Skip to main content
Erschienen in: Journal of Translational Medicine 1/2019

Open Access 01.12.2019 | Research

Identification and validation of an immune cell infiltrating score predicting survival in patients with lung adenocarcinoma

verfasst von: Xiaodong Yang, Yu Shi, Ming Li, Tao Lu, Junjie Xi, Zongwu Lin, Wei Jiang, Weigang Guo, Cheng Zhan, Qun Wang

Erschienen in: Journal of Translational Medicine | Ausgabe 1/2019

Abstract

Background

Immune infiltration may predict survival and have clinical significance in lung cancer. However, immune signatures derived from immune profiling based on bulk tumor transcriptomes have not been systematically established in lung adenocarcinoma. We aimed to construct an immune cell infiltrating score, using a new algorithm for evaluating immune infiltration, to improve the prognostic model of lung adenocarcinoma.

Methods

Public datasets of lung adenocarcinoma from the Gene Expression Omnibus and The Cancer Genome Atlas were adopted as the training and validation cohorts. Fractions of different immune cell subtypes in each sample were estimated using the CIBERSORT algorithm. The immune infiltrating score was further developed by a least absolute shrinkage and selection operator regression model. The prognostic value and clinical relationship of the model was then further explored.

Results

An immune infiltrating score model was established on the basis of the immune cells in the training cohort. A high score was associated with significantly worse survival in patients with lung adenocarcinoma (P < 0.001). The prognostic value of the score was confirmed in the validation cohort. The immune infiltrating score could improve the accuracy of predictions of survival when combined with the staging system. Furthermore, the score was potentially associated with patient smoking status and histologic subtype of lung adenocarcinoma. Its possible association with the efficacy of adjuvant chemotherapy was not statistically significant.

Conclusion

The immune cell infiltrating score has prognostic significance in predicting overall survival in patients with lung adenocarcinoma.
Begleitmaterial
Hinweise

Electronic supplementary material

The online version of this article (https://​doi.​org/​10.​1186/​s12967-019-1964-6) contains supplementary material, which is available to authorized users.
Xiaodong Yang and Yu Shi contributed equally to this work

Publisher's Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Background

Lung cancer remains the leading cause of cancer incidence and death worldwide in 2018, when it was estimated to comprise 2.1 million new cases and cause 1.8 million deaths [1]. Among all patients with lung cancer, lung adenocarcinoma accounts for the largest proportion, and its incidence continues to increase [2]. The literature has shown that phenotypes of cancers are dependent on the tumor microenvironment, especially tumor-infiltrating immune cells [3, 4]. The immune response is characterized by many types of cells, and differences in their potential prognostic value depend on the cancer type [5]. Computational approaches were adopted to characterize immune-interactions in lung adenocarcinoma, mainly focused on B cell and CD8+ T cell [6]. Meta-analysis also showed that CD8+, CD3+ and FOXP3+ T cells infiltration had significantly prognostic value in lung adenocarcinoma [7]. Furthermore, the prognostic significance of integrated immune infiltration in some cancers has been validated, and this has been proposed to supplement the tumor, node, and metastasis (TNM) staging system for predicting patient survival [813]. However, very few studies reported integrated immune infiltration in lung adenocarcinoma.
Most previous studies have adopted an immunohistochemistry method to evaluate the landscape of tumor-infiltrating immune cell subtypes. However, a bioinformatics tool, known as the CIBERSORT algorithm, has been previously developed for computational enumeration of immune cell subtypes. The landscape and proportions of tumor-infiltrating immune cells are evaluated on the basis of the changes in expression of immune-relative and other genes in one sample. This meta-gene approach has been well designed and validated in previous studies [9, 1416].
In the present study, CIBERSORT was used to estimate the fraction of immune cell types based on different lung adenocarcinoma gene expression profiles. Least absolute shrinkage and selection operator (LASSO) Cox regression analysis was utilized to establish an immune cell infiltrating score model in both training and validation cohorts. We believe that the immune infiltrating score could assist in predicting survival, along with clinical factors, in lung adenocarcinoma.

Methods

Study cohort and data processing

We adopted the public datasets from the Gene Expression Omnibus (GEO) (https://​www.​ncbi.​nlm.​nih.​gov/​geo/​) as the training cohort. Four GEO datasets (GSE30219, GSE37745, GSE50081 and GSE68465, the latter also known as the Director’s Challenge Consortium) were included in this study. All of the four datasets were derived from the genechips of Affymetrix® (Santa Clara, California, USA). GSE30219, GSE37745 and GSE50081 were based on the GPL570 genechip, while GSE68465 were from the GPL96 microarray. Then, raw data and genechip files were downloaded directly. A total of 85, 106, 127 and 443 samples of lung adenocarcinoma from GSE30219, GSE37745, GSE50081 and GSE68465 datasets were enrolled. We used a robust multichip average method via RMAExpress for background adjustment, quantile normalization and summary to process the gene profiles [1719].
Level 3 RNA sequence data from lung adenocarcinoma samples were also downloaded from The Cancer Genome Atlas (TCGA) before June 5, 2018 (https://​portal.​gdc.​cancer.​gov/​). A total of 594 samples were obtained, comprising 535 adenocarcinoma and 59 normal lung samples. Baseline clinicopathological factors and treatment information were also acquired from TCGA. Primary lung adenocarcinoma samples with complete follow-up and baseline information (age, sex and pathological stage) were included as the validation cohort in this study. Due to the original records of all included datasets, the seventh edition of the TNM staging method was utilized in our study. Information regarding histologic subtypes of lung adenocarcinoma was intergraded with records from the University of California Santa Cruz Xena database (http://​xena.​ucsc.​edu/​) and previous literature [2022]. According to previous studies, tumors were classified into three subgroups (low-, intermediate- and high-grade adenocarcinoma) regarding the survival data for each predominant lung adenocarcinoma subtype [23, 24].

Estimation of tumor-infiltrating immune cell types

The CIBERSORT method was adopted to quantify the proportions of the immune cell in both training and validation cohorts [25, 26]. Common methods, like flow cytometry and immunohistochemistry, rely on few phenotype markers and tissue disaggregation prior to cytometry, may cause damaged or lost cells, altering analyses [26]. The CIBERSORT was developed to accurately evaluate the relative levels of the 22 immune cell phenotypes, especially closely related types, using signature gens within complex expression mixtures [26]. Such mixtures could derive from both patients’ solid tissues and blood profiled by array or RNA-sequencing [26]. The 22 immune cells are mainly composed of B cells, T cells, macrophages, dendritic cells, plasma cells, natural killer cells and mast cells. To process data by the CIBERSORT, the targeted gene profile was uploaded to its website (https://​cibersort.​stanford.​edu). Then, a heat-map table with levels of 22 immune cell subtypes returned as the final results. As previously described, we set the threshold P-value < 0.05 and excluded samples without immune infiltration in training (5 patients) and validation (78 patients) cohorts [25]. The optimal cut-off values of the proportions of different immune cells in the training cohort were calculated on the basis of the prognostic significance using X-Tile software [27]. Each immune cell fraction level of samples from both cohorts was then divided into two groups based on the cut-off point. The immune cell fraction level divided by the cut-off value was valued as 0 or 1 in the subsequent scoring formula.

Statistical analysis

All statistical analyses in this study were performed using R version 3.5.0 (R Foundation for Statistical Computing, Vienna, Austria) and IBM SPSS Statistics 22.0 (IBM, Inc., Armonk, NY, USA). We used the LASSO Cox regression model to select the ideal prognostic model among the estimated immune cell subtypes using the glmnet package in R [28]. The optimal model parameter λ and corresponding coefficients were determined by tenfold cross validations. We chose λ via 1 − SE (standard error) criteria as previously recommended [8, 9]. Based on the final immune infiltrating score model and corresponding coefficients, a formula for the score was constructed using the training cohort. The sensitivity and specificity of the prognosis prediction of the immune cell infiltrating score were evaluated by a time-dependent receiver operating characteristic (ROC) curve in R software. The nearest neighbor estimation method in the ROC curve was adopted with cut-off times of 1, 3 and 5 years. Survival curves were estimated using the Kaplan–Meier method and the log-rank test was utilized for comparing survival curves. Univariate and multivariate Cox proportional hazard models were adopted to identify all significant prognostic factors. The prognostic value of the included variables was evaluated by Harrell’s concordance index (c-index) [29]. The score stratified by subgroups were compared using the one-way ANOVA method, if normal distribution and homogeneity of variance were followed. Correlation analysis was also conducted to explore the potential relationship between the score and the expression of immune checkpoint regulators. In this study, a two-tailed P-value of < 0.05 was considered statistically significant.

Results

Construction of the immune cell infiltrating score model

After the initial selection process, 751 and 418 patients were enrolled as the training and validation cohorts, respectively. The characteristics of the patients are listed in Table 1 and Additional file 1: Table S1. Optimal cut-off values for the fraction of each immune cell type in the training cohort were generated as described in “Methods” section (Additional file 2: Table S2). LASSO Cox regression analysis was performed, based on the fraction of immune cells in the training cohort (Fig. 1a, b). Finally, the formula of the immune cell infiltrating score for lung adenocarcinoma in the training cohort was built (Additional file 3). The prognostic accuracy of the scoring model was evaluated using time-dependent ROC analysis in the training cohort. The predictive time points were set at 1, 3 and 5 years, respectively (Fig. 2a). The area under the ROC curve for the immune infiltrating score at each time point was 0.674, 0.684 and 0.675, respectively.
Table 1
Baseline characteristics of patients with lung adenocarcinoma in the training cohort
Variable
Baseline characteristics (n = 751)
Affymetrix® platform
 HG-U133 Plus 2.0 (GPL570)
313 (41.7)
 HG-U133A (GPL96)
438 (58.3)
Age
64.6 ± 10.0
Sex
 Male
394 (52.5)
 Female
357 (47.5)
Tumor stage
 Stage I
515 (68.6)
 Stage II–III
232 (30.9)
 Stage IV
4 (0.5)
Smoking status
 Current smoker
67 (8.9)
 Ex-smoker
322 (42.9)
 Non-smoker
71 (9.5)
 Unknown
291 (38.7)
Adjuvant chemotherapy
 Yes
102 (13.6)
 No
377 (50.2)
 Unknown
272 (36.2)
Continuous data (age) was presented as mean ± standard deviation and categorical data as number (proportion of that the subgroup accounted for the whole group)
Survival analyses were performed stratified by the score, in the training cohort (Fig. 2b). Patients were separated into low- and high-score groups by the median score of the cohort. We observed that patients with a high score had significantly worse survival than those with a low score (P < 0.001, Fig. 2b). A high score was also associated with significantly shorter progression-free survival (P < 0.001, Additional file 4: Figure S1).
As a continuous variable, the immune cell infiltrating score was found to have significantly prognostic value using univariate Cox regression analysis (P < 0.001, Table 2). Multivariate analysis confirmed that the immune infiltrating score was an independent prognostic factor in the training cohort, along with the TNM staging system (P < 0.001, Table 2). According to the corresponding c-index, the combination of the scoring model and TNM stage showed a significantly better predictive value than the TNM stage alone in this cohort (P < 0.001, Table 3).
Table 2
Results of univariate and multivariate Cox regression of overall survival in the training cohort
Cohort
Training cohort
Variable
Univariate
Multivariate
HR
95% CI
P-value
HR
95% CI
P-value
Immune infiltrating score
6.459
4.409–9.462
< 0.001
5.322
3.574–7.926
< 0.001
Age
1.025
1.014–1.035
< 0.001
1.024
1.014–1.035
< 0.001
Sex
  
0.031
  
0.079
 Female
Reference
  
Reference
  
 Male
1.245
1.021–1.518
0.031
1.196
0.979–1.460
0.079
TNM stage
  
< 0.001
  
< 0.001
 Stage I
Reference
  
Reference
  
 Stage II–III
2.682
2.195–3.278
< 0.001
2.443
1.996–2.990
< 0.001
 Stage IV
2.256
0.721–7.053
0.162
1.835
0.586–5.740
0.297
HR hazard ratio, CI confidence interval
Table 3
Comparison of the accuracy of survival prediction between the TNM stage with and without the immune infiltrating score
Cohort
TNM stage
Immune infiltrating score + TNM stage
P-value
c-index
95% CI
c-index
95% CI
Training cohort
0.619
0.594–0.643
0.686
0.659–0.714
< 0.001
Validation cohort
0.661
0.621–0.700
0.695
0.650–0.740
< 0.001
CI confidence interval

Validation of the immune cell infiltrating score model

We adopted a validation group based on the dataset from TCGA to evaluate the prognostic value of the proposed scoring model. The same formula for the immune cell infiltrating score and optimal cut-off point for each immune cells were applied to the validation group. Likewise, ROC analysis was adopted to assess the prognostic value of the scoring model. The area under the curve was 0.650, 0.616 and 0.567 at the predictive time of 1, 3 and 5 years, respectively (Fig. 2c). In the validation group, a high score was associated with significantly worse prognosis (P = 0.013, Fig. 2d).
Consistent with the previous findings, the score was an independent prognostic factor in the validation cohort, based on the univariate and multivariate Cox regression model (P = 0.001 and 0.002, Additional file 5: Table S3). Furthermore, the combination of the scoring model and TNM stage improved the prognostic model of lung adenocarcinoma in this cohort (P < 0.001, Table 3).

Clinical significance and bioinformatics analyses based on the immune cell infiltrating score

In the training group, data regarding adjuvant chemotherapy were documented only in the GSE68465 and GSE37745 datasets. Patients were divided into low and high score groups based on the score median value. Given the differences in staging methods and initial study design, patients with stage I disease were excluded from this analysis. The survival advantage of the low-score group was observed in patients with or without adjuvant chemotherapy (P = 0.004 and 0.002, Additional file 6: Figure S2A, B). Moreover, there was a trend for better prognosis in patients with a low score who received adjuvant chemotherapy, although the difference was not statistically significant (P = 0.909, Fig. 3a). No significant prognostic difference regarding adjuvant chemotherapy was observed in high score group (P = 0.764, Fig. 3b). We also extracted data on adjuvant chemotherapy from the validation group. There was a trend for better survival in low score group who received chemotherapy (P = 0.084, Additional file 6: Figure S2C). No difference was found in patients without chemotherapy (P = 0.761, Additional file 6: Figure S2D). Furthermore, patients with a low score may benefit from adjuvant chemotherapy (P = 0.010, Fig. 3c), while no difference was observed in high-score group (P = 0.213, Fig. 3d).
Smoking status was included in two datasets (GSE50081 and GSE68465) for the training cohort. Patients with unknown smoking status were excluded. We observed that patients who are current smokers had significantly higher score than those who do not smoke (P < 0.001, Fig. 4a). In the validation cohort, patients who are current or reformed smokers (< 15 years) had higher scores than non-smokers or reformed smokers (> 15 years), although the difference was not statistically significant (P = 0.932, Fig. 4b).
Furthermore, we explored potential relationships between the immune cell infiltrating score and histologic subtypes of lung adenocarcinoma. In the validation cohort, 186 patients with information on histologic subtypes were included. We found that high-grade adenocarcinoma subtypes were associated with significantly higher score than low-grade tumors (P = 0.003, micropapillary/solid vs acinar/papillary vs lepidic predominant lung adenocarcinoma, Fig. 4c).
Owing to the difference of the microarray platforms used for the training cohort, correlation analyses between the immune infiltrating score and the expression of immune checkpoint regulators or inflammatory mediators were performed in the validation cohort. We found that there was a significant positive correlation between the score and some of the regulators and mediators, including PD-L1 (P = 0.002), IDO1 (P = 0.003) and LAG3 (P = 0.017), IFNB1 (P = 0.002), IL-1A (P = 0.005), TNFA (P = 0.010) and IL-6 (P = 0.007) (Additional file 7: Table S4).

Discussion

In this study, we adopted the CIBERSORT method to calculate the fraction of 22 immune cell in lung adenocarcinoma. This newly developed algorithm can work accurately for bulk tumor samples profiled by microarray or RNA-Sequencing. And unlike flow or mass cytometry-based methods, it is applicable to archived RNA and cellular specimens [26]. Previous studies have validated the efficacy of the CIBERSORT method [1416]. On the basis of the estimated fractions of signature immune cells, LASSO regression was utilized to construct an immune cell infiltrating score model, of which the predictive accuracy has been demonstrated previously [14, 28, 30, 31]. In both cohorts, the prognostic value of the scoring model was confirmed. A lower score was associated with significantly better prognosis, which may be due to more immunity-activating lymphocyte infiltrations. Further analyses also implied that the score could significantly improve the accuracy of survival prediction when combined with the TNM staging system. Significant correlation between the score and the expression of common immune checkpoint regulators or inflammatory mediators was also observed, including PD-L1, IDO1 and LAG3, which further confirmed its potential values. Previous studies also indicated that the expression of PD-L1 and LAG-3 was related with early recurrence and poor prognosis [32, 33]. In a word, we hope that our work will provide new insights into the construction of staging or prognostic models in lung adenocarcinoma.
The role of different tumor-infiltrating lymphocytes in lung adenocarcinoma has been explored separately [7, 34]. Kinoshita et al. [35] found that CD8+ T-cells accumulation was identified as a negative prognostic factor in lung adenocarcinoma, but not in lung squamous carcinoma. Comprehensive immune profiling of lung adenocarcinoma revealed plasma cell infiltration was related to worse prognosis [36]. Mast cell exosomes were also reported to promote lung adenocarcinoma cell proliferation [37]. The function of mast cells against or in favor of the cancers still requires investigations [38, 39]. Previous studies also observed that the prognostic implication differs according to histological types and smoking status [35, 40]. The potential role of tumor-infiltrating lymphocytes may require exploration comprehensively according to immune microenvironment. The functionality of CD8+ tumor-infiltrating lymphocytes was reported to be affected by competition between antitumor and exhaustion programs [41]. Choi and Na investigated the relationship between the immune landscape and tumor glucose metabolism in lung adenocarcinoma [42]. Varn et al. adopted computational approaches to characterize tumor-immune interactions, mainly focused on six immune cell subtypes [6]. In our study, we adopted the CIBERSORT method with 22 cell phenotypes in lung adenocarcinoma like the study of Gentles et al. [5]. The final formula of the immune cell infiltrating score was composed of 13 types of immune cells using LASSO regression, which may better characterize the potential internal interactions and microenvironment.
Several studies have focused on the relationships between tumor-infiltrating lymphocytes and the efficacy of adjuvant chemotherapy [8, 4345]. In this study, the potential associations with chemotherapy were not all statistically significant. The weak effect may be due to small sample size in this part of analyses and the difficulty in finding the optimal cut-off value for the score. Previous studies showed that the chemotherapy sensitivity may depend on the lymphocytes infiltrating the tumor [8, 9]. One potential mechanism is that the interferon secreted by lymphocytes could sensitize cells to chemotherapy [46]. The innate and adaptive immune responses may also be activated by immunogenic tumor cell death [47]. More investigations may be required for the underlying mechanisms [48]. Our study also indicated that the score was significantly associated with patient smoking status and histologic subtype, which may help us to better characterize patient subgroups and choose the optimal treatment.
The strength of this study is the large cohort derived from several institutions, which outnumbered than most similar researches of lung adenocarcinoma. We adopted a newly developed CIBERSORT method to estimate the level of tumor-infiltrating lymphocytes, which outperformed other methods (like immunohistochemistry) regarding noise, unknown mixture content and closely related cell types. There are also some limitations that should be noted. First, the public datasets in this study were based on different genechips, but we adopted the RMAExpress method which designed for Affymetrix® microarrays and merged the raw data. All the datasets were collected with different study purposes, which may lead to heterogeneities in baseline features and therapies. Also, only retrospective analyses were made. Future prospective studies will be needed to confirm the findings. Second, some clinicopathologic factors were missing or incomplete, especially histological subtype, smoking status and adjuvant chemotherapy. Further investigations should collect more clinical factors and endpoints, along with possible alternative methods for testing and validation, which may provide more evidence for lung adenocarcinoma [13]. In addition, optimal cut-off value and practical approaches are essential for future clinical application.

Conclusion

An immune cell infiltrating score model was established based on the immune cells in lung adenocarcinoma. A high score was associated with significantly worse survival. The scoring model could improve the accuracy of predictions of survival when combined with the staging system. It was also shown that the score was closely associated with the smoking status, histologic subtype and the expression of some immune checkpoint regulators or inflammatory mediators. This study may provide new insights into the construction of staging or prognostic models in lung adenocarcinoma. The close associations between the immune infiltrating score and clinical factors may help us to better identify patient subgroups and choose the optimal treatment for patients with lung adenocarcinoma.

Acknowledgements

We would like to thank International Science Editing Co. for the language editing service.
Appropriate Ethics Committees of Zhongshan Hospital approved this study to be exempt research.
Not applicable.

Competing interests

The authors declare that they have no competing interests.
Open AccessThis article is distributed under the terms of the Creative Commons Attribution 4.0 International License (http://​creativecommons.​org/​licenses/​by/​4.​0/​), which permits unrestricted use, distribution, and reproduction in any medium, provided you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons license, and indicate if changes were made. The Creative Commons Public Domain Dedication waiver (http://​creativecommons.​org/​publicdomain/​zero/​1.​0/​) applies to the data made available in this article, unless otherwise stated.

Publisher's Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Anhänge

Additional files

Literatur
1.
Zurück zum Zitat Bray F, Ferlay J, Soerjomataram I, et al. Global cancer statistics 2018: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J Clin. 2018;68(6):394–424.CrossRef Bray F, Ferlay J, Soerjomataram I, et al. Global cancer statistics 2018: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J Clin. 2018;68(6):394–424.CrossRef
2.
Zurück zum Zitat Meza R, Meernik C, Jeon J, Cote ML. Lung cancer incidence trends by gender, race and histology in the United States, 1973–2010. PLoS ONE. 2015;10(3):e0121323.CrossRef Meza R, Meernik C, Jeon J, Cote ML. Lung cancer incidence trends by gender, race and histology in the United States, 1973–2010. PLoS ONE. 2015;10(3):e0121323.CrossRef
3.
Zurück zum Zitat Mantovani A, Allavena P, Sica A, Balkwill F. Cancer-related inflammation. Nature. 2008;454(7203):436–44.CrossRef Mantovani A, Allavena P, Sica A, Balkwill F. Cancer-related inflammation. Nature. 2008;454(7203):436–44.CrossRef
4.
Zurück zum Zitat Swann JB, Smyth MJ. Immune surveillance of tumors. J Clin Invest. 2007;117(5):1137–46.CrossRef Swann JB, Smyth MJ. Immune surveillance of tumors. J Clin Invest. 2007;117(5):1137–46.CrossRef
5.
Zurück zum Zitat Gentles AJ, Newman AM, Liu CL, et al. The prognostic landscape of genes and infiltrating immune cells across human cancers. Nat Med. 2015;21(8):938–45.CrossRef Gentles AJ, Newman AM, Liu CL, et al. The prognostic landscape of genes and infiltrating immune cells across human cancers. Nat Med. 2015;21(8):938–45.CrossRef
6.
Zurück zum Zitat Varn FS, Tafe LJ, Amos CI, Cheng C. Computational immune profiling in lung adenocarcinoma reveals reproducible prognostic associations with implications for immunotherapy. Oncoimmunology. 2018;7(6):e1431084.CrossRef Varn FS, Tafe LJ, Amos CI, Cheng C. Computational immune profiling in lung adenocarcinoma reveals reproducible prognostic associations with implications for immunotherapy. Oncoimmunology. 2018;7(6):e1431084.CrossRef
7.
Zurück zum Zitat Geng Y, Shao Y, He W, et al. Prognostic role of tumor-infiltrating lymphocytes in lung cancer: a meta-analysis. Cell Physiol Biochem. 2015;37(4):1560–71.CrossRef Geng Y, Shao Y, He W, et al. Prognostic role of tumor-infiltrating lymphocytes in lung cancer: a meta-analysis. Cell Physiol Biochem. 2015;37(4):1560–71.CrossRef
8.
Zurück zum Zitat Jiang Y, Zhang Q, Hu Y, et al. ImmunoScore signature: a prognostic and predictive tool in gastric cancer. Ann Surg. 2018;267(3):504–13.CrossRef Jiang Y, Zhang Q, Hu Y, et al. ImmunoScore signature: a prognostic and predictive tool in gastric cancer. Ann Surg. 2018;267(3):504–13.CrossRef
9.
Zurück zum Zitat Zeng D, Zhou R, Yu Y, et al. Gene expression profiles for a prognostic immunoscore in gastric cancer. Br J Surg. 2018;105(10):1338–48.CrossRef Zeng D, Zhou R, Yu Y, et al. Gene expression profiles for a prognostic immunoscore in gastric cancer. Br J Surg. 2018;105(10):1338–48.CrossRef
10.
Zurück zum Zitat Xiong Y, Wang K, Zhou H, et al. Profiles of immune infiltration in colorectal cancer and their clinical significant: a gene expression-based study. Cancer Med. 2018;7(9):4496–508.CrossRef Xiong Y, Wang K, Zhou H, et al. Profiles of immune infiltration in colorectal cancer and their clinical significant: a gene expression-based study. Cancer Med. 2018;7(9):4496–508.CrossRef
11.
Zurück zum Zitat Zhang XM, Song LJ, Shen J, et al. Prognostic and predictive values of immune infiltrate in patients with head and neck squamous cell carcinoma. Hum Pathol. 2018;82:104–12.CrossRef Zhang XM, Song LJ, Shen J, et al. Prognostic and predictive values of immune infiltrate in patients with head and neck squamous cell carcinoma. Hum Pathol. 2018;82:104–12.CrossRef
12.
Zurück zum Zitat Duan J, Xie Y, Qu L, et al. A nomogram-based immunoprofile predicts overall survival for previously untreated patients with esophageal squamous cell carcinoma after esophagectomy. J Immunother Cancer. 2018;6(1):100.CrossRef Duan J, Xie Y, Qu L, et al. A nomogram-based immunoprofile predicts overall survival for previously untreated patients with esophageal squamous cell carcinoma after esophagectomy. J Immunother Cancer. 2018;6(1):100.CrossRef
13.
Zurück zum Zitat Pages F, Mlecnik B, Marliot F, et al. International validation of the consensus Immunoscore for the classification of colon cancer: a prognostic and accuracy study. Lancet. 2018;391(10135):2128–39.CrossRef Pages F, Mlecnik B, Marliot F, et al. International validation of the consensus Immunoscore for the classification of colon cancer: a prognostic and accuracy study. Lancet. 2018;391(10135):2128–39.CrossRef
14.
Zurück zum Zitat Fu H, Zhu Y, Wang Y, et al. Identification and validation of stromal immunotype predict survival and benefit from adjuvant chemotherapy in patients with muscle-invasive bladder cancer. Clin Cancer Res. 2018;24(13):3069–78.CrossRef Fu H, Zhu Y, Wang Y, et al. Identification and validation of stromal immunotype predict survival and benefit from adjuvant chemotherapy in patients with muscle-invasive bladder cancer. Clin Cancer Res. 2018;24(13):3069–78.CrossRef
15.
Zurück zum Zitat Mao Y, Feng Q, Zheng P, et al. Low tumor infiltrating mast cell density confers prognostic benefit and reflects immunoactivation in colorectal cancer. Int J Cancer. 2018;143(9):2271–80.CrossRef Mao Y, Feng Q, Zheng P, et al. Low tumor infiltrating mast cell density confers prognostic benefit and reflects immunoactivation in colorectal cancer. Int J Cancer. 2018;143(9):2271–80.CrossRef
16.
Zurück zum Zitat Zhou L, Xu L, Chen L, et al. Tumor-infiltrating neutrophils predict benefit from adjuvant chemotherapy in patients with muscle invasive bladder cancer. OncoImmunology. 2017;6(4):e1293211.CrossRef Zhou L, Xu L, Chen L, et al. Tumor-infiltrating neutrophils predict benefit from adjuvant chemotherapy in patients with muscle invasive bladder cancer. OncoImmunology. 2017;6(4):e1293211.CrossRef
17.
Zurück zum Zitat Bolstad BM, Irizarry RA, Astrand M, Speed TP. A comparison of normalization methods for high density oligonucleotide array data based on variance and bias. Bioinformatics. 2003;19(2):185–93.CrossRef Bolstad BM, Irizarry RA, Astrand M, Speed TP. A comparison of normalization methods for high density oligonucleotide array data based on variance and bias. Bioinformatics. 2003;19(2):185–93.CrossRef
18.
Zurück zum Zitat Irizarry RA, Bolstad BM, Collin F, et al. Summaries of Affymetrix GeneChip probe level data. Nucleic Acids Res. 2003;31(4):e15.CrossRef Irizarry RA, Bolstad BM, Collin F, et al. Summaries of Affymetrix GeneChip probe level data. Nucleic Acids Res. 2003;31(4):e15.CrossRef
19.
Zurück zum Zitat Irizarry RA, Hobbs B, Collin F, et al. Exploration, normalization, and summaries of high density oligonucleotide array probe level data. Biostatistics. 2003;4(2):249–64.CrossRef Irizarry RA, Hobbs B, Collin F, et al. Exploration, normalization, and summaries of high density oligonucleotide array probe level data. Biostatistics. 2003;4(2):249–64.CrossRef
20.
Zurück zum Zitat Cancer Genome Atlas Research Network. Comprehensive molecular profiling of lung adenocarcinoma. Nature. 2014;511(7511):543–50.CrossRef Cancer Genome Atlas Research Network. Comprehensive molecular profiling of lung adenocarcinoma. Nature. 2014;511(7511):543–50.CrossRef
21.
Zurück zum Zitat Travis WD, Brambilla E, Noguchi M, et al. International association for the study of lung cancer/American Thoracic Society/European Respiratory Society international multidisciplinary classification of lung adenocarcinoma. J Thorac Oncol. 2011;6(2):244–85.CrossRef Travis WD, Brambilla E, Noguchi M, et al. International association for the study of lung cancer/American Thoracic Society/European Respiratory Society international multidisciplinary classification of lung adenocarcinoma. J Thorac Oncol. 2011;6(2):244–85.CrossRef
22.
Zurück zum Zitat Travis WD, Brambilla E, Nicholson AG, et al. The 2015 World Health Organization classification of lung tumors: impact of genetic, clinical and radiologic advances since the 2004 classification. J Thorac Oncol. 2015;10(9):1243–60.CrossRef Travis WD, Brambilla E, Nicholson AG, et al. The 2015 World Health Organization classification of lung tumors: impact of genetic, clinical and radiologic advances since the 2004 classification. J Thorac Oncol. 2015;10(9):1243–60.CrossRef
23.
Zurück zum Zitat Lee HY, Cha MJ, Lee KS, et al. Prognosis in resected invasive mucinous adenocarcinomas of the lung: related factors and comparison with resected nonmucinous adenocarcinomas. J Thorac Oncol. 2016;11(7):1064–73.CrossRef Lee HY, Cha MJ, Lee KS, et al. Prognosis in resected invasive mucinous adenocarcinomas of the lung: related factors and comparison with resected nonmucinous adenocarcinomas. J Thorac Oncol. 2016;11(7):1064–73.CrossRef
24.
Zurück zum Zitat Russell PA, Wainer Z, Wright GM, et al. Does lung adenocarcinoma subtype predict patient survival?: a clinicopathologic study based on the new International Association for the Study of Lung Cancer/American Thoracic Society/European Respiratory Society international multidisciplinary lung adenocarcinoma classification. J Thorac Oncol. 2011;6(9):1496–504.CrossRef Russell PA, Wainer Z, Wright GM, et al. Does lung adenocarcinoma subtype predict patient survival?: a clinicopathologic study based on the new International Association for the Study of Lung Cancer/American Thoracic Society/European Respiratory Society international multidisciplinary lung adenocarcinoma classification. J Thorac Oncol. 2011;6(9):1496–504.CrossRef
25.
Zurück zum Zitat Ali HR, Chlon L, Pharoah PD, et al. Patterns of immune infiltration in breast cancer and their clinical implications: a gene-expression-based retrospective study. PLoS Med. 2016;13(12):e1002194.CrossRef Ali HR, Chlon L, Pharoah PD, et al. Patterns of immune infiltration in breast cancer and their clinical implications: a gene-expression-based retrospective study. PLoS Med. 2016;13(12):e1002194.CrossRef
26.
Zurück zum Zitat Newman AM, Liu CL, Green MR, et al. Robust enumeration of cell subsets from tissue expression profiles. Nat Methods. 2015;12(5):453–7.CrossRef Newman AM, Liu CL, Green MR, et al. Robust enumeration of cell subsets from tissue expression profiles. Nat Methods. 2015;12(5):453–7.CrossRef
27.
Zurück zum Zitat Camp RL, Dolled-Filhart M, Rimm DL. X-tile: a new bio-informatics tool for biomarker assessment and outcome-based cut-point optimization. Clin Cancer Res. 2004;10(21):7252–9.CrossRef Camp RL, Dolled-Filhart M, Rimm DL. X-tile: a new bio-informatics tool for biomarker assessment and outcome-based cut-point optimization. Clin Cancer Res. 2004;10(21):7252–9.CrossRef
28.
Zurück zum Zitat Tibshirani R. The lasso method for variable selection in the Cox model. Stat Med. 1997;16(4):385–95.CrossRef Tibshirani R. The lasso method for variable selection in the Cox model. Stat Med. 1997;16(4):385–95.CrossRef
29.
Zurück zum Zitat Harrell FJ, Califf RM, Pryor DB, et al. Evaluating the yield of medical tests. JAMA. 1982;247(18):2543–6.CrossRef Harrell FJ, Califf RM, Pryor DB, et al. Evaluating the yield of medical tests. JAMA. 1982;247(18):2543–6.CrossRef
30.
Zurück zum Zitat Qu L, Wang Z, Chen Q, et al. Prognostic value of a long non-coding RNA signature in localized clear cell renal cell carcinoma. Eur Urol. 2018;74(6):756–63.CrossRef Qu L, Wang Z, Chen Q, et al. Prognostic value of a long non-coding RNA signature in localized clear cell renal cell carcinoma. Eur Urol. 2018;74(6):756–63.CrossRef
31.
Zurück zum Zitat Dai W, Feng Y, Mo S, et al. Transcriptome profiling reveals an integrated mRNA–lncRNA signature with predictive value of early relapse in colon cancer. Carcinogenesis. 2018;39(10):1235–44.CrossRef Dai W, Feng Y, Mo S, et al. Transcriptome profiling reveals an integrated mRNA–lncRNA signature with predictive value of early relapse in colon cancer. Carcinogenesis. 2018;39(10):1235–44.CrossRef
32.
Zurück zum Zitat He Y, Yu H, Rozeboom L, et al. LAG-3 protein expression in non-small cell lung cancer and its relationship with PD-1/PD-L1 and tumor-infiltrating lymphocytes. J Thorac Oncol. 2017;12(5):814–23.CrossRef He Y, Yu H, Rozeboom L, et al. LAG-3 protein expression in non-small cell lung cancer and its relationship with PD-1/PD-L1 and tumor-infiltrating lymphocytes. J Thorac Oncol. 2017;12(5):814–23.CrossRef
33.
Zurück zum Zitat Zhang ML, Kem M, Mooradian MJ, et al. Differential expression of PD-L1 and IDO1 in association with the immune microenvironment in resected lung adenocarcinomas. Mod Pathol. 2019;32(4):511–23.CrossRef Zhang ML, Kem M, Mooradian MJ, et al. Differential expression of PD-L1 and IDO1 in association with the immune microenvironment in resected lung adenocarcinomas. Mod Pathol. 2019;32(4):511–23.CrossRef
34.
Zurück zum Zitat Bremnes RM, Busund LT, Kilvaer TL, et al. The role of tumor-infiltrating lymphocytes in development, progression, and prognosis of non-small cell lung cancer. J Thorac Oncol. 2016;11(6):789–800.CrossRef Bremnes RM, Busund LT, Kilvaer TL, et al. The role of tumor-infiltrating lymphocytes in development, progression, and prognosis of non-small cell lung cancer. J Thorac Oncol. 2016;11(6):789–800.CrossRef
35.
Zurück zum Zitat Kinoshita T, Kudo-Saito C, Muramatsu R, et al. Determination of poor prognostic immune features of tumour microenvironment in non-smoking patients with lung adenocarcinoma. Eur J Cancer. 2017;86:15–27.CrossRef Kinoshita T, Kudo-Saito C, Muramatsu R, et al. Determination of poor prognostic immune features of tumour microenvironment in non-smoking patients with lung adenocarcinoma. Eur J Cancer. 2017;86:15–27.CrossRef
36.
Zurück zum Zitat Kurebayashi Y, Emoto K, Hayashi Y, et al. Comprehensive immune profiling of lung adenocarcinomas reveals four immunosubtypes with plasma cell subtype a negative indicator. Cancer Immunol Res. 2016;4(3):234–47.CrossRef Kurebayashi Y, Emoto K, Hayashi Y, et al. Comprehensive immune profiling of lung adenocarcinomas reveals four immunosubtypes with plasma cell subtype a negative indicator. Cancer Immunol Res. 2016;4(3):234–47.CrossRef
37.
Zurück zum Zitat Xiao H, Lasser C, Shelke GV, et al. Mast cell exosomes promote lung adenocarcinoma cell proliferation—role of KIT-stem cell factor signaling. Cell Commun Signal. 2014;12:64.PubMedPubMedCentral Xiao H, Lasser C, Shelke GV, et al. Mast cell exosomes promote lung adenocarcinoma cell proliferation—role of KIT-stem cell factor signaling. Cell Commun Signal. 2014;12:64.PubMedPubMedCentral
38.
Zurück zum Zitat Derakhshani A, Vahidian F, Alihasanzadeh M, et al. Mast cells: a double-edged sword in cancer. Immunol Lett. 2019;209:28–35.CrossRef Derakhshani A, Vahidian F, Alihasanzadeh M, et al. Mast cells: a double-edged sword in cancer. Immunol Lett. 2019;209:28–35.CrossRef
39.
Zurück zum Zitat Reddy SM, Reuben A, Barua S, et al. Poor response to neoadjuvant chemotherapy correlates with mast cell infiltration in inflammatory breast cancer. Cancer Immunol Res. 2019;7(6):1025–35.CrossRef Reddy SM, Reuben A, Barua S, et al. Poor response to neoadjuvant chemotherapy correlates with mast cell infiltration in inflammatory breast cancer. Cancer Immunol Res. 2019;7(6):1025–35.CrossRef
40.
Zurück zum Zitat Kinoshita T, Muramatsu R, Fujita T, et al. Prognostic value of tumor-infiltrating lymphocytes differs depending on histological type and smoking habit in completely resected non-small-cell lung cancer. Ann Oncol. 2016;27(11):2117–23.CrossRef Kinoshita T, Muramatsu R, Fujita T, et al. Prognostic value of tumor-infiltrating lymphocytes differs depending on histological type and smoking habit in completely resected non-small-cell lung cancer. Ann Oncol. 2016;27(11):2117–23.CrossRef
41.
Zurück zum Zitat O’Brien SM, Klampatsa A, Thompson JC, et al. Function of human tumor-infiltrating lymphocytes in early-stage non-small cell lung cancer. Cancer Immunol Res. 2019;7(6):896–909.CrossRef O’Brien SM, Klampatsa A, Thompson JC, et al. Function of human tumor-infiltrating lymphocytes in early-stage non-small cell lung cancer. Cancer Immunol Res. 2019;7(6):896–909.CrossRef
42.
Zurück zum Zitat Choi H, Na KJ. Integrative analysis of imaging and transcriptomic data of the immune landscape associated with tumor metabolism in lung adenocarcinoma: clinical and prognostic implications. Theranostics. 2018;8(7):1956–65.CrossRef Choi H, Na KJ. Integrative analysis of imaging and transcriptomic data of the immune landscape associated with tumor metabolism in lung adenocarcinoma: clinical and prognostic implications. Theranostics. 2018;8(7):1956–65.CrossRef
43.
Zurück zum Zitat Jang N, Kwon HJ, Park MH, et al. Prognostic value of tumor-infiltrating lymphocyte density assessed using a standardized method based on molecular subtypes and adjuvant chemotherapy in invasive breast cancer. Ann Surg Oncol. 2018;25(4):937–46.CrossRef Jang N, Kwon HJ, Park MH, et al. Prognostic value of tumor-infiltrating lymphocyte density assessed using a standardized method based on molecular subtypes and adjuvant chemotherapy in invasive breast cancer. Ann Surg Oncol. 2018;25(4):937–46.CrossRef
44.
Zurück zum Zitat Zhang H, Liu H, Shen Z, et al. Tumor-infiltrating neutrophils is prognostic and predictive for postoperative adjuvant chemotherapy benefit in patients with gastric cancer. Ann Surg. 2018;267(2):311–8.CrossRef Zhang H, Liu H, Shen Z, et al. Tumor-infiltrating neutrophils is prognostic and predictive for postoperative adjuvant chemotherapy benefit in patients with gastric cancer. Ann Surg. 2018;267(2):311–8.CrossRef
45.
Zurück zum Zitat Ignatiadis M, Van den Eynden G, Roberto S, et al. Tumor-infiltrating lymphocytes in patients receiving trastuzumab/pertuzumab-based chemotherapy: a TRYPHAENA substudy. J Natl Cancer Inst. 2019;111(1):69–77.CrossRef Ignatiadis M, Van den Eynden G, Roberto S, et al. Tumor-infiltrating lymphocytes in patients receiving trastuzumab/pertuzumab-based chemotherapy: a TRYPHAENA substudy. J Natl Cancer Inst. 2019;111(1):69–77.CrossRef
46.
Zurück zum Zitat Galluzzi L, Buqué A, Kepp O, et al. Immunological effects of conventional chemotherapy and targeted anticancer agents. Cancer Cell. 2015;28(6):690–714.CrossRef Galluzzi L, Buqué A, Kepp O, et al. Immunological effects of conventional chemotherapy and targeted anticancer agents. Cancer Cell. 2015;28(6):690–714.CrossRef
47.
Zurück zum Zitat Ghiringhelli F, Apetoh L. The interplay between the immune system and chemotherapy: emerging methods for optimizing therapy. Expert Rev Clin Immunol. 2014;10(1):19–30.CrossRef Ghiringhelli F, Apetoh L. The interplay between the immune system and chemotherapy: emerging methods for optimizing therapy. Expert Rev Clin Immunol. 2014;10(1):19–30.CrossRef
48.
Zurück zum Zitat Zitvogel L, Apetoh L, Ghiringhelli F, et al. The anticancer immune response: indispensable for therapeutic success? J Clin Invest. 2008;118(6):1991–2001.CrossRef Zitvogel L, Apetoh L, Ghiringhelli F, et al. The anticancer immune response: indispensable for therapeutic success? J Clin Invest. 2008;118(6):1991–2001.CrossRef
Metadaten
Titel
Identification and validation of an immune cell infiltrating score predicting survival in patients with lung adenocarcinoma
verfasst von
Xiaodong Yang
Yu Shi
Ming Li
Tao Lu
Junjie Xi
Zongwu Lin
Wei Jiang
Weigang Guo
Cheng Zhan
Qun Wang
Publikationsdatum
01.12.2019
Verlag
BioMed Central
Erschienen in
Journal of Translational Medicine / Ausgabe 1/2019
Elektronische ISSN: 1479-5876
DOI
https://doi.org/10.1186/s12967-019-1964-6

Weitere Artikel der Ausgabe 1/2019

Journal of Translational Medicine 1/2019 Zur Ausgabe

Leitlinien kompakt für die Innere Medizin

Mit medbee Pocketcards sicher entscheiden.

Seit 2022 gehört die medbee GmbH zum Springer Medizin Verlag

Echinokokkose medikamentös behandeln oder operieren?

06.05.2024 DCK 2024 Kongressbericht

Die Therapie von Echinokokkosen sollte immer in spezialisierten Zentren erfolgen. Eine symptomlose Echinokokkose kann – egal ob von Hunde- oder Fuchsbandwurm ausgelöst – konservativ erfolgen. Wenn eine Op. nötig ist, kann es sinnvoll sein, vorher Zysten zu leeren und zu desinfizieren. 

Umsetzung der POMGAT-Leitlinie läuft

03.05.2024 DCK 2024 Kongressbericht

Seit November 2023 gibt es evidenzbasierte Empfehlungen zum perioperativen Management bei gastrointestinalen Tumoren (POMGAT) auf S3-Niveau. Vieles wird schon entsprechend der Empfehlungen durchgeführt. Wo es im Alltag noch hapert, zeigt eine Umfrage in einem Klinikverbund.

Proximale Humerusfraktur: Auch 100-Jährige operieren?

01.05.2024 DCK 2024 Kongressbericht

Mit dem demographischen Wandel versorgt auch die Chirurgie immer mehr betagte Menschen. Von Entwicklungen wie Fast-Track können auch ältere Menschen profitieren und bei proximaler Humerusfraktur können selbst manche 100-Jährige noch sicher operiert werden.

Die „Zehn Gebote“ des Endokarditis-Managements

30.04.2024 Endokarditis Leitlinie kompakt

Worauf kommt es beim Management von Personen mit infektiöser Endokarditis an? Eine Kardiologin und ein Kardiologe fassen die zehn wichtigsten Punkte der neuen ESC-Leitlinie zusammen.

Update Innere Medizin

Bestellen Sie unseren Fach-Newsletter und bleiben Sie gut informiert.