1
|
Ohba S, Teranishi T, Matsumura K, Kumon M, Kojima D, Fujiwara E, Nakao K, Kuwahara K, Murayama K, Pareira ES, Yamada S, Joko M, Nakae S, Muto J, Nishiyama Y, Adachi K, Sasaki H, Abe M, Hasegawa M, Hirose Y. Factors involved in maintaining Karnofsky Performance Status (≥ 50%) in glioblastoma, IDH-wildtype patients treated with temozolomide and radiotherapy. Sci Rep 2025; 15:1750. [PMID: 39799218 PMCID: PMC11724988 DOI: 10.1038/s41598-025-85339-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2024] [Accepted: 01/02/2025] [Indexed: 01/15/2025] Open
Abstract
Karnofsky Performance Status (KPS) is a widely used scale to assess performance status. KPS ≥ 50% implies that patients can live at home. Therefore, maintaining KPS ≥ 50% is important to improve the quality of life of patients with glioblastoma, whose median survival is less than 2 years. This study aimed to identify the factors associated with survival time with maintenance of KPS ≥ 50% (survival with KPS ≥ 50%) in patients with glioblastoma, IDH-wildtype. Ninety-eight patients with glioblastomas, IDH-wildtype, who were treated with concomitant radiotherapy (RT) and temozolomide (TMZ) followed by maintenance TMZ therapy, and whose KPS at the start of RT was ≥ 50%, were included. The median survival with KPS ≥ 50% was 13.3 months. In univariate analysis, preoperative KPS (≥ 80%), KPS at the start of RT (≥ 80%), residual tumor size (< 2 cm3), methylated MGMT promotor, and implantation of BCNU wafer were associated with survival with KPS ≥ 50%. In multivariate analysis, KPS at the start of RT (≥ 80%), methylated MGMT promotor, and residual tumor size (< 2 cm3) were significantly associated with increased survival with KPS ≥ 50%. A strategy of maximum possible tumor resection without compromising KPS is desirable to prolong the survival time with KPS ≥ 50%.
Collapse
Affiliation(s)
- Shigeo Ohba
- Department of Neurosurgery, Fujita Health University School of Medicine, 1-98 Dengakugakubo Kutsukake-cho, Toyoake, Aichi, 470-1192, Japan.
| | - Takao Teranishi
- Department of Neurosurgery, Fujita Health University School of Medicine, 1-98 Dengakugakubo Kutsukake-cho, Toyoake, Aichi, 470-1192, Japan
| | - Kazuyasu Matsumura
- Department of Neurosurgery, Fujita Health University School of Medicine, 1-98 Dengakugakubo Kutsukake-cho, Toyoake, Aichi, 470-1192, Japan
| | - Masanobu Kumon
- Department of Neurosurgery, Fujita Health University School of Medicine, 1-98 Dengakugakubo Kutsukake-cho, Toyoake, Aichi, 470-1192, Japan
| | - Daijiro Kojima
- Department of Neurosurgery, Fujita Health University School of Medicine, 1-98 Dengakugakubo Kutsukake-cho, Toyoake, Aichi, 470-1192, Japan
| | - Eiji Fujiwara
- Department of Neurosurgery, Fujita Health University School of Medicine, 1-98 Dengakugakubo Kutsukake-cho, Toyoake, Aichi, 470-1192, Japan
| | - Kazutaka Nakao
- Department of Neurosurgery, Fujita Health University School of Medicine, 1-98 Dengakugakubo Kutsukake-cho, Toyoake, Aichi, 470-1192, Japan
| | - Kiyonori Kuwahara
- Department of Neurosurgery, Fujita Health University School of Medicine, 1-98 Dengakugakubo Kutsukake-cho, Toyoake, Aichi, 470-1192, Japan
| | - Kazuhiro Murayama
- Department of Radiology, Fujita Health University School of Medicine, Toyoake, Aichi, Japan
| | | | - Seiji Yamada
- Department of Diagnostic Pathology, Fujita Health University School of Medicine, Toyoake, Aichi, Japan
| | - Masahiro Joko
- Department of Neurosurgery, Fujita Health University School of Medicine, 1-98 Dengakugakubo Kutsukake-cho, Toyoake, Aichi, 470-1192, Japan
| | - Shunsuke Nakae
- Department of Neurosurgery, Fujita Health University School of Medicine, 1-98 Dengakugakubo Kutsukake-cho, Toyoake, Aichi, 470-1192, Japan
| | - Jun Muto
- Department of Neurosurgery, Fujita Health University School of Medicine, 1-98 Dengakugakubo Kutsukake-cho, Toyoake, Aichi, 470-1192, Japan
| | - Yuya Nishiyama
- Department of Neurosurgery, Fujita Health University School of Medicine, 1-98 Dengakugakubo Kutsukake-cho, Toyoake, Aichi, 470-1192, Japan
| | - Kazuhide Adachi
- Department of Neurosurgery, Fujita Health University School of Medicine, 1-98 Dengakugakubo Kutsukake-cho, Toyoake, Aichi, 470-1192, Japan
| | - Hikaru Sasaki
- Department of Neurosurgery, Tokyo Dental College Ichikawa General Hospital, Ichikawa, Chiba, Japan
| | - Masato Abe
- Department of Pathology, Fujita Health University School of Health Sciences, Toyoake, Aichi, Japan
| | - Mitsuhiro Hasegawa
- Department of Neurosurgery, Fujita Health University School of Medicine, 1-98 Dengakugakubo Kutsukake-cho, Toyoake, Aichi, 470-1192, Japan
| | - Yuichi Hirose
- Department of Neurosurgery, Fujita Health University School of Medicine, 1-98 Dengakugakubo Kutsukake-cho, Toyoake, Aichi, 470-1192, Japan
| |
Collapse
|
2
|
Spallotta F, Illi B. The Role of HDAC6 in Glioblastoma Multiforme: A New Avenue to Therapeutic Interventions? Biomedicines 2024; 12:2631. [PMID: 39595195 PMCID: PMC11591585 DOI: 10.3390/biomedicines12112631] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2024] [Revised: 11/14/2024] [Accepted: 11/15/2024] [Indexed: 11/28/2024] Open
Abstract
Despite the great advances in basic research results, glioblastoma multiforme (GBM) still remains an incurable tumour. To date, a GBM diagnosis is a death sentence within 15-18 months, due to the high recurrence rate and resistance to conventional radio- and chemotherapy approaches. The effort the scientific community is lavishing on the never-ending battle against GBM is reflected by the huge number of clinical trials launched, about 2003 on 10 September 2024. However, we are still far from both an in-depth comprehension of the biological and molecular processes leading to GBM onset and progression and, importantly, a cure. GBM is provided with high intratumoral heterogeneity, immunosuppressive capacity, and infiltrative ability due to neoangiogenesis. These features impact both tumour aggressiveness and therapeutic vulnerability, which is further limited by the presence in the tumour core of niches of glioblastoma stem cells (GSCs) that are responsible for the relapse of this brain neoplasm. Epigenetic alterations may both drive and develop along GBM progression and also rely on changes in the expression of the genes encoding histone-modifying enzymes, including histone deacetylases (HDACs). Among them, HDAC6-a cytoplasmic HDAC-has recently gained attention because of its role in modulating several biological aspects of GBM, including DNA repair ability, massive growth, radio- and chemoresistance, and de-differentiation through primary cilia disruption. In this review article, the available information related to HDAC6 function in GBM will be presented, with the aim of proposing its inhibition as a valuable therapeutic route for this deadly brain tumour.
Collapse
Affiliation(s)
- Francesco Spallotta
- Department of Biology and Biotechnology Charles Darwin, Sapienza University, 00185 Rome, Italy;
- Istituto Pasteur Italia-Fondazione Cenci Bolognetti, Sapienza University, 00185 Rome, Italy
| | - Barbara Illi
- Institute of Molecular Biology and Pathology, National Research Council (IBPM-CNR), 00185 Rome, Italy
| |
Collapse
|
3
|
Dashtaki ME, Karimi E, Ghasemi S. Genetic Variants Impacting Angiogenesis Signaling Pathways in Glioblastoma Multiforme: A Systematic Review of Mutations and Polymorphisms. Curr Mol Med 2024; 24:1346-1357. [PMID: 37489777 DOI: 10.2174/1566524023666230725115812] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2023] [Revised: 06/11/2023] [Accepted: 06/19/2023] [Indexed: 07/26/2023]
Abstract
BACKGROUND Several signaling pathways are involved in the process of angiogenesis, which is one of the most important hallmarks of glioblastoma multiforme (GBM). Identifying related gene variants can help researchers work out what causes anti-angiogenesis drug resistance. OBJECTIVE The goal of this systematic analysis was to identify all mutations and polymorphisms involved in angiogenesis pathways in GBM and their impact on clinical outcomes. METHODS The keywords include glioblastoma, angiogenesis, signaling pathway, mutation, polymorphism, and related terms used to search ISI, PubMed, and Scopus for relevant articles published up to January 2022. The PRISMA protocol was used to conduct our systematic review. The related articles were taken into consideration. The risk of bias in the associated articles was surveyed, as well as the article scoring. Two authors collaborated on data extraction. RESULTS The inclusion criteria were included in 32 articles out of a total of 787 articles. VEGF, HIF1a, EGFR, PI3K, and MAPK are the pathways that have been studied the most. IDH1, VEGF, VEGFR, EGFR, and HIF1a are the genes with the highest frequency of mutations or polymorphisms. CONCLUSION In conclusion, this study found that angiogenesis in primary or recurrent GBM is linked to gene changes in eleven signaling pathways. However, some of these gene mutations have been researched numerous times in relation to angiogenesis, while others have only been studied once. Understanding these changes will help us employ combination therapies more effectively for GBM patients' survival and personal medicine.
Collapse
Affiliation(s)
| | - Elham Karimi
- Department of Medical Genetics, School of medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Sorayya Ghasemi
- Cancer Research Center, Shahrekord University of Medical Sciences, Shahrekord, Iran
| |
Collapse
|
4
|
Zhong S, Yang W, Zhang Z, Xie Y, Pan L, Ren J, Ren F, Li Y, Xie H, Chen H, Deng D, Lu J, Li H, Wu B, Chen Y, Peng F, Puduvalli VK, Sai K, Li Y, Cheng Y, Mou Y. Association between viral infections and glioma risk: a two-sample bidirectional Mendelian randomization analysis. BMC Med 2023; 21:487. [PMID: 38053181 PMCID: PMC10698979 DOI: 10.1186/s12916-023-03142-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/17/2023] [Accepted: 10/30/2023] [Indexed: 12/07/2023] Open
Abstract
BACKGROUND Glioma is one of the leading types of brain tumor, but few etiologic factors of primary glioma have been identified. Previous observational research has shown an association between viral infection and glioma risk. In this study, we used Mendelian randomization (MR) analysis to explore the direction and magnitude of the causal relationship between viral infection and glioma. METHODS We conducted a two-sample bidirectional MR analysis using genome-wide association study (GWAS) data. Summary statistics data of glioma were collected from the largest meta-analysis GWAS, involving 12,488 cases and 18,169 controls. Single-nucleotide polymorphisms (SNPs) associated with exposures were used as instrumental variables to estimate the causal relationship between glioma and twelve types of viral infections from corresponding GWAS data. In addition, sensitivity analyses were performed. RESULTS After correcting for multiple tests and sensitivity analysis, we detected that genetically predicted herpes zoster (caused by Varicella zoster virus (VZV) infection) significantly decreased risk of low-grade glioma (LGG) development (OR = 0.85, 95% CI: 0.76-0.96, P = 0.01, FDR = 0.04). No causal effects of the other eleven viral infections on glioma and reverse causality were detected. CONCLUSIONS This is one of the first and largest studies in this field. We show robust evidence supporting that genetically predicted herpes zoster caused by VZV infection reduces risk of LGG. The findings of our research advance understanding of the etiology of glioma.
Collapse
Affiliation(s)
- Sheng Zhong
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-Sen University Cancer Center, Guangzhou, 510060, People's Republic of China
| | - Wenzhuo Yang
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-Sen University Cancer Center, Guangzhou, 510060, People's Republic of China
| | - Zhiyun Zhang
- Department of Plastic Surgery, The First Hospital of Jilin University, Changchun, 130000, People's Republic of China
| | - Yangyiran Xie
- Vanderbilt University School of Medicine, Vanderbilt University, 1161 21St Ave S # D3300, Nashville, TN, 37232, USA
| | - Lin Pan
- Clinical College, Jilin University, Street Xinmin 828, Changchun, People's Republic of China
| | - Jiaxin Ren
- Stroke Center, Department of Neurology, The First Hospital of Jilin University, Chang Chun, People's Republic of China
| | - Fei Ren
- Clinical College, Jilin University, Street Xinmin 828, Changchun, People's Republic of China
| | - Yifan Li
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-Sen University Cancer Center, Guangzhou, 510060, People's Republic of China
| | - Haoqun Xie
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-Sen University Cancer Center, Guangzhou, 510060, People's Republic of China
| | - Hongyu Chen
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-Sen University Cancer Center, Guangzhou, 510060, People's Republic of China
| | - Davy Deng
- Dana Farber Cancer Institute, 450 Brookline Ave, Boston, MA, 02215, USA
| | - Jie Lu
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-Sen University Cancer Center, Guangzhou, 510060, People's Republic of China
| | - Hui Li
- Stroke Center, Department of Neurology, The First Hospital of Jilin University, Chang Chun, People's Republic of China
| | - Bo Wu
- Department of Orthopaedics, The First Hospital of Jilin University, No.71, Street Xinmin Road, Chaoyang District, Changchun, Jilin, People's Republic of China
| | - Youqi Chen
- Clinical College, Jilin University, Street Xinmin 828, Changchun, People's Republic of China
| | - Fei Peng
- Department of Medicine, Division of Endocrinology, Diabetes and Metabolism, Baylor College of Medicine, Houston, TX, USA
| | - Vinay K Puduvalli
- Department of Neuro-Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
- Department of Neurosurgery, The First Hospital of Jilin University, Changchun, People's Republic of China
| | - Ke Sai
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-Sen University Cancer Center, Guangzhou, 510060, People's Republic of China.
| | - Yunqian Li
- Department of Neurosurgery, The First Hospital of Jilin University, Changchun, People's Republic of China.
| | - Ye Cheng
- Department of Neurosurgery, Xuanwu Hospital, Capital Medical University, Beijing, People's Republic of China.
| | - Yonggao Mou
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-Sen University Cancer Center, Guangzhou, 510060, People's Republic of China.
| |
Collapse
|
5
|
Ohba S, Murayama K, Teranishi T, Kumon M, Nakae S, Yui M, Yamamoto K, Yamada S, Abe M, Hasegawa M, Hirose Y. Three-Dimensional Amide Proton Transfer-Weighted Imaging for Differentiating between Glioblastoma, IDH-Wildtype and Primary Central Nervous System Lymphoma. Cancers (Basel) 2023; 15:952. [PMID: 36765909 PMCID: PMC9913574 DOI: 10.3390/cancers15030952] [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: 12/07/2022] [Revised: 01/26/2023] [Accepted: 01/28/2023] [Indexed: 02/05/2023] Open
Abstract
Distinguishing primary central nervous system lymphoma (PCNSL) from glioblastoma, isocitrate dehydrogenase (IDH)-wildtype is sometimes hard. Because the role of operation on them varies, accurate preoperative diagnosis is crucial. In this study, we evaluated whether a specific kind of chemical exchange saturation transfer imaging, i.e., amide proton transfer-weighted (APTw) imaging, was useful to distinguish PCNSL from glioblastoma, IDH-wildtype. A total of 14 PCNSL and 27 glioblastoma, IDH-wildtype cases were evaluated. There was no significant difference in the mean APTw signal values between the two groups. However, the percentile values from the 1st percentile to the 20th percentile APTw signals and the width1-100 APTw signals significantly differed. The highest area under the curve was 0.796, which was obtained from the width1-100 APTw signal values. The sensitivity and specificity values were 64.3% and 88.9%, respectively. APTw imaging was useful to distinguish PCNSL from glioblastoma, IDH-wildtype. To avoid unnecessary aggressive surgical resection, APTw imaging is recommended for cases in which PCNSL is one of the differential diagnoses.
Collapse
Affiliation(s)
- Shigeo Ohba
- Department of Neurosurgery, Fujita Health University School of Medicine, Toyoake 470-1192, Aichi, Japan
| | - Kazuhiro Murayama
- Department of Radiology, Fujita Health University School of Medicine, Toyoake 470-1192, Aichi, Japan
| | - Takao Teranishi
- Department of Neurosurgery, Fujita Health University School of Medicine, Toyoake 470-1192, Aichi, Japan
| | - Masanobu Kumon
- Department of Neurosurgery, Fujita Health University School of Medicine, Toyoake 470-1192, Aichi, Japan
| | - Shunsuke Nakae
- Department of Neurosurgery, Fujita Health University School of Medicine, Toyoake 470-1192, Aichi, Japan
| | - Masao Yui
- Canon Medical Systems Corporation, Otawara 324-8550, Tochigi, Japan
| | - Kaori Yamamoto
- Canon Medical Systems Corporation, Otawara 324-8550, Tochigi, Japan
| | - Seiji Yamada
- Department of Diagnostic Pathology, Fujita Health University School of Medicine, Toyoake 470-1192, Aichi, Japan
| | - Masato Abe
- Department of Pathology, Fujita Health University School of Health Sciences, Toyoake 470-1192, Aichi, Japan
| | - Mitsuhiro Hasegawa
- Department of Neurosurgery, Fujita Health University School of Medicine, Toyoake 470-1192, Aichi, Japan
| | - Yuichi Hirose
- Department of Neurosurgery, Fujita Health University School of Medicine, Toyoake 470-1192, Aichi, Japan
| |
Collapse
|
6
|
Zhu Q, Shen S, Yang C, Li M, Zhang X, Li H, Zhao X, Li M, Cui Y, Ren X, Lin S. A prognostic estimation model based on mRNA-sequence data for patients with oligodendroglioma. Front Neurol 2022; 13:1074593. [PMID: 36588901 PMCID: PMC9795846 DOI: 10.3389/fneur.2022.1074593] [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: 10/19/2022] [Accepted: 11/21/2022] [Indexed: 12/15/2022] Open
Abstract
Background The diagnosis of oligodendroglioma based on the latest World Health Organization Classification of Tumors of the Central Nervous System (WHO CNS 5) criteria requires the codeletion of chromosome arms 1p and 19q and isocitrate dehydrogenase gene (IDH) mutation (mut). Previously identified prognostic indicators may not be completely suitable for patients with oligodendroglioma based on the new diagnostic criteria. To find potential prognostic indicators for oligodendroglioma, we analyzed the expression of mRNAs of oligodendrogliomas in Chinese Glioma Genome Atlas (CGGA). Methods We collected 165 CGGA oligodendroglioma mRNA-sequence datasets and divided them into two cohorts. Patients in the two cohorts were further classified into long-survival and short-survival subgroups. The most predictive mRNAs were filtered out of differentially expressed mRNAs (DE mRNAs) between long-survival and short-survival patients in the training cohort by least absolute shrinkage and selection operator (LASSO), and risk scores of patients were calculated. Univariate and multivariate analyses were performed to screen factors associated with survival and establish the prognostic model. qRT-PCR was used to validate the expression differences of mRNAs. Results A total of 88 DE mRNAs were identified between the long-survival and the short-survival groups in the training cohort. Seven RNAs were selected to calculate risk scores. Univariate analysis showed that risk level, age, and primary-or-recurrent status (PRS) type were statistically correlated with survival and were used as factors to establish a prognostic model for patients with oligodendroglioma. The model showed an optimal predictive accuracy with a C-index of 0.912 (95% CI, 0.679-0.981) and harbored a good agreement between the predictions and observations in both training and validation cohorts. Conclusion We established a prognostic model based on mRNA-sequence data for patients with oligodendroglioma. The predictive ability of this model was validated in a validation cohort, which demonstrated optimal accuracy. The 7 mRNAs included in the model would help predict the prognosis of patients and guide personalized treatment.
Collapse
Affiliation(s)
- Qinghui Zhu
- Department of Neurosurgical Oncology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Shaoping Shen
- Department of Neurosurgical Oncology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Chuanwei Yang
- Department of Neurosurgical Oncology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Mingxiao Li
- Department of Neurosurgical Oncology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Xiaokang Zhang
- Department of Neurosurgical Oncology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Haoyi Li
- Department of Neurosurgical Oncology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Xuzhe Zhao
- Department of Neurosurgical Oncology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Ming Li
- Department of Neurosurgical Oncology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Yong Cui
- Department of Neurosurgical Oncology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China,Department of Neurosurgery, Beijing Neurosurgical Institute, Capital Medical University, Beijing, China
| | - Xiaohui Ren
- Department of Neurosurgical Oncology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China,Department of Neurosurgery, Beijing Neurosurgical Institute, Capital Medical University, Beijing, China
| | - Song Lin
- Department of Neurosurgical Oncology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China,Department of Neurosurgery, Beijing Neurosurgical Institute, Capital Medical University, Beijing, China,*Correspondence: Song Lin
| |
Collapse
|
7
|
Govindarajan V, Shah AH, Di L, Rivas S, Suter RK, Eichberg DG, Luther E, Lu V, Morell AA, Ivan ME, Komotar RJ, Ayad N, De La Fuente M. Systematic Review of Epigenetic Therapies for Treatment of IDH-mutant Glioma. World Neurosurg 2022; 162:47-56. [PMID: 35314408 PMCID: PMC9177782 DOI: 10.1016/j.wneu.2022.03.051] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2022] [Revised: 03/11/2022] [Accepted: 03/12/2022] [Indexed: 12/11/2022]
Abstract
BACKGROUND Isocitrate dehydrogenase (IDH) mutations are present in 70% of World Health Organization grade II and III gliomas. IDH mutation induces accumulation of the oncometabolite 2-hydroxyglutarate. Therefore, therapies targeting reversal of epigenetic dysregulation in gliomas have been suggested. However, the utility of epigenetic treatments in gliomas remains unclear. Here, we present the first clinical systematic review of epigenetic therapies in treatment of IDH-mutant gliomas and highlight their safety and efficacy. METHODS We conducted a systematic search of electronic databases from 2000 to January 2021 following PRISMA guidelines. Articles were screened to include clinical usage of epigenetic therapies in case reports, prospective case series, or clinical trials. Primary and secondary outcomes included safety/tolerability of epigenetic therapies and progression-free survival/overall survival, respectively. RESULTS A total of 133 patients across 8 clinical studies were included in our analysis. IDH inhibitors appear to have the best safety profile, with an overall grade 3/grade 4 adverse event rate of 9%. Response rates to IDH-mutant inhibitors were highest in nonenhancing gliomas (stable disease achieved in 55% of patients). In contrast, histone deacetylase inhibitors demonstrate a lower safety profile with single-study adverse events as high as 28%. CONCLUSION IDH inhibitors appear promising given their benign toxicity profile and ease of monitoring. Histone deacetylase inhibitors appear to have a narrow therapeutic index, as lower concentrations do not appear effective, while increased doses can produce severe immunosuppressive effects. Preliminary data suggest that epigenetic therapies are generally well tolerated and may control disease in certain patient groups, such as those with nonenhancing lesions.
Collapse
Affiliation(s)
- Vaidya Govindarajan
- Department of Neurological Surgery, University of Miami Miller School of Medicine, Miami, Florida, USA
| | - Ashish H Shah
- Department of Neurological Surgery, University of Miami Miller School of Medicine, Miami, Florida, USA.
| | - Long Di
- Department of Neurological Surgery, University of Miami Miller School of Medicine, Miami, Florida, USA
| | - Sarah Rivas
- Surgical Neurology Branch, National Institute of Health, Bethesda, Maryland, USA
| | - Robert K Suter
- Department of Oncology, Georgetown University, Washington, DC, USA
| | - Daniel G Eichberg
- Department of Neurological Surgery, University of Miami Miller School of Medicine, Miami, Florida, USA
| | - Evan Luther
- Department of Neurological Surgery, University of Miami Miller School of Medicine, Miami, Florida, USA
| | - Victor Lu
- Department of Neurological Surgery, University of Miami Miller School of Medicine, Miami, Florida, USA
| | - Alexis A Morell
- Department of Neurological Surgery, University of Miami Miller School of Medicine, Miami, Florida, USA
| | - Michael E Ivan
- Department of Neurological Surgery, University of Miami Miller School of Medicine, Miami, Florida, USA
| | - Ricardo J Komotar
- Department of Neurological Surgery, University of Miami Miller School of Medicine, Miami, Florida, USA
| | - Nagi Ayad
- Department of Oncology, Georgetown University, Washington, DC, USA
| | - Macarena De La Fuente
- Department of Neurology, University of Miami Miller School of Medicine, Miami, Florida, USA
| |
Collapse
|
8
|
Protein Tyrosine Phosphatase Receptor Type Z in Central Nervous System Disease. Int J Mol Sci 2022; 23:ijms23084414. [PMID: 35457233 PMCID: PMC9024684 DOI: 10.3390/ijms23084414] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2022] [Revised: 04/12/2022] [Accepted: 04/14/2022] [Indexed: 01/06/2023] Open
Abstract
Gliomas are among the most common tumors of the central nervous system and include highly malignant subtypes, such as glioblastoma, which are associated with poor prognosis. Effective treatments are therefore urgently needed. Despite the recent advances in neuroimaging technologies, differentiating gliomas from other brain diseases such as multiple sclerosis remains challenging in some patients, and often requires invasive brain biopsy. Protein tyrosine phosphatase receptor type Z (PTPRZ) is a heavily glycosylated membrane protein that is highly expressed in the central nervous system. Several reports analyzing mouse tumor models suggest that PTPRZ may have potential as a therapeutic target for gliomas. A soluble cleaved form of PTPRZ (sPTPRZ) in the cerebrospinal fluid is markedly upregulated in glioma patients, making it another promising diagnostic biomarker. Intriguingly, PTPRZ is also involved in the process of remyelination in multiple sclerosis. Indeed, lowered PTPRZ glycosylation by deletion of the glycosyltransferase gene leads to reduced astrogliosis and enhanced remyelination in mouse models of demyelination. Here, we review the expression, molecular structure, and biological roles of PTPRZ. We also discuss glioma and demyelinating diseases, as well as the pathological role of PTPRZ and its application as a diagnostic marker and therapeutic target.
Collapse
|
9
|
Guo Y, Li Y, Li J, Tao W, Dong W. DNA Methylation-Driven Genes for Developing Survival Nomogram for Low-Grade Glioma. Front Oncol 2022; 11:629521. [PMID: 35111661 PMCID: PMC8801588 DOI: 10.3389/fonc.2021.629521] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2020] [Accepted: 12/20/2021] [Indexed: 12/13/2022] Open
Abstract
Low-grade gliomas (LGG) are heterogeneous, and the current predictive models for LGG are either unsatisfactory or not user-friendly. The objective of this study was to establish a nomogram based on methylation-driven genes, combined with clinicopathological parameters for predicting prognosis in LGG. Differential expression, methylation correlation, and survival analysis were performed in 516 LGG patients using RNA and methylation sequencing data, with accompanying clinicopathological parameters from The Cancer Genome Atlas. LASSO regression was further applied to select optimal prognosis-related genes. The final prognostic nomogram was implemented together with prognostic clinicopathological parameters. The predictive efficiency of the nomogram was internally validated in training and testing groups, and externally validated in the Chinese Glioma Genome Atlas database. Three DNA methylation-driven genes, ARL9, CMYA5, and STEAP3, were identified as independent prognostic factors. Together with IDH1 mutation status, age, and sex, the final prognostic nomogram achieved the highest AUC value of 0.930, and demonstrated stable consistency in both internal and external validations. The prognostic nomogram could predict personal survival probabilities for patients with LGG, and serve as a user-friendly tool for prognostic evaluation, optimizing therapeutic regimes, and managing LGG patients.
Collapse
Affiliation(s)
- Yingyun Guo
- Department of Gastroenterology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Yuan Li
- Department of Oncology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Jiao Li
- Department of Gastroenterology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Weiping Tao
- Department of Oncology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Weiguo Dong
- Department of Gastroenterology, Renmin Hospital of Wuhan University, Wuhan, China
| |
Collapse
|
10
|
Mazurek M, Szczepanek D, Orzyłowska A, Rola R. Analysis of Factors Affecting 5-ALA Fluorescence Intensity in Visualizing Glial Tumor Cells-Literature Review. Int J Mol Sci 2022; 23:ijms23020926. [PMID: 35055109 PMCID: PMC8779265 DOI: 10.3390/ijms23020926] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2021] [Revised: 01/11/2022] [Accepted: 01/13/2022] [Indexed: 01/27/2023] Open
Abstract
Glial tumors are one of the most common lesions of the central nervous system. Despite the implementation of appropriate treatment, the prognosis is not successful. As shown in the literature, maximal tumor resection is a key element in improving therapeutic outcome. One of the methods to achieve it is the use of fluorescent intraoperative navigation with 5-aminolevulinic acid. Unfortunately, often the level of fluorescence emitted is not satisfactory, resulting in difficulties in the course of surgery. This article summarizes currently available knowledge regarding differences in the level of emitted fluorescence. It may depend on both the histological type and the genetic profile of the tumor, which is reflected in the activity and expression of enzymes involved in the intracellular metabolism of fluorescent dyes, such as PBGD, FECH, UROS, and ALAS. The transport of 5-aminolevulinic acid and its metabolites across the blood–brain barrier and cell membranes mediated by transporters, such as ABCB6 and ABCG2, is also important. Accompanying therapies, such as antiepileptic drugs or steroids, also have an impact on light emission by tumor cells. Accurate determination of the factors influencing the fluorescence of 5-aminolevulinic acid-treated cells may contribute to the improvement of fluorescence navigation in patients with highly malignant gliomas.
Collapse
|
11
|
Huang Z, Li G, Li Z, Sun S, Zhang Y, Hou Z, Xie J. Contralesional Structural Plasticity in Different Molecular Pathologic Subtypes of Insular Glioma. Front Neurol 2021; 12:636573. [PMID: 33935941 PMCID: PMC8079625 DOI: 10.3389/fneur.2021.636573] [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: 12/01/2020] [Accepted: 03/08/2021] [Indexed: 12/25/2022] Open
Abstract
Neuroplasticity may preserve neurologic function in insular glioma, thereby improving prognosis following resection. However, the anatomic and molecular bases of this phenomenon are not known. To address this gap in knowledge, the present study investigated contralesional compensation in different molecular pathologic subtypes of insular glioma by high-resolution three-dimensional T1-weighted structural magnetic resonance imaging. A total of 52 patients with insular glioma were examined. We compared the gray matter volume (GMV) of the contralesional insula according to histological grade [low-grade glioma (LGG) and high-grade glioma (HGG)] and molecular pathology status [isocitrate dehydrogenase (IDH) mutation, telomerase reverse-transcriptase (TERT) promoter mutation, and 1p19q codeletion] by voxel-based morphometry (VBM). A cluster of 320 voxels in contralesional insula with higher GMV was observed in glioma with IDH mutation as compared to IDH wild-type tumors by region of interest-based VBM analysis (family-wise error-corrected at p < 0.05). The GMV of the entire contralesional insula was also larger in insular glioma patients with IDH mutation than in patients with wild-type IDH. However, there was no association between histological grade, TERT promoter mutation, or 1p19q codeletion and GMV in the contralesional insula. Thus, IDH mutation is associated with greater structural compensation in insular glioma. These findings may be useful for predicting neurocognitive and functional outcomes in patients undergoing resection surgery.
Collapse
Affiliation(s)
- Zhenxing Huang
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China.,China National Clinical Research Centre for Neurological Diseases, Beijing, China
| | - Gen Li
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China.,China National Clinical Research Centre for Neurological Diseases, Beijing, China
| | - Zhenye Li
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China.,China National Clinical Research Centre for Neurological Diseases, Beijing, China
| | - Shengjun Sun
- China National Clinical Research Centre for Neurological Diseases, Beijing, China.,Neuroimaging Center, Beijing Neurosurgical Institute, Capital Medical University, Beijing, China
| | - Yazhuo Zhang
- China National Clinical Research Centre for Neurological Diseases, Beijing, China.,Beijing Neurosurgical Institute, Capital Medical University, Beijing, China
| | - Zonggang Hou
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China.,China National Clinical Research Centre for Neurological Diseases, Beijing, China
| | - Jian Xie
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China.,China National Clinical Research Centre for Neurological Diseases, Beijing, China
| |
Collapse
|
12
|
Integrated Gene Expression and Methylation Analyses Identify DLL3 as a Biomarker for Prognosis of Malignant Glioma. J Mol Neurosci 2021; 71:1622-1635. [PMID: 33713320 DOI: 10.1007/s12031-021-01817-7] [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: 12/05/2020] [Accepted: 02/15/2021] [Indexed: 12/21/2022]
Abstract
Glioma is one of the most common neurological malignancies worldwide. Delta-like ligand 3 (DLL3), an inhibitory ligand-driven activation of the Notch pathway, has been shown to be significantly associated with overall survival in patients with glioma. Therefore, the purpose of this study was to determine whether DLL3 as a biomarker in glioma is associated with patients' clinicopathological features and prognosis. We identified differences in transcriptome and promoter methylation in the Chinese Glioma Genome Atlas (CGGA) in patients with malignant glioma with shorter (less than 1 year) and longer (greater than 3 years) survival time. Further analysis of The Cancer Genome Atlas (TCGA) revealed that four genes (DLL3, TSPAN15, RTN1, PAK7) are highly associated with patient prognosis and play an indispensable role in evolution. We chose the expression level of DLL3 in glioma patients for our study. Patients were divided into groups with low and high expression of DLL3 according to the cutoff values obtained, and Kaplan-Meier and Cox analysis were used to examine the correlation between DLL3 gene expression and patient survival. We then performed a gene set enrichment analysis (GSEA) to identify significantly enriched signaling pathways. Our results confirmed that the overall survival of patients with low DLL3 expression was significantly shorter than that of patients with high DLL3 expression. GSEA showed that the signaling pathways of the immune process and immune response, among others, were enhanced with the DLL3 low-expression phenotype. Collectively, our findings signify that DLL3 is a potent prognostic factor for glioma, which can provide a viable approach for glioma prognostic assessment and valuable insights for anti-tumor immune-targeted therapies.
Collapse
|
13
|
Ohba S, Murayama K, Kuwahara K, Pareira ES, Nakae S, Nishiyama Y, Adachi K, Yamada S, Sasaki H, Yamamoto N, Abe M, Mukherjee J, Hasegawa M, Pieper RO, Hirose Y. The Correlation of Fluorescence of Protoporphyrinogen IX and Status of Isocitrate Dehydrogenase in Gliomas. Neurosurgery 2021; 87:408-417. [PMID: 31833548 DOI: 10.1093/neuros/nyz524] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2019] [Accepted: 10/01/2019] [Indexed: 11/12/2022] Open
Abstract
BACKGROUND The extent of resection has been reported to be associated with overall survival in gliomas. The use of 5-aminolevulinic acid (5-ALA) has been recognized to increase the extent of tumor resection. OBJECTIVE To evaluate what factors affect the intraoperative fluorescence after administration of 5-ALA in gliomas. METHODS Correlation of intraoperative fluorescence and several clinical, radiographic, molecular biologic, and histopathologic characters was retrospectively evaluated in 104 patients (53 males and 51 females; mean age 54.2 yr) with gliomas at our institution. To clarify the mechanisms that mutant isocitrate dehydrogenase (IDH) affect the intraoperative fluorescence, in Vitro experiments using genetically engineered glioma cells harboring mutant IDH1 were performed. RESULTS Intraoperative fluorescence was observed in 82 patients (78.8%). In addition to age, magnetic resonance imaging enhancement, World Health Organization grades, and MIB-1 index, the status of IDH was revealed to be correlated with intraoperative fluorescence. In Vitro assay revealed that mutant IDH indirectly reduced the amount of exogenous 5-ALA-derived protoporphyrinogen IX in glioma cells by increasing activity of ferrochelatase and heme oxygenase 1. CONCLUSION Mutant IDH1/2-induced metabolite changes of exogenous 5-ALA were suggested to contribute to the lesser intraoperative fluorescence in gliomas with mutant IDH1/2 than in those without.
Collapse
Affiliation(s)
- Shigeo Ohba
- Department of Neurosurgery, Fujita Health University, Toyoake, Japan
| | | | - Kiyonori Kuwahara
- Department of Neurosurgery, Fujita Health University, Toyoake, Japan
| | | | - Shunsuke Nakae
- Department of Neurosurgery, Fujita Health University, Toyoake, Japan
| | - Yuya Nishiyama
- Department of Neurosurgery, Fujita Health University, Toyoake, Japan
| | - Kazuhide Adachi
- Department of Neurosurgery, Fujita Health University, Toyoake, Japan
| | - Seiji Yamada
- Department of Pathology, Fujita Health University, Toyoake, Japan
| | - Hikaru Sasaki
- Department of Neurosurgery, Keio University School of Medicine, Tokyo, Japan
| | - Naoki Yamamoto
- Laboratory of Molecular Biology, Fujita Health University Institute of Joint Research, Toyoake, Japan
| | - Masato Abe
- Department of Pathology, School of Health Sciences, Fujita Health University, Toyoake, Japan
| | - Joydeep Mukherjee
- Department of Neurological Surgery, UCSF Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, California
| | | | - Russell O Pieper
- Department of Neurological Surgery, UCSF Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, California
| | - Yuichi Hirose
- Department of Neurosurgery, Fujita Health University, Toyoake, Japan
| |
Collapse
|
14
|
Correlation between IDH, ATRX, and TERT promoter mutations in glioma. Brain Tumor Pathol 2020; 37:33-40. [PMID: 32227259 DOI: 10.1007/s10014-020-00360-4] [Citation(s) in RCA: 45] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2020] [Accepted: 03/11/2020] [Indexed: 12/12/2022]
Abstract
According to the 2016 World Health Organization (WHO) classification of central nervous system tumors, diffuse astrocytic and oligodendroglial tumors are differentiated by the presence of isocitrate dehydrogenase 1 or 2 (IDH1/2) mutation and the combined loss of the short arm of chromosome 1 and the long arm of chromosome 19 (1p/19q co-deletion). IDH-mutant astrocytoma often has p53 and alpha-thalassemia/mental retardation syndrome X-linked (ATRX) mutation, showing the alternative lengthening of telomeres (ALT) phenotype, while IDH-mutant and 1p/19q-co-deleted oligodendroglioma often have wild-type p53 and telomerase reverse transcriptase (TERT) promoter mutation, showing telomerase activation. This study analyzed IDH, ATRX, and TERT promoter mutations, and the correlation between them. Immortalized cells overcome the telomere-related crisis by activating telomerase or ALT. In glioma, telomerase is mainly activated by TERT promoter mutation, while ALT is usually associated with ATRX mutation. Although the mechanism of how ATRX mutation induces ALT remains unclear, ATRX loss alone is believed to be insufficient to induce ALT. Treatments targeting telomere maintenance are promising.
Collapse
|
15
|
Bhavya B, Anand CR, Madhusoodanan UK, Rajalakshmi P, Krishnakumar K, Easwer HV, Deepti AN, Gopala S. To be Wild or Mutant: Role of Isocitrate Dehydrogenase 1 (IDH1) and 2-Hydroxy Glutarate (2-HG) in Gliomagenesis and Treatment Outcome in Glioma. Cell Mol Neurobiol 2020; 40:53-63. [PMID: 31485826 PMCID: PMC11448936 DOI: 10.1007/s10571-019-00730-3] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2019] [Accepted: 08/23/2019] [Indexed: 12/20/2022]
Abstract
Molecular and clinical research based on isocitrate dehydrogenase (IDH) mutations is much sought after in glioma research since a decade of its discovery in 2008. IDH enzyme normally catalyzes isocitrate to α-keto-glutarate (α-KG), but once the gene is mutated it produces an 'oncometabolite', 2-hydroxyglutarate (2-HG). 2-HG is proposed to inhibit α-KG-dependent dioxygenases and also blocks cellular differentiation. Here, we discuss the role of the IDH1 mutation in gliomagenesis. The review also focuses on the effect of 2-HG on glioma epigenetics, the cellular signaling involved in IDH1 mutant glioma cells and the therapeutic response seen in mutant IDH1(mIDH1) harboring glioma patients in comparison to the patients with wild-type IDH1. The review encompasses the debatable impacts of the mutation on immune microenvironment a propos of various mIDH1 inhibitors in practice or in trials. Recent studies revealing the relation of IDH mutation with the immune microenvironment and inflammatory status in untreated versus treated glioblastoma patients are highlighted with respect to prospective therapeutic targets. Also at the molecular level, the association of mIDH1/2-HG with the intracellular components such as mitochondria and other neighboring cells is discussed.
Collapse
Affiliation(s)
- Bharathan Bhavya
- Department of Biochemistry, Sree Chitra Tirunal Institute for Medical Sciences and Technology, Trivandrum, Kerala, 695011, India
| | - C R Anand
- Department of Biochemistry, Sree Chitra Tirunal Institute for Medical Sciences and Technology, Trivandrum, Kerala, 695011, India
| | - U K Madhusoodanan
- Department of Biochemistry, Sree Chitra Tirunal Institute for Medical Sciences and Technology, Trivandrum, Kerala, 695011, India
| | - P Rajalakshmi
- Department of Pathology, Sree Chitra Tirunal Institute for Medical Sciences and Technology, Trivandrum, Kerala, 695011, India
| | - K Krishnakumar
- Department of Neurosurgery, Sree Chitra Tirunal Institute for Medical Sciences and Technology, Trivandrum, Kerala, 695011, India
| | - H V Easwer
- Department of Neurosurgery, Sree Chitra Tirunal Institute for Medical Sciences and Technology, Trivandrum, Kerala, 695011, India
| | - A N Deepti
- Department of Pathology, Sree Chitra Tirunal Institute for Medical Sciences and Technology, Trivandrum, Kerala, 695011, India
| | - Srinivas Gopala
- Department of Biochemistry, Sree Chitra Tirunal Institute for Medical Sciences and Technology, Trivandrum, Kerala, 695011, India.
| |
Collapse
|
16
|
Laba P, Wang J, Zhang J. Low level of isocitrate dehydrogenase 1 predicts unfavorable postoperative outcomes in patients with clear cell renal cell carcinoma. BMC Cancer 2018; 18:852. [PMID: 30153799 PMCID: PMC6114787 DOI: 10.1186/s12885-018-4747-1] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2018] [Accepted: 08/14/2018] [Indexed: 12/16/2022] Open
Abstract
Background The purpose of this study was to investigate the role of isocitrate dehydrogenase 1 (IDH1) expression on prognosis of patients with clear cell renal cell carcinoma (ccRCC) following nephrectomy. Methods We retrospectively enrolled 358 ccRCC patients undergoing nephrectomy in Renji Hospital. Clinicopathologic features, overall survival (OS) and recurrence-free survival (RFS) of ccRCC patents were all collected. IDH1 expression level was assessed by immunohistochemistry and its association with clinicopathologic features and outcomes were also evaluated. Kaplan-Meier method with the log-rank test was applied to compare survival curves. Multivariate cox regression models were applied to analyze the prognostic value of each factor on OS and RFS of ccRCC patients. Moreover, two nomograms with factors selected by multivariate analysis were constructed to evaluate the prognosis of ccRCC patients, and the calibration plots were built to assess the predictive accuracy of nomograms. Results Our data indicated that IDH1 expression level was down-regulated in ccRCC tissues, and it negatively correlated with tumor Fuhrman grade (p = 0.025). Low IDH1 expression was associated with worse OS and RFS for cccRCC patients (OS, p = 0.004; RFS, p = 0.03). In addition, IDH1 could significantly stratify patients’ OS and RFS in intermediate/high risk patients (UISS score ≥ 4) (p = 0.049 and p = 0.004, respectively). Furthermore, incorporating IDH1 with other prognostic factors could predict ccRCC patients’ OS and RFS (OS, c-index = 0.779; RFS, c-index = 0.798) and perform better than TNM and SSIGN system. Conclusions Low IDH1 expression level might be an adverse prognostic biomarker for clinical outcomes of ccRCC patients, and two nomograms with IDH1 are potential effective prognostic models for ccRCC.
Collapse
Affiliation(s)
- Pingcuo Laba
- Department of Urology, Shigatse People's Hospital, Shigatse, 85700, China
| | - Jianfeng Wang
- Department of Urology, Renji Hospital, School of Medicine, Shanghai Jiaotong University, No.160, Pujian Road, Shanghai, 200127, China
| | - Jin Zhang
- Department of Urology, Shigatse People's Hospital, Shigatse, 85700, China. .,Department of Urology, Renji Hospital, School of Medicine, Shanghai Jiaotong University, No.160, Pujian Road, Shanghai, 200127, China.
| |
Collapse
|
17
|
Association between mutant IDHs and tumorigenesis in gliomas. Med Mol Morphol 2018; 51:194-198. [PMID: 29633022 DOI: 10.1007/s00795-018-0189-8] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2018] [Accepted: 04/02/2018] [Indexed: 01/14/2023]
Abstract
To become immortalized, cells need to maintain the telomere length via the activation of telomerase or alternative lengthening of telomere. Mutations in IDH1/2 are strongly associated with the early stage of gliomagenesis. Previous work has shown that the accumulation of 2-HG, which is induced by mutant IDH1/2, inhibits α-KG-dependent deoxygenase and leads to genome-wide histone and DNA methylation alterations. These alterations are believed to contribute to tumorigenesis. H-Ras can transform human astrocytes with the inactivation of p53/pRb and expression of hTERT; however, mutant IDH1 can also transform cells. Moreover, mutant IDH1 can drive the immortalization and transformation of p53-/pRb-deficient astrocytes by reactivating telomerase and stabilizing telomeres in combination with increased histone lysine methylation and c-Myc/Max binding at the TERT promoter. It remains unclear whether mutant IDH1/2 acts only as the initial driver of gliomagenesis or it maintains transformed cells. Clinical studies are being performed to assess the use of mutant IDH1/2 inhibitors for treating gliomas.
Collapse
|
18
|
Guadagno E, Presta I, Maisano D, Donato A, Pirrone CK, Cardillo G, Corrado SD, Mignogna C, Mancuso T, Donato G, Del Basso De Caro M, Malara N. Role of Macrophages in Brain Tumor Growth and Progression. Int J Mol Sci 2018; 19:ijms19041005. [PMID: 29584702 PMCID: PMC5979398 DOI: 10.3390/ijms19041005] [Citation(s) in RCA: 81] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2018] [Revised: 03/10/2018] [Accepted: 03/23/2018] [Indexed: 12/16/2022] Open
Abstract
The role of macrophages in the growth and the progression of tumors has been extensively studied in recent years. A large body of data demonstrates that macrophage polarization plays an essential role in the growth and progression of brain tumors, such as gliomas, meningiomas, and medulloblastomas. The brain neoplasm cells have the ability to influence the polarization state of the tumor associated macrophages. In turn, innate immunity cells have a decisive role through regulation of the acquired immune response, but also through humoral cross-talking with cancer cells in the tumor microenvironment. Neoangiogenesis, which is an essential element in glial tumor progression, is even regulated by the tumor associated macrophages, whose activity is linked to other factors, such as hypoxia. In addition, macrophages play a decisive role in establishing the entry into the bloodstream of cancer cells. As is well known, the latter phenomenon is also present in brain tumors, even if they only rarely metastasize. Looking ahead in the future, we can imagine that characterizing the relationships between tumor and tumor associated macrophage, as well as the study of circulating tumor cells, could give us useful tools in prognostic evaluation and therapy. More generally, the study of innate immunity in brain tumors can boost the development of new forms of immunotherapy.
Collapse
Affiliation(s)
- Elia Guadagno
- Department of Advanced Biomedical Sciences-Pathology Section, University of Naples "Federico II"-via Pansini 5, 80131 Naples, Italy.
| | - Ivan Presta
- Department of Health Sciences, University of Catanzaro "Magna Græcia"-viale Europa, 88100 Catanzaro, Italy.
| | - Domenico Maisano
- Department of Health Sciences, University of Catanzaro "Magna Græcia"-viale Europa, 88100 Catanzaro, Italy.
| | - Annalidia Donato
- Department of Medical and Surgical Sciences-University of Catanzaro "Magna Graecia"-viale Europa, 88100 Catanzaro, Italy.
| | - Caterina Krizia Pirrone
- Department of Health Sciences, University of Catanzaro "Magna Græcia"-viale Europa, 88100 Catanzaro, Italy.
| | - Gabriella Cardillo
- Department of Health Sciences, University of Catanzaro "Magna Græcia"-viale Europa, 88100 Catanzaro, Italy.
| | - Simona Domenica Corrado
- Department of Health Sciences, University of Catanzaro "Magna Græcia"-viale Europa, 88100 Catanzaro, Italy.
| | - Chiara Mignogna
- Department of Health Sciences, University of Catanzaro "Magna Græcia"-viale Europa, 88100 Catanzaro, Italy.
| | - Teresa Mancuso
- Department of Medical and Surgical Sciences-University of Catanzaro "Magna Graecia"-viale Europa, 88100 Catanzaro, Italy.
| | - Giuseppe Donato
- Department of Health Sciences, University of Catanzaro "Magna Græcia"-viale Europa, 88100 Catanzaro, Italy.
| | - Marialaura Del Basso De Caro
- Department of Advanced Biomedical Sciences-Pathology Section, University of Naples "Federico II"-via Pansini 5, 80131 Naples, Italy.
| | - Natalia Malara
- Department of Clinical and Experimental Medicine-University of Catanzaro "Magna Graecia"-viale Europa, 88100 Catanzaro, Italy.
| |
Collapse
|