101
|
Quantitative Analysis of the MGMT Methylation Status of Glioblastomas in Light of the 2021 WHO Classification. Cancers (Basel) 2022; 14:cancers14133149. [PMID: 35804921 PMCID: PMC9264886 DOI: 10.3390/cancers14133149] [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: 05/20/2022] [Revised: 06/22/2022] [Accepted: 06/24/2022] [Indexed: 01/27/2023] Open
Abstract
Background: Glioblastomas with methylation of the promoter region of the O(6)-methylguanine-DNA methyltransferase (MGMT) gene exhibit increased sensitivity to alkylating chemotherapy. Quantitative assessment of the MGMT promoter methylation status might provide additional prognostic information. The aim of our study was to determine a quantitative methylation threshold for better survival among patients with glioblastomas. Methods: We included consecutive patients ≥18 years treated at our department between 11/2010 and 08/2018 for a glioblastoma, IDH wildtype, undergoing quantitative MGMT promoter methylation analysis. The primary endpoint was overall survival. Results: A total of 321 patients were included. Median overall survival was 12.6 months. Kaplan−Meier and adjusted Cox regression analysis showed better survival for the groups with 16−30%, 31−60%, and 61−100% methylation. In contrast, survival in the group with 1−15% methylation was similar to those with unmethylated promoter regions. A secondary analysis confirmed this threshold. Conclusions: Better survival is observed in patients with glioblastomas with ≥16% methylation of the MGMT promoter region than with <16% methylation. Survival with tumors with 1−15% methylation is similar to with unmethylated tumors. Above 16% methylation, we found no additional benefit with increasing methylation.
Collapse
|
102
|
Chromatin structure predicts survival in glioma patients. Sci Rep 2022; 12:8221. [PMID: 35581287 PMCID: PMC9114333 DOI: 10.1038/s41598-022-11019-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2022] [Accepted: 03/15/2022] [Indexed: 11/08/2022] Open
Abstract
The pathological changes in epigenetics and gene regulation that accompany the progression of low-grade to high-grade gliomas are under-studied. The authors use a large set of paired atac-seq and RNA-seq data from surgically resected glioma specimens to infer gene regulatory relationships in glioma. Thirty-eight glioma patient samples underwent atac-seq sequencing and 16 samples underwent additional RNA-seq analysis. Using an atac-seq/RNA-seq correlation matrix, atac-seq peaks were paired with genes based on high correlation values (|r2| > 0.6). Samples clustered by IDH1 status but not by grade. Surprisingly there was a trend for IDH1 mutant samples to have more peaks. The majority of peaks are positively correlated with survival and positively correlated with gene expression. Constructing a model of the top six atac-seq peaks created a highly accurate survival prediction model (r2 = 0.68). Four of these peaks were still significant after controlling for age, grade, pathology, IDH1 status and gender. Grade II, III, and IV (primary) samples have similar transcription factors and gene modules. However, grade IV (recurrent) samples have strikingly few peaks. Patient-derived glioma cultures showed decreased peak counts following radiation indicating that this may be radiation-induced. This study supports the notion that IDH1 mutant and IDH1 wildtype gliomas have different epigenetic landscapes and that accessible chromatin sites mapped by atac-seq peaks tend to be positively correlated with expression. The data in this study leads to a new model of treatment response wherein glioma cells respond to radiation therapy by closing open regions of DNA.
Collapse
|
103
|
Radiation-induced contrast enhancement following proton radiotherapy for low-grade glioma depends on tumor characteristics and is rarer in children than adults. Radiother Oncol 2022; 172:54-64. [PMID: 35568281 DOI: 10.1016/j.radonc.2022.05.005] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2022] [Revised: 04/21/2022] [Accepted: 05/05/2022] [Indexed: 12/24/2022]
Abstract
BACKGROUND AND PURPOSE Proton beam radiotherapy (PRT) is used in the treatment of low-grade glioma (LGG) to mitigate long-term sequelae. Following PRT, increased rates of radiation-induced contrast enhancements (RICE) are suspected but poorly understood. MATERIALS AND METHODS We analyzed consecutive 227 patients (42 children and 185 adults) treated with PRT (54Gy RBE) for LGG from 2010 to 2020 and followed with serial clinical exams and magnetic resonance imaging for in median 5.6 years. RESULTS Tumors were graded WHO 1 in a minority (n = 22, 12%) of adults, but a majority of children (n = 29, 69%). In contrast, tumors were graded WHO 2 in the majority (n = 160, 87%) of adults and a minority of children (n = 10, 24%). Five-year overall survival following PRT was 81% in adults and 91% in children. The risk of RICE was 5-fold more frequent in adults (25%) versus children (5%) (p = 0.0043). In children and adults, RICE were symptomatic in 50% and 55% (n=1 and 26) of cases with CTCAE grade 0 in 47% (n=23), grade 1 in 25% (n=12), 0% grade 2 (n=0) and 29% grade 3 (n=14), respectively. In adults, RICE risk was associated to WHO grading (8% in WHO grade 1 vs. 24% in WHO grade 2, p = 0.026), independent of age (p=0.44) and irradiation dose (p=0.005), but not independent of IDH mutational status. CONCLUSIONS These data demonstrate effectiveness of PRT for LGG in both children and adults. The RICE risk is lower in children which are a main target group for PRT and differs with WHO grading.
Collapse
|
104
|
Nardone V, Desideri I, D’Ambrosio L, Morelli I, Visani L, Di Giorgio E, Guida C, Clemente A, Belfiore MP, Cioce F, Spadafora M, Vinciguerra C, Mansi L, Reginelli A, Cappabianca S. Nuclear medicine and radiotherapy in the clinical management of glioblastoma patients. Clin Transl Imaging 2022. [DOI: 10.1007/s40336-022-00495-8] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Abstract
Introduction
The aim of the narrative review was to analyse the applications of nuclear medicine (NM) techniques such as PET/CT with different tracers in combination with radiotherapy for the clinical management of glioblastoma patients.
Materials and methods
Key references were derived from a PubMed query. Hand searching and clinicaltrials.gov were also used.
Results
This paper contains a narrative report and a critical discussion of NM approaches in combination with radiotherapy in glioma patients.
Conclusions
NM can provide the Radiation Oncologist several aids that can be useful in the clinical management of glioblastoma patients. At the same, these results need to be validated in prospective and multicenter trials.
Collapse
|
105
|
Dono A, Alfaro-Munoz K, Yan Y, Lopez-Garcia CA, Soomro Z, Williford G, Takayasu T, Robell L, Majd NK, de Groot J, Esquenazi Y, Kamiya-Matsuoka C, Ballester LY. Molecular, Histological, and Clinical Characteristics of Oligodendrogliomas: A Multi-Institutional Retrospective Study. Neurosurgery 2022; 90:515-522. [PMID: 35179134 PMCID: PMC9514747 DOI: 10.1227/neu.0000000000001875] [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: 03/18/2021] [Accepted: 11/01/2021] [Indexed: 11/19/2022] Open
Abstract
BACKGROUND Reports suggest that phosphatidylinositol 3-kinase pathway alterations confer increased risk of progression and poor prognosis in oligodendroglioma, IDH-mutant, and 1p/19q-codeleted molecular oligodendrogliomas (mODG). However, factors that affect prognosis in mODG have not been thoroughly studied. In addition, the benefits of adjuvant radiation and temozolomide (TMZ) in mODGs remain to be determined. OBJECTIVE To evaluate the role of PIK3CA mutations in mODGs. METHODS One hundred seven mODGs (2008-2019) diagnosed at 2 institutions were included. A retrospective review of clinical characteristics, molecular alterations, treatments, and outcomes was performed. RESULTS The median age was 37 years, and 61 patients (57%) were male. There were 64 (60%) World Health Organization (WHO) grade 2 and 43 (40%) WHO grade 3 tumors. Eighty-two patients (77%) were stratified as high risk (age 40 years or older and/or subtotal resection per Radiation Treatment Oncology Group-9802). Gross-total resection was achieved in 47 patients (45%). Treatment strategies included observation (n = 15), TMZ (n = 11), radiation (n = 13), radiation/TMZ (n = 62), and others (n = 6). Our results show a benefit of TMZ vs observation in progression-free survival (PFS). No difference in PFS or overall survival (OS) was observed between radiation and radiation/TMZ. PIK3CA mutations were detected in 15 (14%) mODG, and shorter OS was observed in PIK3CA-mutant compared with PIK3CA wild-type mODGs (10.7 years vs 15.1 years, P = .009). WHO grade 3 tumors showed a shorter PFS, but no significant difference in OS was observed between WHO grades. CONCLUSION Our findings suggest that mODGs harboring PIK3CA mutations have worse OS. Except for an advantage in PFS with TMZ treatment, adjuvant TMZ, radiation, or a combination of the two showed no significant improvement in OS.
Collapse
Affiliation(s)
- Antonio Dono
- Vivian L. Smith Department of Neurosurgery, School of Biomedical Informatics, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, Texas, USA
- Department of Pathology and Laboratory Medicine, School of Biomedical Informatics, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, Texas, USA
| | | | - Yuanqing Yan
- Vivian L. Smith Department of Neurosurgery, School of Biomedical Informatics, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, Texas, USA
| | - Carlos A. Lopez-Garcia
- Department of Pathology and Laboratory Medicine, School of Biomedical Informatics, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, Texas, USA
| | - Zaid Soomro
- Department of Neuro-Oncology, MD Anderson Cancer Center, Houston, Texas, USA
| | - Garret Williford
- Department of Neuro-Oncology, MD Anderson Cancer Center, Houston, Texas, USA
| | - Takeshi Takayasu
- Department of Pathology and Laboratory Medicine, School of Biomedical Informatics, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, Texas, USA
| | - Lindsay Robell
- Department of Neuro-Oncology, MD Anderson Cancer Center, Houston, Texas, USA
| | - Nazanin K. Majd
- Department of Neuro-Oncology, MD Anderson Cancer Center, Houston, Texas, USA
| | - John de Groot
- Department of Neuro-Oncology, MD Anderson Cancer Center, Houston, Texas, USA
| | - Yoshua Esquenazi
- Vivian L. Smith Department of Neurosurgery, School of Biomedical Informatics, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, Texas, USA
- Center of Precision Health, School of Biomedical Informatics, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, Texas, USA
- Memorial Hermann Hospital, Houston, Texas, USA
| | | | - Leomar Y. Ballester
- Vivian L. Smith Department of Neurosurgery, School of Biomedical Informatics, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, Texas, USA
- Department of Pathology and Laboratory Medicine, School of Biomedical Informatics, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, Texas, USA
- Memorial Hermann Hospital, Houston, Texas, USA
| |
Collapse
|
106
|
Wu J, Shen S, Liu T, Ren X, Zhu C, Liang Q, Cui X, Chen L, Cheng P, Cheng W, Wu A. Chemerin enhances mesenchymal features of glioblastoma by establishing autocrine and paracrine networks in a CMKLR1-dependent manner. Oncogene 2022; 41:3024-3036. [PMID: 35459783 PMCID: PMC9122825 DOI: 10.1038/s41388-022-02295-w] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2021] [Revised: 03/14/2022] [Accepted: 03/23/2022] [Indexed: 11/18/2022]
Abstract
Glioblastoma multiforme (GBM) with mesenchymal features exhibits enhanced chemotherapeutic resistance and results in reduced overall survival. Recent studies have suggested that there is a positive correlation between the GBM mesenchymal status and immune cell infiltration. However, the mechanisms by which GBM acquires its mesenchymal features in a tumor immune microenvironment-dependent manner remains unknown. Here, we uncovered a chemerin-mediated autocrine and paracrine network by which the mesenchymal phenotype of GBM cells is strengthened. We identified chemerin as a prognostic secretory protein mediating the mesenchymal phenotype-promoting network between tumor-associated macrophages (TAMs) and tumor cells in GBM. Mechanistically, chemerin promoted the mesenchymal features of GBM by suppressing the ubiquitin-proteasomal degradation of CMKLR1, a chemerin receptor predominantly expressed on TAMs and partially expressed on GBM cells, thereby enhancing NF-κB pathway activation. Moreover, chemerin was found to be involved in the recruitment of TAMs in the GBM tumor microenvironment. We revealed that chemerin also enhances the mesenchymal phenotype-promoting ability of TAMs and promotes their M2 polarization via a CMKLR1/NF-κB axis, which further exacerbates the mesenchymal features of GBM. Blocking the chemerin/CMKLR1 axis with 2-(α-naphthoyl) ethyltrimethylammonium iodide disrupted the mesenchymal network and suppressed tumor growth in GBM. These results suggest the therapeutic potential of targeting the chemerin/CMKLR1 axis to block the mesenchymal network in GBM.
Collapse
Affiliation(s)
- Jianqi Wu
- Department of Neurosurgery, The First Hospital of China Medical University, Shenyang, China
| | - Shuai Shen
- Department of Neurosurgery, The First Hospital of China Medical University, Shenyang, China
| | - Tianqi Liu
- Department of Neurosurgery, The First Hospital of China Medical University, Shenyang, China
| | - Xiufang Ren
- Departement of Pathology, Shengjing Hospital of China Medical University, Shenyang, China
| | - Chen Zhu
- Department of Neurosurgery, The First Hospital of China Medical University, Shenyang, China
| | - Qingyu Liang
- Department of Neurosurgery, The First Hospital of China Medical University, Shenyang, China
| | - Xiao Cui
- Department of Neurosurgery, The First Hospital of China Medical University, Shenyang, China
| | - Ling Chen
- Department of Neurosurgery, Chinese People's Liberation Army of China (PLA) General Hospital, Medical School of Chinese PLA, Institute of Neurosurgery of Chinese PLA, Beijing, China
| | - Peng Cheng
- Department of Neurosurgery, The First Hospital of China Medical University, Shenyang, China
| | - Wen Cheng
- Department of Neurosurgery, The First Hospital of China Medical University, Shenyang, China.
| | - Anhua Wu
- Department of Neurosurgery, The First Hospital of China Medical University, Shenyang, China.
| |
Collapse
|
107
|
Zhao M, Li X, Chen Y, Wang S. MD2 Is a Potential Biomarker Associated with Immune Cell Infiltration in Gliomas. Front Oncol 2022; 12:854598. [PMID: 35372062 PMCID: PMC8968038 DOI: 10.3389/fonc.2022.854598,] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2022] [Accepted: 02/23/2022] [Indexed: 09/03/2024] Open
Abstract
BACKGROUND Glioma is the most common primary malignant tumor in the central nervous system. Myeloid differentiation protein 2 (MD2) acts as a coreceptor of toll-like receptor 4 (TLR4) to mediate innate immune response. However, the actual roles of MD2 in the regulation of progression and immune cell infiltration in gliomas remain largely unclear. This study aims to explore whether MD2 could be an independent prognostic factor through the mediation of immune cell infiltration in gliomas. METHODS The mRNA expression and DNA methylation differential analyses of MD2 were performed using CGGA, TCGA and Rembrandt databases and survival analyses were performed using Kaplan-Meier plotter. Univariate and multivariate Cox regression was applied to analyze the prognostic value of MD2 and nomograms were constructed to evaluate the clinical value of MD2. Then, Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) were utilized to analyze MD2-related signal pathways. Furthermore, correlations between MD2 and immune cell infiltration were calculated by TIMER and CIBERSOPT. The correlation between MD2 expression and the infiltrations of macrophages and neutrophils was experimentally verified by the knockdown of MD2 expression using small interfering RNA (siRNA) in glioma cells. RESULTS We found that MD2 was overexpressed and associated with a poor prognosis in gliomas. Meanwhile, higher expression of MD2 could be a result of lower DNA methylation of MD2 gene in gliomas. In addition, univariate and multivariate Cox regression analysis indicated that MD2 could be an independent prognostic factor for gliomas. Further functional enrichment analysis revealed that the functions of MD2 were closely related to immune responses. Moreover, the expression level of MD2 was strongly correlated with the infiltration and polarization of pro-tumor phenotype of tumor-associated macrophages and tumor-associated neutrophils in gliomas. CONCLUSIONS These findings have provided strong evidence that MD2 could be served as a valuable immune-related biomarker to diagnose and predict the progression of gliomas.
Collapse
Affiliation(s)
| | | | - Yijun Chen
- State Key Laboratory of Natural Medicines and Laboratory of Chemical Biology, School of Life Science and Technology, China Pharmaceutical University, Nanjing, China
| | - Shuzhen Wang
- State Key Laboratory of Natural Medicines and Laboratory of Chemical Biology, School of Life Science and Technology, China Pharmaceutical University, Nanjing, China
| |
Collapse
|
108
|
Zhao M, Li X, Chen Y, Wang S. MD2 Is a Potential Biomarker Associated with Immune Cell Infiltration in Gliomas. Front Oncol 2022; 12:854598. [PMID: 35372062 PMCID: PMC8968038 DOI: 10.3389/fonc.2022.854598] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2022] [Accepted: 02/23/2022] [Indexed: 11/30/2022] Open
Abstract
BACKGROUND Glioma is the most common primary malignant tumor in the central nervous system. Myeloid differentiation protein 2 (MD2) acts as a coreceptor of toll-like receptor 4 (TLR4) to mediate innate immune response. However, the actual roles of MD2 in the regulation of progression and immune cell infiltration in gliomas remain largely unclear. This study aims to explore whether MD2 could be an independent prognostic factor through the mediation of immune cell infiltration in gliomas. METHODS The mRNA expression and DNA methylation differential analyses of MD2 were performed using CGGA, TCGA and Rembrandt databases and survival analyses were performed using Kaplan-Meier plotter. Univariate and multivariate Cox regression was applied to analyze the prognostic value of MD2 and nomograms were constructed to evaluate the clinical value of MD2. Then, Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) were utilized to analyze MD2-related signal pathways. Furthermore, correlations between MD2 and immune cell infiltration were calculated by TIMER and CIBERSOPT. The correlation between MD2 expression and the infiltrations of macrophages and neutrophils was experimentally verified by the knockdown of MD2 expression using small interfering RNA (siRNA) in glioma cells. RESULTS We found that MD2 was overexpressed and associated with a poor prognosis in gliomas. Meanwhile, higher expression of MD2 could be a result of lower DNA methylation of MD2 gene in gliomas. In addition, univariate and multivariate Cox regression analysis indicated that MD2 could be an independent prognostic factor for gliomas. Further functional enrichment analysis revealed that the functions of MD2 were closely related to immune responses. Moreover, the expression level of MD2 was strongly correlated with the infiltration and polarization of pro-tumor phenotype of tumor-associated macrophages and tumor-associated neutrophils in gliomas. CONCLUSIONS These findings have provided strong evidence that MD2 could be served as a valuable immune-related biomarker to diagnose and predict the progression of gliomas.
Collapse
Affiliation(s)
| | | | - Yijun Chen
- State Key Laboratory of Natural Medicines and Laboratory of Chemical Biology, School of Life Science and Technology, China Pharmaceutical University, Nanjing, China
| | - Shuzhen Wang
- State Key Laboratory of Natural Medicines and Laboratory of Chemical Biology, School of Life Science and Technology, China Pharmaceutical University, Nanjing, China
| |
Collapse
|
109
|
Xia H, Liu B, Shen N, Xue J, Chen S, Guo H, Zhou X. circRNA-0002109 promotes glioma malignant progression via modulating the miR-129-5P/EMP2 axis. MOLECULAR THERAPY. NUCLEIC ACIDS 2022; 27:1-15. [PMID: 34938603 PMCID: PMC8646083 DOI: 10.1016/j.omtn.2021.11.011] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/24/2021] [Revised: 09/05/2021] [Accepted: 11/16/2021] [Indexed: 02/08/2023]
Abstract
Glioma is a common intracranial malignant tumor with high mortality and high recurrence rate. In recent years, increasing evidence has demonstrated that circular RNAs (circRNAs) are potential biomarkers and therapeutic targets for many tumors. However, the role of circRNAs in glioma remains unclear. In this study, we found that circRNA-0002109 was highly expressed in glioma tissues and cell lines. Downregulation of circRNA-0002109 expression inhibited the proliferation, migration, and invasion of glioma cells and inhibited the malignant progression of tumors in vivo. Investigations into the relevant mechanisms showed that circRNA-0002109 upregulated the expression of EMP2 through endogenous competitive binding of microRNA-129-5P (miR-129-5P), which partially alleviated the inhibitory effect of miR-129-5P on epithelial membrane protein-2 (EMP2) and ultimately promoted the malignant development of glioma. Our results indicate that circRNA-0002109 plays an important role in the proliferation, invasion, and migration of glioma cells by regulating the miR-129-5P/EMP2 axis, which provides a new potential therapeutic target for glioma.
Collapse
Affiliation(s)
- Haibin Xia
- Institute of Neuroscience and the Second Affiliated Hospital of Guangzhou Medical University, 250 Chang-gang East Road, Guangzhou 510260, China.,Trauma Center, First Affiliated Hospital of Gannan Medical University, Ganzhou 341000, China
| | - Boyang Liu
- Neurosurgery Center, The National Key Clinical Specialty, The Engineering Technology Research Center of Education Ministry of China on Diagnosis and Treatment of Cerebrovascular Disease, Guangdong Provincial Key Laboratory on Brain Function Repair and Regeneration, The Neurosurgery Institute of Guangdong Province, Zhujiang Hospital, Southern Medical University, Guangzhou 510282, China
| | - Nanxiang Shen
- Institute of Neuroscience and the Second Affiliated Hospital of Guangzhou Medical University, 250 Chang-gang East Road, Guangzhou 510260, China
| | - Jinhua Xue
- Department of Physiology, School of Basic Medical Sciences, Gannan Medical University, Ganzhou 341000, China
| | - Siyu Chen
- Institute of Neuroscience and the Second Affiliated Hospital of Guangzhou Medical University, 250 Chang-gang East Road, Guangzhou 510260, China
| | - Hongbo Guo
- Neurosurgery Center, The National Key Clinical Specialty, The Engineering Technology Research Center of Education Ministry of China on Diagnosis and Treatment of Cerebrovascular Disease, Guangdong Provincial Key Laboratory on Brain Function Repair and Regeneration, The Neurosurgery Institute of Guangdong Province, Zhujiang Hospital, Southern Medical University, Guangzhou 510282, China
| | - Xiaozhong Zhou
- Trauma Center, First Affiliated Hospital of Gannan Medical University, Ganzhou 341000, China
| |
Collapse
|
110
|
Blonski M, Obara T, Brzenczek C, Pouget C, Dillier C, Meyer M, Lavigne L, Forthoffer N, Broussois A, Gauchotte G, Baron MH, Rech F, Mézières S, Gaudeau Y, Verger A, Vogin G, Anxionnat R, Moureaux JM, Taillandier L. Initial PCV Chemotherapy Followed by Radiotherapy Is Associated With a Prolonged Response But Late Neurotoxicity in 20 Diffuse Low-Grade Glioma Patients. Front Oncol 2022; 12:827897. [PMID: 35311144 PMCID: PMC8931287 DOI: 10.3389/fonc.2022.827897] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2021] [Accepted: 02/03/2022] [Indexed: 11/13/2022] Open
Abstract
BackgroundStudy RTOG 9802 in high-risk diffuse low-grade gliomas (DLGGs) showed the potential synergistic effect on survival of the procarbazine, CCNU, and vincristine (PCV) radiotherapy (RT) combination. Limited data on long-term neurocognitive impact and quality of life (QoL) have yet been reported.Patients and MethodsWe described a monocentric series of patients treated at first line by the combination of PCV immediately followed by RT between January 01, 1982 and January 01, 2017. Radiological data were collected and included volume, velocity of diametric expansion (VDE), and MRI aspects. Long-term neurocognitive and QoL were analyzed.ResultsTwenty patients fulfilled the eligibility criteria. The median response rate was 65.1% (range, 9.6%–99%) at the time of maximal VDE decrease corresponding to a median volume reduction of 79.7 cm3 (range, 3.1 to 174.2 cm3), which occurred after a median period of 7.2 years (range, 0.3–21.9) after the end of RT. An ongoing negative VDE was measured in 13/16 patients after the end of RT, with a median duration of 6.7 years (range, 9 months–21.9 years). The median follow-up since radiological diagnosis was 17.5 years (range, 4.8 to 29.5). Estimated median survival was 17.4 years (95% CI: 12; NR). After a long-term follow-up, substantial neurotoxicity was noticed with dementia in six progression-free patients (30%), leading to ventriculo-peritoneal shunt procedures in three, and premature death in five. Thirteen patients (65%) were unable to work with disability status. Successive longitudinal neurocognitive assessments for living patients showed verbal episodic memory deterioration.ConclusionsPCV-RT combination seems to have not only an oncological synergy but also a long-term neurotoxic synergy to consider before initial therapeutic decision.
Collapse
Affiliation(s)
- Marie Blonski
- Department of Neurology, Neurooncology Unit, CHRU, Nancy, France
- Centre de Recherche en Automatique Nancy France - UMR 7039 - BioSiS Department, Faculty of Medicine, Université de Lorraine, Vandoeuvre-lès Nancy, France
- *Correspondence: Marie Blonski,
| | - Tiphaine Obara
- Department of Neurology, Neurooncology Unit, CHRU, Nancy, France
- Centre de Recherche en Automatique Nancy France - UMR 7039 - BioSiS Department, Faculty of Medicine, Université de Lorraine, Vandoeuvre-lès Nancy, France
| | - Cyril Brzenczek
- Centre de Recherche en Automatique Nancy France - UMR 7039 - BioSiS Department, Faculty of Medicine, Université de Lorraine, Vandoeuvre-lès Nancy, France
| | - Celso Pouget
- Department of Pathology, CHRU, Nancy, France
- Centre de ressources Biologiques, BB-0033-00035, CHRU, Nancy, France
| | - Céline Dillier
- Department of Neurology, Neurooncology Unit, CHRU, Nancy, France
| | - Mylène Meyer
- Department of Neurology, Neurooncology Unit, CHRU, Nancy, France
| | - Laura Lavigne
- Department of Neurology, Neurooncology Unit, CHRU, Nancy, France
| | | | | | - Guillaume Gauchotte
- Department of Pathology, CHRU, Nancy, France
- Centre de ressources Biologiques, BB-0033-00035, CHRU, Nancy, France
| | | | - Fabien Rech
- Centre de Recherche en Automatique Nancy France - UMR 7039 - BioSiS Department, Faculty of Medicine, Université de Lorraine, Vandoeuvre-lès Nancy, France
- Department of Neurosurgery, CHRU, Nancy, France
| | - Sophie Mézières
- Department of Mathematics, Elie Cartan Institute, Nancy, France
- INRIA Biology, Genetics and Statistics, Nancy, France
| | - Yann Gaudeau
- Centre de Recherche en Automatique Nancy France - UMR 7039 - BioSiS Department, Faculty of Medicine, Université de Lorraine, Vandoeuvre-lès Nancy, France
| | - Antoine Verger
- Department of Nuclear Medicine and Nancyclotep Imaging Platform, CHRU, Nancy, France
- IADI, INSERM U1254, Université de Lorraine, Vandoeuvre-lès Nancy, France
| | - Guillaume Vogin
- Department of Radiation Therapy, Baclesse Radiation Therapy Center, Esch/Alzette, Luxembourg
- UMR 7635 CNRS, IMoPA Biopole Lorraine University Faculty of Medicine, Université de Lorraine, Vandoeuvre-lès-Nancy, France
| | | | - Jean-Marie Moureaux
- Centre de Recherche en Automatique Nancy France - UMR 7039 - BioSiS Department, Faculty of Medicine, Université de Lorraine, Vandoeuvre-lès Nancy, France
| | - Luc Taillandier
- Department of Neurology, Neurooncology Unit, CHRU, Nancy, France
- Centre de Recherche en Automatique Nancy France - UMR 7039 - BioSiS Department, Faculty of Medicine, Université de Lorraine, Vandoeuvre-lès Nancy, France
| |
Collapse
|
111
|
Tom MC, Milano MT, Chao ST, Soltys SG, Knisely JP, Sahgal A, Nagpal S, Lo SS, Jabbari S, Wang TJ, Ahluwalia MS, Simonson M, Palmer JD, Gephart MH, Halasz LM, Garg AK, Chiang VL, Chang EL. Executive summary of american radium society’s appropriate use criteria for the postoperative management of lower grade gliomas. Radiother Oncol 2022; 170:79-88. [DOI: 10.1016/j.radonc.2022.03.018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2022] [Revised: 03/22/2022] [Accepted: 03/28/2022] [Indexed: 10/18/2022]
|
112
|
Byrne E, Abel S, Yu A, Shepard M, Karlovits SM, Wegner RE. Trends in radiation dose for low grade gliomas across the United States. J Neurooncol 2022; 157:197-205. [PMID: 35199246 DOI: 10.1007/s11060-022-03962-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2021] [Accepted: 02/03/2022] [Indexed: 11/30/2022]
Abstract
PURPOSE Adjuvant radiation is often used in patients with low grade gliomas with high-risk characteristics with a recommended dose of 45-54 Gy. We used the National Cancer Database (NCDB) to see which doses were being used, and if any difference was seen in outcome. METHODS We queried the NCDB for patients with WHO Grade 2 primary brain tumors treated with surgery and adjuvant radiotherapy. We divided the cohort into dose groups: 45-50 Gy, 50.4-54 Gy, and > 54 Gy. Multivariable logistic regression was used to identify predictors of low and high dose radiation. Propensity matching was used to account for indication bias. RESULTS We identified 1437 patients meeting inclusion criteria. Median age was 45 years and 62% of patients were > 40 years old. Nearly half of patients (48%) had astrocytoma subtype and 70% had subtotal resection. The majority of patients (69%) were treated to doses between 50.4 and 54 Gy. Predictors of high dose radiation (> 54 Gy) were increased income, astrocytoma subtype, chemotherapy receipt, and treatment in later year (2014). The main predictors of survival were age > 40, astrocytoma subtype, and insurance type. Patients treated to a dose of > 54 Gy had a median survival of 73.5 months and was not reached in those treated to a lower dose (p = 0.0041). CONCLUSIONS This analysis shows that 50.4-54 Gy is the most widely used radiation regimen for the adjuvant treatment of low-grade gliomas. There appeared to be no benefit to higher doses, although unreported factors may impact interpretation of the results.
Collapse
Affiliation(s)
- Emma Byrne
- Drexel University College of Medicine, Philadelphia, PA, USA
| | - Stephen Abel
- Division of Radiation Oncology, Allegheny Health Network Cancer Institute, Pittsburgh, PA, USA
| | - Alexander Yu
- Department of Neurosurgery, Allegheny Health Network, Pittsburgh, PA, USA
| | - Matthew Shepard
- Department of Neurosurgery, Allegheny Health Network, Pittsburgh, PA, USA
| | - Stephen M Karlovits
- Division of Radiation Oncology, Allegheny Health Network Cancer Institute, Pittsburgh, PA, USA
| | - Rodney E Wegner
- Division of Radiation Oncology, Allegheny Health Network Cancer Institute, Pittsburgh, PA, USA.
| |
Collapse
|
113
|
Jo J, van den Bent MJ, Nabors B, Wen PY, Schiff D. Surveillance imaging frequency in adult patients with lower-grade (WHO Grade 2 and 3) gliomas. Neuro Oncol 2022; 24:1035-1047. [PMID: 35137214 PMCID: PMC9248400 DOI: 10.1093/neuonc/noac031] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
With improved outcome following aggressive treatment in patients with grade 2 and 3 IDH-mutant (IDHmt), 1p/19q codeleted oligodendroglioma and IDHmt, non-codeleted astrocytoma, prolonged surveillance is desirable for early detection of tumor growth and malignant transformation. Current National Comprehensive Cancer Network (NCCN) guidelines provide imaging follow-up recommendations based on molecular classification of lower-grade gliomas, although individualized imaging guidelines based on treatments received and after tumor recurrence are not clearly specified. Other available guidelines have yet to incorporate the molecular biomarkers that inform the WHO classification of gliomas, and in some cases do not adequately consider current knowledge on IDHmt glioma growth rate and recurrence patterns. Moreover, these guidelines also do not provide specific recommendations for concerning clinical symptoms or radiographic findings warranting imaging studies out of prespecified intervals. Focusing on molecularly defined grade 2 and 3 IDHmt astrocytomas and oligodendrogliomas, we review current knowledge of tumor growth rates and time to tumor progression for each tumor type and propose a range of recommended MRI surveillance intervals for both the newly diagnosed and recurrent tumor setting. Additionally, we summarize situations in which imaging is advisable outside of these intervals.
Collapse
Affiliation(s)
- Jasmin Jo
- Department of Internal Medicine, Division of Hematology and Oncology, East Carolina University, Greenville, North Carolina, USA
| | - Martin J van den Bent
- Department of Neuro-Oncology/Neurology, Erasmus MC Cancer Institute, Erasmus MC University Medical Center, Rotterdam, Netherland
| | - Burt Nabors
- Department of Neurology, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Patrick Y Wen
- Center for Neuro-Oncology, Dana-Farber/Brigham and Women’s Cancer Center; Division of Neuro-Oncology, Department of Neurology, Brigham and Women’s Hospital, and Harvard Medical School, Boston, Massachusetts, USA
| | - David Schiff
- Corresponding Author: David Schiff, MD, University of Virginia Neuro-Oncology Center, Box 800432 Charlottesville, VA 22908-0432, USA ()
| |
Collapse
|
114
|
Yang K, Wu Z, Zhang H, Zhang N, Wu W, Wang Z, Dai Z, Zhang X, Zhang L, Peng Y, Ye W, Zeng W, Liu Z, Cheng Q. Glioma targeted therapy: insight into future of molecular approaches. Mol Cancer 2022; 21:39. [PMID: 35135556 PMCID: PMC8822752 DOI: 10.1186/s12943-022-01513-z] [Citation(s) in RCA: 402] [Impact Index Per Article: 134.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2021] [Accepted: 01/12/2022] [Indexed: 12/13/2022] Open
Abstract
Gliomas are the common type of brain tumors originating from glial cells. Epidemiologically, gliomas occur among all ages, more often seen in adults, which males are more susceptible than females. According to the fifth edition of the WHO Classification of Tumors of the Central Nervous System (WHO CNS5), standard of care and prognosis of gliomas can be dramatically different. Generally, circumscribed gliomas are usually benign and recommended to early complete resection, with chemotherapy if necessary. Diffuse gliomas and other high-grade gliomas according to their molecule subtype are slightly intractable, with necessity of chemotherapy. However, for glioblastoma, feasible resection followed by radiotherapy plus temozolomide chemotherapy define the current standard of care. Here, we discuss novel feasible or potential targets for treatment of gliomas, especially IDH-wild type glioblastoma. Classic targets such as the p53 and retinoblastoma (RB) pathway and epidermal growth factor receptor (EGFR) gene alteration have met failure due to complex regulatory network. There is ever-increasing interest in immunotherapy (immune checkpoint molecule, tumor associated macrophage, dendritic cell vaccine, CAR-T), tumor microenvironment, and combination of several efficacious methods. With many targeted therapy options emerging, biomarkers guiding the prescription of a particular targeted therapy are also attractive. More pre-clinical and clinical trials are urgently needed to explore and evaluate the feasibility of targeted therapy with the corresponding biomarkers for effective personalized treatment options.
Collapse
Affiliation(s)
- Keyang Yang
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, China
- Xiangya School of Medicine, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Zhijing Wu
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, China
- Xiangya School of Medicine, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Hao Zhang
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Nan Zhang
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
- One-Third Lab, College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, China
| | - Wantao Wu
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
- Department of Oncology, Xiangya Hospital, Central South University, Changsha, China
| | - Zeyu Wang
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Ziyu Dai
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Xun Zhang
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Liyang Zhang
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Yun Peng
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
- Department of Geriatrics, Xiangya Hospital, Central South University, Changsha, China
- Teaching and Research Section of Clinical Nursing, Xiangya Hospital of Central South University, Changsha, China
| | - Weijie Ye
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha, China
| | - Wenjing Zeng
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha, China
| | - Zhixiong Liu
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, China.
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China.
| | - Quan Cheng
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, China.
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China.
| |
Collapse
|
115
|
Li C, Wan Y, Zhang Y, Fu LH, Blum NT, Cui R, Wu B, Zheng R, Lin J, Li Z, Huang P. In Situ Sprayed Starvation/Chemodynamic Therapeutic Gel for Post-Surgical Treatment of IDH1 (R132H) Glioma. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2022; 34:e2103980. [PMID: 34775641 DOI: 10.1002/adma.202103980] [Citation(s) in RCA: 59] [Impact Index Per Article: 19.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/25/2021] [Revised: 10/24/2021] [Indexed: 06/13/2023]
Abstract
Complete resection of isocitrate dehydrogenase 1 (IDH1) (R132H) glioma is unfeasible and the classic post-surgical chemo/radiotherapy suffers from high recurrence and low survival rate. IDH1 (R132H) cells are sensitive to low concentrations of glucose and high concentrations of reactive oxygen species (ROS) due to inherent metabolism reprograming. Hence, a starvation/chemodynamic therapeutic gel is developed to combat residual IDH1 (R132H) tumor cells after surgery. Briefly, glucose oxidase (GOx) is mineralized with manganese-doped calcium phosphate to form GOx@MnCaP nanoparticles, which are encapsulated into the fibrin gel (GOx@MnCaP@fibrin). After spraying gel in the surgical cavity, GOx catalyzes the oxidation of glucose in residual IDH1 (R132H) cells and produces H2 O2 . The generated H2 O2 is further converted into highly lethal hydroxyl radicals (•OH) by a Mn2+ -mediated Fenton-like reaction to further kill the residual IDH1 (R132H) cells. The as-prepared starvation/chemodynamic therapeutic gel shows much higher therapeutic efficacy toward IDH1 (R132H) cells than IDH1 (WT) cells, and achieves long-term survival.
Collapse
Affiliation(s)
- Chunying Li
- Department of Dermatology and Venereology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, China
- Marshall Laboratory of Biomedical Engineering, International Cancer Center, Laboratory of Evolutionary Theranostics (LET), School of Biomedical Engineering, Shenzhen University Health Science Center, Shenzhen, 518060, China
| | - Yilin Wan
- Marshall Laboratory of Biomedical Engineering, International Cancer Center, Laboratory of Evolutionary Theranostics (LET), School of Biomedical Engineering, Shenzhen University Health Science Center, Shenzhen, 518060, China
| | - Yifan Zhang
- Marshall Laboratory of Biomedical Engineering, International Cancer Center, Laboratory of Evolutionary Theranostics (LET), School of Biomedical Engineering, Shenzhen University Health Science Center, Shenzhen, 518060, China
| | - Lian-Hua Fu
- Marshall Laboratory of Biomedical Engineering, International Cancer Center, Laboratory of Evolutionary Theranostics (LET), School of Biomedical Engineering, Shenzhen University Health Science Center, Shenzhen, 518060, China
| | - Nicholas Thomas Blum
- Marshall Laboratory of Biomedical Engineering, International Cancer Center, Laboratory of Evolutionary Theranostics (LET), School of Biomedical Engineering, Shenzhen University Health Science Center, Shenzhen, 518060, China
| | - Run Cui
- Marshall Laboratory of Biomedical Engineering, International Cancer Center, Laboratory of Evolutionary Theranostics (LET), School of Biomedical Engineering, Shenzhen University Health Science Center, Shenzhen, 518060, China
| | - Boda Wu
- Department of Dermatology and Venereology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, China
- Marshall Laboratory of Biomedical Engineering, International Cancer Center, Laboratory of Evolutionary Theranostics (LET), School of Biomedical Engineering, Shenzhen University Health Science Center, Shenzhen, 518060, China
| | - Rui Zheng
- Department of Dermatology and Venereology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, China
- Marshall Laboratory of Biomedical Engineering, International Cancer Center, Laboratory of Evolutionary Theranostics (LET), School of Biomedical Engineering, Shenzhen University Health Science Center, Shenzhen, 518060, China
| | - Jing Lin
- Marshall Laboratory of Biomedical Engineering, International Cancer Center, Laboratory of Evolutionary Theranostics (LET), School of Biomedical Engineering, Shenzhen University Health Science Center, Shenzhen, 518060, China
| | - Zhiming Li
- Department of Dermatology and Venereology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, China
| | - Peng Huang
- Department of Dermatology and Venereology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, China
- Marshall Laboratory of Biomedical Engineering, International Cancer Center, Laboratory of Evolutionary Theranostics (LET), School of Biomedical Engineering, Shenzhen University Health Science Center, Shenzhen, 518060, China
| |
Collapse
|
116
|
Ben Ahmed K, Hall LO, Goldgof DB, Gatenby R. Ensembles of Convolutional Neural Networks for Survival Time Estimation of High-Grade Glioma Patients from Multimodal MRI. Diagnostics (Basel) 2022; 12:diagnostics12020345. [PMID: 35204436 PMCID: PMC8871067 DOI: 10.3390/diagnostics12020345] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2021] [Revised: 01/13/2022] [Accepted: 01/24/2022] [Indexed: 11/16/2022] Open
Abstract
Glioma is the most common type of primary malignant brain tumor. Accurate survival time prediction for glioma patients may positively impact treatment planning. In this paper, we develop an automatic survival time prediction tool for glioblastoma patients along with an effective solution to the limited availability of annotated medical imaging datasets. Ensembles of snapshots of three dimensional (3D) deep convolutional neural networks (CNN) are applied to Magnetic Resonance Image (MRI) data to predict survival time of high-grade glioma patients. Additionally, multi-sequence MRI images were used to enhance survival prediction performance. A novel way to leverage the potential of ensembles to overcome the limitation of labeled medical image availability is shown. This new classification method separates glioblastoma patients into long- and short-term survivors. The BraTS (Brain Tumor Image Segmentation) 2019 training dataset was used in this work. Each patient case consisted of three MRI sequences (T1CE, T2, and FLAIR). Our training set contained 163 cases while the test set included 46 cases. The best known prediction accuracy of 74% for this type of problem was achieved on the unseen test set.
Collapse
Affiliation(s)
- Kaoutar Ben Ahmed
- Department of Computer Science and Engineering, University of South Florida, Tampa, FL 33620, USA; (L.O.H.); (D.B.G.)
- Correspondence:
| | - Lawrence O. Hall
- Department of Computer Science and Engineering, University of South Florida, Tampa, FL 33620, USA; (L.O.H.); (D.B.G.)
| | - Dmitry B. Goldgof
- Department of Computer Science and Engineering, University of South Florida, Tampa, FL 33620, USA; (L.O.H.); (D.B.G.)
| | - Robert Gatenby
- Department of Diagnostic Imaging and Interventional Radiology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL 33612, USA;
| |
Collapse
|
117
|
Liu L, Cheng M, Zhang T, Chen Y, Wu Y, Wang Q. Mesenchymal stem cell-derived extracellular vesicles prevent glioma by blocking M2 polarization of macrophages through a miR-744-5p/TGFB1-dependent mechanism. Cell Biol Toxicol 2022; 38:649-665. [PMID: 34978010 DOI: 10.1007/s10565-021-09652-7] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2021] [Accepted: 09/03/2021] [Indexed: 12/20/2022]
Abstract
AIM Our current study is conducted with intention to explore the regulatory mechanism of mesenchymal stem cell (MSC)-derived extracellular vesicle (EV)-miR-744-5p in glioma. METHODS Expression patterns of TGFB1, TGFBR1, and miR-744-5p were determined. EVs were isolated from human MSCs, which were characterized. Then, macrophages were co-cultured with MSCs with ectopic miR-744-5p expression to explore its role in cell proliferation, invasion, and migration capabilities. A nude mouse model of glioma xenograft was developed to observe the tumorigenesis and metastasis ability of glioma in vivo. RESULTS TGFB1 and TGFBR1 were upregulated in glioma. TGFB1 promoted M2 polarization of macrophages through theMAPK signaling, thereby promoting the progression of glioma. MSC-EVs suppressed TGFB1 expression in macrophages and inhibited M2 polarization of macrophages. MSC-EVs-miR-744-5p/TGFB1/MAPK axis inhibited M2 polarization of macrophages and reduced the malignant phenotypes of glioma cells. In vivo experiments verified that MSC-EVs-miR-744-5p inhibited the polarization of macrophage M2 and prevented glioma progression. CONCLUSION Taken together, MSC-EVs-miR-744-5p may suppress the MAPK signaling activity by downregulating TGFB1, and then inhibit polarization of macrophages M2, thereby preventing the progression of glioma. Graphical Headlights 1. TGFB1 promotes the M2 polarization of macrophages via the MAPK signaling. 2. miR-744-5p carried by MSC-EVs targets and inhibits TGFB1. 3. MSC-EV-miR-744-5p inhibits M2 polarization of macrophages to prevent glioma progression. 4. miR-744-5p loaded by MSC-EVs may be a preventive strategy against glioma.
Collapse
Affiliation(s)
- Ling Liu
- Department of Neurosurgery, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, No. 32, West Section 2, 1st Ring Road, Chengdu, 610072, Sichuan Province, People's Republic of China
| | - Meixiong Cheng
- Department of Neurosurgery, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, No. 32, West Section 2, 1st Ring Road, Chengdu, 610072, Sichuan Province, People's Republic of China
| | - Tian Zhang
- Department of Neurosurgery, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, No. 32, West Section 2, 1st Ring Road, Chengdu, 610072, Sichuan Province, People's Republic of China
| | - Yong Chen
- Department of Neurosurgery, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, No. 32, West Section 2, 1st Ring Road, Chengdu, 610072, Sichuan Province, People's Republic of China
| | - Yaqiu Wu
- Department of Neurosurgery Intensive Care Unit, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, No. 32, West Section 2, 1st Ring Road, Chengdu, 610072, Sichuan Province, People's Republic of China.
| | - Qi Wang
- Department of Neurosurgery, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, No. 32, West Section 2, 1st Ring Road, Chengdu, 610072, Sichuan Province, People's Republic of China.
| |
Collapse
|
118
|
Gritsch S, Batchelor TT, Gonzalez Castro LN. Diagnostic, therapeutic, and prognostic implications of the 2021 World Health Organization classification of tumors of the central nervous system. Cancer 2022; 128:47-58. [PMID: 34633681 DOI: 10.1002/cncr.33918] [Citation(s) in RCA: 176] [Impact Index Per Article: 58.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2021] [Revised: 08/16/2021] [Accepted: 08/17/2021] [Indexed: 12/17/2022]
Abstract
The 2016 revised fourth edition of the World Health Organization (WHO) classification of central nervous system (CNS) tumors incorporated molecular features with histologic grading, revolutionizing how oncologists conceptualize primary brain and spinal cord tumors as well as providing new insights into their management and prognosis. The 2021 revised fifth edition of the WHO classification further integrates molecular alterations for CNS tumor categorization, updating current understanding of the pathophysiology of many of these disease entities. Here, the authors review changes in the new classification for the most common primary adult tumors-gliomas (including astrocytomas, oligodendrogliomas, and ependymomas) and meningiomas-highlighting the key genomic alterations for each group classification to help clinicians interpret them as they consider therapeutic options-including clinical trials and targeted therapies-and discuss the prognosis of these tumors with their patients. The revised, updated 2021 WHO classification also further integrates molecular alterations in the classification of pediatric CNS tumors, but those are not covered in the current review.
Collapse
Affiliation(s)
- Simon Gritsch
- Cancer Center, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
| | - Tracy T Batchelor
- Department of Neurology, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts
- Center for Neuro-Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts
| | - L Nicolas Gonzalez Castro
- Cancer Center, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
- Department of Neurology, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts
- Center for Neuro-Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts
| |
Collapse
|
119
|
He X, Zhao W, Huang J, Xu J, Niu S, Zhang Q, Zhang N, Jin H, Shen G. Characteristics and trends of globally registered glioma clinical trials in the past 16 years. Ther Adv Neurol Disord 2022; 15:17562864221114355. [PMID: 35923217 PMCID: PMC9340892 DOI: 10.1177/17562864221114355] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2022] [Accepted: 06/29/2022] [Indexed: 11/15/2022] Open
Abstract
Background: Advancement in the treatment of glioma has been vacant since temozolomide has proved its therapeutic value in glioblastoma in 2005. Aim: To help investigators understand the landscape of glioma clinical research, we analyzed the characteristics and trends of globally registered glioma trials in the past decades. Methods: This is a cross-sectional analysis of glioma trials registered on ClinicalTrials.gov between January 2006 and December 2021. Characteristics regarding phase, enrollment number, study design and type, funding source, tumor site, pathology, patient status, age of population, trial purpose, and participating country were abstracted, and chronological shifts were analyzed. Results: There were 1531 registered glioma trials involved 58 participating countries. The trial purpose concerning surgery, radiotherapy, chemotherapy, targeted therapy, tumor-treating fields, immunotherapy, other antiglioma therapy and non-antiglioma research trial accounts for 3.5%, 6.5%, 9.5%, 28.9%, 2.0%, 16.4%, 12.5%, and 20.6%, respectively. In the past 16 years, the numbers of chemotherapy and targeted therapy trials declined; tumor-treating fields and immune checkpoint inhibitor application trials sprang at the latter half period; Immunotherapy, other antiglioma therapy and non-antiglioma research trials escalated (all above ptrend < 0.005). The trend also showed the phased trials registered diminishingly and that the trials which focused on glioblastoma registered incrementally (those two ptrend < 0.05). Among 784 drug therapy trials, it was included 45 cytotoxic drugs, 186 targeted drugs, 19 immune checkpoint inhibitors, 78 other drugs, and five immunomodulatory drugs. Two trials belonged to Bayesian adaptive randomized design. By the end of December 2021, 309 trials had publications. Only everolimus and tumor-treating fields exhibited meaningful survival benefit in specific glioma patients in phase 3 trials. Conclusion: Meaningful effective treatments regarding drugs or methods for glioma were difficult to be found. Bayesian adaptive platform trials may accelerate clinical research in glioma. Development of novel treatment modalities for glioma is still challenged.
Collapse
Affiliation(s)
- Xiaofang He
- Department of Radiation Oncology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, P.R. China
| | - Wenbin Zhao
- Department of Radiation Oncology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, P.R. China
| | - Jianwen Huang
- Department of Radiation Oncology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, P.R. China
| | - Jia Xu
- Department of Emergency, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, P.R. China
| | - Shaoqing Niu
- Department of Radiation Oncology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, P.R. China
| | - Qun Zhang
- Department of Radiation Oncology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, P.R. China
| | - Nu Zhang
- Department of Neurosurgery, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, P.R. China
- Guangdong Provincial Key Laboratory of Brain Function and Disease, Guangzhou, P.R. China
| | - Huawei Jin
- Department of Neurosurgery, The First Affiliated Hospital of Sun Yat-sen University, No.58, Zhongshan Road 2, Guangzhou 510080, Guangdong Province, P.R. China
| | - Guoping Shen
- Department of Radiation Oncology, The First Affiliated Hospital of Sun Yat-sen University, No.58, Zhongshan Road 2, Guangzhou 510080, Guangdong Province, P.R. China
| |
Collapse
|
120
|
Mohile NA, Messersmith H, Gatson NTN, Hottinger AF, Lassman AB, Morton J, Ney D, Nghiemphu PL, Olar A, Olson J, Perry J, Portnow J, Schiff D, Shannon A, Shih HA, Strowd R, van den Bent M, Ziu M, Blakeley J. Therapy for Diffuse Astrocytic and Oligodendroglial Tumors in Adults: ASCO-SNO Guideline. Neuro Oncol 2021. [DOI: 10.1093/neuonc/noab279] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Abstract
Purpose
To provide guidance to clinicians regarding therapy for diffuse astrocytic and oligodendroglial tumors in adults.
Methods
ASCO and the Society for Neuro-Oncology convened an Expert Panel and conducted a systematic review of the literature.
Results
Fifty-nine randomized trials focusing on therapeutic management were identified.
Recommendations
Adults with newly diagnosed oligodendroglioma, isocitrate dehydrogenase (IDH)–mutant, 1p19q codeleted CNS WHO grade 2 and 3 should be offered radiation therapy (RT) and procarbazine, lomustine, and vincristine (PCV). Temozolomide (TMZ) is a reasonable alternative for patients who may not tolerate PCV, but no high-level evidence supports upfront TMZ in this setting. People with newly diagnosed astrocytoma, IDH-mutant, 1p19q non-codeleted CNS WHO grade 2 should be offered RT with adjuvant chemotherapy (TMZ or PCV). People with astrocytoma, IDH-mutant, 1p19q non-codeleted CNS WHO grade 3 should be offered RT and adjuvant TMZ. People with astrocytoma, IDH-mutant, CNS WHO grade 4 may follow recommendations for either astrocytoma, IDH-mutant, 1p19q non-codeleted CNS WHO grade 3 or glioblastoma, IDH-wildtype, CNS WHO grade 4. Concurrent TMZ and RT should be offered to patients with newly diagnosed glioblastoma, IDH-wildtype, CNS WHO grade 4 followed by 6 months of adjuvant TMZ. Alternating electric field therapy, approved by the US Food and Drug Administration, should be considered for these patients. Bevacizumab is not recommended. In situations in which the benefits of 6-week RT plus TMZ may not outweigh the harms, hypofractionated RT plus TMZ is reasonable. In patients age ≥ 60 to ≥ 70 years, with poor performance status or for whom toxicity or prognosis are concerns, best supportive care alone, RT alone (for MGMTpromoter unmethylated tumors), or TMZ alone (for MGMT promoter methylated tumors) are reasonable treatment options. Additional information is available at www.asco.org/neurooncology-guidelines.
Collapse
Affiliation(s)
- Nimish A Mohile
- Department of Neurology and Wilmot Cancer Institute, University of Rochester Medical Center, Rochester, NY, USA
| | | | - Na Tosha N Gatson
- Banner MD Anderson Cancer Center, Phoenix, AZ, USA
- Geisinger Neuroscience Institute, Danville, PA, USA
| | - Andreas F Hottinger
- Department of Clinical Neurosciences and Oncology, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
| | | | - Jordan Morton
- University of Oklahoma Health Sciences, Oklahoma City, OK, USA
| | - Douglas Ney
- University of Colorado School of Medicine, Aurora, CO, USA
| | | | | | - Jeffery Olson
- Emory University, Atlanta, GA, USA
- Sunnybrook Health Sciences Center, Toronto, Ontario, Canada
| | - James Perry
- City of Hope National Medical Center, Duarte, CA, USA
| | - Jana Portnow
- City of Hope National Medical Center, Duarte, CA, USA
| | - David Schiff
- University of Virginia Medical Center, Charlottesville, VA, USA
| | | | | | - Roy Strowd
- Wake Forest Baptist Health Medical Center, Winston-Salem, NC, USA
| | - Martin van den Bent
- The Brain Tumor Center at Erasmus MC Cancer Institute, University Medical Center Rotterdam, Rotterdam, the Netherlands
| | - Mateo Ziu
- INOVA Neurosciences and Inova Schar Cancer Institute, Falls Church, VA, USA
| | | |
Collapse
|
121
|
Mohile NA, Messersmith H, Gatson NT, Hottinger AF, Lassman A, Morton J, Ney D, Nghiemphu PL, Olar A, Olson J, Perry J, Portnow J, Schiff D, Shannon A, Shih HA, Strowd R, van den Bent M, Ziu M, Blakeley J. Therapy for Diffuse Astrocytic and Oligodendroglial Tumors in Adults: ASCO-SNO Guideline. J Clin Oncol 2021; 40:403-426. [PMID: 34898238 DOI: 10.1200/jco.21.02036] [Citation(s) in RCA: 90] [Impact Index Per Article: 22.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
PURPOSE To provide guidance to clinicians regarding therapy for diffuse astrocytic and oligodendroglial tumors in adults. METHODS ASCO and the Society for Neuro-Oncology convened an Expert Panel and conducted a systematic review of the literature. RESULTS Fifty-nine randomized trials focusing on therapeutic management were identified. RECOMMENDATIONS Adults with newly diagnosed oligodendroglioma, isocitrate dehydrogenase (IDH)-mutant, 1p19q codeleted CNS WHO grade 2 and 3 should be offered radiation therapy (RT) and procarbazine, lomustine, and vincristine (PCV). Temozolomide (TMZ) is a reasonable alternative for patients who may not tolerate PCV, but no high-level evidence supports upfront TMZ in this setting. People with newly diagnosed astrocytoma, IDH-mutant, 1p19q non-codeleted CNS WHO grade 2 should be offered RT with adjuvant chemotherapy (TMZ or PCV). People with astrocytoma, IDH-mutant, 1p19q non-codeleted CNS WHO grade 3 should be offered RT and adjuvant TMZ. People with astrocytoma, IDH-mutant, CNS WHO grade 4 may follow recommendations for either astrocytoma, IDH-mutant, 1p19q non-codeleted CNS WHO grade 3 or glioblastoma, IDH-wildtype, CNS WHO grade 4. Concurrent TMZ and RT should be offered to patients with newly diagnosed glioblastoma, IDH-wildtype, CNS WHO grade 4 followed by 6 months of adjuvant TMZ. Alternating electric field therapy, approved by the US Food and Drug Administration, should be considered for these patients. Bevacizumab is not recommended. In situations in which the benefits of 6-week RT plus TMZ may not outweigh the harms, hypofractionated RT plus TMZ is reasonable. In patients age ≥ 60 to ≥ 70 years, with poor performance status or for whom toxicity or prognosis are concerns, best supportive care alone, RT alone (for MGMT promoter unmethylated tumors), or TMZ alone (for MGMT promoter methylated tumors) are reasonable treatment options. Additional information is available at www.asco.org/neurooncology-guidelines.
Collapse
Affiliation(s)
- Nimish A Mohile
- Department of Neurology and Wilmot Cancer Institute, University of Rochester Medical Center, Rochester, NY
| | | | - Na Tosha Gatson
- Banner MD Anderson Cancer Center, Phoenix, AZ.,Geisinger Neuroscience Institute. Danville, PA
| | - Andreas F Hottinger
- Departments of Clinical Neurosciences and Oncology, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
| | | | - Jordan Morton
- University of Oklahoma Health Sciences, Oklahoma City, OK
| | - Douglas Ney
- University of Colorado School of Medicine, Aurora, CO
| | | | | | | | - James Perry
- Sunnybrook Health Sciences Center, Toronto, Ontario, Canada
| | - Jana Portnow
- City of Hope National Medical Center, Duarte, CA
| | - David Schiff
- University of Virginia Medical Center, Charlottesville, VA
| | | | | | - Roy Strowd
- Wake Forest Baptist Health Medical Center, Winston-Salem, NC
| | - Martin van den Bent
- The Brain Tumor Center at Erasmus MC Cancer Institute, University Medical Center Rotterdam, Rotterdam, the Netherlands
| | - Mateo Ziu
- INOVA Neurosciences and Inova Schar Cancer Institute, Falls Church, VA
| | | |
Collapse
|
122
|
Antoni D, Feuvret L, Biau J, Robert C, Mazeron JJ, Noël G. Radiation guidelines for gliomas. Cancer Radiother 2021; 26:116-128. [PMID: 34953698 DOI: 10.1016/j.canrad.2021.08.006] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Gliomas are the most frequent primary brain tumour. The proximity of organs at risk, the infiltrating nature, and the radioresistance of gliomas have to be taken into account in the choice of prescribed dose and technique of radiotherapy. The management of glioma patients is based on clinical factors (age, KPS) and tumour characteristics (histology, molecular biology, tumour location), and strongly depends on available and associated treatments, such as surgery, radiation therapy, and chemotherapy. The knowledge of molecular biomarkers is currently essential, they are increasingly evolving as additional factors that facilitate diagnostics and therapeutic decision-making. We present the update of the recommendations of the French society for radiation oncology on the indications and the technical procedures for performing radiation therapy in patients with gliomas.
Collapse
Affiliation(s)
- D Antoni
- Service de radiothérapie, institut cancérologie Strasbourg Europe (ICANS), 17, rue Albert-Calmette, 67200 Strasbourg cedex, France.
| | - L Feuvret
- Service de radiothérapie, CHU Pitié-Salpêtrière, Assistance publique-hôpitaux de Paris (AP-HP), 47-83, boulevard de l'Hôpital, 75013 Paris, France
| | - J Biau
- Département universitaire de radiothérapie, centre Jean-Perrin, Unicancer, 58, rue Montalembert, BP 392, 63011 Clermont-Ferrand cedex 01, France
| | - C Robert
- Département de radiothérapie, institut de cancérologie Gustave-Roussy, 39, rue Camille-Desmoulin, 94800 Villejuif, France
| | - J-J Mazeron
- Service de radiothérapie, CHU Pitié-Salpêtrière, Assistance publique-hôpitaux de Paris (AP-HP), 47-83, boulevard de l'Hôpital, 75013 Paris, France
| | - G Noël
- Service de radiothérapie, institut cancérologie Strasbourg Europe (ICANS), 17, rue Albert-Calmette, 67200 Strasbourg cedex, France
| |
Collapse
|
123
|
Guo W, Ma S, Zhang Y, Liu H, Li Y, Xu JT, Yang B, Guan F. Genome-wide methylomic analyses identify prognostic epigenetic signature in lower grade glioma. J Cell Mol Med 2021; 26:449-461. [PMID: 34894053 PMCID: PMC8743658 DOI: 10.1111/jcmm.17101] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2021] [Revised: 11/17/2021] [Accepted: 11/19/2021] [Indexed: 12/19/2022] Open
Abstract
Glioma is the most malignant and aggressive type of brain tumour with high heterogeneity and mortality. Although some clinicopathological factors have been identified as prognostic biomarkers, the individual variants and risk stratification in patients with lower grade glioma (LGG) have not been fully elucidated. The primary aim of this study was to identify an efficient DNA methylation combination biomarker for risk stratification and prognosis in LGG. We conducted a retrospective cohort study by analysing whole genome DNA methylation data of 646 patients with LGG from the TCGA and GEO database. Cox proportional hazard analysis was carried out to screen and construct biomarker model that predicted overall survival (OS). The Kaplan‐Meier survival curves and time‐dependent ROC were constructed to prove the efficiency of the signature. Then, another independent cohort was used to further validate the finding. A two‐CpG site DNA methylation signature was identified by multivariate Cox proportional hazard analysis. Further analysis indicated that the signature was an independent survival predictor from other clinical factors and exhibited higher predictive accuracy compared with known biomarkers. This signature was significantly correlated with immune‐checkpoint blockade, immunotherapy‐related signatures and ferroptosis regulator genes. The expression pattern and functional analysis showed that these two genes corresponding with two methylation sites contained in the model were correlated with immune infiltration level, and involved in MAPK and Rap1 signalling pathway. The signature may contribute to improve the risk stratification of patients and provide a more accurate assessment for precision medicine in the clinic.
Collapse
Affiliation(s)
- Wenna Guo
- School of Life Sciences, Zhengzhou University, Zhengzhou, China.,School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, China
| | - Shanshan Ma
- School of Life Sciences, Zhengzhou University, Zhengzhou, China
| | - Yanting Zhang
- School of Life Sciences, Zhengzhou University, Zhengzhou, China
| | - Hongtao Liu
- School of Life Sciences, Zhengzhou University, Zhengzhou, China
| | - Ya Li
- School of Life Sciences, Zhengzhou University, Zhengzhou, China
| | - Ji-Tian Xu
- School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, China
| | - Bo Yang
- Department of Neurosurgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Fangxia Guan
- School of Life Sciences, Zhengzhou University, Zhengzhou, China
| |
Collapse
|
124
|
Qiu X, Chen Y, Bao Z, Chen L, Jiang T. Chemoradiotherapy with temozolomide vs. radiotherapy alone in patients with IDH wild-type and TERT promoter mutation WHO grade II/III gliomas: A prospective randomized study. Radiother Oncol 2021; 167:1-6. [PMID: 34902368 DOI: 10.1016/j.radonc.2021.12.009] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2021] [Revised: 11/22/2021] [Accepted: 12/05/2021] [Indexed: 12/15/2022]
Abstract
PURPOSE Patients with grade II/III diffuse glioma (lower grade glioma, LGG) with isocitrate dehydrogenase wild-type (IDH-wt) and telomerase reverse-transcriptase promoter mutation (TERTp-mut) experience shorter overall survival (OS) time than IDH mutant patients. The optimal treatment strategy for these patients is unclear. We compared the effects of radiotherapy (RT) alone vs. RT concurrent with temozolomide (TMZ) followed by adjuvant TMZ in these LGG patients. PATIENTS AND METHODS Thirty-seven LGG patients with IDH-wt and TERTp-mut were randomly allocated to either RT alone treatment (RT group, n = 18; 60 Gy in 30 daily fractions) or RT concurrent with TMZ (75 mg/m2/d, 7 d/week) followed by adjuvant TMZ (CRT group, n = 19). The median follow-up duration was 17 months. Log-rank test was used for OS and PFS comparisons. RESULTS The 1-year OS rate was 94.1% [95% confidence interval (CI) 82.9-100] in the CRT group and 74.6% (95% CI, 52.9-96.4) in the RT group. The median OS values in the CRT and RT groups were statistically different [25 vs. 17 months, respectively; hazard ratio (HR) 0.271; 95% CI, 0.092-0.793; P = 0.017], while PFS values were not (16 vs. 7 months, respectively; HR, 0.917; 95% CI, 0.397-2.120; P = 0.840). Multivariate analysis indicated that CRT treatment and female sex were associated with significantly longer OS (P = 0.001, P = 0.016, respectively). CONCLUSION CRT treatment for IDH-wt/TERTp-mut grade II/III gliomas resulted in significantly longer OS than RT alone. Female sex was a significant favorable prognostic factor.
Collapse
Affiliation(s)
- Xiaoguang Qiu
- Department of Radiation Oncology, Beijing Tiantan Hospital, Capital Medical University, China
| | - Yidong Chen
- Department of Radiation Oncology, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Zhaoshi Bao
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, China; Neurosurgical Institute, Capital Medical University, Beijing, China
| | - Li Chen
- Department of Radiation Oncology, Beijing Tiantan Hospital, Capital Medical University, China
| | - Tao Jiang
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, China; Neurosurgical Institute, Capital Medical University, Beijing, China.
| |
Collapse
|
125
|
Krauze AV, Camphausen K. Molecular Biology in Treatment Decision Processes-Neuro-Oncology Edition. Int J Mol Sci 2021; 22:13278. [PMID: 34948075 PMCID: PMC8703419 DOI: 10.3390/ijms222413278] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2021] [Revised: 12/02/2021] [Accepted: 12/03/2021] [Indexed: 11/30/2022] Open
Abstract
Computational approaches including machine learning, deep learning, and artificial intelligence are growing in importance in all medical specialties as large data repositories are increasingly being optimised. Radiation oncology as a discipline is at the forefront of large-scale data acquisition and well positioned towards both the production and analysis of large-scale oncologic data with the potential for clinically driven endpoints and advancement of patient outcomes. Neuro-oncology is comprised of malignancies that often carry poor prognosis and significant neurological sequelae. The analysis of radiation therapy mediated treatment and the potential for computationally mediated analyses may lead to more precise therapy by employing large scale data. We analysed the state of the literature pertaining to large scale data, computational analysis, and the advancement of molecular biomarkers in neuro-oncology with emphasis on radiation oncology. We aimed to connect existing and evolving approaches to realistic avenues for clinical implementation focusing on low grade gliomas (LGG), high grade gliomas (HGG), management of the elderly patient with HGG, rare central nervous system tumors, craniospinal irradiation, and re-irradiation to examine how computational analysis and molecular science may synergistically drive advances in personalised radiation therapy (RT) and optimise patient outcomes.
Collapse
Affiliation(s)
- Andra V. Krauze
- Radiation Oncology Branch, Center for Cancer Research, National Cancer Institute, NIH, 9000 Rockville Pike, Building 10, Bethesda, MD 20892, USA;
| | | |
Collapse
|
126
|
Greuter L, Guzman R, Soleman J. Pediatric and Adult Low-Grade Gliomas: Where Do the Differences Lie? CHILDREN (BASEL, SWITZERLAND) 2021; 8:1075. [PMID: 34828788 PMCID: PMC8624473 DOI: 10.3390/children8111075] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/03/2021] [Revised: 11/17/2021] [Accepted: 11/17/2021] [Indexed: 12/21/2022]
Abstract
Two thirds of pediatric gliomas are classified as low-grade (LGG), while in adults only around 20% of gliomas are low-grade. However, these tumors do not only differ in their incidence but also in their location, behavior and, subsequently, treatment. Pediatric LGG constitute 65% of pilocytic astrocytomas, while in adults the most commonly found histology is diffuse low-grade glioma (WHO II), which mostly occurs in eloquent regions of the brain, while its pediatric counterpart is frequently found in the infratentorial compartment. The different tumor locations require different skillsets from neurosurgeons. In adult LGG, a common practice is awake surgery, which is rarely performed on children. On the other hand, pediatric neurosurgeons are more commonly confronted with infratentorial tumors causing hydrocephalus, which more often require endoscopic or shunt procedures to restore the cerebrospinal fluid flow. In adult and pediatric LGG surgery, gross total excision is the primary treatment strategy. Only tumor recurrences or progression warrant adjuvant therapy with either chemo- or radiotherapy. In pediatric LGG, MEK inhibitors have shown promising initial results in treating recurrent LGG and several ongoing trials are investigating their role and safety. Moreover, predisposition syndromes, such as neurofibromatosis or tuberous sclerosis complex, can increase the risk of developing LGG in children, while in adults, usually no tumor growth in these syndromes is observed. In this review, we discuss and compare the differences between pediatric and adult LGG, emphasizing that pediatric LGG should not be approached and managed in the same way as adult LCG.
Collapse
Affiliation(s)
- Ladina Greuter
- Department of Neurosurgery, University Hospital of Basel, 4031 Basel, Switzerland; (R.G.); (J.S.)
- Department of Neurosurgery, King’s College Hospital, NHS Foundation Trust, London SE5 9RS, UK
| | - Raphael Guzman
- Department of Neurosurgery, University Hospital of Basel, 4031 Basel, Switzerland; (R.G.); (J.S.)
- Division of Pediatric Neurosurgery, University Children’s Hospital of Basel, 4056 Basel, Switzerland
- Faculty of Medicine, University of Basel, 4056 Basel, Switzerland
| | - Jehuda Soleman
- Department of Neurosurgery, University Hospital of Basel, 4031 Basel, Switzerland; (R.G.); (J.S.)
- Division of Pediatric Neurosurgery, University Children’s Hospital of Basel, 4056 Basel, Switzerland
- Faculty of Medicine, University of Basel, 4056 Basel, Switzerland
| |
Collapse
|
127
|
Liu Y, Liu S, Li G, Li Y, Chen L, Feng J, Yang Y, Jiang T, Qiu X. Association of high-dose radiotherapy with improved survival in patients with newly diagnosed low-grade gliomas. Cancer 2021; 128:1085-1092. [PMID: 34780673 PMCID: PMC9299029 DOI: 10.1002/cncr.34028] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2021] [Revised: 10/28/2021] [Accepted: 10/29/2021] [Indexed: 11/10/2022]
Abstract
BACKGROUND The radiation dose for patients with low-grade gliomas (LGGs) is controversial. The objective of this study was to investigate the impact of the radiation dose on survival for patients with LGGs and especially for molecularly defined subgroups. METHODS Three hundred fifty-one patients with newly diagnosed LGGs from the multicenter Chinese Glioma Cooperative Group received postoperative radiotherapy (RT) in 2005-2018. The RT dose, as a continuous variable, was entered into a Cox regression model using penalized spline regression to allow for a nonlinear relationship between the RT dose and overall survival (OS) or progression-free survival (PFS). Inverse probability of treatment weighting (IPTW)-adjusted propensity scores were used to correct for potential confounders. Dose effects on survival within IDH mutation and 1p/19q codeletion defined subgroups were analyzed. RESULTS The risk of mortality and disease progression decreased sharply until 54 Gy. High-dose RT (≥54 Gy) was associated with significantly better 5-year OS (81.7% vs 64.0%; hazard ratio [HR], 0.33; P < .001) and PFS (77.4% vs 54.5%; HR, 0.46; P < .001) than low-dose RT (<54 Gy). IPTW correction confirmed the associations (HR for OS, 0.44; P = .001; HR for PFS, 0.48; P = .003). High-dose RT was associated with longer PFS (HR, 0.25; P = .002; HR, 0.21; P = .039) and OS (HR, 0.27; P = .006; HR, 0.07; P = .017) in IDH-mutant/1p/19q noncodeleted and IDH wild-type subgroups, respectively. No significant difference in survival was observed with high-dose RT in the IDH-mutant/1p/19q codeleted subgroup. CONCLUSIONS High-dose RT (≥54 Gy) was effective in LGGs. Patients with an IDH mutation/1p/19q noncodeletion or IDH wild-type may need to be considered for high-dose RT. LAY SUMMARY The radiotherapy dose-response was observed in patients with low-grade gliomas, and high-dose radiotherapy (≥54 Gy) was associated with improved survival. Patients with an IDH mutation/1p/19q noncodeletion or wild-type IDH may have improved survival with the administration of high-dose radiotherapy.
Collapse
Affiliation(s)
- Yanwei Liu
- Department of Radiation Oncology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China.,Department of Molecular Neuropathology, Beijing Neurosurgical Institute, Capital Medical University, Beijing, China
| | - Shuai Liu
- Department of Radiation Oncology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China.,Department of Molecular Neuropathology, Beijing Neurosurgical Institute, Capital Medical University, Beijing, China
| | - Guanzhang Li
- Department of Molecular Neuropathology, Beijing Neurosurgical Institute, Capital Medical University, Beijing, China.,Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Yanong Li
- Department of Radiation Oncology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Li Chen
- Department of Radiation Oncology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Jin Feng
- Department of Radiation Oncology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Yong Yang
- Department of Radiation Oncology, Fujian Medical University Union Hospital, Fuzhou, China
| | - Tao Jiang
- Department of Molecular Neuropathology, Beijing Neurosurgical Institute, Capital Medical University, Beijing, China.,Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China.,National Clinical Research Center for Neurological Diseases, Beijing, China
| | - Xiaoguang Qiu
- Department of Radiation Oncology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China.,Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China.,National Clinical Research Center for Neurological Diseases, Beijing, China
| |
Collapse
|
128
|
Babikir H, Wang L, Shamardani K, Catalan F, Sudhir S, Aghi MK, Raleigh DR, Phillips JJ, Diaz AA. ATRX regulates glial identity and the tumor microenvironment in IDH-mutant glioma. Genome Biol 2021; 22:311. [PMID: 34763709 PMCID: PMC8588616 DOI: 10.1186/s13059-021-02535-4] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2021] [Accepted: 10/28/2021] [Indexed: 12/26/2022] Open
Abstract
BACKGROUND Recent single-cell transcriptomic studies report that IDH-mutant gliomas share a common hierarchy of cellular phenotypes, independent of genetic subtype. However, the genetic differences between IDH-mutant glioma subtypes are prognostic, predictive of response to chemotherapy, and correlate with distinct tumor microenvironments. RESULTS To reconcile these findings, we profile 22 human IDH-mutant gliomas using scATAC-seq and scRNA-seq. We determine the cell-type-specific differences in transcription factor expression and associated regulatory grammars between IDH-mutant glioma subtypes. We find that while IDH-mutant gliomas do share a common distribution of cell types, there are significant differences in the expression and targeting of transcription factors that regulate glial identity and cytokine elaboration. We knock out the chromatin remodeler ATRX, which suffers loss-of-function alterations in most IDH-mutant astrocytomas, in an IDH-mutant immunocompetent intracranial murine model. We find that both human ATRX-mutant gliomas and murine ATRX-knockout gliomas are more heavily infiltrated by immunosuppressive monocytic-lineage cells derived from circulation than ATRX-intact gliomas, in an IDH-mutant background. ATRX knockout in murine glioma recapitulates gene expression and open chromatin signatures that are specific to human ATRX-mutant astrocytomas, including drivers of astrocytic lineage and immune-cell chemotaxis. Through single-cell cleavage under targets and tagmentation assays and meta-analysis of public data, we show that ATRX loss leads to a global depletion in CCCTC-binding factor association with DNA, gene dysregulation along associated chromatin loops, and protection from therapy-induced senescence. CONCLUSIONS These studies explain how IDH-mutant gliomas from different subtypes maintain distinct phenotypes and tumor microenvironments despite a common lineage hierarchy.
Collapse
Affiliation(s)
- Husam Babikir
- Department of Neurological Surgery, University of California, Aaron Diaz, 1450 3rd Street, San Francisco, CA, 94158, USA
| | - Lin Wang
- Department of Neurological Surgery, University of California, Aaron Diaz, 1450 3rd Street, San Francisco, CA, 94158, USA
| | - Karin Shamardani
- Department of Neurological Surgery, University of California, Aaron Diaz, 1450 3rd Street, San Francisco, CA, 94158, USA
| | - Francisca Catalan
- Department of Neurological Surgery, University of California, Aaron Diaz, 1450 3rd Street, San Francisco, CA, 94158, USA
| | - Sweta Sudhir
- Department of Neurological Surgery, University of California, Aaron Diaz, 1450 3rd Street, San Francisco, CA, 94158, USA
| | - Manish K Aghi
- Department of Neurological Surgery, University of California, Aaron Diaz, 1450 3rd Street, San Francisco, CA, 94158, USA
| | - David R Raleigh
- Department of Neurological Surgery, University of California, Aaron Diaz, 1450 3rd Street, San Francisco, CA, 94158, USA
| | - Joanna J Phillips
- Department of Neurological Surgery, University of California, Aaron Diaz, 1450 3rd Street, San Francisco, CA, 94158, USA
| | - Aaron A Diaz
- Department of Neurological Surgery, University of California, Aaron Diaz, 1450 3rd Street, San Francisco, CA, 94158, USA.
| |
Collapse
|
129
|
Mellinghoff IK, Chang SM, Jaeckle KA, van den Bent M. Isocitrate Dehydrogenase Mutant Grade II and III Glial Neoplasms. Hematol Oncol Clin North Am 2021; 36:95-111. [PMID: 34711457 DOI: 10.1016/j.hoc.2021.08.008] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Mutations in isocitrate dehydrogenase (IDH) 1 or IDH2 occur in most of the adult low-grade gliomas and, less commonly, in cholangiocarcinoma, chondrosarcoma, acute myeloid leukemia, and other human malignancies. Cancer-associated mutations alter the function of the enzyme, resulting in production of R(-)-2-hydroxyglutarate and broad epigenetic dysregulation. Small molecule IDH inhibitors have received regulatory approval for the treatment of IDH mutant (mIDH) leukemia and are under development for the treatment of mIDH solid tumors. This article provides a current view of mIDH adult astrocytic and oligodendroglial tumors, including their clinical presentation and treatment, and discusses novel approaches and challenges toward improving the treatment of these tumors.
Collapse
Affiliation(s)
- Ingo K Mellinghoff
- Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, NY 10065, USA
| | - Susan M Chang
- Department of Neurological Surgery, University of California San Francisco, 505 Parnassus Room M 774SF, San Francisco, CA 94142-0112, USA
| | - Kurt A Jaeckle
- Department of Neurology and Oncology, Mayo Clinic Florida, Mangurian 4415, 4500 San Pablo Road, Jacksonville, FL 32224, USA
| | - Martin van den Bent
- Department of Neuro-onoclogy, Brain Tumor Center at Erasmus MC Cancer Institute, Nt-542, Dr Molenwaterplein 40, Rotterdam 3015 GD, The Netherlands.
| |
Collapse
|
130
|
Kagawa Y, Umaru BA, Kanamori M, Zama R, Shil SK, Miyazaki H, Kobayashi S, Wannakul T, Yang S, Tominaga T, Owada Y. Nuclear FABP7 regulates cell proliferation of wild-type IDH1 glioma through caveolae formation. Mol Oncol 2021; 16:289-306. [PMID: 34716958 PMCID: PMC8732344 DOI: 10.1002/1878-0261.13130] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2021] [Revised: 10/15/2021] [Accepted: 10/28/2021] [Indexed: 12/15/2022] Open
Abstract
Isocitrate dehydrogenase 1 (IDH1) is a key enzyme in cellular metabolism. IDH1 mutation (IDH1mut) is the most important genetic alteration in lower grade glioma, whereas glioblastoma (GB), the most common malignant brain tumor, often has wild‐type IDH1 (IDH1wt). Although there is no effective treatment yet for neither IDH1wt nor IDHmut GB, it is important to note that the survival span of IDH1wt GB patients is significantly shorter than those with IDH1mut GB. Thus, understanding IDH1wt GB biology and developing effective molecular‐targeted therapies is of paramount importance. Fatty acid‐binding protein 7 (FABP7) is highly expressed in GB, and its expression level is negatively correlated with survival in malignant glioma patients; however, the underlying mechanisms of FABP7 involvement in tumor proliferation are still unknown. In this study, we demonstrate that FABP7 is highly expressed and localized in nuclei in IDH1wt glioma. Wild‐type FABP7 (FABP7wt) overexpression in IDH1wt U87 cells increased cell proliferation rate, caveolin‐1 expression, and caveolae/caveosome formation. In addition, FABP7wt overexpression increased the levels of H3K27ac on the caveolin‐1 promoter through controlling the nuclear acetyl‐CoA level via the interaction with ACLY. Consistent results were obtained using a xenograft model transplanted with U87 cells overexpressing FABP7. Interestingly, in U87 cells with mutant FABP7 overexpression, both in vitro and in vivo phenotypes shown by FABP7wt overexpression were disrupted. Furthermore, IDH1wt patient GB showed upregulated caveolin‐1 expression, increased levels of histone acetylation, and increased levels of acetyl‐CoA compared with IDH1mut patient GB. Taken together, these data suggest that nuclear FABP7 is involved in cell proliferation of GB through caveolae function/formation regulated via epigenetic regulation of caveolin‐1, and this mechanism is critically important for IDH1wt tumor biology.
Collapse
Affiliation(s)
- Yoshiteru Kagawa
- Department of Organ Anatomy, Tohoku University Graduate School of Medicine, Sendai, Japan
| | | | - Masayuki Kanamori
- Department of Neurosurgery, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Ryo Zama
- Department of Organ Anatomy, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Subrata Kumar Shil
- Department of Organ Anatomy, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Hirofumi Miyazaki
- Department of Organ Anatomy, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Shuhei Kobayashi
- Department of Organ Anatomy, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Tunyanat Wannakul
- Department of Organ Anatomy, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Shuhan Yang
- Department of Organ Anatomy, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Teiji Tominaga
- Department of Neurosurgery, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Yuji Owada
- Department of Organ Anatomy, Tohoku University Graduate School of Medicine, Sendai, Japan
| |
Collapse
|
131
|
Feng S, Liu H, Dong X, Du P, Guo H, Pang Q. Identification and validation of an autophagy-related signature for predicting survival in lower-grade glioma. Bioengineered 2021; 12:9692-9708. [PMID: 34696669 PMCID: PMC8810042 DOI: 10.1080/21655979.2021.1985818] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
Abnormal levels of autophagy have been implicated in the pathogenesis of multiple diseases, including cancer. However, little is known about the role of autophagy-related genes (ARGs) in low-grade gliomas (LGG). Accordingly, the aims of this study were to assess the prognostic values of ARGs and to establish a genetic signature for LGG prognosis. Expression profile data from patients with and without primary LGG were obtained from The Cancer Genome Atlas (TCGA) and Genome Tissue Expression databases, respectively, and consensus clustering was used to identify clusters of patients with distinct prognoses. Nineteen differentially expressed ARGs were selected with threshold values of FDR < 0.05 and |log2 fold change (FC)| ≥ 2, and functional analysis revealed that these genes were associated with autophagy processes as expected. An autophagy-related signature was established using a Cox regression model of six ARGs that separated patients from TCGA training cohort into high- and low-risk groups. Univariate and multivariate Cox regression analysis indicated that the signature-based risk score was an independent prognostic factor. The signature was successfully validated using the TCGA testing, TCGA entire, and Chinese Glioma Genome Atlas cohorts. Stratified analyses demonstrated that the signature was associated with clinical features and prognosis, and gene set enrichment analysis revealed that autophagy- and cancer-related pathways were more enriched in high-risk patients than in low-risk patients. The prognostic value and expression of the six signature-related genes were also investigated. Thus, the present study constructed and validated an autophagy-related prognostic signature that could optimize individualized survival prediction in LGG patients.
Collapse
Affiliation(s)
- Shaobin Feng
- Department of Neurosurgery, Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
| | - Huiling Liu
- Department of Otolaryngology-Head and Neck Surgery, Shandong Provincial ENT Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
| | - Xushuai Dong
- Department of Neurosurgery, Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
| | - Peng Du
- Department of Neurosurgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China
| | - Hua Guo
- Department of Neurosurgery, Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China.,Department of Neurosurgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China
| | - Qi Pang
- Department of Neurosurgery, Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
| |
Collapse
|
132
|
Optimal Combinations of Chemotherapy and Radiotherapy in Low-Grade Gliomas: A Mathematical Approach. J Pers Med 2021; 11:jpm11101036. [PMID: 34683177 PMCID: PMC8537400 DOI: 10.3390/jpm11101036] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2021] [Revised: 09/30/2021] [Accepted: 10/11/2021] [Indexed: 12/16/2022] Open
Abstract
Low-grade gliomas (LGGs) are brain tumors characterized by their slow growth and infiltrative nature. Treatment options for these tumors are surgery, radiation therapy and chemotherapy. The optimal use of radiation therapy and chemotherapy is still under study. In this paper, we construct a mathematical model of LGG response to combinations of chemotherapy, specifically to the alkylating agent temozolomide and radiation therapy. Patient-specific parameters were obtained from longitudinal imaging data of the response of real LGG patients. Computer simulations showed that concurrent cycles of radiation therapy and temozolomide could provide the best therapeutic efficacy in-silico for the patients included in the study. The patient cohort was extended computationally to a set of 3000 virtual patients. This virtual cohort was subject to an in-silico trial in which matching the doses of radiotherapy to those of temozolomide in the first five days of each cycle improved overall survival over concomitant radio-chemotherapy according to RTOG 0424. Thus, the proposed treatment schedule could be investigated in a clinical setting to improve combination treatments in LGGs with substantial survival benefits.
Collapse
|
133
|
Rudà R, Bruno F, Ius T, Silvani A, Minniti G, Pace A, Lombardi G, Bertero L, Pizzolitto S, Pollo B, Conti Nibali M, Pellerino A, Migliore E, Skrap M, Bello L, Soffietti R. IDH wild-type grade 2 diffuse astrocytomas: prognostic factors and impact of treatments within molecular subgroups. Neuro Oncol 2021; 24:809-820. [PMID: 34651653 DOI: 10.1093/neuonc/noab239] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
BACKGROUND Prognostic factors and role of treatments are not well known in isocitrate dehydrogenase (IDH) wild-type (wt) grade 2 astrocytomas. The aim of this study was to define in these tumours clinical features, molecular characteristics and prognostic factors, with particular focus on molecular subgroups defined by cIMPACT-NOW update 3. METHODS We analysed 120 patients with confirmed diagnosis of grade 2 IDHwt astrocytoma according to WHO 2016, collected from 7 Italian centres between 1999 and 2017. RESULTS Median PFS and OS of the whole cohort were 18.9 and 32.6 months. Patients older than 40 years and patients with modest contrast enhancement on MRI had a shorter PFS and OS. Gross total resection yielded superior PFS and OS over non-gross total resection. PFS and OS of patients with either pTERT mutation or EGRF amplification were significantly shorter. The prognostic value of age, contrast enhancement on MRI and extent of surgery was different within the molecular subgroups. Gross total resection was associated with increased PFS (not reached versus 14 months, p = 0.023) and OS (117.9 versus 20 months, p = 0.023) in patients without EGFR amplification, and with increased OS in those without pTERT mutation (NR vs 53.7 months, p = 0.05). Conversely, for patients with EGFR amplification or pTERT mutation, gross total resection did not yield a significant survival benefit. CONCLUSION Patients without EGFR amplification and pTERT mutation could be observed after gross total resection.
Collapse
Affiliation(s)
- Roberta Rudà
- Department of Neuro-Oncology, University and City of Health and Science Hospital, Turin, Italy.,Department of Neurology, Castelfranco Veneto and Brain Tumor Board Treviso Hospital, Italy
| | - Francesco Bruno
- Department of Neuro-Oncology, University and City of Health and Science Hospital, Turin, Italy
| | - Tamara Ius
- Neurosurgery Unit, Department of Neurosciences, Santa Maria della Misericordia University Hospital, Udine, Italy
| | - Antonio Silvani
- Department of Neuro-Oncology, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan, Italy
| | - Giuseppe Minniti
- Radiation Oncology Unit, Department of Medicine, Surgery and Neurosciences, University Hospital, Siena, Italy
| | - Andrea Pace
- Neuro-Oncology Unit, Regina Elena National Cancer Institute, Rome, Italy
| | | | - Luca Bertero
- Pathology Unit, Department of Medical Sciences, University of Turin, Italy
| | - Stefano Pizzolitto
- Department of Pathology, Santa Maria della Misericordia University Hospital, Udine, Italy
| | - Bianca Pollo
- Neuropathology Unit, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan, Italy
| | - Marco Conti Nibali
- Neurosurgical Oncology Division, Department of Oncology and Hemato-Oncology, University of Milan, Italy
| | - Alessia Pellerino
- Department of Neuro-Oncology, University and City of Health and Science Hospital, Turin, Italy
| | - Enrica Migliore
- Unit of Cancer Epidemiology (CPO Piemonte), University of Turin, Turin, Italy
| | - Miran Skrap
- Neurosurgery Unit, Department of Neurosciences, Santa Maria della Misericordia University Hospital, Udine, Italy
| | - Lorenzo Bello
- Neurosurgical Oncology Division, Department of Oncology and Hemato-Oncology, University of Milan, Italy
| | - Riccardo Soffietti
- Department of Neuro-Oncology, University and City of Health and Science Hospital, Turin, Italy
| |
Collapse
|
134
|
Karschnia P, Weller J, Blobner J, Stoecklein VM, Dorostkar MM, Rejeski K, Forbrig R, Niyazi M, von Baumgarten L, Dietrich J, Tonn JC, Thon N. Subventricular zone involvement is associated with worse outcome in glioma WHO grade 2 depending on molecular markers. Sci Rep 2021; 11:20045. [PMID: 34625590 PMCID: PMC8501091 DOI: 10.1038/s41598-021-97714-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2021] [Accepted: 08/26/2021] [Indexed: 02/06/2023] Open
Abstract
Neural stem cells within the subventricular zone were identified as cells of origin driving growth of high-grade gliomas, and anatomical involvement of the subventricular zone has been associated with an inferior clinical outcome. Whether the association between poor outcome and subventricular zone involvement also applies to glioma of lower grades is unclear. We therefore analysed a retrospective cohort of 182 patients with glioma grade 2 (according to the WHO 2016 classification) including 78 individuals (43%) with subventricular zone involvement. Patients with and without subventricular zone involvement did not differ in regard to demographics, histopathology, and molecular markers. Notably, subventricular zone involvement was a negative prognostic marker for malignant progression and overall survival on uni- and multivariate analysis. When patients were stratified according to the cIMPACT-NOW update 6, subventricular zone involvement was negatively associated with outcome in IDH-wildtype astrocytomas and 1p19q-codeleted oligodendrogliomas but not in IDH-mutant astrocytomas. Collectively, subventricular zone involvement may represent a risk factor for worse outcome in glioma WHO grade 2 depending on the molecular tumor signature. The present data confirm the relevance of molecular glioma classifications as proposed by the cIMPACT-NOW update 6. These findings warrant evaluation in prospective cohorts.
Collapse
Affiliation(s)
- Philipp Karschnia
- Department of Neurosurgery, Ludwig Maximilians University, Marchioninistrasse 15, 81377, Munich, Germany. .,German Cancer Consortium (DKTK), Partner Site Munich, Munich, Germany. .,Department of Neurology, Massachusetts General Hospital Cancer Center, Harvard Medical School, Boston, MA, USA.
| | - Jonathan Weller
- Department of Neurosurgery, Ludwig Maximilians University, Marchioninistrasse 15, 81377, Munich, Germany.,German Cancer Consortium (DKTK), Partner Site Munich, Munich, Germany
| | - Jens Blobner
- Department of Neurosurgery, Ludwig Maximilians University, Marchioninistrasse 15, 81377, Munich, Germany.,German Cancer Consortium (DKTK), Partner Site Munich, Munich, Germany
| | - Veit M Stoecklein
- Department of Neurosurgery, Ludwig Maximilians University, Marchioninistrasse 15, 81377, Munich, Germany.,German Cancer Consortium (DKTK), Partner Site Munich, Munich, Germany
| | - Mario M Dorostkar
- German Cancer Consortium (DKTK), Partner Site Munich, Munich, Germany.,Center for Neuropathology and Prion Research, Ludwig-Maximilians-University, Munich, Germany
| | - Kai Rejeski
- German Cancer Consortium (DKTK), Partner Site Munich, Munich, Germany.,Department of Medicine III, Ludwig-Maximilians-University, Munich, Germany
| | - Robert Forbrig
- German Cancer Consortium (DKTK), Partner Site Munich, Munich, Germany.,Department of Neuroradiology, Ludwig-Maximilians-University, Munich, Germany
| | - Maximilian Niyazi
- German Cancer Consortium (DKTK), Partner Site Munich, Munich, Germany.,Department of Radiation Oncology, Ludwig-Maximilians-University, Munich, Germany
| | - Louisa von Baumgarten
- Department of Neurosurgery, Ludwig Maximilians University, Marchioninistrasse 15, 81377, Munich, Germany.,German Cancer Consortium (DKTK), Partner Site Munich, Munich, Germany.,Department of Neurology, Ludwig-Maximilians-University, Munich, Germany
| | - Jorg Dietrich
- Department of Neurology, Massachusetts General Hospital Cancer Center, Harvard Medical School, Boston, MA, USA
| | - Joerg-Christian Tonn
- Department of Neurosurgery, Ludwig Maximilians University, Marchioninistrasse 15, 81377, Munich, Germany.,German Cancer Consortium (DKTK), Partner Site Munich, Munich, Germany
| | - Niklas Thon
- Department of Neurosurgery, Ludwig Maximilians University, Marchioninistrasse 15, 81377, Munich, Germany.,German Cancer Consortium (DKTK), Partner Site Munich, Munich, Germany
| |
Collapse
|
135
|
Chen Z, Wu H, Yang H, Fan Y, Zhao S, Zhang M. Identification and validation of RNA-binding protein-related gene signature revealed potential associations with immunosuppression and drug sensitivity in glioma. Cancer Med 2021; 10:7418-7439. [PMID: 34482648 PMCID: PMC8525098 DOI: 10.1002/cam4.4248] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2021] [Revised: 07/31/2021] [Accepted: 08/22/2021] [Indexed: 12/11/2022] Open
Abstract
Background Glioma is the most common central nervous system tumor in adults, and a considerable part of them are high‐degree ones with high malignancy and poor prognosis. At present, the classification and treatment of glioma are mainly based on its histological characteristics, so studies at the molecular level are needed. Methods RNA‐seq data from The Cancer Genome Atlas (TCGA) datasets (n = 703) and Chinese Glioma Genome Atlas (CGGA) were utilized to find out the differentially expressed RNA‐binding proteins (RBPs) between normal cerebral tissue and glioma. A prediction system for the prognosis of glioma patients based on 11 RBPs was established and validated using uni‐ and multi‐variate Cox regression analyses. STITCH and CMap databases were exploited to identify putative drugs and their targets. Single sample gene set enrichment analysis (ssGSEA) was used to calculate scores of specific immune‐related gene sets. IC50 of over 20,000 compounds in 60 cancer cell lines was collected from the CellMiner database to test the drug sensitivity prediction value of the RBP‐based signature. Results We established a reliable prediction system for the prognosis of glioma patients based on 11 RBPs including THOC3, LSM11, SARNP, PABPC1L2B, SMN1, BRCA1, ZC3H8, DZIP1L, HEXIM2, LARP4B, and ZC3H12B. These RBPs were primarily associated with ribosome and post‐transcriptional regulation. RBP‐based risk scores were closely related to immune cells and immune function. We also confirmed the potential of the signature to predict the drug sensitivity of currently approved or evaluated drugs. Conclusions Differentially expressed RBPs in glioma can be used as a basis for prognosis prediction, new drugs screening and drug sensitivity prediction. As RBP‐based glioma risk scores were associated with immunity, immunotherapy may become an important treatment for glioma in the future.
Collapse
Affiliation(s)
- Zhuohui Chen
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, China
| | - Haiyue Wu
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, China
| | - Haojun Yang
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, China
| | - Yishu Fan
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, China
| | - Songfeng Zhao
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, China
| | - Mengqi Zhang
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, China
| |
Collapse
|
136
|
Fleming JL, Pugh SL, Fisher BJ, Lesser GJ, Macdonald DR, Bell EH, McElroy JP, Becker AP, Timmers CD, Aldape KD, Rogers CL, Doyle TJ, Werner-Wasik M, Bahary JP, Yu HHM, D'Souza DP, Laack NN, Sneed PK, Kwok Y, Won M, Mehta MP, Chakravarti A. Long-Term Report of a Comprehensive Molecular and Genomic Analysis in NRG Oncology/RTOG 0424: A Phase II Study of Radiation and Temozolomide in High-Risk Grade II Glioma. JCO Precis Oncol 2021; 5:PO.21.00112. [PMID: 34589661 PMCID: PMC8462570 DOI: 10.1200/po.21.00112] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2021] [Revised: 05/18/2021] [Accepted: 07/27/2021] [Indexed: 01/16/2023] Open
Abstract
PURPOSE This study sought to determine the prognostic significance of the WHO-defined glioma molecular subgroups along with additional alterations, including MGMT promoter methylation and mutations in ATRX, CIC, FUBP1, TERT, and TP53, in NRG/RTOG 0424 using long-term follow-up data. METHODS Mutations were determined using an Ion Torrent sequencing panel. 1p/19q co-deletion and MGMT promoter methylation were determined by Affymetrix OncoScan and Illumina 450K arrays. Progression-free survival (PFS) and overall survival (OS) were estimated using the Kaplan-Meier method and tested using the log-rank test. Hazard ratios were calculated using the Cox proportional hazard model. Multivariable analyses (MVAs) included patient pretreatment characteristics. RESULTS We obtained complete molecular data to categorize 80/129 eligible patients within the WHO subgroups. Of these, 26 (32.5%) were IDHmutant/co-deleted, 28 (35%) were IDHmutant/non-co-deleted, and 26 (32.5%) were IDHwild-type. Upon single-marker MVA, both IDHmutant subgroups were associated with significantly better OS and PFS (P values < .001), compared with the IDHwild-type subgroup. MGMT promoter methylation was obtained on 76 patients, where 58 (76%) were methylated and 18 (24%) were unmethylated. Single-marker MVAs demonstrated that MGMT promoter methylation was statistically significant for OS (P value < .001) and PFS (P value = .003). In a multimarker MVA, one WHO subgroup comparison (IDHmutant/co-deleted v IDHwild-type) was significant for OS (P value = .045), whereas MGMT methylation did not retain significance. CONCLUSION This study reports the long-term prognostic effect of the WHO molecular subgroups, MGMT promoter methylation, and other mutations in NRG/RTOG 0424. These results demonstrate that the WHO molecular classification and MGMT both serve as strong prognostic indicators, but that MGMT does not appear to add statistically significant prognostic value to the WHO subgrouping, above and beyond IDH and 1p/19q status.
Collapse
Affiliation(s)
| | - Stephanie L. Pugh
- NRG Oncology Statistics and Data Management Center, Philadelphia, PA
| | | | | | | | - Erica H. Bell
- Ohio State University Comprehensive Cancer Center, Columbus, OH
| | | | - Aline P. Becker
- Ohio State University Comprehensive Cancer Center, Columbus, OH
| | | | | | - C. Leland Rogers
- Barrow Neurological Institute, Phoenix, AZ (accruals under Arizona Oncology Services Foundation)
| | | | | | - Jean-Paul Bahary
- Centre Hospitalier de l`université De Montréal, Montreal, QC, Canada
| | | | | | | | | | - Young Kwok
- University of Maryland/Greenebaum Cancer Center, Baltimore, MA
| | - Minhee Won
- NRG Oncology Statistics and Data Management Center, Philadelphia, PA
| | | | | |
Collapse
|
137
|
Xu Z, Chen Q, Zeng X, Li M, Liao J. lnc-NLC1-C inhibits migration, invasion and autophagy of glioma cells by targeting miR-383 and regulating PRDX-3 expression. Oncol Lett 2021; 22:640. [PMID: 34386062 PMCID: PMC8299021 DOI: 10.3892/ol.2021.12901] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2020] [Accepted: 05/11/2021] [Indexed: 01/10/2023] Open
Abstract
Long non-coding RNAs (lncRNAs) serve an important role in tumor progression, and their abnormal expression is associated with tumor development. The lncRNA narcolepsy candidate region 1 gene C (lnc-NLC1-C) is involved in numerous types of cancer, but its biological function in glioma remains unknown. In the present study, lnc-NLC1-C expression was detected using reverse transcription-quantitative (RT-q)PCR in U251, SHG44, U87MG and U118MG glioma cells. U87MG cells were transfected with lnc-NLC1-C overexpression or interference vectors. Cell proliferation was detected using a Cell Counting Kit-8 assay. Cell migration and invasion were examined using a Transwell assay, while apoptosis, cell cycle and reactive oxygen species production were evaluated using flow cytometry, and the expression levels of lnc-NLC1-C, microRNA (miR)-383 and peroxiredoxin 3 (PRDX-3) were measured using western blotting and RT-qPCR. Rescue experiments were performed to verify the function of the lnc-NLC1-C/miR-383/PRDX-3 axis. The highest expression levels of lnc-NLC1-C were identified in U87MG glioma cells. Overexpression of lnc-NLC1-C expression promoted cell proliferation, G1 phase blocking, migration and invasion, while inhibiting apoptosis and autophagy in U87MG cells. Mechanistically, miR-383 could bind to lnc-NLC1-C to regulate PRDX-3 expression and improve its oncogenic effect. Rescue experiments confirmed that the lnc-NLC1-C/miR-383/PRDX-3 axis was involved in the molecular mechanism of glioma progression. Therefore, lnc-NLC1-C may be a tumor promoter that affects multiple biological functions, such as migration, invasion and autophagy, in glioma cells.
Collapse
Affiliation(s)
- Zhou Xu
- Department of Neurosurgery, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, P.R. China
| | - Qianxue Chen
- Department of Neurosurgery, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, P.R. China
| | - Xingnuo Zeng
- Department of Nephrology and Rheumatology, The Central Hospital of Wuhan, Huazhong University of Science and Technology, Wuhan, Hubei 430014, P.R. China
| | - Mingchang Li
- Department of Neurosurgery, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, P.R. China
| | - Jianming Liao
- Department of Neurosurgery, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, P.R. China
| |
Collapse
|
138
|
Mellinghoff IK, Penas-Prado M, Peters KB, Burris HA, Maher EA, Janku F, Cote GM, de la Fuente MI, Clarke JL, Ellingson BM, Chun S, Young RJ, Liu H, Choe S, Lu M, Le K, Hassan I, Steelman L, Pandya SS, Cloughesy TF, Wen PY. Vorasidenib, a Dual Inhibitor of Mutant IDH1/2, in Recurrent or Progressive Glioma; Results of a First-in-Human Phase I Trial. Clin Cancer Res 2021; 27:4491-4499. [PMID: 34078652 PMCID: PMC8364866 DOI: 10.1158/1078-0432.ccr-21-0611] [Citation(s) in RCA: 157] [Impact Index Per Article: 39.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2021] [Revised: 04/01/2021] [Accepted: 05/25/2021] [Indexed: 11/16/2022]
Abstract
PURPOSE Lower grade gliomas (LGGs) are malignant brain tumors. Current therapy is associated with short- and long-term toxicity. Progression to higher tumor grade is associated with contrast enhancement on MRI. The majority of LGGs harbor mutations in the genes encoding isocitrate dehydrogenase 1 or 2 (IDH1/IDH2). Vorasidenib (AG-881) is a first-in-class, brain-penetrant, dual inhibitor of the mutant IDH1 and mutant IDH2 enzymes. PATIENTS AND METHODS We conducted a multicenter, open-label, phase I, dose-escalation study of vorasidenib in 93 patients with mutant IDH1/2 (mIDH1/2) solid tumors, including 52 patients with glioma that had recurred or progressed following standard therapy. Vorasidenib was administered orally, once daily, in 28-day cycles until progression or unacceptable toxicity. Enrollment is complete; this trial is registered with ClinicalTrials.gov, NCT02481154. RESULTS Vorasidenib showed a favorable safety profile in the glioma cohort. Dose-limiting toxicities of elevated transaminases occurred at doses ≥100 mg and were reversible. The protocol-defined objective response rate per Response Assessment in Neuro-Oncology criteria for LGG in patients with nonenhancing glioma was 18% (one partial response, three minor responses). The median progression-free survival was 36.8 months [95% confidence interval (CI), 11.2-40.8] for patients with nonenhancing glioma and 3.6 months (95% CI, 1.8-6.5) for patients with enhancing glioma. Exploratory evaluation of tumor volumes in patients with nonenhancing glioma showed sustained tumor shrinkage in multiple patients. CONCLUSIONS Vorasidenib was well tolerated and showed preliminary antitumor activity in patients with recurrent or progressive nonenhancing mIDH LGG.
Collapse
Affiliation(s)
- Ingo K Mellinghoff
- Department of Neurology and Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, New York.
| | | | - Katherine B Peters
- The Preston Robert Tisch Brain Tumor Center, Duke University Medical Center, Durham, North Carolina
| | | | - Elizabeth A Maher
- Department of Neurology and Neurotherapeutics, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Filip Janku
- Department of Investigational Cancer Therapeutics, Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Gregory M Cote
- Massachusetts General Hospital Cancer Center, Boston, Massachusetts
| | - Macarena I de la Fuente
- Department of Neurology and Sylvester Comprehensive Cancer Center, University of Miami, Miami, Florida
| | - Jennifer L Clarke
- Weill Institute for Neurosciences, University of California San Francisco, San Francisco, California
| | - Benjamin M Ellingson
- UCLA Brain Tumor Imaging Laboratory, Department of Radiological Sciences, David Geffen School of Medicine, University of California, Los Angeles, California
| | - Saewon Chun
- Department of Neurology, Ronald Reagan UCLA Medical Center, University of California, Los Angeles, California
| | - Robert J Young
- Neuroradiology Service, Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Hua Liu
- Agios Pharmaceuticals, Inc., Cambridge, Massachusetts
| | - Sung Choe
- Agios Pharmaceuticals, Inc., Cambridge, Massachusetts
| | - Min Lu
- Agios Pharmaceuticals, Inc., Cambridge, Massachusetts
| | - Kha Le
- Agios Pharmaceuticals, Inc., Cambridge, Massachusetts
| | - Islam Hassan
- Agios Pharmaceuticals, Inc., Cambridge, Massachusetts
| | - Lori Steelman
- Agios Pharmaceuticals, Inc., Cambridge, Massachusetts
| | | | - Timothy F Cloughesy
- Department of Neurology, Ronald Reagan UCLA Medical Center, University of California, Los Angeles, California
| | - Patrick Y Wen
- Center for Neuro-Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
| |
Collapse
|
139
|
Adjuvant and concurrent temozolomide for 1p/19q non-co-deleted anaplastic glioma. Lancet Oncol 2021; 22:e345. [PMID: 34339648 DOI: 10.1016/s1470-2045(21)00378-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2021] [Revised: 06/11/2021] [Accepted: 06/14/2021] [Indexed: 01/25/2023]
|
140
|
ITGB2 as a prognostic indicator and a predictive marker for immunotherapy in gliomas. Cancer Immunol Immunother 2021; 71:645-660. [PMID: 34313821 DOI: 10.1007/s00262-021-03022-2] [Citation(s) in RCA: 46] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2021] [Accepted: 07/20/2021] [Indexed: 12/15/2022]
Abstract
PURPOSE Glioma is the most common primary tumor in the brain, accounting for 81% of intracranial malignancies. Nowadays, cancer immunotherapy has become a novel and revolutionary treatment for patients with advanced, highly aggressive tumors. However, to date, there are no effective biomarkers to reflect the response of glioma patients to immunotherapy. In this study, we aimed to assess the clinical predictive value of ITGB2 in patients with glioma. METHODS The correlation between ITGB2 expression levels and glioma progression was explored and validated using data from CGGA, TCGA, GEO datasets, and patient samples from our hospital. Univariate and multivariate cox regression models were developed to determine the predictive role of ITGB2 on the prognosis of patients with glioma. The relationship between ITGB2 and immune activation was then analyzed. Finally, we predicted the immunotherapy response in both high and low ITGB2 expression subgroups. RESULTS ITGB2 was significantly elevated in gliomas with higher malignancy and predicted poor prognosis. In multivariate analysis, the hazard ratio for ITGB2 expression (low versus high) was 0.71 with 95% CI (0.59-0.85) (P < 0.001). Furthermore, we found that ITGB2 stratified glioma patients into high and low ITGB2 expression subgroups, exhibiting different clinical outcomes and immune activation status. At last, we demonstrated that glioma patients with high ITGB2 expression levels had better immunotherapy response. CONCLUSIONS This study demonstrated ITGB2 as a novel predictor for clinical prognosis and response to immunotherapy in gliomas. Assessing expression levels of ITGB2 is a promising method to discover patients that may benefit from immunotherapy.
Collapse
|
141
|
Taillandier L, Obara T, Duffau H. What Does Quality of Care Mean in Lower-Grade Glioma Patients: A Precision Molecular-Based Management of the Tumor or an Individualized Medicine Centered on Patient's Choices? Front Oncol 2021; 11:719014. [PMID: 34354956 PMCID: PMC8329449 DOI: 10.3389/fonc.2021.719014] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2021] [Accepted: 07/02/2021] [Indexed: 12/02/2022] Open
Affiliation(s)
- Luc Taillandier
- Centre de Recherche en Automatique Nancy France - UMR 7039 - BioSiS Department, Faculty of Medicine, Université de Lorraine, Vandoeuvre-lès-Nancy, France
- Neurology Departement, Neurooncology Unit, CHRU, Nancy, France
| | - Tiphaine Obara
- Centre de Recherche en Automatique Nancy France - UMR 7039 - BioSiS Department, Faculty of Medicine, Université de Lorraine, Vandoeuvre-lès-Nancy, France
- Neurology Departement, Neurooncology Unit, CHRU, Nancy, France
| | - Hugues Duffau
- Department of Neurosurgery, Gui de Chauliac Hospital, Montpellier University Medical Center, Montpellier, France
- Team “Plasticity of Central Nervous System, Stem Cells and Glial Tumors”, National Institute for Health and Medical Research (INSERM), U1191 Laboratory, Institute of Functional Genomics, University of Montpellier, Montpellier, France
| |
Collapse
|
142
|
Anand S, Chatterjee A, Gupta T, Panda P, Moiyadi A, Epari S, Patil V, Krishnatry R, Goda JS, Jalali R. Upfront Therapy of Aggressive/High-Risk Low-Grade Glioma: Single-Institution Outcome Analysis of Temozolomide-Based Radio-Chemotherapy and Adjuvant Chemotherapy. World Neurosurg 2021; 154:e176-e184. [PMID: 34245877 DOI: 10.1016/j.wneu.2021.07.002] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2021] [Revised: 06/29/2021] [Accepted: 07/01/2021] [Indexed: 01/12/2023]
Abstract
OBJECTIVE To report clinical outcomes of temozolomide (TMZ)-based radio-chemotherapy and adjuvant chemotherapy in patients with aggressive/high-risk low-grade glioma (LGG). METHODS Medical records of patients defined as aggressive/high-risk LGG based on clinicoradiologic and/or histomorphologic features treated between 2009 and 2016 in an academic neuro-oncology unit with upfront postoperative radiotherapy at time of initial diagnosis with concurrent and adjuvant TMZ were reviewed, retrospectively. RESULTS In total, 64 patients with median age of 38 years at initial diagnosis were included. Histomorphologically, patients were classified into oligodendroglioma, mixed oligoastrocytoma, and astrocytoma. Molecular markers such as isocitrate dehydrogenase (IDH) mutation and 1p/19q codeletion were used to classify 37 of 64 (58%) patients into molecularly defined entities comprising oligodendroglioma (IDH-mutant with 1p/19q codeletion), IDH-mutant astrocytoma (immunohistochemistry or gene sequencing), and IDH-wild-type astrocytoma (gene sequencing). All 64 patients completed planned conventionally fractionated focal conformal radiotherapy (median dose 55.8 Gy) with concurrent TMZ. Fifty-nine patients received further adjuvant TMZ for a median of 12 cycles. Adjuvant TMZ was stopped prematurely in 6 (9%) patients due to toxicity or early disease progression. At a median follow-up of 56.7 months, 5-year Kaplan-Meier estimates of progression-free survival and overall survival for the study cohort were 74.6% and 84.3%, respectively. Five-year overall survival was 87.5%, 90.4%, and 71.9% for oligodendroglioma, mixed oligoastrocytoma, and astrocytoma, respectively (P = 0.42) Similar estimates for molecularly defined oligodendroglioma, IDH-mutant astrocytoma, and IDH-wild-type astrocytoma were 85.8%, 90%, and 66.7%, respectively (P = 0.87). CONCLUSIONS Upfront TMZ-based concurrent radio-chemotherapy and adjuvant TMZ chemotherapy provides acceptable survival outcomes in aggressive/high-risk LGG with modest toxicity.
Collapse
Affiliation(s)
- Sachith Anand
- Department of Radiation Oncology, TMH/ACTREC, Tata Memorial Centre, Homi Bhabha National Institute (HBNI), Mumbai, India
| | - Abhishek Chatterjee
- Department of Radiation Oncology, TMH/ACTREC, Tata Memorial Centre, Homi Bhabha National Institute (HBNI), Mumbai, India
| | - Tejpal Gupta
- Department of Radiation Oncology, TMH/ACTREC, Tata Memorial Centre, Homi Bhabha National Institute (HBNI), Mumbai, India.
| | - Pankaj Panda
- Department of Clinical Research Secretariat, TMH/ACTREC, Tata Memorial Centre, Homi Bhabha National Institute (HBNI), Mumbai, India
| | - Aliasgar Moiyadi
- Department of Neuro-surgical Oncology, TMH/ACTREC, Tata Memorial Centre, Homi Bhabha National Institute (HBNI), Mumbai, India
| | - Sridhar Epari
- Department of Pathology, TMH/ACTREC, Tata Memorial Centre, Homi Bhabha National Institute (HBNI), Mumbai, India
| | - Vijay Patil
- Department of Medical Oncology, TMH/ACTREC, Tata Memorial Centre, Homi Bhabha National Institute (HBNI), Mumbai, India
| | - Rahul Krishnatry
- Department of Radiation Oncology, TMH/ACTREC, Tata Memorial Centre, Homi Bhabha National Institute (HBNI), Mumbai, India
| | - Jayant Sastri Goda
- Department of Radiation Oncology, TMH/ACTREC, Tata Memorial Centre, Homi Bhabha National Institute (HBNI), Mumbai, India
| | - Rakesh Jalali
- Department of Radiation Oncology, TMH/ACTREC, Tata Memorial Centre, Homi Bhabha National Institute (HBNI), Mumbai, India
| |
Collapse
|
143
|
Zhao K, Sun G, Wang Q, Xue Z, Liu G, Xia Y, Yao A, Zhao Y, You N, Yang C, Xu B. The Diagnostic Value of Conventional MRI and CT Features in the Identification of the IDH1-Mutant and 1p/19q Co-Deletion in WHO Grade II Gliomas. Acad Radiol 2021; 28:e189-e198. [PMID: 32359929 DOI: 10.1016/j.acra.2020.03.008] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2020] [Revised: 03/05/2020] [Accepted: 03/06/2020] [Indexed: 12/21/2022]
Abstract
RATIONALE AND OBJECTIVES The classification of patients based on pathology and molecular features is important for improving WHO grade II glioma patient prognosis, especially for the initially diagnosed patients. Less invasive and more convenient methods for the prediction of the pathological type and gene status are desired. MATERIALS AND METHODS This study investigates the ability to use conventional magnetic resonance imaging (MRI) and computed tomography (CT) features for determining the Isocitrate Dehydrogenase (IDH)-mutant and 1p/19q-codeletion status, through a retrospective review of information obtained from 189 WHO grade II glioma patients. Diffuse astrocytoma (IDH-mutant), Diffuse astrocytoma (IDH- wildtype) and Oligodendroglioma (IDH-mutant and 1p/19q co-deletion) were included in this cohort. All patients were divided into IDH-mutant group and IDH-wildtype group according to the IDH R132H mutation status. Moreover, all patients were divided into 1p/19q co-deletion group and 1p/19q non-codeletion group according to the 1p and 19q chromosome status. Patients underwent pre-operative CT and MRI scans, followed by operation and histopathological analyses, including immunohistochemistry and polymerase chain reaction analysis for IDH mutants, and fluorescence capillary electrophoresis analysis for the 1p/19q co-deletion. The χ2 test, logistical regression and receiver operating characteristic curve analysis were conducted for statistical analysis. RESULTS IDH-mutant group patients exhibited a higher calcification frequency (25.2% vs 2.4%, p = 0.006) and lower frequency of T1 enhancement (20.4% vs 38.1%, p = 0.028) comparing patients in IDH-wildtype group, while 1p/19q co-deletion group patients exhibited a higher calcification frequency (46.67% vs 2.6%, p < 0.001) and lower homogenous signal frequency in T2WI (12.0% vs 31.6%, p = 0.014), sharp lesion margins (14.7% vs 43.0%, p = 0.010), T2/fluid attenuated inversion recovery mismatch signs (22.7% vs 50.9%, p = 0.001), and subventricular zone involvement (64.0% vs 15.8%, p = 0.021) comparing patients in 1p/19q non-codeletion group. According to the results of receiver operating characteristic analysis, these features were observed to have certain diagnostic abilities, especially with regard to combination parameters, which had a high diagnostic capability, with an area under the curve of 0.848. CONCLUSION Conventional MRI and CT features, which still represent the most convenient and widely used predictive method, might be a promising noninvasive predictor for differentiating between varied WHO grade II gliomas. Patients with calcification and T1 nonenhancement are more likely to be IDH-mutant. Moreover, patients with noncalcification, homogenous signal, sharp lesion margins, subventricular zone involvement on T2 and T2/fluid attenuated inversion recovery mismatch signs are more likely to be 1p/19q non-codeletion.
Collapse
|
144
|
Ouyang J, Jiang Y, Deng C, Zhong Z, Lan Q. Doxorubicin Delivered via ApoE-Directed Reduction-Sensitive Polymersomes Potently Inhibit Orthotopic Human Glioblastoma Xenografts in Nude Mice. Int J Nanomedicine 2021; 16:4105-4115. [PMID: 34163162 PMCID: PMC8214541 DOI: 10.2147/ijn.s314895] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2021] [Accepted: 05/19/2021] [Indexed: 12/14/2022] Open
Abstract
Purpose Glioblastoma multiforme (GBM) poorly responds to chemotherapy owing to the existence of blood-brain barriers (BBB). It has been a long desire to develop BBB-permeable vehicles to facilitate drug targeting to GBM. Method and Results Here, we report that doxorubicin hydrochloride loaded in ApoE peptide-functionalized reduction-sensitive polymersomes (ApoE-PS-DOX) induces potent therapy of orthotopic U-87 MG model in nude mice. ApoE-PS-DOX with varying amount of ApoE (10~30 mol%) all had stable DOX loading and small sizes (< 90 nm). As revealed by flow cytometry, confocal microscopy, apoptosis and MTT assays, ApoE-PS-DOX with 20 mol.% ApoE induced the best cellular uptake and inhibitory effect to U-87 MG cells, which were much better than the non-targeted PS-DOX and liposomal doxorubicin (Lipo-DOX) used in the clinic. ApoE-PS-DOX revealed a pharmacokinetic profile comparable to PS-DOX but induced considerably better growth inhibition of orthotopically xenografted U-87 MG tumors in nude mice than PS-DOX and Lipo-DOX, leading to significant survival benefits with a median survival time of 44 days, which was almost doubled relative to the phosphate-buffered saline (PBS) group. Moreover, in contrast to mice treated with Lipo-DOX and PS-DOX, ApoE-PS-DOX group exhibited little body weight loss, signifying that ApoE-PS-DOX not only has low side effects but also can effectively inhibit glioblastoma invasion. Conclusion This ApoE-docked multifunctional polymersomal doxorubicin induces potent and safe chemotherapy of orthotopic U-87 MG model in nude mice offering an alternative treatment modality for GBM.
Collapse
Affiliation(s)
- Jia Ouyang
- Department of Neurosurgery, The Second Affiliated Hospital of Soochow University, Suzhou, 215004, People's Republic of China
| | - Yu Jiang
- Biomedical Polymers Laboratory, College of Chemistry, Chemical Engineering and Materials Science, and State Key Laboratory of Radiation Medicine and Protection, Soochow University, Suzhou, 215123, People's Republic of China
| | - Chao Deng
- Biomedical Polymers Laboratory, College of Chemistry, Chemical Engineering and Materials Science, and State Key Laboratory of Radiation Medicine and Protection, Soochow University, Suzhou, 215123, People's Republic of China
| | - Zhiyuan Zhong
- Biomedical Polymers Laboratory, College of Chemistry, Chemical Engineering and Materials Science, and State Key Laboratory of Radiation Medicine and Protection, Soochow University, Suzhou, 215123, People's Republic of China
| | - Qing Lan
- Department of Neurosurgery, The Second Affiliated Hospital of Soochow University, Suzhou, 215004, People's Republic of China
| |
Collapse
|
145
|
Abstract
PURPOSE OF REVIEW This review discusses current and investigative strategies for targeting DNA repair in the management of glioma. RECENT FINDINGS Recent strategies in glioma treatment rely on the production of overwhelming DNA damage and inhibition of repair mechanisms, resulting in lethal cytotoxicity. Many strategies are effective in preclinical glioma models while clinical feasibility remains under investigation. The presence of glioma biomarkers, including IDH mutation and/or MGMT promoter methylation, may confer particular susceptibility to DNA damage and inhibition of repair. These biomarkers have been adopted as eligibility criteria in the design of multiple ongoing clinical trials. Targeting DNA repair mechanisms with novel agents or therapeutic combinations is a promising approach to the treatment of glioma. Further investigations are underway to optimize this approach in the clinical setting.
Collapse
|
146
|
Chen Y, Wang Y, He Q, Wang W, Zhang T, Wang Z, Dong J, Lan Q, Zhao J. Integrative analysis of TP73 profile prognostic significance in WHO grade II/III glioma. Cancer Med 2021; 10:4644-4657. [PMID: 34121368 PMCID: PMC8267133 DOI: 10.1002/cam4.4016] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2021] [Revised: 04/24/2021] [Accepted: 04/26/2021] [Indexed: 12/17/2022] Open
Abstract
Due to the extremely intrinsic heterogeneity among glioma patients, the outcomes of these patients are tremendously different. Therefore, the exploitation of novel biomarker classification of glioma is vitally important for deep insight into the essence and predicting the prognosis of glioma. We aim to analyze the correlation between TP73 mRNA expression, DNA methylated alteration and the prognosis of WHO grade II/III glioma, utilizing bioinformatics to evaluate its significance as a risk‐factor in predicting the prognosis of these glioma patients. The analysis found that TP73 expression was positively correlated with the grade of glioma, and showed a strong correlation with glioma molecular classification, which revealed significantly higher TP73 expression in IDH‐wildtype than in IDH‐mutant subtype of WHO grade II/III glioma. Cox regression analysis indicated that high expression of TP73 shared an independent high‐risk factor impacting the prognosis of WHO grade II/III glioma. We discovered 8 DNA promoter methylation sites with prognostic significance, which were negatively associated with TP73 expression, and positively associated with beneficial overall survival (OS) and progression‐free survival (PFS). Integrating with four independent glioma datasets, subsequent Meta‐analysis verified that low expression of TP73 was closely related to favorable OS, especially in IDH‐mutant subtype. Moreover, we found that 1p/19qCodel/TP73low subgroup shared the most favorable OS, 1p/19qNon−codel/TP73high subgroup suffered the worst OS. Meanwhile, the enrichment analysis of TP73‐related differential mRNAs demonstrated that TP73 aberration in WHO grade II/III glioma might be closely related to cell cycle and P53 signaling pathways. Finally, TP73 expression of 53 glioma specimens was measured by qRT‐PCR, which was consistent with the previous analytical result, and TP73 high‐expression subgroup suffered worse PFS than TP73 low‐expression subgroup. In summary, our funding supports that TP73 gene can perform as a reliable biomarker to evaluate the survival outcome of patients diagnosed with WHO grade II/III glioma.
Collapse
Affiliation(s)
- Yanming Chen
- Department of Neurosurgery, The Second Affiliated Hospital of Soochow University, Suzhou, China.,China National Clinical Research Center for Neurological Diseases, Beijing, China
| | - Ye Wang
- Heath Management Center, The Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Qiheng He
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Wen Wang
- China National Clinical Research Center for Neurological Diseases, Beijing, China.,Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Tan Zhang
- Department of Neurosurgery, The Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Zhongyong Wang
- Department of Neurosurgery, The Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Jun Dong
- Department of Neurosurgery, The Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Qing Lan
- Department of Neurosurgery, The Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Jizong Zhao
- Department of Neurosurgery, The Second Affiliated Hospital of Soochow University, Suzhou, China.,China National Clinical Research Center for Neurological Diseases, Beijing, China.,Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| |
Collapse
|
147
|
IDH Inhibitors and Beyond: The Cornerstone of Targeted Glioma Treatment. Mol Diagn Ther 2021; 25:457-473. [PMID: 34095989 DOI: 10.1007/s40291-021-00537-3] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/11/2021] [Indexed: 12/12/2022]
Abstract
Diffuse low-grade gliomas account for approximately 20% of all primary brain tumors, they arise from glial cells and show infiltrative growth without histological features of malignancy. Mutations of the IDH1 and IDH2 genes constitute a reliable molecular signature of low-grade gliomas and are the earliest driver mutations occurring during gliomagenesis, representing a relevant biomarker with diagnostic, prognostic, and predictive value. IDH mutations induce a neomorphic enzyme that converts α-ketoglutarate to the oncometabolite D-2-hydroxyglutarate, which leads to widespread effects on cellular epigenetics and metabolism. Currently, there are no approved molecularly targeted therapies and the standard treatment for low-grade gliomas consists of radiation therapy and chemotherapy, with rising concern about treatment-related toxicities. Targeting D-2-hydroxyglutarate is considered a novel attractive therapeutic approach for low-grade gliomas and the insights from clinical trials suggest that mutant-selective IDH inhibitors are the ideal candidates, with a favorable benefit/risk ratio. A pivotal question is whether blocking IDH neomorphic activity may activate alternative oncogenetic pathways, inducing acquired resistance to IDH inhibitors. Based on this rationale, combination therapies to enhance the antitumor activity of IDH inhibitors and approaches aimed at exploiting, rather than inhibiting, the metabolism of IDH-mutant cancer cells, such as poly (adenosine 5'-diphosphate-ribose) polymerase inhibitors, are emerging from preclinical research and clinical trials. In this review, we discuss the pivotal role of IDH mutations in gliomagenesis and the complex interactions between the genomic and epigenetic landscapes, providing an overview of how, in the last decade, therapeutic approaches for low-grade gliomas have evolved.
Collapse
|
148
|
Nie S, Zhu Y, Yang J, Xin T, Xue S, Zhang X, Sun J, Mu D, Gao Y, Chen Z, Ding X, Yu J, Hu M. Determining optimal clinical target volume margins in high-grade glioma based on microscopic tumor extension and magnetic resonance imaging. Radiat Oncol 2021; 16:97. [PMID: 34098965 PMCID: PMC8186169 DOI: 10.1186/s13014-021-01819-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2021] [Accepted: 05/10/2021] [Indexed: 11/22/2022] Open
Abstract
Introduction In this study, we performed a consecutive macropathologic analysis to assess microscopic extension (ME) in high-grade glioma (HGG) to determine appropriate clinical target volume (CTV) margins for radiotherapy. Materials and methods The study included HGG patients with tumors located in non-functional areas, and supratotal resection was performed. The ME distance from the edge of the tumor to the microscopic tumor cells surrounding brain tissue was measured. Associations between the extent of ME and clinicopathological characteristics were evaluated by multivariate linear regression (MVLR) analysis. An ME predictive model was developed based on the MVLR model. Results Between June 2017 and July 2019, 652 pathologic slides obtained from 30 HGG patients were analyzed. The mean ME distance was 1.70 cm (range, 0.63 to 2.87 cm). The MVLR analysis identified that pathologic grade, subventricular zone (SVZ) contact and O6-methylguanine-DNA methyltransferase (MGMT) methylation, isocitrate dehydrogenase (IDH) mutation and 1p/19q co-deletion status were independent variables predicting ME (all P < 0.05). A multivariable prediction model was developed as follows: YME = 0.672 + 0.513XGrade + 0.380XSVZ + 0.439XMGMT + 0.320XIDH + 0.333X1p/19q. The R-square value of goodness of fit was 0.780. The receiver operating characteristic curve proved that the area under the curve was 0.964 (P < 0.001). Conclusion ME was heterogeneously distributed across different grades of gliomas according to the tumor location and molecular marker status, which indicated that CTV delineation should be individualized. The model could predict the ME of HGG, which may help clinicians determine the CTV for individual patients. Trial registration The trial was registered with Chinese Clinical Trial Registry (ChiCTR2100046106). Registered 4 May 2021-Retrospectively registered. Supplementary Information The online version contains supplementary material available at 10.1186/s13014-021-01819-0.
Collapse
Affiliation(s)
- Shulun Nie
- Department of Radiation Oncology, Shandong First Medical University and Shandong Academy of Medical Sciences, Qingdao Road 6699, Jinan, 250117, Shandong, People's Republic of China.,Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jiyan Road 440, Jinan, 250117, Shandong, People's Republic of China
| | - Yufang Zhu
- Department of Neurosurgery, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong, People's Republic of China
| | - Jia Yang
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jiyan Road 440, Jinan, 250117, Shandong, People's Republic of China
| | - Tao Xin
- Department of Neurosurgery, The First Affiliated Hospital of Shandong First Medical University, Jinan, Shandong, People's Republic of China
| | - Song Xue
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jiyan Road 440, Jinan, 250117, Shandong, People's Republic of China
| | - Xianbin Zhang
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jiyan Road 440, Jinan, 250117, Shandong, People's Republic of China
| | - Jujie Sun
- Department of Pathology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong, People's Republic of China
| | - Dianbin Mu
- Department of Pathology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong, People's Republic of China
| | - Yongsheng Gao
- Department of Pathology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong, People's Republic of China
| | - Zhaoqiu Chen
- Department of Radiology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong, People's Republic of China
| | - Xingchen Ding
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jiyan Road 440, Jinan, 250117, Shandong, People's Republic of China
| | - Jinming Yu
- Department of Radiation Oncology, Shandong First Medical University and Shandong Academy of Medical Sciences, Qingdao Road 6699, Jinan, 250117, Shandong, People's Republic of China. .,Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jiyan Road 440, Jinan, 250117, Shandong, People's Republic of China.
| | - Man Hu
- Department of Radiation Oncology, Shandong First Medical University and Shandong Academy of Medical Sciences, Qingdao Road 6699, Jinan, 250117, Shandong, People's Republic of China. .,Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jiyan Road 440, Jinan, 250117, Shandong, People's Republic of China.
| |
Collapse
|
149
|
Berzero G, Di Stefano AL, Ronchi S, Bielle F, Villa C, Guillerm E, Capelle L, Mathon B, Laurenge A, Giry M, Schmitt Y, Marie Y, Idbaih A, Hoang-Xuan K, Delattre JY, Mokhtari K, Sanson M. IDH-wildtype lower-grade diffuse gliomas: the importance of histological grade and molecular assessment for prognostic stratification. Neuro Oncol 2021; 23:955-966. [PMID: 33173941 PMCID: PMC8168809 DOI: 10.1093/neuonc/noaa258] [Citation(s) in RCA: 89] [Impact Index Per Article: 22.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
BACKGROUND Isocitrate dehydrogenase (IDH) wildtype (wt) grade II gliomas are a rare and heterogeneous entity. Survival and prognostic factors are poorly defined. METHODS We searched retrospectively all patients diagnosed with diffuse World Health Organization (WHO) grades II and III gliomas at our center (1989-2020). RESULTS Out of 517 grade II gliomas, 47 were "diffuse astrocytomas, IDHwt." Tumors frequently had fronto-temporo-insular location (28/47, 60%) and infiltrative behavior. We found telomerase reverse transcriptase (TERT) promoter mutations (23/45, 51%), whole chromosome 7 gains (10/37, 27%), whole chromosome 10 losses (10/41, 24%), and EGFR amplifications (4/43, 9%), but no TP53 mutations (0/22, 0%). Median overall survival (OS) was 59 months (vs 19 mo for IDHwt grade III gliomas) (P < 0.0001). Twenty-nine patients (29/43, 67%) met the definition of molecular glioblastoma according to cIMPACT-NOW update 3. Median OS in this subset was 42 months, which was shorter compared with patients with IDHwt grade II gliomas not meeting this definition (median OS: 57 mo), but substantially longer compared with IDHwt grade III gliomas meeting the definition for molecular glioblastoma (median OS: 17 mo, P < 0.0001). Most patients with IDHwt grade II gliomas met cIMPACT criteria because of isolated TERT promoter mutations (16/26, 62%), which were not predictive of poor outcome (median OS: 88 mo). Actionable targets, including 5 gene fusions involving FGFR3, were found in 7 patients (24%). CONCLUSIONS Our findings highlight the importance of histological grading and molecular profiling for the prognostic stratification of IDHwt gliomas and suggest some caution when assimilating IDHwt grade II gliomas to molecular glioblastomas, especially those with isolated TERT promoter mutation.
Collapse
Affiliation(s)
- Giulia Berzero
- Sorbonne University, Brain and Spinal Cord Institute, Paris, France
- University Hospitals of Pitié Salpêtrière, Charles Foix, Department of Neurology 2 Mazarin, Paris, France
- Department of Brain and Behavioral Sciences, University of Pavia, Italy
| | - Anna Luisa Di Stefano
- Sorbonne University, Brain and Spinal Cord Institute, Paris, France
- University Hospitals of Pitié Salpêtrière, Charles Foix, Department of Neurology 2 Mazarin, Paris, France
- Department of Neurology, Foch Hospital, Suresnes, France
| | - Susanna Ronchi
- University Hospitals of Pitié Salpêtrière, Charles Foix, R Escourolle Laboratory, Paris, France
| | - Franck Bielle
- University Hospitals of Pitié Salpêtrière, Charles Foix, R Escourolle Laboratory, Paris, France
| | - Chiara Villa
- Department of Pathology, Foch Hospital, Suresnes, France
| | - Erell Guillerm
- University Hospitals of La Pitié Salpêtrière, Charles Foix, Functional Unit of Oncogenetics and Molecular Angiogenetics, Department of Genetics, Paris, France
| | - Laurent Capelle
- University Hospitals of La Pitié Salpêtrière, Charles Foix, Department of Neurology 2, Paris, France
| | - Bertrand Mathon
- University Hospitals of La Pitié Salpêtrière, Charles Foix, Department of Neurology 2, Paris, France
| | - Alice Laurenge
- Sorbonne University, Brain and Spinal Cord Institute, Paris, France
- University Hospitals of Pitié Salpêtrière, Charles Foix, Department of Neurology 2 Mazarin, Paris, France
| | - Marine Giry
- Sorbonne University, Brain and Spinal Cord Institute, Paris, France
| | - Yohann Schmitt
- Sorbonne University, Brain and Spinal Cord Institute, Paris, France
| | - Yannick Marie
- Sorbonne University, Brain and Spinal Cord Institute, Paris, France
- Onconeurotek Tumor Bank, University Hospitals of Pitié Salpêtrière, Charles Foix, Paris, France
| | - Ahmed Idbaih
- Sorbonne University, Brain and Spinal Cord Institute, Paris, France
- University Hospitals of Pitié Salpêtrière, Charles Foix, Department of Neurology 2 Mazarin, Paris, France
| | - Khe Hoang-Xuan
- Sorbonne University, Brain and Spinal Cord Institute, Paris, France
- University Hospitals of Pitié Salpêtrière, Charles Foix, Department of Neurology 2 Mazarin, Paris, France
| | - Jean-Yves Delattre
- Sorbonne University, Brain and Spinal Cord Institute, Paris, France
- University Hospitals of Pitié Salpêtrière, Charles Foix, Department of Neurology 2 Mazarin, Paris, France
- Onconeurotek Tumor Bank, University Hospitals of Pitié Salpêtrière, Charles Foix, Paris, France
| | - Karima Mokhtari
- Sorbonne University, Brain and Spinal Cord Institute, Paris, France
- University Hospitals of Pitié Salpêtrière, Charles Foix, R Escourolle Laboratory, Paris, France
- Onconeurotek Tumor Bank, University Hospitals of Pitié Salpêtrière, Charles Foix, Paris, France
| | - Marc Sanson
- Corresponding Author: Marc Sanson, MD PhD, Service de Neurologie 2, GH Pitié-Salpêtrière, 47 bd de l’Hôpital, 75013 Paris, France ()
| |
Collapse
|
150
|
Trifiletti DM, Brown PD. Cognitive outcomes in patients with low-grade glioma. Neuro Oncol 2021; 23:709-710. [PMID: 33704493 DOI: 10.1093/neuonc/noab033] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Affiliation(s)
| | - Paul D Brown
- Department of Radiation Oncology, Mayo Clinic, Rochester, Minnesota, USA
| |
Collapse
|