1
|
He D, Wei S, Geng F, Li L, Li F, Ge Y, Lv R, Li W, Hao Z, Jiang F, Meng C, Lu S, Zhang S. Prophylactic cerebral irradiation sensitizes relapsed sensitive small cell lung cancer to temozolomide: A retrospective cohort study. Oncol Lett 2025; 30:327. [PMID: 40370642 PMCID: PMC12076553 DOI: 10.3892/ol.2025.15073] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2024] [Accepted: 02/24/2025] [Indexed: 05/16/2025] Open
Abstract
Prophylactic cerebral irradiation (PCI) reduces the rate of brain metastasis and improves the prognosis of patients with small cell lung cancer (SCLC), but little is known about the effect of PCI on second-line chemotherapy in patients with relapsed sensitive SCLC. This retrospective cohort study included a total of 164 patients with relapsed sensitive SCLC, 20 of whom were administered temozolomide (TMZ). Categorical clinical variables were compared between subgroups with the chi-square test or Fisher's exact test, continuous clinical variables were compared with the t-test or one-way ANOVA, and the impact on overall survival (OS) was assessed using Kaplan-Meier analysis with the log-rank test. In general, TMZ prolonged the OS of patients with SCLC with brain metastasis from 12.0 to 19.0 months [P=0.0109, hazard ratio (HR): 0.4789, 95% CI: 0.2470-0.9287]. Furthermore, the administration of PCI improved the effects of TMZ on patients with SCLC with brain metastasis, with an increase in OS from 16.0 to 36.5 months (P=0.0017, HR: 3.634, 95% CI: 1.083-12.20); additionally, no difference was observed on the basis of the history of chemotherapy or state of brain metastasis. For the local response evaluation, the overall response rate reached 75.0% for both brain metastasis and extracranial lesions in the two-cycle evaluation, remained at 30.0 and 25.0% in the four-cycle and more-cycle evaluations, respectively, and was minimally influenced by the history of chemotherapy or PCI. In conclusion, the results of this study suggest that PCI may be valuable for patients with relapsed sensitive SCLC with brain metastasis who are receiving TMZ treatment, and it may also serve as an effective regimen to prevent local progression of extracranial lesions; however, more evidence is needed.
Collapse
Affiliation(s)
- Dan He
- Department of Oncology, The Second Affiliated Hospital of Chengdu Medical College, China National Nuclear Corporation 416 Hospital, Chengdu, Sichuan 610041, P.R. China
- Department of Radiation Medicine, West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu, Sichuan 610041, P.R. China
| | - Shuxia Wei
- Department of Oncology, The Hospital of 81st Group Army, Zhangjiakou, Hebei 075000, P.R. China
| | - Fenghao Geng
- Department of Radiation Medicine, West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu, Sichuan 610041, P.R. China
- Department of Oncology, The Hospital of 81st Group Army, Zhangjiakou, Hebei 075000, P.R. China
- Radiotherapy Center, Radiation Oncology Key Laboratory of Sichuan Province, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Hospital & Institute, Affiliated Cancer Hospital of University of Electronic Science and Technology of China, Chengdu, Sichuan 610041, P.R. China
| | - Lintao Li
- Radiotherapy Center, Radiation Oncology Key Laboratory of Sichuan Province, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Hospital & Institute, Affiliated Cancer Hospital of University of Electronic Science and Technology of China, Chengdu, Sichuan 610041, P.R. China
| | - Fengyu Li
- Department of Oncology, The Hospital of 81st Group Army, Zhangjiakou, Hebei 075000, P.R. China
| | - Yanli Ge
- Department of Oncology, The Hospital of 81st Group Army, Zhangjiakou, Hebei 075000, P.R. China
| | - Ruichang Lv
- Department of Oncology, The Hospital of 81st Group Army, Zhangjiakou, Hebei 075000, P.R. China
| | - Weiwei Li
- Department of Oncology, The Hospital of 81st Group Army, Zhangjiakou, Hebei 075000, P.R. China
| | - Zhijun Hao
- Department of Oncology, The Hospital of 81st Group Army, Zhangjiakou, Hebei 075000, P.R. China
| | - Fengdi Jiang
- Department of Radiation Medicine, West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu, Sichuan 610041, P.R. China
| | - Chao Meng
- Department of Nephrology, Huayao Hospital of North China Medical and Health Group, Shijiazhuang, Hebei 050011, P.R. China
| | - Shun Lu
- Radiotherapy Center, Radiation Oncology Key Laboratory of Sichuan Province, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Hospital & Institute, Affiliated Cancer Hospital of University of Electronic Science and Technology of China, Chengdu, Sichuan 610041, P.R. China
| | - Shuyu Zhang
- Department of Radiation Medicine, West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu, Sichuan 610041, P.R. China
- Department of Nuclear Medicine, The Second Affiliated Hospital of Chengdu Medical College, China National Nuclear Corporation 416 Hospital, Chengdu, Sichuan 610041, P.R. China
- National Health Commission Key Laboratory of Nuclear Technology Medical Transformation, Mianyang Central Hospital, Mianyang, Sichuan 621000, P.R. China
| |
Collapse
|
2
|
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
|
3
|
Morfouace M, Bielle F, Razis E, Estrade F, Rubio A, Bautista F, de Rojas T, Vieito M, Meade S, Sanson M, Marques AC, Preusser M, Hatcher H, Balasubramanian GP, Pineda E, D'Hondt L, Duerinck J, Michotte A, Mawrin C, Ribalta T, Marucci G, Golfinopoulos V, Pfister SM, Jones DT, McCabe MG. Molecular analysis of adolescent and young adult high grade gliomas in the SPECTA-AYA study: Poorly characterised tumours with frequent germline alterations. Eur J Cancer 2025; 223:115493. [PMID: 40393126 DOI: 10.1016/j.ejca.2025.115493] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2024] [Revised: 04/22/2025] [Accepted: 05/04/2025] [Indexed: 05/22/2025]
Abstract
BACKGROUND Adolescent and young adult (AYA) high grade gliomas (HGG) have the worst survival of AYA malignancies yet are poorly represented in large-scale molecular datasets. METHODS 50 AYAs aged 12-29 with newly diagnosed or recurrent HGG and other high risk central nervous system (CNS) tumours were prospectively recruited to the EORTC SPECTA platform study and underwent whole exome sequencing, RNA sequencing and methylation profiling, with central pathological review. Actionable mutations were reported and patients followed up for therapies and outcome. RESULTS From 46 locally diagnosed HGGs and 4 other recurrent CNS tumours, molecular and pathology review resulted in histological grade re-classification (n = 10), diagnostic refinement (n = 9) and revised diagnoses (n = 12) in a substantial proportion. Pathogenic constitutional alterations were present in 14 % overall and were largely limited to cases with IDH-wildtype glioblastoma and paediatric-type diffuse HGGs. 91 % of HGGs had potentially actionable alterations affecting RAS/RAF/MAPK (60 %), PI3K/AKT/mTOR (27 %) and cell cycle genes (11 %). High tumour mutational burden (> 10 somatic non-synonymous mutations per Mb of genome targeted) was present in 12 % at diagnosis and 18 % at recurrence, all in histological grade 4 tumours. Ten patients' treatment was modified on the basis of molecular profile, of whom 5 remained on treatment at last follow-up. CONCLUSION AYA HGGs comprise a diverse group of entities; accurate, molecularly-defined diagnosis is critical to direct primary treatment, determine risk of genetic predisposition and guide molecularly-directed therapy. Current services fail to routinely address diagnosis, personalised molecular profiling or investigation of therapeutic opportunities for this high risk, poor prognosis group of rare cancer patients.
Collapse
Affiliation(s)
- Marie Morfouace
- EORTC Headquarters, Brussels, Belgium; Gustave Roussy, Villejuif, France
| | - Franck Bielle
- Sorbonne Université, Paris Brain Institute-ICM, Inserm, CNRS, AP-HP, Hôpitaux Universitaires La Pitié Salpêtrière-Charles Foix, Department of Neuropathology, Paris, France
| | - Evangelia Razis
- Third Medical Oncology Dept, Hygeia Hospital, Athens, Greece
| | | | - Alba Rubio
- Hospital Niño Jesús, Department of Pediatric Oncology, Hematology and Stem Cell Transplantation, Madrid, Spain
| | - Francisco Bautista
- Hospital Niño Jesús, Department of Pediatric Oncology, Hematology and Stem Cell Transplantation, Madrid, Spain; Princess Maxima Center for Pediatric Oncology, Utrecht, the Netherlands
| | | | - Maria Vieito
- GU, Sarcoma and Neuroncology Unit, Vall d'Hebron University Hospital and Drug Development Unit, Vall d'Hebron Institute of Oncology (VHIO), Barcelona, Spain
| | - Sara Meade
- UHB-Queen Elizabeth Medical Centre, Birmingham, UK
| | - Marc Sanson
- Sorbonne Université, Paris Brain Institute-ICM, Inserm, CNRS, AP-HP, Hôpitaux Universitaires La Pitié Salpêtrière-Charles Foix, Department of Neuro-oncology, Paris, France
| | | | - Matthias Preusser
- Division of Oncology, Department of Medicine I, Medical University of Vienna, Vienna, Austria
| | - Helen Hatcher
- Department of Medical Oncology, Addenbrookes Hospital, Cambridge, UK
| | - Gnana Prakash Balasubramanian
- Hopp Children´s Cancer Center Heidelberg (KiTZ), German Consortium for Translational Cancer Research (DKTK), Germany; Division Pediatric Neurooncology, German Cancer Research Center (DKFZ), Heidelberg, Germany; National Center for Tumor Diseases (NCT), Heidelberg, Germany
| | | | - Lionel D'Hondt
- Department of Oncology CHU UCL Namur site Godinne, Yvoir, Belgium
| | - Johnny Duerinck
- Department of Neurosurgery, Universitair Ziekenhuis Brussel (UZ Brussel), Vrije Universiteit Brussel (VUB), Laarbeeklaan 101, Brussels 1090, Belgium
| | - Alex Michotte
- Dept of Pathology (Neuropathology), Universitair Ziekenhuis Brussel, Brussels, Belgium
| | | | - Teresa Ribalta
- Pathology Department, Hospital Clinic, Institute of Biomedical Research August Pi i Sunyer (IDIBAPS), University of Barcelona, Barcelona, Spain
| | - Gianluca Marucci
- Neuropathology Unit, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan, Italy
| | | | - Stefan M Pfister
- Hopp Children´s Cancer Center Heidelberg (KiTZ), German Consortium for Translational Cancer Research (DKTK), Germany; Division Pediatric Neurooncology, German Cancer Research Center (DKFZ), Heidelberg, Germany; National Center for Tumor Diseases (NCT), Heidelberg, Germany
| | - David Tw Jones
- National Center for Tumor Diseases (NCT), Heidelberg, Germany; Division of Pediatric Glioma Research, Hopp Children's Cancer Center Heidelberg (KiTZ), Heidelberg, Germany
| | - Martin G McCabe
- Division of Cancer Sciences, University of Manchester, Manchester, UK; The Christie NHS Foundation Trust, Manchester, UK.
| |
Collapse
|
4
|
Qiu Y, Li Y, Jiang C, Wu X, Liu W, Fan C, Ye X, He L, Xiao S, Zhao Q, Wu W, Chen K, Tan C, Li Y, Wang H, Liu F. Toxicity and Efficacy of Different Target Volume Delineations of Radiation Therapy Based on the Updated Radiation Therapy Oncology Group/National Research Group and European Organization for Research and Treatment of Cancer Guidelines in Patients With Grade 3-4 Glioma: A Randomized Controlled Clinical Trial. Int J Radiat Oncol Biol Phys 2025; 121:1168-1181. [PMID: 39615657 DOI: 10.1016/j.ijrobp.2024.11.094] [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: 04/10/2024] [Revised: 11/12/2024] [Accepted: 11/15/2024] [Indexed: 12/22/2024]
Abstract
PURPOSE Our study aimed to evaluate the safety and efficacy of radiation therapy (RT) in the treatment of grade 3-4 glioma by comparing the updated Radiation Therapy Oncology Group (RTOG)/National Research Group (NRG) with European Organization for Research and Treatment of Cancer (EORTC) guidelines for target volume delineation. METHODS AND MATERIALS A total of 245 patients with newly diagnosed World Health Organization grade 3-4 glioma were enrolled and randomly assigned (1:1 ratio) to undergo postoperative RT with concurrent and maintenance temozolomide. The radiation target volume delineation was determined by using either the updated RTOG/NRG (n = 122) or EORTC guidelines (n = 123). The primary endpoint was the toxicity associated with treatment. Progression-free survival (PFS) and overall survival (OS) were considered secondary endpoints. RESULTS No differences in low- or high-grade toxicities between the 2 groups, and neither group exhibited grade 5 toxicities. No significant differences in neurologic toxicities were observed between the RTOG/NRG and EORTC groups. The median PFS in the RTOG/NRG group and the EORTC group was 11.0 months (95% confidence interval [CI], 7.1-14.9 months) and 10.0 months (95% CI, 3.8-16.2 months), respectively (P = .73). The median OS in the RTOG/NRG group and the EORTC group was 19.5 months (95% CI, 14.2-24.8 months) and 18.5 months (95% CI, 12.8-24.2 months), respectively (P = .80). In patients with isocitrate dehydrogenase wild-type glioblastoma, there were no significant differences between the RTOG/NRG group and the EORTC group in median PFS (8.0 months [95% CI, 6.8-9.2 months] vs. 8.0 months [95% CI, 7.0-9.0 months], P = .38) and median OS (12.0 months [95% CI, 7.2-16.8 months] vs. 11.0 months [95% CI, 9.7-12.3 months], P = .10). CONCLUSIONS Compared with EORTC principles, postoperative RT according to RTOG/NRG principles did not increase treatment-related toxicities and was equally effective for patients with grade 3-4 glioma, including the subgroup of patients with isocitrate dehydrogenase wild-type glioblastoma.
Collapse
Affiliation(s)
- Yanfang Qiu
- Department of Radiation Oncology, The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University/Hunan Cancer Hospital, Hunan, China
| | - Yanxian Li
- Department of Radiation Oncology, The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University/Hunan Cancer Hospital, Hunan, China
| | - Cuihong Jiang
- Department of Radiation Oncology, The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University/Hunan Cancer Hospital, Hunan, China
| | - Xiangwei Wu
- Department of Radiation Oncology, The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University/Hunan Cancer Hospital, Hunan, China
| | - Wen Liu
- Department of Radiation Oncology, The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University/Hunan Cancer Hospital, Hunan, China
| | - Changgen Fan
- Department of Radiation Oncology, The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University/Hunan Cancer Hospital, Hunan, China
| | - Xu Ye
- Department of Radiation Oncology, The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University/Hunan Cancer Hospital, Hunan, China
| | - Lili He
- Department of Radiation Oncology, The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University/Hunan Cancer Hospital, Hunan, China
| | - Shuai Xiao
- Department of Radiation Oncology, The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University/Hunan Cancer Hospital, Hunan, China
| | - Qi Zhao
- Department of Radiation Oncology, The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University/Hunan Cancer Hospital, Hunan, China
| | - Wenqiong Wu
- Department of Radiation Oncology, The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University/Hunan Cancer Hospital, Hunan, China
| | - Kailin Chen
- Department of Radiation Oncology, The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University/Hunan Cancer Hospital, Hunan, China
| | - Chao Tan
- Department of Radiation Oncology, The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University/Hunan Cancer Hospital, Hunan, China
| | - Yuyi Li
- Department of Radiation Oncology, The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University/Hunan Cancer Hospital, Hunan, China
| | - Hui Wang
- Department of Radiation Oncology, The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University/Hunan Cancer Hospital, Hunan, China; Key Laboratory of Translational Radiation Oncology, Hunan Province, The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University/Hunan Cancer Hospital, Changsha, Hunan, China.
| | - Feng Liu
- Department of Radiation Oncology, The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University/Hunan Cancer Hospital, Hunan, China.
| |
Collapse
|
5
|
Maddula M, McNamee N, Gan HK, Satgunaseelan L, Koh ES, Han CH, Thavaneswaran S. Contemporary Patterns of Care for Low-Grade Glioma in Australia and New Zealand. Curr Oncol 2025; 32:183. [PMID: 40136387 PMCID: PMC11941661 DOI: 10.3390/curroncol32030183] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2025] [Revised: 03/14/2025] [Accepted: 03/17/2025] [Indexed: 03/27/2025] Open
Abstract
AIM The management of low-grade gliomas (LGGs) is evolving with new insights into disease biology. Furthermore, recently, the phase III INDIGO1 study highlighted the benefits of an IDH inhibitor, vorasidenib, in treating residual or recurrent grade 2 IDH-mutant gliomas following surgery alone. We aimed to characterise the current patterns of care for patients with LGGs in Australia and New Zealand, including the role of vorasidenib. METHODS An online survey examining respondents' practice setting, caseload, and preferred treatment approach to three clinical scenarios was distributed through the Cooperative Trials Group for Neuro-Oncology, New Zealand Aotearoa Neuro-Oncology Society, and the Australian and New Zealand Society for Neuropathology in December 2023 with three reminders in April, June, and September of 2024. RESULTS The survey response rate was 19.6% (57/291), 87.7% from Australia, and 12.3% from New Zealand, spanning medical oncology (45.7%), pathology (22.8%), radiation oncology (17.5%), and neurosurgery (14.0%). Case 1 examined an IDH-mutant grade 2 astrocytoma following gross total resection. Observation alone was recommended by 93%. Case 2 examined an incompletely resected IDH-mutant grade 2 astrocytoma. If feasible, 38% recommended further surgery and 83% adjuvant chemotherapy and radiotherapy. After 12 months of disease stability, 53% of the respondents preferred vorasidenib over the existing therapies. Case 3 examined an incompletely resected IDH-mutant grade 3 oligodendroglioma. No respondents recommended observation alone, with 26% recommending salvage surgery and 97% recommending further chemotherapy and radiotherapy. CONCLUSIONS This study describes current management practices for LGGs in Australia and New Zealand, showing ongoing variation and a cautious approach to integrating IDH inhibitors. This highlights the critical role of multidisciplinary team-based decision-making in increasingly complex clinical situations.
Collapse
Affiliation(s)
- Meghana Maddula
- The Kinghorn Cancer Centre, St. Vincent’s Hospital, 370 Victoria St., Darlinghurst, NSW 2010, Australia;
- Faculty of Medicine and Health, University of New South Wales, Sydney, NSW 2052, Australia
| | - Nicholas McNamee
- The Kinghorn Cancer Centre, St. Vincent’s Hospital, 370 Victoria St., Darlinghurst, NSW 2010, Australia;
| | - Hui K. Gan
- Austin Health, Austin Hospital, Heidelberg, VIC 3084, Australia;
- Cancer Therapies and Biology Group, Centre of Research Excellence in Brain Tumours, Olivia Newton-John Cancer Wellness and Research Centre, Austin Hospital, Heidelberg, VIC 3084, Australia
- School of Cancer Medicine, La Trobe University, Heidelberg, VIC 3079, Australia
| | - Laveniya Satgunaseelan
- Department of Neuropathology, Royal Prince Alfred Hospital, Sydney, NSW 2050, Australia;
| | - Eng-Siew Koh
- Department of Radiation Oncology, Liverpool Hospital, Sydney, NSW 2170, Australia;
- School of Clinical Medicine, South West Sydney Clinical Campuses, Faculty of Medicine and Health, University of New South Wales, Liverpool, NSW 2170, Australia
| | - Catherine H. Han
- Auckland Oncology & Auckland City Hospital, Auckland 1023, New Zealand;
- Department of Pharmacology and Clinical Pharmacology, Faculty of Medical and Health Sciences University of Auckland, Auckland 1023, New Zealand
| | - Subotheni Thavaneswaran
- The Kinghorn Cancer Centre, St. Vincent’s Hospital, 370 Victoria St., Darlinghurst, NSW 2010, Australia;
- Faculty of Medicine and Health, University of New South Wales, Sydney, NSW 2052, Australia
- NHMRC Clinical Trials Centre, University of Sydney, Sydney, NSW 2050, Australia
| |
Collapse
|
6
|
Shi Z, Li KK, Kwan JS, Chung NY, Wong S, Chu AW, Chen H, Chan DT, Mao Y, Ng H. The molecular history of IDH-mutant astrocytomas without adjuvant treatment. Brain Pathol 2025; 35:e13300. [PMID: 39473241 PMCID: PMC11835445 DOI: 10.1111/bpa.13300] [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: 01/23/2024] [Accepted: 08/05/2024] [Indexed: 02/20/2025] Open
Abstract
Hypermutation and malignant transformation are potential complications arising from temozolomide treatment of IDH-mutant gliomas. However, the natural history of IDH-mutant low-grade gliomas without temozolomide treatment is actually under-studied. We retrieved retrospectively from our hospitals paired tumors from 19 patients with IDH-mutant, 1p19q non-codeleted Grade 2 astrocytomas where no interim adjuvant treatment with either temozolomide or radiotherapy was given between primary resections and first recurrences. Tissues from multiple recurrences were available from two patients and radiotherapy but not temozolomide was given before the last specimens were resected. We studied the natural molecular history of these low-grade IDH-mutant astrocytomas without pressure of temozolomide with DNA methylation profiling and copy number variation (CNV) analyses, targeted DNA sequencing, TERTp sequencing, FISH for ALT and selected biomarkers. Recurrences were mostly higher grades (15/19 patients) and characterized by new CNVs not present in the primary tumors (17/19 cases). Few novel mutations were identified in recurrences. Tumors from 17/19 (89.5%) patients showed either CDKN2A homozygous deletion, MYC or PDGFRA focal and non-focal gains at recurrences. There was no case of hypermutation. Phylogenetic trees constructed for tumors for the two patients with multiple recurrences suggested a lack of subclone development in their evolution when under no pressure from temozolomide. In summary, our studies demonstrated, in contrast to the phenomenon of temozolomide-induced hypermutation, IDH-mutant, 1p19q non-codeleted Grade 2 astrocytomas which had not been treated by temozolomide, acquired new CNVs at tumor recurrences. These findings improve our understanding of the molecular life history of IDH-mutant astrocytomas.
Collapse
Affiliation(s)
- Zhi‐Feng Shi
- Department of NeurosurgeryHuashan Hospital, Fudan UniversityShanghaiChina
- Hong Kong and Shanghai Brain Consortium (HSBC)Hong KongChina
| | - Kay Ka‐Wai Li
- Department of Anatomical and Cellular PathologyThe Chinese University of Hong KongHong KongChina
| | - Johnny Sheung‐Him Kwan
- Department of Anatomical and Cellular PathologyThe Chinese University of Hong KongHong KongChina
- Hong Kong Genome InstituteHong Kong Science ParkShatin, Hong KongChina
| | - Nellie Yuk‐Fei Chung
- Department of Anatomical and Cellular PathologyThe Chinese University of Hong KongHong KongChina
| | - Sze‐Ching Wong
- Department of Anatomical and Cellular PathologyThe Chinese University of Hong KongHong KongChina
| | - Abby Wai‐Yan Chu
- Department of Anatomical and Cellular PathologyThe Chinese University of Hong KongHong KongChina
| | - Hong Chen
- Department of Pathology, Huashan HospitalFudan UniversityShanghaiChina
| | - Danny Tat‐Ming Chan
- Division of Neurosurgery, Department of SurgeryThe Chinese University of Hong KongShatin, Hong KongChina
| | - Ying Mao
- Department of NeurosurgeryHuashan Hospital, Fudan UniversityShanghaiChina
- Hong Kong and Shanghai Brain Consortium (HSBC)Hong KongChina
| | - Ho‐Keung Ng
- Hong Kong and Shanghai Brain Consortium (HSBC)Hong KongChina
- Department of Anatomical and Cellular PathologyThe Chinese University of Hong KongHong KongChina
| |
Collapse
|
7
|
Roth P, Capper D, Calabrese E, Halasz LM, Jakola AS. Role of the tumor board when prescribing mutant isocitrate dehydrogenase inhibitors to patients with isocitrate dehydrogenase-mutant glioma. Neurooncol Pract 2025; 12:i29-i37. [PMID: 39776528 PMCID: PMC11703365 DOI: 10.1093/nop/npae100] [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: 01/11/2025] Open
Abstract
Isocitrate dehydrogenase (IDH)-mutant gliomas, comprising both astrocytomas and oligodendrogliomas, represent a distinct group of tumors that pose an interdisciplinary challenge. Addressing the needs of affected patients requires close collaboration among various disciplines, including neuropathology, neuroradiology, neurosurgery, radiation oncology, neurology, medical oncology, and other relevant specialties when necessary. Interdisciplinary tumor boards are central in determining the ideal diagnostic and therapeutic strategies for these patients. The key tasks of a tumor board include the evaluation of imaging findings, selecting the appropriate surgical approach, discussing additional treatment options, and identification/determination of tumor recurrence and progression. In addition to established treatments such as radiotherapy and alkylating chemotherapy, patients with an isocitrate dehydrogenase (IDH)-mutant glioma for whom additional treatment is indicated may now also have the option of receiving treatment with an mutant isocitrate dehydrogenase inhibitor such as vorasidenib or ivosidenib. In this regard, the collaborative nature of tumor boards becomes even more crucial for evaluating comprehensively the needs of these patients. Through interdisciplinary discussions, tumor boards aim to develop personalized treatment strategies that maximize therapeutic efficacy while minimizing potential side effects and preserving patients' quality of life.
Collapse
Affiliation(s)
- Patrick Roth
- University of Zurich, Zurich, Switzerland
- Department of Neurology and Brain Tumor Center, University Hospital Zurich, Zurich, Switzerland
| | - David Capper
- German Cancer Consortium (DKTK), Partner Site Berlin, German Cancer Research Center (DKFZ), Heidelberg, Germany
- Department of Neuropathology, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Evan Calabrese
- Department of Radiology, Division of Neuroradiology, Duke University Medical Center, Durham, North Carolina
| | - Lia M Halasz
- Department of Radiation Oncology, University of Washington
| | - Asgeir S Jakola
- Institute of Neuroscience and Physiology, Section of Clinical Neuroscience, Sahlgrenska Academy, Gothenburg, Sweden
- Department of Neurosurgery, Sahlgrenska University Hospital, Gothenburg, Sweden
| |
Collapse
|
8
|
Himstead AS, Chen JW, Chu E, Perez-Rosendahl MA, Zheng M, Mathew S, Yuen CA. Expanded Use of Vorasidenib in Non-Enhancing Recurrent CNS WHO Grade 3 Oligodendroglioma. Biomedicines 2025; 13:201. [PMID: 39857783 PMCID: PMC11762706 DOI: 10.3390/biomedicines13010201] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2024] [Revised: 12/31/2024] [Accepted: 01/13/2025] [Indexed: 01/27/2025] Open
Abstract
Background/Objectives: Anaplastic oligodendrogliomas (AOs) are central nervous system (CNS) World Health Organization (WHO) grade 3 gliomas characterized by isocitrate dehydrogenase (IDH) mutation (m)IDH and 1p/19q codeletion. AOs are typically treated with surgery and chemoradiation. However, chemoradiation can cause detrimental late neurocognitive morbidities and an accelerated disease course. The recently regulatory-approved vorasidenib, a brain-penetrating oral inhibitor of IDH1/2, has altered the treatment paradigm for recurrent/residual non-enhancing surgically resected CNS WHO grade 2 mIDH gliomas. Though vorasidenib can delay the time to chemoradiation for grade 2 gliomas, the implications for vorasidenib in non-grade 2 mIDH gliomas are not well understood. Results: We present a case of a 71-year-old male with a grade 3 non-enhancing oligodendroglioma successfully treated with vorasidenib with an 11% reduction in residual tumor volume. Vorasidenib was well tolerated in our patient with a mild elevation in his liver transaminases that resolved following a brief interruption in treatment. Conclusions: Our case suggests that vorasidenib may impart therapeutic benefits in this setting. This case illustrates the need for further investigation into these less commonly addressed scenarios and treatment strategies that extend beyond current guidelines.
Collapse
Affiliation(s)
- Alexander S. Himstead
- Department of Neurological Surgery, University of California, Irvine, CA 92697, USA; (A.S.H.); (J.W.C.)
| | - Jefferson W. Chen
- Department of Neurological Surgery, University of California, Irvine, CA 92697, USA; (A.S.H.); (J.W.C.)
| | - Eleanor Chu
- Department of Radiological Sciences, University of California, Irvine, CA 92697, USA;
| | - Mari A. Perez-Rosendahl
- Department of Pathology & Laboratory Medicine, University of California, Irvine, CA 92697, USA;
| | - Michelle Zheng
- UC Irvine Charlie Dunlop School of Biological Sciences, University of California, Irvine, CA 92697, USA
| | - Sherin Mathew
- Department of Research, University of California, Irvine, CA 92697, USA
| | - Carlen A. Yuen
- Department of Neurology, Division of Neuro-Oncology, University of California, Irvine, CA 92697, USA
| |
Collapse
|
9
|
Diaz M, Pan PC. Management of Low-Grade Gliomas. Cancer J 2025; 31:e0760. [PMID: 39841424 PMCID: PMC11801446 DOI: 10.1097/ppo.0000000000000760] [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: 01/23/2025]
Abstract
ABSTRACT The term "low-grade glioma" historically refers to adult diffuse gliomas that exhibit a less aggressive course than the more common high-grade gliomas. In the current molecular era, "low-grade" refers to World Health Organization central nervous system grade 2 gliomas almost always with an isocitrate dehydrogenase (IDH) mutation (astrocytomas and oligodendrogliomas). The term "lower-grade gliomas" has emerged encompassing grades 2 and 3 IDH-mutant astrocytomas and oligodendrogliomas, to acknowledge that histological grade is not as important a prognostic factor as molecular features, and distinguishing them from grade 4 glioblastomas, which lack an IDH mutation. These grades 2 and 3 IDH-mutant tumors are characterized by indolent growth but are ultimately incurable in most cases, presenting significant management challenges. Physicians must carefully weigh all available evidence to balance improvements in survival from new treatments against treatment toxicities. This review summarizes the evidence guiding the treatment of these patients.
Collapse
|
10
|
Fleming JL, Chakravarti A. Recent Advancements and Future Perspectives on Molecular Biomarkers in Adult Lower-Grade Gliomas. Cancer J 2025; 31:e0758. [PMID: 39841423 DOI: 10.1097/ppo.0000000000000758] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2025]
Abstract
ABSTRACT There has been a significant paradigm shift in the clinical management of lower-grade glioma patients given the recent updates to the 2021 World Health Organization classification along with long-term results from randomized phase III clinical trials. As a result, we are now better able to diagnose and assign patients to the most appropriate treatment course. This review provides a comprehensive summary of the most robust and reliable molecular biomarkers for adult lower-grade gliomas and discusses current challenges facing this patient population that future correlative biology studies combined with advancements in technologies could help overcome.
Collapse
Affiliation(s)
- Jessica L Fleming
- From the Department of Radiation Oncology, Ohio State University Comprehensive Cancer Center, Columbus, OH
| | | |
Collapse
|
11
|
Ziu M, Halasz LM, Kumthekar PU, McGranahan TM, Lo SS, Olson JJ. Congress of Neurological Surgeons systematic review and evidence-based guidelines for the role of chemotherapy in newly diagnosed WHO Grade II diffuse glioma in adults: update. J Neurooncol 2025; 171:279-298. [PMID: 39565459 DOI: 10.1007/s11060-024-04861-6] [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: 08/21/2024] [Accepted: 10/18/2024] [Indexed: 11/21/2024]
Abstract
Questions and recommendations from the prior version of these guidelines without changeTarget populationAdult patients (older than 18 years of age) with newly diagnosed World Health Organization (WHO) Grade II gliomas (Oligodendroglioma, astrocytoma, mixed oligoastrocytoma).QuestionIs there a role for chemotherapy as adjuvant therapy of choice in treatment of patients with newly diagnosed low-grade gliomas?RecommendationLevel III: Chemotherapy is recommended as a treatment option to postpone the use of radiotherapy, to slow tumor growth and to improve progression free survival (PFS), overall survival (OS) and clinical symptoms in adult patients with newly diagnosed LGG.QuestionWho are the patients with newly diagnosed LGG that would benefit the most from chemotherapy?RecommendationLevel III: Chemotherapy is recommended as an optional component alone or in combination with radiation as the initial adjuvant therapy for all patients who cannot undergo gross total resection (GTR) of a newly diagnosed LGG. Patients with residual tumor >1 cm on post-operative MRI, presenting diameter of 4 cm or older than 40 years of age should be considered for adjuvant therapy as well.QuestionAre there tumor markers that can predict which patients can benefit the most from initial treatment with chemotherapy?RecommendationLevel III: The addition of chemotherapy to standard RT is recommended in LGG patients that carry IDH mutation. In addition, temozolomide (TMZ) is recommended as a treatment option to slow tumor growth in patients who harbor the 1p/19q co-deletion.QuestionHow soon should the chemotherapy be started once the diagnosis of LGG is confirmed?RecommendationThere is insufficient evidence to make a definitive recommendation on the timing of starting chemotherapy after surgical/pathological diagnosis of LGG has been made. However, using the 12 weeks mark as the latest timeframe to start adjuvant chemotherapy is suggested. It is recommended that patients be enrolled in properly designed clinical trials to assess the timing of chemotherapy initiation once diagnosis is confirmed for this target population.QuestionWhat chemotherapeutic agents should be used for treatment of newly diagnosed LGG?RecommendationThere is insufficient evidence to make a recommendation of one particular regimen. Enrollment of subjects in properly designed trials comparing the efficacy of these or other agents is recommended so as to determine which of these regimens is superior.QuestionWhat is the optimal duration and dosing of chemotherapy as initial treatment for LGG?RecommendationInsufficient evidence exists regarding the duration of any specific cytotoxic drug regimen for treatment of newly diagnosed LGG. Enrollment of subjects in properly designed clinical investigations assessing the optimal duration of this therapy is recommended.QuestionShould chemotherapy be given alone or in conjunction with RT as initial therapy for LGG?RecommendationInsufficient evidence exists to make recommendations in this regard. Hence, enrollment of patients in properly designed clinical trials assessing the difference between chemotherapy alone, RT alone or a combination of them is recommended.QuestionShould chemotherapy be given in addition to other type of adjuvant therapy to patients with newly diagnosed LGG?RecommendationLevel II: It is recommended that chemotherapy be added to the RT in patients with unfavorable LGG to improve their progression free survival.Updated Question and Recommendations from the Prior Version of These GuidelinesQuestionIn adult patients with pathologically confirmed WHO Grade II diffuse glioma does chemotherapy alone, combined with radiation therapy or after radiation therapy compared to radiotherapy alone result in better overall survival, progression free survival, local control, fewer complications, neurocognitive preservation, and quality of life?RecommendationLevel I: It is recommended that chemotherapy (PCV) be added to radiation therapy (RT) in all patients with newly diagnosed high-risk WHO Grade II diffuse glioma (Patients younger than 40 unable to get gross total resection and older than 40 regardless of the degree of resection) to improve their overall survival. LEVEL II It is recommended that chemotherapy be added to radiation therapy in all patients with newly diagnosed high-risk WHO Grade II diffuse glioma to improve overall survival without a decline in neurocognitive function. LEVEL III It is suggested that chemotherapy (temozolomide) be added to RT in all patients with newly diagnosed high-risk WHO Grade II diffuse glioma to improve progression free survival and overall survival. LEVEL III It is suggested that chemotherapy alone should be considered in patients with newly diagnosed WHO Grade II diffuse glioma in cases with 1p/19q co-deletion.New questions and recommendationsTarget populationThese recommendations apply to adult patients diagnosed with WHO Grade II diffuse glioma.QuestionIn adult patients with newly diagnosed WHO grade II diffuse glioma does administration of chemotherapy prior to surgical resection improve extent of resection, provide longer progression free survival and overall survival when compared to chemotherapy alone?RecommendationLevel III: Neo-adjuvant temozolomide may be used in patients with WHO Grade II diffuse gliomas deemed unsafe for resection due to infiltration of eloquent areas or with large contralateral extension as an initial step to improve the extent of resection.There is insufficient evidence to support a recommendation regarding the ability of chemotherapy provided prior to surgical resection to improve progression free survival (PFS) and overall survival (OS).QuestionIn adult patients with newly diagnosed WHO grade II diffuse glioma does the administration of temozolomide increase the rate of malignant transformation when compared to no chemotherapy or other chemotherapy regimens?RecommendationThere is insufficient evidence to support a recommendation against the use of temozolomide for WHO Grade II diffuse gliomas due to concern over increasing the rate of malignant transformation.QuestionIn adult patients with newly diagnosed WHO grade II diffuse glioma does administration of multi-agent chemotherapy improve progression free survival and overall survival when compared to administration of single-agent chemotherapy?RecommendationThere is insufficient evidence to support a recommendation for or against the use of multi-agent chemotherapy to improve progression free survival and overall survival when compared to administration of single-agent chemotherapy in patients with newly diagnosed WHO Grade II diffuse glioma.
Collapse
Affiliation(s)
- Mateo Ziu
- Department of Neurosurgery, Inova Neurosciences, Inova Health System, Fairfax, VA, USA.
| | - Lia M Halasz
- Departments of Radiation Oncology and Neurological Surgery, University of Washington/ Fred Hutch Cancer Center, Seatle, WA, USA
| | - Priya U Kumthekar
- Department of Neurology, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Tresa M McGranahan
- Division of Hematology and Oncology, Scripps Cancer Center, San Diego, CA, USA
| | - Simon S Lo
- Department of Radiation Oncology and Neurological Surgery, University of Washington/Fred Hutch Cancer Center, Seattle, WA, USA
| | - Jeffrey J Olson
- Department of Neurosurgery, Emory University School of Medicine, Atlanta, GA, USA
| |
Collapse
|
12
|
Darlix A, Bady P, Deverdun J, Lefort K, Rigau V, Le Bars E, Meriadec J, Carrière M, Coget A, Santarius T, Matys T, Duffau H, Hegi ME. Clinical value of the MGMT promoter methylation score in IDHmt low-grade glioma for predicting benefit from temozolomide treatment. Neurooncol Adv 2025; 7:vdae224. [PMID: 40041202 PMCID: PMC11877643 DOI: 10.1093/noajnl/vdae224] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/06/2025] Open
Abstract
Background Diffuse IDH mutant low-grade gliomas (IDHmt LGG) (World Health Organization grade 2) typically affect young adults. The outcome is variable, with survival ranging from 5 to over 20 years. The timing and choice of initial treatments after surgery remain controversial. In particular, radiotherapy is associated with early and late cognitive toxicity. Over 90% of IDHmt LGG exhibit some degree of promoter methylation of the repair gene O(6)-methylguanine-DNA methytransferase (MGMTp) that when expressed blunts the effect of alkylating agent chemotherapy, for example, temozolomide (TMZ). However, the clinical value of MGMTp methylation predicting benefit from TMZ in IDHmt LGG is unclear. Methods Patients treated in the EORTC-22033 phase III trial comparing TMZ versus radiotherapy served as training set to establish a cutoff based on the MGMT-STP27 methylation score. A validation cohort was established with patients treated in a single-center first-line with TMZ after surgery/surgeries. Results The MGMT-STP27 methylation score was associated with better progression-free survival (PFS) in the training cohort treated with TMZ, but not radiotherapy. In the validation cohort, an association with next treatment-free survival (P = .045) after TMZ was observed, and a trend using RANO criteria (P = .07). A cutoff value set above the 95% confidence interval of being methylated was significantly associated with PFS in the TMZ-treated training cohort, but not in the radiotherapy arm. However, this cutoff could not be confirmed in the test cohort. Conclusions While the MGMTp methylation score was associated with better outcomes in TMZ-treated IDHmt LGG, a cutoff could not be established to guide treatment decisions.
Collapse
Affiliation(s)
- Amélie Darlix
- Institute of Functional Genomics IGF, University of Montpellier, CNRS, INSERM, Montpellier, France
- Department of Medical Oncology, Institut Régional du Cancer de Montpellier, University of Montpellier, Montpellier, France
| | - Pierre Bady
- SIB Swiss Institute of Bioinformatics, Lausanne, Switzerland
- Translational Data Science & Biomedical Data Science Center, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
- Neuroscience Research Center and Service of Neurosurgery, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
| | - Jérémy Deverdun
- Department of Neuroradiology, Gui de Chauliac Hospital, Montpellier University Medical Center, Montpellier, France
- Department of Neuroradiology, I2FH, Institut d’Imagerie Fonctionnelle Humaine, Montpellier University Medical Center, Montpellier, France
| | - Karine Lefort
- Department of Laboratory Medicine and Pathology, Institute of Pathology, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
| | - Valérie Rigau
- Department of Neuropathology, Gui de Chauliac Hospital, Montpellier University Medical Center, Montpellier, France
- Institute of Functional Genomics IGF, University of Montpellier, CNRS, INSERM, Montpellier, France
| | - Emmanuelle Le Bars
- Department of Neuroradiology, Gui de Chauliac Hospital, Montpellier University Medical Center, Montpellier, France
- Department of Neuroradiology, I2FH, Institut d’Imagerie Fonctionnelle Humaine, Montpellier University Medical Center, Montpellier, France
| | - Justine Meriadec
- Department of Neuroradiology, Gui de Chauliac Hospital, Montpellier University Medical Center, Montpellier, France
| | - Mathilde Carrière
- Department of Neuroradiology, Gui de Chauliac Hospital, Montpellier University Medical Center, Montpellier, France
| | - Arthur Coget
- Department of Neuroradiology, Gui de Chauliac Hospital, Montpellier University Medical Center, Montpellier, France
| | - Thomas Santarius
- Department of Neurosurgery, Cambridge University Hospitals NHS Foundation Trust, Cambridge, UK
| | - Tomasz Matys
- Department of Radiology, University of Cambridge and Cambridge University Hospitals NHS Foundation Trust, Cambridge, UK
| | - Hugues Duffau
- Department of Neurosurgery, Gui de Chauliac Hospital, Montpellier University Medical Center, Montpellier, France
- Institute of Functional Genomics IGF, University of Montpellier, CNRS, INSERM, Montpellier, France
| | - Monika E Hegi
- Departments of Oncology and Clinical Neurosciences, L. Lundin and Family Brain Tumor Research Center, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
- Neuroscience Research Center and Service of Neurosurgery, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
| |
Collapse
|
13
|
Cayuela N, Izquierdo C, Vaquero L, Càmara E, Bruna J, Simó M. Mapping glioma's impact on cognition: Insights from macrostructure, microstructure, and beyond. Neurooncol Adv 2025; 7:vdaf003. [PMID: 39911704 PMCID: PMC11795312 DOI: 10.1093/noajnl/vdaf003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2025] Open
Abstract
Background Cognitive impairment (CI) significantly impacts the quality of life of glioma patients. The main contributing risk factors include tumor characteristics, treatment-related factors, and their complex interplay. This review explores the role of advanced structural neuroimaging techniques in understanding CI in glioma patients. Methods A literature search was conducted in PubMed, PsycINFO, and ISI Web of Knowledge using specific keywords. We included studies with advanced magnetic resonance imaging techniques and objective neuropsychological exams. Results At diagnosis, during the pre-surgery phase, associations between glioma characteristics and cognitive outcomes have been described. Specifically, patients with isocitrate dehydrogenase (IDH)-wild-type gliomas exhibit more adverse cognitive outcomes, accompanied by disruptions in gray (GM) and white matter (WM) networks when compared to IDH-mutant. In addition, pre- and post-surgery imaging analyses highlight the importance of preserving specific WM tracts, such as the inferior longitudinal and arcuate fasciculus, in mitigating verbal memory and language processing decline. Furthermore, examining gliomas in perisylvian regions emphasizes deleterious effects on various cognitive domains. Additionally, it has been suggested that neuroplastic reorganization could serve as a compensatory mechanism against CI. Lastly, a limited number of studies suggest long-term CI linked to GM atrophy and leukoencephalopathy induced by radiotherapy ± chemotherapy in glioma survivors, highlighting the need for improving treatment approaches, particularly for patients with extended survival expectations. Conclusion This review underscores the need for nuanced understanding and an individual approach in the management of glioma patients. Neuroplastic insights offer clinicians valuable guidance in surgical decision-making and personalized therapeutic approaches thus improving patient outcomes in neuro-oncology.
Collapse
Affiliation(s)
- Nuria Cayuela
- Neurology Department, Complex Hospitalari Moisès Broggi, Barcelona, Spain
| | - Cristina Izquierdo
- Department of Neuroscience, Hospital Universitari Germans Trias i Pujol, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Lucía Vaquero
- Music and Audio Research Lab (MARL), New York University, New York, USA
- Center for Language Music and Emotion (CLaME) – Max Plank Institute of Empirical Aesthetics, New York University, New York, USA
- Research Group in Digital Culture and Social Movements (Cibersomosaguas), and Department of Experimental Psychology, Cognitive Processes and Speech Therapy, Complutense University of Madrid, Madrid, Spain
| | - Estela Càmara
- Cognition and Brain Plasticity Group, IDIBELL, Barcelona, Spain
| | - Jordi Bruna
- Neuro-Oncology Unit, Hospital Universitari de Bellvitge-ICO l’Hospitalet, IDIBELL (Oncobell Program), Barcelona, Spain
| | - Marta Simó
- Neuro-Oncology Unit, Hospital Universitari de Bellvitge-ICO l’Hospitalet, IDIBELL (Oncobell Program), Barcelona, Spain
- Cognition and Brain Plasticity Group, IDIBELL, Barcelona, Spain
| |
Collapse
|
14
|
Baumert BG, P M Jaspers J, Keil VC, Galldiks N, Izycka-Swieszewska E, Timmermann B, Grosu AL, Minniti G, Ricardi U, Dhermain F, Weber DC, van den Bent M, Rudà R, Niyazi M, Erridge S. ESTRO-EANO guideline on target delineation and radiotherapy for IDH-mutant WHO CNS grade 2 and 3 diffuse glioma. Radiother Oncol 2025; 202:110594. [PMID: 39454886 DOI: 10.1016/j.radonc.2024.110594] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2024] [Accepted: 10/12/2024] [Indexed: 10/28/2024]
Abstract
PURPOSE This guideline will discuss radiotherapeutic management of IDH-mutant grade 2 and grade 3 diffuse glioma, using the latest 2021 WHO (5th) classification of brain tumours focusing on: imaging modalities, tumour volume delineation, irradiation dose and fractionation. METHODS The ESTRO Guidelines Committee, CNS subgroup, nominated 15 European experts who identified questions for this guideline. Four working groups were established addressing specific questions concerning imaging, target volume delineation, radiation techniques and fractionation. A literature search was performed, and available literature was discussed. A modified two-step Delphi process was used with majority voting resulted in a decision or highlighting areas of uncertainty. RESULTS Key issues identified and discussed included imaging needed to define target definition, target delineation and the size of margins, and technical aspects of treatment including different planning techniques such as proton therapy. CONCLUSIONS The GTV should include any residual tumour volume after surgery, as well as the resection cavity. Enhancing lesions on T1 imaging should be included if they are indicative of residual tumour. In grade 2 tumours, T2/FLAIR abnormalities should be included in the GTV. In grade 3 tumours, T2/FLAIR abnormalities should also be included, except areas that are considered to be oedema which should be omitted from the GTV. A GTV to CTV expansion of 10 mm is recommended in grade 2 tumours and 15 mm in grade 3 tumours. A treatment dose of 50.4 Gy in 28 fractions is recommended in grade 2 tumours and 59.4 Gy in 33 fractions in grade 3 tumours. Radiation techniques with IMRT are the preferred approach.
Collapse
Affiliation(s)
- Brigitta G Baumert
- Institute of Radiation-Oncology, Cantonal Hospital Graubunden, Chur, Switzerland.
| | - Jaap P M Jaspers
- Department of Radiation Oncology, Radboud University Medical Center, Nijmegen, Netherlands
| | - Vera C Keil
- Department of Radiology and Nuclear Medicine, Amsterdam University Medical Center, Amsterdam, Netherlands
| | - Norbert Galldiks
- Department of Neurology, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany; Institute of Neuroscience and Medicine (IMN-3), Research Center Juelich, Juelich, Germany; Center for Integrated Oncology Aachen Bonn Cologne Duesseldorf (CIO ABCD), Germany
| | - Ewa Izycka-Swieszewska
- Department of Pathology and Neuropathology, Medical University of Gdansk, Gdansk, Poland
| | - Beate Timmermann
- West German Proton Therapy Centre Essen (WPE), University Hospital Essen, Essen, Germany; Department of Particle Therapy, University Hospital Essen, Essen, Germany; West German Cancer Centre (WTZ), German Cancer Consortium (DKTK), Essen, Germany
| | - Anca L Grosu
- Department of Radiation Oncology, University Medical Center, Medical Faculty, University of Freiburg, Freiburg, Germany
| | - Giuseppe Minniti
- Radiation Oncology Unit, Department of Radiological Sciences, Sapienza University of Rome, Rome, Italy
| | | | - Frédéric Dhermain
- Radiation Oncology Department, Gustave Roussy University Hospital, Villejuif, France
| | - Damien C Weber
- Center for Proton Therapy, Paul Scherrer Institute, Villigen PSI, Villingen, Switzerland
| | - Martin van den Bent
- The Brain Tumor Center at Erasmus MC Cancer Institute, University Medical Center Rotterdam, Rotterdam, Netherlands
| | - Roberta Rudà
- Division of Neuro-Oncology, Department of Neuroscience, University of Turin, Turin, Italy
| | - Maximilian Niyazi
- Center for Neuro-Oncology, Comprehensive Cancer Center Tübingen-Stuttgart, University Hospital Tübingen, Tübingen, Germany; Department of Radiation Oncology, University Hospital Tübingen, Tübingen, Germany
| | - Sara Erridge
- Edinburgh Cancer Centre, Western General Hospital, Edinburgh, UK; Institute of Genetics and Cancer, University of Edinburgh, Edinburgh, UK
| |
Collapse
|
15
|
Gardner MM, Winter SF, Stahl F, Gerstner ER, Shih HA, Sherman J, Dietrich J, Parsons MW. Brain volume loss after cranial irradiation: a controlled comparison study between photon vs proton radiotherapy for WHO grade 2-3 gliomas. J Neurooncol 2025; 171:351-363. [PMID: 39400662 DOI: 10.1007/s11060-024-04850-9] [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: 08/25/2024] [Accepted: 10/01/2024] [Indexed: 10/15/2024]
Abstract
PURPOSE Radiation therapy (RT) is an integral treatment component in patients with glioma but associated with neurotoxicity. Proton RT (PRT), as compared with photon RT (XRT), reduces excess radiation to nontarget tissue. We used a retrospective method to evaluate brain imaging metrics of neurotoxicity after treatment with PRT and XRT for glioma. METHODS We analyzed brain volume change in thirty-four patients with WHO grade 2-3 gliomas treated with either PRT (n = 17) or XRT (n = 17). Both groups were carefully matched by demographic/clinical criteria and assessed longitudinally for two years post-radiotherapy. Brain volume change was measured as ventricular volume expansion in the tumor free hemisphere (contralateral to RT target) as a proxy indicator of brain volume loss. We further assessed the impact of volumetric changes on cognition in PRT patients, who completed neuropsychological testing as part of an outcome study. RESULTS We found significant ventricular volume increases in the contralesional hemisphere in both groups at two years post-RT (F(1, 31) = 18.45, p < 0.000, partial η2 = 0.373), with greater volume change observed in XRT (26.55%) vs. PRT (12.03%) (M = 12.03%, SD = 16.26; F(1,31) = 4.26, p = 0.048, partial η2 = 0.121). Although, there was no group-level change on any cognitive test in PRT treated patients, individual changes on cognitive screening, working memory, processing speed and visual memory tasks correlated with contralesional brain volume loss. CONCLUSION This study suggests progressive brain volume loss following cranial irradiation, with greater severity after XRT vs. PRT. Radiation-induced brain volume loss appears to be associated with measurable cognitive changes on an individual level. Prospective studies are warranted to validate these findings and their impacts on long-term cognitive function and quality of life. An improved understanding of the structural and functional consequences of cranial radiation is essential to develop neuroprotective strategies.
Collapse
Affiliation(s)
- Melissa M Gardner
- Division of Neuro-Oncology, Mass General Cancer Center, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
- Department of Psychiatry, Psychology Assessment Center, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Sebastian F Winter
- Division of Neuro-Oncology, Mass General Cancer Center, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Franziska Stahl
- Department of Neurology, Schoen Clinic Munich Schwabing, Munich Schwabing, Germany
| | - Elizabeth R Gerstner
- Division of Neuro-Oncology, Mass General Cancer Center, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Helen A Shih
- Department of Radiation Oncology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Janet Sherman
- Department of Psychiatry, Psychology Assessment Center, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Jorg Dietrich
- Division of Neuro-Oncology, Mass General Cancer Center, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Michael W Parsons
- Division of Neuro-Oncology, Mass General Cancer Center, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA.
- Department of Psychiatry, Psychology Assessment Center, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
16
|
Kinslow CJ, Mehta MP. Future Directions in the Treatment of Low-Grade Gliomas. Cancer J 2025; 31:e0759. [PMID: 39841425 DOI: 10.1097/ppo.0000000000000759] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2025]
Abstract
ABSTRACT There is major interest in deintensifying therapy for isocitrate dehydrogenase-mutant low-grade gliomas, including with single-agent cytostatic isocitrate dehydrogenase inhibitors. These efforts need head-to-head comparisons with proven modalities, such as chemoradiotherapy. Ongoing clinical trials now group tumors by intrinsic molecular subtype, rather than classic clinical risk factors. Advances in imaging, surgery, and radiotherapy have improved outcomes in low-grade gliomas. Emerging biomarkers, targeted therapies, immunotherapy, radionuclides, and novel medical devices are a promising frontier for future treatment. Diverse representation in glioma research and clinical trials will help to ensure that advancements in care are realized by all groups.
Collapse
Affiliation(s)
| | - Minesh P Mehta
- Department of Radiation Oncology, Miami Cancer Institute, Baptist Health South Florida, Miami, FL
| |
Collapse
|
17
|
Wen K, Zhu W, Luo Z, Wang W. Machine learning-based identification of histone deacetylase-associated prognostic factors and prognostic modeling for low-grade glioma. Discov Oncol 2024; 15:824. [PMID: 39714729 DOI: 10.1007/s12672-024-01713-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/04/2024] [Accepted: 12/16/2024] [Indexed: 12/24/2024] Open
Abstract
BACKGROUND Low-grade glioma (LGG) is a slow-growing but invasive tumor that affects brain function. Histone deacetylases (HDACs) play a critical role in gene regulation and tumor progression. This study aims to develop a prognostic model based on HDAC-related genes to aid in risk stratification and predict therapeutic responses. METHODS Expression data from The Cancer Genome Atlas (TCGA) and Chinese Glioma Genome Atlas (CGGA) were analyzed to identify an optimal HDAC-related risk signature from 73 genes using 10 machine learning algorithms. Patients were stratified into high- and low-risk groups based on the median risk score. Prognostic accuracy was evaluated using Kaplan-Meier survival analysis and receiver operating characteristic (ROC) curves. Functional enrichment analyses, including Gene Ontology (GO), Kyoto Encyclopedia of Genes and Genomes (KEGG), and Gene Set Enrichment Analysis (GSEA), were performed to explore pathways linked to the gene signature. Immune infiltration and tumor microenvironment characteristics were assessed using Single Sample Gene Set Enrichment Analysis (ssGSEA) and ESTIMATE algorithm. SubMap was applied to predict responsiveness to immune checkpoint inhibitors, and chemotherapeutic sensitivity was analyzed via the Genomics of Drug Sensitivity in Cancer (GDSC) database. RESULTS A prognostic model consisting of four HDAC-related genes-SP140, BAZ1B, SP100, and SIRT1-was identified. This signature displayed strong prognostic accuracy, achieving a C-index of 0.945. Individuals with LGG were systematically divided into high-risk and low-risk cohorts based on the median risk value, enabling more precise risk stratification. The survival prognosis was significantly worse in the high-risk cohort compared to the low-risk group, highlighting distinct survival trajectories. Notably, the two cohorts exhibited marked shifts in immune checkpoint gene transcriptional profiles and immune cell infiltration maps, underscoring fundamental biological differences that contribute to these differing prognoses. CONCLUSION We developed an HDAC-related four-gene prognostic model that correlates with survival, immune landscape, and therapeutic response in LGG patients. This model may guide personalized treatment strategies and improve prognostic accuracy, warranting further validation in clinical settings.
Collapse
Affiliation(s)
- Keshan Wen
- Department of Radiation Oncology, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Weijie Zhu
- Department of Neurology, Shenzhen Longhua District Central Hospital, Shenzhen, 518110, China
| | - Ziyi Luo
- Department of Radiation Oncology, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Wei Wang
- Department of Radiation Oncology, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China.
| |
Collapse
|
18
|
Liang X, Li J, Lyu Y, Wang X. PIN4 could function as a prognostic biomarker for glioma. Asian J Surg 2024:S1015-9584(24)02609-5. [PMID: 39645496 DOI: 10.1016/j.asjsur.2024.11.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2024] [Accepted: 11/04/2024] [Indexed: 12/09/2024] Open
Affiliation(s)
- Xianyin Liang
- The Fifth Affiliated Hospital of Zhengzhou University, Henan, Zhengzhou, China; The Third Affiliated Hospital of Zhengzhou University, Medical Research Center, Henan, Zhengzhou, China; Tianjian Laboratory of Advanced Biomedical Sciences, School of Life Sciences, Zhengzhou University, Zhengzhou, Henan, China; Henan Engineering Research Center of Precise Diagnosis and Treatment of Pediatric Glioma, Henan, Zhengzhou, China; Henan International Joint Laboratory of Glioma Metabolism and Microenvironment Research, Zhengzhou, Henan, China; Maternal and Child Neurological Disorders International Joint Research Center, Zhengzhou, Henan, China; Zhengzhou University, Institute of Neuroscience, Zhengzhou, Henan, China; The Third Affiliated Hospital of Zhengzhou University, Department of Neurosurgery, Zhengzhou, Henan, China.
| | - Junqi Li
- The Third Affiliated Hospital of Zhengzhou University, Medical Research Center, Henan, Zhengzhou, China; Tianjian Laboratory of Advanced Biomedical Sciences, School of Life Sciences, Zhengzhou University, Zhengzhou, Henan, China; Henan Engineering Research Center of Precise Diagnosis and Treatment of Pediatric Glioma, Henan, Zhengzhou, China.
| | - Yuan Lyu
- Maternal and Child Neurological Disorders International Joint Research Center, Zhengzhou, Henan, China; Zhengzhou University, Institute of Neuroscience, Zhengzhou, Henan, China; The Third Affiliated Hospital of Zhengzhou University, Department of Neurosurgery, Zhengzhou, Henan, China.
| | - Xinjun Wang
- The Third Affiliated Hospital of Zhengzhou University, Medical Research Center, Henan, Zhengzhou, China; Tianjian Laboratory of Advanced Biomedical Sciences, School of Life Sciences, Zhengzhou University, Zhengzhou, Henan, China; Henan Engineering Research Center of Precise Diagnosis and Treatment of Pediatric Glioma, Henan, Zhengzhou, China; Henan International Joint Laboratory of Glioma Metabolism and Microenvironment Research, Zhengzhou, Henan, China; Maternal and Child Neurological Disorders International Joint Research Center, Zhengzhou, Henan, China; Zhengzhou University, Institute of Neuroscience, Zhengzhou, Henan, China; The Third Affiliated Hospital of Zhengzhou University, Department of Neurosurgery, Zhengzhou, Henan, China.
| |
Collapse
|
19
|
Szivos L, Virga J, Mészár Z, Rostás M, Bakó A, Zahuczki G, Hortobágyi T, Klekner Á. Prognostic Role of Invasion-Related Extracellular Matrix Molecules in Diffusely Infiltrating Grade 2 and 3 Astrocytomas. Brain Sci 2024; 14:1157. [PMID: 39595920 PMCID: PMC11592374 DOI: 10.3390/brainsci14111157] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2024] [Revised: 11/12/2024] [Accepted: 11/14/2024] [Indexed: 11/28/2024] Open
Abstract
BACKGROUND Astrocytoma, an IDH-mutant is a common primary brain tumor. Total surgical resection is not feasible due to peritumoral infiltration mediated by extracellular matrix (ECM) molecules. METHODS This study aimed at determining the expression pattern of ECM molecules in different prognostic groups of WHO grade 2 and grade 3 patients and identifying the effect of onco-radiotherapy on tumor cell invasion of grade 3 patients. Gene and protein expression of ECM molecules was determined by qRT-PCR and immunohistochemistry, respectively. RESULTS In the different prognostic groups of grade 2 tumors HMMR, IDH-1, MKI-67, PDGF-A and versican, in grade 3 tumors integrin α-3, and in both groups integrin α-3 and IDH-1 mRNA expression was significantly different. Regarding protein expression, only integrin αV expression changed significantly in the prognostic groups of grade 2 tumors. CONCLUSIONS Based on the invasion spectrum determined by this joint gene and protein expression analysis, there was a sensitivity of 87.5% and a negative predictive value of 88.9% regarding the different prognostic groups of grade 2 astrocytoma. For grade 3 tumors, the applied standard oncotherapeutic modalities apparently lacked significant anti-invasive effects.
Collapse
Affiliation(s)
- László Szivos
- Department of Neurosurgery, University of Debrecen, H-4032 Debrecen, Hungary or (L.S.); (J.V.)
- Department of Neurosurgery, University of Szeged, H-6725 Szeged, Hungary
| | - József Virga
- Department of Neurosurgery, University of Debrecen, H-4032 Debrecen, Hungary or (L.S.); (J.V.)
- Department of Oncology, University of Debrecen, H-4032 Debrecen, Hungary;
| | - Zoltán Mészár
- Department of Anatomy, Histology and Embryology, University of Debrecen, H-4032 Debrecen, Hungary;
| | - Melinda Rostás
- Department of Biochemistry and Molecular Biology, University of Debrecen, H-4032 Debrecen, Hungary;
| | - Andrea Bakó
- Department of Oncology, University of Debrecen, H-4032 Debrecen, Hungary;
| | - Gábor Zahuczki
- UD-GenoMed Medical Genomic Technologies Ltd., H-4032 Debrecen, Hungary;
| | - Tibor Hortobágyi
- Department of Neurology, University of Debrecen, H-4032 Debrecen, Hungary;
- Institute of Neuropathology, University Hospital Zurich, 8091 Zurich, Switzerland
| | - Álmos Klekner
- Department of Neurosurgery, University of Debrecen, H-4032 Debrecen, Hungary or (L.S.); (J.V.)
| |
Collapse
|
20
|
Hiong A, Lapidus AH, Gately L, Sinclair G, Ameratunga M. Herpes zoster in patients with glioma treated with temozolomide. J Clin Neurosci 2024; 129:110816. [PMID: 39222582 DOI: 10.1016/j.jocn.2024.110816] [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: 06/26/2024] [Revised: 08/16/2024] [Accepted: 08/25/2024] [Indexed: 09/04/2024]
Abstract
BACKGROUND The risk of herpes zoster in patients treated with temozolomide is poorly defined in the literature. We aimed to evaluate the incidence of and risk factors for herpes zoster in individuals receiving temozolomide for glioma. METHODS A retrospective observational study was conducted on a series of patients treated with temozolomide for glioma at a single centre between 1 October 2018 and 30 September 2023. RESULTS 131 patients were treated with temozolomide for glioma with a median age of 55 years. 4 out of 131 patients (3.1 %) developed herpes zoster during temozolomide treatment. All cases of herpes zoster occurred in patients who had lymphocyte nadirs of less than 0.7 x 109/L and were receiving corticosteroids concomitantly. The estimated herpes zoster incidence rates were 45.44 per 1000 person-years (95 % confidence interval (CI) 12.38-116.34 per 1000 person-years) in the overall study population and 224.97 per 1000 person-years (95 % CI 61.30-576.02 per 1000 person-years) in subjects who were treated with corticosteroids and had a lymphocyte nadir of less than 1.0 x 109/L. CONCLUSION Use of temozolomide, particularly in conjunction with lymphopaenia or corticosteroid use, poses a risk of herpes zoster. Further research into the benefits of prophylactic antiviral measures in this population is recommended.
Collapse
Affiliation(s)
- Alison Hiong
- Department of Oncology, Alfred Health, Melbourne, Victoria, Australia
| | - Adam H Lapidus
- Department of Oncology, Alfred Health, Melbourne, Victoria, Australia
| | - Lucy Gately
- Department of Oncology, Alfred Health, Melbourne, Victoria, Australia
| | - Grace Sinclair
- Department of Oncology, Alfred Health, Melbourne, Victoria, Australia
| | - Malaka Ameratunga
- Department of Oncology, Alfred Health, Melbourne, Victoria, Australia; School of Translational Medicine, Monash University, Melbourne, Victoria, Australia.
| |
Collapse
|
21
|
Ng S, Rigau V, Moritz-Gasser S, Gozé C, Darlix A, Herbet G, Duffau H. Long-term autonomy, professional activities, cognition, and overall survival after awake functional-based surgery in patients with IDH-mutant grade 2 gliomas: a retrospective cohort study. THE LANCET REGIONAL HEALTH. EUROPE 2024; 46:101078. [PMID: 39381547 PMCID: PMC11458993 DOI: 10.1016/j.lanepe.2024.101078] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 06/19/2024] [Revised: 09/04/2024] [Accepted: 09/09/2024] [Indexed: 10/10/2024]
Abstract
Background In isocitrate dehydrogenase (IDH)-mutant low-grade gliomas (LGGs), awake functional-based resection (i.e., resection based on intraoperative functional responses rather than anatomical margins) has emerged as an efficient method to reduce tumour volume (TV) while minimizing postoperative deficits. Here, our goal was to assess the long-term onco-functional outcomes after awake functional-based resection in IDH-mutant LGGs, in conjunction with clinico-radiological and molecular factors. Methods We retrospectively studied a consecutive cohort (June 1997-January 2023) of 949 patients. Six hundred patients with IDH-mutant LGGs benefited from an awake functional-based resection with a median follow-up of 7.8 years (95% Confidence interval [CI]: 7.1-8.4 years). The main outcomes were the overall survival (OS), the OS with Karnofsky performance status ≥80% (OSKPS ≥ 80%), cognition measures, and professional activities at 12 months post-surgery. Findings 600 patients were included in the cohort (274 female [46.0%], median age: 36 years [Interquartile range, IQR: 30-44 years]). The rate of return to work was 93.7%. The impact of surgery on cognition was of limited magnitude. The median postsurgical TV of 2.5 mL (IQR: 0-8.0 mL). The median OS was over 20 years (median: NA, 95% CI: 17.0-NA years). The median OSKPS ≥ 80% was 14.7 years (95% CI: 13.2-17.2 years). Factors associated with longer OS and OSKPS ≥ P80% were 1p19q codeletion (Hazard ratio [HR]OS: 0.27, 95% CI: 0.16-0.43, HRKPS ≥ 80%:0.25, 95% CI: 0.17-0.36), supratotal resection (HROS: 0.08, 95% CI: 0.005-0.40, HRKPS ≥ 80%:0.12, 95% CI: 0.03-0.34) and total resection (HROS: 0.31, 95% CI: 0.16-0.59, HRKPS ≥ 80%:0.21, 95% CI: 0.12-0.36). Recursive partitioning analyses established three OS and OSKPS ≥ 80% prognostic groups, highlighting the contributions of histomolecular status, extent of resection, postsurgical and presurgical TV. Further propensity-matching analyses confirmed the oncological benefits of supratotal resections. Interpretation Awake functional-based resection surgery in newly diagnosed IDH-mutant grade 2 LGG, was an effective strategy associated with long survival (median OS over 20 years) and long-term preservation of autonomy. More complete tumor resections favored better onco-functional outcomes across all molecularly-defined subtypes. Short-term effects were of limited magnitude regarding postoperative cognitive and professional outcomes. Supratotal functional-based resections offered additional survival benefits. Funding None.
Collapse
Affiliation(s)
- Sam Ng
- Department of Neurosurgery, Gui de Chauliac Hospital, Montpellier University Medical Center, 80 Av Augustin Fliche, 34295, Montpellier, France
- Institute of Functional Genomics, University of Montpellier, INSERM, CNRS, Team “Plasticity of Central Nervous System, Stem Cells and Glial Tumors,”, National Institute for Health and Medical Research (INSERM), U1191 Laboratory, 34091, Montpellier, France
| | - Valérie Rigau
- Institute of Functional Genomics, University of Montpellier, INSERM, CNRS, Team “Plasticity of Central Nervous System, Stem Cells and Glial Tumors,”, National Institute for Health and Medical Research (INSERM), U1191 Laboratory, 34091, Montpellier, France
- Department of Pathology and Onco-Biology, Gui de Chauliac Hospital, Montpellier University Medical Center, 80 Av Augustin Fliche, 34295, Montpellier, France
| | - Sylvie Moritz-Gasser
- Department of Neurosurgery, Gui de Chauliac Hospital, Montpellier University Medical Center, 80 Av Augustin Fliche, 34295, Montpellier, France
- Institute of Functional Genomics, University of Montpellier, INSERM, CNRS, Team “Plasticity of Central Nervous System, Stem Cells and Glial Tumors,”, National Institute for Health and Medical Research (INSERM), U1191 Laboratory, 34091, Montpellier, France
- University of Montpellier, 163 rue Broussonnet, 34000, Montpellier, France
| | - Catherine Gozé
- Institute of Functional Genomics, University of Montpellier, INSERM, CNRS, Team “Plasticity of Central Nervous System, Stem Cells and Glial Tumors,”, National Institute for Health and Medical Research (INSERM), U1191 Laboratory, 34091, Montpellier, France
- Department of Pathology and Onco-Biology, Gui de Chauliac Hospital, Montpellier University Medical Center, 80 Av Augustin Fliche, 34295, Montpellier, France
| | - Amélie Darlix
- Institute of Functional Genomics, University of Montpellier, INSERM, CNRS, Team “Plasticity of Central Nervous System, Stem Cells and Glial Tumors,”, National Institute for Health and Medical Research (INSERM), U1191 Laboratory, 34091, Montpellier, France
- Department of Medical Oncology, Montpellier Regional Cancer Institute, 34298, Montpellier, France
| | - Guillaume Herbet
- Department of Neurosurgery, Gui de Chauliac Hospital, Montpellier University Medical Center, 80 Av Augustin Fliche, 34295, Montpellier, France
- University of Montpellier, 163 rue Broussonnet, 34000, Montpellier, France
- Praxiling Laboratory, UMR 5267, CNRS, Paul Valéry – Montpellier 3 University, rue de Mende, 34090, Montpellier, France
- Institut Universitaire de France, Paris, France
| | - Hugues Duffau
- Department of Neurosurgery, Gui de Chauliac Hospital, Montpellier University Medical Center, 80 Av Augustin Fliche, 34295, Montpellier, France
- Institute of Functional Genomics, University of Montpellier, INSERM, CNRS, Team “Plasticity of Central Nervous System, Stem Cells and Glial Tumors,”, National Institute for Health and Medical Research (INSERM), U1191 Laboratory, 34091, Montpellier, France
| |
Collapse
|
22
|
Vaz-Salgado MÁ, García BC, Pérez IF, Munárriz BJ, Domarco PS, González AH, Villar MV, Caro RL, Delgado MLV, Sánchez JMS. SEOM-GEINO clinical guidelines for grade 2 gliomas (2023). Clin Transl Oncol 2024; 26:2856-2865. [PMID: 38662171 PMCID: PMC11467015 DOI: 10.1007/s12094-024-03456-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/08/2024] [Indexed: 04/26/2024]
Abstract
The 2021 World Health Organization (WHO) classification has updated the definition of grade 2 gliomas and the presence of isocitrate dehydrogenase (IDH) mutation has been deemed the cornerstone of diagnosis. Though slow-growing and having a low proliferative index, grade 2 gliomas are incurable by surgery and complementary treatments are vital to improving prognosis. This guideline provides recommendations on the multidisciplinary treatment of grade 2 astrocytomas and oligodendrogliomas based on the best evidence available.
Collapse
Affiliation(s)
- María Ángeles Vaz-Salgado
- Medical Oncology Department, Hospital Universitario Ramón y Cajal, Instituto Ramón y Cajal de Investigación Sanitaria (Irycis) CIBERONC, Madrid, Spain.
| | - Belén Cigarral García
- Medical Oncology Department, Complejo Asistencial Universitario de Salamanca, Salamanca, Spain
| | - Isaura Fernández Pérez
- Medical Oncology Department, Hospital Alvaro Cunqueiro-Complejo Hospitalario Universitario de Vigo, Pontevedra, Spain
| | | | - Paula Sampedro Domarco
- Medical Oncology Department, Complexo Hospitalario Universitario de Ourense (CHUO), Orense, Spain
| | - Ainhoa Hernández González
- Medical Oncology Department, Hospital Germans Trias I Pujol(ICO)-Badalona, Instituto Catalán de Oncología, Barcelona, Spain
| | - María Vieito Villar
- Medical Oncology Department, Hospital Universitario Vall D'Hebron, Barcelona, Spain
| | - Raquel Luque Caro
- Medical Oncology Department, Hospital Universitario Virgen de las Nieves, Instituto de Investigación Biosanitaria Ibs.Granada, Granada, Spain
| | | | - Juan Manuel Sepúlveda Sánchez
- Neuro-Oncology Unit, HM Universitario Sanchinarro-CIOCC, Madrid, Spain.
- Medical Oncology Department, Hospital Universitario 12 de Octubre, Instituto de Investigación 12 de Octubre (I+12), Madrid, Spain.
| |
Collapse
|
23
|
Gately L, Drummond K, Dowling A, Bennett I, Freilich R, Phillips C, Ahern E, Campbell D, Dumas M, Campbell R, Harrup R, Kim GY, Reeves S, Collins IM, Gibbs P. Evolving Practice and Outcomes in Grade 2 Glioma: Real-World Data from a Multi-Institutional Registry. Cancers (Basel) 2024; 16:3514. [PMID: 39456608 PMCID: PMC11506835 DOI: 10.3390/cancers16203514] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2024] [Revised: 10/10/2024] [Accepted: 10/16/2024] [Indexed: 10/28/2024] Open
Abstract
Background: Grade-2 gliomas (G2-glioma) are uncommon. In 2016, RTOG9802 established the addition of chemotherapy after radiation (CRT) as a new standard of care for patients with high-risk G2-glioma, defined as subtotal resection or age ≥40 yrs. Here, we report current practices using real-world data. Methods: Patients diagnosed with G2-glioma from 1 January 2016 to 31 December 2022 were identified in BRAIN, a prospective clinical registry collecting data on patients with brain tumours. High- and low-risk were defined as per RTOG9802. Two time periods, January 2016-December 2019 (TP1) and January 2020-December 2022 (TP2), were defined. Survival was estimated using the Kaplan-Meier method. Results: 224 patients were identified. Overall, 38 (17%) were low-risk, with 35 (91%) observed without further treatment. A total of 186 (83%) were high-risk, with 96 (52%) observed, 63 (34%) receiving CRT, and 19 (10%) receiving radiation. Over time, CRT use increased (TP1 vs. TP2: 22% vs. 36%, p = 0.004), and the rate of biopsy (TP1 vs. TP2: 35% vs. 20%, p = 0.02) and radiotherapy alone (TP1 vs. TP2: 14% vs. 4%, p = 0.01) decreased. Median progression-free survival (PFS) was significantly longer in high-risk patients who received CRT (NR) over observation (39 months) (HR 0.49, p = 0.007). In high-risk patients who were observed, 59 (61%) were progression-free at 12 months and 10 (10%) at 5 years. OS data remains immature. Conclusions: Congruent with RTOG9802, real-world BRAIN data shows CRT is associated with improved PFS compared to observation in high-risk G2-glioma. Whilst CRT use has increased over time, observation after surgery remains the most common strategy, with some high-risk patients achieving clinically meaningful PFS. Validated biomarkers are urgently required to better inform patient management.
Collapse
Affiliation(s)
- Lucy Gately
- Personalised Oncology Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC 3052, Australia
- Department of Medical Oncology, Alfred Health, Melbourne, VIC 3004, Australia
| | - Katharine Drummond
- Department of Neurosurgery, Royal Melbourne Hospital, Parkville, VIC 3052, Australia
- Department of Surgery, University of Melbourne, Parkville, VIC 3010, Australia
| | - Anthony Dowling
- Department of Medical Oncology, St Vincent’s Hospital Melbourne, Fitzroy, VIC 3065, Australia
- Department of Medicine, University of Melbourne, Melbourne, VIC 3010, Australia
| | - Iwan Bennett
- Department of Neurosurgery, The Alfred, Melbourne, VIC 3004, Australia
| | | | - Claire Phillips
- Department of Radiation Oncology, Peter MacCallum Cancer Centre, University of Melbourne, Melbourne, VIC 3010, Australia
- Sir Peter MacCallum Department of Oncology, University of Melbourne, Melbourne, VIC 3010, Australia
| | - Elizabeth Ahern
- Department of Medical Oncology, Monash Health, Clayton VIC 3168, Australia
| | - David Campbell
- Department of Medical Oncology, University Hospital Geelong, Barwon Health, Geelong, VIC 3220, Australia
| | - Megan Dumas
- Personalised Oncology Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC 3052, Australia
| | | | - Rosemary Harrup
- Cancer & Blood Services, Royal Hobart Hospital, Hobart, TAS 7000, Australia
- Menzies Research Institute, University of Tasmania, Hobart, TAS 7005, Australia
| | - Grace Y. Kim
- Personalised Oncology Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC 3052, Australia
| | - Simone Reeves
- Ballarat Austin Radiation Oncology Centre, Ballarat, VIC 3350, Australia
| | - Ian M. Collins
- Department of Medical Oncology, South West Regional Cancer Centre, Geelong, VIC 3220, Australia
| | - Peter Gibbs
- Personalised Oncology Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC 3052, Australia
| |
Collapse
|
24
|
Chen T, Meng J, Yu K, Huang T, Zhao J. Chromatin Licensing and DNA Replication Factor 1 (CDT1) Is a Potential Prognostic Biomarker Involved in the Malignant Biological Behavior of Glioma. ACS Pharmacol Transl Sci 2024; 7:3131-3143. [PMID: 39416957 PMCID: PMC11475523 DOI: 10.1021/acsptsci.4c00312] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2024] [Revised: 09/06/2024] [Accepted: 09/13/2024] [Indexed: 10/19/2024]
Abstract
Glioma is the primary malignant tumor with the highest incidence rate in the adult central nervous system. The application of bioinformatics methods to analyze the RNA sequences of multiple gliomas revealed that the CDT1 gene has a significant impact on the cell cycle of glioma cells. Subsequently, we comprehensively and systematically investigated the expression of CDT1 in gliomas through bioinformatics analysis, clinical tissue specimens, and in vitro functional experiments. Our study is the first to report the expression of CDT1 in glioma. Our findings demonstrate that CDT1 plays a crucial role in the proliferation and invasion of glioma. Additionally, our bioinformatics analysis identified several other genes and signaling pathways that are dysregulated in multifocal gliomas, providing potential targets for further research and drug development.
Collapse
Affiliation(s)
- Tiange Chen
- Department
of Neurosurgery, Hainan General Hospital/Hainan
Affiliated Hospital of Hainan Medical University, Haikou 570311, China
| | - Jiawei Meng
- Department
of Laboratory Medicine, The Third Xiangya
Hospital, Central South University, Changsha, Hunan 410013, China
| | - Ke Yu
- Department
of Laboratory Medicine, The Third Xiangya
Hospital, Central South University, Changsha, Hunan 410013, China
| | - Tianxiang Huang
- Department
of Neurosurgery, and National Clinical Research Center of Geriatric
Disorders, Xiangya Hospital, Central South
University, 87 Xiangya Road, Changsha, Hunan 410008, China
| | - Jiannong Zhao
- Department
of Neurosurgery, Hainan General Hospital/Hainan
Affiliated Hospital of Hainan Medical University, Haikou 570311, China
| |
Collapse
|
25
|
Kinslow CJ, Roy S, Iwamoto FM, Brown PD, DeStephano DM, Canoll PD, Qureshi SS, Gallito M, Sisti MB, Bruce JN, Horowitz DP, Kachnic LA, Neugut AI, Yu JB, Mehta MP, Cheng SK, Wang TJC. The IDH paradox: Meta-analysis of alkylating chemotherapy in IDH-wild type and -mutant lower grade gliomas. Neuro Oncol 2024; 26:1839-1849. [PMID: 38943513 PMCID: PMC11449043 DOI: 10.1093/neuonc/noae102] [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: 07/01/2024] Open
Abstract
BACKGROUND IDH-wild type (-wt) status is a prerequisite for the diagnosis of glioblastoma (GBM); however, IDH-wt gliomas with low-grade or anaplastic morphology have historically been excluded from GBM trials and may represent a distinct prognostic entity. While alkylating agent chemotherapy improves overall survival (OS) and progression-free survival (PFS) for IDH-wt GBM and also IDH-mutant gliomas, irrespective of grade, the benefit for IDH-wt diffuse histologic lower-grade gliomas is unclear. METHODS We performed a meta-analysis of randomized clinical trials for World Health Organization (WHO) grades 2-3 gliomas (2009 to present) to determine the effect of alkylating chemotherapy on IDH-wt and -mutant gliomas using a random-effects model with inverse-variance pooling. RESULTS We identified 6 trials with 1204 patients (430 IDH-wt, 774 IDH-mutant) that evaluated alkylating chemoradiotherapy versus radiotherapy alone, allowing us to perform an analysis focused on the value of adding alkylating chemotherapy to radiotherapy. For patients with IDH-wt tumors, alkylating chemotherapy added to radiotherapy was associated with improved PFS (HR:0.77 [95% CI: 0.62-0.97], P = .03) but not OS (HR:0.87 [95% CI: 0.64-1.18], P = .17). For patients with IDH-mutant tumors, alkylating chemotherapy added to radiotherapy improved both OS (HR:0.52 [95% CI: 0.42-0.64], P < .001) and PFS (HR = 0.47 [95% CI: 0.39-0.57], P < .001) compared to radiotherapy alone. The magnitude of benefit was similar for IDH-mutant gliomas with or without 1p19q-codeletion. CONCLUSIONS Alkylating chemotherapy reduces mortality by 48% and progression by 53% for patients with IDH-mutant gliomas. Optimal management of IDH-wt diffuse histologic lower-grade gliomas remains to be determined, as there is little evidence supporting an OS benefit from alkylating chemotherapy.
Collapse
Affiliation(s)
- Connor J Kinslow
- Herbert Irving Comprehensive Cancer Center, Columbia University Vagelos College of Physicians and Surgeons and NewYork-Presbyterian, New York, New York, USA
- Department of Radiation Oncology, Columbia University Vagelos College of Physicians and Surgeons and NewYork-Presbyterian, New York, New York, USA
| | - Soumyajit Roy
- Department of Radiation Oncology, Rush University Medical Center, Chicago, Illinois, USA
| | - Fabio M Iwamoto
- Department of Neurology, Columbia University Vagelos College of Physicians and Surgeons and NewYork-Presbyterian, New York, New York, USA
- Herbert Irving Comprehensive Cancer Center, Columbia University Vagelos College of Physicians and Surgeons and NewYork-Presbyterian, New York, New York, USA
| | - Paul D Brown
- Department of Radiation Oncology, Mayo Clinic, Rochester, Minnesota, USA
| | - David M DeStephano
- Herbert Irving Comprehensive Cancer Center, Columbia University Vagelos College of Physicians and Surgeons and NewYork-Presbyterian, New York, New York, USA
- Department of Radiation Oncology, Columbia University Vagelos College of Physicians and Surgeons and NewYork-Presbyterian, New York, New York, USA
| | - Peter D Canoll
- Departments of Pathology and Cell Biology, Columbia University Vagelos College of Physicians and Surgeons and NewYork-Presbyterian, New York, New York, USA
- Herbert Irving Comprehensive Cancer Center, Columbia University Vagelos College of Physicians and Surgeons and NewYork-Presbyterian, New York, New York, USA
| | - Summer S Qureshi
- Department of Radiation Oncology, Columbia University Vagelos College of Physicians and Surgeons and NewYork-Presbyterian, New York, New York, USA
| | - Matthew Gallito
- Herbert Irving Comprehensive Cancer Center, Columbia University Vagelos College of Physicians and Surgeons and NewYork-Presbyterian, New York, New York, USA
- Department of Radiation Oncology, Columbia University Vagelos College of Physicians and Surgeons and NewYork-Presbyterian, New York, New York, USA
| | - Michael B Sisti
- Department of Neurological Surgery, Columbia University Vagelos College of Physicians and Surgeons and NewYork-Presbyterian, New York, New York, USA
- Herbert Irving Comprehensive Cancer Center, Columbia University Vagelos College of Physicians and Surgeons and NewYork-Presbyterian, New York, New York, USA
| | - Jeffrey N Bruce
- Department of Neurological Surgery, Columbia University Vagelos College of Physicians and Surgeons and NewYork-Presbyterian, New York, New York, USA
- Herbert Irving Comprehensive Cancer Center, Columbia University Vagelos College of Physicians and Surgeons and NewYork-Presbyterian, New York, New York, USA
| | - David P Horowitz
- Herbert Irving Comprehensive Cancer Center, Columbia University Vagelos College of Physicians and Surgeons and NewYork-Presbyterian, New York, New York, USA
- Department of Radiation Oncology, Columbia University Vagelos College of Physicians and Surgeons and NewYork-Presbyterian, New York, New York, USA
| | - Lisa A Kachnic
- Herbert Irving Comprehensive Cancer Center, Columbia University Vagelos College of Physicians and Surgeons and NewYork-Presbyterian, New York, New York, USA
- Department of Radiation Oncology, Columbia University Vagelos College of Physicians and Surgeons and NewYork-Presbyterian, New York, New York, USA
| | - Alfred I Neugut
- Department of Medicine, Vagelos College of Physicians and Surgeons, and Department of Epidemiology, Mailman School of Public Health, Columbia University Vagelos College of Physicians and Surgeons and NewYork-Presbyterian, New York, New York, USA
- Herbert Irving Comprehensive Cancer Center, Columbia University Vagelos College of Physicians and Surgeons and NewYork-Presbyterian, New York, New York, USA
| | - James B Yu
- Department of Medical Oncology, Yale School of Medicine, New Haven, Connecticut, USA
- Department of Radiation Oncology Medical Oncology, Saint Francis Hospital, Hartford, Connecticut, USA
| | - Minesh P Mehta
- Department of Radiation Oncology, Miami Cancer Institute, Baptist Health South Florida, Miami, Florida, USA
| | - Simon K Cheng
- Department of Radiation Oncology, James J. Peters Veterans Affairs Medical Center, Bronx, New York, USA
- Herbert Irving Comprehensive Cancer Center, Columbia University Vagelos College of Physicians and Surgeons and NewYork-Presbyterian, New York, New York, USA
- Department of Radiation Oncology, Columbia University Vagelos College of Physicians and Surgeons and NewYork-Presbyterian, New York, New York, USA
| | - Tony J C Wang
- Herbert Irving Comprehensive Cancer Center, Columbia University Vagelos College of Physicians and Surgeons and NewYork-Presbyterian, New York, New York, USA
- Department of Radiation Oncology, Columbia University Vagelos College of Physicians and Surgeons and NewYork-Presbyterian, New York, New York, USA
| |
Collapse
|
26
|
Li M, Liu J, Weng J, Dong G, Chen X, Cui Y, Ren X, Shen S, Jiang H, Zhang X, Zhao X, Li M, Wang X, Ren H, Li Q, Zhang Y, Cheng Q, Yu Y, Lin S. Unveiling hierarchy and spatial distribution of O 6-methylguanine-DNA methyltransferase promoter methylation in World Health Organization grade 2-3 gliomas. Cancer Sci 2024; 115:3403-3414. [PMID: 39101880 PMCID: PMC11447971 DOI: 10.1111/cas.16268] [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: 03/30/2024] [Revised: 06/13/2024] [Accepted: 06/24/2024] [Indexed: 08/06/2024] Open
Abstract
This study investigated the role of O6-methylguanine-DNA methyltransferase promoter (MGMTp) methylation hierarchy and heterogeneity in grade 2-3 gliomas, focusing on variations in chemotherapy benefits and resection dependency. A cohort of 668 newly diagnosed grade 2-3 gliomas, with comprehensive clinical, radiological, and molecular data, formed the basis of this analysis. The extent of resection was categorized into gross total resection (GTR ≥100%), subtotal resection (STR >90%), and partial resection (PR ≤90%). MGMTp methylation levels were examined using quantitative pyrosequencing. Our findings highlighted the critical role of GTR in improving the prognosis for astrocytomas (IDH1/2-mutant and 1p/19q non-codeleted), contrasting with its lesser significance for oligodendrogliomas (IDH1/2 mutation and 1p/19q codeletion). Oligodendrogliomas demonstrated the highest average MGMTp methylation levels (median: 28%), with a predominant percentage of methylated cases (average methylation levels >20%). Astrocytomas were more common in the low-methylated group (10%-20%), while IDH wild-type gliomas were mostly unmethylated (<10%). Spatial distribution analysis revealed a decrement in frontal lobe involvement from methylated, low-methylated to unmethylated cases (72.8%, 59.3%, and 47.8%, respectively). In contrast, low-methylated and unmethylated cases were more likely to invade the temporal-insular region (19.7%, 34.3%, and 40.4%, respectively). Astrocytomas with intermediate MGMTp methylation were notably associated with temporal-insular involvement, potentially indicating a moderate response to temozolomide and underscoring the importance of aggressive resection strategies. In conclusion, our study elucidates the complex interplay of MGMTp methylation hierarchy and heterogeneity among grade 2-3 gliomas, providing insights into why astrocytomas and IDH wild-type lower-grade glioma might derive less benefit from chemotherapy.
Collapse
Affiliation(s)
- Mingxiao Li
- Department of Neurosurgery, China-Japan Friendship Hospital, Beijing, China
- Department of Neurosurgical Oncology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Jiang Liu
- Department of Neurosurgery, China-Japan Friendship Hospital, Beijing, China
| | - Jiancong Weng
- Department of Neurosurgery, China-Japan Friendship Hospital, Beijing, China
| | - Gehong Dong
- Department of Pathology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Xuzhu Chen
- Department of Radiology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Yong Cui
- Department of Neurosurgical Oncology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
- Department of Neurosurgery, Beijing Neurosurgical Institute, Capital Medical University, Beijing, China
| | - Xiaohui Ren
- Department of Neurosurgical Oncology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
- Department of Neurosurgery, Beijing Neurosurgical Institute, Capital Medical University, Beijing, China
| | - Shaoping Shen
- Department of Neurosurgical Oncology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
- Department of Neurosurgery, Beijing Neurosurgical Institute, Capital Medical University, Beijing, China
| | - Haihui Jiang
- Department of Neurosurgery, Peking University Third Hospital, Beijing, China
| | - Xiaokang Zhang
- Department of Neurosurgical Oncology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
- Department of Neurosurgery, Beijing Neurosurgical Institute, Capital Medical University, Beijing, China
| | - Xuzhe Zhao
- Department of Neurosurgical Oncology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
- Department of Neurosurgery, Beijing Neurosurgical Institute, Capital Medical University, Beijing, China
| | - Ming Li
- Department of Neurosurgical Oncology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
- Department of Neurosurgery, Beijing Neurosurgical Institute, Capital Medical University, Beijing, China
| | - Xijie Wang
- Department of Neurosurgical Oncology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
- Department of Neurosurgery, Beijing Neurosurgical Institute, Capital Medical University, Beijing, China
| | - Hongxiang Ren
- Department of Neurosurgery, China-Japan Friendship Hospital, Beijing, China
| | - Qiang Li
- Department of Neurosurgery, China-Japan Friendship Hospital, Beijing, China
| | - Yulian Zhang
- Department of Neurosurgery, China-Japan Friendship Hospital, Beijing, China
| | - Quan Cheng
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Yanbing Yu
- Department of Neurosurgery, China-Japan Friendship Hospital, Beijing, China
| | - Song Lin
- Department of Neurosurgical Oncology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
- Department of Neurosurgery, Beijing Neurosurgical Institute, Capital Medical University, Beijing, China
| |
Collapse
|
27
|
Albert NL, Le Rhun E, Minniti G, Mair MJ, Galldiks N, Tolboom N, Jakola AS, Niyazi M, Smits M, Verger A, Cicone F, Weller M, Preusser M. Translating the theranostic concept to neuro-oncology: disrupting barriers. Lancet Oncol 2024; 25:e441-e451. [PMID: 39214115 DOI: 10.1016/s1470-2045(24)00145-1] [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: 01/20/2024] [Revised: 02/23/2024] [Accepted: 03/08/2024] [Indexed: 09/04/2024]
Abstract
Theranostics integrate molecular imaging and targeted radionuclide therapy for personalised cancer therapy. Theranostic treatments have shown meaningful efficacy in randomised clinical trials and are approved for clinical use in prostate cancer and neuroendocrine tumours. Brain tumours represent an unmet clinical need and theranostics might offer effective treatment options, although specific issues need to be considered for clinical development. In this Policy Review, we discuss opportunities and challenges of developing targeted radionuclide therapies for the treatment of brain tumours including glioma, meningioma, and brain metastasis. The rational choice of molecular treatment targets is highlighted, including the potential relevance of different types of targeted radionuclide therapeutics, and the role of the blood-brain barrier and blood-tumour barrier. Furthermore, we discuss considerations for effective clinical trial design and conduct, as well as logistical and regulatory challenges for implementation of radionuclide therapies into neuro-oncological practice. Rational development will foster successful translation of the theranostic concept to brain tumours.
Collapse
Affiliation(s)
- Nathalie L Albert
- Department of Nuclear Medicine, Ludwig Maximilians University Hospital, Ludwig Maximilians University Munich, Munich, Germany
| | - Emilie Le Rhun
- Department of Neurology, University Hospital and University of Zurich, Zurich, Switzerland; Department of Neurosurgery, University Hospital and University of Zurich, Zurich, Switzerland
| | - Giuseppe Minniti
- Department of Radiological Sciences, Oncology and Anatomical Pathology, Sapienza University, Rome, Italy
| | - Maximilian J Mair
- Department of Nuclear Medicine, Ludwig Maximilians University Hospital, Ludwig Maximilians University Munich, Munich, Germany; Division of Oncology, Department of Medicine I, Medical University of Vienna, Vienna, Austria
| | - Norbert Galldiks
- Department of Neurology, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany; Institute of Neuroscience and Medicine (INM-3), Research Center Juelich, Juelich, Germany; Center for Integrated Oncology Aachen Bonn Cologne Duesseldorf, Germany
| | - Nelleke Tolboom
- Department of Radiology and Nuclear Medicine, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Asgeir S Jakola
- Institute of Neuroscience and Physiology, Department of Clinical Neuroscience, Sahlgrenska Academy, Gothenburg, Sweden; Department of Neurosurgery, Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Maximilian Niyazi
- Department of Radiation Oncology, University Hospital Tübingen, Tübingen, Germany
| | - Marion Smits
- Department of Radiology & Nuclear Medicine, Erasmus MC, Rotterdam, The Netherlands; Medical Delta, Delft, The Netherlands
| | - Antoine Verger
- Department of Nuclear Medicine and Nancyclotep Imaging Platform, CHRU-Nancy, IADI, INSERM, UMR 1254, Université de Lorraine, Nancy, France
| | - Francesco Cicone
- Nuclear Medicine Unit, Department of Experimental and Clinical Medicine, Magna Graecia University of Catanzaro, Catanzaro, Italy
| | - Michael Weller
- Department of Neurology, University Hospital and University of Zurich, Zurich, Switzerland
| | - Matthias Preusser
- Division of Oncology, Department of Medicine I, Medical University of Vienna, Vienna, Austria.
| |
Collapse
|
28
|
Andrew Awuah W, Shah MH, Tan JK, Ranganathan S, Sanker V, Darko K, Tenkorang PO, Adageba BB, Ahluwalia A, Shet V, Aderinto N, Kundu M, Abdul‐Rahman T, Atallah O. Immunotherapeutic advances in glioma management: The rise of vaccine-based approaches. CNS Neurosci Ther 2024; 30:e70013. [PMID: 39215399 PMCID: PMC11364516 DOI: 10.1111/cns.70013] [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: 02/19/2024] [Revised: 07/23/2024] [Accepted: 08/12/2024] [Indexed: 09/04/2024] Open
Abstract
BACKGROUND Gliomas, particularly glioblastoma multiforme (GBM), are highly aggressive brain tumors that present significant challenges in oncology due to their rapid progression and resistance to conventional therapies. Despite advancements in treatment, the prognosis for patients with GBM remains poor, necessitating the exploration of novel therapeutic approaches. One such emerging strategy is the development of glioma vaccines, which aim to stimulate the immune system to target and destroy tumor cells. AIMS This review aims to provide a comprehensive evaluation of the current landscape of glioma vaccine development, analyzing the types of vaccines under investigation, the outcomes of clinical trials, and the challenges and opportunities associated with their implementation. The goal is to highlight the potential of glioma vaccines in advancing more effective and personalized treatments for glioma patients. MATERIALS AND METHODS This narrative review systematically assessed the role of glioma vaccines by including full-text articles published between 2000 and 2024 in English. Databases such as PubMed/MEDLINE, EMBASE, the Cochrane Library, and Scopus were searched using key terms like "glioma," "brain tumor," "glioblastoma," "vaccine," and "immunotherapy." The review incorporated both pre-clinical and clinical studies, including descriptive studies, animal-model studies, cohort studies, and observational studies. Exclusion criteria were applied to omit abstracts, case reports, posters, and non-peer-reviewed studies, ensuring the inclusion of high-quality evidence. RESULTS Clinical trials investigating various glioma vaccines, including peptide-based, DNA/RNA-based, whole-cell, and dendritic-cell vaccines, have shown promising results. These vaccines demonstrated potential in extending survival rates and managing adverse events in glioma patients. However, significant challenges remain, such as therapeutic resistance due to tumor heterogeneity and immune evasion mechanisms. Moreover, the lack of standardized guidelines for evaluating vaccine responses and issues related to ethical considerations, regulatory hurdles, and vaccine acceptance among patients further complicate the implementation of glioma vaccines. DISCUSSION Addressing the challenges associated with glioma vaccines involves exploring combination therapies, targeted approaches, and personalized medicine. Combining vaccines with traditional therapies like radiotherapy or chemotherapy may enhance efficacy by boosting the immune system's ability to fight tumor cells. Personalized vaccines tailored to individual patient profiles present an opportunity for improved outcomes. Furthermore, global collaboration and equitable distribution are critical for ensuring access to glioma vaccines, especially in low- and middle-income countries with limited healthcare resources CONCLUSION: Glioma vaccines represent a promising avenue in the fight against gliomas, offering hope for improving patient outcomes in a disease that is notoriously difficult to treat. Despite the challenges, continued research and the development of innovative strategies, including combination therapies and personalized approaches, are essential for overcoming current barriers and transforming the treatment landscape for glioma patients.
Collapse
Affiliation(s)
| | | | | | | | - Vivek Sanker
- Department of NeurosurgeryTrivandrum Medical CollegeTrivandrumKeralaIndia
| | - Kwadwo Darko
- Department of NeurosurgeryKorle Bu Teaching HospitalAccraGhana
| | | | - Bryan Badayelba Adageba
- Kwame Nkrumah University of Science and Technology School of Medicine and DentistryKumasiGhana
| | | | - Vallabh Shet
- Faculty of MedicineBangalore Medical College and Research InstituteBangaloreKarnatakaIndia
| | - Nicholas Aderinto
- Department of Internal MedicineLAUTECH Teaching HospitalOgbomosoNigeria
| | - Mrinmoy Kundu
- Institute of Medical Sciences and SUM HospitalBhubaneswarOdishaIndia
| | | | - Oday Atallah
- Department of Neurosurgery, Hannover Medical SchoolHannoverGermany
| |
Collapse
|
29
|
Zhu E, Wang J, Shi W, Chen Z, Zhu M, Xu Z, Li L, Shan D. Utilizing machine learning to tailor radiotherapy and chemoradiotherapy for low-grade glioma patients. PLoS One 2024; 19:e0306711. [PMID: 39163387 PMCID: PMC11335161 DOI: 10.1371/journal.pone.0306711] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2023] [Accepted: 06/23/2024] [Indexed: 08/22/2024] Open
Abstract
BACKGROUND There is ongoing uncertainty about the effectiveness of various adjuvant treatments for low-grade gliomas (LGGs). Machine learning (ML) models that predict individual treatment effects (ITE) and provide treatment recommendations could help tailor treatments to each patient's needs. OBJECTIVE We sought to discern the individual suitability of radiotherapy (RT) or chemoradiotherapy (CRT) in LGG patients using ML models. METHODS Ten ML models, trained to infer ITE in 4,042 LGG patients, were assessed. We compared patients who followed treatment recommendations provided by the models with those who did not. To mitigate the risk of treatment selection bias, we employed inverse probability treatment weighting (IPTW). RESULTS The Balanced Survival Lasso-Network (BSL) model showed the most significant protective effect among all the models we tested (hazard ratio (HR): 0.52, 95% CI, 0.41-0.64; IPTW-adjusted HR: 0.58, 95% CI, 0.45-0.74; the difference in restricted mean survival time (DRMST): 9.11, 95% CI, 6.19-12.03; IPTW-adjusted DRMST: 9.17, 95% CI, 6.30-11.83). CRT presented a protective effect in the 'recommend for CRT' group (IPTW-adjusted HR: 0.60, 95% CI, 0.39-0.93) yet presented an adverse effect in the 'recommend for RT' group (IPTW-adjusted HR: 1.64, 95% CI, 1.19-2.25). Moreover, the models predict that younger patients and patients with overlapping lesions or tumors crossing the midline are better suited for CRT (HR: 0.62, 95% CI, 0.42-0.91; IPTW-adjusted HR: 0.59, 95% CI, 0.36-0.97). CONCLUSION Our findings underscore the potential of the BSL model in guiding the choice of adjuvant treatment for LGGs patients, potentially improving survival time. This study emphasizes the importance of ML in customizing patient care, understanding the nuances of treatment selection, and advancing personalized medicine.
Collapse
Affiliation(s)
- Enzhao Zhu
- School of Medicine, Tongji University, Shanghai, China
| | - Jiayi Wang
- School of Medicine, Tongji University, Shanghai, China
| | - Weizhong Shi
- Shanghai Hospital Development Center, Shanghai, China
| | - Zhihao Chen
- School of Business, East China University of Science and Technology, Shanghai, China
| | - Min Zhu
- Department of Computer Science and Technology, School of Electronics and Information Engineering, Tongji University, Shanghai, China
| | - Ziqin Xu
- Department of Biobehavioral Sciences, Columbia University, New York, NY, United States of America
| | - Linlin Li
- School of Medicine, Tongji University, Shanghai, China
| | - Dan Shan
- Department of Biobehavioral Sciences, Columbia University, New York, NY, United States of America
- School of Medicine, National University of Ireland, Galway, Ireland
| |
Collapse
|
30
|
Schiff D. Low-Grade Gliomas: A New Mutation, New Targeted Therapy, and Many Questions. Neurology 2024; 103:e209688. [PMID: 39008801 DOI: 10.1212/wnl.0000000000209688] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/17/2024] Open
Abstract
The discovery in 2008 that many adult gliomas harbor a hitherto unknown mutation in the metabolic gene isocitrate dehydrogenase (IDH) initiated revolutionary advances in our understanding of the biology, and correspondingly our classification, of gliomas. IDH mutations are found in most nonglioblastoma adult gliomas and portend a better prognosis. Massive efforts have unraveled many of the pleiotropic cellular effects of these mutations and spawned several lines of investigation to target the effect to therapeutic benefit. In this article are reviewed the implications of the IDH mutation in gliomas, in particular focusing on recent studies that have culminated in a rare positive phase 3 trial in these generally refractory tumors.
Collapse
Affiliation(s)
- David Schiff
- From the Departments of Neurology, Neurological Surgery, and Medicine, University of Virginia Health System
| |
Collapse
|
31
|
Budhiraja S, McManus G, Baisiwala S, Perrault EN, Cho S, Saathoff M, Chen L, Park CH, Kazi HA, Dmello C, Lin P, James CD, Sonabend AM, Heiland DH, Ahmed AU. ARF4-mediated retrograde trafficking as a driver of chemoresistance in glioblastoma. Neuro Oncol 2024; 26:1421-1437. [PMID: 38506351 PMCID: PMC11300013 DOI: 10.1093/neuonc/noae059] [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: 10/17/2023] [Indexed: 03/21/2024] Open
Abstract
BACKGROUND Cellular functions hinge on the meticulous orchestration of protein transport, both spatially and temporally. Central to this process is retrograde trafficking, responsible for targeting proteins to the nucleus. Despite its link to many diseases, the implications of retrograde trafficking in glioblastoma (GBM) are still unclear. METHODS To identify genetic drivers of TMZ resistance, we conducted comprehensive CRISPR-knockout screening, revealing ADP-ribosylation factor 4 (ARF4), a regulator of retrograde trafficking, as a major contributor. RESULTS Suppressing ARF4 significantly enhanced TMZ sensitivity in GBM patient-derived xenograft (PDX) models, leading to improved survival rates (P < .01) in both primary and recurrent lines. We also observed that TMZ exposure stimulates ARF4-mediated retrograde trafficking. Proteomics analysis of GBM cells with varying levels of ARF4 unveiled the influence of this pathway on EGFR signaling, with increased nuclear trafficking of EGFR observed in cells with ARF4 overexpression and TMZ treatment. Additionally, spatially resolved RNA-sequencing of GBM patient tissues revealed substantial correlations between ARF4 and crucial nuclear EGFR (nEGFR) downstream targets, such as MYC, STAT1, and DNA-PK. Decreased activity of DNA-PK, a DNA repair protein downstream of nEGFR signaling that contributes to TMZ resistance, was observed in cells with suppressed ARF4 levels. Notably, treatment with DNA-PK inhibitor, KU-57788, in mice with a recurrent PDX line resulted in prolonged survival (P < .01), highlighting the promising therapeutic implications of targeting proteins reliant on ARF4-mediated retrograde trafficking. CONCLUSIONS Our findings demonstrate that ARF4-mediated retrograde trafficking contributes to the development of TMZ resistance, cementing this pathway as a viable strategy to overcome chemoresistance in GBM.
Collapse
Affiliation(s)
- Shreya Budhiraja
- Department of Neurological Surgery, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
- Northwestern Medicine Malnati Brain Tumor Institute of the Lurie Comprehensive Cancer Center, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| | - Graysen McManus
- Department of Neurological Surgery, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| | | | - Ella N Perrault
- Department of Neurological Surgery, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| | - Sia Cho
- Department of Neurobiology, Northwestern University, Evanston, Illinois, USA
| | - Miranda Saathoff
- Department of Neurological Surgery, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| | - Li Chen
- Department of Neurological Surgery, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| | - Cheol H Park
- Department of Neurological Surgery, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| | - Hasaan A Kazi
- Department of Neurological Surgery, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| | - Crismita Dmello
- Department of Neurological Surgery, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| | - Peiyu Lin
- Department of Neurological Surgery, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| | - C David James
- Department of Neurological Surgery, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
- Northwestern Medicine Malnati Brain Tumor Institute of the Lurie Comprehensive Cancer Center, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| | - Adam M Sonabend
- Department of Neurological Surgery, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
- Northwestern Medicine Malnati Brain Tumor Institute of the Lurie Comprehensive Cancer Center, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| | - Dieter H Heiland
- Department of Neurological Surgery, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
- Microenvironment and Immunology Research Laboratory, Medical Center - University of Freiburg, Freiburg, Germany
- Department of Neurosurgery, Medical Center - University of Freiburg, Freiburg, Germany
- Faculty of Medicine, University of Freiburg, Freiburg, Germany
- German Cancer Consortium (DKTK), Freiburg, Germany
| | - Atique U Ahmed
- Department of Neurological Surgery, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
- Northwestern Medicine Malnati Brain Tumor Institute of the Lurie Comprehensive Cancer Center, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| |
Collapse
|
32
|
Staicu GA, Tataranu LG, Tache DE, Popescu SO, Artene SA, Danoiu S, Sfredel V, Barcan EN, Baloi SC, Dricu A. Evaluation of Single and Combined Temozolomide and Doxorubicin Treatment Responses in Low- and High-Grade Glioma In Vitro. Cureus 2024; 16:e66928. [PMID: 39280423 PMCID: PMC11401637 DOI: 10.7759/cureus.66928] [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] [Accepted: 08/14/2024] [Indexed: 09/18/2024] Open
Abstract
BACKGROUND Astrocytoma, the most common type of glioma, can histologically be low or high grade. Treatment recommendations for astrocytic tumors are based on the histopathological and molecular phenotype. For grade 2 astrocytoma, the combination of radiotherapy and adjuvant chemotherapy with procarbazine, lomustine, and vincristine (PCV) is better than radiotherapy alone. Temozolomide (TMZ) is being increasingly recognized as a replacement for PCV in brain tumor therapy, due to the lower myelotoxicity. TMZ is currently a well-established first-line treatment for grade 3 astrocytoma, grade 4 astrocytoma, and glioblastoma and it is also sporadically used for grade 2 astrocytoma. However, TMZ faces multiple challenges such as adverse effects and drug resistance. METHODS In this study, we compared the cytotoxic effect induced by TMZ and doxorubicin (DOXO), alone and in combination, on a low-grade astrocytoma cell line (AC1B) and a high-grade glioma cell line (GB1B). RESULTS We found that TMZ and DOXO, each produced a cytotoxic effect in monotherapy. GB1B cell line was more sensitive to the treatment than AC1B cells, at a 7- and 10-day exposure to the DOXO. However, when the duration of the treatment was extended to 14 days, GB1B cells became more resistant to DOXO treatment, compared to AC1B cells. Regarding the treatment with TMZ, GB1B exhibited greater resistance to TMZ compared to AC1B, across all studied intervals and the resistance to treatment of GB1B increased with longer exposure time. However, in combined therapy, the drugs did not exert a synergistic effect on any astrocytic cell line. CONCLUSIONS The current data suggest that both TMZ and DOXO exhibit efficient therapeutic effects on low- and high-grade glioma cells. However, no synergistic effect was observed for combined therapy.
Collapse
Affiliation(s)
| | - Ligia G Tataranu
- Department of Neurosurgery, Bagdasar-Arseni Clinical Emergency Hospital, Bucharest, ROU
- Department of Neurosurgery, Carol Davila University of Medicine and Pharmacy, Bucharest, ROU
| | - Daniela Elise Tache
- Department of Biochemistry, University of Medicine and Pharmacy of Craiova, Craiova, ROU
| | - Stefana Oana Popescu
- Department of Biochemistry, University of Medicine and Pharmacy of Craiova, Craiova, ROU
| | | | - Suzana Danoiu
- Department of Physiology, University of Medicine and Pharmacy of Craiova, Craiova, ROU
| | - Veronica Sfredel
- Department of Physiology, University of Medicine and Pharmacy of Craiova, Craiova, ROU
| | - Edmond Nicolae Barcan
- Department of Biochemistry, University of Medicine and Pharmacy of Craiova, Craiova, ROU
| | - Stefania Carina Baloi
- Department of Biochemistry, University of Medicine and Pharmacy of Craiova, Craiova, ROU
| | - Anica Dricu
- Department of Biochemistry, University of Medicine and Pharmacy of Craiova, Craiova, ROU
| |
Collapse
|
33
|
Kotecha R, Schiff D, Chakravarti A, Fleming JL, Brown PD, Puduvalli VK, Vogelbaum MA, Gondi V, Gallus M, Okada H, Mehta MP. Multidisciplinary Management of Isocitrate Dehydrogenase-Mutated Gliomas in a Contemporary Molecularly Defined Era. J Clin Oncol 2024; 42:2588-2598. [PMID: 38833641 PMCID: PMC11283772 DOI: 10.1200/jco.23.02195] [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: 10/08/2023] [Revised: 03/04/2024] [Accepted: 04/04/2024] [Indexed: 06/06/2024] Open
Abstract
Mutations in isocitrate dehydrogenase (IDH) genes, an early step in the ontogeny of lower-grade gliomas, induce global epigenetic changes characterized by a hypermethylation phenotype and are critical to tumor classification, treatment decision making, and estimation of patient prognosis. The introduction of IDH inhibitors to block the oncogenic neomorphic function of the mutated protein has resulted in new therapeutic options for these patients. To appreciate the implications of these recent IDH inhibitor results, it is important to juxtapose historical outcomes with chemoradiotherapy. Herein, we rationally evaluate recent IDH inhibitor data within historical precedents to guide contemporary decisions regarding the role of observation, maximal safe resection, adjuvant therapies, and the import of patient and tumor variables. The biological underpinnings of the IDH pathway and the mechanisms, impact, and limitations of IDH inhibitors, the actual magnitude of tumor regression and patient benefit, and emergence of resistance pathways are presented to guide future trial development. Management in the current, molecularly defined era will require careful patient selection and risk factor assessment, followed by an open dialog about the results of studies such as INDIGO, as well as mature data from legacy trials, and a discussion about risk-versus-benefit for the choice of treatment, with multidisciplinary decision making as an absolute prerequisite.
Collapse
Affiliation(s)
- Rupesh Kotecha
- Department of Radiation Oncology, Miami Cancer Institute, Baptist Health South Florida, Miami, FL
| | - David Schiff
- Division of Neuro-Oncology, Departments of Neurology, Neurological Surgery, and Medicine, University of Virginia Health System, Charlottesville, VA
| | - Arnab Chakravarti
- Department of Radiation Oncology, James Cancer Hospital and Solove Research Institute, The Ohio State University College of Medicine, Columbus, OH
| | - Jessica L. Fleming
- Department of Radiation Oncology, James Cancer Hospital and Solove Research Institute, The Ohio State University College of Medicine, Columbus, OH
| | - Paul D. Brown
- Department of Radiation Oncology, Mayo Clinic, Rochester, MN
| | - Vinay K. Puduvalli
- Department of Neuro-Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX
| | | | - Vinai Gondi
- Department of Radiation Oncology, Northwestern Medicine West Region, Lou & Jean Malnati Brain Tumor Institute, Northwestern University, Warrenville, IL
| | - Marco Gallus
- Department of Neurosurgery, UCSF, San Francisco, CA
| | - Hideho Okada
- Department of Neurosurgery, UCSF, San Francisco, CA
| | - Minesh P. Mehta
- Department of Radiation Oncology, Miami Cancer Institute, Baptist Health South Florida, Miami, FL
| |
Collapse
|
34
|
Tao Q, Yang S, Wang S, Yang Y, Yu S, Pan Y, Li Y, Zhang J, Hu C. Neural Progenitor Cell-Mediated Magnetic Nanoparticles for Magnetic Resonance Imaging and Photothermal Therapy of Glioma. ACS APPLIED BIO MATERIALS 2024; 7:4553-4561. [PMID: 38875521 DOI: 10.1021/acsabm.4c00414] [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: 06/16/2024]
Abstract
Glioma is the most common primary malignant tumor in the brain. The diagnostic accuracy and treatment efficiency of glioma are facing great challenges due to the presence of the blood-brain barrier (BBB) and the high infiltration of glioma. There is an urgent need to explore the combination of diagnostic and therapeutic approaches to achieve a more accurate diagnosis, as well as guidance before and after surgery. In this work, we induced human induction of pluripotent stem cell into neural progenitor cells (NPCs) and synthesized nanoprobes labeled with enhanced green fluorescent protein (EGFP, abbreviated as MFe3O4-labeled EGFP-NPCs) for photothermal therapy. Nanoprobes carried by NPCs can effectively penetrate the BBB and target glioma for the purpose of magnetic resonance imaging and guiding surgery. More importantly, MFe3O4-labeled EGFP-NPCs can effectively induce local photothermal therapy, conduct preoperative tumor therapy, and inhibit the recurrence of postoperative glioma. This work shows that MFe3O4-labeled EGFP-NPCs is a promising nanoplatform for glioma diagnosis, accurate imaging-guided surgery, and effective photothermal therapy.
Collapse
Affiliation(s)
- Qing Tao
- Department of Radiology, The First Affiliated Hospital of Soochow University, Suzhou 215000, Jiangsu, China
- Institute of Medical Imaging, Soochow University, Suzhou 215000, Jiangsu, China
| | - Shuang Yang
- Department of Radiology, The First Affiliated Hospital of Soochow University, Suzhou 215000, Jiangsu, China
- Institute of Medical Imaging, Soochow University, Suzhou 215000, Jiangsu, China
| | - Sheng Wang
- Department of Radiology, The First Affiliated Hospital of Soochow University, Suzhou 215000, Jiangsu, China
- Institute of Medical Imaging, Soochow University, Suzhou 215000, Jiangsu, China
| | - Yiwen Yang
- Department of Radiology, The First Affiliated Hospital of Soochow University, Suzhou 215000, Jiangsu, China
- Institute of Medical Imaging, Soochow University, Suzhou 215000, Jiangsu, China
| | - Shuang Yu
- Suzhou Institute of Biomedical Engineering and Technology, Chinese Academy of Sciences, Suzhou 215163, China
| | - Yue Pan
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangdong-Hong Kong Joint Laboratory for RNA Medicine, Medical Research Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, China
| | - Yonggang Li
- Department of Radiology, The First Affiliated Hospital of Soochow University, Suzhou 215000, Jiangsu, China
- Institute of Medical Imaging, Soochow University, Suzhou 215000, Jiangsu, China
| | - Jingzhong Zhang
- Suzhou Institute of Biomedical Engineering and Technology, Chinese Academy of Sciences, Suzhou 215163, China
| | - Chunhong Hu
- Department of Radiology, The First Affiliated Hospital of Soochow University, Suzhou 215000, Jiangsu, China
- Institute of Medical Imaging, Soochow University, Suzhou 215000, Jiangsu, China
| |
Collapse
|
35
|
Rudà R, Horbinski C, van den Bent M, Preusser M, Soffietti R. IDH inhibition in gliomas: from preclinical models to clinical trials. Nat Rev Neurol 2024; 20:395-407. [PMID: 38760442 DOI: 10.1038/s41582-024-00967-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/26/2024] [Indexed: 05/19/2024]
Abstract
Gliomas are the most common malignant primary brain tumours in adults and cannot usually be cured with standard cancer treatments. Gliomas show intratumoural and intertumoural heterogeneity at the histological and molecular levels, and they frequently contain mutations in the isocitrate dehydrogenase 1 (IDH1) or IDH2 gene. IDH-mutant adult-type diffuse gliomas are subdivided into grade 2, 3 or 4 IDH-mutant astrocytomas and grade 2 or 3 IDH-mutant, 1p19q-codeleted oligodendrogliomas. The product of the mutated IDH genes, D-2-hydroxyglutarate (D-2-HG), induces global DNA hypermethylation and interferes with immunity, leading to stimulation of tumour growth. Selective inhibitors of mutant IDH, such as ivosidenib and vorasidenib, have been shown to reduce D-2-HG levels and induce cellular differentiation in preclinical models and to induce MRI-detectable responses in early clinical trials. The phase III INDIGO trial has demonstrated superiority of vorasidenib, a brain-penetrant pan-mutant IDH inhibitor, over placebo in people with non-enhancing grade 2 IDH-mutant gliomas following surgery. In this Review, we describe the pathway of development of IDH inhibitors in IDH-mutant low-grade gliomas from preclinical models to clinical trials. We discuss the practice-changing implications of the INDIGO trial and consider new avenues of investigation in the field of IDH-mutant gliomas.
Collapse
Affiliation(s)
- Roberta Rudà
- Division of Neuro-Oncology, Department of Neuroscience 'Rita Levi Montalcini', University of Turin, Turin, Italy.
| | - Craig Horbinski
- Department of Pathology, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
- Department of Neurological Surgery, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Martin van den Bent
- Brain Tumour Center at Erasmus MC Cancer Institute, Rotterdam, The Netherlands
| | - Matthias Preusser
- Division of Oncology, Department of Medicine I, Medical University of Vienna, Vienna, Austria
| | - Riccardo Soffietti
- Division of Neuro-Oncology, Department of Neuroscience 'Rita Levi Montalcini', University of Turin, Turin, Italy
| |
Collapse
|
36
|
Picca A, Touat M, Belin L, Gourmelon C, Harlay V, Cuzzubbo S, Cohen-Jonathan Moyal E, Bronnimann C, Di Stefano AL, Laurent I, Lerond J, Carpentier C, Bielle F, Ducray F, Dehais C. REVOLUMAB: A phase II trial of nivolumab in recurrent IDH mutant high-grade gliomas. Eur J Cancer 2024; 202:114034. [PMID: 38537315 DOI: 10.1016/j.ejca.2024.114034] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2024] [Accepted: 03/20/2024] [Indexed: 04/21/2024]
Abstract
BACKGROUND Novel effective treatments are needed for recurrent IDH mutant high-grade gliomas (IDHm HGGs). The aim of the multicentric, single-arm, phase II REVOLUMAB trial (NCT03925246) was to assess the efficacy and safety of the anti-PD1 Nivolumab in patients with recurrent IDHm HGGs. PATIENTS AND METHODS Adult patients with IDHm WHO grade 3-4 gliomas recurring after radiotherapy and ≥ 1 line of alkylating chemotherapy were treated with intravenous Nivolumab until end of treatment (12 months), progression, unacceptable toxicity, or death. The primary endpoint was the 24-week progression-free survival rate (24w-PFS) according to RANO criteria. RESULTS From July 2019 to June 2020, 39 patients with recurrent IDHm HGGs (twenty-one grade 3, thirteen grade 4, five grade 2 with radiological evidence of anaplastic transformation; 39% 1p/19q codeleted) were enrolled. Median time since diagnosis was 5.7 years, and the median number of previous systemic treatments was two. The 24w-PFS was 28.2% (11/39, CI95% 15-44.9%). Median PFS and OS were 1.84 (CI95% 1.81-5.89) and 14.7 months (CI95% 9.18-NR), respectively. Four patients (10.3%) achieved partial response according to RANO criteria. There were no significant differences in clinical or histomolecular features between responders and non-responders. The safety profile of Nivolumab was consistent with prior studies. CONCLUSIONS We report the results of the first trial of immune checkpoint inhibitors in IDHm gliomas. Nivolumab failed to achieve its primary endpoint. However, treatment was well tolerated, and long-lasting responses were observed in a subset of patients, supporting further evaluation in combination with other agents (e.g. IDH inhibitors).
Collapse
Affiliation(s)
- Alberto Picca
- Service de Neuro-oncologie, Institut de Neurologie, AP-HP, Hôpital de la Pitié-Salpêtrière, Paris, France; Sorbonne Université, Inserm, CNRS, UMR S 1127, Paris Brain Institute (ICM), Paris, France
| | - Mehdi Touat
- Service de Neuro-oncologie, Institut de Neurologie, AP-HP, Hôpital de la Pitié-Salpêtrière, Paris, France; Sorbonne Université, Inserm, CNRS, UMR S 1127, Paris Brain Institute (ICM), Paris, France
| | - Lisa Belin
- Sorbonne Université, INSERM, Institut Pierre Louis d'Epidémiologie et de Santé Publique, Département de Santé Publique, Unité de Recherche Clinique PSL-CFX, CIC-1901, AP-HP, Hôpital de la Pitié-Salpêtrière, Paris, France
| | - Carole Gourmelon
- Department of Medical Oncology, Institut de Cancérologie de l'Ouest, Nantes, France
| | - Vincent Harlay
- Department of Neuro-Oncology, AP-HM, University Hospital Timone, Marseille, France
| | - Stefania Cuzzubbo
- Department of Neurology, Hôpital Saint-Louis, Université Paris Cité, APHP, Paris, France
| | - Elizabeth Cohen-Jonathan Moyal
- Department of Radiotherapy, Oncopole Claudius Regaud, Cancer University Institute of Toulouse, Oncopole Paul Sabatier University, Toulouse III, Cancer Research Center of Toulouse (CRCT), Toulouse, France
| | - Charlotte Bronnimann
- Hôpital Saint-André Bordeaux, Centre Hospitalier Universitaire, Service d'Oncologie, 33000 Bordeaux, France
| | - Anna Luisa Di Stefano
- Department of Neurology, Foch Hospital, 92150 Suresnes, France; Division of Neurosurgery, Spedali Riuniti di Livorno-USL Toscana Nord-Ovest, 57124 Livorno, Italy
| | - Isaura Laurent
- Unité de Recherche Clinique PSL-CFX, CIC-1901, AP-HP, Hôpital de la Pitié-Salpêtrière, Paris, France
| | - Julie Lerond
- Sorbonne Université, Inserm, CNRS, UMR S 1127, Paris Brain Institute (ICM), Paris, France
| | - Catherine Carpentier
- Sorbonne Université, Inserm, CNRS, UMR S 1127, Paris Brain Institute (ICM), Paris, France
| | - Franck Bielle
- Sorbonne Université, Inserm, CNRS, UMR S 1127, Paris Brain Institute (ICM), Paris, France; Department of Neuropathology, AP-HP, Hôpital de la Pitié-Salpêtrière, Paris, France
| | - François Ducray
- Department of Neuro-Oncology, East Group Hospital, Hospices Civils de Lyon, Lyon, France
| | - Caroline Dehais
- Service de Neuro-oncologie, Institut de Neurologie, AP-HP, Hôpital de la Pitié-Salpêtrière, Paris, France; Sorbonne Université, Inserm, CNRS, UMR S 1127, Paris Brain Institute (ICM), Paris, France.
| |
Collapse
|
37
|
Wu G, Shi Z, Li Z, Xie X, Tang Q, Zhu J, Yang Z, Wang Y, Wu J, Yu J. Study of radiochemotherapy decision-making for young high-risk low-grade glioma patients using a macroscopic and microscopic combined radiomics model. Eur Radiol 2024; 34:2861-2872. [PMID: 37889272 DOI: 10.1007/s00330-023-10378-9] [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: 06/28/2023] [Revised: 09/06/2023] [Accepted: 09/16/2023] [Indexed: 10/28/2023]
Abstract
OBJECTIVES As a few types of glioma, young high-risk low-grade gliomas (HRLGGs) have higher requirements for postoperative quality of life. Although adjuvant chemotherapy with delayed radiotherapy is the first treatment strategy for HRLGGs, not all HRLGGs benefit from it. Accurate assessment of chemosensitivity in HRLGGs is vital for making treatment choices. This study developed a multimodal fusion radiomics (MFR) model to support radiochemotherapy decision-making for HRLGGs. METHODS A MFR model combining macroscopic MRI and microscopic pathological images was proposed. Multiscale features including macroscopic tumor structure and microscopic histological layer and nuclear information were grabbed by unique paradigm, respectively. Then, these features were adaptively incorporated into the MFR model through attention mechanism to predict the chemosensitivity of temozolomide (TMZ) by means of objective response rate and progression free survival (PFS). RESULTS Macroscopic tumor texture complexity and microscopic nuclear size showed significant statistical differences (p < 0.001) between sensitivity and insensitivity groups. The MFR model achieved stable prediction results, with an area under the curve of 0.950 (95% CI: 0.942-0.958), sensitivity of 0.833 (95% CI: 0.780-0.848), specificity of 0.929 (95% CI: 0.914-0.936), positive predictive value of 0.833 (95% CI: 0.811-0.860), and negative predictive value of 0.929 (95% CI: 0.914-0.934). The predictive efficacy of MFR was significantly higher than that of the reported molecular markers (p < 0.001). MFR was also demonstrated to be a predictor of PFS. CONCLUSIONS A MFR model including radiomics and pathological features predicts accurately the response postoperative TMZ treatment. CLINICAL RELEVANCE STATEMENT Our MFR model could identify young high-risk low-grade glioma patients who can have the most benefit from postoperative upfront temozolomide (TMZ) treatment. KEY POINTS • Multimodal radiomics is proposed to support the radiochemotherapy of glioma. • Some macro and micro image markers related to tumor chemotherapy sensitivity are revealed. • The proposed model surpasses reported molecular markers, with a promising area under the curve (AUC) of 0.95.
Collapse
Affiliation(s)
- Guoqing Wu
- School of Information Science and Technology, Fudan University, Shanghai, China
- Key Laboratory of Medical Imaging Computing and Computer Assisted Intervention of Shanghai, Shanghai, China
| | - Zhifeng Shi
- Department of Neurosurgery, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai, China
- National Center for Neurological Disorders, Shanghai, China
- Shanghai Key Laboratory of Brain Function Restoration and Neural Regeneration, Shanghai, China
- Neurosurgical Institute of Fudan University, Shanghai, China
| | - Zeyang Li
- Department of Neurosurgery, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai, China
- National Center for Neurological Disorders, Shanghai, China
- Shanghai Key Laboratory of Brain Function Restoration and Neural Regeneration, Shanghai, China
- Neurosurgical Institute of Fudan University, Shanghai, China
| | - Xuan Xie
- School of Information Science and Technology, Fudan University, Shanghai, China
- Key Laboratory of Medical Imaging Computing and Computer Assisted Intervention of Shanghai, Shanghai, China
| | - Qisheng Tang
- Department of Neurosurgery, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai, China
- National Center for Neurological Disorders, Shanghai, China
- Shanghai Key Laboratory of Brain Function Restoration and Neural Regeneration, Shanghai, China
- Neurosurgical Institute of Fudan University, Shanghai, China
| | - Jingjing Zhu
- Department of Pathology, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai, China
| | - Zhong Yang
- Department of Radiology, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai, China
| | - Yuanyuan Wang
- School of Information Science and Technology, Fudan University, Shanghai, China
- Key Laboratory of Medical Imaging Computing and Computer Assisted Intervention of Shanghai, Shanghai, China
| | - Jinsong Wu
- Department of Neurosurgery, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai, China.
- National Center for Neurological Disorders, Shanghai, China.
- Shanghai Key Laboratory of Brain Function Restoration and Neural Regeneration, Shanghai, China.
- Neurosurgical Institute of Fudan University, Shanghai, China.
| | - Jinhua Yu
- School of Information Science and Technology, Fudan University, Shanghai, China.
- Key Laboratory of Medical Imaging Computing and Computer Assisted Intervention of Shanghai, Shanghai, China.
- The AI Lab of Huashan Hospital, Fudan University, Shanghai, China.
| |
Collapse
|
38
|
Hu X, Zhao M, Bai M, Xue Z, Wang F, Zhu Z, Yu J, Yue J. PARP inhibitor plus radiotherapy reshape the immune suppressive microenvironment and potentiate the efficacy of immune checkpoint inhibitors in tumors with IDH1 mutation. Cancer Lett 2024; 586:216676. [PMID: 38278469 DOI: 10.1016/j.canlet.2024.216676] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2023] [Revised: 01/22/2024] [Accepted: 01/23/2024] [Indexed: 01/28/2024]
Abstract
Isocitrate dehydrogenase 1 mutant (IDH1mut) tumors respond poorly to immunotherapy, but are more sensitive to chemoradiotherapy and poly (ADP-ribose) polymerase inhibition (PARPi). Accordingly, some efforts have aimed to capitalize on the IDH1 mutation rather than reverse it. Moreover, radiotherapy (RT) and PARPi can stimulate antitumor immunity, raising the possibility of reversing the immunosuppression caused by IDH1 mutation while killing the tumor. To assess this possibility, we treated IDH1mut tumors and cells with RT + PARPi. RT + PARPi showed enhanced efficacy over either modality alone both in vitro and in vivo. RT + PARPi induced more DNA damage and activated the cGAS-STING pathway more. IFNβ, CXCL10, and CCL5 were also more highly expressed at both the mRNA and protein levels. In two different tumor models, RT + PARPi increased infiltration and cytolytic function of CD8+ T cells, with one model also showing increased CD8+T cell proliferation. RT+PARPi also increased PD-L1 expression and enhanced checkpoint inhibition. Knocking out cGAS reversed the increased CD8+ T cell infiltration and the antitumor effect of RT+PARPi. We conclude that RT + PARPi reshapes the IDH1mut tumor immunosuppressive microenvironment, thereby augmenting checkpoint inhibition.
Collapse
Affiliation(s)
- Xiaoyu Hu
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong, China
| | - Mengyu Zhao
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong, China; Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Menglin Bai
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong, China; Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Zhuang Xue
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong, China
| | - Fei Wang
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong, China
| | - Ziyuan Zhu
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong, China
| | - Jinming Yu
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong, China; Cheeloo College of Medicine, Shandong University, Jinan, China.
| | - Jinbo Yue
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong, China; Cheeloo College of Medicine, Shandong University, Jinan, China.
| |
Collapse
|
39
|
Nie J, Li Q, Yin H, Yang J, Li M, Li Q, Fan X, Zhao Q, Wen Z. NPS-2143 inhibit glioma progression by suppressing autophagy through mediating AKT-mTOR pathway. J Cell Mol Med 2024; 28:e18221. [PMID: 38509759 PMCID: PMC10955153 DOI: 10.1111/jcmm.18221] [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: 11/13/2023] [Revised: 02/16/2024] [Accepted: 02/22/2024] [Indexed: 03/22/2024] Open
Abstract
Gliomas are the most common tumours in the central nervous system. In the present study, we aimed to find a promising anti-glioma compound and investigate the underlying molecular mechanism. Glioma cells were subjected to the 50 candidate compounds at a final concentration of 10 μM for 72 h, and CCK-8 was used to evaluate their cytotoxicity. NPS-2143, an antagonist of calcium-sensing receptor (CASR), was selected for further study due to its potent cytotoxicity to glioma cells. Our results showed that NPS-2143 could inhibit the proliferation of glioma cells and induce G1 phase cell cycle arrest. Meanwhile, NPS-2143 could induce glioma cell apoptosis by increasing the caspase-3/6/9 activity. NPS-2143 impaired the immigration and invasion ability of glioma cells by regulating the epithelial-mesenchymal transition process. Mechanically, NPS-2143 could inhibit autophagy by mediating the AKT-mTOR pathway. Bioinformatic analysis showed that the prognosis of glioma patients with low expression of CASR mRNA was better than those with high expression of CASR mRNA. Gene set enrichment analysis showed that CASR was associated with cell adhesion molecules and lysosomes in glioma. The nude mice xenograft model showed NPS-2143 could suppress glioma growth in vivo. In conclusion, NPS-2143 can suppress the glioma progression by inhibiting autophagy.
Collapse
Affiliation(s)
- Jia‐Li Nie
- Department of PharmacyAffiliated Hospital of Guizhou Medical UniversityGuiyangP.R. China
- College of PharmacyGuizhou Medical UniversityGuiyangP.R. China
| | - Qi Li
- Department of PharmacyAffiliated Hospital of Guizhou Medical UniversityGuiyangP.R. China
- College of PharmacyGuizhou Medical UniversityGuiyangP.R. China
| | - Hai‐Tang Yin
- Department of PharmacyAffiliated Hospital of Guizhou Medical UniversityGuiyangP.R. China
- College of PharmacyGuizhou Medical UniversityGuiyangP.R. China
| | - Ji‐Hong Yang
- Department of PharmacyAffiliated Hospital of Guizhou Medical UniversityGuiyangP.R. China
- College of PharmacyGuizhou Medical UniversityGuiyangP.R. China
| | - Ming Li
- Department of PharmacyAffiliated Hospital of Guizhou Medical UniversityGuiyangP.R. China
- College of PharmacyGuizhou Medical UniversityGuiyangP.R. China
| | - Qin Li
- Centre of Clinical TrialsAffiliated Hospital of Guizhou Medical UniversityGuiyangP.R. China
| | - Xing‐Hua Fan
- Department of PharmacyAffiliated Hospital of Guizhou Medical UniversityGuiyangP.R. China
- College of PharmacyGuizhou Medical UniversityGuiyangP.R. China
| | - Qing‐Qing Zhao
- Clinical Research CenterAffiliated Hospital of Guizhou Medical UniversityGuiyangP.R. China
| | - Zhi‐Peng Wen
- Department of PharmacyAffiliated Hospital of Guizhou Medical UniversityGuiyangP.R. China
- College of PharmacyGuizhou Medical UniversityGuiyangP.R. China
| |
Collapse
|
40
|
Sasaki H, Kitamura Y, Toda M, Hirose Y, Yoshida K. Oligodendroglioma, IDH-mutant and 1p/19q-codeleted-prognostic factors, standard of care and chemotherapy, and future perspectives with neoadjuvant strategy. Brain Tumor Pathol 2024; 41:43-49. [PMID: 38564040 DOI: 10.1007/s10014-024-00480-1] [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: 01/10/2024] [Accepted: 03/25/2024] [Indexed: 04/04/2024]
Abstract
Oligodendroglioma, IDH-mutant and 1p/19q-codeleted is known for their relative chemosensitivity and indolent clinical course among diffuse gliomas of adult type. Based on the data from phase 3 clinical trials, the standard of post-surgical care for those tumors is considered to be initial chemoradiotherapy regardless of histopathological grade, particularly with PCV. However, partly due to its renewed definition in late years, prognostic factors in patients with those tumors are not well established. Moreover, the survival rate declines over 15 years, with only a 37% OS rate at 20 years for grade 3 tumors, even with the current standard of care. Given that most of this disease occurs in young or middle-aged adults, further improvements in treatment and management are necessary. Here, we discuss prognostic factors, standard of care and chemotherapy, and future perspectives with neoadjuvant strategy in those tumors.
Collapse
Affiliation(s)
- Hikaru Sasaki
- Department of Neurosurgery, Tokyo Dental College Ichikawa General Hospital, 5-11-13 Sugano, Ichikawa, Chiba, 272-8523, Japan.
- Department of Neurosurgery, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo, 160-8582, Japan.
| | - Yohei Kitamura
- Department of Neurosurgery, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo, 160-8582, Japan
| | - Masahiro Toda
- Department of Neurosurgery, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo, 160-8582, Japan
| | - Yuichi Hirose
- Department of Neurosurgery, Fujita Health University School of Medicine, 1-98 Dengakugakubo, Kutsukakecho, Toyoake, Aichi, 470-1192, Japan
| | - Kazunari Yoshida
- Department of Neurosurgery, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo, 160-8582, Japan
| |
Collapse
|
41
|
Wei J, Li Y, Zhou W, Ma X, Hao J, Wen T, Li B, Jin T, Hu M. The construction of a novel prognostic prediction model for glioma based on GWAS-identified prognostic-related risk loci. Open Med (Wars) 2024; 19:20240895. [PMID: 38584840 PMCID: PMC10996933 DOI: 10.1515/med-2024-0895] [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: 08/03/2023] [Revised: 11/17/2023] [Accepted: 12/08/2023] [Indexed: 04/09/2024] Open
Abstract
Backgrounds Glioma is a highly malignant brain tumor with a grim prognosis. Genetic factors play a role in glioma development. While some susceptibility loci associated with glioma have been identified, the risk loci associated with prognosis have received less attention. This study aims to identify risk loci associated with glioma prognosis and establish a prognostic prediction model for glioma patients in the Chinese Han population. Methods A genome-wide association study (GWAS) was conducted to identify risk loci in 484 adult patients with glioma. Cox regression analysis was performed to assess the association between GWAS-risk loci and overall survival as well as progression-free survival in glioma. The prognostic model was constructed using LASSO Cox regression analysis and multivariate Cox regression analysis. The nomogram model was constructed based on the single nucleotide polymorphism (SNP) classifier and clinical indicators, enabling the prediction of survival rates at 1-year, 2-year, and 3-year intervals. Additionally, the receiver operator characteristic (ROC) curve was employed to evaluate the prediction value of the nomogram. Finally, functional enrichment and tumor-infiltrating immune analyses were conducted to examine the biological functions of the associated genes. Results Our study found suggestive evidence that a total of 57 SNPs were correlated with glioma prognosis (p < 5 × 10-5). Subsequently, we identified 25 SNPs with the most significant impact on glioma prognosis and developed a prognostic model based on these SNPs. The 25 SNP-based classifier and clinical factors (including age, gender, surgery, and chemotherapy) were identified as independent prognostic risk factors. Subsequently, we constructed a prognostic nomogram based on independent prognostic factors to predict individualized survival. ROC analyses further showed that the prediction accuracy of the nomogram (AUC = 0.956) comprising the 25 SNP-based classifier and clinical factors was significantly superior to that of each individual variable. Conclusion We identified a SNP classifier and clinical indicators that can predict the prognosis of glioma patients and established a prognostic prediction model in the Chinese Han population. This study offers valuable insights for clinical practice, enabling improved evaluation of patients' prognosis and informing treatment options.
Collapse
Affiliation(s)
- Jie Wei
- College of Life Science, Northwest University, Xi’an 710127, Shaanxi, China
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, Northwest University, Xi’an 710069, Shaanxi, China
- Shaanxi Provincial Key Laboratory of Biotechnology, Northwest University, Xi’an710069, Shaanxi, China
| | - Yujie Li
- College of Life Science, Northwest University, Xi’an 710127, Shaanxi, China
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, Northwest University, Xi’an 710069, Shaanxi, China
- Shaanxi Provincial Key Laboratory of Biotechnology, Northwest University, Xi’an710069, Shaanxi, China
| | - Wenqian Zhou
- College of Life Science, Northwest University, Xi’an 710127, Shaanxi, China
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, Northwest University, Xi’an 710069, Shaanxi, China
- Shaanxi Provincial Key Laboratory of Biotechnology, Northwest University, Xi’an710069, Shaanxi, China
| | - Xiaoya Ma
- College of Life Science, Northwest University, Xi’an 710127, Shaanxi, China
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, Northwest University, Xi’an 710069, Shaanxi, China
- Shaanxi Provincial Key Laboratory of Biotechnology, Northwest University, Xi’an710069, Shaanxi, China
| | - Jie Hao
- College of Life Science, Northwest University, Xi’an 710127, Shaanxi, China
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, Northwest University, Xi’an 710069, Shaanxi, China
- Shaanxi Provincial Key Laboratory of Biotechnology, Northwest University, Xi’an710069, Shaanxi, China
| | - Ting Wen
- College of Life Science, Northwest University, Xi’an 710127, Shaanxi, China
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, Northwest University, Xi’an 710069, Shaanxi, China
- Shaanxi Provincial Key Laboratory of Biotechnology, Northwest University, Xi’an710069, Shaanxi, China
| | - Bin Li
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, Northwest University, Xi’an 710069, Shaanxi, China
- Biomedicine Key Laboratory of Shaanxi Province, Northwest University, Xi’an710069, Shaanxi, China
| | - Tianbo Jin
- College of Life Science, Northwest University, Xi’an 710127, Shaanxi, China
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, Northwest University, Xi’an 710069, Shaanxi, China
- Shaanxi Provincial Key Laboratory of Biotechnology, Northwest University, Xi’an710069, Shaanxi, China
| | - Mingjun Hu
- College of Life Science, Northwest University, Xi’an 710127, Shaanxi, China
- School of Medicine, Northwest University, Xi’an710127, Shaanxi, China
- Department of Neurosurgery, Xi’an Chest Hospital, Xi’an710100, Shaanxi, China
- Department of Neurosurgery, Xi’an Chang’an District Hospital, Xi’an710118, Shaanxi, China
| |
Collapse
|
42
|
Malta TM, Sabedot TS, Morosini NS, Datta I, Garofano L, Vallentgoed W, Varn FS, Aldape K, D'Angelo F, Bakas S, Barnholtz-Sloan JS, Gan HK, Hasanain M, Hau AC, Johnson KC, Cazacu S, deCarvalho AC, Khasraw M, Kocakavuk E, Kouwenhoven MC, Migliozzi S, Niclou SP, Niers JM, Ormond DR, Paek SH, Reifenberger G, Sillevis Smitt PA, Smits M, Stead LF, van den Bent MJ, Van Meir EG, Walenkamp A, Weiss T, Weller M, Westerman BA, Ylstra B, Wesseling P, Lasorella A, French PJ, Poisson LM, Verhaak RG, Iavarone A, Noushmehr H. The Epigenetic Evolution of Glioma Is Determined by the IDH1 Mutation Status and Treatment Regimen. Cancer Res 2024; 84:741-756. [PMID: 38117484 PMCID: PMC10911804 DOI: 10.1158/0008-5472.can-23-2093] [Citation(s) in RCA: 20] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2023] [Revised: 09/15/2023] [Accepted: 12/13/2023] [Indexed: 12/21/2023]
Abstract
Tumor adaptation or selection is thought to underlie therapy resistance in glioma. To investigate longitudinal epigenetic evolution of gliomas in response to therapeutic pressure, we performed an epigenomic analysis of 132 matched initial and recurrent tumors from patients with IDH-wildtype (IDHwt) and IDH-mutant (IDHmut) glioma. IDHwt gliomas showed a stable epigenome over time with relatively low levels of global methylation. The epigenome of IDHmut gliomas showed initial high levels of genome-wide DNA methylation that was progressively reduced to levels similar to those of IDHwt tumors. Integration of epigenomics, gene expression, and functional genomics identified HOXD13 as a master regulator of IDHmut astrocytoma evolution. Furthermore, relapse of IDHmut tumors was accompanied by histologic progression that was associated with survival, as validated in an independent cohort. Finally, the initial cell composition of the tumor microenvironment varied between IDHwt and IDHmut tumors and changed differentially following treatment, suggesting increased neoangiogenesis and T-cell infiltration upon treatment of IDHmut gliomas. This study provides one of the largest cohorts of paired longitudinal glioma samples with epigenomic, transcriptomic, and genomic profiling and suggests that treatment of IDHmut glioma is associated with epigenomic evolution toward an IDHwt-like phenotype. SIGNIFICANCE Standard treatments are related to loss of DNA methylation in IDHmut glioma, resulting in epigenetic activation of genes associated with tumor progression and alterations in the microenvironment that resemble treatment-naïve IDHwt glioma.
Collapse
Affiliation(s)
- Tathiane M. Malta
- School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo, Ribeirão Preto, São Paulo, Brazil
| | - Thais S. Sabedot
- Hermelin Brain Tumor Center, Henry Ford Health System, Detroit, Michigan
| | | | - Indrani Datta
- Hermelin Brain Tumor Center, Henry Ford Health System, Detroit, Michigan
| | - Luciano Garofano
- Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Miami, Florida
| | - Wies Vallentgoed
- Neurology Department, The Brain Tumour Center, Erasmus MC Cancer Institute, Rotterdam, the Netherlands
| | - Frederick S. Varn
- The Jackson Laboratory for Genomic Medicine, Farmington, Connecticut
| | | | - Fulvio D'Angelo
- Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Miami, Florida
- Department of Neurological Surgery, University of Miami Miller School of Medicine, Miami, Florida
| | - Spyridon Bakas
- Center for Biomedical Image Computing and Analytics, University of Pennsylvania, Philadelphia, Pennsylvania
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
- Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | | | - Hui K. Gan
- Olivia Newton-John Cancer Research Institute, Austin Health, Heidelberg, Melbourne, Australia
| | - Mohammad Hasanain
- Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Miami, Florida
| | | | - Kevin C. Johnson
- Department of Neurosurgery, Yale School of Medicine, New Haven, Connecticut
| | - Simona Cazacu
- Hermelin Brain Tumor Center, Henry Ford Health System, Detroit, Michigan
| | - Ana C. deCarvalho
- Hermelin Brain Tumor Center, Henry Ford Health System, Detroit, Michigan
| | | | - Emre Kocakavuk
- Department of Neurosurgery, Yale School of Medicine, New Haven, Connecticut
- Department of Hematology and Stem Cell Transplantation, West German Cancer Center (WTZ), National Center for Tumor Diseases (NCT) West, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Mathilde C.M. Kouwenhoven
- Department of Neurology, Cancer Center Amsterdam, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, the Netherlands
| | - Simona Migliozzi
- Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Miami, Florida
| | | | - Johanna M. Niers
- Department of Neurology, Cancer Center Amsterdam, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, the Netherlands
| | - D. Ryan Ormond
- University of Colorado School of Medicine, Department of Neurosurgery, Aurora, Colorado
| | - Sun Ha Paek
- Department of Neurosurgery, Cancer Research Institute, Hypoxia Ischemia Disease Institute, Seoul National University, Seoul, Republic of Korea (South)
| | - Guido Reifenberger
- Institute of Neuropathology, Heinrich Heine University, Dusseldorf, Germany
| | - Peter A. Sillevis Smitt
- Department of Neurology, Cancer Center Amsterdam, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, the Netherlands
- The Brain Tumour Centre, Erasmus MC Cancer Institute, Rotterdam, the Netherlands
| | - Marion Smits
- Department of Radiology and Nuclear Medicine, Erasmus MC, Rotterdam, the Netherlands
| | - Lucy F. Stead
- Leeds Institute of Medical Research, University of Leeds, Leeds, United Kingdom
| | - Martin J. van den Bent
- Department of Neurology, Cancer Center Amsterdam, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, the Netherlands
- The Brain Tumour Centre, Erasmus MC Cancer Institute, Rotterdam, the Netherlands
| | - Erwin G. Van Meir
- Department of Neurosurgery and O'Neal Comprehensive Cancer Center, University of Alabama at Birmingham, Birmingham, Alabama
| | | | - Tobias Weiss
- Department of Neurology, University Hospital and University of Zurich, Zurich, Switzerland
| | - Michael Weller
- Department of Neurology, University Hospital and University of Zurich, Zurich, Switzerland
| | - Bart A. Westerman
- Department of Neurology, Cancer Center Amsterdam, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, the Netherlands
| | - Bauke Ylstra
- Department of Pathology, Cancer Center Amsterdam, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, the Netherlands
| | - Pieter Wesseling
- Department of Pathology, Cancer Center Amsterdam, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, the Netherlands
- Brain Tumor Center Amsterdam, Cancer Center Amsterdam, Amsterdam UMC, VU University Medical Center, Amsterdam, the Netherlands
- Laboratory for Childhood Cancer Pathology, Princess Máxima Center for Pediatric Oncology, Utrecht, the Netherlands
| | - Anna Lasorella
- Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Miami, Florida
- Department of Biochemistry and Molecular Biology, University of Miami Miller School of Medicine, Miami, Florida
| | - Pim J. French
- Neurology Department, The Brain Tumour Center, Erasmus MC Cancer Institute, Rotterdam, the Netherlands
| | - Laila M. Poisson
- Hermelin Brain Tumor Center, Henry Ford Health System, Detroit, Michigan
| | - Roel G.W. Verhaak
- Department of Neurosurgery, Yale School of Medicine, New Haven, Connecticut
- Department of Neurosurgery, Amsterdam University Medical Center, Amsterdam, the Netherlands
| | - Antonio Iavarone
- Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Miami, Florida
- Department of Neurological Surgery, University of Miami Miller School of Medicine, Miami, Florida
| | - Houtan Noushmehr
- Hermelin Brain Tumor Center, Henry Ford Health System, Detroit, Michigan
| |
Collapse
|
43
|
Mohamed AA, Alshaibi R, Faragalla S, Mohamed Y, Lucke-Wold B. Updates on management of gliomas in the molecular age. World J Clin Oncol 2024; 15:178-194. [PMID: 38455131 PMCID: PMC10915945 DOI: 10.5306/wjco.v15.i2.178] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/28/2023] [Revised: 01/06/2024] [Accepted: 01/25/2024] [Indexed: 02/20/2024] Open
Abstract
Gliomas are primary brain tumors derived from glial cells of the central nervous system, afflicting both adults and children with distinct characteristics and therapeutic challenges. Recent developments have ushered in novel clinical and molecular prognostic factors, reshaping treatment paradigms based on classification and grading, determined by histological attributes and cellular lineage. This review article delves into the diverse treatment modalities tailored to the specific grades and molecular classifications of gliomas that are currently being discussed and used clinically in the year 2023. For adults, the therapeutic triad typically consists of surgical resection, chemotherapy, and radiotherapy. In contrast, pediatric gliomas, due to their diversity, require a more tailored approach. Although complete tumor excision can be curative based on the location and grade of the glioma, certain non-resectable cases demand a chemotherapy approach usually involving, vincristine and carboplatin. Additionally, if surgery or chemotherapy strategies are unsuccessful, Vinblastine can be used. Despite recent advancements in treatment methodologies, there remains a need of exploration in the literature, particularly concerning the efficacy of treatment regimens for isocitrate dehydrogenase type mutant astrocytomas and fine-tuned therapeutic approaches tailored for pediatric cohorts. This review article explores into the therapeutic modalities employed for both adult and pediatric gliomas in the context of their molecular classification.
Collapse
Affiliation(s)
- Ali Ahmed Mohamed
- Charles E. Schmidt College of Medicine, Florida Atlantic University, Boca Raton, FL 33431, United States
| | - Rakan Alshaibi
- Herbert Wertheim College of Medicine, Florida International University, Miami, FL 33199, United States
| | - Steven Faragalla
- Charles E. Schmidt College of Medicine, Florida Atlantic University, Boca Raton, FL 33431, United States
| | - Youssef Mohamed
- College of Osteopathic Medicine, Kansas City University, Joplin, MO 64804, United States
| | - Brandon Lucke-Wold
- Department of Neurosurgery, University of Florida, Gainesville, FL 32611, United States
| |
Collapse
|
44
|
Zheng S, Rammohan N, Sita T, Teo PT, Wu Y, Lesniak M, Sachdev S, Thomas TO. GlioPredictor: a deep learning model for identification of high-risk adult IDH-mutant glioma towards adjuvant treatment planning. Sci Rep 2024; 14:2126. [PMID: 38267516 PMCID: PMC10808248 DOI: 10.1038/s41598-024-51765-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2023] [Accepted: 01/09/2024] [Indexed: 01/26/2024] Open
Abstract
Identification of isocitrate dehydrogenase (IDH)-mutant glioma patients at high risk of early progression is critical for radiotherapy treatment planning. Currently tools to stratify risk of early progression are lacking. We sought to identify a combination of molecular markers that could be used to identify patients who may have a greater need for adjuvant radiation therapy machine learning technology. 507 WHO Grade 2 and 3 glioma cases from The Cancer Genome Atlas, and 1309 cases from AACR GENIE v13.0 datasets were studied for genetic disparities between IDH1-wildtype and IDH1-mutant cohorts, and between different age groups. Genetic features such as mutations and copy number variations (CNVs) correlated with IDH1 mutation status were selected as potential inputs to train artificial neural networks (ANNs) to predict IDH1 mutation status. Grade 2 and 3 glioma cases from the Memorial Sloan Kettering dataset (n = 404) and Grade 3 glioma cases with subtotal resection (STR) from Northwestern University (NU) (n = 21) were used to further evaluate the best performing ANN model as independent datasets. IDH1 mutation is associated with decreased CNVs of EGFR (21% vs. 3%), CDKN2A (20% vs. 6%), PTEN (14% vs. 1.7%), and increased percentage of mutations for TP53 (15% vs. 63%), and ATRX (10% vs. 54%), which were all statistically significant (p < 0.001). Age > 40 was unable to identify high-risk IDH1-mutant with early progression. A glioma early progression risk prediction (GlioPredictor) score generated from the best performing ANN model (6/6/6/6/2/1) with 6 inputs, including CNVs of EGFR, PTEN and CDKN2A, mutation status of TP53 and ATRX, patient's age can predict IDH1 mutation status with over 90% accuracy. The GlioPredictor score identified a subgroup of high-risk IDH1-mutant in TCGA and NU datasets with early disease progression (p = 0.0019, 0.0238, respectively). The GlioPredictor that integrates age at diagnosis, CNVs of EGFR, CDKN2A, PTEN and mutation status of TP53, and ATRX can identify a small cohort of IDH-mutant with high risk of early progression. The current version of GlioPredictor mainly incorporated clinically often tested genetic biomarkers. Considering complexity of clinical and genetic features that correlate with glioma progression, future derivatives of GlioPredictor incorporating more inputs can be a potential supplement for adjuvant radiotherapy patient selection of IDH-mutant glioma patients.
Collapse
Affiliation(s)
- Shuhua Zheng
- Department of Radiation Oncology, Northwestern University Feinberg School of Medicine, Chicago, IL, USA.
| | - Nikhil Rammohan
- Department of Radiation Oncology, Washington University School of Medicine, St. Louis, MO, USA
| | - Timothy Sita
- Department of Radiation Oncology, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - P Troy Teo
- Department of Radiation Oncology, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Yilin Wu
- Department of Mathematics, DigiPen Institute of Technology, Redmond, WA, USA
| | - Maciej Lesniak
- Department of Neurological Surgery, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Sean Sachdev
- Department of Radiation Oncology, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Tarita O Thomas
- Department of Radiation Oncology, Northwestern University Feinberg School of Medicine, Chicago, IL, USA.
- Department of Radiation Oncology, Northwestern Medical Group, Northwestern Memorial Hospital, Northwestern University Feinberg School of Medicine, Chicago, USA.
| |
Collapse
|
45
|
Chen J, Lu R, Peng Y, Lai Y, Cai Z, Zheng D, Xie A, Huang K, Liang C, Zhang H. MCU promotes the migration of glioma cells by activating p38 through TFEB-mediated autophagy. J Cancer 2024; 15:1257-1270. [PMID: 38356708 PMCID: PMC10861810 DOI: 10.7150/jca.89485] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2023] [Accepted: 12/13/2023] [Indexed: 02/16/2024] Open
Abstract
Changes in calcium signalling are crucial for the development of glioma cells. Whether mitochondrial calcium balance is involved in glial cell development is still unknown. Mitochondrial Calcium Uniporter (MCU) plays an important role in regulating glioma progression. In this work, we found that MCU and p38 expression were positively correlated with glioma grade and the degree tumour progression. MCU increases glioma cell migration by upregulating p38. Furthermore, p38 promotes glioma progression by activating Transcription Factor EB (TFEB)-mediated autophagy. Thus, MCU promotes glioma cell migration by activating autophagy in a p38/TFEB pathway-dependent manner, which provides a theoretical basis for new therapeutic targets for gliomas.
Collapse
Affiliation(s)
- Jialong Chen
- ✉ Corresponding authors: Jialong Chen, He Zhang; Dongguan Key Laboratory of Environmental Medicine, Affiliated Hospital of Guangdong Medical University, School of Public Health, Guangdong Medical University, Dongguan, 523808, P.R. China. Phone: +86-75922896573; E-mail: ;
| | | | | | | | | | | | | | | | | | - He Zhang
- Dongguan Key Laboratory of Environmental Medicine, The First Dongguan Affiliated Hospital, School of Public Health, Guangdong Medical University, Dongguan, 523808
| |
Collapse
|
46
|
Shikalov A, Koman I, Kogan NM. Targeted Glioma Therapy-Clinical Trials and Future Directions. Pharmaceutics 2024; 16:100. [PMID: 38258110 PMCID: PMC10820492 DOI: 10.3390/pharmaceutics16010100] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2023] [Revised: 01/05/2024] [Accepted: 01/08/2024] [Indexed: 01/24/2024] Open
Abstract
Glioblastoma multiforme (GBM) is the most common type of glioma, with a median survival of 14.6 months post-diagnosis. Understanding the molecular profile of such tumors allowed the development of specific targeted therapies toward GBM, with a major role attributed to tyrosine kinase receptor inhibitors and immune checkpoint inhibitors. Targeted therapeutics are drugs that work by specific binding to GBM-specific or overexpressed markers on the tumor cellular surface and therefore contain a recognition moiety linked to a cytotoxic agent, which produces an antiproliferative effect. In this review, we have summarized the available information on the targeted therapeutics used in clinical trials of GBM and summarized current obstacles and advances in targeted therapy concerning specific targets present in GBM tumor cells, outlined efficacy endpoints for major classes of investigational drugs, and discussed promising strategies towards an increase in drug efficacy in GBM.
Collapse
Affiliation(s)
| | | | - Natalya M. Kogan
- Department of Molecular Biology, Institute of Personalized and Translational Medicine, Ariel University, Ariel 40700, Israel; (A.S.); (I.K.)
| |
Collapse
|
47
|
Weller M, Felsberg J, Hentschel B, Gramatzki D, Kubon N, Wolter M, Reusche M, Roth P, Krex D, Herrlinger U, Westphal M, Tonn JC, Regli L, Maurage CA, von Deimling A, Pietsch T, Le Rhun E, Reifenberger G. Improved prognostic stratification of patients with isocitrate dehydrogenase-mutant astrocytoma. Acta Neuropathol 2024; 147:11. [PMID: 38183430 PMCID: PMC10771615 DOI: 10.1007/s00401-023-02662-1] [Citation(s) in RCA: 22] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2023] [Revised: 11/19/2023] [Accepted: 11/20/2023] [Indexed: 01/08/2024]
Abstract
Prognostic factors and standards of care for astrocytoma, isocitrate dehydrogenase (IDH)-mutant, CNS WHO grade 4, remain poorly defined. Here we sought to explore disease characteristics, prognostic markers, and outcome in patients with this newly defined tumor type. We determined molecular biomarkers and assembled clinical and outcome data in patients with IDH-mutant astrocytomas confirmed by central pathology review. Patients were identified in the German Glioma Network cohort study; additional cohorts of patients with CNS WHO grade 4 tumors were identified retrospectively at two sites. In total, 258 patients with IDH-mutant astrocytomas (114 CNS WHO grade 2, 73 CNS WHO grade 3, 71 CNS WHO grade 4) were studied. The median age at diagnosis was similar for all grades. Karnofsky performance status at diagnosis inversely correlated with CNS WHO grade (p < 0.001). Despite more intensive treatment upfront with higher grade, CNS WHO grade was strongly prognostic: median overall survival was not reached for grade 2 (median follow-up 10.4 years), 8.1 years (95% CI 5.4-10.8) for grade 3, and 4.7 years (95% CI 3.4-6.0) for grade 4. Among patients with CNS WHO grade 4 astrocytoma, median overall survival was 5.5 years (95% CI 4.3-6.7) without (n = 58) versus 1.8 years (95% CI 0-4.1) with (n = 12) homozygous CDKN2A deletion. Lower levels of global DNA methylation as detected by LINE-1 methylation analysis were strongly associated with CNS WHO grade 4 (p < 0.001) and poor outcome. MGMT promoter methylation status was not prognostic for overall survival. Histomolecular stratification based on CNS WHO grade, LINE-1 methylation level, and CDKN2A status revealed four subgroups of patients with significantly different outcomes. In conclusion, CNS WHO grade, global DNA methylation status, and CDKN2A homozygous deletion are prognostic in patients with IDH-mutant astrocytoma. Combination of these parameters allows for improved prediction of outcome. These data aid in designing upcoming trials using IDH inhibitors.
Collapse
Affiliation(s)
- Michael Weller
- Department of Neurology, University Hospital Zurich, Frauenklinikstrasse 26, 8091, Zurich, Switzerland.
- Department of Neurology, University of Zurich, Zurich, Switzerland.
| | - Jörg Felsberg
- Institute of Neuropathology, Heinrich Heine University, Medical Faculty, and University Hospital Düsseldorf, Düsseldorf, Germany
| | - Bettina Hentschel
- Institute for Medical Informatics, Statistics and Epidemiology, University Leipzig, Leipzig, Germany
| | - Dorothee Gramatzki
- Department of Neurology, University Hospital Zurich, Frauenklinikstrasse 26, 8091, Zurich, Switzerland
| | - Nadezhda Kubon
- Institute of Neuropathology, Heinrich Heine University, Medical Faculty, and University Hospital Düsseldorf, Düsseldorf, Germany
| | - Marietta Wolter
- Institute of Neuropathology, Heinrich Heine University, Medical Faculty, and University Hospital Düsseldorf, Düsseldorf, Germany
| | - Matthias Reusche
- Institute for Medical Informatics, Statistics and Epidemiology, University Leipzig, Leipzig, Germany
| | - Patrick Roth
- Department of Neurology, University Hospital Zurich, Frauenklinikstrasse 26, 8091, Zurich, Switzerland
- Department of Neurology, University of Zurich, Zurich, Switzerland
| | - Dietmar Krex
- Faculty of Medicine, Department of Neurosurgery, Technische Universität Dresden, University Hospital Carl Gustav Carus, Dresden, Germany
| | | | - Manfred Westphal
- Department of Neurosurgery, University of Hamburg, Hamburg, Germany
| | - Joerg C Tonn
- Department of Neurosurgery, Ludwig-Maximilians-University Munich, Munich, Germany
- German Cancer Consortium (DKTK), Partner Site Munich, Munich, Germany
| | - Luca Regli
- Department of Neurosurgery, University Hospital Zurich, Zurich, Switzerland
- Department of Neurosurgery, University of Zurich, Zurich, Switzerland
| | - Claude-Alain Maurage
- Department of Pathology, Centre Biologie Pathologie, Lille University Hospital, Hopital Nord, Lille, France
| | - Andreas von Deimling
- Department of Neuropathology, University Hospital Heidelberg, Heidelberg, Germany
- Clinical Cooperation Unit Neuropathology, German Cancer Center (DKFZ), and German Cancer Consortium (DKTK), Partner Site Heidelberg, Heidelberg, Germany
| | - Torsten Pietsch
- Department of Neuropathology, University of Bonn Medical Center, DGNN Brain Tumor Reference Center, Bonn, Germany
| | - Emilie Le Rhun
- Department of Neurology, University Hospital Zurich, Frauenklinikstrasse 26, 8091, Zurich, Switzerland
- Department of Neurology, University of Zurich, Zurich, Switzerland
- Department of Neurosurgery, University Hospital Zurich, Zurich, Switzerland
- Department of Neurosurgery, University of Zurich, Zurich, Switzerland
- Department of Neurosurgery, Lille University Hospital, Lille, France
| | - Guido Reifenberger
- Institute of Neuropathology, Heinrich Heine University, Medical Faculty, and University Hospital Düsseldorf, Düsseldorf, Germany
- German Cancer Consortium (DKTK), Partner Site Essen/Düsseldorf, Düsseldorf, Germany
| |
Collapse
|
48
|
Williamson A, Houston P, Paterson J, Chalmers AJ, McLoone P, Fullerton N, Foo SY, James A, Nowicki S. Dosimetric comparison of hippocampal-sparing technologies in patients with low-grade glioma. Neurooncol Adv 2024; 6:vdae131. [PMID: 39220244 PMCID: PMC11364934 DOI: 10.1093/noajnl/vdae131] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/04/2024] Open
Abstract
Background Radiotherapy (RT) plays an integral role in the management of low-grade gliomas (LGG). Late toxicity from RT can cause progressive neurocognitive dysfunction. Radiation-induced damage to the hippocampus (HCP) plays a considerable role in memory decline. Advancements in photon planning software have resulted in the development of multi-criteria optimization (MCO) and HyperArc technologies which may improve HCP sparing while maintaining planning target volume (PTV) target coverage. Methods Three planning methods for hippocampal sparing (HS) were compared, volumetric modulated arc therapy (VMAT) without HS (VMAT_noHS), VMAT with HS (VMAT_HS), MCO with HS (MCO_HS), and HyperArc with HS (HyperArc_HS). Results Twenty-five patients were identified. The contralateral HCP was spared in 16 patients and bilateral HCP in 9 patients with superiorly located tumors. All 3 HS planning techniques showed significant reductions in dose to the spared HCP in contralateral cases but only VMAT_HS and MCO_HS achieved this in bilateral cases (P < .008). Only MCO_HS was superior to VMAT_HS in lowering the dose to both contralateral HCP and bilateral HCP in all measured metrics (P < .008). PTV and OAR (organ at risk) dose constraints were achieved for all plans. Conclusions This retrospective dosimetric study demonstrated the feasibility of HS for low-grade glioma. All 3 HS planning techniques achieved significant dose reductions to the spared contralateral hippocampus, but only MCO_HS and VMAT_HS achieved this in bilateral cases. MCO was superior to other planning techniques for sparing both bilateral and contralateral hippocampi.
Collapse
Affiliation(s)
- Aoife Williamson
- Department of Clinical Oncology, Beatson West of Scotland Cancer Centre, Glasgow, UK
| | - Peter Houston
- Department of Clinical Oncology, Beatson West of Scotland Cancer Centre, Glasgow, UK
| | - Jennifer Paterson
- Department of Clinical Oncology, Beatson West of Scotland Cancer Centre, Glasgow, UK
| | | | - Philip McLoone
- School of Health & Wellbeing, University of Glasgow, Glasgow, UK
| | - Natasha Fullerton
- Department of Neuroradiology, Institute of Neurosciences, QEUH, Glasgow, UK
| | - Sin Yee Foo
- Department of Neuroradiology, Institute of Neurosciences, QEUH, Glasgow, UK
| | - Allan James
- Department of Clinical Oncology, Beatson West of Scotland Cancer Centre, Glasgow, UK
| | - Stefan Nowicki
- Department of Clinical Oncology, Beatson West of Scotland Cancer Centre, Glasgow, UK
| |
Collapse
|
49
|
Darlix A, Monnier M, Castan F, Coutant L, Fabbro M, Denis-Chammas È, Carrière M, Menjot-de-Champfleur N, Rigau V, Duffau H, Guerdoux E. Longitudinal assessment of quality of life, neurocognition, and psychopathology in patients with low-grade glioma on first-line temozolomide: A feasibility study. Neurooncol Adv 2024; 6:vdae084. [PMID: 38946878 PMCID: PMC11212068 DOI: 10.1093/noajnl/vdae084] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/02/2024] Open
Abstract
Background The treatment timing and choice after neurosurgical resection in patients with newly diagnosed diffuse low-grade glioma (DLGG) remain controversial. Indeed, the effect of such treatments must be balanced with the possible side effects. This study evaluated the feasibility of longitudinal exhaustive quality of life (QoL) and neuropsychological assessments in patients with DLGG receiving first-line temozolomide. Methods QoL, neurocognition, and psychological disorders were assessed prospectively until disease progression, using testing, clinician-reported, and self-reported questionnaires. The primary endpoint was the participation and adherence to this complete assessment at Baseline (before temozolomide initiation), months 6 and 12 of treatment, and month 6 post-treatment. The QoL and neuropsychological changes over time also were described. Results Twenty-six of the twenty-nine eligible patients were enrolled (participation rate: 89.7%, 95% CI: 72.6-97.8). The adherence rate was 95.7% (95% CI: 78.1-99.9; n = 23 because 3 patients progressed in the first 12 months of treatment). Up to month 6 post-treatment, QoL and fatigue remained stable (EORTC QLQC30 and BN20, MFI-20); some specific symptoms were transitory. Both subjective (FACT-Cog) and objective (Z-scores of neurocognitive tests) neurocognitive outcomes remained stable or tended to improve. The percentage of patients with severe depression (BDI-II), anxiety (STAI-Y), or anger (STAXI-II) was stable over time. Conclusions This prospective study demonstrated the feasibility of an exhaustive and longitudinal evaluation of QoL, neurocognition, and psychological disorders, with high acceptability by patients with DLGG undergoing chemotherapy. First-line temozolomide seems to have limited short-term effects on QoL and neurocognition. These findings must be confirmed in the long term and in a larger cohort.
Collapse
Affiliation(s)
- Amélie Darlix
- Institut de Génomique Fonctionnelle, University of Montpellier, CNRS, INSERM, Montpellier, France
- Department of Medical Oncology, Montpellier Cancer Institute (ICM), University of Montpellier, Montpellier, France
| | - Maëva Monnier
- Biometrics Unit, Montpellier Cancer Institute (ICM), University of Montpellier, Montpellier, France
| | - Florence Castan
- Biometrics Unit, Montpellier Cancer Institute (ICM), University of Montpellier, Montpellier, France
| | - Louise Coutant
- Department of Supportive Care, Psycho-Oncology Unit, Montpellier Cancer Institute (ICM), University of Montpellier, Montpellier, France
| | - Michel Fabbro
- Department of Medical Oncology, Montpellier Cancer Institute (ICM), University of Montpellier, Montpellier, France
| | - Ève Denis-Chammas
- Department of Neuroradiology, University of Montpellier, CHU Montpellier, Montpellier, France
| | - Mathilde Carrière
- Department of Neuroradiology, University of Montpellier, CHU Montpellier, Montpellier, France
| | | | - Valérie Rigau
- Institut de Génomique Fonctionnelle, University of Montpellier, CNRS, INSERM, Montpellier, France
- Department of Pathology, University of Montpellier, CHU Montpellier, Montpellier, France
| | - Hugues Duffau
- Institut de Génomique Fonctionnelle, University of Montpellier, CNRS, INSERM, Montpellier, France
- Department of Neurosurgery, University of Montpellier, CHU Montpellier, Montpellier, France
| | - Estelle Guerdoux
- Department of Supportive Care, Psycho-Oncology Unit, Montpellier Cancer Institute (ICM), University of Montpellier, Montpellier, France
- Desbrest Institute of Epidemiology and Public Health (IDESP), INSERM, Montpellier Cancer Institute (ICM), University of Montpellier, France
| |
Collapse
|
50
|
Wu C, Li Y, Luo Y, Dai Y, Qin J, Liu N, Xu R, Li X, Zhang P. Analysis of glutathione Stransferase mu class 5 gene methylation as a prognostic indicator in low-grade gliomas. Technol Health Care 2024; 32:3925-3942. [PMID: 39031395 PMCID: PMC11612950 DOI: 10.3233/thc-231316] [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: 09/16/2023] [Accepted: 05/22/2024] [Indexed: 07/22/2024]
Abstract
BACKGROUND Low-grade gliomas (LGG) are a variety of brain tumors that show different clinical outcomes. The methylation of the GSTM5 gene has been noted in the development of LGG, however, its prognostic importance remains uncertain. OBJECTIVE The objective of this study was to examine the correlation between GSTM5 DNA methylation and clinical outcomes in individuals diagnosed with LGG. METHODS Analysis of GSTM5 methylation levels in LGG samples was conducted using data from The Cancer Genome Atlas (TCGA) and Gene Expression Omnibus (GEO) datasets. The overall survival based on GSTM5 methylation status was evaluated using Kaplan-Meier curves. The DNA methylation heatmap for particular CpG sites in the GSTM5 gene was visualized using the "pheatmap" R package. RESULTS The study analyzed that LGG tumors had higher levels of GSTM5 methylation than normal tissues. There was an inverse relationship discovered between GSTM5 expression and methylation. LGG patients with hypermethylation of GSTM5 promoter experienced a positive outcome. Age, grade, and GSTM5 methylation were determined as independent prognostic factors in LGG through both univariate and multivariate Cox regression analyses. CONCLUSION Methylation of GSTM5 DNA, specifically at certain CpG sites, is linked to a positive outlook in patients with LGG. Utilizing the "pheatmap" R package to visualize GSTM5 methylation patterns offers important information for identifying prognostic markers and therapeutic targets in low-grade gliomas.
Collapse
Affiliation(s)
- Cuiying Wu
- Department of Neurosurgery, The Seventh Medical Centre of PLA General Hospital, Beijing, China
- Department of Neurobiology, Beijing Institute of Basic Medical Sciences, Beijing, China
| | - Yunjun Li
- Department of Neurosurgery, The Seventh Medical Centre of PLA General Hospital, Beijing, China
- Department of Neurosurgery, Senior Department of Neurosurgery, The First Medical Centre of PLA General Hospital, Beijing, China
| | - Yongchun Luo
- Department of Neurosurgery, Senior Department of Neurosurgery, The First Medical Centre of PLA General Hospital, Beijing, China
| | - Yiwu Dai
- Department of Neurosurgery, Senior Department of Neurosurgery, The First Medical Centre of PLA General Hospital, Beijing, China
| | - Jiazhen Qin
- Department of Neurosurgery, Senior Department of Neurosurgery, The First Medical Centre of PLA General Hospital, Beijing, China
| | - Ning Liu
- Department of Neurosurgery, The Seventh Medical Centre of PLA General Hospital, Beijing, China
| | - Ruxiang Xu
- Department of Neurosurgery, Sichuan Academy of Medical Science and Sichuan Provincial People’s Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Xuezhen Li
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Peng Zhang
- Department of Neurosurgery, The Seventh Medical Centre of PLA General Hospital, Beijing, China
- Department of Neurosurgery, Senior Department of Neurosurgery, The First Medical Centre of PLA General Hospital, Beijing, China
| |
Collapse
|