Open Access

Prognostic value of immune related genes in lung adenocarcinoma

  • Authors:
    • Han Wang
    • Meng-Sen Wang
    • Ying Wang
    • Yue-Qing Huang
    • Jian-Ping Shi
    • Zhi-Liang Ding
    • Wen-Jie Wang
  • View Affiliations

  • Published online on: September 18, 2020     https://doi.org/10.3892/ol.2020.12122
  • Article Number: 259
  • Copyright: © Wang et al. This is an open access article distributed under the terms of Creative Commons Attribution License.

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


Abstract

Lung cancer has the highest incidence and mortality rates of all cancers in China. Immune‑related genes and immune infiltrating lymphocytes are involved in tumor growth, and in the past decade, immunotherapy has become increasingly important in the treatment of lung cancer. Using the edgeR package, differentially expressed genes and immune‑related genes (DEIRGs) were identified in patients with lung adenocarcinoma (LUAD). Functional enrichment analysis of DEIRGs was performed using Gene Ontology annotation and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway analyses. Survival‑associated immune‑related genes (IRGs) were selected using univariate Cox regression analysis and the prognostic model was assessed using multivariate Cox regression analysis. Overall, 273 DEIRGs were identified in LUAD, and KEGG pathway analysis of IRGs showed that ‘cytokine‑cytokine receptor interaction’ was the most significantly enriched pathway. Furthermore, six survival associated IRGs were screened to establish a prognostic model; patients in the high risk score group had less favorable survival times, and the prognostic model was negatively associated with B cell infiltration. The present study established a prognostic model using analysis of survival‑related immune‑related genes, which were associated with B cell infiltration.

Introduction

In terms of incidence and mortality rate, lung cancer ranks first among all types of cancer globally, with <20% of patients surviving <5 years after diagnosis, in 2017 (1). There are two forms of lung cancer: Non-small cell lung cancer (NSCLC) and small cell lung cancer (2). NSCLC is further subdivided into lung adenocarcinoma (LUAD), squamous cell carcinoma and large cell carcinoma (3). Adenocarcinoma accounts for the largest proportion of cases, and its incidence has been increasing over the last 10 years, worldwide (4).

The primary treatments available for patients with lung cancer include surgery, chemo- and radiotherapy, molecular targeted therapy and immunotherapy. In the past few decades, researchers have improved our understanding of the role of the immune system in cancer development, and thus, immunotherapy has improved the field of tumor treatment. Of late, checkpoint inhibitors have been developed for the treatment of lung cancer (5,6); blockade of immune checkpoint proteins, including programmed death-1 (PD-1)/programmed death-ligand1 (PD-L1) and cytotoxic T-lymphocyte-associated protein 4, has shown promise in the treatment of several types of cancer, reducing tumor burden and prolonging the survival time of patients (7).

By comprehensively exploring the prognostic value of immune-related genes (IRGS), a recent study assessed individualized immune characteristics to improve the prognoses of patients with NSCLC (8). Previous studies have reported that tumor-infiltrating B cells are closely associated with a more favorable prognosis in NSCLC, cervical cancer and breast cancer (911). Nielsen et al (11) reported that CD20+ tumor-infiltrating lymphocytes (TILs) colocalized with activated CD8+ TILs expressed markers of antigen presentation. The group proposed that the association between CD20+ TILs and patient survival may reflect a supportive role in cytolytic immune responses.

By investigating survival associated immune-related genes, the present study aimed to elucidate the underlying molecular mechanisms of immune genes in LUAD, with a view to establish therapeutic targets and provide a basis for personalized treatment.

Materials and methods

Patients

In total, 10 pairs of LUAD and adjacent normal tissues were obtained from patients with LUAD (4 men and 6 women; median age, 55 years; age range, 33–69 years), undergoing surgery at the Jining Cancer Hospital (Jining, China) from November 2018 to March 2019. None of the patients had received chemo- or radiotherapy prior to surgery. The present study was approved by the Medical Ethics Committees of Jining Cancer Hospital, and written informed consent was provided by all patients prior to surgery. A total of 497 patients were assessed from The Cancer Genome Atlas (TCGA) database (cancer.gov/tcga), including 229 men and 268 women; median age, 66 years; age range, 33–88 years.

Data acquisition and processing

The LUAD dataset (12) containing transcriptome RNA-sequencing and clinical data of patients with LUAD was downloaded from TCGA database. A total of 497 LUAD tissues and 54 normal lung tissues were included in the present study. The list of IRGs was downloaded from the Immunology Database and Analysis Portal (ImmPort) database (13). The inclusion criteria were as follows: Patients with histologically or cytologically confirmed lung adenocarcinoma and patients with complete clinical information. The exclusion criteria were as follows: Patients with histologically or cytologically confirmed cancer other than lung adenocarcinoma and patients with OS time <10 days.

Identification of differentially expressed genes (DEGs), differentially expressed immune-related genes (DEIRGs) and survival-associated immune related genes (IRGs)

DEGs were identified using the edgeR package (version 3.53) in Rand further analyzed. A |log2 fold change| >2.0 and false discovery rate adjusted to P<0.01 were set as the thresholds (14). In addition, volcano and heat maps of the DEGs were constructed using the gplots and heat map components of the edgeR package, respectively. DEIRGs were obtained by comparison with the immune gene lists. Survival-associated IRGs were selected using univariate Cox regression analysis, which was performed using the survival package in R.

Functional enrichment analysis

To understand the underlying biological mechanisms of the IRGs, Gene Ontology (GO) annotation and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway analyses were performed using The Database for Annotation, Visualization and Integrated Discovery (david.ncifcrf.gov/) online tool (15) and cluster profiler, an R package for functional classification and enrichment of gene clusters using the hypergeometric distribution (16,17). The results of the GO and KEGG analyses were displayed using the GOplot package in R, and analyses were based on a threshold of P<0.01.

Development of the immune-related gene prognostic model (IRGPM)

Overall survival time was measured from the date of diagnosis to mortality or the last clinical evaluation. Survival-associated IRGs were selected via univariate Cox regression analysis using the R survival package. Using multivariate Cox regression analysis via the Akaike Information Criterion (18), patients with LUAD were then divided into high-risk and low-risk groups according to the median risks core value. The risk score was calculated using the following formula:

Survival Risk Score(SRS)=∑i=1k(Ci×Vi)

Where k represents the number of mRNAs, Ci represents the coefficient of mRNA in multivariate Cox regression analysis and Vi represents the mRNA expression level. Kaplan-Meier plots were used to divide patients into high and low risk score groups, according to OS time.

Relationship between IRGPM and immune cell infiltration

The Tumor Immune Estimation Resource (TIMER) online database analyzes and creates a visualization of tumor infiltrating immune cells (19). TIMER reanalyzes gene expression data, which includes 10,897 samples across 32 cancer types from TCGA, to estimate the abundance of six subtypes of tumor-infiltrating immune cells, including CD4 T cells, CD8 T cells, B cells, macrophages, dendritic cells (DCs) and neutrophils. Thus, TIMER can easily be used to determine the relationship between immune cell infiltration and other parameters. Data regarding immune infiltration levels among patients with LUAD were obtained, and the association between IRGPM and immune cell infiltration was assessed.

Reverse transcription-quantitative (RT-q) PCR

Total RNA was obtained from the LUAD and corresponding adjacent normal tissues of 10 patients using TRIzol® reagent (Invitrogen; Thermo Fisher Scientific, Inc.) and then reverse transcribed into cDNA using the First Strand cDNA Synthesis kit (New England Biolabs, Inc.), according to the manufacturer's protocol. PCR amplification was performed with a SYBR Green PCR kit (ABM, Inc.), according to the manufacturer's protocol, using the Applied Biosystems 7500Real-Time PCR system (Applied Biosystems; Thermo Fisher Scientific, Inc.). The primer sequences used for qPCR are presented in Table I. The following thermocycling conditions were used for qPCR: Initial denaturation of 95°C for 10 min; 40 cycles of 95°C for 30 sec, 60°C for 30 sec and 72°C for 30 sec; and a final extension at 75°C for 7 min. Relative mRNA expression levels were measured using the 2−ΔΔCq method (20) and normalized to the internal reference gene GAPDH. All experiments were performed in triplicate.

Table I.

Primer sequences for reverse transcription quantitative-PCR.

Table I.

Primer sequences for reverse transcription quantitative-PCR.

PrimerSequence, 5′→3′
IL11
  Forward GTGGCCAAGATACAGCTGTCGC
  Reverse GGTAGGACAGTAGGTCCGCTC
LGR4
  Forward TCCACCTGGAAAGTCTGA
  Reverse GGTTAGATTTGATTACGCTGT
CRABP1
  Forward ATTCTCGAGCCACCATGCCCAACTTC
  ReverseACAGGATCCC TGCCTTCACTCTCGG
GAPDH
  Forward CAACGAATTTGGCTACAGCA
  Reverse AGGGGTCTACATGGCAACTG
Statistical analysis

Survival analysis of data from patients in the prognostic model was performed using the R survival package. Survival curves were generated using the Kaplan-Meier method and a log-rank test was used to compare the differences between the two groups. To validate the performance of the prognostic signature, the area under the survival receiver operating characteristic (ROC) curve was calculated using the R survival ROC package (21). The expression levels of genes between different groups were evaluated using the unpaired Student's t test. P<0.05 was considered to indicate a statistically significant difference.

Results

Identification of DEGs

Using the edgeR package, 2,672 DEGs were identified in patients with LUAD, 2,191 and 481 of which were upregulated and downregulated, respectively (Fig. 1A and B). Upon further comparison with immune gene lists from the ImmPort database, 273 DEIRGs were identified, 210 and 63 of which were up-and downregulated, respectively (Fig. 1C and D).

Construction of the prognostic model

The median gene expression was set as the cut-off value to divide all genes into two groups, high expression group and low expression group. Univariate analysis was used to identify survival-associated IRGS. The results demonstrated that high expression levels of: S100P [hazard ratio (HR), 1.218; 95% confidence interval (CI), 1.578–2.018; P=0.005), CPABP1 (HR, 1.343; 95% CI, 1.659–2.106; P=0.005), BIRC5 (HR, 1.645; 95% CI, 1.388–1.951; P=0.002), IGKV4-1 (HR, 1.665; 95% CI, 1.322–2.098; P=0.009), IL11 (HR, 1.728; 95% CI, 1.349–2.636; P<0.001), INHA (HR, 1.226; 95% CI, 0.972–2.265; P=0.004), INSL4 (HR, 1.978; 95% CI, 1.493–2.872; P=0.007) and LGR4 (HR, 1.678; 95% CI, 1.433–2.172; P=0.001) were associated with worse OS compared with the low expression group. Conversely, low expression levels of: ADRB2 (HR, 0.711; 95% CI, 0.553–0.712; P=0.004) and VIPR1 (HR, 0.651; 95% CI, 0.413–0.732; P<0.001) were associated with worse OS compared with the high expression group (Table II). Based on multivariate Cox regression analysis of survival-associated IRGs, a prognostic model was constructed which divided the patients into two groups (high risk score group and low risk score), according to OS time and the median risk score, using the following formula: (Expression levels of CRABP1*0.00326) + (expression levels of IGKV4-1*-0.00036) + (expression levels of IL-11*0.14555) + (expression levels of INHA*0.00475) + (expression levels of LGR4*0.01757) + (expression levels of VIPR1*-0.17506). The results demonstrated that the expression levels of IGKV4-1 and VIPR1 in high risk score group were significantly higher than low risk score group, while the expression levels of CRABP1, IL11, INHA and LGR4 in high risk score group were significantly lower than low risk score group (Fig. 2A). The risk coefficient (Fig. 2B) and mortality (Fig. 2C) were significantly higher in high risk score group compared with the low risk score group, respectively.

Table II.

Univariate Cox regression analysis of immune related genes of patients with lung adenocarcinoma.

Table II.

Univariate Cox regression analysis of immune related genes of patients with lung adenocarcinoma.

Overall survival Univariate analysis

Immune related geneHR (95% CI)P-value
S100P1.218 (1.578–2.018)0.005
CPABP11.343 (1.659–2.106)0.005
BIRC51.645 (1.388–1.951)0.002
IGKV4-11.665 (1.322–2.098)0.009
IL111.728 (1.349–2.636)<0.001
INHA1.226 (0.972–2.265)0.004
INSL41.978 (1.493–2.872)0.007
ADRB20.711 (0.553–0.712)0.004
LGR41.678 (1.433–2.172)0.001
VIPR10.651 (0.413–0.732)<0.001

[i] CI, confidence interval; HR, hazard ratio.

Gene functional enrichment analysis of differentially expressed IRGs

The biological functions of 273 IRGs were further investigated using GO and KEGG analyses. The results showed that the ‘extracellular region part’ was the most frequent GO biological process category (P<0.05; Fig. 3A). The top 10 enriched GO networks and top 40 genes involved in GO networks are presented in Fig. 3A and B, respectively.

The top significantly enriched pathways were obtained using KEGG pathway analysis (Fig. 4A); these were ‘cytokine-cytokine receptor interaction’, ‘neuroactive ligand-receptor interaction’, ‘JAK-STAT signaling pathway’, ‘IL-17 signaling pathway’ and ‘viral protein interaction with cytokine and cytokine receptor’. Based on the relationship between IRGs and the top 5 KEGG pathways, a visual network was constructed using Cytoscape version 3.6.1 (Fig. 4B).

Clinical outcome of patients with LUAD using the prognostic model

Kaplan-Meier plots were used to divide patients into high and low risk score groups according to OS time. The area under the ROC curve was 0.800, suggesting that a prognostic model based on IRGs could be used to monitoring survival (Fig. 5A and B). Univariate analyses showed that high American Joint Committee on Cancer (AJCC) stage (22) (HR, 1.645; 95% CI, 1.388–1.951; P<0.001), high tumor stage (22) (HR, 1.665; 95%; CI, 1.322–2.098; P<0.001), high node stage (22) (HR, 1.928; 95% CI, 1.549–2.426; P<0.001) and high risk score (HR, 1.978; 95% CI, 1.493–2.872; P<0.001) were significant risk factors for a poor prognosis (Table III). Using multivariate analysis, a high risks core (HR, 2.071; 95% CI, 1.313–3.425; P<0.001) was found to be independently associated with a less favorable OS time (Table III). Collectively, these data indicate that the risk scores are significantly higher among patients with advanced T (Fig. 5C) and high AJCC stages (Fig. 5D).

Table III.

Univariate and Multivariate Cox regression analysis of prognostic model (risk score) and clinical features of patients with lung adenocarcinoma.

Table III.

Univariate and Multivariate Cox regression analysis of prognostic model (risk score) and clinical features of patients with lung adenocarcinoma.

Overall survival

Univariate analysisMultivariate analysis


Risk factorsHR (95% CI)P-valueHR (95% CI)P-value
Age, years
  (>65 vs. ≤65)0.998 (0.978–1.018)0.8421.002 (0.981–1.023)0.863
Sex
  (Male vs. Female)0.843 (0.659–1.406)0.8430.864 (0.581–1.286)0.472
AJCC stage
  (I–II vs. III–IV)1.645 (1.388–1.951)<0.0011.547 (0.909–2.640)0.108
T stage
  (T1-2 vs. T3-4)1.665 (1.322–2.098)<0.0011.280 (0.985–1.662)0.065
N stage
  (N0 vs. N1-3)1.928 (1.549–2.426)<0.0011.281 (0.795–2.062)0.309
M stage
  (M0 vs. M1)1.226 (0.972–2.265)0.0961.408 (0.541–1.966)0.277
Risk score
  (Low vs. High)1.978 (1.493–2.872)<0.0012.071 (1.313–3.425)<0.001

[i] AJCC, American Joint Committee on Cancer; CI, confidence interval; HR, hazard ratio; M, metastasis; N, Node; T, Tumor.

Correlation analysis of the prognostic model and immune cell infiltration

Among the six immune cell types investigated (B cells, CD4 T cells, CD8 T cells, DCs, macrophages and neutrophils), the risk factors identified in the prognostic model were negatively correlated with B cell infiltration (r=−0.158; P=0.001; Fig. 6A); however, risk score was not associated with CD4+ T cells (r=−0.078; P=0.112; Fig. 6B), CD8+ T cells (r=−0.015; P=0.756; Fig. 6C), dendritic cells (r=−0.080; P=0.102; Fig. 6D), macrophages (r=−0.068; P=0.164; Fig. 6E) or neutrophils (r=0.018; P=0.715; Fig. 6F).

Analysis and validation of gene expression

To further validate the expression of relevant key genes in the prognostic model, three mRNAs (IL11, CARBP1 and LGR4) were randomly selected and their expression levels were evaluated in 10 pairs of LUAD and adjacent normal tissues. The expression levels of IL11, CRABP1 and LGR4 were higher in tumor tissues compared with adjacent normal tissues (Fig. 7A-C), which was consistent with the findings observed in TCGA database.

Discussion

The incidence and mortality rates of lung cancer in China are still increasing (23). A previous study indicated that IRGs are promising prognostic indicators of early stage lung cancer (8). A particular study screened for 40 genes and classified patients into high-risk and low-risk groups according to the immune signature; Patients with early non-squamous lung cancer demonstrated independent prognostic factors (8). As is well known, the treatment of advanced stage unresectable or metastatic lung cancer in China is difficult (24). Immunotherapy, such as immune checkpoint inhibitors, is primarily used for patients with metastatic lung cancer (5,6); therefore, the present study aimed to predict the prognoses of patients with LUAD using IRGs.

A total of six IRGs associated with prognosis were identified in patients with LUAD using TCGA database, and a prognostic model was established based on these genes (CRABP1, IGKV4-1, IL11, INHA, LGR4 and VIPR1). In this model, the overall survival duration of patients with high-risk disease was significantly shorter compared with patients with low-risk disease.

CRABP1 is a member of the fatty acid binding family of proteins, which binds to retinoic acid with high affinity (25). There are few studies on CRABP1 and lung cancer and the underlying molecular mechanisms of CRABP1 function in lung cancer remain unclear. Favorskaya et al demonstrated thatCRABP1 significantly alters the expression levels of CRABP in NSCLC samples (26). IGKV4-1 is inherently autoreactive and has been implicated in B-cell mediated autoimmune diseases and dysregulated B-cell tolerance (2729). However, the function of IGKV4-1 in lung cancer remains unknown. VIPR1 is a G protein-coupled receptor that is widely distributed in the normal tissues of humans, and that serves a role in physiological functions (30). Downregulation and deletion of VIPR1 have been detected in patients with LUAD (31). Gong et al (32) demonstrated that LGR4 was expressed in LUAD tissues but not in normal lung tissue. The group reported that LGR4 and IQGAP1 served a role in the regulation of tumor growth and metastasis in lung cancer cells. IL-11 is a member of the IL-6 group and binds to its corresponding receptors. IL-11 is an important inflammatory mediator that can affect the activity of a variety of immune cells (3335). Increased IL-11 expression levels have been associated with various types of cancer, including LUAD (3638).

To further explain some of these potential mechanisms, gene functional enrichment analysis was performed. It was demonstrated that IRGs were primarily enriched in ‘cytokine-cytokine receptor interaction’, ‘neuroactive ligand-receptor interaction’ and the ‘JAK-STAT signaling pathway’. Among the above prognosis-related immune genes, IL-11 was associated with ‘cytokine-cytokine receptor interaction’ and the ‘JAK-STAT signaling pathway’.

Cytokines are secreted glycoproteins that function as intercellular mediators, promoting cellular proliferation, differentiation and apoptosis (39). On the other hand, cytokines secreted by tumors can promote the recruitment of immunosuppressive cells, resulting in tumor metastasis (40). Previous studies have identified a variety of cytokines that can regulate hematopoiesis, induce inflammatory responses and control immune responses through the Janus kinase (JAK) signaling pathway (41,42). The JAK family contains four members: JAK1, JAK2, JAK3 and TYK2 (43). JAK kinases are a potential target for the treatment of tumors due to the oncogenic effects and the promotion of tumor inflammatory responses via JAK signaling (41). When cytokines bind to their cognate receptors, JAK is activated and phosphorylates downstream signaling and transcriptional activator (STAT), ultimately leading to tumor invasion, angiogenesis, apoptosis and metastasis (41). IL-11 activates downstream JAK/STAT signaling proteins via a gp130 homodimer (42). The suppressor of cytokine signaling proteins regulate JAK/STAT signaling pathways by serving as feedback inhibitors of activated JAK (44). Currently, few studies have investigated IL-11 and JAK signaling pathways in tumors, and the underlying molecular mechanisms of action remain unknown. In the present study, KEGG analysis revealed that IRGs are mainly enriched in these two signaling pathways. Further network construction revealed that IL-11 is closely associated with these two pathways. The explanation of this relationship between the two pathways in the present study may provide a basis for determining the prognosis of lung cancer.

In the present study, the immune gene-related prognostic index was not only associated with the prognosis of LUAD but was also negatively correlated with immune B cell infiltration. Tumor-infiltrating B lymphocytes have seemingly conflicting effects in tumors. On the one hand, B cells function in the inhibition of tumor cell proliferation via antigen restricted tumoricidal responses; on the other hand, B cells also act by suppressing the immune system, thus promoting tumor growth, proliferation and metastasis (45). Tumor-infiltrating B lymphocyte-derived lymphotoxin has been reported to promote the progression of androgen-independent prostate cancer by activating the Nuclear Factor κ-B and STAT3 signaling pathways (46). Previous studies have shown that tumor-infiltrating CD20+ B cells reside in close proximity to CD8+ T cells, and in patients with ovarian cancer, infiltration of CD20+ B and CD8+ T cells prolongs DSS (disease-specific survival) compared with CD8+ T cell infiltration alone (11). Pinto et al (47) demonstrated that patients with LUAD, with mutated K-RAS had associated B cell infiltration. B cells also exert a number of anti-tumor effects. First, they can stimulate other immune cells to produce cytokines, particularly those that enhance the activity of CTL (cytotoxic T-lymphocytes) (48). Secondly, B cells secrete granzyme B to directly kill tumor cells (48). Furthermore, B cells can suppress pancreatic cancer via antibody-dependent mechanisms (48,49). However, the functions of a number of prognosis-related genes in lung cancer remain unclear. For example, the IGKV4-1 gene encodes a B cell receptor (50). Previous studies have not described the relationship between IGKV4-1 and lung cancer. It is unclear whether the IGKV4-1 gene serves a role in B cell infiltration in lung cancer, and further research is required to investigate the possible underlying molecular mechanisms. Due to the negative correlation between the prognostic model and B cell infiltration in the present study, some patients may have had low risk scores due to the anti-tumor effects exerted by infiltrating B lymphocytes.

The present study was not without limitations. The primary limitation was the small sample size, which will be increased in future studies. Although the conclusions of the present study were drawn based on evidence from TCGA database, only gene expression was verified. Thus, it remains critical to further verify the applicability of survival models.

In summary, the present study identified prognosis-related immune genes using TCGA database and established a prognostic model for patients with LUAD. Using multivariate analysis with other clinicopathological features, such as age, gender and TNM stage, risk score was revealed to be an independent prognostic factor, hence, the present model can predict the prognoses of patients with LUAD. In addition, the prognostic model was associated with B cell infiltration and the present study may provide novel evidence for the prognosis and immunotherapy of patients with LUAD in the future.

Acknowledgements

The authors would like to thank Dr Wei Shan (Department of Gastrointestinal Surgery, Renmin Hospital of Wuhan University) for his assistance regarding statistics.

Funding

The present study was supported by The Science and Education for Health Foundation of Suzhou for Youth (grant nos. kjxw2018030 and kjxw2018032), The Science and Technology Project Foundation of Suzhou (grant no. SS201651), The Education Research Project Foundation of Nanjing Medical University (grant no. FZS-ZD-201701), The Jiangsu Province Medical Key Discipline (grant no. ZDXKC2016007) and Suzhou Oncology Clinical Center (grant no. Szzx201506).

Availability of data and materials

The datasets generated and analyzed during the current study are available in The Cancer Genome Atlas repository (https://portal.gdc.cancer.gov/).

Authors' contributions

ZLD and WJW conceived and designed the study. YQH and JPS performed the statistical analysis. HW, MSW and YW were involved in the writing of the manuscript and in the interpretation of the results. All authors read and approved the final manuscript.

Ethics approval and consent to participate

The present study was approved by The Medical Ethics Committees of Jining Cancer Hospital (approval no. 20190067). Written informed consent was provided by all patients prior to the study start.

Patient consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

Glossary

Abbreviations

Abbreviations:

AJCC

American Joint Committee on Cancer

CI

confidence interval

DCs

dendritic cells

DEIRGs

differentially expressed immune-related genes

GO

gene ontology

HR

hazard ratio

IRGPM

immune-related gene prognostic model

IRGs

immune-related genes

JAK-STAT

Janus kinase-signaling and transcriptional activator

KEGG

Kyoto Encyclopedia of Genes and Genomes

LUAD

lung adenocarcinoma

NSCLC

non-small cell lung cancer

OS

overall survival

PD-1

programmed death-1

PD-L1

programmed death-ligand 1

ROC

receiver operating characteristic

TCGA

The Cancer Genome Atlas

TILs

tumor-infiltrating lymphocytes

References

1 

Siegel RL, Miller KD and Jemal A: Cancer statistics, 2017. CA Cancer J Clin. 67:7–30. 2017. View Article : Google Scholar : PubMed/NCBI

2 

Sher T, Dy GK and Adjei AA: Small cell lung cancer. Mayo Clin Proc. 83:355–367. 2008. View Article : Google Scholar : PubMed/NCBI

3 

Duffy MJ and O'Byrne K: Tissue and blood biomarkers in lung cancer: A review. Adv Clin Chem. 86:1–21. 2018. View Article : Google Scholar : PubMed/NCBI

4 

Meza R, Meernik C, Jeon J and Cote ML: Lung cancer incidence trends by gender, race and histology in the United States, 1973–2010. PLoS One. 10:e01213232015. View Article : Google Scholar : PubMed/NCBI

5 

Garon EB, Rizvi NA, Hui R, Leighl N, Balmanoukian AS, Eder JP, Patnaik A, Aggarwal C, Gubens M, Horn L, et al: Pembrolizumab for the treatment of non-small-cell lung cancer. N Engl J Med. 372:2018–2028. 2015. View Article : Google Scholar : PubMed/NCBI

6 

Hellmann MD, Rizvi NA, Goldman JW, Gettinger SN, Borghaei H, Brahmer JR, Ready NE, Gerber DE, Chow LQ, Juergens RA, et al: Nivolumab plus ipilimumab as first-line treatment for advanced non-small-cell lung cancer (CheckMate 012): Results of an open-label, phase 1, multicohort study. Lancet Oncol. 18:31–41. 2017. View Article : Google Scholar : PubMed/NCBI

7 

Postow MA, Callahan MK and Wolchok JD: Immune checkpoint blockade in cancer therapy. J Clin Oncol. 33:1974–1982. 2015. View Article : Google Scholar : PubMed/NCBI

8 

Li B, Cui Y, Diehn M and Li R: Development and validation of an individualized immune prognostic signature in early-stage nonsquamous non-small cell lung cancer. JAMA Oncol. 3:1529–1537. 2017. View Article : Google Scholar : PubMed/NCBI

9 

Al-Shibli KI, Donnem T, Al-Saad S, Persson M, Bremnes RM and Busund LT: Prognostic effect of epithelial and stromal lymphocyte infiltration in non-small cell lung cancer. Clin Cancer Res. 14:5220–5227. 2008. View Article : Google Scholar : PubMed/NCBI

10 

Nedergaard BS, Ladekarl M, Nyengaard JR and Nielsen K: A comparative study of the cellular immune response in patients with stage IB cervical squamous cell carcinoma. Low numbers of several immune cell subtypes are strongly associated with relapse of disease within 5 years. Gynecol Oncol. 108:106–111. 2008. View Article : Google Scholar : PubMed/NCBI

11 

Nielsen JS, Sahota RA, Milne K, Kost SE, Nesslinger NJ, Watson PH and Nelson BH: CD20+ tumor -infiltrating lymphocytes have an atypical CD27- memory phenotype and together with CD8+ T cells promote favorable prognosis in ovarian cancer. Clin Cancer Res. 18:3281–3292. 2012. View Article : Google Scholar : PubMed/NCBI

12 

Liu J, Lichtenberg T, Hoadley KA, Poisson LM, Lazar AJ, Cherniack AD, Kovatich AJ, Benz CC, Levine DA, Lee AV, et al: An integrated TCGA pan-cancer clinical data resource to drive high-quality survival outcome analytics. Cell. 173:400–416.e11. 2018. View Article : Google Scholar : PubMed/NCBI

13 

Bhattacharya S, Andorf S, Gomes L, Dunn P, Schaefer H, Pontius J, Berger P, Desborough V, Smith T, Campbell J, et al: ImmPort: Disseminating data to the public for the future of immunology. Immunol Res. 58:234–239. 2014. View Article : Google Scholar : PubMed/NCBI

14 

Robinson MD, McCarthy DJ and Smyth GK: EdgeR: A bioconductor package for differential expression analysis of digital gene expression data. Bioinformatics. 26:139–140. 2010. View Article : Google Scholar : PubMed/NCBI

15 

Dennis G Jr, Sherman BT, Hosack DA, Yang J, Gao W, Lane HC and Lempicki RA: DAVID: Database for annotation, visualization, and integrated discovery. Genome Biol. 4:P32003. View Article : Google Scholar : PubMed/NCBI

16 

Yu G, Wang LG, Han Y and He QY: ClusterProfiler: An R package for comparing biological themes among gene clusters. OMICS. 16:284–287. 2012. View Article : Google Scholar : PubMed/NCBI

17 

Huang da W, Sherman BT and Lempicki RA: Bioinformatics enrichment tools: Paths toward the comprehensive functional analysis of large gene lists. Nucleic Acids Res. 37:1–13. 2009. View Article : Google Scholar : PubMed/NCBI

18 

Wagenmakers EJ and Farrell S: AIC model selection using Akaike weights. Psychon Bull Rev. 11:192–196. 2004. View Article : Google Scholar : PubMed/NCBI

19 

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

20 

Livak KJ and Schmittgen TD: Analysis of relative gene expression data using real-time quantitative PCR and the 2(-Delta Delta C(T)) method. Methods. 25:402–408. 2001. View Article : Google Scholar : PubMed/NCBI

21 

Heagerty PJ, Lumley T and Pepe MS: Time-dependent ROC curves for censored survival data and a diagnostic marker. Biometrics. 56:337–344. 2000. View Article : Google Scholar : PubMed/NCBI

22 

Detterbeck FC, Boffa DJ, Kim AW and Tanoue LT: The eighth edition lung cancer stage classification. Chest. 151:193–203. 2017. View Article : Google Scholar : PubMed/NCBI

23 

Chen W, Zheng R, Baade PD, Zhang S, Zeng H, Bray F, Jemal A, Yu XQ and He J: Cancer statistics in China, 2015. CA Cancer J Clin. 66:115–132. 2016. View Article : Google Scholar : PubMed/NCBI

24 

Chinese Association for Clinical Oncologists, . Clinical practice guideline for stage IV primary lung cancer in China(2020 version). Zhonghua Zhong Liu Za Zhi. 42:1–16. 2020.(In Chinese). PubMed/NCBI

25 

Dong D, Ruuska SE, Levinthal DJ and Noy N: Distinct roles for cellular retinoic acid-binding proteins I and II in regulating signaling by retinoic acid. J Biol Chem. 274:23695–23698. 1999. View Article : Google Scholar : PubMed/NCBI

26 

Favorskaya I, Kainov Y, Chemeris G, Komelkov A, Zborovskaya I and Tchevkina E: Expression and clinical significance of CRABP1 and CRABP2 in non-small cell lung cancer. Tumour Biol. 35:10295–10300. 2014. View Article : Google Scholar : PubMed/NCBI

27 

Yurasov S, Wardemann H, Hammersen J, Tsuiji M, Meffre E, Pascual V and Nussenzweig MC: Defective B cell tolerance checkpoints in systemic lupus erythematosus. J Exp Med. 201:703–711. 2005. View Article : Google Scholar : PubMed/NCBI

28 

Pugh-Bernard AE, Silverman GJ, Cappione AJ, Villano ME, Ryan DH, Insel RA and Sanz I: Regulation of inherently autoreactive VH4-34 B cells in the maintenance of human B cell tolerance. J Clin Invest. 108:1061–1070. 2001. View Article : Google Scholar : PubMed/NCBI

29 

Cappione A III, Anolik JH, Pugh-Bernard A, Barnard J, Dutcher P, Silverman G and Sanz I: Germinal center exclusion of autoreactive B cells is defective in human systemic lupus erythematosus. J Clin Invest. 115:3205–3216. 2005. View Article : Google Scholar : PubMed/NCBI

30 

Reubi JC, Laderach U, Waser B, Gebbers JO, Robberecht P and Laissue JA: Vasoactive intestinal peptide/pituitary adenylate cyclase-activating peptide receptor subtypes in human tumors and their tissues of origin. Cancer Res. 60:3105–3112. 2000.PubMed/NCBI

31 

Mlakar V, Strazisar M, Sok M and Glavac D: Oligonucleotide DNA microarray profiling of lung adenocarcinoma revealed significant downregulation and deletions of vasoactive intestinal peptide receptor 1. Cancer Invest. 28:487–494. 2010. View Article : Google Scholar : PubMed/NCBI

32 

Gong X, Yi J, Carmon KS, Crumbley CA, Xiong W, Thomas A, Fan X, Guo S, An Z, Chang JT and Liu QJ: Aberrant RSPO3-LGR4 signaling in Keap1-deficient lung adenocarcinomas promotes tumor aggressiveness. Oncogene. 34:4692–4701. 2015. View Article : Google Scholar : PubMed/NCBI

33 

Zhang J, Zhang Y, Dutta DJ, Argaw AT, Bonnamain V, Seto J, Braun DA, Zameer A, Hayot F, Lòpez CB, et al: Proapoptotic and antiapoptotic actions of Stat1 versus Stat3 underlie neuroprotective and immunoregulatory functions of IL-11. J Immunol. 187:1129–1141. 2011. View Article : Google Scholar : PubMed/NCBI

34 

Trepicchio WL, Wang L, Bozza M and Dorner AJ: IL-11 regulates macrophage effector function through the inhibition of nuclear factor-kappaB. J Immunol. 159:5661–5670. 1997.PubMed/NCBI

35 

Curti A, Ratta M, Corinti S, Girolomoni G, Ricci F, Tazzari P, Siena M, Grande A, Fogli M, Tura S and Lemoli RM: Interleukin-11 induces Th2 polarization of human CD4(+) T cells. Blood. 97:2758–2763. 2001. View Article : Google Scholar : PubMed/NCBI

36 

Pastor MD, Nogal A, Molina-Pinelo S, Quintanal-Villalonga Á, Meléndez R, Ferrer I, Romero-Romero B, De Miguel MJ, López-Campos JL, Corral J, et al: IL-11 and CCL-1: Novel protein diagnostic biomarkers of lung adenocarcinoma in bronchoalveolar lavage fluid (BALF). J Thorac Oncol. 11:2183–2192. 2016. View Article : Google Scholar : PubMed/NCBI

37 

Yamazumi K, Nakayama T, Kusaba T, Wen CY, Yoshizaki A, Yakata Y, Nagayasu T and Sekine I: Expression of interleukin-11 and interleukin-11 receptor alpha in human colorectal adenocarcinoma; immunohistochemical analyses and correlation with clinicopathological factors. World J Gastroenterol. 12:317–321. 2006. View Article : Google Scholar : PubMed/NCBI

38 

Hanavadi S, Martin TA, Watkins G, Mansel RE and Jiang WG: Expression of interleukin 11 and its receptor and their prognostic value in human breast cancer. Ann Surg Oncol. 13:802–808. 2006. View Article : Google Scholar : PubMed/NCBI

39 

Morris R, Kershaw NJ and Babon JJ: The molecular details of cytokine signaling via the JAK/STAT pathway. Protein Sci. 27:1984–2009. 2018. View Article : Google Scholar : PubMed/NCBI

40 

Hanahan D and Coussens LM: Accessories to the crime: Functions of cells recruited to the tumor microenvironment. Cancer Cell. 21:309–322. 2012. View Article : Google Scholar : PubMed/NCBI

41 

O'Shea JJ, Schwartz DM, Villarino AV, Gadina M, McInnes IB and Laurence A: The JAK-STAT pathway: Impact on human disease and therapeutic intervention. Annu Rev Med. 66:311–328. 2015. View Article : Google Scholar : PubMed/NCBI

42 

Buzzelli JN, O'Connor L, Scurr M, Chung Nien Chin S, Catubig A, Ng GZ, Oshima M, Oshima H, Giraud AS, Sutton P, et al: Overexpression of IL-11 promotes premalignant gastric epithelial hyperplasia in isolation from germline gp130-JAK-STAT driver mutations. Am J Physiol Gastrointest Liver Physiol. 316:G251–G262. 2019. View Article : Google Scholar : PubMed/NCBI

43 

Stark GR and Darnell JE Jr: The JAK-STAT pathway at twenty. Immunity. 36:503–514. 2012. View Article : Google Scholar : PubMed/NCBI

44 

Inagaki-Ohara K, Kondo T, Ito M and Yoshimura A: SOCS, inflammation, and cancer. JAKSTAT. 2:e240532013.PubMed/NCBI

45 

Guo S, Contratto M, Miller G, Leichman L and Wu J: Immunotherapy in pancreatic cancer: Unleash its potential through novel combinations. World J Clin Oncol. 8:230–240. 2017. View Article : Google Scholar : PubMed/NCBI

46 

Ammirante M, Luo JL, Grivennikov S, Nedospasov S and Karin M: B-cell-derived lymphotoxin promotes castration-resistant prostate cancer. Nature. 464:302–305. 2010. View Article : Google Scholar : PubMed/NCBI

47 

Pinto R, Petriella D, Lacalamita R, Montrone M, Catino A, Pizzutilo P, Botticella MA, Zito FA, Del Bene G, Zonno A, et al: KRAS-driven lung adenocarcinoma and B cell infiltration: Novel insights for immunotherapy. Cancers (Basel). 11(pii): E11452019. View Article : Google Scholar : PubMed/NCBI

48 

Tsou P, Katayama H, Ostrin EJ and Hanash SM: The emerging role of B cells in tumor immunity. Cancer Res. 76:5597–5601. 2016. View Article : Google Scholar : PubMed/NCBI

49 

Lee KE, Spata M, Bayne LJ, Buza EL, Durham AC, Allman D, Vonderheide RH and Simon MC: Hif1a deletion reveals pro-neoplastic function of B cells in pancreatic neoplasia. Cancer Discov. 6:256–269. 2016. View Article : Google Scholar : PubMed/NCBI

50 

Asare A, Kanaparthi S, Lim N, Phippard D, Vincenti F, Friedewald J, Pavlakis M, Poggio E, Heeger P, Mannon R, et al: B cell receptor genes associated with tolerance identify a cohort of immunosuppressed patients with improved renal allograft graft function. Am J Transplant. 17:2627–2639. 2017. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

November-2020
Volume 20 Issue 5

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
Wang H, Wang M, Wang Y, Huang Y, Shi J, Ding Z and Wang W: Prognostic value of immune related genes in lung adenocarcinoma. Oncol Lett 20: 259, 2020
APA
Wang, H., Wang, M., Wang, Y., Huang, Y., Shi, J., Ding, Z., & Wang, W. (2020). Prognostic value of immune related genes in lung adenocarcinoma. Oncology Letters, 20, 259. https://doi.org/10.3892/ol.2020.12122
MLA
Wang, H., Wang, M., Wang, Y., Huang, Y., Shi, J., Ding, Z., Wang, W."Prognostic value of immune related genes in lung adenocarcinoma". Oncology Letters 20.5 (2020): 259.
Chicago
Wang, H., Wang, M., Wang, Y., Huang, Y., Shi, J., Ding, Z., Wang, W."Prognostic value of immune related genes in lung adenocarcinoma". Oncology Letters 20, no. 5 (2020): 259. https://doi.org/10.3892/ol.2020.12122