首页 > 最新文献

Glioma最新文献

英文 中文
A multifaceted review of temozolomide resistance mechanisms in glioblastoma beyond O-6-methylguanine-DNA methyltransferase 除O-6-甲基鸟嘌呤-DNA甲基转移酶外,替莫唑胺在胶质母细胞瘤中耐药性机制的多方面综述
Pub Date : 2019-03-01 DOI: 10.4103/glioma.glioma_3_19
P. Woo, Yi Li, Anna H. Chan, S. Ng, H. Loong, D. Chan, G. Wong, W. Poon
Temozolomide (TMZ), an oral alkylating agent, is a cornerstone of standard-of-care multimodality therapy for glioblastoma. In spite of significant efforts to treat this disease, the tumor carries a poor prognosis and is considered incurable largely due to the development of chemoresistance. One of the main mechanisms for this phenomenon is the activation of tumor DNA repair systems, such as O-6-methylguanine-DNA methyltransferase, that removes TMZ-induced DNA adducts and restores genomic integrity. Recent advances in the understanding of TMZ resistance oncobiology have introduced several novel independent molecular mechanisms involving epigenetic, transcriptomic, proteomic aberrations as well as alterations in apoptosis-autophagy processes, receptor tyrosine kinase activity, the tumor microenvironment, and the emergence of glioma stem cells. This article describes a multifaceted summary of the latest proposed causes for TMZ resistance and their complex interactions. It is believed that only by comprehending this growing network of interdependent mechanisms can effective combinatorial oncologic therapeutic strategies be developed to improve patient overall survival.
替莫唑胺(TMZ)是一种口服烷基化剂,是胶质母细胞瘤标准护理多模式治疗的基石。尽管在治疗这种疾病方面做出了重大努力,但该肿瘤的预后很差,并且被认为是不可治愈的,这主要是由于化疗耐药性的发展。这种现象的主要机制之一是肿瘤DNA修复系统的激活,如O-6-甲基鸟嘌呤-DNA甲基转移酶,其去除TMZ诱导的DNA加合物并恢复基因组完整性。TMZ抗性肿瘤生物学的最新进展介绍了几种新的独立分子机制,涉及表观遗传学、转录组学、蛋白质组学畸变以及细胞凋亡自噬过程、受体酪氨酸激酶活性、肿瘤微环境和神经胶质瘤干细胞的出现。本文对TMZ耐药性的最新原因及其复杂的相互作用进行了多方面的总结。人们相信,只有理解这种日益增长的相互依赖的机制网络,才能制定有效的组合肿瘤学治疗策略来提高患者的整体生存率。
{"title":"A multifaceted review of temozolomide resistance mechanisms in glioblastoma beyond O-6-methylguanine-DNA methyltransferase","authors":"P. Woo, Yi Li, Anna H. Chan, S. Ng, H. Loong, D. Chan, G. Wong, W. Poon","doi":"10.4103/glioma.glioma_3_19","DOIUrl":"https://doi.org/10.4103/glioma.glioma_3_19","url":null,"abstract":"Temozolomide (TMZ), an oral alkylating agent, is a cornerstone of standard-of-care multimodality therapy for glioblastoma. In spite of significant efforts to treat this disease, the tumor carries a poor prognosis and is considered incurable largely due to the development of chemoresistance. One of the main mechanisms for this phenomenon is the activation of tumor DNA repair systems, such as O-6-methylguanine-DNA methyltransferase, that removes TMZ-induced DNA adducts and restores genomic integrity. Recent advances in the understanding of TMZ resistance oncobiology have introduced several novel independent molecular mechanisms involving epigenetic, transcriptomic, proteomic aberrations as well as alterations in apoptosis-autophagy processes, receptor tyrosine kinase activity, the tumor microenvironment, and the emergence of glioma stem cells. This article describes a multifaceted summary of the latest proposed causes for TMZ resistance and their complex interactions. It is believed that only by comprehending this growing network of interdependent mechanisms can effective combinatorial oncologic therapeutic strategies be developed to improve patient overall survival.","PeriodicalId":12731,"journal":{"name":"Glioma","volume":"2 1","pages":"68 - 82"},"PeriodicalIF":0.0,"publicationDate":"2019-03-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"41520237","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 19
Glioma special issue introduction 胶质瘤特刊简介
Pub Date : 2019-01-01 DOI: 10.4103/glioma.glioma_2_19
Christopher M. Jackson, M. Lim
In the past decade, cancer immunotherapy has emerged from experimental protocols in select pathologies to a mainstay of frontline cancer therapy that has drastically improved outcomes for thousands of patients. Since the approval of the first anticancer vaccine (sipuleucel-T) in 2010, strategies for stimulating the immune system to fight cancer have evolved from monotherapy vaccination approaches adapted from infectious diseases to immune checkpoint inhibition, adoptive cell therapy, viral therapy, and many others. These clinical advances reflect pivotal discoveries in how the immune system interacts with the tumor microenvironment, leading to the identification of targetable pathways and rationally designed algorithms that account for pathology, patient, and iatrogenic factors. Despite the challenges of treating gliomas with immunotherapy, intensive efforts aimed at elucidating the underpinnings of glioma immunology and next-generation immunotherapeutic strategies have placed successful translation of the first glioma immunotherapies seemingly within reach.
在过去的十年中,癌症免疫疗法已经从选择病理的实验方案发展成为一线癌症治疗的支柱,极大地改善了成千上万患者的预后。自2010年首个抗癌疫苗(sipuleucel-T)获批以来,刺激免疫系统对抗癌症的策略已经从适用于传染病的单疗法疫苗接种方法发展到免疫检查点抑制、过继细胞治疗、病毒治疗和许多其他方法。这些临床进展反映了免疫系统如何与肿瘤微环境相互作用的关键发现,从而确定了可靶向的途径,并合理设计了考虑病理、患者和医源性因素的算法。尽管用免疫疗法治疗胶质瘤面临挑战,但旨在阐明胶质瘤免疫学基础和下一代免疫治疗策略的密集努力已经使第一批胶质瘤免疫疗法的成功翻译似乎指日可及。
{"title":"Glioma special issue introduction","authors":"Christopher M. Jackson, M. Lim","doi":"10.4103/glioma.glioma_2_19","DOIUrl":"https://doi.org/10.4103/glioma.glioma_2_19","url":null,"abstract":"In the past decade, cancer immunotherapy has emerged from experimental protocols in select pathologies to a mainstay of frontline cancer therapy that has drastically improved outcomes for thousands of patients. Since the approval of the first anticancer vaccine (sipuleucel-T) in 2010, strategies for stimulating the immune system to fight cancer have evolved from monotherapy vaccination approaches adapted from infectious diseases to immune checkpoint inhibition, adoptive cell therapy, viral therapy, and many others. These clinical advances reflect pivotal discoveries in how the immune system interacts with the tumor microenvironment, leading to the identification of targetable pathways and rationally designed algorithms that account for pathology, patient, and iatrogenic factors. Despite the challenges of treating gliomas with immunotherapy, intensive efforts aimed at elucidating the underpinnings of glioma immunology and next-generation immunotherapeutic strategies have placed successful translation of the first glioma immunotherapies seemingly within reach.","PeriodicalId":12731,"journal":{"name":"Glioma","volume":"2 1","pages":"1 - 2"},"PeriodicalIF":0.0,"publicationDate":"2019-01-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"44939305","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 1
Gamma-delta T cells in glioblastoma immunotherapy γ - δ T细胞在胶质母细胞瘤免疫治疗中的应用
Pub Date : 2019-01-01 DOI: 10.4103/glioma.glioma_48_18
Sadhak Sengupta
Conventional immunotherapy in the treatment of glioblastoma (GBM) has essentially produced no significant advantage over the use of chemotherapeutic drugs. A strongly immunosuppressive tumor microenvironment and lack of antigen-presenting major histocompatibility expression on tumor cells have made GBM a poor immunological target. Molecular heterogeneity of GBMs, both within the tumor and across patients, results in the immunological escape of tumors that do not express target antigens. Therefore, the development of nonconventional immunotherapy for GBM is continuously being sought. γδ T cells are a minor subset of the human T-cell repertoire with unique antitumor properties that have been shown to be functionally superior to conventional αβ T-cell receptor expressing T cell-based immunotherapy for cancer, including GBM. Unlike, the more abundant αβ T cells, γδ T cells do not require major histocompatibility proteins for activation. In addition to the γδ T-cell receptor, these cells express a plethora of other antigenic receptors that recognize external stimuli, as well as several self-peptides, which make these cells a strong candidate for the development of cancer immunotherapeutics. A higher threshold of activation-induced cell death and resistance to inducing graft-versus-host disease are also characteristics of these T cells. In this review, we discuss the biology and immunological characteristics of γδ T cells and review current research using γδ T cells in GBM immunotherapy to explore whether these cells can be the potential next-gen immunotherapeutic candidate for this dreadful disease.
在治疗胶质母细胞瘤(GBM)中,传统的免疫疗法基本上没有产生比使用化疗药物显著的优势。强烈的免疫抑制肿瘤微环境和肿瘤细胞上缺乏抗原呈递的主要组织相容性表达使GBM成为一个较差的免疫靶点。肿瘤内和患者间GBMs的分子异质性导致不表达靶抗原的肿瘤的免疫逃逸。因此,对GBM的非常规免疫治疗的发展一直是人们不断寻求的。γδ T细胞是人类T细胞库中的一个小子集,具有独特的抗肿瘤特性,已被证明在功能上优于传统的αβ T细胞受体表达的基于T细胞的癌症免疫疗法,包括GBM。与更丰富的αβ T细胞不同,γδ T细胞不需要主要的组织相容性蛋白来激活。除了γδ t细胞受体外,这些细胞还表达大量识别外部刺激的其他抗原受体,以及几种自肽,这使得这些细胞成为癌症免疫治疗发展的有力候选者。激活诱导的细胞死亡和抗移植物抗宿主病的更高阈值也是这些T细胞的特征。本文综述了γδ T细胞的生物学和免疫学特性,并对γδ T细胞在GBM免疫治疗中的研究现状进行了综述,以探讨γδ T细胞是否可以作为GBM免疫治疗的潜在候选细胞。
{"title":"Gamma-delta T cells in glioblastoma immunotherapy","authors":"Sadhak Sengupta","doi":"10.4103/glioma.glioma_48_18","DOIUrl":"https://doi.org/10.4103/glioma.glioma_48_18","url":null,"abstract":"Conventional immunotherapy in the treatment of glioblastoma (GBM) has essentially produced no significant advantage over the use of chemotherapeutic drugs. A strongly immunosuppressive tumor microenvironment and lack of antigen-presenting major histocompatibility expression on tumor cells have made GBM a poor immunological target. Molecular heterogeneity of GBMs, both within the tumor and across patients, results in the immunological escape of tumors that do not express target antigens. Therefore, the development of nonconventional immunotherapy for GBM is continuously being sought. γδ T cells are a minor subset of the human T-cell repertoire with unique antitumor properties that have been shown to be functionally superior to conventional αβ T-cell receptor expressing T cell-based immunotherapy for cancer, including GBM. Unlike, the more abundant αβ T cells, γδ T cells do not require major histocompatibility proteins for activation. In addition to the γδ T-cell receptor, these cells express a plethora of other antigenic receptors that recognize external stimuli, as well as several self-peptides, which make these cells a strong candidate for the development of cancer immunotherapeutics. A higher threshold of activation-induced cell death and resistance to inducing graft-versus-host disease are also characteristics of these T cells. In this review, we discuss the biology and immunological characteristics of γδ T cells and review current research using γδ T cells in GBM immunotherapy to explore whether these cells can be the potential next-gen immunotherapeutic candidate for this dreadful disease.","PeriodicalId":12731,"journal":{"name":"Glioma","volume":"2 1","pages":"30 - 36"},"PeriodicalIF":0.0,"publicationDate":"2019-01-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"44192060","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 2
Combination of oncolytic viruses and immune checkpoint inhibitors in glioblastoma 溶瘤病毒和免疫检查点抑制剂在胶质母细胞瘤中的联合应用
Pub Date : 2019-01-01 DOI: 10.4103/glioma.glioma_5_19
Kunal Desai, Anne Hubben, M. Ahluwalia
Glioblastoma is associated with poor prognosis with a mean survival of 15 months after diagnosis. The current standard of care includes surgery, radiation, and temozolomide with the use of tumor-treating fields in select patient population. The past decade has witnessed a convergence in our understanding of tumor biology and the role of the immune system in fighting cancer. The highly immunosuppressive tumor microenvironment exerted by glioblastoma cells has contributed to the lack of success of novel immunotherapies till date (including checkpoint inhibitors). Oncolytic viral-based approaches are of renewed interest given advances in tumor cell tropism, pathogenicity, and immunogenicity. More importantly, oncolytic viruses have been shown to initiate a broad immune response through various mechanisms including dual activation of the innate and adaptive arms of the host immune system. Because the initial clinical studies with monotherapy checkpoint inhibition in glioblastoma have failed to demonstrate a survival advantage, most trials in glioblastoma are testing combinations that seek to augment the immune response through mutually reinforcing approaches that can overcome the immunosuppressive milieu. Preclinical data in glioblastoma models with combined oncolytic viruses therapy and checkpoint blockade are favorable and provide rationale to initiate first-in-human trials. Even though the number of clinical trials testing this combination in glioblastoma is limited, more studies are expected in the future.
胶质母细胞瘤与不良预后相关,诊断后平均生存期为15个月。目前的治疗标准包括手术、放疗和替莫唑胺,并在选定的患者群体中使用肿瘤治疗领域。过去的十年见证了我们对肿瘤生物学和免疫系统在抗癌中的作用的理解的融合。胶质母细胞瘤细胞施加的高度免疫抑制肿瘤微环境导致迄今为止缺乏成功的新型免疫疗法(包括检查点抑制剂)。鉴于肿瘤细胞嗜性、致病性和免疫原性方面的进展,基于溶瘤病毒的方法重新引起了人们的兴趣。更重要的是,溶瘤病毒已被证明可以通过多种机制启动广泛的免疫反应,包括双重激活宿主免疫系统的先天和适应性臂。由于在胶质母细胞瘤中单药检查点抑制的初步临床研究未能证明其具有生存优势,因此大多数胶质母细胞瘤试验都在测试通过相互加强的方法来增强免疫反应的组合,从而克服免疫抑制环境。溶瘤病毒联合治疗和检查点阻断在胶质母细胞瘤模型中的临床前数据是有利的,并为开展首次人体试验提供了依据。尽管在胶质母细胞瘤中测试这种组合的临床试验数量有限,但预计未来会有更多的研究。
{"title":"Combination of oncolytic viruses and immune checkpoint inhibitors in glioblastoma","authors":"Kunal Desai, Anne Hubben, M. Ahluwalia","doi":"10.4103/glioma.glioma_5_19","DOIUrl":"https://doi.org/10.4103/glioma.glioma_5_19","url":null,"abstract":"Glioblastoma is associated with poor prognosis with a mean survival of 15 months after diagnosis. The current standard of care includes surgery, radiation, and temozolomide with the use of tumor-treating fields in select patient population. The past decade has witnessed a convergence in our understanding of tumor biology and the role of the immune system in fighting cancer. The highly immunosuppressive tumor microenvironment exerted by glioblastoma cells has contributed to the lack of success of novel immunotherapies till date (including checkpoint inhibitors). Oncolytic viral-based approaches are of renewed interest given advances in tumor cell tropism, pathogenicity, and immunogenicity. More importantly, oncolytic viruses have been shown to initiate a broad immune response through various mechanisms including dual activation of the innate and adaptive arms of the host immune system. Because the initial clinical studies with monotherapy checkpoint inhibition in glioblastoma have failed to demonstrate a survival advantage, most trials in glioblastoma are testing combinations that seek to augment the immune response through mutually reinforcing approaches that can overcome the immunosuppressive milieu. Preclinical data in glioblastoma models with combined oncolytic viruses therapy and checkpoint blockade are favorable and provide rationale to initiate first-in-human trials. Even though the number of clinical trials testing this combination in glioblastoma is limited, more studies are expected in the future.","PeriodicalId":12731,"journal":{"name":"Glioma","volume":"2 1","pages":"7 - 19"},"PeriodicalIF":0.0,"publicationDate":"2019-01-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"42309229","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 2
Importance of iatrogenic immunosuppression in the treatment of patients with high-grade glioma with immunotherapy 医源性免疫抑制在高级别胶质瘤患者免疫治疗中的重要性
Pub Date : 2019-01-01 DOI: 10.4103/glioma.glioma_8_19
Anna F Piotrowski, S. Grossman
Treatment-related lymphopenia is a poor prognostic factor for overall survival in patients with high-grade glioma and predicts suboptimal response to immune therapies. Immunotherapy is conceptually an appealing approach in adults with high-grade glioma given that effector lymphocytes are capable of penetrating the blood–brain barrier. However, 40% of these patients develop severe lymphopenia (CD4 counts <200) following concurrent radiation and temozolomide. These low lymphocyte counts are associated with inferior survival. Research suggests that this iatrogenic immunosuppression is attributed to the inadvertent radiation of circulating lymphocytes as they traverse the irradiated field. Lymphocyte subtypes are universally affected by this radiation toxicity. These findings have been reproduced in animal studies, and clinical correlations have been demonstrated in patients with various malignancies. This lymphopenia has been linked with failure to respond to immunologic interventions. Recent insights into the etiology of this radiation-induced lymphopenia have triggered a variety of novel approaches to prevent or restore immunologic function in this patient population. These include altering radiation plans, reducing the number of lymphocytes passing through the radiation field, harvesting lymphocytes before and reinfusing them after radiation, and using growth factors to restore lymphocyte counts. This manuscript reviews critical relationships between treatment-related lymphopenia and immunotherapy outcomes in patients with high-grade gliomas and novel approaches to these issues.
治疗相关性淋巴细胞减少症是影响高级别胶质瘤患者总生存率的一个不良预后因素,并预示着免疫治疗的次优反应。考虑到效应淋巴细胞能够穿透血脑屏障,免疫疗法在概念上是治疗成人高级别胶质瘤的一种吸引人的方法。然而,这些患者中有40%在同时放疗和替莫唑胺后出现严重淋巴细胞减少(CD4计数<200)。淋巴细胞计数低与生存率低有关。研究表明,这种医源性免疫抑制是由于循环淋巴细胞在穿过辐照场时的无意辐射。淋巴细胞亚型普遍受到这种辐射毒性的影响。这些发现在动物研究中得到了重复,并且在各种恶性肿瘤患者中证实了临床相关性。这种淋巴细胞减少与免疫干预无效有关。最近对这种放射诱导淋巴细胞减少的病因学的深入研究引发了各种新的方法来预防或恢复该患者群体的免疫功能。这些方法包括改变放射计划,减少通过放射场的淋巴细胞数量,在放射前收集淋巴细胞并在放射后重新输注,以及使用生长因子来恢复淋巴细胞计数。这篇文章回顾了治疗相关淋巴细胞减少症和高级别胶质瘤患者免疫治疗结果之间的关键关系,以及解决这些问题的新方法。
{"title":"Importance of iatrogenic immunosuppression in the treatment of patients with high-grade glioma with immunotherapy","authors":"Anna F Piotrowski, S. Grossman","doi":"10.4103/glioma.glioma_8_19","DOIUrl":"https://doi.org/10.4103/glioma.glioma_8_19","url":null,"abstract":"Treatment-related lymphopenia is a poor prognostic factor for overall survival in patients with high-grade glioma and predicts suboptimal response to immune therapies. Immunotherapy is conceptually an appealing approach in adults with high-grade glioma given that effector lymphocytes are capable of penetrating the blood–brain barrier. However, 40% of these patients develop severe lymphopenia (CD4 counts <200) following concurrent radiation and temozolomide. These low lymphocyte counts are associated with inferior survival. Research suggests that this iatrogenic immunosuppression is attributed to the inadvertent radiation of circulating lymphocytes as they traverse the irradiated field. Lymphocyte subtypes are universally affected by this radiation toxicity. These findings have been reproduced in animal studies, and clinical correlations have been demonstrated in patients with various malignancies. This lymphopenia has been linked with failure to respond to immunologic interventions. Recent insights into the etiology of this radiation-induced lymphopenia have triggered a variety of novel approaches to prevent or restore immunologic function in this patient population. These include altering radiation plans, reducing the number of lymphocytes passing through the radiation field, harvesting lymphocytes before and reinfusing them after radiation, and using growth factors to restore lymphocyte counts. This manuscript reviews critical relationships between treatment-related lymphopenia and immunotherapy outcomes in patients with high-grade gliomas and novel approaches to these issues.","PeriodicalId":12731,"journal":{"name":"Glioma","volume":"2 1","pages":"3 - 6"},"PeriodicalIF":0.0,"publicationDate":"2019-01-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"43422866","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Evidence of calcium-activated potassium channel subunit alpha-1 as a key promoter of glioma growth and tumorigenicity 钙激活的钾通道亚单位α-1是神经胶质瘤生长和致瘤性的关键启动子的证据
Pub Date : 2019-01-01 DOI: 10.4103/glioma.glioma_44_18
D. Khaitan, N. Ningaraj
Background and Aim: Mechanisms of glioma progression are poorly understood. Upregulation of calcium-activated potassium channel subunit alpha-1 (KCNMA1), which encodes the α-subunit of maxi-calcium-activated potassium (BKCa) channels, is shown to be a novel mechanism for the malignant phenotype of brain tumor cells. The aim of this study was to establish the functional role of KCNMA1 in glioma biology. Materials and Methods: U-87-MG (U-87) cells were transfected to increase BKCa channel expression and activity. Glioma cell proliferation, invasiveness, and transendothelial migration were then measured. BKCa channels were blocked with iberiotoxin or short hairpin RNA (shRNA), which significantly inhibited K+ currents and growth of U-87 cells. It was tested whether KCNMA1 overexpression enhanced tumorogenecity in glioma xenograft mouse models by injecting wild-type and KCNMA1- overexpressing U87-MG cells. In parallel experiment, it was studied whether shRNA KCNMA1-expressing U-87 cells show attenuated glioma growth in mice. The study protocol was approved by the Institutional Animal Care and Use Committee, Mercer University (A0706007_01), Atlanta, GA, USA on July 20, 2007. Results: The effect of KCNMA1 overexpression in glioma growth as well as on associated cell biology functions such as proliferation, invasion, and migration was presented in this study. Messenger RNA and protein analyses revealed that KCNMA1 was amplified in 90% of high-grade gliomas and in high-grade glioma cell line U-87. In contrast, KCNMA1 amplification was not found in normal brain tissues. These data indicate that KCNMA1 plays critical role in glioma biology by interacting with several cellular processes. The data demonstrate that KCNMA1 amplification drives glioma cell proliferation and growth, which can be attenuated by its downregulation. Conclusion: KCNMA1 is a regulator of glioma cell proliferation and growth and thus qualifies as a promising diagnostic and therapeutic target in the treatment of glioma.
背景与目的:胶质瘤进展的机制尚不清楚。编码最大钙活化钾(BKCa)通道α-亚基的钙活化钾通道亚基α- 1 (KCNMA1)的上调被证明是脑肿瘤细胞恶性表型的新机制。本研究的目的是确定KCNMA1在胶质瘤生物学中的功能作用。材料和方法:转染U-87- mg (U-87)细胞,提高BKCa通道的表达和活性。然后测量胶质瘤细胞增殖、侵袭性和跨内皮迁移。用iberiotoxin或short hairpin RNA (shRNA)阻断BKCa通道,可显著抑制U-87细胞的K+电流和生长。通过注射野生型和过表达KCNMA1的U87-MG细胞,检测KCNMA1过表达是否增强了胶质瘤异种移植小鼠模型的肿瘤发生。在平行实验中,我们研究了表达shRNA kcnma1的U-87细胞在小鼠中是否表现出胶质瘤生长减弱的现象。本研究方案于2007年7月20日获得美国亚特兰大美世大学机构动物护理和使用委员会(A0706007_01)批准。结果:本研究揭示了KCNMA1过表达对胶质瘤生长及相关细胞生物学功能如增殖、侵袭和迁移的影响。信使RNA和蛋白分析显示KCNMA1在90%的高级别胶质瘤和高级别胶质瘤细胞系U-87中扩增。相反,在正常脑组织中未发现KCNMA1扩增。这些数据表明KCNMA1通过与几个细胞过程相互作用在胶质瘤生物学中起关键作用。数据表明,KCNMA1扩增可驱动胶质瘤细胞的增殖和生长,其下调可减弱其增殖和生长。结论:KCNMA1是胶质瘤细胞增殖和生长的调节因子,是治疗胶质瘤的一个有前景的诊断和治疗靶点。
{"title":"Evidence of calcium-activated potassium channel subunit alpha-1 as a key promoter of glioma growth and tumorigenicity","authors":"D. Khaitan, N. Ningaraj","doi":"10.4103/glioma.glioma_44_18","DOIUrl":"https://doi.org/10.4103/glioma.glioma_44_18","url":null,"abstract":"Background and Aim: Mechanisms of glioma progression are poorly understood. Upregulation of calcium-activated potassium channel subunit alpha-1 (KCNMA1), which encodes the α-subunit of maxi-calcium-activated potassium (BKCa) channels, is shown to be a novel mechanism for the malignant phenotype of brain tumor cells. The aim of this study was to establish the functional role of KCNMA1 in glioma biology. Materials and Methods: U-87-MG (U-87) cells were transfected to increase BKCa channel expression and activity. Glioma cell proliferation, invasiveness, and transendothelial migration were then measured. BKCa channels were blocked with iberiotoxin or short hairpin RNA (shRNA), which significantly inhibited K+ currents and growth of U-87 cells. It was tested whether KCNMA1 overexpression enhanced tumorogenecity in glioma xenograft mouse models by injecting wild-type and KCNMA1- overexpressing U87-MG cells. In parallel experiment, it was studied whether shRNA KCNMA1-expressing U-87 cells show attenuated glioma growth in mice. The study protocol was approved by the Institutional Animal Care and Use Committee, Mercer University (A0706007_01), Atlanta, GA, USA on July 20, 2007. Results: The effect of KCNMA1 overexpression in glioma growth as well as on associated cell biology functions such as proliferation, invasion, and migration was presented in this study. Messenger RNA and protein analyses revealed that KCNMA1 was amplified in 90% of high-grade gliomas and in high-grade glioma cell line U-87. In contrast, KCNMA1 amplification was not found in normal brain tissues. These data indicate that KCNMA1 plays critical role in glioma biology by interacting with several cellular processes. The data demonstrate that KCNMA1 amplification drives glioma cell proliferation and growth, which can be attenuated by its downregulation. Conclusion: KCNMA1 is a regulator of glioma cell proliferation and growth and thus qualifies as a promising diagnostic and therapeutic target in the treatment of glioma.","PeriodicalId":12731,"journal":{"name":"Glioma","volume":"2 1","pages":"46 - 54"},"PeriodicalIF":0.0,"publicationDate":"2019-01-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"42981683","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 4
A rare case of gliosarcoma after acute hemorrhage 急性出血后发生胶质瘤1例
Pub Date : 2019-01-01 DOI: 10.4103/glioma.glioma_7_19
Y. Diansari, S. Mutiara, Hasan Baraqbah, M. Syahrir, Hanna Marsinta, A. Erwin, Dini Andriani
Gliosarcoma (GSM) is a rare primary neoplasm of the brain, characterized by a biphasic tissue pattern with alternating areas displaying glial and mesenchymal differentiation. GSM is usually treated as glioblastoma multiforme (GBM) and the prognosis is poor. Here, we discuss a 50-year-old male presenting with sudden decreased consciousness and a history of recent headache. A noncontrasted computed tomography of the brain revealed a well-circumscribed, hyperdense lesion in the left frontal lobe with significant perilesional edema, suggestive of an intracranial hemorrhage. The patient underwent emergency craniotomy to evacuate the hematoma. Intraoperatively, a mass was present, and fluid around the mass was collected and analyzed. Cytology revealed the presence of malignant cells. A 1-week postmagnetic resonance imaging (MRI) revealed a heterogenously enhancing mass in the left frontal lobe, suggestive of a high-grade glioma. The patient was then treated with radiation followed by adjuvant treatment with temozolomide plus bevacizumab. One year after the surgery, the patient was readmitted with headache, right-sided hemiparesis, and seizure. A follow-up MRI found a residual mass in the same region. A second surgery was performed. Histopathology examination showed GSM, and this was confirmed with an immunohistochemistry panel including glial fibrillary acid protein and vimentin. GSM is a rare variant of GBM. Intracranial hemorrhage is an uncommon clinical presentation of GSM. Diagnosis is established by histopathology. Treatment is surgery, followed by radiotherapy and chemotherapy. The study was approved by the Health Research Review Committee of Mohammad Hoesin Central General Hospital and Faculty of Medicine Sriwijaya University (No. 089/kepkrsmhunsri/2019) on March 1, 2019.
神经胶质瘤(GSM)是一种罕见的原发性脑肿瘤,其特征是双相组织模式,交替显示胶质细胞和间质分化。GSM通常作为多形性胶质母细胞瘤(GBM)治疗,预后较差。在这里,我们讨论一个50岁的男性表现为突然意识下降和最近头痛的历史。脑部非对比计算机断层扫描显示左额叶一边界清晰的高密度病变,病灶周围明显水肿,提示颅内出血。病人接受紧急开颅以排出血肿。术中发现肿块,收集并分析肿块周围的液体。细胞学检查显示存在恶性细胞。1周后磁共振成像(MRI)显示左侧额叶有一个非均匀强化肿块,提示高级别胶质瘤。患者随后接受放射治疗,随后使用替莫唑胺加贝伐单抗进行辅助治疗。手术一年后,患者因头痛、右侧偏瘫和癫痫再次入院。随后的核磁共振检查在同一区域发现了一个残余肿块。进行了第二次手术。组织病理学检查显示GSM,免疫组织化学检查包括神经胶质纤维酸蛋白和波形蛋白证实了这一点。GSM是GBM的一种罕见变种。颅内出血是一种罕见的临床表现。诊断由组织病理学确定。治疗方法是手术,然后是放疗和化疗。该研究于2019年3月1日获得穆罕默德·胡辛中央总医院和斯里维加亚大学医学院卫生研究审查委员会(No. 089/kepkrsmhunsri/2019)的批准。
{"title":"A rare case of gliosarcoma after acute hemorrhage","authors":"Y. Diansari, S. Mutiara, Hasan Baraqbah, M. Syahrir, Hanna Marsinta, A. Erwin, Dini Andriani","doi":"10.4103/glioma.glioma_7_19","DOIUrl":"https://doi.org/10.4103/glioma.glioma_7_19","url":null,"abstract":"Gliosarcoma (GSM) is a rare primary neoplasm of the brain, characterized by a biphasic tissue pattern with alternating areas displaying glial and mesenchymal differentiation. GSM is usually treated as glioblastoma multiforme (GBM) and the prognosis is poor. Here, we discuss a 50-year-old male presenting with sudden decreased consciousness and a history of recent headache. A noncontrasted computed tomography of the brain revealed a well-circumscribed, hyperdense lesion in the left frontal lobe with significant perilesional edema, suggestive of an intracranial hemorrhage. The patient underwent emergency craniotomy to evacuate the hematoma. Intraoperatively, a mass was present, and fluid around the mass was collected and analyzed. Cytology revealed the presence of malignant cells. A 1-week postmagnetic resonance imaging (MRI) revealed a heterogenously enhancing mass in the left frontal lobe, suggestive of a high-grade glioma. The patient was then treated with radiation followed by adjuvant treatment with temozolomide plus bevacizumab. One year after the surgery, the patient was readmitted with headache, right-sided hemiparesis, and seizure. A follow-up MRI found a residual mass in the same region. A second surgery was performed. Histopathology examination showed GSM, and this was confirmed with an immunohistochemistry panel including glial fibrillary acid protein and vimentin. GSM is a rare variant of GBM. Intracranial hemorrhage is an uncommon clinical presentation of GSM. Diagnosis is established by histopathology. Treatment is surgery, followed by radiotherapy and chemotherapy. The study was approved by the Health Research Review Committee of Mohammad Hoesin Central General Hospital and Faculty of Medicine Sriwijaya University (No. 089/kepkrsmhunsri/2019) on March 1, 2019.","PeriodicalId":12731,"journal":{"name":"Glioma","volume":"2 1","pages":"122 - 125"},"PeriodicalIF":0.0,"publicationDate":"2019-01-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"70735559","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 1
Immune checkpoint modulation: Tenets and implications in glioblastoma 免疫检查点调节:胶质母细胞瘤的原理和意义
Pub Date : 2019-01-01 DOI: 10.4103/glioma.glioma_47_18
John P. Lynes, Victoria E. Sanchez, Anthony K. Nwankwo, Gifty A. Dominah, E. Nduom
Glioblastoma (GBM) is the most common primary central nervous system tumor, and despite advances made in traditional chemotherapy and radiation, it continues to carry a poor prognosis. The discovery of the profound immunosuppressive microenvironment created by GBM has given insight on the aggressiveness of this recalcitrant disease. This has led many to believe that immune therapy may yield the improvement in survival that the neuro-oncology community is seeking. In other cancers, the targeting of immune checkpoints has been the most promising immunotherapeutic strategy to date. Immune checkpoints modulate the function of the immune system by increasing or decreasing immune activity. Checkpoint inhibitors and more recently agonists target molecules that regulate immune response to increase immune function either directly or by removal of inhibitory signals. These molecules modulate immunity in the physiologic state to maintain homeostasis, but they are co-opted by cancer to avoid immune detection and attack. The use of checkpoint inhibition to improve cancer therapy has revolutionized the field of oncology, leading to unprecedented improvements in survival from many systemic malignancies. Utilizing PubMed and ClinicalTrials.gov to compile published findings and ongoing trials, we review immune checkpoints and their modulators from bench to bedside over several decades. In this review, the discovery of different checkpoint molecules and the development of drugs used to target them are addressed. In addition, the current state of checkpoint inhibition in GBM, presenting completed and ongoing preclinical and clinical studies utilizing these therapies, is discussed. Finally, we conclude by reviewing the current limitations and potential future directions for the use of checkpoint blockade in the treatment of GBM.
胶质母细胞瘤(GBM)是最常见的原发性中枢神经系统肿瘤,尽管传统的化疗和放疗取得了进展,但其预后仍然很差。GBM产生的深刻的免疫抑制微环境的发现,让我们深入了解了这种顽固性疾病的侵袭性。这让许多人相信,免疫治疗可能会提高神经肿瘤学界正在寻求的生存率。在其他癌症中,靶向免疫检查点是迄今为止最有前途的免疫治疗策略。免疫检查点通过增加或降低免疫活性来调节免疫系统的功能。检查点抑制剂和最近的激动剂靶向调节免疫反应的分子,以直接或通过去除抑制信号来增加免疫功能。这些分子在生理状态下调节免疫以维持体内平衡,但它们与癌症共存以避免免疫检测和攻击。使用检查点抑制来改善癌症治疗已经彻底改变了肿瘤学领域,导致许多系统性恶性肿瘤的生存率得到前所未有的提高。利用PubMed和ClinicalTrials.gov汇编已发表的研究结果和正在进行的试验,我们回顾了几十年来从试验台到床边的免疫检查点及其调节剂。在这篇综述中,介绍了不同检查点分子的发现和用于靶向它们的药物的开发。此外,还讨论了GBM检查点抑制的现状,介绍了利用这些疗法进行的已完成和正在进行的临床前和临床研究。最后,我们通过回顾检查点阻断在GBM治疗中的当前局限性和潜在的未来方向来得出结论。
{"title":"Immune checkpoint modulation: Tenets and implications in glioblastoma","authors":"John P. Lynes, Victoria E. Sanchez, Anthony K. Nwankwo, Gifty A. Dominah, E. Nduom","doi":"10.4103/glioma.glioma_47_18","DOIUrl":"https://doi.org/10.4103/glioma.glioma_47_18","url":null,"abstract":"Glioblastoma (GBM) is the most common primary central nervous system tumor, and despite advances made in traditional chemotherapy and radiation, it continues to carry a poor prognosis. The discovery of the profound immunosuppressive microenvironment created by GBM has given insight on the aggressiveness of this recalcitrant disease. This has led many to believe that immune therapy may yield the improvement in survival that the neuro-oncology community is seeking. In other cancers, the targeting of immune checkpoints has been the most promising immunotherapeutic strategy to date. Immune checkpoints modulate the function of the immune system by increasing or decreasing immune activity. Checkpoint inhibitors and more recently agonists target molecules that regulate immune response to increase immune function either directly or by removal of inhibitory signals. These molecules modulate immunity in the physiologic state to maintain homeostasis, but they are co-opted by cancer to avoid immune detection and attack. The use of checkpoint inhibition to improve cancer therapy has revolutionized the field of oncology, leading to unprecedented improvements in survival from many systemic malignancies. Utilizing PubMed and ClinicalTrials.gov to compile published findings and ongoing trials, we review immune checkpoints and their modulators from bench to bedside over several decades. In this review, the discovery of different checkpoint molecules and the development of drugs used to target them are addressed. In addition, the current state of checkpoint inhibition in GBM, presenting completed and ongoing preclinical and clinical studies utilizing these therapies, is discussed. Finally, we conclude by reviewing the current limitations and potential future directions for the use of checkpoint blockade in the treatment of GBM.","PeriodicalId":12731,"journal":{"name":"Glioma","volume":"2 1","pages":"20 - 29"},"PeriodicalIF":0.0,"publicationDate":"2019-01-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"48684300","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
IDH1 mutation decreases the invasiveness of glioma by downregulating the expression and activity of TAZ IDH1突变通过下调TAZ的表达和活性来降低胶质瘤的侵袭性
Pub Date : 2019-01-01 DOI: 10.4103/glioma.glioma_46_18
Ningning Li, Rui Zhang, Yi Sun, Chenyue Xu, Yin Wang, J. Xiong, Qi Chen, Y. Liu
Background and Aim: Gliomas carrying mutated isocitrate dehydrogenase 1 (IDH1) have an improved prognosis, but how this mutation improves survival is not known. In this study, we evaluated the correlation of expression of the gene transcriptional coactivator with PDZ-binding motif (TAZ) with IDH1 mutation in astrocytomas of different grades. Materials and Methods: We analyzed the expression of TAZ by immunohistochemistry in a cohort of 90 formalin-fixed paraffin-embedded human astrocytoma samples. A human glioblastoma cell line (U87) was transfected with mutated IDH1R132H; the expression and subcellular location of TAZ were analyzed by western blot assay, quantitative real-time polymerase chain reaction, and immunofluorescence staining. We detected activation of the Hippo signaling pathway by western blot. Octyl-2-hydroxyglutarate (Octyl-2-HG), an analog of 2-HG, was used to treat IDH1 wild-type U87 cells to determine its influence on the expression of TAZ. Cell viability assay, flow cytometry, Transwell migration assay, and scratch assay were used to analyze cell proliferation and invasive capacity. To verify that those changes were caused by the expression of TAZ, we did a rescue experiment by transfecting TAZ in the IDH1R132H cells. The study was approved by the Ethics Committee of Fudan University (approval No. 2016-Y013) on January 18, 2016. Results: TAZ expression was significantly lower in IDH1-mutated astrocytoma than the wild type in the same tumor grade. In IDH1-mutant cells, the nuclear TAZ location was decreased and the Hippo signaling pathway was activated as determined by TAZ phosphorylation and increased 14-3-3e expression. Treatment with Octyl-2-HG reduced the expression of TAZ. IDH1R132H cells showed decreased invasion proliferation compared with IDH1 wild-type cells. Overexpression of TAZ rescued cell migration and invasion capacity. Conclusion: In glioma, IDH1R132H mutation decreases TAZ expression and significantly reduces the invasive character of glioma cells.
背景和目的:携带异柠檬酸脱氢酶1 (IDH1)突变的胶质瘤预后改善,但这种突变如何改善生存尚不清楚。在本研究中,我们评估了不同级别星形细胞瘤中pdz结合基序基因转录共激活因子(TAZ)的表达与IDH1突变的相关性。材料与方法:采用免疫组化方法分析90例经福尔马林固定石蜡包埋的人星形细胞瘤标本中TAZ的表达。用突变的IDH1R132H转染人胶质母细胞瘤细胞系(U87);采用western blot、实时定量聚合酶链反应和免疫荧光染色分析TAZ的表达和亚细胞定位。我们通过western blot检测Hippo信号通路的激活。采用2-HG类似物Octyl-2-hydroxyglutarate (Octyl-2-HG)处理IDH1野生型U87细胞,观察其对TAZ表达的影响。采用细胞活力法、流式细胞术、Transwell迁移法和划痕法分析细胞增殖和侵袭能力。为了验证这些变化是由TAZ的表达引起的,我们在IDH1R132H细胞中转染TAZ进行了拯救实验。本研究已于2016年1月18日获复旦大学伦理委员会批准(批准号:2016- y013)。结果:在相同肿瘤级别下,idh1突变的星形细胞瘤中TAZ的表达明显低于野生型。在idh1突变细胞中,通过TAZ磷酸化和14-3-3e表达增加确定核TAZ位置减少,Hippo信号通路被激活。辛烷基-2-汞处理降低了TAZ的表达。与IDH1野生型细胞相比,IDH1R132H细胞的侵袭性增殖降低。TAZ的过表达可改善细胞的迁移和侵袭能力。结论:在胶质瘤中,IDH1R132H突变降低了TAZ的表达,显著降低了胶质瘤细胞的侵袭性。
{"title":"IDH1 mutation decreases the invasiveness of glioma by downregulating the expression and activity of TAZ","authors":"Ningning Li, Rui Zhang, Yi Sun, Chenyue Xu, Yin Wang, J. Xiong, Qi Chen, Y. Liu","doi":"10.4103/glioma.glioma_46_18","DOIUrl":"https://doi.org/10.4103/glioma.glioma_46_18","url":null,"abstract":"Background and Aim: Gliomas carrying mutated isocitrate dehydrogenase 1 (IDH1) have an improved prognosis, but how this mutation improves survival is not known. In this study, we evaluated the correlation of expression of the gene transcriptional coactivator with PDZ-binding motif (TAZ) with IDH1 mutation in astrocytomas of different grades. Materials and Methods: We analyzed the expression of TAZ by immunohistochemistry in a cohort of 90 formalin-fixed paraffin-embedded human astrocytoma samples. A human glioblastoma cell line (U87) was transfected with mutated IDH1R132H; the expression and subcellular location of TAZ were analyzed by western blot assay, quantitative real-time polymerase chain reaction, and immunofluorescence staining. We detected activation of the Hippo signaling pathway by western blot. Octyl-2-hydroxyglutarate (Octyl-2-HG), an analog of 2-HG, was used to treat IDH1 wild-type U87 cells to determine its influence on the expression of TAZ. Cell viability assay, flow cytometry, Transwell migration assay, and scratch assay were used to analyze cell proliferation and invasive capacity. To verify that those changes were caused by the expression of TAZ, we did a rescue experiment by transfecting TAZ in the IDH1R132H cells. The study was approved by the Ethics Committee of Fudan University (approval No. 2016-Y013) on January 18, 2016. Results: TAZ expression was significantly lower in IDH1-mutated astrocytoma than the wild type in the same tumor grade. In IDH1-mutant cells, the nuclear TAZ location was decreased and the Hippo signaling pathway was activated as determined by TAZ phosphorylation and increased 14-3-3e expression. Treatment with Octyl-2-HG reduced the expression of TAZ. IDH1R132H cells showed decreased invasion proliferation compared with IDH1 wild-type cells. Overexpression of TAZ rescued cell migration and invasion capacity. Conclusion: In glioma, IDH1R132H mutation decreases TAZ expression and significantly reduces the invasive character of glioma cells.","PeriodicalId":12731,"journal":{"name":"Glioma","volume":"2 1","pages":"37 - 45"},"PeriodicalIF":0.0,"publicationDate":"2019-01-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"41703686","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 1
Diffuse midline glioma, H3-K27M mutant: Awareness leads to identification 弥漫性中线胶质瘤,H3-K27M突变体:意识导致识别
Pub Date : 2019-01-01 DOI: 10.4103/glioma.glioma_1_19
Sadhana Tiwari, I. Pant, S. Chaturvedi, Gurbachan Singh
Midline astrocytic neoplasms have distinct molecular characteristics, quite different from astrocytic neoplasms with similar morphology but not located in the midline. It is imperative that neuropathologists should be aware of the existence of these tumors, so they can be correctly diagnosed. Here, we discuss the case of a 14-year-old boy who presented with acute onset of vomiting followed by loss of consciousness. Subsequent magnetic resonance imaging revealed an ill-defined exophytic lesion arising from the brainstem and extending into the left cerebellopontine angle, with areas of hemorrhage and patchy restricted diffusion. The tumor was resected. Microscopy revealed medium-sized tumor cells in diffuse sheets, having round nuclei, granular chromatin, and scant cytoplasm. Microvascular and endothelial cell proliferation in small necrotic areas were seen. Mitosis was 0–1 per high-power field. By routine histopathological analysis, all features were consistent with the diagnosis of glioblastoma. Tumor cells were immunopositive for glial fibrillary acidic protein and isocitrate dehydrogenase-1 mutation (R132H), immunonegative for p53, and retained alpha thalassemia/mental retardation syndrome X-linked. It also showed a strong immunopositivity for H3-K27M mutation. A diagnosis of a diffuse midline glioma with H3-K27M mutation corresponding to the World Health Organization Grade IV was made. This case highlights the importance of exploring signature mutations in well-defined tumor categories such as H3-K27M-mutant diffuse midline glioma.
中线星形细胞肿瘤具有明显的分子特征,与形态相似但不位于中线的星形细胞肿瘤有很大不同。神经病理学家必须意识到这些肿瘤的存在,这样才能正确诊断。在这里,我们讨论的情况下,14岁的男孩谁提出急性发作呕吐,随后失去意识。随后的磁共振成像显示一不明确的外生病变,起源于脑干,延伸到左小脑桥脑角,伴有出血和斑片状扩散受限。肿瘤被切除。显微镜显示中等大小的肿瘤细胞呈弥散片状,核圆,染色质颗粒状,胞质稀少。小坏死区微血管和内皮细胞增生。每高倍视场有丝分裂0-1。经常规组织病理学分析,所有特征符合胶质母细胞瘤的诊断。肿瘤细胞对胶质纤维酸性蛋白和异柠檬酸脱氢酶-1突变(R132H)免疫阳性,对p53免疫阴性,并保留α地中海贫血/智力低下综合征x连锁。对H3-K27M突变株也表现出较强的免疫阳性。诊断为弥漫性中线胶质瘤伴H3-K27M突变,符合世界卫生组织ⅳ级标准。该病例强调了在明确定义的肿瘤类别(如h3 - k27m突变弥漫性中线胶质瘤)中探索特征突变的重要性。
{"title":"Diffuse midline glioma, H3-K27M mutant: Awareness leads to identification","authors":"Sadhana Tiwari, I. Pant, S. Chaturvedi, Gurbachan Singh","doi":"10.4103/glioma.glioma_1_19","DOIUrl":"https://doi.org/10.4103/glioma.glioma_1_19","url":null,"abstract":"Midline astrocytic neoplasms have distinct molecular characteristics, quite different from astrocytic neoplasms with similar morphology but not located in the midline. It is imperative that neuropathologists should be aware of the existence of these tumors, so they can be correctly diagnosed. Here, we discuss the case of a 14-year-old boy who presented with acute onset of vomiting followed by loss of consciousness. Subsequent magnetic resonance imaging revealed an ill-defined exophytic lesion arising from the brainstem and extending into the left cerebellopontine angle, with areas of hemorrhage and patchy restricted diffusion. The tumor was resected. Microscopy revealed medium-sized tumor cells in diffuse sheets, having round nuclei, granular chromatin, and scant cytoplasm. Microvascular and endothelial cell proliferation in small necrotic areas were seen. Mitosis was 0–1 per high-power field. By routine histopathological analysis, all features were consistent with the diagnosis of glioblastoma. Tumor cells were immunopositive for glial fibrillary acidic protein and isocitrate dehydrogenase-1 mutation (R132H), immunonegative for p53, and retained alpha thalassemia/mental retardation syndrome X-linked. It also showed a strong immunopositivity for H3-K27M mutation. A diagnosis of a diffuse midline glioma with H3-K27M mutation corresponding to the World Health Organization Grade IV was made. This case highlights the importance of exploring signature mutations in well-defined tumor categories such as H3-K27M-mutant diffuse midline glioma.","PeriodicalId":12731,"journal":{"name":"Glioma","volume":"2 1","pages":"55 - 59"},"PeriodicalIF":0.0,"publicationDate":"2019-01-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"42815075","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 1
期刊
Glioma
全部 Acc. Chem. Res. ACS Applied Bio Materials ACS Appl. Electron. Mater. ACS Appl. Energy Mater. ACS Appl. Mater. Interfaces ACS Appl. Nano Mater. ACS Appl. Polym. Mater. ACS BIOMATER-SCI ENG ACS Catal. ACS Cent. Sci. ACS Chem. Biol. ACS Chemical Health & Safety ACS Chem. Neurosci. ACS Comb. Sci. ACS Earth Space Chem. ACS Energy Lett. ACS Infect. Dis. ACS Macro Lett. ACS Mater. Lett. ACS Med. Chem. Lett. ACS Nano ACS Omega ACS Photonics ACS Sens. ACS Sustainable Chem. Eng. ACS Synth. Biol. Anal. Chem. BIOCHEMISTRY-US Bioconjugate Chem. BIOMACROMOLECULES Chem. Res. Toxicol. Chem. Rev. Chem. Mater. CRYST GROWTH DES ENERG FUEL Environ. Sci. Technol. Environ. Sci. Technol. Lett. Eur. J. Inorg. Chem. IND ENG CHEM RES Inorg. Chem. J. Agric. Food. Chem. J. Chem. Eng. Data J. Chem. Educ. J. Chem. Inf. Model. J. Chem. Theory Comput. J. Med. Chem. J. Nat. Prod. J PROTEOME RES J. Am. Chem. Soc. LANGMUIR MACROMOLECULES Mol. Pharmaceutics Nano Lett. Org. Lett. ORG PROCESS RES DEV ORGANOMETALLICS J. Org. Chem. J. Phys. Chem. J. Phys. Chem. A J. Phys. Chem. B J. Phys. Chem. C J. Phys. Chem. Lett. Analyst Anal. Methods Biomater. Sci. Catal. Sci. Technol. Chem. Commun. Chem. Soc. Rev. CHEM EDUC RES PRACT CRYSTENGCOMM Dalton Trans. Energy Environ. Sci. ENVIRON SCI-NANO ENVIRON SCI-PROC IMP ENVIRON SCI-WAT RES Faraday Discuss. Food Funct. Green Chem. Inorg. Chem. Front. Integr. Biol. J. Anal. At. Spectrom. J. Mater. Chem. A J. Mater. Chem. B J. Mater. Chem. C Lab Chip Mater. Chem. Front. Mater. Horiz. MEDCHEMCOMM Metallomics Mol. Biosyst. Mol. Syst. Des. Eng. Nanoscale Nanoscale Horiz. Nat. Prod. Rep. New J. Chem. Org. Biomol. Chem. Org. Chem. Front. PHOTOCH PHOTOBIO SCI PCCP Polym. Chem.
×
引用
GB/T 7714-2015
复制
MLA
复制
APA
复制
导出至
BibTeX EndNote RefMan NoteFirst NoteExpress
×
0
微信
客服QQ
Book学术公众号 扫码关注我们
反馈
×
意见反馈
请填写您的意见或建议
请填写您的手机或邮箱
×
提示
您的信息不完整,为了账户安全,请先补充。
现在去补充
×
提示
您因"违规操作"
具体请查看互助需知
我知道了
×
提示
现在去查看 取消
×
提示
确定
Book学术官方微信
Book学术文献互助
Book学术文献互助群
群 号:481959085
Book学术
文献互助 智能选刊 最新文献 互助须知 联系我们:info@booksci.cn
Book学术提供免费学术资源搜索服务,方便国内外学者检索中英文文献。致力于提供最便捷和优质的服务体验。
Copyright © 2023 Book学术 All rights reserved.
ghs 京公网安备 11010802042870号 京ICP备2023020795号-1