1
|
Zhou Y, Lu Q. Identification and analysis of amino acid metabolism-related subtypes in lung adenocarcinoma. Am J Physiol Regul Integr Comp Physiol 2025; 328:R470-R480. [PMID: 39745726 DOI: 10.1152/ajpregu.00217.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2024] [Revised: 12/16/2024] [Accepted: 12/23/2024] [Indexed: 03/21/2025]
Abstract
We aimed to explore the role of amino acid metabolism (AAM) and identify biomarkers for prognosis management and treatment of lung adenocarcinoma. Differentially expressed genes (DEGs) associated with AAM in lung adenocarcinoma were selected from public databases. Samples were clustered into varying subtypes using ConsensusClusterPlus based on gene levels. Survival analysis was conducted using a survival package, and immune analysis was performed using ssGSEA and ESTIMATE. Enrichment analysis was performed using gene set enrichment analysis, and a protein-protein interaction network of DEGs between subgroups was established through STRING. Hub genes were screened and verified using survival analysis, and drug sensitivity prediction was performed. One hundred sixty-three DEGs associated with AAM in lung adenocarcinoma were obtained, and two AAM-associated subtypes were identified. Cluster 1 showed higher survival rates and immune levels compared with cluster 2. The two subtypes were mainly enriched in immune-related signaling pathways, such as B cell receptor, Jak-Stat, and natural killer cell-mediated cytotoxicity. In addition, the mutation landscape between the two groups was significantly different. F2, AHSG, and APOA1 were key hub genes that significantly affected the prognosis differences between the two subtypes. Cluster 2 showed higher sensitivity to drugs, such as mithramycin, depsipeptide, and actinomycin than cluster 1. This study identified two AAM-associated gene subtypes and their biomarkers and predicted the immune status and drug treatment sensitivity of varying subtypes. The results are instructive in the clinical treatment of lung adenocarcinoma.NEW & NOTEWORTHY Two amino acid metabolism-related subtypes were identified based on differentially expressed genes associated with amino acid metabolism. Cluster 1 showed higher survival rates and immune levels compared with cluster 2. Cluster 2 showed higher sensitivity to drugs, such as mithramycin, depsipeptide, and actinomycin compared with cluster 1.
Collapse
Affiliation(s)
- Yifan Zhou
- Department of Thoracic Surgery, Guangxi Academy of Medical Sciences and the People's Hospital of Guangxi Zhuang Autonomous Region, Nanning, People's Republic of China
| | - Qiangchang Lu
- Department of Thoracic Surgery, Guangxi Academy of Medical Sciences and the People's Hospital of Guangxi Zhuang Autonomous Region, Nanning, People's Republic of China
| |
Collapse
|
2
|
Zhang J, Hu D, Fang P, Qi M, Sun G. Deciphering key roles of B cells in prognostication and tailored therapeutic strategies for lung adenocarcinoma: a multi-omics and machine learning approach towards predictive, preventive, and personalized treatment strategies. EPMA J 2025; 16:127-163. [PMID: 39991096 PMCID: PMC11842682 DOI: 10.1007/s13167-024-00390-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2024] [Accepted: 11/24/2024] [Indexed: 02/25/2025]
Abstract
Background Lung adenocarcinoma (LUAD) remains a significant global health challenge, with an urgent need for innovative predictive, preventive, and personalized medicine (PPPM) strategies to improve patient outcomes. This study leveraged multi-omics and machine learning approaches to uncover the prognostic roles of B cells in LUAD, thereby reinforcing the PPPM approach. Methods We integrated multi-omics data, including bulk RNA, ATAC-seq, single-cell RNA, and spatial transcriptomics sequencing, to characterize the B cell landscape in LUAD within the PPPM framework. Subsequently, we developed an integrative machine learning program that generated the Scissor+ related B cell score (SRBS). This score was validated in the training and validation sets, and its prognostic value was assessed along with clinical features to develop predictive nomograms. This study further assessed the role of SRBS and SRBS genes in response to immunotherapy and identified personalized drug targets for distinct risk subgroups, with gene expression verified experimentally to ensure tailored medical interventions. Results Our analysis identified 79 Scissor+ B cell genes linked to LUAD prognosis, supporting the predictive aspect of PPPM. The SRBS model, which utilizes multiple machine learning algorithms, performed excellently in predicting prognosis and clinical transformation, embodying the preventive and personalized aspects of PPPM. Multifactorial analysis confirmed that SRBS was an independent prognostic factor. We observed varying biological functions and immune cell infiltration in the tumor immune microenvironment (TIME) between the high- and low-SRBS groups, underscoring personalized treatment approaches. Notably, patients with elevated SRBS may exhibit resistance to immunotherapy but show increased sensitivity to chemotherapy and targeted therapies. Additionally, we found that LDHA, as an SRBS gene with significant clinical implications, may regulate the sensitivity of LUAD cells to cisplatin. Conclusion This study presents a B cell-associated gene signature that serves as a prognostic marker to facilitate personalized treatment for patients with LUAD, adhering to the principles of PPPM. Supplementary Information The online version contains supplementary material available at 10.1007/s13167-024-00390-4.
Collapse
Affiliation(s)
- Jinjin Zhang
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Anhui Medical University, Hefei, 230022 Anhui Province China
| | - Dingtao Hu
- Clinical Cancer Institute, Center for Translational Medicine, Naval Medical University, Shanghai, China
| | - Pu Fang
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Anhui Medical University, Hefei, 230022 Anhui Province China
| | - Min Qi
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Anhui Medical University, Hefei, 230022 Anhui Province China
| | - Gengyun Sun
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Anhui Medical University, Hefei, 230022 Anhui Province China
| |
Collapse
|
3
|
Xiang H, Kasajima R, Azuma K, Tagami T, Hagiwara A, Nakahara Y, Saito H, Igarashi Y, Wei F, Ban T, Yoshihara M, Nakamura Y, Sato S, Koizume S, Tamura T, Sasada T, Miyagi Y. Multi-omics analysis-based clinical and functional significance of a novel prognostic and immunotherapeutic gene signature derived from amino acid metabolism pathways in lung adenocarcinoma. Front Immunol 2024; 15:1361992. [PMID: 39735553 PMCID: PMC11671776 DOI: 10.3389/fimmu.2024.1361992] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2023] [Accepted: 07/30/2024] [Indexed: 12/31/2024] Open
Abstract
Background Studies have shown that tumor cell amino acid metabolism is closely associated with lung adenocarcinoma (LUAD) development and progression. However, the comprehensive multi-omics features and clinical impact of the expression of genes associated with amino acid metabolism in the LUAD tumor microenvironment (TME) are yet to be fully understood. Methods LUAD patients from The Cancer Genome Atlas (TCGA) database were enrolled in the training cohort. Using least absolute shrinkage and selection operator Cox regression analysis, we developed PTAAMG-Sig, a signature based on the expression of tumor-specific amino acid metabolism genes associated with overall survival (OS) prognosis. We evaluated its predictive performance for OS and thoroughly explored the effects of the PTAAMG-Sig risk score on the TME. The risk score was validated in two Gene Expression Omnibus (GEO) cohorts and further investigated against an original cohort of chemotherapy combined with immune checkpoint inhibitors (ICIs). Somatic mutation, chemotherapy response, immunotherapy response, gene set variation, gene set enrichment, immune infiltration, and plasma-free amino acids (PFAAs) profile analyses were performed to identify the underlying multi-omics features. Results TCGA datasets based PTAAMG-Sig model consisting of nine genes, KYNU, PSPH, PPAT, MIF, GCLC, ACAD8, TYRP1, ALDH2, and HDC, could effectively stratify the OS in LUAD patients. The two other GEO-independent datasets validated the robust predictive power of PTAAMG-Sig. Our differential analysis of somatic mutations in the high- and low-risk groups in TCGA cohort showed that the TP53 mutation rate was significantly higher in the high-risk group and negatively correlated with OS. Prediction from transcriptome data raised the possibility that PTAAMG-Sig could predict the response to chemotherapy and ICIs therapy. Our immunotherapy cohort confirmed the predictive ability of PTAAMG-Sig in the clinical response to ICIs therapy, which correlated with the infiltration of immune cells (e.g., T lymphocytes and nature killer cells). Corresponding to the concentrations of PFAAs, we discovered that the high PTAAMG-Sig risk score patients showed a significantly lower concentration of plasma-free α-aminobutyric acid. Conclusion In patients with LUAD, the PTAAMG-Sig effectively predicted OS, drug sensitivity, and immunotherapy outcomes. These findings are expected to provide new targets and strategies for personalized treatment of LUAD patients.
Collapse
Affiliation(s)
- Huihui Xiang
- Molecular Pathology & Genetics Division, Kanagawa Cancer Center Research Institute, Yokohama, Japan
- Department of Pathology, Kanagawa Cancer Center, Yokohama, Japan
| | - Rika Kasajima
- Molecular Pathology & Genetics Division, Kanagawa Cancer Center Research Institute, Yokohama, Japan
- Center for Cancer Genome Medicine, Kanagawa Cancer Center, Yokohama, Japan
| | - Koichi Azuma
- Department of Internal Medicine, Kurume University School of Medicine, Kurume, Japan
| | - Tomoyuki Tagami
- Research Institute for Bioscience Products and Fine Chemicals, Ajinomoto Co., Inc., Kanagawa, Japan
| | - Asami Hagiwara
- Research Institute for Bioscience Products and Fine Chemicals, Ajinomoto Co., Inc., Kanagawa, Japan
| | - Yoshiro Nakahara
- Department of Thoracic Oncology, Kanagawa Cancer Center, Yokohama, Kanagawa, Japan
- Department of Respiratory Medicine, Kitasato University School of Medicine, Sagamihara, Kanagawa, Japan
| | - Haruhiro Saito
- Department of Thoracic Oncology, Kanagawa Cancer Center, Yokohama, Kanagawa, Japan
| | - Yuka Igarashi
- Division of Cancer Immunotherapy, Kanagawa Cancer Center Research Institute, Yokohama, Japan
- Cancer Vaccine and Immunotherapy Center, Kanagawa Cancer Center, Yokohama, Japan
| | - Feifei Wei
- Division of Cancer Immunotherapy, Kanagawa Cancer Center Research Institute, Yokohama, Japan
- Cancer Vaccine and Immunotherapy Center, Kanagawa Cancer Center, Yokohama, Japan
| | - Tatsuma Ban
- Department of Immunology, Yokohama City University Graduate School of Medicine, Yokohama, Japan
| | - Mitsuyo Yoshihara
- Molecular Pathology & Genetics Division, Kanagawa Cancer Center Research Institute, Yokohama, Japan
- Morphological Analysis Laboratory, Kanagawa Cancer Center Research Institute, Yokohama, Japan
| | - Yoshiyasu Nakamura
- Molecular Pathology & Genetics Division, Kanagawa Cancer Center Research Institute, Yokohama, Japan
- Morphological Analysis Laboratory, Kanagawa Cancer Center Research Institute, Yokohama, Japan
| | - Shinya Sato
- Molecular Pathology & Genetics Division, Kanagawa Cancer Center Research Institute, Yokohama, Japan
- Department of Pathology, Kanagawa Cancer Center, Yokohama, Japan
- Morphological Analysis Laboratory, Kanagawa Cancer Center Research Institute, Yokohama, Japan
| | - Shiro Koizume
- Molecular Pathology & Genetics Division, Kanagawa Cancer Center Research Institute, Yokohama, Japan
- Department of Pathology, Kanagawa Cancer Center, Yokohama, Japan
| | - Tomohiko Tamura
- Department of Immunology, Yokohama City University Graduate School of Medicine, Yokohama, Japan
| | - Tetsuro Sasada
- Division of Cancer Immunotherapy, Kanagawa Cancer Center Research Institute, Yokohama, Japan
- Cancer Vaccine and Immunotherapy Center, Kanagawa Cancer Center, Yokohama, Japan
| | - Yohei Miyagi
- Molecular Pathology & Genetics Division, Kanagawa Cancer Center Research Institute, Yokohama, Japan
- Department of Pathology, Kanagawa Cancer Center, Yokohama, Japan
| |
Collapse
|
4
|
Li C, Mao Y, Hu J, Su C, Li M, Tan H. Integrating machine learning and multi-omics analysis to develop an asparagine metabolism immunity index for improving clinical outcome and drug sensitivity in lung adenocarcinoma. Immunol Res 2024; 72:1447-1469. [PMID: 39320693 DOI: 10.1007/s12026-024-09544-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2024] [Accepted: 09/13/2024] [Indexed: 09/26/2024]
Abstract
Lung adenocarcinoma (LUAD) is a malignancy affecting the respiratory system. Most patients are diagnosed with advanced or metastatic lung cancer due to the fact that most of their clinical symptoms are insidious, resulting in a bleak prognosis. Given that abnormal reprogramming of asparagine metabolism (AM) has emerged as an emerging therapeutic target for anti-tumor therapy. However, the clinical significance of abnormal reprogramming of AM in LUAD patients is unclear. In this study, we collected 864 asparagine metabolism-related genes (AMGs) and used a machine-learning computational framework to develop an asparagine metabolism immunity index (AMII) for LUAD patients. Through the utilization of median AMII scores, LUAD patients were segregated into either a low-AMII group or a high-AMII group. We observed outstanding performance of AMII in predicting survival prognosis in LUAD patients in the TCGA-LUAD cohort and in three externally independently validated GEO cohorts (GSE72094, GSE37745, and GSE30219), and poorer prognosis for LUAD patients in the high-AMII group. The results of univariate and multivariate analyses showed that AMII can be used as an independent risk factor for LUAD patients. In addition, the results of C-index analysis and decision analysis showed that AMII-based nomograms had a robust performance in terms of accuracy of prognostic prediction and net clinical benefit in patients with LUAD. Excitingly, LUAD patients in the low-AMII group were more sensitive to commonly used chemotherapeutic drugs. Consequently, AMII is expected to be a novel diagnostic tool for clinical classification, providing valuable insights for clinical decision-making and personalized management of LUAD patients.
Collapse
Affiliation(s)
- Chunhong Li
- Central Laboratory, The Second Affiliated Hospital of Guilin Medical University, Guilin , 541199, Guangxi, China.
- Guangxi Health Commission Key Laboratory of Glucose and Lipid Metabolism Disorders, The Second Affiliated Hospital of Guilin Medical University, Guilin, 541199, Guangxi, China.
- Central Laboratory, Guangxi Health Commission Key Laboratory of Glucose and Lipid Metabolism Disorders, The Second Affiliated Hospital of Guilin Medical University, Guilin, 541199, Guangxi, China.
| | - Yuhua Mao
- Department of Obstetrics, The Second Affiliated Hospital of Guilin Medical University, Guilin, 541199, Guangxi, China
| | - Jiahua Hu
- Central Laboratory, The Second Affiliated Hospital of Guilin Medical University, Guilin , 541199, Guangxi, China
- Guangxi Health Commission Key Laboratory of Glucose and Lipid Metabolism Disorders, The Second Affiliated Hospital of Guilin Medical University, Guilin, 541199, Guangxi, China
| | - Chunchun Su
- Department of Laboratory Medicine, The Second Affiliated Hospital of Guilin Medical University, Guilin, 541199, Guangxi, China
| | - Mengqin Li
- College of Pharmacy, Guilin Medical University, Guilin, 541199, Guangxi, China
| | - Haiyin Tan
- School of Medical Laboratory Medicine, Guilin Medical University, Guilin, 541004, Guangxi, China
| |
Collapse
|
5
|
Wang Z, Zuo C, Fei J, Chen H, Wang L, Xie Y, Zhang J, Min S, Wang X, Lian C. Development of a novel centrosome-related risk signature to predict prognosis and treatment response in lung adenocarcinoma. Discov Oncol 2024; 15:717. [PMID: 39592523 PMCID: PMC11599701 DOI: 10.1007/s12672-024-01615-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/11/2024] [Accepted: 11/21/2024] [Indexed: 11/28/2024] Open
Abstract
BACKGROUND Abnormalities of centrosomes, the major microtubular organizing centers of animal cells and regulators of cell cycle progression, usually accelerate tumor progression, but their prognostic value in lung adenocarcinoma (LUAD) remains insufficiently explored. METHODS We collected centrosome genes from the literature and identified LUAD-specific centrosome-related genes (CRGs) using the single-sample gene set enrichment analysis (ssGSEA) algorithm and weighted gene co-expression network analysis (WGCNA). Univariate Cox was performed to screen prognostic CRGs. Consistent clustering was performed to classify LUAD patients into two subgroups, and centrosome-related risk score signatures were constructed by Lasso and multivariate Cox regression to predict overall survival (OS). We further explored the correlation between CRS and patient prognosis, clinical manifestations, mutation status, tumor microenvironment, and response to different treatments. RESULTS We constructed centrosome-associated prognostic features and verified that CRS could effectively predict 1-, 3-, and 5-year survival in LUAD patients. In addition, patients in the high-risk group exhibited elevated tumor mutational loads and reduced levels of immune infiltration, particularly of T and B cells. Patients in the high-risk group were resistant to immunotherapy and sensitive to 5-fluoropyrimidine and gefitinib. The key gene spermine synthase (SRM) is highly expressed at the mRNA and protein levels in LUAD. DISCUSSION Our work develops a novel centrosome-related prognostic signature that accurately predicts OS in LUAD and can assist in clinical diagnosis and treatment.
Collapse
Affiliation(s)
- Ziqiang Wang
- Anhui Province Key Laboratory of Respiratory Tumor and Infectious Disease, Department of Pulmonary and Critical Care Medicine, Molecular Diagnosis Center, First Affiliated Hospital of Bengbu Medical University, Bengbu, 233030, China
- Research Center of Clinical Laboratory Science, Bengbu Medical University, Bengbu, 233030, China
| | - Chao Zuo
- Department of Clinical Laboratory, Affiliated Hospital of Guilin Medical University, Guilin, 541001, China
| | - Jiaojiao Fei
- Anhui Province Key Laboratory of Respiratory Tumor and Infectious Disease, Department of Pulmonary and Critical Care Medicine, Molecular Diagnosis Center, First Affiliated Hospital of Bengbu Medical University, Bengbu, 233030, China
| | - Huili Chen
- Research Center of Clinical Laboratory Science, Bengbu Medical University, Bengbu, 233030, China
| | - Luyao Wang
- Department of Genetics, School of Life Sciences, Bengbu Medical University, Bengbu, 233030, China
| | - Yiluo Xie
- Department of Clinical Medicine, Bengbu Medical University, Bengbu, 233030, China
| | - Jing Zhang
- Department of Genetics, School of Life Sciences, Bengbu Medical University, Bengbu, 233030, China
| | - Shengping Min
- Anhui Province Key Laboratory of Respiratory Tumor and Infectious Disease, Department of Pulmonary and Critical Care Medicine, Molecular Diagnosis Center, First Affiliated Hospital of Bengbu Medical University, Bengbu, 233030, China
| | - Xiaojing Wang
- Anhui Province Key Laboratory of Respiratory Tumor and Infectious Disease, Department of Pulmonary and Critical Care Medicine, Molecular Diagnosis Center, First Affiliated Hospital of Bengbu Medical University, Bengbu, 233030, China.
- Joint Research Center for Regional Diseases of Institute of Health and Medicine (IHM), Hefei Comprehensive National Science Center, Bengbu Medical University, Bengbu, 233030, China.
| | - Chaoqun Lian
- Research Center of Clinical Laboratory Science, Bengbu Medical University, Bengbu, 233030, China.
| |
Collapse
|
6
|
Wu D, Liu Y, Liu J, Ma L, Tong X. Myeloid cell differentiation-related gene signature for predicting clinical outcome, immune microenvironment, and treatment response in lung adenocarcinoma. Sci Rep 2024; 14:17460. [PMID: 39075165 PMCID: PMC11286868 DOI: 10.1038/s41598-024-68111-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2024] [Accepted: 07/19/2024] [Indexed: 07/31/2024] Open
Abstract
Considering the key role of myeloid cell differentiation-related genes in the tumor microenvironment (TME), we aimed to build a prognostic risk model using these genes for Lung adenocarcinoma (LUAD). The mRNA gene expression profiles of LUAD patients from The Cancer Genome Atlas (TCGA) and Gene Expression Omnibus (GEO) databases were downloaded as the training and validation sets. Then, "edgeR" R package was applied to screen out the differentially expressed genes (DEGs) and univariate cox regression, backward stepwise selection analyses were performed to construct a prognostic model for LUAD. ESTIMATE, TIMER, XCELL, CIBERSORT abs, QUANTISEQ, MCPCOUNTER, EPIC, and CIBERSORT algorithms were conducted to access the association of risk levels with the stromal and immune cell infiltration levels in LUAD. Six genes (F2RL1, PRKDC, TNFSF11, INHA, PLA2G3 and TUBB1) were utilized to construct the prognostic model. The risk model showed excellent prognostic performance for LUAD in both TCGA and GEO datasets. Also, compared to the low-risk patients, the high-risk patients had higher expression of immune checkpoint molecules and showed a lower IC50 value to the chemotherapy agents. Our findings provided a myeloid cell differentiation-related gene signature that could effectively predict prognosis and guide treatment strategies for LUAD patients.
Collapse
Affiliation(s)
- Di Wu
- Experimental Research Center, Qingpu Branch of Zhongshan Hospital Affiliated to Fudan University, Shanghai, 201700, China
| | - Yibing Liu
- Experimental Research Center, Qingpu Branch of Zhongshan Hospital Affiliated to Fudan University, Shanghai, 201700, China
| | - Jian Liu
- Department of Otolaryngology-Head and Neck Surgery, Qingpu Branch of Zhongshan Hospital Affiliated to Fudan University, Shanghai, China
| | - Li Ma
- Experimental Research Center, Qingpu Branch of Zhongshan Hospital Affiliated to Fudan University, Shanghai, 201700, China
| | - Xiaoxia Tong
- Experimental Research Center, Qingpu Branch of Zhongshan Hospital Affiliated to Fudan University, Shanghai, 201700, China.
| |
Collapse
|
7
|
Zhang L, Zhang X, Guan M, Zeng J, Yu F, Lai F. Identification of a novel ADCC-related gene signature for predicting the prognosis and therapy response in lung adenocarcinoma. Inflamm Res 2024; 73:841-866. [PMID: 38507067 DOI: 10.1007/s00011-024-01871-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2023] [Revised: 03/03/2024] [Accepted: 03/05/2024] [Indexed: 03/22/2024] Open
Abstract
BACKGROUND Previous studies have largely neglected the role of ADCC in LUAD, and no study has systematically compiled ADCC-associated genes to create prognostic signatures. METHODS In this study, 1564 LUAD patients, 2057 NSCLC patients, and more than 5000 patients with various cancer types from diverse cohorts were included. R package ConsensusClusterPlus was utilized to classify patients into different subtypes. A number of machine-learning algorithms were used to construct the ADCCRS. GSVA and ClusterProfiler were used for enrichment analyses, and IOBR was used to quantify immune cell infiltration level. GISTIC2.0 and maftools were used to analyze the CNV and SNV data. The Oncopredict package was used to predict drug information based on the GDSC1. Three immunotherapy cohorts were used to evaluate patient response to immunotherapy. The Seurat package was used to process single-cell data, the AUCell package was used to calculate cells' geneset activity scores, and the Scissor algorithm was used to identify ADCCRS-associated cells. RESULTS Through unsupervised clustering, two distinct subtypes of LUAD were identified, each exhibiting distinct clinical characteristics. The ADCCRS, consisted of 16 genes, was constructed by integrated machine-learning methods. The prognostic power of ADCCRS was validated in 28 independent datasets. Further, ADCCRS shows better predictive abilities than 102 previously published signatures in predicting LUAD patients' survival. A nomogram incorporating ADCCRS and clinical features was constructed, demonstrating high predictive performance. ADCCRS positively correlates with patients' gene mutation, and integrated analysis of bulk and single-cell transcriptome data revealed the association of ADCCRS with TME modulators. Cells representing high-ADCCRS phenotype exhibited more malignant features. LUAD patients with high ADCCRS levels exhibited sensitivity to chemotherapy and targeted therapy, while displaying resistance to immunotherapy. In pan-cancer analysis, ADCCRS still exhibited significant prognostic value and was found to be a risk factor for most cancer patients. CONCLUSIONS ADCCRS offers a critical prognostic insight for patients with LUAD, shedding light on the tumor microenvironment and forecasting treatment responsiveness.
Collapse
Affiliation(s)
- Liangyu Zhang
- Department of Thoracic Surgery, The First Affiliated Hospital, Fujian Medical University, Fuzhou, 350005, China
- Department of Thoracic Surgery, National Regional Medical Center, Binhai Campus of the Fitst Affiliated Hospiral, Fujian Medical University, Fuzhou, 350212, China
| | - Xun Zhang
- Department of Thoracic Surgery, The First Affiliated Hospital, Fujian Medical University, Fuzhou, 350005, China
- Department of Thoracic Surgery, National Regional Medical Center, Binhai Campus of the Fitst Affiliated Hospiral, Fujian Medical University, Fuzhou, 350212, China
| | - Maohao Guan
- Department of Thoracic Surgery, The First Affiliated Hospital, Fujian Medical University, Fuzhou, 350005, China
- Department of Thoracic Surgery, National Regional Medical Center, Binhai Campus of the Fitst Affiliated Hospiral, Fujian Medical University, Fuzhou, 350212, China
| | - Jianshen Zeng
- Department of Thoracic Surgery, The First Affiliated Hospital, Fujian Medical University, Fuzhou, 350005, China
- Department of Thoracic Surgery, National Regional Medical Center, Binhai Campus of the Fitst Affiliated Hospiral, Fujian Medical University, Fuzhou, 350212, China
| | - Fengqiang Yu
- Department of Thoracic Surgery, The First Affiliated Hospital, Fujian Medical University, Fuzhou, 350005, China.
- Department of Thoracic Surgery, National Regional Medical Center, Binhai Campus of the Fitst Affiliated Hospiral, Fujian Medical University, Fuzhou, 350212, China.
| | - Fancai Lai
- Department of Thoracic Surgery, The First Affiliated Hospital, Fujian Medical University, Fuzhou, 350005, China.
- Department of Thoracic Surgery, National Regional Medical Center, Binhai Campus of the Fitst Affiliated Hospiral, Fujian Medical University, Fuzhou, 350212, China.
| |
Collapse
|