1
|
Yang Y, Li D, Nie J, Wang J, Huang H, Hang X. A Nomogram for Predicting Survival in Patients with SARS-CoV-2 Omicron Variant Pneumonia Based on Admission Data. Infect Drug Resist 2025; 18:2093-2104. [PMID: 40303607 PMCID: PMC12039831 DOI: 10.2147/idr.s509178] [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: 11/28/2024] [Accepted: 04/15/2025] [Indexed: 05/02/2025] Open
Abstract
Purpose Patients with severe SARS-CoV-2 omicron variant pneumonia pose a serious challenge. This study aimed to develop a nomogram for predicting survival using chest computed tomography (CT) imaging features and laboratory test results based on admission data. Patients and Methods A total of 436 patients with SARS-CoV-2 pneumonia (323 and 113 in the training and validation groups, respectively) were enrolled. Pneumonitis volume, assessed on chest CT scans at admission, was used to identify low- and high-risk groups. Risk analysis was performed using clinical symptoms, laboratory findings, and chest CT imaging features. A predictive algorithm was developed using Cox multivariate analysis. Results The high-risk group had a shorter survival duration than the low-risk group. Significant differences in mortality rate, neutrophil and lymphocyte counts, C-reactive protein (CRP) concentration, and urea nitrogen level were observed between the two groups. In the training group, age, pneumonia volume, total bilirubin, and blood urea nitrogen were independent prognostic factors. In the validation group, age, pneumonia volume, neutrophil count, and CRP were independent prognostic factors. A personalized prediction model for survival outcomes was developed using independent predictors. Conclusion A personalized prediction model was created to forecast the 5-, 10-, 15-, 20-, and 30-day survival rates of patients with COVID-19 omicron variant pneumonia based on admission data, and can be used to determine the survival rate and early treatment of severe patients.
Collapse
Affiliation(s)
- Yinghao Yang
- Department of Infectious Diseases, Changzheng Hospital, Naval Medical University, Shanghai, People’s Republic of China
- Department of Infectious Diseases, the 988th Hospital of the Joint Logistic Support Force, Zhengzhou, People’s Republic of China
| | - Dong Li
- Department of Infectious Diseases, Changzheng Hospital, Naval Medical University, Shanghai, People’s Republic of China
- Department of Gastroenterology, The 971th Hospital of PLA Navy, Qingdao, People’s Republic of China
| | - Jinqiu Nie
- Department of Infectious Diseases, Changzheng Hospital, Naval Medical University, Shanghai, People’s Republic of China
| | - Junxue Wang
- Department of Infectious Diseases, Changzheng Hospital, Naval Medical University, Shanghai, People’s Republic of China
| | - Huili Huang
- Department of Infectious Diseases, Changzheng Hospital, Naval Medical University, Shanghai, People’s Republic of China
| | - Xiaofeng Hang
- Department of Infectious Diseases, Changzheng Hospital, Naval Medical University, Shanghai, People’s Republic of China
| |
Collapse
|
2
|
Smith R, Sapkota R, Antony B, Sun J, Aboud O, Bloch O, Daly M, Fragoso R, Yiu G, Liu YA. A novel predictive model utilizing retinal microstructural features for estimating survival outcome in patients with glioblastoma. Clin Neurol Neurosurg 2025; 250:108790. [PMID: 39987704 PMCID: PMC11911018 DOI: 10.1016/j.clineuro.2025.108790] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2025] [Accepted: 02/15/2025] [Indexed: 02/25/2025]
Abstract
PURPOSE Glioblastoma is a highly aggressive brain tumor with poor prognosis despite surgery and chemoradiation. The visual sequelae of glioblastoma have not been well characterized. This study assessed visual outcomes in glioblastoma patients through neuro-ophthalmic exams, imaging of the retinal microstructures/microvasculature, and perimetry. METHODS A total of 19 patients with glioblastoma (9 male, 10 female, average age at diagnosis 69 years) were enrolled. Tumor characteristic, neuro-ophthalmic exam data, Optical Coherence Tomography (OCT) and OCT-Angiography data of all patient eyes were analyzed using Microsoft Excel and a Machine Learning algorithm. RESULTS Best-corrected visual acuity ranged from 20/20 - 20/50. Occipital tumors showed worse visual fields than frontal tumors (mean deviation -14.9 and -0.23, respectively, p < 0.0001). Those with overall survival (OS)< 15 months demonstrated thinner retinal nerve fiber layer and ganglion cell complex (p < 0.0001) and enlarged foveal avascular zone starting from 4 months post-diagnosis (p = 0.006). There was no significant difference between eyes ipsilateral and contralateral to radiation fields (average doses were 1370 cGy and 1180 cGy, respectively, p = 0.42). A machine learning algorithm using retinal microstructure and visual fields predicted patients with long (≥15 months) progression-free and overall survival with 78 % accuracy. CONCLUSION Glioblastoma patients frequently present with visual field defects despite normal visual acuity. Patients with poor survival duration demonstrated significant retinal thinning and decreased microvascular density. A machine learning algorithm predicted survival though further validation is warranted.
Collapse
Affiliation(s)
- Rebekah Smith
- School of Medicine, University of California, Davis, USA
| | - Ranjit Sapkota
- Institute of Innovation, Science & Sustainability, Federation University Australia, Mt Helen, Australia
| | - Bhavna Antony
- Institute of Innovation, Science & Sustainability, Federation University Australia, Mt Helen, Australia
| | - Jinger Sun
- Department of Radiation Oncology, University of California, Davis, USA
| | - Orwa Aboud
- Department of Neurology, University of California, Davis, USA; Comprehensive Cancer Center, University of California, Davis, USA; Department of Neurological Surgery, University of California, Davis, USA
| | - Orin Bloch
- Comprehensive Cancer Center, University of California, Davis, USA; Department of Neurological Surgery, University of California, Davis, USA
| | - Megan Daly
- Department of Radiation Oncology, University of California, Davis, USA; Comprehensive Cancer Center, University of California, Davis, USA
| | - Ruben Fragoso
- Department of Radiation Oncology, University of California, Davis, USA; Comprehensive Cancer Center, University of California, Davis, USA
| | - Glenn Yiu
- Department of Ophthalmology & Vision Science, University of California, Davis, USA
| | - Yin Allison Liu
- Department of Neurology, University of California, Davis, USA; Department of Neurological Surgery, University of California, Davis, USA; Department of Ophthalmology & Vision Science, University of California, Davis, USA.
| |
Collapse
|
3
|
Wang J, Yan S. Integration of histone modification-based risk signature with drug sensitivity analysis reveals novel therapeutic strategies for lower-grade glioma. Front Pharmacol 2025; 15:1523779. [PMID: 39872055 PMCID: PMC11770009 DOI: 10.3389/fphar.2024.1523779] [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: 11/06/2024] [Accepted: 12/18/2024] [Indexed: 01/29/2025] Open
Abstract
Background Lower-grade glioma (LGG) exhibits significant heterogeneity in clinical outcomes, and current prognostic markers have limited predictive value. Despite the growing recognition of histone modifications in tumor progression, their role in LGG remains poorly understood. This study aimed to develop a histone modification-based risk signature and investigate its relationship with drug sensitivity to guide personalized treatment strategies. Methods We performed single-cell RNA sequencing analysis on LGG samples (n = 4) to characterize histone modification patterns. Through integrative analysis of TCGA-LGG (n = 513) and CGGA datasets (n = 693 and n = 325), we constructed a histone modification-related risk signature (HMRS) using machine learning approaches. The model's performance was validated in multiple independent cohorts. We further conducted comprehensive analyses of molecular mechanisms, immune microenvironment, and drug sensitivity associated with the risk stratification. Results We identified distinct histone modification patterns across five major cell populations in LGG and developed a robust 20-gene HMRS from 129 candidate genes that effectively stratified patients into high- and low-risk groups with significantly different survival outcomes (training set: AUC = 0.77, 0.73, and 0.71 for 1-, 3-, and 5-year survival; P < 0.001). Integration of HMRS with clinical features further improved prognostic accuracy (C-index >0.70). High-risk tumors showed activation of TGF-β and IL6-JAK-STAT3 signaling pathways, and distinct mutation profiles including TP53 (63% vs 28%), IDH1 (68% vs 85%), and ATRX (46% vs 20%) mutations. The high-risk group demonstrated significantly elevated immune and stromal scores (P < 0.001), with distinct patterns of immune cell infiltration, particularly in memory CD4+ T cells (P < 0.001) and CD8+ T cells (P = 0.001). Drug sensitivity analysis revealed significant differential responses to six therapeutic agents including Temozolomide and targeted drugs (P < 0.05). Conclusion Our study establishes a novel histone modification-based prognostic model that not only accurately predicts LGG patient outcomes but also reveals potential therapeutic targets. The identified associations between risk stratification and drug sensitivity provide valuable insights for personalized treatment strategies. This integrated approach offers a promising framework for improving LGG patient care through molecular-based risk assessment and treatment selection.
Collapse
Affiliation(s)
- Jingyuan Wang
- Department of Neurological Surgery, The First Affiliated Hospital of China Medical University, Shenyang, China
| | - Shuai Yan
- Department of Neurological Function Examination, Affiliated Hospital of Hebei University, Baoding, China
| |
Collapse
|
4
|
Xu H, Liu B, Wang Y, Zhu R, Jiang S, Soliman LAFA, Chai H, Sun M, Chen J, Li KKW, Ng HK, Zhang Z, Wei J, Shi Z, Mao Y. Multi-center real-world data-driven web calculator for predicting outcomes in IDH-mutant gliomas: Integrating molecular subtypes and treatment modalities. Neurooncol Adv 2025; 7:vdae221. [PMID: 39844832 PMCID: PMC11751580 DOI: 10.1093/noajnl/vdae221] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2025] Open
Abstract
Background Isocitrate dehydrogenase (IDH)-mutant gliomas generally have a better prognosis than IDH-wild-type glioblastomas, and the extent of resection significantly impacts prognosis. However, there is a lack of integrated tools for predicting outcomes based on molecular subtypes and treatment modalities. This study aimed to identify factors influencing gross total resection (GTR) rates and to develop a clinical prognostic tool for IDH-mutant gliomas. Methods We analyzed 650 patients with IDH-mutant gliomas from 3 Chinese medical centers (Shanghai, Hong Kong, and Zhengzhou). Data included age, sex, extent of resection, radiotherapy status, tumor grade, histology, and molecular markers (1p19q, TERT promoter, BRAF, EGFR, 10q). Patients were categorized based on GTR status, and a nomogram predicting 3-, 5-, and 10-year overall survival (OS) was developed using Cox proportional hazards regression and validated with time-dependent ROC and calibration plot analyses. Results Non-GTR was associated with diffuse astrocytoma (73.0% vs. 53.5%), 1p19q non-codeletion (67.9% vs. 48.7%), and wildtype TERT promoter (63.6% vs. 52.4%). The nomogram, incorporating age, TERT promoter status, extent of resection, grade, and radiotherapy status, demonstrated strong discriminatory ability (AUC > 0.75) and good calibration. Decision curve analysis indicated that it outperformed WHO grade-based classification in identifying high-risk patients. An online calculator was developed for clinical use (http://www.szflab.site/nomogram/). Conclusion We developed and validated a nomogram and online tool that integrates molecular and clinical factors for predicting outcomes in IDH-mutant gliomas, enhancing clinical decision-making.
Collapse
Affiliation(s)
- Houshi Xu
- Department of Neurosurgery, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai, China
- Research Unit of New Technologies of Micro-Endoscopy Combination in Skull Base Surgery (2018RU008), Chinese Academy of Medical Sciences and Peking Union Medical College (CAMS & PUMC), Shanghai, China
- Department of Neurosurgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College (CAMS & PUMC), Beijing, China
| | - Beining Liu
- Shanghai Medical College, Fudan University, Shanghai, China
- Department of Neurosurgery, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai, China
- Research Unit of New Technologies of Micro-Endoscopy Combination in Skull Base Surgery (2018RU008), Chinese Academy of Medical Sciences and Peking Union Medical College (CAMS & PUMC), Shanghai, China
| | - Yue Wang
- Department of Neurosurgery, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai, China
- Research Unit of New Technologies of Micro-Endoscopy Combination in Skull Base Surgery (2018RU008), Chinese Academy of Medical Sciences and Peking Union Medical College (CAMS & PUMC), Shanghai, China
- Department of Neurosurgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College (CAMS & PUMC), Beijing, China
| | - Ruize Zhu
- Shanghai Medical College, Fudan University, Shanghai, China
- Department of Neurosurgery, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai, China
- Research Unit of New Technologies of Micro-Endoscopy Combination in Skull Base Surgery (2018RU008), Chinese Academy of Medical Sciences and Peking Union Medical College (CAMS & PUMC), Shanghai, China
| | - Shan Jiang
- Shanghai Medical College, Fudan University, Shanghai, China
- Department of Neurosurgery, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai, China
- Research Unit of New Technologies of Micro-Endoscopy Combination in Skull Base Surgery (2018RU008), Chinese Academy of Medical Sciences and Peking Union Medical College (CAMS & PUMC), Shanghai, China
| | | | - Huihui Chai
- Shanghai Medical College, Fudan University, Shanghai, China
- Department of Neurosurgery, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai, China
- Research Unit of New Technologies of Micro-Endoscopy Combination in Skull Base Surgery (2018RU008), Chinese Academy of Medical Sciences and Peking Union Medical College (CAMS & PUMC), Shanghai, China
| | - Maoyuan Sun
- Shanghai Medical College, Fudan University, Shanghai, China
- Department of Neurosurgery, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai, China
- Research Unit of New Technologies of Micro-Endoscopy Combination in Skull Base Surgery (2018RU008), Chinese Academy of Medical Sciences and Peking Union Medical College (CAMS & PUMC), Shanghai, China
| | - Jiawen Chen
- Shanghai Medical College, Fudan University, Shanghai, China
- Department of Neurosurgery, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai, China
- Research Unit of New Technologies of Micro-Endoscopy Combination in Skull Base Surgery (2018RU008), Chinese Academy of Medical Sciences and Peking Union Medical College (CAMS & PUMC), Shanghai, China
| | - Kay Ka-Wai Li
- Department of Anatomical and Cellular Pathology, The Chinese University of Hong Kong, Shatin, Hong Kong SAR, China
| | - Ho-Keung Ng
- Department of Anatomical and Cellular Pathology, The Chinese University of Hong Kong, Shatin, Hong Kong SAR, China
| | - Zhenyu Zhang
- Department of Neurosurgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Junji Wei
- Research Unit of New Technologies of Micro-Endoscopy Combination in Skull Base Surgery (2018RU008), Chinese Academy of Medical Sciences and Peking Union Medical College (CAMS & PUMC), Shanghai, China
- Department of Neurosurgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College (CAMS & PUMC), Beijing, China
| | - Zhifeng Shi
- Shanghai Medical College, Fudan University, Shanghai, China
- Department of Neurosurgery, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai, China
- Research Unit of New Technologies of Micro-Endoscopy Combination in Skull Base Surgery (2018RU008), Chinese Academy of Medical Sciences and Peking Union Medical College (CAMS & PUMC), Shanghai, China
| | - Ying Mao
- Shanghai Medical College, Fudan University, Shanghai, China
- Department of Neurosurgery, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai, China
- Research Unit of New Technologies of Micro-Endoscopy Combination in Skull Base Surgery (2018RU008), Chinese Academy of Medical Sciences and Peking Union Medical College (CAMS & PUMC), Shanghai, China
| |
Collapse
|
5
|
Gu C, Chen X, Wu J, Zhang Y, Zhong L, Luo H, Luo W, Yang F. SOCS1: A potential diagnostic and prognostic marker for aggressive gliomas and a new target for immunotherapy. Medicine (Baltimore) 2024; 103:e40632. [PMID: 39654174 PMCID: PMC11630960 DOI: 10.1097/md.0000000000040632] [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: 09/24/2024] [Revised: 10/31/2024] [Accepted: 11/04/2024] [Indexed: 12/12/2024] Open
Abstract
Gliomas, the most common and deadly cancers of the central nervous system, present a unique immunological barrier that severely undermines the effectiveness of immunotherapies. Suppressor of cytokine signaling 1 (SOCS1), belonging to the SOCS protein family and playing a pivotal role in various cancer treatment strategies and is abundant in high-grade gliomas. This study conducted a comparative analysis of SOCS1 and glioma immune checkpoints. It underscores the feasibility of leveraging SOCS1 as a promising diagnostic and prognostic marker for aggressive gliomas, thus offering novel targets for glioma immunotherapy. Comprehensive gene expression analyses and clinical data validations were performed across multiple databases. The expression and biological functions of SOCS1 were examined through an array of techniques including pan-cancer analysis, functional enrichment, gene set variation analysis, and immune microenvironment examination. This was done alongside a comparison of the similarities between SOCS1 and various glioma immune checkpoints. Utilizing clinical information from patients, a bespoke predictive model was developed to further corroborate the prognostic capabilities of SOCS1. The investigation revealed considerable similarities between SOCS1 and several immune checkpoints such as CTLA4, demonstrating SOCS1's role as an independent prognostic factor positively influencing glioma patient outcomes. The inclusion of SOCS1 in the developed predictive model significantly enhanced its precision. Our findings highlight SOCS1's potential as an innovative target for glioma immunotherapy, providing a novel strategy to overcome the immunological barriers posed by gliomas. Furthermore, identifying SOCS1 as a viable diagnostic marker for aggressive gliomas improves the accuracy of prognostic predictions for affected patients.
Collapse
Affiliation(s)
- Chuanshen Gu
- The Fourth Clinical Medical College of Guangzhou University of Chinese Medicine, Shenzhen, Guangdong, China
- Shenzhen Traditional Chinese Medicine Hospital, Shenzhen, Guangdong, China
| | - Xinyi Chen
- The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| | - Jiayan Wu
- The Fourth Clinical Medical College of Guangzhou University of Chinese Medicine, Shenzhen, Guangdong, China
- Shenzhen Traditional Chinese Medicine Hospital, Shenzhen, Guangdong, China
| | - Yiwen Zhang
- The Fourth Clinical Medical College of Guangzhou University of Chinese Medicine, Shenzhen, Guangdong, China
- Shenzhen Traditional Chinese Medicine Hospital, Shenzhen, Guangdong, China
| | - Linyu Zhong
- The Fourth Clinical Medical College of Guangzhou University of Chinese Medicine, Shenzhen, Guangdong, China
- Shenzhen Traditional Chinese Medicine Hospital, Shenzhen, Guangdong, China
| | - Han Luo
- College of Acupuncture and Tuina, Guangxi University of Chinese Medicine, Nanning, Guangxi, China
| | - Wenshu Luo
- The Fourth Clinical Medical College of Guangzhou University of Chinese Medicine, Shenzhen, Guangdong, China
- Shenzhen Traditional Chinese Medicine Hospital, Shenzhen, Guangdong, China
| | - Fuxia Yang
- The Fourth Clinical Medical College of Guangzhou University of Chinese Medicine, Shenzhen, Guangdong, China
- Shenzhen Traditional Chinese Medicine Hospital, Shenzhen, Guangdong, China
| |
Collapse
|
6
|
Zhang C, Wang J, Niu Z, Zhang K, Wang C, Wang S, Hou S, Yu D, Lin N. Identification of a nomogram predicting overall survival based on ADAP2-related apoptosis genes in gliomas. Int Immunopharmacol 2024; 142:113084. [PMID: 39243555 DOI: 10.1016/j.intimp.2024.113084] [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: 06/25/2024] [Revised: 08/25/2024] [Accepted: 09/02/2024] [Indexed: 09/09/2024]
Abstract
BACKGROUND Apoptosis continues to be a pivotal area of investigation in glioma research. ADAP2 mediates the malignant progression of gliomas through the inhibition of apoptosis and predicts the overall survival(OS) of glioma patients based on prognostic modeling of the apoptotic gene set. METHODS The study encompassed 686 glioma patients, with 413 allocated to the training group and 273 to the validation group. Differential expression of ADAP2 across various glioma subtypes was assessed through bioinformatics analysis and Western blotting. The correlation between ADAP2 and apoptosis was examined using Gene Set Enrichment Analysis (GSEA). Multivariate Cox regression analysis and LASSO dimension reduction analysis were employed to identify apoptosis-related genes with prognostic significance in glioma patients and to construct a nomogram. Biological functions and mechanisms associated with risk scores were explored via Gene Ontology (GO), Kyoto Encyclopedia of Genes and Genomes (KEGG), and GSEA analyses, with validation through Western blotting, flow cytometry, and AM/PI staining. RESULTS ADAP2 was found to be enriched in more aggressive glioma subtypes and was closely linked to glioma cell apoptosis, modulating this process via the NF-κB and P53 signaling pathway. A nomogram for OS in glioma patients was constructed using thirteen apoptosis-related genes. Additionally, ROC curves, calibration curves, and C-indices confirmed the robust applicability of the nomogram. CONCLUSION ADAP2 functions as a prognostic biomarker for glioma patients, regulating glioma cell apoptosis through the NF-κB and P53 signaling pathway. Moreover, prognostic models based on apoptosis-related genes can accurately predict OS for glioma patients at 1, 2, 3, 5, and 10 years.
Collapse
Affiliation(s)
- Chao Zhang
- Department of Neurosurgery, The Affliated Chuzhou Hospital of Anhui Medical University, The First People's Hospital of Chuzhou, Chuzhou 239000, China
| | - Jiajun Wang
- Department of Neurosurgery, The Affliated Chuzhou Hospital of Anhui Medical University, The First People's Hospital of Chuzhou, Chuzhou 239000, China
| | - Zihui Niu
- Department of Neurosurgery, The Affliated Chuzhou Hospital of Anhui Medical University, The First People's Hospital of Chuzhou, Chuzhou 239000, China
| | - Kang Zhang
- Department of Neurosurgery, The Affliated Chuzhou Hospital of Anhui Medical University, The First People's Hospital of Chuzhou, Chuzhou 239000, China
| | - Chengcheng Wang
- Department of Neurosurgery, The Affliated Chuzhou Hospital of Anhui Medical University, The First People's Hospital of Chuzhou, Chuzhou 239000, China
| | - Shuai Wang
- Department of Neurosurgery, The Affliated Chuzhou Hospital of Anhui Medical University, The First People's Hospital of Chuzhou, Chuzhou 239000, China
| | - Shiqiang Hou
- Department of Neurosurgery, The Affliated Chuzhou Hospital of Anhui Medical University, The First People's Hospital of Chuzhou, Chuzhou 239000, China
| | - Dong Yu
- Department of Neurosurgery, The Affliated Chuzhou Hospital of Anhui Medical University, The First People's Hospital of Chuzhou, Chuzhou 239000, China.
| | - Ning Lin
- Department of Neurosurgery, The Affliated Chuzhou Hospital of Anhui Medical University, The First People's Hospital of Chuzhou, Chuzhou 239000, China.
| |
Collapse
|
7
|
Yang P, Jiao X. A Glycolysis and gluconeogenesis-related model for breast cancer prognosis. Cancer Biomark 2024; 41:18758592241296278. [PMID: 40095490 DOI: 10.1177/18758592241296278] [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] [Indexed: 03/19/2025]
Abstract
BackgroundBreast cancer is a malignant tumor with high morbidity and mortality, which seriously endangers the health of women around the world. Biomarker-based exploration will be effective for better diagnosis, prediction and targeted therapy.ObjectiveTo construct biomarker models related to glycolysis and gluconeogenesis in breast cancer.MethodsThe gene expression of 932 breast cancer patients in the Cancer Genome Atlas (TCGA) database was analyzed by Gene Set Variation Analysis (GSVA) using glycolysis and gluconeogenesis-related pathways. Differential expression genes were searched for by the T-test. Univariate Cox proportional hazards model (COX) regression, Least Absolute Shrinkage and Selection Operator (LASSO) regression, and Multivariate COX regression were used to find clinically significant genes for prognostic survival. After that, the constructed gene signature was externally validated through the Gene Expression Omnibus (GEO). Finally, a nomogram was constructed to predict the survival of patients. In addition, analyzing the role of biomarkers in pan-cancer.ResultsA risk scoring model associated with glycolysis and gluconeogenesis was developed and validated. A nomogram was created to predict 2-, 3-, and 5- survival.ConclusionsThe predictive model accurately predicted the prognosis of breast cancer patients.
Collapse
Affiliation(s)
- Penglu Yang
- College of Artificial Intelligence, Taiyuan University of Technology, Jinzhong, China
| | - Xiong Jiao
- College of Artificial Intelligence, Taiyuan University of Technology, Jinzhong, China
| |
Collapse
|
8
|
Jiao Y, Ye J, Zhao W, Fan Z, Kou Y, Guo S, Chao M, Fan C, Ji P, Liu J, Zhai Y, Wang Y, Wang N, Wang L. Development and validation of a deep learning-based survival prediction model for pediatric glioma patients: A retrospective study using the SEER database and Chinese data. Comput Biol Med 2024; 182:109185. [PMID: 39341114 DOI: 10.1016/j.compbiomed.2024.109185] [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: 01/05/2024] [Revised: 09/20/2024] [Accepted: 09/20/2024] [Indexed: 09/30/2024]
Abstract
OBJECTIVE Develop a time-dependent deep learning model to accurately predict the prognosis of pediatric glioma patients, which can assist clinicians in making precise treatment decisions and reducing patient risk. STUDY DESIGN The study involved pediatric glioma patients from the Surveillance, Epidemiology, and End Results (SEER) Registry (2000-2018) and Tangdu Hospital in China (2010-2018) within specific time frames. For training, we selected two neural network-based algorithms (DeepSurv, neural multi-task logistic regression [N-MTLR]) and one ensemble learning-based algorithm (random survival forest [RSF]). Additionally, a multivariable Cox proportional hazard (CoxPH) model was developed for comparison purposes. The SEER dataset was randomly divided into 80 % for training and 20 % for testing, while the Tangdu Hospital dataset served as an external validation cohort. Super-parameters were fine-tuned through 1000 repeated random searches and 5-fold cross-validation on the training cohort. Model performance was assessed using the concordance index (C-index), Brier score, and Integrated Brier Score (IBS). Furthermore, the accuracy of predicting survival at 1, 3, and 5 years was evaluated using receiver operating characteristic (ROC) curves, calibration curves, and the area under the ROC curves (AUC). The generalization ability of the model was assessed using the C-index of the Tangdu Hospital data, ROC curves for 1, 3, and 5 years, and AUC values. Lastly, decision curve analysis (DCA) curves for 1, 3, and 5-year time frames are provided to assess the net benefits across different models. RESULTS A total of 9532 patients with pediatric glioma were included in this study, comprising 9274 patients from the SEER database and 258 patients from Tangdu Hospital in China. The average age at diagnosis was 9.4 ± 6.2 years, and the average survival time was 96 ± 66 months. Through comprehensive performance comparison, the DeepSurv model demonstrated the highest effectiveness, with a C-index of 0.881 on the training cohort. Furthermore, it exhibited excellent accuracy in predicting the 1-year, 3-year, and 5-year survival rates (AUC: 0.903-0.939). Notably, the DeepSurv model also achieved remarkable performance and accuracy on the Chinese dataset (C-index: 0.782, AUC: 0.761-0.852). Comprehensive analysis of DeepSurv, N-MTLR, and RSF revealed that tumor stage, radiotherapy, histological type, tumor size, chemotherapy, age, and surgical method are all significant factors influencing the prognosis of pediatric glioma. Finally, an online version of the pediatric glioma survival predictor based on the DeepSurv model has been established and can be accessed through https://pediatricglioma-tangdu.streamlit.app. CONCLUSIONS The DeepSurv model exhibits exceptional efficacy in predicting the survival of pediatric glioma patients, demonstrating strong performance in discrimination, calibration, stability, and generalization. By utilizing the online version of the pediatric glioma survival predictor, which is based on the DeepSurv model, clinicians can accurately predict patient survival and offer personalized treatment options.
Collapse
Affiliation(s)
- Yang Jiao
- Department of Neurosurgery, Tangdu Hospital of Air Force Medical University, Xi'an, China
| | - Jianan Ye
- School of Engineering Medicine and School of Biological Science and Medical Engineering, Beihang University, Beijing, China
| | - Wenjian Zhao
- Department of Neurosurgery, Tangdu Hospital of Air Force Medical University, Xi'an, China
| | - Zhicheng Fan
- Department of Neurosurgery, Tangdu Hospital of Air Force Medical University, Xi'an, China
| | - Yunpeng Kou
- Department of Neurosurgery, Tangdu Hospital of Air Force Medical University, Xi'an, China
| | - Shaochun Guo
- Department of Neurosurgery, Tangdu Hospital of Air Force Medical University, Xi'an, China
| | - Min Chao
- Department of Neurosurgery, Tangdu Hospital of Air Force Medical University, Xi'an, China
| | - Chao Fan
- Department of Neurosurgery, Tangdu Hospital of Air Force Medical University, Xi'an, China
| | - Peigang Ji
- Department of Neurosurgery, Tangdu Hospital of Air Force Medical University, Xi'an, China
| | - Jinghui Liu
- Department of Neurosurgery, Tangdu Hospital of Air Force Medical University, Xi'an, China
| | - Yulong Zhai
- Department of Neurosurgery, Tangdu Hospital of Air Force Medical University, Xi'an, China
| | - Yuan Wang
- Department of Neurosurgery, Tangdu Hospital of Air Force Medical University, Xi'an, China
| | - Na Wang
- Department of Neurosurgery, Tangdu Hospital of Air Force Medical University, Xi'an, China
| | - Liang Wang
- Department of Neurosurgery, Tangdu Hospital of Air Force Medical University, Xi'an, China.
| |
Collapse
|
9
|
Hao S, Gao M, Li Q, Shu L, Wang P, Hao G. Machine learning predicts cuproptosis-related lncRNAs and survival in glioma patients. Sci Rep 2024; 14:22323. [PMID: 39333603 PMCID: PMC11437180 DOI: 10.1038/s41598-024-72664-w] [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: 07/05/2024] [Accepted: 09/06/2024] [Indexed: 09/29/2024] Open
Abstract
Gliomas are the most common tumor in the central nervous system in adults, with glioblastoma (GBM) representing the most malignant form, while low-grade glioma (LGG) is a less severe. The prognosis for glioma remains poor even after various treatments, such as chemotherapy and immunotherapy. Cuproptosis is a newly defined form of programmed cell death, distinct from ferroptosis and apoptosis, primarily caused by the accumulation of the copper within cells. In this study, we compared the difference between the expression of cuproptosis-related genes in GBM and LGG, respectively, and conducted further analysis on the enrichment pathways of the exclusive expressed cuproptosis-related mRNAs in GBM and LGG. We established two prediction models for survival status using xgboost and random forest algorithms and applied the ROSE algorithm to balance the dataset to improve model performance.
Collapse
Affiliation(s)
- Shaocai Hao
- Department of Neurosurgery, General Hospital of Ningxia Medical University, Yinchuan, Ningxia, China
- Department of Neurosurgery, The First Dongguan Affiliated Hospital of Guangdong Medical University, Dongguan, Guangdong, China
| | - Maoxiang Gao
- Department of Neurosurgery, General Hospital of Ningxia Medical University, Yinchuan, Ningxia, China
| | - Qin Li
- Department of Medicine, Zhejiang Zhongwei Medical Research Center, Hangzhou, 310018, Zhejiang, China
| | - Lilu Shu
- Department of Medicine, Zhejiang Zhongwei Medical Research Center, Hangzhou, 310018, Zhejiang, China
| | - Peter Wang
- Department of Medicine, Zhejiang Zhongwei Medical Research Center, Hangzhou, 310018, Zhejiang, China.
| | - Guangshan Hao
- Department of Neurosurgery, The First Dongguan Affiliated Hospital of Guangdong Medical University, Dongguan, Guangdong, China.
| |
Collapse
|
10
|
Zhang J, Gao A, Meng X, Li K, Li Q, Zhang X, Fan Z, Rong Y, Zhang H, Yu Z, Zhang X, Liang H. Prediction model for poor short-term prognosis in patients with chronic subdural hematoma after burr hole drainage: a retrospective cohort study. Neurosurg Rev 2024; 47:633. [PMID: 39292301 DOI: 10.1007/s10143-024-02752-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2023] [Revised: 06/26/2024] [Accepted: 08/23/2024] [Indexed: 09/19/2024]
Abstract
Chronic subdural hematoma (CSDH) is a common condition in neurosurgery. With an aging population, there is increasing attention on the prognosis of patients following surgical intervention. We developed a postoperative short-term prognostic prediction model using preoperative clinical indicators, aiming to assist in perioperative medical decision-making and management. The dataset was randomly divided into training and validation cohorts. An mRS score greater than 2 one month after discharge was considered indicative of a poor prognosis. In the training cohort, the least absolute shrinkage and selection operator (LASSO) regression analysis was used for multivariate analysis to identify independent risk factors and construct a prediction nomogram for poor prognosis one month after discharge. The performance of the nomogram was assessed using the Receiver Operating Characteristic (ROC) curve and calibration curve. A Decision Curve Analysis (DCA) was also conducted to determine the net benefit threshold of the prediction model. Among the 505 participants, 18.8% (95/505) had a poor prognosis one month after discharge. The baseline characteristics did not significantly differ between the training cohort and the validation cohort. LASSO regression analysis in the training cohort reduced the predictors to four potential factors. Further multivariate logistic analyses in the training cohort identified four independent predictors: age, admission Glasgow Coma Scale (GCS) score, hemiparesis, and hemoglobin count. These predictors were incorporated into the nomogram prediction model. Internal validation using ROC analysis, calibration curves, and other methods demonstrated a strong correlation between the observed and predicted likelihood of poor prognosis one month after discharge. The visualized nomogram prediction model we developed for short-term postoperative prognosis of chronic subdural hematoma after burr hole drainage aids in predicting short-term outcomes and guiding clinical treatment decisions. Further external validation is needed in the future to confirm its effectiveness.
Collapse
Affiliation(s)
- Jie Zhang
- Department of Neurosurgery, The First Affiliated Hospital of Harbin Medical University, Youzheng Street 23, Nangang District, Harbin, Heilongjiang Province, 150001, P. R. China
- NHC Key Laboratory of Cell Transplantation, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang Province, 150001, P. R. China
| | - Aili Gao
- School of Life Science, Northeast Agricultural University, Harbin, PR China
| | - Xiangyi Meng
- Department of Neurosurgery, The First Affiliated Hospital of Harbin Medical University, Youzheng Street 23, Nangang District, Harbin, Heilongjiang Province, 150001, P. R. China
- NHC Key Laboratory of Cell Transplantation, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang Province, 150001, P. R. China
| | - Kuo Li
- School of Life Science, Northeast Agricultural University, Harbin, PR China
| | - Qi Li
- Department of Neurosurgery, The First Affiliated Hospital of Harbin Medical University, Youzheng Street 23, Nangang District, Harbin, Heilongjiang Province, 150001, P. R. China
- NHC Key Laboratory of Cell Transplantation, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang Province, 150001, P. R. China
| | - Xi Zhang
- Department of Neurosurgery, The First Affiliated Hospital of Harbin Medical University, Youzheng Street 23, Nangang District, Harbin, Heilongjiang Province, 150001, P. R. China
- NHC Key Laboratory of Cell Transplantation, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang Province, 150001, P. R. China
| | - Zhaoxin Fan
- Department of Neurosurgery, The First Affiliated Hospital of Harbin Medical University, Youzheng Street 23, Nangang District, Harbin, Heilongjiang Province, 150001, P. R. China
- NHC Key Laboratory of Cell Transplantation, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang Province, 150001, P. R. China
| | - Yiwei Rong
- Department of Neurosurgery, The First Affiliated Hospital of Harbin Medical University, Youzheng Street 23, Nangang District, Harbin, Heilongjiang Province, 150001, P. R. China
- NHC Key Laboratory of Cell Transplantation, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang Province, 150001, P. R. China
| | - Haopeng Zhang
- Department of Neurosurgery, The First Affiliated Hospital of Harbin Medical University, Youzheng Street 23, Nangang District, Harbin, Heilongjiang Province, 150001, P. R. China
- NHC Key Laboratory of Cell Transplantation, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang Province, 150001, P. R. China
| | - Zhao Yu
- Department of Neurosurgery, The First Affiliated Hospital of Harbin Medical University, Youzheng Street 23, Nangang District, Harbin, Heilongjiang Province, 150001, P. R. China
- NHC Key Laboratory of Cell Transplantation, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang Province, 150001, P. R. China
| | - Xiangtong Zhang
- Department of Neurosurgery, The First Affiliated Hospital of Harbin Medical University, Youzheng Street 23, Nangang District, Harbin, Heilongjiang Province, 150001, P. R. China.
| | - Hongsheng Liang
- Department of Neurosurgery, The First Affiliated Hospital of Harbin Medical University, Youzheng Street 23, Nangang District, Harbin, Heilongjiang Province, 150001, P. R. China.
- NHC Key Laboratory of Cell Transplantation, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang Province, 150001, P. R. China.
| |
Collapse
|
11
|
Wang Y, Suo J, Wang Z, Ran K, Tian Y, Han W, Liu Y, Peng X. The PTPRZ1-MET/STAT3/ISG20 axis in glioma stem-like cells modulates tumor-associated macrophage polarization. Cell Signal 2024; 120:111191. [PMID: 38685521 DOI: 10.1016/j.cellsig.2024.111191] [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: 03/13/2024] [Revised: 04/19/2024] [Accepted: 04/24/2024] [Indexed: 05/02/2024]
Abstract
Recent studies have revealed that PTPRZ1-MET (ZM) fusion plays a pivotal role in the progression of glioma to glioblastoma multiforme (GBM), thus serving as a biomarker to distinguish between primary GBM and secondary GBM (sGBM). However, the mechanisms through which ZM fusion influences this progression remain to be elucidated. GBMs with ZM showed poorer prognoses and greater infiltration of tumor-associated macrophages (TAMs) than those without ZM. Glioma stem-like cells (GSCs) and TAMs play complex roles in glioma recurrence, glioma progression and therapy resistance. In this study, we analyzed RNA-seq data from sGBM patients' glioma tissues with or without ZM fusion, and found that stemness and macrophage markers were more highly expressed in sGBM patients harboring ZM than in those without ZM fusion. ZM enhanced the self-renewal and proliferation of GSCs, thereby accelerating glioma progression. In addition, ZM-positive GSCs facilitated the infiltration of TAMs and drove their polarization toward an immunosuppressive phenotype, which was primarily accomplished through the extracellular secretion of ISG20. Our research identified the MET-STAT3-ISG20 axis within GSCs, thus demonstrating the critical role of ZM in GBM initiation and progression. Our study demonstrated that, in contrast to ZM-positive differentiated glioma cells, ZM-positive GSCs upregulated ISG20 expression through the MET-STAT3-ISG20 axis. The extracellular secretion of ISG20 recruited and induced M2-like polarization in macrophages, thereby promoting tumor progression. Our results reveal a novel mechanism involved in ZM-positive GBM pathogenesis and identify potential therapeutic targets.
Collapse
Affiliation(s)
- Yuxin Wang
- Department of Molecular Biology and Biochemistry, Medical Primate Research Center, Neuroscience Center, Institute of Basic Medical Sciences Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical College, Beijing 100005, China; State Key Laboratory of Common Mechanism Research for Major Diseases, Institute of Basic Medical Sciences Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical College, Beijing 100005, China
| | - Jinghao Suo
- Department of Molecular Biology and Biochemistry, Medical Primate Research Center, Neuroscience Center, Institute of Basic Medical Sciences Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical College, Beijing 100005, China; State Key Laboratory of Common Mechanism Research for Major Diseases, Institute of Basic Medical Sciences Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical College, Beijing 100005, China
| | - Zhixing Wang
- Department of Molecular Biology and Biochemistry, Medical Primate Research Center, Neuroscience Center, Institute of Basic Medical Sciences Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical College, Beijing 100005, China; State Key Laboratory of Common Mechanism Research for Major Diseases, Institute of Basic Medical Sciences Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical College, Beijing 100005, China
| | - Kunnian Ran
- Department of Molecular Biology and Biochemistry, Medical Primate Research Center, Neuroscience Center, Institute of Basic Medical Sciences Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical College, Beijing 100005, China; State Key Laboratory of Common Mechanism Research for Major Diseases, Institute of Basic Medical Sciences Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical College, Beijing 100005, China
| | - Yuan Tian
- Department of Molecular Biology and Biochemistry, Medical Primate Research Center, Neuroscience Center, Institute of Basic Medical Sciences Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical College, Beijing 100005, China; State Key Laboratory of Common Mechanism Research for Major Diseases, Institute of Basic Medical Sciences Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical College, Beijing 100005, China
| | - Wei Han
- Department of Molecular Biology and Biochemistry, Medical Primate Research Center, Neuroscience Center, Institute of Basic Medical Sciences Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical College, Beijing 100005, China; State Key Laboratory of Common Mechanism Research for Major Diseases, Institute of Basic Medical Sciences Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical College, Beijing 100005, China.
| | - Yanwei Liu
- Department of Radiation Oncology, Beijing Tiantan Hospital, Capital Medical University, Beijing 100070, China.
| | - Xiaozhong Peng
- Department of Molecular Biology and Biochemistry, Medical Primate Research Center, Neuroscience Center, Institute of Basic Medical Sciences Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical College, Beijing 100005, China; State Key Laboratory of Respiratory Health and Multimorbidity, Institute of Laboratory Animal Science, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100021, China.
| |
Collapse
|
12
|
Shi S, Liang H, Huang Q, Sun X. Identification of Novel Prognostic Signature of Recurrent Low-Grade Glioma. World Neurosurg 2024:S1878-8750(24)01287-7. [PMID: 39069129 DOI: 10.1016/j.wneu.2024.07.147] [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: 06/03/2024] [Revised: 07/18/2024] [Accepted: 07/20/2024] [Indexed: 07/30/2024]
Abstract
OBJECTIVES The prognosis of patients with recurrent low-grade glioma (rLGG) varies greatly. Some patients can survive >10 years after recurrence, whereas other patients have <1 year of survival. METHODS To identify the related risk factors affecting the prognosis of patients with rLGG, we performed a series of bioinformatics analyses on RNA sequencing data of rLGG based on the Chinese Glioma Genome Altas database. RESULTS We constructed a 12-gene prognostic signature, dividing all the patients with rLGG into high- and low-risk subgroups. The result showed an excellent predictive effect in both the training cohort and the validation cohort using LASSO-Cox regression. Moreover, multivariate Cox analysis identified 4 independent prognostic factors of rLGG; among them, ZCWPW1 is identified as a high-value protective factor. CONCLUSIONS In all, this prognostic model displayed robust predictive capability for the overall survival of patients with rLGG, providing a new monitoring method for rLGG. The 4 independent prognostic factors, especially ZCWPW1, can be potential targets for rLGG, bringing new possibilities for the treatment of patients with rLGG.
Collapse
Affiliation(s)
- Shenbao Shi
- Department of Neurosurgery, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Hui Liang
- Department of Neurosurgery, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Qinhong Huang
- Department of Neurosurgery, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Xinlin Sun
- Department of Neurosurgery, Zhujiang Hospital, Southern Medical University, Guangzhou, China.
| |
Collapse
|
13
|
Huang Q, Liang H, Shi S, Ke Y, Wang J. Identification of TNFAIP6 as a reliable prognostic indicator of low-grade glioma. Heliyon 2024; 10:e33030. [PMID: 38948040 PMCID: PMC11211890 DOI: 10.1016/j.heliyon.2024.e33030] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2024] [Revised: 06/12/2024] [Accepted: 06/13/2024] [Indexed: 07/02/2024] Open
Abstract
Glioma is the most common primary malignant tumor in the brain, characterizing by high disability rate and high recurrence rate. Although low-grade glioma (LGG) has a relative benign biological behavior, the prognosis of LGG patients still varies greatly. Glioma stem cells (GSCs) are considered as the chief offenders of glioma cell proliferation, invasion and resistance to therapies. Our study screened a series of glioma stem cell-related genes (GSCRG) based on mDNAsi and WCGNA, and finally established a reliable single-gene prognostic model through 101 combinations of 10 machine learning methods. Our result suggested that the expression level of TNFAIP6 is negatively correlated with the prognosis of LGG patients, which may be the result of pro-cancer signaling pathways activation and immunosuppression. In general, this study revealed that TNFAIP6 is a robust and valuable prognostic factor in LGG, and may be a new target for LGG treatment.
Collapse
Affiliation(s)
| | | | - Shenbao Shi
- The National Key Clinical Specialty, The Engineering Technology Research Center of Education Ministry of China, Guangdong Provincial Key Laboratory on Brain Function Repair and Regeneration, Department of Neurosurgery, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Yiquan Ke
- The National Key Clinical Specialty, The Engineering Technology Research Center of Education Ministry of China, Guangdong Provincial Key Laboratory on Brain Function Repair and Regeneration, Department of Neurosurgery, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Jihui Wang
- The National Key Clinical Specialty, The Engineering Technology Research Center of Education Ministry of China, Guangdong Provincial Key Laboratory on Brain Function Repair and Regeneration, Department of Neurosurgery, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| |
Collapse
|
14
|
Zhang Y, Gao T, Wu M, Xu Z, Hu H. Value analysis of ITLN1 in the diagnostic and prognostic assessment of colorectal cancer. Transl Cancer Res 2024; 13:2877-2891. [PMID: 38988920 PMCID: PMC11231763 DOI: 10.21037/tcr-24-137] [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: 01/18/2024] [Accepted: 04/28/2024] [Indexed: 07/12/2024]
Abstract
BACKGROUND Colorectal cancer (CRC) remains the leading cause of cancer death worldwide. Less than half of the patients are diagnosed when the cancer is locally advanced. Several studies have shown that intelectin-1 (ITLN1) can serve as a key prognostic and therapeutic target for CRC. The purpose of this study was to investigate the clinical value of ITLN1 in CRC and to analyse its potential as a predictive biomarker for CRC. METHODS Colon adenocarcinoma (COAD) is the main type of CRC. COAD project in The Cancer Genome Atlas (TCGA) database served as the training cohort, and GSE39582 series in the Gene Expression Omnibus (GEO) database served as the external independent validation cohort. First, the difference in the expression level of ITLN1 between COAD tissue and normal tissue was analysed, and the results were verified via immunohistochemistry. The relationship between ITLN1 expression and the prognosis of COAD patients was evaluated via the heatmap and the Kaplan-Meier (KM) curve. The ITLN1 coexpressed gene set obtained by Pearson correlation analysis was used. The prognostic signatures that were significantly correlated with survival status were screened by Cox and least absolute shrinkage and selection operator (LASSO) regression analyses. Finally, a nomogram related to ITLN1 was constructed based on the risk score of the prognostic signature and routine clinicopathological variables. RESULTS ITLN1 is significantly underexpressed in tumour tissues and can be used as a valuable tool to distinguish COAD. The high-expression group of ITLN1 was verified to have a greater survival rate. ITLN1 is significantly associated with a good prognosis in COAD patients. Six candidate genes (ITLN1 and MORC2, SH2D7, LGALS4, ATOH1, and NAT2) were selected for use in the Cox-LASSO regression analysis to calculate the risk score. Finally, a nomogram was constructed with a comprehensive risk score and clinicopathologic factors to successfully predict and verify the 1-year, 3-year, and 5-year survival probability. CONCLUSIONS Our study established ITLN1 as an effective tool for CRC screening, diagnosis, and prognostic assessment, provided a basis for further study of the molecular function of ITLN1, and provided new insights for the mechanistic exploration and treatment of CRC.
Collapse
Affiliation(s)
- Yun Zhang
- Department of Medical Engineering, Wannan Medical College, Wuhu, China
| | - Tianyuan Gao
- Department of Pathology, The Second Affiliated Hospital of Wannan Medical College, Wuhu, China
| | - Min Wu
- Sixteen Inpatient Ward, The Fourth People’s Hospital of Wuhu, Wuhu, China
| | - Zhengyuan Xu
- Department of Medical Engineering, Wannan Medical College, Wuhu, China
| | - Huixian Hu
- Department of Medical Engineering, Wannan Medical College, Wuhu, China
| |
Collapse
|
15
|
Smith R, Sapkota R, Antony B, Sun J, Aboud O, Bloch O, Daly M, Fragoso R, Yiu G, Liu YA. A Novel Predictive Model Utilizing Retinal Microstructural Features for Estimating Survival Outcome in Patients with Glioblastoma. RESEARCH SQUARE 2024:rs.3.rs-4420925. [PMID: 38798600 PMCID: PMC11118691 DOI: 10.21203/rs.3.rs-4420925/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/29/2024]
Abstract
Glioblastoma is a highly aggressive brain tumor with poor prognosis despite surgery and chemoradiation. The visual sequelae of glioblastoma have not been well characterized. This study assessed visual outcomes in glioblastoma patients through neuro-ophthalmic exams, imaging of the retinal microstructures/microvasculature, and perimetry. A total of 19 patients (9 male, 10 female, average age at diagnosis 69 years) were enrolled. Best-corrected visual acuity ranged from 20/20-20/50. Occipital tumors showed worse visual fields than frontal tumors (mean deviation - 14.9 and - 0.23, respectively, p < 0.0001). Those with overall survival (OS) < 15 months demonstrated thinner retinal nerve fiber layer and ganglion cell complex (p < 0.0001) and enlarged foveal avascular zone starting from 4 months post-diagnosis (p = 0.006). There was no significant difference between eyes ipsilateral and contralateral to radiation fields (average doses were 1370 cGy and 1180 cGy, respectively, p = 0.42). A machine learning algorithm using retinal microstructure and visual fields predicted patients with long (≥ 15 months) progression free and overall survival with 78% accuracy. Glioblastoma patients frequently present with visual field defects despite normal visual acuity. Patients with poor survival duration demonstrated significant retinal thinning and decreased microvascular density. A machine learning algorithm predicted survival; further validation is warranted.
Collapse
Affiliation(s)
| | - Ranjit Sapkota
- Institute of Innovation, Science & Sustainability, Federation University Australia, Mt Helen, Australia
| | - Bhavna Antony
- Institute of Innovation, Science & Sustainability, Federation University Australia, Mt Helen, Australia
| | | | - Orwa Aboud
- Department of Neurology, University of California, Davis
| | | | | | | | - Glenn Yiu
- Department of Ophthalmology & Vision Science, University of California, Davis
| | - Yin Allison Liu
- Department of Ophthalmology & Vision Science, University of California, Davis
| |
Collapse
|
16
|
Yang S, Wang X, Huan R, Deng M, Kong Z, Xiong Y, Luo T, Jin Z, Liu J, Chu L, Han G, Zhang J, Tan Y. Machine learning unveils immune-related signature in multicenter glioma studies. iScience 2024; 27:109317. [PMID: 38500821 PMCID: PMC10946333 DOI: 10.1016/j.isci.2024.109317] [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: 09/16/2023] [Revised: 01/11/2024] [Accepted: 02/17/2024] [Indexed: 03/20/2024] Open
Abstract
In glioma molecular subtyping, existing biomarkers are limited, prompting the development of new ones. We present a multicenter study-derived consensus immune-related and prognostic gene signature (CIPS) using an optimal risk score model and 101 algorithms. CIPS, an independent risk factor, showed stable and powerful predictive performance for overall and progression-free survival, surpassing traditional clinical variables. The risk score correlated significantly with the immune microenvironment, indicating potential sensitivity to immunotherapy. High-risk groups exhibited distinct chemotherapy drug sensitivity. Seven signature genes, including IGFBP2 and TNFRSF12A, were validated by qRT-PCR, with higher expression in tumors and prognostic relevance. TNFRSF12A, upregulated in GBM, demonstrated inhibitory effects on glioma cell proliferation, migration, and invasion. CIPS emerges as a robust tool for enhancing individual glioma patient outcomes, while IGFBP2 and TNFRSF12A pose as promising tumor markers and therapeutic targets.
Collapse
Affiliation(s)
- Sha Yang
- Guizhou University Medical College, Guiyang 550025, Guizhou Province, China
| | - Xiang Wang
- Department of Neurosurgery, the Affiliated Hospital of Guizhou Medical University, Guiyang 550004, China
| | - Renzheng Huan
- Department of Neurosurgery, The Second Affiliated Hospital of Chongqing Medical University, Chongqing 400010, China
| | - Mei Deng
- Department of Neurosurgery, Guizhou Provincial People’s Hospital, Guiyang, China
| | - Zhuo Kong
- Department of Neurosurgery, Guizhou Provincial People’s Hospital, Guiyang, China
| | - Yunbiao Xiong
- Department of Neurosurgery, Guizhou Provincial People’s Hospital, Guiyang, China
| | - Tao Luo
- Department of Neurosurgery, Guizhou Provincial People’s Hospital, Guiyang, China
| | - Zheng Jin
- Department of Neurosurgery, Guizhou Provincial People’s Hospital, Guiyang, China
| | - Jian Liu
- Guizhou University Medical College, Guiyang 550025, Guizhou Province, China
- Department of Neurosurgery, Guizhou Provincial People’s Hospital, Guiyang, China
| | - Liangzhao Chu
- Department of Neurosurgery, the Affiliated Hospital of Guizhou Medical University, Guiyang 550004, China
| | - Guoqiang Han
- Department of Neurosurgery, Guizhou Provincial People’s Hospital, Guiyang, China
| | - Jiqin Zhang
- Department of Anesthesiology, Guizhou Provincial People’s Hospital, Guiyang, China
| | - Ying Tan
- Department of Neurosurgery, Guizhou Provincial People’s Hospital, Guiyang, China
| |
Collapse
|
17
|
He X, Zhou H, Huang Q, Li Y. The mitotic cell cycle-associated nomogram predicts overall survival in lung adenocarcinoma. Cancer Med 2023; 12:21519-21530. [PMID: 37930238 PMCID: PMC10726878 DOI: 10.1002/cam4.6676] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2023] [Revised: 09/26/2023] [Accepted: 09/29/2023] [Indexed: 11/07/2023] Open
Abstract
BACKGROUND This study aimed to develop a prognostic model for lung adenocarcinoma (LUAD) associated with mitotic cell cycle. The model will predict the probability of survival at different time points and serve as a reference tool to evaluate the effectiveness of LUAD treatment. METHODS A cohort of 442 patients with LUAD from the gene expression omnibus (GEO) database was randomly divided into a training group (n = 299) and a validation group (n = 99). The least absolute shrinkage and selection operator (LASSO)-COX algorithm was used to reduce the number of predictors based on the clinicopathological and RNA sequencing data to establish mutant characteristics that could predict patient survival. Additionally, gene ontology (GO), Kyoto Encyclopedia of Genes and Genomes (KEGG), gene set variation analysis (GSVA), and gene set enrichment analysis (GSEA) analyses were conducted on the mutant characteristics. The performance of the developed nomogram was evaluated using calibration curves and the C-index. RESULTS The mutant characteristics had prognostic value for LUAD and acted as an independent prognostic factor. The mutant characteristics profile derived from the LASSO-COX algorithm demonstrated a significant association with overall survival in patients with LUAD. Functional annotation based on the mutant score, its involvement in the phase transition of the mitotic cell cycle, and its regulatory processes. The nomogram, which combined the mutant score with clinical factors associated with prognosis, showed robust accuracy in both the training and validation groups. CONCLUSION This study presents the first individualized model that establishes a mutant score for predicting survival in LUAD. This model can be used as a predictive tool for determining 1-, 2-, 3-, and 5-year survival probabilities in patients with LUAD.
Collapse
Affiliation(s)
- Xu He
- Department of Cardio‐Thoracic SurgeryThe First Affiliated Hospital of Guangxi Medical UniversityNanningChina
| | - Huafu Zhou
- Department of Cardio‐Thoracic SurgeryThe First Affiliated Hospital of Guangxi Medical UniversityNanningChina
| | - Qianyu Huang
- Department of Respiratory and Critical Care MedicineThe First Affiliated Hospital of Guangxi Medical UniversityNanningChina
| | - Yue Li
- Department of Cardio‐Thoracic SurgeryThe First Affiliated Hospital of Guangxi Medical UniversityNanningChina
| |
Collapse
|
18
|
Wan Y, Li G, Deng J, Zhu H, Ma X. A gene signature predicting prognosis of patients with lower-grade gliomas receiving temozolomide therapy. Discov Oncol 2023; 14:202. [PMID: 37955724 PMCID: PMC10643648 DOI: 10.1007/s12672-023-00818-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/24/2023] [Accepted: 11/02/2023] [Indexed: 11/14/2023] Open
Abstract
Temozolomide (TMZ) has been used as a first-line therapy against lower-grade gliomas (LGGs) combined with other chemotherapy drugs. However, there has been no reliable index predicting TMZ response of patients with LGGs. In this study, we aim to investigate the relationship between gene expressions and the prognosis of TMZ therapy in LGGs. We integrated transcriptome and clinical data of 171 LGGs from the Chinese Glioma Genome Atlas (CGGA). Consensus LASSO Cox regression was used to identify 14 key genes related to different clinical outcomes under TMZ chemotherapy. We constructed and evaluated a risk score based on the 14 genes. Patients with LGGs of lower risk scores (low-risk group) generally had better survival than those LGGs of higher risk scores (high-risk group), which is independent of clinicopathological factors. High-risk patients showed activation of innate and humoral-type immunity. The prognostic contribution of the risk score was validated in an independent validation cohort of 65 patients. Besides, combined with three independent predictors (grade, IDH1 mutation status, and chr1p19q co-deletion status), we further developed a nomogram to predict the benefit of TMZ treatment in LGGs. Our results indicate that a transcriptome-based index can optimize the treatment strategy for patients with LGGs under TMZ therapy.
Collapse
Affiliation(s)
- Yanzhi Wan
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan, China
- West China School of Medicine, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Guangqi Li
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Junyue Deng
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan, China
- West China School of Medicine, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Hong Zhu
- Department of Medical Oncology, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China.
| | - Xuelei Ma
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan, China.
| |
Collapse
|
19
|
Jin G, Hu W, Zeng L, Ma B, Zhou M. Prediction of long-term mortality in patients with ischemic stroke based on clinical characteristics on the first day of ICU admission: An easy-to-use nomogram. Front Neurol 2023; 14:1148185. [PMID: 37122313 PMCID: PMC10140521 DOI: 10.3389/fneur.2023.1148185] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2023] [Accepted: 03/15/2023] [Indexed: 05/02/2023] Open
Abstract
Background This study aimed to establish and validate an easy-to-use nomogram for predicting long-term mortality among ischemic stroke patients. Methods All raw data were obtained from the Medical Information Mart for Intensive Care IV database. Clinical features associated with long-term mortality (1-year mortality) among ischemic stroke patients were identified using least absolute shrinkage and selection operator regression. Then, binary logistic regression was used to construct a nomogram, the discrimination of which was evaluated by the concordance index (C-index), integrated discrimination improvement (IDI), and net reclassification index (NRI). Finally, a calibration curve and decision curve analysis (DCA) were employed to study calibration and net clinical benefit, compared to the Glasgow Coma Scale (GCS) and the commonly used disease severity scoring system. Results Patients who were identified with ischemic stroke were randomly assigned into developing (n = 1,443) and verification (n = 646) cohorts. The following factors were associated with 1-year mortality among ischemic stroke patients, including age on ICU admission, marital status, underlying dementia, underlying malignant cancer, underlying metastatic solid tumor, heart rate, respiratory rate, oxygen saturation, white blood cells, anion gap, mannitol injection, invasive mechanical ventilation, and GCS. The construction of the nomogram was based on the abovementioned features. The C-index of the nomogram in the developing and verification cohorts was 0.820 and 0.816, respectively. Compared with GCS and the commonly used disease severity scoring system, the IDI and NRI of the constructed nomogram had a statistically positive improvement in predicting long-term mortality in both developing and verification cohorts (all with p < 0.001). The actual mortality was consistent with the predicted mortality in the developing (p = 0.862) and verification (p = 0.568) cohorts. Our nomogram exhibited greater net clinical benefit than GCS and the commonly used disease severity scoring system. Conclusion This proposed nomogram has good performance in predicting long-term mortality among ischemic stroke patients.
Collapse
Affiliation(s)
- Guangyong Jin
- Department of Critical Care Medicine, Affiliated Hangzhou First People’s Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Wei Hu
- Department of Critical Care Medicine, Affiliated Hangzhou First People’s Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Longhuan Zeng
- Department of Critical Care Medicine, Affiliated Hangzhou First People’s Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Buqing Ma
- Department of Critical Care Medicine, Affiliated Hangzhou First People’s Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Menglu Zhou
- Department of Neurology, Affiliated Hospital of Hangzhou Normal University, Hangzhou, China
- *Correspondence: Menglu Zhou,
| |
Collapse
|
20
|
Li G, Wu F, Zeng F, Zhai Y, Feng Y, Chang Y, Wang D, Jiang T, Zhang W. A novel DNA repair-related nomogram predicts survival in low-grade gliomas. CNS Neurosci Ther 2020; 27:186-195. [PMID: 33063446 PMCID: PMC7816205 DOI: 10.1111/cns.13464] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Revised: 08/20/2020] [Accepted: 09/26/2020] [Indexed: 12/17/2022] Open
Abstract
Aims We aimed to create a tumor recurrent‐based prediction model to predict recurrence and survival in patients with low‐grade glioma. Methods This study enrolled 291 patients (188 in the training group and 103 in the validation group) with clinicopathological information and transcriptome sequencing data. LASSO‐COX algorithm was applied to shrink predictive factor size and build a predictive recurrent signature. GO, KEGG, and GSVA analyses were performed for function annotations of the recurrent signature. The calibration curves and C‐Index were assessed to evaluate the nomogram's performance. Results This study found that DNA repair functions of tumor cells were significantly enriched in recurrent low‐grade gliomas. A predictive recurrent signature, built by the LASSO‐COX algorithm, was significantly associated with overall survival and progression‐free survival in low‐grade gliomas. Moreover, function annotations analysis of the predictive recurrent signature exhibited that the signature was associated with DNA repair functions. The nomogram, combining the predictive recurrent signature and clinical prognostic predictors, showed powerful prognostic ability in the training and validation groups. Conclusion An individualized prediction model was created to predict 1‐, 2‐, 3‐, 5‐, and 10‐year survival and recurrent rate of patients with low‐grade glioma, which may serve as a potential tool to guide postoperative individualized care.
Collapse
Affiliation(s)
- Guanzhang Li
- Department of Molecular Neuropathology, Beijing Neurosurgical Institute, Capital Medical University, Beijing, China
| | - Fan Wu
- Department of Molecular Neuropathology, Beijing Neurosurgical Institute, Capital Medical University, Beijing, China
| | - Fan Zeng
- Department of Molecular Neuropathology, Beijing Neurosurgical Institute, Capital Medical University, Beijing, China
| | - You Zhai
- Department of Molecular Neuropathology, Beijing Neurosurgical Institute, Capital Medical University, Beijing, China
| | - Yuemei Feng
- Department of Molecular Neuropathology, Beijing Neurosurgical Institute, Capital Medical University, Beijing, China
| | - Yuanhao Chang
- Department of Molecular Neuropathology, Beijing Neurosurgical Institute, Capital Medical University, Beijing, China
| | - Di Wang
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Tao Jiang
- Department of Molecular Neuropathology, Beijing Neurosurgical Institute, Capital Medical University, Beijing, China.,Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China.,Center of Brain Tumor, Beijing Institute for Brain Disorders, Beijing, China.,China National Clinical Research Center for Neurological Diseases, Beijing, China.,Chinese Glioma Genome Atlas Network (CGGA), Asian Glioma Genome Atlas Network (AGGA)
| | - Wei Zhang
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China.,China National Clinical Research Center for Neurological Diseases, Beijing, China.,Chinese Glioma Genome Atlas Network (CGGA), Asian Glioma Genome Atlas Network (AGGA)
| |
Collapse
|