RNA-seq and bulk RNA-seq data analysis of cancer-related fibroblasts (CAF) in LUAD to construct a CAF-based risk signature
RNA-seq and bulk RNA-seq data analysis of cancer-related fibroblasts (CAF) in LUAD to construct a CAF-based risk signature
com/scientificreports
Abbreviations
NSCLC Non-small cell lung cancer
LUAD Lung adenocarcinoma
ICIs Immune-checkpoint inhibitors
TME Tumor microenvironment
CAFs Cancer associated fibroblasts
ScRNA-seq ScRNA-seq Single-cell RNA-sequencing
GEO Gene Expression Omnibus
UMI Unique Molecular Identifier
TCGA The Cancer Genome Atlas
1Department of Radiology, Shandong Cancer Hospital and Institute, Shandong First Medical University, Shandong
Academy of Medical Sciences, Jinan, Shandong, China. 2Department of Oncology, Binzhou Medical University
Hospital, Binzhou, Shandong, China. 3Department of Radiation Oncology, Shandong Cancer Hospital and Institute,
Shandong First Medical University, Shandong Academy of Medical Sciences, Jinan, Shandong, China. email:
[email protected]
Lung cancer is estimated to contribute to 25% of cancer-associated deaths, and non-small cell lung cancer
(NSCLC) alone accounts for 85% of cases1, lung adenocarcinoma (LUAD) and squamous carcinoma are two
major subtypes of NSCLC. Metastatic LUAD treatment has been revolutionized by immunotherapy since it was
approved by the FDA in 2015. Despite many factors associated with intrinsic resistance to immune-checkpoint
inhibitors (ICIs), LUAD has been hampered by a lack of predictive biomarkers and limited understanding of how
ICIs affect the disease3. It was possible to predict the immunotherapy clinical outcomes of LUAD using omic
data-derived signatures4. Consequently, multigene signatures can be valuable for predicting LUAD outcomes.
Tumor microenvironment (TME) is a specialized ecosystem of host components designed by tumor cells to
support the development and metastasis of tumors5. There are diverse immune cell types in the TME, as well as
cancer-associated fibroblasts, endothelial cells, pericytes, and other types of cells residing in the tissue. Host cells
were once thought to play no role in tumorigenesis, but are now known to play a crucial role in the development
of cancer6. In order to understand the spatial and temporal regulation of immune therapeutic interventions, it
is important to gain a deeper understanding of the diversity of immune cells, stromal components, repertoire
profiling, and the neoantigen prediction of TME5. TME is dominated by cancer associated fibroblasts (CAFs)
that affect cancer features7. As a result of growth factors, inflammatory ligands, and extracellular matrix proteins,
they promote the proliferation of cancer cells, the resistance to therapy, and the exclusion of the immune
system8. With CAFs being highly heterogeneous, it remains unclear how cancer treatments can be personalized
based on CAFs in a patient’s tumor as they are highly heterogeneous9. Although many studies have investigated
CAF characteristics in LUAD, the relationship between CAF characteristics and prognosis and immunotherapy
response remains poorly understood10–12.
From the TCGA databases, we obtained LUAD single-cell RNA-sequencing (scRNA-seq) data and
transcriptome data. In this study, we distinguished CAF subclusters and identified CAF-based risk signatures
for patients with LUAD who received immunotherapy. An analysis of the immunologic landscape and
responsiveness to immunotherapy underlying the CAF-based signature was conducted to determine its clinical
relevance. A novel nomogram has been created to facilitate the clinical application of CAF features in LUAD
prognosis combining the CAF-based risk signature and clinicopathological features. The study may provide
new insights into the pathophysiology of LUAD, allowing for more tailored treatments and better outcomes for
patients.
Definition of CAF
A comprehensive CAF signature was characterized using the Seurat package using scRNA-seq data from LUAD.
Firstly, we removed the cells with over 6000 or below 250 expressed genes, then we normalized the expression of
these genes based on log ratios. Using the FindIntegration Anchors function, batch effects were eliminated for 4
samples. With a resolution of 0.2, the uniform manifold approximation and projection method was used for non-
linear dimensional reduction. By using the FindNeighbors and FindClusters functions, cells were clustered into
different subgroups by using the functions of FindNeighbors and FindClusters (dim = 40 and resolution = 0.2).
RunTSNE was then used to reduce the dimensions using t-distributed stochastic neighbor embeddings.
Four marker genes were identified in fibroblasts, including ACTA2, FAP, PDGFRB, and NOTCH312,14–16.
FindNeighbors and FindClusters functions were used to re-cluster the fibroblasts. Moreover, TSNE was used
to lessen the dimensionality of fibroblast clusters. By comparing one CAF cluster with the others and using
the FindAllMarkers function with logFC = 0.5, minpct = 0.35, and an adjusted p value < 0.05, we were able to
determine the markers of each cluster. Using the clusterProfiler software(Version 4.12.6; https://yulab-smu.
top/contribution-knowledge-mining/), the Kyoto Encyclopedia of Genes and Genomes (KEGG) enrichment
analysis was carried out on the marker genes of CAF clusters, the “clusterProfiler” R package was utilized to
conduct KEGG analyses17–19. The CNV characteristics within CAF clusters were analyzed using the CopyKAT R
package in order to differentiate between tumor cells and normal cells.
Statistical analysis
The statistical analysis was performed using R software (version 3.6.3). Spearman correlation and Pearson
correlation were used to analyze the correlation matrices. Comparing the two groups was carried out using the
Wilcoxon test. A Log-rank test was used to compare survival differences using K-M curves. P-value < 0.05 was
considered statistical significance.
Results
scRNA-seq analysis of CAFs
A detailed analysis of data preprocessing can be found in Figure S1. Using four marker genes, including ACTA2,
FAP, PDGFRB, and NOTCH3, nine CAF populations were identified after log-normalization and dimensionality
reduction (Figures S1A, B). We extracted the cells of 5 CAF populations for further clustering and dimensionality
reduction. We further clustered CAF populations using the same clustering algorithm and identified five clusters
(Figures S1C, D). All five CAF clusters did not express the epithelial cell specific gene, supporting accurate
CAF identification (Figure S2). UMAP plots of 4 sample distributions are shown in Fig. 1A. This resulted in the
generation of five CAF clusters that were used for further analysis (Fig. 1B). 735 DEGs were identified among
the 5 CAF clusters, and Fig. 1C shows the expression of the top 5 DEGs (determined as markers of CAF clusters)
in the 5 clusters. Figure 1D illustrates the proportion of the 5 clusters in each cohort. According to Fig. 1E, these
DEGs were enriched in multiple pathways, including ECM-receptor interaction, focal adhesion, ribosome, and
protein digestion and absorption. According to the CNV characteristic of the 5 CAF clusters, 347 tumor cells
and 274 normal cells are present (Fig. 1F).
Fig. 1. The identification of CAF clusters based on a scRNA-seq ddata of LUAD patients. (A) Distribution
of 4 samples as represented by uMAP; (B) Distribution of five fibroblasts by uMAP after clustering; (C) Dot
plot of top 5 gene expression markers of subgroups; (D) CAF proportion and number of subgroups for cancer
tissue and metastatic lymph node cells; (E) KEGG enrichment analysis of 5 subsets of fibroblasts; (F) uMAP
distribution map of malignant and non-malignant cells predicted by copykat package.
malignant cells than the other four clusters. Among the ten tumor-related pathways with in each CAF cluster, we
found slight differences in GSVA scores between malignant and non-malignant cells (Fig. 2 C-G).
We first calculated the ssGSEA score of the marker genes (the top 5 DEGs of CAF clusters in Fig. 1C) of each
CAF cluster based on the TCGA cohort to determine the associations between CAF clusters and prognosis.
The CAF cluster scored significantly higher in normal samples than in tumor samples (Fig. 3A). By using the
survminer R package, LUAD samples from the TCGA dataset were divided into high- and low-CAF score groups.
CAF_1 and CAF_2 clusters had a better prognosis in the high-CAF score group than those in the low-CAF
score group, CAF_0, CAF_3 and CAF_4 did not have any effect on the prognosis (Fig.3 B). CAF_1 and CAF_2
distribution in tumor tissue decreased as T stage increased, whereas CAF_0, CAF_3, and CAF_4 distribution
did not change as T stage increased. CAF_1 and CAF_2 clusters may have lot impact on LUAD progression (Fig.
3 C).
Fig. 2. Characteristics of tumor-related pathways in CAF clusters. (A) Heatmap of 10 tumor-related pathways
enriched in CAF cells; (B) Comparison of CAF clusters in malignant and non-malignant cells; Comparison of
GSVA scores between malignant and non-malignant cells in CAF_0 (C), CAF_1 (D), CAF_2 (E), CAF_3 (F)
and CAF_4 cluster (G). (wilcox.test, *P < 0.05; **P < 0.01; ***P < 0.001; ****P < 0.0001and ns, not significant).
related CAF clusters. Additionally, 234 genes exhibited prognostic values based on univariate Cox regression
analysis (Fig. 4B). The number of genes was narrowed down by Lasso Cox regression analysis, and 8 genes
retained lambda = 0.0185 (Fig. 4C). Finally, we included eight genes in the risk signature, including cell
division cycle 25 C (CDC25C), exonuclease 1 (EXO1), cell cycle-related proteins cyclin B1 (CCNB1),
dihydropyrimidinase-like 2 (DPYSL2), methyltransferase like 7 A (METTL7A), C-Type Lectin Domain
Family 3 Member B (CLEC3B), brutons tyrosine kinase (BTK), glutamate receptor (GRIA) after multivariate
Cox regression analysis using stepwise regression (Fig. 4E). The final 8-gene signature formula is as follows:
RiskScore = -0.183*BTK + 0.051*CCNB1 + 0.116*CDC25C + 0.077* CLEC3B+-0.01*DPYSL2 + 0.086*EXO1+
-0.484*GRIA1+-0.04*METTL7A. After z-mean normalization, we calculated risk scores for each sample and
divided them into high- and low-risk groups. According to Kaplan-Meier survival analyses, high-risk patients
had significantly poorer survival outcomes (Fig. 4 F-K). In the 6 cohort (Fig. 4 F-K), the AUC values for 1-, 3-,
and 5-year survival were very good predictors of 3- and 5-year survival.
Fig. 3. Correlation between the five CAF clusters and prognosis for LUAD patients. (A) Comparison of five
CAF scores in cancerous and normal tissue; (B) K-M curves for the high and low CAF scores in the CAF_0–4
cluster; (C) CAF scores for tumors at different stages in the CAF_0–4 cluster. **P < 0.01, ****P < 0.0001.
gene. The correlation between these eight genes and 41 pathways is shown in Fig. 6A, these pathways include
allograft rejection, antigen processing and presenation, and apoptosis, etc.
Fig. 4. Identification of the hub predictive genes for constructing a risk signature. (A) Volcano plot of
differentially expressed genes of cancer and normal tissues in TCGA cohort; (B) Univariate Cox regression
analysis identified genes related to prognosis as volcano plots; (C) The trajectory of each independent
variable with lambda; (D) Plots of the produced coefficient distributions for the logarithmic (lambda) series
for parameter selection (lambda); (E) The multivariate Cox coefficients for each genes in the risk signature;
(F-K) Multivariate Cox coefficients for each gene in the risk signature; ROC and K-M curves of risk model
constructed by 8 genes in GSE3141 (F), GSE31210 (G), GSE37745 (H), GSE50081 (I), GSE68465 (J) and
TCGA cohort (K).
There was a significant negative correlation between DPYSL2, GRIA1, and METTL7A and the majority of T
cells according to the correlation analysis. Moreover, CCNB1, CDC25C, and HMGXB3 significantly positive
correlated with M0 macrophages and Dendritic cells resting (Fig. 7D). Through the correlation analysis of
immune cell abundance and gene expression, we found that BTK was significantly correlated with immune cell
infiltration (Fig. 7E).
Fig. 5. Identifies the mutation characteristics of the genes included in the risk signature. (A) A waterfall
diagram of SNV mutations in 8 main genes; (B) Analysis of co-existence and mutual exclusion of key
genes and the 10 most mutated genes in tumors; (C) Carcinogenic pathway analysis of LUAD in 10
oncogenic signaling pathways; (D) Heatmaps of 8 key genes correlated with Aneuploidy Score, Homologous
Recombination Defects, Fraction Altered, Number of Segments, and Nonsilent Mutation Rate.
Fig. 6. Identification of pathways that the risk genes involved in. (A) Gene-pathway correlation heatmap; (B)
Enrichment score heatmap for key pathways. *P < 0.05, **P < 0.01, ***P < 0.001.
Fig. 7. The relationship between risk genes and immune landscape. (A-B) Correlation matrix between 8 genes
and immune score, stromal score and estimate score. (C) A comparison of the immune score (Wilcox.test) of
the risk genes according to high and low expression of key genes. (D-E) CIBERSORT (D) and MCPcounter
(E) analysis correlation between immune cell score and key genes. *P < 0.05; **P < 0.01; ***P < 0.001; and
****P < 0.0001.
Discussion
Tumor cells and non-malignant cells such as immune cells and stromal cells are among the components of the
TME. These cells influence tumor genesis, development, metastasis, and resistance to antitumor therapies, as
well as establishing complex signals in the body20. Cancer cells often co-opt stromal cell functions to promote
Fig. 8. Response of risk score to PD-L1 blockade immunotherapy in IMvigor210 cohort. (A) Risk score
differences between immunotherapy response groups in the IMvigor210 cohort; (B) Immunotherapy
response distribution among risk score groups in the IMvigor210 cohort; (C) OS differences among risk score
groups in the IMvigor210 cohort; (D) PFS differences between risk score groups in the IMvigor210 cohort;
(E-F) Distribution of immunotherapy responses among risk score groups in the GSE78220 cohort. (G) OS
differences between risk score groups in advanced patients in the GSE78220; (H) PFS differences in risk score
groups in the GSE78220 cohort. *P < 0.05; ****P < 0.0001.
Fig. 9. Development of a nomogram for predicting LUAD prognosis. (A-B) Multivariate (A) and univariate
(B) Cox analyses of the risk score and clinicopathological characteristics; (C) Construction of a nomogram
model that incorporates the risk score and stage; (D) Calibration curves for 1, 3, and 5 years of nomogram
and decision curve for nomogram; (E) Time-ROC analysis was used to compare the predictive capacity of
clinicopathological features and the nomogram. ***P < 0.001.
tumor progression and resistance to treatment. A similar treatment is administered to stromal cells in the
microenvironment as it is to cancer cells. Some of the responses induced by these therapies may contribute
to tumor progression, due to the damage induced by these therapies21. CAF cells play an important role in
both primary and metastatic cancer progression through their interaction with other types of cells in the tumor
microenvironment22. Based on scRNA-seq data, we performed a systematic characterization and classification
of CAFs of LUAD to determine their diversity. CAF functional classification correlates with patients’ clinical
response to targeted therapies and with the tumor immune microenvironment, providing an avenue to guide
customized treatment9. Based on a score derived from DEGs across the five clusters, three clusters were
consistently associated with LUAD prognosis. In an interesting finding, we observed HIPPO and RTK-RAS
cluster differences among CAF clusters may play a role in CAF prognosis.
Based on the prognostic significance of the five CAF clusters, we developed a risk signature consisting of 8
genes centered on CAFs. It consisted of one protective gene (BTK) and three risk genes (CCNB1, CDC25C and
EXO1). In our study, there were more SNV mutations in BTK, EXO1 and GRIA1, while none were observed
in CCNB1, CLEC3B and METTL7A. Mutations in the Sense SNV influence the activity or functionality of
proteins, which can contribute to the onset of HCC or impact its progression23. While independent studies have
not established a direct connection between SNV mutations in these risk genes and the progression of LUAD,
our findings also indicate that SNV mutations in these genes may play a significant role in the advancement
of LUAD. Additionally, we discovered that the 8 genes showed significant associations with 41 pathways, with
distinct pathway signatures evident between protective genes and risk genes. For example, the protective gene
was significantly positively associated with antigen processing and presentation, apoptosis, B cell receptor
signaling pathway, chemokine signaling pathway, et al., whereas risk genes were significantly connected with
DNA replication, mismatch repair, p53 signaling pathway. The alterations of DNA replication play an important
role in NSCLC and stress to promote chromosomal instability early in cancer evolution24. Mutations in the
mismatch repair system predict the response to ICIs in NSCLC25. Therefore, this information guides us in
investigating the regulation of these risk genes in LUAD further.
According to recent research, CAFs may promote tumor progression through their interaction with TMEs26.
Through CAF-tumor interaction, Li et al. found that the HNRNPK/CLCN3 axis facilitates LUAD progression27.
Given the prognostic values of three CAF clusters, we established a CAF-based risk signature with 8 genes. It
consisted of one protective genes (BTK) and three risk genes (CCNB1, CDC25C and EXO1). There is potential
cross-talk between these genes and TME in LUAD, and these genes could be therapeutic targets for LUAD.
By interacting with these immune cells, CAFs can form an immunosuppressive TME, enabling tumor cells to
evade immune surveillance28. CAF-based risk signatures were significantly correlated with stromal scores and
immune cells, according to Yu et al. study14. In our study, SNV mutations were observed in CCNB1, CLEC3B
and METTL7A without significant co-occurrence probability. A plot of SNV and CNV genes shows that LUAD
high-risk individuals have obvious oncogene amplifications as well as tumor suppressor deletions, whereas
LUAD low-risk individuals have both tumor suppressor deletions and tumor suppressor amplifications, along
with a few oncogene amplifications29. CAF in LUAD primarily enhance the glutamine metabolism through a
CAF-specific long noncoding RNA (LINC01614), which directly interacts with ANXA2 and p65 to facilitate
the activation of NF-κB, leading to the upregulation of the glutamine transporters SLC38A2 and SLC7A5
and eventually enhances the glutamine influx of cancer cells10. In addition, we found that the 8 genes were
significantly correlated with 41 pathways, while protective genes and risk genes showed clear differences in
pathways. Therefore, these data provide us with a direction for further research on LUAD’s regulation of these
risk genes.
Evidence suggests that tumor progression can be promoted by the interaction between CAFs and TMEs12. In
our study, one predictive gene showed a significant positive correlation with immune score, while three risk genes
showed a negative correlation. These data indicate that these genes may interact with TME in LUAD, suggesting
they may be useful as therapeutic targets. A TME is comprised of several immune cells and determines the
antitumor immune status synergistically14. It plays a critical role in tumorigenesis and immunosuppression in the
TME when CAFs interact with immune cells that suppress the immune response30. In the risk signature, multiple
types of T cells were negatively associated with the predictive genes. Based on the CAF signature, Ren et al. found
that comprehensive characterization of LUAD can predict the response of LUAD to immunotherapy, and that
EXP1 facilitates invasion and tumor cell growth31. Hence, CAF may play an important role in tumorigenesis
and development, as well as regulating the microenvironment of tumors, thereby enhancing the effectiveness of
nutritional immunotherapy.
Immunotherapy has shown clinically significant benefits for non-small-cell lung cancer, but innate (primary)
and acquired resistance remain challenges32. As a result of our data, we were able to identify patients more
likely to benefit from immunotherapies based on their risk signature. Also, POSTN + CAFs, which may
cooperate with SPP1 + macrophages to promote the formation of desmoplastic architecture and suppress the
immune system, and POSTN + CAFs associated with cancer progression and poor clinical outcomes, may
provide new insights into NSCLC treatment33. A positive correlation was found between the risk genes and
M0 macrophages, indicating that the risk genes play a role in macrophage polarization. Single-cell analysis
reveals that COL11A1 + fibroblasts promote tumor progression by remodeling ECM and suppressing antitumor
immune responses34. Additionally, we found that CAF-based signatures could predict response to anti-PD-L1
immunotherapy. There data provided novel clues of the role of CAF in remodeling the cancer niche and immune
status in TME. LUAD immunotherapy relies on CAF-TME communication, but further experiments are needed
to clarify its role in LUAD.
In spite of this, there are a number of limitations in our study that should be acknowledged. In the first
step, the CAF clusters and CAF-based risk signature were created from retrospective data extracted from public
databases. Therefore, prospective LUAD cohorts and multicenter trials should be conducted in the future in order
to validate the method. Second, the CAF-based risk signature was only investigated for its potential prognostic
value, so further research is needed to determine the mechanisms involved in the development of LUAD.
Conclusion
As a result of this study, five distinct CAF clusters were identified in LUAD, each with distinct characteristics.
The DEGs among the four clusters were enriched in cell-substrate adhension, collagen-containing extracellular
matrix, extracellular matrix structural constituent and Focal adhesion signaling pathway, etc. A CAF-based
prognostic risk signature with eight genes was constructed using three of the cluster’s significant associations
with LUAD prognosis. PD-L1 blockade immunotherapy response could be predicted using CAF-based gene
signatures connected to the immune landscape. Finally, we developed a novel nomogram that integrated risk
signatures and clinicopathological features, resulting in improved prediction of the clinical outcome of LUAD
patients.
Data availability
The NSCLC scRNA-seq datasets can be downloaded from the Gene Expression Omnibus (GEO) database under
accession number GSE149655. Due to survival analysis data can be downloaded from the following database
GSE3141, GSE31210, GSE37745, GSE50081, GSE68465 and TCGA cohort. The original code in the study can be
requested from the corresponding author.
References
1. Srivastava, S. et al. Emerging role in prognosis, heterogeneity, and therapeutics. Sem. Cancer Biol. 86 (Pt 2), 233–246 (2022).
2. Desai, A. & Peters, S. Immunotherapy-based combinations in metastatic NSCLC. Cancer Treat. Rev. 116, 102545 (2023).
3. Otano, I., Ucero, A. C., Zugazagoitia, J. & Paz-Ares, L. At the crossroads of immunotherapy for oncogene-addicted subsets of
NSCLC. Nat. Reviews Clin. Oncol. 20 (3), 143–159 (2023).
4. Song, X. et al. Spatial multi-omics revealed the impact of tumor ecosystem heterogeneity on immunotherapy efficacy in patients
with advanced non-small cell lung cancer treated with bispecific antibody. J. Immunother. Cancer ; 11(2). (2023).
5. Tiwari, A., Trivedi, R. & Lin, S. Y. Tumor microenvironment: barrier or opportunity towards effective cancer therapy. J. Biomed.
Sci. 29 (1), 83 (2022).
6. de Visser, K. E. & Joyce, J. A. The evolving tumor microenvironment: from cancer initiation to metastatic outgrowth. Cancer cell.
41 (3), 374–403 (2023).
7. Luo, H. et al. Pan-cancer single-cell analysis reveals the heterogeneity and plasticity of cancer-associated fibroblasts in the tumor
microenvironment. Nat. Commun. 13 (1), 6619 (2022).
8. Biffi, G. & Tuveson, D. A. Diversity and Biology of Cancer-Associated fibroblasts. Physiol. Rev. 101 (1), 147–176 (2021).
9. Hu, H. et al. Three subtypes of lung cancer fibroblasts define distinct therapeutic paradigms. Cancer cell. 39 (11), 1531–47e10
(2021).
10. Liu, T. et al. Cancer-associated fibroblast-specific lncRNA LINC01614 enhances glutamine uptake in lung adenocarcinoma. J.
Hematol. Oncol. 15 (1), 141 (2022).
11. Yang, H. et al. Multi-scale integrative analyses identify THBS2(+) cancer-associated fibroblasts as a key orchestrator promoting
aggressiveness in early-stage lung adenocarcinoma. Theranostics. 12 (7), 3104–3130 (2022).
12. Tang, P. C. et al. Smad3 Promotes Cancer-Associated Fibroblasts Generation via Macrophage-Myofibroblast Transition. Advanced
science (Weinheim, Baden-Wurttemberg, Germany). ; 9(1):e2101235. (2022).
13. Sanchez-Vega, F. et al. Oncogenic signaling pathways in the Cancer Genome Atlas. Cell. 173 (2), 321–37e10 (2018).
14. Yu, L. et al. Characterization of cancer-related fibroblasts (CAF) in hepatocellular carcinoma and construction of CAF-based risk
signature based on single-cell RNA-seq and bulk RNA-seq data. Front. Immunol. 13, 1009789 (2022).
15. Xiang, H. et al. Single-cell analysis identifies NOTCH3-Mediated interactions between stromal cells that promote Microenvironment
Remodeling and Invasion in Lung Adenocarcinoma. Cancer Res. 84 (9), 1410–1425 (2024).
16. Li, X. et al. Single-cell RNA sequencing reveals a pro-invasive cancer-associated fibroblast subgroup associated with poor clinical
outcomes in patients with gastric cancer. Theranostics. 12 (2), 620–638 (2022).
17. Kanehisa, M., Furumichi, M., Sato, Y., Kawashima, M. & Ishiguro-Watanabe, M. KEGG for taxonomy-based analysis of pathways
and genomes. Nucleic Acids Res. 51 (D1), D587–d92 (2023).
18. Kanehisa, M. & Goto, S. KEGG: kyoto encyclopedia of genes and genomes. Nucleic Acids Res. 28 (1), 27–30 (2000).
19. Kanehisa, M. Toward understanding the origin and evolution of cellular organisms. Protein Science: Publication Protein Soc. 28
(11), 1947–1951 (2019).
20. Zhao, Y. et al. Stromal cells in the tumor microenvironment: accomplices of tumor progression? Cell Death Dis. 14 (9), 587 (2023).
21. Berg, T. J. & Pietras, A. Radiotherapy-induced remodeling of the tumor microenvironment by stromal cells. Sem. Cancer Biol. 86
(Pt 3), 846–856 (2022).
22. Chen, Y., McAndrews, K. M. & Kalluri, R. Clinical and therapeutic relevance of cancer-associated fibroblasts. Nat. Reviews Clin.
Oncol. 18 (12), 792–804 (2021).
23. Wang, H., Cao, H., Xu, Z., Wang, D. & Zeng, Y. SNP rs2596542G > A in MICA is associated with risk of hepatocellular carcinoma:
a meta-analysis. Biosci. Rep.; 39(5). (2019).
24. Venkatesan, S. et al. Induction of APOBEC3 exacerbates DNA replication stress and chromosomal instability in early breast and
Lung Cancer Evolution. Cancer Discov. 11 (10), 2456–2473 (2021).
25. Olivares-Hernández, A. et al. Influence of DNA mismatch repair (MMR) system in survival and response to Immune Checkpoint
inhibitors (ICIs) in Non-small Cell Lung Cancer (NSCLC): retrospective analysis. Biomedicines; 10(2). (2022).
26. Barrett, R. L. & Puré, E. Cancer-associated fibroblasts and their influence on tumor immunity and immunotherapy. eLife; 9. (2020).
27. Li, Y. et al. HNRNPK/CLCN3 axis facilitates the progression of LUAD through CAF-tumor interaction. Int. J. Biol. Sci. 18 (16),
6084–6101 (2022).
28. Mao, X. et al. Crosstalk between cancer-associated fibroblasts and immune cells in the tumor microenvironment: new findings and
future perspectives. Mol. Cancer. 20 (1), 131 (2021).
29. Zengin, T. & Önal-Süzek, T. Comprehensive profiling of genomic and transcriptomic differences between Risk Groups of Lung
Adenocarcinoma and lung squamous cell carcinoma. J. Personalized Med.; 11(2). (2021).
30. Wang, S. et al. Integrative analyses of bulk and single-cell RNA-seq identified cancer-associated fibroblasts-related signature as a
prognostic factor for immunotherapy in NSCLC. Cancer Immunol. Immunotherapy: CII. 72 (7), 2423–2442 (2023).
31. Ren, Q. et al. A novel signature predicts prognosis and immunotherapy in lung adenocarcinoma based on cancer-associated
fibroblasts. Front. Immunol. 14, 1201573 (2023).
32. Frisone, D., Friedlaender, A., Addeo, A. & Tsantoulis, P. The Landscape of Immunotherapy Resistance in NSCLC. Front. Oncol. 12,
817548 (2022).
33. Chen, C. et al. Single-cell and spatial transcriptomics reveal POSTN(+) cancer-associated fibroblasts correlated with immune
suppression and tumour progression in non-small cell lung cancer. Clin. Translational Med. 13 (12), e1515 (2023).
34. Zhang, J. et al. Single-cell analysis reveals the COL11A1(+) fibroblasts are cancer-specific fibroblasts that promote tumor
progression. Front. Pharmacol. 14, 1121586 (2023).
Author contributions
SYJ and ZZH conducted statistical analyses of the data and prepared the draft manuscript. MXJ and ZKK edited
the manuscript. All authors checked and proofread the final version of the manuscript.
Funding
This work was supported by the Clinical research fund of Shandong Medical Association National (grant
numbers YXH2022ZX02029), the National Natural Science Foundation of China (grant numbers 81972796,
81972863).
Declarations
Competing interests
The authors declare no competing interests.
Additional information
Supplementary Information The online version contains supplementary material available at https://doi.
org/10.1038/s41598-024-74336-1.
Correspondence and requests for materials should be addressed to K.Z.
Reprints and permissions information is available at www.nature.com/reprints.
Publisher’s note Springer Nature remains neutral with regard to jurisdictional claims in published maps and
institutional affiliations.
Open Access This article is licensed under a Creative Commons Attribution-NonCommercial-NoDerivatives
4.0 International License, which permits any non-commercial use, sharing, distribution and reproduction in
any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide
a link to the Creative Commons licence, and indicate if you modified the licensed material. You do not have
permission under this licence to share adapted material derived from this article or parts of it. The images or
other third party material in this article are included in the article’s Creative Commons licence, unless indicated
otherwise in a credit line to the material. If material is not included in the article’s Creative Commons licence and
your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain
permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/
licenses/by-nc-nd/4.0/.