1
|
Uttam V, Kapoor HS, Rana MK, Yadav R, Prakash H, Jain M, Tuli HS, Jain A. Immune-Related Long Non-Coding RNA Signature Determines Prognosis and Immunotherapeutic Coherence in Esophageal Cancer. Cancer Inform 2024; 23:11769351241276757. [PMID: 39282627 PMCID: PMC11401149 DOI: 10.1177/11769351241276757] [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: 04/01/2024] [Accepted: 07/31/2024] [Indexed: 09/19/2024] Open
Abstract
Objectives Aim of this study was to explore the immune-related lncRNAs having prognostic role and establishing risk score model for better prognosis and immunotherapeutic coherence for esophageal cancer (EC) patients. Methods To determine the role of immune-related lncRNAs in EC, we analyzed the RNA-seq expression data of 162 EC patients and 11 non-cancerous individuals and their clinically relevant information from the cancer genome atlas (TCGA) database. Bioinformatic and statistical analysis such as Differential expression analysis, co-expression analysis, Kaplan Meier survival analysis, Cox proportional hazards model, ROC analysis of risk model was employed. Results Utilizing a cutoff criterion (log2FC > 1 + log2FC < -1 and FDR < 0.01), we identified 3737 RNAs were significantly differentially expressed in EC patients. Among these, 2222 genes were classified as significantly differentially expressed mRNAs (demRNAs), and 966 were significantly differentially expressed lncRNAs (delncRNA). Through Pearson correlation analysis between differentially expressed lncRNAs and immune related-mRNAs, we identified 12 immune-related lncRNAs as prognostic signatures for EC. Notably, through Kaplan-Meier analysis on these lncRNAs, we found the low-risk group patients showed significantly improved survival compared to the high-risk group. Moreover, this prognostic signature has consistent performance across training, testing and entire validation cohort sets. Using ESTIMATE and CIBERSORT algorithm we further observed significant enriched infiltration of naive B cells, regulatory T cells resting CD4+ memory T cells, and, plasma cells in the low-risk group compared to high-risk EC patients group. On the contrary, tumor-associated M2 macrophages were highly enriched in high-risk patients. Additionally, we confirmed immune-related biological functions and pathways such as inflammatory, cytokines, chemokines response and natural killer cell-mediated cytotoxicity, toll-like receptor signaling pathways, JAK-STAT signaling pathways, chemokine signaling pathways significantly associated with identified IRlncRNA signature and their co-expressed immune genes. Furthermore, we assessed the predictive potential of the lncRNA signature in immune checkpoint inhibitors; we found that programed cell death ligand 1 (PD-L1; P-value = .048), programed cell death ligand 2 (PD-L2; P-value = .002), and T cell immunoglobulin and mucin-domain containing-3 (TIM-3; P-value = .045) expression levels were significantly higher in low-risk patients compared to high-risk patients. Conclusion We believe this study will contribute to better prognosis prediction and targeted treatment of EC in the future.
Collapse
Affiliation(s)
- Vivek Uttam
- Non-Coding RNA and Cancer Biology Lab, Department of Zoology, Central University of Punjab, Ghudda, Punjab, India
| | | | - Manjit Kaur Rana
- Department of Pathology/Lab Medicine, AIIMS, Bathinda, Punjab, India
| | - Ritu Yadav
- Non-Coding RNA and Cancer Biology Lab, Department of Zoology, Central University of Punjab, Ghudda, Punjab, India
| | | | - Manju Jain
- Department of Biochemistry, Central University of Punjab, Ghudda, Punjab, India
| | - Hardeep Singh Tuli
- Department of Biotechnology, Maharishi Markandeshwar (Deemed to be University), Mullana, Ambala, Haryana, India
| | - Aklank Jain
- Non-Coding RNA and Cancer Biology Lab, Department of Zoology, Central University of Punjab, Ghudda, Punjab, India
| |
Collapse
|
2
|
Masrour M, Khanmohammadi S, Fallahtafti P, Hashemi SM, Rezaei N. Long non-coding RNA as a potential diagnostic and prognostic biomarker in melanoma: A systematic review and meta-analysis. J Cell Mol Med 2024; 28:e18109. [PMID: 38193829 PMCID: PMC10844705 DOI: 10.1111/jcmm.18109] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2023] [Revised: 11/25/2023] [Accepted: 11/30/2023] [Indexed: 01/10/2024] Open
Abstract
Recently, long noncoding RNAs (lncRNAs) have been applied as biomarkers for melanoma patients. In this systematic review and meta-analysis, we investigated the diagnostic and prognostic value of lncRNAs. We used the keywords 'lncRNA' and 'melanoma' to search databases for studies published before June 14th, 2023. The specificity, sensitivity and AUC were utilized to assess diagnostic accuracy and the prognostic value was assessed using overall survival, progression-free survival and disease-free survival hazard ratios. After screening 1191 articles, we included seven studies in the diagnostic evaluation section and 17 studies in the prognosis evaluation section. The Reitsma bivariate model estimated a cumulative sensitivity of 0.724 (95% CI: 0.659-0.781, p < 0.001) and specificity of 0.812 (95% CI: 0.752-0.859, p < 0.001). The pooled AUC was 0.780 (95% CI: 0.749-0.811, p < 0.0001). The HR for overall survival was 2.723 (95% CI: 2.259-3.283, p < 0.0001). Two studies reported an HR for overall survival less than one, with an HR of 0.348 (95% CI: 0.200-0.607, p < 0.0002). The HR for progression-free survival was 2.913 (95% CI: 2.050-4.138, p < 0.0001). Four studies reported an HR less than one, with an HR of 0.457 (95% CI: 0.256-0.817). The HR for disease-free survival was 2.760 (95% CI: 2.009-3.792, p < 0.0001). In conclusion, the expression of lncRNAs in melanoma patients affects survival and prognosis. LncRNAs can also be employed as diagnostic biomarkers.
Collapse
Affiliation(s)
- Mahdi Masrour
- School of MedicineTehran University of Medical SciencesTehranIran
| | - Shaghayegh Khanmohammadi
- School of MedicineTehran University of Medical SciencesTehranIran
- Research Center for Immunodeficiencies, Pediatrics Center of Excellence, Children's Medical CenterTehran University of Medical SciencesTehranIran
- Non‐Communicable Diseases Research Center, Endocrinology and Metabolism Population Sciences InstituteTehran University of Medical SciencesTehranIran
| | - Parisa Fallahtafti
- School of MedicineTehran University of Medical SciencesTehranIran
- Tehran Heart Center, Cardiovascular Diseases Research InstituteTehran University of Medical SciencesTehranIran
| | - Seyedeh Melika Hashemi
- School of MedicineTehran University of Medical SciencesTehranIran
- Tehran Heart Center, Cardiovascular Diseases Research InstituteTehran University of Medical SciencesTehranIran
| | - Nima Rezaei
- Research Center for Immunodeficiencies, Pediatrics Center of Excellence, Children's Medical CenterTehran University of Medical SciencesTehranIran
- Non‐Communicable Diseases Research Center, Endocrinology and Metabolism Population Sciences InstituteTehran University of Medical SciencesTehranIran
- Department of Immunology, School of MedicineTehran University of Medical SciencesTehranIran
| |
Collapse
|
3
|
Roccuzzo G, Bongiovanni E, Tonella L, Pala V, Marchisio S, Ricci A, Senetta R, Bertero L, Ribero S, Berrino E, Marchiò C, Sapino A, Quaglino P, Cassoni P. Emerging prognostic biomarkers in advanced cutaneous melanoma: a literature update. Expert Rev Mol Diagn 2024; 24:49-66. [PMID: 38334382 DOI: 10.1080/14737159.2024.2314574] [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: 08/05/2023] [Accepted: 02/01/2024] [Indexed: 02/10/2024]
Abstract
INTRODUCTION Over the past two years, the scientific community has witnessed an exponential growth in research focused on identifying prognostic biomarkers for melanoma, both in pre-clinical and clinical settings. This surge in studies reflects the need of developing effective prognostic indicators in the field of melanoma. AREAS COVERED The aim of this work is to review the scientific literature on the most recent findings on the development or validation of prognostic biomarkers in melanoma, in the attempt of providing both clinicians and researchers with an updated broad synopsis of prognostic biomarkers in cutaneous melanoma. EXPERT OPINION While the field of prognostic biomarkers in melanoma appears promising, there are several complexities and limitations to address. The interdependence of clinical, histological, and molecular features requires accurate classification of different biomarker families. Correlation does not imply causation, and adjustments for confounding factors are often overlooked. In this scenario, large-scale studies based on high-quality clinical trial data can provide more reliable evidence. It is essential to avoid oversimplification by focusing on a single biomarker, as the interactions among multiple factors contribute to define the disease course and patient's outcome. Furthermore, implementing well-supported evidence in real-life settings can help advance prognostic biomarker research in melanoma.
Collapse
Affiliation(s)
- Gabriele Roccuzzo
- Department of Medical Sciences, Section of Dermatology, University of Turin, Turin, Italy
| | - Eleonora Bongiovanni
- Department of Medical Sciences, Section of Dermatology, University of Turin, Turin, Italy
| | - Luca Tonella
- Department of Medical Sciences, Section of Dermatology, University of Turin, Turin, Italy
| | - Valentina Pala
- Department of Medical Sciences, Section of Dermatology, University of Turin, Turin, Italy
| | - Sara Marchisio
- Pathology Unit, Department of Medical Sciences, University of Turin, Turin, Italy
| | - Alessia Ricci
- Pathology Unit, Department of Medical Sciences, University of Turin, Turin, Italy
| | - Rebecca Senetta
- Pathology Unit, Department of Medical Sciences, University of Turin, Turin, Italy
| | - Luca Bertero
- Pathology Unit, Department of Medical Sciences, University of Turin, Turin, Italy
| | - Simone Ribero
- Department of Medical Sciences, Section of Dermatology, University of Turin, Turin, Italy
| | - Enrico Berrino
- Candiolo Cancer Institute, FPO-IRCCS, Candiolo, Italy
- Department of Medical Sciences, University of Turin, Turin, Italy
| | - Caterina Marchiò
- Candiolo Cancer Institute, FPO-IRCCS, Candiolo, Italy
- Department of Medical Sciences, University of Turin, Turin, Italy
| | - Anna Sapino
- Candiolo Cancer Institute, FPO-IRCCS, Candiolo, Italy
- Department of Medical Sciences, University of Turin, Turin, Italy
| | - Pietro Quaglino
- Department of Medical Sciences, Section of Dermatology, University of Turin, Turin, Italy
| | - Paola Cassoni
- Pathology Unit, Department of Medical Sciences, University of Turin, Turin, Italy
| |
Collapse
|
4
|
Natarelli N, Boby A, Aflatooni S, Tran JT, Diaz MJ, Taneja K, Forouzandeh M. Regulatory miRNAs and lncRNAs in Skin Cancer: A Narrative Review. Life (Basel) 2023; 13:1696. [PMID: 37629553 PMCID: PMC10455148 DOI: 10.3390/life13081696] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2023] [Revised: 07/26/2023] [Accepted: 07/28/2023] [Indexed: 08/27/2023] Open
Abstract
Non-coding RNAs (ncRNAs) have a significant regulatory role in the pathogenesis of skin cancer, despite the fact that protein-coding genes have generally been the focus of research efforts in the field. We comment on the actions of long non-coding RNAs (lncRNAs) and microRNAs (miRNAs) in the current review with an eye toward potential therapeutic treatments. LncRNAs are remarkably adaptable, acting as scaffolding, guides, or decoys to modify key signaling pathways (i.e., the Wnt/β-catenin pathway) and gene expression. As post-transcriptional gatekeepers, miRNAs control gene expression by attaching to messenger RNAs and causing their degradation or suppression during translation. Cell cycle regulation, cellular differentiation, and immunological responses are all affected by the dysregulation of miRNAs observed in skin cancer. NcRNAs also show promise as diagnostic biomarkers and prognostic indicators. Unraveling the complexity of the regulatory networks governed by ncRNAs in skin cancer offers unprecedented opportunities for groundbreaking targeted therapies, revolutionizing the landscape of dermatologic care.
Collapse
Affiliation(s)
- Nicole Natarelli
- Morsani College of Medicine, University of South Florida, Tampa, FL 33602, USA
| | - Aleena Boby
- Morsani College of Medicine, University of South Florida, Tampa, FL 33602, USA
| | - Shaliz Aflatooni
- Morsani College of Medicine, University of South Florida, Tampa, FL 33602, USA
| | - Jasmine Thuy Tran
- School of Medicine, University of Indiana, Indianapolis, IN 46202, USA;
| | | | - Kamil Taneja
- Renaissance School of Medicine, Stony Brook University, Stony Brook, NY 11794, USA
| | - Mahtab Forouzandeh
- Department of Dermatology, University of Florida, Gainesville, FL 32606, USA
| |
Collapse
|
5
|
Li W, Zhan Y, Peng C, Wang Z, Xu T, Liu M. A model based on immune-related lncRNA pairs and its potential prognostic value in immunotherapy for melanoma. Funct Integr Genomics 2023; 23:91. [PMID: 36939945 DOI: 10.1007/s10142-023-01029-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2022] [Revised: 03/10/2023] [Accepted: 03/13/2023] [Indexed: 03/21/2023]
Abstract
A model based on long non-coding RNA (lncRNA) pairs independent of expression quantification was constructed to evaluate prognosis melanoma and response to immunotherapy in melanoma. RNA sequencing data and clinical information were retrieved and downloaded from The Cancer Genome Atlas and the Genotype-Tissue Expression databases. We identified differentially expressed immune-related lncRNAs (DEirlncRNAs), matched them, and used least absolute shrinkage and selection operator and Cox regression to construct predictive models. The optimal cutoff value of the model was determined using a receiver operating characteristic curve and used to categorize melanoma cases into high-risk and low-risk groups. The predictive efficacy of the model with respect to prognosis was compared with that of clinical data and ESTIMATE (Estimation of STromal and Immune cells in MAlignant Tumor tissues using Expression data). Then, we analyzed the correlations of risk score with clinical characteristics, immune cell invasion, anti-tumor, and tumor-promoting activities. Differences in survival, degree of immune cell infiltration, and intensity of anti-tumor and tumor-promoting activities were also evaluated in the high- and low-risk groups. A model based on 21 DEirlncRNA pairs was established. Compared with ESTIMATE score and clinical data, this model could better predict outcomes of melanoma patients. Follow-up analysis of the model's effectiveness showed that patients in the high-risk group had poorer prognosis and were less likely to benefit from immunotherapy compared with those in the low-risk group. Moreover, there were differences in tumor-infiltrating immune cells between the high-risk and low-risk groups. By pairing the DEirlncRNA, we constructed a model to evaluate the prognosis of cutaneous melanoma independent of a specific level of lncRNA expression.
Collapse
Affiliation(s)
- Wenshuai Li
- Department of Clinical Laboratory, Key Laboratory of Laboratory Medicine, The Affiliated Hospital of Qingdao University, 16 Jiangsu Road, Qingdao, 266003, China
| | - Yingxuan Zhan
- Department of Clinical Laboratory, Key Laboratory of Laboratory Medicine, The Affiliated Hospital of Qingdao University, 16 Jiangsu Road, Qingdao, 266003, China
| | - Chong Peng
- Department of Clinical Laboratory, Key Laboratory of Laboratory Medicine, The Affiliated Hospital of Qingdao University, 16 Jiangsu Road, Qingdao, 266003, China
| | - Zhan Wang
- Department of Clinical Laboratory, Key Laboratory of Laboratory Medicine, The Affiliated Hospital of Qingdao University, 16 Jiangsu Road, Qingdao, 266003, China
| | - Tiantian Xu
- Department of Clinical Laboratory, Key Laboratory of Laboratory Medicine, The Affiliated Hospital of Qingdao University, 16 Jiangsu Road, Qingdao, 266003, China
| | - Mingjun Liu
- Department of Clinical Laboratory, Key Laboratory of Laboratory Medicine, The Affiliated Hospital of Qingdao University, 16 Jiangsu Road, Qingdao, 266003, China.
| |
Collapse
|
6
|
Li AA, Li F, Lan M, Zhang Y, Xie D, Yan MY. A novel endoplasmic reticulum stress-related lncRNA prognostic risk model for cutaneous melanoma. J Cancer Res Clin Oncol 2022; 148:3227-3241. [PMID: 35687183 DOI: 10.1007/s00432-022-04086-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: 04/06/2022] [Accepted: 05/20/2022] [Indexed: 12/24/2022]
Abstract
OBJECTIVE Endoplasmic reticulum stress (ERS) and long non-coding RNAs (lncRNAs) are important in melanoma development and progression. This study aimed to explore the prognostic value of ERS-associated lncRNA profiles in cutaneous melanoma (CM). METHODS The Cancer Genome Atlas (TCGA) provides the raw data of CM. GSEA website was used to obtain ERS-related genes, and mRNA and LncRNA co-expression network were used to obtain ERS-related lncRNAs. A Lasso regression analysis was used to identify a prognostic risk model for the composition of ERS-related lncRNAs. Patients were divided into high- and low-risk groups based on the model's risk score. The researchers then compared the two groups' survival rates, immune infiltration, chemotherapeutic drug sensitivity, and immune checkpoint gene expression. RESULTS Thirty-nine ERS-related lncRNAs were discovered to be prognostic. A prognostic risk model made up of ten ERS-related lncRNAs was discovered. Patients in the low-risk group had a better prognosis than those in the high-risk group. An examination of tumor microenvironment revealed that risk scores correlated with immune cell infiltration in eight cases. Dacarbazine, paclitaxel, and cisplatin, three chemotherapy drugs, were more sensitive in the low-risk group than in the high-risk group. CONCLUSION This study identified a risk model of ten ERS-related lncRNAs that have significant prognostic value in CM and could help guide clinical treatment.
Collapse
Affiliation(s)
- An-An Li
- Department of Orthopedic Surgery, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, People's Republic of China
| | - Fan Li
- Ji'an College, Ji'an, Jiangxi, People's Republic of China
| | - Min Lan
- Department of Orthopedic Surgery, Jiangxi Provincial People's Hospital, Nanchang, Jiangxi, People's Republic of China
| | - Yu Zhang
- Department of Orthopedic Surgery, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, People's Republic of China
| | - Dong Xie
- Department of Dermatology, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, People's Republic of China
| | - Mei-Ying Yan
- Medical Imaging Center, The Second Affiliated Hospital of Nanchang University, No. 1 Mingde Road, Donghu District, Nanchang, 330006, Jiangxi, People's Republic of China.
| |
Collapse
|
7
|
Huang A, Lv B, Zhang Y, Yang J, Li J, Li C, Yu Z, Xia J. Construction of a tumor immune infiltration macrophage signature for predicting prognosis and immunotherapy response in liver cancer. Front Mol Biosci 2022; 9:983840. [PMID: 36120553 PMCID: PMC9479109 DOI: 10.3389/fmolb.2022.983840] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2022] [Accepted: 08/03/2022] [Indexed: 11/13/2022] Open
Abstract
Liver cancer is an extraordinarily heterogeneous malignant disease. The tumor microenvironment (TME) and tumor-associated macrophages (TAMs) are the major drivers of liver cancer initiation and progression. It is critical to have a better understanding of the complicated interactions between liver cancer and the immune system for the development of cancer immunotherapy. Based on the gene expression profiles of tumor immune infiltration cells (TIICs), upregulated genes in TAMs and downregulated genes in other types of immune cells were identified as macrophage-specific genes (MSG). In this study, we combined MSG, immune subtypes, and clinical information on liver cancer to develop a tumor immune infiltration macrophage signature (TIMSig). A four-gene signature (S100A9, SLC22A15, TRIM54, and PPARGC1A) was identified as the TAM-related prognostic genes for liver cancer, independent of multiple clinicopathological parameters. Survival analyses showed that patients with low TIMSig had a superior survival rate than those with high TIMSig. Additionally, clinical immunotherapy response and TIMSig was observed as highly relevant. In addition, TIMSig could predict the response to chemotherapy. Collectively, the TIMSig could be a potential tool for risk-stratification, clinical decision making, treatment planning, and oncology immunotherapeutic drug development.
Collapse
Affiliation(s)
- Anmin Huang
- Key Laboratory of Diagnosis and Treatment of Severe Hepato-Pancreatic Diseases of Zhejiang Province, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
- Translational Medicine Laboratory, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
- The First Clinical Medical College, Wenzhou Medical University, Wenzhou, China
| | - Bei Lv
- Department of Radiation Oncology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Yunjie Zhang
- Key Laboratory of Diagnosis and Treatment of Severe Hepato-Pancreatic Diseases of Zhejiang Province, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
- Translational Medicine Laboratory, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
- The First Clinical Medical College, Wenzhou Medical University, Wenzhou, China
| | - Junhui Yang
- Key Laboratory of Diagnosis and Treatment of Severe Hepato-Pancreatic Diseases of Zhejiang Province, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
- Translational Medicine Laboratory, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
- The First Clinical Medical College, Wenzhou Medical University, Wenzhou, China
| | - Jie Li
- Key Laboratory of Diagnosis and Treatment of Severe Hepato-Pancreatic Diseases of Zhejiang Province, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
- Translational Medicine Laboratory, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
- The First Clinical Medical College, Wenzhou Medical University, Wenzhou, China
| | - Chengjun Li
- Key Laboratory of Diagnosis and Treatment of Severe Hepato-Pancreatic Diseases of Zhejiang Province, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
- Translational Medicine Laboratory, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
- The First Clinical Medical College, Wenzhou Medical University, Wenzhou, China
| | - Zhijie Yu
- Wenzhou Key Laboratory of Hematology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
- *Correspondence: Zhijie Yu, ; Jinglin Xia,
| | - Jinglin Xia
- Key Laboratory of Diagnosis and Treatment of Severe Hepato-Pancreatic Diseases of Zhejiang Province, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
- Department of Intervention, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
- Liver Cancer Institute, Zhongshan Hospital, Fudan University, Shanghai, China
- *Correspondence: Zhijie Yu, ; Jinglin Xia,
| |
Collapse
|
8
|
Xu Y, Chen Y, Niu Z, Yang Z, Xing J, Yin X, Guo L, Zhang Q, Yang Y, Han Y. Ferroptosis-related lncRNA signature predicts prognosis and immunotherapy efficacy in cutaneous melanoma. Front Surg 2022; 9:860806. [PMID: 35937602 PMCID: PMC9354448 DOI: 10.3389/fsurg.2022.860806] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2022] [Accepted: 07/07/2022] [Indexed: 11/30/2022] Open
Abstract
Purpose Ferroptosis-related lncRNAs are promising biomarkers for predicting the prognosis of many cancers. However, a ferroptosis-related signature to predict the prognosis of cutaneous melanoma (CM) has not been identified. The purpose of this study was to construct a ferroptosis-related lncRNA signature to predict prognosis and immunotherapy efficacy in CM. Methods Ferroptosis-related differentially expressed genes (FDEGs) and lncRNAs (FDELs) were identified using TCGA, GTEx, and FerrDb datasets. We performed Cox and LASSO regressions to identify key FDELs, and constructed a risk score to stratify patients into high- and low-risk groups. The lncRNA signature was evaluated using the areas under the receiver operating characteristic curves (AUCs) and Kaplan-Meier analyses in the training, testing, and entire cohorts. Multivariate Cox regression analyses including the lncRNA signature and common clinicopathological characteristics were performed to identify independent predictors of overall survival (OS). A nomogram was developed for clinical use. We performed gene set enrichment analyses (GSEA) to identify significantly enriched pathways. Differences in the tumor microenvironment (TME) between the 2 groups were assessed using 7 algorithms. To predict the efficacy of immune checkpoint inhibitors (ICI), we analyzed the association between PD1 and CTLA4 expression and the risk score. Finally, differences in Tumor Mutational Burden (TMB) and molecular drugs Sensitivity between the 2 groups were performed. Results We identified 5 lncRNAs (AATBC, AC145423.2, LINC01871, AC125807.2, and AC245041.1) to construct the risk score. The AUC of the lncRNA signature was 0.743 in the training cohort and was validated in the testing and entire cohorts. Kaplan-Meier analyses revealed that the high-risk group had poorer prognosis. Multivariate Cox regression showed that the lncRNA signature was an independent predictor of OS with higher accuracy than traditional clinicopathological features. The 1-, 3-, and 5-year survival probabilities for CM patients were 92.7%, 57.2%, and 40.2% with an AUC of 0.804, indicating a good accuracy and reliability of the nomogram. GSEA showed that the high-risk group had lower ferroptosis and immune response. TME analyses confirmed that the high-risk group had lower immune cell infiltration (e.g., CD8+ T cells, CD4+ memory-activated T cells, and M1 macrophages) and lower immune functions (e.g., immune checkpoint activation). Low-risk patients whose disease expressed PD1 or CTLA4 were likely to respond better to ICIs. The analysis demonstrated that the TMB had significantly difference between low- and high- risk groups. Chemotherapy drugs, such as sorafenib, Imatinib, ABT.888 (Veliparib), Docetaxel, and Paclitaxel showed Significant differences in the estimated IC50 between the two risk groups. Conclusion Our novel ferroptosis-related lncRNA signature was able to accurately predict the prognosis and ICI outcomes of CM patients. These ferroptosis-related lncRNAs might be potential biomarkers and therapeutic targets for CM.
Collapse
Affiliation(s)
- Yujian Xu
- Department of Plastic and Reconstructive Surgery, Chinese PLA General Hospital, Beijing, China
| | - Youbai Chen
- Department of Plastic and Reconstructive Surgery, Chinese PLA General Hospital, Beijing, China
| | - Zehao Niu
- Department of Plastic and Reconstructive Surgery, Chinese PLA General Hospital, Beijing, China
| | - Zheng Yang
- Department of Plastic and Reconstructive Surgery, Chinese PLA General Hospital, Beijing, China
| | - Jiahua Xing
- Department of Plastic and Reconstructive Surgery, Chinese PLA General Hospital, Beijing, China
| | - Xiangye Yin
- Department of Plastic and Reconstructive Surgery, Chinese PLA General Hospital, Beijing, China
| | - Lingli Guo
- Department of Plastic and Reconstructive Surgery, Chinese PLA General Hospital, Beijing, China
| | - Qixu Zhang
- Department of Plastic Surgery, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Yi Yang
- Department of Dermatology, Chinese PLA General Hospital, Beijing, China
- Correspondence: Yan Han Yi Yang
| | - Yan Han
- Department of Plastic and Reconstructive Surgery, Chinese PLA General Hospital, Beijing, China
- Correspondence: Yan Han Yi Yang
| |
Collapse
|
9
|
Melixetian M, Pelicci PG, Lanfrancone L. Regulation of LncRNAs in Melanoma and Their Functional Roles in the Metastatic Process. Cells 2022; 11:577. [PMID: 35159386 PMCID: PMC8834033 DOI: 10.3390/cells11030577] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2022] [Revised: 02/02/2022] [Accepted: 02/05/2022] [Indexed: 02/06/2023] Open
Abstract
Long non-coding RNAs (lncRNAs) are key regulators of numerous intracellular processes leading to tumorigenesis. They are frequently deregulated in cancer, functioning as oncogenes or tumor suppressors. As they act through multiple mechanisms, it is not surprising that they may exert dual functions in the same tumor. In melanoma, a highly invasive and metastatic tumor with the propensity to rapidly develop drug resistance, lncRNAs play different roles in: (i) guiding the phenotype switch and leading to metastasis formation; (ii) predicting the response of melanoma patients to immunotherapy; (iii) triggering adaptive responses to therapy and acquisition of drug resistance phenotypes. In this review we summarize the most recent findings on the lncRNAs involved in melanoma growth and spreading to distant sites, focusing on their role as biomarkers for disease diagnosis and patient prognosis, or targets for novel therapeutic approaches.
Collapse
Affiliation(s)
- Marine Melixetian
- Department of Experimental Oncology, IEO, European Institute of Oncology IRCCS, 20139 Milan, Italy; (M.M.); (P.G.P.)
| | - Pier Giuseppe Pelicci
- Department of Experimental Oncology, IEO, European Institute of Oncology IRCCS, 20139 Milan, Italy; (M.M.); (P.G.P.)
- Department of Oncology and Hemato-Oncology, University of Milan, 20122 Milan, Italy
| | - Luisa Lanfrancone
- Department of Experimental Oncology, IEO, European Institute of Oncology IRCCS, 20139 Milan, Italy; (M.M.); (P.G.P.)
| |
Collapse
|
10
|
Li FW, Luo SK. Identification and Construction of a Predictive Immune-Related lncRNA Signature Model for Melanoma. Int J Gen Med 2021; 14:9227-9235. [PMID: 34880662 PMCID: PMC8647169 DOI: 10.2147/ijgm.s340025] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2021] [Accepted: 11/25/2021] [Indexed: 01/18/2023] Open
Abstract
Objective The occurrence and development mechanisms of melanoma are related to immunity and lncRNAs. Therefore, it is necessary to systematically explore immune-related lncRNA profiles to help improve the prognosis of melanoma. Methods We integrated immune-related lncRNAs and the basic clinical information of melanoma patients in the TCGA dataset. Immune-associated lncRNAs were selected by differential expression screening and enriched for analysis. After univariate and multivariate Cox regression analyses, a new prognostic indicator based on immune-associated lncRNAs was established. Results Overall, differentially expressed immune-related lncRNAs were significantly associated with clinical outcomes in patients with melanoma. A prognostic model was then established based on 14 immune-associated lncRNAs (LRRC8C-DT, AC021188.1, MALINC1, CCR5AS, EIF2AK3-DT, AC022306.2, AC242842.1, AL034376.1, AL662844.4, AC009065.3, AC099811.3, AC125807.2, SPINT1-AS1 and AC009495.2). Melanoma patients in the high-risk group had worse overall survival than those in the low-risk group. The AUC of the risk score was 0.786. Conclusion This study identified several clinically significant immune-related lncRNAs and established a relevant prognostic model, which provided a molecular analysis of immunity in melanoma and potential prognostic lncRNAs for melanoma.
Collapse
Affiliation(s)
- Fang-Wei Li
- Department of Plastic and Reconstructive Surgery, Guangdong Second Provincial General Hospital, Guangzhou City, Guangdong Province, 510317, People's Republic of China
| | - Sheng-Kang Luo
- Department of Plastic and Reconstructive Surgery, Guangdong Second Provincial General Hospital, Guangzhou City, Guangdong Province, 510317, People's Republic of China
| |
Collapse
|
11
|
Zhang W, Xin J, Lai J, Zhang W. LncRNA LINC00184 promotes docetaxel resistance and immune escape via miR-105-5p/PD-L1 axis in prostate cancer. Immunobiology 2021; 227:152163. [PMID: 34896914 DOI: 10.1016/j.imbio.2021.152163] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2021] [Revised: 11/16/2021] [Accepted: 12/02/2021] [Indexed: 12/25/2022]
Abstract
BACKGROUND Docetaxel (DTX) resistance is a common factor in metastatic prostate cancer (PC) chemotherapy that leads to treatment failure. Because lncRNA is involved in a variety of regulatory processes in tumor progression, this study aimed to explore the function and mechanism of LINC00184 in docetaxel resistance of PC. METHODS Two PC cell lines and their docetaxel resistant cell lines (DU145/DTX and PC3/DTX) were used. The expression of LINC00184 in both cell lines and PC patient samples were evaluated. SiRNA knocking down was used to test the function of LINC00184 in proliferation and colony formation. Interaction between LINC00184 and its target miR-105-5p, as well as miR-105-5p and PD-L1 was checked by luciferase reporter assay and RNA pull-down assay. PC cell line and CD8 + T cell co-culture system was established, miR-105-5p inhibitor was co-transfected with LINC00184 siRNA to investigate the underline mechanism. RESULTS LINC00184 was found to be associated with docetaxel resistance and adverse prognosis of prostate cancer. It regulated docetaxel resistance and T-cell-mediated immune response in prostate cancer cells. LINC00184 was induced by adsorption of miR-105-5p and negatively regulated it, subsequently inhibited the expression level of PD-L1. CONCLUSIONS LINC00184 promoted docetaxel resistance and immune escape in prostate cancer cells by adsorption of miR-105-5p, resulted in upregulation of the expression of PD-L1. LINC00184 could possibly be considered as a potential target for treatment in prostate cancer patients with docetaxel-resistance.
Collapse
Affiliation(s)
- Wei Zhang
- Department of Urology, Quanzhou First Hospital Affiliated to Fujian Medical University, No. 248-252 East Street, Licheng District, Quanzhou 362000, People's Republic of China
| | - Jun Xin
- Department of Urology, Quanzhou First Hospital Affiliated to Fujian Medical University, No. 248-252 East Street, Licheng District, Quanzhou 362000, People's Republic of China
| | - Jinjin Lai
- Department of Urology, Quanzhou First Hospital Affiliated to Fujian Medical University, No. 248-252 East Street, Licheng District, Quanzhou 362000, People's Republic of China
| | - Wenbin Zhang
- Department of Urology, Quanzhou First Hospital Affiliated to Fujian Medical University, No. 248-252 East Street, Licheng District, Quanzhou 362000, People's Republic of China.
| |
Collapse
|
12
|
Xiao B, Liu L, Li A, Wang P, Xiang C, Li H, Xiao T. Identification and validation of immune-related lncRNA prognostic signatures for melanoma. IMMUNITY INFLAMMATION AND DISEASE 2021; 9:1044-1054. [PMID: 34077998 PMCID: PMC8342236 DOI: 10.1002/iid3.468] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/12/2021] [Accepted: 05/14/2021] [Indexed: 12/15/2022]
Abstract
Introduction Melanoma is a highly aggressive malignant skin tumor as well as the primary reason for skin cancer‐specific deaths. We first identified immune‐related long noncoding RNA (lncRNA) prognostic signature and found potential immunotherapeutic targets for melanoma cancer. Methods RNA‐seq data and clinical features of melanoma samples were obtained from The Cancer Genome Atlas. Samples of melanoma were randomly assigned to the training and testing cohort. The immune‐related lncRNA signature was then obtained via using univariate, LASSO, and multivariate Cox analysis of patients in the training cohort. Eight significant immune‐related lncRNA signature was then subsequently obtained through correlation analysis between immune‐related genes and lncRNAs. The association between risk score and immune cell infiltration was finally assessed using TIMER and CIBERSORT. Results Three hundred and fifty‐six immune‐related lncRNAs were obtained. Among them, eight immune‐related lncRNAs were identified to build a prognostic risk signature model. The model's performance was then confirmed using the Kaplan–Meier curves, risk plots, and time‐dependent receiver‐operating characteristic curves in the training cohort. The risk score was identified and confirmed as an independent prognostic factor through univariate and multivariate Cox regression analyses. These results were further verified in the testing and whole cohorts. CIBERSORT algorithm showed that the infiltration levels of T cells CD8, M1 macrophages, plasma cells, T cells CD4 memory activated, T cells gamma delta, and mast cells activated were significantly lower in the high‐risk group while the infiltration level of macrophages M0 was significantly lower in the low‐risk group. Conclusion The immune‐related lncRNA signature offers prognostic markers and potential immunotherapeutic targets for melanoma.
Collapse
Affiliation(s)
- Bo Xiao
- Department of OrthopedicsThe Second Xiangya Hospital of Central South UniversityChangshaHunanChina
| | - Liyan Liu
- Department of OrthopedicsThe Second Xiangya Hospital of Central South UniversityChangshaHunanChina
| | - Aoyu Li
- Department of OrthopedicsThe Second Xiangya Hospital of Central South UniversityChangshaHunanChina
| | - Pingxiao Wang
- Department of OrthopedicsThe Second Xiangya Hospital of Central South UniversityChangshaHunanChina
| | - Cheng Xiang
- Department of OrthopedicsThe Second Xiangya Hospital of Central South UniversityChangshaHunanChina
| | - Hui Li
- Department of OrthopedicsThe Second Xiangya Hospital of Central South UniversityChangshaHunanChina
| | - Tao Xiao
- Department of OrthopedicsThe Second Xiangya Hospital of Central South UniversityChangshaHunanChina
| |
Collapse
|