首页 > 最新文献

Neuro-oncology最新文献

英文 中文
Inhibition of Mitochondrial Bioenergetics and Hypoxia to Radiosensitize Diffuse Intrinsic Pontine Glioma. 抑制线粒体生物能和缺氧使弥漫性内生性桥脑胶质瘤放射敏化
IF 16.4 1区 医学 Q1 CLINICAL NEUROLOGY Pub Date : 2024-11-22 DOI: 10.1093/neuonc/noae255
Han Shen, Faiqa Mudassar, Shiyong Ma, Xingyu Wang, Sandy Nguyen, Neha Bal, Quy-Susan Huynh, Dongwei Wang, Cecilia Chang, Prunella Ing, Winny Varikatt, Joey Lai, Brian Gloss, Jeff Holst, Geraldine M O'Neill, Harriet Gee, Kristina M Cook, Eric Hau

Background: Diffuse Intrinsic Pontine Gliomas (DIPG) and other H3K27M-mutated diffuse midline gliomas (DMGs) are brain tumors that primarily affect children. Radiotherapy is the standard of care but only provides temporary symptomatic relief due to radioresistance. While hypoxia is a major driver of radioresistance in other tumors, there is no definitive evidence that DIPGs are hypoxic. DIPGs often contain histone mutations, which alter tumor metabolism and are also associated with radioresistance. Our objective was to identify the metabolic profiles of DIPG cells, detect hypoxia signatures, and uncover metabolism-linked mechanisms of radioresistance to improve tumor radiosensitivity.

Method: Using DIPG models combined with clinical datasets, we examined mitochondrial metabolism and signatures of hypoxia. We explored DIPG reliance on mitochondrial metabolism using extracellular flux assays and targeted metabolomics. In vitro and in vivo models were used to explore the mechanisms of targeting mitochondrial bioenergetics and hypoxia for radiosensitization. Treatment-induced transcriptomics and metabolomics were also investigated.

Results: Comprehensive analyses of DIPG cells show signatures of enhanced oxidative phosphorylation (OXPHOS). We also identified increased expression of specific OXPHOS related genes and signatures of hypoxia gene expression in datasets obtained from DIPG patients. We found the presence of hypoxia in orthotopic mouse models bearing DIPG tumors. These findings enabled us to develop a proof-of-concept treatment strategy to enhance radiosensitivity of DIPGs in vitro and in animal models.

Conclusion: DIPG cells rely on mitochondrial metabolism for growth, and targeting mitochondria disrupts bioenergetics, alleviates hypoxia, and enhances radiosensitivity. These findings warrant further exploration of OXPHOS inhibition as a radiosensitizing strategy for DIPG treatment.

背景:弥漫性桥脑胶质瘤(DIPG)和其他H3K27M突变的弥漫性中线胶质瘤(DMGs)是主要影响儿童的脑肿瘤。放疗是标准的治疗方法,但由于放射抗药性,只能暂时缓解症状。虽然缺氧是其他肿瘤放射抗性的主要驱动因素,但没有确切证据表明DIPGs缺氧。DIPGs通常含有组蛋白突变,这种突变会改变肿瘤的新陈代谢,也与放射抗性有关。我们的目标是确定DIPG细胞的代谢特征,检测缺氧特征,并发现与代谢相关的放射抗性机制,以提高肿瘤的放射敏感性:我们利用 DIPG 模型和临床数据集,研究了线粒体代谢和缺氧特征。我们利用细胞外通量测定和靶向代谢组学探索了DIPG对线粒体代谢的依赖。我们使用体外和体内模型来探索针对线粒体生物能和缺氧进行放射增敏的机制。此外,还研究了治疗诱导的转录组学和代谢组学:结果:对 DIPG 细胞的综合分析显示出氧化磷酸化(OXPHOS)增强的特征。我们还在从 DIPG 患者获得的数据集中发现了特定 OXPHOS 相关基因表达的增加和缺氧基因表达的特征。我们发现,在携带 DIPG 肿瘤的正位小鼠模型中存在缺氧现象。这些发现使我们能够开发一种概念验证治疗策略,在体外和动物模型中增强 DIPGs 的放射敏感性:结论:DIPG细胞的生长依赖线粒体代谢,靶向线粒体可破坏生物能,缓解缺氧,增强放射敏感性。这些发现值得进一步探索将抑制 OXPHOS 作为 DIPG 治疗的放射增敏策略。
{"title":"Inhibition of Mitochondrial Bioenergetics and Hypoxia to Radiosensitize Diffuse Intrinsic Pontine Glioma.","authors":"Han Shen, Faiqa Mudassar, Shiyong Ma, Xingyu Wang, Sandy Nguyen, Neha Bal, Quy-Susan Huynh, Dongwei Wang, Cecilia Chang, Prunella Ing, Winny Varikatt, Joey Lai, Brian Gloss, Jeff Holst, Geraldine M O'Neill, Harriet Gee, Kristina M Cook, Eric Hau","doi":"10.1093/neuonc/noae255","DOIUrl":"https://doi.org/10.1093/neuonc/noae255","url":null,"abstract":"<p><strong>Background: </strong>Diffuse Intrinsic Pontine Gliomas (DIPG) and other H3K27M-mutated diffuse midline gliomas (DMGs) are brain tumors that primarily affect children. Radiotherapy is the standard of care but only provides temporary symptomatic relief due to radioresistance. While hypoxia is a major driver of radioresistance in other tumors, there is no definitive evidence that DIPGs are hypoxic. DIPGs often contain histone mutations, which alter tumor metabolism and are also associated with radioresistance. Our objective was to identify the metabolic profiles of DIPG cells, detect hypoxia signatures, and uncover metabolism-linked mechanisms of radioresistance to improve tumor radiosensitivity.</p><p><strong>Method: </strong>Using DIPG models combined with clinical datasets, we examined mitochondrial metabolism and signatures of hypoxia. We explored DIPG reliance on mitochondrial metabolism using extracellular flux assays and targeted metabolomics. In vitro and in vivo models were used to explore the mechanisms of targeting mitochondrial bioenergetics and hypoxia for radiosensitization. Treatment-induced transcriptomics and metabolomics were also investigated.</p><p><strong>Results: </strong>Comprehensive analyses of DIPG cells show signatures of enhanced oxidative phosphorylation (OXPHOS). We also identified increased expression of specific OXPHOS related genes and signatures of hypoxia gene expression in datasets obtained from DIPG patients. We found the presence of hypoxia in orthotopic mouse models bearing DIPG tumors. These findings enabled us to develop a proof-of-concept treatment strategy to enhance radiosensitivity of DIPGs in vitro and in animal models.</p><p><strong>Conclusion: </strong>DIPG cells rely on mitochondrial metabolism for growth, and targeting mitochondria disrupts bioenergetics, alleviates hypoxia, and enhances radiosensitivity. These findings warrant further exploration of OXPHOS inhibition as a radiosensitizing strategy for DIPG treatment.</p>","PeriodicalId":19377,"journal":{"name":"Neuro-oncology","volume":" ","pages":""},"PeriodicalIF":16.4,"publicationDate":"2024-11-22","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142687761","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Distinct epigenetic and transcriptional profiles of Epstein-Barr virus (EBV) positive and negative primary CNS lymphomas. Epstein-Barr 病毒 (EBV) 阳性和阴性原发性中枢神经系统淋巴瘤不同的表观遗传学和转录谱。
IF 16.4 1区 医学 Q1 CLINICAL NEUROLOGY Pub Date : 2024-11-22 DOI: 10.1093/neuonc/noae251
Ling Hai, Dennis Friedel, Felix Hinz, Dirk C Hoffmann, Sofia Doubrovinskaia, Hannah Rohdjess, Katharina Weidenauer, Evgeniya Denisova, Georg T Scheffler, Tobias Kessler, Alexandros Kourtesakis, Christel Herold-Mende, Octavian Henegariu, Joachim M Baehring, Jorg Dietrich, Benedikt Brors, Wolfgang Wick, Felix Sahm, Leon D Kaulen

Background: Epstein-Barr virus (EBV)+ and EBV- primary CNS lymphomas (PCNSL) carry distinct mutational landscapes, but their transcriptional and epigenetic profiles have not been integrated and compared. This precludes further insights into pathobiology and molecular differences, relevant for classification and targeted therapy.

Methods: 23 EBV- and 15 EBV+ PCNSL, histologically classified as diffuse large B-cell lymphomas, were subjected to RNA-Sequencing and EPIC methylation arrays. Unsupervised clustering analyses were performed. Differentially expressed and differentially methylated genes were identified and integrated.

Results: Two distinct transcriptional clusters were found, which separated EBV-and EBV+PCNSL (p < 0.0001). The EBV+ transcriptional signature contained genes (GPR15, FCER2/CD23, SLAMF1/CD150) closely regulated by EBV oncogenes in B-cells. Pathway enrichment analysis uncovered enhanced B-cell receptor (BCR) and WNT/beta-catenin signaling in EBV-lymphomas, whereas Interleukin-10, NOTCH, and viral life cycle pathways were upregulated in EBV+PCNSL. Correspondingly, BCR-associated SYK kinase activity was enriched in EBV-tumors while JAK2 was overrepresented in EBV+PCNSL. Epigenetic profiling revealed reduced global promoter methylation in EBV+PCNSL. Two methylation clusters were recognized, which separated EBV-and EBV+PCNSL (p < 0.0001). Epigenetic profiles were distinct from 2,788 other brain tumor and non-malignant reference samples. Promoter region hypermethylation of CD79B, a BCR subunit critical for sustained proliferation in EBV-disease, highly correlated (R = -0.7) with its transcriptional downregulation in EBV+PCNSL.

Conclusions: EBV+ and EBV- PCNSL harbor distinct transcriptional and epigenetic profiles, corroborating them as distinctive biological subtypes. Uncovered differences provide novel insights into their pathobiology, may guide molecular diagnostics and targeted therapies.

背景:爱泼斯坦-巴氏病毒(EBV)+和EBV-原发性中枢神经系统淋巴瘤(PCNSL)具有不同的突变特征,但它们的转录和表观遗传学特征尚未进行整合和比较。方法:对 23 例 EBV- 和 15 例 EBV+ PCNSL(组织学分类为弥漫大 B 细胞淋巴瘤)进行了 RNA 序列分析和 EPIC 甲基化阵列分析。进行了无监督聚类分析。对差异表达基因和差异甲基化基因进行了鉴定和整合:结果:发现了两个不同的转录集群,将 EBV 和 EBV+PCNSL 区分开来(p < 0.0001)。EBV+转录特征包含受B细胞中EBV致癌基因密切调控的基因(GPR15、FCER2/CD23、SLAMF1/CD150)。通路富集分析发现,EBV淋巴瘤中B细胞受体(BCR)和WNT/beta-catenin信号转导增强,而EBV+PCNSL中白细胞介素-10、NOTCH和病毒生命周期通路上调。相应地,BCR相关的SYK激酶活性在EBV肿瘤中富集,而JAK2在EBV+PCNSL中的比例过高。表观遗传学分析表明,EBV+PCNSL 的启动子甲基化程度降低。发现了两个甲基化簇,它们将 EBV 和 EBV+PCNSL 区分开来(p < 0.0001)。表观遗传学特征与 2788 个其他脑肿瘤和非恶性参考样本不同。CD79B是一种对EBV疾病中持续增殖至关重要的BCR亚基,其启动子区域的高甲基化与EBV+PCNSL中CD79B的转录下调高度相关(R = -0.7):结论:EBV+ PCNSL 和 EBV- PCNSL 具有不同的转录和表观遗传学特征,证实它们是不同的生物学亚型。所发现的差异为了解它们的病理生物学提供了新的视角,可为分子诊断和靶向治疗提供指导。
{"title":"Distinct epigenetic and transcriptional profiles of Epstein-Barr virus (EBV) positive and negative primary CNS lymphomas.","authors":"Ling Hai, Dennis Friedel, Felix Hinz, Dirk C Hoffmann, Sofia Doubrovinskaia, Hannah Rohdjess, Katharina Weidenauer, Evgeniya Denisova, Georg T Scheffler, Tobias Kessler, Alexandros Kourtesakis, Christel Herold-Mende, Octavian Henegariu, Joachim M Baehring, Jorg Dietrich, Benedikt Brors, Wolfgang Wick, Felix Sahm, Leon D Kaulen","doi":"10.1093/neuonc/noae251","DOIUrl":"https://doi.org/10.1093/neuonc/noae251","url":null,"abstract":"<p><strong>Background: </strong>Epstein-Barr virus (EBV)+ and EBV- primary CNS lymphomas (PCNSL) carry distinct mutational landscapes, but their transcriptional and epigenetic profiles have not been integrated and compared. This precludes further insights into pathobiology and molecular differences, relevant for classification and targeted therapy.</p><p><strong>Methods: </strong>23 EBV- and 15 EBV+ PCNSL, histologically classified as diffuse large B-cell lymphomas, were subjected to RNA-Sequencing and EPIC methylation arrays. Unsupervised clustering analyses were performed. Differentially expressed and differentially methylated genes were identified and integrated.</p><p><strong>Results: </strong>Two distinct transcriptional clusters were found, which separated EBV-and EBV+PCNSL (p < 0.0001). The EBV+ transcriptional signature contained genes (GPR15, FCER2/CD23, SLAMF1/CD150) closely regulated by EBV oncogenes in B-cells. Pathway enrichment analysis uncovered enhanced B-cell receptor (BCR) and WNT/beta-catenin signaling in EBV-lymphomas, whereas Interleukin-10, NOTCH, and viral life cycle pathways were upregulated in EBV+PCNSL. Correspondingly, BCR-associated SYK kinase activity was enriched in EBV-tumors while JAK2 was overrepresented in EBV+PCNSL. Epigenetic profiling revealed reduced global promoter methylation in EBV+PCNSL. Two methylation clusters were recognized, which separated EBV-and EBV+PCNSL (p < 0.0001). Epigenetic profiles were distinct from 2,788 other brain tumor and non-malignant reference samples. Promoter region hypermethylation of CD79B, a BCR subunit critical for sustained proliferation in EBV-disease, highly correlated (R = -0.7) with its transcriptional downregulation in EBV+PCNSL.</p><p><strong>Conclusions: </strong>EBV+ and EBV- PCNSL harbor distinct transcriptional and epigenetic profiles, corroborating them as distinctive biological subtypes. Uncovered differences provide novel insights into their pathobiology, may guide molecular diagnostics and targeted therapies.</p>","PeriodicalId":19377,"journal":{"name":"Neuro-oncology","volume":" ","pages":""},"PeriodicalIF":16.4,"publicationDate":"2024-11-22","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142687742","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
EANO guideline on molecular testing of meningiomas for targeted therapy selection. 脑膜瘤分子检测用于靶向治疗选择的 EANO 指南。
IF 16.4 1区 医学 Q1 CLINICAL NEUROLOGY Pub Date : 2024-11-22 DOI: 10.1093/neuonc/noae253
Felix Sahm, Luca Bertero, Sebastian Brandner, David Capper, Roland Goldbrunner, Michael D Jenkinson, Michel Kalamarides, Katrin Lamszus, Nathalie L Albert, Maximilian J Mair, Anna S Berghoff, Christian Mawrin, Hans-Georg Wirsching, Sybren Ln Maas, David R Raleigh, Guido Reifenberger, Leonille Schweizer, Abigail K Suwala, Ghazaleh Tabatabai, Emeline Tabouret, Susan Short, Patrick Y Wen, Michael Weller, Emilie Le Rhun, Pieter Wesseling, Martin van den Bent, Matthias Preusser

Meningiomas are the most common primary intracranial tumors of adults. For meningiomas that progress or recur despite surgical resection and radiotherapy, additional treatment options are limited due to lack of proven efficacy. Meningiomas show recurring molecular aberrations, which may serve as predictive markers for systemic pharmacotherapies with targeted drugs or immunotherapy, radiotherapy or radioligand therapy. Here, we review the evidence for a predictive role of a wide range of molecular alterations and markers including NF2, AKT1, SMO, SMARCE1, PIK3CA, CDKN2A/B, CDK4/6, TERT, TRAF7, BAP1, KLF4, ARID1/2, SUFU, PD-L1, SSTR2A, PR/ER, mTOR, VEGFR, PDGFR, as well as homologous recombination deficiency (HRD), genomic copy number variations, DNA methylation classes and combined gene expression profiles. In our assessment based on the established ESMO ESCAT (European Society for Medical Oncology Scale for Clinical Actionability of molecular Targets) evidence level criteria, no molecular target reached ESCAT I ("ready for clinical use") classification and only mTOR pathway activation and NF2 alterations reached ESCAT II ("investigational") classification, respectively. Our evaluations may guide targeted therapy selection in clinical practice and clinical trial efforts and highlight areas for which additional research is warranted.

脑膜瘤是成人最常见的原发性颅内肿瘤。对于手术切除和放疗后仍有进展或复发的脑膜瘤,由于缺乏经证实的疗效,其他治疗方案十分有限。脑膜瘤会反复出现分子畸变,这些畸变可作为靶向药物或免疫疗法、放射疗法或放射性同位素疗法等全身药物疗法的预测标志物。SUFU、PD-L1、SSTR2A、PR/ER、mTOR、VEGFR、PDGFR,以及同源重组缺陷(HRD)、基因组拷贝数变异、DNA 甲基化类别和综合基因表达谱。在我们根据既定的ESMO ESCAT(欧洲肿瘤内科学会分子靶点临床可操作性量表)证据等级标准进行的评估中,没有一个分子靶点达到ESCAT I("可用于临床")等级,只有mTOR通路激活和NF2改变分别达到ESCAT II("研究中")等级。我们的评估结果可以指导临床实践和临床试验中靶向治疗的选择,并强调需要进一步研究的领域。
{"title":"EANO guideline on molecular testing of meningiomas for targeted therapy selection.","authors":"Felix Sahm, Luca Bertero, Sebastian Brandner, David Capper, Roland Goldbrunner, Michael D Jenkinson, Michel Kalamarides, Katrin Lamszus, Nathalie L Albert, Maximilian J Mair, Anna S Berghoff, Christian Mawrin, Hans-Georg Wirsching, Sybren Ln Maas, David R Raleigh, Guido Reifenberger, Leonille Schweizer, Abigail K Suwala, Ghazaleh Tabatabai, Emeline Tabouret, Susan Short, Patrick Y Wen, Michael Weller, Emilie Le Rhun, Pieter Wesseling, Martin van den Bent, Matthias Preusser","doi":"10.1093/neuonc/noae253","DOIUrl":"https://doi.org/10.1093/neuonc/noae253","url":null,"abstract":"<p><p>Meningiomas are the most common primary intracranial tumors of adults. For meningiomas that progress or recur despite surgical resection and radiotherapy, additional treatment options are limited due to lack of proven efficacy. Meningiomas show recurring molecular aberrations, which may serve as predictive markers for systemic pharmacotherapies with targeted drugs or immunotherapy, radiotherapy or radioligand therapy. Here, we review the evidence for a predictive role of a wide range of molecular alterations and markers including NF2, AKT1, SMO, SMARCE1, PIK3CA, CDKN2A/B, CDK4/6, TERT, TRAF7, BAP1, KLF4, ARID1/2, SUFU, PD-L1, SSTR2A, PR/ER, mTOR, VEGFR, PDGFR, as well as homologous recombination deficiency (HRD), genomic copy number variations, DNA methylation classes and combined gene expression profiles. In our assessment based on the established ESMO ESCAT (European Society for Medical Oncology Scale for Clinical Actionability of molecular Targets) evidence level criteria, no molecular target reached ESCAT I (\"ready for clinical use\") classification and only mTOR pathway activation and NF2 alterations reached ESCAT II (\"investigational\") classification, respectively. Our evaluations may guide targeted therapy selection in clinical practice and clinical trial efforts and highlight areas for which additional research is warranted.</p>","PeriodicalId":19377,"journal":{"name":"Neuro-oncology","volume":" ","pages":""},"PeriodicalIF":16.4,"publicationDate":"2024-11-22","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142693323","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
G-quadruplex stabilizer CX-5461 effectively combines with radiotherapy to target ATRX-deficient malignant glioma. G-四叠体稳定剂CX-5461与放疗有效结合,可靶向ATRX缺陷型恶性胶质瘤。
IF 16.4 1区 医学 Q1 CLINICAL NEUROLOGY Pub Date : 2024-11-21 DOI: 10.1093/neuonc/noae248
Sharvari Dharmaiah, Prit Benny Malgulwar, William E Johnson, Brandon A Chen, Vladislav Sharin, Benjamin T Whitfield, Christian Alvarez, Vasudev Tadimeti, Ahsan S Farooqi, Jason T Huse

Background: Inactivation of α-thalassaemia/mental retardation X-linked (ATRX) represents a defining molecular feature in large subsets of malignant glioma. ATRX deficiency gives rise to abnormal G-quadruplex (G4) DNA secondary structures, enhancing replication stress and genomic instability. Building on earlier work, we evaluated the extent to which pharmacological G4 stabilization selectively enhances DNA damage and cell death in ATRX-deficient preclinical glioma models.

Methods: Using the G4 stabilizer CX-5461, we treated patient-derived glioma stem cells (GSCs) in vitro and GSC flank and intracranial murine xenografts in vivo to evaluate efficacy as both a single agent and in combination with ionizing radiation (IR), the latter a central element of current treatment standards.

Results: CX-5461 promoted dose-sensitive lethality in ATRX-deficient GSCs relative to ATRX-intact controls. Mechanistic studies revealed that CX-5461 disrupted histone variant H3.3 deposition, enhanced replication stress and DNA damage, activated p53-independent apoptosis, and induced G2/M arrest to a greater extent in ATRX-deficient GSCs than in ATRX-intact counterparts. These data were corroborated in vivo, where CX-5461/IR treatment profoundly delayed tumor growth and prolonged survival in mice bearing ATRX-deficient flank xenografts. Histopathological analyses revealed decreased proliferation, increased apoptosis, and significant G4 induction, replication stress, and DNA damage in CX-5461-treated tumors, both alone and in combination with IR. Finally, despite suboptimal blood-brain-barrier penetration, systemic CX-5461 treatment induced tangible pharmacodynamic effects in ATRX-deficient intracranial GSC models.

Conclusions: In totality, our work substantively demonstrates efficacy and defines mechanisms of action for G4 stabilization as a novel therapeutic strategy targeting ATRX-deficient malignant glioma, laying the groundwork for clinical translation.

背景:α-地中海贫血/智力迟钝X连锁(ATRX)失活是大量恶性胶质瘤亚群的一个决定性分子特征。ATRX 缺乏会导致异常的 G-四叠体(G4)DNA 二级结构,增强复制压力和基因组不稳定性。在先前工作的基础上,我们评估了药理 G4 稳定在多大程度上选择性地增强了 ATRX 缺陷临床前胶质瘤模型中的 DNA 损伤和细胞死亡:我们使用 G4 稳定剂 CX-5461 在体外处理了源自患者的胶质瘤干细胞(GSCs),并在体内处理了 GSC 侧面和颅内小鼠异种移植物,以评估其作为单药以及与电离辐射(IR)(后者是当前治疗标准的核心要素)联合使用的疗效:结果:CX-5461促进了ATRX缺陷GSC相对于ATRX完整对照组的剂量敏感致死率。机理研究显示,CX-5461破坏了组蛋白变体H3.3的沉积,增强了复制应激和DNA损伤,激活了p53依赖性凋亡,并在更大程度上诱导了ATRX缺陷型GSCs的G2/M停滞。这些数据在体内得到了证实,CX-5461/IR 治疗可显著延缓肿瘤生长,并延长携带 ATRX 缺失侧腹异种移植物的小鼠的存活时间。组织病理学分析表明,CX-5461单独或与IR联合治疗后,肿瘤增殖减少,凋亡增加,G4诱导、复制应激和DNA损伤显著。最后,尽管CX-5461的血脑屏障穿透性欠佳,但在ATRX缺陷的颅内GSC模型中,CX-5461的全身治疗诱导了明显的药效学效应:总之,我们的研究工作有力地证明了 G4 稳定化作为一种针对 ATRX 缺失型恶性胶质瘤的新型治疗策略的疗效,并确定了其作用机制,为临床转化奠定了基础。
{"title":"G-quadruplex stabilizer CX-5461 effectively combines with radiotherapy to target ATRX-deficient malignant glioma.","authors":"Sharvari Dharmaiah, Prit Benny Malgulwar, William E Johnson, Brandon A Chen, Vladislav Sharin, Benjamin T Whitfield, Christian Alvarez, Vasudev Tadimeti, Ahsan S Farooqi, Jason T Huse","doi":"10.1093/neuonc/noae248","DOIUrl":"https://doi.org/10.1093/neuonc/noae248","url":null,"abstract":"<p><strong>Background: </strong>Inactivation of α-thalassaemia/mental retardation X-linked (ATRX) represents a defining molecular feature in large subsets of malignant glioma. ATRX deficiency gives rise to abnormal G-quadruplex (G4) DNA secondary structures, enhancing replication stress and genomic instability. Building on earlier work, we evaluated the extent to which pharmacological G4 stabilization selectively enhances DNA damage and cell death in ATRX-deficient preclinical glioma models.</p><p><strong>Methods: </strong>Using the G4 stabilizer CX-5461, we treated patient-derived glioma stem cells (GSCs) in vitro and GSC flank and intracranial murine xenografts in vivo to evaluate efficacy as both a single agent and in combination with ionizing radiation (IR), the latter a central element of current treatment standards.</p><p><strong>Results: </strong>CX-5461 promoted dose-sensitive lethality in ATRX-deficient GSCs relative to ATRX-intact controls. Mechanistic studies revealed that CX-5461 disrupted histone variant H3.3 deposition, enhanced replication stress and DNA damage, activated p53-independent apoptosis, and induced G2/M arrest to a greater extent in ATRX-deficient GSCs than in ATRX-intact counterparts. These data were corroborated in vivo, where CX-5461/IR treatment profoundly delayed tumor growth and prolonged survival in mice bearing ATRX-deficient flank xenografts. Histopathological analyses revealed decreased proliferation, increased apoptosis, and significant G4 induction, replication stress, and DNA damage in CX-5461-treated tumors, both alone and in combination with IR. Finally, despite suboptimal blood-brain-barrier penetration, systemic CX-5461 treatment induced tangible pharmacodynamic effects in ATRX-deficient intracranial GSC models.</p><p><strong>Conclusions: </strong>In totality, our work substantively demonstrates efficacy and defines mechanisms of action for G4 stabilization as a novel therapeutic strategy targeting ATRX-deficient malignant glioma, laying the groundwork for clinical translation.</p>","PeriodicalId":19377,"journal":{"name":"Neuro-oncology","volume":" ","pages":""},"PeriodicalIF":16.4,"publicationDate":"2024-11-21","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142681613","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Neuroimmune-competent human brain organoid model of Diffuse Midline Glioma. 弥漫中线胶质瘤的神经免疫功能健全人脑类器官模型。
IF 16.4 1区 医学 Q1 CLINICAL NEUROLOGY Pub Date : 2024-11-19 DOI: 10.1093/neuonc/noae245
Katharina Sarnow, Emma Majercak, Qurbonali Qurbonov, Gustavo A V Cruzeiro, Daeun Jeong, Ishraq A Harque, Andrew Khalil, Lissa C Baird, Mariella G Filbin, Xin Tang

Background: Pediatric high-grade gliomas, such as diffuse midline glioma (DMG), have a poor prognosis and lack curative treatments. Current research models of DMG primarily rely on human DMG cell lines cultured in vitro or xenografted into the brains of immunodeficient mice. However, these models are insufficient to recapitulate the complex cell-cell interactions between DMG and the tumor immune microenvironment (TIME), therefore fall short of accurately reflecting how efficacious therapeutic agents or combinations will be in the clinical setting.

Methods: To address these challenges, we developed a neuroimmune-competent brain/tumor fusion organoid model system consisting entirely of human cells to investigate the interactions between DMG cells and the primary innate immune cells of the brain, microglia, in the TIME at both cellular and subcellular levels. We generated microglia-containing brain organoids (MiCBO) that carry morphologically mature, motile microglia and multiple subtypes of neurons to mimic the brain tumor microenvironment. These organoids were then fused with H3K27M mutant, TP53P27R/K132R DMG tumor spheroids to create the MiCBO-tumor fusion (MiCBO-TF) model.

Results: We utilized live imaging methods to simultaneously track the mobility of microglial cell bodies and the motility of their process, as well as the behavior of tumor cells within a human brain tissue environment. Our MiCBO-TF model faithfully recapitulated the diffuse infiltration pattern of DMG into brain tissue and revealed that microglial mobility and interactions with tumor cells are highly influenced by external factors and surrounding tissue environment.

Conclusions: The MiCBO-TF model represents a powerful platform for both mechanistic investigations and the development of precision medicine approaches for DMG.

背景:小儿高级别胶质瘤,如弥漫中线胶质瘤(DMG),预后较差,缺乏根治性治疗方法。目前的DMG研究模型主要依赖于体外培养的人类DMG细胞系或异种移植到免疫缺陷小鼠的大脑中。然而,这些模型不足以再现DMG与肿瘤免疫微环境(TIME)之间复杂的细胞-细胞相互作用,因此无法准确反映治疗药物或组合在临床环境中的疗效:为了应对这些挑战,我们开发了一种完全由人体细胞组成的神经免疫功能脑/肿瘤融合类器官模型系统,以研究DMG细胞与脑内主要先天性免疫细胞小胶质细胞在TIME中的细胞和亚细胞水平上的相互作用。我们生成了含小胶质细胞的脑器官组织(MiCBO),它携带形态成熟、运动的小胶质细胞和多种亚型神经元,以模拟脑肿瘤微环境。然后将这些有机体与H3K27M突变体、TP53P27R/K132R DMG肿瘤球体融合,创建了MiCBO-肿瘤融合(MiCBO-TF)模型:我们利用活体成像方法同时追踪了小胶质细胞体的移动性及其运动过程,以及肿瘤细胞在人脑组织环境中的行为。我们的 MiCBO-TF 模型忠实再现了 DMG 向脑组织的弥漫浸润模式,并揭示了小胶质细胞的移动性以及与肿瘤细胞的相互作用受到外部因素和周围组织环境的高度影响:MiCBO-TF模型是研究DMG机理和开发精准医疗方法的强大平台。
{"title":"Neuroimmune-competent human brain organoid model of Diffuse Midline Glioma.","authors":"Katharina Sarnow, Emma Majercak, Qurbonali Qurbonov, Gustavo A V Cruzeiro, Daeun Jeong, Ishraq A Harque, Andrew Khalil, Lissa C Baird, Mariella G Filbin, Xin Tang","doi":"10.1093/neuonc/noae245","DOIUrl":"https://doi.org/10.1093/neuonc/noae245","url":null,"abstract":"<p><strong>Background: </strong>Pediatric high-grade gliomas, such as diffuse midline glioma (DMG), have a poor prognosis and lack curative treatments. Current research models of DMG primarily rely on human DMG cell lines cultured in vitro or xenografted into the brains of immunodeficient mice. However, these models are insufficient to recapitulate the complex cell-cell interactions between DMG and the tumor immune microenvironment (TIME), therefore fall short of accurately reflecting how efficacious therapeutic agents or combinations will be in the clinical setting.</p><p><strong>Methods: </strong>To address these challenges, we developed a neuroimmune-competent brain/tumor fusion organoid model system consisting entirely of human cells to investigate the interactions between DMG cells and the primary innate immune cells of the brain, microglia, in the TIME at both cellular and subcellular levels. We generated microglia-containing brain organoids (MiCBO) that carry morphologically mature, motile microglia and multiple subtypes of neurons to mimic the brain tumor microenvironment. These organoids were then fused with H3K27M mutant, TP53P27R/K132R DMG tumor spheroids to create the MiCBO-tumor fusion (MiCBO-TF) model.</p><p><strong>Results: </strong>We utilized live imaging methods to simultaneously track the mobility of microglial cell bodies and the motility of their process, as well as the behavior of tumor cells within a human brain tissue environment. Our MiCBO-TF model faithfully recapitulated the diffuse infiltration pattern of DMG into brain tissue and revealed that microglial mobility and interactions with tumor cells are highly influenced by external factors and surrounding tissue environment.</p><p><strong>Conclusions: </strong>The MiCBO-TF model represents a powerful platform for both mechanistic investigations and the development of precision medicine approaches for DMG.</p>","PeriodicalId":19377,"journal":{"name":"Neuro-oncology","volume":" ","pages":""},"PeriodicalIF":16.4,"publicationDate":"2024-11-19","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142676296","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Longitudinal multimodal profiling of IDH-wildtype glioblastoma reveals the molecular evolution and cellular phenotypes underlying prognostically different treatment responses. IDH-野生型胶质母细胞瘤的纵向多模态特征分析揭示了预后不同的治疗反应背后的分子演变和细胞表型。
IF 16.4 1区 医学 Q1 CLINICAL NEUROLOGY Pub Date : 2024-11-19 DOI: 10.1093/neuonc/noae214
Calixto-Hope G Lucas, Nadeem N Al-Adli, Jacob S Young, Rohit Gupta, Ramin A Morshed, Jasper Wu, Ajay Ravindranathan, Anny Shai, Nancy Ann Oberheim Bush, Jennie W Taylor, John de Groot, Javier E Villanueva-Meyer, Melike Pekmezci, Arie Perry, Andrew W Bollen, Philip V Theodosopoulos, Manish K Aghi, Edward F Chang, Shawn L Hervey-Jumper, David R Raleigh, Annette M Molinaro, Joseph F Costello, Aaron A Diaz, Jennifer L Clarke, Nicholas A Butowski, Joanna J Phillips, Susan M Chang, Mitchel S Berger, David A Solomon

Background: Despite recent advances in the biology of IDH-wildtype glioblastoma, it remains a devastating disease with median survival of less than 2 years. However, the molecular underpinnings of the heterogeneous response to the current standard-of-care treatment regimen consisting of maximal safe resection, adjuvant radiation, and chemotherapy with temozolomide remain unknown.

Methods: Comprehensive histopathologic, genomic, and epigenomic evaluation of paired initial and recurrent glioblastoma specimens from 106 patients was performed to investigate the molecular evolution and cellular phenotypes underlying differential treatment responses.

Results: While TERT promoter mutation and CDKN2A homozygous deletion were early events during gliomagenesis shared by initial and recurrent tumors, most other recurrent genetic alterations (eg, EGFR, PTEN, and NF1) were commonly private to initial or recurrent tumors indicating acquisition later during clonal evolution. Furthermore, glioblastomas exhibited heterogeneous epigenomic evolution with subsets becoming more globally hypermethylated, hypomethylated, or remaining stable. Glioblastoma that underwent sarcomatous transformation had shorter interval to recurrence and were significantly enriched in NF1, TP53, and RB1 alterations and the mesenchymal epigenetic class. Patients who developed somatic hypermutation following temozolomide treatment had significantly longer interval to disease recurrence and prolonged overall survival, and increased methylation at 4 specific CpG sites in the promoter region of MGMT was significantly associated with this development of hypermutation. Finally, an epigenomic evolution signature incorporating change in DNA methylation levels across 347 critical CpG sites was developed that significantly correlated with clinical outcomes.

Conclusions: Glioblastoma undergoes heterogeneous genetic, epigenetic, and cellular evolution that underlies prognostically different treatment responses.

背景:尽管IDH-野生型胶质母细胞瘤的生物学研究取得了最新进展,但它仍然是一种毁灭性疾病,中位生存期不到2年。然而,对于目前由最大安全切除、辅助放疗和替莫唑胺化疗组成的标准治疗方案的异质性反应,其分子基础仍然未知:方法:对106名患者的配对初发和复发胶质母细胞瘤标本进行组织病理学、基因组学和表观基因组学综合评估,研究不同治疗反应背后的分子演变和细胞表型:结果:TERT启动子突变和CDKN2A同基因缺失是初发和复发肿瘤共有的胶质瘤发生过程中的早期事件,而大多数其他复发基因改变(如表皮生长因子受体、PTEN和NF1)通常发生在初发或复发肿瘤中,这表明它们是在克隆进化的后期获得的。此外,胶质母细胞瘤还表现出表观基因组的异质性进化,其亚群变得更全面地高甲基化、低甲基化或保持稳定。发生肉瘤样转化的胶质母细胞瘤复发间隔较短,NF1、TP53和RB1改变以及间质表观遗传类别明显丰富。在接受替莫唑胺治疗后发生体细胞高突变的患者,其疾病复发间隔时间明显更长,总生存期也更长,而MGMT启动子区4个特定CpG位点的甲基化增加与高突变的发生显著相关。最后,347个关键CpG位点的DNA甲基化水平变化被纳入表观基因组进化特征,该特征与临床结果有显著相关性:结论:胶质母细胞瘤经历了遗传学、表观遗传学和细胞学的异质性演变,这是预后不同的治疗反应的基础。
{"title":"Longitudinal multimodal profiling of IDH-wildtype glioblastoma reveals the molecular evolution and cellular phenotypes underlying prognostically different treatment responses.","authors":"Calixto-Hope G Lucas, Nadeem N Al-Adli, Jacob S Young, Rohit Gupta, Ramin A Morshed, Jasper Wu, Ajay Ravindranathan, Anny Shai, Nancy Ann Oberheim Bush, Jennie W Taylor, John de Groot, Javier E Villanueva-Meyer, Melike Pekmezci, Arie Perry, Andrew W Bollen, Philip V Theodosopoulos, Manish K Aghi, Edward F Chang, Shawn L Hervey-Jumper, David R Raleigh, Annette M Molinaro, Joseph F Costello, Aaron A Diaz, Jennifer L Clarke, Nicholas A Butowski, Joanna J Phillips, Susan M Chang, Mitchel S Berger, David A Solomon","doi":"10.1093/neuonc/noae214","DOIUrl":"10.1093/neuonc/noae214","url":null,"abstract":"<p><strong>Background: </strong>Despite recent advances in the biology of IDH-wildtype glioblastoma, it remains a devastating disease with median survival of less than 2 years. However, the molecular underpinnings of the heterogeneous response to the current standard-of-care treatment regimen consisting of maximal safe resection, adjuvant radiation, and chemotherapy with temozolomide remain unknown.</p><p><strong>Methods: </strong>Comprehensive histopathologic, genomic, and epigenomic evaluation of paired initial and recurrent glioblastoma specimens from 106 patients was performed to investigate the molecular evolution and cellular phenotypes underlying differential treatment responses.</p><p><strong>Results: </strong>While TERT promoter mutation and CDKN2A homozygous deletion were early events during gliomagenesis shared by initial and recurrent tumors, most other recurrent genetic alterations (eg, EGFR, PTEN, and NF1) were commonly private to initial or recurrent tumors indicating acquisition later during clonal evolution. Furthermore, glioblastomas exhibited heterogeneous epigenomic evolution with subsets becoming more globally hypermethylated, hypomethylated, or remaining stable. Glioblastoma that underwent sarcomatous transformation had shorter interval to recurrence and were significantly enriched in NF1, TP53, and RB1 alterations and the mesenchymal epigenetic class. Patients who developed somatic hypermutation following temozolomide treatment had significantly longer interval to disease recurrence and prolonged overall survival, and increased methylation at 4 specific CpG sites in the promoter region of MGMT was significantly associated with this development of hypermutation. Finally, an epigenomic evolution signature incorporating change in DNA methylation levels across 347 critical CpG sites was developed that significantly correlated with clinical outcomes.</p><p><strong>Conclusions: </strong>Glioblastoma undergoes heterogeneous genetic, epigenetic, and cellular evolution that underlies prognostically different treatment responses.</p>","PeriodicalId":19377,"journal":{"name":"Neuro-oncology","volume":" ","pages":""},"PeriodicalIF":16.4,"publicationDate":"2024-11-19","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142667847","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Phase 2 Trial of Veliparib, Local Irradiation and Temozolomide in Patients with Newly Diagnosed High-Grade Glioma: A Children's Oncology Group Study. 针对新诊断高级别胶质瘤患者的 Veliparib、局部照射和替莫唑胺 2 期试验:儿童肿瘤学小组的一项研究。
IF 16.4 1区 医学 Q1 CLINICAL NEUROLOGY Pub Date : 2024-11-19 DOI: 10.1093/neuonc/noae247
Matthias A Karajannis, Arzu Onar-Thomas, Tong Lin, Patricia A Baxter, Daniel R Boué, Bonnie L Cole, Christine Fuller, Sofia Haque, Nada Jabado, John T Lucas, Shannon M MacDonald, Celeste Matsushima, Namrata Patel, Christopher R Pierson, Mark M Souweidane, Diana L Thomas, Michael F Walsh, Wafik Zaky, Sarah E S Leary, Amar Gajjar, Maryam Fouladi, Kenneth J Cohen

Background: The outcome for pediatric patients with high-grade glioma (HGG) remains poor. Veliparib, a potent oral poly(adenosine diphosphate-ribose) polymerase (PARP) 1/2 inhibitor, enhances the activity of radiotherapy and DNA-damaging chemotherapy.

Methods: We conducted a single-arm, non-randomized phase 2 clinical trial to determine whether treatment with veliparib and radiotherapy, followed by veliparib and temozolomide, improves progression-free survival in pediatric patients with newly diagnosed HGG without H3 K27M or BRAF mutations compared to patient level data from historical cohorts with closely matching clinical and molecular features. Following surgical resection, newly diagnosed children with non-metastatic HGG were screened by rapid central pathology review and molecular testing. Eligible patients were enrolled on Stratum 1 (IDH wild-type) or Stratum 2 (IDH mutant).

Results: Both strata were closed to accrual for futility after planned interim analyses. Among the 23 eligible patients who enrolled on Stratum 1 and received protocol therapy, the 1-year event-free survival (EFS) was 23% (standard error, SE = 9%) and 1-year overall survival (OS) was 64% (SE = 10%). Among the 14 eligible patients who enrolled on Stratum 2 and received protocol therapy, the 1-year EFS was 57% (SE = 13%) and 1-year OS was 93% (SE = 0.7%).

Conclusions: Rapid central pathology review and molecular testing for eligibility was feasible. The protocol therapy including radiation, veliparib and temozolomide was well tolerated but failed to improve outcome compared to clinically and molecularly matched historical control cohorts treated with higher doses of alkylator chemotherapy.

背景:小儿高级别胶质瘤(HGG)患者的预后仍然很差。Veliparib是一种强效口服多(腺苷二磷酸核糖)聚合酶(PARP)1/2抑制剂,可增强放疗和DNA损伤化疗的活性:我们开展了一项单臂、非随机的2期临床试验,以确定与临床和分子特征密切匹配的历史队列中的患者水平数据相比,使用维利帕尼和放疗,再使用维利帕尼和替莫唑胺治疗是否能改善新诊断为HGG且无H3 K27M或BRAF突变的儿童患者的无进展生存期。手术切除后,新确诊的非转移性 HGG 儿童通过快速中央病理审查和分子检测进行筛查。符合条件的患者被纳入第1层(IDH野生型)或第2层(IDH突变型):结果:在计划的中期分析后,两个分层均因无效而终止。在23名符合条件的第1层患者中,接受方案治疗的1年无事件生存期(EFS)为23%(标准误差,SE = 9%),1年总生存期(OS)为64%(SE = 10%)。在14名符合条件的第2层患者中,接受方案治疗的1年无事件生存率为57%(标准误差=13%),1年总生存率为93%(标准误差=0.7%):结论:快速的中央病理审查和分子检测是可行的。包括放疗、维力帕利和替莫唑胺在内的方案治疗耐受性良好,但与临床和分子匹配的历史对照组相比,使用更高剂量的烷化剂化疗未能改善疗效。
{"title":"Phase 2 Trial of Veliparib, Local Irradiation and Temozolomide in Patients with Newly Diagnosed High-Grade Glioma: A Children's Oncology Group Study.","authors":"Matthias A Karajannis, Arzu Onar-Thomas, Tong Lin, Patricia A Baxter, Daniel R Boué, Bonnie L Cole, Christine Fuller, Sofia Haque, Nada Jabado, John T Lucas, Shannon M MacDonald, Celeste Matsushima, Namrata Patel, Christopher R Pierson, Mark M Souweidane, Diana L Thomas, Michael F Walsh, Wafik Zaky, Sarah E S Leary, Amar Gajjar, Maryam Fouladi, Kenneth J Cohen","doi":"10.1093/neuonc/noae247","DOIUrl":"10.1093/neuonc/noae247","url":null,"abstract":"<p><strong>Background: </strong>The outcome for pediatric patients with high-grade glioma (HGG) remains poor. Veliparib, a potent oral poly(adenosine diphosphate-ribose) polymerase (PARP) 1/2 inhibitor, enhances the activity of radiotherapy and DNA-damaging chemotherapy.</p><p><strong>Methods: </strong>We conducted a single-arm, non-randomized phase 2 clinical trial to determine whether treatment with veliparib and radiotherapy, followed by veliparib and temozolomide, improves progression-free survival in pediatric patients with newly diagnosed HGG without H3 K27M or BRAF mutations compared to patient level data from historical cohorts with closely matching clinical and molecular features. Following surgical resection, newly diagnosed children with non-metastatic HGG were screened by rapid central pathology review and molecular testing. Eligible patients were enrolled on Stratum 1 (IDH wild-type) or Stratum 2 (IDH mutant).</p><p><strong>Results: </strong>Both strata were closed to accrual for futility after planned interim analyses. Among the 23 eligible patients who enrolled on Stratum 1 and received protocol therapy, the 1-year event-free survival (EFS) was 23% (standard error, SE = 9%) and 1-year overall survival (OS) was 64% (SE = 10%). Among the 14 eligible patients who enrolled on Stratum 2 and received protocol therapy, the 1-year EFS was 57% (SE = 13%) and 1-year OS was 93% (SE = 0.7%).</p><p><strong>Conclusions: </strong>Rapid central pathology review and molecular testing for eligibility was feasible. The protocol therapy including radiation, veliparib and temozolomide was well tolerated but failed to improve outcome compared to clinically and molecularly matched historical control cohorts treated with higher doses of alkylator chemotherapy.</p>","PeriodicalId":19377,"journal":{"name":"Neuro-oncology","volume":" ","pages":""},"PeriodicalIF":16.4,"publicationDate":"2024-11-19","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142668032","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Treatment of glioblastoma patients with personalized vaccines outside clinical trials: Lessons ignored? 在临床试验之外使用个性化疫苗治疗胶质母细胞瘤患者:被忽视的教训?
IF 16.4 1区 医学 Q1 CLINICAL NEUROLOGY Pub Date : 2024-11-19 DOI: 10.1093/neuonc/noae225
Ghazaleh Tabatabai, Michael Platten, Matthias Preusser, Michael Weller, Wolfgang Wick, Martin van den Bent
{"title":"Treatment of glioblastoma patients with personalized vaccines outside clinical trials: Lessons ignored?","authors":"Ghazaleh Tabatabai, Michael Platten, Matthias Preusser, Michael Weller, Wolfgang Wick, Martin van den Bent","doi":"10.1093/neuonc/noae225","DOIUrl":"https://doi.org/10.1093/neuonc/noae225","url":null,"abstract":"","PeriodicalId":19377,"journal":{"name":"Neuro-oncology","volume":" ","pages":""},"PeriodicalIF":16.4,"publicationDate":"2024-11-19","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142676219","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Measurable disease as baseline criterion for response assessment in glioblastoma: A comparison of PET -based (PET RANO 1.0) and MRI-based (RANO) assessments. 将可测量疾病作为胶质母细胞瘤反应评估的基线标准:基于 PET(PET RANO 1.0)和基于 MRI(RANO)的评估比较。
IF 16.4 1区 医学 Q1 CLINICAL NEUROLOGY Pub Date : 2024-11-19 DOI: 10.1093/neuonc/noae208
Katharina J Müller, Robert Forbrig, Jonas Reis, Lilian Wiegand, Enio Barci, Sophie C Kunte, Lena Kaiser, Stephan Schönecker, Christian Schichor, Patrick N Harter, Niklas Thon, Louisa von Baumgarten, Matthias Preusser, Nathalie L Albert

Background: Recently, criteria based on amino acid positron emission tomography (PET) have been proposed for response assessment in diffuse gliomas (PET RANO 1.0). In this study, we compare the prevalence of measurable disease according to PET RANO 1.0 with magnetic resonance imaging (MRI)-based Response Assessment in Neuro-Oncology (RANO) criteria in glioblastoma.

Methods: We retrospectively identified patients with newly diagnosed IDH-wild-type glioblastoma who underwent [18F] Fluoroethyltyrosine (FET) PET and MRI after resection or biopsy and before radio-/radiochemotherapy. Two independent investigators analyzed measurable disease according to PET RANO 1.0 or MRI-RANO criteria. Additionally, lesion size, congruency patterns, and uptake intensity on [18F]FET PET images were assessed.

Results: We evaluated 125 patients including 49 cases after primary resection and 76 cases after biopsy. Using PET criteria, 113 out of 125 patients (90.4%) had measurable disease, with a median PET-positive volume of 15.34 cm3 (8.83-38.03). With MRI, a significantly lower proportion of patients had measurable disease (57/125, 45.6%; P < .001) with a median sum of maximum cross-sectional diameters of 35.65 mm (26.18-45.98). None of the 12 patients without measurable disease on PET had measurable disease on MRI. Contrariwise, 56/68 patients (82.4%) without measurable disease on MRI exhibited measurable disease on PET. Clinical performance status correlated significantly with PET-positive volume and MRI-based sum of diameters (P < .0059, P < .0087, respectively).

Conclusions: [18F]FET PET identifies a higher number of patients with measurable disease compared to conventional MRI in newly diagnosed glioblastoma. PET-based assessment may serve as a novel baseline parameter for evaluating residual tumor burden and improving patient stratification in glioblastoma studies. Further validation in prospective trials is warranted.

背景:最近提出了基于氨基酸正电子发射断层扫描(PET)的弥漫性胶质瘤反应评估标准(PET RANO 1.0)。在这项研究中,我们比较了根据 PET RANO 1.0 和基于磁共振成像(MRI)的神经肿瘤学反应评估(RANO)标准在胶质母细胞瘤中可测量疾病的发生率:我们回顾性地鉴定了新诊断的IDH-Wild型胶质母细胞瘤患者,这些患者在切除术或活检后、放射/放射化疗前接受了[18F]氟乙酪氨酸(FET)PET和MRI检查。两名独立研究人员根据 PET RANO 1.0 或 MRI-RANO 标准对可测量疾病进行了分析。此外,还评估了[18F]FET PET图像上的病灶大小、一致性模式和摄取强度:我们对 125 例患者进行了评估,其中 49 例患者接受了原发性切除术,76 例患者接受了活组织检查。根据 PET 标准,125 例患者中有 113 例(90.4%)有可测量的疾病,PET 阳性体积中位数为 15.34 立方厘米(8.83-38.03)。通过核磁共振成像检查,可测量疾病的患者比例明显较低(57/125,45.6%;P 结论:[18F]FET PET 可识别癌症:与传统磁共振成像相比,[18F]FET PET 能识别出更多新诊断胶质母细胞瘤患者的可测量疾病。基于 PET 的评估可作为胶质母细胞瘤研究中评估残余肿瘤负荷和改善患者分层的新基线参数。有必要在前瞻性试验中进行进一步验证。
{"title":"Measurable disease as baseline criterion for response assessment in glioblastoma: A comparison of PET -based (PET RANO 1.0) and MRI-based (RANO) assessments.","authors":"Katharina J Müller, Robert Forbrig, Jonas Reis, Lilian Wiegand, Enio Barci, Sophie C Kunte, Lena Kaiser, Stephan Schönecker, Christian Schichor, Patrick N Harter, Niklas Thon, Louisa von Baumgarten, Matthias Preusser, Nathalie L Albert","doi":"10.1093/neuonc/noae208","DOIUrl":"https://doi.org/10.1093/neuonc/noae208","url":null,"abstract":"<p><strong>Background: </strong>Recently, criteria based on amino acid positron emission tomography (PET) have been proposed for response assessment in diffuse gliomas (PET RANO 1.0). In this study, we compare the prevalence of measurable disease according to PET RANO 1.0 with magnetic resonance imaging (MRI)-based Response Assessment in Neuro-Oncology (RANO) criteria in glioblastoma.</p><p><strong>Methods: </strong>We retrospectively identified patients with newly diagnosed IDH-wild-type glioblastoma who underwent [18F] Fluoroethyltyrosine (FET) PET and MRI after resection or biopsy and before radio-/radiochemotherapy. Two independent investigators analyzed measurable disease according to PET RANO 1.0 or MRI-RANO criteria. Additionally, lesion size, congruency patterns, and uptake intensity on [18F]FET PET images were assessed.</p><p><strong>Results: </strong>We evaluated 125 patients including 49 cases after primary resection and 76 cases after biopsy. Using PET criteria, 113 out of 125 patients (90.4%) had measurable disease, with a median PET-positive volume of 15.34 cm3 (8.83-38.03). With MRI, a significantly lower proportion of patients had measurable disease (57/125, 45.6%; P < .001) with a median sum of maximum cross-sectional diameters of 35.65 mm (26.18-45.98). None of the 12 patients without measurable disease on PET had measurable disease on MRI. Contrariwise, 56/68 patients (82.4%) without measurable disease on MRI exhibited measurable disease on PET. Clinical performance status correlated significantly with PET-positive volume and MRI-based sum of diameters (P < .0059, P < .0087, respectively).</p><p><strong>Conclusions: </strong>[18F]FET PET identifies a higher number of patients with measurable disease compared to conventional MRI in newly diagnosed glioblastoma. PET-based assessment may serve as a novel baseline parameter for evaluating residual tumor burden and improving patient stratification in glioblastoma studies. Further validation in prospective trials is warranted.</p>","PeriodicalId":19377,"journal":{"name":"Neuro-oncology","volume":" ","pages":""},"PeriodicalIF":16.4,"publicationDate":"2024-11-19","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142676292","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Alternative lengthening of telomere-based immortalization renders H3G34R -mutant diffuse hemispheric glioma hypersensitive to PARP inhibitor combination regimens. 基于端粒永生的替代性延长使H3G34R突变的弥漫性半球胶质瘤对PARP抑制剂联合疗法不敏感。
IF 16.4 1区 医学 Q1 CLINICAL NEUROLOGY Pub Date : 2024-11-18 DOI: 10.1093/neuonc/noae228
Anna Laemmerer, Christian Lehmann, Lisa Mayr, Katharina Bruckner, Lisa Gabler, Daniel Senfter, Philipp Meyer, Theresa Balber, Christine Pirker, Carola N Jaunecker, Dominik Kirchhofer, Petra Vician, Michelle Griesser, Sabine Spiegl-Kreinecker, Maria T Schmook, Tatjana Traub-Weidinger, Peter Kuess, Franziska Eckert, Aniello Federico, Sibylle Madlener, Natalia Stepien, Bernhard Robl, Alicia Baumgartner, Johannes A Hainfellner, Karin Dieckmann, Christian Dorfer, Karl Roessler, Nina S Corsini, Klaus Holzmann, Wolfgang M Schmidt, Andreas Peyrl, Amedeo A Azizi, Christine Haberler, Alexander Beck, Stefan M Pfister, Julia Schueler, Daniela Loetsch-Gojo, Jürgen A Knoblich, Walter Berger, Johannes Gojo

Background: Diffuse hemispheric glioma, H3G34R/V-mutant (DHG-H3G34) is characterized by poor prognosis and lack of effective treatment options. DHG-H3G34R further harbor deactivation of Alpha-Thalassemia/Mental Retardation Syndrome X-linked protein (ATRX; DHG-H3G34R_ATRX) suggesting a unique interaction of these two oncogenic alterations. In this study, we dissect their cell biological interplay, investigate the impact on telomere stabilization and, consequently, validate a targeted therapy approach.

Methods: We characterized patient-derived primary pediatric high-grade glioma (pHGG) models for telomere-maintenance mechanisms, DNA damage stress (including protein expression, pH2AX/Rad51 foci, cell-cycle arrest) and their sensitivity towards poly-ADP polymerase inhibitor (PARPi) combinations. Human induced pluripotent stem cells (iPSCs) were used for modelling the disease. The anticancer activity of PARPi combinations in vivo was studied in Chorioallantoic Membrane (CAM) and orthotopic in vivo experiments. Finally, we treated a DHG-H3G34R_ATRX patient with a PARPi combination therapy.

Results: We elaborate that alternative lengthening of telomeres (ALT) is a key characteristic of DHG-H3G34R_ATRX. A dominant cooperative effect between H3G34R and ATRX loss in ALT activation also became apparent in iPSCs, which endogenously exert telomerase activity. In both, patient-derived DHG-H3G34R_ATRX models and H3G34R+/ATRX- iPSCs, the ALT phenotype was associated with increased basal DNA damage stress, mediating synergistic susceptibility towards PARPi (talazoparib, niraparib) combinations with topoisomerase-I inhibitors (topotecan, irinotecan). In a first-of-its-kind case, treatment of a DHG-H3G34R_ATRX patient with the brain-penetrant PARP inhibitor niraparib and topotecan resulted in a significant tumor reduction.

Conclusion: Our preclinical and clinical data strongly support the further development of PARPis together with DNA damage stress-inducing treatment regimens for DHG-H3G34R_ATRX.

背景:弥漫性大脑半球胶质瘤、H3G34R/V-突变体(DHG-H3G34)的特点是预后不良和缺乏有效的治疗方案。DHG-H3G34R还携带α-地中海贫血/智力迟钝综合征X连锁蛋白(ATRX;DHG-H3G34R_ATRX)的失活,这表明这两种致癌改变之间存在独特的相互作用。在这项研究中,我们剖析了它们在细胞生物学上的相互作用,研究了它们对端粒稳定的影响,从而验证了一种靶向治疗方法:方法:我们对源自患者的原发性儿科高级别胶质瘤(pHGG)模型的端粒维持机制、DNA损伤应激(包括蛋白质表达、pH2AX/Rad51病灶、细胞周期停滞)及其对多聚ADP聚合酶抑制剂(PARPi)组合的敏感性进行了鉴定。人类诱导多能干细胞(iPSCs)被用于建立疾病模型。在绒毛膜(CAM)和正位体内实验中研究了 PARPi 组合的体内抗癌活性。最后,我们用PARPi联合疗法治疗了一名DHG-H3G34R_ATRX患者:结果:我们阐述了端粒替代性延长(ALT)是DHG-H3G34R_ATRX的一个关键特征。在内源性端粒酶活性的 iPSCs 中,H3G34R 和 ATRX 缺失在 ALT 激活过程中的优势合作效应也很明显。在患者衍生的 DHG-H3G34R_ATRX 模型和 H3G34R+/ATRX- iPSCs 中,ALT 表型与基础 DNA 损伤应激的增加有关,它介导了 PARPi(他拉唑帕利、尼拉帕利)与拓扑异构酶-I 抑制剂(拓扑替康、伊立替康)的协同易感性。在一个首例病例中,DHG-H3G34R_ATRX患者接受脑穿透性PARP抑制剂尼拉帕利和拓博替康治疗后,肿瘤明显缩小:我们的临床前和临床数据有力地支持了针对 DHG-H3G34R_ATRX 的 PARPis 和 DNA 损伤应激诱导治疗方案的进一步发展。
{"title":"Alternative lengthening of telomere-based immortalization renders H3G34R -mutant diffuse hemispheric glioma hypersensitive to PARP inhibitor combination regimens.","authors":"Anna Laemmerer, Christian Lehmann, Lisa Mayr, Katharina Bruckner, Lisa Gabler, Daniel Senfter, Philipp Meyer, Theresa Balber, Christine Pirker, Carola N Jaunecker, Dominik Kirchhofer, Petra Vician, Michelle Griesser, Sabine Spiegl-Kreinecker, Maria T Schmook, Tatjana Traub-Weidinger, Peter Kuess, Franziska Eckert, Aniello Federico, Sibylle Madlener, Natalia Stepien, Bernhard Robl, Alicia Baumgartner, Johannes A Hainfellner, Karin Dieckmann, Christian Dorfer, Karl Roessler, Nina S Corsini, Klaus Holzmann, Wolfgang M Schmidt, Andreas Peyrl, Amedeo A Azizi, Christine Haberler, Alexander Beck, Stefan M Pfister, Julia Schueler, Daniela Loetsch-Gojo, Jürgen A Knoblich, Walter Berger, Johannes Gojo","doi":"10.1093/neuonc/noae228","DOIUrl":"10.1093/neuonc/noae228","url":null,"abstract":"<p><strong>Background: </strong>Diffuse hemispheric glioma, H3G34R/V-mutant (DHG-H3G34) is characterized by poor prognosis and lack of effective treatment options. DHG-H3G34R further harbor deactivation of Alpha-Thalassemia/Mental Retardation Syndrome X-linked protein (ATRX; DHG-H3G34R_ATRX) suggesting a unique interaction of these two oncogenic alterations. In this study, we dissect their cell biological interplay, investigate the impact on telomere stabilization and, consequently, validate a targeted therapy approach.</p><p><strong>Methods: </strong>We characterized patient-derived primary pediatric high-grade glioma (pHGG) models for telomere-maintenance mechanisms, DNA damage stress (including protein expression, pH2AX/Rad51 foci, cell-cycle arrest) and their sensitivity towards poly-ADP polymerase inhibitor (PARPi) combinations. Human induced pluripotent stem cells (iPSCs) were used for modelling the disease. The anticancer activity of PARPi combinations in vivo was studied in Chorioallantoic Membrane (CAM) and orthotopic in vivo experiments. Finally, we treated a DHG-H3G34R_ATRX patient with a PARPi combination therapy.</p><p><strong>Results: </strong>We elaborate that alternative lengthening of telomeres (ALT) is a key characteristic of DHG-H3G34R_ATRX. A dominant cooperative effect between H3G34R and ATRX loss in ALT activation also became apparent in iPSCs, which endogenously exert telomerase activity. In both, patient-derived DHG-H3G34R_ATRX models and H3G34R+/ATRX- iPSCs, the ALT phenotype was associated with increased basal DNA damage stress, mediating synergistic susceptibility towards PARPi (talazoparib, niraparib) combinations with topoisomerase-I inhibitors (topotecan, irinotecan). In a first-of-its-kind case, treatment of a DHG-H3G34R_ATRX patient with the brain-penetrant PARP inhibitor niraparib and topotecan resulted in a significant tumor reduction.</p><p><strong>Conclusion: </strong>Our preclinical and clinical data strongly support the further development of PARPis together with DNA damage stress-inducing treatment regimens for DHG-H3G34R_ATRX.</p>","PeriodicalId":19377,"journal":{"name":"Neuro-oncology","volume":" ","pages":""},"PeriodicalIF":16.4,"publicationDate":"2024-11-18","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142648280","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
期刊
Neuro-oncology
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