1
|
Xiao Y, He S, Xie B, Zhao W, Ji D. Unveiling the impact of cell death-related genes and immune dynamics on drug resistance in lung adenocarcinoma: a risk score model and functional insights. Discov Oncol 2024; 15:441. [PMID: 39269650 PMCID: PMC11399484 DOI: 10.1007/s12672-024-01336-y] [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: 08/13/2024] [Accepted: 09/11/2024] [Indexed: 09/15/2024] Open
Abstract
Lung adenocarcinoma (LUAD), characterized by its heterogeneity and complex pathogenesis, is the focus of this study which investigates the association between cell death-related genes and LUAD. Through machine learning, a risk score model was developed using the Coxboost rsf algorithm, demonstrating strong prognostic accuracy in both validation (GSE30219, GSE31210, GSE72094) and training (TCGA-LUAD) datasets with C-indices of 0.93, 0.67, 0.68, and 0.64, respectively. The study reveals that the expression of Keratin 18 (KRT18), a key cytoskeletal protein, varies across LUAD cell lines (DV-90, PC-9, A549) compared to normal bronchial epithelial cells (BEAS-2B), suggesting its potential role in LUAD's pathogenesis. Kaplan-Meier survival curves further validate the model, indicating longer survival in the low-risk group. A comprehensive analysis of gene expression, functional differences, immune infiltration, and mutations underscores significant variations between risk groups, highlighting the high-risk group's immunological dysfunction. This points to a more intricate tumor immune environment and the possibility of alternative therapeutic strategies. The study also delves into drug sensitivity, showing distinct responses between risk groups, underscoring the importance of risk stratification in treatment decisions for LUAD patients. Additionally, it explores KRT18's epigenetic regulation and its correlation with immune cell infiltration and immune regulatory molecules, suggesting KRT18's significant role in the tumor immune landscape. This research not only offers a valuable prognostic tool for LUAD but also illuminates the complex interplay between cell death-related genes, drug sensitivity, and immune infiltration, positioning KRT18 as a potential therapeutic or prognostic target to improve patient outcomes by personalizing LUAD treatment strategies.
Collapse
Affiliation(s)
- Yuanyuan Xiao
- Department of Critical Care Medicine, The Fifth People's Hospital of Ganzhou City, Ganzhou, 341000, China
- Ganzhou Institute of Liver Disease, Ganzhou, 341000, China
| | - Shancheng He
- Department of Critical Care Medicine, The Fifth People's Hospital of Ganzhou City, Ganzhou, 341000, China
- Ganzhou Institute of Liver Disease, Ganzhou, 341000, China
| | - Baochang Xie
- Department of Critical Care Medicine, The Fifth People's Hospital of Ganzhou City, Ganzhou, 341000, China
- Ganzhou Institute of Liver Disease, Ganzhou, 341000, China
| | - Wenqi Zhao
- Department of Critical Care Medicine, The Fifth People's Hospital of Ganzhou City, Ganzhou, 341000, China
- Ganzhou Institute of Liver Disease, Ganzhou, 341000, China
| | - Dengliang Ji
- Department of Critical Care Medicine, The Fifth People's Hospital of Ganzhou City, Ganzhou, 341000, China.
- Ganzhou Institute of Liver Disease, Ganzhou, 341000, China.
| |
Collapse
|
2
|
Styliara EI, Astrakas LG, Alexiou G, Xydis VG, Zikou A, Kafritsas G, Voulgaris S, Argyropoulou MI. Survival Outcome Prediction in Glioblastoma: Insights from MRI Radiomics. Curr Oncol 2024; 31:2233-2243. [PMID: 38668068 PMCID: PMC11048751 DOI: 10.3390/curroncol31040165] [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: 03/09/2024] [Revised: 04/10/2024] [Accepted: 04/12/2024] [Indexed: 04/28/2024] Open
Abstract
Background: Extracting multiregional radiomic features from multiparametric MRI for predicting pretreatment survival in isocitrate dehydrogenase (IDH) wild-type glioblastoma (GBM) patients is a promising approach. Methods: MRI data from 49 IDH wild-type glioblastoma patients pre-treatment were utilized. Diffusion and perfusion maps were generated, and tumor subregions segmented. Radiomic features were extracted for each tissue type and map. Feature selection on 1862 radiomic features identified 25 significant features. The Cox proportional-hazards model with LASSO regularization was used to perform survival analysis. Internal and external validation used a 38-patient training cohort and an 11-patient validation cohort. Statistical significance was set at p < 0.05. Results: Age and six radiomic features (shape and first and second order) from T1W, diffusion, and perfusion maps contributed to the final model. Findings suggest that a small necrotic subregion, inhomogeneous vascularization in the solid non-enhancing subregion, and edema-related tissue damage in the enhancing and edema subregions are linked to poor survival. The model's C-Index was 0.66 (95% C.I. 0.54-0.80). External validation demonstrated good accuracy (AUC > 0.65) at all time points. Conclusions: Radiomics analysis, utilizing segmented perfusion and diffusion maps, provide predictive indicators of survival in IDH wild-type glioblastoma patients, revealing associations with microstructural and vascular heterogeneity in the tumor.
Collapse
Affiliation(s)
- Effrosyni I. Styliara
- Department of Radiology, Faculty of Medicine, University of Ioannina, 45110 Ioannina, Greece; (E.I.S.); (V.G.X.); (A.Z.); (M.I.A.)
| | - Loukas G. Astrakas
- Medical Physics Lab, Faculty of Medicine, University of Ioannina, 45110 Ioannina, Greece;
| | - George Alexiou
- Department of Neurosurgery, Faculty of Medicine, University of Ioannina, 45110 Ioannina, Greece; (G.K.); (S.V.)
| | - Vasileios G. Xydis
- Department of Radiology, Faculty of Medicine, University of Ioannina, 45110 Ioannina, Greece; (E.I.S.); (V.G.X.); (A.Z.); (M.I.A.)
| | - Anastasia Zikou
- Department of Radiology, Faculty of Medicine, University of Ioannina, 45110 Ioannina, Greece; (E.I.S.); (V.G.X.); (A.Z.); (M.I.A.)
| | - Georgios Kafritsas
- Department of Neurosurgery, Faculty of Medicine, University of Ioannina, 45110 Ioannina, Greece; (G.K.); (S.V.)
| | - Spyridon Voulgaris
- Department of Neurosurgery, Faculty of Medicine, University of Ioannina, 45110 Ioannina, Greece; (G.K.); (S.V.)
| | - Maria I. Argyropoulou
- Department of Radiology, Faculty of Medicine, University of Ioannina, 45110 Ioannina, Greece; (E.I.S.); (V.G.X.); (A.Z.); (M.I.A.)
| |
Collapse
|
3
|
Obrador E, Moreno-Murciano P, Oriol-Caballo M, López-Blanch R, Pineda B, Gutiérrez-Arroyo JL, Loras A, Gonzalez-Bonet LG, Martinez-Cadenas C, Estrela JM, Marqués-Torrejón MÁ. Glioblastoma Therapy: Past, Present and Future. Int J Mol Sci 2024; 25:2529. [PMID: 38473776 PMCID: PMC10931797 DOI: 10.3390/ijms25052529] [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: 12/23/2023] [Revised: 02/10/2024] [Accepted: 02/16/2024] [Indexed: 03/14/2024] Open
Abstract
Glioblastoma (GB) stands out as the most prevalent and lethal form of brain cancer. Although great efforts have been made by clinicians and researchers, no significant improvement in survival has been achieved since the Stupp protocol became the standard of care (SOC) in 2005. Despite multimodality treatments, recurrence is almost universal with survival rates under 2 years after diagnosis. Here, we discuss the recent progress in our understanding of GB pathophysiology, in particular, the importance of glioma stem cells (GSCs), the tumor microenvironment conditions, and epigenetic mechanisms involved in GB growth, aggressiveness and recurrence. The discussion on therapeutic strategies first covers the SOC treatment and targeted therapies that have been shown to interfere with different signaling pathways (pRB/CDK4/RB1/P16ink4, TP53/MDM2/P14arf, PI3k/Akt-PTEN, RAS/RAF/MEK, PARP) involved in GB tumorigenesis, pathophysiology, and treatment resistance acquisition. Below, we analyze several immunotherapeutic approaches (i.e., checkpoint inhibitors, vaccines, CAR-modified NK or T cells, oncolytic virotherapy) that have been used in an attempt to enhance the immune response against GB, and thereby avoid recidivism or increase survival of GB patients. Finally, we present treatment attempts made using nanotherapies (nanometric structures having active anti-GB agents such as antibodies, chemotherapeutic/anti-angiogenic drugs or sensitizers, radionuclides, and molecules that target GB cellular receptors or open the blood-brain barrier) and non-ionizing energies (laser interstitial thermal therapy, high/low intensity focused ultrasounds, photodynamic/sonodynamic therapies and electroporation). The aim of this review is to discuss the advances and limitations of the current therapies and to present novel approaches that are under development or following clinical trials.
Collapse
Affiliation(s)
- Elena Obrador
- Scientia BioTech S.L., 46002 Valencia, Spain; (P.M.-M.); (M.O.-C.); (R.L.-B.); (J.M.E.)
- Department of Physiology, Faculty of Medicine and Odontology, University of Valencia, 46010 Valencia, Spain;
| | - Paz Moreno-Murciano
- Scientia BioTech S.L., 46002 Valencia, Spain; (P.M.-M.); (M.O.-C.); (R.L.-B.); (J.M.E.)
| | - María Oriol-Caballo
- Scientia BioTech S.L., 46002 Valencia, Spain; (P.M.-M.); (M.O.-C.); (R.L.-B.); (J.M.E.)
- Department of Physiology, Faculty of Medicine and Odontology, University of Valencia, 46010 Valencia, Spain;
| | - Rafael López-Blanch
- Scientia BioTech S.L., 46002 Valencia, Spain; (P.M.-M.); (M.O.-C.); (R.L.-B.); (J.M.E.)
- Department of Physiology, Faculty of Medicine and Odontology, University of Valencia, 46010 Valencia, Spain;
| | - Begoña Pineda
- Department of Physiology, Faculty of Medicine and Odontology, University of Valencia, 46010 Valencia, Spain;
| | - Julia Lara Gutiérrez-Arroyo
- Department of Medicine, Jaume I University of Castellon, 12071 Castellon, Spain; (J.L.G.-A.); (A.L.); (C.M.-C.)
| | - Alba Loras
- Department of Medicine, Jaume I University of Castellon, 12071 Castellon, Spain; (J.L.G.-A.); (A.L.); (C.M.-C.)
| | - Luis G. Gonzalez-Bonet
- Department of Neurosurgery, Castellon General University Hospital, 12004 Castellon, Spain;
| | - Conrado Martinez-Cadenas
- Department of Medicine, Jaume I University of Castellon, 12071 Castellon, Spain; (J.L.G.-A.); (A.L.); (C.M.-C.)
| | - José M. Estrela
- Scientia BioTech S.L., 46002 Valencia, Spain; (P.M.-M.); (M.O.-C.); (R.L.-B.); (J.M.E.)
- Department of Physiology, Faculty of Medicine and Odontology, University of Valencia, 46010 Valencia, Spain;
- Department of Physiology, Faculty of Pharmacy, University of Valencia, 46100 Burjassot, Spain
| | | |
Collapse
|
4
|
Chen L, Chen R, Li T, Huang L, Tang C, Li Y, Zeng Z. MRI radiomics model for predicting TERT promoter mutation status in glioblastoma. Brain Behav 2023; 13:e3324. [PMID: 38054695 PMCID: PMC10726789 DOI: 10.1002/brb3.3324] [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: 08/22/2023] [Revised: 09/05/2023] [Accepted: 10/31/2023] [Indexed: 12/07/2023] Open
Abstract
BACKGROUND AND PURPOSE The presence of TERT promoter mutations has been associated with worse prognosis and resistance to therapy for patients with glioblastoma (GBM). This study aimed to determine whether the combination model of different feature selections and classification algorithms based on multiparameter MRI can be used to predict TERT subtype in GBM patients. METHODS A total of 143 patients were included in our retrospective study, and 2553 features were obtained. The datasets were randomly divided into training and test sets in a ratio of 7:3. The synthetic minority oversampling technique was used to achieve data balance. The Pearson correlation coefficients were used for dimension reduction. Three feature selections and five classification algorithms were used to model the selected features. Finally, 10-fold cross validation was applied to the training dataset. RESULTS A model with eight features generated by recursive feature elimination (RFE) and linear discriminant analysis (LDA) showed the greatest diagnostic performance (area under the curve values for the training, validation, and testing sets: 0.983, 0.964, and 0.926, respectively), followed by relief and random forest (RF), analysis of variance and RF. Furthermore, the relief was the optimal feature selection for separately evaluating those five classification algorithms, and RF was the most preferable algorithm for separately assessing the three feature selectors. ADC entropy was the parameter that made the greatest contribution to the discrimination of TERT mutations. CONCLUSIONS Radiomics model generated by RFE and LDA mainly based on ADC entropy showed good performance in predicting TERT promoter mutations in GBM.
Collapse
Affiliation(s)
- Ling Chen
- Department of RadiologyThe First Affiliated Hospital of Guangxi Medical UniversityNanningGuangxiChina
- Department of RadiologyLiuzhou Worker's HospitalThe Fourth Affiliated HospitalGuangxi Medical UniversityNanningGuangxiChina
| | - Runrong Chen
- Department of RadiologyThe First Affiliated Hospital of Guangxi Medical UniversityNanningGuangxiChina
| | - Tao Li
- Department of RadiologyLiuzhou Worker's HospitalThe Fourth Affiliated HospitalGuangxi Medical UniversityNanningGuangxiChina
| | - Lizhao Huang
- Department of RadiologyLiuzhou Worker's HospitalThe Fourth Affiliated HospitalGuangxi Medical UniversityNanningGuangxiChina
| | - Chuyun Tang
- Department of RadiologyThe First Affiliated Hospital of Guangxi Medical UniversityNanningGuangxiChina
| | - Yao Li
- Department of NeurosurgeryLiuzhou Worker's HospitalThe Fourth Affiliated HospitalGuangxi Medical UniversityNanningGuangxiChina
| | - Zisan Zeng
- Department of RadiologyThe First Affiliated Hospital of Guangxi Medical UniversityNanningGuangxiChina
| |
Collapse
|
5
|
Ge M, Zhu Y, Wei M, Piao H, He M. Improving the efficacy of anti-EGFR drugs in GBM: Where we are going? Biochim Biophys Acta Rev Cancer 2023; 1878:188996. [PMID: 37805108 DOI: 10.1016/j.bbcan.2023.188996] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2023] [Revised: 09/11/2023] [Accepted: 09/29/2023] [Indexed: 10/09/2023]
Abstract
The therapies targeting mutations of driver genes in cancer have advanced into clinical trials for a variety of tumors. In glioblastoma (GBM), epidermal growth factor receptor (EGFR) is the most commonly mutated oncogene, and targeting EGFR has been widely investigated as a promising direction. However, the results of EGFR pathway inhibitors have not been satisfactory. Limited blood-brain barrier (BBB) permeability, drug resistance, and pathway compensation mechanisms contribute to the failure of anti-EGFR therapies. This review summarizes recent research advances in EGFR-targeted therapy for GBM and provides insight into the reasons for the unsatisfactory results of EGFR-targeted therapy. By combining the results of preclinical studies with those of clinical trials, we discuss that improved drug penetration across the BBB, the use of multi-target combinations, and the development of peptidomimetic drugs under the premise of precision medicine may be promising strategies to overcome drug resistance in GBM.
Collapse
Affiliation(s)
- Manxi Ge
- Department of Pharmacology, School of Pharmacy, China Medical University, Shenyang, China; Liaoning Key Laboratory of Molecular Targeted Anti-Tumor Drug Development and Evaluation, Liaoning Cancer Immune Peptide Drug Engineering Technology Research Center, Shenyang, China
| | - Yan Zhu
- Department of Pharmacology, School of Pharmacy, China Medical University, Shenyang, China; Liaoning Key Laboratory of Molecular Targeted Anti-Tumor Drug Development and Evaluation, Liaoning Cancer Immune Peptide Drug Engineering Technology Research Center, Shenyang, China
| | - Minjie Wei
- Department of Pharmacology, School of Pharmacy, China Medical University, Shenyang, China; Liaoning Key Laboratory of Molecular Targeted Anti-Tumor Drug Development and Evaluation, Liaoning Cancer Immune Peptide Drug Engineering Technology Research Center, Shenyang, China; Liaoning Medical Diagnosis and Treatment Center, Shenyang, China.
| | - Haozhe Piao
- Department of Neurosurgery, Liaoning Cancer Hospital and Institute, Cancer Hospital of China Medical University, Shenyang, China.
| | - Miao He
- Department of Pharmacology, School of Pharmacy, China Medical University, Shenyang, China; Liaoning Key Laboratory of Molecular Targeted Anti-Tumor Drug Development and Evaluation, Liaoning Cancer Immune Peptide Drug Engineering Technology Research Center, Shenyang, China.
| |
Collapse
|
6
|
Wang J, Liu Y, Liu F, Gan S, Roy S, Hasan I, Zhang B, Guo B. Emerging extracellular vesicle-based carriers for glioblastoma diagnosis and therapy. NANOSCALE 2023. [PMID: 37337814 DOI: 10.1039/d3nr01667f] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/21/2023]
Abstract
Glioblastoma (GBM) treatment is still a big clinical challenge because of its highly malignant, invasive, and lethal characteristics. After treatment with the conventional therapeutic paradigm of surgery combined with radio- and chemotherapy, patients bearing GBMs generally exhibit a poor prognosis, with high mortality and a high disability rate. The main reason is the existence of the formidable blood-brain barrier (BBB), aggressive growth, and the infiltration nature of GBMs. Especially, the BBB suppresses the delivery of imaging and therapeutic agents to lesion sites, and thus this leads to difficulties in achieving a timely diagnosis and treatment. Recent studies have demonstrated that extracellular vesicles (EVs) exhibit favorable merits including good biocompatibility, a strong drug loading capacity, long circulation time, good BBB crossing efficiency, specific targeting to lesion sites, and high efficiency in the delivery of a variety of cargos for GBM therapy. Importantly, EVs inherit physiological and pathological molecules from the source cells, which are ideal biomarkers for molecularly tracking the malignant progression of GBMs. Herein, we start by introducing the pathophysiology and physiology of GBMs, followed by presenting the biological functions of EVs in GBMs with a special focus on their role as biomarkers for GBM diagnosis and as messengers in the modulation of the GBM microenvironment. Furthermore, we provide an update on the recent progress of using EVs in biology, functionality, and isolation applications. More importantly, we systematically summarize the most recent advances of EV-based carriers for GBM therapy by delivering different drugs including gene/RNA-based drugs, chemotherapy drugs, imaging agents, and combinatory drugs. Lastly, we point out the challenges and prospects of future research on EVs for diagnosing and treating GBMs. We hope this review will stimulate interest from researchers with different backgrounds and expedite the progress of GBM treatment paradigms.
Collapse
Affiliation(s)
- Jingjing Wang
- Shenzhen Key Laboratory of Advanced Functional Carbon Materials Research and Comprehensive Application, Shenzhen Key Laboratory of Flexible Printed Electronics Technology, and School of Science, Harbin Institute of Technology, Shenzhen 518055, China.
- Shenzhen Key Laboratory of Flexible Printed Electronics Technology, Harbin Institute of Technology, Shenzhen 518055, China
| | - Yue Liu
- Shenzhen Key Laboratory of Advanced Functional Carbon Materials Research and Comprehensive Application, Shenzhen Key Laboratory of Flexible Printed Electronics Technology, and School of Science, Harbin Institute of Technology, Shenzhen 518055, China.
- Shenzhen Key Laboratory of Flexible Printed Electronics Technology, Harbin Institute of Technology, Shenzhen 518055, China
| | - Fengbo Liu
- Shenzhen Key Laboratory of Advanced Functional Carbon Materials Research and Comprehensive Application, Shenzhen Key Laboratory of Flexible Printed Electronics Technology, and School of Science, Harbin Institute of Technology, Shenzhen 518055, China.
- Shenzhen Key Laboratory of Flexible Printed Electronics Technology, Harbin Institute of Technology, Shenzhen 518055, China
| | - Shaoyan Gan
- Shenzhen Key Laboratory of Advanced Functional Carbon Materials Research and Comprehensive Application, Shenzhen Key Laboratory of Flexible Printed Electronics Technology, and School of Science, Harbin Institute of Technology, Shenzhen 518055, China.
- Shenzhen Key Laboratory of Flexible Printed Electronics Technology, Harbin Institute of Technology, Shenzhen 518055, China
| | - Shubham Roy
- Shenzhen Key Laboratory of Advanced Functional Carbon Materials Research and Comprehensive Application, Shenzhen Key Laboratory of Flexible Printed Electronics Technology, and School of Science, Harbin Institute of Technology, Shenzhen 518055, China.
- Shenzhen Key Laboratory of Flexible Printed Electronics Technology, Harbin Institute of Technology, Shenzhen 518055, China
| | - Ikram Hasan
- Shenzhen Key Laboratory of Advanced Functional Carbon Materials Research and Comprehensive Application, Shenzhen Key Laboratory of Flexible Printed Electronics Technology, and School of Science, Harbin Institute of Technology, Shenzhen 518055, China.
- Shenzhen Key Laboratory of Flexible Printed Electronics Technology, Harbin Institute of Technology, Shenzhen 518055, China
| | - Baozhu Zhang
- Department of Oncology, People's Hospital of Shenzhen Baoan District, The Second Affiliated Hospital of Shenzhen University, Shenzhen, Guangdong 518055, China.
| | - Bing Guo
- Shenzhen Key Laboratory of Advanced Functional Carbon Materials Research and Comprehensive Application, Shenzhen Key Laboratory of Flexible Printed Electronics Technology, and School of Science, Harbin Institute of Technology, Shenzhen 518055, China.
- Shenzhen Key Laboratory of Flexible Printed Electronics Technology, Harbin Institute of Technology, Shenzhen 518055, China
| |
Collapse
|
7
|
Wang Y, Li J, Cao Y, Chen W, Xing H, Guo X, Shi Y, Wang Y, Liang T, Ye L, Liu D, Yang T, Wang Y, Ma W. Characteristic analysis and identification of novel molecular biomarkers in elderly glioblastoma patients using the 2021 WHO Classification of Central Nervous System Tumors. Front Neurosci 2023; 17:1165823. [PMID: 37360159 PMCID: PMC10288210 DOI: 10.3389/fnins.2023.1165823] [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: 02/14/2023] [Accepted: 05/19/2023] [Indexed: 06/28/2023] Open
Abstract
Introduction Elderly glioblastoma (GBM) patients is characterized by high incidence and poor prognosis. Currently, however, there is still a lack of adequate molecular characterization of elderly GBM patients. The fifth edition of the WHO Classification of Central Nervous System Tumors (WHO5) gives a new classification approach for GBM, and the molecular characteristics of elderly GBM patients need to be investigated under this new framework. Methods The clinical and radiological features of patients with different classifications and different ages were compared. Potential prognostic molecular markers in elderly GBM patients under the WHO5 classification were found using Univariate Cox regression and Kaplan-Meier survival analysis. Results A total of 226 patients were included in the study. The prognostic differences between younger and elderly GBM patients were more pronounced under the WHO5 classification. Neurological impairment was more common in elderly patients (p = 0.001), while intracranial hypertension (p = 0.034) and epilepsy (p = 0.038) were more common in younger patients. Elderly patients were more likely to have higher Ki-67(p = 0.013), and in elderly WHO5 GBM patients, KMT5B (p = 0.082), KRAS (p = 0.1) and PPM1D (p = 0.055) were each associated with overall survival (OS). Among them, KRAS and PPM1D were found to be prognostic features unique to WHO5 elderly GBM patients. Conclusion Our study demonstrates that WHO5 classification can better distinguish the prognosis of elderly and younger GBM. Furthermore, KRAS and PPM1D may be potential prognostic predictors in WHO5 elderly GBM patients. The specific mechanism of these two genes in elderly GBM remains to be further studied.
Collapse
Affiliation(s)
- Yaning Wang
- Department of Neurosurgery, Center for Malignant Brain Tumors, National Glioma MDT Alliance, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Junlin Li
- Department of Neurosurgery, Center for Malignant Brain Tumors, National Glioma MDT Alliance, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
- Eight-Year Medical Doctor Program, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yaning Cao
- Department of Neurosurgery, Center for Malignant Brain Tumors, National Glioma MDT Alliance, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
- Eight-Year Medical Doctor Program, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Wenlin Chen
- Department of Neurosurgery, Center for Malignant Brain Tumors, National Glioma MDT Alliance, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Hao Xing
- Department of Neurosurgery, Center for Malignant Brain Tumors, National Glioma MDT Alliance, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Xiaopeng Guo
- Department of Neurosurgery, Center for Malignant Brain Tumors, National Glioma MDT Alliance, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
- China Anti-Cancer Association Specialty Committee of Glioma, Beijing, China
| | - Yixin Shi
- Department of Neurosurgery, Center for Malignant Brain Tumors, National Glioma MDT Alliance, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
- Eight-Year Medical Doctor Program, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yuekun Wang
- Department of Neurosurgery, Center for Malignant Brain Tumors, National Glioma MDT Alliance, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Tingyu Liang
- Department of Neurosurgery, Center for Malignant Brain Tumors, National Glioma MDT Alliance, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Liguo Ye
- Department of Neurosurgery, Center for Malignant Brain Tumors, National Glioma MDT Alliance, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Delin Liu
- Department of Neurosurgery, Center for Malignant Brain Tumors, National Glioma MDT Alliance, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
- Eight-Year Medical Doctor Program, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Tianrui Yang
- Department of Neurosurgery, Center for Malignant Brain Tumors, National Glioma MDT Alliance, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
- Eight-Year Medical Doctor Program, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yu Wang
- Department of Neurosurgery, Center for Malignant Brain Tumors, National Glioma MDT Alliance, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
- China Anti-Cancer Association Specialty Committee of Glioma, Beijing, China
| | - Wenbin Ma
- Department of Neurosurgery, Center for Malignant Brain Tumors, National Glioma MDT Alliance, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
- China Anti-Cancer Association Specialty Committee of Glioma, Beijing, China
| |
Collapse
|
8
|
Gatto L, Franceschi E, Tosoni A, Di Nunno V, Bartolini S, Brandes AA. Glioblastoma treatment slowly moves toward change: novel druggable targets and translational horizons in 2022. Expert Opin Drug Discov 2023; 18:269-286. [PMID: 36718723 DOI: 10.1080/17460441.2023.2174097] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
INTRODUCTION Glioblastoma (GBM) is the most common primary brain tumor in adults. GBM treatment options have been the same for the past 30 years and have only modestly extended survival, despite aggressive multimodal treatments. The progressively better knowledge of GBM biology and a comprehensive analysis of its genomic profile have elucidated GBM heterogeneity, contributing to a more effective molecular classification and to the development of innovative targeted therapeutic approaches. AREAS COVERED This article reports all the noteworthy innovations for immunotherapy and targeted therapy, providing insights into the current advances in trial designs, including combination therapies with immuno-oncology agents and target combinations. EXPERT OPINION GBM molecular heterogeneity and brain anatomical characteristics critically restrain drug effectiveness. Nevertheless, stimulating insights for future research and drug development come from innovative treatment strategies for GBM, such as multi-specific 'off-the-shelf' CAR-T therapy, oncolytic viral therapy and autologous dendritic cell vaccination. Disappointing results from targeted therapies-clinical trials are mainly due to complex interferences between signaling pathways and biological processes leading to drug resistance: hence, it is imperative in the future to develop combinatorial approaches and multimodal therapies.
Collapse
Affiliation(s)
- Lidia Gatto
- Department of Oncology, AUSL Bologna, Bologna, Italy
| | - Enrico Franceschi
- Nervous System Medical Oncology Department, IRCCS Istituto Delle Scienze Neurologiche Di Bologna, Bologna, Italy
| | - Alicia Tosoni
- Nervous System Medical Oncology Department, IRCCS Istituto Delle Scienze Neurologiche Di Bologna, Bologna, Italy
| | | | - Stefania Bartolini
- Nervous System Medical Oncology Department, IRCCS Istituto Delle Scienze Neurologiche Di Bologna, Bologna, Italy
| | - Alba Ariela Brandes
- Nervous System Medical Oncology Department, IRCCS Istituto Delle Scienze Neurologiche Di Bologna, Bologna, Italy
| |
Collapse
|
9
|
Makowska M, Smolarz B, Romanowicz H. microRNAs (miRNAs) in Glioblastoma Multiforme (GBM)-Recent Literature Review. Int J Mol Sci 2023; 24:3521. [PMID: 36834933 PMCID: PMC9965735 DOI: 10.3390/ijms24043521] [Citation(s) in RCA: 21] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2022] [Revised: 01/25/2023] [Accepted: 02/08/2023] [Indexed: 02/12/2023] Open
Abstract
Glioblastoma multiforme (GBM) is the most common, malignant, poorly promising primary brain tumor. GBM is characterized by an infiltrating growth nature, abundant vascularization, and a rapid and aggressive clinical course. For many years, the standard treatment of gliomas has invariably been surgical treatment supported by radio- and chemotherapy. Due to the location and significant resistance of gliomas to conventional therapies, the prognosis of glioblastoma patients is very poor and the cure rate is low. The search for new therapy targets and effective therapeutic tools for cancer treatment is a current challenge for medicine and science. microRNAs (miRNAs) play a key role in many cellular processes, such as growth, differentiation, cell division, apoptosis, and cell signaling. Their discovery was a breakthrough in the diagnosis and prognosis of many diseases. Understanding the structure of miRNAs may contribute to the understanding of the mechanisms of cellular regulation dependent on miRNA and the pathogenesis of diseases underlying these short non-coding RNAs, including glial brain tumors. This paper provides a detailed review of the latest reports on the relationship between changes in the expression of individual microRNAs and the formation and development of gliomas. The use of miRNAs in the treatment of this cancer is also discussed.
Collapse
Affiliation(s)
- Marianna Makowska
- Department of Anesthesiology and Operative Intensive Care Medicine, Charité–Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Augustenburger Platz 1, 13353 Berlin, Germany
| | - Beata Smolarz
- Laboratory of Cancer Genetics, Department of Pathology, Polish Mother’s Memorial Hospital Research Institute, Rzgowska 281/289, 93-338 Lodz, Poland
| | - Hanna Romanowicz
- Laboratory of Cancer Genetics, Department of Pathology, Polish Mother’s Memorial Hospital Research Institute, Rzgowska 281/289, 93-338 Lodz, Poland
| |
Collapse
|
10
|
Ma Y, Wang Y, Nie C, Lin Y. The efficacy of targeted therapy combined with radiotherapy and temozolomide-based chemotherapy in the treatment of glioma: A systemic review and meta-analysis of phase II/III randomized controlled trials. Front Oncol 2023; 13:1082539. [PMID: 36776303 PMCID: PMC9909217 DOI: 10.3389/fonc.2023.1082539] [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: 10/28/2022] [Accepted: 01/04/2023] [Indexed: 01/27/2023] Open
Abstract
Background Glioma is the most common intracranial tumor, accounting for about half of the primary intracranial tumors, with the characteristics of hidden onset and high mortality. Even after surgery, radiotherapy and chemotherapy, the prognosis of glioma is not ideal. Targeted therapy has developed rapidly in the treatment of other malignant tumors, which is also an important direction in the research and development of new therapies for glioma. So far, targeting combined with radiotherapy and chemotherapy have been used as the treatment of glioma in many clinical trials, but the role of targeted combined radiotherapy and chemotherapy in the treatment of glioma is still controversial. The purpose of this study was to evaluate the efficacy of targeted therapy combined with radiotherapy and temozolomide (TMZ)-based chemotherapy in the treatment of glioma. Methods Phase II or phase III clinical trials involving targeted therapy combined with radiotherapy and chemotherapy and temozolomide-based radiotherapy and chemotherapy for gliomas were searched using PubMed, Embase and Web of Science databases, and a comprehensive meta-analysis was conducted. The primary outcome was overall survival time (OS) and progression-free survival time (PFS), and the secondary outcome was adverse reaction. The time-to-event data is summarized as hazard ratio (HR), and the binary results are summarized as odds ratio (OR). Two researchers conducted literature screening, data extraction and quality evaluation according to inclusion and exclusion criteria. Stata16.0 software was used for analysis, random effect model was used for data merging, and forest map was used for display. Results A total of 11 eligible literatures and 12 prospective randomized controlled clinical trials of 1284 cases were included in the meta-analysis. The results showed that compared with radiotherapy and chemotherapy alone, targeted drugs combined with temozolomide-based radiotherapy and chemotherapy could significantly improve OS in phase II trial, but there was no improvement in Phase III trial, and PFS of newly diagnosed glioma patients was improved (HR=0.82(0.71-0.94) 95%CI, p =0.005). The PFS of the third phase of the experiment also improved. Compared with radiotherapy and chemotherapy alone, there was no statistically significant increase in adverse events in targeted combined radiotherapy and chemotherapy group. Systematic review registration https://www.crd.york.ac.uk/prospero, identifier CRD42022326012.
Collapse
Affiliation(s)
- Yifan Ma
- Department of Neurology, The Second Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, China
| | - Yue Wang
- Department of Medical Oncology, The Second Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, China
| | - Chen Nie
- Department of Neurology, The Second Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, China,*Correspondence: Chen Nie, ; Yongzhong Lin,
| | - Yongzhong Lin
- Department of Neurology, The Second Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, China,*Correspondence: Chen Nie, ; Yongzhong Lin,
| |
Collapse
|
11
|
Cheng YW, Chen YY, Lin CJ, Chen YT, Lieu AS, Tsai HP, Kwan AL. High expression of NLRP12 predicts poor prognosis in patients with intracranial glioma. J Chin Med Assoc 2023; 86:88-97. [PMID: 36599143 DOI: 10.1097/jcma.0000000000000830] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
BACKGROUND Intracranial gliomas are the most common primary central nervous system tumors in humans, and glioblastoma multiforme is the most malignant intracranial glioma. The nucleotide-binding domain leucine-rich repeat (NLR)-containing family are crucial regulators of inflammatory and innate immune responses. NLRP12 codes for the monarch-1 protein, which regulates immune responses in humans. Data from a next-generation sequencing database indicated that NLRP12 expression is increased in glioma cells. However, the relationship between NLRP12 levels and gliomas is unclear. METHODS To explore the role of NLRP12-related translation factors and proteins in glioma, we evaluated the clinical data and paraffin sections from glioma patients. The expression of NLRP12 was evaluated using immunohistochemical analysis, and clinical parameters were analyzed using chi-square and Kaplan-Meier survival tests. RESULTS The degree of malignancy and prognosis highly correlated with NLRP12 levels. In addition, the siRNA-mediated downregulation of NLRP12 in glioma cell lines decreased proliferation, invasion, and migration. The levels of VEGF, N-cadherin, and cyclin D1 were downregulated after knockdown of NRLP12 in glioma cell lines, as observed using western blotting in vitro. Knockdown of NLRP12 attenuated the tumor progression in vivo. CONCLUSION The expression of NLRP12 may be an independent prognostic factor and a potential target for the treatment of intracranial glioma.
Collapse
Affiliation(s)
- Yu-Wen Cheng
- Department of Neurosurgery, Kaohsiung Veterans General Hospital, Kaohsiung, Taiwan, ROC
- Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan, ROC
| | - Yang-Yi Chen
- Department of Dermatology, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung, Taiwan, ROC
| | - Chien-Ju Lin
- School of Pharmacy, College of Pharmacy, Kaohsiung Medical University, Kaohsiung, Taiwan, ROC
| | - Yi-Ting Chen
- Department of Pathology, Kaohsiung Medical University Hospital, Kaohsiung, Taiwan, ROC
- Department of Pathology, School of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan, ROC
| | - Ann-Shung Lieu
- Division of Neurosurgery, Department of Surgery, Kaohsiung Medical University Hospital, Kaohsiung, Taiwan, ROC
- Department of Surgery, School of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan, ROC
| | - Hung-Pei Tsai
- Division of Neurosurgery, Department of Surgery, Kaohsiung Medical University Hospital, Kaohsiung, Taiwan, ROC
| | - Aij-Lie Kwan
- Division of Neurosurgery, Department of Surgery, Kaohsiung Medical University Hospital, Kaohsiung, Taiwan, ROC
- Department of Surgery, School of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan, ROC
- Department of Neurosurgery, University of Virginia, Charlottesville, VA, USA
| |
Collapse
|
12
|
Rom M, Schott R, Pencreac'h E, Cébula H, Cox D, Bender L, Antoni D, Lhermitte B, Noel G. [Impact of NGS results on patient outcome with a multiform glioblastoma]. Cancer Radiother 2022; 26:987-993. [PMID: 35715358 DOI: 10.1016/j.canrad.2022.01.002] [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: 10/25/2021] [Revised: 01/01/2022] [Accepted: 01/17/2022] [Indexed: 11/18/2022]
Abstract
PURPOSE Although some genetic alterations in glioblastoma (GBM) have been characterized, the prognostic value of these gene mutations is not yet established in patients treated with standard therapy. PATIENTS AND METHOD 40 patients with newly diagnosed GBM, treated between July 2017 and December 2019, and who had genomic analysis were analyzed. Next-generation sequencing techniques (NGS) were used with a panel of 26 genes. Patients were grouped according to MGMT status, the presence or absence of at least one mutated gene on the panel, and p53 expression by immunohistochemistry. RESULTS the median follow-up was 11.5 months (1.0-37). For all patients, the median duration of progression-free survival was 8 months (95% CI, 5.3-10.7) and the median overall survival (OS) was 17 months (95% CI, 7.5-26.5). Progression-free and overall survival were significantly different according to MGMT status but not according to NGS and p53 status. Three groups of patients according to different combined status could be distinguished due to significant differences in overall survival. CONCLUSION we have shown that the presence of MGMT promoter methylation is a good prognostic factor. By grouping the patients according to their MGMT, NGS and p53 status, three groups of patients could be separated according to their overall survival. However, these results must be confirmed on a larger number of patients.
Collapse
Affiliation(s)
- M Rom
- ICANS-service de radiothérapie, Institut du Cancer Strasbourg-Europe, 17, rue Albert Calmette, 67033 Strasbourg, France; Service de radiothérapie - Hôpital Calmette, No. 3, Monivong Bvld, Sangkat Sras Chok, Khan Daun Penh, Phnom Penh, Royaume du Cambodge
| | - R Schott
- ICANS-service d'oncologie médicale, Institut du Cancer Strasbourg-Europe, 17, rue Albert Calmette, 67033 Strasbourg, France
| | - E Pencreac'h
- Service de biologie, CHU Hautepierre, 1, rue Molière, 67200 Strasbourg, France
| | - H Cébula
- Service de neurochirurgie - CHU Hautepierre, 1, rue Molière, 67200 Strasbourg, France
| | - D Cox
- IRFAC, Inserm U1113, 3, avenue Molière, 67000 Strasbourg, France; Research, Development in Precision Medicine, Institut de Cancérologie Strasbourg Europe (ICANS), 17, rue Albert Calmette, 67200 Strasbourg, France
| | - L Bender
- ICANS-service d'oncologie médicale, Institut du Cancer Strasbourg-Europe, 17, rue Albert Calmette, 67033 Strasbourg, France
| | - D Antoni
- ICANS-service de radiothérapie, Institut du Cancer Strasbourg-Europe, 17, rue Albert Calmette, 67033 Strasbourg, France
| | - B Lhermitte
- Service d'anatomopathologie, CHU Hautepierre, 1, rue Molière, 67200 Strasbourg, France
| | - G Noel
- ICANS-service de radiothérapie, Institut du Cancer Strasbourg-Europe, 17, rue Albert Calmette, 67033 Strasbourg, France.
| |
Collapse
|
13
|
Cocola C, Magnaghi V, Abeni E, Pelucchi P, Martino V, Vilardo L, Piscitelli E, Consiglio A, Grillo G, Mosca E, Gualtierotti R, Mazzaccaro D, La Sala G, Di Pietro C, Palizban M, Liuni S, DePedro G, Morara S, Nano G, Kehler J, Greve B, Noghero A, Marazziti D, Bussolino F, Bellipanni G, D'Agnano I, Götte M, Zucchi I, Reinbold R. Transmembrane Protein TMEM230, a Target of Glioblastoma Therapy. Front Cell Neurosci 2021; 15:703431. [PMID: 34867197 PMCID: PMC8636015 DOI: 10.3389/fncel.2021.703431] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Accepted: 10/12/2021] [Indexed: 11/13/2022] Open
Abstract
Glioblastomas (GBM) are the most aggressive tumors originating in the brain. Histopathologic features include circuitous, disorganized, and highly permeable blood vessels with intermittent blood flow. These features contribute to the inability to direct therapeutic agents to tumor cells. Known targets for anti-angiogenic therapies provide minimal or no effect in overall survival of 12–15 months following diagnosis. Identification of novel targets therefore remains an important goal for effective treatment of highly vascularized tumors such as GBM. We previously demonstrated in zebrafish that a balanced level of expression of the transmembrane protein TMEM230/C20ORF30 was required to maintain normal blood vessel structural integrity and promote proper vessel network formation. To investigate whether TMEM230 has a role in the pathogenesis of GBM, we analyzed its prognostic value in patient tumor gene expression datasets and performed cell functional analysis. TMEM230 was found necessary for growth of U87-MG cells, a model of human GBM. Downregulation of TMEM230 resulted in loss of U87 migration, substratum adhesion, and re-passaging capacity. Conditioned media from U87 expressing endogenous TMEM230 induced sprouting and tubule-like structure formation of HUVECs. Moreover, TMEM230 promoted vascular mimicry-like behavior of U87 cells. Gene expression analysis of 702 patients identified that TMEM230 expression levels distinguished high from low grade gliomas. Transcriptomic analysis of patients with gliomas revealed molecular pathways consistent with properties observed in U87 cell assays. Within low grade gliomas, elevated TMEM230 expression levels correlated with reduced overall survival independent from tumor subtype. Highest level of TMEM230 correlated with glioblastoma and ATP-dependent microtubule kinesin motor activity, providing a direction for future therapeutic intervention. Our studies support that TMEM230 has both glial tumor and endothelial cell intracellular and extracellular functions. Elevated levels of TMEM230 promote glial tumor cell migration, extracellular scaffold remodeling, and hypervascularization and abnormal formation of blood vessels. Downregulation of TMEM230 expression may inhibit both low grade glioma and glioblastoma tumor progression and promote normalization of abnormally formed blood vessels. TMEM230 therefore is both a promising anticancer and antiangiogenic therapeutic target for inhibiting GBM tumor cells and tumor-driven angiogenesis.
Collapse
Affiliation(s)
- Cinzia Cocola
- Institute for Biomedical Technologies, National Research Council, Milan, Italy.,Consorzio Italbiotec, Milan, Italy
| | - Valerio Magnaghi
- Department of Pharmacological and Biomolecular Sciences, University of Milan, Milan, Italy
| | - Edoardo Abeni
- Institute for Biomedical Technologies, National Research Council, Milan, Italy
| | - Paride Pelucchi
- Institute for Biomedical Technologies, National Research Council, Milan, Italy
| | - Valentina Martino
- Institute for Biomedical Technologies, National Research Council, Milan, Italy
| | - Laura Vilardo
- Institute for Biomedical Technologies, National Research Council, Milan, Italy
| | - Eleonora Piscitelli
- Institute for Biomedical Technologies, National Research Council, Milan, Italy
| | - Arianna Consiglio
- Institute for Biomedical Technologies, National Research Council, Milan, Italy
| | - Giorgio Grillo
- Institute for Biomedical Technologies, National Research Council, Milan, Italy
| | - Ettore Mosca
- Institute for Biomedical Technologies, National Research Council, Milan, Italy
| | - Roberta Gualtierotti
- Department of Pathophysiology and Transplantation, Università degli Studi di Milano, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Daniela Mazzaccaro
- Operative Unit of Vascular Surgery, IRCCS Policlinico San Donato, San Donato Milanese, Italy
| | - Gina La Sala
- Institute of Biochemistry and Cell Biology, Italian National Research Council, Rome, Italy
| | - Chiara Di Pietro
- Institute of Biochemistry and Cell Biology, Italian National Research Council, Rome, Italy
| | - Mira Palizban
- Department of Gynecology and Obstetrics, University Hospital of Münster, Münster, Germany
| | - Sabino Liuni
- Institute for Biomedical Technologies, National Research Council, Milan, Italy
| | - Giuseppina DePedro
- Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
| | | | - Giovanni Nano
- Operative Unit of Vascular Surgery, IRCCS Policlinico San Donato, San Donato Milanese, Italy.,Department of Biomedical Sciences for Health, Università degli Studi di Milano, Milan, Italy
| | - James Kehler
- National Institutes of Health, NIDDK, Laboratory of Cell and Molecular Biology, Bethesda, MD, United States
| | - Burkhard Greve
- Department of Radiation Therapy and Radiation Oncology, University Hospital of Münster, Münster, Germany
| | - Alessio Noghero
- Lovelace Biomedical Research Institute, Albuquerque, NM, United States.,Department of Oncology, University of Turin, Orbassano, Italy
| | - Daniela Marazziti
- Institute of Biochemistry and Cell Biology, Italian National Research Council, Rome, Italy
| | - Federico Bussolino
- Department of Oncology, University of Turin, Orbassano, Italy.,Laboratory of Vascular Oncology Candiolo Cancer Institute - IRCCS, Candiolo, Italy
| | - Gianfranco Bellipanni
- Department of Biology, Center for Biotechnology, Sbarro Institute for Cancer Research and Molecular Medicine, Temple University, Philadelphia, PA, United States
| | - Igea D'Agnano
- Institute for Biomedical Technologies, National Research Council, Milan, Italy
| | - Martin Götte
- Department of Gynecology and Obstetrics, University Hospital of Münster, Münster, Germany
| | - Ileana Zucchi
- Institute for Biomedical Technologies, National Research Council, Milan, Italy
| | - Rolland Reinbold
- Institute for Biomedical Technologies, National Research Council, Milan, Italy
| |
Collapse
|
14
|
MiR-137 Targets the 3' Untranslated Region of MSH2: Potential Implications in Lynch Syndrome-Related Colorectal Cancer. Cancers (Basel) 2021; 13:cancers13184662. [PMID: 34572889 PMCID: PMC8470766 DOI: 10.3390/cancers13184662] [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: 08/05/2021] [Revised: 09/13/2021] [Accepted: 09/15/2021] [Indexed: 11/17/2022] Open
Abstract
Mismatch Repair (MMR) gene dysregulation plays a fundamental role in Lynch Syndrome (LS) pathogenesis, a form of hereditary colorectal cancer. Loss or overexpression of key MMR genes leads to genome instability and tumorigenesis; however, the mechanisms controlling MMR gene expression are unknown. One such gene, MSH2, exerts an important role, not only in MMR, but also in cell proliferation, apoptosis, and cell cycle control. In this study, we explored the functions and underlying molecular mechanisms of increased MSH2 expression related to a c.*226A>G variant in the 3'untranslated (UTR) region of MSH2 that had been previously identified in a subject clinically suspected of LS. Bioinformatics identified a putative binding site for miR-137 in this region. To verify miRNA targeting specificity, we performed luciferase gene reporter assays using a MSH2 3'UTR psiCHECK-2 vector in human SW480 cells over-expressing miR-137, which showed a drastic reduction in luciferase activity (p > 0.0001). This effect was abolished by site-directed mutagenesis of the putative miR-137 seed site. Moreover, in these cells we observed that miR-137 levels were inversely correlated with MSH2 expression levels. These results were confirmed by results in normal and tumoral tissues from the patient carrying the 3'UTR c.*226A>G variant in MSH2. Finally, miR-137 overexpression in SW480 cells significantly suppressed cell proliferation in a time- and dose-dependent manner (p < 0.0001), supporting a role for MSH2 in apoptosis and cell proliferation processes. Our findings suggest miR-137 helps control MSH2 expression via its 3'UTR and that dysregulation of this mechanism appears to promote tumorigenesis in colon cells.
Collapse
|
15
|
Yang Y, Chu L, Zeng Z, Xu S, Yang H, Zhang X, Jia J, Long N, Hu Y, Liu J. Four specific biomarkers associated with the progression of glioblastoma multiforme in older adults identified using weighted gene co-expression network analysis. Bioengineered 2021; 12:6643-6654. [PMID: 34516348 PMCID: PMC8806527 DOI: 10.1080/21655979.2021.1975980] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022] Open
Abstract
Glioblastoma multiforme (GBM) is the most common primary intracranial malignancy in adults. Owing to individual tolerance and tumor heterogeneity, the therapy methods for young adults do not apply to older adults. The present study aimed to identify specific biomarkers for GBM in older adults using weighted gene co-expression network analysis (WGCNA). Gene expression profiles of older adults with GBM were downloaded from The Cancer Genome Atlas (TCGA) and set as a discovery cohort to construct WGCNA. Core genes of clinically significant modules were used to perform functional enrichment, protein-protein interaction, and Pearson correlation analyses. Gene expression profiles of young in TCGA and older GBM patients from our research group were set as verification cohorts for hub gene expression and diagnostic value. Four significant gene modules associated clinically with older adults with GBM were identified, whereas 251 genes were core genes with module membership>0.8 and gene significance>0.2. Ermin (ERMN), myelin-associated oligodendrocyte basic protein (MOBP), proteolipid protein 1 (PLP1), and oligodendrocytic myelin paranodal and inner loop protein (OPALIN) genes had significant relationships with the Karnofsky score (KPS) in older GBM patients. ERMN, MOBP, PLP1, and OPALIN had no relationship with KPS in young GBM patients. These genes were upregulated in GBM tissues from older patients with low but not high KPS and had high diagnostic value. In conclusion, ERMN, MOBP, PLP1, and OPALIN may serve as specific biomarkers for the progression of GBM in older adults.
Collapse
Affiliation(s)
- Yushi Yang
- College of Basic Medicine, Guizhou Medical University, Guiyang, Guizhou, China.,Department of Pathology, Guizhou Medical University, Guiyang, Guizhou, China
| | - Liangzhao Chu
- Department of Cerebral Surgery, The Affiliated Hospital of Guizhou Medical University, Guiyang, Guizhou, China
| | - Zhirui Zeng
- College of Basic Medicine, Guizhou Medical University, Guiyang, Guizhou, China
| | - Shu Xu
- Department of Pathology, Guizhou Medical University, Guiyang, Guizhou, China
| | - Hua Yang
- Department of Cerebral Surgery, The Affiliated Hospital of Guizhou Medical University, Guiyang, Guizhou, China
| | - Xuelin Zhang
- Department of Physical Examination Center, The Affiliated Hospital of Guizhou Medical University, Guiyang, Guizhou, China
| | - Jun Jia
- College of Clinical Medicine, Guizhou Medical University, Guiyang, Guizhou, China
| | - Niya Long
- Department of Cerebral Surgery, The Affiliated Hospital of Guizhou Medical University, Guiyang, Guizhou, China
| | - Yaxin Hu
- Department of Prenatal Diagnosis, The Affiliated Hospital of Guizhou Medical University, Guiyang, Guizhou, China
| | - Jian Liu
- College of Basic Medicine, Guizhou Medical University, Guiyang, Guizhou, China.,Department of Cerebral Surgery, Guizhou Provincial People's Hospital, Guiyang, Guizhou, China
| |
Collapse
|
16
|
Sfifou F, Hakkou EM, Bouaiti ELA, Slaoui M, Errihani H, Al Bouzidi A, Abouqal R, El Ouahabi A, Cherradi N. Correlation of immunohistochemical expression of HIF-1alpha and IDH1 with clinicopathological and therapeutic data of moroccan glioblastoma and survival analysis. Ann Med Surg (Lond) 2021; 69:102731. [PMID: 34466221 PMCID: PMC8384773 DOI: 10.1016/j.amsu.2021.102731] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2021] [Revised: 08/15/2021] [Accepted: 08/15/2021] [Indexed: 11/27/2022] Open
Abstract
INTRODUCTION Glioblastomas are aggressive primary intracranial tumours of the central nervous system causing significant mortality and morbidity worldwide. OBJECTIVE This study aims to evaluate the prognostic value of tissue expression by immunostaining of hypoxia-inducible factor (HIF-1α), isocitrate dehydrogenase 1 (IDH1), and tumour protein p53 in glioblastoma in Moroccan patients. The association of HIF-1α, IDH1, and p53 expression with the clinicopathological data and overall patient survival (OS) was also evaluated. MATERIALS AND METHODS Confirmed glioblastomas were included in this study. Twenty-two tissue samples were obtained by neurosurgical intervention resulting from total resection, and subtotal resection or biopsy. Karnofsky index, histological type of tumour, and the status of IDH1, p53 protein, and HIF-1α expression by immunostaining were reported. RESULTS The majority of the patients were males (64%) with a sex ratio of 1.75. The average age was 54 ± 13. Median follow-up was 10.10 months and median overall survival was 10 months. The expression of HIF-1α was high in 10 samples (45%) and low in 12 (55%). There was a statistically significant difference in OS of 85% at 12 months for the subgroup of patients "HIF-1α negative IDH1 positive" p = 0.038, the unadjusted analysis showed that the group "HIF-1α positive, IDH1 positive" was a poor prognostic factor, the HR was 0.08 (95% CI: 0.009-0.756, p = 0.027). CONCLUSION Patients with negative HIF-1α expression and positive IDH1 expression have a better prognosis, suggesting that these two biomarkers may be useful in the search for new approaches for targeted therapy in glioblastoma.
Collapse
Affiliation(s)
- Fatima Sfifou
- Research's Pedagogic Unit of Pathological Anatomy, Laboratory of Pathological Anatomy. Research Team in Tumour Pathology, Faculty of Medicine and Pharmacy, Mohammed V University in Rabat, Morocco
- Pathological Anatomy Department, Hospital of Specialities in Rabat, Morocco
| | - El Mehdi Hakkou
- Neurosurgery Department, Hospital of Specialities in Rabat, Morocco
| | - EL Arbi Bouaiti
- Laboratory of Biostatistics, Clinical Research and Epidemiology, Rabat Faculty of Medicine and Pharmacy, Mohammed V University in Rabat, Morocco
| | - Meriem Slaoui
- Research's Pedagogic Unit of Pathological Anatomy, Laboratory of Pathological Anatomy. Research Team in Tumour Pathology, Faculty of Medicine and Pharmacy, Mohammed V University in Rabat, Morocco
| | - Hassan Errihani
- National Oncology Centre Sidi Mohamed Ben Abdallah in Rabat, Morocco
| | - Abderrahmane Al Bouzidi
- Research's Pedagogic Unit of Pathological Anatomy, Laboratory of Pathological Anatomy. Research Team in Tumour Pathology, Faculty of Medicine and Pharmacy, Mohammed V University in Rabat, Morocco
| | - Redouane Abouqal
- Laboratory of Biostatistics, Clinical Research and Epidemiology, Rabat Faculty of Medicine and Pharmacy, Mohammed V University in Rabat, Morocco
| | | | - Nadia Cherradi
- Research's Pedagogic Unit of Pathological Anatomy, Laboratory of Pathological Anatomy. Research Team in Tumour Pathology, Faculty of Medicine and Pharmacy, Mohammed V University in Rabat, Morocco
- Pathological Anatomy Department, Hospital of Specialities in Rabat, Morocco
| |
Collapse
|
17
|
Park Y, Park J, Ahn JW, Sim JM, Kang SJ, Kim S, Hwang SJ, Han SH, Sung KS, Lim J. Transcriptomic Landscape of Lower Grade Glioma Based on Age-Related Non-Silent Somatic Mutations. ACTA ACUST UNITED AC 2021; 28:2281-2295. [PMID: 34205437 PMCID: PMC8293196 DOI: 10.3390/curroncol28030210] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2021] [Revised: 06/06/2021] [Accepted: 06/16/2021] [Indexed: 12/13/2022]
Abstract
Glioma accounts for 80% of all malignant brain tumours and is the most common adult primary brain tumour. Age is an important factor affecting the development of cancer, as somatic mutations accumulate with age. Here, we aimed to analyse the significance of age-dependent non-silent somatic mutations in glioma prognosis. Histological tumour grade depends on age at diagnosis in patients with IDH1, TP53, ATRX, and EGFR mutations. Age of patients with wild-type IDH1 and EGFR increased with increase in tumour grade, while the age of patients with IDH1 or EGFR mutation remained constant. However, the age of patients with EGFR mutation was higher than that of patients with IDH1 mutation. The hierarchical clustering of patients was dominantly separated by IDH1 and EGFR mutations. Furthermore, patients with IDH1 mutation were dominantly separated by TP53 and ATRX double mutation and its double wild-type counterpart. The age of patients with ATRX and TP53 mutation was lower than that of patients with wild-type ATRX and TP53. Patients with the double mutation showed poorer prognosis than those with the double wild type genotype. Unlike IDH1 mutant, IDH1 wild-type showed upregulation of expression of epithelial mesenchymal transition associated genes.
Collapse
Affiliation(s)
- YoungJoon Park
- Institute Department of Biomedical Science, College of Life Science, CHA University, Seongnam 13488, Korea; (Y.P.); (J.P.); (J.W.A.); (S.J.K.)
- Department of Neurosurgery, Bundang CHA Medical Center, CHA University College of Medicine, Seongnam 13496, Korea; (J.M.S.); (S.K.); (S.J.H.)
| | - JeongMan Park
- Institute Department of Biomedical Science, College of Life Science, CHA University, Seongnam 13488, Korea; (Y.P.); (J.P.); (J.W.A.); (S.J.K.)
- Department of Neurosurgery, Bundang CHA Medical Center, CHA University College of Medicine, Seongnam 13496, Korea; (J.M.S.); (S.K.); (S.J.H.)
| | - Ju Won Ahn
- Institute Department of Biomedical Science, College of Life Science, CHA University, Seongnam 13488, Korea; (Y.P.); (J.P.); (J.W.A.); (S.J.K.)
- Department of Neurosurgery, Bundang CHA Medical Center, CHA University College of Medicine, Seongnam 13496, Korea; (J.M.S.); (S.K.); (S.J.H.)
| | - Jeong Min Sim
- Department of Neurosurgery, Bundang CHA Medical Center, CHA University College of Medicine, Seongnam 13496, Korea; (J.M.S.); (S.K.); (S.J.H.)
| | - Su Jung Kang
- Institute Department of Biomedical Science, College of Life Science, CHA University, Seongnam 13488, Korea; (Y.P.); (J.P.); (J.W.A.); (S.J.K.)
- Department of Neurosurgery, Bundang CHA Medical Center, CHA University College of Medicine, Seongnam 13496, Korea; (J.M.S.); (S.K.); (S.J.H.)
| | - Suwan Kim
- Department of Neurosurgery, Bundang CHA Medical Center, CHA University College of Medicine, Seongnam 13496, Korea; (J.M.S.); (S.K.); (S.J.H.)
| | - So Jung Hwang
- Department of Neurosurgery, Bundang CHA Medical Center, CHA University College of Medicine, Seongnam 13496, Korea; (J.M.S.); (S.K.); (S.J.H.)
- Global Research Supporting Center, Bundang CHA Medical Center, CHA University College of Medicine, Seongnam 13496, Korea
| | - Song-Hee Han
- Department of Pathology, Dong-A University College of Medicine, Dong-A University, Busan 49201, Korea;
| | - Kyoung Su Sung
- Department of Neurosurgery, Dong-A University Hospital, Dong-A University College of Medicine, Busan 49201, Korea
- Correspondence: or (K.S.S.); or (J.L.); Tel.: +82-51-240-5241 (K.S.S.); +82-31-780-5688 (J.L.)
| | - Jaejoon Lim
- Department of Neurosurgery, Bundang CHA Medical Center, CHA University College of Medicine, Seongnam 13496, Korea; (J.M.S.); (S.K.); (S.J.H.)
- Correspondence: or (K.S.S.); or (J.L.); Tel.: +82-51-240-5241 (K.S.S.); +82-31-780-5688 (J.L.)
| |
Collapse
|
18
|
Li J, Wang W, Wang J, Cao Y, Wang S, Zhao J. Viral Gene Therapy for Glioblastoma Multiforme: A Promising Hope for the Current Dilemma. Front Oncol 2021; 11:678226. [PMID: 34055646 PMCID: PMC8155537 DOI: 10.3389/fonc.2021.678226] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2021] [Accepted: 04/29/2021] [Indexed: 12/23/2022] Open
Abstract
Glioblastoma multiforme (GBM), as one of the most common malignant brain tumors, was limited in its treatment effectiveness with current options. Its invasive and infiltrative features led to tumor recurrence and poor prognosis. Effective treatment and survival improvement have always been a challenge. With the exploration of genetic mutations and molecular pathways in neuro-oncology, gene therapy is becoming a promising therapeutic approach. Therapeutic genes are delivered into target cells with viral vectors to act specific antitumor effects, which can be used in gene delivery, play an oncolysis effect, and induce host immune response. The application of engineering technology makes the virus vector used in genetics a more prospective future. Recent advances in viral gene therapy offer hope for treating brain tumors. In this review, we discuss the types and designs of viruses as well as their study progress and potential applications in the treatment of GBM. Although still under research, viral gene therapy is promising to be a new therapeutic approach for GBM treatment in the future.
Collapse
Affiliation(s)
- Junsheng Li
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China.,China National Clinical Research Center for Neurological Diseases, Beijing, China.,Center of Stroke, Beijing Institute for Brain Disorders, Beijing, China.,Beijing Key Laboratory of Translational Medicine for Cerebrovascular Disease, Beijing, China.,Beijing Translational Engineering Center for 3D Printer in Clinical Neuroscience, Beijing, China
| | - Wen Wang
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China.,China National Clinical Research Center for Neurological Diseases, Beijing, China.,Center of Stroke, Beijing Institute for Brain Disorders, Beijing, China.,Beijing Key Laboratory of Translational Medicine for Cerebrovascular Disease, Beijing, China.,Beijing Translational Engineering Center for 3D Printer in Clinical Neuroscience, Beijing, China
| | - Jia Wang
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China.,China National Clinical Research Center for Neurological Diseases, Beijing, China.,Center of Stroke, Beijing Institute for Brain Disorders, Beijing, China.,Beijing Key Laboratory of Translational Medicine for Cerebrovascular Disease, Beijing, China.,Beijing Translational Engineering Center for 3D Printer in Clinical Neuroscience, Beijing, China
| | - Yong Cao
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China.,China National Clinical Research Center for Neurological Diseases, Beijing, China.,Center of Stroke, Beijing Institute for Brain Disorders, Beijing, China.,Beijing Key Laboratory of Translational Medicine for Cerebrovascular Disease, Beijing, China.,Beijing Translational Engineering Center for 3D Printer in Clinical Neuroscience, Beijing, China
| | - Shuo Wang
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China.,China National Clinical Research Center for Neurological Diseases, Beijing, China.,Center of Stroke, Beijing Institute for Brain Disorders, Beijing, China.,Beijing Key Laboratory of Translational Medicine for Cerebrovascular Disease, Beijing, China.,Beijing Translational Engineering Center for 3D Printer in Clinical Neuroscience, Beijing, China
| | - Jizong Zhao
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China.,China National Clinical Research Center for Neurological Diseases, Beijing, China.,Center of Stroke, Beijing Institute for Brain Disorders, Beijing, China.,Beijing Key Laboratory of Translational Medicine for Cerebrovascular Disease, Beijing, China.,Beijing Translational Engineering Center for 3D Printer in Clinical Neuroscience, Beijing, China.,Savaid Medical School, University of the Chinese Academy of Sciences, Beijing, China
| |
Collapse
|
19
|
Tsamis KI, Sakkas H, Giannakis A, Ryu HS, Gartzonika C, Nikas IP. Evaluating Infectious, Neoplastic, Immunological, and Degenerative Diseases of the Central Nervous System with Cerebrospinal Fluid-Based Next-Generation Sequencing. Mol Diagn Ther 2021; 25:207-229. [PMID: 33646562 PMCID: PMC7917176 DOI: 10.1007/s40291-021-00513-x] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/07/2021] [Indexed: 12/24/2022]
Abstract
Cerebrospinal fluid (CSF) is a clear and paucicellular fluid that circulates within the ventricular system and the subarachnoid space of the central nervous system (CNS), and diverse CNS disorders can impact its composition, volume, and flow. As conventional CSF testing suffers from suboptimal sensitivity, this review aimed to evaluate the role of next-generation sequencing (NGS) in the work-up of infectious, neoplastic, neuroimmunological, and neurodegenerative CNS diseases. Metagenomic NGS showed improved sensitivity—compared to traditional methods—to detect bacterial, viral, parasitic, and fungal infections, while the overall performance was maximized in some studies when all diagnostic modalities were used. In patients with primary CNS cancer, NGS findings in the CSF were largely concordant with the molecular signatures derived from tissue-based molecular analysis; of interest, additional mutations were identified in the CSF in some glioma studies, reflecting intratumoral heterogeneity. In patients with metastasis to the CNS, NGS facilitated diagnosis, prognosis, therapeutic management, and monitoring, exhibiting higher sensitivity than neuroimaging, cytology, and plasma-based molecular analysis. Although evidence is still rudimentary, NGS could enhance the diagnosis and pathogenetic understanding of multiple sclerosis in addition to Alzheimer and Parkinson disease. To conclude, NGS has shown potential to aid the research, facilitate the diagnostic approach, and improve the management outcomes of all the aforementioned CNS diseases. However, to establish its role in clinical practice, the clinical validity and utility of each NGS protocol should be determined. Lastly, as most evidence has been derived from small and retrospective studies, results from randomized control trials could be of significant value.
Collapse
Affiliation(s)
- Konstantinos I Tsamis
- Department of Neurology, University Hospital of Ioannina, 45500, Ioannina, Greece. .,School of Medicine, European University Cyprus, 2404, Nicosia, Cyprus.
| | - Hercules Sakkas
- Microbiology Department, Faculty of Medicine, School of Health Sciences, University of Ioannina, 45110, Ioannina, Greece
| | - Alexandros Giannakis
- Department of Neurology, University Hospital of Ioannina, 45500, Ioannina, Greece
| | - Han Suk Ryu
- Department of Pathology, Seoul National University Hospital, Seoul, 03080, Korea
| | - Constantina Gartzonika
- Microbiology Department, Faculty of Medicine, School of Health Sciences, University of Ioannina, 45110, Ioannina, Greece
| | - Ilias P Nikas
- School of Medicine, European University Cyprus, 2404, Nicosia, Cyprus
| |
Collapse
|
20
|
Zhang Y, Wang Q, Luo N, Liu J, Ren H, Shao X, Zhang L, Yu Y. MicroRNA-1269a Promotes Proliferation and Arrest of Apoptosis of Glioma Cells by Directly Targeting ATRX. Front Oncol 2020; 10:563901. [PMID: 33194637 PMCID: PMC7659443 DOI: 10.3389/fonc.2020.563901] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2020] [Accepted: 08/20/2020] [Indexed: 12/16/2022] Open
Abstract
Glioma is one of the deadliest malignant brain tumors in adults worldwide. MicroRNA (miR) has been reported to be a pivotal regulator in human tumors. The aim of this study was to determine the expression, function, and mechanism of action of miR-1269a in glioma progression. The expression of miR-1269a was higher in both glioma cases reported in databases and glioma cell lines, and it was highly associated with poorer prognosis. Next, it was shown in vitro that mimic of miR-1269a could promote glioma progression and arrest apoptosis, whereas the inhibition of miR-1269a exhibited the opposite effects. In addition, miR-1269a was found to directly target ATRX chromatin remodeler by a dual-luciferase reporter assay. Moreover, ATRX overexpression could reverse the suppressive effects of miR-1269a on proliferation and apoptosis in vitro. In vivo subcutaneous xenograft tumor assay was also performed to confirm the phenotypes and molecular mechanism involved. Taking the findings together, our study implies that the miR-1269a/ATRX axis is a novel therapeutic target of glioma.
Collapse
Affiliation(s)
- Yulian Zhang
- Department of Neurosurgery, Peking University China-Japan Friendship School of Clinical Medicine, Beijing, China.,Department of Neurosurgery, China-Japan Friendship Hospital, Beijing, China
| | - Qi Wang
- Department of Neurosurgery, China-Japan Friendship Hospital, Beijing, China
| | - Na Luo
- Department of Neurosurgery, Peking University China-Japan Friendship School of Clinical Medicine, Beijing, China.,Department of Neurosurgery, China-Japan Friendship Hospital, Beijing, China
| | - Jiang Liu
- Department of Neurosurgery, China-Japan Friendship Hospital, Beijing, China
| | - Hongxiang Ren
- Department of Neurosurgery, China-Japan Friendship Hospital, Beijing, China
| | - Xu Shao
- Department of Neurosurgery, China-Japan Friendship Hospital, Beijing, China
| | - Li Zhang
- Department of Neurosurgery, Peking University China-Japan Friendship School of Clinical Medicine, Beijing, China.,Department of Neurosurgery, China-Japan Friendship Hospital, Beijing, China.,Department of Neurosurgery, Graduate School of Peking Union Medical College, Beijing, China
| | - Yanbing Yu
- Department of Neurosurgery, Peking University China-Japan Friendship School of Clinical Medicine, Beijing, China.,Department of Neurosurgery, China-Japan Friendship Hospital, Beijing, China.,Department of Neurosurgery, Graduate School of Peking Union Medical College, Beijing, China
| |
Collapse
|
21
|
Dusart P, Hallström BM, Renné T, Odeberg J, Uhlén M, Butler LM. A Systems-Based Map of Human Brain Cell-Type Enriched Genes and Malignancy-Associated Endothelial Changes. Cell Rep 2020; 29:1690-1706.e4. [PMID: 31693905 DOI: 10.1016/j.celrep.2019.09.088] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2019] [Revised: 04/09/2019] [Accepted: 09/27/2019] [Indexed: 02/02/2023] Open
Abstract
Changes in the endothelium of the cerebral vasculature can contribute to inflammatory, thrombotic, and malignant disorders. The importance of defining cell-type-specific genes and their modification in disease is increasingly recognized. Here, we develop a bioinformatics-based approach to identify normal brain cell-enriched genes, using bulk RNA sequencing (RNA-seq) data from 238 normal human cortex samples from 2 independent cohorts. We compare endothelial cell-enriched gene profiles with astrocyte, oligodendrocyte, neuron, and microglial cell profiles. Endothelial changes in malignant disease are explored using RNA-seq data from 516 lower-grade gliomas and 401 glioblastomas. Lower-grade gliomas appear to be an "endothelial intermediate" between normal brain and glioblastoma. We apply our method for the prediction of glioblastoma-specific endothelial biomarkers, providing potential diagnostic or therapeutic targets. In summary, we provide a roadmap of endothelial cell identity in normal and malignant brain, using a method developed to resolve bulk RNA-seq into constituent cell-type-enriched profiles.
Collapse
Affiliation(s)
- Philip Dusart
- Science for Life Laboratory, Department of Protein Science, Royal Institute of Technology (KTH), 171 21 Stockholm, Sweden; K.G. Jebsen Thrombosis Research and Expertise Centre, Department of Clinical Medicine, The Arctic University of Norway, 9019 Tromsø, Norway
| | - Björn Mikael Hallström
- Science for Life Laboratory, Department of Protein Science, Royal Institute of Technology (KTH), 171 21 Stockholm, Sweden
| | - Thomas Renné
- Institute for Clinical Chemistry and Laboratory Medicine, University Medical Centre Hamburg-Eppendorf, 20246 Hamburg, Germany
| | - Jacob Odeberg
- Science for Life Laboratory, Department of Protein Science, Royal Institute of Technology (KTH), 171 21 Stockholm, Sweden; K.G. Jebsen Thrombosis Research and Expertise Centre, Department of Clinical Medicine, The Arctic University of Norway, 9019 Tromsø, Norway; The University Hospital of North Norway (UNN), PB100, 9038 Tromsø, Norway; Department of Hematology, Karolinska University Hospital, 171 77 Stockholm, Sweden
| | - Mathias Uhlén
- Science for Life Laboratory, Department of Protein Science, Royal Institute of Technology (KTH), 171 21 Stockholm, Sweden
| | - Lynn Marie Butler
- Science for Life Laboratory, Department of Protein Science, Royal Institute of Technology (KTH), 171 21 Stockholm, Sweden; K.G. Jebsen Thrombosis Research and Expertise Centre, Department of Clinical Medicine, The Arctic University of Norway, 9019 Tromsø, Norway; Institute for Clinical Chemistry and Laboratory Medicine, University Medical Centre Hamburg-Eppendorf, 20246 Hamburg, Germany; Clinical Chemistry and Blood Coagulation Research, Department of Molecular Medicine and Surgery, Karolinska Institute, 171 76 Stockholm, Sweden.
| |
Collapse
|
22
|
Petre G, Durand H, Pelletier L, Poulenard M, Nugue G, Ray PF, Rendu J, Coutton C, Berger F, Bidart M. Rapid Proteomic Profiling by MALDI-TOF Mass Spectrometry for Better Brain Tumor Classification. Proteomics Clin Appl 2020; 14:e1900116. [PMID: 32198817 DOI: 10.1002/prca.201900116] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2019] [Revised: 02/21/2020] [Indexed: 12/21/2022]
Abstract
PURPOSE Glioblastoma is one of the most aggressive primary brain cancers. The precise grading of tumors is important to adopt the best follow-up treatment but complementary methods to histopathological diagnosis still lack in achieving an unbiased and reliable classification. EXPERIMENTAL DESIGN To progress in the field, a rapid Matrix Assisted Laser Desorption Ionization - Time of Flight Mass spectrometry (MALDI-TOF MS) protocole, devised for the identification and taxonomic classification of microorganisms and based on the analysis of whole cell extracts, was applied to glioma cell lines. RESULTS The analysis of different human glioblastoma cell lines permitted to identify distinct proteomic profiles thus demonstrating the ability of MALDI-TOF to distinguish different malignant cell types. CONCLUSIONS AND CLINICAL RELEVANCE In the study, the authors showed the ability of MALDI-TOF profiling to discriminate glioblastoma cell lines, demonstrating that this technique could be used in complement to histological tumor classification. The proposed procedure is rapid and inexpensive and could be used to improve brain tumors classification and help propose a personalized and more efficient treatment.
Collapse
Affiliation(s)
- Graciane Petre
- UMR1205, Brain Tech Lab, Grenoble Alpes University, Grenoble, 38000, France
| | - Harmonie Durand
- UMR1205, Brain Tech Lab, Grenoble Alpes University, Grenoble, 38000, France
| | - Laurent Pelletier
- Université Grenoble Alpes, Grenoble Institut des Neurosciences, GIN, F-38000, Grenoble, France
| | - Margot Poulenard
- UMR1205, Brain Tech Lab, Grenoble Alpes University, Grenoble, 38000, France
| | - Guillaume Nugue
- UMR1205, Brain Tech Lab, Grenoble Alpes University, Grenoble, 38000, France
| | - Pierre F Ray
- Genetic Epigenetic and Therapies of Infertility, Institute for Advanced Biosciences, Inserm U1209, CNRS UMR 5309, Université Grenoble Alpes, Grenoble, 38000, France.,Unité Médicale de génétique de l'infertilité et DPI moléculaire (GI-DPI), Pôle Biologie, Institut de Biologie et de Pathologie, Centre Hospitalier Universitaire Grenoble Alpes, La Tronche, 38700, France
| | - John Rendu
- Université Grenoble Alpes, Grenoble Institut des Neurosciences, GIN, F-38000, Grenoble, France.,Unité Médicale de Génétique Moléculaire: Maladies Héréditaires et Oncologie, Pôle Biologie, Institut de Biologie et de Pathologie, Centre Hospitalier Universitaire Grenoble Alpes, La Tronche, 38700, France
| | - Charles Coutton
- Genetic Epigenetic and Therapies of Infertility, Institute for Advanced Biosciences, Inserm U1209, CNRS UMR 5309, Université Grenoble Alpes, Grenoble, 38000, France.,Unité Médicale de Génétique Chromosomique, Hopital Couple Enfant, Centre Hospitalier Universitaire Grenoble Alpes, La Tronche, 38700, France
| | - Francois Berger
- UMR1205, Brain Tech Lab, Grenoble Alpes University, Grenoble, 38000, France
| | - Marie Bidart
- UMR1205, Brain Tech Lab, Grenoble Alpes University, Grenoble, 38000, France.,Unité Médicale de Génétique Moléculaire: Maladies Héréditaires et Oncologie, Pôle Biologie, Institut de Biologie et de Pathologie, Centre Hospitalier Universitaire Grenoble Alpes, La Tronche, 38700, France
| |
Collapse
|
23
|
The histomolecular criteria established for adult anaplastic pilocytic astrocytoma are not applicable to the pediatric population. Acta Neuropathol 2020; 139:287-303. [PMID: 31677015 PMCID: PMC6989446 DOI: 10.1007/s00401-019-02088-8] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2019] [Revised: 10/21/2019] [Accepted: 10/22/2019] [Indexed: 12/18/2022]
Abstract
Pilocytic astrocytoma (PA) is the most common pediatric glioma, arising from a single driver MAPK pathway alteration. Classified as a grade I tumor according to the 2016 WHO classification, prognosis is excellent with a 10-year survival rate > 95% after surgery. However, rare cases present with anaplastic features, including an unexpected high mitotic/proliferative index, thus posing a diagnostic and therapeutic challenge. Based on small histomolecular series and case reports, such tumors arising at the time of diagnosis or recurrence have been designated by many names including pilocytic astrocytoma with anaplastic features (PAAF). Recent DNA methylation-profiling studies performed mainly on adult cases have revealed that PAAF exhibit a specific methylation signature, thus constituting a distinct methylation class from typical PA [methylation class anaplastic astrocytoma with piloid features-(MC-AAP)]. However, the diagnostic and prognostic significance of MC-AAP remains to be determined in children. We performed an integrative work on the largest pediatric cohort of PAAF, defined according to strict criteria: morphology compatible with the diagnosis of PA, with or without necrosis, ≥ 4 mitoses for 2.3 mm2, and MAPK pathway alteration. We subjected 31 tumors to clinical, imaging, morphological and molecular analyses, including DNA methylation profiling. We identified only one tumor belonging to the MC-AAP (3%), the others exhibiting a methylation profile typical for PA (77%), IDH-wild-type glioblastoma (7%), and diffuse leptomeningeal glioneuronal tumor (3%), while three cases (10%) did not match to a known DNA methylation class. No significant outcome differences were observed between PAAF with necrosis versus no necrosis (p = 0.07), or with 4-6 mitoses versus 7 or more mitoses (p = 0.857). Our findings argue that the diagnostic histomolecular criteria established for anaplasia in adult PA are not of diagnostic or prognostic value in a pediatric setting. Further extensive and comprehensive integrative studies are necessary to accurately define this exceptional entity in children.
Collapse
|
24
|
Valdebenito S, D'Amico D, Eugenin E. Novel approaches for glioblastoma treatment: Focus on tumor heterogeneity, treatment resistance, and computational tools. Cancer Rep (Hoboken) 2019; 2:e1220. [PMID: 32729241 PMCID: PMC7941428 DOI: 10.1002/cnr2.1220] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2018] [Revised: 06/05/2019] [Accepted: 07/02/2019] [Indexed: 09/20/2023] Open
Abstract
BACKGROUND Glioblastoma (GBM) is a highly aggressive primary brain tumor. Currently, the suggested line of action is the surgical resection followed by radiotherapy and treatment with the adjuvant temozolomide, a DNA alkylating agent. However, the ability of tumor cells to deeply infiltrate the surrounding tissue makes complete resection quite impossible, and, in consequence, the probability of tumor recurrence is high, and the prognosis is not positive. GBM is highly heterogeneous and adapts to treatment in most individuals. Nevertheless, these mechanisms of adaption are unknown. RECENT FINDINGS In this review, we will discuss the recent discoveries in molecular and cellular heterogeneity, mechanisms of therapeutic resistance, and new technological approaches to identify new treatments for GBM. The combination of biology and computer resources allow the use of algorithms to apply artificial intelligence and machine learning approaches to identify potential therapeutic pathways and to identify new drug candidates. CONCLUSION These new approaches will generate a better understanding of GBM pathogenesis and will result in novel treatments to reduce or block the devastating consequences of brain cancers.
Collapse
Affiliation(s)
- Silvana Valdebenito
- Department of Neuroscience, Cell Biology, and AnatomyUniversity of Texas Medical Branch (UTMB)GalvestonTexas
| | - Daniela D'Amico
- Department of Neuroscience, Cell Biology, and AnatomyUniversity of Texas Medical Branch (UTMB)GalvestonTexas
- Department of Biomedicine and Clinic NeuroscienceUniversity of PalermoPalermoItaly
| | - Eliseo Eugenin
- Department of Neuroscience, Cell Biology, and AnatomyUniversity of Texas Medical Branch (UTMB)GalvestonTexas
| |
Collapse
|
25
|
Youssef GC, Gomez-Manzano C, Sawaya R, Fueyo J. Antitumor immune response during glioma virotherapy. Neuro Oncol 2019; 21:1087-1088. [PMID: 31348516 DOI: 10.1093/neuonc/noz114] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Affiliation(s)
- Gilbert C Youssef
- Department of Neuro-Oncology, Brain Tumor Program, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Candelaria Gomez-Manzano
- Department of Neuro-Oncology, Brain Tumor Program, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Raymond Sawaya
- Department of Neurosurgery, Brain Tumor Program, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Juan Fueyo
- Department of Neuro-Oncology, Brain Tumor Program, The University of Texas MD Anderson Cancer Center, Houston, Texas.,Department of Neurosurgery, Brain Tumor Program, The University of Texas MD Anderson Cancer Center, Houston, Texas
| |
Collapse
|
26
|
Jovčevska I. Genetic secrets of long-term glioblastoma survivors. Bosn J Basic Med Sci 2019; 19:116-124. [PMID: 30114377 DOI: 10.17305/bjbms.2018.3717] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2018] [Accepted: 07/25/2018] [Indexed: 12/12/2022] Open
Abstract
Glioblastomas are the most aggressive and lethal primary astrocytic tumors of the central nervous system. They account for 60% to 70% of all gliomas and the majority are diagnosed in Caucasian male patients at advanced age. Genetic analyses of glioblastoma show a great intra- and inter-tumor heterogeneity, which opens up a debate about its cellular origin. Different types of brain cells, including astrocytes, neural stem cells, oligodendrocyte precursor cells and glioblastoma stem cells are proposed to have a role in tumor initiation and spreading; however, data is still inconclusive. Due to short life expectancy, long-term glioblastoma survivors are defined as patients who live longer than two years post-diagnosis. Extreme survivors, living 10 years or more after diagnosis, comprise less than 1% of all patients. Molecular testing indicates genetic differences between short- and long-term survivors with glioblastoma. The most informative are IDH1/2 gene mutations and MGMT promoter methylation, which are associated with a better response to standard clinical care. Moreover, a decreased expression of the CHI3L1, FBLN4, EMP3, IGFBP2, IGFBP3, LGALS3, MAOB, PDPN, SERPING1 and TIMP1 genes has been associated with prolonged survival. In addition, emerging evidence suggests the role of different microRNAs in predicting patient survival. Other factors that may affect the survival of glioblastoma patients include clinical/demographic characteristics such as seizures at presentation, age at diagnosis, and the extent of surgical resection. Because of the small number of long-term survivors with glioblastoma, comparative studies on genetic differences between short- and long-term survivors are challenging. To improve patient management and clinical outcomes, a thorough "omics" approach is necessary for identifying differences between short- and long-term survivors with glioblastoma.
Collapse
Affiliation(s)
- Ivana Jovčevska
- Medical Center for Molecular Biology, Institute of Biochemistry, Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia.
| |
Collapse
|