Next Article in Journal
Iron-Gallic Acid Peptide Nanoparticles as a Versatile Platform for Cellular Delivery with Synergistic ROS Enhancement Effect
Next Article in Special Issue
Bio-Enhanced Neoligaments Graft Bearing FE002 Primary Progenitor Tenocytes: Allogeneic Tissue Engineering & Surgical Proofs-of-Concept for Hand Ligament Regenerative Medicine
Previous Article in Journal
Colloidal Silver Nanoparticles Obtained via Radiolysis: Synthesis Optimization and Antibacterial Properties
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Therapies Targeting Immune Cells in Tumor Microenvironment for Non-Small Cell Lung Cancer

1
School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510091, China
2
People’s Hospital of Longhua, Affiliated Longhua People’s Hospital, Southern Medical University, Shenzhen 518109, China
*
Author to whom correspondence should be addressed.
Pharmaceutics 2023, 15(7), 1788; https://doi.org/10.3390/pharmaceutics15071788
Submission received: 27 April 2023 / Revised: 2 June 2023 / Accepted: 15 June 2023 / Published: 21 June 2023
(This article belongs to the Special Issue Where Are We Now and Where Is Cell Therapy Headed?)

Abstract

:
The tumor microenvironment (TME) plays critical roles in immune modulation and tumor malignancies in the process of cancer development. Immune cells constitute a significant component of the TME and influence the migration and metastasis of tumor cells. Recently, a number of therapeutic approaches targeting immune cells have proven promising and have already been used to treat different types of cancer. In particular, PD-1 and PD-L1 inhibitors have been used in the first-line setting in non-small cell lung cancer (NSCLC) with PD-L1 expression ≥1%, as approved by the FDA. In this review, we provide an introduction to the immune cells in the TME and their efficacies, and then we discuss current immunotherapies in NSCLC and scientific research progress in this field.

1. Introduction

Cancer is one of the leading causes of mortality worldwide [1], contributing to a reduction in life expectancy in many nations [2]. The Global Burden of Cancer 2020 (GLOBOCAN 2020) database from the World Health Organization estimates the most recent incidence and mortality rates for 36 varieties of cancer as well as cancer trends in 185 nations. This latest cancer report demonstrated that there were almost 19.3 million new cases of cancer and close to 10 million cancer-related deaths in 2020, and that by 2040 there would be 28.4 million new cancer cases worldwide [3,4]. The most recent edition of the International Agency for Research on Cancer (IARC) World Cancer Report shows that cancer is the primary cause of death among people aged 30 to 69 [5]. Lung cancer is the leading cause of cancer-related deaths and ranks second in terms of cancer incidence worldwide, with an estimated 1.8 million deaths (18%) in 2020 [4]. Non-small cell lung cancer (NSCLC) accounts for 85% of all lung cancer cases and is one of the most fatal tumors worldwide [6]. According to the Chinese National Cancer Center’s statistics for 2016, the number of lung cancer cases was about 787,000 and the number of deaths was about 630,000, making it the primary cause of mortality (21.7%) in China [7]. In recent years, the survival rate of NSCLC patients has increased due to the development of surgery, radiotherapy, and other treatments. However, the 5-year survival rate remains low, with a rate of 15% [8].
The tumor microenvironment (TME) is widely considered to be vital in dynamically modulating tumor growth and affecting therapeutic effectiveness, and numerous therapeutic approaches targeting components of the TME have been designed recently. The TME is defined as the cellular environment. It is composed of various cell types, such as cancer cells, immune cells, stromal cells, and cytokines as well as chemokines [9,10]. Much scientific research indicates that regulating a particular TME component can increase cancer survival rates. Garon et al., revealed that the combination of docetaxel and ramucirumab, which targeted vascular endothelial growth factor receptor-2 (VEGFR-2), could effectively prolong the lifetime of NSCLC patients [11]. Gocheva et al., illustrated that Tenascin-C, one of the components of the extracellular matrix, was much more prevalent in primary and metastatic lung cancers than in normal tissue, which was closely related to lung cancer patient survival [12]. Moreover, a study by Lambrechts analyzed the TME cell profile of lung cancer and discovered that a subpopulation of Tregs from NSCLC was related to a poor prognosis for lung adenocarcinoma [13].
Immune cells have been shown to be crucial in the TME. Tumor cells can develop a variety of immunosuppressive mechanisms, causing cancer cells to evade immune surveillance and undergo immune escape, resulting in tumor proliferation after treatment [14]. Different types of immune cells within the TME are able to activate an immune response through a number of interactions, and lead to a substantial impact on tumor progression and metastasis [15]. In the past few decades, a number of immunotherapies have already been used to treat different types of cancer. Immunotherapy is an essential part of cancer therapy, with better efficacy and fewer side effects than conventional chemotherapies. Rodriguez-Garcia demonstrated that CAR-T cell therapy, which was approved by the FDA for use in clinical treatment for B cell acute lymphoblastic leukemia, is an extremely successful therapy for hematological malignancies [16,17]. There is also increased promise for treatment of tumors with immunotherapies targeting Dendritic Cells (DCs), such as DC vaccines. DCs can induce tumor-directed immune responses in cancer patients by activating cytotoxic T cells [18]. The treatment of advanced NSCLC has undergone a fundamental change due to the advent of immunotherapy in recent years. The use of immune checkpoint inhibitors (ICIs) alone or in combination with chemotherapy has become the primary immunotherapy strategy for advanced or metastatic NSCLC without driver mutations [19]. The FDA has approved the use of ICI therapies such as nivolumab, pembrolizumab and atezolizumab as first-line therapy for NSCLC patients with high PD-L1 expression [20]. Nevertheless, a large proportion of patients still suffer from disease progression.
In this review, we focus on the immune cells in the TME and tumor development. We also discuss the mainstream immunotherapies currently used to treat NSCLC and some novel therapeutic approaches used in the clinic, highlighting therapies that are under clinical trials or that have been approved.

2. Immune Cells in Tumor Microenvironment

Immune cells are a fundamental part of the TME and are essential for both pro- and antitumor immune responses [21]. The cells of the immune system include both innate immune cells, such as macrophages, neutrophils, mast cells, DCs, and MDSC and NK cells, and adaptive T and B lymphatic immune cells [22]. Early studies using immunohistochemistry to identify immune cells in NSCLC found that NSCLC tumors contain multiple types of immune cells, including T cells, B cells, macrophages, NK cells, and DCs, and many studies have demonstrated a crucial link between immune cells and the survival of NSCLC patients [23,24]. It is clear that immune cells are a non-negligible presence in the TME. Most importantly, the immune system is normally capable of monitoring, recognizing, and destroying cancerous cells [25]. However, due to their close proximity to tumor cells, or as a result of chemokine and cytokine communication, the immune cells within the TME are able to evade identification and annihilation by the host immune system [26]. The occurrence of lung cancer and the entire process from early lung cancer to metastatic lung cancer are considered to depend on the immune escape mechanism [27,28]. Therefore, the role that immune cells play in the TME and the mechanisms by which they influence tumor growth deserve to be discussed. In the following part, we introduce the main immune cells in the TME, including T cells, tumor-associated macrophages, dendritic cells, and natural killer cells (Figure 1).

2.1. T Cells

T cells are an essential part of the immune system, which is responsible for defending cells against cancer and viruses [29]. T cells can be further divided into three subpopulations: cytotoxic T lymphocytes (CTLs), T helper cells (Ths), and Tregs [30]. CTLs are the cells in the TME with direct tumor-killing power, Ths cells help to postpone tumor growth, and Treg cells are typically associated with poor outcomes in tumor patients.
CTLs (which include cytotoxic CD8+ T cells and CD4+ T cells) in the TME can directly destroy malignant tumors. As major killers of pathogens and cancers, CD8+ T cells are the preferred immune cells in the fight against cancer [31,32]. The presence of cytotoxic CD8+ T lymphocytes is also linked to better outcomes in NSCLC patients [33]. When CD8+ T cells develop into cytotoxic T cells, naive CD8+ T cells are connected to the peptide major histocompatibility complex (MHC) on antigen-presenting cells (APCs) via their T-cell receptors (TCRs), and are activated by co-stimulatory signals and extracellular cytokines. These CD8+ T cells subsequently become effectors, and their activation enables them to attack cancer cells directly [34]. In addition to actively destroying tumor cells, CD8+ T cells have the capacity to prevent tumor angiogenesis by secreting interferon-γ (IFN-γ) [35]. However, prolonged antigenic stimulation transforms CD8+ T cells in the TME into a T cell-depleted hypoactive state (including progressive loss of effector function and persistent expression of inhibitory receptors), which prevents the tumor cell-killing function of CD8+ T cells from proceeding normally [36,37]. Apart from CD8+ T cells, CD4+ T cells are essential for tumor elimination. CD4+ T cells can directly contribute to CD8+ T cell activation, support CD8+ T cells in the formation of memory CTL, and enhance CD8+ T cell proliferation by generating interleukin(IL)-2 [38,39]. Furthermore, CD4+ T cells can help activate CD8+ T cells by delivering tumor antigens to CD8+ T cells or by triggering the production of cytokines and suppressor molecules [40].
Ths cells are generated by cytokine-specific polarization or TCR-signaling activation in the TME, and they can assist CD4+ T lymphocytes in producing cytotoxicity and destroying tumor cells [41]. Th1 and Th9 cells in the TME boost CD8+ T cells’ ability to fight tumors, mainly by secreting IL-4 and IFN-γ, which are linked to a favorable prognosis in various cancer types [9,42].
Tregs normally inhibit autoimmunity by restricting the immune system’s reaction to autoantigens [43]. However, Tregs in the TME can promote cancer growth by reducing the function of lymphatic and bone marrow immune cells [44]. Many studies have reported that Treg cells in the TME suppress effector T cells directly by generating higher levels of cytokines than those associated with T-cell activation, resulting in immunosuppressive TME and the progression of tumors [45,46,47,48]. On the other hand, Treg cells are capable of promoting tumor growth and metastasis by secreting growth factors, interacting with stromal cells, and undergoing immune escape [43]. A higher Treg-to-effector T-cell ratio in NSCLC tumor tissue has been linked to a worse prognosis [49].

2.2. Tumor-Associated Macrophages (TAMs)

The majority of immune cells that infiltrate the TME are tumor-associated macrophages, which influence tumor angiogenesis and metastasis and are related to a poor prognosis for various cancers [50,51]. In the TME, TAMs may be differentiated into classical activated macrophages (M1) or, alternatively, activated macrophages (M2) [52,53]. Phenotype M1 is able to directly kill tumor cells by producing nitric oxide and reactive oxygen species, or by secreting pro-inflammatory cytokines such as tumor necrosis factor-α (TNF-α), IL-6, and IFN-γ [50,54,55]. Additionally, through the process of antibody-dependent cell-mediated cytotoxicity (ADCC), M1 has the capacity to eradicate malignant cells [56]. In contrast, M2 stimulates angiogenesis, tissue healing, and cancer-cell proliferation by secreting anti-inflammatory cytokines such as IL-4, IL-1, and VEGF [57,58,59]. As macrophages are highly plastic in the TME, they convert between M1 and M2 in response to microenvironmental factors (cytokines, growth factors, chemokines, inflammation, hypoxia, and infection), thereby inhibiting the antitumor response and producing immunosuppressive TME. This is a process known as macrophage polarization [52,59,60]. It has been found that tumor cells express significant amounts of colony-stimulating factor 1, C-C pattern ligand 2 (CCL2), and monocyte chemotactic protein-1, something which contributes to TAMs’ function in tumor development [61]. At the same time, TAMs activate tumor cells by activating transcription factors (STAT3 and NF-κB) to produce VEGF and matrix metalloproteinases (MMPs) to promote tumor angiogenesis [51,62]. As the relationship between TAMs and cancer becomes clearer, TAMs have become a desirable target for the development of novel immunotherapies. It has been demonstrated that activation of TAM receptors reduces resistance to ICI treatment in NSCLC patients, indicating that the combination of the two strategies may be beneficial for these patients [63].

2.3. Dendritic Cells (DCs)

As the most powerful APCs, DCs serve as central regulators of the adaptive immune response and connect innate and adaptive immunity [64,65,66]. DCs are inherently plastic and can be stratified into a variety of subtypes [15]. According to their phenotypes and their functions in the TME, DCs are typically divided into two major subgroups: plasmacytoid DCs (pDCs) and myeloid DCs (mDCs) [67,68].
A primary function of pDCs is to produce IFN-Ⅰ, which is needed to counteract the antiviral immune response [65,69]. It has also been suggested that pDCs may have potential as APCs due to the MHC-Ⅱ and co-stimulatory molecules they express at steady state [70]. In the TME, the function of pDCs is controversial. Tumor-infiltrating pDCs are reported to produce immunosuppressive and carcinogenic effects; however, due to their ability to secrete INF-Ⅰ and TNF-α, pDCs are potential therapeutic targets for combating cancer [71,72].
In the TME, conventional DCs (cDCs), as the typical representatives of mDCs, can be further subdivided into cDC1 and cDC2 spectrums [73]. cDC1 is regulated by transcription factors IRF8, ID2, and BATF3, while cDC2 is regulated by transcription factors IRF4, ID2, ZEB2, and Notch2 [74]. cDC1 is able to cross-present antigens and present exogenous antigens to CD8+ T cells on MHC-I, activating CD8+ T cells and making them the primary mediators of antitumor immunity [75]. Hence, cDC1 has the unique ability to induce intracellular infections and encourage the immune system to reject malignant cells [15,65,76]. The cDC2 exhibits enhanced presentation of the MHC II antigen. In contrast to cDC1, cDC2 presents only exogenous antigens without cross-presentation and activates CD4+ T cells selectively, making it the most effective activation and expansion agent for CD4+ T cells [77,78].
It has been shown that immunogenic DCs secrete substantial quantities of inflammatory cytokines, including TNF-α, IL-12, IL-6, and IL-8, which may improve the clinical outcomes of tumor patients [79,80]. Despite this, the uptake of antigen by DCs in the TME is often insufficient. The phenomenon is caused by aberrant expression of some tumor-derived chemokines on DCs within the TME, resulting in the suppression of DC invasion [67]. The maturation state of DCs and specific markers expressed by different subgroups of DCs have been considered significant indicators of prognosis and prediction in human cancer. DC-based vaccines have been developed to induce and maintain immune responses [78]. Consequently, the use of DC therapy in conjunction with other immunotherapies to reduce immunosuppressive TME can serve as a future treatment strategy for cancer. Zhong et al., found that chemotherapy combined with a DC vaccine significantly inhibited tumor growth in a mouse model of NSCLC [81].

2.4. Natural Killer Cells (NK Cells)

As innate lymphocytes, NK cells can stimulate the immune system to destroy cancer cells [82]. It has been demonstrated that NK cells can kill tumors by direct cytotoxic killing of target cells [83]. Besides, NK cells can inhibit tumor growth by inducing pro-inflammatory cytokines, which include IFN-α, TNF-α, and granulocyte macrophage colony-stimulating factor [84].
NK cells have been divided into two distinct subpopulations based on CD56 expression levels, CD56brightCD16 and CD56dimCD16 [85]. When CD56bright NK cells are exposed to pro-inflammatory cytokines including IL-12, IL-15, and IL-18, they produce additional cytokines to regulate adaptive immunity and cause cytotoxicity [86,87]. CD56dim NK cells can directly mediate the death of infected and cancer cells via cytotoxicity and cytokine production [86,88]. NK cells may also eliminate MHC class I tumors and carry out antigen presentation on their own, creating a connection between innate and adaptive immunity [89].
NK cells are associated with both the killing of tumor cells and the prevention of cancer progression. However, NK cell toxicity is decreased in the TME [9,90]. A change in angiogenesis may be responsible for this, as well as insufficient access to nutrients and oxygen. Many studies have reported that hypoxia and the secretion of transforming growth factor-β (TGF-β) and other cytokines in the TME limit the activity of NK cells [91,92,93]. TGF-β can reduce the antitumor effect of NK cells by transforming peripheral CD4+ T cells activated into Treg cells [92,94]. One study found that delivering TGF-β inhibitors and selenocysteine to breast cancer cells can reduce TGF-β in the TME, improve NK cell activity, and increase antitumor capability, suggesting that NK-targeted immunotherapies may be promising [95].

2.5. Other Types of Immune Cells That Affect Tumor Development

Apart from the immune cells discussed above, neutrophils, MDSCs, and B cells also play crucial roles in the TME.
Neutrophils, the most abundant innate immune cells in the bone marrow and peripheral blood, serve a complex and important function in cancer development [96]. In many cancer patients, neutrophil peripheral blood counts are elevated, and neutrophil-to-lymphocyte ratios are independent prognostic factors [97,98]. In the TME, neutrophils produce MMP-9 to stimulate angiogenesis, boosting cancer growth and offering extra immune-escape pathways for cancer cells [97]. The neutrophil extracellular trap (NET) can promote tumor progression, metastasis, invasion, and angiogenesis [99,100]. Several studies have shown that mice with lung cancer, breast cancer, and chronic myeloid leukemia are more likely to release NET than healthy mice [100,101,102].
MDSCs are a heterogeneous cell population composed of progenitor cells derived from bone marrow, immature macrophages, immature granulocytes, and immature DCs [103]. As part of the TME, they promote tumor angiogenesis and metastasis [104]. In Hinshaw’s study, it was found that MDSC could be stimulated in the TME and released chemokines including NO, CCL3, and CCL4, which inhibited innate and adaptive immune responses [15]. In addition, MDSCs promote epithelial mesenchymal transition by secreting IL-6, which increases angiogenesis and speeds up tumor progression [105,106].
Tumor-infiltrating B cells are responsible for antibody production, antigen presentation, and cytokine secretion in tumors, as well as for complementing T-cell-mediated antitumor immunity [9,107]. Recent studies demonstrated that B cells in the TME have prognostic value in various tumors [108,109].

3. Target Immune Cells against NSCLC

Given the crucial roles of immune cells in immunomodulation and tumor progression in the TME, strategies to target immune cells in the TME hold significant promise. In the past few decades, a number of immunotherapies have already been used to treat different types of cancer. Current research indicates that immunotherapies targeting ICIs when utilized in NSCLC patients have exhibited promising results in terms of five-year survival and progression-free survival (PFS) when compared with previous treatments [110,111].
In the following part of our review, the immunotherapies (Figure 1) used in NSCLC in clinical trials and in scientific research are discussed.

3.1. Target T Cells against NSCLC

T cells are usually recognized as the most essential immune cells in attacking malignancies [112]. Among T-cell-targeted immunotherapies, treatments using ICIs have undergone the most extensive research and been approved by the FDA. This strategy keeps the antitumor immune response in the TME, bringing a new era of cancer immunotherapy [113,114]. An ICI is an immune checkpoint protein that maintains the ability of T cells to destroy tumors by binding to their partner proteins. Immune checkpoint molecules include CTLA-4, PD-1, etc. (Figure 2), and their role is to maintain an immune balance and prevent an immune response from attacking normal tissues and cells in the body. Immunotherapy drugs are mainly monoclonal antibodies, which can block immunosuppressive effect, allowing T cells to recognize and attack cancer cells, thereby enhancing immune responses, by binding with their partner proteins. These drugs (antibodies) are preferentially designed as ICIs [115]. The main agents undergoing clinical trial and research are nivolumab and pembrolizumab (PD-1 antibody), atezolizumab (PD-L1 antibody), and ipilimumab (CTLA-4 antibody) [116].
According to scientific studies, ICIs have a significant impact on patients’ prognoses, and have become the first-line therapy for most individuals with advanced lung cancer [117,118,119]. PD-1 receptor is mainly expressed on activated T cells, and by adhering to ligands PD-L1 and PD-L2, it suppresses the antitumor activity of T cells [120,121]. Expression of PD-L1 is widespread in cancer cells and elevated in the TME, leading to suppression of the antitumor immune response by limiting the signals that activate T cells [112,121]. Nivolumab is an antibody against PD-1. As demonstrated in a clinical trial (NCT01642004), among patients with metastatic NSCLC (mNSCLC) (stage III patients) for whom platinum-based chemotherapy had failed, improved overall survival (OS) and substantially improved PFS were seen in individuals treated with nivolumab [122]. An additional study comparing the efficacy of nivolumab and docetaxel (a chemotherapy drug) in patients with mNSCLC (NCT01673867) also discovered positive results for nivolumab [123]. As a result, nivolumab is the first ICI targeting PD-1 to receive FDA approval for use in the treatment of patients with mNSCLC. Pembrolizumab is also an anti-PD-1 antibody. Patients with mNSCLC who participated in a randomized, phase III trial (NCT02142738) demonstrated significant improvements in 6-month PFS, OS, and objective response rate (ORR) when compared with platinum-based chemotherapy [124]. Furthermore, a phase III trial (NCT03850444) comparing pembrolizumab to platinum-based chemotherapy in patients with advanced or mNSCLC found that pembrolizumab had a meaningful effect in individuals whose PD-L1 expression was higher than 50% [125]. The FDA has approved pembrolizumab as a first-line treatment for patients with PD-L1 levels ≥ 1%, and these results have led to the authorization of pembrolizumab by the European Medicines Agency (EMA) as a first-line treatment for patients with PD-L1 levels ≥ 50% [20]. Atezolizumab, a PD-L1 antibody, has also been approved by the FDA and the EMA as a single-agent treatment for patients with mNSCLC with high PD-L1 expression [20,126], having shown significant benefits in treating patients with NSCLC in clinical trial NCT02409342 (Table 1).
Moreover, ICIs includes antibodies against CTLA-4, which inhibits T-cell proliferation by competing binding with CD28 on the cell surface [127]. Since the antitumor effect of CTLA-4 antibodies is mainly related to T-cell initiation in lymph nodes and the TME, this may offset some ICI resistance [120,128,129]. Ipilimumab is a therapeutic antibody targeting CTLA-4 [130]. A trial (NCT02477826) has been carried out on patients with stage IV NSCLC to compare the efficacies of three different strategies: nivolumab alone, the combination of nivolumab and ipilimumab, and the combination of chemotherapy with nivolumab plus platinum [131]. Recent findings from this trial indicated that the combination of nivolumab and ipilimumab enhanced OS for those patients with PD-L1 expression levels of more than 1%, suggesting that the ICI two-drug combination could be considered as a first-line therapy for NSCLC. Meanwhile, the results of this trial also implied a positive outcome from the combination of chemotherapy and anti-PD-1 drugs [132]. In addition, in a clinical trial of patients with metastatic squamous NSCLC treated with carboplatin-paclitaxel/nab-paclitaxel with or without pembrolizumab (NCT03875092), it was found that PFS and OS improved significantly when anti-PD-1 medications were taken in conjunction with chemotherapy [133] (Table 1). Combination therapy with ICIs has been shown to have great potential in terms of antitumor response. Apart from chemotherapy and ICIs, many studies indicate that inhibitors targeting IDO, VEGF, lymphocyte activation3 and T-cell immunoglobulin mucin 3 may also be considered as future combination options [134,135,136,137].
In addition to the immunotherapy of ICIs against NSCLC, chimeric antigen receptor-modified T cells (CAR-T cells, a type of genetically engineered T cells) treatment is another very rapidly developing form of cellular immunotherapy [138]. Synthetic CAR vectors are genetically engineered to recognize and bind specifically to tumor cell surface antigens (such as CD19) to achieve antitumor effects in this therapy [139,140]. However, the presence of tumor-associated antigens (TAAs) in NSCLC has been heterogeneous in terms of both intensity and distribution, severely limiting the clinical efficacy of CAR-T therapy [141].
Recently, understanding of the molecular typing of NSCLC has been significantly enhanced, because of research into the causative genes of NSCLC patients [142,143]. It is clear that the molecular typing of NSCLC can be classified into three groups. The first category refers to genes with clinical relevance, such as KRAS, HER2, ROS1, TP53, BRAF, and NTRK, and relevant medications targeting these genes are currently being researched [142,144]. Several studies have demonstrated that patients with mutant NSCLC, such as KRAS, BRAF, and TP53, can benefit from ICIs [145,146,147]. The second category consists of treatable mutation-driver genes. The current focus of therapies based on mutation-driver genes in NSCLC are epidermal growth factor receptor (EGFR) and anaplastic lymphoma kinase (ALK) [142]. Because of the mutations in EGRF or ALK, the efficacy of ICI treatment in these NSCLC patients is not obvious [19,148]. Many targeted medicines that target the driver genes of EGFR and ALK have had some success [143]. The third category includes biomarkers that have some association efficacy, such as PD-L1 and TMB [149,150]. In NSCLC patients with mutations in EGFR, the PD-L1 expression is predominantly reduced. Consequently, anti-PD-1 ICIs are ineffective in this group of patients [151]. However, EGFR-tyrosine kinase inhibitors (EGFR-TKIs) have been developed to treat NSCLC patients. First-generation EGFR-TKIs, such as gefitinib, erlotinib, and ecotinib, have been shown to be effective in treating patients with EGFR-sensitive mutant NSCLC. Afatinib is a second-generation EGFR-TKI that was approved by the FDA and the EMA in 2013 used to treat adults with advanced, EGFR mutation-positive NSCLC [152]. However, long-term use of EGFR-TKI would result in drug resistance and the loss of the original therapeutic effect [142]. Osimertinib, a third-generation EGFR-TKI, was developed to treat NSCLC patients who acquired the EGFR T790M resistance mutation [153]. Due to the drug resistance of EGFR-TKIs, future strategies must be developed to cope with resistance to next generation EGFR-TKIs, including conventional combinations of EGFR-TKIs and other agents and treatments combined with ICIs.
Although ICIs targeting PD-1 and CTLA4 have been shown to be beneficial in mNSCLC, the efficiency in NSCLC is, however, only 8–30% [154]. Emerging evidence indicates that driver oncogenes have different effects on the TME that influence the potential benefit from treatment with ICIs. For instance, patients harboring BRAF, KRAS or TP53 co-mutations benefit from ICIs [155,156]. However, certain patient populations do not respond to these drugs targeting ICIs [112]. Further strategies to enlarge the efficacy of ICIs are to synergistically combine them with CAR-T, EGFR-TKIs and other agents.
Table 1. Relevant immunotherapies for the treatment of NSCLC.
Table 1. Relevant immunotherapies for the treatment of NSCLC.
ClinicalTrials.gov IdentifierTargetDrugStudy DesignCancer TypesPhaseStatusEnrollmentOutcomesReferences
NCT01642004T cells, PD-1NivolumabNivolumab vs. Docetaxel Advanced or metastatic squamous cell NSCLCCompleted352OS↑[122]
NCT01673867T cells, PD-1NivolumabNivolumab vs. DocetaxelMetastatic non-squamous NSCLCCompleted792OS↑[123]
NCT02142738T cells, PD-1PembrolizumabPembrolizumab vs. Paclitaxel + Carboplatin vs. Pemetrexed + Carboplatin vs. Pemetrexed + Cisplatin vs. Gemcitabine + Carboplatin vs. Gemcitabine + CisplatinMetastatic NSCLCCompleted305PFS↑[124]
NCT03850444T cells, PD-1PembrolizumabPembrolizumab vs. Carboplatin + Paclitaxel + PemetrexedPD-L1 positive advanced or metastatic NSCLCActive, not recruiting262Undergoing[125]
NCT02409342Tumor cells, PD-L1Atezolizumab(Carboplatin/Cisplatin) + (Pemetrexed/Gemcitabine) vs. AtezolizumabStage IV non-squamous or squamous NSCLCCompleted572OS↑, PFS↑[126]
NCT02477826T cells, CTLA-4IpilimumabNivolumab vs. Nivolumab + IpilimumabStage IV or recurrent NSCLCActive, not recruiting2748Undergoing[131]
NCT03875092T cells, PD-1PembrolizumabPembrolizumab + Chemotherapy vs. ChemotherapyMetastatic squamous NSCLCActive, not recruiting125Undergoing[133]
NCT05467748TAMs, EZH2TazemetostatTazemetostat + PembrolizumabAdvanced NSCLCIb/IINot yet recruiting66Undergoing[157]
NCT05094804TAMs, CD163OR2805OR2805 vs. OR2805 + PD-1 inhibitorAdvanced malignanciesI/IIRecruiting130Undergoing[158]
NCT04762199TAMs, MerTKMRX-2843Osimertinib + MRX-2843Advanced EGFR mutant NSCLCRecruiting69Undergoing[159]
NCT00103116DCAutologous dendritic cell cancer vaccineAutologous dendritic cell cancer vaccineStage I, Stage II, or Stage III NSCLCIICompleted32Safety, no related adverse events[160]
NCT02808416DCPersonalized cellular vaccinePersonalized cellular vaccinePatients with brain metastases from solid tumorsCompleted10Induced specific CD4+ and CD8+ T cell responses [161]
NCT02843204NK cellsPembrolizumab and NK immunotherapyPembrolizumab + NK immunotherapy vs. PembrolizumabMalignant solid tumorI/IICompleted110OS↑, PFS↑[162]
NCT02843815NK cellsNK immunotherapy and cryosurgeryCryosurgery + NK immunotherapy vs. CryosurgeryAdvanced NSCLCI/IICompleted30Relief degree↑[163]
NCT02118415NK cellsHsp70-peptide TKD/IL-2 activated, autologous NK cells(Hsp70-peptide TKD/IL-2 activated, autologous NK cells) vs. Control groupNSCLC Stage IIIA/BSuspended90Undergoing[164]
↑ means an improvement in the outcomes.

3.2. Target TAMs against NSCLC

Crosstalk between TAMs and other cells in the TME results in an immunosuppression that promotes tumor cell proliferation, angiogenesis, tumor metastasis, and the development of cancer-therapy resistance [165]. It is widely known that macrophage polarization may cause a direct switch between M1 and M2 in the TME, while subtype M1 could kill tumor cells and M2 can promote tumor progression [52]. Small molecule inhibitors or monoclonal antibodies against TAM signaling can consequently inhibit the progression and metastasis of cancer [129]. Recently, the main immunotherapies targeting TAMs and undergoing clinical or scientific trials are tazemetostat, OR2805, and MRX-2843.
Tazemetostat, an enhancer of zeste homolog 2 (EZH2) inhibitor, has been used individually or in combination with ICIs against advanced NSCLC in clinical trials. It has been demonstrated that EZH2 could stimulate the expression of CCL5 and result in the recruitment of macrophages and the invasion of lung cancer [166]. When EZH2 is inhibited, the quantity of M2 decreases, thereby preventing the progression of lung cancer [166]. An open-label, single-arm, phase Ib/II clinical trial (NCT05467748) is underway, in which pembrolizumab in combination with tazemetostat is used to treat patients with advanced NSCLC who have relapsed following initial or second-line therapy (Table 1) [157].
OR2805 is a monoclonal antibody against CD163. CD163 is one of the biomarkers of M2 macrophage polarization, which can inhibit PD-1 and PD-L1 signaling and stimulate macrophage polarization to the M1 phenotype, resulting in enhanced antitumor activity [167,168]. Therefore, OR2805 can be used as a monotherapy or in combination with PD-1 inhibitors to treat patients with advanced solid tumors, and is a valuable targeted TAMs therapy. An open-label, multicenter, first-in-human dose escalation and extension phase I, II trial (NCT05094804) to verify the safety, tolerability, pharmacokinetics, pharmacodynamics, and preliminary anticancer activity of OR2805 is at the recruitment stage, with individuals with NSCLC being expected to enroll (Table 1) [158].
MRX-2843 is a mer tyrosine kinase (MerTK) inhibitor. MerTK is a macrophage-specific tyrosine kinase receptor (RTK) that can induce tumor immunological tolerance by inducing macrophage death, polarizing macrophages towards the M2 phenotype, and inhibiting the secretion of inflammatory factors [169,170]. In contrast, blocking MerTK signaling repolarizes macrophages to M1 phenotypes and enhances antitumor immunity [171]. MerTK antibodies stimulate T-cell activation and are synergistic with anti-PD-1 and anti-PD-L1 treatments [172]. Blocking the macrophage MerTK protein may increase antitumor immunity and induce tumor immunogenicity, a promising method for treating cancer. An ongoing phase Ⅰb safety and pharmacodynamic study is being conducted with MRX-2843 in conjunction with osimertinib in patients with advanced EGFR-mutated NSCLC (NCT04762199) (Table 1) [159]. Melittin (MEL-dKLA) is a promising option for NSCLC patients because it selectively induces cell death in M2 macrophages in vitro without interacting with the normal function of other cell types [173,174]. In addition, a preclinical study showed that the construction of targeted nano-micelles (called ‘nano-dandelion’) for simultaneous delivery of curcumin (Cur) and baicalin (Bai) was effective in overcoming tumor resistance, which resulted in the transforming of TAMs to tumor-killing M1-type macrophages, indicating an adjuvant therapy for tumors [175].
Due to the high level of macrophage infiltration, the prognosis for NSCLC patients is poor. Macrophage polarization promotes the cancer development in the TME, thus targeting TAMs has emerged as a promising therapeutic strategy and has led to the development of numerous relevant drugs undergoing clinical trials.

3.3. Target DCs against NSCLC

As the primary APCs, DCs trigger antigen-specific CD8+ T lymphocyte activation, which initiates innate and adaptive immune responses that are capable of killing tumors [176,177]. Usually, DC-targeted treatments include autologous DC vaccination and individualized cellular vaccination. In the majority of studies, patients are vaccinated with DCs via numerous subcutaneous injections of primed DCs [177]. Moreover, combination therapy with DC and cytokine-induced killer cells (DC-CIK) has been confirmed to be an effective treatment for NSCLC patients.
In a clinical trial (NCT00103116), an autologous DC vaccine was involved in the treatment of individuals with stage I, II, and III NSCLC. Specific T-cell activity in the blood was compared before and six months after vaccination. As a result of the trial, there are 22 out of 32 patients who demonstrated an immune response to the vaccine six months after vaccination, and 20 patients out of 32 who were still alive five years after dual vaccination. These pieces of evidence suggest that the DC vaccine has a favorable safety profile [160]. Moreover, one study was conducted in ten patients with tumor brain metastases (BM) (NCT02808416), five of whom were NSCLC patients. The trial was to examine the security and effectiveness of an individualized cellular tumor vaccination (DC vaccine) in patients with BM [161]. These patients had a good OS rate and an excellent objective response; however, due to the limited number of samples, additional clinical trials are required in the future [178]. Details of the above two tests are shown in Table 1. Based on the outcomes of the aforementioned trials, DC vaccines have demonstrated significant potential for treating NSCLC patients.
In mouse models, DC vaccination was found to be effective when used in combination with chemotherapy and radiotherapy for the treatment of NSCLC [81,179]. Studies have compared the combined effects of chemotherapy or radiotherapy, DC vaccination, and cytokine-induced killer cells (CIK) in humans [180,181,182,183]. Consequently, a combination therapy consisting of DC-CIK may have a stronger antitumor effect. Clinical trials have shown that DC-CIK immunotherapy is effective in controlling the disease, improving immune function, and delaying the progression of advanced NSCLC without increasing adverse effects, indicating a promising future in the treatment of NSCLC patients [184].
DC-related immunotherapy for NSCLC patients focuses on DC vaccines and DC-CIK therapy. The safety of these treatments has been recognized, but their exact efficacy still needs to be further determined.

3.4. Target NK Cells against NSCLC

NK cells can destroy tumor cells directly or indirectly through the release of pro-inflammatory factors [83,185]. Many researchers have discovered that CD56bright CD16 NK cells that release high levels of cytokines (such as VEGF, placental growth factor, and IL-8/CXCL8) which promote tumor neoangiogenesis exhibit low cytotoxicity in the lung TME [89,185,186]. In addition, recent research has demonstrated that NK cells can identify and eliminate cancer progenitor cells in solid tumors, and have a beneficial effect on lung cancer [187,188].
Immunotherapies targeting NK cells are mostly utilized as an additional therapy with other treatments and are seldom used alone in clinical studies. It has been revealed that the mixture of NK and T lymphocytes cells (NKTm) improves OS and 2-year survival among patients with NSCLC [189]. In addition, NK immunotherapy in combination with cryosurgery or the PD-1 representative drug Pembrolizumab is currently under research. Hsp70-peptide TKD/IL-2 activated autologous NK cells are being considered as a new treatment for NSCLC patients after demonstrating their potential to treat cancer in preclinical models.
In a clinical trial (NCT02843204), NK immunotherapy combined with ICI pembrolizumab was used to treat patients with advanced relapsed NSCLC. Results from the study indicated that pembrolizumab combined with NK cell therapy improved OS and PFS in patients with previously treated advanced PD-L1 NSCLC without causing any serious adverse effects compared to pembrolizumab alone, showing significant potential for NK cell immunotherapy (Table 1) [162,190]. In a separate trial (NCT02843815), the safety and efficacy of cryosurgery combined with allogeneic NK cell immunotherapy for advanced NSCLC were initially determined (Table 1) [163,191]. Preclinical models demonstrated that the combination of NK cells and anti-PD-1 therapy provides long-term tumor control with a substantial infiltration of CD8+ T cells and NK cells [192]. A combination-therapy study using anti-PD-1 antibodies and autologous NK cells has made significant progress in view of the enormous potential NK cells have demonstrated in tumor therapy.
Currently, a trial (NCT02118415) is taking place to test the efficacy of sequential immunotherapy using autologous NK cells and Hsp70-peptide TKD/IL-2 in patients with NSCLC (stage IIIa, b) after radiotherapy and chemotherapy (Table 1) [164]. It has been shown in a preclinical model of lung cancer that autologous NK cells-based therapies are highly effective against tumors selectively expressing membrane-type Hsp70 [193]. When combined with other ICIs or modulators, autologous NK cell therapy or other therapies that target tumors expressing mHsp70 may be effective in inhibiting tumor growth and progression [194]. In current NSCLC research, relay transfer of NK cells is being evaluated as a potential treatment method.
Furthermore, CAR-NK therapy has emerged as one of the most promising new research topics targeting NK cells for the treatment of NSCLC patients over the past few years [195]. CAR-NK cells have a number of benefits over CAR-T cells due to the fact that they can be produced from pre-existing cell lines or allogeneic NK cells with mismatched MHC. Moreover, they can kill cancer cells through CAR-dependent and CAR-independent pathways with less toxicity [196]. However, CAR-NK cells still have some limitations, such as the short half-life of NK cells and the extra-tumor toxicity [196,197]. Optimizing the CAR-NK structure and prolonging the survival of CAR-NK cells in vivo are the main research goals for the future.
The efficacy of NK cells immunotherapies in combination with other therapies has demonstrated promising results for NSCLC patients. The benefits of recently developed CAR-NK immunotherapies have also been recognized. However, immunotherapies associated with NK cells are still immature, and further research is needed to increase the treatment options for NSCLC patients.

4. Conclusions and Future Directions

The TME is engaged in the whole process of tumor growth and serves a crucial role. Different immune cells in the TME are required to mediate pro- and antitumor immune responses, which affect tumor destiny and the capacity for tumors to become aggressive and grow [21,198]. Due to the significance of immune cells in tumor progression, several immunotherapies have been designed for use in different types of cancer. Over the decades, there have been breakthroughs in immunotherapy, especially ICI immunotherapy applied to NSCLC patients, suggesting immunotherapy’s enormous potential for cancer treatment.
Anti-PD-1/PD-L1 immunotherapies are some of the most common ICIs. The FDA has authorized the representative PD-1- or PD-L1-blocking antibodies nivolumab, pembrolizumab, and atezolizumab for the treatment of NSCLC patients, and pembrolizumab has been used as a first-line treatment for patients with PD-L1 levels ≥ 1%. Recent clinical trial studies targeting PD-L1 include avelumab and durvalumab, and the most recent follow-up results for avelumab and the positive results shown with durvalumab in NSCLC patients after radiotherapy provide a potential treatment option [116]. In addition, it has been demonstrated that the anti-PD-1 drug cemiplimab substantially improved clinical outcomes in advanced NSCLC patients with more than 50% PD-L1 expression, indicating that this drug may be beneficial for NSCLC patients [116]. ICIs also contain antibodies against CTLA-4, such as ipilimumab. In the treatment of NSCLC, two-drug combinations of ICIs (e.g., nivolumab and ipilimumab) have the potential to be first-line therapies. Combination regimens of multiple ICIs offer additional options for future NSCLC patients. In addition, CAR-T genetically engineered T cell-related immunotherapy is being studied to improve the adverse effects seen in patients during the current trials as well as to make the treatment more effective for NSCLC patients in the future. For patients with EGRF-mutant NSCLC for whom ICIs therapy is ineffective, some EGRF-TKI drugs have been developed which have improved the survival rate among these patients. However, the long-term use of these drugs may result in a resistance; future research may focus on developing new drugs to reduce such resistance. TAMs-related immunotherapies are often used in combination with other immunotherapies to treat patients with NSCLC. ICIs with tazemetostat (an EZH2 inhibitor which targets TAMs) is an option for patients with NSCLC whose disease has progressed following first- or second-line therapy. Other TAMs-targeting cancer immunotherapies, such as MRX-2843, OR2805, and related nanotherapies, are currently undergoing clinical testing as promising NSCLC treatments.
NSCLC patients with DC-related immunotherapies are mainly vaccinated with autologous DCs, and studies have shown that they are both safe and effective, though their exact efficacy remains unclear.
Targeted NK cells therapies are mostly used in combination with other therapies for patients with NSCLC. The efficacy of NKTm cell transplantation, as well as cryosurgery combined with allogeneic NK immunotherapy, has been demonstrated. Autologous NK cells treatment or other medicines targeting mHsp70-expressing tumors, in conjunction with ICIs, may be beneficial in preventing the progression of NSCLC. Genetically engineered CAR-NK immunotherapies are being developed to improve NSCLC treatment in the future.
However, immunotherapy still has some drawbacks. ICIs boost T-cell activation in NSCLC patients by blocking negative regulators of T-cell function, which leads to an uncontrolled immune response and results in immune-related adverse events, including colitis, hypophysis, and pneumonitis [199]. Furthermore, checkpoint inhibitor pneumonia seems to be an uncommon but harmful side effect of anti-PD-1 and PD-L1 ICIs [193,200]. Thus, more scientific research and clinical studies are needed to improve treatment plans, reduce side effects, and control the immune response.
Moreover, sitravatinib (a TAM-targeting MerTK inhibitor) has exhibited an antitumor effect and modification of the TME in preclinical trials [201]. A retrospective analysis of NSCLC patients with tumor progression after ICI treatment showed that sitravatinib does not have significant antitumor activity when used singly. On the basis of its immunomodulatory effect on the TME, studies of sitravatinib in combination with ICI will be one of the primary areas of future research.
In summary, immune cells within the TME not only affect tumor growth and metastasis but are also used for therapy against tumors. For patients with advanced NSCLC, a number of immunotherapeutic strategies have been suggested as first-line therapies, and preclinical and clinical studies have proven their effectiveness. The combination of multiple medicines and the development of new therapeutic targets for NSCLC should be investigated in greater depth in the future, along with existing immunotherapy strategies, in order to increase the survival rate of cancer patients.

Author Contributions

W.Y. provided the idea and wrote the manuscript. M.L. searched the literature. K.L. illustrated the figures and revised the manuscript. All authors contributed to the article and approved the submitted version. All authors have read and agreed to the published version of the manuscript.

Funding

This work was supported by the National Natural Science Foundation of China [82104457] and the Shenzhen Longhua District Science and Technology Innovation Fund [SZLHQJCYJ2021001].

Institutional Review Board Statement

Exclude this statement.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Conflicts of Interest

All authors declare that there is no conflict of interest.

References

  1. Bray, F.; Laversanne, M.; Weiderpass, E.; Soerjomataram, I. The ever-increasing importance of cancer as a leading cause of premature death worldwide. Cancer 2021, 127, 3029–3030. [Google Scholar] [CrossRef] [PubMed]
  2. Brustugun, O.T.; Møller, B.; Helland, Å. Years of life lost as a measure of cancer burden on a national level. Br. J. Cancer 2014, 111, 1014–1020. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  3. Deo, S.V.S.; Sharma, J.; Kumar, S. GLOBOCAN 2020 Report on Global Cancer Burden: Challenges and Opportunities for Surgical Oncologists. Ann. Surg. Oncol. 2022, 29, 6497–6500. [Google Scholar] [CrossRef] [PubMed]
  4. 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 Cancer J. Clin. 2021, 71, 209–249. [Google Scholar] [CrossRef]
  5. Wild, C.P.; Weiderpass, E.; Stewart, B.W. World Cancer Report: Cancer Research for Cancer Prevention; IARC: Lyon, France, 2020. [Google Scholar]
  6. Siegel, R.L.; Miller, K.D.; Jemal, A. Cancer statistics, 2020. CA Cancer J. Clin. 2020, 70, 7–30. [Google Scholar] [CrossRef] [PubMed]
  7. Chen, W.; Zheng, R.; Baade, P.D.; Zhang, S.; Zeng, H.; Bray, F.; Jemal, A.; Yu, X.Q.; He, J. Cancer statistics in China, 2015. CA Cancer J. Clin. 2016, 66, 115–132. [Google Scholar] [CrossRef] [Green Version]
  8. Bray, F.; Ferlay, J.; Soerjomataram, I.; Siegel, R.L.; Torre, L.A.; Jemal, A. Global cancer statistics 2018: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J. Clin. 2018, 68, 394–424. [Google Scholar] [CrossRef] [Green Version]
  9. Anderson, N.M.; Simon, M.C. Tumor Microenvironment. Curr. Biol. 2020, 30, R921–R925. [Google Scholar] [CrossRef]
  10. Arneth, B. Tumor Microenvironment. Medicina 2019, 56, 15. [Google Scholar] [CrossRef] [Green Version]
  11. Garon, E.B.; Ciuleanu, T.-E.; Arrieta, O.; Prabhash, K.; Syrigos, K.N.; Goksel, T.; Park, K.; Gorbunova, V.; Kowalyszyn, R.D.; Pikiel, J.; et al. Ramucirumab plus docetaxel versus placebo plus docetaxel for second-line treatment of stage IV non-small-cell lung cancer after disease progression on platinum-based therapy (REVEL): A multicentre, double-blind, randomised phase 3 trial. Lancet 2014, 384, 665–673. [Google Scholar] [CrossRef]
  12. Gocheva, V.; Naba, A.; Bhutkar, A.; Guardia, T.; Miller, K.M.; Li, C.M.-C.; Dayton, T.L.; Sanchez-Rivera, F.J.; Kim-Kiselak, C.; Jailkhani, N.; et al. Quantitative proteomics identify Tenascin-C as a promoter of lung cancer progression and contributor to a signature prognostic of patient survival. Proc. Natl. Acad. Sci. USA 2017, 114, E5625–E5634. [Google Scholar] [CrossRef] [Green Version]
  13. Lambrechts, D.; Wauters, E.; Boeckx, B.; Aibar, S.; Nittner, D.; Burton, O.; Bassez, A.; Decaluwé, H.; Pircher, A.; Van den Eynde, K.; et al. Phenotype molding of stromal cells in the lung tumor microenvironment. Nat. Med. 2018, 24, 1277–1289. [Google Scholar] [CrossRef]
  14. Jiang, X.; Wang, J.; Deng, X.; Xiong, F.; Ge, J.; Xiang, B.; Wu, X.; Ma, J.; Zhou, M.; Li, X.; et al. Role of the tumor microenvironment in PD-L1/PD-1-mediated tumor immune escape. Mol. Cancer 2019, 18, 10. [Google Scholar] [CrossRef] [Green Version]
  15. Hinshaw, D.C.; Shevde, L.A. The Tumor Microenvironment Innately Modulates Cancer Progression. Cancer Res. 2019, 79, 4557–4566. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  16. Rodriguez-Garcia, A.; Palazon, A.; Noguera-Ortega, E.; Powell, D.J.; Guedan, S. CAR-T Cells Hit the Tumor Microenvironment: Strategies to Overcome Tumor Escape. Front. Immunol. 2020, 11, 1109. [Google Scholar] [CrossRef] [PubMed]
  17. Ma, S.; Li, X.; Wang, X.; Cheng, L.; Li, Z.; Zhang, C.; Ye, Z.; Qian, Q. Current Progress in CAR-T Cell Therapy for Solid Tumors. Int. J. Biol. Sci. 2019, 15, 2548–2560. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  18. Wculek, S.K.; Cueto, F.J.; Mujal, A.M.; Melero, I.; Krummel, M.F.; Sancho, D. Dendritic cells in cancer immunology and immunotherapy. Nat. Rev. Immunol. 2020, 20, 7–24. [Google Scholar] [CrossRef]
  19. Doroshow, D.B.; Sanmamed, M.F.; Hastings, K.; Politi, K.; Rimm, D.L.; Chen, L.; Melero, I.; Schalper, K.A.; Herbst, R.S. Immunotherapy in Non-Small Cell Lung Cancer: Facts and Hopes. Clin. Cancer Res. 2019, 25, 4592–4602. [Google Scholar] [CrossRef] [Green Version]
  20. Reck, M.; Remon, J.; Hellmann, M.D. First-Line Immunotherapy for Non-Small-Cell Lung Cancer. J. Clin. Oncol. 2022, 40, 586–597. [Google Scholar] [CrossRef]
  21. Pansy, K.; Uhl, B.; Krstic, J.; Szmyra, M.; Fechter, K.; Santiso, A.; Thüminger, L.; Greinix, H.; Kargl, J.; Prochazka, K.; et al. Immune Regulatory Processes of the Tumor Microenvironment under Malignant Conditions. Int. J. Mol. Sci. 2021, 22, 13311. [Google Scholar] [CrossRef]
  22. Wu, T.; Dai, Y. Tumor microenvironment and therapeutic response. Cancer Lett. 2017, 387, 61–68. [Google Scholar] [CrossRef] [PubMed]
  23. Donnem, T.; Kilvaer, T.K.; Andersen, S.; Richardsen, E.; Paulsen, E.E.; Hald, S.M.; Al-Saad, S.; Brustugun, O.T.; Helland, A.; Lund-Iversen, M.; et al. Strategies for clinical implementation of TNM-Immunoscore in resected nonsmall-cell lung cancer. Ann. Oncol. 2016, 27, 225–232. [Google Scholar] [CrossRef] [PubMed]
  24. Banat, G.-A.; Tretyn, A.; Pullamsetti, S.S.; Wilhelm, J.; Weigert, A.; Olesch, C.; Ebel, K.; Stiewe, T.; Grimminger, F.; Seeger, W.; et al. Immune and Inflammatory Cell Composition of Human Lung Cancer Stroma. PLoS ONE 2015, 10, e0139073. [Google Scholar] [CrossRef] [PubMed]
  25. Saab, S.; Zalzale, H.; Rahal, Z.; Khalifeh, Y.; Sinjab, A.; Kadara, H. Insights Into Lung Cancer Immune-Based Biology, Prevention, and Treatment. Front. Immunol. 2020, 11, 159. [Google Scholar] [CrossRef] [Green Version]
  26. Hanahan, D.; Weinberg, R.A. Hallmarks of cancer: The next generation. Cell 2011, 144, 646–674. [Google Scholar] [CrossRef] [Green Version]
  27. Tian, Y.; Zhai, X.; Yan, W.; Zhu, H.; Yu, J. Clinical outcomes of immune checkpoint blockades and the underlying immune escape mechanisms in squamous and adenocarcinoma NSCLC. Cancer Med. 2020, 10, 3–14. [Google Scholar] [CrossRef]
  28. Qu, J.; Jiang, M.; Wang, L.; Zhao, D.; Qin, K.; Wang, Y.; Tao, J.; Zhang, X. Mechanism and potential predictive biomarkers of immune checkpoint inhibitors in NSCLC. Biomed. Pharmacother. 2020, 127, 109996. [Google Scholar] [CrossRef]
  29. Khedri, M.; Samei, A.; Fasihi-Ramandi, M.; Taheri, R.A. The immunopathobiology of T cells in stress condition: A review. Cell Stress Chaperones 2020, 25, 743–752. [Google Scholar] [CrossRef]
  30. Kumar, B.V.; Connors, T.J.; Farber, D.L. Human T Cell Development, Localization, and Function throughout Life. Immunity 2018, 48, 202–213. [Google Scholar] [CrossRef] [Green Version]
  31. Farhood, B.; Najafi, M.; Mortezaee, K. CD8+ cytotoxic T lymphocytes in cancer immunotherapy: A review. J. Cell. Physiol. 2019, 234, 8509–8521. [Google Scholar] [CrossRef]
  32. Raskov, H.; Orhan, A.; Christensen, J.P.; Gögenur, I. Cytotoxic CD8+ T cells in cancer and cancer immunotherapy. Br. J. Cancer 2021, 124, 359–367. [Google Scholar] [CrossRef] [PubMed]
  33. Gueguen, P.; Metoikidou, C.; Dupic, T.; Lawand, M.; Goudot, C.; Baulande, S.; Lameiras, S.; Lantz, O.; Girard, N.; Seguin-Givelet, A.; et al. Contribution of resident and circulating precursors to tumor-infiltrating CD8+ T cell populations in lung cancer. Sci. Immunol. 2021, 6, eabd5778. [Google Scholar] [CrossRef] [PubMed]
  34. Iwahori, K. Cytotoxic CD8+ Lymphocytes in the Tumor Microenvironment. In Tumor Microenvironment: Hematopoietic Cells—Part A; Birbrair, A., Ed.; Advances in Experimental Medicine and Biology; Springer International Publishing: Cham, Switzerland, 2020; pp. 53–62. ISBN 978-3-030-35723-8. [Google Scholar]
  35. Paul, M.S.; Ohashi, P.S. The Roles of CD8+ T Cell Subsets in Antitumor Immunity. Trends Cell Biol. 2020, 30, 695–704. [Google Scholar] [CrossRef] [PubMed]
  36. Wherry, E.J. T cell exhaustion. Nat. Immunol. 2011, 12, 492–499. [Google Scholar] [CrossRef]
  37. Jiang, W.; He, Y.; He, W.; Wu, G.; Zhou, X.; Sheng, Q.; Zhong, W.; Lu, Y.; Ding, Y.; Lu, Q.; et al. Exhausted CD8+ T Cells in the Tumor Immune Microenvironment: New Pathways to Therapy. Front. Immunol. 2021, 11, 622509. [Google Scholar] [CrossRef]
  38. Lei, X.; Lei, Y.; Li, J.-K.; Du, W.-X.; Li, R.-G.; Yang, J.; Li, J.; Li, F.; Tan, H.-B. Immune cells within the tumor microenvironment: Biological functions and roles in cancer immunotherapy. Cancer Lett. 2020, 470, 126–133. [Google Scholar] [CrossRef]
  39. Borst, J.; Ahrends, T.; Bąbała, N.; Melief, C.J.M.; Kastenmüller, W. CD4+ T cell help in cancer immunology and immunotherapy. Nat. Rev. Immunol. 2018, 18, 635–647. [Google Scholar] [CrossRef]
  40. Ahrends, T.; Borst, J. The opposing roles of CD4+ T cells in anti-tumour immunity. Immunology 2018, 154, 582–592. [Google Scholar] [CrossRef] [Green Version]
  41. Basu, A.; Ramamoorthi, G.; Albert, G.; Gallen, C.; Beyer, A.; Snyder, C.; Koski, G.; Disis, M.L.; Czerniecki, B.J.; Kodumudi, K. Differentiation and Regulation of TH Cells: A Balancing Act for Cancer Immunotherapy. Front. Immunol. 2021, 12, 669474. [Google Scholar] [CrossRef]
  42. Maier, E.; Duschl, A.; Horejs-Hoeck, J. STAT6-dependent and -independent mechanisms in Th2 polarization. Eur. J. Immunol. 2012, 42, 2827–2833. [Google Scholar] [CrossRef] [Green Version]
  43. Halvorsen, E.C.; Mahmoud, S.M.; Bennewith, K.L. Emerging roles of regulatory T cells in tumour progression and metastasis. Cancer Metastasis Rev. 2014, 33, 1025–1041. [Google Scholar] [CrossRef] [PubMed]
  44. Beyer, M.; Schultze, J.L. Regulatory T cells in cancer. Blood 2006, 108, 804–811. [Google Scholar] [CrossRef] [Green Version]
  45. Vignali, D.A.A.; Collison, L.W.; Workman, C.J. How regulatory T cells work. Nat. Rev. Immunol. 2008, 8, 523–532. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  46. Lim, W.C.; Olding, M.; Healy, E.; Millar, T.M. Human Endothelial Cells Modulate CD4+ T Cell Populations and Enhance Regulatory T Cell Suppressive Capacity. Front. Immunol. 2018, 9, 565. [Google Scholar] [CrossRef] [Green Version]
  47. Comito, G.; Iscaro, A.; Bacci, M.; Morandi, A.; Ippolito, L.; Parri, M.; Montagnani, I.; Raspollini, M.R.; Serni, S.; Simeoni, L.; et al. Lactate modulates CD4+ T-cell polarization and induces an immunosuppressive environment, which sustains prostate carcinoma progression via TLR8/miR21 axis. Oncogene 2019, 38, 3681–3695. [Google Scholar] [CrossRef] [PubMed]
  48. Huppert, L.A.; Green, M.D.; Kim, L.; Chow, C.; Leyfman, Y.; Daud, A.I.; Lee, J.C. Tissue-specific Tregs in cancer metastasis: Opportunities for precision immunotherapy. Cell Mol. Immunol. 2022, 19, 33–45. [Google Scholar] [CrossRef]
  49. Tao, H.; Mimura, Y.; Aoe, K.; Kobayashi, S.; Yamamoto, H.; Matsuda, E.; Okabe, K.; Matsumoto, T.; Sugi, K.; Ueoka, H. Prognostic potential of FOXP3 expression in non-small cell lung cancer cells combined with tumor-infiltrating regulatory T cells. Lung. Cancer 2012, 75, 95–101. [Google Scholar] [CrossRef]
  50. Cheng, N.; Bai, X.; Shu, Y.; Ahmad, O.; Shen, P. Targeting tumor-associated macrophages as an antitumor strategy. Biochem. Pharmacol. 2021, 183, 114354. [Google Scholar] [CrossRef]
  51. Lin, Y.; Xu, J.; Lan, H. Tumor-associated macrophages in tumor metastasis: Biological roles and clinical therapeutic applications. J. Hematol. Oncol. 2019, 12, 76. [Google Scholar] [CrossRef]
  52. Pan, Y.; Yu, Y.; Wang, X.; Zhang, T. Tumor-Associated Macrophages in Tumor Immunity. Front. Immunol. 2020, 11, 583084. [Google Scholar] [CrossRef]
  53. Biswas, S.K.; Mantovani, A. Macrophage plasticity and interaction with lymphocyte subsets: Cancer as a paradigm. Nat. Immunol. 2010, 11, 889–896. [Google Scholar] [CrossRef] [PubMed]
  54. Li, M.; He, L.; Zhu, J.; Zhang, P.; Liang, S. Targeting tumor-associated macrophages for cancer treatment. Cell Biosci. 2022, 12, 85. [Google Scholar] [CrossRef] [PubMed]
  55. Bernsmeier, C.; van der Merwe, S.; Périanin, A. Innate immune cells in cirrhosis. J. Hepatol. 2020, 73, 186–201. [Google Scholar] [CrossRef] [PubMed]
  56. Bruns, H.; Büttner, M.; Fabri, M.; Mougiakakos, D.; Bittenbring, J.T.; Hoffmann, M.H.; Beier, F.; Pasemann, S.; Jitschin, R.; Hofmann, A.D.; et al. Vitamin D-dependent induction of cathelicidin in human macrophages results in cytotoxicity against high-grade B cell lymphoma. Sci. Transl. Med. 2015, 7, 282ra47. [Google Scholar] [CrossRef] [PubMed]
  57. Movahedi, K.; Laoui, D.; Gysemans, C.; Baeten, M.; Stangé, G.; Van den Bossche, J.; Mack, M.; Pipeleers, D.; In’t Veld, P.; De Baetselier, P.; et al. Different tumor microenvironments contain functionally distinct subsets of macrophages derived from Ly6C(high) monocytes. Cancer Res. 2010, 70, 5728–5739. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  58. Ngambenjawong, C.; Gustafson, H.H.; Pun, S.H. Progress in tumor-associated macrophage (TAM)-targeted therapeutics. Adv. Drug Deliv. Rev. 2017, 114, 206–221. [Google Scholar] [CrossRef] [Green Version]
  59. Dallavalasa, S.; Beeraka, N.M.; Basavaraju, C.G.; Tulimilli, S.V.; Sadhu, S.P.; Rajesh, K.; Aliev, G.; Madhunapantula, S.V. The Role of Tumor Associated Macrophages (TAMs) in Cancer Progression, Chemoresistance, Angiogenesis and Metastasis—Current Status. Curr. Med. Chem. 2021, 28, 8203–8236. [Google Scholar] [CrossRef]
  60. Fu, L.-Q.; Du, W.-L.; Cai, M.-H.; Yao, J.-Y.; Zhao, Y.-Y.; Mou, X.-Z. The roles of tumor-associated macrophages in tumor angiogenesis and metastasis. Cell Immunol. 2020, 353, 104119. [Google Scholar] [CrossRef]
  61. Gazzaniga, S.; Bravo, A.I.; Guglielmotti, A.; van Rooijen, N.; Maschi, F.; Vecchi, A.; Mantovani, A.; Mordoh, J.; Wainstok, R. Targeting tumor-associated macrophages and inhibition of MCP-1 reduce angiogenesis and tumor growth in a human melanoma xenograft. J. Investig. Dermatol. 2007, 127, 2031–2041. [Google Scholar] [CrossRef] [Green Version]
  62. Cortese, N.; Soldani, C.; Franceschini, B.; Barbagallo, M.; Marchesi, F.; Torzilli, G.; Donadon, M. Macrophages in Colorectal Cancer Liver Metastases. Cancers 2019, 11, 633. [Google Scholar] [CrossRef] [Green Version]
  63. Msaouel, P.; Genovese, G.; Gao, J.; Sen, S.; Tannir, N.M. TAM kinase inhibition and immune checkpoint blockade- a winning combination in cancer treatment? Expert Opin. Ther. Targets 2021, 25, 141–151. [Google Scholar] [CrossRef] [PubMed]
  64. Constantino, J.; Gomes, C.; Falcão, A.; Neves, B.M.; Cruz, M.T. Dendritic cell-based immunotherapy: A basic review and recent advances. Immunol. Res 2017, 65, 798–810. [Google Scholar] [CrossRef] [PubMed]
  65. Gardner, A.; Ruffell, B. Dendritic Cells and Cancer Immunity. Trends Immunol. 2016, 37, 855–865. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  66. Salmon, H.; Idoyaga, J.; Rahman, A.; Leboeuf, M.; Remark, R.; Jordan, S.; Casanova-Acebes, M.; Khudoynazarova, M.; Agudo, J.; Tung, N.; et al. Expansion and activation of CD103+ dendritic cell progenitors at the tumor site enhances tumor responses to therapeutic PD-L1 and BRAF inhibition. Immunity 2016, 44, 924–938. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  67. Zhu, S.; Yang, N.; Wu, J.; Wang, X.; Wang, W.; Liu, Y.-J.; Chen, J. Tumor microenvironment-related dendritic cell deficiency: A target to enhance tumor immunotherapy. Pharmacol. Res. 2020, 159, 104980. [Google Scholar] [CrossRef]
  68. Escors, D. Tumour immunogenicity, antigen presentation and immunological barriers in cancer immunotherapy. N. J. Sci. 2014, 2014, 734515. [Google Scholar] [CrossRef] [Green Version]
  69. Mathan, T.S.M.; Figdor, C.G.; Buschow, S.I. Human Plasmacytoid Dendritic Cells: From Molecules to Intercellular Communication Network. Front. Immunol. 2013, 4, 372. [Google Scholar] [CrossRef] [Green Version]
  70. Bi, E.; Li, R.; Bover, L.C.; Li, H.; Su, P.; Ma, X.; Huang, C.; Wang, Q.; Liu, L.; Yang, M.; et al. E-cadherin expression on multiple myeloma cells activates tumor-promoting properties in plasmacytoid DCs. J. Clin. Investig. 2018, 128, 4821–4831. [Google Scholar] [CrossRef] [Green Version]
  71. Yang, L.-L.; Mao, L.; Wu, H.; Chen, L.; Deng, W.-W.; Xiao, Y.; Li, H.; Zhang, L.; Sun, Z.-J. pDC depletion induced by CD317 blockade drives the antitumor immune response in head and neck squamous cell carcinoma. Oral. Oncol. 2019, 96, 131–139. [Google Scholar] [CrossRef]
  72. Kranz, L.M.; Diken, M.; Haas, H.; Kreiter, S.; Loquai, C.; Reuter, K.C.; Meng, M.; Fritz, D.; Vascotto, F.; Hefesha, H.; et al. Systemic RNA delivery to dendritic cells exploits antiviral defence for cancer immunotherapy. Nature 2016, 534, 396–401. [Google Scholar] [CrossRef]
  73. Mildner, A.; Jung, S. Development and function of dendritic cell subsets. Immunity 2014, 40, 642–656. [Google Scholar] [CrossRef] [Green Version]
  74. Collin, M.; Bigley, V. Human dendritic cell subsets: An update. Immunology 2018, 154, 3–20. [Google Scholar] [CrossRef] [Green Version]
  75. Joffre, O.P.; Segura, E.; Savina, A.; Amigorena, S. Cross-presentation by dendritic cells. Nat. Rev. Immunol. 2012, 12, 557–569. [Google Scholar] [CrossRef]
  76. Fucikova, J.; Palova-Jelinkova, L.; Bartunkova, J.; Spisek, R. Induction of Tolerance and Immunity by Dendritic Cells: Mechanisms and Clinical Applications. Front. Immunol. 2019, 10, 2393. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  77. Motta, J.M.; Rumjanek, V.M. Sensitivity of Dendritic Cells to Microenvironment Signals. J. Immunol. Res. 2016, 2016, 4753607. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  78. Verneau, J.; Sautés-Fridman, C.; Sun, C.-M. Dendritic cells in the tumor microenvironment: Prognostic and theranostic impact. Semin. Immunol. 2020, 48, 101410. [Google Scholar] [CrossRef] [PubMed]
  79. Gkirtzimanaki, K.; Kabrani, E.; Nikoleri, D.; Polyzos, A.; Blanas, A.; Sidiropoulos, P.; Makrigiannakis, A.; Bertsias, G.; Boumpas, D.T.; Verginis, P. IFNα Impairs Autophagic Degradation of mtDNA Promoting Autoreactivity of SLE Monocytes in a STING-Dependent Fashion. Cell Rep. 2018, 25, 921–933.e5. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  80. Subbiah, V.; Murthy, R.; Hong, D.S.; Prins, R.M.; Hosing, C.; Hendricks, K.; Kolli, D.; Noffsinger, L.; Brown, R.; McGuire, M.; et al. Cytokines Produced by Dendritic Cells Administered Intratumorally Correlate with Clinical Outcome in Patients with Diverse Cancers. Clin. Cancer Res. 2018, 24, 3845–3856. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  81. Zhong, H.; Han, B.; Tourkova, I.L.; Lokshin, A.; Rosenbloom, A.; Shurin, M.R.; Shurin, G.V. Low-Dose Paclitaxel Prior to Intratumoral Dendritic Cell Vaccine Modulates Intratumoral Cytokine Network and Lung Cancer Growth. Clin. Cancer Res. 2007, 13, 5455–5462. [Google Scholar] [CrossRef] [Green Version]
  82. Mattiola, I. Immune Circuits to Shape Natural Killer Cells in Cancer. Cancers 2021, 13, 3225. [Google Scholar] [CrossRef]
  83. Marofi, F.; Al-Awad, A.S.; Sulaiman Rahman, H.; Markov, A.; Abdelbasset, W.K.; Ivanovna Enina, Y.; Mahmoodi, M.; Hassanzadeh, A.; Yazdanifar, M.; Stanley Chartrand, M.; et al. CAR-NK Cell: A New Paradigm in Tumor Immunotherapy. Front. Oncol. 2021, 11, 673276. [Google Scholar] [CrossRef] [PubMed]
  84. Carnevalli, L.S.; Ghadially, H.; Barry, S.T. Therapeutic Approaches Targeting the Natural Killer-Myeloid Cell Axis in the Tumor Microenvironment. Front. Immunol. 2021, 12, 633685. [Google Scholar] [CrossRef] [PubMed]
  85. Jewett, A.; Kos, J.; Kaur, K.; Safaei, T.; Sutanto, C.; Chen, W.; Wong, P.; Namagerdi, A.K.; Fang, C.; Fong, Y.; et al. Natural Killer Cells: Diverse Functions in Tumor Immunity and Defects in Pre-Neoplastic and Neoplastic Stages of Tumorigenesis. Mol. Ther. Oncolytics 2019, 16, 41–52. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  86. Melaiu, O.; Lucarini, V.; Cifaldi, L.; Fruci, D. Influence of the Tumor Microenvironment on NK Cell Function in Solid Tumors. Front. Immunol. 2020, 10, 3038. [Google Scholar] [CrossRef] [PubMed]
  87. Wagner, J.A.; Rosario, M.; Romee, R.; Berrien-Elliott, M.M.; Schneider, S.E.; Leong, J.W.; Sullivan, R.P.; Jewell, B.A.; Becker-Hapak, M.; Schappe, T.; et al. CD56bright NK cells exhibit potent antitumor responses following IL-15 priming. J. Clin. Investig. 2017, 127, 4042–4058. [Google Scholar] [CrossRef] [Green Version]
  88. Prager, I.; Liesche, C.; van Ooijen, H.; Urlaub, D.; Verron, Q.; Sandström, N.; Fasbender, F.; Claus, M.; Eils, R.; Beaudouin, J.; et al. NK cells switch from granzyme B to death receptor–mediated cytotoxicity during serial killing. J. Exp. Med. 2019, 216, 2113–2127. [Google Scholar] [CrossRef] [Green Version]
  89. Gemelli, M.; Noonan, D.M.; Carlini, V.; Pelosi, G.; Barberis, M.; Ricotta, R.; Albini, A. Overcoming Resistance to Checkpoint Inhibitors: Natural Killer Cells in Non-Small Cell Lung Cancer. Front. Oncol. 2022, 12, 886440. [Google Scholar] [CrossRef]
  90. Gaggero, S.; Witt, K.; Carlsten, M.; Mitra, S. Cytokines Orchestrating the Natural Killer-Myeloid Cell Crosstalk in the Tumor Microenvironment: Implications for Natural Killer Cell-Based Cancer Immunotherapy. Front. Immunol. 2021, 11, 621225. [Google Scholar] [CrossRef]
  91. Terrén, I.; Orrantia, A.; Vitallé, J.; Zenarruzabeitia, O.; Borrego, F. NK Cell Metabolism and Tumor Microenvironment. Front. Immunol. 2019, 10, 2278. [Google Scholar] [CrossRef]
  92. Myers, J.A.; Miller, J.S. Exploring the NK cell platform for cancer immunotherapy. Nat. Rev. Clin. Oncol. 2021, 18, 85–100. [Google Scholar] [CrossRef]
  93. Vitale, M.; Cantoni, C.; Pietra, G.; Mingari, M.C.; Moretta, L. Effect of tumor cells and tumor microenvironment on NK-cell function. Eur. J. Immunol. 2014, 44, 1582–1592. [Google Scholar] [CrossRef] [PubMed]
  94. Wahl, S.M.; Wen, J.; Moutsopoulos, N.M. The kiss of death: Interrupted by NK-cell close encounters of another kind. Trends Immunol. 2006, 27, 161–164. [Google Scholar] [CrossRef] [PubMed]
  95. Liu, C.; Lai, H.; Chen, T. Boosting Natural Killer Cell-Based Cancer Immunotherapy with Selenocystine/Transforming Growth Factor-Beta Inhibitor-Encapsulated Nanoemulsion. ACS Nano 2020, 14, 11067–11082. [Google Scholar] [CrossRef]
  96. Mollinedo, F. Neutrophil Degranulation, Plasticity, and Cancer Metastasis. Trends Immunol. 2019, 40, 228–242. [Google Scholar] [CrossRef]
  97. Xiong, S.; Dong, L.; Cheng, L. Neutrophils in cancer carcinogenesis and metastasis. J. Hematol. Oncol. 2021, 14, 173. [Google Scholar] [CrossRef]
  98. Templeton, A.J.; McNamara, M.G.; Šeruga, B.; Vera-Badillo, F.E.; Aneja, P.; Ocaña, A.; Leibowitz-Amit, R.; Sonpavde, G.; Knox, J.J.; Tran, B.; et al. Prognostic role of neutrophil-to-lymphocyte ratio in solid tumors: A systematic review and meta-analysis. J. Natl. Cancer Inst. 2014, 106, dju124. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  99. Masucci, M.T.; Minopoli, M.; Del Vecchio, S.; Carriero, M.V. The Emerging Role of Neutrophil Extracellular Traps (NETs) in Tumor Progression and Metastasis. Front. Immunol. 2020, 11, 1749. [Google Scholar] [CrossRef] [PubMed]
  100. Mouchemore, K.A.; Anderson, R.L.; Hamilton, J.A. Neutrophils, G-CSF and their contribution to breast cancer metastasis. FEBS J. 2018, 285, 665–679. [Google Scholar] [CrossRef] [Green Version]
  101. Jung, H.S.; Gu, J.; Kim, J.-E.; Nam, Y.; Song, J.W.; Kim, H.K. Cancer cell-induced neutrophil extracellular traps promote both hypercoagulability and cancer progression. PLoS ONE 2019, 14, e0216055. [Google Scholar] [CrossRef] [Green Version]
  102. Demers, M.; Wagner, D.D. Neutrophil extracellular traps: A new link to cancer-associated thrombosis and potential implications for tumor progression. Oncoimmunology 2013, 2, e22946. [Google Scholar] [CrossRef] [Green Version]
  103. Gabrilovich, D.I. Myeloid-Derived Suppressor Cells. Cancer Immunol. Res. 2017, 5, 3–8. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  104. Li, Y.; He, H.; Jihu, R.; Zhou, J.; Zeng, R.; Yan, H. Novel Characterization of Myeloid-Derived Suppressor Cells in Tumor Microenvironment. Front. Cell Dev. Biol. 2021, 9, 698532. [Google Scholar] [CrossRef] [PubMed]
  105. Condamine, T.; Dominguez, G.A.; Youn, J.-I.; Kossenkov, A.V.; Mony, S.; Alicea-Torres, K.; Tcyganov, E.; Hashimoto, A.; Nefedova, Y.; Lin, C.; et al. Lectin-type oxidized LDL receptor-1 distinguishes population of human polymorphonuclear myeloid-derived suppressor cells in cancer patients. Sci. Immunol. 2016, 1, aaf8943. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  106. Condamine, T.; Ramachandran, I.; Youn, J.-I.; Gabrilovich, D.I. Regulation of tumor metastasis by myeloid-derived suppressor cells. Annu. Rev. Med. 2015, 66, 97–110. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  107. Engelhard, V.; Conejo-Garcia, J.R.; Ahmed, R.; Nelson, B.H.; Willard-Gallo, K.; Bruno, T.C.; Fridman, W.H. B cells and cancer. Cancer Cell 2021, 39, 1293–1296. [Google Scholar] [CrossRef]
  108. Kroeger, D.R.; Milne, K.; Nelson, B.H. Tumor-Infiltrating Plasma Cells Are Associated with Tertiary Lymphoid Structures, Cytolytic T-Cell Responses, and Superior Prognosis in Ovarian Cancer. Clin. Cancer Res. J. Am. Assoc. Cancer Res. 2016, 22, 3005–3015. [Google Scholar] [CrossRef] [Green Version]
  109. Sautès-Fridman, C.; Petitprez, F.; Calderaro, J.; Fridman, W.H. Tertiary lymphoid structures in the era of cancer immunotherapy. Nat. Rev. Cancer 2019, 19, 307–325. [Google Scholar] [CrossRef]
  110. West, H.; McCleod, M.; Hussein, M.; Morabito, A.; Rittmeyer, A.; Conter, H.J.; Kopp, H.-G.; Daniel, D.; McCune, S.; Mekhail, T.; et al. Atezolizumab in combination with carboplatin plus nab-paclitaxel chemotherapy compared with chemotherapy alone as first-line treatment for metastatic non-squamous non-small-cell lung cancer (IMpower130): A multicentre, randomised, open-label, phase 3 trial. Lancet Oncol. 2019, 20, 924–937. [Google Scholar] [CrossRef]
  111. Neoadjuvant Chemo-ICI Boosts NSCLC Survival. Cancer Discov. 2022, 12, 2228. [CrossRef]
  112. Xia, L.; Liu, Y.; Wang, Y. PD-1/PD-L1 Blockade Therapy in Advanced Non-Small-Cell Lung Cancer: Current Status and Future Directions. Oncologist 2019, 24, S31–S41. [Google Scholar] [CrossRef] [Green Version]
  113. Kono, K.; Nakajima, S.; Mimura, K. Current status of immune checkpoint inhibitors for gastric cancer. Gastric Cancer 2020, 23, 565–578. [Google Scholar] [CrossRef] [PubMed]
  114. Yi, M.; Jiao, D.; Qin, S.; Chu, Q.; Wu, K.; Li, A. Synergistic effect of immune checkpoint blockade and anti-angiogenesis in cancer treatment. Mol. Cancer 2019, 18, 60. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  115. Carlino, M.S.; Larkin, J.; Long, G.V. Immune checkpoint inhibitors in melanoma. Lancet 2021, 398, 1002–1014. [Google Scholar] [CrossRef]
  116. Tang, S.; Qin, C.; Hu, H.; Liu, T.; He, Y.; Guo, H.; Yan, H.; Zhang, J.; Tang, S.; Zhou, H. Immune Checkpoint Inhibitors in Non-Small Cell Lung Cancer: Progress, Challenges, and Prospects. Cells 2022, 11, 320. [Google Scholar] [CrossRef] [PubMed]
  117. Sharma, P.; Allison, J.P. Immune checkpoint targeting in cancer therapy: Toward combination strategies with curative potential. Cell 2015, 161, 205–214. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  118. Bravo Montenegro, G.; Farid, S.; Liu, S.V. Immunotherapy in lung cancer. J. Surg. Oncol. 2021, 123, 718–729. [Google Scholar] [CrossRef]
  119. Grant, M.J.; Herbst, R.S.; Goldberg, S.B. Selecting the optimal immunotherapy regimen in driver-negative metastatic NSCLC. Nat. Rev. Clin. Oncol. 2021, 18, 625–644. [Google Scholar] [CrossRef]
  120. Keir, M.E.; Butte, M.J.; Freeman, G.J.; Sharpe, A.H. PD-1 and its ligands in tolerance and immunity. Annu. Rev. Immunol. 2008, 26, 677–704. [Google Scholar] [CrossRef] [Green Version]
  121. Sun, C.; Mezzadra, R.; Schumacher, T.N. Regulation and Function of the PD-L1 Checkpoint. Immunity 2018, 48, 434–452. [Google Scholar] [CrossRef] [Green Version]
  122. Bristol-Myers Squibb An Open-Label Randomized Phase III Trial of BMS-936558 (Nivolumab) Versus Docetaxel in Previously Treated Advanced or Metastatic Squamous Cell Non-Small Cell Lung Cancer (NSCLC). 2012. Available online: https://ClinicalTrials.gov/show/NCT01642004 (accessed on 13 April 2023).
  123. Bristol-Myers Squibb Study of BMS-936558 (Nivolumab) Compared to Docetaxel in Previously Treated Metastatic Non-squamous NSCLC (CheckMate057). 2012. Available online: https://ClinicalTrials.gov/show/NCT01673867 (accessed on 13 April 2023).
  124. Merck Sharp & Dohme LLC A Randomized Open-Label Phase III Trial of Pembrolizumab Versus Platinum Based Chemotherapy in 1L Subjects with PD-L1 Strong Metastatic Non-Small Cell Lung Cancer. 2014. Available online: https://ClinicalTrials.gov/show/NCT02142738 (accessed on 13 April 2023).
  125. Merck Sharp & Dohme LLC A Randomized, Open Label, Phase III Study of Overall Survival Comparing Pembrolizumab (MK-3475) Versus Platinum Based Chemotherapy in Treatment Naïve Subjects With PD-L1 Positive Advanced or Metastatic Non-Small Cell Lung Cancer (Keynote 042). 2019. Available online: https://ClinicalTrials.gov/show/NCT03850444 (accessed on 13 April 2023).
  126. Hoffmann-La Roche A Phase III, Open Label, Randomized Study of Atezolizumab (Anti-PD-L1 Antibody) Compared with a Platinum Agent (Cisplatin or Carboplatin) in Combination with Either Pemetrexed or Gemcitabine for PD-L1-Selected, Chemotherapy-Naive Patients with Stage IV Non-Squamous Or Squamous Non-Small Cell Lung Cancer. 2015. Available online: https://ClinicalTrials.gov/show/NCT02409342 (accessed on 13 April 2023).
  127. Krummel, M.F.; Allison, J.P. CD28 and CTLA-4 have opposing effects on the response of T cells to stimulation. J. Exp. Med. 1995, 182, 459–465. [Google Scholar] [CrossRef] [Green Version]
  128. Wei, S.C.; Sharma, R.; Anang, N.-A.A.S.; Levine, J.H.; Zhao, Y.; Mancuso, J.J.; Setty, M.; Sharma, P.; Wang, J.; Pe’er, D.; et al. Negative Co-Stimulation Constrains T Cell Differentiation by Imposing Boundaries on Possible Cell States. Immunity 2019, 50, 1084–1098. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  129. Buchbinder, E.I.; Desai, A. CTLA-4 and PD-1 Pathways. Am. J. Clin. Oncol. 2016, 39, 98–106. [Google Scholar] [CrossRef] [Green Version]
  130. Patel, S.A.; Weiss, J. Advances in the Treatment of Non-Small Cell Lung Cancer: Immunotherapy. Clin. Chest. Med. 2020, 41, 237–247. [Google Scholar] [CrossRef] [PubMed]
  131. Bristol-Myers Squibb An Open-Label, Randomized Phase 3 Trial of Nivolumab, or Nivolumab Plus Ipilimumab, or Nivolumab Plus Platinum Doublet Chemotherapy Versus Platinum Doublet Chemotherapy in Subjects with Chemotherapy-Naïve Stage IV or Recurrent Non-Small Cell Lung Cancer (NSCLC). 2015. Available online: https://ClinicalTrials.gov/show/NCT02477826 (accessed on 13 April 2023).
  132. Hellmann, M.D.; Paz-Ares, L.; Bernabe Caro, R.; Zurawski, B.; Kim, S.-W.; Carcereny Costa, E.; Park, K.; Alexandru, A.; Lupinacci, L.; de la Mora Jimenez, E.; et al. Nivolumab plus Ipilimumab in Advanced Non-Small-Cell Lung Cancer. N. Engl. J. Med. 2019, 381, 2020–2031. [Google Scholar] [CrossRef] [PubMed]
  133. Merck Sharp & Dohme LLC A Randomized, Double-Blind, Phase III Study of Carboplatin-Paclitaxel/Nab-Paclitaxel Chemotherapy with or Without Pembrolizumab (MK-3475) in First Line Metastatic Squamous Non-small Cell Lung Cancer Subjects (KEYNOTE-407). 2019. Available online: https://ClinicalTrials.gov/show/NCT03875092 (accessed on 13 April 2023).
  134. Epacadostat Shows Value in Two SCCHN Trials. Cancer Discov. 2017, 7, OF2. [CrossRef] [Green Version]
  135. Einstein, D.J.; McDermott, D.F. Combined blockade of vascular endothelial growth factor and programmed death 1 pathways in advanced kidney cancer. Clin. Adv. Hematol. Oncol. 2017, 15, 478–488. [Google Scholar]
  136. He, Y.; Yu, H.; Rozeboom, L.; Rivard, C.J.; Ellison, K.; Dziadziuszko, R.; Suda, K.; Ren, S.; Wu, C.; Hou, L.; et al. LAG-3 Protein Expression in Non-Small Cell Lung Cancer and Its Relationship with PD-1/PD-L1 and Tumor-Infiltrating Lymphocytes. J. Thorac. Oncol. 2017, 12, 814–823. [Google Scholar] [CrossRef] [Green Version]
  137. Liu, J.; Ma, S.; Mao, L.; Bu, L.; Yu, G.; Li, Y.; Huang, C.; Deng, W.; Kulkarni, A.B.; Zhang, W.; et al. T-cell immunoglobulin mucin 3 blockade drives an antitumor immune response in head and neck cancer. Mol. Oncol. 2017, 11, 235–247. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  138. Zhang, L.; Li, H.; Zhang, F.; Wang, S.; Li, G. CAR-T Immunotherapy and Non-small Cell Lung Cancer: Bottleneck and Dawn. Zhongguo Fei Ai Za Zhi 2020, 23, 916–920. [Google Scholar] [CrossRef]
  139. Srivastava, S.; Riddell, S.R. Engineering CAR-T Cells: Design Concepts. Trends Immunol. 2015, 36, 494–502. [Google Scholar] [CrossRef] [Green Version]
  140. Ying, Z.; Huang, X.F.; Xiang, X.; Liu, Y.; Kang, X.; Song, Y.; Guo, X.; Liu, H.; Ding, N.; Zhang, T.; et al. A safe and potent anti-CD19 CAR T cell therapy. Nat. Med. 2019, 25, 947–953. [Google Scholar] [CrossRef] [PubMed]
  141. Qu, J.; Mei, Q.; Chen, L.; Zhou, J. Chimeric antigen receptor (CAR)-T-cell therapy in non-small-cell lung cancer (NSCLC): Current status and future perspectives. Cancer Immunol. Immunother. 2021, 70, 619–631. [Google Scholar] [CrossRef] [PubMed]
  142. Dong, J.; Li, B.; Lin, D.; Zhou, Q.; Huang, D. Advances in Targeted Therapy and Immunotherapy for Non-small Cell Lung Cancer Based on Accurate Molecular Typing. Front. Pharmacol. 2019, 10, 230. [Google Scholar] [CrossRef] [PubMed]
  143. Zhao, J.; Xiong, J. Advances on driver oncogenes of non-small cell lung cancer. Zhongguo Fei Ai Za Zhi 2015, 18, 42–47. [Google Scholar] [CrossRef] [PubMed]
  144. Mogi, A.; Kuwano, H. TP53 mutations in nonsmall cell lung cancer. J. Biomed. Biotechnol. 2011, 2011, 583929. [Google Scholar] [CrossRef] [Green Version]
  145. Borghaei, H.; Paz-Ares, L.; Horn, L.; Spigel, D.R.; Steins, M.; Ready, N.E.; Chow, L.Q.; Vokes, E.E.; Felip, E.; Holgado, E.; et al. Nivolumab versus Docetaxel in Advanced Non-squamous Non-small Cell Lung Cancer. N. Engl. J. Med. 2015, 373, 1627. [Google Scholar] [CrossRef]
  146. Mazieres, J.; Drilon, A.; Lusque, A.; Mhanna, L.; Cortot, A.B.; Mezquita, L.; Thai, A.A.; Mascaux, C.; Couraud, S.; Veillon, R.; et al. Immune checkpoint inhibitors for patients with advanced lung cancer and oncogenic driver alterations: Results from the IMMUNOTARGET registry. Ann. Oncol. 2019, 30, 1321–1328. [Google Scholar] [CrossRef]
  147. Schoenfeld, A.J.; Rizvi, H.; Bandlamudi, C.; Sauter, J.L.; Travis, W.D.; Rekhtman, N.; Plodkowski, A.J.; Perez-Johnston, R.; Sawan, P.; Beras, A.; et al. Clinical and molecular correlates of PD-L1 expression in patients with lung adenocarcinomas. Ann. Oncol. J. Eur. Soc. Med. Oncol. 2020, 31, 599–608. [Google Scholar] [CrossRef]
  148. Qiao, M.; Jiang, T.; Liu, X.; Mao, S.; Zhou, F.; Li, X.; Zhao, C.; Chen, X.; Su, C.; Ren, S.; et al. Immune Checkpoint Inhibitors in EGFR-Mutated NSCLC: Dusk or Dawn? J. Thorac. Oncol. 2021, 16, 1267–1288. [Google Scholar] [CrossRef]
  149. Imyanitov, E.N.; Iyevleva, A.G.; Levchenko, E.V. Molecular testing and targeted therapy for non-small cell lung cancer: Current status and perspectives. Crit. Rev. Oncol. Hematol. 2021, 157, 103194. [Google Scholar] [CrossRef]
  150. Bravaccini, S.; Bronte, G.; Ulivi, P. TMB in NSCLC: A Broken Dream? Int. J. Mol. Sci. 2021, 22, 6536. [Google Scholar] [CrossRef] [PubMed]
  151. Masuda, K.; Horinouchi, H.; Tanaka, M.; Higashiyama, R.; Shinno, Y.; Sato, J.; Matsumoto, Y.; Okuma, Y.; Yoshida, T.; Goto, Y.; et al. Efficacy of anti-PD-1 antibodies in NSCLC patients with an EGFR mutation and high PD-L1 expression. J. Cancer Res. Clin. Oncol. 2021, 147, 245–251. [Google Scholar] [CrossRef] [PubMed]
  152. Yang, J.C.-H.; Sequist, L.V.; Geater, S.L.; Tsai, C.-M.; Mok, T.S.K.; Schuler, M.; Yamamoto, N.; Yu, C.-J.; Ou, S.-H.I.; Zhou, C.; et al. Clinical activity of afatinib in patients with advanced non-small-cell lung cancer harbouring uncommon EGFR mutations: A combined post-hoc analysis of LUX-Lung 2, LUX-Lung 3, and LUX-Lung 6. Lancet Oncol. 2015, 16, 830–838. [Google Scholar] [CrossRef]
  153. Remon, J.; Steuer, C.E.; Ramalingam, S.S.; Felip, E. Osimertinib and other third-generation EGFR TKI in EGFR-mutant NSCLC patients. Ann. Oncol. 2018, 29, i20–i27. [Google Scholar] [CrossRef] [PubMed]
  154. Park, S.; Choi, Y.; Kim, J.; Kho, B.; Park, C.; Oh, I.; Kim, Y. Efficacy of immune checkpoint inhibitors according to PD-L1 tumor proportion scores in non-small cell lung cancer. Thorac. Cancer 2020, 11, 408–414. [Google Scholar] [CrossRef]
  155. Uehara, Y.; Watanabe, K.; Hakozaki, T.; Yomota, M.; Hosomi, Y. Efficacy of first-line immune checkpoint inhibitors in patients with advanced NSCLC with KRAS, MET, FGFR, RET, BRAF, and HER2 alterations. Thorac. Cancer 2022, 13, 1703–1711. [Google Scholar] [CrossRef]
  156. Zhang, L.; Zhang, T.; Shang, B.; Li, Y.; Cao, Z.; Wang, H. Prognostic effect of coexisting TP53 and ZFHX3 mutations in non-small cell lung cancer patients treated with immune checkpoint inhibitors. Scand. J. Immunol. 2021, 94, e13087. [Google Scholar] [CrossRef]
  157. VA Office of Research and Development Phase Ib/II Study of Safety and Efficacy of EZH2 Inhibitor, Tazemetostat, and PD-1 Blockade for Treatment of Advanced Non-small Cell Lung Cancer. 2022. Available online: https://ClinicalTrials.gov/show/NCT05467748 (accessed on 13 April 2023).
  158. OncoResponse, Inc. A Phase 1-2 Study of OR2805, a Monoclonal Antibody Targeting CD163, Alone and in Combination With a PD-1 Inhibitor in Subjects with Advanced Malignancies. 2021. Available online: https://ClinicalTrials.gov/show/NCT05094804 (accessed on 13 April 2023).
  159. Steuer, C. A Phase 1b Safety and Pharmacodynamic Study of MER Tyrosine Kinase Inhibitor, MRX-2843, in Combination with Osimertinib in Advanced EGFR Mutant Non-Small Cell Lung Cancer. 2021. Available online: https://ClinicalTrials.gov/show/NCT04762199 (accessed on 13 April 2023).
  160. Hirschowitz, E. Autologous Dendritic Cell Vaccines in Non-small Cell Lung Cancer (NSCLC). 2005. Available online: https://ClinicalTrials.gov/show/NCT00103116 (accessed on 13 April 2023).
  161. Personalized Cellular Vaccine for Brain Metastases (PERCELLVAC3)—Tabular View—ClinicalTrials.gov. Available online: https://clinicaltrials.gov/ct2/show/record/NCT02808416 (accessed on 13 April 2023).
  162. Fuda Cancer Hospital, Guangzhou Combination of Anti-PD-1 and NK Immunotherapy for Recurrent Solid Tumors. 2016. Available online: https://ClinicalTrials.gov/show/NCT02843204 (accessed on 13 April 2023).
  163. Combination of Cryosurgery and NK Immunotherapy for Advanced Non-Small Cell Lung Cancer—Full Text View—ClinicalTrials.gov. Available online: https://clinicaltrials.gov/ct2/show/NCT02843815 (accessed on 13 April 2023).
  164. Targeted Natural Killer (NK) Cell Based Adoptive Immunotherapy for the Treatment of Patients with Non-Small Cell Lung Cancer (NSCLC) After Radiochemotherapy (RCT)—Full Text View—ClinicalTrials.gov. Available online: https://clinicaltrials.gov/ct2/show/NCT02118415 (accessed on 13 April 2023).
  165. Wang, N.; Wang, S.; Wang, X.; Zheng, Y.; Yang, B.; Zhang, J. Research trends in pharmacological modulation of tumor-associated macrophages. Clin. Transl. Med. 2021, 11, e288. [Google Scholar] [CrossRef]
  166. Xia, L.; Zhu, X.; Zhang, L.; Xu, Y.; Chen, G.; Luo, J. EZH2 enhances expression of CCL5 to promote recruitment of macrophages and invasion in lung cancer. Biotechnol. Appl. Biochem. 2020, 67, 1011–1019. [Google Scholar] [CrossRef] [Green Version]
  167. Sica, A.; Allavena, P.; Mantovani, A. Cancer related inflammation: The macrophage connection. Cancer Lett. 2008, 267, 204–215. [Google Scholar] [CrossRef]
  168. Van Gorp, H.; Delputte, P.L.; Nauwynck, H.J. Scavenger receptor CD163, a Jack-of-all-trades and potential target for cell-directed therapy. Mol. Immunol. 2010, 47, 1650–1660. [Google Scholar] [CrossRef] [PubMed]
  169. Davra, V.; Kumar, S.; Geng, K.; Calianese, D.; Mehta, D.; Gadiyar, V.; Kasikara, C.; Lahey, K.C.; Chang, Y.-J.; Wichroski, M.; et al. Axl and Mertk Receptors Cooperate to Promote Breast Cancer Progression by Combined Oncogenic Signaling and Evasion of Host Antitumor Immunity. Cancer Res. 2021, 81, 698–712. [Google Scholar] [CrossRef] [PubMed]
  170. Zizzo, G.; Hilliard, B.A.; Monestier, M.; Cohen, P.L. Efficient clearance of early apoptotic cells by human macrophages requires “M2c” polarization and MerTK induction. J. Immunol. 2012, 189, 3508–3520. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  171. Caetano, M.S.; Younes, A.I.; Barsoumian, H.B.; Quigley, M.; Menon, H.; Gao, C.; Spires, T.; Reilly, T.P.; Cadena, A.P.; Cushman, T.R.; et al. Triple Therapy with MerTK and PD1 Inhibition plus Radiotherapy Promotes Abscopal Antitumor Immune Responses. Clin. Cancer Res. 2019, 25, 7576–7584. [Google Scholar] [CrossRef] [Green Version]
  172. Zhou, Y.; Fei, M.; Zhang, G.; Liang, W.-C.; Lin, W.; Wu, Y.; Piskol, R.; Ridgway, J.; McNamara, E.; Huang, H.; et al. Blockade of the Phagocytic Receptor MerTK on Tumor-Associated Macrophages Enhances P2X7R-Dependent STING Activation by Tumor-Derived cGAMP. Immunity 2020, 52, 357–373. [Google Scholar] [CrossRef]
  173. Lee, C.; Bae, S.-J.S.; Joo, H.; Bae, H. Melittin suppresses tumor progression by regulating tumor-associated macrophages in a Lewis lung carcinoma mouse model. Oncotarget 2017, 8, 54951–54965. [Google Scholar] [CrossRef] [Green Version]
  174. Lee, C.; Jeong, H.; Bae, Y.; Shin, K.; Kang, S.; Kim, H.; Oh, J.; Bae, H. Targeting of M2-like tumor-associated macrophages with a melittin-based pro-apoptotic peptide. J. Immunother. Cancer 2019, 7, 147. [Google Scholar] [CrossRef] [Green Version]
  175. Wang, B.; Zhang, W.; Zhou, X.; Liu, M.; Hou, X.; Cheng, Z.; Chen, D. Development of dual-targeted nano-dandelion based on an oligomeric hyaluronic acid polymer targeting tumor-associated macrophages for combination therapy of non-small cell lung cancer. Drug Deliv. 2019, 26, 1265–1279. [Google Scholar] [CrossRef] [Green Version]
  176. Wang, J.-B.; Huang, X.; Li, F.-R. Impaired dendritic cell functions in lung cancer: A review of recent advances and future perspectives. Cancer Commun. 2019, 39, 43. [Google Scholar] [CrossRef] [Green Version]
  177. Van der Hoorn, I.A.E.; Flórez-Grau, G.; van den Heuvel, M.M.; de Vries, I.J.M.; Piet, B. Recent Advances and Future Perspective of DC-Based Therapy in NSCLC. Front. Immunol. 2021, 12, 704776. [Google Scholar] [CrossRef]
  178. Wang, Q.-T.; Nie, Y.; Sun, S.-N.; Lin, T.; Han, R.-J.; Jiang, J.; Li, Z.; Li, J.-Q.; Xiao, Y.-P.; Fan, Y.-Y.; et al. Tumor-associated antigen-based personalized dendritic cell vaccine in solid tumor patients. Cancer Immunol. Immunother. 2020, 69, 1375–1387. [Google Scholar] [CrossRef] [PubMed]
  179. Flieswasser, T.; Van Loenhout, J.; Freire Boullosa, L.; Van den Eynde, A.; De Waele, J.; Van Audenaerde, J.; Lardon, F.; Smits, E.; Pauwels, P.; Jacobs, J. Clinically Relevant Chemotherapeutics Have the Ability to Induce Immunogenic Cell Death in Non-Small Cell Lung Cancer. Cells 2020, 9, 1474. [Google Scholar] [CrossRef] [PubMed]
  180. Hu, R.-H.; Shi, S.-B.; Qi, J.-L.; Tian, J.; Tang, X.-Y.; Liu, G.-F.; Chang, C.-X. Pemetrexed plus dendritic cells as second-line treatment for patients with stage IIIB/IV non-small cell lung cancer who had treatment with TKI. Med. Oncol 2014, 31, 63. [Google Scholar] [CrossRef] [PubMed]
  181. Zhao, M.; Li, H.; Li, L.; Zhang, Y. Effects of a gemcitabine plus platinum regimen combined with a dendritic cell-cytokine induced killer immunotherapy on recurrence and survival rate of non-small cell lung cancer patients. Exp. Ther. Med. 2014, 7, 1403–1407. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  182. Zhang, L.; Yang, X.; Sun, Z.; Li, J.; Zhu, H.; Li, J.; Pang, Y. Dendritic cell vaccine and cytokine-induced killer cell therapy for the treatment of advanced non-small cell lung cancer. Oncol. Lett. 2016, 11, 2605–2610. [Google Scholar] [CrossRef] [Green Version]
  183. Zhao, Y.; Qiao, G.; Wang, X.; Song, Y.; Zhou, X.; Jiang, N.; Zhou, L.; Huang, H.; Zhao, J.; Morse, M.A.; et al. Combination of DC/CIK adoptive T cell immunotherapy with chemotherapy in advanced non-small-cell lung cancer (NSCLC) patients: A prospective patients’ preference-based study (PPPS). Clin. Transl. Oncol. 2019, 21, 721–728. [Google Scholar] [CrossRef]
  184. Zhong, R.; Teng, J.; Han, B.; Zhong, H. Dendritic cells combining with cytokine-induced killer cells synergize chemotherapy in patients with late-stage non-small cell lung cancer. Cancer Immunol. Immunother. 2011, 60, 1497–1502. [Google Scholar] [CrossRef]
  185. Hamilton, G.; Plangger, A. The Impact of NK Cell-Based Therapeutics for the Treatment of Lung Cancer for Biologics: Targets and Therapy. Biologics 2021, 15, 265–277. [Google Scholar] [CrossRef]
  186. Bruno, A.; Focaccetti, C.; Pagani, A.; Imperatori, A.S.; Spagnoletti, M.; Rotolo, N.; Cantelmo, A.R.; Franzi, F.; Capella, C.; Ferlazzo, G.; et al. The Proangiogenic Phenotype of Natural Killer Cells in Patients with Non-Small Cell Lung Cancer. Neoplasia 2013, 15, 133–142. [Google Scholar] [CrossRef] [Green Version]
  187. Luna, J.I.; Grossenbacher, S.K.; Murphy, W.J.; Canter, R.J. Natural Killer Cell Immunotherapy Targeting Cancer Stem Cells. Expert. Opin. Biol. Ther. 2017, 17, 313–324. [Google Scholar] [CrossRef] [Green Version]
  188. Kim, S.; Iizuka, K.; Aguila, H.L.; Weissman, I.L.; Yokoyama, W.M. In vivo natural killer cell activities revealed by natural killer cell-deficient mice. Proc. Natl. Acad. Sci. USA 2000, 97, 2731–2736. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  189. Zhang, G.; Zhao, H.; Wu, J.; Li, J.; Xiang, Y.; Wang, G.; Wu, L.; Jiao, S. Adoptive immunotherapy for non-small cell lung cancer by NK and cytotoxic T lymphocytes mixed effector cells: Retrospective clinical observation. Int. Immunopharmacol. 2014, 21, 396–405. [Google Scholar] [CrossRef] [PubMed]
  190. Lin, M.; Luo, H.; Liang, S.; Chen, J.; Liu, A.; Niu, L.; Jiang, Y. Pembrolizumab plus allogeneic NK cells in advanced non–small cell lung cancer patients. J. Clin. Investig. 2020, 130, 2560–2569. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  191. Lin, M.; Liang, S.-Z.; Wang, X.-H.; Liang, Y.-Q.; Zhang, M.-J.; Niu, L.-Z.; Chen, J.-B.; Li, H.-B.; Xu, K.-C. Clinical efficacy of percutaneous cryoablation combined with allogenic NK cell immunotherapy for advanced non-small cell lung cancer. Immunol. Res. 2017, 65, 880–887. [Google Scholar] [CrossRef]
  192. Iliopoulou, E.G.; Kountourakis, P.; Karamouzis, M.V.; Doufexis, D.; Ardavanis, A.; Baxevanis, C.N.; Rigatos, G.; Papamichail, M.; Perez, S.A. A phase I trial of adoptive transfer of allogeneic natural killer cells in patients with advanced non-small cell lung cancer. Cancer Immunol. Immunother. 2010, 59, 1781–1789. [Google Scholar] [CrossRef]
  193. Shevtsov, M.; Pitkin, E.; Ischenko, A.; Stangl, S.; Khachatryan, W.; Galibin, O.; Edmond, S.; Lobinger, D.; Multhoff, G. Ex vivo Hsp70-Activated NK Cells in Combination With PD-1 Inhibition Significantly Increase Overall Survival in Preclinical Models of Glioblastoma and Lung Cancer. Front. Immunol. 2019, 10, 454. [Google Scholar] [CrossRef]
  194. Pockley, A.G.; Vaupel, P.; Multhoff, G. NK cell-based therapeutics for lung cancer. Expert Opin. Biol. Ther. 2020, 20, 23–33. [Google Scholar] [CrossRef]
  195. Daher, M.; Rezvani, K. Outlook for new CAR-based therapies with a focus on CAR-NK cells: What lies beyond CAR-engineered T cells in the race against cancer. Cancer Discov. 2021, 11, 45–58. [Google Scholar] [CrossRef]
  196. Pan, K.; Farrukh, H.; Chittepu, V.C.S.R.; Xu, H.; Pan, C.; Zhu, Z. CAR race to cancer immunotherapy: From CAR T, CAR NK to CAR macrophage therapy. J. Exp. Clin. Cancer Res. 2022, 41, 119. [Google Scholar] [CrossRef]
  197. Zhang, Y.; Wallace, D.L.; de Lara, C.M.; Ghattas, H.; Asquith, B.; Worth, A.; Griffin, G.E.; Taylor, G.P.; Tough, D.F.; Beverley, P.C.L.; et al. In vivo kinetics of human natural killer cells: The effects of ageing and acute and chronic viral infection. Immunology 2007, 121, 258–265. [Google Scholar] [CrossRef]
  198. Roma-Rodrigues, C.; Mendes, R.; Baptista, P.V.; Fernandes, A.R. Targeting Tumor Microenvironment for Cancer Therapy. Int. J. Mol. Sci. 2019, 20, 840. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  199. Freeman-Keller, M.; Kim, Y.; Cronin, H.; Richards, A.; Gibney, G.; Weber, J.S. Nivolumab in Resected and Unresectable Metastatic Melanoma: Characteristics of Immune-Related Adverse Events and Association with Outcomes. Clin. Cancer Res. 2016, 22, 886–894. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  200. Suresh, K.; Naidoo, J.; Lin, C.T.; Danoff, S. Immune Checkpoint Immunotherapy for Non-Small Cell Lung Cancer. Chest 2018, 154, 1416–1423. [Google Scholar] [CrossRef] [PubMed]
  201. Bauer, T.; Cho, B.C.; Heist, R.; Bazhenova, L.; Werner, T.; Goel, S.; Kim, D.-W.; Adkins, D.; Carvajal, R.D.; Alva, A.; et al. First-in-human phase 1/1b study to evaluate sitravatinib in patients with advanced solid tumors. Investig. New Drugs 2022, 40, 990–1000. [Google Scholar] [CrossRef]
Figure 1. Tumor microenvironment and immunotherapies. The TME is composed of various cell types, such as cancer cells, immune cells, and stromal cells, and cytokines as well as chemokines. These growth factors and cytokines are essential for communication between the TME and other cell types. Due to the importance of immune cells in the TME and in tumor progression, a number of immunotherapies have been developed against tumors in recent years. Immunotherapies that have been approved for use or that are undergoing clinical trials focus on TAMs, DCs, T cells, and NK cells as their main targets.
Figure 1. Tumor microenvironment and immunotherapies. The TME is composed of various cell types, such as cancer cells, immune cells, and stromal cells, and cytokines as well as chemokines. These growth factors and cytokines are essential for communication between the TME and other cell types. Due to the importance of immune cells in the TME and in tumor progression, a number of immunotherapies have been developed against tumors in recent years. Immunotherapies that have been approved for use or that are undergoing clinical trials focus on TAMs, DCs, T cells, and NK cells as their main targets.
Pharmaceutics 15 01788 g001
Figure 2. Two of the most common targeting pathways for ICIs (anti-CTLA-4 and anti-PD-1/PD-L1). PD-1 receptor on T cells leads to T cell exhaustion by binding to its ligand PD-L1 on tumor cells. Targeted antibodies that block PD-1 or PD-L1 prevent T cell exhaustion and maintain the ability of T cells to destroy tumors. T cells can be activated through two primary mechanisms. T cell receptors (TCRs) on antigen-presenting cells (APCs) present antigens with the major histocompatibility complex (MHC) and activate T lymphocytes. Additionally, the binding of CD28 on T cells and B7 on APCs can result in T-cell activation and antitumor activity. CTLA-4 antigen limits T cell-activation by competing for CD28 binding to B7 on the T cell surface, resulting in a decrease in T cell antitumor capacity. Therefore, the use of anti-CTLA-4 drugs can prevent the competitive behavior of CTLA-4 antibodies and support normal T cell-activation.
Figure 2. Two of the most common targeting pathways for ICIs (anti-CTLA-4 and anti-PD-1/PD-L1). PD-1 receptor on T cells leads to T cell exhaustion by binding to its ligand PD-L1 on tumor cells. Targeted antibodies that block PD-1 or PD-L1 prevent T cell exhaustion and maintain the ability of T cells to destroy tumors. T cells can be activated through two primary mechanisms. T cell receptors (TCRs) on antigen-presenting cells (APCs) present antigens with the major histocompatibility complex (MHC) and activate T lymphocytes. Additionally, the binding of CD28 on T cells and B7 on APCs can result in T-cell activation and antitumor activity. CTLA-4 antigen limits T cell-activation by competing for CD28 binding to B7 on the T cell surface, resulting in a decrease in T cell antitumor capacity. Therefore, the use of anti-CTLA-4 drugs can prevent the competitive behavior of CTLA-4 antibodies and support normal T cell-activation.
Pharmaceutics 15 01788 g002
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

Ye, W.; Li, M.; Luo, K. Therapies Targeting Immune Cells in Tumor Microenvironment for Non-Small Cell Lung Cancer. Pharmaceutics 2023, 15, 1788. https://doi.org/10.3390/pharmaceutics15071788

AMA Style

Ye W, Li M, Luo K. Therapies Targeting Immune Cells in Tumor Microenvironment for Non-Small Cell Lung Cancer. Pharmaceutics. 2023; 15(7):1788. https://doi.org/10.3390/pharmaceutics15071788

Chicago/Turabian Style

Ye, Wei, Meiye Li, and Kewang Luo. 2023. "Therapies Targeting Immune Cells in Tumor Microenvironment for Non-Small Cell Lung Cancer" Pharmaceutics 15, no. 7: 1788. https://doi.org/10.3390/pharmaceutics15071788

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