Next Article in Journal
Atezolizumab Induces Necroptosis and Contributes to Hepatotoxicity of Human Hepatocytes
Next Article in Special Issue
Investigation of Mutated in Colorectal Cancer (MCC) Gene Family Evolution History Indicates a Putative Role in Th17/Treg Differentiation
Previous Article in Journal
The Influence of Pedo-Climatic Conditions on the Micromorphological, Phytochemical Features, and Biological Properties of Leaves of Saponaria sicula Raf
Previous Article in Special Issue
Upgrading Treatment and Molecular Diagnosis in Endometrial Cancer—Driving New Tools for Endometrial Preservation?
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Immune Checkpoint Inhibitors, Small-Molecule Immunotherapies and the Emerging Role of Neutrophil Extracellular Traps in Therapeutic Strategies for Head and Neck Cancer

by
Connor H. O’Meara
1,*,
Zuhayr Jafri
2 and
Levon M. Khachigian
2
1
Department of Otorhinolaryngology, Head and Neck Surgery, Prince of Wales Hospital, Randwick, NSW 2031, Australia
2
Vascular Biology and Translational Research, School of Biomedical Sciences, UNSW Faculty of Medicine and Health, University of New South Wales, Sydney, NSW 2052, Australia
*
Author to whom correspondence should be addressed.
Int. J. Mol. Sci. 2023, 24(14), 11695; https://doi.org/10.3390/ijms241411695
Submission received: 21 June 2023 / Revised: 13 July 2023 / Accepted: 17 July 2023 / Published: 20 July 2023
(This article belongs to the Special Issue Cancer Prevention with Molecular Target Therapies 4.0)

Abstract

:
Immune checkpoint inhibitor (ICI) therapy has revolutionized the treatment of many cancer types, including head and neck cancers (HNC). When checkpoint and partner proteins bind, these send an “off” signal to T cells, which prevents the immune system from destroying tumor cells. However, in HNC, and indeed many other cancers, more people do not respond and/or suffer from toxic effects than those who do respond. Hence, newer, more effective approaches are needed. The challenge to durable therapy lies in a deeper understanding of the complex interactions between immune cells, tumor cells and the tumor microenvironment. This will help develop therapies that promote lasting tumorlysis by overcoming T-cell exhaustion. Here we explore the strengths and limitations of current ICI therapy in head and neck squamous cell carcinoma (HNSCC). We also review emerging small-molecule immunotherapies and the growing promise of neutrophil extracellular traps in controlling tumor progression and metastasis.

1. Introduction

The immune system is a dynamic and equipped mechanism, an intricate system of “recognition” and “on-off” switches. Unfortunately, cancers utilize this system to enable growth and escape. The role of the immune system in tumor regulation is particularly evident in the immunocompromised. Iatrogenic solid organ transplant, diabetes, autoimmunity requiring immunosuppressive therapy, HIV and hemoproliferative malignant disease or disorders and aging, are all associated with an increased risk of developing head and neck cancer (HNC) and worse outcomes [1,2,3,4,5,6,7,8,9,10,11]. Proliferating tumors utilize many forms of immunosuppression to tip the balance of immunoediting toward tumor progression [12]. Identifying therapies capable of shifting this balance back toward immunosurveillance should play an integral role in reducing morbidity- and mortality-associated HNC.
HNC, the sixth most common group of malignancies worldwide, results in 680,000 new cases annually, with squamous cell carcinoma (SCC) being the most common [13,14,15]. The incidence of HNC is increasing due to a range of factors including smoking, alcohol, human papillomavirus (HPV) infection and extended life expectancy [16].
Despite the vital role played by traditional therapies for HNSCC, namely surgery, radiotherapy and chemotherapy, prognosis remains poor and survival remains correlated to stage, with a 5-year survival rate of 50–60% and more than 60% presenting in the advanced stage [17,18]. More than 50% of HNSCCs have tumor recurrence and metastasis in less than 3 years [19]. Targeting the epidermal growth factor receptor (EGFR) was hailed a paradigm shift in personalizing HNSCC treatment, with the monoclonal antibody cetuximab demonstrating promise [20,21]; however, this has since demonstrated limited efficacy [22].
Compared with traditional therapies, new immunotherapy agents, namely antibodies targeting the PD-1/PD-L1 system, so-called immune checkpoint inhibitors (ICI) provide improved efficacy and comparatively lower toxicity for patients with advanced HNSCC [23,24,25,26]. KEYNOTE-048 (NCT02358031), a randomized open-label phase 3 study comparing the humanized monoclonal antibody pembrolizumab (Keytruda) targeting PD-1 alone or in conjunction with chemotherapy (platinum and 5-fluorouracil) against cetuximab with chemotherapy, demonstrated overall survival improvement in both treatment arms over standard-of-care therapy in recurrent or metastatic HNSCC [27]. Pembrolizumab was subsequently approved as a first-line therapeutic drug for patients with metastatic, unresectable and recurrent HNSCC. Unfortunately, the objective response rate (ORR) of pembrolizumab (or nivolumab/Optivo) in HNSCC is only 15%, with short-term durability [28,29]. In addition, immune-related adverse events (irAEs) secondary to immunotherapy treatment were identified in over 50% of patients, impacting clinical outcomes [30], with adverse-event-associated mortality evident in 0.3–1.3% of patients [31]. Common irAEs include gastrointestinal, dermatologic and endocrine toxicities, more specifically dermatitis, rash, nausea/vomiting, fever, headache, myalgia, hypothyroidism and fatigue [32]. Rarely, irAEs can be severe, resulting in carditis, nephritis, hepatitis, pneumonitis, gastrointestinal perforation and severe hematological dysfunction [33]. irAEs in ICI therapy have been associated with benefits, namely improvements in PFS, OS and ORR [34,35,36,37]. Consequently, balancing immunotherapy de-escalation or commencement of immunosuppressive therapy against a sub-optimal oncological outcome can be difficult.
Predictive biomarkers may be the key to identifying patients at risk of irAEs. To date, circulating blood counts and ratios, autoantibodies and autoantigens, microRNAs, gastrointestinal microbiome, T-cell diversification and expansion and cytokines are all being investigated; however, they remain to be validated for clinical use [38].
Biological, etiological, phenotypic and clinical heterogeneities characterize HNSCC and challenge the development of personalized medicine. However, poor survival, significant morbidity and compromised quality of life emphasize the requirement for innovative therapy. Immunoediting is the process through which the immune system can promote and constrain tumor development [39]. This article explores current and developing therapies in immunomodulation and the developing role of neutrophil extracellular traps (NETs), net-like structures comprised of DNA-histone complexes and proteins in immune-mediated tumorigenesis.

2. Immune Checkpoint Inhibitor Targets and Therapies

A successful objective ICI response revitalizes the immune system to recognize and target cancer cells. The roles of known key immune checkpoints CTLA-4, PD-1 and LAG-3 are summarized in Figure 1.

2.1. CTLA-4 and PD-1/PD-L1

CTLA-4 (cytotoxic T-lymphocyte associated protein 4, also known as cluster of differentiation 152, CD152) and programmed cell death protein 1 (PD-1) (and its ligands PD-L1 and PD-L2) are immune checkpoints targeted by humanized antibodies for the treatment of HNSCC. CTLA-4 is bound by ipilimumab (Yervoy), whereas PD-1 is targeted by pembrolizumab and nivolumab [32]. The antibodies atezolizumab (Tecentriq), durvalumab (Imfinzi) and avelumab (Bavencio) have also been approved as inhibitors of PD-L1 [33]. Both checkpoints regulate different stages of the immune response. CTLA-4 is considered the “leader” of the immune response and prevents the stimulation of autoreactive T-cells in the initial stage of naïve T-cell activation, whereas PD-1 is thought to regulate previously activated T-cells at the later stages of the immune response [32].
CTLA-4 is a homolog of CD28, but unlike CD28, CTLA-4 activation has an immunosuppressive effect opposite to the stimulatory effect of CD28 and the T-cell receptor (TCR) [40]. The binding of CD80/CD86 on antigen-presenting cells to CTLA-4 on T-cells in the tumor microenvironment suppresses the immune system, enabling tumor proliferation [41]. PD-1’s interaction with PD-L1 and PD-L2 has an immunosuppressive effect [41]. PD-L1 and PD-L2 are expressed by a range of tumors including HNSCC [42]. Critically, increased PD-1 levels serve as a biomarker for T cell exhaustion; this state of exhaustion is linked to T-cell dysfunction, which can facilitate tumor proliferation [43]. PD-L1′s interaction with PD-1 has an immunosuppressive effect, thus protecting cancer cells from lysis by activated T-cells [44].
Despite ICI therapy demonstrating survival advantage, comparatively few patients develop an effective response, the durability of which attenuates with acquired tumor resistance. Acquired resistance leads to tumor progression, and both arms of the immune system, innate and adaptive, can play a critical role in this change. Mechanisms of resistance to immunotherapy can be either intrinsic (tumor cell-mediated) or extrinsic (processes associated with T-cell activation) and shift the balance of immunomodulation towards tumor proliferation. Intrinsic resistance can include the downregulation of antigen-presenting machinery (APM) [45], the up-regulation of signaling pathways promoting T-cell exhaustion [46], the expression of multiple checkpoint inhibitors to mitigate T-cell activation [47], changes in tumor cell DNA repair, damage and genomic instability [48] and altered kinase signaling pathways [49]. Extrinsic resistance involves the complex interplay between tumor cells and the tumor microenvironment and its ability to regulate phenotypical characteristics of immune cells, especially TANs, TAMs, Tregs, MDSCs, T-cells, their associated regulatory cytokines and signaling pathways and a newly identified player, NETs [50,51,52,53,54,55,56].
Despite the clear improvements in overall survival due to immune checkpoint therapy, such treatments have limitations. For example, since CTLA-4 prevents the stimulation of autoreactive T-cells, inhibiting CTLA-4 can lead to grade 3 or 4 autoimmune-related adverse effects in 10–15% of patients [57]. Immune checkpoint immunotherapies are also associated with low response rates. For example, pembrolizumab has a response rate of only 15% in HNSCC [58].
To improve therapeutic failure and overcome immunotherapy resistance, significant energy is being invested in exploring biomarkers to predict clinical response and combinational therapies or changes in adjuvant delivery of immunotherapy to increase success rates. Biomarkers that have shown potential to determine improved clinical response in HNSCC include the tumor mutational burden, CCND1 amplification (CCND1 encodes cyclin D1, which regulates the retinoblastoma protein activity and cell-cycle progression), PD-1, IFN-γ, tumor-infiltrating lymphocytes (TILs) and cancer-associated fibroblasts (CAFs), CTLA-4, exosomes, CXCL, MTAP and SFR4/CPXM1/COL5A1 molecules [25,59,60,61,62,63,64,65,66,67,68,69].
Clinical trials exploring combinational immunotherapy in HNSCC are underway. The phase 3 randomized trial CheckMate 651 NCT02741570), which compared nivolumab and ipilimumab against EXTREME (platinum/5-fluorouracil/cetuximab) for R/M HNSCC, was unsuccessful in demonstrating OS improvement, although there was an association between elevated CPS and OS and durable response [70]. Other combination ICI therapy clinical trials have been largely unsuccessful (Table 1).
Concurrent neoadjuvant and adjuvant delivery of ICIs has recently demonstrated benefits in surgically resectable advanced melanoma (Stage IIIB to IVC). In a recently completed Phase 2 randomized study (NCT03698019), neoadjuvant-adjuvant delivery of pembrolizumab was compared to an adjuvant alone in demonstrating an event-free survival of 72% in the neoadjuvant-adjuvant group compared to 49% in the adjuvant group after 2 years [76].

2.2. LAG-3

LAG-3 is expressed on activated human T-cells and natural killer cells and plays a similar role in T-cell regulation to CTLA-4 and PD-1 [77]. LAG-3 may represent an intrinsic resistance mechanism to PD-1 inhibitors due to its synergistic co-expression with PD-1 on exhausted T-cells [77]. To combat resistance, the FDA-approved drug opdualag® (combined LAG-3 and PD-1 inhibitor) became a first-line treatment for unresectable or metastatic melanoma in March 2022 [77]. Opdualag has shown success in clinical trials, more than doubling progression-free survival compared to melanoma patients treated with nivolumab alone [78].

2.3. Tim-3 and CD39

T cell immunoglobulin and mucin domain-containing protein 3 (Tim-3) is a co-inhibitory receptor expressed on IFN-γ-producing T-cells Tim-3. Studies by Liu et al. showed that Tim-3 is linked to immunosuppression in HNSCC and that targeting Tim-3 (with monoclonal antibodies) can enhance the anti-tumor immune response by reducing Tregs in HNSCC [79]. Similarly, the expression of the cell-surface ectonucleosidase CD39 in HNSCC positively correlates with tumor stage and predicts poor prognosis [80]. There are no approved inhibitors of Tim-3 or CD39, and opdualag has not yet been approved for HNSCC.

3. Emerging Immunotherapeutic Targets and Strategies

Small-molecule immunotherapy (SMI) may represent the paradigm shift required to improve quality of life (QOL) and survival in HNSCC. Unlike current ICI therapies, small molecules can be delivered orally, are potentially less expensive than antibodies and utilized to target intracellular signaling and transcriptional pathways upstream of receptors expressed on the cell surface. Several promising SMIs in various phases of development are listed in Table 2 and described below.

3.1. STAT3

Among the intracellular signal transducer and activator of transcription (STAT) proteins, STAT3 plays an important hemostatic role in normal cells by helping to regulate cell growth, survival, differentiation, angiogenesis, immune response and cellular respiration [87,88]. STAT3 can be activated by both JAK and EGFR (via Src) signaling, subsequently binding target DNA to regulate gene expression [89,90]. Importantly, STAT3 activation can upregulate multiple survival proteins, namely Bcl-xL, survivin and Bcl-2 [91]. STAT3 is also an upstream regulator of PD-1 expression [92,93]. STAT3 can behave as an oncogene and is expressed in approximately 70% of human cancers [19]. Overexpression of STAT3 regulates tumor progression and is associated with poor prognosis in various malignancies [94]. Elevated levels of IL-6, released by tumor-infiltrating lymphocytes, M2 phenotype macrophages [95], cancer-associated fibroblasts [96] and tumor cells [97,98], can activate a pro-tumorigenic IL-6/STAT3 pathway [99], inhibiting dendritic cell maturation, suppressing CD8+ T-cell and NK cell activation [100,101,102] and promoting CD4+ T cell differentiation to a T regulatory phenotype [103]. This pathway supports the survival of immunosuppressive MDSCs and M2 phenotype macrophages [104,105] and supports tumor survival, invasiveness and proliferation [106]. STAT3 can also regulate metabolism-related genes that favor cancer progression [107] and promote angiogenesis via the upregulation of VEGF [108]. STAT3 activation was identified to be 10.6- and 8.8-fold higher in tumors and normal mucosa, respectively, of HNSCC patients compared to the mucosa of non-cancer patients, supporting the concept of “field cancerization” [109]. Furthermore, there is a strong association between downstream proteins transcribed by STAT3 and locoregional metastasis, stage, recurrence and mortality in OCSCC [110]. There is also evidence that constitutive STAT3 activation plays a prominent role in mediating drug resistance to many targeted cancer therapies and chemotherapies, including the poor response to the EGFR blockade, with less than 15% of patients benefiting from cetuximab as a single agent or 36% when combined with chemotherapy [111]. Certainly, STAT3 inhibition has improved radiotherapy and cetuximab responsiveness in HNSCC cell lines [91,112]. Hence preclinical and early clinical trial data suggest that targeting STAT3 is a promising therapeutic strategy [113].
Flubendazole is a benzimidazole, long utilized as a macrofilaricide in humans and animals. Recently, it has been repurposed and recognized as a promising anti-cancer agent, effective in breast cancer, melanoma, neuroblastoma, colorectal, liver and squamous cell carcinoma [114,115,116,117,118,119,120,121,122,123,124]. In melanoma, flubendazole reduced the expression of phosphorylated STAT3 in tumor tissue and the expression of PD-1 expression, while also decreasing MDSC levels in tumors [125]. Immunological signature gene sets, including those associated with T cell differentiation, proliferation and function correlated with FLU treatment [126]. Flubendazole has also been found to have synergistic antiproliferative effects in vitro with 5-fluorouracil [127], which raised the potential benefit of use topically in conjunction with 5-FU for the treatment of premalignant and malignant non-melanoma skin cancers [128,129,130]. Flubendazole has not entered clinical trials for HNC.
An alternative approach is provided by danvatirsen (AZD9150), an antisense oligonucleotide inhibitor of STAT3 comprised of 16 nucleotides, which has demonstrated antiproliferative effects in xenograft models showing reduced STAT3 expression, paving its way to clinical trial [131]. Combining durvalumab (MEDI4736, PD-L1 inhibitor) with danvatirsen or AZD5069 (CXCR2 inhibitor) (NCT02499328) in patients with advanced solid malignancies and HNSCC improved anticancer activity as compared to PD-L1 monotherapy [81].

3.2. STING

STING (stimulator of interferon genes) is a cytosolic pattern-recognition receptor (PRR) that recognizes non-self-dsDNA, upregulating type 1 interferon [132]. Recent evidence indicates that type 1 IFN plays an important role in many anticancer modalities, including immunotherapy, helping to promote dendritic cell activation and prime and recruit cytotoxic CD8+ T-cells against tumor-associated antigens [133,134,135]. Evidence suggests that the STING pathway may help potentiate checkpoint blockade therapy [136,137].

3.3. PPAR

Peroxisome proliferator-activated receptors (PPARs) regulate a multitude of cellular functions. They are a family of ligand-inducible nuclear hormone receptors belonging to the steroid receptor superfamily. PPAR-α is commonly expressed in skeletal muscle, liver, heart and brown adipose tissue. Its activation suppresses NF-κB signaling, which decreases the inflammatory cytokine production by different cells and modulates the proliferation, differentiation and survival of macrophages, B-cells and T-cells, whilst also playing a role in angiogenesis, homeostasis and glucose and lipid metabolism [138,139,140,141]. Notwithstanding a pleiotropic role in cancer, which appears type and tumor microenvironment (TME)-dependent, increasing evidence is demonstrating that PPAR-γ can modulate carcinogenesis, showing promise as a focus for cancer therapies. PPAR-γ agonists have been shown to inhibit cancer cell proliferation and Warburg effects.

3.4. RTKs

There are over 50 known RTKs in humans. These are transmembrane receptors integral to cell-to-cell communication and the regulation of cell growth, metabolism, motility and cell differentiation. These mediate the activation of a variety of signaling pathways, including JAK/STAT, PI-3K/AKT/mTOR, PLC/PKC and RAS/MAPK. Their dysregulation plays a role in multiple human disease processes, including carcinogenesis, which can confer constitutive activation by genomic amplification, chromosomal rearrangements, autocrine activation, gain-of-function mutations or kinase domain duplication [142,143].
There is evidence that IL-33, although a pleiotropic cytokine in HNSCC [144], can regulate immune cells in the TME, namely CD4+ T-helper cells, CD8+ T-cells, NK cells, DCs and macrophages [144]. Developing evidence suggests that IL-33 may regulate the immune response through a signaling complex between IL-33R and EGFR in gastrointestinal helminth infections [145]; however, we are not aware of current research supporting this pathway in cancer.
EGFR is a prototypic RTK and is well recognized to be susceptible to gain-of-function mutations and is commonly overexpressed in HNSCC. These mutations can hyperactivate the kinase and its downstream signaling, conferring oncogenic properties [146]. Eighty to ninety percent of HNSCCs overexpress or demonstrate EGFR mutation, with these changes detrimentally affecting both PFS and OS [147,148]. Certainly, EGFR status has been identified as a survival predictor and guide to the effectiveness of chemoradiation [149]. Although known mutations in EGFR are rare in HNC, its overexpression with TGF-α is common, and auto or paracrine activation is important in HNC EGFR function. Unfortunately, mutation commonly alters drug binding dynamics, leading to resistance, a similar phenomenon leading to a reduced radiation response and overall survival in HNSCC. Mutation status is also associated with the tumor stage [150]. Chromosomal rearrangements have been identified in the RET kinase in thyroid cancer [151] and TRKA, TRKB and TRKC tyrosine kinases in thyroid and HNC.
The primary site of action of TKIs on EGFR is the intracellular tyrosine kinase domain, inhibiting downstream signaling. Geftinib and erlotinib have been ineffective in HNC, comparatively lapatinib, afatinib and dacomitinib have demonstrated benefits in clinical trials and can target VEGFR to reduce tumor angiogenesis. Cetuximab has shown an ORR of 13% as a monotherapy, and gefitinib and erlotinib demonstrated ORR of 1.4% and 10.6% with a median OS of 5.5 and 8.1 months, respectively [152,153,154]. Anlotinib (AL3818) is a novel multi-target RTK antagonist against PDGFR, FGFR, VEGFR and c-Kit. In human OCSCC cell lines, it effectively reduced tumor cell proliferation and promoted apoptosis [155].

3.5. AHR

AHR (aryl hydrocarbon receptor) is a ligand-activated transcription factor activated by both anthropogenic and natural agonists, with recent studies reporting a key role in regulating host immunity [156,157]. As a transcription factor, AHR can regulate the expression of cytochrome P450 family genes. Chronically active AHR is capable of driving cancer cell invasion, migration, cancer stem cell characteristics and survival [158]. Tumor expression of AHR can result in an autocrine AHR-IL-6/STAT3 signaling loop via kynurenine, an immunosuppressive AHR agonist ligand produced by the metabolism of the essential amino acid tryptophan [159].

4. Neutrophils Extracellular Traps (NETs)

Neutrophils are the largest group of leukocytes within the blood and play an integral role in immune-mediated host defense mechanisms. As activated phagocytes, these secrete neutrophil elastase (NE), reactive oxygen species (ROS), nicotinamide adenine dinucleotide phosphate oxidase (NADPH) and myeloperoxidase (MPO) to digest pathogens [160]. Chemotactically attracted to the TME, tumor-associated neutrophils (TANs) phenotypically polarize to either N1 or N2 sub-types. Similar to M1 phenotype tumor-associated macrophages (TAMs), N1 TANs are anti-tumorigenic, whilst N2 TANs are pro-tumorigenic and regulate immunosuppression, tumor cell proliferation, angiogenesis and metastasis [161,162,163].
Derived from neutrophils undergoing a signal-mediated cell death program known as NETosis, NETs are extracellular “spider webs” of unwound chromatin, comprising histones, neutrophil elastase and granular antimicrobial enzymes [164,165] (Figure 2). These are key antimicrobial components of the innate defense system that sequester and contribute to bacterial cytotoxicity and phagocytosis [166,167]. Their antimicrobial purview includes inhibiting replication, containing and eliminating viral infections via the activation of TLR4, 7 and 8 pathways, PKC pathway blockade or aggregation and neutralizing effects of cationic histones, particularly arginine-rich H3 and H4 [168,169,170,171,172]. NETs are important in clearing large pathogens and are activated by β-glucan on fungal hyphae [173,174,175]. NETs recognize the activation of platelets and monocytes and limit the dissemination of parasites by trapping and killing these with histones, neutrophil elastase, MPO and collagenase mediating cytotoxicity [176,177,178,179]. Unfortunately, NET dysregulation is pivotal to the pathogenesis of numerous diseases, including sepsis [180,181], acute respiratory disease syndromes [182,183], ischemia-reperfusion injury [184], diabetes [185], venous thromboembolism [186] and chemotherapy-induced peripheral neuropathy [187].
New evidence indicates that NETs potentiate pro-tumorigenic effects, with neutrophils attracted to the tumor microenvironment being reprogrammed by tumor-associated factors to undergo NETosis and potentiate tumor activity. In this regard, NETs are capable of suppressing tumor cell apoptosis and promoting tumor cell invasion [188,189,190,191,192,193,194,195]. Factors involved in tumor-mediated NET formation include tumor-derived inflammatory and chemoattractant cytokines (IL-8, IL-6, TNF-α, G-CSF and IL-1β) [189,190,196,197,198,199], tumor extracellular vesicles [200], tumor-activated platelets [201,202], tumor-derived HMGB1 [203,204,205,206], KRAS oncogene mutation [207] and hypoxia [208].
Recent evidence has highlighted the ability of NETs to actively drive tumor growth and metastasis. NET-associated HMGB1 promotes tumor cell proliferation involving interaction with tumor RAGE, activating and NF-κB signaling [209]. Additionally, neutrophil elastase, via the PI3K signaling pathway, promotes the proliferation of adenocarcinoma cells [210]. NETs play a key role in shielding tumor cells from tumor-recognizing NK- and cytotoxic CD8+ T-cells [211,212], whilst promoting cytotoxic CD8+ T-cell exhaustion through the upregulation of PD-L1 [213]. Their immunosuppressive role also extends to the programming of T regulatory phenotype cells, which inhibit macrophage, dendritic, cytotoxic CD8+ T cell anti-tumor effects via a TLR4-dependent mechanism [214,215,216], which may be histone dependent, while they also play a key role in thrombosis [217,218,219,220,221]. Further compromising immunorecognition, NET-activated platelets may facilitate plasma membrane transfer, enabling tumor cell expression of platelet markers and MHC receptors to camouflage their presence within the platelet aggregate attached to a NET scaffold [222,223,224].
NETs can affect all arms of Virchow’s triad [225], activating platelets and endothelial cells, aggregating erythrocytes [226] and promoting tissue factor release from both platelets and endothelial cells. Specifically, by way of histone expression, NETs induced an endothelial cell shift toward a pro-coagulant phenotype [227,228,229]. NETs also promote thrombosis by acting as a scaffold that can capture and activate platelets, facilitate fibrin deposition and express TF for coagulation [230]. There is likely significant crosstalk between NETs and platelets, working in concert to enhance tumor cell survival and metastasis. Platelet TLR4 can trigger NET production, and NET-expressed histone H3 and H4 can activate platelets in a positive feedback loop [217,231].
The ability of NETs to promote thrombosis and capture circulating tumor cells enhances the ability of tumor cells to metastasize. However, their further ability to facilitate distant tumor growth is much more extensive. NETs can promote tumor cell migration and enhance invasiveness, with the rearrangement of the cytoskeleton elements, via their CCDC25 receptor, which may aid tumor cell transmigration across the endothelium [232]. Although the mechanism remains unclear, NETs can reprogram the epithelial-to-mesenchymal transition in tumor cells, essentially allowing them to disconnect cell-to-cell and cell-to-extracellular matrix chelation and commence migration and invasion [233,234,235]. NETs can also revive dormant tumor cells that have metastasized to an unfavorable microenvironment. Evidence suggests that extracellular matrix NET remodeled laminin binds dormant tumor a3B1 integrin, driving their activation via FAK/ERK/MLCK/YAP signalling, in turn supporting proliferation [236].
Angiogenesis is also a priority for hypoxic tumor cells. NETs have been demonstrated to promote angiogenesis [237,238,239] and this may well be via a histone-dependent mechanism [237]. Aldabbous and colleagues showed that NETs increased the vascularization of Matrigel plugs and release of MMP-9, TGF-β1 latency-associated peptide, HB-EGFGF and uPA, also promoting endothelial permeability and cell motility [238].
Interactions between NETs and cancer cells are also thought to drive resistance to various cancer therapies, including chemotherapy, immunotherapy and radiation therapies. Therefore, the development of therapies to mitigate the pro-tumorigenic role of NETs is an absolute necessity but should minimize interference with immunity, wound healing, and host defense mechanisms. There are several novel therapies being developed for the management of NETs in sterile systemic inflammatory response syndromes and several agents that may be repurposed to mitigate the pro-tumorigenic role of NETs [240,241,242,243].

5. Potential NET-Based Therapies

5.1. Novel Compounds Facilitating NET Prevention or Modulation

5.1.1. Conceptual

STC3141 (methyl β-cellobioside per-O-sulfate) is a small polyanion (SPA) that interacts electrostatically with histones, neutralizing their pathological effects preclinically in several pathologies [240]. This agent was developed specifically to target histones on NETs, to help preserve the host defense benefit of the protease-labelled chromatin web and facilitate microbe cytotoxicity in sepsis (histone neutralization with NET stabilization). Phase 1b results demonstrate favorable safety profiles and clinical benefits in ARDS. STC3141 is likely to interfere with the pro-tumorigenic functions of NETs, including tumor cell camouflage, migration and dormant cell reactivation, and may represent an effective small-molecule NET modulator. STC3141 may inhibit histone-dependent pathways, including TLR4/histone-dependent TME immunosuppression, histone-dependent endothelial and platelet activation and thrombosis, conferring survival and metastatic ability.

5.1.2. Preclinical

Sivelestat is an inhibitor of the NET-expressed serine protease neutrophil elastase, competitively inhibiting it with high specificity. NE plays a key role in NETosis and NET formation, and the pro-tumorigenic role of NE has been confirmed in breast, lung and colon cancers [244,245,246]. In keeping with the role of NETs in metastasis, Okamoto and colleagues demonstrated that sivelestat reduced NET formation and liver metastasis in a murine model of colorectal cancer (CRC) but had no effect on primary tumor growth or the suppression of liver metastasis if the CRC cells had already metastasized [247].
GSK383 chloramidine is a PAD4 inhibitor. PAD4 is a peptidyl arginine deiminase type IV enzyme, critical to the formation of NETs [248]. In 4T1 murine breast cancer cells, co-culture with GSK383 significantly attenuated NET production and inhibited NET-mediated tumor cell invasion.

5.1.3. Clinical Trials

Given the importance of the CXCR1/2:IL-8 axis in neutrophil/NET-mediated carcinogenesis, there are currently several CXCR1/2 inhibitors undergoing clinical trials in combination with ICIs. SX-682 (a CXCR1/2 antagonist) in combination with pembrolizumab entered Phase 1 trials in metastatic melanoma (NCT03161431), while a combination of avarixin (CXCR1/2 antagonist) and pembrolizumab is being trialed in advanced/metastatic solid tumors in a Phase 2 study (NCT03473925). CXCR1/2 inhibitors are not currently being trialed in HNC.

5.2. Repurposed Compounds Facilitating NET Prevention or Modulation

Preclinical/Clinical Cohort Studies

Aspirin is a COX-1 inhibitor commonly utilized as an antagonist to the primary hemostatic role of platelets. In a murine model of lipopolysaccharide-induced lung injury, aspirin reduced target tissue invasion by neutrophils and NET production. This is thought to be mediated by the amelioration of platelet-dependent release of CXCL4 (PF4) and CCL5 (RANTES), both of which increase neutrophil recruitment. Low-dose aspirin can have an anti-metastatic effect via a COX-1 inhibition-mediated reduction in NET production [249,250,251].
Metformin is a PKC inhibitor that attenuates NETosis. Previous studies identified that circulating NE, citrillinated histone, ds-DNA and proteinase-3 levels were reduced in the presence of metformin, and furthermore, metformin decreased expected NETosis in the presence of stimuli [252]. New evidence in hepatocellular carcinoma (HCC) and pancreatic cancer (PC) demonstrated that metformin attenuated NET production and reduced the metastatic potential of HCC and PC cells [253,254]. The role of metformin in attenuating NET-mediated carcinogenesis was corroborated in further murine models [255]. Interestingly, a recent presentation at AACR’s Annual Meeting revealed that patients with type 2 diabetes mellitus suffering from colorectal cancer had significantly improved DFS in the presence of metformin, with tissue analysis identifying a significant reduction in tumor-associated NETs and a significant increase in CD8+ T-cells. The authors concluded that metformin inhibits neutrophil infiltration and NET expression whilst promoting the infiltration of cytotoxic CD8+ T-cells in the TME [256].

5.3. Compounds Facilitating NET Destruction

Preclinical

Unfractionated heparin (UH) is a glycosaminoglycan that potentiates the enzyme antithrombin III, inactivating thrombin, factor Xa and other proteases. It has a high affinity for extracellular histones and has been shown to promote the degradation of NETs [257]. UH is readily available and no longer under patent but has an off-target side effect in increasing the risk of bleeding.
Dornase alfa (rhDNase 1) is a recombinant human deoxyribonuclease, which selectively cleaves DNA. A co-culture of triple-negative breast cancer cells with neutrophils formed significant NETs, with DNase 1 blocking both NET-mediated cancer cell migration and invasion. In vivo, DNase 1 has demonstrated some ability to attenuate metastasis in a murine lung cancer model [258]. Park and colleagues concluded that this effect could be improved by increasing the half-life of DNase 1 and developing DNase-1-coated nanoparticles, which reduced the metastatic burden in a 4T1 breast cancer murine model [259]. Wang and colleagues also demonstrated that rhDNase 1 mitigated the pro-tumorigenic effects of NETs in murine pancreatic cancer [253]. rhDNase 1 is an FDA-approved therapy to degrade chromatin in cystic fibrosis and is currently undergoing a clinical trial to determine similar benefits in COVID-19-associated ARDs (NCT04402944) [260].

6. Conclusions

While tumor development may be controlled by cytotoxic adaptive and innate immune cells, the challenge to durable therapy lies in understanding the complex interaction between the immune cells, the tumor and its microenvironment and delivering therapies that re-program immunomodulation toward tumorlysis. In this article, we explored the strengths and limitations of current ICI therapies and outlined emerging SMI targets and modalities including our growing understanding of the important role NETs play in immunomodulation and their promise in ameliorating tumor progression and metastasis.

Author Contributions

Conceptualization, C.H.O. and L.M.K.; writing—original draft preparation, C.H.O.; writing—review and editing, C.H.O., Z.J. and L.M.K.; figure preparation, Z.J. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Acknowledgments

The figures were created using BioRender under a licence acquired through the School of Biomedical Sciences, UNSW. CHO was supported by a Prince of Wales Hospital Foundation Grant to complete this review.

Conflicts of Interest

There are no conflict of interest.

References

  1. Tam, S.; Yao, C.M.K.L.; Amit, M.; Gajera, M.; Luo, X.; Treistman, R.; Khanna, A.; Aashiq, M.; Nagarajan, P.; Bell, D.; et al. Association of Immunosuppression with Outcomes of Patients with Cutaneous Squamous Cell Carcinoma of the Head and Neck. JAMA Otolaryngol. Head Neck Surg. 2020, 146, 128–135. [Google Scholar] [CrossRef] [PubMed]
  2. Szturz, P.; Vermorken, J.B. Treatment of Elderly Patients with Squamous Cell Carcinoma of the Head and Neck. Front. Oncol. 2016, 6, 199. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  3. Pritchett, E.N.; Doyle, A.; Shaver, C.M.; Miller, B.; Abdelmalek, M.; Cusack, C.A.; Malat, G.E.; Chung, C.L. Nonmelanoma Skin Cancer in Nonwhite Organ Transplant Recipients. JAMA Dermatol. 2016, 152, 1348–1353. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  4. Gilbert, M.; Liang, E.; Li, P.; Salgia, R.; Abouljoud, M.; Siddiqui, F. Outcomes of Primary Mucosal Head and Neck Squamous Cell Carcinoma in Solid Organ Transplant Recipients. Cureus 2022, 14, e24305. [Google Scholar] [CrossRef]
  5. Mowery, A.J.; Conlin, M.J.; Clayburgh, D.R. Elevated incidence of head and neck cancer in solid organ transplant recipients. Head Neck 2019, 41, 4009–4017. [Google Scholar] [CrossRef]
  6. Mowery, A.; Conlin, M.; Clayburgh, D. Risk of Head and Neck Cancer in Patients with Prior Hematologic Malignant Tumors. JAMA Otolaryngol. Head Neck Surg. 2019, 145, 1121–1127. [Google Scholar] [CrossRef]
  7. Wang, X.; Wang, H.; Zhang, T.; Cai, L.; Dai, E.; He, J. Diabetes and its Potential Impact on Head and Neck Oncogenesis. J. Cancer 2020, 11, 583–591. [Google Scholar] [CrossRef]
  8. Zhong, W.; Mao, Y. Daily Insulin Dose and Cancer Risk among Patients with Type 1 Diabetes. JAMA Oncol. 2022, 8, 1356–1358. [Google Scholar] [CrossRef]
  9. Batista, N.V.R.; Valdez, R.M.A.; Silva, E.; Melo, T.S.; Pereira, J.R.D.; Warnakulasuriya, S.; Santos-Silva, A.R.; Duarte, A.; Mariz, H.A.; Gueiros, L.A. Association between autoimmune rheumatic diseases and head and neck cancer: Systematic review and meta-analysis. J. Oral Pathol. Med. 2022, 52, 357–364. [Google Scholar] [CrossRef]
  10. Li, C.M.; Chen, Z. Autoimmunity as an Etiological Factor of Cancer: The Transformative Potential of Chronic Type 2 Inflammation. Front. Cell Dev. Biol. 2021, 9, 664305. [Google Scholar] [CrossRef]
  11. D’souza, G.; Carey, T.E.; William, W.N., Jr.; Nguyen, M.L.; Ko, E.C.; Riddell, J., IV; Pai, S.I.; Gupta, V.; Walline, H.M.; Lee, J.J.; et al. Epidemiology of head and neck squamous cell cancer among HIV-infected patients. J. Acquir. Immune Defic. Syndr. 2014, 65, 603–610. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  12. Dunn, G.P.; Old, L.J.; Schreiber, R.D. The three Es of cancer immunoediting. Annu. Rev. Immunol. 2004, 22, 329–360. [Google Scholar] [CrossRef] [PubMed]
  13. Dorsey, K.; Agulnik, M. Promising new molecular targeted therapies in head and neck cancer. Drugs 2013, 73, 315–325. [Google Scholar] [CrossRef] [PubMed]
  14. Torre, L.A.; Bray, F.; Siegel, R.L.; Ferlay, J.; Lortet-Tieulent, J.; Jemal, A. Global cancer statistics, 2012. CA Cancer J. Clin. 2015, 65, 87–108. [Google Scholar] [CrossRef] [Green Version]
  15. Haddad, R.I.; Shin, D.M. Recent advances in head and neck cancer. N. Engl. J. Med. 2008, 359, 1143–1154. [Google Scholar] [CrossRef] [Green Version]
  16. Qiao, X.W.; Jiang, J.; Pang, X.; Huang, M.C.; Tang, Y.J.; Liang, X.H.; Tang, Y.L. The Evolving Landscape of PD-1/PD-L1 Pathway in Head and Neck Cancer. Front. Immunol. 2020, 11, 1721. [Google Scholar] [CrossRef]
  17. Leemans, C.R.; Braakhuis, B.J.; Brakenhoff, R.H. The molecular biology of head and neck cancer. Nat. Rev. Cancer 2011, 11, 9–22. [Google Scholar] [CrossRef]
  18. Chow, L.Q.M. Head and Neck Cancer. N. Engl. J. Med. 2020, 382, 60–72. [Google Scholar] [CrossRef]
  19. Chi, A.C.; Day, T.A.; Neville, B.W. Oral cavity and oropharyngeal squamous cell carcinoma–An update. CA Cancer J. Clin. 2015, 65, 401–421. [Google Scholar] [CrossRef]
  20. Mehra, R.; Cohen, R.B.; Burtness, B.A. The role of cetuximab for the treatment of squamous cell carcinoma of the head and neck. Clin. Adv. Hematol. Oncol. 2008, 6, 742–750. [Google Scholar]
  21. Sundvall, M.; Karrila, A.; Nordberg, J.; Grénman, R.; Elenius, K. EGFR targeting drugs in the treatment of head and neck squamous cell carcinoma. Expert Opin. Emerg. Drugs 2010, 15, 185–201. [Google Scholar] [CrossRef] [PubMed]
  22. Sacco, A.G.; Worden, F.P. Molecularly targeted therapy for the treatment of head and neck cancer: A review of the ErbB family inhibitors. Onco Targets Ther. 2016, 9, 1927–1943. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  23. Addeo, R.; Ghiani, M.; Merlino, F.; Ricciardiello, F.; Caraglia, M. CheckMate 141 trial: All that glitters is not gold. Expert Opin. Biol. Ther. 2019, 19, 169–171. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  24. Ferris, R.L.; Blumenschein, G., Jr.; Fayette, J.; Guigay, J.; Colevas, A.D.; Licitra, L.; Harrington, K.; Kasper, S.; Vokes, E.E.; Even, C.; et al. Nivolumab for Recurrent Squamous-Cell Carcinoma of the Head and Neck. N. Engl. J. Med. 2016, 375, 1856–1867. [Google Scholar] [CrossRef] [Green Version]
  25. Seiwert, T.Y.; Burtness, B.; Mehra, R.; Weiss, J.; Berger, R.; Eder, J.P.; Heath, K.; McClanahan, T.; Lunceford, J.; Gause, C.; et al. Safety and clinical activity of pembrolizumab for treatment of recurrent or metastatic squamous cell carcinoma of the head and neck (KEYNOTE-012): An open-label, multicentre, phase 1b trial. Lancet Oncol. 2016, 17, 956–965. [Google Scholar] [CrossRef]
  26. Cohen, E.E.W.; Soulières, D.; Le Tourneau, C.; Dinis, J.; Licitra, L.; Ahn, M.J.; Soria, A.; Machiels, J.P.; Mach, N.; Mehra, R.; et al. Pembrolizumab versus methotrexate, docetaxel, or cetuximab for recurrent or metastatic head-and-neck squamous cell carcinoma (KEYNOTE-040): A randomised, open-label, phase 3 study. Lancet 2019, 393, 156–167. [Google Scholar] [CrossRef]
  27. Burtness, B.; Harrington, K.J.; Greil, R.; Soulières, D.; Tahara, M.; de Castro, G., Jr.; Psyrri, A.; Basté, N.; Neupane, P.; Bratland, Å.; et al. Pembrolizumab alone or with chemotherapy versus cetuximab with chemotherapy for recurrent or metastatic squamous cell carcinoma of the head and neck (KEYNOTE-048): A randomised, open-label, phase 3 study. Lancet 2019, 394, 1915–1928. [Google Scholar] [CrossRef]
  28. Ribas, A.; Wolchok, J.D. Cancer immunotherapy using checkpoint blockade. Science 2018, 359, 1350–1355. [Google Scholar] [CrossRef] [Green Version]
  29. Zandberg, D.P.; Algazi, A.P.; Jimeno, A.; Good, J.S.; Fayette, J.; Bouganim, N.; Ready, N.E.; Clement, P.M.; Even, C.; Jang, R.W.; et al. Durvalumab for recurrent or metastatic head and neck squamous cell carcinoma: Results from a single-arm, phase II study in patients with ≥25% tumour cell PD-L1 expression who have progressed on platinum-based chemotherapy. Eur. J. Cancer 2019, 107, 142–152. [Google Scholar] [CrossRef] [Green Version]
  30. Jamieson, L.; Forster, M.D.; Zaki, K.; Mithra, S.; Alli, H.; O’Connor, A.; Patel, A.; Wong, I.C.K.; Chambers, P. Immunotherapy and associated immune-related adverse events at a large UK centre: A mixed methods study. BMC Cancer 2020, 20, 743. [Google Scholar] [CrossRef]
  31. Wang, D.Y.; Salem, J.E.; Cohen, J.V.; Chandra, S.; Menzer, C.; Ye, F.; Zhao, S.; Das, S.; Beckermann, K.E.; Ha, L.; et al. Fatal Toxic Effects Associated with Immune Checkpoint Inhibitors: A Systematic Review and Meta-analysis. JAMA Oncol. 2018, 4, 1721–1728. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  32. Kessler, R.; Pandruvada, S. Immune-related adverse events following checkpoint inhibitor treatment in head and neck cancers: A comprehensive review. Oral Oncol. Rep. 2023, 6, 100036. [Google Scholar] [CrossRef]
  33. Wu, X.; Gu, Z.; Chen, Y.; Chen, B.; Chen, W.; Weng, L.; Liu, X. Application of PD-1 blockade in cancer immunotherapy. Comput. Struct. Biotechnol. J. 2019, 17, 661–674. [Google Scholar] [CrossRef] [PubMed]
  34. Park, R.; Lopes, L.; Saeed, A. Anti-PD-1/L1-associated immune-related adverse events as harbinger of favorable clinical outcome: Systematic review and meta-analysis. Clin. Transl. Oncol. 2021, 23, 100–109. [Google Scholar] [CrossRef] [PubMed]
  35. Griewing, L.M.; Schweizer, C.; Schubert, P.; Rutzner, S.; Eckstein, M.; Frey, B.; Haderlein, M.; Weissmann, T.; Semrau, S.; Gostian, A.-O. Questionnaire-based detection of immune-related adverse events in cancer patients treated with PD-1/PD-L1 immune checkpoint inhibitors. BMC Cancer 2021, 21, 314. [Google Scholar] [CrossRef]
  36. Schweizer, C.; Schubert, P.; Rutzner, S.; Eckstein, M.; Haderlein, M.; Lettmaier, S.; Semrau, S.; Gostian, A.-O.; Frey, B.; Gaipl, U.S. Prospective evaluation of the prognostic value of immune-related adverse events in patients with non-melanoma solid tumour treated with PD-1/PD-L1 inhibitors alone and in combination with radiotherapy. Eur. J. Cancer 2020, 140, 55–62. [Google Scholar] [CrossRef]
  37. Matsuki, T.; Okamoto, I.; Fushimi, C.; Takahashi, H.; Okada, T.; Kondo, T.; Sato, H.; Ito, T.; Tokashiki, K.; Tsukahara, K. Real-world, long-term outcomes of nivolumab therapy for recurrent or metastatic squamous cell carcinoma of the head and neck and impact of the magnitude of best overall response: A retrospective multicenter study of 88 patients. Cancers 2020, 12, 3427. [Google Scholar] [CrossRef]
  38. Les, I.; Martínez, M.; Pérez-Francisco, I.; Cabero, M.; Teijeira, L.; Arrazubi, V.; Torrego, N.; Campillo-Calatayud, A.; Elejalde, I.; Kochan, G.; et al. Predictive Biomarkers for Checkpoint Inhibitor Immune-Related Adverse Events. Cancers 2023, 15, 1629. [Google Scholar] [CrossRef]
  39. O’Donnell, J.S.; Teng, M.W.L.; Smyth, M.J. Cancer immunoediting and resistance to T cell-based immunotherapy. Nat. Rev. Clin. Oncol. 2019, 16, 151–167. [Google Scholar] [CrossRef]
  40. Masteller, E.L.; Chuang, E.; Mullen, A.C.; Reiner, S.L.; Thompson, C.B. Structural analysis of CTLA-4 function in vivo. J. Immunol. 2000, 164, 5319–5327. [Google Scholar] [CrossRef]
  41. Buchbinder, E.I.; Desai, A. CTLA-4 and PD-1 Pathways: Similarities, Differences, and Implications of Their Inhibition. Am. J. Clin. Oncol. 2016, 39, 98–106. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  42. Qiao, Y.; Liu, C.; Zhang, X.; Zhou, Q.; Li, Y.; Xu, Y.; Gao, Z.; Xu, Y.; Kong, L.; Yang, A.; et al. PD-L2 based immune signature confers poor prognosis in HNSCC. Oncoimmunology 2021, 10, 1947569. [Google Scholar] [CrossRef] [PubMed]
  43. Wherry, E.J. T cell exhaustion. Nat. Immunol. 2011, 12, 492–499. [Google Scholar] [CrossRef] [PubMed]
  44. Hino, R.; Kabashima, K.; Kato, Y.; Yagi, H.; Nakamura, M.; Honjo, T.; Okazaki, T.; Tokura, Y. Tumor cell expression of programmed cell death-1 ligand 1 is a prognostic factor for malignant melanoma. Cancer 2010, 116, 1757–1766. [Google Scholar] [CrossRef]
  45. Rasmussen, M.; Durhuus, J.A.; Nilbert, M.; Andersen, O.; Therkildsen, C. Response to Immune Checkpoint Inhibitors Is Affected by Deregulations in the Antigen Presentation Machinery: A Systematic Review and Meta-Analysis. J. Clin. Med. 2023, 12, 329. [Google Scholar] [CrossRef]
  46. De Sousa Linhares, A.; Leitner, J.; Grabmeier-Pfistershammer, K.; Steinberger, P. Not All Immune Checkpoints Are Created Equal. Front. Immunol. 2018, 9, 1909. [Google Scholar] [CrossRef] [Green Version]
  47. Haibe, Y.; El Husseini, Z.; El Sayed, R.; Shamseddine, A. Resisting Resistance to Immune Checkpoint Therapy: A Systematic Review. Int. J. Mol. Sci. 2020, 21, 6176. [Google Scholar] [CrossRef]
  48. Mouw, K.W.; Goldberg, M.S.; Konstantinopoulos, P.A.; D’Andrea, A.D. DNA Damage and Repair Biomarkers of Immunotherapy Response. Cancer Discov. 2017, 7, 675–693. [Google Scholar] [CrossRef] [Green Version]
  49. García-Aranda, M.; Redondo, M. Targeting Protein Kinases to Enhance the Response to anti-PD-1/PD-L1 Immunotherapy. Int. J. Mol. Sci. 2019, 20, 2296. [Google Scholar] [CrossRef] [Green Version]
  50. Ohue, Y.; Nishikawa, H. Regulatory T (Treg) cells in cancer: Can Treg cells be a new therapeutic target? Cancer Sci. 2019, 110, 2080–2089. [Google Scholar] [CrossRef] [Green Version]
  51. Yan, M.; Zheng, M.; Niu, R.; Yang, X.; Tian, S.; Fan, L.; Li, Y.; Zhang, S. Roles of tumor-associated neutrophils in tumor metastasis and its clinical applications. Front. Cell Dev. Biol. 2022, 10, 938289. [Google Scholar] [CrossRef] [PubMed]
  52. Xiao, M.; He, J.; Yin, L.; Chen, X.; Zu, X.; Shen, Y. Tumor-Associated Macrophages: Critical Players in Drug Resistance of Breast Cancer. Front. Immunol. 2021, 12, 799428. [Google Scholar] [CrossRef] [PubMed]
  53. Ma, T.; Renz, B.W.; Ilmer, M.; Koch, D.; Yang, Y.; Werner, J.; Bazhin, A.V. Myeloid-Derived Suppressor Cells in Solid Tumors. Cells 2022, 11, 310. [Google Scholar] [CrossRef] [PubMed]
  54. Martin-Orozco, E.; Sanchez-Fernandez, A.; Ortiz-Parra, I.; Ayala-San Nicolas, M. WNT Signaling in Tumors: The Way to Evade Drugs and Immunity. Front. Immunol. 2019, 10, 2854. [Google Scholar] [CrossRef] [Green Version]
  55. Berraondo, P.; Sanmamed, M.F.; Ochoa, M.C.; Etxeberria, I.; Aznar, M.A.; Pérez-Gracia, J.L.; Rodríguez-Ruiz, M.E.; Ponz-Sarvise, M.; Castañón, E.; Melero, I. Cytokines in clinical cancer immunotherapy. Br. J. Cancer 2019, 120, 6–15. [Google Scholar] [CrossRef] [Green Version]
  56. Zhong, W.; Wang, Q.; Shen, X.; Du, J. The emerging role of neutrophil extracellular traps in cancer: From lab to ward. Front. Oncol. 2023, 13, 1163802. [Google Scholar] [CrossRef]
  57. Hodi, F.S.; O’Day, S.J.; McDermott, D.F.; Weber, R.W.; Sosman, J.A.; Haanen, J.B.; Gonzalez, R.; Robert, C.; Schadendorf, D.; Hassel, J.C.; et al. Improved Survival with Ipilimumab in Patients with Metastatic Melanoma. N. Engl. J. Med. 2010, 363, 711–723. [Google Scholar] [CrossRef]
  58. Mehra, R.; Seiwert, T.Y.; Gupta, S.; Weiss, J.; Gluck, I.; Eder, J.P.; Burtness, B.; Tahara, M.; Keam, B.; Kang, H.; et al. Efficacy and safety of pembrolizumab in recurrent/metastatic head and neck squamous cell carcinoma: Pooled analyses after long-term follow-up in KEYNOTE-012. Br. J. Cancer 2018, 119, 153–159. [Google Scholar] [CrossRef]
  59. Noji, R.; Tohyama, K.; Kugimoto, T.; Kuroshima, T.; Hirai, H.; Tomioka, H.; Michi, Y.; Tasaki, A.; Ohno, K.; Ariizumi, Y.; et al. Comprehensive Genomic Profiling Reveals Clinical Associations in Response to Immune Therapy in Head and Neck Cancer. Cancers 2022, 14, 3476. [Google Scholar] [CrossRef]
  60. Ayers, M.; Lunceford, J.; Nebozhyn, M.; Murphy, E.; Loboda, A.; Kaufman, D.R.; Albright, A.; Cheng, J.D.; Kang, S.P.; Shankaran, V.; et al. IFN-γ-related mRNA profile predicts clinical response to PD-1 blockade. J. Clin. Investig. 2017, 127, 2930–2940. [Google Scholar] [CrossRef]
  61. Jing, F.; Wang, J.; Zhou, L.; Ning, Y.; Xu, S.; Zhu, Y. Bioinformatics analysis of the role of CXC ligands in the microenvironment of head and neck tumor. Aging 2021, 13, 17789–17817. [Google Scholar] [CrossRef] [PubMed]
  62. de Ruiter, E.J.; Ooft, M.L.; Devriese, L.A.; Willems, S.M. The prognostic role of tumor infiltrating T-lymphocytes in squamous cell carcinoma of the head and neck: A systematic review and meta-analysis. Oncoimmunology 2017, 6, e1356148. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  63. Xu, Q.; Wang, C.; Yuan, X.; Feng, Z.; Han, Z. Prognostic Value of Tumor-Infiltrating Lymphocytes for Patients with Head and Neck Squamous Cell Carcinoma. Transl. Oncol. 2017, 10, 10–16. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  64. Daud, A.I.; Loo, K.; Pauli, M.L.; Sanchez-Rodriguez, R.; Sandoval, P.M.; Taravati, K.; Tsai, K.; Nosrati, A.; Nardo, L.; Alvarado, M.D.; et al. Tumor immune profiling predicts response to anti-PD-1 therapy in human melanoma. J. Clin. Investig. 2016, 126, 3447–3452. [Google Scholar] [CrossRef]
  65. Kao, H.F.; Liao, B.C.; Huang, Y.L.; Huang, H.C.; Chen, C.N.; Chen, T.C.; Hong, Y.J.; Chan, C.Y.; Chia, J.S.; Hong, R.L. Afatinib and Pembrolizumab for Recurrent or Metastatic Head and Neck Squamous Cell Carcinoma (ALPHA Study): A Phase II Study with Biomarker Analysis. Clin. Cancer Res. 2022, 28, 1560–1571. [Google Scholar] [CrossRef] [PubMed]
  66. Mascarella, M.A.; Mannard, E.; Silva, S.D.; Zeitouni, A. Neutrophil-to-lymphocyte ratio in head and neck cancer prognosis: A systematic review and meta-analysis. Head Neck 2018, 40, 1091–1100. [Google Scholar] [CrossRef]
  67. Zhang, L.; Yu, D. Exosomes in cancer development, metastasis, and immunity. Biochim. Biophys. Acta Rev. Cancer 2019, 1871, 455–468. [Google Scholar] [CrossRef]
  68. Chen, Y.; Li, Z.Y.; Zhou, G.Q.; Sun, Y. An Immune-Related Gene Prognostic Index for Head and Neck Squamous Cell Carcinoma. Clin. Cancer Res. 2021, 27, 330–341. [Google Scholar] [CrossRef]
  69. Litchfield, K.; Reading, J.L.; Puttick, C.; Thakkar, K.; Abbosh, C.; Bentham, R.; Watkins, T.B.K.; Rosenthal, R.; Biswas, D.; Rowan, A.; et al. Meta-analysis of tumor- and T cell-intrinsic mechanisms of sensitization to checkpoint inhibition. Cell 2021, 184, 596–614.e14. [Google Scholar] [CrossRef]
  70. Argiris, A.; Harrington, K.; Tahara, M.; Ferris, R.; Gillison, M.; Fayette, J.; Daste, A.; Koralewski, P.; Nin, R.M.; Saba, N. LBA36 Nivolumab (N)+ ipilimumab (I) vs EXTREME as first-line (1L) treatment (tx) for recurrent/metastatic squamous cell carcinoma of the head and neck (R/M SCCHN): Final results of CheckMate 651. Ann. Oncol. 2021, 32, S1310–S1311. [Google Scholar] [CrossRef]
  71. Argiris, A.; Gillison, M.; Ferris, R.; Harrington, K.; Sanchez, T.; Baudelet, C.; Geese, W.; Shaw, J.; Haddad, R. A randomized, open-label, phase 3 study of nivolumab in combination with ipilimumab vs extreme regimen (cetuximab + cisplatin/carboplatin + fluorouracil) as first-line therapy in patients with recurrent or metastatic squamous cell carcinoma of the head and neck-CheckMate 651. Ann. Oncol. 2016, 27, vi350. [Google Scholar]
  72. Seiwert, T.Y.; Weiss, J.; Baxi, S.S.; Ahn, M.-J.; Fayette, J.; Gillison, M.L.; Machiels, J.-P.H.; Takahashi, S.; Melillo, G.; Franks, A. A phase 3, randomized, open-label study of first-line durvalumab (MEDI4736) ± tremelimumab versus standard of care (SoC; EXTREME regimen) in recurrent/metastatic (R/M) SCCHN: KESTREL. J. Clin. Oncol. 2016, 34, TPS6101. [Google Scholar] [CrossRef]
  73. Harrington, K.J.; Ferris, R.L.; Gillison, M.; Tahara, M.; Argiris, A.; Fayette, J.; Schenker, M.; Bratland, Å.; Walker, J.W.T.; Grell, P.; et al. Efficacy and Safety of Nivolumab Plus Ipilimumab vs Nivolumab Alone for Treatment of Recurrent or Metastatic Squamous Cell Carcinoma of the Head and Neck: The Phase 2 CheckMate 714 Randomized Clinical Trial. JAMA Oncol. 2023, 9, 779–789. [Google Scholar] [CrossRef] [PubMed]
  74. Ferris, R.L.; Haddad, R.; Even, C.; Tahara, M.; Dvorkin, M.; Ciuleanu, T.E.; Clement, P.M.; Mesia, R.; Kutukova, S.; Zholudeva, L.; et al. Durvalumab with or without tremelimumab in patients with recurrent or metastatic head and neck squamous cell carcinoma: EAGLE, a randomized, open-label phase III study. Ann. Oncol. 2020, 31, 942–950. [Google Scholar] [CrossRef] [PubMed]
  75. Licitra, L.F.; Haddad, R.I.; Even, C.; Tahara, M.; Dvorkin, M.; Ciuleanu, T.-E.; Clement, P.M.; Mesia, R.; Kutukova, S.I.; Zholudeva, L. EAGLE: A phase 3, randomized, open-label study of durvalumab (D) with or without tremelimumab (T) in patients (pts) with recurrent or metastatic head and neck squamous cell carcinoma (R/M HNSCC). J. Clin. Oncol. 2019, 37, 6012. [Google Scholar] [CrossRef]
  76. Patel, S.P.; Othus, M.; Chen, Y.; Wright, G.P., Jr.; Yost, K.J.; Hyngstrom, J.R.; Hu-Lieskovan, S.; Lao, C.D.; Fecher, L.A.; Truong, T.G.; et al. Neoadjuvant-Adjuvant or Adjuvant-Only Pembrolizumab in Advanced Melanoma. N. Engl. J. Med. 2023, 388, 813–823. [Google Scholar] [CrossRef] [PubMed]
  77. Chocarro, L.; Bocanegra, A.; Blanco, E.; Fernández-Rubio, L.; Arasanz, H.; Echaide, M.; Garnica, M.; Ramos, P.; Piñeiro-Hermida, S.; Vera, R.; et al. Cutting-Edge: Preclinical and Clinical Development of the First Approved Lag-3 Inhibitor. Cells 2022, 11, 2351. [Google Scholar] [CrossRef]
  78. Albrecht, L.J.; Livingstone, E.; Zimmer, L.; Schadendorf, D. The Latest Option: Nivolumab and Relatlimab in Advanced Melanoma. Curr. Oncol. Rep. 2023, 25, 647–657. [Google Scholar] [CrossRef]
  79. Liu, J.F.; Wu, L.; Yang, L.L.; Deng, W.W.; Mao, L.; Wu, H.; Zhang, W.F.; Sun, Z.J. Blockade of TIM3 relieves immunosuppression through reducing regulatory T cells in head and neck cancer. J. Exp. Clin. Cancer Res. 2018, 37, 44. [Google Scholar] [CrossRef] [Green Version]
  80. Mandapathil, M.; Boduc, M.; Roessler, M.; Güldner, C.; Walliczek-Dworschak, U.; Mandic, R. Ectonucleotidase CD39 expression in regional metastases in head and neck cancer. Acta Oto-Laryngol. 2018, 138, 428–432. [Google Scholar] [CrossRef]
  81. Cohen, E.E.; Hong, D.S.; Wise Draper, T.; Nassib William, W.; Schrijvers, D.; Mesia Nin, R.; Scott, M.L.; Lyne, P.; Mugundu, G.; McCoon, P.; et al. Phase 1b/2 Study (SCORES) assessing safety, tolerability, and prelominary anti-tumor activity of durvalumab plus AZD9150 or AZD5069 in patients with advanced solid malignancies and squamous cell carcinoma of the head and neck (SCCHN). Ann. Oncol. 2017, 28, V403. [Google Scholar] [CrossRef] [Green Version]
  82. Harrington, K.J.; Brody, J.; Ingham, M.; Strauss, J.; Cemerski, S.; Wang, M.; Tse, A.; Khilnani, A.; Marabelle, A.; Golan, T. Preliminary results of the first-in-human (FIH) study of MK-1454, an agonist of stimulator of interferon genes (STING), as monotherapy or in combination with pembrolizumab (pembro) in patients with advanced solid tumours of lymphomas. Ann. Oncol. 2018, 29, viii712. [Google Scholar] [CrossRef]
  83. Yarchoan, M.; Powderly, J.D.; Bastos, B.R.; Karasic, T.B.; Crysler, O.V.; Munster, P.N.; McKean, M.; Emens, L.A.; Saenger, Y.M.; Ged, Y.; et al. A phase 1 study of TPST-1120 as a single agent and in combination with nivolumab in subjects with advanced solid tumors. J. Clin. Oncol. 2022, 40, 3005. [Google Scholar] [CrossRef]
  84. Gui, L.; He, X.; Yang, J.; Liu, P.; Qin, Y.; Shi, Y.K. 230MO Pembrolizumab plus anlotinib as first-line treatment in patients of CPS > 1 with recurrent or metastatic head and neck squamous-cell carcinoma: A prospective phase II study. Ann. Oncol. 2022, 33, S1524. [Google Scholar] [CrossRef]
  85. Cohen, E.E.W.; Licitra, L.F.; Burtness, B.; Fayette, J.; Gauler, T.; Clement, P.M.; Grau, J.J.; Del Campo, J.M.; Mailliez, A.; Haddad, R.I.; et al. Biomarkers predict enhanced clinical outcomes with afatinib versus methotrexate in patients with second-line recurrent and/or metastatic head and neck cancer. Ann. Oncol. 2017, 28, 2526–2532. [Google Scholar] [CrossRef]
  86. Cecchini, M.; Zugazagoitia, J.; Lopez, J.S.; Jäger, D.; Oliva, M.; Ochsenreither, S.; Gambardella, V.; Chung, K.Y.; Longo, F.; Razak, A.R.A.; et al. Initial results from a first-in-human, phase I study of immunomodulatory aryl hydrocarbon receptor (AhR) inhibitor BAY2416964 in patients with advanced solid tumors. J. Clin. Oncol. 2023, 41, 2502. [Google Scholar] [CrossRef]
  87. Zhou, Y.; Chen, J.J. STAT3 plays an important role in DNA replication by turning on WDHD1. Cell Biosci. 2021, 11, 10. [Google Scholar] [CrossRef]
  88. Xiong, A.; Yang, Z.; Shen, Y.; Zhou, J.; Shen, Q. Transcription Factor STAT3 as a Novel Molecular Target for Cancer Prevention. Cancers 2014, 6, 926–957. [Google Scholar] [CrossRef] [Green Version]
  89. Greenhill, C.J.; Rose-John, S.; Lissilaa, R.; Ferlin, W.; Ernst, M.; Hertzog, P.J.; Mansell, A.; Jenkins, B.J. IL-6 trans-signaling modulates TLR4-dependent inflammatory responses via STAT3. J. Immunol. 2011, 186, 1199–1208. [Google Scholar] [CrossRef] [Green Version]
  90. Jones, G.W.; McLoughlin, R.M.; Hammond, V.J.; Parker, C.R.; Williams, J.D.; Malhotra, R.; Scheller, J.; Williams, A.S.; Rose-John, S.; Topley, N.; et al. Loss of CD4+ T cell IL-6R expression during inflammation underlines a role for IL-6 trans signaling in the local maintenance of Th17 cells. J. Immunol. 2010, 184, 2130–2139. [Google Scholar] [CrossRef] [Green Version]
  91. Bonner, J.A.; Yang, E.S.; Trummell, H.Q.; Nowsheen, S.; Willey, C.D.; Raisch, K.P. Inhibition of STAT-3 results in greater cetuximab sensitivity in head and neck squamous cell carcinoma. Radiother. Oncol. 2011, 99, 339–343. [Google Scholar] [CrossRef] [PubMed]
  92. Loeuillard, E.; Yang, J.; Buckarma, E.; Wang, J.; Liu, Y.; Conboy, C.; Pavelko, K.D.; Li, Y.; O’Brien, D.; Wang, C.; et al. Targeting tumor-associated macrophages and granulocytic myeloid-derived suppressor cells augments PD-1 blockade in cholangiocarcinoma. J. Clin. Investig. 2020, 130, 5380–5396. [Google Scholar] [CrossRef] [PubMed]
  93. Grauers Wiktorin, H.; Nilsson, M.S.; Kiffin, R.; Sander, F.E.; Lenox, B.; Rydström, A.; Hellstrand, K.; Martner, A. Histamine targets myeloid-derived suppressor cells and improves the anti-tumor efficacy of PD-1/PD-L1 checkpoint blockade. Cancer Immunol. Immunother. 2019, 68, 163–174. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  94. Yu, H.; Lee, H.; Herrmann, A.; Buettner, R.; Jove, R. Revisiting STAT3 signalling in cancer: New and unexpected biological functions. Nat. Rev. Cancer 2014, 14, 736–746. [Google Scholar] [CrossRef]
  95. Radharani, N.N.V.; Yadav, A.S.; Nimma, R.; Kumar, T.V.S.; Bulbule, A.; Chanukuppa, V.; Kumar, D.; Patnaik, S.; Rapole, S.; Kundu, G.C. Tumor-associated macrophage derived IL-6 enriches cancer stem cell population and promotes breast tumor progression via Stat-3 pathway. Cancer Cell Int. 2022, 22, 122. [Google Scholar] [CrossRef]
  96. 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]
  97. Eldesoky, A.; Shouma, A.; Mosaad, Y.; Elhawary, A. Clinical relevance of serum vascular endothelial growth factor and interleukin-6 in patients with colorectal cancer. Saudi J. Gastroenterol. 2011, 17, 170–173. [Google Scholar] [CrossRef]
  98. Riegel, K.; Yurugi, H.; Schlöder, J.; Jonuleit, H.; Kaulich, M.; Kirschner, F.; Arnold-Schild, D.; Tenzer, S.; Schild, H.; Rajalingam, K. ERK5 modulates IL-6 secretion and contributes to tumor-induced immune suppression. Cell Death Dis. 2021, 12, 969. [Google Scholar] [CrossRef]
  99. Chaudhry, A.; Rudra, D.; Treuting, P.; Samstein, R.M.; Liang, Y.; Kas, A.; Rudensky, A.Y. CD4+ regulatory T cells control TH17 responses in a Stat3-dependent manner. Science 2009, 326, 986–991. [Google Scholar] [CrossRef] [Green Version]
  100. Wang, T.; Niu, G.; Kortylewski, M.; Burdelya, L.; Shain, K.; Zhang, S.; Bhattacharya, R.; Gabrilovich, D.; Heller, R.; Coppola, D.; et al. Regulation of the innate and adaptive immune responses by Stat-3 signaling in tumor cells. Nat. Med. 2004, 10, 48–54. [Google Scholar] [CrossRef]
  101. Iwata-Kajihara, T.; Sumimoto, H.; Kawamura, N.; Ueda, R.; Takahashi, T.; Mizuguchi, H.; Miyagishi, M.; Takeda, K.; Kawakami, Y. Enhanced cancer immunotherapy using STAT3-depleted dendritic cells with high Th1-inducing ability and resistance to cancer cell-derived inhibitory factors. J. Immunol. 2011, 187, 27–36. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  102. Herrmann, A.; Kortylewski, M.; Kujawski, M.; Zhang, C.; Reckamp, K.; Armstrong, B.; Wang, L.; Kowolik, C.; Deng, J.; Figlin, R.; et al. Targeting Stat3 in the myeloid compartment drastically improves the in vivo antitumor functions of adoptively transferred T cells. Cancer Res. 2010, 70, 7455–7464. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  103. Yu, H.; Pardoll, D.; Jove, R. STATs in cancer inflammation and immunity: A leading role for STAT3. Nat. Rev. Cancer 2009, 9, 798–809. [Google Scholar] [CrossRef] [PubMed]
  104. Kopf, M.; Baumann, H.; Freer, G.; Freudenberg, M.; Lamers, M.; Kishimoto, T.; Zinkernagel, R.; Bluethmann, H.; Köhler, G. Impaired immune and acute-phase responses in interleukin-6-deficient mice. Nature 1994, 368, 339–342. [Google Scholar] [CrossRef]
  105. Zhao, C.; Zeng, N.; Zhou, X.; Tan, Y.; Wang, Y.; Zhang, J.; Wu, Y.; Zhang, Q. CAA-derived IL-6 induced M2 macrophage polarization by activating STAT3. BMC Cancer 2023, 23, 392. [Google Scholar] [CrossRef]
  106. Zou, S.; Tong, Q.; Liu, B.; Huang, W.; Tian, Y.; Fu, X. Targeting STAT3 in Cancer Immunotherapy. Mol. Cancer 2020, 19, 145. [Google Scholar] [CrossRef]
  107. Valle-Mendiola, A.; Soto-Cruz, I. Energy Metabolism in Cancer: The Roles of STAT3 and STAT5 in the Regulation of Metabolism-Related Genes. Cancers 2020, 12, 124. [Google Scholar] [CrossRef] [Green Version]
  108. Masuda, M.; Ruan, H.Y.; Ito, A.; Nakashima, T.; Toh, S.; Wakasaki, T.; Yasumatsu, R.; Kutratomi, Y.; Komune, S.; Weinstein, I.B. Signal transducers and activators of transcription 3 up-regulates vascular endothelial growth factor production and tumor angiogenesis in head and neck squamous cell carcinoma. Oral Oncol. 2007, 43, 785–790. [Google Scholar] [CrossRef]
  109. Grandis, J.R.; Drenning, S.D.; Zeng, Q.; Watkins, S.C.; Melhem, M.F.; Endo, S.; Johnson, D.E.; Huang, L.; He, Y.; Kim, J.D. Constitutive activation of Stat3 signaling abrogates apoptosis in squamous cell carcinogenesis in vivo. Proc. Natl. Acad. Sci. USA 2000, 97, 4227–4232. [Google Scholar] [CrossRef]
  110. Zhao, Y.; Zhang, J.; Xia, H.; Zhang, B.; Jiang, T.; Wang, J.; Chen, X.; Wang, Y. Stat3 is involved in the motility, metastasis and prognosis in lingual squamous cell carcinoma. Cell Biochem. Funct. 2012, 30, 340–346. [Google Scholar] [CrossRef]
  111. Vermorken, J.B.; Mesia, R.; Rivera, F.; Remenar, E.; Kawecki, A.; Rottey, S.; Erfan, J.; Zabolotnyy, D.; Kienzer, H.R.; Cupissol, D.; et al. Platinum-based chemotherapy plus cetuximab in head and neck cancer. N. Engl. J. Med. 2008, 359, 1116–1127. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  112. Li, X.; Wang, H.; Lu, X.; Di, B. STAT3 blockade with shRNA enhances radiosensitivity in Hep-2 human laryngeal squamous carcinoma cells. Oncol. Rep. 2010, 23, 345–353. [Google Scholar] [CrossRef] [PubMed]
  113. Zhao, C.; Li, H.; Lin, H.J.; Yang, S.; Lin, J.; Liang, G. Feedback Activation of STAT3 as a Cancer Drug-Resistance Mechanism. Trends Pharmacol. Sci. 2016, 37, 47–61. [Google Scholar] [CrossRef] [PubMed]
  114. Lin, S.; Yang, L.; Yao, Y.; Xu, L.; Xiang, Y.; Zhao, H.; Wang, L.; Zuo, Z.; Huang, X.; Zhao, C. Flubendazole demonstrates valid antitumor effects by inhibiting STAT3 and activating autophagy. J. Exp. Clin. Cancer Res. 2019, 38, 293. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  115. Spagnuolo, P.A.; Hu, J.; Hurren, R.; Wang, X.; Gronda, M.; Sukhai, M.A.; Di Meo, A.; Boss, J.; Ashali, I.; Beheshti Zavareh, R.; et al. The antihelmintic flubendazole inhibits microtubule function through a mechanism distinct from Vinca alkaloids and displays preclinical activity in leukemia and myeloma. Blood 2010, 115, 4824–4833. [Google Scholar] [CrossRef] [Green Version]
  116. Čáňová, K.; Rozkydalová, L.; Vokurková, D.; Rudolf, E. Flubendazole induces mitotic catastrophe and apoptosis in melanoma cells. Toxicol. Vitr. 2018, 46, 313–322. [Google Scholar] [CrossRef]
  117. Tao, J.; Zhao, H.; Xie, X.; Luo, M.; Gao, Z.; Sun, H.; Huang, Z. The anthelmintic drug flubendazole induces cell apoptosis and inhibits NF-κB signaling in esophageal squamous cell carcinoma. OncoTargets Ther. 2019, 12, 471–478. [Google Scholar] [CrossRef] [Green Version]
  118. Michaelis, M.; Agha, B.; Rothweiler, F.; Löschmann, N.; Voges, Y.; Mittelbronn, M.; Starzetz, T.; Harter, P.N.; Abhari, B.A.; Fulda, S.; et al. Identification of flubendazole as potential anti-neuroblastoma compound in a large cell line screen. Sci. Rep. 2015, 5, 8202. [Google Scholar] [CrossRef] [Green Version]
  119. Hanušová, V.; Skálová, L.; Králová, V.; Matoušková, P. The Effect of Flubendazole on Adhesion and Migration in SW480 and SW620 Colon Cancer Cells. Anticancer Agents Med. Chem. 2018, 18, 837–846. [Google Scholar] [CrossRef]
  120. Králová, V.; Hanušová, V.; Rudolf, E.; Čáňová, K.; Skálová, L. Flubendazole induces mitotic catastrophe and senescence in colon cancer cells in vitro. J. Pharm. Pharmacol. 2016, 68, 208–218. [Google Scholar] [CrossRef]
  121. Raisová Stuchlíková, L.; Králová, V.; Lněničková, K.; Zárybnický, T.; Matoušková, P.; Hanušová, V.; Ambrož, M.; Šubrt, Z.; Skálová, L. The metabolism of flubendazole in human liver and cancer cell lines. Drug Test. Anal. 2018, 10, 1139–1146. [Google Scholar] [CrossRef] [PubMed]
  122. Kim, Y.J.; Sung, D.; Oh, E.; Cho, Y.; Cho, T.M.; Farrand, L.; Seo, J.H.; Kim, J.Y. Flubendazole overcomes trastuzumab resistance by targeting cancer stem-like properties and HER2 signaling in HER2-positive breast cancer. Cancer Lett. 2018, 412, 118–130. [Google Scholar] [CrossRef] [PubMed]
  123. Oh, E.; Kim, Y.J.; An, H.; Sung, D.; Cho, T.M.; Farrand, L.; Jang, S.; Seo, J.H.; Kim, J.Y. Flubendazole elicits anti-metastatic effects in triple-negative breast cancer via STAT3 inhibition. Int. J. Cancer 2018, 143, 1978–1993. [Google Scholar] [CrossRef] [Green Version]
  124. Hou, Z.J.; Luo, X.; Zhang, W.; Peng, F.; Cui, B.; Wu, S.J.; Zheng, F.M.; Xu, J.; Xu, L.Z.; Long, Z.J.; et al. Flubendazole, FDA-approved anthelmintic, targets breast cancer stem-like cells. Oncotarget 2015, 6, 6326–6340. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  125. Li, Y.; Acharya, G.; Elahy, M.; Xin, H.; Khachigian, L.M. The anthelmintic flubendazole blocks human melanoma growth and metastasis and suppresses programmed cell death protein-1 and myeloid-derived suppressor cell accumulation. Cancer Lett. 2019, 459, 268–276. [Google Scholar] [CrossRef] [PubMed]
  126. Li, Y.; Wu, B.; Hossain, M.J.; Quagliata, L.; O’Meara, C.; Wilkins, M.R.; Corley, S.; Khachigian, L.M. Flubendazole inhibits PD-1 and suppresses melanoma growth in immunocompetent mice. J. Transl. Med. 2023, 21, 467. Available online: https://translational-medicine.biomedcentral.com/articles/10.1186/s12967-023-04289-y (accessed on 16 July 2023). [CrossRef]
  127. Pillaiyar, T.; Meenakshisundaram, S.; Manickam, M.; Sankaranarayanan, M. A medicinal chemistry perspective of drug repositioning: Recent advances and challenges in drug discovery. Eur. J. Med. Chem. 2020, 195, 112275. [Google Scholar] [CrossRef]
  128. Tutrone, W.D.; Saini, R.; Caglar, S.; Weinberg, J.M.; Crespo, J. Topical therapy for actinic keratoses, I: 5-Fluorouracil and imiquimod. Cutis 2003, 71, 365–370. [Google Scholar]
  129. Hamouda, B.; Jamila, Z.; Najet, R.; Slim, T.; Rafiaa, N.; Noureddine, B.; Ahmed, E.M.; Mohamed, F.; Ridha, K.M.; Abderrahman, L. Topical 5-fluorouracil to treat multiple or unresectable facial squamous cell carcinomas in xeroderma pigmentosum. J. Am. Acad. Dermatol. 2001, 44, 1054. [Google Scholar] [CrossRef]
  130. van Ruth, S.; Jansman, F.G.; Sanders, C.J. Total body topical 5-fluorouracil for extensive non-melanoma skin cancer. Pharm. World Sci. 2006, 28, 159–162. [Google Scholar] [CrossRef] [Green Version]
  131. Nishina, T.; Fujita, T.; Yoshizuka, N.; Sugibayashi, K.; Murayama, K.; Kuboki, Y. Safety, tolerability, pharmacokinetics and preliminary antitumour activity of an antisense oligonucleotide targeting STAT3 (danvatirsen) as monotherapy and in combination with durvalumab in Japanese patients with advanced solid malignancies: A phase 1 study. BMJ Open 2022, 12, e055718. [Google Scholar] [CrossRef] [PubMed]
  132. Dhanwani, R.; Takahashi, M.; Sharma, S. Cytosolic sensing of immuno-stimulatory DNA, the enemy within. Curr. Opin. Immunol. 2018, 50, 82–87. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  133. Zhou, L.; Zhang, Y.; Wang, Y.; Zhang, M.; Sun, W.; Dai, T.; Wang, A.; Wu, X.; Zhang, S.; Wang, S.; et al. A Dual Role of Type I Interferons in Antitumor Immunity. Adv. Biosyst. 2020, 4, e1900237. [Google Scholar] [CrossRef] [PubMed]
  134. Zitvogel, L.; Galluzzi, L.; Kepp, O.; Smyth, M.J.; Kroemer, G. Type I interferons in anticancer immunity. Nat. Rev. Immunol. 2015, 15, 405–414. [Google Scholar] [CrossRef] [PubMed]
  135. Flood, B.A.; Higgs, E.F.; Li, S.; Luke, J.J.; Gajewski, T.F. STING pathway agonism as a cancer therapeutic. Immunol. Rev. 2019, 290, 24–38. [Google Scholar] [CrossRef]
  136. Kinkead, H.L.; Hopkins, A.; Lutz, E.; Wu, A.A.; Yarchoan, M.; Cruz, K.; Woolman, S.; Vithayathil, T.; Glickman, L.H.; Ndubaku, C.O.; et al. Combining STING-based neoantigen-targeted vaccine with checkpoint modulators enhances antitumor immunity in murine pancreatic cancer. JCI Insight 2018, 3, e122857. [Google Scholar] [CrossRef] [Green Version]
  137. Ager, C.R.; Reilley, M.J.; Nicholas, C.; Bartkowiak, T.; Jaiswal, A.R.; Curran, M.A. Intratumoral STING Activation with T-cell Checkpoint Modulation Generates Systemic Antitumor Immunity. Cancer Immunol. Res. 2017, 5, 676–684. [Google Scholar] [CrossRef] [Green Version]
  138. Kostadinova, R.; Wahli, W.; Michalik, L. PPARs in diseases: Control mechanisms of inflammation. Curr. Med. Chem. 2005, 12, 2995–3009. [Google Scholar] [CrossRef]
  139. Decara, J.; Rivera, P.; López-Gambero, A.J.; Serrano, A.; Pavón, F.J.; Baixeras, E.; Rodríguez de Fonseca, F.; Suárez, J. Peroxisome Proliferator-Activated Receptors: Experimental Targeting for the Treatment of Inflammatory Bowel Diseases. Front. Pharmacol. 2020, 11, 730. [Google Scholar] [CrossRef]
  140. Yousefnia, S.; Momenzadeh, S.; Seyed Forootan, F.; Ghaedi, K.; Nasr Esfahani, M.H. The influence of peroxisome proliferator-activated receptor γ (PPARγ) ligands on cancer cell tumorigenicity. Gene 2018, 649, 14–22. [Google Scholar] [CrossRef]
  141. Han, L.; Shen, W.J.; Bittner, S.; Kraemer, F.B.; Azhar, S. PPARs: Regulators of metabolism and as therapeutic targets in cardiovascular disease. Part II: PPAR-β/δ and PPAR-γ. Future Cardiol. 2017, 13, 279–296. [Google Scholar] [CrossRef] [PubMed]
  142. Lemmon, M.A.; Schlessinger, J. Cell signaling by receptor tyrosine kinases. Cell 2010, 141, 1117–1134. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  143. Du, Z.; Lovly, C.M. Mechanisms of receptor tyrosine kinase activation in cancer. Mol. Cancer 2018, 17, 58. [Google Scholar] [CrossRef] [PubMed]
  144. Peng, L.; Sun, W.; Chen, L.; Wen, W.P. The Role of Interleukin-33 in Head and Neck Squamous Cell Carcinoma Is Determined by Its Cellular Sources in the Tumor Microenvironment. Front. Oncol. 2020, 10, 588454. [Google Scholar] [CrossRef]
  145. Minutti, C.M.; Drube, S.; Blair, N.; Schwartz, C.; McCrae, J.C.; McKenzie, A.N.; Kamradt, T.; Mokry, M.; Coffer, P.J.; Sibilia, M.; et al. Epidermal Growth Factor Receptor Expression Licenses Type-2 Helper T Cells to Function in a T Cell Receptor-Independent Fashion. Immunity 2017, 47, 710–722.e6. [Google Scholar] [CrossRef] [Green Version]
  146. Wang, Z.; Longo, P.A.; Tarrant, M.K.; Kim, K.; Head, S.; Leahy, D.J.; Cole, P.A. Mechanistic insights into the activation of oncogenic forms of EGF receptor. Nat. Struct. Mol. Biol. 2011, 18, 1388–1393. [Google Scholar] [CrossRef]
  147. Comprehensive genomic characterization of head and neck squamous cell carcinomas. Nature 2015, 517, 576–582. [CrossRef] [Green Version]
  148. Johnson, D.E.; Burtness, B.; Leemans, C.R.; Lui, V.W.Y.; Bauman, J.E.; Grandis, J.R. Head and neck squamous cell carcinoma. Nat. Rev. Dis. Primers 2020, 6, 92. [Google Scholar] [CrossRef]
  149. Byeon, H.K.; Ku, M.; Yang, J. Beyond EGFR inhibition: Multilateral combat strategies to stop the progression of head and neck cancer. Exp. Mol. Med. 2019, 51, 1–14. [Google Scholar] [CrossRef] [Green Version]
  150. Vatte, C.; Al Amri, A.M.; Cyrus, C.; Chathoth, S.; Acharya, S.; Hashim, T.M.; Al Ali, Z.; Alshreadah, S.T.; Alsayyah, A.; Al-Ali, A.K. Tyrosine kinase domain mutations of EGFR gene in head and neck squamous cell carcinoma. OncoTargets Ther. 2017, 10, 1527–1533. [Google Scholar] [CrossRef] [Green Version]
  151. Stransky, N.; Cerami, E.; Schalm, S.; Kim, J.L.; Lengauer, C. The landscape of kinase fusions in cancer. Nat. Commun. 2014, 5, 4846. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  152. Soulieres, D.; Senzer, N.N.; Vokes, E.E.; Hidalgo, M.; Agarwala, S.S.; Siu, L.L. Multicenter phase II study of erlotinib, an oral epidermal growth factor receptor tyrosine kinase inhibitor, in patients with recurrent or metastatic squamous cell cancer of the head and neck. J. Clin. Oncol. 2004, 22, 77–85. [Google Scholar] [CrossRef] [PubMed]
  153. Stewart, J.S.; Cohen, E.E.; Licitra, L.; Van Herpen, C.M.; Khorprasert, C.; Soulieres, D.; Vodvarka, P.; Rischin, D.; Garin, A.M.; Hirsch, F.R.; et al. Phase III study of gefitinib compared with intravenous methotrexate for recurrent squamous cell carcinoma of the head and neck [corrected]. J. Clin. Oncol. 2009, 27, 1864–1871. [Google Scholar] [CrossRef] [PubMed]
  154. Vermorken, J.B.; Trigo, J.; Hitt, R.; Koralewski, P.; Diaz-Rubio, E.; Rolland, F.; Knecht, R.; Amellal, N.; Schueler, A.; Baselga, J. Open-label, uncontrolled, multicenter phase II study to evaluate the efficacy and toxicity of cetuximab as a single agent in patients with recurrent and/or metastatic squamous cell carcinoma of the head and neck who failed to respond to platinum-based therapy. J. Clin. Oncol. 2007, 25, 2171–2177. [Google Scholar] [CrossRef]
  155. Deng, Z.; Liao, W.; Wei, W.; Zhong, G.; He, C.; Zhang, H.; Liu, Q.; Xu, X.; Liang, J.; Liu, Z. Anlotinib as a promising inhibitor on tumor growth of oral squamous cell carcinoma through cell apoptosis and mitotic catastrophe. Cancer Cell Int. 2021, 21, 37. [Google Scholar] [CrossRef]
  156. Boule, L.A.; Burke, C.G.; Jin, G.B.; Lawrence, B.P. Aryl hydrocarbon receptor signaling modulates antiviral immune responses: Ligand metabolism rather than chemical source is the stronger predictor of outcome. Sci. Rep. 2018, 8, 1826. [Google Scholar] [CrossRef] [Green Version]
  157. Gutiérrez-Vázquez, C.; Quintana, F.J. Regulation of the Immune Response by the Aryl Hydrocarbon Receptor. Immunity 2018, 48, 19–33. [Google Scholar] [CrossRef] [Green Version]
  158. Wang, Z.; Snyder, M.; Kenison, J.E.; Yang, K.; Lara, B.; Lydell, E.; Bennani, K.; Novikov, O.; Federico, A.; Monti, S.; et al. How the AHR Became Important in Cancer: The Role of Chronically Active AHR in Cancer Aggression. Int. J. Mol. Sci. 2021, 22, 387. [Google Scholar] [CrossRef]
  159. Dean, J.W.; Zhou, L. Cell-intrinsic view of the aryl hydrocarbon receptor in tumor immunity. Trends Immunol. 2022, 43, 245–258. [Google Scholar] [CrossRef]
  160. Zeng, W.; Song, Y.; Wang, R.; He, R.; Wang, T. Neutrophil elastase: From mechanisms to therapeutic potential. J. Pharm. Anal. 2023, 13, 355–366. [Google Scholar] [CrossRef]
  161. Fridlender, Z.G.; Sun, J.; Kim, S.; Kapoor, V.; Cheng, G.; Ling, L.; Worthen, G.S.; Albelda, S.M. Polarization of tumor-associated neutrophil phenotype by TGF-beta: “N1” versus “N2” TAN. Cancer Cell 2009, 16, 183–194. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  162. Swierczak, A.; Mouchemore, K.A.; Hamilton, J.A.; Anderson, R.L. Neutrophils: Important contributors to tumor progression and metastasis. Cancer Metastasis Rev. 2015, 34, 735–751. [Google Scholar] [CrossRef] [PubMed]
  163. Mantovani, A. The yin-yang of tumor-associated neutrophils. Cancer Cell 2009, 16, 173–174. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  164. Brinkmann, V.; Reichard, U.; Goosmann, C.; Fauler, B.; Uhlemann, Y.; Weiss, D.S.; Weinrauch, Y.; Zychlinsky, A. Neutrophil extracellular traps kill bacteria. Science 2004, 303, 1532–1535. [Google Scholar] [CrossRef]
  165. Neumann, A.; Berends, E.T.; Nerlich, A.; Molhoek, E.M.; Gallo, R.L.; Meerloo, T.; Nizet, V.; Naim, H.Y.; von Köckritz-Blickwede, M. The antimicrobial peptide LL-37 facilitates the formation of neutrophil extracellular traps. Biochem. J. 2014, 464, 3–11. [Google Scholar] [CrossRef]
  166. Mutua, V.; Gershwin, L.J. A Review of Neutrophil Extracellular Traps (NETs) in Disease: Potential Anti-NETs Therapeutics. Clin. Rev. Allergy Immunol. 2021, 61, 194–211. [Google Scholar] [CrossRef]
  167. Delgado-Rizo, V.; Martínez-Guzmán, M.A.; Iñiguez-Gutierrez, L.; García-Orozco, A.; Alvarado-Navarro, A.; Fafutis-Morris, M. Neutrophil Extracellular Traps and Its Implications in Inflammation: An Overview. Front. Immunol. 2017, 8, 81. [Google Scholar] [CrossRef] [Green Version]
  168. Saitoh, T.; Komano, J.; Saitoh, Y.; Misawa, T.; Takahama, M.; Kozaki, T.; Uehata, T.; Iwasaki, H.; Omori, H.; Yamaoka, S.; et al. Neutrophil extracellular traps mediate a host defense response to human immunodeficiency virus-1. Cell Host Microbe 2012, 12, 109–116. [Google Scholar] [CrossRef] [Green Version]
  169. Muraro, S.P.; De Souza, G.F.; Gallo, S.W.; Da Silva, B.K.; De Oliveira, S.D.; Vinolo, M.A.R.; Saraiva, E.M.; Porto, B.N. Respiratory Syncytial Virus induces the classical ROS-dependent NETosis through PAD-4 and necroptosis pathways activation. Sci. Rep. 2018, 8, 14166. [Google Scholar] [CrossRef] [Green Version]
  170. Narasaraju, T.; Yang, E.; Samy, R.P.; Ng, H.H.; Poh, W.P.; Liew, A.A.; Phoon, M.C.; van Rooijen, N.; Chow, V.T. Excessive neutrophils and neutrophil extracellular traps contribute to acute lung injury of influenza pneumonitis. Am. J. Pathol. 2011, 179, 199–210. [Google Scholar] [CrossRef]
  171. Gwyer Findlay, E.; Currie, S.M.; Davidson, D.J. Cationic host defence peptides: Potential as antiviral therapeutics. BioDrugs 2013, 27, 479–493. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  172. Hoeksema, M.; Tripathi, S.; White, M.; Qi, L.; Taubenberger, J.; van Eijk, M.; Haagsman, H.; Hartshorn, K.L. Arginine-rich histones have strong antiviral activity for influenza A viruses. Innate Immun. 2015, 21, 736–745. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  173. Branzk, N.; Lubojemska, A.; Hardison, S.E.; Wang, Q.; Gutierrez, M.G.; Brown, G.D.; Papayannopoulos, V. Neutrophils sense microbe size and selectively release neutrophil extracellular traps in response to large pathogens. Nat. Immunol. 2014, 15, 1017–1025. [Google Scholar] [CrossRef] [PubMed]
  174. Bruns, S.; Kniemeyer, O.; Hasenberg, M.; Aimanianda, V.; Nietzsche, S.; Thywissen, A.; Jeron, A.; Latgé, J.P.; Brakhage, A.A.; Gunzer, M. Production of extracellular traps against Aspergillus fumigatus in vitro and in infected lung tissue is dependent on invading neutrophils and influenced by hydrophobin RodA. PLoS Pathog. 2010, 6, e1000873. [Google Scholar] [CrossRef] [Green Version]
  175. McCormick, A.; Heesemann, L.; Wagener, J.; Marcos, V.; Hartl, D.; Loeffler, J.; Heesemann, J.; Ebel, F. NETs formed by human neutrophils inhibit growth of the pathogenic mold Aspergillus fumigatus. Microbes Infect. 2010, 12, 928–936. [Google Scholar] [CrossRef]
  176. Baker, V.S.; Imade, G.E.; Molta, N.B.; Tawde, P.; Pam, S.D.; Obadofin, M.O.; Sagay, S.A.; Egah, D.Z.; Iya, D.; Afolabi, B.B.; et al. Cytokine-associated neutrophil extracellular traps and antinuclear antibodies in Plasmodium falciparum infected children under six years of age. Malar. J. 2008, 7, 41. [Google Scholar] [CrossRef] [Green Version]
  177. Abi Abdallah, D.S.; Lin, C.; Ball, C.J.; King, M.R.; Duhamel, G.E.; Denkers, E.Y. Toxoplasma gondii triggers release of human and mouse neutrophil extracellular traps. Infect. Immun. 2012, 80, 768–777. [Google Scholar] [CrossRef] [Green Version]
  178. Guimarães-Costa, A.B.; Nascimento, M.T.; Froment, G.S.; Soares, R.P.; Morgado, F.N.; Conceição-Silva, F.; Saraiva, E.M. Leishmania amazonensis promastigotes induce and are killed by neutrophil extracellular traps. Proc. Natl. Acad. Sci. USA 2009, 106, 6748–6753. [Google Scholar] [CrossRef]
  179. Díaz-Godínez, C.; Carrero, J.C. The state of art of neutrophil extracellular traps in protozoan and helminthic infections. Biosci. Rep. 2019, 39, BSR20180916. [Google Scholar] [CrossRef] [Green Version]
  180. Ekaney, M.L.; Otto, G.P.; Sossdorf, M.; Sponholz, C.; Boehringer, M.; Loesche, W.; Rittirsch, D.; Wilharm, A.; Kurzai, O.; Bauer, M.; et al. Impact of plasma histones in human sepsis and their contribution to cellular injury and inflammation. Crit. Care 2014, 18, 543. [Google Scholar] [CrossRef] [Green Version]
  181. Xu, J.; Zhang, X.; Pelayo, R.; Monestier, M.; Ammollo, C.T.; Semeraro, F.; Taylor, F.B.; Esmon, N.L.; Lupu, F.; Esmon, C.T. Extracellular histones are major mediators of death in sepsis. Nat. Med. 2009, 15, 1318–1321. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  182. Abrams, S.T.; Zhang, N.; Manson, J.; Liu, T.; Dart, C.; Baluwa, F.; Wang, S.S.; Brohi, K.; Kipar, A.; Yu, W.; et al. Circulating histones are mediators of trauma-associated lung injury. Am. J. Respir. Crit. Care Med. 2013, 187, 160–169. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  183. Scozzi, D.; Liao, F.; Krupnick, A.S.; Kreisel, D.; Gelman, A.E. The role of neutrophil extracellular traps in acute lung injury. Front. Immunol. 2022, 13, 953195. [Google Scholar] [CrossRef] [PubMed]
  184. Zhang, F.; Li, Y.; Wu, J.; Zhang, J.; Cao, P.; Sun, Z.; Wang, W. The role of extracellular traps in ischemia reperfusion injury. Front. Immunol. 2022, 13, 1022380. [Google Scholar] [CrossRef]
  185. Njeim, R.; Azar, W.S.; Fares, A.H.; Azar, S.T.; Kfoury Kassouf, H.; Eid, A.A. NETosis contributes to the pathogenesis of diabetes and its complications. J. Mol. Endocrinol. 2020, 65, R65–R76. [Google Scholar] [CrossRef]
  186. Middleton, E.A.; He, X.Y.; Denorme, F.; Campbell, R.A.; Ng, D.; Salvatore, S.P.; Mostyka, M.; Baxter-Stoltzfus, A.; Borczuk, A.C.; Loda, M.; et al. Neutrophil extracellular traps contribute to immunothrombosis in COVID-19 acute respiratory distress syndrome. Blood 2020, 136, 1169–1179. [Google Scholar] [CrossRef]
  187. Wang, C.Y.; Lin, T.T.; Hu, L.; Xu, C.J.; Hu, F.; Wan, L.; Yang, X.; Wu, X.F.; Zhang, X.T.; Li, Y.; et al. Neutrophil extracellular traps as a unique target in the treatment of chemotherapy-induced peripheral neuropathy. eBioMedicine 2023, 90, 104499. [Google Scholar] [CrossRef]
  188. Masucci, M.T.; Minopoli, M.; Del Vecchio, S.; Carriero, M.V. The Emerging Role of Neutrophil Extracellular Traps (NETs) in Tumor Progression and Metastasis. Front. Immunol. 2020, 11, 1749. [Google Scholar] [CrossRef]
  189. Podaza, E.; Sabbione, F.; Risnik, D.; Borge, M.; Almejún, M.B.; Colado, A.; Fernández-Grecco, H.; Cabrejo, M.; Bezares, R.F.; Trevani, A.; et al. Neutrophils from chronic lymphocytic leukemia patients exhibit an increased capacity to release extracellular traps (NETs). Cancer Immunol. Immunother. 2017, 66, 77–89. [Google Scholar] [CrossRef]
  190. Li, B.; Liu, Y.; Hu, T.; Zhang, Y.; Zhang, C.; Li, T.; Wang, C.; Dong, Z.; Novakovic, V.A.; Hu, T.; et al. Neutrophil extracellular traps enhance procoagulant activity in patients with oral squamous cell carcinoma. J. Cancer Res. Clin. Oncol. 2019, 145, 1695–1707. [Google Scholar] [CrossRef]
  191. Li, R.; Zou, X.; Zhu, T.; Xu, H.; Li, X.; Zhu, L. Destruction of Neutrophil Extracellular Traps Promotes the Apoptosis and Inhibits the Invasion of Gastric Cancer Cells by Regulating the Expression of Bcl-2, Bax and NF-κB. OncoTargets Ther. 2020, 13, 5271–5281. [Google Scholar] [CrossRef]
  192. Li, M.; Lin, C.; Deng, H.; Strnad, J.; Bernabei, L.; Vogl, D.T.; Burke, J.J.; Nefedova, Y. A Novel Peptidylarginine Deiminase 4 (PAD4) Inhibitor BMS-P5 Blocks Formation of Neutrophil Extracellular Traps and Delays Progression of Multiple Myeloma. Mol. Cancer Ther. 2020, 19, 1530–1538. [Google Scholar] [CrossRef] [PubMed]
  193. Cristinziano, L.; Modestino, L.; Loffredo, S.; Varricchi, G.; Braile, M.; Ferrara, A.L.; de Paulis, A.; Antonelli, A.; Marone, G.; Galdiero, M.R. Anaplastic Thyroid Cancer Cells Induce the Release of Mitochondrial Extracellular DNA Traps by Viable Neutrophils. J. Immunol. 2020, 204, 1362–1372. [Google Scholar] [CrossRef] [PubMed]
  194. Wu, L.; Saxena, S.; Goel, P.; Prajapati, D.R.; Wang, C.; Singh, R.K. Breast Cancer Cell-Neutrophil Interactions Enhance Neutrophil Survival and Pro-Tumorigenic Activities. Cancers 2020, 12, 2884. [Google Scholar] [CrossRef] [PubMed]
  195. Garley, M.; Jabłońska, E.; Miltyk, W.; Grubczak, K.; Surażyński, A.; Ratajczak-Wrona, W.; Grudzińska, M.; Nowacka, K.H.; Moniuszko, M.; Pałka, J.A.; et al. Cancers Cells in Traps? The Pathways of NETs Formation in Response to OSCC in Humans-A Pilot Study. Cancer Control 2020, 27, 1073274820960473. [Google Scholar] [CrossRef] [PubMed]
  196. Cai, Z.; Zhang, M.; Boafo Kwantwi, L.; Bi, X.; Zhang, C.; Cheng, Z.; Ding, X.; Su, T.; Wang, H.; Wu, Q. Breast cancer cells promote self-migration by secreting interleukin 8 to induce NET formation. Gene 2020, 754, 144902. [Google Scholar] [CrossRef]
  197. Thålin, C.; Lundström, S.; Seignez, C.; Daleskog, M.; Lundström, A.; Henriksson, P.; Helleday, T.; Phillipson, M.; Wallén, H.; Demers, M. Citrullinated histone H3 as a novel prognostic blood marker in patients with advanced cancer. PLoS ONE 2018, 13, e0191231. [Google Scholar] [CrossRef]
  198. Guan, X.; Lu, Y.; Zhu, H.; Yu, S.; Zhao, W.; Chi, X.; Xie, C.; Yin, Z. The Crosstalk Between Cancer Cells and Neutrophils Enhances Hepatocellular Carcinoma Metastasis via Neutrophil Extracellular Traps-Associated Cathepsin G Component: A Potential Therapeutic Target. J. Hepatocell. Carcinoma 2021, 8, 451–465. [Google Scholar] [CrossRef]
  199. Gomes, T.; Várady, C.B.S.; Lourenço, A.L.; Mizurini, D.M.; Rondon, A.M.R.; Leal, A.C.; Gonçalves, B.S.; Bou-Habib, D.C.; Medei, E.; Monteiro, R.Q. IL-1β Blockade Attenuates Thrombosis in a Neutrophil Extracellular Trap-Dependent Breast Cancer Model. Front. Immunol. 2019, 10, 2088. [Google Scholar] [CrossRef] [Green Version]
  200. Guimarães-Bastos, D.; Frony, A.C.; Barja-Fidalgo, C.; Moraes, J.A. Melanoma-derived extracellular vesicles skew neutrophils into a pro-tumor phenotype. J. Leukoc. Biol. 2022, 111, 585–596. [Google Scholar] [CrossRef]
  201. Abdol Razak, N.; Elaskalani, O.; Metharom, P. Pancreatic Cancer-Induced Neutrophil Extracellular Traps: A Potential Contributor to Cancer-Associated Thrombosis. Int. J. Mol. Sci. 2017, 18, 487. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  202. Zhang, Y.; Wang, C.; Yu, M.; Zhao, X.; Du, J.; Li, Y.; Jing, H.; Dong, Z.; Kou, J.; Bi, Y.; et al. Neutrophil extracellular traps induced by activated platelets contribute to procoagulant activity in patients with colorectal cancer. Thromb. Res. 2019, 180, 87–97. [Google Scholar] [CrossRef] [PubMed]
  203. Wang, Z.; Yang, C.; Li, L.; Jin, X.; Zhang, Z.; Zheng, H.; Pan, J.; Shi, L.; Jiang, Z.; Su, K.; et al. Tumor-derived HMGB1 induces CD62Ldim neutrophil polarization and promotes lung metastasis in triple-negative breast cancer. Oncogenesis 2020, 9, 82. [Google Scholar] [CrossRef] [PubMed]
  204. Mittal, D.; Saccheri, F.; Vénéreau, E.; Pusterla, T.; Bianchi, M.E.; Rescigno, M. TLR4-mediated skin carcinogenesis is dependent on immune and radioresistant cells. Embo J. 2010, 29, 2242–2252. [Google Scholar] [CrossRef] [PubMed]
  205. Shinde-Jadhav, S.; Mansure, J.J.; Rayes, R.F.; Marcq, G.; Ayoub, M.; Skowronski, R.; Kool, R.; Bourdeau, F.; Brimo, F.; Spicer, J.; et al. Role of neutrophil extracellular traps in radiation resistance of invasive bladder cancer. Nat. Commun. 2021, 12, 2776. [Google Scholar] [CrossRef]
  206. Li, J.; Xia, Y.; Sun, B.; Zheng, N.; Li, Y.; Pang, X.; Yang, F.; Zhao, X.; Ji, Z.; Yu, H.; et al. Neutrophil extracellular traps induced by the hypoxic microenvironment in gastric cancer augment tumour growth. Cell Commun. Signal. 2023, 21, 86. [Google Scholar] [CrossRef]
  207. Shang, A.; Gu, C.; Zhou, C.; Yang, Y.; Chen, C.; Zeng, B.; Wu, J.; Lu, W.; Wang, W.; Sun, Z.; et al. Exosomal KRAS mutation promotes the formation of tumor-associated neutrophil extracellular traps and causes deterioration of colorectal cancer by inducing IL-8 expression. Cell Commun. Signal. 2020, 18, 52. [Google Scholar] [CrossRef] [Green Version]
  208. Tohme, S.; Yazdani, H.O.; Al-Khafaji, A.B.; Chidi, A.P.; Loughran, P.; Mowen, K.; Wang, Y.; Simmons, R.L.; Huang, H.; Tsung, A. Neutrophil Extracellular Traps Promote the Development and Progression of Liver Metastases after Surgical Stress. Cancer Res. 2016, 76, 1367–1380. [Google Scholar] [CrossRef] [Green Version]
  209. Zha, C.; Meng, X.; Li, L.; Mi, S.; Qian, D.; Li, Z.; Wu, P.; Hu, S.; Zhao, S.; Cai, J.; et al. Neutrophil extracellular traps mediate the crosstalk between glioma progression and the tumor microenvironment via the HMGB1/RAGE/IL-8 axis. Cancer Biol. Med. 2020, 17, 154–168. [Google Scholar] [CrossRef]
  210. Houghton, A.M.; Rzymkiewicz, D.M.; Ji, H.; Gregory, A.D.; Egea, E.E.; Metz, H.E.; Stolz, D.B.; Land, S.R.; Marconcini, L.A.; Kliment, C.R.; et al. Neutrophil elastase-mediated degradation of IRS-1 accelerates lung tumor growth. Nat. Med. 2010, 16, 219–223. [Google Scholar] [CrossRef] [Green Version]
  211. Teijeira, Á.; Garasa, S.; Gato, M.; Alfaro, C.; Migueliz, I.; Cirella, A.; de Andrea, C.; Ochoa, M.C.; Otano, I.; Etxeberria, I.; et al. CXCR1 and CXCR2 Chemokine Receptor Agonists Produced by Tumors Induce Neutrophil Extracellular Traps that Interfere with Immune Cytotoxicity. Immunity 2020, 52, 856–871.e8. [Google Scholar] [CrossRef] [PubMed]
  212. Ireland, A.S.; Oliver, T.G. Neutrophils Create an ImpeNETrable Shield between Tumor and Cytotoxic Immune Cells. Immunity 2020, 52, 729–731. [Google Scholar] [CrossRef] [PubMed]
  213. Kaltenmeier, C.; Yazdani, H.O.; Morder, K.; Geller, D.A.; Simmons, R.L.; Tohme, S. Neutrophil Extracellular Traps Promote T Cell Exhaustion in the Tumor Microenvironment. Front. Immunol. 2021, 12, 785222. [Google Scholar] [CrossRef] [PubMed]
  214. Okeke, E.B.; Uzonna, J.E. The Pivotal Role of Regulatory T Cells in the Regulation of Innate Immune Cells. Front. Immunol. 2019, 10, 680. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  215. Wang, H.; Zhang, H.; Wang, Y.; Brown, Z.J.; Xia, Y.; Huang, Z.; Shen, C.; Hu, Z.; Beane, J.; Ansa-Addo, E.A.; et al. Regulatory T-cell and neutrophil extracellular trap interaction contributes to carcinogenesis in non-alcoholic steatohepatitis. J. Hepatol. 2021, 75, 1271–1283. [Google Scholar] [CrossRef] [PubMed]
  216. Velliou, R.I.; Mitroulis, I.; Chatzigeorgiou, A. Neutrophil extracellular traps contribute to the development of hepatocellular carcinoma in NASH by promoting Treg differentiation. Hepatobiliary Surg. Nutr. 2022, 11, 415–418. [Google Scholar] [CrossRef]
  217. Kimball, A.S.; Obi, A.T.; Diaz, J.A.; Henke, P.K. The Emerging Role of NETs in Venous Thrombosis and Immunothrombosis. Front. Immunol. 2016, 7, 236. [Google Scholar] [CrossRef] [Green Version]
  218. Diaz, J.A.; Fuchs, T.A.; Jackson, T.O.; Kremer Hovinga, J.A.; Lämmle, B.; Henke, P.K.; Myers, D.D., Jr.; Wagner, D.D.; Wakefield, T.W. Plasma DNA is Elevated in Patients with Deep Vein Thrombosis. J. Vasc. Surg. Venous Lymphat. Disord. 2013, 1, 341–348.e1. [Google Scholar] [CrossRef] [Green Version]
  219. Fuchs, T.A.; Brill, A.; Duerschmied, D.; Schatzberg, D.; Monestier, M.; Myers, D.D., Jr.; Wrobleski, S.K.; Wakefield, T.W.; Hartwig, J.H.; Wagner, D.D. Extracellular DNA traps promote thrombosis. Proc. Natl. Acad. Sci. USA 2010, 107, 15880–15885. [Google Scholar] [CrossRef]
  220. Brill, A.; Fuchs, T.A.; Savchenko, A.S.; Thomas, G.M.; Martinod, K.; De Meyer, S.F.; Bhandari, A.A.; Wagner, D.D. Neutrophil extracellular traps promote deep vein thrombosis in mice. J. Thromb. Haemost. 2012, 10, 136–144. [Google Scholar] [CrossRef] [Green Version]
  221. Martinod, K.; Wagner, D.D. Thrombosis: Tangled up in NETs. Blood 2014, 123, 2768–2776. [Google Scholar] [CrossRef] [PubMed]
  222. Wang, S.; Li, Z.; Xu, R. Human Cancer and Platelet Interaction, a Potential Therapeutic Target. Int. J. Mol. Sci. 2018, 19, 1246. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  223. Waldmann, T.A. Cytokines in Cancer Immunotherapy. Cold Spring Harb. Perspect. Biol. 2018, 10, a028472. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  224. Placke, T.; Örgel, M.; Schaller, M.; Jung, G.; Rammensee, H.G.; Kopp, H.G.; Salih, H.R. Platelet-derived MHC class I confers a pseudonormal phenotype to cancer cells that subverts the antitumor reactivity of natural killer immune cells. Cancer Res. 2012, 72, 440–448. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  225. Batu, E.D. Neutrophil-mediated Thrombosis and NETosis in Behçet’s Disease: A Hypothesis. J. Korean Med. Sci. 2020, 35, e213. [Google Scholar] [CrossRef] [PubMed]
  226. Kordbacheh, F.; O’Meara, C.H.; Coupland, L.A.; Lelliott, P.M.; Parish, C.R. Extracellular histones induce erythrocyte fragility and anemia. Blood 2017, 130, 2884–2888. [Google Scholar] [CrossRef] [PubMed]
  227. Zhang, H.; Wang, Y.; Qu, M.; Li, W.; Wu, D.; Cata, J.P.; Miao, C. Neutrophil, neutrophil extracellular traps and endothelial cell dysfunction in sepsis. Clin. Transl. Med. 2023, 13, e1170. [Google Scholar] [CrossRef]
  228. Yang, X.; Li, L.; Liu, J.; Lv, B.; Chen, F. Extracellular histones induce tissue factor expression in vascular endothelial cells via TLR and activation of NF-κB and AP-1. Thromb. Res. 2016, 137, 211–218. [Google Scholar] [CrossRef]
  229. Saffarzadeh, M.; Juenemann, C.; Queisser, M.A.; Lochnit, G.; Barreto, G.; Galuska, S.P.; Lohmeyer, J.; Preissner, K.T. Neutrophil extracellular traps directly induce epithelial and endothelial cell death: A predominant role of histones. PLoS ONE 2012, 7, e32366. [Google Scholar] [CrossRef]
  230. Folco, E.J.; Mawson, T.L.; Vromman, A.; Bernardes-Souza, B.; Franck, G.; Persson, O.; Nakamura, M.; Newton, G.; Luscinskas, F.W.; Libby, P. Neutrophil Extracellular Traps Induce Endothelial Cell Activation and Tissue Factor Production Through Interleukin-1α and Cathepsin G. Arterioscler. Thromb. Vasc. Biol. 2018, 38, 1901–1912. [Google Scholar] [CrossRef] [Green Version]
  231. Cools-Lartigue, J.; Spicer, J.; Najmeh, S.; Ferri, L. Neutrophil extracellular traps in cancer progression. Cell. Mol. Life Sci. 2014, 71, 4179–4194. [Google Scholar] [CrossRef] [PubMed]
  232. Yang, L.; Liu, Q.; Zhang, X.; Liu, X.; Zhou, B.; Chen, J.; Huang, D.; Li, J.; Li, H.; Chen, F.; et al. DNA of neutrophil extracellular traps promotes cancer metastasis via CCDC25. Nature 2020, 583, 133–138. [Google Scholar] [CrossRef] [PubMed]
  233. Dongre, A.; Weinberg, R.A. New insights into the mechanisms of epithelial-mesenchymal transition and implications for cancer. Nat. Rev. Mol. Cell Biol. 2019, 20, 69–84. [Google Scholar] [CrossRef]
  234. Martins-Cardoso, K.; Almeida, V.H.; Bagri, K.M.; Rossi, M.I.D.; Mermelstein, C.S.; König, S.; Monteiro, R.Q. Neutrophil Extracellular Traps (NETs) Promote Pro-Metastatic Phenotype in Human Breast Cancer Cells through Epithelial-Mesenchymal Transition. Cancers 2020, 12, 1542. [Google Scholar] [CrossRef]
  235. Zhu, T.; Zou, X.; Yang, C.; Li, L.; Wang, B.; Li, R.; Li, H.; Xu, Z.; Huang, D.; Wu, Q. Neutrophil extracellular traps promote gastric cancer metastasis by inducing epithelial-mesenchymal transition. Int. J. Mol. Med. 2021, 48, 127. [Google Scholar] [CrossRef] [PubMed]
  236. Albrengues, J.; Shields, M.A.; Ng, D.; Park, C.G.; Ambrico, A.; Poindexter, M.E.; Upadhyay, P.; Uyeminami, D.L.; Pommier, A.; Küttner, V.; et al. Neutrophil extracellular traps produced during inflammation awaken dormant cancer cells in mice. Science 2018, 361, eaao4227. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  237. Jung, H.S.; Gu, J.; Kim, J.E.; Nam, Y.; Song, J.W.; Kim, H.K. Cancer cell-induced neutrophil extracellular traps promote both hypercoagulability and cancer progression. PLoS ONE 2019, 14, e0216055. [Google Scholar] [CrossRef] [Green Version]
  238. Aldabbous, L.; Abdul-Salam, V.; McKinnon, T.; Duluc, L.; Pepke-Zaba, J.; Southwood, M.; Ainscough, A.J.; Hadinnapola, C.; Wilkins, M.R.; Toshner, M.; et al. Neutrophil Extracellular Traps Promote Angiogenesis: Evidence From Vascular Pathology in Pulmonary Hypertension. Arterioscler. Thromb. Vasc. Biol. 2016, 36, 2078–2087. [Google Scholar] [CrossRef] [Green Version]
  239. Yuan, K.; Zheng, J.; Huang, X.; Zhang, Y.; Han, Y.; Hu, R.; Jin, X. Neutrophil extracellular traps promote corneal neovascularization-induced by alkali burn. Int. Immunopharmacol. 2020, 88, 106902. [Google Scholar] [CrossRef]
  240. Meara, C.H.O.; Coupland, L.A.; Kordbacheh, F.; Quah, B.J.C.; Chang, C.W.; Simon Davis, D.A.; Bezos, A.; Browne, A.M.; Freeman, C.; Hammill, D.J.; et al. Neutralizing the pathological effects of extracellular histones with small polyanions. Nat. Commun. 2020, 11, 6408. [Google Scholar] [CrossRef]
  241. Volkov, D.V.; Tetz, G.V.; Rubtsov, Y.P.; Stepanov, A.V.; Gabibov, A.G. Neutrophil Extracellular Traps (NETs): Opportunities for Targeted Therapy. Acta Naturae 2021, 13, 15–23. [Google Scholar] [CrossRef] [PubMed]
  242. Clark, S.R.; Ma, A.C.; Tavener, S.A.; McDonald, B.; Goodarzi, Z.; Kelly, M.M.; Patel, K.D.; Chakrabarti, S.; McAvoy, E.; Sinclair, G.D.; et al. Platelet TLR4 activates neutrophil extracellular traps to ensnare bacteria in septic blood. Nat. Med. 2007, 13, 463–469. [Google Scholar] [CrossRef] [PubMed]
  243. Tilgner, J.; von Trotha, K.T.; Gombert, A.; Jacobs, M.J.; Drechsler, M.; Döring, Y.; Soehnlein, O.; Grommes, J. Aspirin, but Not Tirofiban Displays Protective Effects in Endotoxin Induced Lung Injury. PLoS ONE 2016, 11, e0161218. [Google Scholar] [CrossRef] [Green Version]
  244. Vaguliene, N.; Zemaitis, M.; Lavinskiene, S.; Miliauskas, S.; Sakalauskas, R. Local and systemic neutrophilic inflammation in patients with lung cancer and chronic obstructive pulmonary disease. BMC Immunol. 2013, 14, 36. [Google Scholar] [CrossRef] [Green Version]
  245. Kistowski, M.; Dębski, J.; Karczmarski, J.; Paziewska, A.; Olędzki, J.; Mikula, M.; Ostrowski, J.; Dadlez, M. A Strong Neutrophil Elastase Proteolytic Fingerprint Marks the Carcinoma Tumor Proteome. Mol. Cell. Proteom. 2017, 16, 213–227. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  246. Akizuki, M.; Fukutomi, T.; Takasugi, M.; Takahashi, S.; Sato, T.; Harao, M.; Mizumoto, T.; Yamashita, J. Prognostic significance of immunoreactive neutrophil elastase in human breast cancer: Long-term follow-up results in 313 patients. Neoplasia 2007, 9, 260–264. [Google Scholar] [CrossRef] [Green Version]
  247. Okamoto, M.; Mizuno, R.; Kawada, K.; Itatani, Y.; Kiyasu, Y.; Hanada, K.; Hirata, W.; Nishikawa, Y.; Masui, H.; Sugimoto, N.; et al. Neutrophil Extracellular Traps Promote Metastases of Colorectal Cancers through Activation of ERK Signaling by Releasing Neutrophil Elastase. Int. J. Mol. Sci. 2023, 24, 1118. [Google Scholar] [CrossRef]
  248. Hemmers, S.; Teijaro, J.R.; Arandjelovic, S.; Mowen, K.A. PAD4-mediated neutrophil extracellular trap formation is not required for immunity against influenza infection. PLoS ONE 2011, 6, e22043. [Google Scholar] [CrossRef]
  249. Bruno, A.; Dovizio, M.; Tacconelli, S.; Patrignani, P. Mechanisms of the antitumoural effects of aspirin in the gastrointestinal tract. Best Pract. Res. Clin. Gastroenterol. 2012, 26, e1–e13. [Google Scholar] [CrossRef]
  250. Guillem-Llobat, P.; Dovizio, M.; Bruno, A.; Ricciotti, E.; Cufino, V.; Sacco, A.; Grande, R.; Alberti, S.; Arena, V.; Cirillo, M.; et al. Aspirin prevents colorectal cancer metastasis in mice by splitting the crosstalk between platelets and tumor cells. Oncotarget 2016, 7, 32462–32477. [Google Scholar] [CrossRef]
  251. Patrignani, P.; Patrono, C. Aspirin and Cancer. J. Am. Coll. Cardiol. 2016, 68, 967–976. [Google Scholar] [CrossRef] [PubMed]
  252. Menegazzo, L.; Scattolini, V.; Cappellari, R.; Bonora, B.M.; Albiero, M.; Bortolozzi, M.; Romanato, F.; Ceolotto, G.; Vigili de Kreutzeberg, S.; Avogaro, A.; et al. The antidiabetic drug metformin blunts NETosis in vitro and reduces circulating NETosis biomarkers in vivo. Acta Diabetol. 2018, 55, 593–601. [Google Scholar] [CrossRef]
  253. Wang, G.; Gao, H.; Dai, S.; Li, M.; Gao, Y.; Yin, L.; Zhang, K.; Zhang, J.; Jiang, K.; Miao, Y.; et al. Metformin inhibits neutrophil extracellular traps-promoted pancreatic carcinogenesis in obese mice. Cancer Lett. 2023, 562, 216155. [Google Scholar] [CrossRef] [PubMed]
  254. Yang, L.Y.; Shen, X.T.; Sun, H.T.; Zhu, W.W.; Zhang, J.B.; Lu, L. Neutrophil extracellular traps in hepatocellular carcinoma are enriched in oxidized mitochondrial DNA which is highly pro-inflammatory and pro-metastatic. J. Cancer 2022, 13, 1261–1271. [Google Scholar] [CrossRef]
  255. Chen, C.-J.; Wu, C.-C.; Chang, C.-Y.; Li, J.-R.; Ou, Y.-C.; Chen, W.-Y.; Liao, S.-L.; Wang, J.-D. Metformin Mitigated Obesity-Driven Cancer Aggressiveness in Tumor-Bearing Mice. Int. J. Mol. Sci. 2022, 23, 9134. [Google Scholar] [CrossRef] [PubMed]
  256. Saito, A.; Ohzawa, H.; Kaneko, Y.; Tamura, K.; Futoh, Y.; Takahashi, K.; Kimura, Y.; Kawashima, R.; Miyato, H.; Kitayama, J.; et al. Abstract 1268: The effect of metformin for neutrophil extracellular traps (NETs) on tumor immune microenvironment of colorectal cancer with type 2 diabetes mellitus. Cancer Res. 2023, 83, 1268. [Google Scholar] [CrossRef]
  257. Leppkes, M.; Knopf, J.; Naschberger, E.; Lindemann, A.; Singh, J.; Herrmann, I.; Stürzl, M.; Staats, L.; Mahajan, A.; Schauer, C.; et al. Vascular occlusion by neutrophil extracellular traps in COVID-19. eBioMedicine 2020, 58, 102925. [Google Scholar] [CrossRef]
  258. Cools-Lartigue, J.; Spicer, J.; McDonald, B.; Gowing, S.; Chow, S.; Giannias, B.; Bourdeau, F.; Kubes, P.; Ferri, L. Neutrophil extracellular traps sequester circulating tumor cells and promote metastasis. J. Clin. Investig. 2013, 123, 3446–3458. [Google Scholar] [CrossRef]
  259. Park, J.; Wysocki, R.W.; Amoozgar, Z.; Maiorino, L.; Fein, M.R.; Jorns, J.; Schott, A.F.; Kinugasa-Katayama, Y.; Lee, Y.; Won, N.H.; et al. Cancer cells induce metastasis-supporting neutrophil extracellular DNA traps. Sci. Transl. Med. 2016, 8, 361ra138. [Google Scholar] [CrossRef] [Green Version]
  260. Law, S.M.; Gray, R.D. Neutrophil extracellular traps and the dysfunctional innate immune response of cystic fibrosis lung disease: A review. J. Inflamm. 2017, 14, 29. [Google Scholar] [CrossRef]
Figure 1. ICI and SMI actions within the tumor microenvironment. Whilst ICIs influence cell signaling at cell surface receptors, SMIs can interact with “upstream” intracellular signaling pathways-potentially playing a more effective role in abrogating tumor cell progression. MDSC, myeloid-derived suppressor cells; M2 macrophages, pro-tumorigenic macrophages; STING, stimulator of interferon genes; PPAR-α, peroxisome proliferator-activated receptor-α; AHR, aryl hydrocarbon receptor; STAT3, signal transducer and activator of transcription 3; P, phosphorylation of STAT3. Created with BioRender.com.
Figure 1. ICI and SMI actions within the tumor microenvironment. Whilst ICIs influence cell signaling at cell surface receptors, SMIs can interact with “upstream” intracellular signaling pathways-potentially playing a more effective role in abrogating tumor cell progression. MDSC, myeloid-derived suppressor cells; M2 macrophages, pro-tumorigenic macrophages; STING, stimulator of interferon genes; PPAR-α, peroxisome proliferator-activated receptor-α; AHR, aryl hydrocarbon receptor; STAT3, signal transducer and activator of transcription 3; P, phosphorylation of STAT3. Created with BioRender.com.
Ijms 24 11695 g001
Figure 2. Pro-tumorigenic effects of NETs and developing therapeutic strategies. NETs have been demonstrated to play a role in tumor immunomodulation, camouflage, homeostasis/activation, and metastasis. Therapies under development can be grouped into those that prevent or “modulate” NETosis (the generation of NETs) or “dissolve” the chromatin “web” of the neutrophil extracellular trap, mitigating the ability for NETs to play a pro-tumorigenic role. HMGB-1, high mobility group box-1; KRAS oncogene mutation. Created with BioRender.com.
Figure 2. Pro-tumorigenic effects of NETs and developing therapeutic strategies. NETs have been demonstrated to play a role in tumor immunomodulation, camouflage, homeostasis/activation, and metastasis. Therapies under development can be grouped into those that prevent or “modulate” NETosis (the generation of NETs) or “dissolve” the chromatin “web” of the neutrophil extracellular trap, mitigating the ability for NETs to play a pro-tumorigenic role. HMGB-1, high mobility group box-1; KRAS oncogene mutation. Created with BioRender.com.
Ijms 24 11695 g002
Table 1. Combination ICI Therapy Clinical Trials in HNSCC.
Table 1. Combination ICI Therapy Clinical Trials in HNSCC.
TargetCombinationPhaseTrialIntentOutcome
PD-1, CTLA-4Nivolumab, Ipilimumab3NCT027441570 (CheckMate 651) [71] Combination nivolumab + ipilimumab vs. EXTREME Regime (platinum/5-fluorouracil/cetuximab) for R/M HNSCCFailed endpoint (OS).
No difference between dual ICI blockade and EXTREME arm. Improvement in dual ICI arm if CPS > 20 (ns)
PD-L1, CTLA-4Durvalumab, Tremelimumab3NCT02551159 (KESTRAL) [72]Combination durvalumab + tremelimumab vs. duravalumab monotherapy vs. SOC CT in R/M HNSCCResults pending
PD-1, CTLA-4Nivolumab, Ipilimumab2NCT02823574 (CheckMate 714) [73]Combination nivolumab + ipilimumab vs. nivolumab + ipilimumab placebo in R/M HNSCCFailed ORR and OS endpoints. Subpopulation assessment ongoing.
PD-L1, CTLA-4Durvalumab, Tremelimumab3NCT02369874 (EAGLE) [74,75]Combination durvalumab + tremelimumab vs. durvalumab monotherapy vs. SOC in R/M HNSCCFailed to meet primary OS improvement endpoint
Table 2. SMI Targets and Clinical Trials in HNSCC.
Table 2. SMI Targets and Clinical Trials in HNSCC.
TargetDrugPhaseTrialIntentOutcome
STAT3AZD91501b/2NCT02499328 [81]Combination ASD9150 + MED14736 (duravalumab) vs. MED14736 alone; in platinum refractory recurrent metastatic HNSCCAcceptable toxicity profile.
Combination therapy more effective than PD-L1 monotherapy
STINGMK-14541NCT03010176 [82]Combination MK-1454 (ulveostinag) + pembrilizumab vs. MK-1454 monotherapy; in advanced HNSCCAcceptable toxicity profile.
Combination therapy more effective (DCR 48%) than monotherapy (DCR 20%)
PPAR-αTPST-11201/1b03829436 [83]Combination TPST-1120 + nivolumab vs. TPST-1120 monotherapy; in advanced solid tumors; including HNSCCAcceptable toxicity (several patients suffered Grade 3 Adverse reactions.
Optimal disease response in combination therapy (38%)
RTKsAL38182NCT04999800 [84]Combination AL3818 (analotinib) + pembrolizumab; as a first line therapy for platinum refractory recurrent or metastatic HNSCCManageable side effects.
Encouraging anti-tumor activity.
ORR: 46.7% (7/15) & DCR: 100%
Median PFS & OS not reached (median follow-up: 8.2 months.
RTKsAfatinib3NCT01345682 [85]LUX-Head & Neck 1: second-line afatinib therapy vs. methotrexate for platinum refractory recurrent/metastatic HNSCCn = 483 patients.
Median PFS: afatinib over methotrexate (2.7 months vs. 1.6 months)
Afatinib more effective in all tumor subsets except HPV + OPSCC
AHRBAY24169641NCT04069026 [86]AHR antagonist: safety and tumor response study in advanced HNSCC & nSCLCWell tolerated at all dose regimes.
Initial evaluation of biomarkers shows inhibition of AHR and modulation of immune functions.
Encouraging preliminary anti-tumor activity in heavily pretreated patients.
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

O’Meara, C.H.; Jafri, Z.; Khachigian, L.M. Immune Checkpoint Inhibitors, Small-Molecule Immunotherapies and the Emerging Role of Neutrophil Extracellular Traps in Therapeutic Strategies for Head and Neck Cancer. Int. J. Mol. Sci. 2023, 24, 11695. https://doi.org/10.3390/ijms241411695

AMA Style

O’Meara CH, Jafri Z, Khachigian LM. Immune Checkpoint Inhibitors, Small-Molecule Immunotherapies and the Emerging Role of Neutrophil Extracellular Traps in Therapeutic Strategies for Head and Neck Cancer. International Journal of Molecular Sciences. 2023; 24(14):11695. https://doi.org/10.3390/ijms241411695

Chicago/Turabian Style

O’Meara, Connor H., Zuhayr Jafri, and Levon M. Khachigian. 2023. "Immune Checkpoint Inhibitors, Small-Molecule Immunotherapies and the Emerging Role of Neutrophil Extracellular Traps in Therapeutic Strategies for Head and Neck Cancer" International Journal of Molecular Sciences 24, no. 14: 11695. https://doi.org/10.3390/ijms241411695

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