Next Article in Journal
Bottom-Up Strategy to Forecast the Drug Location and Release Kinetics in Antitumoral Electrospun Drug Delivery Systems
Next Article in Special Issue
Mechanisms Contributing to the Comorbidity of COPD and Lung Cancer
Previous Article in Journal
Neuroprotective Effects of Erinacine A on an Experimental Model of Traumatic Optic Neuropathy
Previous Article in Special Issue
B7-H3/CD276 Inhibitors: Is There Room for the Treatment of Metastatic Non-Small Cell Lung Cancer?
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

The Importance of the Immune System and Molecular Cell Signaling Pathways in the Pathogenesis and Progression of Lung Cancer

by
Jolanta Smok-Kalwat
1,
Paulina Mertowska
2,
Sebastian Mertowski
2,
Konrad Smolak
2,
Aleksandra Kozińska
3,
Filip Koszałka
3,
Wojciech Kwaśniewski
4,
Ewelina Grywalska
2,* and
Stanisław Góźdź
1,5
1
Department of Clinical Oncology, Holy Cross Cancer Centre, 3 Artwinskiego Street, 25-734 Kielce, Poland
2
Department of Experimental Immunology, Medical University of Lublin, 4a Chodzki Street, 20-093 Lublin, Poland
3
Student Research Group of Experimental Immunology, Medical University of Lublin, 4a Chodzki Street, 20-093 Lublin, Poland
4
Department of Gynecologic Oncology and Gynecology, Medical University of Lublin, 20-081 Lublin, Poland
5
Institute of Medical Science, Collegium Medicum, Jan Kochanowski University of Kielce, IX Wieków Kielc 19A, 25-317 Kielce, Poland
*
Author to whom correspondence should be addressed.
Int. J. Mol. Sci. 2023, 24(2), 1506; https://doi.org/10.3390/ijms24021506
Submission received: 30 November 2022 / Revised: 4 January 2023 / Accepted: 8 January 2023 / Published: 12 January 2023
(This article belongs to the Special Issue Molecular Mechanisms and Therapies of Lung Cancer and Lung Disease)

Abstract

:
Lung cancer is a disease that in recent years has become one of the greatest threats to modern society. Every year there are more and more new cases and the percentage of deaths caused by this type of cancer increases. Despite many studies, scientists are still looking for answers regarding the mechanisms of lung cancer development and progression, with particular emphasis on the role of the immune system. The aim of this literature review was to present the importance of disorders of the immune system and the accompanying changes at the level of cell signaling in the pathogenesis of lung cancer. The collected results showed that in the process of immunopathogenesis of almost all subtypes of lung cancer, changes in the tumor microenvironment, deregulation of immune checkpoints and abnormalities in cell signaling pathways are involved, which contribute to the multistage and multifaceted carcinogenesis of this type of cancer. We, therefore, suggest that in future studies, researchers should focus on a detailed analysis of tumor microenvironmental immune checkpoints, and to validate their validity, perform genetic polymorphism analyses in a wide range of patients and healthy individuals to determine the genetic susceptibility to lung cancer development. In addition, further research related to the analysis of the tumor microenvironment; immune system disorders, with a particular emphasis on immunological checkpoints and genetic differences may contribute to the development of new personalized therapies that improve the prognosis of patients.

1. Introduction

Lung cancer is one of the diseases with the highest mortality rate in the world. According to the World Health Organization, in 2020 the number of deaths caused by this cancer amounted to 1.80 million people, which accounted for 25% of the recorded deaths due to cancer [1,2]. The American Cancer Society lung cancer statistics for 2022 show that approximately 236,740 new cases of lung cancer were diagnosed in the United States alone (117,910 in men and 118,830 in women) and approximately 130,180 people died from lung cancer (68,820 men and 61,360 women) [3]. In addition to gender and age differences (diagnosed patients are >65 years of age), many environmental, genetic and lifestyle factors are involved in the development of lung cancer (Figure 1) [4,5,6,7,8,9,10,11]. However, more and more scientific studies highlight the extremely important role of genetic changes in the development and progression of this type of cancer [12,13,14,15].
Genetic aberrations and epigenetic changes occurring in the human body have a significant impact on proto-oncogenes (genes that stimulate cell growth, which may be transformed into oncogenes as a result of mutation) and suppressor genes (genes encoding proteins, whose task is to inhibit cell growth and differentiation and to maintain cell stability; mutations in these genes lead to uncontrolled cell proliferation), facilitating the development and migration of cancer cells. However, this is only one aspect of the process [16,17,18,19,20]. Under conditions of homeostasis, the immune system has an extremely important potential for monitoring, recognizing and destroying cancer cells. However, these cells have developed several mechanisms that allow them to escape the surveillance of the immune system [21,22,23,24]. Therefore, all deregulations in the human body related to the functioning of the immune system should be considered as the second potential process involved in the pathogenesis of cancer, including lung cancer [25].
Despite many studies on the risk factors, classification and pathogenesis of lung cancer, very little data are devoted to disorders of the immune system and signal transduction pathways, which, as a result of genetic changes, determine the development of cancer cells. The aim of this publication is to review the literature on the importance of disorders of the immune system and the accompanying molecular implications in the pathogenesis of lung cancer. We paid particular attention to the importance of cancer antigens and neoantigens, as well as the role of immune checkpoints and abnormalities in cell signaling pathways that contribute to the multifaceted and multistage carcinogenesis of lung cancer.

2. Results and Discussions

2.1. Molecular and Histological Classification of Lung Cancer

The data from the literature classified lung cancer histologically into two main types: neuroendocrine cell-derived small-cell lung cancer (SCLC), which accounts for 15–20% of all lung cancer patients, and epithelial cell-derived non-small-cell lung cancer (NSCLC), which accounts for 80–85% [19,26,27,28]. Neuroendocrine neoplasms (NENs) of the lung include two characteristic subtypes: neuroendocrine tumors (NETs), consisting of typical (TCs) and atypical (ACs) carcinoids; and full-fledged carcinoids (NECs), which include large-cell neuroendocrine carcinomas (LCNECs) and small cell carcinomas (SCLCs). Detailed studies resulted in the morphological and molecular characterization of these subtypes (Figure 2) [28,29].
In the case of NSCLC lung cancer, three main histological subtypes can be distinguished: adenocarcinoma (ADC), accounting for about 40% of all lung cases; squamous cell carcinoma (SCC), which is strongly associated with a smoking history and accounted for about 20% of all cases lung cancer; and large cell carcinoma (LCC). As indicated by histological studies, lung cancer is characterized by high heterogeneity, which is also reflected at the molecular level. Thanks to the development of genomic research, it was possible to determine the molecular patterns of each of the subtypes of lung cancer, which, according to research data, also differed depending on the histological subtype [33,34,35,36].
The TSG analysis for selected lung cancer subtypes based on the COSMIC Cancer Gene Census showed that there are certain genes in which mutations occur much more often. We can indicate both genes that are not specific to a specific subtype, such as EGFR or TP53, as well as those that are characteristic of a specific subtype of lung cancer (Table 1).
Furthermore, clinical studies have shown that molecularly defined lung cancer subgroups may be correlated with characteristics such as ethnicity [38,39,40], smoking history [41,42,43], treatment sensitivity [44,45,46] or prognosis [47].
Most of the genes indicated in the literature and databases encode proteins involved in the signal transduction process (kinases, transcription factors, receptors), which, as a result of mutations, introduce disturbances in the signal transduction pathway between cells.
Specific proteins could be used for differential diagnosis, facilitating the diagnosis of a specific type of lung cancer. Examples of proteins that have been tested and assessed for differential diagnosis include p63 in correlation with the TTF-1 transcription factor [48]. MUC5B was also correlated with the transcription factor TTF-1 [49]. The first protein was studied in the context of small-cell lung cancer compared to poorly differentiated non-keratinizing squamous cell carcinoma [48]. In these studies, the researchers demonstrated the utility of evaluating a panel of p63 and TTF-1 antibodies as useful markers in distinguishing SCLC from poorly differentiated non-keratinizing SCC on surgical, biopsy and cytology sections. The researchers based their conclusions on the positive expression of the p63 protein, in the negative correlation of the reaction for TTF-1, shown in subjects with differentiated non-keratinizing SCC, while the opposite results were obtained for SCLC. Similar conclusions were obtained in studies conducted by Veena et al., and in the summary of their research the researchers indicated that obtaining positive results for TTF1 and negative results for p63 can help in the diagnosis of adenocarcinoma, while samples that are positive for p63 and negative for TTF1 negative can be indicated and help in the diagnosis of squamous cell carcinoma [50]. In turn, another study conducted by Nagashio et al. indicated the usefulness of MUC5B protein determination in correlation with TTF-1, which may be useful for differential diagnosis between AC and SCC. In addition, it has been suggested that an assessment of the expression of the above factors could be a prognostic indicator for the prognosis of patients with pulmonary AC [49]. This means that any abnormalities of these genetic factors or disorders in the functioning of the pathway itself should be carefully analyzed in terms of their involvement in the development and progression of lung cancer.

2.2. Disorders of Cell Signaling Pathways as an Element of Lung Cancer Development and Progression

The development of new research techniques in the field of genomic and proteomic research has contributed to the increased interest of many researchers in the interactions between cancer cells, their unique microenvironment and intercellular communication pathways [51]. Analyses have been launched to demonstrate the occurrence of genetic aberrations and epigenetic changes that may determine an increased risk of lung cancer, but also affect the selection of therapeutic therapies and their therapeutic success. In recent years, several papers on this issue have appeared in the literature, which have allowed the classification of the observed disorders into three characteristic pathways, which, as the researchers indicated, can direct cells towards a malignant phenotype. These include signal transduction pathways that stimulate cell growth, pathways associated with tumor suppressor genes and pathways associated with cell evasion of apoptosis [52,53,54]. Detailed characteristics of these three groups of pathways with the most important genes/proteins involved in the development and progression of lung cancer are presented in Table 2.

2.3. Epigenetic Changes Involved in the Pathogenesis of Lung Cancer

All the cells of the human body have almost identical genetic information, yet the cells of different tissues are very different from each other. This remarkable diversity is due to the different use of the same genetic information through epigenetic mechanisms. These mechanisms allow the emergence of some kind of permanent gene expression patterns for a given differentiated cell, with a simultaneous short-term and reversible change in their expression. Research on the epigenome (i.e., a set of DNA and histone protein modifications) developed in recent years has led to the determination of a number of molecular mechanisms that are epigenetic modifications in the course of lung cancer [106,107,108]. Such mechanisms include the following:
(1)
A change in DNA methylation status within the CpG islands of tumor suppressor genes;
(2)
Covalent modifications of histone tails;
(3)
Regulation of genes by micro-RNAs (miRNAs).
The first mechanism can be considered in two ways: in chemical terms (the process in which the methyl group -CH3 is transferred between two molecules) and in biological terms (i.e., the effect of methylation on the functioning of DNA). In response to various external factors, the DNA of each person undergoes changes that are designed to adapt the functioning of the body to changing conditions [109]. Therefore, the correct course of this process plays a key role for the efficient functioning of cells and organs because it determines the expression and activity of individual genes. As a result of disorders of methylation pathways in the human body, there may be abnormalities related to immune reactions or the development of cancer (genetically determined or resulting from spontaneous mutations) [110,111]. Thus, methylation has become the subject of many intensive studies aimed at elucidating its detailed role in the pathogenesis of neoplastic diseases. According to the available literature, three types of abnormal methylation may occur in the development of this type of disease: global hypomethylation, hypermethylation of suppressor genes, and regulation of miRNA activity by methylation [112,113]. Hypomethylation is a process observed in the course of many diseases, especially those of a chronic nature, such as autoimmune diseases and cancer. According to some scientists, this process can reduce the stability of chromosomes and activate proto-oncogenes, and, in non-promoter regions of genes, it can weaken the stability of the genome by demethylating transposons. The phenomenon of global hypomethylation does not depend on local changes in DNA methylation in regulatory regions of genes. Genetic and cellular data support the idea that global DNA methylation is responsible for genome integrity and can also lead to chromosomal aberrations [114,115,116].
Hypermethylation, i.e., an increased degree of methylation, causes silencing of DNA repair genes and tumor suppressor genes (anti-oncogenes). The result of this process is that there is no limit to cell proliferation and there is a high probability that the abnormal cell will not be directed to the apoptotic pathway. In addition, the hypermethylation of DNA repair genes turns off genes that code for proteins responsible for repairing abnormal genes [117]. Since hypermethylation of promoter regions results in a loss of gene function, it has a major impact on cell functioning and can inactivate genes and key biological pathways [118]. In the course of neoplastic diseases, we can observe both the loss of methylation in non-coding regions of the genome, which in turn leads to disturbances in its stability, or the methylation of CpG islands in the tumors themselves, which results in a loss of expression of tumor suppressor genes. In lung cancer, studies have indicated that many genes have been silenced by methylation, including RARB (retinoic acid receptor beta), CDKN2A (cyclin dependent kinase inhibitor 2A), TIMP3 (TIMP metallopeptidase inhibitor 3), MGMT (methylguanine methyltransferase), DAPK (death associated protein kinase 1), CDH13 (cadherin 13) and CDH1 (cadherin 1) [119,120,121,122,123,124,125,126,127,128,129,130]. Researchers suggested that restoring the expression of silenced genes in the course of lung cancer may be a new therapeutic target.
The second mechanism of epigenetic changes involves modifications of histones and their implementing enzymes in response to internal and external stimuli [131]. Disturbances in their functioning can lead to many unfavorable changes, such as chromatin compaction, changes in nucleosome dynamics and abnormalities at the level of transcription. All changes contribute to the imbalance of gene expression, which in the case of the development of cancer, is observed as a gain or loss of function, overexpression, suppression by promoter hypermethylation, chromosome translocation or mutations of histone-modifying enzymes/complexes and even histone modification sites [132]. In lung cancer, scientists have observed a specific combination of histone markers resulting from epigenetic changes that include the deacetylation of H3 and H4 histones, loss of lysine 4 trimethylation of histone H3 and an increase in the trimethylation of H3K9 and H3K27. In 2007, Barlési et al. showed that the acetylation and trimethylation states of H2 and H3 can be a kind of prognostic marker in the course of NSCLC. They showed that the predictive value of epigenetic changes involved many histones, and of particular note were H2A (H2AK5ac) and H3 (H3K4me2, H3K9ac), which were higher in the early development of NSCLC [133].
The team of Van Den Broeck et al. showed in 2008 that there is increased H4K5/H4K8 acetylation as well as a loss of H4K20 trimethylation in NSCLC and pre-invasive bronchial dysplastic lesions. In addition, they found that the loss of H4K20 trimethylation was significantly associated with the early stage I ADC subpopulation, and their data indicated shortened survival [134]. Similar conclusions were also drawn by researchers from the team of Seligson et al. in 2009, who showed that the cellular levels of H3K4me2 and H3K18ac were relatively low, and the identified histone modification trends were independent predictors of prognosis in the course of ADC [135]. Studies presented in the literature suggest that reduced cellular levels of specific histone modifications may be a kind of predictive factor related to the prognosis of patients with lung cancer.
The third mechanism of epigenetic changes concerns the regulation of miRNAs. This is a family of small non-coding RNAs (21–25 nucleotides long) that can negatively affect the translation of messenger RNA (mRNA) and promote its degradation by base pairing with complementary mRNA target sites [135]. Thanks to the development of such a mechanism, miRNAs can change gene expression after the transcription process. The literature describes over 500 miRNAs found in humans, which can function both as oncogenes, but also as tumor suppressor genes [136]. As a result of intensive research in recent years, scientists have managed to classify some miRNAs involved in the pathogenesis of lung cancer into eight groups related to signaling pathways and cellular processes: sustaining proliferative signaling; evading growth suppressors; enabling replicative immortality; activating invasion and metastasis; inducing angiogenesis; deregulating circular energetics; resisting cell death; avoiding immune destruction and tumors promoting inflammation. Detailed data are presented in Table 3. The scientists indicated that the use of miRNAs in the future may allow the identification of histological subtypes of lung cancer, which will increase the diagnostic possibilities and their participation as molecular biomarkers in screening tests, as well as determine whether to distinguish primary from metastatic tumors [137]. In addition, researchers hope that miRNAs will play an important role in determining patient prognoses and tailoring personalized therapies for lung cancer in the future [138].

2.4. The Importance of the Immune System in the Pathogenesis of Lung Cancer

In the condition of immune homeostasis, monitoring, recognition and destruction of foreign antigens and cancer cells take place [299]. The latter group is capable of an uncontrolled proliferation and spread throughout the human body, as well as attacking healthy tissues or cells. Tumor development and progression is a multistage process and involves eight characteristic processes that are also observed in the development of lung cancer (Figure 3) [300].
Understanding the role of the immune system in the development and progression of many types of cancer has become one of the most intensive research objects of recent decades.
Based on this type of research, cancer immunotherapies are being developed that target the immune microenvironment of tumors. Some researchers have indicated that type 1 and type 2 immune responses are important in the progression of cancer. Type 1 immune responses are considered important in fighting cancer, and immunotherapies are aimed at increasing the activity of this type of immune response [301]. Such a conclusion seems correct because in NSCLC tumors Stankovic’s team identified major different subpopulations of T cells, and the second most common type of disease cell in NSCLC tumors was B cells [302].
Still, there are many cases of research in which the scientists have been able to discover only a small part or aspect of how the immune response works in fighting cancer cells.
That is why in order to better understand the interactions between cancer cells and the immune system, researchers looked at the contributions of components of the adaptive and innate immune responses, the involvement of tumor antigens, the importance of the tumor microenvironment and immune checkpoints [25,53].

2.4.1. Importance of Tumor Antigens and Neoantigens

Immune surveillance assumes that cancer cells appear in the human body more often than the clinical manifestation of cancer, but their development is controlled by appropriate immune mechanisms that allow their detection and destruction [303,304]. This process is carried out by both the innate and adaptive immune systems and is assisted by cancer-specific antigens (tumor markers) that can be recognized by the immune system. Tumor markers are specific substances, often proteins, that are produced by the cancer tissue itself or sometimes by the body in response to the appearance of cancer. The presence or elevated levels of tumor markers in the blood or urine can help detect certain types of cancer, including lung cancer [305]. One group of antigens is known as tumor-associated antigens (TAAs), which are antigens that are overexpressed in cancer cells but can also be found in normal tissues, while the other group is tumor-associated antigens (TSAs). Both groups of antigens have become the subject of many studies aimed at supporting the diagnostic process and therapy of patients with cancer. In the case of lung cancer, the majority of the literature has focused on the search for tumor antigens that may be specific biomarker molecules in the course of NSCLC.
Research conducted by the team of Gure et al. in 2005 aimed to determine the level of expression of nine tumor antigens (NY-ESO-1, LAGE-1, MAGE-A1, MAGE-A3, MAGE-A4, MAGE-A10, CT7/MAGE-C1, SSX2 and SSX4) in over 500 patients diagnosed with NSCLC. The researchers showed that the expression of cancer genes in the testis, both cumulative and single, showed a significant relationship with the clinical characteristics of patients, i.e., with male sex, smoking history, cancer stage or the presence of metastases. In addition, the scientists’ Cox regression analysis showed that the expression of NY-ESO-1 and MAGE-A3 was correlated with a poor patient prognosis [306]. Another study carried out by Grunwald et al. in 2006 showed that the expression of seven genes (MAGE-A3, NY-ESO-1, LAGE-1, BRDT, HOM-TES-85, TPX-1 and LDHC) was associated with the development of NSCLC. Researchers have shown that 81% of NSCLC patients tested expressed at least one of the genes tested and 50% of the sample at least two of the genes. Additionally, the researchers showed that all but one of the genes are regulated by genomic methylation, and not all of them are co-expressed [307]. In 2020, the team of Palat et al. conducted research in which 12 selected cancer antigens were determined (CEA, MAGE-A1, MAGE-A3, MAGE-A4, PRAME, hTERT, HER2, MUC1, Survivin, STEAP1, SOX2 and NY-ESO-1), and their involvement in the pathogenesis of lung cancer was studied, with a particular emphasis on NSCLC subtypes [308]. Based on the analyses carried out, the researchers showed a higher expression of TAA SCCs in relation to ACs. In addition, they showed that the patients’ T-cell response to stimulation was significantly lower in patients with SCCs than those with ACs. Researchers have suggested that there are specific differences in T-cell function between NSCLC subtypes [308]. Some researchers have pointed out that cancer proteins circulating in serum or plasma, released from cancer cells, are few and their sensitivity, especially in terms of diagnostics, is low, especially in the early stages of cancer. Therefore, in recent years, scientists have used the fact that TAs are capable of inducing an immune response and stimulating the formation of autoantibodies associated with them (TAAb). They are created not only before, but also during the formation of the cancer itself, which means that they can be potential biomarker molecules that will be involved in the diagnostic process [309]. The researchers pointed out that TAAbs have several significant advantages that outweigh their use over TAs: the response of TAAbs to TAs is often enhanced by immune responses, allowing them to be more easily detected; TAAbs are relatively stable in body fluids, unlike TAs, and, therefore, according to the researchers, they are more stable; and TAAbs are highly specific and easily detectable in small sample volumes [310,311,312].
More and more attention has been paid by scientists to the development of combinations of biomarker molecules found in the blood or sputum of lung cancer patients that could potentially contribute to the early detection of neoplastic changes. According to the data available from the literature, these molecules are usually associated in several combinations and classified into one of four groups: autoantibody-based marker combinations, metabolites, protein-based biomarker combinations and mixed panels of markers (Figure 4) [313,314,315,316,317,318,319,320,321,322,323,324,325,326,327,328,329,330,331,332,333,334,335,336].
Lung cancers carry a high mutational load (point mutations, misfolding, overexpression, abnormal glycosylation, shortening or abnormal degradation), which leads to the formation of unique peptides capable of inducing an immune response in the body, i.e., neoantigens. The formation of abnormal proteins as a result of their mutations can lead to their recognition by the host’s immune system, more specifically by the histocompatibility system (MHC) class I (MHC-I) or MHC class II. Neoantigens in the tumor cytosol are presented on MHC-I and recognized by CD8+ cytotoxic T cells, while neoantigens released into the tumor microenvironment can be taken up by circulating antigen-presenting cells [337,338]. In the literature, we can find studies indicating that neoantigens are specific to the patient and not to the tumor itself, which, according to the researchers, suggests that tumors avoid destruction due to the immune system. These properties make neoantigens increasingly recognized as key mediators of tumor-specific immune activation and they have been identified as potential targets for personalized cancer therapies (including cancer vaccines) [312,337,338,339]. Thanks to the development of molecular biology techniques, including next-generation sequencing, and the participation of bioinformatics, it is possible to use data to predict neoantigens, most often on the basis of their affinity to MHC-I. However, research in recent years has resulted in a much broader approach to the detection of neoantigens in lung cancer (especially NSCLC), taking into account both the stability of binding to MHC-I, as well as the level of gene expression or aggretopicity [340,341,342,343]. According to the literature, the median number of predicted neoantigens in NSCLC is quite variable. In 2016, Karasaki et al. conducted research aimed at identifying individual and specific somatic mutations occurring in NSCLC, which could be a new method of neoantigen-based immunotherapy. The researchers found from 13 to 659 neoantigens for ADC (median 46) and from 10 to 145 neoantigens for SCC (median 95.5) [344]. Other reports in the literature state that the number of neoantigens detected in NSCLC ranged from 63 to 214 neoantigens per tumor. Researchers have observed that increased numbers of neoantigens were noted among patients who had molecular signatures associated with cigarette smoking and increased expression of programmed death ligand 1 (PD-L1) [339,345,346].

2.4.2. Importance of Immune Checkpoints

Immune checkpoints are systems of protein receptors and ligands exposed on the surface of cells of the immune system (especially T lymphocytes) that modulate the body’s immune responses [347,348,349]. Many studies have shown that they are involved in altering the ability of the immune system to recognize malignant cells. In conditions of immune homeostasis, immune checkpoints provide self-tolerance by maintaining T lymphocyte activation, which causes inhibition of the immune response, but in the case of cancer, they are inhibited and the immune system is reactivated so that it is able to launch an attack on cancer cells [350,351]. In the literature, we found data on the detailed mechanisms of action of almost 20 significant modulators of immune checkpoints (Table 4), such as LAG-3, TIM3 and TIGIT, as well as the programmed cell death receptor 1 (PD-1) and a protein associated with cytotoxic T-4 lymphocytes (CTLA-4) (Figure 5), which have found their greatest application in clinical practice, including the treatment of lung cancer [350,351,352,353,354,355].

PD-1/PD-L1 Pathway

The PD-1 molecule together with its PDL-1/PD-L2 ligands as immune checkpoint molecules are responsible for suppressing the effector phase of activated T lymphocytes, reducing the inflammatory response and, consequently, autoimmunity [356]. These molecules can exist in the form of being bound to the cell membrane and soluble in serum and blood plasma [357]. Forms associated with the cell membrane occur, among others, in cells such as T and B lymphocytes, Treg lymphocytes, NK cells and APC cells [356]. In turn, the soluble molecule PD-L1 is detected in plasma as well as in body fluids, such as pleural effusion of lung cancer patients [357].
In a properly functioning body, after the elimination of pathogens or abnormal infected cells, the PD-1/PD-L pathway has a suppressive effect on T lymphocytes, causing their exhaustion and apoptosis, which prevents autoimmunity. Unfortunately, this natural immunosuppression is used by cancer cells to evade surveillance by the immune system [356].
Therefore, monoclonal antibodies directed against PD-1/PD-L1 are currently used as one of the forms of therapy and treatment of cancer, including lung cancer [352].
In clinical trials, the effectiveness of various models of therapy have been observed, in which preparations such as pembrolizumab, nivolumab, atezolizumab and durvalumab have been used [357].
In clinical trials in the first-line treatment of advanced NSCLC, nivolumab demonstrated an improvement in overall survival outcomes for advanced squamous NSCLC compared to docetaxel (a cytotoxic anticancer drug used in cancer chemotherapy). Similar observations were also obtained but with better results in relation to docetaxelem. In the case of Atezolizumab, its positive impact on patient outcomes was also assessed. The obtained results indicated a beneficial therapeutic path using PD-1/PD-L1 antibodies in patients with advanced NSCLC. In addition, such therapy has a more body-friendly safety profile compared to chemotherapy [358].

CTLA4 Pathway

The CTLA-4 molecule, a member of the immunoglobulin superfamily, was expressed primarily by activated T cells. It is an important surface protein and co-inhibitor, localized to activated CD4+/CD8+ T cells to reduce the activity of these cells by binding CD80/CD86/CD28 molecules [359,360].
One of the main tasks of CTLA-4 is to inhibit the function of CD28, a co-stimulatory receptor for T cells. Previous studies suggested that CTLA-4 expression increases upon activation of T cells, while CD28 appears mainly on the surface of naive T cells. Therefore, this molecule seems to have a key role in the regulation of activated T cells, as the literature reports that a lack of CTLA-4 causes unregulated T-cell proliferation [360].
In turn, the deletion or inhibition of T-cell-specific CTLA-4 reduces their ability to control both antitumor and autoimmune responses. Therefore, it is suggested that in the treatment of tumors with a high percentage of T cells that respond to tumor antigens, inhibition of CTLA-4 may be beneficial by restoring anti-tumor immunity by controlling overactive T cells [360].
Currently, as the first-line treatment for lung cancers, such as inoperable malignant pleural mesothelioma (MPM) or NSCLC (with PD-L1 expression in the tumor ≥1% and no EGFR/ALK aberration), treatment with nivolumab and ipilimumab (CTLA-4 inhibitor) is proposed. These formulations, when injected intravenously, have produced favorable treatment results for patients. At the same time, blocking the CTLA-4 molecule using ipilimumab may cause side effects such as colitis and enteritis [361]. Therefore, it is important to find optimal treatments with checkpoint inhibitors so that the benefits outweigh the negative effects. At the same time, current clinical data and basic research give hope for the development of an effective anticancer therapy based on checkpoint inhibitors, because their use can shift the immune balance towards promoting tumor killing and enhancing the immune attack on pathological cells [359].
Table 4. Immune checkpoints considered in lung cancer research.
Table 4. Immune checkpoints considered in lung cancer research.
PathwayMolecules Found on Cancer CellsMolecules Found on T Lymphocytes
NameGene and LocationAmino Acid Length and Molecular WeightFunctionNameGene and LocationAmino acid Length and Molecular WeightFunctionReferences
Inhibitory pathwayA2aRADORA2A,
22q11.23
412 aa;
44.707 kDa
  • Plays a regulatory role in the adaptive immune system;
  • Extracellular adenosine binds to the receptor, causing a G protein-coupled response, resulting in upregulation of TGF-β and PD-1;
  • Interactions with FOXP3 stimulate CD4+ T cells further suppressing the immune response.
No data[362,363,364,365]
VISTAC10orf54,
10q22.1
311 aa,
33.908 kDa
  • Belongs to the B7 family;
  • Its transcription is partially controlled by p53;
  • Produced in large amounts in tumor-infiltrating lymphocytes, such as myeloid-derived suppressor cells and regulatory T cells.
No data[364,366,367]
B7-H3CD276, 15q24.1 534 aa, 57.235 kDa
  • In benign tissues, it mainly plays an inhibitory role in adaptive immunity by inhibiting T-cell activation and proliferation;
  • In malignant tissues, it inhibits immune responses specific to the tumor antigen;
  • Has non-immune prone-forming functions, such as promotion of migration, invasion, angiogenesis and chemoresistance.
No data[368,369,370,371]
PD-L1CD274, 9p24.1 290 aa, 33.275 kDa
  • Plays a major role in suppressing the adaptive response of the immune system;
  • The binding of PD-L1 to the inhibitory checkpoint molecule PD-1 transmits an inhibitory signal based on interaction with phosphatases (SHP-1 or SHP-2) via the immunoreceptor tyrosine switching motif (ITSM).
  • It reduces proliferation of antigen-specific T cells in lymph nodes while reducing apoptosis in regulatory T cells.
PD-1PDCD1,
2q37.3
288 aa,
31.647 kDa
  • Protein on the surface of T and B lymphocytes that plays a role in regulating the immune response and promoting self-tolerance by suppressing the inflammatory activity of T lymphocytes;
  • It prevents autoimmune diseases, but it can also prevent the immune system from killing cancer cells.
[346,372,373,374,375,376,377,378]
PD-L2PDCD1LG2, 9p24.1273, 30,957
CD80CD80, 13q13.3-q21288 aa, 33.048 kDa
  • Can be expressed on antigen presenting cells (APCs) or tumor cells;
  • Interacts with both co-stimulatory (CD28) and coinhibitory receptors (cytotoxic T lymphocytes, antigen 4 (CTLA-4)) and regulates the immune response;
  • The low level of CD80 expression serves as a tumor escape mechanism due to the higher affinity and therefore preferential binding of CTLA-4 to CD80 compared to CD28;
  • In contrast, CD80 overexpression promotes T cell activation and tumor rejection, and CD80 deficiency also increases the immunogenicity of tumor cells.
CTLA-4CTLA-4, 2q33.2223 aa,
24.656 kDa
  • Expressed by activated T cells and transmits an inhibitory signal to T cells;
  • CTLA4 expression has been found in many cancers, including non-small-cell lung cancer (NSCLC) tissues and cells; its function in cancer cells is unknown.
[379,380,381,382,383,384,385,386]
CD86CD86, 3q21323 aa, 37.021 kDa
  • Acts in parallel with CD80 (B7-1) as a natural ligand for CD28 and CTLA-4;
  • Promotes T-cell proliferation, function and survival by interacting with CD28 as a co-stimulator, while in activated T cells it interacts with CTLA-4 and acts as a suppressor.
Galectin 9LGALS9, 17q11.2355 aa,
39.518 kDa
  • HAVCR2/galectin-9 interaction attenuated T-cell expansion and effector function in the tumor microenvironment and chronic infections;
  • Contributes to the formation of tumors through the transformation of cancer cells, regulation of the cell cycle, angiogenesis and cell adhesion.
TIM3HAVCR2,
5q33.3,
301 aa,
33.394 kDa
  • Belongs to the cell surface receptor proteins of the TIM family;
  • Transmembrane protein of T lymphocytes (CD4+ and CD8+ T cells), myeloid cells (monocytes, macrophages, DCs, mast cells, NK cells) or various cells in various types of cancers;
  • Mediates depletion of CD8+ T cells for proliferation and secretion of cytokines such as TNF-alpha, IFN-gamma and IL-2.
[387,388,389,390,391,392]
HVEMTNFRSF14, 1p36.32283 aa,
30.392 kDa
  • Belongs to the superfamily of TNF receptors;
  • Cytoplasmic region of this receptor binds to several members of the TNF receptor-associated factor (TRAF) family, which may mediate signal transduction pathways that activate the immune response;
  • HVEM overexpression in NSCLC patients was significant in patients with N2 lymph node metastases;
  • Shows a negative correlation with PD-L1 expression in NSCLC patients.
BTLABTLA, 3q13.2289 aa,
32.834 kDa
  • Expressed in lymph nodes, thymus and spleen;
  • Through ITIM, it recruits SHP-1 or SHP-2, which act as phosphatases and dephosphorylate tyrosine, which inhibits immune activation;
  • Has a Grb-2 recognition motif which, when recognized by the Grb-2 protein, promotes PI3K activation and additionally mediates cell proliferation and survival;
  • High BTLA expression may predict poor prognosis in NSCLC patients.
[393,394,395,396,397]
MHC IIHLA-DP, -DQ and –DR, chromosome 6-
  • A complex of proteins responsible for the presentation of antigens to T lymphocytes;
  • Correlates with the prognosis of patients with neoplastic diseases and is important in the selection of patients for immunotherapy.
LAG3LAG3, 12p13.31525 aa,57.449 kDa
  • Expressed on activated T and B cells, NK cells, and plasmacytoid dendritic cells;
  • Binds to MHC class II with greater affinity than CD4.
  • Negatively regulates cell proliferation, activation and homeostasis of T cells in a similar way to CTLA-4 and PD-1.
[398,399,400,401,402]
Stimulatory pathwayOX40LTNFSF4, 1q25.1183 aa,
21.050 kDa
  • Expressed on DC2, macrophages and activated B cells;
  • OX40-OX40L ligation is a source of survival signal for T cells and enables the development of memory T cells;
  • Signaling by these two molecules also polarizes towards a Th2 immune response, even in an environment with low levels of the cytokine IL-4.
OX40TNFRSF4, 1p36.33277 aa,
29.341 kDa
  • Present on the surface of activated T cells (mainly CD4+ T cells), but also on NK cells, NKT cells and neutrophils;
  • OX40 plays a key role in NSCLC and SCLC.
[403,404,405,406]
CD40TNFRSF5, 20q12-q13.2117 aa,13.158 kDa
  • Protein found on the surface of antigen-presenting cells, necessary for their activation and providing a co-stimulatory signal to T cells expressing the CD154 molecule;
  • Binding of CD154 to CD40 triggers signaling pathways based on TRAF proteins.
CD40LCD40LG, Xq26261 aa,
29.274 kDa
  • Protein found primarily on T lymphocytes, belonging to the TNF superfamily.
  • Its main function is to bind the CD40 molecule on antigen-presenting cells and deliver the co-stimulatory signal required for lymphocyte activation.
[407,408,409,410]
B7RP1ICOSG, 21q22.3302 aa,
33.349 kDa
  • Belongs to the B7 family of co-stimulatory ligands;
  • Shares 19–20% sequence identity with CD80 and CD86;
  • Causes activation and proliferation of T cells;
  • The ICOS/ICOSLG axis promotes antitumor T-cell responses (when activated in Th1 and other Teffs) or protumor responses when triggered in Tregs.
ICOSICOS, 2q33.2199 aa,
22.625 kDa
  • Costimulatory molecule of the CD28 superfamily that is expressed on activated T cells.
[411,412,413,414,415]
CD70CD70, 19p13.3193 aa,
21.118 kDa
  • Belongs to the tumor necrosis factor (TNF) family of ligands;
  • Surface antigen on activated but not resting T and B lymphocytes;
  • Responsible for inducing the proliferation of co-stimulated T cells, increasing the production of cytolytic T cells and contributes to the activation of T cells.
CD27CD27,
12p13.31
260 aa,
29.137 kDa
  • Necessary for the production and long-term maintenance of T-lymphocyte immunity;
  • Its binding to CD70 activates the signaling cascade leading to differentiation and clonal expansion of T cells;
  • Results in improved survival and memory of cytotoxic T cells and increased production of some cytokines;
  • Transmits signals that lead to the activation of NF-κB and MAPK8 / JNK
[416,417,418,419,420,421]
GITRLTNFSF18, 1q25.1177 aa,
20.308 kDa
  • Regulates T cell responses;
  • May act as a co-stimulator and lower the threshold for T cell activation and T cell proliferation
  • Important for the interaction between activated T cells and endothelial cells;
  • Mediates the activation of NF-kappa-B.
GITRTNFRSF18, 1p36.33241 aa,
26.000 kDa
  • Constitutively expressed on CD25+ CD4+ regulatory T cells and is upregulated in all subsets of T cells upon activation;
  • Signaling may promote anticancer and anti-infective immune responses, but may also be a driver of autoimmune diseases.
[422,423,424,425]
4-1 BBLTNFSF9, 19p13.3254 aa, 26.625 kDa
  • Interaction between 4-1BB and 4-1BBL provides co-stimulating signals for various T cells that can be used to discover cancer immunotherapy.
4-1 BBTNFRSF9, 1p36.23255 aa,
27.899 kDa
  • Expressed by activated T cells of both CD4+ and CD8+ lines;
  • Increased T cell proliferation, IL-2 secretion, survival and cytolytic activity.
[426,427,428,429,430]
CD155PVR, 19q13.31417 aa,
45.303 k Da
  • Type I transmembrane glycoprotein of the immunoglobulin superfamily;
  • Commonly known as the poliovirus receptor (PVR);
  • It mediates NK cell adhesion and triggers NK cell effector functions
TIGITTIGIT, 3q13.31244 aa,
26.319 kDa
  • Binding to CD155 results in increased secretion of IL10 and decreased secretion of IL12B and inhibition of T cell activation by promoting the production of mature immunoregulatory dendritic cells
[431,432,433,434]
Abbreviations: A2aR—adenosine A2A receptor; VISTA—V-domain Ig suppressor of T-cell activation; B7-H3—B7 homolog 3 protein; PD-1—programmed death receptor-1; PD-L1—programmed death-ligand 1; PD-L2—programmed death-ligand 2; CD80—cluster of differentiation 80; CD86—cluster of differentiation 86; TIM-3—T-cell immunoglobulin mucin-3; HVEM—herpes virus entry mediator; BTLA—B and T lymphocyte attenuator; MHC—major histocompatibility complex; LAG3—lymphocyte activating 3; OX40L—OX40 ligand; CD40—cluster of differentiation 40; CD40L—cluster of differentiation 40 ligand; ICOSLG—inducible T-cell costimulator ligand; ICOS (B7RP-1)—inducible T-Cell costimulator; CD70—cluster of differentiation 70; CD27—cluster of differentiation 27; GITR—glucocorticoid-induced tumor necrosis factor receptor-related protein; GITRL—glucocorticoid-induced tumor necrosis factor ligand-related protein; CD155—cluster of differentiation 155; TIGIT—T-cell immunoreceptor with Ig and ITIM domains.

3. Materials and Methods

3.1. Search Strategy

This study was performed on the basis of a meta-analysis of observational studies in accordance with the epidemiological guidelines and the PRISMA statement. To maximize sensitivity, a broad search strategy was driven using the international databases Web of Science, Scopus and PubMed/MEDLINE. The search strategy covered publications from 2002–2022 and used the following keywords: “immune system”; “immune response”; “genetic association”; tumor microenvironment”; “signal transduction”, “immune checkpoint”. Only articles in English were included in the analysis. After removing duplicates, the abstracts of the publications were reviewed by two independent researchers in accordance with the PRISMA abstract list. All disputes were resolved by a third investigator. The detailed search strategy is shown in Figure 6.

3.2. Assessment of (Quality) Bias Risk and Data Synthesis Strategy

The remaining articles were then reviewed according to the PRISMA statement. They were then systematically reviewed and assessed against the inclusion and exclusion criteria. All steps were carried out by two independent investigators, and disputes were resolved through discussion with the third investigator.

4. Conclusions

The aim of our literature review was to present the importance of immune system disorders and the accompanying changes at the level of cell signaling in the pathogenesis of lung cancer. The information presented by us indicates one of the causes of lung cancer development, which are changes in the tumor microenvironment. Moreover, in our review, we indicated the relationship between deregulation of the immune system and disturbances in cell signaling pathways that contribute to the multistage and multifaceted carcinogenesis of this type of cancer. We hope that the data presented by us will attract the attention of researchers and clinicians and will allow the development of more effective therapies. Despite the numerous scientific studies available in the literature on the etiopathogenesis of various types of lung cancer, it seems important for scientists to undertake an interdisciplinary and multifaceted approach, combining molecular, immunohistochemical and histological methods in order to expand the existing knowledge, which will contribute to faster and more effective diagnosis in the early stages of the disease.

Author Contributions

Conceptualization, J.S.-K., P.M., S.M., E.G. and S.G.; methodology, P.M., S.M., K.S., A.K., F.K., W.K. and J.S.-K.; software, P.M., S.M., A.K., F.K. and W.K.; validation, P.M, S.M., A.K., F.K. and K.S.; formal analysis, P.M. and S.M.; investigation, J.S.-K., P.M., S.M. and K.S.; resources, J.S.-K., P.M., S.M. and K.S.; data curation, P.M. and S.M.; writing—original draft preparation, P.M., S.M., K.S., A.K. and F.K.; writing—review and editing, J.S.-K., W.K., E.G. and S.G.; visualization, P.M. and S.M.; supervision, E.G. and S.G.; project administration, P.M. and S.M.; funding acquisition, P.M., E.G. and S.G. All authors have read and agreed to the published version of the manuscript.

Funding

This research was funded by the Medical University of Lublin under grant numbers PBsd166, DS640 and GI/1.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. 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] [PubMed] [Green Version]
  2. De Groot, P.M.; Wu, C.C.; Carter, B.W.; Munden, R.F. The Epidemiology of Lung Cancer. Transl. Lung Cancer Res. 2018, 7, 220–233. [Google Scholar] [CrossRef] [PubMed]
  3. Rahal, Z.; El Nemr, S.; Sinjab, A.; Chami, H.; Tfayli, A.; Kadara, H. Smoking and Lung Cancer: A Geo-Regional Perspective. Front Oncol. 2017, 7, 194. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  4. Shankar, A.; Dubey, A.; Saini, D.; Singh, M.; Prasad, C.P.; Roy, S.; Bharati, S.J.; Rinki, M.; Singh, N.; Seth, T.; et al. Environmental and Occupational Determinants of Lung Cancer. Transl. Lung Cancer Res. 2019, 8, S31–S49. [Google Scholar] [CrossRef]
  5. Tseng, C.-H.; Tsuang, B.-J.; Chiang, C.-J.; Ku, K.-C.; Tseng, J.-S.; Yang, T.-Y.; Hsu, K.-H.; Chen, K.-C.; Yu, S.-L.; Lee, W.-C.; et al. The Relationship Between Air Pollution and Lung Cancer in Nonsmokers in Taiwan. J. Thorac. Oncol. 2019, 14, 784–792. [Google Scholar] [CrossRef]
  6. Cancer. Available online: https://www.who.int/news-room/fact-sheets/detail/cancer (accessed on 20 November 2022).
  7. Lung Cancer Statistics | World Cancer Research Fund International; WCRF International: Londin, UK, 2022.
  8. Lung Cancer Statistics | How Common Is Lung Cancer? Available online: https://www.cancer.org/cancer/lung-cancer/about/key-statistics.html (accessed on 21 November 2022).
  9. Benusiglio, P.R.; Fallet, V.; Sanchis-Borja, M.; Coulet, F.; Cadranel, J. Lung Cancer Is Also a Hereditary Disease. Eur. Respir. Rev. 2021, 30, 210045. [Google Scholar] [CrossRef]
  10. Chen, X.; Xu, B.; Li, Q.; Xu, X.; Li, X.; You, X.; Yu, Z. Genetic Profile of Non-small Cell Lung Cancer (NSCLC): A Hospital-based Survey in Jinhua. Mol. Genet. Genomic. Med. 2020, 8, e1398. [Google Scholar] [CrossRef]
  11. Cline, M.J. The Role of Proto-Oncogenes in Human Cancer: Implications for Diagnosis and Treatment. Int. J. Radiat Oncol. Biol. Phys. 1987, 13, 1297–1301. [Google Scholar] [CrossRef]
  12. El-Telbany, A.; Ma, P.C. Cancer Genes in Lung Cancer. Genes Cancer 2012, 3, 467–480. [Google Scholar] [CrossRef]
  13. Kanwal, M.; Ding, X.-J.; Cao, Y. Familial Risk for Lung Cancer. Oncol. Lett. 2017, 13, 535–542. [Google Scholar] [CrossRef]
  14. Oxnard, G.R.; Binder, A.; Jänne, P.A. New Targetable Oncogenes in Non–Small-Cell Lung Cancer. J. Clin. Oncol. 2013, 31, 1097–1104. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  15. Padmavathi, P.; Raghu, P.S.; Reddy, V.D.; Bulle, S.; Marthadu, S.B.; Maturu, P.; Varadacharyulu, N.C. Chronic Cigarette Smoking-Induced Oxidative/Nitrosative Stress in Human Erythrocytes and Platelets. Mol. Cell Toxicol. 2018, 14, 27–34. [Google Scholar] [CrossRef]
  16. Robinson, L.A.; Jaing, C.J.; Pierce Campbell, C.; Magliocco, A.; Xiong, Y.; Magliocco, G.; Thissen, J.B.; Antonia, S. Molecular Evidence of Viral DNA in Non-Small Cell Lung Cancer and Non-Neoplastic Lung. Br. J. Cancer 2016, 115, 497–504. [Google Scholar] [CrossRef] [PubMed]
  17. Xiong, W.-M.; Xu, Q.-P.; Li, X.; Xiao, R.-D.; Cai, L.; He, F. The Association between Human Papillomavirus Infection and Lung Cancer: A System Review and Meta-Analysis. Oncotarget 2017, 8, 96419–96432. [Google Scholar] [CrossRef] [Green Version]
  18. Barros-Filho, M.C.; Guisier, F.; Rock, L.D.; Becker-Santos, D.D.; Sage, A.P.; Marshall, E.A.; Lam, W.L.; Barros-Filho, M.C.; Guisier, F.; Rock, L.D.; et al. Tumour Suppressor Genes with Oncogenic Roles in Lung Cancer; IntechOpen: London, UK, 2019; ISBN 978-1-78984-427-6. [Google Scholar]
  19. Domagala-Kulawik, J. The Role of the Immune System in Non-Small Cell Lung Carcinoma and Potential for Therapeutic Intervention. Transl. Lung Cancer Res. 2015, 4, 177–190. [Google Scholar] [CrossRef]
  20. Domagala-Kulawik, J.; Osinska, I.; Hoser, G. Mechanisms of Immune Response Regulation in Lung Cancer. Transl. Lung Cancer Res. 2014, 3, 15–22. [Google Scholar] [CrossRef]
  21. Domagala-Kulawik, J.; Raniszewska, A. How to Evaluate the Immune Status of Lung Cancer Patients before Immunotherapy. Breathe 2017, 13, 291–296. [Google Scholar] [CrossRef]
  22. Kuramochi, M.; Fukuhara, H.; Nobukuni, T.; Kanbe, T.; Maruyama, T.; Ghosh, H.P.; Pletcher, M.; Isomura, M.; Onizuka, M.; Kitamura, T.; et al. TSLC1 Is a Tumor-Suppressor Gene in Human Non-Small-Cell Lung Cancer. Nat. Genet. 2001, 27, 427–430. [Google Scholar] [CrossRef] [Green Version]
  23. Lim, R.J.; Liu, B.; Krysan, K.; Dubinett, S.M. Lung Cancer and Immunity Markers. Cancer Epidemiol. Biomark. Prev. 2020, 29, 2423–2430. [Google Scholar] [CrossRef]
  24. Pikor, L.A.; Ramnarine, V.R.; Lam, S.; Lam, W.L. Genetic Alterations Defining NSCLC Subtypes and Their Therapeutic Implications. Lung Cancer 2013, 82, 179–189. [Google Scholar] [CrossRef]
  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] [PubMed] [Green Version]
  26. Zhang, C.; Jiang, M.; Zhou, N.; Hou, H.; Li, T.; Yu, H.; Tan, Y.-D.; Zhang, X. Use Tumor Suppressor Genes as Biomarkers for Diagnosis of Non-Small Cell Lung Cancer. Sci. Rep. 2021, 11, 3596. [Google Scholar] [CrossRef] [PubMed]
  27. Chen, Z.; Fillmore, C.M.; Hammerman, P.S.; Kim, C.F.; Wong, K.-K. Non-Small-Cell Lung Cancers: A Heterogeneous Set of Diseases. Nat. Rev. Cancer 2014, 14, 535–546. [Google Scholar] [CrossRef] [PubMed]
  28. Fisseler-Eckhoff, A.; Demes, M. Neuroendocrine Tumors of the Lung. Cancers 2012, 4, 777–798. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  29. Niemira, M.; Collin, F.; Szalkowska, A.; Bielska, A.; Chwialkowska, K.; Reszec, J.; Niklinski, J.; Kwasniewski, M.; Kretowski, A. Molecular Signature of Subtypes of Non-Small-Cell Lung Cancer by Large-Scale Transcriptional Profiling: Identification of Key Modules and Genes by Weighted Gene Co-Expression Network Analysis (WGCNA). Cancers 2019, 12, 37. [Google Scholar] [CrossRef] [Green Version]
  30. Rodriguez-Canales, J.; Parra-Cuentas, E.; Wistuba, I.I. Diagnosis and Molecular Classification of Lung Cancer. In Lung Cancer: Treatment and Research; Reckamp, K.L., Ed.; Cancer Treatment and Research; Springer International Publishing: Cham, Switzerland, 2016; pp. 25–46. ISBN 978-3-319-40389-2. [Google Scholar]
  31. Savu, C.; Melinte, A.; Diaconu, C.; Stiru, O.; Gherghiceanu, F.; Tudorica Ștefan, D.O.; Dumitrașcu, O.C.; Bratu, A.; Balescu, I.; Bacalbasa, N. Lung Neuroendocrine Tumors: A Systematic Literature Review (Review). Exp. Ther. Med. 2022, 23, 176. [Google Scholar] [CrossRef]
  32. West, L.; Vidwans, S.J.; Campbell, N.P.; Shrager, J.; Simon, G.R.; Bueno, R.; Dennis, P.A.; Otterson, G.A.; Salgia, R. A Novel Classification of Lung Cancer into Molecular Subtypes. PLoS ONE 2012, 7, e31906. [Google Scholar] [CrossRef]
  33. Contributors, W.E. The Types of Lung Cancer. Available online: https://www.webmd.com/lung-cancer/lung-cancer-types (accessed on 21 November 2022).
  34. Cosmic Cancer Gene Census. Available online: http://cancer.sanger.ac.uk/census (accessed on 22 November 2022).
  35. Inamura, K. Lung Cancer: Understanding Its Molecular Pathology and the 2015 WHO Classification. Front Oncol. 2017, 7, 193. [Google Scholar] [CrossRef] [Green Version]
  36. Joshi, A.; Mishra, R.; Desai, S.; Chandrani, P.; Kore, H.; Sunder, R.; Hait, S.; Iyer, P.; Trivedi, V.; Choughule, A.; et al. Molecular Characterization of Lung Squamous Cell Carcinoma Tumors Reveals Therapeutically Relevant Alterations. Oncotarget 2021, 12, 578–588. [Google Scholar] [CrossRef]
  37. Nicholson, A.G.; Tsao, M.S.; Beasley, M.B.; Borczuk, A.C.; Brambilla, E.; Cooper, W.A.; Dacic, S.; Jain, D.; Kerr, K.M.; Lantuejoul, S.; et al. The 2021 WHO Classification of Lung Tumors: Impact of Advances Since 2015. J. Thorac. Oncol. 2022, 17, 362–387. [Google Scholar] [CrossRef]
  38. Schabath, M.B.; Cress, W.D.; Muñoz-Antonia, T. Racial and Ethnic Differences in the Epidemiology of Lung Cancer and the Lung Cancer Genome. Cancer Control. 2016, 23, 338–346. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  39. Soh, J.; Toyooka, S.; Matsuo, K.; Yamamoto, H.; Wistuba, I.I.; Lam, S.; Fong, K.M.; Gazdar, A.F.; Miyoshi, S. Ethnicity Affects EGFR and KRAS Gene Alterations of Lung Adenocarcinoma. Oncol. Lett. 2015, 10, 1775–1782. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  40. Zheng, M. Classification and Pathology of Lung Cancer. Surg. Oncol. Clin. North Am. 2016, 25, 447–468. [Google Scholar] [CrossRef] [PubMed]
  41. Furrukh, M. Tobacco Smoking and Lung Cancer. Sultan Qaboos Univ. Med. J. 2013, 13, 345–358. [Google Scholar] [CrossRef] [Green Version]
  42. Li, Y.; Xiao, X.; Han, Y.; Gorlova, O.; Qian, D.; Leighl, N.; Johansen, J.S.; Barnett, M.; Chen, C.; Goodman, G.; et al. Genome-Wide Interaction Study of Smoking Behavior and Non-Small Cell Lung Cancer Risk in Caucasian Population. Carcinogenesis 2018, 39, 336–346. [Google Scholar] [CrossRef] [Green Version]
  43. Massion, P.P.; Carbone, D.P. The Molecular Basis of Lung Cancer: Molecular Abnormalities and Therapeutic Implications. Respir. Res. 2003, 4, 12. [Google Scholar] [CrossRef] [Green Version]
  44. Qi, J.; Zhang, J.; Liu, N.; Zhao, L.; Xu, B. Prognostic Implications of Molecular Subtypes in Primary Small Cell Lung Cancer and Their Correlation With Cancer Immunity. Front. Oncol. 2022, 12, 424. [Google Scholar] [CrossRef]
  45. Ray, M.R.; Jablons, D.; He, B. Lung Cancer Therapeutics That Target Signaling Pathways: An Update. Expert Rev. Respir. Med. 2010, 4, 631–645. [Google Scholar] [CrossRef] [Green Version]
  46. Schwendenwein, A.; Megyesfalvi, Z.; Barany, N.; Valko, Z.; Bugyik, E.; Lang, C.; Ferencz, B.; Paku, S.; Lantos, A.; Fillinger, J.; et al. Molecular Profiles of Small Cell Lung Cancer Subtypes: Therapeutic Implications. Mol. Ther. Oncolytics 2021, 20, 470–483. [Google Scholar] [CrossRef]
  47. Shtivelman, E.; Hensing, T.; Simon, G.R.; Dennis, P.A.; Otterson, G.A.; Bueno, R.; Salgia, R. Molecular Pathways and Therapeutic Targets in Lung Cancer. Oncotarget 2014, 5, 1392–1433. [Google Scholar] [CrossRef]
  48. Gurguş, D.; Grigoraş, M.L.; Motoc, A.G.M.; Zamfir, C.L.; Cornianu, M.; Faur, C.I.; Pop, D.L.; Folescu, R. Clinical Relevance and Accuracy of P63 and TTF-1 for Better Approach of Small Cell Lung Carcinoma versus Poorly Differentiated Nonkeratinizing Squamous Cell Carcinoma. Rom. J. Morphol. Embryol. 2019, 60, 139–143. [Google Scholar] [PubMed]
  49. Nagashio, R.; Ueda, J.; Ryuge, S.; Nakashima, H.; Jiang, S.-X.; Kobayashi, M.; Yanagita, K.; Katono, K.; Satoh, Y.; Masuda, N.; et al. Diagnostic and Prognostic Significances of MUC5B and TTF-1 Expressions in Resected Non-Small Cell Lung Cancer. Sci. Rep. 2015, 5, 8649. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  50. Veena, V.S.; Saritha, V.N.; George, P.S.; Rajan, K.; Jayasree, K.; Sujathan, K. Immunoexpression of Ttf1 and P63 Differentiates Lung Adenocarcinomas in Sputum Samples. J. Cytol. 2021, 38, 151. [Google Scholar] [CrossRef] [PubMed]
  51. Zito Marino, F.; Bianco, R.; Accardo, M.; Ronchi, A.; Cozzolino, I.; Morgillo, F.; Rossi, G.; Franco, R. Molecular Heterogeneity in Lung Cancer: From Mechanisms of Origin to Clinical Implications. Int. J. Med. Sci. 2019, 16, 981–989. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  52. Abe, Y.; Tanaka, N. The Hedgehog Signaling Networks in Lung Cancer: The Mechanisms and Roles in Tumor Progression and Implications for Cancer Therapy. BioMed Res. Int. 2016, 2016, e7969286. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  53. Brambilla, E.; Gazdar, A. Pathogenesis of Lung Cancer Signaling Pathways: Roadmap for Therapies. Eur. Respir. J. 2009, 33, 1485–1497. [Google Scholar] [CrossRef] [PubMed]
  54. Chuang, J.C.; Stehr, H.; Liang, Y.; Das, M.; Huang, J.; Diehn, M.; Wakelee, H.A.; Neal, J.W. ERBB2-Mutated Metastatic Non-Small Cell Lung Cancer: Response and Resistance to Targeted Therapies. J. Thorac. Oncol. 2017, 12, 833–842. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  55. Hirsch, F.R.; Varella-Garcia, M.; Bunn, P.A.; Di Maria, M.V.; Veve, R.; Bremnes, R.M.; Barón, A.E.; Zeng, C.; Franklin, W.A. Epidermal Growth Factor Receptor in Non–Small-Cell Lung Carcinomas: Correlation Between Gene Copy Number and Protein Expression and Impact on Prognosis. JCO 2003, 21, 3798–3807. [Google Scholar] [CrossRef]
  56. Reiter, J.L.; Maihle, N.J. Characterization and Expression of Novel 60-KDa and 110-KDa EGFR Isoforms in Human Placenta. Ann. New York Acad. Sci. 2003, 995, 39–47. [Google Scholar] [CrossRef]
  57. Roviello, G.; D’Angelo, A.; Sirico, M.; Pittacolo, M.; Conter, F.U.; Sobhani, N. Advances in Anti-BRAF Therapies for Lung Cancer. Investig. New Drugs 2021, 39, 879–890. [Google Scholar] [CrossRef]
  58. Rowinsky, E.K. The ErbB Family: Targets for Therapeutic Development Against Cancer and Therapeutic Strategies Using Monoclonal Antibodies and Tyrosine Kinase Inhibitors. Annu. Rev. Med. 2004, 55, 433–457. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  59. Sharma, S.V.; Bell, D.W.; Settleman, J.; Haber, D.A. Epidermal Growth Factor Receptor Mutations in Lung Cancer. Nat. Rev. Cancer 2007, 7, 169–181. [Google Scholar] [CrossRef] [PubMed]
  60. Singh, V.; Feldman, R.; Sukari, A.; Kim, C.; Mamdani, H.; Spira, A.I.; Bepler, G.; Kim, E.S.; Raez, L.E.; Pai, S.G.; et al. Characterization of ERBB2 Alterations in Non-Small Cell Lung Cancer. JCO 2020, 38, e21553. [Google Scholar] [CrossRef]
  61. Thommen, D.S.; Schreiner, J.; Müller, P.; Herzig, P.; Roller, A.; Belousov, A.; Umana, P.; Pisa, P.; Klein, C.; Bacac, M.; et al. Progression of Lung Cancer Is Associated with Increased Dysfunction of T Cells Defined by Coexpression of Multiple Inhibitory Receptors. Cancer Immunol. Res. 2015, 3, 1344–1355. [Google Scholar] [CrossRef] [Green Version]
  62. Yan, N.; Guo, S.; Zhang, H.; Zhang, Z.; Shen, S.; Li, X. BRAF-Mutated Non-Small Cell Lung Cancer: Current Treatment Status and Future Perspective. Front Oncol. 2022, 12, 863043. [Google Scholar] [CrossRef]
  63. BRAF-Serine/Threonine-Protein Kinase B-Raf-Homo Sapiens (Human) | UniProtKB | UniProt. Available online: https://www.uniprot.org/uniprotkb/P15056/entry (accessed on 22 November 2022).
  64. EGFR-Epidermal Growth Factor Receptor-Homo Sapiens (Human) | UniProtKB | UniProt. Available online: https://www.uniprot.org/uniprotkb/P00533/entry (accessed on 22 November 2022).
  65. ERBB2-Receptor Tyrosine-Protein Kinase ErbB-2-Homo Sapiens (Human) | UniProtKB | UniProt. Available online: https://www.uniprot.org/uniprotkb/P04626/entry (accessed on 22 November 2022).
  66. Cao, L.-H.; Qiao, J.-Y.; Huang, H.-Y.; Fang, X.-Y.; Zhang, R.; Miao, M.-S.; Li, X.-M. PI3K–AKT Signaling Activation and Icariin: The Potential Effects on the Perimenopausal Depression-Like Rat Model. Molecules 2019, 24, 3700. [Google Scholar] [CrossRef] [Green Version]
  67. Cheng, K.T.; Chakrabart, A.; Aruva, M.R.; Thakur, M.L.; Wickstrom, E. 64Cu-N,N’-Bis(S-Benzoyl-Thioglycoloyl)Diaminopropanoate-KRAS-PNA-d(Cys-Ser-Lys-Cys). In Molecular Imaging and Contrast Agent Database (MICAD); National Center for Biotechnology Information (US): Bethesda, MD, USA, 2004. [Google Scholar]
  68. Du, X.; Shao, Y.; Qin, H.; Tai, Y.; Gao, H. ALK-rearrangement in Non-small-cell Lung Cancer (NSCLC). Thorac. Cancer 2018, 9, 423–430. [Google Scholar] [CrossRef]
  69. Engelman, J.A.; Luo, J.; Cantley, L.C. The Evolution of Phosphatidylinositol 3-Kinases as Regulators of Growth and Metabolism. Nat. Rev. Genet 2006, 7, 606–619. [Google Scholar] [CrossRef]
  70. Maeda, Y.; Davé, V.; Whitsett, J.A. Transcriptional Control of Lung Morphogenesis. Physiol. Rev. 2007, 87, 219–244. [Google Scholar] [CrossRef]
  71. Reck, M.; Carbone, D.P.; Garassino, M.; Barlesi, F. Targeting KRAS in Non-Small-Cell Lung Cancer: Recent Progress and New Approaches. Ann. Oncol. 2021, 32, 1101–1110. [Google Scholar] [CrossRef]
  72. Tanaka, H.; Yanagisawa, K.; Shinjo, K.; Taguchi, A.; Maeno, K.; Tomida, S.; Shimada, Y.; Osada, H.; Kosaka, T.; Matsubara, H.; et al. Lineage-Specific Dependency of Lung Adenocarcinomas on the Lung Development Regulator TTF-1. Cancer Res. 2007, 67, 6007–6011. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  73. Wu, F.; Wang, L.; Zhou, C. Lung Cancer in China: Current and Prospect. Curr. Opin. Oncol. 2021, 33, 40–46. [Google Scholar] [CrossRef] [PubMed]
  74. ALK-ALK Tyrosine Kinase Receptor-Homo Sapiens (Human) | UniProtKB | UniProt. Available online: https://www.uniprot.org/uniprotkb/Q9UM73/entry (accessed on 22 November 2022).
  75. KRAS-GTPase KRas-Homo Sapiens (Human) | UniProtKB | UniProt. Available online: https://www.uniprot.org/uniprotkb/P01116/entry (accessed on 22 November 2022).
  76. PIK3CA-Phosphatidylinositol 4,5-Bisphosphate 3-Kinase Catalytic Subunit Alpha Isoform—Homo Sapiens (Human) | UniProtKB | UniProt. Available online: https://www.uniprot.org/uniprotkb/P42336/entry (accessed on 22 November 2022).
  77. TITF1-Thyroid Transcription Factor 1-Canis Lupus Familiaris (Dog) | UniProtKB | UniProt. Available online: https://www.uniprot.org/uniprotkb/P43698/entry (accessed on 22 November 2022).
  78. Bhateja, P.; Chiu, M.; Wildey, G.; Lipka, M.B.; Fu, P.; Yang, M.C.L.; Ardeshir-Larijani, F.; Sharma, N.; Dowlati, A. Retinoblastoma Mutation Predicts Poor Outcomes in Advanced Non Small Cell Lung Cancer. Cancer Med. 2019, 8, 1459–1466. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  79. Farkas, M.; McMahon, S. Unlocking P53 Response Elements: DNA Shape Is the Key. Mol. Cell. Oncol. 2021, 8, 1905489. [Google Scholar] [CrossRef]
  80. Gazzeri, S.; Brambilla, E.; Chauvin, C.; Jacrot, M.; Benabid, A.L.; Brambilla, C. Analysis of the Activation of the Myc Family Oncogene and of Its Stability over Time in Xenografted Human Lung Carcinomas. Cancer Res. 1990, 50, 1566–1570. [Google Scholar]
  81. Offin, M.; Chan, J.M.; Tenet, M.; Rizvi, H.A.; Shen, R.; Riely, G.J.; Rekhtman, N.; Daneshbod, Y.; Quintanal-Villalonga, A.; Penson, A.; et al. Concurrent RB1 and TP53 Alterations Define a Subset of EGFR-Mutant Lung Cancers at Risk for Histologic Transformation and Inferior Clinical Outcomes. J. Thorac. Oncol. 2019, 14, 1784–1793. [Google Scholar] [CrossRef]
  82. Olivier, M.; Petitjean, A.; Marcel, V.; Pétré, A.; Mounawar, M.; Plymoth, A.; de Fromentel, C.C.; Hainaut, P. Recent Advances in P53 Research: An Interdisciplinary Perspective. Cancer Gene Ther. 2009, 16, 1–12. [Google Scholar] [CrossRef] [Green Version]
  83. Sung, H.-J.; Cho, J.-Y. Biomarkers for the Lung Cancer Diagnosis and Their Advances in Proteomics. BMB Reports 2008, 41, 615–625. [Google Scholar] [CrossRef]
  84. Wohlhieter, C.A.; Richards, A.L.; Uddin, F.; Hulton, C.H.; Quintanal-Villalonga, À.; Martin, A.; de Stanchina, E.; Bhanot, U.; Asher, M.; Shah, N.S.; et al. Concurrent Mutations in STK11 and KEAP1 Promote Ferroptosis Protection and SCD1 Dependence in Lung Cancer. Cell Reports 2020, 33, 108444. [Google Scholar] [CrossRef]
  85. MYC-Myc Proto-Oncogene Protein-Homo Sapiens (Human) | UniProtKB | UniProt. Available online: https://www.uniprot.org/uniprotkb/P01106/entry (accessed on 22 November 2022).
  86. MYCL-Protein L-Myc-Homo Sapiens (Human) | UniProtKB | UniProt. Available online: https://www.uniprot.org/uniprotkb/P12524/entry (accessed on 22 November 2022).
  87. MYCN-N-Myc Proto-Oncogene Protein-Homo Sapiens (Human) | UniProtKB | UniProt. Available online: https://www.uniprot.org/uniprotkb/P04198/entry (accessed on 22 November 2022).
  88. RB1-Retinoblastoma-Associated Protein-Homo Sapiens (Human) | UniProtKB | UniProt. Available online: https://www.uniprot.org/uniprotkb/P06400/entry (accessed on 22 November 2022).
  89. STK11-Serine/Threonine-Protein Kinase STK11-Homo Sapiens (Human) | UniProtKB | UniProt. Available online: https://www.uniprot.org/uniprotkb/Q15831/entry (accessed on 22 November 2022).
  90. TP53-Cellular Tumor Antigen P53-Homo Sapiens (Human) | UniProtKB | UniProt. Available online: https://www.uniprot.org/uniprotkb/P04637/entry (accessed on 22 November 2022).
  91. Danial, N.N. BCL-2 Family Proteins: Critical Checkpoints of Apoptotic Cell Death. Clin. Cancer Res. 2007, 13, 7254–7263. [Google Scholar] [CrossRef] [Green Version]
  92. Gavory, G.; Farrow, M.; Balasubramanian, S. Minimum Length Requirement of the Alignment Domain of Human Telomerase RNA to Sustain Catalytic Activity in Vitro. Nucleic Acids Res. 2002, 30, 4470–4480. [Google Scholar] [CrossRef]
  93. Groeger, A.M.; Esposito, V.; De Luca, A.; Cassandro, R.; Tonini, G.; Ambrogi, V.; Baldi, F.; Goldfarb, R.; Mineo, T.C.; Baldi, A.; et al. Prognostic Value of Immunohistochemical Expression of P53, Bax, Bcl-2 and Bcl-XL in Resected Non-Small-Cell Lung Cancers. Histopathology 2004, 44, 54–63. [Google Scholar] [CrossRef] [PubMed]
  94. Porębska, I.; Wyrodek, E.; Kosacka, M.; Adamiak, J.; Jankowska, R.; Harłozińska-Szmyrka, A. Apoptotic Markers P53, Bcl-2 and Bax in Primary Lung Cancer. In Vivo 2006, 20, 599–604. [Google Scholar]
  95. Qi, R.; Wang, D.; Wu, F.; Zhang, S. Correlations between Fas/FasL Expression and Apoptosis as Well as Clinicopathological Features in Non-Small Cell Lung Cancer. Int. J. Clin. Exp. Med. 2020, 6, 4251–4256. [Google Scholar]
  96. Viard-Leveugle, I.; Veyrenc, S.; French, L.E.; Brambilla, C.; Brambilla, E. Frequent Loss of Fas Expression and Function in Human Lung Tumours with Overexpression of FasL in Small Cell Lung Carcinoma. J. Pathol. 2003, 201, 268–277. [Google Scholar] [CrossRef] [PubMed]
  97. 12789-1-AP. Available online: https://www.ptglab.com/products/BCL2-Antibody-12789-1-AP.htm (accessed on 22 November 2022).
  98. 50599-2-Ig. Available online: https://www.ptglab.com/products/BAX-Antibody-50599-2-Ig.htm (accessed on 22 November 2022).
  99. BAX-Apoptosis Regulator BAX-Homo Sapiens (Human) | UniProtKB | UniProt. Available online: https://www.uniprot.org/uniprotkb/Q07812/entry (accessed on 22 November 2022).
  100. BCL2-Apoptosis Regulator Bcl-2-Homo Sapiens (Human) | UniProtKB | UniProt. Available online: https://www.uniprot.org/uniprotkb/P10415/entry (accessed on 22 November 2022).
  101. E2F1-Transcription Factor E2F1-Homo Sapiens (Human) | UniProtKB | UniProt. Available online: https://www.uniprot.org/uniprotkb/Q01094/entry (accessed on 22 November 2022).
  102. FASLG-Tumor Necrosis Factor Ligand Superfamily Member 6-Homo Sapiens (Human) | UniProtKB | UniProt. Available online: https://www.uniprot.org/uniprotkb/P48023/entry (accessed on 22 November 2022).
  103. STK11 Gene: MedlinePlus Genetics. Available online: https://medlineplus.gov/genetics/gene/stk11/ (accessed on 22 November 2022).
  104. TERT-Telomerase Reverse Transcriptase-Homo Sapiens (Human) | UniProtKB | UniProt. Available online: https://www.uniprot.org/uniprotkb/O14746/entry (accessed on 22 November 2022).
  105. Das, P.M.; Singal, R. DNA Methylation and Cancer. JCO 2004, 22, 4632–4642. [Google Scholar] [CrossRef] [Green Version]
  106. Duruisseaux, M.; Esteller, M. Lung Cancer Epigenetics: From Knowledge to Applications. Semin. Cancer Biol. 2018, 51, 116–128. [Google Scholar] [CrossRef] [PubMed]
  107. Langevin, S.M.; Kratzke, R.A.; Kelsey, K.T. Epigenetics of Lung Cancer. Transl. Res. 2015, 165, 74–90. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  108. Sauerwald, A.; Sandin, S.; Cristofari, G.; Scheres, S.H.W.; Lingner, J.; Rhodes, D. Structure of Active Dimeric Human Telomerase. Nat. Struct Mol. Biol. 2013, 20, 454–460. [Google Scholar] [CrossRef] [PubMed]
  109. Shi, Y.-X.; Sheng, D.-Q.; Cheng, L.; Song, X.-Y. Current Landscape of Epigenetics in Lung Cancer: Focus on the Mechanism and Application. J. Oncol. 2019, 2019, e8107318. [Google Scholar] [CrossRef] [Green Version]
  110. Wajed, S.A.; Laird, P.W.; DeMeester, T.R. DNA Methylation: An Alternative Pathway to Cancer. Ann. Surg 2001, 234, 10–20. [Google Scholar] [CrossRef] [PubMed]
  111. Debernardi, C.; Libera, L.; Berrino, E.; Sahnane, N.; Chiaravalli, A.M.; Laudi, C.; Berselli, M.; Sapino, A.; Sessa, F.; Venesio, T.; et al. Evaluation of Global and Intragenic Hypomethylation in Colorectal Adenomas Improves Patient Stratification and Colorectal Cancer Risk Prediction. Clin. Epigenetics 2021, 13, 154. [Google Scholar] [CrossRef] [PubMed]
  112. Ehrlich, M. DNA Hypomethylation in Cancer Cells. Epigenomics 2009, 1, 239–259. [Google Scholar] [CrossRef] [PubMed]
  113. Lujambio, A. CpG Island Hypermethylation of Tumor Suppressor MicroRNAs in Human Cancer. Cell Cycle 2007, 6, 1454–1458. [Google Scholar] [CrossRef]
  114. Nishiyama, A.; Nakanishi, M. Navigating the DNA Methylation Landscape of Cancer. Trends Genet. 2021, 37, 1012–1027. [Google Scholar] [CrossRef] [PubMed]
  115. Wang, S.; Wu, W.; Claret, F.X. Mutual Regulation of MicroRNAs and DNA Methylation in Human Cancers. Epigenetics 2017, 12, 187–197. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  116. Wedge, E.; Hansen, J.W.; Garde, C.; Asmar, F.; Tholstrup, D.; Kristensen, S.S.; Munch-Petersen, H.D.; Ralfkiaer, E.; Brown, P.; Grønbæk, K.; et al. Global Hypomethylation Is an Independent Prognostic Factor in Diffuse Large B Cell Lymphoma. Am. J. Hematol. 2017, 92, 689–694. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  117. Do, H.; Wong, N.C.; Murone, C.; John, T.; Solomon, B.; Mitchell, P.L.; Dobrovic, A. A Critical Re-Assessment of DNA Repair Gene Promoter Methylation in Non-Small Cell Lung Carcinoma. Sci. Rep. 2014, 4, 4186. [Google Scholar] [CrossRef] [Green Version]
  118. Feng, L.; Chen, C.; Li, L. Hypermethylation of Tumor Suppressor Genes Is a Risk Factor for Poor Prognosis in Ovarian Cancer: A Meta-Analysis. Medicine 2019, 98, e14588. [Google Scholar] [CrossRef]
  119. Ng, J.M.-K.; Yu, J. Promoter Hypermethylation of Tumour Suppressor Genes as Potential Biomarkers in Colorectal Cancer. Int. J. Mol. Sci. 2015, 16, 2472–2496. [Google Scholar] [CrossRef] [Green Version]
  120. Tam, K.W.; Zhang, W.; Soh, J.; Stastny, V.; Chen, M.; Sun, H.; Thu, K.; Rios, J.J.; Yang, C.; Marconett, C.N.; et al. CDKN2A/P16 Inactivation Mechanisms and Their Relationship to Smoke Exposure and Molecular Features in Non-Small-Cell Lung Cancer. J. Thorac. Oncol. 2013, 8, 1378–1388. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  121. Walter, R.F.H.; Rozynek, P.; Casjens, S.; Werner, R.; Mairinger, F.D.; Speel, E.J.M.; Zur Hausen, A.; Meier, S.; Wohlschlaeger, J.; Theegarten, D.; et al. Methylation of L1RE1, RARB, and RASSF1 Function as Possible Biomarkers for the Differential Diagnosis of Lung Cancer. PLoS ONE 2018, 13, e0195716. [Google Scholar] [CrossRef] [PubMed]
  122. Czarnecka, K.H.; Szmyd, B.; Barańska, M.; Kaszkowiak, M.; Kordiak, J.; Antczak, A.; Pastuszak-Lewandoska, D.; Brzeziańska-Lasota, E. A Strong Decrease in TIMP3 Expression Mediated by the Presence of MiR-17 and 20a Enables Extracellular Matrix Remodeling in the NSCLC Lesion Surroundings. Front. Oncol. 2019, 9, 1372. [Google Scholar] [CrossRef] [PubMed]
  123. Gu, C.; Lu, J.; Cui, T.; Lu, C.; Shi, H.; Xu, W.; Yuan, X.; Yang, X.; Huang, Y.; Lu, M. Association between MGMT Promoter Methylation and Non-Small Cell Lung Cancer: A Meta-Analysis. PLoS ONE 2013, 8, e72633. [Google Scholar] [CrossRef]
  124. Su, C.-W.; Chang, Y.-C.; Chien, M.-H.; Hsieh, Y.-H.; Chen, M.-K.; Lin, C.-W.; Yang, S.-F. Loss of TIMP3 by Promoter Methylation of Sp1 Binding Site Promotes Oral Cancer Metastasis. Cell Death Dis. 2019, 10, 793. [Google Scholar] [CrossRef] [Green Version]
  125. Li, Y.; Zhu, M.; Zhang, X.; Cheng, D.; Ma, X. Clinical Significance of DAPK Promoter Hypermethylation in Lung Cancer: A Meta-Analysis. Drug Des. Devel. Ther. 2015, 9, 1785–1796. [Google Scholar] [CrossRef] [Green Version]
  126. Pu, W.; Geng, X.; Chen, S.; Tan, L.; Tan, Y.; Wang, A.; Lu, Z.; Guo, S.; Chen, X.; Wang, J. Aberrant Methylation of CDH13 Can Be a Diagnostic Biomarker for Lung Adenocarcinoma. J. Cancer 2016, 7, 2280. [Google Scholar] [CrossRef] [Green Version]
  127. Wang, Y.; Zhang, L.; Yang, J.; Li, B.; Wang, J. CDH13 Promoter Methylation Regulates Cisplatin Resistance of Non-Small Cell Lung Cancer Cells. Oncol. Lett. 2018, 16, 5715–5722. [Google Scholar] [CrossRef]
  128. Yu, Q.; Guo, Q.; Chen, L.; Liu, S. Clinicopathological Significance and Potential Drug Targeting of CDH1 in Lung Cancer: A Meta-Analysis and Literature Review. Drug Des. Devel. Ther. 2015, 9, 2171–2178. [Google Scholar] [CrossRef] [Green Version]
  129. Zhang, Y.; Wu, J.; Huang, G.; Xu, S. Clinicopathological Significance of DAPK Promoter Methylation in Non-Small-Cell Lung Cancer: A Systematic Review and Meta-Analysis. Cancer Manag. Res. 2018, 10, 6897–6904. [Google Scholar] [CrossRef] [Green Version]
  130. Bajbouj, K.; Al-Ali, A.; Ramakrishnan, R.K.; Saber-Ayad, M.; Hamid, Q. Histone Modification in NSCLC: Molecular Mechanisms and Therapeutic Targets. Int. J. Mol. Sci. 2021, 22, 11701. [Google Scholar] [CrossRef]
  131. Barlési, F.; Giaccone, G.; Gallegos-Ruiz, M.I.; Loundou, A.; Span, S.W.; Lefesvre, P.; Kruyt, F.A.E.; Rodriguez, J.A. Global Histone Modifications Predict Prognosis of Resected Non Small-Cell Lung Cancer. J. Clin. Oncol. 2007, 25, 4358–4364. [Google Scholar] [CrossRef] [PubMed]
  132. Seligson, D.B.; Horvath, S.; McBrian, M.A.; Mah, V.; Yu, H.; Tze, S.; Wang, Q.; Chia, D.; Goodglick, L.; Kurdistani, S.K. Global Levels of Histone Modifications Predict Prognosis in Different Cancers. Am. J. Pathol. 2009, 174, 1619–1628. [Google Scholar] [CrossRef]
  133. Van Den Broeck, A.; Brambilla, E.; Moro-Sibilot, D.; Lantuejoul, S.; Brambilla, C.; Eymin, B.; Gazzeri, S. Loss of Histone H4K20 Trimethylation Occurs in Preneoplasia and Influences Prognosis of Non-Small Cell Lung Cancer. Clin. Cancer Res. 2008, 14, 7237–7245. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  134. Ye, T.; Li, J.; Sun, Z.; Liu, D.; Zeng, B.; Zhao, Q.; Wang, J.; Xing, H.R. Cdh1 Functions as an Oncogene by Inducing Self-Renewal of Lung Cancer Stem-like Cells via Oncogenic Pathways. Int. J. Biol. Sci. 2020, 16, 447–459. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  135. Zhao, Z.; Shilatifard, A. Epigenetic Modifications of Histones in Cancer. Genome Biol. 2019, 20, 245. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  136. Asakura, K.; Kadota, T.; Matsuzaki, J.; Yoshida, Y.; Yamamoto, Y.; Nakagawa, K.; Takizawa, S.; Aoki, Y.; Nakamura, E.; Miura, J.; et al. A MiRNA-Based Diagnostic Model Predicts Resectable Lung Cancer in Humans with High Accuracy. Commun. Biol. 2020, 3, 134. [Google Scholar] [CrossRef] [Green Version]
  137. Gajda, E.; Grzanka, M.; Godlewska, M.; Gawel, D. The Role of MiRNA-7 in the Biology of Cancer and Modulation of Drug Resistance. Pharmaceuticals (Basel) 2021, 14, 149. [Google Scholar] [CrossRef]
  138. Zhao, J.; Tao, Y.; Zhou, Y.; Qin, N.; Chen, C.; Tian, D.; Xu, L. MicroRNA-7: A Promising New Target in Cancer Therapy. Cancer Cell Int. 2015, 15, 103. [Google Scholar] [CrossRef] [Green Version]
  139. Zhong, S.; Golpon, H.; Zardo, P.; Borlak, J. MiRNAs in Lung Cancer. A Systematic Review Identifies Predictive and Prognostic MiRNA Candidates for Precision Medicine in Lung Cancer. Transl. Res. 2021, 230, 164–196. [Google Scholar] [CrossRef]
  140. Zhu, X.; Kudo, M.; Huang, X.; Sui, H.; Tian, H.; Croce, C.M.; Cui, R. Frontiers of MicroRNA Signature in Non-Small Cell Lung Cancer. Front. Cell Dev. Biol. 2021, 9, 643942. [Google Scholar] [CrossRef] [PubMed]
  141. Acunzo, M.; Romano, G.; Palmieri, D.; Laganá, A.; Garofalo, M.; Balatti, V.; Drusco, A.; Chiariello, M.; Nana-Sinkam, P.; Croce, C.M. Cross-Talk between MET and EGFR in Non-Small Cell Lung Cancer Involves MiR-27a and Sprouty2. Proc. Natl. Acad. Sci. USA 2013, 110, 8573–8578. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  142. Guan, Y.; Rao, Z.; Chen, C. MiR-30a Suppresses Lung Cancer Progression by Targeting SIRT1. Oncotarget 2017, 9, 4924–4934. [Google Scholar] [CrossRef] [PubMed]
  143. Li, X.; Xu, M.; Ding, L.; Tang, J. MiR-27a: A Novel Biomarker and Potential Therapeutic Target in Tumors. J. Cancer 2019, 10, 2836–2848. [Google Scholar] [CrossRef] [Green Version]
  144. Zhang, J.; Cao, Z.; Yang, G.; You, L.; Zhang, T.; Zhao, Y. MicroRNA-27a (MiR-27a) in Solid Tumors: A Review Based on Mechanisms and Clinical Observations. Front. Oncol. 2019, 9, 893. [Google Scholar] [CrossRef] [Green Version]
  145. MIR27A MicroRNA 27a [Homo Sapiens (Human)]-Gene-NCBI. Available online: https://www.ncbi.nlm.nih.gov/gene/407018 (accessed on 22 November 2022).
  146. MIR30A MicroRNA 30a [Homo Sapiens (Human)]-Gene-NCBI. Available online: https://www.ncbi.nlm.nih.gov/gene/407029 (accessed on 22 November 2022).
  147. Hu, J.; Cheng, Y.; Li, Y.; Jin, Z.; Pan, Y.; Liu, G.; Fu, S.; Zhang, Y.; Feng, K.; Feng, Y. MicroRNA-128 Plays a Critical Role in Human Non-Small Cell Lung Cancer Tumourigenesis, Angiogenesis and Lymphangiogenesis by Directly Targeting Vascular Endothelial Growth Factor-C. Eur. J. Cancer 2014, 50, 2336–2350. [Google Scholar] [CrossRef]
  148. Li, L.; Wang, D. MicroRNA-128-b Regulates Epidermal Growth Factor Receptor Expression in Non-Small Cell Lung Cancer. Mol. Med. Rep. 2019, 20, 4803–4810. [Google Scholar] [CrossRef]
  149. PubChem MIR128-1-MicroRNA 128-1 (Human). Available online: https://pubchem.ncbi.nlm.nih.gov/gene/MIR128-1/human (accessed on 22 November 2022).
  150. Rupaimoole, R.; Yoon, B.; Zhang, W.C.; Adams, B.D.; Slack, F.J. A High-Throughput Small Molecule Screen Identifies Ouabain as Synergistic with MiR-34a in Killing Lung Cancer Cells. iScience 2020, 23, 100878. [Google Scholar] [CrossRef] [Green Version]
  151. Song, K.; Jiang, Y.; Zhao, Y.; Xie, Y.; Zhou, J.; Yu, W.; Wang, Q. Members of the MiR-30 Family Inhibit the Epithelial-to-Mesenchymal Transition of Non-Small-Cell Lung Cancer Cells by Suppressing XB130 Expression Levels. Oncol. Lett. 2020, 20, 68. [Google Scholar] [CrossRef]
  152. Zhang, L.; Liao, Y.; Tang, L. MicroRNA-34 Family: A Potential Tumor Suppressor and Therapeutic Candidate in Cancer. J. Exp. Clin. Cancer Res. 2019, 38, 53. [Google Scholar] [CrossRef] [Green Version]
  153. MIR34A MicroRNA 34a [ Homo Sapiens (Human)]. Available online: https://www.ncbi.nlm.nih.gov/gene/407040 (accessed on 22 November 2022).
  154. Liao, Y.; Wu, X.; Wu, M.; Fang, Y.; Li, J.; Tang, W. Non-Coding RNAs in Lung Cancer: Emerging Regulators of Angiogenesis. J. Transl. Med. 2022, 20, 349. [Google Scholar] [CrossRef] [PubMed]
  155. Liu, B.; Li, J.; Zheng, M.; Ge, J.; Li, J.; Yu, P. MiR-542-3p Exerts Tumor Suppressive Functions in Non-Small Cell Lung Cancer Cells by Upregulating FTSJ2. Life Sci. 2017, 188, 87–95. [Google Scholar] [CrossRef] [PubMed]
  156. Pan, J.-Y.; Zhang, F.; Sun, C.-C.; Li, S.-J.; Li, G.; Gong, F.-Y.; Bo, T.; He, J.; Hua, R.-X.; Hu, W.-D.; et al. MiR-134: A Human Cancer Suppressor? Mol. Ther. Nucleic Acids 2017, 6, 140–149. [Google Scholar] [CrossRef] [PubMed]
  157. PubChem MIR542-MicroRNA 542 (Human). Available online: https://pubchem.ncbi.nlm.nih.gov/gene/MIR542/human (accessed on 22 November 2022).
  158. Qin, Q.; Wei, F.; Zhang, J.; Li, B. MiR-134 Suppresses the Migration and Invasion of Non-small Cell Lung Cancer by Targeting ITGB1. Oncol. Rep. 2017, 37, 823–830. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  159. MIR134 MicroRNA 134 [Homo Sapiens (Human)]-Gene-NCBI. Available online: https://www.ncbi.nlm.nih.gov/gene/406924 (accessed on 22 November 2022).
  160. Chu, D.; Li, J.; Lin, H.; Zhang, X.; Pan, H.; Liu, L.; Yu, T.; Yan, M.; Yao, M. Quantitative Proteomic Analysis of the MiR-148a-Associated Mechanisms of Metastasis in Non-Small Cell Lung Cancer. Oncol. Lett. 2018, 15, 9941–9952. [Google Scholar] [CrossRef]
  161. He, R.; Li, X.; Liang, L.; Xie, Y.; Luo, D.; Ma, J.; Peng, Z.; Hu, X.; Chen, G. The Suppressive Role of MiR-542-5p in NSCLC: The Evidence from Clinical Data and in Vivo Validation Using a Chick Chorioallantoic Membrane Model. BMC Cancer 2017, 17, 655. [Google Scholar] [CrossRef] [Green Version]
  162. Liang, L.; Xu, W.; Shen, A.; Cen, H.; Chen, Z.; Tan, L.; Zhang, L.; Zhang, Y.; Fu, J.; Qin, A.; et al. Promoter Methylation-Regulated MiR-148a-3p Inhibits Lung Adenocarcinoma (LUAD) Progression by Targeting MAP3K9. Acta Pharmacol. Sin. 2022, 43, 2946–2955. [Google Scholar] [CrossRef]
  163. Qian, M.; Xia, Y.; Zhang, G.; Yu, H.; Cui, Y. Research Progress on MicroRNA-1258 in the Development of Human Cancer. Front. Oncol. 2022, 12, 1024234. [Google Scholar] [CrossRef]
  164. MicroRNA-1258 Suppresses Tumour Progression via GRB2/Ras/Erk Pathway in Non-small-cell Lung Cancer; John Wiley & Sons, Ltd.: Hoboken, NJ, USA, 2022. [CrossRef]
  165. MIR148A MicroRNA 148a [Homo Sapiens (Human)]-Gene-NCBI. Available online: https://www.ncbi.nlm.nih.gov/gene/406940 (accessed on 22 November 2022).
  166. MIR1258 MicroRNA 1258 [Homo Sapiens (Human)]-Gene-NCBI. Available online: https://www.ncbi.nlm.nih.gov/gene/100302172 (accessed on 22 November 2022).
  167. Braicu, C.; Gulei, D.; Cojocneanu, R.; Raduly, L.; Jurj, A.; Knutsen, E.; Calin, G.A.; Berindan-Neagoe, I. MiR-181a/b Therapy in Lung Cancer: Reality or Myth? Mol. Oncol. 2019, 13, 9–25. [Google Scholar] [CrossRef] [Green Version]
  168. Ding, H.; Chu, M.; Yue, J.; Huang, H.; Wang, J.; Zhu, L. MiR-96 Induced Non-Small-Cell Lung Cancer Progression through Competing Endogenous RNA Network and Affecting EGFR Signaling Pathway. Iran J. Basic Med. Sci. 2019, 22, 908–914. [Google Scholar] [CrossRef]
  169. Fan, Q.; Hu, X.; Zhang, H.; Wang, S.; Zhang, H.; You, C.; Zhang, C.-Y.; Liang, H.; Chen, X.; Ba, Y. MiR-193a-3p Is an Important Tumour Suppressor in Lung Cancer and Directly Targets KRAS. Cell Physiol. Biochem. 2017, 44, 1311–1324. [Google Scholar] [CrossRef] [PubMed]
  170. Gao, X.; Tang, R.; Xie, Q.; Lin, J.; Shi, H.; Chen, G.; Li, Z. The Clinical Value of MiR-193a-3p in Non-small Cell Lung Cancer and Its Potential Molecular Mechanism Explored in Silico Using RNA-sequencing and Microarray Data. FEBS Open Bio. 2018, 8, 94–109. [Google Scholar] [CrossRef] [PubMed]
  171. Ge, H.; Wang, L.; Chen, W.; Wang, L. Mechanism of MiR-760 Reversing Lung Cancer Immune Escape by Downregulating IDO1 and Eliminating Regulatory T Cells Based on Mathematical Biology. Comput. Math Methods Med. 2022, 2022, 2960773. [Google Scholar] [CrossRef] [PubMed]
  172. Shi, Q.; Zhou, Z.; Ye, N.; Chen, Q.; Zheng, X.; Fang, M. MiR-181a Inhibits Non-Small Cell Lung Cancer Cell Proliferation by Targeting CDK1. Cancer Biomark 2017, 20, 539–546. [Google Scholar] [CrossRef] [PubMed]
  173. Wu, H.; Zhou, J.; Mei, S.; Wu, D.; Mu, Z.; Chen, B.; Xie, Y.; Ye, Y.; Liu, J. Circulating Exosomal MicroRNA-96 Promotes Cell Proliferation, Migration and Drug Resistance by Targeting LMO7. J. Cell Mol. Med. 2017, 21, 1228–1236. [Google Scholar] [CrossRef]
  174. Xue, B.; Chuang, C.-H.; Prosser, H.M.; Fuziwara, C.S.; Chan, C.; Sahasrabudhe, N.; Kühn, M.; Wu, Y.; Chen, J.; Biton, A.; et al. MiR-200 Deficiency Promotes Lung Cancer Metastasis by Activating Notch Signaling in Cancer-Associated Fibroblasts. Genes Dev. 2021, 35, 1109–1122. [Google Scholar] [CrossRef] [PubMed]
  175. Yan, C.; Zhang, W.; Shi, X.; Zheng, J.; Jin, X.; Huo, J. MiR-760 Suppresses Non-Small Cell Lung Cancer Proliferation and Metastasis by Targeting ROS1. Environ Sci Pollut Res Int 2018, 25, 18385–18391. [Google Scholar] [CrossRef]
  176. MIR96 MicroRNA 96 [Homo Sapiens (Human)]-Gene-NCBI. Available online: https://www.ncbi.nlm.nih.gov/gene/407053 (accessed on 22 November 2022).
  177. MIR193A MicroRNA 193a [Homo Sapiens (Human)]-Gene-NCBI. Available online: https://www.ncbi.nlm.nih.gov/gene/406968 (accessed on 22 November 2022).
  178. MIR200C MicroRNA 200c [ Homo Sapiens (Human)]. Available online: https://www.ncbi.nlm.nih.gov/gene/406985 (accessed on 22 November 2022).
  179. MIR760 MicroRNA 760 [Homo Sapiens (Human)]-Gene-NCBI. Available online: https://www.ncbi.nlm.nih.gov/gene/100126348 (accessed on 22 November 2022).
  180. Chen, J.; Cui, J.; Guo, X.; Cao, X.; Li, Q. Increased Expression of MiR-641 Contributes to Erlotinib Resistance in Non-Small-Cell Lung Cancer Cells by Targeting NF1. Cancer Med. 2018, 7, 1394–1403. [Google Scholar] [CrossRef]
  181. Fang, X.; Shi, H.; Sun, F. The MicroRNA-520a-3p Inhibits Invasion and Metastasis by Targeting NF-KappaB Signaling Pathway in Non-Small Cell Lung Cancer. Clin. Transl. Oncol. 2022, 24, 1569–1579. [Google Scholar] [CrossRef]
  182. Kong, Q.; Shu, N.; Li, J.; Xu, N. MiR-641 Functions as a Tumor Suppressor by Targeting MDM2 in Human Lung Cancer. Oncol. Res. 2018, 26, 735–741. [Google Scholar] [CrossRef]
  183. Li, J.; Tan, Q.; Yan, M.; Liu, L.; Lin, H.; Zhao, F.; Bao, G.; Kong, H.; Ge, C.; Zhang, F.; et al. MiRNA-200c Inhibits Invasion and Metastasis of Human Non-Small Cell Lung Cancer by Directly Targeting Ubiquitin Specific Peptidase 25. Mol. Cancer 2014, 13, 166. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  184. Liu, P.-L.; Liu, W.-L.; Chang, J.-M.; Chen, Y.-H.; Liu, Y.-P.; Kuo, H.-F.; Hsieh, C.-C.; Ding, Y.-S.; Chen, W.-W.; Chong, I.-W. MicroRNA-200c Inhibits Epithelial-Mesenchymal Transition, Invasion, and Migration of Lung Cancer by Targeting HMGB1. PLoS ONE 2017, 12, e0180844. [Google Scholar] [CrossRef] [PubMed]
  185. Xie, Y.; Xue, C.; Guo, S.; Yang, L. MicroRNA-520a Suppresses Pathogenesis and Progression of Non-Small-Cell Lung Cancer through Targeting the RRM2/Wnt Axis. Anal. Cell Pathol. (Amst) 2021, 2021, 9652420. [Google Scholar] [CrossRef]
  186. MicroRNA-520a-3p Inhibits Cell Growth and Metastasis of Non-Small Cell Lung Cancer through PI3K/AKT/MTOR Signaling Pathway. Eur. Rev. 2018, 22, 2321–2327.
  187. MIR520A MicroRNA 520a [Homo Sapiens (Human)]-Gene-NCBI. Available online: https://www.ncbi.nlm.nih.gov/gene/574467 (accessed on 22 November 2022).
  188. MIR641 MicroRNA 641 [Homo Sapiens (Human)]-Gene-NCBI. Available online: https://www.ncbi.nlm.nih.gov/gene/693226 (accessed on 22 November 2022).
  189. MIR660 MicroRNA 660 [Homo Sapiens (Human)]-Gene-NCBI. Available online: https://www.ncbi.nlm.nih.gov/gene/724030 (accessed on 22 November 2022).
  190. Andriani, F.; Majorini, M.T.; Mano, M.; Landoni, E.; Miceli, R.; Facchinetti, F.; Mensah, M.; Fontanella, E.; Dugo, M.; Giacca, M.; et al. MiR-16 Regulates the pro-Tumorigenic Potential of Lung Fibroblasts through the Inhibition of HGF Production in an FGFR-1- and MEK1-Dependent Manner. J. Hematol. Oncol. 2018, 11, 45. [Google Scholar] [CrossRef] [Green Version]
  191. Bandi, N.; Zbinden, S.; Gugger, M.; Arnold, M.; Kocher, V.; Hasan, L.; Kappeler, A.; Brunner, T.; Vassella, E. MiR-15a and MiR-16 Are Implicated in Cell Cycle Regulation in a Rb-Dependent Manner and Are Frequently Deleted or down-Regulated in Non-Small Cell Lung Cancer. Cancer Res. 2009, 69, 5553–5559. [Google Scholar] [CrossRef] [Green Version]
  192. Chatterjee, A.; Chattopadhyay, D.; Chakrabarti, G. MiR-16 Targets Bcl-2 in Paclitaxel-Resistant Lung Cancer Cells and Overexpression of MiR-16 along with MiR-17 Causes Unprecedented Sensitivity by Simultaneously Modulating Autophagy and Apoptosis. Cell Signal. 2015, 27, 189–203. [Google Scholar] [CrossRef]
  193. Chen, T.; Xiao, Q.; Wang, X.; Wang, Z.; Hu, J.; Zhang, Z.; Gong, Z.; Chen, S. MiR-16 Regulates Proliferation and Invasion of Lung Cancer Cells via the ERK/MAPK Signaling Pathway by Targeted Inhibition of MAPK Kinase 1 (MEK1). J. Int. Med. Res. 2019, 47, 5194–5204. [Google Scholar] [CrossRef] [Green Version]
  194. Cimmino, A.; Calin, G.A.; Fabbri, M.; Iorio, M.V.; Ferracin, M.; Shimizu, M.; Wojcik, S.E.; Aqeilan, R.I.; Zupo, S.; Dono, M.; et al. MiR-15 and MiR-16 Induce Apoptosis by Targeting BCL2. Proc. Natl. Acad. Sci. USA 2005, 102, 13944–13949. [Google Scholar] [CrossRef] [Green Version]
  195. Fortunato, O.; Boeri, M.; Moro, M.; Verri, C.; Mensah, M.; Conte, D.; Caleca, L.; Roz, L.; Pastorino, U.; Sozzi, G. Mir-660 Is Downregulated in Lung Cancer Patients and Its Replacement Inhibits Lung Tumorigenesis by Targeting MDM2-P53 Interaction. Cell Death Dis. 2014, 5, e1564. [Google Scholar] [CrossRef] [Green Version]
  196. Guo, S.; Li, M.; Li, J.; Lv, Y. Inhibition Mechanism of Lung Cancer Cell Metastasis through Targeted Regulation of Smad3 by MiR-15a. Oncol. Lett. 2020, 19, 1516–1522. [Google Scholar] [CrossRef] [PubMed]
  197. Luo, W.; Huang, B.; Li, Z.; Li, H.; Sun, L.; Zhang, Q.; Qiu, X.; Wang, E. MicroRNA-449a Is Downregulated in Non-Small Cell Lung Cancer and Inhibits Migration and Invasion by Targeting c-Met. PLoS ONE 2013, 8, e64759. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  198. PubChem MIR15A-MicroRNA 15a (Human). Available online: https://pubchem.ncbi.nlm.nih.gov/gene/MIR15A/human (accessed on 22 November 2022).
  199. MIR449A MicroRNA 449a [Homo Sapiens (Human)]-Gene-NCBI. Available online: https://www.ncbi.nlm.nih.gov/gene/554213 (accessed on 22 November 2022).
  200. Hu, S.; Cao, P.; Kong, K.; Han, P.; Deng, Y.; Li, F.; Zhao, B. MicroRNA-449a Delays Lung Cancer Development through Inhibiting KDM3A/HIF-1α Axis. J. Transl. Med. 2021, 19, 224. [Google Scholar] [CrossRef] [PubMed]
  201. Wang, Z.; Zhu, X.; Zhang, T.; Yao, F. MiR-512-5p Suppresses the Progression of Non-small Cell Lung Cancer by Targeting Β-catenin. Oncol. Lett. 2020, 19, 415–423. [Google Scholar] [CrossRef] [Green Version]
  202. Wu, D.; Liu, J.; Chen, J.; He, H.; Ma, H.; Lv, X. MiR-449a Suppresses Tumor Growth, Migration, and Invasion in Non-Small Cell Lung Cancer by Targeting a HMGB1-Mediated NF-ΚB Signaling Pathway. Oncol. Res. 2019, 27, 227–235. [Google Scholar] [CrossRef] [PubMed]
  203. Wu, F.; Ji, A.; Zhang, Z.; Li, J.; Li, P. MiR-491-5p Inhibits the Proliferation and Migration of A549 Cells by FOXP4. Exp. Ther. Med. 2021, 21, 622. [Google Scholar] [CrossRef] [PubMed]
  204. Yuan, Y.; Chen, X.; Yuan, B.; Hu, Y. Effect of Mir-299-3p on the Biological Function of Lung Adenocarcinoma Cell H1299 through Targeting PTPRD. Food Sci. Technol. 2022, 42, e43321. [Google Scholar] [CrossRef]
  205. Zhang, G.; Zheng, H.; Wang, L. MiR-491-3p Functions as a Tumor Suppressor in Non-Small Cell Lung Cancer by Targeting Fibroblast Growth Factor 5. Oncol. Rep. 2022, 48, 164. [Google Scholar] [CrossRef]
  206. Zheng, D.; Dai, Y.; Wang, S.; Xing, X. MicroRNA-299-3p Promotes the Sensibility of Lung Cancer to Doxorubicin through Directly Targeting ABCE1. Int. J. Clin. Exp. Pathol. 2015, 8, 10072–10081. [Google Scholar]
  207. MIR299 MicroRNA 299 [Homo Sapiens (Human)]-Gene-NCBI. Available online: https://www.ncbi.nlm.nih.gov/gene/407023 (accessed on 22 November 2022).
  208. MIR491 MicroRNA 491 [Homo Sapiens (Human)]-Gene-NCBI. Available online: https://www.ncbi.nlm.nih.gov/gene/574444 (accessed on 22 November 2022).
  209. MIR512-1 MicroRNA 512-1 [Homo Sapiens (Human)]-Gene-NCBI. Available online: https://www.ncbi.nlm.nih.gov/gene/574458 (accessed on 22 November 2022).
  210. Bica-Pop, C.; Cojocneanu-Petric, R.; Magdo, L.; Raduly, L.; Gulei, D.; Berindan-Neagoe, I. Overview upon MiR-21 in Lung Cancer: Focus on NSCLC. Cell Mol. Life Sci. 2018, 75, 3539–3551. [Google Scholar] [CrossRef]
  211. Chu, K.; Gao, G.; Yang, X.; Ren, S.; Li, Y.; Wu, H.; Huang, Y.; Zhou, C. MiR-512-5p Induces Apoptosis and Inhibits Glycolysis by Targeting P21 in Non-Small Cell Lung Cancer Cells. Int. J. Oncol. 2016, 48, 577–586. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  212. Fabbri, M.; Garzon, R.; Cimmino, A.; Liu, Z.; Zanesi, N.; Callegari, E.; Liu, S.; Alder, H.; Costinean, S.; Fernandez-Cymering, C.; et al. MicroRNA-29 Family Reverts Aberrant Methylation in Lung Cancer by Targeting DNA Methyltransferases 3A and 3B. Proc. Natl. Acad. Sci. USA 2007, 104, 15805–15810. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  213. Liu, X.; Liu, B.; Li, R.; Wang, F.; Wang, N.; Zhang, M.; Bai, Y.; Wu, J.; Liu, L.; Han, D.; et al. MiR-146a-5p Plays an Oncogenic Role in NSCLC via Suppression of TRAF6. Front Cell Dev. Biol. 2020, 8, 847. [Google Scholar] [CrossRef]
  214. Liu, X.; Lv, X.; Yang, Q.; Jin, H.; Zhou, W.; Fan, Q. MicroRNA-29a Functions as a Tumor Suppressor and Increases Cisplatin Sensitivity by Targeting NRAS in Lung Cancer. Technol. Cancer Res. Treat 2018, 17, 1533033818758905. [Google Scholar] [CrossRef]
  215. Markou, A.; Zavridou, M.; Lianidou, E.S. MiRNA-21 as a Novel Therapeutic Target in Lung Cancer. Lung Cancer (Auckl) 2016, 7, 19–27. [Google Scholar] [CrossRef] [Green Version]
  216. PubChem MIR29A-MicroRNA 29a (Human). Available online: https://pubchem.ncbi.nlm.nih.gov/gene/MIR29A/human (accessed on 22 November 2022).
  217. PubChem MIR146A-MicroRNA 146a (Human). Available online: https://pubchem.ncbi.nlm.nih.gov/gene/MIR146A/human (accessed on 22 November 2022).
  218. Wani, J.A.; Majid, S.; Khan, A.; Arafah, A.; Ahmad, A.; Jan, B.L.; Shah, N.N.; Kazi, M.; Rehman, M.U. Clinico-Pathological Importance of MiR-146a in Lung Cancer. Diagnostics (Basel) 2021, 11, 274. [Google Scholar] [CrossRef] [PubMed]
  219. Zheng, W.; Zhao, J.; Tao, Y.; Guo, M.; Ya, Z.; Chen, C.; Qin, N.; Zheng, J.; Luo, J.; Xu, L. MicroRNA-21: A Promising Biomarker for the Prognosis and Diagnosis of Non-small Cell Lung Cancer (Review). Oncol. Lett. 2018, 16, 2777–2782. [Google Scholar] [CrossRef]
  220. MIR21 MicroRNA 21 [Homo Sapiens (Human)]-Gene-NCBI. Available online: https://www.ncbi.nlm.nih.gov/gene/406991 (accessed on 22 November 2022).
  221. Fan, X.; Tao, S.; Li, Q.; Deng, B.; Tan, Q.-Y.; Jin, H. The MiR-23a/27a/24-2 Cluster Promotes Postoperative Progression of Early-Stage Non-Small Cell Lung Cancer. Mol. Ther. Oncolytics 2022, 24, 205–217. [Google Scholar] [CrossRef]
  222. Feliciano, A.; Garcia-Mayea, Y.; Jubierre, L.; Mir, C.; Hummel, M.; Castellvi, J.; Hernández-Losa, J.; Paciucci, R.; Sansano, I.; Sun, Y.; et al. MiR-99a Reveals Two Novel Oncogenic Proteins E2F2 and EMR2 and Represses Stemness in Lung Cancer. Cell Death Dis. 2017, 8, e3141. [Google Scholar] [CrossRef] [Green Version]
  223. Hetta, H.F.; Zahran, A.M.; Shafik, E.A.; El-Mahdy, R.I.; Mohamed, N.A.; Nabil, E.E.; Esmaeel, H.M.; Alkady, O.A.; Elkady, A.; Mohareb, D.A.; et al. Circulating MiRNA-21 and MiRNA-23a Expression Signature as Potential Biomarkers for Early Detection of Non-Small-Cell Lung Cancer. Microrna 2019, 8, 206–215. [Google Scholar] [CrossRef]
  224. Hsu, Y.-L.; Hung, J.-Y.; Chang, W.-A.; Lin, Y.-S.; Pan, Y.-C.; Tsai, P.-H.; Wu, C.-Y.; Kuo, P.-L. Hypoxic Lung Cancer-Secreted Exosomal MiR-23a Increased Angiogenesis and Vascular Permeability by Targeting Prolyl Hydroxylase and Tight Junction Protein ZO-1. Oncogene 2017, 36, 4929–4942. [Google Scholar] [CrossRef] [PubMed]
  225. Yu, S.; Zhang, C.; Dong, F.; Zhang, Y. MiR-99a Suppresses the Metastasis of Human Non-Small Cell Lung Cancer Cells by Targeting AKT1 Signaling Pathway. J. Cell Biochem. 2015, 116, 268–276. [Google Scholar] [CrossRef] [PubMed]
  226. Zhang, J.; Wang, J.; Zhao, F.; Liu, Q.; Jiang, K.; Yang, G. MicroRNA-21 (MiR-21) Represses Tumor Suppressor PTEN and Promotes Growth and Invasion in Non-Small Cell Lung Cancer (NSCLC). Clin. Chim. Acta 2010, 411, 846–852. [Google Scholar] [CrossRef] [PubMed]
  227. MIR99A MicroRNA 99a [Homo Sapiens (Human)]-Gene-NCBI. Available online: https://www.ncbi.nlm.nih.gov/gene/407055 (accessed on 22 November 2022).
  228. MIR155 MicroRNA 155 [Homo Sapiens (Human)]-Gene-NCBI. Available online: https://www.ncbi.nlm.nih.gov/gene/406947 (accessed on 23 November 2022).
  229. Xue, X.; Liu, Y.; Wang, Y.; Meng, M.; Wang, K.; Zang, X.; Zhao, S.; Sun, X.; Cui, L.; Pan, L.; et al. MiR-21 and MiR-155 Promote Non-Small Cell Lung Cancer Progression by Downregulating SOCS1, SOCS6, and PTEN. Oncotarget 2016, 7, 84508–84519. [Google Scholar] [CrossRef]
  230. Lin, J.; Chen, Y.; Liu, L.; Shen, A.; Zheng, W. MicroRNA-155-5p Suppresses the Migration and Invasion of Lung Adenocarcinoma A549 Cells by Targeting Smad2. Oncol. Lett. 2018, 16, 2444–2452. [Google Scholar] [CrossRef] [Green Version]
  231. The Value of MiR-155 as a Biomarker for the Diagnosis and Prognosis of Lung Cancer: A Systematic Review with Meta-Analysis | BMC Cancer | Full Text. Available online: https://bmccancer.biomedcentral.com/articles/10.1186/s12885-019-6297-6 (accessed on 23 November 2022).
  232. MIR192 MicroRNA 192 [Homo Sapiens (Human)]-Gene-NCBI. Available online: https://www.ncbi.nlm.nih.gov/gene/406967 (accessed on 23 November 2022).
  233. Li, Y.; Zu, L.; Wu, H.; Zhang, F.; Fan, Y.; Pan, H.; Du, X.; Guo, F.; Zhou, Q. MiR-192/NKRF Axis Confers Lung Cancer Cell Chemoresistance to Cisplatin via the NF-κB Pathway. Thorac. Cancer 2022, 13, 430–441. [Google Scholar] [CrossRef]
  234. Wang, L.; Liu, Y.; Lyu, C.; Buchner, A.; Pohla, H. Diagnostic and Prognostic Role of MiR-192 in Different Cancers: A Systematic Review and Meta-Analysis. Biomed. Res. Int. 2021, 2021, 8851035. [Google Scholar] [CrossRef]
  235. Zou, P.; Zhu, M.; Lian, C.; Wang, J.; Chen, Z.; Zhang, X.; Yang, Y.; Chen, X.; Cui, X.; Liu, J.; et al. MiR-192-5p Suppresses the Progression of Lung Cancer Bone Metastasis by Targeting TRIM44. Sci. Rep. 2019, 9, 19619. [Google Scholar] [CrossRef] [Green Version]
  236. Xu, S.; Shi, L. High Expression of MiR-155 and MiR-21 in the Recurrence or Metastasis of Non-Small Cell Lung Cancer. Oncol. Lett. 2019, 18, 758–763. [Google Scholar] [CrossRef] [Green Version]
  237. Yang, M.; Shen, H.; Qiu, C.; Ni, Y.; Wang, L.; Dong, W.; Liao, Y.; Du, J. High Expression of MiR-21 and MiR-155 Predicts Recurrence and Unfavourable Survival in Non-Small Cell Lung Cancer. Eur. J. Cancer 2013, 49, 604–615. [Google Scholar] [CrossRef]
  238. MIR520B MicroRNA 520b [Homo Sapiens (Human)]-Gene-NCBI. Available online: https://www.ncbi.nlm.nih.gov/gene/574473 (accessed on 23 November 2022).
  239. Zhang, L.; Yu, S. Role of MiR-520b in Non-Small Cell Lung Cancer. Exp. Ther. Med. 2018, 16, 3987–3995. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  240. Zhou, Y.; Shen, S. MiR-520f Acts as a Biomarker for the Diagnosis of Lung Cancer. Medicine (Baltimore) 2019, 98, e16546. [Google Scholar] [CrossRef] [PubMed]
  241. MIR497 MicroRNA 497 [Homo Sapiens (Human)]-Gene-NCBI. Available online: https://www.ncbi.nlm.nih.gov/gene/574456 (accessed on 23 November 2022).
  242. Zhao, W.; Wang, Y.; An, Z.; Shi, C.; Zhu, G.; Wang, B.; Lu, M.; Pan, C.; Chen, P. Downregulation of MiR-497 Promotes Tumor Growth and Angiogenesis by Targeting HDGF in Non-Small Cell Lung Cancer. Biochem. Biophys. Res. Commun. 2013, 435, 466–471. [Google Scholar] [CrossRef] [PubMed]
  243. Huang, X.; Wang, L.; Liu, W.; Li, F. MicroRNA-497-5p Inhibits Proliferation and Invasion of Non-Small Cell Lung Cancer by Regulating FGF2. Oncol. Lett. 2019, 17, 3425–3431. [Google Scholar] [CrossRef]
  244. Chae, D.-K.; Park, J.; Cho, M.; Ban, E.; Jang, M.; Yoo, Y.S.; Kim, E.E.; Baik, J.-H.; Song, E.J. MiR-195 and MiR-497 Suppress Tumorigenesis in Lung Cancer by Inhibiting SMURF2-Induced TGF-β Receptor I Ubiquitination. Mol. Oncol. 2019, 13, 2663–2678. [Google Scholar] [CrossRef] [Green Version]
  245. MIR494 MicroRNA 494 [Homo Sapiens (Human)]-Gene-NCBI. Available online: https://www.ncbi.nlm.nih.gov/gene/574452 (accessed on 23 November 2022).
  246. Zhang, J.; Wang, T.; Zhang, Y.; Wang, H.; Wu, Y.; Liu, K.; Pei, C. Upregulation of Serum MiR-494 Predicts Poor Prognosis in Non-Small Cell Lung Cancer Patients. Cancer Biomark 2018, 21, 763–768. [Google Scholar] [CrossRef] [PubMed]
  247. Wang, J.; Chen, H.; Liao, Y.; Chen, N.; Liu, T.; Zhang, H.; Zhang, H. Expression and Clinical Evidence of MiR-494 and PTEN in Non-Small Cell Lung Cancer. Tumour. Biol. 2015, 36, 6965–6972. [Google Scholar] [CrossRef]
  248. Xu, F.; Liu, G.; Wang, L.; Wang, X.; Jin, X.; Bo, W. MiR-494 Promotes Progression of Retinoblastoma via PTEN through PI3K/AKT Signaling Pathway. Oncol. Lett. 2020, 20, 1952–1960. [Google Scholar] [CrossRef]
  249. MIR126 MicroRNA 126 [ Homo Sapiens (Human)]. Available online: https://www.ncbi.nlm.nih.gov/gene/406913 (accessed on 23 November 2022).
  250. Chen, Q.; Chen, S.; Zhao, J.; Zhou, Y.; Xu, L. MicroRNA-126: A New and Promising Player in Lung Cancer. Oncol. Lett. 2021, 21, 35. [Google Scholar] [CrossRef]
  251. Li, M.; Wang, Q.; Zhang, X.; Yan, N.; Li, X. Exosomal MiR-126 Blocks the Development of Non-Small Cell Lung Cancer through the Inhibition of ITGA6. Cancer Cell Int. 2020, 20, 574. [Google Scholar] [CrossRef]
  252. Liu, B.; Peng, X.-C.; Zheng, X.-L.; Wang, J.; Qin, Y.-W. MiR-126 Restoration down-Regulate VEGF and Inhibit the Growth of Lung Cancer Cell Lines in Vitro and in Vivo. Lung Cancer 2009, 66, 169–175. [Google Scholar] [CrossRef] [PubMed]
  253. MIR33B MicroRNA 33b [Homo Sapiens (Human)]-Gene-NCBI. Available online: https://www.ncbi.nlm.nih.gov/gene/693120 (accessed on 23 November 2022).
  254. Zhai, S.; Zhao, L.; Lin, T.; Wang, W. Downregulation of MiR-33b Promotes Non-small Cell Lung Cancer Cell Growth through Reprogramming Glucose Metabolism MiR-33b Regulates Non-small Cell Lung Cancer Cell Growth. J. Cell Biochem. 2019, 120, 6651–6660. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  255. Azizi, M.I.H.N.; Othman, I.; Naidu, R. The Role of MicroRNAs in Lung Cancer Metabolism. Cancers 2021, 13, 1716. [Google Scholar] [CrossRef] [PubMed]
  256. MIR144 MicroRNA 144 [Homo Sapiens (Human)]-Gene-NCBI. Available online: https://www.ncbi.nlm.nih.gov/gene/406936 (accessed on 23 November 2022).
  257. Morais, M.; Dias, F.; Nogueira, I.; Leão, A.; Gonçalves, N.; Araújo, L.; Granja, S.; Baltazar, F.; Teixeira, A.L.; Medeiros, R. Cancer Cells’ Metabolism Dynamics in Renal Cell Carcinoma Patients’ Outcome: Influence of GLUT-1-Related Hsa-MiR-144 and Hsa-MiR-186. Cancers 2021, 13, 1733. [Google Scholar] [CrossRef]
  258. LIU, M.; GAO, J.; HUANG, Q.; JIN, Y.; WEI, Z. Downregulating MicroRNA-144 Mediates a Metabolic Shift in Lung Cancer Cells by Regulating GLUT1 Expression. Oncol. Lett. 2016, 11, 3772–3776. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  259. MIR124-1 MicroRNA 124-1 [Homo Sapiens (Human)]-Gene-NCBI. Available online: https://www.ncbi.nlm.nih.gov/gene/406907 (accessed on 23 November 2022).
  260. Zhao, X.; Lu, C.; Chu, W.; Zhang, B.; Zhen, Q.; Wang, R.; Zhang, Y.; Li, Z.; Lv, B.; Li, H.; et al. MicroRNA-124 Suppresses Proliferation and Glycolysis in Non-Small Cell Lung Cancer Cells by Targeting AKT-GLUT1/HKII. Tumour. Biol. 2017, 39, 1010428317706215. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  261. Jin, H.; Li, Q.; Cao, F.; Wang, S.-N.; Wang, R.-T.; Wang, Y.; Tan, Q.-Y.; Li, C.-R.; Zou, H.; Wang, D.; et al. MiR-124 Inhibits Lung Tumorigenesis Induced by K-Ras Mutation and NNK. Mol. Ther. Nucleic Acids 2017, 9, 145–154. [Google Scholar] [CrossRef] [Green Version]
  262. MIR199A1 MicroRNA 199a-1 [Homo Sapiens (Human)]-Gene-NCBI. Available online: https://www.ncbi.nlm.nih.gov/gene/406976 (accessed on 23 November 2022).
  263. Yang, X.; Zheng, Y.; Tan, J.; Tian, R.; Shen, P.; Cai, W.; Liao, H. MiR-199a-5p–HIF-1α-STAT3 Positive Feedback Loop Contributes to the Progression of Non-Small Cell Lung Cancer. Front Cell Dev. Biol. 2021, 8, 620615. [Google Scholar] [CrossRef]
  264. Wang, L.-M.; Zhang, L.-L.; Wang, L.-W.; Zhu, L.; Ma, X.-X. Influence of MiR-199a on Rats with Non-Small Cell Lung Cancer via Regulating the HIF-1α/VEGF Signaling Pathway. Eur. Rev. Med. Pharmacol. Sci. 2019, 23, 10363–10369. [Google Scholar] [CrossRef]
  265. MIR31 MicroRNA 31 [Homo Sapiens (Human)]-Gene-NCBI. Available online: https://www.ncbi.nlm.nih.gov/gene/407035 (accessed on 23 November 2022).
  266. Davenport, M.L.; Echols, J.B.; Silva, A.D.; Anderson, J.C.; Owens, P.; Yates, C.; Wei, Q.; Harada, S.; Hurst, D.R.; Edmonds, M.D. MiR-31 Displays Subtype Specificity in Lung Cancer. Cancer Res. 2021, 81, 1942–1953. [Google Scholar] [CrossRef]
  267. Zhu, B.; Cao, X.; Zhang, W.; Pan, G.; Yi, Q.; Zhong, W.; Yan, D. MicroRNA-31-5p Enhances the Warburg Effect via Targeting FIH. FASEB J. 2019, 33, 545–556. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  268. MIR28 MicroRNA 28 [Homo Sapiens (Human)]-Gene-NCBI. Available online: https://www.ncbi.nlm.nih.gov/gene/407020 (accessed on 23 November 2022).
  269. Cui, F.; Zhou, Q.; Xiao, K.; Qian, H. MicroRNA-28 Promotes the Proliferation of Non-small-cell Lung Cancer Cells by Targeting PTEN. Mol. Med. Rep. 2020, 21, 2589–2596. [Google Scholar] [CrossRef] [PubMed]
  270. Wang, Q.; Lin, W.; Tang, X.; Li, S.; Guo, L.; Lin, Y.; Kwok, H.F. The Roles of MicroRNAs in Regulating the Expression of PD-1/PD-L1 Immune Checkpoint. Int. J. Mol. Sci. 2017, 18, 2540. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  271. Xing, S.; Xu, Q.; Fan, X.; Wu, S.; Tian, F. Downregulation of MiR-138-5p Promotes Non-small Cell Lung Cancer Progression by Regulating CDK8. Mol. Med. Rep. 2019, 20, 5272–5278. [Google Scholar] [CrossRef]
  272. Song, N.; Li, P.; Song, P.; Li, Y.; Zhou, S.; Su, Q.; Li, X.; Yu, Y.; Li, P.; Feng, M.; et al. MicroRNA-138-5p Suppresses Non-Small Cell Lung Cancer Cells by Targeting PD-L1/PD-1 to Regulate Tumor Microenvironment. Front. Cell Dev. Biol. 2020, 8, 540. [Google Scholar] [CrossRef]
  273. MIR197 MicroRNA 197 [Homo Sapiens (Human)]-Gene-NCBI. Available online: https://www.ncbi.nlm.nih.gov/gene/406974 (accessed on 23 November 2022).
  274. Fujita, Y.; Yagishita, S.; Hagiwara, K.; Yoshioka, Y.; Kosaka, N.; Takeshita, F.; Fujiwara, T.; Tsuta, K.; Nokihara, H.; Tamura, T.; et al. The Clinical Relevance of the MiR-197/CKS1B/STAT3-Mediated PD-L1 Network in Chemoresistant Non-Small-Cell Lung Cancer. Mol. Ther. 2015, 23, 717–727. [Google Scholar] [CrossRef] [Green Version]
  275. Mavridis, K.; Gueugnon, F.; Petit-Courty, A.; Courty, Y.; Barascu, A.; Guyetant, S.; Scorilas, A. The OncomiR MiR-197 Is a Novel Prognostic Indicator for Non-Small Cell Lung Cancer Patients. Br. J. Cancer 2015, 112, 1527–1535. [Google Scholar] [CrossRef]
  276. MIR513A1 MicroRNA 513a-1 [Homo Sapiens (Human)]-Gene-NCBI. Available online: https://www.ncbi.nlm.nih.gov/gene/574509 (accessed on 23 November 2022).
  277. Zhang, X.; Zhu, J.; Xing, R.; Tie, Y.; Fu, H.; Zheng, X.; Yu, B. MiR-513a-3p Sensitizes Human Lung Adenocarcinoma Cells to Chemotherapy by Targeting GSTP1. Lung Cancer 2012, 77, 488–494. [Google Scholar] [CrossRef]
  278. Wang, J.; Sheng, Z.; Cai, Y. Effects of MicroRNA-513b on Cell Proliferation, Apoptosis, Invasion, and Migration by Targeting HMGB3 through Regulation of MTOR Signaling Pathway in Non-Small-Cell Lung Cancer. J. Cell. Physiol. 2019, 234, 10934–10941. [Google Scholar] [CrossRef]
  279. Sp, N.; Kang, D.Y.; Lee, J.-M.; Jang, K.-J. Mechanistic Insights of Anti-Immune Evasion by Nobiletin through Regulating MiR-197/STAT3/PD-L1 Signaling in Non-Small Cell Lung Cancer (NSCLC) Cells. Int. J. Mol. Sci. 2021, 22, 9843. [Google Scholar] [CrossRef]
  280. MIR20A MicroRNA 20a [Homo Sapiens (Human)]-Gene-NCBI. Available online: https://www.ncbi.nlm.nih.gov/gene/406982 (accessed on 23 November 2022).
  281. Gong, J.; Shen, Y.; Jiang, F.; Wang, Y.; Chu, L.; Sun, J.; Shen, P.; Chen, M. MicroRNA-20a Promotes Non-Small Cell Lung Cancer Proliferation by Upregulating PD-L1 by Targeting PTEN. Oncol. Lett. 2022, 23, 148. [Google Scholar] [CrossRef] [PubMed]
  282. Ren, T.; Fan, X.-X.; Wang, M.-F.; Duan, F.-G.; Wei, C.-L.; Li, R.-Z.; Jiang, Z.-B.; Wang, Y.-W.; Yao, X.-J.; Chen, M.-W.; et al. MiR-20b Promotes Growth of Non-Small Cell Lung Cancer through a Positive Feedback Loop of the Wnt/β-Catenin Signaling Pathway. Int. J. Oncol. 2019, 56, 470–479. [Google Scholar] [CrossRef] [PubMed]
  283. MIR130A MicroRNA 130a [Homo Sapiens (Human)]-Gene-NCBI. Available online: https://www.ncbi.nlm.nih.gov/gene/406919 (accessed on 23 November 2022).
  284. Zhang, Q.; Zhang, B.; Sun, L.; Yan, Q.; Zhang, Y.; Zhang, Z.; Su, Y.; Wang, C. MicroRNA-130b Targets PTEN to Induce Resistance to Cisplatin in Lung Cancer Cells by Activating Wnt/Β-catenin Pathway. Cell Biochem. Funct 2018, 36, 194–202. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  285. Wei, M.-C.; Wang, Y.-M.; Wang, D.-W. miR-130a-Mediated KLF3 Can Inhibit the Growth of Lung Cancer Cells. CMAR 2021, 13, 2995–3004. [Google Scholar] [CrossRef] [PubMed]
  286. MIR424 MicroRNA 424 [Homo Sapiens (Human)]-Gene-NCBI. Available online: https://www.ncbi.nlm.nih.gov/gene/494336 (accessed on 23 November 2022).
  287. Wang, Y.; Lv, Z.; Fu, J.; Wang, Z.; Fan, Z.; Lei, T. Endogenous MicroRNA-424 Predicts Clinical Outcome and Its Inhibition Acts as Cancer Suppressor in Human Non-Small Cell Lung Cancer. Biomed. Pharmacother. 2017, 89, 208–214. [Google Scholar] [CrossRef]
  288. Richardsen, E.; Andersen, S.; Al-Saad, S.; Rakaee, M.; Nordby, Y.; Pedersen, M.I.; Ness, N.; Ingebriktsen, L.M.; Fassina, A.; Taskén, K.A.; et al. Low Expression of MiR-424-3p Is Highly Correlated with Clinical Failure in Prostate Cancer. Sci. Rep. 2019, 9, 10662. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  289. Li, S.; Wu, Y.; Zhang, J.; Sun, H.; Wang, X. Role of MiRNA-424 in Cancers. Onco Targets Ther. 2020, 13, 9611–9622. [Google Scholar] [CrossRef]
  290. MIR136 MicroRNA 136 [Homo Sapiens (Human)]-Gene-NCBI. Available online: https://www.ncbi.nlm.nih.gov/gene/406927 (accessed on 23 November 2022).
  291. Shen, S.; Yue, H.; Li, Y.; Qin, J.; Li, K.; Liu, Y.; Wang, J. Upregulation of MiR-136 in Human Non-Small Cell Lung Cancer Cells Promotes Erk1/2 Activation by Targeting PPP2R2A. Tumour. Biol. 2014, 35, 631–640. [Google Scholar] [CrossRef]
  292. Li, T.; Gao, X.; Gao, L.; Gan, B.; Xie, Z.; Zeng, J.; Chen, G. Role of Upregulated MiR-136-5p in Lung Adenocarcinoma: A Study of 1242 Samples Utilizing Bioinformatics Analysis. Pathol. Res. Pract. 2018, 214, 750–766. [Google Scholar] [CrossRef]
  293. Gao, L.; Li, T.; Gan, B.; Gao, X.; Xie, Z.; He, R.; Mo, W.; Chi, X. MiR-136-5p Is Involved in the Pathogenesis of LUSC through Targeting MTDH: A Study Based on RT-QPCR, IHC, Public Database and Dual-Luciferase Reporter Assay. Int. J. Clin. Exp. Med. 2018, 10, 10417–10432. [Google Scholar]
  294. MIR301B MicroRNA 301b [Homo Sapiens (Human)]-Gene-NCBI. Available online: https://www.ncbi.nlm.nih.gov/gene/100126318 (accessed on 23 November 2022).
  295. Liu, H.; Ma, X.; Niu, N.; Zhao, J.; Lu, C.; Yang, F.; Qi, W. MIR-301b-3p Promotes Lung Adenocarcinoma Cell Proliferation, Migration and Invasion by Targeting DLC1. Technol. Cancer Res. Treat. 2021, 20, 1533033821990036. [Google Scholar] [CrossRef] [PubMed]
  296. Guo, Y.; Yao, B.; Zhu, Q.; Xiao, Z.; Hu, L.; Liu, X.; Li, L.; Wang, J.; Xu, Q.; Yang, L.; et al. MicroRNA-301b-3p Contributes to Tumour Growth of Human Hepatocellular Carcinoma by Repressing Vestigial like Family Member 4. J. Cell. Mol. Med. 2019, 23, 5037–5047. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  297. Li, P.; Xing, W.; Xu, J.; Yuan, D.; Liang, G.; Liu, B.; Ma, H. MicroRNA-301b-3p Downregulation Underlies a Novel Inhibitory Role of Long Non-Coding RNA MBNL1-AS1 in Non-Small Cell Lung Cancer. Stem Cell Res. Ther. 2019, 10, 144. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  298. Hirono, T.; Jingushi, K.; Kitae, K.; Nagata, T.; Sato, M.; Minami, K.; Aoki, M.; Harada Takeda, A.; Umehara, T.; Egawa, H.; et al. MiR-301a/b Function as OncomiRs in Non-Small-Cell Lung Cancer. Integr. Mol. Med. 2018, 5, 1–6. [Google Scholar] [CrossRef]
  299. Hanahan, D.; Weinberg, R.A. Hallmarks of Cancer: The Next Generation. Cell 2011, 144, 646–674. [Google Scholar] [CrossRef]
  300. Weston, A.; Harris, C.C. Multistage Carcinogenesis. In Holland-Frei Cancer Medicine, 6th ed.; PMPH USA, Ltd.: Raleigh, NC, USA, 2003. [Google Scholar]
  301. Stankovic, B.; Bjørhovde, H.A.K.; Skarshaug, R.; Aamodt, H.; Frafjord, A.; Müller, E.; Hammarström, C.; Beraki, K.; Bækkevold, E.S.; Woldbæk, P.R.; et al. Immune Cell Composition in Human Non-Small Cell Lung Cancer. Front. Immunol. 2019, 9, 3101. [Google Scholar] [CrossRef] [Green Version]
  302. Yoon, H.J.; Kang, J.; Park, H.; Sohn, I.; Lee, S.-H.; Lee, H.Y. Deciphering the Tumor Microenvironment through Radiomics in Non-Small Cell Lung Cancer: Correlation with Immune Profiles. PLoS ONE 2020, 15, e0231227. [Google Scholar] [CrossRef] [Green Version]
  303. Pennycuick, A.; Teixeira, V.H.; AbdulJabbar, K.; Raza, S.E.A.; Lund, T.; Akarca, A.U.; Rosenthal, R.; Kalinke, L.; Chandrasekharan, D.P.; Pipinikas, C.P.; et al. Immune Surveillance in Clinical Regression of Preinvasive Squamous Cell Lung Cancer. Cancer Discov. 2020, 10, 1489–1499. [Google Scholar] [CrossRef]
  304. Kunimasa, K.; Goto, T. Immunosurveillance and Immunoediting of Lung Cancer: Current Perspectives and Challenges. Int. J. Mol. Sci. 2020, 21, 597. [Google Scholar] [CrossRef] [Green Version]
  305. Bai, R.; Chen, N.; Li, L.; Du, N.; Bai, L.; Lv, Z.; Tian, H.; Cui, J. Mechanisms of Cancer Resistance to Immunotherapy. Front Oncol. 2020, 10, 1290. [Google Scholar] [CrossRef]
  306. Gure, A.O.; Chua, R.; Williamson, B.; Gonen, M.; Ferrera, C.A.; Gnjatic, S.; Ritter, G.; Simpson, A.J.G.; Chen, Y.-T.; Old, L.J.; et al. Cancer-Testis Genes Are Coordinately Expressed and Are Markers of Poor Outcome in Non-Small Cell Lung Cancer. Clin. Cancer Res. 2005, 11, 8055–8062. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  307. Grunwald, C.; Koslowski, M.; Arsiray, T.; Dhaene, K.; Praet, M.; Victor, A.; Morresi-Hauf, A.; Lindner, M.; Passlick, B.; Lehr, H.-A.; et al. Expression of Multiple Epigenetically Regulated Cancer/Germline Genes in Nonsmall Cell Lung Cancer. Int. J. Cancer 2006, 118, 2522–2528. [Google Scholar] [CrossRef] [PubMed]
  308. Palata, O.; Podzimkova Hradilova, N.; Mysiková, D.; Kutna, B.; Mrazkova, H.; Lischke, R.; Spisek, R.; Adkins, I. Detection of Tumor Antigens and Tumor-Antigen Specific T Cells in NSCLC Patients: Correlation of the Quality of T Cell Responses with NSCLC Subtype. Immunol. Lett. 2020, 219, 46–53. [Google Scholar] [CrossRef]
  309. Hanash, S.M.; Ostrin, E.J.; Fahrmann, J.F. Blood Based Biomarkers beyond Genomics for Lung Cancer Screening. Transl. Lung Cancer Res. 2018, 7, 327–335. [Google Scholar] [CrossRef]
  310. Li, N.; Holden, V.K.; Deepak, J.; Todd, N.W.; Jiang, F. Autoantibodies against Tumor-Associated Antigens in Sputum as Biomarkers for Lung Cancer. Transl. Oncol. 2021, 14, 100991. [Google Scholar] [CrossRef] [PubMed]
  311. Murphy, M.A.; O’Leary, J.J.; Cahill, D.J. Assessment of the Humoral Immune Response to Cancer. J. Proteom. 2012, 75, 4573–4579. [Google Scholar] [CrossRef]
  312. Zaenker, P.; Gray, E.S.; Ziman, M.R. Autoantibody Production in Cancer-The Humoral Immune Response toward Autologous Antigens in Cancer Patients. Autoimmun. Rev. 2016, 15, 477–483. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  313. Yang, D.; Zhang, X.; Powell, C.A.; Ni, J.; Wang, B.; Zhang, J.; Zhang, Y.; Wang, L.; Xu, Z.; Zhang, L.; et al. Probability of Cancer in High-Risk Patients Predicted by the Protein-Based Lung Cancer Biomarker Panel in China: LCBP Study. Cancer 2018, 124, 262–270. [Google Scholar] [CrossRef] [Green Version]
  314. Li, X.; Hayward, C.; Fong, P.-Y.; Dominguez, M.; Hunsucker, S.W.; Lee, L.W.; McLean, M.; Law, S.; Butler, H.; Schirm, M.; et al. A Blood-Based Proteomic Classifier for the Molecular Characterization of Pulmonary Nodules. Sci. Transl. Med. 2013, 5, 207ra142. [Google Scholar] [CrossRef] [Green Version]
  315. Pan, Y.W.; Zhou, Z.G.; Wang, M.; Dong, J.Q.; Du, K.P.; Li, S.; Liu, Y.L.; Lv, P.J.; Gao, J.B. Combination of IL-6, IL-10, and MCP-1 with Traditional Serum Tumor Markers in Lung Cancer Diagnosis and Prognosis. Genet Mol. Res. 2016, 15, 4210–4238. [Google Scholar] [CrossRef]
  316. Ostroff, R.M.; Bigbee, W.L.; Franklin, W.; Gold, L.; Mehan, M.; Miller, Y.E.; Pass, H.I.; Rom, W.N.; Siegfried, J.M.; Stewart, A.; et al. Unlocking Biomarker Discovery: Large Scale Application of Aptamer Proteomic Technology for Early Detection of Lung Cancer. PLoS ONE 2010, 5, e15003. [Google Scholar] [CrossRef] [Green Version]
  317. Farlow, E.C.; Patel, K.; Basu, S.; Lee, B.-S.; Kim, A.W.; Coon, J.S.; Faber, L.P.; Bonomi, P.; Liptay, M.J.; Borgia, J.A. Development of a Multiplexed Tumor-Associated Autoantibody-Based Blood Test for the Detection of Non-Small Cell Lung Cancer. Clin. Cancer Res. 2010, 16, 3452–3462. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  318. Schneider, J.; Bitterlich, N.; Velcovsky, H.-G.; Morr, H.; Katz, N.; Eigenbrodt, E. Fuzzy Logic-Based Tumor-Marker Profiles Improved Sensitivity in the Diagnosis of Lung Cancer. Int. J. Clin. Oncol. 2002, 7, 145–151. [Google Scholar] [CrossRef] [PubMed]
  319. Borgia, J.A.; Basu, S.; Faber, L.P.; Kim, A.W.; Coon, J.S.; Kaiser-Walters, K.A.; Fhied, C.; Thomas, S.; Rouhi, O.; Warren, W.H.; et al. Establishment of a Multi-Analyte Serum Biomarker Panel to Identify Lymph Node Metastases in Non-Small Cell Lung Cancer. J. Thorac. Oncol. 2009, 4, 338–347. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  320. Wu, Y.; Wu, Y.; Wang, J.; Yan, Z.; Qu, L.; Xiang, B.; Zhang, Y. An Optimal Tumor Marker Group-Coupled Artificial Neural Network for Diagnosis of Lung Cancer. Expert Syst. Appl. 2011, 38, 11329–11334. [Google Scholar] [CrossRef]
  321. Feng, F.; Wu, Y.; Wu, Y.; Nie, G.; Ni, R. The Effect of Artificial Neural Network Model Combined with Six Tumor Markers in Auxiliary Diagnosis of Lung Cancer. J. Med. Syst. 2012, 36, 2973–2980. [Google Scholar] [CrossRef] [PubMed]
  322. Flores-Fernández, J.M.; Herrera-López, E.J.; Sánchez-Llamas, F.; Rojas-Calvillo, A.; Cabrera-Galeana, P.A.; Leal-Pacheco, G.; González-Palomar, M.G.; Femat, R.; Martínez-Velázquez, M. Development of an Optimized Multi-Biomarker Panel for the Detection of Lung Cancer Based on Principal Component Analysis and Artificial Neural Network Modeling. Expert Syst. Appl. 2012, 39, 10851–10856. [Google Scholar] [CrossRef]
  323. Louis, E.; Adriaensens, P.; Guedens, W.; Bigirumurame, T.; Baeten, K.; Vanhove, K.; Vandeurzen, K.; Darquennes, K.; Vansteenkiste, J.; Dooms, C.; et al. Detection of Lung Cancer through Metabolic Changes Measured in Blood Plasma. J. Thorac. Oncol. 2016, 11, 516–523. [Google Scholar] [CrossRef] [Green Version]
  324. Fahrmann, J.F.; Kim, K.; DeFelice, B.C.; Taylor, S.L.; Gandara, D.R.; Yoneda, K.Y.; Cooke, D.T.; Fiehn, O.; Kelly, K.; Miyamoto, S. Investigation of Metabolomic Blood Biomarkers for Detection of Adenocarcinoma Lung Cancer. Cancer Epidemiol Biomark. Prev. 2015, 24, 1716–1723. [Google Scholar] [CrossRef] [Green Version]
  325. Flores, J.M.; Herrera, E.; Leal, G.; González, M.G.; Sánchez, F.; Rojas, A.; Cabrera, P.A.; Femat, R.; Martínez-Velázquez, M. Artificial Neural Network-Based Serum Biomarkers Analysis Improves Sensitivity in the Diagnosis of Lung Cancer. In Proceedings of the V Latin American Congress on Biomedical Engineering CLAIB 2011, 16–21 May 2011,Habana, Cuba; Folgueras Méndez, J., Aznielle Rodríguez, T.Y., Calderón Marín, C.F., Llanusa Ruiz, S.B., Castro Medina, J., Vega Vázquez, H., Carballo Barreda, M., Rodríguez Rojas, R., Eds.; Springer: Berlin/Heidelberg, Germany, 2011; pp. 882–885. [Google Scholar]
  326. Chapman, C.J.; Healey, G.F.; Murray, A.; Boyle, P.; Robertson, C.; Peek, L.J.; Allen, J.; Thorpe, A.J.; Hamilton-Fairley, G.; Parsy-Kowalska, C.B.; et al. EarlyCDT®-Lung Test: Improved Clinical Utility through Additional Autoantibody Assays. Tumor Biol. 2012, 33, 1319–1326. [Google Scholar] [CrossRef] [Green Version]
  327. Li, P.; Shi, J.-X.; Xing, M.-T.; Dai, L.-P.; Li, J.-T.; Zhang, J.-Y. Evaluation of Serum Autoantibodies against Tumor-Associated Antigens as Biomarkers in Lung Cancer. Tumour Biol. 2017, 39, 1010428317711662. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  328. Dai, L.; Li, J.; Tsay, J.-C.J.; Yie, T.-A.; Munger, J.S.; Pass, H.; Rom, W.N.; Tan, E.M.; Zhang, J.-Y. Identification of Autoantibodies to ECH1 and HNRNPA2B1 as Potential Biomarkers in the Early Detection of Lung Cancer. OncoImmunology 2017, 6, e1310359. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  329. Dai, L.; Tsay, J.-C.J.; Li, J.; Yie, T.-A.; Munger, J.S.; Pass, H.; Rom, W.N.; Zhang, Y.; Tan, E.M.; Zhang, J.-Y. Autoantibodies against Tumor-Associated Antigens in the Early Detection of Lung Cancer. Lung Cancer 2016, 99, 172–179. [Google Scholar] [CrossRef] [PubMed]
  330. Full Article: Serum Anti-MDM2 and Anti-c-Myc Autoantibodies as Biomarkers in the Early Detection of Lung Cancer. Available online: https://www.tandfonline.com/doi/full/10.1080/2162402X.2016.1138200 (accessed on 23 November 2022).
  331. Li, P.; Shi, J.-X.; Dai, L.-P.; Chai, Y.-R.; Zhang, H.-F.; Kankonde, M.; Kankonde, P.; Yu, B.-F.; Zhang, J.-Y. Serum Anti-MDM2 and Anti-c-Myc Autoantibodies as Biomarkers in the Early Detection of Lung Cancer. OncoImmunology 2016, 5, e1138200. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  332. Miyamoto, S.; Taylor, S.L.; Barupal, D.K.; Taguchi, A.; Wohlgemuth, G.; Wikoff, W.R.; Yoneda, K.Y.; Gandara, D.R.; Hanash, S.M.; Kim, K.; et al. Systemic Metabolomic Changes in Blood Samples of Lung Cancer Patients Identified by Gas Chromatography Time-of-Flight Mass Spectrometry. Metabolites 2015, 5, 192–210. [Google Scholar] [CrossRef] [PubMed]
  333. Lam, S.; Boyle, P.; Healey, G.F.; Maddison, P.; Peek, L.; Murray, A.; Chapman, C.J.; Allen, J.; Wood, W.C.; Sewell, H.F.; et al. EarlyCDT-Lung: An Immunobiomarker Test as an Aid to Early Detection of Lung Cancer. Cancer Prev. Res. (Phila) 2011, 4, 1126–1134. [Google Scholar] [CrossRef] [Green Version]
  334. Ros-Mazurczyk, M.; Jelonek, K.; Marczyk, M.; Binczyk, F.; Pietrowska, M.; Polanska, J.; Dziadziuszko, R.; Jassem, J.; Rzyman, W.; Widlak, P. Serum Lipid Profile Discriminates Patients with Early Lung Cancer from Healthy Controls. Lung Cancer 2017, 112, 69–74. [Google Scholar] [CrossRef]
  335. Klupczynska, A.; Dereziński, P.; Garrett, T.J.; Rubio, V.Y.; Dyszkiewicz, W.; Kasprzyk, M.; Kokot, Z.J. Study of Early Stage Non-Small-Cell Lung Cancer Using Orbitrap-Based Global Serum Metabolomics. J. Cancer Res. Clin. Oncol. 2017, 143, 649–659. [Google Scholar] [CrossRef] [Green Version]
  336. Campa, M.J.; Gottlin, E.B.; Herndon, J.E.; Patz, E.F. Rethinking Autoantibody Signature Panels for Cancer Diagnosis. J. Thorac. Oncol. 2017, 12, 1011–1014. [Google Scholar] [CrossRef] [Green Version]
  337. Tran, E.; Robbins, P.F.; Lu, Y.-C.; Prickett, T.D.; Gartner, J.J.; Jia, L.; Pasetto, A.; Zheng, Z.; Ray, S.; Groh, E.M.; et al. T-Cell Transfer Therapy Targeting Mutant KRAS in Cancer. New Engl. J. Med. 2016, 375, 2255–2262. [Google Scholar] [CrossRef] [Green Version]
  338. Tran, E.; Turcotte, S.; Gros, A.; Robbins, P.F.; Lu, Y.-C.; Dudley, M.E.; Wunderlich, J.R.; Somerville, R.P.; Hogan, K.; Hinrichs, C.S.; et al. Cancer Immunotherapy Based on Mutation-Specific CD4+ T Cells in a Patient with Epithelial Cancer. Science 2014, 344, 641–645. [Google Scholar] [CrossRef] [PubMed]
  339. Ye, L.; Creaney, J.; Redwood, A.; Robinson, B. The Current Lung Cancer Neoantigen Landscape and Implications for Therapy. J. Thorac. Oncol. 2021, 16, 922–932. [Google Scholar] [CrossRef] [PubMed]
  340. Fehlings, M.; Nardin, A.; Jhunjhunwala, S. Late-differentiated effector neoantigen-specific CD8+ T cells are enriched in non-small cell lung carcinoma patients responding to atezolizumab treatment. Eur. J. Cancer 2019, 110, S2–S3. [Google Scholar] [CrossRef]
  341. McGranahan, N.; Furness, A.J.S.; Rosenthal, R.; Ramskov, S.; Lyngaa, R.; Saini, S.K.; Jamal-Hanjani, M.; Wilson, G.A.; Birkbak, N.J.; Hiley, C.T.; et al. Clonal Neoantigens Elicit T Cell Immunoreactivity and Sensitivity to Immune Checkpoint Blockade. Science 2016, 351, 1463–1469. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  342. Rizvi, N.A.; Hellmann, M.D.; Snyder, A.; Kvistborg, P.; Makarov, V.; Havel, J.J.; Lee, W.; Yuan, J.; Wong, P.; Ho, T.S.; et al. Mutational Landscape Determines Sensitivity to PD-1 Blockade in Non-Small Cell Lung Cancer. Science 2015, 348, 124–128. [Google Scholar] [CrossRef]
  343. Wells, D.K.; van Buuren, M.M.; Dang, K.K.; Hubbard-Lucey, V.M.; Sheehan, K.C.F.; Campbell, K.M.; Lamb, A.; Ward, J.P.; Sidney, J.; Blazquez, A.B.; et al. Key Parameters of Tumor Epitope Immunogenicity Revealed Through a Consortium Approach Improve Neoantigen Prediction. Cell 2020, 183, 818–834. [Google Scholar] [CrossRef]
  344. Karasaki, T.; Nagayama, K.; Kawashima, M.; Hiyama, N.; Murayama, T.; Kuwano, H.; Nitadori, J.; Anraku, M.; Sato, M.; Miyai, M.; et al. Identification of Individual Cancer-Specific Somatic Mutations for Neoantigen-Based Immunotherapy of Lung Cancer. J. Thorac. Oncol. 2016, 11, 324–333. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  345. Okada, M.; Shimizu, K.; Iyoda, T.; Ueda, S.; Shinga, J.; Mochizuki, Y.; Watanabe, T.; Ohara, O.; Fujii, S. PD-L1 Expression Affects Neoantigen Presentation. iScience 2020, 23, 101238. [Google Scholar] [CrossRef]
  346. Niu, M.; Yi, M.; Li, N.; Luo, S.; Wu, K. Predictive Biomarkers of Anti-PD-1/PD-L1 Therapy in NSCLC. Exp. Hematol. Oncol. 2021, 10, 18. [Google Scholar] [CrossRef]
  347. He, X.; Xu, C. Immune Checkpoint Signaling and Cancer Immunotherapy. Cell Res. 2020, 30, 660–669. [Google Scholar] [CrossRef]
  348. McBride, M.A.; Patil, T.K.; Bohannon, J.K.; Hernandez, A.; Sherwood, E.R.; Patil, N.K. Immune Checkpoints: Novel Therapeutic Targets to Attenuate Sepsis-Induced Immunosuppression. Front. Immunol. 2021, 11, 624272. [Google Scholar] [CrossRef] [PubMed]
  349. Onoi, K.; Chihara, Y.; Uchino, J.; Shimamoto, T.; Morimoto, Y.; Iwasaku, M.; Kaneko, Y.; Yamada, T.; Takayama, K. Immune Checkpoint Inhibitors for Lung Cancer Treatment: A Review. J. Clin. Med. 2020, 9, 1362. [Google Scholar] [CrossRef] [PubMed]
  350. 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]
  351. Wang, C.; Li, J.; Zhang, Q.; Wu, J.; Xiao, Y.; Song, L.; Gong, H.; Li, Y. The Landscape of Immune Checkpoint Inhibitor Therapy in Advanced Lung Cancer. BMC Cancer 2021, 21, 968. [Google Scholar] [CrossRef]
  352. Domagała-Kulawik, J. Immune Checkpoint Inhibitors in Non-Small Cell Lung Cancer—Towards Daily Practice. Adv. Respir. Med. 2018, 86, 142–148. [Google Scholar] [CrossRef] [PubMed]
  353. Huang, Z.; Su, W.; Lu, T.; Wang, Y.; Dong, Y.; Qin, Y.; Liu, D.; Sun, L.; Jiao, W. First-Line Immune-Checkpoint Inhibitors in Non-Small Cell Lung Cancer: Current Landscape and Future Progress. Front. Pharmacol. 2020, 11, 578091. [Google Scholar] [CrossRef]
  354. Xiong, W.; Zhao, Y.; Du, H.; Guo, X. Current Status of Immune Checkpoint Inhibitor Immunotherapy for Lung Cancer. Front. Oncol. 2021, 11, 704336. [Google Scholar] [CrossRef]
  355. Ma, L.-R.; Li, J.-X.; Tang, L.; Li, R.-Z.; Yang, J.-S.; Sun, A.; Leung, E.L.-H.; Yan, P.-Y. Immune Checkpoints and Immunotherapy in Non-small Cell Lung Cancer: Novel Study Progression, Challenges and Solutions (Review). Oncol. Lett. 2021, 22, 787. [Google Scholar] [CrossRef]
  356. Tsoukalas, N.; Kiakou, M.; Tsapakidis, K.; Tolia, M.; Baxevanos, P.; Kyrgias, G.; Theocharis, S. PD-1 and PD-L1 as Immunotherapy Targets and Biomarkers in Non-Small Cell Lung Cancer. J. Buon 2019, 24, 883–888. [Google Scholar]
  357. Abu Hejleh, T.; Furqan, M.; Ballas, Z.; Clamon, G. The Clinical Significance of Soluble PD-1 and PD-L1 in Lung Cancer. Crit. Rev. Oncol. /Hematol. 2019, 143, 148–152. [Google Scholar] [CrossRef]
  358. 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] [PubMed] [Green Version]
  359. Zhang, Q.; Tang, L.; Zhou, Y.; He, W.; Li, W. Immune Checkpoint Inhibitor-Associated Pneumonitis in Non-Small Cell Lung Cancer: Current Understanding in Characteristics, Diagnosis, and Management. Front. Immunol. 2021, 12, 1889. [Google Scholar] [CrossRef] [PubMed]
  360. Reck, M.; Borghaei, H.; O’Byrne, K.J. Nivolumab plus Ipilimumab in Non-Small-Cell Lung Cancer. Future Oncol. 2019, 15, 2287–2302. [Google Scholar] [CrossRef] [PubMed]
  361. Shiravand, Y.; Khodadadi, F.; Kashani, S.M.A.; Hosseini-Fard, S.R.; Hosseini, S.; Sadeghirad, H.; Ladwa, R.; O’Byrne, K.; Kulasinghe, A. Immune Checkpoint Inhibitors in Cancer Therapy. Curr. Oncol. 2022, 29, 3044–3060. [Google Scholar] [CrossRef]
  362. ADORA2A-Adenosine Receptor A2a-Homo Sapiens (Human) | UniProtKB | UniProt. Available online: https://www.uniprot.org/uniprotkb/P29274/entry (accessed on 23 November 2022).
  363. Leone, R.D.; Lo, Y.-C.; Powell, J.D. A2aR Antagonists: Next Generation Checkpoint Blockade for Cancer Immunotherapy. Comput. Struct. Biotechnol. J. 2015, 13, 265–272. [Google Scholar] [CrossRef] [Green Version]
  364. Pardoll, D.M. The Blockade of Immune Checkpoints in Cancer Immunotherapy. Nat. Rev. Cancer 2012, 12, 252–264. [Google Scholar] [CrossRef]
  365. Hinz, S.; Navarro, G.; Borroto-Escuela, D.; Seibt, B.F.; Ammon, Y.-C.; de Filippo, E.; Danish, A.; Lacher, S.K.; Červinková, B.; Rafehi, M.; et al. Adenosine A2A Receptor Ligand Recognition and Signaling Is Blocked by A2B Receptors. Oncotarget 2018, 9, 13593–13611. [Google Scholar] [CrossRef] [Green Version]
  366. VSIR-V-Type Immunoglobulin Domain-Containing Suppressor of T-Cell Activation-Homo Sapiens (Human) | UniProtKB | UniProt. Available online: https://www.uniprot.org/uniprotkb/Q9H7M9/entry (accessed on 23 November 2022).
  367. Huang, X.; Zhang, X.; Li, E.; Zhang, G.; Wang, X.; Tang, T.; Bai, X.; Liang, T. VISTA: An Immune Regulatory Protein Checking Tumor and Immune Cells in Cancer Immunotherapy. J. Hematol. Oncol. 2020, 13, 83. [Google Scholar] [CrossRef]
  368. CD276-CD276 Antigen-Homo Sapiens (Human) | UniProtKB | UniProt. Available online: https://www.uniprot.org/uniprotkb/Q5ZPR3/entry (accessed on 23 November 2022).
  369. Aicher, W.K.; Korn, M.; Reitnauer, L.; Maurer, F.B.; Hennenlotter, J.; Black, P.C.; Todenhofer, T.; Bedke, J.; Stenzl, A. Expression Patterns of the Immune Checkpoint Ligand CD276 in Urothelial Carcinoma. BMC Urol. 2021, 21, 60. [Google Scholar] [CrossRef]
  370. Li, F.; Chen, H.; Wang, D. Silencing of CD276 Suppresses Lung Cancer Progression by Regulating Integrin Signaling. J. Thorac. Dis. 2020, 12, 2137–2145. [Google Scholar] [CrossRef]
  371. Zhang, C.; Hao, X. Prognostic Significance of CD276 in Non-Small Cell Lung Cancer. Open Med. (Wars) 2019, 14, 805–812. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  372. CD274-Programmed Cell Death 1 Ligand 1-Homo Sapiens (Human) | UniProtKB | UniProt. Available online: https://www.uniprot.org/uniprotkb/Q9NZQ7/entry (accessed on 23 November 2022).
  373. PDCD1LG2-Programmed Cell Death 1 Ligand 2-Homo Sapiens (Human) | UniProtKB | UniProt. Available online: https://www.uniprot.org/uniprotkb/Q9BQ51/entry (accessed on 23 November 2022).
  374. Larsen, T.V.; Hussmann, D.; Nielsen, A.L. PD-L1 and PD-L2 Expression Correlated Genes in Non-Small-Cell Lung Cancer. Cancer Commun. (Lond) 2019, 39, 30. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  375. Ludovini, V.; Bianconi, F.; Siggillino, A.; Vannucci, J.; Baglivo, S.; Berti, V.; Tofanetti, F.R.; Reda, M.S.; Bellezza, G.; Mandarano, M.; et al. High PD-L1/IDO-2 and PD-L2/IDO-1 Co-Expression Levels Are Associated with Worse Overall Survival in Resected Non-Small Cell Lung Cancer Patients. Genes (Basel) 2021, 12, 273. [Google Scholar] [CrossRef] [PubMed]
  376. Kwiecień, I.; Rutkowska, E.; Polubiec-Kownacka, M.; Raniszewska, A.; Rzepecki, P.; Domagała-Kulawik, J. Identification of PD-1 Ligands: PD-L1 and PD-L2 on Macrophages in Lung Cancer Milieu by Flow Cytometry. Transl. Lung Cancer Res. 2021, 10, 1679. [Google Scholar] [CrossRef]
  377. PDCD1-Programmed Cell Death Protein 1-Homo Sapiens (Human) | UniProtKB | UniProt. Available online: https://www.uniprot.org/uniprotkb/Q15116/entry (accessed on 23 November 2022).
  378. Santini, F.C.; Hellmann, M.D. PD-1/PD-L1 Axis in Lung Cancer. Cancer J. 2018, 24, 15–19. [Google Scholar] [CrossRef] [PubMed]
  379. CD80-CD80 Antigen (CD28 Antigen Ligand 1, B7-1 Antigen), Isoform CRA_a-Homo Sapiens (Human) | UniProtKB | UniProt. Available online: https://www.uniprot.org/uniprotkb/A0N0P2/entry (accessed on 23 November 2022).
  380. Sato, T.; Takagi, K.; Higuchi, M.; Abe, H.; Kojimahara, M.; Sagawa, M.; Tanaki, M.; Miki, Y.; Suzuki, T.; Hojo, H. Immunolocalization of CD80 and CD86 in Non-Small Cell Lung Carcinoma: CD80 as a Potent Prognostic Factor. Acta Histochem. Cytochem. 2022, 55, 25–35. [Google Scholar] [CrossRef]
  381. Vackova, J.; Polakova, I.; Johari, S.D.; Smahel, M. CD80 Expression on Tumor Cells Alters Tumor Microenvironment and Efficacy of Cancer Immunotherapy by CTLA-4 Blockade. Cancers 2021, 13, 1935. [Google Scholar] [CrossRef]
  382. Qiu, H.; Tian, W.; He, Y.; Li, J.; He, C.; Li, Y.; Liu, N.; Li, J. Integrated Analysis Reveals Prognostic Value and Immune Correlates of CD86 Expression in Lower Grade Glioma. Front. Oncol. 2021, 11, 654350. [Google Scholar] [CrossRef]
  383. CD86-CD86 Antigen-Homo Sapiens (Human) | UniProtKB | UniProt. Available online: https://www.uniprot.org/uniprotkb/A0A0X9R4E0/entry (accessed on 23 November 2022).
  384. CTLA4-Cytotoxic T-Lymphocyte Protein 4-Homo Sapiens (Human) | UniProtKB | UniProt. Available online: https://www.uniprot.org/uniprotkb/P16410/entry (accessed on 23 November 2022).
  385. Paulsen, E.-E.; Kilvaer, T.K.; Rakaee, M.; Richardsen, E.; Hald, S.M.; Andersen, S.; Busund, L.-T.; Bremnes, R.M.; Donnem, T. CTLA-4 Expression in the Non-Small Cell Lung Cancer Patient Tumor Microenvironment: Diverging Prognostic Impact in Primary Tumors and Lymph Node Metastases. Cancer Immunol. Immunother. 2017, 66, 1449–1461. [Google Scholar] [CrossRef] [Green Version]
  386. Shen, K.; Cui, J.; Wei, Y.; Chen, X.; Liu, G.; Gao, X.; Li, W.; Lu, H.; Zhan, P.; Lv, T.; et al. Effectiveness and Safety of PD-1/PD-L1 or CTLA4 Inhibitors Combined with Chemotherapy as a First-Line Treatment for Lung Cancer: A Meta-Analysis. J. Thorac. Dis. 2018, 10, 6636. [Google Scholar] [CrossRef]
  387. LGALS9-Galectin-9-Homo Sapiens (Human) | UniProtKB | UniProt. Available online: https://www.uniprot.org/uniprotkb/O00182/entry (accessed on 23 November 2022).
  388. He, Y.; Jia, K.; Dziadziuszko, R.; Zhao, S.; Zhang, X.; Deng, J.; Wang, H.; Hirsch, F.R.; Zhou, C. Galectin-9 in Non-Small Cell Lung Cancer. Lung Cancer 2019, 136, 80–85. [Google Scholar] [CrossRef] [PubMed]
  389. Chen, P.; Zhang, L.; Zhang, W.; Sun, C.; Wu, C.; He, Y.; Zhou, C. Galectin-9-Based Immune Risk Score Model Helps to Predict Relapse in Stage I-III Small Cell Lung Cancer. J. Immunother. Cancer 2020, 8, e001391. [Google Scholar] [CrossRef] [PubMed]
  390. HAVCR2-Hepatitis A Virus Cellular Receptor 2-Homo Sapiens (Human) | UniProtKB | UniProt. Available online: https://www.uniprot.org/uniprotkb/Q8TDQ0/entry (accessed on 23 November 2022).
  391. Zhuang, X.; Zhang, X.; Xia, X.; Zhang, C.; Liang, X.; Gao, L.; Zhang, X.; Ma, C. Ectopic Expression of TIM-3 in Lung Cancers: A Potential Independent Prognostic Factor for Patients with NSCLC. Am. J. Clin. Pathol. 2012, 137, 978–985. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  392. Zang, K.; Hui, L.; Wang, M.; Huang, Y.; Zhu, X.; Yao, B. TIM-3 as a Prognostic Marker and a Potential Immunotherapy Target in Human Malignant Tumors: A Meta-Analysis and Bioinformatics Validation. Front. Oncol. 2021, 11, 579351. [Google Scholar] [CrossRef] [PubMed]
  393. TNFRSF14-Tumor Necrosis Factor Receptor Superfamily Member 14-Homo Sapiens (Human) | UniProtKB | UniProt. Available online: https://www.uniprot.org/uniprotkb/Q92956/entry (accessed on 23 November 2022).
  394. Ren, S.; Tian, Q.; Amar, N.; Yu, H.; Rivard, C.J.; Caldwell, C.; Ng Terry, L.; Tu, M.; Liu, Y.; Gao, D.; et al. The Immune Checkpoint, HVEM May Contribute to Immune Escape in Non-Small Cell Lung Cancer Lacking PD-L1 Expression. Lung Cancer (Amsterdam, Netherlands) 2018, 125, 115–120. [Google Scholar] [CrossRef]
  395. Demerlé, C.; Gorvel, L.; Olive, D. BTLA-HVEM Couple in Health and Diseases: Insights for Immunotherapy in Lung Cancer. Front Oncol. 2021, 11, 682007. [Google Scholar] [CrossRef]
  396. BTLA-B-and T-Lymphocyte Attenuator-Homo Sapiens (Human) | UniProtKB | UniProt. Available online: https://www.uniprot.org/uniprotkb/Q7Z6A9/entry (accessed on 23 November 2022).
  397. Li, X.; Xu, Z.; Cui, G.; Yu, L.; Zhang, X. BTLA Expression in Stage I–III Non–Small-Cell Lung Cancer and Its Correlation with PD-1/PD-L1 and Clinical Outcomes. Onco Targets Ther. 2020, 13, 215–224. [Google Scholar] [CrossRef] [Green Version]
  398. LAG3-Lymphocyte Activation Gene 3 Protein-Homo Sapiens (Human) | UniProtKB | UniProt. Available online: https://www.uniprot.org/uniprotkb/P18627/entry (accessed on 23 November 2022).
  399. Sun, H.; Dai, J.; Zhao, L.; Zhu, J.; Wang, H.; Chen, P.; Lu, H.; Chen, Q.; Zhang, Z. Lymphocyte Activation Gene-3 Is Associated with Programmed Death-Ligand 1 and Programmed Cell Death Protein 1 in Small Cell Lung Cancer. Ann. Transl. Med. 2021, 9, 1468. [Google Scholar] [CrossRef]
  400. 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]
  401. He, Y.; Rozeboom, L.; Rivard, C.J.; Ellison, K.; Dziadziuszko, R.; Yu, H.; Zhou, C.; Hirsch, F.R. MHC Class II Expression in Lung Cancer. Lung Cancer 2017, 112, 75–80. [Google Scholar] [CrossRef]
  402. Wang, H.; Zhao, S.; Zhang, X.; Jia, K.; Deng, J.; Zhou, C.; He, Y. Major Histocompatibility Complex Class II Molecule in Non-Small Cell Lung Cancer Diagnosis, Prognosis and Treatment. Onco Targets Ther. 2019, 12, 7281–7288. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  403. TNFSF4-Tumor Necrosis Factor Ligand Superfamily Member 4-Homo Sapiens (Human) | UniProtKB | UniProt. Available online: https://www.uniprot.org/uniprotkb/P23510/entry (accessed on 23 November 2022).
  404. Chen, P.; Wang, H.; Zhao, L.; Guo, H.; Zhang, L.; Zhang, W.; Sun, C.; Zhao, S.; Li, W.; Zhu, J.; et al. Immune Checkpoints OX40 and OX40L in Small-Cell Lung Cancer: Predict Prognosis and Modulate Immune Microenvironment. Front Oncol. 2021, 11, 713853. [Google Scholar] [CrossRef]
  405. Zhang, X.; He, Y.; Hirsch, F.R.; Zhou, C. OX40/OX40L Protein Expression in Non-Small Cell Lung Cancer and Its Role in Clinical Outcome and Relationships with Other Immune Biomarkers. Ann. Oncol. 2019, 30, v51. [Google Scholar] [CrossRef]
  406. TNFRSF4-Tumor Necrosis Factor Receptor Superfamily Member 4-Homo Sapiens (Human) | UniProtKB | UniProt. Available online: https://www.uniprot.org/uniprotkb/P43489/entry (accessed on 23 November 2022).
  407. CD40-Homo Sapiens (Human) | UniProtKB | UniProt. Available online: https://www.uniprot.org/uniprotkb/Q09LL4/entry (accessed on 23 November 2022).
  408. CD40LG-CD40 Ligand-Homo Sapiens (Human) | UniProtKB | UniProt. Available online: https://www.uniprot.org/uniprotkb/P29965/entry (accessed on 23 November 2022).
  409. Roselli, M.; Mineo, T.C.; Basili, S.; Martini, F.; Mariotti, S.; Aloe, S.; Del Monte, G.; Ambrogi, V.; Spila, A.; Palmirotta, R.; et al. Soluble CD40 Ligand Plasma Levels in Lung Cancer. Clin. Cancer Res. 2004, 10, 610–614. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  410. Xi, W.; Ke, D.; Min, L.; Lin, W.; Jiahui, Z.; Fang, L.; Zhaowei, G.; Zhe, Z.; Xi, C.; Huizhong, Z. Incorporation of CD40 Ligand Enhances the Immunogenicity of Tumor-associated Calcium Signal Transducer 2 Virus-like Particles against Lung Cancer. Int. J. Mol. Med. 2018, 41, 3671–3679. [Google Scholar] [CrossRef] [PubMed]
  411. ICOSLG-ICOS Ligand-Homo Sapiens (Human) | UniProtKB | UniProt. Available online: https://www.uniprot.org/uniprotkb/O75144/entry (accessed on 23 November 2022).
  412. Zang, X.; Allison, J.P. The B7 Family and Cancer Therapy: Costimulation and Coinhibition. Clin. Cancer Res. 2007, 13, 5271–5279. [Google Scholar] [CrossRef]
  413. Wu, G.; He, M.; Ren, K.; Ma, H.; Xue, Q. Inducible Co-Stimulator ICOS Expression Correlates with Immune Cell Infiltration and Can Predict Prognosis in Lung Adenocarcinoma. Int. J. Gen. Med. 2022, 15, 3739–3751. [Google Scholar] [CrossRef]
  414. ICOS-Inducible T-Cell Costimulator-Homo Sapiens (Human) | UniProtKB | UniProt. Available online: https://www.uniprot.org/uniprotkb/Q9Y6W8/entry (accessed on 23 November 2022).
  415. Marinelli, O.; Nabissi, M.; Morelli, M.B.; Torquati, L.; Amantini, C.; Santoni, G. ICOS-L as a Potential Therapeutic Target for Cancer Immunotherapy. Curr. Protein Pept. Sci. 2018, 19, 1107–1113. [Google Scholar] [CrossRef]
  416. CD70-CD70 Antigen-Homo Sapiens (Human) | UniProtKB | UniProt. Available online: https://www.uniprot.org/uniprotkb/P32970/entry (accessed on 23 November 2022).
  417. Flieswasser, T.; Van den Eynde, A.; Van Audenaerde, J.; De Waele, J.; Lardon, F.; Riether, C.; de Haard, H.; Smits, E.; Pauwels, P.; Jacobs, J. The CD70-CD27 Axis in Oncology: The New Kids on the Block. J. Exp. Clin. Cancer Res. 2022, 41, 12. [Google Scholar] [CrossRef]
  418. Jacobs, J.; Zwaenepoel, K.; Rolfo, C.; Van den Bossche, J.; Deben, C.; Silence, K.; Hermans, C.; Smits, E.; Van Schil, P.; Lardon, F.; et al. Unlocking the Potential of CD70 as a Novel Immunotherapeutic Target for Non-Small Cell Lung Cancer. Oncotarget 2015, 6, 13462–13475. [Google Scholar] [CrossRef] [Green Version]
  419. CD27-CD27 Antigen-Homo Sapiens (Human) | UniProtKB | UniProt. Available online: https://www.uniprot.org/uniprotkb/P26842/entry (accessed on 23 November 2022).
  420. Starzer, A.M.; Berghoff, A.S. New Emerging Targets in Cancer Immunotherapy: CD27 (TNFRSF7). ESMO Open 2020, 4, e000629. [Google Scholar] [CrossRef] [Green Version]
  421. Kashima, J.; Okuma, Y.; Hosomi, Y.; Hishima, T. High Serum Soluble CD27 Level Correlates with Poor Performance Status and Reduced Survival in Patients with Advanced Lung Cancer. Oncology 2019, 97, 365–372. [Google Scholar] [CrossRef]
  422. TNFSF18-Tumor Necrosis Factor Ligand Superfamily Member 18-Homo Sapiens (Human) | UniProtKB | UniProt. Available online: https://www.uniprot.org/uniprotkb/Q9UNG2/entry (accessed on 23 November 2022).
  423. TNFRSF18-Tumor Necrosis Factor Receptor Superfamily Member 18-Homo Sapiens (Human) | UniProtKB | UniProt. Available online: https://www.uniprot.org/uniprotkb/Q9Y5U5/entry (accessed on 23 November 2022).
  424. Zhu, L.X.; Davoodi, M.; Srivastava, M.K.; Kachroo, P.; Lee, J.M.; St. John, M.; Harris-White, M.; Huang, M.; Strieter, R.M.; Dubinett, S.; et al. GITR Agonist Enhances Vaccination Responses in Lung Cancer. Oncoimmunology 2015, 4, e992237. [Google Scholar] [CrossRef] [Green Version]
  425. Kopru, C.Z.; Cagnan, I.; Akar, I.; Esendagli, G.; Korkusuz, P.; Gunel-Ozcan, A. Dual Effect of Glucocorticoid-Induced Tumor Necrosis Factor–Related Receptor Ligand Carrying Mesenchymal Stromal Cells on Small Cell Lung Cancer: A Preliminary in Vitro Study. Cytotherapy 2018, 20, 930–940. [Google Scholar] [CrossRef]
  426. TNFSF9-Tumor Necrosis Factor Ligand Superfamily Member 9-Homo Sapiens (Human) | UniProtKB | UniProt. Available online: https://www.uniprot.org/uniprotkb/P41273/entry (accessed on 23 November 2022).
  427. Qu, Q.; Zhu, X.; Du, W.; Wang, H.; Shen, Y.; Zhu, Y.; Chen, C. 4-1BB Agonism Combined With PD-L1 Blockade Increases the Number of Tissue-Resident CD8+ T Cells and Facilitates Tumor Abrogation. Front. Immunol. 2020, 11, 577. [Google Scholar] [CrossRef]
  428. Vinay, D.S.; Kwon, B.S. 4-1BB (CD137), an Inducible Costimulatory Receptor, as a Specific Target for Cancer Therapy. BMB Rep. 2014, 47, 122–129. [Google Scholar] [CrossRef]
  429. Shah, P.; Forget, M.-A.; Frank, M.L.; Jiang, P.; Sakellariou-Thompson, D.; Federico, L.; Khairullah, R.; Neutzler, C.A.; Wistuba, I.; Chow, C.-W.B.; et al. Combined IL-2, Agonistic CD3 and 4-1BB Stimulation Preserve Clonotype Hierarchy in Propagated Non-Small Cell Lung Cancer Tumor-Infiltrating Lymphocytes. J. Immunother. Cancer 2022, 10, e003082. [Google Scholar] [CrossRef]
  430. TNFRSF9-Tumor Necrosis Factor Receptor Superfamily Member 9-Homo Sapiens (Human) | UniProtKB | UniProt. Available online: https://www.uniprot.org/uniprotkb/Q07011/entry (accessed on 23 November 2022).
  431. PVR-Poliovirus Receptor-Homo Sapiens (Human) | UniProtKB | UniProt. Available online: https://www.uniprot.org/uniprotkb/P15151/entry (accessed on 23 November 2022).
  432. Oyama, R.; Kanayama, M.; Mori, M.; Matsumiya, H.; Taira, A.; Shinohara, S.; Takenaka, M.; Yoneda, K.; Kuroda, K.; Tanaka, F. CD155 Expression and Its Clinical Significance in Non-Small Cell Lung Cancer. Oncol. Lett. 2022, 23, 166. [Google Scholar] [CrossRef]
  433. TIGIT-T-Cell Immunoreceptor with Ig and ITIM Domains-Homo Sapiens (Human) | UniProtKB | UniProt. Available online: https://www.uniprot.org/uniprotkb/Q495A1/entry (accessed on 23 November 2022).
  434. Thao, K.; Huynh, P. Anti-TIGIT and ICI Combinations Supported by Early-Phase Research. Target. Ther. Oncol. 2021, 10. [Google Scholar]
Figure 1. Risk factors for lung cancer (based on [4,5,6,7,8,9,10,11]).
Figure 1. Risk factors for lung cancer (based on [4,5,6,7,8,9,10,11]).
Ijms 24 01506 g001
Figure 2. Classification of different subtypes of lung cancer (based on [30,31,32]).
Figure 2. Classification of different subtypes of lung cancer (based on [30,31,32]).
Ijms 24 01506 g002
Figure 3. Stages of cancerogenesis development and progression (based on [300]).
Figure 3. Stages of cancerogenesis development and progression (based on [300]).
Ijms 24 01506 g003
Figure 4. Biomarker molecules for use in the diagnostic process of lung cancer (based on [313,314,315,316,317,318,319,320,321,322,323,324,325,326,327,328,329,330,331,332,333,334,335,336]). Abbreviations: p53—tumor protein P53; CAGE—cancer-associated gene protein; NY-ESO-1—New York esophageal squamous cell carcinoma-1; SOX2—SRY-box transcription factor 2; MAGEA4—melanoma-associated antigen 4; HuD—ELAV-like protein 4; PGP 9.5—protein gene product 9.5; GAGE7—G antigen 7; MAGEA1—melanoma-associated antigen 1; IMPDH1—inosine monophosphate dehydrogenase 1; PGAM—phosphoglycerate mutase; HSP-9B—heat shock protein family A member 9B; SEC15L2—EXOC1 exocyst complex component 1; XRCC5—X-ray repair cross complementing 5; MALAT1—metastasis-related lung adenocarcinoma transcript 1; HCC1—nuclear protein Hcc-1; BARD1—BRCA1-associated RING domain protein 1; ECH1—Enoyl-CoA hydratase 1; HNRNPA2B1—heterogeneous nuclear ribonucleoprotein A2/B1; ANXA1—annexin A1; FOXP3—forkhead box protein P3; c-MYC—MYC proto-oncogene; MDM2—mouse double minute 2 homolog; NPM1—nucleophosmin 1; p16—cyclin-dependent kinase inhibitor 2A; TTC14—tetratricopeptide repeat domain 14; BRAF—serine/threonine-protein kinase B-raf; CK8—cytokeratin 8; CK20—cytokeratin 20; CDK2—cyclin-dependent kinase 2; CEA—carcinoembryonic antigen; CYFRA 21-1—cytokeratin 19 fragments; CA 125—cancer antigen 125; CRP—C-reactive protein; HGF—hepatocyte growth factor; ENO1—alpha-enolase; ProGRP—progastrin-releasing peptide; MCP1—monocyte chemoattractant protein 1; IL-6—interleukin-6; IL-10—interleukin-10; NSE—neuron-specific enolase; SAA—serum amyloid A; RBP—retinol-binding proteins; A1AT—alpha-1 antitrypsin; CART—cocaine and amphetamine-regulated transcript; MIP-1α—macrophage inflammatory proteins alpha 1; SCF—Skp, Cullin, F-box-containing complex; TNF RI—tumor necrosis factor receptor I; IFN-γ—interferon gamma; TNF-α—tumor necrosis factor alpha; sIL-6R—soluble interleukin-6 receptor; TTR—transthyretin; THBS1—thrombospondin-1; CCL5—CC motif chemokine ligand 5; MIF—macrophage migration inhibitory factor; PAI-1—plasminogen activator inhibitor 1; ERBB2—erb-b2 receptor tyrosine kinase 2; IGF1—insulin-like growth factor 1; RANTES—regulated upon activation, normal T-cell expressed and secreted; AFP—alpha fetoprotein; MMP-1—matrix metallopeptidase 1; MMP-9—matrix metallopeptidase 9; YKL-40—chitinase-3-like protein; IL-1Ra—interleukin 1 receptor antagonist; sIL-2Ra—human soluble interleukin 2 receptor alpha; SDF-1α+β—stromal cell-derived factor 1 alfa + beta.
Figure 4. Biomarker molecules for use in the diagnostic process of lung cancer (based on [313,314,315,316,317,318,319,320,321,322,323,324,325,326,327,328,329,330,331,332,333,334,335,336]). Abbreviations: p53—tumor protein P53; CAGE—cancer-associated gene protein; NY-ESO-1—New York esophageal squamous cell carcinoma-1; SOX2—SRY-box transcription factor 2; MAGEA4—melanoma-associated antigen 4; HuD—ELAV-like protein 4; PGP 9.5—protein gene product 9.5; GAGE7—G antigen 7; MAGEA1—melanoma-associated antigen 1; IMPDH1—inosine monophosphate dehydrogenase 1; PGAM—phosphoglycerate mutase; HSP-9B—heat shock protein family A member 9B; SEC15L2—EXOC1 exocyst complex component 1; XRCC5—X-ray repair cross complementing 5; MALAT1—metastasis-related lung adenocarcinoma transcript 1; HCC1—nuclear protein Hcc-1; BARD1—BRCA1-associated RING domain protein 1; ECH1—Enoyl-CoA hydratase 1; HNRNPA2B1—heterogeneous nuclear ribonucleoprotein A2/B1; ANXA1—annexin A1; FOXP3—forkhead box protein P3; c-MYC—MYC proto-oncogene; MDM2—mouse double minute 2 homolog; NPM1—nucleophosmin 1; p16—cyclin-dependent kinase inhibitor 2A; TTC14—tetratricopeptide repeat domain 14; BRAF—serine/threonine-protein kinase B-raf; CK8—cytokeratin 8; CK20—cytokeratin 20; CDK2—cyclin-dependent kinase 2; CEA—carcinoembryonic antigen; CYFRA 21-1—cytokeratin 19 fragments; CA 125—cancer antigen 125; CRP—C-reactive protein; HGF—hepatocyte growth factor; ENO1—alpha-enolase; ProGRP—progastrin-releasing peptide; MCP1—monocyte chemoattractant protein 1; IL-6—interleukin-6; IL-10—interleukin-10; NSE—neuron-specific enolase; SAA—serum amyloid A; RBP—retinol-binding proteins; A1AT—alpha-1 antitrypsin; CART—cocaine and amphetamine-regulated transcript; MIP-1α—macrophage inflammatory proteins alpha 1; SCF—Skp, Cullin, F-box-containing complex; TNF RI—tumor necrosis factor receptor I; IFN-γ—interferon gamma; TNF-α—tumor necrosis factor alpha; sIL-6R—soluble interleukin-6 receptor; TTR—transthyretin; THBS1—thrombospondin-1; CCL5—CC motif chemokine ligand 5; MIF—macrophage migration inhibitory factor; PAI-1—plasminogen activator inhibitor 1; ERBB2—erb-b2 receptor tyrosine kinase 2; IGF1—insulin-like growth factor 1; RANTES—regulated upon activation, normal T-cell expressed and secreted; AFP—alpha fetoprotein; MMP-1—matrix metallopeptidase 1; MMP-9—matrix metallopeptidase 9; YKL-40—chitinase-3-like protein; IL-1Ra—interleukin 1 receptor antagonist; sIL-2Ra—human soluble interleukin 2 receptor alpha; SDF-1α+β—stromal cell-derived factor 1 alfa + beta.
Ijms 24 01506 g004
Figure 5. Exemplary interactions between a T lymphocyte and a tumor cell and dendritic cell, including immunological checkpoints (based on [350,351,352,353,354,355]).
Figure 5. Exemplary interactions between a T lymphocyte and a tumor cell and dendritic cell, including immunological checkpoints (based on [350,351,352,353,354,355]).
Ijms 24 01506 g005
Figure 6. Strategy for searching articles for literature review based on PRISMA flow diagram (based on https://www.prisma-statement.org/PRISMAStatement, accessed on 22 November 2022).
Figure 6. Strategy for searching articles for literature review based on PRISMA flow diagram (based on https://www.prisma-statement.org/PRISMAStatement, accessed on 22 November 2022).
Ijms 24 01506 g006
Table 1. List of the 20 most mutated genes by lung cancer subtype (adapted from [37]).
Table 1. List of the 20 most mutated genes by lung cancer subtype (adapted from [37]).
ACLCCNCSLCSCLCADC
EGFR (31%)TP53 (68%)EGFR (25%)TP53 (63%)TP53 (57%)
KRAS (17%)KRAS (22%)KRAS (16%)RB1 (46%)EGFR (7%)
TP53 (42%)EGFR (5%)TP53 (31%)LRP1B (25%)LRP1B (37%)
STK11 (11%)STK11 (8%)ARID1A (25%)KMT2D (16%)PIK3CA (8%)
BRAF (3%)PIK3CA (4%)PIK3CA (4%)EGFR (6%)NFE2L2 (12%)
KEAP1 (15%)NRAS (3%)ALK (17%)NOTCH1 (9%)KRAS (4%)
MET (3%)MET (3%)BRAF (2%)PIK3CA (7%)KMT2D (17%)
LRP1B (28%)KEAP1 (21%)MET (2%)PTEN (6%)CDKN2A (11%)
ERBB2 (2%)AR (32%)STK11 (7%)EP300 (8%)FAT4 (16%)
PIK3CA (4%)BRAF (2%)KEAP1 (16%)FAT1 (9%)KMT2C (15%)
SMARCA4 (9%)PTEN (4%)CDKN2A (4%)ATRX (8%)KEAP1 (9%)
NF1 (8%)CDKN2A (5%)ATM (13%)KMT2C (8%)NF1 (11%)
ATM (7%)LRP1B (28%)KMT2D (24%)CREBBP (8%)PTEN (7%)
FAT1 (8%)POLQ (28%)SMARCA4 (10%)GRIN2A (8%)ERBB4 (8%)
PTPRT (9%)HRAS (4%)NF1 (9%)FAT4 (10%)FAT1 (12%)
KMT2C (9%)GNAS (4%)SETBP1 (10%)ROS1 (8%)ROS1 (10%)
RBM10 (8%)ERBB4 (5%)FAT1 (15%)ERBB4 (6%)PREX2 (13%)
KMT2D (7%)ATM (6%)ERBB2 (2%)ARID1A (6%)PDE4DIP (11%)
FAT4 (16%)FLT3 (7%)TET2 (8%)ZNF521 (7%)ZFHX3 (9%)
ERBB4 (5%)ZFHX3 (21%)NOTCH1 (3%)MTOR (5%)GRIN2A (10%)
Abbreviations: EGF—epidermal growth factor receptor; KRAS—Kirsten rat sarcoma virus; TP53—tumor protein p53; STK11—serine/threonine kinase 11; BRAF—B-Raf proto-oncogene, serine/threonine kinase; Keap1-Kelch-like ECH-associated protein 1; MET—MET proto-oncogene, receptor tyrosine kinase; LRP1B—LDL receptor-related protein 1B; ERBB2-Erb-B2 receptor tyrosine kinase 2; PIK3CA—phosphatidylinositol-4,5-bisphosphate 3-kinase catalytic subunit alpha; SMARCA4—SWI/SNF-related, matrix-associated, actin-dependent regulator of chromatin, subfamily A, member 4; NF1—neurofibromin 1; ATM-ATM serine/threonine kinase; FAT1—FAT atypical cadherin 1; PTPRT-protein tyrosine phosphatase receptor type T; KMT2C-lysine methyltransferase 2C; RBM10 (RNA binding motif protein 10; KMT2D—lysine methyltransferase 2D; FAT4—FAT atypical cadherin 4; ERBB4—Erb-B2 receptor tyrosine kinase 4; NRAS—NRAS proto-oncogene, GTPase; AR—androgen receptor; PTEN—phosphatase and tensin homolog; CDKN2A—cyclin-dependent kinase inhibitor 2A; POLQ—DNA polymerase theta; HRAS—HRas proto-oncogene, GTPase; GNAS—GNAS complex locus; FLT3—Fms-related receptor tyrosine kinase 3; ZFHX3—zinc finger homeobox 3; ARID1A—AT-rich interaction domain 1A; ALK—ALK receptor tyrosine kinase; SETBP1—SET binding protein 1; TET2—tet methylcytosine dioxygenase 2; Notch 1—neurogenic locus notch homolog protein 1; RB1—RB transcriptional corepressor 1; EP300 Gene—E1A binding protein P300; ATRX—ATRX chromatin remodeler; CREBBP—CREB binding protein; GRIN2A—glutamate ionotropic receptor NMDA type subunit 2A; ROS1—ROS proto-oncogene 1, receptor tyrosine kinase; ZNF521—zinc finger protein 521; MTOR—mechanistic target of rapamycin kinase; NFE2L2—NFE2-like BZIP transcription factor 2; REX2—phosphatidylinositol-3,4,5-trisphosphate-dependent Rac exchange factor 2; PDE4DIP—phosphodiesterase 4D interacting protein.
Table 2. Disturbances in cell signaling pathways involved in the development and progression of lung cancer.
Table 2. Disturbances in cell signaling pathways involved in the development and progression of lung cancer.
TypeGene and LocationProtein NameAmino Acid LengthMolar Weight [kDa]FunctionFunction in Lung CancerReferences
Cell signaling pathway stimulating cell growthEGFR,
7p11.2
EGFR1210134.277
  • The main regulator of the cell proliferation process;
  • Responsible for endocrine and paracrine regulation of cell growth and proliferation.
  • Acts as a strong oncogene in the case of deregulatory changes;
  • Its dysregulation has been observed in 40% of NSCLC and ADC cases;
  • Its increased expression is associated with a lower prognosis of patients.
[55,56,57,58,59]
BRAF, 7q34BRAF76684.437
  • A serine/threonine kinase involved in the transduction of cellular signals related to growth;
  • Plays a key role in the regulation of MAPK/ERK signaling.
  • Mutations are detected in about 3% of lung cancer cases;
  • It is a proto-oncogene;
  • The V600 mutation in loop A is the most common type of mutation;
  • A mutation is incompatible with RET, Ras mutations in lung cancer.
[60,61,62]
ERBB2,
17q12
HER21255137.910
  • It is a membrane-associated tyrosine kinase and is responsible for regulating cell proliferation.
  • Its mutation occurs in non-smokers;
  • The mutation affects about 2% of lung cancer cases;
  • It is an oncogene.
[63,64,65]
KRAS,
12p12.1
KRAS18921.656
  • Part of the signaling of the RAS/MAPK pathway;
  • Responsible for cell proliferation and differentiation.
  • It is a proto-oncogene;
  • Mutation involved in the pathogenesis of lung adenoma;
  • Mutations are more common in smokers and men.
[66,67,68,69]
PIK3CA, 3q25PIK3CA1068124.284
  • It is a kinase involved in a number of cellular functions such as growth, proliferation, differentiation, motility and secretion.
  • One of the most frequently mutated onco-genes along with KRAS;
  • Relatively little information about its mutation in NSCLC;
  • Lack of complete studies on the relationship of its amplification in lung cancer (SCLC).
[70,71,72]
ALK,
2p32.2
ALK1620176.442
  • Plays a role in RAS activation;
  • Participates in cellular communication and the proper development and functioning of the nervous system.
  • Occurs in 7% of NSCLC cases;
  • Negative for KRAS or EGFR mutations;
  • Mutations favor the development of ADCs;
  • It is associated with a tendency to metastasize to the pleura and pericardium;
  • Mutations are seen more often in women.
[73,74]
TITF1,
14q13.3
TITF137138.596
  • The main transcription factor necessary for the development of peripheral airways
  • It is a specific marker of ADC development;
  • Can also be expressed in SCLC.
[75,76,77]
c-MYC, 8q24.21MYC43948.804
  • It is a transcription factor;
  • Participates in cell proliferation and differentiation.
  • It is a proto-oncogene;
  • l- and n-MYC amplification is specific to SCLC and LCNEC development.
[78,79,80]
l-MYC,
1p34.2
MYC36440.327[80,81]
n-MYC,
2p24.3
MYC46449.561[79,80,82]
Szlak genów supresorowych guzaTP53, 17p13P5339343.653
  • Cell-cycle transcription factor;
  • It is a kind of cellular guardian, protecting against instability and genetic abnormalities;
  • It is also a sensor of stress signals, e.g., DNA damage, the activation of oncogenes;
  • It is a tumor suppressor;
  • Regulated by Mdm2 and p14ARF.
  • It is the most frequently mutated gene in lung cancer: 90% of SCLC and LCNEC and 50% of NSCLC;
  • The mutation is closely related to exposure to carcinogens, especially smoking and benzopyrene;
  • Mutations in regulatory genes: 65 cases of NSCLC were related to Mad2; the mutation in p14ARF affects SCLC, LCNEC and ADC.
[83,84,85]
RB1, 13q14.2Rb1928106.159
  • Tumor suppressor gene;
  • It is an effector of p53-mediated G1/s cycle arrest via activation of p21 kinase inhibitor.
  • The lack of Rb protein is one of the most common escape mechanisms from the G1 checkpoint seen in SCLC; in NSCLC, hyperphosphorylation of Rb occurs;
  • Loss of Rb occurs in 70% of LCNEC cases, 90% of SCLC cases and 15% of NSCLC cases.
[86,87,88]
STK11, 19p13.3STK1143348.636
  • It is a serine/threonine kinase;
  • Acts as a tumor suppressor;
  • Is the precursor kinase to AMPK, an essential component of cellular metabolism and the maintenance of energy homeostasis.
  • A mutation of this gene has been observed in ADC;
  • It is a critical barrier to new lung formation, controls initiation, differentiation and metastasis;
  • Typically, mutations are in smokers and are associated with a mutation in KRAS.
[89,90,91]
Szlak związany z unikaniem apoptozyBax, 19q13.33Bax19221.184
  • Bcl-2 and Bax are key factors of mitochondrial apoptosis involved in the control of outer membrane permeabilization, resulting in the release of cytochrome C.
  • Bax heterodimerizes with Bcl-2 to control the level of susceptibility to apoptosis;
  • Bcl-2 is overexpressed in SCLC and LCNEC and downregulated in NSCLC;
  • Bax is downregulated in SCLC and upregulated in NSCLC;
  • Abnormalities in the Bcl-2:Bax ratio are observed in 95% of SCLC cases and 25% of NSCLC cases.
[92,93,94,95,96]
Bcl-2, 18q21.33Bcl-223926.266[93,94,96,97,98]
FASL, 1q24.3TNFL628131.485
  • Its interaction with the Fas transmembrane receptor (FasR) on target cells induces the course of apoptosis;
  • Has immunoregulatory importance and is involved in the process of carcinogenesis.
  • Like the FasR receptor, FasL has been shown to be downregulated in 70% of NSCLC cases;
  • In SCLC, there is no expression of FasR but high expression of FasL (50% of cases).
[99,100,101]
E2F1, 20q11.22E2F143746.920
  • It is a transcription factor involved in the control of the G1/S cell cycle;
  • Can mediate both p53-dependent and non-p53-dependent proliferation and apoptosis.
  • Low levels of expression are detected in NSCLC and have been suggested to increase cancer cell survival;
  • High levels of expression are seen in SCLC and increase tumor cell proliferation.
[102]
TERT, 5p15.33TERT1132126.997
  • Responsible for maintaining telomere ends;
  • It is involved in the aging process of cells.
  • High expression of telomerase contributes to the early stages of immortalization of cancer cells;
  • Elevated telomerase levels are observed in 80% of NSCLC cases and nearly 100% of SCLC cases;
  • Telomerase is expressed in checkpoint-deficient cancer cells via p53 or Rb.
[103,104,105]
Abbreviations: EGFR—epidermal growth factor receptor; BRAF—serine/threonine-protein kinase B-raf; MAP/ERK—mitogen-activated protein kinase/extracellular signal-regulated kinase ½; ERBB2—receptor tyrosine-protein kinase erbB-2; HER2—human epidermal growth factor receptor 2; RET—ret proto-oncogene; Ras—rat sarcoma virus; KRAS—Kirsten rat sarcoma virus; PIK3CA—phosphatidylinositol-4,5-bisphosphate 3-kinase catalytic subunit alpha; ALK—tyrosine kinase receptor; TITF1—thyroid transcription factor 1; MYC—Myc proto-oncogene protein; TP53—cellular tumor antigen p53; Mdm2—mouse double minute 2 homolog MDM2; p14ARF—ARF tumor suppressor; RB1—retinoblastoma-associated protein; STK11—serine/threonine-protein kinase STK11; AMPK—5’AMP-activated protein kinase; BAX—apoptosis regulator BAX; BCL2—apoptosis regulator Bcl-2; TNFL6—tumor necrosis factor ligand superfamily member 6; E2F1—E2F transcription Factor 1; E2F1—transcription factor E2F1; TERT—telomerase reverse transcriptase.
Table 3. miRNAs involved in the pathogenesis of lung cancer.
Table 3. miRNAs involved in the pathogenesis of lung cancer.
miRNAGenetic LocationTargetMechasimsReferences
miR-7miR-7 is derived from three precursors: pri-miR-7-1 (9q21), pri-miR-7-2 (15q26) and andpri-miR-7-3 (19q13)EGFRSustaining proliferative signaling[139,140]
miR-2719p13.12EGFRSustaining proliferative signaling[141,142,143,144]
miR-306q13EGFRSustaining proliferative signaling[145,146,147]
miR-341p36.22EGFRSustaining proliferative signaling[148,149,150]
PD-L1Avoiding immune destruction and tumors promoting inflammation
miR-1282q21.3EGFRSustaining proliferative signaling[151,152,153,154]
VEGFInducing angiogenesis
miR-13414q32.31EGFRSustaining proliferative signaling[155,156,157]
miR-542Xq26.3EGFRSustaining proliferative signaling[158,159,160]
miR-12582q31.3Grb2Sustaining proliferative signaling[161,162,163]
miR-148a7p15.2SOSSustaining proliferative signaling[164,165,166]
miR-181achromosome 1 (37.p5)RASSustaining proliferative signaling[167,168]
miR-193a17q11.2RASSustaining proliferative signaling[169,170,171]
miR-7601p22.1ROS1Sustaining proliferative signaling[172,173,174]
miR-967q32.2EML4-ALKSustaining proliferative signaling[175,176,177]
miR-200c/12p13.31PI3KSustaining proliferative signaling[178,179,180,181,182]
chromosome 1ZEB1/2Activating invasion and metastasis
miR-200VEGFInducing angiogenesis
PD-L1Avoiding immune destruction and tumors promoting inflammation
miR-520a19q13.42PI3KSustaining proliferative signaling[183,184,185]
miR-64119q13.2MDM2Evading growth suppressors[186,187,188]
miR-660Xp11.23MDM2Evading growth suppressors[189,190]
miR-15a13q14.2CDK/CyclinEvading growth suppressors[191,192,193]
miR-1613q14CDK/CyclinEvading growth suppressors[194,195,196,197]
BclResisting cell death
miR-449a5q11.2E2FEvading growth suppressors[198,199,200,201]
miR-29914q32.31hTERTEnabling replicative immortality[202,203,204]
miR-4919p21.3hTERTEnabling replicative immortality[205,206,207]
miR-51219q13.42hTERTEnabling replicative immortality[208,209,210]
miR-297q32.3DNMTEnabling replicative immortality[211,212,213]
miR-146A5q33.3SmadActivating invasion and metastasis[214,215,216]
miR-2117q23.1TGF-βActivating invasion and metastasis[217,218,219,220,221]
PTENAvoiding immune destruction and Tumors promoting inflammation
miR-23no dataTGF-βActivating invasion and metastasis[222,223,224]
miR-99A21q21.1TGF-βActivating invasion and metastasis[225,226,227]
miR-15521q21.3TGF-βActivating invasion and metastasis[228,229,230,231]
miR-19211q13.1TGF-βActivating invasion and metastasis[232,233,234,235]
miR-115no dataTGF-βActivating invasion and metastasis[236,237]
miR-52019q13.42TGF-βActivating invasion and metastasis[238,239,240]
miR-49717p13.1HDGFInducing angiogenesis[241,242,243,244]
miR-49414q32.31PTENInducing angiogenesis[245,246,247,248]
miR-1269q34.3VEGFInducing angiogenesis[249,250,251,252]
miR-33b17p11.2LDHADeregulating cellular energetics[253,254,255]
miR-14417q11.2GLUTDeregulating cellular energetics[256,257,258]
miR-1248p23.1AktDeregulating cellular energetics[259,260,261]
miR-199a19p13.2HIF-1Deregulating cellular energetics[262,263,264]
miR-31-5p9p21.3FIHDeregulating cellular energetics[265,266,267]
miR-283q28PD-1PTENAvoiding immune destruction and tumors promoting inflammation[268,269,270]
miR-13816q13PD-1Avoiding immune destruction and tumors promoting inflammation[271,272]
PD-L1
miR-1971p13.3CKS1BAvoiding immune destruction and tumors promoting inflammation[273,274,275]
miR-513Xq27.3PD-L1Avoiding immune destruction and tumors promoting inflammation[276,277,278,279]
miR-2013q31.3PTENAvoiding immune destruction and tumors promoting inflammation[280,281,282]
miR-13011q12.1PTENAvoiding immune destruction and tumors promoting inflammation[283,284,285]
miR-424Xq26.3CD80Avoiding immune destruction and tumors promoting inflammation[286,287,288,289]
miR-13614q32.2CTLA-4Avoiding immune destruction and tumors promoting inflammation[290,291,292,293]
miR-301b22q11.21BH3Resisting cell death[294,295,296,297,298]
Abbreviations: EGFR—epidermal growth factor receptor; PD-L1—programmed death-ligand 1; VEGFA—vascular endothelial growth factor A; Grb2—growth factor receptor bound protein 2; SOS—Ras/Rac guanine nucleotide exchange factor; Ras—rat sarcoma virus; ROS1—ROS proto-oncogene 1, receptor tyrosine kinase; EML4—echinoderm microtubule-associated protein-like 4; ALK—anaplastic lymphoma kinase; PI3K—phosphoinositide 3-kinase; ZEB1 zinc finger E-box binding homeobox 1; MDM2—mouse double minute 2 homolog; CDK—cyclin-dependent kinase; Bcl—B-cell lymphoma; hTERT—telomerase reverse transcriptase; DNMT—DNA methyltransferase; SMAD—suppressor of mothers against decapentaplegic; TGF-β—transforming growth factor beta; PTEN—phosphatase and tensin homolog; HDGF—heparin binding growth factor; LDHA—lactate dehydrogenase A; GLUT—glucose transporter; HIF-1—hypoxia inducible factor 1 subunit alpha; FIH—hypoxia-inducible factor inhibitor; CKS1B—CDC28 protein kinase regulatory subunit 1B; CD80—cluster of differentiation 80; CTLA-4—cytotoxic T cell antigen 4; BH3—interacting domain death agonist.
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

Smok-Kalwat, J.; Mertowska, P.; Mertowski, S.; Smolak, K.; Kozińska, A.; Koszałka, F.; Kwaśniewski, W.; Grywalska, E.; Góźdź, S. The Importance of the Immune System and Molecular Cell Signaling Pathways in the Pathogenesis and Progression of Lung Cancer. Int. J. Mol. Sci. 2023, 24, 1506. https://doi.org/10.3390/ijms24021506

AMA Style

Smok-Kalwat J, Mertowska P, Mertowski S, Smolak K, Kozińska A, Koszałka F, Kwaśniewski W, Grywalska E, Góźdź S. The Importance of the Immune System and Molecular Cell Signaling Pathways in the Pathogenesis and Progression of Lung Cancer. International Journal of Molecular Sciences. 2023; 24(2):1506. https://doi.org/10.3390/ijms24021506

Chicago/Turabian Style

Smok-Kalwat, Jolanta, Paulina Mertowska, Sebastian Mertowski, Konrad Smolak, Aleksandra Kozińska, Filip Koszałka, Wojciech Kwaśniewski, Ewelina Grywalska, and Stanisław Góźdź. 2023. "The Importance of the Immune System and Molecular Cell Signaling Pathways in the Pathogenesis and Progression of Lung Cancer" International Journal of Molecular Sciences 24, no. 2: 1506. https://doi.org/10.3390/ijms24021506

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