Prognosis of Different Types of Non-Small Cell Lung Cancer Progression: Current State and Perspectives

 

Anastasia A. Schegolevaa    Anna A. Khozyainovaa    Anton A. Fedorova

Tatiana S. Gerashchenkoa    Evgeny O. Rodionovb    Evgeny B. Topolnitskyc

Nikolay A. Sheferd    Olga V. Pankovae    Anastasia A. Durovaf

Marina V. Zavyalovae,f    Vladimir M. Perelmutere    Evgeny V. Denisova

 

aLaboratory of Cancer Progression Biology, Cancer Research Institute, Tomsk National Research Medical Center, Russian Academy of Sciences, Tomsk, Russia, bDepartment of Thoracic Oncology, Cancer Research Institute, Tomsk National Research Medical Center, Russian Academy of Sciences, Tomsk, Russia, cDepartment of Surgery with a Course of Mobilization Training and Medicine of Disasters, Siberian State Medical University, Tomsk, Russia, dSurgery Department, Tomsk Regional Oncology Center, Tomsk, Russia, eDepartment of General and Molecular Pathology, Cancer Research Institute, Tomsk National Research Medical Center, Russian Academy of Sciences, Tomsk, Russia, fDepartment of Pathological Anatomy, Siberian State Medical University, Tomsk, Russia

 

 

 

 

Key Words

Non-small cell lung cancer • Progression • Recurrence • Metastasis • Marker

 

Abstract

Despite advances in diagnostics and therapy of non-small cell lung cancer (NSCLC), the problem of prognosis and prevention of tumor progression is still highly important. Even if NSCLC is diagnosed in the early stages, almost a quarter of patients develop relapse; most of them die from recurrent disease. A large number of different markers have been proposed to predict the risk of NSCLC progression; however, none of them are used in clinical practice. It is obvious that this situation is related to the economic and methodological complexity of the proposed markers and/or their insufficient efficiency due to a lack of effective study models and tumor heterogeneity. Another reason may be that potential markers are developed for NSCLC progression in general, which is represented by at least four pathogenetically-distinct processes: synchronous lymph node metastasis, local, regional, and distant recurrence. In this review, we summarize data from published literature on clinicopathological, genetic, and molecular factors associated with different types of NSCLC progression and emphasize challenges and approaches to developing prognostic factors. In conclusion, we highlight the importance of further studies to reveal molecular mechanisms of NSCLC progression and the need for differential analysis of markers of local, regional, and distant recurrences for disease prognosis.

 

 

Introduction

 

Despite significant advances in diagnostics and therapy of non-small cell lung cancer (NSCLC), there is still no effective treatment approaches to prevent tumor progression. Even if NSCLC is diagnosed at stages I and II, almost a quarter of patients develop postoperative relapse; most of them die from recurrent disease [1, 2]. This issue is complicated by the fact that in the vast majority of patients relapses are inoperable [3]. As a result, the 5-year relative survival rate of patients with advanced NSCLC is only 6.1%. However, in patients with regional involvement and localized NSCLC, the 5-year relative survival rate is 34.5% and 61.4%, respectively, which indicate the importance of early diagnosis, relapse prognosis, and the development of approaches to prevent the recurrent disease [4].

Many studies described potential markers of NSCLC recurrence including the expression of genes, proteins, and non-coding RNAs, as well as genetic and epigenetic alterations, and circulating molecules. For example, a signature of miR-210, miR-214, and miR-15a has been proposed to predict the probability of lung adenocarcinoma (LUAD) metastasis into the brain (90.4% accuracy) [5]. In another study, a panel of CSF1, EGFR, and CA9 (CA-IX) genes has been developed to predict the risk of distant recurrence in patients with lung squamous cell carcinoma (LUSC) (85% sensitivity and 60% specificity) [6]. Some researches have shown an association of somatic mutations in the ALK, EGFR, and KRAS genes with the risk of NSCLC recurrence [7-13].

Despite a large number of studies, the proposed markers and prognostic signatures are mainly applicable to LUAD and significantly less often to LUSC. Moreover, they have a number of significant limitations, such as insufficient predictive efficiency, economic and methodological complexity of analysis. Because of this, none of the markers are approved to be used in clinical practice. Among other reasons, this may be due to the heterogeneous nature of NSCLC progression that is represented by lymph node metastasis synchronous with the primary tumor, as well as local, regional, and distant recurrence. In present, there is some confusion in the literature regarding definitions of local, regional, and distant recurrence of NSCLC. In this review, we settled on the following definitions. Local recurrence means that cancer reappears in the same lung or at the bronchial stump [14-18]. Regional recurrence is identified as a recurring disease in the ipsilateral hilar, mediastinal, and supraclavicular lymph nodes detected during follow-up after treatment [9, 14, 19]. All other sites of recurrence are referred to as distant recurrence or metastasis [14]. It is interesting to note that metastasis to the pleura is referred to as a form of distant recurrence [9, 10]. However, there is an opinion that pleural metastasis can be an independent type of cancer progression [19]. The probability of distinct types of NSCLC progression and their contribution to the disease outcome are different. It was found that in surgically resected NSCLC patients are more likely to develop distant relapses than local (67.8-79.5% vs. 20.5%-28.8%). The simultaneous development of local and distant relapses is quite rare (3.4-6.4%) [3]. The most favorable outcome is observed in patients with locoregional relapses [20].

Previously, we showed that the probability of locoregional and distant recurrence of NSCLC is associated with the presence of premalignant lesions in the small bronchi distant from the primary tumor. The isolated basal cell hyperplasia (BCH) was related to increased distant metastasis and a decreased locoregional recurrence [21]. On the contrary, the co-presence of BCH and squamous metaplasia (SM) was associated with a high probability of locoregional recurrence and a low risk of distant metastasis [22, 23]. Probably, epithelial-stromal interactions in the small bronchi may reflect constitutive features of the inflammation and can be indirectly related to tumor-stromal interactions and cancer progression. In other words, some individuals are predisposed to locoregional recurrence, while others are predisposed to distant metastasis.

Thus, the biology and clinical significance of NSCLC progression significantly vary, and it is necessary to develop prognostic markers specific to synchronous lymph node metastasis, local, regional, and distant recurrence. Unfortunately, most studies reported the markers that are associated with the NSCLC recurrence in general. The aim of the current review is to systematize the available literature data on clinicopathological parameters, germline and somatic genetic variations, gene and protein expression, non-coding RNAs, and circulating molecules that can be potential markers of synchronous lymph node metastasis, as well as local, regional, and distant recurrence of NSCLC. This review also emphasizes the main challenges and approaches to developing prognostic factors of NSCLC and the importance of further studies of molecular mechanisms of different types of NSCLC progression.

 

 

Synchronous lymph node metastasis

 

Clinicopathological markers

Surgical pathology has an important role in the diagnosis and treatment of lung cancer [24]. Despite the remarkable progress in molecular pathology related mainly to the development of targeted therapies specific for histological types of NSCLC, routine clinical and morphological parameters still remain of significant prognostic value. In particular, it is known that non-upper lobe located NSCLC and micropapillary-predominant LUAD have an increased lymph node metastasis [25]. In addition, tumor size correlates with lymph node involvement [26-29]. For example, tumor size more than 3 cm and central localization of the tumor are associated with lymph node metastasis [30]. Lymph node lesions are also related to tumor grade and pleural involvement [31].

 

Genetic markers

Germline variations. Single nucleotide polymorphisms (SNPs) and copy number variations (CNVs) form the genetic landscape of the individual that controls cellular processes and determines predisposition to different diseases [32]. Many studies demonstrated the contribution of certain SNPs and CNVs to an increased risk of NSCLC [33]. However, only a few studies showed that the probability of lymph node metastasis of NSCLC can be determined by germline variations. For example, a high risk of lymph node involvement is associated with the rs9876 AA genotype of the TNS3 gene [34]. In smokers, lymph node metastasis is related to the MMP1-MMP3 1G/5A haplotype [35].

Somatic alterations. Transformation of normal cells to tumor cells occurs due to acquiring a series of mutations over time. Tumors can harbor tens to hundreds and thousands of different mutations some of whom are critically important for carcinogenesis and called “drivers” whereas other alterations, “passengers”, are considered to be neutral [36]. The mutational landscape of NSCLC is well studied and driver genes have been identified to contribute to carcinogenesis in the respiratory epithelium. LUAD is believed to be associated mainly with alterations in the KRAS, EGFR, BRAF, and MET genes, [37] whereas the development of lung LUSC is predominantly related to changes in the TP53, CDKN2A, PTEN, PIK3CA, KEAP1, MLL2, HLA-A, and NFE2L2 genes [38]. Single studies showed that driver and some other genes can be involved in lymph node metastasis of NSCLC. LUAD patients harboring both a known driver (KRAS, EGFR, NRAS, etc.) or candidate mutation, and TP53 mutations have high rates of lymph node metastasis [39]. ALK-rearranged NSCLC tended to show more frequent lymph node involvement, even in patients with low T stage [11].

 

Molecular markers

Gene and protein expression. Genes, particularly encoded proteins, are the main players in cellular processes in normal physiology and cancer. There is a lot of data demonstrating the association between the expression of genes and proteins and synchronous lymph node metastasis of NSCLC. In general, lymph node involvement of NSCLC is associated with overexpression of hMTH1, SPD, ITGA11, COL11A1, MACC1, S100A4, MTA2, CXCR4 and CPSF3 genes, as well as a high level of PRMT5, Hsp90-beta, annexin A1 (ANXA1), MACC1, integrin αvβ5 and TRIM44 proteins [40-49](Table 1). In LUAD, a high incidence of lymph node metastases is related to overexpression of RACK1, CD147 and underexpression of RASAL2 proteins [50, 51].

Non-coding RNAs. Non-coding RNAs are crucial regulators of cellular processes and represented by three main types: translation-related RNAs (rRNAs and tRNAs), small non-coding RNAs (siRNA, miRNAs, and piRNAs), and long non-coding RNAs [52]. Many studies showed the association of miRNAs and long non-coding RNAs with synchronous lymph node metastasis of NSCLC. In general, lymph node involvement is associated with underexpression of miR-451, miR-449a, miR-486-5p, miR-206, miR-139, miR-187-5p, miR-212, miR-138, and miR-101 and overexpression of miR-9, miR-26a, miR-422a, miR-944 and miR-1290 (Table 1) [53-66]. miR-449, miR-422a, miR-187-5p, and miR-101 microRNAs are known to promote apoptosis, whereas miR-451, miR-486-5p, miR-187-5p, and miR-101 inhibit proliferation of lung cancer cells [53-55, 58, 61, 67]. Other miRNAs, such as miR-212, miR-486-5p, miR-206, miR-139, miR-187-5p, miR-212, miR-138, miR-1290, and miR-101, suppress epithelial–mesenchymal transition (EMT) and decrease migration, invasion, and metastasis of lung cancer cells [55-60, 68-70]. In contrast, miR-944, miR-9, and miR-26a increase migration and invasion of lung cancer cells and promote metastasis [62, 64, 71]. Thus, miR-138, miR-486-5p, miR-212, miR-449a, miR-451, miR-206, miR-139, miR-101, and miR-187-5p work as tumor suppressors, whereas miR-944, miR-422a, miR-1290, miR-9, and miR-26a possess oncogenic functions.

Underexpression of long non-coding RNAs GAS5-AS1, GAS6-AS1, and BANCR and overexpression of CCAT2, ANRIL, and HOTAIR are associated with lymph node metastasis of NSCLC [56, 68, 72-76]. GAS5-AS1, GAS6-AS1, and BANCR are known to be involved in the suppression of EMT, migration, invasion, and metastasis of lung cancer cells, while HOTAIR, CCAT2, and ANRIL, on the contrary, induce cell proliferation, migration, and invasion [56, 68, 72-75].

Circulating molecules. Blood contains different molecules including circulating DNA, RNA, and proteins. They can be used for the diagnosis of tumors, prediction of treatment response, cancer prognosis, and detection of recurrence [77]. In NSCLC, high level of circulating miR-1290, miR-422a, mRNA of the MACC1 gene, and carcinoembryonic antigen (CEA), and low level of miRNA let-7c and miR-152in blood plasma were found to be associated with lymph node metastasis (Table 1) [25, 26, 65, 66, 78, 79]. miR-1290, miR-422a, let-7c, and miR-152 are known to inhibit migration and invasion of lung cancer cells, while miR-422a induces apoptosis [67, 70, 80, 81].

 

Table 1. Molecular markers associated with synchronous lymph node metastasis of NSCLC. DFS, disease-free survival; LncRNAs, long non-coding RNAs; LUAD, adenocarcinoma; LUSC, lung squamous cell carcinoma; NSCLC, non-small cell lung cancer; PFS, progression-free survival; OS, overall survival; RFS, recurrence/relapse-free survival; "-", not available. The numbers in brackets indicate the size of validation cohorts

 

 

Locoregional recurrence

 

Clinicopathological markers

Visceral pleural invasion (VPI), lymphovascular invasion (LVI), and blood vessel invasion (BVI) were shown to be the main risk factors for locoregional recurrence [82-84]. Besides VPI, in patients with early-stage NSCLC, locoregional recurrence is also related to wedge resection, microscopic positive surgical margin, lymphatic permeation, and large tumor size [18, 85]. Moreover, wedge resection is frequently associated with margins less than 1 cm and, thus, a high risk of locoregional recurrence [86]. Segmentectomy is also an independent risk factor for locoregional recurrence. Superior segmentectomies have significantly lower locoregional recurrence rates and excellent recurrence-free survival (RFS) comparable to lower lobectomies [15].

Locoregional relapses are also more often observed in NSCLC patients with the co-presence of BCH and SM in the small bronchi distant from the primary tumor [22, 23]. Spread through air spaces (STAS) is associated with an increased frequency of locoregional recurrence in patients with LUSC [87]. In LUAD, STAS-positive I stage patients, who underwent sublobar resection or limited resection comparing to lobectomy, have a high risk of locoregional recurrence regardless of margin-to-tumor ratio [16, 17, 88]. Among STAS-negative patients, locoregional recurrence is related to a margin-to-tumor ratio of less than 1 [88]. Interestingly, STAS was more frequently observed in LUAD patients with a cribriform component which is associated with increased risk of locoregional recurrence. Moreover, in patients with the cribriform component, STAS-positive cases had a significantly inferior outcome as compared to STAS-negative cases [89].

 

Genetic markers

The frequency of locoregional recurrence depends on the C609T polymorphism of the NQO1 gene. Relapse is more often observed in patients with the T/T genotype compared to C/C and C/T genotype carriers [90]. Locoregional recurrence is also more frequent in NSCLC patients with missense mutations in exons 18 and 21 and exon 19 deletion of the EGFR gene [8]. Interestingly, ALK rearrangements are associated with greater tendency to develop locoregional recurrence than EGFR and KRAS mutations [9].

Despite the presence of studies focusing on locoregional recurrence of NSCLC in general, we believe that the distinguishing local and regional relapse separately is needed due to their different nature and underlying mechanisms. Below, we provide the literature data regarding potential markers of local and regional recurrence.

 

 

Local recurrence

 

VPI, LVI, and STAS mentioned above in case of locoregional recurrence are also associated with local relapse [83, 91, 92]. Besides, local recurrence is related to other clinical and pathological parameters such as younger age, smoking index more than 20 pack-years, central tumor localization, squamous/large cell histology, stage > IA, sublobar resections, lobectomy with negative surgical margins, segmentectomies in the right upper lobe and of basal segments, tumor margin less than 1.0 cm, lower radiation dose, fewer medical comorbidities, and higher body mass index [15, 83, 92-95]. The recent study showed that local recurrence is also more often observed in NSCLC patients that have circulating tumor cells with small and irregular nuclei [96].

In LUAD patients, VEGFR-2 -906C>T and -271G>A variants are associated with tumor size and local recurrence. In particular, -906 CC and -271 GG genotypes significantly correlate with high frequency and early appearance of local relapse [97]. Local recurrence is also more often observed in NSCLC with NRF2/KEAP1 gene mutations compared to wild-type tumors [98] and high expression of MMP12 [99].

 

 

Regional recurrence

 

There are only a few studies regarding potential markers of regional recurrence of NSCLC. In the postoperative period, lymph node involvement is associated with intratumoral LVI (L1-status) [30]. Regional recurrence of NSCLC is also related to a low level of insulin-like growth factor binding protein (IGFBP5 and IGFBP7) in blood plasma[100]. In LUAD, regional recurrence is associated with overexpression of the CUB domain-containing protein (CDCP1) [101].

 

 

Distant recurrence

 

Clinicopathological markers

Non-squamous cell histology, pneumonectomy, and lymphatic invasion have been shown to be associated with distant metastasis in NSCLC patients [85, 95, 102]. Similar to locoregional recurrence, distant recurrence correlates to BVI, large tumor size, LVI, VPI, and STAS [16, 83-85, 87, 103-105]. In contrast to locoregional recurrence, distant metastases are associated with isolated BCH in the small bronchi distant from the tumor [21]. STAS is associated with a high risk of recurrence in LUSC patients undergoing lobectomy, but not sublobar resection; while in LUAD patients, STAS is a prognostic factor of recurrence in sublobar resection group [87]. Interestingly, another study reported that STAS is tightly linked to nodal and distant metastasis in LUAD patients, most of whom underwent lobectomy [106]. In addition, hematogenous metastases of LUAD are related to LVI and the cribriform component in the tumor [89, 107]. Similar to local recurrence, distant metastasis is more often observed in NSCLC patients that harbor circulating tumor cells with small and irregular nuclei [96].

 

Genetic markers

Germline variations. A high risk of distant recurrence is specific for NSCLC patients-carriers of the RETN rs3219175 AG and AG+AA genotypes [108]. Pleural metastasis is more frequently observed in LUAD patients with the EGFR rs712829 -216G/T and -216T/T genotypes [109]. Bone metastasis is associated with the CD44 rs187115 and the OPN 443 C/T polymorphisms, whereas the TGFB1 rs1800469, ATG10 rs10036653, ATG12 rs26532, and ATG16L1 rs2241880 variantscontribute to brain metastasis [110-113]. For example, a high risk of brain metastases was observed in rs26532 AC/CC carriers, while the rs10036653 AT/TT and rs2241880 AG/GG genotypes had a protective effect [111]. Moreover, the transfection of NSCLC cells with ATG16L1 rs2241880 -300T (threonine) encoded by A allele increases their metastatic potential to the brain in a mouse model [111].

Somatic alterations. The data regarding the association of genetic alterations with distant recurrence of NSCLC are contradictory. Mutations in the EGFR gene are associated with a high risk of distant recurrence in general [10] and metastasis to the brain [12, 13], liver [7], and pleura [9]. ALK gene rearrangements are associated with metastasis to the pleura, liver, and pericardium [7], as well as to the brain similar to the ROS1 and RET gene alterations [13, 114]. Mutations in the KRAS gene tend to be frequently detected in NSCLC patients with brain metastases [9]. At the same time, no association was found between metastases to the brain and ALK [7, 9], EGFR [7, 9, 115], and KRAS mutations [7, 115], liver metastases and ALK/EGFR alterations [9], and pleural metastases and aberrations in the EGFR [7] and ALK [9] genes.

 

Molecular markers

Gene and protein expression. Different genes and proteins whose expression is associated with the risk of distant recurrence and organotropic metastasis have been described (Table 2). Distant metastases are more frequently detected in NSCLC patients with overexpression of thymosinβ4 (TMSB4X), eukaryotic translation initiation factor 4A1 (eIF4A1), chemokine receptors CXCR4 and CXCR7, EMT-regulators MCRS1 and SPOCK1, cyclin E (CCNE1), α smooth muscle actin (ACTA2), tyrosine kinase receptor (IR, NTRK1, EGFR, ERBB2, ERBB3, PDGFR-β, FGFR1, and LTK), and underexpression of ribosomal S6 kinase 1 (RSK1) and huntingtin-interacting protein 1 (HIP1) [41, 116-124].NSCLC metastasis to the brain is associated with the expression of the GAP43 gene probably through the promotion of migration of cancer cells by Rac1 activation and prevention of F-actin depolymerization [125].

Several multigenic signatures have been proposed to predict the risk of NSCLC distant metastasis. Skrzypski and colleagues developed a panel of CSF1, EGFR, and CA9 (CA-IX) genes to predict the probability of LUSC metastasis (sensitivity 85%, specificity 60%) [6]. Grinberg-Rashi et al. showed that the expression of CDH2, KIFC1, and FALZ genes is associated with NSCLC metastasis to the brain [126]. Fregni and colleagues described a signature of 4 genes of mesenchymal stem cells (GREM1, LOXL2, ADAMTS12, and ITGA11), whose expression is associated with NSCLC metastasis [127].

Non-coding RNAs. A large number of microRNAs and long non-coding RNAs, whose expression correlates with distant recurrence of NSCLC and organotropic metastasis, have been identified (Table 2). In LUSC, increased risk of distant metastases is associated with expression of miR-10b, miR-662, miR-502-3p, miR-192*, miR-192, and miR-128 [128]. It is known that these microRNAs are involved in the regulation of proliferation (miR-10b and miR-128), apoptosis (miR-10b and miR-128), migration, and invasion of lung cancer cells (miR-10b, miR-192, and miR-662) as well as angiogenesis and lymphangiogenesis (miR-128) [129-133].

Brain metastasis of NSCLC is associated with low level of miR-590, miR-375, miR-145-5p, and miR-1280 and high expression of miR-219-2-3p, miR-219-5p, miR-124, miR-9*, miR-128, and miR-338-3p [134-136]. In LUAD, brain metastasis is also related to miR-214, miR-145, and miR-23a underexpression and overexpression of miR-9*, miR-1471, miR-718, miR-3656, miR-720, miR-423-5p, miR-184, and miR-197 [137-139]. It is known that miR-145, miR-590, miR-718, miR-124, miR-338-3p, and miR-184 [136, 140-144] inhibit proliferation, migration, and invasion of lung cancer cells, while miR-423-5p induces tumor progression [138]. miR-23a and miR-145-5p regulate ЕМТ, miR-9* controls cell cycle, and miR-718 and miR-197 induce apoptosis of lung cancer cells [144-148]. The contribution of miR-375 to the NSCLC prognosis is probably related to VEGF and MMP9 overexpression [134].

Bone metastasis correlates with overexpression of miR-21 and hsv2-miR-H9-5p [149, 150]. miR-21 is known to activate proliferation and inhibit apoptosis through suppression of cytochrome c oxidase (COX)-assembly protein COX-19, and hsv2-miR-H9-5p increases survival, migration, and invasion of lung cancer cells [149, 150].

The expression of lncRNA HOTAIR is significantly higher in brain metastases than in primary NSCLC and associated with low postoperative disease-free survival [151]. It is known that HOTAIR promotes proliferation, survival, invasion, metastasis, and drug resistance of lung cancer cells [152]. Overexpression of lncRNAs MALAT1 and AWPPH is associated with NSCLC metastasis [117, 153]. There is also data that the MALAT1 level is significantly higher in brain metastases of NSCLC [154]. Probably, MALAT1 promotes lung cancer brain metastasis by inducing EMT [154]. In addition, MALAT1 expression displays the strongest association with genes involved in cancer cell growth, movement, proliferation, signaling, and immune regulation [155]. AWPPH overexpression is known to upregulate TGF-β1 and promote NSCLC cell migration and invasion [153].

Based on microRNA profiling data, two signatures were proposed to predict the probability of LUAD metastasis to the brain: 1. miR-450b-3p, miR-29c, miR-145, miR-148a, miR-1, miR-30d, miR-187, miR-218, miR-708, and miR-375 [156] and 2. miR-210, miR-214, and miR-15a [5]. Also, Skrzypski and coauthors developed a signature of miR-10b, miR-662, miR-502-3p, miR-192*, miR-192, and miR-128 to predict LUSC metastasis risk and distant recurrence-free survival (RFS) [128].

Circulating molecules. Several serum proteins and circulating microRNAs have been shown to be associated with distant recurrence of NSCLC (Table 2). Metastases were more often detected in patients with a low level of miR-375 in blood plasma [157]. The high content of СЕА in the plasma of NSCLC patients is associated with brain metastasis [158, 159]. Probably, CEA-positive tumor cells could bond to brain vasculature, favoring central nervous system metastasis, similar to leukocyte transendothelial arrest and migration through the blood-brain barrier [158]. The target blockade of CEA with antibodies inhibits the adhesion, migration, and invasion of several tumor cell lines in vitro and in vivo [158]. Bone metastasis is associated with a low plasma level of miR-139-5p in LUAD patients [160]. miR-139-5p is known to inhibit lung cancer cell invasion [57].

 

Table 2. Molecular markers associated with distant recurrence of NSCLC. BMFS, brain metastasis-free survival; DFS, disease-free survival; LncRNAs, long non-coding RNAs; LUAD, adenocarcinoma; LUSC, lung squamous cell carcinoma; MFS, metastasis-free survival; NSCLC, non-small cell lung cancer; OS, overall survival; PFS, progression-free survival; RFS, recurrence/relapse-free survival; "-", not available. Numbers in brackets indicate the size of validation cohorts

 

 

Differences in the markers of NSCLC progression types

 

It is not surprising that the different types of NSCLC progression are distinct in the biology and clinical significance. However, in most studies, they, especially local, regional, and distant recurrences, are considered together. Here, we conducted a comparative analysis of the above-reviewed literature data to select markers specific for each type of NSCLC progression (Fig. 1).

Most of the clinicopathological parameters including LVI, STAS, tumor size, and some others are common for different types of NSCLC progression. Nevertheless, there are clinicopathological factors specific to certain types of NSCLC progression. In particular, local recurrence is more common in LUSC, whereas distant metastases – in non-squamous cell histology, particularly in LUAD [93, 95]. The underlying mechanisms are unclear; however, it is suggested that tobacco smoke being the main cause of LUSC may have a field effect predisposing to local disease recurrence [93]. In addition, the distant metastasis of NSCLC is related to isolated BCH in the small bronchi distant from the tumor [21], whereas the co-presence of BCH and SM is associated with a high probability of locoregional recurrence [22, 23]. The mechanisms of association between premalignant bronchial lesions and NSCLC progression remain unclear. Probably, individual features of immune-inflammatory reactions, on the one hand, can predispose to isolated BCH or the development of BCH and SM, whereas, on another hand, can promote NSCLC distant metastasis or locoregional recurrence. Distant recurrence of NSCLC is also specifically related to BVI that is considered as a step of tumor cell escape from the primary site to distant organs [83, 103, 105, 161].

In contrast to clinicopathological parameters, genetic and molecular markers common for different types of NSCLC progression are rare and include only EGFR, KRAS, and ALK mutations as well as expression of CXCR4, HOTAIR, and miR-9 and level of circulating CEA. Almost all molecular markers are specifically associated with one or another type of NSCLC progression. Nevertheless, most of these unique markers are related to the regulation of similar processes such as proliferation, cell growth and differentiation, apoptosis, EMT, migration, invasion, and angiogenesis. For example, all types of NSCLC progression are related to molecules involved in the regulation of cell migration: lymph node metastasis – miR-206, miR-101, ITGA11, S100A4, and RACK1 [40, 44, 50, 56, 61], local recurrence – MMP12 [99], regional recurrence – IGFBP5 [100], and distant recurrence – miR-590, miR-375, MALAT1, ACTA2, CD44, TMSB4X, GAP43, and RSK1 [112, 117-119, 125, 134, 136, 137, 154].

Thus, lymph node metastasis, local, regional, and distant recurrences of NSCLC differ in the spectrum of clinicopathological and molecular markers that once again emphasize their attitude to pathogenetically distinct types of cancer progression.

 

Fig. 1. Different types of NSCLC progression and associated markers. The left side of the figure shows the sites of metastases/relapses (red fields). Local recurrence means that cancer reappears in the same lung or at the bronchial stump. Regional recurrence is identified as a recurring disease in the ipsilateral hilar, mediastinal, and supraclavicular lymph nodes detected during follow-up after treatment. All other sites of recurrence outside the hemithorax or in the contralateral lung are referred to as distant recurrence or metastasis. The right side of the figure contains the markers associated with each type of NSCLC progression: clinicopathological parameters (yellow stripe), germline variations (green stripe), somatic alterations (blue stripe), gene and protein expression (violet stripe), miRNAs (orange stripe), long non-coding RNAs (pink stripe), and circulating molecules (red stripe). BCH, basal cell hyperplasia; BVI, blood vessel invasion; LUAD, lung adenocarcinoma; LUSC, lung squamous cell carcinoma; LVI, lymphovascular invasion; SM, squamous metaplasia; STAS, spread through air spaces; VPI, visceral pleural invasion.

 

 

Challenges and approaches to developing NSCLC progression markers

 

A number of clinicopathological parameters, genes, non-coding RNAs, and proteins have been described to be associated with the risk of NSCLC recurrence. However, the proposed markers are applicable mainly to LUAD and much less often to LUSC (Tables 1 and 2). Moreover, most of these markers have a number of significant limitations, such as insufficient predictive accuracy and economic and methodological complexity of analysis that prevents their validation in clinical practice.

Nevertheless, several prognostic markers, such as STAS, LVI, CEA, and mutations in the EGFR and ALK genes, described in multiple studies would seem to be reliable and valid. However, their translation into clinical practice is limited by several factors including lack of uniformity in the definition of STAS [162] and standardization in the methods used for STAS [163, 164] and LVI [165, 166] analysis, non-specificity of CEA for lung cancer [167, 168], its more sensitivity for LUAD, and unclear significance for non-adenocarcinomas [169], as well as contradictory results regarding the prognostic role of EGFR and ALK mutations in distant recurrence [7, 9, 12, 13, 114, 115]. Another problem is that all of these markers are associated with NSCLC progression, in general, that is heterogeneous and represented by clinically and molecularly distinct processes, particularly local, regional, and distant recurrences. In our opinion, the NSCLC prognosis should be built on the basis of markers specific to individual forms of tumor progression. Unfortunately, most of the proposed markers (Fig. 1) have been described in single studies, and additional research is required to validate them. The exception is BVI which has been reported in many studies to be associated predominantly with distant recurrence of NSCLC [83, 103, 105, 161]. However, there are many variations of BVI assessment methods and their standardization and quality control are needed [170].

The intermingling of different types of NSCLC progression and their presentation as cancer recurrence, in general, can be a possible explanation for the low effectiveness of suggested approaches to predict the risk of metastatic and recurrent disease. In this case, there also is some confusion regarding definitions of local, regional, and distant recurrence of NSCLC. For example, the definition of local recurrence, on which we relied, implies that cancer reappears in the same lung or at the bronchial stump [14-18]. However, according to other authors, local recurrence also includes recurring disease in ipsilateral hemithorax and mediastinum, pleural cavity, and lymph node regions [10, 95, 102, 103]. It is important to note that the term "regional recurrence" is often not used and the clinical manifestations of this type of progression are referred to as local recurrence [10, 95, 102, 103]. Regional recurrence is defined in this review as a recurring disease in the ipsilateral hilar, mediastinal, and supraclavicular lymph nodes detected during follow-up after treatment [9, 14, 19]. All other sites of recurrence are referred to as distant recurrence or metastasis [14]. Nevertheless, some studies refer relapse to second ipsilateral lobe or stump, as well as ipsilateral pleural recurrence, to regional recurrence, and metastases to supraclavicular lymph nodes are described as distant recurrence [16, 84, 87].

Also, the reason for the insufficient predictive power of the proposed markers may be tumor heterogeneity and the use of inadequate study models. Besides these challenges, the situation is complicated by the absence of highly effective methods to prevent NSCLC progression. Different therapeutics with anti-metastatic and anti-relapse effects have been suggested [171-173]; however, none of them are still used in clinical practice.

In our opinion, the use of the prognostic value of premalignant bronchial lesions can be one of the potential strategies to overcome the above-mentioned challenges. Previously, we showed that the probability of locoregional and distant recurrence of NSCLC is associated with the presence of premalignant lesions in the small bronchi distant from the primary tumor. In particular, the isolated BCH is related to increased distant metastasis and a decreased locoregional recurrence [21], whereas the co-presence of BCH and SM is associated with a high probability of locoregional recurrence and a low risk of distant metastasis [22, 23]. Importantly, neoadjuvant chemotherapy combined with intraoperative radiation shows high efficacy in the prevention of locoregional recurrence in high-risk NSCLC patients [23]. In this regard, we suggest that the analysis of premalignant changes in the bronchial epithelium at a distance from the primary tumor can be an effective instrument to assess the risk of NSCLC progression, whereas high- and low-risk patients may represent potential populations for the identification of highly reliable markers of cancer metastasis/recurrence. The analysis of the constitutive features of these patients, for example, germline variations and immune system parameters, and the investigation of the mutational landscape of their tumors can be also used to find potential molecular targets for the development of novel therapeutics aimed at the prevention of NSCLC progression.

 

 

Conclusion

 

The problem of prognosis and prevention of the risk of NSCLC progression is highly important. Despite advances in diagnostics and treatment, NSCLC continues to be diagnosed at the late stages. Even if NSCLC is detected early, the probability of recurrence is high. A huge amount of prognostic markers have been proposed; however, none of them are validated and recommended for the prediction of NSCLC progression in clinics. Moreover, there are no effective approaches to prevent cancer metastasis/recurrence. These problems are mainly related to the heterogeneous nature of NSCLC progression. Further studies should be conducted to investigate the mechanisms of different types of NSCLC progression including synchronous lymph node metastasis, local, regional, and distant recurrence, and to develop prognostic markers and potential therapeutic targets.

 

 

Abbreviations

 

BCH (basal cell hyperplasia); BVI (blood vessel invasion); LUAD (lung adenocarcinoma); LUSC (lung squamous cell carcinoma); LVI (lymphovascular invasion); NSCLC (non-small cell lung cancer); SM (squamous metaplasia); STAS (spread through air spaces); VPI (visceral pleural invasion).

 

 

Acknowledgements

 

We thank Ms. Ekaterina Khitrinskaya for the preparation of the figure.

 

Author Contributions

AAS and AAK wrote the text regarding molecular markers of different types of NSCLC progression. AAF and TSG focused on genetic markers. EOR, EBT, NAS, OVP, and AAD prepared subsections related to clinicopathological markers. MVZ, VMP, and EVD contributed to the conception and design and revised the review article.

 

Funding Source

This work was supported by the Russian Science Foundation [grant number 20-75-10060].

 

Statement of Ethics

The authors have no ethical conflicts to disclose.

 

 

Disclosure Statement

 

The authors have no conflicts of interest to declare.

 

 

References

 

1 Boyd JA, Hubbs JL, Kim DW, Hollis D, Marks LB, Kelsey CR: Timing of local and distant failure in resected lung cancer: implications for reported rates of local failure. J Thorac Oncol 2010;5:211-214.
https://doi.org/10.1097/JTO.0b013e3181c20080

 

2 Lou F, Huang J, Sima CS, Dycoco J, Rusch V, Bach PB: Patterns of recurrence and second primary lung cancer in early-stage lung cancer survivors followed with routine computed tomography surveillance. J Thorac Cardiovasc Surg 2013;145:75-81; discussion 81-72.
https://doi.org/10.1016/j.jtcvs.2012.09.030

 

3 Pfannschmidt J: Editorial on "Long-term survival outcome after postoperative recurrence of non-small cell lung cancer: who is 'cured' from postoperative recurrence?". J Thorac Dis 2018;10:610-613.
https://doi.org/10.21037/jtd.2018.01.02

 

4 Howlader N, Noone AM, Krapcho M, Miller D, Brest A, Yu M, et al.: SEER Cancer Statistics Review 1975-2016, National Cancer Institute, Bethesda, MD, USA. Updated April 9, 2020. URL: https://seer.cancer.gov/archive/csr/1975_2016/.

 

5 Zhao S, Yu J, Wang L: Machine Learning Based Prediction of Brain Metastasis of Patients with IIIA-N2 Lung Adenocarcinoma by a Three-miRNA Signature. Transl Oncol 2018;11:157-167.
https://doi.org/10.1016/j.tranon.2017.12.002

 

6 Skrzypski M, Jassem E, Taron M, Sanchez JJ, Mendez P, Rzyman W, et al.: Three-gene expression signature predicts survival in early-stage squamous cell carcinoma of the lung. Clin Cancer Res 2008;14:4794-4799.
https://doi.org/10.1158/1078-0432.CCR-08-0576

 

7 Doebele RC, Lu X, Sumey C, Maxson DA, Weickhardt AJ, Oton AB, et al.: Oncogene status predicts patterns of metastatic spread in treatment-naive nonsmall cell lung cancer. Cancer 2012;118:4502-4511.
https://doi.org/10.1002/cncr.27409

 

8 Hayashi H, Okamoto I, Kimura H, Sakai K, Nishimura Y, Nishio K, et al.: Clinical outcomes of thoracic radiotherapy for locally advanced NSCLC with EGFR mutations or EML4-ALK rearrangement. Anticancer Res 2012;32:4533-4537.

 

9 Mizuno T, Yatabe Y, Kuroda H, Sakakura N, Sakao Y: Impact of the oncogenic status on the mode of recurrence in resected non-small cell lung cancer. Jpn J Clin Oncol 2016;46:928-934.
https://doi.org/10.1093/jjco/hyw094

 

10 Na, II, Kim HR, Lee JK, Park SH, Kim CH, Koh JS, et al.: Epidermal growth factor receptor mutations in female patients with postoperative recurrent non-small-cell lung cancer. J Cancer Res Ther 2012;8:373-378.
https://doi.org/10.4103/0973-1482.103515

 

11 Paik JH, Choi CM, Kim H, Jang SJ, Choe G, Kim DK, et al.: Clinicopathologic implication of ALK rearrangement in surgically resected lung cancer: a proposal of diagnostic algorithm for ALK-rearranged adenocarcinoma. Lung Cancer 2012;76:403-409.
https://doi.org/10.1016/j.lungcan.2011.11.008

 

12 Shin DY, Na II, Kim CH, Park S, Baek H, Yang SH: EGFR mutation and brain metastasis in pulmonary adenocarcinomas. J Thorac Oncol 2014;9:195-199.
https://doi.org/10.1097/JTO.0000000000000069

 

13 Wang H, Wang Z, Zhang G, Zhang M, Zhang X, Li H, et al.: Driver genes as predictive indicators of brain metastasis in patients with advanced NSCLC: EGFR, ALK, and RET gene mutations. Cancer Med 2020;9:487-495.
https://doi.org/10.1002/cam4.2706

 

14 Martini N, Bains MS, Burt ME, Zakowski MF, McCormack P, Rusch VW, et al.: Incidence of local recurrence and second primary tumors in resected stage I lung cancer. J Thorac Cardiovasc Surg 1995;109:120-129.
https://doi.org/10.1016/S0022-5223(95)70427-2

 

15 Nishio W, Yoshimura M, Maniwa Y, Kitamura Y, Tane K, Takenaka D, et al.: Re-Assessment of Intentional Extended Segmentectomy for Clinical T1aN0 Non-Small Cell Lung Cancer. Ann Thorac Surg 2016;102:1702-1710.
https://doi.org/10.1016/j.athoracsur.2016.05.071

 

16 Kadota K, Nitadori JI, Sima CS, Ujiie H, Rizk NP, Jones DR, et al.: Tumor Spread through Air Spaces is an Important Pattern of Invasion and Impacts the Frequency and Location of Recurrences after Limited Resection for Small Stage I Lung Adenocarcinomas. J Thorac Oncol 2015;10:806-814.
https://doi.org/10.1097/JTO.0000000000000486

 

17 Kadota K, Kushida Y, Kagawa S, Ishikawa R, Ibuki E, Inoue K, et al.: Limited Resection Is Associated With a Higher Risk of Locoregional Recurrence than Lobectomy in Stage I Lung Adenocarcinoma With Tumor Spread Through Air Spaces. Am J Surg Pathol 2019;43:1033-1041.
https://doi.org/10.1097/PAS.0000000000001285

 

18 Koike T, Koike T, Yoshiya K, Tsuchida M, Toyabe S: Risk factor analysis of locoregional recurrence after sublobar resection in patients with clinical stage IA non-small cell lung cancer. J Thorac Cardiovasc Surg 2013;146:372-378.
https://doi.org/10.1016/j.jtcvs.2013.02.057

 

19 Eriguchi T, Takeda A, Tsurugai Y, Sanuki N, Kibe Y, Hara Y, et al.: Pleural contact decreases survival in clinical T1N0M0 lung cancer patients undergoing SBRT. Radiother Oncol 2019;134:191-198.
https://doi.org/10.1016/j.radonc.2019.02.005

 

20 Sekihara K, Hishida T, Yoshida J, Oki T, Omori T, Katsumata S, et al.: Long-term survival outcome after postoperative recurrence of non-small-cell lung cancer: who is 'cured' from postoperative recurrence? Eur J Cardiothorac Surg 2017;52:522-528.
https://doi.org/10.1093/ejcts/ezx127

 

21 Perelmuter VM, Pankova OV: Method for prediction of onset of hematogenous metastases accompanying squamous cell lung cancer. Russian Federation Patent RU 2489718 C1. August 10, 2013.

 

22 Pankova OV, Denisov EV, Ponomaryova AA, Gerashchenko TS, Tuzikov SA, Perelmuter VM: Recurrence of squamous cell lung carcinoma is associated with the co-presence of reactive lesions in tumor-adjacent bronchial epithelium. Tumour Biol 2016;37:3599-3607.
https://doi.org/10.1007/s13277-015-4196-2

 

23 Pankova OV, Rodionov EO, Miller SV, Tuzikov SA, Tashireva LA, Gerashchenko TS, et al.: Neoadjuvant chemotherapy combined with intraoperative radiotherapy is effective to prevent recurrence in high-risk non- small cell lung cancer (NSCLC) patients. Translational lung cancer research 2020;9:988-999.
https://doi.org/10.21037/tlcr-19-719

 

24 Cagle PT, Allen TC, Dacic S, Beasley MB, Borczuk AC, Chirieac LR, et al.: Revolution in lung cancer: new challenges for the surgical pathologist. Arch Pathol Lab Med 2011;135:110-116.
https://doi.org/10.5858/2010-0567-RA.1

 

25 Bao F, Yuan P, Yuan X, Lv X, Wang Z, Hu J: Predictive risk factors for lymph node metastasis in patients with small size non-small cell lung cancer. J Thorac Dis 2014;6:1697-1703.

 

26 Cho S, Song IH, Yang HC, Kim K, Jheon S: Predictive factors for node metastasis in patients with clinical stage I non-small cell lung cancer. Ann Thorac Surg 2013;96:239-245.
https://doi.org/10.1016/j.athoracsur.2013.03.050

 

27 Seok Y, Yang HC, Kim TJ, Lee KW, Kim K, Jheon S, et al.: Frequency of lymph node metastasis according to the size of tumors in resected pulmonary adenocarcinoma with a size of 30 mm or smaller. J Thorac Oncol 2014;9:818-824.
https://doi.org/10.1097/JTO.0000000000000169

 

28 Zhang J, Gold KA, Lin HY, Swisher SG, Xing Y, Lee JJ, et al.: Relationship between tumor size and survival in non-small-cell lung cancer (NSCLC): an analysis of the surveillance, epidemiology, and end results (SEER) registry. J Thorac Oncol 2015;10:682-690.
https://doi.org/10.1097/JTO.0000000000000456

 

29 Shan Q, Fan Y, Guo J, Han X, Wang H, Wang Z: Relationship between tumor size and metastatic site in patients with stage IV non-small cell lung cancer: A large SEER-based study. PeerJ 2019;7:e7822.
https://doi.org/10.7717/peerj.7822

 

30 Moulla Y, Gradistanac T, Wittekind C, Eichfeld U, Gockel I, Dietrich A: Predictive risk factors for lymph node metastasis in patients with resected non-small cell lung cancer: a case control study. J Cardiothorac Surg 2019;14:11.
https://doi.org/10.1186/s13019-019-0831-0

 

31 Suzuki K, Nagai K, Yoshida J, Nishimura M, Nishiwaki Y: Predictors of lymph node and intrapulmonary metastasis in clinical stage IA non-small cell lung carcinoma. Ann Thorac Surg 2001;72:352-356.
https://doi.org/10.1016/S0003-4975(01)02748-5

 

32 Chung CC, Magalhaes WC, Gonzalez-Bosquet J, Chanock SJ: Genome-wide association studies in cancer--current and future directions. Carcinogenesis 2010;31:111-120.
https://doi.org/10.1093/carcin/bgp273

 

33 Wang J, Liu Q, Yuan S, Xie W, Liu Y, Xiang Y, et al.: Genetic predisposition to lung cancer: comprehensive literature integration, meta-analysis, and multiple evidence assessment of candidate-gene association studies. Sci Rep 2017;7:8371.
https://doi.org/10.1038/s41598-017-07737-0

 

34 Yan S, Sun R, Wu S, Jin T, Zhang S, Niu F, et al.: Single nucleotide polymorphism in the 3' untranslated region of LPP is a risk factor for lung cancer: a case-control study. BMC Cancer 2019;19:35.
https://doi.org/10.1186/s12885-018-5241-5

 

35 Fang S, Jin X, Wang R, Li Y, Guo W, Wang N, et al.: Polymorphisms in the MMP1 and MMP3 promoter and non-small cell lung carcinoma in North China. Carcinogenesis 2005;26:481-486.
https://doi.org/10.1093/carcin/bgh327

 

36 Vogelstein B, Papadopoulos N, Velculescu VE, Zhou S, Diaz LA, Kinzler KW: Cancer Genome Landscapes. Science 2013;339:1546-1558.
https://doi.org/10.1126/science.1235122

 

37 Cancer Genome Atlas Research Network: Comprehensive molecular profiling of lung adenocarcinoma. Nature 2014;511:543-550.
https://doi.org/10.1038/nature13385

 

38 Cancer Genome Atlas Research Network: Comprehensive genomic characterization of squamous cell lung cancers. Nature 2012;489:519-525.
https://doi.org/10.1038/nature11404

 

39 Seo JS, Ju YS, Lee WC, Shin JY, Lee JK, Bleazard T, et al.: The transcriptional landscape and mutational profile of lung adenocarcinoma. Genome Res 2012;22:2109-2119.
https://doi.org/10.1101/gr.145144.112

 

40 Chong IW, Chang MY, Chang HC, Yu YP, Sheu CC, Tsai JR, et al.: Great potential of a panel of multiple hMTH1, SPD, ITGA11 and COL11A1 markers for diagnosis of patients with non-small cell lung cancer. Oncol Rep 2006;16:981-988.
https://doi.org/10.3892/or.16.5.981

 

41 Liang JX, Gao W, Liang Y, Zhou XM: Chemokine receptor CXCR4 expression and lung cancer prognosis: a meta-analysis. Int J Clin Exp Med 2015;8:5163-5174.

 

42 Jing P, Zhao N, Ye M, Zhang Y, Zhang Z, Sun J, et al.: Protein arginine methyltransferase 5 promotes lung cancer metastasis via the epigenetic regulation of miR-99 family/FGFR3 signaling. Cancer Lett 2018;427:38-48.
https://doi.org/10.1016/j.canlet.2018.04.019

 

43 Ning Y, Liu W, Guan X, Xie X, Zhang Y: CPSF3 is a promising prognostic biomarker and predicts recurrence of non-small cell lung cancer. Oncol Lett 2019;18:2835-2844.
https://doi.org/10.3892/ol.2019.10659

 

44 Chen XL, Zhang WG, Chen XY, Sun ZM, Liu SH: [Correlations of S100A4 protein expression to invasion and metastasis of non-small cell lung cancer]. Ai Zheng 2006;25:1134-1137.

 

45 Biaoxue R, Xiling J, Shuanying Y, Wei Z, Xiguang C, Jinsui W, et al.: Upregulation of Hsp90-beta and annexin A1 correlates with poor survival and lymphatic metastasis in lung cancer patients. J Exp Clin Cancer Res 2012;31:70.
https://doi.org/10.1186/1756-9966-31-70

 

46 Bai SY, Xu N, Chen C, Song YL, Hu J, Bai CX: Integrin αvβ5 as a biomarker for the assessment of non-small cell lung cancer metastasis and overall survival. Clin Respir J 2015;9:457-467.
https://doi.org/10.1111/crj.12163

 

47 Zhang B, Zhang H, Shen G: Metastasis-associated protein 2 (MTA2) promotes the metastasis of non-small-cell lung cancer through the inhibition of the cell adhesion molecule Ep-CAM and E-cadherin. Jpn J Clin Oncol 2015;45:755-766.
https://doi.org/10.1093/jjco/hyv062

 

48 Xing Y, Meng Q, Chen X, Zhao Y, Liu W, Hu J, et al.: TRIM44 promotes proliferation and metastasis in non‑small cell lung cancer via mTOR signaling pathway. Oncotarget 2016;7:30479-30491.
https://doi.org/10.18632/oncotarget.8586

 

49 Wang Z, Li Z, Wu C, Wang Y, Xia Y, Chen L, et al.: MACC1 overexpression predicts a poor prognosis for non-small cell lung cancer. Med Oncol 2014;31:790.
https://doi.org/10.1007/s12032-013-0790-6

 

50 Zhong X, Li M, Nie B, Wu F, Zhang L, Wang E, et al.: Overexpressions of RACK1 and CD147 associated with poor prognosis in stage T1 pulmonary adenocarcinoma. Ann Surg Oncol 2013;20:1044-1052.
https://doi.org/10.1245/s10434-012-2377-4

 

51 Li N, Li S: RASAL2 promotes lung cancer metastasis through epithelial-mesenchymal transition. Biochem Biophys Res Commun 2014;455:358-362.
https://doi.org/10.1016/j.bbrc.2014.11.020

 

52 Diamantopoulos MA, Tsiakanikas P, Scorilas A: Non-coding RNAs: the riddle of the transcriptome and their perspectives in cancer. Ann Transl Med 2018;6:241.
https://doi.org/10.21037/atm.2018.06.10

 

53 Wang R, Wang ZX, Yang JS, Pan X, De W, Chen LB: MicroRNA-451 functions as a tumor suppressor in human non-small cell lung cancer by targeting ras-related protein 14 (RAB14). Oncogene 2011;30:2644-2658.
https://doi.org/10.1038/onc.2010.642

 

54 Luo W, Huang B, Li Z, Li H, Sun L, Zhang Q, et al.: MicroRNA-449a is downregulated in non-small cell lung cancer and inhibits migration and invasion by targeting c-Met. PLoS One 2013;8:e64759.
https://doi.org/10.1371/journal.pone.0064759

 

55 Wang J, Tian X, Han R, Zhang X, Wang X, Shen H, et al.: Downregulation of miR-486-5p contributes to tumor progression and metastasis by targeting protumorigenic ARHGAP5 in lung cancer. Oncogene 2014;33:1181-1189.
https://doi.org/10.1038/onc.2013.42

 

56 Lin L, Gu ZT, Chen WH, Cao KJ: Increased expression of the long non-coding RNA ANRIL promotes lung cancer cell metastasis and correlates with poor prognosis. Diagn Pathol 2015;10:14.
https://doi.org/10.1186/s13000-015-0247-7

 

57 Watanabe K, Amano Y, Ishikawa R, Sunohara M, Kage H, Ichinose J, et al.: Histone methylation-mediated silencing of miR-139 enhances invasion of non-small-cell lung cancer. Cancer Med 2015;4:1573-1582.
https://doi.org/10.1002/cam4.505

 

58 Mao M, Wu Z, Chen J: MicroRNA-187-5p suppresses cancer cell progression in non-small cell lung cancer (NSCLC) through down-regulation of CYP1B1. Biochem Biophys Res Commun 2016;478:649-655.
https://doi.org/10.1016/j.bbrc.2016.08.001

 

59 Tang T, Huan L, Zhang S, Zhou H, Gu L, Chen X, et al.: MicroRNA-212 functions as a tumor-suppressor in human non-small cell lung cancer by targeting SOX4. Oncol Rep 2017;38:2243-2250.
https://doi.org/10.3892/or.2017.5885

 

60 Ye Z, Fang B, Pan J, Zhang N, Huang J, Xie C, et al.: miR-138 suppresses the proliferation, metastasis and autophagy of non-small cell lung cancer by targeting Sirt1. Oncol Rep 2017;37:3244-3252.
https://doi.org/10.3892/or.2017.5619

 

61 Han L, Chen W, Xia Y, Song Y, Zhao Z, Cheng H, et al.: MiR-101 inhibits the proliferation and metastasis of lung cancer by targeting zinc finger E-box binding homeobox 1. Am J Transl Res 2018;10:1172-1183.

 

62 Liu B, Wu X, Liu B, Wang C, Liu Y, Zhou Q, et al.: MiR-26a enhances metastasis potential of lung cancer cells via AKT pathway by targeting PTEN. Biochim Biophys Acta 2012;1822:1692-1704.
https://doi.org/10.1016/j.bbadis.2012.07.019

 

63 Xu T, Liu X, Han L, Shen H, Liu L, Shu Y: Up-regulation of miR-9 expression as a poor prognostic biomarker in patients with non-small cell lung cancer. Clin Transl Oncol 2014;16:469-475.
https://doi.org/10.1007/s12094-013-1106-1

 

64 Ma J, Mannoor K, Gao L, Tan A, Guarnera MA, Zhan M, et al.: Characterization of microRNA transcriptome in lung cancer by next-generation deep sequencing. Mol Oncol 2014;8:1208-1219.
https://doi.org/10.1016/j.molonc.2014.03.019

 

65 Wu L, Hu B, Zhao B, Liu Y, Yang Y, Zhang L, et al.: Circulating microRNA-422a is associated with lymphatic metastasis in lung cancer. Oncotarget 2017;8:42173-42188.
https://doi.org/10.18632/oncotarget.15025

 

66 Mo D, Gu B, Gong X, Wu L, Wang H, Jiang Y, et al.: miR-1290 is a potential prognostic biomarker in non-small cell lung cancer. J Thorac Dis 2015;7:1570-1579.

 

67 Li WQ, Zhang JP, Wang YY, Li XZ, Sun L: MicroRNA-422a functions as a tumor suppressor in non-small cell lung cancer through SULF2-mediated TGF-β/SMAD signaling pathway. Cell Cycle 2019;18:1727-1744.
https://doi.org/10.1080/15384101.2019.1632135

 

68 Han L, Kong R, Yin DD, Zhang EB, Xu TP, De W, et al.: Low expression of long noncoding RNA GAS6-AS1 predicts a poor prognosis in patients with NSCLC. Med Oncol 2013;30:694.
https://doi.org/10.1007/s12032-013-0694-5

 

69 Xiao L, Zhou H, Li XP, Chen J, Fang C, Mao CX, et al.: MicroRNA-138 acts as a tumor suppressor in non small cell lung cancer via targeting YAP1. Oncotarget 2016;7:40038-40046.
https://doi.org/10.18632/oncotarget.9480

 

70 Jin JJ, Liu YH, Si JM, Ni R, Wang J: Overexpression of miR-1290 contributes to cell proliferation and invasion of non small cell lung cancer by targeting interferon regulatory factor 2. Int J Biochem Cell Biol 2018;95:113-120.
https://doi.org/10.1016/j.biocel.2017.12.017

 

71 Xu G, Shao G, Pan Q, Sun L, Zheng D, Li M, et al.: MicroRNA-9 regulates non-small cell lung cancer cell invasion and migration by targeting eukaryotic translation initiation factor 5A2. Am J Transl Res 2017;9:478-488.

 

72 Liu XH, Liu ZL, Sun M, Liu J, Wang ZX, De W: The long non-coding RNA HOTAIR indicates a poor prognosis and promotes metastasis in non-small cell lung cancer. BMC Cancer 2013;13:464.
https://doi.org/10.1186/1471-2407-13-464

 

73 Qiu M, Xu Y, Yang X, Wang J, Hu J, Xu L, et al.: CCAT2 is a lung adenocarcinoma-specific long non-coding RNA and promotes invasion of non-small cell lung cancer. Tumour Biol 2014;35:5375-5380.
https://doi.org/10.1007/s13277-014-1700-z

 

74 Sun M, Liu XH, Wang KM, Nie FQ, Kong R, Yang JS, et al.: Downregulation of BRAF activated non-coding RNA is associated with poor prognosis for non-small cell lung cancer and promotes metastasis by affecting epithelial-mesenchymal transition. Mol Cancer 2014;13:68.
https://doi.org/10.1186/1476-4598-13-68

 

75 Wu Y, Lyu H, Liu H, Shi X, Song Y, Liu B: Downregulation of the long noncoding RNA GAS5-AS1 contributes to tumor metastasis in non-small cell lung cancer. Sci Rep 2016;6:31093.
https://doi.org/10.1038/srep31093

 

76 Zhao W, An Y, Liang Y, Xie XW: Role of HOTAIR long noncoding RNA in metastatic progression of lung cancer. Eur Rev Med Pharmacol Sci 2014;18:1930-1936.

 

77 Cree IA: Liquid biopsy for cancer patients: Principles and practice. Pathogenesis 2015;2:1-4.
https://doi.org/10.1016/j.pathog.2015.05.001

 

78 Dou H, Wang Y, Su G, Zhao S: Decreased plasma let-7c and miR-152 as noninvasive biomarker for non-small-cell lung cancer. Int J Clin Exp Med 2015;8:9291-9298.

 

79 Wang Z, Cai M, Weng Y, Zhang F, Meng D, Song J, et al.: Circulating MACC1 as a novel diagnostic and prognostic biomarker for nonsmall cell lung cancer. J Cancer Res Clin Oncol 2015;141:1353-1361.
https://doi.org/10.1007/s00432-014-1903-0

 

80 Cheng Z, Ma R, Tan W, Zhang L: MiR-152 suppresses the proliferation and invasion of NSCLC cells by inhibiting FGF2. Exp Mol Med 2014;46:e112.
https://doi.org/10.1038/emm.2014.51

 

81 Zhao B, Han H, Chen J, Zhang Z, Li S, Fang F, et al.: MicroRNA let-7c inhibits migration and invasion of human non-small cell lung cancer by targeting ITGB3 and MAP4K3. Cancer Lett 2014;342:43-51.
https://doi.org/10.1016/j.canlet.2013.08.030

 

82 Luo H, Qiao L, Liang N, Zhang J: Risk factors for recurrence in patients with resected N1 non-small cell lung cancer - a systematic review and meta-analysis. J BUON 2015;20:791-799.

 

83 Dziedzic DA, Rudzinski P, Langfort R, Orlowski T: Risk Factors for Local and Distant Recurrence After Surgical Treatment in Patients With Non-Small-Cell Lung Cancer. Clin Lung Cancer 2016;17:e157-e167.
https://doi.org/10.1016/j.cllc.2015.12.013

 

84 Tian D, Pei Y, Zheng Q, Zhang J, Li S, Wang X, et al.: Effect of visceral pleural invasion on the prognosis of patients with lymph node negative non-small cell lung cancer. Thorac Cancer 2017;8:97-105.
https://doi.org/10.1111/1759-7714.12412

 

85 Shiono S, Endo M, Suzuki K, Hayasaka K, Yanagawa N: Spread through air spaces in lung cancer patients is a risk factor for pulmonary metastasis after surgery. J Thorac Dis 2019;11:177-187.
https://doi.org/10.21037/jtd.2018.12.21

 

86 El-Sherif A, Fernando HC, Santos R, Pettiford B, Luketich JD, Close JM, et al.: Margin and local recurrence after sublobar resection of non-small cell lung cancer. Ann Surg Oncol 2007;14:2400-2405.
https://doi.org/10.1245/s10434-007-9421-9

 

87 Lu S, Tan KS, Kadota K, Eguchi T, Bains S, Rekhtman N, et al.: Spread through Air Spaces (STAS) Is an Independent Predictor of Recurrence and Lung Cancer-Specific Death in Squamous Cell Carcinoma. J Thorac Oncol 2017;12:223-234.
https://doi.org/10.1016/j.jtho.2016.09.129

 

88 Eguchi T, Kameda K, Lu S, Bott MJ, Tan KS, Montecalvo J, et al.: Lobectomy Is Associated with Better Outcomes than Sublobar Resection in Spread through Air Spaces (STAS)-Positive T1 Lung Adenocarcinoma: A Propensity Score-Matched Analysis. J Thorac Oncol 2019;14:87-98.
https://doi.org/10.1016/j.jtho.2018.09.005

 

89 Ding Q, Chen D, Wang X, Wen J, Chen C, Zhang Y, et al.: Characterization of lung adenocarcinoma with a cribriform component reveals its association with spread through air spaces and poor outcomes. Lung Cancer 2019;134:238-244.
https://doi.org/10.1016/j.lungcan.2019.06.027

 

90 Song SY, Jeong SY, Park HJ, Park SI, Kim DK, Kim YH, et al.: Clinical significance of NQO1 C609T polymorphisms after postoperative radiation therapy in completely resected non-small cell lung cancer. Lung Cancer 2010;68:278-282.
https://doi.org/10.1016/j.lungcan.2009.06.009

 

91 Yanagawa N, Shiono S, Abiko M, Ogata SY, Sato T, Tamura G: Prognostic impact and initial recurrence site of lymphovascular and visceral pleural invasion in surgically resected stage I non-small-cell lung carcinoma. Eur J Cardiothorac Surg 2013;44:e200-206.
https://doi.org/10.1093/ejcts/ezt309

 

92 Masai K, Sakurai H, Sukeda A, Suzuki S, Asakura K, Nakagawa K, et al.: Prognostic Impact of Margin Distance and Tumor Spread Through Air Spaces in Limited Resection for Primary Lung Cancer. J Thorac Oncol 2017;12:1788-1797.
https://doi.org/10.1016/j.jtho.2017.08.015

 

93 Kelsey CR, Marks LB, Hollis D, Hubbs JL, Ready NE, D'Amico TA, et al.: Local recurrence after surgery for early stage lung cancer: an 11-year experience with 975 patients. Cancer 2009;115:5218-5227.
https://doi.org/10.1002/cncr.24625

 

94 Woody NM, Stephans KL, Andrews M, Zhuang T, Gopal P, Xia P, et al.: A Histologic Basis for the Efficacy of SBRT to the lung. J Thorac Oncol 2017;12:510-519.
https://doi.org/10.1016/j.jtho.2016.11.002

 

95 Hung JJ, Jeng WJ, Hsu WH, Chou TY, Huang BS, Wu YC: Predictors of death, local recurrence, and distant metastasis in completely resected pathological stage-I non-small-cell lung cancer. J Thorac Oncol 2012;7:1115-1123.
https://doi.org/10.1097/JTO.0b013e31824cbad8

 

96 Wu Y, Niu Y, Lv F, Gao W, Shen X: Morphology classification of circulating tumor cells could be a predictor of recurrent disease in patients with non-small cell lung cancer after surgery. J Clin Oncol 2020;38:e15530-e15530.
https://doi.org/10.1200/JCO.2020.38.15_suppl.e15530

 

97 Uzunoglu FG, Kaufmann C, Wikman H, Güngör C, Bohn BA, Nentwich MF, et al.: Vascular endothelial growth factor receptor 2 gene polymorphisms as predictors for tumor recurrence and overall survival in non-small-cell lung cancer. Ann Surg Oncol 2012;19:2159-2168.
https://doi.org/10.1245/s10434-012-2227-4

 

98 Jeong Y, Hoang NT, Lovejoy A, Stehr H, Newman AM, Gentles AJ, et al.: Role of KEAP1/NRF2 and TP53 Mutations in Lung Squamous Cell Carcinoma Development and Radiation Resistance. Cancer Discov 2017;7:86-101.
https://doi.org/10.1158/2159-8290.CD-16-0127

 

99 Hofmann HS, Hansen G, Richter G, Taege C, Simm A, Silber RE, et al.: Matrix metalloproteinase-12 expression correlates with local recurrence and metastatic disease in non-small cell lung cancer patients. Clin Cancer Res 2005;11:1086-1092.

 

100 Shersher DD, Vercillo MS, Fhied C, Basu S, Rouhi O, Mahon B, et al.: Biomarkers of the insulin-like growth factor pathway predict progression and outcome in lung cancer. Ann Thorac Surg 2011;92:1805-1811; discussion 1811.
https://doi.org/10.1016/j.athoracsur.2011.06.058

 

101 Ikeda J, Oda T, Inoue M, Uekita T, Sakai R, Okumura M, et al.: Expression of CUB domain containing protein (CDCP1) is correlated with prognosis and survival of patients with adenocarcinoma of lung. Cancer Sci 2009;100:429-433.
https://doi.org/10.1111/j.1349-7006.2008.01066.x

 

102 Varlotto JM, Recht A, Flickinger JC, Medford-Davis LN, Dyer AM, Decamp MM: Factors associated with local and distant recurrence and survival in patients with resected nonsmall cell lung cancer. Cancer 2009;115:1059-1069.
https://doi.org/10.1002/cncr.24133

 

103 Shoji F, Haro A, Yoshida T, Ito K, Morodomi Y, Yano T, et al.: Prognostic significance of intratumoral blood vessel invasion in pathologic stage IA non-small cell lung cancer. Ann Thorac Surg 2010;89:864-869.
https://doi.org/10.1016/j.athoracsur.2009.09.047

 

104 Sung SY, Kwak YK, Lee SW, Jo IY, Park JK, Kim KS, et al.: Lymphovascular Invasion Increases the Risk of Nodal and Distant Recurrence in Node-Negative Stage I-IIA Non-Small-Cell Lung Cancer. Oncology 2018;95:156-162.
https://doi.org/10.1159/000488859

 

105 Bodendorf MO, Haas V, Laberke HG, Blumenstock G, Wex P, Graeter T: Prognostic value and therapeutic consequences of vascular invasion in non-small cell lung carcinoma. Lung Cancer 2009;64:71-78.
https://doi.org/10.1016/j.lungcan.2008.07.011

 

106 Warth A, Muley T, Kossakowski CA, Goeppert B, Schirmacher P, Dienemann H, et al.: Prognostic Impact of Intra-alveolar Tumor Spread in Pulmonary Adenocarcinoma. Am J Surg Pathol 2015;39:793-801.
https://doi.org/10.1097/PAS.0000000000000409

 

107 Higgins KA, Chino JP, Ready N, D'Amico TA, Berry MF, Sporn T, et al.: Lymphovascular invasion in non-small-cell lung cancer: implications for staging and adjuvant therapy. J Thorac Oncol 2012;7:1141-1147.
https://doi.org/10.1097/JTO.0b013e3182519a42

 

108 Hu WW, Tang CH, Sun Y, Lu TT, Jiang P, Wu YM, et al.: Correlation between resistin gene polymorphism and clinical aspects of lung cancer. Medicine (Baltimore) 2017;96:e9485.
https://doi.org/10.1097/MD.0000000000009485

 

109 Guo H, Xing Y, Liu R, Chen S, Bian X, Wang F, et al.: -216G/T (rs712829), a functional variant of the EGFR promoter, is associated with the pleural metastasis of lung adenocarcinoma. Oncol Lett 2013;6:693-698.
https://doi.org/10.3892/ol.2013.1442

 

110 Chen Y, Liu H, Wu W, Li Y, Li J: Osteopontin genetic variants are associated with overall survival in advanced non-small-cell lung cancer patients and bone metastasis. J Exp Clin Cancer Res 2013;32:45.
https://doi.org/10.1186/1756-9966-32-45

 

111 Li QX, Zhou X, Huang TT, Tang Y, Liu B, Peng P, et al.: The Thr300Ala variant of ATG16L1 is associated with decreased risk of brain metastasis in patients with non-small cell lung cancer. Autophagy 2017;13:1053-1063.
https://doi.org/10.1080/15548627.2017.1308997

 

112 Liu Y, Qing H, Su X, Wang C, Li Z, Liu S: Association of CD44 Gene Polymorphism with Survival of NSCLC and Risk of Bone Metastasis. Med Sci Monit 2015;21:2694-2700.
https://doi.org/10.12659/MSM.894357

 

113 Wang HB, Song WG, Liu HQ, Fang F, Xiao Y: Role of TGFB1 polymorphism in the development of metastatic brain tumors in non-small cell lung cancer patients. Genet Mol Res 2015;14:3545-3550.
https://doi.org/10.4238/2015.April.17.3

 

114 Patil T, Smith DE, Bunn PA, Aisner DL, Le AT, Hancock M, et al.: The Incidence of Brain Metastases in Stage IV ROS1-Rearranged Non-Small Cell Lung Cancer and Rate of Central Nervous System Progression on Crizotinib. J Thorac Oncol 2018;13:1717-1726.
https://doi.org/10.1016/j.jtho.2018.07.001

 

115 Hendriks LE, Smit EF, Vosse BA, Mellema WW, Heideman DA, Bootsma GP, et al.: EGFR mutated non-small cell lung cancer patients: more prone to development of bone and brain metastases? Lung Cancer 2014;84:86-91.
https://doi.org/10.1016/j.lungcan.2014.01.006

 

116 Iwakiri S, Mino N, Takahashi T, Sonobe M, Nagai S, Okubo K, et al.: Higher expression of chemokine receptor CXCR7 is linked to early and metastatic recurrence in pathological stage I nonsmall cell lung cancer. Cancer 2009;115:2580-2593.
https://doi.org/10.1002/cncr.24281

 

117 Ji P, Diederichs S, Wang W, Boing S, Metzger R, Schneider PM, et al.: MALAT-1, a novel noncoding RNA, and thymosin beta4 predict metastasis and survival in early-stage non-small cell lung cancer. Oncogene 2003;22:8031-8041.
https://doi.org/10.1038/sj.onc.1206928

 

118 Lara R, Mauri FA, Taylor H, Derua R, Shia A, Gray C, et al.: An siRNA screen identifies RSK1 as a key modulator of lung cancer metastasis. Oncogene 2011;30:3513-3521.
https://doi.org/10.1038/onc.2011.61

 

119 Lee HW, Park YM, Lee SJ, Cho HJ, Kim DH, Lee JI, et al.: Alpha-smooth muscle actin (ACTA2) is required for metastatic potential of human lung adenocarcinoma. Clin Cancer Res 2013;19:5879-5889.
https://doi.org/10.1158/1078-0432.CCR-13-1181

 

120 Liu MX, Zhou KC, Cao Y: MCRS1 overexpression, which is specifically inhibited by miR-129*, promotes the epithelial-mesenchymal transition and metastasis in non-small cell lung cancer. Mol Cancer 2014;13:245.
https://doi.org/10.1186/1476-4598-13-245

 

121 Miao L, Wang Y, Xia H, Yao C, Cai H, Song Y: SPOCK1 is a novel transforming growth factor-beta target gene that regulates lung cancer cell epithelial-mesenchymal transition. Biochem Biophys Res Commun 2013;440:792-797.
https://doi.org/10.1016/j.bbrc.2013.10.024

 

122 Muller-Tidow C, Diederichs S, Bulk E, Pohle T, Steffen B, Schwable J, et al.: Identification of metastasis-associated receptor tyrosine kinases in non-small cell lung cancer. Cancer Res 2005;65:1778-1782.
https://doi.org/10.1158/0008-5472.CAN-04-3388

 

123 Muller-Tidow C, Metzger R, Kugler K, Diederichs S, Idos G, Thomas M, et al.: Cyclin E is the only cyclin-dependent kinase 2-associated cyclin that predicts metastasis and survival in early stage non-small cell lung cancer. Cancer Res 2001;61:647-653.

 

124 Hsu CY, Lin CH, Jan YH, Su CY, Yao YC, Cheng HC, et al.: Huntingtin-Interacting Protein-1 Is an Early-Stage Prognostic Biomarker of Lung Adenocarcinoma and Suppresses Metastasis via Akt-mediated Epithelial-Mesenchymal Transition. Am J Respir Crit Care Med 2016;193:869-880.
https://doi.org/10.1164/rccm.201412-2226OC

 

125 Zhang F, Ying L, Jin J, Feng J, Chen K, Huang M, et al.: GAP43, a novel metastasis promoter in non-small cell lung cancer. J Transl Med 2018;16:310.
https://doi.org/10.1186/s12967-018-1682-5

 

126 Grinberg-Rashi H, Ofek E, Perelman M, Skarda J, Yaron P, Hajdúch M, et al.: The Expression of Three Genes in Primary Non-Small Cell Lung Cancer Is Associated with Metastatic Spread to the Brain. Clin Cancer Res 2009;15:1755-1761.
https://doi.org/10.1158/1078-0432.CCR-08-2124

 

127 Fregni G, Quinodoz M, Möller E, Vuille J, Galland S, Fusco C, et al.: Reciprocal modulation of mesenchymal stem cells and tumor cells promotes lung cancer metastasis. EBioMedicine 2018;29:128-145.
https://doi.org/10.1016/j.ebiom.2018.02.017

 

128 Skrzypski M, Czapiewski P, Goryca K, Jassem E, Wyrwicz L, Pawlowski R, et al.: Prognostic value of microRNA expression in operable non-small cell lung cancer patients. Br J Cancer 2014;110:991-1000.
https://doi.org/10.1038/bjc.2013.786

 

129 Filipska M, Skrzypski M, Czetyrbok K, Stokowy T, Stasiłojć G, Supernat A, et al.: MiR-192 and miR-662 enhance chemoresistance and invasiveness of squamous cell lung carcinoma. Lung Cancer 2018;118:111-118.
https://doi.org/10.1016/j.lungcan.2018.02.002

 

130 Hu J, Cheng Y, Li Y, Jin Z, Pan Y, Liu G, et al.: 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.
https://doi.org/10.1016/j.ejca.2014.06.005

 

131 Huang J, Sun C, Wang S, He Q, Li D: microRNA miR-10b inhibition reduces cell proliferation and promotes apoptosis in non-small cell lung cancer (NSCLC) cells. Mol Biosyst 2015;11:2051-2059.
https://doi.org/10.1039/C4MB00752B

 

132 Li M, Fu W, Wo L, Shu X, Liu F, Li C: miR-128 and its target genes in tumorigenesis and metastasis. Exp Cell Res 2013;319:3059-3064.
https://doi.org/10.1016/j.yexcr.2013.07.031

 

133 Liu Y, Li M, Zhang G, Pang Z: MicroRNA-10b overexpression promotes non-small cell lung cancer cell proliferation and invasion. Eur J Med Res 2013;18:41.
https://doi.org/10.1186/2047-783X-18-41

 

134 Chen LJ, Li XY, Zhao YQ, Liu WJ, Wu HJ, Liu J, et al.: Down-regulated microRNA-375 expression as a predictive biomarker in non-small cell lung cancer brain metastasis and its prognostic significance. Pathol Res Pract 2017;213:882-888.
https://doi.org/10.1016/j.prp.2017.06.012

 

135 Donzelli S, Mori F, Bellissimo T, Sacconi A, Casini B, Frixa T, et al.: Epigenetic silencing of miR-145-5p contributes to brain metastasis. Oncotarget 2015;6:35183-35201.
https://doi.org/10.18632/oncotarget.5930

 

136 Wang FF, Wang S, Xue WH, Cheng JL: microRNA-590 suppresses the tumorigenesis and invasiveness of non-small cell lung cancer cells by targeting ADAM9. Mol Cell Biochem 2016;423:29-37.
https://doi.org/10.1007/s11010-016-2822-y

 

137 Zhao C, Xu Y, Zhang Y, Tan W, Xue J, Yang Z, et al.: Downregulation of miR-145 contributes to lung adenocarcinoma cell growth to form brain metastases. Oncol Rep 2013;30:2027-2034.
https://doi.org/10.3892/or.2013.2728

 

138 Sun G, Ding X, Bi N, Wu L, Wang J, Zhang W, et al.: MiR-423-5p in brain metastasis: potential role in diagnostics and molecular biology. Cell Death Dis 2018;9:936.
https://doi.org/10.1038/s41419-018-0955-5

 

139 Remon J, Alvarez-Berdugo D, Majem M, Moran T, Reguart N, Lianes P: miRNA-197 and miRNA-184 are associated with brain metastasis in EGFR-mutant lung cancers. Clin Transl Oncol 2016;18:153-159.
https://doi.org/10.1007/s12094-015-1347-2

 

140 Cho WC, Chow AS, Au JS: MiR-145 inhibits cell proliferation of human lung adenocarcinoma by targeting EGFR and NUDT1. RNA Biol 2011;8:125-131.
https://doi.org/10.4161/rna.8.1.14259

 

141 Hong-Yuan W, Xiao-Ping C: miR-338-3p suppresses epithelial-mesenchymal transition and metastasis in human nonsmall cell lung cancer. Indian J Cancer 2015;52:E168-171.
https://doi.org/10.4103/0019-509X.186569

 

142 Lin J, Xu K, Wei J, Heimberger AB, Roth JA, Ji L: MicroRNA-124 suppresses tumor cell proliferation and invasion by targeting CD164 signaling pathway in non-small cell lung cancer. J Gene Ther 2016;2:6.
https://doi.org/10.13188/2381-3326.1000006

 

143 Lin TC, Lin PL, Cheng YW, Wu TC, Chou MC, Chen CY, et al.: MicroRNA-184 Deregulated by the MicroRNA-21 Promotes Tumor Malignancy and Poor Outcomes in Non-small Cell Lung Cancer via Targeting CDC25A and c-Myc. Ann Surg Oncol 2015;22:S1532-1539.
https://doi.org/10.1245/s10434-015-4595-z

 

144 Wang S, Sun H, Zhan X, Wang Q: MicroRNA‑718 serves a tumor‑suppressive role in non‑small cell lung cancer by directly targeting CCNB1. Int J Mol Med 2020;45:33-44.
https://doi.org/10.3892/ijmm.2019.4396

 

145 Cao M, Seike M, Soeno C, Mizutani H, Kitamura K, Minegishi Y, et al.: MiR-23a regulates TGF-β-induced epithelial-mesenchymal transition by targeting E-cadherin in lung cancer cells. Int J Oncol 2012;41:869-875.
https://doi.org/10.3892/ijo.2012.1535

 

146 Fiori ME, Barbini C, Haas TL, Marroncelli N, Patrizii M, Biffoni M, et al.: Antitumor effect of miR-197 targeting in p53 wild-type lung cancer. Cell Death Differ 2014;21:774-782.
https://doi.org/10.1038/cdd.2014.6

 

147 Su YH, Zhou Z, Yang KP, Wang XG, Zhu Y, Fa XE: MIR-142-5p and miR-9 may be involved in squamous lung cancer by regulating cell cycle related genes. Eur Rev Med Pharmacol Sci 2013;17:3213-3220.

 

148 Yin Q, Han Y, Zhu D, Li Z, Shan S, Jin W, et al.: miR-145 and miR-497 suppress TGF-β-induced epithelial-mesenchymal transition of non-small cell lung cancer by targeting MTDH. Cancer Cell Int 2018;18:105.
https://doi.org/10.1186/s12935-018-0601-4

 

149 Wang X, Liu S, Zhou Z, Yan H, Xiao J: A herpes simplex virus type 2-encoded microRNA promotes tumor cell metastasis by targeting suppressor of cytokine signaling 2 in lung cancer. Tumour Biol 2017;39:1010428317701633.
https://doi.org/10.1177/1010428317701633

 

150 Guo Q, Zhang H, Zhang L, He Y, Weng S, Dong Z, et al.: MicroRNA-21 regulates non-small cell lung cancer cell proliferation by affecting cell apoptosis via COX-19. Int J Clin Exp Med 2015;8:8835-8841.

 

151 Nakagawa T, Endo H, Yokoyama M, Abe J, Tamai K, Tanaka N, et al.: Large noncoding RNA HOTAIR enhances aggressive biological behavior and is associated with short disease-free survival in human non-small cell lung cancer. Biochem Biophys Res Commun 2013;436:319-324.
https://doi.org/10.1016/j.bbrc.2013.05.101

 

152 Loewen G, Jayawickramarajah J, Zhuo Y, Shan B: Functions of lncRNA HOTAIR in lung cancer. J Hematol Oncol 2014;7:90.
https://doi.org/10.1186/s13045-014-0090-4

 

153 Tang L, Wang T, Zhang Y, Zhang J, Zhao H, Wang H, et al.: Long Non-Coding RNA AWPPH Promotes Postoperative Distant Recurrence in Resected Non-Small Cell Lung Cancer by Upregulating Transforming Growth Factor beta 1 (TGF-β1). Med Sci Monit 2019;25:2535-2541.
https://doi.org/10.12659/MSM.912876

 

154 Shen L, Chen L, Wang Y, Jiang X, Xia H, Zhuang Z: Long noncoding RNA MALAT1 promotes brain metastasis by inducing epithelial-mesenchymal transition in lung cancer. J Neurooncol 2015;121:101-108.
https://doi.org/10.1007/s11060-014-1613-0

 

155 Schmidt LH, Spieker T, Koschmieder S, Schäffers S, Humberg J, Jungen D, et al.: The long noncoding MALAT-1 RNA indicates a poor prognosis in non-small cell lung cancer and induces migration and tumor growth. J Thorac Oncol 2011;6:1984-1992.
https://doi.org/10.1097/JTO.0b013e3182307eac

 

156 Lu Y, Govindan R, Wang L, Liu PY, Goodgame B, Wen W, et al.: MicroRNA profiling and prediction of recurrence/relapse-free survival in stage I lung cancer. Carcinogenesis 2012;33:1046-1054.
https://doi.org/10.1093/carcin/bgs100

 

157 Yu H, Jiang L, Sun C, Li Guo L, Lin M, Huang J, et al.: Decreased circulating miR-375: a potential biomarker for patients with non-small-cell lung cancer. Gene 2014;534:60-65.
https://doi.org/10.1016/j.gene.2013.10.024

 

158 Arrieta O, Saavedra-Perez D, Kuri R, Aviles-Salas A, Martinez L, Mendoza-Posada D, et al.: Brain metastasis development and poor survival associated with carcinoembryonic antigen (CEA) level in advanced non-small cell lung cancer: a prospective analysis. BMC Cancer 2009;9:119.
https://doi.org/10.1186/1471-2407-9-119

 

159 Lee DS, Kim YS, Jung SL, Lee KY, Kang JH, Park S, et al.: The relevance of serum carcinoembryonic antigen as an indicator of brain metastasis detection in advanced non-small cell lung cancer. Tumour Biol 2012;33:1065-1073.
https://doi.org/10.1007/s13277-012-0344-0

 

160 Xu S, Yang F, Liu R, Li X, Fan H, Liu J, et al.: Serum microRNA-139-5p is downregulated in lung cancer patients with lytic bone metastasis. Oncol Rep 2018;39:2376-2384.
https://doi.org/10.3892/or.2018.6316

 

161 Kessler R, Gasser B, Massard G, Roeslin N, Meyer P, Wihlm JM, et al.: Blood vessel invasion is a major prognostic factor in resected non-small cell lung cancer. Ann Thorac Surg 1996;62:1489-1493.
https://doi.org/10.1016/0003-4975(96)00540-1

 

162 Warth A: Spread through air spaces (STAS): a comprehensive update. Transl Lung Cancer Res 2017;6:501-507.
https://doi.org/10.21037/tlcr.2017.06.08

 

163 Jia M, Yu S, Gao H, Sun PL: Spread Through Air Spaces (STAS) in Lung Cancer: A Multiple-Perspective and Update Review. Cancer Manag Res 2020;12:2743-2752.
https://doi.org/10.2147/CMAR.S249790

 

164 Mino-Kenudson M: Significance of tumor spread through air spaces (STAS) in lung cancer from the pathologist perspective. Transl Lung Cancer Res 2020;9:847-859.
https://doi.org/10.21037/tlcr.2020.01.06

 

165 He KW, Sun JJ, Liu ZB, Zhuo PY, Ma QH, Liu ZY, et al.: Prognostic significance of lymphatic vessel invasion diagnosed by D2-40 in Chinese invasive breast cancers. Medicine (Baltimore) 2017;96:e8490.
https://doi.org/10.1097/MD.0000000000008490

 

166 Mollberg NM, Bennette C, Howell E, Backhus L, Devine B, Ferguson MK: Lymphovascular invasion as a prognostic indicator in stage I non-small cell lung cancer: a systematic review and meta-analysis. Ann Thorac Surg 2014;97:965-971.
https://doi.org/10.1016/j.athoracsur.2013.11.002

 

167 Akcam TI, Cagirici U, Ergonul AG, Ozdil A, Kavurmaci O, Turhan K, et al.: The prognostic value of carcinoembryonic antigen levels in blood and intraoperative pleural lavage fluid in non-small-cell lung cancer. Kardiochir Torakochirurgia Pol 2017;14:104-109.
https://doi.org/10.5114/kitp.2017.68740

 

168 Abbas M, Kassim SA, Habib M, Li X, Shi M, Wang ZC, et al.: Clinical Evaluation of Serum Tumor Markers in Patients With Advanced-Stage Non-Small Cell Lung Cancer Treated With Palliative Chemotherapy in China. Front Oncol 2020;10:800.
https://doi.org/10.3389/fonc.2020.00800

 

169 Shintani T, Matsuo Y, Iizuka Y, Mitsuyoshi T, Mizowaki T, Hiraoka M: Prognostic Significance of Serum CEA for Non-small Cell Lung Cancer Patients Receiving Stereotactic Body Radiotherapy. Anticancer Res 2017;37:5161-5167.
https://doi.org/10.21873/anticanres.11937

 

170 Wang J, Chen J, Chen X, Wang B, Li K, Bi J: Blood vessel invasion as a strong independent prognostic indicator in non-small cell lung cancer: a systematic review and meta-analysis. PLoS One 2011;6:e28844.
https://doi.org/10.1371/journal.pone.0028844

 

171 Fontebasso Y, Dubinett SM: Drug Development for Metastasis Prevention. Crit Rev Oncog 2015;20:449-473.
https://doi.org/10.1615/CritRevOncog.v20.i5-6.150

 

172 Gandalovičová A, Rosel D, Fernandes M, Veselý P, Heneberg P, Čermák V, et al.: Migrastatics-Anti-metastatic and Anti-invasion Drugs: Promises and Challenges. Trends Cancer 2017;3:391-406.
https://doi.org/10.1016/j.trecan.2017.04.008

 

173 Steeg PS: Targeting metastasis. Nat Rev Cancer 2016;16:201-218.
https://doi.org/10.1038/nrc.2016.25