[go: up one dir, main page]
More Web Proxy on the site http://driver.im/ Skip to content
BY 4.0 license Open Access Published by De Gruyter Open Access September 16, 2023

A pan-cancer analysis of STAT3 expression and genetic alterations in human tumors

  • Junyin Tan and Ronghao Feng EMAIL logo
From the journal Open Medicine

Abstract

Combined cancer immunotherapy and targeted therapy have proven to be effective against various cancers and therefore have recently become the focus of cancer research. Signal transducer and activator of transcription 3 (STAT3) is a member of the STAT protein family of transcription factors. Several studies have shown that STAT3 can affect the prognosis of cancer patients by regulating immune microenvironment (IME). Therefore, STAT3 may have high research value for the development of combined immunotherapy/targeted therapy approaches for the treatment of cancer patients. We found differences in STAT3 expression between tumor and normal tissues. Kaplan−Meier survival and Cox regression analyses showed that high expression of STAT3 is associated with poor prognosis in low-grade glioma (LGG) patients. The results of the analysis of the area under the curve of the receiver operating characteristic curve further suggested that the expression of STAT3 is an effective way to evaluate the prognosis of patients with glioma. The results of the IME analysis revealed that the immune and matrix scores of LGGs were positively correlated with the expression of STAT3 (P < 0.05). The results of immune cell infiltration analysis showed that STAT3 was positively correlated with resting dendritic cells, eosinophils, neutrophils, M0 macrophages, M1 macrophages, CD4 memory resting T cells, and CD8 T cells in LGG patients, but negatively correlated with activated mast cells and M2 macrophages (P < 0.05). Our gene set enrichment analysis identified 384 enriched pathways. According to the enrichment scores, the top ten most significantly upregulated pathways were related to immune response. The top ten most significantly downregulated pathways were related to cell signal transduction and the regulation of cell survival, proliferation, and metabolism. Genetic alteration analysis showed that missense mutations in STAT3 account for the majority of mutations, and STAT3 mutations mostly occur in the Src homology domain. In conclusion overexpression of STAT3 can promote the development and growth of tumors by regulating IME, which is significantly related to the poor prognosis of cancer patients. Therefore, targeted inhibition of STAT3 expression may have high research value for the development of combined immunotherapy/targeted therapy approaches for the treatment of cancer patients.

1 Introduction

Combined immunotherapy and targeted therapy have become a hot topic in cancer research, and immune therapy targeting immunosuppressive genes, such as CD274, PDCD1, CTLA4, LAG3, TIGIT, etc., have been shown to be effective. However, many clinical and experimental studies have revealed that not all tumors respond to immunotherapy against these targets. Therefore, finding new potential targets is critical for the development of more effective cancer therapies.

The signal transducer and activator of transcription (STAT) protein family of transcription factors in mammals consists of seven members (STAT1, STAT2, STAT3, STAT4, STAT5A, STAT5B, and STAT6), which are associated with the regulation of the cell cycle, cell survival, and immune response [111]. STAT3, which was first reported by Akira et al. [12], is a protein composed of 770 amino acids with six functionally conserved domains, including the amino-terminal domain, coiled-coil domain, DNA-binding domain, linker domain, Src homology 2 (SH2) domain, and trans-activation domain [13]. In most cancers, STAT3 is overactivated and promotes tumor progression by regulating various biological processes, such as proliferation, apoptosis, angiogenesis, and immune response, which are generally associated with poor clinical prognosis [10,1417]. Therefore, targeting the STAT3 signaling pathway has been recognized as a promising therapeutic strategy for numerous cancers [5].

Additionally, many studies have shown that STAT3 can affect the prognosis of tumor patients by regulating the immune microenvironment (IME) [1820]. There are studies to prove that derivatives of secondary metabolites can play an anticancer effect by regulating the STAT3 pathway, and show cytotoxicity to cancer cells but no toxicity to non-cancer cell lines, which may inspire development of new drug-like substances with improved cytotoxicity on cancer [2123]. However, the specific molecular mechanisms of STAT3 in the pathogenesis of different tumors remain unclear, as is their value in the human pan-cancer analysis. In addition, through literature search, we have found few studies on pan-cancer analysis of STAT3 from the perspective of overall tumor microenvironment. Therefore, in this study, we mainly used bioinformatics methods to investigate the impact of changes in STAT3 expression and genetic alterations on the development of cancer from the pan-cancer perspective, to provide new insights into the transformation and application of STAT3 in the development of more effective cancer treatments.

2 Materials and methods

2.1 Data retrieval and pre-processing

In order to unify the standard, we obtained the gene expression data, clinical data, and sample information of 33 cancers from The Cancer Genome Atlas (TCGA) database from the University of California, Santa Cruz (UCSC) Xena database (http://xena.ucsc.edu/) [24]. Also, we downloaded mutation data from TCGA (https://portal.gdc.cancer.gov/repository) [25]. In addition, we obtained the annotation information of the genes from the Ensembl human genome browser GRCh38. P13 (http://asia.ensembl.org/index.html) [26]. Additionally, the data of 1,018 glioma samples were downloaded from the Chinese Glioma Genome Atlas (CGGA, http://www.cgga.org.cn/) and used for subsequent verification [27].

2.2 Gene expression analysis

Wilcoxon test analysis was performed to determine significant differences in STAT3 expression between cancer samples and normal samples. Additionally, we also analyzed the protein expression dataset obtained from The National Cancer Institute’s Clinical Proteomic Tumor Analysis Consortium (CPTAC) through the UALCAN website (http://ualcan.path.uab.edu/analysis-prot.html) [28]. To this end, we opened the UALCAN website and typed “STAT3” to obtain the total protein expression level of STAT3 between primary tumor and normal tissues.

2.3 Prognostic analysis

First, 33 types of cancer were divided into high and low expression groups according to the median level of expression of STAT3. The overall survival (OS) time and progression-free survival (PFS) of the high and low expression groups were analyzed by Kaplan−Meier (K−M) and Cox survival analyses using the “survival” package in R. The K−M survival curve and Cox forest plot were plotted with the “survminer” and “forestplot” packages, respectively. The dataset downloaded from the CGGA was used to verify the prognostic role of STAT3 in low-grade glioma (LGG). Additionally, we also performed univariate and multivariate prognostic analysis and receiver operating characteristic (ROC) curve analysis on the CGGA dataset to determine whether STAT3 expression can be an independent prognostic factor for glioma patients and its accuracy. The immunohistochemical staining data of STAT3 protein in normal and glioma tissues were obtained from the Human Protein Atlas (HPA, https://www.proteinatlas.org/).

2.4 Immune correlation analysis

Immunotherapy and IME have long been the focus of tumor research. In order to further understand the mechanism by which STAT3 affects cancer prognosis from the perspective of immunity, we performed immune correlation analysis on the cancers with statistical significance in the survival analysis described in the previous section. We first evaluated the IME of each tumor based on TCGA expression data using the “estimate” package in R to obtain an immune score and a stromal score for each tumor, followed by a “Spearman” correlation analysis between these scores and STAT3 expression. Then, we used the CIBERSORT algorithm to evaluate the degree of infiltration of 22 immune cells in each cancer type [29]. Subsequently, we calculated the correlation between the degree of infiltration of each immune cell and STAT3 expression using the “Spearman” test. We also performed immune checkpoint correlation analysis, determined the correlation between common immune checkpoints and STAT3 expression levels by “Spearman” correlation analysis, and visualized the results as a heat map.

2.5 Enrichment analysis

We performed gene set enrichment analysis (GSEA) to identify the pathways through which activated STAT3 promotes tumor development. To this end, we first downloaded the gene set database file “c2.cp.kegg.v7.1.symbols.gmt” from the “downloads” in the Molecular Signatures Database (MSigDB) in the GSEA website (http://www.gsea-msigdb.org/) [30]. The data of 33 cancer types of TCGA were divided into high and low expression groups according to the median level of expression of STAT3, and the data were downloaded from the “org.Hs.eg.db” “clusterProfiler” “enrichplot” R package for GSEA analysis, and a P value less than 0.05 was considered significant.

2.6 Genetic alteration analysis

According to the mutation data downloaded from TCGA, we determined the tumor mutational burden (TMB) and microsatellite instability (MSI) in each tumor. Then, we analyzed the correlation between the TMB and MSI for each tumor and the expression of STAT3, determined the correlation coefficient and P-value, and used the “fmsb” package in R to visualize the results as a correlation radar map. The CBioPortal database (http://www.cbioportal.org/) was used to obtain, visualize, and analyze multidimensional cancer genomic data for subsequent analysis of STAT3 gene alterations [31,32]. We selected “Pan-cancer analysis of whole genomes (ICGC/TCGA, Nature 2020)” in the “Query” module, clicked the “Query by gene” button, and entered the “STAT3” gene. The results of the structural variation data, mutation data, and CNA data are shown in the “Cancer Types Summary” module. The analysis results of STAT3 mutation and its three-dimensional (3D) structure are in the “Mutation” module. The Catalogue of Somatic Mutations in Cancer (COSMIC, http://www.sanger.ac.uk/cosmic) is a database that preserves somatic mutation data and related information for further analysis of STAT3 mutations [33]. We typed “STAT3” in the query module and clicked “SEARCH”. In the results, the Gene view, Tissue distribution, Variants, Mutation distribution, and 3D structure of the STAT3 gene can be easily seen by clicking “STAT3”.

3 Results

3.1 Gene expression analysis

Ten types of tumors, namely breast cancer (BRCA), ovarian cancer, colon cancer, clear cell renal cell carcinoma (ccRCC), uterine corpus endometrial carcinoma (UCEC), lung cancer, head and neck squamous cell carcinoma (HNSC), glioblastoma, liver cancer, and pancreatic cancer in the CPTAC dataset were analyzed using the UALCAN web tool. The analysis revealed significant differences in STAT3 expression between the different types of tumors and normal tissues (P < 0.05, Figure 1). STAT3 was highly expressed in BRCA, ccRCC, UCEC, lung cancer, HNSC, pancreatic cancer, and glioblastoma, but lowly expressed in ovarian cancer, colon cancer, and liver cancer.

Figure 1 
                  Gene expression analysis between normal and tumor tissues that are curated by UALCAN website (P < 0.005).
Figure 1

Gene expression analysis between normal and tumor tissues that are curated by UALCAN website (P < 0.005).

3.2 Prognostic analysis

We first performed the K−M survival and Cox regression analyses of TCGA data. The K−M survival analysis revealed that the OS of patients with LGG (Figure 2a), tenosynovial giant cell tumors (TGCT) (Figure 2b), and skin cutaneous melanoma (SKCM) (Figure 2c) in the STAT3 high expression group was significantly different (P < 0.05). In particular, in LGG and TGCT patients, the OS in the low expression group was higher than that in the high expression group, while in SKCM patients the OS in the high expression group was higher than that in the low expression group. Cox regression analysis revealed that the OS of patients with LGG and SKCM (Figure 2d) was significantly correlated with the expression of STAT3 (LGG: hazard ratio [HR] 2.424, ranging from 1.692 to 3.474, P < 0.001; SKCM: HR 0.659, ranging from 0.531 to 0.819, P < 0.001). The PFS of patients with LGG (Figure 2e), HNSC (Figure 2f), SKCM (Figure 2g), prostate adenocarcinoma (PRAD) (Figure 2h), and colon adenocarcinoma (COAD) (Figure 2i) was significantly different, and the PFS of the low STAT3 expression group was significantly higher than that of the high expression group in LGG. Cox regression analysis (Figure 2j) revealed that only patients with LGG had significant difference in PFS and STAT3 expression (HR 2.230, ranging from 1.639 to 3.034, P < 0.001). Thus, it is evident that the expression of STAT3 in LGG is correlated with OS and PFS, and the difference is statistically significant. In addition, we validated the results with the CGGA glioma dataset and OS, and found that the results were similar to those mentioned above (K−M: P < 0.001, Figure 3a; Cox: HR 1.887, ranging from 1.670 to 2.132, P < 0.001, Table 1). The results of univariate (U) and multivariate (M) analysis showed that the change in STAT3 expression was statistically significant in evaluating the prognosis of patients with LGG (U: HR 1.887, ranging from 1.670 to 2.132, P < 0.001, Figure 3b; M: HR 1.314, ranging from 1.160 to 1.488, P < 0.001, Figure 3c). The area under curve results obtained by ROC curve analysis (1 year: 0.637, 3 years: 0.688, and 5 years: 0.714, Figure 3d) further indicated that measuring the expression of STAT3 is an effective way to evaluate the prognosis of patients with glioma. Moreover, we examined the STAT3 protein expression levels in LGG and normal tissues obtained from the HPA database, and the immunohistochemical staining images showed negative immunostaining of STAT3 in normal tissue (Figure 3e) and moderately positive immunostaining in LGG (Figure 3f).

Figure 2 
                  Prognostic survival analysis. (a)−(c) K−M survival analysis showed that the OS of LGG, TGCT, and SKCM in STAT3 high expression group was significantly different. (d) COX regression analysis showed that the OS of LGG and SKCM were significantly correlated with the expression of STAT3. (e)−(i) PFS of LGG, HNSC, SKCM, PRAD, and COAD were significantly different. (j) COX regression analysis showed that only LGG had significant difference in PFS and STAT3 expression.
Figure 2

Prognostic survival analysis. (a)−(c) K−M survival analysis showed that the OS of LGG, TGCT, and SKCM in STAT3 high expression group was significantly different. (d) COX regression analysis showed that the OS of LGG and SKCM were significantly correlated with the expression of STAT3. (e)−(i) PFS of LGG, HNSC, SKCM, PRAD, and COAD were significantly different. (j) COX regression analysis showed that only LGG had significant difference in PFS and STAT3 expression.

Figure 3 
                  Verification of CGGA: (a) result of K−M survival analysis, (b) results of univariate analysis, (c) results of multivariate analysis, (d) ROC curve, (e) immunohistochemical staining showed that STAT3 was negative staining in normal tissue, and (f) immunohistochemical staining showed that STAT3 was medium positive in LGG.
Figure 3

Verification of CGGA: (a) result of K−M survival analysis, (b) results of univariate analysis, (c) results of multivariate analysis, (d) ROC curve, (e) immunohistochemical staining showed that STAT3 was negative staining in normal tissue, and (f) immunohistochemical staining showed that STAT3 was medium positive in LGG.

Table 1

Result of COX regression analysis in CGGA

Cancer LGG
HR 1.887
HR.95L 1.670
HR.95H 2.132
P value <0.001

3.3 Immune correlation analysis

The survival analysis revealed that the expression of STAT3 in LGG is correlated with OS and PFS, and the difference is statistically significant. Therefore, we took LGG as an example for immune correlation analysis to further investigate the relationship between STAT3 expression and tumor immunity and the mechanism by which STAT3 affects prognosis of LGG patients. The results of the IME analysis revealed that the immune score (Figure 4a) and matrix score (Figure 4b) of LGG were positively correlated with the expression levels of STAT3 (P < 0.05). The results of immune cell infiltration showed that STAT3 was positively correlated with resting dendritic cells (DCs) (Figure 4c), eosinophils (Figure 4d), neutrophils (Figure 4e), M0 macrophages (Figure 4f), M1 macrophages (Figure 4g), CD4 memory resting T cells (Figure 4h), and CD8 T cells (Figure 4i) in LGG, but negatively correlated with activated mast cells (Figure 4j) and M2 macrophages (Figure 4k), for all the above P values were less than 0.05. We also analyzed the common immune checkpoints, and the correlation between immune checkpoints and STAT3 expression. As shown in Figure 4l, in LGG, the common immune checkpoint proteins CD274 (also known as PD-L1), PDCD1 (also known as PD-1), CTLA4, LAG3, and TIGIT showed a significant positive correlation with STAT3 expression.

Figure 4 
                  Results of immune correlation analysis: (a) immune score of LGG by IME analysis, (b) matrix score of LGG by IME analysis, (c)−(k) results of immune cell infiltration, and (l) correlations between immune checkpoints and STAT3 expression.
Figure 4

Results of immune correlation analysis: (a) immune score of LGG by IME analysis, (b) matrix score of LGG by IME analysis, (c)−(k) results of immune cell infiltration, and (l) correlations between immune checkpoints and STAT3 expression.

3.4 GSEA

Our GSEA identified 384 enrichment gene sets. According to the enrichment scores, the top ten upregulated pathways were mainly related to the immune response and included the following: immunoglobulin complex, immunoglobulin complex circulating, immunoglobulin receptor binding, phagocytosis recognition, humoral immune response mediated by circulating immunoglobulins, T cell tolerance induction, antigen binding, complement activation, opsonization, and Fc receptor mediated stimulatory signaling pathway (Figure 5a). The top ten downregulated pathways were mainly related to cell signal transduction and regulation of cell survival, proliferation, and metabolism and included the following: opioid receptor signaling pathway, amine binding, U2 snRNP, negative regulation of phosphatidylinositol 3 kinase (PI3K) signaling, regulation of guanylate cyclase activity, Gaba gated chloride ion channel activity, Gaba receptor complex, negative regulation of vascular associated smooth muscle cell migration, anchored component of synaptic vesicle membrane, and inhibitory extracellular ligand gated ion channel activity (Figure 5b).

Figure 5 
                  GSEA enrichment analysis: (a) first ten upregulated pathways and (b) first ten downregulated pathways.
Figure 5

GSEA enrichment analysis: (a) first ten upregulated pathways and (b) first ten downregulated pathways.

3.5 Genetic alteration analysis

As shown in the radar plot, the expression of STAT3 is negatively correlated with the TMB in BRCA, thyroid cancer, stomach adenocarcinoma (STAD), sarcoma (SARC), PRAD, pancreatic adenocarcinoma (PAAD), lung squamous cell carcinoma, liver hepatocellular carcinoma, and kidney renal papillary cell carcinoma, and positively correlated with thymoma (THYM), LGG, and COAD (Figure 6a, P < 0.05). The expression of STAT3 was negatively correlated with the MSI in STAD, SKCM, SARC, PRAD, PAAD, HNSC, esophageal carcinoma, and diffuse large B-cell lymphoma, but positively correlated with COAD (Figure 6b). Further analysis of the gene alterations in the CBioPortal database revealed, as shown in the total gene alteration histogram (Figure 6c), that embryonic tumors, endometrial carcinoma, and mature B-cell lymphoma are the top three cancers with the highest frequency of STAT3 alteration. In the “Mutations” module, the STAT3 mutation lollipop chart provides information on the mutation sites, and mutation types, as shown in Figure 6d, reveals that most of the STAT3 mutations occur in the SH2 domain, where both mutations in Y640F were “Missense”. The 3D structures of the STAT3 protein and the Y640F site are shown in Figure 6e. We further characterized the STAT3 gene mutation using the COSMIC database for STAT3 mutation analysis. The results of this analysis revealing the point mutations, copy number variation, overexpression or underexpression, and methylation of STAT3 in each group are shown in Table 2. The results in Table 2, which are sorted according to the point mutation frequency in descending order, show that vagina, penis, skin, hematopoietic, lymphoid, and liver tissues have the higher STAT3 mutation frequency. The “Missense substitution” of STAT3 accounted for the majority (51.88%) of mutations, as can be seen in the sector map of the mutation type (Figure 6f). The STAT3 protein sequence features map (Figure 6g) and its 3D structure map (Figure 6h) reveal that most of these missense substitutions are concentrated in the SH2 domain, which is consistent with the results of the analysis of the CBioPortal database.

Figure 6 
                  Genetic alteration analysis: (a) radar map shows the correlation between STAT3 expression and TMB, (b) radar map shows the correlation between STAT3 expression and MSI, (c) alteration frequency of ADAM12 in different tumors, (d) mutation lollipop chart provides information on the mutation sites and mutation types, (e) 3D structure of the STAT3 protein and the Y640F site, (f) an overview of the types of mutations observed that are curated by COSMIC, (g) protein sequence features of STAT3 protein curated by COSMIC, and (h) 3D structure and missense mutation frequency of STAT3 protein that are curated by COSMIC (the redder the color, the higher the missense mutation frequency).
Figure 6 
                  Genetic alteration analysis: (a) radar map shows the correlation between STAT3 expression and TMB, (b) radar map shows the correlation between STAT3 expression and MSI, (c) alteration frequency of ADAM12 in different tumors, (d) mutation lollipop chart provides information on the mutation sites and mutation types, (e) 3D structure of the STAT3 protein and the Y640F site, (f) an overview of the types of mutations observed that are curated by COSMIC, (g) protein sequence features of STAT3 protein curated by COSMIC, and (h) 3D structure and missense mutation frequency of STAT3 protein that are curated by COSMIC (the redder the color, the higher the missense mutation frequency).
Figure 6

Genetic alteration analysis: (a) radar map shows the correlation between STAT3 expression and TMB, (b) radar map shows the correlation between STAT3 expression and MSI, (c) alteration frequency of ADAM12 in different tumors, (d) mutation lollipop chart provides information on the mutation sites and mutation types, (e) 3D structure of the STAT3 protein and the Y640F site, (f) an overview of the types of mutations observed that are curated by COSMIC, (g) protein sequence features of STAT3 protein curated by COSMIC, and (h) 3D structure and missense mutation frequency of STAT3 protein that are curated by COSMIC (the redder the color, the higher the missense mutation frequency).

Table 2

Distribution of mutations across the primary tissue types that are curated by COSMIC

Point mutations Copy number variation Gene expression Methylation
Mutated (%) Tested Variant (%) Tested Regulated (%) (over/under) Tested Diff. methylated (%) Tested
Vagina 50 2
Penis 11.76 17
Skin 4.75 2,482 4.44/1.27 473
Hematopoietic and lymphoid 4.71 11,951 5.43/3.17 221
Liver 4.01 2,917 0.3 663 3.75/− 373 244
Endometrium 3.86 1,062 0.17 586 3.99/4.65 602 398
Vulva 3.33 30
Placenta 2.94 34
Prostate 2.48 3,102 0.21 949 2.01/0.4 498
Cervix 2.31 389 6.19/− 307
Urinary tract 2.26 1,285 0.25 399 2.45/− 408
Biliary tract 2.22 1,173
Large intestine 2.06 4,717 3.44/3.28 610 0.36 281
Upper aerodigestive tract 2.04 1,811 3.26/0.19 522 496
Ovary 1.92 1,460 0.29 684 1.5/0.38 266
Stomach 1.88 1,912 0.42 472 2.11/− 285
Pancreas 1.84 2,550 0.11 898 3.91/5.03 179
Meninges 1.52 198
Breast 1.45 5,445 0.27 1,492 4.35/2.17 1,104 707
NS 1.3 460
Esophagus 1.28 1,800 0.39 510 2.4/0.8 125
Salivary gland 1.12 267
Lung 1.07 5,619 0.4 1,006 4.32/1.37 1,019 717
Soft tissue 1.02 1,561 3.8/1.9 263
Small intestine 0.66 305
Thyroid 0.66 1,976 2.53/1.56 513 510
Kidney 0.56 2,870 0.1 995 3.67/3.83 600 2.53 513
Central nervous system 0.36 3,370 0.1 1,035 4.45/0.14 697
Adrenal gland 0.31 654 0.37 267 2.53/3.8 79
Autonomic ganglia 0.08 1,231
Bone 737
Eye 176
Fallopian tube 3
Gastrointestinal tract (site indeterminate) 67
Genital tract 126
Parathyroid 35
Perineum 1
Peritoneum 38
Pituitary 86
Pleura 356 1.15 87
Testis 458
Thymus 180
Uterine adnexa 4

4 Discussion

Increasing evidence shows that aberrant activation of STAT3 is involved in the proliferation and survival of tumor cells. The purpose of this study is to investigate the effects of genetic alterations of STAT3 and its expression on the development of cancer from the perspective of pan-cancer. First, we analyzed the differential expression between normal tissues and pan-cancer tissues, and found that the expression of STAT3 was different in different tumors and different tissues, and there were significant differences between most tumors and normal tissues. We also evaluated the effect of STAT3 expression on the prognosis of cancer patients by performing survival analysis, which revealed that, in LGG, the prognosis of OS and PFS in the STAT3 high expression group was worse than that in the low expression group. To some extent, this finding indicated that the high expression of STAT3 was related to the poor prognosis of gliomas. Therefore, we focused our study on the effect of STAT3 on gliomas. We further analyzed the glioma dataset downloaded from the CGGA database to verify the aforementioned results of STAT3 in glioma, and the results similarly suggested that the STAT3 high expression group had a poor prognosis. Additionally, univariate and multivariate regression analysis and ROC curve analysis indicated that STAT3 can be used as an independent prognostic factor of glioma with a certain degree of robustness. The importance of the IME in tumorigenesis and malignant progression is currently a hot research topic. Numerous studies have shown that the IME can promote the progression of cancer and lead to drug resistance, especially to cancer immunotherapy [34,35]. Therefore, we performed immune correlation analysis and GSEA to further investigate the mechanism by which activated STAT3 leads to poor cancer prognosis.

The immune correlation analysis found that the immune score and matrix score were higher in the group with high expression of STAT3. Further analysis of the infiltration of immune cell infiltration revealed that STAT3 was positively correlated with DCs, eosinophils, neutrophils, M0 macrophages, M1 macrophages, CD4 T cells, and CD8 T cells, and negatively correlated with activated mast cells and M2 macrophages. Previous studies have shown that abnormal STAT3 activation promotes the recruitment immune cells and impairs their function, resulting in immune escape of tumor cells [36]. First, aberrant activation of STAT3 in tumor cells plays an important role in the maturation of DCs. DCs are key antigen presenting cells of the immune system and play an important role in initiating the response of T cells to tumors, while immature DCs usually induce immune tolerance [37]. Overactivation of STAT3 in tumor cells can interfere with the antigen presentation process of DCs in various ways, such as decreasing the expression of BCL2 in DCs by inhibiting the expression of IL12 and TNF [3840], and inhibiting the maturation of DCs and innate immunity by negatively regulating the expression of interferon gamma inducible protein 10 and CC chemokine ligand 5 [41]. Moreover, since immature DCs cannot activate antigen-specific CD8+ T cells, the antitumor effect of CD8+ T cells will be decreased accordingly. In addition, other studies have found that STAT3 plays a major role in the expansion of regulatory T cells, and regulatory T cells can promote tumor progression by inhibiting the antitumor immune response mediated by TH1 CD4+ T and CD8+ T cells [40,42,43]. Furthermore, overactivation of STAT3 can promote tumor progression by inducing the polarization of type M2 macrophages and the expression of CD274 [44]. In summary, it is clear that overexpression of STAT3 can regulate in various ways the tumor IME, which besides generally promoting tumor progression, is related to poor prognosis, and is consistent with our findings. Through GSEA, we found that many immune response pathways are activated in the STAT3 high expression group, such as antigen–antibody binding, immunoglobulin complex formation, phagocytosis recognition, complement activation, etc. It is worth noting that among the downregulated pathway, the “negative regulation of PI3K signal transduction pathway” is downregulated. PI3K is a major regulatory factor of cancer, which can affect the progression of cancer by affecting the growth, proliferation, survival, and angiogenesis of tumor cells [4551]. The results of this study suggest that the negative regulation of PI3K signaling is downregulated in the group with high expression of STAT3. Previous studies have revealed that there is a certain correlation between STAT3 and the PI3K signaling pathway. First, Hart et al. identified the dependent transcription between PI3K and STAT3 by analyzing stable isotope labeling with amino acids in cell culture of PI3K transformed cells [52]. Subsequently, Hart et al. further studied the STAT3 and PI3K pathways and reported the following findings: (1) The p110 α-H1047R mutant transformed cells of PI3K showed increased tyrosine phosphorylation of STAT3. (2) The dominant-negative mutation of STAT3 interferes with PI3K-induced tumorigenesis. (3) GDC-0941, a specific inhibitor of PI3K, can reduce the phosphorylation level of STAT3. (4) In some human tumor cell lines, the enhanced phosphorylation of STAT3 is inhibited by PI3K and Tec kinase inhibitors. In summary, the study of the regulatory relationship between PI3K and STAT3 is of great significance to understand the development of cancer, and the inhibition of STAT3 expression may represent a breakthrough in the treatment of human tumors [53,54]. In order to develop new approaches to target STAT3 inhibitors, we also performed genetic alteration analysis. The TMB and MSI have been considered as predictive biomarkers of immune checkpoint blocking responses. Our study revealed that the expression of STAT3 is associated with TMB and MSI in many tumors, which suggests the possibility that, to some extent, STAT3 may serve as an immune checkpoint in these tumors [5456]. By examining the impact of genetic alterations in STAT3, we found that there are alterations in STAT3 in many tumors. In the mutational analysis of STAT3, our analysis showed that missense mutations in STAT3 account for the vast majority of mutations, and STAT3 mutations mostly occurred in the SH2 domain. The SH2 domain is the most conserved STAT domain, which drives transcription by binding to a specific phosphotyrosine motif that is essential for molecular activation and nuclear accumulation of phosphorylated STAT dimers. Even slight changes in the electronic or stereo structure of the SH2 domain can significantly change the activity of STAT3 [13,57,58]. To date, using high-throughput screening and a structure-based virtual screening system, a variety of small molecular peptides of STAT3 directly targeting the SH2 domain of STAT3 have been reported, which can also significantly change the activity of STAT3 [53]. For example, PY*LKTK (where Y* is the phosphorylated tyrosine) [59], S3I-M2001 [60], S3I-1757 [61], curcumin-proline [62], cryptotashinone [63], STA-21 [64], Stattic [65], S3I-201 [66], SD-36 [57], etc.

Undoubtedly, this study has certain limitations, including the following: this study is based on the analysis of multiple databases, and there are some differences in statistical analysis methods among different databases. More importantly, this is only a bioinformatics analysis study, and more genetic, experimental studies, and multicenter clinical studies are needed to verify the above inferences for more effective clinical application.

5 Conclusion

Overexpression of STAT3 promotes the growth and development of tumor cells by regulating the IME, which is significantly related to poor prognosis in cancer patients. Therefore, targeted inhibition of STAT3 expression or activity may have important research value for the development of combined immunotherapy and targeted therapy approaches for the treatment of cancer patients.


# Junyin Tan and Ronghao Feng contributed equally to this work.


  1. Funding information: There is no funding.

  2. Author contributions: Conceptualization, Ronghao Feng; methodology, Junyin Tan; validation and formal analysis, Ronghao Feng and Junyin Tan; writing and editing, Ronghao Feng and Junyin Tan. All authors have read and agreed to the published version of the manuscript.

  3. Conflict of interest: The authors declare that there is no conflict of interest regarding the publication of this article.

  4. Data availability statement: University of California, Santa Cruz (UCSC) Xena database: http://xena.ucsc.edu/; The Cancer Genome Atlas (TCGA): https://portal.gdc.cancer.gov/repository; GRCh38. P13: http://asia.ensembl.org/index.html; Chinese Glioma Genome Atlas (CGGA): http://www.cgga.org.cn/; UALCAN website: http://ualcan.path.uab.edu/analysis-prot.html; Molecular Signatures Database (MSigDB): http://www.gsea-msigdb.org/; CBioPortal database: http://www.cbioportal.org/; Catalogue Of Somatic Mutations In Cancer (COSMIC): http://www.sanger.ac.uk/cosmic.

References

[1] Qin JJ, Yan L, Zhang J, Zhang WD. STAT3 as a potential therapeutic target in triple negative breast cancer: a systematic review. J Exp Clin Cancer Res. 2019;38(1):195.10.1186/s13046-019-1206-zSearch in Google Scholar PubMed PubMed Central

[2] He W, Wu J, Shi J, Huo YM, Dai W, Geng J, et al. IL22RA1/STAT3 signaling promotes stemness and tumorigenicity in pancreatic cancer. Cancer Res. 2018;78(12):3293–305.10.1158/0008-5472.CAN-17-3131Search in Google Scholar PubMed

[3] Lu J, Tang Y, Cheng Y, Zhang G, Yip A, Martinka M, et al. ING4 regulates JWA in angiogenesis and their prognostic value in melanoma patients. Br J Cancer. 2013;109(11):2842–52.10.1038/bjc.2013.670Search in Google Scholar PubMed PubMed Central

[4] Moon EJ, Mello SS, Li CG, Chi JT, Thakkar K, Kirkland JG, et al. The HIF target MAFF promotes tumor invasion and metastasis through IL11 and STAT3 signaling. Nat Commun. 2021;12(1):4308.10.1038/s41467-021-24631-6Search in Google Scholar PubMed PubMed Central

[5] Zheng ZY, Chu MY, Lin W, Zheng YQ, Xu XE, Chen Y, et al. Blocking STAT3 signaling augments MEK/ERK inhibitor efficacy in esophageal squamous cell carcinoma. Cell Death Dis. 2022;13(5):496.10.1038/s41419-022-04941-3Search in Google Scholar PubMed PubMed Central

[6] Li Z, Li Y, Li N, Shen L, Liu A. Silencing GOLGA8B inhibits cell invasion and metastasis by suppressing STAT3 signaling pathway in lung squamous cell carcinoma. Clin Sci (Lond). 2022;136(11):895–909.10.1042/CS20220128Search in Google Scholar PubMed

[7] Yanagimura N, Takeuchi S, Fukuda K, Arai S, Tanimoto A, Nishiyama A, et al. STAT3 inhibition suppresses adaptive survival of ALK-rearranged lung cancer cells through transcriptional modulation of apoptosis. NPJ Precis Oncol. 2022;6(1):11.10.1038/s41698-022-00254-ySearch in Google Scholar PubMed PubMed Central

[8] Huynh J, Chand A, Gough D, Ernst M. Therapeutically exploiting STAT3 activity in cancer – using tissue repair as a road map. Nat Rev Cancer. 2019;19(2):82–96.10.1038/s41568-018-0090-8Search in Google Scholar PubMed

[9] Yu H, Pardoll D, Jove R. STATs in cancer inflammation and immunity: a leading role for STAT3. Nat Rev Cancer. 2009;9(11):798–809.10.1038/nrc2734Search in Google Scholar PubMed PubMed Central

[10] Johnson DE, O’Keefe RA, Grandis JR. Targeting the IL-6/JAK/STAT3 signalling axis in cancer. Nat Rev Clin Oncol. 2018;15(4):234–48.10.1038/nrclinonc.2018.8Search in Google Scholar PubMed PubMed Central

[11] Kumar V, Cheng P, Condamine T, Mony S, Languino LR, McCaffrey JC, et al. CD45 Phosphatase inhibits STAT3 transcription factor activity in myeloid cells and promotes tumor-associated macrophage differentiation. Immunity. 2016;44(2):303–15.10.1016/j.immuni.2016.01.014Search in Google Scholar PubMed PubMed Central

[12] Akira S, Nishio Y, Inoue M, Wang XJ, Wei S, Matsusaka T, et al. Molecular cloning of APRF, a novel IFN-stimulated gene factor 3 p91-related transcription factor involved in the gp130-mediated signaling pathway. Cell. 1994;77(1):63–71.10.1016/0092-8674(94)90235-6Search in Google Scholar PubMed

[13] Sgrignani J, Garofalo M, Matkovic M, Merulla J, Catapano CV, Cavalli A. Structural biology of STAT3 and its implications for anticancer therapies development. Int J Mol Sci. 2018;19(6):1591.10.3390/ijms19061591Search in Google Scholar PubMed PubMed Central

[14] Wang H, Huo X, Yang XR, He J, Cheng L, Wang N, et al. STAT3-mediated upregulation of lncRNA HOXD-AS1 as a ceRNA facilitates liver cancer metastasis by regulating SOX4. Mol Cancer. 2017;16(1):136.10.1186/s12943-017-0680-1Search in Google Scholar PubMed PubMed Central

[15] Yang Y, Jin G, Liu H, Liu K, Zhao J, Chen X, et al. Metformin inhibits esophageal squamous cell carcinoma-induced angiogenesis by suppressing JAK/STAT3 signaling pathway. Oncotarget. 2017;8(43):74673–87.10.18632/oncotarget.20341Search in Google Scholar PubMed PubMed Central

[16] Huang X, Meng B, Iqbal J, Ding BB, Perry AM, Cao W, et al. Activation of the STAT3 signaling pathway is associated with poor survival in diffuse large B-cell lymphoma treated with R-CHOP. J Clin Oncol. 2013;31(36):4520–8.10.1200/JCO.2012.45.6004Search in Google Scholar PubMed PubMed Central

[17] Wang LL, Luo J, He ZH, Liu YQ, Li HG, Xie D, et al. STEAP3 promotes cancer cell proliferation by facilitating nuclear trafficking of EGFR to enhance RAC1-ERK-STAT3 signaling in hepatocellular carcinoma. Cell Death Dis. 2021;12(11):1052.10.1038/s41419-021-04329-9Search in Google Scholar PubMed PubMed Central

[18] Zerdes I, Wallerius M, Sifakis EG, Wallmann T, Betts S, Bartish M, et al. STAT3 activity promotes programmed-death ligand 1 expression and suppresses immune responses in breast cancer. Cancers (Basel). 2019;11(10):1479.10.3390/cancers11101479Search in Google Scholar PubMed PubMed Central

[19] Kim SJ, Garcia-Recio S, Creighton CJ, Perou CM, Rosen JM. Alterations in Wnt- and/or STAT3 signaling pathways and the immune microenvironment during metastatic progression. Oncogene. 2019;38(31):5942–58.10.1038/s41388-019-0852-0Search in Google Scholar PubMed PubMed Central

[20] Li Y, Song Z, Han Q, Zhao H, Pan Z, Lei Z, et al. Targeted inhibition of STAT3 induces immunogenic cell death of hepatocellular carcinoma cells via glycolysis. Mol Oncol. 2022;16(15):2861–80.10.1002/1878-0261.13263Search in Google Scholar PubMed PubMed Central

[21] Chung CC, Huang TY, Chu HR, De Luca R, Candelotti E, Huang CH, et al. Heteronemin and tetrac derivatives suppress non-small cell lung cancer growth via ERK1/2 inhibition. Food Chem Toxicol. 2022;161:112850.10.1016/j.fct.2022.112850Search in Google Scholar PubMed

[22] Liu C, Liu H, Wen Y, Huang H, Hao J, Lv Y, et al. Aspernolide A inhibits the proliferation of human laryngeal carcinoma cells through the mitochondrial apoptotic and STAT3 signaling pathways. Molecules. 2019;24(6):1074.10.3390/molecules24061074Search in Google Scholar PubMed PubMed Central

[23] Pejin B, Iodice C, Tommonaro G, Bogdanovic G, Kojic V, De, Rosa S. Further in vitro evaluation of cytotoxicity of the marine natural product derivative 4’-leucine-avarone. Nat Prod Res. 2014;28(5):347–50.10.1080/14786419.2013.863201Search in Google Scholar PubMed

[24] Haeussler M, Zweig AS, Tyner C, Speir ML, Rosenbloom KR, Raney BJ, et al. The UCSC genome browser database: 2019 update. Nucleic Acids Res. 2019;47(D1):D853–8.10.1093/nar/gky1095Search in Google Scholar PubMed PubMed Central

[25] Weinstein JN, Collisson EA, Mills GB, Shaw KR, Ozenberger BA, Ellrott K, et al. The Cancer Genome Atlas Pan-Cancer analysis project. Nat Genet. 2013;45(10):1113–20.10.1038/ng.2764Search in Google Scholar PubMed PubMed Central

[26] Cunningham F, Allen JE, Allen J, Alvarez-Jarreta J, Amode MR, Armean IM, et al. Ensembl 2022. Nucleic Acids Res. 2022;50(D1):D988–95.10.1093/nar/gkab1049Search in Google Scholar PubMed PubMed Central

[27] Zhao Z, Zhang KN, Wang Q, Li G, Zeng F, Zhang Y, et al. Chinese glioma genome atlas (CGGA): a comprehensive resource with functional genomic data from chinese glioma patients. Genom Proteom Bioinform. 2021;19(1):1–12.10.1016/j.gpb.2020.10.005Search in Google Scholar PubMed PubMed Central

[28] Chandrashekar DS, Bashel B, Balasubramanya SAH, Creighton CJ, Ponce-Rodriguez I, Chakravarthi B, et al. UALCAN: a portal for facilitating tumor subgroup gene expression and survival analyses. Neoplasia. 2017;19(8):649–58.10.1016/j.neo.2017.05.002Search in Google Scholar PubMed PubMed Central

[29] Newman AM, Liu CL, Green MR, Gentles AJ, Feng W, Xu Y, et al. Robust enumeration of cell subsets from tissue expression profiles. Nat Methods. 2015;12(5):453–7.10.1038/nmeth.3337Search in Google Scholar PubMed PubMed Central

[30] Subramanian A, Tamayo P, Mootha VK, Mukherjee S, Ebert BL, Gillette MA, et al. Gene set enrichment analysis: a knowledge-based approach for interpreting genome-wide expression profiles. Proc Natl Acad Sci U S A. 2005;102(43):15545–50.10.1073/pnas.0506580102Search in Google Scholar PubMed PubMed Central

[31] Gao J, Aksoy BA, Dogrusoz U, Dresdner G, Gross B, Sumer SO, et al. Integrative analysis of complex cancer genomics and clinical profiles using the cBioPortal. Sci Signal. 2013;6(269):pl1.10.1126/scisignal.2004088Search in Google Scholar PubMed PubMed Central

[32] Cerami E, Gao J, Dogrusoz U, Gross BE, Sumer SO, Aksoy BA, et al. The cBio cancer genomics portal: an open platform for exploring multidimensional cancer genomics data. Cancer Discov. 2012;2(5):401–4.10.1158/2159-8290.CD-12-0095Search in Google Scholar PubMed PubMed Central

[33] Bamford S, Dawson E, Forbes S, Clements J, Pettett R, Dogan A, et al. The COSMIC (Catalogue of Somatic Mutations in Cancer) database and website. Br J Cancer. 2004;91(2):355–8.10.1038/sj.bjc.6601894Search in Google Scholar PubMed PubMed Central

[34] Martin JD, Cabral H, Stylianopoulos T, Jain RK. Improving cancer immunotherapy using nanomedicines: progress, opportunities and challenges. Nat Rev Clin Oncol. 2020;17(4):251–66.10.1038/s41571-019-0308-zSearch in Google Scholar PubMed PubMed Central

[35] Phuengkham H, Ren L, Shin IW, Lim YT. Nanoengineered immune niches for reprogramming the immunosuppressive tumor microenvironment and enhancing cancer immunotherapy. Adv Mater. 2019;31(34):e1803322.10.1002/adma.201803322Search in Google Scholar PubMed

[36] Jung JE, Kim HS, Lee CS, Shin YJ, Kim YN, Kang GH, et al. STAT3 inhibits the degradation of HIF-1alpha by pVHL-mediated ubiquitination. Exp Mol Med. 2008;40(5):479–85.10.3858/emm.2008.40.5.479Search in Google Scholar PubMed PubMed Central

[37] Giovanelli P, Sandoval TA, Cubillos-Ruiz JR. Dendritic cell metabolism and function in tumors. Trends Immunol. 2019;40(8):699–718.10.1016/j.it.2019.06.004Search in Google Scholar PubMed

[38] Wang T, Niu G, Kortylewski M, Burdelya L, Shain K, Zhang S, et al. Regulation of the innate and adaptive immune responses by Stat-3 signaling in tumor cells. Nat Med. 2004;10(1):48–54.10.1038/nm976Search in Google Scholar PubMed

[39] Szulc-Dąbrowska L, Struzik J, Cymerys J, Winnicka A, Nowak Z, Toka FN, et al. The in vitro inhibitory effect of ectromelia virus infection on innate and adaptive immune properties of GM-CSF-derived bone marrow cells is mouse strain-independent. Front Microbiol. 2017;8:2539.10.3389/fmicb.2017.02539Search in Google Scholar PubMed PubMed Central

[40] Kortylewski M, Kujawski M, Wang T, Wei S, Zhang S, Pilon-Thomas S, et al. Inhibiting Stat3 signaling in the hematopoietic system elicits multicomponent antitumor immunity. Nat Med. 2005;11(12):1314–21.10.1038/nm1325Search in Google Scholar PubMed

[41] Zou W. Regulatory T cells, tumour immunity and immunotherapy. Nat Rev Immunol. 2006;6(4):295–307.10.1038/nri1806Search in Google Scholar PubMed

[42] Kortylewski M, Xin H, Kujawski M, Lee H, Liu Y, Harris T, et al. Regulation of the IL-23 and IL-12 balance by Stat3 signaling in the tumor microenvironment. Cancer Cell. 2009;15(2):114–23.10.1016/j.ccr.2008.12.018Search in Google Scholar PubMed PubMed Central

[43] Matsumura Y, Kobayashi T, Ichiyama K, Yoshida R, Hashimoto M, Takimoto T, et al. Selective expansion of foxp3-positive regulatory T cells and immunosuppression by suppressors of cytokine signaling 3-deficient dendritic cells. J Immunol. 2007;179(4):2170–9.10.4049/jimmunol.179.4.2170Search in Google Scholar PubMed

[44] Fujiwara Y, Takeya M, Komohara Y. A novel strategy for inducing the antitumor effects of triterpenoid compounds: blocking the protumoral functions of tumor-associated macrophages via STAT3 inhibition. Biomed Res Int. 2014;2014:348539.10.1155/2014/348539Search in Google Scholar PubMed PubMed Central

[45] Yang J, Nie J, Ma X, Wei Y, Peng Y, Wei X. Targeting PI3K in cancer: mechanisms and advances in clinical trials. Mol Cancer. 2019;18(1):26.10.1186/s12943-019-0954-xSearch in Google Scholar PubMed PubMed Central

[46] Fresno Vara JA, Casado E, de Castro J, Cejas P, Belda-Iniesta C, González-Barón M. PI3K/Akt signalling pathway and cancer. Cancer Treat Rev. 2004;30(2):193–204.10.1016/j.ctrv.2003.07.007Search in Google Scholar PubMed

[47] Alzahrani AS. PI3K/Akt/mTOR inhibitors in cancer: at the bench and bedside. Semin Cancer Biol. 2019;59:125–32.10.1016/j.semcancer.2019.07.009Search in Google Scholar PubMed

[48] Fattahi S, Amjadi-Moheb F, Tabaripour R, Ashrafi GH, Akhavan-Niaki H. PI3K/AKT/mTOR signaling in gastric cancer: epigenetics and beyond. Life Sci. 2020;262:118513.10.1016/j.lfs.2020.118513Search in Google Scholar PubMed

[49] Zoncu R, Efeyan A, Sabatini DM. mTOR: from growth signal integration to cancer, diabetes and ageing. Nat Rev Mol Cell Biol. 2011;12(1):21–35.10.1038/nrm3025Search in Google Scholar PubMed PubMed Central

[50] Engelman JA, Luo J, Cantley LC. The evolution of phosphatidylinositol 3-kinases as regulators of growth and metabolism. Nat Rev Genet. 2006;7(8):606–19.10.1038/nrg1879Search in Google Scholar PubMed

[51] Cantley LC. The phosphoinositide 3-kinase pathway. Science. 2002;296(5573):1655–7.10.1126/science.296.5573.1655Search in Google Scholar PubMed

[52] Hart JR, Liao L, Ueno L, Yates III JR, Vogt PK. Protein expression profiles of C3H 10T1/2 murine fibroblasts and of isogenic cells transformed by the H1047R mutant of phosphoinositide 3-kinase (PI3K). Cell Cycle. 2011;10(6):971–6.10.4161/cc.10.6.15077Search in Google Scholar PubMed PubMed Central

[53] Vogt PK, Hart JR. PI3K and STAT3: a new alliance. Cancer Discov. 2011;1(6):481–6.10.1158/2159-8290.CD-11-0218Search in Google Scholar PubMed PubMed Central

[54] Chan TA, Yarchoan M, Jaffee E, Swanton C, Quezada SA, Stenzinger A, et al. Development of tumor mutation burden as an immunotherapy biomarker: utility for the oncology clinic. Ann Oncol. 2019;30(1):44–56.10.1093/annonc/mdy495Search in Google Scholar PubMed PubMed Central

[55] Yu HY, Li CP, Huang YH, Hsu SJ, Wang YP, Hsieh YC, et al. Microsatellite instability, epstein-barr virus, and programmed cell death ligand 1 as predictive markers for immunotherapy in gastric cancer. Cancers (Basel). 2022;14(1):218.10.3390/cancers14010218Search in Google Scholar PubMed PubMed Central

[56] Alam MR, Abdul-Ghafar J, Yim K, Thakur N, Lee SH, Jang HJ, et al. Recent applications of artificial intelligence from histopathologic image-based prediction of microsatellite instability in solid cancers: a systematic review. Cancers (Basel). 2022;14(1):2590.10.3390/cancers14112590Search in Google Scholar PubMed PubMed Central

[57] Bai L, Zhou H, Xu R, Zhao Y, Chinnaswamy K, McEachern D, et al. A potent and selective small-molecule degrader of STAT3 achieves complete tumor regression in vivo. Cancer Cell. 2019;36(5):498–511.e417.10.1016/j.ccell.2019.10.002Search in Google Scholar PubMed PubMed Central

[58] de Araujo ED, Orlova A, Neubauer HA, Bajusz D, Seo HS, Dhe-Paganon S, et al. Structural implications of STAT3 and STAT5 SH2 domain mutations. Cancers (Basel). 2019;11(11):1757.10.3390/cancers11111757Search in Google Scholar PubMed PubMed Central

[59] Turkson J, Ryan D, Kim JS, Zhang Y, Chen Z, Haura E, et al. Phosphotyrosyl peptides block Stat3-mediated DNA binding activity, gene regulation, and cell transformation. J Biol Chem. 2001;276(48):45443–55.10.1074/jbc.M107527200Search in Google Scholar PubMed

[60] Siddiquee KA, Gunning PT, Glenn M, Katt WP, Zhang S, Schrock C, et al. An oxazole-based small-molecule Stat3 inhibitor modulates Stat3 stability and processing and induces antitumor cell effects. ACS Chem Biol. 2007;2(12):787–98.10.1021/cb7001973Search in Google Scholar PubMed

[61] Zhang X, Sun Y, Pireddu R, Yang H, Urlam MK, Lawrence HR, et al. A novel inhibitor of STAT3 homodimerization selectively suppresses STAT3 activity and malignant transformation. Cancer Res. 2013;73(6):1922–33.10.1158/0008-5472.CAN-12-3175Search in Google Scholar PubMed PubMed Central

[62] Kumar A, Bora U. Molecular docking studies on inhibition of Stat3 dimerization by curcumin natural derivatives and its conjugates with amino acids. Bioinformation. 2012;8(20):988–93.10.6026/97320630008988Search in Google Scholar PubMed PubMed Central

[63] Shin DS, Kim HN, Shin KD, Yoon YJ, Kim SJ, Han DC, et al. Cryptotanshinone inhibits constitutive signal transducer and activator of transcription 3 function through blocking the dimerization in DU145 prostate cancer cells. Cancer Res. 2009;69(1):193–202.10.1158/0008-5472.CAN-08-2575Search in Google Scholar PubMed

[64] 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 U S A. 2005;102(13):4700–5.10.1073/pnas.0409894102Search in Google Scholar PubMed PubMed Central

[65] Schust J, Sperl B, Hollis A, Mayer TU, Berg T. Stattic: a small-molecule inhibitor of STAT3 activation and dimerization. Chem Biol. 2006;13(11):1235–42.10.1016/j.chembiol.2006.09.018Search in Google Scholar PubMed

[66] Fletcher S, Page BD, Zhang X, Yue P, Li ZH, Sharmeen S, et al. Antagonism of the Stat3-Stat3 protein dimer with salicylic acid based small molecules. ChemMedChem. 2011, 6(8):1459–70.10.1002/cmdc.201100194Search in Google Scholar PubMed PubMed Central

Received: 2022-09-04
Revised: 2023-03-30
Accepted: 2023-08-08
Published Online: 2023-09-16

© 2023 the author(s), published by De Gruyter

This work is licensed under the Creative Commons Attribution 4.0 International License.

Downloaded on 29.1.2025 from https://www.degruyter.com/document/doi/10.1515/med-2023-0792/html
Scroll to top button