ArticlePDF Available

Identification of a new gene signature for prognostic evaluation in cervical cancer: based on cuproptosis-associated angiogenesis and multi-omics analysis

Authors:

Abstract and Figures

Patients with recurrent or metastatic cervical cancer are in urgent need of novel prognosis assessment or treatment approaches. In this study, a novel prognostic gene signature was discovered by utilizing cuproptosis-related angiogenesis (CuRA) gene scores obtained through weighted gene co-expression network analysis (WGCNA) of The Cancer Genome Atlas (TCGA) and Gene Expression Omnibus (GEO) datasets. To enhance its reliability, the gene signature was refined by integrating supplementary clinical variables and subjected to cross-validation. Meanwhile, the activation of the VEGF pathway was inferred from an analysis of cell-to-cell communication, based on the expression of ligands and receptors in cell transcriptomic datasets. High-CuRA patients had less infiltration of CD8 + T cells and reduced expression of most of immune checkpoint genes, which indicated greater difficulty in immunotherapy. Lower IC50 values of imatinib, pazopanib, and sorafenib in the high-CuRA group revealed the potential value of these drugs. Finally, we verified an independent prognostic gene SFT2D1 was highly expressed in cervical cancer and positively correlated with the microvascular density. Knockdown of SFT2D1 significantly inhibited ability of the proliferation, migration, and invasive in cervical cancer cells. CuRA gene signature provided valuable insights into the prediction of prognosis and immune microenvironment of cervical cancer, which could help develop new strategies for individualized precision therapy for cervical cancer patients. Supplementary Information The online version contains supplementary material available at 10.1186/s12935-023-03189-x.
This content is subject to copyright. Terms and conditions apply.
RESEARCH Open Access
© The Author(s) 2024. Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use,
sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and
the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this
article are included in the article’s Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included
in the article’s Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will
need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by/4.0/. The
Creative Commons Public Domain Dedication waiver (http://creativecommons.org/publicdomain/zero/1.0/) applies to the data made available
in this article, unless otherwise stated in a credit line to the data.
Kang et al. Cancer Cell International (2024) 24:23
https://doi.org/10.1186/s12935-023-03189-x
Introduction
Cervical cancer ranks rst in the female reproductive
tract [1] and consists mainly of squamous and adeno-
carcinoma types of the cervix. Patients diagnosed with
early or locally advanced cervical cancer have achieved
some remission and have experienced high survival rates
with radical resection or concurrent radiotherapy [2, 3],
which have signicantly reduced mortality, especially in
developing countries and poor regions [4]. Nevertheless,
the prognosis and treatment outcomes for patients with
refractory cervical cancer, encompassing those aicted
with recurrent, persistent, or metastatic forms, remain
Cancer Cell International
Jiawen Kang and Jingwen Jiang have contributed equally to this
work and share rst authorship.
*Correspondence:
Yong Zhang
374794955@qq.com
Jie Tang
tangjie_73@163.com
Lesai Li
LLS0731@126.com
1Department of Gynecologic Oncology, School of Medicine, Hunan
Cancer Hospital/The Aliated Cancer Hospital of Xiangya, Central South
University, Changsha, Hunan, China
2Department of Clinical Medicine, Medical College of Hunan Normal
University, Changsha, Hunan, China
Abstract
Patients with recurrent or metastatic cervical cancer are in urgent need of novel prognosis assessment or treatment
approaches. In this study, a novel prognostic gene signature was discovered by utilizing cuproptosis-related
angiogenesis (CuRA) gene scores obtained through weighted gene co-expression network analysis (WGCNA) of
The Cancer Genome Atlas (TCGA) and Gene Expression Omnibus (GEO) datasets. To enhance its reliability, the
gene signature was rened by integrating supplementary clinical variables and subjected to cross-validation.
Meanwhile, the activation of the VEGF pathway was inferred from an analysis of cell-to-cell communication, based
on the expression of ligands and receptors in cell transcriptomic datasets. High-CuRA patients had less inltration
of CD8 + T cells and reduced expression of most of immune checkpoint genes, which indicated greater diculty
in immunotherapy. Lower IC50 values of imatinib, pazopanib, and sorafenib in the high-CuRA group revealed the
potential value of these drugs. Finally, we veried an independent prognostic geneSFT2D1was highly expressed
in cervical cancer and positively correlated with the microvascular density. Knockdown ofSFT2D1signicantly
inhibited ability of the proliferation, migration, and invasive in cervical cancer cells. CuRA gene signature provided
valuable insights into the prediction of prognosis and immune microenvironment of cervical cancer, which could
help develop new strategies for individualized precision therapy for cervical cancer patients.
Keywords Cervical cancer, Cuproptosis, Angiogenesis, Prognosis, Immune inltration, Cell communication
Identication of a new gene signature
for prognostic evaluation in cervical
cancer: based on cuproptosis-associated
angiogenesis and multi-omics analysis
JiawenKang1,2†, JingwenJiang2†, XiaoqingXiang2, YongZhang2*, JieTang1* and LesaiLi1*
Content courtesy of Springer Nature, terms of use apply. Rights reserved.
Page 2 of 15
Kang et al. Cancer Cell International (2024) 24:23
disheartening [57]. e implementation of novel scien-
tic methodologies and an enhanced comprehension of
tumor pathogenesis are poised to augment our under-
standing of cervical cancer mechanisms and ultimately
ameliorate the prognosis for individuals grappling with
refractory cervical cancer. It is necessary to explore new
ways to improve the prognosis of patients with refractory
cervical cancer.
Tumor angiogenesis, inammatory inltration of the
tumor microenvironment, and programmed cell death
processes have been identied as contributing factors
to tumor metastasis [8]. Tumor cells exhibit elevated
secretion of pro-angiogenic factors, which stimulate the
development of heterogeneous and immature neovascu-
larization. is heteromorphic neovascularization often
leads to a hypoxic microenvironment caused by inade-
quate perfusion, thereby favoring the survival and growth
of more aggressive tumor cells [9]. Simultaneously, the
presence of pro-angiogenic factors within the tumor
microenvironment facilitates the process of angiogenesis
and immunosuppression [10]. Consequently, angiogen-
esis fosters the tumor’s ability to evade the immune sys-
tem and engenders drug resistance. Tumor angiogenesis
stands as a contributing factor to recurrence, prompting
the clinical utilization of anti-angiogenic drugs in the
management of advanced or recurrent cervical cancer,
resulting in notable enhancements in survival rates [11].
However, the limited applicability of targeting mature
stable vessels [12] and the presence of various treatment-
related side eects have necessitated the exploration of
novel therapeutic approaches [13]. Notably, the remark-
able eectiveness of immunotherapy in cervical cancer
has highlighted the signicance of targeting angiogen-
esis in the tumor microenvironment for immunothera-
peutic interventions [14, 15]. Consequently, for patients
with metastatic, persistent, and recurrent cervical cancer
who exhibit PD-L1 positivity, the combination of pabli-
zumab and chemotherapy, with or without bevacizumab,
has emerged as the preferred rst-line treatment option
[6]. Investigating modications in the immune microen-
vironment and immune checkpoint genes within tumors
experiencing varying angiogenic states can provide valu-
able insights into the development of precise combina-
tions of vascular targeting therapy and immunotherapy.
Recent ndings indicate that several well-established
regulators of programmed cell death play a role in pro-
moting angiogenesis [8, 1620]. Additionally, cupropto-
sis, a distinct form of programmed cell death, is primarily
characterized by the excessive accumulation of intracel-
lular copper, leading to cell death [21]. It has been shown
increased intratumor copper concentrations promote
tumor growth and invasion as well as treatment resis-
tance [22]. Serum copper concentrations have been
found to exhibit a correlation with tumor progression
and morbidity [23]. Additionally, cuproptosis, a recently
identied mode of cell death, has been reported to play
a role in tumor growth, angiogenesis, and tumor metas-
tasis [24, 25]. Studies have demonstrated that copper
facilitates tumor angiogenesis by activating various
angiogenic factors, such as basic broblast growth factor
(bFGF) and vascular endothelial growth factor (VEGF)
[26]. Moreover, copper is implicated in signal transduc-
tion processes within endothelial cells, thereby inuenc-
ing angiogenesis [23]. In summary, copper assumes an
indispensable function in the advancement of tumors
as a trace element crucial for the proliferation of cancer
cells and the formation of blood vessels within tumors.
Further exploration of the correlation between angiogen-
esis and cuproptosis is imperative, as it holds potential
for novel treatment approaches [19, 27]. e association
between angiogenesis and cuproptosis in cervical cancer
has yet to be investigated, thus necessitating a meticu-
lous and proactive investigation employing innovative
methodologies.
e emergence of precision oncology and the integra-
tion of big data have facilitated the utilization of bulk
RNA sequencing to uncover the mean gene expression
in tissues, thereby enabling exploration into the realm
of cognitive dierential gene expression [28]. Further-
more, the progression of technological tools has allowed
for the implementation of single cell sequencing, which
has proven instrumental in discerning dierential gene
expression among cells and investigating intricate cell
populations [2931]. is technique has signicantly
contributed to the elds of tumor diagnosis, targeted
therapy, and prognosis prediction [32, 33]. e analy-
sis of intercellular communication in cell populations
aids in the elucidation of communication and signaling
mechanisms among diverse cells [34]. Additionally, cor-
relation analysis of receptor-ligand pairs enables a deeper
comprehension of cellular functionality and regulatory
networks. In this study, we have developed a prognos-
tic model for the CuRA gene using both single-cell RNA
sequencing and bulk RNA sequencing. is model holds
promise for the development of innovative prognostic
prediction models and treatment approaches for individ-
uals with cervical cancer.
Materials and methods
Data acquisition
e cervical cancer patient dataset (the TCGA-CESC
cohort) was obtained from the Cancer Genome Atlas
as the training group (TCGA, https://portal.gdc.cancer.
gov/). For external validation, we used 55 cervical cancer
patients from the GSE52903 dataset in Gene Expression
Omnibus (GEO, https://www.ncbi.nlm.nih.gov/geo/)
databases. We also included single-cell sequencing datas-
ets GSE168652 in cervical cancer using “Seurat” package
Content courtesy of Springer Nature, terms of use apply. Rights reserved.
Page 3 of 15
Kang et al. Cancer Cell International (2024) 24:23
[35] for calculating genetic correlations to score cells and
patients. e scores of CuRA gene-sets were calculated
by applying the “Percentage FeatureSet” function. e
GeneCards website (https://www.genecards.org/) was
used to obtain 1245 angiogenesis-related genes using the
“angiogenesis” keyword with a correlation > 1. R pack-
age “limma” [36] was used to obtain cuproptosis-related
angiogenesis (CuRA) genes. Genes with dierential
expression in normal and cervical tissues were obtained
from the GEPIA website (http://gepia.cancer-pku.cn/).
WGCNA
Single-sample gene set enrichment analysis (ssGSEA),
which estimated the relative enrichment of a particular
gene set in each sample by comparing the gene expression
data of that sample with that set. We performed weighted
gene co-expression network analysis (WGCNA) using
the “WGCNA” package [37]. e threshold of clustering
cut tree was set to 210, and the minimum threshold was
set to 80. We merged the modules with threshold < 0.5.
en, we performed analysis and included all genes in
modules of CuRA phenotypes with P < 0.05 for subse-
quent analysis.
Construction of the CuRA model
We congured the alpha parameter of the elastic network
to 0.5 and computed the errors for ridge regression, lasso
regression and elastic network regression. e model
regression was constructed using the “glmnet” package
[38]. e “timeROC” package [39] and “survivalROC”
package [40] were performed to plot ROC curves for
survival outcomes at dierent time points. Nomogram
based on logistic regression and Cox regression was con-
structed using the “rms” package [41].
Immune inltration analysis
“IOBR” package [42] was used to immune inltration
analysis. Six algorithm CIBERSORT, EPIC, MCP, XCELL,
TIMER, QUANTISEQ was performed to compare the
dierences between the high and low-CuRA groups. Dif-
ferences in the expression of immune checkpoint genes
were also compared. e relevant mutation data were
obtained from Cbioportal (https://www.cbioportal.org/
datasets). e “maftools” package [43] was performed for
visualization.
Cell communication analysis
We performed cell communication analysis using the R
package “CellChat” [34]. We ltered out cell communi-
cation with less than 10 cells and obtained the cell com-
munication relationship between each cell. We inferred
cell-to-cell communication at the pathway level, deduced
pathway-level interaction networks, and obtained the
interaction relationship between receptor-ligand pairs
and cell communication.
GSEA
e GSEA software (version 3.0) was downloaded from
the GSEA (http://software.broadinstitute.org/gsea/
index.jsp) website, divided the samples into high and
low expression groups based on the expression levels
ofSFT2D1. APvalue of < 0.05 and an FDR value of < 0.25
were considered statistically signicant. e correspond-
ing data was listed in Additional le 1: Table S5.
Drug sensitivity analysis
We searched the GDSC database to predict drug sensi-
tivity by comparing the IC50 of drugs among dierent
groups based on the CuRA scores. By analyzing in the
Drug Signatures Database (DSigDB, http://tanlab.ucden-
ver.edu/DSigDB), we listed corresponding small mol-
ecule drugs of relevant modeling genes (Additional le 1:
Table S4).
Cell culture
e ECT1/E6E7 cell line (ATCC: CRL-2614™), the
SiHa cell line (ATCC: HTB-35™), the CaSki cell line
(ATCC: CRM-CRL-1550™) were obtained from Ameri-
can Type Culture Collection (ATCC). Ect1/E6E7 cells,
SiHa cells were cultured in DMEM medium (Procell,
Wuhan,China) containing 10% fetal bovine serum (Pro-
cell, Wuhan, China), 100 U/mL penicillin and 100 µg/mL
streptomycin, and incubated at 37 °C under conditions
of 5% CO2. CaSki cells were cultured with the same con-
ditions in 1640 medium (Procell, Wuhan, China). ese
cells were transfected with synthetic small interfering
RNAs (GenePharma, Shanghai, China) by Lipo8000™
Transfection Reagent (Beyotime, Shanghai, China), and
the siRNA sequences targeting SFT2D1 gene are pro-
vided in the Additional le 1: Table S6.
Real-time uorescence quantitative PCR
We extracted total RNA of cells using TRIZOL reagent
(Vazyme, China), followed by adding chloroform for cen-
trifugation. e supernatant was collected and mixed
with isopropanol. e RNA pellet was washed with 75%
ethanol and air-dried. e purity of RNA was measured
using a spectrophotometer (ermo Fisher Scientic,
USA). e cDNA was synthesized using reverse tran-
scription reagent (TransGen, China) for uorescence
quantication (RT-qPCR).
Immunohistochemistry
We collected normal cervical tissue and cervical can-
cer tissue at the Hunan Provincial Cancer Hospital for
immunohistochemical staining, and it has been reviewed
and approved by the Ethics Committee of Hunan
Content courtesy of Springer Nature, terms of use apply. Rights reserved.
Page 4 of 15
Kang et al. Cancer Cell International (2024) 24:23
Cancer Hospital. After dewaxing of sections, heat antigen
retrieval was performed. e primary antibodies SFT2D1
(Immunoway, USA, 1:100) and CD31 (ZenBio, China,
1:100) were incubated overnight at 4 °C. e secondary
antibodies were incubated for 20 min using the PV-9000
kit (ZSGB-BIO, China). DAB reagent (ZSGB-BIO, China)
was used for antibody staining, with brown-yellow indi-
cating positive signal areas. Cell nuclei were stained blue
with hematoxylin (Servicebio, China). Images were cap-
tured using microscope (Zeiss, Germany) and analyzed
using Image J software (1.53, USA).
Western blotting
We added a mixture of cell lysis buer (Servicebio,
China) and protease inhibitor PMSF to the cells. e
sample was then denatured by adding SDS-loading buer
and subjected to electrophoresis. e PVDF membrane
(Millipore, USA) was wet-transferred at a constant cur-
rent. After blocking with skim milk at room tempera-
ture for 2 h, the primary antibody SFT2D1 (Immunoway,
USA, 1:1000) was incubated at 4 overnight. e sec-
ondary antibody (bioworld, USA, 1:10000) was incubated
at room temperature for 1 h, followed by detection with a
developing solution.
CCK-8 assay
After transfection, the appropriate amounts of resus-
pended cervical cancer cells in the logarithmic phase of
growth were added in 96-well plates with trypsin diges-
tion down and set up 5 sub-wells per group (NC, si-
SFT2D1). When the cells were adhered to the wall, the
solution in Cell Counting Kit-8 (CCK-8, APE, USA) was
added after replacing the fresh medium, and the absor-
bance value was measured at 450 nm after incubation
with the cells for 2 h at 37 in an MicroplateReader
Instrument (Biotek, USA). First data were grouped into
the 0 h group. And the readings of 0 h, 24 h, 48 h, 72 and
96 h were recorded to calculate the proliferative capacity
of the cells.
Wound scratch experiment
After transfection, resuspended cells were added in
6-well plates by trypsin digestion. e cells incubated
under conditions of constant temperature and constant
CO2, a straight line was drawn vertically in the center of
the 6-well plate with the pipette tip, and then the width of
the straight line was photographed and recorded under
the microscope. Replace the medium with serum-free
medium to continue incubation for 24-48 h, and then
take pictures with the microscope to record the growth
of cells. Cell migration rate = (0 h scratch width - scratch
width after incubation)/0 h scratch width × 100%, which
was analyzed by ImageJ software (1.53, USA) to calculate
the migration ability of cells.
Transwell migration and invasion assay
e chambers were hydrated with serum-free DMEM
medium for 30 min. After aspirating the medium a cell
suspension mixed with appropriate amount of serum-
free medium was added to the upper chamber, and the
lower chamber was incubated with medium containing
10% serum for 24 h. After xation in methanol and stain-
ing with crystal violet, the cells that did not pass through
the upper chamber were wiped away, and the cells that
passed through the lower chamber were observed and
counted under the microscope, and the migration abil-
ity of the cells was judged according to the number of
cells. e cells were observed and counted in the lower
chamber under the microscope. e matrix gel was pur-
chased from Corning (USA). e gel was spread on the
upper chamber surface, and after 2 h, the gel was allowed
to solidify and then hydrated with medium, and the same
procedure was followed to determine the invasion ability
of the cells according to the number of cells.
Statistical analyses
We used R software (version 4.2) and GraphPad prism
(version 8.3.0) for relative analyses and drawings.
T-test was performed to analysis dierences between
two groups. ANOVA was used to analysis dierences
between three or more groups.P < 0.05 was considered as
statistically dierent.
Results
Flow chart
e ow chart was shown in Fig. 1.
Identication of phenotype -related dierent CuRA genes
by WGCNA
19 cuproptosis genes was shown in Additional le 1:
Table S1. en we downloaded angiogenesis-associated
genes with correlation > 1 from the GENECARD website.
Based on the gene expression of TCGA-CESC patients,
as a screening condition of |cor|>0.3 andP < 0.05, nally
533 CuRA genes were included. At the same time, we
performed univariate cox analysis to obtain 66 prognos-
tic CuRA genes (Additional le 1: Figure S1) from 533
genes. To explore genes that are dierentially expressed
between the normal cervix and cervical cancer, we
downloaded 6057 dierential genes from the GEPIA
website(|log2FC|>1). We took the intersection of 6057
dierential genes with the 66 prognostic CuRA genes
(Fig. 2A) above and nally obtained 20 CuRA genes with
signicant dierences (Fig. 2B, Additional le 1: Table
S2). Based on the scores of 20 CuRA gene-sets, each cell
was divided into high-CuRA and low-CuRA cells groups
according to the median value in GSE168652 dataset (Fig.
2C, D). In the TCGA-CESC cohort, we quantied and
visualized the level of immune inltration in dierent
Content courtesy of Springer Nature, terms of use apply. Rights reserved.
Page 5 of 15
Kang et al. Cancer Cell International (2024) 24:23
patients by ssGSEA (Fig. 2E). Meanwhile, we performed
CuRA scores for each patient by the ssGSEA algorithm,
then plotted circle plots to see the dierences in scoring
for each patient (Fig. 2F). WGCNA was performed to
obtain phenotype-related modules for CuRA genes (Fig.
2G). We included all genes (P < 0.05) in non-grey mod-
ules (e grey module contained genes that couldn’t be
classied as any module): pink, brown, magenta, purple,
and yellow modules for the follow-up study (Fig. 2H).
Construction of a CuRA prognostic model
We obtained CuRA intersection genes by intersecting the
dierent genes of high-CuRA and low-CuRA cells groups
of GSE168652 with the WGCNA phenotype-related
modular genes of patients. We congured the alpha
parameter of the elastic network to 0.5 and computed the
errors for primary methods. e results revealed that the
error for ridge regression is 3.055991, for lasso regression
it is 0.0002452, and for elastic network it is 0.0002547035.
After comparing the errors of the regression methods, we
opted for lasso regression to construct the model (Addi-
tional le 1: Figure S2). en a prognostic model was
constructed based on 10 CuRA genes by lasso regres-
sion according to optimal lambda value (Additional le
1: Figure S3A, B). CuRA modeling scores = 0.001007113
*IRF6+ 0.001993273*THBD+ 0.007235823*EFEMP2+ 4.
45E-04*SNX9+ 0.012465781*PCDH18+ 7.23E-05*MFA
P4+ 0.005812479*ADAM9+ 0.004983918*EHBP1+ 0.05
5328306*AVL9+ 0.00770984*SFT2D1. We included the
GSE52903 as validation set. en we analyzed the dier-
ences between TCGA patients (Additional le 1: Figure
S3C) and GSE52903 patients (Additional le 1: Figure
S3D) according to CuRA modeling scores by PCA. Fur-
ther, we assessed ecacy of the model to predict progno-
sis. Heatmaps and point chart of risk scores were drawn
showing dierences in expression of CuRA model genes
Fig. 1 Flow chart of the full text. CuRA, cuproptosis-related angiogenesis gene. DEGs, dierential genes. WGCNA, weighted correlation network analysis.
ROC, receiver operating characteristic
Content courtesy of Springer Nature, terms of use apply. Rights reserved.
Page 6 of 15
Kang et al. Cancer Cell International (2024) 24:23
in TCGA patients (Fig. 3A) and GEO patients (Fig. 3D.
In TCGA dataset (Fig. 3B) and GEO dataset (Fig. 3E),
patients in the high-CuRA group had signicantly lower
survival than those in the low-CuRA group. In the ROC
curves, the 1, 2, 3, and 5-year AUC values were 0.653,
0.759, 0.748, and 0.799 for TCGA patients, respectively
(Fig. 3C). e AUC values for 2, 3, and 5-year survival for
GEO patients were 0.653, 0.646, and 0.604, respectively
(Fig. 3F). Results indicated the model had better predic-
tive eect on prognosis in both TCGA and GEO datasets.
Meanwhile, we included clinical data of TCGA patients
and then combined T1-2 patients into early stage and
T3-4 stage patients into late stage of T-stage for univari-
ate COX analysis, and we found CuRA modeling scores,
N_stage were risk factors (Additional le 1: Figure S3E).
Further, we performed a multivariate COX analysis and
the results showed CuRA modeling scores, N_stage, and
T_stage were independent risk factors (Additional le 1:
Figure S3F).
Immune inltration landscape and mutational landscape
Based on the modeling gene scores, we explored the dif-
ferences in immune inltration and tumor mutations
of high-CuRA and low-CuRA groups. We introduced
Fig. 2 Selection of CuRA phenotype-related genes by WGCNA. (A) Venn diagram of intersection of 6057 dierential genes with 66 prognostic genes. (B)
Histogram of 20 CuRA genes validated at the GEPIA website. (C) Scores of CuRA genes in GSE168652, with the darker the purple, the higher the scores.
(D) Grouping into high-CuRA and low-CuRA groups, red indicating high-CuRA group, blue indicating low-CuRA group. (E) Quantication of immune
inltration levels by ssGSEA. (F) Circle plot of CuRA genes scores of patients. (G) Waterfall plot of WGCNA. (H) Heatmap of phenotype-related modules
Content courtesy of Springer Nature, terms of use apply. Rights reserved.
Page 7 of 15
Kang et al. Cancer Cell International (2024) 24:23
six algorithms to perform a comprehensive analysis of
immune cell inltration in two groups (Additional le
1: Figure S5). e six algorithms showed inltration of
the overall immune cells was signicantly lower in the
high-CuRA group than in the low-CuRA group. Among
them, it showed a signicant dierence between the two
groups by xCell algorithm. Next, we compared the 10
types of major immune cells, and found CD8 + T cells
were signicantly reduced in patients in the high-CuRA
group (Fig. 4A). Also, we analyzed the expression of com-
mon immune checkpoint genes, and we found the clini-
cally common immune checkpoint genesCD274(PD-L1)
and CTLA4 did not dier signicantly between two
groups (Fig. 4B). erefore, we speculate patients clas-
sied into high-CuRA and low-CuRA groups may not
dier in treatment benet by applyingPD-L1inhibitors
orCTLA4 inhibitors. Also, the results showed most of
the immune checkpoint genes were signicantly less
expressed in the high-CuRA group than in the low-
CuRA group. We also found IL10RB, KDR, TGFB1,
and TGFBR1 genes were signicantly more expressed
in the high-CuRA group than in the low-CuRA group.
Results suggested patients in the high-CuRA group may
get better therapeutic outcomes by using inhibitors tar-
geting these 4 genes. Next, we analyzed the mutation
landscape in both groups (Fig. 4C, D), and found the top
3 genes with the highest mutation frequencies were PI
K3CA(31%), TTN(31%),SYNE1(18%) in 127 patients of
the high-CuRA group, while in the low-CuRA group, the
top 3 genes with the highest mutation frequencies were
TTN(33%),PIK3CA(27%),KMT2C(22%) in 113 patients.
We foundSYNE1showed a higher mutation frequency
(23 cases) in the high-CuRA group and a lower muta-
tion frequency (11 cases) in the low-CuRA group with
signicant dierence between the two groups (odds ratio
(OR) = 0.412) (Additional le 1: Figure S6). Also, other
genes such as RELN, SPATA31D1, TCOF1 had higher
mutations in the high-CuRA group.
Analysis of clinical characterization and construction of
nomogram
We analyzed prognosis with dierent clinical character-
istics, and constructed a nomogram based on the CuRA
model. Results showed the N_stage and CuRA model-
ing scores contributed signicantly to the model. e
predicted mortality of the patient was 0.571, 0.98, and
0.997 at 1, 3, and 5-years, respectively (Fig. 5A), and
its odds ratio of status was 8.19 (Fig. 5B). ROC curves
were performed to predict the accuracy of nomogram.
AUC values at 1, 3, and 5-years were 0.71, 0.78, and
0.83, respectively, which indicated the nomogram had
good predictive accuracy (Fig. 5C). Finally, we plotted
Fig. 3 Construction and verication of a CuRA prognostic model. Heatmap and point chart of TCGA patients (A) and GEO patients (D). Survival curve plot
of TCGA patients (B) and GEO patients (E). ROC curve of TCGA patients (C) and GEO patients (F)
Content courtesy of Springer Nature, terms of use apply. Rights reserved.
Page 8 of 15
Kang et al. Cancer Cell International (2024) 24:23
the DCA decision curves, and results showed the model
had a strong predictive eect on the survival rate at 1, 3,
and5-years (Fig. 5D).
Drug sensitivity prediction
To explore the dierent drugs that may be eective
against treating the high- and low-CuRA groups, we used
GDSC database by predicting the IC50 to determine the
dierences in drug sensitivity between the high- and
low-CuRA groups. Results showed the IC50 of Imatinib,
Pazopanib, and Sorafenib was signicantly lower for the
high-CuRA group than for the low-CuRA group, suggest-
ing they may have better ecacy when applied with the
high-CuRA patient group (Fig. 6A–C). Similarly, for the
low-CuRA group, the application of AMG.706, CEP.701,
Sunitinib, ABT.888 (Veliparib), AZD.2281 (Olaparib),
and MS.275 (Entinostat) may lead to better therapeutic
remission (Fig. 6D–I).
Cell communication analysis
We performed cell communication analysis by single-cell
sequencing dataset GSE168652 from GEO. We grouped
the cells into 25 clusters and divided the cell clusters into
8 types based on annotation, which are: endothelial cells,
FDX1 + tumor/epithelial cells, broblasts, lymphocytes,
macrophages, smooth muscle cells, tumor/epithelial cells
(other types), and VEGFA + tumor/epithelial cells (Fig.
7A). We observed the expression and localization of 10
modeling genes in GSE168652 (Additional le 1: Figure
S4). SinceFDX1was a representative gene for cupropto-
sis, we labeled FDX1-positive or VEGFA-positive tumor/
epithelial cells here in the hope of exploring the relation-
ship between cuproptosis and angiogenesis-associated
tumor/epithelial cells in intercellular communication.
Multidirectional cell communication was discovered in
each cell subpopulation (Fig. 7B). en we identied the
cell-extrinsic communication patterns. We analyzed the
signaling pathways of both incoming and outgoing sig-
nals in the samples. Our results revealed the main outgo-
ing signals of FDX1 + tumor/epithelial cells were PDGF,
WNT, CD46, MHC-1, MIF, and MK pathways, while the
primary incoming signals were IFN-II and other path-
ways. As for VEGFA + tumor/epithelial cells, the major
outgoing signals were WNT, EGF, and VEGF, while the
main incoming signals involved numerous signaling
pathways, including COLLAGEN (Fig. 7C). Specically,
Fig. 4 Immune inltration and tumor mutation analysis. (A) Dierences in inltration of 10 types of immune cells between two CuRA groups. (B) Dif-
ferences in expression of immune checkpoints gene between the two groups. (C) Tumor mutation characteristics in the high-CuRA group. (D) Tumor
mutation characteristics in the low-CuRA group
Content courtesy of Springer Nature, terms of use apply. Rights reserved.
Page 9 of 15
Kang et al. Cancer Cell International (2024) 24:23
we focused on the involvement of VEGF pathway in cell
communication as the key pathway of angiogenesis. We
observed higher expression of the key gene VEGFA in the
VEGFA + tumors/epithelial cells, tumor/epithelial cells,
and FDX1 + tumors/epithelial cells in the VEGF signaling
pathway (Fig. 7D). In signal transduction, by calculating
the network centrality indices for each cell population,
we found VEGFA + tumor/epithelial cells were the domi-
nant signaler in the intercellular communication net-
work, endothelial cells were the main receivers and
inuencer, and FDX1 + tumors/epithelial cells also played
an important role in inuencer (Fig. 7E). In the visual cir-
cular and hierarchical plots of the VEGF signaling path-
way (Additional le 1: Figure S9), our analysis revealed
VEGFA + tumor/epithelial cells and FDX1 + tumor/epi-
thelial cells had the most signicant eect on endothelial
cells. In addition, we analyzed relevant receptor-ligand
pairs in FDX1 + or VEGFA + tumors/epithelial cells com-
municating with other cells respectively (Additional le
1: Figure S10).
Critical functional role of SFT2D1 in cervical cancer
We selected prognosis-related model genes
among 10 model genes (Additional le 1: Figure
S7).ADAM9,EHBP1, andSFT2D1gene were shown sig-
nicantly aecting the survival of patients. Meanwhile,
we also identied SFT2D1 as an independent risk fac-
tor by multivariate analysis along with clinical features
(Additional le 1: Table S3). en we performed GSEA
of SFT2D1, and results showed SFT2D1 was mainly
involved in the regulation of autophagy, glycosaminogly-
can degradation, RNA degradation, riboavin metabo-
lism, mTOR signaling pathway. We then investigate the
eect of SFT2D1 on immune microenvironment. e
total scores, stromal scores and immune scores were
signicantly lower in the high-SFT2D1 group than in
the low-SFT2D1group, and the immune related scores
were negatively correlated with SFT2D1 expression
(Additional le 1: Figure S8). Meanwhile, we performed
immunohistochemical analysis of SFT2D1 and the neo-
vascularization marker CD31 in paran sections of cer-
vical cancer patients. e results showed both SFT2D1
and CD31 were expressed up-regulated in cervical can-
cer tissues (Fig. 8A). Correlation analysis also showed
Fig. 5 Construction and validation of nomogram. (A) Cox regression to construct nomogram. (B) Logistic regression to construct nomogram. (C) Curves
of ROC for 1, 3, and 5-years. (D) DCA decision curves for 1, 3, and 5-years
Content courtesy of Springer Nature, terms of use apply. Rights reserved.
Page 10 of 15
Kang et al. Cancer Cell International (2024) 24:23
positive correlation in SFT2D1 and CD31 (Fig. 8B). en
we performed RT-qPCR to verify SFT2D1 was highly
expressed in cervical cancer cell lines compared to nor-
mal cervical cell lines ECT1/E6E7 (Fig. 8C). Meanwhile,
western blotting showed that SFT2D1 was upregulated
in SiHa, CaSki cervical cell lines (Fig. 8D). Since the role
ofSFT2D1in cervical cancer has not yet been explored,
we authenticated the eect ofSFT2D1on the function of
SiHa and CaSki cells by in vitro experiments. RT-qRCR
showed that four siRNAs signicantly suppressed the
expression ofSFT2D1in transfected SiHa and CaSki cells
(Fig. 8E, F). We selected the two siRNAs with the high-
est knockdown eciency among them: si-SFT2D1-2,
si-SFT2D1-3 for subsequent experiments. CCK-8 analy-
sis showed that knockdown of SFT2D1 signicantly
inhibited the proliferative ability of SiHa and CaSki cells
(Fig. 8G, H). Wound scratch assay showed that knock-
down ofSFT2D1signicantly inhibited the migration of
cervical cancer cells (Fig. 8I, J). Transwell assay showed
that knockdown of SFT2D1 signicantly inhibited the
migratory and invasive abilities of SiHa and CaSki cells
(Fig. 8K, L).
Fig. 6 Drug sensitivity analysis of the high and low-CuRA group.(A-I)The IC50 of Imatinib, Pazopanib, and Sorafenib was lower in the high-CuRA group
than for the low-CuRA group contrary to AMG.706, CEP.701, Sunitinib, ABT.888, AZD.2281, and MS.275. The vertical coordinates displayed as drug names,
demonstration of drugs with statistical signicance
Content courtesy of Springer Nature, terms of use apply. Rights reserved.
Page 11 of 15
Kang et al. Cancer Cell International (2024) 24:23
Discussion
In this study, we developed and validated a prognostic
model based on CuRA genes in cervical cancer. We also
analyzed clinical characteristics and the immune micro-
environment between the high and low-CuRA patient
groups. Based on the modeling scores, we analyzed drug
sensitivity of the high or low-CuRA patient group to pro-
vide guidance on drug administration.SFT2D1, as a key
gene involved in the progression of cervical cancer, it was
associated with the cuproptosis-dependent angiogenesis
pathway.
Cuproptosis and tumor angiogenesis are closely linked
in the tumor microenvironment. CuRA genes may help
explain the potential link between cuproptosis and angio-
genesis, which could improve the prognosis of cervi-
cal cancer patients. We constructed and validated our
model based on CuRA signatures using patient data from
TCGA and GEO. Several web tools enable us to extract
prognostic variable characteristics from multi-omics data
by selecting clinical variables or subgroup variables (lasso,
elastic network regularization, and network regularized
high-dimensional Cox regression) [44]. is implies the
necessity of choosing the optimal regression method for
subsequent studies. After comparing the errors of these
three regression methods, we observed that lasso regres-
sion minimized the error. Consequently, we have opted
for the lasso regression method to construct the CuRA
model. e AUC values for 1, 2, 3, and 5-year survival
in TCGA patients were 0.653, 0.759, 0.748, and 0.799,
respectively, while the AUC values for 2, 3, and 5-year
survival in GEO patients were 0.653, 0.646, and 0.604,
respectively. e shorter survival time of patients in the
high CuRA group may indicate that tumor cells pro-
mote tumor progression through cuproptosis-associated
angiogenesis. In the analysis of clinical characteristics,
CuRA modeling scores, N_stage, and T_stage were inde-
pendent risk factors, suggesting that modeling scores
Fig. 7 Analysis of cellular communication network. (A) Cell annotation of 8 types of cells. (B) Number of interactions and interaction weights of samples
in GSE168652. (C) Schematic diagram of the incoming and outgoing signals of samples. (D) Visualization of the expression of key genes in the VEGF signal-
ing pathway in 8 types of cells. (E) Visualization of cells involved as senders, receivers, mediators and inuencers in the VEGF pathway
Content courtesy of Springer Nature, terms of use apply. Rights reserved.
Page 12 of 15
Kang et al. Cancer Cell International (2024) 24:23
could independently contribute to cervical cancer pro-
gression as a risk factor. Our CuRA model is represented
by 10 genes involved in programmed death-related path-
ways, membrane vesicle transport, and tumorigenesis
and progression. Among them, SFT2 Domain Containing
1 (SFT2D1) is involved in protein and vesicle-mediated
translocation and is also associated with poor survival in
patients with high-risk neuroblastoma [45]. GSEA path-
way analysis results suggest thatSFT2D1plays an impor-
tant role in tumor-related pathways and is associated
with the invasion and progression of cervical cancer. e
immune microenvironment was scored forSFT2D1, and
patients with highSFT2D1expression had a higher CuRA
score and a worse prognosis, showing a more aggressive
immunosuppressive phenotype. We veriedSFT2D1was
signicantly upregulated in cervical cancer cells by west-
ern blotting, RT-qPCR, and immunohistochemistry.
erefore,SFT2D1, a CuRA modeling gene, may serve as
a marker gene and provide a new reference for the treat-
ment of cervical cancer patients.
Antitumor strategies targeting angiogenesis have been
used in the clinical management of patients with meta-
static or recurrent cervical cancer. However, improve-
ments in overall survival (OS) and progression-free
survival (PFS) times for patients are still limited. Com-
bining immune checkpoint inhibitors (ICIs) with
vascular targeting therapy has demonstrated synergis-
tic sensitization in the treatment of various tumors,
Fig. 8 SFT2D1was involved in the progression of cervical cancer. (A) SFT2D1 and CD31 were highly expressed in cervical cancer by IHC. (B) Correlation
analysis between SFT2D1 and CD31. (C) Validation ofSFT2D1expression by RT-qPCR. (D) Validation of SFT2D1 expression by western blotting. Four siRNAs
suppressed the expression ofSFT2D1in SiHa (E)and CaSki (F) cells by RT-qPCR. CCK-8 assay in in SiHa (G) and CaSki (H) cells. Wound scratch assay in SiHa
(I) and CaSki (J) cells. Transwell assay in SiHa (K) and CaSki (L) cells
Content courtesy of Springer Nature, terms of use apply. Rights reserved.
Page 13 of 15
Kang et al. Cancer Cell International (2024) 24:23
including hepatocellular carcinoma [46, 47], Non-small
cell lung cancer [48], gastric cancer [49]. In cervical
cancer, a phase III randomized controlled trial showed
increased overall survival in patients with recurrent or
metastatic cervical cancer after treatment with pabli-
zumab combined with chemotherapy and bevacizumab
[50]. erefore, researching the dierences in immune
microenvironment in cervical cancer patients with dier-
ent CuRA scores can help promote the precise combina-
tion of immunotherapy and vascular targeting therapy,
and enable personalized treatment selection of immune
checkpoint inhibitors for patients with CuRA-related
cervical cancer. Results showed that the group of patients
with high-CuRA scores exhibited immunosuppression,
while patients with low-CuRA scores may have a more
signicant therapeutic eect with immune agents target-
ing CD8 + T cells compared to patients in the high-CuRA
scores group. Patients with high-CuRA scores had lower
expression in most immune checkpoint genes. However,
a minority of immune checkpoint genes presented high
expression in the high-CuRA group, which suggests that
treatment with anti-IL10RB, anti-KDR, anti-TGFB1, and
anti-TGFBR1 may be considered to improve prognosis of
patients in the high-CuRA group.
Cuproptosis plays an important role in tumor cell pro-
liferation and angiogenesis. Cell-cell communication
analysis based on single-cell sequencing helps to reveal
the tumor immune microenvironment and changes
in the tumor itself [51]. Previous studies have shown
thatFDX1, as a key gene in cuproptosis, is involved in the
progression of hepatocellular carcinoma [52] and glioma
[53] as well as tumor immunity and drug sensitivity [54].
To investigate the relationship between angiogenesis and
cuproptosis in cervical cancer, we identied two types of
cells, FDX1 + tumor/epithelial cells and VEGFA + tumor/
epithelial cells, and conducted cell-cell communica-
tion analysis. We found that the genesVEGFAandPGF,
which promote angiogenesis, are highly expressed in
these two types of cells. Meanwhile, both types of cells
are sent as signals to endothelial cells, indicating that
they can aect the process of transendothelial migra-
tion and promote angiogenesis. In addition, both types
of cells can transmit cell signals to macrophages to aect
the macrophage migration inhibitory factor (MIF) path-
way. Previous studies have shown that MIF is involved in
multiple immune processes and mediates immune escape
leading to tumor metastasis [5558]. Our results suggest
that VEGFA + tumor/epithelial cells and FDX1 + tumor/
epithelial cells may play irreplaceable roles in the tumor
immune microenvironment.
We conducted drug sensitivity analysis for dierent
CuRA groups of patients, which may lay the foundation
for personalized treatment. e GDSC provides informa-
tion on the sensitivity of tumor cell lines to drugs, with
a smaller IC50 indicating greater sensitivity of the bio-
logical system to the compound. Our analysis showed
that among small molecule tyrosine kinase inhibitors,
imatinib, pazopanib, and sorafenib may provide bet-
ter ecacy for patients with high CuRA scores, while
AMG.706, CEP.701, and sunitinib may provide better
ecacy for patients with lower CuRA scores. Receptor
tyrosine kinase inhibitors have signicant anti-angiogenic
eects and have made great progress in the treatment of
gynecological tumors [59]. For example, pazopanib has
improved PFS and OS in patients with advanced or recur-
rent cervical cancer, while AMG.706 (motesanib) inhibits
angiogenesis in recurrent ovarian cancer and CEP.701
(lestaurtinib) inhibits the growth of cervical cancer cells
[6062]. Studies have shown dierences in IC50 values
of drugs such as pazopanib, sorafenib, sunitinib, and
imatinib among dierent CuRA subgroups of clear cell
renal cell carcinoma, bladder cancer, and triple-negative
breast cancer [6365]. Our study also suggested that in
the high CuRA score group, IC50 values of PARP inhibi-
tors ABT.888 (Veliparib), AZD.2281 (Olaparib), and
MS.275 (Entinostat) were higher, indicating that these
drugs were eective bythe cuproptosis-dependent angio-
genesis pathway ese studies supported our results of
drug sensitivity analysis and suggested that research on
anti-cuproptosis-related angiogenesis targeted drugs may
bring new treatment ideas and expand the application of
drugs in the current dilemma.
In conclusion, we have conducted a novel cuproptosis-
related angiogenesis (CuRA) gene signature using single-
cell RNA sequencing and bulk RNA sequencing data,
which provides signicant predictive value for patients
with cervical cancer. Of course, there are limitations to
our results, and future relevant mechanisms need to be
further validated in in vivo and in vitro experiments.
Supplementary Information
The online version contains supplementary material available at https://doi.
org/10.1186/s12935-023-03189-x.
Supplementary Material 1:Figure S1: Forest plot of 66 CuRA prognostic
genes by univariate COX analysis. Figure S2: Comparison of errors in ridge
regression, lasso regression and elastic network regression and compre-
hensive comparison of the errors in three regression algorithms.Figure
S3: Construction and verication of a CuRA prognostic model. Figure
S4: Localization and validation of 10 modeling genes in the GSE168652
dataset. Figure S5: Immune inltration landscape in high and low CuRA-
groups calculated by CIBERSORT, EPIC, MCP, Quanti-seq, TIMER, xCell
algorithms. Figure S6: Dierences in mutation frequency between high
and low CuRA groups. Figure S7: Survival analysis of 10 modeling genes.
Figure S8: Assessment of regulatory pathways and immune microen-
vironment of SFT2D1. Figure S9: Visual circular and hierarchical plots
showing cellular communication in the VEGF pathway. Figure S10: Cell
Communication Analysis
Supplementary Material 2:Table S1: Genelist of Cuproptosis-Related
Gene.Table S2: Genelist of Cuproptosis-Related Angiogenesis Gene
(CuRA).Table S3: Univariate Analysis and Multivariate Analysis of SFT2D1
and Clinical Characteristics.Table S4: Prediction of small molecule drugs
Content courtesy of Springer Nature, terms of use apply. Rights reserved.
Page 14 of 15
Kang et al. Cancer Cell International (2024) 24:23
by modeling genes.Table S5: GSEA to assess SFT2D1-related path-
ways.Table S6: Oligonucleotides used in research
Acknowledgements
Not applicable.
Author contributions
Conceptualization, Yong Zhang and Lesai Li; Data curation, Jiawen
Kang; Investigation, Jingwen Jiang; Methodology, Yong Zhang; Project
administration, Jie Tang; Validation, Xiaoqing Xiang; Writing – original draft,
Jiawen Kang; Writing – review & editing, Lesai Li.
Funding
This research was funded by Natural Science Foundation of Hunan Province
(Grant. 2022JJ30415), Hunan Cancer Hospital Climb Plan (ZX2020004-3).
Data availability
The data and materials in the current study are available under the permission
of author.
Declarations
Ethics approval and consent to participate
The study was conducted in accordance with the Declaration of Helsinki, and
approved by the Ethics Committee of Hunan Cancer Hospital (No. SBQLL-
2021-289). All patients gave informed consent.
Consent for publication
All authors of this paper consent for publishing the manuscript and gures in
the journal.
Competing interests
The authors declare no competing interests.
Received: 30 September 2023 / Accepted: 23 December 2023
References
1. Sung H, Ferlay J, Siegel RL, Laversanne M, Soerjomataram I, Jemal A, Bray
F. Global Cancer statistics 2020: GLOBOCAN estimates of incidence and
Mortality Worldwide for 36 cancers in 185 countries. CA Cancer J Clin.
2021;71(3):209–49.
2. Aviki EM, Chen L, Dessources K, Leitao MM Jr., Wright JD. Impact of hospital
volume on surgical management and outcomes for early-stage Cervical
cancer. Gynecol Oncol. 2020;157(2):508–13.
3. Abu-Rustum NR, Yashar CM, Bean S, Bradley K, Campos SM, Chon HS, Chu C,
Cohn D, Crispens MA, Damast S, et al. NCCN guidelines insights: Cervical Can-
cer, Version 1.2020. J Natl Compr Cancer Network: JNCCN. 2020;18(6):660–6.
4. Salehiniya H, Momenimovahed Z, Allahqoli L, Momenimovahed S, Alkatout
I. Factors related to Cervical cancer screening among Asian women. Eur Rev
Med Pharmacol Sci. 2021;25(19):6109–22.
5. Adiga D, Eswaran S, Pandey D, Sharan K, Kabekkodu SP. Molecular landscape
of recurrent Cervical cancer. Crit Rev Oncol/Hematol. 2021;157:103178.
6. Mutlu L, Tymon-Rosario J, Harold J, Menderes G. Targeted treatment options
for the management of metastatic/persistent and recurrent Cervical cancer.
Expert Rev Anticancer Ther. 2022;22(6):633–45.
7. Li H, Wu X, Cheng X. Advances in diagnosis and treatment of metastatic
Cervical cancer. J Gynecologic Oncol. 2016;27(4):e43.
8. Su Z, Yang Z, Xu Y, Chen Y, Yu Q. Apoptosis, autophagy, necroptosis, and
cancer Metastasis. Mol Cancer. 2015;14:48.
9. Viallard C, Larrivée B. Tumor angiogenesis and vascular normalization: alterna-
tive therapeutic targets. Angiogenesis. 2017;20(4):409–26.
10. Albini A, Bruno A, Noonan DM, Mortara L. Contribution to Tumor Angiogen-
esis from Innate Immune cells within the Tumor Microenvironment: implica-
tions for Immunotherapy. Front Immunol. 2018;9:527.
11. Tewari KS, Sill MW, Long HJ 3rd, Penson RT, Huang H, Ramondetta LM,
Landrum LM, Oaknin A, Reid TJ, Leitao MM, et al. Improved survival with
bevacizumab in advanced Cervical cancer. N Engl J Med. 2014;370(8):734–43.
12. Carmeliet P, Jain RK. Principles and mechanisms of vessel normaliza-
tion for cancer and other angiogenic Diseases. Nat Rev Drug Discovery.
2011;10(6):417–27.
13. Alameddine RS, Yakan AS, Skouri H, Mukherji D, Temraz S, Shamseddine A.
Cardiac and vascular toxicities of angiogenesis inhibitors: the other side of
the coin. Crit Rev Oncol/Hematol. 2015;96(2):195–205.
14. Chitsike L, Duerksen-Hughes P. The potential of Immune Checkpoint Block-
ade in Cervical Cancer: can combinatorial regimens maximize response? A
review of the literature. Curr Treat Options Oncol. 2020;21(12):95.
15. Mortara L, Benest AV, Derosa L, Chouaib S, Ribatti D. Editorial: the intricate
innate immune-cancer cell relationship in the context of Tumor angio-
genesis, immunity and microbiota: the angiogenic switch in the Tumor
microenvironment as a key target for immunotherapies. Front Immunol.
2022;13:1045074.
16. Gong C, Bauvy C, Tonelli G, Yue W, Deloménie C, Nicolas V, Zhu Y, Domergue
V, Marin-Esteban V, Tharinger H, et al. Beclin 1 and autophagy are required
for the tumorigenicity of Breast cancer stem-like/progenitor cells. Oncogene.
2013;32(18):2261–72. 2272e.2261 – 2211.
17. Tisch N, Freire-Valls A, Yerbes R, Paredes I, La Porta S, Wang X, Martín-Pérez
R, Castro L, Wong WW, Coultas L, et al. Caspase-8 modulates physiological
and pathological angiogenesis during retina development. J Clin Investig.
2019;129(12):5092–107.
18. Zhang F, Li Y, Tang Z, Kumar A, Lee C, Zhang L, Zhu C, Klotzsche-von Ameln
A, Wang B, Gao Z, et al. Proliferative and survival eects of PUMA promote
angiogenesis. Cell Rep. 2012;2(5):1272–85.
19. Tisch N, Ruiz de Almodóvar C. Contribution of cell death signaling to blood
vessel formation. Cell Mol Life Sci. 2021;78(7):3247–64.
20. Yang L, Joseph S, Sun T, Homann J, Thevissen S, Oermanns S, Strilic B. TAK1
regulates endothelial cell necroptosis and Tumor Metastasis. Cell Death Dier.
2019;26(10):1987–97.
21. Tsvetkov P, Coy S, Petrova B, Dreishpoon M, Verma A, Abdusamad M,
Rossen J, Joesch-Cohen L, Humeidi R, Spangler RD, et al. Copper induces
cell death by targeting lipoylated TCA cycle proteins. Sci (New York NY).
2022;375(6586):1254–61.
22. Lelièvre P, Sancey L, Coll JL, Deniaud A, Busser B. The multifaceted roles of
copper in Cancer: a Trace Metal element with Dysregulated Metabolism, but
also a target or a bullet for Therapy. Cancers 2020, 12(12).
23. Das A, Ash D, Fouda AY, Sudhahar V, Kim YM, Hou Y, Hudson FZ, Stans-
eld BK, Caldwell RB, McMenamin M, et al. Cysteine oxidation of copper
transporter CTR1 drives VEGFR2 signalling and angiogenesis. Nat Cell Biol.
2022;24(1):35–50.
24. Chi H, Peng G, Wang R, Yang F, Xie X, Zhang J, Xu K, Gu T, Yang X, Tian G.
Cuprotosis Programmed-Cell-Death-Related lncRNA Signature Predicts
Prognosis and Immune Landscape in PAAD Patients. Cells 2022, 11(21).
25. Ruiz LM, Libedinsky A, Elorza AA. Role of copper on mitochondrial function
and metabolism. Front Mol Biosci. 2021;8:711227.
26. Li Y. Copper homeostasis: emerging target for cancer treatment. IUBMB Life.
2020;72(9):1900–8.
27. Naito H, Iba T, Wakabayashi T, Tai-Nagara I, Suehiro JI, Jia W, Eino D, Sakimoto
S, Muramatsu F, Kidoya H, et al. TAK1 prevents endothelial apoptosis and
maintains Vascular Integrity. Dev Cell. 2019;48(2):151–166e157.
28. Li X, Wang CY. From bulk, single-cell to spatial RNA sequencing. Int J Oral Sci.
2021;13(1):36.
29. Hwang B, Lee JH, Bang D. Single-cell RNA sequencing technologies and
bioinformatics pipelines. Exp Mol Med. 2018;50(8):1–14.
30. Sun D, Guan X, Moran AE, Wu LY, Qian DZ, Schedin P, Dai MS, Danilov AV,
Alumkal JJ, Adey AC, et al. Identifying phenotype-associated subpopula-
tions by integrating bulk and single-cell sequencing data. Nat Biotechnol.
2022;40(4):527–38.
31. Li X, Sun Z, Peng G, Xiao Y, Guo J, Wu B, Li X, Zhou W, Li J, Li Z, et al. Single-cell
RNA sequencing reveals a pro-invasive cancer-associated broblast sub-
group associated with poor clinical outcomes in patients with gastric cancer.
Theranostics. 2022;12(2):620–38.
32. Lei Y, Tang R, Xu J, Wang W, Zhang B, Liu J, Yu X, Shi S. Applications of single-
cell sequencing in cancer research: progress and perspectives. J Hematol
Oncol. 2021;14(1):91.
33. Kim C, Gao R, Sei E, Brandt R, Hartman J, Hatschek T, Crosetto N, Foukakis
T, Navin NE. Chemoresistance Evolution in Triple-negative Breast Cancer
delineated by single-cell sequencing. Cell. 2018;173(4):879–893e813.
34. Jin S, Guerrero-Juarez CF, Zhang L, Chang I, Ramos R, Kuan CH, Myung P,
Plikus MV, Nie Q. Inference and analysis of cell-cell communication using
CellChat. Nat Commun. 2021;12(1):1088.
Content courtesy of Springer Nature, terms of use apply. Rights reserved.
Page 15 of 15
Kang et al. Cancer Cell International (2024) 24:23
35. Mangiola S, Doyle MA, Papenfuss AT. Interfacing Seurat with the R tidy
universe. Bioinf (Oxford England). 2021;37(22):4100–7.
36. Ritchie ME, Phipson B, Wu D, Hu Y, Law CW, Shi W, Smyth GK. Limma powers
dierential expression analyses for RNA-sequencing and microarray studies.
Nucleic Acids Res. 2015;43(7):e47.
37. Langfelder P, Horvath S. WGCNA: an R package for weighted correlation
network analysis. BMC Bioinformatics. 2008;9:559.
38. Engebretsen S, Bohlin J. Statistical predictions with glmnet. Clin Epigenetics.
2019;11(1):123.
39. Wang QW, Lin WW, Zhu YJ. Comprehensive analysis of a TNF family based-sig-
nature in diuse gliomas with regard to prognosis and immune signicance.
Cell Communication and Signaling: CCS. 2022;20(1):6.
40. Huang R, Chen Z, Li W, Fan C, Liu J. Immune system–associated genes
increase malignant progression and can be used to predict clinical outcome
in patients with hepatocellular carcinoma. Int J Oncol. 2020;56(5):1199–211.
41. Sun D, Zhu Y, Zhao H, Bian T, Li T, Liu K, Feng L, Li H, Hou H. Loss of ARID1A
expression promotes lung adenocarcinoma Metastasis and predicts a poor
prognosis. Cell Oncol (Dordrecht). 2021;44(5):1019–34.
42. Zeng D, Ye Z, Shen R, Yu G, Wu J, Xiong Y, Zhou R, Qiu W, Huang N, Sun L, et al.
IOBR: Multi-omics Immuno-Oncology Biological Research to Decode Tumor
Microenvironment and signatures. Front Immunol. 2021;12:687975.
43. Mayakonda A, Lin DC, Assenov Y, Plass C, Koeer HP. Maftools: ecient
and comprehensive analysis of somatic variants in cancer. Genome Res.
2018;28(11):1747–56.
44. Pak K, Oh SO, Goh TS, Heo HJ, Han ME, Jeong DC, Lee CS, Sun H, Kang J, Choi
S, et al. A User-Friendly, web-based Integrative Tool (ESurv) for Survival Analy-
sis: Development and Validation Study. J Med Internet Res. 2020;22(5):e16084.
45. Ognibene M, Morini M, Garaventa A, Podestà M, Pezzolo A. Identication of a
minimal region of loss on chromosome 6q27 associated with poor survival of
high-risk neuroblastoma patients. Cancer Biol Ther. 2020;21(5):391–9.
46. Finn RS, Qin S, Ikeda M, Galle PR, Ducreux M, Kim TY, Kudo M, Breder V, Merle
P, Kaseb AO, et al. Atezolizumab plus Bevacizumab in Unresectable Hepato-
cellular Carcinoma. N Engl J Med. 2020;382(20):1894–905.
47. Ren Z, Xu J, Bai Y, Xu A, Cang S, Du C, Li Q, Lu Y, Chen Y, Guo Y, et al. Sintilimab
plus a bevacizumab biosimilar (IBI305) versus sorafenib in unresectable
hepatocellular carcinoma (ORIENT-32): a randomised, open-label, phase 2–3
study. Lancet Oncol. 2021;22(7):977–90.
48. Reckamp KL, Redman MW, Dragnev KH, Minichiello K, Villaruz LC, Faller B,
Al Baghdadi T, Hines S, Everhart L, Highleyman L, et al. Phase II random-
ized study of Ramucirumab and Pembrolizumab Versus Standard of Care
in Advanced Non-small-cell Lung Cancer previously treated with immuno-
therapy-Lung-MAP S1800A. J Clin Oncology: Ocial J Am Soc Clin Oncol.
2022;40(21):2295–306.
49. Song X, Qi W, Guo J, Sun L, Ding A, Zhao G, Li H, Qiu W, Lv J. Immune check-
point inhibitor combination therapy for gastric cancer: Research progress.
Oncol Lett. 2020;20(4):46.
50. Schmidt MW, Battista MJ, Schmidt M, Garcia M, Siepmann T, Hasenburg A,
Anic K. Ecacy and Safety of Immunotherapy for Cervical Cancer-A System-
atic Review of clinical trials. Cancers 2022, 14(2).
51. Liu Y, Shou Y, Zhu R, Qiu Z, Zhang Q, Xu J. Construction and validation of a
Ferroptosis-Related Prognostic Signature for Melanoma based on single-cell
RNA sequencing. Front cell Dev Biology. 2022;10:818457.
52. Zhang Z, Zeng X, Wu Y, Liu Y, Zhang X, Song Z. Cuproptosis-related risk score
predicts prognosis and characterizes the Tumor Microenvironment in Hepa-
tocellular Carcinoma. Front Immunol. 2022;13:925618.
53. Lu H, Zhou L, Zhang B, Xie Y, Yang H, Wang Z. Cuproptosis key gene FDX1 is
a prognostic biomarker and associated with immune inltration in glioma.
Front Med. 2022;9:939776.
54. Yang L, Zhang Y, Wang Y, Jiang P, Liu F, Feng N. Ferredoxin 1 is a cuproptosis-
key gene responsible for Tumor immunity and drug sensitivity: a pan-cancer
analysis. Front Pharmacol. 2022;13:938134.
55. Sumaiya K, Langford D, Natarajaseenivasan K, Shanmughapriya S. Macro-
phage migration inhibitory factor (MIF): a multifaceted cytokine regulated by
genetic and physiological strategies. Pharmacol Ther. 2022;233:108024.
56. Noe JT, Mitchell RA. MIF-Dependent Control of Tumor Immunity. Front Immu-
nol. 2020;11:609948.
57. Rendon BE, Willer SS, Zundel W, Mitchell RA. Mechanisms of macrophage
migration inhibitory factor (MIF)-dependent Tumor microenvironmental
adaptation. Exp Mol Pathol. 2009;86(3):180–5.
58. Nishihira J, Ishibashi T, Fukushima T, Sun B, Sato Y, Todo S. Macrophage migra-
tion inhibitory factor (MIF): its potential role in Tumor growth and tumor-
associated angiogenesis. Ann N Y Acad Sci. 2003;995:171–82.
59. Qin S, Li A, Yi M, Yu S, Zhang M, Wu K. Recent advances on anti-angiogenesis
receptor tyrosine kinase inhibitors in cancer therapy. J Hematol Oncol.
2019;12(1):27.
60. Wei XW, Zhang ZR, Wei YQ. Anti-angiogenic Drugs currently in phase II
clinical trials for gynecological cancer treatment. Expert Opin Investig Drugs.
2013;22(9):1181–92.
61. Monk BJ, Mas Lopez L, Zarba JJ, Oaknin A, Tarpin C, Termrungruanglert W,
Alber JA, Ding J, Stutts MW, Pandite LN. Phase II, open-label study of pazo-
panib or lapatinib monotherapy compared with pazopanib plus lapatinib
combination therapy in patients with advanced and recurrent Cervical
cancer. J Clin Oncology: Ocial J Am Soc Clin Oncol. 2010;28(22):3562–9.
62. Miller SC, Huang R, Sak amuru S, Shukla SJ, Attene-Ramos MS, Shinn P, Van
Leer D, Leister W, Austin CP, Xia M. Identication of known Drugs that act as
inhibitors of NF-kappaB signaling and their mechanism of action. Biochem
Pharmacol. 2010;79(9):1272–80.
63. Shen J, Wang L, Bi J. Bioinformatics analysis and experimental validation of
cuproptosis-related lncRNA LINC02154 in clear cell renal cell carcinoma. BMC
Cancer. 2023;23(1):160.
64. Song Q, Zhou R, Shu F, Fu W. Cuproptosis scoring system to predict the
clinical outcome and immune response in Bladder cancer. Front Immunol.
2022;13:958368.
65. Sha S, Si L, Wu X, Chen Y, Xiong H, Xu Y, Liu W, Mei H, Wang T, Li M. Prognostic
analysis of cuproptosis-related gene in triple-negative Breast cancer. Front
Immunol. 2022;13:922780.
Publisher’s Note
Springer Nature remains neutral with regard to jurisdictional claims in
published maps and institutional aliations.
Content courtesy of Springer Nature, terms of use apply. Rights reserved.
1.
2.
3.
4.
5.
6.
Terms and Conditions
Springer Nature journal content, brought to you courtesy of Springer Nature Customer Service Center GmbH (“Springer Nature”).
Springer Nature supports a reasonable amount of sharing of research papers by authors, subscribers and authorised users (“Users”), for small-
scale personal, non-commercial use provided that all copyright, trade and service marks and other proprietary notices are maintained. By
accessing, sharing, receiving or otherwise using the Springer Nature journal content you agree to these terms of use (“Terms”). For these
purposes, Springer Nature considers academic use (by researchers and students) to be non-commercial.
These Terms are supplementary and will apply in addition to any applicable website terms and conditions, a relevant site licence or a personal
subscription. These Terms will prevail over any conflict or ambiguity with regards to the relevant terms, a site licence or a personal subscription
(to the extent of the conflict or ambiguity only). For Creative Commons-licensed articles, the terms of the Creative Commons license used will
apply.
We collect and use personal data to provide access to the Springer Nature journal content. We may also use these personal data internally within
ResearchGate and Springer Nature and as agreed share it, in an anonymised way, for purposes of tracking, analysis and reporting. We will not
otherwise disclose your personal data outside the ResearchGate or the Springer Nature group of companies unless we have your permission as
detailed in the Privacy Policy.
While Users may use the Springer Nature journal content for small scale, personal non-commercial use, it is important to note that Users may
not:
use such content for the purpose of providing other users with access on a regular or large scale basis or as a means to circumvent access
control;
use such content where to do so would be considered a criminal or statutory offence in any jurisdiction, or gives rise to civil liability, or is
otherwise unlawful;
falsely or misleadingly imply or suggest endorsement, approval , sponsorship, or association unless explicitly agreed to by Springer Nature in
writing;
use bots or other automated methods to access the content or redirect messages
override any security feature or exclusionary protocol; or
share the content in order to create substitute for Springer Nature products or services or a systematic database of Springer Nature journal
content.
In line with the restriction against commercial use, Springer Nature does not permit the creation of a product or service that creates revenue,
royalties, rent or income from our content or its inclusion as part of a paid for service or for other commercial gain. Springer Nature journal
content cannot be used for inter-library loans and librarians may not upload Springer Nature journal content on a large scale into their, or any
other, institutional repository.
These terms of use are reviewed regularly and may be amended at any time. Springer Nature is not obligated to publish any information or
content on this website and may remove it or features or functionality at our sole discretion, at any time with or without notice. Springer Nature
may revoke this licence to you at any time and remove access to any copies of the Springer Nature journal content which have been saved.
To the fullest extent permitted by law, Springer Nature makes no warranties, representations or guarantees to Users, either express or implied
with respect to the Springer nature journal content and all parties disclaim and waive any implied warranties or warranties imposed by law,
including merchantability or fitness for any particular purpose.
Please note that these rights do not automatically extend to content, data or other material published by Springer Nature that may be licensed
from third parties.
If you would like to use or distribute our Springer Nature journal content to a wider audience or on a regular basis or in any other manner not
expressly permitted by these Terms, please contact Springer Nature at
onlineservice@springernature.com
ResearchGate has not been able to resolve any citations for this publication.
Article
Full-text available
Background Clear cell renal cell carcinoma (ccRCC) is common in urinary system tumors. Cuproptosis is a non-apoptotic cell death pathway. Copper binds to fatty acylated mitochondrial proteins and activates various forms of cell death. LncRNA LINC02154 is significantly highly expressed in cells and tissues of many types of tumors, and the risk signature of LINC02154 in some tumors has been validated for effectiveness. Methods We constructed a risk prognostic signature by obtaining differentially expressed long noncoding RNAs (lncRNAs) associated with ccRCC outcomes and cuproptosis from The Cancer Genome Atlas (TCGA). We used TCGA to construct training and testing sets to analyze the risk signature and the impact of LINC02154, and we performed relevant survival analyses. Tumor mutational burdens were analyzed in different LINC02154 expression groups and risk score groups. We next analyzed the immune microenvironment of LINC20154. We performed LINC20154-related drug sensitivity analyses. We also investigated the cellular function of LINC02154 in the ACHN cell line and performed CCK-8 assay, EdU, wound-healing assay, and Transwell assay. The essential genes FDX1 and DLST of cuproptosis were detected by western blot. Results We demonstrated that LINC02154’s impact on outcomes was statistically significant. We also demonstrated the association of different ages, genders, stages, and grades with LINC02154 and risk models. The results showed a significant difference in tumor mutation burden between the groups, which was closely related to clinical prognosis. We found differences in immune cells among groups with different levels of LINC02154 expression and significant differences in immune function, immunotherapeutic positive markers, and critical steps of the immune cycle. The sensitivity analysis showed that differential expression of LINC02154 discriminated between sensitivity to axitinib, doxorubicin, gemcitabine, pazopanib, sorafenib, sunitinib, and temsirolimus. This difference was also present in the high-risk group and low-risk group. We demonstrated that the proliferation and migration of t ACHN cells in the LINC02154 knockdown group were inhibited. The western blot results showed that the knockdown of LINC02154 significantly affected the expression of FDX1 and DLST, critical genes of cuproptosis. Conclusion Finally, we demonstrated that LINC02154 and our constructed risk signature could predict outcomes and have potential clinical value.
Article
Full-text available
Recent studies have found that the protein encoded by the FDX1 gene is involved in mediating Cuproptosis as a regulator of protein lipoylation and related to immune response process of tumors. However, the specific biological function of FDX1 in glioma is currently unclear. To explore the potential function of FDX1 , this study explored the correlation between the expression of FDX1 in cancers and survival prognosis by analyzing the public databases of GEPIA and Cbioportal. Immune infiltration was analyzed by the TIMER2.0 database in tumors. The possible biological processes and functions of FDX1-related in glioma were annotated through gene enrichment. Relationship between Cuproptosis and autophagy was explored through gene co-expression studies. Summary and conclusions of this study: (1) FDX1 is highly expressed in gliomas and associated with poor prognosis in low-grade gliomas (LGG). (2) Gene annotation indicates that FDX1 is mainly involved in the tumor protein lipoylation and cell death. (3) FDX1 expression is positively correlated with the infiltration of immune cells. (4) LIPT2 and NNAT , two other genes involved in lipoylation, may be unidentified marker gene for Cuproptosis. And the Cuproptosis genes related to FDX1 were positively correlated with the expression of autophagy marker genes Atg5 , Atg12 , and BECN-1 . This evidence suggests that there may be some interaction between FDX1 mediated Cuproptosis and autophagy. In summary, FDX1 may serve as a potential immunotherapy target and prognostic marker for Glioma.
Article
Full-text available
In terms of mortality and survival, pancreatic cancer is one of the worst malignancies. Known as a unique type of programmed cell death, cuprotosis contributes to tumor cell growth, angiogenesis, and metastasis. Cuprotosis programmed-cell-death-related lncRNAs (CRLs) have been linked to PAAD, although their functions in the tumor microenvironment and prognosis are not well understood. This study included data from the TCGA-PAAD cohort. Random sampling of PAAD data was conducted, splitting the data into two groups for use as a training set and test set (7:3). We searched for differentially expressed genes that were substantially linked to prognosis using univariate Cox and Lasso regression analysis. Through the use of multivariate Cox proportional risk regression, a risk-rating system for prognosis was developed. Correlations between the CRL signature and clinicopathological characteristics, tumor microenvironment, immunotherapy response, and chemotherapy sensitivity were further evaluated. Lastly, qRT-PCR was used to compare CRL expression in healthy tissues to that in tumors. Some CRLs are thought to have strong correlations with PAAD outcomes. These CRLs include AC005332.6, LINC02041, LINC00857, and AL117382.1. The CRL-based signature construction exhibited outstanding predictive performance and offers a fresh approach to evaluating pre-immune effectiveness, paving the way for future studies in precision immuno-oncology.
Article
Full-text available
Ferredoxin 1 (FDX1) functions by transferring electrons from NADPH to mitochondrial cytochrome P450 via the ferredoxin reductase and is the key regulator in copper-dependent cell death. Although mounting evidence supports a vital role for FDX1 in tumorigenesis of some cancers, no pan-cancer analysis of FDX1 has been reported. Therefore, we aimed to explore the prognostic value of FDX1 in pan-cancer and investigate its potential immune function. Based on data from The Cancer Genome Atlas, Cancer Cell Line Encyclopedia, Genotype Tissue-Expression, Human Protein Atlas, and Gene Set Cancer Analysis, we used a range of bioinformatics approaches to explore the potential carcinogenic role of FDX1, including analyzing the relationship between FDX1 expression and prognosis, DNA methylation, RNA methylation-related genes, mismatch repair (MMR) gene, microsatellite instability (MSI), tumor mutation burden (TMB), tumor microenvironment (TME), immune-related genes, and drug sensitivity in different tumors. The results show that FDX1 was lowly expressed in most cancers but higher in glioblastoma multiforme, stomach adenocarcinoma, and uterine corpus endometrial carcinoma. Moreover, FDX1 expression was positively or negatively associated with prognosis in different cancers. FDX1 expression was significantly associated with DNA methylation in 6 cancers, while there was a correlation between FDX1 expression and RNA methylation-related genes and MMR gene in most cancers. Furthermore, FDX1 expression was significantly associated with MSI in 8 cancers and TMB in 10 cancers. In addition, FDX1 expression was also significantly correlated with immune cell infiltration, immune-related genes, TME, and drug resistance in various cancers. An experiment in vitro showed FDX1 is downregulated by elesclomol, resulting in inhibiting cell viability of bladder cancer, clear cell renal cell carcinoma, and prostate cancer cells. Our study reveals that FDX1 can serve as a potential therapeutic target and prognostic marker for various malignancies due to its vital role in tumorigenesis and tumor immunity.
Article
Full-text available
Cuproptosis is a novel copper ion-dependent cell death type being regulated in cells, and this is quite different from the common cell death patterns such as apoptosis, pyroptosis, necroptosis, and ferroptosis. Interestingly, like with death patterns, cuproptosis-related genes have recently been reported to regulate the occurrence and progression of various tumors. However, in bladder cancer, the link between cuproptosis and clinical outcome, tumor microenvironment (TME) modification, and immunotherapy is unknown. To determine the role of cuprotosis in the tumor microenvironment, we systematically examined the characteristic patterns of 10 cuproptosis-related genes in bladder cancer (BLCA). By analyzing principal component data, we established a cuproptosis score to determine the degree of cuproptosis among patients. Finally, we evaluated the potential of these values in predicting BLCA prognosis and treatment responses. A comprehensive study of the mutations of cuproptosis-related genes in BLCA specimens was conducted at the genetic level, and their expression and survival patterns were evaluated using The Cancer Genome Atlas (TCGA) and Gene Expression Omnibus (GEO). Two cuproptosis patterns were constructed based on the transcription level of 10 cuproptosis-related genes, featuring differences in the prognosis and the infiltrating landscape of immune cells (especially T and dendritic cells) with interactions between cuproptosis and the TME. Our study further demonstrated that cuproptosis score may predict prognosis, immunophenotype sensitivity to chemotherapy, and immunotherapy response among bladder cancer patients. The development and progression of bladder cancer are likely to be influenced by cuproptosis, which may involve a diverse and complex TME. The cuproptosis pattern evaluated in our study may enhance understanding of immune infiltrations and guide more potent immunotherapy interventions.
Article
Full-text available
Background Cuproptosis is a copper-dependent cell death mechanism that is associated with tumor progression, prognosis, and immune response. However, the potential role of cuproptosis-related genes (CRGs) in the tumor microenvironment (TME) of triple-negative breast cancer (TNBC) remains unclear. Patients and methods In total, 346 TNBC samples were collected from The Cancer Genome Atlas database and three Gene Expression Omnibus datasets, and were classified using R software packages. The relationships between the different subgroups and clinical pathological characteristics, immune infiltration characteristics, and mutation status of the TME were examined. Finally, a nomogram and calibration curve were constructed to predict patient survival probability to improve the clinical applicability of the CRG_score. Results We identified two CRG clusters with immune cell infiltration characteristics highly consistent with those of the immune-inflamed and immune-desert clusters. Furthermore, we demonstrated that the gene signature can be used to evaluate tumor immune cell infiltration, clinical features, and prognostic status. Low CRG_scores were characterized by high tumor mutation burden and immune activation, good survival probability, and more immunoreactivity to CTLA4, while high CRG_scores were characterized by the activation of stromal pathways and immunosuppression. Conclusion This study revealed the potential effects of CRGs on the TME, clinicopathological features, and prognosis of TNBC. The CRGs were closely associated with the tumor immunity of TNBC and are a potential tool for predicting patient prognosis. Our data provide new directions for the development of novel drugs in the future.
Article
Full-text available
Aims Cuproptosis is a recently identified form of programmed cell death; however, its role in hepatocellular carcinoma (HCC) remains unclear. Methods A set of bioinformatic tools was integrated to analyze the expression and prognostic significance of ferredoxin 1 (FDX1), the key regulator of cuproptosis. A cuproptosis-related risk score (CRRS) was developed via correlation analyses, least absolute shrinkage and selection operator (LASSO) Cox regression, and multivariate Cox regression. The metabolic features, mutation signatures, and immune profile of CRRS-classified HCC patients were investigated, and the role of CRRS in therapy guidance was analyzed. Results FDX1 was significantly downregulated in HCC, and its high expression was associated with longer survival time. HCC patients in the high-CRRS group showed a significantly lower overall survival (OS) and enriched in cancer-related pathways. Mutation analyses revealed that the high-CRRS HCC patients had a high mutational frequency of some tumor suppressors such as tumor protein P53 (TP53) and Breast-cancer susceptibility gene 1 (BRCA1)-associated protein 1 (BAP1) and a low frequency of catenin beta 1 (CTNNB1). Besides, HCC patients with high CRRS showed an increase of protumor immune infiltrates and a high expression of immune checkpoints. Moreover, the area under the curve (AUC) values of CRRS in predicting the efficiency of sorafenib and the non-responsiveness to transcatheter arterial chemoembolization (TACE) in HCC patients reached 0.877 and 0.764, respectively. Significance The cuproptosis-related signature is helpful in prognostic prediction and in guiding treatment for HCC patients.
Article
Full-text available
PURPOSE Resistance to immune checkpoint inhibition (ICI) in advanced non–small-cell lung cancer (NSCLC) represents a major unmet need. Combining ICI with vascular endothelial growth factor (VEGF)/VEGF receptor inhibition has yielded promising results in multiple tumor types. METHODS In this randomized phase II Lung-MAP nonmatch substudy (S1800A), patients ineligible for a biomarker-matched substudy with NSCLC previously treated with ICI and platinum-based chemotherapy and progressive disease at least 84 days after initiation of ICI were randomly assigned to receive ramucirumab plus pembrolizumab (RP) or investigator's choice standard of care (SOC: docetaxel/ramucirumab, docetaxel, gemcitabine, and pemetrexed). With a goal of 130 eligible patients, the primary objective was to compare overall survival (OS) using a one-sided 10% level using the better of a standard log-rank (SLR) and weighted log-rank (WLR; G[rho = 0, gamma = 1]) test. Secondary end points included objective response, duration of response, investigator-assessed progression-free survival, and toxicity. RESULTS Of 166 patients enrolled, 136 were eligible (69 RP; 67 SOC). OS was significantly improved with RP (hazard ratio [80% CI]: 0.69 [0.51 to 0.92]; SLR one-sided P = .05; WLR one-sided P = .15). The median (80% CI) OS was 14.5 (13.9 to 16.1) months for RP and 11.6 (9.9 to 13.0) months for SOC. OS benefit for RP was seen in most subgroups. Investigator-assessed progression-free survival (hazard ratio [80% CI]: 0.86 [0.66 to 1.14]; one-sided SLR, P = .25 and .14 for WLR) and response rates (22% RP v 28% SOC, one-sided P = .19) were similar between arms. Grade ≥ 3 treatment-related adverse events occurred in 42% of patients in the RP group and 60% on SOC. CONCLUSION This randomized phase II trial demonstrated significantly improved OS with RP compared with SOC in patients with advanced NSCLC previously treated with ICI and chemotherapy. The safety was consistent with known toxicities of both drugs. These data warrant further evaluation.
Article
Introduction: Cervical cancer is the overall fourth most common malignancy and the fourth most common cause of cancer related deaths worldwide. Despite vaccination and screening programs, many women continue to present with advanced stage cervical cancer, wherein the treatment options have been limited. Areas covered: In this review, immunotherapy and the potential targeted therapies that have demonstrated promise in the treatment of persistent, recurrent, and metastatic cervical cancer are discussed. Expert opinion: Our global goal in the gynecologic oncology community is to eliminate cervical cancer, by increasing the uptake of preventive vaccination and screening programs. For unfortunate patients who present with metastatic, persistent, and recurrent cervical cancer, pembrolizumab with chemotherapy, with or without bevacizumab is the new first line therapy for PD-L1 positive patients. For this patient population as a second line therapy, tisotumab vedotin (i.e. ADC) has shown significant efficacy in Phase II trials, leading to FDA approval. Combination regimens inclusive of immune checkpoint inhibitors, DNA damage repair inhibitors, antibody drug conjugates are potential breakthrough treatment strategies and are currently being investigated.