Skip to main content
Erschienen in: Journal of Translational Medicine 1/2021

Open Access 01.12.2021 | Research

Immune and stromal scoring system associated with tumor microenvironment and prognosis: a gene-based multi-cancer analysis

verfasst von: Zihang Zeng, Jiali Li, Jianguo Zhang, Yangyi Li, Xingyu Liu, Jiarui Chen, Zhengrong Huang, Qiuji Wu, Yan Gong, Conghua Xie

Erschienen in: Journal of Translational Medicine | Ausgabe 1/2021

insite
INHALT
download
DOWNLOAD
print
DRUCKEN
insite
SUCHEN

Abstract

Background

Tumor microenvironment (TME) is associated with tumor progression and prognosis. Previous studies provided tools to estimate immune and stromal cell infiltration in TME. However, there is still a lack of single index to reflect both immune and stromal status associated with prognosis and immunotherapy responses.

Methods

A novel immune and stromal scoring system named ISTMEscore was developed. A total of 15 datasets were used to train and validate this system, containing 2965 samples from lung adenocarcinoma, skin cutaneous melanoma and head and neck squamous cell carcinoma.

Results

The patients with high immune and low stromal scores (HL) were associated with low ratio of T cell co-inhibitory/stimulatory molecules and low levels of angiogenesis markers, while the patients with low immune and high stromal scores (LH) had the opposite characteristics. The HL patients had immune-centered networks, while the patients with low immune and low stromal scores (LL) had desert-like networks. Moreover, copy number alteration burden was decreased in the HL patients. For the clinical characteristics, our TME classification was an independent prognostic factor. In the 5 cohorts with immunotherapy, the LH patients were linked to the lowest response rate.

Conclusions

ISTMEscore system could reflect the TME status and predict the prognosis. Compared to previous TME scores, our ISTMEscore was superior in the prediction of prognosis and immunotherapy response.
Begleitmaterial
Hinweise

Supplementary Information

The online version contains supplementary material available at https://​doi.​org/​10.​1186/​s12967-021-03002-1.

Publisher's Note

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

Background

With the discovery of immune checkpoint molecules, immunotherapy becomes more promising strategy for cancer patients to elicit clinical responses durably [1]. It has been approved for the treatment of lung adenocarcinoma (LUAD), skin cutaneous melanoma (SKCM), and head and neck squamous cell carcinoma (HNSC). These tumors have high PD-L1 and CD8A expression levels [2]. Nevertheless, immunotherapy only benefits a minor subset of patients for long-term survival [3]. Identification of potentially therapeutic indexes linked to tumor prognosis and immunotherapy responses will remarkedly contributed to precision medicine.
Tumor microenvironment (TME) consists of immune and non-immune stromal components, both of which were reported to be closely associated with oncogenesis and malignant behaviors of tumors [4]. The existing evidences demonstrate that abundant immune components in TME are positively associated with immunotherapy responses [5]. To date, a range of algorithms have been developed to estimate the immune and stromal cell infiltration including CIBERSORT, TIMER, ESTIMATE and MCPcounter [69]. These tools perform well in the estimation of TME cells, but not in the prediction of tumor prognosis and immunotherapy responses. Although ESTIMATE provides the immune and stromal infiltration scores [8], LUAD, SKCM and HNSC were not included in the training datasets. There is still a lack of single index to reflect both immune and stromal activation signals associated with prognosis and immunotherapy responses.
Here we developed a novel immune and stromal scoring system named ISTMEscore, which followed a unique design: (1) Isolate TME signals associated with prognosis from bulk gene expression data; (2) Extract specific gene signatures from the above TME signals; (3) Calculate ISTME scores with single-sample gene set enrichment analysis (ssGSEA) algorithm [10]. In addition, we collected 15 datasets with 2965 patients to train and validate our ISTMEscore system, and depicted the landscapes of immune and stromal cell infiltration, transcriptome, genome, prognosis and immunotherapy responses in patients with different ISTME scores. Finally, we compared ISTMEscore with previous TME indexes on prediction of TME status and cancer prognosis.

Methods

Data collection and processing

Transcriptomic cohorts

We collected 15 transcriptomic cohorts for LUAD, SKCM and HNSC with clinical annotations from The Cancer Genome Atlas (TCGA) [11], Gene Expression Omnibus (GEO) [1217] and PubMed [5]. Their basic information was presented in Additional file 9: Table S1. Samples without prognostic information were excluded. The normalized data of 3 TCGA cohorts (TCGA LUAD, TCGA SKCM and TCGA HNSC) were downloaded from Firehose RSEM files [18]. The microarray data from GEO were processed with RMA standardization, except that NanoString nCounter data were processed using Housekeeper genes [19]. All transcriptomic data were normalized by Z-score and min–max for further analysis [20]. R sva package was used for batch effect correction [21].

Mutation and copy number alteration data

The TCGA somatic mutations were called by Mutect2 [22]. Only non-silent mutations were included in this study. Copy number alteration (CNA) of the TCGA cohorts was detected by Affymetrix SNP 6.0 array and GISTIC2 after germline subtraction [23].

Immunotherapy cohorts

A total of 5 independent transcriptomic cohorts containing patients with immune checkpoint inhibitor (ICI) treatment were used to validate the ISTMEscore system [5, 2427]. The detailed information of these immunotherapy cohorts was presented in Additional file 10: Table S2. The immunotherapy dataset 1 [5] included 28 melanoma patients before and after anti-PD-1/CTLA4 therapies. The immunotherapy dataset 2 (GSE91061) [24] included 65 melanoma patients before and after anti-PD-1 therapies. The immunotherapy dataset 3 (GSE93157) [25] included 35 NSCLC, 5 HNSC and 25 melanoma patients before anti-PD-1 therapy. The immunotherapy dataset 4 (GSE67501) [26] included 11 renal cell carcinoma patients before anti-PD-1 therapy. The immunotherapy dataset 5 (GSE35640) [27] included 56 melanoma patients before anti-MAGE-A3 therapy.

Development of novel immune and stromal scores

Step 1: Extraction of low dimensional feature associated with TME signals via non-negative matrix factorization

Non-negative matrix factorization (NMF) was an unsupervised algorithm for low dimensional feature extraction, which was performed by R NMF package based on brunet methods [28]. The specific formula was as follows:
$$\mathrm{V}{\in {R}_{+}}^{i*j};\mathrm{w}{\in {R}_{+}}^{i*k};\mathrm{H}{\in {R}_{+}}^{k*j}$$
$${\mathrm{V}}_{i*j}\approx {\mathrm{W}}_{i*k}*{\mathrm{H}}_{k*j}$$
where \({\mathrm{V}}_{i*j}\) is gene expression matrix with i gene and j sample, \({\mathrm{W}}_{i*k}\) represents basis matrix with i gene and k the low dimensional features (LDF). \({\mathrm{H}}_{k*j}\) is the coefficient matrix considered as a low dimensional matrix of \({\mathrm{V}}_{i*j}\). The k means the number of NMF clusters. The j sample and i gene can be divided into the k clusters, respectively:
$${\mathrm{sample}\_\mathrm{cluster}}_{j}=k, \quad when\; {\mathrm{H}}_{k*j}=Max({\mathrm{H}}_{j})$$
$${\mathrm{gene}\_\mathrm{cluster}}_{i}=k, \quad when\; {\mathrm{W}}_{i*k}=Max({\mathrm{W}}_{i})$$
The number of NMF clusters (k) was determined by hierarchical clustering of gene expression matrix. Normally, \(\mathrm{k}*\left(\mathrm{i}+\mathrm{j}\right)<i*j\), thus high dimensional genes in transcriptome data (i gene) were transformed into low dimensional eigenvalues (k feature).
In this study, we first defined k as 11 through hierarchical clustering of \({\mathrm{V}}_{i*j}\) from the training datasets. Next, enrichment analysis was performed based on genes with high NMF weights (top 100) to identify clusters associated with immune and stromal signals. In addition, we extracted eigenvalues (\({\mathrm{H}}_{k}\)) from each cluster by NMF, and used univariate Cox regression to identify prognosis-related clusters. Finally, we identified one immune- (\({k}_{immu}\)-th) and one stromal-related clusters (\({k}_{stro}\)-th), both of which were associated with immune/stromal signals and the overall survival (OS).

Step 2: Identification of TME-related signatures with the ℓ2,1-norm multitask learning linear model

Multitask learning (MTL) was an ensemble approach, which can train multiple tasks at the same time. By introducing ℓ2,1 regularization term into cost function, the output of regression coefficient matrix was sparse. The cost function of the MTL model was based on least squares loss:
$$\sum_{t=1}^{m}||{W}_{t}^{T}{X}_{t}-{Y}_{t}|{|}_{F}^{2}+{\rho }_{1}{||W||}_{\mathrm{2,1}}+{\rho }_{L2}{|\left|W\right||}_{F}^{2}$$
$${||W||}_{\mathrm{2,1}}=\sum_{i=1}^{n}\sqrt{\sum_{j=1}^{m}{W}_{i,j}^{2}}$$
$${|\left|W\right||}_{F}=\sqrt{\sum_{i=1}^{n}\sum_{j=1}^{m}{W}_{i,j}^{2}}$$
where \({W}_{t}\) denotes the coefficient matrix of multi-factor linear regression model in the task t, \({X}_{t}\) is the gene expression matrix (\({\mathrm{V}}_{i*j}\) in NMF) of the task t, \({Y}_{t}\) is sample label (immune or stromal scores), \({||W||}_{\mathrm{2,1}}\) denotes ℓ2,1-norm term, and \({|\left|W\right||}_{F}\) denotes Frobenius-norm term. The accelerated gradient methods were used to minimize the cost function. Compared to Lasso or Ridge, the ℓ2,1-norm regularization resulted in grouped sparsity across sub-tasks. Thus, we selected the small subset of genes with the effective information in the input \({\mathrm{V}}_{i*j}\) matrix. The MTL code was based on MATLAB from the previous studies [29].
In this study, there were 2 sub-tasks (t = 2), and \({Y}_{t}\) was defined as follow:
$${Y}_{t}=\left[{Y}_{1},{Y}_{2}\right]=\left[{H}_{{k}_{immu}},{H}_{{k}_{stro}}\right]$$
where \({H}_{{k}_{immu}}\) is \({k}_{immu}\)-th column of coefficient matrix of NMF, and \({H}_{{k}_{stro}}\) is \({k}_{stro}\)-th column of coefficient matrix of NMF. Moreover, addition of ℓ2,1 regularization term contributed to avoid impact of multicollinearity among genes. Through ℓ2,1-norm MTL, we identified the genes associated with TME-related LDF (criterion: the coefficient of each gene in coefficient matrix \({W}_{t}\) \(\ne \) 0).

Step 3: Optimization of the gene list through different gene expression analysis and consensus clustering

Consensus clustering (number of clusters = 2) for MTL genes was performed to divide the TCGA training datasets into 2 clusters through NMFConsensus (GenePattern module) [30]. Different gene expression (DGE) analysis was then used to identify differentially expressed genes in these 2 consensus clusters by R limma package [31]. Benjamini-Hochberg (BH) method was used for multiple hypothesis adjustment of DGE [32]. To verify whether the consensus clustering was related to TME, enrichment analysis of the genes with top 1,000 positive and top 1,000 negative logFC was performed. According to results of enrichment analysis, we defined the 108 genes from MTL list with P < 0.05 and logFC > 0 as the immune-related genes and the 58 genes with P < 0.05 and logFC < 0 as the stromal-related genes. DGE had 2 effects: (1) Remove the genes not significantly expressed in the consensus clusters; (2) Distinguish immune- and stromal-related genes by the direction of logFC.

Step 4: Quantification of novel immune and stromal scores through ssGSEA

The ssGSEA was used to construct novel immune and stromal scores based on immune- and stromal-related genes using the R GSVA package [10]. Compare with the generalized linear model, the normalized ssGSEA score was the enrichment score based on gene rank, and was therefore not sensitive to different platforms (microarray or RNA-seq) or incomplete information of a few genes.

ISTMEscore R package

We provided an R package named ISTMEscore to calculate our immune and stromal scores, and to estimate TME classification (GitHub: https://​github.​com/​ZengZihang/​ISTMEscore).

Cell-receptor-ligand communication networks

The connections of cells, receptors and ligands were retrieved from the FANTOM5 resource [33]. The edges of interaction networks in each TME subtype were determined by Thorsson’s method [34]. Core subnetworks were determined by the Molecular Complex Detection tool in Cytoscape [35].

Cell infiltration scores

Cell infiltration was estimated by the MCPcounter [9], containing T cells, CD8+ T cells, cytotoxic lymphocytes, NK cells, B lineages, monocytic lineages, myeloid dendritic cells, neutrophils, endothelial cells and fibroblasts. Estimation of stromal and immune cells in malignant tumors using expression (ESTIMATE) [8] scores provided the purity of immune and stromal components through ssGSEA.

Propensity score matching analysis

Propensity score matching (PSM) through 1:1 nearest neighbor matching with caliper was performed to adjust clinical confounders. Sample pairs with similar propensity scores (calipers = 0.05) were matched one-to-one in different groups to make the baseline levels consistent. PSM was performed by R MatchIt package [36].

Gene enrichment analysis

Gene set enrichment and over-representation analysis were performed by R clusterProfiler packages based on Gene Ontology (GO) corpus [37].

Decision curve analysis

Decision curve analysis (DCA) evaluated the clinical net benefit of risk prediction model using the R rmda package [38].

Statistical analysis

All statistical analysis was implemented by R software 3.6.1. R survival package was used to perform Cox proportional hazards regression, Kaplan–Meier analysis and calculation of concordance index (C-index) [39]. R stats package was used to perform Pearson, Spearman correlation and chi square test. The receiver operating characteristic (ROC) curve analysis was performed by R pROC package [40]. P value less than 0.05 was considered as statistically significant. All the P values were two-sided. The significant threshold of false discovery rate (FDR) was 0.05 in enrichment analysis.

Additional analysis

To explore the possible pathways of the stromal-related genes, we performed additional analysis (Additional file 18). The relevant parts were included in the supplementary materials.

Results

Feature extraction of TME signals and construction of ISTMEscore

The designs of our studies were demonstrated in Figs. 1, 2 and Additional file 1: Figure S1. To isolated the TME-related signals from mixed tumor tissues, we first divided RNA-seq data of the training cohort (TCGA LUAD) [11] into 11 heterogeneous clusters based on NMF (Fig. 3A). Enrichment analysis of clusters' high NMF weight genes (top 100) was performed to identify clusters with immune or stromal signals. Only one cluster was linked with T cell activation and IFN-γ production (Additional file 11: Table S3, Fig. 3B). We extracted the LDF of this cluster by NMF. Univariate Cox regression indicated that the LDF was a favorable prognostic factor (Cox-P < 0.0001). Therefore, this cluster was defined as "immune activation cluster". On the other hand, extracellular matrix organization, cell–matrix adhesion and TGF-β signal were enriched in another cluster (Additional file 12: Table S4, Fig. 3B), and its LDF was identified as an adverse prognostic factor (Cox-P < 0.0001), suggesting this cluster was a "stromal activation cluster". Enrichment results of other 9 clusters were displayed in Additional file 13: Table S5.
We next identified gene signatures from the immune and stromal clusters using our novel workflow (Fig. 2, see “Methods”—“Development of novel immune and stromal scores” for details). A total of 166 genes were identified as TME-related signatures, containing 108 immune- (e.g. CD40LG, CD8A, IFNG, PTPRC, CXCL10) and 58 stromal-related genes (e.g. MMP13, FN1, COL1A1, COL1A2, COL11A1, COL3A1, VEGFA, Additional file 14: Table S6). Novel immune and stromal scores were calculated by immune- and stromal-related signatures using ssGSEA.

ISTMEscore was linked to prognostic, cellular and molecular characteristics

The association of ISTMEscore with prognostic, cellular and molecular patterns were then investigated. Kaplan–Meier curves indicated that our immune scores were significantly associated with better OS, while stromal scores were associated with poorer OS in the training datasets (Fig. 3C, D). We next characterized the association between ISTMEscore and TME cell infiltration (estimated by MCPcounter) in the TCGA LUAD training dataset [11]. All 6 immune cells (T cells, CD8+ T cells, B cells, NK cells, monocytic lineage and myeloid dendritic cells) were positively correlated with the immune scores (Spearman correlation P < 0.0001, Fig. 3E), and the stromal scores were positively correlated with fibroblasts and negatively correlated with myeloid dendritic cells (Spearman correlation P < 0.0001).
Differentially expressed gene analysis was then performed in patients with high-low immune and stromal scores (Up 50% vs. Low 50%). The GSEA of differentially expressed genes suggested that T cell activation was highly enriched in the patients with high immune scores, while leukocyte activation was less enriched in the patients with high stromal scores (Fig. 3F, G).
The correlation between T cell co-stimulatory/inhibitory molecules and our scores were subsequently analyzed. The stromal scores were negatively correlated with CD28 and CD40LG, and positively correlated with PD-1 (Additional file 2: Figure S2), suggesting that the stromal scores were positively associated with exhaustive immunity.

Identification of TME subtypes by ISTMEscore

To explore the roles of immune and stromal activation in tumors, patients were divided into 4 TME subtypes according to the median values of ISTME scores. The patients with high immune and stromal scores were considered as the “HH type”, and high immune and low stromal scores were identified as the “HL type”. The “LH type” patients had low immune and high stromal scores, and the patients with low immune and stromal scores were defined as the “LL type”.

Comprehensive analysis of multi-dimensional landscape of the 4 TME subtypes in LUAD, SKCM and HNSC

The landscape of TME cell infiltration in different TME subtypes

To depict the TME immune landscape of the 4 subtypes, the infiltration of immune cells was calculated using MCPcounter. In the TCGA LUAD, SKCM and HNSC cohorts, the HH and HL patients had significantly high levels of immune cell infiltration, including T cells, CD8+ T cells, cytotoxic lymphocytes, NK cells, B lineages, monocytic lineages and myeloid dendritic cells (Fig. 4A–C). We further assessed the cellular patterns of TME subtypes in other independent cohorts, containing GSE11969 (NSCLC, n = 149) [12], GSE68465 (LUAD, n = 442) [13], GSE68571 (LUAD, n = 86) [14], GSE37745 (NSCLC, n = 196) [15], GSE50081 (NSCLC, n = 172) [16] and GSE65904 (SKCM, n = 214) [17]. Consistent with the outcomes in the TCGA cohorts, the HH and HL patients also had high immune cell infiltration (Additional file 3: Figure S3), except GSE68571, due to the lack of mapping genes.

TME subtype was associated with the functional markers of T cells and angiogenesis

Downregulation of co-stimulatory molecules (CD28, CD40LG) and upregulation of co-inhibitory molecules (PD-1, CTLA4, LAG3, TIM-3) were detected in the exhaustive T cells. In the TCGA LUAD dataset, the ratio of co-inhibitory/co-stimulatory molecules (PD-1 + CTLA4 + LAG3 + TIM-3)/(CD28 + CD40LG) was significantly different in the 4 TME subtypes (analysis of variance, ANOVA, P < 0.0001, Fig. 4D). Contrary to the LH patients, the HL patients had the lowest ratio. Similar patterns were also found in the TCGA SKCM (ANOVA, P = 0.0072) and TCGA HNSC datasets (ANOVA, P < 0.0001).
In addition, angiogenic markers, VEGFA levels and ANGPT1/ANGPT2 ratios had significantly different levels among TME subtypes in the TCGA LUAD and TCGA HNSC datasets (ANOVA, P < 0.0001, Fig. 4E, F). The HL patients had low levels of VEGFA and high ANGPT1/ANGPT2 ratios, suggesting inhibited angiogenesis. VEGFA levels were associated with TME subtypes in the TCGA SKCM dataset (ANOVA, P = 0.0004).

TME subtype was associated with distinct TME networks composed of cells, receptors and ligands

In TME, signal transmission between cells was mediated by secreted proteins and direct communications. The TME communication networks were established to characterize cell interactions, as well as receptors and ligands, with Thorsson's method in the training datasets (Fig. 4G) [34]. The TME networks of the HH type composed of CD8+ T cells, B cells, NK cells and fibroblasts as the center of the subnetworks mediated by integrins and chemokines. The immune cell-centered network was identified in the HL type. The fibroblast-centered networks were observed in the LH type. However, no network was identified in the LL type, suggesting its desert-like TME.

TME subtype was linked to clinicopathological characteristics

According to the above findings, our TME subtypes could represent TME patterns and were associated with immune regulation in LUAD, SKCM and HNSC. The correlations between TME subtypes and clinicopathological characteristics were next analyzed. In the TCGA LUAD dataset, the LH type was concentrated on young patients at pathologic N2–3 and stage III–IV (Table 1), which were related to higher malignancy and poorer survival. In the TCGA HNSC dataset, the LH type was significantly enriched in male patients at pathologic T3–4, which was contrary to HL. In the TCGA SKCM dataset, TME subtype was independent of clinicopathological characteristics.
Table 1
The clinical characteristics of the 4 subgroups in TCGA LUAD, TCGA SKCM and TCGA HNSC
Variable
TCGA LUAD
TCGA SKCM
TCGA HNSC
Subtypes
HH
HL
LH
LL
P
HH
HL
LH
LL
P
HH
HL
LH
LL
P
Gender (%)/Chi-squared test
    
0.35
    
0.14
    
0.0016**
 Male
41 (43.2)
66 (42.6)
75 (48.4)
51 (53.1)
 
47 (58.0)
53 (55.8)
66 (69.5)
55 (67.9)
 
76 (75.2)
102 (65.4)
131 (84.0)
70 (69.3)
 
 Female
54 (56.8)
89 (57.4)
80 (51.6)
45 (46.9)
 
34 (42.0)
42 (44.2)
29 (30.5)
26 (32.1)
 
25 (24.8)
54 (34.6)
25 (16.0)
31 (30.7)
 
Median age (IQR)/ANOVA
66.5 (57.7–72.2)
69.4 (62.7–75.0)
63.3 (56.2–71.6)
65.3 (60.5–72.0)
 < 0.0001****
56.7 (46.4–64.8)
56.3 (45.0–71.6)
56.1 (49.0–68.7)
60.0 (46.4–70.9)
0.71
60.77 (53.51–68.99)
61.11 (55.03–69.86)
60.85 (53.03–67.88)
61.72 (53.04–69.02)
0.72
Pathologic T (%)/Chi-squared test
    
0.16
    
0.068
    
0.018*
 T0–2
86 (90.5)
139 (90.8)
129 (83.8)
81 (84.4)
 
38 (58.5)
39 (52.0)
35 (45.5)
24 (36.4)
 
37 (41.1)
64 (48.5)
41 (30.0)
40 (43.0)
 
 T3–4
9 (9.5)
14 (9.2)
25 (16.2)
15 (15.6)
 
27 (41.5)
36 (48.0)
42 (54.5)
42 (63.6)
 
53 (58.9)
68 (51.5)
96 (70.0)
53 (57.0)
 
Pathologic N (%)/Chi-squared test
    
 < 0.0001****
    
0.3
    
0.42
 N0–1
81 (85.3)
139 (93.9)
115 (75.2)
83 (88.3)
 
50 (68.5)
66 (77.6)
59 (68.6)
55 (78.6)
 
40 (49.4)
71 (60.2)
77 (58.8)
51 (60.0)
 
 N2–3
14 (14.7)
9 (6.1)
38 (24.8)
11 (11.7)
 
23 (31.5)
19 (22.4)
27 (31.4)
15 (21.4)
 
41 (50.6)
47 (39.8)
54 (41.2)
34 (40.0)
 
Pathologic M (%)/Fisher's test
    
0.3
    
0.27
    
0.16
 M0
69 (94.5)
95 (96.0)
105 (92.1)
63 (88.7)
 
70 (93.3)
86 (96.6)
87 (94.6)
64 (88.9)
 
40 (100)
56 (100)
57 (100)
29 (96.7)
 
 M1
4 (5.5)
4 (4.0)
9 (7.9)
8 (11.3)
 
5 (6.7)
3 (3.4)
5 (5.4)
8 (11.1)
 
0
0
0
1 (3.3)
 
Pathologic stage (%)/Chi-squared test
    
 < 0.0001****
    
0.38
    
0.16
 Stage I
47 (49.5)
106 (69.3)
64 (41.3)
54 (56.3)
 
19 (25.7)
24 (28.9)
18 (19.8)
14 (19.4)
 
2 (2.2)
11 (8.5)
5 (3.7)
9 (9.9)
 
 Stage II
28 (29.5)
31 (20.3)
43 (27.7)
19 (19.8)
 
12 (16.2)
19 (22.9)
29 (31.9)
23 (31.9)
 
13 (14.6)
28 (21.7)
17 (12.6)
15 (16.5)
 
 Stage III
16 (16.8)
11 (7.2)
39 (25.2)
14 (14.6)
 
38 (51.4)
37 (44.6)
39 (42.9)
28 (38.9)
 
18 (20.2)
20 (15.5)
24 (17.8)
17 (18.7)
 
 Stage IV
4 (4.2)
5 (3.3)
9 (5.8)
9 (9.4)
 
5 (6.8)
3 (3.6)
5 (5.5)
7 (9.7)
 
56 (62.9)
70 (54.3)
89 (65.9)
50 (54.9)
 
Survival rate (%)/log-rank
    
 < 0.0001****
    
 < 0.0001****
    
0.39
 3-year OS
60.7
78.4
45.5
69.2
 
75
85.1
60.8
79.4
 
56.2
65.6
52.7
54.4
 
 5-year OS
41.2
55
30.2
40.1
 
65.1
78.4
43.2
63.3
 
46.2
50.3
47
47.4
 
LUAD lung adenocarcinoma, SKCM skin cutaneous melanoma, HNSC head and neck squamous cell carcinoma, NS not significant
* < 0.05; ** < 0.01; *** < 0.001; **** < 0.0001

TME subtype was an independent prognostic factor

The prognostic features (age, gender, pathologic stage and TME subtype) were identified by clinical value and univariate Cox regression (Fig. 5A–C). Pathological T, N and M have collinearity with pathologic stage, thus we only included the pathologic stages in the Cox model. In the TCGA LUAD dataset, multivariate Cox regression analysis indicated that age (HR = 1.02, P = 0.03), pathologic stage (HR = 1.58, P < 0.0001) and TME subtype (HL vs. LH, HR = 0.42, P < 0.0001) were independently prognostic factors (Fig. 5D). In the TCGA SKCM dataset, age (HR = 1.02, P = 0.0006), pathologic stage (HR = 1.36, P = 0.002) and TME subgroup (HL vs. LH, HR = 0.39, P < 0.0001) were independently prognostic factors (Fig. 5E). In the TCGA HNSC dataset, age (HR = 1.02, P = 0.006), gender (male vs. female, HR = 0.76, P = 0.1), pathologic stage (HR = 1.45, P < 0.001) and TME subgroup (HL vs. HH, HR = 0.74, P = 0.168) were independently prognostic factors (Fig. 5F). Combining the 4 prognostic factors using Cox model, DCA revealed that the inclusion of TME subtype had higher potential clinical utility (Fig. 5G–I).
In the Kaplan–Meier curves (Fig. 5J), the HL patients had the best OS in the TCGA LUAD (logrank-P < 0.0001), SKCM (logrank-P < 0.0001) and HNSC (logrank-P = 0.39) datasets. To adjust the impacts of prognostic covariates (age, gender and pathologic stage) on prognosis of TME subtypes, we performed PSM to baseline correction. In the TCGA LUAD dataset, the HL type had significantly favorable OS, while the LH type had unfavorable OS (logrank-P = 0.043, Additional file 4: Figure S4A&B) after PSM (For all subtypes: age, ANOVA, P = 0.986; gender, Chi-squared test, P = 0.418; stage, Chi-squared test, P = 0.228). In the TCGA SKCM dataset, the survival curve was similar to that of the TCGA LUAD dataset (logrank-P = 0.00061, Additional file 4: Figure S4C&D) after PSM (For all subtypes: age, ANOVA, P = 0.882; gender, Chi-squared test, P = 0.43; stage, Chi-squared test, P = 0.796). In the TCGA HNSC dataset, survival curve also showed difference (logrank-P = 0.096, HL vs. LH: P = 0.03, Additional file 4: Figure S4E&F) after PSM (For all subtypes: age, ANOVA, P = 0.622; gender, Chi-squared test, P = 0.931; stage, Chi-squared test, P = 0.471).
To validate our TME subtypes in the other 6 independent datasets [1217], we performed Kaplan–Meier analysis in each dataset. The HL patients were associated with significantly favorable prognosis in all validation datasets (Fig. 5J).

TME subtype was associated with mutation profile and copy number alteration

To determine whether TME subtypes were driven by gene mutations, we analyzed the somatic mutation profile in the 3 cancers from TCGA (Fig. 6A–C). Gene mutations in the 4 TME subtypes were compared using Fisher's test (Additional file 5: Figure S5A–F). However, there was no duplicated differentially mutated genes in the 3 cancers. The mutation frequencies of traditional driving genes were then compared (Additional file 6: Figure S6A–I). In LUAD and HNSC, the mutation frequency of TP53 was significantly downregulated in the HL type, and the mutation sites of TP53 were concentrated at DNA binding domains (Additional file 7: Figure S7A&B). In SKCM, there were no differently mutated driver genes in the 4 TME subtypes.
Despite the controversy, tumor mutation burden (TMB) was the novel biomarker for ICI therapy according to CheckMate-032 (SCLC) and CheckMate-026 (NSCLC) [41, 42], and its roles in prognosis of patients without ICIs remained unclear. The non-silent mutation counts in the exon region represent relative values of TMB [43]. In this study, the HH and LH types were associated with high non-silent mutation count (ANOVA P < 0.0001, Fig. 6D), which was linked to better OS (logrank-P = 0.055, Fig. 6E) in the TCGA LUAD dataset. In the TCGA SKCM and HNSC datasets, the TME subtype had no significant mutation count that was also linked to prognosis.
The association between CNA burden and prognosis was still unclear. Here, we defined CNA count burden (CNACB) as the total gene counts with CNA in each sample. In the 3 TCGA cohorts, the HL type had lowest CNACB (Fig. 6F), which was an unfavorable prognostic factor in HNSC (logrank-P = 0.0056) and LUAD (logrank-P = 0.1), but a favorable factor in SKCM (logrank-P = 0.1, Fig. 6G).
Five independent transcriptomic cohorts containing patients with immunotherapy were used to validate the predictive effects of our TME subtype on ICI responses. In the immunotherapy dataset 1 (Chen et al.) of 53 melanoma cases [5], there were 5 biopsy timepoints: pre-anti-CTLA4, on-anti-CTLA4, pre-anti-PD-1, on-anti-PD-1 and prog-anti-PD-1. Immune scores were markedly increased in on-anti-CTLA4 (P = 0.078) and on-anti-PD-1 (P = 0.0002) responders (Fig. 7A), while stromal scores were decreased in on-anti-PD-1 therapy (P = 0.0013) responders (Fig. 7B). The HL patients showed higher responses than the LH patients with on-anti-CTLA4/PD-1 therapy (HH vs. HL vs. LH vs. LL: 100% vs. 100% vs. 0% vs. 0%, Fisher's test P = 0.0012, Fig. 7C). Notably, 41.9% patients experienced transition of TME subtype at the 5 biopsy timepoints (Fig. 7D). There were 8 patients with changes from the LH and LL types before anti-CTLA4 therapy to HH and HL types at later times, and 75% patients were responsive.
The immunotherapy dataset 2 (GSE91061) included 65 melanoma patients with anti-PD-1 therapy [24]. Similar to immunotherapy dataset 1, immune scores were significantly higher in responders with on-anti-PD-1 therapy (P = 0.0049, Fig. 7E), but stromal scores were not significantly lower (Fig. 7F). The LH patients were linked to low responses of on-anti-PD-1 therapy (HH vs. HL vs. LH vs. LL: 54.5% vs. 78.9% vs. 37.5% vs. 44.4%, Fisher's test P = 0.07, Fig. 7G). Moreover, 29% patients experienced transition of TME subtype before and during treatment (Fig. 7H). Furthermore, 66.7% patients, who changed to LH and LL types, were not responsive, while 66.7% patients, who changed to HH and HL types, were responsive.
Other immunotherapy datasets only included transcriptomic data before treatment. There was no statistical significance between TME subtype and immunotherapy response in these datasets (Fig. 7I–L).

Comparison of ISTMEscore system with other studies

The overlap between our TME gene signatures and existing signatures

To test the robustness of this study, we compared our TME signatures (“Results”—“Feature extraction of TME signals and construction of ISTMEscore”) with existing signatures [4449]. There were 30.6% and 31% overlaps of ISTMEscore signatures with ESTIMATE and MCPcounter, respectively (Additional file 15: Table S7).

Comparison with other algorithms for prediction of prognosis and ICI responses

Since ESTIMATE [8] and MCPcounter [9] algorithms also provided TME-related scores, it was necessary to compare these indicators with our ISTMEscore. We first calculated C-index for each score on prognosis prediction in all 9 datasets with prognostic annotation (Additional file 16: Table S8). The top 5 indicators of C-index were our stromal score (mean C-index: 0.587), our immune score (mean rank: 0.58), B lineage infiltration (mean C-index: 0.578), myeloid dendritic cell infiltration (mean C-index: 0.567), ESTIMATE immune score (mean C-index: 0.566). We next applied ROC analysis on ICI responses, and calculated area under receiver (AUC) of ROC in the 5 immunotherapy cohorts (Additional file 16: Table S8). The top 5 indicators of AUC were monocytic lineage infiltration (mean AUC: 0.73462), our immune score (mean AUC: 0.72636), myeloid dendritic cell infiltration (mean AUC: 0.72574), T cell infiltration (mean AUC: 0.72034) and our stromal score (mean AUC: 0.71854).
Moreover, we compared our scores with ESTIMATE scores in GSE9014 [50], which included 111 arrays of stroma through Laser Capture Microdissected from 53 breast cancer patients. Only our stromal scores were significantly associated with the histologic GRADE (Additional file 8: Figure S8A-D, P < 0.0001). Table of abbreviations was displayed in Additional file 17: Table S9.

Discussion

In this study, we built the novel ISTMEscore system with unique workflow, and depicted the multi-dimensional landscape of different TME subtypes. Our TME subtypes could represent TME patterns, and were associated with clinical features and immunotherapy responses in LUAD, SKCM and HNSC. Additional analysis suggested that high collagen, matrix metalloproteinases, glycolysis and acid environment, VEGF signaling were the integral parts of stromal activation. Whether the stromal signals were involved in immune exhaustion remained to be further studied.
Although immunotherapy benefited LUAD, SKCM and HNSC patients, only a small proportion of patients had long-term survival [5]. Identification of potentially sensitive population for ICIs helped to decrease medical expenses and improve quality of life. Our studies found that the LH type with malignant TME and the LL type with desert-like TME had low ICI responses (Fig. 7). Interestingly, the TME subtypes determined by transcriptome before treatment did not demonstrate significant association with immunotherapy response. In the 3 immunotherapy cohorts with biopsies taken before treatment, TME subtype was not significantly associated with ICI response: HH vs. HL vs. LH vs. LL: 100% vs. 94.7% vs. 94.7% vs. 78.6%, Fisher's test P = 0.2 in GSE93157 (Fig. 7I) [25]; and HH vs. HL vs. LH vs. LL: 100% vs. 33.3% vs. 33.3% vs. 0%, Fisher’s test P = 0.26 in GSE67501 (Fig. 7J) [26]; and HH vs. HL vs. LH vs. LL: 50% vs. 52.9% vs. 13.3% vs. 40%, Fisher’s test P = 0.09 in GSE35640 (Fig. 7K) [27]. In GSE93157, there was a difference of progression-free survival only in subgroup comparisons of HL vs. LH (logrank-P = 0.056, Fig. 7L). Chen et al. also found positive implications for dynamic monitoring of the immune microenvironment [5]. The protein levels of CD3, CD4, CD8, PD-1, PD-L1 and LAG3 during the treatment could reflect the responses (all, P < 0.01) better than those before treatment. Moreover, Riaz et al. found that patients with TME of immune activation or “hot tumor” were associated with high CR/PR rates in the group receiving ICIs in advance, whereas TME immune infiltration was not linked to ICI responses in the patients without prior immunotherapy [24]. In stromal environment, the desmoplastic stroma was the physical barrier for tumor to resist immunotherapy and chemotherapy [51]. Kim et al. reported the single-cell sequencing analysis of the longitudinal samples from 20 triple-negative breast cancer patients during neoadjuvant chemotherapy [52]. They found that degradation of ECM and angiogenesis signals were upregulated in the chemoresistant tumors. In preclinical studies, cancer-associated fibroblasts were reported to compensate immunotherapy through crosstalk with myeloid-derived suppressor cells and CD8+ T cells [53, 54]. In this study, we found that the majority of patients, who switched to the LH and LL types during ICI therapy, were non-responders. Accordingly, longitudinal detection of TME-related indicators might be a better choice for patients with ICI treatment, which required studies with large sample size.
The HL patients had high immune activation, inhibitory angiogenesis and long OS, which was considered as balanced states of TME. As previous studies, there were synergistic effects between immune normalization and vascular normalization [55]. VEGF induced immunosuppressive cells such as myeloid suppressive cells, tumor-related macrophages and Tregs, which developed immune exhaustion. On the other hand, the infiltration and activation of intratumoral effector T cells promoted the remodeling and normalization of vessels. The immune and vascular normalization might explain the results of IMpower150 study, in which the first-line immunotherapy combined with anti-angiogenic drugs benefited non-squamous NSCLC patients [56]. The immune and vascular normalization seemed to correspond to the HL type in our study.
Compared with the existing methods (ESTIMATE and MCPcounter) [8, 9], our scores showed advantages on the prediction of prognosis and immunotherapy response. However, the improvement in predictive performance was not large. B lineage and myeloid dendritic cell infiltration scores of MCPcounter were also good predictors of prognosis (Additional file 16: Table S8). For prediction of ICI responses, monocytic lineage infiltration scores also had excellent performance.
The effects of TMB on prognosis were controversial. Some clinical studies revealed opposite prognostic effects of TMB in NSCLC patients without immunotherapy. In LACE-Bio-II study with 908 NSCLC patients [57], high TMB group (≥ 8 m/Mb) showed better OS, while the low TMB group (< 4 m/Mb) had worse prognosis (P = 0.016). However, another clinical study indicated that higher TMB (≥ 62 m/Mb) was correlated with worse OS in the 90 NSCLC patients (P = 0.0003) [58]. TMB may not be a very robust prognostic marker due to lack of consideration on the threshold, complex effects of mutation, as well as the gene mutations from immune or stromal cells.
There were some limitations in this study: All datasets were retrospective, requiring prospective clinical trials to further verify. Whether our scores worked in other tumors, especially tumors lacking immune cell infiltration, remained to be further investigated. In addition, the link between our ISMEscore system and ICI response was not strong. Our system needed to be applied cautiously for the prediction of immunotherapeutic efficacy.

Conclusions

In conclusion, we proposed the novel ISTMEscore and TME classification. We comprehensively depicted TME classification associated with the cellular, molecular, TME communication networks, mutation, CNA burden and clinical features of LUAD, SKCM and HNSC. Our TME classification was an independent prognostic factor and associated with immunotherapy responses, which was superior to previous studies.

Acknowledgements

Not applicable.

Declarations

Not applicable.
Not applicable.

Competing interests

The authors declare that they have no competing interests.
Open AccessThis article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article's Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://​creativecommons.​org/​licenses/​by/​4.​0/​. 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 in a credit line to the data.

Publisher's Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
insite
INHALT
download
DOWNLOAD
print
DRUCKEN
Anhänge

Supplementary Information

Literatur
1.
Zurück zum Zitat Luke JJ, Flaherty KT, Ribas A, Long GV. Targeted agents and immunotherapies: optimizing outcomes in melanoma. Nat Rev Clin Oncol. 2017;14(8):463–82.PubMedCrossRef Luke JJ, Flaherty KT, Ribas A, Long GV. Targeted agents and immunotherapies: optimizing outcomes in melanoma. Nat Rev Clin Oncol. 2017;14(8):463–82.PubMedCrossRef
2.
Zurück zum Zitat Ock CY, Keam B, Kim S, Lee JS, Kim M, Kim TM, et al. Pan-cancer immunogenomic perspective on the tumor microenvironment based on PD-L1 and CD8 T-cell infiltration. Clin Cancer Res. 2016;22(9):2261–70.PubMedCrossRef Ock CY, Keam B, Kim S, Lee JS, Kim M, Kim TM, et al. Pan-cancer immunogenomic perspective on the tumor microenvironment based on PD-L1 and CD8 T-cell infiltration. Clin Cancer Res. 2016;22(9):2261–70.PubMedCrossRef
3.
Zurück zum Zitat Chen YP, Wang YQ, Lv JW, Li YQ, Chua MLK, Le QT, et al. Identification and validation of novel microenvironment-based immune molecular subgroups of head and neck squamous cell carcinoma: implications for immunotherapy. Ann Oncol. 2019;30(1):68–75.PubMedCrossRef Chen YP, Wang YQ, Lv JW, Li YQ, Chua MLK, Le QT, et al. Identification and validation of novel microenvironment-based immune molecular subgroups of head and neck squamous cell carcinoma: implications for immunotherapy. Ann Oncol. 2019;30(1):68–75.PubMedCrossRef
4.
Zurück zum Zitat Bremnes RM, Donnem T, Al-Saad S, Al-Shibli K, Andersen S, Sirera R, et al. The role of tumor stroma in cancer progression and prognosis: emphasis on carcinoma-associated fibroblasts and non-small cell lung cancer. J Thorac Oncol. 2011;6(1):209–17.PubMedCrossRef Bremnes RM, Donnem T, Al-Saad S, Al-Shibli K, Andersen S, Sirera R, et al. The role of tumor stroma in cancer progression and prognosis: emphasis on carcinoma-associated fibroblasts and non-small cell lung cancer. J Thorac Oncol. 2011;6(1):209–17.PubMedCrossRef
5.
Zurück zum Zitat Chen PL, Roh W, Reuben A, Cooper ZA, Spencer CN, Prieto PA, et al. Analysis of immune signatures in longitudinal tumor samples yields insight into biomarkers of response and mechanisms of resistance to immune checkpoint blockade. Cancer Discov. 2016;6(8):827–37.PubMedPubMedCentralCrossRef Chen PL, Roh W, Reuben A, Cooper ZA, Spencer CN, Prieto PA, et al. Analysis of immune signatures in longitudinal tumor samples yields insight into biomarkers of response and mechanisms of resistance to immune checkpoint blockade. Cancer Discov. 2016;6(8):827–37.PubMedPubMedCentralCrossRef
7.
Zurück zum Zitat Li T, Fan J, Wang B, Traugh N, Chen Q, Liu JS, et al. TIMER: a web server for comprehensive analysis of tumor-infiltrating immune cells. Can Res. 2017;77(21):e108–10.CrossRef Li T, Fan J, Wang B, Traugh N, Chen Q, Liu JS, et al. TIMER: a web server for comprehensive analysis of tumor-infiltrating immune cells. Can Res. 2017;77(21):e108–10.CrossRef
8.
Zurück zum Zitat Yoshihara K, Shahmoradgoli M, Martinez E, Vegesna R, Kim H, Torres-Garcia W, et al. Inferring tumour purity and stromal and immune cell admixture from expression data. Nat Commun. 2013;4:2612.PubMedCrossRef Yoshihara K, Shahmoradgoli M, Martinez E, Vegesna R, Kim H, Torres-Garcia W, et al. Inferring tumour purity and stromal and immune cell admixture from expression data. Nat Commun. 2013;4:2612.PubMedCrossRef
9.
Zurück zum Zitat Becht E, Giraldo N, Lacroix L, Buttard B, Elarouci N, Petitprez F, et al. Estimating the population abundance of tissue-infiltrating immune and stromal cell populations using gene expression. Genome Biol. 2016;17:218.PubMedPubMedCentralCrossRef Becht E, Giraldo N, Lacroix L, Buttard B, Elarouci N, Petitprez F, et al. Estimating the population abundance of tissue-infiltrating immune and stromal cell populations using gene expression. Genome Biol. 2016;17:218.PubMedPubMedCentralCrossRef
10.
Zurück zum Zitat Hanzelmann S, Castelo R, Guinney J. GSVA: gene set variation analysis for microarray and RNA-seq data. BMC Bioinform. 2013;14:7.CrossRef Hanzelmann S, Castelo R, Guinney J. GSVA: gene set variation analysis for microarray and RNA-seq data. BMC Bioinform. 2013;14:7.CrossRef
11.
Zurück zum Zitat Weinstein JN, Collisson EA, Mills GB, Shaw KR, Ozenberger BA, Ellrott K, et al. The cancer genome atlas pan-cancer analysis project. Nat Genet. 2013;45(10):1113–20.PubMedPubMedCentralCrossRef Weinstein JN, Collisson EA, Mills GB, Shaw KR, Ozenberger BA, Ellrott K, et al. The cancer genome atlas pan-cancer analysis project. Nat Genet. 2013;45(10):1113–20.PubMedPubMedCentralCrossRef
12.
Zurück zum Zitat Takeuchi T, Tomida S, Yatabe Y, Kosaka T, Osada H, Yanagisawa K, et al. Expression profile-defined classification of lung adenocarcinoma shows close relationship with underlying major genetic changes and clinicopathologic behaviors. J Cli Oncol. 2006;24(11):1679–88.CrossRef Takeuchi T, Tomida S, Yatabe Y, Kosaka T, Osada H, Yanagisawa K, et al. Expression profile-defined classification of lung adenocarcinoma shows close relationship with underlying major genetic changes and clinicopathologic behaviors. J Cli Oncol. 2006;24(11):1679–88.CrossRef
13.
Zurück zum Zitat Shedden K, Taylor JM, Enkemann SA, Tsao MS, Yeatman TJ, Gerald WL, et al. Gene expression-based survival prediction in lung adenocarcinoma: a multi-site, blinded validation study. Nat Med. 2008;14(8):822–7.PubMedPubMedCentralCrossRef Shedden K, Taylor JM, Enkemann SA, Tsao MS, Yeatman TJ, Gerald WL, et al. Gene expression-based survival prediction in lung adenocarcinoma: a multi-site, blinded validation study. Nat Med. 2008;14(8):822–7.PubMedPubMedCentralCrossRef
14.
Zurück zum Zitat Beer DG, Kardia SL, Huang CC, Giordano TJ, Levin AM, Misek DE, et al. Gene-expression profiles predict survival of patients with lung adenocarcinoma. Nat Med. 2002;8(8):816–24.PubMedCrossRef Beer DG, Kardia SL, Huang CC, Giordano TJ, Levin AM, Misek DE, et al. Gene-expression profiles predict survival of patients with lung adenocarcinoma. Nat Med. 2002;8(8):816–24.PubMedCrossRef
15.
Zurück zum Zitat Botling J, Edlund K, Lohr M, Hellwig B, Holmberg L, Lambe M, et al. Biomarker discovery in non-small cell lung cancer: integrating gene expression profiling, meta-analysis, and tissue microarray validation. Clin Cancer Res. 2013;19(1):194–204.PubMedCrossRef Botling J, Edlund K, Lohr M, Hellwig B, Holmberg L, Lambe M, et al. Biomarker discovery in non-small cell lung cancer: integrating gene expression profiling, meta-analysis, and tissue microarray validation. Clin Cancer Res. 2013;19(1):194–204.PubMedCrossRef
16.
Zurück zum Zitat Der SD, Sykes J, Pintilie M, Zhu CQ, Strumpf D, Liu N, et al. Validation of a histology-independent prognostic gene signature for early-stage, non-small-cell lung cancer including stage IA patients. J Thorac Oncol. 2014;9(1):59–64.PubMedCrossRef Der SD, Sykes J, Pintilie M, Zhu CQ, Strumpf D, Liu N, et al. Validation of a histology-independent prognostic gene signature for early-stage, non-small-cell lung cancer including stage IA patients. J Thorac Oncol. 2014;9(1):59–64.PubMedCrossRef
17.
Zurück zum Zitat Cabrita R, Lauss M, Sanna A, Donia M, Skaarup Larsen M, Mitra S, et al. Tertiary lymphoid structures improve immunotherapy and survival in melanoma. Nature. 2020;577(7791):561–5.PubMedCrossRef Cabrita R, Lauss M, Sanna A, Donia M, Skaarup Larsen M, Mitra S, et al. Tertiary lymphoid structures improve immunotherapy and survival in melanoma. Nature. 2020;577(7791):561–5.PubMedCrossRef
18.
Zurück zum Zitat Deng M, Brägelmann J, Kryukov I, Saraiva-Agostinho N, Perner S. FirebrowseR: an R client to the Broad Institute’s Firehose Pipeline. Database (Oxford). 2017;2017:160.CrossRef Deng M, Brägelmann J, Kryukov I, Saraiva-Agostinho N, Perner S. FirebrowseR: an R client to the Broad Institute’s Firehose Pipeline. Database (Oxford). 2017;2017:160.CrossRef
19.
Zurück zum Zitat Harbron C, Chang KM, South MC. RefPlus: an R package extending the RMA Algorithm. Bioinformatics. 2007;23(18):2493–4.PubMedCrossRef Harbron C, Chang KM, South MC. RefPlus: an R package extending the RMA Algorithm. Bioinformatics. 2007;23(18):2493–4.PubMedCrossRef
20.
Zurück zum Zitat Patro SG, Sahu KK. Normalization: a preprocessing stage. IARJSET; 2015. Patro SG, Sahu KK. Normalization: a preprocessing stage. IARJSET; 2015.
21.
Zurück zum Zitat Leek JT, Johnson WE, Parker HS, Jaffe AE, Storey JD. The sva package for removing batch effects and other unwanted variation in high-throughput experiments. Bioinformatics. 2012;28(6):882–3.PubMedPubMedCentralCrossRef Leek JT, Johnson WE, Parker HS, Jaffe AE, Storey JD. The sva package for removing batch effects and other unwanted variation in high-throughput experiments. Bioinformatics. 2012;28(6):882–3.PubMedPubMedCentralCrossRef
22.
Zurück zum Zitat Van der Auwera GA, Carneiro MO, Hartl C, Poplin R, Del Angel G, Levy-Moonshine A, et al. From FastQ data to high confidence variant calls: the Genome Analysis Toolkit best practices pipeline. Curr Protoc Bioinform. 2013;43:11.0.1-33. Van der Auwera GA, Carneiro MO, Hartl C, Poplin R, Del Angel G, Levy-Moonshine A, et al. From FastQ data to high confidence variant calls: the Genome Analysis Toolkit best practices pipeline. Curr Protoc Bioinform. 2013;43:11.0.1-33.
23.
Zurück zum Zitat Mermel CH, Schumacher SE, Hill B, Meyerson ML, Beroukhim R, Getz G. GISTIC2.0 facilitates sensitive and confident localization of the targets of focal somatic copy-number alteration in human cancers. Genome Biol. 2011;12(4):R41.PubMedPubMedCentralCrossRef Mermel CH, Schumacher SE, Hill B, Meyerson ML, Beroukhim R, Getz G. GISTIC2.0 facilitates sensitive and confident localization of the targets of focal somatic copy-number alteration in human cancers. Genome Biol. 2011;12(4):R41.PubMedPubMedCentralCrossRef
24.
Zurück zum Zitat Riaz N, Havel JJ, Makarov V, Desrichard A, Urba WJ, Sims JS, et al. Tumor and microenvironment evolution during immunotherapy with nivolumab. Cell. 2017;171(4):934-49.e16.PubMedPubMedCentralCrossRef Riaz N, Havel JJ, Makarov V, Desrichard A, Urba WJ, Sims JS, et al. Tumor and microenvironment evolution during immunotherapy with nivolumab. Cell. 2017;171(4):934-49.e16.PubMedPubMedCentralCrossRef
25.
Zurück zum Zitat Prat A, Navarro A, Paré L, Reguart N, Galván P, Pascual T, et al. Immune-Related gene expression profiling after PD-1 blockade in non-small cell lung carcinoma, head and neck squamous cell carcinoma, and melanoma. Can Res. 2017;77(13):3540–50.CrossRef Prat A, Navarro A, Paré L, Reguart N, Galván P, Pascual T, et al. Immune-Related gene expression profiling after PD-1 blockade in non-small cell lung carcinoma, head and neck squamous cell carcinoma, and melanoma. Can Res. 2017;77(13):3540–50.CrossRef
26.
Zurück zum Zitat Ascierto ML, McMiller TL, Berger AE, Danilova L, Anders RA, Netto GJ, et al. The Intratumoral balance between metabolic and immunologic gene expression is associated with anti-PD-1 response in patients with renal cell carcinoma. Cancer Immunol Res. 2016;4(9):726–33.PubMedPubMedCentralCrossRef Ascierto ML, McMiller TL, Berger AE, Danilova L, Anders RA, Netto GJ, et al. The Intratumoral balance between metabolic and immunologic gene expression is associated with anti-PD-1 response in patients with renal cell carcinoma. Cancer Immunol Res. 2016;4(9):726–33.PubMedPubMedCentralCrossRef
27.
Zurück zum Zitat Ulloa-Montoya F, Louahed J, Dizier B, Gruselle O, Spiessens B, Lehmann FF, et al. Predictive gene signature in MAGE-A3 antigen-specific cancer immunotherapy. J Clin Oncol. 2013;31(19):2388–95.PubMedCrossRef Ulloa-Montoya F, Louahed J, Dizier B, Gruselle O, Spiessens B, Lehmann FF, et al. Predictive gene signature in MAGE-A3 antigen-specific cancer immunotherapy. J Clin Oncol. 2013;31(19):2388–95.PubMedCrossRef
28.
Zurück zum Zitat Gaujoux R, Seoighe C. A flexible R package for nonnegative matrix factorization. BMC Bioinform. 2010;11:367.CrossRef Gaujoux R, Seoighe C. A flexible R package for nonnegative matrix factorization. BMC Bioinform. 2010;11:367.CrossRef
29.
Zurück zum Zitat Zhou J, Chen J, Ye J. MALSAR: Multi-tAsk Learning via StructurAl Regularization; 2012. Zhou J, Chen J, Ye J. MALSAR: Multi-tAsk Learning via StructurAl Regularization; 2012.
30.
Zurück zum Zitat Reich M, Liefeld T, Gould J, Lerner J, Tamayo P, Mesirov JP. GenePattern 2.0. Nat Genet. 2006;38(5):500–1.PubMedCrossRef Reich M, Liefeld T, Gould J, Lerner J, Tamayo P, Mesirov JP. GenePattern 2.0. Nat Genet. 2006;38(5):500–1.PubMedCrossRef
31.
Zurück zum Zitat Ritchie ME, Phipson B, Wu D, Hu Y, Law CW, Shi W, et al. limma powers differential expression analyses for RNA-sequencing and microarray studies. Nucleic Acids Res. 2015;43(7):e47.PubMedPubMedCentralCrossRef Ritchie ME, Phipson B, Wu D, Hu Y, Law CW, Shi W, et al. limma powers differential expression analyses for RNA-sequencing and microarray studies. Nucleic Acids Res. 2015;43(7):e47.PubMedPubMedCentralCrossRef
32.
Zurück zum Zitat Benjamini Y, Hochberg Y. Controlling the false discovery rate—a practical and powerful approach to multiple testing. J R Stat Soc Ser B. 1995;57:289–300. Benjamini Y, Hochberg Y. Controlling the false discovery rate—a practical and powerful approach to multiple testing. J R Stat Soc Ser B. 1995;57:289–300.
33.
Zurück zum Zitat Ramilowski JA, Goldberg T, Harshbarger J, Kloppmann E, Lizio M, Satagopam VP, et al. A draft network of ligand–receptor-mediated multicellular signalling in human. Nat Commun. 2015;6(1):7866.PubMedCrossRef Ramilowski JA, Goldberg T, Harshbarger J, Kloppmann E, Lizio M, Satagopam VP, et al. A draft network of ligand–receptor-mediated multicellular signalling in human. Nat Commun. 2015;6(1):7866.PubMedCrossRef
34.
35.
Zurück zum Zitat Shannon P, Markiel A, Ozier O, Baliga NS, Wang JT, Ramage D, et al. Cytoscape: a software environment for integrated models of biomolecular interaction networks. Genome Res. 2003;13(11):2498–504.PubMedPubMedCentralCrossRef Shannon P, Markiel A, Ozier O, Baliga NS, Wang JT, Ramage D, et al. Cytoscape: a software environment for integrated models of biomolecular interaction networks. Genome Res. 2003;13(11):2498–504.PubMedPubMedCentralCrossRef
36.
Zurück zum Zitat Ho DE, Imai K, King G, Stuart EA. Matching as nonparametric preprocessing for reducing model dependence in parametric causal inference. Polit Anal. 2017;15(3):199–236.CrossRef Ho DE, Imai K, King G, Stuart EA. Matching as nonparametric preprocessing for reducing model dependence in parametric causal inference. Polit Anal. 2017;15(3):199–236.CrossRef
37.
38.
Zurück zum Zitat Kerr KF, Brown MD, Zhu K, Janes H. Assessing the clinical impact of risk prediction models with decision curves: guidance for correct interpretation and appropriate use. J Clin Oncol. 2016;34(21):2534–40.PubMedPubMedCentralCrossRef Kerr KF, Brown MD, Zhu K, Janes H. Assessing the clinical impact of risk prediction models with decision curves: guidance for correct interpretation and appropriate use. J Clin Oncol. 2016;34(21):2534–40.PubMedPubMedCentralCrossRef
39.
Zurück zum Zitat Therneau T, Grambsch P. Modeling survival data: extending the cox model. New York: Springer; 2013. Therneau T, Grambsch P. Modeling survival data: extending the cox model. New York: Springer; 2013.
40.
Zurück zum Zitat Robin X, Turck N, Hainard A, Tiberti N, Lisacek F, Sanchez J-C, et al. pROC: an open-source package for R and S+ to analyze and compare ROC curves. BMC Bioinform. 2011;12(1):77.CrossRef Robin X, Turck N, Hainard A, Tiberti N, Lisacek F, Sanchez J-C, et al. pROC: an open-source package for R and S+ to analyze and compare ROC curves. BMC Bioinform. 2011;12(1):77.CrossRef
41.
Zurück zum Zitat Antonia SJ, Lopez-Martin JA, Bendell J, Ott PA, Taylor M, Eder JP, et al. Nivolumab alone and nivolumab plus ipilimumab in recurrent small-cell lung cancer (CheckMate 032): a multicentre, open-label, phase 1/2 trial. Lancet Oncol. 2016;17(7):883–95.PubMedCrossRef Antonia SJ, Lopez-Martin JA, Bendell J, Ott PA, Taylor M, Eder JP, et al. Nivolumab alone and nivolumab plus ipilimumab in recurrent small-cell lung cancer (CheckMate 032): a multicentre, open-label, phase 1/2 trial. Lancet Oncol. 2016;17(7):883–95.PubMedCrossRef
42.
Zurück zum Zitat Carbone DP, Reck M, Paz-Ares L, Creelan B, Horn L, Steins M, et al. First-line nivolumab in stage IV or recurrent non-small-cell lung cancer. N Engl J Med. 2017;376(25):2415–26.PubMedPubMedCentralCrossRef Carbone DP, Reck M, Paz-Ares L, Creelan B, Horn L, Steins M, et al. First-line nivolumab in stage IV or recurrent non-small-cell lung cancer. N Engl J Med. 2017;376(25):2415–26.PubMedPubMedCentralCrossRef
43.
Zurück zum Zitat Cao R, Yuan L, Ma B, Wang G, Tian Y. Tumour microenvironment (TME) characterization identified prognosis and immunotherapy response in muscle-invasive bladder cancer (MIBC). Cancer Immunol Immunother. 2020;70:1–18.PubMedCrossRef Cao R, Yuan L, Ma B, Wang G, Tian Y. Tumour microenvironment (TME) characterization identified prognosis and immunotherapy response in muscle-invasive bladder cancer (MIBC). Cancer Immunol Immunother. 2020;70:1–18.PubMedCrossRef
44.
Zurück zum Zitat Chifman J, Pullikuth A, Chou JW, Bedognetti D, Miller LD. Conservation of immune gene signatures in solid tumors and prognostic implications. BMC Cancer. 2016;16(1):911.PubMedPubMedCentralCrossRef Chifman J, Pullikuth A, Chou JW, Bedognetti D, Miller LD. Conservation of immune gene signatures in solid tumors and prognostic implications. BMC Cancer. 2016;16(1):911.PubMedPubMedCentralCrossRef
45.
Zurück zum Zitat Jiang L, Chughtai K, Purvine SO, Bhujwalla ZM, Raman V, Paša-Tolić L, et al. MALDI-mass spectrometric imaging revealing hypoxia-driven lipids and proteins in a breast tumor model. Anal Chem. 2015;87(12):5947–56.PubMedPubMedCentralCrossRef Jiang L, Chughtai K, Purvine SO, Bhujwalla ZM, Raman V, Paša-Tolić L, et al. MALDI-mass spectrometric imaging revealing hypoxia-driven lipids and proteins in a breast tumor model. Anal Chem. 2015;87(12):5947–56.PubMedPubMedCentralCrossRef
46.
Zurück zum Zitat Rodrigues-Lisoni FC, Peitl P Jr, Vidotto A, Polachini GM, Maniglia JV, Carmona-Raphe J, et al. Genomics and proteomics approaches to the study of cancer-stroma interactions. BMC Med Genomics. 2010;3:14.PubMedPubMedCentralCrossRef Rodrigues-Lisoni FC, Peitl P Jr, Vidotto A, Polachini GM, Maniglia JV, Carmona-Raphe J, et al. Genomics and proteomics approaches to the study of cancer-stroma interactions. BMC Med Genomics. 2010;3:14.PubMedPubMedCentralCrossRef
47.
Zurück zum Zitat Hanley CJ, Noble F, Ward M, Bullock M, Drifka C, Mellone M, et al. A subset of myofibroblastic cancer-associated fibroblasts regulate collagen fiber elongation, which is prognostic in multiple cancers. Oncotarget. 2016;7(5):6159–74.PubMedCrossRef Hanley CJ, Noble F, Ward M, Bullock M, Drifka C, Mellone M, et al. A subset of myofibroblastic cancer-associated fibroblasts regulate collagen fiber elongation, which is prognostic in multiple cancers. Oncotarget. 2016;7(5):6159–74.PubMedCrossRef
48.
Zurück zum Zitat Reuben A, Spencer CN, Prieto PA, Gopalakrishnan V, Reddy SM, Miller JP, et al. Genomic and immune heterogeneity are associated with differential responses to therapy in melanoma. NPJ Genomic Med. 2017;2:10.CrossRef Reuben A, Spencer CN, Prieto PA, Gopalakrishnan V, Reddy SM, Miller JP, et al. Genomic and immune heterogeneity are associated with differential responses to therapy in melanoma. NPJ Genomic Med. 2017;2:10.CrossRef
49.
Zurück zum Zitat Roufas C, Chasiotis D, Makris A, Efstathiades C, Dimopoulos C, Zaravinos A. The expression and prognostic impact of immune cytolytic activity-related markers in human malignancies: a comprehensive meta-analysis. Front Oncol. 2018;8:27.PubMedPubMedCentralCrossRef Roufas C, Chasiotis D, Makris A, Efstathiades C, Dimopoulos C, Zaravinos A. The expression and prognostic impact of immune cytolytic activity-related markers in human malignancies: a comprehensive meta-analysis. Front Oncol. 2018;8:27.PubMedPubMedCentralCrossRef
50.
Zurück zum Zitat Finak G, Bertos N, Pepin F, Sadekova S, Souleimanova M, Zhao H, et al. Stromal gene expression predicts clinical outcome in breast cancer. Nat Med. 2008;14(5):518–27.PubMedCrossRef Finak G, Bertos N, Pepin F, Sadekova S, Souleimanova M, Zhao H, et al. Stromal gene expression predicts clinical outcome in breast cancer. Nat Med. 2008;14(5):518–27.PubMedCrossRef
51.
Zurück zum Zitat Galluzzi L, Chan TA. The hallmarks of successful anticancer immunotherapy. Sci Transl Med. 2018;10(459):eaat7807.PubMedCrossRef Galluzzi L, Chan TA. The hallmarks of successful anticancer immunotherapy. Sci Transl Med. 2018;10(459):eaat7807.PubMedCrossRef
52.
Zurück zum Zitat Kim C, Gao R, Sei E, Brandt R, Hartman J, Hatschek T, et al. Chemoresistance evolution in triple-negative breast cancer delineated by single-cell sequencing. Cell. 2018;173(4):879-93.e13.PubMedPubMedCentralCrossRef Kim C, Gao R, Sei E, Brandt R, Hartman J, Hatschek T, et al. Chemoresistance evolution in triple-negative breast cancer delineated by single-cell sequencing. Cell. 2018;173(4):879-93.e13.PubMedPubMedCentralCrossRef
53.
Zurück zum Zitat Ford K, Hanley CJ, Mellone M, Szyndralewiez C, Heitz F, Wiesel P, et al. NOX4 inhibition potentiates immunotherapy by overcoming cancer-associated fibroblast-mediated CD8 T-cell exclusion from tumors. Can Res. 2020;80(9):1846.CrossRef Ford K, Hanley CJ, Mellone M, Szyndralewiez C, Heitz F, Wiesel P, et al. NOX4 inhibition potentiates immunotherapy by overcoming cancer-associated fibroblast-mediated CD8 T-cell exclusion from tumors. Can Res. 2020;80(9):1846.CrossRef
55.
Zurück zum Zitat Tian L, Goldstein A, Wang H, Ching Lo H, Sun Kim I, Welte T, et al. Mutual regulation of tumour vessel normalization and immunostimulatory reprogramming. Nature. 2017;544(7649):250–4.PubMedPubMedCentralCrossRef Tian L, Goldstein A, Wang H, Ching Lo H, Sun Kim I, Welte T, et al. Mutual regulation of tumour vessel normalization and immunostimulatory reprogramming. Nature. 2017;544(7649):250–4.PubMedPubMedCentralCrossRef
56.
Zurück zum Zitat Reck M, Mok TSK, Nishio M, Jotte RM, Cappuzzo F, Orlandi F, et al. Atezolizumab plus bevacizumab and chemotherapy in non-small-cell lung cancer (IMpower150): key subgroup analyses of patients with EGFR mutations or baseline liver metastases in a randomised, open-label phase 3 trial. Lancet Respir Med. 2019;7(5):387–401.PubMedCrossRef Reck M, Mok TSK, Nishio M, Jotte RM, Cappuzzo F, Orlandi F, et al. Atezolizumab plus bevacizumab and chemotherapy in non-small-cell lung cancer (IMpower150): key subgroup analyses of patients with EGFR mutations or baseline liver metastases in a randomised, open-label phase 3 trial. Lancet Respir Med. 2019;7(5):387–401.PubMedCrossRef
58.
Zurück zum Zitat Owada-Ozaki Y, Muto S, Takagi H, Inoue T, Watanabe Y, Fukuhara M, et al. Prognostic impact of tumor mutation burden in patients with completely resected non-small cell lung cancer: brief report. J Thorac Oncol. 2018;13(8):1217–21.PubMedCrossRef Owada-Ozaki Y, Muto S, Takagi H, Inoue T, Watanabe Y, Fukuhara M, et al. Prognostic impact of tumor mutation burden in patients with completely resected non-small cell lung cancer: brief report. J Thorac Oncol. 2018;13(8):1217–21.PubMedCrossRef
Metadaten
Titel
Immune and stromal scoring system associated with tumor microenvironment and prognosis: a gene-based multi-cancer analysis
verfasst von
Zihang Zeng
Jiali Li
Jianguo Zhang
Yangyi Li
Xingyu Liu
Jiarui Chen
Zhengrong Huang
Qiuji Wu
Yan Gong
Conghua Xie
Publikationsdatum
01.12.2021
Verlag
BioMed Central
Erschienen in
Journal of Translational Medicine / Ausgabe 1/2021
Elektronische ISSN: 1479-5876
DOI
https://doi.org/10.1186/s12967-021-03002-1

Weitere Artikel der Ausgabe 1/2021

Journal of Translational Medicine 1/2021 Zur Ausgabe

Leitlinien kompakt für die Innere Medizin

Mit medbee Pocketcards sicher entscheiden.

Seit 2022 gehört die medbee GmbH zum Springer Medizin Verlag

Ist Fasten vor Koronarinterventionen wirklich nötig?

Wenn Eingriffe wie eine Koronarangiografie oder eine Koronarangioplastie anstehen, wird häufig empfohlen, in den Stunden zuvor nüchtern zu bleiben. Ein französisches Forscherteam hat diese Maßnahme hinterfragt.

Typ-2-Diabetes: Ernährungsunsicherheit vervierfacht Risiko für schwere Hypoglykämien

04.06.2024 Typ-2-Diabetes Nachrichten

Wenn ältere Menschen mit Typ-2-Diabetes Schwierigkeiten beim Beschaffen und Zubereiten von Mahlzeiten haben, geht dies mit einem deutlich gesteigerten Risiko für schwere Hypoglykämien einher.

Mehr Brustkrebs, aber weniger andere gynäkologische Tumoren mit Levonorgestrel-IUS

04.06.2024 Levonorgestrel Nachrichten

Unter Frauen, die ein Levonorgestrel-freisetzendes intrauterines System (IUS) verwenden, ist die Brustkrebsrate um 13% erhöht. Dafür kommt es deutlich seltener zu Endometrium-, Zervix- und Ovarialkarzinomen.

GLP-1-Agonist Semaglutid wirkt kardio- und nephroprotektiv

03.06.2024 Semaglutid Nachrichten

Der GLP-1-Agonist Semaglutid hat in der FLOW-Studie bewiesen, dass sich damit die Progression chronischer Nierenerkrankungen bei Patienten mit Typ-2-Diabetes bremsen lässt. Auch in kardiovaskulärer Hinsicht war die Therapie erfolgreich.

Update Innere Medizin

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