Open Access

NKX2‑1 copy number alterations are associated with oncogenic, immunological and prognostic remodeling in non‑small cell lung cancer

  • Authors:
    • Herdee Gloriane C. Luna
    • Marcelo Severino Imasa
    • Necy Juat
    • Katherine V. Hernandez
    • Treah May Sayo
    • Gloria Cristal‑Luna
    • Sheena Marie Asur‑Galang
    • Mirasol Bellengan
    • Kent John Duga
    • Bien Brian Buenaobra
    • Marvin I. De Los Santos
    • Daniel Medina
    • Jamirah Samo
    • Venus Minerva Literal
    • Sullian Sy‑Naval
  • View Affiliations

  • Published online on: May 8, 2024     https://doi.org/10.3892/ol.2024.14436
  • Article Number: 303
  • Copyright: © Luna et al. This is an open access article distributed under the terms of Creative Commons Attribution License [CC BY_NC 4.0].

Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

NK2 homeobox 1 (NKX2‑1) copy number alterations (CNAs) are frequently observed in lung cancer. However, little is known about the complete landscape of focal alterations in NKX2‑1 copy number (CN), their clinical significance and their therapeutic implications in non‑small cell lung cancer (NSCLC). The correlations between NKX2‑1 expression and EGFR driver mutations and programmed death ligand 1 (PD‑L1) co‑expression were studied using immunohistochemistry and PCR from the tumors of recruited Filipino patients (n=45). Clinical features of NSCLC with NKX2‑1 CNAs were resolved at the tumor and clonal levels using the molecular profiles of patients with lung adenocarcinoma and lung squamous cell carcinoma from The Cancer Genome Atlas (n=1,130), and deconvoluted single‑cell RNA‑seq data from the Bivona project (n=1,654), respectively. Despite a significant and positive correlation between expression and CN (r=0.264; P<0.001), NKX2‑1 CNAs exerted a stronger influence on the combined EGFR and PD‑L1 status of NSCLC tumors than expression. NKX2‑1 CN gain was prognostic of favorable survival (P=0.018) and a better response to targeted therapy. NKX2‑1 CN loss predicted a worse survival (P=0.041). Mutational architecture in the Y‑chromosome differentiated the two prognostic groups. There were 19,941 synonymous mutations and 1,408 genome‑wide CN perturbations associated with NKX2‑1 CNAs. Tumors with NKX2‑1 CN gain expressed lymphocyte markers more heterogeneously than those with CN loss. Higher expression of tumor‑infiltrating lymphocyte gene signatures in CN gain was prognostic of longer disease‑free survival (P=0.005). Tumors with NKX2‑1 CN gain had higher B‑cell (P<0.001) and total T‑cell estimates (P=0.003). NKX2‑1 CN loss was associated with immunologically colder tumors due to higher M2 macrophage infiltrates (P=0.011) and higher expression of immune checkpoint proteins, CD274 (P=0.025), VTCN1 (P<0.001) and LGALS9 (P=0.002). In conclusion, NKX2‑1 CNAs are associated with tumors that exhibit clinically diverse characteristics, and with unique oncogenic, immunological and prognostic signatures.

Introduction

Copy number alterations (CNAs) refer to the gain or loss of sections of genetic material, which can contribute to disease development by modifying gene dosage and functionality (1,2). In non-small cell lung cancer (NSCLC), CNAs are common and diverse, affecting critical biological processes that promote tumorigenesis (3,4). Genome-wide copy number (CN) analysis has been proposed as a diagnostic, pathological and prognostic tool for NSCLC (57).

The investigation of focal CNAs in programmed death ligand 1 (PD-L1) and EGFR genes has gained interest due to their close association with targeted therapy. Notably, alterations in PD-L1 CN have been shown to influence PD-L1 immunopositivity, tumor proportion score (TPS) and patient survival (810). Meanwhile, variations in EGFR CN have been found to influence the overall survival of NSCLC patients with EGFR-mutated tumors and can predict lung cancer metastasis to the brain (11,12). These findings have indicated the importance of CNA analysis in understanding lung cancer pathophysiology and predicting treatment outcomes in patients with lung cancer. However, CNAs are complex and dynamic, and the landscape of CN perturbations in NSCLC remains largely unexplored.

Previously, we reported that the lineage-survival oncogene NK2 homeobox 1 (NKX2-1), also known as thyroid transcription factor 1 (TTF-1), may upregulate the expression of oncogenic proteins, and could influence the signaling pathways associated with EGFR and PD-L1 (13). Additionally, NKX2-1 CN is significantly correlated with NKX2-1 protein expression (14), which justifies the interest in studying the prognostic implications of focal CN amplification (14,15). However, NKX2-1 CN is variable in lung cancer and only ~15% of cases of NSCLC have amplifications (16). Thus, the elucidation and comparison of tumor biology in NSCLC with focal loss and gain of NKX2-1 CN remain poorly investigated in lung cancer. Additionally, the clinical significance of NKX2-1 CNAs and their association with targeted therapy remain elusive. The present study reports on a comprehensive investigation involving the molecular features associated with focal alterations in NKX2-1 CN, their prognostic significance in patient survival, association with chromosomal instability and implications for targeted therapy.

Materials and methods

Study design and cohorts

The present study was conducted using clinical data from the Clinical Proteomics for Cancer Initiative program described elsewhere (Filipino cohort) (13,17,18); lung adenocarcinoma (LUAD) and lung squamous cell carcinoma (LUSC) data from The Cancer Genome Atlas (TCGA cohort) (19); and longitudinal lung cancer single-cell RNA-sequencing (scRNA-seq) project data (Bivona cohort) (20). All participants were confirmed as having NSCLC with either adenocarcinoma or squamous histology. A total of 45, 49 and 1,130 enrolled participants were included in the analyses from the Filipino, Bivona and TCGA cohorts, respectively. The tumor molecular data of TCGA cohort were obtained from 501 patients with LUSC and 629 with LUAD. The tumor sequencing data from the Bivona cohort had 1,654 unique scRNA-seq profiles. In the Filipino cohort, NKX2-1/TTF-1 and PD-L1 expression levels were analyzed using immunohistochemistry (IHC), whereas EGFR mutational frequency was determined using PCR. In both TCGA and Bivona cohorts, expression levels, mutational frequencies and CNs were retrieved using the curated databases of UCSC Xena platform (21) and GDC Data Portal (22). The study involving the Filipino cohort was approved by The Institutional Ethics Review Board (approval no. LCP-CS-001-2019) of the Lung Center of the Philippines (Quezon City, Philippines). All patients provided written informed consent for genetic testing, as well as for the use of their clinical data. The clinicodemographic and pathological deidentified data of TCGA and Bivona cohorts were described in each study publication. The clinicopathological data of the Filipino cohort are described in Table SI.

PCR analysis and IHC

In accordance with the status as a multicenter study, the protocol for specimen collection was approved by The Single Joint Research Ethics Board of the Department of Health, Manila, Philippines (approval no. SJREB-2020-97). Blood and tissues samples were collected from recruited Filipino patients with NSCLC and were immediately processed for plasma testing, histopathological examination or cryopreservation at −80°C, as previously described (18). The specimens were tested for the presence of EGFR-sensitizing mutations using PCR, and assayed for PD-L1 and TTF-1 protein expression using IHC. EGFR mutations were assessed using the AmoyDx EGFR 29 Mutations Detection Kit (designed to detect mutations in EGFR exons 19–21; Amoy Diagnostics Co., Ltd.) as previously described (23). EGFR without driver mutations, with single and co-mutations were scored as 0, 1 and 2, respectively. Expression of PD-L1 and NKX2-1/TTF-1 in NSCLC were evaluated by IHC using the pharmDx 22C3 kit (cat no. SK006; Agilent Technologies, Inc.) (24) and clone 8G7G3/1 (cat no. M3575; Agilent Technologies, Inc.) (25), respectively. After pathology confirmation, expression was classified as negative (scored 0) if the TPS was <1%, or positive (scored as 1) if TPS was ≥1%. The representative histopathological data are shown in Fig. S1.

Analysis of tumor molecular profiles

Gene expression, copy number variations (CNVs), mutational frequencies and treatment modalities of patients from TCGA and Bivona cohorts were obtained through the UCSC Xena platform (https://xenabrowser.net). Gene expression units in TCGA cohort (LUAD and LUSC) were expressed in log2(TPM+1,) while expression units in the Bivona cohort were equalized to the depth of sequencing per cell, where log-normalized counts were scaled by linear regression against the number of reads (scaled expression) (20). CNVs in TCGA and Bivona cohorts were expressed as log2-transformed (https://xenabrowser.net/datapages/) and raw proportions of tumor to normal counts (https://singlecell.xenabrowser.net/datapages/), respectively. CNAs were synchronized between cohorts by deriving log2-transformed values from raw proportion data, or vice versa. List of driver genes, type of driver mutations, presence of secondary mutations and treatment interventions of participants in the Bivona cohort were queried using the ‘Phenotypic Data’ field, whereas whole genome CNV profile was queried using the ‘Analytic Data’ field. Tumor mutational architecture (TMA) in chromosomes 7, X, Y, 15 and 14 were analyzed using tumor profiles in TCGA cohort by querying ‘Somatic Mutation Dataset’ with chromosomal length according to hg19 assembly. Tumor mutational burden (TMB) of LUAD and LUSC tumors in TCGA cohort were extracted from the Data Exploration of GDC Data Portal (https://portal.gdc.cancer.gov/exploration). Proteome and phosphoproteome data of tumor samples were retrieved from Proteomic Data Commons (https://proteomic.datacommons.cancer.gov/pdc/) with IDs PDC000153, PDC000149 and PDC000149.

Survival analysis

Survival of patients with NSCLC (LUAD and LUSC) in TCGA cohort were analyzed using the Kaplan Meier Plot through the Xena platform (21) and survival analysis in Gene Expression Profiling Interactive Analysis (26). Median cutoff was used to assess the survival of patients with high and low CN or gene expression. Quartile cutoff was used to assess the survival associated with CN gain and loss. Binary grouping was used to compare certain subgroups with the rest of the cohort. Log-rank P-values were reported for each survival analysis with P<0.05 being considered significant.

Gene set operations and clustering analysis

Mutational signatures associated with NKX2-1 CN gain (>0.1875; n=251) and loss (<-0.0767; n=251) were assessed by comparing the TMB of patients with CN gain and loss with the overall TMB of participants in TCGA cohort (n=1,130). Unique mutational features of the two prognostic groups were identified by performing ‘Set Operations Analysis’ in the GDC Data Portal (https://portal.gdc.cancer.gov/analysis). Mutational clusters were presented as classic or Edwards’ constructed Venn diagram generated using jvenn (27). Frequency of genes with commonly or uniquely altered CN between the two groups was identified using the set operation in jvenn (http://jvenn.toulouse.inra.fr). Symbols of genes that were significantly mutated or had altered CNs in patients with NKX2-1 CN gain and loss were plotted as a word cloud using WordClouds (https://www.wordclouds.com/).

Enrichment analysis

Functional annotation of molecular, cellular and biological processes that were potentially affected by genetic mutations and alteration of CNs was performed using Enrichr (28,29). Affected pathways were enriched using BioPlanet (30) and Kyoto Encyclopedia of Genes and Genomes (31), while perturbed processes were enriched using Gene Ontology (32). Chromosomal locations (hg19) were enriched using the sequence annotation of UCSC Genome Browser (33). Subcellular enrichment was performed using COMPARTMENTS (34), and cellular enrichments were performed using CellMarker (35) and Human Gene Atlas in BioGPS (36) curations. Enrichments with P<0.05 were considered significant.

Tumor-infiltrating lymphocyte (TIL) gene marker analysis

The expression signature of 53 widely used gene markers for TILs (20,37,38) from TCGA cohort was analyzed against NKX2-1 CNVs. Significantly correlating genes that were common or unique in patients with NKX2-1 CN gain and loss were identified and visualized using jvenn. Direction of linear correlation (positive or negative) and labelling of significant gene markers was visualized as volcano plots using VolcaNoseR (39).

Heatmap analysis

Expression levels of JAK, STAT and TIL marker genes were plotted against NKX2-1 expression or CNV without data labels. Unsupervised clustering analysis was performed through Heatmapper using Euclidian average linkage (40). Cluster dendrograms were applied to data columns.

TIL proportion estimation

Immune infiltration estimation using the gene expression profiles of 53 TIL markers was performed using CIBERSORT (41), CIBERSORT-ABS (41), MCPCounter (42) and TIMER (43) algorithms through TIMER 2.0 (http://timer.cistrome.org/). Proportions of estimated lymphocytes were compared between NKX2-1 CN gain and loss. Overlapping immune estimation was consolidated based on the highest absolute significance or lowest P-value.

Statistical analysis

Data are presented as the mean ± standard deviation using box charts, percentages as pie charts and frequencies/distributions as bar graphs. Frequency statistics were used to describe each result of IHC and PCR tests of participants in the Filipino cohort, NKX2-1 CNV in TCGA cohort, as well as treatment modalities and genome-wide CNV differences between patient groups in the Bivona cohort. Pearson's correlation analysis was used to compare the trend and significance of two variables in one or two groups. One sample t-test was used to identify the variability of NKX2-1 CN among NSCLC tumors. Independent sample t-test was used to determine the statistical difference of two samples, while one-way ANOVA (without post hoc test) was used to determine the statistical difference of three or more samples. P-values and different levels of significance were reported in the figures and/or data legends. All statistical analysis was performed using JASP version 0.16.3 (University of Amsterdam). P<0.05 was considered to indicate a statistically significant difference.

Results

NKX2-1 CNAs have a stronger correlation with combined EGFR and PD-L1 status of NSCLC tumors than expression

The correlation of NKX2-1/TTF-1 expression with the co-occurrence of EGFR-sensitizing mutations and PD-L1 co-expression in the Filipino cohort was first examined. Immunohistochemical analysis revealed that most tumors expressed NKX2-1/TTF-1 (98%) and more than half expressed PD-L1 (51%). The majority of participants had exon19del (44%) and exon21L858R (33%) mutations (Fig. 1A-C). Pearson's correlation revealed that NKX2-1/TTF-1 expression was significantly associated with EGFR mutations (P=0.039). By contrast, PD-L1 positivity had no significant correlation with NKX2-1/TTF-1 expression (P=0.334) or EGFR mutation (P=0.096) (Fig. 1D-F). This observation was in agreement with previous reports (44,45) and may justify their independent prognostic value.

A previous expression-based analysis in adenocarcinoma proposed the potential involvement of NKX2-1 in EGFR and PD-L1 signaling through the PI3K-STAT pathway (13). In vitro experiments found that NKX2-1 transactivates the receptor tyrosine kinase-like orphan receptor 1 (ROR1), which in turn promotes EGFR-induced ERBB3-dependent activation of the PI3K pathway (46). ERBB3 also regulates the expression of PD-L1 through the activation of the PI3K/PDK1/RSK/CREB signaling axis (47). In addition, ROR1 activates the proto-oncogene tyrosine-protein kinase Src (c-Src), which in turn activates the PI3K pathway (46) and constitutively activates STAT signaling (48). STAT signaling is known to regulate the expression of PD-L1 (49). This NKX2-1-mediated ROR1-dependent PI3K-STAT signaling activation could link the influence of NKX2-1 in the combined EGFR and PD-L1 status of lung tumors. Notably, a previous study partially revealed the crosstalk between EGFR and PD-L1 pathways through interleukin-mediated PI3K and STAT signaling (50,51). Expression analysis in NSCLC showed that ROR1 transcript levels were significantly correlated with the upregulation of NKX2-1 (r=0.499; P<0.001), and ROR1 protein levels were positively associated with ERBB3 (r=0.221; P<0.01), PIK3s (r=0.222–0.266; P<0.01), STATs (r=0.282–0.306; P<0.001) and c-SRC (r=0.186; P<0.01) protein expression (Fig. S2A and B). Phosphoproteome analysis of LUAD tumors retrieved from Proteomic Data Commons revealed that the phosphorylation of ERBB3 was positively associated with phosphoactivation of c-SRC at different serine, threonine and tyrosine residues (Fig. S2C). Additionally, ERBB3 phosphorylation was positively associated with the phosphorylation of EGFR and PIK3 proteins (Fig. S2D), confirming the contribution of ERBB3-dependent activation of the PI3K pathway in NSCLC. Lastly, c-SRC phosphorylation was also positively associated with the phosphorylation of PIK3 and STAT proteins (Fig. S2E), confirming the c-SRC-mediated activation of PI3K and STAT signaling in NSCLC. Collectively, these findings support the involvement of NKX2-1 in influencing the combined EGFR and PD-L1 status in tumors through ROR1 signaling.

Next, the expression levels of PIK3 and STAT genes in NSCLC tumors from TCGA cohort were examined. Pearson's correlation analysis revealed that NKX2-1 expression was significantly correlated with the positive regulation of PIK3CG, PIK3CD, PIK3R5, PIK3R6, PIK3C2A/B, STAT3, STAT4, STAT5A/B and STAT6 (all P<0.001). Meanwhile, CNV had a positive correlation with PIK3C2A and STAT4 only (both P<0.01; Fig. S2F and G), despite a strong positive correlation detected between NKX2-1 CNV and expression (P<0.001; Fig. 2A). The variability of NKX2-1 CN among NSCLC tumors was significantly high (P<0.001; Fig. 2B). Subgrouping according to NKX2-1 CN gain and loss revealed that CN loss, but not gain, was significantly correlated with the positive regulation of PIK3CG, PIK3CD, PIK3R5, PIK3R6, STAT4, STAT5A (all P<0.001) and STAT5B (P<0.05; Fig. S2H and I). Unsupervised heatmap analysis revealed the close association between NKX2-1 gene expression and CN loss (Fig. S2J). These results signify that CNAs may influence the combined EGFR and PD-L1 profiles of patients with lung cancer.

To test this hypothesis, the molecular profiles of patients with NSCLC from TCGA and Bivona cohorts were explored. Consistent with the IHC results, NKX2-1 transcript levels did not correlate with CD274 (PD-L1) expression (P=0.184; Fig. 2C). However, NKX2-1 CNAs were significantly correlated with CD274 downregulation (P=0.047 in loss; P=0.001 in gain) and EGFR upregulation (P<0.001 in gain), at the clonal level (Fig. 2D-G). EGFR-driver mutations in tumors with NKX2-1 CNAs were significantly higher compared to other oncogenes such as ALK, BRAF and KRAS (70 vs. 15, 14 and 1%, respectively; P<0.001; Fig. 2H). Additionally, NKX2-1 and EGFR CNVs were found co-altered in NSCLC (P=0.008; Fig. 2I). These results suggested that the EGFR and PD-L1 profiles of NSCLC tumors may be strongly associated with NKX2-1 CNAs rather than expression.

NKX2-1 CNAs differentially prognose the survival of patients with NSCLC

Germline and somatic CNVs may influence the survival of patients with lung cancer (7). Therefore, the prognostic value of NKX2-1 CNAs, with and without germline CNVs was examined. The results showed that NKX2-1 CN with and without germline CNVs had positive and significant correlation (r=0.996; P<0.001), which suggested that NKX2-1 CNAs were most likely acquired somatically (Fig. S3A). Survival analysis based on high and low NKX2-1 CN yielded comparable survival curves, regardless of whether they were with or without germline CNVs (P=0.079 and P=0.066, respectively; Fig. S3B and C). Quartile subgrouping by NKX2-1 CNAs showed that patients with NKX2-1 CN loss had significantly shorter survival than those with CN gain, with (P=0.019) or without germline CNVs (P=0.004; Fig. 3A and B).

The present study aimed to identify the contribution of NKX2-1 CNAs that developed somatically and defined the threshold of NKX2-1 CN gain [>0.1875 log2(tumor/normal)] and NKX2-1 CN loss [<-0.0767 log2(tumor/normal)]. When the survival of these two patient subgroups were compared with the rest of the NSCLC cohort, it was found that patients with NKX2-1 CN gain had significantly longer survival than the rest of the cohort (P=0.018; Fig. 3C). While patients with CN loss had significantly shorter survival than the rest of the cohort (P=0.041; Fig. 3D). EGFR mutational subgrouping showed that the survival of patients with EGFR mutant and wildtype tumors had no statistical difference, regardless of NKX2-1 CN gain or loss (Fig. S3D and E). This is potentially due to the small sample size of participants with EGFR driver mutations (e.g., T90M, L858R and 19del). Furthermore, NKX2-1 CN gain significantly prognosed shorter progression-free interval in patients with EGFR driver mutations than those with the wildtype sequence (P=0.037), while subgrouping by CN loss did not show a significant difference in the survival of patients with EGFR mutant and wildtype tumors (Fig. S3F and G). Lastly, quartile subgrouping by NKX2-1 expression failed to provide significant prognostic value (P=0.088; Fig. 3E). Collectively, these results suggested that NKX2-1 CNAs differentially influence patient survival, and CN gain may stratify a prognostically favorable subgroup, while CN loss may prognose worse survival in patients with NSCLC. Additionally, the prognostic value of NKX2-1 CNAs may be more superior than expression. Further studies are needed to verify the prognostic value of NKX2-1 CN gain and loss in patients with EGFR driver mutations and wildtype sequence.

Mutational burden in chromosome Y may differentiate the mutational architecture of tumors with NKX2-1 CNAs

Next, the present study aimed to determine the unique molecular features of tumors from the two prognostic groups (CN gain and CN loss) to identify theragnostic markers and to understand the physiologies associated with different survival outcomes. The two groups were characterized with high EGFR mutational frequencies (Fig. 2H) and high mutational burden in chromosome 7 (Fig. 4A), suggesting culprits in their tumor mutational architectures (TMAs). Genome-wide mutational analysis revealed that among the reported 20,506 mutated genes in NSCLC from TCGA cohort, there were 20,134 and 20,157 mutations associated with NKX2-1 CN loss and gain, respectively. A total of ~19,941 (97.24%) synonymous mutations were found. There were 216 (1.05%) and 193 (0.94%) uniquely mutated genes associated with CN gain and loss, respectively (Fig. 4B). Gene set analysis revealed that most affected genes were found in adenocarcinoma, rather than in squamous carcinomas (Fig. S4A).

Enrichment analysis showed that uniquely mutated genes (Figs. 4C and S4B) associated with NKX2-1 CNAs were found significantly distributed in chromosomes X, 15 and 14 (P<0.05) for both CN gain and loss. Additionally, Y-specific mutational signatures were found in patients with CN loss (P<0.05; Fig. 4D). BioPlanet enrichment analysis revealed that the mutated genes were found to play a significant role in a number of immune processes. Genes involved in PD-1 signaling were preferentially affected in CN gain; whereas, genes of the innate immune system were found enriched for CN loss. No significant GO biological process enrichment was found for CN gain, but mutated genes in CN loss were found involved in immunoregulation (Fig. 4E). These results suggested that NKX2-1 CNAs are closely associated with mutational clusters in chromosomes X, 14 and 15. Y-specific mutations may differentiate the two prognostic groups. TMA in tumors with NKX2-1 CNAs are potentially linked to dysregulation of antitumor activity of the immune system.

NKX2-1 CNAs are associated with high frequency of chromosomal instability

Y-specific mutations were significantly higher in tumors with NKX2-1 CN loss (P=0.014) but the number of mutational frequencies in chromosomes X, 15 and 14 had no significant differences between the two prognostic groups (P>0.05; Fig. 5A). Additionally, a number of biological processes were found to be dysregulated, which are independent of genes located from these chromosomes (Fig. S4C), suggesting that other critical tumorigenic drivers exist. Thus, the present study next examined the degree of clonal chromosomal instability of tumors with NKX2-1 CNAs from the Bivona cohort. A total of 1,408 affected genes in clones with CNAs were identified, and 61% of these genes significantly differentiated the clonal features of the two prognostic groups (Fig. 5B). Genes with altered CN were found to be higher in clones with NKX2-1 CN loss (33%) than in gain (28%). Genes with altered CN were found significantly enriched in chromosomes 1, 19, 3, 17, 2 and 4 (Fig. 5C and D). This suggested that chromosomal instability may also influence the disease outcomes of patients with NXK2-1 CNAs.

Focal alteration in EGFR CN was positively correlated with NKX2-1 CN gain (Fig. 5E), which may justify the sensitization of cancer cells with therapeutic interventions leading to favorable outcomes (Fig. 3B). Consistently, patients with NKX2-1 CN gain had a higher frequency of developing secondary mutations compared to those with CN loss (Fig. 5F), which may suggest that NKX2-1 CN gain could serve as biomarker to stratify patients with higher chromosomal instability that could predict better survival outcomes using tyrosine kinase inhibitors (TKIs; Fig. 5G). Genes with significantly altered CNs were also found enriched for immune processes (Figs. 5H, S4D and E).

NKX2-1 CNAs can influence tumor infiltration by lymphocytes

To gain deeper insights into how NKX2-1 CNAs could influence the immune microenvironment, the expression of the CD2 marker in tumors from the Bivona and TCGA cohorts was measured. CD2 expression was statistically comparable between tumors with NKX2-1 CN gain and loss (P=0.866; Fig. S5A). However, CD2 expression was significantly correlated with NKX2-1 CN loss (P<0.001; Fig. S5B), which indicated a dynamic lymphocyte infiltration in tumors with NKX2-1 CNAs.

The expression of 53 gene markers of TILs in NSCLC with NKX2-1 CNAs was next compared. Unsupervised clustering analysis showed that the expression of TIL markers was heterogenous in CN gain, whereas, some markers strongly clustered with CN loss (Fig. 6A). There were 37 markers that were significantly correlated with NKX2-1 CN loss, while four were correlated with CN gain (Figs. 6B and S5C). Excluding DPP4, which was common for both, the higher expression of three (NGF, SCN3A and GPNMB) unique TIL signatures in CN gain was prognostic of longer disease-free survival than those with lower expression (P=0.005; Fig. 6C). By contrast, the higher expression of 36 TIL marker genes (associated with NKX2-1 CN loss) had no significant effect on the survival of patients with lower expression (P=0.89). These results suggested that NKX2-1 CNAs could be linked to differential TIL profiles of NSCLC tumors, and that the immune infiltration was prognostic of survival (52).

The multivariate correlations of the 53 genes varied significantly, which may link to differences in lymphocyte proportions (Fig. S5D). There were significant differences in TIL estimates between tumors with focal gain and loss of NKX2-1 CN (Fig. 6D). T-cell count was generally higher in tumors with CN gain (P=0.003) and was dominated by the CD4+ subtype (P<0.001). Meanwhile, CD8+ T cells were found higher in CN loss (P<0.001). B cell (naïve, memory and plasma) infiltrations were significantly higher in CN gain (P<0.001). Monocyte, neutrophil, activated mast cell and resting natural killer cell levels were significantly higher in CN loss (P<0.001, P=0.002, P=0.007 and P=0.008, respectively). M0 macrophage levels were higher in CN gain (P<0.001), whereas the tumor-promoting M2 macrophage levels were found higher in CN loss (P=0.011; Fig. 6E). The proportion of regulatory T cells did not significantly differ between the two groups (P=0.948; Fig. S6A). Patients with NKX2-1 CN loss previously associated with a higher degree of chromosomal instability (Fig. 4D) had a significantly higher CD8+ infiltration (P=0.011; Fig. S6B) and higher cytotoxicity score (P<0.001; Fig. S6C) than those with CN gain. This suggested a potential ongoing battle between immune evolution and clonal tumor heterogeneity, and that the loss of NKX2-1 CN may be indicative of tumor maintenance (53). Whereas NKX2-1 CN gain may represent co-perturbation events that constrain the evolution of NSCLC subclones, justifying their favorable outcomes (Fig. 3C). These results suggested that NKX2-1 CNAs may be associated with immunologically dynamic tumors.

NKX2-1 CNAs as biomarkers for NSCLC-targeted therapy

The extent to which co-alteration in NKX2-1 and EGFR CNVs (Fig. 2I) is significant for EGFR-targeted therapy was unclear. Therefore, the survival of patients with NKX2-1 CNAs who did and did not receive targeted therapy was compared. The survival of patients with CN gain (P=0.3006) and loss (P=0.7019) was found to be statistically comparable. However, patients with NKX2-1 CN gain had a relatively longer survival than those with CN loss who had received targeted therapy (Fig. S3H and I). Analysis of driver genes in those two patient groups showed that there was a higher frequency of EGFR-sensitizing mutation (exon19 del and exon21L858R) in CN gain than in loss (Fig. S3J). These results signified that NKX2-1 CN gain may be associated with higher occurrence of EGFR-targetable mutations and could be linked to EGFR-targeted stratification, which was not observed previously because past reports failed to compare CN amplification with deletion (14,15).

Notably, NKX2-1 CN loss had a negative and significant correlation with CD274 expression (Fig. 2D) and associated with infiltration of M2 macrophages (Fig. 6E). Thus, the expression-based association of immune checkpoint proteins with NKX2-1 CNAs was explored. It was found that the positive regulation of CD80 and CD40LG, and the negative regulation of TNFSF9, VTCN1 and LGALS9 were significantly correlated with NKX2-1 CNVs (Fig. S7). The expression of CD274, VTCN1 and LGALS9 were significantly higher in CN loss than in gain. The expression of CD80 and CD40LG had no significant difference between the two groups (Fig. 6F). The higher expression of immune checkpoint proteins provided additional evidence that NSCLC with NKX2-1 CN loss may be associated with a ‘cold’ immune microenvironment, which may justify their poor prognostic outcomes.

Further correlation analysis showed that CD274 may not be the dominant immune checkpoint in NSCLC tumors with NKX2-1 CN loss. The immunoexpression of VTCN1 and LGALS9 were more significant (P<0.001 and P=0.002, respectively) than CD274 (P=0.090). Co-expression analysis revealed that VTCN1 was significantly correlated with the negative regulation of CD274 (P<0.001), whereas LGALS9 was significantly correlated with the positive regulation of CD274 (P<0.001). No significant correlation was observed between the expression of LGALS9 and VTCN1 (P=0.288; Fig. 6G). These results suggested that NKX2-1 CNAs could be associated with differential immune checkpoint profiles in NSCLC tumors.

Discussion

Alterations in NKX2-1 CN are one of the most frequently observed consequences of genomic instability in lung cancer (16). However, beyond amplification, little is known about the clinical significance of NKX2-1 CNVs, the disease burden associated with focal CNAs and their implications for targeted therapy. In the present study, it was reported that NKX2-1 CNAs had a stronger correlation with the combined EGFR and PD-L1 status of tumors than RNA and protein expression. Furthermore, it was shown that focal loss and gain in NKX2-1 CN were prognostic in NSCLC. CN gain may represent a prognostically favorable subgroup, whereas CN loss may be associated with poor survival. With the enigma surrounding TTF-1 expression studies, including inconsistencies in patient stratification and immunostaining specificities (13), the prognostic implication of CNAs could be superior to expression. In the present study, the prognostic threshold associated with NKX2-1 CN gain was defined as >0.1875 and loss was defined as <-0.0767 [log2(tumor/normal)].

The present analysis revealed that NKX2-1 and EGFR CNs were significantly co-altered. Consistently, tumors with NKX2-1 CNAs had a high frequency of missense mutations in chromosome 7 where the EGFR gene is located, suggesting that NKX2-1 CNAs may serve as a biomarker for EGFR-directed therapy. Additionally, patients with NKX2-1 CN gain and loss had 97.25% synonymous mutations, with 216 and 193 distinct mutational signatures, respectively. The mutational burden in the Y-chromosome was found to be significantly higher in tumors with NKX2-1 CN loss, indicating that the unique Y-specific mutational cluster associated with CN loss tends to negatively impact survival. The findings of the present study justify the foundational basis for exploring the Y-specific contribution of tumor mutational signatures in NSCLC and how sex could influence treatment outcomes in lung cancer (54).

The results of the present study also revealed the difference in the degree of genome-wide CNAs in cancer clones with focal gain and loss in NKX2-1 CN. Out of 1,408 genes with CNAs, more than half (61%) were significantly co-altered with NKX2-1. Most genome-wide alterations were found in clones with NKX2-1 CN loss than in gain (33 vs. 28%, respectively). The majority of the affected genes were enriched in chromosomes 1, 19, 3, 17, 2 and 4, which suggested that NKX2-1 CNAs are closely linked to a high frequency of chromosomal instability.

A recent study showed that chromosomal instability could predict treatment outcomes of patients with NSCLC receiving TKIs (55). The researchers showed that chromosomal instability resulting in Chr 1p13.3-p13.1 gain could predict poor response to EGFR-TKI. However, CN gain in Chr 14q31.1-q31.3 and Chr 7q31.1-q31.31, and CN loss in Chr 8p23.3-p23.1 and Chr 10q21.2-q22.1 were found to have favorable survival outcomes in patients receiving TKIs. This suggests that contrary to the general assumption that high chromosomal instability could negatively impact survival, the chromosomal location and the affected genes have a more significant impact on patient outcomes. The results of the present study showed that NKX2-1 CN gain may be associated with chromosomal instability that resulted in the development of more secondary mutations, which could confer a better response to TKI treatments. Additionally, the present study showed that the unique genomic CN perturbations and instabilities in chromosomes 1, 19, 3, 17, 2 and 4 among patients with NSCLC with NKX2-1 CN gain could predict better treatment outcomes with TKIs.

Genes with perturbed CN and uniquely mutated in tumors with NKX2-1 CNAs were found enriched for a number of immunological processes, suggesting a close association between NKX2-1 CNAs and immune microenvironment remodeling. The results of the present study showed that the TIL profiles of patients with NKX2-1 CN gain and loss were different. TIL estimation analysis showed that total T-cell and B-cell proportions were higher in tumors with CN gain than loss. This suggested that tumors with NKX2-1 CN gain had higher adaptive infiltration and robust humoral responses that resulted in better survival. Tumors with CN loss were found to have a ‘colder’ immune environment due to significantly higher M2 macrophage infiltrates and immune checkpoint protein expression.

Currently, there is a limited number of effective biomarkers that may accurately prognose whether patients will benefit, develop resistance or experience severe side effects from therapy, particularly using TKIs and immune checkpoint inhibitors (ICIs). Subgrouping by PD-L1 positivity, EGFR mutational status and NKX2-1 expression produced insufficient results (5658). The findings of the present study revealed that patients with NKX2-1 CN gain may benefit from either EGFR-TKIs or ICIs due to a higher frequency of EGFR-driver mutations and higher adaptive infiltrates.

Consequently, enrichment of EGFR-targetable mutations in CN gain could also present a negative impact due to a higher tendency of developing resistant mutations (e.g., T790M) and co-mutations. Indeed, NKX2-1 CN amplification prognosed a poor response to EGFR-TKIs in patients with recurrent tumors after surgical treatment (59). However, this study stratified patients as NKX2-1 CN amplified or unamplified only, and patients designated as unamplified could have harbored CN deletions, which could radically change the survival outcomes of the cohort. Also, the study failed to correlate CN amplification with other genomic alterations that may confound with the prognostic value of NKX2-1 CN amplification. Lastly, the detection of NKX2-1 CNAs remains unstandardized at present, and studies utilizing fluorescence in situ hybridization and PCR are normally hampered by variations and complexities of DNA breakpoints in analyzing CNVs (14,60). Thus, the dynamic and complete changes in CN are often not captured from studies utilizing these techniques.

A previous paper that utilized next generation sequencing to analyze EGFR-TKI outcomes in patients with lung cancer with concurrent genomic alterations identified a lower frequency of NKX2-1 CN amplification in tumors that developed resistance to EGFR-TKI than those that were naïve (11 vs. 15%), and reported that acquired T790M mutation, co-mutation in TP53 and co-amplification of EGFR had higher frequencies in EGFR-TKI resistant tumors than NKX2-1 CN amplification alone (61). In fact, there was only a small frequency of T790M mutations in TCGA cohort with NKX2-1 CN gain. Thus, there is a need for a more quantitative approach in determining the extent to which NKX2-1 CN gain could be predictive for patients receiving EGFR-TKIs. With the advent of more sophisticated tools, such as sequencing platforms, the development of quantitative CNV subgrouping is now possible. To the best of our knowledge, the present study is the first to provide retrospective data and propose a quantitative approach to stratifying patients as having NKX2-1 CN gain with measurable CNVs [>0.1875 log2(tumor/normal)] that may benefit from EGFR-TKIs. However, this hypothesis requires further clinical analysis.

Additionally, previous studies failed to account NKX2-1 CN deletion, and thus a comprehensive and quantitative comparison of the prognostic value of NKX2-1 CNAs is still elusive. The present analysis also showed that NKX2-1 CN loss with measurable CNVs [<-0.0767 log2(tumor/normal)] was associated with lower TMB, higher genome-wide CNV perturbations, reduced adaptive infiltrations and increased expression of immunosuppressive molecules. All these qualifications have been linked to poor responses that may be unlikely to benefit from combinatorial TKI–ICI therapy. However, these hypotheses require further clinical validations. Lastly, the present findings emphasized the need to develop other target approaches, other than CD274, LGALS9 and VTCN1, that could be potential targets in NSCLC.

Limitations of the present study include the small sample size, particularly in the Filipino cohort, which may impact correlation analysis. Additionally, subgrouping by EGFR mutation did not reach significant survival probabilities due to the small sample size and requires further study. Second, the involvement of ROR1 in the combined EGFR and PD-L1 status needs to be further explored through knock-in and knock-out mouse models, supported by protein-protein interaction experiments and functional mice studies. Third, the CNV data from TCGA and the Bivona cohorts were detected using Affymetrix SNP 6.0 arrays and RNA sequencing, respectively. Although CNV was normalized to tumor-normal ratio, difference in methods may have contributed to variations in measurements. Fourth, the expression data in TCGA and the Bivona cohorts were scaled differently, which may have affected statistical analyses, although data from each cohort were interpreted independently. Lastly, the analysis of TILs depended on 53 select gene markers, which may have influenced the estimation analysis.

Supplementary Material

Supporting Data
Supporting Data

Acknowledgements

The authors would like to thank Dr. Neil Andrew Bascos (National Institute of Molecular Biology and Biotechnology, University of the Philippines, Quezon City, Philippines) for his insights and contributions in the study.

Funding

This study was funded by the Philippine Council for Health Research and Development and the Grants-In-Aid program of The Department of Science and Technology (grant no. IC-21-02421).

Availability of data and materials

The datasets used and/or analyzed during the current study available from the corresponding author on reasonable request.

Authors' contributions

HGCL, MSI, NJ, KVH, TMS, GCL, SSN and MIDLS conceived and designed the study; HGCL, DM and JS provided administrative support; HGCL, MSI, NJ, KVH, TMS, GCL, SSN and MIDLS provided study materials or patients; SMAG, MB, KJD, BBB, MIDLS, VML, JS and DM performed collection and assembly of the data; HGCL, SMAG, MB, KJD, BBB, MIDLS and VML performed data analysis and interpretation; and HGCL, MSI, NJ, KVH, TMS, GCL, SSN, SMAG, MB, KJD, BBB, MIDLS, VML, DM and JS wrote the manuscript. MIDLS and HGCL confirm the authenticity of all the raw data. All authors read and approved the final manuscript.

Ethics approval and consent to participate

The study was conducted in accordance with The Declaration of Helsinki (as revised in 2013). The study was approved by The Institutional Ethics Review Board (approval no. LCP-CS-001-2019) of the Lung Center of the Philippines (Quezon City, Philippines). The protocol for specimen collection was approved by The Single Joint Research Ethics Board (approval no. SJREB-2020-97) of the Department of Health, Manila, Philippines. All patients provided written informed consent for genetic testing, as well as the use of their clinical data.

Patient consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

References

1 

Feuk L, Carson AR and Scherer SW: Structural variation in the human genome. Nat Rev Genet. 7:85–97. 2006. View Article : Google Scholar : PubMed/NCBI

2 

Steele CD, Abbasi A, Islam SMA, Bowes AL, Khandekar A, Haase K, Hames-Fathi S, Ajayi D, Verfaillie A, Dhami P, et al: Signatures of copy number alterations in human cancer. Nature. 606:984–991. 2022. View Article : Google Scholar : PubMed/NCBI

3 

Bjaanæs MM, Nilsen G, Halvorsen AR, Russnes HG, Solberg S, Jørgensen L, Brustugun OT, Lingjærde OC and Helland Å: Whole genome copy number analyses reveal a highly aberrant genome in TP53 mutant lung adenocarcinoma tumors. BMC Cancer. 21:10892021. View Article : Google Scholar : PubMed/NCBI

4 

Shao X, Lv N, Liao J, Long J, Xue R, Ai N, Xu D and Fan X: Copy number variation is highly correlated with differential gene expression: A pan-cancer study. BMC Med Genet. 20:1752019. View Article : Google Scholar : PubMed/NCBI

5 

Heo Y, Heo J, Han SS, Kim WJ, Cheong HS and Hong Y: Difference of copy number variation in blood of patients with lung cancer. Int J Biol Markers. 36:3–9. 2020. View Article : Google Scholar : PubMed/NCBI

6 

Qiu ZW, Bi JH, Gazdar AF and Song K: Genome-wide copy number variation pattern analysis and a classification signature for non-small cell lung cancer. Genes Chromosomes Cancer. 56:559–569. 2017. View Article : Google Scholar : PubMed/NCBI

7 

Chen S, Lu L, Xian J, Shi C, Chen J, Rao B, Qiu F, Lu J and Yang L: Prognostic value of germline copy number variants and environmental exposures in Non-small cell lung cancer. Front Genet. 12:681852021.

8 

Aujla S, Aloe C, Vannitamby A, Hendry S, Rangamuwa K, Wang H, Vlahos R, Selemidis S, Leong T, Steinfort D and Bozinovski S: Programmed Death-Ligand 1 Copy number loss in NSCLC associates with reduced programmed Death-Ligand 1 tumor staining and a cold immunophenotype. J Thorac Oncol. 17:675–687. 2022. View Article : Google Scholar : PubMed/NCBI

9 

Alden S, Ricciuti B, Spurr L, Gupta H, Lamberti G, Li Y, Sholl L, Cherniack A and Awad A: P33.04 programmed death-ligand 1 (PD-L1) changes in non-small-cell lung cancer (NSCLC): Clinical, pathologic, and genomic correlates. J Thorac Oncol. 16:S406–S407. 2021. View Article : Google Scholar

10 

Inoue Y, Yoshimura K, Mori K, Kurabe N, Kahyo T, Mori H, Kawase A, Tanahashi M, Ogawa H, Inui N, et al: Clinical significance of PD-L1 and PD-L2 copy number gains in non-small-cell lung cancer. Oncotarget. 7:32113–32128. 2016. View Article : Google Scholar : PubMed/NCBI

11 

Wei J, Meng P, Terpstra MM, van Rijk A, Tamminga M, Scherpen F, Ter Elst A, Alimohamed MZ, Johansson LF, Stigt J, et al: Clinical Value of EGFR Copy number gain determined by amplicon-based targeted next generation sequencing in patients with EGFR-Mutated NSCLC. Target Oncol. 16:215–226. 2021. View Article : Google Scholar : PubMed/NCBI

12 

Peng D, Liang P, Zhong C, Xu P, He Y, Luo Y, Wang X, Liu A and Zeng Z: Effect of EGFR amplification on the prognosis of EGFR-mutated advanced non-small-cell lung cancer patients: A prospective observational study. BMC Cancer. 22:13232022. View Article : Google Scholar : PubMed/NCBI

13 

Gloriane C Luna H, Severino Imasa M, Juat N, Hernandez KV, May Sayo T, Cristal-Luna G, Marie Asur-Galang S, Bellengan M, John Duga K, Brian Buenaobra B, et al: Expression landscapes in non-small cell lung cancer shaped by the thyroid transcription factor 1. Lung Cancer. 176:121–131. 2023. View Article : Google Scholar

14 

Yoshimura K, Inoue Y, Mori K, Iwashita Y, Kahyo T, Kawase A, Tanahashi M, Ogawa H, Inui N, Funai K, et al: Distinct prognostic roles and heterogeneity of TTF1 copy number and TTF1 protein expression in non-small cell lung cancer. Genes Chromosomes Cancer. 56:570–581. 2017. View Article : Google Scholar : PubMed/NCBI

15 

Li X, Wan L, Shen H, Geng J, Nie J, Wang G, Jia N, Dai M and Bai X: Thyroid transcription Factor-1 amplification and expressions in lung adenocarcinoma tissues and pleural effusions predict patient survival and prognosis. J Thorac Oncol. 7:76–84. 2012. View Article : Google Scholar : PubMed/NCBI

16 

Clarke N, Biscocho J, Kwei KA, Davidson JM, Sridhar S, Gong X and Pollack JR: Integrative genomics implicates EGFR as a downstream mediator in NKX2-1 amplified non-small cell lung cancer. PLoS One. 10:e01420612015. View Article : Google Scholar : PubMed/NCBI

17 

Luna HG, Prieto E, Dimayacyac-Esleta BR, Imasa MS, Juat N, Hernandez KV, Sayo TM, Cristal-Luna GR, Asur-Galang SM, Bellengan M, et al: 342P Prognostic implications of PD-L1 co-expression among Filipino EGFR MT mNSCLC. Ann Oncol. 33 (Suppl):S15762022. View Article : Google Scholar

18 

Luna HGC, Imasa MS, Juat N, Hernandez KV, Sayo TM, Cristal-Luna G, Asur-Galang SM, Bellengan M and Duga KJ: The differential prognostic implications of PD-L1 expression in the outcomes of Filipinos with EGFR-mutant NSCLC treated with tyrosine kinase inhibitors. Transl Lung Cancer Res. 12:1896–1911. 2023. View Article : Google Scholar : PubMed/NCBI

19 

Cancer Genome Atlas Research Network, . Weinstein JN, Collisson EA, Mills GB, Shaw KR, Ozenberger BA, Ellrott K, Shmulevich I, Sander C and Stuart JM: The cancer genome atlas pan-cancer analysis project. Nat Genet. 45:1113–1120. 2013. View Article : Google Scholar : PubMed/NCBI

20 

Maynard A, McCoach CE, Rotow JK, Harris L, Haderk F, Kerr DL, Yu EA, Schenk EL, Tan W, Zee A, et al: Therapy-Induced evolution of human lung cancer revealed by single-cell RNA sequencing. Cell. 182:1232–1251.e22. 2020. View Article : Google Scholar : PubMed/NCBI

21 

Goldman MJ, Craft B, Hastie M, Repečka K, McDade F, Kamath A, Banerjee A, Luo Y, Rogers D, Brooks AN, et al: Visualizing and interpreting cancer genomics data via the Xena platform. Nat Biotechnol. 38:675–678. 2020. View Article : Google Scholar : PubMed/NCBI

22 

Grossman RL, Heath AP, Ferretti V, Varmus HE, Lowy DR, Kibbe WA and Staudt LM: Toward a shared vision for cancer genomic data. N Engl J Med. 375:1109–1112. 2016. View Article : Google Scholar : PubMed/NCBI

23 

Luna HGC, Imasa MS, Juat N, Hernandez V, Sayo MT, Cristal-Luna G, Asur-Galang SM, Bellengan M, Duga KJ, Buenaobra BB, et al: Prognostic Value of PD-L1 in Metastatic NSCLC with EGFR-Sensitizing Mutations: A Benchmark Filipino Cohort Study. SSRN. 2022.doi: 10.2139/ssrn.4286198. View Article : Google Scholar

24 

Torous VF, Rangachari D, Gallant BP, Shea M, Costa DB and VanderLaan PA: PD-L1 testing using the clone 22C3 pharmDx kit for selection of patients with non-small cell lung cancer to receive immune checkpoint inhibitor therapy: Are cytology cell blocks a viable option? J Am Soc Cytopathol. 7:133–141. 2018. View Article : Google Scholar : PubMed/NCBI

25 

Gu K, Shah V, Ma C, Zhang L and Yang M: Cytoplasmic immunoreactivity of thyroid transcription Factor-1 (Clone 8G7G3/1) in hepatocytestrue positivity or cross-reaction? Am J Clin Pathol. 128:382–388. 2007. View Article : Google Scholar : PubMed/NCBI

26 

Tang Z, Li C, Kang B, Gao G, Li C and Zhang Z: GEPIA: A web server for cancer and normal gene expression profiling and interactive analyses. Nucleic Acids Res. 45:W98–W102. 2017. View Article : Google Scholar : PubMed/NCBI

27 

Bardou P, Mariette J, Escudié F, Djemiel C and Klopp C: Jvenn: An interactive Venn diagram viewer. BMC Bioinformatics. 15:2932014. View Article : Google Scholar : PubMed/NCBI

28 

Kuleshov MV, Jones MR, Rouillard AD, Fernandez NF, Duan Q, Wang Z, Koplev S, Jenkins SL, Jagodnik KM, Lachmann A, et al: Enrichr: A comprehensive gene set enrichment analysis web server 2016 update. Nucleic Acids Res. 44:W90–W97. 2016. View Article : Google Scholar : PubMed/NCBI

29 

Xie Z, Bailey A, Kuleshov M V, Clarke DJB, Evangelista JE, Jenkins SL, Lachmann A, Wojciechowicz ML, Kropiwnicki E, Jagodnik KM, et al: Gene set knowledge discovery with enrichr. Curr Protoc. 1:e902021. View Article : Google Scholar : PubMed/NCBI

30 

Huang R, Grishagin I, Wang Y, Zhao T, Greene J, Obenauer JC, Ngan D, Nguyen DT, Guha R, Jadhav A, et al: The NCATS BioPlanet-An integrated platform for exploring the universe of cellular signaling pathways for toxicology, systems biology, and chemical genomics. Front Pharmacol. 10:4452019. View Article : Google Scholar : PubMed/NCBI

31 

Kanehisa M, Furumichi M, Sato Y, Ishiguro-Watanabe M and Tanabe M: KEGG: Integrating viruses and cellular organisms. Nucleic Acids Res. 49:D545–D551. 2021. View Article : Google Scholar : PubMed/NCBI

32 

Gene Ontology Consortium: The Gene Ontology resource: Enriching a GOld mine. Nucleic Acids Res. 49:D325–D334. 2021. View Article : Google Scholar : PubMed/NCBI

33 

Lee BT, Barber GP, Benet-Pagès A, Casper J, Clawson H, Diekhans M, Fischer C, Gonzalez JN, Hinrichs AS, Lee CM, et al: The UCSC Genome Browser database: 2022 update. Nucleic Acids Res. 50:D1115–D1122. 2022. View Article : Google Scholar : PubMed/NCBI

34 

Binder JX, Pletscher-Frankild S, Tsafou K, Stolte C, O'Donoghue SI, Schneider R and Jensen LJ: COMPARTMENTS: Unification and visualization of protein subcellular localization evidence. Database (Oxford). 2014:bau0122014. View Article : Google Scholar : PubMed/NCBI

35 

Zhang X, Lan Y, Xu J, Quan F, Zhao E, Deng C, Luo T, Xu L, Liao G, Yan M, et al: CellMarker: A manually curated resource of cell markers in human and mouse. Nucleic Acids Res. 47:D721–D728. 2019. View Article : Google Scholar : PubMed/NCBI

36 

Wu C, MacLeod I and Su AI: BioGPS and MyGene.info: Organizing online, gene-centric information. Nucleic Acids Res. 41:D561–D565. 2013. View Article : Google Scholar : PubMed/NCBI

37 

Tang Z, Kang B, Li C, Chen T and Zhang Z: GEPIA2: An enhanced web server for large-scale expression profiling and interactive analysis. Nucleic Acids Res. 47:W556–W560. 2019. View Article : Google Scholar : PubMed/NCBI

38 

Wu M, Mei F, Liu W and Jiang J: Comprehensive characterization of tumor infiltrating natural killer cells and clinical significance in hepatocellular carcinoma based on gene expression profiles. Biomed Pharmacother. 121:1096372020. View Article : Google Scholar : PubMed/NCBI

39 

Goedhart J and Luijsterburg MS: VolcaNoseR is a web app for creating, exploring, labeling and sharing volcano plots. Sci Reports. 10:205602020.PubMed/NCBI

40 

Babicki S, Arndt D, Marcu A, Liang Y, Grant JR, Maciejewski A and Wishart DS: Heatmapper: Web-enabled heat mapping for all. Nucleic Acids Res. 44:W147–W153. 2016. View Article : Google Scholar : PubMed/NCBI

41 

Newman AM, Liu CL, Green MR, Gentles AJ, Feng W, Xu Y, Hoang CD, Diehn M and Alizadeh AA: Robust enumeration of cell subsets from tissue expression profiles. Nat Methods. 12:453–457. 2015. View Article : Google Scholar : PubMed/NCBI

42 

Becht E, Giraldo NA, Lacroix L, Buttard B, Elarouci N, Petitprez F, Selves J, Laurent-Puig P, Sautès-Fridman C, Fridman WH and de Reyniès A: Estimating the population abundance of tissue-infiltrating immune and stromal cell populations using gene expression. Genome Biol. 17:2182016. View Article : Google Scholar : PubMed/NCBI

43 

Li T, Fan J, Wang B, Traugh N, Chen Q, Liu JS, Li B and Liu XS: TIMER: A web server for comprehensive analysis of tumor-infiltrating immune cells. Cancer Res. 77:e108–e110. 2017. View Article : Google Scholar : PubMed/NCBI

44 

Galland L, Le Page AL, Lecuelle J, Bibeau F, Oulkhouir Y, Derangère V, Truntzer C and Ghiringhelli F: Prognostic value of thyroid transcription Factor-1 expression in lung adenocarcinoma in patients treated with anti PD-1/PD-L1. Oncoimmunology. 10:19576032021. View Article : Google Scholar : PubMed/NCBI

45 

Lamberti G, Spurr LF, Li Y, Ricciuti B, Recondo G, Umeton R, Nishino M, Sholl LM, Meyerson ML, Cherniack AD and Awad MM: Clinicopathological and genomic correlates of programmed cell death ligand 1 (PD-L1) expression in nonsquamous non-small-cell lung cancer. Ann Oncol. 31:807–814. 2020. View Article : Google Scholar : PubMed/NCBI

46 

Yamaguchi T, Yanagisawa K, Sugiyama R, Hosono Y, Shimada Y, Arima C, Kato S, Tomida S, Suzuki M, Osada H and Takahashi T: NKX2-1/TITF1/TTF-1-Induced ROR1 is required to sustain EGFR survival signaling in lung adenocarcinoma. Cancer Cell. 21:348–361. 2012. View Article : Google Scholar : PubMed/NCBI

47 

Li Y, Liu Z, Zhao Y, Yang J, Xiao TS, Conlon RA and Wang Z: PD-L1 expression is regulated by ATP-binding of the ERBB3 pseudokinase domain. Genes Dis. 10:1702–1713. 2023. View Article : Google Scholar : PubMed/NCBI

48 

Hu X, Li J, Fu M, Zhao X and Wang W: The JAK/STAT signaling pathway: From bench to clinic. Signal Transduct Target Ther. 6:4022021. View Article : Google Scholar : PubMed/NCBI

49 

Jahangiri A, Dadmanesh M and Ghorban K: STAT3 inhibition reduced PD-L1 expression and enhanced antitumor immune responses. J Cell Physiol. 235:9457–9463. 2020. View Article : Google Scholar : PubMed/NCBI

50 

Zegeye MM, Lindkvist M, Fälker K, Kumawat AK, Paramel G, Grenegård M, Sirsjö A and Ljungberg LU: Activation of the JAK/STAT3 and PI3K/AKT pathways are crucial for IL-6 trans-signaling-mediated pro-inflammatory response in human vascular endothelial cells. Cell Commun Signal. 16:552018. View Article : Google Scholar : PubMed/NCBI

51 

Zhang N, Zeng Y, Du W, Zhu J, Shen D, Liu Z and Huang JA: The EGFR pathway is involved in the regulation of PDL1 expression via the IL-6/JAK/STAT3 signaling pathway in EGFR-mutated non-small cell lung cancer. Int J Oncol. 49:1360–1368. 2016. View Article : Google Scholar : PubMed/NCBI

52 

Federico L, McGrail DJ, Bentebibel SE, Haymaker C, Ravelli A, Forget MA, Karpinets T, Jiang P, Reuben A, Negrao MV, et al: Distinct tumor-infiltrating lymphocyte landscapes are associated with clinical outcomes in localized non-small-cell lung cancer. Ann Oncol. 33:42–56. 2022. View Article : Google Scholar : PubMed/NCBI

53 

Jamal-Hanjani M, Wilson GA, McGranahan N, Birkbak NJ, Watkins TBK, Veeriah S, Shafi S, Johnson DH, Mitter R, Rosenthal R, et al: Tracking the evolution of non-small-cell lung cancer. N Engl J Med. 376:2109–2121. 2017. View Article : Google Scholar : PubMed/NCBI

54 

Pinto JA, Vallejos CS, Raez LE, Mas LA, Ruiz R, Torres-Roman JS, Morante Z, Araujo JM, Gómez HL, Aguilar A, et al: Gender and outcomes in non-small cell lung cancer: An old prognostic variable comes back for targeted therapy and immunotherapy? ESMO Open. 3:e0003442018. View Article : Google Scholar : PubMed/NCBI

55 

He H, Ma H, Chen Z, Chen J, Wu D, Lv X and Zhu J: Chromosomal copy number variation predicts EGFR-TKI response and prognosis for patients with non-small cell lung cancer. Pharmgenomics Pers Med. 16:835–846. 2023.PubMed/NCBI

56 

Hong L, Dibaj S, Negrao MV, Reuben A, Roarty E, Rinsurongkawong W, Lewis J, Gibbons DL, Sepesi B, Papadimitrakopoulou V, et al: Spatial and temporal heterogeneity of PD-L1 and its impact on benefit from immune checkpoint blockade in non-small cell lung cancer (NSCLC). J Clin Oncol. 37:90172019. View Article : Google Scholar

57 

De-Rui Huang D and Chih-Hsin Yang J: Checkpoint inhibitor combined with tyrosine kinase inhibitor-the end or beginning? J Thorac Oncol. 15:305–307. 2020. View Article : Google Scholar : PubMed/NCBI

58 

Nakahama K, Kaneda H, Osawa M, Izumi M, Yoshimoto N, Sugimoto A, Nagamine H, Ogawa K, Matsumoto Y, Sawa K, et al: Association of thyroid transcription factor-1 with the efficacy of immune-checkpoint inhibitors in patients with advanced lung adenocarcinoma. Thorac Cancer. 13:2309–2317. 2022. View Article : Google Scholar : PubMed/NCBI

59 

Lee JS, Kim HR, Lee CY, Shin M and Shim HS: EGFR and TTF-1 gene amplification in surgically resected lung adenocarcinomas: Clinicopathologic significance and effect on response to EGFR-tyrosine kinase inhibitors in recurred cases. Ann Surg Oncol. 20:3015–3022. 2013. View Article : Google Scholar : PubMed/NCBI

60 

Pös O, Radvanszky J, Styk J, Pös Z, Buglyó G, Kajsik M, Budis J and Nagy B and Nagy B: Copy number variation: Methods and clinical applications. Appl Sci. 11:8192021. View Article : Google Scholar

61 

Yu HA, Suzawa K, Jordan E, Zehir A, Ni A, Kim R, Kris MG, Hellmann MD, Li BT, Somwar R, et al: Concurrent alterations in EGFR-mutant lung cancers associated with resistance to EGFR kinase inhibitors and characterization of MTOR as a mediator of resistance. Clin Cancer Res. 24:3108–3118. 2018. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

July-2024
Volume 28 Issue 1

Print ISSN: 1792-1074
Online ISSN:1792-1082

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Luna HC, Imasa MS, Juat N, Hernandez KV, Sayo TM, Cristal‑Luna G, Asur‑Galang SM, Bellengan M, Duga KJ, Buenaobra BB, Buenaobra BB, et al: NKX2‑1 copy number alterations are associated with oncogenic, immunological and prognostic remodeling in non‑small cell lung cancer. Oncol Lett 28: 303, 2024
APA
Luna, H.C., Imasa, M.S., Juat, N., Hernandez, K.V., Sayo, T.M., Cristal‑Luna, G. ... Sy‑Naval, S. (2024). NKX2‑1 copy number alterations are associated with oncogenic, immunological and prognostic remodeling in non‑small cell lung cancer. Oncology Letters, 28, 303. https://doi.org/10.3892/ol.2024.14436
MLA
Luna, H. C., Imasa, M. S., Juat, N., Hernandez, K. V., Sayo, T. M., Cristal‑Luna, G., Asur‑Galang, S. M., Bellengan, M., Duga, K. J., Buenaobra, B. B., De Los Santos, M. I., Medina, D., Samo, J., Literal, V. M., Sy‑Naval, S."NKX2‑1 copy number alterations are associated with oncogenic, immunological and prognostic remodeling in non‑small cell lung cancer". Oncology Letters 28.1 (2024): 303.
Chicago
Luna, H. C., Imasa, M. S., Juat, N., Hernandez, K. V., Sayo, T. M., Cristal‑Luna, G., Asur‑Galang, S. M., Bellengan, M., Duga, K. J., Buenaobra, B. B., De Los Santos, M. I., Medina, D., Samo, J., Literal, V. M., Sy‑Naval, S."NKX2‑1 copy number alterations are associated with oncogenic, immunological and prognostic remodeling in non‑small cell lung cancer". Oncology Letters 28, no. 1 (2024): 303. https://doi.org/10.3892/ol.2024.14436