Skip to main content
Erschienen in: Journal of Experimental & Clinical Cancer Research 1/2019

Open Access 01.12.2019 | Review

STAT3 as a potential therapeutic target in triple negative breast cancer: a systematic review

verfasst von: Jiang-Jiang Qin, Li Yan, Jia Zhang, Wei-Dong Zhang

Erschienen in: Journal of Experimental & Clinical Cancer Research | Ausgabe 1/2019

Abstract

Triple negative breast cancer (TNBC), which is typically lack of expression of estrogen receptor (ER), progesterone receptor (PR), and human epidermal growth factor receptor 2 (HER2), represents the most aggressive and mortal subtype of breast cancer. Currently, only a few treatment options are available for TNBC due to the absence of molecular targets, which underscores the need for developing novel therapeutic and preventive approaches for this disease. Recent evidence from clinical trials and preclinical studies has demonstrated a pivotal role of signal transducer and activator of transcription 3 (STAT3) in the initiation, progression, metastasis, and immune evasion of TNBC. STAT3 is overexpressed and constitutively activated in TNBC cells and contributes to cell survival, proliferation, cell cycle progression, anti-apoptosis, migration, invasion, angiogenesis, chemoresistance, immunosuppression, and stem cells self-renewal and differentiation by regulating the expression of its downstream target genes. STAT3 small molecule inhibitors have been developed and shown excellent anticancer activities in in vitro and in vivo models of TNBC. This review discusses the recent advances in the understanding of STAT3, with a focus on STAT3’s oncogenic role in TNBC. The current targeting strategies and representative small molecule inhibitors of STAT3 are highlighted. We also propose potential strategies that can be further examined for developing more specific and effective inhibitors for TNBC prevention and therapy.
Hinweise
Jiang-Jiang Qin and Li Yan contributed equally to this work.
Abkürzungen
ABC
ATP-binding cassette
AML
Acute myeloid leukemia
APE/Ref-1
Apurinic/apyrimidinic endonuclease-1/redox factor-1
ATO
Arsenic trioxide
Bax
Bcl-2-associated x protein
Bcl-2
B-cell lymphoma-2
Bcl-xL
B-cell lymphoma-extra Large
bFGF
Basic fibroblast growth factor
CBP
CREB-binding protein
CCD
Coiled-coil domain
CLL
Chronic lymphocytic leukemia
DBD
DNA binding domain
DLBCL
Diffuse large B-cell lymphoma
DRD2
Dopamine receptor D2
EGF
Epidermal growth factor
EGFR
Epidermal growth factor receptor
ER
Estrogen receptor
EZH2
Enhancer of zeste homolog 2
FAK
Focal adhesion kinase
FGFR
Fibroblast growth factor receptor
GGNBP2
Gametogenetin-binding protein 2
GIC
Gastrointestinal cancer
gp130
Glycoprotein 130
GPER
G protein-coupled estrogen receptor
GPER-1
G protein-coupled estrogen receptor-1
GRIM-19
Gene associated with retinoic-interferon-induced mortality 19
HCC
Hepatocellular carcinoma
HER2
Human epidermal growth factor receptor 2
HGF
Hepatocyte growth factor
HIF-1α
Hypoxia-inducible factor 1-alpha
HN1L
Hematological and neurological expressed 1-like
HNSCC
Head and neck squamous cell carcinoma
HSP90
Heat shock protein 90
IGF-1R
IGF-1 receptor
IGFR
Insulin-like growth factor receptor
IL-10R
Interleukin-10 receptor
IL-22
Interleukin-22
IL-6
Interleukin-6
IL-6R
Interleukin-6 receptor
IR
Ionizing irradiation
JAKs
Janus kinases
LECs
Lymphatic endothelial cells
LEPRb
Long form of leptin receptor
LVs
Lymphatic vessels
MCC
Metastatic colorectal cancer
miRNA
MicroRNA
MMP
Matrix metalloproteinase
MRP2
Multidrug resistance protein 2
MSK1
Mitogen- and stress-activated protein kinase-1
MSM
Methylsulfonyl-methane
NF-κB
Nuclear factor-kappa B
NHL
Non-Hodgkin lymphoma
NSCLC
Non-small cell lung cancer
Oct-4
Octamer-binding transcription factor-4
PARP
Poly (ADP-ribose) polymerase
pATF2
Phosphorylated activating transcription factor 2
pc-Jun
Phosphorylated c-Jun
PD-L1
Programmed death ligand 1
PGG
1,2,3,4,6-penta-O-galloyl-beta-D-glucose
PIAS
Protein inhibitor of activated STAT
PR
Progesterone receptor
PROTACs
Proteolysis targeting chimeras
pSTAT3
Phosphorylated STAT3
PTPRT
Protein tyrosine phosphatase receptor T
PTPs
Protein tyrosine phosphatases
ROS
Reactive oxygen species
SH2
SRC homology 2
SHP-1/2
Src homology domain-containing tyrosine phosphatases 1/2
SLL
Small lymphocytic leukemia
SMYD2
SET and MYND domain 2
SOCS
Suppressors of cytokine signaling
Sox2
SRY-related HMG-box 2
STAT3
Signal transducer and activator of transcription 3
TAD
Transactivation domain
TAK1
Transforming growth factor β-activated kinase 1
TC-PTP
T-cell protein-tyrosine phosphatase
tDRs
Transfer RNA-derived fragments
TNBC
Triple negative breast cancer
TNFRSF1A
Tumor necrosis factor receptor superfamily member 1A
TRAF6
Tumor necrosis factor receptor-associated factor 6
UTR
3′-untranslated region
VEGF
Vascular endothelial growth factor
Wwox
WW domain-containing oxidoreductase
XPO1
Exportin 1

Background

Triple negative breast cancer (TNBC) is the most aggressive form of breast cancer and accounts for much higher recurrence and metastasis rates [1]. Due to the absence of the expression of estrogen receptor (ER), progesterone receptor (PR), and human epidermal growth factor receptor 2 (HER2), TNBC is unresponsive to endocrine and HER2-targeted therapies, which results in the high mortality of patients with this disease [1]. When patients are diagnosed with TNBC at the early stage, combination chemotherapy (anthracyclines, taxanes, platinum salts, etc.) with or without radiotherapy is used as standard non-surgical therapy and has shown some efficacy in patients with both primary and metastatic diseases [2]. Because of the inter- and the intratumoral heterogeneities of TNBC, the intrinsic chemoresistance as well as severe side effects are often observed and lead to limited success in the clinic [3, 4]. Targeted therapies (e.g., poly (ADP-ribose) polymerase (PARP) inhibitors and epidermal growth factor receptor (EGFR) inhibitors) and immunotherapies have also shown some promise in preliminary clinical studies, but further investigations are critically needed [57]. More recently, many efforts have been made to identify targetable molecules for treating TNBC via genomic profiling and several critical alternations have been discovered, including the overexpression and aberrant activation of signal transducer and activator of transcription 3 (STAT3) [8, 9]. The emerging data suggest that STAT3 may be a potential molecular target and biomarker for TNBC.
The STAT family of transcription factors is comprised of seven members with high structural and functional similarity, including STAT1, STAT2, STAT3, STAT4, STAT5a, STAT5b, and STAT6 [10, 11]. All STAT proteins consist of an amino acid domain (NH2), a coiled-coil domain (CCD) for binding with interactive proteins, a DNA binding domain (DBD), a linker domain, a SRC homology 2 (SH2) domain for phosphorylation and dimerization, and a C-terminal transactivation domain (TAD) [11]. Most of these domains are highly conserved among STAT proteins and only TAD is divergent and mainly contributes to their structure diversity [12]. STAT3 was initially discovered to bind to DNA in response to interleukin-6 (IL-6) and epidermal growth factor (EGF) in 1994 [13, 14]. Over the past decades, STAT3 has become one of the most investigated oncogenic transcription factors and is highly associated with cancer initiation, progression, metastasis, chemoresistance, and immune evasion [15, 16]. The recent evidence from both preclinical and clinical studies have demonstrated that STAT3 plays a critical role in TNBC and STAT3 inhibitors have shown efficacy in inhibiting TNBC tumor growth and metastasis.
Considering that there is an unmet medical need for TNBC treatment and innovative therapeutic agents are urgently required, an in-depth understanding of the roles of STAT3 in TNBC will facilitate the development of STAT3-targeted therapeutics and pave the way for a novel TNBC treatment approach. In this review, we focus on the recent findings related to STAT3’s role in TNBC as well as STAT3 inhibitors and current targeting strategies. We also discuss other potential strategies for developing new STAT3 inhibitors for TNBC treatment.

The STAT3 signaling pathway

The classical STAT3 signaling pathway that is activated through the binding of cytokines or growth factors to their corresponding cell surface receptors has been extensively reviewed [1618]. Here, we present a brief overview of the STAT3 signaling pathway, nonreceptor tyrosine kinases of STAT3, and its intrinsic inhibitors and coactivators, which are depicted in Fig. 1. Briefly, the overexpressed cytokine receptors, e.g., interleukin-6 receptor (IL-6R) and interleukin-10 receptor (IL-10R) and the hyperactive growth factor receptors, e.g., epidermal growth factor receptor (EGFR), fibroblast growth factor receptor (FGFR) and insulin-like growth factor receptor (IGFR) always trigger the tyrosine phosphorylation cascade through the binding of ligands to these receptors, leading to the aberrant activation of STAT3 and the transcription of its downstream target genes [17]. Once the ligands bind to their receptors on the cell surface, these receptors further form dimers and successively recruit glycoprotein 130 (gp130) and Janus kinases (JAKs), thus phosphorylating and activating JAKs [19]. Conversely, the cytoplasmic tyrosine residues of these receptors are phosphorylated by the activated JAKs and then interact with the SH2 domain of STAT3, resulting in STAT3 phosphorylation at Tyr705 by JAKs [16]. In addition, STAT3 can be phosphorylated and activated by several nonreceptor tyrosine kinases, e.g., Src and Abl [20]. The phosphorylated STAT3 (pSTAT3) further forms a homodimer through interaction between their phosphorylated Tyr705 site and SH2 domain, triggering the dissociation of STAT3 dimers from the cell surface receptors and its translocation from cytoplasm to the nucleus [21, 22]. With the help of a variety of coactivator proteins, including NCOA/SRC1a, apurinic/apyrimidinic endonuclease-1/redox factor-1 (APE/Ref-1), and CREB-binding protein (CBP)/p300, the nuclear STAT3 binds to specific DNA sequences and activates the transcription of genes that regulate various phenotypes of cancer cells [17, 18].
STAT3 is also highly expressed in some normal tissues and organs, including the bone marrow, peripheral nervous system, and digestive tract and plays a physiological role [2325]. In the normal physiological conditions, STAT3 phosphorylation and activation are tightly controlled by several intrinsic inhibitors, including protein tyrosine phosphatases (PTPs), the suppressors of cytokine signaling (SOCS), and the protein inhibitor of activated STAT (PIAS) [26]. The Src homology domain-containing tyrosine phosphatases 1/2 (SHP-1/2) directly interact and dephosphorylate JAK and STAT3, resulting in their inactivation [27, 28]. The nuclear PTPs, including TC45 and T-cell protein-tyrosine phosphatase (TC-PTP) induce the inactivation of STAT3 through its dephosphorylation and translocation from nucleus to the cytoplasm [29, 30]. Other PTPs, such as PTP1B and PTPeC have also been reported to regulate STAT3 dephosphorylation and inactivation [31]. Moreover, SOCS directly interacts with JAK and STAT3 and inhibits their phosphorylation and activation via forming a negative feedback loop with JAK-STAT3 signaling pathway [32]. PIAS inhibits the binding of nuclear STAT3 to DNA and induces STAT3 dephosphorylation via protein tyrosine phosphatase receptor T (PTPRT), leading to the reduced expression of its downstream target genes [33]. In addition, the stability of STAT3 protein is also regulated by the ubiquitin-proteasome system via the ubiquitin ligase TRAF6 (tumor necrosis factor receptor-associated factor 6) [34]. Recent studies have also reported that miR-544 directly targets the 3′-untranslated region (UTR) on STAT3 mRNA, thus down-regulating STAT3 expression in TNBC cells [35]. Due to the presence of these endogenous inhibitors, STAT3 is strictly governed to exert its physiological functions in normal cells [36]. Herein, both direct inhibition of STAT3 and activation of the endogenous inhibitors may be considered as potential STAT3-inhibiting strategies for developing novel cancer therapeutics.

The STAT3 signaling pathway in triple negative breast cancer

The oncogenic potential of STAT3 has been widely recognized through its involvement in regulating the expression of genes related to cancer cell proliferation, anti-apoptosis, migration, invasion, angiogenesis, chemoresistance, immune suppression, stem cell self-renewal and maintenance, and autophagy (as shown in Fig. 2) [17, 18]. Importantly, STAT3 is overexpressed and constitutively activated in TNBC, which is highly related to TNBC initiation, progression, metastasis, resistance to chemotherapy, and the poor survival outcomes [8]. STAT3 is not only capable of eliciting the expression of cancer-related genes, but also physically interacts and functionally cooperates with other oncogenic transcription factors, e.g., GLI1, promoting the aggressiveness of TNBC [8]. A recent study has also found a reduction of the gene associated with retinoic-interferon-induced mortality 19 (GRIM-19), an intrinsic inhibitor of STAT3 transcription accompanied by STAT3 overexpression in TNBC [37]. In addition, TCPTP, including two splice variants TC45 and TC48 are down-regulated in TNBC cells in vitro and in vivo, which also contributes to the activation of STAT3 signaling [38]. Indeed, STAT3 has also been found to localize in the mitochondria, where it is termed mitoSTAT3 and regulates the mitochondrial functions, including electron transport chain, ATP synthesis, calcium homeostasis, and reactive oxygen species (ROS) accumulation [39, 40]. Moreover, mitoSTAT3 has been shown to promote breast cancer cell growth, in which the phosphorylation of Serine 727 plays a critical role [41].
A recent study has shown that acetylated STAT3 is highly elevated in TNBC, causing the methylation and inactivation of tumor-suppressor gene promoters [42]. Importantly, mutation of STAT3 at Lys685 or reducing STAT3 acetylation by resveratrol could induce demethylation and activation of the estrogen receptor-α gene and sensitize TNBC cells to antiestrogens. Considering the emerging data that demonstrate the critical role of STAT3 in TNBC, we herein present a comprehensive overview of its oncogenic functions in this section.

Role of STAT3 in TNBC cell proliferation and anti-apoptosis

Several studies have demonstrated that STAT3 promotes cell proliferation and inhibits apoptosis in TNBC by increasing the expression of target genes, including survivin, c-Myc, cyclin D1, B-cell lymphoma-2 (Bcl-2), and B-cell lymphoma-extra large (Bcl-xL) [21]. In TNBC, STAT3 directly binds to the survivin promoter and promotes its transcription [43, 44], which can be blocked by inhibiting the nuclear export factor, exportin 1 (XPO1) and CBP-mediated STAT3 acetylation [45]. In addition, Galectin-1, a β-galactoside binding protein has also been shown to contribute to TNBC progression through binding to integrin β1 and activating the integrin β1/FAK/c-Src/ERK/STAT3/survivin pathway [46]. Conversely, WW domain-containing oxidoreductase (Wwox) inhibits TNBC cell proliferation by interacting with JAK2 and suppressing JAK2 and STAT3 phosphorylation [47]. Wwox also represses the binding of STAT3 to the IL-6 promoter, therefore decreasing the expression of IL-6 cytokine. A tumor suppressor gene, gametogenetin-binding protein 2 (GGNBP2) has been found to inhibit breast cancer cell proliferation and induce apoptosis, independent of ER expression [48]. A further study has indicated that the inhibition of IL-6/STAT3 signaling by GGNBP2 is mainly responsible for its inhibitory effects on TNBC growth and metastasis [48].
STAT3 also promotes TNBC cell proliferation and inhibits apoptosis through the crosstalk with SET and MYND domain 2 (SMYD2) and nuclear factor-kappa B (NF-κB) [49]. SMYD2 is highly expressed in TNBC cell lines and tissues, which is correlated with increased TNBC cell proliferation and survival. Mechanistically, SMYD2 physically interacts with STAT3 and NF-κB p65 and increases their methylation and phosphorylation, promoting tumor growth and metastasis [49]. STAT3 recruits the acetyltransferase p300 to enhance NF-κB acetylation and prolong its nuclear retention [50]. In addition, STAT3 and NF-κB also contribute to each other’s activation via SMYD2 [49]. Interestingly, a recent study has reported an opposite role of STAT3 in TNBC cells [51]. It was observed that STAT3 knockdown did not inhibit but promoted the growth of MDA-MB-231 cells-derived xenograft tumors, implying that the oncogenic role of STAT3 in TNBC might be context-specific [51].

Role of STAT3 in TNBC cell migration and invasion

The role of STAT3 in promoting cell migration and invasion has been linked to the upregulated expression of matrix metalloproteinase 2 (MMP2), MMP9, TWIST, and Vimentin [52]. As discussed earlier, the STAT3 signaling is frequently activated through the binding of cytokines and growth factors to their corresponding receptors in cancer cells. A newly discovered cytokine termed interleukin-22 (IL-22) was recently reported to promote the migration of TNBC cells and induce their chemoresistance by activating the JAK/STAT3/MAPKs/AKT signaling pathway. The increased levels of the IL-22 producing (Th22) cells were also observed in normal, paratumor, and tumor tissues from patients with TNBC, which confirmed the importance of IL-22/JAK/STAT3/MAPKs/AKT in metastasis of this disease [53].
Recent studies reported that several upstream regulators of STAT3 signaling are involved in TNBC metastasis. Wwox blocks JAK2-STAT3 interaction and inhibits STAT3 phosphorylation, therefore repressing STAT3-driven TNBC metastasis [47]. G protein-coupled estrogen receptor (GPER) has been demonstrated as a TNBC metastasis suppressor. Mechanistically, activation of GPER can inhibit the NF-κB/IL-6/STAT3 signals, cause STAT3 dephosphorylation and inactivation, and then suppress migration and angiogenesis of TNBC [54]. GPER also triggers Y397 phosphorylation of focal adhesion kinase (FAK) in TNBC while the activation of both GPER and FAK promotes the migration of TNBC cells by increasing STAT3 nuclear accumulation and gene expression [55].

Role of STAT3 in angiogenesis of TNBC

The pro-angiogenic role of STAT3 has been partially attributed to the upregulation of vascular endothelial growth factor (VEGF), hypoxia-inducible factor 1-alpha (HIF-1α), hepatocyte growth factor (HGF), and basic fibroblast growth factor (bFGF) via STAT3 transactivation [52]. A recent study demonstrated that lymphatic endothelial cells (LECs) promote angiogenesis and metastasis through pSTAT3-mediated CCL5 expression in TNBC [56]. LECs are an important component of lymphatic vessels (LVs), which are prevailingly considered as the routes for cancer metastasis. Lee et al. have found that IL-6 secretion from TNBC cells causes STAT3 phosphorylation and activation, therefore inducing HIF-1α and VEGF expression. pSTAT3 also forms a ternary complex with phosphorylated c-Jun (pc-Jun) and phosphorylated activating transcription factor 2 (pATF2), which induces CCL5 expression in LECs and accelerates metastasis [56]. It was also observed that estrogen activates G protein-coupled estrogen receptor-1 (GPER-1), inhibits the expression VEGF at both protein and mRNA levels, and suppresses the tumor growth and angiogenesis in TNBC xenograft tumor models, in which STAT3 is involved [57].

Role of STAT3 in chemoresistance of TNBC

It has frequently been observed that blocking STAT3 signaling enhances the anticancer activity of chemotherapies in TNBC cells in vitro and in vivo, which endorses a critical role of STAT3 in chemosensitivity of TNBC [5861]. Several recent studies revealed the mechanisms underlying STAT3-mediated chemoresistance in different subsets of TNBC cell lines [62]. NF-κB is highly associated with resistance to cancer therapies, while the overexpression and constitutive activation STAT3-NF-κB signaling pathway have been shown to confer chemoresistance in TNBC cells [63]. Mechanistically, STAT3 upregulates the expression of a target gene TNFRSF1A (tumor necrosis factor receptor superfamily member 1A), which recruits TNFα to the cell surface and triggers the activation of NF-κB signaling pathway [64]. The aberrant activation of STAT3 also increases the expression levels of pluripotency transcription factors octamer-binding transcription factor-4 (Oct-4) and c-Myc, which regulate stemness-mediated doxorubicin resistance in TNBC [65]. The restoration of doxorubicin sensitivity of TNBC cells by a STAT3 inhibitor WP1066 further confirms a pivotal role of this oncogene in chemoresistance.
STAT3-mediated microRNA (miRNA) expression is emerging as a mechanism for regulating chemoresistance in TNBC. Niu et al. found that miR-181a expression is increased in TNBC due to doxorubicin treatment and contributes to acquired resistance and metastasis of this disease through repressing the expression of its target gene Bax (Bcl-2-associated x protein) [66]. Further studies have indicated that pSTAT3 at S727 not only directly binds to MIR181A1 promoter but also recruits MSK1 (mitogen- and stress-activated protein kinase-1) and stabilizes its binding to MIR181A1 promoter, facilitating the transactivation [67]. The effectiveness of targeting STAT3-mediated MIR181A1 transactivation for sensitizing cells to chemotherapy and preventing metastasis has also been validated in a TNBC orthotopic model.
STAT3 is also involved in hypoxia-induced chemoresistance in TNBC [67]. Under hypoxia, the intracellular uptake of chemotherapy, especially cisplatin is dramatically reduced due to the upregulated expression of ATP-binding cassette (ABC) drug transporters. Although the expression level and activity of HIF-1α was increased by hypoxia in TNBC, no significant improvement in chemoresistance was observed in TNBC cells that were treated by HIF-1α siRNA. Intriguingly, STAT3 was found to increase the expression levels of ABC transporters, especially ABCC2 (also known as multidrug resistance protein 2, MRP2) and ABCC6 (also known as MRP6) in hypoxia-treated TNBC cells, therefore conferring chemoresistance to cisplatin [67, 68]. However, another study reported that IL-6-mediated STAT3 activation induces HIF-1α expression in TNBC cells, which consequently attenuates chemotherapy-induced cytotoxicity and cell apoptosis through regulating the expression of apoptosis-related proteins (Bax and Bcl-2) and drug transporters (P-glycoprotein and MRP1) [68]. The transfer RNA-derived fragments (tDRs), particularly tDR-0009 and tDR-7336 are upregulated in TNBC under hypoxia and facilitate the doxorubicin resistance through phosphorylating and activating STAT3 [69]. In addition, the combination treatment with HIF-1α and STAT3 inhibitors significantly enhances the cytotoxicity of cisplatin against TNBC cells and overcomes hypoxia-induced chemoresistance [70]. However, the role of STAT3-induced HIF-1α expression in hypoxia-induced chemoresistance is not clear so far, and further investigation is critically needed.

Role of STAT3 in immune suppression

Recent findings have established STAT3 as a powerful regulator of tumor-mediated immune suppression [21, 71]. STAT3 is not only overexpressed and activated in cancer cells but also in tumor-associated immune cells, inducing the expression of immune-suppression related genes, including IL-6, IL-10, TGF-β and VEGF and driving the escape of cancer cells from immune-mediated elimination [71]. In TNBC, STAT3 and its homolog STAT1 are also involved in regulating the expression of programmed death ligand 1 (PD-L1), a critical immune checkpoint that modulates the magnitude and the functional profile of T cell responses [72]. PD-L1 and PD-L2 are actually also amplified and overexpressed in TNBC cell lines due to JAK-mediated STAT3 phosphorylation and activation [73]. The mechanism studies have shown that pSTAT1 and pSTAT3 form heterodimers in the cytoplasm and translocate into the nucleus, where the pSTAT1-pSTAT3 dimers bind to the PD-L1 promoter and activate its transcription [72]. Another study has shown that syntenin1 is highly expressed in TNBC tissues and increases the expression level of PD-L1 by activating STAT3, consequently attenuates the response of TNBC to anti-PD-L1 treatment [74]. Moreover, direct inhibition of STAT3 overcomes the resistance of TNBC to immunotherapies, which confirms its immunosuppressive activity [72, 74].

Role of STAT3 in TNBC stem cell phenotypes

Early studies on STAT3 signaling disclosed an important role in stem cells self-renewal and differentiation [75]. The increasing evidence has also demonstrated that the constitutive activation of IL-6/STAT3 signaling pathway contributes to the stemness of TNBC stem cells under both normal and hypoxia conditions [76, 77]. In addition, the VEGF-VEGFR-2 binding-induced STAT3 phosphorylation and activation was found to promote the self-renewal of breast cancer cells, especially TNBC cells by upregulating the expression of Myc and Sox2 (SRY-related HMG-box 2) [78]. The crosstalk of STAT3 with NF-κB and Wnt signaling pathways was also observed in TNBC cells and serves as a feed-forward loop for regulating the TNBC stem cell function [79]. Moreover, Syndecan-1 (CD138) is highly expressed in TNBC, especially inflammatory TNBC and contributes to the poor prognosis of this disease [80]. Syndecan-1 was recently reported to promote TNBC stem cells through modulating the STAT3, NF-κB, and Wnt signaling pathways together [76]. Another study by Ibrahim et al. has demonstrated the importance of IL-6/STAT3 signaling pathway in Syndecan-1-modulated cancer stem cell phenotype [81]. Furthermore, Notch and EGFR signaling pathways are also implicated in the modulatory effects of Syndecan-1 on TNBC stem cells [81].
Except for cytokines and growth factors, adipokines, e.g., Leptin are also involved in the constitutive activation of the STAT3 signaling pathway. Leptin and its long form of leptin receptor (LEPRb) are enriched in breast cancer tissues and promote cell proliferation, migration, and angiogenesis [82]. Recently studies have shown that the binding of Leptin to LEPRb initiates the activation of JAK2/STAT3 signaling pathway, which further induces self-renewal and maintains the stem-cell state in TNBC stem cells [83]. Moreover, a new upstream regulator of the LEPR-STAT3 signaling pathway termed hematological and neurological expressed 1-like (HN1L) was also discovered to promote TNBC stem cell properties [84]. HN1L is overexpressed in TNBC tissues and correlates with the shorter survival of patients with this disease. The HN1L silencing experiments further confirmed its regulatory effects on LEPR-STAT3 signaling pathway and on TNBC stem cell population and lung metastasis [84].

Role of STAT3 in autophagy of TNBC cells

Autophagy is capable of regulating STAT3 phosphorylation status in TNBC cells [85]. Maycotte et al. discovered that the autophagy-dependent survival under unstressed conditions is enriched in TNBC, which reduces the response of cancer cells to therapy. Further studies have indicated that autophagy promotes TNBC cell survival by regulating STAT3 phosphorylation and activation [85]. Therefore, pharmacological inhibition of STAT3 may be a promising strategy for treating autophagy-dependent TNBC.

Targeting STAT3 for TNBC prevention and therapy

Abundant evidence has suggested that STAT3 may be a promising molecular target for TNBC therapy [86]. Various STAT3 inhibitors have been developed and shown some efficacy in TNBC models in vitro and in vivo, which have been summarized in Table 1. In this section, we discuss the current STAT3-targeting strategies (as shown in Fig. 3) for treating and preventing TNBC, as well as the challenges in developing more specific and effective STAT3 inhibitors.
Table 1
Summary of STAT3 inhibitors and their mechanisms of action for TNBC therapy
Inhibitors
Mechanisms of action
In vitro activity
In vivo activity
Reference
Strategy 1: Target upstream regulators of STAT3
 Carfilzomib
Inhibits IL-6/STAT3 signaling pathway
Inhibits mitosis and proliferation and induces apoptosis
Reduces serum IL-6 levels in tumor-bearing mice
[87]
 Manuka honey
Inhibits IL-6/STAT3 signaling pathway
Inhibits cell viability and colony formation, induces apoptosis, impairs cell migration and invasion, and inhibits angiogenesis
NR
[88]
 Bazedoxifene
Inhibits IL6/gp130/STAT3 signaling pathway
Inhibits cell viability, colony formation and cell migration and synergistically enhances the activity of paclitaxel
Suppresses tumor growth
[89, 90]
Ganoderma lucidum extract
Inhibits IL-6/JAK/STAT3 signaling pathway
Inhibits cell viability and induces apoptosis
Suppresses tumor growth
[91]
 Arsenic trioxide
Inhibits EZH2/NF-κB/IL-6/STAT3/VEGF signaling pathway
Inhibits angiogenesis
NR
[96]
 Deguelin
Inhibits EGFR/STAT3 signaling pathway
Inhibits cell viability
Suppresses tumor growth
[92]
 Picrasidine G
Inhibits EGFR/STAT3 signaling pathway
Inhibits cell viability and induces apoptosis
NR
[93]
 Cantharidin
Inhibits EGFR/STAT3 signaling pathway
Inhibits cell viability and induces apoptosis
NR
[94]
 Silibinin
Inhibits JAK2/STAT3/MMP2 signaling pathway
Inhibits cell viability, migration and invasion
NR
[97]
Inhibits EGFR/STAT3/Fibronectin signaling pathway
NR
NR
[95]
 Ganoderic acid A
Inhibits JAK2/STAT3 signaling pathway
Inhibits cell viability and invasive capacity and induces apoptosis
NR
[98]
 Nintedanib
Modulates SHP-1/p-STAT3 signaling pathway
Inhibits cell viability and induces apoptosis
Suppresses tumor growth
[99]
 SC-78
Modulates SHP-1/p-STAT3/VEGF-A signaling pathway
Inhibits cell migration and tube formation
Suppresses tumor growth and metastasis
[100]
 1,2,3,4,6-penta-O-galloyl-beta-D-glucose
Modulates SHP-1/p-STAT3 signaling pathway
NR
Suppresses tumor growth and metastasis
[101]
 SC-2001
Modulates RFX-1/SHP-1/p-STAT3 signaling pathway
Inhibits cell growth and induces apoptosis
Suppresses tumor growth
[95, 102]
 Isolinderalactone
Enhances SOCS3-mediated STAT3 dephosphorylation
Inhibits cell viability and colony formation and induces apoptosis
Suppresses tumor growth
[103]
 Compound 57
Binds to HSP90 and inhibits the expression and phosphorylation of STAT3
Inhibits cell viability
NR
[104]
 L80
Binds to HSP90 and inhibits the expression and phosphorylation of STAT3
Inhibits cell viability induces apoptosis, and suppresses BCSC-like properties
Suppresses the growth of BCSC-enriched TNBC tumors and distant metastasis
[105]
 Nor-wogonin
Inhibits TAK1-mediated STAT3 activation
Inhibits cell viability and proliferation and induces G1 and G2/M phases arrest and apoptosis
NR
[106]
 Thioridazine
Inhibits DRD2-mediated STAT3 activation
Inhibits cell self-renewal, proliferation, and viability and induces G1 arrest
NR
[107]
Strategy 2: Directly bind to STAT3 and inhibit its activation
 Bt354
Directly binds to SH2 domain of STAT3 and inhibits its phosphorylation
Inhibits cell viability, induces G2/M phase arrest and apoptosis, and impairs cell migration
Suppresses tumor growth
[108]
 Osthole
Directly binds to STAT3 and inhibits its phosphorylation
Inhibits cell viability and induces G2/M phase arrest and apoptosis
Suppresses tumor growth
[109]
 Arctigenin
Directly binds to SH2 domain of STAT3 and inhibits its phosphorylation and DNA binding ability
Inhibits cell viability, induces apoptosis, impairs cell migration and invasion, and sensitizes cells to chemotherapy
Suppresses tumor growth
[110]
 Alantolactone
Directly binds to SH2 domain of STAT3 and inhibits its phosphorylation
Inhibits cell viability and colony formation and impairs cell migration and invasion
Suppresses tumor growth
[111]
 KYZ3
Directly binds to SH2 domain of STAT3 and inhibits its phosphorylation
Inhibits cell viability, induces apoptosis, and impairs cell migration
Suppresses tumor growth
[113]
Strategy 3: Inhibit STAT3 phosphorylation or acetylation
 Sesquiterpene lactones fraction of Inula helenium L.
Inhibits STAT3 phosphorylation and nuclear translocation
Inhibits cell viability and induces apoptosis
Suppresses tumor growth
[114]
Rhus coriaria
Inhibits STAT3 phosphorylation
Inhibits angiogenesis and impairs cell migration and invasion
Suppresses tumor growth and metastasis
[115]
 Schisandrin B
Inhibits STAT3 phosphorylation and nuclear translocation
Inhibits cell viability and colony formation, induces cell cycle arrest and apoptosis, and impairs cell migration
Suppresses tumor growth
[116]
 Eupalinolide J
Inhibits STAT3 phosphorylation and activation
Inhibits cell viability
NR
[117]
 Galiellalactone analogues 16 and 17
Inhibits STAT3 phosphorylation and activation
Inhibits cell viability
NR
[118]
 FZU-03,010
Inhibits STAT3 phosphorylation and activation
Inhibits cell viability and induces G1 phase arrest and apoptosis
NR
[119]
 Niclosamide
Inhibits STAT3 phosphorylation and nuclear translocation
Reverses acquired radioresistance
Sensitizes tumors to irradiation
[120]
 Flubendazole
Inhibits STAT3 phosphorylation
Inhibits cell viability, induces G2/M phase arrest and apoptosis, and suppresses BCSC-like phenotype
Suppresses tumor growth, angiogenesis and metastasis
[121]
 Disulfiram
Inhibits STAT3 expression and phosphorylation
Inhibits cell viability, induces apoptosis, and impairs cancer stem cell-like properties
Suppresses tumor growth and BCSC-like properties
[122]
 Salinomycin
Inhibits STAT3 phosphorylation and activation
Inhibits cell viability, promotes anoikis, impairs cell migration and invasion, and decreases CD44+/CD24 stem-like population
NR
[123]
 Metformin
Inhibits STAT3 phosphorylation
Inhibits cell viability
NR
[124]
 SH-I-14
Inhibits STAT3 acetylation and disrupts DNMT1-STAT3 interaction
Inhibits cell viability
Suppresses tumor growth
[126]
Strategy 4: Block STAT3-DNA binding
 Methylsulfonyl-methane
Inhibits the bindings of STAT3 to VEGF promoter and STAT5 to IGF-1R promoter
Inhibits cell viability and induces apoptosis
Suppresses tumor growth
[127]
 Isoharringtonine
Inhibits STAT3-mediated Nanog expression
Inhibits cell viability, impairs cell migration, and decreases proportion of BCSC population
NR
[128]
 Salidroside
Inhibits the bindings of STAT3 to MMP2 promoter
Inhibits cell migration, invasion and angiogenesis
NR
[129]
NR, not reported

Target upstream regulators of STAT3

The majority of STAT3 inhibitors have been identified to target the upstream regulators of STAT3 signaling. STAT3 activation is often initiated through the binding of cytokines and growth factors to their corresponding cell surface receptors. Therefore, small molecules and natural products that are able to inhibit IL-6 secretion and production, e.g., carfilzomib [87], manuka honey [88], bazedoxifene [89, 90], and Ganoderma lucidum extract [91] or suppress EGFR expression and phosphorylation, e.g., deguelin [92], picrasidine G [93], cantharidin [94], and silibinin [95] have shown significant inhibitory effects on STAT3 signaling as well as the expression of its downstream target genes in TNBC cell lines. In addition, arsenic trioxide (ATO) was reported to inhibit IL-6-mediated STAT3 activation, consequently reducing the expression of VEGF and suppressing angiogenesis [96]. Further studies have demonstrated that ATO blocks the interaction between enhancer of zeste homolog 2 (EZH2) and NF-κB p65, herein suppressing the activity of NF-κB and reducing the expression of IL-6. All these indirect STAT3 inhibitors have exhibited potent in vitro and in vivo anti-TNBC activities (Table 1). However, most of them have also been found to inhibit other signaling pathways that are triggered by ligand-cell surface receptor binding in cancer cells, indicating a low level of specificity in targeting the STAT3 signaling pathway.
As discussed earlier, several protein tyrosine kinases, such as JAK2 contribute to STAT3 phosphorylation and activation in both receptor-dependent and/or receptor-independent manners. JAK2 inhibitors, including silibinin [97] and ganoderic acid A [98] were found to inhibit TNBC cell viability, migration, and invasion and induce apoptosis in vitro through inhibiting the JAK2/STAT3 signaling pathway. However, their in vivo efficacy still needs further investigation. Targeting the intrinsic STAT3 inhibitors, such as PTPs and SOCS have been considered as a potential strategy for repressing STAT3 signaling pathway. Several natural and synthetic compounds were identified to activate one of the STAT3 PTPs, SHP-1. Among them, nintedanib and SC-78 significantly increase SHP-1 activity without affecting its expression [99, 100], while 1,2,3,4,6-penta-O-galloyl-beta-D-glucose (PGG) and SC-2001 largely induce the expression of SHP-1 [101, 102]. All these SHP-1 activators were also shown to inhibit STAT3 phosphorylation and the expression of its downstream target genes, thus suppressing TNBC cell growth and migration and inducing apoptosis in vitro and in vivo [99102]. In addition, isolinderalactone was reported to increase SOCS3 expression and then enhance SOCS3-mediated STAT3 dephosphorylation and inactivation [103].
As one of the major client proteins of heat shock protein 90 (HSP90), STAT3 can be degraded through inhibiting HSP90. Two deguelin-derived HSP90 inhibitors, termed compound 57 and L80 have been observed to inhibit STAT3 expression and phosphorylation by interacting with the C-terminal ATP-binding pocket of HSP90 and blocking its function [104, 105]. Both compounds have also exerted their anticancer activities in TNBC models in vitro and in vivo [104, 105]. Moreover, nor-wogonin was found to inhibit the expression of transforming growth factor β-activated kinase 1 (TAK1), therefore dephosphorylating STAT3 without affecting its total expression level [106]. The dopamine receptor D2 (DRD2)-targeting drug thioridazine inhibits TNBC cell self-renewal through reducing DRD2-mediated STAT3 activation [107]. Due to the highly conserved structures among STAT family members, targeting the upstream regulators always results in the wide-spectrum inhibition of all STAT proteins, causing off-target effects. Therefore, directly targeting STAT3 and/or inhibiting its functions may be more promising strategies for developing safe and effective anticancer therapeutics.

Directly bind to STAT3 and inhibit its activation

Due to advances in the understanding of the structural biology of STAT3, small molecule inhibitors have been developed to directly bind to STAT3 and inhibit its activity. Currently, many small molecule inhibitors have been designed to target the SH2 domain and block its phosphorylation, dimerization, and nuclear translocation. Several STAT3-binding small molecule inhibitors that are under preclinical and clinical investigations have shown excellent efficacy in TNBC cells in vitro and in vivo.
Recently, a dual-luciferase assay-based screening of 1563 compounds for STAT3 inhibitors was performed, leading to the identification of Bt354 [108]. Further studies have shown that Bt354 inhibits STAT3 phosphorylation and nuclear translocation, which may be attributed to the binding of this compound to the SH2 domain of STAT3. Bt354 did not cause significant changes in the expression of STAT3 upstream regulators JAK2 and Src, indicating a specific targeting effect on STAT3 [108]. Moreover, this small molecule inhibitor also suppresses the viability of TNBC cells with constitutively activated STAT3, induces the G2/M phase arrest and late apoptosis, and impairs cell migration in vitro and represses the growth of TNBC xenograft tumors in vivo [108]. Additionally, several natural products, including osthole [109], arctigenin [110], and alantolactone [111] have also been shown to directly bind to the SH2 domain of STAT3, inhibit its phosphorylation and activation, and suppress the growth and metastasis of TNBC in vitro and in vivo. Cryptotanshinone is a well-documented natural product inhibitor of STAT3, which also binds to the SH2 domain and inhibits the phosphorylation and dimerization of STAT3 [112]. KYZ3, a synthetic derivative of cryptotanshinone has recently been developed and shown to exert anticancer activity in TNBC cells in vitro and in vivo through binding to and inhibiting STAT3 activation [113]. However, none of these compounds have been evaluated for their binding affinity to STAT3. Their selectivity among STAT3 and other STAT family members is yet to be determined.

Inhibit STAT3 phosphorylation or acetylation

Except for the STAT3-binding small molecule inhibitors that we discussed above, a number of natural products and their derivatives were found to inhibit STAT3 phosphorylation and/or nuclear translocation without affecting the upstream regulators. Sesquiterpene lactones, which are enriched in the hexane fraction from Inula helenium L. have been shown to suppress tumor growth in vitro and in vivo by inhibiting STAT3 phosphorylation and decreasing the expression of the downstream target genes, including cyclin D1, c-Myc, and Bcl-2 [114]. Another crude extract from the fruits of Rhus coriaria was also discovered to inhibit angiogenesis, tumor growth and metastasis in TNBC models in vitro and in vivo by repressing STAT3 phosphorylation and STAT3-mediated VEGF expression [115]. Moreover, several natural compounds and derivatives, including schisandrin B [116], eupalinolide J [117], galiellalactone analogs 16 and 17 [118], and ursolic acid derivative FZU-03,010 [119] have shown in vitro and in vivo efficacy in TNBC models through inhibition of STAT3 phosphorylation and/or nuclear translocation. None of them have been investigated for the binding ability with STAT3. Considering that these compounds did not show any significant effects on STAT3 regulators and interactive proteins, further studies for examining the potential binding between STAT3 and these compounds would provide important information regarding their underlying molecular mechanisms.
Of note, several approved drugs have shown potent inhibitory effects on pSTAT3 and may be repositioned as anticancer drugs. Niclosamide, an FDA-approved anthelmintic drug was identified as a potent STAT3 inhibitor. A recent study demonstrated that niclosamide not only inhibits TNBC cell viability but also sensitizes TNBC cells to ionizing irradiation (IR) by blocking IR-induced STAT3 phosphorylation and activation [120]. Flubendazole, another wildly used anthelmintic agent and disulfiram, a clinical drug for treating chronic alcoholism were found to eradicate TNBC stem cells-like cells that express high levels of pSTAT3 [121, 122]. Further studies showed that both drugs were able to cause TNBC cell growth arrest and apoptosis in vitro and suppress TNBC tumor growth, angiogenesis, and metastasis in vivo by inhibiting STAT3 [121, 122]. Moreover, salinomycin, an antibacterial and coccidiostat ionophore therapeutic drug and metformin, an antidiabetic drug have exhibited potent inhibitory effects on STAT3 phosphorylation and TNBC cell growth in vitro [123, 124]. However, further evaluation of their anti-TNBC efficacy in in vivo models is critically needed.
Recent studies have disclosed that targeting STAT3 acetylation may be a potential therapeutic approach for treating cancer. SH-I-14, a newly synthesized carbazole was shown to inhibit STAT3 phosphorylation through increasing SHP-1 expression [125]. A follow-up study reported that SH-I-14 also inhibited STAT3 acetylation and disrupted DNMT1-STAT3 interaction, resulting in DNA demethylation and re-expression of tumor suppressor genes [126]. Its in vitro and in vivo activity has also been demonstrated in TNBC model, suggesting the effectiveness of inhibiting STAT3 acetylation in TNBC therapy.

Block STAT3-DNA binding

STAT3 induces the expression of its downstream targets through binding to DNA and activating the transcription. Therefore, inhibition of STAT3-DNA binding has been considered as a promising strategy to develop targeted cancer therapies. Several STAT3-DNA binding inhibitors have been developed and shown potent anticancer efficacy in TNBC cells. Methylsulfonyl-methane (MSM), a dietary supplement was found to inhibit TNBC cell viability and induce apoptosis by blocking the DNA binding abilities of STAT3 to VEGF promoter and STAT5 to IGF-1R (IGF-1 receptor) promoter and repressing the expression of VEGF and IGF-1R [127]. Considering the extremely low toxicity of MSM, it could be developed as a preventive agent for cancers harboring overexpressed and aberrantly activated STAT3. Two natural compounds, isoharringtonine and salidroside have also been demonstrated to exert their anti-TNBC activities by blocking the binding of STAT3 to Nanog and MMP2 promoters, respectively [128, 129]. However, their binding affinity to STAT3 and in vivo efficacy are yet to be studied.
As discussed above, several strategies (as shown in Fig. 3) have been developed to inhibit STAT3 signaling, i.e. 1) targeting the upstream regulators, 2) directly binding to STAT3 SH2 domain and inhibiting its activation, 3) inhibiting STAT3 phosphorylation or acetylation, and 4) blocking STAT3-DNA binding. Many small molecules have been developed and shown efficacy in preventing and treating TNBC in preclinical studies (Table 1). Several STAT3 inhibitors also enter clinical trials [130148], which have been summarized in Table 2. There are other STAT3-targeting strategies (as shown in Fig. 3) that have not been examined, including 1) inhibiting the binding of STAT3 with its co-activators (e.g., NCOA/SRC1a, APE/Ref-1, and CBP/p300) and repressing its transcriptional activity, 2) modulating the binding of STAT3 with other interactive proteins (e.g., SMYD2 and TRAF6) that regulate its activity and stability, and 3) developing STAT3-targeting PROTACs (proteolysis targeting chimeras) for promoting STAT3 ubiquitination and proteasomal degradation. Because most of the small molecule STAT3 inhibitors have been developed to inhibit its phosphorylation and activation but not affect the protein stability, long-term treatment of these inhibitors may result in the compensatory activation of other signaling pathways, finally causing drug resistance. Therefore, small molecules, such as PROTACs that can induce STAT3 protein degradation may be used more efficiently in combination with current inhibitors for cancer therapy.
Table 2
Summary of STAT3 inhibitors in clinical trials
Inhibitors
Target
ClinicalTrials ID
Condition or disease
Phase
References
STAT3 DECOY
STAT3
NCT00696176
Head and neck cancer
Early phase 1
[130]
AZD9150
(IONIS-STAT3Rx or ISIS-STAT3Rx)
STAT3
NCT01563302
Advanced cancers, DLBCL
Phases 1 & 2
[131, 132]
NCT02417753
Ovarian cancer, GIC
Phase 2
NCT01839604
HCC
Phase 1
NCT02983578
GIC, lung cancer, etc.
Phase 2
NCT03527147
NHL, DLBCL, NHL, DLBCL
Phase 1
NCT02549651
DLBCL
Phase 1
NCT03421353
Advanced solid tumors
Phases 1 & 2
TTI-101
(C188–9)
STAT3
NCT03195699
Breast cancer, HNSCC, NSCLC, etc.
Phase 1
[133]
OPB-51602
STAT3
NCT02058017
Nasopharyngeal carcinoma
Phase 1
[134]
NCT01867073
Advanced solid tumors
Phase 1
NCT01423903
Advanced cancer
Phase 1
OPB-31121
STAT3
NCT00955812
Advanced cancer, solid tumor
Phase 1
[136]
OPB-111077
STAT3
NCT01711034
Solid tumors
Phase 1
[137]
Napabucasin (BBI608 or GB201)
STAT3
NCT03647839
MCC
Phase 2
[135]
NCT03522649
Previously treated MCC
Phase 3
NCT02826161
NSCLC
Phase 3
NCT02993731
Pancreatic ductal carcinoma
Phase 3
Pyrimethamine
STAT3
NCT01066663
CLL, SLL
Phases 1 & 2
[138]
NCT03057990
Myelodysplastic syndromes
Phase 1
Simvastatin
STAT3
NCT02390843
Retinoblastoma, clear cell sarcoma, renal cell carcinoma, rhabdoid tumor, etc.
Phase 1
[139]
DSP-0337
STAT3
NCT03416816
Neoplasms
Phase 1
[140]
Cetuximab
EGFR
NCT01445405
Squamous carcinoma, head and neck cancer, etc.
Phase 1
[141]
Lapatinib
EGFR
NCT00105950
Breast neoplasms
Phase 2
[142]
Dasatinib
c-Src
NCT02680951
AML
Phase 1
[143]
SC-43
SHP-1
NCT03443622
Refractory solid tumor
Phase 1
[144]
ASN002
JAK
NCT02440685
Lymphoma, leukemia
Phases 1 & 2
[145]
SAR302503
JAK2
NCT01420783
Hematopoietic neoplasm
Phase 2
[146]
AZD1480
JAK2
NCT01112397
Solid malignancies
Phase 1
[147]
WP1066
JAK2
NCT01904123
Metastatic melanoma, recurrent glioblastoma, etc.
Phase 1
[148]
AML Acute myeloid leukemia, CLL Chronic lymphocytic leukemia, DLBCL Diffuse large B-cell lymphoma, GIC Gastrointestinal cancer, HCC Hepatocellular carcinoma, HNSCC Head and neck squamous cell carcinoma, MCC Metastatic colorectal cancer, NHL Non-Hodgkin lymphoma, NSCLC Non-small cell lung cancer, SLL Small lymphocytic leukemia

Conclusions

TNBC is still a treatable but incurable disease with complex genetic heterogeneity. The STAT3 oncogene is overexpressed and constitutively activated in TNBC and is associated with the high metastatic risk and poor survival outcomes. Moreover, STAT3 not only acts as a transcription factor to activate the expression of its downstream target genes but also localizes to mitochondria and regulates its functions, then regulating the various aspects of TNBC cells. Many STAT3-targeted therapies have been successfully developed and shown efficacy in preclinical models of TNBC in vitro and in vivo; several STAT3 inhibitors even enter clinical trials and are currently under investigation in various human cancers, including TNBC. In addition to its role in cancer cells, STAT3 also plays a pivotal role in the immune system. Indeed, STAT3 inhibitors have been found to suppress tumor cells but also boost immune cell responses. Therefore, the STAT3 oncogene is a promising target for TNBC prevention and therapy.
Of note, targeting STAT3 alone has shown excellent anti-TNBC activities in preclinical settings. However, TNBC has been reported to harbor multiple genetic alterations, including STAT3 overexpression and constitutive activation which contribute to the initiation, progression, metastasis, and drug resistance of this disease. Therefore, STAT3 inhibition combined with other targeted therapies may be more effective in treating TNBC. Considering that STAT3 plays a crucial role in chemoresistance, the combination of STAT3 inhibitors with other chemotherapies may exert synergistic effects in treating TNBC. Therefore, further studies are warranted to demonstrate the preventive and therapeutic efficacy of STAT3 inhibitors alone or in combination with chemotherapy and/or other targeted therapies in clinical studies. Moreover, new targeting strategies, i.e. inducing the degradation of STAT3 protein through PROTAC or inhibiting the binding of STAT3 to its co-activators and other interactive proteins can be examined, which may lead to more specific and effective inhibitors for TNBC prevention and therapy.

Acknowledgements

We thank the current and former members of our laboratories and collaborators for their contributions to the publications cited in this review article. The research field in STAT3 is rapidly growing, and we apologize for not being able to cite all the recent publications, due to space limitation.

Funding

JJQ was supported by Zhejiang Chinese Medical University Startup Funding (111100E014). WDZ was supported by Professor of Chang Jiang Scholars Program, NSFC (81520108030, 21472238), Shanghai Engineering Research Center for the Preparation of Bioactive Natural Products (16DZ2280200), the Scientific Foundation of Shanghai China (13401900103, 13401900101), and the National Key Research and Development Program of China (2017YFC1700200).

Availability of data and materials

Not applicable.
Not applicable.
Not applicable.

Competing interests

The authors declare that they have no competing interests.

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Open AccessThis article is distributed under the terms of the Creative Commons Attribution 4.0 International License (http://​creativecommons.​org/​licenses/​by/​4.​0/​), which permits unrestricted use, distribution, and reproduction in any medium, provided you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons license, and indicate if changes were made. The Creative Commons Public Domain Dedication waiver (http://​creativecommons.​org/​publicdomain/​zero/​1.​0/​) applies to the data made available in this article, unless otherwise stated.
Literatur
2.
Zurück zum Zitat Sharma P. Update on the treatment of early-stage triple-negative breast Cancer. Curr Treat Options in Oncol. 2018;19(5):22.CrossRef Sharma P. Update on the treatment of early-stage triple-negative breast Cancer. Curr Treat Options in Oncol. 2018;19(5):22.CrossRef
3.
Zurück zum Zitat Walsh EM, Keane MM, Wink DA, Callagy G, Glynn SA. Review of triple negative breast Cancer and the impact of inducible nitric oxide synthase on tumor biology and patient outcomes. Crit Rev Oncog. 2016;21(5–6):333–51.PubMedPubMedCentralCrossRef Walsh EM, Keane MM, Wink DA, Callagy G, Glynn SA. Review of triple negative breast Cancer and the impact of inducible nitric oxide synthase on tumor biology and patient outcomes. Crit Rev Oncog. 2016;21(5–6):333–51.PubMedPubMedCentralCrossRef
4.
Zurück zum Zitat Kim C, Gao R, Sei E, Brandt R, Hartman J, Hatschek T, et al. Chemoresistance evolution in triple-negative breast Cancer delineated by single-cell sequencing. Cell. 2018;173(4):879–93 e813.PubMedPubMedCentralCrossRef Kim C, Gao R, Sei E, Brandt R, Hartman J, Hatschek T, et al. Chemoresistance evolution in triple-negative breast Cancer delineated by single-cell sequencing. Cell. 2018;173(4):879–93 e813.PubMedPubMedCentralCrossRef
5.
Zurück zum Zitat Robert M, Patsouris A, Frenel JS, Gourmelon C, Augereau P, Campone M. Emerging PARP inhibitors for treating breast cancer. Expert Opin Emerg Drugs. 2018;23(3):211–21.PubMedCrossRef Robert M, Patsouris A, Frenel JS, Gourmelon C, Augereau P, Campone M. Emerging PARP inhibitors for treating breast cancer. Expert Opin Emerg Drugs. 2018;23(3):211–21.PubMedCrossRef
6.
Zurück zum Zitat Costa R, Shah AN, Santa-Maria CA, Cruz MR, Mahalingam D, Carneiro BA, et al. Targeting epidermal growth factor receptor in triple negative breast cancer: new discoveries and practical insights for drug development. Cancer Treat Rev. 2017;53:111–9.PubMedCrossRef Costa R, Shah AN, Santa-Maria CA, Cruz MR, Mahalingam D, Carneiro BA, et al. Targeting epidermal growth factor receptor in triple negative breast cancer: new discoveries and practical insights for drug development. Cancer Treat Rev. 2017;53:111–9.PubMedCrossRef
7.
8.
Zurück zum Zitat Sirkisoon SR, Carpenter RL, Rimkus T, Anderson A, Harrison A, Lange AM, et al. Interaction between STAT3 and GLI1/tGLI1 oncogenic transcription factors promotes the aggressiveness of triple-negative breast cancers and HER2-enriched breast cancer. Oncogene. 2018;37(19):2502–14.PubMedPubMedCentralCrossRef Sirkisoon SR, Carpenter RL, Rimkus T, Anderson A, Harrison A, Lange AM, et al. Interaction between STAT3 and GLI1/tGLI1 oncogenic transcription factors promotes the aggressiveness of triple-negative breast cancers and HER2-enriched breast cancer. Oncogene. 2018;37(19):2502–14.PubMedPubMedCentralCrossRef
9.
Zurück zum Zitat Gupta I, Sareyeldin RM, Al-Hashimi I, Al-Thawadi HA, Al Farsi H, Vranic S, et al. Triple Negative Breast Cancer Profile, from Gene to microRNA, in Relation to Ethnicity. Cancers (Basel). 2019;11(3):363.PubMedCentralCrossRef Gupta I, Sareyeldin RM, Al-Hashimi I, Al-Thawadi HA, Al Farsi H, Vranic S, et al. Triple Negative Breast Cancer Profile, from Gene to microRNA, in Relation to Ethnicity. Cancers (Basel). 2019;11(3):363.PubMedCentralCrossRef
10.
11.
Zurück zum Zitat Furtek SL, Backos DS, Matheson CJ, Reigan P. Strategies and approaches of targeting STAT3 for Cancer treatment. ACS Chem Biol. 2016;11(2):308–18.PubMedCrossRef Furtek SL, Backos DS, Matheson CJ, Reigan P. Strategies and approaches of targeting STAT3 for Cancer treatment. ACS Chem Biol. 2016;11(2):308–18.PubMedCrossRef
13.
Zurück zum Zitat Akira S, Nishio Y, Inoue M, Wang XJ, Wei S, Matsusaka T, et al. Molecular cloning of APRF, a novel IFN-stimulated gene factor 3 p91-related transcription factor involved in the gp130-mediated signaling pathway. Cell. 1994;77(1):63–71.PubMedCrossRef Akira S, Nishio Y, Inoue M, Wang XJ, Wei S, Matsusaka T, et al. Molecular cloning of APRF, a novel IFN-stimulated gene factor 3 p91-related transcription factor involved in the gp130-mediated signaling pathway. Cell. 1994;77(1):63–71.PubMedCrossRef
14.
Zurück zum Zitat Zhong Z, Wen Z, Darnell JE Jr. Stat3: a STAT family member activated by tyrosine phosphorylation in response to epidermal growth factor and interleukin-6. Science. 1994;264(5155):95–8.PubMedCrossRef Zhong Z, Wen Z, Darnell JE Jr. Stat3: a STAT family member activated by tyrosine phosphorylation in response to epidermal growth factor and interleukin-6. Science. 1994;264(5155):95–8.PubMedCrossRef
15.
Zurück zum Zitat Huynh J, Chand A, Gough D, Ernst M. Therapeutically exploiting STAT3 activity in cancer - using tissue repair as a road map. Nat Rev Cancer. 2019;19(2):82–96.PubMedCrossRef Huynh J, Chand A, Gough D, Ernst M. Therapeutically exploiting STAT3 activity in cancer - using tissue repair as a road map. Nat Rev Cancer. 2019;19(2):82–96.PubMedCrossRef
17.
Zurück zum Zitat Yu H, Lee H, Herrmann A, Buettner R, Jove R. Revisiting STAT3 signalling in cancer: new and unexpected biological functions. Nat Rev Cancer. 2014;14(11):736–46.CrossRefPubMed Yu H, Lee H, Herrmann A, Buettner R, Jove R. Revisiting STAT3 signalling in cancer: new and unexpected biological functions. Nat Rev Cancer. 2014;14(11):736–46.CrossRefPubMed
18.
Zurück zum Zitat Guanizo AC, Fernando CD, Garama DJ, Gough DJ. STAT3: a multifaceted oncoprotein. Growth Factors. 2018;36(1–2):1–14.PubMedCrossRef Guanizo AC, Fernando CD, Garama DJ, Gough DJ. STAT3: a multifaceted oncoprotein. Growth Factors. 2018;36(1–2):1–14.PubMedCrossRef
19.
Zurück zum Zitat Garbers C, Aparicio-Siegmund S, Rose-John S. The IL-6/gp130/STAT3 signaling axis: recent advances towards specific inhibition. Curr Opin Immunol. 2015;34:75–82.PubMedCrossRef Garbers C, Aparicio-Siegmund S, Rose-John S. The IL-6/gp130/STAT3 signaling axis: recent advances towards specific inhibition. Curr Opin Immunol. 2015;34:75–82.PubMedCrossRef
20.
Zurück zum Zitat Karras JG, Wang Z, Huo L, Howard RG, Frank DA, Rothstein TL. Signal transducer and activator of transcription-3 (STAT3) is constitutively activated in normal, self-renewing B-1 cells but only inducibly expressed in conventional B lymphocytes. J Exp Med. 1997;185(6):1035–42.PubMedPubMedCentralCrossRef Karras JG, Wang Z, Huo L, Howard RG, Frank DA, Rothstein TL. Signal transducer and activator of transcription-3 (STAT3) is constitutively activated in normal, self-renewing B-1 cells but only inducibly expressed in conventional B lymphocytes. J Exp Med. 1997;185(6):1035–42.PubMedPubMedCentralCrossRef
21.
Zurück zum Zitat Wang Y, Shen Y, Wang S, Shen Q, Zhou X. The role of STAT3 in leading the crosstalk between human cancers and the immune system. Cancer Lett. 2018;415:117–28.PubMedCrossRef Wang Y, Shen Y, Wang S, Shen Q, Zhou X. The role of STAT3 in leading the crosstalk between human cancers and the immune system. Cancer Lett. 2018;415:117–28.PubMedCrossRef
22.
Zurück zum Zitat Yu H, Kortylewski M, Pardoll D. Crosstalk between cancer and immune cells: role of STAT3 in the tumour microenvironment. Nat Rev Immunol. 2007;7(1):41–51.PubMedCrossRef Yu H, Kortylewski M, Pardoll D. Crosstalk between cancer and immune cells: role of STAT3 in the tumour microenvironment. Nat Rev Immunol. 2007;7(1):41–51.PubMedCrossRef
23.
Zurück zum Zitat Huang YH, Molavi O, Alshareef A, Haque M, Wang Q, Chu MP, et al. Constitutive activation of STAT3 in myeloma cells cultured in a three-dimensional, reconstructed bone marrow model. Cancers (Basel). 2018;10(6):206.PubMedCentralCrossRef Huang YH, Molavi O, Alshareef A, Haque M, Wang Q, Chu MP, et al. Constitutive activation of STAT3 in myeloma cells cultured in a three-dimensional, reconstructed bone marrow model. Cancers (Basel). 2018;10(6):206.PubMedCentralCrossRef
24.
Zurück zum Zitat Quarta S, Baeumer BE, Scherbakov N, Andratsch M, Rose-John S, Dechant G, et al. Peripheral nerve regeneration and NGF-dependent neurite outgrowth of adult sensory neurons converge on STAT3 phosphorylation downstream of neuropoietic cytokine receptor gp130. J Neurosci. 2014;34(39):13222–33.PubMedPubMedCentralCrossRef Quarta S, Baeumer BE, Scherbakov N, Andratsch M, Rose-John S, Dechant G, et al. Peripheral nerve regeneration and NGF-dependent neurite outgrowth of adult sensory neurons converge on STAT3 phosphorylation downstream of neuropoietic cytokine receptor gp130. J Neurosci. 2014;34(39):13222–33.PubMedPubMedCentralCrossRef
25.
Zurück zum Zitat Hruz P, Dann SM, Eckmann L. STAT3 and its activators in intestinal defense and mucosal homeostasis. Curr Opin Gastroenterol. 2010;26(2):109–15.PubMedCrossRef Hruz P, Dann SM, Eckmann L. STAT3 and its activators in intestinal defense and mucosal homeostasis. Curr Opin Gastroenterol. 2010;26(2):109–15.PubMedCrossRef
26.
27.
Zurück zum Zitat Bousquet C, Susini C, Melmed S. Inhibitory roles for SHP-1 and SOCS-3 following pituitary proopiomelanocortin induction by leukemia inhibitory factor. J Clin Invest. 1999;104(9):1277–85.PubMedPubMedCentralCrossRef Bousquet C, Susini C, Melmed S. Inhibitory roles for SHP-1 and SOCS-3 following pituitary proopiomelanocortin induction by leukemia inhibitory factor. J Clin Invest. 1999;104(9):1277–85.PubMedPubMedCentralCrossRef
28.
Zurück zum Zitat Kim H, Baumann H. Dual signaling role of the protein tyrosine phosphatase SHP-2 in regulating expression of acute-phase plasma proteins by interleukin-6 cytokine receptors in hepatic cells. Mol Cell Biol. 1999;19(8):5326–38.PubMedPubMedCentralCrossRef Kim H, Baumann H. Dual signaling role of the protein tyrosine phosphatase SHP-2 in regulating expression of acute-phase plasma proteins by interleukin-6 cytokine receptors in hepatic cells. Mol Cell Biol. 1999;19(8):5326–38.PubMedPubMedCentralCrossRef
29.
Zurück zum Zitat Kim DJ, Tremblay ML, Digiovanni J. Protein tyrosine phosphatases, TC-PTP, SHP1, and SHP2, cooperate in rapid dephosphorylation of Stat3 in keratinocytes following UVB irradiation. PLoS One. 2010;5(4):e10290.PubMedPubMedCentralCrossRef Kim DJ, Tremblay ML, Digiovanni J. Protein tyrosine phosphatases, TC-PTP, SHP1, and SHP2, cooperate in rapid dephosphorylation of Stat3 in keratinocytes following UVB irradiation. PLoS One. 2010;5(4):e10290.PubMedPubMedCentralCrossRef
30.
Zurück zum Zitat Wakahara R, Kunimoto H, Tanino K, Kojima H, Inoue A, Shintaku H, et al. Phospho-Ser727 of STAT3 regulates STAT3 activity by enhancing dephosphorylation of phospho-Tyr705 largely through TC45. Genes Cells. 2012;17(2):132–45.PubMedCrossRef Wakahara R, Kunimoto H, Tanino K, Kojima H, Inoue A, Shintaku H, et al. Phospho-Ser727 of STAT3 regulates STAT3 activity by enhancing dephosphorylation of phospho-Tyr705 largely through TC45. Genes Cells. 2012;17(2):132–45.PubMedCrossRef
31.
Zurück zum Zitat Martin-Granados C, Prescott AR, Le Sommer S, Klaska IP, Yu T, Muckersie E, et al. A key role for PTP1B in dendritic cell maturation, migration, and T cell activation. J Mol Cell Biol. 2015;7(6):517–28.PubMedCrossRef Martin-Granados C, Prescott AR, Le Sommer S, Klaska IP, Yu T, Muckersie E, et al. A key role for PTP1B in dendritic cell maturation, migration, and T cell activation. J Mol Cell Biol. 2015;7(6):517–28.PubMedCrossRef
32.
Zurück zum Zitat Naka T, Narazaki M, Hirata M, Matsumoto T, Minamoto S, Aono A, et al. Structure and function of a new STAT-induced STAT inhibitor. Nature. 1997;387(6636):924–9.PubMedCrossRef Naka T, Narazaki M, Hirata M, Matsumoto T, Minamoto S, Aono A, et al. Structure and function of a new STAT-induced STAT inhibitor. Nature. 1997;387(6636):924–9.PubMedCrossRef
33.
Zurück zum Zitat Yagil Z, Nechushtan H, Kay G, Yang CM, Kemeny DM, Razin E. The enigma of the role of protein inhibitor of activated STAT3 (PIAS3) in the immune response. Trends Immunol. 2010;31(5):199–204.PubMedCrossRef Yagil Z, Nechushtan H, Kay G, Yang CM, Kemeny DM, Razin E. The enigma of the role of protein inhibitor of activated STAT3 (PIAS3) in the immune response. Trends Immunol. 2010;31(5):199–204.PubMedCrossRef
34.
Zurück zum Zitat Wei J, Yuan Y, Jin C, Chen H, Leng L, He F, et al. The ubiquitin ligase TRAF6 negatively regulates the JAK-STAT signaling pathway by binding to STAT3 and mediating its ubiquitination. PLoS One. 2012;7(11):e49567.PubMedPubMedCentralCrossRef Wei J, Yuan Y, Jin C, Chen H, Leng L, He F, et al. The ubiquitin ligase TRAF6 negatively regulates the JAK-STAT signaling pathway by binding to STAT3 and mediating its ubiquitination. PLoS One. 2012;7(11):e49567.PubMedPubMedCentralCrossRef
35.
Zurück zum Zitat Zhu Z, Wang S, Zhu J, Yang Q, Dong H, Huang J. MicroRNA-544 down-regulates both Bcl6 and Stat3 to inhibit tumor growth of human triple negative breast cancer. Biol Chem. 2016;397(10):1087–95.PubMedCrossRef Zhu Z, Wang S, Zhu J, Yang Q, Dong H, Huang J. MicroRNA-544 down-regulates both Bcl6 and Stat3 to inhibit tumor growth of human triple negative breast cancer. Biol Chem. 2016;397(10):1087–95.PubMedCrossRef
36.
Zurück zum Zitat Zhang W, Qu X, Chen B, Snyder M, Wang M, Li B, et al. Critical roles of STAT3 in beta-adrenergic functions in the heart. Circulation. 2016;133(1):48–61.PubMedCrossRef Zhang W, Qu X, Chen B, Snyder M, Wang M, Li B, et al. Critical roles of STAT3 in beta-adrenergic functions in the heart. Circulation. 2016;133(1):48–61.PubMedCrossRef
37.
Zurück zum Zitat Zhou T, Chao L, Rong G, Wang C, Ma R, Wang X. Down-regulation of GRIM-19 is associated with STAT3 overexpression in breast carcinomas. Hum Pathol. 2013;44(9):1773–9.PubMedCrossRef Zhou T, Chao L, Rong G, Wang C, Ma R, Wang X. Down-regulation of GRIM-19 is associated with STAT3 overexpression in breast carcinomas. Hum Pathol. 2013;44(9):1773–9.PubMedCrossRef
38.
Zurück zum Zitat Shields BJ, Wiede F, Gurzov EN, Wee K, Hauser C, Zhu HJ, et al. TCPTP regulates SFK and STAT3 signaling and is lost in triple-negative breast cancers. Mol Cell Biol. 2013;33(3):557–70.PubMedPubMedCentralCrossRef Shields BJ, Wiede F, Gurzov EN, Wee K, Hauser C, Zhu HJ, et al. TCPTP regulates SFK and STAT3 signaling and is lost in triple-negative breast cancers. Mol Cell Biol. 2013;33(3):557–70.PubMedPubMedCentralCrossRef
40.
Zurück zum Zitat Wegrzyn J, Potla R, Chwae YJ, Sepuri NB, Zhang Q, Koeck T, et al. Function of mitochondrial Stat3 in cellular respiration. Science. 2009;323(5915):793–7.PubMedPubMedCentralCrossRef Wegrzyn J, Potla R, Chwae YJ, Sepuri NB, Zhang Q, Koeck T, et al. Function of mitochondrial Stat3 in cellular respiration. Science. 2009;323(5915):793–7.PubMedPubMedCentralCrossRef
41.
Zurück zum Zitat Zhang Q, Raje V, Yakovlev VA, Yacoub A, Szczepanek K, Meier J, et al. Mitochondrial localized Stat3 promotes breast cancer growth via phosphorylation of serine 727. J Biol Chem. 2013;288(43):31280–8.PubMedPubMedCentralCrossRef Zhang Q, Raje V, Yakovlev VA, Yacoub A, Szczepanek K, Meier J, et al. Mitochondrial localized Stat3 promotes breast cancer growth via phosphorylation of serine 727. J Biol Chem. 2013;288(43):31280–8.PubMedPubMedCentralCrossRef
42.
Zurück zum Zitat Lee H, Zhang P, Herrmann A, Yang C, Xin H, Wang Z, et al. Acetylated STAT3 is crucial for methylation of tumor-suppressor gene promoters and inhibition by resveratrol results in demethylation. Proc Natl Acad Sci U S A. 2012;109(20):7765–9.PubMedPubMedCentralCrossRef Lee H, Zhang P, Herrmann A, Yang C, Xin H, Wang Z, et al. Acetylated STAT3 is crucial for methylation of tumor-suppressor gene promoters and inhibition by resveratrol results in demethylation. Proc Natl Acad Sci U S A. 2012;109(20):7765–9.PubMedPubMedCentralCrossRef
43.
Zurück zum Zitat Diaz N, Minton S, Cox C, Bowman T, Gritsko T, Garcia R, et al. Activation of stat3 in primary tumors from high-risk breast cancer patients is associated with elevated levels of activated SRC and survivin expression. Clin Cancer Res. 2006;12(1):20–8.PubMedCrossRef Diaz N, Minton S, Cox C, Bowman T, Gritsko T, Garcia R, et al. Activation of stat3 in primary tumors from high-risk breast cancer patients is associated with elevated levels of activated SRC and survivin expression. Clin Cancer Res. 2006;12(1):20–8.PubMedCrossRef
44.
Zurück zum Zitat Gritsko T, Williams A, Turkson J, Kaneko S, Bowman T, Huang M, et al. Persistent activation of stat3 signaling induces survivin gene expression and confers resistance to apoptosis in human breast cancer cells. Clin Cancer Res. 2006;12(1):11–9.PubMedCrossRef Gritsko T, Williams A, Turkson J, Kaneko S, Bowman T, Huang M, et al. Persistent activation of stat3 signaling induces survivin gene expression and confers resistance to apoptosis in human breast cancer cells. Clin Cancer Res. 2006;12(1):11–9.PubMedCrossRef
45.
Zurück zum Zitat Cheng Y, Holloway MP, Nguyen K, McCauley D, Landesman Y, Kauffman MG, et al. XPO1 (CRM1) inhibition represses STAT3 activation to drive a survivin-dependent oncogenic switch in triple-negative breast cancer. Mol Cancer Ther. 2014;13(3):675–86.PubMedPubMedCentralCrossRef Cheng Y, Holloway MP, Nguyen K, McCauley D, Landesman Y, Kauffman MG, et al. XPO1 (CRM1) inhibition represses STAT3 activation to drive a survivin-dependent oncogenic switch in triple-negative breast cancer. Mol Cancer Ther. 2014;13(3):675–86.PubMedPubMedCentralCrossRef
46.
Zurück zum Zitat Nam K, Son SH, Oh S, Jeon D, Kim H, Noh DY, et al. Binding of galectin-1 to integrin beta1 potentiates drug resistance by promoting survivin expression in breast cancer cells. Oncotarget. 2017;8(22):35804–23.PubMedPubMedCentralCrossRef Nam K, Son SH, Oh S, Jeon D, Kim H, Noh DY, et al. Binding of galectin-1 to integrin beta1 potentiates drug resistance by promoting survivin expression in breast cancer cells. Oncotarget. 2017;8(22):35804–23.PubMedPubMedCentralCrossRef
47.
Zurück zum Zitat Chang R, Song L, Xu Y, Wu Y, Dai C, Wang X, et al. Loss of Wwox drives metastasis in triple-negative breast cancer by JAK2/STAT3 axis. Nat Commun. 2018;9(1):3486.PubMedPubMedCentralCrossRef Chang R, Song L, Xu Y, Wu Y, Dai C, Wang X, et al. Loss of Wwox drives metastasis in triple-negative breast cancer by JAK2/STAT3 axis. Nat Commun. 2018;9(1):3486.PubMedPubMedCentralCrossRef
48.
Zurück zum Zitat Liu J, Liu L, Yague E, Yang Q, Pan T, Zhao H, et al. GGNBP2 suppresses triple-negative breast cancer aggressiveness through inhibition of IL-6/STAT3 signaling activation. Breast Cancer Res Treat. 2019;174(1):65–78.PubMedCrossRef Liu J, Liu L, Yague E, Yang Q, Pan T, Zhao H, et al. GGNBP2 suppresses triple-negative breast cancer aggressiveness through inhibition of IL-6/STAT3 signaling activation. Breast Cancer Res Treat. 2019;174(1):65–78.PubMedCrossRef
49.
Zurück zum Zitat Li LX, Zhou JX, Calvet JP, Godwin AK, Jensen RA, Li X. Lysine methyltransferase SMYD2 promotes triple negative breast cancer progression. Cell Death Dis. 2018;9(3):326.PubMedPubMedCentralCrossRef Li LX, Zhou JX, Calvet JP, Godwin AK, Jensen RA, Li X. Lysine methyltransferase SMYD2 promotes triple negative breast cancer progression. Cell Death Dis. 2018;9(3):326.PubMedPubMedCentralCrossRef
50.
Zurück zum Zitat Lee H, Herrmann A, Deng JH, Kujawski M, Niu G, Li Z, et al. Persistently activated Stat3 maintains constitutive NF-kappaB activity in tumors. Cancer Cell. 2009;15(4):283–93.PubMedPubMedCentralCrossRef Lee H, Herrmann A, Deng JH, Kujawski M, Niu G, Li Z, et al. Persistently activated Stat3 maintains constitutive NF-kappaB activity in tumors. Cancer Cell. 2009;15(4):283–93.PubMedPubMedCentralCrossRef
51.
Zurück zum Zitat Banerjee K, Pru C, Pru JK, Resat H. STAT3 knockdown induces tumor formation by MDA-MB-231 cells. Clin Oncol Res. 2018;1(1). Banerjee K, Pru C, Pru JK, Resat H. STAT3 knockdown induces tumor formation by MDA-MB-231 cells. Clin Oncol Res. 2018;1(1).
53.
Zurück zum Zitat Wang S, Yao Y, Yao M, Fu P, Wang W. Interleukin-22 promotes triple negative breast cancer cells migration and paclitaxel resistance through JAK-STAT3/MAPKs/AKT signaling pathways. Biochem Biophys Res Commun. 2018;503(3):1605–9.PubMedCrossRef Wang S, Yao Y, Yao M, Fu P, Wang W. Interleukin-22 promotes triple negative breast cancer cells migration and paclitaxel resistance through JAK-STAT3/MAPKs/AKT signaling pathways. Biochem Biophys Res Commun. 2018;503(3):1605–9.PubMedCrossRef
54.
Zurück zum Zitat Liang S, Chen Z, Jiang G, Zhou Y, Liu Q, Su Q, et al. Activation of GPER suppresses migration and angiogenesis of triple negative breast cancer via inhibition of NF-kappaB/IL-6 signals. Cancer Lett. 2017;386:12–23.PubMedCrossRef Liang S, Chen Z, Jiang G, Zhou Y, Liu Q, Su Q, et al. Activation of GPER suppresses migration and angiogenesis of triple negative breast cancer via inhibition of NF-kappaB/IL-6 signals. Cancer Lett. 2017;386:12–23.PubMedCrossRef
55.
Zurück zum Zitat Rigiracciolo DC, Santolla MF, Lappano R, Vivacqua A, Cirillo F, Galli GR, et al. Focal adhesion kinase (FAK) activation by estrogens involves GPER in triple-negative breast cancer cells. J Exp Clin Cancer Res. 2019;38(1):58.PubMedPubMedCentralCrossRef Rigiracciolo DC, Santolla MF, Lappano R, Vivacqua A, Cirillo F, Galli GR, et al. Focal adhesion kinase (FAK) activation by estrogens involves GPER in triple-negative breast cancer cells. J Exp Clin Cancer Res. 2019;38(1):58.PubMedPubMedCentralCrossRef
56.
Zurück zum Zitat Lee E, Fertig EJ, Jin K, Sukumar S, Pandey NB, Popel AS. Breast cancer cells condition lymphatic endothelial cells within pre-metastatic niches to promote metastasis. Nat Commun. 2014;5:4715.PubMedCrossRef Lee E, Fertig EJ, Jin K, Sukumar S, Pandey NB, Popel AS. Breast cancer cells condition lymphatic endothelial cells within pre-metastatic niches to promote metastasis. Nat Commun. 2014;5:4715.PubMedCrossRef
57.
Zurück zum Zitat Wang C, Li J, Ye S, Zhang Y, Li P, Wang L, et al. Oestrogen inhibits VEGF expression and angiogenesis in triple-negative breast Cancer by activating GPER-1. J Cancer. 2018;9(20):3802–11.PubMedPubMedCentralCrossRef Wang C, Li J, Ye S, Zhang Y, Li P, Wang L, et al. Oestrogen inhibits VEGF expression and angiogenesis in triple-negative breast Cancer by activating GPER-1. J Cancer. 2018;9(20):3802–11.PubMedPubMedCentralCrossRef
58.
Zurück zum Zitat Liu CY, Su JC, Huang TT, Chu PY, Huang CT, Wang WL, et al. Sorafenib analogue SC-60 induces apoptosis through the SHP-1/STAT3 pathway and enhances docetaxel cytotoxicity in triple-negative breast cancer cells. Mol Oncol. 2017;11(3):266–79.PubMedPubMedCentralCrossRef Liu CY, Su JC, Huang TT, Chu PY, Huang CT, Wang WL, et al. Sorafenib analogue SC-60 induces apoptosis through the SHP-1/STAT3 pathway and enhances docetaxel cytotoxicity in triple-negative breast cancer cells. Mol Oncol. 2017;11(3):266–79.PubMedPubMedCentralCrossRef
59.
Zurück zum Zitat Liu CY, Chen KF, Chao TI, Chu PY, Huang CT, Huang TT, et al. Sequential combination of docetaxel with a SHP-1 agonist enhanced suppression of p-STAT3 signaling and apoptosis in triple negative breast cancer cells. J Mol Med (Berl). 2017;95(9):965–75.CrossRef Liu CY, Chen KF, Chao TI, Chu PY, Huang CT, Huang TT, et al. Sequential combination of docetaxel with a SHP-1 agonist enhanced suppression of p-STAT3 signaling and apoptosis in triple negative breast cancer cells. J Mol Med (Berl). 2017;95(9):965–75.CrossRef
60.
Zurück zum Zitat Fatehi D, Soltani A, Ghatrehsamani M. SRT1720, a potential sensitizer for radiotherapy and cytotoxicity effects of NVB-BEZ235 in metastatic breast cancer cells. Pathol Res Pract. 2018;214(6):889–95.PubMedCrossRef Fatehi D, Soltani A, Ghatrehsamani M. SRT1720, a potential sensitizer for radiotherapy and cytotoxicity effects of NVB-BEZ235 in metastatic breast cancer cells. Pathol Res Pract. 2018;214(6):889–95.PubMedCrossRef
61.
Zurück zum Zitat Tzeng YT, Liu PF, Li JY, Liu LF, Kuo SY, Hsieh CW, et al. Kinome-wide siRNA screening identifies Src-enhanced resistance of chemotherapeutic drugs in triple-negative breast Cancer cells. Front Pharmacol. 2018;9:1285.PubMedPubMedCentralCrossRef Tzeng YT, Liu PF, Li JY, Liu LF, Kuo SY, Hsieh CW, et al. Kinome-wide siRNA screening identifies Src-enhanced resistance of chemotherapeutic drugs in triple-negative breast Cancer cells. Front Pharmacol. 2018;9:1285.PubMedPubMedCentralCrossRef
62.
Zurück zum Zitat Moreira MP, da Conceicao Braga L, Cassali GD, Silva LM. STAT3 as a promising chemoresistance biomarker associated with the CD44(+/high)/CD24(−/low)/ALDH(+) BCSCs-like subset of the triple-negative breast cancer (TNBC) cell line. Exp Cell Res. 2018;363(2):283–90.PubMedCrossRef Moreira MP, da Conceicao Braga L, Cassali GD, Silva LM. STAT3 as a promising chemoresistance biomarker associated with the CD44(+/high)/CD24(−/low)/ALDH(+) BCSCs-like subset of the triple-negative breast cancer (TNBC) cell line. Exp Cell Res. 2018;363(2):283–90.PubMedCrossRef
63.
Zurück zum Zitat Kuo WY, Hwu L, Wu CY, Lee JS, Chang CW, Liu RS. STAT3/NF-kappaB-regulated lentiviral TK/GCV suicide gene therapy for cisplatin-resistant triple-negative breast Cancer. Theranostics. 2017;7(3):647–63.PubMedPubMedCentralCrossRef Kuo WY, Hwu L, Wu CY, Lee JS, Chang CW, Liu RS. STAT3/NF-kappaB-regulated lentiviral TK/GCV suicide gene therapy for cisplatin-resistant triple-negative breast Cancer. Theranostics. 2017;7(3):647–63.PubMedPubMedCentralCrossRef
64.
Zurück zum Zitat Egusquiaguirre SP, Yeh JE, Walker SR, Liu S, Frank DA. The STAT3 target gene TNFRSF1A modulates the NF-kappaB pathway in breast Cancer cells. Neoplasia. 2018;20(5):489–98.PubMedPubMedCentralCrossRef Egusquiaguirre SP, Yeh JE, Walker SR, Liu S, Frank DA. The STAT3 target gene TNFRSF1A modulates the NF-kappaB pathway in breast Cancer cells. Neoplasia. 2018;20(5):489–98.PubMedPubMedCentralCrossRef
65.
Zurück zum Zitat Cheng CC, Shi LH, Wang XJ, Wang SX, Wan XQ, Liu SR, et al. Stat3/Oct-4/c-Myc signal circuit for regulating stemness-mediated doxorubicin resistance of triple-negative breast cancer cells and inhibitory effects of WP1066. Int J Oncol. 2018;53(1):339–48.PubMed Cheng CC, Shi LH, Wang XJ, Wang SX, Wan XQ, Liu SR, et al. Stat3/Oct-4/c-Myc signal circuit for regulating stemness-mediated doxorubicin resistance of triple-negative breast cancer cells and inhibitory effects of WP1066. Int J Oncol. 2018;53(1):339–48.PubMed
66.
Zurück zum Zitat Niu J, Xue A, Chi Y, Xue J, Wang W, Zhao Z, et al. Induction of miRNA-181a by genotoxic treatments promotes chemotherapeutic resistance and metastasis in breast cancer. Oncogene. 2016;35(10):1302–13.PubMedCrossRef Niu J, Xue A, Chi Y, Xue J, Wang W, Zhao Z, et al. Induction of miRNA-181a by genotoxic treatments promotes chemotherapeutic resistance and metastasis in breast cancer. Oncogene. 2016;35(10):1302–13.PubMedCrossRef
67.
Zurück zum Zitat Soleymani Abyaneh H, Gupta N, Radziwon-Balicka A, Jurasz P, Seubert J, Lai R, et al. STAT3 but not HIF-1alpha is important in mediating hypoxia-induced Chemoresistance in MDA-MB-231, a triple negative breast Cancer cell line. Cancers (Basel). 2017;9(10):137.PubMedCentralCrossRef Soleymani Abyaneh H, Gupta N, Radziwon-Balicka A, Jurasz P, Seubert J, Lai R, et al. STAT3 but not HIF-1alpha is important in mediating hypoxia-induced Chemoresistance in MDA-MB-231, a triple negative breast Cancer cell line. Cancers (Basel). 2017;9(10):137.PubMedCentralCrossRef
68.
Zurück zum Zitat Wang K, Zhu X, Zhang K, Yin Y, Chen Y, Zhang T. Interleukin-6 contributes to chemoresistance in MDA-MB-231 cells via targeting HIF-1alpha. J Biochem Mol Toxicol. 2018;32(3):e22039.PubMedCrossRef Wang K, Zhu X, Zhang K, Yin Y, Chen Y, Zhang T. Interleukin-6 contributes to chemoresistance in MDA-MB-231 cells via targeting HIF-1alpha. J Biochem Mol Toxicol. 2018;32(3):e22039.PubMedCrossRef
69.
Zurück zum Zitat Cui Y, Huang Y, Wu X, Zheng M, Xia Y, Fu Z, et al. Hypoxia-induced tRNA-derived fragments, novel regulatory factor for doxorubicin resistance in triple-negative breast cancer. J Cell Physiol. 2019;234(6):8740–51.PubMedCrossRef Cui Y, Huang Y, Wu X, Zheng M, Xia Y, Fu Z, et al. Hypoxia-induced tRNA-derived fragments, novel regulatory factor for doxorubicin resistance in triple-negative breast cancer. J Cell Physiol. 2019;234(6):8740–51.PubMedCrossRef
70.
Zurück zum Zitat Soleymani Abyaneh H, Soleimani AH, Vakili MR, Soudy R, Kaur K, Cuda F, et al. Modulation of hypoxia-induced Chemoresistance to polymeric micellar cisplatin: the effect of ligand modification of micellar carrier versus inhibition of the mediators of drug resistance. Pharmaceutics. 2018;10(4):196.PubMedCentralCrossRef Soleymani Abyaneh H, Soleimani AH, Vakili MR, Soudy R, Kaur K, Cuda F, et al. Modulation of hypoxia-induced Chemoresistance to polymeric micellar cisplatin: the effect of ligand modification of micellar carrier versus inhibition of the mediators of drug resistance. Pharmaceutics. 2018;10(4):196.PubMedCentralCrossRef
71.
Zurück zum Zitat Kitamura H, Ohno Y, Toyoshima Y, Ohtake J, Homma S, Kawamura H, et al. Interleukin-6/STAT3 signaling as a promising target to improve the efficacy of cancer immunotherapy. Cancer Sci. 2017;108(10):1947–52.PubMedPubMedCentralCrossRef Kitamura H, Ohno Y, Toyoshima Y, Ohtake J, Homma S, Kawamura H, et al. Interleukin-6/STAT3 signaling as a promising target to improve the efficacy of cancer immunotherapy. Cancer Sci. 2017;108(10):1947–52.PubMedPubMedCentralCrossRef
72.
Zurück zum Zitat Sasidharan Nair V, Toor SM, Ali BR, Elkord E. Dual inhibition of STAT1 and STAT3 activation downregulates expression of PD-L1 in human breast cancer cells. Expert Opin Ther Targets. 2018;22(6):547–57.PubMedCrossRef Sasidharan Nair V, Toor SM, Ali BR, Elkord E. Dual inhibition of STAT1 and STAT3 activation downregulates expression of PD-L1 in human breast cancer cells. Expert Opin Ther Targets. 2018;22(6):547–57.PubMedCrossRef
73.
Zurück zum Zitat Chen M, Pockaj B, Andreozzi M, Barrett MT, Krishna S, Eaton S, et al. JAK2 and PD-L1 amplification enhance the dynamic expression of PD-L1 in triple-negative breast Cancer. Clin Breast Cancer. 2018;18(5):e1205–15.PubMedCrossRef Chen M, Pockaj B, Andreozzi M, Barrett MT, Krishna S, Eaton S, et al. JAK2 and PD-L1 amplification enhance the dynamic expression of PD-L1 in triple-negative breast Cancer. Clin Breast Cancer. 2018;18(5):e1205–15.PubMedCrossRef
74.
Zurück zum Zitat Liu J, Yang Y, Wang H, Wang B, Zhao K, Jiang W, et al. Syntenin1/MDA-9 (SDCBP) induces immune evasion in triple-negative breast cancer by upregulating PD-L1. Breast Cancer Res Treat. 2018;171(2):345–57.PubMedCrossRef Liu J, Yang Y, Wang H, Wang B, Zhao K, Jiang W, et al. Syntenin1/MDA-9 (SDCBP) induces immune evasion in triple-negative breast cancer by upregulating PD-L1. Breast Cancer Res Treat. 2018;171(2):345–57.PubMedCrossRef
76.
Zurück zum Zitat Ibrahim SA, Hassan H, Vilardo L, Kumar SK, Kumar AV, Kelsch R, et al. Syndecan-1 (CD138) modulates triple-negative breast cancer stem cell properties via regulation of LRP-6 and IL-6-mediated STAT3 signaling. PLoS One. 2013;8(12):e85737.PubMedPubMedCentralCrossRef Ibrahim SA, Hassan H, Vilardo L, Kumar SK, Kumar AV, Kelsch R, et al. Syndecan-1 (CD138) modulates triple-negative breast cancer stem cell properties via regulation of LRP-6 and IL-6-mediated STAT3 signaling. PLoS One. 2013;8(12):e85737.PubMedPubMedCentralCrossRef
77.
Zurück zum Zitat Soleymani Abyaneh H, Gupta N, Alshareef A, Gopal K, Lavasanifar A, Lai R. Hypoxia induces the Acquisition of Cancer Stem-like Phenotype via Upregulation and Activation of signal transducer and activator of Transcription-3 (STAT3) in MDA-MB-231, a triple negative breast Cancer cell line. Cancer Microenviron. 2018;11(2–3):141–52.PubMedPubMedCentralCrossRef Soleymani Abyaneh H, Gupta N, Alshareef A, Gopal K, Lavasanifar A, Lai R. Hypoxia induces the Acquisition of Cancer Stem-like Phenotype via Upregulation and Activation of signal transducer and activator of Transcription-3 (STAT3) in MDA-MB-231, a triple negative breast Cancer cell line. Cancer Microenviron. 2018;11(2–3):141–52.PubMedPubMedCentralCrossRef
78.
Zurück zum Zitat Zhao D, Pan C, Sun J, Gilbert C, Drews-Elger K, Azzam DJ, et al. VEGF drives cancer-initiating stem cells through VEGFR-2/Stat3 signaling to upregulate Myc and Sox2. Oncogene. 2015;34(24):3107–19.PubMedCrossRef Zhao D, Pan C, Sun J, Gilbert C, Drews-Elger K, Azzam DJ, et al. VEGF drives cancer-initiating stem cells through VEGFR-2/Stat3 signaling to upregulate Myc and Sox2. Oncogene. 2015;34(24):3107–19.PubMedCrossRef
79.
80.
Zurück zum Zitat Nguyen TL, Grizzle WE, Zhang K, Hameed O, Siegal GP, Wei S. Syndecan-1 overexpression is associated with nonluminal subtypes and poor prognosis in advanced breast cancer. Am J Clin Pathol. 2013;140(4):468–74.PubMedCrossRef Nguyen TL, Grizzle WE, Zhang K, Hameed O, Siegal GP, Wei S. Syndecan-1 overexpression is associated with nonluminal subtypes and poor prognosis in advanced breast cancer. Am J Clin Pathol. 2013;140(4):468–74.PubMedCrossRef
81.
Zurück zum Zitat Ibrahim SA, Gadalla R, El-Ghonaimy EA, Samir O, Mohamed HT, Hassan H, et al. Syndecan-1 is a novel molecular marker for triple negative inflammatory breast cancer and modulates the cancer stem cell phenotype via the IL-6/STAT3, notch and EGFR signaling pathways. Mol Cancer. 2017;16(1):57.PubMedPubMedCentralCrossRef Ibrahim SA, Gadalla R, El-Ghonaimy EA, Samir O, Mohamed HT, Hassan H, et al. Syndecan-1 is a novel molecular marker for triple negative inflammatory breast cancer and modulates the cancer stem cell phenotype via the IL-6/STAT3, notch and EGFR signaling pathways. Mol Cancer. 2017;16(1):57.PubMedPubMedCentralCrossRef
82.
Zurück zum Zitat Zheng Q, Dunlap SM, Zhu J, Downs-Kelly E, Rich J, Hursting SD, et al. Leptin deficiency suppresses MMTV-Wnt-1 mammary tumor growth in obese mice and abrogates tumor initiating cell survival. Endocr Relat Cancer. 2011;18(4):491–503.PubMedPubMedCentralCrossRef Zheng Q, Dunlap SM, Zhu J, Downs-Kelly E, Rich J, Hursting SD, et al. Leptin deficiency suppresses MMTV-Wnt-1 mammary tumor growth in obese mice and abrogates tumor initiating cell survival. Endocr Relat Cancer. 2011;18(4):491–503.PubMedPubMedCentralCrossRef
83.
Zurück zum Zitat Thiagarajan PS, Zheng Q, Bhagrath M, Mulkearns-Hubert EE, Myers MG, Lathia JD, et al. STAT3 activation by leptin receptor is essential for TNBC stem cell maintenance. Endocr Relat Cancer. 2017;24(8):415–26.PubMedPubMedCentralCrossRef Thiagarajan PS, Zheng Q, Bhagrath M, Mulkearns-Hubert EE, Myers MG, Lathia JD, et al. STAT3 activation by leptin receptor is essential for TNBC stem cell maintenance. Endocr Relat Cancer. 2017;24(8):415–26.PubMedPubMedCentralCrossRef
84.
Zurück zum Zitat Liu Y, Choi DS, Sheng J, Ensor JE, Liang DH, Rodriguez-Aguayo C, et al. HN1L promotes triple-negative breast Cancer stem cells through LEPR-STAT3 pathway. Stem Cell Reports. 2018;10(1):212–27.PubMedCrossRef Liu Y, Choi DS, Sheng J, Ensor JE, Liang DH, Rodriguez-Aguayo C, et al. HN1L promotes triple-negative breast Cancer stem cells through LEPR-STAT3 pathway. Stem Cell Reports. 2018;10(1):212–27.PubMedCrossRef
85.
Zurück zum Zitat Maycotte P, Gearheart CM, Barnard R, Aryal S, Mulcahy Levy JM, Fosmire SP, et al. STAT3-mediated autophagy dependence identifies subtypes of breast cancer where autophagy inhibition can be efficacious. Cancer Res. 2014;74(9):2579–90.PubMedPubMedCentralCrossRef Maycotte P, Gearheart CM, Barnard R, Aryal S, Mulcahy Levy JM, Fosmire SP, et al. STAT3-mediated autophagy dependence identifies subtypes of breast cancer where autophagy inhibition can be efficacious. Cancer Res. 2014;74(9):2579–90.PubMedPubMedCentralCrossRef
86.
Zurück zum Zitat Huynh J, Etemadi N, Hollande F, Ernst M, Buchert M. The JAK/STAT3 axis: a comprehensive drug target for solid malignancies. Semin Cancer Biol. 2017;45:13–22.PubMedCrossRef Huynh J, Etemadi N, Hollande F, Ernst M, Buchert M. The JAK/STAT3 axis: a comprehensive drug target for solid malignancies. Semin Cancer Biol. 2017;45:13–22.PubMedCrossRef
87.
Zurück zum Zitat Vyas D, Lopez-Hisijos N, Shah P, Deshpande KS, Basson MD, Vyas A, et al. A second-generation proteasome inhibitor and doxorubicin modulates IL-6, pSTAT-3 and NF-kB activity in MDA-MB-231 breast Cancer cells. J Nanosci Nanotechnol. 2017;17(1):175–85.PubMedCrossRef Vyas D, Lopez-Hisijos N, Shah P, Deshpande KS, Basson MD, Vyas A, et al. A second-generation proteasome inhibitor and doxorubicin modulates IL-6, pSTAT-3 and NF-kB activity in MDA-MB-231 breast Cancer cells. J Nanosci Nanotechnol. 2017;17(1):175–85.PubMedCrossRef
88.
Zurück zum Zitat Aryappalli P, Al-Qubaisi SS, Attoub S, George JA, Arafat K, Ramadi KB, et al. The IL-6/STAT3 signaling pathway is an early target of Manuka honey-induced suppression of human breast Cancer cells. Front Oncol. 2017;7:167.PubMedPubMedCentralCrossRef Aryappalli P, Al-Qubaisi SS, Attoub S, George JA, Arafat K, Ramadi KB, et al. The IL-6/STAT3 signaling pathway is an early target of Manuka honey-induced suppression of human breast Cancer cells. Front Oncol. 2017;7:167.PubMedPubMedCentralCrossRef
89.
Zurück zum Zitat Tian J, Chen X, Fu S, Zhang R, Pan L, Cao Y, et al. Bazedoxifene is a novel IL-6/GP130 inhibitor for treating triple-negative breast cancer. Breast Cancer Res Treat. 2019. Tian J, Chen X, Fu S, Zhang R, Pan L, Cao Y, et al. Bazedoxifene is a novel IL-6/GP130 inhibitor for treating triple-negative breast cancer. Breast Cancer Res Treat. 2019.
90.
Zurück zum Zitat Fu S, Chen X, Lo HW, Lin J. Combined bazedoxifene and paclitaxel treatments inhibit cell viability, cell migration, colony formation, and tumor growth and induce apoptosis in breast cancer. Cancer Lett. 2019;448:11–9.PubMedCrossRef Fu S, Chen X, Lo HW, Lin J. Combined bazedoxifene and paclitaxel treatments inhibit cell viability, cell migration, colony formation, and tumor growth and induce apoptosis in breast cancer. Cancer Lett. 2019;448:11–9.PubMedCrossRef
91.
Zurück zum Zitat Rios-Fuller TJ, Ortiz-Soto G, Lacourt-Ventura M, Maldonado-Martinez G, Cubano LA, Schneider RJ, et al. Ganoderma lucidum extract (GLE) impairs breast cancer stem cells by targeting the STAT3 pathway. Oncotarget. 2018;9(89):35907–21.PubMedPubMedCentralCrossRef Rios-Fuller TJ, Ortiz-Soto G, Lacourt-Ventura M, Maldonado-Martinez G, Cubano LA, Schneider RJ, et al. Ganoderma lucidum extract (GLE) impairs breast cancer stem cells by targeting the STAT3 pathway. Oncotarget. 2018;9(89):35907–21.PubMedPubMedCentralCrossRef
92.
Zurück zum Zitat Mehta R, Katta H, Alimirah F, Patel R, Murillo G, Peng X, et al. Deguelin action involves c-met and EGFR signaling pathways in triple negative breast cancer cells. PLoS One. 2013;8(6):e65113.PubMedPubMedCentralCrossRef Mehta R, Katta H, Alimirah F, Patel R, Murillo G, Peng X, et al. Deguelin action involves c-met and EGFR signaling pathways in triple negative breast cancer cells. PLoS One. 2013;8(6):e65113.PubMedPubMedCentralCrossRef
93.
Zurück zum Zitat Yamashita N, Kondo M, Zhao S, Li W, Koike K, Nemoto K, et al. Picrasidine G decreases viability of MDA-MB 468 EGFR-overexpressing triple-negative breast cancer cells through inhibition of EGFR/STAT3 signaling pathway. Bioorg Med Chem Lett. 2017;27(11):2608–12.PubMedCrossRef Yamashita N, Kondo M, Zhao S, Li W, Koike K, Nemoto K, et al. Picrasidine G decreases viability of MDA-MB 468 EGFR-overexpressing triple-negative breast cancer cells through inhibition of EGFR/STAT3 signaling pathway. Bioorg Med Chem Lett. 2017;27(11):2608–12.PubMedCrossRef
94.
Zurück zum Zitat Chun J, Park MK, Ko H, Lee K, Kim YS. Bioassay-guided isolation of cantharidin from blister beetles and its anticancer activity through inhibition of epidermal growth factor receptor-mediated STAT3 and Akt pathways. J Nat Med. 2018;72(4):937–45.PubMedCrossRef Chun J, Park MK, Ko H, Lee K, Kim YS. Bioassay-guided isolation of cantharidin from blister beetles and its anticancer activity through inhibition of epidermal growth factor receptor-mediated STAT3 and Akt pathways. J Nat Med. 2018;72(4):937–45.PubMedCrossRef
95.
Zurück zum Zitat Kim S, Jeon M, Lee J, Han J, Oh SJ, Jung T, et al. Induction of fibronectin in response to epidermal growth factor is suppressed by silibinin through the inhibition of STAT3 in triple negative breast cancer cells. Oncol Rep. 2014;32(5):2230–6.PubMedCrossRef Kim S, Jeon M, Lee J, Han J, Oh SJ, Jung T, et al. Induction of fibronectin in response to epidermal growth factor is suppressed by silibinin through the inhibition of STAT3 in triple negative breast cancer cells. Oncol Rep. 2014;32(5):2230–6.PubMedCrossRef
96.
Zurück zum Zitat Jiang F, Li Y, Si L, Zhang Z, Li Z. Interaction of EZH2 and P65 is involved in the arsenic trioxide-induced anti-angiogenesis in human triple-negative breast cancer cells. Cell Biol Toxicol. 2019. Jiang F, Li Y, Si L, Zhang Z, Li Z. Interaction of EZH2 and P65 is involved in the arsenic trioxide-induced anti-angiogenesis in human triple-negative breast cancer cells. Cell Biol Toxicol. 2019.
97.
Zurück zum Zitat Byun HJ, Darvin P, Kang DY, Sp N, Joung YH, Park JH, et al. Silibinin downregulates MMP2 expression via Jak2/STAT3 pathway and inhibits the migration and invasive potential in MDA-MB-231 cells. Oncol Rep. 2017;37(6):3270–8.PubMedCrossRef Byun HJ, Darvin P, Kang DY, Sp N, Joung YH, Park JH, et al. Silibinin downregulates MMP2 expression via Jak2/STAT3 pathway and inhibits the migration and invasive potential in MDA-MB-231 cells. Oncol Rep. 2017;37(6):3270–8.PubMedCrossRef
98.
Zurück zum Zitat Yang Y, Zhou H, Liu W, Wu J, Yue X, Wang J, et al. Ganoderic acid a exerts antitumor activity against MDA-MB-231 human breast cancer cells by inhibiting the Janus kinase 2/signal transducer and activator of transcription 3 signaling pathway. Oncol Lett. 2018;16(5):6515–21.PubMedPubMedCentral Yang Y, Zhou H, Liu W, Wu J, Yue X, Wang J, et al. Ganoderic acid a exerts antitumor activity against MDA-MB-231 human breast cancer cells by inhibiting the Janus kinase 2/signal transducer and activator of transcription 3 signaling pathway. Oncol Lett. 2018;16(5):6515–21.PubMedPubMedCentral
99.
Zurück zum Zitat Liu CY, Huang TT, Chu PY, Huang CT, Lee CH, Wang WL, et al. The tyrosine kinase inhibitor nintedanib activates SHP-1 and induces apoptosis in triple-negative breast cancer cells. Exp Mol Med. 2017;49(8):e366.PubMedPubMedCentralCrossRef Liu CY, Huang TT, Chu PY, Huang CT, Lee CH, Wang WL, et al. The tyrosine kinase inhibitor nintedanib activates SHP-1 and induces apoptosis in triple-negative breast cancer cells. Exp Mol Med. 2017;49(8):e366.PubMedPubMedCentralCrossRef
100.
Zurück zum Zitat Su JC, Mar AC, Wu SH, Tai WT, Chu PY, Wu CY, et al. Disrupting VEGF-A paracrine and autocrine loops by targeting SHP-1 suppresses triple negative breast cancer metastasis. Sci Rep. 2016;6:28888.PubMedPubMedCentralCrossRef Su JC, Mar AC, Wu SH, Tai WT, Chu PY, Wu CY, et al. Disrupting VEGF-A paracrine and autocrine loops by targeting SHP-1 suppresses triple negative breast cancer metastasis. Sci Rep. 2016;6:28888.PubMedPubMedCentralCrossRef
101.
Zurück zum Zitat Lee HJ, Seo NJ, Jeong SJ, Park Y, Jung DB, Koh W, et al. Oral administration of penta-O-galloyl-beta-D-glucose suppresses triple-negative breast cancer xenograft growth and metastasis in strong association with JAK1-STAT3 inhibition. Carcinogenesis. 2011;32(6):804–11.PubMedPubMedCentralCrossRef Lee HJ, Seo NJ, Jeong SJ, Park Y, Jung DB, Koh W, et al. Oral administration of penta-O-galloyl-beta-D-glucose suppresses triple-negative breast cancer xenograft growth and metastasis in strong association with JAK1-STAT3 inhibition. Carcinogenesis. 2011;32(6):804–11.PubMedPubMedCentralCrossRef
102.
Zurück zum Zitat Liu CY, Su JC, Ni MH, Tseng LM, Chu PY, Wang DS, et al. Obatoclax analog SC-2001 inhibits STAT3 phosphorylation through enhancing SHP-1 expression and induces apoptosis in human breast cancer cells. Breast Cancer Res Treat. 2014;146(1):71–84.PubMedCrossRef Liu CY, Su JC, Ni MH, Tseng LM, Chu PY, Wang DS, et al. Obatoclax analog SC-2001 inhibits STAT3 phosphorylation through enhancing SHP-1 expression and induces apoptosis in human breast cancer cells. Breast Cancer Res Treat. 2014;146(1):71–84.PubMedCrossRef
103.
Zurück zum Zitat Yen MC, Shih YC, Hsu YL, Lin ES, Lin YS, Tsai EM, et al. Isolinderalactone enhances the inhibition of SOCS3 on STAT3 activity by decreasing miR-30c in breast cancer. Oncol Rep. 2016;35(3):1356–64.PubMedCrossRef Yen MC, Shih YC, Hsu YL, Lin ES, Lin YS, Tsai EM, et al. Isolinderalactone enhances the inhibition of SOCS3 on STAT3 activity by decreasing miR-30c in breast cancer. Oncol Rep. 2016;35(3):1356–64.PubMedCrossRef
104.
Zurück zum Zitat Kim HS, Hoang VH, Hong M, Chul Kim K, Ann J, Nguyen CT, et al. Investigation of B,C-ring truncated deguelin derivatives as heat shock protein 90 (HSP90) inhibitors for use as anti-breast cancer agents. Bioorg Med Chem. 2019;27(7):1370–81.PubMedCrossRef Kim HS, Hoang VH, Hong M, Chul Kim K, Ann J, Nguyen CT, et al. Investigation of B,C-ring truncated deguelin derivatives as heat shock protein 90 (HSP90) inhibitors for use as anti-breast cancer agents. Bioorg Med Chem. 2019;27(7):1370–81.PubMedCrossRef
105.
Zurück zum Zitat Cho TM, Kim JY, Kim YJ, Sung D, Oh E, Jang S, et al. C-terminal HSP90 inhibitor L80 elicits anti-metastatic effects in triple-negative breast cancer via STAT3 inhibition. Cancer Lett. 2019;447:141–53.PubMedCrossRef Cho TM, Kim JY, Kim YJ, Sung D, Oh E, Jang S, et al. C-terminal HSP90 inhibitor L80 elicits anti-metastatic effects in triple-negative breast cancer via STAT3 inhibition. Cancer Lett. 2019;447:141–53.PubMedCrossRef
106.
Zurück zum Zitat Abd El-Hafeez AA, Khalifa HO, Mahdy EAM, Sharma V, Hosoi T, Ghosh P, et al. Anticancer effect of nor-wogonin (5, 7, 8-trihydroxyflavone) on human triple-negative breast cancer cells via downregulation of TAK1, NF-kappaB, and STAT3. Pharmacol Rep. 2019;71(2):289–98.PubMedCrossRefPubMedCentral Abd El-Hafeez AA, Khalifa HO, Mahdy EAM, Sharma V, Hosoi T, Ghosh P, et al. Anticancer effect of nor-wogonin (5, 7, 8-trihydroxyflavone) on human triple-negative breast cancer cells via downregulation of TAK1, NF-kappaB, and STAT3. Pharmacol Rep. 2019;71(2):289–98.PubMedCrossRefPubMedCentral
107.
Zurück zum Zitat Tegowski M, Fan C, Baldwin AS. Thioridazine inhibits self-renewal in breast cancer cells via DRD2-dependent STAT3 inhibition, but induces a G1 arrest independent of DRD2. J Biol Chem. 2018;293(41):15977–90.PubMedCrossRefPubMedCentral Tegowski M, Fan C, Baldwin AS. Thioridazine inhibits self-renewal in breast cancer cells via DRD2-dependent STAT3 inhibition, but induces a G1 arrest independent of DRD2. J Biol Chem. 2018;293(41):15977–90.PubMedCrossRefPubMedCentral
108.
Zurück zum Zitat Chen Y, Ji M, Zhang S, Xue N, Xu H, Lin S, et al. Bt354 as a new STAT3 signaling pathway inhibitor against triple negative breast cancer. J Drug Target. 2018;26(10):920–30.PubMedCrossRef Chen Y, Ji M, Zhang S, Xue N, Xu H, Lin S, et al. Bt354 as a new STAT3 signaling pathway inhibitor against triple negative breast cancer. J Drug Target. 2018;26(10):920–30.PubMedCrossRef
109.
Zurück zum Zitat Dai X, Yin C, Zhang Y, Guo G, Zhao C, Wang O, et al. Osthole inhibits triple negative breast cancer cells by suppressing STAT3. J Exp Clin Cancer Res. 2018;37(1):322.PubMedPubMedCentralCrossRef Dai X, Yin C, Zhang Y, Guo G, Zhao C, Wang O, et al. Osthole inhibits triple negative breast cancer cells by suppressing STAT3. J Exp Clin Cancer Res. 2018;37(1):322.PubMedPubMedCentralCrossRef
110.
Zurück zum Zitat Feng T, Cao W, Shen W, Zhang L, Gu X, Guo Y, et al. Arctigenin inhibits STAT3 and exhibits anticancer potential in human triple-negative breast cancer therapy. Oncotarget. 2017;8(1):329–44.PubMedCrossRef Feng T, Cao W, Shen W, Zhang L, Gu X, Guo Y, et al. Arctigenin inhibits STAT3 and exhibits anticancer potential in human triple-negative breast cancer therapy. Oncotarget. 2017;8(1):329–44.PubMedCrossRef
111.
Zurück zum Zitat Chun J, Li RJ, Cheng MS, Kim YS. Alantolactone selectively suppresses STAT3 activation and exhibits potent anticancer activity in MDA-MB-231 cells. Cancer Lett. 2015;357(1):393–403.PubMedCrossRef Chun J, Li RJ, Cheng MS, Kim YS. Alantolactone selectively suppresses STAT3 activation and exhibits potent anticancer activity in MDA-MB-231 cells. Cancer Lett. 2015;357(1):393–403.PubMedCrossRef
112.
Zurück zum Zitat Shin DS, Kim HN, Shin KD, Yoon YJ, Kim SJ, Han DC, et al. Cryptotanshinone inhibits constitutive signal transducer and activator of transcription 3 function through blocking the dimerization in DU145 prostate cancer cells. Cancer Res. 2009;69(1):193–202.PubMedCrossRef Shin DS, Kim HN, Shin KD, Yoon YJ, Kim SJ, Han DC, et al. Cryptotanshinone inhibits constitutive signal transducer and activator of transcription 3 function through blocking the dimerization in DU145 prostate cancer cells. Cancer Res. 2009;69(1):193–202.PubMedCrossRef
113.
Zurück zum Zitat Zhang W, Yu W, Cai G, Zhu J, Zhang C, Li S, et al. A new synthetic derivative of cryptotanshinone KYZ3 as STAT3 inhibitor for triple-negative breast cancer therapy. Cell Death Dis. 2018;9(11):1098.PubMedPubMedCentralCrossRef Zhang W, Yu W, Cai G, Zhu J, Zhang C, Li S, et al. A new synthetic derivative of cryptotanshinone KYZ3 as STAT3 inhibitor for triple-negative breast cancer therapy. Cell Death Dis. 2018;9(11):1098.PubMedPubMedCentralCrossRef
114.
Zurück zum Zitat Chun J, Song K, Kim YS. Sesquiterpene lactones-enriched fraction of Inula helenium L. induces apoptosis through inhibition of signal transducers and activators of transcription 3 signaling pathway in MDA-MB-231 breast cancer cells. Phytother Res. 2018;32(12):2501–9.PubMedCrossRef Chun J, Song K, Kim YS. Sesquiterpene lactones-enriched fraction of Inula helenium L. induces apoptosis through inhibition of signal transducers and activators of transcription 3 signaling pathway in MDA-MB-231 breast cancer cells. Phytother Res. 2018;32(12):2501–9.PubMedCrossRef
115.
Zurück zum Zitat El Hasasna H, Saleh A, Al Samri H, Athamneh K, Attoub S, Arafat K, et al. Rhus coriaria suppresses angiogenesis, metastasis and tumor growth of breast cancer through inhibition of STAT3, NFkappaB and nitric oxide pathways. Sci Rep. 2016;6:21144.PubMedPubMedCentralCrossRef El Hasasna H, Saleh A, Al Samri H, Athamneh K, Attoub S, Arafat K, et al. Rhus coriaria suppresses angiogenesis, metastasis and tumor growth of breast cancer through inhibition of STAT3, NFkappaB and nitric oxide pathways. Sci Rep. 2016;6:21144.PubMedPubMedCentralCrossRef
116.
Zurück zum Zitat Dai X, Yin C, Guo G, Zhang Y, Zhao C, Qian J, et al. Schisandrin B exhibits potent anticancer activity in triple negative breast cancer by inhibiting STAT3. Toxicol Appl Pharmacol. 2018;358:110–9.PubMedCrossRef Dai X, Yin C, Guo G, Zhang Y, Zhao C, Qian J, et al. Schisandrin B exhibits potent anticancer activity in triple negative breast cancer by inhibiting STAT3. Toxicol Appl Pharmacol. 2018;358:110–9.PubMedCrossRef
117.
Zurück zum Zitat Yang B, Shen JW, Zhou DH, Zhao YP, Wang WQ, Zhu Y, et al. Precise discovery of a STAT3 inhibitor from Eupatorium lindleyanum and evaluation of its activity of anti-triple-negative breast cancer. Nat Prod Res. 2019;33:477-85.PubMedCrossRef Yang B, Shen JW, Zhou DH, Zhao YP, Wang WQ, Zhu Y, et al. Precise discovery of a STAT3 inhibitor from Eupatorium lindleyanum and evaluation of its activity of anti-triple-negative breast cancer. Nat Prod Res. 2019;33:477-85.PubMedCrossRef
118.
Zurück zum Zitat Kim HS, Kim T, Ko H, Lee J, Kim YS, Suh YG. Identification of galiellalactone-based novel STAT3-selective inhibitors with cytotoxic activities against triple-negative breast cancer cell lines. Bioorg Med Chem. 2017;25(19):5032–40.PubMedCrossRef Kim HS, Kim T, Ko H, Lee J, Kim YS, Suh YG. Identification of galiellalactone-based novel STAT3-selective inhibitors with cytotoxic activities against triple-negative breast cancer cell lines. Bioorg Med Chem. 2017;25(19):5032–40.PubMedCrossRef
119.
Zurück zum Zitat Li W, Zhang H, Nie M, Tian Y, Chen X, Chen C, et al. Ursolic acid derivative FZU-03,010 inhibits STAT3 and induces cell cycle arrest and apoptosis in renal and breast cancer cells. Acta Biochim Biophys Sin Shanghai. 2017;49(4):367–73.PubMedCrossRef Li W, Zhang H, Nie M, Tian Y, Chen X, Chen C, et al. Ursolic acid derivative FZU-03,010 inhibits STAT3 and induces cell cycle arrest and apoptosis in renal and breast cancer cells. Acta Biochim Biophys Sin Shanghai. 2017;49(4):367–73.PubMedCrossRef
120.
Zurück zum Zitat Lu L, Dong J, Wang L, Xia Q, Zhang D, Kim H, et al. Activation of STAT3 and Bcl-2 and reduction of reactive oxygen species (ROS) promote radioresistance in breast cancer and overcome of radioresistance with niclosamide. Oncogene. 2018;37(39):5292–304.PubMedCrossRef Lu L, Dong J, Wang L, Xia Q, Zhang D, Kim H, et al. Activation of STAT3 and Bcl-2 and reduction of reactive oxygen species (ROS) promote radioresistance in breast cancer and overcome of radioresistance with niclosamide. Oncogene. 2018;37(39):5292–304.PubMedCrossRef
121.
Zurück zum Zitat Oh E, Kim YJ, An H, Sung D, Cho TM, Farrand L, et al. Flubendazole elicits anti-metastatic effects in triple-negative breast cancer via STAT3 inhibition. Int J Cancer. 2018;143(8):1978–93.PubMedCrossRef Oh E, Kim YJ, An H, Sung D, Cho TM, Farrand L, et al. Flubendazole elicits anti-metastatic effects in triple-negative breast cancer via STAT3 inhibition. Int J Cancer. 2018;143(8):1978–93.PubMedCrossRef
122.
Zurück zum Zitat Kim YJ, Kim JY, Lee N, Oh E, Sung D, Cho TM, et al. Disulfiram suppresses cancer stem-like properties and STAT3 signaling in triple-negative breast cancer cells. Biochem Biophys Res Commun. 2017;486(4):1069–76.PubMedCrossRef Kim YJ, Kim JY, Lee N, Oh E, Sung D, Cho TM, et al. Disulfiram suppresses cancer stem-like properties and STAT3 signaling in triple-negative breast cancer cells. Biochem Biophys Res Commun. 2017;486(4):1069–76.PubMedCrossRef
123.
Zurück zum Zitat An H, Kim JY, Oh E, Lee N, Cho Y, Seo JH. Salinomycin promotes Anoikis and decreases the CD44+/CD24- stem-like population via inhibition of STAT3 activation in MDA-MB-231 cells. PLoS One. 2015;10(11):e0141919.PubMedPubMedCentralCrossRef An H, Kim JY, Oh E, Lee N, Cho Y, Seo JH. Salinomycin promotes Anoikis and decreases the CD44+/CD24- stem-like population via inhibition of STAT3 activation in MDA-MB-231 cells. PLoS One. 2015;10(11):e0141919.PubMedPubMedCentralCrossRef
124.
Zurück zum Zitat Deng XS, Wang S, Deng A, Liu B, Edgerton SM, Lind SE, et al. Metformin targets Stat3 to inhibit cell growth and induce apoptosis in triple-negative breast cancers. Cell Cycle. 2012;11(2):367–76.PubMedCrossRef Deng XS, Wang S, Deng A, Liu B, Edgerton SM, Lind SE, et al. Metformin targets Stat3 to inhibit cell growth and induce apoptosis in triple-negative breast cancers. Cell Cycle. 2012;11(2):367–76.PubMedCrossRef
125.
Zurück zum Zitat Hou S, Yi YW, Kang HJ, Zhang L, Kim HJ, Kong Y, et al. Novel carbazole inhibits phospho-STAT3 through induction of protein-tyrosine phosphatase PTPN6. J Med Chem. 2014;57(15):6342–53.PubMedPubMedCentralCrossRef Hou S, Yi YW, Kang HJ, Zhang L, Kim HJ, Kong Y, et al. Novel carbazole inhibits phospho-STAT3 through induction of protein-tyrosine phosphatase PTPN6. J Med Chem. 2014;57(15):6342–53.PubMedPubMedCentralCrossRef
126.
Zurück zum Zitat Kang HJ, Yi YW, Hou SJ, Kim HJ, Kong Y, Bae I, et al. Disruption of STAT3-DNMT1 interaction by SH-I-14 induces re-expression of tumor suppressor genes and inhibits growth of triple-negative breast tumor. Oncotarget. 2017;8(48):83457–68.PubMedCrossRef Kang HJ, Yi YW, Hou SJ, Kim HJ, Kong Y, Bae I, et al. Disruption of STAT3-DNMT1 interaction by SH-I-14 induces re-expression of tumor suppressor genes and inhibits growth of triple-negative breast tumor. Oncotarget. 2017;8(48):83457–68.PubMedCrossRef
127.
Zurück zum Zitat Lim EJ, Hong DY, Park JH, Joung YH, Darvin P, Kim SY, et al. Methylsulfonylmethane suppresses breast cancer growth by down-regulating STAT3 and STAT5b pathways. PLoS One. 2012;7(4):e33361.PubMedPubMedCentralCrossRef Lim EJ, Hong DY, Park JH, Joung YH, Darvin P, Kim SY, et al. Methylsulfonylmethane suppresses breast cancer growth by down-regulating STAT3 and STAT5b pathways. PLoS One. 2012;7(4):e33361.PubMedPubMedCentralCrossRef
128.
Zurück zum Zitat Chen W, Wang H, Cheng M, Ni L, Zou L, Yang Q, et al. Isoharringtonine inhibits breast cancer stem-like properties and STAT3 signaling. Biomed Pharmacother. 2018;103:435–42.PubMedCrossRef Chen W, Wang H, Cheng M, Ni L, Zou L, Yang Q, et al. Isoharringtonine inhibits breast cancer stem-like properties and STAT3 signaling. Biomed Pharmacother. 2018;103:435–42.PubMedCrossRef
129.
Zurück zum Zitat Kang DY, Sp N, Kim DH, Joung YH, Lee HG, Park YM, et al. Salidroside inhibits migration, invasion and angiogenesis of MDAMB 231 TNBC cells by regulating EGFR/Jak2/STAT3 signaling via MMP2. Int J Oncol. 2018;53(2):877–85.PubMed Kang DY, Sp N, Kim DH, Joung YH, Lee HG, Park YM, et al. Salidroside inhibits migration, invasion and angiogenesis of MDAMB 231 TNBC cells by regulating EGFR/Jak2/STAT3 signaling via MMP2. Int J Oncol. 2018;53(2):877–85.PubMed
130.
Zurück zum Zitat Njatcha C, Farooqui M, Kornberg A, Johnson DE, Grandis JR, Siegfried JM. STAT3 cyclic Decoy demonstrates robust antitumor effects in non-small cell lung Cancer. Mol Cancer Ther. 2018;17(9):1917–26.PubMedCrossRefPubMedCentral Njatcha C, Farooqui M, Kornberg A, Johnson DE, Grandis JR, Siegfried JM. STAT3 cyclic Decoy demonstrates robust antitumor effects in non-small cell lung Cancer. Mol Cancer Ther. 2018;17(9):1917–26.PubMedCrossRefPubMedCentral
131.
Zurück zum Zitat Reilley MJ, McCoon P, Cook C, Lyne P, Kurzrock R, Kim Y, et al. STAT3 antisense oligonucleotide AZD9150 in a subset of patients with heavily pretreated lymphoma: results of a phase 1b trial. J Immunother Cancer. 2018;6(1):119.PubMedPubMedCentralCrossRef Reilley MJ, McCoon P, Cook C, Lyne P, Kurzrock R, Kim Y, et al. STAT3 antisense oligonucleotide AZD9150 in a subset of patients with heavily pretreated lymphoma: results of a phase 1b trial. J Immunother Cancer. 2018;6(1):119.PubMedPubMedCentralCrossRef
132.
Zurück zum Zitat Hong D, Kurzrock R, Kim Y, Woessner R, Younes A, Nemunaitis J, et al. AZD9150, a next-generation antisense oligonucleotide inhibitor of STAT3 with early evidence of clinical activity in lymphoma and lung cancer. Sci Transl Med. 2015;7(314):314ra185.PubMedPubMedCentralCrossRef Hong D, Kurzrock R, Kim Y, Woessner R, Younes A, Nemunaitis J, et al. AZD9150, a next-generation antisense oligonucleotide inhibitor of STAT3 with early evidence of clinical activity in lymphoma and lung cancer. Sci Transl Med. 2015;7(314):314ra185.PubMedPubMedCentralCrossRef
133.
Zurück zum Zitat Jung KH, Yoo W, Stevenson HL, Deshpande D, Shen H, Gagea M, et al. Multifunctional effects of a small-molecule STAT3 inhibitor on NASH and hepatocellular carcinoma in mice. Clin Cancer Res. 2017;23(18):5537–46.PubMedPubMedCentralCrossRef Jung KH, Yoo W, Stevenson HL, Deshpande D, Shen H, Gagea M, et al. Multifunctional effects of a small-molecule STAT3 inhibitor on NASH and hepatocellular carcinoma in mice. Clin Cancer Res. 2017;23(18):5537–46.PubMedPubMedCentralCrossRef
134.
Zurück zum Zitat Wong AL, Soo RA, Tan DS, Lee SC, Lim JS, Marban PC, et al. Phase I and biomarker study of OPB-51602, a novel signal transducer and activator of transcription (STAT) 3 inhibitor, in patients with refractory solid malignancies. Ann Oncol. 2015;26(5):998–1005.PubMedCrossRef Wong AL, Soo RA, Tan DS, Lee SC, Lim JS, Marban PC, et al. Phase I and biomarker study of OPB-51602, a novel signal transducer and activator of transcription (STAT) 3 inhibitor, in patients with refractory solid malignancies. Ann Oncol. 2015;26(5):998–1005.PubMedCrossRef
135.
Zurück zum Zitat Jonker DJ, Nott L, Yoshino T, Gill S, Shapiro J, Ohtsu A, et al. Napabucasin versus placebo in refractory advanced colorectal cancer: a randomised phase 3 trial. Lancet Gastroenterol Hepatol. 2018;3(4):263–70.PubMedCrossRef Jonker DJ, Nott L, Yoshino T, Gill S, Shapiro J, Ohtsu A, et al. Napabucasin versus placebo in refractory advanced colorectal cancer: a randomised phase 3 trial. Lancet Gastroenterol Hepatol. 2018;3(4):263–70.PubMedCrossRef
136.
Zurück zum Zitat Oh DY, Lee SH, Han SW, Kim MJ, Kim TM, Kim TY, et al. Phase I study of OPB-31121, an Oral STAT3 inhibitor, in patients with advanced solid tumors. Cancer Res Treat. 2015;47(4):607–15.PubMedPubMedCentralCrossRef Oh DY, Lee SH, Han SW, Kim MJ, Kim TM, Kim TY, et al. Phase I study of OPB-31121, an Oral STAT3 inhibitor, in patients with advanced solid tumors. Cancer Res Treat. 2015;47(4):607–15.PubMedPubMedCentralCrossRef
137.
Zurück zum Zitat Yoo C, Kang J, Lim HY, Kim JH, Lee MA, Lee KH, et al. Phase I dose-finding study of OPB-111077, a novel STAT3 inhibitor, in patients with advanced hepatocellular carcinoma. Cancer Res Treat. 2019;51(2):510–8.PubMedCrossRef Yoo C, Kang J, Lim HY, Kim JH, Lee MA, Lee KH, et al. Phase I dose-finding study of OPB-111077, a novel STAT3 inhibitor, in patients with advanced hepatocellular carcinoma. Cancer Res Treat. 2019;51(2):510–8.PubMedCrossRef
138.
Zurück zum Zitat Khan MW, Saadalla A, Ewida AH, Al-Katranji K, Al-Saoudi G, Giaccone ZT, et al. The STAT3 inhibitor pyrimethamine displays anti-cancer and immune stimulatory effects in murine models of breast cancer. Cancer Immunol Immunother. 2018;67(1):13–23.PubMedCrossRef Khan MW, Saadalla A, Ewida AH, Al-Katranji K, Al-Saoudi G, Giaccone ZT, et al. The STAT3 inhibitor pyrimethamine displays anti-cancer and immune stimulatory effects in murine models of breast cancer. Cancer Immunol Immunother. 2018;67(1):13–23.PubMedCrossRef
139.
Zurück zum Zitat Wang ST, Ho HJ, Lin JT, Shieh JJ, Wu CY. Simvastatin-induced cell cycle arrest through inhibition of STAT3/SKP2 axis and activation of AMPK to promote p27 and p21 accumulation in hepatocellular carcinoma cells. Cell Death Dis. 2017;8(2):e2626.PubMedPubMedCentralCrossRef Wang ST, Ho HJ, Lin JT, Shieh JJ, Wu CY. Simvastatin-induced cell cycle arrest through inhibition of STAT3/SKP2 axis and activation of AMPK to promote p27 and p21 accumulation in hepatocellular carcinoma cells. Cell Death Dis. 2017;8(2):e2626.PubMedPubMedCentralCrossRef
141.
Zurück zum Zitat Venook AP, Niedzwiecki D, Lenz HJ, Innocenti F, Fruth B, Meyerhardt JA, et al. Effect of first-line chemotherapy combined with Cetuximab or bevacizumab on overall survival in patients with KRAS wild-type advanced or metastatic colorectal Cancer: a randomized clinical trial. JAMA. 2017;317(23):2392–401.PubMedPubMedCentralCrossRef Venook AP, Niedzwiecki D, Lenz HJ, Innocenti F, Fruth B, Meyerhardt JA, et al. Effect of first-line chemotherapy combined with Cetuximab or bevacizumab on overall survival in patients with KRAS wild-type advanced or metastatic colorectal Cancer: a randomized clinical trial. JAMA. 2017;317(23):2392–401.PubMedPubMedCentralCrossRef
142.
Zurück zum Zitat Medina PJ, Goodin S. Lapatinib: a dual inhibitor of human epidermal growth factor receptor tyrosine kinases. Clin Ther. 2008;30(8):1426–47.PubMedCrossRef Medina PJ, Goodin S. Lapatinib: a dual inhibitor of human epidermal growth factor receptor tyrosine kinases. Clin Ther. 2008;30(8):1426–47.PubMedCrossRef
143.
Zurück zum Zitat Song L, Morris M, Bagui T, Lee FY, Jove R, Haura EB. Dasatinib (BMS-354825) selectively induces apoptosis in lung cancer cells dependent on epidermal growth factor receptor signaling for survival. Cancer Res. 2006;66(11):5542–8.PubMedCrossRef Song L, Morris M, Bagui T, Lee FY, Jove R, Haura EB. Dasatinib (BMS-354825) selectively induces apoptosis in lung cancer cells dependent on epidermal growth factor receptor signaling for survival. Cancer Res. 2006;66(11):5542–8.PubMedCrossRef
144.
Zurück zum Zitat Chung SY, Chen YH, Lin PR, Chao TC, Su JC, Shiau CW, et al. Two novel SHP-1 agonists, SC-43 and SC-78, are more potent than regorafenib in suppressing the in vitro stemness of human colorectal cancer cells. Cell Death Discov. 2018;4:25.PubMedCrossRef Chung SY, Chen YH, Lin PR, Chao TC, Su JC, Shiau CW, et al. Two novel SHP-1 agonists, SC-43 and SC-78, are more potent than regorafenib in suppressing the in vitro stemness of human colorectal cancer cells. Cell Death Discov. 2018;4:25.PubMedCrossRef
145.
Zurück zum Zitat Bissonnette R, Maari C, Forman S, Bhatia N, Lee M, Fowler J, et al. The Oral JAK/SYK inhibitor ASN002 demonstrates efficacy and improves associated systemic inflammation in patients with moderate-to-severe atopic dermatitis: results from a randomised, double-blind, placebo-controlled study. Br J Dermatol. 2019. Bissonnette R, Maari C, Forman S, Bhatia N, Lee M, Fowler J, et al. The Oral JAK/SYK inhibitor ASN002 demonstrates efficacy and improves associated systemic inflammation in patients with moderate-to-severe atopic dermatitis: results from a randomised, double-blind, placebo-controlled study. Br J Dermatol. 2019.
146.
Zurück zum Zitat Zhang M, Xu CR, Shamiyeh E, Liu F, Yin JY, von Moltke LL, et al. A randomized, placebo-controlled study of the pharmacokinetics, pharmacodynamics, and tolerability of the oral JAK2 inhibitor fedratinib (SAR302503) in healthy volunteers. J Clin Pharmacol. 2014;54(4):415–21.PubMedCrossRef Zhang M, Xu CR, Shamiyeh E, Liu F, Yin JY, von Moltke LL, et al. A randomized, placebo-controlled study of the pharmacokinetics, pharmacodynamics, and tolerability of the oral JAK2 inhibitor fedratinib (SAR302503) in healthy volunteers. J Clin Pharmacol. 2014;54(4):415–21.PubMedCrossRef
147.
Zurück zum Zitat Hedvat M, Huszar D, Herrmann A, Gozgit JM, Schroeder A, Sheehy A, et al. The JAK2 inhibitor AZD1480 potently blocks Stat3 signaling and oncogenesis in solid tumors. Cancer Cell. 2009;16(6):487–97.PubMedPubMedCentralCrossRef Hedvat M, Huszar D, Herrmann A, Gozgit JM, Schroeder A, Sheehy A, et al. The JAK2 inhibitor AZD1480 potently blocks Stat3 signaling and oncogenesis in solid tumors. Cancer Cell. 2009;16(6):487–97.PubMedPubMedCentralCrossRef
148.
Zurück zum Zitat Verstovsek S, Manshouri T, Quintas-Cardama A, Harris D, Cortes J, Giles FJ, et al. WP1066, a novel JAK2 inhibitor, suppresses proliferation and induces apoptosis in erythroid human cells carrying the JAK2 V617F mutation. Clin Cancer Res. 2008;14(3):788–96.PubMedCrossRef Verstovsek S, Manshouri T, Quintas-Cardama A, Harris D, Cortes J, Giles FJ, et al. WP1066, a novel JAK2 inhibitor, suppresses proliferation and induces apoptosis in erythroid human cells carrying the JAK2 V617F mutation. Clin Cancer Res. 2008;14(3):788–96.PubMedCrossRef
Metadaten
Titel
STAT3 as a potential therapeutic target in triple negative breast cancer: a systematic review
verfasst von
Jiang-Jiang Qin
Li Yan
Jia Zhang
Wei-Dong Zhang
Publikationsdatum
01.12.2019
Verlag
BioMed Central
Erschienen in
Journal of Experimental & Clinical Cancer Research / Ausgabe 1/2019
Elektronische ISSN: 1756-9966
DOI
https://doi.org/10.1186/s13046-019-1206-z

Weitere Artikel der Ausgabe 1/2019

Journal of Experimental & Clinical Cancer Research 1/2019 Zur Ausgabe

Blutdrucksenkung könnte Uterusmyome verhindern

Frauen mit unbehandelter oder neu auftretender Hypertonie haben ein deutlich erhöhtes Risiko für Uterusmyome. Eine Therapie mit Antihypertensiva geht hingegen mit einer verringerten Inzidenz der gutartigen Tumoren einher.

Alphablocker schützt vor Miktionsproblemen nach der Biopsie

16.05.2024 alpha-1-Rezeptorantagonisten Nachrichten

Nach einer Prostatabiopsie treten häufig Probleme beim Wasserlassen auf. Ob sich das durch den periinterventionellen Einsatz von Alphablockern verhindern lässt, haben australische Mediziner im Zuge einer Metaanalyse untersucht.

Antikörper-Wirkstoff-Konjugat hält solide Tumoren in Schach

16.05.2024 Zielgerichtete Therapie Nachrichten

Trastuzumab deruxtecan scheint auch jenseits von Lungenkrebs gut gegen solide Tumoren mit HER2-Mutationen zu wirken. Dafür sprechen die Daten einer offenen Pan-Tumor-Studie.

Mammakarzinom: Senken Statine das krebsbedingte Sterberisiko?

15.05.2024 Mammakarzinom Nachrichten

Frauen mit lokalem oder metastasiertem Brustkrebs, die Statine einnehmen, haben eine niedrigere krebsspezifische Mortalität als Patientinnen, die dies nicht tun, legen neue Daten aus den USA nahe.

Update Onkologie

Bestellen Sie unseren Fach-Newsletter und bleiben Sie gut informiert.