 3. Blood. 2008;111(10):5054–63.

    Article  CAS  PubMed  Google Scholar 

  238. Xu G, Niu L, Wang Y, Yang G, Zhu X, Yao Y, et al. HDAC6-dependent deacetylation of TAK1 enhances sIL-6R release to promote macrophage M2 polarization in colon cancer. Cell Death Dis. 2022;13(10):888.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  239. Banik D, Noonepalle S, Hadley M, Palmer E, Gracia-Hernandez M, Zevallos-Delgado C, et al. HDAC6 plays a noncanonical role in the regulation of antitumor immune responses, dissemination, and invasiveness of breast cancer. Cancer Res. 2020;80(17):3649–62.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  240. Wang YC, Wu YS, Hung CY, Wang SA, Young MJ, Hsu TI, et al. USP24 induces IL-6 in tumor-associated microenvironment by stabilizing p300 and beta-TrCP and promotes cancer malignancy. Nat Commun. 2018;9(1):3996.

    Article  PubMed  PubMed Central  Google Scholar 

  241. Chauvistre H, Kustermann C, Rehage N, Klisch T, Mitzka S, Felker P, et al. Dendritic cell development requires histone deacetylase activity. Eur J Immunol. 2014;44(8):2478–88.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  242. De Sa Fernandes C, Novoszel P, Gastaldi T, Krauss D, Lang M, Rica R, et al. The histone deacetylase HDAC1 controls dendritic cell development and anti-tumor immunity. Cell Rep. 2024;43(6):114308.

    Article  PubMed  Google Scholar 

  243. Ning Y, Ding J, Sun X, Xie Y, Su M, Ma C et al. HDAC9 deficiency promotes tumor progression by decreasing the CD8(+) dendritic cell infiltration of the tumor microenvironment. J Immunother Cancer. 2020;8(1).

  244. Cheng F, Lienlaf M, Wang HW, Perez-Villarroel P, Lee C, Woan K, et al. A novel role for histone deacetylase 6 in the regulation of the tolerogenic STAT3/IL-10 pathway in apcs. J Immunol. 2014;193(6):2850–62.

    Article  CAS  PubMed  Google Scholar 

Download references

Acknowledgements

The figure is created in BioRender.

Funding

JMR is supported by NCI CA148761 and Komen SAC232150.

Author information

Authors and Affiliations

Authors

Contributions

X.Y. wrote the manuscript. J.R. reviewed and edited the manuscript.

Corresponding author

Correspondence to Jeffrey M. Rosen.

Ethics declarations

Ethics approval and consent to participate

Not applicable.

Competing interests

The authors declare no competing interests.

Additional information

Publisher’s note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Rights and permissions

Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article’s Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article’s Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by/4.0/.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Yuan, X., Rosen, J.M. Histone acetylation modulators in breast cancer. Breast Cancer Res 27, 49 (2025). https://doiorg.publicaciones.saludcastillayleon.es/10.1186/s13058-025-02006-9

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doiorg.publicaciones.saludcastillayleon.es/10.1186/s13058-025-02006-9

Keywords