1
|
Pham J, Cote DJ, Kang K, Briggs RG, Gomez D, Prasad A, Daggupati S, Sisti J, Chow F, Attenello F, Chen CC, Zada G. Treatment practices and survival outcomes for IDH-wildtype glioblastoma patients according to MGMT promoter methylation status: insights from the U.S. National Cancer Database. J Neurooncol 2025; 172:655-665. [PMID: 39907975 PMCID: PMC11968476 DOI: 10.1007/s11060-025-04952-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2024] [Accepted: 01/23/2025] [Indexed: 02/06/2025]
Abstract
PURPOSE Methylation of the O6-methylguanine-DNA methyltransferase (MGMT) promoter is an important prognostic marker in glioblastoma (GBM); however, its implementation in clinical practice remains understudied. Here, we assessed the prevalence of MGMT methylation status among GBM patients in the United States. Additionally, we evaluated treatment practices and survival outcomes of GBM patients according to MGMT promoter methylation status. METHODS The National Cancer Database was queried to identify all adult U.S. patients (≥ 18 years) diagnosed with IDH-wildtype GBM between 2018 and 2020. Treatment regimen was grouped into no chemotherapy and no radiotherapy, chemotherapy alone (without radiotherapy), radiotherapy alone (without chemotherapy), and chemoradiotherapy (chemotherapy and radiotherapy). Survival data were analyzed using Kaplan-Meier survival curves, log-rank tests, and multivariable Cox proportional hazard modeling. RESULTS A total of 20,734 patients were included, of whom 6,404 (30.9%) had MGMT-methylated GBM, 9,065 (43.7%) had MGMT-unmethylated tumors, and 5,265 (25.4%) had unknown methylation status. The median and three-year overall survival were 12.4 months and 15.5%, respectively, for the entire cohort (16.4 months and 23.9% for MGMT-methylated patients and 11.8 months and 9.8% for MGMT-unmethylated patients, p < 0.001). Chemoradiotherapy was less commonly used for elderly (≥ 70 years, 58.5%) than non-elderly (< 70 years, 79.2%) patients. Among elderly patients, radiotherapy alone was more commonly administered than chemotherapy alone for patients with MGMT-unmethylated tumors (11.2% vs. 2.1%) and MGMT-methylated tumors (6.6% vs. 3.9%). However, chemotherapy alone was associated with a lower mortality risk (HR 0.71, 95% CI 0.51-0.99, p = 0.04) than radiotherapy alone for elderly patients with MGMT-methylated tumors, while chemotherapy alone was associated with a higher mortality risk (HR 1.63, 95% CI 1.09-2.44, p = 0.02) than radiotherapy alone for elderly patients with MGMT-unmethylated tumors. Patients who were elderly, uninsured, insured through Medicaid, lived in zip codes with lower median education levels, or received care at non-academic programs were less likely to undergo MGMT testing. CONCLUSION A high proportion of GBM patients in the United States undergo MGMT promoter testing, though significant sociodemographic disparities exist. While there was a decrease in chemoradiotherapy use with increasing age, radiotherapy alone was more commonly administered to elderly patients than chemotherapy alone irrespective of MGMT promoter methylation status.
Collapse
Affiliation(s)
- John Pham
- Department of Neurosurgery, The Warren Alpert Medical School of Brown University, Providence, RI, 02903, USA.
| | - David J Cote
- Department of Neurosurgery, University of Southern California Keck School of Medicine, Los Angeles, CA, 90033, USA
| | - Keiko Kang
- Department of Neurosurgery, University of Southern California Keck School of Medicine, Los Angeles, CA, 90033, USA
| | - Robert G Briggs
- Department of Neurosurgery, University of Southern California Keck School of Medicine, Los Angeles, CA, 90033, USA
| | - David Gomez
- Department of Neurosurgery, University of Southern California Keck School of Medicine, Los Angeles, CA, 90033, USA
| | - Apurva Prasad
- Department of Neurosurgery, University of Southern California Keck School of Medicine, Los Angeles, CA, 90033, USA
| | - Sindhu Daggupati
- Department of Neurosurgery, University of Southern California Keck School of Medicine, Los Angeles, CA, 90033, USA
| | - Jonathan Sisti
- Department of Neurosurgery, University of Southern California Keck School of Medicine, Los Angeles, CA, 90033, USA
| | - Frances Chow
- Department of Neurosurgery, University of Southern California Keck School of Medicine, Los Angeles, CA, 90033, USA
| | - Frank Attenello
- Department of Neurosurgery, University of Southern California Keck School of Medicine, Los Angeles, CA, 90033, USA
| | - Clark C Chen
- Department of Neurosurgery, The Warren Alpert Medical School of Brown University, Providence, RI, 02903, USA
| | - Gabriel Zada
- Department of Neurosurgery, University of Southern California Keck School of Medicine, Los Angeles, CA, 90033, USA
| |
Collapse
|
2
|
Cataldi S, Feraco P, Marrale M, Alongi P, Geraci L, La Grutta L, Caruso G, Bartolotta TV, Midiri M, Gagliardo C. Intra-tumoral susceptibility signals in brain gliomas: where do we stand? FRONTIERS IN RADIOLOGY 2025; 5:1546069. [PMID: 40052095 PMCID: PMC11882858 DOI: 10.3389/fradi.2025.1546069] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/16/2024] [Accepted: 01/28/2025] [Indexed: 03/09/2025]
Abstract
Nowadays, the genetic and biomolecular profile of neoplasms-related with their biological behaviour-have become a key issue in oncology, as they influence many aspects of both diagnosis and treatment. In the neuro-oncology field, neuroradiological research has recently explored the potential of non-invasively predicting the molecular phenotype of primary brain neoplasms, particularly gliomas, based on magnetic resonance imaging (MRI), using both conventional and advanced imaging techniques. Among these, diffusion-weighted imaging (DWI), perfusion-weighted imaging (PWI), MR spectroscopy (MRS) and susceptibility-weighted imaging (SWI) and have been used to explore various aspects of glioma biology, including predicting treatment response and understanding treatment-related changes during follow-up imaging. Recently, intratumoral susceptibility signals (ITSSs)-visible on SWI-have been recognised as an important new imaging tool in the evaluation of brain gliomas, as they offer a fast and simple non-invasive window into their microenvironment. These intratumoral hypointensities reflect critical pathological features such as microhemorrhages, calcifications, necrosis and vascularization. Therefore, ITSSs can provide neuroradiologists with more biological information for glioma differential diagnosis, grading and subtype differentiation, providing significant clinical support in prognosis assessment, therapeutic management and treatment response evaluation. This review summarizes recent advances in ITSS applications in glioma assessment, emphasizing both its potential and limitations while referencing key studies in the field.
Collapse
Affiliation(s)
- Simone Cataldi
- Department of Biomedicine, Neurosciences and Advanced Diagnostics (BIND), University of Palermo, Palermo, Italy
| | - Paola Feraco
- Centre for Medical Sciences (CISMed), University of Trento, Trento, Italy
| | - Maurizio Marrale
- Department of Physics and Chemistry “Emilio Segrè”, University of Palermo, Palermo, Italy
| | - Pierpaolo Alongi
- Department of Biomedicine, Neurosciences and Advanced Diagnostics (BIND), University of Palermo, Palermo, Italy
- Nuclear Medicine Unit, Department of Radiological Sciences, A.R.N.A.S. Civico, Palermo, Italy
| | - Laura Geraci
- Neuroradiology Unit, Department of Radiological Sciences, A.R.N.A.S. Civico, Palermo, Italy
| | - Ludovico La Grutta
- Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialties (ProMISE), University of Palermo, Palermo, Italy
| | - Giuseppe Caruso
- Department of Biomedicine, Neurosciences and Advanced Diagnostics (BIND), University of Palermo, Palermo, Italy
| | - Tommaso Vincenzo Bartolotta
- Department of Biomedicine, Neurosciences and Advanced Diagnostics (BIND), University of Palermo, Palermo, Italy
| | - Massimo Midiri
- Department of Biomedicine, Neurosciences and Advanced Diagnostics (BIND), University of Palermo, Palermo, Italy
| | - Cesare Gagliardo
- Department of Biomedicine, Neurosciences and Advanced Diagnostics (BIND), University of Palermo, Palermo, Italy
- Neuroradiology Unit, University-Hospital Paolo Giaccone, Palermo, Italy
| |
Collapse
|
3
|
Rafanan J, Ghani N, Kazemeini S, Nadeem-Tariq A, Shih R, Vida TA. Modernizing Neuro-Oncology: The Impact of Imaging, Liquid Biopsies, and AI on Diagnosis and Treatment. Int J Mol Sci 2025; 26:917. [PMID: 39940686 PMCID: PMC11817476 DOI: 10.3390/ijms26030917] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2024] [Revised: 01/18/2025] [Accepted: 01/20/2025] [Indexed: 02/16/2025] Open
Abstract
Advances in neuro-oncology have transformed the diagnosis and management of brain tumors, which are among the most challenging malignancies due to their high mortality rates and complex neurological effects. Despite advancements in surgery and chemoradiotherapy, the prognosis for glioblastoma multiforme (GBM) and brain metastases remains poor, underscoring the need for innovative diagnostic strategies. This review highlights recent advancements in imaging techniques, liquid biopsies, and artificial intelligence (AI) applications addressing current diagnostic challenges. Advanced imaging techniques, including diffusion tensor imaging (DTI) and magnetic resonance spectroscopy (MRS), improve the differentiation of tumor progression from treatment-related changes. Additionally, novel positron emission tomography (PET) radiotracers, such as 18F-fluoropivalate, 18F-fluoroethyltyrosine, and 18F-fluluciclovine, facilitate metabolic profiling of high-grade gliomas. Liquid biopsy, a minimally invasive technique, enables real-time monitoring of biomarkers such as circulating tumor DNA (ctDNA), extracellular vesicles (EVs), circulating tumor cells (CTCs), and tumor-educated platelets (TEPs), enhancing diagnostic precision. AI-driven algorithms, such as convolutional neural networks, integrate diagnostic tools to improve accuracy, reduce interobserver variability, and accelerate clinical decision-making. These innovations advance personalized neuro-oncological care, offering new opportunities to improve outcomes for patients with central nervous system tumors. We advocate for future research integrating these tools into clinical workflows, addressing accessibility challenges, and standardizing methodologies to ensure broad applicability in neuro-oncology.
Collapse
Affiliation(s)
| | | | | | | | | | - Thomas A. Vida
- Department of Medical Education, Kirk Kerkorian School of Medicine at UNLV, 625 Shadow Lane, Las Vegas, NV 89106, USA; (J.R.); (N.G.); (S.K.); (A.N.-T.); (R.S.)
| |
Collapse
|
4
|
Russo M, Martella N, Gargano D, Fantasma F, Marcovecchio C, Russo V, Oliva MA, Segatto M, Saviano G, Di Bartolomeo S, Arcella A. Lavender Essential Oil and Its Terpenic Components Negatively Affect Tumor Properties in a Cell Model of Glioblastoma. Molecules 2024; 29:6044. [PMID: 39770132 PMCID: PMC11676467 DOI: 10.3390/molecules29246044] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2024] [Revised: 12/18/2024] [Accepted: 12/19/2024] [Indexed: 01/11/2025] Open
Abstract
Glioblastoma (GBM) is the most common and aggressive form of brain cancer in adults, characterized by extensive growth, a high recurrence rate, and resistance to treatment. Growing research interest is focusing on the biological roles of natural compounds due to their potential beneficial effects on health. Our research aimed to investigate the effects of lavender essential oil (LEO) on a GBM cell model. Chemical characterization using GC-MS analysis indicated that LEO contains several terpenes, compounds that have been found to exhibit anticancer properties by interfering with key cancer-related pathways in several cancer models. By means of cell biology assays, we demonstrated that LEO impairs cell proliferation and migration, and also reduces oxidative stress in U87 cells. We further observed that Terpinen-4-ol, contained in LEO, was capable of reproducing the effects of the oil on GBM cells. Our results suggest that the terpenic molecules present in LEO could be considered valuable allies alongside conventional therapies against GBM.
Collapse
Affiliation(s)
- Miriam Russo
- Department of Biosciences and Territory, University of Molise, 86090 Pesche, Italy; (M.R.); (N.M.); (D.G.); (F.F.); (C.M.); (M.S.)
| | - Noemi Martella
- Department of Biosciences and Territory, University of Molise, 86090 Pesche, Italy; (M.R.); (N.M.); (D.G.); (F.F.); (C.M.); (M.S.)
| | - Deborah Gargano
- Department of Biosciences and Territory, University of Molise, 86090 Pesche, Italy; (M.R.); (N.M.); (D.G.); (F.F.); (C.M.); (M.S.)
| | - Francesca Fantasma
- Department of Biosciences and Territory, University of Molise, 86090 Pesche, Italy; (M.R.); (N.M.); (D.G.); (F.F.); (C.M.); (M.S.)
| | - Chiara Marcovecchio
- Department of Biosciences and Territory, University of Molise, 86090 Pesche, Italy; (M.R.); (N.M.); (D.G.); (F.F.); (C.M.); (M.S.)
| | - Veronica Russo
- IRCCS Istituto Neurologico Mediterraneo NEUROMED, Via Atinense 18, 86077 Pozzilli, Italy; (V.R.); (M.A.O.); (A.A.)
| | - Maria Antonietta Oliva
- IRCCS Istituto Neurologico Mediterraneo NEUROMED, Via Atinense 18, 86077 Pozzilli, Italy; (V.R.); (M.A.O.); (A.A.)
| | - Marco Segatto
- Department of Biosciences and Territory, University of Molise, 86090 Pesche, Italy; (M.R.); (N.M.); (D.G.); (F.F.); (C.M.); (M.S.)
| | - Gabriella Saviano
- Department of Biosciences and Territory, University of Molise, 86090 Pesche, Italy; (M.R.); (N.M.); (D.G.); (F.F.); (C.M.); (M.S.)
| | - Sabrina Di Bartolomeo
- Department of Biosciences and Territory, University of Molise, 86090 Pesche, Italy; (M.R.); (N.M.); (D.G.); (F.F.); (C.M.); (M.S.)
| | - Antonietta Arcella
- IRCCS Istituto Neurologico Mediterraneo NEUROMED, Via Atinense 18, 86077 Pozzilli, Italy; (V.R.); (M.A.O.); (A.A.)
| |
Collapse
|
5
|
Zhang Q, Dai Z, Chen Y, Li Q, Guo Y, Zhu Z, Tu M, Cai L, Lu X. Endosome associated trafficking regulator 1 promotes tumor growth and invasion of glioblastoma multiforme via inhibiting TNF signaling pathway. J Neurooncol 2024; 166:113-127. [PMID: 38191954 DOI: 10.1007/s11060-023-04527-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2023] [Accepted: 11/30/2023] [Indexed: 01/10/2024]
Abstract
PURPOSE Endosome associated trafficking regulator 1 (ENTR1) is a novel endosomal protein, which can affect multiple cellular biological behavior by remodeling plasma membrane structures. However, little is known regarding its function and underlying mechanisms in glioblastoma multiforme. METHODS Expression profile and clinical signature were obtained from The Public Database of human tumor. Immunohistochemical staining and western blotting assays were used to measure ENTR1 expression level. Human primary GBM tumor cells and human GBM cell lines A172, U87 and U251 were used to clarify the precise role of ENTR1. CCK-8 assays, wound healing and transwell invasion assays were designed to investigate cell viability, invasion and migration of GBM cells, respectively. Underlying molecular mechanisms of ENTR1 were determined via RNA-seq analysis. Tumor formation assay was used to validate the influence of ENTR1 in vivo. RESULTS Compared with normal brain tissues, ENTR1 was highly expressed in gliomas and correlated with malignant grades of gliomas and poor overall survival time. The proliferation and invasion of GBM cells could be weaken and the sensitivity to temozolomide (TMZ) chemotherapy increased after knocking down ENTR1. Overexpression of ENTR1 could reverse this effect. RNA-seq analysis showed that tumor necrosis factor (TNF) signaling pathway might be a putative regulatory target of ENTR1. Tumor formation assay validated that ENTR1 was a significant factor in tumor growth. CONCLUSION Our results indicated that ENTR1 played an important role in cell proliferation, invasion and chemotherapeutic sensitivity of GBM, suggesting that ENTR1 might be a novel prognostic marker and significant therapeutic target for GBM.
Collapse
Affiliation(s)
- Qian Zhang
- Department of Neurosurgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, Zhejiang, China
| | - Zhang'an Dai
- Department of Neurosurgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, Zhejiang, China
| | - Yingyu Chen
- Department of Neurosurgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, Zhejiang, China
| | - Qun Li
- Department of Neurosurgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, Zhejiang, China
| | - Yuhang Guo
- Department of Neurosurgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, Zhejiang, China
- Department of Neurosurgery, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, China
| | - Zhangzhang Zhu
- Department of Neurosurgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, Zhejiang, China
| | - Ming Tu
- Department of Neurosurgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, Zhejiang, China
| | - Lin Cai
- Department of Neurosurgery, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, China.
| | - Xianghe Lu
- Department of Neurosurgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, Zhejiang, China.
| |
Collapse
|
6
|
Lu Z, Zhou X, Fu L, Li Y, Tian T, Liu Q, Zou H. Prognostic prediction models for oropharyngeal squamous cell carcinoma (OPSCC): a protocol for systematic review, critical appraisal and meta-analysis. BMJ Open 2023; 13:e073375. [PMID: 37827742 PMCID: PMC10582955 DOI: 10.1136/bmjopen-2023-073375] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/03/2023] [Accepted: 08/25/2023] [Indexed: 10/14/2023] Open
Abstract
INTRODUCTION Oropharyngeal squamous cell carcinoma (OPSCC) is increasingly prevalent and has significantly heterogeneous risks of survival for diagnosed individuals due to the inter-related risk factors. Precise prediction of the risk of survival for an individual patient with OPSCC presents a useful adjunct to therapeutic decision-making regarding the management of OPSCC. The aim of this systematic review, critical appraisal and meta-analysis is to assess prognostic prediction models for OPSCC and lay a foundation for future research programmes to develop and validate prognostic prediction models for OPSCC. METHODS AND ANALYSIS This protocol will follow the Preferred Reporting Items for Systematic Review and Meta-Analyses Protocol statement. Based on predefined criteria, electronic databases including MEDLINE, Embase, Web of Science, the Cochrane Library and China National Knowledge Infrastructure (CNKI) will be searched for relevant studies without language restrictions from inception of databases to present. This study will systematically review published prognostic prediction models for survival outcomes in patients with OPSCC, describe their characteristics, compare performance and assess risk of bias and real-world clinical utility. Selection of eligible studies, data extraction and critical appraisal will be conducted independently by two reviewers. A third reviewer will resolve any disagreements. Included studies will be systematically summarised using appropriate tools designed for prognostic prediction modelling studies. Risk of bias and quality of studies will be assessed using the Prediction Model Risk of Bias Assessment Tool and the Transparent Reporting of a multivariable prediction model for individual prognosis or diagnosis. Performance measures of these models will be pooled and analysed with meta-analyses if feasible. ETHICS AND DISSEMINATION This review will be conducted completely based on published data, so approval from an ethics committee or written consent is not required. The results will be disseminated through a peer-reviewed publication. PROSPERO REGISTRATION NUMBER CRD42023400272.
Collapse
Affiliation(s)
- Zhen Lu
- School of Public Health (Shenzhen), Sun Yat-sen University, Shenzhen, Guangdong, China
| | - Xinyi Zhou
- School of Public Health (Shenzhen), Sun Yat-sen University, Shenzhen, Guangdong, China
| | - Leiwen Fu
- School of Public Health (Shenzhen), Sun Yat-sen University, Shenzhen, Guangdong, China
| | - Yuwei Li
- School of Public Health (Shenzhen), Sun Yat-sen University, Shenzhen, Guangdong, China
| | - Tian Tian
- School of Public Health (Shenzhen), Sun Yat-sen University, Shenzhen, Guangdong, China
| | - Qi Liu
- School of Public Health (Shenzhen), Sun Yat-sen University, Shenzhen, Guangdong, China
| | - Huachun Zou
- School of Public Health, Fudan University, Shanghai, China
- Fujian Maternity and Child Health Hospital, College of Clinical Medicine for Obstetrics and Gynecology and Pediatrics, Fujian Medical University, Fuzhou, Fujian, China
- School of Public Health, Southwest Medical University, Luzhou, Sichuan, China
- Kirby Institute, University of New South Wales, Sydney, New South Wales, Australia
| |
Collapse
|
7
|
Ashkan K, Baig Mirza A, Soumpasis C, Syrris C, Kalaitzoglou D, Sharma C, James ZJ, Khoja AK, Ahmed R, Vastani A, Bartram J, Chia K, Al-Salihi O, Swampilai A, Brazil L, Laxton R, Reisz Z, Bodi I, King A, Gullan R, Vergani F, Bhangoo R, Al-Sarraj S, Lavrador JP. MGMT Promoter Methylation: Prognostication beyond Treatment Response. J Pers Med 2023; 13:999. [PMID: 37373988 DOI: 10.3390/jpm13060999] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2023] [Revised: 06/08/2023] [Accepted: 06/09/2023] [Indexed: 06/29/2023] Open
Abstract
MGMT promoter methylation is related to the increased sensitivity of tumour tissue to chemotherapy with temozolomide (TMZ) and thus to improved patient survival. However, it is unclear how the extent of MGMT promoter methylation affects outcomes. In our study, a single-centre retrospective study, we explore the impact of MGMT promoter methylation in patients with glioblastoma who were operated upon with 5-ALA. Demographic, clinical and histology data, and survival rates were assessed. A total of 69 patients formed the study group (mean age 53.75 ± 15.51 years old). Positive 5-ALA fluorescence was noted in 79.41%. A higher percentage of MGMT promoter methylation was related to lower preoperative tumour volume (p = 0.003), a lower likelihood of 5-ALA positive fluorescence (p = 0.041) and a larger extent of resection EoR (p = 0.041). A higher MGMT promoter methylation rate was also related to improved progression-free survival (PFS) and overall survival (OS) (p = 0.008 and p = 0.006, respectively), even when adjusted for the extent of resection (p = 0.034 and p = 0.042, respectively). A higher number of adjuvant chemotherapy cycles was also related to longer PFS and OS (p = 0.049 and p = 0.030, respectively). Therefore, this study suggests MGMT promoter methylation should be considered as a continuous variable. It is a prognostic factor that goes beyond sensitivity to chemotherapy treatment, as a higher percentage of methylation is related not only to increased EoR and increased PFS and OS, but also to lower tumour volume at presentation and a lower likelihood of 5-ALA fluorescence intraoperatively.
Collapse
Affiliation(s)
- Keyoumars Ashkan
- Kings College Hospital NHS Foundation Trust, Denmark Hill, London SE5 9RS, UK
| | - Asfand Baig Mirza
- Kings College Hospital NHS Foundation Trust, Denmark Hill, London SE5 9RS, UK
| | - Christos Soumpasis
- Kings College Hospital NHS Foundation Trust, Denmark Hill, London SE5 9RS, UK
| | - Christoforos Syrris
- Kings College Hospital NHS Foundation Trust, Denmark Hill, London SE5 9RS, UK
| | | | - Chaitanya Sharma
- GKT School of Medicine, Kings College London, London SE1 1UL, UK
| | | | | | - Razna Ahmed
- GKT School of Medicine, Kings College London, London SE1 1UL, UK
| | - Amisha Vastani
- Kings College Hospital NHS Foundation Trust, Denmark Hill, London SE5 9RS, UK
| | - James Bartram
- Kings College Hospital NHS Foundation Trust, Denmark Hill, London SE5 9RS, UK
| | - Kazumi Chia
- Kings College Hospital NHS Foundation Trust, Denmark Hill, London SE5 9RS, UK
| | - Omar Al-Salihi
- Department of Neuro-Oncology, Cancer Centre, Guys Hospital, Great Maze Pond, London SE1 9RT, UK
| | - Angela Swampilai
- Department of Neuro-Oncology, Cancer Centre, Guys Hospital, Great Maze Pond, London SE1 9RT, UK
| | - Lucy Brazil
- Department of Neuro-Oncology, Cancer Centre, Guys Hospital, Great Maze Pond, London SE1 9RT, UK
| | - Ross Laxton
- Department of Neuropathology, Kings College London, London SE5 9RS, UK
| | - Zita Reisz
- Department of Neuropathology, Kings College London, London SE5 9RS, UK
| | - Istvan Bodi
- Department of Neuropathology, Kings College London, London SE5 9RS, UK
| | - Andrew King
- Department of Neuropathology, Kings College London, London SE5 9RS, UK
| | - Richard Gullan
- Kings College Hospital NHS Foundation Trust, Denmark Hill, London SE5 9RS, UK
| | - Francesco Vergani
- Kings College Hospital NHS Foundation Trust, Denmark Hill, London SE5 9RS, UK
| | - Ranjeev Bhangoo
- Kings College Hospital NHS Foundation Trust, Denmark Hill, London SE5 9RS, UK
| | - Safa Al-Sarraj
- Kings College Hospital NHS Foundation Trust, Denmark Hill, London SE5 9RS, UK
| | - Jose Pedro Lavrador
- Kings College Hospital NHS Foundation Trust, Denmark Hill, London SE5 9RS, UK
| |
Collapse
|
8
|
Gan HK. MGMT methylation: Is it time to embrace the shades of grey? Neurooncol Pract 2023; 10:111-112. [PMID: 36970166 PMCID: PMC10037944 DOI: 10.1093/nop/npad003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Affiliation(s)
- Hui K Gan
- Cancer Therapies and Biology Group, and Centre for Research Excellence in Brain Cancer, Olivia Newton-John Cancer Research Institute, Austin Health, Melbourne, Australia
- La Trobe University School of Cancer Medicine, Heidelberg, Victoria, Australia
- Department of Medicine, University of Melbourne, Heidelberg, Victoria, Australia
- Australian Brain Cancer Research Alliance, Melbourne, Australia
| |
Collapse
|
9
|
Zapata Laguado M, Baez JM, Luna A, Mantilla C, Palencia M. Bone Metastasis From Glioblastoma Multiforme: A Case Report. Cureus 2022; 14:e25464. [PMID: 35800795 PMCID: PMC9246435 DOI: 10.7759/cureus.25464] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/29/2022] [Indexed: 11/05/2022] Open
|
10
|
Yang K, Wu Z, Zhang H, Zhang N, Wu W, Wang Z, Dai Z, Zhang X, Zhang L, Peng Y, Ye W, Zeng W, Liu Z, Cheng Q. Glioma targeted therapy: insight into future of molecular approaches. Mol Cancer 2022; 21:39. [PMID: 35135556 PMCID: PMC8822752 DOI: 10.1186/s12943-022-01513-z] [Citation(s) in RCA: 386] [Impact Index Per Article: 128.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2021] [Accepted: 01/12/2022] [Indexed: 12/13/2022] Open
Abstract
Gliomas are the common type of brain tumors originating from glial cells. Epidemiologically, gliomas occur among all ages, more often seen in adults, which males are more susceptible than females. According to the fifth edition of the WHO Classification of Tumors of the Central Nervous System (WHO CNS5), standard of care and prognosis of gliomas can be dramatically different. Generally, circumscribed gliomas are usually benign and recommended to early complete resection, with chemotherapy if necessary. Diffuse gliomas and other high-grade gliomas according to their molecule subtype are slightly intractable, with necessity of chemotherapy. However, for glioblastoma, feasible resection followed by radiotherapy plus temozolomide chemotherapy define the current standard of care. Here, we discuss novel feasible or potential targets for treatment of gliomas, especially IDH-wild type glioblastoma. Classic targets such as the p53 and retinoblastoma (RB) pathway and epidermal growth factor receptor (EGFR) gene alteration have met failure due to complex regulatory network. There is ever-increasing interest in immunotherapy (immune checkpoint molecule, tumor associated macrophage, dendritic cell vaccine, CAR-T), tumor microenvironment, and combination of several efficacious methods. With many targeted therapy options emerging, biomarkers guiding the prescription of a particular targeted therapy are also attractive. More pre-clinical and clinical trials are urgently needed to explore and evaluate the feasibility of targeted therapy with the corresponding biomarkers for effective personalized treatment options.
Collapse
Affiliation(s)
- Keyang Yang
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, China
- Xiangya School of Medicine, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Zhijing Wu
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, China
- Xiangya School of Medicine, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Hao Zhang
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Nan Zhang
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
- One-Third Lab, College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, China
| | - Wantao Wu
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
- Department of Oncology, Xiangya Hospital, Central South University, Changsha, China
| | - Zeyu Wang
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Ziyu Dai
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Xun Zhang
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Liyang Zhang
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Yun Peng
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
- Department of Geriatrics, Xiangya Hospital, Central South University, Changsha, China
- Teaching and Research Section of Clinical Nursing, Xiangya Hospital of Central South University, Changsha, China
| | - Weijie Ye
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha, China
| | - Wenjing Zeng
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha, China
| | - Zhixiong Liu
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, China.
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China.
| | - Quan Cheng
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, China.
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China.
| |
Collapse
|
11
|
Wesseling P, Rozowsky JS. Neurooncology: 2022 update. FREE NEUROPATHOLOGY 2022; 3:4. [PMID: 37284148 PMCID: PMC10209868 DOI: 10.17879/freeneuropathology-2022-3804] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 02/14/2022] [Accepted: 02/23/2022] [Indexed: 06/08/2023]
Abstract
This 'Neurooncology: 2022 update' presents topics that were selected by the authors as top ten discoveries published in 2021 in the broader field of neurooncological pathology. This time, the spectrum of topics includes: papers with a direct impact on daily diagnostic practice of CNS tumors in general and with information on how to improve grading of meningiomas; studies shedding new light on the oncogenesis of gliomas (in particular 'optic gliomas' and H3-mutant gliomas); several 'multi-omic' investigations unraveling the intra-tumoral heterogeneity of especially glioblastomas further; a study indicating the potential of 'repurposing' Prozac® for the treatment of glioblastomas; liquid biopsy using CSF for assessment of residual medulloblastoma. In the last part of this review some other papers are mentioned that didn't make it to this (quite subjective) top ten list.
Collapse
Affiliation(s)
- Pieter Wesseling
- Department of Pathology, Amsterdam University Medical Centers/VUmc, Brain Tumor Center Amsterdam, De Boelelaan 1117, 1081HV AmsterdamThe Netherlands
- Laboratory for Childhood Cancer Pathology, Princess Máxima Center for Pediatric Oncology, Heidelberglaan 25, 3584 CS UtrechtThe Netherlands
| | - Jacob S. Rozowsky
- Department of Pathology, Amsterdam University Medical Centers/VUmc, Brain Tumor Center Amsterdam, De Boelelaan 1117, 1081HV AmsterdamThe Netherlands
| |
Collapse
|
12
|
Dai Z, Cai L, Chen Y, Wang S, Zhang Q, Wang C, Tu M, Zhu Z, Li Q, Lu X. Brusatol Inhibits Proliferation and Invasion of Glioblastoma by Down-Regulating the Expression of ECM1. Front Pharmacol 2022; 12:775680. [PMID: 34970146 PMCID: PMC8713816 DOI: 10.3389/fphar.2021.775680] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2021] [Accepted: 11/30/2021] [Indexed: 12/13/2022] Open
Abstract
Brusatol (Bru), a Chinese herbal extract, has a variety of anti-tumor effects. However, little is known regarding its role and underlying mechanism in glioblastoma cells. Here, we found that Bru could inhibit the proliferation of glioblastoma cells in vivo and in vitro. Besides, it also had an inhibitory effect on human primary glioblastoma cells. RNA-seq analysis indicated that Bru possibly achieved these effects through inhibiting the expression of extracellular matrix protein 1 (ECM1). Down-regulating the expression of ECM1 via transfecting siRNA could weaken the proliferation and invasion of glioblastoma cells and promote the inhibitory effect of Bru treatment. Lentivirus-mediated overexpression of ECM1 could effectively reverse this weakening effect. Our findings indicated that Bru could inhibit the proliferation and invasion of glioblastoma cells by suppressing the expression of ECM1, and Bru might be a novel effective anticancer drug for glioblastoma cells.
Collapse
Affiliation(s)
- Zhang'an Dai
- Department of Neurosurgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Lin Cai
- Department of Neurosurgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China.,Department of Neurosurgery, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China
| | - Yingyu Chen
- Department of Neurosurgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Silu Wang
- Department of Neurosurgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Qian Zhang
- Department of Neurosurgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Chengde Wang
- Department of Neurosurgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Ming Tu
- Department of Neurosurgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Zhangzhang Zhu
- Department of Neurosurgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Qun Li
- Department of Neurosurgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Xianghe Lu
- Department of Neurosurgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| |
Collapse
|
13
|
Independently validated sex-specific nomograms for predicting survival in patients with newly diagnosed glioblastoma: NRG Oncology RTOG 0525 and 0825. J Neurooncol 2021; 155:363-372. [PMID: 34761331 PMCID: PMC8651582 DOI: 10.1007/s11060-021-03886-5] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2021] [Accepted: 10/25/2021] [Indexed: 02/08/2023]
Abstract
Background/purpose Glioblastoma (GBM) is the most common primary malignant brain tumor. Sex has been shown to be an important prognostic factor for GBM. The purpose of this study was to develop and independently validate sex-specific nomograms for estimation of individualized GBM survival probabilities using data from 2 independent NRG Oncology clinical trials. Methods This analysis included information on 752 (NRG/RTOG 0525) and 599 (NRG/RTOG 0825) patients with newly diagnosed GBM. The Cox proportional hazard models by sex were developed using NRG/RTOG 0525 and significant variables were identified using a backward selection procedure. The final selected models by sex were then independently validated using NRG/RTOG 0825. Results Final nomograms were built by sex. Age at diagnosis, KPS, MGMT promoter methylation and location of tumor were common significant predictors of survival for both sexes. For both sexes, tumors in the frontal lobes had significantly better survival than tumors of multiple sites. Extent of resection, and use of corticosteroids were significant predictors of survival for males. Conclusions A sex specific nomogram that assesses individualized survival probabilities (6-, 12- and 24-months) for patients with GBM could be more useful than estimation of overall survival as there are factors that differ between males and females. A user friendly online application can be found here—https://npatilshinyappcalculator.shinyapps.io/SexDifferencesInGBM/. Supplementary Information The online version contains supplementary material available at 10.1007/s11060-021-03886-5.
Collapse
|
14
|
Affiliation(s)
- Monika E Hegi
- Neuroscience Research Center and Service of Neurosurgery, Lausanne University Hospital and University of Lausanne, Lausanne Switzerland
| | - Koichi Ichimura
- Department of Brain Disease Translational Research, Juntendo University Faculty of Medicine, Tokyo; Japan
| |
Collapse
|
15
|
Sulman EP, Eisenstat DD. World Cancer Day 2021 - Perspectives in Pediatric and Adult Neuro-Oncology. Front Oncol 2021; 11:659800. [PMID: 34041027 PMCID: PMC8142853 DOI: 10.3389/fonc.2021.659800] [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] [Received: 01/28/2021] [Accepted: 04/07/2021] [Indexed: 12/13/2022] Open
Abstract
Significant advances in our understanding of the molecular genetics of pediatric and adult brain tumors and the resulting rapid expansion of clinical molecular neuropathology have led to improvements in diagnostic accuracy and identified new targets for therapy. Moreover, there have been major improvements in all facets of clinical care, including imaging, surgery, radiation and supportive care. In selected cohorts of patients, targeted and immunotherapies have resulted in improved patient outcomes. Furthermore, adaptations to clinical trial design have facilitated our study of new agents and other therapeutic innovations. However, considerable work remains to be done towards extending survival for all patients with primary brain tumors, especially children and adults with diffuse midline gliomas harboring Histone H3 K27 mutations and adults with isocitrate dehydrogenase (IDH) wild-type, O6 guanine DNA-methyltransferase gene (MGMT) promoter unmethylated high grade gliomas. In addition to improvements in therapy and care, access to the advances in technology, such as particle radiation or biologic therapy, neuroimaging and molecular diagnostics in both developing and developed countries is needed to improve the outcome of patients with brain tumors.
Collapse
Affiliation(s)
- Erik P. Sulman
- Section of Neuro-oncology & Neurosurgical Oncology, Frontiers in Oncology and Frontiers in Neurology, Lausanne, Switzerland
- Department of Radiation Oncology, NYU Grossman School of Medicine, New York, NY, United States
- Brain and Spine Tumor Center, Laura and Isaac Perlmutter Cancer Center, New York, NY, United States
- NYU Langone Health, New York, NY, United States
| | - David D. Eisenstat
- Section of Neuro-oncology & Neurosurgical Oncology, Frontiers in Oncology and Frontiers in Neurology, Lausanne, Switzerland
- Children’s Cancer Centre, Royal Children’s Hospital, Parkville, VIC, Australia
- Murdoch Children’s Research Institute, Parkville, VIC, Australia
- Department of Paediatrics, University of Melbourne, Parkville, VIC, Australia
| |
Collapse
|
16
|
McAleenan A, Kelly C, Spiga F, Kernohan A, Cheng HY, Dawson S, Schmidt L, Robinson T, Brandner S, Faulkner CL, Wragg C, Jefferies S, Howell A, Vale L, Higgins JPT, Kurian KM. Prognostic value of test(s) for O6-methylguanine-DNA methyltransferase (MGMT) promoter methylation for predicting overall survival in people with glioblastoma treated with temozolomide. Cochrane Database Syst Rev 2021; 3:CD013316. [PMID: 33710615 PMCID: PMC8078495 DOI: 10.1002/14651858.cd013316.pub2] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
BACKGROUND Glioblastoma is an aggressive form of brain cancer. Approximately five in 100 people with glioblastoma survive for five years past diagnosis. Glioblastomas that have a particular modification to their DNA (called methylation) in a particular region (the O6-methylguanine-DNA methyltransferase (MGMT) promoter) respond better to treatment with chemotherapy using a drug called temozolomide. OBJECTIVES To determine which method for assessing MGMT methylation status best predicts overall survival in people diagnosed with glioblastoma who are treated with temozolomide. SEARCH METHODS We searched MEDLINE, Embase, BIOSIS, Web of Science Conference Proceedings Citation Index to December 2018, and examined reference lists. For economic evaluation studies, we additionally searched NHS Economic Evaluation Database (EED) up to December 2014. SELECTION CRITERIA Eligible studies were longitudinal (cohort) studies of adults with diagnosed glioblastoma treated with temozolomide with/without radiotherapy/surgery. Studies had to have related MGMT status in tumour tissue (assessed by one or more method) with overall survival and presented results as hazard ratios or with sufficient information (e.g. Kaplan-Meier curves) for us to estimate hazard ratios. We focused mainly on studies comparing two or more methods, and listed brief details of articles that examined a single method of measuring MGMT promoter methylation. We also sought economic evaluations conducted alongside trials, modelling studies and cost analysis. DATA COLLECTION AND ANALYSIS Two review authors independently undertook all steps of the identification and data extraction process for multiple-method studies. We assessed risk of bias and applicability using our own modified and extended version of the QUality In Prognosis Studies (QUIPS) tool. We compared different techniques, exact promoter regions (5'-cytosine-phosphate-guanine-3' (CpG) sites) and thresholds for interpretation within studies by examining hazard ratios. We performed meta-analyses for comparisons of the three most commonly examined methods (immunohistochemistry (IHC), methylation-specific polymerase chain reaction (MSP) and pyrosequencing (PSQ)), with ratios of hazard ratios (RHR), using an imputed value of the correlation between results based on the same individuals. MAIN RESULTS We included 32 independent cohorts involving 3474 people that compared two or more methods. We found evidence that MSP (CpG sites 76 to 80 and 84 to 87) is more prognostic than IHC for MGMT protein at varying thresholds (RHR 1.31, 95% confidence interval (CI) 1.01 to 1.71). We also found evidence that PSQ is more prognostic than IHC for MGMT protein at various thresholds (RHR 1.36, 95% CI 1.01 to 1.84). The data suggest that PSQ (mainly at CpG sites 74 to 78, using various thresholds) is slightly more prognostic than MSP at sites 76 to 80 and 84 to 87 (RHR 1.14, 95% CI 0.87 to 1.48). Many variants of PSQ have been compared, although we did not see any strong and consistent messages from the results. Targeting multiple CpG sites is likely to be more prognostic than targeting just one. In addition, we identified and summarised 190 articles describing a single method for measuring MGMT promoter methylation status. AUTHORS' CONCLUSIONS PSQ and MSP appear more prognostic for overall survival than IHC. Strong evidence is not available to draw conclusions with confidence about the best CpG sites or thresholds for quantitative methods. MSP has been studied mainly for CpG sites 76 to 80 and 84 to 87 and PSQ at CpG sites ranging from 72 to 95. A threshold of 9% for CpG sites 74 to 78 performed better than higher thresholds of 28% or 29% in two of three good-quality studies making such comparisons.
Collapse
Affiliation(s)
- Alexandra McAleenan
- Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, UK
| | - Claire Kelly
- Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, UK
| | - Francesca Spiga
- Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, UK
| | - Ashleigh Kernohan
- Population Health Sciences Institute, Newcastle University, Newcastle upon Tyne, UK
| | - Hung-Yuan Cheng
- Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, UK
| | - Sarah Dawson
- Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, UK
- NIHR Applied Research Collaboration West (ARC West) , University Hospitals Bristol and Weston NHS Foundation Trust, Bristol, UK
| | - Lena Schmidt
- Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, UK
| | - Tomos Robinson
- Population Health Sciences Institute, Newcastle University, Newcastle upon Tyne, UK
| | - Sebastian Brandner
- Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, London, UK
- Division of Neuropathology, The National Hospital for Neurology and Neurosurgery, University College London Hospitals NHS Foundation Trust, London, UK
| | - Claire L Faulkner
- Bristol Genetics Laboratory, Pathology Sciences, Southmead Hospital, Bristol, UK
| | - Christopher Wragg
- Bristol Genetics Laboratory, Pathology Sciences, Southmead Hospital, Bristol, UK
| | - Sarah Jefferies
- Department of Oncology, Addenbrooke's Hospital, Cambridge, UK
| | - Amy Howell
- Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, UK
| | - Luke Vale
- Population Health Sciences Institute, Newcastle University, Newcastle upon Tyne, UK
| | - Julian P T Higgins
- Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, UK
- NIHR Applied Research Collaboration West (ARC West) , University Hospitals Bristol and Weston NHS Foundation Trust, Bristol, UK
- NIHR Bristol Biomedical Research Centre, University Hospitals Bristol and Weston NHS Foundation Trust, Bristol, UK
| | - Kathreena M Kurian
- Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, UK
- Bristol Medical School: Brain Tumour Research Centre, Public Health Sciences, University of Bristol, Bristol, UK
| |
Collapse
|
17
|
Poon MTC, Keni S, Vimalan V, Ip C, Smith C, Erridge S, Weir CJ, Brennan PM. Extent of MGMT promoter methylation modifies the effect of temozolomide on overall survival in patients with glioblastoma: a regional cohort study. Neurooncol Adv 2021; 3:vdab171. [PMID: 34988453 PMCID: PMC8704383 DOI: 10.1093/noajnl/vdab171] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
BACKGROUND MGMT methylation in glioblastoma predicts response to temozolomide but dichotomizing methylation status may mask the true prognostic value of quantitative MGMT methylation. This study evaluated whether extent of MGMT methylation interacts with the effect of temozolomide on overall survival. METHODS We included consecutive glioblastoma patients aged ≥16 years diagnosed (April 2012-May 2020) at a neuro-oncology center. All patients had quantitative MGMT methylation measured using pyrosequencing. Those with MGMT methylated tumors were stratified into high and low methylation groups based on a cut-off using Youden index on 2-year survival. Our accelerated failure time survival models included extent of MGMT methylation, age, postoperative Karnofsky performance score, extent of resection, temozolomide regimen, and radiotherapy. RESULTS There were 414 patients. Optimal cut-off point using Youden index was 25.9% MGMT methylation. The number of patients in the unmethylated, low and high methylation groups was 223 (53.9%), 81 (19.6%), and 110 (26.6%), respectively. In the adjusted model, high (hazard ratio [HR] 0.60, 95% confidence intervals [CI] 0.46-0.79, P = 0.005) and low (HR 0.67, 95% CI 0.50-0.89, P < 0.001) methylation groups had better survival compared to unmethylated group. There was no evidence for interaction between MGMT methylation and completed temozolomide regimen (interaction term for low methylation P = 0.097; high methylation P = 0.071). This suggests no strong effect of MGMT status on survival in patients completing temozolomide regimen. In patients not completing the temozolomide regimen, higher MGMT methylation predicted better survival (interaction terms P < 0.001). CONCLUSIONS Quantitative MGMT methylation may provide additional prognostic value. This is important when assessing clinical and research therapies.
Collapse
Affiliation(s)
- Michael T C Poon
- Centre for Medical Informatics, Usher Institute, University of Edinburgh, UK
- Department of Clinical Neuroscience, Royal Infirmary of Edinburgh, NHS Lothian, UK
| | - Shivank Keni
- Edinburgh Medical School, University of Edinburgh, UK
| | - Vineeth Vimalan
- Department of Clinical Neuroscience, Royal Infirmary of Edinburgh, NHS Lothian, UK
| | - Chak Ip
- Royal Infirmary of Edinburgh, NHS Lothian, UK
| | - Colin Smith
- Centre for Comparative Pathology, University of Edinburgh, UK
| | - Sara Erridge
- Clinical Oncology, Western General Hospital, NHS Lothian, UK
| | - Christopher J Weir
- Edinburgh Clinical Trials Unit, Usher Institute, University of Edinburgh, UK
| | - Paul M Brennan
- Department of Clinical Neuroscience, Royal Infirmary of Edinburgh, NHS Lothian, UK
- Cancer Research UK Brain Tumour Centre of Excellence, CRUK Edinburgh Centre, University of Edinburgh, UK
- Translational Neurosurgery, Centre for Clinical Brain Sciences, University of Edinburgh, UK
| |
Collapse
|