1
|
Yang Y, Zhang B, Xu Y, Zhu W, Zhu Z, Zhang X, Wu W, Chen J, Yu Z. An immunotherapeutic hydrogel booster inhibits tumor recurrence and promotes wound healing for postoperative management of melanoma. Bioact Mater 2024; 42:178-193. [PMID: 39285910 PMCID: PMC11402546 DOI: 10.1016/j.bioactmat.2024.08.028] [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: 01/24/2024] [Revised: 08/07/2024] [Accepted: 08/26/2024] [Indexed: 09/19/2024] Open
Abstract
Low tumor immunogenicity, immunosuppressive tumor microenvironment, and bacterial infections have emerged as significant challenges in postsurgical immunotherapy and skin regeneration for preventing melanoma recurrence. Herein, an immunotherapeutic hydrogel booster (GelMA-CJCNPs) was developed to prevent postoperative tumor recurrence and promote wound healing by incorporating ternary carrier-free nanoparticles (CJCNPs) containing chlorine e6 (Ce6), a BRD4 inhibitor (JQ1), and a glutaminase inhibitor (C968) into methacrylic anhydride-modified gelatin (GelMA) dressings. GelMA-CJCNPs reduced glutathione production by inhibiting glutamine metabolism, thereby preventing the destruction of reactive oxygen species generated by photodynamic therapy, which could amplify oxidative stress to induce severe cell death and enhance immunogenic cell death. In addition, GelMA-CJCNPs reduced M2-type tumor-associated macrophage polarization by blocking glutamine metabolism to reverse the immunosuppressive tumor microenvironment, recruiting more tumor-infiltrating T lymphocytes. GelMA-CJCNPs also downregulated IFN-γ-induced expression of programmed cell death ligand 1 to mitigate acquired immune resistance. Benefiting from the amplified systemic antitumor immunity, GelMA-CJCNPs markedly inhibited the growth of both primary and distant tumors. Moreover, GelMA-CJCNPs demonstrated satisfactory photodynamic antibacterial effects against Staphylococcus aureus infections, thereby promoting postsurgical wound healing. Hence, this immunotherapeutic hydrogel booster, as a facile and effective postoperative adjuvant, possesses a promising potential for inhibiting tumor recurrence and accelerating skin regeneration.
Collapse
Affiliation(s)
- Yuanyuan Yang
- Department of Laboratory Medicine, Dongguan Institute of Clinical Cancer Research, Dongguan Key Laboratory of Precision Diagnosis and Treatment for Tumors, The Tenth Affiliated Hospital of Southern Medical University (Dongguan People's Hospital), Dongguan, 523058, China
| | - Bo Zhang
- Jingzhou Central Hospital, Jingzhou Hospital Affiliated to Yangtze University, Jingzhou, 434020, China
| | - Yangtao Xu
- Department of Laboratory Medicine, Dongguan Institute of Clinical Cancer Research, Dongguan Key Laboratory of Precision Diagnosis and Treatment for Tumors, The Tenth Affiliated Hospital of Southern Medical University (Dongguan People's Hospital), Dongguan, 523058, China
- Institute of Biomedical Health Technology and Engineering, Shenzhen Bay Laboratory, Shenzhen, 518132, China
| | - Wenxiang Zhu
- Department of Laboratory Medicine, Dongguan Institute of Clinical Cancer Research, Dongguan Key Laboratory of Precision Diagnosis and Treatment for Tumors, The Tenth Affiliated Hospital of Southern Medical University (Dongguan People's Hospital), Dongguan, 523058, China
- Institute of Biomedical Health Technology and Engineering, Shenzhen Bay Laboratory, Shenzhen, 518132, China
| | - Zinian Zhu
- Department of Laboratory Medicine, Dongguan Institute of Clinical Cancer Research, Dongguan Key Laboratory of Precision Diagnosis and Treatment for Tumors, The Tenth Affiliated Hospital of Southern Medical University (Dongguan People's Hospital), Dongguan, 523058, China
| | - Xibo Zhang
- Department of Laboratory Medicine, Dongguan Institute of Clinical Cancer Research, Dongguan Key Laboratory of Precision Diagnosis and Treatment for Tumors, The Tenth Affiliated Hospital of Southern Medical University (Dongguan People's Hospital), Dongguan, 523058, China
| | - Wenze Wu
- Jingzhou Central Hospital, Jingzhou Hospital Affiliated to Yangtze University, Jingzhou, 434020, China
| | - Jierong Chen
- Laboratory Medicine, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, 510080, China
| | - Zhiqiang Yu
- Department of Laboratory Medicine, Dongguan Institute of Clinical Cancer Research, Dongguan Key Laboratory of Precision Diagnosis and Treatment for Tumors, The Tenth Affiliated Hospital of Southern Medical University (Dongguan People's Hospital), Dongguan, 523058, China
| |
Collapse
|
2
|
Hong P, Hu Z, Lin J, Cui K, Gao Z, Tian X, Shi Q, Lin T, Wei G. Multi-omics revealed that ELAVL3 regulates MYCN in neuroblastoma via immunogenic cell death: Risk stratification and experimental research. Int J Biol Macromol 2024; 282:137045. [PMID: 39486730 DOI: 10.1016/j.ijbiomac.2024.137045] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2024] [Revised: 10/12/2024] [Accepted: 10/28/2024] [Indexed: 11/04/2024]
Abstract
Neuroblastoma (NB), a common and highly lethal malignant disease in pediatrics, still lacks an effective therapeutic approach that addresses all conditions. Immunogenic Cell Death (ICD) plays a crucial role in tumor cell death and triggers a potent anti-tumor immune response. In this study, we report an ICD-related index (ICDR-Index) in NB through various machine learning methodologies, utilizing bulk transcriptome data from 1244 NB samples and 16 scRNA-seq datasets. Our results showed that the ICDR-Index could accurately identify different risk subtypes of patients with NB and provide predictive value for prognosis. Importantly, we found that high-risk patients with NB exhibited significantly poor overall survival (OS) rates, adverse clinical phenotypes, poor immune cell infiltration, and low sensitivity to immunotherapy. Furthermore, we identified ELAVL3 as a key gene within the ICDR-Index, where high expression levels were associated with malignancy and poor OS in NB. Additionally, targeted silencing of ELAVL3 down-regulated MYCN gene expression and reduced the malignancy of NB cells. Notably, the si-ELAVL3-transfected NB cells enhanced the anti-tumor activity of NK cells. Collectively, this study offers avenues for predicting the risk stratification of patients with NB and reveals a potential mechanism by which ELAVL3 regulates NB cell death.
Collapse
Affiliation(s)
- Peng Hong
- Department of Urology, Children's Hospital of Chongqing Medical University; National Clinical Research Center for Child Health and Disorders; Ministry of Education Key Laboratory of Child Development and Disorders; Chongqing Key Laboratory of Structural Birth Defect and Reconstruction, China
| | - Zaihong Hu
- Department of Urology, Children's Hospital of Chongqing Medical University; National Clinical Research Center for Child Health and Disorders; Ministry of Education Key Laboratory of Child Development and Disorders; Chongqing Key Laboratory of Structural Birth Defect and Reconstruction, China
| | - Jie Lin
- Department of Urology, Children's Hospital of Chongqing Medical University; National Clinical Research Center for Child Health and Disorders; Ministry of Education Key Laboratory of Child Development and Disorders; Chongqing Key Laboratory of Structural Birth Defect and Reconstruction, China
| | - Kongkong Cui
- Department of Urology, Children's Hospital of Chongqing Medical University; National Clinical Research Center for Child Health and Disorders; Ministry of Education Key Laboratory of Child Development and Disorders; Chongqing Key Laboratory of Structural Birth Defect and Reconstruction, China
| | - Zhiqiang Gao
- Department of Urology, Children's Hospital of Chongqing Medical University; National Clinical Research Center for Child Health and Disorders; Ministry of Education Key Laboratory of Child Development and Disorders; Chongqing Key Laboratory of Structural Birth Defect and Reconstruction, China
| | - Xiaomao Tian
- Department of Urology, Children's Hospital of Chongqing Medical University; National Clinical Research Center for Child Health and Disorders; Ministry of Education Key Laboratory of Child Development and Disorders; Chongqing Key Laboratory of Structural Birth Defect and Reconstruction, China
| | - Qinlin Shi
- Department of Urology, Children's Hospital of Chongqing Medical University; National Clinical Research Center for Child Health and Disorders; Ministry of Education Key Laboratory of Child Development and Disorders; Chongqing Key Laboratory of Structural Birth Defect and Reconstruction, China.
| | - Tao Lin
- Department of Urology, Children's Hospital of Chongqing Medical University; National Clinical Research Center for Child Health and Disorders; Ministry of Education Key Laboratory of Child Development and Disorders; Chongqing Key Laboratory of Structural Birth Defect and Reconstruction, China
| | - Guanghui Wei
- Department of Urology, Children's Hospital of Chongqing Medical University; National Clinical Research Center for Child Health and Disorders; Ministry of Education Key Laboratory of Child Development and Disorders; Chongqing Key Laboratory of Structural Birth Defect and Reconstruction, China
| |
Collapse
|
3
|
Gong Z, Zhou D, Shen H, Ma C, Wu D, Hou L, Wang H, Xu T. Development of a prognostic model related to homologous recombination deficiency in glioma based on multiple machine learning. Front Immunol 2024; 15:1452097. [PMID: 39434883 PMCID: PMC11491349 DOI: 10.3389/fimmu.2024.1452097] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2024] [Accepted: 09/13/2024] [Indexed: 10/23/2024] Open
Abstract
Background Despite advances in neuro-oncology, treatments of glioma and tools for predicting the outcome of patients remain limited. The objective of this research is to construct a prognostic model for glioma using the Homologous Recombination Deficiency (HRD) score and validate its predictive capability for glioma. Methods We consolidated glioma datasets from TCGA, various cancer types for pan-cancer HRD analysis, and two additional glioma RNAseq datasets from GEO and CGGA databases. HRD scores, mutation data, and other genomic indices were calculated. Using machine learning algorithms, we identified signature genes and constructed an HRD-related prognostic risk model. The model's performance was validated across multiple cohorts. We also assessed immune infiltration and conducted molecular docking to identify potential therapeutic agents. Results Our analysis established a correlation between higher HRD scores and genomic instability in gliomas. The model, based on machine learning algorithms, identified seven key genes, significantly predicting patient prognosis. Moreover, the HRD score prognostic model surpassed other models in terms of prediction efficacy across different cancers. Differential immune cell infiltration patterns were observed between HRD risk groups, with potential implications for immunotherapy. Molecular docking highlighted several compounds, notably Panobinostat, as promising for high-risk patients. Conclusions The prognostic model based on the HRD score threshold and associated genes in glioma offers new insights into the genomic and immunological landscapes, potentially guiding therapeutic strategies. The differential immune profiles associated with HRD-risk groups could inform immunotherapeutic interventions, with our findings paving the way for personalized medicine in glioma treatment.
Collapse
Affiliation(s)
- Zhenyu Gong
- Department of Neurosurgery, Changzheng Hospital, Naval Medical University, Shanghai, China
- Department of Neurosurgery, Klinikum rechts der Isar, Technical University of Munich, Munich, Germany
| | - Dairan Zhou
- Department of Neurosurgery, Changzheng Hospital, Naval Medical University, Shanghai, China
| | - Haotian Shen
- Department of Neurosurgery, Changzheng Hospital, Naval Medical University, Shanghai, China
| | - Chao Ma
- Department of Neurosurgery, The Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
| | - Dejun Wu
- Department of Neurosurgery, The Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
| | - Lijun Hou
- Department of Neurosurgery, Changzheng Hospital, Naval Medical University, Shanghai, China
| | - Hongxiang Wang
- Department of Neurosurgery, Changhai Hospital, Naval Medical University, Shanghai, China
| | - Tao Xu
- Department of Neurosurgery, Changzheng Hospital, Naval Medical University, Shanghai, China
| |
Collapse
|
4
|
Dai SL, Pan JQ, Su ZR. Multi-omics features of immunogenic cell death in gastric cancer identified by combining single-cell sequencing analysis and machine learning. Sci Rep 2024; 14:21751. [PMID: 39294296 PMCID: PMC11410816 DOI: 10.1038/s41598-024-73071-x] [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: 07/22/2024] [Accepted: 09/13/2024] [Indexed: 09/20/2024] Open
Abstract
Gastric cancer (GC) is a prevalent malignancy with high mortality rates. Immunogenic cell death (ICD) is a unique form of programmed cell death that is closely linked to antitumor immunity and plays a critical role in modulating the tumor microenvironment (TME). Nevertheless, elucidating the precise effect of ICD on GC remains a challenging endeavour. ICD-related genes were identified in single-cell sequencing datasets and bulk transcriptome sequencing datasets via the AddModuleScore function, weighted gene co-expression network (WGCNA), and differential expression analysis. A robust signature associated with ICD was constructed using a machine learning computational framework incorporating 101 algorithms. Furthermore, multiomics analysis, including single-cell sequencing analysis, bulk transcriptomic analysis, and proteomics analysis, was conducted to verify the correlation of these hub genes with the immune microenvironment features of GC and with GC invasion and metastasis. We screened 59 genes associated with ICD and developed a robust ICD-related gene signature (ICDRS) via a machine learning computational framework that integrates 101 different algorithms. Furthermore, we identified five key hub genes (SMAP2, TNFAIP8, LBH, TXNIP, and PIK3IP1) from the ICDRS. Through single-cell analysis of GC tumor s, we confirmed the strong correlations of the hub genes with immune microenvironment features. Among these five genes, LBH exhibited the most significant associations with a poor prognosis and with the invasion and metastasis of GC. Finally, our findings were validated through immunohistochemical staining of a large clinical sample set, and the results further supported that LBH promotes GC cell invasion by activating the epithelial-mesenchymal transition (EMT) pathway.
Collapse
Affiliation(s)
- Shu-Long Dai
- Department of General Surgery, Deqing People's Hospital, Deqing Campus, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, 120 South Yingxi Road, Deqing, 313200, Zhejiang, P. R. China.
| | - Jian-Qiang Pan
- Department of Pathology, Deqing People's Hospital, Deqing Campus, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, 120 South Yingxi Road, Deqing, 313200, Zhejiang, P. R. China
| | - Zhen-Rong Su
- Department of General Surgery, Deqing People's Hospital, Deqing Campus, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, 120 South Yingxi Road, Deqing, 313200, Zhejiang, P. R. China
| |
Collapse
|
5
|
Qin X, Chen Z, Wu L, Ding R. A disulfidptosis-associated long noncoding RNA signature to predict low-grade glioma classification, prognosis, tumor microenvironment, and therapy regimens: Observational study. Medicine (Baltimore) 2024; 103:e39316. [PMID: 39183405 PMCID: PMC11346906 DOI: 10.1097/md.0000000000039316] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/18/2023] [Revised: 04/23/2024] [Accepted: 07/25/2024] [Indexed: 08/27/2024] Open
Abstract
This study aimed to investigate the function of disulfidptosis-associated long noncoding RNAs (DAlncRNAs) in low-grade gliomas (LGG) through bioinformatics analysis and construct a signature to predict the classification, prognosis, tumor microenvironment, and selection of immunotherapy and chemotherapy in LGG. Genomic, clinical, and mutational information of 526 patients with LGG was retrieved from The Cancer Genome Atlas repository. A nonnegative matrix factorization algorithm was applied to classify patients with LGG. Univariate, LASSO regression, and multivariate Cox regression analyses were performed to determine prognostic DAlncRNAs. Following the median risk score, we defined the sample as a high-risk (HR) or low-risk group. Finally, survival, receiver operating characteristic curve, risk curve, principal component, independent prognosis, risk difference, functional enrichment, tumor microenvironment, immune cell infiltration, mutation, and drug sensitivity analyses were performed. Patients were classified into C1 and C2 subtypes associated with disulfidptosis. Eight prognostic DAlncRNAs (AC003035.2, AC010157.2, AC010273.3, AC011444.3, AC092667.1, AL450270.1, AL645608.2, and LINC01571) were identified, and a prognostic signature of LGG was developed. The DAlncRNA-based signature was found to be an independent prognostic factor in patients with LGG, thereby constructing a nomogram. In addition, in the HR group, immune function was more active and the tumor mutation burden was higher. The patients were mainly composed of subtype C2, and their prognosis was worse. Immunotherapy and chemotherapy were predicted in the HR and low-risk groups, respectively. Our study, based on DAlncRNAs, highlights 2 disulfidptosis-associated LGG subtypes with different prognostic and immune characteristics and creates a novel disulfidptosis-associated prognostic signature, which may inform the classification, prognosis, molecular pathogenesis, and therapeutic strategies for patients with LGG.
Collapse
Affiliation(s)
- Xiaohong Qin
- Department of Neurosurgery, Renmin Hospital of Wuhan University, Wuhan, Hubei, China
| | - Zhibiao Chen
- Department of Neurosurgery, Renmin Hospital of Wuhan University, Wuhan, Hubei, China
| | - Liquan Wu
- Department of Neurosurgery, Renmin Hospital of Wuhan University, Wuhan, Hubei, China
| | - Rui Ding
- Department of Neurosurgery, Renmin Hospital of Wuhan University, Wuhan, Hubei, China
| |
Collapse
|
6
|
Luo M, Luan X, Yang C, Chen X, Yuan S, Cao Y, Zhang J, Xie J, Luo Q, Chen L, Li S, Xiang W, Zhou J. Revisiting the potential of regulated cell death in glioma treatment: a focus on autophagy-dependent cell death, anoikis, ferroptosis, cuproptosis, pyroptosis, immunogenic cell death, and the crosstalk between them. Front Oncol 2024; 14:1397863. [PMID: 39184045 PMCID: PMC11341384 DOI: 10.3389/fonc.2024.1397863] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2024] [Accepted: 07/22/2024] [Indexed: 08/27/2024] Open
Abstract
Gliomas are primary tumors that originate in the central nervous system. The conventional treatment options for gliomas typically encompass surgical resection and temozolomide (TMZ) chemotherapy. However, despite aggressive interventions, the median survival for glioma patients is merely about 14.6 months. Consequently, there is an urgent necessity to explore innovative therapeutic strategies for treating glioma. The foundational study of regulated cell death (RCD) can be traced back to Karl Vogt's seminal observations of cellular demise in toads, which were documented in 1842. In the past decade, the Nomenclature Committee on Cell Death (NCCD) has systematically classified and delineated various forms and mechanisms of cell death, synthesizing morphological, biochemical, and functional characteristics. Cell death primarily manifests in two forms: accidental cell death (ACD), which is caused by external factors such as physical, chemical, or mechanical disruptions; and RCD, a gene-directed intrinsic process that coordinates an orderly cellular demise in response to both physiological and pathological cues. Advancements in our understanding of RCD have shed light on the manipulation of cell death modulation - either through induction or suppression - as a potentially groundbreaking approach in oncology, holding significant promise. However, obstacles persist at the interface of research and clinical application, with significant impediments encountered in translating to therapeutic modalities. It is increasingly apparent that an integrative examination of the molecular underpinnings of cell death is imperative for advancing the field, particularly within the framework of inter-pathway functional synergy. In this review, we provide an overview of various forms of RCD, including autophagy-dependent cell death, anoikis, ferroptosis, cuproptosis, pyroptosis and immunogenic cell death. We summarize the latest advancements in understanding the molecular mechanisms that regulate RCD in glioma and explore the interconnections between different cell death processes. By comprehending these connections and developing targeted strategies, we have the potential to enhance glioma therapy through manipulation of RCD.
Collapse
Affiliation(s)
- Maowen Luo
- Department of Neurosurgery, the Affiliated Hospital, Southwest Medical University, Luzhou, Sichuan, China
- School of Clinical Medicine, Southwest Medical University, Luzhou, Sichuan, China
| | - Xingzhao Luan
- Department of Neurosurgery, the Affiliated Hospital of Panzhihua University, Panzhihua, Sichuan, China
- School of Clinical Medicine, the Affiliated Hospital of Panzhihua University, Panzhihua, Sichuan, China
| | - Chaoge Yang
- Department of Neurosurgery, the Affiliated Hospital, Southwest Medical University, Luzhou, Sichuan, China
- School of Clinical Medicine, Southwest Medical University, Luzhou, Sichuan, China
- School of Clinical Medicine, Sichuan Clinical Research Center for Neurosurgery, Luzhou, Sichuan, China
| | - Xiaofan Chen
- Department of Neurosurgery, the Affiliated Hospital, Southwest Medical University, Luzhou, Sichuan, China
- School of Clinical Medicine, Southwest Medical University, Luzhou, Sichuan, China
| | - Suxin Yuan
- School of Clinical Medicine, the Affiliated Hospital of Panzhihua University, Panzhihua, Sichuan, China
| | - Youlin Cao
- Department of Neurosurgery, the Affiliated Hospital of Panzhihua University, Panzhihua, Sichuan, China
- School of Clinical Medicine, the Affiliated Hospital of Panzhihua University, Panzhihua, Sichuan, China
| | - Jing Zhang
- School of Clinical Medicine, the Affiliated Hospital of Panzhihua University, Panzhihua, Sichuan, China
| | - Jiaying Xie
- Department of Neurosurgery, the Affiliated Hospital, Southwest Medical University, Luzhou, Sichuan, China
- School of Clinical Medicine, Southwest Medical University, Luzhou, Sichuan, China
| | - Qinglian Luo
- Department of Neurosurgery, the Affiliated Hospital, Southwest Medical University, Luzhou, Sichuan, China
- School of Clinical Medicine, Southwest Medical University, Luzhou, Sichuan, China
- School of Clinical Medicine, Sichuan Clinical Research Center for Neurosurgery, Luzhou, Sichuan, China
| | - Ligang Chen
- Department of Neurosurgery, the Affiliated Hospital, Southwest Medical University, Luzhou, Sichuan, China
- School of Clinical Medicine, Southwest Medical University, Luzhou, Sichuan, China
- School of Clinical Medicine, Sichuan Clinical Research Center for Neurosurgery, Luzhou, Sichuan, China
| | - Shenjie Li
- Department of Neurosurgery, the Affiliated Hospital, Southwest Medical University, Luzhou, Sichuan, China
- School of Clinical Medicine, Southwest Medical University, Luzhou, Sichuan, China
- School of Clinical Medicine, Sichuan Clinical Research Center for Neurosurgery, Luzhou, Sichuan, China
| | - Wei Xiang
- Department of Neurosurgery, the Affiliated Hospital, Southwest Medical University, Luzhou, Sichuan, China
- School of Clinical Medicine, Southwest Medical University, Luzhou, Sichuan, China
- School of Clinical Medicine, Sichuan Clinical Research Center for Neurosurgery, Luzhou, Sichuan, China
| | - Jie Zhou
- Department of Neurosurgery, the Affiliated Hospital, Southwest Medical University, Luzhou, Sichuan, China
- School of Clinical Medicine, Southwest Medical University, Luzhou, Sichuan, China
- School of Clinical Medicine, Sichuan Clinical Research Center for Neurosurgery, Luzhou, Sichuan, China
| |
Collapse
|
7
|
Li MP, Long SP, Liu WC, Long K, Gao XH. EMT-related gene classifications predict the prognosis, immune infiltration, and therapeutic response of osteosarcoma. Front Pharmacol 2024; 15:1419040. [PMID: 39170698 PMCID: PMC11335561 DOI: 10.3389/fphar.2024.1419040] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2024] [Accepted: 07/26/2024] [Indexed: 08/23/2024] Open
Abstract
Background Osteosarcoma (OS), a bone tumor with high ability of invasion and metastasis, has seriously affected the health of children and adolescents. Many studies have suggested a connection between OS and the epithelial-mesenchymal transition (EMT). We aimed to integrate EMT-Related genes (EMT-RGs) to predict the prognosis, immune infiltration, and therapeutic response of patients with OS. Methods We used consensus clustering to identify potential EMT-Related OS molecular subtypes. Somatic mutation, tumor immune microenvironment, and functional enrichment analyses were performed for each subtype. We next constructed an EMT-Related risk signature and evaluated it by Kaplan-Meier (K-M) analysis survival and receiver operating characteristic (ROC) curves. Moreover, we constructed a nomogram to more accurately predict OS patients' clinical outcomes. Response effects of immunotherapy in OS patients was analyzed by Tumor Immune Dysfunction and Exclusion (TIDE) analysis, while sensitivity for chemotherapeutic agents was analyzed using oncoPredict. Finally, the expression patterns of hub genes were investigated by single-cell RNA sequencing (scRNA-seq) data analysis. Results A total of 53 EMT-RDGs related to prognosis were identified, separating OS samples into two separate subgroups. The EMT-high subgroup showed favourable overall survival and more active immune response. Significant correlations were found between EMT-Related DEGs and functions as well as pathways linked to the development of OS. Additionally, a risk signature was established and OS patients were divided into two categories based on the risk scores. The signature presented a good predictive performance and could be recognized as an independent predictive factor for OS. Furthermore, patients with higher risk scores exhibited better sensitivity for five drugs, while no significant difference existed in immunotherapy response between the two risk subgroups. scRNA-seq data analysis displayed different expression patterns of the hub genes. Conclusion We developed a novel EMT-Related risk signature that can be considered as an independent predictor for OS, which may help improve clinical outcome prediction and guide personalized treatments for patients with OS.
Collapse
Affiliation(s)
- Meng-Pan Li
- Department of Orthopedics, Guangzhou First People’s Hospital, South China University of Technology, Guangzhou, China
- Department of Orthopedics, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- The First Clinical Medical College of Nanchang University, Nanchang, China
| | - Si-Ping Long
- The Fourth Clinical Medical College of Nanchang University, Nanchang, China
| | - Wen-Cai Liu
- Department of Orthopedics, Shanghai Sixth People’s Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Kun Long
- The First Clinical Medical College of Nanchang University, Nanchang, China
| | - Xing-Hua Gao
- Department of Orthopedics, Guangzhou First People’s Hospital, South China University of Technology, Guangzhou, China
| |
Collapse
|
8
|
Gulla A, Morelli E, Johnstone M, Turi M, Samur MK, Botta C, Cifric S, Folino P, Vinaixa D, Barello F, Clericuzio C, Favasuli VK, Maisano D, Talluri S, Prabhala R, Bianchi G, Fulciniti M, Wen K, Kurata K, Liu J, Penailillo J, Bragoni A, Sapino A, Richardson PG, Chauhan D, Carrasco RD, Hideshima T, Munshi NC, Anderson KC. Loss of GABARAP mediates resistance to immunogenic chemotherapy in multiple myeloma. Blood 2024; 143:2612-2626. [PMID: 38551812 DOI: 10.1182/blood.2023022777] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2023] [Accepted: 03/16/2024] [Indexed: 06/21/2024] Open
Abstract
ABSTRACT Immunogenic cell death (ICD) is a form of cell death by which cancer treatments can induce a clinically relevant antitumor immune response in a broad range of cancers. In multiple myeloma (MM), the proteasome inhibitor bortezomib is an ICD inducer and creates durable therapeutic responses in patients. However, eventual relapse and resistance to bortezomib appear inevitable. Here, by integrating patient transcriptomic data with an analysis of calreticulin (CRT) protein interactors, we found that GABA type A receptor-associated protein (GABARAP) is a key player whose loss prevented tumor cell death from being perceived as immunogenic after bortezomib treatment. GABARAP is located on chromosome 17p, which is commonly deleted in patients with high risk MM. GABARAP deletion impaired the exposure of the eat-me signal CRT on the surface of dying MM cells in vitro and in vivo, thus reducing tumor cell phagocytosis by dendritic cells and the subsequent antitumor T-cell response. Low GABARAP was independently associated with shorter survival in patients with MM and reduced tumor immune infiltration. Mechanistically, we found that GABARAP deletion blocked ICD signaling by decreasing autophagy and altering Golgi apparatus morphology, with consequent defects in the downstream vesicular transport of CRT. Conversely, upregulating autophagy using rapamycin restored Golgi morphology, CRT exposure, and ICD signaling in GABARAPKO cells undergoing bortezomib treatment. Therefore, coupling an ICD inducer, such as bortezomib, with an autophagy inducer, such as rapamycin, may improve patient outcomes in MM, in which low GABARAP in the form of del(17p) is common and leads to worse outcomes.
Collapse
Affiliation(s)
- Annamaria Gulla
- Department of Medical Oncology, Candiolo Cancer Institute, Fondazione del Piemonte per l'Oncologia-IRCCS, Candiolo, Italy
| | - Eugenio Morelli
- Department of Medical Oncology, Candiolo Cancer Institute, Fondazione del Piemonte per l'Oncologia-IRCCS, Candiolo, Italy
| | - Megan Johnstone
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA
| | - Marcello Turi
- Department of Medical Oncology, Candiolo Cancer Institute, Fondazione del Piemonte per l'Oncologia-IRCCS, Candiolo, Italy
| | - Mehmet K Samur
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA
- Department of Data Sciences, Dana-Farber Cancer Institute, Boston, MA
- Department of Biostatistics, Harvard T.H. Chan School of Public Health, Boston, MA
| | - Cirino Botta
- Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialties, University of Palermo, Palermo, Italy
| | - Selma Cifric
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA
| | - Pietro Folino
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA
| | - Delaney Vinaixa
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA
- Northeastern University, Boston, MA
| | - Francesca Barello
- Department of Medical Oncology, Candiolo Cancer Institute, Fondazione del Piemonte per l'Oncologia-IRCCS, Candiolo, Italy
| | - Cole Clericuzio
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA
- Northeastern University, Boston, MA
| | - Vanessa Katia Favasuli
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA
| | - Domenico Maisano
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA
| | - Srikanth Talluri
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA
- VA Boston Healthcare System, Boston, MA
| | - Rao Prabhala
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA
- VA Boston Healthcare System, Boston, MA
| | - Giada Bianchi
- Division of Hematology, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA
| | - Mariateresa Fulciniti
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA
| | - Kenneth Wen
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA
| | - Keiji Kurata
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA
- Division of Medical Oncology/Hematology, Department of Medicine, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Jiye Liu
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA
| | - Johany Penailillo
- Department of Oncologic Pathology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA
| | - Alberto Bragoni
- Department of Medical Oncology, Candiolo Cancer Institute, Fondazione del Piemonte per l'Oncologia-IRCCS, Candiolo, Italy
- Department of Medical Sciences, University of Turin, Turin, Italy
| | - Anna Sapino
- Department of Medical Oncology, Candiolo Cancer Institute, Fondazione del Piemonte per l'Oncologia-IRCCS, Candiolo, Italy
- Department of Medical Sciences, University of Turin, Turin, Italy
| | - Paul G Richardson
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA
| | - Dharminder Chauhan
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA
| | - Ruben D Carrasco
- Department of Oncologic Pathology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA
- Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA
| | - Teru Hideshima
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA
| | - Nikhil C Munshi
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA
- VA Boston Healthcare System, Boston, MA
| | - Kenneth C Anderson
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA
| |
Collapse
|
9
|
Meng LB, Li Y, Lv T, Lv C, Liu L, Zhang P. Joint effects of CD8A and ICOS in Long QT Syndrome (LQTS) and Beckwith-Wiedemann Syndrome (BWS). J Cardiothorac Surg 2024; 19:321. [PMID: 38845009 PMCID: PMC11155187 DOI: 10.1186/s13019-024-02804-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2024] [Accepted: 05/25/2024] [Indexed: 06/10/2024] Open
Abstract
BACKGROUND Long QT Syndrome (LQTS) and Beckwith-Wiedemann Syndrome (BWS) are complex disorders with unclear origins, underscoring the need for in-depth molecular investigations into their mechanisms. The main aim of this study is to identify the shared key genes between LQTS and BWS, shedding light on potential common molecular pathways underlying these syndromes. METHODS The LQTS and BWS datasets are available for download from the GEO database. Differential expression genes (DEGs) were identified. Weighted gene co-expression network analysis (WGCNA) was used to detect significant modules and central genes. Gene enrichment analysis was performed. CIBERSORT was used for immune cell infiltration analysis. The predictive protein interaction (PPI) network of core genes was constructed using STRING, and miRNAs regulating central genes were screened using TargetScan. RESULTS Five hundred DEGs associated with Long QT Syndrome and Beckwith-Wiedemann Syndrome were identified. GSEA analysis revealed enrichment in pathways such as T cell receptor signaling, MAPK signaling, and adrenergic signaling in cardiac myocytes. Immune cell infiltration indicated higher levels of memory B cells and naive CD4 T cells. Four core genes (CD8A, ICOS, CTLA4, LCK) were identified, with CD8A and ICOS showing low expression in the syndromes and high expression in normal samples, suggesting potential inverse regulatory roles. CONCLUSION The expression of CD8A and ICOS is low in long QT syndrome and Beckwith-Wiedemann syndrome, indicating their potential as key genes in the pathogenesis of these syndromes. The identification of shared key genes between LQTS and BWS provides insights into common molecular mechanisms underlying these disorders, potentially facilitating the development of targeted therapeutic strategies.
Collapse
Affiliation(s)
- Ling-Bing Meng
- Department of Cardiology, Beijing Tsinghua Changgung Hospital, School of Clinical Medicine, Tsinghua University, Beijing, China
| | - Yongchao Li
- Department of Cardiac Surgery, Beijing Tsinghua Changgung Hospital, School of Clinical Medicine, Tsinghua University, Beijing, China
| | - Tingting Lv
- Department of Cardiology, Beijing Tsinghua Changgung Hospital, School of Clinical Medicine, Tsinghua University, Beijing, China
| | - Changhua Lv
- Department of Cardiology, Beijing Tsinghua Changgung Hospital, School of Clinical Medicine, Tsinghua University, Beijing, China
| | - Lianfeng Liu
- Department of Cardiology, Beijing Tsinghua Changgung Hospital, School of Clinical Medicine, Tsinghua University, Beijing, China
| | - Ping Zhang
- Department of Cardiology, Beijing Tsinghua Changgung Hospital, School of Clinical Medicine, Tsinghua University, Beijing, China.
| |
Collapse
|
10
|
Yu M, Huo D, Yu K, Zhou K, Xu F, Meng Q, Cai Y, Chen X. Crosstalk of different cell-death patterns predicts prognosis and drug sensitivity in glioma. Comput Biol Med 2024; 175:108532. [PMID: 38703547 DOI: 10.1016/j.compbiomed.2024.108532] [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: 02/27/2024] [Revised: 04/17/2024] [Accepted: 04/28/2024] [Indexed: 05/06/2024]
Abstract
BACKGROUND Glioma is a malignant brain tumor originating from glial cells, and there still a challenge to accurately predict the prognosis. Programmed cell death (PCD) plays a key role in tumorigenesis and immune response. However, the crosstalk and potential role of various PCDs in prognosis and tumor microenvironment remains unknown. Therefore, we comprehensively discussed the relationship between different models of PCD and the prognosis of glioma and provided new ideas for the optimal targeted therapy of glioma. MATERIALS AND METHODS We compared and analyzed the role of 14 PCD patterns on the prognosis from different levels. We constructed the cell death risk score (CDRS) index and conducted a comprehensive analysis of CDRS and TME characteristics, clinical characteristics, and drug response. RESULTS Effects of different PCDs at the genomic, functional, and immune microenvironment levels were discussed. CDRS index containing 6 gene signatures and a nomogram were established. High CDRS is associated with a worse prognosis. Through transcriptome and single-cell data, we found that patients with high CDRS showed stronger immunosuppressive characteristics. Moreover, the high-CDRS group was resistant to the traditional glioma chemotherapy drug Vincristine, but more sensitive to the Temozolomide and the clinical experimental drug Bortezomib. In addition, we identified 19 key potential therapeutic targets during malignant differentiation of tumor cells. CONCLUSION Overall, we provide the first systematic description of the role of 14 PCDs in glioma. A new CDRS model was built to predict the prognosis and to provide a new idea for the targeted therapy of glioma.
Collapse
Affiliation(s)
- Meini Yu
- Department of pharmacogenomics, College of Bioinformatics Science and Technology, Harbin Medical University, 150081, Harbin, Heilongjiang, China
| | - Diwei Huo
- Fourth Affiliated Hospital of Harbin Medical University, China
| | - Kexin Yu
- Department of pharmacogenomics, College of Bioinformatics Science and Technology, Harbin Medical University, 150081, Harbin, Heilongjiang, China
| | - Kun Zhou
- Department of pharmacogenomics, College of Bioinformatics Science and Technology, Harbin Medical University, 150081, Harbin, Heilongjiang, China
| | - Fei Xu
- Department of pharmacogenomics, College of Bioinformatics Science and Technology, Harbin Medical University, 150081, Harbin, Heilongjiang, China
| | - Qingkang Meng
- Department of pharmacogenomics, College of Bioinformatics Science and Technology, Harbin Medical University, 150081, Harbin, Heilongjiang, China
| | - Yiyang Cai
- Department of pharmacogenomics, College of Bioinformatics Science and Technology, Harbin Medical University, 150081, Harbin, Heilongjiang, China
| | - Xiujie Chen
- Department of pharmacogenomics, College of Bioinformatics Science and Technology, Harbin Medical University, 150081, Harbin, Heilongjiang, China.
| |
Collapse
|
11
|
Tian Z, Jia W, Wang Z, Mao H, Zhang J, Shi Q, Li X, Song S, Zhang J, Zhu Y, Yang B, Huang C, Huang J. Clinical significance of immune-related antigen CD58 in gliomas and analysis of its potential core related gene clusters. Heliyon 2024; 10:e29275. [PMID: 38699747 PMCID: PMC11063413 DOI: 10.1016/j.heliyon.2024.e29275] [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: 03/23/2024] [Accepted: 04/03/2024] [Indexed: 05/05/2024] Open
Abstract
Background The clinical significance of immune-related antigen CD58 in gliomas remains uncertain. The aim of this study was to examine the clinical importance and possible core related genes of CD58 in gliomas. Methods Pan-cancer analysis was to observe the association between CD58 and different tumors, glioma RNA sequencing data and clinical sample analyses were used to observe the relationship between CD58 and glioma, shRNA interference models were to observe the impact of CD58 on glioma cell function, and four glioma datasets and two online analysis platforms were used to explore the core related genes affecting the correlation between CD58 and glioma. Results High CD58 expression was associated with worse prognosis in various tumors and higher malignancy in glioma. Down regulation of CD58 expression was linked to decreased proliferation, increased apoptosis, and reduced metastasis in glioma cells. The pathways involved in CD58-related effects were enriched for immune cell adhesion and immune factor activation, and the core genes were CASP1, CCL2, IL18, MYD88, PTPRC, and TLR2. The signature of CD58 and its core-related genes showed superior predictive power for glioma prognosis. Conclusion High CD58 expression is correlated with more malignant glioma types, and also an independent risk factor for mortality in glioma. CD58 and its core-related genes may serve as novel biomarkers for diagnosing and treating glioma.
Collapse
Affiliation(s)
- Zhi Tian
- Department of Neurosurgery, First Affiliated Hospital, Jishou University, Jishou, Hunan, 416000 PR China
| | - Wei Jia
- Medical College of Jishou University, Jishou, Hunan, 416000 PR China
| | - Zhao Wang
- Department of Neurosurgery, First Affiliated Hospital, Jishou University, Jishou, Hunan, 416000 PR China
| | - Hui Mao
- Department of Neurosurgery, First Affiliated Hospital, Jishou University, Jishou, Hunan, 416000 PR China
| | - Jingjing Zhang
- Department of Neurosurgery, First Affiliated Hospital, Jishou University, Jishou, Hunan, 416000 PR China
| | - Qiongya Shi
- Medical College of Jishou University, Jishou, Hunan, 416000 PR China
| | - Xing Li
- Medical College of Jishou University, Jishou, Hunan, 416000 PR China
| | - Shaoyu Song
- Department of Neurosurgery, First Affiliated Hospital, Jishou University, Jishou, Hunan, 416000 PR China
| | - Jiao Zhang
- Department of Neurosurgery, First Affiliated Hospital, Jishou University, Jishou, Hunan, 416000 PR China
| | - Yingjie Zhu
- Department of Neurosurgery, First Affiliated Hospital, Jishou University, Jishou, Hunan, 416000 PR China
| | - Bo Yang
- Department of Pathology, First Affiliated Hospital, Jishou University, Jishou, Hunan, 416000 PR China
| | - Chunhai Huang
- Department of Neurosurgery, First Affiliated Hospital, Jishou University, Jishou, Hunan, 416000 PR China
| | - Jun Huang
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, Hunan 410008 PR China
| |
Collapse
|
12
|
Yang Q, Yang G, Wu Y, Zhang L, Song Z, Yang D. Bioinformatics analysis and validation of genes related to paclitaxel's anti-breast cancer effect through immunogenic cell death. Heliyon 2024; 10:e28409. [PMID: 38560098 PMCID: PMC10979210 DOI: 10.1016/j.heliyon.2024.e28409] [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: 09/28/2023] [Revised: 03/14/2024] [Accepted: 03/18/2024] [Indexed: 04/04/2024] Open
Abstract
Research indicated that Paclitaxel (PTX) can induce immunogenic cell death (ICD) through immunogenic modulation. However, the combination of PTX and ICD has not been extensively studied in breast cancer (BRCA). The TCGA-BRCA and GSE20685 datasets were enrolled in this study. Samples from the TCGA-BRCA dataset were consistently clustered based on selected immunogenic cell death-related genes (ICD-RGs). Next, candidate genes were obtained by overlapping differentially expressed genes (DEGs) between BRCA and normal groups, intersecting genes common to DEGs between cluster1 and cluster2 and hub module genes, and target genes of PTX from five databases. The univariate Cox algorithm and the least absolute shrinkage and selection operator (LASSO) were performed to obtain biomarkers and build a risk model. Following observing the immune microenvironment in differential risk subgroups, single-gene gene set enrichment analysis (GSEA) was carried out in all biomarkers. Finally, the expression of biomarkers was analyzed. Enrichment analysis showed that 626 intersecting genes were linked with inflammatory response. Further five biomarkers (CHI3L1, IL18, PAPLN, SH2D2A, and UBE2L6) were identified and a risk model was built. The model's performance was validated using GSE20685 dataset. Furthermore, the biomarkers were enriched with adaptive immune response. Lastly, the experimental results indicated that the alterations in IL18, SH2D2A, and CHI3L1 expression after treatment matched those in the public database. In this study, Five PTX-ICD-related biomarkers (CHI3L1, IL18, PAPLN, SH2D2A, and UBE2L6) were identified to aid in predicting BRCA treatment outcomes.
Collapse
Affiliation(s)
- Qianmei Yang
- School of Pharmaceutical Science & Yunnan Provincial Key Laboratory of Pharmacology for Natural Products, Kunming Medical University, Kunming, Yunnan, 650500, PR China
- Yunnan College of Modern Biomedical Industry, Kunming, Yunnan, 650500, PR China
| | - Guimei Yang
- School of Pharmaceutical Science & Yunnan Provincial Key Laboratory of Pharmacology for Natural Products, Kunming Medical University, Kunming, Yunnan, 650500, PR China
- Yunnan College of Modern Biomedical Industry, Kunming, Yunnan, 650500, PR China
| | - Yi Wu
- Science and Technology Achievement Incubation Center, Kunming Medical University, Kunming, Yunnan, 650500, PR China
| | - Lun Zhang
- School of Pharmaceutical Science & Yunnan Provincial Key Laboratory of Pharmacology for Natural Products, Kunming Medical University, Kunming, Yunnan, 650500, PR China
| | - Zhuoyang Song
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, 325035, PR China
| | - Dan Yang
- School of Pharmaceutical Science & Yunnan Provincial Key Laboratory of Pharmacology for Natural Products, Kunming Medical University, Kunming, Yunnan, 650500, PR China
| |
Collapse
|
13
|
Li F, Feng Q, Tao R. Machine learning-based cell death signature for predicting the prognosis and immunotherapy benefit in stomach adenocarcinoma. Medicine (Baltimore) 2024; 103:e37314. [PMID: 38457593 PMCID: PMC10919539 DOI: 10.1097/md.0000000000037314] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/30/2023] [Revised: 01/27/2024] [Accepted: 01/29/2024] [Indexed: 03/10/2024] Open
Abstract
Stomach adenocarcinoma (STAD) is a one of most common malignancies with high mortality-to-incidence ratio. Programmed cell death (PCD) exerts vital functions in the progression of cancer. The role of PCD-related genes (PRGs) in STAD are not fully clarified. Using TCGA, GSE15459, GSE26253, GSE62254 and GSE84437 datasets, PCD-related signature (PRS) was constructed with an integrative procedure including 10 machine learning methods. The role of PRS in predicting the immunotherapy benefits was evaluated by several predicting score and 3 immunotherapy datasets (GSE91061, GSE78220, and IMvigor210). The model developed by Lasso + CoxBoost algorithm having a highest average C-index of 0.66 was considered as the optimal PRS. As an independent risk factor for STAD patients, PRS had a good performance in predicting the overall survival rate of patients, with an AUC of 1-, 3-, and 5-year ROC curve being 0.771, 0.751 and 0.827 in TCGA cohort. High PRS score demonstrated a lower gene set score of some immune-activated cells and immune-activated activities. Patient with high PRS score had a higher TIDE score, higher immune escape score, lower PD1&CTLA4 immunophenoscore, lower TMB score, lower response rate and poor prognosis, indicating a less immunotherapy response. The IC50 value of some drugs correlated with chemotherapy and targeted therapy was higher in high PRS score group. Our investigation developed an optimal PRS in STAD and it acted as an indicator for predicting the prognosis, stratifying risk and guiding treatment for STAD patients.
Collapse
Affiliation(s)
- Fan Li
- Department of Emergency, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China
| | - Qian Feng
- Department of Emergency, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China
| | - Ran Tao
- Department of Emergency, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China
| |
Collapse
|
14
|
Huang S, Liu W, Zhao Q, Chen T, Huang R, Dong L, Nian Z, Yang L. Immunogenic Cell Death-related Signature Evaluates the Tumor Microenvironment and Predicts the Prognosis in Diffuse Large B-Cell Lymphoma. Biochem Genet 2024:10.1007/s10528-024-10697-6. [PMID: 38446321 DOI: 10.1007/s10528-024-10697-6] [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: 10/25/2023] [Accepted: 01/10/2024] [Indexed: 03/07/2024]
Abstract
Current literatures suggest a growing body of evidence highlighting the pivotal role of Immunogenic Cell Death (ICD) in multiple tumor types. Nevertheless, the potential and mechanisms of ICD in diffuse large B-cell lymphoma (DLBCL) remain inadequately studied. To address this gap, our current study aims to examine the impact of ICD on DLBCL and identify a corresponding gene signature in DLBC. Using the expression profiles of ICD-associated genes, the gene expression omnibus (GEO) samples were segregated into ICD-high and ICD-low subtypes utilizing non-negative matrix factorization clustering. Next, univariate and LASSO Cox regression analyses were employed to establish the ICD-related gene signature. Subsequently, the CIBERSORT tool, ssGSEA, and ESTIMATE algorithm were utilized to examine the association between the signature and tumor immune microenvironment of DLBC. Finally, the oncoPredict algorithm was implemented to evaluate the drug sensitivity prediction of DLBCL patients. These findings suggest that the immune microenvironment of the ICD-high group with a poor prognosis was significantly suppressed. An 8-gene ICD-related signature was identified and validated to prognosticate and evaluate the tumor immune microenvironment in DLBCL. Similarly, the high-risk group exhibited a worse prognosis compared to the low-risk group, and the immune function was considerably suppressed. Moreover, the results of oncoPredict algorithm indicated that patients in the high-risk group exhibited higher sensitivity to Cisplatin, Cytarabine, Epirubicin, Oxaliplatin, and Vincristine with low IC50. In conclusion, the present study provides novel insights into the role of ICD in DLBCL by identifying a new biomarker for the disease and may have implications for the development of immune-targeted therapies for the tumor.
Collapse
Affiliation(s)
- Shengqiang Huang
- Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, No.420, Fuma Road, Fuzhou, Fujian, China
| | - Wenbin Liu
- Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, No.420, Fuma Road, Fuzhou, Fujian, China
| | - Qiuling Zhao
- Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, No.420, Fuma Road, Fuzhou, Fujian, China
| | - Ting Chen
- Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, No.420, Fuma Road, Fuzhou, Fujian, China
| | - Ruyi Huang
- Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, No.420, Fuma Road, Fuzhou, Fujian, China
| | - Liangliang Dong
- Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, No.420, Fuma Road, Fuzhou, Fujian, China
| | - Zilin Nian
- Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, No.420, Fuma Road, Fuzhou, Fujian, China
| | - Lin Yang
- Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, No.420, Fuma Road, Fuzhou, Fujian, China.
| |
Collapse
|
15
|
Dong P, Zhao L, Zhao L, Zhang J, Lu G, Zhang H, Ma M. A model based on immunogenic cell death-related genes predicts prognosis and response to immunotherapy in kidney renal clear cell carcinoma. Transl Cancer Res 2024; 13:249-267. [PMID: 38410237 PMCID: PMC10894358 DOI: 10.21037/tcr-23-214] [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: 02/14/2023] [Accepted: 10/18/2023] [Indexed: 02/28/2024]
Abstract
Background The prognosis of patients with kidney renal clear cell carcinoma (KIRC), a life-threatening condition, is poor. Immunogenic cell death (ICD) induces regulated cell death via immunogenic signal secretion and exposure. ICD induces regulated cell death through immunogenic signal secretion and exposure. ICD plays an essential role in tumorigenesis, however, the role of ICD in KIRC remains unclear. Methods This study examined the expression levels of 34 ICD-related genes in The Cancer Genome Atlas (TCGA) data set. Signature genes linked to KIRC survival were identified using Cox regression. Next, a prognostic risk model (RM) was built. Subsequently, the KIRC patients were divided into low- and high-risk groups. Kaplan-Meier curves and receiver operating characteristic (ROC) curves were plotted. Gene Ontology and Kyoto Encyclopedia of Genes and Genomes analyses were carried out to investigate the possible role of differential gene expression between the two groups. The immune microenvironment (IME) was assessed using Estimation of STromal and Immune cells in MAlignant Tumor tissues using Expression, CIBERSORT, and single-sample gene-set enrichment analysis algorithms. An enrichment analysis was used to determine the biological significance of these regulatory networks we conducted. The relationship between immune checkpoint gene expression and risk score, and the relationship between treatment outcome and gene expression were assessed using correlation analyses. Results We developed a KIRC RM based on five ICD-related genes (i.e., FOXP3, IFNB1, IL6, LY96, and TLR4), which were identified as the prognostic signature genes. Using the TCGA data set, we conducted a survival analysis and found that the 3-year RM had an area under the curve (AUC) of 0.735, which validated the reliability of the signature. Similarly, using the International Cancer Genome Consortium (ICGC) data set, we found that the 3-year RM had an AUC of 0.732. Conclusions A RM based on five ICD-related genes was built to predict the prognosis of KIRC patients. This RM predicted patient prognosis and reflected the tumor IME of KIRC patients. Thus, this RM could be used to promote individualized treatments and provide potential novel targets for immunotherapy.
Collapse
Affiliation(s)
- Pei Dong
- Department of Clinical Laboratory, The Fourth Hospital of Hebei Medical University, Shijiazhuang, China
| | - Lincong Zhao
- Information Security Center, Information and Communication Branch of State Grid Hebei Electric Power Co. Ltd., Shijiazhuang, China
| | - Lianmei Zhao
- Research Center, The Fourth Hospital of Hebei Medical University, Shijiazhuang, China
| | - Jinyan Zhang
- Department of Clinical Laboratory, The Fourth Hospital of Hebei Medical University, Shijiazhuang, China
| | - Gang Lu
- Department of Clinical Laboratory, The Fourth Hospital of Hebei Medical University, Shijiazhuang, China
| | - Hong Zhang
- Department of Clinical Laboratory, The Fourth Hospital of Hebei Medical University, Shijiazhuang, China
| | - Ming Ma
- Department of Clinical Laboratory, The Fourth Hospital of Hebei Medical University, Shijiazhuang, China
| |
Collapse
|
16
|
Mishchenko TA, Turubanova VD, Gorshkova EN, Krysko O, Vedunova MV, Krysko DV. Glioma: bridging the tumor microenvironment, patient immune profiles and novel personalized immunotherapy. Front Immunol 2024; 14:1299064. [PMID: 38274827 PMCID: PMC10809268 DOI: 10.3389/fimmu.2023.1299064] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2023] [Accepted: 12/11/2023] [Indexed: 01/27/2024] Open
Abstract
Glioma is the most common primary brain tumor, characterized by a consistently high patient mortality rate and a dismal prognosis affecting both survival and quality of life. Substantial evidence underscores the vital role of the immune system in eradicating tumors effectively and preventing metastasis, underscoring the importance of cancer immunotherapy which could potentially address the challenges in glioma therapy. Although glioma immunotherapies have shown promise in preclinical and early-phase clinical trials, they face specific limitations and challenges that have hindered their success in further phase III trials. Resistance to therapy has been a major challenge across many experimental approaches, and as of now, no immunotherapies have been approved. In addition, there are several other limitations facing glioma immunotherapy in clinical trials, such as high intra- and inter-tumoral heterogeneity, an inherently immunosuppressive microenvironment, the unique tissue-specific interactions between the central nervous system and the peripheral immune system, the existence of the blood-brain barrier, which is a physical barrier to drug delivery, and the immunosuppressive effects of standard therapy. Therefore, in this review, we delve into several challenges that need to be addressed to achieve boosted immunotherapy against gliomas. First, we discuss the hurdles posed by the glioma microenvironment, particularly its primary cellular inhabitants, in particular tumor-associated microglia and macrophages (TAMs), and myeloid cells, which represent a significant barrier to effective immunotherapy. Here we emphasize the impact of inducing immunogenic cell death (ICD) on the migration of Th17 cells into the tumor microenvironment, converting it into an immunologically "hot" environment and enhancing the effectiveness of ongoing immunotherapy. Next, we address the challenge associated with the accurate identification and characterization of the primary immune profiles of gliomas, and their implications for patient prognosis, which can facilitate the selection of personalized treatment regimens and predict the patient's response to immunotherapy. Finally, we explore a prospective approach to developing highly personalized vaccination strategies against gliomas, based on the search for patient-specific neoantigens. All the pertinent challenges discussed in this review will serve as a compass for future developments in immunotherapeutic strategies against gliomas, paving the way for upcoming preclinical and clinical research endeavors.
Collapse
Affiliation(s)
- Tatiana A. Mishchenko
- Institute of Biology and Biomedicine, National Research Lobachevsky State University of Nizhny Novgorod, Nizhny Novgorod, Russia
| | - Victoria D. Turubanova
- Institute of Biology and Biomedicine, National Research Lobachevsky State University of Nizhny Novgorod, Nizhny Novgorod, Russia
- Neuroscience Research Institute, National Research Lobachevsky State University of Nizhny Novgorod, Nizhny Novgorod, Russia
| | - Ekaterina N. Gorshkova
- Institute of Biology and Biomedicine, National Research Lobachevsky State University of Nizhny Novgorod, Nizhny Novgorod, Russia
| | - Olga Krysko
- Cell Death Investigation and Therapy Laboratory, Anatomy and Embryology Unit, Department of Human Structure and Repair, Faculty of Medicine and Health Sciences, Ghent University, Ghent, Belgium
| | - Maria V. Vedunova
- Institute of Biology and Biomedicine, National Research Lobachevsky State University of Nizhny Novgorod, Nizhny Novgorod, Russia
- Faculty of Biology and Biotechnologies, National Research University Higher School of Economics, Moscow, Russia
| | - Dmitri V. Krysko
- Institute of Biology and Biomedicine, National Research Lobachevsky State University of Nizhny Novgorod, Nizhny Novgorod, Russia
- Cell Death Investigation and Therapy Laboratory, Anatomy and Embryology Unit, Department of Human Structure and Repair, Faculty of Medicine and Health Sciences, Ghent University, Ghent, Belgium
- Department of Pathophysiology, Sechenov First Moscow State Medical University (Sechenov University), Moscow, Russia
- Cancer Research Institute Ghent, Ghent, Belgium
| |
Collapse
|
17
|
Han Y, Dong Z, Xing Y, Zhan Y, Zou J, Wang X. Establishment of a prognosis prediction model for lung squamous cell carcinoma related to PET/CT: basing on immunogenic cell death-related lncRNA. BMC Pulm Med 2023; 23:511. [PMID: 38102594 PMCID: PMC10724919 DOI: 10.1186/s12890-023-02792-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: 04/27/2023] [Accepted: 11/28/2023] [Indexed: 12/17/2023] Open
Abstract
BACKGROUND Immunogenic cell death (ICD) stimulates adaptive immunity and holds significant promise in cancer therapy. Nevertheless, the influence of ICD-associated long non-coding RNAs (lncRNAs) on the prognosis of patients with lung squamous cell carcinoma (LUSC) remains unexplored. METHODS We employed data from the The Cancer Genome Atlas (TCGA)database to identify ICD-related lncRNAs associated with the prognosis of LUSC using univariate Cox regression analysis. Subsequently, we utilized the LOSS regression model to construct a predictive risk model for assessing the prognosis of LUSC patients based on ICD-related lncRNAs. Our study randomly allocated187 TCGA patients into a training group and 184 patients for testing the predictive model. Furthermore, we conducted quantitative polymerase chain reaction (qPCR) analysis on 43 tumor tissues from LUSC patients to evaluate lncRNA expression levelsPearson correlation analysis was utilized to analyze the correlation of risk scores with positron emission tomography/computed tomography (PET/CT) parameters among LUSC patients. RESULTS The findings from the univariate Cox regression revealed 16 ICD-associated lncRNAs linked to LUSC prognosis, with 12 of these lncRNAs integrated into our risk model utilizing the LOSS regression. Survival analysis indicated a markedly higher overall survival time among patients in the low-risk group compared to those in the high-risk group. The area under the Receiver operating characteristic (ROC) curve to differentiate high-risk and low-risk patients was 0.688. Additionally, the overall survival rate was superior in the low-risk group compared to the high-risk group. Correlation analysis demonstrated a positive association between the risk score calculated based on the ICD-lncRNA risk model and the maximum standard uptake value (SUVmax) (r = 0.427, P = 0.0043) as well as metabolic volume (MTV)of PET-CT (r = 0.360, P = 0.0177) in 43 LUSC patients. CONCLUSION We have successfully developed a risk model founded on ICD-related lncRNAs that proves effective in predicting the overall survival of LUSC patients.
Collapse
Affiliation(s)
- Yu Han
- Nuclear medicine, Cangzhou Central Hospital, Cangzhou, China
| | - Zhiqiang Dong
- 2nd Department of Hepatobiliary and Pancreatic Surgery, Cangzhou People's Hospital, Cangzhou, China
| | - Yu Xing
- Nuclear medicine, Cangzhou Central Hospital, Cangzhou, China
| | - Yingying Zhan
- Nuclear medicine, Cangzhou Central Hospital, Cangzhou, China
| | - Jinhai Zou
- Nuclear medicine, Cangzhou Central Hospital, Cangzhou, China.
| | - Xiaodong Wang
- Department of Pathology, Zhangjiakou Integrated Traditional Chinese and Western Medicine Hospital, Zhangjiakou, China
| |
Collapse
|
18
|
Yu C, Yang W, Tian L, Qin Y, Gong Y, Cheng W. Construction of immunogenic cell death-related molecular subtypes and prognostic signature in colorectal cancer. Open Med (Wars) 2023; 18:20230836. [PMID: 38025525 PMCID: PMC10655694 DOI: 10.1515/med-2023-0836] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2023] [Revised: 09/22/2023] [Accepted: 10/10/2023] [Indexed: 12/01/2023] Open
Abstract
Immunotherapy is a promising treatment for advanced colorectal cancers (CRCs). However, immunotherapy resistance remains a common problem. Immunogenic cell death (ICD), a form of regulated cell death, induces adaptive immunity, thereby enhancing anti-tumor immunity. Research increasingly suggests that inducing ICD is a promising avenue for cancer immunotherapy and identifying ICD-related biomarkers for CRCs would create a new direction for targeted therapies. Thus, this study used bioinformatics to address these questions and create a prognostic signature, aiming to improve individualized CRC treatment. We identified two ICD -related molecular subtypes of CRCs. The high subtype showed pronounced immune cell infiltration, high immune activity, and high expression of human leukocyte antigen and immune checkpoints genes. Subsequently, we constructed and validated a prognostic signature comprising six genes (CD1A, TSLP, CD36, TIMP1, MC1R, and NRG1) using random survival forest analyses. Further analysis using this prediction model indicated that patients with CRCs in the low-risk group exhibited favorable clinical outcomes and better immunotherapy responses than those in the high-risk group. Our findings provide novel insights into determining the prognosis and design of personalized immunotherapeutic strategies for patients with CRCs.
Collapse
Affiliation(s)
- Chun Yu
- Department of Gastroenterology, Jiangsu Province Hospital and Nanjing Medical University First Affiliated Hospital, Nanjing210029, China
| | - Weixuan Yang
- Department of Gastroenterology, The Fifth People’s Hospital of Huai’an, Huai’an223300, China
| | - Li Tian
- Department of Gastroenterology, Zigong Fourth People’s Hospital, Zigong643000, China
| | - Yue Qin
- Department of Gastroenterology, Jiangsu Province Hospital and Nanjing Medical University First Affiliated Hospital, Nanjing210029, China
| | - Yaoyao Gong
- Department of Gastroenterology, Jiangsu Province Hospital and Nanjing Medical University First Affiliated Hospital, Nanjing210029, China
| | - Wenfang Cheng
- Department of Gastroenterology, Jiangsu Province Hospital and Nanjing Medical University First Affiliated Hospital, Nanjing210029, China
| |
Collapse
|
19
|
Shi X, Ding H, Tao J, Zhu Y, Zhang X, He G, Yang J, Wu X, Liu X, Yu X. Comprehensive evaluation of cell death-related genes as novel diagnostic biomarkers for breast cancer. Heliyon 2023; 9:e21341. [PMID: 38027811 PMCID: PMC10643282 DOI: 10.1016/j.heliyon.2023.e21341] [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: 07/29/2023] [Revised: 10/17/2023] [Accepted: 10/19/2023] [Indexed: 12/01/2023] Open
Abstract
Background Breast cancer (BRCA) ranks first among cancers in terms of incidence and mortality rates in women, primarily owing to metastasis, chemo-resistance, and heterogeneity. To predict long-term prognosis and design novel therapies for BRCA, more sensitive markers need to be explored. Methods Data from 1089 BRCA patients were downloaded from TCGA database. Pearson's correlation analysis and univariate and multivariate Cox regression analyses were performed to assess the role of cell death-related genes (CDGs) in predicting BRCA prognosis. Kaplan-Meier survival curves were generated to compare the overall survival in the two subgroups. A nomogram was constructed using risk scores based on the five CDGs and other clinicopathological features. CCK-8, EdU incorporation, and colony formation assays were performed to verify the inhibitory effect of NFKBIA on BRCA cell proliferation. Transwell assay, flow cytometry, and immunohistochemistry analyses were performed to ascertain the biological function of NFKBIA. Results Five differentially expressed CDGs were detected among 156 CDGs. The risk score for each patient was then calculated based on the expression levels of the five CDGs. Distinct differences in immune infiltration, expression of immune-oncological targets, mutation status, and half-maximal inhibitory concentration values of some targeted drugs were observed between the high- and low-risk groups. Finally, in vitro cell experiments verified that NFKBIA overexpression suppresses the proliferation and migration of BRCA cells. Conclusions Our study revealed that some CDGs, especially NFKBIA, could serve as sensitive markers for predicting the prognosis of patients with BRCA and designing more personalized clinical therapies.
Collapse
Affiliation(s)
- Xiaoyue Shi
- Department of Breast Surgery, The First Affiliated Hospital of Nanjing Medical University, 300 Guangzhou Road, Nanjing, Jiangsu, 210029, People's Republic of China
| | - Hao Ding
- Department of Breast Surgery, Baoying Maternal and Child Health Hospital, 120 Anyi East Road, Yangzhou, Jiangsu 225800, People's Republic of China
| | - Jing Tao
- Department of Thyroid-Breast Surgery, Nanjing Pukou Hospital, The Fourth Affiliated Hospital of Nanjing Medical University, 18 Puyuan Road, Nanjing, Jiangsu 210031, People's Republic of China
| | - Yanhui Zhu
- Department of Breast Surgery, The First Affiliated Hospital of Nanjing Medical University, 300 Guangzhou Road, Nanjing, Jiangsu, 210029, People's Republic of China
| | - Xiaoqiang Zhang
- Department of Breast Surgery, The First Affiliated Hospital of Nanjing Medical University, 300 Guangzhou Road, Nanjing, Jiangsu, 210029, People's Republic of China
| | - Gao He
- Department of Breast Surgery, The First Affiliated Hospital of Nanjing Medical University, 300 Guangzhou Road, Nanjing, Jiangsu, 210029, People's Republic of China
| | - Junzhe Yang
- Department of Breast Surgery, The First Affiliated Hospital of Nanjing Medical University, 300 Guangzhou Road, Nanjing, Jiangsu, 210029, People's Republic of China
| | - Xian Wu
- Department of Breast Surgery, The First Affiliated Hospital of Nanjing Medical University, 300 Guangzhou Road, Nanjing, Jiangsu, 210029, People's Republic of China
| | - Xiaoan Liu
- Department of Breast Surgery, The First Affiliated Hospital of Nanjing Medical University, 300 Guangzhou Road, Nanjing, Jiangsu, 210029, People's Republic of China
| | - Xiafei Yu
- Department of Breast Surgery, The First Affiliated Hospital of Nanjing Medical University, 300 Guangzhou Road, Nanjing, Jiangsu, 210029, People's Republic of China
| |
Collapse
|
20
|
Luo Q, Zhuang J, Zheng D, Miao C, Luo H, Peng J, Zheng C, Qin C, Lan C, Chen M, Xia Y, Huang D, Chen Z. IGFBP2 from a novel copper metabolism-associated biomarker promoted glioma progression and response to immunotherapy. Front Immunol 2023; 14:1282734. [PMID: 37928523 PMCID: PMC10620745 DOI: 10.3389/fimmu.2023.1282734] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2023] [Accepted: 10/02/2023] [Indexed: 11/07/2023] Open
Abstract
Introduction Copper metabolism encompasses all cellular metabolic processes involving copper ions and plays a significant role in the pathogenesis of diseases, including cancer. Furthermore, copper is intricately involved in various processes related to nucleotide metabolism. However, a comprehensive analysis of copper metabolism in gliomas remains lacking despite its importance. Methods To address this gap, glioma patients were stratified based on the expression levels of copper metabolism-related genes. By utilizing machine learning techniques, a novel copper metabolism-associated biomarker was developed. The potential of this biomarker in prognosis, mutation analysis, and predicting immunotherapy response efficiency in gliomas was systematically investigated. Results Notably, IGFBP2, identified as a glioma tumor promoter, was found to promote disease progression and influence immunotherapy response. Additionally, glioma-derived IGFBP2 was observed to enhance microglial migration. High IGFBP2 expression in GBM cells facilitated macrophage interactions through the EGFR, CD63, ITGB1, and CD44 signaling pathways. Discussion: Overall, the copper metabolism-associated biomarker shows promising potential to enhance the clinical management of gliomas, offering valuable insights into disease prognosis and treatment strategies.
Collapse
Affiliation(s)
- Qisheng Luo
- Department of Neurosurgery, Affiliated Hospital of Youjiang Medical University for Nationalities, Baise, Guangxi, China
| | - Junhong Zhuang
- Department of Neurology, Affiliated Haikou Hospital of Xiangya Medical School, Central South University, Haikou, China
| | - Dandan Zheng
- Department of Radiation Oncology, The First Affiliated Hospital Zhejiang University, Hangzhou, China
| | - Changfeng Miao
- Department of Laboratory Medicine, Neurosurgery Second Branche, Hunan Provincial People’s Hospital (The First affiliated Hospital of Hunan Normal University), Changsha, Hunan, China
| | - Hongcheng Luo
- Department of Laboratory Medicine, Affiliated Hospital of Youjiang Medical University for Nationalities, Baise, Guangxi, China
| | - Jun Peng
- Department of Neurosurgery, Affiliated Haikou Hospital of Xiangya Medical School, Central South University, Haikou, China
| | - Chuanhua Zheng
- Department of Neurosurgery, Affiliated Hospital of Youjiang Medical University for Nationalities, Baise, Guangxi, China
| | - Chengjian Qin
- Department of Neurosurgery, Affiliated Hospital of Youjiang Medical University for Nationalities, Baise, Guangxi, China
| | - Chuanliu Lan
- Department of Neurosurgery, Affiliated Hospital of Youjiang Medical University for Nationalities, Baise, Guangxi, China
| | - Meiqin Chen
- Department of Radiation Oncology, Affiliated Jinhua Hospital, Zhejiang University School of Medicine, Jinhua, Zhejiang, China
| | - Ying Xia
- Department of Neurosurgery, Affiliated Haikou Hospital of Xiangya Medical School, Central South University, Haikou, China
| | - Deyou Huang
- Department of Radiology, Affiliated Hospital of Youjiang Medical University for Nationalities, Baise, Guangxi, China
| | - Zigui Chen
- Department of Neurosurgery, Affiliated Haikou Hospital of Xiangya Medical School, Central South University, Haikou, China
| |
Collapse
|
21
|
Li Z, Wang B, Liang H, Li Y, Zhang Z, Han L. A three-stage eccDNA based molecular profiling significantly improves the identification, prognosis assessment and recurrence prediction accuracy in patients with glioma. Cancer Lett 2023; 574:216369. [PMID: 37640198 DOI: 10.1016/j.canlet.2023.216369] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2023] [Revised: 08/15/2023] [Accepted: 08/24/2023] [Indexed: 08/31/2023]
Abstract
Glioblastoma (GBM) progression is influenced by intratumoral heterogeneity. Emerging evidence has emphasized the pivotal role of extrachromosomal circular DNA (eccDNA) in accelerating tumor heterogeneity, particularly in GBM. However, the eccDNA landscape of GBM has not yet been elucidated. In this study, we first identified the eccDNA profiles in GBM and adjacent tissues using circle- and RNA-sequencing data from the same samples. A three-stage model was established based on eccDNA-carried genes that exhibited consistent upregulation and downregulation trends at the mRNA level. Combinations of machine learning algorithms and stacked ensemble models were used to improve the performance and robustness of the three-stage model. In stage 1, a total of 113 combinations of machine learning algorithms were constructed and validated in multiple external cohorts to accurately distinguish between low-grade glioma (LGG) and GBM in patients with glioma. The model with the highest area under the curve (AUC) across all cohorts was selected for interpretability analysis. In stage 2, a total of 101 combinations of machine learning algorithms were established and validated for prognostic prediction in patients with glioma. This prognostic model performed well in multiple glioma cohorts. Recurrent GBM is invariably associated with aggressive and refractory disease. Therefore, accurate prediction of recurrence risk is crucial for developing individualized treatment strategies, monitoring patient status, and improving clinical management. In stage 3, a large-scale GBM cohort (including primary and recurrent GBM samples) was used to fit the GBM recurrence prediction model. Multiple machine learning and stacked ensemble models were fitted to select the model with the best performance. Finally, a web tool was developed to facilitate the clinical application of the three-stage model.
Collapse
Affiliation(s)
- Zesheng Li
- Tianjin Neurological Institute, Key Laboratory of Post-Neuro Injury, Neuro-repair and Regeneration in Central Nervous System, Ministry of Education and Tianjin City, Tianjin Medical University General Hospital, Tianjin, 300052, China
| | - Bo Wang
- Tianjin Neurological Institute, Key Laboratory of Post-Neuro Injury, Neuro-repair and Regeneration in Central Nervous System, Ministry of Education and Tianjin City, Tianjin Medical University General Hospital, Tianjin, 300052, China
| | - Hao Liang
- Tianjin Neurological Institute, Key Laboratory of Post-Neuro Injury, Neuro-repair and Regeneration in Central Nervous System, Ministry of Education and Tianjin City, Tianjin Medical University General Hospital, Tianjin, 300052, China
| | - Ying Li
- Tianjin Neurological Institute, Key Laboratory of Post-Neuro Injury, Neuro-repair and Regeneration in Central Nervous System, Ministry of Education and Tianjin City, Tianjin Medical University General Hospital, Tianjin, 300052, China
| | - Zhenyu Zhang
- Department of Neurosurgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, 480082, China.
| | - Lei Han
- Tianjin Neurological Institute, Key Laboratory of Post-Neuro Injury, Neuro-repair and Regeneration in Central Nervous System, Ministry of Education and Tianjin City, Tianjin Medical University General Hospital, Tianjin, 300052, China.
| |
Collapse
|
22
|
Chen W, Shu K, Cai C, Ding J, Zhang X, Zhang W, Wang K. Prognostic value and immune landscapes of immunogenic cell death-related lncRNAs in hepatocellular carcinoma. Biosci Rep 2023; 43:BSR20230634. [PMID: 37584192 PMCID: PMC10500227 DOI: 10.1042/bsr20230634] [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: 04/02/2023] [Revised: 05/31/2023] [Accepted: 08/15/2023] [Indexed: 08/17/2023] Open
Abstract
BACKGROUND Both immunogenic cell death (ICD) and long noncoding RNAs (lncRNAs) are strongly associated with tumor development, but the mechanism of action of ICD-associated lncRNAs in hepatocellular carcinoma (HCC) remains unclear. METHODS We collected data from 365 HCC patients from The Cancer Genome Atlas (TCGA) database. We formulated a prognostic signature of ICD-associated lncRNAs and a nomogram to predict prognosis. To explore the potential mechanisms and provide clinical guidance, survival analysis, enrichment analysis, tumor microenvironment analysis, tumor mutation burden (TMB), and drug sensitivity prediction were conducted based on the subgroups obtained from the risk score. RESULTS A prognostic signature of seven ICD-associated lncRNAs was constructed. Kaplan-Meier (K-M) survival curves showed a more unfavorable outcome in high-risk patients. The nomogram had a higher predictive value than the nomogram constructed without the risk model. Enrichment analysis confirmed that risk lncRNAs were closely associated with cell proliferation and mitosis. Most of the immune checkpoints currently used in therapy (e.g., PDCD1 and CTLA4) appeared to be elevated in high-risk patients. Tumor microenvironment analysis showed differential expression of lymphocytes (including natural killer cells, regulatory T cells, etc.) in the high-risk group. TMB had a higher incidence of mutations in the high-risk group (P=0.004). Chemotherapy drug sensitivity prediction provides effective guidelines for individual therapy. RT-qPCR of human HCC tissues verified the accuracy of the model. CONCLUSION We constructed an effective prognostic signature for patients with HCC using seven ICD-lncRNAs, which provides guidance for the prognostic assessment and personalized treatment of patients.
Collapse
Affiliation(s)
- Wanying Chen
- Department of Thoracic Surgery, The Second Affiliated Hospital of Nanchang University, Nanchang 330006, China
- Jiangxi Medical College, Nanchang University, Nanchang 330006, China
| | - Kexin Shu
- Jiangxi Medical College, Nanchang University, Nanchang 330006, China
- Department of Hepatobiliary Surgery, The Second Affiliated Hospital of Nanchang University, Nanchang 330006, China
| | - Chenxi Cai
- Jiangxi Medical College, Nanchang University, Nanchang 330006, China
- Department of Hepatobiliary Surgery, The Second Affiliated Hospital of Nanchang University, Nanchang 330006, China
| | - Jiatong Ding
- Jiangxi Medical College, Nanchang University, Nanchang 330006, China
- Department of Hepatobiliary Surgery, The Second Affiliated Hospital of Nanchang University, Nanchang 330006, China
| | - Xin Zhang
- Department of Thoracic Surgery, The Second Affiliated Hospital of Nanchang University, Nanchang 330006, China
| | - Wenxiong Zhang
- Department of Thoracic Surgery, The Second Affiliated Hospital of Nanchang University, Nanchang 330006, China
| | - Kang Wang
- Department of Traditional Chinese Medicine, The Second Affiliated Hospital of Nanchang University, Nanchang 330006, China
| |
Collapse
|
23
|
Cai C, Shu K, Chen W, Ding J, Guo Z, Wei Y, Zhang W. Construction and validation of a model based on immunogenic cell death-associated lncRNAs to predict prognosis and direct therapy for kidney renal clear cell carcinoma. Aging (Albany NY) 2023; 15:5304-5338. [PMID: 37379129 PMCID: PMC10333057 DOI: 10.18632/aging.204741] [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: 01/31/2023] [Accepted: 05/09/2023] [Indexed: 06/30/2023]
Abstract
BACKGROUND Immunogenic cell death (ICD) is an important part of the antitumor effect, yet the role played by long noncoding RNAs (lncRNAs) remains unclear. We explored the value of ICD-related lncRNAs in tumor prognosis assessment in kidney renal clear cell carcinoma (KIRC) patients to provide a basis for answering the above questions. METHODS Data on KIRC patients were obtained from The Cancer Genome Atlas (TCGA) database, prognostic markers were identified, and their accuracy was verified. An application-validated nomogram was developed based on this information. Furthermore, we performed enrichment analysis, tumor mutational burden (TMB) analysis, tumor microenvironment (TME) analysis, and drug sensitivity prediction to explore the mechanism of action and clinical application value of the model. RT-qPCR was performed to detect the expression of lncRNAs. RESULTS The risk assessment model constructed using eight ICD-related lncRNAs provided insight into patient prognoses. Kaplan-Meier (K-M) survival curves showed a more unfavorable outcome in high-risk patients (p<0.001). The model had good predictive value for different clinical subgroups, and the nomogram constructed based on this model worked well (risk score AUC=0.765). Enrichment analysis revealed that mitochondrial function-related pathways were enriched in the low-risk group. The adverse prognosis of the higher-risk cohort might correspond to a higher TMB. The TME analysis revealed a higher resistance to immunotherapy in the increased-risk subgroup. Drug sensitivity analysis can guide the selection and application of antitumor drugs in different risk groups. CONCLUSIONS This prognostic signature based on eight ICD-associated lncRNAs has significant implications for prognostic assessment and treatment selection in KIRC.
Collapse
Affiliation(s)
- Chenxi Cai
- Department of Thoracic Surgery, The Second Affiliated Hospital of Nanchang University, Nanchang 330006, China
- Jiangxi Medical College, Nanchang University, Nanchang 330006, China
| | - Kexin Shu
- Jiangxi Medical College, Nanchang University, Nanchang 330006, China
- Department of Urinary Surgery, The Second Affiliated Hospital of Nanchang University, Nanchang 330006, China
| | - Wanying Chen
- Jiangxi Medical College, Nanchang University, Nanchang 330006, China
- Department of Urinary Surgery, The Second Affiliated Hospital of Nanchang University, Nanchang 330006, China
| | - Jiatong Ding
- Jiangxi Medical College, Nanchang University, Nanchang 330006, China
- Department of Urinary Surgery, The Second Affiliated Hospital of Nanchang University, Nanchang 330006, China
| | - Zishun Guo
- Department of Thoracic Surgery, The Second Affiliated Hospital of Nanchang University, Nanchang 330006, China
| | - Yiping Wei
- Department of Thoracic Surgery, The Second Affiliated Hospital of Nanchang University, Nanchang 330006, China
| | - Wenxiong Zhang
- Department of Thoracic Surgery, The Second Affiliated Hospital of Nanchang University, Nanchang 330006, China
| |
Collapse
|
24
|
Dong B, Wu Y, Zhang J, Gu Y, Xie R, He X, Pang X, Wang X, Cui Y. A novel immunogenic cell death-related subtype classification and risk signature for predicting prognosis and immunotherapy efficacy in gastric cancer. Front Immunol 2023; 14:1162876. [PMID: 37215130 PMCID: PMC10196197 DOI: 10.3389/fimmu.2023.1162876] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2023] [Accepted: 04/18/2023] [Indexed: 05/24/2023] Open
Abstract
The majority of gastric cancer (GC) patients are in a progressive stage at the initial stage of treatment, and the overall response rate to immunotherapy remains unsatisfactory largely due to the lack of effective prognostic biomarkers. Immunogenic cell death (ICD) was identified as a new form of regulated cell death that can activate adaptive immune responses and further promote immunotherapy efficacy. Therefore, we attempted to characterize the ICD-associated signature to stratify patients who could benefit from immunotherapy. In our study, two subgroups of patients were identified based on the data of 34 ICD-related genes extracted from The Cancer Genome Atlas database via consensus clustering. The estimated scores, stromal scores, immune scores, tumor purity, and survival rate showed significant differences between the low and high ICD groups. Then, we constructed an ICD-related risk signature, including IFNB1, IL6, LY96, and NT5E, using least absolute shrinkage and selection operator Cox regression analysis; then, high- and low-risk groups could be clearly distinguished. Notably, the risk score is a reliable predictor of the prognosis and immunotherapy outcome in GC, which was further validated in an immunohistochemistry assay. These results suggest that ICD is closely associated with the prognosis and tumor immune microenvironment in GC. Taken together, this study first constructed and validated a prognostic ICD-related signature to predict the survival and effect of immunotherapy in GC, which provided new insight for potent individualized immunotherapy strategies.
Collapse
Affiliation(s)
- Bingqi Dong
- Department of General Surgery, Peking University First Hospital, Beijing, China
| | - Yingchao Wu
- Department of General Surgery, Peking University First Hospital, Beijing, China
| | - Junling Zhang
- Department of General Surgery, Peking University First Hospital, Beijing, China
| | - Yanlun Gu
- Department of Pharmacy, Peking University First Hospital, Beijing, China
- Institute of Clinical Pharmacology, Peking University, Beijing, China
| | - Ran Xie
- Department of Pharmacy, Peking University First Hospital, Beijing, China
- Institute of Clinical Pharmacology, Peking University, Beijing, China
| | - Xu He
- Department of Pharmacy, Peking University First Hospital, Beijing, China
- Institute of Clinical Pharmacology, Peking University, Beijing, China
| | - Xiaocong Pang
- Department of Pharmacy, Peking University First Hospital, Beijing, China
- Institute of Clinical Pharmacology, Peking University, Beijing, China
| | - Xin Wang
- Department of General Surgery, Peking University First Hospital, Beijing, China
| | - Yimin Cui
- Department of Pharmacy, Peking University First Hospital, Beijing, China
- Institute of Clinical Pharmacology, Peking University, Beijing, China
| |
Collapse
|
25
|
Li X, Kang J, Yue J, Xu D, Liao C, Zhang H, Zhao J, Liu Q, Jiao J, Wang L, Li G. Identification and validation of immunogenic cell death-related score in uveal melanoma to improve prediction of prognosis and response to immunotherapy. Aging (Albany NY) 2023; 15:3442-3464. [PMID: 37142279 PMCID: PMC10449274 DOI: 10.18632/aging.204680] [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: 02/13/2023] [Accepted: 04/17/2023] [Indexed: 05/06/2023]
Abstract
BACKGROUND Immunogenic cell death (ICD) could activate innate and adaptive immune response. In this work, we aimed to develop an ICD-related signature in uveal melanoma (UVM) patients and facilitate assessment of their prognosis and immunotherapy. METHODS A set of machine learning methods, including non-negative matrix factorization (NMF) method and least absolute shrinkage and selection operator (LASSO) logistic regression model, and bioinformatics analytic tools were integrated to construct an ICD-related risk score (ICDscore). CIBERSORT and ESTIMATE algorithms were used to evaluate the infiltration of immune cells. The Genomics of Drug Sensitivity in Cancer (GDSC), cellMiner and tumor immune dysfunction and exclusion (TIDE) databases were used for therapy sensitivity analyses. The predictive performance between ICDscore with other mRNA signatures was also compared. RESULTS The ICDscore could predict the prognosis of UVM patients in both the training and four validating cohorts. The ICDscore outperformed 19 previously published signatures. Patients with high ICDscore exhibited a substantial increase in immune cell infiltration and expression of immune checkpoint inhibitor-related genes, leading to a higher response rate to immunotherapy. Furthermore, the downregulation of poly (ADP-ribose) polymerase family member 8 (PARP8), a critical gene involved in the development of the ICDscore, resulted in decreased cell proliferation and slower migration of UVM cells. CONCLUSION In conclusion, we developed a robust and powerful ICD-related signature for evaluating the prognosis and benefits of immunotherapy that could serve as a promising tool to guide decision-making and surveillance for UVM patients.
Collapse
Affiliation(s)
- Xiaoyan Li
- Department of Central Laboratory, Shanxi Provincial People’s Hospital, Taiyuan, Shanxi, China
- Department of Blood Transfusion, Shanxi Provincial People’s Hospital, Taiyuan, Shanxi, China
| | - Jing Kang
- Department of Clinical Laboratory, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Third Hospital of Shanxi Medical University, Taiyuan, Shanxi, China
| | - Jing Yue
- Department of Clinical Laboratory, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Third Hospital of Shanxi Medical University, Taiyuan, Shanxi, China
| | - Dawei Xu
- Department of Blood Transfusion, Shanxi Provincial People’s Hospital, Taiyuan, Shanxi, China
| | - Chunhua Liao
- Department of Physiotherapy and Rehabilitation, The Second Affiliated Hospital of Air Force Military Medical University, Xi'an, Shaanxi, China
| | - Huina Zhang
- College of Life Science and Agronomy, Zhoukou Normal University, Zhoukou, Henan, China
| | - Jin Zhao
- College of Life Science and Agronomy, Zhoukou Normal University, Zhoukou, Henan, China
| | - Qiongwen Liu
- College of Life Science and Agronomy, Zhoukou Normal University, Zhoukou, Henan, China
| | - Jinke Jiao
- College of Life Science and Agronomy, Zhoukou Normal University, Zhoukou, Henan, China
| | - Lin Wang
- Department of Geriatrics, Xijing Hospital, The Air Force Military Medical University, Xi'an, Shaanxi, China
| | - Guoyin Li
- College of Life Science and Agronomy, Zhoukou Normal University, Zhoukou, Henan, China
- Key Laboratory of Modern Teaching Technology, Ministry of Education, Shaanxi Normal University, Xi’an, Shaanxi, China
- Academy of Medical Science, Zhengzhou University, Zhengzhou, Henan, China
| |
Collapse
|
26
|
Dong CC, Zhang QH, Zhang Y, Zhang Y, Ruan H, Qin T, Zhao JH, Wu G, Zhu Z, Yang JR. Comprehensive landscape of the IPAF inflammasomes in pan-cancer: A bulk omics research and single-cell sequencing validation. Comput Biol Med 2023; 155:106622. [PMID: 36780800 DOI: 10.1016/j.compbiomed.2023.106622] [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: 08/26/2022] [Revised: 12/28/2022] [Accepted: 01/28/2023] [Indexed: 02/05/2023]
Abstract
BACKGROUND IPAF (ICE-protease Activating Factor) is a nucleotide-binding/leucine-rich repeat (NLR) protein known as the cysteine-associated recruitment domain 12 (CARD12). Previous studies only discuss the role of IPAF inflammasomes in specific tumors. The role of IPAF inflammasomes in pan-cancer is still unclear. Therefore, we performed a comprehensive analysis of IPAF inflammasome in 33 tumors. METHODS We used databases like The Cancer Genome Atlas (TCGA) and Genotype-Tissue Expression (GTEx) from the UCSC XENA (http://xena.ucsc.edu/) to retrieve and analyze gene expression. The influence of IPAF inflammasome on the prognosis of tumor patients was analyzed using univariate Cox regression analysis and Kaplan-Meier survival analysis. Furthermore, we conducted the following analysis: Single-sample gene set enrichment analysis, single-cell level functional state analysis, single-cell sequencing, immune cell infiltration analysis, and tumor immune dysfunction and exclusion (TIDE) score. RESULTS First, the differential expression of IPAF inflammasome-related genes (IPAF-RGs) in 33 tumors were analyzed. The results revealed that IPAF-RGs were significantly and differentially expressed in eight tumors. The prognostic significance of IPAF inflammasome scores was different in different tumors. A positive correlation was observed between IPAF inflammasomes scores and CD8+ T cells in most tumors. Further analysis revealed that IPAF inflammasome might affect tumor immunity mainly by mediating effector T cell recruitment via the expression of chemokines such as CXCL9, CXCL10, and CCL5. The analysis of TIDE and IPAF inflammasome scores revealed a significant negative correlation between IPAF inflammasome and TIDE scores in 11 tumors. CONCLUSION A pan-cancer analysis of IPAF inflammasome in various tumors was performed. The results highlight the potential value of IPAF inflammasome in response to immunotherapy in patients and provide a new direction for future immunotherapy.
Collapse
Affiliation(s)
- Chen-Cheng Dong
- Department of Colorectal and Anal Surgery, The People's Hospital of Guangxi Zhuang Autonomous Region, Guangxi Academy of Medical Sciences, China
| | - Qiu-Huan Zhang
- Department of Colorectal and Anal Surgery, The People's Hospital of Guangxi Zhuang Autonomous Region, Guangxi Academy of Medical Sciences, China
| | - Yan Zhang
- Department of Colorectal and Anal Surgery, Affiliated Tumor Hospital of Guangxi Medical University, China
| | - Yujie Zhang
- Department of Colorectal and Anal Surgery, Affiliated Tumor Hospital of Guangxi Medical University, China
| | - Hanyi Ruan
- Department of Colorectal and Anal Surgery, Affiliated Tumor Hospital of Guangxi Medical University, China
| | - Tianyu Qin
- Department of Colorectal and Anal Surgery, Affiliated Tumor Hospital of Guangxi Medical University, China
| | - Jie-Hua Zhao
- Department of Breast and Thyroid Surgery, The People's Hospital of Guangxi Zhuang Autonomous Region, Guangxi Academy of Medical Sciences, China
| | - Guo Wu
- Department of Colorectal and Anal Surgery, The People's Hospital of Guangxi Zhuang Autonomous Region, Guangxi Academy of Medical Sciences, China
| | - Zhou Zhu
- Department of Colorectal and Anal Surgery, The People's Hospital of Guangxi Zhuang Autonomous Region, Guangxi Academy of Medical Sciences, China.
| | - Jian-Rong Yang
- Department of Breast and Thyroid Surgery, The People's Hospital of Guangxi Zhuang Autonomous Region, Guangxi Academy of Medical Sciences, China.
| |
Collapse
|
27
|
Identification of Immunogenic Cell Death-Related Signature for Glioma to Predict Survival and Response to Immunotherapy. Cancers (Basel) 2022; 14:cancers14225665. [PMID: 36428756 PMCID: PMC9688866 DOI: 10.3390/cancers14225665] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2022] [Revised: 11/16/2022] [Accepted: 11/17/2022] [Indexed: 11/19/2022] Open
Abstract
Immunogenic cell death (ICD) is a type of regulated cell death (RCD) and is correlated with the progression, prognosis, and therapy of tumors, including glioma. Numerous studies have shown that the immunotherapeutic and chemotherapeutic agents of glioma might induce ICD. However, studies on the comprehensive analysis of the role of ICD-related genes and their correlations with overall survival (OS) in glioma are lacking. The genetic, transcriptional, and clinical data of 1896 glioma samples were acquired from five distinct databases and analyzed in terms of genes and transcription levels. The method of consensus unsupervised clustering divided the patients into two disparate molecular clusters: A and B. All of the patients were randomly divided into training and testing groups. Employing the training group data, 14 ICD-related genes were filtered out to develop a risk-score model. The correlations between our risk groups and prognosis, cells in the tumor microenvironment (TME) and immune cells infiltration, chemosensitivity and cancer stem cell (CSC) index were assessed. A highly precise nomogram model was constructed to enhance and optimize the clinical application of the risk score. The results demonstrated that the risk score could independently predict the OS rate and the immunotherapeutic response of glioma patients. This study analyzed the ICD-related genes in glioma and evaluated their role in the OS, clinicopathological characteristics, TME and immune cell infiltration of glioma. Our results may help in assessing the OS of glioma and developing better immunotherapeutic strategies.
Collapse
|