Abstract
Background: The immune checkpoint blockade (ICB) with anti-programmed
cell death protein 1(PD-1) on HNSCC is not as effective as on other
tumors. In this study, we try to find out the key factors in the
heterogeneous tumor-associated monocyte/macrophage (TAMM) that could
regulate immune responses and predict the validity of ICB on HNSCC.
Experimental Design: To explore the correlation of the TAMM
heterogeneity with the immune properties and prognosis of HNSCC, we
established the differentiation trajectory of TAMM by analyzing the
single-cell RNA-seq data of HNSCC, by which the HNSCC patients were
divided into different sub-populations. Then, we exploited the topology
of the network to screen out the genes critical for immune hot
phenotype of HNSCC, as well as their roles in TAMM differentiation,
tumor immune cycle, and progression. Finally, these key genes were used
to construct a neural net model via deep-learning framework to predict
the validity of treatment with anti-PD-1/PDL-1
Results: According to the differentiation trajectory, the genes
involved in TAMM differentiation were categorized into early and later
groups. Then, the early group genes divided the HNSCC patients into
sub-populations with more detailed immune properties. Through network
topology, CXCL9, 10, 11, and CLL5 related to TAMM differentiation in
the TME were identified as the key genes initiating and maintaining the
immune hot phenotype in HNSCC by remarkably strengthening immune
responses and infiltration. Genome wide, CASP8 mutations were found to
be key to triggering immune responses in the immune hot phenotype. On
the other hand, in the immune cold phenotype, the evident changes in
CNV resulted in immune evasion by disrupting immune balance. Finally,
based on the framework of CXCL9-11, CLL5, CD8^+, CD4^+ T cells, and
Macrophage M1, the neural network model could predict the validity of
PD-1/PDL-1 therapy with 75% of AUC in the test cohort.
Conclusion: We concluded that the CXCL9, 10,11, and CCL5 mediated TAMM
differentiation and constructed immune hot phenotype of HNSCC. Since
they positively regulated immune cells and immune cycle in HNSCC, the
CXCL9-11 and CCL5 could be used to predict the effects of
anti-PD-1/PDL-1 therapy on HNSCC.
Keywords: squamous cell carcinoma of head and neck (HNSCC),
tumor-associated monocyte/macrophage (TAMM), immune checkpoint blockade
(ICB), tumor micro-environment (TME), CXCL, CCL5, PD-1/PD-L1
Introduction
Application of immune checkpoint blockage (ICB) has significantly
improved the prognosis of multiple tumors, but exerted limited effects
on head and neck squamous cell carcinoma (HNSCC) because less than 30%
of the patients got a better prognosis ([44]Curran et al., 2010;
[45]Topalian et al., 2012; [46]Seiwert et al., 2016; [47]Clarke et al.,
2021; [48]García Campelo et al., 2021). To find out the HNSCC
sub-populations susceptible to ICB therapy, various criteria have been
proposed for HNSCC classification, by which HNSCC was classified into
the enhanced and decreased immune subtypes with the immune-related
genes ([49]Cao et al., 2018), into the CD8^+ high and CD8^+ low
subtypes with the density of infiltrating CD8^+ T cells ([50]Saloura et
al., 2019), into the basal, mesenchymal, atypical, and classical
subtypes with the integrated genomic characteristics ([51]Walter et
al., 2013), and even into the m6A^high and m6A^low subtypes with the
N6-methyladenosine (m6A) methylation levels on mRNAs ([52]Yi et al.,
2020). Although these criteria explicated the clinical and immune
characteristics of HNSCC from different perspectives, how these HNSCC
subtypes formed and the roles of the genes involved in it remained
elusive.
When inflammation took place, the tumor-associated monocyte/macrophages
in circulating blood were motivated into the inflammatory focus to
maintain homeostasis, eliminate pathogens, and balance immune responses
([53]Shi & Pamer, 2011). There were three types of tumor associated
monocyte/macrophages, namely, the classical (CD14^+; CD16^−), the
non-classical (CD16^+), and the intermediate tumor associated
monocyte/macrophages (CD14^+; CD16^+) ([54]Ziegler-Heitbrock et al.,
2010). During tumorigenesis, the tumor-associated monocyte/macrophages
in circulation (mainly the classical type) were chemoattracted into
tumor focus, and differentiated gradually into dendritic cells (DC) and
Tumor Associated Macrophages (TAM) ([55]Movahedi et al., 2010;
[56]Franklin et al., 2014; [57]Guilliams et al., 2014; [58]Li B et al.,
2020). More than 50% of the immune cells in tumor micro-environment
(TME) were TAM that affected the migration, invasion, angiogenesis, and
drug-resistance of tumors ([59]Watters et al., 2005; [60]Kimura et al.,
2007; [61]Zheng et al., 2018). The M1-like phenotype of TAM exhibited
the inhibitory effects on tumors, such as the promoted inflammatory
response and chemoattraction of immune cells ([62]Goswami et al.,
2017). Reversely, M2-like phenotype of TAM suppressed inflammatory
response, and enhanced immune evasion, angiogenesis, and metastasis,
which resulted in a poor prognosis ([63]Hu et al., 2016; [64]Seminerio
et al., 2018). However, recent studies reported that M1-like phenotype
of TAM was also related to the poor prognosis in HNSCC and
medulloblastoma by suppressing inflammatory response and promoting
metastasis ([65]Lee et al., 2018; [66]Xiao et al., 2018). Previously,
TAM was thought mainly to be macrophage M2, while the increasing
evidence indicated that TAM also exhibited the phenotype of macrophage
M1, suggesting that TAM contained the third population of macrophages
other than macrophage M1 and M2. The third macrophage population was
supposed to co-express the M1 and M2 characteristics and transform into
M1 or M2 in certain instances ([67]Estko et al., 2015; L. ; [68]Gao et
al., 2016; [69]Kloepper et al., 2016). All the above findings indicated
that the role of TAM in tumor progression could be complicated and not
simply attributed to macrophage M1 and M2. Since the TAM was
differentiated from the tumor-associated monocyte/macrophages
gradually, the differentiating and differentiated TAM were termed as
tumor-associated monocyte/macrophages/Macrophages (TAMM) in recent
studies ([70]Cassetta et al., 2019; [71]Singhal et al., 2019). More and
more studies implicated the TAMM as the potential target of ICB
therapy. The relevance between TAMM responses and ICB therapy has been
established by bioinformatic methods. In triple negative breast cancer,
machine learning identified the TAMM-expressed genes which were highly
associated with the prognosis and ICB therapy, and constructed a model
predicting the response to ICB therapy with the 100% validation queue
AUC ([72]Bao et al., 2021). Moreover, WGCNA was used to find that the
marker genes expressed in TAMM of glioblastoma, which were highly
correlated with prognosis and ICB therapy, and were more active in the
patients susceptible to ICB therapy ([73]Zhang et al., 2021). Despite
this, there are relatively few studies on HNSCC concerning the role of
TAMM in immune response and ICB therapy. Since TAMM differentiation
endowed TAMM with heterogeneity dynamically, instead of statically, we
proposed a criterion that combined the genes involved in TAMM
differentiation with the immune cells to depict the immune phenotype
and prognosis of HNSCC in more detail.
Materials and Methods
Data Collection
The single cell RNA-seq (scRNA-seq) data of [74]GSE139324 (10X
genomics), including the tumor infiltrating immune cells from 16 HPV
negative patients and the immune cells from the peripheral blood of a
healthy donor, and [75]GSE103322 (Smart-seq2), containing 5,902 single
cells from 18 HNSCC patients, were obtained from Gene Expression
Omnibus (GEO). Multiomics data and clinical data of 502 HNSCC patients
obtained from The Cancer Genome Atlas (TCGA) database
([76]Supplementary Table S1), including mRNA expression (level 3,
Illumina RNA-Seq), miRNA expression (level 3, Illumina miRNA-Seq),
somatic copy number variation (CNV level 3, Affymetrix SNP 6.0), and
somatic mutation (level 4, MAF files), were obtained from UCSC Xena
browser. The array data and clinical data of five HNSCC cohorts,
[77]GSE65858 (n = 270), [78]GSE40774 (n = 134), [79]GSE39366 (n = 138),
[80]GSE117973 (n = 77), and [81]GSE41613 (N = 97), were obtained from
Gene Expression Omnibus (GEO) database ([82]Supplementary Table S1).
The bulk transcriptome data and clinical data of six cohorts accepted
the PDL-1/PD-L1 antibody immunotherapy, namely [83]GSE93157 (n = 65,
Non-Small Cell Lung Carcinoma, HNSCC and Melanoma), [84]GSE154538 (n =
8, gastrointestinal cancer), [85]GSE141119 (n = 12, melanoma),
[86]GSE91061 (n = 109, melanoma and non-small cell lung cancer),
[87]GSE78220 (n = 28, melanomas), [88]GSE176307 (n = 88, Metastatic
Urothelial Cancer), and the IMvigor210 (n = 348, bladder cancer), were
obtained from Gene Expression Omnibus (GEO) and the IMvigor210 database
([89]Supplementary Table S2). [90]GSE93157 was array data, while
[91]GSE154538, [92]GSE141119, [93]GSE91061, [94]GSE78220,
[95]GSE176307, and the IMvigor210 were bulk transcriptome data. The
mRNA-seq data from the HNSCC cell line that accepted the treatment of
anti-tumor drugs were obtained from Genomics of Drug Sensitivity in
Cancer (GDSC).
Data Processing
In the scRNA-seq from [96]GSE139324, with the exclusion of the genes
detected in fewer than three cells, the cells containing mRNA more than
4,500 or less than 200, and the cells expressing mitochondria genes
more than 10% transcripts, there were 19,718 genes from 39,711
qualified cells of total 39,994 cells (283 cells were screened out).
Through SCT in seurant package SCT, the data from the 18 patients in
[97]GSE139324 cohort were integrated to screen out the non-biological
inferences, such as batch effect. Similarly, in the scRNA-seq of
[98]GSE103322 (Smart2-seq without screen), there were 21,519 genes and
5,844 qualified cells from 18 patients integrated by SCT. The HNSCC
RNA-seq counts [log[2](rawcounts+1)] obtained from USCS through
exp[log[2](rawcounts+1)-1] were restored to raw counts, and then the
log2(fpkm-uq+1) from USCS was used to compare them with the data from
other databases. There were 501 HNSCC samples (one normal sample was
excluded) for the subsequent analyses. The [99]GSE65858, [100]GSE40774,
[101]GSE39366, [102]GSE117973, and [103]GSE41613 were normalized prior
to following analyses. There were 501 samples in [104]GSE93157,
[105]GSE154538, [106]GSE141119, [107]GSE91061, [108]GSE78220,
[109]GSE176307, and the IMvigor210 for the following analyses except
defective and reiterated data. The data from the RNA-seq of GDSC2 cell
line were normalized with TPM for subsequent processing.
Analyses on Squamous Cell Carcinoma of Head and Neck scRNA-Seq Data
For the [110]GSE139324 cohort: 1) the data integrated with “SCT” Seurat
package was applied for PCA analysis to find out the first 50 principal
component analysis (PCA); 2) Umap (Uniform Manifold Approximation and
Projection for Dimension Reduction) dimension reduction was performed
on the 50 PCAs. In this unsupervised clustering, the function of
FindNeighbors in Seurat package was used to construct a KNN graph based
on the Euclidean distance in PCA space (top 50 PCAs, k = 20), and then,
the function of FindClusters (Louvain algorithm) was used to cluster
the cells with the resolution of 0.1. The K-NN clustering classified
the consequences undergoing the dimension reduction into four clusters,
which were annotated by SingleR as NK cells (n = 14,925), T cells (n =
14,073), B cells (n = 2,789), and tumor-associated monocyte/macrophages
cells (n = 7,924). 3) Tumor-associated monocyte/macrophages cells were
classified by K-NN into seven further clusters. Cluster 0, 1, 2, and 4
were annotated by SingleR as tumor-associated monocyte/macrophages (n =
6,875), while the cluster3 (n = 312), 5 (n = 478), and 6 (n = 259) as T
and B cells. 4) The T cells were applied for Multimodal reference
mapping ([111]Hao et al., 2021) and divided into eight clusters,
namely, the CD4 CTL, CD4 Navie cells, CD4 TCM, CD4 TEM, CD8 Navie
cells, CD8 TCM, CD8 TEM, and proliferating T cells ([112]Supplementary
Table S3). 5) The tumor-associated monocyte/macrophages were applied
for GSVA analysis for function enrichment. 6) The tumor-associated
monocyte/macrophages were applied for pseudotime analysis through
Monocle2 package and Destiny package, which adopted different manners
to reduce the dimensions of the high-dimensional data. The single cell
was separated and projected into low-dimensional space to form a
differentiation trajectory with knots. Each knot represented a similar
status of differentiation. (1) Through the data of single cell lineage,
Monocle 2 adopted the embedding converse diagraph to learn the explicit
principal graph ([113]Packer et al., 2019). 2) Destiny adopted the
diffuse maps (differentiating cells follow noisy diffusion-like
dynamics) to mimic the division from multipotent cells ([114]Coifman et
al., 2005). 7) The “InferCNV” R package 1.10.1 ([115]Patel et al.,
2014) and CellPhoneDB (Python edition) ([116]Efremova et al., 2020)
were performed on all clusters for CNV analysis (normal blood cells as
control) and cell communication analysis.
[MATH:
CNVk(i)=∑j=
i−50i+
50Ek(Oj
)/101 :MATH]
, where CNV(i) was the estimated relative copy number, and
[MATH:
Ek(O
j) :MATH]
was mRNA level, of the i^th gene in the cell k at the whole genomic
scope. 8) The differentially expressed genes (DEG) between
tumor-associated monocyte/macrophages C1 and C0 were summarized with
the “Findmarker” Seurant package. Setting |log[2]fold Change|>1.3 and
FDR<0.05 as the cutoff criteria, the Log[2]Fold Change >1.3 was
regarded as the characteristics of the genes for the early
differentiation of TAMM, while Log[2]FoldChange<-1.3 as the
characteristics of the genes for the late differentiation of TAMM
([117]Supplementary Table S4). For [118]GSE103322 cohort: 1) PCA
analysis was performed on the data integrated by “SCT” Seurat package.
According to the specific markers, the cells were divided into the
malignant epithelial cells (KRT14, KRT6A, EPCAM, n = 1939), Cancer
associated fibroblasts (FAP, PDRN, n = 1,697), T cells (CD2, CD3D, n =
1,633), B cells (SLAMF7, CD79A n = 354), endothelial cells (PECAM1, VWF
n = 75), and mono-macrophage cells (CD14, CD163, CD68, n = 146). 2) T
cells were further classified with Multimodal reference mapping into
eight clusters of CD4 CTL, CD4 Navie cells, CD4 TCM, CD4 TEM, CD8 Navie
cells, CD8 TCM, CD8 TEM, and proliferating T cells ([119]Supplementary
Table S5).
CIBERSORT and ESTIMATE for Immune Cell and Stromal Scores
For the one TCGA HNSCC and five GEO HNSCC cohorts, “CIBERSORT” and
“ESTIMATE” R package were applied to calculate the contents of the 22
kinds of immune cells (1,000 permutations) and immune and stromal
score.
The Unsupervised Clustering on TCGA Squamous Cell Carcinoma of Head and Neck
and [120]GSE65858 Cohorts
According to the scores of the genes in the early TAMM differentiation
and the 22 kinds of immune cells, the unsupervised clustering (through
“ConsensuClusterPlus” R package) was applied to the samples with
Euclidean distance and Ward (unsquared distances) linkage to get the
sub-populations with different immune phenotype and prognosis.
Associations of TCGA Squamous Cell Carcinoma of Head and Neck Subtype With
DNA Methylation, CNVs and Mutations
The data of methylation probes were normalized with “wateRmelon” R
package, and the difference in methylation probes were analyzed with
“limma” R package. The evidently altered regions in genome were
screened with GISTIC2.0. The numeric focal CNV values larger than 0.2
meant gain, while less than 0.2 meant loss. Through Somatic mutation
data, the TMB (the number of non-synonymous mutations in every million
bases of somatic cells) of each patient in the TCGA HNSCC subtype were
calculated.
Search for the DEGs in the Subtype of TCGA HNSCS and [121]GSE65858 Cohorts
The DEGs were obtained by comparing the A3 to A1 subtype, and the A3 to
B subtype in TCGA HNSCC and [122]GSE65858 cohorts with “limma” in R
package. The TCGA HNSCC cohorts were produced by RNA-seq, while the
[123]GSE65858 resulted from micro-array. One criterion failed to
satisfy the cohorts from a different sequencing approach. If the
threshold of [124]GSE65858 was identical to that for TCGA HNSC, the
DEGs would be rare. For the TCGA HNSCC cohort, |Log[2]Fold Change|>1
and FDR<0.05 were set up as standard. According to the DEGs in
[125]GSE65858 array, a threshold of |Log[2]Fold Change|>0.2 and
FDR<0.05 was selected to keep the numbers of DEGs in the two cohorts
from varying too much. FDR was the p value calibrated using the
Benjamini–Hochberg method.
Confirmation of the Key Genes
In the two HNSCC cohorts, the comparison between A3 and B subtypes gave
rise to 181 overlapped candidate genes. Centiscape was applied to the
analysis on the protein crosstalk network and was constructed with PPI
database. Each knot in the PPI network constructed with 181 DEGs was
evaluated with the centiscape of cytoscape for the
topo-characteristics, namely, Degree, Eigenvector Centrality, and
Betweennesss.
Degree was the most direct and classical index evaluating the
regulatory and importance of knot, which was defined as the nodes
directly connected to a given node.
Eccentricity
[MATH:
Cecc(v) :MATH]
represent the reciprocal inverse of the longest path between the knot v
and all other knots. The eccentricity of a node in a biological network
can be interpreted as easiness of a protein to be functionally
influenced by all other proteins in the same network.
*
[MATH:
Cecc
msub>(v)=1max{d
mi>ist(v, w) : w ∈ V }
:MATH]
, in which v and w were the nodes in network (V)
S.-P. Betweenness
[MATH:
Cspb(v)
:MATH]
represents the ratio of the path number connecting the knot s and t
through v to the total number of path. A high S.-P. Betweenness score
meant that the node, for certain paths, was crucial to maintain node
connections.
*
[MATH:
Cspb
msub>(v)=<
mstyle
displaystyle="true">∑s≠v∈V∑t≠v∈V
σst(v
)σs
t :MATH]
, in which s, t and v were nodes in network (V)
Our purpose was to find out the knot with the higher values on the
topo-characteristics, because the higher the value the more significant
it was. Since the relative significance of Degree was higher than
Eigenvector Centrality, and the Eigenvector Centrality equaled
Betweenness, we selected the first 40 knots with the higher Degree.
Then, we selected the first 20 DEGs with the higher Eigenvector
centrality and Betweenness, respectively. The thresholds of 40 and 20
were set empirically, and had no effect on the outcomes, because the
key knots with the higher values of the topo-characteristics would vary
with the threshold. According to the descending order of Degree, the
first 40 candidate genes were selected. According to the descending
sequence of Eigenvector Centrality and Betweenness, the first 20 genes
were selected from the 40 candidates ([126]Supplementary Table S6).
Finally, 12 genes included in both above populations were set up as the
hub genes. KEGG database was applied for pathway correlation, CluoGO
for visulization, and all the manipulations were based on Cytoscape.
From the DEGs by comparing the A3 to A1 subtype of the two HNSCC
cohorts, 41 genes were selected for the protein crosstalk network
constructed with PPI database.
Pathway Enrichment Analysis
The DEGs from the comparison between the A3 and B subtype in both the
TCGA HNSCC and [127]GEO65858 cohorts were applied for GSEA enrichment.
Then, the Enrichment map was visualized and annotated. Sample Gene Set
Enrichment analysis (ssGSEA) was performed on the TCGA HNSCC,
[128]GSE65858, [129]GSE39366, [130]GSE117973, [131]GSE40774, and
[132]GSE41613 cohorts with “GSVA” R package to grade the 29 immune
signatures ([133]He et al., 2018).
[MATH:
Fold−Change=1n1∑i∈
immune hotimmune rela
ted scorei− 1n2∑i∈
immune coldimmune rel
ated scorei :MATH]
Where n1 and n2 were the number of immune hot and immune cold samples,
respectively. Immune related score was the sum of 22 immune cells
scores obtained by Cibersort and 29 immune signature scores obtained by
GSVA.
The GSVA Scores of the Four Chemokines and the Confirmation of the Immune Hot
and Immune Cold Subtype
According to the mRNA levels of the four chemokines, CXCL9, CXCL10,
CXCL11, and CCL5, TCGA HNSCC, [134]GSE65858, [135]GSE39366,
[136]GSE117973, [137]GSE40774, and [138]GSE41613 cohorts were applied
for ssGSEA with GSVA in R package and divided by the median grade into
the high- and low-graded subtype, namely, the immune hot and immune
cold subtype.
Survival Analysis
The survival curve was generated by “Survminer” R package. The
statistical differences among the immune subtype of TCGA HNSCC and
[139]GSE65858 cohorts were obtained by log rank test.
Construction of Neural Network
By “neurnet” R package, a neural network containing an input layer, two
hiding layers (there were 20 neurons in the first layer, and five
neurons in the second layer. Both layers were in dropout), and an
output layer.
Activation Function:
[MATH:
11+e
−x :MATH]
; Loss Function:
[MATH: 1N∑i−(yi ∗
log(pi)+(1
−yi) ∗
log(1−p<
/mi>i))
mrow> :MATH]
Statistical Analysis
All the statistical analyses were performed with R software version
4.0.4. The t test and Wilcoxon test were applied for the comparison
between two subtypes, while ANOVA was used for comparison among more
than two subtype. Fisher exact test was applied for the classified
variations between and among subtypes. Pearson or Spearman coefficients
were applied for the relevance between two variations. All statistical
tests were two-sided and when p <0.05, the difference was regarded as
significant.
Results
The Heterogeneity of Tumor-Associated Monocyte/Macrophage in Squamous Cell
Carcinoma of Head and Neck
A schematic diagram of the study design and principal findings is shown
in [140]Supplementary Figure S1. To classify the TAMM in HNSCC
according to their differentiation status, 19,718 genes were selected
from 39,711 leukocytes of HNSCC patients ([141]Supplementary Figures
S2A, S2B) qualified for dimensionality reduction with PCA and UMAP
(Uniform Manifold Approximation and Projection for Dimension
Reduction). The cluster classification analysis with K-NN gave rise to
four clusters, which were annotated as NK cells (n = 14,925), T cells
(n = 14,073), B cells (n = 2,789), and tumor-associated
monocyte/macrophages cells (TAMM; n = 7,924) by SingleR. The 7924 TAMM
were further classified with K-NN into seven clusters, in which the
cluster 0, 1, 2, and 4 were annotated by SingleR as tumor-associated
monocyte/macrophages cells (n = 6,875; [142]Figure 1A), while the
cluster 3 (n = 312), 5 (n = 478), and 6 (n = 259) as T cells and B
cells (data not shown). In the TAMM clusters, TAMMC0, TAMMC1, and
TAMMC2 were regarded as TAMM because of the higher CD68 expression,
while the TAMMC4 with the lower CD68 expression was considered as
dendritic cells ([143]Figure 1B). Furthermore, the mature TAM-related
genes, such as CD206, CD81 (marker of macrophage M2), TSPO, HLA-DRA,
IRF (marker of macrophage M1), and METTL14 (C1q+), were mainly
expressed in TAMMC0 and TAMMC2 (the expression in TAMMC0 was higher
than that in TAMMC2), but almost silenced in TAMMC1 ([144]Figure 1C),
indicating TAMMC0 as the mature TAM, TAMMC1 as the early
tumor-associated monocytes (TAM-M0), and TAMMC2 as the transforming
TAM-M0 from monocytes to macrophages. Differential gene expression
analysis between TAMMC1 and TAMMC0 classified 54 genes highly activated
in TAMMC1, including S100A12, S100A8, VCAN, PTGS2, and CD55, into the
early group of TAMM differentiation, and the other 51 genes robustly
expressed in TAMMC0, such as C1QB, C1QC, MMP12, and SPP1, into the late
group. Such a difference was also proven by pseudotime clustering heat
map ([145]Figure 1D, [146]Supplementary Figure S1C). By analyzing the
scRNA-seq data with GSVA and CIBERSORT, the gene function in TAMMC1 was
enriched in cellular toxicity and immune inflammation, as well as the
stemness and metabolism. In contrast, the gene function in TAMMC0 was
less enriched in stemness and metabolism, but more highly enriched in
cellular toxicity and immune inflammation, as well as the pathways of
hypoxia and angiogenesis. The enriched gene function of TAMMC2 was
medially located between TAMMC0 and TAMMC1 ([147]Figure 1D;
[148]Supplementary Figure S3). Therefore, TAMMC1 was highly scored as
early tumor-associated monocyte/macrophages and TAMMC0 as mature
macrophages.
FIGURE 1.
[149]FIGURE 1
[150]Open in a new tab
Cellular heterogeneity of tumor-associated
monocyte/macrophages/macrophages at the single cell level. (A) Umap
plots of all clusters annotated by SingleR. (B) CD68 expressions levels
of TAMM sub-clusters (C0, C1, C2, C4). (C) CD14, CD16, ITGAX, CD201,
CD81, TSPO, HLA-DRA, IRF5, SPP1, and METT14 expressions levels of TAMM
sub-clusters (C0, C1, C2). (D) GSVA revealed the enrichment scores of
TAMM sub-clusters in the pathways of tumor invasion, immunity,
stemness, and metabolism.
Differentiation Trajectory and Copy Number Variation Verified the
Heterogeneity of Tumor-Associated Monocyte/Macrophage
According to the above differential gene expression, the
differentiation trajectory of TAMM was established, in which TAMMC1 was
located in the early stage, TAMMC0 in the late stage, and TAMMC2
diffusely distributed in the early and late stages ([151]Figure 2A).
Along with the time progression, TAMMC1 was decreased with the increase
of TAMMC0, while TAMMC2 was increased and then decreased in the
diffusion maps ([152]Figures 2B,C). In the cell communication network,
the centrally located TAMMC2 exhibited a strong connection with both
TAMMC0 and TAMMC1 ([153]Figure 2D), which coincided with the finding
that TAMMC2 was located medially between TAMMC0 and TAMMC1 in the
differentiation trajectory. Similarly, CNV assay revealed that the copy
number and deficiency in TAMMC1 genome were relatively lower compared
to those in TAMMC0 and TAMMC2 ([154]Figure 2E). Therefore, in the
heterogeneous TAMM subpopulations of HNSCC, both the gene expression
profile and genomic properties indicated that TAMMC0 represented the
mature TAM, TAMMC1 stood for the early differentiating monocytes, and
TAMMC2 was the monocytes transforming into macrophages.
FIGURE 2.
FIGURE 2
[155]Open in a new tab
Differentiation trajectory and CNV changes of TAMM. (A) Monocle2
reveals the differentiation trajectory of TAMM. (B) Three-dimensional
diffusion map embedding of macrophages reveals the different
differentiation states of TAMM sub-clusters. (C) Density diffusion maps
model revealed the content of sub-clusters of TAMM at pseudo-time. (D)
Cell-Cell interaction network of different TAMM sub-clusters, node
represent TAMM sub-cluster, and the number of lines represent ligand
interactions between two sub-clusters. (E) Heatmap of the inferred CNV
in which genes were sorted by genomic location.
A Criterion Classifying Squamous Cell Carcinoma of Head and Neck With
Different Immune Phenotypes and Prognosis by Combining Tumor-Associated
Monocyte/Macrophage Differentiation and Immune cells
As mentioned above, to explore the heterogeneity of TAMM in HNSCC
patients, we identified 54 genes as the signature of early TAMM
differentiation, and another 51 genes as the signature of late TAMM
differentiation. To disclose the correlation of TAMM differentiation
with the immune phenotypes of HNSCC, we combined the differentiation
signatures with 22 immune cells to form a two-step classifier. First,
the scores of 22 immune cells estimated by CIBERSORT were applied for
the unsupervised clustering. Both the TCGA HNSCC and the [156]GSE65858
cohorts were classified into A and B subtypes (TCGA HNSCC A = 265, B =
138; GSE56858 A = 175, B = 95) ([157]Supplementary Figures S4A, S4D).
The PD-1L and IFNG expression was higher in the A subtype than those in
B subtype in both cohorts (p < 0.001, [158]GSE65858: PD-1L p < 0.1).
Second, unsupervised clustering was performed in the A subtype with the
54 genes as the early TAMM differentiation signatures and the 51 genes
as the late TAMM differentiation signatures. The unsupervised
clustering with the early TAMM differentiation signatures could divide
A subtype into three subtypes (TCGA HNSCC A1 = 40, A2 = 96, A3 = 127;
[159]GSE65858 A1 = 32, A2 = 74, A3 = 69) with different clinical
outcomes and immune signatures ([160]Supplementary Figures S4B, S4C,
S4E, S4F). Interestingly, the three subtypes from A subtype also showed
the distinct immune infiltration and immune excluded signatures. Both
the immune and stromal scores of the A1 and A3 subtypes were
significantly increased compared to those in A2 and B subtypes
([161]Supplementary Figure S5A). Moreover, the A1 subtype exhibited the
stronger immune infiltration and immune excluded signatures, the A2
subtypes in both cohorts displayed the weaker immune infiltration and
immune excluded signatures, while both the A3 subtypes possessed the
stronger immune infiltration signatures and the weaker immune excluded
signatures. In contrast to A subtype, the B subtype were weaker in
immune infiltration signatures and stronger in immune excluded
signatures ([162]Figures 3A,B,F,G). The PCA with the 54 early TAMM
differentiation signatures also supported this notion ([163]Figures
3C,H). On the other hand, the unsupervised clustering with the late
TAMM differentiation signatures failed to distinguish the immune
phenotypes of HNSCC (data not shown). Thus, the A3 subtypes were
defined as the high immune infiltration type, and the B subtype as the
high immune evasion type. The following survival assay revealed the
different prognoses among the subtypes, especially between A3 and B
subtype (p < 0.05; [164]Figures 3D,I). According to TMN staging, the A3
subtypes of both cohorts exhibited a lower ratio of IV stage patients
compared to other subtypes ([165]Figures 3E,J). To further verify the
correlation between TAMM differentiation and immune phenotypes, we
performed GSEA analysis on the differentially expressed genes between
the A3 subtype and B subtype. The genes highly expressed in the A3
subtype were enriched in immune-associated pathways, such as activation
of immune cells, adherence, proliferation, immune response, and
regulation, while the genes highly expressed in the B subtype were
enriched in cellular development and ECM-related pathways, for
instance, mesenchymal development, pattern formation, and
cytodifferentiation ([166]Figures 3K,L). These results suggested that
the early differentiation signatures of TAMM were associated with the
HNSCC immune phenotypes and prognosis.
FIGURE 3.
[167]FIGURE 3
[168]Open in a new tab
A two-step molecular classification combining the early differentiation
features of TAMM and 22 immune cell scores. (A,F) Heatmaps of
immune-related components, stromal score, and tumor purity score of the
HNSCC subtypes in TCGA HNSCC (A) and [169]GSE65858 cohorts (F). (B,G)
IFNG, PD-L1 expression level of subtype of TCGA HNSCC (B) and
[170]GSE65858 (G). (C,H) PCA of the mRNA expression of 54 early
differentiation feature genes from the HNSCC patients in the TCGA (C)
and [171]GSE65858 cohorts (H). (D,I) Kaplan-Meier curves for overall
survival (OS) of all HNSCC patients in TCGA (D) and [172]GSE65858 (I)
within A3 and B subtypes. (E,J) The pie chart showed the proportion of
TMN stages with four different immunophenotypes in TCGA (E) and
[173]GSE65858 cohorts (J). (K,L) GSEA network of DEGs in A3 vs B
subtypes using Enrichment map in TCGA (K) and [174]GSE65858 cohorts
(L).
Multiomic Characteristics Associated With the Immune Phenotypes of the
Different Squamous Cell Carcinoma of Head and Neck Subtypes
Finally, CNV, SNP, and methylation levels were examined to further
explore the immune phenotype in the subtype of TCGA HNSCC cohort. It
was found that the mutation frequency of tumor mutation loading and
tumor driver genes (TP53, TTN, etc.) in the A2 subtype was noticeably
higher than that in other subtypes ([175]Supplementary Figures S5B,
S5C). CNV analysis found that the focal copy numbers in 3p, 11q, and 2p
were significantly distinguishable between the A3 subtype and B subtype
([176]Supplementary Figures S5D–E). The methylation assay revealed that
there were 96 genes highly expressed in B subtype overlapped with the
methylation probe highly expressed in A3 subtype, while only 13 genes
highly expressed in A3 subtype were detected by the methylation probes
highly expressed in B subtype ([177]Supplementary Figures S5F–G). These
findings implicated that CNV, SNP, and methylation levels also
contributed to the different immune phenotypes in the HNSCC subtypes.
Construction of Gene Regulatory Network Based on Differential Genes Between
Squamous Cell Carcinoma of Head and Neck Subtype
To explore the mechanisms regulating the formation of different
subtypes, we compared the differential expressed genes (DEGs) between
A3 and B subtypes, and between A3 and A1 subtypes. There were 451
highly DEGs in the A3 subtype compared to the B subtype in the TCGA
HNSCC cohort ([178]Figure 4A), and 567 highly DEGs in the A3 subtype
compared to the B subtype in [179]GSE65858 cohort ([180]Figure 4B).
There were 181 overlapped genes in the two groups of the highly DEGs
([181]Figure 4C), which represented the high immune infiltration
associated genes in HNSCC. On the other hand, we obtained 659 lowly
DEGs in the A3 subtype from the comparison to the A1 subtype of TCGA
HNSCC cohort ([182]Figure 4D), and 596 lowly DEGs in the A3 subtype
from the comparison to the A1 subtype of [183]GSE65858 cohort
([184]Figure 4E). In the two groups of lowly DEGs, 41 genes were
overlapped ([185]Figure 4F). Thus, the high immune
infiltration-associated genes overlapped evidently between different
cohorts, while the immune evasion-related genes showed diversity
between different cohorts even in the instance of high immune
infiltration. By exploiting STRING database, the 181 highly and 41
lowly DEGs were constructed into a protein crosstalk net ([186]Figure
4G). Moreover, in the 41 immune evasion-related genes, those correlated
with the genes encoding extracellular matrix (POSTN, COL6A3, COL1A2,
etc.) resided in the core of the network.
FIGURE 4.
FIGURE 4
[187]Open in a new tab
Differential genes between subtypes and protein regulatory network.
(A,B,C) Volcano plot and Venn diagram show DEGs of A3 vs B in TCGA
HNSCC and [188]GSE65858 cohorts. (D,E,G) Volcano plot and Venn diagram
showed the DEGs of A3 vs A1 in TCGA HNSCC and [189]GSE65858 cohorts.
(F) PPI protein regulatory network of 181 overlapping DEGs up in A3 (A3
vs (B) and the 40 overlapping DEGs down in A3 (A3 vs A1).
Tumor-Associated Monocyte/Macrophage-Associated Chemokines-CXCL9, CXCL10, and
CXCL11-and Inflammatory Chemokine- CCLL5 Were Key Nodes in Gene Regulatory
Network
To screen out the key nodes in the gene regulatory network of the high
immune infiltration we assumed three criteria: at the center of the
regulatory network, belong to the same pathway, and highly correlated
expression. In the network constituted by the 181 highly DEGs, we
screened out 40 highly regulated genes with the Degree more than 30.
Although the correlation matrix also verified the high association
among the 40 highly regulated genes ([190]Supplementary Figure S6A),
the Degree and correlation are insufficient for the identity of the key
genes. Thus, the 40 candidate genes were arranged in the order of
Betweenness which represented the center value of the node ([191]Figure
5A) and Eigenvector according to the importance of integrating adjacent
nodes ([192]Figure 5B), respectively. Then, by comparing the first 20
genes arranged with Betweenness to the first 20 genes arranged with
Eigenvector, 12 overlapped genes were chosen as the hub genes
([193]Figure 5C). Finally, the function of these 12 hub genes were
applied for KEGG pathway correlation with CluoGO ([194]Figure 5D)
through which four chemokines (CCL5, CXCL9, CXCL10, and CXCL11) were
screened out. Although not included in the 12 hub genes, CXCL11 shared
the same family with CXCL9 and CXCL10, and was highly correlated with
their expression levels. So CXCL11 was also identified as one of the
driver genes. These four chemokines were regarded as the key node in
gene regulatory network of the high immune infiltration, because of
these characteristics: 1) the core genes with the higher Degree,
Betweenness, and Eigenvector in the network; 2) robust relevance at the
transcription level ([195]Supplementary Figures S6B–D), and the
remarkably higher difference in CXCL9, CXCL10, and CXCL11 (A3 vs B)
than the other eight genes ([196]Supplementary Figure S6E); and 3)
belong to the same pathway and share the higher topological signs.
Moreover, during TAMM differentiation, CXCL9, CXCL10, and CXCL11 were
increased with the time progression in Pseudotime analysis ([197]Figure
5E). Taken together, the four chemokines with the strongest functional
co-regulation and co-expression could be regarded as the pivotal genes
screening the high immune evasion of HNSCC.
FIGURE 5.
[198]FIGURE 5
[199]Open in a new tab
The topological feature of the network composed of 181 genes. (A)
Dotplot of -log2(Degree+1) and log2(Betweenness+1) in selected 181
nodes. (B) Dotplot of log2(Degree+1) and log2(Eigenvector+1) in
selected 181 nodes. (C) Venn diagram of top 20 genes in A or (B) (D)
KEGG pathway association network of 12 hub genes. (E) Trend of mRNA
expression levels of the four chemokines (CXCL9,10,11, and CCL5)
following the differentiation trajectory of TAMM. (F) Aggregation of
ATAC-seq peaks of the four chemokines of nine patients in the
transcription initiation region. (G) Methylation levels of the four
chemokines in different HNSCC subtypes.
The Transcription of the four Chemokines-CXCL9, CXCL10, CXCL11, and CCLL5-Was
Influenced by Epigenetic and Health Factors
To further explore the endogenous (epigenetic) and exogenous (health
manner) factors impacting the expression of the four chemokines-CXCL9,
CXCL10, CXCL11, and CCL5--the chromatin accessibility and the
methylation status at the transcription initiation regions of CXCL9,
CXCL10, CXCL11, and CCL5 were analyzed in the whole genome with TCGA
HNSCC ATAC and methylation database. The enriched reads were evidently
concentrated at the transcription initiation regions of CXCL9, CXCL10,
CXCL11, and CCL5 in the A3 subtype, compared to the A1 and A2 subtypes
([200]Figure 5F), implicating a more active transcription of CXCL9,
CXCL10, CXCL11, and CCL5 in the A3 subtype. In contrast, the
methylation levels of CXCL9, CXCL10, CXCL11, and CCL5 showed
insignificant difference among the subtype, implying that the four
chemokines were epigenetically regulated by the manners rather than DNA
methylation. However, the methylation of CXCL9, CXCL10, CXCL11, and
CCL5 in the control patients were higher ([201]Figure 5G), suggesting
de-methylation of the four chemokines was crucial for HNSCC genesis. We
also found that the transcription and methylation levels of CXCL9,
CXCL10, CXCL11, and CCL5 were correlated with age, smoking, alcohol
consumption, and HPV infection. The higher mRNA levels of the four
chemokines were detected in the HNSCC population with older age and
lower consumption of tobacco and alcohol ([202]Supplementary Figure
S7A, S7C, S7E; t test, p < 0.05). The HPV positive HNSCC group
exhibited a higher CXCL10 mRNA level and an increased methylation of
CXCL9 and CXCL11 compared with the HPV negative HNSCC group
([203]Supplementary Figures S7B, S7D; t test, p < 0.05). Thus, it was
concluded that, although not associated with the immune phenotypes of
HNSCC, the transcription of the four chemokines regulated by DNA
methylation and health factors were critical for HNSCC genesis.
The Four Chemokines-CXCL9, CXCL10, CXCL11, and CCL5-Positively Regulated
Immune Responses and Were Associated With the Low CNV and CASP8 Mutations in
the Squamous Cell Carcinoma of Head and Neck Genome
According to the mRNA levels of the four chemokines, six HNSCC cohorts
(one TCGA cohort and five GEO cohorts) were graded with GSVA, and then
divided into the high- and low-graded groups with the median grade to
evaluate the correlation of the four chemokines with immune response.
There was a remarkable difference between the high- and low-graded
groups in the immune signature and genome. The TCGA and most
high-graded cohorts showed a higher enrichment of immune cells
(Macrophages M1, CD4 T cells memory activated, and CD8 T cell) in the
infiltration grading of the 22 kinds of the immune cells, got higher
scores in the enrichment of antigen present during tumor immune circle,
immune cell infiltration, and the recognizing and killing of tumor
cells by effector T cells in the 29 immune signatures assay
([204]Figure 6A), and was given the lower scores in the TIDE assay. All
of the results suggested a better response to immune therapy and was
verified by the cohorts of immune therapy, in which the GSVA grades of
the four chemokines in the CR group were higher than those in PR, SD,
and PD groups ([205]Figure 6B). Based on these findings, we classified
HNSCC into the immune hot and immune cold phenotype according to the
GSVA grades of the four chemokines. Then, we estimated the distribution
of the mutations from the first 30 HNSCC driver genes with the highest
frequency of mutation (TP53, TTN, CSMD3, SYNE1, etc.,) in the hot and
cold immune groups, and found that except for CASP8, all other driver
genes had an elevated frequency of mutation in the low-graded group
([206]Figure 6C; [207]Supplementary Table S7), implying that the
mutations of CASP8 endowed HNSCC with a stronger immunity. Moreover,
CNV analysis revealed that in the immune cold group, an active CNV was
detected in several hot spot regions (gain: 3p, 8q, 17q, 18p. loss: 2q,
7q, 13q) ([208]Figure 6D). In combination with Kech classification, we
found that the most immune hot was BA type, while the most immune cold
was CL type ([209]Figure 6E). All the above results suggested that the
four chemokines could not only act as the markers identifying the HNSCC
with high concentration of immune cells (CD8 T cell, Macrophage M1,
etc.), but also reflect the HNSCC characteristics comprehensively. It
was also suggested that the immune hot subtype of HNSCC could enhance
the immune responses through CASP8 mutations, and the immune cold
subtype also circumvented immune responses through gene mutations.
Further exploration on the crosstalk among the four chemokines, TME,
and immune cells in the immune circle by analyzing the relevance in
TCGA cohort disclosed that CXCL9, CXCL10, CXCL11, and CCL5 showed a
strongly positive association with immune cells (Macrophages M1, CD4 T
cells memory activate and CD8 T cells), and the three stages of tumor
immune circle ([210]Figure 6F). Since the similar association was also
detected in other HNSCC cohorts, the four chemokines were proven to
enhance the anti-tumor immune capability. Moreover, we also found that
macrophages M1 was strongly positively associated with the activation
of dormant CD8 and CD4 T cells ([211]Figure 6G).
FIGURE 6.
[212]FIGURE 6
[213]Open in a new tab
The roles of the four chemokines (CXCL9,10,11, and CCL5) in HNSCC TME
and their association with tumor genomic changes. (A) Dotplot
summarized the scores of 22 immune cells estimated by GIBESORTRT and
GSVA scoring on the fold change and p.adjust of 29 immune signature
between the immune hot and cold group classified by the median GSVA of
the four chemokines (only p < 0.05 was given). (B) Vilionplot
summarized the GSVA scores of CXCL9, CXCL10, CXCL11, and CCL5 of four
outcomes (CR, PR, PD, SD) in the cohort receiving anti-PD-1/PD-L1
immunotherapy. (C) The OncoPrint was constructed between high and low
scores of the top 30 genes with the highest mutation frequency. (D) CNV
plot showed the frequency of copy-number gains (red) and deletions
(blue) among immune hot and cold groups of the TCGA-HNSC cohort. (E)
The stacking histogram showed the distribution of Keck classification
in the immune hot and cold groups of TCGA HNSCC and gse65858 cohorts.
(F) Correlation between the four chemokines and GIBESORTRT score of 22
immune cells in HNSCC-TCGA and 29 immune signature GSVA score. The cell
charts in the upper-right triangular exhibited the correlation among
the 22 immune cell scores, and the cell charts in the -lower triangular
showed the correlation among the 17 immune signature scores (Red stood
for positive and green for negative, the darkness and lightness of the
colors for the high and low of the coefficients, and the size of the
cell for p value). The lines between two cell charts represent the
correlation of the mRNA of the four chemokines with the bilateral
immune scores (Red stood for positive and green for negative, and the
thickness of the lines for the high and low of the coefficients). (G)
Dotplot summarized the correlation coefficients between the scores of
21 immune cells estimated by GIBESORTRT and the scores of Macrophage M1
estimated by GIBESORTRT (only p < 0.05 was given). (H,J) Heatmap of
average expression level of ligand–receptor interactions in all
clusters in [214]GSE103322 and [215]GSE139324. (I,K) Histogram of
expression levels of CCL5, CXCL9, CXCL10, CXCL11, CXCR3, and CCR5 in
each cell in [216]GSE103322 and [217]GSE139324 cohorts. (L) Dotplot
summarized the correlation coefficients between the expression level of
CXCL9, CXCL10, CXCL11, and CCL5, and the GSVA of 10 immune signatures
in each cells classified as TAMM.
The Relationship Between the Four Chemokines and the Sub-Populations in the
Squamous Cell Carcinoma of Head and Neck Tumor Micro-Environment at Single
Cell Level
The cohorts of [218]GSE10332 and [219]GSE139324 (the [220]GSE10332
cohort contained all kinds of cells in TME, while the [221]GSE139324
cohort only contained infiltrative leukocytes) were applied for the
relationship assay at the single cell level to disclose the effects of
CXCL9, CXCL10, CXCL11, and CCL5 on the HNSCC sub-populations in TME.
Cell communication assay with CellPhoneDB found that TAM, DC,
tumor-associated fibroblasts (CAF), and malignant epithelium cells
communicated with themselves and other cells intensively ([222]Figures
6H,J). Interestingly, CXCL9, CXCL10, and CXCL11 were expressed robustly
in TAM, malignant epithelium cells, and CAFs, but weakly in CD4 and CD8
T cells. However, CXCR3, the common receptor for CXCL9, CXCL10, and
CXCL11, was expressed in CD4 and CD8 T cells ([223]Figure 6I),
indicating that CD4 and CD8 T cells were chemo-attracted to immune
focus by the CXCL9, CXCL10, and CXCL11 emanated from TAM, malignant
epithelium cells, and CAF. Moreover, CCL5 and its receptor, CCR5, were
mainly expressed in CD4, CD8 T cells, and NK cells (especially CD8 T
cells; [224]Figure 5K), suggesting that CCL5 influenced T cells through
autocrine or paracrine. Worthy of note, the mRNA peaks of CXCL9,
CXCL10, CXCL11, and CCL5 in TAM were distributed in multiple
sub-populations ([225]Figure 6K), suggesting that although the four
chemokines were highly correlated at the bulk RNA-seq level, such
correlation could be inconsistent at the single cell level due to the
heterogeneity of TAMM. Finally, the relevance assay on TAM clusters at
the single cell level disclosed that the mRNA levels of the four
chemokines were positively correlated to the pathways involved in
immune infiltration and immune inflammation response ([226]Figure 6L).
Therefore, the expression of the four chemokines were associated with
the immune sub-populations in TAMM.
The Immune Hot Subtype of Squamous Cell Carcinoma of Head and Neck
Characterized by the High Expression of the Four Chemokines was Sensitive to
Anti-Cancer Drug Targeting ERK1-MARK and RAS Pathway
The GSEA assay on the DEGs between the high- and low-graded groups of
the six HNSCC cohorts revealed that the pathways enriched in the
high-graded groups were mainly involved in chemoattraction and immune
response of immune cells, as well as cell proliferation and
differentiation, which were consistent in different cohorts. In
contrast, the pathways enriched in the low-graded groups were
relatively sparse in different cohorts, though mainly concentrated in
tumorigenesis and progression, such as stemness, metastasis,
metabolism, and hypoxia ([227]Figure 7A). The relevance assay on the
IC50 of the HNSCC cell lines treated with anti-tumor drugs in GDSC
database disclosed that the GSVA scores of CXCL9, CXCL10, CXCL11, and
CCL5 were negatively correlated to ERK1-MARK ([228]Figures 7B–H) and
RAS pathway ([229]Figures 7I,J) targeted by anti-HNSCC drugs,
implicating that the susceptibility of the immune hot subtype of HNSCC
to the drug originated from the influence on ERK1-MARK and RAS pathway.
FIGURE 7.
[230]FIGURE 7
[231]Open in a new tab
Enriched tumorigenesis pathways in immune hot and cold phenotypes
determined by the four chemokines (CXCL9,10,11, and CCL5) and immune
hot sensitive drugs. (A) Dotplot summarized the GSEA NES scores of
signal pathways related to tumor immunity and tumorigenesis between
immune hot and cold groups in the six HNSCC cohorts (only p.adjust<0.05
were given). (B–J) Scatter diagram summarized the correlation
coefficients between GSVA score of CXCL9, CXCL10, CXCL11, and CCL5, and
the log[2](IC50) in seven drugs targeting ERK-MAPK pathway (B–I) and
two drugs targeting RAS pathway (I, J).
A Neural Network Predicting Model With the Four Chemokines and Three Immune
Cells
Through the above studies, we found out a strongly positive correlation
between four chemokines (CXCL9, CXCL10, CXCL11, and CCL5) and three
kinds of immune cells (Macrophage M1, CD8, and CD4 T cells) in the
immune phenotypes of HNSCC. Since the above seven factors were located
in the core of the immune response in HNSCC, we designed a 4-layer
neural network to predict the response (response: CR, PR. not response:
SD, PD) to the treatment with auti-PD-1/PD-L1 ([232]Figure 8A), in
which [233]GSE154538, [234]GSE141119, [235]GSE91061, [236]GSE78220,
[237]GSE176307, and the IMvigor210 acted as the training set (n = 501),
and [238]GSE93157 as the test set (n = 65). The AUC of the training set
and the test set in the prediction model reached 95% and 74.6%,
respectively ([239]Figure 8B). The confusion matrix of the training and
test sets was displayed in [240]Figures 8C,D.
FIGURE 8.
[241]FIGURE 8
[242]Open in a new tab
The four chemokines and three immune cells were used to construct a
neural network model to predict the response to anti-PD-1/PD-L1
immunotherapy. (A) Schematic diagram of the neural network. (B) ROC
plot of train cohort and test cohort. (C,D) The confusion matrix in
test cohort and train cohort.
Discussion
Despite the great progress made by ICB in multiple tumors, only a
minority of HNSCC patients have benefited from ICB therapy. It is of
major importance to characterize the HNSCC sub-population susceptible
to ICB therapy, and the key genes maintaining the sub-population
([243]Curran et al., 2010). Although a variety of criteria were
proposed previously, few of them concern the association of TAMM with
the immune responses in HNSCC. Since TAMM in TME contributed greatly to
tumorigenesis and progression ([244]Estko et al., 2015; [245]Gao et
al., 2016), a lot of researchers focused on the potentials of TAMM in
immune therapy ([246]Coifman et al., 2005). However, most previous
studies and strategies ignored the heterogeneity of TAMM, but treated
the TAMM as a static entity, which meant the immune therapy was
challenged by TAMM heterogeneity. In the present study, we applied
bioinformatical methods from multiple dimensions to explore the
heterogeneity of the TAMM during differentiation, as well as its
correlation with the immune responses in HNSCC. Although CIBEOSORT was
not reported to be applied in the matrix of single-cell RNA-Seq, we
think that, according to the resolve of non-negative matrix in the
algorithm theory of CIBEOSORT, the bulk RNA-seq could be resolved into
the matrix of single-cell RNA-Seq timing the matrix of cell type
clusters. In our study, the single-cell RNA-Seq matrix, namely, the
Leukocyte signature matrix (LM22), was set as the decision variable,
and the single sample in the bulk RNA-seq as response variable for SVM
linear deconvolution. The outcome weighted vector was regarded as the
cell type abundance of each sample. Therefore, it is sound to transform
the bulk RNA-seq matrix into Single-cell RNA-Seq matrix by treating
each cell as a sample in the bulk RNA-seq. By treating the Single-cell
RNA-Seq data with the above algorithm theory, we found that combining
the genes characterizing early TAMM differentiation with the immune
cells in HNSCC could provide a criterion classifying the immune
subtypes of HNSCC more precisely. Furthermore, the four
chemokines-CXCL9, CXCL10, CXCL11, and CCL5-were identified not only as
the driver genes initiating and maintaining the immune hot subtype of
HNSCC, but also the nodes connecting TAM (macrophage M1), CD4, and CD8
T cells together.
The heterogeneity of TAMM in HNSCC was found to result from not only
the different subpopulations, but also the different stages during
differentiation. Molecularly, the gene expression and genomic
constitution of TAMM underwent remarkable alterations during the
differentiation from tumor-associated monocyte to tumor-associated
macrophages, which was also supported by the in vitro assay
([247]Singhal et al., 2019). Previously, a criterion exploiting 22
kinds of immune cells was once proposed to classify HNSCC into the high
and low immune enrichment subtype ([248]Feng et al., 2020; [249]Zhang
et al., 2020), however, it was too broad to characterize the immune
subtype and prognosis. To establish a practical and accurate criterion,
we divided the gene expression profile during TAMM differentiation into
the early and later groups, and found out the DEGs between the early
and later groups. By using multiple bioinformatical methods, the DEGs
in the early TAMM differentiation were found, indicating a better
prognosis in the subtype with high immune infiltration and low immune
evasion ([250]Sanmamed and Chen., 2018). Thus, the DEGs in early TAMM
differentiation could reflect more details in the immune infiltration,
evasion (which could estimate the immune phenotype more
comprehensively) ([251]Turan et al., 2021), and prognosis of HNSCC.
Previous studies reported a correlation of the better prognosis with
the elevated tumor mutation burden (TMB) ([252]Chan et al., 2015;
[253]Spencer et al., 2016; [254]Büttner et al., 2019). However, in our
study, the A2 subtype which had the highest TMB in the TCGA HNSCC
cohort displayed the lower immune infiltration and evasion, implicating
the insusceptibility to ICB therapy. Since similar characteristics were
also detected in another HNSCC clinical cohort ([255]Kim et al., 2020),
it was still unclear whether the HNSCC subtype with a higher TMB was
susceptible to PD-1/PD-L1 therapy.
The DEGs between A3 and B subtype (high immune infiltration vs high
immune evasion) were applied to construct the protein crosstalk network
from which the genes highly correlated in expression at the nodes of
regulatory network and sharing the same pathway were identified as the
driver genes initiating and maintaining the immune subtype. The DEGs
between the high immune infiltration and high immune evasion (A3
subtype vs B subtype) showed a high similarity in different cohorts. In
contrast, the DEGs between high immune infiltration subtype (A1 vs A3
subtype) were different from one another in different cohorts.
Therefore, we concentrated on the DEGs between the high immune
infiltration and high immune evasion (A3 subtype vs B subtype) to
explore the key genes regulating the immune hot phenotype. Since both
the A3 and A1 subtypes stood for the high immune infiltration subtypes
in the two cohorts, the difference between A3 and A1 subtypes
represented the discrepancy between the high immune infiltration
subtypes. From [256]Figure 5G, it can be seen that such discrepancy
could be partially attributed to the various extents of the immune
evasion. However, because the number of the intersected DEGs in the A3
and A1 subtypes was too low to contribute to the difference, the
factors resulting in the difference were implicated varying
dramatically in different cohorts. Since the A3 and B subtypes in the
two cohorts exhibited the converse immune signatures, their comparison
was supposed to get the core genes associating immune infiltration with
the HNSCC. The numerous overlapped DEGs from the two cohorts reflecting
the similarity in the high immune infiltration between the two cohorts
allowed the following exploration of the pivotal genes in the HNSCC
with immune infiltration phenotype. Taking all above findings into
account, we concluded that the TAMM differentiation related
chemokines-CXCL9, CXCL10, and CXCL11, and inflammatory chemokine
CCL5-were the driver genes initiating and maintaining the high immune
infiltration phenotype of HNSCC. A series of previous reports supported
that the four chemokines could work as the potential targets of immune
therapy. During tumorigenesis and progression, the CXCL9-11/CXCR3 axis
regulated the differentiation and chemoattraction of T cells
([257]Tokunaga et al., 2018), and the CCL5/CCR5 axis influenced growth
and metastasis ([258]Aldinucci et al., 2020). Previous reports showed
that CXCL9, CXCL10, and CCL5 could mark T cell–inflamed phenotype of
pancreatic cancer ([259]Romero et al., 2020). CXCL9 and CCL5 activated
immune responses and enhanced ICB therapy in mouse model of ovary
cancer ([260]Dangaj et al., 2019), and the melanoma in CXCR3 knock-out
mice exhibited decreased immune infiltration and poor prognosis
([261]Korniejewska et al., 2011). However, we have to acknowledge that
there was subjective opinion in the criteria of the pivotal genes.
Actually, besides the four chemokines, the other eight candidate hub
genes were also verified to act as the pivotal genes establishing and
maintaining the high immune infiltration to some extent. However, we
think that CXCL9, CXCL10, CXCL11, and CCL5 were the optimal
combination, because they satisfied the three criteria: 1). they shared
multiple pathways, implicating that they could collectively reflect the
activity of immune pathway, instead of independently. Other genes were
located in different pathways, which raised the uncertainty for their
function, though they possessed the higher network topology; and 2).
CXCL9, CXCL10, CXCL11, and CCL5 were highly correlated in expression.
We tested the correlated expression of the 181 overlapped DEGs in TCGA
cohorts and confirmed a higher correlation among CXCL9, CXCL10, and
CXCL11 than among other DEGs. Although CXCL11 was excluded from the 12
candidate genes with the highest topological signatures, it had the
higher correlated expression, belonged to the same family and shared
multiple pathways with the other three chemokines. Therefore, we
selected CXCL9, CXCL10, CXCL11, and CCL5 as the pivotal genes in the
high immune infiltration of HNSCC for the following study.
Because the roles of the four chemokines in HNSCC have been little
studied, to explore their pivotal effects in the immune responses to
HNSCC, we divided the immune circle into infiltration and evasion
stages. The immune infiltration included: 1) the release and convey of
tumor cell antigen, 2) chemoattraction and infiltration of immune cells
into TME by the cytokines and inflammation, and 3) recognition and
elimination of HNSCC cells by CTL. With the exhaustion of CTL and the
expression of immune suppression factors (TGF-beta, PD-L1, etc.) by
HNSCC, the immune evasion commenced. The bulk RNA-seq data revealed
that CXCL9, CXCL10, CXCL11, and CCL5 were positively correlated to all
three steps of immune infiltration, but negatively to immune evasion.
The scRNA-seq data further disclosed that the four chemokines and their
receptors were highly expressed in DCs, enhancing the antigen
presentation in TME; the high CCL5 expression in CD8 T cells, NK cells,
and certain TAM promoted inflammation; the tumor cells highly
expressing CXCL9, CXCL10, and CXCL11 attracted the CD8 T cells, NK
cells, and TAM, which eliminated tumor cells by perforating cell
membrane, digesting through serine proteases and apoptosis via ligand
binding ([262]Lee et al., 2018). All of these results supported the
role of the four chemokines in initiating and maintaining the immune
hot phenotype of HNSCC.
According to the GSVA scores of the four chemokines, the HNSCC cohorts
were divided into the subtypes of immune hot and immune cold. The
immune cold subtype was characterized by the evident alteration on CNV,
which was correlated to tumor invasion and decreased immune responses
([263]Davoli et al., 2017). This finding also implied that the
decreased immune responses in immune cold subtype resulted from the
imbalanced immune gene expression caused by CNV alteration, as opposed
to the dysfunction of single immune gene. The other sign of immune cold
subtype was the higher mutation frequency, including the tumor driver
genes of TP53, TTN, etc. Reversely, the frequency of CASP8 in immune
hot subtype was noticeably higher than that in immune cold subtype.
Since several studies demonstrated that the CASP8 mutations
characterized the local activation of immune cells and inflammation
([264]Rooney et al., 2015; [265]Tummers and Green., 2017), it suggested
that CASP8 mutation was capable of activating immune responses in the
immune hot subtype.
Worthy of note, the inflammation resulting from the four chemokines
could also increase the tumor invasion. As shown in previous studies,
the VEGF-PIK3/AKT pathway activated by CCL5 promoted tumor metastasis
by stimulating angiogenesis and ECM remodeling ([266]Karnoub et al.,
2007; [267]Wang et al., 2012), and the robust expression of CXCL10 also
enhanced gastric cancer invasion and metastasis by binding the receptor
CXCR3A ([268]Yang et al., 2016). Thus, further exploration was still
required to elucidate the relationship between the four chemokines and
HNSCC prognosis. Additionally, the four chemokines were strongly
associated with the check points on HNSCC surface, implicating that
HNSCC cells could circumvent immune attacks by conveying the signal of
“Don’t eat me” to CTL through the check point molecules (PD-L1, PD-1,
etc.). Thus, the four chemokines might also indirectly enhance the
tumor invasion and metastasis even when directly attacked by tumor
cells. Since CCL5 was reported to rapidly induce cyclin D1, c-Myc,
Ha-Ras through MARK-ERK, and Jak/STAT signaling, as well as glucose
in-take and ATP production to stimulate tumor growth ([269]Ding et al.,
2016; D. ; [270]Gao & Fish, 2018; [271]Murooka, Rahbar, & Fish, 2009),
the HNSCC cell lines with the high expression of the four chemokines
were sensitive to the anti-tumor drug targeting MARK-ERK and RAS
pathways.
The four chemokines were associated positively with Macrophages M1,
activated CD4, and CD8 T cells, but negatively with Macrophages M0 (in
TCGA HNSCC cohort) and tumor-associated monocyte/macrophages (GEO
cohort), suggesting the lower expression of the four chemokines in the
early tumor-associated monocyte/macrophages or macrophages M0, and the
gradually elevated expression in differentiating TAMM. Moreover, the
higher expression of the four chemokines were intensively detected in
macrophage M1, instead of M2, also implicating their association with
the polarization of macrophages, which was supported by the recent
studies that found that the lack of CCL5 promoted the polarization in
macrophages M2 (M. [272]Li M et al., 2020), and CXCL9 and CXCL10
induced the polarization in macrophages M1 ([273]Kohler et al., 2019).
The scRNA-seq revealed that the mRNA peaks of the four chemokines in
TAM were distributed in different subpopulations of TAMM, suggesting
that the strong and exclusive co-regulation of the four chemokines
disclosed by the bulk RNA-seq were inconsistent with the single cell
level, which required further investigation.
In the immunotherapy cohort receiving anti-PD-L1/PD-1, the mRNA levels
of the four chemokines were elevated significantly in CR group,
verifying their positive roles in immune responses. It also encouraged
us to establish a criterion predicting the patients’ responses to ICB
therapy. In the premise of the substantial immune capacity of resisting
tumors, ICB therapy facilitated T cells to eliminate tumor cells by
blocking immune evasion, namely, the validity of ICB therapy depended
on the patients’ immunity. Considering the crucial roles of the four
chemokines (CXCL9, CXCL10, CXCL11, and CCL5) and the three pivotal
immune cells (Macrophages M1, CD 4, and CD8 T cells) in tumor immunity,
we established a criterion predicting the validity of ICB therapy
through neural network, in which the AUC in training and test sets
achieved 100% and 74%, respectively.
We have to acknowledge the shortcomings in this study. First, although
they have been verified in other tumors, work is still required to
verify the roles of the four chemokines in HNSCC found by the
bioinformatical analyses. Second, the exploration on the mechanisms
resulting in the immune evasion in the high immune infiltration subtype
was insufficient. Third, the predicting and verifying cohorts were not
HNSCC cohorts, which might weaken the prediction accuracy in HNSCC
cohorts because of the variations among tumors. Fourth, despite the
impressive promotion in tumor immunity, the roles of the four
chemokines-CXCL9, CXCL10, CXCL11, and CCL5-in tumorigenesis and
progression were still in debates.
Conclusion
In summary, we indeed established the core roles of the four
chemokines-CXCL9, CXCL10, CXCL11, and CLL5-in HNSCC immunity by
combining TAMM differentiation and HNSCC TME. From the perspective of
the four chemokines associating TAMM with HNSCC immunity, we found the
limitation of treating TAM as a static entity and the potential values
of the four chemokines in tumor immunity. Considering the few studies
on HNSCC immunity, our present study provided bio-informatics support
for future explorations.
Acknowledgments