ArticlePDF Available

Immune-related LncRNAs scores predicts chemotherapeutic responses and prognosis in cervical cancer patients

Authors:

Abstract and Figures

Background Long non-coding RNAs (LncRNAs) regulating the immune microenvironment of cancer is a hot spot. But little is known about the influence of the immune-related lncRNA (IRlncRs) on the chemotherapeutic responses and prognosis of cervical cancer (CC) patients. The purpose of the study was to identify an immune-related lncRNAs (IRlncRs)-based model for the prospective prediction of clinical outcomes in CC patients. Methods CC patients’ relevant data was acquired from The Cancer Genome Atlas (TCGA). Correlation analysis and Cox regression analyses were applied. A risk score formula was formulated. Prognostic factors were combined into a nomogram, while sensitivity for chemotherapy drugs was analyzed using the OncoPredict algorithm. Results Eight optimal IRlncRs(ATP2A1-AS1, LINC01943, AL158166.1, LINC00963, AC009065.8, LIPE-AS1, AC105277.1, AC098613.1.) were incorporated in the IRlncRs model. The overall survival (OS) of the high-risk group of the model was inferior to those in the low-risk group. Further analysis demonstrated this eight-IRlncRs model as a useful prognostic marker. The Nomogram had a concordance index of survival prediction of 0.763(95% CI 0.746–0.780) and more robust predictive accuracy. Furthermore, patients in the low-risk group were found to be more sensitive to chemotherapy, including Paclitaxel, Rapamycin, Epirubicin, Vincristine, Docetaxel and Vinorelbine. Conclusions An eight-IRlncRs-based prediction model was identified that has the potential to be an important tool to predict chemotherapeutic responses and prognosis for CC patients.
Content may be subject to copyright.
Vol.:(0123456789)
Discover Oncology (2024) 15:119 | https://doi.org/10.1007/s12672-024-00979-1
Discover Oncology
Research
Immune‑related LncRNAs scores predicts chemotherapeutic responses
andprognosis incervical cancer patients
WeijieTian1· SongsongTan1· JunWang1· PingShen1· QingfenQin1· DanZi1
Received: 12 September 2023 / Accepted: 10 April 2024
© The Author(s) 2024 OPEN
Abstract
Background Long non-coding RNAs (LncRNAs) regulating the immune microenvironment of cancer is a hot spot. But little
is known about the inuence of the immune-related lncRNA (IRlncRs) on the chemotherapeutic responses and prognosis
of cervical cancer (CC) patients. The purpose of the study was to identify an immune-related lncRNAs (IRlncRs)-based
model for the prospective prediction of clinical outcomes in CC patients.
Methods CC patients’ relevant data was acquired from The Cancer Genome Atlas (TCGA). Correlation analysis and Cox
regression analyses were applied. A risk score formula was formulated. Prognostic factors were combined into a nomo-
gram, while sensitivity for chemotherapy drugs was analyzed using the OncoPredict algorithm.
Results Eight optimal IRlncRs(ATP2A1-AS1, LINC01943, AL158166.1, LINC00963, AC009065.8, LIPE-AS1, AC105277.1,
AC098613.1.) were incorporated in the IRlncRs model. The overall survival (OS) of the high-risk group of the model was
inferior to those in the low-risk group. Further analysis demonstrated this eight-IRlncRs model as a useful prognostic
marker. The Nomogram had a concordance index of survival prediction of 0.763(95% CI 0.746–0.780) and more robust
predictive accuracy. Furthermore, patients in the low-risk group were found to be more sensitive to chemotherapy,
including Paclitaxel, Rapamycin, Epirubicin, Vincristine, Docetaxel and Vinorelbine.
Conclusions An eight-IRlncRs-based prediction model was identied that has the potential to be an important tool to
predict chemotherapeutic responses and prognosis for CC patients.
Keywords Cervical squamous cell cancer· Immune· Long non-coding RNA· Prognosis· Chemotherapeutic Responses
Abbreviations
CC Cervical cancer
CSCC Cervical squamous cell carcinoma
FIGO International Federation of Gynaecology and Obstetrics
HPV Human papillomavirus
IRlncRs Immune-related long noncoding RNAs
lncRNAs Long noncoding RNAs
Weijie Tian, Songsong Tan should be considered similar in the author order of the study.
Qingfen Qin and Dan Zi are contributed equally.
Supplementary Information The online version contains supplementary material available at https:// doi. org/ 10. 1007/ s12672- 024-
00979-1.
* Qingfen Qin, tanqingfeng@gz5055.com; * Dan Zi, zidangy08@163.com | 1Department ofGynecology, Guizhou Provincial Peoples
Hospital, Medical College ofGuizhou University, Guiyang, Guizhou, People’sRepublicofChina.
Vol:.(1234567890)
Research Discover Oncology (2024) 15:119 | https://doi.org/10.1007/s12672-024-00979-1
ncRNAs Noncoding RNAs
OS Overall survival
sIRlncRs Survival-related IRlncRNAs
TCGA The Cancer Genome Atlas
TMM Trimmed mean of M-values
PCA Principal components analysis
GSEA Gene set enrichment analysis
ROC Receiver operating characteristic
AUC Area under the curve
IC50 Half-maximal inhibitory concentration
1 Introduction
Cervical cancer (CC) is still the common cause of disease-related mortalities in women, leading to nearly 300,000 deaths
worldwide [1]. Cervical squamous cell carcinoma (CSCC) is the primary pathological subtype and comprises the most CC
cases [2]. Despite the utilization of treatment modalities such as surgery, radiotherapy, and chemotherapy, these therapies
have shown limited ecacy in patients with advanced-stage disease [35]. The International Federation of Gynaecology
and Obstetrics (FIGO) stage is the primary prognostic indicator for CC. However, the FIGO stage cannot dierentiate the
various heterogeneity of CC in terms of clinical behavior. Patients with the same FIGO stage may often present obviously
dierent clinical outcomes. Therefore, identifying new prognostic indicators reecting the heterogeneity of CC is essential
and can facilitate individualized treatments for patients with an otherwise poor prognosis.
Increasing evidence showed that immune system disruption might lead to tumour progression and metastasis [6, 7].
Malignant cells can escape immunosurveillance by reducing the expression of major histocompatibility complex class
I molecules. Cervical adenocarcinoma has impaired recruitment of CDC1 and CD8 + T cells [8, 9]. Higher CCL22 + cell
inltration is negatively associated with prognosis in CC patients [9]. LINC00240 promotes natural killer T cell cytotoxic
activity in CC and enhanced the growth, invasion, and migration of CC cells [10].
LncRNAs are a class of non-coding RNAs (ncRNAs) with longer than 200 nucleotides. With the development of tran-
scriptome sequencing, it is clear that over 70% of the genome is transcribed into RNA, and the majority of them are
ncRNAs [11]. lncRNAs are involved in various transcriptional and post-transcriptional gene regulatory processes and play
crucial roles in the tumour immune response, including immune recognition and immune inltration [11]. Numerous
tumor-associated lncRNAs have been recognized as tumor cell factors that regulate tumor cell escape of immunosur-
veillance. These immune-related lncRNAs (IRlncRs) may play essential parts in immunotherapy resistance and further
impact the prognosis of cancer patients [12]. Specically, a subset of lncRNAs act as immune-related lncRNAs (IRlncRs)
by regulating immune responses in the tumor microenvironment. This subset of lncRNAs has been shown to play a key
role in modulating tumor immunosurveillance, immune cell inltration into the tumor microenvironment, and sensitiv-
ity of cancer cells to immunotherapy treatment [12, 13]. Cao etal. identied an immune-related ve-lncRNAs signature
positively correlated with tumour immune cell inltration and the poor prognosis in bladder cancer patients. The asso-
ciation of IRlncRs expression and the clinical outcomes of CC patients is reported, but the results lack validation [12].
The development of new prognostic markers is essential considering the inherent heterogeneity of cervical cancer,
guiding personalized treatment strategies. Reliable assessment of chemotherapy responses and determination of prog-
nostic risk would enable physicians to tailor more accurate treatment plans to improve outcomes. The IRlncRs signature
holds strong potential in risk stratication and chemotherapy selection for cervical cancer patients. By evaluating prog-
nosis and chemotherapy sensitivity, the IRlncRs model can provide a basis for clinical decision-making, oering patients
the most likely successful treatment strategies based on their molecular risk proles. To achieve this, we developed an
innovative IRlncRs model and performed preliminary invitro validation, demonstrating its ability to distinguish high-
risk and low-risk cervical cancer patients with signicant dierences in overall survival. Further analysis conrmed the
prognostic predictive capability of this model. Additionally, this model demonstrated utility in predicting chemotherapy
response, with high-risk patients showing resistance to several commonly used drugs.
Vol.:(0123456789)
Discover Oncology (2024) 15:119 | https://doi.org/10.1007/s12672-024-00979-1 Research
2 Materials andmethods
2.1 Patient datasets
CC transcriptome RNA-seq data in the format of Fragments Per Kilobase Million and the corresponding clinical data of
The Cancer Genome Atlas (TCGA) (GDC, https:// gdc. cancer. gov/) were downloaded.
2.2 Data preprocessing andnormalization
The raw RNA-sequencing count data from TCGA cervical cancer cohorts was preprocessed to ensure normalization
and integrity for downstream analyses. Briey, quality control was rst performed using FastQC to assess attributes like
guanine-cytosine content, overrepresented sequences, and duplication levels. Reads were then trimmed and ltered
to remove adapters and low-quality bases using Trimmomatic.
The RNA-seq pre-processed data was quantied and normalized using the Trinity pipeline (https:// github. com/
NCIP/ Trini ty_ CTAT) to generate normalized gene-level count data. This pipeline maps reads, assembles transcripts,
estimates abundances, and extracts dierentially expressed features. Normalization was conducted using the trimmed
mean of M-values (TMM) method to account for dierences in sequencing depth between samples using the EdgeR R/
Bioconductor package. TMM normalization controls for library size variability via scaling based on the ratio of read counts
between samples. Normalized expression data is represented as counts per million (CPM).
Samples with > 50% missing lncRNA expression data were excluded from analysis to avoid technical bias. For the
remaining samples, missing values were imputed using a k-nearest neighbor algorithm with k = 10 neighbors. Imputation
was conducted using the Bioconductor impute package. This allowed retention of samples with some missing data
rather than complete exclusion.
Additional ltering of lncRNAs was conducted to restrict analysis to those with evidence of abundance and variation
across samples. LncRNAs expressed at 0.5 CPM in at least 10% of samples and with an interquartile range greater than
0 were retained.
2.3 LncRNA profile mining
Three gene sets, “immune response(M19817)”, “immune system development(M3457)”, and “immune system
process(M13664)”, were acquired from the Molecular Signatures Database. LncRNAs with abundance lower than 0.5
and lncRNAs of normal tissues were excluded. Then, immune-related genes were acquired from the above three gene
sets. The Pearson correlation test analyses the correlation between the immune-related genes and lncRNAs. Absolute
value of correlation coecient > 0.5 and p < 0.001 were dened as IRlncRs.
2.4 Real‑time quantitative PCR
Total RNA from cell lines (Hela cell and HCerEpiC cell) was isolated using Trizol reagent (Invitrogen, USA) according
to the manufacturer’s instructions. cDNA Synthesis Kit (TaKaRa, Japan) was utilized to generate cDNA. 4.5μL diluted
cDNA (1:50) was used as the template in a 10μL qPCR reaction using the ABI 7500 fast real-time PCR system (Applied
Biosystems). GAPDH was used as a reference. The relative expression level was calculated by the 2−ΔCt method. Table1
shows the sequences of the forward and reverse primers of eight examined IRlncRs (ATP2A1-AS1, LINC01943, AL158166.1,
LINC00963, AC009065.8, LIPE-AS1, AC105277.1, AC098613.1.).
2.5 Model development
Univariate and multivariate Cox regression models were utilized to identify lncRNAs prognostic of overall survival and
build a predictive risk score formula. The Cox proportional hazards regression model was selected because it allows
assessment of the association between continuous gene expression data and censored survival outcomes while adjust-
ing for the eects of other covariates. Univariate Cox regression was carried out to extract IRlncRs correlated with the
OS of patients with CC at p < 0.05. Next, only the IRlncRs with a statistical signicance of p < 0.01 were further enrolled
in the stepwise multivariate Cox regression analysis to extract optimal IRlncRs independently associated with prognosis
Vol:.(1234567890)
Research Discover Oncology (2024) 15:119 | https://doi.org/10.1007/s12672-024-00979-1
at p < 0.05. Finally, the survival-related IRlncRs(sIRlncRs) model (risk score) was constructed according to the regression
coecients with lncRNA expression. In other words, the prognostic risk score was formulated based on a linear combi-
nation of the expression level of theses IRlncRs multiplied by the regression coecients derived from the multivariate
Cox regression analysis, as mentioned above [14]. Patients were grouped into a high- and low-risk group according to
the median value of risk scores.
Kaplan–Meier analysis was employed to validate survival dierences between the high-risk and low-risk groups
stratied by the risk score formula. This non-parametric analysis was chosen because it is well-suited for estimating
group survival functions over time while accounting for censoring, which was essential for the overall survival endpoint
that had censored observations (patients still alive at last follow-up). The Kaplan–Meier estimator also provides median
survival times and key quantied survival statistics for each risk group. Using this method enabled validation of the risk
score by verifying poorer survival prognosis in the high-risk group compared to the low-risk group in a time-to-event
analysis context.
2.6 Independent prognostic analysis
We applied both single and multifactorial analyses to validate the validity of the risk score being an independent
prognostic marker for CC. The receiver operator characteristic (ROC) curve was utilized to assess whether the risk score’s
predictive power was reliable. The relationship between clinical traits and sIRlncRs was also studied. We also employed
the PCA (principal components analysis) method to demonstrate the distribution patterns between the low- and high-
risk groups. GSEA was applied to explore the distinct functional phenotypes between the high-risk and low-risk groups.
2.7 Construction apredictive nomogram
A prognostic nomogram including risk scores and clinical features for predicting the likelihood of 3-, and 5-year OS was
developed by R “rms” package. The calibration curves and C-index were used to evaluate the predictive accuracy of the
nomogram [15].
2.8 Prediction ofchemotherapeutic response
The clinical response of each CC patient in high- and low-risk groups to chemotherapy was estimated based on
the Genomics of Drug Sensitivity in Cancer (GDSC; https:// www. cance rrxge ne. org/) data. Twenty commonly used
chemotherapy drugs of CC, were selected for the chemotherapeutic response prediction through the ridge regression
Table 1 The forward and
reverse primer sequences
of eight examined IRlncRs
(ATP2A1-AS1, LINC01943,
AL158166.1, LINC00963,
AC009065.8, LIPE-AS1,
AC105277.1, AC098613.1.)
for performing real-time PCR
assay
Gene symbol Primer Primer Sequence (5-3)
ATP2A1-AS1 Primer_F GAG GAG AAT CCG CAC CAG GA
Primer_R TAG CCA CAA AGT CTT GGG TGT
LINC01943 Primer-F CAG GAA GCG TGA GGA CAG AA
Primer-R AAC CAG ACT GAT GCC ACA GG
AL158166.1 Primer-F TGA GCA TAG CCT CCA CTC CT
Primer-R AGA CAG CAC TGT CAG TCA CG
LINC00963 Primer-F GAA CTG CCT TTG GAA GCA AG
Primer-R AGG AGT TCG AGG CTG CAG TA
AC009065.8 Primer-F TTA GCT GGG CTG CGT TTA CA
Primer-R CCA CTC TCC CAC CTC CCT TA
LIPE-AS1 Primer-F CTC TGT CTC CGC CCC CTA AT
Primer-R TTC TCA AGC ATG CGT CGT TC
AC105277.1 Primer-F GTG ACC AGG TAC TGG GGA AA
Primer-R AAT GAG GTT CCA CAC CTG CT
AC098613.1 Primer-F GGG GAA AAT CAT CTC CCA TT
Primer-R TCA CAT TGC TCT GCC TCA TC
Vol.:(0123456789)
Discover Oncology (2024) 15:119 | https://doi.org/10.1007/s12672-024-00979-1 Research
using the “OncoPredict” R package [16]. The half-maximal inhibitory concentration (IC50) predicted of each CC patient
was used to assess dierential chemotherapeutic response.
2.9 Statistical analysis
Statistical analyses were carried out by the R statistical programming environment (version 4.0.2). Correlations between
the immune-related genes and lncRNAs were tested using the Pearson correlation test. Sensitivity and specicity of
signature were determined by ROC curves representing its power to dierentiate the dierent groups. The R package
“survivalROC” was used to calculate the area under the curve [17], and the “survival” R package was loaded to gure
survival analysis [18].
3 Results
3.1 The analysis process ofthis study
Figure1 displays the analysis process of our study. We downloaded transcriptome RNA-seq data and corresponding
clinical data of 289 cases of CC from the TCGA database. Among these cases, there were 253 CSCC patients, 33 cervical
adenocarcinoma patients, and 3 healthy control patients (Additional file3: TableS1). Then, the RNA-seq data were
Fig. 1 Analysis of the work-
ow of this study
Vol:.(1234567890)
Research Discover Oncology (2024) 15:119 | https://doi.org/10.1007/s12672-024-00979-1
divided into mRNA and lncRNAs data. LncRNAs with an abundance less than 0.5 and normal tissue lncRNAs were
excluded. We identified 331 immune-related genes from gene sets of MSigDB, of which 255 lncRNAs were IRlncRs
validated by correlation analysis (Additional file1: Fig. S1). Next, we identified 28 IRlncRs that were associated with the
prognosis of CC. We further optimized theses IRlncRs by stepwise multivariate Cox regression, and eight sIRlncRs were
utlized to formulate the risk score model. Finally, we utilized the risk score model for a series of subsequent analyses,
including survival analysis, risk score analysis, clinicopathological characteristics, ROC curve analysis, PCA, and GSEA.
3.2 Construction ofanIRlncRs‑based risk score model
Of the 28 IRlncRs related to the prognosis of CC (p < 0.01), 25 were low-risk factors, and 3 were high-risk factors
(Additional file3: TableS2). Eight sIRlncRs were finally incorporated to formulate the risk score model, including
ATP2A1-AS1, LINC01943, AL158166.1, LINC00963, AC009065.8, LIPE-AS1, AC105277.1, AC098613.1 (Table2). All CC
samples were categorized into low-and the high-risk groups using the median risk score as a boundary (Fig.2A). The
vital status of each patient was plotted. The proportion of death events in different risk groups was also analyzed. The
mortality rate increased faster in the high-risk group than in the low-risk group (Fig.2B). The differentially expressed
genes (DEGs) displayed that the expression levels of AL158166.1 and AC105277.1 had a positive coefficient and acted
as risk factors. The other six sIRlncRs showed negative coefficients, including ATP2A1-AS1, LINC01943, LINC00963,
AC009065.8, LIPE-AS1, AC098613.1, and served as protective factors (Fig.2C).
To verify the clinical value of the selected sIRlncRs in predicting prognosis, we compared the expression levels of
the eight sIRlncRs in cervical cancer cells to that in normal human cervical epithelial cells. As illustrated in Fig.2D,
five of the six sIRlncRs serving as protective factors showed a significant decrease in the cervical cancer cells. One of
the two IRlncRs acting as risk factors showed a significant increase in the cervical cancer cells.
Moreover, Kaplan–Meier survival analysis was used to evaluate the above prognosis model’s impact on CC patients’
survival. Survival was inferior in the high-risk group than in the low-risk group (Fig.3).
3.3 Independent prognostic analysis
To explore the relationship between the selected IRlncRs and clinical features of CC, the potential association of
the eight IRlncRs with the clinicopathological features, including T-stage, N-stage, and tumor grading, was investi-
gated. The results presented that the expression level of LINC00963 negatively correlated with the grading, while
AL158166.1 was positively related to advanced grading (Fig.4A). The expression of LINC00963 and AC105277.1
decreased with progressive T-stages (Fig.4B), and the expression of LIPE−AS1 increased with the progression of the
N-stage (Fig.4C). We then performed independent risk analysis, and it showed the risk score model, N-stage, and
T-stage were negatively related to the OS in univariate analysis (p < 0.05) (Fig.5A). The results were further confirmed
in the multivariate analysis showing that the risk score model, N-stage, and T-stage were significantly associated with
OS (p < 0.05) (Fig.5B). The ROC (Receiver Operating Characteristic) curve analysis validated this finding, demonstrat-
ing the predictive accuracy of the model. The AUC values for grade, T-stage, N-stage and risk score model were 0.516,
0.704, 0.633, and 0.710, respectively (Fig.6). These results demonstrated the risk score model as an independently
reliable prognostic factor.
Table 2 Eight immune-related
lncRNAs identied from
multivariate Cox regression
analysis
Gene symbol Ensembl ID coef HR Low95 High95 p-value
ATP2A1-AS1 ENSG00000260442 −0.36 0.7 0.48 1 0.05
LINC01943 ENSG00000280721 −0.94 0.39 0.14 1.11 0.08
AL158166.1 ENSG00000227076 0.57 1.76 1.15 2.69 0.01
LINC00963 ENSG00000204054 −0.48 0.62 0.39 0.98 0.04
AC009065.8 ENSG00000261532 −0.56 0.57 0.3 1.07 0.08
LIPE-AS1 ENSG00000213904 −0.58 0.56 0.29 1.09 0.09
AC105277.1 ENSG00000232453.7 0.83 2.29 1.36 3.85 0
AC098613.1 ENSG00000121797 −0.85 0.43 0.14 1.3 0.14
Vol.:(0123456789)
Discover Oncology (2024) 15:119 | https://doi.org/10.1007/s12672-024-00979-1 Research
Fig. 2 Construction of an IRlncRs-based risk score model. A The eight IRlncRs-based risk score distribution; B The eight-IRlncRs-based risk
score distribution for CC patient survival status. C Heatmap of the eight-IRlncRs expression proles in the high-risk and low-risk subgroups;
D Relative expression of the 8 IRlncRs
Vol:.(1234567890)
Research Discover Oncology (2024) 15:119 | https://doi.org/10.1007/s12672-024-00979-1
3.4 Construction ofthenomogram
The factors of age, grade, T-stage, N-stage and risk score were further combined to construct a compound nomogram for
predicting the OS of patients with CC at 3- and 5-year (Fig.7A). The points for the factors indicated their corresponding
contribution to the survival probability. The total points of each patient provided the estimated 3- and 5-year OS. The
C-index of our nomogram was 0.763(95% CI 0.746–0.780, p < 0.05). The actual recurrence rate and nomogram-predicted
survival rate matched well at 3years (Fig.7B) and 5years (Fig.7C), as shown by the calibration curves (Fig.7B, C).
3.5 The immune status ofthelow andhigh‑risk groups
We performed PCA to explore the dispersion of the low-and high-risk groups based on genome-wide expression sets
and the immune gene sets. Considering the immune gene sets, the low-and high-risk groups showed clustering (Fig.8A),
although there was no signicant separation of the two groups based on the genome-wide expression proles (Fig.8B).
The GSEA further veried the dierences in functional annotation. As shown in Fig.8C, D, the low-risk group’s genes
were predominantly mapped to the immune-related activities, such as immune response and immune system process.
However, there was no gene enriched in the high-risk group (p > 0.05).
3.6 Analysis ofchemotherapeutic responses inhigh‑ andlow‑risk groups
A total of 198 drugs were analyzed, and drug response to twenty commonly used chemotherapy drugs for CC were
analyzed using the Wilcoxon rank-sum test. There were signicantly lower IC50 levels for Paclitaxel, Rapamycin, Epiru-
bicin, Vincristine, Docetaxel, and Vinorelbine in the low-risk group compared with the high-risk group (Fig.9, p < 0.05),
indicating that the low-risk group was more sensitive to these drugs. Among the 20 drugs, only docetaxel and lapatinib
showed no signicant dierence in IC50 values (Additional le2: Fig. S2), which indicated that our IRlncRs-based risk
model might act as a potential predictor for chemosensitivity.
4 Discussion
Eight IRlncRs correlated with the overall survival of CC patients were identied. The risk score model based on these
eight IRlncRs demonstrated a strong ability to distinguish CC patients into low- and high-risk groups, which exhibited
signicant dierences in OS. Further multivariate analysis showed that the eight-IRlncRs model is a valid marker of OS
when accounting for other clinical characteristics, including T-stage and N-stage. The prognostic factors were further
analyzed and integrated into a well-designed nomogram that demonstrated high potential for clinical application.
Fig. 3 Survival curve of
CC patients. Kaplan–Meier
survival curve of OS among
CC patients from the low-risk
groups and high-risk groups.
The high-risk group show the
poorer prognosis
Vol.:(0123456789)
Discover Oncology (2024) 15:119 | https://doi.org/10.1007/s12672-024-00979-1 Research
Therefore, the eight-lncRNA model demonstrated promising value as a prognostic predictor of chemotherapeutic
responses in CC. PCA based on the immune gene sets demonstrated that the low- and high-risk groups exhibited
distinct immune statuses, with more abundant immune-related processes and responses observed in the low-risk group.
Finally, OncoPredict analysis revealed that the tissues from the high-risk group were resistant to six commonly used
chemotherapy drugs for CC.
Patients in the low-risk group may possess a better immune status, making them more sensitive to chemotherapy
drugs; conversely, patients in the high-risk group might exhibit an immunosuppressed state, leading to resistance
against chemotherapy drugs. The low-risk group presented enhanced antitumor immune pathways in the tumor
Fig. 4 The relationships between the sIRlncRs and clinical features. A grading; B T-stage; C N-stage
Vol:.(1234567890)
Research Discover Oncology (2024) 15:119 | https://doi.org/10.1007/s12672-024-00979-1
microenvironment. Higher expression of immunostimulatory molecules promotes inltration and activity of immune cells
such as T cells and natural killer (NK) cells [19]. Robust immune activation better sensitizes tumor cells to chemotherapy
through increased antigen presentation and vulnerability to immune-mediated killing [20]. In contrast, the high-
risk group exhibited an immunosuppressed state. Downregulation of critical immune modulators reduces tumor
immunogenicity through diminished antigen presentation and a decreased presence of cytotoxic lymphocytes [21].
This allows for immune evasion and subsequent resistance against chemotherapy, which relies on the immune system
recognizing and responding to cancer cells damaged by drug treatment [22].
Certain immune cell and mRNAs predictors have been studied to predict treatment outcomes of gynecological cancer.
Several risk score models based on dierentially expressed genes have been developed to assess the outcomes of women
with female reproductive cancers. Pan etal. reported 149 genes that were correlated with the survival of CSCC patients,
and most of these genes were closely related to T cell activation [23]. Mairinger etal. developed a predictive scoring
Fig. 5 Cox regression. A Uni-
variate Cox regression showed
that the T stage, N stage, and
risk score model were corre-
lated with the prognosis of CC
patients. B Multivariate Cox
regression showed that the T
stage, N stage, and risk score
model were an independent
risk factor for CC patients
Fig. 6 Receiver operating
characteristic (ROC) curve.
ROC curves demonstrated the
prognostic value of the inde-
pendent prognostic factors
Vol.:(0123456789)
Discover Oncology (2024) 15:119 | https://doi.org/10.1007/s12672-024-00979-1 Research
system based on immune-related genes to predict the therapy response and prognosis of epithelial ovarian cancer;
however, the system was not validated for OS prediction in two datasets [24]. Yang etal. utilized 11 immune-related
genes to formulate an immune signature for predicting clinical outcomes and the response to immune checkpoint
inhibitors in CC patients [25]. Although several prediction models based on immune-related genes have been previously
developed [23, 24], they face challenges such as a large number of genes that aect their practical utility [23] or a lack of
validation across multiple datasets [24]. Compared to other mRNA-based prediction models, the lncRNA-based model
constructed in this study exhibits higher specicity and provides a more precise reection of the actual tumor condition
[26]. Moreover, this study goes beyond constructing a prediction model and delves into the dierences in immune status
and chemotherapy sensitivity between high-risk and low-risk groups, thereby supporting the clinical application of
this model. The nomogram demonstrated excellent predictive performance (C-index of 0.763), highlighting its robust
potential for clinical application. Additionally, among the eight immune-related lncRNAs identied in this study, only
LINC00963 and AC098613.1 have been previously reported [27, 28], while the other six have not. The current study
represents the rst discovery of their association with cervical cancer prognosis.
Fig. 7 The Nomogram for predicting overall survival of CC patients. A The Nomogram integrating the signature risk score with the clinical
characteristics for predicting OS. B The calibration curve for the Nomogram in TCGA cohort for predicting 3-year overall survival. C The cali-
bration curve for the Nomogram in TCGA cohort for predicting 5-year overall survival
Vol:.(1234567890)
Research Discover Oncology (2024) 15:119 | https://doi.org/10.1007/s12672-024-00979-1
lncRNAs may have more specicity in presenting the actual tumor condition than other types of markers. A ten-lncRNA
signature for predicting the survival of patients with CC showed potential value as a prognostic biomarker for CC patients
(He etal. [27]). Compared to regular lncRNAs, immune-related lncRNAs (IRlncRs) are highly associated with the immune
system and exhibit distinct functions in the development of tumors [29]. Regular lncRNAs primarily regulate biological
processes such as tumor cell growth, proliferation, apoptosis, and migration, directly inuencing tumor formation and
progression. In contrast, the key role of IRlncRs lies in regulating the immune response in the tumor microenvironment
and participating in the process of determining whether tumor cells can evade immune surveillance. Specically, IRlncRs
can impact antigen expression on tumor cell surfaces, alter the local immune microenvironment of tumors, and thereby
aect whether tumor cells can be recognized and eliminated by immune cells. This is quite dierent from the direct eects
of regular lncRNAs on the biological functions of tumor cells [12, 30]. Therefore, the expression levels of IRlncRs often
correlate with clinical outcomes such as sensitivity to immunotherapy and prognosis [13], highlighting their advantage
as tumor biomarkers.
LINC00963 participates in the progression of several types of cancers, including lung cancer [31], prostate cancer
[28], and breast cancer [32]. LINC00963 can activate the oncogenic AKT/mTOR signaling pathway or EGFR signaling
pathway to enhance cancer cell metastasis [28]. AC098613.1 was also included in a four-lncRNA risk score serving as
an independent marker to predict the survival of bladder urothelial cancer patients [27]. However, the remaining six
Fig. 8 Principal components analysis (PCA) and gene set enrichment analysis (GSEA). A PCA plot showing high-risk group and low-risk
groups based on the immune-related gene sets. B PCA plot showing high-risk group and low-risk group based on the whole protein-coding
gene sets. C, D GSEA implied remarkable enrichment of immune-related phenotype in the low-risk group;
Vol.:(0123456789)
Discover Oncology (2024) 15:119 | https://doi.org/10.1007/s12672-024-00979-1 Research
IRlncRs have not been reported in the literature to date, and GSEA was conducted to predict their potential functional
annotations. The results showed more abundant immune-related processes in the low-risk group compared to the high-
risk group. Consistent infection by human papillomavirus (HPV), the primary etiology of CC, can lead to the shutdown
of host immune detection and the establishment of a local immunosuppressive status in HPV-associated CC [33]. These
six IRlncRs may play a signicant role in regulating these immune-related processes, and their modes of action warrant
further research.
This study primarily utilized bioinformatics analysis methods to establish the association between immune-related
long non-coding RNAs (IRlncRs) and the prognosis of CC, based on the reported lncRNA expression prole data from TCGA
database. The use of this large-scale database signicantly reduces the experimental workload and allows for ecient
identication of candidate biomarkers associated with prognosis. However, relying solely on bioinformatics predictions
has certain limitations, including potential biases resulting from selective population sampling, the retrospective nature
that may overlook important variables, and the absence of extensive external validation to ensure wider applicability.
Furthermore, the investigation of IRlncRs as prognostic markers shows promise but is still in its early stages, requiring
further research to understand their complex mechanisms and interactions in CC. The insucient comprehensive
metastasis data, the necessity for more rigorous experimental validation to conrm quantitative polymerase chain
reaction (qPCR) results, and the evaluation of clinical usefulness comprise the limitations and constraints of this study,
providing directions for future work.
Further studies could also consider validating the accuracy of this model in peripheral blood samples. Compared to
tissue samples, peripheral blood samples are more readily accessible and provide a comprehensive reection of the
body’s immune status, potentially resulting in higher accuracy of the predictive model [34]. However, the consistency
of lncRNA expression patterns between tumor tissue and peripheral blood may vary depending on the cancer type
and specic lncRNA. Certain lncRNAs have been explored as potential blood-based biomarkers for various cancers.
Fig. 9 Dierential chemotherapeutic responses of 6 drugs in low- and high-risk CC patients (AF)
Vol:.(1234567890)
Research Discover Oncology (2024) 15:119 | https://doi.org/10.1007/s12672-024-00979-1
For example, HOTAIR and LINC00152 show high specicity in identifying colorectal and gastric cancers, respectively.
However, it is notable that the diagnostic performance of many circulatory lncRNAs is still relatively poor when detected
individually, suggesting dierences in their expression patterns between blood and tumor tissue [34]. Despite the
challenges, developing predictive models using circulatory immune-related lncRNAs should be feasible, and this requires
a multidisciplinary approach involving molecular biology, bioinformatics, clinical research, and ethical considerations.
5 Conclusion
In conclusion, we identied an eight-IRlncRs signature that has the potential to be an important prognostic tool for CC
patients. We expect this IRlncRs model to be practical for forecasting clinical behaviour and guide precision medicine
approaches.
Acknowledgements The contents of the manuscript do not previously appear online. The authors would like to thank the TCGA databases
for the availability of the data.
Author contributions Collection and assembly of data: Wei-Jie Tian. Data analysis and interpretation: Songsong Tan and Jun Wang. Manuscript
writing: Weijie Tian, Qingfeng Qin and Ping Shen. Paper revision: Dan Zi, Qingfeng Qin. All authors have read and agreed to the published
version of the manuscript.
Funding The present study was supported by the Science and Technology Program of Guizhou Province, China (Grant numbers: Qian Ke He
Ji Chu-ZK[2023] Yi Ban 204) and the Talent Project for Guizhou Provincial People’s Hospital(Grant numbers: Yuan Ren Cai Project [2022] -31).
Data availability IThe data used to support the ndings of this study are available from the TCGA open database (https:// tcgad ata. nci. nih.
gov/ tcga/; LUAD).
Declarations
Competing interests The authors declare no competing interests.
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:// creat iveco mmons. or g/ lic en ses/ b y/4. 0/.
References
1. Siegel RL, Miller KD, Jemal A. Cancer statistics, 2019. CA Cancer J Clin. 2019;69(1):7–34.
2. Arbyn M, Weiderpass E, Bruni L, de Sanjosé S, Saraiya M, Ferlay J, etal. Estimates of incidence and mortality of cervical cancer in 2018: a
worldwide analysis. Lancet Glob Health. 2020;8(2):e191-203.
3. Cohen PA, Jhingran A, Oaknin A, Denny L. Cervical cancer. Lancet. 2019;393(10167):169–82.
4. Ye Q, Yang Y, Tang X, Li J, Li X, Zhang Y. Neoadjuvant chemotherapy followed by radical surgery versus radiotherapy (with or without
chemotherapy) in patients with stage IB2, IIA, or IIB cervical cancer: a systematic review and meta-analysis. Dis Markers. 2020. https://
doi. org/ 10. 1155/ 2020/ 74150 56.
5. Grigsby PW, Massad LS, Mutch DG, Powell MA, Thaker PH, McCourt C, etal. FIGO 2018 staging criteria for cervical cancer: Impact on stage
migration and survival. Gynecol Oncol. 2020;157(3):639–43.
6. 홍인선. Stimulatory versus suppressive eects of GM-CSF on tumor progression in multiple cancer types. Exp Mol Med. 2016;48:1–8.
7. Quail DF, Joyce JA. Microenvironmental regulation of tumor progression and metastasis. Nat Med. 2013;19(11):1423–37.
8. Wang Q, Schmoeckel E, Kost BP, Kuhn C, Vattai A, Vilsmaier T, etal. Higher CCL22+ cell inltration is associated with poor prognosis in
cervical cancer patients. Cancers. 2019;11(12):2004.
9. Rotman J, Heeren AM, Gassama AA, Lougheed SM, Pocorni N, Stam AG, etal. Adenocarcinoma of the uterine cervix shows impaired
recruitment of cDC1 and CD8+ T cells and elevated β-catenin activation compared with squamous cell carcinoma. Clin Cancer Res.
2020;26(14):3791–802.
10. Zhang Y, Li X, Zhang J, Liang H. Natural killer T cell cytotoxic activity in cervical cancer is facilitated by the LINC00240/microRNA-124-3p/
STAT3/MICA axis. Cancer Lett. 2020;474:63–73.
11. Djebali S, Davis CA, Merkel A, Dobin A, Lassmann T, Mortazavi A, etal. Landscape of transcription in human cells. Nature.
2012;489(7414):101–8.
Vol.:(0123456789)
Discover Oncology (2024) 15:119 | https://doi.org/10.1007/s12672-024-00979-1 Research
12. Denaro N, Merlano MC, Lo NC. Long noncoding RNA s as regulators of cancer immunity. Mol Oncol. 2019;13(1):61–73.
13. Egranov SD, Hu Q, Lin C, Yang L. LncRNAs as tumor cell intrinsic factors that aect cancer immunotherapy. RNA Biol. 2020;17(11):1625–7.
14. Chen HY, Yu SL, Chen CH, Chang GC, Chen CY, Yuan A, etal. A ve-gene signature and clinical outcome in non–small-cell lung cancer. N
Engl J Med. 2007;356(1):11–20.
15. Park SY. Nomogram: an analogue tool to deliver digital knowledge. J Thorac Cardiovasc Surg. 2018;155(4):1793.
16. Maeser D, Gruener RF, Huang RS. oncoPredict: an R package for predicting invivo or cancer patient drug response and biomarkers from
cell line screening data. Brief Bioinform. 2021;22(6):260.
17. Lorent M, Giral M, Foucher Y. Net time -dependent ROC curves: a solution for evaluating the accuracy of a marker to predict disease-related
mortality. Stat Med. 2014;33(14):2379–89.
18. Holleczek B, Brenner H. Model based period analysis of absolute and relative survival with R: data preparation, model tting and deriva-
tion of survival estimates. Comput Methods Programs Biomed. 2013;110(2):192–202.
19. Han X, Vesely MD. Stimulating T cells against cancer with agonist immunostimulatory monoclonal antibodies. Int Rev Cell Mol Biol.
2019;342:1–25.
20. Fabian KP, Wolfson B, Hodge JW. From immunogenic cell death to immunogenic modulation: select chemotherapy regimens induce a
spectrum of immune-enhancing activities in the tumor microenvironment. Front Oncol. 2021;11:728018.
21. Escors D. Tumour immunogenicity, antigen presentation, and immunological barriers in cancer immunotherapy. New J Sci. 2014. https://
doi. org/ 10. 1155/ 2014/ 734515.
22. Muenst S, Läubli H, Soysal SD, Zippelius A, Tzankov A, Hoeller S. The immune system and cancer evasion strategies: therapeutic concepts.
J Intern Med. 2016;279(6):541–62.
23. Pan XB, Lu Y, Huang JL, Long Y, Yao DS. Prognostic genes in the tumor microenvironment in cervical squamous cell carcinoma. Aging.
2019;11(22):10154.
24. Mairinger F, Bankfalvi A, Schmid KW, Mairinger E, Mach P, Walter RF, etal. Digital immune-related gene expression signatures in high-grade
serous ovarian carcinoma: developing prediction models for platinum response. Cancer Manag Res. 2019;11:9571–83.
25. Yang S, Wu Y, Deng Y, Zhou L, Yang P, Zheng Y, etal. Identication of a prognostic immune signature for cervical cancer to predict survival
and response to immune checkpoint inhibitors. OncoImmunology. 2019;8(12):e1659094.
26. Shen L, Yu H, Liu M, Wei D, Liu W, Li C, etal. A ten-long non-coding RNA signature for predicting prognosis of patients with cervical cancer.
OncoTargets Ther. 2018;11:6317–26.
27. He RQ, Huang ZG, Li TY, Wei YP, Chen G, Lin XG, etal. RNA-sequencing data reveal a prognostic four-lncRNA-based risk score for bladder
urothelial carcinoma: an in silico update. Cell Physiol Biochem. 2018;50(4):1474–95.
28. Wang L, Han S, Jin G, Zhou X, Li M, Ying X, etal. Linc00963: a novel, long non-coding RNA involved in the transition of prostate cancer
from androgen-dependence to androgen-independence. Int J Oncol. 2014;44(6):2041–9.
29. Hu Q, Egranov SD, Lin C, Yang L. Long noncoding RNA loss in immune suppression in cancer. Pharmacol Ther. 2020;213:107591.
30. Mercer TR, Dinger ME, Mattick JS. Long non-coding RNAs: insights into functions. Nat Rev Genet. 2009;10(3):155–9.
31. Yu T, Zhao Y, Hu Z, Li J, Chu D, Zhang J, etal. MetaLnc9 facilitates lung cancer metastasis via a PGK1-activated AKT/mTOR pathway. Cancer
Res. 2017;77(21):5782–94.
32. Wu Z, Wang W, Wang Y, Wang X, Sun S, Yao Y, etal. Long noncoding RNA LINC00963 promotes breast cancer progression by functioning
as a molecular sponge for microRNA-625 and thereby upregulating HMGA1. Cell Cycle. 2020;19(5):610–24.
33. Luo X, Donnelly CR, Gong W, Heath BR, Hao Y, Donnelly LA, etal. HPV16 drives cancer immune escape via NLRX1-mediated degradation
of STING. J Clin Invest. 2020;130(4):1635–52.
34. Badowski C, He B, Garmire LX. Blood-derived lncRNAs as biomarkers for cancer diagnosis: the good, the bad and the beauty. NPJ Precis
Oncol. 2022;6(1):40.
Publisher’s Note Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional aliations.
ResearchGate has not been able to resolve any citations for this publication.
Article
Full-text available
Cancer ranks as one of the deadliest diseases worldwide. The high mortality rate associated with cancer is partially due to the lack of reliable early detection methods and/or inaccurate diagnostic tools such as certain protein biomarkers. Cell-free nucleic acids (cfNA) such as circulating long noncoding RNAs (lncRNAs) have been proposed as a new class of potential biomarkers for cancer diagnosis. The reported correlation between the presence of tumors and abnormal levels of lncRNAs in the blood of cancer patients has notably triggered a worldwide interest among clinicians and oncologists who have been actively investigating their potentials as reliable cancer biomarkers. In this report, we review the progress achieved (“the Good”) and challenges encountered (“the Bad”) in the development of circulating lncRNAs as potential biomarkers for early cancer diagnosis. We report and discuss the diagnostic performance of more than 50 different circulating lncRNAs and emphasize their numerous potential clinical applications (“the Beauty”) including therapeutic targets and agents, on top of diagnostic and prognostic capabilities. This review also summarizes the best methods of investigation and provides useful guidelines for clinicians and scientists who desire conducting their own clinical studies on circulating lncRNAs in cancer patients via RT-qPCR or Next Generation Sequencing (NGS).
Article
Full-text available
Cancer treatment has rapidly entered the age of immunotherapy, and it is becoming clear that the effective therapy of established tumors necessitates rational multi-combination immunotherapy strategies. But even in the advent of immunotherapy, the clinical role of standard-of-care chemotherapy regimens still remains significant and may be complementary to emerging immunotherapeutic approaches. Depending on dose, schedule, and agent, chemotherapy can induce immunogenic cell death, resulting in the release of tumor antigens to stimulate an immune response, or immunogenic modulation, sensitizing surviving tumor cells to immune cell killing. While these have been previously defined as distinct processes, in this review we examine the published mechanisms supporting both immunogenic cell death and immunogenic modulation and propose they be reclassified as similar effects termed “immunogenic cell stress.” Treatment-induced immunogenic cell stress is an important result of cytotoxic chemotherapy and future research should consider immunogenic cell stress as a whole rather than just immunogenic cell death or immunogenic modulation. Cancer treatment strategies should be designed specifically to take advantage of these effects in combination immunotherapy, and novel chemotherapy regimens should be designed and investigated to potentially induce all aspects of immunogenic cell stress.
Article
Full-text available
Background: The predictive value of soluble Klotho (sKlotho) for adverse outcomes in patients on maintenance hemodialysis (MHD) is controversial. In this study, we aimed to clarify the potential association of sKlotho levels with adverse outcomes in this patient population. Materials: A total of 211 patients on MHD were identified and stratified according to the median sKlotho level. Patients were followed up for adverse outcomes including cardiovascular (CV) morbidity and all-cause mortality. Results: During the 36-month follow-up, 75 patients [51 CV events (including 16 CV deaths) and 40 deaths] experienced adverse outcomes. After stratification according to median sKlotho level, patients with a lower sKlotho level had a greater risk of CV events (38.2% vs. 19.5%, p = 0.006), all-cause mortality (28.4% vs. 11.6%, p = 0.003), and combined adverse outcomes (51.0% vs. 24.2%, p < 0.001). Similar observations were made from analyses using Kaplan-Meier survival curves. Cox regression analysis showed that a low sKlotho level was strongly correlated with CV morbidity [1.942 (1.030-3.661), p = 0.040)], all-cause mortality [2.073 (1.023-4.203), p = 0.043], and combined adverse outcomes [1.818 (1.092-3.026), p = 0.021] in fully adjusted models. Conclusions: The sKlotho level was an independent predictive factor of adverse outcomes including CV morbidity and mortality in patients on MHD.
Article
Full-text available
Background: This study was to compare the efficacy and safety between neoadjuvant chemotherapy followed by radical surgery (NACT+RS) and radiotherapy only (RT) or concurrent chemoradiotherapy (CCRT) for treatment of patients with stage IB2, IIA, or IIB cervical cancer. Method: The electronic databases of PubMed, Embase, and the Cochrane Library were searched to screen relevant studies from their inception to October 2018. Clinical data including overall survival (OS), disease-free survival (DFS), and adverse events were extracted. Egger's test was used to evaluate the publication bias, and sensitivity analysis was conducted to estimate the robustness of results. Results: Finally, three randomized controlled trials (RCTs) and two case-control studies consisting of 1,275 patients with stage IB2, IIA, or IIB cervical cancer were included in the current study. Overall, pooled results showed no significant differences in OS ((hazard ratio (HR) = 0.603, 95%CI = 0.350 - 1.038) and DFS (HR = 0.678, 95%CI = 0.242 - 1.904) for patients treated with NACT+RS compared with RT only or CCRT, but the subgroup analysis showed that the OS and DFS were significantly longer in the NACT+RS groups than the RT or CCRT group (OS: HR = 0.431, 95%CI = 0.238 - 0.781, p = 0.006; DFS: HR = 0.300, 95%CI = 0.187 - 0.482, p < 0.001) for the population with median follow-up time of more than 60 months. For adverse events, the incidence of thrombocytopenia in the NACT+RS group was significantly higher than that in the RT only or CCRT group (relative risk (RR) = 3.240, 95% CI 1.575-6.662), while the incidence of diarrhea was significantly lower than that in the RT only or CCRT group (RR = 0.452, 95% CI =0.230-0.890). Conclusion: These findings suggest that the short-term therapeutic effects of the two treatments may be possibly equal for patients with stage IB2-IIB cervical cancer, but the long-term effects for improving OS and DFS may be better using NACT+RS compared with the RT only or CCRT.
Article
Full-text available
Long noncoding RNAs (lncRNAs) have been found to associate with all major types of malignancies and play important roles in regulating several hallmarks of cancer by interacting with proteins, DNA, and RNA. The possible functions of lncRNAs and their roles in the regulation of tumour growth will be reported and discussed in the present review. In our recent report, based on genetic mice models and a series of systematic analyses, we suggested that lncRNAs also play critical roles in the regulation of antigen presentation in tumour cells and allow tumour cells to escape immune surveillance, which further broadens the scope of understanding lncRNA functions and how they relate to cancer immunotherapy resistance.
Article
Cell line drug screening datasets can be utilized for a range of different drug discovery applications from drug biomarker discovery to building translational models of drug response. Previously, we described three separate methodologies to (1) correct for general levels of drug sensitivity to enable drug-specific biomarker discovery, (2) predict clinical drug response in patients and (3) associate these predictions with clinical features to perform in vivo drug biomarker discovery. Here, we unite and update these methodologies into one R package (oncoPredict) to facilitate the development and adoption of these tools. This new OncoPredict R package can be applied to various in vitro and in vivo contexts for drug and biomarker discovery.
Article
Long noncoding RNAs (lncRNAs) have multiple functions in the regulation of cellular homeostasis. In recent years, numerous studies have shown that tumor-associated lncRNAs play key roles in promoting and maintaining tumor initiation and progression by shaping the tumor microenvironment through changing tumor cell intrinsic properties. Here, we focus on the roles of lncRNAs in cancer immunology. In the first part, we provide an overview of the roles played by lncRNAs and their deregulation in cancer at the cancer cell- and tumor microenvironment-associated immune cell levels. We go on to describe preclinical strategies for targeting lncRNAs, particularly highlighting the effects on tumor microenvironments. We then discuss the possibility of combining lncRNA targeting and tumor immune checkpoint inhibitor antibodies to treat cancer.
Article
Objective To compare FIGO 2009 and FIGO 2018 cervical cancer staging criteria with a focus on stage migration and treatment outcomes. Methods This study is based on a database cohort of 1282 patients newly diagnosed with cervical cancer from 1997 to 2019. All underwent standard clinical examination and whole-body FDG-PET. Tumor stage was recorded using the FIGO 2009 system, which excluded surgical pathologic, FDG-PET and other advanced imaging findings, and then re-classified to the FIGO 2018 system, including surgical pathologic and imaging findings. Patient management was based on clinical, surgical, and imaging findings. Stage migration and prognosis were evaluated. Results The distribution per the 2009 staging system was stage I in 593 (46%), stage II in 342 (27%), stage III in 263 (21%), and stage IV in 84 (7%) and the 2018 staging system was stage I in 354 (28%), stage II in 156 (12%), stage III in 601 (47%), and stage IV in 171 (13%). No patients were down-staged. Stage migration occurred in 53% (676/1282) and was attributable to detection of occult lymph node metastasis in 520 (41%), occult distant metastasis in 90 (7%), and tumor size and extent in 66 (5%). The 5-year progression-free survivals (PFS) by FIGO 2009 versus FIGO 2018 were as follows: stage I, 80% vs. 87% (p = 0.02); stage II, 59% vs. 71% (p = 0.002); stage III, 35% vs. 55% (p < 0.001), and stage IV, 20% vs. 16% (p = 0.41). Conclusion Inclusion of surgical pathologic and imaging findings resulted in upward stage migration in the majority, mostly related to nodal and distant metastasis. While FIGO 2018 improves survival discriminatory ability for stages I and IV patients, survival remains heterogeneous among stage III substages.
Article
Purpose: Adenocarcinoma (AC) of the uterine cervix is the second most common type of cervical cancer after squamous cell carcinoma (SCC). Although both subtypes are treated similarly, patients with AC have a worse prognosis. In this study, immunologic features of the tumor microenvironment in these two subsets were pursued with potential therapeutic implications. Experimental design: The immune microenvironment of primary tumors (PT) and non-metastatic tumor-draining lymph nodes (TDLN) was compared between patients with cervical AC (n = 16) and SCC (n = 20) by polychromatic flow cytometry and by transcriptional profiling of the PT (n = 299) using publicly available data from The Cancer Genome Atlas (TCGA). Results: Flowcytometric analyses revealed intact T-cell differentiation in TDLN but hampered effector T-cell trafficking to the PT in AC, as compared to SCC. TCGA analysis demonstrated higher expression of chemokines involved in effector T-cell homing (CXCL9/10/11) in SCC PT as compared to AC PT, which was highly correlated to a transcriptional signature for type 1 conventional dendritic cells (cDC1). This was consistent with elevated frequencies of CD141+/BDCA3+ cDC1 in PT SCC samples relative to AC and correspondingly elevated levels of CXCL9 and CXCL10 in 24h ex-vivo cultures. Hampered cDC1 recruitment in AC was in turn related to lower transcript levels of cDC1-recruiting chemokines and an elevated β-catenin activation score, and was associated with poor overall survival. Conclusions: Our data have identified an opportunity for the investigation of potentially novel therapeutic interventions in AC of the cervix, i.e. β-catenin inhibition and cDC1 mobilization.
Article
Extensive research has shown that LINC00963 is aberrantly expressed in human cancers, and that dysregulation of LINC00963 is implicated in the initiation and progression of human cancers. The expression and functions of LINC00963 in breast cancer are still unclear. Our aims were to measure the expression of LINC00963 in breast cancer, determine its effects on malignant behaviors of tumor cells, and uncover the molecular events underlying the actions of LINC00963 in breast cancer. Herein, LINC00963 was found to be overexpressed in breast cancer samples, and its overexpression was correlated with lymph node metastasis, TNM stage and differentiation grade. Patients with breast cancer harboring higher LINC00963 expression showed shorter overall survival than did the patients with lower LINC00963 expression. Functional experiments revealed that depletion of LINC00963 inhibited breast cancer cell proliferation, migration, and invasion and facilitated apoptosis in vitro and impaired tumor growth in vivo. Mechanism investigation revealed that LINC00963 can interact with microRNA-625 (miR-625). LINC00963 worked as a competitive endogenous RNA for miR-625 to weaken the suppressive effect of miR-625 on high mobility group AT-hook 1 (HMGA1) in breast cancer cells. Furthermore, miR-625 inhibition and HMGA1 restoration both abrogated the effects of LINC00963 silencing on breast cancer cells. Our findings indicate that the LINC00963–miR-625–HMGA1 pathway plays an important role in the malignancy of breast cancer in vitro and in vivo. Hence, targeting this pathway may be a novel strategy against breast cancer.