Next Article in Journal
Spiroleiferthione A and Oleiferthione A: Two Unusual Isothiocyanate-Derived Thioketone Alkaloids from Moringa oleifera Lam. Seeds
Next Article in Special Issue
Advances in Non-Small Cell Lung Cancer (NSCLC) Treatment—A Paradigm Shift in Oncology
Previous Article in Journal
Propolis: A Detailed Insight of Its Anticancer Molecular Mechanisms
Previous Article in Special Issue
Prognostic Value of CXCL12 in Non-Small Cell Lung Cancer Patients Undergoing Tumor Resection
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Targeting Inflammation in Non-Small Cell Lung Cancer through Drug Repurposing

by
Thiviyadarshini Rajasegaran
1,
Chee Wun How
2,
Anoosha Saud
1,
Azhar Ali
3,† and
Jonathan Chee Woei Lim
1,*,†
1
Department of Medicine, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, Serdang 43400, Selangor, Malaysia
2
School of Pharmacy, Monash University Malaysia, Bandar Sunway, Subang Jaya 47500, Selangor, Malaysia
3
Cancer Science Institute Singapore, National University of Singapore, Singapore 117599, Singapore
*
Author to whom correspondence should be addressed.
These authors contributed equally to this work.
Pharmaceuticals 2023, 16(3), 451; https://doi.org/10.3390/ph16030451
Submission received: 13 February 2023 / Revised: 12 March 2023 / Accepted: 14 March 2023 / Published: 16 March 2023

Abstract

:
Lung cancer is the most common cause of cancer-related deaths. Lung cancers can be classified as small-cell (SCLC) or non-small cell (NSCLC). About 84% of all lung cancers are NSCLC and about 16% are SCLC. For the past few years, there have been a lot of new advances in the management of NSCLC in terms of screening, diagnosis and treatment. Unfortunately, most of the NSCLCs are resistant to current treatments and eventually progress to advanced stages. In this perspective, we discuss some of the drugs that can be repurposed to specifically target the inflammatory pathway of NSCLC utilizing its well-defined inflammatory tumor microenvironment. Continuous inflammatory conditions are responsible to induce DNA damage and enhance cell division rate in lung tissues. There are existing anti-inflammatory drugs which were found suitable for repurposing in non-small cell lung carcinoma (NSCLC) treatment and drug modification for delivery via inhalation. Repurposing anti-inflammatory drugs and their delivery through the airway is a promising strategy to treat NSCLC. In this review, suitable drug candidates that can be repurposed to treat inflammation-mediated NSCLC will be comprehensively discussed together with their administration via inhalation from physico-chemical and nanocarrier perspectives.

1. Introduction

According to GLOBOCAN 2020, lung cancer tops the cancer list, with highest fatality rate and accounts for 18% of all cancer deaths worldwide [1]. About 84% of lung cancer cases belongs to non-small cell lung carcinoma (NSCLC) and the remaining 15% belongs to small cell lung carcinoma (SCLC) [2]. NSCLC is categorized into three sub-types: adenocarcinoma, squamous cell carcinoma, and large cell carcinoma. Adenocarcinoma is the major subtype with about 45% of all NSCLC followed by squamous cell carcinoma with 25–30% and the remaining 5–10% is large cell carcinoma subtype [3]. Late diagnosis of disease (at stage III and IV) is the major factor for the poor survival rate in lung cancer patients as the disease has progressed to the metastatic stage. About 92% of patients diagnosed at stage IA1 could survived for 5 years or more compared to 10% of patients diagnosed at stage IV. Furthermore, slight enlargement in tumor size from <1 cm (stage IA1) to >2 cm (stage IA3) could reduce the 5-year survival rate of patients from 92% to 77% [4].
Current NCSLC therapies, including surgery, chemotherapy, and radiotherapy, are insufficient to reduce the high mortality rates. These approaches lack precision and are usually limited by low drug bioavailability due to high first pass metabolism. Furthermore, serious adverse effects occur due to non-specificity where the chemotherapeutics adversely affect healthy cells [5]. To improve the survival of NSCLC patients, personalized medicine is preferred. Recent molecular targeted therapies, such as epidermal growth factor receptor tyrosine kinase inhibitors (EGFR-TKIs), could restrict growth and proliferation of lung tumors with EGFR mutations. On the other hand, targeting ROS could inhibit signaling pathways, such as MAPK/ERK, JAK/STAT, and P13K/AKT/mTOR. Then, targeting BRAF can interfere in cell proliferation and growth [6]. However, this treatment is only effective for a short duration due to subsequent development of acquired drug resistance [7]. Anaplastic lymphoma kinase (ALK) mutation is another example of a successful targeted therapy approach. Crizotinib is an FDA-approved agent that targets tyrosine kinases, such as ALK, c-mesenchymal-epithelial transition (c-MET), and c-ros oncogene 1 (ROS). Crizotinib has shown promising improvement in progression-free survival, and was the first ALK-tyrosine kinase inhibitor approved in the treatment of ALK-rearranged NSCLC [8]. However, treatment with Lorlatinib have indicated a survival rate of only 12 out 37 patients who were ALK positive, while 8 out of 14 patients who were positive for ROS survived [9].
Another targeted approach, immunotherapy, is also currently being applied in NSCLC treatment. Anti-PD-1/PDL-1 inhibitors, such as nivolumab and Atezolizumab, have shown substantive clinical activity in metastatic lung cancer and are approved for first or subsequent lines of therapy. However, only 20% of NSCLC patients showed significant remission and clinical benefit due to development of resistance [10]. Dysregulation of immune balance was found involved in promoting the progression of cancer. Tumor cell vaccines and antigen-specific vaccines have been suggested to shift immune balance in favor of host and elicit an immune response against the antigens. Belagenpumatucel-L is an allogenic tumor vaccine comprising a pool of four irradiated transforming-growth factor (TGF)-β-modified NSCLC lines. A group of randomized patients who had earlier completed chemotherapy within 12 weeks were treated with this vaccine and showed improvement in overall survival by 21 months when compared to the placebo group, which showed improvement of overall survival by 14 months [3]. The goal of these therapies, nevertheless, aims to lengthen progression-free survival instead of prevention. Lifestyle and environmental factors are well known to be tightly associated with lung cancer. Cigarette smoking accounts for 85–90% of lung cancer, depending on the extent of smoking and exposure to other carcinogenic factors such as asbestos. Other risk factors include ionizing radiation, radon, environmental toxins, metals (arsenic, chromium and nickel), history of pulmonary fibrosis, HIV infection, and alcohol consumption [11].
Among other factors, inflammation plays an integral role in initiating and supporting tumor growth. Physiologically, inflammation is an important process where the body responds to stimuli such as irritants, pathogens, injuries and eliminates tissue and cell damage. Nevertheless, unregulated inflammation can induce organ fibrosis and cancer [12]. Tumor inflammation is now recognized as one of the “10 characteristics of cancer”. Inflammation is thought to enhance tumor progression and development by supplying pro-tumorigenic constituents to tumor microenvironment [13]. It also contributes at all stages of tumorigenesis, from malignant transformation and tumor initiation to invasion and metastasis [14]. Here, we review the impact of inflammation-mediated mechanisms in the development and progression of NSCLC, and identify potential targets for therapeutic intervention. We will also examine current drugs and natural compounds with anti-inflammatory properties for drug repurposing, and the delivery of repurposed drugs by inhalation to improve treatment efficacy.

2. Inflammation in NSCLC Initiation and Progression

Lung cancer often occurs with a well-defined inflammatory tumor microenvironment. The tumor microenvironment (TME) is formed by various events known as tumorigenesis, progression, invasion, and metastasis [14]. The TME is highly complex, comprising cytokines, vasculature, growth factorsof different populations of stromal cells, such as tumor-associated macrophages (TAMs), tumor-associated fibroblasts (TAFs), and myeloid-derived suppressor cells (MDSCs) [15]. Continuous inflammatory conditions can induce DNA damage and mutations and increased cell division rate damage to lung tissue. Chronic infection, physical inactivity, diet, (visceral) obesity, social isolation, intestinal dysbiosis, sleep disruption, mental stress, and circadian rhythm disruption as well as exposure to xenobiotics such as air pollutants, oxidants, hazardous waste products, gases, poisons, smoking, and lung diseases, have all been found to elevate the risk of developing inflammation in the lung [16,17]. Inflammation can either initiate or promote anti-apoptotic signals and elevate risk of lung cancer. Additionally, inflammation promotes angiogenesis or growth of new blood vessels to provide nutrients to tumor cells [14].
It requires long-term exposure to these factors to induce cancer via inflammation. In a recent clinical study involving 311 NSCLC patients, inflammation was utilized as a prognostic marker for treatment outcome after receiving first-line chemotherapy or targeted therapy for advanced NSCLC. The Systemic Immune-Inflammation Index (SII), which was derived by analyzing neutrophils, platelet counts, and lymphocytes from peripheral patient blood samples, was designed and utilized to measure the impact of inflammation towards cancer progression. Out of 311 patients, 179 were categorized into group A, with SII ≥ 1270, while the remaining patients were categorized into group B, with SII < 1270. The median overall survival was 12.4 months for patients in Group A and 21.7 months for patients in Group B. Median progression free survival was 3.3 and 5.2 months, respectively for both groups. The study concluded that inflammation played a major impact in the prognostic outcome of NSCLC. Furthermore, SII was highly recommended to be included as a tool to measure prognostic impact of patients and this tool applies not only for locally advanced but also for metastatic NSCLC [18]. A meta-analysis study on the prognostic value of pre-treatment advanced lung cancer inflammation (ALI) index in NSCLC was recently conducted. ALI is a tool used to access systemic inflammation in patients by assessing their BMI, serum albumin, and neutrophil-to-lymphocyte ratio. A combination of 14 studies involving 3607 patients concluded that pretreatment ALI was found to be a reliable prognostic marker for NSCLC at both early and late stage. Lower pretreatment ALI indicates poor overall survival and progression free survival [19]. Comparing SII and pretreatment ALI, SII has more inflammatory background compared to pretreatment ALI, as the pretreatment ALI correlated with the immune-nutritional status of the patients.
Lung tumor progression occurs as a result of interaction between intrinsic (genetic) and extrinsic (environment) pathways. Intrinsic pathway is driven strongly by genetic events, such as oncogenes and genetic aberrations, leading to neoplastic transformation and also initiates the construction of an inflammatory microenvironment [20]. In the extrinsic pathway, the inflammatory condition is mainly developed by inflammatory leukocytes, particularly macrophages and soluble mediators such as histamine, serotonin, bradykinin, and other vasoactive amines including eicosanoids, such as prostaglandins, leukotrienes, and thromboxanes [21].
Abnormal inflammation can be induced by oncogene activation and/or silencing of tumor suppressor genes. Genetic alterations in EGFR and KRAS are commonly found in human lung adenocarcinoma. EGFR mutations are more frequent among non-smokers, whereas KRAS mutations are more common in tobacco smokers [12]. KRAS mutation causes elevated secretion of VEGF and CXCL1, which support cancer progression and development [22]. COPD-like inflammation was recently reported to promote lung cancer in KRAS mutant mouse model. Both HIF-1 and HIF-1α activities were found significantly elevated in lung tumors of KRAS mutant mice. Regardless of either absence or presence of COPD, the number of lung tumor surface was found to be significantly reduced in HIF-1α deficient mice. Angiogenesis and cell proliferation activities were also found to be reduced. However, these observations were seen only with HIF-1α expression, indicating a key target for lung cancer progression [23]. HIF-1α is a transcription factor and modulates cellular response to low oxygen through orchestrating a metabolic switch to allow cell survival. Inflamed tissues are often hypoxic, and HIF-α allows immune cells such as macrophages, dendritic cells, neutrophils, T cells, and B cells to adapt by regulating cellular metabolism and expression of immune-related genes to suppress activity of immune cells in early stages of tumor development [24]. Under hypoxic condition, HIF-1α was highly expressed in cancer-associated fibroblasts in human lung cancer tissues and spontaneous lung tumors in mice. Knocking out or inhibiting HIF-1α significantly attenuated fibroblast activation by downregulating NF-ĸB signaling with significant reduction of CCL5, a potent pro-inflammatory chemokine and restricted tumor growth [25].
Acute lung inflammation is an early event where neutrophils play a major role. During acute inflammation, neutrophils migration and chemokine production initiate granulation tissue formation comprising endothelial cells. cellular matrix, fibroblasts, and leukocytes [12]. When the acute inflammation is not resolved, chronic inflammation takes place. During chronic inflammation, other inflammatory immune cells such as macrophages and lymphocytes induce a more severe inflammation in lung, which eventually elevates cancer risks by promoting all stages of tumorigenesis (Figure 1). Tumors arising from chronic inflammation regions are often described with infiltrating leukocytes (mostly macrophages), growth factors, cytokines, and metastatic-promoting enzymes. Depending on type of immune cell infiltrate, the infiltrating leukocytes can exert either pro-tumor or antitumor effects.
A hallmark of lung inflammation is immune cell infiltration. Immune cells infiltrate can be divided into innate and adaptive immune cells [26]. Innate immune cells, such as dendritic cells, myeloid-derived suppressor cells (MDSCs), TAMs, and neutrophils, can either promote or suppress tumor initiation and progression during tissue injury [27]. Dendritic cells mediate antitumoral immunity by cross-presenting tumor antigens to activate T-lymphocytes in lymph nodes [28]. MDSCs degrade L-arganine, and inhibit NK cell-derived IFN-γ production as well as CD4 and CD8 T cell IFN-γ production [29]. MDSCs curb T cell activity by downregulating pro-inflammatory cytokines, such as prostaglandin E2 (PGE2) and IL-12, reducing host immunity to target cancerous cells (Huang et al., 2019). Anti-CD33 antibodies such as Gemtuzumab and ozogamicin were able to restore T cell immunity against cancer [30]. Accumulation of MDSCs is boosted by HIF-1α, released within the hypoxic condition of TME, to facilitate immune evasion of tumor cells [31]. TAMs can be divided into macrophage 1 (M1) and macrophage 2 (M2). M2 macrophages are usually associated with tumor progression, whereas M1 macrophages generate reactive oxygen and nitrogen intermediates, which induces DNA damage in proliferating cells leading to neoplastic transformation. The biological behavior of A549 NSCLC cells, after co-culturing with various macrophage subtypes, were examined in one such study. In this study, M2 macrophages stimulated A549 cell invasion and tumor growth and in contrast, M1 macrophages inhibited A549 cell proliferation and viability by triggering apoptosis and senescence. The results from this study suggested an increased expression of DNA damage-induced proteins, where GADD34 and GADD153 in M1-A549 cells are highly susceptible to cisplatin [32].
Under normal physiology, inflammatory cytokines are released by macrophages to stimulate repair of damaged cells [33]. However, overexpression of inflammatory cytokines, chemokines, and growth factors by TAMs inhibited key enzymes, such as xanthine oxidase (XO), NADPH oxidase, and NOS, which induce accumulation of DNA-damaging agents and disrupt genome integrity and stability [14,33]. TNF-α, IL-6, IL-1β, and IL-8 are often secreted by macrophages in response to smoking or inhaling toxic chemicals from cigarette smoke [34]. Accumulation of TNF-α triggers angiogenesis cascade by inducing “tip-cell” phenotype in endothelial cells with the help of an NF-κB-dependent mechanism [35]. IL-6 plays the role as an activator of JAK and STAT 3 [36] as well as inducing lung cancer metastasis [37]. In chronic inflammation, neutrophils promote carcinogenesis either by supporting tumor-related inflammation, angiogenesis, and metastasis or restricting tumor growth through expressions of antitumor and cytotoxic mediators [38]. Accumulation of neutrophils in the lung enhances lung metastasis by boosting release and production of MMP9. The surge increase of MMP9 by neutrophils results in breakdown of collagen and induce production of inflammation-generated extracellular matrix fragments ac-PGP (N-acetyl-proline-glycine-proline), which act as chemoattractant to stimulate cancer cell migration [39]. Other from MMP9, degranulation of azurophilic granules in neutrophils also elevates other enzymes, such as Ser proteases, cathepsin G, and elastase, leading to degradation of antitumorigenic factor thrombospondin-1 (Tsp-1). Without Tsp-1, tumor growth in metastatic organs will be uncontrollable [40]. Furthermore, neutrophils also assist in contributing neo-plasticity in TME by releasing reactive oxygen species (ROS), thus favoring tumor formation [12].
Apart from changes in innate immunity, adaptive immune cells such as T-lymphocytes, immunosuppressive FOXP3+ T regulatory cells, and CD8+ cytotoxic T-cells are involved in lung cancer development and progression [26]. T-lymphocytes execute cytotoxic effects in tumor microenvironment, and these responses are linked to immune checkpoint inhibition (ICI). Immunosuppressive FOXP3+ T regulatory cells, induced by cyclooxygenase 2 (COX-2), can enhance tumor burden [41]. FOXP3 is essential for Treg development and differentiation. Tregs coordinate cellular and molecular networks to create an immunosuppressive environment and encourages tumorigenesis [42]. CD8+ cytotoxic T cells secrete cytotoxic molecules or make cell-to-cell contact to induce tumor cell apoptosis [43]. Gamma-delta (γδ) T cells are a type of T cells that initiate inflammatory responses of myeloid and lymphoid cell lineages. These γδ T cells can be easily activated by inflammation provoked by local microbiota, leading to the development of lung adenocarcinoma. Germ-free KRAS-mutated and p53 null mice were found protected from lung tumor development after treatment with combination of antibiotics (ampicillin, neomycin, metronidazole, and vancomycin). It was postulated that local microbiota stimulated production of IL-23 and IL-1β in myeloid cells were responsible for the proliferation and stimulation of γδ T cells in these mice. Activated γδ T cells also produce and release IL-17 and other effector molecules to induce inflammation and tumor proliferation [44].
Inflammation supports lung cancer progression by providing essential molecules to the TME. This is achieved with the help of extracellular vesicles (EVs). EVs are lipid bilayers released by a variety of cells. They are also called macrovesicles, exosomes, or apoptotic bodies [45]. EVs harbor cargo molecules, such as RNA, lipids, and proteins, and these cargo molecules are transferred to recipient cells, acting as intercellular communicators within lung TME. Smoking is a major risk factor for lung cancer. In a recent study, smoking-induced extracellular vesicles were characterized in NSCLC smokers, where higher amounts of EVs were found in bronchoalveolar lavage. Furthermore, long non-coding RNAs (MALAT1, FOXD2-AS1, HOTAIR, HOTTIP, HOXA11-AS, AGAP2-AS1, ATB, TCF7, PCAF1, and BCAR4) were detected in smoke-induced EVs and were significantly higher in NSCLC smokers versus non-smokers. In the same study, signaling pathways involving proteoglycans, ErbB, fatty acid biosynthesis, Hippo, Rap1, TGF-β, Wnt, AMPK, and Ras were heavily enriched in EVs from NSCLC patients through bioinformatic analysis identification [46]. The oncogenic contents in EVs could further drive disease progression, as they could be easily absorbed by other cells and influence cellular programming at transcriptional and post-transcriptional levels. These EVs can further inflict damage by attracting cancer-associated fibroblasts, promoting angiogenesis and remodeling of the extracellular matrix to support metastasis within TME [45]. Release of these cargo molecules within the TME changes inflammatory cytokines and growth factor levels, such as transforming growth factor (TGF)-β, IL-1β, IL-6, IL-4, IL-11, IL-12, and MCP-1, and TNF-α activating pro-inflammatory signaling cascades, such as MAP kinases and NF-ĸB pathways [47]. EVs produced by vascular and blood cells can also contribute to the development of atherosclerosis in several ways. Together with LDL cholesterol, they increase thrombotic risks by promoting inflammation, vascular dysfunction, leukocyte adhesion, and tissue remodeling [45].

3. Inflammatory Cytokines in NSCLC

Both TGF-β and IL-6 are produced in response to inflammation in NSCLC. In erlotinib-naïve NSCLC-derived cell lines and early-stage NSCLC tumors, intrinsic erlotinib-resistant cell subpopulations displayed features suggestive of epithelial-to-mesenchymal transition (EMT). TGF-β is a potent driver of EMT in most cancers. Activation of TGF-β signaling induces not only the EMT phenotype, but also promotes TGF-β-dependent IL-6 secretion, which elevates inflammation in TME. Increased inflammatory response in TME has been shown to adversely affect tumor response to EGFR-TKI, erlotinib [48]. Eph receptors participate in tumor progression and their expression is governed by the TGF-β-activated Smad 2 pathway. Eph was found upregulated in NSCLC patient biopsies and significant increase of Eph receptor expression was found correlated with poor survival [49]. Nevertheless, the role of EpH in NSCLC via inflammation is yet to be fully explored. IL-6 is a multipotent pro-inflammatory cytokine, which activates the JAK-STAT3 signaling pathway. In a cancer context, it plays a significant role in multiple tumors, such as lung, colon, breast, prostate, ovarian, and multiple myeloma [36]. Fibroblasts, isolated from human lung cancer tissues, were found to actively secrete IL-6 and enhance metastatic activity in human lung cancer cell lines through JAK2 and STAT3 signal transduction activation [37]. In K-ras mutant lung cancer mouse model, IL-6 was found overexpressed and blocking IL-6 with monoclonal antibody significantly reduced airway inflammation and dampened lung tumor cell proliferation and angiogenesis [50]. However, another study highlighted that IL-6 deficient mice developed higher numbers but smaller-sized lung tumors after activation of mutant KRAS in lung. IL-6 could prevent growth of lung tumors in the early cancer stage by maintaining lung homeostasis through regulation of lung macrophages and cytotoxic CD-8 T cells with IL-6/STAT3 signaling activation promoted tumor progression with more tumor colonies through induction of cell proliferation regulator cyclin D1 under KRAS oncogenic stress [51]. As such, it is vital to keep optimal IL-6 levels to avoid disease progression.
IL-1β, mainly produced by myeloid cells, such as macrophages, mast cells, and neutrophils, regulate various cellular activities including cell differentiation, differentiation, apoptosis, and proliferation. This pro-inflammatory cytokine is often found in lungs of patients with COPD and asthma [52]. The CANTOS trial (Canakinumab Anti-inflammatory Thrombosis Outcomes Study) was a clinical trial conducted to evaluate the safety and efficacy of canakinumab, a monoclonal antibody which targets IL-1β. This trial was conducted in patients with a history of myocardial infarction and high levels of high-sensitivity C-reactive protein (hsCRP), an inflammatory biomarker. Besides reducing the recurrence of cardiovascular events compared to placebo group, the results also indicated that canakinumab significant impact on lung cancer. CANTOS trial revealed IL-1β and inflammasome inhibition could significantly lower incidents of lung cancer [53]. This notorious inflammatory cytokine also promotes blood vessels and lymphatic angiogenesis after inflammasome activation to support tumor development [54]. Angiogenesis is required for tumor progression through formation of new blood vessels to facilitate long distance migration of tumor cells. Pro-inflammatory cytokines including TNF-α and IL-1β activate NF-κB pathway, a key regulator of cell proliferation and growth. However, in tumor cells, persistent NF-κB activity in TME induces angiogenesis and apoptosis, and promotes tumor cell invasion and EMT. Furthermore, it can elevate cyclin D and E expressions, which increases the transition from G1 phase to S phase of dividing cells [14].
IL-8 signaling axis is associated with pathogenesis of inflammatory-based diseases, including cystic fibrosis, asthma, chronic obstructive pulmonary diseases (COPD), and cancer. Secretion of IL-8, by tumor and other cells within the stroma, is critical for cancer progression and metastasis [55]. In NSCLC, epigenetic modification of IL-1β, IL-8, and IL-6 expressions could affect inflammatory response during cancer development [56]. The same observation was seen in tumor cells from NSCLC patients, where rapid lowering of IL-8 serum levels were observed after surgical tumor excision. In human NSCLC, serum IL-8 levels were found correlated with tumor burden and could be utilized as a biomarker to predict tumor burden [57].
TNF-α is a notorious inflammatory cytokine and is often associated with hormone non-responsiveness, poor prognosis, and cachexia [58]. It plays an important role in activating the NF-κB signaling pathway in tumor promotion [59]. TNF-α promotes pleural effusion of lung cancers by causing excessive permeability of airway blood vessels [60]. Surprisingly, TNF-α has been proposed to be used for cancer treatment, as it possesses the ability to induce vascular hyperpermeability and destruction of vascular lining in tumor-associated vasculature. This strategy should aid the accumulation of administered cytotoxic drugs in tumor after vasculature destruction [61].
MCP-1 regulates monocyte chemotaxis and lymphocyte differentiation through CC chemokine receptor 2 (CCR2) binding and plays a significant role in pathogenesis of inflammatory diseases, such as asthma, COPD, and cancer [62]. In the cancer microenvironment, cancer cells and non-cancerous stromal cells, including inflammatory cells, endothelial cells, and fibroblasts, produce MCP-1, which enhances cancer cell migration, survival, and proliferation [63]. MCP-1 expression in solid tumors were evaluated through meta-analysis, and results showed that high level of MCP-1 were related with decreased survival rate (hazard ratio 1.95, 95% CI 1.32–2.88) [64]. In bone cancer, MCP-1 enhances metastasis by promoting interaction between host-derived chemokines and tumor-derived factors [65]. Table 1 shows a summary list of common targets of inflammation in NSCLC.

4. Drugs and Molecules Targeting Inflammation in NSCLC

Inflammation occurs in both early and late phases of lung cancer, and is a highly complex process. The proteins involved in this process can be utilized as candidate targets to treat lung cancer. Targeting inflammation can be applied both to lung cancer induced by intrinsic factors and extrinsic factors.
Mutations in leucine-rich-repeat kinase 2 (LRRK2) are common in immune-related disorders, such as inflammatory bowel disease and Parkinson’s disease [77]. LRRK2 can modulate inflammation during microbial infection in mouse model. LRRK2 mutations are associated with worsened survival of infected animals [78]. LRRK2 is highly expressed in immune cells and has important roles, including regulation of cytokine release, autophagy, and phagocytosis [77]. In a recent study, loss of LRRK2 was observed to promote carcinogen-induced lung tumorigenesis in both patient and mouse lung cancer models. In NSCLC patients, reduced LRRK2 levels led to immunosuppression, altered surfactant metabolism, and lessened differentiated lung adenocarcinoma. It is proposed that the developmental program of growth and differentiation of tumor is strongly associated with weakened activation of inflammatory activities within the region. This observation was fully supported in a carcinogen-induced murine lung cancer model, where LRRK2 knockout led to a significant increase of both tumor numbers and sizes [75]. LRRK2 kinase and GRPase inhibitors, such as MLi-2, PF-06447475, GNE-0877, compound 68 and 70, and FX2149, initially developed for treatment of Parkinson’ disease [79], could be repurposed to treat LRRK2-associated lung cancer.
Isochorismatase domain containing 1 (ISOC1) is a potential biomarker in gastrointestinal cancer, but its role in cancer remains unknown [73]. ISOC1 has also been reported to regulate the growth of breast and pancreatic cancer cell lines. ISOC1 knockdown in these cell lines reduces growth and cell proliferation, induces cell apoptosis, and elevates caspase-3/7 [80]. Elevated ISOC1 expression was seen in NSCLC patients with records of unfavorable disease-free survival. ISOC-1 overexpression in NSCLC cells induced cell proliferation, viability, migration, and invasion, whereas ISOC1 knockout in mouse xenograft model led to significant tumor growth inhibition [73]. ISOC1 suppression also inhibited cell proliferation and migration and induced apoptosis in colon cancer cells [81]. Using RNA sequencing analysis, signaling pathways mediated by ISOC-1 were mainly inflammation related [73].
The enzymatic subunit of polycomb repressive complex 2 (PRC2) is known as an enhancer of zeste homolog 2 (EZH2) and has been identified to activate oncogenes, inhibits tumor suppressor factors, promoting metastasis, altering immunity and metabolism, as well as inducing drug resistance [82]. NSCLC tumors were found to possess high levels of EZH2. In orthotropic KRAS-driven EZH positive NSCLC grafts, treatment with EZH2 inhibitor GSK126 could amplify inflammation through activation of NF-ĸB and genes residing within the PRC-2 regulated chromatin. The inflammation allowed tumor cells to overcome GSK126 antiproliferative effects, an unfavorable event and possibly rendering EZH2 inhibitors ineffective against KRAS-driven NSCLC. In the same study by Serresi et al., GSK126-treated NSCLC in vivo displayed enhanced response towards nemisulide (NSAID) and bortezomib combination treatment [74]. Aspirin, naproxen, sulindac acid, amino salicylic acid, and celecoxib are NSAIDs that should be considered for use in combination with EZH2 inhibitors for KRAS-driven NSCLC. These lines of evidence indicate that combination of anti-inflammatory drugs is a plausible strategy to resolve EZH inhibitor ineffectiveness in KRAS-driven NSCLC.
Molecules targeting signaling pathways such as NF-ĸB and STAT3 are often studied in inflammatory-based diseases including cancer. STAT3 pathway transmits extracellular signals to the nucleus and regulates immunity, inflammation, and tumorigenesis. Activation of STAT3 mediates various cellular processes including survival, proliferation, invasion, inflammation, angiogenesism and metastasis [83,84]. STAT3 pathway activation in NSCLC induces tumor resistance towards conventional and small molecule targeted therapy [85]. STAT3 often interacts with other signaling pathways, such as NF-ĸB, commonly associated with lung inflammation and confers robustness in tumor progression. Hyperactivation of STAT3 leads to a series of tumor promoting events, such as immunosuppression in tumor-infiltrating cells, dampening antigen presentation, and inhibition of tumor-killing activities [86]. Existing drugs targeting the NF-ĸB pathway, such as thioridazine, imatinib, mesylate, clemastine, and ibudilast, can be repurposed for lung cancer treatment [87,88,89]. Imatinib, an oral anticancer agent that inhibits tyrosine kinase activity, is used to inhibit BCR-ABL1 fusion oncoprotein, c-kit, platelet-derived growth factor receptor (PDGFR), and native tyrosine-protein kinase Abelson murine leukemia (ABL1 kinase) [90]. Imatinib can modulate immune response by inhibiting IL-6 and other proinflammatory cytokines through suppressing NF-ĸB activity [90]. Two clinical phase 2 trials of Imatinib, in combination with docetaxel or paclitaxel, reported poor clinical outcomes due to poor therapeutic responses and unwanted side effects, such as chronic gastrointestinal toxicity (nausea, vomiting, and diarrhea) and cardiac toxicity (cardiomyocyte injury) [91,92]. The antagonistic effects of Imatinib suggest that caution should be taken administering in combination with other drugs. However, the unwanted side effects can be reduced if Imatinib is given directly to the lung instead of the usual i.v. route.
Pro-inflammatory cytokines are important in ensuring that inflammation is regulated at an optimal level to promote lung carcinogenesis. Lowering pro-inflammatory cytokines, such as IL-1β, IL-2, IL-6, IL-8, IL-10, IL-12, interferon γ (IFN-γ), TNF-α, and granulocyte-macrophage colony-stimulating factor (CSF), can reduce lung cancer risk, particularly among smokers [93]. These pro-inflammatory cytokines can be inhibited with existing biologics such as antibodies targeting either a cytokine or its receptor. Sarilumab, Tocilizumab, and siltuximab are existing FDA-approved IL-6 inhibitors for rheumatoid arthritis and COVID-19 to reduce damages caused by IL-6-induced inflammation [94,95]. On the other hand, glucocorticoids, which are wide-spectrum anti-inflammatory agents, reduced pro-inflammatory cytokine expression via genomic and non-genomic pathways in COVID-19-induced acute respiratory distress syndrome (ARDS) patients [96]. In addition to that, dimethyl fumarate (DMF) also inhibits more extensive pro-inflammatory cytokines, especially IL-1 and IL-6 [97]. Three IL-1 inhibitors (anakinra, rilonacept, and canakinumab), used as a single agent or in combination for treatment of rheumatoid arthritis and IL-1, induced autoimmune disease [98], and can be utilized to reduce IL-1 induced lung inflammation. Results from a recent CANOPY-1 Phase III study showed that locally advanced or metastatic NSCLC patients treated with canakinumab did not achieve its primary endpoints of overall survival and progression-free survival. However, the study recommended the use of canakinumab in patients with elevated inflammatory biomarkers at early stages of lung cancer, as canakinumab-treated patients showed improved progression-free survival and overall survival [99].
PD-1 immune checkpoint pathway is an attractive NSCLC therapy where it prevents T-cell activation by downregulating immune system response, promoting self-tolerance and reducing auto-immunity. However, the PD-1/PD-L1 pathway has also been associated with significant inflammatory effects. Besides cancer, this pathway has served as a target in other inflammatory-based diseases, including autoimmune responses, chronic infections, and sepsis [100]. Nivolumab, a human immunoglobulin G4, has demonstrated superior overall survival in patients with advanced squamous NSCLC patients when compared to docetaxel [3]. In cancer immunotherapy, many inhibitors of pro-inflammatory cytokines, such as TNF-α, TGF-β, and CSF, have been used in combination with anti-PD-L1 or anti-PD-1 agents, and have shown promising improvement in therapeutic outcomes, in comparison to monotherapy agents Cytokines, which exert therapeutic efficacy by potentiating immune response and inhibiting the immunosuppressive activity. Currently, there are ongoing clinical trials to evaluate therapeutic effect of cytokines by combining them with various anti-PD-L1 and anti-PD-1 agents [101].
Neutralizing the effects of pro-inflammatory cytokines with existing biologics, small molecules, cytokine traps, or RNA interference should be further explored. Interestingly, expressions of estrogen receptor, progesterone receptors, and aromatase have been associated with poor prognostic outcome in post-menopausal NSCLC in females [102]. In a tobacco carcinogen-induced lung tumor mouse model, the combination of aromatase inhibitor (anastrozole) with an NSAID (ibuprofen or aspirin) resulted in stronger tumor prevention effects in comparison to a single agent. The combination treatment reduced infiltrating macrophages, deactivated MAPK and STAT3 signaling, and inhibited inflammatory markers, such as IL-6 and IL-17A [102].
Excessive production of ROS can damage normal and cancer cells and regulate the production of oxidants at an optimal level for survival. The ROS produced activate many oncogenes, which induces production of inflammatory cytokines and factors. Statins are low-density lipoprotein cholesterol lowering drugs that inhibit enzyme HMG-CoA reductase. Rosuvastatin was reported to inhibit pro-inflammatory cytokines including tumor necrosis factor-α (TNF-α), IL6, and TGF-β in mice, leading to tumor shrinkage [103]. Liposomal pravastatin treatment was found to effectively inhibit inflammatory cytokine production, such as GM-CSF, IGF-II, IL-1α, IL-1β, leptin, IL-6, and TNF-α [104]. A recent study investigated the association between statin exposure and lung cancer risk in a population of COPD patients (n = 39,879) and the results showed that statin use significantly reduced the risk of lung cancer The results indicated that statin use may specifically reduce non-small cell lung cancer and not small cell lung cancer. Lung cancer risk is reduced by statins via a reduction in systemic inflammation, which also leads to slowing down of the decline in lung function and reduction in all-cause mortality [105]. Despite possessing potent anti-inflammatory effects, the exact anti-inflammatory mechanism of statins is unknown. Statins are known to exert pleiotropic effects, such as reducing cell proliferation, angiogenesis, and invasion. However, given statin’s potency, this drug remains highly attractive for the prevention and treatment of lung cancer [103].
The hypoxia-inducible factor (HIF) pathway plays a crucial role in solid tumors including lung cancer. Among the two sub-units HIF-α and HIF-β, HIF-α plays a more crucial role in regulating inflammation [23,24,25]. There are several suitable HIF inhibitors currently in phase II and III of clinical trials, and results have shown promising outcomes for gliobastoma, melanoma, lymphoma, colorectal, and mesothelioma malignancies. HIF-α inhibitors, including 2ME2 NCD (panzem), 17-AAG (tanespimycin), Vorinostat (SAHA), EZN-2208 (pegylated SN-38), and CRLX101, are potential drugs to be considered for the treatment of lung cancer [31].
Fingolimod (FTY720), a sphingosine-1-phosphate receptor modulator, is commonly used as an immunomodulator in multiple sclerosis treatment [106]. Inflammatory mediators, such as TNF-α, IL-6, and IL-1β, were found induced in rats with renal ischemia/reperfusion, which resulted in lung injury. Intraperitoneal FTY720 administration were found to protect against acute lung injury by reducing pulmonary inflammation through downregulation of sphingosine-1-phosphate metabolism [73]. Additionally, FTY720 also prevented pulmonary cell apoptosis in a renal ischemia/reperfusion model [73] and induced fibrosis in a bleomycin-induced lung injury model [107], indicating that a combination of strong apoptosis inducer and antifibrotic agents could be given for effective lung cancer treatment. Table 2 provides a summary of existing drugs with anti-inflammation properties for NSCLC treatment and their chemical structures are included in Figure 2. Table 3 shows natural compounds that are currently undergoing clinical trials for the treatment of non-small cell lung cancer and their chemical structures are included in Figure 3.

5. Natural Compounds Targeting Inflammation in NSCLC

Natural products remain a potential source for new and innovative drug discovery, given that many have shown to possess anti-inflammatory properties. They are rich with secondary metabolites, such as flavonoids, terpenes, and alkaloids. Several herbal medicinal plants have been actively studied for their anti-inflammatory properties. In recent years, many natural products have been reported to exert effects against lung TME. Most are recognized with potential to be developed as new plant-derived chemotherapy agents due to their ability to modulate angiogenesis, the extracellular matrix, MDSC, TAMs, and immune checkpoint [113].
Cinnamon contains secondary metabolites, such as cinnamaldehyde, cinnamic acid, 2-hydroxycinnamaldehyde, 2-methoxycinnamaldehyde, and eugenol, and possesses potent anti-inflammatory effect by reducing pro-inflammatory IL6, IL-1β, and TNF-α and suppressing NF-ĸB-mediated COX-2 and iNOS pathways [114]. With regards to NSCLC, cinnamon extract was found to suppress invasion of A549 and H1299 cells by regulating the expression of FAK and ERK pathways [115]. Combination therapy with cinnamaldehyde and hyperthermia was also found to induce apoptosis of A549 cells by regulation of reactive oxygen species and the MAPK pathway [116].
Hochuekkito (TJ-41) is a Japanese traditional herbal kampo medicine comprising 10 natural herbs. TJ-41 was shown effective in attenuating lung inflammation in the COPD mouse model and LPS-induced macrophage cell line through TNF-α ablation [117]. Secondary plant metabolites found to possess potent anti-inflammatory properties include Andrographolide, baicalein, curcumin, Pterostilbene, Dihydroisotanshinone I, Ginsenoside Rh2, vitamin D, and zerumbone.
Immune cell regulation is a crucial event in lung inflammation, and manipulating these immune cells can prevent inflammation and impede cancer progression, particularly in the early stages. Andrographolide, the primary active component found in Andrographis paniculata, is a labdane diterpene known for its potent anti-inflammatory properties [118]. It has been reported to inhibit the production of several pro-inflammatory cytokines and chemokines, such as TNF-α, IL-6, and IL-8, and suppress the activation of the NF-κB and MAPK signaling pathways, which are crucial regulators of inflammation [119,120]. Additionally, andrographolide has been found to exhibit anticancer effects in various cancers.
Andrographolide has been reported to suppress migration of macrophages towards chemo-attractants, such as complement 5a (C5a), through the inhibition of phosphorylation of mitogen-activated protein kinase (MAPK) kinase 1/2 (MEK1/2) and downstream p42/p44 MAPK (aka extracellular signal-related kinase 1/2, ERK1/2) and Akt signaling pathways [121]. Furthermore, andrographolide acts on other cellular pathways regulation, including mTOR, Wnt/β-catenin, TRAIL-mediated apoptosis, as well as VEGF-mediated intracellular signaling, and adversely affects tumor development [122]. Interestingly, andrographolide can also inhibit human NSCLC cellular proliferation and induces apoptosis by reprogramming host glucose metabolism [123]. In addition to growth inhibition, andrographolide could suppress aggressive metastatic cancer, including luminal-like breast cancer through NF-ĸB pathway inhibition [124]. Data from high-throughput metabolomics analysis revealed that this compound exerted its anticancer properties by enhancing immune system activity, reduces inflammation, tumor cell metastasis, and balancing visceral metabolism in a Lewis lung cancer model [125]. Resistance to cisplatin in NSCLC, achieved through autophagy, is a hindrance and andrographolide is found capable of inhibiting autophagy in cisplatin-resistant NSCLC by activating the Akt/mTOR pathway, and re-sensitizes tumor cells towards cisplatin [126].
Pterostilbene, Dihydroisotanshinone I, and Ginsenoside Rh2 are natural compounds capable of inhibiting TAMs activity [113]. Pterostilbene is a natural analogue of resveratrol, which has metabolic stability and superior pharmacological activities. Pterostilbene was initially extracted from red sandalwood (Pterocarpus santalinus) and can primarily be found in a few natural sources, such as grapes, blueberries, and Pterocarpus marsupium [127]. Pterostilbene treatment led to reduced expressions of NF-ĸB, CD133, MUC1, β-catenin, and Sox2 in inflammatory lung TAMs. This also led to a significant loss of stemness by TAMs with decreased side-population cells and suppression of self-renewal ability in TAM-co-cultured lung cancer cells [128]. In another study, female Balb/C mice were treated with varying doses of pterostilbene to examine its impact on cell proliferation, cell death, and the p53 pathway. The study observed a reduction in Ki-67 expression and an increase in caspase-3 expression, leading to a decrease in cyclin D1 and cyclin E2 protein expression, causing cell cycle arrest. Furthermore, pterostilbene increased p53, p21, and p27 protein expression [127].
Dihydroisotanshinone I is a pure compound extracted from danshen, which is a Chinese medicinal herb. Dihydroisotanshinone I could inhibit tumor migration and cell motility, block macrophage recruitment by lung cancer cells, reduce CCL2 secretion, and suppress p-STAT3 signaling in NSCLC A549 and H460 cells [129].
Ginsenoside Rh2, found in ginseng, possessed the ability to convert TAMs from M2 to M1 subtype and prevented cancer cell migration by curbing TAMs activity in TME [130]. Ginsenoside Rh2 was also found to inhibit hypoxia-induced cell migration by increasing the expression of mir-491, which subsequently downregulated the expression of MMP-9. In this study, the effects of ginsenoside Rh2 on hypoxia-induced migration in lung adenocarcinoma was studied. Rh2 was found to inhibit hypoxia-induced cell migration in A549 and H1299 cell lines through the upregulation of mir-491 expression. Additionally, mir-491 antisense oligonucleotide suppressed hypoxia-induced migration and the expression of matrix metalloproteinase (MMP)-9 expression in Rh2-treated A549 cells [131].
Vitamin D, found in sea fish and animal liver, was reported to reduce hyperinflammation in both macrophages and MDSCs in COVID-19 patients. A complication, commonly seen in the lungs of COVID-19 patients, is hyperinflammation-induced acute respiratory distress syndrome. In patients who suffered from acute respiratory distress syndrome and lacked vitamin D, symptoms were reduced after vitamin D supplementation [132]. Vitamin D was also found to improve the survival of patients with cancer found by a meta-analysis of randomized clinical trials performed [133]. Additionally, in a randomized controlled trial with 155 patients NSCLC, vitamin D supplementation show a significant difference in relapse-free survival (RFS) or overall survival (OS) compared to placebo among the subgroup of patients with early-stage adenocarcinoma and low levels of 25(OH)D [134].
Resveratrol is a naturally occurring non-flavone polyphenol compound that is derived from various plants, such as Polygonum cuspidatum, Cassia tora Linn, and Vitis vinifera. It belongs to the stilbene family. Resveratol was found to activate autophagy and apoptosis in A549 cell by regulating the NGFR-AMPK-mTOR signaling pathway [135]. Resveratrol, found in grape skin and seeds, and silymarin, from Silybum marianum, were shown to modulate MDSCs in lung cancer in vivo model [113].
Baicalein is a major bioactive compound found in the root of Scutellaria baicalensis, a traditional Chinese herb. Baicalein was reported to effectively inhibit NSCLC cell invasion and metastasis without any toxicity. This flavonoid significantly reduces ezrin tension by reducing cellular ezrin S-nitrosylation (SNO) levels and iNOS expression in the inflammatory microenvironment of NSCLC [136]. Baicalein has been proven to exert anti-airway inflammation in cigarette smoke-induced chronic obstructive pulmonary rat model by regulating pro- and anti-inflammatory balance [137], and in an OVA-induced allergic airway inflammation model through iNOS and NF-ĸB signaling inhibition [138]. Dimethyl fumarate is a promising fumaric acid ester and possesses strong anti-oxidative, anti-inflammatory, and immunomodulation properties [139]. Given to mice with chronic exposure to diesel exhaust particles, peroxynitrite, total reactive oxygen species, and nitric oxide levels in the lung were significantly reduced, whereas expression of products such as nitrotyrosine, glutathione peroxidase-1/2, and catalase were significantly elevated [140]. The observed changes were possibly due to downregulation of NF-ĸB pathway. Dimethyl fumarate has been reported to inhibit metastasis in cutaneous T cell lymphoma and melanoma through NF-ĸB pathway inhibition as well [139].
Curcumin is a natural compound found in Curcuma longa, possessing a variety of pharmacological properties, including antidiabetic, neuroprotective, anticancer, and anti-inflammation [141]. In NSCLC, migratory and invasive ability of A549 cells is reduced by curcumin through inhibition of adiponectin, an acid peptide hormone, via the NF-ĸB pathway [142]. Zerumbone, a monocyclic sesquiterpene compound, found in Zingiber zerumbet rhizomes has a broad range of pharmacological activities and anti-inflammatory effects. Recently, zerumbone was shown to suppress LPS-Induced inflammation in macrophages through inhibition of NLRP3 inflammasome. Furthermore, NF-ĸB activity and production of inflammatory cytokines, such as IL-1β and IL-6, were also significantly reduced [143]. The same inhibitory effect could also be seen in TNF-α-activated fibroblasts treated with zerumbone, in which tumor-promoting cytokines TNF-α, TGF-β, IL-33, SDF-1, and MCP-1 were significantly reduced in comparison to TNF-α-activated fibroblasts [144]. Astaxanthin, a naturally occurring xanthophyll carotenoid is found in marine organisms, such as algae, shrimp, and salmon [145]. Asthaxanthin possesses both anti-inflammatory and anti-oxidant properties and is shown to be capable of protecting the lung against inflammatory-based diseases. This compound exerts these effects by regulating nuclear factor erythroid 2-related factor/heme oxygenase-1, NF-ĸB, MAPK, JAK-STAT3, PI3-kinase/Akt pathways, and modulating immune response [145,146,147].
Farnesoid X receptor, known to regulate immune responses and inflammation in immune-mediated diseases, can promote tumor cell proliferation in NSCLC. Overexpression of this receptor in a Lewis lung carcinoma (LLC) syngeneic mouse model resulted in downregulation of PD-L1 [148]. The immunosuppressive role of farnesoid X receptor suggests it is a candidate for drug development. Additionally, it offers the opportunity for existing anti-PD-1 therapy to be fully utilized in treatment of NSCLC patients with high PD-1 expression. Bile acid and non-bile antagonists, such as Tauro-β-muricholic acid (T-β-MCA), taurochenodeoxycholic acid, glycoursodeoxycholic acid, guggulsterone, epiallopregnanolone sulfate, 3,5-disubstituted oxadiazole core, stigmasterol, tuberatolides, and andrographolide, are known to inhibit farnesoid X receptor [72]. These versatile natural compounds offer an alternative approach to curb cancer progression. Table 4 provides a list of natural compounds possessing anti-inflammation properties for NSCLC treatment.

6. Repurposing Drugs with Anti-Inflammation Properties and Their Delivery via Inhalation in NSCLC

Despite advances in treatment options, NSCLC’s mortality rates are still at an alarming state, and hence, a more precise and target-specific treatment is urgently needed to overcome this problem. Chronic inflammation plays an inevitable role in tumor initiation and progression. Thus, targeting inflammation presents an important tool in NSCLC treatment. There are various anti-inflammatory drugs and compounds that are readily available, which can be repurposed for NSCLC treatment.
Drug repurposing, also known as drug repositioning, is a process where the therapeutic use of an old or existing drug is explored for treatment of other indications. This is a highly efficient strategy with minimum risk of failure [152]. Another significant advantage of drug repurposing is that these drug candidates have already been clinically proven to be safe. Therefore, repurposed drugs are cost- and time-efficient as the pharmacokinetics and drug safety profile of the approved drugs have been fully investigated [153,154]. Drug repurposing is, hence, a promising option in targeting inflammation in NSCLC. Many compounds derived from natural products have shown promising anti-inflammatory activity and exhibit anticancer activity in NSCLC in pre-clinical studies. However, most suffer from a poor pharmacokinetic profile, which restricts further development.
Though many drugs have been shown to possess anti-inflammatory properties, there is a still a need to improve delivery of these drugs to target site. Localization of drugs at the intended site is vital to avoid toxicity issues related to their original indication. Statins, indicated to treat hypercholesterolemia, have low bioavailability, high first-pass metabolism in liver and intestine, rapid systemic clearance, and low half-life when administered orally. To overcome this issue, a high oral dose is, thus, required, which induces hepatotoxicity, rhabdomyolysis, nephrotoxicity, myalgia, and multiple drug interactions with other drugs and food [108]. Furthermore, NSAIDs can induce severe side effects in the cardiovascular, gastrointestinal, and renal systems, resulting in system failure in severe cases [155]. Thus, localized therapy provides an option to overcome these drawbacks.
Inhalation therapy, a localized therapy, directly delivers drug to the lung and minimizes systemic exposure and toxicity. Inhalation therapy provides rapid clinical response due to the ability to bypass therapeutic barriers, such as gastrointestinal absorption and first-pass liver metabolism [156]. The large surface area of lungs, highly dispersible nature of aerosol, and good epithelial permeability allow small molecules to be deposited in lungs and absorbed into circulation. Drug-metabolizing enzymes are lower in concentrations in lungs compared to the gastrointestinal tract and liver, thus allowing inhaled molecules to remain longer [157]. In addition, inhalation therapy can achieve either an equivalent or better therapeutic effect at a fraction of the systemic dose. An oral dose of 2–4 mg of salbutamol has been found to be equivalent to 100–200 μg by inhalation therapeutically [156]. Therefore, pulmonary drug administration is a promising route of drug delivery to improve the clinical efficacy of drug administered and minimizes unnecessary side-effects.
The administration of repurposed anti-inflammatory drugs for the treatment of NSCLC is one of the major challenges in achieving therapeutic efficacy. The non-localized administration of repurposed drug may cause other severe side effects, such as systemic toxicity and other toxicities which might be related to their original indications. To illustrate that, simvastatin, a type of drug indicated for the treatment of hyperlipidemia, exhibited a hypolipidemic effect when administered orally for cancer treatment, as it tends to accumulate in the liver [158]. Therefore, non-localized administration of repurposed drug cause unwanted effects, such as potential accumulation in organs such as the liver, spleen, and kidney. As a result, this causes failure of the repurposed drug in suppressing tumor growth in a more effective way. When dosage and frequency of drug administration was increased to overcome the above-mentioned disadvantage, there was the occurrence of multi-drug resistance and increased side effects. In addition to that, non-localized administration of the repurposed drug may also result in tumor recurrence [159]. Apart from that, naproxen was repurposed to treat bladder cancer and resulted in severe side effects, such as gastrointestinal, cardiovascular, and renal complications [160]. Thus, in order to specifically target NSCLC, localized therapy with inhalation therapy would help in improving the therapeutic efficacy and also in reducing unwanted systemic toxicity.
Various physicochemical properties influence drug deposition in various regions of the lung. Particle size, pKa, shape, lipophilicity (log P), and solubility are key factors moderating successful drug delivery to the lung [160]. Among these, particle size is a primary factor that influences particle deposition due to diminishing diameter of the airway towards the alveoli. Particles with larger molecular sizes have poorer ability to cross the air–blood barrier. Large drug molecules, however, possess a high receptor binding affinity, which can help reduce off-target effects [161]. Large molecular size drugs with sizes ranging from 1–3 μm are efficiently deposited in the lungs [162]. However, the effect of particle size can also be adjusted by incorporation of polymers. Nanoparticles with a diameter less than 200 nm, coated with polyethylene glycol (PEG), were found to be moving rapidly within mucus mesh. PEG-coated nanoparticles with a diameter larger than 500 nm were found almost immobilized in mucus mesh. Conversely, particle size can influence particles to escape clearance by macrophages where macrophages usually opsonized larger particles, with sizes ranging from 0.5 to 5 μm [163].
The rate and extent of drug uptake by lungs are altered by drug lipophilicity (log P) and pKa as the drug passes through surfactant, alveolar macrophage, mucus, and mucociliary clearance. Lipophilicity of drug impacts residence time of drug on surface of airways and influences therapeutic efficacy of the drug. Lipophilic drugs, having slower dissolution rates, undergo slower mucociliary clearance [164]. For example, inhalable corticosteroids have shown longer residence time in the lungs due to their slow dissolution rate and high lipophilicity [160]. Furthermore, drug delivery via inhalation is regulated by the morphology of drug particles. The aerodynamic performance of particles is influenced by particle shape, which affects their deposition. Particles with amorphous shape exhibit greater lung permeability compared to needle (crystalline)-shaped drug particle when administered through inhalation. In addition, pollen-shaped drug particles showed elevated lung deposition compared to drug particles of other shapes with the same aerodynamic size range [165].
Nanoformulation approaches for direct pulmonary delivery of repurposed drugs have extensively evolved since its first introduction in 1960s. In particular, lipid nanoparticles are important biodegradable and biocompatible delivery systems offering higher drug entrapment and site-specific delivery [166]. Nanoemulsions or microemulsions are thermodynamically stable oil-in-water emulsions and optically isotropic and transparent single-phase liquid suspensions. Microemulsions, such as Sandimmun Optoral and Neoral pre-concentrates, have shown superior solubilizing capacities compared to micellar solutions [167]. Nanoemulsions sizes range between 100 to 500 nm and are now commonly used as drug carriers in lipophilic active ingredients. Etomidat Lipuro, Diazepam Lipuro, Disoprivan, and Stesolid are among the current pharmaceutical nanoemulsions in the market [168]. Advantages of nanoemulsions over microemulsions are reduced local and systemic side effects, lesser pain during injection, and reduced hemolytic events.
Liposomes are spherical vesicles with membranes comprising one or more phospholipid (amphiphilic) bilayers, which are separated by an aqueous compartment. In the design of liposomes as drug carriers, natural phospholipids are often selected for their biological inertness, weak immunogenicity, and low intrinsic toxicity. Liposomes are classified based on vesicle size, lamellarity, and preparation. Unilamellar vesicles comprise one lipid bilayer with diameters between 50 to 500 nm, while multi-lamellar vesicles are envisioned as an onion skin-like arrangement comprising several concentric lipid bilayers with diameters between 1 to 5 µm. The multilamellar vesicles have high lipid content and can passively entrap drugs [169].
Using drug-loaded liposomes as delivery systems, drug concentrations at sites of action are seven- to nine-fold higher than using drugs alone without carriers [170]. The fact that liposomes may be administered parenterally renders it a good candidate as inhalable cargo to achieve an effective pulmonary drug delivery [160]. Currently, the downside of liposomes is that they are susceptible to removal by a mononuclear phagocytic system through non-specific binding of nanoparticle opsonizing serum proteins [171].

7. Conclusions

Comprehensive genomic profiling has allowed a more precise and personalized approach to treatment through tumor mutation profiles and PD-L1 expression in advanced NSCLC patients. However, lung cancer remains a leading cause of cancer-related deaths, despite the development and implementation of targeted therapy and immunotherapy in NSCLC. Inflammation exerts extensive roles within the lung TME to promote the development and progression of NSCLC. Repurposing of existing drugs and natural compounds exhibiting anti-inflammation properties provides an alternative paradigm in NSCLC therapy. Besides, repurposed anti-inflammatory drugs also provides a therapeutic option for NSCLC patients at an affordable cost in a short period of time. Repurposed drugs are time and cost effectiveness, as these drugs are already established on the market with regulatory approval. This reduces the time frame and money investment for the development of these repurposed drugs. However, most of these drugs are administered orally and are limited by their low bioavailability in the system to exert the desired effects. Localized therapy, such as pulmonary drug administration, tends to increase the drug deposition at the targeted organ with reduced unwanted toxicity and increased therapeutic effect. This requires the formulation of repurposed drugs to allow direct delivery to the lungs.
For a drug particle/molecule to diffuse through the extracellular matrices and reach the target organ and cells, it must possess favorable properties, such as small molecular weight, ideal lipophilicity, good solubility, and appropriate pKa, among others [160]. However, these desirable parameters have always been referred to as non-existent due to the highly complex interplay between molecule structure and pharmacodynamic/pharmacokinetic profiles. These requirements have severely restricted commercialization of many drugs for lung delivery. Although the ideal parameters could not fully be attained, they can still contribute to the overall effectiveness of a drug. Hence, an overall understanding to the challenges involved in drug distribution to reach the pulmonary site is highly necessary to ensure the success of drug repurposing and its effectiveness.

Author Contributions

Conceptualization, A.A. and J.C.W.L.; Methodology, T.R., C.W.H. and J.C.W.L.; Software, A.S.; Writing—original draft preparation, T.R., C.W.H. and J.C.W.L.; Writing—review and editing, A.A., J.C.W.L.; Visualization, A.S.; Supervision, A.A., C.W.H. and J.C.W.L.; Project administration, A.A. and J.C.W.L.; Funding acquisition, A.A. All authors have read and agreed to the published version of the manuscript.

Funding

This review is funded by National University of Singapore Grant (A-0001263-00-00).

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Sung, H.; Ferlay, J.; Siegel, R.L.; Laversanne, M.; Soerjomataram, I.; Jemal, A.; Bray, F. Global Cancer Statistics 2020: GLOBOCAN Estimates of Incidence and Mortality Worldwide for 36 Cancers in 185 Countries. CA A Cancer J. Clin. 2021, 71, 209–249. [Google Scholar] [CrossRef] [PubMed]
  2. Society, A.C. Key Statistics for Lung Cancer; American Cancer Society: Kennesaw, GA, USA, 2022. [Google Scholar]
  3. Zappa, C.; Mousa, S.A. Non-small cell lung cancer: Current treatment and future advances. Transl. Lung Cancer Res. 2016, 5, 288–300. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  4. Kalinke, L.; Thakrar, R.; Janes, S.M. The promises and challenges of early non-small cell lung cancer detection: Patient perceptions, low-dose CT screening, bronchoscopy and biomarkers. Mol. Oncol. 2021, 15, 2544–2564. [Google Scholar] [CrossRef] [PubMed]
  5. Sharma, P.; Mehta, M.; Dhanjal, D.S.; Kaur, S.; Gupta, G.; Singh, H.; Thangavelu, L.; Rajeshkumar, S.; Tambuwala, M.; Bakshi, H.A.; et al. Emerging trends in the novel drug delivery approaches for the treatment of lung cancer. Chem. Biol. Interact. 2019, 309, 108720. [Google Scholar] [CrossRef]
  6. Mustachio, L.M.; Roszik, J. Current Targeted Therapies for the Fight against Non-Small Cell Lung Cancer. Pharmaceuticals 2020, 13, 374. [Google Scholar] [CrossRef]
  7. Schrank, Z.; Chhabra, G.; Lin, L.; Iderzorig, T.; Osude, C.; Khan, N.; Kuckovic, A.; Singh, S.; Miller, R.J.; Puri, N. Current Molecular-Targeted Therapies in NSCLC and Their Mechanism of Resistance. Cancers 2018, 10, 224. [Google Scholar] [CrossRef] [Green Version]
  8. Fukui, T.; Tachihara, M.; Nagano, T.; Kobayashi, K. Review of Therapeutic Strategies for Anaplastic Lymphoma Kinase-Rearranged Non-Small Cell Lung Cancer. Cancers 2022, 14, 1184. [Google Scholar] [CrossRef]
  9. Hochmair, M.J.; Fabikan, H.; Illini, O.; Weinlinger, C.; Setinek, U.; Krenbek, D.; Prosch, H.; Rauter, M.; Schumacher, M.; Wöll, E.; et al. Later-Line Treatment with Lorlatinib in ALK- and ROS1-Rearrangement-Positive NSCLC: A Retrospective, Multicenter Analysis. Pharmaceuticals 2020, 13, 371. [Google Scholar] [CrossRef]
  10. Sui, H.; Ma, N.; Wang, Y.; Li, H.; Liu, X.; Su, Y.; Yang, J. Anti-PD-1/PD-L1 Therapy for Non-Small-Cell Lung Cancer: Toward Personalized Medicine and Combination Strategies. J. Immunol. Res. 2018, 2018, 6984948. [Google Scholar] [CrossRef] [Green Version]
  11. Duma, N.; Santana-Davila, R.; Molina, J.R. Non-Small Cell Lung Cancer: Epidemiology, Screening, Diagnosis, and Treatment. Mayo Clin. Proc. 2019, 94, 1623–1640. [Google Scholar] [CrossRef]
  12. Nakano-Narusawa, Y.; Yokohira, M.; Yamakawa, K.; Ye, J.; Tanimoto, M.; Wu, L.; Mukai, Y.; Imaida, K.; Matsuda, Y. Relationship between Lung Carcinogenesis and Chronic Inflammation in Rodents. Cancers 2021, 13, 2910. [Google Scholar] [CrossRef]
  13. Hanahan, D.; Weinberg, R.A. Hallmarks of cancer: The next generation. Cell 2011, 144, 646–674. [Google Scholar] [CrossRef] [Green Version]
  14. Tan, Z.; Xue, H.; Sun, Y.; Zhang, C.; Song, Y.; Qi, Y. The Role of Tumor Inflammatory Microenvironment in Lung Cancer. Front. Pharmacol. 2021, 12, 688625. [Google Scholar] [CrossRef] [PubMed]
  15. Yang, Q.; Zhang, H.; Wei, T.; Lin, A.; Sun, Y.; Luo, P.; Zhang, J. Single-Cell RNA Sequencing Reveals the Heterogeneity of Tumor-Associated Macrophage in Non-Small Cell Lung Cancer and Differences Between Sexes. Front. Immunol. 2021, 12, 756722. [Google Scholar] [CrossRef] [PubMed]
  16. Furman, D.; Campisi, J.; Verdin, E.; Carrera-Bastos, P.; Targ, S.; Franceschi, C.; Ferrucci, L.; Gilroy, D.W.; Fasano, A.; Miller, G.W.; et al. Chronic inflammation in the etiology of disease across the life span. Nat. Med. 2019, 25, 1822–1832. [Google Scholar] [CrossRef]
  17. Li, R. Tumor-Promoting Inflammation in Non-Small Cell Lung Cancer. UCLA. 2016. Available online: https://escholarship.org/uc/item/6972h04z (accessed on 5 January 2023).
  18. Berardi, R.; Santoni, M.; Rinaldi, S.; Bower, M.; Tiberi, M.; Morgese, F.; Caramanti, M.; Savini, A.; Ferrini, C.; Torniai, M.; et al. Pre-treatment systemic immune-inflammation represents a prognostic factor in patients with advanced non-small cell lung cancer. Ann. Transl. Med. 2019, 7, 572. [Google Scholar] [CrossRef] [PubMed]
  19. Tang, Y.; Ji, Y.; Yang, M. Prognostic value of pre-treatment advanced lung cancer inflammation index in non-small cell lung cancer: A meta-analysis. Biomarkers 2022, 27, 441–447. [Google Scholar] [CrossRef] [PubMed]
  20. Singh, N.; Baby, D.; Rajguru, J.P.; Patil, P.B.; Thakkannavar, S.S.; Pujari, V.B. Inflammation and cancer. Ann. Afr. Med. 2019, 18, 121–126. [Google Scholar] [CrossRef]
  21. Begum, R.; Mohammad, A.; Tabassum, R.M.; Shaeri, N. The Role of Introns for the Development of Inflammation-Mediated Cancer Cell. In Inflammation in the 21st Century; Vijay, K., Aguilera, S.A., Shamsadin, A.S., Eds.; IntechOpen: Rijeka, Croatia, 2021; p. 9. [Google Scholar]
  22. Wislez, M.; Fujimoto, N.; Izzo, J.G.; Hanna, A.E.; Cody, D.D.; Langley, R.R.; Tang, H.; Burdick, M.D.; Sato, M.; Minna, J.D.; et al. High expression of ligands for chemokine receptor CXCR2 in alveolar epithelial neoplasia induced by oncogenic kras. Cancer Res. 2006, 66, 4198–4207. [Google Scholar] [CrossRef] [Green Version]
  23. De la Garza, M.M.; Cumpian, A.M.; Daliri, S.; Castro-Pando, S.; Umer, M.; Gong, L.; Khosravi, N.; Caetano, M.S.; Ramos-Castañeda, M.; Flores, A.G.; et al. COPD-Type lung inflammation promotes K-ras mutant lung cancer through epithelial HIF-1α mediated tumor angiogenesis and proliferation. Oncotarget 2018, 9, 32972–32983. [Google Scholar] [CrossRef] [Green Version]
  24. McGettrick, A.F.; O’Neill, L.A.J. The Role of HIF in Immunity and Inflammation. Cell Metab. 2020, 32, 524–536. [Google Scholar] [CrossRef] [PubMed]
  25. Zhang, Y.; Bian, Y.; Wang, Y.; Wang, Y.; Duan, X.; Han, Y.; Zhang, L.; Wang, F.; Gu, Z.; Qin, Z. HIF-1α is necessary for activation and tumour-promotion effect of cancer-associated fibroblasts in lung cancer. J. Cell. Mol. Med. 2021, 25, 5457–5469. [Google Scholar] [CrossRef] [PubMed]
  26. Altorki, N.K.; Markowitz, G.J.; Gao, D.; Port, J.L.; Saxena, A.; Stiles, B.; McGraw, T.; Mittal, V. The lung microenvironment: An important regulator of tumour growth and metastasis. Nat. Rev. Cancer 2019, 19, 9–31. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  27. Grivennikov, S.I.; Greten, F.R.; Karin, M. Immunity, inflammation, and cancer. Cell 2010, 140, 883–899. [Google Scholar] [CrossRef] [Green Version]
  28. Kastenmüller, W.; Kastenmüller, K.; Kurts, C.; Seder, R.A. Dendritic cell-targeted vaccines—Hope or hype? Nat. Rev. Immunol. 2014, 14, 705–711. [Google Scholar] [CrossRef]
  29. Goh, C.C.; Roggerson, K.M.; Lee, H.C.; Golden-Mason, L.; Rosen, H.R.; Hahn, Y.S. Hepatitis C Virus-Induced Myeloid-Derived Suppressor Cells Suppress NK Cell IFN-γ Production by Altering Cellular Metabolism via Arginase-1. J. Immunol. 2016, 196, 2283–2292. [Google Scholar] [CrossRef] [Green Version]
  30. Lv, M.; Wang, K.; Huang, X.-J. Myeloid-derived suppressor cells in hematological malignancies: Friends or foes. J. Hematol. Oncol. 2019, 12, 105. [Google Scholar] [CrossRef] [Green Version]
  31. Fallah, J.; Rini, B.I. HIF Inhibitors: Status of Current Clinical Development. Curr. Oncol. Rep. 2019, 21, 6. [Google Scholar] [CrossRef]
  32. Yuan, A.; Hsiao, Y.J.; Chen, H.Y.; Chen, H.W.; Ho, C.C.; Chen, Y.Y.; Liu, Y.C.; Hong, T.H.; Yu, S.L.; Chen, J.J.; et al. Opposite Effects of M1 and M2 Macrophage Subtypes on Lung Cancer Progression. Sci. Rep. 2015, 5, 14273. [Google Scholar] [CrossRef] [Green Version]
  33. Conway, E.M.; Pikor, L.A.; Kung, S.H.; Hamilton, M.J.; Lam, S.; Lam, W.L.; Bennewith, K.L. Macrophages, Inflammation, and Lung Cancer. Am. J. Respir. Crit. Care Med. 2016, 193, 116–130. [Google Scholar] [CrossRef]
  34. Arnson, Y.; Shoenfeld, Y.; Amital, H. Effects of tobacco smoke on immunity, inflammation and autoimmunity. J. Autoimmun. 2010, 34, J258–J265. [Google Scholar] [CrossRef] [PubMed]
  35. Yehya, A.H.S.; Asif, M.; Petersen, S.H.; Subramaniam, A.V.; Kono, K.; Majid, A.; Oon, C.E. Angiogenesis: Managing the Culprits behind Tumorigenesis and Metastasis. Medicina 2018, 54, 8. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  36. Jin, S.; Mutvei, A.P.; Chivukula, I.V.; Andersson, E.R.; Ramsköld, D.; Sandberg, R.; Lee, K.L.; Kronqvist, P.; Mamaeva, V.; Ostling, P.; et al. Non-canonical Notch signaling activates IL-6/JAK/STAT signaling in breast tumor cells and is controlled by p53 and IKKα/IKKβ. Oncogene 2013, 32, 4892–4902. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  37. Wang, L.; Cao, L.; Wang, H.; Liu, B.; Zhang, Q.; Meng, Z.; Wu, X.; Zhou, Q.; Xu, K. Cancer-associated fibroblasts enhance metastatic potential of lung cancer cells through IL-6/STAT3 signaling pathway. Oncotarget 2017, 8, 76116–76128. [Google Scholar] [CrossRef] [Green Version]
  38. Sionov, R.V.; Fridlender, Z.G.; Granot, Z. The Multifaceted Roles Neutrophils Play in the Tumor Microenvironment. Cancer Microenviron. 2015, 8, 125–158. [Google Scholar] [CrossRef] [Green Version]
  39. Bekaert, S.; Fillet, M.; Detry, B.; Pichavant, M.; Marée, R.; Noel, A.; Rocks, N.; Cataldo, D. Inflammation-Generated Extracellular Matrix Fragments Drive Lung Metastasis. Cancer Growth Metastasis 2017, 10, 5539. [Google Scholar] [CrossRef]
  40. El Rayes, T.; Catena, R.; Lee, S.; Stawowczyk, M.; Joshi, N.; Fischbach, C.; Powell, C.A.; Dannenberg, A.J.; Altorki, N.K.; Gao, D.; et al. Lung inflammation promotes metastasis through neutrophil protease-mediated degradation of Tsp-1. Proc. Natl. Acad. Sci. USA 2015, 112, 16000–16005. [Google Scholar] [CrossRef] [Green Version]
  41. Markowitz, G.J.; Havel, L.S.; Crowley, M.J.; Ban, Y.; Lee, S.B.; Thalappillil, J.S.; Narula, N.; Bhinder, B.; Elemento, O.; Wong, S.T.; et al. Immune reprogramming via PD-1 inhibition enhances early-stage lung cancer survival. JCI Insight 2018, 3, e96836. [Google Scholar] [CrossRef] [Green Version]
  42. Saleh, R.; Elkord, E. FoxP3(+) T regulatory cells in cancer: Prognostic biomarkers and therapeutic targets. Cancer Lett. 2020, 490, 174–185. [Google Scholar] [CrossRef]
  43. Lee, I.K.; Song, H.; Kim, H.; Kim, I.S.; Tran, N.L.; Kim, S.H.; Oh, S.J.; Lee, J.M. RORα Regulates Cholesterol Metabolism of CD8(+) T Cells for Anticancer Immunity. Cancers 2020, 12, 1733. [Google Scholar] [CrossRef]
  44. Jin, C.; Lagoudas, G.K.; Zhao, C.; Bullman, S.; Bhutkar, A.; Hu, B.; Ameh, S.; Sandel, D.; Liang, X.S.; Mazzilli, S.; et al. Commensal Microbiota Promote Lung Cancer Development via γδ T Cells. Cell 2019, 176, 998–1013.e1016. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  45. Beck, S.; Hochreiter, B.; Schmid, J.A. Extracellular Vesicles Linking Inflammation, Cancer and Thrombotic Risks. Front. Cell Dev. Biol. 2022, 10, 859863. [Google Scholar] [CrossRef] [PubMed]
  46. Wu, F.; Yin, Z.; Yang, L.; Fan, J.; Xu, J.; Jin, Y.; Yu, J.; Zhang, D.; Yang, G. Smoking Induced Extracellular Vesicles Release and Their Distinct Properties in Non-Small Cell Lung Cancer. J. Cancer 2019, 10, 3435–3443. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  47. Mohan, A.; Agarwal, S.; Clauss, M.; Britt, N.S.; Dhillon, N.K. Extracellular vesicles: Novel communicators in lung diseases. Respir. Res. 2020, 21, 175. [Google Scholar] [CrossRef] [PubMed]
  48. Yao, Z.; Fenoglio, S.; Gao, D.C.; Camiolo, M.; Stiles, B.; Lindsted, T.; Schlederer, M.; Johns, C.; Altorki, N.; Mittal, V.; et al. TGF-beta IL-6 axis mediates selective and adaptive mechanisms of resistance to molecular targeted therapy in lung cancer. Proc. Natl. Acad. Sci. USA 2010, 107, 15535–15540. [Google Scholar] [CrossRef] [Green Version]
  49. Wang, L.; Peng, Q.; Sai, B.; Zheng, L.; Xu, J.; Yin, N.; Feng, X.; Xiang, J. Ligand-independent EphB1 signaling mediates TGF-β-activated CDH2 and promotes lung cancer cell invasion and migration. J. Cancer 2020, 11, 4123–4131. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  50. Caetano, M.S.; Zhang, H.; Cumpian, A.M.; Gong, L.; Unver, N.; Ostrin, E.J.; Daliri, S.; Chang, S.H.; Ochoa, C.E.; Hanash, S.; et al. IL6 Blockade Reprograms the Lung Tumor Microenvironment to Limit the Development and Progression of K-ras-Mutant Lung Cancer. Cancer Res. 2016, 76, 3189–3199. [Google Scholar] [CrossRef] [Green Version]
  51. Qu, Z.; Sun, F.; Zhou, J.; Li, L.; Shapiro, S.D.; Xiao, G. Interleukin-6 Prevents the Initiation but Enhances the Progression of Lung Cancer. Cancer Res. 2015, 75, 3209–3215. [Google Scholar] [CrossRef] [Green Version]
  52. Lappalainen, U.; Whitsett, J.A.; Wert, S.E.; Tichelaar, J.W.; Bry, K. Interleukin-1beta causes pulmonary inflammation, emphysema, and airway remodeling in the adult murine lung. Am. J. Respir. Cell Mol. Biol. 2005, 32, 311–318. [Google Scholar] [CrossRef] [PubMed]
  53. Pretre, V.; Papadopoulos, D.; Regard, J.; Pelletier, M.; Woo, J. Interleukin-1 (IL-1) and the inflammasome in cancer. Cytokine 2022, 153, 155850. [Google Scholar] [CrossRef]
  54. Bent, R.; Moll, L.; Grabbe, S.; Bros, M. Interleukin-1 Beta-A Friend or Foe in Malignancies? Int. J. Mol. Sci. 2018, 19, 2155. [Google Scholar] [CrossRef] [Green Version]
  55. Ha, H.; Debnath, B.; Neamati, N. Role of the CXCL8-CXCR1/2 Axis in Cancer and Inflammatory Diseases. Theranostics 2017, 7, 1543–1588. [Google Scholar] [CrossRef] [PubMed]
  56. Tekpli, X.; Landvik, N.E.; Anmarkud, K.H.; Skaug, V.; Haugen, A.; Zienolddiny, S. DNA methylation at promoter regions of interleukin 1B, interleukin 6, and interleukin 8 in non-small cell lung cancer. Cancer Immunol. Immunother. 2013, 62, 337–345. [Google Scholar] [CrossRef] [PubMed]
  57. Sanmamed, M.F.; Carranza-Rua, O.; Alfaro, C.; Oñate, C.; Martín-Algarra, S.; Perez, G.; Landazuri, S.F.; Gonzalez, A.; Gross, S.; Rodriguez, I.; et al. Serum interleukin-8 reflects tumor burden and treatment response across malignancies of multiple tissue origins. Clin. Cancer Res. 2014, 20, 5697–5707. [Google Scholar] [CrossRef] [Green Version]
  58. Kim, H.J.; Kim, H.J.; Yun, J.; Kim, K.H.; Kim, S.H.; Lee, S.C.; Bae, S.B.; Kim, C.K.; Lee, N.S.; Lee, K.T.; et al. Pathophysiological role of hormones and cytokines in cancer cachexia. J. Korean Med. Sci. 2012, 27, 128–134. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  59. Greten, F.R.; Eckmann, L.; Greten, T.F.; Park, J.M.; Li, Z.W.; Egan, L.J.; Kagnoff, M.F.; Karin, M. IKKbeta links inflammation and tumorigenesis in a mouse model of colitis-associated cancer. Cell 2004, 118, 285–296. [Google Scholar] [CrossRef] [Green Version]
  60. Stathopoulos, G.T.; Kollintza, A.; Moschos, C.; Psallidas, I.; Sherrill, T.P.; Pitsinos, E.N.; Vassiliou, S.; Karatza, M.; Papiris, S.A.; Graf, D.; et al. Tumor necrosis factor-alpha promotes malignant pleural effusion. Cancer Res. 2007, 67, 9825–9834. [Google Scholar] [CrossRef] [Green Version]
  61. Van Horssen, R.; Ten Hagen, T.L.; Eggermont, A.M. TNF-alpha in cancer treatment: Molecular insights, antitumor effects, and clinical utility. Oncologist 2006, 11, 397–408. [Google Scholar] [CrossRef]
  62. Bianconi, V.; Sahebkar, A.; Atkin, S.L.; Pirro, M. The regulation and importance of monocyte chemoattractant protein-1. Curr. Opin. Hematol. 2018, 25, 44–51. [Google Scholar] [CrossRef]
  63. Yoshimura, T. The chemokine MCP-1 (CCL2) in the host interaction with cancer: A foe or ally? Cell. Mol. Immunol. 2018, 15, 335–345. [Google Scholar] [CrossRef] [Green Version]
  64. Wang, H.; Zhang, Q.; Kong, H.; Zeng, Y.; Hao, M.; Yu, T.; Peng, J.; Xu, Z.; Chen, J.; Shi, H. Monocyte chemotactic protein-1 expression as a prognosic biomarker in patients with solid tumor: A meta analysis. Int. J. Clin. Exp. Pathol. 2014, 7, 3876–3886. [Google Scholar] [PubMed]
  65. Mulholland, B.S.; Forwood, M.R.; Morrison, N.A. Monocyte Chemoattractant Protein-1 (MCP-1/CCL2) Drives Activation of Bone Remodelling and Skeletal Metastasis. Curr. Osteoporos. Rep. 2019, 17, 538–547. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  66. Kaneko, N.; Kurata, M.; Yamamoto, T.; Morikawa, S.; Masumoto, J. The role of interleukin-1 in general pathology. Inflamm. Regen. 2019, 39, 12. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  67. Chonov, D.C.; Ignatova, M.M.K.; Ananiev, J.R.; Gulubova, M.V. IL-6 Activities in the Tumour Microenvironment. Part 1. Open Access Maced. J. Med. Sci. 2019, 7, 2391–2398. [Google Scholar] [CrossRef] [Green Version]
  68. Heim, L.; Yang, Z.; Tausche, P.; Hohenberger, K.; Chiriac, M.T.; Koelle, J.; Geppert, C.I.; Kachler, K.; Miksch, S.; Graser, A.; et al. IL-9 Producing Tumor-Infiltrating Lymphocytes and Treg Subsets Drive Immune Escape of Tumor Cells in Non-Small Cell Lung Cancer. Front. Immunol. 2022, 13, 859738. [Google Scholar] [CrossRef] [PubMed]
  69. Bezel, P.; Valaperti, A.; Steiner, U.; Scholtze, D.; Wieser, S.; Vonow-Eisenring, M.; Widmer, A.; Kowalski, B.; Kohler, M.; Franzen, D.P. Evaluation of cytokines in the tumor microenvironment of lung cancer using bronchoalveolar lavage fluid analysis. Cancer Immunol. Immunother. 2021, 70, 1867–1876. [Google Scholar] [CrossRef]
  70. Ritzmann, F.; Jungnickel, C.; Vella, G.; Kamyschnikow, A.; Herr, C.; Li, D.; Menger, M.M.; Angenendt, A.; Hoth, M.; Lis, A.; et al. IL-17C-mediated innate inflammation decreases the response to PD-1 blockade in a model of Kras-driven lung cancer. Sci. Rep. 2019, 9, 10353. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  71. Melese, E.S.; Franks, E.; Cederberg, R.A.; Harbourne, B.T.; Shi, R.; Wadsworth, B.J.; Collier, J.L.; Halvorsen, E.C.; Johnson, F.; Luu, J.; et al. CCL5 production in lung cancer cells leads to an altered immune microenvironment and promotes tumor development. Oncoimmunology 2022, 11, 2010905. [Google Scholar] [CrossRef]
  72. Li, C.; Yang, J.; Wang, Y.; Qi, Y.; Yang, W.; Li, Y. Farnesoid X Receptor Agonists as Therapeutic Target for Cardiometabolic Diseases. Front. Pharmacol. 2020, 11, 1247. [Google Scholar] [CrossRef]
  73. Shi, J.; Yang, F.; Zhou, N.; Jiang, Y.; Zhao, Y.; Zhu, J.; Prelaj, A.; Malhotra, J.; Normanno, N.; Danese, E.; et al. Isochorismatase domain-containing protein 1 (ISOC1) participates in DNA damage repair and inflammation-related pathways to promote lung cancer development. Transl. Lung Cancer Res. 2021, 10, 1444–1456. [Google Scholar] [CrossRef]
  74. Serresi, M.; Siteur, B.; Hulsman, D.; Company, C.; Schmitt, M.J.; Lieftink, C.; Morris, B.; Cesaroni, M.; Proost, N.; Beijersbergen, R.L.; et al. Ezh2 inhibition in Kras-driven lung cancer amplifies inflammation and associated vulnerabilities. J. Exp. Med. 2018, 215, 3115–3135. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  75. Lebovitz, C.; Wretham, N.; Osooly, M.; Milne, K.; Dash, T.; Thornton, S.; Tessier-Cloutier, B.; Sathiyaseelan, P.; Bortnik, S.; Go, N.E.; et al. Loss of Parkinson’s susceptibility gene LRRK2 promotes carcinogen-induced lung tumorigenesis. Sci. Rep. 2021, 11, 2097. [Google Scholar] [CrossRef]
  76. Takahashi, H.; Ogata, H.; Nishigaki, R.; Broide, D.H.; Karin, M. Tobacco smoke promotes lung tumorigenesis by triggering IKKbeta- and JNK1-dependent inflammation. Cancer Cell 2010, 17, 89–97. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  77. Wallings, R.L.; Tansey, M.G. LRRK2 regulation of immune-pathways and inflammatory disease. Biochem. Soc. Trans. 2019, 47, 1581–1595. [Google Scholar] [CrossRef] [PubMed]
  78. Shutinoski, B.; Hakimi, M.; Harmsen, I.E.; Lunn, M.; Rocha, J.; Lengacher, N.; Zhou, Y.Y.; Khan, J.; Nguyen, A.; Hake-Volling, Q.; et al. Lrrk2 alleles modulate inflammation during microbial infection of mice in a sex-dependent manner. Sci. Transl. Med. 2019, 11, eaas9292. [Google Scholar] [CrossRef] [PubMed]
  79. Wojewska, D.N.; Kortholt, A. LRRK2 Targeting Strategies as Potential Treatment of Parkinson’s Disease. Biomolecules 2021, 11, 1101. [Google Scholar] [CrossRef]
  80. Cheng, L.; Zhao, Y.; Tang, M.; Luo, Z.; Wang, X. Knockdown of ISOC1 suppresses cell proliferation in pancreatic cancer in vitro. Oncol. Lett. 2019, 17, 4263–4270. [Google Scholar] [CrossRef] [Green Version]
  81. Gao, B.; Zhao, L.; Wang, F.; Bai, H.; Li, J.; Li, M.; Hu, X.; Cao, J.; Wang, G. Knockdown of ISOC1 inhibits the proliferation and migration and induces the apoptosis of colon cancer cells through the AKT/GSK-3β pathway. Carcinogenesis 2020, 41, 1123–1133. [Google Scholar] [CrossRef] [Green Version]
  82. Duan, R.; Du, W.; Guo, W. EZH2: A novel target for cancer treatment. J. Hematol. Oncol. 2020, 13, 104. [Google Scholar] [CrossRef]
  83. Dutta, P.; Sabri, N.; Li, J.; Li, W.X. Role of STAT3 in lung cancer. Jakstat 2014, 3, e999503. [Google Scholar] [CrossRef]
  84. Siveen, K.S.; Sikka, S.; Surana, R.; Dai, X.; Zhang, J.; Kumar, A.P.; Tan, B.K.; Sethi, G.; Bishayee, A. Targeting the STAT3 signaling pathway in cancer: Role of synthetic and natural inhibitors. Biochim. Biophys. Acta 2014, 1845, 136–154. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  85. Looyenga, B.D.; Hutchings, D.; Cherni, I.; Kingsley, C.; Weiss, G.J.; Mackeigan, J.P. STAT3 is activated by JAK2 independent of key oncogenic driver mutations in non-small cell lung carcinoma. PLoS ONE 2012, 7, e30820. [Google Scholar] [CrossRef] [PubMed]
  86. 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] [PubMed]
  87. Baig, M.S.; Roy, A.; Saqib, U.; Rajpoot, S.; Srivastava, M.; Naim, A.; Liu, D.; Saluja, R.; Faisal, S.M.; Pan, Q.; et al. Repurposing Thioridazine (TDZ) as an anti-inflammatory agent. Sci. Rep. 2018, 8, 12471. [Google Scholar] [CrossRef] [Green Version]
  88. Ciarcia, R.; Damiano, S.; Montagnaro, S.; Pagnini, U.; Ruocco, A.; Caparrotti, G.; d’Angelo, D.; Boffo, S.; Morales, F.; Rizzolio, F.; et al. Combined effects of PI3K and SRC kinase inhibitors with imatinib on intracellular calcium levels, autophagy, and apoptosis in CML-PBL cells. Cell Cycle 2013, 12, 2839–2848. [Google Scholar] [CrossRef] [Green Version]
  89. Roberti, A.; Chaffey, L.E.; Greaves, D.R. NF-κB Signaling and Inflammation-Drug Repurposing to Treat Inflammatory Disorders? Biology 2022, 11, 372. [Google Scholar] [CrossRef]
  90. Bernal-Bello, D.; Jaenes-Barrios, B.; Morales-Ortega, A.; Ruiz-Giardin, J.M.; García-Bermúdez, V.; Frutos-Pérez, B.; Farfán-Sedano, A.I.; de Ancos-Aracil, C.; Bermejo, F.; García-Gil, M.; et al. Imatinib might constitute a treatment option for lung involvement in COVID-19. Autoimmun. Rev. 2020, 19, 102565. [Google Scholar] [CrossRef] [PubMed]
  91. Boussios, S.; Pentheroudakis, G.; Katsanos, K.; Pavlidis, N. Systemic treatment-induced gastrointestinal toxicity: Incidence, clinical presentation and management. Ann. Gastroenterol. 2012, 25, 106–118. [Google Scholar]
  92. Tsao, A.S.; Liu, S.; Fujimoto, J.; Wistuba, I.I.; Lee, J.J.; Marom, E.M.; Charnsangavej, C.; Fossella, F.V.; Tran, H.T.; Blumenschein, G.R.; et al. Phase II trials of imatinib mesylate and docetaxel in patients with metastatic non-small cell lung cancer and head and neck squamous cell carcinoma. J. Thorac. Oncol. 2011, 6, 2104–2111. [Google Scholar] [CrossRef] [Green Version]
  93. Brenner, D.R.; Fanidi, A.; Grankvist, K.; Muller, D.C.; Brennan, P.; Manjer, J.; Byrnes, G.; Hodge, A.; Severi, G.; Giles, G.G.; et al. Inflammatory Cytokines and Lung Cancer Risk in 3 Prospective Studies. Am. J. Epidemiol. 2017, 185, 86–95. [Google Scholar] [CrossRef] [Green Version]
  94. Choy, E.H.; De Benedetti, F.; Takeuchi, T.; Hashizume, M.; John, M.R.; Kishimoto, T. Translating IL-6 biology into effective treatments. Nat. Rev. Rheumatol. 2020, 16, 335–345. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  95. Mariette, X.; Hermine, O.; Tharaux, P.-L.; Resche-Rigon, M.; Porcher, R.; Ravaud, P.; Bureau, S.; Dougados, M.; Tibi, A.; Azoulay, E.; et al. Sarilumab in adults hospitalised with moderate-to-severe COVID-19 pneumonia (CORIMUNO-SARI-1): An open-label randomised controlled trial. Lancet Rheumatol. 2022, 4, e24–e32. [Google Scholar] [CrossRef]
  96. Salton, F.; Confalonieri, P.; Campisciano, G.; Cifaldi, R.; Rizzardi, C.; Generali, D.; Pozzan, R.; Tavano, S.; Bozzi, C.; Lapadula, G.; et al. Cytokine Profiles as Potential Prognostic and Therapeutic Markers in SARS-CoV-2-Induced ARDS. J. Clin. Med. 2022, 11, 2951. [Google Scholar] [CrossRef]
  97. Timpani, C.A.; Rybalka, E. Calming the (Cytokine) Storm: Dimethyl Fumarate as a Therapeutic Candidate for COVID-19. Pharmaceuticals 2020, 14, 15. [Google Scholar] [CrossRef] [PubMed]
  98. Moll, M.; Kuemmerle-Deschner, J.B. Inflammasome and cytokine blocking strategies in autoinflammatory disorders. Clin. Immunol. 2013, 147, 242–275. [Google Scholar] [CrossRef] [PubMed]
  99. Lythgoe, M.P.; Prasad, V. Repositioning canakinumab for non-small cell lung cancer-important lessons for drug repurposing in oncology. Br. J. Cancer 2022, 127, 785–787. [Google Scholar] [CrossRef] [PubMed]
  100. Qin, W.; Hu, L.; Zhang, X.; Jiang, S.; Li, J.; Zhang, Z.; Wang, X. The Diverse Function of PD-1/PD-L Pathway Beyond Cancer. Front. Immunol. 2019, 10, 2298. [Google Scholar] [CrossRef] [Green Version]
  101. 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]
  102. Stabile, L.P.; Farooqui, M.; Kanterewicz, B.; Abberbock, S.; Kurland, B.F.; Diergaarde, B.; Siegfried, J.M. Preclinical Evidence for Combined Use of Aromatase Inhibitors and NSAIDs as Preventive Agents of Tobacco-Induced Lung Cancer. J. Thorac. Oncol. 2018, 13, 399–412. [Google Scholar] [CrossRef] [Green Version]
  103. Amin, F.; Fathi, F.; Reiner, Ž.; Banach, M.; Sahebkar, A. The role of statins in lung cancer. Arch. Med. Sci. 2022, 18, 141–152. [Google Scholar] [CrossRef]
  104. Coimbra, M.; Banciu, M.; Fens, M.H.; de Smet, L.; Cabaj, M.; Metselaar, J.M.; Storm, G.; Schiffelers, R.M. Liposomal pravastatin inhibits tumor growth by targeting cancer-related inflammation. J. Control. Release 2010, 148, 303–310. [Google Scholar] [CrossRef] [PubMed]
  105. Raymakers, A.; Sin, D.D.; Sadatsafavi, M.; FitzGerald, J.M.; Marra, C.A.; Lynd, L.D. Statin use and lung cancer risk in chronic obstructive pulmonary disease patients: A population-based cohort study. Respir. Res. 2020, 21, 118. [Google Scholar] [CrossRef]
  106. Huwiler, A.; Zangemeister-Wittke, U. The sphingosine 1-phosphate receptor modulator fingolimod as a therapeutic agent: Recent findings and new perspectives. Pharmacol. Ther. 2018, 185, 34–49. [Google Scholar] [CrossRef] [PubMed]
  107. Gendron, D.R.; Lemay, A.M.; Lecours, P.B.; Perreault-Vallières, V.; Huppé, C.A.; Bossé, Y.; Blanchet, M.R.; Dion, G.; Marsolais, D. FTY720 promotes pulmonary fibrosis when administered during the remodelling phase following a bleomycin-induced lung injury. Pulm. Pharmacol. Ther. 2017, 44, 50–56. [Google Scholar] [CrossRef] [PubMed]
  108. Bradbury, P.; Traini, D.; Ammit, A.J.; Young, P.M.; Ong, H.X. Repurposing of statins via inhalation to treat lung inflammatory conditions. Adv. Drug Deliv. Rev. 2018, 133, 93–106. [Google Scholar] [CrossRef] [PubMed]
  109. Kumar, V.; Harjai, K.; Chhibber, S. Thalidomide treatment modulates macrophage pro-inflammatory function and cytokine levels in Klebsiella pneumoniae B5055 induced pneumonia in BALB/c mice. Int. Immunopharmacol. 2010, 10, 777–783. [Google Scholar] [CrossRef] [PubMed]
  110. Wang, P.; Yuan, Y.; Lin, W.; Zhong, H.; Xu, K.; Qi, X. Roles of sphingosine-1-phosphate signaling in cancer. Cancer Cell Int. 2019, 19, 295. [Google Scholar] [CrossRef] [PubMed]
  111. Zhang, Z.; Chen, F.; Shang, L. Advances in antitumor effects of NSAIDs. Cancer Manag. Res. 2018, 10, 4631–4640. [Google Scholar] [CrossRef] [Green Version]
  112. Malcova, H.; Strizova, Z.; Milota, T.; Striz, I.; Sediva, A.; Cebecauerova, D.; Horvath, R. IL-1 Inhibitors in the Treatment of Monogenic Periodic Fever Syndromes: From the Past to the Future Perspectives. Front. Immunol. 2020, 11, 619257. [Google Scholar] [CrossRef]
  113. Yang, Y.; Li, N.; Wang, T.M.; Di, L. Natural Products with Activity against Lung Cancer: A Review Focusing on the Tumor Microenvironment. Int. J. Mol. Sci. 2021, 22, 10827. [Google Scholar] [CrossRef]
  114. Zareie, A.; Soleimani, D.; Askari, G.; Jamialahmadi, T.; Guest, P.C.; Bagherniya, M.; Sahebkar, A. Cinnamon: A Promising Natural Product Against COVID-19. Adv. Exp. Med. Biol. 2021, 1327, 191–195. [Google Scholar] [CrossRef] [PubMed]
  115. Wu, H.C.; Horng, C.T.; Lee, Y.L.; Chen, P.N.; Lin, C.Y.; Liao, C.Y.; Hsieh, Y.S.; Chu, S.C. Cinnamomum Cassia Extracts Suppress Human Lung Cancer Cells Invasion by Reducing u-PA/MMP Expression through the FAK to ERK Pathways. Int. J. Med. Sci. 2018, 15, 115–123. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  116. Park, J.; Baek, S.H. Combination Therapy with Cinnamaldehyde and Hyperthermia Induces Apoptosis of A549 Non-Small Cell Lung Carcinoma Cells via Regulation of Reactive Oxygen Species and Mitogen-Activated Protein Kinase Family. Int. J. Mol. Sci. 2020, 21, 6229. [Google Scholar] [CrossRef] [PubMed]
  117. Isago, H.; Mitani, A.; Kohno, S.; Nagoshi, S.; Ishimori, T.; Saito, M.; Tamiya, H.; Miyashita, N.; Ishii, T.; Matsuzaki, H.; et al. The Japanese Herbal (Kampo) Medicine Hochuekkito Attenuates Lung Inflammation in Lung Emphysema. Biol. Pharm. Bull. 2021, 44, 39–45. [Google Scholar] [CrossRef]
  118. Vetvicka, V.; Vannucci, L. Biological properties of andrographolide, an active ingredient of Andrographis Paniculata: A narrative review. Ann. Transl. Med. 2021, 9, 1186. [Google Scholar] [CrossRef]
  119. He, W.; Sun, J.; Zhang, Q.; Li, Y.; Fu, Y.; Zheng, Y.; Jiang, X. Andrographolide exerts anti-inflammatory effects in Mycobacterium tuberculosis-infected macrophages by regulating the Notch1/Akt/NF-κB axis. J. Leukoc. Biol. 2020, 108, 1747–1764. [Google Scholar] [CrossRef] [PubMed]
  120. Burgos, R.A.; Alarcón, P.; Quiroga, J.; Manosalva, C.; Hancke, J. Andrographolide, an Anti-Inflammatory Multitarget Drug: All Roads Lead to Cellular Metabolism. Molecules 2020, 26, 5. [Google Scholar] [CrossRef]
  121. Tsai, H.R.; Yang, L.M.; Tsai, W.J.; Chiou, W.F. Andrographolide acts through inhibition of ERK1/2 and Akt phosphorylation to suppress chemotactic migration. Eur. J. Pharmacol. 2004, 498, 45–52. [Google Scholar] [CrossRef]
  122. Farooqi, A.A.; Attar, R.; Sabitaliyevich, U.Y.; Alaaeddine, N.; de Sousa, D.P.; Xu, B.; Cho, W.C. The Prowess of Andrographolide as a Natural Weapon in the War against Cancer. Cancers 2020, 12, 2159. [Google Scholar] [CrossRef]
  123. Chen, Z.; Tang, W.J.; Zhou, Y.H.; Chen, Z.M.; Liu, K. Andrographolide inhibits non-small cell lung cancer cell proliferation through the activation of the mitochondrial apoptosis pathway and by reprogramming host glucose metabolism. Ann. Transl. Med. 2021, 9, 1701. [Google Scholar] [CrossRef]
  124. Li, J.; Huang, L.; He, Z.; Chen, M.; Ding, Y.; Yao, Y.; Duan, Y.; Zixuan, L.; Qi, C.; Zheng, L.; et al. Andrographolide Suppresses the Growth and Metastasis of Luminal-Like Breast Cancer by Inhibiting the NF-κB/miR-21-5p/PDCD4 Signaling Pathway. Front. Cell Dev. Biol. 2021, 9, 643525. [Google Scholar] [CrossRef] [PubMed]
  125. Luo, W.; Jia, L.; Zhang, J.W.; Wang, D.J.; Ren, Q.; Zhang, W. Andrographolide Against Lung Cancer-New Pharmacological Insights Based on High-Throughput Metabolomics Analysis Combined with Network Pharmacology. Front. Pharmacol. 2021, 12, 596652. [Google Scholar] [CrossRef] [PubMed]
  126. Mi, S.; Xiang, G.; Yuwen, D.; Gao, J.; Guo, W.; Wu, X.; Wu, X.; Sun, Y.; Su, Y.; Shen, Y.; et al. Inhibition of autophagy by andrographolide resensitizes cisplatin-resistant non-small cell lung carcinoma cells via activation of the Akt/mTOR pathway. Toxicol. Appl. Pharmacol. 2016, 310, 78–86. [Google Scholar] [CrossRef]
  127. Surien, O.; Ghazali, A.R.; Masre, S.F. Chemopreventive effects of pterostilbene through p53 and cell cycle in mouse lung of squamous cell carcinoma model. Sci. Rep. 2021, 11, 14862. [Google Scholar] [CrossRef]
  128. Huang, W.C.; Chan, M.L.; Chen, M.J.; Tsai, T.H.; Chen, Y.J. Modulation of macrophage polarization and lung cancer cell stemness by MUC1 and development of a related small-molecule inhibitor pterostilbene. Oncotarget 2016, 7, 39363–39375. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  129. Wu, C.Y.; Cherng, J.Y.; Yang, Y.H.; Lin, C.L.; Kuan, F.C.; Lin, Y.Y.; Lin, Y.S.; Shu, L.H.; Cheng, Y.C.; Liu, H.T.; et al. Danshen improves survival of patients with advanced lung cancer and targeting the relationship between macrophages and lung cancer cells. Oncotarget 2017, 8, 90925–90947. [Google Scholar] [CrossRef] [Green Version]
  130. Li, H.; Huang, N.; Zhu, W.; Wu, J.; Yang, X.; Teng, W.; Tian, J.; Fang, Z.; Luo, Y.; Chen, M.; et al. Modulation the crosstalk between tumor-associated macrophages and non-small cell lung cancer to inhibit tumor migration and invasion by ginsenoside Rh2. BMC Cancer 2018, 18, 579. [Google Scholar] [CrossRef] [Green Version]
  131. Chen, Y.; Zhang, Y.; Song, W.; Zhang, Y.; Dong, X.; Tan, M. Ginsenoside Rh2 Inhibits Migration of Lung Cancer Cells under Hypoxia via mir-491. Anticancer Agents Med. Chem. 2019, 19, 1633–1641. [Google Scholar] [CrossRef]
  132. Kloc, M.; Ghobrial, R.M.; Lipińska-Opałka, A.; Wawrzyniak, A.; Zdanowski, R.; Kalicki, B.; Kubiak, J.Z. Effects of vitamin D on macrophages and myeloid-derived suppressor cells (MDSCs) hyperinflammatory response in the lungs of COVID-19 patients. Cell. Immunol. 2021, 360, 104259. [Google Scholar] [CrossRef]
  133. Akutsu, T.; Kitamura, H.; Himeiwa, S.; Kitada, S.; Akasu, T.; Urashima, M. Vitamin D and Cancer Survival: Does Vitamin D Supplementation Improve the Survival of Patients with Cancer? Curr. Oncol. Rep. 2020, 22, 62. [Google Scholar] [CrossRef]
  134. Akiba, T.; Morikawa, T.; Odaka, M.; Nakada, T.; Kamiya, N.; Yamashita, M.; Yabe, M.; Inagaki, T.; Asano, H.; Mori, S.; et al. Vitamin D Supplementation and Survival of Patients with Non-small Cell Lung Cancer: A Randomized, Double-Blind, Placebo-Controlled Trial. Clin. Cancer Res. 2018, 24, 4089–4097. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  135. Li, J.; Fan, Y.; Zhang, Y.; Liu, Y.; Yu, Y.; Ma, M. Resveratrol Induces Autophagy and Apoptosis in Non-Small-Cell Lung Cancer Cells by Activating the NGFR-AMPK-mTOR Pathway. Nutrients 2022, 14, 2413. [Google Scholar] [CrossRef] [PubMed]
  136. Zhang, X.; Ruan, Q.; Zhai, Y.; Lu, D.; Li, C.; Fu, Y.; Zheng, Z.; Song, Y.; Guo, J. Baicalein inhibits non-small-cell lung cancer invasion and metastasis by reducing ezrin tension in inflammation microenvironment. Cancer Sci. 2020, 111, 3802–3812. [Google Scholar] [CrossRef]
  137. Wang, G.; Mohammadtursun, N.; Lv, Y.; Zhang, H.; Sun, J.; Dong, J. Baicalin Exerts Anti-Airway Inflammation and Anti-Remodelling Effects in Severe Stage Rat Model of Chronic Obstructive Pulmonary Disease. Evid. Based Complement. Alternat. Med. 2018, 2018, 7591348. [Google Scholar] [CrossRef]
  138. Xu, T.; Ge, X.; Lu, C.; Dai, W.; Chen, H.; Xiao, Z.; Wu, L.; Liang, G.; Ying, S.; Zhang, Y.; et al. Baicalein attenuates OVA-induced allergic airway inflammation through the inhibition of the NF-κB signaling pathway. Aging 2019, 11, 9310–9327. [Google Scholar] [CrossRef] [PubMed]
  139. Kourakis, S.; Timpani, C.A.; de Haan, J.B.; Gueven, N.; Fischer, D.; Rybalka, E. Dimethyl Fumarate and Its Esters: A Drug with Broad Clinical Utility? Pharmaceuticals 2020, 13, 306. [Google Scholar] [CrossRef]
  140. Cattani-Cavalieri, I.; da Maia Valença, H.; Moraes, J.A.; Brito-Gitirana, L.; Romana-Souza, B.; Schmidt, M.; Valença, S.S. Dimethyl Fumarate Attenuates Lung Inflammation and Oxidative Stress Induced by Chronic Exposure to Diesel Exhaust Particles in Mice. Int. J. Mol. Sci. 2020, 21, 9658. [Google Scholar] [CrossRef]
  141. Ashrafizadeh, M.; Najafi, M.; Makvandi, P.; Zarrabi, A.; Farkhondeh, T.; Samarghandian, S. Versatile role of curcumin and its derivatives in lung cancer therapy. J. Cell. Physiol. 2020, 235, 9241–9268. [Google Scholar] [CrossRef]
  142. Tsai, J.R.; Liu, P.L.; Chen, Y.H.; Chou, S.H.; Cheng, Y.J.; Hwang, J.J.; Chong, I.W. Curcumin Inhibits Non-Small Cell Lung Cancer Cells Metastasis through the Adiponectin/NF-κb/MMPs Signaling Pathway. PLoS ONE 2015, 10, e0144462. [Google Scholar] [CrossRef] [Green Version]
  143. Su, C.C.; Wang, S.C.; Chen, I.C.; Chiu, F.Y.; Liu, P.L.; Huang, C.H.; Huang, K.H.; Fang, S.H.; Cheng, W.C.; Huang, S.P.; et al. Zerumbone Suppresses the LPS-Induced Inflammatory Response and Represses Activation of the NLRP3 Inflammasome in Macrophages. Front. Pharmacol. 2021, 12, 652860. [Google Scholar] [CrossRef]
  144. Radaei, Z.; Zamani, A.; Najafi, R.; Saidijam, M.; Jalilian, F.A.; Ezati, R.; Solgi, G.; Amini, R. Promising Effects of Zerumbone on the Regulation of Tumor-promoting Cytokines Induced by TNF-α-activated Fibroblasts. Curr. Med. Sci. 2020, 40, 1075–1084. [Google Scholar] [CrossRef] [PubMed]
  145. Cheng, J.; Eroglu, A. The Promising Effects of Astaxanthin on Lung Diseases. Adv. Nutr. 2021, 12, 850–864. [Google Scholar] [CrossRef] [PubMed]
  146. Wu, C.; Zhang, J.; Liu, T.; Jiao, G.; Li, C.; Hu, B. Astaxanthin inhibits proliferation and promotes apoptosis of A549 lung cancer cells via blocking JAK1/STAT3 pathway. Xi Bao Yu Fen Zi Mian Yi Xue Za Zhi 2016, 32, 784–788. [Google Scholar]
  147. Akduman, H.; Tayman, C.; Çakir, U.; Çakir, E.; Dilli, D.; Türkmenoğlu, T.T.; Gönel, A. Astaxanthin Prevents Lung Injury Due to Hyperoxia and Inflammation. Comb. Chem. High Throughput Screen. 2021, 24, 1243–1250. [Google Scholar] [CrossRef]
  148. You, W.; Li, L.; Sun, D.; Liu, X.; Xia, Z.; Xue, S.; Chen, B.; Qin, H.; Ai, J.; Jiang, H. Farnesoid X Receptor Constructs an Immunosuppressive Microenvironment and Sensitizes FXR(high)PD-L1(low) NSCLC to Anti-PD-1 Immunotherapy. Cancer Immunol. Res. 2019, 7, 990–1000. [Google Scholar] [CrossRef]
  149. Almatroodi, S.A.; Alrumaihi, F.; Alsahli, M.A.; Alhommrani, M.F.; Khan, A.; Rahmani, A.H. Curcumin, an Active Constituent of Turmeric Spice: Implication in the Prevention of Lung Injury Induced by Benzo(a) Pyrene (BaP) in Rats. Molecules 2020, 25, 724. [Google Scholar] [CrossRef] [Green Version]
  150. Chai, Y.S.; Chen, Y.Q.; Lin, S.H.; Xie, K.; Wang, C.J.; Yang, Y.Z.; Xu, F. Curcumin regulates the differentiation of naïve CD4+T cells and activates IL-10 immune modulation against acute lung injury in mice. Biomed. Pharmacother. 2020, 125, 109946. [Google Scholar] [CrossRef]
  151. Hasan, M.; Paul, N.C.; Paul, S.K.; Saikat, A.S.M.; Akter, H.; Mandal, M.; Lee, S.S. Natural Product-Based Potential Therapeutic Interventions of Pulmonary Fibrosis. Molecules 2022, 27, 1481. [Google Scholar] [CrossRef]
  152. Mehta, P.P.; Dhapte-Pawar, V.S. Repurposing drug molecules for new pulmonary therapeutic interventions. Drug Deliv. Transl. Res. 2021, 11, 1829–1848. [Google Scholar] [CrossRef] [PubMed]
  153. To, K.K.; Cho, W.C. Drug Repurposing in Cancer Therapy: Approached and Applications; Academic Press: Cambridge, MA, USA, 2020. [Google Scholar]
  154. Talevi, A.; Bellera, C.L. Challenges and opportunities with drug repurposing: Finding strategies to find alternative uses of therapeutics. Expert Opin. Drug Discov. 2020, 15, 397–401. [Google Scholar] [CrossRef] [Green Version]
  155. Bindu, S.; Mazumder, S.; Bandyopadhyay, U. Non-steroidal anti-inflammatory drugs (NSAIDs) and organ damage: A current perspective. Biochem. Pharmacol. 2020, 180, 114147. [Google Scholar] [CrossRef] [PubMed]
  156. Claus, S.; Weiler, C.; Schiewe, J.; Friess, W. How can we bring high drug doses to the lung? Eur. J. Pharm. Biopharm. 2014, 86, 1–6. [Google Scholar] [CrossRef] [PubMed]
  157. Patton, J.S.; Byron, P.R. Inhaling medicines: Delivering drugs to the body through the lungs. Nat. Rev. Drug Discov. 2007, 6, 67–74. [Google Scholar] [CrossRef] [PubMed]
  158. Anzar, N.; Mirza, M.A.; Anwer, K.; Khuroo, T.; Alshetaili, A.S.; Alshahrani, S.M.; Meena, J.; Hasan, N.; Talegaonkar, S.; Panda, A.K.; et al. Preparation, evaluation and pharmacokinetic studies of spray dried PLGA polymeric submicron particles of simvastatin for the effective treatment of breast cancer. J. Mol. Liq. 2018, 249, 609–616. [Google Scholar] [CrossRef]
  159. Lee, W.H.; Loo, C.Y.; Ghadiri, M.; Leong, C.R.; Young, P.M.; Traini, D. The potential to treat lung cancer via inhalation of repurposed drugs. Adv. Drug Deliv. Rev. 2018, 133, 107–130. [Google Scholar] [CrossRef]
  160. Kumbhar, P.; Manjappa, A.; Shah, R.; Jha, N.K.; Singh, S.K.; Dua, K.; Disouza, J.; Patravale, V. Inhalation delivery of repurposed drugs for lung cancer: Approaches, benefits and challenges. J. Control. Release 2022, 341, 1–15. [Google Scholar] [CrossRef]
  161. Guagliardo, R.; Pérez-Gil, J.; De Smedt, S.; Raemdonck, K. Pulmonary surfactant and drug delivery: Focusing on the role of surfactant proteins. J. Control. Release 2018, 291, 116–126. [Google Scholar] [CrossRef] [PubMed]
  162. Ahmed, S.; Fahad, S.H.B.; Nesamony, J. Mechanism and Ways of Pulmonary Drug Administration. In Handbook of Lung Targeted Drug Delivery Systems; Pathak, N.I.Y., Ed.; CRC Press: Boca Raton, FL, USA, 2021. [Google Scholar]
  163. He, Y.; Liang, Y.; Han, R.; Lu, W.L.; Mak, J.C.W.; Zheng, Y. Rational particle design to overcome pulmonary barriers for obstructive lung diseases therapy. J. Control. Release 2019, 314, 48–61. [Google Scholar] [CrossRef]
  164. Bäckman, P.; Arora, S.; Couet, W.; Forbes, B.; de Kruijf, W.; Paudel, A. Advances in experimental and mechanistic computational models to understand pulmonary exposure to inhaled drugs. Eur. J. Pharm. Sci. 2018, 113, 41–52. [Google Scholar] [CrossRef] [Green Version]
  165. Zellnitz, S.; Zellnitz, L.; Müller, M.T.; Meindl, C.; Schröttner, H.; Fröhlich, E. Impact of drug particle shape on permeability and cellular uptake in the lung. Eur. J. Pharm. Sci. 2019, 139, 105065. [Google Scholar] [CrossRef]
  166. Hou, X.; Zaks, T.; Langer, R.; Dong, Y. Lipid nanoparticles for mRNA delivery. Nat. Rev. Mater. 2021, 6, 1078–1094. [Google Scholar] [CrossRef] [PubMed]
  167. Tiwari, N.; Sivakumar, A. Biomedical application of microemulsion delivery systems: A review. J. Res. Pharm. 2022, 26, 1052–1064. [Google Scholar] [CrossRef]
  168. İsar, S.; Akbaba, H.; Akbaba, G.E.; Baspinar, Y. Development and characterization of cationic nanoemulsions as non-viral vectors for plasmid DNA delivery. J. Res. Pharm. 2020, 24, 952–960. [Google Scholar] [CrossRef]
  169. Gbian, D.L.; Omri, A. Lipid-Based Drug Delivery Systems for Diseases Managements. Biomedicines 2022, 10, 2137. [Google Scholar] [CrossRef]
  170. Ajeeshkumar, K.K.; Aneesh, P.A.; Raju, N.; Suseela, M.; Ravishankar, C.N.; Benjakul, S. Advancements in liposome technology: Preparation techniques and applications in food, functional foods, and bioactive delivery: A review. Compr. Rev. Food Sci. Food Saf. 2021, 20, 1280–1306. [Google Scholar] [CrossRef]
  171. Loh, J.S.; Tan, L.K.S.; Lee, W.L.; Ming, L.C.; How, C.W.; Foo, J.B.; Kifli, N.; Goh, B.H.; Ong, Y.S. Do Lipid-based Nanoparticles Hold Promise for Advancing the Clinical Translation of Anticancer Alkaloids? Cancers 2021, 13, 5346. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Neutrophils have a significant impact on early event of acute lung inflammation. The formation of granulation tissue, which is made up of cellular matrix, fibroblasts, endothelial cells, and leukocytes, is orchestrated by neutrophil migration and chemokine production during acute inflammation. Inflammation becomes chronic when acute inflammation is not treated. Other inflammatory immune cells, such as macrophages and lymphocytes, intensify lung inflammation during chronic inflammation, increasing the risk of cancer by fostering tumorigenesis at all stages, including initiation, invasion, and metastasis. Lung cancer progression is also aided by inflammation because it supplies the tumor microenvironment with vital molecules by the help of extracellular vesicles.
Figure 1. Neutrophils have a significant impact on early event of acute lung inflammation. The formation of granulation tissue, which is made up of cellular matrix, fibroblasts, endothelial cells, and leukocytes, is orchestrated by neutrophil migration and chemokine production during acute inflammation. Inflammation becomes chronic when acute inflammation is not treated. Other inflammatory immune cells, such as macrophages and lymphocytes, intensify lung inflammation during chronic inflammation, increasing the risk of cancer by fostering tumorigenesis at all stages, including initiation, invasion, and metastasis. Lung cancer progression is also aided by inflammation because it supplies the tumor microenvironment with vital molecules by the help of extracellular vesicles.
Pharmaceuticals 16 00451 g001
Figure 2. Chemical structures of the drug to be repurposed.
Figure 2. Chemical structures of the drug to be repurposed.
Pharmaceuticals 16 00451 g002aPharmaceuticals 16 00451 g002bPharmaceuticals 16 00451 g002c
Figure 3. Chemical structures of the natural compounds to be repurposed.
Figure 3. Chemical structures of the natural compounds to be repurposed.
Pharmaceuticals 16 00451 g003
Table 1. List of common targets of inflammation in NSCLC.
Table 1. List of common targets of inflammation in NSCLC.
TargetsRole in Lung Cancer InflammationReferences
Inflammatory cells
NeutrophilsRelease proteases which degrade Tsp-1 and promotes tumor metastasis[40]
MacrophagesGenerate reactive oxygen and nitrogen intermediates, which induces DNA damage in proliferating cells, leading to neoplastic transformation[15,21,33]
Myeloid derived suppressor cells (MDSC)Degrade L-arginine, produce ROS, and secrete anti-inflammatory cytokines, such as IL-10 and TGF-β, to suppress the activity of other immune cells[29,30]
Gamma-delta (γδ) T cellsProduce and release IL-17 and other effector molecules, which promotes inflammation and tumor proliferation[44]
FibroblastsProduce and release inflammatory cytokines, such as MCP-1 and IL-6, in the tumor microenvironment[37]
Inflammatory cytokines
IL-1βIncreased IL-1β expression is linked to aggressive tumor biology and tumor invasiveness[66]
IL-4
IL-6Produced by macrophages, T-lymphocytes, B-lymphocytes, and monocytes and promotes tumor cell proliferation, angiogenesis invasion, and metastasis[67]
IL-8Produced by endothelial cells, epithelial cells, and fibroblasts to promote angiogenesis, proliferation, and cancer cell invasion[67]
IL-9Through its effects on tumor-infiltrating T cells and tumor cell survival, promotes immune escape of lung tumor cells[68]
IL-13IL-13 has been linked to lung cancer metastasis and progression[69]
IL-17CPromotes tumorigenesis in Kras-driven lung cancer by inducing inflammation[70]
CCL5CCL5 production changes the immune microenvironment and encourages tumor growth[71]
HIF-1αKey mediator of adaptation to hypoxic condition and promotes tumorigenesis via inflammation[25]
TNF-αTumor necrosis factor-alpha (TNF-α) controls inflammation and tumor development in non-small cell lung cancer (NSCLC)[72]
Inflammatory gene expressions
ISOC1Participates in DNA damage repair and inflammation to promote lung cancer development[73]
Ezh2Ezh2 inhibition amplifies inflammation in Kras-driven lung cancer[74]
LRRK2Loss of LRRK2 promotes tumor initiation and size (tumorigenesis)[75]
Signaling Proteins
NF-ĸBPromotes tumor formation by inducing inflammation[76]
JAK/STAT3[36]
JNK1[76]
Table 2. List of existing drugs with anti-inflammation properties for NSCLC treatment.
Table 2. List of existing drugs with anti-inflammation properties for NSCLC treatment.
DrugMechanism of ActionInitial PurposePerformance RemarksReference
Statins
-
Simvastatin
-
Pravastatin
-
Rosuvastatin
-
Pitavastatin
-
Atorvastatin
Inhibits 3-hydroxy-3-methylglutaryl coenzyme A (HMG CoA) reductaseTo treat hypercholesterolemiaIn vivo—Atorvastatin showed better anti-inflammatory properties than simvastatin[108]
ThioridazineInhibits IκBα protein degradation, NF-ĸB activationAnti-psychotic drug against schizophreniaIn vivo—potent anti-inflammatory target specific drug[87]
ThalidomideInhibits the production of pro-inflammatory cytokines (TNF-α, IL-1α)To treat morning sickness in pregnant womenIn vivo—significant reduction in pro-inflammatory cytokines in pneumonia-induced acute lung inflammation[109]
Fingolimod (FTY720)Inhibits SphK/S1P signaling and S1PR3 in lung cancer metastasisTo treat multiple sclerosis [110]
AnastrozoleIn combination with non-steroidal anti-inflammatory drug (Aspirin) to reduce circulating Beta-estradiol, pro-inflammatory cytokines, and macrophages recruitment in a tobacco induced lung cancer model Hormone therapyIn vivo—downregulation of SOX-2 expression in the lungs[102]
NSAIDS
-
Aspirin
-
Aspirin/Naproxen
-
Sulindac Acid
-
Amino salicylic acid
-
Celecoxib
  • Downregulate COX-2 expression
  • Upregulation of bax expression and downregulation of bcl-2 expression
  • Degrade antiapoptotic protein bcl-XL
  • Increase ROS
  • Increase p53 gene expression
To treat inflammation, antipyretic, analgesics [111]
Tyrosine Kinase Inhibitor
-
Imatinib mesylate
Inhibits LPS-induced production of TNF-a, IL-6, and IL-8, via inhibition of nuclear factor kappa B (NF-ĸB)To treat leukemias characterized by the presence of the Philadelphia chromosome. Recently, it has been proposed to treat inflammation linked to COVID-19 infectionSignificant decrease of NF-ĸB in chronic myelogenous leukemia patients[88,90]
H1 histamine antagonist
-
Clemastine
Reduces NF-ĸB activity and TLR4 expressionTo treat allergy symptoms [89]
Phosphodiesterase Inhibitor
-
Ibudilast
Inhibit NF-ĸB by preventing nuclear translocationTo treat asthma and stroke [89]
Interleukin-6 inhibitors
-
Sarilumab
-
Tocilizumab
-
Siltuximab
Monocolonal antibodies inhibit IL-6 receptor and IL-6Treatment of inflammatory diseases such as rheumatoid arthritis and COVID-19 infection [95]
Interleukin-1 inhibitors
-
Anakinra
-
Rilonacept
-
Canakinumab
Inhibits IL-1 directly or binds to IL-1 receptor [112]
HIF-1α inhibitors
-
2ME2 NCD (panzem)
-
17-AAG (tanespimycin)
-
Vorinostat (SAHA, Zolinza)
-
PT2385
-
PT2977
-
EZN-2208 (Pegylated SN-38)
-
CRLX101
Inhibits HIF-1α by either inhibiting its production, promoting the degradation, interfering the signaling pathway, or direct binding [31]
Table 3. Drugs currently undergoing clinical trials for the treatment of non-small cell lung cancer.
Table 3. Drugs currently undergoing clinical trials for the treatment of non-small cell lung cancer.
Trial NumberPhaseStatusEstimated Completion DateTreatment
NCT046480331RecruitingDecember 2027Vancomycin + Stereotactic Body Radiation Therapy
NCT049053162RecruitingMay 2024Canakinumab + Durvalumab
+ Radiation therapy
+ Chemotherapy
NCT043823002RecruitingApril 2023Pyrotinib + thalidomide
NCT027797511Active, not recruitingSeptember 2023Abemaciclib + Pembrolizumab + Anastrozole
NCT04184921-Active, not recruitingDecember 2023Aspirin + Osimertinib
NCT004084602CompletedApril 2017Imatinib Mesylate + paclitaxel
NCT057046341Not yet recruitingJanuary 2028Cemiplimab + Sarilumab
NCT046918172Not yet recruitingApril-2026Atezolizumab + Tocilizumab
NCT033376981 and 2RecruitingAugust 2025Atezolizumab + Cobimetinib
+ RO6958688 + Docetaxel
+ CPI-444 + Pemetrexed
+ Carboplatin + Gemcitabine
+ Linagliptin + Tocilizumab
+ Ipatasertib + Bevacizumab
+ Sacituzumab Govitecan
+ Radiation + Evolocumab
NCT026380901 and 2Active, not recruitingJanuary 2024Vorinostat + Pembrolizumab
NCT013807692CompletedFebruary 2014CRLX101
NCT056365921RecruitingDecember 2027Statins + PD-1/PD-L1 inhibitors
NCT054457913RecruitingJuly 2025Metformin hydrochloride
Table 4. List of natural compounds to target inflammation in NSCLC.
Table 4. List of natural compounds to target inflammation in NSCLC.
CompoundMechanism of ActionInitial PurposePerformance RemarksReference
AndrographolideInhibition of NF-ĸBTreatment of upper airway disorders [122,124,125]
AsthaxanthinRegulating the nuclear factor erythroid 2-related factor/heme oxygenase-1 pathway, NF-ĸB signaling, MAPK signaling, JAK-STAT 3 signaling, Pi3-kinase/Akt pathway, and modulating immune responseDietary supplement [145,146,147]
CurcuminInhibition of NF-ĸBDietary supplement [149,150]
Fumaric Acid EstersAlters leukocyte, keratinocyte, and/or endothelial functionsTo treat psoriasis and multiple sclerosis [139]
BaicaleinInhibits metastasis (exact mechanism of action yet to be confirmed) [136]
Kampo medicine, Hochuekkito, TJ-41Inhibited influenza A virus replication by IFN-α upregulationTo treat infectious disease, possesses virological activityIn vivo and in vitro study shows positive results[117]
Cinnamon
(cinnamaldehyde, cinnamic acid, 2-hydroxycinnamaldehyde, 2-methoxycinnamaldehyde, and eugenol)
Suppressed nitric oxide (NO), IL-6, TNF-α, and IL-1β production. Production and blocking of nuclear factor-ĸB (NF-ĸB) and mitogen-activated protein kinase (MAPK)Has immunomodulator, antiseptic and antiviral properties [114]
β-carbolinesInhibits NF-κB/p65 and EMT transitionTo treat altitude sickness and possess anti-inflammatory properties [151]
Magnesium isoglycyrrhizinate (MgIG)Inhibits fibroblast differentiation via the p38MAPK/Nox4/Akt pathwayRespiratory disorders, hyperdipsia, epilepsy, fever [151]
Polydatin (PD)NLRP3 inflammasome and NF-κB pathwayUsed to reduce symptoms of menopause, digestive system [151]
Zingerone (vanillylacetone)Inhibiting NF-κB and MAPKsTo treat infections, nausea, bronchitis, dysentery, heartburn, cough, flatulence, diarrhea, loss of appetite [151]
ZerumboneInhibits TNF-α or LPS-induced production inflammatory cytokines via inhibition of NF-ĸBTo treat fever, sprains, asthma, torment, severe sprains, toothache, allergies, wounds, and stomachache [143]
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

Rajasegaran, T.; How, C.W.; Saud, A.; Ali, A.; Lim, J.C.W. Targeting Inflammation in Non-Small Cell Lung Cancer through Drug Repurposing. Pharmaceuticals 2023, 16, 451. https://doi.org/10.3390/ph16030451

AMA Style

Rajasegaran T, How CW, Saud A, Ali A, Lim JCW. Targeting Inflammation in Non-Small Cell Lung Cancer through Drug Repurposing. Pharmaceuticals. 2023; 16(3):451. https://doi.org/10.3390/ph16030451

Chicago/Turabian Style

Rajasegaran, Thiviyadarshini, Chee Wun How, Anoosha Saud, Azhar Ali, and Jonathan Chee Woei Lim. 2023. "Targeting Inflammation in Non-Small Cell Lung Cancer through Drug Repurposing" Pharmaceuticals 16, no. 3: 451. https://doi.org/10.3390/ph16030451

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