Skip to main content
Erschienen in: BMC Cancer 1/2022

Open Access 01.12.2022 | Research

Co-deficiency of B7-H3 and B7-H4 identifies high CD8 + T cell infiltration and better prognosis in pancreatic cancer

verfasst von: Shuping Si, Lei Wang, Hui Cao, Yuhua Xu, Qiang Zhan

Erschienen in: BMC Cancer | Ausgabe 1/2022

Abstract

Background

Immunotherapy is a novel hotspot for the treatment of pancreatic adenocarcinoma (PAAD). However, potential biomarkers which could identify the inflamed tumor microenvironment (TME) are urgently required.

Methods

In the present study, we measured the levels of B7-H3, B7-H4, and major tumor-infiltrating immune cells (TIICs) using bioinformatics analyses and immunohistochemistry (IHC) staining on PAAD samples represented in the tissue microarray (TMA) format. Statistical analysis and figures exhibition were performed using R 4.1.0, SPSS 26.0, and GraphPad Prism 6.0.

Results

B7-H3 and B7-H4 were up-regulated in PAAD compared with para-tumor tissues, and their expression exhibited no tight correlation in PAAD tissues. B7-H3 and B7-H4 were lowly expressed in well-differentiated PAAD tissues and correlated with poorly differentiated grades. Besides, single B7-H3 or B7-H4 expression exhibited limited prognostic value, but co-deficiency of B7-H3 and B7-H4 predicted a better prognosis in PAAD. Moreover, co-deficiency of B7-H3 and B7-H4 indicated immuno-hot tumors with high CD8 + T cell infiltration.

Conclusions

Overall, combined B7-H3 and B7-H4 expression is a promising stratification strategy to assess prognosis and immunogenicity in PAAD, which could be used as a novel classifier in clinical practice.
Hinweise

Supplementary Information

The online version contains supplementary material available at https://​doi.​org/​10.​1186/​s12885-022-09294-w.

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Abkürzungen
PAAD
Pancreatic adenocarcinoma
TME
Tumor microenvironment
TIICs
Tumor-infiltrating immune cells
PD-L1
Programmed cell death ligand 1
RNA-seq
RNA-sequencing
TCGA
The Cancer Genome Atlas
TMA
Tissue microarray
IHC
Immunohistochemistry
DAB
Diaminobenzidine
OS
Overall survival
PFS
Progression-free survival
APC
Antigen-presenting cells

Background

Pancreatic adenocarcinoma (PAAD) is one of the most fatal malignant tumors in the world, featured with dreadful invasiveness, powerful proliferative potential, and poor clinical outcome. The early diagnosis of PAAD is rare on account of the obscure symptoms, and the morbidity of PAAD has been significantly elevated over the last few decades. Although PAAD does not account for a high proportion of all patients, survival is lowest for cancers of the pancreas (10%) [1]. With the rapid progress of emerging therapeutic programs, immunotherapy is becoming a promising hotspot for the treatment of PAAD [2]. It has been revealed that the response for immunotherapy is low in PAAD due to its non-inflamed tumor microenvironment (TME) [35]. Growing evidence indicates that tumor progression and therapeutic response were critically affected by host immune response, which depends on the abundance of tumor-infiltrating immune cells (TIICs) in TME [6, 7]. Thus, potential biomarkers which could identify the abundance of TIICs in TME of PAAD are urgently required in clinical practice.
In recent years, the roles of co-stimulatory B7 family molecules in regulating tumor immunity have been widely concerned, specially programmed cell death ligand 1 (PD-L1), also named as B7-H1 [8]. PD-L1 expression is usually correlated with inflamed TME phenotype and predicts a high response rate to immunotherapy in the clinic [9, 10]. In addition to B7-H1, B7-H3 and B7-H4 are becoming promising hotspots [11]. According to previous reports, B7-H3 and B7-H4 are significantly up-regulated in PAAD tissues compared with non-tumor or normal pancreas tissues [12, 13]. Besides, co-expressed or mutually-exclusive patterns of B7 molecules predict inflamed or non-inflamed TME in multiple human cancers [14, 15]. However, the correlation between B7-H3 and B7-H4 expression and TIICs abundance as well as the predictive value of combined B7-H3 and B7-H4 in assessing prognosis has not been investigated yet.
In this research, we first analyzed the expression of B7-H3 and B7-H4 as well as their associations between clinic-pathological features in PAAD. Besides, the prognostic values and immuno-correlations of B7-H3, B7-H4, and combined expression were also evaluated. As result, we found that B7-H3 and B7-H4 were upregulated in PAAD tissues and correlated with advanced differentiated grades. Moreover, co-deficiency of B7-H3 and B7-H4 in PAAD predicted better clinical outcomes and identifies high CD8 + T cell infiltration. Overall, co-deficiency of B7-H3 and B7-H4 is a promising prognostic and immunogenic biomarker in PAAD.

Methods

Acquisition of TCGA data

Normalized RNA-sequencing (RNA-seq) data and corresponding clinical information of PAAD samples in the Cancer Genome Atlas (TCGA) database were downloaded from the UCSC Xena website (https://​xenabrowser.​net/​datapages/​). Patients with missing or insufficient data were excluded from this research. Finally, a total of 178 tumor samples were retained for further analysis.

Analyses of the GEPIA and CPTAC databases

GEPIA (http://​gepia.​cancer-pku.​cn/​) was an interactive website based on the TCGA and GTEx databases and used for RNA expression analyses [16]. In the present study, the GEPIA website was used to explore the expression levels of B7-H3 and B7-H4 in PAAD and adjacent pancreas tissues. In addition, to further compare the differential expressions of B7-H3 and B7-H4 at protein levels, the proteome data of the CPTAC dataset (http://​ualcan.​path.​uab.​edu/​analysis-prot.​html) were also used for differential analyses of B7-H3 and B7-H4 [17].

Immune infiltration analysis

Tumor Immune Estimation Resource (TIMER) database is an online tool for systematic analysis of immune cell infiltration across diverse cancer types from TCGA [18]. We evaluated the correlation of B7-H3 & B7-H4 expressions with the infiltration of main types of immune cells, including B cells, CD8 + T cells, CD4 + T cells, neutrophils, macrophages, and dendritic cells (DCs).
The relative abundance of more types of infiltrating immune cells was analyzed using the xCell algorithm (https://​xcell.​ucsf.​edu/​), an emerging tool to estimate the abundance of 64 immune and stromal cell types based on gene expression profiles [19]. Pre-calculated infiltrating data of TIICs corresponding to TCGA-PAAD samples were downloaded from the xCell website.

Clinical samples

The PAAD tissue microarray (TMA, Cat. no HPanA150CS04) was purchased from Outdo BioTech (Shanghai, China). A total of 120 PAAD and 30 paired para-tumor tissues were included in the TMA. Detailed clinic-pathological characteristics of these patients were also provided by Outdo BioTech. Ethical approval for the use of the TMA was granted by the Clinical Research Ethics Committee (Outdo BioTech).

Immunohistochemistry

Immunohistochemistry (IHC) staining was performed on the TMA of PAAD tissues. The primary antibodies used in the research were as follows: anti-B7-H3 (1:8000 dilution, Cat. no ab219648, Abcam, Cambridge, UK), anti-B7-H4 (1:50 dilution, Cat. no ab252438, Abcam, Cambridge, UK), and anti-CD8 (Ready-to-use, Cat. no PA067, Abcarta, Suzhou, China). Antibody staining was visualized using diaminobenzidine (DAB) and hematoxylin counterstain, and stained TMA was scanned using Aperio Digital Pathology Slide Scanners.

Semi-quantitative assessment

A total of 104 TMA points were retained for further analysis after the exfoliated points were removed. All stained points were independently assessed by two senior pathologists. For semi-quantitative evaluation of B7-H3 and B7-H4 staining, the percentage of positively stained tumor cells was scored as 0–4: 0 (< 1%), 1 (1–5%), 2 (6–25%), 3 (26–50%) and 4 (> 50%). The staining intensity was scored as 0–3: 0 (negative), 1 (weak), 2 (moderate) and 3 (strong). The immune-reactivity score (IRS) equals the percentages of positive cells multiplied with staining intensity. For semi-quantitative evaluation of CD8 staining, the infiltration level of CD8 + immune cells was evaluated by estimating the percentage of cells with strong intensity of membrane staining in the stroma cells [20].

Statistical analysis

Statistical analysis and figures exhibition were performed using R 4.1.0, SPSS 26.0, and GraphPad Prism 6.0. Most of the data were analyzed by Student’s t-test, Mann–Whitney test, and one-way ANOVA. Kaplan–Meier survival plots were generated with survival curves compared by log-rank test. The Chi-square test was used to assess differences in clinic-pathological features between groups with different risks. For all analyses, differences were deemed statistically significant when P-value was less than or equal 0.05.

Results

B7-H3 and B7-H4 are up-regulated in PAAD compared with para-tumor tissues

As described previously, several research groups reported that B7-H3 and B7-H4 are up-regulated in multiple cancers [21, 22]. In the GEPIA and CPTAC databases, B7-H3 was upregulated in PAAD tissues, while B7-H4 showed no difference between tumor and para-tumor tissues (Figure S1A-D). We also assessed B7-H3 and B7-H4 expression based on IHC staining. As shown in Fig. 1A, the immuno-reactivity of B7-H3 was mostly localized to the cytomembrane of tumor cells and tumor stroma. After the semi-quantitative analysis, we found that the IRS of B7-H3 in PAAD tissues was significantly higher than para-cancerous tissues (Fig. 1B). Similar to B7-H3, the immuno-reactivity of B7-H4 was also localized to the cytomembrane of tumor cells and but not stroma (Fig. 1C). Besides, the expression of B7-H4 was notably up-regulated in PAAD tissues compared with para-cancerous tissues (Fig. 1D). We also evaluated the correlation between B7-H3 and B7-H4 expression, and the results showed that the protein expression of B7-H3 and B7-H4 had no obvious correlation (Fig. 1E). However, in the TCGA database, B7-H3 mRNA was positively correlated with B7-H4 mRNA (Fig. 1F). Overall, these data suggest that the expression of B7-H3 and B7-H4 proteins are up-regulated in PAAD tissues and have no notable correlation.

B7-H3 and B7-H4 are lowly expressed in well-differentiated PAAD tissues

Next, the associations between clinic-pathological features and B7 molecules expression were evaluated in the current patients’ cohort. As shown in Table 1, the expression levels of B7-H3 and B7-H4 were not associated with gender, age, T stage, N stage, M stage, and clinical stage. However, these two B7 molecules were significantly associated with differentiation (Table 1). We next compared the expression levels of B7-H3 and B7-H4 in well-differentiated and moderate & poor-differentiated groups, and the results exhibited that B7-H3 and B7-H4 were notably downregulated in well-differentiated PAAD tissues (Fig. 2A-D). Besides, in the TCGA database, B7-H3 was significantly correlated with advanced differentiated grades (Fig. 2E). Although B7-H4 tended to be upregulated with advanced differentiated grades, the difference was not statistically significant (Fig. 2F). Overall, deficiency of B7-H3 and/or B7-H4 identifies well-differentiated tumors in PAAD.
Table 1
Association between B7-H3 & B7-H4 expression and clinic-pathological parameters in PAAD
Clinic-pathological parameters
Cases
B7-H3 expression
χ2 value
P value
B7-H4 expression
χ2 value
P value
Low
High
Low
High
Gender
 Female
46
24
22
0.156
0.693
24
22
0.698
0.404
 Male
58
28
30
35
23
Age
  ≤ 60
47
25
22
0.349
0.554
22
25
3.439
0.064
  > 60
57
27
30
37
20
Differentiation
 Well
65
39
26
5.455
0.020
42
23
6.292
0.012
 Moderate & poor
34
12
22
13
21
T stage
 T1-2
29
17
12
0.829
0.363
19
10
1.466
0.226
 T3-4
56
27
29
29
27
N stage
 N0
48
25
23
0.157
0.692
28
20
0.009
0.925
 N1
54
26
28
 
31
23
M stage
 M0
98
51
47
/
0.205*
58
40
/
0.083*
 M1
6
1
5
1
5
Clinical stage
 1-2A
46
25
21
0.624
0.430
27
19
0.130
0.719
 2B-4
58
27
31
32
26
Note: *P value was calculated by Fisher test

Correlations between B7-H3 & B7-H4 and infiltration of main types of immune cells

Given B7-H3 & B7-H4 were correlated with TIICs in other cancers [23, 24], we also assessed the correlations between B7-H3 & B7-H4 and infiltration of main types of immune cells. B7-H3 was positively correlated with CD8 + T cells, CD4 + T cells, neutrophils, macrophages, and DCs, while B7-H4 was only positively correlated with CD8 + T cells (Fig. 3A-B). To validate the results, we performed IHC staining using anti-CD8 antibody. However, neither B7-H3 nor B7-H4 was correlated with CD8 + T cell infiltration (Fig. 3C-D). Thus, the correlations between B7-H3 & B7-H4 and immune cells infiltration are contradictory and need to be further confirmed.

Co-deficiency of B7-H3 and B7-H4 predicts a better prognosis

We further definite the prognostic values of these two B7 molecules in patients with PAAD. Patients in the TCGA cohort were divided into low (n = 89) and high (n = 89) groups at the cut-off value of the median expression. The Kaplan–Meier curves exhibited B7-H3 and B7-H4 could not effectively predict overall survival (OS) in patients with PAAD (Fig. 4A, C). In term of progression-free survival (PFS), patients with high B7-H3 expression had a significantly worse prognosis than those with low expression (Fig. 4B). However, B7-H4 could not effectively predict PFS in PAAD patients (Fig. 4D). Furthermore, combined B7-H3 and B7-H4 expression was a promising prognostic biomarker. Co-deficiency of B7-H3 and B7-H4 predicted better prognosis in terms of both OS and PFS (Fig. 4E-F) in PAAD. Taken together, these results indicated that co-deficiency of B7-H3 and B7-H4 was a favorable prognostic factor in PAAD patients.

Co-deficiency of B7-H3 and B7-H4 indicates high CD8 + T cell infiltration

Given co-expressed or mutually-exclusive patterns of B7 molecules predict inflamed or non-inflamed TME in multiple human cancers [14, 15], we next assess whether co-deficiency of B7-H3 and B7-H4 predicted specific TME features. The xCell tool was used to estimate the abundance of 64 immune and stromal cell types in the TCGA database, and the abundance of these cells in the co-low, single-high and co-high groups were next compared. A subset of non-tumor cells was different in the three groups, and total CD8 + T cells and CD8 + Tcm cells were increased in the co-low groups (Table 2, Fig. 5A-B). As expected, the infiltrating abundance of CD8 + T cell was highest in the co-low group among these three groups (Fig. 5C-D). Overall, co-deficiency of B7-H3 and B7-H4 predicts high CD8 + T cell infiltration, which may explain the better prognosis in the co-low group of PAAD patients.
Table 2
Differences of immune cells levels estimated by xCell algorithm
Immune cells
Average
F value
P value
co-low
single-high
co-high
aDC
0.080
0.108
0.097
2.582
0.079
Adipocytes
0.108
0.076
0.031
3.456
0.034
Astrocytes
0.040
0.087
0.106
13.338
0.000
B cells
0.108
0.111
0.062
0.538
0.585
Basophils
0.038
0.012
0.007
6.142
0.003
CD4 + memory T cells
0.137
0.125
0.107
1.028
0.360
CD4 + naïve T cells
0.060
0.064
0.045
0.577
0.563
CD4 + T cells
0.004
0.002
0.000
0.622
0.538
CD4 + Tcm
0.006
0.004
0.003
2.194
0.115
CD4 + Tem
0.014
0.011
0.011
0.850
0.429
CD8 + naïve T cells
0.008
0.009
0.007
1.943
0.146
CD8 + T cells
0.034
0.015
0.011
10.264
0.000
CD8 + Tcm
0.022
0.014
0.010
3.071
0.049
CD8 + Tem
0.003
0.002
0.000
2.196
0.114
cDC
0.026
0.022
0.019
0.701
0.498
Chondrocytes
0.126
0.161
0.163
4.418
0.013
Class switched memory B cells
0.034
0.025
0.020
1.493
0.228
CLP
0.048
0.042
0.044
1.146
0.320
CMP
0.000
0.000
0.000
1.071
0.345
DC
0.060
0.071
0.056
1.160
0.316
Endothelial cells
0.142
0.143
0.145
0.010
0.990
Eosinophils
0.030
0.036
0.038
3.502
0.032
Epithelial cells
0.726
0.891
0.918
7.364
0.001
Erythrocytes
0.000
0.000
0.000
0.943
0.391
Fibroblasts
0.121
0.138
0.137
0.559
0.573
GMP
0.004
0.002
0.000
2.493
0.086
Hepatocytes
0.085
0.090
0.098
0.516
0.598
HSC
0.196
0.221
0.215
1.009
0.367
iDC
0.119
0.131
0.114
0.591
0.555
Keratinocytes
0.092
0.137
0.142
5.336
0.006
Ly Endothelial cells
0.154
0.157
0.154
0.016
0.984
Macrophages
0.054
0.074
0.072
2.933
0.056
Macrophages M1
0.046
0.065
0.065
5.435
0.005
Macrophages M2
0.015
0.017
0.013
1.055
0.350
Mast cells
0.032
0.029
0.024
3.281
0.040
Megakaryocytes
0.014
0.014
0.012
1.286
0.279
Melanocytes
0.013
0.012
0.011
0.447
0.640
Memory B cells
0.015
0.016
0.009
0.305
0.738
MEP
0.035
0.030
0.027
1.792
0.170
Mesangial cells
0.081
0.093
0.106
8.969
0.000
Monocytes
0.021
0.033
0.021
2.337
0.100
MPP
0.000
0.000
0.000
0.056
0.945
MSC
0.140
0.265
0.310
28.875
0.000
Mv Endothelial cells
0.052
0.057
0.059
0.483
0.618
Myocytes
0.006
0.005
0.004
1.011
0.366
Naïve B cells
0.010
0.013
0.007
0.398
0.672
Neurons
0.066
0.015
0.014
17.406
0.000
Neutrophils
0.001
0.001
0.001
0.094
0.910
NK cells
0.000
0.000
0.000
0.458
0.633
NKT
0.024
0.022
0.026
0.959
0.385
Osteoblast
0.077
0.036
0.034
10.543
0.000
pDC
0.072
0.058
0.046
1.981
0.141
Pericytes
0.074
0.099
0.115
3.779
0.025
Plasma cells
0.011
0.008
0.005
6.987
0.001
Platelets
0.001
0.000
0.000
7.379
0.001
Preadipocytes
0.009
0.002
0.001
3.916
0.022
Pro B cells
0.002
0.002
0.001
0.319
0.727
Sebocytes
0.408
0.533
0.534
4.005
0.020
Skeletal muscle
0.000
0.000
0.000
2.332
0.100
Smooth muscle
0.386
0.332
0.333
12.517
0.000
Tgd cells
0.000
0.000
0.000
0.263
0.769
Th1 cells
0.058
0.046
0.047
1.671
0.191
Th2 cells
0.009
0.010
0.009
0.041
0.960
Tregs
0.016
0.011
0.009
3.210
0.043
Abbreviations: aDC Activated dendritic cells, CD4 + Tcm, CD4 + central memory T-cells, CD4 + Tem CD4 + effector memory T-cells, CD8 + Tcm CD8 + central memory T-cells, CD8 + Tem CD8 + effector memory T-cells, cDC Xonventional dendritic cells, CLP Common lymphoid progenitors, CMP Common myeloid progenitors, DC Dendritic cells, GMP Granulocyte–macrophage progenitors, HSC Hematopoietic stem cells, iDC Immature dendritic cells, ly endothelial cells Lymphatic endothelial cells, MEP Megakaryocyte–erythroid progenitors, MPP Multipotent rogenitors, MSC Mesenchymal stem cells, mv endothelial cells Microvascular endothelial cells, NKT Natural killer T-cells, pDC Plasmacytoid dendritic cells, Tgd cells Gamma delta T-cells, Th1 cells Type 1 T-helper cells, Th2 cells Type 2 T-helper cells, Tregs Regulatory T-cells

Discussion

It has been proved that increased CD8 + T cell infiltration is one of the notable features of immuno-hot tumors, which indicates a better prognosis and high therapeutic response [2527]. Thus, reliable biomarkers for the identification of immuno-hot tumors in PAAD are urgent in clinical practice. In the current research, we analyzed the expression patterns of B7-H3 and B7-H4 in PAAD and combined their expression as a novel stratification strategy. We found that B7-H3 and B7-H4 were highly expressed in PAAD tissues and higher in poorly differentiated tumors. Moreover, co-deficiency of B7-H3 and B7-H4 indicates a better prognosis and high CD8 + T cell infiltration.
B7-H3 is a negative regulator and inhibits T cell proliferation and cytokine production mediated by antibody to CD3 [28]. In cancers, B7-H3 acts as an inhibitory immune checkpoint that negatively regulates anti-tumor immunity. Overexpression of B7-H3 in tumor tissues is a poor prognostic biomarker in prostate cancer [29], upper tract urothelial carcinoma [30], small cell lung cancer [31], etc. Besides, inhibition of B7-H3 expression is a promising therapeutic strategy for human cancer. In previous research, B7-H3 targeted therapies have been mentioned, which shows promising applications, including monoclonal antibodies against B7-H3, specific antibody-dependent cell-mediated cytotoxicity, antibody drug conjugates, specific small-molecule inhibitor, and chimeric antigen receptor T-cell therapy [21]. B7-H3 expression shows no notable correlation with major TIICs, including CD3 + , CD8 + and CD20 + TIICs in small cell lung cancer [32], whereas B7-H3 expression is positively correlated with the abundance of CD45 + and CD8 + TIICs in non-small-cell lung cancer [33]. In PAAD, B7-H3 was overexpressed and promoted tumor progression [34]. In addition, tumor high B7-H3 expression was independently associated with poor survival [35]. In our research, B7-H3 was positively correlated with CD8 + T cells, CD4 + T cells, neutrophils, macrophages, and DCs estimated by TIMER algorithm, but B7-H3 expression was not correlated with the abundance of CD8 + TIICs in our validated cohort. The contradictory results need to be further confirmed.
Similar to B7-H3, B7-H4 is also an inhibitory immune checkpoint and predicts poor prognosis in multiple human cancers [3638]. Besides, immunotherapy targeting B7-H4 is under pre-clinical investigation [22]. For example, pharmacologic inhibition of B7-H4 glycosylation restores anti-tumor immunity in immuno-cold breast cancer [39]. It has been reported that B7-H4 expression is inversely correlated with T cell infiltration in clear cell ovarian cancer [40] and breast cancer [24]. In PAAD, B7-H4 promoted cancer progression and inhibited apoptosis in PAAD cells [41]. In addition, a meta-analysis revealed that high expression of B7-H4 was an unfavorable prognostic factor for patients with PAAD [42]. In the current research, we assessed the expression of B7-H4 in tumor and para-tumor tissues in PAAD. However, the results revealed by the GEPIA and CPTAC databases showed no difference between tumor and para-tumor tissues, but IHC staining uncovered that B7-H4 was significantly overexpressed in PAAD tissues. Since B7-H4 was almost only expressed in tumor cells and not in the tumor stroma, we speculated that bulk-RNA sequencing could not distinguish cell subtypes, leading to the false low expression in tumor tissues.
Interestingly, growing numbers of studies have suggested that B7 molecules exhibit limited co-expression patterns [15, 32, 43]. B7-H3 and B7-H4 exhibited no tight correlation in PAAD in our research, but no obvious pattern of mutually exclusive expression was observed as well. Novel prognostic and/or immunogenic classifiers based on different expression patterns of B7 molecules have been preliminarily investigated. For example, B7-H4 is negatively correlated with PD-L1 and identifies immuno-cold tumors in glioma [15]. In our research, we found that co-deficiency of B7-H3 and B7-H4 indicates better prognosis and immuno-hot tumors with high CD8 + T cell infiltration, which could be applied as a novel classifier for prognostic and immunogenic assessment in PAAD.

Conclusion

To sum up, we analyze the expression patterns and prognostic values of B7-H3 and B7-H4 in PAAD. Single B7-H3 or B7-H4 expression exhibits limited prognostic value for assessment of clinical outcome in PAAD, while combined their expression is a promising stratification strategy to evaluate prognosis and immunogenicity in PAAD.

Acknowledgements

Not applicable.

Declarations

Ethical approval for the use of the TMA was granted by the Clinical Research Ethics Committee (Outdo BioTech). Informed consent was obtained from all subjects and/or their legal guardian(s). In addition, all methods were performed in accordance with the relevant guidelines and regulations.
Not applicable.

Competing interests

The authors declare no conflict of interest.
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.
Literatur
1.
Zurück zum Zitat Siegel RL, Miller KD, Fuchs HE, Jemal A. Cancer Statistics, 2021. CA Cancer J Clin. 2021;71(1):7–33.CrossRef Siegel RL, Miller KD, Fuchs HE, Jemal A. Cancer Statistics, 2021. CA Cancer J Clin. 2021;71(1):7–33.CrossRef
2.
Zurück zum Zitat Foley K, Kim V, Jaffee E, Zheng L. Current progress in immunotherapy for pancreatic cancer. Cancer Lett. 2016;381(1):244–51.CrossRef Foley K, Kim V, Jaffee E, Zheng L. Current progress in immunotherapy for pancreatic cancer. Cancer Lett. 2016;381(1):244–51.CrossRef
3.
Zurück zum Zitat Hilmi M, Bartholin L, Neuzillet C. Immune therapies in pancreatic ductal adenocarcinoma: Where are we now? World J Gastroenterol. 2018;24(20):2137–51.CrossRef Hilmi M, Bartholin L, Neuzillet C. Immune therapies in pancreatic ductal adenocarcinoma: Where are we now? World J Gastroenterol. 2018;24(20):2137–51.CrossRef
4.
Zurück zum Zitat Banerjee K, Kumar S, Ross KA, Gautam S, Poelaert B, Nasser MW, Aithal A, Bhatia R, Wannemuehler MJ, Narasimhan B, et al. Emerging trends in the immunotherapy of pancreatic cancer. Cancer Lett. 2018;417:35–46.CrossRef Banerjee K, Kumar S, Ross KA, Gautam S, Poelaert B, Nasser MW, Aithal A, Bhatia R, Wannemuehler MJ, Narasimhan B, et al. Emerging trends in the immunotherapy of pancreatic cancer. Cancer Lett. 2018;417:35–46.CrossRef
5.
Zurück zum Zitat Schizas D, Charalampakis N, Kole C, Economopoulou P, Koustas E, Gkotsis E, Ziogas D, Psyrri A, Karamouzis MV. Immunotherapy for pancreatic cancer: A 2020 update. Cancer Treat Rev. 2020;86:102016.CrossRef Schizas D, Charalampakis N, Kole C, Economopoulou P, Koustas E, Gkotsis E, Ziogas D, Psyrri A, Karamouzis MV. Immunotherapy for pancreatic cancer: A 2020 update. Cancer Treat Rev. 2020;86:102016.CrossRef
6.
Zurück zum Zitat Tahkola K, Mecklin JP, Wirta EV, Ahtiainen M, Helminen O, Bohm J, Kellokumpu I. High immune cell score predicts improved survival in pancreatic cancer. Virchows Arch. 2018;472(4):653–65.CrossRef Tahkola K, Mecklin JP, Wirta EV, Ahtiainen M, Helminen O, Bohm J, Kellokumpu I. High immune cell score predicts improved survival in pancreatic cancer. Virchows Arch. 2018;472(4):653–65.CrossRef
7.
Zurück zum Zitat Stenzel PJ, Schindeldecker M, Tagscherer KE, Foersch S, Herpel E, Hohenfellner M, Hatiboglu G, Alt J, Thomas C, Haferkamp A, et al. Prognostic and Predictive Value of Tumor-infiltrating Leukocytes and of Immune Checkpoint Molecules PD1 and PDL1 in Clear Cell Renal Cell Carcinoma. Transl Oncol. 2020;13(2):336–45.CrossRef Stenzel PJ, Schindeldecker M, Tagscherer KE, Foersch S, Herpel E, Hohenfellner M, Hatiboglu G, Alt J, Thomas C, Haferkamp A, et al. Prognostic and Predictive Value of Tumor-infiltrating Leukocytes and of Immune Checkpoint Molecules PD1 and PDL1 in Clear Cell Renal Cell Carcinoma. Transl Oncol. 2020;13(2):336–45.CrossRef
8.
Zurück zum Zitat Leung J, Suh WK. The CD28-B7 Family in Anti-Tumor Immunity: Emerging Concepts in Cancer Immunotherapy. Immune Netw. 2014;14(6):265–76.CrossRef Leung J, Suh WK. The CD28-B7 Family in Anti-Tumor Immunity: Emerging Concepts in Cancer Immunotherapy. Immune Netw. 2014;14(6):265–76.CrossRef
9.
Zurück zum Zitat Li YM, Yu JM, Liu ZY, Yang HJ, Tang J, Chen ZN. Programmed Death Ligand 1 Indicates Pre-Existing Adaptive Immune Response by Tumor-Infiltrating CD8(+) T Cells in Non-Small Cell Lung Cancer. Int J Mol Sci. 2019;20(20):5138.CrossRef Li YM, Yu JM, Liu ZY, Yang HJ, Tang J, Chen ZN. Programmed Death Ligand 1 Indicates Pre-Existing Adaptive Immune Response by Tumor-Infiltrating CD8(+) T Cells in Non-Small Cell Lung Cancer. Int J Mol Sci. 2019;20(20):5138.CrossRef
10.
Zurück zum Zitat Wei XL, Liu QW, Liu FR, Yuan SS, Li XF, Li JN, Yang AL, Ling YH. The clinicopathological significance and predictive value for immunotherapy of programmed death ligand-1 expression in Epstein-Barr virus-associated gastric cancer. Oncoimmunology. 2021;10(1):1938381.CrossRef Wei XL, Liu QW, Liu FR, Yuan SS, Li XF, Li JN, Yang AL, Ling YH. The clinicopathological significance and predictive value for immunotherapy of programmed death ligand-1 expression in Epstein-Barr virus-associated gastric cancer. Oncoimmunology. 2021;10(1):1938381.CrossRef
11.
Zurück zum Zitat Ni L, Dong C. New B7 Family Checkpoints in Human Cancers. Mol Cancer Ther. 2017;16(7):1203–11.CrossRef Ni L, Dong C. New B7 Family Checkpoints in Human Cancers. Mol Cancer Ther. 2017;16(7):1203–11.CrossRef
12.
Zurück zum Zitat Yamato I, Sho M, Nomi T, Akahori T, Shimada K, Hotta K, Kanehiro H, Konishi N, Yagita H, Nakajima Y. Clinical importance of B7–H3 expression in human pancreatic cancer. Br J Cancer. 2009;101(10):1709–16.CrossRef Yamato I, Sho M, Nomi T, Akahori T, Shimada K, Hotta K, Kanehiro H, Konishi N, Yagita H, Nakajima Y. Clinical importance of B7–H3 expression in human pancreatic cancer. Br J Cancer. 2009;101(10):1709–16.CrossRef
13.
Zurück zum Zitat Shen L, Qian Y, Wu W, Weng T, Wang FXC, Hong B, Wu Z, Wang Q, Sang Y, Zhang H, et al. B7–H4 is a prognostic biomarker for poor survival in patients with pancreatic cancer. Hum Pathol. 2017;66:79–85.CrossRef Shen L, Qian Y, Wu W, Weng T, Wang FXC, Hong B, Wu Z, Wang Q, Sang Y, Zhang H, et al. B7–H4 is a prognostic biomarker for poor survival in patients with pancreatic cancer. Hum Pathol. 2017;66:79–85.CrossRef
14.
Zurück zum Zitat Cherif B, Triki H, Charfi S, Bouzidi L, Kridis WB, Khanfir A, Chaabane K, Sellami-Boudawara T, Rebai A. Immune checkpoint molecules B7–H6 and PD-L1 co-pattern the tumor inflammatory microenvironment in human breast cancer. Sci Rep. 2021;11(1):7550.CrossRef Cherif B, Triki H, Charfi S, Bouzidi L, Kridis WB, Khanfir A, Chaabane K, Sellami-Boudawara T, Rebai A. Immune checkpoint molecules B7–H6 and PD-L1 co-pattern the tumor inflammatory microenvironment in human breast cancer. Sci Rep. 2021;11(1):7550.CrossRef
15.
Zurück zum Zitat Chen D, Li G, Ji C, Lu Q, Qi Y, Tang C, Xiong J, Hu J, Yasar FBA, Zhang Y, et al. Enhanced B7–H4 expression in gliomas with low PD-L1 expression identifies super-cold tumors. J Immunother Cancer. 2020;8(1):e000154.CrossRef Chen D, Li G, Ji C, Lu Q, Qi Y, Tang C, Xiong J, Hu J, Yasar FBA, Zhang Y, et al. Enhanced B7–H4 expression in gliomas with low PD-L1 expression identifies super-cold tumors. J Immunother Cancer. 2020;8(1):e000154.CrossRef
16.
Zurück zum Zitat Tang Z, Li C, Kang B, Gao G, Li C, Zhang Z. GEPIA: a web server for cancer and normal gene expression profiling and interactive analyses. Nucleic Acids Res. 2017;45(W1):W98–102.CrossRef Tang Z, Li C, Kang B, Gao G, Li C, Zhang Z. GEPIA: a web server for cancer and normal gene expression profiling and interactive analyses. Nucleic Acids Res. 2017;45(W1):W98–102.CrossRef
17.
Zurück zum Zitat Chen F, Chandrashekar DS, Varambally S, Creighton CJ. Pan-cancer molecular subtypes revealed by mass-spectrometry-based proteomic characterization of more than 500 human cancers. Nat Commun. 2019;10(1):5679.CrossRef Chen F, Chandrashekar DS, Varambally S, Creighton CJ. Pan-cancer molecular subtypes revealed by mass-spectrometry-based proteomic characterization of more than 500 human cancers. Nat Commun. 2019;10(1):5679.CrossRef
18.
Zurück zum Zitat Li T, Fan J, Wang B, Traugh N, Chen Q, Liu JS, Li B, Liu XS. TIMER: A Web Server for Comprehensive Analysis of Tumor-Infiltrating Immune Cells. Cancer Res. 2017;77(21):e108–10.CrossRef Li T, Fan J, Wang B, Traugh N, Chen Q, Liu JS, Li B, Liu XS. TIMER: A Web Server for Comprehensive Analysis of Tumor-Infiltrating Immune Cells. Cancer Res. 2017;77(21):e108–10.CrossRef
19.
Zurück zum Zitat Aran D, Hu Z, Butte AJ. xCell: digitally portraying the tissue cellular heterogeneity landscape. Genome Biol. 2017;18(1):220.CrossRef Aran D, Hu Z, Butte AJ. xCell: digitally portraying the tissue cellular heterogeneity landscape. Genome Biol. 2017;18(1):220.CrossRef
20.
Zurück zum Zitat Cai Y, Ji W, Sun C, Xu R, Chen X, Deng Y, Pan J, Yang J, Zhu H, Mei J. Interferon-Induced Transmembrane Protein 3 Shapes an Inflamed Tumor Microenvironment and Identifies Immuno-Hot Tumors. Front Immunol. 2021;12(3162):704965. Cai Y, Ji W, Sun C, Xu R, Chen X, Deng Y, Pan J, Yang J, Zhu H, Mei J. Interferon-Induced Transmembrane Protein 3 Shapes an Inflamed Tumor Microenvironment and Identifies Immuno-Hot Tumors. Front Immunol. 2021;12(3162):704965.
21.
Zurück zum Zitat Liu S, Liang J, Liu Z, Zhang C, Wang Y, Watson AH, Zhou C, Zhang F, Wu K, Zhang F, et al. The role of CD276 in Cancers. Front Oncol. 2021;11:654684.CrossRef Liu S, Liang J, Liu Z, Zhang C, Wang Y, Watson AH, Zhou C, Zhang F, Wu K, Zhang F, et al. The role of CD276 in Cancers. Front Oncol. 2021;11:654684.CrossRef
22.
Zurück zum Zitat Wang JY, Wang WP. B7-H4, a promising target for immunotherapy. Cell Immunol. 2020;347:104008. Wang JY, Wang WP. B7-H4, a promising target for immunotherapy. Cell Immunol. 2020;347:104008.
23.
Zurück zum Zitat Fauci JM, Straughn JM Jr, Ferrone S, Buchsbaum DJ. A review of B7–H3 and B7–H4 immune molecules and their role in ovarian cancer. Gynecol Oncol. 2012;127(2):420–5.CrossRef Fauci JM, Straughn JM Jr, Ferrone S, Buchsbaum DJ. A review of B7–H3 and B7–H4 immune molecules and their role in ovarian cancer. Gynecol Oncol. 2012;127(2):420–5.CrossRef
24.
Zurück zum Zitat Kim NI, Park MH, Kweon SS, Lee JS. B7–H3 and B7–H4 expression in breast cancer and their association with clinicopathological variables and T Cell infiltration. Pathobiology. 2020;87(3):179–92.CrossRef Kim NI, Park MH, Kweon SS, Lee JS. B7–H3 and B7–H4 expression in breast cancer and their association with clinicopathological variables and T Cell infiltration. Pathobiology. 2020;87(3):179–92.CrossRef
25.
Zurück zum Zitat Craig SG, Humphries MP, Alderdice M, Bingham V, Richman SD, Loughrey MB, Coleman HG, Viratham-Pulsawatdi A, McCombe K, Murray GI, et al. Immune status is prognostic for poor survival in colorectal cancer patients and is associated with tumour hypoxia. Br J Cancer. 2020;123(8):1280–8.CrossRef Craig SG, Humphries MP, Alderdice M, Bingham V, Richman SD, Loughrey MB, Coleman HG, Viratham-Pulsawatdi A, McCombe K, Murray GI, et al. Immune status is prognostic for poor survival in colorectal cancer patients and is associated with tumour hypoxia. Br J Cancer. 2020;123(8):1280–8.CrossRef
26.
Zurück zum Zitat Ren F, Zhao Q, Minghai Z, Shaogong Z, Liu B, Bukhari I, Zhang K, Wu W, Yuming F, Yu Y, et al. Immune infiltration profiling in gastric cancer and their clinical implications. Cancer Sci. 2021;112(9):3569–84.CrossRef Ren F, Zhao Q, Minghai Z, Shaogong Z, Liu B, Bukhari I, Zhang K, Wu W, Yuming F, Yu Y, et al. Immune infiltration profiling in gastric cancer and their clinical implications. Cancer Sci. 2021;112(9):3569–84.CrossRef
27.
Zurück zum Zitat Hu R, Han Q, Zhang J. STAT3: A key signaling molecule for converting cold to hot tumors. Cancer Lett. 2020;489:29–40.CrossRef Hu R, Han Q, Zhang J. STAT3: A key signaling molecule for converting cold to hot tumors. Cancer Lett. 2020;489:29–40.CrossRef
28.
Zurück zum Zitat Suh WK, Gajewska BU, Okada H, Gronski MA, Bertram EM, Dawicki W, Duncan GS, Bukczynski J, Plyte S, Elia A, et al. The B7 family member B7–H3 preferentially down-regulates T helper type 1-mediated immune responses. Nat Immunol. 2003;4(9):899–906.CrossRef Suh WK, Gajewska BU, Okada H, Gronski MA, Bertram EM, Dawicki W, Duncan GS, Bukczynski J, Plyte S, Elia A, et al. The B7 family member B7–H3 preferentially down-regulates T helper type 1-mediated immune responses. Nat Immunol. 2003;4(9):899–906.CrossRef
29.
Zurück zum Zitat Nunes-Xavier CE, Kildal W, Kleppe A, Danielsen HE, Waehre H, Llarena R, Maelandsmo GM, Fodstad O, Pulido R, Lopez JI. Immune checkpoint B7–H3 protein expression is associated with poor outcome and androgen receptor status in prostate cancer. Prostate. 2021;81(12):838–48.CrossRef Nunes-Xavier CE, Kildal W, Kleppe A, Danielsen HE, Waehre H, Llarena R, Maelandsmo GM, Fodstad O, Pulido R, Lopez JI. Immune checkpoint B7–H3 protein expression is associated with poor outcome and androgen receptor status in prostate cancer. Prostate. 2021;81(12):838–48.CrossRef
30.
Zurück zum Zitat Koyama Y, Morikawa T, Miyama Y, Miyakawa J, Kawai T, Kume H, Sawabe M, Ushiku T. B7–H3 expression in upper tract urothelial carcinoma associates with adverse clinicopathological features and poor survival. Pathol Res Pract. 2020;216(12):153219.CrossRef Koyama Y, Morikawa T, Miyama Y, Miyakawa J, Kawai T, Kume H, Sawabe M, Ushiku T. B7–H3 expression in upper tract urothelial carcinoma associates with adverse clinicopathological features and poor survival. Pathol Res Pract. 2020;216(12):153219.CrossRef
31.
Zurück zum Zitat Qiu MJ, Xia Q, Chen YB, Fang XF, Li QT, Zhu LS, Jiang X, Xiong ZF, Yang SL. The expression of three negative Co-Stimulatory B7 family molecules in small cell lung cancer and their effect on prognosis. Front Oncol. 2021;11:600238.CrossRef Qiu MJ, Xia Q, Chen YB, Fang XF, Li QT, Zhu LS, Jiang X, Xiong ZF, Yang SL. The expression of three negative Co-Stimulatory B7 family molecules in small cell lung cancer and their effect on prognosis. Front Oncol. 2021;11:600238.CrossRef
32.
Zurück zum Zitat Carvajal-Hausdorf D, Altan M, Velcheti V, Gettinger SN, Herbst RS, Rimm DL, Schalper KA. Expression and clinical significance of PD-L1, B7–H3, B7–H4 and TILs in human small cell lung Cancer (SCLC). J Immunother Cancer. 2019;7(1):65.CrossRef Carvajal-Hausdorf D, Altan M, Velcheti V, Gettinger SN, Herbst RS, Rimm DL, Schalper KA. Expression and clinical significance of PD-L1, B7–H3, B7–H4 and TILs in human small cell lung Cancer (SCLC). J Immunother Cancer. 2019;7(1):65.CrossRef
33.
Zurück zum Zitat Yim J, Koh J, Kim S, Song SG, Ahn HK, Kim YA, Jeon YK, Chung DH. Effects of B7–H3 expression on tumour-infiltrating immune cells and clinicopathological characteristics in non-small-cell lung cancer. Eur J Cancer. 2020;133:74–85.CrossRef Yim J, Koh J, Kim S, Song SG, Ahn HK, Kim YA, Jeon YK, Chung DH. Effects of B7–H3 expression on tumour-infiltrating immune cells and clinicopathological characteristics in non-small-cell lung cancer. Eur J Cancer. 2020;133:74–85.CrossRef
34.
Zurück zum Zitat Zhao X, Li DC, Zhu XG, Gan WJ, Li Z, Xiong F, Zhang ZX, Zhang GB, Zhang XG, Zhao H. B7–H3 overexpression in pancreatic cancer promotes tumor progression. Int J Mol Med. 2013;31(2):283–91.CrossRef Zhao X, Li DC, Zhu XG, Gan WJ, Li Z, Xiong F, Zhang ZX, Zhang GB, Zhang XG, Zhao H. B7–H3 overexpression in pancreatic cancer promotes tumor progression. Int J Mol Med. 2013;31(2):283–91.CrossRef
35.
Zurück zum Zitat Inamura K, Takazawa Y, Inoue Y, Yokouchi Y, Kobayashi M, Saiura A, Shibutani T, Ishikawa Y. Tumor B7–H3 (CD276) Expression and Survival in Pancreatic Cancer. J Clin Med. 2018;7(7):172.CrossRef Inamura K, Takazawa Y, Inoue Y, Yokouchi Y, Kobayashi M, Saiura A, Shibutani T, Ishikawa Y. Tumor B7–H3 (CD276) Expression and Survival in Pancreatic Cancer. J Clin Med. 2018;7(7):172.CrossRef
36.
Zurück zum Zitat Niu N, Shen W, Zhong Y, Bast RC Jr, Jazaeri A, Sood AK, Liu J. Expression of B7–H4 and IDO1 is associated with drug resistance and poor prognosis in high-grade serous ovarian carcinomas. Hum Pathol. 2021;113:20–7.CrossRef Niu N, Shen W, Zhong Y, Bast RC Jr, Jazaeri A, Sood AK, Liu J. Expression of B7–H4 and IDO1 is associated with drug resistance and poor prognosis in high-grade serous ovarian carcinomas. Hum Pathol. 2021;113:20–7.CrossRef
37.
Zurück zum Zitat Mizuno T, Kamai T, Tsuzuki T, Nishihara D, Kijima T, Arai K, Yoshida KI. Elevated expression of B7 homolog 4 is associated with disease progression in upper urinary tract urothelial carcinoma. Cancer Immunol Immunother. 2021;2022(3):565–78. https://doi.org/10.1007/s00262-021-03011-5. Mizuno T, Kamai T, Tsuzuki T, Nishihara D, Kijima T, Arai K, Yoshida KI. Elevated expression of B7 homolog 4 is associated with disease progression in upper urinary tract urothelial carcinoma. Cancer Immunol Immunother. 2021;2022(3):565–78. https://​doi.​org/​10.​1007/​s00262-021-03011-5.
38.
Zurück zum Zitat Ding S, Lv X, Liu Z, Zhan S, Xu Y, Zhang X, Liu C, Cao L. Overexpression of B7–H4 is associated with infiltrating immune cells and poor prognosis in metastatic colorectal cancer. Int Immunopharmacol. 2021;90:107144.CrossRef Ding S, Lv X, Liu Z, Zhan S, Xu Y, Zhang X, Liu C, Cao L. Overexpression of B7–H4 is associated with infiltrating immune cells and poor prognosis in metastatic colorectal cancer. Int Immunopharmacol. 2021;90:107144.CrossRef
39.
Zurück zum Zitat Song X, Zhou Z, Li H, Xue Y, Lu X, Bahar I, Kepp O, Hung MC, Kroemer G, Wan Y. Pharmacologic Suppression of B7–H4 Glycosylation Restores Antitumor Immunity in Immune-Cold Breast Cancers. Cancer Discov. 2020;10(12):1872–93.CrossRef Song X, Zhou Z, Li H, Xue Y, Lu X, Bahar I, Kepp O, Hung MC, Kroemer G, Wan Y. Pharmacologic Suppression of B7–H4 Glycosylation Restores Antitumor Immunity in Immune-Cold Breast Cancers. Cancer Discov. 2020;10(12):1872–93.CrossRef
40.
Zurück zum Zitat Pagnotti GM, Atkinson RM, Romeiser J, Akalin A, Korman MB, Shroyer KR. B7–H4 is Inversely Correlated With T-Cell Infiltration in Clear Cell but Not Serous or Endometrioid Ovarian Cancer. Appl Immunohistochem Mol Morphol. 2019;27(7):515–22.CrossRef Pagnotti GM, Atkinson RM, Romeiser J, Akalin A, Korman MB, Shroyer KR. B7–H4 is Inversely Correlated With T-Cell Infiltration in Clear Cell but Not Serous or Endometrioid Ovarian Cancer. Appl Immunohistochem Mol Morphol. 2019;27(7):515–22.CrossRef
41.
Zurück zum Zitat Qian Y, Hong B, Shen L, Wu Z, Yao H, Zhang L. B7–H4 enhances oncogenicity and inhibits apoptosis in pancreatic cancer cells. Cell Tissue Res. 2013;353(1):139–51.CrossRef Qian Y, Hong B, Shen L, Wu Z, Yao H, Zhang L. B7–H4 enhances oncogenicity and inhibits apoptosis in pancreatic cancer cells. Cell Tissue Res. 2013;353(1):139–51.CrossRef
42.
Zurück zum Zitat Chen X, Tao L, Yuan C, Xiu D. The prognostic value of B7–H4 in pancreatic cancer: Systematic review and meta-analysis. Medicine (Baltimore). 2018;97(12):e0088.CrossRef Chen X, Tao L, Yuan C, Xiu D. The prognostic value of B7–H4 in pancreatic cancer: Systematic review and meta-analysis. Medicine (Baltimore). 2018;97(12):e0088.CrossRef
43.
Zurück zum Zitat Altan M, Pelekanou V, Schalper KA, Toki M, Gaule P, Syrigos K, Herbst RS, Rimm DL. B7–H3 Expression in NSCLC and Its Association with B7–H4, PD-L1 and Tumor-Infiltrating Lymphocytes. Clin Cancer Res. 2017;23(17):5202–9.CrossRef Altan M, Pelekanou V, Schalper KA, Toki M, Gaule P, Syrigos K, Herbst RS, Rimm DL. B7–H3 Expression in NSCLC and Its Association with B7–H4, PD-L1 and Tumor-Infiltrating Lymphocytes. Clin Cancer Res. 2017;23(17):5202–9.CrossRef
Metadaten
Titel
Co-deficiency of B7-H3 and B7-H4 identifies high CD8 + T cell infiltration and better prognosis in pancreatic cancer
verfasst von
Shuping Si
Lei Wang
Hui Cao
Yuhua Xu
Qiang Zhan
Publikationsdatum
01.12.2022
Verlag
BioMed Central
Erschienen in
BMC Cancer / Ausgabe 1/2022
Elektronische ISSN: 1471-2407
DOI
https://doi.org/10.1186/s12885-022-09294-w

Weitere Artikel der Ausgabe 1/2022

BMC Cancer 1/2022 Zur Ausgabe

Bei seelischem Stress sind Checkpoint-Hemmer weniger wirksam

03.06.2024 NSCLC Nachrichten

Wie stark Menschen mit fortgeschrittenem NSCLC von einer Therapie mit Immun-Checkpoint-Hemmern profitieren, hängt offenbar auch davon ab, wie sehr die Diagnose ihre psychische Verfassung erschüttert

Antikörper mobilisiert Neutrophile gegen Krebs

03.06.2024 Onkologische Immuntherapie Nachrichten

Ein bispezifischer Antikörper formiert gezielt eine Armee neutrophiler Granulozyten gegen Krebszellen. An den Antikörper gekoppeltes TNF-alpha soll die Zellen zudem tief in solide Tumoren hineinführen.

Erhebliches Risiko für Kehlkopfkrebs bei mäßiger Dysplasie

29.05.2024 Larynxkarzinom Nachrichten

Fast ein Viertel der Personen mit mäßig dysplastischen Stimmlippenläsionen entwickelt einen Kehlkopftumor. Solche Personen benötigen daher eine besonders enge ärztliche Überwachung.

15% bedauern gewählte Blasenkrebs-Therapie

29.05.2024 Urothelkarzinom Nachrichten

Ob Patienten und Patientinnen mit neu diagnostiziertem Blasenkrebs ein Jahr später Bedauern über die Therapieentscheidung empfinden, wird einer Studie aus England zufolge von der Radikalität und dem Erfolg des Eingriffs beeinflusst.

Update Onkologie

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