Next Article in Journal
Clomiphene Citrate Shows Effective and Sustained Antimicrobial Activity against Mycobacterium abscessus
Next Article in Special Issue
Autoantibodies from Patients with Scleroderma Renal Crisis Promote PAR-1 Receptor Activation and IL-6 Production in Endothelial Cells
Previous Article in Journal
A Presurgical Study of Curcumin Combined with Anthocyanin Supplements in Patients with Colorectal Adenomatous Polyps
Previous Article in Special Issue
Interleukin-1β in Multifactorial Hypertension: Inflammation, Vascular Smooth Muscle Cell and Extracellular Matrix Remodeling, and Non-Coding RNA Regulation
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

IL-6 in the Ecosystem of Head and Neck Cancer: Possible Therapeutic Perspectives

by
Michal Španko
1,2,*,
Karolína Strnadová
1,3,
Aleš Jan Pavlíček
1,4,
Pavol Szabo
1,
Ondřej Kodet
1,3,4,
Jaroslav Valach
2,
Barbora Dvořánková
1,3,
Karel Smetana, Jr.
1,3 and
Lukáš Lacina
1,3,4
1
Institute of Anatomy, First Faculty of Medicine, Charles University, 12800 Prague, Czech Republic
2
Department of Stomatology, First Faculty of Medicine, Charles University and General University Hospital in Prague, 12801 Prague, Czech Republic
3
BIOCEV, Biotechnology and Biomedicine Centre, Academy of Science and Charles University, 25250 Vestec, Czech Republic
4
Department of Dermatovenereology, First Faculty of Medicine, Charles University and General University Hospital in Prague, 12800 Prague, Czech Republic
*
Author to whom correspondence should be addressed.
Int. J. Mol. Sci. 2021, 22(20), 11027; https://doi.org/10.3390/ijms222011027
Submission received: 23 August 2021 / Revised: 25 September 2021 / Accepted: 29 September 2021 / Published: 13 October 2021

Abstract

:
Interleukin-6 (IL-6) is a highly potent cytokine involved in multiple biological processes. It was previously reported to play a distinct role in inflammation, autoimmune and psychiatric disorders, ageing and various types of cancer. Furthermore, it is understood that IL-6 and its signaling pathways are substantial players in orchestrating the cancer microenvironment. Thus, they appear to be potential targets in anti-tumor therapy. The aim of this article is to elucidate the role of IL-6 in the tumor ecosystem and to review the possible therapeutic approaches in head and neck cancer.

1. Introduction

Head and neck cancer (HNC) is a significant cause of morbidity and mortality worldwide. Its incidence is increasing; approximately 550,000 new cases are diagnosed every year [1]. Head and neck squamous cell carcinoma (HNSCC) is its most frequent histological type [2]. Smoking, alcohol consumption, paan (betel) abuse in some regions and HPV infection are pivotal etiological causes of HNSCC [3]. Furthermore, ageing is substantially involved, as gene-repair mechanisms are reduced after the age of 50. The risk of malignancy is thus further enhanced [4]. In HNSCC, it is alarming that approximately two-thirds of cases are diagnosed at an advanced stage, and roughly 50% of the patients die within two years after the initial diagnosis [5]. Since the number of oncological cases is rising, novel therapeutic approaches need to be developed [6].
The tumor mass is a complex ecosystem. It is formed of cancer cells and a supporting tissue called tumor stroma. It comprises various cell populations (e.g., activated fibroblasts, immune cells, endothelial cells, adipocytes, mesenchymal stem cells) and extracellular matrix [7,8,9,10]. The biological behavior of cancer cells can be significantly modified by stromal cells through intensive intercellular cross-talk. This can be based on their mutual intercellular contacts or paracrine production of bioactive factors [11,12,13].
Cancer-associated fibroblasts (CAFs) produce a plethora of cytokines. Their effect is pleiotropic, i.e., they can modulate functions of various cell types. The function can be either synergistic or antagonistic. Their interplay is a highly complex phenomenon. Chemokines and interleukins are the major cytokines involved in tumor growth and metastatic spread [3].
Interleukin-6 (IL-6) is a crucial component of the cancer microenvironment. It can be produced by cancer cells or their stromal counterparts, e.g., CAFs [10,14,15]. The synthesis and activity of IL-6, though, is not limited to the cells of the cancer ecosystem. It can easily leak out and affect distant tissues and cells. It can also be found in, e.g., hematopoietic cells of bone marrow, epithelial cells, connective tissues and muscle cells [16]. This contributes to the formation of a premetastatic niche. Therefore, IL-6 signaling seems to be a promising target in anti-tumor therapy [6].
Of note, IL-6 is also involved in a plethora of other biological processes such as inflammation, ageing, autoimmune (rheumatoid arthritis) and psychiatric disorders (bipolar disorder and anorexia) [6]. However, it is beyond the scope of this review.
IL-6 is vitally important for many malignant cells; moreover, it is a critical molecule significantly shaping their tumor microenvironment landscape. We believe that this complex mechanism of multifaceted intercellular communication in tumors is worthy of attention. As the IL-6 signaling pathway offers several druggable targets, it was earlier clinically exploited to treat various conditions. We believe that it also holds therapeutic potential for the oncology of solid tumors, including HNC. In this review, we, therefore, aimed to summarize available data on the IL-6 signaling pathway mechanisms and emphasize the IL-6 importance in the context of the tumor microenvironment. We also offer a review of selected therapeutic options available for the purposes of IL-6 pathway regulation.

2. IL-6 Signaling Pathway

IL-6 is a glycosylated protein formed of 184 amino acids (MW 23.7 kDa) [17]. It was originally described to stimulate B lymphocytes and hepatocytes [18]. It is synthesized by multiple cell types including cancer and stromal cells in many types of human malignancies [16,17].
There are two principal pathways of IL-6 signaling. The first, referred to as classic signaling, utilizes the membrane-bound receptor (IL-6R). It is expressed only on hepatocytes and certain types of leukocytes [19,20]. The signal is subsequently transduced by membrane-bound α-receptor glycoprotein 130 (gp130), which initiates a cascade of intracellular processes. The second pathway, named trans-signaling, is activated by the binding of IL-6 to the soluble form of IL-6R (sIL-6R). sIL-6R is produced by proteolytic cleavage (shedding) of IL-6R from the cell membrane by metalloproteases ADAM10 and ADAM17 [21,22]. Of note, human sIL-6R can also be a product of alternative splicing of IL-6R mRNA. However, this mechanism is of minor significance [23]. The trans-signaling pathway can activate any cells since gp130 is expressed ubiquitously. It primarily involves pro-inflammatory reactions, whilst the classic signaling is dedicated to anti-inflammatory and regenerative processes [6,21]. IL-6R also appears to be pro-cancerogenic [24]. The soluble form of gp130 (sgp130), which is most likely formed by alternative splicing, is known to inhibit the trans-signaling pathway naturally [21,25].
As mentioned above, gp130 is the inceptive step of intracellular signaling. There are three possible consequent pathways: JAK-STAT (Janus kinase and signal transducer and activator of transcription) pathway, MAPK (mitogen-activated protein kinase) cascade and PI3K (phosphatidylinositol-3-kinase) cascade [22,26]. The cross-talk between these signaling units still requires detailed elucidation [22].
Interestingly, the cytokine receptors lack intrinsic kinase activity [22]. Janus kinases (JAK) are associated with the cytoplasmic part of gp130. Once IL-6 is bound to IL-6R, JAK kinases are activated by autophosphorylation. Of note, there is a family of JAK kinases associated with IL-6 signaling: JAK1, JAK2 and Tyk2. JAK1 is, though, the essential type. The activation of JAKs causes phosphorylation of tyrosine motifs located on the cytoplasmic part of gp130. The four most distal phosphorylated motifs (Y767, Y814, Y905 and Y915) bind STAT1 and STAT3 monomers. They are consequently phosphorylated within their tyrosine sites (STAT1: Y701, STAT3: Y705), dimerized and translocated into the nucleus [22,26,27]. This traditional description, though, appears to be simplified. Recent studies have revealed that STATs can dimerize even without cytokine stimulation and tyrosine phosphorylation [28,29,30,31]. The pre-dimerization step requires the N-terminal region of the protein, whereas the cytokine-dependent dimerization requires tyrosine Y705 phosphorylation [32]. Even the translocation of STAT3 into the nucleus is not strictly based upon phosphorylation. It requires the N-terminal domain. In fact, regardless of cytokine stimulation, non-phosphorylated STATs can transfer from the cytoplasm to the nucleus and vice versa [32]. However, IL-6-dependent tyrosine phosphorylation increases the pool of STAT3 in the nucleus [33]. Such a finding corresponds to the original concept of JAK-STAT signaling.
The nuclear activity of STAT3 engages the CBP/p300 transcriptional adaptor. Its function is to regulate transcription and the course of the cell cycle by histone acetylation [22,34]. Moreover, it acetylates STAT3 on both N-terminal (lysines 49 and 87) [35] and C-terminal domains (lysine 685) [36]. Such a modification enhances the interaction between STAT3 and CBP. Thus, STAT3-dependent gene expression is potentiated [35,36,37]. On the other hand, STAT1 signaling is considered to be of minor significance [22].
Apart from JAK-STAT signaling, IL-6 can initiate the MAPK (mitogen-activated protein kinase) cascade and the PI3K (phosphatidylinositol-3-kinase) cascade. The cytoplasmic tyrosine-759 motif of gp130 is responsible for a balance between the STAT3 and MAPK cascades. Such an equilibrium is essential for the organism’s homeostasis [38,39] and its disturbance may lead to the development of inflammatory and proliferative diseases [40,41]. STAT3 regulates expression of cyclins D2, D3 and A, phosphatase cdc24A and cell cycle inhibitors p21 and p27 [39]; activated MAPK is a crucial mitogenic impulse [38]. Hyperactivity of both STAT3 and MAPK correlates with splenomegaly and thrombocytosis. In contrast, diminished activity of STAT3 and MAPK is associated with reduced acute-phase response and thrombocytopenia. Attenuated phosphorylation of the tyrosine-759 motif increases IL-6-dependent STAT3 activity [42,43] and reduces MAPK activity [44,45], which is related to gastric adenomas, autoimmune arthritis and increased Th1 cytokine expression. Conversely, phosphorylation of the tyrosine-759 motif reduces STAT3 activity and potentiates MAPK activity. This leads to reduced acute-phase gene expression and increased Th2 cytokine expression [46] (Figure 1).
STAT3 can be inactivated through binding of JAK feedback inhibitor SOCS3 to the phosphorylated tyrosine-759 motif in gp130. The activation of MAPK is based on the binding of the adapter protein and tyrosine phosphatase SHP2 to the same tyrosine-759 motif [38,44,45,47,48]. Both of these proteins utilize SH2 domains for binding to gp130 [49]. Additionally, phosphorylation of the tyrosine-759 motif of gp130 leads to recruitment of protein Gab1 [50]. Its function is to connect MAPK and PI3K with the IL-6 receptor complex [51].
IL-6 signaling can be suppressed in multiple ways. As previously mentioned, SOCS3 and SHP2 bind the phosphorylated 759-tyrosine motif and inhibit JAK-STAT signaling [47]. The STAT-independent signaling (MAPK, PI3K) is thus initiated. IL-6 signaling can also be regulated by the receptor complex internalization and its subsequent degradation [52,53]. This happens both constitutively and by IL-6R activation. Of note, cytokines such as IL-1α and TNFα also contribute to receptor internalization [54]. Eventually, activated STAT dimers can be negatively regulated by PIAS (protein inhibitor of activated STAT). PIAS3 is known to interfere with the phosphorylated tyrosine-705 domain of STAT3 [55].

3. IL-6 in the Context of Head and Neck Cancer

3.1. Tumor Microenvironment

Despite the recent advances in clinical medicine, head and neck carcinoma remains a significant cause of morbidity and mortality worldwide [1]. The limited efficacy of conventional anti-tumor therapy originates in the eradication of quickly dividing cancer cells [56,57]. In contrast, slowly proliferating progenitors and stem cells remain relatively spared and protected by a surrounding niche called tumor microenvironment (TME) [58]. In the search for new therapeutic approaches, IL-6 appears to be a promising target since it is a potent player within the TME. A number of adverse cancer-associated effects are related to IL-6 signaling [6].
The tumor microenvironment is a very complex structure. The cancer cells are surrounded by a variety of supporting cell types (e.g., cancer-associated fibroblasts, immune cells, endothelial cells, adipocytes, mesenchymal stem cells) and extracellular matrix [7,8,9,10]. All types of these cells produce IL-6 [59]. Notably, the stromal cells are not just quiet bystanders—they are substantially involved in the tumor growth regulation [6,60].
IL-6 signaling is complex and is dependent on the available signal (IL-6 cytokine), receptor equipment (IL-6R) and signal transducer gp130. Glycoprotein gp130 is present ubiquitously on all cell types. In contrast, cytokine production and receptor expression are more restricted. Interleukin can act directly on the cell in an autocrine manner. It can also influence the neighboring cells in the vicinity of the cytokine source in a paracrine manner. However, IL-6 can also enter the circulation and act at long distances, and eve exert systemic effects of endocrine nature.
Such versatile arrangement of cytokine production and release along with receptor mobility in the soluble form is a prerequisite for precise orchestration of the tumor cells and their supporting stromal population into one malignant machinery. One cell type can easily support and foster the other components in case of a lack of any component of the receptor complex. In this view, it is also possible that the membrane-bound IL-6/IL-6R complexes on transmitter cells activate the gp130 receptors on neighboring receiver cells [61]. Moreover, such versatility justifies the concept of cluster signaling [62], where some cells are able to trans-present IL-6 through the complex containing IL-6R bound on their membrane and thus interact with gp130 expressed on another cell in intimate proximity [63].
Having said all that, IL-6 seems to fit very well to the concept of the tumor microenvironment. Should the intention of targeting IL-6 become a successful therapeutic strategy in oncology one day, it is inevitable that the structure and rules of the tissue microenvironment must be observed.

3.2. Cancer-Associated Fibroblasts (CAFs)

Immune cells were the first recognized producers of IL-6. However, CAFs are also known to be a potent source of IL-6 in the context of the tumor microenvironment. We will focus on them in detail because they have been extensively studied in recent years [64]. CAFs represent the most abundant cell type in the tumor stroma and secrete many regulatory molecules—growth factors, cytokines and immune modulators [65]. They can be readily recruited from the local fibroblasts. Besides that, CAFs can also to some extent arise from bone marrow mesenchymal cells, pericytes, endothelial cells and smooth muscle cells [60]. Although CAFs represent a heterogeneous group of cells [66], conserved expression of α-smooth muscle actin (α-SMA), vimentin, tenascin C, fibroblast-specific protein-1, fibroblast-activating protein and neural-glial protein is observed [65,67]. CAFs play an important role in tumorigenesis, local invasiveness, tumor angiogenesis and chemoresistance. CAFs can induce epithelial-mesenchymal transition (EMT) and thus facilitate distant metastasizing [68]. It has been reported that fibroblasts isolated from squamous cell carcinoma and basal cell carcinoma significantly differ from normal fibroblasts, since their phenotype is shifted towards epithelial cells [69,70,71,72].
There are numerous studies describing remarkable similarities between a tumor and a healing wound [10,15,73,74]. Fibroblasts are substantially involved in the formation of both tumor stroma and granulation tissue [13,71,74,75]. Fibroblasts can transform into myofibroblasts and hence modulate wound contraction [76]. Myofibroblasts also influence the biological behavior of the tumor [75]. Wound healing is a cascade of precisely regulated events. It comprises blood clotting, inflammation, proliferation and maturation with consequent remodeling [58]. All these steps are also precisely orchestrated in terms of their timing. If temporal and spatial regulation fails, wound healing is suboptimal. Of note, the granulation tissue, product of the proliferative phase of normal healing, resembles the tumor stroma.
Fibroblasts embedded in these tissues readily produce not only structural molecules of ECM, but also multiple cytokines and growth factors, and thus potentiate angiogenesis and epithelial cell proliferation [77,78,79,80]. This can lead to rapid reepithelization of the wounded area in normal healing tissue. However, in cancer, identical stimuli will result in increased tumor mass. The pathways are shared; the result is given by the current tissue context. According to whole-genome transcriptome profiling, certain cytokines and ECM proteins have similar regulatory roles in both tumor growth and wound healing—namely, IL-6, IL-8, chemokine (C-X-C domain) ligand 1, galectin-1, and fibronectin [15]. Interestingly, basal and squamous cell carcinomas were referred to as caricatures of a healing skin wound [58].
The cancer field theory was initially coined on the background of oral cancer; however, it was later applied to models of many cancer types [81]. It was primarily proposed to describe the wide impact of cancerogenic genotoxic agents affecting epithelial cells and consequently triggering neoplastic growth in some of them. However, these harmful stimuli also inevitably impact the surrounding fibroblasts. Therefore, the precise diagnostic distinction of normal or abnormal cells should not be applied to cancerous cells only, but should also be extended to the broader landscape forming the tumor-supportive microenvironment [10,13]. These cells, including CAFs, are per se not genetically malignant, but their activity in the tissue is at least tumor-promoting [82]. Therefore, the control of their key signaling mechanisms could have therapeutic implications.

3.3. Effects of IL-6 Expression

IL-6 is understood to modulate the tumor microenvironment significantly. It likely keeps the low-grade differentiation of cells in squamous cell carcinoma [15] and promotes their epithelial-mesenchymal transition (EMT) [83]. IL-6 produced by different cell types in the cancer microenvironment stimulates migration of cancer stem cells, hence the metastatic potential increases [84,85]. Overexpression of IL-6 indicates a poor response to chemo- [86,87] and radiotherapy [86,88,89]. Eventually, an elevated level of IL-6 may contribute to bone metastasizing [90]. IL-6 also affects the metabolism of oncological patients by induction of cancer cachexia and afflicts the central nervous system, where it may induce anorexia and depression (Figure 2) [6].

3.4. Serum and Salivary Concentrations of IL-6 in HNSCC and Their Potential Role in Diagnostics

The concentration of IL-6 in the serum and other biological fluids is increased in HNSCC patients [91,92,93,94]. The physiological serum level of IL-6 is lower than 10 pg/mL [95]. Riedel et al. reported higher serum IL-6 concentrations in HNSCC patients compared to healthy controls [96]. In contrast, a different study by Andersson et al. did not prove a statistically significant difference between HNSCC patients and healthy individuals in terms of serum IL-6 concentrations [97]. It should be noted here that many HNSCC cases are primarily exophytic tumors. Therefore, an increased level of IL-6 in the serum might not be an optimal biomarker of early disease. However, this does not diminish its critical importance during the progression and metastasis formation.
Oral squamous cell carcinoma (OSCC), a topographic subtype of HNSCC, was also reported to have elevated serum IL-6 concentrations [94,98]. There is robust evidence that elevated IL-6 expression correlates with tumor progression and consequently also with lower patient overall survival [85,86,87,89,91,96,98,99,100,101,102,103,104,105,106]. In detail, increased IL-6 levels were associated with increased tumor size and the presence of nodal metastasis in OSCC patients [99]. Another study also reported higher T-stage, clinical stage, bone invasion and tumor depths in OSCC [98]. HNSCC patients with T3/T4-stage, positive nodal metastases and advanced stage were also found to have their IL-6 levels elevated [96,105,106].
We can conclude that IL-6 seems to be a candidate biomarker related to the overall- and disease-free survival in OSCC patients [98]. Its value could be further increased in combination with other biomarkers. Such multiplexing can further facilitate diagnostic interpretation. For instance, overexpression of both IL-6 and nuclear myoferlin indicates more dismal survival rates when compared with isolated myoferlin overexpression [107]. A similar result can be found when there is overexpression of both IL-6 and protein arginine methyltransferase-5 [108]. Also, analyzing the serum IL-6 concentration jointly with salivary IL-8 concentration can improve the sensitivity and specificity [109].
Of note, increased systemic availability of IL-6 can have rather unexpected implications. Human behavior is extremely complex, and it is challenging to apply principles of scientific reductionism strictly to it and follow the increase of a single cytokine. However, it is worth mentioning that circulating IL-6 has a clinically documented impact on several behavioral aspects. All of these behavioral patterns might have health-related consequences. Duffy et al. analyzed the possible relationship between health-related behavior and IL-6 levels in patients prior to HNSCC treatment. The results showed that particular addictive behavior (represented by smoking status), weak sleep scores, higher age, and more advanced tumor stage were significantly associated with increased serum IL-6 concentrations. Interestingly, tumor location, alcohol consumption, physical activity, body mass index, as well as ethnicity, education and financial status had no relevant correlation with serum IL-6 levels [110]. We can only speculate regarding the causality of these factors.
Likely, the early disease stages may not be fully represented by biomarkers detected in circulating body fluids. More proximal samples could overcome this issue in localized disease. Therefore, saliva has attracted the interest of researchers during recent years [111,112,113]. It is sometimes referred to as “liquid biopsy”. A non-invasive procedure is required for sample collection, sampling is cost-effective and cancer cells are located within its vicinity [114]. Hence, saliva seems to be a valuable substrate for the early detection of pathological conditions of the oral cavity [112,115].
The physiological salivary level of IL-6 oscillates around 10 pg/mL [116]. Significantly higher concentrations have been reported in OSCC patients compared to healthy individuals [92,117,118,119,120,121,122,123,124,125]. Moreover, some studies showed that OSCC had significantly higher IL-6 levels than oral potentially malignant disorders (OPMD; e.g., leukoplakia, erythroplakia, lichen planus, oral submucous fibrosis) [116,119,123,124,125,126]. Notably, it was also higher compared to chronic oral inflammatory diseases (e.g., chronic periodontitis) [123].
OSCC and these premalignant conditions are sometimes difficult to treat radically by surgery. This increases the importance of consequent watchful follow-up. A sensitive biomarker of early recurrence would thus be of very great diagnostic value. IL-6 concentrations dropped significantly after the successful treatment of OSCC. Salivary IL-6 was therefore proposed as a useful marker to monitor the treatment response [127] and to screen the locoregional recurrence during the patient’s follow up [128,129]. However, it is necessary to associate the laboratory finding with the clinical status of the patient. Of note (as of mid-2021), there are no regulatory authority-approved salivary diagnostic tests available in the USA or Europe for evaluating the risk of oral diseases, including head and neck cancer.

3.5. Anti-IL-6 Therapy

Anti-IL-6 targeting is a promising therapeutic approach in many ways, since activation of IL-6 signaling is associated, for instance, with aggressive tumor growth [99], metastasizing [84,85] and poor responsiveness to chemo- and radiotherapy [86]. A beneficial effect of the blockade can also be increased if used in combinations. Such benefit was observed when IL-6 was targeted together with TNF-α. It is known that TNF-α, similarly to IL-6, is engaged in the occurrence of cancer cachexia and depression [6,130,131]. There are other reports suggesting that a simultaneous blockade of IL-6 and IL-8 can further potentiate the therapeutic outcome [15,132,133,134].
IL-6 can be produced at the tumor site by various cells, but it can also leak and be active in remote body parts. The presence of tumor-associated cytokines in systemic circulation can be a critically important signal for the establishment of premetastatic niche [135], the prerequisite for circulating malignant cell harboring and consequent metastasis formation [10]. Deprivation of tumor cells from these supportive and protective stimuli provided by the microenvironment leaves malignant cells more vulnerable. Therefore, control over IL-6 production could be a clinically attractive strategy. Of note, cytokine synthesis can be inhibited without affecting the viability of cells. This suggests good tolerance and safety profile of this approach and makes this strategy convenient for therapeutic combinations.
Moreover, a plethora of drugs for lowering IL-6 production and release have recently become available on the market and are ready for therapeutic repurposing. The list of these drugs includes well-known anti-inflammatory substances such as acetylsalicylic acid (Aspirin), indomethacin, corticoids, steroidal pregnanes or statins [136,137,138].
On the background of the COVID-19 pandemic, this topic has attracted much attention recently [139]. IL-6 is a multifactorial cytokine and plays a central role in the acute inflammatory response resulting in cytokine storm, a condition which might be fatal for some patients [140]. Upon this ground, several old drugs were extensively investigated for their potential benefits in severe COVID-19 infection. Particularly great attention was dedicated to antimalarials, namely chloroquine and hydroxychloroquine. Based on the recently available Cochrane database analysis, the outcomes of this treatment are controversial [141]. Despite recently strong and polarized opinions on antimalarials among scientists, politicians and the general public, we have to highlight that these substances successfully stood the test of time and were widely used as treatment of several serious autoimmune conditions such as lupus and rheumatoid arthritis. There are evidence-based indications for antimalarials and scientifically sound data suggesting that antimalarials can lower the production of IL-6 in lupus [142] and arthritis [143] patients.
Mechanistically, the deregulated immunity of autoimmune conditions and smoldering protumorigenic inflammation observed in cancer [144] share multiple important features and regulatory molecules, including IL-6 [59]. The immune deregulation of cytokine storm is a very different concept of immune reaction. Failure of antimalarials in acute conditions does not diminish their potential value in chronic inflammation. However, the data on the application of antimalarials in cancer is somewhat sparse and requires careful attention in the future. There are recently running clinical trials focusing on the beneficial role of antimalarials (namely derivatives of artemisinin) in hepatocellular cancer (NCT02304289). A commercially available antimalarial drug, hydroxychloroquine, was used in clinical trials focusing on brain tumors, breast cancer, lung cancer, melanoma, myeloma and other malignancies [145]. For head and neck cancer, a promising effect of quinacrine in combination with platinum derivatives was reported in mouse models [146]. Collectively, antimalarials could potentially exert effects on cell invasion, chemotaxis, cellular trans-differentiation, and clonogenicity in cancer; they are also inhibitors of autophagy [147].
However, when targeting IL-6 production, it is worthy of notice that there are also important physiological functions of this molecule that should not be suppressed entirely. IL-6 is also, e.g., a prominent myokine regulating the homeostasis of skeletal muscles [148]. Here, significant elimination of IL-6 synthesis could have deleterious effects. This function of myokine is of autocrine or paracrine nature.
Therefore, targeting of released and circulating IL-6 represents another therapeutic concept. The first experience with the therapeutic use of anti-IL-6 antibodies dates back to 1988, when mouse anti-IL-6 was tested in patients with multiple myeloma [149]. In the second approved indication, anti-IL-6 was used in patients with Castleman’s disease, where overproduction of IL-6 from the germinal centers of hyperplastic lymph nodes was described [150]. In recent years, the manufacturing of monoclonal antibodies in large quantities for therapeutic purposes has become an affordable option for the pharma industry. It is also possible to produce chimeric or even achieve fully humanized antibodies [151]. These antibodies bind to and neutralize human IL-6 directly, thus decreasing the levels of available free IL-6, and prevent binding to its receptor. For this purpose, siltuximab (CNTO 328) as a chimeric, human-murine immunoglobulin monoclonal antibody produced in Chinese hamster ovary cells was introduced to the market. Later, it was followed by sirukumab (also known as CNTO136), which is a human anti-interleukin (IL)-6 immunoglobulin G1 kappa (IgG1k) monoclonal antibody (mAb), and clazakizumab [152]. For routine use in rheumatoid arthritis approval, additional studies were requested to further define the safety profile of sirukumab [152].
Clazakizumab (in clinical trials also described as ALD518 and BMS-945429), humanized rabbit monoclonal antibody, was raised against interleukin-6 and suggested for treatment of musculoskeletal aspects of psoriatic arthritis and recently also for the management of Late Antibody-Mediated Kidney Transplant Rejection [153].
Olokizumab was also developed for the treatment of rheumatoid arthritis [154]. Mechanistically, it inhibits binding between the IL-6/6R complex and signal transducer gp130. Concerning the availability of IL-6, this offers a particular therapeutic advantage, because this blockade increases systemic levels of available free IL-6 to a lesser degree than other monoclonal antibodies preventing binding of IL-6 and IL-6R (as discussed below).
Full humanization seems critically essential in these applications, because administration of any biologics with immunogenic potential can elicit immune responses leading to the consequent loss of their therapeutic effect. However, this seems to be relatively rare in recently available biologics.
Strategically, targeting the redundantly produced cytokine via antibodies might be a rather labor-intensive, costly and sometimes ungrateful effort. Concerning the ratio of available cytokine/receptor molecules, it seems more prudent and favorable to focus the therapeutic intervention on the IL-6 receptor side [155].
The anti-IL-6R biologics can target either extracellular or intracellular signaling components of the receptor. It is more demanding to administer the drugs within the intracellular compartment since the structures are less accessible [22]. Additionally, downstream effectors, namely JAKs and STATs, could also be targeted. However, it is worthy of notice that these messengers are activated (via their phosphorylation) by a broader range of cytokines, not exclusively by IL-6 [27]. Therefore, we touch them only briefly in our review; however, we have included them in Tables—if their development has reached clinically relevant stage. Similarly, we have deliberately decided to limit the extent of our attention dedicated to highly experimental approaches using gene editing, microRNAs, gene knockouts of signaling cascade members, etc., because these are recently far from clinical applicability [63].
As indicated above, probably the most significant experience with IL-6 blockade was gained in the treatment of rheumatic diseases, especially rheumatoid arthritis (RA), using an antibody against the IL-6 receptor, Tocilizumab [156].
Tocilizumab recognizes the IL-6-binding site of the human IL-6 receptor and thus inhibits IL-6 signaling through competitive blockade of IL-6 binding. Regular dosing of tocilizumab in patients causes no antibody-dependent cellular cytotoxicity or complement-dependent cytotoxicity in cells expressing IL-6R. Tocilizumab recognizes both IL-6R on the cell membrane and soluble IL-6R circulating in body fluids and inhibits both IL-6 signaling and trans-signaling.
Tocilizumab improved clinical signs and symptoms of RA, laboratory parameters, radiological manifestations and ameliorated RA’s effects on patient-reported outcomes, activities of daily living and quality of life when administered as monotherapy or in combination with conventional synthetic disease-modifying antirheumatic drugs [157]. A relatively fundamental study of tumor biology was also presented in clinical trials studying the anti-proliferative effect of Tocilizumab in non-small cell lung cancer cells [158]. Alraouji and coworkers described the effect of Tocilizumab in combination with cisplatin in triple-negative breast cancer. In this study, Tocilizumab potentiated toxicity for cancer stem cells [159]. Thus, tumor stem cells can be a source of self-renewal of tumor clones and failure of conventional chemotherapeutics. The introduction of checkpoint inhibitors into tumor therapy was completely revolutionary. Cemiplimab, as an anti-PD-1 antibody, has shown an excellent therapeutic response in patients with advanced cutaneous squamous cell carcinoma, and it is only a matter of time before it is introduced into HNSCC therapy [160,161]. However, in some patients, there is a weak or no therapeutic response to checkpoint inhibitors. In addition, the immune-mediated mechanisms of resistance have been shown to be challenging to overcome. IL-6 may be one of the molecules involved in the processes of this resistance. It is evidenced by a study of blocking IL-6 and anti-PD-1 simultaneously. Higher levels of IL-6 were detected in some melanoma patients treated with anti-PD-1, which was also associated with a worse prognosis. A simultaneous blockade of IL-6 and this signaling led to an increase in the expression of INF-γ produced by CD4+ T cells and consequent enhancement of anti-PD-1 therapy in a mouse model of melanoma [162]. Insufficient expression of INF-γ in the tumor microenvironment is also considered a mechanism of resistance to checkpoint inhibitors.
Tocilizumab was the “first on the market” representative of this class. It was followed in later years by sarilumab with comparable therapeutic outcomes [163]. Very recently, Satralizumab, a humanized IgG2 monoclonal antibody binding soluble and membrane-bound human IL-6R, was approved for autoimmune optic neuromyelitis [164].
Similarly to IL-6 neutralizing antibodies, there are potential risks associated with the immunogenicity of IL-6R-blocking antibodies. However, these occur rarely in the case of tocilizumab.
Another important issue is the stability of serum concentrations after parenteral administration. The half-life of tocilizumab is approximately two weeks; however, in case of olokizumab, it is around one month. This parameter can be modified, e.g., by engineering using an Fc region mutation to prolong the half-life, as proved by Gerilimzumab, a humanized llama antibody with femtomolar potency [165].
To improve this important pharmacological parameter even further, Vobarilizumab (ALX-0061) was designed as an anti-IL-6R nanobody linked to the anti-human serum albumin domain [166]. Bispecific peptide nanobodies represent single-domain antibody fragments that combine the advantages of conventional biologics with some of the features of small-molecule drugs. A nanobody is much smaller than conventional immunoglobulin (conventional IgG monoclonal antibody MW is 144–148 kDa versus nanobody MW of 26 kDa). The small size improves bioavailability; albumin binding provides increased in vivo half-life of the therapeutic molecule.
It has been extensively studied whether IL-6R can also be inhibited to some extent by small drug inhibitors. A smaller molecule can be easily administered, e.g., orally, which can offer better comfort to the patient. Unlike in cases of e.g., CXCLR1/2 or TGFBRI/II, surprisingly we do not have any highly selective small drug inhibitors of IL-6R available currently.
As mentioned earlier, the IL-6 signaling uses glycoprotein gp130 as a signal transducer. Therefore, this molecule represents an attractive therapeutic target. It is expressed by virtually all cell types, which further increases its value. Attempts to provide therapeutic blockade via monoclonal antibodies raised against gp130 were published already in the 1990s [167]. Surprisingly, different antibodies were able to antagonize the biological activities of all cytokines belonging to the IL-6 cytokine family in a specific manner.
For many years, the chemical synthesis of gp130 inhibitors was thought to be extremely challenging. This gave gp130 the reputation of a conventionally undruggable target. However, some natural compounds can bind to the extracellular domain of gp130, namely Madindoline-A. The binding is specific and non-covalent, and displays relatively low affinity [168]. Synthetic analogues of Madindoline-A with improved pharmacologic parameters were synthesized later [168].
Using strategies of molecular design, potent inhibitors were finally synthesized in the last decade—namely SC144 [169], LMT-28 [170].
More recently, it was discovered that bazedoxifene and raloxifene and their derivatives, routinely used drugs for postmenopausal osteoporosis in clinics, inhibit IL-6/gp130 protein–protein interactions. By this mechanism, drugs of the bazedoxifene type and their derivatives cause significant inhibition of IL-6-dependent STAT3 phosphorylation [171]. Recent data suggested that even these well-known drugs can be repurposed for cancer treatment. The proof of this concept was confirmed in gastrointestinal cancer in a mouse model [172].
For signal transduction, gp130 is typically bound to the cell membrane. However, the soluble gp130 form is also known. The mechanism leading to the production of soluble gp130 has been discussed; whether it is formed via alternative splicing, or as suggested more recently, is a product of BACE cleavage [173]. In any case, the soluble form of gp130 binds the IL-6/IL-6R complex, but does not transduce the signal to the cell anymore. This can also be understood as an inhibitory effect, specifically inhibiting trans-signaling. Of note, trans-signaling inhibitor olamkicept was well tolerated in the treatment of inflammatory bowel disease, and this drug was administered without signs of immunosuppression in patients [174]. Gp130 is a large molecule, but it could be manufactured in a miniaturized form, which could be a pharmacologically important modification [173].
Multiple preclinical studies on IL-6 targeting have been completed (see Table 1) [63,88,175,176,177,178,179,180,181,182,183,184,185,186,187,188,189,190,191,192,193,194,195,196,197,198,199,200,201,202,203,204,205,206,207,208,209,210,211]. All focused both on the production of IL-6 and its signaling. There is an array of therapeutics possibly applicable to HNSCC treatment. They were tested either in vitro or in vivo; a few of the studies combined both approaches. Interestingly, some of the investigated therapeutics are already commercially available for the treatment of autoimmune diseases (e.g., Tocilizumab in rheumatoid arthritis [16]). Generally, state of the art anti-IL-6 compounds interfere with IL-6 production, IL-6 availability, inhibition of IL-6R or the IL-6/sIL-6R complex, inhibition of IL-6R signal transducer gp130 and downstream signaling effector molecules [22]. As mentioned above, the results are predominantly preclinical.
On the other hand, there is a rapidly increasing number of approved IL-6-related therapeutic agents, which have not been tested in HNSCC yet (see Supplementary Table S1). As these drugs have already acquired approval and are marketed for other indications, this also offers certain hopes for HNSCC. It is known very well that drug repurposing has a number of interrelated advantages, such as simplification in the regulatory procedures for introducing a previously approved drug on the market. These medications are already well characterized in terms of the drug’s safety and toxicity. This makes the process considerably faster, and therefore also cheaper compared to de novo drug discovery. Hence, robust clinical trials need to be conducted in order to verify the experimental conclusions in clinical practice.

4. Conclusions

HNSCC remains a challenging field of medicine because of the high incidence and unfavorable result of therapy in numerous patients. In many cases, standard surgical and oncological therapy fails. There is a critical need for novel therapeutic approaches. IL-6, as a highly potent cytokine, appears to be a promising target. The concept of IL-6 targeting was proved in rheumatologic diseases.
However, several issues relating to IL-6 biology remain unanswered. It is unclear why IL-6 signal inhibition leads to clinically significant benefits for patients with some, but not all, rheumatologic conditions.
Tumors can have different underlying genetic causes, which can result in disease heterogeneity. Due to this heterogeneity, any therapy can be effective in only a subset of the patient population. Such inherent variability of cancer requires increased diagnostic precision of reliable biomarkers and personalized approaches to therapy.
If IL-6 is rightfully believed to be a biologically powerful player in cancer biology, here comes an inevitable question: Why has IL-6 not gained a similar position as a recognized biomarker (of any cancer type including HNSCC) so far? We believe that IL-6 and IL-6-related signaling differ greatly from the routinely used biomarkers in cancer research. More conventional biomarkers are based mostly on malignant cell properties in terms of DNA mutation, RNA expression, protein presence or release, etc. This simplistic approach centered on a malignant cell might be misleading in the case of IL-6. The IL-6 signaling mechanism is far more complex and exceeds a single cell type. It cannot be assigned to the malignant cell population only. It requires a broader view, and one must take the whole tumor microenvironment into account, as was also proposed for HNSCC in our previous review [13]. It is not critical whether a particular cell type expresses the cytokine or the receptor, because the other components of the tumor ecosystem can readily substitute for whatever is missing. The cytokine can diffuse and reach areas remote from where it was produced. Moreover, the soluble receptor from donor cells can activate acceptor cells that do not express it on their own. Thus, IL-6 signaling deregulation is more likely a marker of overall sick social liaisons of the tumor microenvironment rather than a tool for the precise assessment of individual members of this ecosystem, which was also demonstrated for HNSCC in vitro and in clinical material [15]. This is highly challenging for routine analytic methods such as immunohistochemistry. Approaches based on multiplexing could be very helpful in this in the future.
Of note, individualization of therapy can lead to unsustainable expenses in healthcare systems. Therefore, a careful selection of targets applicable to broader patient populations can be beneficial. IL-6 is an essential component in the cancer microenvironment orchestration and contributes to some of the adverse effects of the malignant disease (e.g., cachexia and depression).
A plethora of potential anti-IL-6 therapeutics have been preclinically investigated; many available drugs could also be repurposed for application in cancer therapy (shortlisted in Supplementary Table S1; [25,146,154,155,165,169,170,173,174,228,229,230,231,232,233,234,235,236,237,238,239,240,241,242,243,244,245,246,247,248,249,250,251,252,253,254]). Available data indicate potential benefits for certain types of solid malignancies, which could also apply to HNSCC patients. However, there is still a need for validation of these preliminary data in sufficiently large clinical studies that could finally prove the efficacy and safety of this newly emerging therapeutic concept.

Supplementary Materials

The following are available online at https://www.mdpi.com/article/10.3390/ijms222011027/s1.

Author Contributions

Conceptualization: K.S.J., M.Š. and L.L.; data collection: O.K., P.S., J.V., K.S., A.J.P. and B.D., original draft writing: M.Š., K.S.J. and L.L., funding acquisition: K.S.J. All authors have read and agreed to the published version of the manuscript.

Funding

This research was funded by the project “Centre for Tumor Ecology—Research of the Cancer Microenvironment Supporting Cancer Growth and Spread “(No. CZ.02.1.01/0.0/0.0/16_019/0000785), supported by the Operational Program “Research, Development and Education” and by a project of Charles University in Prague (PROGRES Q28).

Acknowledgments

The authors are grateful to Šárka Takáčová for the language revision of the manuscript.

Conflicts of Interest

The authors declare no conflict of interest.

Abbreviations

Aside from official gene names, the following abbreviations were used in the text and legends:
α-SMAα-Smooth muscle actin
CAFCancer-associated fibroblast
CBPCREB-binding protein
ECMExtracellular matrix
EMTEpithelial to mesenchymal transition
GAB1GRB2-associated-binding protein
gp130Β-Receptor glycoprotein 130
HNCHead and neck cancer
HNSCCHead and neck squamous cell carcinoma
HPVHuman papilloma virus
IFNInterferon
IL-6Interleukin 6
IL-8Interleukin 8
IL-6RReceptor for IL-6
JAKJanus kinase
JAK-STATJanus kinase and signal transducer and activator of transcription
MAPKMitogen-activated protein kinase
OPMDOral potentially malignant disorder
OSCCOral squamous cell carcinoma
PIASProtein inhibitor of activated STAT
PI3KPhosphatidylinositol-3-kinase
RARheumatoid arthritis
SOCS3Suppressor of cytokine signaling 3
STATSignal transducer and activator of transcription
TMETumor microenvironment
TNF-αTumor necrosis factor α

References

  1. Warnakulasuriya, S. Global Epidemiology of Oral and Oropharyngeal Cancer. Oral Oncol. 2009, 45, 309–316. [Google Scholar] [CrossRef]
  2. Vokes, E.E.; Weichselbaum, R.R.; Lippman, S.M.; Hong, W.K. Head and Neck Cancer. N. Engl. J. Med. 1993, 328. [Google Scholar] [CrossRef] [PubMed]
  3. Novák, Š.; Bandurová, V.; Mifková, A.; Kalfeřt, D.; Fík, Z.; Lukeš, P.; Szabo, P.; Plzák, J.; Smetana, K., Jr. Tumor Microenvironment. Otorinolaryng. A Foniat. 2019, 68, 41–51. [Google Scholar]
  4. Smetana, K.; Lacina, L.; Szabo, P.; Dvořánková, B.; Brož, P.; Šedo, A. Ageing as an Important Risk Factor for Cancer. Anticancer Res. 2016, 36, 5009–5017. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  5. Saussez, S.; Duray, A.; Demoulin, S.; Hubert, P.; Delvenne, P. Immune Suppression in Head and Neck Cancers: A Review. Clin. Dev. Immunol. 2010, 2010, 15. [Google Scholar] [CrossRef]
  6. Lacina, L.; Brábek, J.; Král, V.; Kodet, O.; Smetana, K. Interleukin-6: A Molecule with Complex Biological Impact in Cancer. Histol. Histopathol. 2019, 34, 125–136. [Google Scholar] [CrossRef]
  7. Hamburger, A.W.; Salmon, S.E. Primary Bioassay of Human Tumor Stem Cells. Science 1977, 197, 461–463. [Google Scholar] [CrossRef]
  8. Metwaly, H.; Maruyama, S.; Yamazaki, M.; Tsuneki, M.; Abé, T.; Jen, K.Y.; Cheng, J.; Saku, T. Parenchymal-Stromal Switching for Extracellular Matrix Production on Invasion of Oral Squamous Cell Carcinoma. Hum. Pathol. 2012, 43, 1973–1981. [Google Scholar] [CrossRef]
  9. Polyak, K.; Haviv, I.; Campbell, I.G. Co-Evolution of Tumor Cells and Their Microenvironment. Trends Genet. 2009, 25, 30–38. [Google Scholar] [CrossRef]
  10. Lacina, L.; Plzak, J.; Kodet, O.; Szabo, P.; Chovanec, M.; Dvorankova, B.; Smetana, K. Cancer Microenvironment: What Can We Learn from the Stem Cell Niche. Int. J. Mol. Sci. 2015, 16, 24094–24110. [Google Scholar] [CrossRef]
  11. Li, H.; Fan, X.; Houghton, J.M. Tumor Microenvironment: The Role of the Tumor Stroma in Cancer. J. Cell. Biochem. 2007, 101, 805–815. [Google Scholar] [CrossRef]
  12. Lorusso, G.; Rüegg, C. The Tumor Microenvironment and Its Contribution to Tumor Evolution toward Metastasis. Histochem. Cell Biol. 2008, 130, 1091–1103. [Google Scholar] [CrossRef] [Green Version]
  13. Plzák, J.; Lacina, L.; Chovanec, M.; Dvořánková, B.; Szabo, P.; Čada, Z.; Smetana, K. Epithelial–Stromal Interaction in Squamous Cell Epithelium-Derived Tumors: An Important New Player in the Control of Tumor Biological Properties. Anticancer Res. 2010, 30. [Google Scholar]
  14. Fisher, D.T.; Appenheimer, M.M.; Evans, S.S. The Two Faces of IL-6 in the Tumor Microenvironment. Semin. Immunol. 2014, 26, 38–47. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  15. Kolář, M.; Szabo, P.; Dvořánková, B.; Lacina, L.; Gabius, H.J.; Strnad, H.; Šáchová, J.; Vlček, Č.; Plzák, J.; Chovanec, M.; et al. Upregulation of IL-6, IL-8 and CXCL-1 Production in Dermal Fibroblasts by Normal/Malignant Epithelial Cells in Vitro: Immunohistochemical and Transcriptomic Analyses. Biol. Cell 2012, 104, 738–751. [Google Scholar] [CrossRef] [PubMed]
  16. Rossi, J.F.; Lu, Z.Y.; Jourdan, M.; Klein, B. Interleukin-6 as a Therapeutic Target. Clin. Cancer Res. 2015, 21, 1248–1257. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  17. Yamasaki, K.; Taga, T.; Hirata, Y.; Yawata, H.; Kawanishi, Y.; Seed, B.; Taniguchi, T.; Hirano, T.; Kishimoto, T. Cloning and Expression of the Human Interleukin-6 (BSF-2/IFNβ 2) Receptor. Science 1988, 241, 825–828. [Google Scholar] [CrossRef] [PubMed]
  18. Hirano, T.; Yasukawa, K.; Harada, H.; Taga, T.; Watanabe, Y.; Matsuda, T.; Kashiwamura, S.I.; Nakajima, K.; Koyama, K.; Iwamatsu, A.; et al. Complementary DNA for a Novel Human Interleukin (BSF-2) That Induces B Lymphocytes to Produce Immunoglobulin. Nature 1986, 324, 73–76. [Google Scholar] [CrossRef]
  19. Scheller, J.; Rose-John, S. Interleukin-6 and Its Receptor: From Bench to Bedside. Med. Microbiol. Immunol. 2006, 195, 173–183. [Google Scholar] [CrossRef] [PubMed]
  20. Rose-John, S. Interleukin-6 Biology Is Coordinated by Membrane-Bound and Soluble Receptors: Role in Inflammation and Cancer. J. Leukoc. Biol. 2006, 80, 227–236. [Google Scholar] [CrossRef] [Green Version]
  21. Wolf, J.; Rose-John, S.; Garbers, C. Interleukin-6 and Its Receptors: A Highly Regulated and Dynamic System. Cytokine 2014, 70, 11–20. [Google Scholar] [CrossRef] [PubMed]
  22. Schaper, F.; Rose-John, S. Interleukin-6: Biology, Signaling and Strategies of Blockade. Cytokine Growth Factor Rev. 2015, 26, 475–487. [Google Scholar] [CrossRef] [PubMed]
  23. Dimitrov, S.; Lange, T.; Benedict, C.; Nowell, M.A.; Jones, S.A.; Scheller, J.; Rose-John, S.; Born, J.; Dimitrov, S.; Lange, T.; et al. Sleep Enhances IL-6 Trans-signaling in Humans. FASEB J. 2006, 20, 2174–2176. [Google Scholar] [CrossRef] [PubMed]
  24. Scheller, J.; Garbers, C.; Rose-John, S. Interleukin-6: From Basic Biology to Selective Blockade of pro-Inflammatory Activities. Semin. Immunol. 2014, 26, 2–12. [Google Scholar] [CrossRef] [PubMed]
  25. Jostock, T.; Müllberg, J.; Özbek, S.; Atreya, R.; Blinn, G.; Voltz, N.; Fischer, M.; Neurath, M.F.; Rose-John, S. Soluble Gp130 Is the Natural Inhibitor of Soluble Interleukin-6 Receptor Transsignaling Responses. Eur. J. Biochem. 2001, 268, 160–167. [Google Scholar] [CrossRef]
  26. 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, 1–20. [Google Scholar] [CrossRef] [Green Version]
  27. Eulenfeld, R.; Dittrich, A.; Khouri, C.; Müller, P.J.; Mütze, B.; Wolf, A.; Schaper, F. Interleukin-6 Signalling: More than Jaks and STATs. Eur. J. Cell Biol. 2012, 91, 486–495. [Google Scholar] [CrossRef]
  28. Kretzschmar, A.K.; Dinger, M.C.; Henze, C.; Brocke-Heidrich, K.; Horn, F. Analysis of Stat3 (Signal Transducer and Activator of Transcription 3) Dimerization by Fluorescence Resonance Energy Transfer in Living Cells. Biochem. J. 2004, 377, 289–297. [Google Scholar] [CrossRef] [Green Version]
  29. Braunstein, J.; Brutsaert, S.; Olson, R.; Schindler, C. STATs Dimerize in the Absence of Phosphorylation. J. Biol. Chem. 2003, 278, 34133–34140. [Google Scholar] [CrossRef] [Green Version]
  30. Haan, S.; Knotholes, M.; Behrmann, I.; Müller-Esterl, W.; Heinrich, P.C.; Schaper, F. Cytoplasmic STAT Proteins Associate Prior to Activation. Biochem. J. 2000, 345, 417–421. [Google Scholar] [CrossRef]
  31. Novak, U.; Ji, H.; Kanagasundaram, V.; Simpson, R.; Paradiso, L. STAT3 Forms Stable Homodimers in the Presence of Divalent Cations Prior to Activation. Biochem. Biophys. Res. Commun. 1998, 247, 558–563. [Google Scholar] [CrossRef]
  32. Vogt, M.; Domoszlai, T.; Kleshchanok, D.; Lehmann, S.; Schmitt, A.; Poli, V.; Richtering, W.; Müller-Newen, G. The Role of the N-Terminal Domain in Dimerization and Nucleocytoplasmic Shuttling of Latent STAT3. J. Cell Sci. 2011, 124, 900–909. [Google Scholar] [CrossRef] [Green Version]
  33. Pranada, A.L.; Metz, S.; Herrmann, A.; Heinrich, P.C.; Müller-Newen, G. Real Time Analysis of STAT3 Nucleocytoplasmic Shuttling. J. Biol. Chem. 2004, 279, 15114–15123. [Google Scholar] [CrossRef] [Green Version]
  34. Ogryzko, V.V.; Schiltz, R.L.; Russanova, V.; Howard, B.H.; Nakatani, Y. The Transcriptional Coactivators P300 and CBP Are Histone Acetyltransferases. Cell 1996, 87, 953–959. [Google Scholar] [CrossRef] [Green Version]
  35. Ray, S.; Boldogh, I.; Brasier, A.R. STAT3 NH2-Terminal Acetylation Is Activated by the Hepatic Acute-Phase Response and Required for IL-6 Induction of Angiotensinogen. Gastroenterology 2005, 129, 1616–1632. [Google Scholar] [CrossRef] [PubMed]
  36. Yuan, Z.L.; Guan, Y.J.; Chatterjee, D.; Chin, Y.E. Stat3 Dimerization Regulated by Reversible Acetylation of a Single Lysine Residue. Science 2005, 307, 269–273. [Google Scholar] [CrossRef] [PubMed]
  37. Hou, T.; Ray, S.; Lee, C.; Brasier, A.R. The STAT3 NH2-Terminal Domain Stabilizes Enhanceosome Assembly by Interacting with the P300 Bromodomain. J. Biol. Chem. 2008, 283, 30725–30734. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  38. Fukada, T.; Hibi, M.; Yamanaka, Y.; Takahashi-Tezuka, M.; Fujitani, Y.; Yamaguchi, T.; Nakajima, K.; Hirano, T. Two Signals Are Necessary for Cell Proliferation Induced by a Cytokine Receptor Gp130: Involvement of STAT3 in Anti-Apoptosis. Immunity 1996, 5, 449–460. [Google Scholar] [CrossRef] [Green Version]
  39. Fukada, T.; Ohtani, T.; Yoshida, Y.; Shirogane, T.; Nishida, K.; Nakajima, K.; Hibi, M.; Hirano, T. STAT3 Orchestrates Contradictory Signals in Cytokine-Induced G1 to S Cell-Cycle Transition. EMBO J. 1998, 17, 6670–6677. [Google Scholar] [CrossRef] [Green Version]
  40. Judd, L.M.; Alderman, B.M.; Howlett, M.; Shulkes, A.; Dow, C.; Moverley, J.; Grail, D.; Jenkins, B.J.; Ernst, M.; Giraud, A.S. Gastric Cancer Development in Mice Lacking the SHP2 Binding Site on the IL-6 Family Co-Receptor Gp130. Gastroenterology 2004, 126, 196–207. [Google Scholar] [CrossRef]
  41. Tebbutt, N.C.; Giraud, A.S.; Inglese, M.; Jenkins, B.; Waring, P.; Clay, F.J.; Malki, S.; Alderman, B.M.; Grail, D.; Hollande, F.; et al. Reciprocal Regulation of Gastrointestinal Homeostasis by SHP2 and STAT-Mediated Trefoil Gene Activation in Gp130 Mutant Mice. Nat. Med. 2002, 8, 1089–1097. [Google Scholar] [CrossRef]
  42. Schaper, F.; Gendo, C.; Eck, M.; Schmitz, J.; Grimm, C.; Anhuf, D.; Kerr, I.M.; Heinrich, P.C. Activation of the Protein Tyrosine Phosphatase SHP2 via the Interleukin-6 Signal Transducing Receptor Protein Gp130 Requires Tyrosine Kinase Jak1 and Limits Acute-Phase Protein Expression. Biochem. J. 1998, 335, 557–565. [Google Scholar] [CrossRef]
  43. Symes, A.; Stahl, N.; Reeves, S.A.; Farruggella, T.; Servidei, T.; Gearan, T.; Yancopoulos, G.; Stephen Fink, J. The Protein Tyrosine Phosphatase SHP-2 Negatively Regulates Ciliary Neurotrophic Factor Induction of Gene Expression. Curr. Biol. 1997, 7, 697–700. [Google Scholar] [CrossRef] [Green Version]
  44. Schiemann, W.P.; Bartoe, J.L.; Nathanson, N.M. Box 3-Independent Signaling Mechanisms Are Involved in Leukemia Inhibitory Factor Receptor α- and Gp130-Mediated Stimulation of Mitogen-Activated Protein Kinase. Evidence for Participation of Multiple Signaling Pathways Which Converge at Ras. J. Biol. Chem. 1997, 272, 16631–16636. [Google Scholar] [CrossRef] [Green Version]
  45. Lai, C.F.; Ripperger, J.; Wang, Y.; Kim, H.; Hawley, R.B.; Baumann, H. The STAT3-Independent Signaling Pathway by Glycoprotein 130 in Hepatic Cells. J. Biol. Chem. 1999, 274, 7793–7802. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  46. Ernst, M.; Jenkins, B.J. Acquiring Signalling Specificity from the Cytokine Receptor Gp130. Trends Genet. 2004, 20, 23–32. [Google Scholar] [CrossRef]
  47. Lehmann, U.; Schmitz, J.; Weissenbach, M.; Sobota, R.M.; Hörtner, M.; Friederichs, K.; Behrmann, I.; Tsiaris, W.; Sasaki, A.; Schneider-Mergener, J.; et al. SHP2 and SOCS3 Contribute to Tyr-759-Dependent Attenuation of Interleukin-6 Signaling through Gp130. J. Biol. Chem. 2003, 278, 661–671. [Google Scholar] [CrossRef] [Green Version]
  48. Kim, H.; Baumann, H. Dual Signaling Role of the Protein Tyrosine Phosphatase SHP-2 in Regulating Expression of Acute-Phase Plasma Proteins by Interleukin-6 Cytokine Receptors in Hepatic Cells. Mol. Cell. Biol. 1999, 19, 5326–5338. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  49. De Souza, D.; Fabri, L.J.; Nash, A.; Hilton, D.J.; Nicola, N.A.; Baca, M. SH2 Domains from Suppressor of Cytokine Signaling-3 and Protein Tyrosine Phosphatase SHP-2 Have Similar Binding Specificities. Biochemistry 2002, 41, 9229–9236. [Google Scholar] [CrossRef]
  50. Eulenfeld, R.; Schaper, F. A New Mechanism for the Regulation of Gab1 Recruitment to the Plasma Membrane. J. Cell Sci. 2009, 122, 55–64. [Google Scholar] [CrossRef] [Green Version]
  51. Takahashi-Tezuka, M.; Yoshida, Y.; Fukada, T.; Ohtani, T.; Yamanaka, Y.; Nishida, K.; Nakajima, K.; Hibi, M.; Hirano, T. Gab1 Acts as an Adapter Molecule Linking the Cytokine Receptor Gp130 to ERK Mitogen-Activated Protein Kinase. Mol. Cell. Biol. 1998, 18, 4109–4117. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  52. Wang, Y.; Fuller, G.M. Phosphorylation and Internalization of Gp130 Occur after IL-6 Activation of Jak2 Kinase in Hepatocytes. Mol. Biol. Cell 1994, 5, 819–828. [Google Scholar] [CrossRef] [Green Version]
  53. Zohlnhöfer, D.; Graeve, L.; Rose-John, S.; Schooltink, H.; Dittrich, E.; Heinrich, P.C. The Hepatic Interleukin-6 Receptor Down-Regulation of the Interleukin-6 Binding Subunit (Gp80) by Its Ligand. FEBS Lett. 1992, 306, 219–222. [Google Scholar] [CrossRef] [Green Version]
  54. Radtke, S.; Wüller, S.; Yang, X.P.; Lippok, B.E.; Mütze, B.; Mais, C.; Schmitz-Van De Leur, H.; Bode, J.G.; Gaestel, M.; Heinrich, P.C.; et al. Cross-Regulation of Cytokine Signalling: Pro-Inflammatory Cytokines Restrict IL-6 Signalling through Receptor Internalisation and Degradation. J. Cell Sci. 2010, 123, 947–959. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  55. Chung, C.D.; Liao, J.; Liu, B.; Rao, X.; Jay, P.; Berta, P.; Shuai, K. Specific Inhibition of Stat3 Signal Transduction by PIAS3. Science 1997, 278, 1803–1805. [Google Scholar] [CrossRef]
  56. Scatena, R.; Bottoni, P.; Pontoglio, A.; Giardina, B. Cancer Stem Cells: The Development of New Cancer Therapeutics. Expert Opin. Biol. Ther. 2011, 11, 875–892. [Google Scholar] [CrossRef]
  57. Korkaya, H.; Liu, S.; Wicha, M.S. Breast Cancer Stem Cells, Cytokine Networks, and the Tumor Microenvironment. J. Clin. Investig. 2011, 121, 3804–3809. [Google Scholar] [CrossRef]
  58. Gál, P.; Varinská, L.; Fáber, L.; Novák, Š.; Szabo, P.; Mitrengová, P.; Mirossay, A.; Mučaji, P.; Smetana, K. How Signaling Molecules Regulate Tumor Microenvironment: Parallels to Wound Repair. Molecules 2017, 22, 1818. [Google Scholar] [CrossRef] [Green Version]
  59. Brábek, J.; Jakubek, M.; Vellieux, F.; Novotný, J.; Kolář, M.; Lacina, L.; Szabo, P.; Strnadová, K.; Rösel, D.; Dvořánková, B.; et al. Interleukin-6: Molecule in the Intersection of Cancer, Ageing and COVID-19. Int. J. Mol. Sci. 2020, 21, 7937. [Google Scholar] [CrossRef]
  60. Östman, A.; Augsten, M. Cancer-Associated Fibroblasts and Tumor Growth - Bystanders Turning into Key Players. Curr. Opin. Genet. Dev. 2009, 19, 67–73. [Google Scholar] [CrossRef] [PubMed]
  61. Lamertz, L.; Rummel, F.; Polz, R.; Baran, P.; Hansen, S.; Waetzig, G.H.; Moll, J.M.; Floss, D.M.; Scheller, J. Soluble Gp130 Prevents Interleukin-6 and Interleukin-11 Cluster Signaling but Not Intracellular Autocrine Responses. Sci. Signal. 2018, 11. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  62. Heink, S.; Yogev, N.; Garbers, C.; Herwerth, M.; Aly, L.; Gasperi, C.; Husterer, V.; Croxford, A.L.; Möller-Hackbarth, K.; Bartsch, H.S.; et al. Trans-Presentation of Interleukin-6 by Dendritic Cells Is Required for Priming Pathogenic TH17 Cells. Nat. Immunol. 2017, 18, 74. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  63. Yu, C.C.; Tsai, L.L.; Wang, M.L.; Yu, C.H.; Lo, W.L.; Chang, Y.C.; Chiou, G.Y.; Chou, M.Y.; Chiou, S.H. MiR145 Targets the SOX9/ADAM17 Axis to Inhibit Tumor-Initiating Cells and IL-6-Mediated Paracrine Effects in Head and Neck Cancer. Cancer Res. 2013, 73, 3425–3440. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  64. Szabo, P.; Valach, J.; Smetana, K., Jr.; Dvorankova, B. Comparative Analysis of IL-8 and CXCL-1 Production by Normal and Cancer Stromal Fibroblasts. Folia Biol. 2013, 59, 134–137. [Google Scholar]
  65. Bremnes, R.M.; Dønnem, T.; Al-Saad, S.; Al-Shibli, K.; Andersen, S.; Sirera, R.; Camps, C.; Marinez, I.; Busund, L.T. The Role of Tumor Stroma in Cancer Progression and Prognosis: Emphasis on Carcinoma-Associated Fibroblasts and Non-Small Cell Lung Cancer. J. Thorac. Oncol. 2011, 6, 209–217. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  66. Novotný, J.; Strnadová, K.; Dvořánková, B.; Kocourková, Š.; Jakša, R.; Dundr, P.; Pačes, V.; Smetana, K.; Kolář, M.; Lacina, L. Single-Cell RNA Sequencing Unravels Heterogeneity of the Stromal Niche in Cutaneous Melanoma Heterogeneous Spheroids. Cancers 2020, 12, 3324. [Google Scholar] [CrossRef]
  67. Erez, N.; Truitt, M.; Olson, P.; Hanahan, D. Cancer-Associated Fibroblasts Are Activated in Incipient Neoplasia to Orchestrate Tumor-Promoting Inflammation in an NF-ΚB-Dependent Manner. Cancer Cell 2010, 17, 135–147. [Google Scholar] [CrossRef] [Green Version]
  68. Ganguly, D.; Chandra, R.; Karalis, J.; Teke, M.; Aguilera, T.; Maddipati, R.; Wachsmann, M.B.; Ghersi, D.; Siravegna, G.; Zeh, H.J.; et al. Cancer-Associated Fibroblasts: Versatile Players in the Tumor Microenvironment. Cancers 2020, 12, 2652. [Google Scholar] [CrossRef]
  69. Lacina, L.; Smetana, K.; Dvořánková, B.; Pytlík, R.; Kideryová, L.; Kučerová, L.; Plzáková, Z.; Štork, J.; Gabius, H.J.; André, S. Stromal Fibroblasts from Basal Cell Carcinoma Affect Phenotype of Normal Keratinocytes. Br. J. Dermatol. 2007, 156, 819–829. [Google Scholar] [CrossRef]
  70. Lacina, L.; Dvořánkova, B.; Smetana, K.; Chovanec, M.; Plzǎk, J.; Tachezy, R.; Kideryovǎ, L.; Kučerová, L.; Čada, Z.; Bouček, J.; et al. Marker Profiling of Normal Keratinocytes Identifies the Stroma from Squamous Cell Carcinoma of the Oral Cavity as a Modulatory Microenvironment in Co-Culture. Int. J. Radiat. Biol. 2007, 83, 837–848. [Google Scholar] [CrossRef]
  71. Strnad, H.; Lacina, L.; Kolář, M.; Čada, Z.; Vlček, Č.; Dvořánková, B.; Betka, J.; Plzák, J.; Chovanec, M.; Šáchova, J.; et al. Head and Neck Squamous Cancer Stromal Fibroblasts Produce Growth Factors Influencing Phenotype of Normal Human Keratinocytes. Histochem. Cell Biol. 2010, 133, 201–211. [Google Scholar] [CrossRef]
  72. Szabó, P.; Kolář, M.; Dvořánková, B.; Lacina, L.; Štork, J.; Vlček, Č.; Strnad, H.; Tvrdek, M.; Smetana, K. Mouse 3T3 Fibroblasts under the Influence of Fibroblasts Isolated from Stroma of Human Basal Cell Carcinoma Acquire Properties of Multipotent Stem Cells. Biol. Cell 2011, 103, 233–248. [Google Scholar] [CrossRef]
  73. Flier, J.S.; Underhill, L.H.; Dvorak, H.F. Tumors: Wounds That Do Not Heal. N. Engl. J. Med. 1986, 315, 1650–1659. [Google Scholar] [CrossRef]
  74. Dvoánková, B.; Szabo, P.; Lacina, L.; Gal, P.; Uhrova, J.; Zima, T.; Kaltner, H.; André, S.; Gabius, H.J.; Sykova, E.; et al. Human Galectins Induce Conversion of Dermal Fibroblasts into Myofibroblasts and Production of Extracellular Matrix: Potential Application in Tissue Engineering and Wound Repair. Cells Tissues Organs 2011, 194, 469–480. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  75. Valach, J.; Fík, Z.; Strnad, H.; Chovanec, M.; Plzák, J.; Čada, Z.; Szabo, P.; Šáchová, J.; Hroudová, M.; Urbanová, M.; et al. Smooth Muscle Actin-Expressing Stromal Fibroblasts in Head and Neck Squamous Cell Carcinoma: Increased Expression of Galectin-1 and Induction of Poor Prognosis Factors. Int. J. Cancer 2012, 131, 2499–2508. [Google Scholar] [CrossRef] [PubMed]
  76. Hinz, B. Formation and Function of the Myofibroblast during Tissue Repair. J. Investig. Dermatol. 2007, 127, 526–537. [Google Scholar] [CrossRef] [PubMed]
  77. Clark, R.A.F.; Ghosh, K.; Tonnesen, M.G. Tissue Engineering for Cutaneous Wounds. J. Investig. Dermatol. 2007, 127, 1018–1029. [Google Scholar] [CrossRef] [Green Version]
  78. Deonarine, K.; Panelli, M.C.; Stashower, M.E.; Jin, P.; Smith, K.; Slade, H.B.; Norwood, C.; Wang, E.; Marincola, F.M.; Stroncek, D.F. Gene Expression Profiling of Cutaneous Wound Healing. J. Transl. Med. 2007, 5, 11. [Google Scholar] [CrossRef] [Green Version]
  79. Smetana, K.; Szabo, P.; Gál, P.; André, S.; Gabius, H.J.; Kodet, O.; Dvořánková, B. Emerging Role of Tissue Lectins as Microenvironmental Effectors in Tumors and Wounds. Histol. Histopathol. 2015, 30, 293–309. [Google Scholar] [CrossRef]
  80. Werner, S.; Krieg, T.; Smola, H. Keratinocyte-Fibroblast Interactions in Wound Healing. J. Investig. Dermatol. 2007, 127, 998–1008. [Google Scholar] [CrossRef] [Green Version]
  81. Slaughter, D.P.; Southwick, H.W.; Smejkal, W. “Field Cancerization” in Oral Stratified Squamous Epithelium. Clinical Implications of Multicentric Origin. Cancer 1953, 6, 963–968. [Google Scholar] [CrossRef]
  82. Ishii, T.; Suzuki, A.; Kuwata, T.; Hisamitsu, S.; Hashimoto, H.; Ohara, Y.; Yanagihara, K.; Mitsunaga, S.; Yoshino, T.; Kinoshita, T.; et al. Drug-Exposed Cancer-Associated Fibroblasts Facilitate Gastric Cancer Cell Progression Following Chemotherapy. Gastric Cancer 2021, 24. [Google Scholar] [CrossRef]
  83. Bharti, R.; Dey, G.; Mandal, M. Cancer Development, Chemoresistance, Epithelial to Mesenchymal Transition and Stem Cells: A Snapshot of IL-6 Mediated Involvement. Cancer Lett. 2016, 375, 51–61. [Google Scholar] [CrossRef]
  84. Krishnamurthy, S.; Warner, K.A.; Dong, Z.; Imai, A.; Nör, C.; Ward, B.B.; Helman, J.I.; Taichman, R.S.; Bellile, E.L.; McCauley, L.K.; et al. Endothelial Interleukin-6 Defines the Tumorigenic Potential of Primary Human Cancer Stem Cells. Stem Cells 2014, 32, 2845–2857. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  85. Kim, H.S.; Chen, Y.-C.; Nör, F.; Warner, K.A.; Andrews, A.; Wagner, V.P.; Zhang, Z.; Zhang, Z.; Martins, M.D.; Pearson, A.T.; et al. Endothelial-Derived Interleukin-6 Induces Cancer Stem Cell Motility by Generating a Chemotactic Gradient towards Blood Vessels. Oncotarget 2017, 8, 100339–100352. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  86. Jinno, T.; Kawano, S.; Maruse, Y.; Matsubara, R.; Goto, Y.; Sakamoto, T.; Hashiguchi, Y.; Kaneko, N.; Tanaka, H.; Kitamura, R.; et al. Increased Expression of Interleukin-6 Predicts Poor Response to Chemoradiotherapy and Unfavorable Prognosis in Oral Squamous Cell Carcinoma. Oncol. Rep. 2015, 33, 2161–2168. [Google Scholar] [CrossRef] [Green Version]
  87. Gao, J.; Zhao, S.; Halstensen, T.S. Increased Interleukin-6 Expression Is Associated with Poor Prognosis and Acquired Cisplatin Resistance in Head and Neck Squamous Cell Carcinoma. Oncol. Rep. 2016, 35, 3265–3274. [Google Scholar] [CrossRef] [Green Version]
  88. Matsuoka, Y.; Nakayama, H.; Yoshida, R.; Hirosue, A.; Nagata, M.; Tanaka, T.; Kawahara, K.; Sakata, J.; Arita, H.; Nakashima, H.; et al. IL-6 Controls Resistance to Radiation by Suppressing Oxidative Stress via the Nrf2-Antioxidant Pathway in Oral Squamous Cell Carcinoma. Br. J. Cancer 2016, 115, 1234–1244. [Google Scholar] [CrossRef] [Green Version]
  89. De Schutter, H.; Landuyt, W.; Verbeken, E.; Goethals, L.; Hermans, R.; Nuyts, S. The Prognostic Value of the Hypoxia Markers CA IX and GLUT I and the Cytokines VEGF and IL 6 in Head and Neck Squamous Cell Carcinoma Treated by Radiotherapy ± Chemotherapy. BMC Cancer 2005, 5, 42. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  90. Ara, T.; DeClerck, Y.A. Interleukin-6 in Bone Metastasis and Cancer Progression. Eur. J. Cancer 2010, 46, 1223–1231. [Google Scholar] [CrossRef] [Green Version]
  91. Duffy, S.A.; Taylor, J.M.G.; Terrell, J.E.; Islam, M.; Li, Y.; Fowler, K.E.; Wolf, G.T.; Teknos, T.N. Interleukin-6 Predicts Recurrence and Survival among Head and Neck Cancer Patients. Cancer 2008, 113, 750–757. [Google Scholar] [CrossRef]
  92. Brailo, V.; Vucicevic-Boras, V.; Lukac, J.; Biocina-Lukenda, D.; Zilic-Alajbeg, I.; Milenovic, A.; Balija, M. Salivary and Serum Interleukin 1 Beta, Interleukin 6 and Tumor Necrosis Factor Alpha in Patients with Leukoplakia and Oral Cancer. Med. Oral Patol. Oral Cir. Bucal 2012, 17, e10. [Google Scholar] [CrossRef]
  93. Zhang, S.; Zhang, X.; Yin, K.E.; Li, T.; Bao, Y.; Chen, Z. Variation and Significance of Secretory Immunoglobulin A, Interleukin 6 and Dendritic Cells in Oral Cancer. Oncol. Lett. 2017, 13, 2297–2303. [Google Scholar] [CrossRef] [Green Version]
  94. Lotfi, A.; Shahidi, N.; Bayazian, G.; Abdollahi Fakhim, S.; Estakhri, R.; Esfahani, A.; Notash, R. Serum Level of Interleukin-6 in Patients with Oral Tongue Squamous Cell Carcinoma. Iran. J. Otorhinolaryngol. 2015, 27, 207–211. [Google Scholar] [CrossRef]
  95. Garbers, C.; Hermanns, H.M.; Schaper, F.; Müller-Newen, G.; Grötzinger, J.; Rose-John, S.; Scheller, J. Plasticity and Cross-Talk of Interleukin 6-Type Cytokines. Cytokine Growth Factor Rev. 2012, 23, 85–97. [Google Scholar] [CrossRef]
  96. Riedel, F.; Zaiss, I.; Herzog, D.; Götte, K.; Naim, R.; Hörmann, K. Serum Levels of Interleukin-6 in Patients with Primary Head and Neck Squamous Cell Carcinoma. Anticancer Res. 2005, 25, 2761–2765. [Google Scholar]
  97. Andersson, B.-Å.; Lewin, F.; Lundgren, J.; Nilsson, M.; Rutqvist, L.-E.; Löfgren, S.; Laytragoon-Lewin, N. Plasma Tumor Necrosis Factor-α and C-Reactive Protein as Biomarker for Survival in Head and Neck Squamous Cell Carcinoma. J. Cancer Res. Clin. Oncol. 2014, 140, 515–519. [Google Scholar] [CrossRef] [PubMed]
  98. Chang, K.P.; Kao, H.K.; Wu, C.C.; Fang, K.H.; Chang, Y.L.; Huang, Y.C.; Liu, S.C.; Cheng, M.H. Pretreatment Interleukin-6 Serum Levels Are Associated with Patient Survival for Oral Cavity Squamous Cell Carcinoma. Otolaryngol.-Head Neck Surg. 2013, 148, 786–791. [Google Scholar] [CrossRef]
  99. Chen, C.C.; Chen, W.C.; Lu, C.H.; Wang, W.H.; Lin, P.Y.; der Lee, K.; Chen, M.F. Significance of Interleukin-6 Signaling in the Resistance of Pharyngeal Cancer to Irradiation and the Epidermal Growth Factor Receptor Inhibitor. Int. J. Radiat. Oncol. Biol. Phys. 2010, 76, 1214–1224. [Google Scholar] [CrossRef] [PubMed]
  100. Shinagawa, K.; Yanamoto, S.; Naruse, T.; Kawakita, A.; Morishita, K.; Sakamoto, Y.; Rokutanda, S.; Umeda, M. Clinical Roles of Interleukin-6 and STAT3 in Oral Squamous Cell Carcinoma. Pathol. Oncol. Res. 2017, 23, 425–431. [Google Scholar] [CrossRef] [PubMed]
  101. Chen, Z.; Yan, B.; van Waes, C. Role of the NF-ΚB Transcriptome and Proteome as Biomarkers Human Head and Neck Squamous Cell Carcinomas. Biomark. Med. 2008, 2, 409–429. [Google Scholar] [CrossRef] [Green Version]
  102. Hao, W.; Zhu, Y.; Zhou, H. Prognostic Value of Interleukin-6 and Interleukin-8 in Laryngeal Squamous Cell Cancer. Med. Oncol. 2013, 30, 333. [Google Scholar] [CrossRef]
  103. Chen, C.J.; Sung, W.W.; Lin, Y.M.; Chen, M.K.; Lee, C.H.; Lee, H.; Yeh, K.T.; Ko, J.L. Gender Difference in the Prognostic Role of Interleukin 6 in Oral Squamous Cell Carcinoma. PLoS ONE 2012, 7, e50104. [Google Scholar] [CrossRef]
  104. Schiegnitz, E.; Kämmerer, P.W.; Schön, H.; Blatt, S.; Berres, M.; Sagheb, K.; Al-Nawas, B. Proinflammatory Cytokines as Serum Biomarker in Oral Carcinoma—A Prospective Multi-Biomarker Approach. J. Oral Pathol. Med. 2018, 47, 268–274. [Google Scholar] [CrossRef]
  105. Li, C.; Zhao, Y.; Zhang, W.; Zhang, W. Increased Prevalence of TH17 Cells in the Peripheral Blood of Patients with Head and Neck Squamous Cell Carcinoma. Oral Surg. Oral Med. Oral Pathol. Oral Radiol. Endodontol. 2011, 112, 81–89. [Google Scholar] [CrossRef]
  106. Sparano, A.; Lathers, D.M.; Achille, N.; Petruzzelli, G.J.; Young, M.R. Modulation of Th1 and Th2 Cytokine Profiles and Their Association with Advanced Head and Neck Squamous Cell Carcinoma. Otolaryngol.-Head Neck Surg. Off. J. Am. Acad. Otolaryngol.-Head Neck Surg. 2004, 131, 573–576. [Google Scholar] [CrossRef] [PubMed]
  107. Kumar, B.; Brown, N.V.; Swanson, B.J.; Schmitt, A.C.; Old, M.; Ozer, E.; Agrawal, A.; Schuller, D.E.; Teknos, T.N.; Kumar, P. High Expression of Myoferlin Is Associated with Poor Outcome in Oropharyngeal Squamous Cell Carcinoma Patients and Is Inversely Associated with HPV-Status. Oncotarget 2016, 7, 18665. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  108. Kumar, B.; Yadav, A.; Brown, N.V.; Zhao, S.; Cipolla, M.J.; Wakely, P.E.; Schmitt, A.C.; Baiocchi, R.A.; Teknos, T.N.; Old, M.; et al. Nuclear PRMT5, Cyclin D1 and IL-6 Are Associated with Poor Outcome in Oropharyngeal Squamous Cell Carcinoma Patients and Is Inversely Associated with P16-Status. Oncotarget 2017, 8, 14847. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  109. St. John, M.A.R.; Li, Y.; Zhou, X.; Denny, P.; Ho, C.M.; Montemagno, C.; Shi, W.; Qi, F.; Wu, B.; Sinha, U.; et al. Interleukin 6 and Interleukin 8 as Potential Biomarkers for Oral Cavity and Oropharyngeal Squamous Cell Carcinoma. Arch. Otolaryngol.-Head Neck Surg. 2004, 130, 929–935. [Google Scholar] [CrossRef] [Green Version]
  110. Duffy, S.A.; Teknos, T.; Taylor, J.M.G.; Fowler, K.E.; Islam, M.; Wolf, G.T.; McLean, S.; Ghanem, T.A.; Terrell, J.E. Health Behaviors Predict Higher Interleukin-6 Levels Among Patients Newly Diagnosed with Head and Neck Squamous Cell Carcinoma. Cancer Epidemiol. Biomark. Prev. A Publ. Am. Assoc. Cancer Res. Cosponsored Am. Soc. Prev. Oncol. 2013, 22, 374. [Google Scholar] [CrossRef] [Green Version]
  111. Meleti, M.; Cassi, D.; Vescovi, P.; Setti, G.; Pertinhez, T.A.; Pezzi, M.E. Salivary Biomarkers for Diagnosis of Systemic Diseases and Malignant Tumors. A Systematic Review. Med. Oral Patol. Oral Cir. Bucal 2020, 25, e299. [Google Scholar] [CrossRef] [PubMed]
  112. Setti, G.; Pezzi, M.E.; Viani, M.V.; Pertinhez, T.A.; Cassi, D.; Magnoni, C.; Bellini, P.; Musolino, A.; Vescovi, P.; Meleti, M. Salivary MicroRNA for Diagnosis of Cancer and Systemic Diseases: A Systematic Review. Int. J. Mol. Sci. 2020, 21, 907. [Google Scholar] [CrossRef] [Green Version]
  113. Meleti, M.; Quartieri, E.; Antonelli, R.; Pezzi, M.E.; Ghezzi, B.; Viani, M.V.; Setti, G.; Casali, E.; Ferrari, E.; Ciociola, T.; et al. Metabolic Profiles of Whole, Parotid and Submandibular/Sublingual Saliva. Metabolites 2020, 10, 318. [Google Scholar] [CrossRef] [PubMed]
  114. Ferrari, E.; Pezzi, M.E.; Cassi, D.; Pertinhez, T.A.; Spisni, A.; Meleti, M. Salivary Cytokines as Biomarkers for Oral Squamous Cell Carcinoma: A Systematic Review. Int. J. Mol. Sci. 2021, 22, 6795. [Google Scholar] [CrossRef]
  115. Cristaldi, M.; Mauceri, R.; di Fede, O.; Giuliana, G.; Campisi, G.; Panzarella, V. Salivary Biomarkers for Oral Squamous Cell Carcinoma Diagnosis and Follow-Up: Current Status and Perspectives. Front. Physiol. 2019, 10, 1476. [Google Scholar] [CrossRef] [PubMed]
  116. Dineshkumar, T.; Ashwini, B.K.; Rameshkumar, A.; Rajashree, P.; Ramya, R.; Rajkumar, K. Salivary and Serum Interleukin-6 Levels in Oral Premalignant Disorders and Squamous Cell Carcinoma: Diagnostic Value and Clinicopathologic Correlations. Asian Pac. J. Cancer Prev. APJCP 2016, 17, 4899. [Google Scholar] [CrossRef]
  117. Katakura, A.; Kamiyama, I.; Takano, N.; Shibahara, T.; Muramatsu, T.; Ishihara, K.; Takagi, R.; Shouno, T. Comparison of Salivary Cytokine Levels in Oral Cancer Patients and Healthy Subjects. Bull. Tokyo Dent. Coll. 2007, 48, 199–203. [Google Scholar] [CrossRef] [Green Version]
  118. Rhodus, N.L.; Ho, V.; Miller, C.S.; Myers, S.; Ondrey, F. NF-ΚB Dependent Cytokine Levels in Saliva of Patients with Oral Preneoplastic Lesions and Oral Squamous Cell Carcinoma. Cancer Detect. Prev. 2005, 29, 42–45. [Google Scholar] [CrossRef]
  119. Juretić, M.; Cerović, R.; Belušić-Gobić, M.; Brekalo Pršo, I.; Kqiku, L.; Špalj, S.; Pezelj-Ribarić, S. Salivary Levels of TNF-α and IL-6 in Patients with Oral Premalignant and Malignant Lesions. Folia Biol. 2014, 59, 99–102. [Google Scholar]
  120. Korostoff, A.; Reder, L.; Masood, R.; Sinha, U.K. The Role of Salivary Cytokine Biomarkers in Tongue Cancer Invasion and Mortality. Oral Oncol. 2011, 47, 282–287. [Google Scholar] [CrossRef]
  121. Hamad, A.; Gaphor, S.; Shawagfeh, M.T.; Al-Talabani, N. Study of Serum and Salivary Levels of Proinflammatory Cytokines, Potential Biomarkers in the Diagnosis of Oral Squamous Cell Carcinoma. Acad. J. Cancer Res. 2011, 4, 47–55. [Google Scholar]
  122. Sahebjamee, M.; Eslami, M.; Atarbashimoghadam, F.; Sarafnejad, A. Salivary Concentration of TNFα, IL1α, IL6, and IL8 in Oral Squamous Cell Carcinoma. Med. Oral Patol. Oral Cir. Bucal 2008, 13, E292–E295. [Google Scholar] [PubMed]
  123. Cheng, Y.-S.L.; Jordan, L.; Gorugantula, L.M.; Schneiderman, E.; Chen, H.-S.; Rees, T. Salivary Interleukin-6 and -8 in Patients With Oral Cancer and Patients With Chronic Oral Inflammatory Diseases. J. Periodontol. 2014, 85, 956–965. [Google Scholar] [CrossRef] [PubMed]
  124. Selvam, N.P.; Sadaksharam, J. Salivary Interleukin-6 in the Detection of Oral Cancer and Precancer. Asia-Pac. J. Clin. Oncol. 2015, 11, 236–241. [Google Scholar] [CrossRef]
  125. Dikova, V.; Jantus-Lewintre, E.; Bagan, J. Potential Non-Invasive Biomarkers for Early Diagnosis of Oral Squamous Cell Carcinoma. J. Clin. Med. 2021, 10, 1658. [Google Scholar] [CrossRef] [PubMed]
  126. Van der Waal, I. Potentially Malignant Disorders of the Oral and Oropharyngeal Mucosa; Terminology, Classification and Present Concepts of Management. Oral Oncol. 2009, 45, 317–323. [Google Scholar] [CrossRef]
  127. Sato, J.; Goto, J.; Murata, T.; Kitamori, S.; Yamazaki, Y.; Satoh, A.; Kitagawa, Y. Changes in Saliva Interleukin-6 Levels in Patients with Oral Squamous Cell Carcinoma. Oral Surg. Oral Med. Oral Pathol. Oral Radiol. Endod. 2010, 110, 330–336. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  128. Sato, J.; Ohuchi, M.; Abe, K.; Satoh, T.; Abe, T.; Yamazaki, Y.; Satoh, A.; Notani, K.; Kitagawa, Y. Correlation between Salivary Interleukin-6 Levels and Early Locoregional Recurrence in Patients with Oral Squamous Cell Carcinoma: Preliminary Study. Head Neck 2013, 35, 889–894. [Google Scholar] [CrossRef]
  129. Sato, J.; Ohuchi, M.; Wada, M.; Ohga, N.; Asaka, T.; Yoshikawa, K.; Miyakoshi, M.; Hata, H.; Satoh, A.; Kitagawa, Y. Differences in Sequential Posttreatment Salivary IL-6 Levels between Patients with and Patients without Locoregional Recurrences of Oral Squamous Cell Carcinoma: Part III of a Cohort Study. Oral Surg. Oral Med. Oral Pathol. Oral Radiol. 2015, 120, 751–760.e2. [Google Scholar] [CrossRef]
  130. Fan, N.; Luo, Y.; Ou, Y.; He, H. Altered Serum Levels of TNF-α, IL-6, and IL-18 in Depressive Disorder Patients. Hum. Psychopharmacol. 2017, 32, e2588. [Google Scholar] [CrossRef]
  131. Patel, H.J.; Patel, B.M. TNF-α and Cancer Cachexia: Molecular Insights and Clinical Implications. Life Sci. 2017, 170, 56–63. [Google Scholar] [CrossRef] [PubMed]
  132. Smetana, K., Jr.; Dvořánková, B.; Lacina, L.; Strnad, H.; Kolář, M.; Chovanec, M.; Plzák, J.; Čada, Z.; Vlček, Č.; Szabo, P.; et al. Combination of Antibodies or Fab Fragments Thereof for Use as Medicament, and Pharmaceutical Composition Containing Said Antibodies or Fab Fragments Thereof. Czech Patent No. B6 303227, 6 June 2012. (In Czech). [Google Scholar]
  133. Jayatilaka, H.; Tyle, P.; Chen, J.J.; Kwak, M.; Ju, J.; Kim, H.J.; Lee, J.S.H.; Wu, P.H.; Gilkes, D.M.; Fan, R.; et al. Synergistic IL-6 and IL-8 Paracrine Signalling Pathway Infers a Strategy to Inhibit Tumour Cell Migration. Nat. Commun. 2017, 8, 1–12. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  134. Wirtz, D.G. and Yatilaka H. Cancer Cell Migration Inhibitors and Their Use in Therapeutic Treatments. U.S. Patent 2017/0165363 A1, 15 June 2017. [Google Scholar]
  135. Kodet, O.; Dvořánková, B.; Bendlová, B.; Sýkorová, V.; Krajsová, I.; Štork, J.; Kučera, J.; Szabo, P.; Strnad, H.; Kolář, M.; et al. Microenvironment-Driven Resistance to B-Raf Inhibition in a Melanoma Patient Is Accompanied by Broad Changes of Gene Methylation and Expression in Distal Fibroblasts. Int. J. Mol. Med. 2018, 41, 2687–2703. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  136. Loppnow, H.; Zhang, L.; Buerke, M.; Lautenschläger, M.; Chen, L.; Frister, A.; Schlitt, A.; Luther, T.; Song, N.; Hofmann, B.; et al. Statins Potently Reduce the Cytokine-Mediated IL-6 Release in SMC/MNC Cocultures. J. Cell. Mol. Med. 2011, 15, 994. [Google Scholar] [CrossRef] [PubMed]
  137. Awasthi, S.; Wagner, T.; Venkatakrishnan, A.J.; Puranik, A.; Hurchik, M.; Agarwal, V.; Conrad, I.; Kirkup, C.; Arunachalam, R.; O’Horo, J.; et al. Plasma IL-6 Levels Following Corticosteroid Therapy as an Indicator of ICU Length of Stay in Critically Ill COVID-19 Patients. Cell Death Discov. 2021, 7, 1–15. [Google Scholar] [CrossRef] [PubMed]
  138. Jain, M.K.; Ridker, P.M. Anti-Inflammatory Effects of Statins: Clinical Evidence and Basic Mechanisms. Nat. Rev. Drug Discov. 2005, 4, 977–987. [Google Scholar] [CrossRef]
  139. Smetana, K.; Smetana, K.; Brábek, J.; Brábek, J. Role of Interleukin-6 in Lung Complications in Patients with COVID-19: Therapeutic Implications. In Vivo 2020, 34, 1589–1592. [Google Scholar] [CrossRef]
  140. Atal, S.; Fatima, Z. IL-6 Inhibitors in the Treatment of Serious COVID-19: A Promising Therapy? Pharm. Med. 2020, 34, 223–231. [Google Scholar] [CrossRef] [PubMed]
  141. Gould, S.; Norris, S.L. Contested Effects and Chaotic Policies: The 2020 Story of (Hydroxy) Chloroquine for Treating COVID-19. Cochrane Database Syst. Rev. 2021, 3, ED000151. [Google Scholar] [CrossRef]
  142. Wozniacka, A.; Lesiak, A.; Narbutt, J.; McCauliffe, D.P.; Sysa-Jedrzejowska, A. Chloroquine Treatment Influences Proinflammatory Cytokine Levels in Systemic Lupus Erythematosus Patients. Lupus 2006, 15, 268–275. [Google Scholar] [CrossRef]
  143. Van den Borne, B.E.; Dijkmans, B.A.; de Rooij, H.H.; le Cessie, S.; Verweij, C.L. Chloroquine and Hydroxychloroquine Equally Affect Tumor Necrosis Factor-Alpha, Interleukin 6, and Interferon-Gamma Production by Peripheral Blood Mononuclear Cells. J. Rheumatol. 1997, 24, 55–60. [Google Scholar]
  144. Mantovani, A.; Allavena, P.; Sica, A.; Balkwill, F. Cancer-Related Inflammation. Nature 2008, 454, 436–444. [Google Scholar] [CrossRef]
  145. Varisli, L.; Cen, O.; Vlahopoulos, S. Dissecting Pharmacological Effects of Chloroquine in Cancer Treatment: Interference with Inflammatory Signaling Pathways. Immunology 2020, 159, 257–278. [Google Scholar] [CrossRef] [Green Version]
  146. Bryant, J.; Batis, N.; Franke, A.C.; Clancey, G.; Hartley, M.; Ryan, G.; Brooks, J.; Southam, A.D.; Barnes, N.; Parish, J.; et al. Repurposed Quinacrine Synergizes with Cisplatin, Reducing the Effective Dose Required for Treatment of Head and Neck Squamous Cell Carcinoma. Oncotarget 2019, 10, 5229–5244. [Google Scholar] [CrossRef] [Green Version]
  147. Duarte, D.; Vale, N. New Trends for Antimalarial Drugs: Synergism between Antineoplastics and Antimalarials on Breast Cancer Cells. Biomolecules 2020, 10, 1623. [Google Scholar] [CrossRef] [PubMed]
  148. Muñoz-Cánoves, P.; Scheele, C.; Pedersen, B.K.; Serrano, A.L. Interleukin-6 Myokine Signaling in Skeletal Muscle: A Double-edged Sword? FEBS J. 2013, 280, 4131. [Google Scholar] [CrossRef]
  149. Kawano, M.; Hirano, T.; Matsuda, T.; Taga, T.; Horii, Y.; Iwato, K.; Asaoku, H.; Tang, B.; Tanabe, O.; Tanaka, H.; et al. Autocrine Generation and Requirement of BSF-2/IL-6 for Human Multiple Myelomas. Nature 1988, 332, 83–85. [Google Scholar] [CrossRef] [PubMed]
  150. Beck, J.T.; Hsu, S.-M.; Wijdenes, J.; Bataille, R.; Klein, B.; Vesole, D.; Hayden, K.; Jagannath, S.; Barlogie, B. Alleviation of Systemic Manifestations of Castleman’s Disease by Monoclonal Anti-Interleukin-6 Antibody. N. Engl. J. Med. 2010, 330, 602–605. [Google Scholar] [CrossRef]
  151. Van Rhee, F.; Fayad, L.; Voorhees, P.; Furman, R.; Lonial, S.; Borghaei, H.; Sokol, L.; Crawford, J.; Cornfeld, M.; Qi, M.; et al. Siltuximab, a Novel Anti-Interleukin-6 Monoclonal Antibody, for Castleman’s Disease. J. Clin. Oncol. 2010, 28. [Google Scholar] [CrossRef] [PubMed]
  152. Choy, E.H.; de Benedetti, F.; Takeuchi, T.; Hashizume, M.; John, M.R.; Kishimoto, T. Translating IL-6 Biology into Effective Treatments. Nat. Rev. Rheumatol. 2020, 16, 335–345. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  153. Doberer, K.; Duerr, M.; Halloran, P.F.; Eskandary, F.; Budde, K.; Regele, H.; Reeve, J.; Borski, A.; Kozakowski, N.; Reindl-Schwaighofer, R.; et al. A Randomized Clinical Trial of Anti⇓IL-6 Antibody Clazakizumab in Late Antibody-Mediated Kidney Transplant Rejection. J. Am. Soc. Nephrol. 2021, 32, 708–722. [Google Scholar] [CrossRef] [PubMed]
  154. Shaw, S.; Bourne, T.; Meier, C.; Carrington, B.; Gelinas, R.; Henry, A.; Popplewell, A.; Adams, R.; Baker, T.; Rapecki, S.; et al. Discovery and Characterization of Olokizumab: A Humanized Antibody Targeting Interleukin-6 and Neutralizing Gp130-Signaling. mAbs 2014, 6, 773–781. [Google Scholar] [CrossRef] [Green Version]
  155. Burger, R.; Günther, A.; Klausz, K.; Staudinger, M.; Peipp, M.; Penas, E.M.M.; Rose-John, S.; Wijdenes, J.; Gramatzki, M. Due to Interleukin-6 Type Cytokine Redundancy Only Glycoprotein 130 Receptor Blockade Efficiently Inhibits Myeloma Growth. Haematologica 2017, 102, 381. [Google Scholar] [CrossRef] [Green Version]
  156. Nishimoto, N.; Kishimoto, T. Humanized Antihuman IL-6 Receptor Antibody, Tocilizumab. Handb. Exp. Pharmacol. 2008, 181, 151–160. [Google Scholar] [CrossRef]
  157. Genovese, M.C.; McKay, J.D.; Nasonov, E.L.; Mysler, E.F.; da Silva, N.A.; Alecock, E.; Woodworth, T.; Gomez-Reino, J.J. Interleukin-6 Receptor Inhibition with Tocilizumab Reduces Disease Activity in Rheumatoid Arthritis with Inadequate Response to Disease-Modifying Antirheumatic Drugs: The Tocilizumab in Combination with Traditional Disease-Modifying Antirheumatic Drug Therapy Study. Arthritis Rheum. 2008, 58, 2968–2980. [Google Scholar] [CrossRef]
  158. Kim, N.; Kim, S.; Kim, D.; Zhang, D.; Park, J.; Yi, H.; Kim, J.; Shin, H. Anti-proliferative Action of IL-6R-Targeted Antibody Tocilizumab for Non-Small Cell Lung Cancer Cells. Oncol. Lett. 2015, 9, 2283–2288. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  159. Alraouji, N.N.; Al-Mohanna, F.H.; Ghebeh, H.; Arafah, M.; Almeer, R.; Al-Tweigeri, T.; Aboussekhra, A. Tocilizumab Potentiates Cisplatin Cytotoxicity and Targets Cancer Stem Cells in Triple-Negative Breast Cancer. Mol. Carcinog. 2020, 59, 1041–1051. [Google Scholar] [CrossRef] [PubMed]
  160. Varra, V.; Smile, T.D.; Geiger, J.L.; Koyfman, S.A. Recent and Emerging Therapies for Cutaneous Squamous Cell Carcinomas of the Head and Neck. Curr. Treat. Options Oncol. 2020, 21, 1–12. [Google Scholar] [CrossRef]
  161. Migden, M.R.; Rischin, D.; Schmults, C.D.; Guminski, A.; Hauschild, A.; Lewis, K.D.; Chung, C.H.; Hernandez-Aya, L.; Lim, A.M.; Chang, A.L.S.; et al. PD-1 Blockade with Cemiplimab in Advanced Cutaneous Squamous-Cell Carcinoma. N. Engl. J. Med. 2018, 379, 341–351. [Google Scholar] [CrossRef] [Green Version]
  162. Tsukamoto, H.; Fujieda, K.; Miyashita, A.; Fukushima, S.; Ikeda, T.; Kubo, Y.; Senju, S.; Ihn, H.; Nishimura, Y.; Oshiumi, H. Combined Blockade of IL6 and PD-1/PD-L1 Signaling Abrogates Mutual Regulation of Their Immunosuppressive Effects in the Tumor Microenvironment. Cancer Res. 2018, 78, 5011–5022. [Google Scholar] [CrossRef] [Green Version]
  163. Boyce, E.G.; Rogan, E.L.; Vyas, D.; Prasad, N.; Mai, Y. Sarilumab: Review of a Second IL-6 Receptor Antagonist Indicated for the Treatment of Rheumatoid Arthritis. Ann. Pharmacother. 2018, 52, 780–791. [Google Scholar] [CrossRef]
  164. Heo, Y.A. Satralizumab: First Approval. Drugs 2020, 80, 1477–1482. [Google Scholar] [CrossRef] [PubMed]
  165. Glicklich, A.; Grayson, P.; Blanchetot, C.; Zhou, Q.; Kretz-Rommel, A. The Development of a New Anti–Interleukin 6 Blocker for Rheumatoid Arthritis Patients-ACR Meeting Abstracts. In Arthritis & Rheumatology; Wiley: Hoboken, NJ, USA, 2016; Volume 68. [Google Scholar]
  166. Rinaldi, M.; van Bogaert, T.; van Roy, M.; Bontinck, L.; Hohlbaum, A.; Snoeck, V.; Dombrecht, E.; van Beneden, K.; Schoen, P.; Ulrichts, H. Assessment of Dose Dependent Effects of Vobarilizumab, an Anti-IL6 Receptor (IL-6R) Nanobody®, on Systemic Biomarkers in Patients with Moderate-to-Severe Rheumatoid Arthritis (RA): Results of Two Phase 2b Studies-ACR Meeting Abstracts. Arthritis Rheumatol. 2017, 69, 2476. [Google Scholar]
  167. Chevalier, S.; Fourcin, M.; Robledo, O.; Wijdenes, J.; Pouplard-Barthelaix, A.; Gascan, H. Interleukin-6 Family of Cytokines Induced Activation of Different Functional Sites Expressed by Gp130 Transducing Protein. J. Biol. Chem. 1996, 271. [Google Scholar] [CrossRef] [Green Version]
  168. Yamamoto, D.; Sunazuka, T.; Hirose, T.; Kojima, N.; Kaji, E.; Omura, S. Design, Synthesis, and Biological Activities of Madindoline Analogues. Bioorg. Med. Chem. Lett. 2006, 16, 2807–2811. [Google Scholar] [CrossRef] [PubMed]
  169. Xu, S.; Grande, F.; Garofalo, A.; Neamati, N. Discovery of a Novel Orally Active Small-Molecule Gp130 Inhibitor for the Treatment of Ovarian Cancer. Mol. Cancer Ther. 2013, 12, 937–949. [Google Scholar] [CrossRef] [Green Version]
  170. Hong, S.-S.; Choi, J.H.; Lee, S.Y.; Park, Y.-H.; Park, K.-Y.; Lee, J.Y.; Kim, J.; Gajulapati, V.; Goo, J.-I.; Singh, S.; et al. A Novel Small-Molecule Inhibitor Targeting the IL-6 Receptor β Subunit, Glycoprotein 130. J. Immunol. 2015, 195, 237–245. [Google Scholar] [CrossRef] [Green Version]
  171. Li, H.; Xiao, H.; Lin, L.; Jou, D.; Kumari, V.; Lin, J.; Li, C. Drug Design Targeting Protein-Protein Interactions (PPIs) Using Multiple Ligand Simultaneous Docking (MLSD) and Drug Repositioning: Discovery of Raloxifene and Bazedoxifene as Novel Inhibitors of IL-6/GP130 Interface. J. Med. Chem. 2014, 57, 632–641. [Google Scholar] [CrossRef]
  172. Thilakasiri, P.; Huynh, J.; Poh, A.R.; Tan, C.W.; Nero, T.L.; Tran, K.; Parslow, A.C.; Afshar-Sterle, S.; Baloyan, D.; Hannan, N.J.; et al. Repurposing the Selective Estrogen Receptor Modulator Bazedoxifene to Suppress Gastrointestinal Cancer Growth. EMBO Mol. Med. 2019, 11, e9539. [Google Scholar] [CrossRef]
  173. Heise, D.; Derrac Soria, A.; Hansen, S.; Dambietz, C.; Akbarzadeh, M.; Berg, A.F.; Waetzig, G.H.; Jones, S.A.; Dvorsky, R.; Ahmadian, M.R.; et al. Selective Inhibition of IL-6 Trans-Signaling by a Miniaturized, Optimized Chimeric Soluble Gp130 Inhibits T H 17 Cell Expansion. Sci. Signal. 2021, 14, eabc3480. [Google Scholar] [CrossRef]
  174. Schreiber, S.; Aden, K.; Bernardes, J.P.; Conrad, C.; Tran, F.; Höper, H.; Volk, V.; Mishra, N.; Blase, J.I.; Nikolaus, S.; et al. Therapeutic Interleukin-6 Trans-Signaling Inhibition by Olamkicept (Sgp130Fc) in Patients With Active Inflammatory Bowel Disease. Gastroenterology 2021, 160, 2354–2366.e11. [Google Scholar] [CrossRef]
  175. Uz, U.; Eskiizmir, G. Association between Interleukin-6 and Head and Neck Squamous Cell Carcinoma: A Systematic Review. Clin. Exp. Otorhinolaryngol. 2021, 14, 50–60. [Google Scholar] [CrossRef] [PubMed]
  176. Cui, N.; Nomura, T.; Noma, H.; Yokoo, K.; Takagi, R.; Hashimoto, S.; Okamoto, M.; Sato, M.; Yu, G.; Guo, C.; et al. Effect of YM529 on a Model of Mandibular Invasion by Oral Squamous Cell Carcinoma in Mice. Clin. Cancer Res. 2005, 11, 2713–2719. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  177. Wichmann, G.; Cedra, S.; Schlegel, D.; Kolb, M.; Wiegand, S.; Boehm, A.; Hofer, M.; Dietz, A. Cilengitide and Cetuximab Reduce Cytokine Production and Colony Formation of Head and Neck Squamous Cell Carcinoma Cells Ex Vivo. Anticancer Res. 2017, 37, 521–527. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  178. Wolf, J.S.; Li, G.; Varadhachary, A.; Petrak, K.; Schneyer, M.; Li, D.; Ongkasuwan, J.; Zhang, X.; Taylor, R.J.; Strome, S.E.; et al. Oral Lactoferrin Results in T Cell-Dependent Tumor Inhibition of Head and Neck Squamous Cell Carcinoma in Vivo. Clin. Cancer Res. 2007, 13, 1601–1610. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  179. Tu, D.G.; Lin, W.T.; Yu, C.C.; Lee, S.S.; Peng, C.Y.; Lin, T.; Yu, C.H. Chemotherapeutic Effects of Luteolin on Radio-Sensitivity Enhancement and Interleukin-6/Signal Transducer and Activator of Transcription 3 Signaling Repression of Oral Cancer Stem Cells. J. Formos. Med. Assoc. 2016, 115, 1032–1038. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  180. Tamatani, T.; Azuma, M.; Motegi, K.; Takamaru, N.; Kawashima, Y.; Bando, T. Cepharanthin-Enhanced Radiosensitivity through the Inhibition of Radiation-Induced Nuclear Factor-ΚB Activity in Human Oral Squamous Cell Carcinoma Cells. Int. J. Oncol. 2007, 31, 761–768. [Google Scholar] [CrossRef] [PubMed]
  181. Macha, M.A.; Matta, A.; Chauhan, S.S.; Michael Siu, K.W.; Ralhan, R. Guggulsterone (GS) Inhibits Smokeless Tobacco and Nicotine-Induced NF-ΚB and STAT3 Pathways in Head and Neck Cancer Cells. Carcinogenesis 2011, 32, 368–380. [Google Scholar] [CrossRef] [Green Version]
  182. Lin, H.Y.; Hou, S.C.; Chen, S.C.; Kao, M.C.; Yu, C.C.; Funayama, S.; Ho, C.T.; der Way, T. (-)-Epigallocatechin Gallate Induces Fas/CD95-Mediated Apoptosis through Inhibiting Constitutive and IL-6-Induced JAK/STAT3 Signaling in Head and Neck Squamous Cell Carcinoma Cells. J. Agric. Food Chem. 2012, 60, 2480–2489. [Google Scholar] [CrossRef]
  183. Cohen, A.N.; Veena, M.S.; Srivatsan, E.S.; Wang, M.B. Suppression of Interleukin 6 and 8 Production in Head and Neck Cancer Cells with Curcumin via Inhibition of Iκβ Kinase. Arch. Otolaryngol.-Head Neck Surg. 2009, 135, 190–197. [Google Scholar] [CrossRef] [Green Version]
  184. Chakravarti, N.; Myers, J.N.; Aggarwal, B.B. Targeting Constitutive and Interleukin-6-Inducible Signal Transducers and Activators of Transcription 3 Pathway in Head and Neck Squamous Cell Carcinoma Cells by Curcumin (Diferuloylmethane). Int. J. Cancer 2006, 119, 1268–1275. [Google Scholar] [CrossRef]
  185. DiNatale, B.C.; Schroeder, J.C.; Perdew, G.H. Ah Receptor Antagonism Inhibits Constitutive and Cytokine Inducible IL6 Production in Head and Neck Tumor Cell Lines. Mol. Carcinog. 2011, 50, 173–183. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  186. DiNatale, B.C.; Smith, K.; John, K.; Krishnegowda, G.; Amin, S.G.; Perdew, G.H. Ah Receptor Antagonism Represses Head and Neck Tumor Cell Aggressive Phenotype. Mol. Cancer Res. 2012, 10, 1369–1379. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  187. Tang, C.H.; Chuang, J.Y.; Fong, Y.C.; Maa, M.C.; der Way, T.; Hung, C.H. Bone-Derived SDF-1 Stimulates IL-6 Release via CXCR4, ERK and NF-ΚB Pathways and Promotes Osteoclastogenesis in Human Oral Cancer Cells. Carcinogenesis 2008, 29, 1483–1492. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  188. Siavash, H.; Nikitakis, N.G.; Sauk, J.J. Abrogation of IL-6-Mediated JAK Signalling by the Cyclopentenone Prostaglandin 15d-PGJ2 in Oral Squamous Carcinoma Cells. Br. J. Cancer 2004, 91, 1074–1080. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  189. Poth, K.J.; Guminski, A.D.; Thomas, G.P.; Leo, P.J.; Jabbar, I.A.; Saunders, N.A. Cisplatin Treatment Induces a Transient Increase in Tumorigenic Potential Associated with High Interleukin-6 Expression in Head and Neck Squamous Cell Carcinoma. Mol. Cancer Ther. 2010, 9, 2430–2439. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  190. Stanam, A.; Love-Homan, L.; Joseph, T.S.; Espinosa-Cotton, M.; Simons, A.L. Upregulated Interleukin-6 Expression Contributes to Erlotinib Resistance in Head and Neck Squamous Cell Carcinoma. Mol. Oncol. 2015, 9, 1371–1383. [Google Scholar] [CrossRef] [Green Version]
  191. Shinriki, S.; Jono, H.; Ueda, M.; Ota, K.; Ota, T.; Sueyoshi, T.; Oike, Y.; Ibusuki, M.; Hiraki, A.; Nakayama, H.; et al. Interleukin-6 Signalling Regulates Vascular Endothelial Growth Factor-C Synthesis and Lymphangiogenesis in Human Oral Squamous Cell Carcinoma. J. Pathol. 2011, 225, 142–150. [Google Scholar] [CrossRef] [PubMed]
  192. Shinriki, S.; Jono, H.; Ota, K.; Ueda, M.; Kudo, M.; Ota, T.; Oike, Y.; Endo, M.; Ibusuki, M.; Hiraki, A.; et al. Humanized Anti-Interleukin-6 Receptor Antibody Suppresses Tumor Angiogenesis and in Vivo Growth of Human Oral Squamous Cell Carcinoma. Clin. Cancer Res. 2009, 15, 5426–5434. [Google Scholar] [CrossRef] [Green Version]
  193. Islam, M.; Sharma, S.; Teknos, T.N. RhoC Regulates Cancer Stem Cells in Head and Neck Squamous Cell Carcinoma by Overexpressing IL-6 and Phosphorylation of STAT3. PLoS ONE 2014, 9, 88527. [Google Scholar] [CrossRef]
  194. Hwang, Y.S.; Ahn, S.Y.; Moon, S.; Zheng, Z.; Cha, I.H.; Kim, J.; Zhang, X. Insulin-like Growth Factor-II MRNA Binding Protein-3 and Podoplanin Expression Are Associated with Bone Invasion and Prognosis in Oral Squamous Cell Carcinoma. Arch. Oral Biol. 2016, 69, 25–32. [Google Scholar] [CrossRef]
  195. Finkel, K.A.; Warner, K.A.; Kerk, S.; Bradford, C.R.; McLean, S.A.; Prince, M.E.; Zhong, H.; Hurt, E.M.; Hollingsworth, R.E.; Wicha, M.S.; et al. IL-6 Inhibition With MEDI5117 Decreases The Fraction of Head and Neck Cancer Stem Cells and Prevents Tumor Recurrence. Neoplasia 2016, 18, 273–281. [Google Scholar] [CrossRef] [Green Version]
  196. Ekshyyan, O.; Khandelwal, A.R.; Rong, X.; Moore-Medlin, T.; Ma, X.; Alexander, J.S.; Nathan, C.A.O. Rapamycin Targets Interleukin 6 (IL-6) Expression and Suppresses Endothelial Cell Invasion Stimulated by Tumor Cells. Am. J. Transl. Res. 2016, 8, 4822–4830. [Google Scholar]
  197. Riebe, C.; Pries, R.; Kemkers, A.; Wollenberg, B. Increased Cytokine Secretion in Head and Neck Cancer upon P38 Mitogen-Activated Protein Kinase Activation. Int. J. Mol. Med. 2007, 20, 883–887. [Google Scholar] [CrossRef] [Green Version]
  198. Jing, Z.; Xu, H.; Chen, X.; Zhong, Q.; Huang, J.; Zhang, Y.; Guo, W.; Yang, Z.; Ding, S.; Chen, P.; et al. The Proton-Sensing G-Protein Coupled Receptor GPR4 Promotes Angiogenesis in Head and Neck Cancer. PLoS ONE 2016, 11, e0152789. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  199. Kross, K.W.; Heimdal, J.H.; Olsnes, C.; Olofson, J.; Aarstad, H.J. Tumour-Associated Macrophages Secrete IL-6 and MCP-1 in Head and Neck Squamous Cell Carcinoma Tissue. Acta Oto-Laryngol. 2007, 127, 532–539. [Google Scholar] [CrossRef] [PubMed]
  200. Mishra, P.B.; Lobo, A.S.; Joshi, K.S.; Rathos, M.J.; Kumar, G.A.; Padigaru, M. Molecular Mechanisms of Anti-Tumor Properties of P276-00 in Head and Neck Squamous Cell Carcinoma. J. Transl. Med. 2013, 11, 1–11. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  201. Sen, M.; Johnston, P.A.; Pollock, N.I.; DeGrave, K.; Joyce, S.C.; Freilino, M.L.; Hua, Y.; Camarco, D.P.; Close, D.A.; Huryn, D.M.; et al. Mechanism of Action of Selective Inhibitors of IL-6 Induced STAT3 Pathway in Head and Neck Cancer Cell Lines. J. Chem. Biol. 2017, 10, 129–141. [Google Scholar] [CrossRef] [Green Version]
  202. Teknos, T.N.; Islam, M.; Arenberg, D.A.; Pan, Q.; Carskadon, S.L.; Abarbanell, A.M.; Marcus, B.; Paul, S.; Vandenberg, C.D.; Carron, M.; et al. The Effect of Tetrathiomolybdate on Cytokine Expression, Angiogenesis, and Tumor Growth in Squamous Cell Carcinoma of the Head and Neck. Arch. Otolaryngol.-Head Neck Surg. 2005, 131, 204–211. [Google Scholar] [CrossRef] [Green Version]
  203. Van Tubergen, E.; vander Broek, R.; Lee, J.; Wolf, G.; Carey, T.; Bradford, C.; Prince, M.; Kirkwood, K.L.; D’Silva, N.J. Tristetraprolin Regulates Interleukin-6, Which Is Correlated with Tumor Progression in Patients with Head and Neck Squamous Cell Carcinoma. Cancer 2011, 117, 2677–2689. [Google Scholar] [CrossRef]
  204. Van Tubergen, E.A.; Banerjee, R.; Liu, M.; vander Broek, R.; Light, E.; Kuo, S.; Feinberg, S.E.; Willis, A.L.; Wolf, G.; Carey, T.; et al. Inactivation or Loss of TTP Promotes Invasion in Head and Neck Cancer via Transcript Stabilization and Secretion of MMP9, MMP2, and IL-6. Clin. Cancer Res. 2013, 19, 1169–1179. [Google Scholar] [CrossRef] [Green Version]
  205. Zhou, X.; Ren, Y.; Liu, A.; Han, L.; Zhang, K.; Li, S.; Li, P.; Li, P.; Kang, C.; Wang, X.; et al. STAT3 Inhibitor WP1066 Attenuates MiRNA-21 to Suppress Human Oral Squamous Cell Carcinoma Growth in Vitro and in Vivo. Oncol. Rep. 2014, 31, 2173–2180. [Google Scholar] [CrossRef] [Green Version]
  206. Te Chang, M.; Lee, S.P.; Fang, C.Y.; Hsieh, P.L.; Liao, Y.W.; Lu, M.Y.; Tsai, L.L.; Yu, C.C.; Liu, C.M. Chemosensitizing Effect of Honokiol in Oral Carcinoma Stem Cells via Regulation of IL-6/Stat3 Signaling. Environ. Toxicol. 2018, 33, 1105–1112. [Google Scholar] [CrossRef]
  207. Wang, T.H.; Fang, J.Y.; Wu, S.J.; Liu, Y.W.; Chan, C.W.; Chuang, S.Y.; Chen, C.Y. 2-O-Methylmagnolol Induces Apoptosis and Inhibits IL-6/STAT3 Signaling in Oral Squamous Cell Carcinoma. Cell. Physiol. Biochem. 2018, 50, 883–892. [Google Scholar] [CrossRef] [PubMed]
  208. Gehrke, T.; Scherzad, A.; Hackenberg, S.; Ickrath, P.; Schendzielorz, P.; Hagen, R.; Kleinsasser, N. Additive Antitumor Effects of Celecoxib and Simvastatin on Head and Neck Squamous Cell Carcinoma in Vitro. Int. J. Oncol. 2017, 51, 931–938. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  209. Yadav, A.; Kumar, B.; Teknos, T.N.; Kumar, P. Bazedoxifene Enhances the Anti-Tumor Effects of Cisplatin and Radiation Treatment by Blocking IL-6 Signaling in Head and Neck Cancer. Oncotarget 2017, 8, 66912. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  210. Meyer, C.; Pries, R.; Wollenberg, B. Established and Novel NF-ΚB Inhibitors Lead to Downregulation of TLR3 and the Proliferation and Cytokine Secretion in HNSCC. Oral Oncol. 2011, 47, 818–826. [Google Scholar] [CrossRef] [PubMed]
  211. Ruan, M.; Thorn, K.; Liu, S.; Li, S.; Yang, W.; Zhang, C.; Zhang, C. The Secretion of IL-6 by CpG-ODN-Treated Cancer Cells Promotes T-Cell Immune Responses Partly through the TLR-9/AP-1 Pathway in Oral Squamous Cell Carcinoma. Int. J. Oncol. 2014, 45, 2103–2110. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  212. A First-in-Humans Dose Finding Study for an Aryl Hydrocarbon Receptor Inhibitor (AhRi) in Patients With Advanced Cancer-Full Text View-ClinicalTrials.Gov. Available online: https://clinicaltrials.gov/ct2/show/study/NCT04069026?term=IL-6&cond=HNSCC&draw=2&rank=9 (accessed on 12 September 2021).
  213. Zafar, E.; Maqbool, M.F.; Iqbal, A.; Maryam, A.; Shakir, H.A.; Irfan, M.; Khan, M.; Li, Y.; Ma, T. A Comprehensive Review on Anticancer Mechanism of Bazedoxifene. Biotechnol. Appl. Biochem. 2021. [Google Scholar] [CrossRef] [PubMed]
  214. Erbitux (Cetuximab)-Summary of Product Characteristics. Available online: https://www.ema.europa.eu/en/documents/product-information/erbitux-epar-product-information_en.pdf (accessed on 10 October 2021).
  215. The Effect of Curcumin for Treatment of Cancer Anorexia-Cachexia Syndrome in Patients with Stage III-IV of Head and Neck Cancer-Full Text View-ClinicalTrials.Gov. Available online: https://clinicaltrials.gov/ct2/show/NCT04208334?cond=head+and+neck+cancer+curcumin&draw=2&rank=2 (accessed on 12 September 2021).
  216. Kotha, R.R.; Luthria, D.L. Curcumin: Biological, Pharmaceutical, Nutraceutical, and Analytical Aspects. Molecules 2019, 24, 2930. [Google Scholar] [CrossRef] [Green Version]
  217. Celebrex (Celecoxib) Information | FDA. Available online: https://www.fda.gov/drugs/postmarket-drug-safety-information-patients-and-providers/celebrex-celecoxib-information (accessed on 12 September 2021).
  218. Simvastatin Information | FDA. Available online: https://www.fda.gov/drugs/postmarket-drug-safety-information-patients-and-providers/simvastatin-information (accessed on 12 September 2021).
  219. Du, G.-J.; Zhang, Z.; Wen, X.-D.; Yu, C.; Calway, T.; Yuan, C.-S.; Wang, C.-Z. Epigallocatechin Gallate (EGCG) Is the Most Effective Cancer Chemopreventive Polyphenol in Green Tea. Nutrients 2012, 4, 1679. [Google Scholar] [CrossRef]
  220. 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.; et al. Guggulsterone Enhances Head and Neck Cancer Therapies via Inhibition of Signal Transducer and Activator of Transcription-3. Carcinogenesis 2009, 30, 1848. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  221. Rauf, A.; Olatunde, A.; Imran, M.; Alhumaydhi, F.A.; Aljohani, A.S.M.; Khan, S.A.; Uddin, M.S.; Mitra, S.; bin Emran, T.; Khayrullin, M.; et al. Honokiol: A Review of Its Pharmacological Potential and Therapeutic Insights. Phytomedicine 2021, 90, 153647. [Google Scholar] [CrossRef] [PubMed]
  222. Lin, Y.; Shi, R.; Wang, X.; Shen, H.-M. Luteolin, a Flavonoid with Potentials for Cancer Prevention and Therapy. Curr. Cancer Drug Targets 2008, 8, 634. [Google Scholar] [CrossRef]
  223. Rapamune-SPC. Available online: https://www.ema.europa.eu/en/documents/product-information/rapamune-epar-product-information_en.pdf (accessed on 19 September 2021).
  224. Angevin, E.; Tabernero, J.; Elez, E.; Cohen, S.J.; Bahleda, R.; van Laethem, J.-L.; Ottensmeier, C.; Lopez-Martin, J.A.; Clive, S.; Joly, F.; et al. A Phase I/II, Multiple-Dose, Dose-Escalation Study of Siltuximab, an Anti-Interleukin-6 Monoclonal Antibody, in Patients with Advanced Solid Tumors. Clin. Cancer Res. 2014, 20, 2192–2204. [Google Scholar] [CrossRef] [Green Version]
  225. A Safety, Efficacy and Pharmacokinetic Study of Siltuximab (CNTO 328) in Participants with Solid Tumors-Full Text View-ClinicalTrials.Gov. Available online: https://clinicaltrials.gov/ct2/show/NCT00841191 (accessed on 13 September 2021).
  226. Interleukin-6 Inhibitors | COVID-19 Treatment Guidelines. Available online: https://www.covid19treatmentguidelines.nih.gov/therapies/immunomodulators/interleukin-6-inhibitors/ (accessed on 12 September 2021).
  227. Actemra-Highlights of Prescribing Information. Available online: https://www.accessdata.fda.gov/drugsatfda_docs/label/2017/125276s114lbl.pdf (accessed on 19 September 2021).
  228. Kerschbaumer, A.; Sepriano, A.; Smolen, J.S.; van der Heijde, D.; Dougados, M.; van Vollenhoven, R.; McInnes, I.B.; Bijlsma, J.W.J.; Burmester, G.R.; de Wit, M.; et al. Efficacy of Pharmacological Treatment in Rheumatoid Arthritis: A Systematic Literature Research Informing the 2019 Update of the EULAR Recommendations for Management of Rheumatoid Arthritis. Ann. Rheum. Dis. 2020, 79, 744. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  229. Fleischmann, R.; van Adelsberg, J.; Lin, Y.; da Castelar-Pinheiro, G.R.; Brzezicki, J.; Hrycaj, P.; Graham, N.M.H.; van Hoogstraten, H.; Bauer, D.; Burmester, G.R. Sarilumab and Nonbiologic Disease-Modifying Antirheumatic Drugs in Patients With Active Rheumatoid Arthritis and Inadequate Response or Intolerance to Tumor Necrosis Factor Inhibitors. Arthritis Rheumatol. 2017, 69, 277–290. [Google Scholar] [CrossRef]
  230. Kevzara-Highlights of Prescribing Information. Available online: https://www.accessdata.fda.gov/drugsatfda_docs/label/2017/761037s000lbl.pdf (accessed on 19 September 2021).
  231. Roche-Enspryng (Satralizumab). Available online: https://www.roche.com/products/product-details.htm?productId=6f4b47a2-7ca9-4768-bf1a-6d813702f5b6 (accessed on 16 September 2021).
  232. Eskandary, F.; Dürr, M.; Budde, K.; Doberer, K.; Reindl-Schwaighofer, R.; Waiser, J.; Wahrmann, M.; Regele, H.; Spittler, A.; Lachmann, N.; et al. Clazakizumab in Late Antibody-Mediated Rejection: Study Protocol of a Randomized Controlled Pilot Trial. Trials 2019, 20, 1–13. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  233. Rovin, B.H.; van Vollenhoven, R.F.; Aranow, C.; Wagner, C.; Gordon, R.; Zhuang, Y.; Belkowski, S.; Hsu, B. A Multicenter, Randomized, Double-Blind, Placebo-Controlled Study to Evaluate the Efficacy and Safety of Treatment With Sirukumab (CNTO 136) in Patients With Active Lupus Nephritis. Arthritis Rheumatol. 2016, 68, 2174–2183. [Google Scholar] [CrossRef] [Green Version]
  234. Reeh, H.; Rudolph, N.; Billing, U.; Christen, H.; Streif, S.; Bullinger, E.; Schliemann-Bullinger, M.; Findeisen, R.; Schaper, F.; Huber, H.J.; et al. Response to IL-6 Trans- and IL-6 Classic Signalling Is Determined by the Ratio of the IL-6 Receptor α to Gp130 Expression: Fusing Experimental Insights and Dynamic Modelling. Cell Commun. Signal. 2019, 17, 1–21. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  235. Jakafi-Highlights of Prescribing Information. Available online: https://www.accessdata.fda.gov/drugsatfda_docs/label/2019/202192s017lbl.pdf (accessed on 19 September 2021).
  236. Xeljanz-Highlights of Prescribing Information. Available online: https://www.accessdata.fda.gov/drugsatfda_docs/label/2018/203214s018lbl.pdf (accessed on 19 September 2021).
  237. Goel, P.; Gerriets, V. Chloroquine. In StatPearls; StatPearls Publishing: Treasure Island, FL, USA, 2021; pp. 1–7. [Google Scholar]
  238. Jang, C.-H.; Choi, J.-H.; Byun, M.-S.; Jue, D.-M. Chloroquine Inhibits Production of TNF-α, IL-1β and IL-6 from Lipopolysaccharide-Stimulated Human Monocytes/Macrophages by Different Modes. Rheumatology 2006, 45, 703–710. [Google Scholar] [CrossRef] [Green Version]
  239. Aralen. Available online: https://www.accessdata.fda.gov/drugsatfda_docs/label/2017/006002s044lbl.pdf (accessed on 19 September 2021).
  240. Bansal, P.; Goyal, A.; Cusick, A., IV; Lahan, S.; Dhaliwal, H.S.; Bhyan, P.; Bhattad, P.B.; Aslam, F.; Ranka, S.; Dalia, T.; et al. Hydroxychloroquine: A Comprehensive Review and Its Controversial Role in Coronavirus Disease 2019. Ann. Med. 2021, 53, 117. [Google Scholar] [CrossRef] [PubMed]
  241. Plaquenil. Available online: https://www.accessdata.fda.gov/drugsatfda_docs/label/2017/009768s037s045s047lbl.pdf (accessed on 19 September 2021).
  242. Wang, Y.; Huang, Z.; Wang, L.; Meng, S.; Fan, Y.; Chen, T.; Cao, J.; Jiang, R.; Wang, C. The Anti-Malarial Artemisinin Inhibits pro-Inflammatory Cytokines via the NF-ΚB Canonical Signaling Pathway in PMA-Induced THP-1 Monocytes. Int. J. Mol. Med. 2011, 27, 233–241. [Google Scholar] [CrossRef] [PubMed]
  243. Artesunate-Highlights of Prescribing Information. Available online: https://www.accessdata.fda.gov/drugsatfda_docs/label/2020/213036s000lbl.pdf (accessed on 19 September 2021).
  244. Lin, L.; Benson, D.M.; DeAngelis, S.; Bakan, C.E.; Li, P.-K.; Li, C.; Lin, J. A Small Molecule, LLL12 Inhibits Constitutive STAT3 and IL-6-Induced STAT3 Signaling and Exhibits Potent Growth Suppressive Activity in Human Multiple Myeloma Cells. Int. J. Cancer 2012, 130, 1459–1469. [Google Scholar] [CrossRef] [Green Version]
  245. Shi, W.; Yan, D.; Zhao, C.; Xiao, M.; Wang, Y.; Ma, H.; Liu, T.; Qin, H.; Zhang, C.; Li, C.; et al. Inhibition of IL-6/STAT3 Signaling in Human Cancer Cells Using Evista. Biochem. Biophys. Res. Commun. 2017, 491, 159–165. [Google Scholar] [CrossRef]
  246. Evista-Highlights of Prescribing Information. Available online: https://www.accessdata.fda.gov/drugsatfda_docs/label/2007/020815s018lbl.pdf (accessed on 19 September 2021).
  247. Hayashi, M.; Rho, M.-C.; Enomoto, A.; Fukami, A.; Kim, Y.-P.; Kikuchi, Y.; Sunazuka, T.; Hirose, T.; Komiyama, K.; Omura, S. Suppression of Bone Resorption by Madindoline A, a Novel Nonpeptide Antagonist to Gp130. Proc. Natl. Acad. Sci. USA 2002, 99, 14728. [Google Scholar] [CrossRef] [Green Version]
  248. Singh, S.; Gajulapati, V.; Gajulapati, K.; Goo, J.; Park, Y.H.; Jung, H.Y.; Lee, S.Y.; Choi, J.H.; Kim, Y.K.; Lee, K.; et al. Structure–Activity Relationship Study of a Series of Novel Oxazolidinone Derivatives as IL-6 Signaling Blockers. Bioorganic Med. Chem. Lett. 2016, 26, 1282–1286. [Google Scholar] [CrossRef]
  249. Zhang, Z.; Zhou, L.; Xie, N.; Nice, E.C.; Zhang, T.; Cui, Y.; Huang, C. Overcoming Cancer Therapeutic Bottleneck by Drug Repurposing. Signal Transduct. Target. Ther. 2020, 5, 1–25. [Google Scholar] [CrossRef]
  250. Gyebi, G.A.; Ogunyemi, O.M.; Ibrahim, I.M.; Afolabi, S.O.; Adebayo, J.O. Dual Targeting of Cytokine Storm and Viral Replication in COVID-19 by Plant-Derived Steroidal Pregnanes: An in Silico Perspective. Comput. Biol. Med. 2021, 134, 104406. [Google Scholar] [CrossRef]
  251. Results of a Phase IIb Study of Vobarilizumab, an Anti-Interleukin 6 Receptor Nanobody®, in Patients with Moderate-to-Severe Rheumatoid Arthritis Despite Treatment with Methotrexate. Available online: https://www.ablynx.com/uploads/data/files/cra2017_alx-0061_abstract%20210_poster_final.pdf (accessed on 3 October 2021).
  252. Genovese, M.C.; Durez, P.; Fleischmann, R.; Tanaka, Y.; Furst, D.; Yamanaka, H.; Korneva, E.; Vasyutin, I.; Takeuchi, T. Long-Term Safety and Efficacy of Olokizumab in Patients with Rheumatoid Arthritis and Inadequate Response to Tumor Necrosis Factor Inhibitor Therapy in Phase II Studies. Eur. J. Rheumatol. 2021, 8, 120–129. [Google Scholar] [CrossRef] [PubMed]
  253. Baricitinib Letter of Authorization Revised July 28 2021. Available online: https://www.fda.gov/media/143822/download (accessed on 3 October 2021).
  254. FDA Briefing Document Pharmacy Compounding Advisory Committee (PCAC) Meeting. Available online: https://www.fda.gov/media/95976/download (accessed on 3 October 2021).
Figure 1. (A): All cells express signal transducer glycoprotein gp130. This molecule can be membrane-bound (gp130) or soluble (sgp130). Expression of IL-6 and its receptor, IL-6R, can be cell-type restricted. IL-6R, if expressed, can be bound to the membrane or it can be released as a soluble receptor (sIL-6R) due to the protease activity (by ADAM10 or ADAM 17). Once released from the membrane (shedding), it can also reach cells that do not express their own IL-6R. The IL-6 cytokine can act on the cell of the same origin (autocrine effect), on the neighboring cells (paracrine effect) or even exert systemic effects of endocrine nature (it is transported via the blood stream into capillaries). (B): Activation of gp130 by the IL-6/IL-6R complex (classic signaling) or by IL-6/sIL-6R (trans-signaling) leads to tyrosine phosphorylation (“P”) of transcription factors JAKs/STATs, PI3K and MAPK. Upon their phosphorylation, the regulatory molecules transmit the signal downstream, translocate into the nucleus and bind to enhancers of IL-6-type cytokine target genes.
Figure 1. (A): All cells express signal transducer glycoprotein gp130. This molecule can be membrane-bound (gp130) or soluble (sgp130). Expression of IL-6 and its receptor, IL-6R, can be cell-type restricted. IL-6R, if expressed, can be bound to the membrane or it can be released as a soluble receptor (sIL-6R) due to the protease activity (by ADAM10 or ADAM 17). Once released from the membrane (shedding), it can also reach cells that do not express their own IL-6R. The IL-6 cytokine can act on the cell of the same origin (autocrine effect), on the neighboring cells (paracrine effect) or even exert systemic effects of endocrine nature (it is transported via the blood stream into capillaries). (B): Activation of gp130 by the IL-6/IL-6R complex (classic signaling) or by IL-6/sIL-6R (trans-signaling) leads to tyrosine phosphorylation (“P”) of transcription factors JAKs/STATs, PI3K and MAPK. Upon their phosphorylation, the regulatory molecules transmit the signal downstream, translocate into the nucleus and bind to enhancers of IL-6-type cytokine target genes.
Ijms 22 11027 g001
Figure 2. Effects of IL-6 expression in HNSCC [6,15,83,84,85,86,88,89,90]. The role of the IL-6 cytokine is multifaceted in HNSCC. In the brain, IL-6 can be linked to the onset of depression and can also cause food intake disorders (anorexia), leading to wasting (cachexia). At the cellular level, IL-6 promotes epithelial-to-mesenchymal transition, which leads to the formation of metastatic lesions. Maintenance of low differentiation is associated with certain stemness features in tumor cells, predisposing to a poor response to chemo- and radiotherapy. However, many of these factors can further potentiate each other interdependently.
Figure 2. Effects of IL-6 expression in HNSCC [6,15,83,84,85,86,88,89,90]. The role of the IL-6 cytokine is multifaceted in HNSCC. In the brain, IL-6 can be linked to the onset of depression and can also cause food intake disorders (anorexia), leading to wasting (cachexia). At the cellular level, IL-6 promotes epithelial-to-mesenchymal transition, which leads to the formation of metastatic lesions. Maintenance of low differentiation is associated with certain stemness features in tumor cells, predisposing to a poor response to chemo- and radiotherapy. However, many of these factors can further potentiate each other interdependently.
Ijms 22 11027 g002
Table 1. List of recently tested anti-IL-6 therapeutics.
Table 1. List of recently tested anti-IL-6 therapeutics.
AgentMode of Action Tested in HNSCCApproved for Human Application in (Primary Indication):
In VitroIn Vivo (Animal Study)Clinical Trial
(Registered on Clinicaltrials.gov)
2-O-Methylmagnolol◼ downstream signaling inhibitionWang et al., 2018 [207]Wang et al., 2018 [207]--
AMD3100
(CXCR4 inhibitor)
inhibition of IL-6 productionTang et al., 2008 [187]---
PD98059
(MEK inhibitor)
inhibition of IL-6 productionTang et al., 2008 [187]---
Ammonium pyrrolidinecarbodithioate
(PDTC)
inhibition of IL-6 productionTang et al., 2008 [187]---
Aryl hydrocarbon receptor antagonist
(BAY 2416964)
inhibition of IL-6 productionDiNatale et al., 2011, 2012 [185,186]-NCT04069026 § [212]-
Bazedoxifene◆ gp130 blockadeYadav et al., 2017 [209]Yadav et al., 2017 [209]-Postmenopausal osteoporosis [213]
Cepharanthin inhibition of IL-6 productionTamatani et al., 2007 [180]---
Cilengitide (EMD 121974)
+ cetuximab
inhibition of IL-6 productionWichmann et al., 2017 [177]--Cetuximab–metastatic colorectal cancer, advanced HNSCC [214]
Curcumin inhibition of IL-6 production
◼ downstream signaling inhibition
Chakravarti et al., 2006 [184]
Cohen et al., 2009 [183]
Meyer et al., 2011 [210]
Yu et al., 2013 [63]NCT04208334; Thambamroong et al., 2016 [215]Dietary supplement [216]
Celecoxib + Simvastatin inhibition of IL-6 productionGehrke et al. 2017 [208]--Celecoxib - NSAID (non-steroidal anti-inflammatory drug) [217]
Simvastatin - reduction of LDL (low-density lipoprotein) cholesterol blood levels [218]
Cyclopentenone prostaglandin 15d-PGJ2◼ downstream signaling inhibitionSiavash et al., 2004 [188]---
Epigallocatechin gallate◼ downstream signaling inhibitionLin et al., 2012 [182]--Dietary supplement [219]
Guggulsterone◼ downstream signaling inhibitionMacha et al., 2011 [181]
Leeman-Neill et al. [220]
Leeman-Neill et al. [220]-Dietary supplement [220]
Honokiol inhibition of IL-6 production
◼ downstream signaling inhibition
Chang et al., 2018 [206]--Dietary supplement [221]
Lactoferrin (Human, recombinant) inhibition of IL-6 productionWolf et al. [178]Wolf et al. [178]--
RhoC knockdown
(lentiviral vector-based shRNA)
inhibition of IL-6 productionIslam et al. [193]---
L-leucine-methylester
(lysosomotropic agent)
inhibition of IL-6 productionKross et al. [199]---
Luteolin◼ downstream signaling inhibitionTu et al. [179]--Dietary supplement, popular in chinese traditional medicine [222]
MEDI5117
(anti-IL-6 humanized monoclonal antibody, IgG1, with Enhanced Serum Half-Life; also known as WBP216)
IL-6 neutralizationFinkel et al., 2016 [195]Finkel et al., 2016 [195]--
P276-00
(cyclin-dependent kinase inhibitor)
inhibition of IL-6 productionMishra et al., 2013 [200]Mishra et al., 2013 [200]--
Rapamycin inhibition of IL-6 productionEkshyyan et al., 2016 [196]--Immunosuppressive drug [223]
SB203580
(p38 MAPK inhibitor)
inhibition of IL-6 productionRiebe et al., 2007 [197]
Jing et al., 2016 [198]
Jing et al., 2016 [198]--
Siltuximab
(anti-IL-6 chimeric (human-murine) monoclonal recombinant antibody (IgG1κ))
IL-6 neutralization--NCT00841191; Angevin et al., 2014 [224,225]Management of systemic inflammation in COVID-19 [226]
Insulin-like growth factor-II mRNA binding protein-3 and podoplanin knockdown (lentiviral vector-based shRNA) inhibition of IL-6 productionHwang et al., 2016 [194]Hwang et al., 2016 [194]--
TLR-9- knockdown (siRNAs) inhibition of IL-6 productionRuan et al., 2014 [211]---
Tristetraprolin knockdown (lentiviral vector-based shRNA) inhibition of IL-6 productionVan Tubergen et al., 2011, 2013 [203,204]Van Tubergen et al., 2013 [204]--
Tetrathiomolybdate inhibition of IL-6 productionTeknos et al., 2005 [202]Teknos et al., 2005 [202]--
Tocilizumab
(anti-IL-6R humanised IgG1 monoclonal recombinant antibody )
❖ IL-6R blockadeMatsuoka et al., 2016 [88]
Stanam et al., 2015 [190]
Shinriki et al., 2009, 2011 [191,192]
Poth et al., 2010 [189]
Stanam et al., 2015 [190]
Shinriki et al., 2009, 2011 [191,192]
-Rheumatoid arthritis [227]
Giant cell arteritis [227]
Polyarticular Juvenile idiopathic arthritis [227]
Systemic Juvenile Idiopathic arthritis [227]
Cytokine Release syndrome [227]
Management of systemic inflammation in COVID-19 [226]
Triazolothiadiazine◆ gp130 blockadeSen et al., 2017 [201]---
Oxazole-piperazine◆ gp130 blockadeSen et al., 2017 [201] -
WP1066
(JAK inhibitor)
◼ Downstream signaling inhibitionZhou et al., 2014 [205]Zhou et al., 2014 [205]--
YM529 (third-generation bisphosphonate, Minodronic acid) inhibition of IL-6 production-Cui et al., 2005 [176]--
§ ongoing clinical trial, IL-6 production, IL-6 neutralization, ❖ IL-6R blocking, ◆ gp130 blocking, ◼ downstream signaling inhibition.
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Španko, M.; Strnadová, K.; Pavlíček, A.J.; Szabo, P.; Kodet, O.; Valach, J.; Dvořánková, B.; Smetana, K., Jr.; Lacina, L. IL-6 in the Ecosystem of Head and Neck Cancer: Possible Therapeutic Perspectives. Int. J. Mol. Sci. 2021, 22, 11027. https://doi.org/10.3390/ijms222011027

AMA Style

Španko M, Strnadová K, Pavlíček AJ, Szabo P, Kodet O, Valach J, Dvořánková B, Smetana K Jr., Lacina L. IL-6 in the Ecosystem of Head and Neck Cancer: Possible Therapeutic Perspectives. International Journal of Molecular Sciences. 2021; 22(20):11027. https://doi.org/10.3390/ijms222011027

Chicago/Turabian Style

Španko, Michal, Karolína Strnadová, Aleš Jan Pavlíček, Pavol Szabo, Ondřej Kodet, Jaroslav Valach, Barbora Dvořánková, Karel Smetana, Jr., and Lukáš Lacina. 2021. "IL-6 in the Ecosystem of Head and Neck Cancer: Possible Therapeutic Perspectives" International Journal of Molecular Sciences 22, no. 20: 11027. https://doi.org/10.3390/ijms222011027

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