Abstract
Osteosarcoma is generally considered a cold tumor and is characterized
by epigenetic alterations. Although tumor cells are surrounded by many
immune cells such as macrophages, T cells may be suppressed, be
inactivated, or not be presented due to various mechanisms, which
usually results in poor prognosis and insensitivity to immunotherapy.
Immunotherapy is considered a promising anti-cancer therapy in
osteosarcoma but requires more research, but osteosarcoma does not
currently respond well to this therapy. The cancer immunity cycle (CIC)
is essential for anti-tumor immunity, and is epigenetically regulated.
Therefore, it is possible to modulate the immune microenvironment of
osteosarcoma by targeting epigenetic factors. In this study, we
explored the correlation between epigenetic modulation and CIC in
osteosarcoma through bioinformatic methods. Based on the RNA data from
TARGET and [42]GSE21257 cohorts, we identified epigenetic related
subtypes by NMF clustering and constructed a clinical prognostic model
by the LASSO algorithm. ESTIMATE, Cibersort, and xCell algorithms were
applied to analyze the tumor microenvironment. Based on eight
epigenetic biomarkers (SFMBT2, SP140, CBX5, HMGN2, SMARCA4, PSIP1,
ACTR6, and CHD2), two subtypes were identified, and they are mainly
distinguished by immune response and cell cycle regulation. After
excluding ACTR6 by LASSO regression, the prognostic model was
established and it exhibited good predictive efficacy. The risk score
showed a strong correlation with the tumor microenvironment, drug
sensitivity and many immune checkpoints. In summary, our study sheds a
new light on the CIC-related epigenetic modulation mechanism of
osteosarcoma and helps search for potential drugs for osteosarcoma
treatment.
Subject terms: Bone cancer, Cancer microenvironment, Tumour immunology,
Epigenomics
Introduction
Osteosarcoma (OS) is one of the most common bone tumors occurring
predominantly in children and adolescents. Another prone population is
the people with age > 60 years, which is generally correlated with
Paget’s disease of bone^[43]1,[44]2. As a solid tumor with a high level
of heterogeneity, osteosarcoma possesses a complex immune environment.
Macrophages are the primary immune cells surrounding the malignant
cells, and other innate immune cells, such as T-lymphocytes, dendritic
cells, mast cells, neutrophils, and B cells, also exhibit a certain
level of infiltration^[45]3,[46]4. However, T cells may not be present
or may be suppressed by various mechanisms involving the lack of T-cell
clonal diversity and low expression of programmed death ligand (PDL) in
the tumor^[47]5,[48]6. Due to this specific immune microenvironment,
osteosarcoma generally is considered a “cold” tumor, which results in a
poor response to immunotherapy^[49]7. Immunotherapy is an emerging and
promising therapy method utilizing the immune system to battle
cancer^[50]8. Immune checkpoint blockade (ICB) is typical of
immunotherapy, which achieves unprecedented advances in cancer
treatment. PD-L1, a vital target for ICB, was found expressed in
osteosarcoma cell lines and correlated with drug resistance of
osteosarcoma. In addition, anti-PD1 and anti-PD-L1 therapy exhibited
good effectiveness in an osteosarcoma mouse model^[51]9. Hence, ICB is
considered a potential promising therapy for osteosarcoma patients.
However, the application of ICB therapy in clinical treatment encounter
some obstacles. Only a small subset of patients benefit from ICB and
immune-related adverse events (irAEs) occur in partial patients^[52]10.
In order to improve the effectiveness and application extent of
immunotherapy, there is an urgent need to realize the regulation
mechanism of anti-cancer immunity. The cancer immunity cycle (CIC) is
defined as a series of stepwise events required for anti-cancer
immunity, and the therapy targeting these steps is a strategy favoring
immunotherapy^[53]11,[54]12. Firstly, dendritic cells capture cancer
antigens released from tumor cells and present them to naïve T cells
that prime and activate effector T cells. Then the effector T cells
infiltrate the tumor microenvironment (TME) and kill tumor cells.
Finally, cancer antigens are released again and the next cycle
starts^[55]13. The dysregulation of CIC can result in the immune escape
and survival of tumor cells^[56]13,[57]14. In order to accelerate the
development and application of immunotherapy in osteosarcoma, it is
essential to explore the regulatory mechanism of CIC.
Epigenetic factors affect tumors not only by regulating the activities
of tumor cells but also through impacting TME^[58]15. Epigenetic
modification, comprising DNA methylation, histone modification,
nucleosome remodeling, RNA modification, and non-coding RNA regulation,
is a critical regulation mechanism in phenotypic alteration without
changes in DNA sequences^[59]16,[60]17. Osteosarcoma displays global
hypomethylation and focal hypermethylation at CpG islands^[61]18.
Abnormal DNA methylation is able to regulate RNA expression levels. For
instance, TSSC3, a pro-apoptosis gene, is silenced in osteosarcoma
cells due to the hypermethylation at promoter regions^[62]19. Histone
modification refers to the post-translational modification of histone
protein tails, which can impact nucleosome dynamics, and
transcription^[63]20. Many enzymes modifying histone proteins are
involved in the tumorigenesis of osteosarcoma. Histone demethylase
KDM4A is upregulated in osteosarcoma and alleviates tumor cell
ferroptosis through mediating H3K9me3 demethylation in the promoter
region of SLC7A11^[64]21. Histone acetyltransferase HBO1 is increased
in osteosarcoma and serves as an oncogene^[65]22. RNA modification and
non-coding RNA contribute to osteosarcoma progression via regulating
the translation of mRNA to proteins. N6-methylation (m6A) is the most
common RNA modification in tumors, and various m6A related enzymes play
a regulatory role in osteosarcoma. ALKBH5, a type of m6A eraser, is
found to inactive STAT3 pathway in osteosarcoma through decreasing m6A
modification of SOCS3^[66]23. These enzymes also can interact with
non-coding RNAs. PVT1, a vital oncogenic long noncoding RNA (lncRNA),
is regulated by ALKBH5 via a m6A-depended manner^[67]24. Interestingly,
epigenetic regulation also plays a vital role in the immune system, and
the therapy targeting epigenetic modulation is considered a meaningful
complement to immunotherapy^[68]25. It was found that immunomodulatory
pathway genes were concentrated in late-replicating partial methylation
domains with DNA methylation loss^[69]26. A pan-cancer analysis also
revealed that tumor immunogenicity was inversely associated with
methylation aberrancy^[70]27. Wholescale epigenetic remodeling was
observed in exhausted T cells that weakens the response to
immunotherapy^[71]28. Epigenetic therapy not only directly inhibits
tumors, but also assists the function of immunotherapy^[72]29. Several
epigenetic drugs have been approved for anti-cancer treatment by the
Food and Drug Administration (FDA) of America, including DNMT
inhibitors, HDAC inhibitors, and so on^[73]30. Many studies found that
a part of epigenetic drugs can work as inducers of tumor immunogenic
cell death (ICD)^[74]31. Besides, HDAC inhibitors were found to
increase the expression levels of CTLA-4, GITR, and PD-1 in Treg
cells^[75]32. Although increasing clinical data suggest the great
potential of the combination of epigenetic drugs and immunotherapy,
there remain a series of obstacles in the clinical implementation of
this combination therapy, such as the ubiquitous distribution of
epigenetic targets in normal and tumor cells^[76]33.
In osteosarcoma, the interaction between epigenetic modulation and
immune response remains largely unexplored, and the value of
combination therapy including epigenetic modulators and immunotherapy
requires further evaluation. Hence, we explored the correlation between
CIC events and epigenetic factors in osteosarcoma, and identified two
molecular subtypes with different epigenetic and immune
characteristics, based on which a clinical risk model was constructed
to predict the clinical outcomes of osteosarcoma patients.
Material and methods
Data source and process
The current study collected 801 epigenetic factor encoding genes (Table
[77]S1) from EpiFactors Database
([78]https://epifactors.autosome.org/). In this database, epigenetic
factors are defined as proteins and lncRNAs involved in epigenetic
regulation. Protein factors consist of modulators of chromatin,
cofactors forming complex with epigenetic factors, histones and
corresponding variants, protamines, histone chaperones, and DNA/RNA
modification regulators^[79]34.
Two cohort datasets were included in this study. The RNA expression
data and corresponding clinical data of 95 patients were downloaded
from the TARGET database ([80]https://ocg.cancer.gov/programs/target).
The [81]GSE21257 dataset (Platform: [82]GPL10295), containing gene
expression data and clinical data of 53 patients, was downloaded from
the Gene Expression Omnibus public database (GEO:
[83]https://www.ncbi.nlm.nih.gov/geo/). The two datasets were merged
and then applied to clustering. The batch effect was corrected by the
“sva” R package. In the construction of prognostic model, the TARGET
dataset was set as a train cohort, and the [84]GSE21257 dataset was
used as a validation cohort. The RNA expression data of 148 samples was
presented in Table [85]S2, and corresponding clinical information was
shown in Table [86]S3.
Identification of CIC-related epigenetic factors with prognostic value
Using Tracking Tumor Immunophenotype database (TIP:
[87]http://biocc.hrbmu.edu.cn/TIP/), CIC score, an indicator to
represent the activity levels of CIC steps, was calculated based on RNA
expression data of 148 osteosarcoma samples (Table [88]S4). TIP
database provides a user-friendly web tool to evaluate and visualize
the activity of anticancer immunophenotypes^[89]35. The correlations
between CIC score and epigenetic factors were estimated by Pearson
test. The correlations with P < 0.05 and |R|> 0.3 were considered
significant, which were shown in Table [90]S5 and were visualized using
Cytoscape 3.8. We then screened the CIC-related epigenetic factors that
were associated with prognosis by univariate Cox analysis. The
epigenetic factors with P < 0.05 were considered significant (Table
[91]S6).
Molecular subtypes of osteosarcoma
In this study, non-negative matrix factorization (NMF) was applied to
clustering the molecular subtypes of osteosarcoma samples. NMF R
package was used in the clustering, where the method was set as
“brunet”, the rank was set as 2–10, and the iteration number was set as
1000. The optimal rank was determined by cophenetic, dispersion and
silhouette indicators. Kaplan–Meier survival curves were drawn to
evaluate the prognostic value of this clustering. Furthermore,
principal component analysis (PCA) was performed to validate if this
clustering method could distinguish osteosarcoma samples with different
molecular features. PCA is an unsupervised learning method that can
transform high-dimensional data into fewer dimensions, which works for
capturing features^[92]36.
Functional and pathway enrichment analysis
In order to deeply analyze the differences between osteosarcoma samples
from different molecular subtypes, functional and pathway enrichment
analysis were performed based on the Kyoto encyclopedia of genes and
genomes (KEGG) database and the Gene ontology (GO) database. Gene set
variation analysis (GSVA) was applied to explore the enriched pathways
based on the KEGG database. Gene set enrichment analysis (GSEA) was
applied to explore the functional difference based on the GO database.
Enrichment analysis was performed in R software, and R packages “GSVA”
and “clusterProfiler” were adopted in the analysis.
The TME landscape of molecular subtypes of osteosarcoma
The TME landscape of osteosarcoma was portrayed by “estimate”,
“cibersort” and “xCell” R packages. Firstly, using “estimate” R
package, we calculated stromal score, immune score and tumor purity,
which reflected the content of stromal cells, immune cells, and tumor
cells respectively. Then, we calculated the priority of 22 immune cells
for each sample by “cibersort” R package. Finally, we calculated the
enrichment score for 64 immune cells and stromal cells in osteosarcoma
via “xCell” R package. The abbreviations of these cell types were
provided in the “Abbreviation Section”.
In addition, we compared RNA expression levels of immune checkpoint
genes in different molecular subtypes and evaluated immune features for
each sample. Immune checkpoint genes were summarized by some recent
studies^[93]37,[94]38. The levels of immune features were estimated by
single sample gene set enrichment analysis (ssGSEA).
Construction and validation of clinical risk model and nomogram
Least absolute shrinkage and selection operator regression (LASSO) is a
regression analysis method which can be used to exclude irrelevant
variables and consequently downscale data. In the current study,
LASSO-Cox regression was applied to screen critical genes and construct
a clinical prognostic model. The TARGET cohort (n = 95) was set as a
train cohort, and the [95]GSE21257 cohort (n = 53) was set as
a validation cohort. The optimal value of tuning parameter (lamda) was
determined by ten-time cross validation using minimum criteria. The
risk score was calculated by the formula:
[MATH: RiskScore=∑iCo
efficien
ti×Expressionofgenei :MATH]
. According to the median value of risk score (− 0.0835), patients were
divided into a high risk group and a low risk group. Kaplan–Meier curve
and Receiver operating characteristic curve (ROC) were plotted to
identify the prognostic value of the risk score.
To predict 1-, 3-, and 5- years survival probabilities, we generated a
nomogram using a R package “rms”. Risk score and three clinical
features were adopted in the construction of a nomogram. Clinical
features included age, gender, and metastasis status. Correction curves
were plotted to compare prediction accuracy between the observed and
model-predicted outcomes. Furthermore, ROC was performed to evaluate
the efficiency of the nomogram.
Comprehensive analysis for the role of risk score in osteosarcoma
In order to comprehensively understand the role of risk score in
osteosarcoma, we explored the correlations of risk score with clinical
characteristics, immune characteristics, drug sensitivity, and response
to immunotherapy.
The clinical characteristic information of osteosarcoma includes age,
gender, histologic response, and metastasis status, among which the
information of histologic response is incomplete. Immune
characteristics were evaluated using the infiltration of immune cells,
and the expression levels of immune modulator genes (immune inhibitor
genes and immune stimulator genes). The immune modulator genes were
summarized by the TISIDB database
([96]http://cis.hku.hk/TISIDB/index.php^[97]39. The drug sensitivity
was defined by the half maximal inhibitory concentration (IC50). The R
package “oncoPredict” provides an algorithm to estimate IC50 of drugs
based on the gene expression data, which was used in this study to
calculate IC50 of drugs for every osteosarcoma patient using drug data
from Genomics of Drug Sensitivity in Cancer (GDSC) as train data. The
lower IC50 means higher drug sensitivity. Tumor inflammation signature
score (TIS) and Immunophenoscore (IPS) were applied to indicate the
response to immunotherapy. IPS is a scoring system used to demonstrate
immune function, including antigen presentation, effector cells,
checkpoints, and suppressor cells. IPS consists of four categories, MHC
molecules IPS (MHC_IPS), immunomodulators IPS (CP_IPS), effector cells
IPS (EC_IPS), and suppressor cells IPS (SC_IPS), which were calculated
based on the RNA expressions of corresponding biomarkers^[98]40. In our
study, the TIS score was calculated using GSVA tools, and the IPS was
calculated through “IOBR” R package.
Statistical analysis
All statistical analysis in our study was performed in R software
(version 4.1.3). The prognostic values of CIC-related epigenetic
factors were estimated using univariate Cox analysis. The Wilcoxon test
was used to compare the differences between the two molecular subtypes,
and the differences in clinical features between high and low risk
groups. Chi-squared test was used to check if there was a significant
difference of risk distribution within groups with different clinical
features. Spearman correlation coefficients were employed to evaluate
the associations between immune characteristics, drug sensitivity,
response to immunotherapy, and risk score.
Result
Identification of CIC-related epigenetic factors with prognostic value in
osteosarcoma
Figure [99]1 presents the design of our research. Firstly, we
identified CIC-related epigenetic factors with prognostic value, and
then we selected significant genes to cluster osteosarcoma and
construct a clinical risk model. The clinical information of 148
patients from the TARGET cohort and the [100]GSE21257 cohort were
summarized in Table [101]1.
Figure 1.
[102]Figure 1
[103]Open in a new tab
The flowchart of this study.
Table 1.
The clinical information of TARGET cohort and [104]GSE21257 cohort.
TARGET_OS (N = 95) [105]GSE21257 (N = 53) Overall (N = 148)
Sex
Male 55 (57.9%) 34 (64.2%) 89 (60.1%)
Female 40 (42.1%) 19 (35.8%) 59 (39.9%)
Age (years)
Mean (SD) 15.4 (5.32) 18.7 (12.2) 16.6 (8.56)
Median [Min, Max] 14.8 [3.56, 39.9] 16.7 [3.08, 79.0] 15.3 [3.08,
79.0]
Follow-up to main event (years)
Mean (SD) 4.00 (3.00) 5.71 (4.94) 4.62 (3.88)
Median [Min, Max] 3.33 [0.203, 16.0] 3.75 [0.333, 20.5] 3.48 [0.203,
20.5]
Status
Alive 57 (60.0%) 30 (56.6%) 87 (58.8%)
Dead 38 (40.0%) 23 (43.4%) 61 (41.2%)
Metastasis
Yes 23 (24.2%) 34 (64.2%) 57 (38.5%)
No 72 (75.8%) 19 (35.8%) 91 (61.5%)
Histologic response
Stage 1/2 28 (29.5%) 29 (54.7%) 57 (38.5%)
Stage 3/4 16 (16.8%) 18 (34.0%) 34 (23.0%)
Unknown 51 (53.7%) 6 (11.3%) 57 (38.5%)
[106]Open in a new tab
CIC steps were quantified by the TIP web tool in the form of a score,
which is exhibited in Fig. [107]2A. Based on the CIC score and RNA
expression data, the correlation between CIC steps and epigenetic
factors was identified. In total, 149 epigenetic factors were
identified associated with CIC (P < 0.05, |R|> 0.3), which were
presented in Fig. [108]2B. Then we further screened prognosis related
genes among CIC-related epigenetic factors. Finally, eight epigenetic
factors were found highly correlated with prognosis. As shown in
Fig. [109]2C, SFMBT2, SP140, CBX5, and HMGN2 were identified as
protective factors, whereas SMARCA4, PSIP1, ACTR6, and CHD2 were
identified as risk factors. An interaction network for these epigenetic
factors was constructed. Eight CIC-related factors with prognostic
value were surrounded by 20 genes, among which eight genes exhibited
high correlation with CHD family genes (CHD1, CHD6, CHD7, CHD8, CHD9),
CBX family genes (CBX1, CBX3, CBX8) and HMGN family genes (HMGN1,
HMGN3, HMGN4) (Fig. [110]2D).
Figure 2.
[111]Figure 2
[112]Open in a new tab
Identification of CIC-related epigenetic factors with prognostic value.
(A) The heatmap of CIC score of osteosarcoma samples. (B) The network
diagram of CIC events and epigenetic factors. Red represented CIC
events, and green represented epigenetic factors. (C) The forest plot
of eight epigenetic factors with prognostic value. (D) The network plot
of epigenetic factors with prognostic value using the GeneMANIA online
tool.
Identification and comprehensive analysis of molecular subtypes of
osteosarcoma
NMF model was used to cluster osteosarcoma patients. Rank 2 was
selected as the optimal cluster number as it exhibited the most
significant decrease in the cophenetic correlation coefficient
(Fig. [113]3A). 148 samples were divided into 2 subtypes. Cluster 1
contains 53 patients, and cluster 2 contains 95 patients. As shown in
Fig. [114]3B, this clustering could distinguish osteosarcoma samples.
The patients in cluster 1 showed better prognosis compared with cluster
2 (Fig. [115]3C, P < 0.001). Finally, PCA was applied to validate the
accuracy of this clustering. 3D dot plot presented that the patients of
osteosarcoma were clearly separated (Fig. [116]3D). The subtype
information is presented in Table [117]S7.
Figure 3.
[118]Figure 3
[119]Open in a new tab
Molecular clustering of osteosarcoma based on CIC-related epigenetic
factors. (A, B) The rank curve and heatmap of NMF clustering. (C) KM
curve of osteosarcoma patients in different molecular subtypes. (D) 3D
dot plot of PCA analysis. (E) The plot of GSVA analysis based on the
KEGG database. The blue columns present the top 15 KEGG pathways with
the highest t-value, and the green columns present the top 15 KEGG
pathways with the lowest t-value. (F) The result of GSEA based on the
GO database. The top 5 terms of cellular component (CC), molecular
function (MF), and biological process (BP) enriched in Cluster 1 and
Cluster 2 were presented.
To reveal the functional difference between cluster 1 and cluster 2,
GSVA and GSEA analysis were performed. GSVA presented that the
molecular subtype of osteosarcoma mainly differed in immune related
pathways. “Primary immunodeficiency”, “chemokine signaling pathway” and
“leishmania infection” were upregulated in cluster1, whereas “cell
cycle”, “one carbon pool by folate” and “steroid biosynthesis” were
upregulated in cluster 2 (Fig. [120]3E). The top 5 terms of cellular
component (CC), molecular function (MF), and biological process (BP)
enriched in cluster 1 and cluster 2 were shown in Fig. [121]3F. The
total results of GSVA and GSEA were presented in Table [122]S8 and
[123]S9 respectively.
The TME landscape of osteosarcoma samples were analyzed by “ESTIMATE”,
“Cibersort” and “xCell” algorithms. “ESTIMATE” algorithm provided
stromal score, immune score, ESTIMATE score, and tumor purity, which
were exhibited in Fig. [124]4A. Stromal score, immune score and
ESTIMATE score were higher in cluster 1, whereas tumor purity was
higher in cluster 2. Figure [125]4B shows the abundance of 22 immune
cells in 148 osteosarcoma samples. As shown in this figure, compared
with other immune cells, M2 and M0 macrophages were the immune cells
with the highest proportion. Based on the result of the “xCell”
algorithm, 64 stromal cells and immune cells were compared between
cluster 1 and cluster 2. 29 cells presented differences between cluster
1 and cluster 2 (Fig. [126]4C). In order to further explore the
differences of molecular subtypes in the immune system, immune
checkpoint genes, and immune features were researched. 17 immune
checkpoint genes were differentially expressed in molecular subtypes,
among which IFGN, HAVCR2, CD8A, TNF, PDCD1, VTCN1, GZMA, GZMB, CTLA4,
LAG3, CD274, TLR4, PRF1, CX3CL1, CXCL9, CXCL10, EDNRB and CD276 were
the most significant genes (P < 0.0001) (Fig. [127]4D). Except for type
II IFN response, 12 immune features exhibited differences between
cluster 1 and cluster 2 (Fig. [128]4E). The results of “ESTIMATE”,
“Cibersort”, “xCell” and immune features were exhibited in Table
[129]S10.
Figure 4.
[130]Figure 4
[131]Open in a new tab
The TME landscape in osteosarcoma. (A) Difference analysis of stromal
score, immune score, ESTIMATE score, and tumor purity in cluster 1 and
cluster 2. (B) The abundance of 22 immune cells in 148 osteosarcoma
samples based on the “CIBERSORT” algorithm. The length of the bar plot
represents the relative abundance of immune cells in each sample. (C)
Difference analysis of 64 stromal and immune cells in cluster 1 and
cluster 2 based on “xCell” algorithm. (D) Different expression of
immune checkpoint genes in cluster 1 and cluster 2. (E) Difference
analysis of immune features in cluster 1 and cluster 2. *P < 0.05,
**P < 0.01, ***P < 0.001, ****P < 0.0001.
The construction and validation of clinical risk prognostic model
Based on the RNA expression data and clinical information of the TARGET
cohort, the LASSO Cox regression algorithm was applied to extract the
most critical genes. Seven epigenetic factors were identified as
optimal gene signatures, including PSIP1, CHD2, SMARCA4, HMGN2, SP140,
CBX5, and SFMBT2 (Fig. [132]5A–C). Among seven factors, PSIP1, CHD2,
and SMARCA4 were risk gene signatures, whereas HMGN2, SP140, CBX5, and
SFMBT2 were protective gene signature. Based on the expression levels
of seven genes and corresponding coefficients, the risk score was
calculated for each osteosarcoma patient as following formula:
[MATH:
Risksco
re=0.457×PSIP1+0.428×CHD2+0.279×<
/mo>SMARCA4
mn>-0.266×HMG
N2-0.447×SP<
mn>140-0.577×CBX<
/mi>5-0.487×SFMBT2 :MATH]
. This risk model was validated in the TARGET cohort and [133]GSE21257
cohort. In the TARGET database, the high risk group had 48 patients and
low risk group had 47 patients. The Kaplan–Meier curve plot showed that
the low-risk group had better clinical outcomes compared with the
high-risk group (Fig. [134]5D, [135]P < 0.001). The predictive accuracy
of the risk model in the TARGET cohort was assessed by 1-, 3- and
5-years ROC analysis, of which area under the curve (AUC) values are
0.703, 0.707 and 0.696 (Fig. [136]5E). Figure [137]5F–H exhibited the
distribution of risk score, survival status and RNA expression profile
of selected TFs in the TARGET cohort.
Figure 5.
[138]Figure 5
[139]Open in a new tab
The construction and validation of the clinical risk prognostic model.
(A) Ten-fold cross-validation for tuning parameter selection in the
LASSO model. (B) LASSO coefficient profiles of the CIC-related TFs with
prognostic value. (C) The coefficients of selected genes. Red
represented coefficient > 0, and blue represented coefficient < 0. (D)
Kaplan–Meier curve plot of overall survival for patients from
the TARGET cohort. (E) The 1-, 3- and 5- year ROC plots for the TARGET
cohort. (F–H) Distributions of risk score, survival status, and RNA
expression profile of selected TFs in TARGET cohort. (I) Kaplan–Meier
curve plot of overall survival for patients from the [140]GSE21257
cohort. (J) The 1-, 3-, 5- year ROC plots for the [141]GSE21257 cohort.
(K-M) Distributions of risk score, survival status, and RNA expression
profile of selected TFs in the [142]GSE21257. The color in heatmaps (H
& M) represents the relative RNA expression.
The risk model exhibited similar predictive efficiency in
the [143]GSE21257 cohort, where the high risk group had 30 patients and
the low risk group had 23 patients. Low risk group members owned better
prognosis compared with the high-risk group (F[144]ig. [145]5I). The
AUC values of 1-, 3- and 5-ROC curves were 0.638, 0.781, and 0.772
(Fig. [146]5J). The distribution of risk score, survival status and RNA
expression profile of 8 TFs were presented in Fig. [147]5K–M. The risk
scores of patients from the TARGET cohort and [148]GSE21257 cohort were
presented in Table [149]S11.
The construction and validation of nomogram
The risk score and three clinical features, age, gender, and metastasis
status, were incorporated into the construction of the nomogram. Based
on the information of TARGET cohort, a nomogram was built, which is
capable of predicting the survival probability of 1-, 3-, and 5-years
(Fig. [150]6A). A nomogram is like a rule that can help clinicians
quickly estimate the survival probability of a patient. Each
characteristic of a patient corresponds to a score on the Points scale,
and these scores add up to a final score on the Total Points scale,
which corresponds to the survival rate of the patient. TARGET cohort
and [151]GSE21257 cohort were used to validate the nomogram. In
the TARGET cohort, the calibration curves at 1-, 3-, and 5-years showed
outstanding predictive performance (Fig. [152]6B), and ROC curves at
5-years showed that the AUC values of age, gender, metastasis, risk
score, and nomogram were 0.496, 0.525, 0.576, 0.696, and 0.766
respectively (Fig. [153]6C). In the [154]GSE21257 cohort, the
calibration curves at 1-, 3- and 5-years showed good predictive
performance as well (Fig. [155]6D), and the AUC values of ROC curves of
age, gender, metastasis, risk score, and nomogram were 0.480, 0.523,
1.000, 0.772, 0.884 (Fig. [156]6E).
Figure 6.
[157]Figure 6
[158]Open in a new tab
The construction and validation of predictive nomogram. (A) The
nomogram for predicting 1-, 3-, and 5-year overall survival, was
constructed based on age, gender, metastasis, and risk score of
patients from the TARGET cohort. Each feature of patients corresponds
to a score on the Point scale, and the final score on the Total Points
scale results from the addition of the above scores, which corresponds
to the survival probability. Point scale: 0–100. The score assigned to
each of the characteristics: Age (0.00–1.87); Gender (Male: 0.00;
Female: 1.53); Metastasis (No: 0.00; Yes: 25.00); riskScore
(0.00–100.00). (B) The calibration curve of the nomogram in terms of
the agreement between predicted and observed outcomes in the TARGET
cohort. (C) The time dependent ROC curves of age, gender, metastasis,
risk score and nomogram with AUC values of 0.496, 0.525, 0.576, 0.696,
and 0.766 at 5 years in the TARGET cohort. (D) The calibration of
nomogram in the [159]GSE21257 cohort. (E) The ROC curves of age,
gender, metastasis, risk score and nomogram with AUC values of 0.480,
0.523, 1.000, 0.772, 0.884 at 5 years in the [160]GSE21257 cohort.
The role of risk score in clinical characteristics and immune infiltration
Four clinical features, age, gender, histologic response, and
metastasis, were incorporated into the following study (Fig. [161]7A).
The distribution of age, gender, and metastasis did not differ between
the high-risk group and low-risk group (chi-square test,
X-square = 1.453, 0.051 and 0.058 respectively, P = 0.228, 0.821 and
0.809 respectively), whereas the percentage of patients with histologic
response stage 1/2 was higher in high-risk group (chi-square test,
X-square = 5.107, P = 0.024). And the risk score of patients with
histological response stage 1/2 were higher than those with stage 3/4
(P = 0.0026). However, the risk scores of patients did not differ in
age, gender, and metastasis status (Wilcoxon test, P = 0.24, 0.61, and
0.22 respectively).
Figure 7.
[162]Figure 7
[163]Open in a new tab
The association of risk score with clinical features and TME. (A) The
association of risk score with age, gender, histologic response, and
metastasis. Histograms presented the distribution of clinical features
between the high-risk group and the low-risk group in the TARGET
cohort. Boxplots presented the differential analysis for risk scores
between different clinical features. (B) The differential analysis of
stromal score, immune score, ESTIMATE score, and tumor purity between
high-risk group and low-risk group. (C) The correlations of risk score
with 64 stromal cells and immune cells. The r represents the
correlation between different cells. The correlation between risk score
and immune cells was presented by the characteristics of the line:
dotted line represents negative correlation, and the solid line
represents positive correlation; The color of the line represents the P
value; The width of the line represents the correlation value. (D) The
correlation of risk score with immune inhibitor genes and immune
stimulator genes. Red represented a positive correlation, and blue
represented a negative correlation. The height of the bar plot
represents the correlation value. The inner circle represented a
correlation coefficient of − 0.5, and the outer circle represented
a correlation coefficient of 0.5. *P < 0.05, **P < 0.01, ***P < 0.001.
Risk score exhibited a strong association with TME. In the TARGET
cohort, the stromal score, immune score, and ESTIMATE score were higher
in the low-risk group (P = 2.5e−4, 4.4e−6 and 9.6e−7 respectively),
whereas tumor purity was higher in the high-risk group (P = 9.6e−7)
(Fig. [164]7B). The relationship between risk score and 64 stromal
cells and immune cells was also investigated respectively in the TARGET
cohort. 30 types of cells were identified correlated with risk score
(P < 0.05), among which Astrocytes, B-cells, CD4+ naïve T-cells, CD4+
Tem, CD8+ T-cells, CD8+ Tcm, CD8+ Tem, Class-switched memory B-cells,
DC, Endothelial cells, iDC, ly Endothelial cells, Macrophages,
Macrophages M1, Macrophages M2, Mast cells, Memory C-cells, Mesangial
cells, Monocytes, MPP, mv Endothelial cells, naïve B-cells, pDC, and
Tgd cells were negatively correlated with risk score, whereas CD4+ Tcm,
CLP, Melanoccytes, MEP, Osteoblast and Sebocytes were positively
correlated with risk score (Fig. [165]7C). In addition, the expression
levels of immune inhibitor genes and immune stimulator genes were
associated with risk scores (Fig. [166]7D). Among immune inhibitor
genes, LGALS9 was highly negatively correlated with risk score
(r = − 0.478, P < 0.001), followed by LAG3 (r = − 0.502, P < 0.001) and
HAVCR2 (r = − 0.457, P < 0.001), and VTCN1 was the only gene positively
correlated with risk score (r = 0.267, P = 0.009). Among immune
stimulator genes, CD48 was the gene most negatively correlated with
risk score (r = − 0.506, P < 0.001), followed by TNFSF13 (r = − 0.491,
P < 0.001) and CD80 (r = − 0.459, P < 0.001), and HHLA2 was the only
gene positively correlated with risk score (r = 0.219, P = 0.033). The
correlation results of risk score with TME cells and immune related
genes were presented in Table [167]S12.
Prediction of drug sensitivity and response to immunotherapy
Risk score was found to correlate with drug sensitivity and response to
immune therapy. The estimated IC50 of drugs is presented in Table
[168]S13. In total, 22 drugs were identified positively correlated with
risk score, whereas 19 drugs were identified negatively correlated with
risk score (Fig. [169]8A). Figure [170]8B presented the top 3 drugs
(XAV939, Entospletinib, and ERK_6604) positively correlated with the
risk score and the top 3 drugs (BI-2536, Lapatinib, and Uprosertib)
negatively correlated with the risk score. XAV939 was the drug with the
highest positive correlation with risk score (r = 0.386, P < 0.001),
followed by Entospletinib (r = 0.365, P < 0.001), ERK_6604 (r = 0.344,
P < 0.001). BI-2536 (r = − 0.371, P < 0.001) was the drug with the
highest negative correlation with risk score, followed by Lapatinib
(r = − 0.349, P < 0.001) and Uprosertib (r = − 0.347, P < 0.001). The
result of correlation analysis between drugs and risk score was
presented in Table [171]S14. TIS and IPS were used to predict the
responses of patients to immunotherapy. TIS were negatively correlated
with risk score (r = − 0.54, P < 0.001) (Fig. [172]8C). The
correlations between risk and four IPS scores are shown in
Fig. [173]8D. MHC_IPS and EC_IPS were negatively correlated with risk
score (r = − 0.45, P < 0.001; r = − 0.54, P < 0.001), whereas SC_IPS
and CP_IPS were positively correlated with risk score (r = 0.58,
P < 0.001; r = 0.42, P < 0.001). The TIS and IPS scores of each
osteosarcoma patient are presented in Table [174]S15.
Figure 8.
[175]Figure 8
[176]Open in a new tab
The role of risk score in drug sensitivity and immune therapy. (A) The
correlation between risk score and drug sensitivity. Red represented
a positive correlation, and green represented a negative correlation.
(B) Scatter charts presented 3 drugs (XAV939, Entospletinib, and
ERK_6604) most positively correlated with a risk score and 3 drugs
(BI-2536, lapatinib, and Uprosertib) most negatively correlated with
risk score. (C) The correlation of Tumor Inflammation Signature (TIS)
with risk score. (D) The correlation of risk score with MHC molecules
IPS (MHC_IPS), immunomodulators IPS (CP_IPS), effector cells IPS
(EC_IPS), and suppressor cells IPS (SC_IPS). *P < 0.05, **P < 0.01,
***P < 0.001.
Discussion
Osteosarcoma is one of the most common malignant childhood tumors
occurring in bone tissue. The prognosis of osteosarcoma has not been
improved since the 1980s because of the stagnation of treatment
methods. Immunotherapy is a novel method using the immune system to
attack tumor cells. Given the promising results in clinical trials of
many cancers, immunotherapy is expected to lead to a breakthrough in
survival. CIC events involve the release of tumor antigens and the
infiltration and activation of immune cells, which are highly
correlated with the effectiveness of immunotherapy. In addition, a
growing number of studies demonstrated the pivotal role of epigenetic
modulation in the regulation of immune cells. Therefore, we hypothesize
that the cross-talk between CIC and epigenetic modulation is a
potential impact factor on the clinical result of immunotherapy,
targeting which is a method to overcome the shortcomings of
immunotherapy. Both epigenetic modulators and immunotherapy drugs are
double-edged swords. The targets of epigenetic drugs occur in tumor
tissues as well as normal tissues, and immunotherapy efficacy is also
accompanied by drug resistance and irAEs. The combination of epigenetic
drugs and immunotherapy is expected to make up for each other’s
disadvantages. To advance this combination therapy in osteosarcoma, we
explored the molecular signature of epigenetic and immune responses.
The impact of epigenetic changes on CIC events has been identified by
numerous studies. In our study, “infiltration of immune cells in
tumors” was the CIC event correlated with the most epigenetic factors.
In tumor tissues, tumor cells are surrounded by a wide variety of
immune cells, such as macrophages, Treg cells, NK cells, B cells etc.,
which is defined as TME. TME is the soil in which the tumor cells grow,
and it is required for the onset and dissemination of
osteosarcoma^[177]41. Through univariate cox analysis, 8 CIC-related
epigenetic factors were identified highly correlated with prognosis.
SFMBT2, one of the polycomb group proteins, downregulates the
expression level of matrix metalloproteinases (MMPs) via interacting
with YY1 and various repressive histone marks^[178]42. What’s more, the
downregulation of SFMBT2 was found to advance the infiltration of tumor
associated macrophages (TAMs) in prostate cancer^[179]43. SP140 is an
epigenetic reader containing bromodomain, the loss-of-function mutation
of which is correlated with multiple autoimmune diseases, such as
Crohn’s disease and multiple sclerosis. Recently, SP140 was identified
as a repressor of macrophage topoisomerases through a global proteomic
strategy^[180]44. SMARCA4 encodes a transcriptional activator protein
called BRG1 that forms the core subunit of the SWI/SNF complex^[181]45.
20% of human cancers were found accompanied by mutations in subunits of
the SWI/SNF complex that has been linked to enhanced interferon
response. Besides, the SWI/SNF complex opposes to PRC2 transcriptional
repression, whose core enzymatic subunit is EZH2, a histone
methyltransferase^[182]46. Therefore, SMARCA4 is a potential biomarker
for evaluating whether to adopt epigenetic modulators or not. SMARCA4
mutation is found in adult-onset epithelial and mesenchymal
tumors^[183]47. A pan-cancer analysis based on the data from TCGA and
GTEx database revealed that SMARCA4 was observed upregulated in most
cancers, which was correlated with poor overall survival in ACC, MESO,
SARC, and SKCM^[184]48. PSIP1 was identified to control the survival of
T cells via its structure changes induced by L-arginine^[185]49. ACTR6
was found associated with TAMs in lung cancer. Compared with M1
macrophages, ACTR6 was downregulated in M2 macrophages^[186]50. CBX5
encodes HP1α, a member of the human heterochromatin protein 1 family,
which binds H3 di- or tri-methylated at position lysine 9^[187]51. HP1α
also participates in the differentiation and angiogenic function of
endothelial progenitor cells through modulating the expression of
angiogenic genes and progenitor cell marker genes^[188]52. CHD2 is
required for neural circuit and its mutation is a driver of abnormal
brain function, early onset epileptic encephalopathy and intellectual
disability^[189]53,[190]54. In addition, CHD2 mutation was frequently
reported in chronic lymphocytic leukaemia and CHD2 was identified
essential for myeloid differentiation^[191]55. HMGN2 protein not only
is expressed in tumor cell lines but also work as an anti-tumor
effector molecular released by CD8+ T cells^[192]56,[193]57.
Based on the expression of these eight genes, osteosarcoma was
clustered into two subtypes. Function analysis revealed that the two
clusters predominantly differed in immune function. The cluster 1 with
a better prognosis was enriched with multiple immune-related pathways
and GO terms, such as chemokine signaling pathway, Toll-like receptor
signaling pathway, Nod like receptor signaling pathway, Natural killer
cell mediated cytotoxicity, antigen binding, chemokine activity,
activation of immune response, and so on. It was obvious that this
clustering was capable of distinguishing the immune characteristics of
osteosarcoma patients. Subsequent immune-related analysis also
corroborated this result. ESTIMATE score indicated that the
osteosarcoma samples in cluster 1 were rich in stromal and immune
cells. The higher infiltration level of immune cells is a potential
reason for cluster 1 to have a better prognosis. It was found that CD8+
T cell, CD8+ Tem (effector memory T cell), CD8+ Tcm (central memory T
cell), B cell, and class-switched memory B cell were enriched in
cluster 1. CD8+ T cell is the core of a variety of immunotherapy
strategies. Anti-PD1/PDL1 therapy activates CD8+ T cells to attack
tumor cells via targeting PD1/PDL1^[194]58. The high abundance of T
cells in cluster 1 suggested that the patients in this cluster were
more likely to benefit from immunotherapy. In this study, Macrophage,
M1 macrophage, and M2 macrophage were also higher in cluster 1. In
addition, M2 macrophages accounted for the vast majority of immune
cells in TME of osteosarcoma. M1 macrophages produce pro-inflammatory
cytokines, whereas M2 macrophages produce anti-inflammatory cytokines
and promote vasculogenesis. The polarization between M1 and M2
macrophages plays a critical role in inflammation and cancer^[195]59.
Besides PD1/PDL1, more and more immune checkpoints emerge and exhibit
promising clinical values. Compared with the cluster 2, the cluster 1
had higher expression levels of immune checkpoint genes. The analysis
of immune features also demonstrated that the two clusters had great
heterogeneity in terms of immunity.
Finally, a prognostic risk model was developed using the LASSO Cox
regression algorithm. When the mean squared error was the minimum
value, seven genes were identified as significant features, and the
coefficient of ACTR6 was zero. Therefore, PSIP1, CHD2, SMARCA4, HMGN2,
SP140, CBX5, and SFMBT2 were included in the development of risk model.
Although these genes have been linked to various diseases, only a part
of them are explored in osteosarcoma. Some studies revealed that HMGN2
plays an anti-tumor role in osteosarcoma. HMGN2 is one of the
no-histone nuclear proteins with the most abundance in vertebrates, and
its overexpression is able to inhibit cell growth of SaO2 and U2OS cell
lines^[196]57. Another study reported that exogenous HMGN2 protein can
inhibit the migration and invasion of osteosarcoma cell lines^[197]60.
Interestingly, CD8+ T cells can release HMGN2 proteins that are
transported into tumor cells and induce tumor apoptosis in a
dose-dependent manner^[198]56. It is reported that SMARCA4 is involved
in other sarcomas. Small cell carcinomas of the ovary hypercalcemic
type (SCCOHT) is characterized by SMARCA4 alterations, and exhibits
good response to immunotherapy^[199]45. The functions of these genes
depend on organization and particular cell types, and their roles
require investigation through experiments in vivo and in vitro. The
patients with the low risk scores in both the train cohort and test
cohort owned better prognosis. With the nomogram, clinicians can
quickly give patients survival probability. However, the generalization
and application of this model in clinical require the inclusion of more
training samples in the future to improve accuracy. We found that risk
score was mainly correlated with histological response, which indicated
that these selected epigenetic factors modulated the response to
chemotherapy in osteosarcoma. A recent clinical trial reported that,
for patients with localized disease and complete remission after
surgery, poor histological response referred to a worse effect of
surgery therapy^[200]61. The investigation into the role of risk score
in the TME of osteosarcoma revealed that risk score was a good
indicator to predict immune status in the osteosarcoma sample. The risk
score was highly negatively associated with CD8+ T cells. Besides, risk
score also exhibited a wide correlation with immune modulator genes.
Most immune modulator genes were negatively correlated with the risk
score, including PDCD1 and CD274. VTCN1 and HHLA2 were the only two
immune modulator genes positively correlated with risk score. They are
both the members of B7 family. VTCN1, also known as B7-H4, is
abnormally upregulated in tumor cells and TAMs, and works as a negative
regulatory factor of T cell immune response^[201]62,[202]63. Song et
al.^[203]64 reported that the inhibition of glycosylation of B7-H4 by
NGI-1 improved the immunogenic properties of tumor cells and enhancing
the anti-tumor effect of dendritic cells as well as T cells. The role
of VTCN1 in the osteosarcoma has been initially investigated by Qiang
Dong and Xinlong Ma. They examined the expression level of VTCN1 in
osteosarcoma sample by immunohistochemistry, and found that VTCN1 was
upregulated in the tumor samples compared with the paired normal tissue
samples^[204]65. HHLA2 is predominantly expressed in various tumor
cells and monocytes, but not in normal tissues other than breast,
gallbladder, kidney, intestines, and placenta^[205]66. The role of
HHLA2 in cancers is various. In epithelial ovarian cancer, HHLA2 was
reported positively correlated with tumor differentiation, the
infiltration level of CD8+ T cells, and prognosis^[206]67. However, in
non-small cell lung cancer, HHLA2 deficiency inhibited tumor cell
proliferation, migration, and invasion in vitro, and blocked the
polarization of M2 macrophages^[207]68. In osteosarcoma, HHLA2 is
upregulated in metastasis tumor samples and associated with worse
clinical outcomes^[208]69. Combining our result and previous studies,
we hypothesized that, for osteosarcoma patients without response to
anti-PD1/PDL1 therapy, targeting VTCN1 or HHLA2 was a potentially
promising treatment method.
Risk score is highly correlated with drug sensitivity. XAV939, a
Wnt/β-catenin pathway modulator^[209]70, was the drug with the highest
positive correlation with risk score in this study. It has been
confirmed that XAV939 suppresses the proliferation and migration of
A549 cells from lung adenocarcinoma in vitro^[210]71. In osteosarcoma,
the blockade of the Wnt/β-catenin pathway by XAV939 reduces the
Adriamycin resistance of U2OS cells^[211]72. It is widely known that
chemotherapy resistance is a key issue in the treatment of
osteosarcoma. Hence, it is possible that the combination of XAV939 and
traditional chemotherapy drugs can enhance the effectiveness of
treatment. The drug with the highest negative correlation with risk
score was BI-2536, a selective inhibitor of polo-like kinase 1^[212]73.
In vitro and in vivo tests demonstrated that BI-2536 inhibited the
proliferation of osteosarcoma cell lines^[213]74,[214]75. What’s more,
BI-2536 was reported to enhance the effects of various conventional
chemotherapeutic agents^[215]76–[216]78. Hence, for patients with the
low risk scores, BI-2536 is a potentially potent complement to
chemotherapy or neoadjuvant chemotherapy. Beyond drug sensitivity, risk
score exhibited a correlation with indicates that are used to predict
response to immunotherapy in pan-cancer. Generally, the patients with
higher TIS scores benefit more from anti-PD1 therapy^[217]79. In the
current study, the TIS score was highly negatively associated with the
risk score, which means that the patients with high risk scores seem to
benefit less from immunotherapy. MHC_IPS and EC_IPS were negatively
correlated with risk score, whereas SC_IPS and CP_IPS were positively
correlated with risk score. It seems like that the TME in patients with
high risk scores is deficient in antigen presentation and effector
cells. Epigenetic modulation in osteosarcoma is complex and the role of
risk score is different in different immunotherapies. Due to the lack
of high-quality osteosarcoma immunotherapy cohorts, it is hard to
establish a link between scores and response to immunotherapy. It is
essential to explore the application of risk scores in the
immunotherapy cohorts in the future.
We have to admit that there were several limitations in this study.
Firstly, only two datasets were included in this research, and the risk
model required validation in more independent datasets. Secondly,
epigenetic gene signatures were identified via bioinformatic methods
and required experimental validations. Thirdly, because of the lack
of an immunotherapy cohort of osteosarcoma, several well-established
algorithms were applied to predict immunotherapy response and the role
of risk score needs to be explored in real-world data.
Conclusion
In conclusion, we explored the association between epigenetic factors
and CIC events in osteosarcoma. Based on eight factors highly
correlated with CIC events, two epigenetic subtypes in osteosarcoma
were identified via NMF clustering. The two subtypes were mainly
distinguished by immune response and cell cycle regulation. Finally, a
clinical risk model and a nomogram were established, which can help
clinicians quickly predict the survival probability of patients. Risk
score is strongly correlated with drug sensitivity, immune
infiltration, and immune checkpoint genes. Our study could shed a novel
light on the epigenetic modulation mechanism of osteosarcoma and helps
search for potential novel drugs.
Supplementary Information
[218]Supplementary Legends.^ (14.3KB, docx)
[219]Supplementary Table S1.^ (18.6KB, xlsx)
[220]Supplementary Table S2.^ (25.5MB, xlsx)
[221]Supplementary Table S3.^ (16KB, xlsx)
[222]Supplementary Table S4.^ (33.7KB, xlsx)
[223]Supplementary Table S5.^ (21.4KB, xlsx)
[224]Supplementary Table S6.^ (8.9KB, xlsx)
[225]Supplementary Table S7.^ (11.7KB, xlsx)
[226]Supplementary Table S8.^ (389.6KB, xlsx)
[227]Supplementary Table S9.^ (394.2KB, xlsx)
[228]Supplementary Table S10.^ (205.8KB, xlsx)
[229]Supplementary Table S11.^ (13KB, xlsx)
[230]Supplementary Table S12.^ (16.5KB, xlsx)
[231]Supplementary Table S13.^ (351.2KB, xlsx)
[232]Supplementary Table S14.^ (18.5KB, xlsx)
[233]Supplementary Table S15.^ (15.7KB, xlsx)
Acknowledgements