Next Article in Journal
Decoding the Role of CD271 in Melanoma
Next Article in Special Issue
The Role of PPARγ Ligands in Breast Cancer: From Basic Research to Clinical Studies
Previous Article in Journal
Sonographic Risk Stratification Systems for Thyroid Nodules as Rule-Out Tests in Older Adults
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

STAT3, the Challenge for Chemotherapeutic and Radiotherapeutic Efficacy

1
The Key Laboratory of Molecular Biology for High Cancer Incidence Coastal Chaoshan Area, Shantou University Medical College, Shantou 515041, Guangdong, China
2
Institute of Oncologic Pathology, Shantou University Medical College, Shantou 515041, Guangdong, China
3
Department of Biochemistry and Molecular Biology, Shantou University Medical College, Shantou 515041, Guangdong, China
*
Authors to whom correspondence should be addressed.
Cancers 2020, 12(9), 2459; https://doi.org/10.3390/cancers12092459
Submission received: 26 July 2020 / Revised: 25 August 2020 / Accepted: 27 August 2020 / Published: 30 August 2020
(This article belongs to the Special Issue Tumor Suppressor Genes: Insight into the Cancer Therapy)

Abstract

:

Simple Summary

STAT3, an oncogene, contributes to insensitivity of chemotherapy and radiotherapy in tumor, reduces the clinical efficacy. Meanwhile, STAT3β, a STAT3 splicing isoform, is related to the inhibition of tumor growth and chemosensitivity. STAT3 may become a potential target to overcome the chemo(radio)resistance, which benefit for developing novel drugs targeting STAT3 or alternative splicing regulators.

Abstract

Chemoradiotherapy is one of the most effective and extensively used strategies for cancer treatment. Signal transducer and activator of transcription 3 (STAT3) regulates vital biological processes, such as cell proliferation and cell growth. It is constitutively activated in various cancers and limits the application of chemoradiotherapy. Accumulating evidence suggests that STAT3 regulates resistance to chemotherapy and radiotherapy and thereby impairs therapeutic efficacy by mediating its feedback loop and several target genes. The alternative splicing product STAT3β is often identified as a dominant-negative regulator, but it enhances sensitivity to chemotherapy and offers a new and challenging approach to reverse therapeutic resistance. We focus here on exploring the role of STAT3 in resistance to receptor tyrosine kinase (RTK) inhibitors and radiotherapy, outlining the potential of targeting STAT3 to overcome chemo(radio)resistance for improving clinical outcomes, and evaluating the importance of STAT3β as a potential therapeutic approach to overcomes chemo(radio)resistance. In this review, we discuss some new insights into the effect of STAT3 and its subtype STAT3β on chemoradiotherapy sensitivity, and we explore how these insights influence clinical treatment and drug development for cancer.

1. Introduction

Chemoradiotherapy is the main treatment for various solid malignancies through the induction of tumor DNA damage, and it can be used alone or in combination with surgery. Because of the genetic and epigenetic heterogeneity of cancer, the response to chemoradiotherapy is variable, and accompanied by therapeutic toxicities and resistances that limit the effectiveness of chemoradiotherapy [1,2,3,4]. In recent years, many reports have demonstrated that STAT3 contributes to resistance to chemotherapy and radiotherapy [5,6,7].
Signal transducer and activator of transcription (STAT) proteins are a family of cytoplasmic transcription factors and were discovered by James E. Darnell while investigating interferon signal transduction [8]. Seven mammalian members (STAT1, STAT2, STAT3, STAT4, STAT5a, STAT5b and STAT6) contribute to multiple biological functions, including cell differentiation, development, proliferation, apoptosis and inflammation [9]. STAT3 shares a common structure with other STAT proteins, and consists of an amino-terminal domain (NTD) that is required for cooperative DNA binding [10], a coiled-coil domain (CCD) for STAT3 recruitment to a receptor, a DNA-binding domain (DBD), a linker domain, an Src homology 2 (SH2) domain, which is involved in STAT3 dimerization, and a C-terminal transcriptional activation domain (TAD) for transcription activation (Figure 1) [11,12].
STAT3 is identified as an acute-phase response factor that is activated in response to IL-6, growth factors and hormones [13,14,15,16,17]. Once STAT3 Tyr705 is phosphorylated by receptor tyrosine kinases (JAK) or non-receptor tyrosine kinases (Src, Abl), STAT3 homodimerizes or heterodimerizes through its SH2 domain; it then translocates into the nucleus and binds to its recognition GAS (TTCN2-4GAA) elements in target genes to modulate gene transcription [18,19]. STAT3β is an alternatively spliced variant, in which the 55 C-terminal amino acid residues of STAT3α, including serine 727, are replaced by seven amino acid residues (FIDAVWK). Poly-C binding protein-1 (PCBP1) can interact with an exonic splicing suppressor in STAT3 exon 23 by binding to the sequence “UCCCCCCG”, and can then regulate the proportion of STAT3α/STAT3β (Figure 1) [20]. STAT3β is generally considered a dominant-negative regulator of STAT3α. However, it still acts as a significant transcriptional regulator for its own specific genes [21]. Constitutively-activated STAT3 has been frequently detected in human cancers and contributes to cancer progression [22,23,24,25,26,27,28,29,30]. STAT3 activation is also associated with maintaining embryonic stem cell self-renewal. While STAT3 deficiency causes early embryonic lethality, STAT3β itself can rescue the embryonic lethality [31,32,33].
Herein, this review focuses on the mechanism of STAT3 in resistance to chemo(radio)therapy and improvement in the therapeutic effect of chemoradiotherapy (CRT)-resistant tumors by targeting STAT3.

2. Feedback Loop Leading to STAT3 Activation

Mammalian cells use various regulatory mechanisms, involving feedback loops and crosstalk between signaling pathways, to adjust to perturbations in cellular homeostasis [34]. These mechanisms are pivotal in determining cell fate under dynamic physiological circumstances and pose a barrier to the effective therapy of cancer. The development of small-molecule inhibitors has significantly improved progression-free survival against receptor tyrosine kinases (RTKs), but disappointing outcomes remain due to the emergence of drug resistance (Figure 2). Interestingly, erlotinib, as an RTK inhibitor, increases STAT3 phosphorylation (Tyr705) in EGFR-mutant ono-small-cell lung cancer (NSCLC) cells through an autocrine loop, whereas knockdown of STAT3 decreases erlotinib-resistant colony numbers in the presence of erlotinib. This suggests that the interruption of a STAT3 feedback loop blocks chemotherapy resistance [35]. A similar result was reported for HER2-overexpressing breast and gastric cancers, where hyperactivated STAT3 signaling-mediated trastuzumab resistance occurred via a feedback loop comprising FN/EGF/IL-6 upstream mediators [36]. Moreover, MEK inhibitor induced STAT3 feedback activation, leading to resistance in KRAS mutant lung cancer cells [37]. MEK blockage has poor clinical outcome in KRAS-mutant (KRASMT) colorectal cancer cells, and siRNA-mediated knockdown of macrophage inhibitory factor (MIF) restored sensitivity to refametinib by decreasing STAT3 phosphorylation [38]. In addition, combined STAT3 and BCR-ABL1 inhibition can attenuate BCR-ABL1 kinase-dependent TKI resistance in chronic myeloid leukemia; additionally, combined with MEK inhibitors AZD6244, PD98059 and trametinib in KRASMT pancreatic and colon cancer cells, it increases the suppression of tumor growth [39,40]. These results confirm the cross-talk between STAT3 signaling and RTKs. Some phenomena suggest that STAT3 is activated through cross talk between RTKs. Upon MEK inhibitor treatment, c-MET, but not IL-6, upregulates phosphorylation of STAT3. The combination of JAK1/2 and c-MET inhibitors increased the effect of MEK inhibition in KRASMT xenograft models [41]. In addition, EGFR and IL-6 signaling could in turn activate STAT3 after treatment with erlotinib [35]. Clearly, a combination of STAT3 pathway inhibitors and RTKs may offer a promising strategy to improve chemotherapeutic efficacy.
Drug-sensitive, oncogene-addicted cancer cells secrete factors that contribute to drug resistance via feedback mechanisms. For example, the secretion of IL-6 in turn stimulates the STAT3 pathway and mediates resistance to PI3K inhibitors [42]. IL-11, a member of the IL-6 family, is upregulated in cisplatin-stimulated cancer-associated fibroblasts, and then IL-11 activates the STAT3 signaling pathway, leading to lung adenocarcinoma cell resistance to cisplatin [43]. DeClerck and colleagues revealed the relationship between environment-mediated drug resistance (EMDR) and STAT3. In the presence of soluble IL-6 receptor (sIL-6R), IL-6 effectively activated STAT3 in neuroblastoma and prevented drug-induced apoptosis in neuroblastoma in a STAT3-dependent manner. Furthermore, monocytes, non-myeloid cells and Treg cells were part of the reciprocal loop of STAT3 activation [44]. IL-6 can be involved in regulating leukemic stem cell chemoresistance in acute myeloid leukemia (AML) via osteopontin (OPN) autocrine/paracrine signaling. AML cell proliferation and apoptotic inhibition were mediated through the AKT/VEGF/STAT3/CXCR4/IL-6 loop under three-drug (daunorubicin, cytarabine and idarubicin) treatment [45]. Another cytokine, IL-10, causes STAT3 phosphorylation to decrease the sensitivity of B16 melanoma cells to chemotherapeutic agents through an autocrine loop [46]. IL-10RA upregulation drives the resistance to crizotinib in anaplastic large cell lymphoma (ALCL) through an autocrine positive feedback loop. The IL-10/IL-10RA signaling pathway can reverse the effects of crizotinib-inhibition on STAT3 activity. Activated STAT3 further binds to the transcriptional start sites of IL-10/IL-10RA/IL-10RB in ALCL cells [47]. Trastuzumab treatment rendered resistance to itself, facilitated epithelial-to-mesenchymal transition and enhanced metastatic potential in gastric cancer cells through an IL6/STAT3/Jagged-1/Notch positive feedback loop [48]. MicroRNAs that regulate STAT3 activation are additional molecular mediators that contribute towards chemotherapeutic resistance and tumorigenicity. The IL-6R/STAT3/miR-34a feedback loop was required for maintenance of the mesenchymal phenotype and promoted invasion and metastasis in colorectal cancer [49]. Similarly, an IL6R/STAT3/miR-204 feedback loop was correlated with the cisplatin resistance of epithelial ovarian cancer cells [50]. These reports indicate that miRNA-dependent regulation of the STAT3 axis could provide a chance to overcoming the chemoresistance of cancer patients.
In addition, STAT3 on Tyr705 correlates with Burkitt lymphoma (BL) chemoresistance. Zhong’s group revealed that STAT3 confers chemoresistance to cells by mediating the antioxidant feedback [51]. In response to reactive oxygen species (ROS) after chemotherapy, STAT3 is phosphorylated and increases the expression of glutathione peroxidase (GPx1) and superoxide dismutase 2 (SOD2), thus reducing the level of ROS. The reduction of ROS is related to multidrug (CODOX, cyclophosphamide, vincristine and doxorubicin) resistance in BL cells [51].

3. STAT3 Activation by Radiation

Radiotherapy, alone or in combination with surgery and chemotherapy, is an indispensable link in comprehensive cancer treatment. Radiotherapy causes DNA damage directly or indirectly through generation of ROS and induces various forms of cancer cell death, including apoptosis, autography, mitotic catastrophe, necrosis and senescence. Radioresistance of tumor cells is one of the main biological factors that affect clinical outcomes [52,53,54,55]. Accumulating evidence indicates a link between STAT3 and radioresistance [56,57,58,59,60]. STAT3 Y705 activation occurs in a dose- and time-dependent manner upon radiation, mediates hepatobiliary malignancies and maintains stem cell self-renewal and tumorigenic potential [61,62,63,64,65]. STAT3 correlates with resistance to radiotherapy in HER2-positive breast cancer tissue, and radiation resistance in breast cancer stem cells is strongly associated with a high expression of STAT3 [66,67]. Potential STAT3 inhibitors can decrease the production of ROS and down-regulate phosphorylation of STAT3 and Bcl-2, restoring sensitivity to radiation, which offers an effective approach for treating triple-negative breast cancer cells [68]. Treg cells can regulate resistance to radiation in head and neck cancer. STAT3 antisense oligonucleotide (ASO) inhibited the phosphorylation of STAT3. ASO, in combination with radiation, decreased the population of M2 macrophages, Treg cells and myeloid-derived suppressor cells and resulted in potential antitumor immune responses [69]. Constitutive phosphorylation of STAT3 Tyr705 is present in B-1 lymphocytes and contributes to radioresistance. The microenvironment of the peritoneal cavity is important for B-1 cell radiation-induced apoptosis (RiA) with high levels of IL-6Rα and IL-10RI. These cytokines are a requirement for B-1 cell radioresistance via activation of STAT3 [56,70]. STAT3 Ser727 phosphorylation is also associated with radioresistance and has been reported to correlate with shortened overall survival and progression-free survival of GBM (glioblastoma) patients [71,72]. Table 1 lists the correlation between STAT3 and chemo(radio)sensitization through the modulation of the STAT3 pathway [59,67,68,73,74,75,76,77,78,79,80,81].
The development of radioresistance following radiation therapy coincides with the radiation-induced appearance of pSTAT3, epithelial–mesenchymal transition (EMT) and self-renewal. IL6/STAT3/TWIST signaling pathway activation mediates esophageal squamous carcinoma EMT, whereas inactivation of STAT3 by an IL-6 inhibitor or siRNA-mediated downregulation of Twist enhances cancer cell apoptosis and reverses radiation-induced EMT [80]. The STAT3/Slug axis is also connected to EMT phenotypes and cancer stemness that contribute to radioresistance in glioblastoma [82]. Upregulating ALDH1 expression can also induce EMT-associated CSC-like properties and lead to radioresistance via STAT3 activation [83]. Park and colleagues identified signaling as a mechanism for CRC (colorectal cancer) stem cell persistence and radioresistance [78]. Lip-FLLL32 sensitized pancreatic CSCs to chemotherapy and radiotherapy both in vitro and in vivo [7]. ROS have been demonstrated in IR-induced EMT. Some reports describe higher levels of phospho-STAT3 and lower levels of ROS than those in radiosensitive cancer cells [68,84]. Radiation also leads to DNA damage. Several studies report that STAT3 signaling pathway activation is involved in DNA repair. For example, high expression of MRE11, a core protein of the MRN (MRE11/RAD50/NBS1) repair complex, enhances breast cancer cell proliferation and invasion and promotes DNA repair through STAT3 [85]. DNA repair-associated molecules, such as ATM, ATR and BRCA1/2, are upregulated via IL-6/STAT3 signaling in radiation-resistant prostate cancer [86].
Together, these findings provide convincing evidence that STAT3 is activated by radiation and contributes to the cancer phenotype. Both STAT3 Y705 and S727 are potential targets for overcoming clinical radioresistance. Interrupting the STAT3 pathway by using STAT3 inhibitors, RNA interference and short hairpin RNA could sensitize radioresistant cancer cells and improve clinical outcomes.
Table 1. STAT3 contributes to chemo(radio)resistance in various carcinoma types.
Table 1. STAT3 contributes to chemo(radio)resistance in various carcinoma types.
CancersCell LinesTreatmentInhibition of STAT3 Activation via Different Methods In Vitro or In VivoEffectsReference
Acute myeloid leukemiaKG-1/U937DNR/Ara-C/IDRSimvastatin/OPN siRNAin vitroSignificantly decreases the viability of cells and the expression level of STAT3, which sensitizes cells to chemotherapy.[45]
Anaplastic large cell lymphomaSUP-M2crizotinibStatticin vitroInhibition of STAT3 can reverse the resistance to crizotinib in IR10RA-overexpressed cells.[47]
B-1 lymphomaB-1 cellsRTSTAT3−/− micein vitro and in vivoIn STAT3−/− mice, B cells are more susceptible to radiation-induced apoptosis.[56]
Lung cancerA549/H358/H157RTNiclosamidein vitro and in vivoBlocks IR-induced activation of the STAT3/Bcl-2/Bcl-xL pathway and enhances apoptosis.[59]
A549RTAZD0530in vitroInhibits cell migration by blocking Src and enhances sensitivity to IR.[73]
A549RT/cisplatinshRNAin vitro and in vivoEnhances radiosensitivity both in vitro and in vivo and enhances chemosensitivity in vitro.[74,81]
HCC2429RTTG101209in vitro and in vivoInhibits STAT3 activation and survivin expression, induces apoptosis and decreases proliferation. [75]
PC-9ErlotinibSTAT3 siRNA/STAT3 shRNAin vitro and in vivoInhibition of STAT3 feedback sensitized lung adenocarcinoma to MEK inhibition.[35]
Breast cancerMCF-7RTXanthohumolin vitroSuppresses MDR1, EGFR and STAT3 expression, while increasing death receptor (DR)-4 and DR5 expression, and restores sensitivity to IR and doxorubicin. [77]
MDA-MB-231/MDA-MB-468 RTNiclosamide/STAT3 shRNAin vitro and in vivoNiclosamide inhibits STAT3 and Bcl-2 and increases ROS generation in vitro and in vivo; it is identified as a radiosensitizer. shRNA of STAT3 sensitizes breast cells to IR.[68]
SKBR3RTLapatinib/S3I-201in vitro and in vivoInhibition of the HER2-STAT3-survivin axis increases sensitivity in SKBR3 cells.[67]
BT474TrastuzumabS3I-201in vitro and in vivoSTAT3 inhibition significantly inhibits tumor growth and sensitizes breast cancer cells to trastuzumab.[36]
Colorectal cancerHCT116/LoVoRTJAK2 shRNAin vitro and in vivoDownregulation of JAK2/STAT3/CCND2 signaling to sensitize cells to radiotherapy and impair cancer stemness.[78]
HCT116/HCT295-FU/RTSelumetinibin vitro and in vivoIncreases mitotic catastrophe and apoptosis and decreases STAT3 activation and survivin expression; it also enhances sensitivity to radiotherapy in vivo. [79]
HCT116Refametinib4-IPPin vitroInhibition of 4-IPP sensitizes cancer cells to refametinib.[38]
Esophageal carcinomaECa109RTAG490in vitroReverses the IR-induced EMT phenotypes and gene expression via regulation of the IL-6/STAT3/Twist signaling pathway. Attenuates radioresistance under AG490 treatment.[80]
ECa109/TE13/KYSE150RTStatticin vitro and in vivoStattic inhibits STAT3 activation, downregulates HIF-1α and VEGF expression; and confers radiosensitivity in vitro and in vivo.[87]
ECa109/TE13RTNSC74859in vitro and in vivoSensitizes cells to radiotherapy in vitro and in vivo via inhibiting STAT3 activation and downregulation of HIF-1α and VEGF expression.[88]
GlioblastomaD456 GSCsRTIbrutinibin vitroCombines with radiotherapy to disrupt tumor growth and mainly disrupts glioma stem cells by inhibiting bone marrow and X-linked (BMX)/STAT3 activation.[64]
GBM-R212RTSTAT3 shRNAin vitroReduces STAT3, decrease Slug expression and suppresses cell invasion; inhibits cancer stem cell properties and enhances radiotherapy.[82]
GSC-2/ GSC-11RTStattic/WP1066in vitroEnhances radiosensitivity of GSC lines by inhibition of STAT3 activation, mainly impacting pSTAT3 on Serine727.[72]
U251/U87RTCryptotanshinone/WP1066/S3I-201in vitro and in vivoRe-sensitizes radioresistant cells to radiotherapy by inhibition of STAT3; when combined with ERK1/2 inhibitors, it remarkably eliminates resistant cells and inhibits tumor regrowth.[89]
Head and neck carcinomaUMSCC-17BRTStatticin vitro and in vivoReduces STAT3-mediated HIF-1α expression in response to Stattic.[90]
UMSCC-22A/UMSCC-22BRTLinifanibin vitroCell growth inhibition, G2/M cell cycle arrest and induction of cell death via apoptosis; it overcomes radioresistance by reducing pSTAT3 and expression of its target genes, e.g., cyclin D1, survivin.[91]
HNSCC-CD44(+)ALDH1(+) cellRTCucurbitacin Iin vitro and in vivoCombines with radiotherapy to suppress tumorigenesis and metastasis and also reduces CSC-like cells. [76]
HepatoblastomaHepG2.2.15RTSTAT3D Suppresses RT-induced hepatitis B virus DNA replication and impairs hepatobiliary malignancies.[62]
PLC5/Huh-7/Sk-Hep1/Hep3B RTSorafenib/STAT3 siRNAIn vitro and in vivoEnhances radiation-induced apoptosis by inhibiting STAT3; it also downregulates Mcl-1, cyclin D1 and survivin expression, overcomes resistance to radiation in cells and suppresses tumor growth in vivo. [92]
5-FU, 5-fluorouracil; Ara-C, cytarabine; Bcl-xL, B-cell lymphoma-extra large; DNR, daunorubicin; DR, death receptor; HIF-1α, hypoxia-inducible factor 1α; IDR, idarubicin; IR, irradiation; MDR1, multi-drug resistance 1; OPN, osteopontin; RT, radiation therapy; siRNA, small interfering RNA; shRNA, short hairpin RNA; STAT3D, dominant-negative mutant STAT3.

4. STAT3 Target Genes Impact Chemoresistance and Radioresistance

Activated STAT3 binds to DNA sequences (GAS element) in target genes and usually correlates with the enhanced expression of anti-apoptotic proteins, prevention of cell cycle arrest and promotion of cell proliferation [21]. A number of target genes, including Bcl-2 [68], Bcl-xL [5], Slug [82], Snail [93], survivin [94] and cyclin D1 [95], which have STAT3 binding sites in their promoters, regulate chemoresistance and radioresistance in a variety of cancer cells. Our group demonstrated that blocking the transcriptional activity of STAT3α by STAT3 isoform STAT3β sensitized esophageal squamous cell carcinoma (ESCC) cells to chemotherapeutic agents, namely, cisplatin and 5-FU both in vitro and in vivo [96]. The role of STAT3 target genes in chemo- and radioresistance is discussed in the following section.

4.1. Bcl-2 Family

Bcl-2, Bcl-xL and Mcl-1 are anti-apoptotic members of the Bcl-2 family and frequently dysregulated in various cancers. STAT3 has been shown to significantly enhance Bcl-2 and Bcl-xL promoter activity. Chromatin immunoprecipitation analysis indicates that activated STAT3 directly binds to the promoters of Bcl-2, Bcl-xL and Mcl-1 genes [97,98]. Hajime et al. further revealed the site of STAT3 that directly binds to the putative consensus sequences (position -92/-83) in the Mcl-1 promoter [99]. Thus, aberrant activation of STAT3 can activate Bcl-2 family proteins in various cancer cells [100]. Yu and colleagues suggested that STAT3 upregulated the expression of various target genes (Bcl-2, cyclin D1, c-myc) through a miR-197/CKS1B/STAT3 axis, which conferred chemoresistance to non-small-cell lung cancer [101]. Activated STAT3 increases RANTES (regulated upon activation, normal T cell expressed and secreted factor) and anti-apoptotic Bcl-2 and Bcl-xL expression levels that contributes to tamoxifen-resistance in breast cancer [97,100,102,103]. For cisplatin-induced mitochondrial-related apoptosis, knockdown of STAT3, by using siRNA, attenuated the expression of anti-apoptotic proteins Bcl-xL and Bcl-2 and increased the release of cytochrome C and the expression of Bax [102]. A similar result was found in ovarian cancer cells after treatment with the conditioned medium of cancer-associated fibroblasts [104]. In cisplatin-resistant ovarian cancer cells, NCX-4016 (nitro derivative of aspirin) treatment significantly decreases the protein levels of Bcl-2 and Bcl-xL and induces apoptosis in a time-dependent manner. The study further showed that the downregulation of STAT3 signaling could also inhibit tumor growth both in vitro and in vivo [103]. Doxorubicin-resistant chronic myeloid leukemia cells enhanced phosphorylation of STAT3 and dampened the tumor growth inhibition of TNF-α. STAT3 dephosphorylation can sensitize doxorubicin-resistant Dalton lymphoma to dendritic cell (DC)-derived TNF-α through the downregulation of Bcl-2 upon cucurbitacin I (STAT3 inhibitor) treatment in DC (chronic myeloid leukemia) patients [105]. (-)-Gossypol mainly disrupts Bcl-xL heterodimerization with Bax and Bad and overcomes the protection of prostate cancer cells by Bcl-xL. It also enhances the chemotherapy of docetaxel in inhibiting tumor growth and inducing apoptosis both in PC-3 cells and in xenograft model of prostate cancer [106].
The STAT3/Mcl-1 signaling axis is a target for overcoming resistance to BH3 mimetic ATB-737 (Bcl-2 inhibitor). Sorafenib, a multiple tyrosine kinase inhibitor, has been found to target Mcl-1 and synergize with ATB-737 to induce apoptosis in glioma cells [107]. However, (-)-gossypol also increases the expression levels of Mcl-1 [106], which may result in drug resistance. Sorafenib sensitizes prostate cancer cells to (-)-gossypol through the attenuation of Mcl-1 expression in vitro and in vivo. Using siRNA to downregulate Mcl-1, Lian et al.’s study also verified that prostate cancer cells were sensitized to (-)-gossypol [108].
Exposure of cells to IR (irradiation) can also increase Bcl-2 expression levels, which are associated with radioresistance [109]. In another study, radiotherapy resulted in the activation of STAT3 and Bcl-2 in triple-negative breast cancer. Persistent activation of STAT3 and Bcl-2 contributes to radioresistance [68]. Moreover, radioresistance was observed in DAB2 interactive protein-deficient prostate cells due to DNA double-strand break repair and the evasion of apoptosis with significantly higher levels of Bcl-2 and STAT3 [110]. Radiation induces the activation of STAT3 and increases the levels of Bcl-2/Bcl-xL in human lung cancer cells. Treatment with niclosamide can sensitize radioresistant lung cancer cells to ionizing radiation [59]. STAT3 and Mcl-1 are also induced by irradiation and are involved in the radioresistance of ESCC cells. A study further suggested that Mcl-1 inhibition was involved in the induction of apoptosis and the enhancement of DNA damage through SOCS1 overexpression [5]. Taking these results together, the upregulation of the Bcl-2 family proteins is the inescapable causative factor of chemo- and radioresistance.

4.2. EMT Regulators

Indeed, STAT3 is also important for initiating the epithelial-mesenchymal transition (EMT) program, which is associated with cancer therapy resistance [111]. Snail, Slug and Twist proteins are involved in regulating EMT [112]. Reports have shown that STAT3 can directly bind to the Slug promoters in a chromatin immunoprecipitation assay [82]. It also binds to Snail [93] and Twist1 [113] promoter and enhances their promoter activity. Reports have shown that STAT3/Slug was associated with GBM recurrence. STAT3 enhancement of tumor motility and EMT-like characteristics in GBM cells correlated with Slug expression. The efficacy of radiotherapy can be improved in vitro and in vivo by blocking STAT3/Slug signaling. Moreover, the blockage of STAT3/Slug also improves survival in GBM-R2I2 xenografts [82]. Slug was also upregulated in ovarian cancer and conferred chemoresistance to cells through the activation of STAT3 and the cross talk between p53/RAS signaling [114]. In other study, ovatodiolide (Ova), a diterpenoid isolate of Anisomeles indica, could overcome resistance to temozolomide (an alkylating agent) in glioblastoma. It reduced Slug, Vimentin, N-cadherin and β-catenin and also disrupted STAT3 signaling [115]. Moreover, Ova can significantly inhibit nasopharyngeal cancer cell tumor growth and enhance sensitivity to cisplatin in vivo. The study also found that Ova reduced Slug expression and inhibited EMT via abrogation of STAT3 signaling [116].
STAT3 upregulates the expression of Snail, contributes to temozolomide resistance in GBM and is associated with recurrent GBM tumors [117]. Another report also showed Snail/Slug-mediated chemoresistance to cisplatin in ovarian cancer cells [114]. Radioresistant head and neck squamous cell carcinoma cells showed high expression of Snail and Twist as the activation of STAT3 levels increased [76]. Increased expression of Snail was correlated with a poor prognosis in CRC patients. CRC cells that overexpressed Snail were also found to be more resistant to 5-FU [118]. Rectal cancer cells were resistant to ionizing radiation and 5-FU treatment due to the activation of STAT3 and the TGF-β/Smad signaling pathway. Treatment with metformin increased the sensitivity of rectal cancer cells by increasing apoptotic cell death as well as by downregulating Snail and Twist [119].
Twist basic helix loop helix transcription factor 1 (Twist1), a regulator of EMT, is upregulated in cisplatin-resistant ovarian cancer cells via STAT3 activation [120]. Inhibition of the IL6/STAT3/Twist signaling pathway could be a useful strategy to reverse radiation -induced EMT and radioresistance in ESCC [80]. Moreover, the inhibition of STAT3 activity and Twist1 transcription could suppress EMT and inhibit tumor progression and chemoresistance in ovarian cancer and renal cancer cells [113]. Wu et al. reported that DAB2 interactive protein suppressed the expression of Twist1 and the activation of STAT3. The report also demonstrated that Twist1 and STAT3 were crucial for the pirarubicin chemoresistance and tumor recurrence in non-muscle invasion bladder cancer, and this result could be reversed via DAB2 interactive protein [121].

4.3. Survivin

Survivin is an inhibitor of the apoptosis protein family, and its aberrant expression correlates with a poor prognosis and contributes to chemo(radio)resistance [122]. STAT3 is a potential transcriptional regulator of the survivin gene and binds to the survivin prompter at sites -264 to -256 [94]. Activation of STAT3 and survivin expression also confers resistance to chemotherapeutic agents (5-FU or cisplatin) in gastric cancer [123], hepatocellular carcinoma [124], NSCC [125] and ovarian cancer [104]. Survivin inhibitor MX106 effectively overcomes paclitaxel resistance in ovarian cancer cells [126]. In one study, STAT3 inhibition downregulated the expression of Bcl-xL, cyclin D1 and survivin, and induced apoptosis in a hepatocellular carcinoma xenograft model. The study also demonstrated that STAT3 inhibition enhanced chemosensitivity to cisplatin [127].
STAT3/survivin signaling regulates a poor response to radiotherapy in HER2-positive breast cancer [67], ESCC [5] and lung cancer [75]. Treatment with linifanib resulted in the induction of cell death via apoptosis and reduced activation of STAT3. It also decreased the expression of cyclin D1 and survivin and overcame radioresistance of head and neck squamous cell carcinoma [93]. Furthermore, using an inhibitor of JAK2, which is upstream of STAT3, affected survivin expression and sensitized lung cancer to radiation in vitro and in vivo [75]. Thus, inhibiting the expression of pSTAT3 and survivin can be efficient in improving the response to chemo- and radiotherapy.

4.4. Cyclin D1

Cyclin D1 peaks during mid-G1 when growth factor-deprived cells re-enter the cell cycle. Previous reports demonstrate that cyclin D1 confers chemo- and radioresistance to several tumor cells [128,129,130]. Activated STAT3 increases cyclin D1 mRNA expression, and binds to the positions -984, -568, -239 and -27 in human cyclin D1 promoters [95]. Several studies reported that inhibition of the STAT3 signaling pathway reduced the expression of cyclin D1 and enhanced the drug sensitivity of the cancer cells, including gastric carcinoma [131], cholangiocarcinoma [132], bladder cancer [133] and hepatocellular carcinoma [134]. An antisense cyclin D1 sequence in head and neck squamous cell carcinoma (HSNCC) cells inhibits cell growth, induces apoptosis and enhances sensitivity to chemotherapeutic agents both in vitro and in vivo [135,136]. Furthermore, Muneyuki et al. revealed that the levels of pSTAT3 played a causative role in the overexpression of cyclin D1 and provided a poor prognosis in HNSCC [137]. The researchers also demonstrated that a dominant-negative STAT3 strongly enhanced the cellular sensitivity of HNSCC cells to 5-FU and radiation [138].
Cyclin D1 can be stimulated by low-dose ionizing radiation (LDIR) in human keratinocytes. Inhibition of cyclin D1 using siRNA can inhibit the LDIR-associated anti-apoptotic response and eliminate LDIR-induced adaptive radioresistance [139]. Radioresistant ESCC cells possessed the EMT characteristic with cyclin D1 overexpression. Inhibition of cyclin D1 by using siRNA significantly decreased the cell proliferation rate and resulted in G0/G1 arrest, and it also enhanced sensitivity to radiation and reversed EMT [140]. Pestell’s group investigated the effect of cyclin D1 on radiosensitivity in prostate cancer cells. Cyclin D1 promoted the clonogenic survival of LNCaP cells by 6-fold upon radiation. Cyclin D1 knockdown using shRNA resulted in a radiation-dose dependent precipitous decline in clonogenic survival. The data suggested that cyclin D1 promoted radioresistance in LNCaP cells [141]. Cyclin D1 has also been implicated in nasopharyngeal carcinoma (NPC), resulting in cell survival by the cyclin D1-dependent DNA repair machinery. Moreover, controlling cyclin D1 by knocking down insulinoma-associated protein1 enhanced the sensitivity of NPC cells to radiation both in vitro and in vivo [142].

4.5. Immunosuppressive Molecules

Activation of STAT3 is important for tumor-induced immunosuppression, which regulates the expression of immunosuppressive molecules [143]. Yu’s study showed that blocking STAT3 signaling with STAT3β or antisense STAT3 oligonucleotide activated dendritic cells (DCs) by inducing the proinflammatory mediators IL-6, regulated upon activation normal T cell expressed and secreted factor (RANTES) and interferon-inducible protein-10 (IP-10) in tumor cells [144]. Further, they revealed that the expression of MHC class II, CD80 and CD86 was increased in tumor-infiltrating DCs of STAT3-null mice [145]. Moreover, T cells, DCs and natural killer (NK) cells are immune cells that are directly involved in anti-tumor responses regardless of the tumor’s sensitivity to the STAT3 inhibitor [145]. STAT3 influenced the maturation of DCs involving Paxillin-3-forkhead (PAX3-FKHR)-expressing cells. PXA3-FKHR interacted with STAT3 to reduce the expression of MHC and generate immunoinhibitory secreted factors such as IL-10 in tumor cells. Thus, the altered transcription induced by the PAX3-FKHR-STAT3 complex suppressed local inflammatory and immunological responses [146]. Another study demonstrated that tumor-derived factors prevented immature myeloid cells from differentiating into mature DCs via STAT3 activation. Except for the decrease in the expression of molecules (CD40, CD86) for T cell activation, the expression of MHC class II proteins also decreased [147].
IL-10 and TGF-β have been demonstrated to be involved in the suppression of the anti-tumor immune response of Treg cells [148]. Studies have reported that STAT3 bound to the promoters of the encoding genes of IL-10 [149] and TGF-β [150]. STAT3 is a key regulator of the Treg cells phenotype with the expression of IL-10, TGF-β and FOXP3 of nucleophosmin/anaplastic lymphoma kinase (NPM/ALK)-carrying T cell lymphoma (ALK+TCL) cells. IL-10 from ALK+TCL cells displays immunosuppressive activity, inhibits CD3/CD28 antibody-stimulated peripheral blood mononuclear cells and impairs the activation of T lymphocytes [151]. CD4+ CD25+ regulatory T cells are an important population of immunosuppressive cells. Treg cells from 12B1 tumor-bearing animals suppressed the function of DCs in a TGF-β- and IL-10-dependent manner and were associated with the activation of STAT3 [148]. Suppressor of cytokine signaling 3 (SOCS3)-deficient CD4+ T cells have a lower type 2 T helper cell (Th2) immune response and considerably increase the expression level of IL-10 and TGF-β for Th3-like differentiation via the enhanced recruitment of STAT3 to their promoters [150].
STAT3 has been identified on the promoter of vascular endothelial growth factor (VEGF) and was found to promote the growth and metastasis of human pancreatic cancer cells [152]. Chronic lymphocytic leukemia B cells secrete VEGF and activate the VEGF receptor (VEGFR). VEGF–VEGFR signaling is associated with abundant phosphorylated-STAT3 in the cell nucleus, which conferred apoptotic resistance to these cells [153]. Recently, Ong and colleagues found that a novel STAT3 mutation (p.D427H, E616G, p.E616K and p.E696K) increased phosphorylation of STAT3 at Tyr705 in STAT3 mutants. Further, p.E616K regulated programmed death-ligand 1(PD-L1) expression via the binding of STAT3 to the promoter of PD-L1 in NK/T-cell lymphoma (NKTL). The team suggested that inhibitors of STAT3 and PD-L1 might be a promising therapeutic strategy for NKTL [154]. Castration-resistant prostate cancer (CRPC) cells increased the expression of PD-L1 which participated in tumor immune evasion under hypoxic conditions. Silencing STAT3 signaling can reverse the level of PD-L1 and enhance the susceptibility of CRPC cells to NK cell immunity [155].

5. The Alternative Splicing Product STAT3β

5.1. Antitumorigenic Potential of STAT3β

In various cells and tissues, the ratio of STAT3α to STAT3β ranges from 3:1 to 10:1 at the mRNA level, and from 1:3 to 10:1 at the protein level. Biethahn and colleagues found that STAT3β was expressed at a high level in CD34+ cells by Western blot analysis [156,157,158]. Because of the low expression levels of STAT3β, its role remained elusive for a long time. Woude and colleagues reported that STAT3β had no effects on branching morphogenesis or invasion of leiomyosarcoma cells. However, STAT3β significantly inhibited cell growth in soft agar in a dose-dependent manner [159], while cell-specific STAT3β expression in macrophages reduced the invasion and cell mobility of breast cancer cells and significantly suppressed breast tumor growth [160].
Given a truncated C-terminal transactivation domain, transcriptional activation by STAT3β probably involves cooperation with other transcription factors, such as c-Jun and STRA13 [161,162,163]. STAT3β has more prolonged Y705 phosphorylation and nuclear retention kinetics than STAT3α, impacts STAT3α-dependent transcription and regulates a number of genes [21,164]. Cooperating with c-Jun, STAT3β increases Fas expression and the sensitization of melanoma cells to Fas ligand-induced apoptosis [161,165]. Zammarchi and colleagues revealed that the STAT3β switch leads to tumor regression in vivo. Furthermore, they revealed that STAT3β has its own specific set of target genes (such as Cyclin C, IL-8 and PEX1) and increases breast cancer cell death [166]. Transient transfection of STAT3β strongly inhibits transcriptional activation of c-Src dependent Bcl-xL [167]. As a potential antitumorigenic molecule, overexpression of STAT3β inhibits the transcriptional activity of STAT3α, down-regulates the anti-apoptotic gene Bcl-xL and the cell cycle-related gene cyclin D1, and suppresses the proliferation of lung cancer cells [168]. In breast cancer cells, STAT3β mediates cells’ growth inhibition and leads to cell cycle arrest and apoptosis. Further, Niu and colleagues showed that STAT3β can induce the expression of TRAIL (tumor necrosis factor-related apoptosis-inducing ligand), which is a tumor-specific apoptotic effector [169]. Intriguingly, the expression of STAT3β can rescue the embryonic lethality of STAT3 deficient mice and induce the expression of specific STAT3 target genes [33].
STAT3β has gained attention as a potent tumor suppressor and a favorable prognostic factor in ESCC with or without chemoradiotherapy, and it strongly correlates with longer overall survival and recurrence-free survival [96]. Recently, it has been found that STAT3β correlates with a favorable prognosis and prolonged survival, and also has a tumor-suppressive effect in acute myeloid leukemia via the regulation of its target gene, SELL [170]. Taking these findings together, STAT3β is a potential protective prognostic marker.

5.2. STAT3β Switch Modulates Chemo(Radio)therapy Sensitivity

As mentioned earlier, STAT3β is a truncated form of STAT3α due to alternative splicing and could be of value if one could control STAT3 alternative splicing. Nevo-Caspi and colleagues show that the level of ADAR editing enzymes increases upon deferoxamine treatment in lymphoblastoid cells, elevating the level of editing in Alu sequences residing in STAT3 introns [171]. They further demonstrate that ADAR1 affects STAT3 alternative splicing by RNA editing, which increases the expression of STAT3β [172].
Few studies have focused on switching STAT3α to STAT3β, which contributes to the inhibition of cancer cell growth (Figure 2). As mentioned above, PCBP1can regulate the proportion of STAT3α/STAT3β. PCBP1 inhibits oral squamous cell carcinoma cell growth and the expression of Bcl-xL and survivin, and it reverses the function of STAT3α [172,173]. Recently, phosphorodiamidate morpholino oligomers (morpholinos) were targeted to a splicing enhancer, and then STAT3α was specifically switched to STAT3β, which led to apoptosis and cell cycle arrest in breast cancer cells [166]. The splicing reprogramming offers an effective therapeutic approach for cancer. Similar results were obtained by using STAT3α-to-β expression-switching oligonucleotides to inhibit breast cancer cell growth and migration [174].
STAT3 has been reported in chemoresistance and radioresistance. In contrast, STAT3β plays an essential tumor-suppressive role in cancer [19,175]. Phosphorylation of STAT3 Ser727 is related to intrinsic radioresistance. STAT3β lacks the C-terminal 55 amino acids (containing S727), which indicates that STAT3β may enhance sensitivity to radiation. The roles of STAT3β in conferring radiation sensitivity to cells are still unclear to date [71,176]. Currently, STAT3β has been shown to be involved in regulating chemoresistance. STAT3β-transfected gastric and breast cancer cells inhibited the invasion of gastric cancer cells, promoted apoptosis and improved sensitivity to chemotherapy [177]. Interestingly, STAT3β expression is correlated with overall survival and recurrence-free survival in esophageal squamous cell carcinoma (ESCC) patients with or without chemoradiotherapy and sensitizes tumor xenografts to chemotherapy (cisplatin and 5-FU) mainly by blocking the transcriptional activity of STAT3α [96]. Taken together, these findings indicated that STAT3β is a potential molecule for sensitization to chemo(radio)therapy in cancer patients.

6. STAT3 as a Therapeutic Target

Early clinical studies demonstrated the promise of therapy that targets STAT3. However, no inhibitors that directly target STAT3 have been approved by the FDA for clinical application. In the following section, we focus on inhibitors that directly target STAT3 through its N-terminal domain, DNA-binding domain, linker domain and SH2 domain (Figure 3).

6.1. Targeting the STAT3 N-Terminal Domain

The N-terminal domain is essential for DNA binding, nuclear translation and protein–protein interactions, and a crystal structure is available for designing STAT3 NTD inhibitors [178]. A structural study has suggested that the overall fold of the domain is similar for different STATs. STAT4 structural data and sequence homology are used to design peptide analogs of STAT3 helix 2. The peptide (DTRYLEQLHQLY) was found to directly and specifically bind to STAT3, as determined by fluorescence resonance energy transfer (FRET) in cells expressing GFP-STAT3. The derivative potently inhibited survival and induced apoptosis in breast cancer cells, but not normal breast cells [179]. Furthermore, ST3-H2A2 (LDTRYLEQLHKLY) bound to both phosphorylated and unphosphorylated STAT3 in prostate cancer cells, decreased STAT3 DNA binding ability and induced apoptotic cell death [180].

6.2. Targeting the STAT3 DNA-Binding Domain

Blocking STAT3 by targeting the DBD domain could affect DNA binding ability and has the potential to inhibit STAT3-dependent gene transcription. Galiellalactone (Figure 3a), a natural product, was isolated from fermentations of the ascomycete strain A111-95. It can block IL-6 signaling by binding to the DNA binding sites of activated STAT3 dimers, but it does not affect the activation of STAT3 Tyr701 and Ser727 [181]. These anticancer agents have been investigated in prostate cancer cells and stem cell-like ALDH-positive prostate cancer cells both in vivo and in vitro. They led to a reduction in anti-apoptotic or cell cycle regulatory proteins through the inhibition of STAT3 signaling [182,183]. In a study on the effectiveness of galiellalactone in prostate cancer cells, Don-Doncow et al. showed that it acts as a cysteine reactive inhibitor and binds to one or more cysteines in STAT3 (Cys-367, Cys-468, Cys-542). The compound could lead to the inhibition of STAT3 binding to DNA [184]. Recently, it was shown to suppress the resistance of prostate cancer cells to enzalutamide [185], and its analogs SG-1709 (Figure 3b) and SG-1721 (Figure 3c) exhibited even greater effectiveness in blocking STAT3 signaling and inducing apoptosis. Interestingly, SG-1721 combined with radiotherapy showed enhanced apoptosis in triple-negative breast cancer cells [186]. To deal with the oral bioavailability of galiellalactone, the galiellalactone prodrug GPA512 (Figure 3d) was synthesized and shown to be able to reduce tumor growth in vivo through oral administration. The effect of GPA512 was similar to that of galiellalactone [187].
Niclosamide, an FDA-approved anthelmintic drug, as well as other small-molecule inhibitors, inhibits STAT3 activity. In combination with radiation, it can overcome the radioresistance of lung cancer xenografts [59]. Even though it potently inhibits the activation and transcriptional function of STAT3, its poor aqueous solubility and bioavailability limit its clinical development. A series of novel O-alkylamino-tethered niclosamide derivatives have been designed, with one, denoted HJC0152, being able to suppress breast cancer cell tumor growth in vivo. Its excellent aqueous solubility means that it can be taken orally in clinical cancer therapy [188].
Structure-based drug design and computational docking have been used for screening small-molecule inhibitors that target the DNA-binding domain of STAT3. The DBD-binding drug inS3-54 (Figure 3e) selectively inhibits the DNA-binding activity of STAT3, but it has off-target effects. The same team further designed an improved lead compound (inS3-54A18, Figure 3f) that not only binds to DBD directly, but also inhibits the expression of downstream STAT3 target genes. Its oral availability and anti-tumor effects in lung xenograft models argue against the previously thought “undruggable” nature of a DBD [189,190]. Ligand-based pharmacophore models of STAT3 inhibitors are being used in combination with a 3D shape- and electrostatic-based drug approach. LC28 is identified as a STAT3 DBD inhibitor through the pharmacophore of the inS3-45 analog A18. The compound inhibits the survival of cisplatin-resistant ovarian cancer cells [191]. An LC28 analog, MMPP exhibits the strongest binding affinity to STAT3 and binds selectively to the DBD, especially to T456. MMPP effectively inhibits STAT3 in vitro and in vivo and induces G1-phase cell cycle arrest and apoptosis [192]. On the basis of the conjugation of a diarylidenyl-piperidone (DAP) backbone to a nitroxide precursor, Kellie identified a novel small-molecule inhibitor, HO-3867, a curcumin analog that directly interacts with the STAT3 DNA-binding domain. HO-3867 has been studied in ovarian and pancreatic cancer cells, and in an ovarian cancer xenograft mouse model [193,194,195]. A molecular dynamics simulation was used for virtual ligand screening of a chemical library, and Buettner et al. found that C48 is a STAT3 inhibitor that selectively binds to Cys468 of STAT3 and blocks phosphorylation of STAT3 to inhibit tumor growth in vivo [196].

6.3. Targeting the STAT3 Linker Domain

Researchers have found a small-molecule inhibitor of the STAT3 linker domain. Tatsuya et al. identified the benzoquinone derivative BPMB as a STAT3 inhibitor. BPMB could selectively inhibit proliferation in constitutively-activated STAT3 breast cancer cells. From an analysis by matrix-assisted laser desorption/ionization-mass spectrometry, Cys550 was found to be an important residue for crosslinking between STAT3 and BPMB. Cys550 could be a drug target for the development of irreversible STAT3 inhibitors [197]. Furthermore, Berg and colleagues identified catechol bisphosphate as a STAT5b inhibitor that exhibited a higher degree of selectivity than STAT5a, which was determined by the linker domain (position 566). Given the homology of the STAT family, it is extremely likely that the linker domain governs selectivity in a similar manner in STAT3 [198].

6.4. Targeting the STAT3 SH2 Domain

Phosphorylation of tyrosine residues in the SH2 domain of STAT3 is essential for receptor recognition as well as dimerization. In a search to identify small molecule inhibitors of STAT3, an SH2 domain-binding phosphopeptide PY*LKTK (Y* represents phosphotyrosine) was identified and formed inactive STAT3·PY*LKTK complexes. It has significant in vivo activity and suppresses v-Src transformation by blocking STAT3 signaling [20]. Since the first peptide inhibitor of STAT3 was identified, a number of small-molecule inhibitors that inhibit STAT3 have been demonstrated. On the basis on the lead compound PY*L (PY*LKTK tripeptide), Turkson found a selective and more potent peptide mimetic, denoted ISS 610. It disrupts STAT3 activity in vitro, inhibits cell growth and induces apoptosis in Src-transformed fibroblasts [199]. Derived from the STAT3 SH2 domain, peptide mimetic SPI has been tested in pancreatic, breast, prostate and non-small lung cancer cells. SPI was shown to have good effectiveness, including viability loss and apoptosis, in all cell lines tested [200]. Other peptides and phosphopeptide mimetics have been reported and used in cancer cells, such as CJ-1383 and PM-73G. Both can selectively inhibit STAT3. CJ-1383 induced breast cancer cell apoptosis in a dose-dependent manner, while inhibition of tumor growth by the phosphopeptide mimetic PM-73G occurred without apoptosis or changes in the expression of cyclin D1 or survivin in xenograft models [201].
High-throughput screening of chemical libraries and computational docking studies have been used to discover inhibitors of STAT3. Stattic (6-nitro-benzo(b)thiophene-1,1-dioxide 1) was found to bind to the STAT3 SH2 domain. Another study demonstrated that Stattic enhanced radiosensitivity [202,203]. STA-21 (Figure 3g), a natural product analog of tetrangomycin, was discovered via structure-based virtual screening to bind to the SH2 domain and form hydrogen bonds with Arg595, Arg609 and Ile634 [204]. Phase I/II clinical trials (NCT01047943, 13 January 2010) for STA-21 have been performed for the treatment of psoriasis [205,206]. Analogs of STA-21 were reported later, such as LLL-3 (Figure 3h) and LLL-12 (Figure 3i), which can suppress cancer cell proliferation and induce apoptosis [207,208,209]. Using in silico site-directed fragment-based drug design (FBDD), the small-molecule STAT3 inhibitor LY5 (Figure 3j), an analog of STA-21, was tested in colon cancer cells. LY5 inhibited STAT3 phosphorylation and suppressed colon tumor growth in vivo [210]. The results suggest that LY5 can be an effective agent in cancer patients with constitutive activation of STAT3, and represents a promising approach for overcoming drug resistance induced by feedback activation of STAT3.
S3I-201 (NSC 74859, Figure 3k) is a selective chemical probe inhibitor of STAT3 and binds to the STAT3 phosphotyrosine peptide (PY*LKT) with the potential to become a radiosensitizer for esophageal cancer radiotherapy [88,211]. S3I-201 alkylates multiple Cys residues of STAT3 (Cys 108, 259, 367, 542 and 687), and is identified as a non-selective alkylating agent due to its leaving group O-tosyl [212]. Molecular modeling, using S3I-201 as the lead compound, led to S3I-1757, which could interact with Y705 in the binding site of the SH2 domain and decrease nuclear phosphotyrosine STAT3 levels [213]. To optimize S3I-201, Steven et al. synthesized a large set of S3I-201 analogs, among which SF-1-066 (known as S3I-201.1066, Figure 3l) and SF-1-121 showed impressive potency in whole-cell viability assays, and SF-1-066 also induced an antitumor response in breast tumor xenografts [214,215]. Further improvement of the parent compound SF-1-066 led to BP-5-087 (Figure 3m), a highly potent and selective STAT3 SH2 domain inhibitor, and when used in combination with imatinib, it could reverse TKI-resistant CML [39]. BP-1-102 (Figure 3n), identified as an analog of SF-1-066 via computer-aided design, also exhibited similar effects in breast cancer xenografts [216]. BP-1-102 is orally bioavailable and detectable at the micrograms levels in tumor tissues, which is enough to inhibit tumor growth. Even though micromolar concentrations of BP-1-102 in plasma were detected, the blood levels exceeded the IC50 (inhibitory concentration 50%, 6.8 ± 0.8 μM) [216]. To reduce the pharmacokinetic lability of BP-1-102, SH-4-54 was designed as a STAT3 SH2 domain inhibitor, which showed better therapeutic efficacy than BP-1-102, and exhibited blood–brain barrier permeability and controlled glioma tumor growth [217].
More recently, OPB-31121 (Otsuka pharmaceuticals Co. ltd.) and OPB-51602, potent small molecule inhibitors of STAT3, were found to strongly inhibit STAT3 phosphorylation and lead to effective antitumor responses [218,219,220,221]. They interact with the SH2 domain of STAT3 with high affinity and inhibit STAT3 Y705 and S727 phosphorylation. Computational docking and molecular dynamics simulations were used to examine the binding of OPB-31121 to STAT3, and OPB-31121 bound to S636 and V637 residues without overlapping with other STAT3 inhibitors [221,222]. OPB-51602 can directly interfere with mitochondrial STAT3 and induce the formation of STAT3 proteotoxic aggregates that are lethal to cancer cells [223,224]. These two inhibitors show the most potential among STAT3 SH2 domain inhibitors to date, based on preclinical studies that describe antitumor ability against various cancers. Phase I/II clinical trials for both OPB-31121 and OPB-51602 have been completed. These include a Phase I clinical trial for OPB-31121 in advanced cancer and solid tumor in Korea (NCT00657176, 18 April 2008) [225] and the USA (NCT00955812, 12 February 2013) [226], a Phase I/II trial in patients with progressive hepatocellular carcinoma (NCT01406574, 8 June 2015), and a Phase I OPB-51602 clinical trial for hematological malignancies in Japan (NCT01344876, 8 June 2015) [221] and for refractory solid tumors (NCT01184807, 26 March 2014) [220].

6.5. The Challenge of STAT3 Inhibitors

Aberrant STAT3 activation is related to cell malignant transformation, tumor proliferation, differentiation and anti-apoptosis [175]. Thus, STAT3 is considered a promising target in the development of anti-tumor drugs. Clearly, an accumulating number of STAT3 inhibitors have been discovered, as mentioned above. Most of them are mainly in the preclinical stage for screening drug activity, with few pharmacological and toxicological studies in animals and few compounds used for clinical trial.
The peptide (DTRYLEQLHQLY) and ST3-H2A2 (LDTRYLEQLHKLY) provide an additional strategy in the discovery of anti-STAT3 therapeutic agents as a novel anticancer therapy. However, the mechanisms of the peptides’ interaction with the N-terminal domain of STAT3 still need to be investigated. The ”undruggable” nature of a DBD has been proven druggable in the case of STAT3. Inhibitors targeting the DBD have been in a minor development stage and will face challenges. In recent years, silibinin has been shown to have unique characteristics whereby it binds directly with high affinity to both the SH2 domain and DBD of STAT3, making it a promising lead candidate for designing novel and effective STAT3 inhibitors [227]. Although few small-molecule inhibitors of the STAT3 linker domain have been discovered, STAT3 mutants with a mutation in the linker domain exhibit very weak transcriptional activity [228]. The linker domain between the DNA-binding domain and the SH2 domain offers a new strategy for developing small-molecule inhibitors of STAT3.
At present, the main focus is on STAT3 inhibitors that bind to the tyrosine phosphorylation site of the SH2 domain of STAT3. The general structure of the SH2 domain and the mode of phosphopeptide recognition are conserved. The site is rich in positively charged residues, which requires a high negative charge of small molecules [229]. Therefore, inhibitors for targeting the SH2 domain generally have large side effect and poor pharmacokinetic characteristics. Faced with this challenge, Zhang and co-workers recently identified allosteric sites and a modulator of the coiled-coil domain of STAT3 via AlloFinder. The team confirmed that K116 both inhibited STAT3 Tyr705 and promoted apoptosis in a dose-dependent manner [230].
In summary, small-molecule STAT3 inhibitors have antitumor activities and inhibit STAT3 phosphorylation but are yet to be approved for clinical cancer therapy. Inhibitors that target the SH2 domain of STAT3, such as STA-21, OPB-31121 and OPB-51602, have completed Phase I/II studies. These compounds are expected to progress in further clinical trials in the future and pave a new avenue for cancer therapy. The inhibitors targeting STAT3 may induce unexpected side-effects because of the repression of STAT3β, and regulators and drugs for the regulation of STAT3 alternative splicing remain to be investigated.

7. Conclusions and future perspectives

Chemoradiotherapy represents an important treatment for cancers. However, the intrinsic and acquired resistances of drug or radiation treatment limit its effectiveness in the clinic. Current evidence indicates that STAT3 is an oncogene, is constitutively activated in a variety of cancers, and regulates resistance to chemotherapy and radiotherapy. Signal transduction pathways influencing radiosensitivity mainly upregulate anti-apoptotic and cell cycle genes, induce EMT or modulate DNA repair [54]. Targeting the STAT3 pathway alone or in combination with other drugs can reverse resistance to chemotherapy or radiotherapy to significantly improve the effectiveness of therapy [4,35,36,48]. These findings show STAT3 to be a promising molecular target for the treatment of cancer. Inhibitors of different STAT3 domains have been identified via the screening of chemical libraries and computational docking. At present, few inhibitors have completed Phase I/II clinical trials. It is extremely urgent and important that we discover clinically useful inhibitors that target STAT3.
Conversely, constitutively active STAT3 inhibits cell proliferation and invasion in PTEN-deficient cancer cells [231,232]. The anti-tumor effects of STAT3 have also been reported for p19ARF-negative hepatocytes [233]. As mentioned above, STAT3β functions as a tumor suppressor and sensitizes cancer cells and ESCC patients to chemotherapy. The conflicting functions of STAT3 in cancers further indicate that STAT3 inhibitors must be used cautiously and could lead to enhanced tumor progression. To date, the binding sites of STAT3 inhibitors have docked to the crystal structure of STAT3β. These inhibitors probably bind to both STAT3α and STAT3β, and the selective binding affinities have not been characterized. Because of the unique structures of STAT3α and STAT3β, drugs targeting the C-terminal domain can be developed. We hope that promising drugs that selectively target STAT3α are forthcoming and that these drugs can reduce the possibility of side effects.
Interestingly, the ratio of STAT3α/STAT3β impacts the fate of a tumor. Splicing mediators, oligonucleotides and morpholinos can switch STAT3α to STAT3β in several cancer cells. Hopefully, in the next few years, several drugs that induce the expression of STAT3β by affecting alternative splicing regulators and increase chemo- and radiotherapeutic efficiency will be successfully applied in the treatment of cancer patients.

Author Contributions

P.-L.Y., L.-X.L., E.-M.L. and L.-Y.X. contributed to the literature review and manuscript preparation. All authors have read and agreed to the published version of the manuscript.

Funding

The authors of the current report were supported by the National Natural Science Foundation of China (No. 81772532).

Acknowledgments

De–Chen Lin (Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, California, USA) polished the manuscript at the beginning.

Conflicts of Interest

The authors declare no conflict of interest.

Abbreviations

AKTprotein kinase BJAKJanus kinase
ArgarginineKRASKirsten rat sarcoma 2 viral oncogene homolog
ATMataxia telangiectasia mutatedlncRNA-SRLRsorafenib resistance-associated lncRNA in RCC
BaxBcl2 associated XMEKmitogen-activated protein kinase kinase 7
Bcl-2B-cell lymphoma 2NKTLNK/T-cell lymphoma
Bcl-xLB-cell lymphoma-extra largeMLSmyxoid liposarcoma
BLACAT1bladder cancer associated transcript 1NTDamino-terminal domain
BLBurkitt lymphomaNSCLCnon-small-cell lung cancer
CCDcoiled-coil domainPCBP1poly-C binding protein-1
CKS1Bcyclin-dependent kinase regulatory subunit 1BPD-L1programmed death ligand 1
CMLchronic myeloid leukemiaPI3Kphosphonosinol-3 kinase
CRCcolorectal cancerPTENphosphatase and tensin homolog
CRTchemoradiotherapyRCCrenal cell carcinoma
CSCcancer stem cellROSreactive oxygen species
DAPdiarylidenyl-piperidoneRTradiation therapy
DBDDNA-binding domainRTKsreceptor tyrosine kinases
DCdendritic cellSer727serine 727
EGFepidermal growth factorSELLselectin L
EGFRepidermal growth factor receptorSH2Src homology 2
EMTepithelial-mesenchymal transitionSTATsignal transducer and activator of transcription
ESCCesophageal squamous-cell carcinomaTADtranscriptional activation domain
FBDDfragment-based drug designTGF-βtransforming growth factor β
FNFibronectinTKIstyrosine kinase inhibitors
FRETfluorescence resonance energy transferTwist1twist basic helix loop helix transcription factor 1
GASinterferon γ-activated sequenceValvaline
GBMGlioblastomaVEGFRvascular endothelial growth factor receptor
HER2human epidermal growth factor receptor 2Tyr705tyrosine 705
IL-6interleukin-6Ileisoleucine

References

  1. Hoek, J.; Bloemendal, K.M.; van der Velden, L.A.; van Diessen, J.N.; van Werkhoven, E.; Klop, W.M.; Tesselaar, M.E. Nephrotoxicity as a Dose-Limiting Factor in a High-Dose Cisplatin-Based Chemoradiotherapy Regimen for Head and Neck Carcinomas. Cancers 2016, 8, 21. [Google Scholar] [CrossRef] [Green Version]
  2. Shah, B.A.; Qureshi, M.M.; Logue, J.M.; Cooley, T.P.; Zaner, K.S.; Jalisi, S.; Truong, M.T. Assessing cumulative acute toxicity of chemoradiotherapy in head and neck cancer with or without induction chemotherapy. Am. J. Otolaryngol. 2017, 38, 456–461. [Google Scholar] [CrossRef]
  3. Hanahan, D.; Weinberg, R.A. Hallmarks of cancer: The next generation. Cell 2011, 144, 646–674. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  4. Spitzner, M.; Ebner, R.; Wolff, H.A.; Ghadimi, B.M.; Wienands, J.; Grade, M. STAT3: A Novel Molecular Mediator of Resistance to Chemoradiotherapy. Cancers 2014, 6, 1986–2011. [Google Scholar] [CrossRef]
  5. Sugase, T.; Takahashi, T.; Serada, S.; Fujimoto, M.; Hiramatsu, K.; Ohkawara, T.; Tanaka, K.; Miyazaki, Y.; Makino, T.; Kurokawa, Y.; et al. SOCS1 Gene Therapy Improves Radiosensitivity and Enhances Irradiation-Induced DNA Damage in Esophageal Squamous Cell Carcinoma. Cancer Res. 2017, 77, 6975–6986. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  6. Spitzner, M.; Roesler, B.; Bielfeld, C.; Emons, G.; Gaedcke, J.; Wolff, H.A.; Rave-Frank, M.; Kramer, F.; Beissbarth, T.; Kitz, J.; et al. STAT3 inhibition sensitizes colorectal cancer to chemoradiotherapy in vitro and in vivo. Int. J. Cancer 2014, 134, 997–1007. [Google Scholar] [CrossRef] [PubMed]
  7. Wu, X.; Tang, W.; Marquez, R.T.; Li, K.; Highfill, C.A.; He, F.; Lian, J.; Lin, J.; Fuchs, J.R.; Ji, M.; et al. Overcoming chemo/radio-resistance of pancreatic cancer by inhibiting STAT3 signaling. Oncotarget 2016, 7, 11708–11723. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  8. Shuai, K.; Stark, G.R.; Kerr, I.M.; Darnell, J.E., Jr. A single phosphotyrosine residue of Stat91 required for gene activation by interferon-gamma. Science 1993, 261, 1744–1746. [Google Scholar] [CrossRef]
  9. Loh, C.Y.; Arya, A.; Naema, A.F.; Wong, W.F.; Sethi, G.; Looi, C.Y. Signal Transducer and Activator of Transcription (STATs) Proteins in Cancer and Inflammation: Functions and Therapeutic Implication. Front. Oncol. 2019, 9, 48. [Google Scholar] [CrossRef] [Green Version]
  10. Xu, X.; Sun, Y.L.; Hoey, T. Cooperative DNA binding and sequence-selective recognition conferred by the STAT amino-terminal domain. Science 1996, 273, 794–797. [Google Scholar] [CrossRef]
  11. Shuai, K.; Horvath, C.M.; Huang, L.H.T.; Qureshi, S.A.; Cowburn, D.; Darnell, J.E. Interferon activation of the transcription factor Stat91 involves dimerization through SH2-phosphotyrosyl peptide interactions. Cell 1994, 76, 821–828. [Google Scholar] [CrossRef]
  12. Zhang, T.; Kee, W.H.; Seow, K.T.; Fung, W.; Cao, X. The coiled-coil domain of Stat3 is essential for its SH2 domain-mediated receptor binding and subsequent activation induced by epidermal growth factor and interleukin-6. Mol. Cell. Biol. 2000, 20, 7132–7139. [Google Scholar] [CrossRef] [Green Version]
  13. Wegenka, U.M.; Buschmann, J.; Lutticken, C.; Heinrich, P.C.; Horn, F. Acute-phase response factor, a nuclear factor binding to acute-phase response elements, is rapidly activated by interleukin-6 at the posttranslational level. Mol. Cell. Biol. 1993, 13, 276–288. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  14. Akira, S.; Nishio, Y.; Inoue, M.; Wang, X.J.; Wei, S.; Matsusaka, T.; Yoshida, K.; Sudo, T.; Naruto, M.; Kishimoto, T. 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, 63–71. [Google Scholar] [CrossRef]
  15. Zhong, Z.; Wen, Z.; Darnell, J.E., Jr. Stat3: A STAT family member activated by tyrosine phosphorylation in response to epidermal growth factor and interleukin-6. Science 1994, 264, 95–98. [Google Scholar] [CrossRef]
  16. Tian, S.S.; Lamb, P.; Seidel, H.M.; Stein, R.B.; Rosen, J. Rapid activation of the STAT3 transcription factor by granulocyte colony-stimulating factor. Blood 1994, 84, 1760–1764. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  17. Vaisse, C.; Halaas, J.L.; Horvath, C.M.; Darnell, J.E., Jr.; Stoffel, M.; Friedman, J.M. Leptin activation of Stat3 in the hypothalamus of wild-type and ob/ob mice but not db/db mice. Nat. Genet. 1996, 14, 95–97. [Google Scholar] [CrossRef]
  18. Lim, C.P.; Cao, X. Structure, function, and regulation of STAT proteins. Mol. Biosyst. 2006, 2, 536–550. [Google Scholar] [CrossRef]
  19. Zhang, H.X.; Yang, P.L.; Li, E.M.; Xu, L.Y. STAT3beta, a distinct isoform from STAT3. Int. J. Biochem. Cell Biol. 2019, 110, 130–139. [Google Scholar] [CrossRef]
  20. Turkson, J.; Ryan, D.; Kim, J.S.; Zhang, Y.; Chen, Z.; Haura, E.; Laudano, A.; Sebti, S.; Hamilton, A.D.; Jove, R. Phosphotyrosyl peptides block Stat3-mediated DNA binding activity, gene regulation, and cell transformation. J. Biol. Chem. 2001, 276, 45443–45455. [Google Scholar] [CrossRef] [Green Version]
  21. Ng, I.H.; Ng, D.C.; Jans, D.A.; Bogoyevitch, M.A. Selective STAT3-alpha or -beta expression reveals spliceform-specific phosphorylation kinetics, nuclear retention and distinct gene expression outcomes. Biochem. J. 2012, 447, 125–136. [Google Scholar] [CrossRef] [PubMed]
  22. Lieblein, J.C.; Ball, S.; Hutzen, B.; Sasser, A.K.; Lin, H.J.; Huang, T.H.; Hall, B.M.; Lin, J. STAT3 can be activated through paracrine signaling in breast epithelial cells. BMC Cancer 2008, 8, 302. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  23. Lin, J.; Jin, X.; Rothman, K.; Lin, H.J.; Tang, H.; Burke, W. Modulation of signal transducer and activator of transcription 3 activities by p53 tumor suppressor in breast cancer cells. Cancer Res. 2002, 62, 376–380. [Google Scholar]
  24. Li, L.; Tang, W.; Wu, X.; Karnak, D.; Meng, X.; Thompson, R.; Hao, X.; Li, Y.; Qiao, X.T.; Lin, J.; et al. HAb18G/CD147 promotes pSTAT3-mediated pancreatic cancer development via CD44s. Clin. Cancer Res. 2013, 19, 6703–6715. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  25. Corvinus, F.M.; Orth, C.; Moriggl, R.; Tsareva, S.A.; Wagner, S.; Pfitzner, E.B.; Baus, D.; Kaufmann, R.; Huber, L.A.; Zatloukal, K.; et al. Persistent STAT3 activation in colon cancer is associated with enhanced cell proliferation and tumor growth. Neoplasia 2005, 7, 545–555. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  26. Park, J.W.; Han, C.R.; Zhao, L.; Willingham, M.C.; Cheng, S.Y. Inhibition of STAT3 activity delays obesity-induced thyroid carcinogenesis in a mouse model. Endocr. Relat. Cancer 2016, 23, 53–63. [Google Scholar] [CrossRef] [Green Version]
  27. Niu, G.; Wright, K.L.; Huang, M.; Song, L.; Haura, E.; Turkson, J.; Zhang, S.; Wang, T.; Sinibaldi, D.; Coppola, D.; et al. Constitutive Stat3 activity up-regulates VEGF expression and tumor angiogenesis. Oncogene 2002, 21, 2000–2008. [Google Scholar] [CrossRef] [Green Version]
  28. Zhao, M.; Gao, F.H.; Wang, J.Y.; Liu, F.; Yuan, H.H.; Zhang, W.Y.; Jiang, B. JAK2/STAT3 signaling pathway activation mediates tumor angiogenesis by upregulation of VEGF and bFGF in non-small-cell lung cancer. Lung Cancer 2011, 73, 366–374. [Google Scholar] [CrossRef]
  29. Ho, P.L.; Lay, E.J.; Jian, W.; Parra, D.; Chan, K.S. Stat3 activation in urothelial stem cells leads to direct progression to invasive bladder cancer. Cancer Res. 2012, 72, 3135–3142. [Google Scholar] [CrossRef] [Green Version]
  30. Haura, E.B.; Zheng, Z.; Song, L.; Cantor, A.; Bepler, G. Activated epidermal growth factor receptor-Stat-3 signaling promotes tumor survival in vivo in non-small cell lung cancer. Clin. Cancer Res. 2005, 11, 8288–8294. [Google Scholar] [CrossRef] [Green Version]
  31. Takeda, K.; Noguchi, K.; Shi, W.; Tanaka, T.; Matsumoto, M.; Yoshida, N.; Kishimoto, T.; Akira, S. Targeted disruption of the mouse Stat3 gene leads to early embryonic lethality. Proc. Natl. Acad. Sci. USA 1997, 94, 3801–3804. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  32. Niwa, H.; Burdon, T.; Chambers, I.; Smith, A. Self-renewal of pluripotent embryonic stem cells is mediated via activation of STAT3. Genes Dev. 1998, 12, 2048–2060. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  33. Maritano, D.; Sugrue, M.L.; Tininini, S.; Dewilde, S.; Strobl, B.; Fu, X.; Murray-Tait, V.; Chiarle, R.; Poli, V. The STAT3 isoforms alpha and beta have unique and specific functions. Nat. Immunol. 2004, 5, 401–409. [Google Scholar] [CrossRef] [PubMed]
  34. Sun, C.; Bernards, R. Feedback and redundancy in receptor tyrosine kinase signaling: Relevance to cancer therapies. Trends Biochem. Sci. 2014, 39, 465–474. [Google Scholar] [CrossRef] [PubMed]
  35. Lee, H.J.; Zhuang, G.; Cao, Y.; Du, P.; Kim, H.J.; Settleman, J. Drug resistance via feedback activation of Stat3 in oncogene-addicted cancer cells. Cancer Cell 2014, 26, 207–221. [Google Scholar] [CrossRef] [Green Version]
  36. Li, G.; Zhao, L.; Li, W.; Fan, K.; Qian, W.; Hou, S.; Wang, H.; Dai, J.; Wei, H.; Guo, Y. Feedback activation of STAT3 mediates trastuzumab resistance via upregulation of MUC1 and MUC4 expression. Oncotarget 2014, 5, 8317–8329. [Google Scholar] [CrossRef] [Green Version]
  37. Yoon, Y.K.; Kim, H.P.; Han, S.W.; Oh, D.Y.; Im, S.A.; Bang, Y.J.; Kim, T.Y. KRAS mutant lung cancer cells are differentially responsive to MEK inhibitor due to AKT or STAT3 activation: Implication for combinatorial approach. Mol. Carcinog. 2010, 49, 353–362. [Google Scholar] [CrossRef]
  38. Cheon, S.K.; Kim, H.P.; Park, Y.L.; Jang, J.E.; Lim, Y.; Song, S.H.; Han, S.W.; Kim, T.Y. Macrophage migration inhibitory factor promotes resistance to MEK blockade in KRAS mutant colorectal cancer cells. Mol. Oncol. 2018, 12, 1398–1409. [Google Scholar] [CrossRef] [Green Version]
  39. Eiring, A.M.; Page, B.D.G.; Kraft, I.L.; Mason, C.C.; Vellore, N.A.; Resetca, D.; Zabriskie, M.S.; Zhang, T.Y.; Khorashad, J.S.; Engar, A.J.; et al. Combined STAT3 and BCR-ABL1 inhibition induces synthetic lethality in therapy-resistant chronic myeloid leukemia. Leukemia 2015, 29, 586–597. [Google Scholar] [CrossRef] [Green Version]
  40. Zhao, C.; Xiao, H.; Wu, X.; Li, C.; Liang, G.; Yang, S.; Lin, J. Rational combination of MEK inhibitor and the STAT3 pathway modulator for the therapy in K-Ras mutated pancreatic and colon cancer cells. Oncotarget 2015, 6, 14472–14487. [Google Scholar] [CrossRef]
  41. Van Schaeybroeck, S.; Kalimutho, M.; Dunne, P.D.; Carson, R.; Allen, W.; Jithesh, P.V.; Redmond, K.L.; Sasazuki, T.; Shirasawa, S.; Blayney, J.; et al. ADAM17-dependent c-MET-STAT3 signaling mediates resistance to MEK inhibitors in KRAS mutant colorectal cancer. Cell Rep. 2014, 7, 1940–1955. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  42. Le Naour, A.; Mevel, R.; Thibault, B.; Courtais, E.; Chantalat, E.; Delord, J.P.; Couderc, B.; Guillermet-Guibert, J.; Martinez, A. Effect of combined inhibition of p110 alpha PI3K isoform and STAT3 pathway in ovarian cancer platinum-based resistance. Oncotarget 2018, 9, 27220–27232. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  43. Tao, L.; Huang, G.; Wang, R.; Pan, Y.; He, Z.; Chu, X.; Song, H.; Chen, L. Cancer-associated fibroblasts treated with cisplatin facilitates chemoresistance of lung adenocarcinoma through IL-11/IL-11R/STAT3 signaling pathway. Sci. Rep. 2016, 6, 38408. [Google Scholar] [CrossRef] [PubMed]
  44. Ara, T.; Nakata, R.; Sheard, M.; Shimada, H.; Buettner, R.; Groshen, S.; Ji, L.; Yu, H.; Jove, R.; Seeger, R.; et al. Critical role of STAT3 in IL-6-mediated drug resistance in human neuroblastoma. Cancer Res. 2013, 73, 3852–3864. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  45. Mirzaei, A.; Mohammadi, S.; Ghaffari, S.H.; Nikbakht, M.; Bashash, D.; Alimoghaddam, K.; Ghavamzadeh, A. Osteopontin b and c isoforms: Molecular Candidates Associated with Leukemic Stem Cell Chemoresistance in Acute Myeloid Leukemia. Asian Pac. J. Cancer Prev. 2017, 18, 1707–1715. [Google Scholar] [CrossRef] [PubMed]
  46. Sredni, B.; Weil, M.; Khomenok, G.; Lebenthal, I.; Teitz, S.; Mardor, Y.; Ram, Z.; Orenstein, A.; Kershenovich, A.; Michowiz, S.; et al. Ammonium trichloro(dioxoethylene-o,o’)tellurate (AS101) sensitizes tumors to chemotherapy by inhibiting the tumor interleukin 10 autocrine loop. Cancer Res. 2004, 64, 1843–1852. [Google Scholar] [CrossRef] [Green Version]
  47. Prokoph, N.; Probst, N.A.; Lee, L.C.; Monahan, J.M.; Matthews, J.D.; Liang, H.C.; Bahnsen, K.; Montes-Mojarro, I.A.; Karaca Atabay, E.; Sharma, G.G.; et al. IL10RA Modulates Crizotinib Sensitivity in NPM1-ALK-positive Anaplastic Large Cell Lymphoma. Blood 2020. [Google Scholar] [CrossRef]
  48. Yang, Z.; Guo, L.; Liu, D.; Sun, L.; Chen, H.; Deng, Q.; Liu, Y.; Yu, M.; Ma, Y.; Guo, N.; et al. Acquisition of resistance to trastuzumab in gastric cancer cells is associated with activation of IL-6/STAT3/Jagged-1/Notch positive feedback loop. Oncotarget 2015, 6, 5072–5087. [Google Scholar] [CrossRef] [Green Version]
  49. Rokavec, M.; Oner, M.G.; Li, H.; Jackstadt, R.; Jiang, L.; Lodygin, D.; Kaller, M.; Horst, D.; Ziegler, P.K.; Schwitalla, S.; et al. IL-6R/STAT3/miR-34a feedback loop promotes EMT-mediated colorectal cancer invasion and metastasis. J. Clin. Investig. 2014, 124, 1853–1867. [Google Scholar] [CrossRef] [Green Version]
  50. Zhu, X.; Shen, H.; Yin, X.; Long, L.; Chen, X.; Feng, F.; Liu, Y.; Zhao, P.; Xu, Y.; Li, M.; et al. IL-6R/STAT3/miR-204 feedback loop contributes to cisplatin resistance of epithelial ovarian cancer cells. Oncotarget 2017, 8, 39154–39166. [Google Scholar] [CrossRef] [Green Version]
  51. Zeng, R.; Tang, Y.; Zhou, H.; Liu, Y.; Huang, J.; Li, L.; Liu, W.; Feng, Y.; Zhou, Y.; Chen, T.; et al. STAT3 mediates multidrug resistance of Burkitt lymphoma cells by promoting antioxidant feedback. Biochem. Biophys. Res. Commun. 2017, 488, 182–188. [Google Scholar] [CrossRef] [PubMed]
  52. Roentgen, W.C. [On a new kind of ray (first report)]. Munch. Med. Wochenschr. (1950) 1959, 101, 1237–1239. [Google Scholar]
  53. Thariat, J.; Hannoun-Levi, J.M.; Sun Myint, A.; Vuong, T.; Gerard, J.P. Past, present, and future of radiotherapy for the benefit of patients. Nat. Rev. Clin. Oncol. 2013, 10, 52–60. [Google Scholar] [CrossRef]
  54. Begg, A.C.; Stewart, F.A.; Vens, C. Strategies to improve radiotherapy with targeted drugs. Nat. Rev. Cancer 2011, 11, 239–253. [Google Scholar] [CrossRef] [PubMed]
  55. Kim, B.M.; Hong, Y.; Lee, S.; Liu, P.; Lim, J.H.; Lee, Y.H.; Lee, T.H.; Chang, K.T.; Hong, Y. Therapeutic Implications for Overcoming Radiation Resistance in Cancer Therapy. Int. J. Mol. Sci. 2015, 16, 26880–26913. [Google Scholar] [CrossRef] [Green Version]
  56. Otero, D.C.; Poli, V.; David, M.; Rickert, R.C. Cutting edge: Inherent and acquired resistance to radiation-induced apoptosis in B cells: A pivotal role for STAT3. J. Immunol. 2006, 177, 6593–6597. [Google Scholar] [CrossRef] [Green Version]
  57. Li, X.; Wang, H.; Lu, X.; Di, B. Silencing STAT3 with short hairpin RNA enhances radiosensitivity of human laryngeal squamous cell carcinoma xenografts in vivo. Exp. Ther. Med. 2010, 1, 947–953. [Google Scholar] [CrossRef] [Green Version]
  58. Wu, C.T.; Chen, M.F.; Chen, W.C.; Hsieh, C.C. The role of IL-6 in the radiation response of prostate cancer. Radiat. Oncol. 2013, 8, 159. [Google Scholar] [CrossRef] [Green Version]
  59. You, S.; Li, R.; Park, D.; Xie, M.; Sica, G.L.; Cao, Y.; Xiao, Z.Q.; Deng, X. Disruption of STAT3 by niclosamide reverses radioresistance of human lung cancer. Mol. Cancer Ther. 2014, 13, 606–616. [Google Scholar] [CrossRef] [Green Version]
  60. He, N.; Kong, Y.; Lei, X.; Liu, Y.; Wang, J.; Xu, C.; Wang, Y.; Du, L.; Ji, K.; Wang, Q.; et al. MSCs inhibit tumor progression and enhance radiosensitivity of breast cancer cells by down-regulating Stat3 signaling pathway. Cell Death Dis. 2018, 9, 1026. [Google Scholar] [CrossRef] [Green Version]
  61. Gao, L.; Li, F.S.; Chen, X.H.; Liu, Q.W.; Feng, J.B.; Liu, Q.J.; Su, X. Radiation induces phosphorylation of STAT3 in a dose- and time-dependent manner. Asian Pac. J. Cancer Prev. 2014, 15, 6161–6164. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  62. Chou, C.H.; Chen, P.J.; Jeng, Y.M.; Cheng, A.L.; Huang, L.R.; Cheng, J.C. Synergistic effect of radiation and interleukin-6 on hepatitis B virus reactivation in liver through STAT3 signaling pathway. Int. J. Radiat. Oncol. Biol. Phys. 2009, 75, 1545–1552. [Google Scholar] [CrossRef] [PubMed]
  63. Yi, F.T.; Lu, Q.P. Mucin 1 promotes radioresistance in hepatocellular carcinoma cells through activation of JAK2/STAT3 signaling. Oncol. Lett. 2017, 14, 7571–7576. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  64. Shi, Y.; Guryanova, O.A.; Zhou, W.; Liu, C.; Huang, Z.; Fang, X.; Wang, X.; Chen, C.; Wu, Q.; He, Z.; et al. Ibrutinib inactivates BMX-STAT3 in glioma stem cells to impair malignant growth and radioresistance. Sci. Transl. Med. 2018, 10. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  65. Arnold, K.M.; Opdenaker, L.M.; Flynn, N.J.; Appeah, D.K.; Sims-Mourtada, J. Radiation induces an inflammatory response that results in STAT3-dependent changes in cellular plasticity and radioresistance of breast cancer stem-like cells. Int. J. Radiat. Biol. 2020, 96, 434–447. [Google Scholar] [CrossRef]
  66. Duru, N.; Fan, M.; Candas, D.; Menaa, C.; Liu, H.C.; Nantajit, D.; Wen, Y.; Xiao, K.; Eldridge, A.; Chromy, B.A.; et al. HER2-associated radioresistance of breast cancer stem cells isolated from HER2-negative breast cancer cells. Clin. Cancer Res. 2012, 18, 6634–6647. [Google Scholar] [CrossRef] [Green Version]
  67. Kim, J.S.; Kim, H.A.; Seong, M.K.; Seol, H.; Oh, J.S.; Kim, E.K.; Chang, J.W.; Hwang, S.G.; Noh, W.C. STAT3-survivin signaling mediates a poor response to radiotherapy in HER2-positive breast cancers. Oncotarget 2016, 7, 7055–7065. [Google Scholar] [CrossRef]
  68. Lu, L.; Dong, J.; Wang, L.; Xia, Q.; Zhang, D.; Kim, H.; Yin, T.; Fan, S.; Shen, Q. 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, 5292–5304. [Google Scholar] [CrossRef]
  69. Oweida, A.J.; Darragh, L.; Phan, A.; Binder, D.; Bhatia, S.; Mueller, A.; Court, B.V.; Milner, D.; Raben, D.; Woessner, R.; et al. STAT3 Modulation of Regulatory T Cells in Response to Radiation Therapy in Head and Neck Cancer. J. Natl. Cancer Inst. 2019, 111, 1339–1349. [Google Scholar] [CrossRef]
  70. Karras, J.G.; Wang, Z.; Huo, L.; Howard, R.G.; Frank, D.A.; Rothstein, T.L. 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, 1035–1042. [Google Scholar] [CrossRef]
  71. Ouedraogo, Z.G.; Muller-Barthelemy, M.; Kemeny, J.L.; Dedieu, V.; Biau, J.; Khalil, T.; Raoelfils, L.I.; Granzotto, A.; Pereira, B.; Beaudoin, C.; et al. STAT3 Serine 727 Phosphorylation: A Relevant Target to Radiosensitize Human Glioblastoma. Brain Pathol. 2016, 26, 18–30. [Google Scholar] [CrossRef] [PubMed]
  72. Masliantsev, K.; Pinel, B.; Balbous, A.; Guichet, P.O.; Tachon, G.; Milin, S.; Godet, J.; Duchesne, M.; Berger, A.; Petropoulos, C.; et al. Impact of STAT3 phosphorylation in glioblastoma stem cells radiosensitization and patient outcome. Oncotarget 2018, 9, 3968–3979. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  73. Purnell, P.R.; Mack, P.C.; Tepper, C.G.; Evans, C.P.; Green, T.P.; Gumerlock, P.H.; Lara, P.N.; Gandara, D.R.; Kung, H.J.; Gautschi, O. The Src inhibitor AZD0530 blocks invasion and may act as a radiosensitizer in lung cancer cells. J. Thorac. Oncol. 2009, 4, 448–454. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  74. Yin, Z.J.; Jin, F.G.; Liu, T.G.; Fu, E.Q.; Xie, Y.H.; Sun, R.L. Overexpression of STAT3 potentiates growth, survival, and radioresistance of non-small-cell lung cancer (NSCLC) cells. J. Surg. Res. 2011, 171, 675–683. [Google Scholar] [CrossRef]
  75. Sun, Y.; Moretti, L.; Giacalone, N.J.; Schleicher, S.; Speirs, C.K.; Carbone, D.P.; Lu, B. Inhibition of JAK2 signaling by TG101209 enhances radiotherapy in lung cancer models. J. Thorac. Oncol. 2011, 6, 699–706. [Google Scholar] [CrossRef] [Green Version]
  76. Chen, Y.W.; Chen, K.H.; Huang, P.I.; Chen, Y.C.; Chiou, G.Y.; Lo, W.L.; Tseng, L.M.; Hsu, H.S.; Chang, K.W.; Chiou, S.H. Cucurbitacin I suppressed stem-like property and enhanced radiation-induced apoptosis in head and neck squamous carcinoma--derived CD44(+)ALDH1(+) cells. Mol. Cancer Ther. 2010, 9, 2879–2892. [Google Scholar] [CrossRef] [Green Version]
  77. Kang, Y.; Park, M.A.; Heo, S.W.; Park, S.Y.; Kang, K.W.; Park, P.H.; Kim, J.A. The radio-sensitizing effect of xanthohumol is mediated by STAT3 and EGFR suppression in doxorubicin-resistant MCF-7 human breast cancer cells. Biochim. Biophys. Acta 2013, 1830, 2638–2648. [Google Scholar] [CrossRef]
  78. Park, S.Y.; Lee, C.J.; Choi, J.H.; Kim, J.H.; Kim, J.W.; Kim, J.Y.; Nam, J.S. The JAK2/STAT3/CCND2 Axis promotes colorectal Cancer stem cell persistence and radioresistance. J. Exp. Clin. Cancer Res. 2019, 38, 399. [Google Scholar] [CrossRef] [Green Version]
  79. Urick, M.E.; Chung, E.J.; Shield, W.P., 3rd; Gerber, N.; White, A.; Sowers, A.; Thetford, A.; Camphausen, K.; Mitchell, J.; Citrin, D.E. Enhancement of 5-fluorouracil-induced in vitro and in vivo radiosensitization with MEK inhibition. Clin. Cancer Res. 2011, 17, 5038–5047. [Google Scholar] [CrossRef] [Green Version]
  80. Zang, C.; Liu, X.; Li, B.; He, Y.; Jing, S.; He, Y.; Wu, W.; Zhang, B.; Ma, S.; Dai, W.; et al. IL-6/STAT3/TWIST inhibition reverses ionizing radiation-induced EMT and radioresistance in esophageal squamous carcinoma. Oncotarget 2017, 8, 11228–11238. [Google Scholar] [CrossRef] [Green Version]
  81. Yin, Z.; Zhang, Y.; Li, Y.; Lv, T.; Liu, J.; Wang, X. Prognostic significance of STAT3 expression and its correlation with chemoresistance of non-small cell lung cancer cells. Acta Histochem. 2012, 114, 151–158. [Google Scholar] [CrossRef] [PubMed]
  82. Lin, J.C.; Tsai, J.T.; Chao, T.Y.; Ma, H.I.; Liu, W.H. The STAT3/Slug Axis Enhances Radiation-Induced Tumor Invasion and Cancer Stem-like Properties in Radioresistant Glioblastoma. Cancers 2018, 10, 512. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  83. Lee, J.H.; Choi, S.I.; Kim, R.K.; Cho, E.W.; Kim, I.G. Tescalcin/c-Src/IGF1Rbeta-mediated STAT3 activation enhances cancer stemness and radioresistant properties through ALDH1. Sci. Rep. 2018, 8, 10711. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  84. Chiang, P.K.; Tsai, W.K.; Chen, M.; Lin, W.R.; Chow, Y.C.; Lee, C.C.; Hsu, J.M.; Chen, Y.J. Zerumbone Regulates DNA Repair Responding to Ionizing Radiation and Enhances Radiosensitivity of Human Prostatic Cancer Cells. Integr. Cancer Ther. 2018, 17, 292–298. [Google Scholar] [CrossRef] [Green Version]
  85. Yuan, S.S.; Hou, M.F.; Hsieh, Y.C.; Huang, C.Y.; Lee, Y.C.; Chen, Y.J.; Lo, S. Role of MRE11 in cell proliferation, tumor invasion, and DNA repair in breast cancer. J. Natl. Cancer Inst. 2012, 104, 1485–1502. [Google Scholar] [CrossRef] [Green Version]
  86. Chen, X.; Chen, F.; Ren, Y.; Weng, G.; Xu, L.; Xue, X.; Keng, P.C.; Lee, S.O.; Chen, Y. IL-6 signaling contributes to radioresistance of prostate cancer through key DNA repair-associated molecules ATM, ATR, and BRCA 1/2. J. Cancer Res. Clin. Oncol. 2019, 145, 1471–1484. [Google Scholar] [CrossRef]
  87. Zhang, Q.; Zhang, C.; He, J.; Guo, Q.; Hu, D.; Yang, X.; Wang, J.; Kang, Y.; She, R.; Wang, Z.; et al. STAT3 inhibitor stattic enhances radiosensitivity in esophageal squamous cell carcinoma. Tumour Biol. 2015, 36, 2135–2142. [Google Scholar] [CrossRef]
  88. Zhang, C.; Yang, X.; Zhang, Q.; Guo, Q.; He, J.; Qin, Q.; Zhu, H.; Liu, J.; Zhan, L.; Lu, J.; et al. STAT3 inhibitor NSC74859 radiosensitizes esophageal cancer via the downregulation of HIF-1alpha. Tumour Biol. 2014, 35, 9793–9799. [Google Scholar] [CrossRef]
  89. Xie, B.; Zhang, L.; Hu, W.; Fan, M.; Jiang, N.; Duan, Y.; Jing, D.; Xiao, W.; Fragoso, R.C.; Lam, K.S.; et al. Dual blockage of STAT3 and ERK1/2 eliminates radioresistant GBM cells. Redox Biol. 2019, 24, 101189. [Google Scholar] [CrossRef]
  90. Adachi, M.; Cui, C.; Dodge, C.T.; Bhayani, M.K.; Lai, S.Y. Targeting STAT3 inhibits growth and enhances radiosensitivity in head and neck squamous cell carcinoma. Oral Oncol. 2012, 48, 1220–1226. [Google Scholar] [CrossRef] [Green Version]
  91. Hsu, H.W.; Gridley, D.S.; Kim, P.D.; Hu, S.; de Necochea-Campion, R.; Ferris, R.L.; Chen, C.S.; Mirshahidi, S. Linifanib (ABT-869) enhances radiosensitivity of head and neck squamous cell carcinoma cells. Oral Oncol. 2013, 49, 591–597. [Google Scholar] [CrossRef] [PubMed]
  92. Huang, C.Y.; Lin, C.S.; Tai, W.T.; Hsieh, C.Y.; Shiau, C.W.; Cheng, A.L.; Chen, K.F. Sorafenib enhances radiation-induced apoptosis in hepatocellular carcinoma by inhibiting STAT3. Int. J. Radiat. Oncol. Biol. Phys. 2013, 86, 456–462. [Google Scholar] [CrossRef] [PubMed]
  93. Saitoh, M.; Endo, K.; Furuya, S.; Minami, M.; Fukasawa, A.; Imamura, T.; Miyazawa, K. STAT3 integrates cooperative Ras and TGF-beta signals that induce Snail expression. Oncogene 2016, 35, 1049–1057. [Google Scholar] [CrossRef] [PubMed]
  94. Gu, L.; Chiang, K.Y.; Zhu, N.; Findley, H.W.; Zhou, M. Contribution of STAT3 to the activation of survivin by GM-CSF in CD34+ cell lines. Exp. Hematol. 2007, 35, 957–966. [Google Scholar] [CrossRef] [PubMed]
  95. Leslie, K.; Lang, C.; Devgan, G.; Azare, J.; Berishaj, M.; Gerald, W.; Kim, Y.B.; Paz, K.; Darnell, J.E.; Albanese, C.; et al. Cyclin D1 is transcriptionally regulated by and required for transformation by activated signal transducer and activator of transcription 3. Cancer Res. 2006, 66, 2544–2552. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  96. Zhang, H.F.; Chen, Y.; Wu, C.; Wu, Z.Y.; Tweardy, D.J.; Alshareef, A.; Liao, L.D.; Xue, Y.J.; Wu, J.Y.; Chen, B.; et al. The Opposing Function of STAT3 as an Oncoprotein and Tumor Suppressor Is Dictated by the Expression Status of STAT3beta in Esophageal Squamous Cell Carcinoma. Clin. Cancer Res. 2016, 22, 691–703. [Google Scholar] [CrossRef] [Green Version]
  97. Stephanou, A.; Brar, B.K.; Knight, R.A.; Latchman, D.S. Opposing actions of STAT-1 and STAT-3 on the Bcl-2 and Bcl-x promoters. Cell Death Differ. 2000, 7, 329–330. [Google Scholar] [CrossRef] [Green Version]
  98. Rahaman, S.O.; Vogelbaum, M.A.; Haque, S.J. Aberrant Stat3 signaling by interleukin-4 in malignant glioma cells: Involvement of IL-13Ralpha2. Cancer Res. 2005, 65, 2956–2963. [Google Scholar] [CrossRef] [Green Version]
  99. Isomoto, H.; Kobayashi, S.; Werneburg, N.W.; Bronk, S.F.; Guicciardi, M.E.; Frank, D.A.; Gores, G.J. Interleukin 6 upregulates myeloid cell leukemia-1 expression through a STAT3 pathway in cholangiocarcinoma cells. Hepatology 2005, 42, 1329–1338. [Google Scholar] [CrossRef]
  100. Yi, E.H.; Lee, C.S.; Lee, J.K.; Lee, Y.J.; Shin, M.K.; Cho, C.H.; Kang, K.W.; Lee, J.W.; Han, W.; Noh, D.Y.; et al. STAT3-RANTES autocrine signaling is essential for tamoxifen resistance in human breast cancer cells. Mol. Cancer Res. 2013, 11, 31–42. [Google Scholar] [CrossRef] [Green Version]
  101. Fujita, Y.; Yagishita, S.; Hagiwara, K.; Yoshioka, Y.; Kosaka, N.; Takeshita, F.; Fujiwara, T.; Tsuta, K.; Nokihara, H.; Tamura, T.; et al. The clinical relevance of the miR-197/CKS1B/STAT3-mediated PD-L1 network in chemoresistant non-small-cell lung cancer. Mol. Ther. 2015, 23, 717–727. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  102. Han, Z.; Feng, J.; Hong, Z.; Chen, L.; Li, W.; Liao, S.; Wang, X.; Ji, T.; Wang, S.; Ma, D.; et al. Silencing of the STAT3 signaling pathway reverses the inherent and induced chemoresistance of human ovarian cancer cells. Biochem. Biophys. Res. Commun. 2013, 435, 188–194. [Google Scholar] [CrossRef] [PubMed]
  103. Selvendiran, K.; Bratasz, A.; Tong, L.; Ignarro, L.J.; Kuppusamy, P. NCX-4016, a nitro-derivative of aspirin, inhibits EGFR and STAT3 signaling and modulates Bcl-2 proteins in cisplatin-resistant human ovarian cancer cells and xenografts. Cell Cycle 2008, 7, 81–88. [Google Scholar] [CrossRef] [PubMed]
  104. Yan, H.; Guo, B.Y.; Zhang, S. Cancer-associated fibroblasts attenuate Cisplatin-induced apoptosis in ovarian cancer cells by promoting STAT3 signaling. Biochem. Biophys. Res. Commun. 2016, 470, 947–954. [Google Scholar] [CrossRef] [PubMed]
  105. Hira, S.K.; Mondal, I.; Bhattacharya, D.; Gupta, K.K.; Manna, P.P. Downregulation of STAT3 phosphorylation enhances tumoricidal effect of IL-15-activated dendritic cell against doxorubicin-resistant lymphoma and leukemia via TNF-alpha. Int. J. Biochem. Cell Biol. 2015, 67, 1–13. [Google Scholar] [CrossRef]
  106. Meng, Y.; Tang, W.; Dai, Y.; Wu, X.; Liu, M.; Ji, Q.; Ji, M.; Pienta, K.; Lawrence, T.; Xu, L. Natural BH3 mimetic (-)-gossypol chemosensitizes human prostate cancer via Bcl-xL inhibition accompanied by increase of Puma and Noxa. Mol. Cancer Ther. 2008, 7, 2192–2202. [Google Scholar] [CrossRef] [Green Version]
  107. Kiprianova, I.; Remy, J.; Milosch, N.; Mohrenz, I.V.; Seifert, V.; Aigner, A.; Kogel, D. Sorafenib Sensitizes Glioma Cells to the BH3 Mimetic ABT-737 by Targeting MCL1 in a STAT3-Dependent Manner. Neoplasia 2015, 17, 564–573. [Google Scholar] [CrossRef] [Green Version]
  108. Lian, J.; Ni, Z.; Dai, X.; Su, C.; Smith, A.R.; Xu, L.; He, F. Sorafenib sensitizes (-)-gossypol-induced growth suppression in androgen-independent prostate cancer cells via Mcl-1 inhibition and Bak activation. Mol. Cancer Ther. 2012, 11, 416–426. [Google Scholar] [CrossRef] [Green Version]
  109. Rosser, C.J.; Reyes, A.O.; Vakar-Lopez, F.; Levy, L.B.; Kuban, D.A.; Hoover, D.C.; Lee, A.K.; Pisters, L.L. Bcl-2 is significantly overexpressed in localized radio-recurrent prostate carcinoma, compared with localized radio-naive prostate carcinoma. Inter. J. Radiat. Oncol. Biol. Phys. 2003, 56, 1–6. [Google Scholar] [CrossRef]
  110. Kong, Z.; Xie, D.; Boike, T.; Raghavan, P.; Burma, S.; Chen, D.J.; Habib, A.A.; Chakraborty, A.; Hsieh, J.T.; Saha, D. Downregulation of human DAB2IP gene expression in prostate cancer cells results in resistance to ionizing radiation. Cancer Res. 2010, 70, 2829–2839. [Google Scholar] [CrossRef] [Green Version]
  111. Van Staalduinen, J.; Baker, D.; Ten Dijke, P.; van Dam, H. Epithelial-mesenchymal-transition-inducing transcription factors: New targets for tackling chemoresistance in cancer? Oncogene 2018, 37, 6195–6211. [Google Scholar] [CrossRef] [PubMed]
  112. Thiery, J.P.; Acloque, H.; Huang, R.Y.; Nieto, M.A. Epithelial-mesenchymal transitions in development and disease. Cell 2009, 139, 871–890. [Google Scholar] [CrossRef] [PubMed]
  113. Chong, K.Y.; Kang, M.; Garofalo, F.; Ueno, D.; Liang, H.; Cady, S.; Madarikan, O.; Pitruzzello, N.; Tsai, C.H.; Hartwich, T.M.P.; et al. Inhibition of Heat Shock Protein 90 suppresses TWIST1 Transcription. Mol. Pharmacol. 2019, 96, 168–179. [Google Scholar] [CrossRef] [PubMed]
  114. Liang, F.; Ren, C.; Wang, J.; Wang, S.; Yang, L.; Han, X.; Chen, Y.; Tong, G.; Yang, G. The crosstalk between STAT3 and p53/RAS signaling controls cancer cell metastasis and cisplatin resistance via the Slug/MAPK/PI3K/AKT-mediated regulation of EMT and autophagy. Oncogenesis 2019, 8, 59. [Google Scholar] [CrossRef] [Green Version]
  115. Su, Y.K.; Bamodu, O.A.; Tzeng, Y.M.; Hsiao, M.; Yeh, C.T.; Lin, C.M. Ovatodiolide inhibits the oncogenicity and cancer stem cell-like phenotype of glioblastoma cells, as well as potentiate the anticancer effect of temozolomide. Phytomedicine 2019, 61, 152840. [Google Scholar] [CrossRef]
  116. Liu, S.C.; Huang, C.M.; Bamodu, O.A.; Lin, C.S.; Liu, B.L.; Tzeng, Y.M.; Tsai, J.T.; Lee, W.H.; Chen, T.M. Ovatodiolide suppresses nasopharyngeal cancer by targeting stem cell-like population, inducing apoptosis, inhibiting EMT and dysregulating JAK/STAT signaling pathway. Phytomedicine 2019, 56, 269–278. [Google Scholar] [CrossRef]
  117. Liang, H.; Chen, G.; Li, J.; Yang, F. Snail expression contributes to temozolomide resistance in glioblastoma. Am. J. Transl. Res. 2019, 11, 4277–4289. [Google Scholar]
  118. Wang, H.; Li, J.M.; Wei, W.; Yang, R.; Chen, D.; Ma, X.D.; Jiang, G.M.; Wang, B.L. Regulation of ATP-binding cassette subfamily B member 1 by Snail contributes to chemoresistance in colorectal cancer. Cancer Sci. 2020, 111, 84–97. [Google Scholar] [CrossRef]
  119. Park, J.H.; Kim, Y.H.; Park, E.H.; Lee, S.J.; Kim, H.; Kim, A.; Lee, S.B.; Shim, S.; Jang, H.; Myung, J.K.; et al. Effects of metformin and phenformin on apoptosis and epithelial-mesenchymal transition in chemoresistant rectal cancer. Cancer Sci. 2019, 110, 2834–2845. [Google Scholar] [CrossRef]
  120. Yang, J.; Xing, H.; Hong, L.; Lu, D.H.; Li, B.S.; Tang, J.M.; Min, J. STAT3-mediated Twist1 upregulation contributes to epithelial-mesenehymal transition in cisplatin resistant ovarian cancer. Int. J. Clin. Exp. Med. 2018, 11, 6749–6757. [Google Scholar]
  121. Wu, K.; Wang, B.; Chen, Y.; Zhou, J.; Huang, J.; Hui, K.; Zeng, J.; Zhu, J.; Zhang, K.; Li, L.; et al. DAB2IP regulates the chemoresistance to pirarubicin and tumor recurrence of non-muscle invasive bladder cancer through STAT3/Twist1/P-glycoprotein signaling. Cell Signal. 2015, 27, 2515–2523. [Google Scholar] [CrossRef] [PubMed]
  122. Mita, A.C.; Mita, M.M.; Nawrocki, S.T.; Giles, F.J. Survivin: Key regulator of mitosis and apoptosis and novel target for cancer therapeutics. Clin. Cancer Res. 2008, 14, 5000–5005. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  123. Pandey, A.; Vishnoi, K.; Mahata, S.; Tripathi, S.C.; Misra, S.P.; Misra, V.; Mehrotra, R.; Dwivedi, M.; Bharti, A.C. Berberine and Curcumin Target Survivin and STAT3 in Gastric Cancer Cells and Synergize Actions of Standard Chemotherapeutic 5-Fluorouracil. Nutr. Cancer 2015, 67, 1293–1304. [Google Scholar] [CrossRef] [PubMed]
  124. Wang, C.; Yang, M.; Zhao, J.; Li, X.; Xiao, X.; Zhang, Y.; Jin, X.; Liao, M. Bile salt (glycochenodeoxycholate acid) induces cell survival and chemoresistance in hepatocellular carcinoma. J. Cell. Physiol. 2019, 234, 10899–10906. [Google Scholar] [CrossRef]
  125. Hu, W.; Jin, P.; Liu, W. Periostin Contributes to Cisplatin Resistance in Human Non-Small Cell Lung Cancer A549 Cells via Activation of Stat3 and Akt and Upregulation of Survivin. Cell. Physiol. Biochem. 2016, 38, 1199–1208. [Google Scholar] [CrossRef] [PubMed]
  126. Zhao, G.; Wang, Q.; Wu, Z.; Tian, X.; Yan, H.; Wang, B.; Dong, P.; Watari, H.; Pfeffer, L.M.; Guo, Y.; et al. Ovarian Primary and Metastatic Tumors Suppressed by Survivin Knockout or a Novel Survivin Inhibitor. Mol. Cancer Ther. 2019, 18, 2233–2245. [Google Scholar] [CrossRef] [Green Version]
  127. Lau, C.K.; Yang, Z.F.; Lam, S.P.; Lam, C.T.; Ngai, P.; Tam, K.H.; Poon, R.T.; Fan, S.T. Inhibition of Stat3 activity by YC-1 enhances chemo-sensitivity in hepatocellular carcinoma. Cancer Biol. Ther. 2007, 6, 1900–1907. [Google Scholar] [CrossRef] [Green Version]
  128. Biliran, H., Jr.; Wang, Y.; Banerjee, S.; Xu, H.; Heng, H.; Thakur, A.; Bollig, A.; Sarkar, F.H.; Liao, J.D. Overexpression of cyclin D1 promotes tumor cell growth and confers resistance to cisplatin-mediated apoptosis in an elastase-myc transgene-expressing pancreatic tumor cell line. Clin. Cancer Res. 2005, 11, 6075–6086. [Google Scholar] [CrossRef] [Green Version]
  129. Dong, X.F.; Liu, T.Q.; Zhi, X.T.; Zou, J.; Zhong, J.T.; Li, T.; Mo, X.L.; Zhou, W.; Guo, W.W.; Liu, X.; et al. COX-2/PGE2 Axis Regulates HIF2alpha Activity to Promote Hepatocellular Carcinoma Hypoxic Response and Reduce the Sensitivity of Sorafenib Treatment. Clin. Cancer Res. 2018, 24, 3204–3216. [Google Scholar] [CrossRef] [Green Version]
  130. Shimura, T. Targeting the AKT/cyclin D1 pathway to overcome intrinsic and acquired radioresistance of tumors for effective radiotherapy. Int. J. Radiat. Biol. 2017, 93, 381–385. [Google Scholar] [CrossRef]
  131. Li, H.; Lu, H.; Lv, M.; Wang, Q.; Sun, Y. Parthenolide facilitates apoptosis and reverses drug-resistance of human gastric carcinoma cells by inhibiting the STAT3 signaling pathway. Oncol. Lett. 2018, 15, 3572–3579. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  132. Fu, Z.; Ma, K.; Dong, B.; Zhao, C.; Che, C.; Dong, C.; Zhang, R.; Wang, H.; Wang, X.; Liang, R. The synergistic antitumor effect of Huaier combined with 5-Florouracil in human cholangiocarcinoma cells. BMC Complement. Altern. Med. 2019, 19, 203. [Google Scholar] [CrossRef] [PubMed]
  133. Lei, T.; Zhou, S.; Meng, Q.; Zhang, M. STAT3 signaling pathway in drug-resistant bladder cancer cell line. J. Biol. Regul. Homeost. Agents 2019, 33, 1347–1357. [Google Scholar] [CrossRef] [PubMed]
  134. Tai, W.T.; Cheng, A.L.; Shiau, C.W.; Liu, C.Y.; Ko, C.H.; Lin, M.W.; Chen, P.J.; Chen, K.F. Dovitinib induces apoptosis and overcomes sorafenib resistance in hepatocellular carcinoma through SHP-1-mediated inhibition of STAT3. Mol. Cancer Ther. 2012, 11, 452–463. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  135. Nakashima, T.; Clayman, G.L. Antisense inhibition of cyclin D1 in human head and neck squamous cell carcinoma. Arch. Otolaryngol. Head Neck Surg. 2000, 126, 957–961. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  136. Sauter, E.R.; Nesbit, M.; Litwin, S.; Klein-Szanto, A.J.; Cheffetz, S.; Herlyn, M. Antisense cyclin D1 induces apoptosis and tumor shrinkage in human squamous carcinomas. Cancer Res. 1999, 59, 4876–4881. [Google Scholar]
  137. Masuda, M.; Suzui, M.; Yasumatu, R.; Nakashima, T.; Kuratomi, Y.; Azuma, K.; Tomita, K.; Komiyama, S.; Weinstein, I.B. Constitutive activation of signal transducers and activators of transcription 3 correlates with cyclin D1 overexpression and may provide a novel prognostic marker in head and neck squamous cell carcinoma. Cancer Res. 2002, 62, 3351–3355. [Google Scholar]
  138. Masuda, M.; Toh, S.; Koike, K.; Kuratomi, Y.; Suzui, M.; Deguchi, A.; Komiyama, S.; Weinstein, I.B. The roles of JNK1 and Stat3 in the response of head and neck cancer cell lines to combined treatment with all-trans-retinoic acid and 5-fluorouracil. Jpn. J. Cancer Res. 2002, 93, 329–339. [Google Scholar] [CrossRef]
  139. Ahmed, K.M.; Fan, M.; Nantajit, D.; Cao, N.; Li, J.J. Cyclin D1 in low-dose radiation-induced adaptive resistance. Oncogene 2008, 27, 6738–6748. [Google Scholar] [CrossRef] [Green Version]
  140. Su, H.; Jin, X.; Shen, L.; Fang, Y.; Fei, Z.; Zhang, X.; Xie, C.; Chen, X. Inhibition of cyclin D1 enhances sensitivity to radiotherapy and reverses epithelial to mesenchymal transition for esophageal cancer cells. Tumour Biol. 2016, 37, 5355–5363. [Google Scholar] [CrossRef]
  141. Casimiro, M.C.; Di Sante, G.; Ju, X.; Li, Z.; Chen, K.; Crosariol, M.; Yaman, I.; Gormley, M.; Meng, H.; Lisanti, M.P.; et al. Cyclin D1 Promotes Androgen-Dependent DNA Damage Repair in Prostate Cancer Cells. Cancer Res. 2016, 76, 329–338. [Google Scholar] [CrossRef] [Green Version]
  142. Song, R.; Wei, X.; Wang, Y.; Hu, S.; Ba, Y.; Xiao, X.; Zhang, J. Insulinoma-associated protein 1 controls nasopharyngeal carcinoma to radiotherapy by modulating cyclin D1-dependent DNA repair machinery. Carcinogenesis 2020, 41, 326–333. [Google Scholar] [CrossRef] [PubMed]
  143. Yu, H.; Kortylewski, M.; Pardoll, D. Crosstalk between cancer and immune cells: Role of STAT3 in the tumour microenvironment. Nat. Rev. Immunol. 2007, 7, 41–51. [Google Scholar] [CrossRef]
  144. Wang, T.; Niu, G.; Kortylewski, M.; Burdelya, L.; Shain, K.; Zhang, S.; Bhattacharya, R.; Gabrilovich, D.; Heller, R.; Coppola, D.; et al. Regulation of the innate and adaptive immune responses by Stat-3 signaling in tumor cells. Nat. Med. 2004, 10, 48–54. [Google Scholar] [CrossRef] [PubMed]
  145. Kortylewski, M.; Kujawski, M.; Wang, T.; Wei, S.; Zhang, S.; Pilon-Thomas, S.; Niu, G.; Kay, H.; Mulé, J.; Kerr, W.G.; et al. Inhibiting Stat3 signaling in the hematopoietic system elicits multicomponent antitumor immunity. Nat. Med. 2005, 11, 1314–1321. [Google Scholar] [CrossRef] [PubMed]
  146. Nabarro, S.; Himoudi, N.; Papanastasiou, A.; Gilmour, K.; Gibson, S.; Sebire, N.; Thrasher, A.; Blundell, M.; Hubank, M.; Canderan, G.; et al. Coordinated oncogenic transformation and inhibition of host immune responses by the PAX3-FKHR fusion oncoprotein. J. Exp. Med. 2005, 202, 1399–1410. [Google Scholar] [CrossRef] [Green Version]
  147. Nefedova, Y.; Huang, M.; Kusmartsev, S.; Bhattacharya, R.; Cheng, P.; Salup, R.; Jove, R.; Gabrilovich, D. Hyperactivation of STAT3 is involved in abnormal differentiation of dendritic cells in cancer. J. Immunol. 2004, 172, 464–474. [Google Scholar] [CrossRef]
  148. Larmonier, N.; Marron, M.; Zeng, Y.; Cantrell, J.; Romanoski, A.; Sepassi, M.; Thompson, S.; Chen, X.; Andreansky, S.; Katsanis, E. Tumor-derived CD4(+)CD25(+) regulatory T cell suppression of dendritic cell function involves TGF-beta and IL-10. Cancer Immunol. Immunother. 2007, 56, 48–59. [Google Scholar] [CrossRef]
  149. Benkhart, E.M.; Siedlar, M.; Wedel, A.; Werner, T.; Ziegler-Heitbrock, H.W. Role of Stat3 in lipopolysaccharide-induced IL-10 gene expression. J. Immunol. 2000, 165, 1612–1617. [Google Scholar] [CrossRef] [Green Version]
  150. Kinjyo, I.; Inoue, H.; Hamano, S.; Fukuyama, S.; Yoshimura, T.; Koga, K.; Takaki, H.; Himeno, K.; Takaesu, G.; Kobayashi, T.; et al. Loss of SOCS3 in T helper cells resulted in reduced immune responses and hyperproduction of interleukin 10 and transforming growth factor-beta 1. J. Exp. Med. 2006, 203, 1021–1031. [Google Scholar] [CrossRef]
  151. Kasprzycka, M.; Marzec, M.; Liu, X.; Zhang, Q.; Wasik, M.A. Nucleophosmin/anaplastic lymphoma kinase (NPM/ALK) oncoprotein induces the T regulatory cell phenotype by activating STAT3. Proc. Natl Acad. Sci. USA 2006, 103, 9964–9969. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  152. Wei, D.; Le, X.; Zheng, L.; Wang, L.; Frey, J.A.; Gao, A.C.; Peng, Z.; Huang, S.; Xiong, H.Q.; Abbruzzese, J.L.; et al. Stat3 activation regulates the expression of vascular endothelial growth factor and human pancreatic cancer angiogenesis and metastasis. Oncogene 2003, 22, 319–329. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  153. Lee, Y.K.; Shanafelt, T.D.; Bone, N.D.; Strege, A.K.; Jelinek, D.F.; Kay, N.E. VEGF receptors on chronic lymphocytic leukemia (CLL) B cells interact with STAT 1 and 3: Implication for apoptosis resistance. Leukemia 2005, 19, 513–523. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  154. Song, T.L.; Nairismägi, M.L.; Laurensia, Y.; Lim, J.Q.; Tan, J.; Li, Z.M.; Pang, W.L.; Kizhakeyil, A.; Wijaya, G.C.; Huang, D.C.; et al. Oncogenic activation of the STAT3 pathway drives PD-L1 expression in natural killer/T-cell lymphoma. Blood 2018, 132, 1146–1158. [Google Scholar] [CrossRef] [Green Version]
  155. Xu, L.J.; Ma, Q.; Zhu, J.; Li, J.; Xue, B.X.; Gao, J.; Sun, C.Y.; Zang, Y.C.; Zhou, Y.B.; Yang, D.R.; et al. Combined inhibition of JAK1,2/Stat3-PD-L1 signaling pathway suppresses the immune escape of castration-resistant prostate cancer to NK cells in hypoxia. Mol. Med. Rep. 2018, 17, 8111–8120. [Google Scholar] [CrossRef] [Green Version]
  156. Shao, H.; Quintero, A.J.; Tweardy, D.J. Identification and characterization of cis elements in the STAT3 gene regulating STAT3 alpha and STAT3 beta messenger RNA splicing. Blood 2001, 98, 3853–3856. [Google Scholar] [CrossRef]
  157. Biethahn, S.; Alves, F.; Wilde, S.; Hiddemann, W.; Spiekermann, K. Expression of granulocyte colony-stimulating factor- and granulocyte-macrophage colony-stimulating factor-associated signal transduction proteins of the JAK/STAT pathway in normal granulopoiesis and in blast cells of acute myelogenous leukemia. Exp. Hematol. 1999, 27, 885–894. [Google Scholar] [CrossRef]
  158. Chakraborty, A.; White, S.M.; Schaefer, T.S.; Ball, E.D.; Dyer, K.F.; Tweardy, D.J. Granulocyte colony-stimulating factor activation of Stat3 alpha and Stat3 beta in immature normal and leukemic human myeloid cells. Blood 1996, 88, 2442–2449. [Google Scholar] [CrossRef] [Green Version]
  159. Zhang, Y.W.; Wang, L.M.; Jove, R.; Vande Woude, G.F. Requirement of Stat3 signaling for HGF/SF-Met mediated tumorigenesis. Oncogene 2002, 21, 217–226. [Google Scholar] [CrossRef]
  160. Dang, W.; Tang, H.; Cao, H.; Wang, L.; Zhang, X.; Tian, W.; Pang, X.; Li, K.; Chen, T. Strategy of STAT3beta cell-specific expression in macrophages exhibits antitumor effects on mouse breast cancer. Gene Ther. 2015, 22, 977–983. [Google Scholar] [CrossRef] [Green Version]
  161. Schaefer, T.S.; Sanders, L.K.; Nathans, D. Cooperative transcriptional activity of Jun and Stat3 beta, a short form of Stat3. Proc. Natl. Acad. Sci. USA 1995, 92, 9097–9101. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  162. Schaefer, T.S.; Sanders, L.K.; Park, O.K.; Nathans, D. Functional differences between Stat3alpha and Stat3beta. Mol. Cell. Biol. 1997, 17, 5307–5316. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  163. Ivanova, A.V.; Ivanov, S.V.; Zhang, X.; Ivanov, V.N.; Timofeeva, O.A.; Lerman, M.I. STRA13 interacts with STAT3 and modulates transcription of STAT3-dependent targets. J. Mol. Biol. 2004, 340, 641–653. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  164. Ng, I.H.; Bogoyevitch, M.A.; Jans, D.A. Cytokine-induced slowing of STAT3 nuclear import; faster basal trafficking of the STAT3beta isoform. Traffic 2014, 15, 946–960. [Google Scholar] [CrossRef] [PubMed]
  165. Ivanov, V.N.; Krasilnikov, M.; Ronai, Z. Regulation of Fas expression by STAT3 and c-Jun is mediated by phosphatidylinositol 3-kinase-AKT signaling. J. Biol. Chem. 2002, 277, 4932–4944. [Google Scholar] [CrossRef] [Green Version]
  166. Zammarchi, F.; de Stanchina, E.; Bournazou, E.; Supakorndej, T.; Martires, K.; Riedel, E.; Corben, A.D.; Bromberg, J.F.; Cartegni, L. Antitumorigenic potential of STAT3 alternative splicing modulation. Proc. Natl. Acad. Sci. USA 2011, 108, 17779–17784. [Google Scholar] [CrossRef] [Green Version]
  167. Karni, R.; Jove, R.; Levitzki, A. Inhibition of pp60c-Src reduces Bcl-XL expression and reverses the transformed phenotype of cells overexpressing EGF and HER-2 receptors. Oncogene 1999, 18, 4654–4662. [Google Scholar] [CrossRef] [Green Version]
  168. Xu, G.; Zhang, C.; Zhang, J. Dominant negative STAT3 suppresses the growth and invasion capability of human lung cancer cells. Mol. Med. Rep. 2009, 2, 819–824. [Google Scholar] [CrossRef] [Green Version]
  169. Niu, G.; Shain, K.H.; Huang, M.; Ravi, R.; Bedi, A.; Dalton, W.S.; Jove, R.; Yu, H. Overexpression of a dominant-negative signal transducer and activator of transcription 3 variant in tumor cells leads to production of soluble factors that induce apoptosis and cell cycle arrest. Cancer Res. 2001, 61, 3276–3280. [Google Scholar]
  170. Aigner, P.; Mizutani, T.; Horvath, J.; Eder, T.; Heber, S.; Lind, K.; Just, V.; Moll, H.P.; Yeroslaviz, A.; Fischer, M.J.M.; et al. STAT3beta is a tumor suppressor in acute myeloid leukemia. Blood Adv. 2019, 3, 1989–2002. [Google Scholar] [CrossRef] [Green Version]
  171. Nevo-Caspi, Y.; Amariglio, N.; Rechavi, G.; Paret, G. A-to-I RNA editing is induced upon hypoxia. Shock 2011, 35, 585–589. [Google Scholar] [CrossRef] [PubMed]
  172. Goldberg, L.; Abutbul-Amitai, M.; Paret, G.; Nevo-Caspi, Y. Alternative Splicing of STAT3 Is Affected by RNA Editing. DNA Cell Biol. 2017, 36, 367–376. [Google Scholar] [CrossRef] [PubMed]
  173. Wang, X.; Guo, J.; Che, X.; Jia, R. PCBP1 inhibits the expression of oncogenic STAT3 isoform by targeting alternative splicing of STAT3 exon 23. Int. J. Biol. Sci. 2019, 15, 1177–1186. [Google Scholar] [CrossRef] [PubMed]
  174. Tano, V.; Jans, D.A.; Bogoyevitch, M.A. Oligonucleotide-directed STAT3 alternative splicing switch drives anti-tumorigenic outcomes in MCF10 human breast cancer cells. Biochem. Biophys. Res. Commun. 2019, 513, 1076–1082. [Google Scholar] [CrossRef] [PubMed]
  175. Aigner, P.; Just, V.; Stoiber, D. STAT3 isoforms: Alternative fates in cancer? Cytokine 2019, 118, 27–34. [Google Scholar] [CrossRef] [PubMed]
  176. Drubay, V.; Skrypek, N.; Cordiez, L.; Vasseur, R.; Schulz, C.; Boukrout, N.; Duchene, B.; Coppin, L.; Van Seuningen, I.; Jonckheere, N. TGF-betaRII Knock-down in Pancreatic Cancer Cells Promotes Tumor Growth and Gemcitabine Resistance. Importance of STAT3 Phosphorylation on S727. Cancers 2018, 10, 254. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  177. Banerjee, A.S.; Pal, A.D.; Banerjee, S. Epstein-Barr virus-encoded small non-coding RNAs induce cancer cell chemoresistance and migration. Virology 2013, 443, 294–305. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  178. Hu, T.; Yeh, J.E.; Pinello, L.; Jacob, J.; Chakravarthy, S.; Yuan, G.C.; Chopra, R.; Frank, D.A. Impact of the N-Terminal Domain of STAT3 in STAT3-Dependent Transcriptional Activity. Mol. Cell. Biol. 2015, 35, 3284–3300. [Google Scholar] [CrossRef] [Green Version]
  179. Timofeeva, O.A.; Gaponenko, V.; Lockett, S.J.; Tarasov, S.G.; Jiang, S.; Michejda, C.J.; Perantoni, A.O.; Tarasova, N.I. Rationally designed inhibitors identify STAT3 N-domain as a promising anticancer drug target. ACS Chem. Biol. 2007, 2, 799–809. [Google Scholar] [CrossRef]
  180. Timofeeva, O.A.; Tarasova, N.I.; Zhang, X.; Chasovskikh, S.; Cheema, A.K.; Wang, H.; Brown, M.L.; Dritschilo, A. STAT3 suppresses transcription of proapoptotic genes in cancer cells with the involvement of its N-terminal domain. Proc. Natl. Acad. Sci. USA 2013, 110, 1267–1272. [Google Scholar] [CrossRef] [Green Version]
  181. Weidler, M.; Rether, J.; Anke, T.; Erkel, G. Inhibition of interleukin-6 signaling by galiellalactone. FEBS Lett. 2000, 484, 1–6. [Google Scholar] [CrossRef]
  182. Hellsten, R.; Johansson, M.; Dahlman, A.; Dizeyi, N.; Sterner, O.; Bjartell, A. Galiellalactone is a novel therapeutic candidate against hormone-refractory prostate cancer expressing activated Stat3. Prostate 2008, 68, 269–280. [Google Scholar] [CrossRef] [PubMed]
  183. Hellsten, R.; Johansson, M.; Dahlman, A.; Sterner, O.; Bjartell, A. Galiellalactone inhibits stem cell-like ALDH-positive prostate cancer cells. PLoS ONE 2011, 6, e22118. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  184. Don-Doncow, N.; Escobar, Z.; Johansson, M.; Kjellstrom, S.; Garcia, V.; Munoz, E.; Sterner, O.; Bjartell, A.; Hellsten, R. Galiellalactone is a direct inhibitor of the transcription factor STAT3 in prostate cancer cells. J. Biol. Chem. 2014, 289, 15969–15978. [Google Scholar] [CrossRef] [Green Version]
  185. Thaper, D.; Vahid, S.; Kaur, R.; Kumar, S.; Nouruzi, S.; Bishop, J.L.; Johansson, M.; Zoubeidi, A. Galiellalactone inhibits the STAT3/AR signaling axis and suppresses Enzalutamide-resistant Prostate Cancer. Sci. Rep. 2018, 8, 17307. [Google Scholar] [CrossRef]
  186. Ko, H.; Lee, J.H.; Kim, H.S.; Kim, T.; Han, Y.T.; Suh, Y.G.; Chun, J.; Kim, Y.S.; Ahn, K.S. Novel Galiellalactone Analogues Can Target STAT3 Phosphorylation and Cause Apoptosis in Triple-Negative Breast Cancer. Biomolecules 2019, 9, 170. [Google Scholar] [CrossRef] [Green Version]
  187. Escobar, Z.; Bjartell, A.; Canesin, G.; Evans-Axelsson, S.; Sterner, O.; Hellsten, R.; Johansson, M.H. Preclinical Characterization of 3beta-(N-Acetyl l-cysteine methyl ester)-2abeta,3-dihydrogaliellalactone (GPA512), a Prodrug of a Direct STAT3 Inhibitor for the Treatment of Prostate Cancer. J. Med. Chem. 2016, 59, 4551–4562. [Google Scholar] [CrossRef]
  188. Chen, H.; Yang, Z.; Ding, C.; Chu, L.; Zhang, Y.; Terry, K.; Liu, H.; Shen, Q.; Zhou, J. Discovery of O-Alkylamino Tethered Niclosamide Derivatives as Potent and Orally Bioavailable Anticancer Agents. ACS Med. Chem. Lett. 2013, 4, 180–185. [Google Scholar] [CrossRef]
  189. Huang, W.; Dong, Z.; Wang, F.; Peng, H.; Liu, J.Y.; Zhang, J.T. A small molecule compound targeting STAT3 DNA-binding domain inhibits cancer cell proliferation, migration, and invasion. ACS Chem. Biol. 2014, 9, 1188–1196. [Google Scholar] [CrossRef] [Green Version]
  190. Huang, W.; Dong, Z.; Chen, Y.; Wang, F.; Wang, C.J.; Peng, H.; He, Y.; Hangoc, G.; Pollok, K.; Sandusky, G.; et al. Small-molecule inhibitors targeting the DNA-binding domain of STAT3 suppress tumor growth, metastasis and STAT3 target gene expression in vivo. Oncogene 2016, 35, 783–792. [Google Scholar] [CrossRef]
  191. Huang, W.; Liu, Y.; Wang, J.; Yuan, X.; Jin, H.W.; Zhang, L.R.; Zhang, J.T.; Liu, Z.M.; Cui, J.R. Small-molecule compounds targeting the STAT3 DNA-binding domain suppress survival of cisplatin-resistant human ovarian cancer cells by inducing apoptosis. Eur. J. Med. Chem. 2018, 157, 887–897. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  192. Son, D.J.; Zheng, J.; Jung, Y.Y.; Hwang, C.J.; Lee, H.P.; Woo, J.R.; Baek, S.Y.; Ham, Y.W.; Kang, M.W.; Shong, M.; et al. MMPP Attenuates Non-Small Cell Lung Cancer Growth by Inhibiting the STAT3 DNA-Binding Activity via Direct Binding to the STAT3 DNA-Binding Domain. Theranostics 2017, 7, 4632–4642. [Google Scholar] [CrossRef] [PubMed]
  193. Selvendiran, K.; Ahmed, S.; Dayton, A.; Kuppusamy, M.L.; Rivera, B.K.; Kalai, T.; Hideg, K.; Kuppusamy, P. HO-3867, a curcumin analog, sensitizes cisplatin-resistant ovarian carcinoma, leading to therapeutic synergy through STAT3 inhibition. Cancer Biol. Ther. 2011, 12, 837–845. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  194. Mast, J.M.; Tse, D.; Shee, K.; Lakshmi Kuppusamy, M.; Kmiec, M.M.; Kalai, T.; Kuppusamy, P. Diarylidenylpiperidones, H-4073 and HO-3867, Induce G2/M Cell-Cycle Arrest, Apoptosis and Inhibit STAT3 Phosphorylation in Human Pancreatic Cancer Cells. Cell Biochem. Biophys. 2019, 77, 109–119. [Google Scholar] [CrossRef]
  195. Selvendiran, K.; Tong, L.; Bratasz, A.; Kuppusamy, M.L.; Ahmed, S.; Ravi, Y.; Trigg, N.J.; Rivera, B.K.; Kalai, T.; Hideg, K.; et al. Anticancer efficacy of a difluorodiarylidenyl piperidone (HO-3867) in human ovarian cancer cells and tumor xenografts. Mol. Cancer Ther. 2010, 9, 1169–1179. [Google Scholar] [CrossRef] [Green Version]
  196. Buettner, R.; Corzano, R.; Rashid, R.; Lin, J.; Senthil, M.; Hedvat, M.; Schroeder, A.; Mao, A.; Herrmann, A.; Yim, J.; et al. Alkylation of cysteine 468 in Stat3 defines a novel site for therapeutic development. ACS Chem. Biol. 2011, 6, 432–443. [Google Scholar] [CrossRef]
  197. Koseki, T.; Suehiro, N.; Masuda, Y.; Miyoshi, N.; Muraoka, D.; Ogo, N.; Asai, A. Discovery of a New STAT3 Inhibitor Acting on the Linker Domain. Biol. Pharm. Bull. 2019, 42, 792–800. [Google Scholar] [CrossRef] [Green Version]
  198. Grab, J.; Berg, A.; Blechschmidt, L.; Kluver, B.; Rubner, S.; Fu, D.Y.; Meiler, J.; Graber, M.; Berg, T. The STAT5b Linker Domain Mediates the Selectivity of Catechol Bisphosphates for STAT5b over STAT5a. ACS Chem. Biol. 2019, 14, 796–805. [Google Scholar] [CrossRef]
  199. Turkson, J.; Kim, J.S.; Zhang, S.; Yuan, J.; Huang, M.; Glenn, M.; Haura, E.; Sebti, S.; Hamilton, A.D.; Jove, R. Novel peptidomimetic inhibitors of signal transducer and activator of transcription 3 dimerization and biological activity. Mol. Cancer Ther. 2004, 3, 261–269. [Google Scholar]
  200. Zhao, W.; Jaganathan, S.; Turkson, J. A cell-permeable Stat3 SH2 domain mimetic inhibits Stat3 activation and induces antitumor cell effects in vitro. J. Biol. Chem. 2010, 285, 35855–35865. [Google Scholar] [CrossRef] [Green Version]
  201. Auzenne, E.J.; Klostergaard, J.; Mandal, P.K.; Liao, W.S.; Lu, Z.; Gao, F.; Bast, R.C., Jr.; Robertson, F.M.; McMurray, J.S. A phosphopeptide mimetic prodrug targeting the SH2 domain of Stat3 inhibits tumor growth and angiogenesis. J. Exp. Ther. Oncol. 2012, 10, 155–162. [Google Scholar] [PubMed]
  202. Schust, J.; Sperl, B.; Hollis, A.; Mayer, T.U.; Berg, T. Stattic: A small-molecule inhibitor of STAT3 activation and dimerization. Chem. Biol. 2006, 13, 1235–1242. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  203. Xu, G.; Zhu, L.; Wang, Y.; Shi, Y.; Gong, A.; Wu, C. Stattic Enhances Radiosensitivity and Reduces Radio-Induced Migration and Invasion in HCC Cell Lines through an Apoptosis Pathway. Biomed Res. Int. 2017, 2017, 1832494. [Google Scholar] [CrossRef] [PubMed]
  204. Song, H.; Wang, R.; Wang, S.; Lin, J. A low-molecular-weight compound discovered through virtual database screening inhibits Stat3 function in breast cancer cells. Proc. Natl. Acad. Sci. USA 2005, 102, 4700–4705. [Google Scholar] [CrossRef] [Green Version]
  205. Miyoshi, K.; Takaishi, M.; Nakajima, K.; Ikeda, M.; Kanda, T.; Tarutani, M.; Iiyama, T.; Asao, N.; DiGiovanni, J.; Sano, S. Stat3 as a therapeutic target for the treatment of psoriasis: A clinical feasibility study with STA-21, a Stat3 inhibitor. J. Investig. Dermatol. 2011, 131, 108–117. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  206. Park, J.S.; Kwok, S.K.; Lim, M.A.; Kim, E.K.; Ryu, J.G.; Kim, S.M.; Oh, H.J.; Ju, J.H.; Park, S.H.; Kim, H.Y.; et al. STA-21, a promising STAT-3 inhibitor that reciprocally regulates Th17 and Treg cells, inhibits osteoclastogenesis in mice and humans and alleviates autoimmune inflammation in an experimental model of rheumatoid arthritis. Arthritis Rheumatol. 2014, 66, 918–929. [Google Scholar] [CrossRef] [PubMed]
  207. Mencalha, A.L.; Du Rocher, B.; Salles, D.; Binato, R.; Abdelhay, E. LLL-3, a STAT3 inhibitor, represses BCR-ABL-positive cell proliferation, activates apoptosis and improves the effects of Imatinib mesylate. Cancer Chemother. Pharmacol. 2010, 65, 1039–1046. [Google Scholar] [CrossRef]
  208. Fuh, B.; Sobo, M.; Cen, L.; Josiah, D.; Hutzen, B.; Cisek, K.; Bhasin, D.; Regan, N.; Lin, L.; Chan, C.; et al. LLL-3 inhibits STAT3 activity, suppresses glioblastoma cell growth and prolongs survival in a mouse glioblastoma model. Br. J. Cancer 2009, 100, 106–112. [Google Scholar] [CrossRef] [Green Version]
  209. Lin, L.; Hutzen, B.; Li, P.K.; Ball, S.; Zuo, M.; DeAngelis, S.; Foust, E.; Sobo, M.; Friedman, L.; Bhasin, D.; et al. A novel small molecule, LLL12, inhibits STAT3 phosphorylation and activities and exhibits potent growth-suppressive activity in human cancer cells. Neoplasia 2010, 12, 39–50. [Google Scholar] [CrossRef] [Green Version]
  210. Zhao, C.; Wang, W.; Yu, W.; Jou, D.; Wang, Y.; Ma, H.; Xiao, H.; Qin, H.; Zhang, C.; Lu, J.; et al. A novel small molecule STAT3 inhibitor, LY5, inhibits cell viability, colony formation, and migration of colon and liver cancer cells. Oncotarget 2016, 7, 12917–12926. [Google Scholar] [CrossRef] [Green Version]
  211. Siddiquee, K.; Zhang, S.; Guida, W.C.; Blaskovich, M.A.; Greedy, B.; Lawrence, H.R.; Yip, M.L.; Jove, R.; McLaughlin, M.M.; Lawrence, N.J.; et al. Selective chemical probe inhibitor of Stat3, identified through structure-based virtual screening, induces antitumor activity. Proc. Natl. Acad. Sci. USA 2007, 104, 7391–7396. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  212. Ball, D.P.; Lewis, A.M.; Williams, D.; Resetca, D.; Wilson, D.J.; Gunning, P.T. Signal transducer and activator of transcription 3 (STAT3) inhibitor, S3I-201, acts as a potent and non-selective alkylating agent. Oncotarget 2016, 7, 20669–20679. [Google Scholar] [CrossRef] [PubMed]
  213. Zhang, X.; Sun, Y.; Pireddu, R.; Yang, H.; Urlam, M.K.; Lawrence, H.R.; Guida, W.C.; Lawrence, N.J.; Sebti, S.M. A novel inhibitor of STAT3 homodimerization selectively suppresses STAT3 activity and malignant transformation. Cancer Res. 2013, 73, 1922–1933. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  214. Fletcher, S.; Singh, J.; Zhang, X.; Yue, P.; Page, B.D.; Sharmeen, S.; Shahani, V.M.; Zhao, W.; Schimmer, A.D.; Turkson, J.; et al. Disruption of transcriptionally active Stat3 dimers with non-phosphorylated, salicylic acid-based small molecules: Potent in vitro and tumor cell activities. Chembiochem 2009, 10, 1959–1964. [Google Scholar] [CrossRef]
  215. Zhang, X.; Yue, P.; Fletcher, S.; Zhao, W.; Gunning, P.T.; Turkson, J. A novel small-molecule disrupts Stat3 SH2 domain-phosphotyrosine interactions and Stat3-dependent tumor processes. Biochem. Pharmacol. 2010, 79, 1398–1409. [Google Scholar] [CrossRef] [Green Version]
  216. Zhang, X.; Yue, P.; Page, B.D.; Li, T.; Zhao, W.; Namanja, A.T.; Paladino, D.; Zhao, J.; Chen, Y.; Gunning, P.T.; et al. Orally bioavailable small-molecule inhibitor of transcription factor Stat3 regresses human breast and lung cancer xenografts. Proc. Natl. Acad. Sci. USA 2012, 109, 9623–9628. [Google Scholar] [CrossRef] [Green Version]
  217. Haftchenary, S.; Luchman, H.A.; Jouk, A.O.; Veloso, A.J.; Page, B.D.; Cheng, X.R.; Dawson, S.S.; Grinshtein, N.; Shahani, V.M.; Kerman, K.; et al. Potent Targeting of the STAT3 Protein in Brain Cancer Stem Cells: A Promising Route for Treating Glioblastoma. ACS Med. Chem. Lett. 2013, 4, 1102–1107. [Google Scholar] [CrossRef] [Green Version]
  218. Hayakawa, F.; Sugimoto, K.; Harada, Y.; Hashimoto, N.; Ohi, N.; Kurahashi, S.; Naoe, T. A novel STAT inhibitor, OPB-31121, has a significant antitumor effect on leukemia with STAT-addictive oncokinases. Blood Cancer J. 2013, 3, e166. [Google Scholar] [CrossRef]
  219. Kim, M.J.; Nam, H.J.; Kim, H.P.; Han, S.W.; Im, S.A.; Kim, T.Y.; Oh, D.Y.; Bang, Y.J. OPB-31121, a novel small molecular inhibitor, disrupts the JAK2/STAT3 pathway and exhibits an antitumor activity in gastric cancer cells. Cancer Lett. 2013, 335, 145–152. [Google Scholar] [CrossRef]
  220. Wong, A.L.; Soo, R.A.; Tan, D.S.; Lee, S.C.; Lim, J.S.; Marban, P.C.; Kong, L.R.; Lee, Y.J.; Wang, L.Z.; Thuya, W.L.; 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, 998–1005. [Google Scholar] [CrossRef]
  221. Ogura, M.; Uchida, T.; Terui, Y.; Hayakawa, F.; Kobayashi, Y.; Taniwaki, M.; Takamatsu, Y.; Naoe, T.; Tobinai, K.; Munakata, W.; et al. Phase I study of OPB-51602, an oral inhibitor of signal transducer and activator of transcription 3, in patients with relapsed/refractory hematological malignancies. Cancer Sci. 2015, 106, 896–901. [Google Scholar] [CrossRef] [PubMed]
  222. Brambilla, L.; Genini, D.; Laurini, E.; Merulla, J.; Perez, L.; Fermeglia, M.; Carbone, G.M.; Pricl, S.; Catapano, C.V. Hitting the right spot: Mechanism of action of OPB-31121, a novel and potent inhibitor of the Signal Transducer and Activator of Transcription 3 (STAT3). Mol. Oncol. 2015, 9, 1194–1206. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  223. Genini, D.; Brambilla, L.; Laurini, E.; Merulla, J.; Civenni, G.; Pandit, S.; D’Antuono, R.; Perez, L.; Levy, D.E.; Pricl, S.; et al. Mitochondrial dysfunction induced by a SH2 domain-targeting STAT3 inhibitor leads to metabolic synthetic lethality in cancer cells. Proc. Natl. Acad. Sci. USA 2017, 114, E4924–E4933. [Google Scholar] [CrossRef] [Green Version]
  224. Orlova, A.; Wagner, C.; de Araujo, E.D.; Bajusz, D.; Neubauer, H.A.; Herling, M.; Gunning, P.T.; Keseru, G.M.; Moriggl, R. Direct Targeting Options for STAT3 and STAT5 in Cancer. Cancers 2019, 11, 1930. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  225. Oh, D.Y.; Lee, S.H.; Han, S.W.; Kim, M.J.; Kim, T.M.; Kim, T.Y.; Heo, D.S.; Yuasa, M.; Yanagihara, Y.; Bang, Y.J. Phase I Study of OPB-31121, an Oral STAT3 Inhibitor, in Patients with Advanced Solid Tumors. Cancer Res. Treat. 2015, 47, 607–615. [Google Scholar] [CrossRef] [PubMed]
  226. Bendell, J.C.; Hong, D.S.; Burris, H.A., 3rd; Naing, A.; Jones, S.F.; Falchook, G.; Bricmont, P.; Elekes, A.; Rock, E.P.; Kurzrock, R. Phase 1, open-label, dose-escalation, and pharmacokinetic study of STAT3 inhibitor OPB-31121 in subjects with advanced solid tumors. Cancer Chemother. Pharmacol. 2014, 74, 125–130. [Google Scholar] [CrossRef]
  227. Verdura, S.; Cuyas, E.; Llorach-Pares, L.; Perez-Sanchez, A.; Micol, V.; Nonell-Canals, A.; Joven, J.; Valiente, M.; Sanchez-Martinez, M.; Bosch-Barrera, J.; et al. Silibinin is a direct inhibitor of STAT3. Food Chem. Toxicol. 2018, 116, 161–172. [Google Scholar] [CrossRef]
  228. Mertens, C.; Haripal, B.; Klinge, S.; Darnell, J.E. Mutations in the linker domain affect phospho-STAT3 function and suggest targets for interrupting STAT3 activity. Proc. Natl. Acad. Sci. USA 2015, 112, 14811–14816. [Google Scholar] [CrossRef] [Green Version]
  229. Kraskouskaya, D.; Duodu, E.; Arpin, C.C.; Gunning, P.T. Progress towards the development of SH2 domain inhibitors. Chem. Soc. Rev. 2013, 42, 3337–3370. [Google Scholar] [CrossRef]
  230. Huang, M.; Song, K.; Liu, X.; Lu, S.; Shen, Q.; Wang, R.; Gao, J.; Hong, Y.; Li, Q.; Ni, D.; et al. AlloFinder: A strategy for allosteric modulator discovery and allosterome analyses. Nucleic Acids Res. 2018, 46, W451–W458. [Google Scholar] [CrossRef]
  231. De la Iglesia, N.; Konopka, G.; Lim, K.L.; Nutt, C.L.; Bromberg, J.F.; Frank, D.A.; Mischel, P.S.; Louis, D.N.; Bonni, A. Deregulation of a STAT3-interleukin 8 signaling pathway promotes human glioblastoma cell proliferation and invasiveness. J. Neurosci. 2008, 28, 5870–5878. [Google Scholar] [CrossRef] [PubMed]
  232. De la Iglesia, N.; Konopka, G.; Puram, S.V.; Chan, J.A.; Bachoo, R.M.; You, M.J.; Levy, D.E.; Depinho, R.A.; Bonni, A. Identification of a PTEN-regulated STAT3 brain tumor suppressor pathway. Genes Dev. 2008, 22, 449–462. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  233. Schneller, D.; Machat, G.; Sousek, A.; Proell, V.; van Zijl, F.; Zulehner, G.; Huber, H.; Mair, M.; Muellner, M.K.; Nijman, S.M.; et al. p19(ARF) /p14(ARF) controls oncogenic functions of signal transducer and activator of transcription 3 in hepatocellular carcinoma. Hepatology 2011, 54, 164–172. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Schematic diagram of the structure and the alternative splicing pattern of STAT3. PCBP1 binds to “UCCCCCCG” and promotes the usage of an alternative 3’ splicing site in exon 23, which increases the expression of STAT3β. The TAD (transcriptional activation domain) of STAT3α is replaced by FIDAVWK.
Figure 1. Schematic diagram of the structure and the alternative splicing pattern of STAT3. PCBP1 binds to “UCCCCCCG” and promotes the usage of an alternative 3’ splicing site in exon 23, which increases the expression of STAT3β. The TAD (transcriptional activation domain) of STAT3α is replaced by FIDAVWK.
Cancers 12 02459 g001
Figure 2. Schematic representation of different ratios of STAT3α/STAT3β and their functions in therapy sensitivity. (a) The STAT3 signaling pathway confers therapy resistance to cells, mainly by mediating its target genes for cell survival, proliferation and inhibition of apoptosis. It promotes epithelial-mesenchymal transition, restores cancer stem cell properties, enhances DNA repair and attenuates ROS levels. RTK inhibitors can augment cell survival and proliferation via feedback activation of STAT3. Thus, in combination with RTK inhibitors, STAT3 inhibitors can be an effective strategy for therapy resistance. (b) STAT3β activation is better for cell growth inhibition and apoptosis, in contrast with STAT3α functions. Switch regulation for STAT3α to STAT3β is a challenging approach for resensitization to chemotherapy. In the context of a high expression level of STAT3β, RTK inhibitors that activate STAT3 via a feedback loop may be of benefit for cell apoptosis. Abbreviations: AKT, protein kinase B; CSCs, cancer stem cells; EGFR, epidermal growth factor receptor; EMT, epithelial-mesenchymal transition; JAK, Janus kinase; P, phosphorylation; PCBP1, poly-C binding protein-1; ROS, reactive oxygen species; RTKi, receptor tyrosine kinase inhibitor; RTKs, receptor tyrosine kinases; STAT3, signal transducer and activator of transcription 3.
Figure 2. Schematic representation of different ratios of STAT3α/STAT3β and their functions in therapy sensitivity. (a) The STAT3 signaling pathway confers therapy resistance to cells, mainly by mediating its target genes for cell survival, proliferation and inhibition of apoptosis. It promotes epithelial-mesenchymal transition, restores cancer stem cell properties, enhances DNA repair and attenuates ROS levels. RTK inhibitors can augment cell survival and proliferation via feedback activation of STAT3. Thus, in combination with RTK inhibitors, STAT3 inhibitors can be an effective strategy for therapy resistance. (b) STAT3β activation is better for cell growth inhibition and apoptosis, in contrast with STAT3α functions. Switch regulation for STAT3α to STAT3β is a challenging approach for resensitization to chemotherapy. In the context of a high expression level of STAT3β, RTK inhibitors that activate STAT3 via a feedback loop may be of benefit for cell apoptosis. Abbreviations: AKT, protein kinase B; CSCs, cancer stem cells; EGFR, epidermal growth factor receptor; EMT, epithelial-mesenchymal transition; JAK, Janus kinase; P, phosphorylation; PCBP1, poly-C binding protein-1; ROS, reactive oxygen species; RTKi, receptor tyrosine kinase inhibitor; RTKs, receptor tyrosine kinases; STAT3, signal transducer and activator of transcription 3.
Cancers 12 02459 g002
Figure 3. The current status of STAT3 inhibitors in (pre)clinical trials and their chemical structures. The N-terminal domain, DNA-binding domain (DBD), coiled-coil domain (CCD), linker domain and Src homology 2 domain (SH2) are components of the STAT3 structure. a Inhibitors of Phase I/II clinical trials registered at https://clinicaltrials.gov. (ad), the structure of galiellalactone and its analogs. (ef), the structure of inS3-45 and inS3-54A18. (gj), the structure of STA-21 and its analogs. (kn), the structure of S3I-201 and its analogs.
Figure 3. The current status of STAT3 inhibitors in (pre)clinical trials and their chemical structures. The N-terminal domain, DNA-binding domain (DBD), coiled-coil domain (CCD), linker domain and Src homology 2 domain (SH2) are components of the STAT3 structure. a Inhibitors of Phase I/II clinical trials registered at https://clinicaltrials.gov. (ad), the structure of galiellalactone and its analogs. (ef), the structure of inS3-45 and inS3-54A18. (gj), the structure of STA-21 and its analogs. (kn), the structure of S3I-201 and its analogs.
Cancers 12 02459 g003

Share and Cite

MDPI and ACS Style

Yang, P.-L.; Liu, L.-X.; Li, E.-M.; Xu, L.-Y. STAT3, the Challenge for Chemotherapeutic and Radiotherapeutic Efficacy. Cancers 2020, 12, 2459. https://doi.org/10.3390/cancers12092459

AMA Style

Yang P-L, Liu L-X, Li E-M, Xu L-Y. STAT3, the Challenge for Chemotherapeutic and Radiotherapeutic Efficacy. Cancers. 2020; 12(9):2459. https://doi.org/10.3390/cancers12092459

Chicago/Turabian Style

Yang, Ping-Lian, Lu-Xin Liu, En-Min Li, and Li-Yan Xu. 2020. "STAT3, the Challenge for Chemotherapeutic and Radiotherapeutic Efficacy" Cancers 12, no. 9: 2459. https://doi.org/10.3390/cancers12092459

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

Article Metrics

Back to TopTop