1
|
Sun C, Cheng X, Xu J, Chen H, Tao J, Dong Y, Wei S, Chen R, Meng X, Ma Y, Tian H, Guo X, Bi S, Zhang C, Kang J, Zhang M, Lv H, Shang Z, Lv W, Zhang R, Jiang Y. A review of disease risk prediction methods and applications in the omics era. Proteomics 2024; 24:e2300359. [PMID: 38522029 DOI: 10.1002/pmic.202300359] [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: 09/15/2023] [Revised: 03/08/2024] [Accepted: 03/12/2024] [Indexed: 03/25/2024]
Abstract
Risk prediction and disease prevention are the innovative care challenges of the 21st century. Apart from freeing the individual from the pain of disease, it will lead to low medical costs for society. Until very recently, risk assessments have ushered in a new era with the emergence of omics technologies, including genomics, transcriptomics, epigenomics, proteomics, and so on, which potentially advance the ability of biomarkers to aid prediction models. While risk prediction has achieved great success, there are still some challenges and limitations. We reviewed the general process of omics-based disease risk model construction and the applications in four typical diseases. Meanwhile, we highlighted the problems in current studies and explored the potential opportunities and challenges for future clinical practice.
Collapse
Affiliation(s)
- Chen Sun
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, China
- The EWAS Project, Harbin, China
| | - Xiangshu Cheng
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, China
- The EWAS Project, Harbin, China
| | - Jing Xu
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, China
- The EWAS Project, Harbin, China
| | - Haiyan Chen
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, China
| | - Junxian Tao
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, China
- The EWAS Project, Harbin, China
| | - Yu Dong
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, China
- The EWAS Project, Harbin, China
| | - Siyu Wei
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, China
- The EWAS Project, Harbin, China
| | - Rui Chen
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, China
| | - Xin Meng
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, China
| | - Yingnan Ma
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, China
- The EWAS Project, Harbin, China
| | - Hongsheng Tian
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, China
| | - Xuying Guo
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, China
| | - Shuo Bi
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, China
| | - Chen Zhang
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, China
| | - Jingxuan Kang
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, China
| | - Mingming Zhang
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, China
| | - Hongchao Lv
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, China
| | - Zhenwei Shang
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, China
| | - Wenhua Lv
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, China
| | - Ruijie Zhang
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, China
| | - Yongshuai Jiang
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, China
- The EWAS Project, Harbin, China
| |
Collapse
|
2
|
Wang Y, Yao J, Zhang Z, Wei L, Wang S. Generation of novel lipid metabolism-based signatures to predict prognosis and immunotherapy response for colorectal adenocarcinoma. Sci Rep 2024; 14:17158. [PMID: 39060344 PMCID: PMC11282063 DOI: 10.1038/s41598-024-67549-x] [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: 01/12/2024] [Accepted: 07/12/2024] [Indexed: 07/28/2024] Open
Abstract
Lipid metabolism reprogramming involves in epithelial-mesenchymal transition (EMT), cancer stemness and immune checkpoints (ICs), which influence the metastasis of cancer. This study aimed to generate lipid metabolism-based signatures to predict prognosis, immunotherapy and chemotherapy response for colorectal adenocarcinoma (COAD). Transcriptome data and clinical information of COAD patients were collected from the cancer genome atlas (TCGA) database. The expression of EMT-, stem cell-, and IC-related genes were assessed between COAD and control samples. Modules and genes correlated EMT, ICs and stemness signatures were identified through weighted gene co-expression network analysis (WGCNA). Prognostic signatures were generated and then the distribution of risk genes was evaluated using single-cell RNA sequencing (scRNA-seq) data from GSE132465 dataset. COAD patients exhibited increased EMT score and stemness along with decreased ICs. Next, 12 hub genes (PIK3CG, ALOX5AP, PIK3R5, TNFAIP8L2, DPEP2, PIK3CD, PIK3R6, GGT5, ELOVL4, PTGIS, CYP7B1 and PRKD1) were found within green and yellow modules correlated with EMT, stemness and ICs. Lipid metabolism-based prognostic signatures were generated based on PIK3CG, GGT5 and PTGIS. Patients with high-risk group had poor prognosis, elevated ESTIMATEScore and StromalScore, 100% mutation rate and higher TIDE score. Samples in low-risk group had more immunogenicity on ICIs. Notably, PIK3CG was expressed in B cells, while GGT5 and PTGIS were expressed in stromal cells. This study generates lipid metabolism-based signatures correlated with EMT, stemness and ICs for predicting prognosis of COAD, and provides potential therapeutic targets for immunotherapy in COAD.
Collapse
Affiliation(s)
- Yi Wang
- Department of Oncology and Hematology, Suzhou Kowloon Hospital, Shanghai Jiao Tong University School of Medicine, Suzhou, 215127, China
| | - Jun Yao
- Department of General Surgery, The Fourth Affiliated Hospital of Soochow University, Suzhou, 215127, China
| | - Zhe Zhang
- Department of General Surgery, The Fourth Affiliated Hospital of Soochow University, Suzhou, 215127, China
| | - Luxin Wei
- Department of General Surgery, The Fourth Affiliated Hospital of Soochow University, Suzhou, 215127, China
| | - Sheng Wang
- Department of General Surgery, The Fourth Affiliated Hospital of Soochow University, Suzhou, 215127, China.
| |
Collapse
|
3
|
Shpilman Z, Kidane D. Dysregulation of base excision repair factors associated with low tumor immunogenicity in head and neck cancer: implication for immunotherapy. Ther Adv Med Oncol 2024; 16:17588359241248330. [PMID: 38680291 PMCID: PMC11047243 DOI: 10.1177/17588359241248330] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2023] [Accepted: 04/03/2024] [Indexed: 05/01/2024] Open
Abstract
Background Head and neck squamous carcinoma (HNSCC) is caused by different exogenous risk factors including smoking cigarettes, alcohol consumption, and HPV infection. Base excision repair (BER) is the frontline to repair oxidative DNA damage, which is initiated by the DNA N-glycosylase proteins (OGG1) and other BER factors including DNA polymerase β (POLB). Objective Explore whether BER genes' (OGG1, POLB) overexpression in HNSCC alters genomic integrity, immunogenicity, and its role in prognostic value. Design RNA sequencing (RNA-Seq) and clinical information (age, gender, histological grade, survival status, and stage) of 530 patients of HNSCC were retrieved from the Cancer Genome Atlas. Patients' data are categorized HPV positive or negative to analyze the tumor data including the tumor stage, POLB, and OGG1 gene expression. Methods RNA-Seq of HNSCC data retrieved and mutation count and aneuploidy score were compared using an unpaired t-test. The TIMER algorithm was used to calculate the tumor abundance of six infiltrating immune cells (CD4+ T cells, CD8+ T cells, B cells, neutrophils, macrophages, and dendritic cells) based on RNA-Seq expression profile data. The correlation between the POLB, OGG1, and immune cells was calculated by Spearman correlation analysis using TIMER 2.0. Results Our data analysis reveals that BER genes frequently overexpressed in HNSCC tumors and increase mutation count. In addition, OGG1 and POLB overexpression are associated with low infiltration of immune cells, low immune checkpoint gene expression (PD-1, cytotoxic T-lymphocyte antigen 4, program death ligand 1, and program death ligand 2), and innate immune signaling genes. Furthermore, dysregulated BER factors in Human papillomavirus (HPV) positive tumors had better overall survival. Conclusion Our analysis suggests that dysregulation of the BER genes panel might be a potential prognosis marker and/or an attractive target for an immune checkpoint blockade in HNSCC cancers. However, our observation still requires further experimental-based scientific validation studies.
Collapse
Affiliation(s)
- Zackary Shpilman
- Department of Chemistry and Biochemistry, University of Maryland, College Park, MD, USA
- Department of Physiology and Biophysics, College of Medicine, Howard University, Washington, DC, USA
| | - Dawit Kidane
- Department of Physiology and Biophysics, College of Medicine, Howard University, 520 W Street, Northwestern Washington, DC 20059, USA
| |
Collapse
|
4
|
Wang L, He Y, Bai Y, Zhang S, Pang B, Chen A, Wu X. Construction and validation of a folate metabolism-related gene signature for predicting prognosis in HNSCC. J Cancer Res Clin Oncol 2024; 150:198. [PMID: 38625586 PMCID: PMC11021263 DOI: 10.1007/s00432-024-05731-4] [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: 01/30/2024] [Accepted: 03/28/2024] [Indexed: 04/17/2024]
Abstract
PURPOSE Metabolic reprogramming is currently considered a hallmark of tumor and immune development. It is obviously of interest to identify metabolic enzymes that are associated with clinical prognosis in head and neck squamous cell carcinomas (HNSCC). METHODS Candidate genes were screened to construct folate metabolism scores by Cox regression analysis. Functional enrichment between high- and low-folate metabolism groups was explored by GO, KEGG, GSVA, and ssGSEA. EPIC, MCPcounter, and xCell were utilized to explore immune cell infiltration between high- and low-folate metabolism groups. Relevant metabolic scores were calculated and visually analyzed by the "IOBR" software package. RESULTS To investigate the mechanism behind metabolic reprogramming of HNSCC, 2886 human genes associated with 86 metabolic pathways were selected. Folate metabolism is significantly enriched in HNSCC, and that the six-gene (MTHFD1L, MTHFD2, SHMT2, ATIC, MTFMT, and MTHFS) folate score accurately predicts and differentiates folate metabolism levels. Reprogramming of folate metabolism affects CD8T cell infiltration and induces immune escape through the MIF signaling pathway. Further research found that SHMT2, an enzyme involved in folate metabolism, inhibits CD8T cell infiltration and induces immune escape by regulating the MIF/CD44 signaling axis, which in turn promotes HNSCC progression. CONCLUSIONS Our study identified a novel and robust folate metabolic signature. A folate metabolic signature comprising six genes was effective in assessing the prognosis and reflecting the immune status of HNSCC patients. The target molecule of folate metabolic reprogramming, SHMT2, probably plays a very important role in HNSCC development and immune escape.
Collapse
Affiliation(s)
- Lu Wang
- Department of Otolaryngology Head and Neck Surgery, Xiangya Hospital of Central South University, 87 Xiangya Road, Changsha, 410008, Hunan, China
| | - Ye He
- Department of Otolaryngology Head and Neck Surgery, Xiangya Hospital of Central South University, 87 Xiangya Road, Changsha, 410008, Hunan, China
| | - Yijiang Bai
- Department of Otolaryngology Head and Neck Surgery, Xiangya Hospital of Central South University, 87 Xiangya Road, Changsha, 410008, Hunan, China
| | - Shuai Zhang
- Department of Otolaryngology Head and Neck Surgery, Xiangya Hospital of Central South University, 87 Xiangya Road, Changsha, 410008, Hunan, China
| | - Bo Pang
- Department of Otolaryngology Head and Neck Surgery, Xiangya Hospital of Central South University, 87 Xiangya Road, Changsha, 410008, Hunan, China
| | - Anhai Chen
- Department of Otolaryngology Head and Neck Surgery, Xiangya Hospital of Central South University, 87 Xiangya Road, Changsha, 410008, Hunan, China.
| | - Xuewen Wu
- Department of Otolaryngology Head and Neck Surgery, Xiangya Hospital of Central South University, 87 Xiangya Road, Changsha, 410008, Hunan, China.
| |
Collapse
|
5
|
Sun J, Wang Y, Zhang K, Shi S, Gao X, Jia X, Cong B, Zheng C. Molecular subtype construction and prognosis model for stomach adenocarcinoma characterized by metabolism-related genes. Heliyon 2024; 10:e28413. [PMID: 38596054 PMCID: PMC11002599 DOI: 10.1016/j.heliyon.2024.e28413] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2023] [Revised: 03/18/2024] [Accepted: 03/18/2024] [Indexed: 04/11/2024] Open
Abstract
Background Metabolic reprogramming is implicated in cancer progression. However, the impact of metabolism-associated genes in stomach adenocarcinomas (STAD) has not been thoroughly reviewed. Herein, we characterized metabolic transcription-correlated STAD subtypes and evaluated a metabolic RiskScore for evaluation survival. Method Genes related to metabolism were gathered from previous study and metabolic subtypes were screened using ConsensusClusterPlus in TCGA-STAD and GSE66229 dataset. The ssGSEA, MCP-Count, ESTIMATE and CIBERSORT determined the immune infiltration. A RiskScore model was established using the WGCNA and LASSO Cox regression in the TCGA-STAD queue and verified in the GSE66229 datasets. RT-qPCR was employed to measure the mRNA expressions of genes in the model. Result Two metabolism-related subtypes (C1 and C2) of STAD were constructed on account of the expression profiles of 113 prognostic metabolism genes with different immune outcomes and apparently distinct metabolic characteristic. The overall survival (OS) of C2 subtype was shorter than that of C1 subtype. Four metabolism-associated genes in turquoise model, which closely associated with C2 subtype, were employed to build the RiskScore (MATN3, OSBPL1A, SERPINE1, CPNE8) in TCGA-train dataset. Patients developed a poorer prognosis if they had a high RiskScore than having a low RiskScore. The promising effect of RiskScore was verified in the TCGA-test, TCGA-STAD and GSE66229 datasets. The prediction reliability of the RiskScore was validated by time-dependent receiver operating characteristic curve (ROC) and nomogram. Moreover, samples with high RiskScore had an enhanced immune status and TIDE score. Moreover, MATN3, OSBPL1A, SERPINE1 and CPNE8 mRNA levels were all elevated in SGC7901 cells. Inhibition of OSBPL1A decreased SGC7901 cells invasion numbers. Conclusion This work provided a new perspective into heterogeneity in metabolism and its association with immune escape in STAD. RiskScore was considered to be a strong prognostic label that could help individualize the treatment of STAD patients.
Collapse
Affiliation(s)
- Jie Sun
- Department of Gastrointestinal Surgery, Shandong Provincial Third Hospital, Jinan, 250031, China
| | - Yuanyuan Wang
- Department of Oncology and Hematology, Second Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, 250001, China
| | - Kai Zhang
- General Surgery Department, Wenshang County People's Hospital, Wenshang, 272501, China
| | - Sijia Shi
- Shandong Provincial Hospital, Jinan, 250001, China
| | - Xinxin Gao
- Gastrointestinal Surgery, Shandong First Medical University Affiliated Provincial Hospital, Jinan, 250001, China
| | - Xianghao Jia
- Gastrointestinal Surgery, Shandong Provincial Hospital, Jinan, 250001, China
| | - Bicong Cong
- Gastrointestinal Surgery, Shandong First Medical University Affiliated Provincial Hospital, Jinan, 250001, China
| | - Chunning Zheng
- Gastrointestinal Surgery, Shandong Provincial Hospital, Jinan, 250001, China
| |
Collapse
|
6
|
Su J, Zhong G, Qin W, Zhou L, Ye J, Ye Y, Chen C, Liang P, Zhao W, Xiao X, Wen W, Luo W, Zhou X, Zhang Z, Cai Y, Li C. Integrating iron metabolism-related gene signature to evaluate prognosis and immune infiltration in nasopharyngeal carcinoma. Discov Oncol 2024; 15:112. [PMID: 38602575 PMCID: PMC11009181 DOI: 10.1007/s12672-024-00969-3] [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: 02/21/2024] [Accepted: 04/03/2024] [Indexed: 04/12/2024] Open
Abstract
BACKGROUND Dysregulation of iron metabolism has been shown to have significant implications for cancer development. We aimed to investigate the prognostic and immunological significance of iron metabolism-related genes (IMRGs) in nasopharyngeal carcinoma (NPC). METHODS Multiple Gene Expression Omnibus (GEO) and The Cancer Genome Atlas (TCGA) datasets were analyzed to identify key IMRGs associated with prognosis. Additionally, the immunological significance of IMRGs was explored. RESULTS A novel risk model was established using the LASSO regression algorithm, incorporating three genes (TFRC, SLC39A14, and ATP6V0D1).This model categorized patients into low and high-risk groups, and Kaplan-Meier analysis revealed significantly shorter progression-free survival for the high-risk group (P < 0.0001). The prognostic model's accuracy was additionally confirmed by employing time-dependent Receiver Operating Characteristic (ROC) curves and conducting Decision Curve Analysis (DCA). High-risk patients were found to correlate with advanced clinical stages, specific tumor microenvironment subtypes, and distinct morphologies. ESTIMATE analysis demonstrated a significant inverse relationship between increased immune, stromal, and ESTIMATE scores and lowered risk score. Immune analysis indicated a negative correlation between high-risk score and the abundance of most tumor-infiltrating immune cells, including dendritic cells, CD8+ T cells, CD4+ T cells, and B cells. This correlation extended to immune checkpoint genes such as PDCD1, CTLA4, TIGIT, LAG3, and BTLA. The protein expression patterns of selected genes in clinical NPC samples were validated through immunohistochemistry. CONCLUSION This study presents a prognostic model utilizing IMRGs in NPC, which could assist in assessing patient prognosis and provide insights into new therapeutic targets for NPC.
Collapse
Affiliation(s)
- Jiaming Su
- Department of Otolaryngology-Head and Neck Surgery, First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China
| | - Guanlin Zhong
- Department of Otolaryngology-Head and Neck Surgery, First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China
| | - Weiling Qin
- Department of Clinical Laboratory, Wuzhou Red Cross Hospital, #3-1 Xinxing Yi Road, Wuzhou, 543002, Guangxi, China
| | - Lu Zhou
- Department of Otolaryngology-Head and Neck Surgery, First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China
| | - Jiemei Ye
- Guangxi Health Commission Key Laboratory of Molecular Epidemiology of Nasopharyngeal Carcinoma, Wuzhou Red Cross Hospital, Guangxi, China
| | - Yinxing Ye
- Department of Clinical Laboratory, Wuzhou Red Cross Hospital, #3-1 Xinxing Yi Road, Wuzhou, 543002, Guangxi, China
| | - Chang Chen
- Department of Pathology, Wuzhou Red Cross Hospital, #3-1 Xinxing Yi Road, Wuzhou, 543002, Guangxi, China
| | - Pan Liang
- Department of Otolaryngology-Head and Neck Surgery, First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China
| | - Weilin Zhao
- Department of Otolaryngology-Head and Neck Surgery, First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China
| | - Xue Xiao
- Department of Otolaryngology-Head and Neck Surgery, First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China
| | - Wensheng Wen
- Department of Otolaryngology-Head and Neck Surgery, First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China
| | - Wenqi Luo
- Department of Pathology, Affiliated Tumor Hospital of Guangxi Medical University, Nanning, Guangxi, China
| | - Xiaoying Zhou
- Key Laboratory of High-Incidence-Tumor Prevention & Treatment (Guangxi Medical University), Ministry of Education, Nanning, China
| | - Zhe Zhang
- Department of Otolaryngology-Head and Neck Surgery, First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China
| | - Yonglin Cai
- Department of Clinical Laboratory, Wuzhou Red Cross Hospital, #3-1 Xinxing Yi Road, Wuzhou, 543002, Guangxi, China.
- Guangxi Health Commission Key Laboratory of Molecular Epidemiology of Nasopharyngeal Carcinoma, Wuzhou Red Cross Hospital, Guangxi, China.
| | - Cheng Li
- Department of Pathology, Wuzhou Red Cross Hospital, #3-1 Xinxing Yi Road, Wuzhou, 543002, Guangxi, China.
| |
Collapse
|
7
|
Xue H, Sun Q, Zhang H, Huang H, Xue H. Disulfidptosis features and prognosis in head and neck squamous cell carcinoma patients: unveiling and validating the prognostic signature across cohorts. J Cancer Res Clin Oncol 2024; 150:156. [PMID: 38526631 PMCID: PMC10963584 DOI: 10.1007/s00432-024-05691-9] [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: 12/11/2023] [Accepted: 03/05/2024] [Indexed: 03/27/2024]
Abstract
BACKGROUND Head and neck squamous cell carcinoma (HNSCC) is a significant health concern with a variable global incidence and is linked to regional lifestyle factors and HPV infections. Despite treatment advances, patient prognosis remains variable, necessitating an understanding of its molecular mechanisms and the identification of reliable prognostic biomarkers. METHODS We analyzed 959 HNSCC samples and employed batch correction to obtain consistent transcriptomic data across cohorts. We examined 79 disulfidptosis-related genes to determine consensus clusters and utilized high-throughput sequencing to identify genetic heterogeneity within tumors. We established a disulfidptosis prognostic signature (DSPS) using least absolute shrinkage and selection operator (LASSO) regression and developed a prognostic nomogram integrating the DSPS with clinical factors. Personalized chemotherapy prediction was performed using the "pRRophetic" R package. RESULTS Batch corrections were used to harmonize gene expression data, revealing two distinct disulfidptosis subtypes, C1 and C2, with differential gene expression and survival outcomes. Subtype C1, characterized by increased expression of the MYH family genes ACTB, ACTN2, and FLNC, had a mortality rate of 48.4%, while subtype C2 had a mortality rate of 38.7% (HR = 0.77, 95% CI: 0.633-0.934, P = 0.008). LASSO regression identified 15 genes that composed the DSPS prognostic model, which independently predicted survival (HR = 2.055, 95% CI: 1.420-2.975, P < 0.001). The prognostic nomogram, which included the DSPS, age, and tumor stage, predicted survival with AUC values of 0.686, 0.704, and 0.789 at 3, 5, and 8 years, respectively, indicating strong predictive capability. In the external validation cohort (cohort B), the DSPS successfully identified patients at greater risk, with worse overall survival outcomes in the high-DSPS subgroup (HR = 1.54, 95% CI: 1.17-2.023, P = 0.002) and AUC values of 0.601, 0.644, 0.636, and 0.748 at 3, 5, 8, and 10 years, respectively, confirming the model's robustness. CONCLUSION The DSPS provides a robust prognostic tool for HNSCC, underscoring the complexity of this disease and the potential for tailored treatment strategies. This study highlights the importance of molecular signatures in oncology, offering a step toward personalized medicine and improved patient outcomes in HNSCC management.
Collapse
Affiliation(s)
- Hao Xue
- Department of Stomatology, The First Affiliated Hospital of Anhui Medical University, Hefei, 230022, Anhui, China
| | - Qianyu Sun
- Department of Stomatology, The First Affiliated Hospital of Anhui Medical University, Hefei, 230022, Anhui, China
| | - Heqing Zhang
- Department of Stomatology, The First Affiliated Hospital of Anhui Medical University, Hefei, 230022, Anhui, China
| | - Hanxiao Huang
- Department of Stomatology, The First Affiliated Hospital of Anhui Medical University, Hefei, 230022, Anhui, China
| | - Haowei Xue
- Department of Stomatology, The First Affiliated Hospital of Anhui Medical University, Hefei, 230022, Anhui, China.
| |
Collapse
|
8
|
Yang J, Shay C, Saba NF, Teng Y. Cancer metabolism and carcinogenesis. Exp Hematol Oncol 2024; 13:10. [PMID: 38287402 PMCID: PMC10826200 DOI: 10.1186/s40164-024-00482-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2023] [Accepted: 01/22/2024] [Indexed: 01/31/2024] Open
Abstract
Metabolic reprogramming is an emerging hallmark of cancer cells, enabling them to meet increased nutrient and energy demands while withstanding the challenging microenvironment. Cancer cells can switch their metabolic pathways, allowing them to adapt to different microenvironments and therapeutic interventions. This refers to metabolic heterogeneity, in which different cell populations use different metabolic pathways to sustain their survival and proliferation and impact their response to conventional cancer therapies. Thus, targeting cancer metabolic heterogeneity represents an innovative therapeutic avenue with the potential to overcome treatment resistance and improve therapeutic outcomes. This review discusses the metabolic patterns of different cancer cell populations and developmental stages, summarizes the molecular mechanisms involved in the intricate interactions within cancer metabolism, and highlights the clinical potential of targeting metabolic vulnerabilities as a promising therapeutic regimen. We aim to unravel the complex of metabolic characteristics and develop personalized treatment approaches to address distinct metabolic traits, ultimately enhancing patient outcomes.
Collapse
Affiliation(s)
- Jianqiang Yang
- Department of Hematology and Medical Oncology, Winship Cancer Institute, Emory University School of Medicine, 201 Dowman Dr, Atlanta, GA, 30322, USA
| | - Chloe Shay
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA, 30322, USA
| | - Nabil F Saba
- Department of Hematology and Medical Oncology, Winship Cancer Institute, Emory University School of Medicine, 201 Dowman Dr, Atlanta, GA, 30322, USA
| | - Yong Teng
- Department of Hematology and Medical Oncology, Winship Cancer Institute, Emory University School of Medicine, 201 Dowman Dr, Atlanta, GA, 30322, USA.
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA, 30322, USA.
| |
Collapse
|
9
|
Eldridge RC, Qin ZS, Saba NF, Houser MC, Hayes DN, Miller AH, Bruner DW, Jones DP, Xiao C. Unsupervised Hierarchical Clustering of Head and Neck Cancer Patients by Pre-Treatment Plasma Metabolomics Creates Prognostic Metabolic Subtypes. Cancers (Basel) 2023; 15:3184. [PMID: 37370794 PMCID: PMC10296258 DOI: 10.3390/cancers15123184] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2023] [Revised: 06/07/2023] [Accepted: 06/09/2023] [Indexed: 06/29/2023] Open
Abstract
There is growing evidence that the metabolism is deeply intertwined with head and neck squamous cell carcinoma (HNSCC) progression and survival but little is known about circulating metabolite patterns and their clinical potential. We performed unsupervised hierarchical clustering of 209 HNSCC patients via pre-treatment plasma metabolomics to identify metabolic subtypes. We annotated the subtypes via pathway enrichment analysis and investigated their association with overall and progression-free survival. We stratified the survival analyses by smoking history. High-resolution metabolomics extracted 186 laboratory-confirmed metabolites. The optimal model created two patient clusters, of subtypes A and B, corresponding to 41% and 59% of the study population, respectively. Fatty acid biosynthesis, acetyl-CoA transport, arginine and proline, as well as the galactose metabolism pathways differentiated the subtypes. Relative to subtype B, subtype A patients experienced significantly worse overall and progression-free survival but only among ever-smokers. The estimated three-year overall survival was 61% for subtype A and 86% for subtype B; log-rank p = 0.001. The association with survival was independent of HPV status and other HNSCC risk factors (adjusted hazard ratio = 3.58, 95% CI: 1.46, 8.78). Our findings suggest that a non-invasive metabolomic biomarker would add crucial information to clinical risk stratification and raise translational research questions about testing such a biomarker in clinical trials.
Collapse
Affiliation(s)
- Ronald C. Eldridge
- Nell Hodgson Woodruff School of Nursing, Emory University, Atlanta, GA 30322, USA; (M.C.H.); (D.W.B.); (C.X.)
| | - Zhaohui S. Qin
- Department of Biostatistics and Bioinformatics, Rollins School of Public Health, Emory University, Atlanta, GA 30322, USA;
| | - Nabil F. Saba
- Winship Cancer Institute, Emory University, Atlanta, GA 30322, USA;
| | - Madelyn C. Houser
- Nell Hodgson Woodruff School of Nursing, Emory University, Atlanta, GA 30322, USA; (M.C.H.); (D.W.B.); (C.X.)
| | - D. Neil Hayes
- Department of Medicine, UT/West Institute for Cancer Research, University of Tennessee Health Science Center, Memphis, TN 38163, USA;
| | - Andrew H. Miller
- Department of Psychiatry and Behavioral Sciences, School of Medicine, Emory University, Atlanta, GA 30322, USA;
| | - Deborah W. Bruner
- Nell Hodgson Woodruff School of Nursing, Emory University, Atlanta, GA 30322, USA; (M.C.H.); (D.W.B.); (C.X.)
| | - Dean P. Jones
- Division of Pulmonary, Allergy and Critical Care Medicine, Emory University, Atlanta, GA 30322, USA;
| | - Canhua Xiao
- Nell Hodgson Woodruff School of Nursing, Emory University, Atlanta, GA 30322, USA; (M.C.H.); (D.W.B.); (C.X.)
| |
Collapse
|
10
|
Lin D, Fan W, Zhang R, Zhao E, Li P, Zhou W, Peng J, Li L. Molecular subtype identification and prognosis stratification by a metabolism-related gene expression signature in colorectal cancer. J Transl Med 2021; 19:279. [PMID: 34193202 PMCID: PMC8244251 DOI: 10.1186/s12967-021-02952-w] [Citation(s) in RCA: 36] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2021] [Accepted: 06/19/2021] [Indexed: 01/05/2023] Open
Abstract
BACKGROUND Metabolic reprograming have been associated with cancer occurrence and progression within the tumor immune microenvironment. However, the prognostic potential of metabolism-related genes in colorectal cancer (CRC) has not been comprehensively studied. Here, we investigated metabolic transcript-related CRC subtypes and relevant immune landscapes, and developed a metabolic risk score (MRS) for survival prediction. METHODS Metabolism-related genes were collected from the Molecular Signatures Database and metabolic subtypes were identified using an unsupervised clustering algorithm based on the expression profiles of survival-related metabolic genes in GSE39582. The ssGSEA and ESTIMATE methods were applied to estimate the immune infiltration among subtypes. The MRS model was developed using LASSO Cox regression in the GSE39582 dataset and independently validated in the TCGA CRC and GSE17537 datasets. RESULTS We identified two metabolism-related subtypes (cluster-A and cluster-B) of CRC based on the expression profiles of 539 survival-related metabolic genes with distinct immune profiles and notably different prognoses. The cluster-B subtype had a shorter OS and RFS than the cluster-A subtype. Eighteen metabolism-related genes that were mostly involved in lipid metabolism pathways were used to build the MRS in GSE39582. Patients with higher MRS had worse prognosis than those with lower MRS (HR 3.45, P < 0.001). The prognostic role of MRS was validated in the TCGA CRC (HR 2.12, P = 0.00017) and GSE17537 datasets (HR 2.67, P = 0.039). Time-dependent receiver operating characteristic curve and stratified analyses revealed the robust predictive ability of the MRS in each dataset. Multivariate Cox regression analysis indicted that the MRS could predict OS independent of TNM stage and age. CONCLUSIONS Our study provides novel insight into metabolic heterogeneity and its relationship with immune landscape in CRC. The MRS was identified as a robust prognostic marker and may facilitate individualized therapy for CRC patients.
Collapse
Affiliation(s)
- Dagui Lin
- Department of Colorectal Surgery, State Key Laboratory of Oncology in South China, Collaborative Innovation Center of Cancer Medicine, Sun Yat-Sen University Cancer Center, 651 Dongfeng Road East, Guangzhou, 510060, Guangdong, People's Republic of China
| | - Wenhua Fan
- Department of Colorectal Surgery, State Key Laboratory of Oncology in South China, Collaborative Innovation Center of Cancer Medicine, Sun Yat-Sen University Cancer Center, 651 Dongfeng Road East, Guangzhou, 510060, Guangdong, People's Republic of China
| | - Rongxin Zhang
- Department of Colorectal Surgery, State Key Laboratory of Oncology in South China, Collaborative Innovation Center of Cancer Medicine, Sun Yat-Sen University Cancer Center, 651 Dongfeng Road East, Guangzhou, 510060, Guangdong, People's Republic of China
| | - Enen Zhao
- Department of Colorectal Surgery, State Key Laboratory of Oncology in South China, Collaborative Innovation Center of Cancer Medicine, Sun Yat-Sen University Cancer Center, 651 Dongfeng Road East, Guangzhou, 510060, Guangdong, People's Republic of China
| | - Pansong Li
- Geneplus-Beijing, Beijing, 102206, China
| | - Wenhao Zhou
- Department of Colorectal Surgery, State Key Laboratory of Oncology in South China, Collaborative Innovation Center of Cancer Medicine, Sun Yat-Sen University Cancer Center, 651 Dongfeng Road East, Guangzhou, 510060, Guangdong, People's Republic of China.
| | - Jianhong Peng
- Department of Colorectal Surgery, State Key Laboratory of Oncology in South China, Collaborative Innovation Center of Cancer Medicine, Sun Yat-Sen University Cancer Center, 651 Dongfeng Road East, Guangzhou, 510060, Guangdong, People's Republic of China.
| | - Liren Li
- Department of Colorectal Surgery, State Key Laboratory of Oncology in South China, Collaborative Innovation Center of Cancer Medicine, Sun Yat-Sen University Cancer Center, 651 Dongfeng Road East, Guangzhou, 510060, Guangdong, People's Republic of China.
| |
Collapse
|
11
|
Li Y, Weng Y, Pan Y, Huang Z, Chen X, Hong W, Lin T, Wang L, Liu W, Qiu S. A Novel Prognostic Signature Based on Metabolism-Related Genes to Predict Survival and Guide Personalized Treatment for Head and Neck Squamous Carcinoma. Front Oncol 2021; 11:685026. [PMID: 34195087 PMCID: PMC8236898 DOI: 10.3389/fonc.2021.685026] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2021] [Accepted: 05/27/2021] [Indexed: 12/30/2022] Open
Abstract
Metabolic reprogramming contributes to patient prognosis. Here, we aimed to reveal the comprehensive landscape in metabolism of head and neck squamous carcinoma (HNSCC), and establish a novel metabolism-related prognostic model to explore the clinical potential and predictive value on therapeutic response. We screened 4752 metabolism-related genes (MRGs) and then identified differentially expressed MRGs in HNSCC. A novel 10-MRGs risk model for prognosis was established by the univariate Cox regression analysis and the least absolute shrinkage and selection operator (Lasso) regression analysis, and then verified in both internal and external validation cohort. Kaplan-Meier analysis was employed to explore its prognostic power on the response of conventional therapy. The immune cell infiltration was also evaluated and we used tumor immune dysfunction and exclusion (TIDE) algorithm to estimate potential response of immunotherapy in different risk groups. Nomogram model was constructed to further predict patients’ prognoses. We found the MRGs-related prognostic model showed good prediction performance. Survival analysis indicated that patients suffered obviously poorer survival outcomes in high-risk group (p < 0.001). The metabolism-related signature was further confirmed to be the independent prognostic value of HNSCC (HR = 6.387, 95% CI = 3.281-12.432, p < 0.001), the efficacy of predictive model was also verified by internal and external validation cohorts. We observed that HNSCC patients would benefit from the application of chemotherapy in the low-risk group (p = 0.029). Immunotherapy may be effective for HNSCC patients with high risk score (p < 0.01). Furthermore, we established a predictive nomogram model for clinical application with high performance. Our study constructed and validated a promising 10-MRGs signature for monitoring outcome, which may provide potential indicators for metabolic therapy and therapeutic response prediction in HNSCC.
Collapse
Affiliation(s)
- Ying Li
- Department of Radiation Oncology, Fujian Medical University Cancer Hospital, Fujian Cancer Hospital, Fuzhou, China
| | - Youliang Weng
- Department of Radiation Oncology, Fujian Medical University Cancer Hospital, Fujian Cancer Hospital, Fuzhou, China
| | - Yuhui Pan
- Department of Radiation Oncology, Fujian Medical University Cancer Hospital, Fujian Cancer Hospital, Fuzhou, China
| | - Zongwei Huang
- Department of Radiation Oncology, Fujian Medical University Cancer Hospital, Fujian Cancer Hospital, Fuzhou, China
| | - Xiaochuan Chen
- Department of Radiation Oncology, Fujian Medical University Cancer Hospital, Fujian Cancer Hospital, Fuzhou, China
| | - Wenquan Hong
- Department of Radiation Oncology, Fujian Medical University Cancer Hospital, Fujian Cancer Hospital, Fuzhou, China
| | - Ting Lin
- Department of Radiation Oncology, Fujian Medical University Cancer Hospital, Fujian Cancer Hospital, Fuzhou, China
| | - Lihua Wang
- Department of Radiation Oncology, Fujian Medical University Cancer Hospital, Fujian Cancer Hospital, Fuzhou, China
| | - Wei Liu
- Department of Radiation Oncology, Fujian Medical University Cancer Hospital, Fujian Cancer Hospital, Fuzhou, China
| | - Sufang Qiu
- Department of Radiation Oncology, Fujian Medical University Cancer Hospital, Fujian Cancer Hospital, Fuzhou, China
| |
Collapse
|
12
|
Fang R, Iqbal M, Chen L, Liao J, Luo J, Wei F, Wen W, Sun W. A novel comprehensive immune-related gene signature as a promising survival predictor for the patients with head and neck squamous cell carcinoma. Aging (Albany NY) 2021; 13:11507-11527. [PMID: 33867351 PMCID: PMC8109104 DOI: 10.18632/aging.202842] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2020] [Accepted: 03/03/2021] [Indexed: 02/07/2023]
Abstract
Head and neck squamous cell carcinoma (HNSCC), the most frequent subtype of head and neck cancer, continues to have a poor prognosis with no improvement. The TNM stage is not satisfactory for individualized prognostic assessment and it does not predict response to therapy. In the present study, we downloaded the gene expression profiles from TCGA database to establish a training set and GEO database for a validation set. In the training set, we developed an 10 immune-related genes signature which had superior predictive value compared with TNM stage. A nomogram including clinical characteristics was also constructed for accurate prediction. Furthermore, it was determined that our prognostic signature might act as an independent factor for predicting the survival of HNSCC patients. As for the immune microenvironment, our results showed higher immune checkpoint expression (CLTA-4 and PD-1) in low-risk group which might reflect a positive immunotherapy response. Thus, our signature not only provided a promising biomarker for survival prediction, but might be evaluated as an indicator for personalized immunotherapy in patients with HNSCC.
Collapse
Affiliation(s)
- Ruihua Fang
- Department of Otorhinolaryngology Head and Neck Surgery, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou 510080, Guangdong, P.R. China.,Institute of Otorhinolaryngology Head and Neck Surgery, Sun Yat-Sen University, Guangzhou 510080, Guangdong, P.R. China.,Guangzhou Key Laboratory of Otorhinolaryngology, Guangzhou 510080, Guangdong, P.R. China
| | - Muhammad Iqbal
- Department of Otorhinolaryngology Head and Neck Surgery, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou 510080, Guangdong, P.R. China.,Institute of Otorhinolaryngology Head and Neck Surgery, Sun Yat-Sen University, Guangzhou 510080, Guangdong, P.R. China.,Guangzhou Key Laboratory of Otorhinolaryngology, Guangzhou 510080, Guangdong, P.R. China
| | - Lin Chen
- Department of Otorhinolaryngology Head and Neck Surgery, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou 510080, Guangdong, P.R. China.,Institute of Otorhinolaryngology Head and Neck Surgery, Sun Yat-Sen University, Guangzhou 510080, Guangdong, P.R. China.,Guangzhou Key Laboratory of Otorhinolaryngology, Guangzhou 510080, Guangdong, P.R. China
| | - Jing Liao
- Guangdong Institute of Gastroenterology, The Sixth Affiliated Hospital, Sun Yat-Sen University, Guangzhou 510655, Guangdong, P.R. China
| | - Jierong Luo
- Department of Anesthesia, Guangzhou First People's Hospital, The Second Affiliated Hospital of South China University of Technology, Guangzhou 510080, Guangdong, P.R. China
| | - Fanqin Wei
- Department of Otorhinolaryngology Head and Neck Surgery, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou 510080, Guangdong, P.R. China.,Institute of Otorhinolaryngology Head and Neck Surgery, Sun Yat-Sen University, Guangzhou 510080, Guangdong, P.R. China.,Guangzhou Key Laboratory of Otorhinolaryngology, Guangzhou 510080, Guangdong, P.R. China
| | - Weiping Wen
- Department of Otorhinolaryngology Head and Neck Surgery, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou 510080, Guangdong, P.R. China.,Institute of Otorhinolaryngology Head and Neck Surgery, Sun Yat-Sen University, Guangzhou 510080, Guangdong, P.R. China.,Guangzhou Key Laboratory of Otorhinolaryngology, Guangzhou 510080, Guangdong, P.R. China.,Guangdong Institute of Gastroenterology, The Sixth Affiliated Hospital, Sun Yat-Sen University, Guangzhou 510655, Guangdong, P.R. China
| | - Wei Sun
- Department of Otorhinolaryngology Head and Neck Surgery, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou 510080, Guangdong, P.R. China.,Institute of Otorhinolaryngology Head and Neck Surgery, Sun Yat-Sen University, Guangzhou 510080, Guangdong, P.R. China.,Guangzhou Key Laboratory of Otorhinolaryngology, Guangzhou 510080, Guangdong, P.R. China
| |
Collapse
|
13
|
Developing a risk scoring system based on immune-related lncRNAs for patients with gastric cancer. Biosci Rep 2021; 41:227201. [PMID: 33295609 PMCID: PMC7789809 DOI: 10.1042/bsr20202203] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2020] [Revised: 11/29/2020] [Accepted: 12/07/2020] [Indexed: 12/24/2022] Open
Abstract
The immune system and the tumor interact closely during tumor development. Aberrantly expressed long non-coding RNAs (lncRNAs) may be potentially applied as diagnostic and prognostic markers for gastric cancer (GC). At present, the diagnosis and treatment of GC patients remain a formidable clinical challenge. The present study aimed to build a risk scoring system to improve the prognosis of GC patients. In the present study, ssGSEA was used to evaluate the infiltration of immune cells in GC tumor tissue samples, and the samples were split into a high immune cell infiltration group and a low immune cell infiltration group. About 1262 differentially expressed lncRNAs between the high immune cell infiltration group and the low immune cell infiltration group. About 3204 differentially expressed lncRNAs between GC tumor tissues and paracancerous tissues were identified. Then, 621 immune-related lncRNAs were screened using a Venn analysis based on the above results, and 85 prognostic lncRNAs were identified using a univariate Cox analysis. We constructed a prognostic signature using LASSO analysis and evaluated the predictive performance of the signature using ROC analysis. GO and KEGG enrichment analyses were performed on the lncRNAs using the R package, ‘clusterProfiler’. The TIMER online database was used to analyze correlations between the risk score and the abundances of the six types of immune cells. In conclusion, our study found that specific immune-related lncRNAs were clinically significant. These lncRNAs were used to construct a reliable prognostic signature and analyzed immune infiltrates, which may assist clinicians in developing individualized treatment strategies for GC patients.
Collapse
|
14
|
Wu X, Yao Y, Li Z, Ge H, Wang D, Wang Y. Identification of a Transcriptional Prognostic Signature From Five Metabolic Pathways in Oral Squamous Cell Carcinoma. Front Oncol 2020; 10:572919. [PMID: 33425725 PMCID: PMC7793793 DOI: 10.3389/fonc.2020.572919] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2020] [Accepted: 11/02/2020] [Indexed: 12/18/2022] Open
Abstract
Dysregulated metabolic pathways have been appreciated to be intimately associated with tumorigenesis and patient prognosis. Here, we sought to develop a novel prognostic signature based on metabolic pathways in patients with primary oral squamous cell carcinoma (OSCC). The original RNA-seq data of OSCC from The Cancer Genome Atlas (TCGA) project and Gene Expression Omnibus (GEO) database were transformed into a metabolic pathway enrichment score matrix by single-sample gene set enrichment analysis (ssGSEA). A novel prognostic signature based on metabolic pathways was constructed by LASSO and stepwise Cox regression analysis in the training cohort and validated in both testing and validation cohorts. The optimal cut-off value was obtained using the Youden index by receiver operating characteristic (ROC) curve. The overall survival curves were plotted by the Kaplan-Meier method. A time-dependent ROC curve analysis with 1, 3, 5 years as the defining point was performed to evaluate the predictive value of this prognostic signature. A 5-metabolic pathways prognostic signature (5MPS) for OSCC was constructed which stratified patients into subgroups with favorable or inferior survival. It served as an independent prognostic factor for patient survival and had a satisfactory predictive performance for OSCC. Our results developed a novel prognostic signature based on dysregulated metabolic pathways in OSCC and provided support for aberrant metabolism underlying OSCC tumorigenesis.
Collapse
Affiliation(s)
- Xiang Wu
- Jiangsu Key Laboratory of Oral Disease, Nanjing Medical University, Nanjing, China
| | - Yuan Yao
- Jiangsu Key Laboratory of Oral Disease, Nanjing Medical University, Nanjing, China
| | - Zhongwu Li
- Jiangsu Key Laboratory of Oral Disease, Nanjing Medical University, Nanjing, China
| | - Han Ge
- Jiangsu Key Laboratory of Oral Disease, Nanjing Medical University, Nanjing, China.,Department of Oral and Maxillofacial Surgery, Affiliated Hospital of Stomatology, Nanjing Medical University, Nanjing, China
| | - Dongmiao Wang
- Department of Oral and Maxillofacial Surgery, Affiliated Hospital of Stomatology, Nanjing Medical University, Nanjing, China
| | - Yanling Wang
- Jiangsu Key Laboratory of Oral Disease, Nanjing Medical University, Nanjing, China.,Department of Oral and Maxillofacial Surgery, Affiliated Hospital of Stomatology, Nanjing Medical University, Nanjing, China
| |
Collapse
|
15
|
Gene Expression Clustering and Selected Head and Neck Cancer Gene Signatures Highlight Risk Probability Differences in Oral Premalignant Lesions. Cells 2020; 9:cells9081828. [PMID: 32756466 PMCID: PMC7466020 DOI: 10.3390/cells9081828] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Revised: 07/27/2020] [Accepted: 07/31/2020] [Indexed: 12/21/2022] Open
Abstract
BACKGROUND Oral premalignant lesions (OPLs) represent the most common oral precancerous conditions. One of the major challenges in this field is the identification of OPLs at higher risk for oral squamous cell cancer (OSCC) development, by discovering molecular pathways deregulated in the early steps of malignant transformation. Analysis of deregulated levels of single genes and pathways has been successfully applied to head and neck squamous cell cancers (HNSCC) and OSCC with prognostic/predictive implications. Exploiting the availability of gene expression profile and clinical follow-up information of a well-characterized cohort of OPL patients, we aim to dissect tissue OPL gene expression to identify molecular clusters/signatures associated with oral cancer free survival (OCFS). MATERIALS AND METHODS The gene expression data of 86 OPL patients were challenged with: an HNSCC specific 6 molecular subtypes model (Immune related: HPV related, Defense Response and Immunoreactive; Mesenchymal, Hypoxia and Classical); one OSCC-specific signature (13 genes); two metabolism-related signatures (3 genes and signatures raised from 6 metabolic pathways associated with prognosis in HNSCC and OSCC, respectively); a hypoxia gene signature. The molecular stratification and high versus low expression of the signatures were correlated with OCFS by Kaplan-Meier analyses. The association of gene expression profiles among the tested biological models and clinical covariates was tested through variance partition analysis. RESULTS Patients with Mesenchymal, Hypoxia and Classical clusters showed an higher risk of malignant transformation in comparison with immune-related ones (log-rank test, p = 0.0052) and they expressed four enriched hallmarks: "TGF beta signaling" "angiogenesis", "unfolded protein response", "apical junction". Overall, 54 cases entered in the immune related clusters, while the remaining 32 cases belonged to the other clusters. No other signatures showed association with OCFS. Our variance partition analysis proved that clinical and molecular features are able to explain only 21% of gene expression data variability, while the remaining 79% refers to residuals independent of known parameters. CONCLUSIONS Applying the existing signatures derived from HNSCC to OPL, we identified only a protective effect for immune-related signatures. Other gene expression profiles derived from overt cancers were not able to identify the risk of malignant transformation, possibly because they are linked to later stages of cancer progression. The availability of a new well-characterized set of OPL patients and further research is needed to improve the identification of adequate prognosticators in OPLs.
Collapse
|