Abstract
Cholangiocarcinoma (CCA) is a highly heterogeneous and aggressive
malignancy of the bile ducts with a poor prognosis and high mortality
rate. Effective targeted therapy and accurate prognostic biomarkers are
still lacking. Ferroptosis is a form of regulated cell death implicated
in cancer progression and has emerged as a potential therapeutic target
in various cancers. However, a comprehensive analysis of
ferroptosis-related genes (FRGs) for predicting CCA prognosis and
therapeutic targets and determining the role of ferroptosis in CCA
remain to be performed. Here, we developed a prognostic FRG signature
using a least absolute shrinkage and selection operator Cox regression
analysis in a training cohort. We then validated it using four
independent public datasets. The six-FRG signature was developed to
predict CCA patient survival, stratifying them into low-risk and
high-risk groups based on survival time. Significantly, the high-risk
CCA patients had shorter overall survival. A receiver operating
characteristic curve analysis further confirmed the prognostic FRG
signature’s strong predictive ability, indicating that it was an
independent prognostic indicator for CCA patients. Furthermore, the
high-risk group was associated with fluke infection and high clinical
stages. Cancer-associated fibroblast (CAF) score and CAF markers were
significantly higher in the high-risk group than the low-risk group.
Moreover, our FRG signature could predict immune checkpoint markers for
immunotherapy and drug sensitivity. The mRNA expression levels of the
six-FRG signature was validated in 10 CCA cell lines and dividing them
into low-risk and high-risk groups using the FRG signature. We further
showed that high-risk CCA cell lines were more resistant to ferroptosis
inducers, including erastin and RSL3, than the low-risk CCA cell lines.
Our study constructed a novel FRG signature model to predict CCA
prognoses which might provide prognostic biomarkers and potential
therapeutic targets for CCA patients. Ferroptosis sensitivity in
high-risk and low-risk CCA cell lines suggests that ferroptosis
resistance is associated with high-risk group CCA. Therefore,
ferroptosis could be a promising therapeutic target for precision
therapy in CCA patients.
Keywords: ferroptosis, cholangiocarcinoma, gene signature, prognosis,
risk score
1. Introduction
Cholangiocarcinoma (CCA/CHOL) is a highly heterogeneous malignancy
originating from epithelial bile ducts at any level of the bile duct
tree. Its incidence rate has significantly increased worldwide, with
higher prevalence in Asian countries over the past few years ([27]1).
Due to a lack of effective early diagnosis, most CCA patients are
usually diagnosed at advanced stages where surgical resection cannot be
performed ([28]2, [29]3). While, therapeutic options for CCA patients
are increasing, such as chemotherapy, targeted therapy, and
immunotherapy, their overall prognosis remains unsatisfactory
([30]4–[31]6). Therefore, identifying novel predictive models, accurate
prognostic biomarkers, and novel therapeutic targets are urgently
required to improve overall survival of CCA patients.
Ferroptosis is a novel regulated form of cell death that relies on iron
overload, accumulation of reactive oxygen species (ROS) and
polyunsaturated fatty acids (PUFA), and phospholipid peroxidation
([32]7–[33]9). System
[MATH:
Xc−<
/msubsup> :MATH]
and glutathione peroxidase 4 (GPX4) are key regulators in the
glutathione pathway that control the ferroptosis mechanism. Erastin and
RAS-selective lethal (RSL3) can induce ferroptosis by inhibiting system
[MATH:
Xc−<
/msubsup> :MATH]
and GPX4 activity, respectively ([34]9–[35]11). Recent studies have
shown that ferroptosis represents a novel target for efficient
therapeutic strategies to overcome treatment resistance in several
cancers ([36]12). In addition, ferroptosis-related genes (FRGs) and
ferroptosis signaling dysregulation might be associated with cancer
patient prognosis. However, the role of ferroptosis in CCA progression
and prognosis remain unknown. In addition, very few studies have
explored the therapeutic applications of ferroptosis for CCA patients.
Therefore, discovering key prognostic FRGs predicting CCA prognosis and
clinical outcomes is urgently needed since they might act as novel
biomarkers and therapeutic targets for CCA patients.
This study obtained RNA expression profiles and clinical data from two
cohorts in the Gene Expression Omnibus (GEO) database and developed a
six-FRG signature. We validated our six-FRG signature for CCA prognosis
prediction in four independent cohorts in GEO, The Cancer Genome Atlas
(TCGA), the European Bioinformatics Institute (EMBL-EBI) database, and
the National Omics Data Encyclopedia (NODE) database. Our six-FRG
signature was used to stratify CCA patients into two groups, confirming
their prognosis prediction. In addition, these two patient groups
differed in their functional and biological processes, immune cell
infiltration, cancer-associated fibroblast (CAF) abundance, and drug
sensitivity. Moreover, we confirmed the mRNA expression levels of these
six-FRG signature in a panel of CCA cell lines, dividing them into two
groups based on their mRNA expression levels for these six-FRG
signature. We further investigated the sensitivity of these CCA cell
lines to ferroptosis inducers, including erastin and RSL3.
2. Materials and methods
2.1. Data collection and processing
A flow chart describing the data collection and analysis process is
provided in [37]Figure 1 . Five public RNA expression and clinical
information were downloaded from four platforms. The TCGA-CHOL dataset
was downloaded from the University of California at Santa Cruz (UCSC)
Xena platform ([38]https://xena.ucsc.edu/). The E-MTAB-6389 dataset was
downloaded from the European Bioinformatics Institute (EMBL-EBI)
database ([39]https://www.ebi.ac.uk/). The OEP001105 dataset reported
by a previous study ([40]13) was downloaded from The National Omics
Data Encyclopedia (NODE) database ([41]https://www.biosino.org/node/).
The [42]GSE76297, [43]GSE89749, and [44]GSE107943 datasets were
downloaded from the Gene Expression Omnibus (GEO) database
([45]https://www.ncbi.nlm.nih.gov/geo/). The [46]GSE89749 dataset’s
clinical information was obtained from previous study ([47]14). In
total, 267 FRGs were identified from published studies ([48]15, [49]16)
and the FerrDb database ([50]http://www.zhounan.org/ferrdb/) ([51]17).
CCA patient characteristics in all datasets are summarized in
[52]Supplementary Table S1 .
Figure 1.
[53]Figure 1
[54]Open in a new tab
Flow chart of the study.
2.2. Identification of ferroptosis-related differentially expressed and
prognostic genes
Differentially expressed genes (DEGs) were identified in the
[55]GSE76297 cohort, which includes 91 pairs of tumor and non-tumor
tissues using a paired-sample t-test with a |log[2](fold change)| > 1
and p-value < 0.0001. A univariate Cox regression analysis was
performed to screen out prognostic genes associated with overall
patient survival in the [56]GSE89749 cohort. This cohort includes 111
patients of which only those with an overall survival ≥ 30 days were
included to ensure this study’s reliability. Genes with a p-value
< 0.05 were considered prognostic genes. Finally, overlapping DEGs and
prognostic genes were identified as candidate genes using a Venn
diagram. A protein-protein interaction (PPI) network analysis was
performed on candidate genes by the STRING database
([57]https://www.string-db.org/) ([58]18).
2.3. FRG signature construction
The [59]GSE89749 cohort was used as a training cohort to construct an
FRG signature. A least absolute shrinkage and selection operator
(LASSO) Cox regression analysis was used to develop the FRG signature
from candidate genes using the R statistical software’s “glmnet”
package. Then, the following formula was used to calculate a
ferroptosis-related risk score for each patient: risk score =
(Exp.Gene[1] × Coef.Gene[1]) + (Exp.Gene[2] × Coef.Gene[2]) + … +
(Exp.Gene[n] × Coef.Gene[n]), where Exp.Gene is the expression of FRG
signature genes and Coef.Gene is the regression coefficient obtained
from the LASSO Cox regression analysis. The 111 patients in the
[60]GSE89749 cohort were stratified into low-risk and high-risk groups
based on the median risk score. Kaplan-Meier and log-rank test were
used to compare patient survival between risk groups using R’s
“survminer” package. A time-dependent receiver operating characteristic
(ROC) curve was used to predict the FRG signature’s specificity and
sensitivity for predicting patient survival at 1, 3, and 5 years using
R’s “survivalROC” package. The FRG signature’s prognostic potential was
validated using the OEP001105, E-MTAB-6389, [61]GSE107943, and
TCGA-CHOL datasets.
2.4. Independent prognostic value analysis
A univariate Cox regression analysis was used to evaluate the
prognostic value of the FRG signature and other clinical
characteristics, including age, liver fluke infection, sex, and stage.
A multivariate Cox regression analysis was performed to evaluate
whether the FRG signature was an independent prognostic factor. Each
variable’s hazard ratio (HR) and 95% confidence interval (CI) were
calculated, with p-value < 0.05 considered statistically significant.
Moreover, Fisher’s exact test was used to assess differences in
clinical characteristics between low-risk and high-risk groups.
2.5. Functional gene set enrichment analysis
A GSEA was performed in the training cohort using GSEA software to
identify functions and pathways enriched between low-risk and high-risk
groups based on Gene Ontology (GO; c5.go.v7.5.1.symbols.gmt) and Kyoto
Encyclopedia of Genes and Genomes (KEGG;
c2.cp.kegg.v7.5.1.symbols.gmt). Gene sets with a | normalized
enrichment score (NES)| > 1, p-value < 0.05, and false discovery rate
(FDR) < 0.25 were considered statistically significant.
2.6. Immune cell infiltration and tumor microenvironment analysis
The CIBERSORTx algorithm ([62]https://cibersortx.stanford.edu/)
([63]19) was used to analyze the immune cell fractions of 22 immune
cell types in the training cohort. Moreover, the MCP-counter ([64]20)
and EPIC ([65]http://epic.gfellerlab.org/) ([66]21) algorithms were
used to estimate CAF abundance.
2.7. Drugs sensitivity and immunotherapy prediction
Differences in drugs sensitivity between the two patient groups were
estimated by comparing half-maximal inhibitory concentration (IC[50])
using R’s “pRRophetic” package and the Genomics of Drug Sensitivity in
Cancer (GDSC) database ([67]https://www.cancerrxgene.org/) ([68]22).
Furthermore, the differential expression of common immune checkpoints
in low-risk and high-risk groups was examined to predict potential
immunotherapy targets.
2.8. Cell culture
The human immortalized non-tumor cholangiocyte cell line (MMNK-1) and
CCA cell lines (CCLP-1, HuCCT-1, KKU-055, KKU-100, KKU-213, KKU-214,
RBE, and TFK-1) were obtained from the Japanese Collection of Research
Bioresources (JCRB) Cell Bank (Osaka, Japan). The HuCCA-1 and RMCCA-1
CCA cell lines were developed from Thai patients with CCA ([69]23,
[70]24). All cell lines were grown in Dulbecco’s modification of
Eagle’s medium (DMEM; HyClone Laboratories, Logan, UT, USA)
supplemented with 10% fetal bovine serum (Sigma-Aldrich, St Louis, MO,
USA) and 1% Penicillin–Streptomycin (HyClone Laboratories) and were
cultured in a humidified incubator at 37°C with 5% carbon dioxide. All
cell lines were tested to be negative for mycoplasma contamination.
2.9. Reverse transcription-quantitative PCR
Total RNA was extracted from the cells using GENEzol Reagent (Geneaid
Biotech, Taiwan). Then, 1 μg of RNA was reverse-transcribed using a
Maxime RT PreMix Kit (iNtRON Biotechnology, Seongnam-si, Gyeonggi-do,
Republic of Korea). RT-qPCR was performed using iTaq universal SYBR
Green Supermix (Bio-Rad, Hercules, CA, USA) following the
manufacturer’s instructions. All primers used in this study are listed
in [71]Supplementary Table S2 . Relative expression in CCA cell lines
was normalized to MMNK-1 cell line using the 2^-ΔΔCt method with
β-actin (ACTB) as an internal control. Moreover, average 2^-ΔCt values
were used as each gene’s values to classify CCA cell lines. Then, the
expression values were transformed to z-score in each gene of all CCA
cell lines. These z-score expression values were used to calculate risk
scores in CCA cell lines which were then stratified into low-risk and
high-risk FRG groups.
2.10. Treatment and cell viability assay
Erastin and RSL3 were obtained from ApexBio Technology (Boston, MA,
USA). Two CCA cell lines groups based on the FRG risk groups, including
CCLP-1, KKU-214, RBE, and RMCCA-1 were seeded in 96-well plates and
incubated in a humidified incubator at 37°C with 5% carbon dioxide for
24 h. The cells were treated for 48 h using a two-fold serial dilution
method with erastin concentrations of 0.3125, 0.625, 1.25, 2.5, 5, 10,
20, and 40 μM or RSL3 concentrations of 7.8125, 15.625, 31.25, 62.5,
125, 250, 500, and 1000 nM. Cell viability was determined using
3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyl-2H-tetrazolium bromide (MTT)
assay after 48 h of treatment. Briefly, 10 μl of MTT reagent was added
to each well and incubated in a humidified incubator for 2 h. Next, the
supernatant was removed, and 100 μl of dimethyl sulfoxide (DMSO) was
added to each well. Then, cell viability was determined at 570 nm using
a microplate reader and the percentage of viable cells was calculated
and normalized to the DMSO vehicle control. The IC[50] of erastin and
RSL3 was calculated and compared between two CCA cell line groups.
Three-independent experiments were performed with triplicate samples.
2.11. Statistical analysis
All data were analyzed using the R statistical software (version 4.1.0)
or SPSS (version 22.0, IMM Corp; Armonk, NY, USA). Wilcoxon or
Student’s t-tests were used to assess differences between groups. The
log-rank test was used to assess differences in survival between
low-risk and high-risk groups, and a Kaplan-Meier curve was used to
visualize patient survival. Pearson’s correlation coefficient (r) was
used in all correlation analyses. All results with p-value < 0.05 were
considered statistically significant (*p < 0.05, **p< 0.01, ***p <
0.001, ****p < 0.0001).
3. Results
3.1. Candidate gene identification
This study included 267 FRGs which are listed in [72]Supplementary
Table S3 . Forty-eight ferroptosis-related DEGs were identified between
paired tumor and non-tumor tissues in the [73]GSE76297. Heatmap and
volcano plots visualized their expression and distribution among
samples, with 21 upregulated and 27 downregulated ([74] Figures 2A, B
). One hundred eleven patients with overall survival ≥ 30 days from the
[75]GSE89749 dataset were used a univariate Cox regression analysis to
identify prognostic genes. The 47 prognostic genes associated with
survival are listed in [76]Table 1 . A Venn diagram identified 17
intersecting genes (candidate genes) among the 48 ferroptosis-related
DEGs and 47 prognostic genes ([77] Figure 2C ). The univariate Cox
regression results for these 17 candidate genes were visualized in a
forest plot ([78] Figure 2D ). Four were protective factors (ACO1,
PEBP1, GOT1, and CXCL12), and 13 genes were risk factors (FANCD2, MT1G,
PTGS2, SQLE, NQO1, SLC1A5, TF, MUC1, HELLS, SLC7A5, HAMP, SLC2A1, and
RRM2) in CCA patients. The PPI network indicated associations between
17 candidate genes ([79] Figure 2E ). Correlations among these genes
are shown by a correlation network ([80] Figure 2F ).
Figure 2.
[81]Figure 2
[82]Open in a new tab
Identification of ferroptosis-related candidate genes. (A) Heatmap and
(B) volcano plot showing the expression levels and distribution of
differentially expressed genes (DEGs) in cholangiocarcinoma (CCA)
tissues and normal tissues in the [83]GSE76297 cohort. (C) A Venn
diagram showing the intersection between DEGs and prognostic genes. (D)
A forest plot showing the hazard ratios of candidate genes. (E) A
protein-protein interaction (PPI) network of the candidate genes in the
STRING database. (F) A correlation network of the candidate genes.
Table 1.
Univariate Cox regression analysis of ferroptosis-related genes in
[84]GSE89749 cohort.
Gene Coefficient p-value
CS -1.2200 0.0013
FANCD2 1.1184 0.0069
GSS 0.6494 0.0307
MT1G 0.1409 0.0127
PTGS2 0.3962 <0.0001
SAT1 0.3975 0.0300
ACO1 -0.5487 0.0334
ACACA 0.6612 0.0317
PEBP1 -0.4156 0.0374
SQLE 0.5543 0.0028
NQO1 0.2857 0.0336
SLC1A5 0.3307 0.0249
GOT1 -0.4960 0.0003
ACSF2 0.4344 0.0014
NOX4 0.3524 0.0198
FLT3 -1.2662 0.0445
HRAS 0.4983 0.0423
TF 0.1166 0.0193
BECN1 0.6708 0.0283
WIPI2 -1.2005 0.0192
MAPK3 0.5606 0.0076
YY1AP1 -1.2359 0.0225
HSF1 0.8390 0.0109
MUC1 0.2436 0.0023
HELLS 0.5399 0.0428
SCD 0.2713 0.0441
STAT3 0.8531 0.0045
MTOR 0.5988 0.0361
TP63 0.5130 0.0187
ISCU -0.5828 0.0123
LAMP2 -0.4613 0.0052
UBC 0.9236 0.0245
OXSR1 0.7610 0.0016
DDIT4 0.3507 0.0059
DDIT3 -0.4767 0.0189
SLC7A5 0.2031 0.0301
TRIB3 -0.3108 0.0092
ZFP69B -1.3924 0.0270
GDF15 -0.2765 0.0038
CXCL2 -0.2605 0.0203
HAMP 0.1417 0.0370
MAP3K5 0.5430 0.0013
SLC2A1 0.5045 <0.0001
RRM2 0.5508 0.0333
CAPG 0.6259 0.0011
AURKA 0.3492 0.0125
PRDX1 0.5882 0.0137
[85]Open in a new tab
3.2. FRG signature construction
This study’s reliability was ensured by excluding three out of the 17
candidate genes (MTIG, TF, and HAMP) from gene signature construction.
While these three genes were downregulated in tumor samples, their
higher expression was associated with poorer prognosis. Therefore, the
expression levels of the 14 remaining candidate genes and overall
survival data from the [86]GSE89749 cohort were used as a training
cohort to construct the FRG signature via a LASSO Cox regression
analysis. A six-FRG signature (ACO1, GOT1, PTGS2, SLC2A1, FANCD2, and
SQLE) was identified based on the LASSO Cox regression with the minimum
optimal lambda value using tenfold cross-validation ([87] Figures 3A, B
). Each patient’s risk score was calculated using the LASSO Cox
regression analysis coefficient and patients were divided into low-risk
and high-risk groups based on their median risk score. The six-FRG
signature’s expression in the two risk groups was visualized as a
heatmap ([88] Figure 3C ). Kaplan-Meier curves were used to compare
survival in the two risk groups. They showed that survival was
significantly longer in the low-risk group than in the high-risk group
(p-value < 0.0001; [89]Figure 3D ). The six-FRG signature’s predictive
efficacy was evaluated with a time-dependent ROC curve. The area under
the ROC curves (AUCs) for the six-FRG signature at 1, 3 and 5 years of
survival were 0.7540, 0.8389, and 0.8103, respectively, suggesting that
it had high sensitivity and specificity ([90] Figure 3E ).
Figure 3.
[91]Figure 3
[92]Open in a new tab
Construction of the ferroptosis-related gene (FRG) signature in a
training cohort. (A, B) A least absolute shrinkage and selection
operator (LASSO) Cox regression analysis of the candidate genes. (C) A
heatmap showing the expression levels and distribution of the six-FRG
signature. (D) A Kaplan-Meier curve showing the overall survival of CCA
patients. (E) Area under the curve (AUC) of time-dependent receiver
operating characteristic (ROC) curves showing the six-FRG signature’s
predictive efficacy for survival time in CCA patients.
3.3. FRG signature validation in independent cohorts
The six-FRG signature’s reproducibility was assessed using four
independent cohorts. Patients with overall survival < 30 days were
excluded from the validation cohorts. The model obtained with the
training [93]GSE89749 cohort was used to calculate each patient’s risk
score in the validation cohorts. The OEP001105 (244 patients),
E-MTAB-6389 (75 patients), [94]GSE107943 (30 patients), and TCGA-CHOL
(33 patients) cohorts were divided into low-risk and high-risk groups
according to their median risk score, except for the TCGA-CHOL cohort,
which was divided using best cut-off. Kaplan-Meier curves for the
OEP001105, E-MTAB-6389, and [95]GSE107943 cohorts indicated that
survival was significantly shorter in the high-risk group than in the
low-risk group (p-value < 0.0001, p-value = 0.0003, and p-value =
0.0267, respectively; [96]Figures 4A, C, E ). While the Kaplan-Meier
curve for the TCGA-CHOL cohort was non-significant (p-value = 0.1638),
this might reflect population variation due to small size.
Nevertheless, the TCGA-CHOL cohort’s Kaplan-Meier curve indicated that
the high-risk group tended to have shorter survival times than the
low-risk group, which differed in their median survival ([97] Figure 4G
). ROC curves were used to assess the FRG signature’s accuracy in
predicting patient survival. The AUCs at 1, 3, and 4 years were 0.7522,
0.7078, and 0.6712 in the OEP001105 cohort, respectively ([98]
Figure 4B ). The AUCs at 1, 3, and 5 years were 0.7346, 0.7234, and
0.6433 in the E-MTAB-6389 cohort, respectively ([99] Figure 4D ). These
results indicated that the FRG signature showed the greatest accuracy
at one year, then gradually decreased with increasing years. However,
the AUCs at 1, 3, and 5 years were 0.7694, 0.8575, and 0.7552 in the
[100]GSE107943 cohort, respectively, showing the greatest accuracy at
three years ([101] Figure 4F ). Furthermore, the AUCs at 1, 3, and 5
years were 0.7105, 0.5829, and 0.6308, in the TCGA-CHOL cohort,
respectively ([102] Figure 4H ). Altogether, these results suggested
that our FRG signature could accurately predict CCA prognosis in
patients.
Figure 4.
[103]Figure 4
[104]Open in a new tab
Validation of the six-FRG signature in four independent cohorts. (A, C,
E, G) Kaplan-Meier curves and (B, D, F, H) time-dependent ROC curves
for each independent cohort.
3.4. Independent prognostic value of the six-FRG signature and clinical
characteristics
A univariate Cox regression analysis was used to investigate the
association between risk score and clinical characteristics, including
fluke infection, sex, age, and stage in the [105]GSE89749 and OEP001105
cohorts. A forest plot of the univariate Cox regression showed that
risk score (HR = 3.77), fluke infection (HR = 2.95), and stage (HR =
4.07) were significantly associated with patient survival in the
[106]GSE89749 cohort ([107] Figure 5A ). Similarly, risk score (HR =
2.08) and stage (HR = 2.24) were significantly associated with patient
survival in the forest plot of OEP001105 cohort ([108] Figure 5D ). A
multivariate Cox regression was performed to determine whether risk
score was an independent prognostic factor. The forest plot for the
multivariate Cox regression showed that risk scores were an independent
prognostic factor in the [109]GSE89749 and OEP001105 cohorts (p-value <
0.001; [110]Figures 5B, E ). Furthermore, the high-risk group was also
associated with fluke infection and high clinical stages in the
[111]GSE89749 cohort, but only with high clinical stages in OEP001105
cohort ([112] Figures 5C, F ).
Figure 5.
[113]Figure 5
[114]Open in a new tab
Univariate and multivariate Cox regression analyses of the six-FRG
signature and other clinical characteristics in the (A–C) [115]GSE89749
and (D–F) [116]OEP00105 cohorts.
3.5. GO and KEGG pathway enrichment analyses
GSEA was performed to investigate the underlying differences in
functions and biological processes between the low-risk and high-risk
groups based on GO and KEGG pathways. The GO analysis identified
biological process (BP), cellular component (CC), and molecular
function (MF) enriched in the high-risk CCA patient groups ([117]
Figure 6A ). KEGG pathway analysis identified differential pathway
enrichment between the low-risk and high-risk CCA patient groups ([118]
Figure 6B ).
Figure 6.
Figure 6
[119]Open in a new tab
Functional and pathway enrichment analyses in a training cohort. (A)
Gene Ontology (GO) analysis of differences between risk groups in three
functional categories: Biological processes (BPs), cellular components
(CCs), and molecular functions (MFs). (B) A Kyoto Encyclopedia of Genes
and Genomes (KEGG) pathway enrichment analysis of differences between
risk groups.
3.6. Immune cell infiltration and TME analysis
The 22 immune cell infiltration results estimated by the CIBERSORTx
algorithm showed that plasma cells, regulatory T cells (Tregs), resting
natural killer (NK) cells, and activated dendritic cells were
significantly higher in the high-risk than in the low-risk groups.
Gamma delta T cells and M1 and M2 macrophages were significantly lower
in the high-risk group than in the low-risk group ([120] Figure 7A ).
In addition, EPIC and MCP-counter algorithms were used to estimate CAF
scores in the low-risk and high-risk groups. Both EPIC and MCP-counter
algorithms showed that CAF scores were significantly higher in the
high-risk group than in the low-risk group ([121] Figures 7B, C ).
Furthermore, CAF specific marker expression was compared between the
risk groups. FAP, ACTA2, MFAP5, COL11A1, PDPN, and ITGA11 expression
levels were significantly higher in the high-risk group than in the
low-risk group ([122] Figure 7D ). These results indicated that immune
responses and CAF statuses differed in these two patient groups, which
might be translated to target the TME in CCA.
Figure 7.
[123]Figure 7
[124]Open in a new tab
Immune cell infiltration and tumor microenvironment (TME) analysis. (A)
A box plot showing the differences in multiple immune cells between
low-risk and high-risk groups based on the CIBERSORTx algorithm. (B, C)
A box plot showing differences in cancer-associated fibroblast (CAF)
score based on EPIC and MCP-counter algorithms. (D) A box plot
comparing CAF marker expression levels between risk groups. Key: ns,
not significant; *p < 0.05; **p < 0.01; ***p < 0.001; ****p < 0.0001.
3.7. Potential drugs targeting the two risk groups and an immune checkpoint
analysis
The GDSC database was used to estimate IC[50] of various drugs via R’s
“pRRophetic” package to predict drug sensitivity between the low-risk
and high-risk groups. The estimated IC[50] for 10 drugs (BI-2536,
GW843682X, Afatinib, Paclitaxel, Imatinib, WZ-1-84, [125]GW441756,
PHA-665752, CHIR-99021, and SB-216763) out of 138 screened drugs were
lower in the high-risk group than in the low-risk group, indicating
that the high-risk group was more sensitive to these drugs than the
low-risk group ([126] Figure 8A ). Moreover, an immune checkpoint
analysis showed that CD47, HHLA2, and TNFRSF14 expression was
significantly higher in the high-risk group than in the low-risk group
([127] Figure 8B ). Therefore, these immune checkpoints might be
effective immunotherapy targets in the high-risk CCA patient group.
Figure 8.
[128]Figure 8
[129]Open in a new tab
Drug sensitivity prediction and immune checkpoint analysis. (A)
Differences in estimated half-maximal inhibitory concentration (IC[50])
between risk groups for 10 candidate drugs. (B) A box plot comparing
immune checkpoint marker expression levels between risk groups. Key:
ns, not significant; *p < 0.05; **p < 0.01; ***p < 0.001; ****p <
0.0001.
3.8. Validation of the expression of six-FRG signature proteins in CCA
patients
A total of 8,320 proteins were identified in the OEP001105 cohort. Four
of six-FRG signature genes were present in this database (ACO1, GOT1,
SLC2A1, and SQLE). Therefore, correlations between their mRNA and
protein expression levels were analyzed in this cohort to confirm their
protein expression. Protein and mRNA levels were significantly
correlated for ACO1 (r = 0.78), GOT1 (r = 0.89), SLC2A1 (r = 0.83) and
SQLE (r = 0.78; [130]Figures 9A–D ). In addition, the score formula
obtained from the training cohort was used to evaluate patient survival
in this cohort based on the protein levels of these four genes.
Kaplan-Meier curves showed that survival time was significantly shorter
in the high-risk group than in the low-risk group ([131] Figure 9E ).
The AUCs at 1, 3, and 4 years of survival were 0.7200, 0.7013, and
0.7360, respectively ([132] Figure 9F ). These results suggested that
these four genes could provide an accurate prognostic signature. In
addition, The Human Protein Atlas (HPA) database
([133]https://www.proteinatlas.org/) ([134]25) was used to confirm the
protein expression of the six-FRG signature in CCA. The
immunohistochemistry images from The HPA database showed that ACO1 and
GOT1 levels were lower in tumor compared to normal tissues. In
contrast, FANCD2, PTGS2, SLC2A1, and SQLE were higher in tumor compared
to normal tissues ([135] Supplementary Figure S1 ).
Figure 9.
[136]Figure 9
[137]Open in a new tab
Validation of the six-FRG signature based on protein levels in the
OEP001105 cohort. (A–D) Scatter plots showing the correlation between
mRNA and protein expression levels of ACO1, GOT1, SLC2A1, and SQLE. (E)
A Kaplan-Meier curve comparing survival time between patient groups
defined on six-FRG signature protein levels. (F) Time-dependent ROC
curves.
3.9. Expression of the six-FRG signature in CCA cell lines and ferroptosis
inducer sensitivity
RT-qPCR was used to investigate the expression of six-FRG signature in
10 CCA cell lines relative to a non-tumor cholangiocyte cell line
(MMNK-1). ACO1 and GOT1 expression was downregulated in most CCA cell
lines compared to the MMNK-1 cell line. In contrast, FANCD2, PTGS2,
SLC2A1, and SQLE expression was upregulated in CCA cell line compared
to the MMNK-1 cell line ([138] Figures 10A–F ). Interestingly, the
expression of the six-FRG signature in CCA cell lines showed a similar
trend to their expression in CCA tissues. Moreover, the formula
constructed from the training cohort was used to calculate risk score
for each CCA cell line and divided them into high-risk and low-risk
groups. The functional role of the six-FRG signature in ferroptosis was
investigated by examining the sensitivity of the two CCA cell lines
with the highest (KKU-214 and RMCCA-1) and lowest (CCLP-1 and RBE) risk
scores to ferroptosis inducers ([139] Figure 10G ). Their sensitivity
to ferroptosis inducers erastin and RSL3 was compared using IC[50]
values. Interestingly, the two CCA cell lines with the lowest-risk
scores (CCLP-1 and RBE) were more sensitive to both ferroptosis
inducers. The CCA cell lines with the highest-risk scores (KKU-214 and
RMCCA-1) had higher IC[50] values than the CCA cell lines with the
lowest-risk scores following treatment with both ferroptosis inducers
([140] Figures 10H–J ). These results highlight the association between
the risk score and ferroptosis sensitivity. CCA cell lines with
higher-risk scores were more resistant to ferroptosis, indicating that
protective mechanisms against ferroptosis might be enhanced in such CCA
cell lines.
Figure 10.
[141]Figure 10
[142]Open in a new tab
Expression of the six-FRG signature in CCA cell lines and their
sensitivity to ferroptosis inducers. (A-F) The relative expression
levels of the six-FRG signature in 10 CCA cell lines normalized to a
non-tumor MMNK-1 cell line. (G) Risk scores calculated using six-FRG
signature in 10 CCA cell lines. (H) Cell viability of CCLP-1, RBE,
KKU-214, and RMCCA-1 treated with erastin. (I) Cell viability of
CCLP-1, RBE, KKU-214, and RMCCA-1 treated with RSL3. (J) A table
showing IC[50] of erastin and RSL3 in CCLP-1, RBE, KKU-214, and
RMCCA-1. Key: ns, not significant; *p < 0.05; **p < 0.01; ***p < 0.001.
4. Discussion
CCA is the second most common cancer in the hepatobiliary system.
Patients with CCA have poor prognoses and high mortality rate in which
patients in advanced stages have a low 5-year survival rate of 5% to
10%. Due to tumor heterogeneity and no effective therapy, patients with
CCA have the worst prognosis. Targeting programmed cell death is one of
the most effective cancer treatments, and dysregulation of this pathway
and its-related genes are directly associated with prognosis of
patients. Accumulating evidence has demonstrated that ferroptosis, a
recently identified regulated cell death pathway, is a promising cancer
therapy in several cancers. In addition, several studies have shown the
relationship between FRGs and patient prognosis. While few previous
studies have explored the relationship between FRG signatures and
prognosis in CCA patients, a comprehensive analysis and validation in
more patients and cohorts have not been performed. Moreover, to our
knowledge, studies focusing on the role of prognostic genes and their
associations with ferroptosis are limited. Consequently, how
ferroptosis contributes to CCA remains unclear. Therefore, discovering
a novel FRG signature might aid in predicting prognosis and developing
novel therapeutic targets that can help to improve the overall survival
of CCA patients.
This study constructed an FRG signature to predict the prognosis of CCA
patients. Seventeen DEGs with prognostic values were identified in a
training cohort using paired-sample t-tests and univariate Cox
regression analyses. We then obtained a six-FRG signature related to
patient survival in a LASSO Cox regression analysis in a training
cohort. Following Kaplan-Meier analyses, our FRG signature showed that
CCA patients with high-risk scores had poor prognoses, and low-risk
scores had a longer survival time, indicating that our new FRG
signature had strong prognostic potential in CCA patients. Predictive
ability was further confirmed via ROC curve analyses, where our FRG
signature had adequate predictive power. Furthermore, univariate and
multivariate Cox regression analyses indicated that the risk score
based on this six-FRG signature was an independent prognostic factor.
The risk scores were associated with fluke infection and clinical
stages in patients in the [143]GSE89749 cohort, but only with clinical
stages in patients in the OEP001105 cohort, potentially reflecting the
small number of fluke- infected patients in this cohort. Therefore, our
novel six-FRG signature can accurately predict prognoses for CCA
patients and divide them into low-risk and high-risk groups in which
appropriate therapeutic strategies can be used for personalized
therapy.
High expression of four genes in the six-FRG signature (FANCD2, PTGS2,
SLC2A1, and SQLE) were associated with poor prognosis in CCA patients
and low expression of the six-FRG other two genes (ACO1 and GOT1) were
associated with poor prognosis in CCA patients. Previous studies have
shown that most FRGs in the signature are involved in tumor progression
in CCA and various other cancers. ACO1, also known as IRP1, is an
RNA-binding protein that controls iron homeostasis by regulating TFRC
and FTH1 expression in CCA and hepatocellular carcinoma ([144]26,
[145]27). ACO1 depletion decreased iron levels and suppressed erastin-
and RSL3-induced ferroptosis in melanoma ([146]28). Moreover, GOT1
downregulation suppressed ferroptosis in melanoma by reducing
α-ketoglutarate which is a metabolic intermediate in ROS production
([147]29). However, the role of GOT1 in CCA remains unknown. In
contrast, FANCD2 was found to be a ferroptosis suppressor in bone
marrow injury by regulating the expression of iron metabolism and lipid
peroxidation-related genes ([148]30). FANCD2 was identified as a
prognostic gene associated with poor prognosis in colon cancer, lung
adenocarcinoma, clear cell renal cell carcinoma, and glioma
([149]31–[150]34). PTGS2, also known as COX-2, was identified as an
upregulated ferroptosis marker during ferroptosis ([151]11). PTGS2 was
upregulated in tumor tissues and promoted tumor progression and
chemotherapy resistance in various cancers ([152]35–[153]37). PTGS2 has
been shown to promote tumor development in CCA, while its inhibition
potentiated conventional chemotherapy effects ([154]38–[155]41).
SLC2A1, also known as GLUT1, is a glucose transporters family member.
SLC2A1 was found to be upregulated and play a role in tumor progression
in various cancers ([156]42–[157]45). A previous study identified
SLC2A1 as a prognostic gene in CCA patients ([158]46, [159]47).
Overexpression of SQLE, a key enzyme in cholesterol biosynthesis,
increased lipid peroxidation, leading to ferroptosis ([160]48). Recent
studies found that SQLE promoted tumor progression, and its expression
was associated with poor prognosis in breast cancer, pancreatic
adenocarcinoma, and bladder cancer ([161]49–[162]51). In summary,
consistent with the previous studies, our study showed that ACO1 and
GOT1 were downregulated and correlated with good prognosis in CCA
patients. In contrast, FANCD2, PTGS2, SLC2A1, and SQLE were upregulated
and correlated with poor prognosis in CCA patients. However, their
functional roles in ferroptosis and CCA progression remain unknown, and
mechanistic studies on each prognostic FRG in our signature are needed.
In addition, our GO analysis identified BP, CC, and MF enrichments in
the high-risk group. Among the BPs, the nuclear division process was
enriched in the high-risk group, indicating that they might have higher
proliferation than the low-risk group. The CC results showed that Golgi
apparatus components were enriched in the high-risk group. Previous
studies have shown that the Golgi apparatus plays important roles in
cellular redox control and prevents ferroptosis ([163]52, [164]53). The
MF results showed that cell adhesion molecules, such as laminin and
cadherin binding, and cell adhesion mediator activity were enriched in
the high-risk group. Recently, cell-cell interaction was found to
regulate ferroptosis sensitivity ([165]54). Moreover, a KEGG pathway
analysis was used to identify the main signaling pathways contributing
to CCA by comparing low-risk and high-risk groups. Many signal
transduction pathways frequently dysregulated in cancers were enriched
in the high-risk group, including the phosphatidylinositol, mammalian
target of rapamycin (mTOR), vascular endothelial growth factor (VEGF),
erb-be receptor tyrosine kinase (ERBB), Wnt, and mitogen-activated
protein kinase (MAPK) signaling pathways. These signaling pathways have
been shown to play important roles in CCA and various cancers
([166]55–[167]66). In contrast, amino acid, carbohydrate, and lipid
metabolism were enriched in the low-risk group, while glycan
biosynthesis and metabolism were found to be enriched in the high-risk
group. In summary, our study has shown differences in signaling pathway
enrichment between CCA risk groups, which could be targeted to develop
treatment strategies and improve prognosis in each CCA risk group.
Accumulating evidence has shown the interplay between tumor cells and
other cell types in the TME, including a subset of immune cells and
CAFs that play important roles in tumor progression and developing
therapeutic resistance ([168]67). CCA is a dysmorphic tumor with
abundance CAFs and immunosuppressive cells in the TME ([169]68).
Therefore, we further investigated immune cell and CAF enrichment by
comparing low-risk and high-risk groups. Tregs are an immune system
component that suppressed anticancer immunity and are associated with
poor prognoses in various cancers ([170]68–[171]71). Recent studies
showed that FOXP3+, a Treg marker associated with poor prognosis in CCA
patients ([172]72, [173]73). Plasma cells were found to be a source of
immunosuppressive factor interleukin (IL)-10 ([174]74, [175]75).
Moreover, CAFs are major TME components with tumorigenic properties,
especially, in immunosuppressive TME modulation ([176]76). This study
found that levels of these immunosuppressive components, including
Tregs, plasma cells, and CAFs were significantly higher in the
high-risk group. Therefore, immunosuppressive components might be
promising therapeutic targets to improve the efficacy of CCA treatment
and patient survival.
Drug sensitivity prediction analysis was performed to overcome
treatment resistance and improve CCA patient prognosis, particularly in
the high-risk group. Our analysis predicted 10 effectives potentially
candidate drugs for the high-risk CCA patient group. Paclitaxel,
Imatinib, and Afatinib are US Food and Drug Administration
(FDA)-approved drugs used in clinics to treat various cancers. A recent
study showed that Paclitaxel could be a drug candidate for CCA patients
resistant to conventional therapies ([177]77). Our analysis supported
Paclitaxel potentially having a better effect in treating CCA patients
within the high-risk group. The ABL, KIT, and PDGFR inhibitor Imatinib
was effective in treating gastrointestinal stromal tumor with KIT or
PDGFR overexpression or mutation ([178]78). The epidermal growth factor
receptor (EGFR) and ERBB tyrosine kinase inhibitor Afatinib has been
used as a first-line drug for non-small cell lung cancer patients with
an EGFR mutation ([179]79). However, relatively few studies have
investigated the efficacy of these kinase inhibitors in CCA patients
([180]80–[181]82). Moreover, PLK1 inhibitors BI-2536 and GW843682X were
predicted to be effective drugs for the high-risk group. Consistent
with our results, previous studies have shown that PLK1 was associated
with poor prognosis in CCA patients, and its inhibition was effective
against CCA cells ([182]83–[183]85). NTRK1 fusion has been found in CCA
patients ([184]86) resulting in constitutive TRKA activation, and its
inhibition showed positive responses in specific CCA patient groups
([185]87). In this study, a selective TRKA inhibitor [186]GW441756 was
shown more effective in the high-risk group. In addition, selective
c-Met inhibitor PHA-665752 and GSK3B inhibitors CHIR-99021 and
SB-216763 were shown to be more effective in the high-risk group. Both
c-MET and GSK3B were associated with poor prognosis, and targeting
c-MET or GSK3B has been reported to be potentially effective in
treating CCA patients ([187]88–[188]93). Altogether, based on risk
stratification groups, our study predicted effective candidate drugs
for treating CCA patients, which could be used in precision therapy to
improve their prognosis.
Immunotherapy has been proposed as a potential therapeutic option for
CCA ([189]94, [190]95). However, the outcomes of anti-PD-1/PD-L1 immune
checkpoint inhibitors (ICIs) remain controversial in CCA. In this
study, PD-1 (PDCD-1) and PD-L1 (CD274) expression levels did not differ
between low-risk and high-risk groups. Interestingly, other immune
checkpoint markers, including CD47, HHLA2, and TNFRSF14 showed
significantly higher expression in the high-risk group. Therefore,
these immune checkpoints might be potential targets for developing ICIs
to reactivate antitumor immunity which might improve the survival of
patients with poor prognoses.
Previous studies have analyzed FRG signatures in several cancers.
However, most have analyzed FRG signatures and classified patients into
low-risk and high-risk groups that did not reflect relative ferroptosis
levels ([191]96–[192]98). Therefore, this study calculated risk scores
based on the expression levels of a six-FRG signature in each CCA cell
line and could divide them into low-risk and high-risk groups. Using
two representative CCA cell lines of the low-risk and high-risk groups,
our study showed that low-risk score CCA cell lines were significantly
more sensitive to ferroptosis inducers than high-risk score CCA cell
lines. This result suggests that ferroptosis resistance might explain
the relationship between the high-risk group and poor prognosis.
Consistent with our findings, a previous study showed that activating
ferroptosis suppressor genes enabled CCA cells to evade ferroptosis
([193]99). Therefore, inhibiting ferroptosis suppressor genes and
targeting ferroptosis resistance mechanisms might be effective
therapeutic strategies for improving prognosis, particularly in the
high-risk group. Besides ferroptosis resistance mechanisms, other
factors, such as CCA stage, might contribute to differences in
ferroptosis inducer sensitivity in the two CCA cell line groups.
Unfortunately, information on the stages of CCA cell lines is only
available for the high-risk group (i.e., RMCCA-1: T2N0M0; KKU-214:
stage IVB)
However, our study had some limitation. First, the data used in this
study were collected from public databases. Consequently, they differed
appreciably in their CCA patient numbers, patient heterogeneity,
ethnicity, and etiology. Second, the FRGs were identified from FerrDb
and previous studies. However, the role of ferroptosis in cancers is
still in its infancy. Therefore, some unidentified FRGs might be
missing from our analyses. Finally, further in vitro and in vivo
studies are needed to investigate the roles of these prognostic genes
in ferroptosis and their underlying mechanisms in CCA.
5. Conclusions
In summary, our study showed that a six-FRG signature scoring model
could divide CCA patients into low-risk and high-risk groups. Our novel
FRG signature model effective predicted the prognosis of CCA patients,
potentially providing prognostic biomarkers and potential therapeutic
targets for CCA patients. Moreover, oncogenic signaling pathways,
immune cell and CAF infiltration, drug sensitivity, and immune
checkpoint marker expression differed between two CCA risk groups,
which might represent novel therapeutic targets for improving survival,
particularly of patients with poor prognoses. In addition, we predicted
drug candidates for CCA patients in the high-risk group. Based on
ferroptosis sensitivity in low-risk and high-risk CCA cell lines, our
results suggest that ferroptosis resistance is associated with the
high-risk group and targeting ferroptosis resistance mechanisms in this
group could be a promising therapeutic strategy for these CCA patients.
Data availability statement
The datasets presented in this study can be found in online
repositories. The names of the repository/repositories and accession
number(s) can be found in the article/[194] Supplementary Material .
Author contributions
AS-F collected data, performed all in vitro experiments, analyzed
statistics and bioinformatics, organized the figures and drafted the
manuscript under the supervision of SJ. AM served as the mentor of SJ
for the Research Grant for New Scholars and participated in the editing
of this manuscript. SJ conceived and designed the study; analyzed and
interpreted the data; participated in the writing and editing of this
manuscript. All authors contributed to the article and approved the
submitted version.
Acknowledgments