1
|
Dharmaiah S, Malgulwar PB, Johnson WE, Chen BA, Sharin V, Whitfield BT, Alvarez C, Tadimeti V, Farooqi AS, Huse JT. G-quadruplex stabilizer CX-5461 effectively combines with radiotherapy to target α-thalassemia/mental retardation X-linked-deficient malignant glioma. Neuro Oncol 2025; 27:932-947. [PMID: 39570009 PMCID: PMC12083236 DOI: 10.1093/neuonc/noae248] [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: 03/18/2024] [Indexed: 11/22/2024] Open
Abstract
BACKGROUND Inactivation of α-thalassemia/mental retardation X-linked (ATRX) represents a defining molecular feature in large subsets of malignant glioma. ATRX deficiency gives rise to abnormal G-quadruplex (G4) DNA secondary structures, enhancing replication stress and genomic instability. Building on earlier work, we evaluated the extent to which pharmacological G4 stabilization selectively enhances DNA damage and cell death in ATRX-deficient preclinical glioma models. METHODS Using the G4 stabilizer CX-5461, we treated patient-derived glioma stem cells (GSCs) in vitro and GSC flank and intracranial murine xenografts in vivo to evaluate efficacy as both a single agent and in combination with ionizing radiation (IR), the latter a central element of current treatment standards. RESULTS CX-5461 promoted dose-sensitive lethality in ATRX-deficient GSCs relative to ATRX-intact controls. Mechanistic studies revealed that CX-5461 disrupted histone variant H3.3 deposition, enhanced replication stress and DNA damage, activated p53-independent apoptosis, and induced G2/M arrest to a greater extent in ATRX-deficient GSCs than in ATRX-intact counterparts. These data were corroborated in vivo, where CX-5461/IR treatment profoundly delayed tumor growth and prolonged survival in mice bearing ATRX-deficient flank xenografts. Histopathological analyses revealed decreased proliferation, increased apoptosis, and significant G4 induction, replication stress, and DNA damage in CX-5461-treated tumors, both alone and in combination with IR. Finally, despite suboptimal blood-brain-barrier penetration, systemic CX-5461 treatment induced tangible pharmacodynamic effects in ATRX-deficient intracranial GSC models. CONCLUSIONS In totality, our work substantively demonstrates efficacy and defines mechanisms of action for G4 stabilization as a novel therapeutic strategy targeting ATRX-deficient malignant glioma, laying the groundwork for clinical translation.
Collapse
Affiliation(s)
- Sharvari Dharmaiah
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
- Graduate School of Biomedical Sciences, Cancer Biology, The University of Texas MD Anderson Cancer Center UTHealth Houston, Houston, Texas, USA
| | - Prit Benny Malgulwar
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - William E Johnson
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Brandon A Chen
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Vladislav Sharin
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Benjamin T Whitfield
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
- Graduate School of Biomedical Sciences, Cancer Biology, The University of Texas MD Anderson Cancer Center UTHealth Houston, Houston, Texas, USA
| | - Christian Alvarez
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Vasudev Tadimeti
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Ahsan S Farooqi
- Division of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Jason T Huse
- Department of Pathology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| |
Collapse
|
2
|
Ghadimi K, Abbas I, Karandish A, Crisman C, Eskandar EN, Kobets AJ. Cognitive Decline in Glioblastoma (GB) Patients with Different Treatment Modalities and Insights on Untreated Cases. Curr Oncol 2025; 32:152. [PMID: 40136356 PMCID: PMC11940939 DOI: 10.3390/curroncol32030152] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2025] [Revised: 02/28/2025] [Accepted: 03/04/2025] [Indexed: 03/27/2025] Open
Abstract
BACKGROUND Cognitive decline is common in patients with Glioblastoma (GB), occurring in both treated and untreated cases. It frequently presents as impairments in memory, attention, language, or other cognitive functions. In addition, these cognitive deficits can affect quality of life, functional independence, and overall survival, and they are associated with psychiatric conditions such as anxiety and depression. METHODS This narrative review evaluates cognitive deficits in GB patients, both with and without treatment. It also explores the impact of tumor features such as size, location, and histology, along with patient characteristics such as age and education, and discusses the effects of standard therapies, such as surgery, chemotherapy, and radiotherapy, on cognitive outcomes. RESULTS Cognitive impairment in GB is influenced by tumor- and patient-specific factors, as well as treatment modalities. Initially, combination therapies such as surgery, radiotherapy, and chemotherapy may improve cognitive domains by reducing tumor burden, relieving cerebral edema, and reducing mass effects, subsequently bringing indirect effects of improved mental health and mood. While certain treatments like radiotherapy and chemotherapy carry risks of delayed neurotoxicity, studies indicate that, on balance, treated patients generally show better preservation or improvement in cognitive function than those who go untreated. However, excessive treatment aggressiveness and cumulative neurotoxic effects may diminish cognitive benefits. CONCLUSION Cognitive function is an independent factor in GB, which could affect survival in GB patients, therefore making routine cognitive assessments essential for prognosis, treatment planning, and rehabilitation. Neuroprotective agents, cognitive rehabilitation, and personalized, multidisciplinary strategies can help optimize both survival and cognitive preservation.
Collapse
|
3
|
Kurdi M, Alkhotani A, Alsinani T, Alkhayyat S, Katib Y, Jastaniah Z, Sabbagh AJ, Butt NS, Toonsi FA, Alharbi M, Baeesa S. Effects of Radiotherapy Alone Versus Concomitant Radiotherapy With Temozolomide Chemotherapy on the Outcome of IDH-wildtype Glioblastoma Patients. Clin Oncol (R Coll Radiol) 2025; 38:103741. [PMID: 39742692 DOI: 10.1016/j.clon.2024.103741] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2024] [Revised: 10/25/2024] [Accepted: 12/13/2024] [Indexed: 01/04/2025]
Abstract
BACKGROUND Isocitrate dehydrogenase [IDH]-wildtype glioblastoma is an aggressive brain cancer associated with high recurrence and poor overall survival. AIM Our study aims to explore the prognostic effects of radiotherapy [RT] alone versus concomitant RT with temozolomide [TMZ]. METHODS A multicentre retrospective study included a cohort of 244 patients diagnosed with IDH-wildtype glioblastoma, and it was analysed from 2013 to 2020. All patients underwent complete surgical resection of the tumour followed by standard postsurgical therapies, including RT alone [group A] or concomitant RT with TMZ chemotherapy [group B]. Intra-statistical cohort data analysis was performed. RESULTS The mean age of the patients was 53.9 years [SD 16.3 years], with 87 [35.7%] females and 157 [64.3%] males. Group "A" patient [n = 67, 27.5%] received RT alone, and group "B" patient [n = 177, 72.5%] received concomitant RT with TMZ chemotherapy. All patients' mean progression-free survival [PFS] was 391.8 days (13.1 months). There was a statistically significant difference in PFS between the two treatment groups [P value<0.0001]. The hazard ratio [HR] for PFS in group "b" compared with group "a" was 0.48 [95% CI: 0.36-0.64, P < 0.001] in the univariable analysis, indicating a significant benefit of the combined treatment. This benefit was maintained in the multivariable analysis with an HR of 0.50 [95% CI: 0.37-0.67, P < 0.001]. Age was found to be a significant factor in PFS, with each additional year of age increasing HR by 2% in the univariable analysis [HR: 1.02, 95% CI: 1.01-1.03, P < 0.001] and the multivariable analysis (HR of 1.01 [95% CI: 1.01-1.02, P < 0.001)]. CONCLUSIONS Concomitant RT with TMZ chemotherapy significantly increased PFS beyond that observed from isolated RT in patients with IDH-wildtype glioblastoma.
Collapse
Affiliation(s)
- M Kurdi
- Department of Pathology, Faculty of Medicine, King Abdulaziz University, Rabigh, Saudi Arabia.
| | - A Alkhotani
- Department of Pathology, College of Medicine, Umm Al-Qura University, Makkah, Saudi Arabia
| | - T Alsinani
- Department of Neurosurgery, King Fahad General Hospital, Jeddah, Saudi Arabia
| | - S Alkhayyat
- Department of Internal Medicine, Faculty of Medicine, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Y Katib
- Department of Internal Medicine, Faculty of Medicine, Taibah University, Almadinah, Saudi Arabia
| | - Z Jastaniah
- Department of Internal Medicine, Faculty of Medicine, King Abdulaziz University, Rabigh, Saudi Arabia
| | - A J Sabbagh
- Department of Surgery, Faculty of Medicine, King Abdulaziz University, Jeddah, Saudi Arabia
| | - N S Butt
- Department of Community Medicine, Faculty of Medicine, King Abdulaziz University, Rabigh, Saudi Arabia
| | - F A Toonsi
- Department of Radiology, Faculty of Medicine, King Abdulaziz University, Jeddah, Saudi Arabia
| | - M Alharbi
- Oncology Center, East Jeddah Hospital, First Health Cluster, Jeddah, Saudi Arabia
| | - S Baeesa
- Department of Neurosciences, King Faisal Specialist Hospital and Research Center, Jeddah, Saudi Arabia
| |
Collapse
|
4
|
Juarez TM, Gill JM, Minev BR, Sharma A, Kesari S. Neoadjuvant clinical trials in adults with newly diagnosed high-grade glioma: A systematic review. Crit Rev Oncol Hematol 2025; 206:104596. [PMID: 39675399 DOI: 10.1016/j.critrevonc.2024.104596] [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: 11/13/2024] [Accepted: 12/10/2024] [Indexed: 12/17/2024] Open
Abstract
BACKGROUND High-grade gliomas are devastating cancers that remain incurable with standard surgical resection and radiochemotherapy. Although beneficial against neoplasms, radiation lowers lymphocyte counts, weakens immune activation, and recruits suppressive myeloid cells impairing immune responses. Tumor environments treated with radiation experience long-term immunosuppression, reducing immunotherapy effectiveness and contributing to recurrence. Investigating pre-radiation treatments in newly diagnosed patients could identify active agents, assess immunotherapy impact, and enable multiomic analyses without radiation-induced confounding factors. This literature review was conducted to describe the feasibility, safety, and outcomes of postsurgical, pre-radiation clinical trials for adults with newly diagnosed high-grade glioma. METHODS A systematic review was performed of the English-language literature reporting results of clinical trials for adults with newly diagnosed high-grade glioma administered postsurgical treatment prior to radiation therapy. A search was conducted in PubMed and references cited in research and review articles were also considered. RESULTS From 1991 to 2024, 52 clinical trials were identified: 3 phase I, 38 phase II, 4 phase III, and 7 of unknown phase. Nine trials were randomized, 24 were multicenter trials, 21 investigated temozolomide-containing regimens, and 12 focused on inoperable tumors, involving a total of 2737 patients. CONCLUSION Pre-radiation neoadjuvant studies are feasible and may identify active drugs. This is particularly relevant in the era of personalized medicine with brain-penetrant drugs, targeted therapy, and immuno-oncology advancements. Investigating pre-radiation treatments in newly diagnosed high-grade glioma is a viable approach to rapidly identify active and inactive regimens while the immune system and tumor microenvironment remain intact.
Collapse
Affiliation(s)
| | | | - Boris R Minev
- Calidi Biotherapeutics, San Diego, CA, USA; Department of Radiation Medicine and Applied Sciences, University of California, San Diego, CA 92093, USA
| | - Akanksha Sharma
- Pacific Neuroscience Institute, Santa Monica, CA, USA; Department of Translational Neuroscience, Saint John's Cancer Institute at Providence Saint John's Health Center, Santa Monica, CA, USA
| | - Santosh Kesari
- Pacific Neuroscience Institute, Santa Monica, CA, USA; Department of Translational Neuroscience, Saint John's Cancer Institute at Providence Saint John's Health Center, Santa Monica, CA, USA.
| |
Collapse
|
5
|
Pascoe M, Byrne E, King A, Cooper D, Foldvary-Schaefer N, Mehra R, Lathia J, Gilbert MR, Armstrong TS. Sleep disorders associated with cranial radiation-A systematic review. Neuro Oncol 2025; 27:63-76. [PMID: 39468721 PMCID: PMC11726243 DOI: 10.1093/neuonc/noae174] [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: 02/13/2024] [Indexed: 10/30/2024] Open
Abstract
BACKGROUND Radiation is the standard-of-care treatment for primary brain tumors (PBTs) but may have profound effects on sleep that have not yet been fully characterized. This systematic review aims to further our understanding of radiation therapy on the risk of development of sleep disorders in patients with PBTs, as well as potential opportunities for prevention and treatment. METHODS A systematic search of PubMed, Embase, and Web of Science was performed (last Jan 2024) with predefined inclusion (PBT patients, radiation therapy, somnolence/circadian disruption) and exclusion (reviews/abstracts/cases/chapters, non-PBT cancer, lack of radiation) criteria, yielding 267 papers initially and 38 studies included. Data extraction and analysis (descriptive statistics, individual study summary) focused on the incidence of sleep disturbances, radiation types/doses, and pharmacologic interventions. Risk of bias assessment was conducted with the Effective Public Health Practice Project's Quality Assessment Tool for Quantitative Studies. RESULTS The included 38 studies (n = 2948 patients) demonstrated a high incidence of sleep disturbances in patients with PBTs throughout radiation therapy, but primarily from the end of radiation to 6 months after. Sleep symptoms were associated with radiation (dose-dependent), and pharmacotherapies were helpful in patients with formal sleep disorder diagnoses. Terminology and incidence reporting of sleep symptoms are inconsistent, and many studies had a high risk of bias. CONCLUSIONS This systematic review highlights the ongoing challenges with sleep symptoms/disorders related to cranial irradiation treatment in the primary brain tumor population. Further investigations on the interconnectedness of sleep disturbance constructs and possible pharmacotherapies to alleviate symptoms are warranted.
Collapse
Affiliation(s)
- Maeve Pascoe
- Neuro-Oncology Branch, Center for Cancer Research, National Institutes of Health Neuro-oncology Branch, Bethesda, Maryland, USA
| | - Emma Byrne
- Neuro-Oncology Branch, Center for Cancer Research, National Institutes of Health Neuro-oncology Branch, Bethesda, Maryland, USA
| | - Amanda King
- Neuro-Oncology Branch, Center for Cancer Research, National Institutes of Health Neuro-oncology Branch, Bethesda, Maryland, USA
| | - Diane Cooper
- Division of Library Services, National Institutes of Health, National Institutes of Health Library, Bethesda, Maryland, USA
| | - Nancy Foldvary-Schaefer
- Lerner Research Institute and Neurological Institute, Cleveland Clinic, Cleveland, Ohio, USA
| | - Reena Mehra
- Lerner Research Institute and Neurological Institute, Cleveland Clinic, Cleveland, Ohio, USA
| | - Justin Lathia
- Lerner Research Institute and Neurological Institute, Cleveland Clinic, Cleveland, Ohio, USA
| | - Mark R Gilbert
- Neuro-Oncology Branch, Center for Cancer Research, National Institutes of Health Neuro-oncology Branch, Bethesda, Maryland, USA
| | - Terri S Armstrong
- Neuro-Oncology Branch, Center for Cancer Research, National Institutes of Health Neuro-oncology Branch, Bethesda, Maryland, USA
| |
Collapse
|
6
|
Jackson MR, Richards AR, Oladipupo ABA, Chahal SK, Caragher S, Chalmers AJ, Gomez-Roman N. ClonoScreen3D - A Novel 3-Dimensional Clonogenic Screening Platform for Identification of Radiosensitizers for Glioblastoma. Int J Radiat Oncol Biol Phys 2024; 120:162-177. [PMID: 38493899 DOI: 10.1016/j.ijrobp.2024.02.046] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2023] [Revised: 02/13/2024] [Accepted: 02/18/2024] [Indexed: 03/19/2024]
Abstract
PURPOSE Glioblastoma (GBM) is a lethal brain tumor. Standard-of-care treatment comprising surgery, radiation, and chemotherapy results in median survival rates of 12 to 15 months. Molecular-targeted agents identified using conventional 2-dimensional (2D) in vitro models of GBM have failed to improve outcome in patients, rendering such models inadequate for therapeutic target identification. A previously developed 3D GBM in vitro model that recapitulates key GBM clinical features and responses to molecular therapies was investigated for utility for screening novel radiation-drug combinations using gold-standard clonogenic survival as readout. METHODS AND MATERIALS Patient-derived GBM cell lines were optimized for inclusion in a 96-well plate 3D clonogenic screening platform, ClonoScreen3D. Radiation responses of GBM cells in this system were highly reproducible and comparable to those observed in low-throughout 3D assays. The screen methodology provided quantification of candidate drug single agent activity (half maximal effective concentration or EC50) and the interaction between drug and radiation (radiation interaction ratio). RESULTS The poly(ADP-ribose) polymerase inhibitors talazoparib, rucaparib, and olaparib each showed a significant interaction with radiation by ClonoScreen3D and were subsequently confirmed as true radiosensitizers by full clonogenic assay. Screening a panel of DNA damage response inhibitors revealed the expected propensity of these compounds to interact significantly with radiation (13/15 compounds). A second screen assessed a panel of compounds targeting pathways identified by transcriptomic analysis and demonstrated single agent activity and a previously unreported interaction with radiation of dinaciclib and cytarabine (radiation interaction ratio 1.28 and 1.90, respectively). These compounds were validated as radiosensitizers in full clonogenic assays (sensitizer enhancement ratio 1.47 and 1.35, respectively). CONCLUSIONS The ClonoScreen3D platform was demonstrated to be a robust method to screen for single agent and radiation-drug combination activity. Using gold-standard clonogenicity, this assay is a tool for identification of radiosensitizers. We anticipate this technology will accelerate identification of novel radiation-drug combinations with genuine translational value.
Collapse
Affiliation(s)
- Mark R Jackson
- Wolfson Wohl Cancer Research Centre, School of Cancer Sciences, University of Glasgow, Glasgow, UK
| | - Amanda R Richards
- Wolfson Wohl Cancer Research Centre, School of Cancer Sciences, University of Glasgow, Glasgow, UK
| | | | - Sandeep K Chahal
- Wolfson Wohl Cancer Research Centre, School of Cancer Sciences, University of Glasgow, Glasgow, UK
| | - Seamus Caragher
- Wolfson Wohl Cancer Research Centre, School of Cancer Sciences, University of Glasgow, Glasgow, UK; Division of Plastic and Reconstructive Surgery, Department of Surgery, Massachusetts General Hospital, Massachussetts, USA
| | - Anthony J Chalmers
- Wolfson Wohl Cancer Research Centre, School of Cancer Sciences, University of Glasgow, Glasgow, UK
| | - Natividad Gomez-Roman
- Wolfson Wohl Cancer Research Centre, School of Cancer Sciences, University of Glasgow, Glasgow, UK; Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, Glasgow, UK.
| |
Collapse
|
7
|
Mei Q, Shen H, Chai X, Jiang Y, Liu J. Practical Nomograms and Risk Stratification System for Predicting the Overall and Cancer-specific Survival in Patients with Anaplastic Astrocytoma. World Neurosurg 2024; 189:e391-e403. [PMID: 38909753 DOI: 10.1016/j.wneu.2024.06.076] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2024] [Accepted: 06/16/2024] [Indexed: 06/25/2024]
Abstract
OBJECTIVE Anaplastic astrocytoma (AA) is an uncommon primary brain tumor with highly variable clinical outcomes. Our study aimed to develop practical tools for clinical decision-making in a population-based cohort study. METHODS Data from 2997 patients diagnosed with AA between 2004 and 2015 were retrospectively extracted from the Surveillance, Epidemiology, and End Results database. The Least Absolute Shrinkage and Selection Operator and multivariate Cox regression analyses were applied to select factors and establish prognostic nomograms. The discriminatory ability of these nomogram models was evaluated using the concordance index and receiver operating characteristic curve. Risk stratifications were established based on the nomograms. RESULTS Selected 2997 AA patients were distributed into the training cohort (70%, 2097) and the validation cohort (30%, 900). Age, household income, tumor site, extension, surgery, radiotherapy, and chemotherapy were identified as independent prognostic factors for both overall survival (OS) and cancer-specific survival (CSS). In the training cohort, our nomograms for OS and CSS exhibited good predictive accuracy with concordance index values of 0.752 (95% CI: 0.741-0.764) and 0.753 (95% CI: 0.741-0.765), respectively. Calibration and decision curve analyses curves showed that the nomograms demonstrated considerable consistency and satisfactory clinical utilities. With the establishment of nomograms, we stratified AA patients into high- and low-risk groups, and constructed risk stratification systems for OS and CSS. CONCLUSIONS We constructed two predictive nomograms and risk classification systems to effectively predict the OS and CSS rates in AA patients. These models were internally validated with considerable accuracy and reliability and might be helpful in future clinical practices.
Collapse
Affiliation(s)
- Qing Mei
- Department of Neurology, Beijing Pinggu Hospital, Beijing, China
| | - Hui Shen
- Department of Interventional Neuroradiology, Sanbo Brain Hospital, Capital Medical University, Beijing, China; Beijing Neurosurgical Institute, Capital Medical University, Department of Interventional Neuroradiology, Beijing Tiantan Hospital, Beijing, China
| | - Xubin Chai
- Beijing Neurosurgical Institute, Capital Medical University, Department of Interventional Neuroradiology, Beijing Tiantan Hospital, Beijing, China; State Key Laboratory of Brain and Cognitive Science, Institute of Biophysics, Chinese Academy of Sciences, Beijing, China; University of Chinese Academy of Sciences, Beijing, China
| | - Yuanfeng Jiang
- Department of Interventional Neuroradiology, Sanbo Brain Hospital, Capital Medical University, Beijing, China
| | - Jiachun Liu
- Department of Interventional Neuroradiology, Sanbo Brain Hospital, Capital Medical University, Beijing, China.
| |
Collapse
|
8
|
Rahman R, Shi DD, Reitman ZJ, Hamerlik P, de Groot JF, Haas-Kogan DA, D’Andrea AD, Sulman EP, Tanner K, Agar NYR, Sarkaria JN, Tinkle CL, Bindra RS, Mehta MP, Wen PY. DNA damage response in brain tumors: A Society for Neuro-Oncology consensus review on mechanisms and translational efforts in neuro-oncology. Neuro Oncol 2024; 26:1367-1387. [PMID: 38770568 PMCID: PMC11300028 DOI: 10.1093/neuonc/noae072] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/22/2024] Open
Abstract
DNA damage response (DDR) mechanisms are critical to maintenance of overall genomic stability, and their dysfunction can contribute to oncogenesis. Significant advances in our understanding of DDR pathways have raised the possibility of developing therapies that exploit these processes. In this expert-driven consensus review, we examine mechanisms of response to DNA damage, progress in development of DDR inhibitors in IDH-wild-type glioblastoma and IDH-mutant gliomas, and other important considerations such as biomarker development, preclinical models, combination therapies, mechanisms of resistance and clinical trial design considerations.
Collapse
Affiliation(s)
- Rifaquat Rahman
- Department of Radiation Oncology, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Diana D Shi
- Department of Radiation Oncology, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Zachary J Reitman
- Department of Radiation Oncology, Duke University Medical Center, Durham, North Carolina, USA
| | - Petra Hamerlik
- Division of Cancer Sciences, University of Manchester, Manchester, UK
| | - John F de Groot
- Division of Neuro-Oncology, University of California San Francisco, San Francisco, California, USA
| | - Daphne A Haas-Kogan
- Department of Radiation Oncology, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Alan D D’Andrea
- Department of Radiation Oncology, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Erik P Sulman
- Department of Radiation Oncology, New York University, New York, New York, USA
| | - Kirk Tanner
- National Brain Tumor Society, Newton, Massachusetts, USA
| | - Nathalie Y R Agar
- Department of Neurosurgery and Department of Radiology, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Jann N Sarkaria
- Department of Radiation Oncology, Mayo Clinic, Rochester, Minnesota, USA
| | - Christopher L Tinkle
- Department of Radiation Oncology, St. Jude Children’s Research Hospital, Memphis, Tennessee, USA
| | - Ranjit S Bindra
- Department of Therapeutic Radiology, Yale University, New Haven, Connecticut, USA
| | - Minesh P Mehta
- Miami Cancer Institute, Baptist Hospital, Miami, Florida, USA
| | - Patrick Y Wen
- Center for Neuro-Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts, USA
| |
Collapse
|
9
|
Jiao C, Lao Y, Zhang W, Braunstein S, Salans M, Villanueva-Meyer JE, Hervey-Jumper SL, Yang B, Morin O, Valdes G, Fan Z, Shiroishi M, Zada G, Sheng K, Yang W. Multi-modal fusion and feature enhancement U-Net coupling with stem cell niches proximity estimation for voxel-wise GBM recurrence prediction . Phys Med Biol 2024; 69:10.1088/1361-6560/ad64b8. [PMID: 39019073 PMCID: PMC11308744 DOI: 10.1088/1361-6560/ad64b8] [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: 02/04/2024] [Accepted: 07/17/2024] [Indexed: 07/19/2024]
Abstract
Objective.We aim to develop a Multi-modal Fusion and Feature Enhancement U-Net (MFFE U-Net) coupling with stem cell niche proximity estimation to improve voxel-wise Glioblastoma (GBM) recurrence prediction.Approach.57 patients with pre- and post-surgery magnetic resonance (MR) scans were retrospectively solicited from 4 databases. Post-surgery MR scans included two months before the clinical diagnosis of recurrence and the day of the radiologicaly confirmed recurrence. The recurrences were manually annotated on the T1ce. The high-risk recurrence region was first determined. Then, a sparse multi-modal feature fusion U-Net was developed. The 50 patients from 3 databases were divided into 70% training, 10% validation, and 20% testing. 7 patients from the 4th institution were used as external testing with transfer learning. Model performance was evaluated by recall, precision, F1-score, and Hausdorff Distance at the 95% percentile (HD95). The proposed MFFE U-Net was compared to the support vector machine (SVM) model and two state-of-the-art neural networks. An ablation study was performed.Main results.The MFFE U-Net achieved a precision of 0.79 ± 0.08, a recall of 0.85 ± 0.11, and an F1-score of 0.82 ± 0.09. Statistically significant improvement was observed when comparing MFFE U-Net with proximity estimation couple SVM (SVMPE), mU-Net, and Deeplabv3. The HD95 was 2.75 ± 0.44 mm and 3.91 ± 0.83 mm for the 10 patients used in the model construction and 7 patients used for external testing, respectively. The ablation test showed that all five MR sequences contributed to the performance of the final model, with T1ce contributing the most. Convergence analysis, time efficiency analysis, and visualization of the intermediate results further discovered the characteristics of the proposed method.Significance. We present an advanced MFFE learning framework, MFFE U-Net, for effective voxel-wise GBM recurrence prediction. MFFE U-Net performs significantly better than the state-of-the-art networks and can potentially guide early RT intervention of the disease recurrence.
Collapse
Affiliation(s)
- Changzhe Jiao
- Department of Radiation Oncology, UC San Francisco, San Francisco, CA 94143
| | - Yi Lao
- Department of Radiation Oncology, UC Los Angeles, Los Angeles, CA 90095
| | - Wenwen Zhang
- Department of Radiation Oncology, UC San Francisco, San Francisco, CA 94143
| | - Steve Braunstein
- Department of Radiation Oncology, UC San Francisco, San Francisco, CA 94143
| | - Mia Salans
- Department of Radiation Oncology, UC San Francisco, San Francisco, CA 94143
| | | | | | - Bo Yang
- Department of Radiation Oncology, UC San Francisco, San Francisco, CA 94143
| | - Olivier Morin
- Department of Radiation Oncology, UC San Francisco, San Francisco, CA 94143
| | - Gilmer Valdes
- Department of Radiation Oncology, UC San Francisco, San Francisco, CA 94143
| | - Zhaoyang Fan
- Department of Radiology, University of Southern California, Los Angeles, CA 90033
| | - Mark Shiroishi
- Department of Radiology, University of Southern California, Los Angeles, CA 90033
| | - Gabriel Zada
- Department of Neurosurgery, University of Southern California, Los Angeles, CA 90033
| | - Ke Sheng
- Department of Radiation Oncology, UC San Francisco, San Francisco, CA 94143
| | - Wensha Yang
- Department of Radiation Oncology, UC San Francisco, San Francisco, CA 94143
| |
Collapse
|
10
|
Janneh AH. Sphingolipid Signaling and Complement Activation in Glioblastoma: A Promising Avenue for Therapeutic Intervention. BIOCHEM 2024; 4:126-143. [PMID: 38894892 PMCID: PMC11185840 DOI: 10.3390/biochem4020007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 06/21/2024]
Abstract
Glioblastoma is the most common and aggressive type of malignant brain tumor with a poor prognosis due to the lack of effective treatment options. Therefore, new treatment options are required. Sphingolipids are essential components of the cell membrane, while complement components are integral to innate immunity, and both play a critical role in regulating glioblastoma survival signaling. This review focuses on recent studies investigating the functional roles of sphingolipid metabolism and complement activation signaling in glioblastoma. It also discusses how targeting these two systems together may emerge as a novel therapeutic approach.
Collapse
Affiliation(s)
- Alhaji H Janneh
- Hollings Cancer Center, Department of Biochemistry and Molecular Biology, Medical University of South Carolina, Charleston, SC 29425, USA
| |
Collapse
|
11
|
Lee PY, Wei YT, Chao KSC, Chu CN, Chung WH, Wang TH. Anti-epileptic drug use during adjuvant chemo-radiotherapy is associated with poorer survival in patients with glioblastoma: A nationwide population-based cohort study. J Cancer Res Ther 2024; 20:555-562. [PMID: 38687925 DOI: 10.4103/jcrt.jcrt_750_22] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2022] [Accepted: 10/31/2022] [Indexed: 05/02/2024]
Abstract
INTRODUCTION There are emerging but inconsistent evidences about anti-epileptic drugs (AEDs) as radio- or chemo-sensitizers to improve survival in glioblastoma patients. We conducted a nationwide population-based study to evaluate the impact of concurrent AED during post-operative chemo-radiotherapy on outcome. MATERIAL AND METHODS A total of 1057 glioblastoma patients were identified by National Health Insurance Research Database and Cancer Registry in 2008-2015. Eligible criteria included those receiving surgery, adjuvant radiotherapy and temozolomide, and without other cancer diagnoses. Survival between patients taking concurrent AED for 14 days or more during chemo-radiotherapy (AED group) and those who did not (non-AED group) were compared, and subgroup analyses for those with valproic acid (VPA), levetiracetam (LEV), or phenytoin were performed. Multivariate analyses were used to adjust for confounding factors. RESULTS There were 642 patients in the AED group, whereas 415 in the non-AED group. The demographic data was balanced except trend of more patients in the AED group had previous drug history of AEDs (22.6% vs. 18%, P 0.078). Overall, the AED group had significantly increased risk of mortality (HR = 1.18, P 0.016) compared to the non-AED group. Besides, an adverse dose-dependent relationship on survival was also demonstrated in the AED group (HR = 1.118, P 0.0003). In subgroup analyses, the significant detrimental effect was demonstrated in VPA group (HR = 1.29,P 0.0002), but not in LEV (HR = 1.18, P 0.079) and phenytoin (HR = 0.98, P 0.862). CONCLUSIONS Improved survival was not observed in patients with concurrent AEDs during chemo-radiotherapy. Our real-world data did not support prophylactic use of AEDs for glioblastoma patients.
Collapse
Affiliation(s)
- Peng-Yi Lee
- Department of Radiation Oncology, Show-Chwan Memorial Hospital, No. 542, Section 1 CHUNG-SHAN Road, Changhua, Taiwan
- Department of Radiation Oncology, Lin Shin Hospital, No. 36, Section 3 Huizhong Road, Taichung, Taiwan
- Department of Radiation Oncology, China Medical University Beigang Hospital, Yunlin, Taiwan
- Department of Radiation Oncology, China Medical University Hospital, China Medical University, Taichung, Taiwan
| | - Yu-Ting Wei
- Division of Family Medicine, Department of Community Medicine, China Medical University Hospital, Taichung, Taiwan
- Division of Occupational Medicine, Department of Community Medicine, China Medical University Hospital, Taichung, Taiwan
| | | | - Chin-Nan Chu
- Department of Radiation Oncology, China Medical University Hospital, China Medical University, Taichung, Taiwan
- Graduate Institute of Medical Science, China Medical University, Taichung, Taiwan
| | - Wen-Hui Chung
- Department of Radiation Oncology, China Medical University Hospital, China Medical University, Taichung, Taiwan
| | - Ti-Hao Wang
- Department of Radiation Oncology, China Medical University Hospital, China Medical University, Taichung, Taiwan
| |
Collapse
|
12
|
Nabian N, Ghalehtaki R, Zeinalizadeh M, Balaña C, Jablonska PA. State of the neoadjuvant therapy for glioblastoma multiforme-Where do we stand? Neurooncol Adv 2024; 6:vdae028. [PMID: 38560349 PMCID: PMC10981465 DOI: 10.1093/noajnl/vdae028] [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: 04/04/2024] Open
Abstract
Glioblastoma multiforme (GBM) is the most common malignant primary brain tumor in adults. Despite several investigations in this field, maximal safe resection followed by chemoradiotherapy and adjuvant temozolomide with or without tumor-treating fields remains the standard of care with poor survival outcomes. Many endeavors have failed to make a dramatic change in the outcomes of GBM patients. This study aimed to review the available strategies for newly diagnosed GBM in the neoadjuvant setting, which have been mainly neglected in contrast to other solid tumors.
Collapse
Affiliation(s)
- Naeim Nabian
- Radiation Oncology Research Center, Cancer Research Institute, Tehran University of Medical Sciences, Tehran, Iran
- Department of Radiation Oncology, Cancer Institute, Imam Khomeini Hospital Complex, Tehran University of Medical Sciences, Tehran, Iran
| | - Reza Ghalehtaki
- Radiation Oncology Research Center, Cancer Research Institute, Tehran University of Medical Sciences, Tehran, Iran
- Department of Radiation Oncology, Cancer Institute, Imam Khomeini Hospital Complex, Tehran University of Medical Sciences, Tehran, Iran
| | - Mehdi Zeinalizadeh
- Department of Neurosurgery, Tehran University of Medical Sciences, Tehran, Iran
| | - Carmen Balaña
- B.ARGO (Badalona Applied Research Group of Oncology) Medical Oncology Department, Catalan Institute of Oncology Badalona, Badalona, Spain
| | | |
Collapse
|
13
|
Mistry AM. Perioperative dexamethasone in high-grade gliomas: the short-term benefits and long-term harms. Front Oncol 2023; 13:1335730. [PMID: 38162484 PMCID: PMC10755919 DOI: 10.3389/fonc.2023.1335730] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2023] [Accepted: 12/04/2023] [Indexed: 01/03/2024] Open
Abstract
Dexamethasone has been commonly given to patients with a presumed new GBM in relatively large doses (6-16 mg daily for 1-2 weeks) since the 1960s without any rigorous evidence. This treatment with dexamethasone before the diagnosis and adjuvant therapy makes GBM patients unique compared to other newly diagnosed cancer patients. While dexamethasone may be beneficial, recent studies suggest that this potent immunosuppressant with pleiotropic effects is harmful in the long term. This perspective article summarizes the disadvantages of perioperative dexamethasone from multiple facets. It concludes that these growing data mandate rigorously testing the benefits of using perioperative dexamethasone.
Collapse
Affiliation(s)
- Akshitkumar M. Mistry
- Department of Neurological Surgery, University of Louisville, Louisville, KY, United States
| |
Collapse
|
14
|
Abstract
The care of patients with both high-grade glioma and low-grade glioma necessitates an interdisciplinary collaboration between neurosurgeons, neuro-oncologists, neurologists and other practitioners. In this review, we aim to detail the considerations, approaches and advances in the neurosurgical care of gliomas. We describe the impact of extent-of-resection in high-grade and low-grade glioma, with particular focus on primary and recurrent glioblastoma. We address advances in surgical methods and adjunct technologies such as intraoperative imaging and fluorescence guided surgery that maximize extent-of-resection while minimizing the potential for iatrogenic neurological deficits. Finally, we review surgically-mediated therapies other than resection and discuss the role of neurosurgery in emerging paradigm-shifts in inter-disciplinary glioma management such as serial tissue sampling and "window of opportunity trials".
Collapse
Affiliation(s)
- Andrew A Hardigan
- Department of Neurosurgery, Duke University Medical Center, Durham, North Carolina
| | - Joshua D Jackson
- Department of Neurosurgery, Duke University Medical Center, Durham, North Carolina
| | - Anoop P Patel
- Department of Neurosurgery, Duke University Medical Center, Durham, North Carolina
| |
Collapse
|
15
|
Crompton D, Koffler D, Fekrmandi F, Lehrer EJ, Sheehan JP, Trifiletti DM. Preoperative stereotactic radiosurgery as neoadjuvant therapy for resectable brain tumors. J Neurooncol 2023; 165:21-28. [PMID: 37889441 DOI: 10.1007/s11060-023-04466-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2023] [Accepted: 09/25/2023] [Indexed: 10/28/2023]
Abstract
PURPOSE Stereotactic radiosurgery (SRS) is a method of delivering conformal radiation, which allows minimal radiation damage to surrounding healthy tissues. Adjuvant radiation therapy has been shown to improve local control in a variety of intracranial neoplasms, such as brain metastases, gliomas, and benign tumors (i.e., meningioma, vestibular schwannoma, etc.). For brain metastases, adjuvant SRS specifically has demonstrated positive oncologic outcomes as well as preserving cognitive function when compared to conventional whole brain radiation therapy. However, as compared with neoadjuvant SRS, larger post-operative volumes and greater target volume uncertainty may come with an increased risk of local failure and treatment-related complications, such as radiation necrosis. In addition to its role in brain metastases, neoadjuvant SRS for high grade gliomas may enable dose escalation and increase immunogenic effects and serve a purpose in benign tumors for which one cannot achieve a gross total resection (GTR). Finally, although neoadjuvant SRS has historically been delivered with photon therapy, there are high LET radiation modalities such as carbon-ion therapy which may allow radiation damage to tissue and should be further studied if done in the neoadjuvant setting. In this review we discuss the evolving role of neoadjuvant radiosurgery in the treatment for brain metastases, gliomas, and benign etiologies. We also offer perspective on the evolving role of high LET radiation such as carbon-ion therapy. METHODS PubMed was systemically reviewed using the search terms "neoadjuvant radiosurgery", "brain metastasis", and "glioma". ' Clinicaltrials.gov ' was also reviewed to include ongoing phase III trials. RESULTS This comprehensive review describes the evolving role for neoadjuvant SRS in the treatment for brain metastases, gliomas, and benign etiologies. We also discuss the potential role for high LET radiation in this setting such as carbon-ion radiotherapy. CONCLUSION Early clinical data is very promising for neoadjuvant SRS in the setting of brain metastases. There are three ongoing phase III trials that will be more definitive in evaluating the potential benefits. While there is less data available for neoadjuvant SRS for gliomas, there remains a potential role, particularly to enable dose escalation and increase immunogenic effects.
Collapse
Affiliation(s)
- David Crompton
- Department of Radiation Oncology, Mayo Clinic, 4500 San Pablo Road South, Jacksonville, FL, 32224, USA
| | - Daniel Koffler
- Department of Radiation Oncology, Mayo Clinic, 4500 San Pablo Road South, Jacksonville, FL, 32224, USA
| | - Fatemeh Fekrmandi
- Department of Radiation Oncology, Roswell Park Comprehensive Cancer Center, Buffalo, USA
| | - Eric J Lehrer
- Department of Radiation Oncology, Mayo Clinic, Rochester, USA
| | - Jason P Sheehan
- Department of Neurological Surgery, University of Virginia, Charlottesville, USA
| | - Daniel M Trifiletti
- Department of Radiation Oncology, Mayo Clinic, 4500 San Pablo Road South, Jacksonville, FL, 32224, USA.
| |
Collapse
|
16
|
Nelson TA, Dietrich J. Investigational treatment strategies in glioblastoma: progress made and barriers to success. Expert Opin Investig Drugs 2023; 32:921-930. [PMID: 37796104 PMCID: PMC10764117 DOI: 10.1080/13543784.2023.2267982] [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: 08/06/2023] [Accepted: 10/04/2023] [Indexed: 10/06/2023]
Abstract
INTRODUCTION Glioblastoma, isocitrate dehydrogenase wildtype (IDHwt), remains an incurable disease despite considerable research effort. The current standard of care since 2005 comprises maximal safe resection followed by radiation with concurrent and adjuvant temozolomide; more recently, the addition of tumor treating fields was approved in the newly diagnosed and recurrent disease settings. AREAS COVERED Searches of PubMed, Cochrane Library, and ClinicalTrials.gov provided a foundation for this review. We first describe early research including carmustine wafers, brachytherapy, anti-angiogenesis, and immune checkpoint inhibition for glioblastoma. Next, we discuss challenges precluding the translation of preclinical successes. This is followed by a description of promising treatments such as chimeric antigen receptor T-cell therapy as well as the recent qualified successes of cancer vaccinations. Non-immunotherapy trials are also highlighted, and ongoing or pending phase 2 and 3 clinical trials are codified in study tables. EXPERT OPINION Unfortunately, hundreds of trials, including of agents effective in systemic malignancy, have not drastically changed management of glioblastoma. This may reflect unique resistance mechanisms and highlights a need for multimodality treatments beyond surgery, radiation, and conventional chemotherapy. Novel techniques, such as those in the emerging field of cancer neuroscience, may help uncover tolerable and effective regimens for this lethal malignancy.
Collapse
Affiliation(s)
- Thomas A Nelson
- Pappas Center for Neuro-Oncology, Department of Neurology, Massachusetts General Hospital, Boston, MA USA
| | - Jorg Dietrich
- Pappas Center for Neuro-Oncology, Department of Neurology, Massachusetts General Hospital, Boston, MA USA
| |
Collapse
|
17
|
Niyazi M, Andratschke N, Bendszus M, Chalmers AJ, Erridge SC, Galldiks N, Lagerwaard FJ, Navarria P, Munck Af Rosenschöld P, Ricardi U, van den Bent MJ, Weller M, Belka C, Minniti G. ESTRO-EANO guideline on target delineation and radiotherapy details for glioblastoma. Radiother Oncol 2023; 184:109663. [PMID: 37059335 DOI: 10.1016/j.radonc.2023.109663] [Citation(s) in RCA: 77] [Impact Index Per Article: 38.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2023] [Revised: 03/23/2023] [Accepted: 03/29/2023] [Indexed: 04/16/2023]
Abstract
BACKGROUND AND PURPOSE Target delineation in glioblastoma is still a matter of extensive research and debate. This guideline aims to update the existing joint European consensus on delineation of the clinical target volume (CTV) in adult glioblastoma patients. MATERIAL AND METHODS The ESTRO Guidelines Committee identified 14 European experts in close interaction with the ESTRO clinical committee and EANO who discussed and analysed the body of evidence concerning contemporary glioblastoma target delineation, then took part in a two-step modified Delphi process to address open questions. RESULTS Several key issues were identified and are discussed including i) pre-treatment steps and immobilisation, ii) target delineation and the use of standard and novel imaging techniques, and iii) technical aspects of treatment including planning techniques and fractionation. Based on the EORTC recommendation focusing on the resection cavity and residual enhancing regions on T1-sequences with the addition of a reduced 15 mm margin, special situations are presented with corresponding potential adaptations depending on the specific clinical situation. CONCLUSIONS The EORTC consensus recommends a single clinical target volume definition based on postoperative contrast-enhanced T1 abnormalities, using isotropic margins without the need to cone down. A PTV margin based on the individual mask system and IGRT procedures available is advised; this should usually be no greater than 3 mm when using IGRT.
Collapse
Affiliation(s)
- Maximilian Niyazi
- Department of Radiation Oncology, University Hospital, LMU Munich, Munich, Germany; German Cancer Consortium (DKTK), partner site Munich, Munich, Germany; Bavarian Cancer Research Center (BZKF), Munich, Germany.
| | - Nicolaus Andratschke
- Department of Radiation Oncology, University Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Martin Bendszus
- Department of Neuroradiology, University Hospital Heidelberg, Heidelberg, Germany
| | | | - Sara C Erridge
- Edinburgh Centre for Neuro-Oncology, University of Edinburgh, Western General Hospital, Edinburgh EH4 1EU, UK
| | - Norbert Galldiks
- Department of Neurology, Faculty of Medicine, University Hospital Cologne, University of Cologne, Cologne, Germany; Institute of Neuroscience and Medicine (INM-3), Research Center Juelich, Juelich, Germany; Center for Integrated Oncology (CIO), Universities of Aachen, Bonn, Cologne, and Duesseldorf, Germany
| | - Frank J Lagerwaard
- Department of Radiation Oncology, Amsterdam UMC location Vrije Universiteit Amsterdam, the Netherlands
| | - Pierina Navarria
- Radiotherapy and Radiosurgery Department, IRCCS, Humanitas Research Hospital, Rozzano, MI, Italy
| | - Per Munck Af Rosenschöld
- Radiation Physics, Department of Hematology, Oncology and Radiation Physics, Skåne University Hospital, and Lund University, Lund, Sweden
| | | | | | - Michael Weller
- Department of Neurology, Clinical Neuroscience Center, University Hospital and University of Zurich, Zurich, Switzerland
| | - Claus Belka
- Department of Radiation Oncology, University Hospital, LMU Munich, Munich, Germany; German Cancer Consortium (DKTK), partner site Munich, Munich, Germany; Bavarian Cancer Research Center (BZKF), Munich, Germany
| | - Giuseppe Minniti
- Dept. of Medicine, Surgery and Neuroscience, University of Siena, Siena, Italy; IRCCS Istituto Neurologico Mediterraneo Neuromed, Pozzilli, Italy
| |
Collapse
|
18
|
Dean JA, Tanguturi SK, Cagney D, Shin KY, Youssef G, Aizer A, Rahman R, Hammoudeh L, Reardon D, Lee E, Dietrich J, Tamura K, Aoyagi M, Wickersham L, Wen PY, Catalano P, Haas-Kogan D, Alexander BM, Michor F. Phase I study of a novel glioblastoma radiation therapy schedule exploiting cell-state plasticity. Neuro Oncol 2023; 25:1100-1112. [PMID: 36402744 PMCID: PMC10237407 DOI: 10.1093/neuonc/noac253] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2024] Open
Abstract
BACKGROUND Glioblastomas comprise heterogeneous cell populations with dynamic, bidirectional plasticity between treatment-resistant stem-like and treatment-sensitive differentiated states, with treatment influencing this process. However, current treatment protocols do not account for this plasticity. Previously, we generated a mathematical model based on preclinical experiments to describe this process and optimize a radiation therapy fractionation schedule that substantially increased survival relative to standard fractionation in a murine glioblastoma model. METHODS We developed statistical models to predict the survival benefit of interventions to glioblastoma patients based on the corresponding survival benefit in the mouse model used in our preclinical study. We applied our mathematical model of glioblastoma radiation response to optimize a radiation therapy fractionation schedule for patients undergoing re-irradiation for glioblastoma and developed a first-in-human trial (NCT03557372) to assess the feasibility and safety of administering our schedule. RESULTS Our statistical modeling predicted that the hazard ratio when comparing our novel radiation schedule with a standard schedule would be 0.74. Our mathematical modeling suggested that a practical, near-optimal schedule for re-irradiation of recurrent glioblastoma patients was 3.96 Gy × 7 (1 fraction/day) followed by 1.0 Gy × 9 (3 fractions/day). Our optimized schedule was successfully administered to 14/14 (100%) patients. CONCLUSIONS A novel radiation therapy schedule based on mathematical modeling of cell-state plasticity is feasible and safe to administer to glioblastoma patients.
Collapse
Affiliation(s)
- Jamie A Dean
- Department of Data Science, Dana-Farber Cancer Institute, Boston, Massachusetts, USA
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, Massachusetts, USA
- Department of Biostatistics, Harvard T.H. Chan School of Public Health, Boston, Massachusetts, USA
- Department of Medical Physics and Biomedical Engineering, University College London, London, UK
- UCL Cancer Institute, University College London, London, UK
| | - Shyam K Tanguturi
- Department of Radiation Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts, USA
| | - Daniel Cagney
- Department of Radiation Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts, USA
| | - Kee-Young Shin
- Department of Data Science, Dana-Farber Cancer Institute, Boston, Massachusetts, USA
- Department of Biostatistics, Harvard T.H. Chan School of Public Health, Boston, Massachusetts, USA
| | - Gilbert Youssef
- Center for Neuro-Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts, USA
- Center for Neuro-Oncology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Ayal Aizer
- Department of Radiation Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts, USA
| | - Rifaquat Rahman
- Department of Radiation Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts, USA
| | - Lubna Hammoudeh
- Department of Radiation Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts, USA
| | - David Reardon
- Center for Neuro-Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts, USA
| | - Eudocia Lee
- Center for Neuro-Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts, USA
| | - Jorg Dietrich
- Center for Neuro-Oncology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Kaoru Tamura
- Department of Neurosurgery, Tokyo Medical and Dental University, Tokyo, Japan
| | - Masaru Aoyagi
- Department of Neurosurgery, Tokyo Medical and Dental University, Tokyo, Japan
| | - Lacey Wickersham
- Department of Radiation Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts, USA
| | - Patrick Y Wen
- Center for Neuro-Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts, USA
| | - Paul Catalano
- Department of Data Science, Dana-Farber Cancer Institute, Boston, Massachusetts, USA
- Department of Biostatistics, Harvard T.H. Chan School of Public Health, Boston, Massachusetts, USA
| | - Daphne Haas-Kogan
- Department of Radiation Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts, USA
| | - Brian M Alexander
- Department of Radiation Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts, USA
| | - Franziska Michor
- Department of Data Science, Dana-Farber Cancer Institute, Boston, Massachusetts, USA
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, Massachusetts, USA
- Department of Biostatistics, Harvard T.H. Chan School of Public Health, Boston, Massachusetts, USA
- The Broad Institute of MIT and Harvard, Cambridge, MA, USA
- The Center for Cancer Evolution, Dana-Farber Cancer Institute, Boston, Massachusetts, USA
- The Ludwig Center at Harvard, Boston, Massachusetts, USA
| |
Collapse
|
19
|
Rončević A, Koruga N, Soldo Koruga A, Rončević R, Rotim T, Šimundić T, Kretić D, Perić M, Turk T, Štimac D. Personalized Treatment of Glioblastoma: Current State and Future Perspective. Biomedicines 2023; 11:1579. [PMID: 37371674 DOI: 10.3390/biomedicines11061579] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2023] [Revised: 05/24/2023] [Accepted: 05/27/2023] [Indexed: 06/29/2023] Open
Abstract
Glioblastoma (GBM) is the most aggressive glial tumor of the central nervous system. Despite intense scientific efforts, patients diagnosed with GBM and treated with the current standard of care have a median survival of only 15 months. Patients are initially treated by a neurosurgeon with the goal of maximal safe resection of the tumor. Obtaining tissue samples during surgery is indispensable for the diagnosis of GBM. Technological improvements, such as navigation systems and intraoperative monitoring, significantly advanced the possibility of safe gross tumor resection. Usually within six weeks after the surgery, concomitant radiotherapy and chemotherapy with temozolomide are initiated. However, current radiotherapy regimens are based on population-level studies and could also be improved. Implementing artificial intelligence in radiotherapy planning might be used to individualize treatment plans. Furthermore, detailed genetic and molecular markers of the tumor could provide patient-tailored immunochemotherapy. In this article, we review current standard of care and possibilities of personalizing these treatments. Additionally, we discuss novel individualized therapeutic options with encouraging results. Due to inherent heterogeneity of GBM, applying patient-tailored treatment could significantly prolong survival of these patients.
Collapse
Affiliation(s)
- Alen Rončević
- Department of Neurosurgery, University Hospital Center Osijek, 31000 Osijek, Croatia
- Faculty of Medicine, Josip Juraj Strossmayer University of Osijek, 31000 Osijek, Croatia
| | - Nenad Koruga
- Department of Neurosurgery, University Hospital Center Osijek, 31000 Osijek, Croatia
- Faculty of Medicine, Josip Juraj Strossmayer University of Osijek, 31000 Osijek, Croatia
| | - Anamarija Soldo Koruga
- Faculty of Medicine, Josip Juraj Strossmayer University of Osijek, 31000 Osijek, Croatia
- Department of Neurology, University Hospital Center Osijek, 31000 Osijek, Croatia
| | - Robert Rončević
- Faculty of Medicine, Josip Juraj Strossmayer University of Osijek, 31000 Osijek, Croatia
- Department of Diagnostic and Interventional Radiology, University Hospital Center Osijek, 31000 Osijek, Croatia
| | - Tatjana Rotim
- Faculty of Medicine, Josip Juraj Strossmayer University of Osijek, 31000 Osijek, Croatia
- Department of Diagnostic and Interventional Radiology, University Hospital Center Osijek, 31000 Osijek, Croatia
| | - Tihana Šimundić
- Faculty of Medicine, Josip Juraj Strossmayer University of Osijek, 31000 Osijek, Croatia
- Department of Nephrology, University Hospital Center Osijek, 31000 Osijek, Croatia
| | - Domagoj Kretić
- Faculty of Medicine, Josip Juraj Strossmayer University of Osijek, 31000 Osijek, Croatia
- Department of Diagnostic and Interventional Radiology, University Hospital Center Osijek, 31000 Osijek, Croatia
| | - Marija Perić
- Faculty of Medicine, Josip Juraj Strossmayer University of Osijek, 31000 Osijek, Croatia
- Department of Cytology, University Hospital Center Osijek, 31000 Osijek, Croatia
| | - Tajana Turk
- Faculty of Medicine, Josip Juraj Strossmayer University of Osijek, 31000 Osijek, Croatia
- Department of Diagnostic and Interventional Radiology, University Hospital Center Osijek, 31000 Osijek, Croatia
| | - Damir Štimac
- Faculty of Medicine, Josip Juraj Strossmayer University of Osijek, 31000 Osijek, Croatia
- Department of Radiology, National Memorial Hospital Vukovar, 32000 Vukovar, Croatia
| |
Collapse
|
20
|
Sachdev S, Dmello C, Sonabend AM. Radiosensitization of IDH-Mutated Gliomas through ZMYND8 - a Pathway to Improved Outcomes. Clin Cancer Res 2023; 29:1648-1650. [PMID: 36826993 PMCID: PMC10159893 DOI: 10.1158/1078-0432.ccr-23-0018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2023] [Revised: 02/10/2023] [Accepted: 02/16/2023] [Indexed: 02/25/2023]
Abstract
Isocitrate dehydrogenase 1-mutant (IDH1m) gliomas are recalcitrant tumors for which radiotherapy remains a standard treatment. A recent study identified ZMYND8 as a key mediator of radioresistance for IDH1m gliomas, and pharmacologic targeting of this pathway may heighten radiotherapy-induced tumor response, providing a prospect of improved clinical outcomes. See related article by Carney et al., p. 1763.
Collapse
Affiliation(s)
- Sean Sachdev
- Department of Radiation Oncology, Northwestern Lou and Jean Malnati Brain Tumor Institute, Northwestern University Robert H. Lurie Comprehensive Cancer Center, 676 N. St Clair Street, Suite 1820, Chicago, IL, 60611, USA
- Northwestern Medicine Malnati Brain Tumor Institute of the Lurie Comprehensive Cancer Center, Feinberg School of Medicine, Northwestern University, Chicago IL
| | - Crismita Dmello
- Northwestern Medicine Malnati Brain Tumor Institute of the Lurie Comprehensive Cancer Center, Feinberg School of Medicine, Northwestern University, Chicago IL
- Department of Neurological Surgery, Feinberg School of Medicine, Northwestern University, Chicago IL
| | - Adam M. Sonabend
- Northwestern Medicine Malnati Brain Tumor Institute of the Lurie Comprehensive Cancer Center, Feinberg School of Medicine, Northwestern University, Chicago IL
- Department of Neurological Surgery, Feinberg School of Medicine, Northwestern University, Chicago IL
| |
Collapse
|
21
|
Johansen PM, Hansen PY, Mohamed AA, Girshfeld SJ, Feldmann M, Lucke-Wold B. Focused ultrasound for treatment of peripheral brain tumors. EXPLORATION OF DRUG SCIENCE 2023:107-125. [PMID: 37171968 PMCID: PMC10168685 DOI: 10.37349/eds.2023.00009] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 01/11/2023] [Accepted: 04/13/2023] [Indexed: 05/14/2023]
Abstract
Malignant brain tumors are the leading cause of cancer-related death in children and remain a significant cause of morbidity and mortality throughout all demographics. Central nervous system (CNS) tumors are classically treated with surgical resection and radiotherapy in addition to adjuvant chemotherapy. However, the therapeutic efficacy of chemotherapeutic agents is limited due to the blood-brain barrier (BBB). Magnetic resonance guided focused ultrasound (MRgFUS) is a new and promising intervention for CNS tumors, which has shown success in preclinical trials. High-intensity focused ultrasound (HIFU) has the capacity to serve as a direct therapeutic agent in the form of thermoablation and mechanical destruction of the tumor. Low-intensity focused ultrasound (LIFU) has been shown to disrupt the BBB and enhance the uptake of therapeutic agents in the brain and CNS. The authors present a review of MRgFUS in the treatment of CNS tumors. This treatment method has shown promising results in preclinical trials including minimal adverse effects, increased infiltration of the therapeutic agents into the CNS, decreased tumor progression, and improved survival rates.
Collapse
Affiliation(s)
| | - Payton Yerke Hansen
- Charles E. Schmidt College of Medicine, Florida Atlantic University, Boca Raton, FL 33431, USA
| | - Ali A. Mohamed
- Charles E. Schmidt College of Medicine, Florida Atlantic University, Boca Raton, FL 33431, USA
| | - Sarah J. Girshfeld
- Charles E. Schmidt College of Medicine, Florida Atlantic University, Boca Raton, FL 33431, USA
| | - Marc Feldmann
- College of Medicine, University of Florida, Gainesville, FL 32611, USA
| | - Brandon Lucke-Wold
- Department of Neurosurgery, University of Florida, Gainesville, FL 32611, USA
| |
Collapse
|
22
|
Watowich MB, Gilbert MR, Larion M. T cell exhaustion in malignant gliomas. Trends Cancer 2023; 9:270-292. [PMID: 36681605 PMCID: PMC10038906 DOI: 10.1016/j.trecan.2022.12.008] [Citation(s) in RCA: 47] [Impact Index Per Article: 23.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2022] [Revised: 12/20/2022] [Accepted: 12/22/2022] [Indexed: 01/21/2023]
Abstract
Despite advances in understanding tumor biology, malignant gliomas remain incurable. While immunotherapy has improved outcomes in other cancer types, comparable efficacy has not yet been demonstrated for primary cancers of the central nervous system (CNS). T cell exhaustion, defined as a progressive decrease in effector function, sustained expression of inhibitory receptors, metabolic dysfunction, and distinct epigenetic and transcriptional alterations, contributes to the failure of immunotherapy in the CNS. Herein, we describe recent advances in understanding the drivers of T cell exhaustion in the glioma microenvironment. We discuss the extrinsic and intrinsic factors that contribute to exhaustion and highlight potential avenues for reversing this phenotype. Our ability to directly target specific immunosuppressive drivers in brain cancers would be a major advance in immunotherapy.
Collapse
Affiliation(s)
- Matthew B Watowich
- Neuro-Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Mark R Gilbert
- Neuro-Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Mioara Larion
- Neuro-Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA.
| |
Collapse
|
23
|
Tsien CI, Pugh SL, Dicker AP, Raizer JJ, Matuszak MM, Lallana EC, Huang J, Algan O, Deb N, Portelance L, Villano JL, Hamm JT, Oh KS, Ali AN, Kim MM, Lindhorst SM, Mehta MP. NRG Oncology/RTOG1205: A Randomized Phase II Trial of Concurrent Bevacizumab and Reirradiation Versus Bevacizumab Alone as Treatment for Recurrent Glioblastoma. J Clin Oncol 2023; 41:1285-1295. [PMID: 36260832 PMCID: PMC9940937 DOI: 10.1200/jco.22.00164] [Citation(s) in RCA: 80] [Impact Index Per Article: 40.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2022] [Revised: 06/07/2022] [Accepted: 08/16/2022] [Indexed: 11/20/2022] Open
Abstract
PURPOSE To assess whether reirradiation (re-RT) and concurrent bevacizumab (BEV) improve overall survival (OS) and/or progression-free survival (PFS), compared with BEV alone in recurrent glioblastoma (GBM). The primary objective was OS, and secondary objectives included PFS, response rate, and treatment adverse events (AEs) including delayed CNS toxicities. METHODS NRG Oncology/RTOG1205 is a prospective, phase II, randomized trial of re-RT and BEV versus BEV alone. Stratification factors included age, resection, and Karnofsky performance status (KPS). Patients with recurrent GBM with imaging evidence of tumor progression ≥ 6 months from completion of prior chemo-RT were eligible. Patients were randomly assigned 1:1 to re-RT, 35 Gy in 10 fractions, with concurrent BEV IV 10 mg/kg once in every 2 weeks or BEV alone until progression. RESULTS From December 2012 to April 2016, 182 patients were randomly assigned, of whom 170 were eligible. Patient characteristics were well balanced between arms. The median follow-up for censored patients was 12.8 months. There was no improvement in OS for BEV + RT, hazard ratio, 0.98; 80% CI, 0.79 to 1.23; P = .46; the median survival time was 10.1 versus 9.7 months for BEV + RT versus BEV alone. The median PFS for BEV + RT was 7.1 versus 3.8 months for BEV, hazard ratio, 0.73; 95% CI, 0.53 to 1.0; P = .05. The 6-month PFS rate improved from 29.1% (95% CI, 19.1 to 39.1) for BEV to 54.3% (95% CI, 43.5 to 65.1) for BEV + RT, P = .001. Treatment was well tolerated. There were a 5% rate of acute grade 3+ treatment-related AEs and no delayed high-grade AEs. Most patients died of recurrent GBM. CONCLUSION To our knowledge, NRG Oncology/RTOG1205 is the first prospective, randomized multi-institutional study to evaluate the safety and efficacy of re-RT in recurrent GBM using modern RT techniques. Overall, re-RT was shown to be safe and well tolerated. BEV + RT demonstrated a clinically meaningful improvement in PFS, specifically the 6-month PFS rate but no difference in OS.
Collapse
Affiliation(s)
| | - Stephanie L. Pugh
- NRG Oncology Statistics and Data Management Center, Philadelphia, PA
| | | | | | | | | | - Jiayi Huang
- Washington University School of Medicine in St Louis-Siteman Cancer Center, St. Louis, MO
| | - Ozer Algan
- University of Oklahoma Health Sciences Center, Oklahoma City, OK
| | - Nimisha Deb
- St Luke's University Hospital & Health Network accruals Thomas Jefferson University Hospital, Bethlehem, PA
| | - Lorraine Portelance
- University of Miami Miller School of Medicine-Sylvester Comprehensive Cancer Center, Miami, FL
| | | | - John T. Hamm
- Norton Hospital Pavilion and Medical Campus, Louisville, KY
| | - Kevin S. Oh
- Dana-Farber/Harvard Cancer Center, Boston, MA
| | - Arif N. Ali
- The Hope Center accruals Emory University/Winship Cancer Institute, Dalton, GA
| | - Michelle M. Kim
- University of Michigan Comprehensive Cancer Center, Ann Arbor, MI
| | - Scott M. Lindhorst
- Medical University of South Carolina Minority Underserved NCORP, Charleston, SC
| | | |
Collapse
|
24
|
Knisely JPS, Fine HA. Reirradiation for Recurrent Glioblastoma: What We Know and What We Do Not. J Clin Oncol 2023; 41:1183-1188. [PMID: 36260825 DOI: 10.1200/jco.22.01785] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
The Oncology Grand Rounds series is designed to place original reports published in the Journal into clinical context. A case presentation is followed by a description of diagnostic and management challenges, a review of the relevant literature, and a summary of the authors' suggested management approaches. The goal of this series is to help readers better understand how to apply the results of key studies, including those published in Journal of Clinical Oncology, to patients seen in their own clinical practice.
Collapse
Affiliation(s)
| | - Howard A Fine
- Department of Neurology, Weill Cornell Medicine, New York, NY.,Meyer Cancer Center, Weill Cornell Medicine, New York, NY
| |
Collapse
|
25
|
Adekeye AO, Needham D, Rahman R. Low-Density Lipoprotein Pathway Is a Ubiquitous Metabolic Vulnerability in High Grade Glioma Amenable for Nanotherapeutic Delivery. Pharmaceutics 2023; 15:pharmaceutics15020599. [PMID: 36839921 PMCID: PMC9958636 DOI: 10.3390/pharmaceutics15020599] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2023] [Revised: 02/06/2023] [Accepted: 02/09/2023] [Indexed: 02/12/2023] Open
Abstract
Metabolic reprogramming, through increased uptake of cholesterol in the form of low-density lipoproteins (LDL), is one way by which cancer cells, including high grade gliomas (HGG), maintain their rapid growth. In this study, we determined LDL receptor (LDLR) expression in HGGs using immunohistochemistry on tissue microarrays from intra- and inter tumour regions of 36 adult and 133 paediatric patients to confirm LDLR as a therapeutic target. Additionally, we analysed expression levels in three representative cell line models to confirm their future utility to test LDLR-targeted nanoparticle uptake, retention, and cytotoxicity. Our data show widespread LDLR expression in adult and paediatric cohorts, but with significant intra-tumour variation observed between the core and either rim or invasive regions of adult HGG. Expression was independent of paediatric tumour grade or identified clinicopathological factors. LDLR-expressing tumour cells localized preferentially within perivascular niches, also with significant adult intra-tumour variation. We demonstrated variable levels of LDLR expression in all cell lines, confirming their suitability as models to test LDLR-targeted nanotherapy delivery. Overall, our study reveals the LDLR pathway as a ubiquitous metabolic vulnerability in high grade gliomas across all ages, amenable to future consideration of LDL-mediated nanoparticle/drug delivery to potentially circumvent tumour heterogeneity.
Collapse
Affiliation(s)
- Adenike O. Adekeye
- Biodiscovery Institute, School of Medicine, University of Nottingham, Nottingham NG7 2RD, UK
| | - David Needham
- Department of Mechanical Engineering and Material Science, School of Engineering, Duke University, Durham, NC 27708, USA
| | - Ruman Rahman
- Biodiscovery Institute, School of Medicine, University of Nottingham, Nottingham NG7 2RD, UK
- Correspondence:
| |
Collapse
|
26
|
Shimizu S, Nakai K, Li Y, Mizumoto M, Kumada H, Ishikawa E, Yamamoto T, Matsumura A, Sakurai H. Boron Neutron Capture Therapy for Recurrent Glioblastoma Multiforme: Imaging Evaluation of a Case With Long-Term Local Control and Survival. Cureus 2023; 15:e33898. [PMID: 36819302 PMCID: PMC9937644 DOI: 10.7759/cureus.33898] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/17/2023] [Indexed: 01/20/2023] Open
Abstract
Glioblastoma (GBM) is difficult to cure with conventional multimodal treatment and has an extremely poor prognosis. Boron neutron capture therapy (BNCT) is a new particle therapy for malignant tumors in the brain and head and neck region. This radiotherapy utilizes a nuclear reaction between neutrons and a nonradioactive isotope, boron-10. In this method, a boron compound is administered transvenously into the body. The boron compound has the property of being selectively taken up only by the cells of malignant tumors, and the subsequent irradiation with neutrons can destroy malignant tumor cells without damaging normal cells. Since the irradiation dose to normal tissues is reduced in BNCT, it may be possible to re-irradiate malignant tumors that recur after radiotherapy. Clinical trials have reported prolonged survival and safety of BNCT in a small number of patients with refractory malignancies, including GBM, but these reports do not address quality of life or activities of daily living (ADL) after treatment, and there is no information on the assessment of local control by imaging. Here, we report a case of GBM that recurred after surgery, 60 Gy of conventional radiotherapy and standard treatment with temozolomide. The patient achieved long-term local control and survival over five years after BNCT and was able to maintain ADL at home without any specialist care. We describe the case with evaluation using longitudinal magnetic resonance imaging (MRI).
Collapse
Affiliation(s)
- Shosei Shimizu
- Proton Beam Therapy Center, University of Tsukuba Hospital, Tsukuba, JPN
| | - Kei Nakai
- Radiation Oncology, University of Tsukuba Hospital, Tsukuba, JPN
| | - Yinuo Li
- Proton Beam Therapy Center, University of Tsukuba Hospital, Tsukuba, JPN
| | - Masashi Mizumoto
- Radiation Oncology, University of Tsukuba Hospital, Tsukuba, JPN
| | - Hiroaki Kumada
- Proton Beam Therapy Center, University of Tsukuba Hospital, Tsukuba, JPN
| | | | - Tetsuya Yamamoto
- Radiation Oncology, Yokohama City University Hospital, Tsukuba, JPN
| | | | | |
Collapse
|
27
|
Wang LM, Englander ZK, Miller ML, Bruce JN. Malignant Glioma. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2023; 1405:1-30. [PMID: 37452933 DOI: 10.1007/978-3-031-23705-8_1] [Citation(s) in RCA: 34] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/18/2023]
Abstract
This chapter provides a comprehensive overview of malignant gliomas, the most common primary brain tumor in adults. These tumors are varied in their cellular origin, genetic profile, and morphology under the microscope, but together they share some of the most dismal prognoses of all neoplasms in the body. Although there is currently no cure for malignant glioma, persistent efforts to improve outcomes in patients with these tumors have led to modest increases in survival, and researchers worldwide continue to strive toward a deeper understanding of the factors that influence glioma development and response to treatment. In addition to well-established epidemiology, clinical manifestations, and common histopathologic and radiologic features of malignant gliomas, this section considers recent advances in molecular biology that have led to a more nuanced understanding of the genetic changes that characterize the different types of malignant glioma, as well as their implications for treatment. Beyond the traditional classification of malignant gliomas based on histopathological features, this chapter incorporates the World Health Organization's 2016 criteria for the classification of brain tumors, with special focus on disease-defining genetic alterations and newly established subcategories of malignant glioma that were previously unidentifiable based on microscopic examination alone. Traditional therapeutic modalities that form the cornerstone of treatment for malignant glioma, such as aggressive surgical resection followed by adjuvant chemotherapy and radiation therapy, and the studies that support their efficacy are reviewed in detail. This provides a foundation for additional discussion of novel therapeutic methods such as immunotherapy and convection-enhanced delivery, as well as new techniques for enhancing extent of resection such as fluorescence-guided surgery.
Collapse
Affiliation(s)
- Linda M Wang
- Columbia University Irving Medical Center, New York, NY, 10032, USA
| | | | - Michael L Miller
- Columbia University Irving Medical Center, New York, NY, 10032, USA
| | - Jeffrey N Bruce
- Department of Neurosurgery, Columbia University Irving Medical Center, New York, NY, 10032, USA.
| |
Collapse
|
28
|
New therapeutic strategies based on molecularly targeted therapy in glioblastoma – a case report and review of the literature. CURRENT ISSUES IN PHARMACY AND MEDICAL SCIENCES 2022. [DOI: 10.2478/cipms-2022-0036] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023] Open
Abstract
Abstract
Glioblastomas are the most common and most lethal forms of malignant primary brain tumor. We present a case report of a patient with III-grade glioma who achieved stable disease (SD) and clinical improvement after trametinib administration. We also report a review of the literature to Current Treatment Guidelines of Glioblastoma and new therapeutic strategies based on molecularly targeted therapy. Traditional treatments, including surgery, radiotherapy, and chemotherapy, have many limitations concerning the prognosis of patients with glioblastomas. Unfortunately, these tumors’recur after primary resection in the majority of cases. There is no standard therapy for recurrence of GBM. Targeted therapy offers a promising new treatment strategy. Regardless of those outstanding results much more can be done in the field of therapeutic options. Most urgent concerns include potent combining molecular targeted therapy with other types of treatments, selecting a group of patients for whom they turn out to be the most beneficial, and addressing adverse events of these molecules.
Collapse
|
29
|
Yang F, Dinakaran D, Heikal AA, Yaghoobpour Tari S, Ghosh S, Amanie J, Murtha A, Rowe LS, Roa WH, Patel S. Dosimetric predictors of toxicity in a randomized study of short-course vs conventional radiotherapy for glioblastoma. Radiother Oncol 2022; 177:152-157. [PMID: 36273738 DOI: 10.1016/j.radonc.2022.10.016] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2022] [Revised: 09/12/2022] [Accepted: 10/14/2022] [Indexed: 11/13/2022]
Abstract
PURPOSE There is no consensus on appropriate organ at risk (OAR) constraints for short-course radiotherapy for patients with glioblastoma. Using dosimetry and prospectively-collected toxicity data from a trial of short-course radiotherapy for glioblastoma, this study aims to empirically examine the OAR constraints, with particular attention to left hippocampus dosimetry and impact on neuro-cognitive decline. METHODS AND MATERIALS Data was taken from a randomized control trial of 133 adults (age 18-70 years; ECOG performance score 0-2) with newly diagnosed glioblastoma treated with 60 Gy in 30 (conventional arm) versus 20 (short-course arm) fractions of adjuvant chemoradiotherapy (ClinicalTrials.gov Identifier: NCT02206230). The delivered plan's dosimetry to the OARs was correlated to prospective-collected toxicity and Mini-Mental State Examination (MMSE) data. RESULTS Toxicity events were not significantly increased in the short-course arm versus the conventional arm. Across all OARs, delivered radiation doses within protocol-allowable maximum doses correlated with lack of grade ≥ 2 toxicities in both arms (p < 0.001), while patients with OAR doses at or above protocol limits correlated with increased grade ≥ 2 toxicities across all examined OARs in both arms (p-values 0.063-0.250). Mean left hippocampus dose was significantly associated with post-radiotherapy decline in MMSE scores (p = 0.005), while the right hippocampus mean dose did not reach statistical significance (p = 0.277). Compared to the original clinical plan, RapidPlan left hippocampus sparing model decreased left hippocampus mean dose by 43 % (p < 0.001), without compromising planning target volume coverage. CONCLUSIONS In this trial, protocol OAR constraints were appropriate for limiting grade ≥ 2 toxicities in conventional and short-course adjuvant chemoradiotherapy for glioblastoma. Higher left hippocampal mean doses were predictive for neuro-cognitive decline post-radiotherapy. Routine contouring and use of dose constraints to limit hippocampal dose is recommended to minimize neuro-cognitive decline in patients with glioblastoma treated with chemoradiotherapy.
Collapse
Affiliation(s)
- Fan Yang
- Division of Radiation Oncology, University of Alberta, Cross Cancer Institute, Edmonton, Alberta, Canada
| | - Deepak Dinakaran
- Division of Radiation Oncology, University of Alberta, Cross Cancer Institute, Edmonton, Alberta, Canada
| | - Amr A Heikal
- Division of Medical Physics, University of Alberta, Cross Cancer Institute, Edmonton, Alberta, Canada
| | - Shima Yaghoobpour Tari
- Division of Medical Physics, University of Alberta, Cross Cancer Institute, Edmonton, Alberta, Canada
| | - Sunita Ghosh
- Division of Medical Oncology, University of Alberta, Cross Cancer Institute, Edmonton, Alberta, Canada
| | - John Amanie
- Division of Radiation Oncology, University of Alberta, Cross Cancer Institute, Edmonton, Alberta, Canada
| | - Albert Murtha
- Division of Radiation Oncology, University of Alberta, Cross Cancer Institute, Edmonton, Alberta, Canada
| | - Lindsay S Rowe
- Division of Radiation Oncology, University of Alberta, Cross Cancer Institute, Edmonton, Alberta, Canada
| | - Wilson H Roa
- Division of Radiation Oncology, University of Alberta, Cross Cancer Institute, Edmonton, Alberta, Canada
| | - Samir Patel
- Division of Radiation Oncology, University of Alberta, Cross Cancer Institute, Edmonton, Alberta, Canada.
| |
Collapse
|
30
|
Prognostic Factors for Survival of Patients with Glioblastoma in the Southern Region of Morocco. ARCHIVES OF NEUROSCIENCE 2022. [DOI: 10.5812/ans-132014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Background: Glioblastoma is the most common brain cancer in adults. It is caused by the abnormal proliferation of central nervous system cells called astrocytes, with an incidence rate of 4.32 per 100,000 in the United States. The median survival for glioblastoma is about 1 to 2 years. In Morocco, the survival of patients with glioblastoma is relatively little explored. Objectives: This research aims to study overall survival and these prognostic factors in patients with glioblastoma. Methods: This is a retrospective study; the data were extracted from the files of patients with glioblastoma in the public reference oncology center in the southern region of Morocco; it is a prognostic study including all patients with glioblastoma cancer between 2014 and October 2021. Results: The present study ultimately focused on 71 files of cases diagnosed with glioblastoma. The median age at diagnosis was 57, with a sex ratio of 1.44. The median survival time for all glioblastoma patients in this study was 11 months (95% CI: 9.96 to 12.03 months). Univariate analysis revealed that age, sex, geographical origin, type of treatment, and type of surgery were significant at P = 0.20 and then included in the multivariate model. After adjusting for all factors, the results revealed that only gender, age, and geographical origin were statistically significant predictors of overall survival. Conclusions: The survival rate in patients with glioblastoma is improved with surgery, followed by concomitant radio-chemotherapy. We also confirmed that age and sex are important prognostic factors for the survival of patients with glioblastoma. Moreover, the data suggest the effect of the geographical origin of the patients on the overall survival of the patients as the only modifiable prognostic factor.
Collapse
|
31
|
Reissig F, Mamat C. Strained Ammonium Precursors for Radiofluorinations. ChemistryOpen 2022; 11:e202200039. [PMID: 35736542 PMCID: PMC9220932 DOI: 10.1002/open.202200039] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2022] [Revised: 05/24/2022] [Indexed: 12/11/2022] Open
Abstract
The increasing application of positron emission tomography (PET) in nuclear medicine has stimulated the extensive development of a multitude of novel and versatile techniques to introduce fluorine-18, especially for the radiolabelling of biologically or pharmacologically active molecules. Taking into consideration that the introduction of fluorine-18 (t1/2 =109.8 min) mostly proceeds under harsh conditions, radiolabelling of such molecules represents a challenge and is of enormous interest. Ideally, it should proceed in a regioselective manner under mild physiological conditions, in an acceptable time span, with high yields and high specific activities. Special attention has been drawn to 2-fluoroethyl and 3-fluoropropyl groups, which are often the active sites of radiofluorinated compounds. Precursors containing an ammonium leaving group - such as a strained azetidinium or aziridinium moiety - can help to overcome these obstacles leading to a convenient and mild introduction of [18 F]fluoride with high radiochemical yields.
Collapse
Affiliation(s)
- Falco Reissig
- Institut für Radiopharmazeutische Krebsforschung Helmholtz-Zentrum Dresden-RossendorfBautzner Landstraße 40001328DresdenGermany
- Fakultät Chemie und LebensmittelchemieTechnische Universität Dresden01062DresdenGermany
| | - Constantin Mamat
- Institut für Radiopharmazeutische Krebsforschung Helmholtz-Zentrum Dresden-RossendorfBautzner Landstraße 40001328DresdenGermany
- Fakultät Chemie und LebensmittelchemieTechnische Universität Dresden01062DresdenGermany
| |
Collapse
|
32
|
Ali MY, Oliva CR, Flor S, Goswami PC, Griguer CE. Cytochrome c oxidase mediates labile iron level and radioresistance in glioblastoma. Free Radic Biol Med 2022; 185:25-35. [PMID: 35476930 DOI: 10.1016/j.freeradbiomed.2022.04.012] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/06/2022] [Revised: 04/11/2022] [Accepted: 04/20/2022] [Indexed: 02/06/2023]
Abstract
Radiotherapy is an important treatment modality for glioblastoma (GBM), yet the initial effectiveness of radiotherapy is eventually lost due to the development of adaptive radioresistance during fractionated radiation therapy. Defining the molecular mechanism(s) responsible for the adaptive radioresistance in GBM is necessary for the development of effective treatment options. The cellular labile iron pool (LIP) is very important for determining the cellular response to radiation, as it contributes to radiation-induced production of reactive oxygen species (ROS) such as lipid radicals through Fenton reactions. Recently, cytochrome c oxidase (CcO), a mitochondrial heme-containing enzyme also involved in regulating ROS production, was found to be involved in GBM chemoresistance. However, the role of LIP and CcO in GBM radioresistance is not known. Herein, we tested the hypothesis that CcO-mediated alterations in the level of labile iron contribute to adaptive radioresistance. Using an in vitro model of GBM adaptive radioresistance, we found an increase in CcO activity in radioresistant cells that associated with a decrease in the cellular LIP, decrease in lipid peroxidation, and a switch in the CcO subunit 4 (COX4) isoform expressed, from COX4-2 to COX4-1. Furthermore, knockdown of COX4-1 in radioresistant GBM cells decreased CcO activity and restored radiosensitivity, whereas overexpression of COX4-1 in radiosensitive cells increased CcO activity and rendered the cells radioresistant. Overexpression of COX4-1 in radiosensitive cells also significantly reduced the cellular LIP and lipid peroxidation. Pharmacological manipulation of the cellular labile iron level using iron chelators altered CcO activity and the radiation response. Overall, these results demonstrate a mechanistic link between CcO activity and LIP in GBM radioresistance and identify the CcO subunit isoform switch from COX4-2 to COX4-1 as a novel biochemical node for adaptive radioresistance of GBM. Manipulation of CcO and the LIP may restore the sensitivity to radiation in radioresistant GBM cells and thereby provide a strategy to improve therapeutic outcome in patients with GBM.
Collapse
Affiliation(s)
- Md Yousuf Ali
- Free Radical & Radiation Biology Program, Department of Radiation Oncology, The University of Iowa, Iowa City, IA, 52242, USA; Interdisciplinary Graduate Program in Human Toxicology, The University of Iowa, Iowa City, IA, 52242, USA
| | - Claudia R Oliva
- Free Radical & Radiation Biology Program, Department of Radiation Oncology, The University of Iowa, Iowa City, IA, 52242, USA
| | - Susanne Flor
- Free Radical & Radiation Biology Program, Department of Radiation Oncology, The University of Iowa, Iowa City, IA, 52242, USA
| | - Prabhat C Goswami
- Free Radical & Radiation Biology Program, Department of Radiation Oncology, The University of Iowa, Iowa City, IA, 52242, USA
| | - Corinne E Griguer
- Free Radical & Radiation Biology Program, Department of Radiation Oncology, The University of Iowa, Iowa City, IA, 52242, USA.
| |
Collapse
|
33
|
Lao Y, Ruan D, Vassantachart A, Fan Z, Ye JC, Chang EL, Chin R, Kaprealian T, Zada G, Shiroishi MS, Sheng K, Yang W. Voxelwise Prediction of Recurrent High-Grade Glioma via Proximity Estimation-Coupled Multidimensional Support Vector Machine. Int J Radiat Oncol Biol Phys 2022; 112:1279-1287. [PMID: 34963559 PMCID: PMC8923952 DOI: 10.1016/j.ijrobp.2021.12.153] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2021] [Revised: 12/09/2021] [Accepted: 12/16/2021] [Indexed: 01/28/2023]
Abstract
PURPOSE To provide early and localized glioblastoma (GBM) recurrence prediction, we introduce a novel postsurgery multiparametric magnetic resonance-based support vector machine (SVM) method coupling with stem cell niche (SCN) proximity estimation. METHODS AND MATERIALS This study used postsurgery magnetic resonance imaging (MRI) scans from 50 patients with recurrent GBM, obtained approximately 2 months before clinically diagnosed recurrence. The main prediction pipeline consisted of a proximity-based estimator to identify regions with high risk of recurrence (HRRs) and an SVM classifier to provide voxelwise prediction in HRRs. The HRRs were estimated using the weighted sum of inverse distances to 2 possible origins of recurrence-the SCN and the tumor cavity. Subsequently, multiparametric voxels (from T1, T1 contrast-enhanced, fluid-attenuated inversion recovery, T2, and apparent diffusion coefficient) within the HRR were grouped into recurrent (warped from the clinical diagnosis) and nonrecurrent subregions and fed into the proximity estimation-coupled SVM classifier (SVMPE). The cohort was randomly divided into 40% and 60% for training and testing, respectively. The trained SVMPE was then extrapolated to an earlier time point for earlier recurrence prediction. As an exploratory analysis, the SVMPE predictive cluster sizes and the image intensities from the 5 magnetic resonance sequences were compared across time to assess the progressive subclinical traces. RESULTS On 2-month prerecurrence MRI scans from 30 test cohort patients, the SVMPE classifier achieved a recall of 0.80, a precision of 0.69, an F1-score of 0.73, and a mean boundary distance of 7.49 mm. Exploratory analysis at early time points showed spatially consistent but significantly smaller subclinical clusters and significantly increased T1 contrast-enhanced and apparent diffusion coefficient values over time. CONCLUSIONS We demonstrated a novel voxelwise early prediction method, SVMPE, for GBM recurrence based on clinical follow-up MR scans. The SVMPE is promising in localizing subclinical traces of recurrence 2 months ahead of clinical diagnosis and may be used to guide more effective personalized early salvage therapy.
Collapse
Affiliation(s)
- Yi Lao
- Department of Radiation Oncology, University of California - Los Angeles, USA
| | - Dan Ruan
- Department of Radiation Oncology, University of California - Los Angeles, USA
| | - April Vassantachart
- Department of Radiation Oncology, Keck School of Medicine of USC, Los Angeles, USA
| | - Zhaoyang Fan
- Department of Radiology, Keck School of Medicine of USC, Los Angeles, USA
| | - Jason C. Ye
- Department of Radiation Oncology, Keck School of Medicine of USC, Los Angeles, USA
| | - Eric L. Chang
- Department of Radiation Oncology, Keck School of Medicine of USC, Los Angeles, USA
| | - Robert Chin
- Department of Radiation Oncology, University of California - Los Angeles, USA
| | - Tania Kaprealian
- Department of Radiation Oncology, University of California - Los Angeles, USA
| | - Gabriel Zada
- Department of Neurosurgery, Keck School of Medicine of USC, Los Angeles, USA
| | - Mark S Shiroishi
- Department of Radiology, Keck School of Medicine of USC, Los Angeles, USA
| | - Ke Sheng
- Department of Radiation Oncology, University of California - Los Angeles, USA
| | - Wensha Yang
- Department of Radiation Oncology, Keck School of Medicine of USC, Los Angeles, USA
| |
Collapse
|
34
|
Sun M, Huang N, Tao Y, Wen R, Zhao G, Zhang X, Xie Z, Cheng Y, Mao J, Liu G. The efficacy of temozolomide combined with levetiracetam for glioblastoma (GBM) after surgery: a study protocol for a double-blinded and randomized controlled trial. Trials 2022; 23:234. [PMID: 35346332 PMCID: PMC8962067 DOI: 10.1186/s13063-022-06168-1] [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: 11/02/2021] [Accepted: 03/10/2022] [Indexed: 11/10/2022] Open
Abstract
Background Temozolomide is applied as the standard chemotherapy agent in patients with glioblastoma (GBM) after surgery. However, the benefit of this treatment for patients is limited by the invasive growth of gliomas and drug resistance. There are indications from fundamental experimental and retrospective studies that levetiracetam has the potential to improve the survival rate of patients with GBM. However, it has yet to be determined whether the combination of temozolomide and levetiracetam is more effective than standard temozolomide chemotherapy. Therefore, we designed a randomized clinical trial to investigate the therapeutic effect of the new combined regime for treating GBM. Methods/design This is a double-blind and randomized clinical trial conducted in a single center. One hundred forty-two patients will be recruited and screened for the inclusion and exclusion criteria. Then, eligible participants will be randomly assigned to an experimental group or a control group in a 1:1 ratio. Based on the administration of radiation therapy (RT), participants in the experimental group will be prescribed levetiracetam plus temozolomide chemotherapy for 34 weeks while participants in the control group will receive placebo tablets plus temozolomide for the same duration. A 3-year follow-up will be conducted on all patients after intervention. Accordingly, the primary outcome will be progression-free survival (PFS). The secondary endpoints include overall survival (OS), the Karnofsky Performance Status (KPS), the objective response rate (ORR), and adverse event incidence. Discussion It is expected that the results of this trial will provide high-level evidence regarding the clinical benefits of levetiracetam and temozolomide combined in the treatment of GBM. Trial registration Chinese Clinical Trial Registry, ChiCTR2100049941. Registered on 14 August 2021 Supplementary Information The online version contains supplementary material available at 10.1186/s13063-022-06168-1.
Collapse
Affiliation(s)
- Maoyuan Sun
- Department of Neurosurgery, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, 400000, China
| | - Ning Huang
- Department of Neurosurgery, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, 400000, China
| | - Yihao Tao
- Department of Neurosurgery, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, 400000, China
| | - Rong Wen
- Department of Neurosurgery, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, 400000, China
| | - Guanjian Zhao
- Department of Neurosurgery, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, 400000, China
| | - Xiang Zhang
- Department of Neurosurgery, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, 400000, China
| | - Zongyi Xie
- Department of Neurosurgery, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, 400000, China
| | - Yuan Cheng
- Department of Neurosurgery, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, 400000, China
| | - Jinning Mao
- Department of Cardiology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, 400000, China
| | - Guodong Liu
- Department of Neurosurgery, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, 400000, China. .,Experimental Therapeutics and Molecular Imaging Laboratory, Department of Neurology, Neuro-Oncology Division, Massachusetts General Hospital, Boston, MA, 02114, USA. .,Neuroscience Program, Harvard Medical School, Boston, MA, 02114, USA.
| |
Collapse
|
35
|
Abdelsayed N, Bondranko Z, Montesano P. Schwannomatosis Presenting With a Grade IV Glioblastoma: A Case Report and Literature Review. Cureus 2022; 14:e23381. [PMID: 35475061 PMCID: PMC9022045 DOI: 10.7759/cureus.23381] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/21/2022] [Indexed: 11/17/2022] Open
Abstract
Schwannomatosis is a rare subset of neurofibromatosis. It is a disease process with a predisposition to schwannomas in the absence of bilateral vestibular schwannomas, which differentiates it from neurofibromatosis 2 (NF2). It is occasionally associated with certain tumors such as malignant peripheral nerve sheath tumors or rhabdoid tumors. Currently, there is limited literature to suggest an association between schwannomatosis and glioblastoma (GB). We present a case of a 55-year-old female with a history significant for schwannomatosis who presented after a witnessed first-time seizure with left facial weakness and slurred speech. She was found to have a 3 cm right-sided ring-enhancing lesion that was excised and found to be a grade IV Isocitrate dehydrogenase (IDH) wildtype GB.
Collapse
|
36
|
Robin AM, Pawloski JA, Snyder JM, Walbert T, Rogers L, Mikkelsen T, Noushmehr H, Lee I, Rock J, Kalkanis SN, Rosenblum ML. Neurosurgery's Impact on Neuro-Oncology—“Can We Do Better?”—Lessons Learned Over 50 Years. Neurosurgery 2022; 68:17-26. [DOI: 10.1227/neu.0000000000001879] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2022] [Accepted: 01/08/2022] [Indexed: 11/19/2022] Open
|
37
|
Li R, Wang H, Liang Q, Chen L, Ren J. Radiotherapy for glioblastoma: clinical issues and nanotechnology strategies. Biomater Sci 2022; 10:892-908. [PMID: 34989724 DOI: 10.1039/d1bm01401c] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Glioblastoma multiforme (GBM) is the most common primary brain cancer in adults with poor prognosis. Despite the current state of knowledge on its genetic characteristics, relatively little progress has been made in improving the treatment of patients with this fatal disease. Radiotherapy (RT) has been identified as a crucial treatment for GBM following surgical resection to improve both local control and survival. Unfortunately, radiotherapy resistance is frequently observed in GBM patients, which is the major reason for the high mortality rate of cancer patients. Radioresistance of GBM is often multifactorial and heterogeneous, and associated with the recurrence of GBM after surgery. Nanotechnology has gained increasing attention and has already been investigated for optimization of radiosensitization due to the unique properties of nanobiomaterials, such as photoelectric decay characteristics or potential as carriers for drug delivery to the central nervous system. A large body of preclinical data has accumulated over the past several years, in which nanotechnology-based strategies exhibit promising potential to enhance the radiosensitivity of GBM, both in cellular and animal models. In this review, we summarize the mechanisms of GBM radioresistance, including tumor cell-intrinsic factors as well as tumor microenvironment (TME). We further discuss current nano-biotechnology-based radiosensitizer in the treatment of GBM, summarize the latest findings, highlight challenges, and put forward prospects for the future of nano-radiosensitizers. These data suggest that nanotechnology has the potential to address many of the clinical challenges and nanobiomaterials would become promising next-generation radiotherapy sensitizers for GBM treatment.
Collapse
Affiliation(s)
- Ruiqi Li
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science & Technology, Wuhan 430022, P. R. China.
| | - Haihong Wang
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science & Technology, Wuhan 430022, P. R. China.
| | - Qing Liang
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science & Technology, Wuhan 430022, P. R. China.
| | - Lian Chen
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science & Technology, Wuhan 430022, P. R. China.
| | - Jinghua Ren
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science & Technology, Wuhan 430022, P. R. China.
| |
Collapse
|
38
|
Ebrahimi Zade A, Shahabi Haghighi S, Soltani M. Deep Neural Networks for Neuro-oncology: Towards Patient Individualized Design of Chemo-Radiation Therapy for Glioblastoma Patients. J Biomed Inform 2022; 127:104006. [DOI: 10.1016/j.jbi.2022.104006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2021] [Revised: 01/04/2022] [Accepted: 01/25/2022] [Indexed: 11/28/2022]
|
39
|
Gutierrez-Quintana R, Walker DJ, Williams KJ, Forster DM, Chalmers AJ. Radiation-induced neuroinflammation: a potential protective role for poly(ADP-ribose) polymerase inhibitors? Neurooncol Adv 2022; 4:vdab190. [PMID: 35118383 PMCID: PMC8807076 DOI: 10.1093/noajnl/vdab190] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Radiotherapy (RT) plays a fundamental role in the treatment of glioblastoma (GBM). GBM are notoriously invasive and harbor a subpopulation of cells with stem-like features which exhibit upregulation of the DNA damage response (DDR) and are radioresistant. High radiation doses are therefore delivered to large brain volumes and are known to extend survival but also cause delayed toxicity with 50%-90% of patients developing neurocognitive dysfunction. Emerging evidence identifies neuroinflammation as a critical mediator of the adverse effects of RT on cognitive function. In addition to its well-established role in promoting repair of radiation-induced DNA damage, activation of poly(ADP-ribose) polymerase (PARP) can exacerbate neuroinflammation by promoting secretion of inflammatory mediators. Therefore, PARP represents an intriguing mechanistic link between radiation-induced activation of the DDR and subsequent neuroinflammation. PARP inhibitors (PARPi) have emerged as promising new agents for GBM when given in combination with RT, with multiple preclinical studies demonstrating radiosensitizing effects and at least 3 compounds being evaluated in clinical trials. We propose that concomitant use of PARPi could reduce radiation-induced neuroinflammation and reduce the severity of radiation-induced cognitive dysfunction while at the same time improving tumor control by enhancing radiosensitivity.
Collapse
Affiliation(s)
- Rodrigo Gutierrez-Quintana
- Institute of Cancer Sciences, College of Medical Veterinary and Life Sciences, University of Glasgow, Glasgow, UK
| | - David J Walker
- Institute of Cancer Sciences, College of Medical Veterinary and Life Sciences, University of Glasgow, Glasgow, UK
| | - Kaye J Williams
- Division of Pharmacy and Optometry, School of Health Sciences, Manchester Cancer Research Centre, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, UK
| | - Duncan M Forster
- Division of Informatics, Imaging and Data Sciences, Manchester Molecular Imaging Centre, The University of Manchester, Manchester, UK
| | - Anthony J Chalmers
- Institute of Cancer Sciences, College of Medical Veterinary and Life Sciences, University of Glasgow, Glasgow, UK
| |
Collapse
|
40
|
Budnick HC, Richardson AM, Shiue K, Watson G, Ng SK, Le Y, Shah MV. GammaTile for Gliomas: A Single-Center Case Series. Cureus 2021; 13:e19390. [PMID: 34925992 PMCID: PMC8654117 DOI: 10.7759/cureus.19390] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/08/2021] [Indexed: 12/31/2022] Open
Abstract
GammaTile® (GT Medical Technologies, Tempe, Arizona) is a surgically targeted radiation source, approved by FDA for brachytherapy in primary and secondary brain neoplasms. Each GammaTile is composed of a collagen sponge with four seeds of cesium 131 and is particularly useful in recurrent tumors. We report our early experience in seven patients with recurrent gliomas to assess this type of brachytherapy with particular attention to ease of use, complication, and surgical planning. This study represents a retrospective chart review of surgical use and early clinical outcomes of GammaTile in recurrent gliomas. The number of tiles was planned using pre-operative imaging and dosimetry was planned based on post-operative imaging. Patients were followed during their hospital stay and were followed up after discharge. Parameters such as case length, resection extent, complication, ICU length of stay (LOS), hospital LOS, pre-operative Glasgow Coma Scale (GCS), immediate post-operative GCS, post-operative imaging findings, recurrence at follow-up, length of follow-up, and dosimetry were collected in a retrospective manner. Seven patients were identified that met the inclusion criteria. Two patients were diagnosed with recurrent glioblastoma multiforme (GBM), one lower-grade glioma that recurred as a GBM, one GBM that recurred as a gliosarcoma, and two recurrent oligodendrogliomas. We found that operation time, ICU LOS, hospital LOS, pre- and post-operative GCS, and post-operative complications were within the expected ranges for tumor resection patients. Further, dosimetry data suggests that six out of seven patients received adequate radiation coverage, with the seventh having implantation limitations due to nearby organs at risk. We report no postoperative complications that can be attributed to the GammaTiles themselves. In our cohort, we report seven cases where GammaTiles were implanted in recurrent gliomas. No implant-related post-operative complications were identified. This early data suggests that GammaTile can be a safe form of brachytherapy in recurrent gliomas.
Collapse
Affiliation(s)
- Hailey C Budnick
- Neurological Surgery, Indiana University Health, Indianapolis, USA
| | | | - Kevin Shiue
- Radiation Oncology, Indiana University (IU) Health Simon Cancer Center, Indianapolis, USA
| | - Gordon Watson
- Radiation Oncology, Indiana University (IU) Health Simon Cancer Center, Indianapolis, USA
| | - Sook K Ng
- Radiation Oncology, Indiana University (IU) Health Simon Cancer Center, Indianapolis, USA
| | - Yi Le
- Radiation Oncology, Indiana University (IU) Health Simon Cancer Center, Indianapolis, USA
| | - Mitesh V Shah
- Neurological Surgery, Indiana University Health, Indianapolis, USA
| |
Collapse
|
41
|
Wang Z, Xie F, Wu Y, Wang L, Bai Y, Long J, Wang X. Differential genes and scoring criteria among immunogenomic clusters of lower-grade gliomas. Int Immunopharmacol 2021; 101:108376. [PMID: 34815191 DOI: 10.1016/j.intimp.2021.108376] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2021] [Revised: 10/28/2021] [Accepted: 11/11/2021] [Indexed: 01/06/2023]
Abstract
High incidence of recurrency had been a significant threat among glioma patients. Moreover, the performance of traditional therapies among recurrent gliomas was far from satisfying. Advances in the tumor microenvironment (TME) and immune responses on the brain inspired immunotherapy researches. Nevertheless, verification of classic PD-1/PD-L1 inhibitors failed in phase III clinical trials. Additional gene targets were required for future studies among glioma patients. Immune cell infiltration (ICI) scores, defined based on multiple prognostic genes, were proved as the marker for the sensitivity of immunotherapies in many tumors. However, relevant results were not reported in gliomas. In the study, a retrospective cohort of 495 patients was classified into two ICI score subgroups. High ICI scores were closely related to high tumor mutation burden (TMB) values, indicating a high instability of genes. Furthermore, ICI scores were proved as reliable prognostic predictors. And a predictive model was built based on the ICI scores and multiple clinical features. The model showed its superiority through both internal validation and external validation. The ICI scores and the predictive model showed significant clinical values through decision curve analysis (DCA) since high ICI scores were related to high sensitivity for treatment. The prognostic immune-related gene list provided targets for immunotherapy researches.
Collapse
Affiliation(s)
- Zhile Wang
- Department of Medical Oncology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Fucun Xie
- Department of Medical Oncology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Yijun Wu
- Department of Medical Oncology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Li Wang
- Department of Medical Oncology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Yi Bai
- Department of Hepatobiliary Surgery, Tianjin First Central Hospital, Tianjin, China
| | - Junyu Long
- Department of Liver Surgery, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, China
| | - Xiang Wang
- Department of Medical Oncology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China.
| |
Collapse
|
42
|
Tsuji Y, Nonoguchi N, Okuzaki D, Wada Y, Motooka D, Hirota Y, Toho T, Yoshikawa N, Furuse M, Kawabata S, Miyatake SI, Nakamura H, Yamamoto R, Nakamura S, Kuroiwa T, Wanibuchi M. Chronic pathophysiological changes in the normal brain parenchyma caused by radiotherapy accelerate glioma progression. Sci Rep 2021; 11:22110. [PMID: 34764346 PMCID: PMC8585920 DOI: 10.1038/s41598-021-01475-0] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2021] [Accepted: 10/28/2021] [Indexed: 12/14/2022] Open
Abstract
Radiation therapy is one of standard treatment for malignant glioma after surgery. The microenvironment after irradiation is considered not to be suitable for the survival of tumor cells (tumor bed effect). This study investigated whether the effect of changes in the microenvironment of parenchymal brain tissue caused by radiotherapy affect the recurrence and progression of glioma. 65-Gy irradiation had been applied to the right hemisphere of Fisher rats. After 3 months from irradiation, we extracted RNA and protein from the irradiated rat brain. To study effects of proteins extracted from the brains, we performed WST-8 assay and tube formation assay in vitro. Cytokine production were investigated for qPCR. Additionally, we transplanted glioma cell into the irradiated and sham animals and the median survival time of F98 transplanted rats was also examined in vivo. Immunohistochemical analyses and invasiveness of implanted tumor were evaluated. X-ray irradiation promoted the secretion of cytokines such as CXCL12, VEGF-A, TGF-β1 and TNFα from the irradiated brain. Proteins extracted from the irradiated brain promoted the proliferation and angiogenic activity of F98 glioma cells. Glioma cells implanted in the irradiated brains showed significantly high proliferation, angiogenesis and invasive ability, and the post-irradiation F98 tumor-implanted rats showed a shorter median survival time compared to the Sham-irradiation group. The current study suggests that the microenvironment around the brain tissue in the chronic phase after exposure to X-ray radiation becomes suitable for glioma cell growth and invasion.
Collapse
Affiliation(s)
- Yuichiro Tsuji
- Department of Neurosurgery, Osaka Medical and Pharmaceutical University, 2-7 Daigaku-Machi, Takatsuki, Osaka, 569-8686, Japan.
| | - Naosuke Nonoguchi
- Department of Neurosurgery, Osaka Medical and Pharmaceutical University, 2-7 Daigaku-Machi, Takatsuki, Osaka, 569-8686, Japan.
| | - Daisuke Okuzaki
- grid.136593.b0000 0004 0373 3971Genome Information Research Center, Research Institute for Microbial Diseases, Osaka University, 3-1 Yamadaoka, Suita, Osaka 565-0871 Japan
| | - Yusuke Wada
- grid.261455.10000 0001 0676 0594Department of Advanced Pathobiology, Graduate School of Life and Environmental Sciences, Osaka Prefecture University, 1-58 Rinku Ourai-Kita, Izumisano, Osaka 598-8531 Japan
| | - Daisuke Motooka
- grid.136593.b0000 0004 0373 3971Genome Information Research Center, Research Institute for Microbial Diseases, Osaka University, 3-1 Yamadaoka, Suita, Osaka 565-0871 Japan
| | - Yuki Hirota
- Department of Neurosurgery, Osaka Medical and Pharmaceutical University, 2-7 Daigaku-Machi, Takatsuki, Osaka 569-8686 Japan
| | - Taichiro Toho
- Department of Neurosurgery, Osaka Medical and Pharmaceutical University, 2-7 Daigaku-Machi, Takatsuki, Osaka 569-8686 Japan
| | - Nobuhiko Yoshikawa
- Department of Radiology, Osaka Medical and Pharmaceutical University, Osaka, Japan
| | - Motomasa Furuse
- Department of Neurosurgery, Osaka Medical and Pharmaceutical University, 2-7 Daigaku-Machi, Takatsuki, Osaka 569-8686 Japan
| | - Shinji Kawabata
- Department of Neurosurgery, Osaka Medical and Pharmaceutical University, 2-7 Daigaku-Machi, Takatsuki, Osaka 569-8686 Japan
| | - Shin-Ichi Miyatake
- Department of Neurosurgery, Osaka Medical and Pharmaceutical University, 2-7 Daigaku-Machi, Takatsuki, Osaka 569-8686 Japan ,Division for Advanced Medical Development, Cancer Center, Osaka Medical and Pharmaceutical University, 2-7 Daigaku-Machi, Takatsuki, Osaka 569-8686 Japan
| | - Hiroyuki Nakamura
- grid.32197.3e0000 0001 2179 2105Laboratory for Chemistry and Life Science, Institute of Innovative Research, Tokyo Institute of Technology, 4259 Nagatsuta-cho, Midori-ku, Yokohama, Kanagawa 226-8503 Japan
| | - Ryohei Yamamoto
- grid.261455.10000 0001 0676 0594Department of Advanced Pathobiology, Graduate School of Life and Environmental Sciences, Osaka Prefecture University, 1-58 Rinku Ourai-Kita, Izumisano, Osaka 598-8531 Japan
| | - Shota Nakamura
- grid.136593.b0000 0004 0373 3971Genome Information Research Center, Research Institute for Microbial Diseases, Osaka University, 3-1 Yamadaoka, Suita, Osaka 565-0871 Japan
| | - Toshihiko Kuroiwa
- Department of Neurosurgery, Tesseikai Neurosurgical Hospital, 28-1, Nakanohommachi, Shijyonawate, Osaka 575-8511 Japan
| | - Masahiko Wanibuchi
- Department of Neurosurgery, Osaka Medical and Pharmaceutical University, 2-7 Daigaku-Machi, Takatsuki, Osaka 569-8686 Japan
| |
Collapse
|
43
|
Rahman R, Sulman E, Haas-Kogan D, Cagney DN. Update on Radiation Therapy for Central Nervous System Tumors. Hematol Oncol Clin North Am 2021; 36:77-93. [PMID: 34711456 DOI: 10.1016/j.hoc.2021.08.006] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Radiation therapy has long been a critical modality of treatment of patients with central nervous system tumors, including primary brain tumors, brain metastases, and meningiomas. Advances in radiation technology and delivery have allowed for more precise treatment to optimize patient outcomes and minimize toxicities. Improved understanding of the molecular underpinnings of brain tumors and normal brain tissue response to radiation will allow for continued refinement of radiation treatment approaches to improve clinical outcomes for brain tumor patients. With continued advances in precision and delivery, radiation therapy will continue to be an important modality to achieve optimal outcomes of brain tumor patients.
Collapse
Affiliation(s)
- Rifaquat Rahman
- Department of Radiation Oncology, Dana-Farber/Brigham and Women's Cancer Center, Harvard Medical School, 75 Francis Street, ASB1-L2, Boston, MA 02115, USA
| | - Erik Sulman
- Department of Radiation Oncology, New York University Grossman School of Medicine, 160 East 34th Street, New York, NY 10016, USA
| | - Daphne Haas-Kogan
- Department of Radiation Oncology, Dana-Farber/Brigham and Women's Cancer Center, Harvard Medical School, 75 Francis Street, ASB1-L2, Boston, MA 02115, USA
| | - Daniel N Cagney
- Department of Radiation Oncology, Dana-Farber/Brigham and Women's Cancer Center, Harvard Medical School, 75 Francis Street, ASB1-L2, Boston, MA 02115, USA.
| |
Collapse
|
44
|
Over-expression of lncRNA TMEM161B-AS1 promotes the malignant biological behavior of glioma cells and the resistance to temozolomide via up-regulating the expression of multiple ferroptosis-related genes by sponging hsa-miR-27a-3p. Cell Death Discov 2021; 7:311. [PMID: 34689169 PMCID: PMC8542043 DOI: 10.1038/s41420-021-00709-4] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2021] [Revised: 09/13/2021] [Accepted: 10/11/2021] [Indexed: 01/20/2023] Open
Abstract
A growing body of evidence suggests that long-chain non-coding RNA (lncRNA) plays an important role in the malignant biological behavior and drug resistance of glioblastoma (GBM) cells. In this study, we analyzed the role and potential mechanism of lncRNA TMEM161B-AS1 in the malignant biological behavior of GBM cells and temozolomide (TMZ) resistance. Studies have found that FANCD2 and CD44 are significantly related to the occurrence of GBM, TMZ resistance and the survival of GBM patients. Knockdown of TMEM161B-AS1 down-regulated the expression of FANCD2 and CD44 by sponging hsa-miR-27a-3p, inhibited the proliferation, migration, invasion and promoted apoptosis, ferroptosis of U87 cells and U251 cells. Down-regulation of lncRNA TMEM161B-AS1 and/or over-expression of hsa-miR-27a-3p down-regulated the expression of FANCD2 and CD44, and inhibited the tumor growth in nude mice. These results demonstrated that the lncRNA TMEM161B-AS1-hsa-miR-27a-3p-FANCD2/CD44 signal axis regulated the malignant biological behavior of GBM and TMZ resistance. These findings were expected to provide promising therapeutic targets for the treatment of glioma.
Collapse
|
45
|
Adegboyega G, Ozair A, Kanmounye US, Bandyopadhyay S, Vaqas B. Letter: Is the Stupp Protocol an Expensive and Unsustainable Standard of Care for Glioblastoma in Low- and Middle-Income Country Settings? A Call to Action! Neurosurgery 2021; 89:E249-E251. [PMID: 34318884 DOI: 10.1093/neuros/nyab273] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2021] [Accepted: 06/10/2021] [Indexed: 11/13/2022] Open
Affiliation(s)
- Gideon Adegboyega
- Barts and The London School of Medicine and Dentistry Queen Mary University of London London, UK
- Research Department Association of Future African Neurosurgeons Yaounde, Cameroon
| | - Ahmad Ozair
- Faculty of Medicine King George's Medical University Lucknow, India
| | | | - Soham Bandyopadhyay
- Oxford University Global Surgery Group Nuffield Department of Surgical Sciences University of Oxford Oxford, UK
| | - Babar Vaqas
- Department of Neurosurgery Queens Hospital Romford, UK
- Imperial College London London, UK
| |
Collapse
|
46
|
Ruiz-Garcia H, Ramirez-Loera C, Malouff TD, Seneviratne DS, Palmer JD, Trifiletti DM. Novel Strategies for Nanoparticle-Based Radiosensitization in Glioblastoma. Int J Mol Sci 2021; 22:9673. [PMID: 34575840 PMCID: PMC8465220 DOI: 10.3390/ijms22189673] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2021] [Revised: 09/01/2021] [Accepted: 09/03/2021] [Indexed: 01/09/2023] Open
Abstract
Radiotherapy (RT) is one of the cornerstones in the current treatment paradigm for glioblastoma (GBM). However, little has changed in the management of GBM since the establishment of the current protocol in 2005, and the prognosis remains grim. Radioresistance is one of the hallmarks for treatment failure, and different therapeutic strategies are aimed at overcoming it. Among these strategies, nanomedicine has advantages over conventional tumor therapeutics, including improvements in drug delivery and enhanced antitumor properties. Radiosensitizing strategies using nanoparticles (NP) are actively under study and hold promise to improve the treatment response. We aim to describe the basis of nanomedicine for GBM treatment, current evidence in radiosensitization efforts using nanoparticles, and novel strategies, such as preoperative radiation, that could be synergized with nanoradiosensitizers.
Collapse
Affiliation(s)
- Henry Ruiz-Garcia
- Department of Radiation Oncology, Mayo Clinic, Jacksonville, FL 32224, USA; (H.R.-G.); (T.D.M.); (D.S.S.)
- Department of Neurological Surgery, Mayo Clinic, Jacksonville, FL 32224, USA;
| | | | - Timothy D. Malouff
- Department of Radiation Oncology, Mayo Clinic, Jacksonville, FL 32224, USA; (H.R.-G.); (T.D.M.); (D.S.S.)
| | - Danushka S. Seneviratne
- Department of Radiation Oncology, Mayo Clinic, Jacksonville, FL 32224, USA; (H.R.-G.); (T.D.M.); (D.S.S.)
| | - Joshua D. Palmer
- Department of Radiation Oncology, Ohio State University, Columbus, OH 43210, USA;
| | - Daniel M. Trifiletti
- Department of Radiation Oncology, Mayo Clinic, Jacksonville, FL 32224, USA; (H.R.-G.); (T.D.M.); (D.S.S.)
- Department of Neurological Surgery, Mayo Clinic, Jacksonville, FL 32224, USA;
| |
Collapse
|
47
|
Scheer M, Strauss C, Scheller C, Kubelt C, Skalej M, Mawrin C, Prell J. Case report of recurrent anaplastic oligodendroglioma with mixed astrocytic components and pathological discordance of tumor progression. INTERDISCIPLINARY NEUROSURGERY 2021. [DOI: 10.1016/j.inat.2021.101195] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022] Open
|
48
|
Mazurek M, Rola R. The implications of nitric oxide metabolism in the treatment of glial tumors. Neurochem Int 2021; 150:105172. [PMID: 34461111 DOI: 10.1016/j.neuint.2021.105172] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2021] [Revised: 08/03/2021] [Accepted: 08/21/2021] [Indexed: 12/20/2022]
Abstract
Glial tumors are the most common intracranial malignancies. Unfortunately, despite such a high prevalence, patients' prognosis is usually poor. It is related to the high invasiveness, tendency to relapse and the resistance of tumors to traditional methods of treatment. An important link in the aspect of these issues may be nitric oxide (NO) metabolism. It is a very complex mechanism with multidirectional effects on the neoplastic process. Depending on the concentration axis, it can both exert pro-tumor action as well as contribute to the inhibition of tumorigenesis. The latest observations show that the control of its metabolism can be very helpful in the development of new methods of treating gliomas, as well as in increasing the effectiveness of the agents currently used. The influence of nitric oxide and nitric oxide synthase (NOS) activity on glioma stem cells seem to be of particular importance. The use of specific inhibitors may allow the reduction of tumor growth and its tendency to relapse. Another important feature of GSCs is their conditioning of glioma resistance to traditional forms of treatment. Recent studies have shown that modulation of NO metabolism can suppress this effect, preventing the induction of radio and chemoresistance. Moreover, nitric oxide is involved in the regulation of a number of immune mechanisms. Adequate modulation of its metabolism may contribute to the induction of an anti-tumor response in the patients' immune system.
Collapse
Affiliation(s)
- Marek Mazurek
- Chair and Department of Neurosurgery and Paediatric Neurosurgery, Medical University in Lublin, Poland.
| | - Radosław Rola
- Chair and Department of Neurosurgery and Paediatric Neurosurgery, Medical University in Lublin, Poland
| |
Collapse
|
49
|
Asemi Z, Behnam M, Pourattar MA, Mirzaei H, Razavi ZS, Tamtaji OR. Therapeutic Potential of Berberine in the Treatment of Glioma: Insights into Its Regulatory Mechanisms. Cell Mol Neurobiol 2021; 41:1195-1201. [PMID: 32557203 PMCID: PMC11448641 DOI: 10.1007/s10571-020-00903-5] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2019] [Accepted: 06/10/2020] [Indexed: 01/07/2023]
Abstract
Glioma is known as one of the most common primary intracranial tumors accounting for four-fifths of malignant brain tumors. There are several biological pathways that play a synergistic, pathophysiological role in glioma, including apoptosis, autophagy, oxidative stress, and cell cycle arrest. According to previous rese arches, the drugs used in the treatment of glioma have been associated with significant limitations. Therefore, improved and/or new therapeutic platforms are required. In this regard, multiple flavonoids and alkaloids have been extensively studied in the treatment of glioma. Berberine is a protoberberine alkaloid with wide range of pharmacological activities, applicable to various pathological conditions. Few studies have reported beneficial roles of berberine in glioma. Berberine exerts its pharmacological functions in glioma by controlling different molecular and cellular pathways. We reviewed the existing knowledge supporting the use of berberine in the treatment of glioma and its effects on molecular and cellular mechanisms.
Collapse
Affiliation(s)
- Zatollah Asemi
- Research Center for Biochemistry and Nutrition in Metabolic Diseases, Institute for Basic Sciences, Kashan University of Medical Sciences, Kashan, Islamic Republic of Iran
| | | | - Mohammad Ali Pourattar
- Department of Radiobiology, Iran University of Medical Sciences, Tehran, Islamic Republic of Iran
| | - Hamed Mirzaei
- Research Center for Biochemistry and Nutrition in Metabolic Diseases, Institute for Basic Sciences, Kashan University of Medical Sciences, Kashan, Islamic Republic of Iran
| | - Zahra Sadat Razavi
- Student Research Committee, Kashan University of Medical Sciences, Kashan, Islamic Republic of Iran
| | - Omid Reza Tamtaji
- Research Center for Biochemistry and Nutrition in Metabolic Diseases, Institute for Basic Sciences, Kashan University of Medical Sciences, Kashan, Islamic Republic of Iran.
| |
Collapse
|
50
|
Adaptor Protein Complex 1 Sigma 3 Is Highly Expressed in Glioma and Could Enhance Its Progression. COMPUTATIONAL AND MATHEMATICAL METHODS IN MEDICINE 2021; 2021:5086236. [PMID: 34367317 PMCID: PMC8346305 DOI: 10.1155/2021/5086236] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/06/2021] [Accepted: 07/11/2021] [Indexed: 11/17/2022]
Abstract
Introduction Glioma is the widely occurring deadly neoplasm induced by glial cell canceration in the central nervous system, including the brain and spinal cord. The function of AP1S3 is special in numerous diseases, but its exact role in glioma remains unknown. Methods Bioinformatics analysis was performed at the beginning. Based on TCGA database, differentially expressed genes were obtained. Protein-protein interaction (PPI) network analysis is performed by STRING. The annotation, visualization, and synthesis (DAVID) discovery database program was used for gene ontology enrichment analysis and Kyoto Encyclopedia of Genes and Genomes pathway analysis. The Kaplan-Meier curve was plotted to determine the prognostic value of AP1S3 Also, in vitro experiments were conducted in our research. Results 4370 differentially expressed genes were identified. 215 key genes were screened by protein-protein interaction (PPI) analysis; AP1S3 had a higher degree. The top five enriched pathways related to AP1S3 contain protein processing in the endoplasmic reticulum (ER), extracellular matrix receptor (ECM receptor) interaction, focal adhesion, advanced glycation end product (AGE) receptor for AGE (RAGE) signaling pathway in diabetic complications, and mRNA surveillance pathway. Additionally, the AP1S3 level was dramatically upregulated in glioblastoma (GBM) samples, but greatly reduced in low-grade glioma (LGG) samples when compared to that in normal tissues. The Kaplan-Meier curve data showed that AP1S3 was closely related to the disease-free survival (DFS) of glioma. Our data suggested that the expression of AP1S3 was increased in glioma in comparison with normal tissues, in line with the data of clinical samples. What was more, our data demonstrated that the reduction of AP1S3 in glioma cells could result in the inhibition of cell proliferation, invasion, and migration. Conclusion Collectively, our results implied that AP1S3 was a promising biomarker of glioma diagnosis and displayed as an oncogene in glioma.
Collapse
|