ArticlePDF Available

Abstract

Background/Objectives: Aneuploidy is a prevalent cancer feature that occurs in many solid tumors. For example, high-grade serous ovarian cancer shows a high level of copy number alterations and genomic rearrangements. This makes genomic variants appealing as diagnostic or prognostic biomarkers, as well as for their easy detection. In this study, we focused on copy number (CN) losses shared by ovarian cancer stem cells (CSCs) to identify chromosomal regions that may be important for CSC features and, in turn, for patients’ prognosis. Methods: Array-CGH and bioinformatic analyses on three CSCs subpopulations were performed. Results: Pathway and gene ontology analyses on genes involved in copy number loss in all CSCs revealed a significant decrease in mRNA surveillance pathway, as well as miRNA-mediated gene silencing. Then, starting from these CN losses, we validated their potential prognostic relevance by analyzing the TCGA cohort. Notably, losses of 4q34.3-q35.2, 8p21.2-p21.1, and 18q12.2-q23 were linked to increased genomic instability. Loss of 18q12.2-q23 was also related to a higher tumor stage and poor prognosis. Finally, specific genes mapping in these regions, such as PPP2R2A and TPGS2A, emerged as potential biomarkers. Conclusions: Our findings highlight the importance of genomic alterations in ovarian cancer and their impact on tumor progression and patients’ prognosis, offering advance in understanding of the application of numerical aberrations as prognostic ovarian cancer biomarkers.
Citation: Jemma, A.; Ardizzoia, A.;
Redaelli, S.; Bentivegna, A.; Lavitrano,
M.; Conconi, D. Prognostic Relevance
of Copy Number Losses in Ovarian
Cancer. Genes 2024,15, 1487. https://
doi.org/10.3390/genes15111487
Academic Editors: Albert Jeltsch and
Hilal Arnouk
Received: 3 October 2024
Revised: 29 October 2024
Accepted: 18 November 2024
Published: 19 November 2024
Copyright: © 2024 by the authors.
Licensee MDPI, Basel, Switzerland.
This article is an open access article
distributed under the terms and
conditions of the Creative Commons
Attribution (CC BY) license (https://
creativecommons.org/licenses/by/
4.0/).
Article
Prognostic Relevance of Copy Number Losses in Ovarian Cancer
Andrea Jemma
1
, Alessandra Ardizzoia
1,2
, Serena Redaelli
1
, Angela Bentivegna
1
, Marialuisa Lavitrano
1
and Donatella Conconi 1, *
1School of Medicine and Surgery, University of Milano-Bicocca, 20900 Monza, Italy;
a.jemma@campus.unimib.it (A.J.); a.ardizzoia@campus.unimib.it (A.A.); serena.redaelli@unimib.it (S.R.);
angela.bentivegna@unimib.it (A.B.); marialuisa.lavitrano@unimib.it (M.L.)
2Fondazione Istituto di Oncologia Molecolare ETS (IFOM), The AIRC Institute for Molecular Oncology,
20139 Milan, Italy
*Correspondence: donatella.conconi@unimib.it
Abstract: Background/Objectives: Aneuploidy is a prevalent cancer feature that occurs in many
solid tumors. For example, high-grade serous ovarian cancer shows a high level of copy number
alterations and genomic rearrangements. This makes genomic variants appealing as diagnostic or
prognostic biomarkers, as well as for their easy detection. In this study, we focused on copy number
(CN) losses shared by ovarian cancer stem cells (CSCs) to identify chromosomal regions that may
be important for CSC features and, in turn, for patients’ prognosis. Methods: Array-CGH and
bioinformatic analyses on three CSCs subpopulations were performed. Results: Pathway and gene
ontology analyses on genes involved in copy number loss in all CSCs revealed a significant decrease
in mRNA surveillance pathway, as well as miRNA-mediated gene silencing. Then, starting from these
CN losses, we validated their potential prognostic relevance by analyzing the TCGA cohort. Notably,
losses of 4q34.3-q35.2, 8p21.2-p21.1, and 18q12.2-q23 were linked to increased genomic instability.
Loss of 18q12.2-q23 was also related to a higher tumor stage and poor prognosis. Finally, specific
genes mapping in these regions, such as PPP2R2A and TPGS2A, emerged as potential biomarkers.
Conclusions: Our findings highlight the importance of genomic alterations in ovarian cancer and
their impact on tumor progression and patients’ prognosis, offering advance in understanding of the
application of numerical aberrations as prognostic ovarian cancer biomarkers.
Keywords: ovarian cancer; biomarkers; copy number loss; cancer stem cells; copy number alterations;
numerical aberrations
1. Introduction
Advances in high-throughput “omics” technologies produced a huge amount of data,
including cancer genomics data. Aneuploidy is a hallmark feature in approximately 90% of
solid tumors, reflecting widespread genomic instability [
1
]. Although its exact role in tumor
initiation and progression remains unclear, it is thought to contribute to the complexity of
chromosomal aberrations frequently observed in cancer cells. Traditionally, aneuploidy
refers to numerical changes involving whole chromosomes, but more recent studies have
expanded this concept to include segmental alterations, such as losses or gains of chromo-
some arms, which are often categorized as copy number alterations (CNAs) [
2
]. For this
reason, it can be challenging to compare different studies because numerical alterations
can involve either few genes (CNAs) or entire chromosome region (SCAs—segmental
chromosomal aberrations; copy number gains or losses affecting one chromosome arm or a
segment within a chromosome arm with at least 100 contiguous oligonucleotide probes) [
3
].
Numerical aberrations can readily be detected using conventional and molecular
cytogenetics techniques, routinely used in clinics, making them appealing biomarkers for
patients’ prognosis [
2
]. As a matter of fact, CNA burden or specific CNAs are associated
with poor outcomes in many cancers including neuroblastoma [
3
,
4
], breast cancer [
5
,
6
],
bladder cancer [7], glioblastoma [8,9] and ovarian cancer [1013].
Genes 2024,15, 1487. https://doi.org/10.3390/genes15111487 https://www.mdpi.com/journal/genes
Genes 2024,15, 1487 2 of 14
Cancer stem cells (CSCs) are a subpopulation of tumor cells known for driving tu-
mor growth, treatment failure, and cancer relapse [
14
]. In this context, ovarian cancer
spheroids, found in malignant ascites, represent that subpopulation, responsible for ovar-
ian cancer dissemination, impairment of treatments effectiveness, and unfavorable patient
prognosis [15]. For these reasons, understanding the connection between CNAs and gene
expression in CSCs could be vital for revealing how genetic instability contributes to the
tumor’s aggressiveness. Moreover, several studies show that a large proportion of genomic
changes in cancer cells can be attributed to CNAs, reinforcing their role in driving key
aspects of tumor behavior [16].
In a previous work [
10
], we identified a potential prognostic role of AhRR and
PPP1R3C expression in serous ovarian cancer, starting from a detailed bioinformatics
analysis of copy number gain arising in CSCs by array comparative genomic hybridization
(array-CGH) analysis. In this work, we turn our attention to copy number losses in order
to identify in a similar way chromosomal regions or genes with a specific role in ovarian
cancer stem cells and, in turn, for patients’ prognosis.
2. Materials and Methods
2.1. Cell Lines
Ovarian cancer cell lines Caov3, Ovcar5, and Ovcar8 were purchased from ATCC
(American Type Culture Collection, Manassas, VA, USA) and Sigma-Aldrich (St. Louis, MO,
USA). Caov3 were grown in Dulbecco’s modified Eagle’s medium (DMEM); Ovcar5 and
Ovcar8 were grown in RPMI 1640. Both media were completed with the addition of 10%
fetal bovine serum (FBS) and 1% penicillin–streptomycin. All reagents were purchased from
EuroClone (Milano, Italy). All the cell lines were maintained in a humidified atmosphere at
37 C with 5% CO2.
Ovarian cancer stem cells (represented by ovarian cancer spheroids) were previously
isolated and characterized [
10
]. Briefly, spheroids were generated following an anchorage-
independent growth assay starting from the three different cell lines and then characterized
by stemness markers’ expression and clonogenic nature.
2.2. Array Comparative Genomic Hybridization
Array comparative genomic hybridization (array-CGH) experiments were previously
reported [
10
]. Briefly, DNA was extracted from ovarian CSCs and the corresponding
cell lines using an QIAamp DNA Mini Kit according to the manufacturer’s instructions
(QIAGEN, Hilden, Germany). After quantification with a Nanodrop ND-2000 spectropho-
tometer (Thermo Fisher Scientific, Waltham, MA, USA), samples with a concentration over
10
µ
g/mL and an absorbance ratio A
260/280
over 1.8 and A
260/230
over 1.7 were used for
analysis. Array-CGH analysis was performed using a SurePrint G3 Human CGH Microar-
ray 8
×
60 K (Agilent Technologies, Santa Clara, CA, USA) according to the manufacturer’s
instructions. The arrays were scanned at 2
µ
m resolution and analyzed using Agilent
Feature Extraction and Agilent Cytogenomics v5.2 software (Agilent Technologies, Santa
Clara, CA, USA). The Aberration Detection Method 2 (ADM-2) algorithm was used to
compute and assist the identification of aberrations in a given sample (threshold = 5.0),
assigning a statistical score based on the average quality weighted log ratio (DLRS) of
the sample and reference channels. We applied a filtering option of a minimum of three
aberrant consecutive probes and a minimum absolute average log2 ratio that differs among
all samples and depends on DLRS values. Log2ratio values over 1 identify amplification;
values under
1.7 identify complete loss. Log2ratio values over 0.6 identify non-mosaic
gains; values under
1 identify non-mosaic losses. Accordingly, log2 ratio values for
mosaic gains range between the DLRS value and 0.6 and for mosaic losses between the
DLRS value and
1. As a reference genomic DNA sample, woman-matched DNA was
provided by Agilent (Agilent Technologies).
Genes 2024,15, 1487 3 of 14
2.3. Bioinformatic Analyses
2.3.1. Analysis of Genes Involved in Copy Number Losses
The Database for Annotation, Visualization and Integrated Discovery (DAVID, https:
//david.ncifcrf.gov/tools.jsp, accessed on 29 July 2024) platform was used to identify
enriched REACTOME pathways, Kyoto Encyclopedia of Genes and Genomes (KEGG)
pathways, and GO (Gene Ontology) terms (BP: biological process, and MF: molecular
function) [17]. A p-value less than 0.05 was considered statistically significant.
2.3.2. Correlation of Chromosome Aberrations and Clinical Data
To evaluate a potential prognostic role of segmental chromosome aberrations, we
analyzed the TCGA-OV (The Cancer Genome Atlas Ovarian Cancer) cohort using the
cBioPortal for Cancer Genomics web server (https://www.cbioportal.org/, accessed on
29 July 2024). Ovarian serous cystadenocarcinoma (TCGA PanCancer Atlas) was chosen.
Samples with or without loss were selected to form the different groups (4q34.3-q35.2
loss vs. no loss, 8p21.2-p21.1 loss vs. no loss, and 18q12.2-q23 loss vs. no loss), which
were compared to identify differences in clinical parameters (survival, clinical, protein,
arm-level CNA).
For the cBioportal web server, copy number data from the GDC analysis pipelines
were provided in ASCAT format and converted to discrete GISTIC data using the following
thresholds:
2 or Deep Deletion indicates a homozygous deletion;
1 or Shallow Deletion
indicates a heterozygous deletion; 0 is diploid; 1 or Gain indicates a low-level gain (a few
additional copies); 2 or Amplification indicates a high-level amplification (more copies)
(https://www.cbioportal.org/).
The Genomic Data Commons (GDC) Data Portal (https://portal.gdc.cancer.gov/,
accessed on 29 July 2024) provides access to the subset of TCGA data that has been har-
monized against GRCh38 (hg38) using GDC Bioinformatics Pipelines, which provides
methods to the standardization of biospecimen and clinical data and the generation of
derived data [
18
]. The CNA pipeline uses either NGS or Affymetrix SNP 6.0 (SNP6) array
data to identify genomic regions that are repeated and infer the copy number of these
repeats. Three sets of pipelines have been used for CNV inferences: ASCAT, ABSOLUTE,
and DNAcopy.
We chose the TCGA-OV project, divided the patients into cohorts according to the
tumor stage or age at diagnosis (under or over 63 years), and then we compared the
frequency of copy number gain and loss in each cohort.
2.3.3. Analysis of FHOD3, TPGS2, and KIAA1328 Expression in Ovarian Cancer
GEPIA2 (http://gepia2.cancer-pku.cn/, accessed on 29 July 2024) and Kaplan–Meier
plotter (https://kmplot.com/analysis/, accessed on 29 July 2024) web servers were used
to analyze the correlation between the selected genes’ expressions and patients’ overall
survival and disease-free survival in ovarian cancer. The selected cut-off value was “median
cut-off” (GEPIA2, OV Dataset) and “auto selected best cut-off” (Kaplan–Meier plotter
mRNA Chip). p-value < 0.05 was considered statistically significant. OncoDB web server
(https://oncodb.org/, accessed on 29 July 2024) was consulted for the analysis of correlation
between expression of genes and clinical stage of the tumor. p-value < 0.05 was considered
statistically significant.
3. Results
3.1. Lost Genes Were Involved in mRNA Processing
Ovarian cancer spheroids were obtained from three ovarian cancer cell lines (Caov3,
Ovcar5, and Ovcar8) belonging to high-grade serous histotype [
19
23
]. In our previous
study, we checked the expression of ovarian cancer stemness markers ALDH, CD44, ABCG2,
and NANOG and spheroids’ clonogenic nature through PKH staining [
10
]. All ovarian
cancer spheroids showed an increased expression of stemness markers compared to the
Genes 2024,15, 1487 4 of 14
corresponding cell line and spheroids’ clonogenic nature. These results validate our model
as ovarian cancer stem cells [10].
Firstly, based on the copy number losses shared by all three CSC subpopulations,
we identified a list of 1253 genes, which underwent enrichment analyses in GO terms,
REACTOME pathways, and KEGG pathways through the DAVID platform (Table S1).
KEGG pathway enrichment analysis revealed a significant decrease in the mRNA
surveillance pathway. This pathway serves as a quality control mechanism that identifies
and degrades abnormal mRNAs, and it includes nonsense-mediated mRNA decay (NMD),
nonstop mRNA decay (NSD), and no-go decay (NGD) [
24
]. In particular, lost genes
involved in this pathway were almost exclusively located on chromosome X, except for the
PPP2R2A gene located in 8p21.1: GSPT2 (Xp11.22), UPF3B (Xq24), CSTF2,NXF2,NXF2B,
NXF3 (all Xq22.1), NCBP2L (Xq22.3), NXT2 (Xq23), PABPC1L2A and PABPC1L2B (Xq13.2),
and PABPC5 (Xq21.31). Instead, REACTOME pathway enrichment analysis highlighted a
loss of downregulation of SMAD2/3:SMAD4 transcriptional activity.
GO enrichment analysis identified a strong downregulation of gene silencing by
miRNA (BP: biological process), with 60 downregulated miRNAs in CSCs, confirmed
by a decrease of mRNA binding involved in posttranscriptional gene silencing GO MF
(molecular function), with 31 downregulated miRNAs in CSCs.
Interestingly, some GO terms associated with mRNA processing (such as negative
regulation of transcription by RNA polymerase II, poly(A)+ mRNA export from nucleus,
mRNA 3
-UTR binding, poly(A) binding, and mRNA export from nucleus) were underrep-
resented in our CSC subpopulations. Moreover, CSCs showed upregulation of angiogenesis,
autophagy, G0 to G1 transition, and ERK1 and ERK2 cascade.
Taken together, these results explain some characteristics of our CSCs models and
confirm the importance of copy number losses in tumorigenesis and cancer progression.
3.2. Copy Number Losses Correlated with Clinical Parameters in the cBioPortal Web Server
Analyzing CNAs and SCAs shared by all the three CSCs subpopulations, we identified
three autosomal regions: 4q34.3-q35.2 (nt 178341937-191133609, 206 probes), 8p21.2-p21.1
(nt 25774629-28393917, 61 probes), and 18q12.2-q23 (nt 32483714-75486904, 686 probes)
(
Figure 1
). Moreover, loss of the entire chrX was observed (nt 4239811-152009316,
2467 probes).
To evaluate the potential prognostic role of these copy number losses, we analyzed the
correlation between loss in these regions and clinical parameters from the TCGA-OV (The
Cancer Genome Atlas Ovarian Cancer) cohort using the cBioPortal for Cancer Genomics
web server. Patients were divided into two groups based on the presence or absence of
18q12.2-q23 loss in tumor biopsies. The clinical data comparison between these two groups
displayed a statistically significant difference in disease-free survival (DFS, Figure 2A).
Moreover, an increased aneuploidy score and a higher fraction of genome altered in
ovarian cancer samples carrying 18q12.2-q23 loss were observed (Table 1,
Figure S1A,B
).
Thirteen proteins showed differential expression across the two groups of samples when
protein levels were analyzed using mass spectrometry (Figure 2B). GO and pathway
enrichment analyses on proteins overexpressed in the “no-loss” group of samples showed
only one statistically significant GO term (negative regulation of transcription, DNA-
templated). Enrichment analyses on two proteins overexpressed in the 18q “loss” group of
samples identified a statistically significant increase in cysteine and methionine metabolism
(KEGG pathway).
On the other hand, samples carrying 4q34.3-q35.2 loss showed an increase in the
fraction of genome altered (p< 0.01), as well as in the mutational count (p< 0.01) (Table 1,
Figure S1C,D). Finally, loss of 8p21.2-p21.1 was associated with a higher mutational count
(p< 0.01) and increased tumor mutational burden (TMB) nonsynonymous (p< 0.01) (Table 1,
Figure S1E,F). Taken together, these data suggest that the identified aberrations could
correlate with generalized genetic–genomic instability.
Genes 2024,15, 1487 5 of 14
Genes 2024, 15, x FOR PEER REVIEW 5 of 14
samples identified a statistically significant increase in cysteine and methionine metabo-
lism (KEGG pathway).
Figure 1. Copy number losses shared by all three CSCs subpopulations: 4q34.3-q35.2 (206 probes),
8p21.2-p21.1 (61 probes), 18q12.2-q23 (686 probes), and the entire chrX (2467 probes). Red: CN loss;
blue: CN gain; red cycles: chromosome number.
On the other hand, samples carrying 4q34.3-q35.2 loss showed an increase in the frac-
tion of genome altered (p < 0.01), as well as in the mutational count (p < 0.01) (Table 1,
Figure S1C,D). Finally, loss of 8p21.2-p21.1 was associated with a higher mutational count
(p < 0.01) and increased tumor mutational burden (TMB) nonsynonymous (p < 0.01) (Table
1, Figure S1E,F). Taken together, these data suggest that the identified aberrations could
correlate with generalized geneticgenomic instability.
As a result, we searched for potential variations in arm-level CNAs of other chromo-
somes in samples with the identified CN losses. We found statistically significant differ-
ences in the frequency of in arm-level CNAs in samples with CN losses with respect to
samples without losses, especially in samples with 8p21.2-p21.1 loss (Table 1). This result,
in addition to the previously reported higher tumor mutational burden, underlines that
these samples are characterized by a great genetic instability.
Given the small number of genes in CN losses of chr8 and chr4, we carried out en-
richment analyses in GO terms, REACTOME pathways, and KEGG pathways in order to
uncover a possible explanation for the observed data. A statistically significant GO term
(positive regulation of the apoptotic process, p < 0.01, BP: biological process) was found
Figure 1. Copy number losses shared by all three CSCs subpopulations: 4q34.3-q35.2 (206 probes),
8p21.2-p21.1 (61 probes), 18q12.2-q23 (686 probes), and the entire chrX (2467 probes). Red: CN loss;
blue: CN gain; red cycles: chromosome number.
As a result, we searched for potential variations in arm-level CNAs of other chro-
mosomes in samples with the identified CN losses. We found statistically significant
differences in the frequency of in arm-level CNAs in samples with CN losses with respect
to samples without losses, especially in samples with 8p21.2-p21.1 loss (Table 1). This result,
in addition to the previously reported higher tumor mutational burden, underlines that
these samples are characterized by a great genetic instability.
Given the small number of genes in CN losses of chr8 and chr4, we carried out
enrichment analyses in GO terms, REACTOME pathways, and KEGG pathways in order to
uncover a possible explanation for the observed data. A statistically significant GO term
(positive regulation of the apoptotic process, p< 0.01, BP: biological process) was found
by DAVID analysis of the genes involved in 8p21.2-p21.1 CN, indicating that the loss of
these genes may result in the loss of apoptosis. No interesting cluster was identified for
4q34.3-q35.2 genes.
Table 1. Variations in clinical parameters and chromosome arm-level CNAs in samples carrying
4q34.3-q35.2, 8p21.2-p21.1, or 18q12.2-q23 loss. Only chromosome arms with statistically significant
differences are reported.
4q34.3-q35.2 Loss vs.
No Loss
8p21.2-p21.1 Loss vs.
No Loss
18q12.2-q23 Loss vs.
No Loss
Aneuploidy score unvaried unvaried
Fraction of genome altered unvaried
Mutational count unvaried
TMB (nonsynonymous) unvaried unvaried
2p unvaried unvaried CN gain
2q unvaried CN gain CN gain
3q unvaried CN gain unvaried
Genes 2024,15, 1487 6 of 14
Table 1. Cont.
4q34.3-q35.2 Loss vs.
No Loss
8p21.2-p21.1 Loss vs.
No Loss
18q12.2-q23 Loss vs.
No Loss
4p CN loss unvaried CN loss
4q arm involved in CNA unvaried CN loss
5p unvaried CN gain unvaried
5q unvaried CN loss unvaried
6p unvaried CN gain CN loss
7p unvaried unvaried CN loss
7q unvaried unvaried CN loss
8p unvaried arm involved in CNA unvaried
9p unvaried unvaried CN loss
9q CN loss CN loss unvaried
10p unvaried CN gain unvaried
10q unvaried CN loss unvaried
11q unvaried CN gain CN loss
12q unvaried unvaried CN loss
13q unvaried CN loss unvaried
14q unvaried CN loss unvaried
15q unvaried CN loss unvaried
16p CN loss unvaried CN loss
16q unvaried CN loss CN loss
17p unvaried CN gain unvaried
18p unvaried CN gain CN loss
18q CN loss unvaried arm involved in CNA
19p unvaried unvaried CN loss
20p CN gain unvaried unvaried
20q CN gain CN gain unvaried
: increase; : decrease.
Genes 2024, 15, x FOR PEER REVIEW 6 of 14
by DAVID analysis of the genes involved in 8p21.2-p21.1 CN, indicating that the loss of
these genes may result in the loss of apoptosis. No interesting cluster was identified for
4q34.3-q35.2 genes.
Figure 2. (A) Disease-free survival data of TCGA-OV patients divided into group A (samples with
18q12.2-q23 loss) and group B (samples without 18q12.2-q23 loss). (B) Dierential expressed protein
between samples of group A and group B analyzed using mass spectrometry (cBioPortal for Cancer
Genomics web server). Blue dots: signicant dierentially expressed proteins between the two
groups. Grey dots: not signicant dierentially expressed proteins between the two groups.
Table 1. Variations in clinical parameters and chromosome arm-level CNAs in samples carrying
4q34.3-q35.2, 8p21.2-p21.1, or 18q12.2-q23 loss. Only chromosome arms with statistically signicant
dierences are reported.
4q34.3-q35.2 Loss vs.
no Loss
8p21.2-p21.1 Loss vs.
no Loss
18q12.2-q23 Loss vs.
no Loss
Aneuploidy score
unvaried
unvaried
Fraction of genome altered
unvaried
Mutational count
unvaried
TMB (nonsynonymous)
unvaried
unvaried
2p
unvaried
unvaried
CN gain
2q
unvaried
CN gain
CN gain
3q
unvaried
CN gain
unvaried
4p
CN loss
unvaried
CN loss
4q
arm involved in CNA
unvaried
CN loss
Figure 2. (A) Disease-free survival data of TCGA-OV patients divided into group A (samples with
18q12.2-q23 loss) and group B (samples without 18q12.2-q23 loss). (B) Differential expressed protein
between samples of group A and group B analyzed using mass spectrometry (cBioPortal for Cancer
Genomics web server). Blue dots: significant differentially expressed proteins between the two
groups. Grey dots: not significant differentially expressed proteins between the two groups.
Genes 2024,15, 1487 7 of 14
3.3. Copy Number Losses Correlated with Clinical Parameters in the GDC Data Portal
We additionally investigated the potential correlations between clinical parameters
and CN loss in 4q34.3-q35.2, 8p21.2-p21.1, and 18q12.2-q23 through the GDC Data Portal be-
cause the two repositories process the TCGA data differently, using different bioinformatics
methods to analyze copy number alterations.
First, we divided the patients into two groups according to the median age at diagnosis
(63 years) and checked the percentage of gains and losses in both groups (Figure 3A). A
statistically significant difference (p< 0.05) in 8p21.2-p21.1 CN percentage was found; in
particular, a lower percentage of 8p21.2-p21.1 gain was observed in samples of the youngest
group (age at diagnosis under 63 years), while the percentage of CN loss was similar in
both groups (Figure 3A).
Genes 2024, 15, x FOR PEER REVIEW 8 of 14
Figure 3. (A) 4q34.3-q35.2, 8p21.2-p21.1, and 18q12.2-q23 loss and gain percentages in samples of
patients categorized by age at diagnosis. (B) Distribution of 18q copy number in samples categorized
by clinical stage. #: statistically signicant dierence between stage IV samples and samples of all
other stages, p < 0.05; §: statistically signicant dierence between stage IV and stage II/III samples,
p < 0.05; *: statistically signicant dierence between stage IV and stage III samples, p < 0.05. (C)
Localization of the 18q sub-regions represented in B (red boxes).
3.4. 18.q12.2 Region Correlated with Clinical Stage, Overall Survival, and Progression-Free
Survival
Considering the previous results, we focused our aention on lost genes within the
18q12.2 region that showed a signicant correlation (p < 0.05) with stage IV: FHOD3,
TPGS2, and KIAA1328 (Table S2). Using the GEPIA2 web server, we identified a
statistically signicant association between decreased expression of these genes and
reduced disease-free survival (DFS) in ovarian cancer patients (p < 0.05), suggesting a
prognostic relevance for this region. Moreover, we also found a link between gene
expression and overall survival (OS) (Figure 4A, p < 0.05). The OncoDB web server further
confirmed a correlation between the expression of these three genes and tumor stage,
consistent with the observed CNA (Figure S2). Finally, to evaluate the role of each single
gene, we consulted the KaplanMeier Ploer web server, selecting the serous histotype.
Only TPGS2A showed a strong correlation between expression and both DFS and OS in
ovarian cancer patients (Figure 4B), suggesting its pivotal role in ovarian cancer.
Figure 3. (A) 4q34.3-q35.2, 8p21.2-p21.1, and 18q12.2-q23 loss and gain percentages in samples of
patients categorized by age at diagnosis. (B) Distribution of 18q copy number in samples categorized
by clinical stage. #: statistically significant difference between stage IV samples and samples of
all other stages, p< 0.05; §: statistically significant difference between stage IV and stage II/III
samples, p< 0.05; *: statistically significant difference between stage IV and stage III samples, p< 0.05.
(C) Localization of the 18q sub-regions represented in B (red boxes).
On the other hand, considering the 18q12.2-q23, we evidenced a statistically significant
enrichment in the percentage of stage IV samples with loss of this region (p< 0.05,
Table S2
).
We checked the distribution of gain and loss also within the 18q12.2-q23 sub-regions
(Figure 3B,C). Different percentages of CNAs among stages were identified (Table S2). In
particular, stage IV showed a higher percentage of samples with loss of 18q12.2, as well
as samples with loss of 18q12.3 and 18q12.3-q21.1 compared to other stages (Figure 3B,C).
These findings could validate the role of this CN loss and demonstrate the involvement of
Genes 2024,15, 1487 8 of 14
18q loss in ovarian cancer progression. No correlation between 8p or 4q losses and tumor
stage was evidenced.
3.4. 18.q12.2 Region Correlated with Clinical Stage, Overall Survival, and
Progression-Free Survival
Considering the previous results, we focused our attention on lost genes within the
18q12.2 region that showed a significant correlation (p< 0.05) with stage IV: FHOD3,TPGS2,
and KIAA1328 (Table S2). Using the GEPIA2 web server, we identified a statistically
significant association between decreased expression of these genes and reduced disease-
free survival (DFS) in ovarian cancer patients (p< 0.05), suggesting a prognostic relevance
for this region. Moreover, we also found a link between gene expression and overall
survival (OS) (Figure 4A, p< 0.05). The OncoDB web server further confirmed a correlation
between the expression of these three genes and tumor stage, consistent with the observed
CNA (Figure S2). Finally, to evaluate the role of each single gene, we consulted the Kaplan–
Meier Plotter web server, selecting the serous histotype. Only TPGS2A showed a strong
correlation between expression and both DFS and OS in ovarian cancer patients (Figure 4B),
suggesting its pivotal role in ovarian cancer.
Genes 2024, 15, x FOR PEER REVIEW 9 of 14
Figure 4. (A) GEPIA2 correlation between ovarian cancer patients DFS or OS and 18q12.2 genes
(FHOD3, TPGS2, and KIAA1328, 3 signatures) expression (p-value < 0.05). Red line: high expression
group; blue line: low expression group. (B) KaplanMeier Ploer correlation between patients DFS
or OS and TPGS2 expression (p-value < 0.01). Red line: high expression group; black line: low ex-
pression group.
4. Discussion
Molecular biomarkers are defined as any measurable molecular indicator of risk dis-
ease or patient outcome. This category includes germline or somatic genetic variants, ep-
igenetic signatures, transcriptional changes, and proteomic signatures [25]. Despite the
vast number of investigations, the identication of novel biomarkers in ovarian cancer is
still an urgent need [26].
The principal defining features of high-grade serous ovarian carcinoma (HGSOC),
the most prevalent form of epithelial ovarian cancer, are copy number alterations and ge-
nomic rearrangements [27]. This makes genomic variants appealing as diagnostic or prog-
nostic biomarkers, as well as for their easy detection [13].
Cancer stem cells (CSCs) are a subpopulation of cancer cells that are capable of self-
renewal, differentiation, proliferation, and drug resistance. CSCs are also important in in-
vasiveness and metastatic capability of tumors. For these reasons, it is understandable that
a worse prognosis of the patient correlates with higher expression of the molecular signa-
tures related to CSCs [28].
In a previous study, we isolated and characterized the CSC subpopulation from
Caov3, Ovcar5, and Ovcar8 ovarian cancer cell lines. Caov3 and Ovcar8 are known to be
representatives of HGSOC [19,20,22,29]. Concerning the Ovcar5 cell line, despite its origin
Figure 4. (A) GEPIA2 correlation between ovarian cancer patients’ DFS or OS and 18q12.2 genes
(FHOD3,TPGS2, and KIAA1328, 3 signatures) expression (p-value < 0.05). Red line: high expression
group; blue line: low expression group. (B) Kaplan–Meier Plotter correlation between patients’ DFS
or OS and TPGS2 expression (p-value < 0.01). Red line: high expression group; black line: low
expression group.
Genes 2024,15, 1487 9 of 14
4. Discussion
Molecular biomarkers are defined as any measurable molecular indicator of risk
disease or patient outcome. This category includes germline or somatic genetic variants,
epigenetic signatures, transcriptional changes, and proteomic signatures [
25
]. Despite the
vast number of investigations, the identification of novel biomarkers in ovarian cancer is
still an urgent need [26].
The principal defining features of high-grade serous ovarian carcinoma (HGSOC), the
most prevalent form of epithelial ovarian cancer, are copy number alterations and genomic
rearrangements [
27
]. This makes genomic variants appealing as diagnostic or prognostic
biomarkers, as well as for their easy detection [13].
Cancer stem cells (CSCs) are a subpopulation of cancer cells that are capable of self-
renewal, differentiation, proliferation, and drug resistance. CSCs are also important in
invasiveness and metastatic capability of tumors. For these reasons, it is understandable
that a worse prognosis of the patient correlates with higher expression of the molecular
signatures related to CSCs [28].
In a previous study, we isolated and characterized the CSC subpopulation from
Caov3, Ovcar5, and Ovcar8 ovarian cancer cell lines. Caov3 and Ovcar8 are known to be
representatives of HGSOC [
19
,
20
,
22
,
29
]. Concerning the Ovcar5 cell line, despite its origin
still being debated, it is currently used as an HGSOC model [
21
,
22
], and for this reason, we
decided to utilize three cell lines for our experiment. Remarkably, we identified a potential
prognostic role of AhRR and PPP1R3C in serous ovarian cancer by bioinformatics analyses
of array-CGH data performed on these CSCs [
10
]. In this work, we started from copy
number losses shared by those CSC subpopulations in order to find chromosomal regions
or genes that may be informative about patients’ prognosis.
First, we looked at the enriched pathways and GO terms of the 1253 genes that were
found to be involved in copy number loss in all CSCs subpopulations. KEGG pathway
analysis revealed a significant decrease in the mRNA surveillance pathway, including
nonsense-mediated mRNA decay (NMD), nonstop mRNA decay (NSD), and no-go decay
(NGD). NMD recognizes and degrades transcripts with a premature translation-termination
codon, preventing the production of C-terminally truncated proteins that can have a delete-
rious effect in the cell [30]. Evidence has identified NMD as a key driver of tumorigenesis
in a tumor-specific manner. In some cancers, NMD is enhanced to degrade certain mR-
NAs, including those encoding tumor suppressors. Conversely, in other tumors, NMD is
inhibited, promoting the expression of oncoproteins or other proteins that support tumor
growth and progression [31].
An interesting gene in this pathway was the PPP2R2A gene (the only one located
at 8p21.1), which is deleted at high frequencies in luminal type B breast cancer and non-
small cell lung cancer, as well as being one of the most common breakpoints in prostate
cancer [
32
]. Loss of PPP2R2A inhibits homologous recombination DNA repair, suggesting
it as a potential marker for PARP inhibitor responses in clinics [33].
REACTOME pathway enrichment analysis showed loss of downregulation of SMAD2/
3:SMAD4 transcriptional activity. In the nucleus, the SMAD2/3:SMAD4 heterotrimer
complex acts as a transcriptional regulator. SMAD2, SMAD3, and SMAD4 are considered
to be key mediators of TGF-
β
signaling [
34
]. Ovarian tumors are significantly influenced by
the TGF-
β
pathway and SMAD proteins [
35
]. In particular, upregulation of this pathway
promotes the EMT process and enhances tumor cell resistance to paclitaxel [
36
]. Our results
confirmed the important role of this pathway in ovarian CSCs.
GO enrichment analyses showed a statistically significant downregulation of gene
silencing by miRNA in CSCs, together with a decrease of some GO terms related to mRNA
processing. The maintenance of CSCs’ stemness and malignancy depends on mRNA
modifications [
37
], and miRNA plays a significant role in this process. In fact, aberrant
expression of miRNA, often due to genetic modifications, is essential for the initiation and
progression of human cancers as they act as both tumor suppressors and oncogenes [38].
Genes 2024,15, 1487 10 of 14
Taken together, these results validated our CSCs models, underlined some CSC char-
acteristics, and confirmed the importance of copy number losses in tumorigenesis and
cancer progression.
Subsequently, we identified only three CN losses and the loss of whole chromosome X
shared by all spheroids and investigated a possible correlation with patients’ prognosis
suggested by their presence in all CSCs models. Loss of chromosome X was abundantly
reported in cancer as a potential mechanism of X-linked tumor suppressor gene inactivation,
so we focused our attention on the other three CN losses [39,40].
Loss of 4q34.3-q35.2 correlated with an increase in the fraction of genome altered, as
well as in the mutational count in these samples, suggesting an increased genomic instability.
Terminal 4q loss has been found in colorectal cancer as a marker of advanced stage [
41
], in
hepatoblastoma as a poor prognostic factor [
42
], and in intrahepatic cholangiocarcinoma
associated with a high histological grade [
43
]. Moreover, deletion of 4q34.3 predicted
early relapse after adjuvant chemotherapy in lung adenocarcinoma [
44
]. Frequent LOH
(loss of heterozygosity) in the 4q terminal region in hepatocellular carcinoma, head and
neck squamous cell carcinoma, and oral carcinoma has also been reported [
42
]. With
regard to ovarian cancer, a loss of 4q34.3 in mucinous and clear cell ovarian cancer cell
lines [
12
] and a potential correlation between 4q35.2 loss and chemoresistance [
45
] were
previously reported.
Loss of 8p21.2-p21.1 in ovarian cancer biopsies was associated with a higher mutational
count and increased TMB (nonsynonymous) together with a statistically significant increase
in arm-level CNAs, indicating a great genetic instability of these samples. Bioinformatics
analyses of the genes involved in 8p21.2-p21.1 CNA revealed a statistically significant
cluster in the positive regulation of the apoptotic process, suggesting that the loss of these
genes may lead to the loss of apoptosis.
Frequent deletion (23%) of 8p21.2 was identified in TCGA tumors and reported in
previous studies for ovarian cancer, particularly for serous histology and high-grade and
chemoresistant samples [
46
]. Another study identified loss on 13q32.1 and 8p21.1 as the
most reliable combination for detecting chemoresistant disease, with EXTL3 as a potential
gene linked to antineoplastic drug-resistance [45].
Kaveh et al. analyzed copy number data from breast, ovarian, endometrial, and
cervical cancers and identified 8p21.2 loss in cancers of the reproductive system, indicating
BNIP3L (a proapoptotic gene) and PPP2R2A as interesting tumor suppressor genes [
47
].
Our results support PPP2R2A’s role in ovarian CSCs also.
Moreover, we identified for the first time a statistically significant difference in 8p21.2-
p21.1 gain between early- and late-onset ovarian cancer (cut-off 63 years), suggesting a
potential favorable prognostic role of this gain, in accordance with data observed in samples
with loss.
As shown for loss of 4q34.3-q35.2 and 8p21.2-p21.1, 18q12.2-q23 loss correlated with
clinical parameters related to genomic instability, such as an increased aneuploidy score
and arm-level CNAs, as well as a higher fraction of altered genome. Intriguingly, a
statistically significant difference in disease-free survival of patients with or without loss
was found. Pathway enrichment analyses of differentially expressed proteins in the two
groups revealed a statistically significant increase in cysteine and methionine metabolism
in the 18q loss group (PSAT1 and LDHB proteins).
Phosphoserine aminotransferase 1 (PSAT1) catalyzes the second step of the serine-
glycine biosynthesis pathway, and its overexpression was reported in ovarian cancer [
48
],
lung adenocarcinoma [
49
], and breast cancer [
50
]. Lactate dehydrogenase (LDH) plays key
roles in cancer metabolism reprogramming [
51
]. LDHA directly catalyzes the conversion
of pyruvate to lactate; on the contrary, LDH-B converts lactate to pyruvate. Upregulation
of LDH-B in tumors has been reported and correlated with disease progression and poor
prognosis [5052]. These data could explain the reduced DFS of patients with 18q loss.
Additionally, a link between 18q12.2-q23 and the stage of the tumor was found; in
fact, a statistically significant increase of samples with CN loss in this region was found in
Genes 2024,15, 1487 11 of 14
stage IV. Interestingly, within the 18q12.2-q23 region, different sub-regions with different
percentages of alterations among stages were present. In particular, the percentage of CN
loss of the 18q12.2 region (containing FHOD3,TPGS2, and KIAA1328) was significantly
increased in stage IV samples with respect to all other samples. Genomic observation
was supported by mRNA data from the OncoDB web server that confirmed a correlation
between the expression of these three genes and the clinical stage of the tumor. Moreover,
a strong correlation between TPGS2A expression and both DFS and OS in ovarian cancer
patients could suggest for the first time its prognostic role in ovarian cancer.
5. Conclusions
In conclusion, we analyzed copy number losses shared by our previously isolated and
characterized ovarian CSC subpopulations. Pathway and gene ontology further validated
our CSC models, underlining some CSC characteristics and confirming the importance of
copy number losses in tumorigenesis and cancer progression.
Then, starting from these CN losses, we validated their potential prognostic relevance
by analyzing the TCGA cohort. Our analysis of copy number alterations in ovarian CSCs
revealed novel insights by identifying three specific copy number losses associated with
higher genetic instability and patients’ prognosis. The identified potential candidate genes
not only enhance our understanding of the role of numerical aberrations as biomarkers but
also suggest new avenues for targeted therapies. By elucidating the pathways influenced
by these genetic alterations, our findings could help the development of personalized
treatment strategies, ultimately aiming to improve patient outcomes and more effectively
manage cancer progression.
Supplementary Materials: The following supporting information can be downloaded at https://www.
mdpi.com/article/10.3390/genes15111487/s1. Table S1: KEGG pathway, REACTOME pathway,
and GO (BP: biological process, and MF: molecular function) enrichment analyses of 1253 genes
involved in copy number losses. Figure S1: Correlation between copy number losses and clinical
parameters. Table S2: 18q12.2-q23 loss in different tumor stages. Figure S2: OncoDB correlation
between expressions of FHOD3, TPGS2, and KIAA1328 and the clinical stage of a tumor.
Author Contributions: Conceptualization: D.C.; methodology: A.A., A.J. and D.C.; formal analysis:
A.A., A.J., S.R. and D.C.; investigation: A.A., A.J., S.R. and D.C.; writing—original draft preparation:
A.J. and D.C.; writing—review and editing: A.A., A.J., S.R., A.B., M.L. and D.C.; funding acquisition:
M.L. and D.C.; resources: A.B. and M.L.; supervision: D.C. All authors have read and agreed to the
published version of the manuscript.
Funding: This research was funded by the University of Milano-Bicocca [2021-ATE-0167 and 2023-
ATE-0402 to D.C. and 2023-ATE-0605 to M.L.] and Instand-NGS4P H2020 Project ID: 874719 [to
M.L.].
Institutional Review Board Statement: Not applicable.
Informed Consent Statement: Not applicable.
Data Availability Statement: The data that support the findings of this study are available from the
corresponding author [D.C.] upon reasonable request.
Conflicts of Interest: The authors declare no conflicts of interest.
References
1.
Taylor, A.M.; Shih, J.; Ha, G.; Gao, G.F.; Zhang, X.; Berger, A.C.; Schumacher, S.E.; Wang, C.; Hu, H.; Liu, J.; et al. Genomic and
Functional Approaches to Understanding Cancer Aneuploidy. Cancer Cell. 2018,33, 676–689.e3. [CrossRef] [PubMed]
2. Ben-David, U.; Amon, A. Context is everything: Aneuploidy in cancer. Nat. Rev. Genet. 2020,21, 44–62. [CrossRef] [PubMed]
3.
Pinto, N.; Mayfield, J.R.; Raca, G.; Applebaum, M.A.; Chlenski, A.; Sukhanova, M.; Bagatell, R.; Irwin, M.S.; Little, A.; Rawwas, J.;
et al. Segmental Chromosomal Aberrations in Localized Neuroblastoma Can be Detected in Formalin-Fixed Paraffin-Embedded
Tissue Samples and Are Associated with Recurrence. Pediatr. Blood Cancer 2016,63, 1019–1023. [CrossRef] [PubMed]
Genes 2024,15, 1487 12 of 14
4.
Janoueix-Lerosey, I.; Schleiermacher, G.; Michels, E.; Mosseri, V.; Ribeiro, A.; Lequin, D.; Vermeulen, J.; Couturier, J.; Peuchmaur,
M.; Valent, A.; et al. Overall genomic pattern is a predictor of outcome in neuroblastoma. J. Clin. Oncol. 2009,27, 1026–1033.
[CrossRef]
5.
Hieronymus, H.; Murali, R.; Tin, A.; Yadav, K.; Abida, W.; Moller, H.; Berney, D.; Scher, H.; Carver, B.; Scardino, P.; et al. Tumor
copy number alteration burden is a pan-cancer prognostic factor associated with recurrence and death. Elife 2018,7, e37294.
[CrossRef]
6.
King, L.; Flaus, A.; Holian, E.; Golden, A. Survival outcomes are associated with genomic instability in luminal breast cancers.
PLoS ONE 2021,16, e0245042. [CrossRef]
7.
Conconi, D.; Jemma, A.; Giambra, M.; Redaelli, S.; Croci, G.A.; Dalprà, L.; Lavitrano, M.; Bentivegna, A. Analysis of copy number
alterations in bladder cancer stem cells revealed a prognostic role of LRP1B. World J. Urol. 2022,40, 2267–2273. [CrossRef]
8.
Giambra, M.; Di Cristofori, A.; Conconi, D.; Marzorati, M.; Redaelli, S.; Zambuto, M.; Rocca, A.; Roumy, L.; Carrabba, G.;
Lavitrano, M.; et al. Insights into the Peritumoural Brain Zone of Glioblastoma: CDK4 and EXT2 May Be Potential Drivers of
Malignancy. Int. J. Mol. Sci. 2023,24, 2835. [CrossRef]
9.
Chen, C.H.; Lin, Y.J.; Lin, Y.Y.; Lin, C.H.; Feng, L.Y.; Chang, I.Y.; Wei, K.C.; Huang, C.Y. Glioblastoma Primary Cells Retain the
Most Copy Number Alterations That Predict Poor Survival in Glioma Patients. Front. Oncol. 2021,11, 621432. [CrossRef]
10.
Ardizzoia, A.; Jemma, A.; Redaelli, S.; Silva, M.; Bentivegna, A.; Lavitrano, M.; Conconi, D. AhRR and PPP1R3C: Potential
Prognostic Biomarkers for Serous Ovarian Cancer. Int. J. Mol. Sci. 2023,24, 11455. [CrossRef]
11.
Macintyre, G.; Goranova, T.E.; De Silva, D.; Ennis, D.; Piskorz, A.M.; Eldridge, M.; Sie, D.; Lewsley, L.A.; Hanif, A.; Wilson,
C.; et al. Copy number signatures and mutational processes in ovarian carcinoma. Nat. Genet. 2018,50, 1262–1270. [CrossRef]
[PubMed]
12.
Li, J.; Liang, H.; Xiao, W.; Wei, P.; Chen, H.; Chen, Z.; Yang, R.; Jiang, H.; Zhang, Y. Whole-exome mutational landscape and
molecular marker study in mucinous and clear cell ovarian cancer cell lines 3AO and ES2. BMC Cancer 2023,23, 321. [CrossRef]
[PubMed]
13.
Smith, P.; Bradley, T.; Gavarró, L.M.; Goranova, T.; Ennis, D.P.; Mirza, H.B.; De Silva, D.; Piskorz, A.M.; Sauer, C.M.; Al-Khalidi, S.;
et al. The copy number and mutational landscape of recurrent ovarian high-grade serous carcinoma. Nat. Commun. 2023,14, 4387.
[CrossRef] [PubMed]
14.
Walcher, L.; Kistenmacher, A.K.; Suo, H.; Kitte, R.; Dluczek, S.; Strauß, A.; Blaudszun, A.R.; Yevsa, T.; Fricke, S.; Kossatz-Boehlert,
U. Cancer Stem Cells-Origins and Biomarkers: Perspectives for Targeted Personalized Therapies. Front. Immunol. 2020,11, 1280.
[CrossRef]
15.
Liao, J.; Qian, F.; Tchabo, N.; Mhawech-Fauceglia, P.; Beck, A.; Qian, Z.; Wang, X.; Huss, W.J.; Lele, S.B.; Morrison, C.D.; et al.
Ovarian cancer spheroid cells with stem cell-like properties contribute to tumor generation, metastasis and chemotherapy
resistance through hypoxia-resistant metabolism. PLoS ONE 2014,9, e84941. [CrossRef]
16.
Geiger, T.; Cox, J.; Mann, M. Proteomic changes resulting from gene copy number variations in cancer cells. PLoS Genet. 2010,6,
e1001090. [CrossRef] [PubMed]
17.
Sherman, B.T.; Hao, M.; Qiu, J.; Jiao, X.; Baseler, M.W.; Lane, H.C.; Imamichi, T.; Chang, W. DAVID: A web server for functional
enrichment analysis and functional annotation of gene lists (2021 update). Nucleic Acids Res. 2022,50, W216–W221. [CrossRef]
18.
Silva, T.C.; Colaprico, A.; Olsen, C.; D
Angelo, F.; Bontempi, G.; Ceccarelli, M.; Noushmehr, H. TCGA Workflow: Analyze cancer
genomics and epigenomics data using Bioconductor packages. F1000Research 2016,5, 1542. [CrossRef]
19.
Khin, M.; Davis, L.J.; Lantvit, D.D.; Orjala, J.; Burdette, J.E. Aulosirazole Stimulates FOXO
3
a Nuclear Translocation to Regulate
Apoptosis and Cell-Cycle Progression in High-Grade Serous Ovarian Cancer (HGSOC) Cells. Mol. Pharmacol. 2024,106, 145–154.
[CrossRef]
20.
Ebott, J.; McAdams, J.; Kim, C.; Jansen, C.; Woodman, M.; De La Cruz, P.; Schrol, C.; Ribeiro, J.; James, N. Enhanced amphiregulin
exposure promotes modulation of the high grade serous ovarian cancer tumor immune microenvironment. Front. Pharmacol.
2024,15, 1375421. [CrossRef]
21.
Valdivia, A.; Cowan, M.; Cardenas, H.; Isac, A.M.; Zhao, G.; Huang, H.; Matei, D. E2F1 mediates competition, proliferation and
response to cisplatin in cohabitating resistant and sensitive ovarian cancer cells. Front. Oncol. 2024,14, 1304691. [CrossRef]
[PubMed]
22.
Mitra, A.K.; Davis, D.A.; Tomar, S.; Roy, L.; Gurler, H.; Xie, J.; Lantvit, D.D.; Cardenas, H.; Fang, F.; Liu, Y.; et al.
In vivo
tumor
growth of high-grade serous ovarian cancer cell lines. Gynecol. Oncol. 2015,138, 372–377. [CrossRef] [PubMed]
23.
Fogg, K.C.; Miller, A.E.; Li, Y.; Flanigan, W.; Walker, A.; O
Shea, A.; Kendziorski, C.; Kreeger, P.K. Ovarian cancer cells direct
monocyte differentiation through a non-canonical pathway. BMC Cancer 2020,20, 1008. [CrossRef] [PubMed]
24. Shoemaker, C.J.; Green, R. Translation drives mRNA quality control. Nat. Struct. Mol. Biol. 2012,19, 594–601. [CrossRef]
25. Sarhadi, V.K.; Armengol, G. Molecular Biomarkers in Cancer. Biomolecules 2022,12, 1021. [CrossRef]
26.
López-Portugués, C.; Montes-Bayón, M.; Díez, P. Biomarkers in Ovarian Cancer: Towards Personalized Medicine. Proteomes 2024,
12, 8. [CrossRef]
27.
Vázquez-García, I.; Uhlitz, F.; Ceglia, N.; Lim, J.L.P.; Wu, M.; Mohibullah, N.; Niyazov, J.; Ruiz, A.E.B.; Boehm, K.M.; Bojilova, V.;
et al. Ovarian cancer mutational processes drive site-specific immune evasion. Nature 2022,612, 778–786. [CrossRef]
Genes 2024,15, 1487 13 of 14
28.
Eun, K.; Ham, S.W.; Kim, H. Cancer stem cell heterogeneity: Origin and new perspectives on CSC targeting. BMB Rep. 2017,50,
117–125. [CrossRef] [PubMed]
29.
McCorkle, J.R.; Ahn, R.; Cao, C.D.; Hill, K.S.; Dietrich, C.S.; Kolesar, J.M. Antineoplastic Drug Synergy of Artesunate with
Navitoclax in Models of High-Grade Serous Ovarian Cancer. Cancers 2024,16, 1321. [CrossRef]
30.
Nogueira, G.; Fernandes, R.; García-Moreno, J.F.; Romão, L. Nonsense-mediated RNA decay and its bipolar function in cancer.
Mol. Cancer 2021,20, 72. [CrossRef]
31.
Nagar, P.; Islam, M.R.; Rahman, M.A. Nonsense-Mediated mRNA Decay as a Mediator of Tumorigenesis. Genes 2023,14, 357.
[CrossRef] [PubMed]
32.
Zhao, Z.; Kurimchak, A.; Nikonova, A.S.; Feiser, F.; Wasserman, J.S.; Fowle, H.; Varughese, T.; Connors, M.; Johnson, K.; Makhov,
P.; et al. PPP2R2A prostate cancer haploinsufficiency is associated with worse prognosis and a high vulnerability to B55
α
/PP2A
reconstitution that triggers centrosome destabilization. Oncogenesis 2019,8, 72. [CrossRef] [PubMed]
33.
Kalev, P.; Simicek, M.; Vazquez, I.; Munck, S.; Chen, L.; Soin, T.; Danda, N.; Chen, W.; Sablina, A. Loss of PPP2R2A inhibits
homologous recombination DNA repair and predicts tumor sensitivity to PARP inhibition. Cancer Res. 2012,72, 6414–6424.
[CrossRef] [PubMed]
34.
Sun, Y.; Ding, L.; Zhang, H.; Han, J.; Yang, X.; Yan, J.; Zhu, Y.; Li, J.; Song, H.; Ye, Q. Potentiation of Smad-mediated transcriptional
activation by the RNA-binding protein RBPMS. Nucleic Acids Res. 2006,34, 6314–6326. [CrossRef]
35.
Wang, Q.; Xiong, F.; Wu, G.; Wang, D.; Liu, W.; Chen, J.; Qi, Y.; Wang, B.; Chen, Y. SMAD Proteins in TGF-
β
Signalling Pathway in
Cancer: Regulatory Mechanisms and Clinical Applications. Diagnostics 2023,13, 2769. [CrossRef]
36.
Shi, Y.; Zhang, J.; Liu, M.; Huang, Y.; Yin, L. SMAD3 inducing the transcription of STYK1 to promote the EMT process and
improve the tolerance of ovarian carcinoma cells to paclitaxel. J. Cell. Biochem. 2019,120, 10796–10811. [CrossRef]
37.
Liang, W.; Lin, Z.; Du, C.; Qiu, D.; Zhang, Q. mRNA modification orchestrates cancer stem cell fate decisions. Mol. Cancer 2020,
19, 38. [CrossRef]
38.
Khan, A.Q.; Ahmed, E.I.; Elareer, N.R.; Junejo, K.; Steinhoff, M.; Uddin, S. Role of miRNA-Regulated Cancer Stem Cells in the
Pathogenesis of Human Malignancies. Cells 2019,8, 840. [CrossRef]
39.
Liu, R.; Kain, M.; Wang, L. Inactivation of X-linked tumor suppressor genes in human cancer. Future Oncol. 2012,8, 463–481.
[CrossRef]
40.
Dunford, A.; Weinstock, D.M.; Savova, V.; Schumacher, S.E.; Cleary, J.P.; Yoda, A.; Sullivan, T.J.; Hess, J.M.; Gimelbrant, A.A.;
Beroukhim, R.; et al. Tumor-suppressor genes that escape from X-inactivation contribute to cancer sex bias. Nat. Genet. 2017,49,
10–16. [CrossRef]
41.
Liang, J.W.; Shi, Z.Z.; Zhang, T.T.; Hao, J.J.; Wang, Z.; Wang, X.M.; Yang, H.; Wang, M.R.; Zhou, Z.X.; Zhang, Y. Analysis of
genomic aberrations associated with the clinicopathological parameters of rectal cancer by array-based comparative genomic
hybridization. Oncol. Rep. 2013,29, 1827–1834. [CrossRef] [PubMed]
42.
Arai, Y.; Honda, S.; Haruta, M.; Kasai, F.; Fujiwara, Y.; Ohshima, J.; Sasaki, F.; Nakagawara, A.; Horie, H.; Yamaoka, H.; et al.
Genome-wide analysis of allelic imbalances reveals 4q deletions as a poor prognostic factor and MDM4 amplification at 1q32.1 in
hepatoblastoma. Genes Chromosomes Cancer 2010,49, 596–609. [CrossRef] [PubMed]
43.
Huang, W.T.; Weng, S.W.; Wei, Y.C.; You, H.L.; Wang, J.T.; Eng, H.L. Genome-wide single nucleotide polymorphism array analysis
reveals recurrent genomic alterations associated with histopathologic features in intrahepatic cholangiocarcinoma. Int. J. Clin.
Exp. Pathol. 2014,7, 6841–6851.
44.
Han, X.; Tan, Q.; Yang, S.; Li, J.; Xu, J.; Hao, X.; Hu, X.; Xing, P.; Liu, Y.; Lin, L.; et al. Comprehensive Profiling of Gene Copy
Number Alterations Predicts Patient Prognosis in Resected Stages I-III Lung Adenocarcinoma. Front. Oncol. 2019,9, 556.
[CrossRef]
45.
Kim, S.W.; Kim, J.W.; Kim, Y.T.; Kim, J.H.; Kim, S.; Yoon, B.S.; Nam, E.J.; Kim, H.Y. Analysis of chromosomal changes in
serous ovarian carcinoma using high-resolution array comparative genomic hybridization: Potential predictive markers of
chemoresistant disease. Genes Chromosomes Cancer 2007,46, 1–9. [CrossRef]
46.
Reid, B.M.; Permuth, J.B.; Chen, Y.A.; Fridley, B.L.; Iversen, E.S.; Chen, Z.; Jim, H.; Vierkant, R.A.; Cunningham, J.M.; Barnholtz-
Sloan, J.S.; et al. Genome-wide Analysis of Common Copy Number Variation and Epithelial Ovarian Cancer Risk. Cancer
Epidemiol. Biomark. Prev. 2019,28, 1117–1126. [CrossRef] [PubMed]
47.
Kaveh, F.; Baumbusch, L.O.; Nebdal, D.; Børresen-Dale, A.L.; Lingjærde, O.C.; Edvardsen, H.; Kristensen, V.N.; Solvang, H.K. A
systematic comparison of copy number alterations in four types of female cancer. BMC Cancer 2016,16, 913. [CrossRef]
48.
Zhang, Y.; Li, J.; Dong, X.; Meng, D.; Zhi, X.; Yuan, L.; Yao, L. PSAT1 Regulated Oxidation-Reduction Balance Affects the Growth
and Prognosis of Epithelial Ovarian Cancer. OncoTargets Ther. 2020,13, 5443–5453. [CrossRef]
49.
Luo, M.Y.; Zhou, Y.; Gu, W.M.; Wang, C.; Shen, N.X.; Dong, J.K.; Lei, H.M.; Tang, Y.B.; Liang, Q.; Zou, J.H.; et al. Metabolic and
Nonmetabolic Functions of PSAT1 Coordinate Signaling Cascades to Confer EGFR Inhibitor Resistance and Drive Progression in
Lung Adenocarcinoma. Cancer Res. 2022,82, 3516–3531. [CrossRef]
50.
Gao, S.; Ge, A.; Xu, S.; You, Z.; Ning, S.; Zhao, Y.; Pang, D. PSAT1 is regulated by ATF4 and enhances cell proliferation via the
GSK3β/β-catenin/cyclin D1 signaling pathway in ER-negative breast cancer. J. Exp. Clin. Cancer Res. 2017,36, 179. [CrossRef]
Genes 2024,15, 1487 14 of 14
51.
Vlasiou, M.; Nicolaidou, V.; Papaneophytou, C. Targeting Lactate Dehydrogenase-B as a Strategy to Fight Cancer: Identification
of Potential Inhibitors by In Silico Analysis and In Vitro Screening. Pharmaceutics 2023,15, 2411. [CrossRef] [PubMed]
52.
Wang, R.; Li, J.; Zhang, C.; Guan, X.; Qin, B.; Jin, R.; Qin, L.; Xu, S.; Zhang, X.; Liu, R.; et al. Lactate Dehydrogenase B Is Required
for Pancreatic Cancer Cell Immortalization Through Activation of Telomerase Activity. Front. Oncol. 2022,12, 821620. [CrossRef]
[PubMed]
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual
author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to
people or property resulting from any ideas, methods, instructions or products referred to in the content.
ResearchGate has not been able to resolve any citations for this publication.
Article
Full-text available
High grade serous ovarian cancer (HGSOC) is a lethal gynecologic malignancy in which chemoresistant recurrence rates remain high. Furthermore, HGSOC patients have demonstrated overall low response rates to clinically available immunotherapies. Amphiregulin (AREG), a low affinity epidermal growth factor receptor ligand is known to be significantly upregulated in HGSOC patient tumors following neoadjuvant chemotherapy exposure. While much is known about AREG’s role in oncogenesis and classical immunity, it is function in tumor immunology has been comparatively understudied. Therefore, the objective of this present study was to elucidate how increased AREG exposure impacts the ovarian tumor immune microenvironment (OTIME). Using NanoString IO 360 and protein analysis, it was revealed that treatment with recombinant AREG led to prominent upregulation of genes associated with ovarian pathogenesis and immune evasion (CXCL8, CXCL1, CXCL2) along with increased STAT3 activation in HGSOC cells. In vitro co-culture assays consisting of HGSOC cells and peripheral blood mononuclear cells (PBMCs) stimulated with recombinant AREG (rAREG) led to significantly enhanced tumor cell viability. Moreover, PBMCs stimulated with rAREG exhibited significantly lower levels of IFNy and IL-2. In vivo rAREG treatment promoted significant reductions in circulating levels of IL-2 and IL-5. Intratumoral analysis of rAREG treated mice revealed a significant reduction in CD8⁺ T cells coupled with an upregulation of PD-L1. Finally, combinatorial treatment with an AREG neutralizing antibody and carboplatin led to a synergistic reduction of cell viability in HGSOC cell lines OVCAR8 and PEA2. Overall, this study demonstrates AREG’s ability to modulate cytotoxic responses within the OTIME and highlights its role as a novel HGSOC immune target.
Article
Full-text available
Simple Summary Modern ovarian cancer treatment has not substantially improved outcomes, and superior therapeutic strategies are needed. The aim of this study was to evaluate the efficacy of artesunate and navitoclax drug combination in ovarian cancer. We determined the combination of these two drugs was extraordinarily effective in multiple models of ovarian cancer, in vitro, inhibiting cancer cell proliferation more than expected based on single-agent activities. Unfortunately, we were unable to validate these findings using a mouse model of metastatic ovarian cancer. These data provide valuable information regarding the potential utility and challenges associated with the artesunate/navitoclax drug combination for ovarian cancer therapy. Abstract Artesunate belongs to a class of medications derived from the sweet wormwood plant (Artemisia annua) known as artemisinins. Artesunate has traditionally been used as a frontline treatment for severe malaria but has also demonstrated antineoplastic activity against various malignancies, including ovarian cancer. Data suggest that artesunate exacerbates cellular oxidative stress, triggering apoptosis. In the current study, we investigated the ability of navitoclax, an inhibitor of the antiapoptotic Bcl-2 protein family, to enhance artesunate efficacy in ovarian cancer cells. Artesunate and navitoclax both demonstrated antiproliferative effects on 2D and 3D ovarian cancer cell models as single agents. Upon combination of navitoclax with artesunate, antineoplastic drug synergy was also observed in each of the 2D cell lines and ovarian tumor organoid models tested. Further investigation of this drug combination using intraperitoneal CAOV3 xenograft models in BALB/scid mice showed that the artesunate/navitoclax doublet was superior to single-agent artesunate and vehicle control treatment. However, it did not outperform single-agent navitoclax. With optimization, this drug combination could provide a new therapeutic option for ovarian cancer and warrants further preclinical investigation.
Article
Full-text available
Ovarian cancer is one of the deadliest cancers in women. The lack of specific symptoms, especially at the initial stages of disease development, together with the malignancy heterogeneity, lower the life expectancy of patients. Aiming to improve survival rates, diagnostic and prognostic biomarkers are increasingly employed in clinics, providing gynecologists and oncologists with new tools to guide their treatment decisions. Despite the vast number of investigations, there is still an urgent need to discover more ovarian cancer subtype-specific markers which could further improve patient classification. To this end, high-throughput screening technologies, like mass spectrometry, are applied to deepen the tumoral cellular landscape and describe the malignant phenotypes. As for disease treatment, new targeted therapies, such as those based on PARP inhibitors, have shown great efficacy in destroying the tumoral cells. Likewise, drug-nanocarrier systems targeting the tumoral cells have exhibited promising results. In this narrative review, we summarize the latest achievements in the pursuit of biomarkers for ovarian cancer and recent anti-tumoral therapies.
Article
Full-text available
Background Tumor heterogeneity is one of the key factors leading to chemo-resistance relapse. It remains unknown how resistant cancer cells influence sensitive cells during cohabitation and growth within a heterogenous tumors. The goal of our study was to identify driving factors that mediate the interactions between resistant and sensitive cancer cells and to determine the effects of cohabitation on both phenotypes. Methods We used isogenic ovarian cancer (OC) cell lines pairs, sensitive and resistant to platinum: OVCAR5 vs. OVCAR5 CisR and PE01 vs. PE04, respectively, to perform long term direct culture and to study the phenotypical changes of the interaction of these cells. Results Long term direct co-culture of sensitive and resistant OC cells promoted proliferation (p < 0.001) of sensitive cells and increased the proportion of cells in the G1 and S cell cycle phase in both PE01 and OVCAR5 cells. Direct co-culture led to a decrease in the IC50 to platinum in the cisplatin-sensitive cells (5.92 µM to 2.79 µM for PE01, and from 2.05 µM to 1.51 µM for OVCAR5). RNAseq analysis of co-cultured cells showed enrichment of Cell Cycle Control, Cyclins and Cell Cycle Regulation pathways. The transcription factor E2F1 was predicted as the main effector responsible for the transcriptomic changes in sensitive cells. Western blot and qRT-PCR confirmed upregulation of E2F1 in co-cultured vs monoculture. Furthermore, an E2F1 inhibitor reverted the increase in proliferation rate induced by co-culture to baseline levels. Conclusion Our data suggest that long term cohabitation of chemo-sensitive and -resistant cancer cells drive sensitive cells to a higher proliferative state, more responsive to platinum. Our results reveal an unexpected effect caused by direct interactions between cancer cells with different proliferative rates and levels of platinum resistance, modelling competition between cells in heterogeneous tumors.
Article
Full-text available
Lactate dehydrogenase (LDH) is an enzyme that catalyzes the reversible conversion of lactate to pyruvate while reducing NAD+ to NADH (or oxidizing NADH to NAD+). Due to its central role in the Warburg effect, LDH-A isoform has been considered a promising target for treating several types of cancer. However, research on inhibitors targeting LDH-B isoform is still limited, despite the enzyme’s implication in the development of specific cancer types such as breast and lung cancer. This study aimed to identify small-molecule compounds that specifically inhibit LDH-B. Our in silico analysis identified eight commercially available compounds that may affect LDH-B activity. The best five candidates, namely tucatinib, capmatinib, moxidectin, rifampicin, and acetyldigoxin, were evaluated further in vitro. Our results revealed that two compounds, viz., tucatinib and capmatinib, currently used for treating breast and lung cancer, respectively, could also act as inhibitors of LDH-B. Both compounds inhibited LDH-B activity through an uncompetitive mechanism, as observed in in vitro experiments. Molecular dynamics studies further support these findings. Together, our results suggest that two known drugs currently being used to treat specific cancer types may have a dual effect and target more than one enzyme that facilitates the development of these types of cancers. Furthermore, the results of this study could be used as a new starting point for identifying more potent and specific LDH-B inhibitors.
Article
Full-text available
Suppressor of mother against decapentaplegic (SMAD) family proteins are central to one of the most versatile cytokine signalling pathways in metazoan biology, the transforming growth factor-β (TGF-β) pathway. The TGF-β pathway is widely known for its dual role in cancer progression as both an inhibitor of tumour cell growth and an inducer of tumour metastasis. This is mainly mediated through SMAD proteins and their cofactors or regulators. SMAD proteins act as transcription factors, regulating the transcription of a wide range of genes, and their rich post-translational modifications are influenced by a variety of regulators and cofactors. The complex role, mechanisms, and important functions of SMAD proteins in tumours are the hot topics in current oncology research. In this paper, we summarize the recent progress on the effects and mechanisms of SMAD proteins on tumour development, diagnosis, treatment and prognosis, and provide clues for subsequent research on SMAD proteins in tumours.
Article
Full-text available
The drivers of recurrence and resistance in ovarian high grade serous carcinoma remain unclear. We investigate the acquisition of resistance by collecting tumour biopsies from a cohort of 276 women with relapsed ovarian high grade serous carcinoma in the BriTROC-1 study. Panel sequencing shows close concordance between diagnosis and relapse, with only four discordant cases. There is also very strong concordance in copy number between diagnosis and relapse, with no significant difference in purity, ploidy or focal somatic copy number alterations, even when stratified by platinum sensitivity or prior chemotherapy lines. Copy number signatures are strongly correlated with immune cell infiltration, whilst diagnosis samples from patients with primary platinum resistance have increased rates of CCNE1 and KRAS amplification and copy number signature 1 exposure. Our data show that the ovarian high grade serous carcinoma genome is remarkably stable between diagnosis and relapse and acquired chemotherapy resistance does not select for common copy number drivers.
Article
Full-text available
The lack of effective screening and successful treatment contributes to high ovarian cancer mortality, making it the second most common cause of gynecologic cancer death. Development of chemoresistance in up to 75% of patients is the cause of a poor treatment response and reduced survival. Therefore, identifying potential and effective biomarkers for its diagnosis and prognosis is a strong critical need. Copy number alterations are frequent in cancer, and relevant for molecular tumor stratification and patients’ prognoses. In this study, array-CGH analysis was performed in three cell lines and derived cancer stem cells (CSCs) to identify genes potentially predictive for ovarian cancer patients’ prognoses. Bioinformatic analyses of genes involved in copy number gains revealed that AhRR and PPP1R3C expression negatively correlated with ovarian cancer patients’ overall and progression-free survival. These results, together with a significant association between AhRR and PPP1R3C expression and ovarian cancer stemness markers, suggested their potential role in CSCs. Furthermore, AhRR and PPP1R3C’s increased expression was maintained in some CSC subpopulations, reinforcing their potential role in ovarian cancer. In conclusion, we reported for the first time, to the best of our knowledge, a prognostic role of AhRR and PPP1R3C expression in serous ovarian cancer.
Article
Full-text available
Background Ovarian cancer is one of the most lethal cancers in women because it is often diagnosed at an advanced stage. The molecular markers investigated thus far have been unsatisfactory. Methods We performed whole-exome sequencing on the human ovarian cancer cell lines 3AO and ES2 and the normal ovarian epithelial cell line IOSE-80. Molecular markers of ovarian cancer were screened from shared mutation genes and copy number variation genes in the 6q21-qter region. Results We found that missense mutations were the most common mutations in the gene (93%). The MUC12, FLG and MUC16 genes were highly mutated in 3AO and ES2 cells. Copy number amplification occurred mainly in 4p16.1 and 11q14.3, and copy number deletions occurred in 4q34.3 and 18p11.21. A total of 23 hub genes were screened, of which 16 were closely related to the survival of ovarian cancer patients. The three genes CCDC170, THBS2 and COL14A1 are most significantly correlated with the survival and prognosis of ovarian cancer. In particular, the overall survival of ovarian cancer patients with high CCDC170 gene expression was significantly prolonged (P < 0.001). The expression of CCDC170 in normal tissues was significantly higher than that in ovarian cancer tissues (P < 0.05), and its expression was significantly decreased in advanced ovarian cancer. Western blotting and immunofluorescence assays also showed that the expression of CCDC170 in ovarian cancer cells was significantly lower than that in normal cells (P < 0.001, P < 0.01). Conclusions CCDC170 is expected to become a new diagnostic molecular target and prognostic indicator for ovarian cancer patients, which can provide new ideas for the design of antitumor drugs.
Article
Ovarian cancer, the fifth leading cause of cancer-related mortality in women, is the most lethal gynecological malignancy globally. Within various ovarian cancer subtypes, high-grade serous ovarian cancer is the most prevalent and there is frequent emergence of chemoresistance. Aulosirazole, an isothiazolonaphthoquinone alkaloid, isolated from the cyanobacterium Nostoc sp. UIC 10771, demonstrated cytotoxic activity against OVCAR3 cells (IC50 = 301 ± 80 nM). Using immunocytochemistry, OVCAR3 cells treated with aulosirazole demonstrated increased concentrations of phosphorylated protein kinase B and phosphorylated c-Jun N-terminal kinase with subsequent accumulation of forkhead box O3a (FOXO3a) in the nucleus. The combination of aulosirazole with protein kinase B inhibitors resulted in the most nuclear accumulation of FOXO3a aulosirazole-induced apoptosis based on cleavage of poly(ADP-ribose) polymerase, annexin V staining, and induction of caspase 3/7 activity in OVCAR3, OVCAR5, and OVCAR8. The expression of downstream targets of FOXO3a, including B-cell lymphoma 2 (BCL2) and p53-upregulator modulator of apoptosis, increased following aulosirazole treatment. Aulosirazole upregulated the FOXO3a target, cyclin-dependent kinase inhibitor 1, and increased cell-cycle arrest in the G0/G1 phase. The downregulation of FOXO3a by short hairpin RNA (shRNA) reduced the cytotoxicity after aulosirazole treatment by 3-fold IC50 (949 ± 16 nM) and eliminated its ability to regulate downstream targets of FOXO3a. These findings underscore FOXO3a as a critical mediator of aulosirazole-induced cytotoxicity. Additionally, aulosirazole was able to decrease migration and invasion while increasing cell death in 3D tumor spheroids. However, in vivo OVCAR8 tumor burden was not reduced by aulosirazole using an intraperitoneal tumor model. Given the mechanism of action of aulosirazole, this class of alkaloids represents promising lead compounds to develop treatments against FOXO3a-downregulated cancers. SIGNIFICANCE STATEMENT: Aulosirazole, an isothiazolonaphthoquinone alkaloid, exhibits potent cytotoxic effects against high-grade serous ovarian cancer by promoting forkhead box O3a (FOXO3a) nuclear accumulation and modulating downstream targets. These findings highlight the potential of aulosirazole as a promising therapeutic intervention for cancers characterized by FOXO3a downregulation.