Next Article in Journal
The MEK1/2 Inhibitor ATR-002 (Zapnometinib) Synergistically Potentiates the Antiviral Effect of Direct-Acting Anti-SARS-CoV-2 Drugs
Previous Article in Journal
Biopolymeric Prodrug Systems as Potential Antineoplastic Therapy
Previous Article in Special Issue
Scope and Limitations of Current Antibiotic Therapies against Helicobacter pylori: Reviewing Amoxicillin Gastroretentive Formulations
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Role of Nrf2, STAT3, and Src as Molecular Targets for Cancer Chemoprevention

1
Department of Pharmacy, Faculty of Life and Environmental Sciences, University of Peshawar, Peshawar 25120, Pakistan
2
Department of Pharmacy, CECOS University, Peshawar 25000, Pakistan
3
BK21 FOUR KNU Creative BioResearch Group, School of Life Sciences, Kyungpook National University, Daegu 41566, Korea
*
Author to whom correspondence should be addressed.
Pharmaceutics 2022, 14(9), 1775; https://doi.org/10.3390/pharmaceutics14091775
Submission received: 4 June 2022 / Revised: 23 July 2022 / Accepted: 22 August 2022 / Published: 25 August 2022
(This article belongs to the Special Issue Current and Future Cancer Chemoprevention Strategies)

Abstract

:
Cancer is a complex and multistage disease that affects various intracellular pathways, leading to rapid cell proliferation, angiogenesis, cell motility, and migration, supported by antiapoptotic mechanisms. Chemoprevention is a new strategy to counteract cancer; to either prevent its incidence or suppress its progression. In this strategy, chemopreventive agents target molecules involved in multiple pathways of cancer initiation and progression. Nrf2, STAT3, and Src are promising molecular candidates that could be targeted for chemoprevention. Nrf2 is involved in the expression of antioxidant and phase II metabolizing enzymes, which have direct antiproliferative action as well as indirect activities of reducing oxidative stress and eliminating carcinogens. Similarly, its cross-talk with NF-κB has great anti-inflammatory potential, which can be utilized in inflammation-induced/associated cancers. STAT3, on the other hand, is involved in multiple pathways of cancer initiation and progression. Activation, phosphorylation, dimerization, and nuclear translocation are associated with tumor cell proliferation and angiogenesis. Src, being the first oncogene to be discovered, is important due to its convergence with many upstream stimuli, its cross-talk with other potential molecular targets, such as STAT3, and its ability to modify the cell cytoskeleton, making it important in cancer invasion and metastasis. Therefore, the development of natural/synthetic molecules and/or design of a regimen that can reduce oxidative stress and inflammation in the tumor microenvironment and stop multiple cellular targets in cancer to stop its initiation or retard its progression can form newer chemopreventive agents.

1. Introduction

Cancer chemoprevention is the use of various means (natural and synthetic) to suppress, prevent, or delay cancer by inhibiting tumor development at the initial stages or by retarding the rate at which it grows to delay the malignant tendencies and properties of tumors [1,2].
At the cellular level, chemoprevention involves the halt or retardation of various molecular pathways at any or all three stages of cancer, i.e., initiation, promotion, and progression [3]. Such agents may block DNA damage during initiation or reduce free radical-induced damage. Furthermore, their potent antioxidant activities and ability to repair DNA may be additional mechanisms of chemoprevention [4]. Chemopreventive agents can also have profound effects on the progressive stages of cancer owing to their antiproliferative, anti-angiogenic, and antiapoptotic effects [3,5,6]. These actions are exerted by affecting various cellular signaling pathways, such as Nrf2, NF-κB, STAT3, and Src [5,7,8].

2. Nuclear Factor E2-Related Factor 2 (Nrf2)

2.1. Role and Significance of Nrf2 in Cancer

Nrf2 is an important transcription factor that regulates cancer gene expression and inflammation [9]. In the resting or inactive state, it binds to Keap1. In the bound state, it undergoes constant ubiquitination by Cullin 3-dependent E3 ubiquitin ligases. When activated, Keap1 undergoes modification, and Nrf2 is released from the complex. It enters the nucleus, dimerizes with the Maf proteins, and binds to the antioxidant response elements of the target genes [10] (Figure 1). Activation of Nrf2 may occur via MAPK, such as p38 and JNK [11]. Nrf2 is responsible for the expression of many enzymes involved in phase II metabolism (making xenobiotics more water soluble and readily excretable) and protects cells from oxidative damage. These enzymes include γ-glutamylcysteine synthetase (γGCS), NADPH quinone oxidoreductase 1 (NQO1), and heme oxygenase 1 (HO-1) [12]. The γ-GCS is involved in the biosynthesis of glutathione, which is one of the major antioxidants in human cells. NQO1 is responsible for generating reduced forms of ubiquinone and tocopherols, making them effective in eliminating free radicals. HO-1 plays a significant antioxidant role in maintaining cellular homeostasis against reactive oxygen species (ROS) [13].

2.2. Targeting Nrf2 Signaling for Cancer Chemoprevention

Nrf2 is an effective molecular target to induce chemopreventive effects. Many activators of the Nrf2 pathway have been shown to enhance the defensive capacity of cells against cancer, inflammation, and oxidative damage (Figure 2) [14,15,16,17].
A study by Lida et al. (2007) showed that Nrf2 null mice are more susceptible to developing chemically induced carcinoma of the urinary bladder [18,19]. Ginnalin A, a natural phenolic compound, has been reported to possess chemopreventive properties in human colon cancer via the activation of Nrf2 signaling. This study revealed that the compound suppressed the proliferation of cancer cells by arresting the cell cycle in the S phase. The compound also increased the translocation and expression of Nrf2, along with other antioxidant genes such as HO-1 and NQO1 [20]. Lycopene, a phytoconstituent in tomatoes, has been reported to attenuate the formation of tumors and their proliferative capacity. Lycopene was found to stimulate nuclear translocation of Nrf2 and the expression of various antioxidant enzymes. The study also reported that the release of Nrf2 from the complex was due to the enhanced expression of p62, which caused degradation of Keap1 [21]. Miconazole has also been reported to increase the expression of autophagy protein p62 in bladder carcinoma. The expression of p62 directly correlates with Nrf2 activation and decreases Keap1 expression [22].

2.3. Correlation between Nuclear Factor κB (NF κB) and Nrf2 Expression in Cancer

NF-κB is a transcription factor involved in regulating inflammation and cell proliferation [23]. It is also believed to serve as a link between inflammation and cancer [24]. In the resting state, it binds to the cytoplasmic IκBs. Proteasomal degradation of IκBs induced through their phosphorylation by kinases causes the release of NF-κB, its translocation to the nucleus, binding to specific response elements, and influences gene expression [25]. It also increases the expression of IL6, IL1β, TNFα, COX2, iNOS, and other cytokines. Furthermore, genes affected by NF-κB are involved in cell proliferation, angiogenesis, and metastasis [23,26,27]. NF-κB has also been linked to the emergence of resistance to endocrine therapy in breast cancer, suggesting that modulation of inflammation via this pathway is a significant requirement for cancer management [28]. Therefore, abnormal activation of this pathway may lead to the transition from inflammation to tumorigenesis. Likewise, the inhibition of inflammation may serve as an important mechanism to prevent cancer.
Nrf2 has been employed as a target for designing anti-inflammatory drugs for various chronic disorders such as multiple sclerosis. Dimethyl fumarate, used as a disease-modifying agent in multiple sclerosis, targets Nrf2. This agent is used because of its potent anti-inflammatory effect, which causes symptomatic relief in multiple sclerosis [29]. One study investigated the effects of sappanone A (Caesalpinia sappan) in macrophages. Sappanone A reduces nitric oxide (NO), prostaglandin E2 (PGE2), and IL-6. Isoflavanone also protects C57BL/6 mice from LPS-induced oxidative damage. Sappanone increased the mRNA expression of Nrf2 and its target genes NQO1 and HO-1. This study demonstrated that knockdown of Nrf2 inhibited the induction of HO-1 by sappanone A. Sappanone A also reduced LPS-induced NF-κB activation. The authors concluded that sappanone A exerts its anti-inflammatory effects by regulating Nrf2 and NF-κB signaling. This study links these two pathways, which can be extrapolated to the connection between inflammation and cancer [30]. Another study identified a sesquiterpene coumarin strigoid that could induce Nrf2 and suppress NF-κB simultaneously. Hence, such natural organic compounds may also be molecules of interest that can target both pathways to suppress inflammation in cancer effectively. Moreover, they could prove to be effective chemopreventive agents as monotherapy or combination therapy [31]. Another study evaluated the effects of xanthohumol and phenethyl isothiocyanate on pancreatic cancer cells. The combination of both agents had better anti-inflammatory and anticancer properties than the individual agents. A combination of the two inhibited the binding of NF-κB to DNA by 47–60%. Other effects observed included enhanced expression of Nrf2, NQO1, and superoxide dismutase (SOD). Thus, the combination reduced oxidative damage, angiogenesis, and proliferation of pancreatic cells. This study suggests the use of these two agents as good chemopreventive agents to delay the progressive stages of cancer [32]. Moringa isothiocyanate 1 increases the nuclear accumulation of Nrf2 and reduces the nuclear translocation of NF-κB, both leading to a reduction in inflammation and oxidative stress [33]. Another study reported that Nrf2-mediated stimulation of antioxidant pathways causes a reduction in ROS, which is a stimulant for NF-κB activation. Hence activation of Nrf2 leads to suppression of NF-κB [34]. Therefore, it is pertinent to mention that agents targeting Nrf2 and NF-κB can prove to be a very useful means of chemoprevention by targeting the tumor directly and inhibiting chronic inflammation associated with cancer [26,35]. The same correlation between Nrf2 and NF-κB is shown in Figure 3.

2.4. Targeting Nrf2 in Colitis-Associated Colon Cancer

Colon cancer is a huge health burden, as it is one of the most commonly diagnosed cancers in both men and women [36,37]. It has also been reported that chronic inflammation of the gastrointestinal tract due to infections, aberrant immune responses, or environmental factors can promote the progression of colon cancer [38].
In inflammatory bowel disease (IBD), the luminal side of the epithelial cells is disrupted by inflammation of the intestines by cytokines and chemokines [39]. Continuous and long-term severe IBD increases the risk of colorectal cancer [40,41]. Certain proinflammatory cytokines, such as IL-23, produced in excessive quantities in IBD, can also lead to the aggravation of colon cancer [42,43,44]. Various studies have reported that inflammation of the gastrointestinal tract in various forms helps in the progression of cancer [45,46]. In experimental settings, IBD can be chemically induced in murine models using dextran sodium sulfate (DSS). DSS is toxic to epithelial cells, causes erosion, and reduces membrane integrity [47,48]. The known cause of epithelial damage due to DSS is the overproduction of ROS [49,50,51]. Nrf2 regulates the expression of several cellular antioxidant enzymes to counteract the effects of ROS [52]. It has been shown experimentally that Nrf2 null mice have a greater tendency to develop DSS-induced IBD than wild-type mice. Moreover, the levels of inflammatory cytokines were higher in Nrf2 null mice. This greater propensity for epithelial damage is attributed to a reduction in phase II detoxifying enzymes, whose expression is regulated by Nrf2 [53,54]. It has been postulated that inflammation is one of the prominent characteristics of the tumor microenvironment in colon cancer [55,56]. A study demonstrated that Nrf2 knockout mice have a greater number and size of tumors induced by Azoxymethane or DSS. It reported that the knockout mice had 80% adenocarcinoma lesions compared to 29% in wild type. This shows that Nrf2 exerts a protective role in colitis-associated colon cancer [56].
Many phytoconstituents have been shown to exert beneficial effects in chemically induced colitis through Nrf2 activation. One study demonstrated that the pretreatment of C57BL/6 mice with the isothiocyanate compound sulforaphane (25 mg/kg) reduced the severity of DSS-induced colitis. The loss of body weight and disease activity index were lower than those in the untreated group. The pretreated group also had a longer colon, reduced expression of proinflammatory markers, and increased expression of Nrf2-related genes [57]. Another study has reported the effects of coenzyme Q10 on colitis. Researchers found that Q10 can potentially protect the colon in an Nrf2-dependent manner. They reported that the suppression of colitis and a reduction in the levels of inflammatory markers are due to activation of the Nrf2-dependent HO-1 pathway [58]. A study also demonstrated that the use of agents that cause the breakdown of the Nrf2-Keap1 complex and facilitate its nuclear translocation in colon cells (NCM460) exerted cytoprotective effects against ulcerative colitis. In this study, CPUY192018 was used as an inhibitor of the Nrf2-Keap1 complex [52].

3. Signal Transducers and Activators of Transcription-3 (STAT3)

3.1. Significance of STAT3

STAT3 is one of the main regulators of the cell cycle and is involved in its differentiation, proliferation, apoptosis, and angiogenesis [59,60]. Cancer cells undergoing rapid proliferation require persistent STAT3 activation. In gastric carcinoma, IL-26-induced abnormal STAT3 activation causes upregulation of the antiapoptotic proteins, such as Bcl2, which leads to uncontrolled cell proliferation [61]. Its persistent activation has also been associated with endometrial cell proliferation in uterine cancer [62]. The same has been reported for bladder, colon, and renal carcinoma [63,64]. Cancer cells can speed up glycolysis while downregulating mitochondrial respiration (Kreb’s cycle). This leads to a greater conversion of pyruvic acid to lactic acid (Warburg effect). Increased lactate is indicative of hypoxia leading to induction of HIF-1a. It induces pyruvate kinase leading to greater pyruvate production for further conversion to lactate and consequent HIF-1a release. Pyruvate kinase induction has been associated with greater STAT3 activation [65]. STAT3 is responsible for transcriptional activation of the VEGF gene. It is also associated with increased expression of MMP-2 and VEGF, which are associated with the increased invasive and metastatic transformation of cancers [66,67]. It plays a significant role in G1 -S phase transition by upregulation of cyclin D1, Cdc 25A, and downregulation of p21. STAT3 activation leading to change in expressions and concentrations of various cytokines and transcription factors leading to invasive carcinogenesis is summarized in Figure 4.

3.2. Strategies to Target STAT3

Chemoprevention strategies can be devised using STAT3 as a target molecule. The persistent activation of STAT3 in cancer can be counteracted by various mechanisms such as (i) inhibition of receptors leading to STAT3 activation; (ii) inhibition of ligand binding to STAT3 activating receptor; (iii) inhibition of the phosphorylation of the cytoplasmic tail of the receptor; (iv) inhibition of JAK kinases to cease STAT3 dimerization; and (v) prevention of its nuclear translocation and binding to specific response elements on DNA [68,69,70].
STAT3 is also a downstream cellular mediator of cancer and angiogenesis, induced by IL-6 and EGFR. Various tyrosine kinase inhibitors (AG490 and AZD1480) are currently being studied to block the JAK-STAT3 pathway, thereby inhibiting tumorigenesis and angiogenesis [71,72]. Novel compounds that can block STAT3 dimerization can also be effective chemopreventive agents. Garcinol, a natural compound isolated from Garcinia indica, has been reported to suppress STAT3 signaling in hepatocellular cancer. The mechanism of STAT3 inhibition involves the binding of garcinol to the SH2 domain of STAT3 and inhibition of its dimerization. Additionally, it inhibits STAT3 acetylation, leading to impaired binding to DNA. This results in the suppression of many target genes involved in cell proliferation and angiogenesis. Moreover, an increase in apoptosis was observed. Hence, garcinol and its semisynthetic derivatives may be effective future treatments for chemoprevention and chemotherapy [73]. A recently published study has reported the effects of STAT3 inhibition by ODZ10117. The molecule inhibits dimerization by binding to its SH2 domain. The net result observed was inhibition of phosphorylation and nuclear translocation. This inhibitory effect was stronger than other known STAT3 inhibitors such as STA-21. ODZ10117 suppresses tumor cell migration. It also induced apoptosis and reduced cell invasiveness. Overall, this molecule and its analogs can potentially be used as chemopreventive agents that can delay cancer progression [74]. The functional phosphorylation sites in STAT3 are tyrosine 705 and serine 727. Most natural and synthetic inhibitors discovered/developed block STAT3 phosphorylation at tyrosine 705. A summary of inhibitors of STAT3 phosphorylation is given in Table 1.

3.3. Chemopreventive Agents Targeting STAT3

Guggulsterone, isolated from Commiphora mukul, has also been reported to possess anticancer potential. It induces apoptosis and causes cell cycle arrest. The combination regimen increased the antineoplastic effects of erlotinib, cetuximab, and cisplatin in squamous cell carcinoma of the head and neck. Guggulsterone has been found to reduce the expression of STAT3 and induce apoptosis [75,76]. Another study reported that guggulsterone causes a decrease in the levels of phosphotyrosine STAT3 in multiple myeloma and squamous cell carcinoma. Furthermore, it also inhibits LPS-induced inflammatory cytokines in the NF-κB pathway [77]. A synthetic derivative of guggulsterone, GSD-1, has recently been reported to exert its strong inhibitory effects on NF-κB which helped reduce the metastatic potential of breast cancer cells [78].
Astaxanthin, a ketocarotenoid produced by certain algae, has been reported to block DMBA-induced hamster buccal pouch (HBP) carcinomas by downregulating JAK/STAT signaling. Astaxanthin has been found to reduce the expression of genes involved in the JAK/STAT3 pathway, such as cyclin D1, MMP-2, and VEGF. Thus, it reduces tumor cell proliferation, invasion, and angiogenesis [79].
Curcumin is a natural polyphenolic present in turmeric rhizome. It has been reported to suppress STAT3 and NF-κB signaling. A study demonstrated that a combination of epigallocatechin gallate and curcumin suppresses STAT3 phosphorylation in breast cancer-derived stem cells. It also reduces COX-2 activity and, therefore, can be considered a good agent for combing with those potentiating Nrf2 antioxidant pathways. It also increases cancer cell apoptosis mediated by Bcl-2. Proapoptotic effects are more pronounced when used with Wnt signaling inhibitors [80].
Silibinin is a natural flavonoid with chemopreventive potential. Silibinin has been reported to have anti-inflammatory and antineoplastic properties. In the current study, silibinin significantly inhibited the viability of intestinal tumor cells. The production of inflammatory cytokines and phosphorylation of STAT3 is inhibited in intestinal tumor cells. Silibinin (750 mg/kg) decreased the number and size of tumors induced by azoxymethane/DSS. Colitis and tumor scores decreased. The rate of proliferation also reduced with an increase in tumor cell apoptosis. Moreover, silibinin reduced the production of inflammatory cytokines and attenuated the impairment of the colonic mucosal barrier. Furthermore, an interaction of the probe with cellular molecules showed that silibinin suppressed the LPS-induced upregulation of STAT3 phosphorylation. It also reduced the expression of IL-6. The study concluded that silibinin had chemoprotective potential via the IL-6/STAT3 pathway, giving it dual beneficial activity in cancer and inflammation, both of which are present in colitis-associated cancer [81].
Acetoside is a naturally occurring glycoside found in plants. Its chemopreventive potential was assessed in a rat model of hepatocellular cancer chemically induced by diethyl nitrosamine (DEN). Acetoside was administered at 0.1% and 0.3% of their diet 2 weeks before chemical induction of hepatic cancer. Treatment was continued for 18 weeks. Histological studies have shown that acetoside reduces nodule size in hepatocellular cancer. The levels of biochemical markers of hepatocytic injury (ALT), inflammation (IL-6, IFN-γ, and TNF-α), and apoptosis (Caspase-3) improved after the administration of acetoside. It also ameliorated the DEN-induced DNA damage, cytotoxicity, and genotoxicity. It also considerably reduced oxidative damage. Furthermore, the reduced expression of NF-κB, Bcl2, and STAT3 showed anti-inflammatory, proapoptotic, and anticancer potential. Thus, acetoside induced STAT3-mediated antioxidant and anti-inflammatory effects along with its antiproliferative and proapoptotic properties, making it an agent with good chemopreventive potential [82].

3.4. Targeting STAT3 Improves Sensitivity of Other Anticancer Agents

Since STAT3 is the converging point of many upstream stimuli, receptors, and ligands, combining anti-STAT3 agents with other chemotherapy or immunosuppressants might offer promising ways to delay the progression of aggressive cancers. The STAT3 pathway is involved in the pathogenesis of EGFR-dependent squamous cell carcinoma (SCC). The invasive potential of EGFR-mediated SCC is greatly enhanced by the persistent activation of STAT3. A previous study suggested that combining anti-EGFR and anti-STAT3 agents would prove a practically effective mode of cancer suppression [83]. Since SCC and other solid tumors are resistant to anticancer agents, their suppression and elimination may warrant blocking via multiple pathways [84]. It has also been reported that SCC of the head and neck is more sensitive to cetuximab when combined with a short hairpin RNA knockdown approach for STAT3 inhibition. Combination therapy enhances DNA damage and apoptosis in cancer cells, as STAT3 activation is important for cell survival [83].
In pancreatic cancer, STAT3 activation has also been correlated with developing resistance to MEK inhibitors. Resistance is due to mutations in K-Ras and MEK inhibitors that target the Ras pathway and are less efficacious. The authors reported that the use of MEK inhibitors such as AZD6244 and trametinib caused profound activation of STAT3 in K-Ras mutant pancreatic cancer. We believe that STAT3 may be an important factor in developing resistance to MEK inhibitors in K-Ras-mutated pancreatic cancer. Therefore, a combination of LY5 (STAT3 inhibitor) and trametinib (MEK inhibitor) was administered to assess anticancer efficacy in resistant cancer. The results showed that trametinib displayed better tumor suppression in the presence of a STAT3 inhibitor. The authors concluded that the STAT3 regimen improves the efficacy of anti-MEK agents in resistant pancreatic cancers [85].
Curcumin has been reported to have a good chemopreventive potential. One known mechanism is the suppression of the JAK/STAT3 pathway. Epigallocatechin gallate is another promising candidate as a chemopreventive agent for cancer. This study assessed the effect of the two as monotherapy and combination therapies on angiogenesis in colorectal carcinoma cell lines (HCT116 and HT-29). Although both inhibited angiogenesis via inhibition of JAK/STAT3 signaling, their individual effects were minimal. However, the anti-angiogenic effects were potentiated when a combination of the two was used [86,87].
To devise new therapeutic strategies for cholangiocarcinoma, the combined effects of doxorubicin and β-caryophyllene were studied in Mz-ChA-1 and H69 cholangiocyte cell lines. In the carcinoma cell line Mz-ChA-1, β-caryophyllene synergized with the cytotoxic effect of doxorubicin at lower doses. However, it exerted cytoprotective effects on the H69 cell line (non-malignant cholangiocytes) after exposure for 24 h. Mechanistic insights revealed that the synergistic cytotoxicity of doxorubicin was due to cell cycle arrest in the G2/M phase by β-caryophyllene. It was also observed that the presence of β-caryophyllene improved the suppression of STAT3 signaling by doxorubicin. Hence, this study reports that the sensitivity of cholangiocarcinoma to doxorubicin is improved due to better suppression of STAT3 signaling by the concomitant use of β-caryophyllene, making it a good candidate as a chemosensitizer and chemopreventive agent [88].
Table 1. Various natural and synthetic STAT3 antiphosphorylating agents.
Table 1. Various natural and synthetic STAT3 antiphosphorylating agents.
Inhibitor of STAT3 PhosphorylationClass of CompoundMechanismReferences
1AlantolactoneSesquiterpene lactoneBinds and inhibits phosphorylation at Tyr705 in pancreatic cancer[89]
2S-3I 1757Synthetic; salicylic acid derivativeBinds pTyr 705 at SH2 domain; inhibits dimerization[90]
3B12Synthetic; sulfamoyl benzamide derivativeInhibits phosphorylation at Tyr 705; inhibits STAT3 phosphorylation induced by IL-6 [91]
4CinobufaginNatural; bufadienolideInhibits STAT3 phosphorylation; Inhibits EMT; Inhibits IL-6 mediated STAT3 translocation in colon cancer[92]
5ACT001Synthetic; parthenolide derivative (sesquiterpene lactone)Directly binds STAT3, inhibits phosphorylation; inhibits PD-L1 in glioblastoma[93]
6ResveratrolNatural stilbenoidInhibits IL-6 induced phosphorylation at Tyr 705; inhibits EMT in cervical cancer[94]
7Piperine & piperlongumineNatural alkaloidsCombination inhibits STAT3 phosphorylation; induces apoptosis selectively in breast cancer cells[95]
8Curcubitacin BNatural triterpeneInhibits STAT3 phosphorylation at Tyr 705; inhibits its nuclear translocation; induces apoptosis in gastric cancer [96]
9oleaceinNatural polyphenolic Reduces cell adhesion, migration, inhibits STAT3 phosphorylation; induces apoptosis in neuroblastoma [97]
10HJC0152Synthetic niclosamide derivativeInhibits STAT3 phosphorylation at Tyr 705; reduces glutamine and glutathione causing oxidative stress-mediated apoptosis in lung cancer.[98]
11CostunolideNatural sesquiterpeneInhibits STAT3 phosphorylation at Tyr 705; inhibits metastasis in osteosarcoma[99]
12Ginsenoside Rh1Natural triterpenoid saponinInhibits its phosphorylation, nuclear translocation, and accumulation; inhibits NF-κB in triple-negative breast cancer[100]
13ConvollatoxinNatural glycosideInhibits STAT3 phosphorylation at Tyr 705 & Ser 727; inhibits phosphorylation of JAK1, JAK2 & Src; promotes apoptosis in colon cancer[101]
14SS-4Synthetic Phenoxyacetamide derivativeInhibits STAT3 phosphorylation at Tyr 705; highly potent and STAT3 selective glioblastoma tumor growth inhibitor[102]
15WZ-2-033Synthetic acetamide derivativeInhibits STAT3 phosphorylation at Tyr 705; inhibits its dimerization & nuclear translocation; inhibits metastasis; induces apoptosis in triple-negative breast cancer and gastric cancer[70]

4. Src

Src was the first oncogene to be discovered [103]. It is a prototype of the Src family of kinases, which are non-receptor tyrosine kinases. These include Fyn, Yes, Blk, Yrk, Fgr, Hck, Lck, and Lyn. Src, Fyn, and Yes are ubiquitously expressed in many tissues. Higher concentrations have been observed in neurons, platelets, and osteoclasts [104]. Their basic function is to catalyze the transfer of phosphate from ATP to tyrosine residues at specific positions in proteins. This tyrosine phosphorylation causes activation and transduction of downstream molecules to transmit signals. The ultimate result of this signaling may be the activation of nuclear factors, resulting in gene expression or reorganization of the cell cytoskeleton [105]. Figure 5 shows the upstream stimuli of Src and its downstream pathway molecules, leading to cell survival, proliferation, and cancer progression.

4.1. Significance of Src in Cancer

Studies have shown that upon activation, Src induces cell growth and survival, leading to the promotion of tumor formation, promotion of reorganization of the cell actin cytoskeleton, and p120-mediated disruption of tight junctions, which subsequently facilitates the invasion and motility of cells (Figure 6) [106,107]. Src overactivation has been observed in multiple cancers, including melanoma, glioma, gastric, pancreatic, colorectal, prostate, breast, lung, head, and neck [108,109,110,111,112,113,114,115,116].
It has been shown that the migration and invasion of cancer cells underlie the ‘‘mesenchymal-type” mechanism where matrix-degrading proteases promote the locomotion of cancer cells through pericellular ECM breakdown [117]. Studies have shown that Src is involved in the increased expression of MMPs via diverse pathways, including the Src/Akt and NF-κB pathways [118], ERK and PI3K signaling [119], and PKC/MAPK/AP-1/LEF-1 and PI3K/AP-1/LEF-1 pathways [120]. Furthermore, Src has been reported to upregulate the expression of MMPs. A study reported that WNT5A upregulates Src, which induces MMP-14, all leading to invasion in osteosarcoma cells [121]. Hypoxia also enhances the invasive characteristics of malignant cells and angiogenesis via stimulation of the Src signaling pathway. Cannabidiol has been reported to inhibit Src, which causes a reduction in the hypoxia-inducible factor and consequent angiogenesis in breast cancer cells [122]. Additionally, Src has been shown to facilitate tumor cell extravasation by enhancing VEGF-induced vascular permeability [123].
Epithelial-mesenchymal transition (EMT) is a complicated process through which epithelial cells attain the characteristics of mesenchymal cells with increased migratory potential [124]. Loss of E-cadherin is a hallmark of EMT, in which cells detach from each other and start migrating to other parts of the body [105,125]. It has been shown that the activation of Src phosphorylates the E-cadherin–β-catenin complex, dissociating the latter, and subsequently induces EMT [126]. Upon dissociation from the complex, β-catenin translocates to the nucleus, where it transcriptionally activates various EMT-related targets such as Snail-family members, vimentin, Myc, matrix-degrading proteases, and cyclin D [127,128]. Src-induced EMT is related to and may promote the metastatic potential of cancer cells [129,130,131].

4.2. Chemoprevention via Src Inhibition

The inhibitory effect of apigenin on TGF-β-stimulated VEGF production in human prostate carcinoma cells was analyzed. The authors reported that apigenin blocked VEGF and TGF-β1-induced phosphorylation and was correlated with cancer progression, especially in Smad2/3 and Src/FAK/Akt signaling pathways [132]. It has been shown that activation of Src actively participates in early stage (ER-negative) breast cancer initiation, whereas downregulation of Src significantly inhibits cancer progression involving various underlying mechanisms such as suppression of Myc translation, reduction of GLUT1 transcription, and glucose uptake by tumor cells. These findings highlight the importance of Src inhibitors in the prevention and treatment of ER-negative breast cancer [133].
An investigation reported that shikonin blocked STAT3/FAK/Src signaling and stimulated the suppression of stem cell load in vitro, tumorigenicity, and metastasis in vivo. Compared to individual inhibitors, the combined blockade of STAT3 with Src or FAK decreased cell migration, invasion, and mammosphere formation more significantly [134]. S100 calcium binding proteinA7 (S100A7) is upregulated in many cancers and is associated with the facilitation of metastasis. A study reported that metastasis induced by S100A7 is reduced by flavonoids, such as luteolin and quercetin, via inhibition of Src/STAT3 signaling in A431-III cells. Flavonoids reduce the levels of S100A7, phosphorylated Src, and phosphorylated STAT3. The effect of flavonoids on EMT markers, such as E-cadherin, was also observed. Flavonoids increased the levels of E-cadherin and were found to resist metastasis of cancer cells in zebrafish larvae [135]. Another study reported that the extract of Morus alba could suppress the metastatic potential of non-small cell lung cancer cells. The plant extract also caused the downregulation of EMT markers such as Slug and vimentin. It also upregulates the expression of occludin, a tight junction protein. Insights into the anti-invasive and anti-metastatic mechanisms revealed that the plant extract decreased the activation of STAT3 and Src by inhibiting their phosphorylation [136].

5. Conclusions

Recently, chemoprevention studies have gained popularity because of the basic concept of delaying the production (if absent) or progression (if present) of cancer. Therefore, investigations leading to the discovery of molecular pathways and target cellular molecules are necessary for a complete understanding of chemopreventive mechanisms. The molecular targets for chemopreventive agents described in this review (Nrf2, STAT3, and Src) have shown to be promising candidates with chemopreventive potential in various cancer cell lines or carcinogen-induced tumorigenesis models in animals. Structural modifications of chemoprevention agents into safer and more effective chemoprevention agents are needed. Recently, researchers have synthesized such derivatives that include dimethylaminomethyl curcumin, diarylyheptanoid curcumin [137], and GSD-1. Moreover, effective delivery of therapeutic doses of medicaments requires careful design of drug delivery systems such as lipid-based nanoparticles. These modifications may enable the agent to effectively concentrate at its target sites and regulate the intracellular molecular targets (Nrf2, STAT3, etc.) to counteract cancer. Emphasis must be given to changing the chemical structures such that their aqueous and lipid solubility becomes optimal for optimal drug delivery and bioavailability. Additions of groups capable of binding specific proteins present in the tumor microenvironment may increase their selectivity toward tumor cells. The use of agents acting on Nrf2, STAT3, Src, and NF-κB, etc., can be looked into in various cancers aggravated by oxidative stress, such as estrogen-dependent breast cancer and steatohepatitis. Furthermore, the discovery and synthesis of agents which can act on Nrf2, STAT3, and NF-κB (like physodic and salazinic acids [138]) is yet another area to be explored for effective and timely chemoprevention.

Author Contributions

Y.S.L.; Conceptualization, supervision; H.A. and S.U.I.; Writing—Original Draft; M.B.A.; Designed all the figures. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Conflicts of Interest

The authors declare no conflict of interest, financial or otherwise.

Abbreviations

Bcl-2B-cell lymphoma 2
Bcl-xLB-cell lymphoma-extra-large
bFGFBasic fibroblast growth factor
c-MycCellular myelocytomatosis
CdcCell division cycle
COX-2Cyclooxygenase-2
DSSDextran sodium sulfate
FAKFocal adhesion kinase
GCSGlutamylcysteine synthetase
HIFHypoxia-inducible factor
HO-1Heme oxygenase 1
IBDInflammatory bowel disease
ICAM-1Intercellular adhesion molecule-1
ILInter leukin
iNOSInducible nitric oxide synthase
IκBInhibitor of Nuclear factor-κB
JNKC-Jun N-terminal kinase
LPSLipo polysaccharide
M-CSFMacrophage colony-stimulating factor
MafMusculoaponeurotic fibrosarcoma
MAPKMitogen-activated protein kinase
MMPMatrix metalloproteinase
MUC1Mucin 1
NF-κBNuclear factor κB
NONitric oxide
NQO1NADPH quinone oxidoreductase 1
Nrf2Nuclear factor Erythroid factor 2-related factor 2
PGE2Prostaglandin E2
PI3Kphosphoinositide 3-kinase
ROSReactive oxygen species
RTKreceptor tyrosine kinase
SCCSquamous cell carcinoma
SFKSrc family kinase
SODSuperoxide dismutase
STAT3Signal Transducers and Activators of Transcription-3
TNFTumor necrosis factor
VEGFVascular endothelial growth factor

References

  1. Landis-Piwowar, K.R.; Iyer, N.R. Cancer chemoprevention: Current state of the art. Cancer Growth Metastasis 2014, 7, CGM.S11288. [Google Scholar] [CrossRef] [PubMed]
  2. Sporn, M.B. Approaches to prevention of epithelial cancer during the preneoplastic period. Cancer Res. 1976, 36, 2699–2702. [Google Scholar] [PubMed]
  3. Steward, W.; Brown, K. Cancer chemoprevention: A rapidly evolving field. Br. J. Cancer 2013, 109, 1–7. [Google Scholar] [CrossRef] [PubMed]
  4. Shu, L.; Cheung, K.-L.; Khor, T.O.; Chen, C.; Kong, A.-N. Phytochemicals: Cancer chemoprevention and suppression of tumor onset and metastasis. Cancer Metastasis Rev. 2010, 29, 483–502. [Google Scholar] [CrossRef] [PubMed]
  5. Yang, H.; Landis-Piwowar, K.; Chan, T.H.; Dou, Q.P. Green tea polyphenols as proteasome inhibitors: Implication in chemoprevention. Curr. Cancer Drug Targets 2011, 11, 296–306. [Google Scholar] [CrossRef] [PubMed]
  6. Landis-Piwowar, K.; Chen, D.; Chan, T.H.; Dou, Q.P. Inhibition of catechol-O-methyltransferase activity in human breast cancer cells enhances the biological effect of the green tea polyphenol (-)-EGCG. Oncol. Rep. 2010, 24, 563–569. [Google Scholar] [PubMed]
  7. Gan, F.-F.; Ling, H.; Ang, X.; Reddy, S.A.; Lee, S.S.; Yang, H.; Tan, S.-H.; Hayes, J.D.; Chui, W.-K.; Chew, E.-H. A novel shogaol analog suppresses cancer cell invasion and inflammation, and displays cytoprotective effects through modulation of NF-κB and Nrf2-Keap1 signaling pathways. Toxicol. Appl. Pharmacol. 2013, 272, 852–862. [Google Scholar] [CrossRef] [PubMed]
  8. Din, F.V.; Valanciute, A.; Houde, V.P.; Zibrova, D.; Green, K.A.; Sakamoto, K.; Alessi, D.R.; Dunlop, M.G. Aspirin inhibits mTOR signaling, activates AMP-activated protein kinase, and induces autophagy in colorectal cancer cells. Gastroenterology 2012, 142, e1503. [Google Scholar] [CrossRef]
  9. Chun, K.-S.; Raut, P.K.; Kim, D.-H.; Surh, Y.-J. Role of chemopreventive phytochemicals in NRF2-mediated redox homeostasis in humans. Free Radic. Biol. Med. 2021, 172, 699–715. [Google Scholar] [CrossRef]
  10. Holland, R.; Fishbein, J.C. Chemistry of the cysteine sensors in Kelch-like ECH-associated protein 1. Antioxid. Redox Signal. 2010, 13, 1749–1761. [Google Scholar] [CrossRef]
  11. Taguchi, K.; Yamamoto, M. The KEAP1–NRF2 system as a molecular target of cancer treatment. Cancers 2020, 13, 46. [Google Scholar] [CrossRef] [PubMed]
  12. Wagner, A.E.; Sturm, C.; Piegholdt, S.; Wolf, I.M.; Esatbeyoglu, T.; De Nicola, G.R.; Iori, R.; Rimbach, G. Myrosinase-treated glucoerucin is a potent inducer of the Nrf2 target gene heme oxygenase 1—Studies in cultured HT-29 cells and mice. J. Nutr. Biochem. 2015, 26, 661–666. [Google Scholar] [CrossRef] [PubMed]
  13. Consoli, V.; Sorrenti, V.; Grosso, S.; Vanella, L. Heme oxygenase-1 signaling and redox homeostasis in physiopathological conditions. Biomolecules 2021, 11, 589. [Google Scholar] [CrossRef] [PubMed]
  14. Zhao, R.; Yang, B.; Wang, L.; Xue, P.; Deng, B.; Zhang, G.; Jiang, S.; Zhang, M.; Liu, M.; Pi, J. Curcumin protects human keratinocytes against inorganic arsenite-induced acute cytotoxicity through an NRF2-dependent mechanism. Oxidative Med. Cell. Longev. 2013, 2013, 412576. [Google Scholar] [CrossRef] [PubMed]
  15. Zhou, R.; Lin, J.; Wu, D. Sulforaphane induces Nrf2 and protects against CYP2E1-dependent binge alcohol-induced liver steatosis. Biochim. Et Biophys. Acta BBA Gen. Subj. 2014, 1840, 209–218. [Google Scholar] [CrossRef]
  16. Duan, X.; Liu, D.; Xing, X.; Li, J.; Zhao, S.; Nie, H.; Zhang, Y.; Sun, G.; Li, B. Tert-butylhydroquinone as a phenolic activator of Nrf2 antagonizes arsenic-induced oxidative cytotoxicity but promotes arsenic methylation and detoxication in human hepatocyte cell line. Biol. Trace Elem. Res. 2014, 160, 294–302. [Google Scholar] [CrossRef]
  17. Duan, X.; Li, J.; Li, W.; Xing, X.; Zhang, Y.; Zhao, L.; Sun, G.; Gao, X.-H.; Li, B. Antioxidant tert-butylhydroquinone ameliorates arsenic-induced intracellular damages and apoptosis through induction of Nrf2-dependent antioxidant responses as well as stabilization of anti-apoptotic factor Bcl-2 in human keratinocytes. Free Radic. Biol. Med. 2016, 94, 74–87. [Google Scholar] [CrossRef]
  18. Iida, K.; Itoh, K.; Maher, J.M.; Kumagai, Y.; Oyasu, R.; Mori, Y.; Shimazui, T.; Akaza, H.; Yamamoto, M. Nrf2 and p53 cooperatively protect against BBN-induced urinary bladder carcinogenesis. Carcinogenesis 2007, 28, 2398–2403. [Google Scholar] [CrossRef]
  19. Iida, K.; Itoh, K.; Kumagai, Y.; Oyasu, R.; Hattori, K.; Kawai, K.; Shimazui, T.; Akaza, H.; Yamamoto, M. Nrf2 is essential for the chemopreventive efficacy of oltipraz against urinary bladder carcinogenesis. Cancer Res. 2004, 64, 6424–6431. [Google Scholar] [CrossRef]
  20. Bi, W.; He, C.-N.; Li, X.-X.; Zhou, L.-Y.; Liu, R.-J.; Zhang, S.; Li, G.-Q.; Chen, Z.-C.; Zhang, P.-F. Ginnalin A from Kujin tea (Acer tataricum subsp. ginnala) exhibits a colorectal cancer chemoprevention effect via activation of the Nrf2/HO-1 signaling pathway. Food Funct. 2018, 9, 2809–2819. [Google Scholar] [CrossRef]
  21. Wang, S.; Wu, Y.-Y.; Wang, X.; Shen, P.; Jia, Q.; Yu, S.; Wang, Y.; Li, X.; Chen, W.; Wang, A. Lycopene prevents carcinogen-induced cutaneous tumor by enhancing activation of the Nrf2 pathway through p62-triggered autophagic Keap1 degradation. Aging 2020, 12, 8167. [Google Scholar] [CrossRef] [PubMed]
  22. Tsai, T.-F.; Chen, P.-C.; Lin, Y.-C.; Chou, K.-Y.; Chen, H.-E.; Ho, C.-Y.; Lin, J.-F.; Hwang, T.I.-S. Miconazole contributes to NRF2 activation by noncanonical P62-KEAP1 pathway in bladder cancer cells. Drug Des. Dev. Ther. 2020, 14, 1209. [Google Scholar] [CrossRef] [PubMed]
  23. Zinatizadeh, M.R.; Schock, B.; Chalbatani, G.M.; Zarandi, P.K.; Jalali, S.A.; Miri, S.R. The Nuclear Factor Kappa B (NF-kB) signaling in cancer development and immune diseases. Genes Dis. 2021, 8, 287–297. [Google Scholar] [CrossRef] [PubMed]
  24. Setia, S.; Nehru, B.; Sanyal, S.N. Activation of NF-κB: Bridging the gap between inflammation and cancer in colitis-mediated colon carcinogenesis. Biomed. Pharmacother. 2014, 68, 119–128. [Google Scholar] [CrossRef]
  25. Soleimani, A.; Rahmani, F.; Ferns, G.A.; Ryzhikov, M.; Avan, A.; Hassanian, S.M. Role of the NF-κB signaling pathway in the pathogenesis of colorectal cancer. Gene 2020, 726, 144132. [Google Scholar] [CrossRef]
  26. Sturm, C.; Wagner, A.E. Brassica-derived plant bioactives as modulators of chemopreventive and inflammatory signaling pathways. Int. J. Mol. Sci. 2017, 18, 1890. [Google Scholar] [CrossRef]
  27. Ghasemi, F.; Shafiee, M.; Banikazemi, Z.; Pourhanifeh, M.H.; Khanbabaei, H.; Shamshirian, A.; Moghadam, S.A.; ArefNezhad, R.; Sahebkar, A.; Avan, A. Curcumin inhibits NF-kB and Wnt/β-catenin pathways in cervical cancer cells. Pathol. Res. Pract. 2019, 215, 152556. [Google Scholar] [CrossRef]
  28. Khongthong, P.; Roseweir, A.K.; Edwards, J. The NF-KB pathway and endocrine therapy resistance in breast cancer. Endocr. -Relat. Cancer 2019, 26, R369–R380. [Google Scholar] [CrossRef]
  29. Diaz, G.M.; Hupperts, R.; Fraussen, J.; Somers, V. Dimethyl fumarate treatment in multiple sclerosis: Recent advances in clinical and immunological studies. Autoimmun. Rev. 2018, 17, 1240–1250. [Google Scholar] [CrossRef]
  30. Lee, S.; Choi, S.-Y.; Choo, Y.-Y.; Kim, O.; Tran, P.T.; Dao, C.T.; Min, B.-S.; Lee, J.-H. Sappanone A exhibits anti-inflammatory effects via modulation of Nrf2 and NF-κB. Int. Immunopharmacol. 2015, 28, 328–336. [Google Scholar] [CrossRef]
  31. Rogati, F.; Millán, E.; Appendino, G.; Correa, A.; Caprioglio, D.; Minassi, A.; Muñoz, E. Identification of a strigoterpenoid with dual Nrf2 and Nf-κB modulatory activity. ACS Med. Chem. Lett. 2019, 10, 606–610. [Google Scholar] [CrossRef] [PubMed]
  32. Krajka-Kuźniak, V.; Cykowiak, M.; Szaefer, H.; Kleszcz, R.; Baer-Dubowska, W. Combination of xanthohumol and phenethyl isothiocyanate inhibits NF-κB and activates Nrf2 in pancreatic cancer cells. Toxicol. In Vitr. 2020, 65, 104799. [Google Scholar] [CrossRef] [PubMed]
  33. Sailaja, B.S.; Aita, R.; Maledatu, S.; Ribnicky, D.; Verzi, M.P.; Raskin, I. Moringa isothiocyanate-1 regulates Nrf2 and NF-κB pathway in response to LPS-driven sepsis and inflammation. PLoS ONE 2021, 16, e0248691. [Google Scholar]
  34. Sivandzade, F.; Prasad, S.; Bhalerao, A.; Cucullo, L. NRF2 and NF-κB interplay in cerebrovascular and neurodegenerative disorders: Molecular mechanisms and possible therapeutic approaches. Redox Biol. 2019, 21, 101059. [Google Scholar] [CrossRef] [PubMed]
  35. Yerra, V.G.; Negi, G.; Sharma, S.S.; Kumar, A. Potential therapeutic effects of the simultaneous targeting of the Nrf2 and NF-κB pathways in diabetic neuropathy. Redox Biol. 2013, 1, 394–397. [Google Scholar] [CrossRef]
  36. Fakih, M.G. Metastatic colorectal cancer: Current state and future directions. J. Clin. Oncol. 2015, 33, 1809–1824. [Google Scholar] [CrossRef]
  37. Levin, T.R.; Corley, D.A.; Jensen, C.D.; Schottinger, J.E.; Quinn, V.P.; Zauber, A.G.; Lee, J.K.; Zhao, W.K.; Udaltsova, N.; Ghai, N.R. Effects of organized colorectal cancer screening on cancer incidence and mortality in a large community-based population. Gastroenterology 2018, 155, e1385. [Google Scholar] [CrossRef]
  38. Schmitt, M.; Greten, F.R. The inflammatory pathogenesis of colorectal cancer. Nat. Rev. Immunol. 2021, 21, 653–667. [Google Scholar] [CrossRef]
  39. Sun, M.; He, C.; Cong, Y.; Liu, Z. Regulatory immune cells in regulation of intestinal inflammatory response to microbiota. Mucosal Immunol. 2015, 8, 969–978. [Google Scholar] [CrossRef]
  40. Ullman, T.A.; Itzkowitz, S.H. Intestinal inflammation and cancer. Gastroenterology 2011, 140, 1807–1816.e1801. [Google Scholar] [CrossRef]
  41. Lucafò, M.; Curci, D.; Franzin, M.; Decorti, G.; Stocco, G. Inflammatory Bowel Disease and Risk of Colorectal Cancer: An Overview From Pathophysiology to Pharmacological Prevention. Front. Pharmacol. 2021, 12, 2916. [Google Scholar] [CrossRef] [PubMed]
  42. Neurath, M.F. IL-23 in inflammatory bowel diseases and colon cancer. Cytokine Growth Factor Rev. 2019, 45, 1–8. [Google Scholar] [CrossRef] [PubMed]
  43. Lee, H.H.; Yang, S.S.; Vo, M.-T.; Cho, W.J.; Lee, B.J.; Leem, S.-H.; Lee, S.-H.; Cha, H.J.; Park, J.W. Tristetraprolin down-regulates IL-23 expression in colon cancer cells. Mol. Cells 2013, 36, 571–576. [Google Scholar] [CrossRef]
  44. Ibrahim, S.; Girault, A.; Ohresser, M.; Lereclus, E.; Paintaud, G.; Lecomte, T.; Raoul, W. Monoclonal antibodies targeting the IL-17/IL-17RA axis: An opportunity to improve the efficiency of anti-VEGF therapy in fighting metastatic colorectal cancer? Clin. Colorectal Cancer 2018, 17, e109–e113. [Google Scholar] [CrossRef]
  45. Greten, F.R.; Eckmann, L.; Greten, T.F.; Park, J.M.; Li, Z.-W.; Egan, L.J.; Kagnoff, M.F.; Karin, M. IKKβ links inflammation and tumorigenesis in a mouse model of colitis-associated cancer. Cell 2004, 118, 285–296. [Google Scholar] [CrossRef] [PubMed]
  46. Park, E.J.; Lee, J.H.; Yu, G.-Y.; He, G.; Ali, S.R.; Holzer, R.G.; Österreicher, C.H.; Takahashi, H.; Karin, M. Dietary and genetic obesity promote liver inflammation and tumorigenesis by enhancing IL-6 and TNF expression. Cell 2010, 140, 197–208. [Google Scholar] [CrossRef] [PubMed]
  47. Perše, M.; Cerar, A. Dextran sodium sulphate colitis mouse model: Traps and tricks. J. Biomed. Biotechnol. 2012, 2012, 718617. [Google Scholar] [CrossRef]
  48. Wirtz, S.; Neufert, C.; Weigmann, B.; Neurath, M.F. Chemically induced mouse models of intestinal inflammation. Nat. Protoc. 2007, 2, 541–546. [Google Scholar] [CrossRef]
  49. Lee, I.-A.; Bae, E.-A.; Hyun, Y.-J.; Kim, D.-H. Dextran sulfate sodium and 2, 4, 6-trinitrobenzene sulfonic acid induce lipid peroxidation by the proliferation of intestinal gram-negative bacteria in mice. J. Inflamm. 2010, 7, 1–9. [Google Scholar] [CrossRef]
  50. Yum, H.-W.; Zhong, X.; Park, J.; Na, H.-K.; Kim, N.; Lee, H.S.; Surh, Y.-J. Oligonol inhibits dextran sulfate sodium-induced colitis and colonic adenoma formation in mice. Antioxid. Redox Signal. 2013, 19, 102–114. [Google Scholar] [CrossRef]
  51. Zhu, H.; Li, Y.R. Oxidative stress and redox signaling mechanisms of inflammatory bowel disease: Updated experimental and clinical evidence. Exp. Biol. Med. 2012, 237, 474–480. [Google Scholar] [CrossRef] [PubMed]
  52. Lu, M.-C.; Ji, J.-A.; Jiang, Y.-L.; Chen, Z.-Y.; Yuan, Z.-W.; You, Q.-D.; Jiang, Z.-Y. An inhibitor of the Keap1-Nrf2 protein-protein interaction protects NCM460 colonic cells and alleviates experimental colitis. Sci. Rep. 2016, 6, 1–13. [Google Scholar] [CrossRef] [PubMed]
  53. Khor, T.O.; Huang, M.-T.; Kwon, K.H.; Chan, J.Y.; Reddy, B.S.; Kong, A.-N. Nrf2-deficient mice have an increased susceptibility to dextran sulfate sodium–induced colitis. Cancer Res. 2006, 66, 11580–11584. [Google Scholar] [CrossRef] [PubMed]
  54. Arisawa, T.; Tahara, T.; Shibata, T.; Nagasaka, M.; Nakamura, M.; Kamiya, Y.; Fujita, H.; Yoshioka, D.; Okubo, M.; Sakata, M. Nrf2 gene promoter polymorphism is associated with ulcerative colitis in a Japanese population. Hepato Gastroenterol. 2008, 55, 394–397. [Google Scholar]
  55. Khor, T.O.; Huang, M.-T.; Prawan, A.; Liu, Y.; Hao, X.; Yu, S.; Cheung, W.K.L.; Chan, J.Y.; Reddy, B.S.; Yang, C.S. Increased Susceptibility of Nrf2 Knockout Mice to Colitis-Associated Colorectal CancerColorectal Cancer Susceptibility of Nrf2 Knockout Mice. Cancer Prev. Res. 2008, 1, 187–191. [Google Scholar] [CrossRef]
  56. Hammad, A.; Zheng, Z.-H.; Gao, Y.; Namani, A.; Shi, H.-F.; Tang, X. Identification of novel Nrf2 target genes as prognostic biomarkers in colitis-associated colorectal cancer in Nrf2-deficient mice. Life Sci. 2019, 238, 116968. [Google Scholar] [CrossRef]
  57. Wagner, A.E.; Will, O.; Sturm, C.; Lipinski, S.; Rosenstiel, P.; Rimbach, G. DSS-induced acute colitis in C57BL/6 mice is mitigated by sulforaphane pre-treatment. J. Nutr. Biochem. 2013, 24, 2085–2091. [Google Scholar] [CrossRef]
  58. Khodir, A.E.; Atef, H.; Said, E.; ElKashef, H.A.; Salem, H.A. Implication of Nrf2/HO-1 pathway in the coloprotective effect of coenzyme Q10 against experimentally induced ulcerative colitis. Inflammopharmacology 2017, 25, 119–135. [Google Scholar] [CrossRef]
  59. Ma, J.-H.; Qin, L.; Li, X. Role of STAT3 signaling pathway in breast cancer. Cell Commun. Signal. 2020, 18, 1–13. [Google Scholar] [CrossRef]
  60. Qin, J.-J.; Yan, L.; Zhang, J.; Zhang, W.-D. STAT3 as a potential therapeutic target in triple negative breast cancer: A systematic review. J. Exp. Clin. Cancer Res. 2019, 38, 195. [Google Scholar] [CrossRef]
  61. You, W.; Tang, Q.; Zhang, C.; Wu, J.; Gu, C.; Wu, Z.; Li, X. IL-26 promotes the proliferation and survival of human gastric cancer cells by regulating the balance of STAT1 and STAT3 activation. PLoS ONE 2013, 8, e63588. [Google Scholar] [CrossRef] [PubMed]
  62. Liu, Y.; Lv, L.; Xiao, W.; Gong, C.; Yin, J.; Wang, D.; Sheng, H. Leptin activates STAT3 and ERK1/2 pathways and induces endometrial cancer cell proliferation. J. Huazhong Univ. Sci. Technol. Med. Sci. 2011, 31, 365–370. [Google Scholar] [CrossRef] [PubMed]
  63. Benerini Gatta, L.; Melocchi, L.; Bugatti, M.; Missale, F.; Lonardi, S.; Zanetti, B.; Cristinelli, L.; Belotti, S.; Simeone, C.; Ronca, R. Hyper-activation of STAT3 sustains progression of non-papillary basal-type bladder cancer via FOSL1 regulome. Cancers 2019, 11, 1219. [Google Scholar] [CrossRef] [PubMed]
  64. Zou, S.; Tong, Q.; Liu, B.; Huang, W.; Tian, Y.; Fu, X. Targeting STAT3 in cancer immunotherapy. Mol. Cancer 2020, 19, 1–19. [Google Scholar] [CrossRef]
  65. Bi, Y.-H.; Han, W.-Q.; Li, R.-F.; Wang, Y.-J.; Du, Z.-S.; Wang, X.-J.; Jiang, Y. Signal transducer and activator of transcription 3 promotes the Warburg effect possibly by inducing pyruvate kinase M2 phosphorylation in liver precancerous lesions. World J. Gastroenterol. 2019, 25, 1936. [Google Scholar] [CrossRef]
  66. Niu, G.; Wright, K.L.; Huang, M.; Song, L.; Haura, E.; Turkson, J.; Zhang, S.; Wang, T.; Sinibaldi, D.; Coppola, D. Constitutive Stat3 activity up-regulates VEGF expression and tumor angiogenesis. Oncogene 2002, 21, 2000–2008. [Google Scholar] [CrossRef]
  67. Kujawski, M.; Kortylewski, M.; Lee, H.; Herrmann, A.; Kay, H.; Yu, H. Stat3 mediates myeloid cell–dependent tumor angiogenesis in mice. J. Clin. Investig. 2008, 118, 3367–3377. [Google Scholar] [CrossRef]
  68. Mankan, A.K.; Greten, F.R. Inhibiting signal transducer and activator of transcription 3: Rationality and rationale design of inhibitors. Expert Opin. Investig. Drugs 2011, 20, 1263–1275. [Google Scholar] [CrossRef]
  69. Lin, W.-H.; Chang, Y.-W.; Hong, M.-X.; Hsu, T.-C.; Lee, K.-C.; Lin, C.; Lee, J.-L. STAT3 phosphorylation at Ser727 and Tyr705 differentially regulates the EMT–MET switch and cancer metastasis. Oncogene 2021, 40, 791–805. [Google Scholar] [CrossRef]
  70. Zhong, Y.; Deng, L.; Shi, S.; Huang, Q.-Y.; Ou-Yang, S.-M.; Mo, J.-S.; Zhu, K.; Qu, X.-M.; Liu, P.-Q.; Wang, Y.-X. The novel STAT3 inhibitor WZ-2-033 causes regression of human triple-negative breast cancer and gastric cancer xenografts. Acta Pharmacol. Sin. 2022, 43, 1013–1023. [Google Scholar] [CrossRef]
  71. Hedvat, M.; Huszar, D.; Herrmann, A.; Gozgit, J.M.; Schroeder, A.; Sheehy, A.; Buettner, R.; Proia, D.; Kowolik, C.M.; Xin, H. The JAK2 inhibitor AZD1480 potently blocks Stat3 signaling and oncogenesis in solid tumors. Cancer Cell 2009, 16, 487–497. [Google Scholar] [CrossRef] [PubMed]
  72. Klein, J.D.; Sano, D.; Sen, M.; Myers, J.N.; Grandis, J.R.; Kim, S. STAT3 oligonucleotide inhibits tumor angiogenesis in preclinical models of squamous cell carcinoma. PLoS ONE 2014, 9, e81819. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  73. Sethi, G.; Chatterjee, S.; Rajendran, P.; Li, F.; Shanmugam, M.K.; Wong, K.F.; Kumar, A.P.; Senapati, P.; Behera, A.K.; Hui, K.M. Inhibition of STAT3 dimerization and acetylation by garcinol suppresses the growth of human hepatocellular carcinoma in vitro and in vivo. Mol. Cancer 2014, 13, 1–14. [Google Scholar] [CrossRef] [PubMed]
  74. Kim, B.-H.; Lee, H.; Song, Y.; Park, J.-S.; Gadhe, C.G.; Choi, J.; Lee, C.-G.; Pae, A.N.; Kim, S.; Ye, S.-K. Development of oxadiazole-based ODZ10117 as a small-molecule inhibitor of STAT3 for targeted cancer therapy. J. Clin. Med. 2019, 8, 1847. [Google Scholar] [CrossRef] [PubMed]
  75. Ahn, K.S.; Sethi, G.; Sung, B.; Goel, A.; Ralhan, R.; Aggarwal, B.B. Guggulsterone, a farnesoid X receptor antagonist, inhibits constitutive and inducible STAT3 activation through induction of a protein tyrosine phosphatase SHP-1. Cancer Res. 2008, 68, 4406–4415. [Google Scholar] [CrossRef] [PubMed]
  76. Leeman-Neill, R.J.; Wheeler, S.E.; Singh, S.V.; Thomas, S.M.; Seethala, R.R.; Neill, D.B.; Panahandeh, M.C.; Hahm, E.-R.; Joyce, S.C.; Sen, M. Guggulsterone enhances head and neck cancer therapies via inhibition of signal transducer and activator of transcription-3. Carcinogenesis 2009, 30, 1848–1856. [Google Scholar] [CrossRef]
  77. Zhang, J.-H.; Shangguan, Z.-S.; Chen, C.; Zhang, H.-J.; Lin, Y. Anti-inflammatory effects of guggulsterone on murine macrophage by inhibiting LPS-induced inflammatory cytokines in NF-κB signaling pathway. Drug Des. Dev. Ther. 2016, 10, 1829. [Google Scholar] [CrossRef] [PubMed]
  78. Abdellatef, A.A.; Zhou, Y.; Yamada, A.; Elmekkawy, S.A.; Kohyama, A.; Yokoyama, S.; Meselhy, M.R.; Matsuya, Y.; Sakurai, H.; Hayakawa, Y. Synthetic E-guggulsterone derivative GSD-1 inhibits NF-κB signaling and suppresses the metastatic potential of breast cancer cells. Biomed. Pharmacother. 2021, 140, 111737. [Google Scholar] [CrossRef]
  79. Kowshik, J.; Baba, A.B.; Giri, H.; Deepak Reddy, G.; Dixit, M.; Nagini, S. Astaxanthin inhibits JAK/STAT-3 signaling to abrogate cell proliferation, invasion and angiogenesis in a hamster model of oral cancer. PLoS ONE 2014, 9, e109114. [Google Scholar] [CrossRef]
  80. Prajapati, K.S.; Gupta, S.; Kumar, S. Targeting Breast Cancer-Derived Stem Cells by Dietary Phytochemicals: A Strategy for Cancer Prevention and Treatment. Cancers 2022, 14, 2864. [Google Scholar] [CrossRef]
  81. Zheng, R.; Ma, J.; Wang, D.; Dong, W.; Wang, S.; Liu, T.; Xie, R.; Liu, L.; Wang, B.; Cao, H. Chemopreventive effects of silibinin on colitis-associated tumorigenesis by inhibiting IL-6/STAT3 signaling pathway. Mediat. Inflamm. 2018, 2018, 1562010. [Google Scholar] [CrossRef] [PubMed]
  82. Peerzada, K.J.; Faridi, A.H.; Sharma, L.; Bhardwaj, S.C.; Satti, N.K.; Shashi, B.; Tasduq, S.A. Acteoside-mediates chemoprevention of experimental liver carcinogenesis through STAT-3 regulated oxidative stress and apoptosis. Environ. Toxicol. 2016, 31, 782–798. [Google Scholar] [CrossRef] [PubMed]
  83. Bonner, J.A.; Yang, E.S.; Trummell, H.Q.; Nowsheen, S.; Willey, C.D.; Raisch, K.P. Inhibition of STAT-3 results in greater cetuximab sensitivity in head and neck squamous cell carcinoma. Radiother. Oncol. 2011, 99, 339–343. [Google Scholar] [CrossRef] [PubMed]
  84. Matar, P.; Rojo, F.; Cassia, R.; Moreno-Bueno, G.; Di Cosimo, S.; Tabernero, J.; Guzmán, M.; Rodriguez, S.; Arribas, J.; Palacios, J. Combined epidermal growth factor receptor targeting with the tyrosine kinase inhibitor gefitinib (ZD1839) and the monoclonal antibody cetuximab (IMC-C225): Superiority over single-agent receptor targeting. Clin. Cancer Res. 2004, 10, 6487–6501. [Google Scholar] [CrossRef]
  85. 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. [Google Scholar] [CrossRef]
  86. Jin, G.; Yang, Y.; Liu, K.; Zhao, J.; Chen, X.; Liu, H.; Bai, R.; Li, X.; Jiang, Y.; Zhang, X. Combination curcumin and (−)-epigallocatechin-3-gallate inhibits colorectal carcinoma microenvironment-induced angiogenesis by JAK/STAT3/IL-8 pathway. Oncogenesis 2017, 6, e384. [Google Scholar] [CrossRef]
  87. Alexandrow, M.G.; Song, L.J.; Altiok, S.; Gray, J.; Haura, E.B.; Kumar, N.B. Curcumin: A novel stat 3 pathway inhibitor for chemoprevention of lung cancer. Eur. J. Cancer Prev. 2012, 21, 407. [Google Scholar] [CrossRef]
  88. Di Sotto, A.; Di Giacomo, S.; Rubini, E.; Macone, A.; Gulli, M.; Mammola, C.L.; Eufemi, M.; Mancinelli, R.; Mazzanti, G. Modulation of STAT3 Signaling, cell redox defenses and cell cycle checkpoints by β-caryophyllene in cholangiocarcinoma cells: Possible mechanisms accounting for doxorubicin chemosensitization and chemoprevention. Cells 2020, 9, 858. [Google Scholar] [CrossRef]
  89. Zheng, H.; Yang, L.; Kang, Y.; Chen, M.; Lin, S.; Xiang, Y.; Li, C.; Dai, X.; Huang, X.; Liang, G. Alantolactone sensitizes human pancreatic cancer cells to EGFR inhibitors through the inhibition of STAT3 signaling. Mol. Carcinog. 2019, 58, 565–576. [Google Scholar] [CrossRef]
  90. 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 TransformationSTAT3 Inhibitor Suppresses Malignant Transformation. Cancer Res. 2013, 73, 1922–1933. [Google Scholar] [CrossRef]
  91. Wang, X.; Wu, K.; Fang, L.; Yang, X.; Zheng, N.; Du, Z.; Lu, Y.; Xie, Z.; Liu, Z.; Zuo, Z. Discovery of N-substituted sulfamoylbenzamide derivatives as novel inhibitors of STAT3 signaling pathway based on Niclosamide. Eur. J. Med. Chem. 2021, 218, 113362. [Google Scholar] [CrossRef] [PubMed]
  92. Bai, Y.; Wang, X.; Cai, M.; Ma, C.; Xiang, Y.; Hu, W.; Zhou, B.; Zhao, C.; Dai, X.; Li, X. Cinobufagin suppresses colorectal cancer growth via STAT3 pathway inhibition. Am. J. Cancer Res. 2021, 11, 200. [Google Scholar] [PubMed]
  93. Tong, L.; Li, J.; Li, Q.; Wang, X.; Medikonda, R.; Zhao, T.; Li, T.; Ma, H.; Yi, L.; Liu, P. ACT001 reduces the expression of PD-L1 by inhibiting the phosphorylation of STAT3 in glioblastoma. Theranostics 2020, 10, 5943. [Google Scholar] [CrossRef]
  94. Sun, X.; Xu, Q.; Zeng, L.; Xie, L.; Zhao, Q.; Xu, H.; Wang, X.; Jiang, N.; Fu, P.; Sang, M. Resveratrol suppresses the growth and metastatic potential of cervical cancer by inhibiting STAT3Tyr705 phosphorylation. Cancer Med. 2020, 9, 8685–8700. [Google Scholar] [CrossRef]
  95. Chen, D.; Ma, Y.; Guo, Z.; Liu, L.; Yang, Y.; Wang, Y.; Pan, B.; Wu, L.; Hui, Y.; Yang, W. Two natural alkaloids synergistically induce apoptosis in breast cancer cells by inhibiting STAT3 activation. Molecules 2020, 25, 216. [Google Scholar] [CrossRef] [PubMed]
  96. Xu, J.; Chen, Y.; Yang, R.; Zhou, T.; Ke, W.; Si, Y.; Yang, S.; Zhang, T.; Liu, X.; Zhang, L. Cucurbitacin B inhibits gastric cancer progression by suppressing STAT3 activity. Arch. Biochem. Biophys. 2020, 684, 108314. [Google Scholar] [CrossRef] [PubMed]
  97. Cirmi, S.; Celano, M.; Lombardo, G.E.; Maggisano, V.; Procopio, A.; Russo, D.; Navarra, M. Oleacein inhibits STAT3, activates the apoptotic machinery, and exerts anti-metastatic effects in the SH-SY5Y human neuroblastoma cells. Food Funct. 2020, 11, 3271–3279. [Google Scholar] [CrossRef]
  98. Lu, L.; Li, H.; Wu, X.; Rao, J.; Zhou, J.; Fan, S.; Shen, Q. HJC0152 suppresses human non–small-cell lung cancer by inhibiting STAT3 and modulating metabolism. Cell Prolif. 2020, 53, e12777. [Google Scholar] [CrossRef]
  99. Jin, X.; Wang, C. Costunolide inhibits osteosarcoma growth and metastasis via suppressing STAT3 signal pathway. Biomed. Pharmacother. 2020, 121, 109659. [Google Scholar] [CrossRef]
  100. Jin, Y.; Huynh, D.T.N.; Myung, C.-S.; Heo, K.-S. Ginsenoside Rh1 Prevents Migration and Invasion through Mitochondrial ROS-Mediated Inhibition of STAT3/NF-κB Signaling in MDA-MB-231 Cells. Int. J. Mol. Sci. 2021, 22, 10458. [Google Scholar] [CrossRef]
  101. Zhang, Z.H.; Li, M.Y.; Wang, Z.; Zuo, H.X.; Wang, J.Y.; Xing, Y.; Jin, C.; Xu, G.; Piao, L.; Piao, H. Convallatoxin promotes apoptosis and inhibits proliferation and angiogenesis through crosstalk between JAK2/STAT3 (T705) and mTOR/STAT3 (S727) signaling pathways in colorectal cancer. Phytomedicine 2020, 68, 153172. [Google Scholar] [CrossRef] [PubMed]
  102. Wang, Y.; Yang, C.; Sims, M.M.; Sacher, J.R.; Raje, M.; Deokar, H.; Yue, P.; Turkson, J.; Buolamwini, J.K.; Pfeffer, L.M. SS-4 is a highly selective small molecule inhibitor of STAT3 tyrosine phosphorylation that potently inhibits GBM tumorigenesis in vitro and in vivo. Cancer Lett. 2022, 533, 215614. [Google Scholar] [CrossRef] [PubMed]
  103. Shah, N.H.; Amacher, J.F.; Nocka, L.M.; Kuriyan, J. The Src module: An ancient scaffold in the evolution of cytoplasmic tyrosine kinases. Crit. Rev. Biochem. Mol. Biol. 2018, 53, 535–563. [Google Scholar] [CrossRef] [PubMed]
  104. Ortiz, M.A.; Mikhailova, T.; Li, X.; Porter, B.A.; Bah, A.; Kotula, L. Src family kinases, adaptor proteins and the actin cytoskeleton in epithelial-to-mesenchymal transition. Cell Commun. Signal. 2021, 19, 1–19. [Google Scholar] [CrossRef] [PubMed]
  105. Martellucci, S.; Clementi, L.; Sabetta, S.; Mattei, V.; Botta, L.; Angelucci, A. Src family kinases as therapeutic targets in advanced solid tumors: What we have learned so far. Cancers 2020, 12, 1448. [Google Scholar] [CrossRef]
  106. Tanji, M.; Ishizaki, T.; Ebrahimi, S.; Tsuboguchi, Y.; Sukezane, T.; Akagi, T.; Frame, M.C.; Hashimoto, N.; Miyamoto, S.; Narumiya, S. mDia1 targets v-Src to the cell periphery and facilitates cell transformation, tumorigenesis, and invasion. Mol. Cell. Biol. 2010, 30, 4604–4615. [Google Scholar] [CrossRef]
  107. Williams, K.C.; Coppolino, M.G. SNARE-dependent interaction of Src, EGFR and β1 integrin regulates invadopodia formation and tumor cell invasion. J. Cell Sci. 2014, 127, 1712–1725. [Google Scholar] [CrossRef]
  108. Lin, M.T.; Lin, B.R.; Chang, C.C.; Chu, C.Y.; Su, H.J.; Chen, S.T.; Jeng, Y.M.; Kuo, M.L. IL-6 induces AGS gastric cancer cell invasion via activation of the c-Src/RhoA/ROCK signaling pathway. Int. J. Cancer 2007, 120, 2600–2608. [Google Scholar] [CrossRef]
  109. Xu, S.; Tang, J.; Wang, C.; Liu, J.; Fu, Y.; Luo, Y. CXCR7 promotes melanoma tumorigenesis via Src kinase signaling. Cell Death Dis. 2019, 10, 191. [Google Scholar] [CrossRef]
  110. Dong, C.; Li, X.; Yang, J.; Yuan, D.; Zhou, Y.; Zhang, Y.; Shi, G.; Zhang, R.; Liu, J.; Fu, P. PPFIBP1 induces glioma cell migration and invasion through FAK/Src/JNK signaling pathway. Cell Death Dis. 2021, 12, 827. [Google Scholar] [CrossRef]
  111. Alcalá, S.; Mayoral-Varo, V.; Ruiz-Cañas, L.; López-Gil, J.C.; Heeschen, C.; Martín-Pérez, J.; Sainz, B. Targeting SRC kinase signaling in pancreatic cancer stem cells. Int. J. Mol. Sci. 2020, 21, 7437. [Google Scholar] [CrossRef] [PubMed]
  112. Li, T.; Sun, R.; Lu, M.; Chang, J.; Meng, X.; Wu, H. NDRG3 facilitates colorectal cancer metastasis through activating Src phosphorylation. OncoTargets Ther. 2018, 11, 2843. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  113. Lu, Y.; Dong, B.; Xu, F.; Xu, Y.; Pan, J.; Song, J.; Zhang, J.; Huang, Y.; Xue, W. CXCL1-LCN2 paracrine axis promotes progression of prostate cancer via the Src activation and epithelial-mesenchymal transition. Cell Commun. Signal. 2019, 17, 118. [Google Scholar] [CrossRef] [PubMed]
  114. Xue, Y.; Lai, L.; Lian, W.; Tu, X.; Zhou, J.; Dong, P.; Su, D.; Wang, X.; Cao, X.; Chen, Y. SOX9/FXYD3/Src axis is critical for ER+ breast cancer stem cell function. Mol. Cancer Res. 2019, 17, 238–249. [Google Scholar] [CrossRef]
  115. Gong, W.J.; Liu, J.Y.; Yin, J.Y.; Cui, J.J.; Xiao, D.; Zhuo, W.; Luo, C.; Liu, R.J.; Li, X.; Zhang, W. Resistin facilitates metastasis of lung adenocarcinoma through the TLR 4/Src/EGFR/PI 3K/NF-κB pathway. Cancer Sci. 2018, 109, 2391–2400. [Google Scholar] [CrossRef] [PubMed]
  116. Yu, G.-T.; Mao, L.; Wu, L.; Deng, W.-W.; Bu, L.-L.; Liu, J.-F.; Chen, L.; Yang, L.-L.; Wu, H.; Zhang, W.-F. Inhibition of SRC family kinases facilitates anti-CTLA4 immunotherapy in head and neck squamous cell carcinoma. Cell. Mol. Life Sci. 2018, 75, 4223–4234. [Google Scholar] [CrossRef] [PubMed]
  117. Conlon, G.A.; Murray, G.I. Recent advances in understanding the roles of matrix metalloproteinases in tumour invasion and metastasis. J. Pathol. 2019, 247, 629–640. [Google Scholar] [CrossRef] [PubMed]
  118. Lu, P.; Chen, J.; Yan, L.; Yang, L.; Zhang, L.; Dai, J.; Hao, Z.; Bai, T.; Xi, Y.; Li, Y. RasGRF2 promotes migration and invasion of colorectal cancer cells by modulating expression of MMP9 through Src/Akt/NF-κB pathway. Cancer Biol. Ther. 2019, 20, 435–443. [Google Scholar] [CrossRef]
  119. Yeh, C.M.; Hsieh, M.J.; Yang, J.S.; Yang, S.F.; Chuang, Y.T.; Su, S.C.; Liang, M.Y.; Chen, M.K.; Lin, C.W. Geraniin inhibits oral cancer cell migration by suppressing matrix metalloproteinase-2 activation through the FAK/Src and ERK pathways. Environ. Toxicol. 2019, 34, 1085–1093. [Google Scholar] [CrossRef]
  120. Rivat, C.; Le Floch, N.; Sabbah, M.; Teyrol, I.; Redeuilh, G.; Bruyneel, E.; Mareel, M.; Matrisian, L.M.; Crawford, H.C.; Gespach, C. Synergistic cooperation between the AP-1 and LEF-1 transcription factors in the activation of the matrilysin promoter by the src oncogene: Implications in cellular invasion. FASEB J. 2003, 17, 1721–1723. [Google Scholar] [CrossRef]
  121. Wang, X.; Zhao, X.; Yi, Z.; Ma, B.; Wang, H.; Pu, Y.; Wang, J.; Wang, S. WNT5A promotes migration and invasion of human osteosarcoma cells via SRC/ERK/MMP-14 pathway. Cell Biol. Int. 2018, 42, 598–607. [Google Scholar] [CrossRef] [PubMed]
  122. Jo, M.J.; Kim, B.G.; Kim, W.Y.; Lee, D.-H.; Yun, H.K.; Jeong, S.; Park, S.H.; Kim, B.R.; Kim, J.L.; Kim, D.Y. Cannabidiol Suppresses Angiogenesis and Stemness of Breast Cancer Cells by Downregulation of Hypoxia-Inducible Factors-1α. Cancers 2021, 13, 5667. [Google Scholar] [CrossRef] [PubMed]
  123. Tomita, T.; Kato, M.; Hiratsuka, S. Regulation of vascular permeability in cancer metastasis. Cancer Sci. 2021, 112, 2966–2974. [Google Scholar] [CrossRef] [PubMed]
  124. Pastushenko, I.; Blanpain, C. EMT transition states during tumor progression and metastasis. Trends Cell Biol. 2019, 29, 212–226. [Google Scholar] [CrossRef]
  125. Loh, C.-Y.; Chai, J.Y.; Tang, T.F.; Wong, W.F.; Sethi, G.; Shanmugam, M.K.; Chong, P.P.; Looi, C.Y. The E-cadherin and N-cadherin switch in epithelial-to-mesenchymal transition: Signaling, therapeutic implications, and challenges. Cells 2019, 8, 1118. [Google Scholar] [CrossRef]
  126. Jin, W. Regulation of Src family kinases during colorectal cancer development and its clinical implications. Cancers 2020, 12, 1339. [Google Scholar] [CrossRef]
  127. Guarino, M. Epithelial–mesenchymal transition and tumour invasion. Int. J. Biochem. Cell Biol. 2007, 39, 2153–2160. [Google Scholar] [CrossRef]
  128. Wang, B.; Li, X.; Liu, L.; Wang, M. β-Catenin: Oncogenic role and therapeutic target in cervical cancer. Biol. Res. 2020, 53, 33. [Google Scholar] [CrossRef]
  129. Olea-Flores, M.; Zuñiga-Eulogio, M.; Tacuba-Saavedra, A.; Bueno-Salgado, M.; Sánchez-Carvajal, A.; Vargas-Santiago, Y.; Mendoza-Catalán, M.A.; Pérez Salazar, E.; García-Hernández, A.; Padilla-Benavides, T. Leptin promotes expression of EMT-related transcription factors and invasion in a Src and FAK-dependent pathway in MCF10A mammary epithelial cells. Cells 2019, 8, 1133. [Google Scholar] [CrossRef]
  130. Li, J.; Yang, R.; Yang, H.; Chen, S.; Wang, L.; Li, M.; Yang, S.; Feng, Z.; Bi, J. NCAM regulates the proliferation, apoptosis, autophagy, EMT, and migration of human melanoma cells via the Src/Akt/mTOR/cofilin signaling pathway. J. Cell. Biochem. 2020, 121, 1192–1204. [Google Scholar] [CrossRef]
  131. Cao, X.-Y.; Zhang, X.-X.; Yang, M.-W.; Hu, L.-P.; Jiang, S.-H.; Tian, G.-A.; Zhu, L.-L.; Li, Q.; Sun, Y.-W.; Zhang, Z.-G. Aberrant upregulation of KLK10 promotes metastasis via enhancement of EMT and FAK/SRC/ERK axis in PDAC. Biochem. Biophys. Res. Commun. 2018, 499, 584–593. [Google Scholar] [CrossRef] [PubMed]
  132. Mirzoeva, S.; Franzen, C.A.; Pelling, J.C. Apigenin inhibits TGF-β-induced VEGF expression in human prostate carcinoma cells via a Smad2/3-and Src-dependent mechanism. Mol. Carcinog. 2014, 53, 598–609. [Google Scholar] [CrossRef] [PubMed]
  133. Jain, S.; Wang, X.; Chang, C.-C.; Ibarra-Drendall, C.; Wang, H.; Zhang, Q.; Brady, S.W.; Li, P.; Zhao, H.; Dobbs, J. Src inhibition blocks c-Myc translation and glucose metabolism to prevent the development of breast cancer. Cancer Res. 2015, 75, 4863–4875. [Google Scholar] [CrossRef]
  134. Thakur, R.; Trivedi, R.; Rastogi, N.; Singh, M.; Mishra, D.P. Inhibition of STAT3, FAK and Src mediated signaling reduces cancer stem cell load, tumorigenic potential and metastasis in breast cancer. Sci. Rep. 2015, 5, 10194. [Google Scholar] [CrossRef] [PubMed]
  135. Fan, J.-J.; Hsu, W.-H.; Lee, K.-H.; Chen, K.-C.; Lin, C.-W.; Lee, Y.-L.A.; Ko, T.-P.; Lee, L.-T.; Lee, M.-T.; Chang, M.-S. Dietary flavonoids luteolin and quercetin inhibit migration and invasion of squamous carcinoma through reduction of Src/Stat3/S100A7 signaling. Antioxidants 2019, 8, 557. [Google Scholar] [CrossRef]
  136. Min, T.-R.; Park, H.-J.; Park, M.N.; Kim, B.; Park, S.-H. The Root Bark of Morus alba L. Suppressed the Migration of Human Non-Small-Cell Lung Cancer Cells through Inhibition of Epithelial–Mesenchymal Transition Mediated by STAT3 and Src. Int. J. Mol. Sci. 2019, 20, 2244. [Google Scholar] [CrossRef]
  137. Idoudi, S.; Bedhiafi, T.; Hijji, Y.M.; Billa, N. Curcumin and Derivatives in Nanoformulations with Therapeutic Potential on Colorectal Cancer. AAPS PharmSciTech 2022, 23, 115. [Google Scholar] [CrossRef]
  138. Papierska, K.; Krajka-Kuźniak, V.; Paluszczak, J.; Kleszcz, R.; Skalski, M.; Studzińska-Sroka, E.; Baer-Dubowska, W. Lichen-Derived Depsides and Depsidones Modulate the Nrf2, NF-κB and STAT3 Signaling Pathways in Colorectal Cancer Cells. Molecules 2021, 26, 4787. [Google Scholar] [CrossRef]
Figure 1. States of Nrf2 in normal conditions and under stress by various stimuli. ARE, antioxidant response element; Keap1, Kelch-like ECH-associated protein 1; Maf, avian musculoaponeurotic fibrosarcoma.
Figure 1. States of Nrf2 in normal conditions and under stress by various stimuli. ARE, antioxidant response element; Keap1, Kelch-like ECH-associated protein 1; Maf, avian musculoaponeurotic fibrosarcoma.
Pharmaceutics 14 01775 g001
Figure 2. General mechanism of Nrf2-mediated expression of various detoxification and antioxidant enzymes by potential chemoprevention agents such as phytochemicals.
Figure 2. General mechanism of Nrf2-mediated expression of various detoxification and antioxidant enzymes by potential chemoprevention agents such as phytochemicals.
Pharmaceutics 14 01775 g002
Figure 3. Correlation between Nrf2 and NF-κB in counteracting inflammation-associated cancer. Activation of Nrf2 after binding to antioxidant response element (ARE) leads to the production of antioxidant enzymes (HO-1, UGT, NQO1, GST), which can lead to suppression of NF-κB. This leads to indirect inhibition of NF-κB targets enzymes such as NO, COX-2, and iNOS. The overall effect is a reduction in oxidative stress and inflammation by the Nrf2 pathway, which can serve as a valuable target in preventing inflammation-associated cancer.
Figure 3. Correlation between Nrf2 and NF-κB in counteracting inflammation-associated cancer. Activation of Nrf2 after binding to antioxidant response element (ARE) leads to the production of antioxidant enzymes (HO-1, UGT, NQO1, GST), which can lead to suppression of NF-κB. This leads to indirect inhibition of NF-κB targets enzymes such as NO, COX-2, and iNOS. The overall effect is a reduction in oxidative stress and inflammation by the Nrf2 pathway, which can serve as a valuable target in preventing inflammation-associated cancer.
Pharmaceutics 14 01775 g003
Figure 4. Complex Role of STAT3 in tumorigenesis and its progression.
Figure 4. Complex Role of STAT3 in tumorigenesis and its progression.
Pharmaceutics 14 01775 g004
Figure 5. Upstream stimuli of Src and its downstream target molecules. Src interacts with integrin/focal adhesion kinase (FAK), the receptor of tyrosine kinases (RTKs), and G-protein-coupled receptors (GPCRs), leading to activation of its downstream target proteins, including MAPK/ERK, PI3K, IL-6/JAK/STAT3, and Rho/Rho-associated protein kinase (ROCK), subsequently promoting cancer progression.
Figure 5. Upstream stimuli of Src and its downstream target molecules. Src interacts with integrin/focal adhesion kinase (FAK), the receptor of tyrosine kinases (RTKs), and G-protein-coupled receptors (GPCRs), leading to activation of its downstream target proteins, including MAPK/ERK, PI3K, IL-6/JAK/STAT3, and Rho/Rho-associated protein kinase (ROCK), subsequently promoting cancer progression.
Pharmaceutics 14 01775 g005
Figure 6. Activated SFKs can phosphorylate p120 catenin and FAK, causing destabilization of E-cadherin and adherens junction. The p130cas and paxillin are cell migration mediators. Src-FAK-p130cas axis causes cell invasion via MMP activation. All these cellular changes promote the invasiveness of cells.
Figure 6. Activated SFKs can phosphorylate p120 catenin and FAK, causing destabilization of E-cadherin and adherens junction. The p130cas and paxillin are cell migration mediators. Src-FAK-p130cas axis causes cell invasion via MMP activation. All these cellular changes promote the invasiveness of cells.
Pharmaceutics 14 01775 g006
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Ahsan, H.; Islam, S.U.; Ahmed, M.B.; Lee, Y.S. Role of Nrf2, STAT3, and Src as Molecular Targets for Cancer Chemoprevention. Pharmaceutics 2022, 14, 1775. https://doi.org/10.3390/pharmaceutics14091775

AMA Style

Ahsan H, Islam SU, Ahmed MB, Lee YS. Role of Nrf2, STAT3, and Src as Molecular Targets for Cancer Chemoprevention. Pharmaceutics. 2022; 14(9):1775. https://doi.org/10.3390/pharmaceutics14091775

Chicago/Turabian Style

Ahsan, Haseeb, Salman Ul Islam, Muhammad Bilal Ahmed, and Young Sup Lee. 2022. "Role of Nrf2, STAT3, and Src as Molecular Targets for Cancer Chemoprevention" Pharmaceutics 14, no. 9: 1775. https://doi.org/10.3390/pharmaceutics14091775

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