Abstract
Background/Objectives: Cellular senescence plays a critical role in
tumorigenesis, immune cell infiltration, and treatment response.
Adrenocortical carcinoma (ACC) is a malignant tumor that lacks
effective therapies. This study aimed to construct and validate a
senescence-related gene signature as an independent prognostic
predictor for ACC and explore its impact on the tumor microenvironment,
immunotherapy, and chemotherapy response. Methods: Data were collected
from The Cancer Genome Atlas (TCGA) and the Gene Expression Omnibus
(GEO) database. Using Kaplan–Meier survival analysis, LASSO penalized
Cox regression and multivariable Cox regression, we identified a
prognostic model with four senescence-related genes (HJURP, CDK1,
FOXM1, and CHEK1). The model’s prognostic value was validated through
survival analysis, risk score curves, and receiver operating
characteristic (ROC) curves. Tumor mutation burden was assessed with
maftools, and the tumor microenvironment was analyzed using CIBERSORT
and ESTIMATE. Immune and chemotherapeutic responses were assessed
through Tumor Immune Dysfunction and Exclusion (TIDE) and OncoPredict.
Results: The risk score derived from our model showed a strong
association with overall survival (OS) in ACC patients (p < 0.001, HR =
2.478). Higher risk scores were correlated with more advanced tumor
stages and a greater frequency of somatic mutations. Differentially
expressed genes (DEGs) that were downregulated in the high-risk group
were significantly enriched in immune-related pathways. Furthermore,
high-risk patients were predicted to have reduced sensitivity to
immunotherapy (p = 0.02). Bioinformatics analysis identified potential
chemotherapeutic agents, including BI-2536 and MIM1, as more effective
treatment options for high-risk patients. Conclusions: Our findings
indicate that this prognostic model may serve as a valuable tool for
predicting overall survival (OS) and treatment responses in ACC
patients, including those receiving chemotherapy and immunotherapy.
Keywords: adrenocortical carcinoma, senescence, prognosis, immune
microenvironment, drug sensitivity
1. Introduction
Adrenocortical carcinoma (ACC) is a rare but aggressive endocrine
cancer with limited treatment options. Its incidence is estimated at
0.7 to 2 cases per million annually, with a higher prevalence in
females (female-to-male ratio of 1.5 to 2.5:1) and peak onset in the
40s and 50s [[30]1,[31]2,[32]3]. While surgical resection remains the
primary method of approach, recurrence rates are high, reaching up to
75% even after complete resection [[33]2]. Currently, mitotane is the
only FDA-approved drug specifically for ACC. For patients with
low-grade, localized ACC, post-surgical surveillance is generally
preferred over mitotane therapy [[34]4], while effective options for
advanced-stage ACC remain limited. Despite efforts to introduce new
therapies for ACC—including combination chemotherapy (etoposide,
doxorubicin, cisplatin, and mitotane) [[35]5], immunotherapy
[[36]6,[37]7], tyrosine kinase inhibitors [[38]8,[39]9], and
radiotherapy [[40]10,[41]11]—none have demonstrated significant
efficacy in improving outcomes. Therefore, enhanced prognostic tools
and biomarkers to better stratify ACC patients are urgently needed to
advance precision medicine in this challenging malignancy.
Cellular senescence, marked by an irreversible cessation of cell
division, was initially regarded as a natural defense against tumor
growth. However, recent studies have shown that senescent tumor cells
can persist, releasing a collection of factors known as the
senescence-associated secretory phenotype (SASP), which primarily
comprises proinflammatory chemokines and cytokines
[[42]12,[43]13,[44]14]. These SASP factors are increasingly recognized
as a double-edged sword in cancer, capable of both promoting anti-tumor
immune cell infiltration and driving tumor progression by influencing
the behavior of nearby cells [[45]15,[46]16]. As a result, some
researchers suggest the following novel strategy: first induce tumor
cell senescence with a senescence-inducing agent, followed by the
targeted elimination of these senescent cells using senolytic
modalities [[47]17]. Recent animal studies on ACC have shown that
genetic mutations can trigger cellular senescence, contributing to
adrenal cancer development in older mice [[48]18,[49]19]. Further
research has demonstrated an upregulation of the aging biomarkers p21
and p53 in ACC, along with elevated Ki-67 expression, when compared to
adrenocortical adenomas (ACA) [[50]20,[51]21]. However, the exact role
of senescence in ACC remains unclear, as it could function either as a
protective or a risk factor. Therefore, we hypothesize that gene
expression related to senescence may provide a foundation for
developing a model to predict ACC progression and guide treatment
strategies. In this study, we developed a four-gene signature model
based on senescence-associated genes to predict outcomes in ACC. Using
data from the TCGA-ACC and GEO databases, we assessed the correlation
between these genes and ACC prognosis. Furthermore, we explored how the
signature relates to clinical features, immune cell infiltration,
immune therapy responses, and drug sensitivity in ACC patients. Our
findings provide insights into the association between these genes and
the prognosis of ACC, which may inform future studies on their
potential role in cellular senescence and therapeutic strategies.
2. Materials and Methods
2.1. Data Acquisition
A cellular senescence-associated gene set, consisting of 278 genes
([52]Supplementary Table S1), was sourced from the CellAge database
([53]http://genomics.senescence.info/cells, accessed on 23 April 2023).
To identify differentially expressed genes (DEGs) between ACC and
normal adrenal gland tissue, RNA-seq data from the TCGA-ACC (n = 77)
and Genotype-Tissue Expression (GTEx) (n = 126) datasets were obtained
from the UCSC Toil RNA-seq Recompute project
([54]https://xenabrowser.net/datapages/, accessed on 23 April 2023).
Additionally, DEGs were derived from the [55]GSE12368 and [56]GSE143383
datasets. RNA sequencing data and patient clinical characteristics were
collected from TCGA-ACC database to establish the construction cohort
([57]https://cancergenome.nih.gov/, accessed on 23 April 2023).
Transcriptomic microarray data and survival information for two
validation cohorts were downloaded from the GEO database, specifically
from the [58]GSE10927 and [59]GSE19750 datasets
([60]https://www.ncbi.nlm.nih.gov/geo/, accessed on 23 April 2023).
2.2. Establishment and Validation of Aging-Associated Gene Signature
To identify DEGs between tumor and normal tissues, we conducted
differential expression analysis on the TCGA-ACC and GTEx datasets
using DESeq2, edgeR, and limma. The combined use of these three
packages helped mitigate potential computational biases that could
arise from relying on a single R package. Genes with an adjusted
p-value < 0.05 and a log fold change (logFC) > 1 were selected as DEGs.
To ensure the reliability of our DEG selection, we refined this list by
intersecting DEGs from the three methods with those identified in the
[61]GSE12368 and [62]GSE143383 datasets (analyzed with limma). This
final intersected set of DEGs was visualized using a Venn diagram.
Next, aging-associated prognostic genes were identified through
univariate Cox regression analysis. We then intersected these
prognostic genes with the final set of DEGs, followed by LASSO
regression and Cox proportional hazards modeling (coxph) to construct
our predictive model. The risk model was calculated as follows:
[MATH: risk
mo>score=coefficient 1×Express
ion 1 +<
/mo>coeffic
mi>ient 2×Expression 2+…
+<
/mo>coeffic
mi>ient i×Expression i :MATH]
(1)
Multivariate Cox regression analyses were performed to assess the
independence of the risk score in predicting survival outcomes. In both
the construction (TCGA-ACC) and validation cohorts ([63]GSE19750 and
[64]GSE10927), patients were divided into low-risk and high-risk groups
based on the median risk score. Kaplan–Meier (KM) curves were generated
using the survival package to assess survival outcomes for patients
stratified by risk score. In addition, time-dependent receiver
operating characteristic (ROC) curves were constructed with the
survivalROC package to evaluate the predictive performance of the risk
signature.
2.3. Mutation Analysis
The mutation waterfall plot illustrating the mutation profiles of
TCGA-ACC patients across two risk subgroups was generated using the R
package maftools.
2.4. Differential Gene and Enrichment Analysis Between High-Risk and Low-Risk
Groups
Differential gene expression analysis between high-risk and low-risk
groups was conducted using DESeq2, with a logFC threshold of 1 and a
significance cutoff at p < 0.05. To investigate the biological
functions of the identified DEGs, Gene Set Enrichment Analysis (GSEA)
was performed to identify enriched pathways using the KEGG subset,
while upregulated and downregulated genes in the high-risk group were
analyzed through the WikiPathways subset. Additionally, Gene Ontology
(GO), and the Kyoto Encyclopedia of Genes and Genomes (KEGG) were also
performed with the clusterProfiler package.
2.5. Immune-Related Signature Profiling
We applied ssGSEA to analyze the gene expression signature of 28
distinct immune cell types ([65]Supplementary Table S2), and used
CIBERSORT ([66]https://cibersort.stanford.edu, accessed on 23 April
2023) to assess immune cell infiltration based on the gene signatures
of 22 immune cell types. Both methods were employed to comprehensively
evaluate immune cell infiltration across different stratified risk
groups. Additionally, ESTIMATE (Estimation of STromal and Immune cells
in MAlignant Tumor tissues using Expression data) was used to evaluate
tumor purity by quantifying the proportion of immune and stromal cells
within the tumor microenvironment. We also systematically explored the
correlations between senescence-related genes in our model and genes
involved in antigen processing and presentation, immune checkpoint
genes, and phagocytosis signaling.
2.6. Immunotherapy Response
Tumor Immune Dysfunction and Exclusion (TIDE)
([67]http://tide.dfci.harvard.edu/, accessed on 23 April 2023) was
employed to predict patient immunotherapy response by estimating
multiple published transcriptomic biomarkers.
2.7. Drug Response Prediction
To identify potential drugs for high-risk patients, we employed the
oncoPredict package (Version 1.2, updated on 5 April 2024) to predict
the sensitivity of 198 drugs across two risk groups based on the
transcriptomic data associated with half-maximal inhibitory
concentration (IC50).
2.8. Statistical Analysis
R version 4.4.0 and SPSS Statistics V27.0 were utilized for data
analysis and graph generation. For comparing clinical characteristics
between the high- and low-risk groups, an unpaired Student’s t-test was
employed for normally distributed variables, while non-normally
distributed variables were analyzed using the Wilcoxon rank-sum test.
Categorical variables were compared using the χ^2 test. Statistical
significance was defined as p < 0.05. A flowchart of our study is shown
in [68]Figure 1.
Figure 1.
[69]Figure 1
[70]Open in a new tab
The flow of diagram of our study.
3. Results
3.1. Aging-Related Prognostic Gene Screening and Senescence Model
Construction in ACC
Using RNA-seq count data from TCGA-ACC (n = 77) and GTEx (n = 126), we
identified 2576 upregulated genes through an integrative analysis
combining DESeq2, edgeR, and limma results ([71]Supplementary Figure
S1). We further refined these genes by incorporating upregulated genes
identified from differential expression analysis of the [72]GSE12368
(ACC vs. Normal, 12 vs. 6) (905 upregulated genes) ([73]Supplementary
Figure S2) and [74]GSE143383 (ACC vs. Normal, 57 vs. 5) (333
upregulated genes) ([75]Supplementary Figure S3) datasets, narrowing
the list to 125 genes ([76]Supplementary Figure S4 and Table S3) for
in-depth analysis. To explore the association between cellular
senescence-related genes and survival in TCGA-ACC (n = 77, two patients
diagnosed with sarcomatoid features were excluded), we performed
univariate Cox proportional hazards regression and identified 102 genes
with p < 0.05 ([77]Supplementary Table S4). The intersection of these
125 genes with the 102 significant survival-associated genes resulted
in six aging-related prognostic genes for further investigation
([78]Figure 2A). To develop a prognostic gene signature, we refined
these six candidate genes through Lasso regression analysis using an
optimized λ value, resulting in a risk score formula based on the
expression levels of HJURP, CDK1, FOXM1 and CHEK1 ([79]Figure 2B–E).
The risk score was calculated as follows:
[MATH: risk
mo>score=0.5541×exp
ression<
/mi> of HJU
mi>RP <
/mo>+0.2132×exp
ression<
/mi> of CDK
mi>1 <
/mo>+(0.1329×expression of FO
XM1) <
/mo>+0.001×exp<
/mi>ression
mi> of CHEK1 :MATH]
(2)
Figure 2.
[80]Figure 2
[81]Open in a new tab
Construction and Validation of the Prognostic Model. (A) The Venn
diagram highlights six differential expression genes associated with
prognosis. (B) The forest plot displays the hazard ratios of the six
genes in relation to overall survival (OS). (C) LASSO regression
analysis of OS-related genes. (D) The cross-validation results for
tuning parameter selection in the LASSO regression model. (E) The
forest plot illustrating the multivariate Cox regression analysis of
the four genes included in the final prognostic model. Kaplan–Meier
survival curves, time-dependent ROC curves, distribution of risk
scores, survival status, and expression profiles of the four signature
genes for the TCGA-ACC dataset (F–H). Kaplan–Meier survival curves,
time-dependent ROC curves, distribution of risk scores, survival
status, and expression profiles of the four signature genes for the
[82]GSE19750 dataset (I–K). Kaplan–Meier survival curves,
time-dependent ROC curves, distribution of risk scores, survival
status, and expression profiles of the four signature genes for the
[83]GSE10927 (L–N).
This formula enables patient stratification based on gene expression
levels relative to the median risk score, providing valuable insights
into potential prognostic outcomes. As demonstrated in the KM plot,
patients with higher risk scores had significantly poorer survival
rates than those with lower scores (p < 0.001) ([84]Figure 2F). The ROC
analysis revealed areas under the curve (AUC) of 0.92, 0.93, and 0.85
for 2-, 3-, and 5-year survival, respectively ([85]Figure 2G).
Moreover, the distribution of risk scores, survival status, and gene
expression heatmap for TCGA-ACC patients further highlight the
association between high-risk status and poorer prognosis ([86]Figure
2H). The association between the expression of these four genes and
survival status in ACC patients is detailed in [87]Supplementary Figure
S5.
3.2. Validation the Prognostic Value of the Aging-Related Gene Signature in
Two Independent GEO Cohorts
Two datasets from the GEO database were included to evaluate the
effectiveness of the risk model: [88]GSE19750 (n = 14, platform:
[89]GPL570, raw data in [90]Supplementary Table S5) and [91]GSE10927 (n
= 24, platform: [92]GPL570, raw data in [93]Supplementary Table S6).
Consistent with the predictive cohort, both validation groups showed
that patients in the higher-risk category had worse overall survival
rates. Notably, the KM plot for [94]GSE19750 displayed a significant
difference between the two risk groups (p = 0.03) ([95]Figure 2I), with
ROC curve analysis being performed to assess the model’s predictive
ability, yielding AUC values of 0.71, 0.80, and 0.84 for 2-, 3-, and
5-year survival in [96]GSE19750, respectively ([97]Figure 2J). The
distribution maps of risk score and survival time for [98]GSE19750
further illustrate the model’s capacity to differentiate patient
survival status based on the expression of HJURP, CDK1, FOXM1, and
CHEK1 ([99]Figure 2K). Similar findings were observed in [100]GSE10927,
with the high-risk group showing worse overall survival (OS) compared
to the low-risk group (p = 0.042) ([101]Figure 2L). Due to the limited
number of patients surviving beyond three years (mean survival: 2.49 ±
0.59 years), ROC curve analysis of [102]GSE10927 was conducted for 1-,
2-, and 3-year survival, yielding AUC values of 0.85, 0.78, and 0.65,
respectively ([103]Figure 2M). The lower AUC for 3-year survival may be
attributed to the small number of long-term survivors, with only five
patients living beyond three years. Detailed survival data are provided
in [104]Supplementary Table S7. The survival outcome plots for
[105]GSE10927 further highlight how the model classifies patient
survival based on the expression levels of HJURP, CDK1, FOXM1, and
CHEK1, reinforcing the model’s predictive capacity ([106]Figure 2N).
3.3. Multivariate Cox Proportional Hazards Regression Analysis of the Risk
Score
A comparison of clinical characteristics—including age, gender, hormone
secretion, tumor stage, and resection status—between the high- and
low-risk groups ([107]Table 1 and [108]Figure 3A) revealed
statistically significant differences in tumor stage and resection
status (both p < 0.001). Multivariate Cox regression analysis confirmed
that the risk score was an independent prognostic factor, unaffected by
age, gender, hormone secretion, tumor stage, or resection status
([109]Figure 3B). Due to missing information, the analysis was
conducted on a subset of 70 patients ([110]Supplementary Table S8). No
statistically significant differences were observed in treatment
regimens between the two groups (detailed in [111]Supplementary Table
S9).
Table 1.
Clinical characteristics of high- and low-risk ACC patients.
Low Risk Group
(n = 36) High Risk Group
(n = 34) p Value
Age 46.94 ± 2.62 46.84 ± 2.69 0.335
Gender
Female 23 (63.89%) 23 (67.65%) 0.741
Male 13 (36.11%) 11 (32.35%)
Hormone Secretion
Yes 21 (58.33%) 25 (73.53%) 0.181
No 15 (41.67%) 9 (26.47%)
ENSAT stage <0.001
I 6 (16.67%) 3 (8.82%)
II 24 (66.67%) 10 (29.41%)
III 4 (11.10%) 10 (29.41%)
IV 2 (5.56%) 11 (32.35%)
Resection Status <0.001
R0 34 (94.44%) 16 (47.05%)
R1 0 5 (14.71%)
R2 1 (2.78%) 8 (23.53%)
Rx 1 (2.78%) 5 (14.71%)
[112]Open in a new tab
Figure 3.
[113]Figure 3
[114]Open in a new tab
Clinical features and mutation profile of ACC patients in the two risk
groups. (A) The Sankey diagram shows clinical traits across the two
risk groups. (B) The forest plot depicts the results of a multivariate
Cox regression analysis, highlighting the association between clinical
characteristics and risk score with overall survival.* p < 0.05 (C) The
mutation burden of TCGA-ACC dataset. (D) The waterfall plot visualizes
the characteristics of the top 20 mutation types in the two groups with
violin plot of mutation burden between the two cohorts.
3.4. Mutation Landscape Across the Two Groups
Among the 77 patients in the TCGA-ACC project, 68.83% (n = 53)
exhibited somatic mutations, most of which were missense mutations,
with an average mutation burden of 0.52/MB ([115]Figure 3C,D). The
three most frequently mutated genes were TP53 (18%), CTNNB1 (17%), and
MUC16 (14%). Notably, high-risk patients showed a higher mutation
burden compared to low-risk patients ([116]Figure 3D).
3.5. Pathway Enrichment Analysis of DEGs Expression Between High- and
Low-Risk Cohorts
To gain deeper insight into the mechanisms underlying outcome
differences between these two risk categories, we applied the DESeq2
package, identifying 323 upregulated and 504 downregulated genes in
high-risk patients compared to low-risk patients ([117]Figure 4A).
Notably, GSEA revealed the suppression of several pathways related to
immune system function, including antigen processing and presentation,
cytokine–cytokine receptor interaction and the intestinal immune
network for IgA production when considering all DEGs ([118]Figure 4B).
Similarly, genes downregulated in the high-risk group were found to be
enriched in immune-related pathways, particularly T cell receptor and
costimulatory signaling, as well as cancer immunotherapy through PD-1
blockade, both of which are closely associated with tumor immunotherapy
([119]Figure 4C). Conversely, while upregulated genes in the high-risk
group showed enrichment in certain pathways, these pathways did not
reach statistical significance after adjustment for multiple
comparisons ([120]Figure 4D). GO analyses of all DEGs identified
immune-related pathways, with GO terms such as “antigen
receptor-mediated signaling pathway”, “lymphocyte mediated immunity”
and “antigen binding” ([121]Figure 4E,F) as well as KEGG analyses as
“cytokine-cytokine receptor interaction” when analyzing all DEGs
([122]Supplementary Figure S6).
Figure 4.
[123]Figure 4
[124]Open in a new tab
DEGs and related enrichment pathways between high-risk and low-risk
groups. (A) The volcano plot illustrates the differentially expressed
genes between the high-risk and low-risk groups. (B) The dot plot of
Gene Set Enrichment Analysis (GSEA) shows the enriched pathways in the
differentially expressed genes. (C,D) The enrichment analysis of the
pathways associated with downregulated and upregulated genes in the
high-risk group. (E,F) The GO enrichment analysis of the DEGs
highlights biological processes, molecular functions, and cellular
components.
3.6. Tumor-Infiltrating Immune Cell Profile
ESTIMATE analysis revealed a lower immune score in the high-risk group,
suggesting lower immune cell infiltration in the high-risk group
([125]Figure 5A). Consistently, ssGSEA confirmed significantly lower
scores for activated CD8 T cells, effector memory CD8 T cells, and γδ T
cells in the high-risk group. In contrast, type 2 T helper cells and
activated CD4 T cells were more abundant in the high-risk group
([126]Figure 5B). In line with these findings, a detailed correlation
analysis between the risk score and immune cells is presented in
[127]Figure 5C, showing that most immune cell populations exhibited
negative correlations with the risk score. The 28 tumor-infiltrating
immune cell types displayed weak to moderate correlations ([128]Figure
5D). Moreover, immune cell proportion composition analysis using
CIBERSORT showed CD8 T cells, activated NK cells, and resting mast
cells were all negatively correlated with genes in our model (HJURP,
CDK1, FOXM1, CHEK1), while macrophages M0, activated dendritic cells,
resting NK cells and activated dentritic cells were positively
correlated ([129]Figure 5E). The correlations between these four genes
and immune cell scores, as analyzed by ssGSEA, are provided in
[130]Supplementary Figure S7.
Figure 5.
[131]Figure 5
[132]Open in a new tab
Comparison of tumor microenvironment features between the two risk
cohorts. (A) The violin plot illustrates the stromal score, immune
score, and ESTIMATE score between the high- and low-risk groups. (B)
The box plot shows the scores of 28 immune cell types in the two risk
groups. (C) The correlation analysis between the risk score and the 28
immune cell types (statistically significant results in red). (D) The
heatmap visualization of the correlation among immune cells in ACC. (E)
The heatmap demonstrates the relationship between the proportions of 22
immune cells in the TME and the genes from our model. * p < 0.05, ** p
< 0.01, *** p < 0.001, **** p< 0.0001, ns: no statistical significance.
3.7. Evaluation of Immune Function and Prediction of Immunotherapy Response
Tumor immune evasion operates through camouflage (avoiding recognition
as malignant), coercion (preventing immune cell activation), and
cryoprotection (shielding malignant cells from immune cytotoxicity)
[[133]22]. These mechanisms are largely governed by proteins expressed
on the surfaces of both immune and tumor cells, such as PD-L1 and Human
Leukocyte Antigen (HLA). To further explore the relationship between
the risk score and immune function, we analyzed the correlation of mRNA
expression of genes involved in antigen processing and presentation,
immune checkpoints, and phagocytosis signaling with both the risk score
and genes in our model. As shown in [134]Figure 6A–F, most HLA genes
(HLA-DOA, HLA-DPA1, HLA-DPB1, HLA-DQA1, HLA-DRA, HLA-DPB1, et al.)
exhibited a negative correlation with the model genes. In contrast,
immune checkpoints such as CD276 and NRP1 showed a strong positive
relationship, while TNFSF14 and LAIR1 demonstrated a notable negative
correlation. Among genes involved in phagocytosis signaling, ITGB3 and
TYRO3 were positively correlated, while GAS6 and ITGAM were negatively
associated. Detailed data are presented in [135]Supplementary Table
S10.
Figure 6.
[136]Figure 6
[137]Open in a new tab
Analysis of immune function-related genes and model-based immunotherapy
response prediction. (A) The box plot illustrates the expression of
genes involved in antigen processing and presentation between the two
risk groups. (B) Gene expression of immune checkpoint genes in the two
risk cohorts. (C) Expression of phagocytosis signaling-related genes
across high- and low-risk patients. (D–F) The correlation of HJURP,
CDK1, FOXM1, and CHEK1 with genes involved in antigen processing and
presentation, immune checkpoint regulation, and phagocytosis signaling.
(G) The TIDE scores and immune exclusion scores between the two groups.
(H) The immune characteristics of the tumor microenvironment (TME) in
the two groups. (I) The immunotherapy response prediction for the two
groups. * p < 0.05, ** p < 0.01, *** p < 0.001, **** p < 0.0001, ns: no
statistical significance.
A higher TIDE score, accompanied by an elevated exclusion score,
indicates reduced sensitivity to immunotherapy in the high-risk group
([138]Figure 6G). Consistently, characteristics associated with
Myeloid-Derived Suppressor Cells (MDSCs) and Tumor-Associated
Macrophages M2 (TAM M2), both of which are known to play a crucial role
in tumor immune evasion [[139]23,[140]24], were significantly elevated
in the high-risk group based on our analysis ([141]Figure 6H).
Regarding the prediction of immunotherapy efficacy, 22.5% of high-risk
patients respond to immunotherapy, compared to 46.2% in the low-risk
group (p = 0.02) ([142]Figure 6I). Data analyzed from TIDE was shown in
[143]Supplementary Figure S8 and Table S11.
3.8. Predictive Analysis for Drug Therapy
OncoPredict predicts drug sensitivity based on gene expression levels.
Out of 198 chemotherapeutic agents analyzed, 75 showed a significant
difference in the predicted IC50 values between the two groups (p <
0.05). We highlight the top 20 drugs with the lowest p-values (p <
0.001) in [144]Figure 7. The higher-risk group exhibited lower
predicted IC50 values for BI-2536, OSI-027, Sepantronium bromide, MIM1,
Tozasertib, Daporinad, ML323, Acetalax, Linsitinib, and ULK_4989 and
others, compared to the lower-risk group, suggesting greater drug
sensitivity compared to the lower-risk group. Detailed drug sensitivity
data are provided in [145]Supplementary Table S12, and additional
results are shown in [146]Supplementary Figure S9.
Figure 7.
[147]Figure 7
[148]Open in a new tab
Prediction of drug sensitivity by OncoPredict in high-risk and low-risk
groups. ** p < 0.01, *** p < 0.001, **** p < 0.0001. •: outliers.
4. Discussion
As a malignant tumor originating from the adrenal cortex, the molecular
landscape of ACC has been increasingly defined through multi-omics
studies, facilitated by advances in detection technologies.
Unfortunately, effective treatments for this rare and aggressive
disease remain to be developed. Thus, there is an urgent need to
discover reliable and complementary prognostic indicators and risk
stratification approaches. The first report of cellular senescence
dates back to the 1960s when scientists observed cell cycle arrest in
human diploid fibroblasts [[149]25]. Since then, the mechanisms
underlying senescence in various biological processes have gradually
been uncovered. Studies on senescence in adrenal diseases have shown
that telomerase activity can serve as an effective tool for
distinguishing malignant from benign adrenocortical tumors.
Specifically, telomerase activity is higher in ACC than in
adrenocortical adenomas (ACA) [[150]26], making it a valuable marker
for tumor classification. However, the exact role of senescence in ACC
remains to be fully elucidated. In many other cancers, senescence has
been reported to play a dual role [[151]27]. On one hand, senescence
contributes to tumorigenesis, as evidenced by its role in the invasion
and progression of melanoma and papillary thyroid carcinoma (PTC)
[[152]28,[153]29]. On the other hand, it acts as a tumor-suppressive
mechanism by inducing cell-cycle arrest, thereby inhibiting tumor
growth [[154]30,[155]31]. In our study, we utilized public databases
(TCGA-ACC and GEO) to develop a model based on a four-gene signature
associated with senescence (HJURP, CDK1, FOXM1, and CHEK1). Patients
with a higher risk score calculated by our model exhibited
significantly worse overall survival and more advanced tumor stages.
Additionally, our analysis suggested a potential trend toward a reduced
immunotherapy response in the high-risk group.
Each of the four genes (HJURP, CDK1, FOXM1, and CHEK1) identified in
the model has been implicated in cell cycle regulation and tumor
development. The Holliday junction recognition protein (HJURP) is a
critical mitogenic protein that plays a vital role in nucleosome
assembly during mitosis [[156]32]. Chromosomal instability, a hallmark
of cancer, results from persistent errors in chromosome segregation
during cell division. HJURP is involved in the recruitment and assembly
of the centromere and kinetochore, which are essential for accurate
chromosome segregation, thereby playing a key role in maintaining
chromosomal stability in tumor cells [[157]33]. The upregulation of
HJURP has been observed in various cancers, in which it regulates tumor
progression and chemoresistance through multiple mechanisms, including
the YAP1/NDRG1 transcriptional axis in triple-negative breast cancer
[[158]34], modulation of the MDM2/p53 pathway in pancreatic cancer
[[159]35], and promotion of CDKN1A degradation via the GSK3β/JNK
pathway in prostate cancer cells [[160]36]. Moreover, HJURP has been
linked to poor prognosis in various cancers, including non-small cell
lung cancer, hepatocellular carcinoma, and renal cell carcinoma
[[161]32,[162]37,[163]38]. Another gene in our model, cyclin-dependent
kinase 1 (CDK1), is a key member of the cyclin-dependent kinase family
and plays a pivotal role in regulating cell cycle progression.
Specifically, its interaction with cyclin B, which peaks during the
G2/M phase, ensures the orderly transition of cells into mitosis
[[164]39]. The overexpression of CDK1 has been observed in ACC cell
lines, and where it has been shown to lock ACC cells at the G2/M
checkpoint through interaction with UBE2C and AURKA/B. Additionally,
CDK1 promotes epithelial-to-mesenchymal transition via Slug and Twist
[[165]40]. Recently, several CDK1 inhibitors have been developed as
therapeutic strategies for cancer treatment. For instance, BEY1107, an
orally active CDK1 inhibitor, has been evaluated in patients with
locally advanced or metastatic pancreatic cancer ([166]NCT03579836)
[[167]39]. Furthermore, forkhead Box M1 (FOXM1) is a transcription
factor (TF) belonging to the forkhead box protein family (FOX). It
plays a critical role in upregulating the expression of various genes
associated with DNA replication, G1/S and G2/M transition. These
include cyclin A2, Cdc25A phosphatase, and ATF2 [[168]41,[169]42].
Finally, CHEK1 serves as a crucial regulator of the DNA damage response
(DDR), playing a vital role in detecting DNA replication stress caused
by oncogene activation or dysfunction of the G1 checkpoint [[170]43].
The CHK1/2 inhibitor AZD7762 has been shown to enhance anticancer
efficacy synergistically when combined with other drugs [[171]44].
Senescence occurs when cells respond to various stressors, such as DNA
damage, triggering a cascade of signaling pathways which lead to
sustained cell cycle arrest. This process results in a decline in cell
cycle activity and the downregulation of mitotic proteins [[172]45].
Therefore, we suggest the upregulation of genes in our model work
together in regulating cell cycle and tumor progression, with HJURP
ensuring chromosome stability, CDK1 driving mitotic entry, and FOXM1
promoting proliferation and CHEK1 supporting cell survival under DNA
damage conditions.
It is well established that “cold” tumors, such as ACC, are
characterized by low immune cell infiltration, which is associated with
poorer outcomes and a diminished response to immunotherapy
[[173]46,[174]47]. Clinical trials investigating immunotherapy in ACC
have consistently shown limited efficacy, with low overall response
rates and limited progression-free survival [[175]48,[176]49,[177]50].
This resistance may be attributed to factors such as cortisol secretion
[[178]51] or specific mutation types. Notably, mutations in the
Wnt/β-catenin pathway, one of the most common genetic alterations in
ACC [[179]52] (17% in our study), have been shown to impair anticancer
immune responses [[180]53,[181]54,[182]55]. Individuals in the
high-risk group carry a greater burden of mutation types, and the
enrichment of downregulated genes in this group suggests a suppressed
immune function, particularly in pathways related to T cell activity,
immunotherapy response, and cytokine signaling. These findings suggest
that the poorer OS and more advanced tumor stage in the high-risk group
may be driven by an abnormal immune cell distribution and functional
impairments. This is further supported by reduced tumor purity, as
calculated by ESTIMATE, and diminished immune cell infiltration, as
revealed by ssGSEA and CIBERSORT, in the high-risk group. Among the
immune cells, a notable reduction in activated CD8 T cells-the key
effectors in tumor cell elimination partly explains the immune evasion
observed in these patients. Additionally, the high-risk cohort
exhibited decreased levels of activated dendritic cells, suggesting a
deficiency in immune surveillance. Senescent tumor cells secrete
chemokines that attract immune cells, particularly macrophages, to
facilitate tumor cell elimination [[183]56]. Consistent with our
findings, high-risk patients exhibited fewer immune cell signatures in
their transcriptomes, and the risk score was negatively correlated with
macrophage signatures. This reduced immune infiltration may drive tumor
cells in the high-risk group to favor cell cycle re-entry over
senescence, potentially contributing to their increased proliferative
capacity.
To further assess the immune response in the tumor microenvironment in
relation to our model, we investigated genes involved in the
recognition and elimination of tumor cells by immune cells. Human
Leukocyte Antigen (HLA), which encodes the Major Histocompatibility
Complex (MHC) proteins responsible for immune recognition, is inversely
correlated with the genes from our model. Specifically, MHC class I
molecules, such as HLA-A, interact with CD8+ T cells to facilitate
tumor cell destruction, while MHC class II molecules—HLA-DPA1,
HLA-DPB1, HLA-DQA1, HLA-DQB1, and HLA-DRB1—are predominantly expressed
on antigen-presenting cells, such as dendritic cells and macrophages,
where they are recognized by CD4+ T cells [[184]57]. Regarding immune
checkpoint genes, CD276 and NRP1 are positively correlated with the
genes in our model, while TNFSF14 shows a negative correlation with
these genes in the tumor immune microenvironment of high-risk patients.
CD276 is reported to be expressed on the surface of cancer stem cells
and tumor-associated macrophages, playing a critical role in
suppressing the immune response against tumors [[185]58,[186]59]. NRP1
(Neuropilin-1) functions as an “immune memory checkpoint” limiting the
development of long-lived tumor-specific T cells, which are essential
for maintaining durable antitumor immunity [[187]60]. Additionally,
NRP1 promotes tumor cell survival and progression by interacting with
the hypoxia response and microRNAs [[188]61]. Alternatively, TNFSF14
(Tumor Necrosis Factor Superfamily Member 14), also known as LIGHT,
enhances anti-tumor immune responses by promoting vascular
normalization and the generation of tertiary lymphoid structures
[[189]62]. In terms of phagocytosis signaling, TYRO3 and ITGB3 are
significantly associated with our model genes. TYRO3, a member of the
TYRO3, AXL, and MerTK (TAM) receptor tyrosine kinase (RTK) family, is
upregulated in various cancers [[190]63]. It inhibits ferroptosis and
suppresses innate immunity in TME, making it a potential predictive
biomarker for identifying patients who may benefit from anti-PD-1/PD-L1
therapy [[191]64]. ITGB3, an integrin that binds to fibronectin,
vitronectin, and collagen, facilitates interactions between the cell
cytoskeleton and the extracellular matrix [[192]65]. A recent study has
highlighted its pivotal role in mediating intracellular communication
through extracellular vesicles, which are essential for cancer
metastasis [[193]66]. In summary, ACC tumors in high-risk patients tend
to exhibit immune dysfunction through various mechanisms, potentially
leading to greater resistance to immunotherapy compared to those with
lower-risk groups.
As discussed above, high-risk patients with higher TIDE scores may
exhibit a poorer response to immunotherapy, accompanied by an increased
presence of MDSCs and TAM M2, both of which are immunosuppressive
myeloid cells. MDSCs are primarily generated in the bone marrow, and
upon migrating to the tumor site, they can differentiate into
immunosuppressive macrophages through the activation of the
transcription factor NF-κB by S100A8/A9 proteins [[194]67].
Furthermore, the immunosuppressive activity of MDSCs can be induced by
IFNγ mediated via the activation of the transcription factor STAT1
[[195]68]. TAM M2 can activate Smad2/3 and Smad1/5/8, thereby promoting
the epithelial–mesenchymal transition (EMT) and tumor metastasis
[[196]69]. Moreover, cytokines secreted by TAM M2, including TNF-α,
ICAM-1, and IL-6, further contribute to tumor growth and invasion
[[197]70]. Recent studies have shown that targeting either MDSCs or M2
TAMs can enhance the efficacy of immunotherapy [[198]71,[199]72]. Based
on our model’s high-risk score, machine learning analysis indicates
that the upregulation of MDSCs and TAM M2, coupled with the
downregulation of CD8+ T cells in the tumor microenvironment, may
contribute to the diminished immunotherapy response observed in
high-risk patients.
OncoPredict integrates data from the Cancer Cell Line Encyclopedia
(CCLE) and the Genomics of Drug Sensitivity in Cancer (GDSC) to
identify potent chemotherapy options. As indicated by the results, 10
out of the 20 drugs with a p-value < 0.001 demonstrated efficacy in the
higher-risk group, exhibiting lower IC50 values. These drugs target key
molecular pathways related to tumor cell proliferation, survival, and
metabolism. Specifically, the cell cycle regulators are as follows:
BI-2536 (targeting polo-like kinase 1) [[200]73] and OSI-027 (an mTOR
kinase inhibitor) [[201]74]; the survival signaling pathways are
targeted by Sepantronium bromide (targeting surviving) [[202]75] and
Tozasertib (targeting Aurora kinase B) [[203]76]; Daporinad (inhibit
NAD+ synthesis pathway) [[204]77] affects metabolic pathways; and
Linstinib (an oral kinase inhibitor for IGF-1R and the insulin
receptor) [[205]78] targets insulin/IGF signaling. Notably, all of
these drugs have been registered for clinical trials. In contrast, MIM1
(Mcl-1 protein inhibitor) [[206]79], ML323 (ubiquitin-specific protease
1 inhibitor) [[207]80], Acetalax (a phenylpyridine compound) [[208]81],
and ULK1_4989 (targeting autophagy) [[209]82] remain in preclinical
development. We hypothesize that the model genes in the high-risk group
primarily drive robust mitotic activity in tumor cells, a process that
is strongly dependent on cell cycle and metabolic pathways. This may
explain the increased sensitivity of the high-risk group to the
predicted drugs. Among these, BI-2536 has been shown to exert cytotoxic
effects on adrenocortical carcinoma cell lines (H295R and SW13) in both
in vitro and in vivo studies, including cell line-derived xenograft
(CDX) models in nude mice [[210]83]. Additionally, it has been reported
to induce senescence in colorectal cancer cell lines [[211]84]
suggesting that high-risk patients may be more susceptible to
senescence induction.
There are several limitations in our study. First, the model was
developed and validated using publicly available datasets, which are
constrained by a relatively small sample size and a retrospective
design, with some patients lacking comprehensive clinical data. Second,
although TCGA data were used for model development and two independent
GEO datasets for external validation, potential batch effects may arise
due to variations in data generation platforms, processing methods, or
technologies. Third, all our data were based on machine learning
without experimental validation. To address these limitations, further
validation in independent, larger cohorts with standardized protocols
is essential to assess the model’s robustness and generalizability.
5. Conclusions
In summary, we have developed a four-gene signature related to the cell
cycle that provides insights into the prognosis of adrenocortical
carcinoma (ACC). Using this model, we also investigated clinical
characteristics, transcriptomic features, immune profiles, and drug
sensitivity predictions. Our analysis suggests that low-risk
individuals, as stratified by our model, may demonstrate enhanced
sensitivity to immunotherapy, while high-risk individuals may benefit
more from alternative therapies, such as chemotherapy. This study
represents the first attempt to categorize ACC based on the expression
of senescence-related genes using bioinformatics tools, potentially
opening new therapeutic avenues for ACC.
Acknowledgments