Skip to main content
Erschienen in: European Journal of Medical Research 1/2023

Open Access 01.12.2023 | Research

CD72, a new immune checkpoint molecule, is a novel prognostic biomarker for kidney renal clear cell carcinoma

Erschienen in: European Journal of Medical Research | Ausgabe 1/2023

Abstract

Background

The incidence and mortality of clear cell carcinoma of the kidney increases yearly. There are limited screening methods and advances in treating kidney renal clear cell carcinoma (KIRC). It is important to find new biomarkers to screen, diagnose and predict the prognosis of KIRC. Some studies have shown that CD72 influences the development and progression of colorectal cancer, nasopharyngeal cancer, and acute lymphoid leukemia. However, there is a lack of research on the role of CD72 in the pathogenesis of KIRC. This study aimed to determine whether CD72 is associated with the prognosis and immune infiltration of KIRC, providing an essential molecular basis for the early non-invasive diagnosis and immunotherapy of KIRC.

Methods

Using TCGA, GTE, GEO, and ImmPort databases, we obtained the differentially expressed mRNA (DEmRNA) associated with the prognosis and immunity of KIRC patients. We used the Kruskal–Wallis test to identify clinicopathological parameters associated with target gene expression. We performed univariate and multivariate COX regression analyses to determine the effect of target gene expression and clinicopathological parameters on survival. We analyzed the target genes' relevant functions and signaling pathways through enrichment analysis. Finally, the correlation of target genes with tumor immune infiltration was explored by ssGSEA and Spearman correlation analysis.

Results

The results revealed that patients with KIRC with higher expression of CD72 have a poorer prognosis. CD72 was associated with the Pathologic T stage, Pathologic stage, Pathologic M stage, Pathologic N stage, Histologic grade in KIRC patients, Laterality, and OS event. It was an independent predictor of the overall survival of KIRC patients. Functional enrichment analysis showed that CD72 was significantly enriched in oncogenic and immune-related pathways. According to ssGSEA and Spearman correlation analysis, CD72 expression was significantly associated with tumor immune cells and immune checkpoints.

Conclusion

Our study suggests that CD72 is associated with tumor immunity and may be a biomarker relevant to the diagnosis and prognosis of KIRC patients.
Hinweise
Lv Tian and Yiming Wang contributed equally to this work.

Publisher's Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Abkürzungen
KIRC
Kidney clear cell carcinoma
TCGA
The Cancer Genome Atlas
GEO
Gene Expression Omnibus
GO
Gene Ontology
KEGG
Kyoto Encyclopedia of Genes and Genomes
GSEA
Gene set enrichment analysis
DEGs
Differentially expressed genes
OS
Overall survival
ANOVA
One-factor analysis of variance
IHC
Immunohistochemical

Introduction

Renal cell carcinoma (RCC) is one of the ten most common cancers; 73,750 new cases of RCC and 14,830 RCC deaths were reported in the U.S. in 2020 [1]. Kidney clear cell carcinoma (KIRC) is the main pathologic subtype of RCC [2]. Despite our great advances in diagnosis, screening, surgery, and treatment, the clinical outcomes of KIRC are still unsatisfactory [3]. The prognosis for advanced KIRC is extremely poor due to its inherent resistance to radiotherapy and chemotherapy, and the challenge for clinical management lies in treating the poor prognosis caused by radiotherapy and chemotherapy resistance [4]. KIRC has an immunogenic tumor microenvironment (TME) that contains a variety of tumor-infiltrating T lymphocytes [5]. Features of the tumor microenvironment heavily affect disease biology and may affect responses to systemic therapy [69]. In recent years, advances in immunotherapy, particularly immune checkpoint blockade (ICB) and engineered T cells have revolutionized cancer treatment [10]. ICB or ICB plus TKIs targeting programmed cell death 1 (PD-1), programmed cell death ligand 1 (PD-L1), and cytotoxic T-lymphocyte-associated antigen 4 (CTLA4) are now the standard of care for RCC [11, 12]. In the era of ICB, understanding immunogenic TME will help to find new therapeutic strategies in KIRC management.
CD72 is a type II membrane protein expressed mainly in B cells and is a member of the C-type lectin superfamily [13]. CD72 contains a C-type lectin-like structural domain (CTLD) and an immunoreceptor tyrosine-based inhibitory motif (ITIM) [14, 15]. CD72 overexpression can negatively regulate BCR signaling in B cell lines by recruiting phosphatase 1 (SHP-1) of the SH2 structural domain to phosphorylated ITIM [16]. CD72 functions similarly to the inhibitory co-receptor CD22 in down-regulating B-cell receptor (BCR) signaling and functioning as a molecular switch determining whether apoptosis or proliferation occurs in antigen-stimulated B cells [14]. Furthermore, CD72 and SEMA4D (CD100) interaction enhances the activation of B cells and dendritic cells (DCs) [17]. Recent studies have shown that CD72 is closely associated with developing various immune-related diseases. For example, CD72 is strongly associated with developing systemic lupus erythematosus (SLE) [1820] in autoimmune diseases. CD72 is also closely associated with the tumor microenvironment [21]. Several studies have shown that CD72 is a marker for progenitor cell B-cell leukemia and a new marker for detecting microscopic residual disease in acute lymphoblastic leukemia [22, 23]. In addition, CD72 has been identified as a prognostic gene in the tumor microenvironment of colorectal cancer [24, 25]. CD72 may be an independent predictor of prognosis in nasopharyngeal carcinoma patients [26]. However, there are no studies on the role of CD72 in immune infiltration in KIRC. Therefore, this study aimed to investigate the relationship between CD72 and immune infiltration and prognosis of KIRC and to provide an important molecular basis for the early non-invasive diagnosis and immunotherapy of KRIC.

Materials and methods

Data sources and preprocessing

We used the ImmPort (https://​www.​immport.​org/​shared/​home) database to obtain immune-related genes [27]. The differential RNAseq expression data of CD72 in pan-cancer were obtained from UCSC XENA (https://​xenabrowser.​net/​datapages/​) in the TPM format of the TCGA and GTEx processed uniformly by the Toil process [28]. The differential RNAseq expression data of CD72 in unpaired and paired samples were in level 3 HTSeq‐FPKM format from the TCGA (https://​portal.​gdc.​cancer.​gov/​) KIRC project. FPKM (Fragments Per Kilobase per Million) format RNAseq data were converted to TPM (transcripts per million reads) format and log2 transformed. All final analyses were performed using data in TPM format. The differential analysis data for CD72 in dataset GSE40435, GSE53757 [29, 30] were downloaded from the GEO database using the GEOquery package (version 2.54.1) [31]. These data were obtained by removing probes corresponding to multiple molecules. When probes corresponding to the same molecule were encountered, only the probe with the largest signal value was retained. Then the data were normalized again by the normalize Between Arrays function of the limma package (version 3.42.2) [32]. All statistical analyses and visualizations were performed using R (version 4.2.1).

Cell culture

HK-2 cells were cultured in a specialized medium (Pricella, CM-0109). KIRC cell line (786-O, ACHN) was cultured in high sugar DMEM (Sigma) with 10% FBS (ExCell Bio) and 1% P/S (Solarbio). Both cell lines were cultured at 37 °C in a humidified environment containing 5% CO2.

Real-time PCR

After the cells were washed with PBS, Trizol and chloroform were added sequentially, and after repeated blowing and mixing, the cells were left to stand for 10 min and then centrifuged at 12,000 rpm for 15 min at 4° C. The supernatant was transferred to a new EP tube. An equal volume of isopropanol was added to mix it well, and after centrifugation, the RNA precipitates were cleaned with 75% ethanol and then lysed after air-drying, and the concentration was determined. The reaction system and procedures were constructed according to the instructions of the reverse transcription kit from Beijing TransGen Biotech. Real-time fluorescence quantification was performed by the SYBR Green method, and the reaction conditions were as follows: pre-denaturation at 94 ℃ for 30s, 94 ℃ for 5s, and 60 ℃ for 30s, for a total of 40 cycles. The relative expression levels of target genes were calculated using the 2-ΔΔCt method [33]. The primers were synthesized by Shanghai Sangon Biological Company: β-actin Forward primer 5′-CCTGGCACCCAGCACAAT-3′, reverse primer 5′-GGGCCGGACTCGTCATAC-3′; CD72-3 forward primer 5′-TCCGTCGGGGATGGATAATGC-3′, reverse primer 5′-TGCGTTGTGTATCATCAGTCAA-3′.

Differential expression analysis of CD72

The expression profiles of CD72 across cancers were analyzed for differences using the Mann–Whitney U test (Wilcoxon rank sum test). The Shapiro–Wilk normality test was used to test the normality of the CD72 expression data in paired samples, unpaired samples, and GSE40435, GSE53757, and the independent samples t test was used to analyze the differences in the data in unpaired samples. The paired-samples t test was used to analyze the differences in the data in paired samples, and the Mann–Whitney U test (Wilcoxon rank sum test) was used to analyze the data variance in GSE40435, GSE53757. The results of all the above analyses were visualized using ggplot2 (version 3.3.3) and were considered statistically significant when P < 0.05.

Differential analysis of CD72 protein expression levels in KIRC

Immunohistochemical staining images of CD72 in KIRC and normal tissue sections were downloaded using the HPA database (https://​www.​proteinatlas.​org/​).

Analysis of DEmRNA

We used the DESeq2 package [version 1.36.0] to perform gene differential analysis and CD72 single gene correlation analysis of KIRC in the TCGA database [34]. The results of the genetic difference analysis were used to generate volcano plots using ggplot2 software [version 3.3.3] [35]. |(LogFC)|> 1 and p.adj < 0.05 were used as thresholds for differentially expressed genes (DEGs). CD72 single gene co-expression heatmaps were generated by the ggplot2 [version 3.3.6] package using genes in positively and negatively correlated top15.

Identification of target genes associated with immunity

We used the ImmPort (https://​www.​immport.​org/​shared/​home) database to obtain immune-associated genes. Prognosis-related DEmRNA and immune-related genes were then derived using VennDiagram [version 1.7.3] overlap analysis. Finally, we identified CD72 as the target gene.

Clinical correlation analysis, survival prognosis analysis

The survival data of KIRC patients were statistically analyzed using the survival package (version 3.2‐10), and the results were visualized using the survival package (version 0.4.9) to plot KIRC patients' overall survival (OS), disease‐specific survival (DSS), and progression‐free interval (PFI) of Kaplan–Meier survival curves. We then performed a subgroup analysis of Kaplan–Meier survival curves in KIRC patients, analyzing clinicopathological factors such as age, gender, Pathologic T stage, serum calcium, and hemoglobin. We used these clinicopathological factors to calculate their correlation with CD72 expression and visualized the calculations using ggplot2 (version 3.3.3). ROC analysis of the data was performed with the pROC software package (version 1.17.0.1) to determine the accuracy of CD72 in predicting prognosis. Finally, logistic analysis of different clinicopathologic factors and CD72 expression were performed using the Survival Package (version 3.2-10). All prognostic data for the above survival analyses were obtained from a paper in cell [36].

Functional enrichment analysis

The single gene differential analysis results were analyzed for GO, KEGG, and GSEA functional enrichment using the clusterProfiler software package (version 3.14.3) [37]. Gene ID conversion was performed using the org.Hs.eg.db package (version 3.10.0), and the Z score was calculated using the GOplot package (version 1.0.2) [38], which scores the relevance of CD72 to the enrichment pathway. The reference gene set used for GSEA is c2.cp.all.v2022.1.Hs.symbols.gmt (All Canonical Pathways) [39], and the results were significantly enriched if they met the conditions of false discovery rate (FDR) < 0.25 and p.adjust < 0.05. All the above analyses were visualized using ggplot2 (version 3.3.3).

Immunocorrelation analysis of CD72

Relative infiltration levels of 24 immune cells were analyzed using the GSVA software package (version 24.1.34) [40]. For the immune infiltration algorithm, ssGSEA was employed, and Spearman correlation analysis was applied. Markers for 24 immune cells were obtained from an immunity study [41]. The samples were then divided into low and high-expression groups based on CD72 expression, and enrichment scores for various immune cell infiltrates in different subgroups were calculated and analyzed using the GSVA software package (version 1.34.0). Correlations between CD72 and immune cells and the expression of immune checkpoint-programmed cell death protein 1 (PD-1)-PDCD1, cytotoxic T-lymphocyte-associated protein 4 (CTLA4), and programmed cell death ligand 1 (PD-L1) were assessed using Spearman analysis. Finally, the correlation between immune cell infiltration and CD72 expression was visualized by analyzing immune cells with statistically significant relative infiltration (P < 0.05). The analysis results were visualized with the ggplot2 package (version 3.3.3) [42].

Statistical analysis

Data are expressed as mean ± standard deviation (mean ± SD). Student's t test analyzed differences in CD72 expression in KIRC tumor tissues and adjacent tissues. One-way analysis of variance (ANOVA) was used for comparison between multiple groups. The Mann–Whitney U test was used to analyze the correlation between CD72 expression and clinical data of KIRC patients. Statistical plots were completed using GraphPad Prism 8. P < 0.05 was considered statistically significant.

Results

We found 19,596 DEmRNAs between 541 KIRC and 72 kidney normal tissues. The volcano distribution is shown in Fig. 1A. We downloaded immune-related genes from the ImmPort database. We used Venn overlap analysis to get the overlapping target genes between immune-related genes and DEmRNAs associated with the prognosis of KIRC patients. The results showed 333 overlapping target genes, such as CD72, TNFAIP3, CETP, IL18R1, HTR3A, DEFB124, and RBP7 (Fig. 1B). Through comprehensive comparison, we finally chose CD72 as the target gene. Figure 1C shows a heat map of CD72 and its co-expressed mRNAs. The first 15 genes had a positive correlation with the expression of CD72, and the last 15 genes had a negative correlation with CD72.

A strong correlation exists between high CD72 expression and poor prognosis in KIRC

Figure 2A and B shows the expression of CD72 in the unpaired sample database UCSC XENA versus the paired database TCGA pan-cancer, respectively. The CD72 expression level of KIRC was higher than in kidney tissues (Fig. 2C, D). We then validated the results in the TCGA database using the datasets GSE40435, GSE53757 from the GEO database, and the results are shown in Fig. 2E, F.

Correlation of CD72 expression with clinicopathologic parameters

We analyzed the relationship between CD72 expression and various clinical characteristics of KIRC patients. Chisq test and Yates' correction were used to correlate clinicopathologic factors and CD72 expression, as shown in Table 1. Chisq test showed that CD72 was associated with Pathologic T stage (P < 0.001), Pathologic M stage (P < 0.001), Pathologic stage (P < 0.001), Race (P = 0.024), Histologic grade (< 0.001), Laterality (P < 0.001) were associated. The results of the logistic analysis are shown in Table 2. As shown in Fig. 3A–G, according to the Kruskal–Wallis Test and Dunn's test, CD72 expression correlated with KIRC patients' Pathologic T stage, Pathologic stage, Pathologic M stage, Pathologic N stage, Histologic grade, Laterality, and OS event were correlated (P < 0.05). According to survival analysis, as shown in Fig. 3H–J, high expression of CD72 was associated with poorer overall survival (OS), disease-specific survival (DSS), and progression-free interval (PFI) in KIRC patients. An AUC of 0.954 shows that KIRC may serve as a potential diagnostic biomarker (Fig. 3K).
Table 1
CD72 expression associated with clinicopathological characteristics (baseline data sheet)
Characteristics
Low expression of CD72
High expression of CD72
P value
n
270
271
 
Pathologic T stage, n (%)
  
< 0.001
 T1
164 (30.3%)
115 (21.3%)
 
 T2
30 (5.5%)
41 (7.6%)
 
 T3
76 (14%)
104 (19.2%)
 
 T4
0 (0%)
11 (2%)
 
Pathologic N stage, n (%)
  
0.091
 N0
113 (43.8%)
129 (50%)
 
 N1
4 (1.6%)
12 (4.7%)
 
Pathologic M stage, n (%)
  
< 0.001
 M0
227 (44.7%)
202 (39.8%)
 
 M1
25 (4.9%)
54 (10.6%)
 
Pathologic stage, n (%)
  
< 0.001
 Stage I
161 (29.9%)
112 (20.8%)
 
 Stage II
26 (4.8%)
33 (6.1%)
 
 Stage III
57 (10.6%)
66 (12.3%)
 
 Stage IV
25 (4.6%)
58 (10.8%)
 
Primary therapy outcome, n (%)
  
0.163
 PD
4 (2.7%)
7 (4.8%)
 
 SD
5 (3.4%)
1 (0.7%)
 
 PR
2 (1.4%)
0 (0%)
 
 CR
71 (48.3%)
57 (38.8%)
 
Gender, n (%)
  
0.054
 Female
104 (19.2%)
83 (15.3%)
 
 Male
166 (30.7%)
188 (34.8%)
 
Race, n (%)
  
0.024
 Asian
1 (0.2%)
7 (1.3%)
 
 Black or African American
35 (6.6%)
22 (4.1%)
 
 White
233 (43.6%)
236 (44.2%)
 
Age, n (%)
  
0.764
 <  = 60
136 (25.1%)
133 (24.6%)
 
 > 60
134 (24.8%)
138 (25.5%)
 
Histologic grade, n (%)
  
< 0.001
 G1
11 (2.1%)
3 (0.6%)
 
 G2
136 (25.5%)
100 (18.8%)
 
 G3
92 (17.3%)
115 (21.6%)
 
 G4
24 (4.5%)
52 (9.8%)
 
Serum calcium, n (%)
  
0.366
 Low
114 (31.1%)
90 (24.5%)
 
 Normal
74 (20.2%)
79 (21.5%)
 
 Elevated
5 (1.4%)
5 (1.4%)
 
Hemoglobin, n (%)
  
0.093
 Low
124 (26.9%)
140 (30.4%)
 
 Normal
106 (23%)
86 (18.7%)
 
 Elevated
4 (0.9%)
1 (0.2%)
 
Laterality, n (%)
  
< 0.001
 Left
105 (19.4%)
148 (27.4%)
 
 Right
164 (30.4%)
123 (22.8%)
 
Table 2
The logistic analysis of clinicopathological parameters in patients with KIRC
Characteristics
Total (N)
OR (95% CI)
P value
Pathologic T stage (T1 vs. T2 & T3 & T4)
541
0.476 (0.338–0.671)
< 0.001
Pathologic N stage (N1 vs. N0)
258
2.628 (0.8248.378)
0.102
Pathologic M stage (M1 vs. M0)
508
2.427 (1.4574.045)
< 0.001
Primary therapy outcome (CR vs. PD & SD & PR)
147
1.104 (0.4162.927)
0.843
Gender (male vs. female)
541
1.419 (0.9942.026)
0.054
Laterality (right vs. left)
540
0.532 (0.3780.749)
< 0.001
Pathologic stage (Stage I & Stage II vs. Stage III & Stage IV)
538
0.513 (0.3600.730)
< 0.001
Race (White vs. Asian & Black or African American)
534
1.257 (0.7462.118)
0.389
Age (< = 60 vs. > 60)
541
0.950 (0.6781.330)
0.764
Histologic grade (G1 & G2 vs. G3 & G4)
533
0.487 (0.3440.688)
< 0.001
Serum calcium (Low vs. Normal & Elevated)
367
0.742 (0.4911.122)
0.158
Hemoglobin (Low vs. Normal & Elevated)
461
1.428 (0.9852.068)
0.060

Subgroup analysis of survival prognosis of UNC93B1 expression

Figure 4A–I shows the survival of patients with high or low CD72 expression in different KIRC subgroups.
The results showed that the subgroup of age not older than 60 (HR = 2.00 (1.20–3.32), P = 0.08), the subgroup of sex male (HR = 1.49 (1.02–2.17), P = 0.040, the subgroup of T1 & T2 & T3 of T stage (HR = 1.44 (1.06–1.96), P = 0.021), Low & Normal subgroup of Serum calcium (HR = 1.75 (1.22–2.50), P = 0.040), Low & Normal subgroup of Hemoglobin: (HR = 1.45 (1.05–1.99), P = 0.022), Stage IV subgroup of Pathologic stage (HR = 1.87 (1.13–3.08), P = 0.015), M1 subgroup of M stage (HR = 1.73 (1.03–2.88), P = 0.038) were associated with increased CD72 expression and poor overall survival.

Functional enrichment analysis of CD72 in KIRC

GO, KEGG, and GSEA enrichment analyses were performed using the single gene differential analysis results shown in Fig. 5. GO analysis showed that CD72 was functionally associated with microtubule binding, cytoskeletal motor activity, histone kinase activity, G protein-coupled receptor binding, and long-chain fatty acid binding (Fig. 5A and Table 3). Figure 5B and Table 4 show the results of KEGG analysis that CD72 is associated with signaling pathways such as the Cell cycle, PPAR signaling pathway, p53 signaling pathway, Chemokine signaling pathway, and Cytokine–cytokine receptor interaction. Z scores reflect, to some extent, the relevance of CD72 to these pathways. Negative Z scores indicate a negative correlation, and positive Z scores indicate a positive correlation. Figure 5C, D shows the enrichment and grading results of GSEA, indicating that CD72 is closely associated with signaling pathways such as the Pd 1 signaling pathway, the CTLA4 pathway, the Th17 cell differentiation pathway, the B-cell receptor signaling pathway, costimulation of the CD28 family, primary immune deficiencies, cancer immunotherapies via Pd1 blockade, regulators of TCR signaling, and T-cell activation, as well as other genes associated with tumorigenesis, invasion, and angiogenesis.
Table 3
GO analysis
Ontology
ID
Description
Gene ratio
Bg ratio
P value
p.adjust
Z score
BP
GO:0140014
Mitotic nuclear division
31/197
293/18800
2.61e−22
7.82e−19
−0.5388159
BP
GO:0000070
Mitotic sister chromatid segregation
24/197
171/18800
2.26e−20
1.83e−17
− 0.8164966
BP
GO:0048285
Organelle fission
36/197
493/18800
2.44e−20
1.83e−17
− 1.0000000
BP
GO:0007059
Chromosome segregation
28/197
348/18800
4.75e−17
2.37e−14
− 1.5118579
BP
GO:0051304
Chromosome separation
14/197
97/18800
1.58e−12
4.3e−10
0.5345225
CC
GO:0005819
Spindle
26/203
402/19594
9.72e−14
2.88e−11
− 1.1766968
CC
GO:0072686
Mitotic spindle
17/203
160/19594
8.73e−13
1.29e−10
− 0.7276069
CC
GO:0000775
Chromosome, centromeric region
19/203
227/19594
2.81e−12
2.77e−10
− 1.1470787
CC
GO:0005875
Microtubule-associated complex
11/203
160/19594
9.09e−07
1.92e−05
− 0.9045340
CC
GO:0062023
Collagen-containing extracellular matrix
17/203
429/19594
2.56e−06
5.05e−05
− 0.2425356
MF
GO:0008017
Microtubule binding
19/192
272/18410
7.14e−11
3.28e−08
− 2.0647416
MF
GO:0003774
Cytoskeletal motor activity
9/192
111/18410
2.45e−06
0.0002
− 0.3333333
MF
GO:0035173
Histone kinase activity
4/192
16/18410
1.89e−05
0.0012
1.0000000
MF
GO:0001664
G protein-coupled receptor binding
11/192
288/18410
0.0002
0.0080
− 0.3015113
MF
GO:0036041
Long-chain fatty acid binding
3/192
15/18410
0.0005
0.0152
− 0.5773503
Table 4
KEGG analysis
Ontology
ID
Description
Gene ratio
Bg ratio
P value
p.adjust
Z score
KEGG
hsa04110
Cell cycle
11/95
126/8164
1.99e−07
4.18e−05
2.1105794
KEGG
hsa03320
PPAR signaling pathway
7/95
75/8164
2.41e−05
0.0010
− 0.3779645
KEGG
hsa04115
p53 signaling pathway
5/95
73/8164
0.0015
0.0400
1.3416408
KEGG
hsa04062
Chemokine signaling pathway
8/95
192/8164
0.0017
0.0400
0.0000000
KEGG
hsa04060
Cytokine–cytokine receptor interaction
10/95
295/8164
0.0022
0.0457
0.0000000

Correlation of CD72 expression with tumor immunity

As shown in Fig. 6A, the relationship between the relative number of 24 immune cells and the expression of CD72 in KIRC was evaluated using the ssGSEA algorithm. As shown in Fig. 6B–*I, different types of immune cells were correlated with CD72 expression, including T cells (R = 0.652, P < 0.001), T helper cells (= 0.605, P < 0.001), TReg (R = 0.509, P < 0.001), Th1 cells (R = 0.507, P < 0.001), Cytotoxic cells (R = 0.507, P < 0.001), Mast cells (R = − 0.125, P < 0.004), NK cells (R = − 0.149, P < 0.001), Th17 cells (R = 0.246, P < 0.001). Mann–Whitney U test (Wilcoxon rank sum test) was used to detect the enrichment of immune cells in CD72 high- and low-expression groups. The results showed that compared with the CD72 low-expression group, in the CD72 high-expression group, T cells, T helper cells, Treg, Th1 cells, Cytotoxic cells, aDC, B cells, Macrophages, TFH, Tcm, CD8 T cells, CD56bright cells, Tem, Eosinophils, and CD72 bright cells were enriched. Cells, Tem, Eosinophils, DC, and Neutrophils were more enriched (Fig. 7A–C). In KIRC, the expression of CD72 was positively associated with the expression of PD-1 (PDCD1) (R = 0.741, P < 0.001), CTLA4 (R = 0.744, P < 0.001), and PD-L1 (CD274) (R = 0.329, P < 0.001) using the spearman's analysis (Fig. 8A–C).

Evaluation of CD72 expression

Next, we evaluated the expression of CD72 in KIRC cells 786-O, ACHN, and renal cells HK-2 in Real-time PCR. As shown in Fig. 9A, the expression of CD72 in renal cancer cells was higher than in normal renal cells. In addition, the immunohistochemical results in the HPA database confirmed this result (Fig. 9B, C).

Discussion

KIRC, as the most common renal cell carcinoma, is challenging to diagnose at an early stage, surgery has limitations, and postoperative metastasis and recurrence are the main reasons for its high mortality rate. With the emergence of tyrosine kinase inhibitors targeting vascular endothelial growth factor receptors, rapamycin-targeted protein inhibitors, and immune checkpoint inhibitors, the choice of second-line treatment is expanding, and renal cancer treatment has entered a new era [43]. However, KIRC has high intrinsic heterogeneity and lacks early diagnostic and prognostic biomarkers [44, 45]. In addition, KIRC is one of the most immune-infiltrated tumors [46, 47]. Therefore, searching for new diagnostic and prognostic biomarkers and therapeutic targets and developing new anti-tumor drugs and immunotherapies are essential ways to improve the survival of KIRC patients.
In this study, we obtained clinical and RNA data of KIRC patients from the TCGA database and downloaded immune-related genes from the ImmPort database. Then we used the "limma" package, "survival" package, and Venn overlaps analysis to obtain differentially expressed mRNAs (DEmRNAs) related to the prognosis and immunity of KIRC patients, and we finally chose CD72 as the target gene.
Our study was analyzed using the UCSC XENA and TCGA database, and the results showed that in KIRC, compared with paracancerous tissues, the expression level of CD72 was significantly increased, which correlated with poor patient prognosis. KIRC patients with high expression of CD72 were less likely than those with low expression of CD72 in terms of either OS, DSS, or PFI. These results suggest that CD72 is associated with the occurrence and development of KIRC. In this study, CD72 expression was significantly correlated with the Pathologic T stage, Pathologic stage, Pathologic M stage, Pathologic N stage, Histologic grade, Laterality, and OS event, suggesting that CD72 may play an essential role in the biological function of tumor cells. Play an important role in the biological function of tumor cells. These results suggest that CD72 may be a useful diagnostic molecular marker for KIRC and can predict the outcome of KIRC patients. In addition, based on the ROC diagnostic curve (AUC = 0.954) and logistic analysis, it was further shown that CD72 could be used for KIRC diagnosis.CD72 is expected to be a potential biomarker for determining the prognosis, suggesting that patients with KIRC may benefit from using CD72 for diagnosis and prognosis.
To further understand the molecular mechanism of CD72 in tumorigenesis and development, functional enrichment analysis of GO, KEGG, and GSEA was performed using CD72 and its related differentially expressed genes. GO-based enrichment analysis showed that CD72 and its co-expressed mRNAs were enriched for several molecular functions (e.g., microtubule binding, cytoskeletal motor activity, histone kinase activity, G protein-coupled receptor binding, long-chain fatty acid binding), biological processes (mitotic nuclear division, mitotic sister chromatid segregation, organelle fission, chromosome segregation).
Abnormal expression of microtubule-binding proteins can cause cytoskeletal changes. Abnormal cytoskeletal motility activity may disrupt critical processes such as cell proliferation, migration, and metastasis [48, 49], leading to disorganization of cellular structure and abnormal proliferation of tumor cells [50]. Aberrant activation of histone kinases may lead to disrupted chromatin structure and aberrant gene expression, thereby affecting tumor cell proliferation and transcriptional regulation [51, 52]. Aberrant activity of G protein-coupled receptors may trigger aberrant activation of cell signaling pathways, promoting the proliferation and survival of tumor cells [53, 54]. RCC is essentially a metabolic disease characterized by a reprogramming of energetic metabolism [5558]. In particular the metabolic flux through glycolysis is partitioned [5961], and mitochondrial bioenergetics and OxPhox are impaired, as well as lipid metabolism [59, 6264]. In addition, aberrant expression of long-chain fatty acid binding proteins may lead to disturbances in intracellular energy metabolism and alterations in tumor cell growth [65].
In tumor development, mitotic nuclear division, sister chromatid segregation, and abnormal chromosome segregation lead to genomic instability, increase the risk of chromosomal abnormalities and mutations, and promote tumor formation and development [66, 67]. The correct alignment of the mitotic spindle during cell division is critical for cell fate determination, tissue organization, and development. Changes in the dynamics and control of microtubules that impair the mitotic spindle lead to chromosomal instability, leading to the generation of tumor cells [68, 69]. The molecular mechanism of CD72 in the development of KIRC may be related to its role in cell cycle regulation, cell growth, and cell migration.
KEGG analysis also showed that CD72 function is associated with signaling pathways such as cell cycle, PPAR signaling pathway, p53 signaling pathway, coagulation factor signaling pathway, and cytokine–cytokine receptor interactions. Abnormal cell cycle regulation is essential in tumorigenesis and progression, leading to unlimited cell proliferation [70]. Abnormalities in PPAR and p53 signaling pathways are associated with the progression of multiple tumors [7175]. Abnormalities in chemokine pathways are associated with tumor infiltration, metastasis, and neovascularization [7678]. Cytokine-receptor interactions are critical for regulating cell growth, differentiation, and immunity, and abnormalities can lead to tumor immune escape phenomena [79, 80].
Based on GSEA enrichment analysis, CD72 and its co-expressed mRNAs are enriched in signaling pathways such as Pd 1 Signaling, CTLA4 Pathway, Th17 Cell Differentiation Pathway, B Cell Receptor Signaling Pathway, Costimulation By the CD28 Family, Primary Immunodeficiency, Cancer Immunotherapy By Pd1 Blockade, Modulators of Tcr Signaling and T Cell Activation, Costimulation By the CD28 Family, Primary Immunodeficiency, Cancer Immunotherapy By Pd1 Blockade, Modulators of Tcr Signaling and T Cell Activation, and other Signaling Pathways associated with tumor immunity and tumorigenesis.
Correlation analysis showed a significant association between CD72 and PD-L1 (CD274) and CTLA4. Pd 1 and CTLA4 are two key immune checkpoint molecules with essential roles in regulating T-cell function and activating [81, 82]. When Pd-L1 (ligand for Pd 1) and CTLA4 are upregulated, tumor cells can evade the immune response and promote tumor growth. Activated T cells express PD-1 encoded by the PDCD1 gene, while PD-L1 encoded by the CD274 gene is overexpressed on the membrane surface of tumor cells. PD-1 binds to PD-L1, inhibits T-cell activation and causes their death, and then assists in the immune escape of tumor cells [83]. Abnormal B-cell receptor (BCR) signaling pathway activation may also lead to malignant transformation of B cells, which drives tumorigenesis and progression [84]. In addition, through activation of the Th17 cell differentiation pathway, increased numbers of tumor-infiltrating immune cells, promotion of neovascularization, and alterations in the tumor microenvironment can promote tumorigenesis and progression [85]. Co-stimulatory pathways of the CD28 family impact tumor growth and immune surveillance by influencing T-cell activation, proliferation, and anti-tumor immune responses, as well as modulating immune cell function [86]. The above results suggest that CD72 is essential in tumorigenesis and progression.
The results of immune infiltration showed that the degree of infiltration of T cells, T helper cells, Treg, Th1 cells, cytotoxic cells, aDC, B cells, macrophages, TFH, Tcm, CD8 T cells, CD56bright cells, Tem, eosinophils, DC, and neutrophils showed a significant positive correlation with CD72 expression. Mast cells, NK cells, and Th17 infiltration degree were significantly negatively correlated with CD72 expression. The transition from Th1/Th2 balance to Th2 dominance is critical during tumor progression. Th2 cells are detrimental to cellular immune anti-tumor effects. Restoration of Th1/Th2 balance is vital in tumor therapy [87]. Tregs are usually enriched in the tumor microenvironment, and many Tregs have a poor prognosis [88]. Lack of NK cell numbers and defective NK cell function promote the escape of tumor cells from immune surveillance [89]. Tumor-Associated Macrophages (TAMs) Promote Tumor Growth and Metastasis by Suppressing Tumor Immunosurveillance [90]. In addition, immunomodulatory interactions may be altered between lymphocytes and non-lymphocytes, leading to immune escape, immunosuppression, tumor growth and progression promotion, and concomitant suppression of tumor immunotherapy in KIRC [90]. All these results suggest that up-regulation of CD72 expression can suppress the anti-tumor immune response in KIRC patients.
In this study, correlation analysis showed a significant association between CD72 and PD-L1 (CD274), CTLA4, PD-1 (PDCD1), and immune cells (e.g., T cells, T helper cells, TReg, Th1 cells, Cytotoxic cells, Mast cells, NK cells, Th17 cells, etc.) were significantly associated with each other. In addition, in KIRC patients, compared with the CD72 low-expression group, the CD72 high-expression group showed a significant correlation between immune cells (T cells, T helper cells, Treg, Th1 cells, Cytotoxic cells, aDC, B cells, Macrophages, TFH, Tcm, CD8 T cells, CD56bright cells, Tem, Eosinophils, DC, Neutrophils, etc.) were more enriched. Our results suggest that CD72 is closely associated with immune infiltration and immunosuppression in the microenvironment of KIRC tumors.

Conclusion

In conclusion, we found that CD72 was overexpressed in KIRC and correlated with the clinical stage of patients, and it may be a marker for early diagnosis of KIRC patients. In addition, CD72 was associated with poor patient prognosis and could be an independent prognostic factor for KIRC.CD72 may promote tumor development by regulating the cell cycle and immune-related signaling pathways and facilitating immune cell infiltration.CD72 also showed a significant positive correlation with PD-L1 (CD274), CTLA4, and PD-1 (PDCD1) immunotherapeutic targets. Therefore, CD72 is expected to be a potential diagnostic and prognostic biomarker for KIRC and a new target for anti-tumor drug development. However, the results obtained in this study require additional experiments, such as animal and cellular experiments, to further validate the mechanism by which CD72 promotes KIRC. Since this study was a retrospective study based on the available RNA sequencing data in the TCGA and GEO databases, prospective studies are needed in the future to minimize the bias caused by retrospective studies.

Declarations

Not applicable, this study does not involve any ethical.

Competing interests

The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest.
Open Access This 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.
2.
Zurück zum Zitat Sjöberg E, Frödin M, Lövrot J, Mezheyeuski A, Johansson M, Harmenberg U, et al. A minority-group of renal cell cancer patients with high infiltration of CD20+B-cells is associated with poor prognosis. Br J Cancer. 2018;119:840–6.PubMedPubMedCentralCrossRef Sjöberg E, Frödin M, Lövrot J, Mezheyeuski A, Johansson M, Harmenberg U, et al. A minority-group of renal cell cancer patients with high infiltration of CD20+B-cells is associated with poor prognosis. Br J Cancer. 2018;119:840–6.PubMedPubMedCentralCrossRef
3.
Zurück zum Zitat Hu J, Chen Z, Bao L, Zhou L, Hou Y, Liu L, et al. Single-cell transcriptome analysis reveals intratumoral heterogeneity in ccRCC, which results in different clinical outcomes. Mol Ther. 2020;28:1658–72.PubMedPubMedCentralCrossRef Hu J, Chen Z, Bao L, Zhou L, Hou Y, Liu L, et al. Single-cell transcriptome analysis reveals intratumoral heterogeneity in ccRCC, which results in different clinical outcomes. Mol Ther. 2020;28:1658–72.PubMedPubMedCentralCrossRef
4.
Zurück zum Zitat Makhov P, Joshi S, Ghatalia P, Kutikov A, Uzzo RG, Kolenko VM. Resistance to systemic therapies in clear cell renal cell carcinoma: mechanisms and management strategies. Mol Cancer Ther. 2018;17:1355–64.PubMedPubMedCentralCrossRef Makhov P, Joshi S, Ghatalia P, Kutikov A, Uzzo RG, Kolenko VM. Resistance to systemic therapies in clear cell renal cell carcinoma: mechanisms and management strategies. Mol Cancer Ther. 2018;17:1355–64.PubMedPubMedCentralCrossRef
5.
Zurück zum Zitat Nukui A, Masuda A, Abe H, Arai K, Yoshida KI, Kamai T. Increased serum level of soluble interleukin-2 receptor is associated with a worse response of metastatic clear cell renal cell carcinoma to interferon alpha and sequential VEGF-targeting therapy. BMC Cancer. 2017;17:372.PubMedPubMedCentralCrossRef Nukui A, Masuda A, Abe H, Arai K, Yoshida KI, Kamai T. Increased serum level of soluble interleukin-2 receptor is associated with a worse response of metastatic clear cell renal cell carcinoma to interferon alpha and sequential VEGF-targeting therapy. BMC Cancer. 2017;17:372.PubMedPubMedCentralCrossRef
6.
Zurück zum Zitat Lasorsa F, di Meo NA, Rutigliano M, Milella M, Ferro M, Pandolfo SD, et al. Immune checkpoint inhibitors in renal cell carcinoma: molecular basis and rationale for their use in clinical practice. Biomedicines. 2023;11:1071.PubMedPubMedCentralCrossRef Lasorsa F, di Meo NA, Rutigliano M, Milella M, Ferro M, Pandolfo SD, et al. Immune checkpoint inhibitors in renal cell carcinoma: molecular basis and rationale for their use in clinical practice. Biomedicines. 2023;11:1071.PubMedPubMedCentralCrossRef
7.
Zurück zum Zitat Ghini V, Laera L, Fantechi B, Monte FD, Benelli M, McCartney A, et al. Metabolomics to assess response to immune checkpoint inhibitors in patients with non-small-cell lung cancer. Cancers (Basel). 2020;12:3574.PubMedPubMedCentralCrossRef Ghini V, Laera L, Fantechi B, Monte FD, Benelli M, McCartney A, et al. Metabolomics to assess response to immune checkpoint inhibitors in patients with non-small-cell lung cancer. Cancers (Basel). 2020;12:3574.PubMedPubMedCentralCrossRef
8.
Zurück zum Zitat Lucarelli G, Netti GS, Rutigliano M, Lasorsa F, Loizzo D, Milella M, et al. MUC1 expression affects the immunoflogosis in renal cell carcinoma microenvironment through complement system activation and immune infiltrate modulation. Int J Mol Sci. 2023;24:4814.PubMedPubMedCentralCrossRef Lucarelli G, Netti GS, Rutigliano M, Lasorsa F, Loizzo D, Milella M, et al. MUC1 expression affects the immunoflogosis in renal cell carcinoma microenvironment through complement system activation and immune infiltrate modulation. Int J Mol Sci. 2023;24:4814.PubMedPubMedCentralCrossRef
9.
Zurück zum Zitat Lasorsa F, Rutigliano M, Milella M, Ferro M, Pandolfo SD, Crocetto F, et al. Cellular and molecular players in the tumor microenvironment of renal cell carcinoma. J Clin Med. 2023;12:3888.PubMedPubMedCentralCrossRef Lasorsa F, Rutigliano M, Milella M, Ferro M, Pandolfo SD, Crocetto F, et al. Cellular and molecular players in the tumor microenvironment of renal cell carcinoma. J Clin Med. 2023;12:3888.PubMedPubMedCentralCrossRef
10.
Zurück zum Zitat Shi T, Song X, Wang Y, Liu F, Wei J. Combining oncolytic viruses with cancer immunotherapy: establishing a new generation of cancer treatment. Front Immunol. 2020;11:683.PubMedPubMedCentralCrossRef Shi T, Song X, Wang Y, Liu F, Wei J. Combining oncolytic viruses with cancer immunotherapy: establishing a new generation of cancer treatment. Front Immunol. 2020;11:683.PubMedPubMedCentralCrossRef
11.
Zurück zum Zitat Ueda K, Suekane S, Kurose H, Chikui K, Nakiri M, Nishihara K, et al. Prognostic value of PD-1 and PD-L1 expression in patients with metastatic clear cell renal cell carcinoma. Urol Oncol. 2018;36:499.CrossRef Ueda K, Suekane S, Kurose H, Chikui K, Nakiri M, Nishihara K, et al. Prognostic value of PD-1 and PD-L1 expression in patients with metastatic clear cell renal cell carcinoma. Urol Oncol. 2018;36:499.CrossRef
12.
Zurück zum Zitat Mollica V, Santoni M, Matrana MR, Basso U, De Giorgi U, Rizzo A, et al. Concomitant proton pump inhibitors and outcome of patients treated with nivolumab alone or plus ipilimumab for advanced renal cell carcinoma. Target Oncol. 2022;17:61–8.PubMedCrossRef Mollica V, Santoni M, Matrana MR, Basso U, De Giorgi U, Rizzo A, et al. Concomitant proton pump inhibitors and outcome of patients treated with nivolumab alone or plus ipilimumab for advanced renal cell carcinoma. Target Oncol. 2022;17:61–8.PubMedCrossRef
13.
Zurück zum Zitat Pan C, Baumgarth N, Parnes JR. CD72-deficient mice reveal nonredundant roles of CD72 in B cell development and activation. Immunity. 1999;11:495–506.PubMedCrossRef Pan C, Baumgarth N, Parnes JR. CD72-deficient mice reveal nonredundant roles of CD72 in B cell development and activation. Immunity. 1999;11:495–506.PubMedCrossRef
14.
Zurück zum Zitat Nitschke L, Tsubata T. Molecular interactions regulate BCR signal inhibition by CD22 and CD72. Trends Immunol. 2004;25:543–50.PubMedCrossRef Nitschke L, Tsubata T. Molecular interactions regulate BCR signal inhibition by CD22 and CD72. Trends Immunol. 2004;25:543–50.PubMedCrossRef
15.
Zurück zum Zitat Tsubata T. Role of inhibitory BCR co-receptors in immunity. Infect Disord Drug Targets. 2012;12:181–90.PubMedCrossRef Tsubata T. Role of inhibitory BCR co-receptors in immunity. Infect Disord Drug Targets. 2012;12:181–90.PubMedCrossRef
16.
Zurück zum Zitat Adachi T, Wakabayashi C, Nakayama T, Yakura H, Tsubata T. CD72 negatively regulates signaling through the antigen receptor of B cells. J Immunol. 2000;164:1223–9.PubMedCrossRef Adachi T, Wakabayashi C, Nakayama T, Yakura H, Tsubata T. CD72 negatively regulates signaling through the antigen receptor of B cells. J Immunol. 2000;164:1223–9.PubMedCrossRef
17.
Zurück zum Zitat Kikutani H, Kumanogoh A. Semaphorins in interactions between T cells and antigen-presenting cells. Nat Rev Immunol. 2003;3:159–67.PubMedCrossRef Kikutani H, Kumanogoh A. Semaphorins in interactions between T cells and antigen-presenting cells. Nat Rev Immunol. 2003;3:159–67.PubMedCrossRef
18.
Zurück zum Zitat Vadasz Z, Goldeberg Y, Halasz K, Rosner I, Valesini G, Conti F, et al. Increased soluble CD72 in systemic lupus erythematosus is in association with disease activity and lupus nephritis. Clin Immunol. 2016;164:114–8.PubMedCrossRef Vadasz Z, Goldeberg Y, Halasz K, Rosner I, Valesini G, Conti F, et al. Increased soluble CD72 in systemic lupus erythematosus is in association with disease activity and lupus nephritis. Clin Immunol. 2016;164:114–8.PubMedCrossRef
19.
Zurück zum Zitat Akatsu C, Shinagawa K, Numoto N, Liu Z, Ucar AK, Aslam M, et al. CD72 negatively regulates B lymphocyte responses to the lupus-related endogenous toll-like receptor 7 ligand Sm/RNP. J Exp Med. 2016;213:2691–706.PubMedPubMedCentralCrossRef Akatsu C, Shinagawa K, Numoto N, Liu Z, Ucar AK, Aslam M, et al. CD72 negatively regulates B lymphocyte responses to the lupus-related endogenous toll-like receptor 7 ligand Sm/RNP. J Exp Med. 2016;213:2691–706.PubMedPubMedCentralCrossRef
20.
Zurück zum Zitat Eiza N, Sabag AD, Kessler O, Neufeld G, Vadasz Z. CD72-semaphorin3A axis: a new regulatory pathway in systemic lupus erythematosus. J Autoimmun. 2023;134: 102960.PubMedCrossRef Eiza N, Sabag AD, Kessler O, Neufeld G, Vadasz Z. CD72-semaphorin3A axis: a new regulatory pathway in systemic lupus erythematosus. J Autoimmun. 2023;134: 102960.PubMedCrossRef
21.
Zurück zum Zitat Wu M, Li J, Gao Q, Ye F. The role of Sema4D/CD100 as a therapeutic target for tumor microenvironments and for autoimmune, neuroimmune and bone diseases. Expert Opin Ther Targets. 2016;20:885–901.PubMedCrossRef Wu M, Li J, Gao Q, Ye F. The role of Sema4D/CD100 as a therapeutic target for tumor microenvironments and for autoimmune, neuroimmune and bone diseases. Expert Opin Ther Targets. 2016;20:885–901.PubMedCrossRef
22.
Zurück zum Zitat Schwarting R, Castello R, Moldenhauer G, Pezzutto A, von Hoegen I, Ludwig WD, et al. Human Lyb-2 homolog CD72 is a marker for progenitor B-cell leukemias. Am J Hematol. 1992;41:151–8.PubMedCrossRef Schwarting R, Castello R, Moldenhauer G, Pezzutto A, von Hoegen I, Ludwig WD, et al. Human Lyb-2 homolog CD72 is a marker for progenitor B-cell leukemias. Am J Hematol. 1992;41:151–8.PubMedCrossRef
23.
Zurück zum Zitat Coustan-Smith E, Song G, Clark C, Key L, Liu P, Mehrpooya M, et al. New markers for minimal residual disease detection in acute lymphoblastic leukemia. Blood. 2011;117:6267–76.PubMedPubMedCentralCrossRef Coustan-Smith E, Song G, Clark C, Key L, Liu P, Mehrpooya M, et al. New markers for minimal residual disease detection in acute lymphoblastic leukemia. Blood. 2011;117:6267–76.PubMedPubMedCentralCrossRef
24.
Zurück zum Zitat Liu J, Lan Y, Tian G, Yang J. A systematic framework for identifying prognostic genes in the tumor microenvironment of colon cancer. Front Oncol. 2022;12: 899156.PubMedPubMedCentralCrossRef Liu J, Lan Y, Tian G, Yang J. A systematic framework for identifying prognostic genes in the tumor microenvironment of colon cancer. Front Oncol. 2022;12: 899156.PubMedPubMedCentralCrossRef
25.
Zurück zum Zitat Ai D, Wang M, Zhang Q, Cheng L, Wang Y, Liu X, et al. Regularized survival learning and cross-database analysis enabled identification of colorectal cancer prognosis-related immune genes. Front Genet. 2023;14:1148470.PubMedPubMedCentralCrossRef Ai D, Wang M, Zhang Q, Cheng L, Wang Y, Liu X, et al. Regularized survival learning and cross-database analysis enabled identification of colorectal cancer prognosis-related immune genes. Front Genet. 2023;14:1148470.PubMedPubMedCentralCrossRef
26.
Zurück zum Zitat Lu X, Chen X, Wang X, Qing J, Li J, Pan Y. Construction of lncRNA and mRNA co-expression network associated with nasopharyngeal carcinoma progression. Front Oncol. 2022;12: 965088.PubMedPubMedCentralCrossRef Lu X, Chen X, Wang X, Qing J, Li J, Pan Y. Construction of lncRNA and mRNA co-expression network associated with nasopharyngeal carcinoma progression. Front Oncol. 2022;12: 965088.PubMedPubMedCentralCrossRef
27.
Zurück zum Zitat Dai Y, Qiang W, Lin K, Gui Y, Lan X, Wang D. An immune-related gene signature for predicting survival and immunotherapy efficacy in hepatocellular carcinoma. Cancer Immunol Immunother. 2021;70:967–79.PubMedCrossRef Dai Y, Qiang W, Lin K, Gui Y, Lan X, Wang D. An immune-related gene signature for predicting survival and immunotherapy efficacy in hepatocellular carcinoma. Cancer Immunol Immunother. 2021;70:967–79.PubMedCrossRef
28.
Zurück zum Zitat Vivian J, Rao AA, Nothaft FA, Ketchum C, Armstrong J, Novak A, et al. Toil enables reproducible, open source, big biomedical data analyses. Nat Biotechnol. 2017;35:314–6.PubMedPubMedCentralCrossRef Vivian J, Rao AA, Nothaft FA, Ketchum C, Armstrong J, Novak A, et al. Toil enables reproducible, open source, big biomedical data analyses. Nat Biotechnol. 2017;35:314–6.PubMedPubMedCentralCrossRef
29.
Zurück zum Zitat Gruosso T, Mieulet V, Cardon M, Bourachot B, Kieffer Y, Devun F, et al. Chronic oxidative stress promotes H2AX protein degradation and enhances chemosensitivity in breast cancer patients. EMBO Mol Med. 2016;8:527–49.PubMedPubMedCentralCrossRef Gruosso T, Mieulet V, Cardon M, Bourachot B, Kieffer Y, Devun F, et al. Chronic oxidative stress promotes H2AX protein degradation and enhances chemosensitivity in breast cancer patients. EMBO Mol Med. 2016;8:527–49.PubMedPubMedCentralCrossRef
30.
Zurück zum Zitat Day RS, McDade KK. A decision theory paradigm for evaluating identifier mapping and filtering methods using data integration. BMC Bioinform. 2013;14:223.CrossRef Day RS, McDade KK. A decision theory paradigm for evaluating identifier mapping and filtering methods using data integration. BMC Bioinform. 2013;14:223.CrossRef
31.
Zurück zum Zitat Davis S, Meltzer PS. GEOquery: a bridge between the Gene Expression Omnibus (GEO) and BioConductor. Bioinformatics. 2007;23:1846–7.PubMedCrossRef Davis S, Meltzer PS. GEOquery: a bridge between the Gene Expression Omnibus (GEO) and BioConductor. Bioinformatics. 2007;23:1846–7.PubMedCrossRef
32.
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: 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: e47.PubMedPubMedCentralCrossRef
33.
Zurück zum Zitat Chen J, Xu D, Wang T, Yang Z, Yang Y, He K, et al. HMGB1 promotes the development of castration-resistant prostate cancer by regulating androgen receptor activation. Oncol Rep. 2022;48:197.PubMedPubMedCentralCrossRef Chen J, Xu D, Wang T, Yang Z, Yang Y, He K, et al. HMGB1 promotes the development of castration-resistant prostate cancer by regulating androgen receptor activation. Oncol Rep. 2022;48:197.PubMedPubMedCentralCrossRef
34.
36.
Zurück zum Zitat Liu J, Lichtenberg T, Hoadley KA, Poisson LM, Lazar AJ, Cherniack AD, et al. An integrated TCGA pan-cancer clinical data resource to drive high-quality survival outcome analytics. Cell. 2018;173:400-16.e11.PubMedPubMedCentralCrossRef Liu J, Lichtenberg T, Hoadley KA, Poisson LM, Lazar AJ, Cherniack AD, et al. An integrated TCGA pan-cancer clinical data resource to drive high-quality survival outcome analytics. Cell. 2018;173:400-16.e11.PubMedPubMedCentralCrossRef
37.
38.
Zurück zum Zitat Walter W, Sánchez-Cabo F, Ricote M. GOplot: an R package for visually combining expression data with functional analysis. Bioinformatics. 2015;31:2912–4.PubMedCrossRef Walter W, Sánchez-Cabo F, Ricote M. GOplot: an R package for visually combining expression data with functional analysis. Bioinformatics. 2015;31:2912–4.PubMedCrossRef
39.
Zurück zum Zitat Subramanian A, Tamayo P, Mootha VK, Mukherjee S, Ebert BL, Gillette MA, et al. Gene set enrichment analysis: a knowledge-based approach for interpreting genome-wide expression profiles. Proc Natl Acad Sci U S A. 2005;102:15545–50.PubMedPubMedCentralCrossRef Subramanian A, Tamayo P, Mootha VK, Mukherjee S, Ebert BL, Gillette MA, et al. Gene set enrichment analysis: a knowledge-based approach for interpreting genome-wide expression profiles. Proc Natl Acad Sci U S A. 2005;102:15545–50.PubMedPubMedCentralCrossRef
40.
Zurück zum Zitat Hänzelmann S, Castelo R, Guinney J. GSVA: gene set variation analysis for microarray and RNA-seq data. BMC Bioinform. 2013;14:7.CrossRef Hänzelmann S, Castelo R, Guinney J. GSVA: gene set variation analysis for microarray and RNA-seq data. BMC Bioinform. 2013;14:7.CrossRef
41.
Zurück zum Zitat Bindea G, Mlecnik B, Tosolini M, Kirilovsky A, Waldner M, Obenauf AC, et al. Spatiotemporal dynamics of intratumoral immune cells reveal the immune landscape in human cancer. Immunity. 2013;39:782–95.PubMedCrossRef Bindea G, Mlecnik B, Tosolini M, Kirilovsky A, Waldner M, Obenauf AC, et al. Spatiotemporal dynamics of intratumoral immune cells reveal the immune landscape in human cancer. Immunity. 2013;39:782–95.PubMedCrossRef
42.
Zurück zum Zitat Gu Z, Gu L, Eils R, Schlesner M, Brors B. circlize Implements and enhances circular visualization in R. Bioinformatics. 2014;30:2811–2.PubMedCrossRef Gu Z, Gu L, Eils R, Schlesner M, Brors B. circlize Implements and enhances circular visualization in R. Bioinformatics. 2014;30:2811–2.PubMedCrossRef
43.
Zurück zum Zitat Xu W, Atkins MB, McDermott DF. Checkpoint inhibitor immunotherapy in kidney cancer. Nat Rev Urol. 2020;17:137–50.PubMedCrossRef Xu W, Atkins MB, McDermott DF. Checkpoint inhibitor immunotherapy in kidney cancer. Nat Rev Urol. 2020;17:137–50.PubMedCrossRef
44.
Zurück zum Zitat López JI. Intratumor heterogeneity in clear cell renal cell carcinoma: a review for the practicing pathologist. APMIS. 2016;124:153–9.PubMedCrossRef López JI. Intratumor heterogeneity in clear cell renal cell carcinoma: a review for the practicing pathologist. APMIS. 2016;124:153–9.PubMedCrossRef
45.
Zurück zum Zitat Beksac AT, Paulucci DJ, Blum KA, Yadav SS, Sfakianos JP, Badani KK. Heterogeneity in renal cell carcinoma. Urol Oncol. 2017;35:507–15.PubMedCrossRef Beksac AT, Paulucci DJ, Blum KA, Yadav SS, Sfakianos JP, Badani KK. Heterogeneity in renal cell carcinoma. Urol Oncol. 2017;35:507–15.PubMedCrossRef
46.
47.
Zurück zum Zitat Tamma R, Rutigliano M, Lucarelli G, Annese T, Ruggieri S, Cascardi E, et al. Microvascular density, macrophages, and mast cells in human clear cell renal carcinoma with and without bevacizumab treatment. Urol Oncol. 2019;37:355.CrossRef Tamma R, Rutigliano M, Lucarelli G, Annese T, Ruggieri S, Cascardi E, et al. Microvascular density, macrophages, and mast cells in human clear cell renal carcinoma with and without bevacizumab treatment. Urol Oncol. 2019;37:355.CrossRef
49.
Zurück zum Zitat Dogterom M, Koenderink GH. Actin-microtubule crosstalk in cell biology. Nat Rev Mol Cell Biol. 2019;20:38–54.PubMedCrossRef Dogterom M, Koenderink GH. Actin-microtubule crosstalk in cell biology. Nat Rev Mol Cell Biol. 2019;20:38–54.PubMedCrossRef
50.
Zurück zum Zitat Kanakkanthara A, Miller JH. βIII-tubulin overexpression in cancer: causes, consequences, and potential therapies. Biochim Biophys Acta Rev Cancer. 2021;1876: 188607.PubMedCrossRef Kanakkanthara A, Miller JH. βIII-tubulin overexpression in cancer: causes, consequences, and potential therapies. Biochim Biophys Acta Rev Cancer. 2021;1876: 188607.PubMedCrossRef
51.
Zurück zum Zitat Simboeck E, Gutierrez A, Cozzuto L, Beringer M, Caizzi L, Keyes WM, et al. DPY30 regulates pathways in cellular senescence through ID protein expression. Embo j. 2013;32:2217–30.PubMedPubMedCentralCrossRef Simboeck E, Gutierrez A, Cozzuto L, Beringer M, Caizzi L, Keyes WM, et al. DPY30 regulates pathways in cellular senescence through ID protein expression. Embo j. 2013;32:2217–30.PubMedPubMedCentralCrossRef
52.
Zurück zum Zitat Nacev BA, Feng L, Bagert JD, Lemiesz AE, Gao J, Soshnev AA, et al. The expanding landscape of “oncohistone” mutations in human cancers. Nature. 2019;567:473–8.PubMedPubMedCentralCrossRef Nacev BA, Feng L, Bagert JD, Lemiesz AE, Gao J, Soshnev AA, et al. The expanding landscape of “oncohistone” mutations in human cancers. Nature. 2019;567:473–8.PubMedPubMedCentralCrossRef
53.
Zurück zum Zitat Elangovan S, Pathania R, Ramachandran S, Ananth S, Padia RN, Lan L, et al. The niacin/butyrate receptor GPR109A suppresses mammary tumorigenesis by inhibiting cell survival. Cancer Res. 2014;74:1166–78.PubMedCrossRef Elangovan S, Pathania R, Ramachandran S, Ananth S, Padia RN, Lan L, et al. The niacin/butyrate receptor GPR109A suppresses mammary tumorigenesis by inhibiting cell survival. Cancer Res. 2014;74:1166–78.PubMedCrossRef
54.
Zurück zum Zitat Huang C, Dai XY, Cai JX, Chen J, Wang BB, Zhu W, et al. A screened GPR1 peptide exerts antitumor effects on triple-negative breast cancer. Mol Ther Oncolytics. 2020;18:602–12.PubMedPubMedCentralCrossRef Huang C, Dai XY, Cai JX, Chen J, Wang BB, Zhu W, et al. A screened GPR1 peptide exerts antitumor effects on triple-negative breast cancer. Mol Ther Oncolytics. 2020;18:602–12.PubMedPubMedCentralCrossRef
55.
Zurück zum Zitat di Meo NA, Lasorsa F, Rutigliano M, Milella M, Ferro M, Battaglia M, et al. The dark side of lipid metabolism in prostate and renal carcinoma: novel insights into molecular diagnostic and biomarker discovery. Expert Rev Mol Diagn. 2023;23:297–313.PubMedCrossRef di Meo NA, Lasorsa F, Rutigliano M, Milella M, Ferro M, Battaglia M, et al. The dark side of lipid metabolism in prostate and renal carcinoma: novel insights into molecular diagnostic and biomarker discovery. Expert Rev Mol Diagn. 2023;23:297–313.PubMedCrossRef
56.
Zurück zum Zitat Lucarelli G, Loizzo D, Franzin R, Battaglia S, Ferro M, Cantiello F, et al. Metabolomic insights into pathophysiological mechanisms and biomarker discovery in clear cell renal cell carcinoma. Expert Rev Mol Diagn. 2019;19:397–407.PubMedCrossRef Lucarelli G, Loizzo D, Franzin R, Battaglia S, Ferro M, Cantiello F, et al. Metabolomic insights into pathophysiological mechanisms and biomarker discovery in clear cell renal cell carcinoma. Expert Rev Mol Diagn. 2019;19:397–407.PubMedCrossRef
57.
Zurück zum Zitat di Meo NA, Lasorsa F, Rutigliano M, Loizzo D, Ferro M, Stella A, et al. Renal cell carcinoma as a metabolic disease: an update on main pathways, potential biomarkers, and therapeutic targets. Int J Mol Sci. 2022;23:14360.PubMedPubMedCentralCrossRef di Meo NA, Lasorsa F, Rutigliano M, Loizzo D, Ferro M, Stella A, et al. Renal cell carcinoma as a metabolic disease: an update on main pathways, potential biomarkers, and therapeutic targets. Int J Mol Sci. 2022;23:14360.PubMedPubMedCentralCrossRef
58.
Zurück zum Zitat De Marco S, Torsello B, Minutiello E, Morabito I, Grasselli C, Bombelli S, et al. The cross-talk between Abl2 tyrosine kinase and TGFβ1 signalling modulates the invasion of clear cell renal cell carcinoma cells. FEBS Lett. 2023;597:1098–113.PubMedCrossRef De Marco S, Torsello B, Minutiello E, Morabito I, Grasselli C, Bombelli S, et al. The cross-talk between Abl2 tyrosine kinase and TGFβ1 signalling modulates the invasion of clear cell renal cell carcinoma cells. FEBS Lett. 2023;597:1098–113.PubMedCrossRef
59.
Zurück zum Zitat Bianchi C, Meregalli C, Bombelli S, Di Stefano V, Salerno F, Torsello B, et al. The glucose and lipid metabolism reprogramming is grade-dependent in clear cell renal cell carcinoma primary cultures and is targetable to modulate cell viability and proliferation. Oncotarget. 2017;8:113502–15.PubMedPubMedCentralCrossRef Bianchi C, Meregalli C, Bombelli S, Di Stefano V, Salerno F, Torsello B, et al. The glucose and lipid metabolism reprogramming is grade-dependent in clear cell renal cell carcinoma primary cultures and is targetable to modulate cell viability and proliferation. Oncotarget. 2017;8:113502–15.PubMedPubMedCentralCrossRef
60.
Zurück zum Zitat Ragone R, Sallustio F, Piccinonna S, Rutigliano M, Vanessa G, Palazzo S, et al. Renal cell carcinoma: a study through NMR-based metabolomics combined with transcriptomics. Diseases. 2016;4:7.PubMedPubMedCentralCrossRef Ragone R, Sallustio F, Piccinonna S, Rutigliano M, Vanessa G, Palazzo S, et al. Renal cell carcinoma: a study through NMR-based metabolomics combined with transcriptomics. Diseases. 2016;4:7.PubMedPubMedCentralCrossRef
61.
Zurück zum Zitat Lucarelli G, Galleggiante V, Rutigliano M, Sanguedolce F, Cagiano S, Bufo P, et al. Metabolomic profile of glycolysis and the pentose phosphate pathway identifies the central role of glucose-6-phosphate dehydrogenase in clear cell-renal cell carcinoma. Oncotarget. 2015;6:13371–86.PubMedPubMedCentralCrossRef Lucarelli G, Galleggiante V, Rutigliano M, Sanguedolce F, Cagiano S, Bufo P, et al. Metabolomic profile of glycolysis and the pentose phosphate pathway identifies the central role of glucose-6-phosphate dehydrogenase in clear cell-renal cell carcinoma. Oncotarget. 2015;6:13371–86.PubMedPubMedCentralCrossRef
62.
Zurück zum Zitat Lucarelli G, Rutigliano M, Sallustio F, Ribatti D, Giglio A, Lepore Signorile M, et al. Integrated multi-omics characterization reveals a distinctive metabolic signature and the role of NDUFA4L2 in promoting angiogenesis, chemoresistance, and mitochondrial dysfunction in clear cell renal cell carcinoma. Aging (Albany NY). 2018;10:3957–85.PubMedCrossRef Lucarelli G, Rutigliano M, Sallustio F, Ribatti D, Giglio A, Lepore Signorile M, et al. Integrated multi-omics characterization reveals a distinctive metabolic signature and the role of NDUFA4L2 in promoting angiogenesis, chemoresistance, and mitochondrial dysfunction in clear cell renal cell carcinoma. Aging (Albany NY). 2018;10:3957–85.PubMedCrossRef
63.
Zurück zum Zitat Bombelli S, Torsello B, De Marco S, Lucarelli G, Cifola I, Grasselli C, et al. 36-kDa annexin A3 isoform negatively modulates lipid storage in clear cell renal cell carcinoma cells. Am J Pathol. 2020;190:2317–26.PubMedCrossRef Bombelli S, Torsello B, De Marco S, Lucarelli G, Cifola I, Grasselli C, et al. 36-kDa annexin A3 isoform negatively modulates lipid storage in clear cell renal cell carcinoma cells. Am J Pathol. 2020;190:2317–26.PubMedCrossRef
64.
Zurück zum Zitat Lucarelli G, Rutigliano M, Loizzo D, di Meo NA, Lasorsa F, Mastropasqua M, et al. MUC1 tissue expression and its soluble form CA15–3 identify a clear cell renal cell carcinoma with distinct metabolic profile and poor clinical outcome. Int J Mol Sci. 2022;23:13968.PubMedPubMedCentralCrossRef Lucarelli G, Rutigliano M, Loizzo D, di Meo NA, Lasorsa F, Mastropasqua M, et al. MUC1 tissue expression and its soluble form CA15–3 identify a clear cell renal cell carcinoma with distinct metabolic profile and poor clinical outcome. Int J Mol Sci. 2022;23:13968.PubMedPubMedCentralCrossRef
67.
Zurück zum Zitat McFarlane RJ, Wakeman JA. Meiosis-like functions in oncogenesis: a new view of cancer. Cancer Res. 2017;77:5712–6.PubMedCrossRef McFarlane RJ, Wakeman JA. Meiosis-like functions in oncogenesis: a new view of cancer. Cancer Res. 2017;77:5712–6.PubMedCrossRef
68.
Zurück zum Zitat Marquis C, Fonseca CL, Queen KA, Wood L, Vandal SE, Malaby HLH, et al. Chromosomally unstable tumor cells specifically require KIF18A for proliferation. Nat Commun. 2021;12:1213.PubMedPubMedCentralCrossRef Marquis C, Fonseca CL, Queen KA, Wood L, Vandal SE, Malaby HLH, et al. Chromosomally unstable tumor cells specifically require KIF18A for proliferation. Nat Commun. 2021;12:1213.PubMedPubMedCentralCrossRef
69.
70.
Zurück zum Zitat Bisteau X, Caldez MJ, Kaldis P. The complex relationship between liver cancer and the cell cycle: a story of multiple regulations. Cancers (Basel). 2014;6:79–111.PubMedCrossRef Bisteau X, Caldez MJ, Kaldis P. The complex relationship between liver cancer and the cell cycle: a story of multiple regulations. Cancers (Basel). 2014;6:79–111.PubMedCrossRef
71.
Zurück zum Zitat Iitaka D, Moodley S, Shimizu H, Bai XH, Liu M. PKCδ-iPLA2-PGE2-PPARγ signaling cascade mediates TNF-α induced Claudin 1 expression in human lung carcinoma cells. Cell Signal. 2015;27:568–77.PubMedCrossRef Iitaka D, Moodley S, Shimizu H, Bai XH, Liu M. PKCδ-iPLA2-PGE2-PPARγ signaling cascade mediates TNF-α induced Claudin 1 expression in human lung carcinoma cells. Cell Signal. 2015;27:568–77.PubMedCrossRef
72.
Zurück zum Zitat Ishtiaq SM, Arshad MI, Khan JA. PPARγ signaling in hepatocarcinogenesis: mechanistic insights for cellular reprogramming and therapeutic implications. Pharmacol Ther. 2022;240: 108298.PubMedCrossRef Ishtiaq SM, Arshad MI, Khan JA. PPARγ signaling in hepatocarcinogenesis: mechanistic insights for cellular reprogramming and therapeutic implications. Pharmacol Ther. 2022;240: 108298.PubMedCrossRef
73.
Zurück zum Zitat Hartley A, Ahmad I. The role of PPARγ in prostate cancer development and progression. Br J Cancer. 2023;128:940–5.PubMedCrossRef Hartley A, Ahmad I. The role of PPARγ in prostate cancer development and progression. Br J Cancer. 2023;128:940–5.PubMedCrossRef
74.
Zurück zum Zitat Zhu Z, McGray AJR, Jiang W, Lu B, Kalinski P, Guo ZS. Improving cancer immunotherapy by rationally combining oncolytic virus with modulators targeting key signaling pathways. Mol Cancer. 2022;21:196.PubMedPubMedCentralCrossRef Zhu Z, McGray AJR, Jiang W, Lu B, Kalinski P, Guo ZS. Improving cancer immunotherapy by rationally combining oncolytic virus with modulators targeting key signaling pathways. Mol Cancer. 2022;21:196.PubMedPubMedCentralCrossRef
75.
Zurück zum Zitat Hernández Borrero LJ, El-Deiry WS. Tumor suppressor p53: Biology, signaling pathways, and therapeutic targeting. Biochim Biophys Acta Rev Cancer. 2021;1876: 188556.PubMedCrossRef Hernández Borrero LJ, El-Deiry WS. Tumor suppressor p53: Biology, signaling pathways, and therapeutic targeting. Biochim Biophys Acta Rev Cancer. 2021;1876: 188556.PubMedCrossRef
76.
Zurück zum Zitat Sceneay J, Smyth MJ, Möller A. The pre-metastatic niche: finding common ground. Cancer Metastasis Rev. 2013;32:449–64.PubMedCrossRef Sceneay J, Smyth MJ, Möller A. The pre-metastatic niche: finding common ground. Cancer Metastasis Rev. 2013;32:449–64.PubMedCrossRef
78.
Zurück zum Zitat Netti GS, Lucarelli G, Spadaccino F, Castellano G, Gigante M, Divella C, et al. PTX3 modulates the immunoflogosis in tumor microenvironment and is a prognostic factor for patients with clear cell renal cell carcinoma. Aging (Albany NY). 2020;12:7585–602.PubMedCrossRef Netti GS, Lucarelli G, Spadaccino F, Castellano G, Gigante M, Divella C, et al. PTX3 modulates the immunoflogosis in tumor microenvironment and is a prognostic factor for patients with clear cell renal cell carcinoma. Aging (Albany NY). 2020;12:7585–602.PubMedCrossRef
79.
Zurück zum Zitat Nagarsheth N, Wicha MS, Zou W. Chemokines in the cancer microenvironment and their relevance in cancer immunotherapy. Nat Rev Immunol. 2017;17:559–72.PubMedPubMedCentralCrossRef Nagarsheth N, Wicha MS, Zou W. Chemokines in the cancer microenvironment and their relevance in cancer immunotherapy. Nat Rev Immunol. 2017;17:559–72.PubMedPubMedCentralCrossRef
80.
Zurück zum Zitat Tumino N, Martini S, Munari E, Scordamaglia F, Besi F, Mariotti FR, et al. Presence of innate lymphoid cells in pleural effusions of primary and metastatic tumors: functional analysis and expression of PD-1 receptor. Int J Cancer. 2019;145:1660–8.PubMedPubMedCentralCrossRef Tumino N, Martini S, Munari E, Scordamaglia F, Besi F, Mariotti FR, et al. Presence of innate lymphoid cells in pleural effusions of primary and metastatic tumors: functional analysis and expression of PD-1 receptor. Int J Cancer. 2019;145:1660–8.PubMedPubMedCentralCrossRef
81.
Zurück zum Zitat Yuan J, Gnjatic S, Li H, Powel S, Gallardo HF, Ritter E, et al. CTLA-4 blockade enhances polyfunctional NY-ESO-1 specific T cell responses in metastatic melanoma patients with clinical benefit. Proc Natl Acad Sci U S A. 2008;105:20410–5.PubMedPubMedCentralCrossRef Yuan J, Gnjatic S, Li H, Powel S, Gallardo HF, Ritter E, et al. CTLA-4 blockade enhances polyfunctional NY-ESO-1 specific T cell responses in metastatic melanoma patients with clinical benefit. Proc Natl Acad Sci U S A. 2008;105:20410–5.PubMedPubMedCentralCrossRef
82.
Zurück zum Zitat Chen R, Peng PC, Wen B, Li FY, Xie S, Chen G, et al. Anti-programmed cell death (PD)-1 immunotherapy for malignant tumor: a systematic review and meta-analysis. Transl Oncol. 2016;9:32–40.PubMedPubMedCentralCrossRef Chen R, Peng PC, Wen B, Li FY, Xie S, Chen G, et al. Anti-programmed cell death (PD)-1 immunotherapy for malignant tumor: a systematic review and meta-analysis. Transl Oncol. 2016;9:32–40.PubMedPubMedCentralCrossRef
84.
Zurück zum Zitat Wilson WH, Young RM, Schmitz R, Yang Y, Pittaluga S, Wright G, et al. Targeting B cell receptor signaling with ibrutinib in diffuse large B cell lymphoma. Nat Med. 2015;21:922–6.PubMedPubMedCentralCrossRef Wilson WH, Young RM, Schmitz R, Yang Y, Pittaluga S, Wright G, et al. Targeting B cell receptor signaling with ibrutinib in diffuse large B cell lymphoma. Nat Med. 2015;21:922–6.PubMedPubMedCentralCrossRef
85.
Zurück zum Zitat Marshall EA, Ng KW, Kung SH, Conway EM, Martinez VD, Halvorsen EC, et al. Emerging roles of T helper 17 and regulatory T cells in lung cancer progression and metastasis. Mol Cancer. 2016;15:67.PubMedPubMedCentralCrossRef Marshall EA, Ng KW, Kung SH, Conway EM, Martinez VD, Halvorsen EC, et al. Emerging roles of T helper 17 and regulatory T cells in lung cancer progression and metastasis. Mol Cancer. 2016;15:67.PubMedPubMedCentralCrossRef
86.
Zurück zum Zitat Zhuang X, Long EO. CD28 homolog is a strong activator of natural killer cells for lysis of B7H7(+) tumor cells. Cancer Immunol Res. 2019;7:939–51.PubMedPubMedCentralCrossRef Zhuang X, Long EO. CD28 homolog is a strong activator of natural killer cells for lysis of B7H7(+) tumor cells. Cancer Immunol Res. 2019;7:939–51.PubMedPubMedCentralCrossRef
87.
Zurück zum Zitat Chen H, Xie J, Jin P. Assessment of hazard immune-related genes and tumor immune infiltrations in renal cell carcinoma. Am J Transl Res. 2020;12:7096–113.PubMedPubMedCentral Chen H, Xie J, Jin P. Assessment of hazard immune-related genes and tumor immune infiltrations in renal cell carcinoma. Am J Transl Res. 2020;12:7096–113.PubMedPubMedCentral
88.
Zurück zum Zitat Facciabene A, Peng X, Hagemann IS, Balint K, Barchetti A, Wang LP, et al. Tumour hypoxia promotes tolerance and angiogenesis via CCL28 and T(reg) cells. Nature. 2011;475:226–30.PubMedCrossRef Facciabene A, Peng X, Hagemann IS, Balint K, Barchetti A, Wang LP, et al. Tumour hypoxia promotes tolerance and angiogenesis via CCL28 and T(reg) cells. Nature. 2011;475:226–30.PubMedCrossRef
89.
Zurück zum Zitat Liu P, Chen L, Zhang H. Natural killer cells in liver disease and hepatocellular carcinoma and the NK cell-based immunotherapy. J Immunol Res. 2018;2018:1206737.PubMedPubMedCentralCrossRef Liu P, Chen L, Zhang H. Natural killer cells in liver disease and hepatocellular carcinoma and the NK cell-based immunotherapy. J Immunol Res. 2018;2018:1206737.PubMedPubMedCentralCrossRef
90.
Zurück zum Zitat Wu H, Han Y, Rodriguez Sillke Y, Deng H, Siddiqui S, Treese C, et al. Lipid droplet-dependent fatty acid metabolism controls the immune suppressive phenotype of tumor-associated macrophages. EMBO Mol Med. 2019;11: e10698.PubMedCentralCrossRef Wu H, Han Y, Rodriguez Sillke Y, Deng H, Siddiqui S, Treese C, et al. Lipid droplet-dependent fatty acid metabolism controls the immune suppressive phenotype of tumor-associated macrophages. EMBO Mol Med. 2019;11: e10698.PubMedCentralCrossRef
Metadaten
Titel
CD72, a new immune checkpoint molecule, is a novel prognostic biomarker for kidney renal clear cell carcinoma
Publikationsdatum
01.12.2023
Erschienen in
European Journal of Medical Research / Ausgabe 1/2023
Elektronische ISSN: 2047-783X
DOI
https://doi.org/10.1186/s40001-023-01487-8

Weitere Artikel der Ausgabe 1/2023

European Journal of Medical Research 1/2023 Zur Ausgabe