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

Open Access 01.12.2023 | Research

Multi-omics analysis uncovers clinical, immunological, and pharmacogenomic implications of cuproptosis in clear cell renal cell carcinoma

verfasst von: Maoshu Zhu, Yongsheng Li, Yun Wang, Pingli Lin, Jun Mi, Weimin Zhong

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

download
DOWNLOAD
print
DRUCKEN
insite
SUCHEN

Abstract

Objective

The latest research proposed a novel copper-dependent programmed cell death named cuproptosis. We aimed to elucidate the influence of cuproptosis in clear cell renal cell carcinoma (ccRCC) from a multi-omic perspective.

Methods

This study systematically assessed mRNA expression, methylation, and genetic alterations of cuproptosis genes in TCGA ccRCC samples. Through unsupervised clustering analysis, the samples were classified as different cuproptosis subtypes, which were verified through NTP method in the E-MTAB-1980 dataset. Next, the cuproptosis score (Cuscore) was computed based on cuproptosis-related genes via PCA. We also evaluated clinical and immunogenomic features, drug sensitivity, immunotherapeutic response, and post-transcriptional regulation.

Results

Cuproptosis genes presented multi-layer alterations in ccRCC, and were linked with patients’ survival and immune microenvironment. We defined three cuproptosis subtypes [C1 (moderate cuproptosis), C2 (low cuproptosis), and C3 (high cuproptosis)], and the robustness and reproducibility of this classification was further proven. Overall survival was best in C3, moderate in C1, and worst in C2. C1 had the highest sensitivity to pazopanib, and sorafenib, while C2 was most sensitive to sunitinib. Furthermore, C1 patients benefited more from anti-PD-1 immunotherapy. Patients with high Cuscore presented the notable survival advantage. Cuscore was highly linked with immunogenomic features, and post-transcriptional events that contributed to ccRCC development. Finally, several potential compounds and druggable targets (NMU, RARRES1) were selected for low Cuscore group.

Conclusion

Overall, our study revealed the non-negligible role of cuproptosis in ccRCC development. Evaluation of the cuproptosis subtypes improves our cognition of immunogenomic features and better guides personalized prognostication and precision therapy.
Hinweise

Supplementary Information

The online version contains supplementary material available at https://​doi.​org/​10.​1186/​s40001-023-01221-4.
Maoshu Zhu, Yongsheng Li and Yun Wang have contributed equally to this work.

Publisher's Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Abkürzungen
ccRCC
Clear cell renal cell carcinoma
RCC
Renal cell carcinoma
TCGA
The Cancer Genome Atlas
TPM
Transcripts per million
SCNA
Somatic copy-number alteration
miRNA
MicroRNA
GO
Gene Ontology
KEGG
Kyoto Encyclopedia of Genes and Genomes
GSEA
Gene set enrichment analysis
FDR
False discovery rate
GSVA
Gene set variation analysis
fGSEA
Fast GSEA
NTP
Nearest template prediction
SNV
Single-nucleotide variant
TMB
Tumor mutation burden
CTA
Cancer-testis antigen
mRNAsi
MRNA expression-based stemness index
CCLs
Cancer cell lines
GDSC
Genomics of drug sensitivity in cancer
CTRP
Cancer therapeutics response portal
CCLE
Cancer cell line encyclopedia
ssGSEA
Single-sample GSEA
SubMap
Subclass mapping
Cuscore
Cuproptosis score
DEGs
Differentially expressed genes
PCs
Principal components
K-M
Kaplan–Meier
OS
Overall survival
ROC
Receiver operating characteristic
AUC
Area under the curve
PCA
Principal component analysis
CDF
Cumulative distribution function

Introduction

Renal cell carcinoma (RCC) originating from renal tubular epithelial cells occupies ~ 3.8% of all newly diagnosed cancers [1]. Clear cell RCC (ccRCC) is an aggressive histological subtype that accounts for ~ 75% of all cases [2]. Over one-third of ccRCC patients experience relapse and metastasis after surgery, and patients with metastatic ccRCC have an undesirable prognosis, with a 5-year survival rate of 10% [3]. Currently, the effects of ccRCC clinical treatment modalities mainly incorporating surgical management, conventional chemotherapy, targeted therapy, and immunotherapy, etc., is limited by intratumoral heterogeneity that challenges the molecular characterization of ccRCC and is a confounding factor for treatment selection [4, 5]. Recently, multi-omics technology have led to a significant advantage in understanding the molecular mechanism and cancer management [6, 7]. Extensive genomic characterization has unveiled a few genetic alterations (VHL, PBRM1, etc.) correlated to ccRCC [8]. Moreover, large-scale molecular profiling analysis (The Cancer Genome Atlas (TCGA), etc.) has identified critical biological processes in ccRCC [9]. Nonetheless, molecular profiling is not routinely applied for ccRCC prognostication and therapeutic options. Additional molecular mechanisms may further improve stratification of ccRCC patients into proper risk classifications.
Copper is a fundamental mineral nutrient, and its redox properties make it both beneficial and toxic to cells [10]. Both serum and tumor copper levels are elevated in solid tumors, which directly correlate to cancer progression [11]. Recently, a novel copper-dependent programmed cell death mechanism named cuproptosis has been proposed, which strongly correlates to mitochondrial metabolism and is mediated by protein lipoylation [12]. Evidence demonstrates that cuproptosis genes exhibit aberrant expression in ccRCC and are notably linked with patients’ prognosis [13]. In addition, cuproptosis might potentially predict immunotherapeutic response of ccRCC [14]. However, our cognition of cuproptosis is still in its infancy. Molecular profiling enables to provide crucial prognostic information and treatment guidance for the management of ccRCC patients. In this study, the clinical, immunological and pharmacogenomic role of cuproptosis was investigated in ccRCC, which may shed light on optimizing the precision management of ccRCC patients. The flowchart of our research is illustrated in Fig. 1.

Materials and methods

Publicly available ccRCC datasets

Transcriptome profiling (HTSeq-counts) of ccRCC (n = 539) and normal (n = 72) specimens together with clinical parameters were acquired from TCGA (https://​portal.​gdc.​cancer.​gov/​). Raw counts data were converted into transcripts per million (TPM), followed by log2 transformation. DNA methylation data (Methylation450K), somatic mutation (mutation annotation format), copy-number alteration (SCNA; Masked Copy Number Segment) and microRNA (miRNA) expression data of ccRCC patients were also collected from TCGA. Matrix files of transcriptome profiling and prognostic data of ccRCC patients (n = 101) were acquired from the E-MTAB-1980 dataset (https://​www.​ebi.​ac.​uk/​arrayexpress/​experiments/​E-MTAB-1980/​), which was utilized for external verification. The detailed clinicopathological information is listed in Additional file 2: Table S1.

Collection of cuproptosis genes

Cuproptosis genes were collected from a previously published study [15]. Circos plot containing chromosome positions of cuproptosis genes was drawn via RCircos package [12].

Methylation analysis

For DNA methylation data, only the probes that mapped to the promoter region of cuproptosis genes were retained. For genes with multiple probes, the average β value of all probes was utilized as the methylation level.

Somatic mutation and SCNA analysis

To lower the false-positive rate, only non-silent mutations were retained. Through maftools package [16], the mutation annotation format from TCGA was analyzed. For SCNA, significant amplifications and deletions were identified via GISTIC2.0 [17]. Then, OncoPrint plots of mutations and SCNA were produced with ComplexHeatmap package [18].

Functional enrichment analysis

The enrichment score of Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) gene sets was evaluated between low and high expression of CDKN2A groups using gene set enrichment analysis (GSEA) [19]. GO and KEGG enrichment analysis of cuproptosis-related genes was conducted via clusterProfiler package [20]. Terms with false discovery rate (FDR) < 0.05 were regarded as significant enrichment. Gene set variation analysis (GSVA) [21], and fast GSEA (fGSEA) were used to compute and compare the activity of the 50 hallmark pathways based on the Molecular Signatures Database-derived “h.all.v7.4.entrez.gmt” gene set as the reference [22].

Unsupervised clustering analysis

ccRCC samples were classified as distinct cuproptosis subtypes based on the transcriptome profiling of cuproptosis genes using unsupervised clustering algorithm. The number of clusters was identified via ConsensusClusterPlus package [23], with iterations for increasing the classification reliability.

Subtype validation

The top 200 up-regulated genes in each subtype versus others were identified via limma package [24], which were used to validate the cuproptosis subtypes through nearest template prediction (NTP) algorithm in the E-MTAB-1980 cohort [25].

Immunogenomic and stemness signatures

Single-nucleotide variant (SNV) neoantigens, tumor mutation burden (TMB), SCNA, cancer-testis antigen (CTA) score, homologous recombination defects, intratumor heterogeneity, and aneuploidy score were acquired from previously published literature [26]. In addition, mRNA expression-based stemness index (mRNAsi) was computed utilizing one-class logistic regression machine-learning approach [27].

Drug sensitivity assessment

Drug sensitivity data of cancer cell lines (CCLs) were acquired from Genomics of Drug Sensitivity in Cancer (GDSC) [28], Cancer Therapeutics Response Portal (CTRP) [46], and PRISM [29] databases. GDSC covers the half-maximal inhibitory concentration (IC50) data; meanwhile, both CTRP and PRISM cover the area under the curve (AUC) data as an evaluation indicator of drug sensitivity. Transcriptome profiling of CCLs was acquired from the Cancer Cell Line Encyclopedia (CCLE) [30]. The IC50 of GDSC-derived compounds was evaluated through oncoPredict package [31]. CTRP- and PRISM-derived compounds with more than 20% missing data were removed before K-nearest neighbor imputation. Next, pRRophetic package was employed to estimate the AUC of compounds [32]. Spearman correlation analysis on Cuscore and IC50 or AUC was executed to evaluate the drug response of ccRCC patients to each compound.

Tumor-infiltrating immune cells

Single-sample GSEA (ssGSEA) was utilized to quantify the relative abundance of 22 tumor-infiltrating immune cell types and two stromal components (fibroblasts and endothelial cells) based on the known marker genes (Additional file 3: Table S2) [33]. Immune score and stromal score were estimated through ESTIMATE algorithm [34].

Multi‑omics analysis of immunomodulators

Multi-omics profiling (comprising mRNA expression, SCNA, and DNA methylation) of 75 immunomodulators was observed across distinct cuproptosis subtypes (Additional file 4: Table S3) [35].

Immunotherapy response prediction

Subclass Mapping (SubMap) analysis was adopted to assess the expression similarity between the three cuproptosis subtypes and immunotherapeutic responses [36]. The degree of commonality between the two groups was inferred via GSEA algorithm. Bonferroni-corrected p < 0.05 denoted that two groups were significantly similar.

Generation of the cuproptosis score (Cuscore)

To identify the cuproptosis-related genes based on the three cuproptosis subtypes, differentially expressed genes (DEGs) were screened through limma package with the threshold of |fold change|> 1.5 together with adjusted p < 0.05. Prognostic value of cuproptosis-related genes was assessed via univariate Cox regression analysis. Then, genes with p < 0.05 were retained for computing the Cuscore through adopting PCA, with the principal components (PCs) 1 and 2 as the final signature score. The Cuscore was defined as Cuscore = \(\sum_{n}^{m}(PCn+PCm)\), where n and m denoted the order and total number of prognostic cuproptosis-related genes in ccRCC, respectively.

Nomogram construction

Univariate Cox regression on clinicopathological parameters together with Cuscore was analyzed across ccRCC patients. Thereafter, significant prognostic predictors (p < 0.05) were retained for multivariate Cox regression analysis, and a nomogram was defined through adopting independent predictive variables (p < 0.05). The consistency between predicted and actual prognostic outcome was assessed via calibration curves. Through decision curve analysis, the net benefits of the nomogram, Cuscore and other clinicopathological parameters were measured.

Analysis of post-transcriptional mechanisms

MiRNAs with differential expression were screened between low and high Cuscore groups based on |fold change|> 2 and adjusted p < 0.01. Then, targeted pathways were enriched through KEGG enrichment analysis.

Statistical analysis and visualization

Continuous variables in two or more than two groups were compared utilizing parametric test (Student’s t-test or analysis of variance) if the variables displayed normal distribution or nonparametric test (Wilcoxon rank-sum test or Kruskal–Wallis test). Associations between continuous variables were evaluated via Pearson or Spearman correlation test. Kaplan–Meier (K-M) analysis of overall survival (OS) with log-rank test was conducted using survminer package. Time-dependent receiver operating characteristic (ROC) curves were plotted with timeROC package, and AUC was computed. Relationships of Cuscore with clinicopathological parameters were analyzed through Chi-square test. The discrimination of transcriptome profiling between groups was displayed via principal component analysis (PCA). Uni- and multivariate Cox regression analysis was executed to determine the prognostic genes and independent prognostic parameters via survival package. All statistical analyses were executed through R packages. Statistical significance was set as a two-tailed p < 0.05.

Results

Multi-omics landscape of cuproptosis genes in ccRCC

Figure 2A illustrates the genomic location of each cuproptosis gene. Most cuproptosis genes (FDX1, DLD, DLAT, PDHA1, PDHB, and GLS) were notably down-regulated in ccRCC versus normal tissues, with only up-regulated CDKN2A (Fig. 2B, C). At the transcriptional level, cuproptosis genes positively interacted except CDKN2A in ccRCC (Fig. 2D). Next, we analyzed the reasons for the low expression of cuproptosis genes. Higher methylation levels of most cuproptosis genes were observed in ccRCC than normal tissues (Fig. 2E). In addition, copy number deletion of cuproptosis genes occurred in ccRCC, especially PDHB (Fig. 2F). Hypermethylation and copy number deletion might contribute to the low expression of cuproptosis genes. Their prognostic value was then assessed. Consequently, most cuproptosis genes were linked with better OS outcome of ccRCC, with opposite effect of CDKN2A (Fig. 2G). PCA revealed the remarkable discrimination of transcriptome profiling of cuproptosis genes between ccRCC and normal samples (Fig. 2H). Altogether, cuproptosis might be indispensable for ccRCC initiation and progression. ccRCC is highly immune infiltrated; adaptive and innate immune cells infiltrate the tumor microenvironment and constitute an ecosystem that regulates each aspect of ccRCC development [37]. Most cuproptosis genes were negatively correlated to the infiltration of immune cells, with positive relationships between CDKN2A and immune cell infiltration (Fig. 2I). Based on above findings, CDKN2A was identified as a cuproptosis gene of interest. CDKN2A expression was higher in more advanced grade and stage (Fig. 2J, K). GSEA revealed that CDKN2A positively correlated to immunity (activation of immune response, immunoglobulin production, lymphocyte-mediated immunity, etc.) (Fig. 2L).

Construction and external verification of cuproptosis subtypes in ccRCC

According to the transcriptome profiling of cuproptosis genes, cuproptosis subtypes were established via unsupervised clustering analysis. When the consensus value k was 3, most of the colors in the consensus matrix did not overlap (Fig. 3A). Cumulative distribution function (CDF) as well as item tracking demonstrated the cluster stability when k = 3 (Additional file 1: Figure S1A–C). Taken together, we classified ccRCC as three cuproptosis subtypes, namely C1 (moderate cuproptosis), C2 (low cuproptosis), and C3 (high cuproptosis) (Additional file 5: Table S4; Fig. 3B). PCA illustrates the notable discrepancy of transcriptome profiling among the three cuproptosis subtypes (Fig. 3C). Prognosis difference was observed among the three cuproptosis subtypes, with the best OS in C3, intermediate in C1, and worst in C2 (Fig. 3D). According to fold change > 1 together with adjusted p < 0.05, we determined up-regulated genes in each cuproptosis subtype (Fig. 3E). The top 200 genes were regarded as up-regulated biomarkers of each cuproptosis subtype (Additional file 6: Table S5). To further verify the stability and robustness of cuproptosis subtypes, NTP analysis that may quantify the prediction confidence of each patient based on transcriptome profiling was implemented in the E-MATB-1980 cohort. Consequently, the three cuproptosis subtypes exhibited high reproducibility in the E-MATB-1980 dataset (Fig. 3F). The differences in OS outcome and transcriptome profiling among the three cuproptosis subtypes were proven in this cohort (Fig. 3G, H). Clinicopathological features were prominently different among the three cuproptosis subtypes, with the lowest proportions of grade and stage in C3, intermediate in C1, and highest in C2 (Fig. 3I). Altogether, the cuproptosis-based classification was reproducible and stable in ccRCC.

Genomic alterations, stemness, and drug sensitivity features across cuproptosis subtypes

DNA alterations mainly comprise somatic mutation and SCNA. Overall, somatic mutation exhibited the remarkable heterogeneity among the three cuproptosis subtypes (Fig. 4A–C). Of note, C3 had the highest mutation frequency of VHL (61%), followed by C1 (57%) and C2 (54%). PBRM1 had the highest mutation frequency in C1 (41%), intermediate in C3 (33%), and lowest in C2 (20%). Overall, C3 presented significantly lower SNV neoantigens and TMB score relative to C1 (Fig. 4D, E). Next, we observed the heterogeneity of SCNA profiling in the three cuproptosis subtypes (Fig. 4F). Additionally, C1 presented higher SCNA, CTA score, homologous recombination defects, and intratumor heterogeneity (Fig. 4G–J). We computed mRNAsi for quantifying cancer stemness, and found that C3 had higher mRNAsi (Fig. 4K). Drug sensitivity differences were also assessed among the three cuproptosis subtypes. Consequently, C1 patients had the highest sensitivity to pazopanib, and sorafenib, while C2 patients were most sensitive to sunitinib (Fig. 4L–N).

Three cuproptosis subtypes with distinct immunotherapeutic responses

Next, we observed the remarkable differences in the oncogenic hallmark pathways among the three cuproptosis subtypes, contributing to the intratumor heterogeneity of ccRCC (Fig. 5A). C1 subtype presented the highest expression of common immune checkpoint molecules (PDCD1, CTLA4, etc.) (Fig. 5B). Additionally, the most abundant infiltration of immune cells was observed in C2, with intermediate for C1, and lowest for C3. In Fig. 5C, there were extensive genomic differences in immunomodulators across the three cuproptosis subtypes. Of note, C3 had the highest frequencies of amplification (IFNG, CD27, LAG3, CD40, GZMA, etc.) and deletion (PDCD1LG2, CD274, BTN3A1, BTN3A2, TNF, VEGFA, IFNA1, IFNA2, MICA, MICB, TLR4, PRF1, ENTPD1, etc.) of several immunomodulators, followed by C1 and C2. Considering that the cuproptosis subtypes appear to correlate to the tumor immune microenvironment, we inferred the immunotherapeutic responses of the three cuproptosis subtypes. Consequently, C1 subtype exhibited the high expression similarity to response to anti-PD-1 therapy (Fig. 5D), proven in the E-MTAB-1980 dataset (Fig. 5E). Hence, patients in C1 subtype clinically benefited more from anti-PD-1 immunotherapy.

Definition of the Cuscore for individual ccRCC patients

To assess the functional role of the three cuproptosis subtypes, 95 cuproptosis-related genes were determined (Fig. 6A; Additional file 7: Table S6) through intersecting DEGs between cuproptosis subtypes according to |fold change|> 1.5 and adjusted p < 0.05. Functional enrichment analysis revealed that they were remarkably enriched in diverse metabolism processes (Fig. 6B). Next, the prognostic value of cuproptosis-related genes was investigated. Consequently, all of them were significantly linked with ccRCC patients’ OS (Additional file 8: Table S7). Through PCA computational approach, the Cuscore was defined based on cuproptosis-related genes to quantify cuproptosis of individual ccRCC patients. Next, we investigated the clinical relevance of the Cuscore. ccRCC patients were classified as low and high Cuscore groups, with the prominent distinction of transcriptome profiling (Fig. 6C). Patients with high Cuscore possessed a remarkable survival benefit (Fig. 6D), with 1-, 3- and 5-year OS AUC values of 0.74, 0.69 and 0.70, respectively (Fig. 6E). The E-MTAB-1980 dataset proved the high reproducibility of this Cuscore (Fig. 6F–H).

Associations of the Cuscore with clinicopathological characteristics

Next, clinicopathological characteristics of low and high Cuscore groups were observed. In Fig. 7A, low Cuscore patients exhibited higher proportions of dead status, male, advanced stage, and grade. Additionally, male patients had lower Cuscore relative to female patients (Fig. 7B). With the increase in grade and stage, the Cuscore gradually lowered (Fig. 7C, D). Altogether, the Cuscore may reflect tumor progression and prognostic outcome.

Generation of the Cuscore-based nomogram for predicting ccRCC prognosis

From uni- and multivariate Cox regression results, Cuscore served as an independent protective factor, with clinicopathological variables (age, stage, and grade) as independent risk factors (Fig. 7E, F). To facilitate the clinical utility of the Cuscore in predicting the survival probability, we generated a nomogram that integrated above independent prognostic factors. Calibration curves of 1-, 3- and 5-year OS displayed that the nomogram was close to the actual survival probability (Fig. 7G–I). Moreover, we found that the nomogram had the highest net benefit in clinical assessment based on decision curve analysis, suggesting the significance of the Cuscore in coordination with other clinicopathological variables for survival prediction (Fig. 7J–L).

Immunogenomic features, and oncogenic hallmark pathways associated with Cuscore

To understand the immunogenomic features involved in the Cuscore, we evaluated the relationships of the Cuscore with immunogenomic signatures. Consequently, the Cuscore was negatively linked with aneuploidy score, CTA score, homologous recombination defects, and intratumor heterogeneity in ccRCC (Fig. 8A–D). Additionally, we observed the negative associations of the Cuscore with most immune cells (Fig. 8E), and most immune checkpoint molecules were up-regulated as the Cuscore increased (Fig. 8F). The fGSEA revealed that the Cuscore was prominently correlated to oncogenic hallmark pathways (Fig. 8G).

Post-transcriptional mechanisms involved in Cuscore

Next, this study analyzed miRNAs with differential expression between low and high Cuscore groups. Sixty-nine miRNAs that presented significant differential expression were selected (Additional file 9: Table S8), and KEGG pathway enrichment analysis of their targeted mRNAs was implemented. Consequently, pathways in cancer, cell adhesion molecules (CAMs), Hippo, FoxO and TGF-β signaling pathways were notably enriched (Fig. 8H). Most miRNA-targeted mRNAs in above pathways were up-regulated in high Cuscore group. Above evidence suggested that Cuscore was linked with post-transcriptional mechanisms and pathway regulation.

Identification of Cuscore-related compounds

We further understood the effects of the Cuscore on drug response. The associations of the Cuscore with the response to GDSC-derived compounds were then assessed. The sensitivity to nine drugs (AGI-6780, topotecan, gefitinib, erlotinib, sapitinib, ibrutinib, Eg5_9814, palbociclib and KRAS (G12C) Inhibitor-12) correlated to the high Cuscore, with the correlation between P22077 resistance and the high Cuscore (Fig. 9A). Next, we investigated the pathways involved in these drugs. As illustrated in Fig. 9B, drugs whose sensitivity was linked with the high Cuscore were mainly targeting cell cycle, DNA replication, EGFR and ERK MAPK signaling. In addition, the CTRP and PRISM databases were utilized to predict potential compounds for ccRCC. CR-1-31B, leptomycin B, paclitaxel, vincristine, ouabain, BI-2536, methotrexate, combretastatin-A-4, cabazitaxel, vincristine, PHA-793887, romidepsin, dolastatin-10, gemcitabine, and YM-155 were more suitable for patients with low Cuscore (Fig. 9C, D). Altogether, our findings implied that cuproptosis was correlated to drug response. Therefore, the Cuscore might be a potential biomarker for formulating appropriate therapeutic schedule.

Identification of potential druggable targets for patients with low Cuscore

Potentially druggable targets for patients with low Cuscore were identified. Firstly, correlation between the Cuscore and protein expression of druggable targets was computed, and 23 protein targets were selected according to correlation coefficient < −0.5 and p < 0.05 (Fig. 9E). Then, we conducted correlation analysis of CERES score with the Cuscore, and further screened 486 targets based on correlation coefficient > 0.75 and p < 0.05 (Fig. 9F). Two genes (NMU, RARRES1) were finally determined as potential therapeutic targets by above methods.

Discussion

Copper is a trace metal in cells that is indispensable for maintaining the function of proteins. Nonetheless, excess copper can result in cytotoxicity [38]. Zvetkov et al. firstly proposed a copper accumulation-dependent cuproptosis, which is distinct from other cell death mechanisms [12]. Renal cell carcinoma (RCC) is essentially a metabolic disease characterized by a reprogramming of energetic metabolism [3942]. In particular the metabolic flux through glycolysis is partitioned [4345], and mitochondrial bioenergetics and OxPhox are impaired, as well as lipid metabolism [43, 4648]. Moreover, accumulative researches have been demonstrated that cuproptosis play an important role as regulators of cell metabolism [4951]. Therefore, this study conducted comprehensive analysis of the role of cuproptosis in ccRCC. We described the genomic and transcriptional alterations of 10 cuproptosis genes in ccRCC relative to normal tissues, and observed that methylation and SCNA might result in the aberrant expression of cuproptosis genes. Consistent with previous research [13, 52], most cuproptosis genes (FDX1, LIAS, LIPT1, DLD, DLAT, PDHA1, PDHB, MTF1, and GLS) correlated to more favorable OS outcome of ccRCC, with opposite effect of CDKN2A, indicating their critical implications in ccRCC prognosis.
Based on the transcriptome profiling of cuproptosis genes, we defined the three cuproptosis subtypes (C1 (moderate cuproptosis), C2 (low cuproptosis), and C3 (high cuproptosis)) in ccRCC. NTP approach demonstrated that the cuproptosis-based classification could be effectively repeated in the TCGA dataset and externally verified in the E-MTAB-1980 dataset, proving that this classification was highly repeatable. OS was best in C3, moderate in C1, and worst in C2, indicating that cuproptosis might contribute to favorable survival outcome of ccRCC. Large-scale genomics research has characterized global somatic alteration features in ccRCC, and their relationships with prognosis [53]. Chromosome 3p loss leads to inactivation of several tumor suppressor genes (VHL, PBRM1, etc.), which has been defined as an early driver event in ccRCC [54]. The heterogeneity in genomic alterations occurred in the three cuproptosis subtypes. C1 exhibited higher SCNA, CTA score, homologous recombination defects, and intratumor heterogeneity than others.
The tumor microenvironment is a complex ecosystem comprising heterogeneous cell types. The complex interaction between renal cancer cells and the surrounding tumor microenvironment results in the remarkable intratumoral heterogeneity of ccRCC. In addition, renal cell carcinoma is one of the most immune-infiltrated tumors [55, 56]. Emerging evidence suggests that the activation of specific metabolic pathway have a role in regulating angiogenesis and inflammatory signatures [57, 58]. Features of the tumor microenvironment heavily affect disease biology and may affect responses to systemic therapy [59]. Accumulating evidence proves the essential role of cuproptosis in tumor immunity [6062], but the mechanisms of cuproptosis molecules in ccRCC remain indistinct. Our evidence indicated the high correlation of cuproptosis subtypes with the tumor microenvironment. For example, subtype C1 and C2 were corresponding to a high immune cell infiltration, while C3 accompanied by an immune-desert type.
Moreover, patients in C1 benefited more from anti-PD-1 immunotherapy. Additionally, C1 patients were most sensitive to pazopanib, and sorafenib, and C2 patients had the highest sensitivity to Sunitinib. Altogether, this cuproptosis-based classification might assist therapeutic options for ccRCC patients.
To improve prognostic outcome of ccRCC, understanding of the molecular underpinnings within the spectrum of ccRCC progression is required. We defined the Cuscore system to quantify cuproptosis of individual ccRCC patients. High Cuscore group presented the remarkable survival advantage. Additionally, the Cuscore was highly associated with immunogenomic features, and post-transcriptional events that contributed to ccRCC development. Several potential compounds and druggable targets (NMU, RARRES1) were determined for patients with low Cuscore. NMU is a neuropeptide implicated in energy homeostasis and tumor progression. VHL inactivation leads to remarkable upregulation of NMU in renal cancer cells, and NMU facilitates renal cancer progression through an autocrine effect [63]. RARRES1 can regulate podocyte function, and its expression is up-regulated in glomerular diseases and correlated to disease progression [64]. Highly expressed RARRES1 results in podocyte apoptosis by autocrine and paracrine effects [65]. Of note, RARRES1 expression is stronger in high grade versus low-grade RCC, and RARRES1-positive patients have poorer OS [66]. Combining the existing evidence, NMU and RARRES1 might be potential druggable targets of ccRCC patients with low Cuscore.
Of course, we recognize some limitations of this study. This is a retrospective study. Although we observed the heterogeneity in cuproptosis in as many ccRCC patients as possible, multicenter clinical cohorts are required for further analysis and validation. Our data suggested that cuproptosis may play a crucial role in ccRCC, but the molecular mechanisms involved are still understudied. This is a study of basic research and more in vitro and in vivo studies are necessary in the future.

Conclusion

Altogether, our findings proposed three different cuproptosis subtypes that provided a novel insight into the relationships of cuproptosis with clinical, molecular, and immune characteristics of ccRCC. In addition, we generated the reliable Cuscore that enabled to accurately predict the survival outcomes and immunotherapeutic response in patients with ccRCC. This work provides a roadmap for patients’ stratification, and may help inform personalized follow-up and individualized decision-making strategies for ccRCC immunotherapy, and advance the development of precision immuno-oncology.

Declarations

Not applicable.

Competing interests

The authors declare that there is no competing 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.
Zurück zum Zitat Motzer RJ, Jonasch E, Agarwal N, et al. Kidney cancer, Version 3.2022, NCCN clinical practice guidelines in oncology. J Natl Compr Canc Netw. 2022;20:71–90.PubMedPubMedCentralCrossRef Motzer RJ, Jonasch E, Agarwal N, et al. Kidney cancer, Version 3.2022, NCCN clinical practice guidelines in oncology. J Natl Compr Canc Netw. 2022;20:71–90.PubMedPubMedCentralCrossRef
2.
Zurück zum Zitat Hu J, Chen Z, Bao 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, et al. Single-cell transcriptome analysis reveals intratumoral heterogeneity in ccRCC, which results in different clinical outcomes. Mol Ther. 2020;28:1658–72.PubMedPubMedCentralCrossRef
3.
Zurück zum Zitat Long Z, Sun C, Tang M, et al. Single-cell multiomics analysis reveals regulatory programs in clear cell renal cell carcinoma. Cell Discov. 2022;8:68.PubMedPubMedCentralCrossRef Long Z, Sun C, Tang M, et al. Single-cell multiomics analysis reveals regulatory programs in clear cell renal cell carcinoma. Cell Discov. 2022;8:68.PubMedPubMedCentralCrossRef
4.
Zurück zum Zitat Udayakumar D, Zhang Z, Xi Y, et al. Deciphering intratumoral molecular heterogeneity in clear cell renal cell carcinoma with a radiogenomics platform. Clin Cancer Res. 2021;27:4794–806.PubMedPubMedCentralCrossRef Udayakumar D, Zhang Z, Xi Y, et al. Deciphering intratumoral molecular heterogeneity in clear cell renal cell carcinoma with a radiogenomics platform. Clin Cancer Res. 2021;27:4794–806.PubMedPubMedCentralCrossRef
5.
Zurück zum Zitat Di Lascio G, Sciarra A, Giudice F, et al. Which factors can influence post-operative renal function preservation after nephron-sparing surgery for kidney cancer: a critical review. Cent Eur J Urol. 2022;75:14–27. Di Lascio G, Sciarra A, Giudice F, et al. Which factors can influence post-operative renal function preservation after nephron-sparing surgery for kidney cancer: a critical review. Cent Eur J Urol. 2022;75:14–27.
6.
Zurück zum Zitat Ferro M, Musi G, Marchioni M, et al. Radiogenomics in renal cancer management-current evidence and future prospects. Int J Mol Sci. 2023;24:4615.PubMedPubMedCentralCrossRef Ferro M, Musi G, Marchioni M, et al. Radiogenomics in renal cancer management-current evidence and future prospects. Int J Mol Sci. 2023;24:4615.PubMedPubMedCentralCrossRef
9.
Zurück zum Zitat Linehan WM, Ricketts CJ. The Cancer Genome Atlas of renal cell carcinoma: findings and clinical implications. Nat Rev Urol. 2019;16:539–52.PubMedCrossRef Linehan WM, Ricketts CJ. The Cancer Genome Atlas of renal cell carcinoma: findings and clinical implications. Nat Rev Urol. 2019;16:539–52.PubMedCrossRef
10.
Zurück zum Zitat Ge EJ, Bush AI, Casini A, et al. Connecting copper and cancer: from transition metal signalling to metalloplasia. Nat Rev Cancer. 2022;22:102–13.PubMedCrossRef Ge EJ, Bush AI, Casini A, et al. Connecting copper and cancer: from transition metal signalling to metalloplasia. Nat Rev Cancer. 2022;22:102–13.PubMedCrossRef
11.
Zurück zum Zitat Gupte A, Mumper RJ. Elevated copper and oxidative stress in cancer cells as a target for cancer treatment. Cancer Treat Rev. 2009;35:32–46.PubMedCrossRef Gupte A, Mumper RJ. Elevated copper and oxidative stress in cancer cells as a target for cancer treatment. Cancer Treat Rev. 2009;35:32–46.PubMedCrossRef
12.
Zurück zum Zitat Zhang H, Meltzer P, Davis S. RCircos: an R package for Circos 2D track plots. BMC Bioinform. 2013;14:244.CrossRef Zhang H, Meltzer P, Davis S. RCircos: an R package for Circos 2D track plots. BMC Bioinform. 2013;14:244.CrossRef
13.
Zurück zum Zitat Bian Z, Fan R, Xie L. A novel cuproptosis-related prognostic gene signature and validation of differential expression in clear cell renal cell carcinoma. Genes (Basel). 2022;13:851.PubMedCrossRef Bian Z, Fan R, Xie L. A novel cuproptosis-related prognostic gene signature and validation of differential expression in clear cell renal cell carcinoma. Genes (Basel). 2022;13:851.PubMedCrossRef
14.
Zurück zum Zitat Xu S, Liu D, Chang T, et al. Cuproptosis-associated lncRNA establishes new prognostic profile and predicts immunotherapy response in clear cell renal cell carcinoma. Front Genet. 2022;13:938259.PubMedPubMedCentralCrossRef Xu S, Liu D, Chang T, et al. Cuproptosis-associated lncRNA establishes new prognostic profile and predicts immunotherapy response in clear cell renal cell carcinoma. Front Genet. 2022;13:938259.PubMedPubMedCentralCrossRef
15.
16.
Zurück zum Zitat Mayakonda A, Lin DC, Assenov Y, et al. Maftools: efficient and comprehensive analysis of somatic variants in cancer. Genome Res. 2018;28:1747–56.PubMedPubMedCentralCrossRef Mayakonda A, Lin DC, Assenov Y, et al. Maftools: efficient and comprehensive analysis of somatic variants in cancer. Genome Res. 2018;28:1747–56.PubMedPubMedCentralCrossRef
17.
Zurück zum Zitat Mermel CH, Schumacher SE, Hill B, et al. GISTIC2.0 facilitates sensitive and confident localization of the targets of focal somatic copy-number alteration in human cancers. Genome Biol. 2011;12:R41.PubMedPubMedCentralCrossRef Mermel CH, Schumacher SE, Hill B, et al. GISTIC2.0 facilitates sensitive and confident localization of the targets of focal somatic copy-number alteration in human cancers. Genome Biol. 2011;12:R41.PubMedPubMedCentralCrossRef
19.
Zurück zum Zitat Subramanian A, Tamayo P, Mootha VK, 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, 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
20.
Zurück zum Zitat Wu T, Hu E, Xu S, et al. clusterProfiler 4.0: a universal enrichment tool for interpreting omics data. Innovation (Camb). 2021;2:100141.PubMedPubMedCentral Wu T, Hu E, Xu S, et al. clusterProfiler 4.0: a universal enrichment tool for interpreting omics data. Innovation (Camb). 2021;2:100141.PubMedPubMedCentral
21.
22.
Zurück zum Zitat Liberzon A, Birger C, Thorvaldsdóttir H, et al. The Molecular Signatures Database (MSigDB) hallmark gene set collection. Cell Syst. 2015;1:417–25.PubMedPubMedCentralCrossRef Liberzon A, Birger C, Thorvaldsdóttir H, et al. The Molecular Signatures Database (MSigDB) hallmark gene set collection. Cell Syst. 2015;1:417–25.PubMedPubMedCentralCrossRef
23.
Zurück zum Zitat Wilkerson MD, Hayes DN. ConsensusClusterPlus: a class discovery tool with confidence assessments and item tracking. Bioinformatics. 2010;26:1572–3.PubMedPubMedCentralCrossRef Wilkerson MD, Hayes DN. ConsensusClusterPlus: a class discovery tool with confidence assessments and item tracking. Bioinformatics. 2010;26:1572–3.PubMedPubMedCentralCrossRef
24.
Zurück zum Zitat Ritchie ME, Phipson B, Wu D, 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, et al. limma powers differential expression analyses for RNA-sequencing and microarray studies. Nucleic Acids Res. 2015;43:e47.PubMedPubMedCentralCrossRef
25.
27.
Zurück zum Zitat Malta TM, Sokolov A, Gentles AJ, et al. Machine learning identifies stemness features associated with oncogenic dedifferentiation. Cell. 2018;173:338-354.e315.PubMedPubMedCentralCrossRef Malta TM, Sokolov A, Gentles AJ, et al. Machine learning identifies stemness features associated with oncogenic dedifferentiation. Cell. 2018;173:338-354.e315.PubMedPubMedCentralCrossRef
28.
Zurück zum Zitat Yang W, Soares J, Greninger P, et al. Genomics of Drug Sensitivity in Cancer (GDSC): a resource for therapeutic biomarker discovery in cancer cells. Nucleic Acids Res. 2013;41:D955-961.PubMedCrossRef Yang W, Soares J, Greninger P, et al. Genomics of Drug Sensitivity in Cancer (GDSC): a resource for therapeutic biomarker discovery in cancer cells. Nucleic Acids Res. 2013;41:D955-961.PubMedCrossRef
29.
Zurück zum Zitat Yu C, Mannan AM, Yvone GM, et al. High-throughput identification of genotype-specific cancer vulnerabilities in mixtures of barcoded tumor cell lines. Nat Biotechnol. 2016;34:419–23.PubMedPubMedCentralCrossRef Yu C, Mannan AM, Yvone GM, et al. High-throughput identification of genotype-specific cancer vulnerabilities in mixtures of barcoded tumor cell lines. Nat Biotechnol. 2016;34:419–23.PubMedPubMedCentralCrossRef
30.
Zurück zum Zitat Barretina J, Caponigro G, Stransky N, et al. The cancer cell line encyclopedia enables predictive modelling of anticancer drug sensitivity. Nature. 2012;483:603–7.PubMedPubMedCentralCrossRef Barretina J, Caponigro G, Stransky N, et al. The cancer cell line encyclopedia enables predictive modelling of anticancer drug sensitivity. Nature. 2012;483:603–7.PubMedPubMedCentralCrossRef
32.
Zurück zum Zitat Geeleher P, Cox N, Huang RS. pRRophetic: an R package for prediction of clinical chemotherapeutic response from tumor gene expression levels. PLoS ONE. 2014;9:e107468.PubMedPubMedCentralCrossRef Geeleher P, Cox N, Huang RS. pRRophetic: an R package for prediction of clinical chemotherapeutic response from tumor gene expression levels. PLoS ONE. 2014;9:e107468.PubMedPubMedCentralCrossRef
33.
Zurück zum Zitat Charoentong P, Finotello F, Angelova M, et al. Pan-cancer immunogenomic analyses reveal genotype-immunophenotype relationships and predictors of response to checkpoint blockade. Cell Rep. 2017;18:248–62.PubMedCrossRef Charoentong P, Finotello F, Angelova M, et al. Pan-cancer immunogenomic analyses reveal genotype-immunophenotype relationships and predictors of response to checkpoint blockade. Cell Rep. 2017;18:248–62.PubMedCrossRef
34.
Zurück zum Zitat Yoshihara K, Shahmoradgoli M, Martínez E, et al. Inferring tumour purity and stromal and immune cell admixture from expression data. Nat Commun. 2013;4:2612.PubMedCrossRef Yoshihara K, Shahmoradgoli M, Martínez E, et al. Inferring tumour purity and stromal and immune cell admixture from expression data. Nat Commun. 2013;4:2612.PubMedCrossRef
35.
Zurück zum Zitat Liu Z, Zhang Y, Shi C, et al. A novel immune classification reveals distinct immune escape mechanism and genomic alterations: implications for immunotherapy in hepatocellular carcinoma. J Transl Med. 2021;19:5.PubMedPubMedCentralCrossRef Liu Z, Zhang Y, Shi C, et al. A novel immune classification reveals distinct immune escape mechanism and genomic alterations: implications for immunotherapy in hepatocellular carcinoma. J Transl Med. 2021;19:5.PubMedPubMedCentralCrossRef
36.
38.
Zurück zum Zitat Wang Y, Zhang L, Zhou F. Cuproptosis: a new form of programmed cell death. Cell Mol Immunol. 2022;19:867–8.PubMedCrossRef Wang Y, Zhang L, Zhou F. Cuproptosis: a new form of programmed cell death. Cell Mol Immunol. 2022;19:867–8.PubMedCrossRef
39.
Zurück zum Zitat Meo N, Lasorsa F, Rutigliano 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.PubMedCrossRef Meo N, Lasorsa F, Rutigliano 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.PubMedCrossRef
40.
Zurück zum Zitat Lucarelli G, Loizzo D, Franzin R, et al. Metabolomic insights into pathophysiological mechanisms and biomarker discovery in clear cell - renal cell carcinoma. Expert Rev Mol Diagn. 2019;19:397.PubMedCrossRef Lucarelli G, Loizzo D, Franzin R, et al. Metabolomic insights into pathophysiological mechanisms and biomarker discovery in clear cell - renal cell carcinoma. Expert Rev Mol Diagn. 2019;19:397.PubMedCrossRef
41.
Zurück zum Zitat Meo N, Lasorsa F, Rutigliano M, 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 Meo N, Lasorsa F, Rutigliano M, 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
42.
Zurück zum Zitat Marco S, Torsello B, Minutiello E, et al. The cross-talk between Abl2 tyrosine kinase and TGFβ1 signalling modulates the invasion of clear cell Renal Cell Carcinoma cells. FEBS Lett. 2022;597:1098.PubMedCrossRef Marco S, Torsello B, Minutiello E, et al. The cross-talk between Abl2 tyrosine kinase and TGFβ1 signalling modulates the invasion of clear cell Renal Cell Carcinoma cells. FEBS Lett. 2022;597:1098.PubMedCrossRef
43.
Zurück zum Zitat Bianchi C, Meregalli C, Bombelli S, 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.PubMedPubMedCentralCrossRef Bianchi C, Meregalli C, Bombelli S, 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.PubMedPubMedCentralCrossRef
44.
Zurück zum Zitat Ragone R, Sallustio F, Piccinonna 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, et al. Renal cell carcinoma: a study through NMR-based metabolomics combined with transcriptomics. Diseases. 2016;4:7.PubMedPubMedCentralCrossRef
45.
Zurück zum Zitat Lucarelli G, Galleggiante V, Rutigliano M, 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.PubMedPubMedCentralCrossRef Lucarelli G, Galleggiante V, Rutigliano M, 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.PubMedPubMedCentralCrossRef
46.
Zurück zum Zitat Lucarelli G, Rutigliano M, Sallustio F, 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. 2018;10:3957.PubMedPubMedCentralCrossRef Lucarelli G, Rutigliano M, Sallustio F, 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. 2018;10:3957.PubMedPubMedCentralCrossRef
47.
Zurück zum Zitat Bombelli S, Torsello B, Marco S, et al. 36-kDa Annexin A3 isoform negatively modulates lipid storage in clear cell renal cell carcinoma cells. Am J Pathol. 2020;190:2317.PubMedCrossRef Bombelli S, Torsello B, Marco S, et al. 36-kDa Annexin A3 isoform negatively modulates lipid storage in clear cell renal cell carcinoma cells. Am J Pathol. 2020;190:2317.PubMedCrossRef
48.
Zurück zum Zitat Lucarelli G, Rutigliano M, Loizzo D, et al. MUC1 tissue expression and its soluble form CA 15–3 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, et al. MUC1 tissue expression and its soluble form CA 15–3 identify a clear cell renal cell carcinoma with distinct metabolic profile and poor clinical outcome. Int J Mol Sci. 2022;23:13968.PubMedPubMedCentralCrossRef
51.
Zurück zum Zitat Ke D, Zhang Z, Liu J, et al. Ferroptosis, necroptosis and cuproptosis: Novel forms of regulated cell death in diabetic cardiomyopathy. Front Cardiovasc Med. 2023;10:1135723.PubMedPubMedCentralCrossRef Ke D, Zhang Z, Liu J, et al. Ferroptosis, necroptosis and cuproptosis: Novel forms of regulated cell death in diabetic cardiomyopathy. Front Cardiovasc Med. 2023;10:1135723.PubMedPubMedCentralCrossRef
52.
Zurück zum Zitat Khouja HI, Ashankyty IM, Bajrai LH, et al. Multi-staged gene expression profiling reveals potential genes and the critical pathways in kidney cancer. Sci Rep. 2022;12:7240.PubMedPubMedCentralCrossRef Khouja HI, Ashankyty IM, Bajrai LH, et al. Multi-staged gene expression profiling reveals potential genes and the critical pathways in kidney cancer. Sci Rep. 2022;12:7240.PubMedPubMedCentralCrossRef
53.
Zurück zum Zitat Patel SA, Hirosue S, Rodrigues P, et al. The renal lineage factor PAX8 controls oncogenic signalling in kidney cancer. Nature. 2022;606:999–1006.PubMedPubMedCentralCrossRef Patel SA, Hirosue S, Rodrigues P, et al. The renal lineage factor PAX8 controls oncogenic signalling in kidney cancer. Nature. 2022;606:999–1006.PubMedPubMedCentralCrossRef
54.
55.
56.
Zurück zum Zitat Tamma R, Rutigliano M, Lucarelli G, 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, 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
57.
Zurück zum Zitat Stefano N, Lucarelli G, Spadaccino F, et al. PTX3 modulates the immunoflogosis in tumor microenvironment and is a prognostic factor for patients with clear cell renal cell carcinoma. Aging. 2020;12:7585.CrossRef Stefano N, Lucarelli G, Spadaccino F, et al. PTX3 modulates the immunoflogosis in tumor microenvironment and is a prognostic factor for patients with clear cell renal cell carcinoma. Aging. 2020;12:7585.CrossRef
59.
Zurück zum Zitat Ghini V, Laera L, Fantechi B, et al. Metabolomics to assess response to immune checkpoint inhibitors in patients with non-small-cell lung cancer. Cancers. 2020;12:3574.PubMedPubMedCentralCrossRef Ghini V, Laera L, Fantechi B, et al. Metabolomics to assess response to immune checkpoint inhibitors in patients with non-small-cell lung cancer. Cancers. 2020;12:3574.PubMedPubMedCentralCrossRef
60.
Zurück zum Zitat Lv H, Liu X, Zeng X, et al. Comprehensive analysis of cuproptosis-related genes in immune infiltration and prognosis in melanoma. Front Pharmacol. 2022;13:930041.PubMedPubMedCentralCrossRef Lv H, Liu X, Zeng X, et al. Comprehensive analysis of cuproptosis-related genes in immune infiltration and prognosis in melanoma. Front Pharmacol. 2022;13:930041.PubMedPubMedCentralCrossRef
61.
Zurück zum Zitat Song Q, Zhou R, Shu F, Fu W. Cuproptosis scoring system to predict the clinical outcome and immune response in bladder cancer. Front Immunol. 2022;13:958368.PubMedPubMedCentralCrossRef Song Q, Zhou R, Shu F, Fu W. Cuproptosis scoring system to predict the clinical outcome and immune response in bladder cancer. Front Immunol. 2022;13:958368.PubMedPubMedCentralCrossRef
62.
Zurück zum Zitat Zhang Z, Zeng X, Wu Y, et al. Cuproptosis-related risk score predicts prognosis and characterizes the tumor microenvironment in hepatocellular carcinoma. Front Immunol. 2022;13:925618.PubMedPubMedCentralCrossRef Zhang Z, Zeng X, Wu Y, et al. Cuproptosis-related risk score predicts prognosis and characterizes the tumor microenvironment in hepatocellular carcinoma. Front Immunol. 2022;13:925618.PubMedPubMedCentralCrossRef
63.
Zurück zum Zitat Harten SK, Esteban MA, Shukla D, et al. Inactivation of the von Hippel-Lindau tumour suppressor gene induces Neuromedin U expression in renal cancer cells. Mol Cancer. 2011;10:89.PubMedPubMedCentralCrossRef Harten SK, Esteban MA, Shukla D, et al. Inactivation of the von Hippel-Lindau tumour suppressor gene induces Neuromedin U expression in renal cancer cells. Mol Cancer. 2011;10:89.PubMedPubMedCentralCrossRef
64.
Zurück zum Zitat Chen A, Feng Y, Lai H, et al. Soluble RARRES1 induces podocyte apoptosis to promote glomerular disease progression. J Clin Invest. 2020;130:5523–35.PubMedPubMedCentralCrossRef Chen A, Feng Y, Lai H, et al. Soluble RARRES1 induces podocyte apoptosis to promote glomerular disease progression. J Clin Invest. 2020;130:5523–35.PubMedPubMedCentralCrossRef
66.
Zurück zum Zitat Zimpfer A, Dammert F, Glass A, et al. Expression and clinicopathological correlations of retinoid acid receptor responder protein 1 in renal cell carcinomas. Biomark Med. 2016;10:721–32.PubMedCrossRef Zimpfer A, Dammert F, Glass A, et al. Expression and clinicopathological correlations of retinoid acid receptor responder protein 1 in renal cell carcinomas. Biomark Med. 2016;10:721–32.PubMedCrossRef
Metadaten
Titel
Multi-omics analysis uncovers clinical, immunological, and pharmacogenomic implications of cuproptosis in clear cell renal cell carcinoma
verfasst von
Maoshu Zhu
Yongsheng Li
Yun Wang
Pingli Lin
Jun Mi
Weimin Zhong
Publikationsdatum
01.12.2023
Verlag
BioMed Central
Erschienen in
European Journal of Medical Research / Ausgabe 1/2023
Elektronische ISSN: 2047-783X
DOI
https://doi.org/10.1186/s40001-023-01221-4

Weitere Artikel der Ausgabe 1/2023

European Journal of Medical Research 1/2023 Zur Ausgabe