Skip to main content
American Journal of Cancer Research logoLink to American Journal of Cancer Research
. 2020 Jun 1;10(6):1770–1784.

Novel genetic variants of KIR3DL2 and PVR involved in immunoregulatory interactions are associated with non-small cell lung cancer survival

Yufeng Wu 1,2,4, Sen Yang 1,2,4, Hongliang Liu 2,4, Sheng Luo 5, Thomas E Stinchcombe 2,3, Carolyn Glass 2,6, Li Su 7, Sipeng Shen 7, David C Christiani 7,8, Qiming Wang 1, Qingyi Wei 2,3,4
PMCID: PMC7339263  PMID: 32642289

Abstract

Immunoregulatory interactions play a pivotal role in immune surveillance, recognition, and killing, particularly its internal pathway, likely playing an important role in immune escape. By using two genotyping datasets, one from the Prostate, Lung, Colorectal and Ovarian (PLCO) Cancer screening trial (n = 1,185) as the discovery, and the other from Harvard Lung Cancer Susceptibility (HLCS) study (n = 984) as the validation, we evaluated associations between 4,713 genetic variants (338 genotyped and 4,375 imputed) in 60 genes involved in immunoregulatory interactions and survival of non-small cell lung cancer (NSCLC). We found that 115 SNPs were significantly associated with NSCLC overall survival in the discovery, of which four remained significant after validation by the HLCS dataset after multiple test correction by Bayesian false discovery probability. Final combined analysis identified two independent SNPs (KIR3DL2 rs4487030 A>G and PVR rs35385129 C>A) that predicted NSCLC survival with a combined hazards ratio of 0.84 (95% confidence interval = 0.76-0.93, P = 0.001) and 0.84 (95% confidence interval = 0.73-0.97, P = 0.021), respectively. Besides, expression quantitative trait loci analyses showed that these two survival-associated SNPs of KRI3DL2 and PVR were significantly associated with their mRNA expression levels in both normal lung tissues and whole blood cells. Additional analyses suggested an oncogenic role for KRI3DL2 and a suppressor role for PVR on the survival. Once further validated, genetic variants of KIR3DL2 and PVR may be potential prognostic markers for NSCLC survival.

Keywords: Non-small cell lung cancer, immunoregulatory interactions, single nucleotide polymorphism, survival

Introduction

Lung cancer is the most commonly diagnosed cancer (11.6% of the total cases) and the leading cause of cancer deaths (18.4% of the total cancer deaths) in the world [1]. Non-small cell lung cancer (NSCLC) is the most common histologic type of lung cancer, accounting for about 80-85% of the total cases [2]. In the past decades, chemotherapy, radiotherapy, and surgery are the main treatments for NSCLC until the emergence of targeted therapies and immunotherapeutic drugs. After many years of disappointing results, treatment choice has finally changed, and immunotherapy has become a clinically validated treatment for many cancers, including NSCLC [3]. Although the use of immunotherapy has a much-improved treatment response, the overall 5-year survival rate of NSCLC is still less than 15% [4,5] and current research has focused on identifying additional factors that improve the effectiveness of immunotherapy. Studies have shown that genetic variants, single-nucleotide polymorphisms (SNPs) in particular as one of the most common genetic variations, have a significant impact on outcomes of cancer treatment [6,7].

The idea of using the host immune system to combat cancer dates back to decades ago. Researchers have realized that various components of the immune system play pivotal roles in protecting humans from cancer. Following numerous disappointing efforts and clinical failures, cancer immunotherapy is now widely considered the “fourth-pillar” of cancer therapy in addition to the other three conventional treatments [8]. Currently immune checkpoint inhibitors alone or in combination with chemotherapy are the standard first-line therapy for patients with metastatic non-small cell lung cancer. One of the representative tumor immunotherapy agents is the autologous cellular immunotherapy sipuleucel-T and anti-cytotoxic T lymphocyte-associated protein 4 antibody, also so-called ipilimumab, which was approved for prostate cancer treatment in 2010 [9], and the other is the anti-programmed cell death protein 1 antibody (PD1) for the treatment of melanoma approved in 2014 [10]. However, the proportion of cancer patients with indications for current immunotherapies remains small, and there are also many problems encountered in the immunotherapies, such as the screening of sensitive patients and, more importantly, dealing with immune drug resistance [11-14]. Although the immune system can differentiate protein structures at the molecular level, cancer cells manage to escape the host immune recognition and subsequent immune killing [15]. In the process of immunotherapy, the immune recognition of malignant cells exerts some selective pressure on the developing tumors, resulting in the growth of tumor cells evolving with low immunogenicity but a strong anti-apoptotic ability. In this case, immune escape occurs, leading to the failure of immunotherapy. Therefore, it is crucial to identify additional survival-related factors involved in immunoregulatory interactions.

Genetic variation, including SNPs, in some key genes in the signaling pathway for immunoregulatory interactions may be involved in the disorder or over-activation of the entire signaling pathway, modulating the effect of the immune system on tumor growth and progression. However, the roles of genetic variants in candidate genes involved in the immunoregulatory interaction signaling and their biological functions in tumor growth or progress remain unknown. As a promising hypothesis-driven method in the post-GWAS (genome-wide association study) era, the biological pathway-based approach has been applied to reanalyze published GWAS datasets and to test the cumulative effect of SNPs across multiple genes in the same biological pathway [16]. Therefore, we hypothesized that genetic variants in genes involved in the immunoregulatory interaction pathway are associated with survival of lung cancer patients. We tested this hypothesis by using genotyping data from two independently published GWAS datasets, focusing on those SNPs that may alter their gene functions and thus most likely have biological and functional consequences.

Materials and methods

Study populations

In the present study, we used the GWAS genotyping dataset from the Prostate, Lung, Colorectal and Ovarian (PLCO) Cancer Screening Trial as the discovery dataset. PLCO is a randomized control study conducted by the National Cancer Institute (NCI), involving 77500 men and 77500 women, aged between 55 and 74, registered in 10 medical centers in the United States from 1993 to 2011. All the participants were randomized into either the intervention arm that received a trial screening or the control arm that received standard care, were followed up for at least 13 years after the enrollment [17]. Blood samples and personal information including smoking status, histologic diagnosis, tumor stage, treatment method and family history were provided at enrollment [18]. A total of 1,185 NSCLC patients were eligible for survival analysis after excluding two individuals who had no follow-up information. Genomic DNA extracted from the whole blood samples of the participants were genotyped with Illumina HumanHap240Sv1.0 and HumanHap550v3.0 (dbGaP accession: phs000093.v2.p2 and phs000336.v1.p1) [19,20]. The 1,185 NSCLC patients with both complete follow-up information and genotype data were used for survival analysis. Each institutional review board of the participating institutions had approved the collection and all the participants had provided a written informed consent permitting the PLCO trial and use of the collected data.

Another GWAS dataset of 984 histology-confirmed Caucasian NSCLC patients from the Harvard Lung Cancer Susceptibility (HLCS) Study which began in 1992 was used as the validation dataset [21]. In the HLCS study, the whole blood samples and personal information were collected after diagnosis, and DNA from the blood samples was extracted with Auto Pure Large Sample nucleic acid purification system (QIAGEN Company, Venlo, Limburg, Netherlands) and genotyped by using the Illumina Humanhap610-Quad array. The genotyped data were used for imputation with the MACH software based on the sequencing data from the 1,000 Genomes Project [21].

The use of these two GWAS datasets was approved by both the Internal Review Board of Duke University School of Medicine (#Pro00054575) and the dbGaP database administration (#6404). The comparison of the characteristics between the PLCO trial (n = 1,185) and the HLCS study (n = 984) is presented in Table S1.

Gene and SNP selection

The genes involved in the immunoregulatory interaction signaling pathway were selected using the Molecular Signatures Database (http://software.broadinstitute.org/gsea/msigdb/index.jsp) with the keyword “immunoregulatory AND interactions”. After the removal of five pseudogenes, four genomic positions unavailable, and one gene on the X chromosome, 60 genes remained as candidate genes for further analysis (Table S2). Imputation with IMPUTE2 and the 1,000 Genomes Project data (phase 3) was performed for additional SNPs untyped for these candidate genes with their ± 500-kb flanking regions. After that, SNPs within the genes and their ± 2 kb flanking regions were extracted with the following criteria: an imputation information score ≥0.8 (Figure S1), a genotyping rate ≥95%, a minor allelic frequency (MAF) ≥5%, and Hardy-Weinberg equilibrium (HWE) ≥1×10-5. As a result, a total of 4,713 (338 genotyped and 4,375 imputed) SNPs were included in the PLCO dataset.

Statistical analyses

The endpoints for the analyses included overall survival (OS) and disease-specific survival (DSS). In the single-locus analysis, we first used multivariate Cox proportional hazards regression analysis to assess the association between each of the SNPs and NSCLC survival in an additive model using the PLCO dataset, with adjustment for clinical variables such as age, sex, smoking status, histology, tumor stage, chemotherapy, radiotherapy, surgery and the first four principal components (Table S3) by using the GenABEL package of R software [22]. We then used Bayesian false discovery probability (BFDP) with a cut-off value of 0.80 for multiple testing correction to lower the probability of potentially false-positive results as recommended [23,24], because most of the SNPs under investigation are in high LD as a result of imputation. We assigned a prior probability of 0.10 and detected an upper boundary hazard ratio (HR) of 3.0 for an association with variant genotypes or minor alleles of the SNPs with P<0.05. After that, we validated those chosen SNPs by using the HLCS dataset. Next, we performed an inverse variance weighted meta-analysis to combine the results of both discovery and validation datasets. In the meta-analysis, Cochran’s Q-test and the heterogeneity statistic (I 2) were performed to assess the inter-study heterogeneity. If no heterogeneity was observed between the two datasets (P het>0.10 and I 2<50%), a fixed-effects model was implemented; otherwise, a random-effects model was applied. Furthermore, a multivariate stepwise Cox model, including the first four principal components of the PLCO dataset, available demographic and clinical variables was performed to identify novel and independent SNPs. Finally, the model was further adjusted for 15 previously published SNPs for the survival of NSCLC from the same PLCO GWAS dataset.

We then used the combined genotypes or alleles to evaluate the cumulative effects of the identified SNPs and the Kaplan-Meier (KM) survival curves to show the survival probability associated with the combined genotypes or alleles. We also assessed possible interactions with an χ2-based Q-test between subgroups in the stratified analysis. We then performed the receiver operating characteristic (ROC) curve and time-dependent area under the curve (AUC) with the timeROC package of R software (version 3.5.0) to illustrate the prediction accuracy of the model integrating the effects of both clinical and genetic variables on NSCLC survival [25]. To evaluate the correlations between SNPs and the corresponding mRNA expression levels, we performed the expression quantitative trait loci (eQTL) analyses with a linear regression model performed with the R software. The mRNA expression data of genes were obtained from two sources: 373 European individuals included in the 1,000 Genomes Project as well as the data from whole blood cell samples from 589 subjects and normal lung tissue samples from 454 subjects included in the genotype-tissue expression (GTEx) project [26,27]. The bioinformatics functional prediction for the tagging SNPs was then performed with SNPinfo [28] (https://snpinfo.niehs.nih.gov), RegulomeDB [29] (http://www.regulomedb.org) and HaploReg [30] (http://archive.broadinstitute.org/mammals/haploreg/haploreg.php).

Lastly, the differences in mRNA expression levels were examined in 111 pairs of lung cancer tissues and adjacent normal tissues from the Cancer Genome Atlas (TCGA) database by using a paired t-test. We also assessed the differences in mRNA expression levels in a larger, but not paired, dataset from TCGA (http://ualcan.path.uab.edu), and the KM survival analysis was performed to assess the association between the mRNA expression levels and survival probability (http://kmplot.com/analysis/index.php?p=service&cancer=lung). All statistical analyses were performed with a statistical significance level of P<0.05 and using the SAS software (version 9.4; SAS Institute, Cary, NC, USA) unless otherwise indicated.

Data availability

The datasets are available from the National Center for Biotechnology Information Database of Genotypes and Phenotypes (dbGaP Study Accession: phs000093.v2.p2 and phs000336.v1.p1). Genome-wide imputation was performed based on the 1000 Genomes Project, phase III CEU, utilizing the IMPUTE2 software (October 2014 release).

Results

Associations between SNPs in the immunoregulatory interaction pathway genes and NSCLC survival

The overall flowchart in Figure 1 describes the design in the present study with genotyping data from two previously published GWAS datasets. The discovery genotyping dataset from the PLCO trial included 1,185 NSCLC patients, and the validation genotyping dataset from the HLCS GWAS study enrolled 984 NSCLC patients [31]. In the discovery PLCO dataset, 338 genotyped and 4,375 imputed SNPs in the 60 immunoregulatory interactions pathway genes with NSCLC after single-locus analysis (all imputation information score ≥0.8, Figure S1). As a result, we identified 115 SNPs to be significantly associated with NSCLC OS (P<0.05), with multiple testing correction by BFDP ≤0.80. After further validation by the HLCS genotyping dataset, four SNPs in three genes remained significant. In combined analysis of the PLCO and HLCS genotyping datasets, three of these four newly identified SNPs were associated with a better survival, including rs4487030 A>G of KIR3DL2 (HR = 0.84, P<0.001) and rs35385129 C>A (HR = 0.83, P<0.001) and rs28411142 C>T (HR = 0.84, P<0.001) of PVR, but rs3788142 G>A of ITGB2 was associated with a poor survival (HR = 1.16, P<0.001) (Table 1).

Figure 1.

Figure 1

The flowchart of the present study. Abbreviations: SNP, single nucleotide polymorphism; PLCO, Prostate, Lung, Colorectal and Ovarian cancer screening trial; NSCLC, non-small cell lung cancer; HLCS, Harvard lung cancer susceptibility study; KIR3DL2: killer cell immunoglobulin like receptor, three Ig domains and long cytoplasmic tail 2; PVR: PVR cell adhesion molecule.

Table 1.

Associations between four validated significant SNPs and overall survival in both discovery and validation genotyping datasets from two previously published NSCLC GWASs

SNP Allelea Gene PLCO (n = 1185) Harvard (n = 984) Combined-analysis



EAF HR (95% CI)b P b EAF HR (95% CI)c P c P het d I 2 HR (95% CI)e P e
rs4487030 A>G KIR3DL2 0.44 0.83 (0.75-0.92) <0.001 0.41 0.85 (0.74-0.97) 0.018 0.828 0 0.84 (0.77-0.91) <0.001
rs3788142 G>A ITGB2 0.24 1.17 (1.04-1.32) 0.008 0.24 1.15 (1.01-1.32) 0.030 0.886 0 1.16 (1.07-1.27) <0.001
rs35385129 C>A PVR 0.16 0.83 (0.72-0.96) 0.010 0.15 0.82 (0.70-0.97) 0.018 0.956 0 0.83 (0.74-0.92) <0.001
rs28411142 C>T PVR 0.16 0.83 (0.72-0.96) 0.012 0.15 0.85 (0.72-0.99) 0.037 0.862 0 0.84 (0.75-0.93) 0.001
a

Major allele > minor allele;

b

Adjusted for age, sex, stage, histology, smoking status, chemotherapy, radiotherapy, surgery, PC1, PC2, PC3 and PC4;

c

Adjusted for age, sex, stage, histology, smoking status, chemotherapy, radiotherapy, surgery, PC1, PC2 and PC3;

d

P het: P value for heterogeneity by Cochrane’s Q test;

e

Meta-analysis in the fix-effects model;

Abbreviations: EAF: effect allele frequency; HR: hazards ratio; CI: confidence interval; PLCO: Prostate, Lung, Colorectal, and Ovarian Cancer Screening Trial; HLCS: Harvard Lung Cancer Susceptibility Study.

Independent SNPs associated with NSCLC survival in the PLCO dataset

Subsequently, these four SNPs were tested for their independence in the multivariate stepwise Cox regression model using the PLCO dataset (because the HLCS dataset did not have individual genotyping data). Two SNPs (KIR3DL2 rs4487030 A>G) and (PVR rs35385129 C>A) remained significantly associated with a better survival (Table 2), after adjustment for other 15 previously reported survival-associated SNPs in the same PLCO GWAS dataset. As showed in Figure S2, the two SNPs from both PLCO and HLCS datasets are summarized in a Manhattan plot, respectively, and the regional association plot for each of these two SNPs is also shown in Figure S3.

Table 2.

Three indenpenden SNPs associated with OS in multivariate Cox proportional hazards regression analysis with adjustment for other covariates and previously published SNPs in the PLCO GWAS dataset

Variablesa Category Frequency HR (95% CI)a P a HR (95% CI)b P b
Age Continuous 1185 1.03 (1.02-1.05) <0.001 1.04 (1.02-1.05) <0.001
Sex Male 698 1.00 1.00
Female 487 0.77 (0.67-0.90) 0.001 0.78 (0.66-0.91) 0.002
Smoking status Never 115 1.00 1.00
Current 423 1.64 (1.22-2.19) 0.001 1.86 (1.37-2.50) <0.001
Former 647 1.62 (1.23-2.14) 0.001 1.83 (1.38-2.43) <0.001
Histology AD 577 1.00 1.00
SC 285 1.16 (0.97-1.40) 0.112 1.21 (1.00-1.47) 0.048
others 323 1.31 (1.10-1.55) 0.002 1.35 (1.13-1.61) 0.001
Tumor stage I-IIIA 655 1.00 1.00
IIIB-IV 528 2.89 (2.37-3.51) <0.001 3.05 (2.50-3.72) <0.001
Chemotherapy No 639 1.00 1.00
Yes 538 0.56 (0.47-0.67) <0.001 0.57 (0.47-0.68) <0.001
Radiotherapy No 762 1.00 1.00
Yes 415 0.98 (0.83-1.15) 0.786 0.96 (0.81-1.14) 0.628
Surgery No 637 1.00 1.00
Yes 540 0.21 (0.16-0.27) <0.001 0.20 (0.15-0.26) <0.001
KIR3DL2 rs4487030 A>G AA/GA/GG 381/560/244 0.84 (0.76-0.92) <0.001 0.84 (0.76-0.94) 0.001
PVR rs35385129 C>A CC/CA/AA 826/334/25 0.82 (0.72-0.95) 0.008 0.84 (0.73-0.97) 0.021
a

Stepwise analysis included age, sex, smoking status, tumor stage, tumor histology, chemotherapy, radiotherapy, surgery, PC1, PC2, PC3, PC4, and two newly validated SNPs in an additive model; P values for significant SNPs were in bold.

b

Fifteen previously published SNPs were used for the post-stepwise adjustment. Five SNPs were reported in previous publication (PMID: 27557513); One SNP was reported in the previous publication (PMID: 29978465); Two SNPs were reported in the previous publication (PMID: 30259978); Two SNPs were reported in the previous publication (PMID: 26757251); Three SNPs were reported in the previous publication (PMID: 30650190); Two SNPs were reported in the previous publication (PMID: 30989732);

Abbreviations: OS: overall survival; PLCO: Prostate, Lung, Colorectal, and Ovarian Cancer Screening Trial; GWAS: genome-wide association study; HR: hazards ratio; CI: confidence interval; AD: Adenocarcinoma; SC: Squamous cell carcinoma.

In the PLCO dataset with complete adjustment for available covariates, patients with the protective KIR3DL2 rs4487030 G allele (i.e., AG+GG) or the PVR rs35385129 A allele (i.e., CA+AA) had a better OS and DSS (P trend<0.001 and P trend = 0.003 for KIR3DL2 rs4487030 G, respectively and P trend = 0.010 and P trend = 0.039 for PVR rs35385129 A, respectively) (Table 3). In comparison with the AA genotype, the KIR3DL2 rs4487030 GG genotype was associated with a decreased risk of death (HR = 0.73, 95% CI = 0.54-0.82 and P<0.001 for OS and HR = 0.70, 95% CI = 0.56-0.87 and P = 0.001 for DSS), but the KIR3DL2 rs4487030 AG genotype was associated with a non-significant better survival (HR = 0.94, 95% CI = 0.80-1.11 and P = 0.033 for OS and HR = 0.84, 95% CI = 0.72-0.99 and P = 0.033 for DSS). Meanwhile, in comparison with the CC genotype, the PVR rs35385129 CA genotype was associated with a better significant better survival (HR = 0.84, 95% CI = 0.72-0.99 and P = 0.034 for OS and HR = 0.86, 95% CI = 0.73-1.02 and P = 0.079 for DSS), but the PVR rs35385129 AA genotype was associated with a non-significant survival (HR = 0.63, 95% CI = 0.35-1.12 and P = 0.112 for OS and HR = 0.70, 95% CI = 0.39-1.24 and P = 0.224 for DSS), likely due to a small observations for the AA genotype (Table 3).

Table 3.

Associations between genotypes of two independent SNPs and survival of NSCLC in the PLCO Trial

Genotype Frequency OSa DSSa


Death (%) HR (95% CI) P * Death (%) HR (95% CI) P *
KIR3DL2 rs4487030 A>Gb
    AA 376 264 (70.21) 1.00 234 (62.23) 1.00
    AG 557 377 (67.68) 0.94 (0.80-1.11) 0.486 342 (61.40) 0.98 (0.82-1.16) 0.789
    GG 242 148 (61.16) 0.73 (0.54-0.82) <0.001 * 133 (54.96) 0.70 (0.56-0.87) 0.001 *
    Trend <0.001 * 0.003 *
    AG+GG 799 525 (65.71) 0.85 (0.73-0.99) 0.034* 475 (59.45) 0.88 (0.75-1.04) 0.125
PVR rs35385129 C>Ab
    CC 820 559 (68.17) 1.00 500 (60.98) 1.00
    CA 330 218 (66.06) 0.84 (0.72-0.99) 0.034 * 197 (59.70) 0.86 (0.73-1.02) 0.079
    AA 25 12 (48.00) 0.63 (0.35-1.12) 0.112 12 (48.00) 0.70 (0.39-1.24) 0.224
    Trend 0.010 * 0.039 *
    CA+AA 355 230 (64.79) 0.83 (0.71-0.97) 0.016 * 209 (58.87) 0.85 (0.72-1.00) 0.049 *
NPG1 b,c
    0 254 178 (70.08) 1.00 155 (61.02) 1.00
    1 688 467 (67.88) 0.95 (0.79-1.13) 0.542 424 (61.63) 0.99 (0.82-1.19) 0.884
    2 233 144 (61.80) 0.69 (0.55-0.86) 0.001 * 130 (55.79) 0.73 (0.58-0.93) 0.012 *
    Trend 0.001 * 0.013 *
    0-1 942 645 (68.47) 1.00 579 (61.46)
    2 233 144 (61.80) 0.72 (0.60-0.86) <0.001 * 130 (55.79) 0.74 (0.61-0.90) 0.003 *
a

Adjusted for age, sex, smoking status, histology, tumor stage, chemotherapy, surgery and principal component.

b

10 missing date were excluded;

c

protective genotypes were KIR3DL2 rs4487030 AG+GG and PVR rs35385129 CA+AA;

*

P values for significant genotypes were in bold.

Abbreviations: SNP: single nucleotide polymorphism; NSCLC: non-small cell lung cancer; PLCO: Prostate, Lung, Colorectal and Ovarian cancer screening trial; OS: overall survival; DSS: disease-specific survival; HR: hazards ratio; CI: confidence interval; NPG: number of protective genotypes.

Combined effects of the two independent SNPs in the PLCO dataset

Consequently, we also utilized the PLCO dataset to assess the combined effect of the two independent SNPs on NSCLC OS and DSS. First, we combined the significant protective genotypes (i.e., KIR3DL2 rs4487030 AG+GG and PVR rs35385129 CA+AA) into a genetic score as the number of protective genotypes (NPGs). As shown in Table 3, the increased genetic score was associated with a better survival in the multivariate analysis of the PLCO dataset (P trend = 0.001 for OS and P trend = 0.003 for DSS). When we dichotomized all the patients into genetic scores of 0-1 and 2 NPGs, the score 2 group had a significantly better survival (HR = 0.72, 95% CI = 0.60-0.86 and P<0.001 for OS and HR = 0.74, 95% CI = 0.61-0.90 and P = 0.003 for DSS), in comparison with the score 0-1 group. We further plotted Kaplan-Meier survival curves to depict these associations of protective genotypes with NSCLC OS (Figure 2A and 2B) and DSS (Figure 2C and 2D).

Figure 2.

Figure 2

Prediction of survival with combined protective genotypes. (A) Kaplan-Meier survival curves for the overall survival of the combined protective genotypes (KIR3DL2 rs4487030 AG+GG and PVR rs35385129 CA+AA) and (B) dichotomized groups of the NUG in the PLCO dataset; (C) Kaplan-Meier survival curves for the disease-specific survival of the combined protective genotypes and (D) dichotomized groups of the NUG in the PLCO dataset. Abbreviations: NPG, number of protective genotypes; PLCO, The Prostate, Lung, Colorectal and Ovarian Cancer Screening Trial.

Stratified analysis for associations between NPGs and NSCLC survival

To evaluate whether the combined effects of protective alleles were modified by other clinical variables, we performed stratified analysis by age, sex, smoking status, histology, tumor stage, chemotherapy, radiotherapy and surgery in the PLCO dataset. As showed in Table S4, we found that the smoke status and chemotherapy had a significant effect on DSS. Patients with two protective genotypes had a better DSS (HR = 0.62, 95% CI = 0.48-0.80 and P<0.001) than those with 0-1 protective genotypes in former-smoke patients (Pinter = 0.045). Meanwhile, patients who received chemotherapy had a better OS (HR = 0.61, 95% CI = 0.46-0.81 and P<0.001) and DSS (HR = 0.63, 95% CI = 0.46-0.86 and P<0.003) than those who did not receive chemotherapy (Pinter = 0.047 and 0.039, respectively).

The ROC curves and time-dependent AUC

We further assessed predictive value of the two SNPs with the time-dependent AUC and ROC curves for the five-year survival in the PLCO dataset. Compared with the model of covariates including age, sex, smoking status, histology, tumor stage, chemotherapy, radiotherapy, surgery and the first four principal components, the time-dependent AUC plot with an additional two independent SNPs did not improve prediction performance of the model for five-year OS and DSS (Figure S4A-D). On the other hand, when we calculated the time-dependent AUC and ROC curves for the 5-year (at the 60th month) survival and the combine two independent SNPs together with the previously published 15 SNPs, the prediction performance of the model for OS and DSS has been significantly improved. The AUCs changed from 88.35% to 89.71% (P = 0.010) for OS and from 88.18% to 89.64% (P = 0.014) for DSS (Figure S4F and S4H).

The eQTL analysis

To assess the correlations between two independent SNPs and their corresponding mRNA expression levels, we firstly performed the eQTL analysis by using the data of 589 whole blood samples and 454 normal lung tissues from the GTEx project. We found that the rs4487030 G allele was significantly associated with a decreased mRNA expression level of KIR3DL2 in normal lung tissues (P = 4.3×10-7) (Figure 3A) and whole blood samples (P = 4.6×10-12) (Figure 3B), while the rs35385129 A allele also associated with an increased mRNA expression level of PVR both in normal lung tissues (P = 7.4×10-6) (Figure 3D) and whole blood samples (P = 7.6×10-5) (Figure 3E).

Figure 3.

Figure 3

Correlations between genotypes of the significant SNPs and their corresponding mRNA expression levels. The KIR3DL2 rs4487030 A allele was associated with higher mRNA expression levels of KIR3DL2 (A) in normal lung tissue and (B) whole blood from the GTEx project; higher expression levels of KIR3DL2 (C) were associated with a worse survival in patients with lung cancer; The PVR rs35385129 A allele was associated with higher mRNA expression levels of PVR (D) in normal lung tissue and (E) whole blood from the GTEx project; higher expression levels of PVR (F) were associated with a better survival in patients with lung cancer (GSE30219 dataset). Abbreviations: KIR3DL2: killer cell immunoglobulin like receptor, three Ig domains and long cytoplasmic tail 2; PVR: PVR cell adhesion molecule.

Additional eQTL analysis was performed for the PVR rs35385129 A allele using the RNA-Seq data of lymphoblastoid cell lines from 373 European descendants available from the 1000 Genomes Project (but no data for the rs4487030 G allele). However, there was no significant correlation between the PVR rs35385129 A allele and mRNA expression levels of PVR in all three genetic models (Figure S7A).

Finally, we performed functional prediction for rs4487030 and rs35385129 as well as other SNPs in high LD, utilizing three different online bioinformatics tools, SNPinfo, RegulomeDB, and HaploReg, to predict their biological functions. In summary, rs35385129 may cause missense mutations, with some substantial functions based on RegulomeDB and HaploReg, particularly in the transcription factor binding or DNase peak. Moreover, rs35385129 is in high LD with other SNPs that may have an effect on other genes and proteins (Table S5). On the other hand, rs4487030 may have an impact on proteins CDP and p300 based on HaploReg without predicted functions based on RegulomeDB. The detailed results are summarized in Figure S6 and Table S5.

Differential mRNA expression analysis and survival of NSCLC

Subsequently, we assessed mRNA expression levels of the two genes identified by the SNPs in 111 pairs of tumor and adjacent normal tissue samples obtained from NSCLC patients in the TCGA database and non-paired tumor and normal tissue samples from the UALCAN database (http://ualcan.path.uab.edu/). For non-paired tumor tissues, as shown in Figure S5, mRNA expression levels of KIR3DL2 in lung adenocarcinoma (LUAD) and lung squamous cell carcinoma (LUSC) were significantly lower than that in adjacent normal tissues (P<0.0001 for both), while the mRNA expression levels of PVR were also significantly lower in tumor tissues in LUSC, but not in LUAD, than adjacent normal tissues (P = 0.005 and P = 0.478, respectively) (Figure S7C). For the paired tumor samples, as shown in Figure S7B, compared with adjacent normal tissues, the mRNA expression levels of PVR were significantly lower in LUSC (P = 0.0001) and LUAD+LUSC (P = 0.0005) than adjacent normal tissues, but not in LUAD alone. Subsequently, we analyzed the correlation between the expression levels of KIR3DL2 and PVR and the survival curve of lung cancer by online KM plotter (http://km-plot.com/analysis). As showed in Figure 3C, a higher expression level of KIR3DL2 was associated with a worse NSCLC survival, but the results of PVR were inconsistent in different datasets of the online KM survival curve plotter (Figure S8). In GSE30219 and GSE19188 datasets, PVR was associated with a better survival but with a poor survival in GSE31210 and All-combined databases (Figure 3F).

Discussion

In the present study, we used the existing genotyping data of two previously published GWASs to analyze the association between genetic variation of immunoregulatory interaction genes and survival of NSCLC. We found and verified two potentially functional and independent SNPs (i.e., KIR3DL2 rs4487030 A>G and PVR rs35385129 C>A) that were significantly associated with survival of NSCLC in populations of European descendants. Our eQTL analysis found that the rs4487030 G and rs35385129 A alleles were significantly correlated with mRNA expression levels of KIR3DL2 and PVR in normal lung tissues and whole blood cells from 369 subjects in the GTEx program, respectively (Figure 3). These results were also consistent with the gene expression analysis of paired tumor and adjacent normal tissue samples and survival analysis in the TCGA database.

KIR3DL2 is a member of a killer Ig-like receptors (KIRs), also known as CD158k, and is expressed as a disulfide bond-linked homodimer. Each chain consists of three immunoglobulin-like domains and a long cytoplasmic tail containing two tyrosine inhibitory motifs of immune receptors. At the genomic level, KIRs are located in the leukocyte receptor cluster of chromosomes 19q13.4, while the Ly49 gene of rodents is located in the NK complex of chromosome 6. Human haplotypes encode the KIR content, and their polymorphic alleles are very different, as many as 14 genes and three frame genes, namely KIR2DL4, KIR3DL2 and KIR3DL3 [32]. KIR3DL2 is expressed not only in NK cells but also in rare circulating T lymphocytes, mainly CD8+ [33]. An enriched KIR3DL2 expression in the memory CD45RO+CD28-CCR7-CD62L-T cells has also been reported. For example, KIR3DL2 is capable, upon the ligation at the surface of NK cells, of inhibiting IFNγ production and cytotoxic function [34], which disrupts the tumor-killing capacity of the immune system.

In the present study, we found that the rs4487030 G allele was significantly associated with lower mRNA expression levels of KIR3DL2 in normal lung tissues and whole blood cells (Figure 3); this finding is consistent with the results from other studies [34], suggesting that the rs4487030 G allele may prolong survival of NSCLC patients through decreasing KIR3DL2 mRNA expression levels. Moreover, mRNA of KIR3DL2 expression levels were lower in the non-paired tumor tissues in both LUSC and LUAD than in normal tissues. The observation of low expression levels of KIR3DL2 associated with a poor survival in NSCLC patients suggested an oncogenic role of the gene. There are two explanations for this observation; one is that the inflammatory reaction and immune killing activity in tumor tissues are more significant than in adjacent normal tissues, so a lower KIR3DL2 expression level is the consequence rather than the cause. On the other hand, normal tissues have a complete immune escape. It is evident that immune evasion in tumor tissues has occurred, likely as a result of low KIR3DL2 expression in tumor microenvironment. According to the ENCODE database, rs4487030 is located in a DNase I hypersensitive site with observable levels of histone modifications in H3K4Me1 acetylation, suggesting that the rs4487030 SNP may lead to altered transcriptional activities of KIR3DL2. Therefore, once our findings are validated by other investigators, the exact molecular mechanisms underlying the observed KIR3DL2 rs4487030 G allele-associated survival warrant additional experimental and mechanistic studies.

PVR was initially identified as a poliovirus receptor, with conserved amino acids and domain structure characteristically similar to the immunoglobulin superfamily [35]. PVR is the fifth member in the nectin-like molecule family, also referred to as CD155 or necl-5. Nectin-like molecule family, which has a domain structure similar to the nectins, plays a crucial role in cell adhesion and polarization, and PVR may have a similar function. However, many studies have investigated the biological role of PVR, but the results were inconsistent. For example, it has been reported that overexpression of PVR can promote tumor cell metastasis [36,37], stimulate cell proliferation, or enhance cell proliferation induced by growth factor [38]. In fact, overexpression of PVR can also reduce contact inhibition by decreasing the expression of nectin-3 [39]. In animal experiments, PVR-/- mice displayed reduced tumor growth and metastasis via DNAM-1 upregulation and enhanced effector function of CD8+ T and NK cells, respectively, while PVR-deleted tumor cells also displayed slower tumor growth and reduced metastases, demonstrating the importance of a tumor-intrinsic role of PVR [40]. On the other hand, PVR seems to play a dual function in onco-immunity and is considered to have the characteristics of a tumor suppressor gene. For example, PVR is the ligand for CD226, and interaction of CD226 with PVR triggers NK or T cell-mediated cytotoxicity, accompanied by an increase in cytokine production [41,42]. Further in vitro studies showed that tumor cells with a higher PVR expression were more susceptible to the CD226-induced killing [43,44]. Accordingly, the balance between PVR/CD226 and PVR/TIGIT or PVR/CD96 maintains normal NK and T cell function. However, this balance may be destroyed in the tumor microenvironment, affecting the progression of the tumor. According to the ENCODE database, rs35385129 is located in both H3K4Me1 and H3K27Ac, suggesting that rs35385129 may lead to an altered transcription, too.

In the present study, we found that the rs35385129 A allele was associated with higher mRNA expression levels of PVR in normal lung tissues and whole blood cells (Figure 3). This is consistent with the results of rs35385129 A allele-associated survival in both PLCO and HLCS datasets. Published studies reported that PVR expression is not detectable in most normal tissues but can be up-regulated in a series of human malignant tumors, including colon cancer, lung adenocarcinoma, melanoma, pancreatic cancer and glioblastoma [45]. This is inconsistent with the mRNA expression results in the TCGA database, including paired and non-paired samples. It is likely that these reported studies may have included different study populations or tumor tissues from those of the TCGA database. Apparently, the function of PVR is still controversial, and the reported effects of PVR on the survival of NSCLC are different as well. However, the findings of the present study provide additional support for a tumor suppressor role of PVR.

Although the associations between SNPs in the immunoregulatory interaction pathway genes had been comprehensively analyzed in the present study, it should also be mentioned that the present study has some limitations. Firstly, both the discovery and validation datasets were from Caucasian populations; thus, our findings may not be generalizable to other ethnic communities. Secondly, we did not have any information on the detailed treatment, particularly for immunotherapies. Additionally, we only analyzed the associations of genetic variants in the identified genes with the survival, and the complicated molecular mechanisms that underly these observed associations should be further explored. Finally, the role of viral infection in tumor tissues and their implications on the survival was not evaluated because the related data were not made available.

In conclusion, two independent SNPs, KIR3DL2 rs4487030 A>G and PVR rs35385129 C>A, were found to be significantly associated with NSCLC survival in both the PLCO trial and the HLCS study. The combined analysis revealed that the protective genotypes of the SNPs were associated with a better OS and DSS in a genotype-dose response manner. Such protective effect on survival is likely through the SNP-associated expression regulation of KIR3DL2 and PVR. Additional studies are needed to substantiate our findings.

Acknowledgements

The authors thank all the participants of the PLCO Cancer Screening Trial. The authors also thank the National Cancer Institute for providing access to the data collected by the PLCO trial. The statements contained herein are solely those of the authors and do not represent or imply concurrence or endorsement by the National Cancer Institute. The authors would also like to acknowledge the dbGaP repository for providing cancer genotyping datasets. The accession numbers for the datasets for lung cancer are phs000336.v1.p1 and phs000093.v2.p2. A list of contributing investigators and funding agencies for those studies can be found in the Supplemental Data. Qingyi Wei was supported by the V Foundation for Cancer Research (D2017-19) and also partly supported by the Duke Cancer Institute as part of the P30 Cancer Center Support Grant (Grant ID: NIH/NCI CA014236). Sheng Luo was supported by NIH (grants R01NS091307, R56AG062302). The Harvard Lung Cancer Susceptibility Study was supported by NIH grants U01CA209414, CA092824, CA074386 and CA090578 to David C. Christiani. We wish to thank all of the investigators and funding agencies that enabled the deposition of data in dbGaP and PLCO that we used in the present study: The datasets used for the analyses described in the present study were obtained from dbGaP at http://www.ncbi.nlm.nih.gov/gap through dbGaP accession number phs000336.v1.p1 and phs000093.v2.p2. Principal Investigators: Maria Teresa Landi. Genetic Epidemiology Branch, Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA. Neil E. Caporaso. Genetic Epidemiology Branch, Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA. Funding support for the GWAS of Lung Cancer and Smoking was provided through the NIH Genes, Environment and Health Initiative [GEI] (Z01CP010200). The human subjects participating in the GWAS derive from The Environment and Genetics in Lung Cancer Etiology (EAGLE) case-control study and the Prostate, Lung Colon and Ovary Screening Trial and these studies are supported by intramural resources of the National Cancer Institute. Assistance with phenotype harmonization and genotype cleaning, as well as with general study coordination, was provided by the Gene Environment Association Studies, GENEVA Coordinating Center (U01HG004446). Assistance with data cleaning was provided by the National Center for Biotechnology Information. Funding support for genotyping, which was performed at the Johns Hopkins University Center for Inherited Disease Research, was provided by the NIH GEI (U01HG004438). PLCO was also supported by the Intramural Research Program of the Division of Cancer Epidemiology and Genetics and by contracts from the Division of Cancer Prevention, National Cancer Institute, NIH, DHHS. The authors thank PLCO screening center investigators and staff, and the staff of Information Management Services Inc. and Westat Inc. Most importantly, we acknowledge trial participants for their contributions that made this study possible.

Disclosure of conflict of interest

None.

Abbreviations

NSCLC

Non-small cell lung cancer

SNPs

single nucleotide polymorphisms

GWAS

Genome-Wide Association Study

PLCO

the Prostate, Lung, Colorectal and Ovarian Cancer Screening Trial

HLCS

Harvard Lung Cancer Susceptibility

OS

overall survival

LD

linkage disequilibrium

FDR

false discovery rate

BFDP

Bayesian false discovery probability

eQTL

expression quantitative trait loci

TCGA

the Cancer Genome Atlas

ROC

receiver operating characteristic

KIR3DL2

killer cell immunoglobulin like receptor, three Ig domains and long cytoplasmic tail 2

PVR

PVR cell adhesion molecule

EAF

effect allele frequency

HR

hazards ratio

CI

confidence interval

AUC

area under the receiver operating characteristic curve

ROC

receiver operating characteristic curve

Supporting Information

ajcr0010-1770-f4.pdf (3.8MB, pdf)

References

  • 1.Bray F, Ferlay J, Soerjomataram I, Siegel RL, Torre LA, Jemal A. Global cancer statistics 2018: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J Clin. 2018;68:394–424. doi: 10.3322/caac.21492. [DOI] [PubMed] [Google Scholar]
  • 2.Blandin Knight S, Crosbie PA, Balata H, Chudziak J, Hussell T, Dive C. Progress and prospects of early detection in lung cancer. Open Biol. 2017;7:170070. doi: 10.1098/rsob.170070. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3.Hirsch FR, Scagliotti GV, Mulshine JL, Kwon R, Curran WJ Jr, Wu YL, Paz-Ares L. Lung cancer: current therapies and new targeted treatments. Lancet. 2017;389:299–311. doi: 10.1016/S0140-6736(16)30958-8. [DOI] [PubMed] [Google Scholar]
  • 4.Topalian SL, Hodi FS, Brahmer JR, Gettinger SN, Smith DC, McDermott DF, Powderly JD, Sosman JA, Atkins MB, Leming PD, Spigel DR, Antonia SJ, Drilon A, Wolchok JD, Carvajal RD, McHenry MB, Hosein F, Harbison CT, Grosso JF, Sznol M. Five-year survival and correlates among patients with advanced melanoma, renal cell carcinoma, or non-small cell lung cancer treated with nivolumab. JAMA Oncol. 2019;5:1411–20. doi: 10.1001/jamaoncol.2019.2187. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5.Garon EB, Hellmann MD, Rizvi NA, Carcereny E, Leighl NB, Ahn MJ, Eder JP, Balmanoukian AS, Aggarwal C, Horn L, Patnaik A, Gubens M, Ramalingam SS, Felip E, Goldman JW, Scalzo C, Jensen E, Kush DA, Hui R. Five-year overall survival for patients with advanced nonsmall-cell lung cancer treated with pembrolizumab: results from the phase I KEYNOTE-001 study. J. Clin. Oncol. 2019;37:2518–2527. doi: 10.1200/JCO.19.00934. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6.Feng Y, Wang Y, Liu H, Liu Z, Mills C, Owzar K, Xie J, Han Y, Qian DC, Hung Rj RJ, Brhane Y, McLaughlin J, Brennan P, Bickeböller H, Rosenberger A, Houlston RS, Caporaso N, Landi MT, Brüske I, Risch A, Ye Y, Wu X, Christiani DC, Amos CI, Wei Q. Novel genetic variants in the P38MAPK pathway gene ZAK and susceptibility to lung cancer. Mol Carcinog. 2018;57:216–224. doi: 10.1002/mc.22748. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7.Wen J, Liu H, Wang L, Wang X, Gu N, Liu Z, Xu T, Gomez DR, Komaki R, Liao Z, Wei Q. Potentially functional variants of ATG16L2 predict radiation pneumonitis and outcomes in patients with non-small cell lung cancer after definitive radiotherapy. J Thorac Oncol. 2018;13:660–675. doi: 10.1016/j.jtho.2018.01.028. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8.Mizukoshi E, Kaneko S. Telomerase-targeted cancer immunotherapy. Int J Mol Sci. 2019;20:1823. doi: 10.3390/ijms20081823. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9.Topalian SL, Weiner GJ, Pardoll DM. Cancer immunotherapy comes of age. J. Clin. Oncol. 2011;29:4828–4836. doi: 10.1200/JCO.2011.38.0899. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10.Sharma P, Allison JP. Immune checkpoint targeting in cancer therapy: toward combination strategies with curative potential. Cell. 2015;161:205–214. doi: 10.1016/j.cell.2015.03.030. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11.Reck M, Rodriguez-Abreu D, Robinson AG, Hui R, Csoszi T, Fulop A, Gottfried M, Peled N, Tafreshi A, Cuffe S, O’Brien M, Rao S, Hotta K, Leiby MA, Lubiniecki GM, Shentu Y, Rangwala R, Brahmer JR KEYNOTE-024 Investigators. Pembrolizumab versus chemotherapy for PD-L1-positive non-small-cell lung cancer. N Engl J Med. 2016;375:1823–1833. doi: 10.1056/NEJMoa1606774. [DOI] [PubMed] [Google Scholar]
  • 12.Gandhi L, Rodriguez-Abreu D, Gadgeel S, Esteban E, Felip E, De Angelis F, Domine M, Clingan P, Hochmair MJ, Powell SF, Cheng SY, Bischoff HG, Peled N, Grossi F, Jennens RR, Reck M, Hui R, Garon EB, Boyer M, Rubio-Viqueira B, Novello S, Kurata T, Gray JE, Vida J, Wei Z, Yang J, Raftopoulos H, Pietanza MC, Garassino MC KEYNOTE-189 Investigators. Pembrolizumab plus chemotherapy in metastatic non-small-cell lung cancer. N Engl J Med. 2018;378:2078–2092. doi: 10.1056/NEJMoa1801005. [DOI] [PubMed] [Google Scholar]
  • 13.Hellmann MD, Paz-Ares L, Bernabe Caro R, Zurawski B, Kim SW, Carcereny Costa E, Park K, Alexandru A, Lupinacci L, de la Mora Jimenez E, Sakai H, Albert I, Vergnenegre A, Peters S, Syrigos K, Barlesi F, Reck M, Borghaei H, Brahmer JR, O’Byrne KJ, Geese WJ, Bhagavatheeswaran P, Rabindran SK, Kasinathan RS, Nathan FE, Ramalingam SS. Nivolumab plus ipilimumab in advanced non-small-cell lung cancer. N Engl J Med. 2019;381:2020–2031. doi: 10.1056/NEJMoa1910231. [DOI] [PubMed] [Google Scholar]
  • 14.Socinski MA, Jotte RM, Cappuzzo F, Orlandi F, Stroyakovskiy D, Nogami N, Rodriguez-Abreu D, Moro-Sibilot D, Thomas CA, Barlesi F, Finley G, Kelsch C, Lee A, Coleman S, Deng Y, Shen Y, Kowanetz M, Lopez-Chavez A, Sandler A, Reck M IMpower150 Study Group. Atezolizumab for first-line treatment of metastatic nonsquamous NSCLC. N Engl J Med. 2018;378:2288–2301. doi: 10.1056/NEJMoa1716948. [DOI] [PubMed] [Google Scholar]
  • 15.Wang RF, Wang HY. Immune targets and neoantigens for cancer immunotherapy and precision medicine. Cell Res. 2017;27:11–37. doi: 10.1038/cr.2016.155. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16.Gallagher MD, Chen-Plotkin AS. The post-GWAS era: from association to function. Am J Hum Genet. 2018;102:717–730. doi: 10.1016/j.ajhg.2018.04.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17.Hocking WG, Hu P, Oken MM, Winslow SD, Kvale PA, Prorok PC, Ragard LR, Commins J, Lynch DA, Andriole GL, Buys SS, Fouad MN, Fuhrman CR, Isaacs C, Yokochi LA, Riley TL, Pinsky PF, Gohagan JK, Berg CD PLCO Project Team. Lung cancer screening in the randomized Prostate, Lung, Colorectal, and Ovarian (PLCO) Cancer Screening Trial. J Natl Cancer Inst. 2010;102:722–731. doi: 10.1093/jnci/djq126. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18.Oken MM, Marcus PM, Hu P, Beck TM, Hocking W, Kvale PA, Cordes J, Riley TL, Winslow SD, Peace S, Levin DL, Prorok PC, Gohagan JK PLCO Project Team. Baseline chest radiograph for lung cancer detection in the randomized Prostate, Lung, Colorectal and Ovarian Cancer Screening Trial. J Natl Cancer Inst. 2005;97:1832–1839. doi: 10.1093/jnci/dji430. [DOI] [PubMed] [Google Scholar]
  • 19.Mailman MD, Feolo M, Jin Y, Kimura M, Tryka K, Bagoutdinov R, Hao L, Kiang A, Paschall J, Phan L, Popova N, Pretel S, Ziyabari L, Lee M, Shao Y, Wang ZY, Sirotkin K, Ward M, Kholodov M, Zbicz K, Beck J, Kimelman M, Shevelev S, Preuss D, Yaschenko E, Graeff A, Ostell J, Sherry ST. The NCBI dbGaP database of genotypes and phenotypes. Nat Genet. 2007;39:1181–1186. doi: 10.1038/ng1007-1181. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20.Tryka KA, Hao L, Sturcke A, Jin Y, Wang ZY, Ziyabari L, Lee M, Popova N, Sharopova N, Kimura M, Feolo M. NCBI’s database of genotypes and phenotypes: dbGaP. Nucleic Acids Res. 2014;42:D975–979. doi: 10.1093/nar/gkt1211. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21.Zhai R, Yu X, Wei Y, Su L, Christiani DC. Smoking and smoking cessation in relation to the development of co-existing non-small cell lung cancer with chronic obstructive pulmonary disease. Int J Cancer. 2014;134:961–970. doi: 10.1002/ijc.28414. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22.Aulchenko YS, Ripke S, Isaacs A, van Duijn CM. GenABEL: an R library for genome-wide association analysis. Bioinformatics. 2007;23:1294–1296. doi: 10.1093/bioinformatics/btm108. [DOI] [PubMed] [Google Scholar]
  • 23.Park JH, Geum DI, Eisenhut M, van der Vliet HJ, Shin JI. Bayesian statistical methods in genetic association studies: empirical examination of statistically non-significant Genome Wide Association Study (GWAS) meta-analyses in cancers: a systematic review. Gene. 2019;685:170–178. doi: 10.1016/j.gene.2018.10.057. [DOI] [PubMed] [Google Scholar]
  • 24.Wakefield J. A Bayesian measure of the probability of false discovery in genetic epidemiology studies. Am J Hum Genet. 2007;81:208–227. doi: 10.1086/519024. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25.Chambless LE, Diao G. Estimation of time-dependent area under the ROC curve for long-term risk prediction. Stat Med. 2006;25:3474–3486. doi: 10.1002/sim.2299. [DOI] [PubMed] [Google Scholar]
  • 26.Consortium GT. Human genomics. The Genotype-Tissue Expression (GTEx) pilot analysis: multitissue gene regulation in humans. Science. 2015;348:648–660. doi: 10.1126/science.1262110. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27.Lappalainen T, Sammeth M, Friedlander MR, t Hoen PA, Monlong J, Rivas MA, Gonzalez-Porta M, Kurbatova N, Griebel T, Ferreira PG, Barann M, Wieland T, Greger L, van Iterson M, Almlof J, Ribeca P, Pulyakhina I, Esser D, Giger T, Tikhonov A, Sultan M, Bertier G, MacArthur DG, Lek M, Lizano E, Buermans HP, Padioleau I, Schwarzmayr T, Karlberg O, Ongen H, Kilpinen H, Beltran S, Gut M, Kahlem K, Amstislavskiy V, Stegle O, Pirinen M, Montgomery SB, Donnelly P, McCarthy MI, Flicek P, Strom TM, Geuvadis C, Lehrach H, Schreiber S, Sudbrak R, Carracedo A, Antonarakis SE, Hasler R, Syvanen AC, van Ommen GJ, Brazma A, Meitinger T, Rosenstiel P, Guigo R, Gut IG, Estivill X, Dermitzakis ET. Transcriptome and genome sequencing uncovers functional variation in humans. Nature. 2013;501:506–511. doi: 10.1038/nature12531. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28.Xu Z, Taylor JA. SNPinfo: integrating GWAS and candidate gene information into functional SNP selection for genetic association studies. Nucleic Acids Res. 2009;37:W600–605. doi: 10.1093/nar/gkp290. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29.Boyle AP, Hong EL, Hariharan M, Cheng Y, Schaub MA, Kasowski M, Karczewski KJ, Park J, Hitz BC, Weng S, Cherry JM, Snyder M. Annotation of functional variation in personal genomes using RegulomeDB. Genome Res. 2012;22:1790–1797. doi: 10.1101/gr.137323.112. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30.Ward LD, Kellis M. HaploReg v4: systematic mining of putative causal variants, cell types, regulators and target genes for human complex traits and disease. Nucleic Acids Res. 2016;44:D877–881. doi: 10.1093/nar/gkv1340. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31.Wang Y, Liu H, Ready NE, Su L, Wei Y, Christiani DC, Wei Q. Genetic variants in ABCG1 are associated with survival of nonsmall-cell lung cancer patients. Int J Cancer. 2016;138:2592–2601. doi: 10.1002/ijc.29991. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32.Wende H, Colonna M, Ziegler A, Volz A. Organization of the leukocyte receptor cluster (LRC) on human chromosome 19q13.4. Mamm Genome. 1999;10:154–160. doi: 10.1007/s003359900961. [DOI] [PubMed] [Google Scholar]
  • 33.Chan AT, Kollnberger SD, Wedderburn LR, Bowness P. Expansion and enhanced survival of natural killer cells expressing the killer immunoglobulin-like receptor KIR3DL2 in spondylarthritis. Arthritis Rheum. 2005;52:3586–3595. doi: 10.1002/art.21395. [DOI] [PubMed] [Google Scholar]
  • 34.Schmitt C, Marie-Cardine A, Bensussan A. Therapeutic antibodies to KIR3DL2 and other target antigens on cutaneous T-cell lymphomas. Front Immunol. 2017;8:1010. doi: 10.3389/fimmu.2017.01010. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35.Gao J, Zheng Q, Xin N, Wang W, Zhao C. CD155, an onco-immunologic molecule in human tumors. Cancer Sci. 2017;108:1934–1938. doi: 10.1111/cas.13324. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36.Ikeda W, Kakunaga S, Itoh S, Shingai T, Takekuni K, Satoh K, Inoue Y, Hamaguchi A, Morimoto K, Takeuchi M, Imai T, Takai Y. Tage4/Nectin-like molecule-5 heterophilically trans-interacts with cell adhesion molecule Nectin-3 and enhances cell migration. J Biol Chem. 2003;278:28167–28172. doi: 10.1074/jbc.M303586200. [DOI] [PubMed] [Google Scholar]
  • 37.Ikeda W, Kakunaga S, Takekuni K, Shingai T, Satoh K, Morimoto K, Takeuchi M, Imai T, Takai Y. Nectin-like molecule-5/Tage4 enhances cell migration in an integrin-dependent, Nectin-3-independent manner. J Biol Chem. 2004;279:18015–18025. doi: 10.1074/jbc.M312969200. [DOI] [PubMed] [Google Scholar]
  • 38.Kakunaga S, Ikeda W, Shingai T, Fujito T, Yamada A, Minami Y, Imai T, Takai Y. Enhancement of serum- and platelet-derived growth factor-induced cell proliferation by Necl-5/Tage4/poliovirus receptor/CD155 through the Ras-Raf-MEK-ERK signaling. J Biol Chem. 2004;279:36419–36425. doi: 10.1074/jbc.M406340200. [DOI] [PubMed] [Google Scholar]
  • 39.Minami Y, Ikeda W, Kajita M, Fujito T, Monden M, Takai Y. Involvement of up-regulated Necl-5/Tage4/PVR/CD155 in the loss of contact inhibition in transformed NIH3T3 cells. Biochem Biophys Res Commun. 2007;352:856–860. doi: 10.1016/j.bbrc.2006.11.089. [DOI] [PubMed] [Google Scholar]
  • 40.Li XY, Das I, Lepletier A, Addala V, Bald T, Stannard K, Barkauskas D, Liu J, Aguilera AR, Takeda K, Braun M, Nakamura K, Jacquelin S, Lane SW, Teng MW, Dougall WC, Smyth MJ. CD155 loss enhances tumor suppression via combined host and tumor-intrinsic mechanisms. J Clin Invest. 2018;128:2613–2625. doi: 10.1172/JCI98769. [DOI] [PMC free article] [PubMed] [Google Scholar] [Retracted]
  • 41.Bottino C, Castriconi R, Pende D, Rivera P, Nanni M, Carnemolla B, Cantoni C, Grassi J, Marcenaro S, Reymond N, Vitale M, Moretta L, Lopez M, Moretta A. Identification of PVR (CD155) and Nectin-2 (CD112) as cell surface ligands for the human DNAM-1 (CD226) activating molecule. J Exp Med. 2003;198:557–567. doi: 10.1084/jem.20030788. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42.Tahara-Hanaoka S, Shibuya K, Kai H, Miyamoto A, Morikawa Y, Ohkochi N, Honda S, Shibuya A. Tumor rejection by the poliovirus receptor family ligands of the DNAM-1 (CD226) receptor. Blood. 2006;107:1491–1496. doi: 10.1182/blood-2005-04-1684. [DOI] [PubMed] [Google Scholar]
  • 43.Castriconi R, Dondero A, Corrias MV, Lanino E, Pende D, Moretta L, Bottino C, Moretta A. Natural killer cell-mediated killing of freshly isolated neuroblastoma cells: critical role of DNAX accessory molecule-1-poliovirus receptor interaction. Cancer Res. 2004;64:9180–9184. doi: 10.1158/0008-5472.CAN-04-2682. [DOI] [PubMed] [Google Scholar]
  • 44.Pende D, Spaggiari GM, Marcenaro S, Martini S, Rivera P, Capobianco A, Falco M, Lanino E, Pierri I, Zambello R, Bacigalupo A, Mingari MC, Moretta A, Moretta L. Analysis of the receptor-ligand interactions in the natural killer-mediated lysis of freshly isolated myeloid or lymphoblastic leukemias: evidence for the involvement of the Poliovirus receptor (CD155) and Nectin-2 (CD112) Blood. 2005;105:2066–2073. doi: 10.1182/blood-2004-09-3548. [DOI] [PubMed] [Google Scholar]
  • 45.Bevelacqua V, Bevelacqua Y, Candido S, Skarmoutsou E, Amoroso A, Guarneri C, Strazzanti A, Gangemi P, Mazzarino MC, D’Amico F, McCubrey JA, Libra M, Malaponte G. Nectin like-5 overexpression correlates with the malignant phenotype in cutaneous melanoma. Oncotarget. 2012;3:882–892. doi: 10.18632/oncotarget.594. [DOI] [PMC free article] [PubMed] [Google Scholar]

Associated Data

This section collects any data citations, data availability statements, or supplementary materials included in this article.

Supplementary Materials

ajcr0010-1770-f4.pdf (3.8MB, pdf)

Data Availability Statement

The datasets are available from the National Center for Biotechnology Information Database of Genotypes and Phenotypes (dbGaP Study Accession: phs000093.v2.p2 and phs000336.v1.p1). Genome-wide imputation was performed based on the 1000 Genomes Project, phase III CEU, utilizing the IMPUTE2 software (October 2014 release).


Articles from American Journal of Cancer Research are provided here courtesy of e-Century Publishing Corporation

RESOURCES