Next Article in Journal
The Influence of Motion on the Delivery Accuracy When Comparing Actively Scanned Carbon Ions versus Protons at a Synchrotron-Based Radiotherapy Facility
Next Article in Special Issue
Paracrine IL-6 Signaling Confers Proliferation between Heterogeneous Inflammatory Breast Cancer Sub-Clones
Previous Article in Journal
Prostate Cancer and Sleep Disorders: A Systematic Review
Previous Article in Special Issue
Targeting STAT3 Signaling in COL1+ Fibroblasts Controls Colitis-Associated Cancer in Mice
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Cell Type-Specific Roles of STAT3 Signaling in the Pathogenesis and Progression of K-ras Mutant Lung Adenocarcinoma

by
Michael J. Clowers
1,2 and
Seyed Javad Moghaddam
1,2,*
1
Department of Pulmonary Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
2
The University of Texas MD Anderson Cancer Center UTHealth Graduate School of Biomedical Sciences, Houston, TX 77030, USA
*
Author to whom correspondence should be addressed.
Cancers 2022, 14(7), 1785; https://doi.org/10.3390/cancers14071785
Submission received: 14 February 2022 / Revised: 28 March 2022 / Accepted: 29 March 2022 / Published: 31 March 2022
(This article belongs to the Special Issue Multicellular Effects of STAT3 in the Tumour Microenvironment)

Abstract

:

Simple Summary

Lung adenocarcinomas with mutations in the K-ras gene are hard to target pharmacologically and highly lethal. As a result, there is a need to identify other therapeutic targets that influence K-ras oncogenesis. One contender is STAT3, a transcription factor that is associated with K-ras mutations and aids tumor development and progression through tumor cell intrinsic and extrinsic mechanisms. In this review, we summarize the lung epithelial and infiltrating immune cells that express STAT3, the roles of STAT3 in K-ras mutant lung adenocarcinoma, and therapies that may be able to target STAT3.

Abstract

Worldwide, lung cancer, particularly K-ras mutant lung adenocarcinoma (KM-LUAD), is the leading cause of cancer mortality because of its high incidence and low cure rate. To treat and prevent KM-LUAD, there is an urgent unmet need for alternative strategies targeting downstream effectors of K-ras and/or its cooperating pathways. Tumor-promoting inflammation, an enabling hallmark of cancer, strongly participates in the development and progression of KM-LUAD. However, our knowledge of the dynamic inflammatory mechanisms, immunomodulatory pathways, and cell-specific molecular signals mediating K-ras-induced lung tumorigenesis is substantially deficient. Nevertheless, within this signaling complexity, an inflammatory pathway is emerging as a druggable target: signal transducer and activator of transcription 3 (STAT3). Here, we review the cell type-specific functions of STAT3 in the pathogenesis and progression of KM-LUAD that could serve as a new target for personalized preventive and therapeutic intervention for this intractable form of lung cancer.

1. Introduction

Lung cancer exerts a massive burden on public health, resulting in nearly a quarter of global cancer deaths [1]. Lung adenocarcinoma (LUAD) is a major histological subtype of lung cancer, and 30% of patients with LUAD harbor driver mutations in Kirsten rat sarcoma viral oncogene (K-ras), which we refer to as K-ras mutant LUAD (KM-LUAD) [2]. K-ras mutations, most frequently in the 12th codon, result in constitutive activation leading to overactive proliferative signaling pathways, such as the RAF cascade [3]. Targeted inhibitors of K-ras point mutations are under development and are either in early stages or have been complicated by emerging drug resistance [4]. The path to improved outcomes relies on targeting factors that are downstream of or cooperate with K-ras.
KM-LUAD is notable in that K-ras activation in the setting of the lung can lead to a chronically inflamed tumor microenvironment (TME) [5,6]. In normal inflammation, noxious stimuli induce various forms of cell death, and these cellular contents nourish the recruited immune cells that clear the debris and resolve the inflammatory trigger if possible [7,8]. Tumor-promoting inflammation differs from this transient response in that prolonged, chronic exposure to the noxious stimuli (e.g., cigarette smoke, infection) prevents full clearance, setting the stage for tumor initiation [8]. K-ras mutations correlate with increased myeloid lineage immune cell infiltration, including macrophages and neutrophils skewed towards pro-tumor phenotypes [5,9]. In addition to these cell types, the KM-LUAD TME fosters production of interleukin 6 (IL-6), a pleiotropic cytokine with many context-dependent effects [10,11,12,13]. IL-6 utilizes a well-known signaling pathway which results in the activation of the signal transducer and activator of transcription 3 (STAT3) [14]. STAT3 represents a critical inflammatory transcription factor in KM-LUAD, with patients with KM-LUAD tumors expressing high levels of STAT3 and activated phospho-STAT3 (p-STAT3), which correlate with worse survival [15,16]. STAT3 controls the transcription of numerous target genes that promote cell survival, angiogenesis, stemness, etc., but alarmingly, STAT3 has the ability, in concert with other transcription factors, to stimulate the production of IL-6 [13,17,18]. This feedback mechanism ensures a repeating loop of tumor-promoting inflammation. Moreover, STAT3 leads to the transcription of other soluble mediators that reprogram the TME towards a pro-tumor phenotype [19].
Given the difficulties in targeting K-ras driver mutations, tumor-promoting inflammation and the STAT3 pathway are gaining attention as therapeutic targets. However, STAT3 plays key functions across the body, as evidenced by the fact that STAT3 global deletion is early lethal [20]. Furthermore, in the context of the lung environment, STAT3 is expressed in lung resident/infiltrating immune cells and in the lung epithelium, and its functions in these cellular compartments differ yet work in synch [21]. Future therapeutics must take these cell types into account in order to produce maximal benefits. Here, we discuss the principles of STAT3 signaling and how they differ across the cell types found in KM-LUAD.

2. STAT3 Structure and Signaling: The Basics

The structure of STAT3 contains several functional components: a Src 2 homology (SH2) domain for recruitment and dimer formation, a C-terminal transactivation domain harboring a key phosphorylation site for STAT3 activation (Y705), a nuclear localization sequence (NLS), a DNA binding domain to recognize STAT3 inducible elements (SIEs), and a coiled-coiled domain for protein–protein interactions [22].
STAT3 is activated downstream of a plethora of receptor/non-receptor tyrosine kinases and G proteins, with ligands varying from growth factors to cytokines; however, STAT3 is classically regulated by IL-6 (Figure 1). IL-6 binds to a multimeric receptor complex comprising the IL-6 receptor (IL-6R) and gp130. Janus kinases (JAKs) 1 and 2 are recruited to the cytosolic tails of the receptors using SH2-like domains, where they transphosphorylate one another. STAT3 molecules then recognize the phosphorylated sites using their SH2 domains and are themselves phosphorylated at the Y705 site. This posttranslational modification enables STAT3 to form homodimers (or heterodimers with STAT1/5) and translocate to the nucleus, where the dimers bind SIEs to drive transcription [22,23]. In addition, IL-6R can be proteolytically cleaved from the cell surface to form soluble IL-6R (sIL-6R) [24]. sIL-6R can then induce trans signaling in cells that do not endogenously express IL-6R [24].
A host of target genes for STAT3 have been identified and have been extensively reviewed elsewhere [19], but the main responsive genes can be divided into several functional categories: anti-apoptosis, angiogenesis, invasiveness, cell cycle progression, and stemness (Figure 1). These SIE-directed programs give STAT3 signaling a decidedly pro-survival, pro-tumor flavor, which is advantageous in the setting of KM-LUAD. However, STAT3 also has the ability to modulate immune-related genes, including a mixture of pro- and anti-inflammatory mediators. These immune-related genes can lead to different immune phenotypes and as such are context and cell-type dependent; we explore the immunological specifics of these mediators in the next section. STAT3 heterodimers can also attenuate other immune signaling pathways indirectly. STAT1:STAT3 heterodimer formation is known to inhibit classical STAT1/IFN-γ signaling by “soaking up” STAT1 and preventing STAT1 homodimerization [23].
STAT3 transcriptional activity is also subject to other key post-translational modifications beyond p-Y705. S727 phosphorylation, usually mediated by ERK or other kinases, is generally believed to lead to a stronger realization of STAT3 transcriptional activity [25,26]. Acetylation, methylation, and SUMOylation of STAT3 are also regular events which have recently garnered more attention and which we will discuss in later sections [26,27]. However, it should be noted that unphosphorylated STAT3 (U-STAT3) can also translocate to the nucleus to effect changes in gene expression as a delayed response to IL-6 signaling [28].
STAT3 transcriptional targets are not limited to SIE-regulated genes. Using its coiled-coiled domain, STAT3 can interact with other transcription factors to expand its target gene repertoire. Famously, STAT3 interaction with the pro-inflammatory NF-κB transcription factor leads to expression of IL-6 [18]. This ability to induce IL-6 creates a positive feedback loop which can stimulate more and more STAT3. To counteract this loop, STAT3 also negatively regulates its own expression by transcribing the suppressor of cytokine signaling 3 (SOCS3) [29]. Moreover, phosphatases and protein inhibitors of activated STAT (PIAS) families work to nullify p-STAT3 signaling, the result of which is a relatively short half-life for p-STAT3 molecules [30,31,32].
Now that we have an overview of the mechanisms of STAT3 signaling, we explore the intricacies of this pathway in the cell types found in KM-LUAD.

3. Studying STAT3 by Cell Type in KM-LUAD Models

Multiple cell types comprise the KM-LUAD TME, principally tumor and immune cells. To study STAT3 signaling in these cell types in a preclinical setting, several models are employed (Table 1).
LUAD-derived cell lines, many of which display heightened STAT3 pathway activity, can represent the tumor component of the TME in vitro [33]. Coculture and/or gel matrix culture with immune cells can help to recapitulate the cell–cell interactions in the TME. These cells can be engineered to knock down or overexpress STAT3, and drugs against STAT3 or other cooperating pathways can be pioneered.
In vivo models offer a more realistic glimpse of the TME. STAT3 global knockout is early lethal, but a variety of conditional knockout systems can be used [20]. In lung tissue, Cre recombinase expression can be directed by the SPC or CCSP promoters, with SPC expressed more broadly and CCSP more tightly targeted to the conducting airway epithelium, where most KM-LUAD tumors arise [6,34]. The Cre can be temporally controlled (e.g., doxycycline-induced) if there are concerns about the timing of Cre expression [35]. Mutant K-ras is then expressed in a Cre-dependent manner and will kickstart tumorigenesis [36]. Additional genes of interest can be floxed to conditionally delete them within tumors and the lung epithelium. The same constructs can be introduced by viral vectors into the airways of adult mice, with the main side effect being potential antiviral immune artifacts [37,38]. On top of these genetic models, cigarette smoke, vaping, nicotine-derived nitrosamine ketone (NNK), and urethane can be administered to mimic smoking [39]. Products of certain bacterial species (e.g., Nontypeable Haemophilus influenzae (NTHi) lysate) can be introduced to recapitulate chronic obstructive pulmonary disease (COPD) and the pro-tumor inflammation which it engenders [6,39,40]. To study STAT3 in immune cells, a Cre that targets immune cell compartments can be used, most notably LysMCre, a myeloid-specific driver [41].
These models have been used to a great extent to dissect the cell type-specific roles of STAT3 in KM-LUAD and are referenced in the sections that follow as we look at STAT3 functions cell by cell within the TME (Figure 2).

4. Cell-Specific Functions of STAT3 in KM-LUAD

4.1. Lung Epithelium/Tumor Cells

In healthy lung tissues, STAT3 plays important roles in lung development and differentiation, including branch morphogenesis and lung epithelial cell identity [42,43]. STAT3 is also a crucial component of the epithelial response to infection. STAT3 activation engages pro-survival machinery to reduce cell death while simultaneously triggering immune responses. The epithelium itself shows STAT3-mediated protective effects against a wide array of pathogens, but it is also able to secret immune mediators that activate features of both mucosal and suppressive immunity to preserve lung architecture and, consequently, lung function [44,45].
LUAD arises from distal regions of lung epithelium [46]. Once K-ras is mutated, cellular proliferation runs rampant as pathways such as RAF are hyperactivated [3]. At this time, STAT3 can be activated intrinsically via K-ras downstream kinase activity at the S727 site or extrinsically by cytokine signaling [26,47,48,49].
STAT3 target genes can improve the survival, proliferation, and immune evasion of early cancer cells. The Bcl and cyclin D family of genes are downstream of STAT3 and block apoptosis and drive cell cycle progression, respectively [50,51]. Moreover, the self-renewal ability of lung cancer stem cells appears to be driven in part by STAT3 [19]. STAT3 also increases the expression of immune checkpoint molecules including PD-L1, which serves to shield the cancer from antigen-specific T cell immunity [52]. Additionally, there is increasing interest in the ability of phosphorylated and acetylated STAT3 to localize to the mitochondria (Figure 1), where it increases mitochondrial gene transcription and ATP production, aiding the Warburg Effect in cancer cells [53]. Moreover, STAT3, in conjunction with NF-κB, can activate the IL-6 positive feedback loop and potentiate an autocrine signaling cycle [13]. At the same time, lung cancer cells often downregulate STAT3 inhibitor SOCS3 which exacerbates the IL-6/STAT3 loop; overexpression of SOCS3 and PIAS can restore normal tumor suppression ability [54,55]. Studies have shown that overexpression of constitutively active STAT3 in murine alveolar type II cells is sufficient to induce a strong inflammatory response and produces LUAD-like tumors [56,57].
Hyperactivation of STAT3 signaling also leads to the paracrine secretion of many STAT3-regulated elements and, in turn, a shift in the TME. Matrix metalloproteinases (MMPs) are able to break down the extracellular matrix to improve invasion and metastasis, and nascent tumors gain increased blood flow through VEGF-induced angiogenesis [19]. STAT3 also directly binds to the promotors of two immunosuppressive cytokines, IL-10 and TGF-β, which skew several immune cell populations from pro- to anti-inflammatory [19]. Simultaneously, pro-inflammatory IL-23 is produced and alters T cell functional phenotypes towards pro-tumor, which we explore later [19].
While STAT3 hyperactivation is largely believed to encourage tumor development and progression, this paradigm is clouded by some data suggesting a tumor suppressor role for STAT3. One study has found that STAT3 in the lung epithelium maintains epithelial characteristics and discourages epithelial-to-mesenchymal transition (EMT) [43]. Another group, using an adenoviral Cre model, observed a tumor suppressor function for STAT3 [58]. Still, other work suggests that STAT3 functions as a tumor suppressor in lung tumor initiation but behaves as an oncogene in established tumors [59]. Our group has also identified a sex disparity in which STAT3 deletion in the lung epithelium of a KM-LUAD mouse model is beneficial for females but harmful for males [60]. This sex disparity is characterized by increased NF-κB signaling in males lacking epithelial STAT3, which drives tumor-promoting inflammation. Introducing exogenous estrogen improved outcomes for males, indicating a potential mechanism for estrogen and estrogen receptors to mitigate NK-κB-modulated inflammation [61]. Correlations have been made with LUAD incidence by sex, with female patients experiencing a greater incidence of lung cancer but better overall survival compared to males [62]. Additionally, a recent study suggests that U-STAT3 can suppress lung cancer tumorigenesis through heterochromatin silencing of cell growth genes [63]. While these stories are still unfolding, it is necessary to avoid a purely negative view of STAT3 and pay attention to chronology, sex, or other factors that may influence the role of STAT3 in tumor development and progression.
On the whole, STAT3 hyperactivation in KM-LUAD cancer cells confers survival advantages and initiates a cascade of autocrine and paracrine signaling that alters the TME. Secreted factors in the TME interact with and recruit immune cells, where STAT3 signaling also plays crucial roles in generating tumor-promoting inflammation and immunosuppression.

4.2. Cancer-Associated Fibroblasts

Cancer-associated fibroblasts (CAFs) are stromal, non-malignant, non-immune lineage cells that contribute to the composition of the TME [64]. IL-6/STAT3 signaling is known to be active in CAFs, and a portion of the IL-6, VEGF, and MMPs produced in the TME is CAF-derived [65,66]. In vitro work suggests that STAT3 CAF signaling can enhance the metastatic potential of lung cancer cells, and IL-11/STAT3 signaling may play a role in cisplatin chemoresistance [66,67].

4.3. Macrophages

Macrophages comprise a major portion of the immune cell compartment in the lung. In homeostatic conditions, macrophages are responsible for clearing debris and pathogens to maintain gas exchange function [45]. Lung macrophages are classified based on their physical location within the lung (alveolar or interstitial) and whether or not they are classically or non-classically activated (M1 or M2 respectively) [68,69]. As the front lines of mucosal immunity, lung macrophages are programmed differently than in other tissues: to maintain lung homeostasis, lung macrophages phagocytose debris and pathogens without producing pro-inflammatory mediators, in line with an M2-like phenotype. In fact, these macrophages are major sources of the immunosuppressive cytokines IL-10 and TGF-β in the lung milieu [68].
While IL-10 and TGF-β are important in lung homeostasis, they are a decidedly pro-tumor component in the context of the TME. As previously mentioned, STAT3 signaling in tumor cells leads to the secretion of IL-10 and TGF-β, and this combined with tumor-associated macrophage (TAM) signaling creates a strongly immunosuppressive environment for existing and newly recruited immune cells. Moreover, these M2 macrophages produce VEGF, growth factors, and T cell metabolic inhibitors: arginase-1 and indoleamine 2,3-dioxygenase (IDO) [70,71].
Studies suggest that M2 polarization is in part driven by STAT3 expression. STAT1:STAT3 heterodimerization hampers the anti-tumor M1 phenotype [71]. Genetic ablation of STAT3 in macrophages results in increased pro-inflammatory cytokine production and increased antigen presentation to T cells [72]. In a urethane-induced lung cancer model, myeloid STAT3 deletion resulted in improved anti-tumor immunity, with M2 macrophages replaced by the M1 phenotype [73]. However, at least one study reports that macrophage and myeloid STAT3 hyperactivation offers anti-tumor benefits. In this study, the authors deleted the STAT3 negative regulator SOCS3, which resulted in unchecked STAT3 signaling in macrophages but overall anti-tumor effects [74]. These experiments, however, utilized a metastatic melanoma model rather than an in situ model of lung cancer. As such, the results may not translate to lung cancer development and progression in the context of tumor-promoting inflammation. In our KM-LUAD model, we see the majority of immune cells in the lung are macrophages and that they skew towards an M2 phenotype [15].

4.4. Neutrophils

Neutrophils are the first recruited immune cells during lung infection and are specialists in phagocytosis; however, they become a hinderance in the context of LUAD. Neutrophils are an abundant immune cell type in lung cancers, and we and others found a correlation with K-ras mutations and neutrophil recruitment in KM-LUAD models and non-small cell lung cancer (NSCLC) at large [6,75,76]. Like macrophages, neutrophils can skew between N1 and N2 phenotypes, with N2 producing TGF-β and representing a pro-tumor phenotype [77]. N2 neutrophils also parallel M2 macrophages and produce VEGF, MMPs, and reactive oxygen species (ROS) to promote tumor survival [78,79,80]. Additionally, neutrophils produce a unique immunosuppressive molecule: neutrophil elastase (NE). NE, along with heparanase and collagenase IV, remodel the extracellular matrix to promote invasion and metastasis [81]. Genetic deletion of NE in a KM-LUAD model reduced immunosuppression in the TME and reduced tumor burden [75,82].

4.5. Myeloid-Derived Suppressor Cells

Myeloid-derived suppressor cells (MDSCs) represent a heterogeneous group of immature myeloid immune cells which possess anti-inflammatory functions. They are further sub-characterized as either polymorphonuclear (PMN-MDSCs) or monocytic (M-MDSCs), based on their phenotypic and functional overlap with neutrophils and monocytes, respectively [83]. MDSCs are recruited by various tumor-derived chemotactic factors, including IL-8, IL-17A, and CCL3 [84]. The leading consensus is that STAT3 functions as a master transcription factor in MDSC development and function [85]. Much like M2 macrophages, MDSCs produce STAT3 transcriptional targets, including IL-10, TGF-β, VEGF, arginase-1, and IDO, to generate an immunosuppressed, pro-tumor TME [86,87]. Moreover, MDSCs induce immunosuppression with nitric oxide (NO), ROS, and PD-L1 [88,89,90]. Studies in which myeloid STAT3 is ablated show that the myeloid compartment shifts from STAT3- to STAT1-driven signaling, thereby skewing to an anti-tumor response [72].

4.6. T Cells

T cells are adaptive immune cells that are tailored to recognize specific targets. There are two broad classes: CD4+ helper T (Th) and CD8+ cytotoxic T lymphocytes (CTLs). While CTLs perform the actual work of killing tumors cell by cell, Th cells produce specialized cytokines that support different flavors of immune responses: Th1 (antiviral/anti-tumor), Th2 (anti-parasite/allergy), Th17 (mucosal immunity), Treg (immunosuppression). Favorable cell types for targeting tumors are CTLs and Th1, which elicit high STAT1 signaling. These cells can directly induce tumor apoptosis via granzyme B (CTLs only) and production of IFNγ and TNFα, two cytokines which broadly reprogram the TME and can induce apoptosis in tumor cells [91,92]. On the other hand, Th2, Th17, and Tregs are generally pro-tumor. Within these diverse subtypes, STAT3 signaling is a crucial factor that skews T cells towards pro-tumor Th17 and Treg immunity while also increasing PD-L1 expression, yielding functional exhaustion [52,93].
Th17 differentiation is triggered in response to IL-6 and TGF-β, two cytokines which are downstream of STAT3 and which are found in abundance in the KM-LUAD TME [93]. These cytokines lead to activation of STAT3, which activates the ROR family of transcription factors to drive Th17 fate [94]. T cells that lack STAT3 cannot become Th17 cells and instead differentiate into Th1 cells [95]. Fully fledged Th17 cells are later maintained by IL-23, another STAT3-controlled cytokine found in the TME [96].
True to their name, Th17 cells produce IL-17A, a cytokine which leads to production of IL-8 [97]. Both IL-17A and IL-8 are major chemotactic factors for neutrophils and MDSCs [84,98,99]. Moreover, IL-17A signaling contributes to paracrine TME reprogramming, increased inflammation, and angiogenesis [100,101]. In our own studies, deletion of IL-17A in a KM-LUAD mouse model reduced tumor burden, inflammation, and pro-tumor myeloid cell recruitment and infiltration [102]. Th17 cells can also inhibit other anti-tumor T cells by cleaving extracellular ATP into adenosine, which is immunosuppressive [103].
It is important to note that non-classical γδ T cells and type 3 innate lymphoid cells (ILC3s) are major producers of IL-17A [104]. However, ILC3s have been reported to form tertiary lymphoid structures (TLSs) in lung cancer that are positively prognostic [105]. It should be noted that IL-17A can also play an anti-tumor role, depending on the type of cancer [106]. In additional to making IL-17A, CD4+ and γδ T cells also produce IL-22, a cytokine which acts on non-immune cells and heavily activates the STAT3 pathway [107]. Lung cancer patients present with elevated IL-22 in lung lavage samples [108]. We have shown that deletion of IL-22 in a KM-LUAD mouse model suppressed tumorigenesis, altered the TME to an anti-tumor phenotype, and reduced stemness properties of tumor cells [109].
Treg differentiation is driven by exposure to TGF-β and subsequent activation of FoxP3, a transcription factor that induces and maintains the Treg subtype [110]. Tregs can express immune checkpoint molecules that directly inactivate neighboring effector T cells. They can release immunosuppressive molecules including IL-10, TGF-β, adenosine, etc. They also metabolically compete with anti-tumor immune cells [111]. Treg infiltration is almost universally a negative prognostic marker across cancer types [112]. When knocking down STAT3 in T cells, Tregs are less abundant, indicating a reliance on STAT3 [113].

4.7. Dendritic Cells

Dendritic cells (DCs) are a key bridge between innate and adaptive immunity and prime antigen-specific T cells [114]. The various DC subtypes and their roles in cancer and immunotherapy have been extensively reviewed elsewhere [115], but the principal DC subtypes of relevance are classical DC type 1 (cDC1) and 2 (cDC2). These cDC types largely correlate with Th subsets: cDC1s are considered anti-tumor and prime tumor-specific CTL and Th1 responses. cDC2s, on the other hand, are more heterogeneous in function, but they are largely thought to prime Th2, Th17, and Treg responses, which represents a pro-tumor skewing.
DC maturation often depends on STAT3 [116,117]. However, deletion of STAT3 in the DC compartment leads to fewer but more functional DCs capable of activating robust T cell responses [41]. STAT3-ablated DCs demonstrate increased IL-12 secretion necessary for Th1 differentiation, and tumors with STAT3 inhibited DCs show decreased Treg infiltration with commensurate increases in intratumoral CTLs and repolarization toward type I immunity [41,118]. Interestingly, acetylated STAT3 has been implicated as a precursor to IDO production in DCs, suggesting an additional immunosuppressive mechanism for STAT3 [119].

4.8. Natural Killer Cells

Natural killer (NK) cells are directly cytotoxic immune cells that recognize their targets independent of MHC presentation [120]. NK function is largely inhibited in KM-LUAD by immunosuppressive factors such as IL-10, TGF-β, and IDO [121]. Deletion of STAT3 in the NK compartment drastically improved tumor rejection, immune surveillance, and IFN-γ secretion abilities, and targeting STAT3 with microRNA-130a augmented NK cytotoxicity against NSCLC cells [122,123]. Similarly, deletion of lung epithelial STAT3 in a urethane-induced LUAD model improved NK cell tumor killing function [57].

4.9. B Cells

B cells have recently come under investigation in lung cancer, displaying both pro- and anti-tumor traits. STAT3 appears to be necessary for regulatory B (Breg) cells, a B cell counterpart to Tregs that produces anti-inflammatory IL-10 and is pro-tumor [124]. B cell-specific deletion of STAT3 causes severe autoimmunity in mice, so we can infer potential anti-tumor benefits from targeting STAT3 signaling in B cells [125]. Moreover, Bregs are found more frequently in lung cancer patients [126]. One study suggests that STAT3-regulated CD5 expression on B cells drives IL-6 signaling and disease progression in lung cancer [127].
On the other hand, B cells have the ability to form structures which histologically and functionally resemble lymph nodes and lymphoid tissue, so-called tertiary lymphoid structures (TLSs). TLSs are associated with increased Th1 polarization, CTL infiltration, and DCs in NSCLC. Much like true lymphoid tissue, TLSs develop germinal centers, a process which depends on STAT3 [128].

5. STAT3 Inhibition

In light of the many negative effects of hyperactive STAT3 in the TME, a number of therapeutic interventions have been developed that target different stages of STAT3 signaling. Many of these drugs are already in or finishing clinical trials and have been extensively reviewed elsewhere [129,130]. However, we want to touch on a few of these which hold the most promise.
Multiple small molecule STAT3 inhibitors have been tested in the last decade, and there are still many novel inhibitors which may one day reach clinical application [130,131]. One such STAT3 inhibitor, TTI-101, binds to the SH2 domain to prevent STAT3 homodimerization [132]. TTI-101 has shown preclinical benefits in various mouse cancer models including lung cancer xenografts, and recruitment for clinical trials has begun (NCT03195699) [133]. AZD9150 is an anti-sense oligonucleotide therapy targeting STAT3 on the RNA level. AZD9150 improves anti-tumor immunity in patient-derived lung cancer xenografts [134]. Multiple clinical trials are underway to combine AZD9150 with an immune checkpoint blockade (NCT02983578, NCT03334617, NCT03421353).
It is easier to block cytokines than to target a transcription factor. Guided by this philosophy, a monoclonal antibody (mAb) against IL-6, ALD518, completed phase II trials in 2011 in patients with advanced NSCLC (NCT00866970). ALD518 helped patients maintain body weight while reducing fatigue and cancer-related anemia [135]. A forthcoming clinical trial will test the synergistic effects of Tocilizumab (anti-IL-6) and Atezolizumab (anti-PD-L1) in patients with NSCLC who have previously failed immune checkpoint blockade and are experiencing advanced or metastatic disease (NCT04691817).

6. Conclusions

Despite advances in cancer therapies, there is rarely a one-treatment solution, and KM-LUAD is no exception. To study the complex cell types, signaling pathways, and possible treatment combinations, we need to make full use of the available models that exist and to test treatments that attack multiple aspects of the TME. KM-LUAD models are vital to understanding the mechanisms of STAT3 and to appraise therapies that block the IL-6/STAT3 signaling pathway across the variety of cancer and immune cells within the TME. The importance of cell types will feature prominently in future treatment strategies. For instance, global STAT3 targeting in the TME may be beneficial in the T cell compartment but harmful for B cells; ways to deliver treatments in a targeted manner will take advantage of the diversity within the TME to yield optimal anti-tumor responses. In our own group, we have also seen that targeting STAT3 in tumors depends greatly on the biological context, with sex hormones playing a greater role than we foresaw. By cell type-specific interrogation of the TME, we are creating a mosaic map and learning how to better tailor future interventions.

Author Contributions

Conceptualization, M.J.C. and S.J.M., writing—original draft preparation, M.J.C.; writing—review and editing, M.J.C. and S.J.M.; project administration, S.J.M. All authors have read and agreed to the published version of the manuscript.

Funding

The authors were supported by the National Cancer Institute (NCI) grant R01CA225977 (to S.J.M.).

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Siegel, R.L.; Miller, K.D.; Fuchs, H.E.; Jemal, A. Cancer Statistics, 2021. CA Cancer J. Clin. 2021, 71, 7–33. [Google Scholar] [CrossRef] [PubMed]
  2. Goldstraw, P.; Ball, D.; Jett, J.R.; Le Chevalier, T.; Lim, E.; Nicholson, A.G.; Shepherd, F.A. Non-small-cell lung cancer. Lancet 2011, 378, 1727–1740. [Google Scholar] [CrossRef]
  3. Jančík, S.; Drábek, J.; Radzioch, D.; Hajdúch, M. Clinical relevance of KRAS in human cancers. BioMed Res. Int. 2010, 2010, 150960. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  4. Mayekar, M.K.; Bivona, T.G. Current landscape of targeted therapy in lung cancer. Clin. Pharmacol. Ther. 2017, 102, 757–764. [Google Scholar] [CrossRef]
  5. Ji, H.; Houghton, A.; Mariani, T.; Perera, S.; Kim, C.; Padera, R.; Tonon, G.; McNamara, K.; Marconcini, L.; Hezel, A. K-ras activation generates an inflammatory response in lung tumors. Oncogene 2006, 25, 2105–2112. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  6. Moghaddam, S.J.; Li, H.; Cho, S.-N.; Dishop, M.K.; Wistuba, I.I.; Ji, L.; Kurie, J.M.; Dickey, B.F.; De Mayo, F.J. Promotion of lung carcinogenesis by chronic obstructive pulmonary disease—Like airway inflammation in a K-ras—Induced mouse model. Am. J. Respir. Cell Mol. Biol. 2009, 40, 443–453. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  7. Yang, Y.; Jiang, G.; Zhang, P.; Fan, J. Programmed cell death and its role in inflammation. Mil. Med. Res. 2015, 2, 12. [Google Scholar] [CrossRef] [Green Version]
  8. Greten, F.R.; Grivennikov, S.I. Inflammation and Cancer: Triggers, Mechanisms, and Consequences. Immunity 2019, 51, 27–41. [Google Scholar] [CrossRef]
  9. Busch, S.E.; Hanke, M.L.; Kargl, J.; Metz, H.E.; MacPherson, D.; Houghton, A.M. Lung Cancer Subtypes Generate Unique Immune Responses. J. Immunol. 2016, 197, 4493–4503. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  10. Ryan, B.M.; Pine, S.R.; Chaturvedi, A.K.; Caporaso, N.; Harris, C.C. A combined prognostic serum interleukin-8 and interleukin-6 classifier for stage 1 lung cancer in the prostate, lung, colorectal, and ovarian cancer screening trial. J. Thorac. Oncol. 2014, 9, 1494–1503. [Google Scholar] [CrossRef] [Green Version]
  11. Barrera, L.; Montes-Servín, E.; Barrera, A.; Ramírez-Tirado, L.A.; Salinas-Parra, F.; Bañales-Méndez, J.L.; Sandoval-Ríos, M.; Arrieta, Ó. Cytokine profile determined by data-mining analysis set into clusters of non-small-cell lung cancer patients according to prognosis. Ann. Oncol. 2015, 26, 428–435. [Google Scholar] [CrossRef] [PubMed]
  12. Brooks, G.D.; McLeod, L.; Alhayyani, S.; Miller, A.; Russell, P.A.; Ferlin, W.; Rose-John, S.; Ruwanpura, S.; Jenkins, B.J. IL6 Trans-signaling Promotes KRAS-Driven Lung Carcinogenesis. Cancer Res. 2016, 76, 866–876. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  13. Haura, E.B.; Livingston, S.; Coppola, D. Autocrine interleukin-6/interleukin-6 receptor stimulation in non-small-cell lung cancer. Clin. Lung Cancer 2006, 7, 273–275. [Google Scholar] [CrossRef]
  14. Johnson, D.E.; O’Keefe, R.A.; Grandis, J.R. Targeting the IL-6/JAK/STAT3 signalling axis in cancer. Nat. Rev. Clin. Oncol. 2018, 15, 234–248. [Google Scholar] [CrossRef]
  15. Caetano, M.S.; Zhang, H.; Cumpian, A.M.; Gong, L.; Unver, N.; Ostrin, E.J.; Daliri, S.; Chang, S.H.; Ochoa, C.E.; Hanash, S.; et al. IL6 Blockade Reprograms the Lung Tumor Microenvironment to Limit the Development and Progression of K-ras-Mutant Lung Cancer. Cancer Res. 2016, 76, 3189–3199. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  16. Tong, M.; Wang, J.; Jiang, N.; Pan, H.; Li, D. Correlation between p-STAT3 overexpression and prognosis in lung cancer: A systematic review and meta-analysis. PLoS ONE 2017, 12, e0182282. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  17. Yu, H.; Pardoll, D.; Jove, R. STATs in cancer inflammation and immunity: A leading role for STAT3. Nat. Rev. Cancer 2009, 9, 798–809. [Google Scholar] [CrossRef] [PubMed]
  18. Grivennikov, S.; Karin, M. Autocrine IL-6 signaling: A key event in tumorigenesis? Cancer Cell 2008, 13, 7–9. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  19. Carpenter, R.L.; Lo, H.W. STAT3 Target Genes Relevant to Human Cancers. Cancers 2014, 6, 897–925. [Google Scholar] [CrossRef] [Green Version]
  20. Takeda, K.; Noguchi, K.; Shi, W.; Tanaka, T.; Matsumoto, M.; Yoshida, N.; Kishimoto, T.; Akira, S. Targeted disruption of the mouse Stat3 gene leads to early embryonic lethality. Proc. Natl. Acad. Sci. USA 1997, 94, 3801–3804. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  21. Dutta, P.; Sabri, N.; Li, J.; Li, W.X. Role of STAT3 in lung cancer. JAKSTAT 2014, 3, e999503. [Google Scholar] [CrossRef] [PubMed]
  22. Heinrich, P.C.; Behrmann, I.; Haan, S.; Hermanns, H.M.; Müller-Newen, G.; Schaper, F. Principles of interleukin (IL)-6-type cytokine signalling and its regulation. Biochem. J. 2003, 374 Pt 1, 1–20. [Google Scholar] [CrossRef] [Green Version]
  23. Delgoffe, G.M.; Vignali, D.A. STAT heterodimers in immunity: A mixed message or a unique signal? JAKSTAT 2013, 2, e23060. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  24. Jones, S.A.; Scheller, J.; Rose-John, S. Therapeutic strategies for the clinical blockade of IL-6/gp130 signaling. J. Clin. Investig. 2011, 121, 3375–3383. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  25. Zhang, J.J.; Zhao, Y.; Chait, B.T.; Lathem, W.W.; Ritzi, M.; Knippers, R.; Darnell, J.E. Ser727-dependent recruitment of MCM5 by Stat1alpha in IFN-gamma-induced transcriptional activation. EMBO J. 1998, 17, 6963–6971. [Google Scholar] [CrossRef] [Green Version]
  26. Tesoriere, A.; Dinarello, A.; Argenton, F. The Roles of Post-Translational Modifications in STAT3 Biological Activities and Functions. Biomedicines 2021, 9, 956. [Google Scholar] [CrossRef] [PubMed]
  27. Zhou, Z.; Wang, M.; Li, J.; Xiao, M.; Chin, Y.E.; Cheng, J.; Yeh, E.T.; Yang, J.; Yi, J. SUMOylation and SENP3 regulate STAT3 activation in head and neck cancer. Oncogene 2016, 35, 5826–5838. [Google Scholar] [CrossRef] [Green Version]
  28. Yang, J.; Liao, X.; Agarwal, M.K.; Barnes, L.; Auron, P.E.; Stark, G.R. Unphosphorylated STAT3 accumulates in response to IL-6 and activates transcription by binding to NFkappaB. Genes Dev. 2007, 21, 1396–1408. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  29. Krebs, D.L.; Hilton, D.J. SOCS proteins: Negative regulators of cytokine signaling. Stem Cells 2001, 19, 378–387. [Google Scholar] [CrossRef] [PubMed]
  30. Shuai, K.; Liu, B. Regulation of gene-activation pathways by PIAS proteins in the immune system. Nat. Rev. Immunol. 2005, 5, 593–605. [Google Scholar] [CrossRef] [PubMed]
  31. Xu, D.; Qu, C.K. Protein tyrosine phosphatases in the JAK/STAT pathway. Front. Biosci. 2008, 13, 4925–4932. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  32. Wormald, S.; Zhang, J.G.; Krebs, D.L.; Mielke, L.A.; Silver, J.; Alexander, W.S.; Speed, T.P.; Nicola, N.A.; Hilton, D.J. The comparative roles of suppressor of cytokine signaling-1 and -3 in the inhibition and desensitization of cytokine signaling. J. Biol. Chem. 2006, 281, 11135–11143. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  33. Looyenga, B.D.; Hutchings, D.; Cherni, I.; Kingsley, C.; Weiss, G.J.; Mackeigan, J.P. STAT3 is activated by JAK2 independent of key oncogenic driver mutations in non-small cell lung carcinoma. PLoS ONE 2012, 7, e30820. [Google Scholar] [CrossRef] [PubMed]
  34. Lin, C.; Song, H.; Huang, C.; Yao, E.; Gacayan, R.; Xu, S.M.; Chuang, P.T. Alveolar type II cells possess the capability of initiating lung tumor development. PLoS ONE 2012, 7, e53817. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  35. Tichelaar, J.W.; Lu, W.; Whitsett, J.A. Conditional expression of fibroblast growth factor-7 in the developing and mature lung. J. Biol. Chem. 2000, 275, 11858–11864. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  36. Fisher, G.H.; Wellen, S.L.; Klimstra, D.; Lenczowski, J.M.; Tichelaar, J.W.; Lizak, M.J.; Whitsett, J.A.; Koretsky, A.; Varmus, H.E. Induction and apoptotic regression of lung adenocarcinomas by regulation of a K-Ras transgene in the presence and absence of tumor suppressor genes. Genes Dev. 2001, 15, 3249–3262. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  37. Drosten, M.; Guerra, C.; Barbacid, M. Genetically Engineered Mouse Models of K-Ras-Driven Lung and Pancreatic Tumors: Validation of Therapeutic Targets. Cold Spring Harb. Perspect. Med. 2018, 8, a031542. [Google Scholar] [CrossRef] [PubMed]
  38. Anghelina, D.; Lam, E.; Falck-Pedersen, E. Diminished Innate Antiviral Response to Adenovirus Vectors in cGAS/STING-Deficient Mice Minimally Impacts Adaptive Immunity. J. Virol. 2016, 90, 5915–5927. [Google Scholar] [CrossRef] [Green Version]
  39. Tanner, L.; Single, A.B. Animal Models Reflecting Chronic Obstructive Pulmonary Disease and Related Respiratory Disorders: Translating Pre-Clinical Data into Clinical Relevance. J. Innate Immun. 2020, 12, 203–225. [Google Scholar] [CrossRef] [PubMed]
  40. Zheng, H.C.; Takano, Y. NNK-Induced Lung Tumors: A Review of Animal Model. J. Oncol. 2011, 2011, 635379. [Google Scholar] [CrossRef] [Green Version]
  41. Kortylewski, M.; Kujawski, M.; Wang, T.; Wei, S.; Zhang, S.; Pilon-Thomas, S.; Niu, G.; Kay, H.; Mulé, J.; Kerr, W.G.; et al. Inhibiting Stat3 signaling in the hematopoietic system elicits multicomponent antitumor immunity. Nat. Med. 2005, 11, 1314–1321. [Google Scholar] [CrossRef] [PubMed]
  42. Maeda, Y.; Davé, V.; Whitsett, J.A. Transcriptional control of lung morphogenesis. Physiol. Rev. 2007, 87, 219–244. [Google Scholar] [CrossRef] [PubMed]
  43. D’Amico, S.; Shi, J.; Martin, B.L.; Crawford, H.C.; Petrenko, O.; Reich, N.C. STAT3 is a master regulator of epithelial identity and KRAS-driven tumorigenesis. Genes Dev. 2018, 32, 1175–1187. [Google Scholar] [CrossRef] [Green Version]
  44. Ware, H.H.; Kulkarni, V.V.; Wang, Y.; Pantaleón García, J.; Leiva Juarez, M.; Kirkpatrick, C.T.; Wali, S.; Syed, S.; Kontoyiannis, A.D.; Sikkema, W.K.A.; et al. Inducible lung epithelial resistance requires multisource reactive oxygen species generation to protect against bacterial infections. PLoS ONE 2019, 14, e0208216. [Google Scholar] [CrossRef] [Green Version]
  45. Kumar, V. Pulmonary Innate Immune Response Determines the Outcome of Inflammation During Pneumonia and Sepsis-Associated Acute Lung Injury. Front. Immunol. 2020, 11, 1722. [Google Scholar] [CrossRef]
  46. Hanna, J.M.; Onaitis, M.W. Cell of origin of lung cancer. J. Carcinog. 2013, 12, 6. [Google Scholar] [CrossRef] [PubMed]
  47. Barnes, P.J. The cytokine network in chronic obstructive pulmonary disease. Am. J. Respir. Cell Mol. Biol. 2009, 41, 631–638. [Google Scholar] [CrossRef] [PubMed]
  48. Decker, T.; Kovarik, P. Serine phosphorylation of STATs. Oncogene 2000, 19, 2628–2637. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  49. Alhayyani, S.; McLeod, L.; West, A.C.; Balic, J.J.; Hodges, C.; Yu, L.; Smith, J.A.; Prodanovic, Z.; Bozinovski, S.; Kumar, B.; et al. Oncogenic dependency on STAT3 serine phosphorylation in KRAS mutant lung cancer. Oncogene 2022, 41, 809–823. [Google Scholar] [CrossRef]
  50. Karni, R.; Jove, R.; Levitzki, A. Inhibition of pp60c-Src reduces Bcl-XL expression and reverses the transformed phenotype of cells overexpressing EGF and HER-2 receptors. Oncogene 1999, 18, 4654–4662. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  51. Masuda, M.; Suzui, M.; Yasumatu, R.; Nakashima, T.; Kuratomi, Y.; Azuma, K.; Tomita, K.; Komiyama, S.; Weinstein, I.B. Constitutive activation of signal transducers and activators of transcription 3 correlates with cyclin D1 overexpression and may provide a novel prognostic marker in head and neck squamous cell carcinoma. Cancer Res. 2002, 62, 3351–3355. [Google Scholar] [PubMed]
  52. Marzec, M.; Zhang, Q.; Goradia, A.; Raghunath, P.N.; Liu, X.; Paessler, M.; Wang, H.Y.; Wysocka, M.; Cheng, M.; Ruggeri, B.A.; et al. Oncogenic kinase NPM/ALK induces through STAT3 expression of immunosuppressive protein CD274 (PD-L1, B7-H1). Proc. Natl. Acad. Sci. USA 2008, 105, 20852–20857. [Google Scholar] [CrossRef] [Green Version]
  53. Garama, D.J.; Harris, T.J.; White, C.L.; Rossello, F.J.; Abdul-Hay, M.; Gough, D.J.; Levy, D.E. A Synthetic Lethal Interaction between Glutathione Synthesis and Mitochondrial Reactive Oxygen Species Provides a Tumor-Specific Vulnerability Dependent on STAT3. Mol. Cell. Biol. 2015, 35, 3646–3656. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  54. He, B.; You, L.; Uematsu, K.; Zang, K.; Xu, Z.; Lee, A.Y.; Costello, J.F.; McCormick, F.; Jablons, D.M. SOCS-3 is frequently silenced by hypermethylation and suppresses cell growth in human lung cancer. Proc. Natl. Acad. Sci. USA 2003, 100, 14133–14138. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  55. Ogata, Y.; Osaki, T.; Naka, T.; Iwahori, K.; Furukawa, M.; Nagatomo, I.; Kijima, T.; Kumagai, T.; Yoshida, M.; Tachibana, I.; et al. Overexpression of PIAS3 suppresses cell growth and restores the drug sensitivity of human lung cancer cells in association with PI3-K/Akt inactivation. Neoplasia 2006, 8, 817–825. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  56. Li, Y.; Du, H.; Qin, Y.; Roberts, J.; Cummings, O.W.; Yan, C. Activation of the signal transducers and activators of the transcription 3 pathway in alveolar epithelial cells induces inflammation and adenocarcinomas in mouse lung. Cancer Res. 2007, 67, 8494–8503. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  57. Ihara, S.; Kida, H.; Arase, H.; Tripathi, L.P.; Chen, Y.A.; Kimura, T.; Yoshida, M.; Kashiwa, Y.; Hirata, H.; Fukamizu, R.; et al. Inhibitory roles of signal transducer and activator of transcription 3 in antitumor immunity during carcinogen-induced lung tumorigenesis. Cancer Res. 2012, 72, 2990–2999. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  58. Grabner, B.; Schramek, D.; Mueller, K.M.; Moll, H.P.; Svinka, J.; Hoffmann, T.; Bauer, E.; Blaas, L.; Hruschka, N.; Zboray, K.; et al. Disruption of STAT3 signalling promotes KRAS-induced lung tumorigenesis. Nat. Commun. 2015, 6, 6285. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  59. Zhou, J.; Qu, Z.; Yan, S.; Sun, F.; Whitsett, J.A.; Shapiro, S.D.; Xiao, G. Differential roles of STAT3 in the initiation and growth of lung cancer. Oncogene 2015, 34, 3804–3814. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  60. Caetano, M.S.; Hassane, M.; Van, H.T.; Bugarin, E.; Cumpian, A.M.; McDowell, C.L.; Cavazos, C.G.; Zhang, H.; Deng, S.; Diao, L. Sex specific function of epithelial STAT3 signaling in pathogenesis of K-ras mutant lung cancer. Nat. Commun. 2018, 9, 4589. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  61. Deng, S.; Ramos-Castaneda, M.; Velasco, W.V.; Clowers, M.J.; Gutierrez, B.A.; Noble, O.; Dong, Y.; Zarghooni, M.; Alvarado, L.; Caetano, M.S.; et al. Interplay between Estrogen and Stat3/NF-κB Driven Immunomodulation in Lung Cancer. Carcinogenesis 2020, 41, 1529–1542. [Google Scholar] [CrossRef] [PubMed]
  62. Thomas, L.; Doyle, L.A.; Edelman, M.J. Lung cancer in women: Emerging differences in epidemiology, biology, and therapy. Chest 2005, 128, 370–381. [Google Scholar] [CrossRef] [PubMed]
  63. Dutta, P.; Zhang, L.; Zhang, H.; Peng, Q.; Montgrain, P.R.; Wang, Y.; Song, Y.; Li, J.; Li, W.X. Unphosphorylated STAT3 in heterochromatin formation and tumor suppression in lung cancer. BMC Cancer 2020, 20, 145. [Google Scholar] [CrossRef] [PubMed]
  64. Augsten, M. Cancer-associated fibroblasts as another polarized cell type of the tumor microenvironment. Front. Oncol. 2014, 4, 62. [Google Scholar] [CrossRef]
  65. Wu, F.; Yang, J.; Liu, J.; Wang, Y.; Mu, J.; Zeng, Q.; Deng, S.; Zhou, H. Signaling pathways in cancer-associated fibroblasts and targeted therapy for cancer. Signal Transduct. Target. Ther. 2021, 6, 218. [Google Scholar] [CrossRef]
  66. Wang, L.; Cao, L.; Wang, H.; Liu, B.; Zhang, Q.; Meng, Z.; Wu, X.; Zhou, Q.; Xu, K. Cancer-associated fibroblasts enhance metastatic potential of lung cancer cells through IL-6/STAT3 signaling pathway. Oncotarget 2017, 8, 76116–76128. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  67. Tao, L.; Huang, G.; Wang, R.; Pan, Y.; He, Z.; Chu, X.; Song, H.; Chen, L. Cancer-associated fibroblasts treated with cisplatin facilitates chemoresistance of lung adenocarcinoma through IL-11/IL-11R/STAT3 signaling pathway. Sci. Rep. 2016, 6, 38408. [Google Scholar] [CrossRef] [PubMed]
  68. Ardain, A.; Marakalala, M.J.; Leslie, A. Tissue-resident innate immunity in the lung. Immunology 2020, 159, 245–256. [Google Scholar] [CrossRef] [Green Version]
  69. Yunna, C.; Mengru, H.; Lei, W.; Weidong, C. Macrophage M1/M2 polarization. Eur. J. Pharmacol. 2020, 877, 173090. [Google Scholar] [CrossRef] [PubMed]
  70. Conway, E.M.; Pikor, L.A.; Kung, S.H.; Hamilton, M.J.; Lam, S.; Lam, W.L.; Bennewith, K.L. Macrophages, Inflammation, and Lung Cancer. Am. J. Respir. Crit. Care Med. 2016, 193, 116–130. [Google Scholar] [CrossRef] [PubMed]
  71. Rébé, C.; Végran, F.; Berger, H.; Ghiringhelli, F. STAT3 activation: A key factor in tumor immunoescape. JAKSTAT 2013, 2, e23010. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  72. Kortylewski, M.; Kujawski, M.; Herrmann, A.; Yang, C.; Wang, L.; Liu, Y.; Salcedo, R.; Yu, H. Toll-like receptor 9 activation of signal transducer and activator of transcription 3 constrains its agonist-based immunotherapy. Cancer Res. 2009, 69, 2497–2505. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  73. Zhou, J.; Qu, Z.; Sun, F.; Han, L.; Li, L.; Yan, S.; Stabile, L.P.; Chen, L.F.; Siegfried, J.M.; Xiao, G. Myeloid STAT3 Promotes Lung Tumorigenesis by Transforming Tumor Immunosurveillance into Tumor-Promoting Inflammation. Cancer Immunol. Res. 2017, 5, 257–268. [Google Scholar] [CrossRef] [Green Version]
  74. Hiwatashi, K.; Tamiya, T.; Hasegawa, E.; Fukaya, T.; Hashimoto, M.; Kakoi, K.; Kashiwagi, I.; Kimura, A.; Inoue, N.; Morita, R.; et al. Suppression of SOCS3 in macrophages prevents cancer metastasis by modifying macrophage phase and MCP2/CCL8 induction. Cancer Lett. 2011, 308, 172–180. [Google Scholar] [CrossRef]
  75. Gong, L.; Cumpian, A.M.; Caetano, M.S.; Ochoa, C.E.; De la Garza, M.M.; Lapid, D.J.; Mirabolfathinejad, S.G.; Dickey, B.F.; Zhou, Q.; Moghaddam, S.J. Promoting effect of neutrophils on lung tumorigenesis is mediated by CXCR2 and neutrophil elastase. Mol. Cancer 2013, 12, 154. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  76. Kargl, J.; Busch, S.E.; Yang, G.H.; Kim, K.H.; Hanke, M.L.; Metz, H.E.; Hubbard, J.J.; Lee, S.M.; Madtes, D.K.; McIntosh, M.W.; et al. Neutrophils dominate the immune cell composition in non-small cell lung cancer. Nat. Commun. 2017, 8, 14381. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  77. Shaul, M.E.; Fridlender, Z.G. Neutrophils as active regulators of the immune system in the tumor microenvironment. J. Leukoc. Biol. 2017, 102, 343–349. [Google Scholar] [CrossRef] [Green Version]
  78. Nozawa, H.; Chiu, C.; Hanahan, D. Infiltrating neutrophils mediate the initial angiogenic switch in a mouse model of multistage carcinogenesis. Proc. Natl. Acad. Sci. USA 2006, 103, 12493–12498. [Google Scholar] [CrossRef] [Green Version]
  79. Acuff, H.B.; Carter, K.J.; Fingleton, B.; Gorden, D.L.; Matrisian, L.M. Matrix metalloproteinase-9 from bone marrow-derived cells contributes to survival but not growth of tumor cells in the lung microenvironment. Cancer Res. 2006, 66, 259–266. [Google Scholar] [CrossRef] [Green Version]
  80. Peranzoni, E.; Zilio, S.; Marigo, I.; Dolcetti, L.; Zanovello, P.; Mandruzzato, S.; Bronte, V. Myeloid-derived suppressor cell heterogeneity and subset definition. Curr. Opin. Immunol. 2010, 22, 238–244. [Google Scholar] [CrossRef] [PubMed]
  81. Welch, D.R.; Schissel, D.J.; Howrey, R.P.; Aeed, P.A. Tumor-elicited polymorphonuclear cells, in contrast to “normal” circulating polymorphonuclear cells, stimulate invasive and metastatic potentials of rat mammary adenocarcinoma cells. Proc. Natl. Acad. Sci. USA 1989, 86, 5859–5863. [Google Scholar] [CrossRef] [Green Version]
  82. Houghton, A.M.; Rzymkiewicz, D.M.; Ji, H.; Gregory, A.D.; Egea, E.E.; Metz, H.E.; Stolz, D.B.; Land, S.R.; Marconcini, L.A.; Kliment, C.R.; et al. Neutrophil elastase-mediated degradation of IRS-1 accelerates lung tumor growth. Nat. Med. 2010, 16, 219–223. [Google Scholar] [CrossRef] [Green Version]
  83. Gabrilovich, D.I. The Dawn of Myeloid-Derived Suppressor Cells: Identification of Arginase I as the Mechanism of Immune Suppression. Cancer Res. 2021, 81, 3953–3955. [Google Scholar] [CrossRef] [PubMed]
  84. Karin, N. The Development and Homing of Myeloid-Derived Suppressor Cells: From a Two-Stage Model to a Multistep Narrative. Front. Immunol. 2020, 11, 557586. [Google Scholar] [CrossRef]
  85. Nefedova, Y.; Nagaraj, S.; Rosenbauer, A.; Muro-Cacho, C.; Sebti, S.M.; Gabrilovich, D.I. Regulation of dendritic cell differentiation and antitumor immune response in cancer by pharmacologic-selective inhibition of the janus-activated kinase 2/signal transducers and activators of transcription 3 pathway. Cancer Res. 2005, 65, 9525–9535. [Google Scholar] [CrossRef] [Green Version]
  86. Gabrilovich, D.I. Myeloid-Derived Suppressor Cells. Cancer Immunol. Res. 2017, 5, 3–8. [Google Scholar] [CrossRef] [Green Version]
  87. 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]
  88. Gabrilovich, D.I.; Nagaraj, S. Myeloid-derived suppressor cells as regulators of the immune system. Nat. Rev. Immunol. 2009, 9, 162–174. [Google Scholar] [CrossRef]
  89. Kusmartsev, S.; Nefedova, Y.; Yoder, D.; Gabrilovich, D.I. Antigen-specific inhibition of CD8+ T cell response by immature myeloid cells in cancer is mediated by reactive oxygen species. J. Immunol. 2004, 172, 989–999. [Google Scholar] [CrossRef] [Green Version]
  90. Wölfle, S.J.; Strebovsky, J.; Bartz, H.; Sähr, A.; Arnold, C.; Kaiser, C.; Dalpke, A.H.; Heeg, K. PD-L1 expression on tolerogenic APCs is controlled by STAT-3. Eur. J. Immunol. 2011, 41, 413–424. [Google Scholar] [CrossRef]
  91. Jorgovanovic, D.; Song, M.; Wang, L.; Zhang, Y. Roles of IFN-γ in tumor progression and regression: A review. Biomark. Res. 2020, 8, 49. [Google Scholar] [CrossRef] [PubMed]
  92. Wajant, H.; Pfizenmaier, K.; Scheurich, P. Tumor necrosis factor signaling. Cell Death Differ. 2003, 10, 45–65. [Google Scholar] [CrossRef] [Green Version]
  93. Golubovskaya, V.; Wu, L. Different Subsets of T Cells, Memory, Effector Functions, and CAR-T Immunotherapy. Cancers 2016, 8, 36. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  94. Capone, A.; Volpe, E. Transcriptional Regulators of T Helper 17 Cell Differentiation in Health and Autoimmune Diseases. Front. Immunol. 2020, 11, 348. [Google Scholar] [CrossRef]
  95. Harris, T.J.; Grosso, J.F.; Yen, H.R.; Xin, H.; Kortylewski, M.; Albesiano, E.; Hipkiss, E.L.; Getnet, D.; Goldberg, M.V.; Maris, C.H.; et al. Cutting edge: An in vivo requirement for STAT3 signaling in TH17 development and TH17-dependent autoimmunity. J. Immunol. 2007, 179, 4313–4317. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  96. Zhou, L.; Ivanov, I.I.; Spolski, R.; Min, R.; Shenderov, K.; Egawa, T.; Levy, D.E.; Leonard, W.J.; Littman, D.R. IL-6 programs T(H)-17 cell differentiation by promoting sequential engagement of the IL-21 and IL-23 pathways. Nat. Immunol. 2007, 8, 967–974. [Google Scholar] [CrossRef]
  97. Yao, Z.; Painter, S.L.; Fanslow, W.C.; Ulrich, D.; Macduff, B.M.; Spriggs, M.K.; Armitage, R.J. Human IL-17: A novel cytokine derived from T cells. J. Immunol. 1995, 155, 5483–5486. [Google Scholar] [PubMed]
  98. Tobin, R.P.; Jordan, K.R.; Kapoor, P.; Spongberg, E.; Davis, D.; Vorwald, V.M.; Couts, K.L.; Gao, D.; Smith, D.E.; Borgers, J.S.W.; et al. IL-6 and IL-8 Are Linked With Myeloid-Derived Suppressor Cell Accumulation and Correlate With Poor Clinical Outcomes in Melanoma Patients. Front. Oncol. 2019, 9, 1223. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  99. Akbay, E.A.; Koyama, S.; Liu, Y.; Dries, R.; Bufe, L.E.; Silkes, M.; Alam, M.M.; Magee, D.M.; Jones, R.; Jinushi, M.; et al. Interleukin-17A Promotes Lung Tumor Progression through Neutrophil Attraction to Tumor Sites and Mediating Resistance to PD-1 Blockade. J. Thorac. Oncol. 2017, 12, 1268–1279. [Google Scholar] [CrossRef] [Green Version]
  100. Gorczynski, R.M. IL-17 Signaling in the Tumor Microenvironment. Adv. Exp. Med. Biol. 2020, 1240, 47–58. [Google Scholar] [CrossRef]
  101. Deng, S.; Clowers, M.J.; Velasco, W.V.; Ramos-Castaneda, M.; Moghaddam, S.J. Understanding the Complexity of the Tumor Microenvironment in K-ras Mutant Lung Cancer: Finding an Alternative Path to Prevention and Treatment. Front. Oncol. 2019, 9, 1556. [Google Scholar] [CrossRef]
  102. Chang, S.H.; Mirabolfathinejad, S.G.; Katta, H.; Cumpian, A.M.; Gong, L.; Caetano, M.S.; Moghaddam, S.J.; Dong, C. T helper 17 cells play a critical pathogenic role in lung cancer. Proc. Natl. Acad. Sci. USA 2014, 111, 5664–5669. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  103. Chalmin, F.; Mignot, G.; Bruchard, M.; Chevriaux, A.; Végran, F.; Hichami, A.; Ladoire, S.; Derangère, V.; Vincent, J.; Masson, D.; et al. Stat3 and Gfi-1 transcription factors control Th17 cell immunosuppressive activity via the regulation of ectonucleotidase expression. Immunity 2012, 36, 362–373. [Google Scholar] [CrossRef] [Green Version]
  104. Yanagisawa, H.; Hashimoto, M.; Minagawa, S.; Takasaka, N.; Ma, R.; Moermans, C.; Ito, S.; Araya, J.; Budelsky, A.; Goodsell, A.; et al. Role of IL-17A in murine models of COPD airway disease. Am. J. Physiol. Lung Cell Mol. Physiol. 2017, 312, L122–L130. [Google Scholar] [CrossRef] [PubMed]
  105. Carrega, P.; Loiacono, F.; Di Carlo, E.; Scaramuccia, A.; Mora, M.; Conte, R.; Benelli, R.; Spaggiari, G.M.; Cantoni, C.; Campana, S.; et al. NCR+ILC3 concentrate in human lung cancer and associate with intratumoral lymphoid structures. Nat. Commun. 2015, 6, 8280. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  106. Asadzadeh, Z.; Mohammadi, H.; Safarzadeh, E.; Hemmatzadeh, M.; Mahdian-Shakib, A.; Jadidi-Niaragh, F.; Azizi, G.; Baradaran, B. The paradox of Th17 cell functions in tumor immunity. Cell. Immunol. 2017, 322, 15–25. [Google Scholar] [CrossRef] [PubMed]
  107. Sonnenberg, G.F.; Fouser, L.A.; Artis, D. Border patrol: Regulation of immunity, inflammation and tissue homeostasis at barrier surfaces by IL-22. Nat. Immunol. 2011, 12, 383–390. [Google Scholar] [CrossRef]
  108. Tufman, A.; Huber, R.M.; Völk, S.; Aigner, F.; Edelmann, M.; Gamarra, F.; Kiefl, R.; Kahnert, K.; Tian, F.; Boulesteix, A.L.; et al. Interleukin-22 is elevated in lavage from patients with lung cancer and other pulmonary diseases. BMC Cancer 2016, 16, 409. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  109. Khosravi, N.; Caetano, M.S.; Cumpian, A.M.; Unver, N.; De la Garza Ramos, C.; Noble, O.; Daliri, S.; Hernandez, B.J.; Gutierrez, B.A.; Evans, S.E.; et al. IL22 Promotes Kras Mutant Lung Cancer by Induction of a Pro-Tumor Immune Response and Protection of Stemness Properties. Cancer Immunol. Res. 2018, 6, 788–797. [Google Scholar] [CrossRef] [Green Version]
  110. Zheng, S.G.; Wang, J.; Wang, P.; Gray, J.D.; Horwitz, D.A. IL-2 is essential for TGF-beta to convert naive CD4+CD25 cells to CD25+Foxp3+ regulatory T cells and for expansion of these cells. J. Immunol. 2007, 178, 2018–2027. [Google Scholar] [CrossRef] [Green Version]
  111. Arce-Sillas, A.; Álvarez-Luquín, D.D.; Tamaya-Domínguez, B.; Gomez-Fuentes, S.; Trejo-García, A.; Melo-Salas, M.; Cárdenas, G.; Rodríguez-Ramírez, J.; Adalid-Peralta, L. Regulatory T Cells: Molecular Actions on Effector Cells in Immune Regulation. J. Immunol. Res. 2016, 2016, 1720827. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  112. Shang, B.; Liu, Y.; Jiang, S.J. Prognostic value of tumor-infiltrating FoxP3+ regulatory T cells in cancers: A systematic review and meta-analysis. Sci. Rep. 2015, 5, 15179. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  113. Pallandre, J.R.; Brillard, E.; Créhange, G.; Radlovic, A.; Remy-Martin, J.P.; Saas, P.; Rohrlich, P.S.; Pivot, X.; Ling, X.; Tiberghien, P.; et al. Role of STAT3 in CD4+CD25+FOXP3+ regulatory lymphocyte generation: Implications in graft-versus-host disease and antitumor immunity. J. Immunol. 2007, 179, 7593–7604. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  114. Crowley, M.; Inaba, K.; Steinman, R.M. Dendritic cells are the principal cells in mouse spleen bearing immunogenic fragments of foreign proteins. J. Exp. Med. 1990, 172, 383–386. [Google Scholar] [CrossRef] [Green Version]
  115. Wculek, S.K.; Cueto, F.J.; Mujal, A.M.; Melero, I.; Krummel, M.F.; Sancho, D. Dendritic cells in cancer immunology and immunotherapy. Nat. Rev. Immunol. 2020, 20, 7–24. [Google Scholar] [CrossRef]
  116. Laouar, Y.; Welte, T.; Fu, X.Y.; Flavell, R.A. STAT3 is required for Flt3L-dependent dendritic cell differentiation. Immunity 2003, 19, 903–912. [Google Scholar] [CrossRef] [Green Version]
  117. Park, S.J.; Nakagawa, T.; Kitamura, H.; Atsumi, T.; Kamon, H.; Sawa, S.; Kamimura, D.; Ueda, N.; Iwakura, Y.; Ishihara, K.; et al. IL-6 regulates in vivo dendritic cell differentiation through STAT3 activation. J. Immunol. 2004, 173, 3844–3854. [Google Scholar] [CrossRef] [Green Version]
  118. Iwata-Kajihara, T.; Sumimoto, H.; Kawamura, N.; Ueda, R.; Takahashi, T.; Mizuguchi, H.; Miyagishi, M.; Takeda, K.; Kawakami, Y. Enhanced cancer immunotherapy using STAT3-depleted dendritic cells with high Th1-inducing ability and resistance to cancer cell-derived inhibitory factors. J. Immunol. 2011, 187, 27–36. [Google Scholar] [CrossRef] [PubMed]
  119. Sun, Y.; Chin, Y.E.; Weisiger, E.; Malter, C.; Tawara, I.; Toubai, T.; Gatza, E.; Mascagni, P.; Dinarello, C.A.; Reddy, P. Cutting edge: Negative regulation of dendritic cells through acetylation of the nonhistone protein STAT-3. J. Immunol. 2009, 182, 5899–5903. [Google Scholar] [CrossRef] [PubMed]
  120. Marcus, A.; Gowen, B.G.; Thompson, T.W.; Iannello, A.; Ardolino, M.; Deng, W.; Wang, L.; Shifrin, N.; Raulet, D.H. Recognition of tumors by the innate immune system and natural killer cells. Adv. Immunol. 2014, 122, 91–128. [Google Scholar] [CrossRef] [Green Version]
  121. Morvan, M.G.; Lanier, L.L. NK cells and cancer: You can teach innate cells new tricks. Nat. Rev. Cancer 2016, 16, 7–19. [Google Scholar] [CrossRef]
  122. Gotthardt, D.; Putz, E.M.; Straka, E.; Kudweis, P.; Biaggio, M.; Poli, V.; Strobl, B.; Müller, M.; Sexl, V. Loss of STAT3 in murine NK cells enhances NK cell-dependent tumor surveillance. Blood 2014, 124, 2370–2379. [Google Scholar] [CrossRef]
  123. Zhou, X.; Liu, S.; Liu, J.; Zhang, Z.; Mao, X.; Zhou, H. MicroRNA-130a enhances the killing ability of natural killer cells against non-small cell lung cancer cells by targeting signal transducers and activators of transcription 3. Biochem. Biophys. Res. Commun. 2020, 523, 481–486. [Google Scholar] [CrossRef] [PubMed]
  124. Wang, R.X.; Yu, C.R.; Dambuza, I.M.; Mahdi, R.M.; Dolinska, M.B.; Sergeev, Y.V.; Wingfield, P.T.; Kim, S.H.; Egwuagu, C.E. Interleukin-35 induces regulatory B cells that suppress autoimmune disease. Nat. Med. 2014, 20, 633–641. [Google Scholar] [CrossRef]
  125. Oladipupo, F.O.; Yu, C.R.; Olumuyide, E.; Jittaysothorn, Y.; Choi, J.K.; Egwuagu, C.E. STAT3 deficiency in B cells exacerbates uveitis by promoting expansion of pathogenic lymphocytes and suppressing regulatory B cells (Bregs) and Tregs. Sci. Rep. 2020, 10, 16188. [Google Scholar] [CrossRef] [PubMed]
  126. Zhou, J.; Min, Z.; Zhang, D.; Wang, W.; Marincola, F.; Wang, X. Enhanced frequency and potential mechanism of B regulatory cells in patients with lung cancer. J. Transl. Med. 2014, 12, 304. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  127. Zhang, C.; Xin, H.; Zhang, W.; Yazaki, P.J.; Zhang, Z.; Le, K.; Li, W.; Lee, H.; Kwak, L.; Forman, S.; et al. CD5 Binds to Interleukin-6 and Induces a Feed-Forward Loop with the Transcription Factor STAT3 in B Cells to Promote Cancer. Immunity 2016, 44, 913–923. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  128. Ding, C.; Chen, X.; Dascani, P.; Hu, X.; Bolli, R.; Zhang, H.G.; Mcleish, K.R.; Yan, J. STAT3 Signaling in B Cells Is Critical for Germinal Center Maintenance and Contributes to the Pathogenesis of Murine Models of Lupus. J. Immunol. 2016, 196, 4477–4486. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  129. Wong, A.L.A.; Hirpara, J.L.; Pervaiz, S.; Eu, J.Q.; Sethi, G.; Goh, B.C. Do STAT3 inhibitors have potential in the future for cancer therapy? Expert Opin. Investig. Drugs 2017, 26, 883–887. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  130. Parakh, S.; Ernst, M.; Poh, A.R. Multicellular Effects of STAT3 in Non-small Cell Lung Cancer: Mechanistic Insights and Therapeutic Opportunities. Cancers 2021, 13, 6228. [Google Scholar] [CrossRef] [PubMed]
  131. Yang, L.; Lin, S.; Xu, L.; Lin, J.; Zhao, C.; Huang, X. Novel activators and small-molecule inhibitors of STAT3 in cancer. Cytokine Growth Factor Rev. 2019, 49, 10–22. [Google Scholar] [CrossRef] [PubMed]
  132. Xu, X.; Kasembeli, M.M.; Jiang, X.; Tweardy, B.J.; Tweardy, D.J. Chemical probes that competitively and selectively inhibit Stat3 activation. PLoS ONE 2009, 4, e4783. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  133. Lewis, K.M.; Bharadwaj, U.; Eckols, T.K.; Kolosov, M.; Kasembeli, M.M.; Fridley, C.; Siller, R.; Tweardy, D.J. Small-molecule targeting of signal transducer and activator of transcription (STAT) 3 to treat non-small cell lung cancer. Lung Cancer 2015, 90, 182–190. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  134. Hong, D.; Kurzrock, R.; Kim, Y.; Woessner, R.; Younes, A.; Nemunaitis, J.; Fowler, N.; Zhou, T.; Schmidt, J.; Jo, M.; et al. AZD9150, a next-generation antisense oligonucleotide inhibitor of STAT3 with early evidence of clinical activity in lymphoma and lung cancer. Sci. Transl. Med. 2015, 7, 314ra185. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  135. Bayliss, T.J.; Smith, J.T.; Schuster, M.; Dragnev, K.H.; Rigas, J.R. A humanized anti-IL-6 antibody (ALD518) in non-small cell lung cancer. Expert Opin. Biol. Ther. 2011, 11, 1663–1668. [Google Scholar] [CrossRef] [PubMed]
Figure 1. An overview of STAT3 signaling. Figure created in BioRender.
Figure 1. An overview of STAT3 signaling. Figure created in BioRender.
Cancers 14 01785 g001
Figure 2. Cell type-specific effects of hyperactive and attenuated STAT3 signaling, with anti-tumor effects in green and pro-tumor effects in red. Figure created in BioRender.
Figure 2. Cell type-specific effects of hyperactive and attenuated STAT3 signaling, with anti-tumor effects in green and pro-tumor effects in red. Figure created in BioRender.
Cancers 14 01785 g002
Table 1. Mouse models available to study STAT3 in KM-LUAD.
Table 1. Mouse models available to study STAT3 in KM-LUAD.
ApproachesExamplesInduction Method
Tissue-Specific Cre RecombinaseCCSPCre: lung epithelium
SPCCre: whole lung
rtTA-tetO: dox-inducible
Germline/AdCre
LysMCre: myeloid
CD4Cre: helper T cell
CD19Cre: B cell
NCR1Cre: NK cell
Germline/Adoptive Transfer
MutationsKrasLA2/+
KrasLSL-G12D/+
KrasLSL-G12V/+
±p53lox/lox
Germline/AdCre
COPD-like InflammationNTHi
LPS
Nebulization
Intranasal
Cigarette Smoke
Elastase
Smoke exposure
Intratracheal
CarcinogenNNK
Urethane
Intraperitoneal
Cigarette Smoke
eCigarette
Smoke exposure
Atomized nicotine
XenograftImmunocompromised mouseOrthotopic, subcutaneous, or intravenous
AdCre: Adenoviral delivery of Cre and any other transgenes.
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Clowers, M.J.; Moghaddam, S.J. Cell Type-Specific Roles of STAT3 Signaling in the Pathogenesis and Progression of K-ras Mutant Lung Adenocarcinoma. Cancers 2022, 14, 1785. https://doi.org/10.3390/cancers14071785

AMA Style

Clowers MJ, Moghaddam SJ. Cell Type-Specific Roles of STAT3 Signaling in the Pathogenesis and Progression of K-ras Mutant Lung Adenocarcinoma. Cancers. 2022; 14(7):1785. https://doi.org/10.3390/cancers14071785

Chicago/Turabian Style

Clowers, Michael J., and Seyed Javad Moghaddam. 2022. "Cell Type-Specific Roles of STAT3 Signaling in the Pathogenesis and Progression of K-ras Mutant Lung Adenocarcinoma" Cancers 14, no. 7: 1785. https://doi.org/10.3390/cancers14071785

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