1
|
Lee GA, Hsu JBK, Chang YW, Hsieh LC, Li YT, Wu YC, Chu CY, Chiang YH, Guo WY, Wu CC, Chen LW, Kao HW, Lin WL, Tseng LW, Weng TW, Kuo DP, Cheng SJ, Chen YC, Huang SW, Kung HJ, Chen CY. IL-19 as a promising theranostic target to reprogram the glioblastoma immunosuppressive microenvironment. J Biomed Sci 2025; 32:34. [PMID: 40057744 PMCID: PMC11889942 DOI: 10.1186/s12929-025-01126-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2024] [Accepted: 02/13/2025] [Indexed: 04/30/2025] Open
Abstract
BACKGROUND Glioblastoma multiforme (GBM) is an aggressive brain tumor with chemoresistant, immunosuppressive, and invasive properties. Despite standard therapies, including surgery, radiotherapy, and temozolomide (TMZ) chemotherapy, tumors inevitably recur in the peritumoral region. Targeting GBM-mediated immunosuppressive and invasive properties is a promising strategy to improve clinical outcomes. METHODS We utilized clinical and genomic data from the Taiwan GBM cohort and The Cancer Genome Atlas (TCGA) to analyze RNA sequencing data from patient tumor samples, determining the association of interleukin-19 (Il-19) expression with survival and immunosuppressive activity. Gene set enrichment analysis (GSEA) was performed to assess the relationship between the enrichment levels of immune subsets and Il-19 expression level, and Ingenuity Pathway Analysis (IPA) was used to predict immune responses. Cytokine array and single-cell RNA sequencing were used to examine the effects of IL-19 blockade on tumor immune microenvironment, including tumor-infiltrating leukocyte profiles, differentiation and immunosuppressive genes expression in tumor associated macrophages (TAM). CRISPR Il-19-/- cell lines and Il-19-/- mice were used to examine the role of IL-19 in tumor invasion and M2-like macrophage-mediated immunosuppression. Additionally, we developed novel cholesterol-polyethylene glycol-superparamagnetic iron oxide-IL-19 antibody nanoparticles (CHOL-PEG-SPIO-IL-19), characterized them using dynamic light scattering and transmission electron microscopy, Fourier-Transform Infrared spectroscopy, prussian blue assay, and conducted in vivo magnetic resonance imaging (MRI) in a human glioblastoma stem cell-derived GBM animal model. RESULT Genomic screening and IPA analysis identified IL-19 as a predicted immunosuppressive cytokine in the peritumoral region, associated with poor survival in patients with GBM. Blocking IL-19 significantly inhibited tumor progression of both TMZ-sensitive (TMZ-S) and TMZ-resistant (TMZ-R) GBM-bearing mice, and modulated the immune response within the GBM microenvironment. Single-cell transcriptome analysis reveal that IL-19 antibody treatment led to a marked increase in dendritic cells and monocyte/macrophage subsets associated with interferon-gamma signaling pathways. IL-19 blockade promoted T cell activation and reprogrammed tumor-associated macrophages toward weakened pro-tumoral phenotypes with reduced Arginase 1 expression. Il19-/- M2-like bone marrow-derived macrophages with lower Arginase 1 level lost their ability to suppress CD8 T cell activation. These findings indicated that IL-19 suppression limits TAM-mediated immune suppression. Molecular studies revealed that IL-19 promotes TMZ-resistant GBM cell migration and invasion through a novel IL-19/WISP1 signaling pathway. For clinical translation, we developed a novel CHOL-PEG-SPIO-IL-19 nanoparticles to target IL-19 expression in glioblastoma tissue. MRI imaging demonstrated enhanced targeting efficiency in brain tumors, with in vivo studies showing prominent hypointense areas in T2*-weighted MRI scans of tumor-bearing mice injected with CHOL-PEG-SPIO-IL-19, highlighting nanoparticle presence in IL-19-expressing regions. Prussian blue staining further confirmed the localization of these nanoparticles in tumor tissues, verifying their potential as a diagnostic tool for detecting IL-19 expression in glioblastoma. This system offers a theranostic approach, integrating diagnostic imaging and targeted therapy for IL-19-expressing GBM. CONCLUSION IL-19 is a promising theranostic target for reversing immunosuppression and restricting the invasive activity of chemoresistant GBM cells.
Collapse
Affiliation(s)
- Gilbert Aaron Lee
- Department of Medical Research, Taipei Medical University Hospital, Taipei, Taiwan.
- Department of Microbiology and Immunology, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan.
- Child Development Research Center, Taipei Medical University Hospital, No. 250, Wu Hsing Street, Taipei, 110, Taiwan.
- TMU Research Center for Digestive Medicine, Taipei Medical University, Taipei, Taiwan.
| | - Justin Bo-Kai Hsu
- Department of Computer Science and Engineering, Yuan Ze University, Taoyuan, Taiwan
| | - Yu-Wei Chang
- Department of Medical Research, Taipei Medical University Hospital, Taipei, Taiwan
| | - Li-Chun Hsieh
- Department of Radiology, School of Medicine, College of Medicine, Taipei Medical University, No. 250, Wu Hsing Street, Taipei, 110, Taiwan
- Department of Medical Imaging, Taipei Medical University Hospital, Taipei, Taiwan
| | - Yi-Tien Li
- Translational Imaging Research Center, Taipei Medical University Hospital, Taipei, Taiwan
- Neuroscience Research Center, Taipei Medical University, Taipei, Taiwan
- Ph.D. Program in Medical Neuroscience, College of Medical Science and Technology, Taipei Medical University, Taipei, 11031, Taiwan
| | - Ying Chieh Wu
- Department of Medical Research, Taipei Medical University Hospital, Taipei, Taiwan
| | - Cheng-Ying Chu
- CRISPR Gene Targeting Core, Taipei Medical University, Taipei 110, Taiwan
- TMU Research Center of Cancer Translational Medicine, Taipei Medical University, Taipei 110, Taiwan
| | - Yung-Hsiao Chiang
- Department of Surgery, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Wan-Yuo Guo
- Department of Radiology, Taipei Veterans General Hospital, Taipei, Taiwan
| | - Chih-Chun Wu
- Department of Radiology, Taipei Veterans General Hospital, Taipei, Taiwan
| | - Liang-Wei Chen
- Department of Radiology, Taipei Veterans General Hospital, Taipei, Taiwan
| | - Hung-Wen Kao
- Radiological Diagnosis Department, Tri-Service General Hospital, Taipei, Taiwan
| | - Wan-Li Lin
- Department of Nuclear Medicine, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan
| | - Li-Wen Tseng
- Department of Medical Research, Taipei Medical University Hospital, Taipei, Taiwan
| | - Ting-Wei Weng
- Department of Medical Research, Taipei Medical University Hospital, Taipei, Taiwan
| | - Duen-Pang Kuo
- Department of Radiology, School of Medicine, College of Medicine, Taipei Medical University, No. 250, Wu Hsing Street, Taipei, 110, Taiwan
| | - Sho-Jen Cheng
- Department of Medical Imaging, Taipei Medical University Hospital, Taipei, Taiwan
| | - Yung-Chieh Chen
- Department of Medical Imaging, Taipei Medical University Hospital, Taipei, Taiwan
| | - Shiu-Wen Huang
- Department of Pharmacology, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
- Graduate Institute of Medical Sciences, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Hsing-Jien Kung
- Research Center of Cancer Translational Medicine, Taipei Medical University, Taipei, Taiwan
| | - Cheng-Yu Chen
- Department of Radiology, School of Medicine, College of Medicine, Taipei Medical University, No. 250, Wu Hsing Street, Taipei, 110, Taiwan.
- Department of Medical Imaging, Taipei Medical University Hospital, Taipei, Taiwan.
| |
Collapse
|
2
|
Patel L, Kolundzic N, Abedalthagafi M. Progress in personalized immunotherapy for patients with brain metastasis. NPJ Precis Oncol 2025; 9:31. [PMID: 39880875 PMCID: PMC11779815 DOI: 10.1038/s41698-025-00812-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2024] [Accepted: 01/20/2025] [Indexed: 01/31/2025] Open
Abstract
Brain metastasis leads to poor outcomes and CNS injury, significantly reducing quality of life and survival rates. Advances in understanding the tumor immune microenvironment have revealed the promise of immunotherapies, which, alongside surgery, chemotherapy, and radiation, offer improved survival for some patients. However, resistance to immunotherapy remains a critical challenge. This review explores the immune landscape of brain metastases, current therapies, clinical trials, and the need for personalized, biomarker-driven approaches to optimize outcomes.
Collapse
Affiliation(s)
- Lalit Patel
- Department of Pathology and Lab Medicine, Emory University School of Medicine, Atlanta, GA, USA
| | - Nikola Kolundzic
- Department of Women & Children's Health, Faculty of Life Sciences & Medicine, King's College London, London, UK
- REPROCELL Europe Ltd., Glasgow, UK
| | - Malak Abedalthagafi
- Department of Pathology and Lab Medicine, Emory University School of Medicine, Atlanta, GA, USA.
| |
Collapse
|
3
|
Ahmad O, Ahmad T, Pfister SM. IDH mutation, glioma immunogenicity, and therapeutic challenge of primary mismatch repair deficient IDH-mutant astrocytoma PMMRDIA: a systematic review. Mol Oncol 2024; 18:2822-2841. [PMID: 38339779 PMCID: PMC11619801 DOI: 10.1002/1878-0261.13598] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2023] [Revised: 12/28/2023] [Accepted: 01/22/2024] [Indexed: 02/12/2024] Open
Abstract
In 2021, Suwala et al. described Primary Mismatch Repair Deficient IDH-mutant Astrocytoma (PMMRDIA) as a distinct group of gliomas. In unsupervised clustering, PMMRDIA forms distinct cluster, separate from other IDH-mutant gliomas, including IDH-mutant gliomas with secondary mismatch repair (MMR) deficiency. In the published cohort, three patients received treatment with an immune checkpoint blocker (ICB), yet none exhibited a response, which aligns with existing knowledge about the decreased immunogenicity of IDH-mutant gliomas in comparison to IDH-wildtype. In the case of PMMRDIA, the inherent resistance to the standard-of-care temozolomide caused by MMR deficiency is an additional challenge. It is known that a gain-of-function mutation of IDH1/2 genes produces the oncometabolite R-2-hydroxyglutarate (R-2-HG), which increases DNA and histone methylation contributing to the characteristic glioma-associated CpG island methylator phenotype (G-CIMP). While other factors could be involved in remodeling the tumor microenvironment (TME) of IDH-mutant gliomas, this systematic review emphasizes the role of R-2-HG and the subsequent G-CIMP in immune suppression. This highlights a potential actionable pathway to enhance the response of ICB, which might be relevant for addressing the unmet therapeutic challenge of PMMRDIA.
Collapse
Affiliation(s)
- Olfat Ahmad
- Division of Pediatric NeurooncologyHopp Children's Cancer Center (KiTZ)HeidelbergGermany
- Division of Pediatric Neurooncology, German Cancer Research Center (DKFZ)German Cancer Consortium (DKTK)HeidelbergGermany
- Institute of Human GeneticsUniversity Hospital HeidelbergHeidelbergGermany
- University of OxfordOxfordUK
- King Hussein Cancer Center (KHCC)AmmanJordan
| | - Tahani Ahmad
- Department of Pediatric NeuroradiologyIWK Health CenterHalifaxCanada
- Dalhousie UniversityHalifaxCanada
| | - Stefan M. Pfister
- Division of Pediatric NeurooncologyHopp Children's Cancer Center (KiTZ)HeidelbergGermany
- Division of Pediatric Neurooncology, German Cancer Research Center (DKFZ)German Cancer Consortium (DKTK)HeidelbergGermany
- Department of Pediatric Hematology and OncologyHeidelberg University HospitalHeidelbergGermany
| |
Collapse
|
4
|
Lim J, Kang I, La J, Ku KB, Kang BH, Kim Y, Park WH, Lee HK. Harnessing type I interferon-mediated immunity to target malignant brain tumors. Front Immunol 2023; 14:1203929. [PMID: 37304294 PMCID: PMC10247981 DOI: 10.3389/fimmu.2023.1203929] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2023] [Accepted: 05/15/2023] [Indexed: 06/13/2023] Open
Abstract
Type I interferons have long been appreciated as a cytokine family that regulates antiviral immunity. Recently, their role in eliciting antitumor immune responses has gained increasing attention. Within the immunosuppressive tumor microenvironment (TME), interferons stimulate tumor-infiltrating lymphocytes to promote immune clearance and essentially reshape a "cold" TME into an immune-activating "hot" TME. In this review, we focus on gliomas, with an emphasis on malignant glioblastoma, as these brain tumors possess a highly invasive and heterogenous brain TME. We address how type I interferons regulate antitumor immune responses against malignant gliomas and reshape the overall immune landscape of the brain TME. Furthermore, we discuss how these findings can translate into future immunotherapies targeting brain tumors in general.
Collapse
Affiliation(s)
- Juhee Lim
- Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, Republic of Korea
| | - In Kang
- Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, Republic of Korea
| | - Jeongwoo La
- Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, Republic of Korea
| | - Keun Bon Ku
- Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, Republic of Korea
- Department of Convergent Research of Emerging Virus Infection, Korea Research Institute of Chemical Technology, Daejeon, Republic of Korea
| | - Byeong Hoon Kang
- Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, Republic of Korea
| | - Yumin Kim
- Department of Biological Sciences, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, Republic of Korea
| | - Won Hyung Park
- Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, Republic of Korea
| | - Heung Kyu Lee
- Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, Republic of Korea
- Department of Biological Sciences, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, Republic of Korea
| |
Collapse
|
5
|
Budi HS, Younus LA, Lafta MH, Parveen S, Mohammad HJ, Al-qaim ZH, Jawad MA, Parra RMR, Mustafa YF, Alhachami FR, Karampoor S, Mirzaei R. The role of miR-128 in cancer development, prevention, drug resistance, and immunotherapy. Front Oncol 2023; 12:1067974. [PMID: 36793341 PMCID: PMC9923359 DOI: 10.3389/fonc.2022.1067974] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2022] [Accepted: 12/30/2022] [Indexed: 02/03/2023] Open
Abstract
A growing body of evidence has revealed that microRNA (miRNA) expression is dysregulated in cancer, and they can act as either oncogenes or suppressors under certain conditions. Furthermore, some studies have discovered that miRNAs play a role in cancer cell drug resistance by targeting drug-resistance-related genes or influencing genes involved in cell proliferation, cell cycle, and apoptosis. In this regard, the abnormal expression of miRNA-128 (miR-128) has been found in various human malignancies, and its verified target genes are essential in cancer-related processes, including apoptosis, cell propagation, and differentiation. This review will discuss the functions and processes of miR-128 in multiple cancer types. Furthermore, the possible involvement of miR-128 in cancer drug resistance and tumor immunotherapeutic will be addressed.
Collapse
Affiliation(s)
- Hendrik Setia Budi
- Department of Oral Biology, Dental Pharmacology, Faculty of Dental Medicine, Universitas Airlangga, Surabaya, Indonesia
| | - Laith A. Younus
- Department of Clinical Laboratory Sciences, Faculty of Pharmacy, Jabir Ibn, Hayyan Medical University, Al Najaf Al Ashraf, Iraq
| | | | - Sameena Parveen
- Department of Maxillofacial Surgery and Diagnostic Sciences, College of Dentistry, Jazan University, Jazan, Saudi Arabia
| | | | | | | | | | - Yasser Fakri Mustafa
- Department of Pharmaceutical Chemistry, College of Pharmacy, University of Mosul, Mosul, Iraq
| | - Firas Rahi Alhachami
- Radiology Department, College of Health and Medical Technology, Al-Ayen University, Thi-Qar, Nasiriyah, Iraq
| | - Sajad Karampoor
- Gastrointestinal and Liver Diseases Research Center, Iran University of Medical Sciences, Tehran, Iran
| | - Rasoul Mirzaei
- Venom and Biotherapeutics Molecules Lab, Medical Biotechnology Department, Biotechnology Research Center, Pasteur Institute of Iran, Tehran, Iran
| |
Collapse
|
6
|
Cystic Glioblastoma: A Mimicker of Infection? A Case Report and Literature Review. Case Rep Oncol Med 2023; 2023:7348188. [PMID: 36704640 PMCID: PMC9873443 DOI: 10.1155/2023/7348188] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2022] [Revised: 01/05/2023] [Accepted: 01/06/2023] [Indexed: 01/19/2023] Open
Abstract
Glioblastoma multiforme (GBM) is the most frequent malignant and aggressive type of glioma. Most cases of GBM present as a single solitary solid tumor; however, there are rare instances in which it may present as a cystic lesion. Here, we report an even rarer case of GBM presenting as bilateral multicystic lesions, mimicking infectious etiology. Our case highlights the importance of identifying clinical features of cystic GBM to ensure early diagnosis and treatment. A literature review was conducted in PubMed, looking at the common characteristics and treatment options for cystic GBM.
Collapse
|
7
|
Zhou J, Li L, Jia M, Liao Q, Peng G, Luo G, Zhou Y. Dendritic cell vaccines improve the glioma microenvironment: Influence, challenges, and future directions. Cancer Med 2022; 12:7207-7221. [PMID: 36464889 PMCID: PMC10067114 DOI: 10.1002/cam4.5511] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2022] [Revised: 11/19/2022] [Accepted: 11/24/2022] [Indexed: 12/11/2022] Open
Abstract
INTRODUCTION Gliomas, especially the glioblastomas, are one of the most aggressive intracranial tumors with poor prognosis. This might be explained by the heterogeneity of tumor cells and the inhibitory immunological microenvironment. Dendritic cells (DCs), as the most potent in vivo functional antigen-presenting cells, link innate immunity with adaptive immunity. However, their function is suppressed in gliomas. Therefore, overcoming the dysfunction of DCs in the TME might be critical to treat gliomas. METHOD In this paper we proposed the specificity of the glioma microenvironment, analyzed the pathways leading to the dysfunction of DCs in tumor microenvironment of patients with glioma, summarized influence of DC-based immunotherapy on the tumor microenvironment and proposed new development directions and possible challenges of DC vaccines. RESULT DC vaccines can improve the immunosuppressive microenvironment of glioma patients. It will bring good treatment prospects to patients. We also proposed new development directions and possible challenges of DC vaccines, thus providing an integrated understanding of efficacy on DC vaccines for glioma treatment.
Collapse
Affiliation(s)
- Jing Zhou
- NHC Key Laboratory of Carcinogenesis, Hunan Cancer Hospital and The Affiliated Cancer Hospital of Xiangya School of Medicine Central South University Changsha Hunan China
- Cancer Research Institute, Basic School of Medicine Central South University Changsha Hunan China
- Hunan Key Laboratory of Cancer Metabolism, Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine Central South University Changsha Hunan China
| | - Luohong Li
- NHC Key Laboratory of Carcinogenesis, Hunan Cancer Hospital and The Affiliated Cancer Hospital of Xiangya School of Medicine Central South University Changsha Hunan China
- Cancer Research Institute, Basic School of Medicine Central South University Changsha Hunan China
- Hunan Key Laboratory of Cancer Metabolism, Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine Central South University Changsha Hunan China
| | - Minqi Jia
- Department of Radiation Oncology Peking University Cancer Hospital & Institute Beijing China
| | - Qianjin Liao
- Hunan Key Laboratory of Cancer Metabolism, Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine Central South University Changsha Hunan China
| | - Guiping Peng
- Xiangya School of Medicine Central South University Changsha China
| | - Gengqiu Luo
- Department of Pathology, Xiangya Hospital, Basic School of Medicine Central South University Changsha Hunan China
| | - Yanhong Zhou
- NHC Key Laboratory of Carcinogenesis, Hunan Cancer Hospital and The Affiliated Cancer Hospital of Xiangya School of Medicine Central South University Changsha Hunan China
- Cancer Research Institute, Basic School of Medicine Central South University Changsha Hunan China
- Hunan Key Laboratory of Cancer Metabolism, Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine Central South University Changsha Hunan China
| |
Collapse
|
8
|
The Molecular and Cellular Strategies of Glioblastoma and Non-Small-Cell Lung Cancer Cells Conferring Radioresistance. Int J Mol Sci 2022; 23:ijms232113577. [PMID: 36362359 PMCID: PMC9656305 DOI: 10.3390/ijms232113577] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2022] [Revised: 11/02/2022] [Accepted: 11/03/2022] [Indexed: 11/09/2022] Open
Abstract
Ionizing radiation (IR) has been shown to play a crucial role in the treatment of glioblastoma (GBM; grade IV) and non-small-cell lung cancer (NSCLC). Nevertheless, recent studies have indicated that radiotherapy can offer only palliation owing to the radioresistance of GBM and NSCLC. Therefore, delineating the major radioresistance mechanisms may provide novel therapeutic approaches to sensitize these diseases to IR and improve patient outcomes. This review provides insights into the molecular and cellular mechanisms underlying GBM and NSCLC radioresistance, where it sheds light on the role played by cancer stem cells (CSCs), as well as discusses comprehensively how the cellular dormancy/non-proliferating state and polyploidy impact on their survival and relapse post-IR exposure.
Collapse
|
9
|
Maqbool I, Ali SI, Paul V, Muzaffer U. Immunognetics: a tool for the identification of novel therapeutic targets in immune disorders. CLINICAL APPLICATIONS OF IMMUNOGENETICS 2022:355-376. [DOI: 10.1016/b978-0-323-90250-2.00015-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
|
10
|
Neurofibromatosis Type 1 Gene Alterations Define Specific Features of a Subset of Glioblastomas. Int J Mol Sci 2021; 23:ijms23010352. [PMID: 35008787 PMCID: PMC8745708 DOI: 10.3390/ijms23010352] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2021] [Revised: 12/14/2021] [Accepted: 12/24/2021] [Indexed: 02/08/2023] Open
Abstract
Neurofibromatosis type 1 (NF1) gene mutations or alterations occur within neurofibromatosis type 1 as well as in many different malignant tumours on the somatic level. In glioblastoma, NF1 loss of function plays a major role in inducing the mesenchymal (MES) subtype and, therefore defining the most aggressive glioblastoma. This is associated with an immune signature and mediated via the NF1–MAPK–FOSL1 axis. Specifically, increased invasion seems to be regulated via mutations in the leucine-rich domain (LRD) of the NF1 gene product neurofibromin. Novel targets for therapy may arise from neurofibromin deficiency-associated cellular mechanisms that are summarised in this review.
Collapse
|
11
|
Xu J, Song J, Xiao M, Wang C, Zhang Q, Yuan X, Tian S. RUNX1 (RUNX family transcription factor 1), a target of microRNA miR-128-3p, promotes temozolomide resistance in glioblastoma multiform by upregulating multidrug resistance-associated protein 1 (MRP1). Bioengineered 2021; 12:11768-11781. [PMID: 34895074 PMCID: PMC8810036 DOI: 10.1080/21655979.2021.2009976] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2021] [Revised: 11/15/2021] [Accepted: 11/18/2021] [Indexed: 12/22/2022] Open
Abstract
Glioblastoma multiform (GBM) is the most frequent type of malignant brain tumor with a poor prognosis. After optimal surgery, radiotherapy plus temozolomide (TMZ) is the standard treatment for GBM patients. However, the development of TMZ resistance limits its efficacy in GBM management. Runt Related Transcription Factor 1 (RUNX1) and microRNAs have been implicated in drug resistance of TMZ in GBM. In this study, we revealed the underlying mechanism of TMZ resistance and identified miR-128-3p/RUNX1 axis as a novel target for TMZ resistance in GBM. RUNX1 expression was significantly upregulated in GBM tissues as compared to normal tissues, and its expression was even higher in recurrent GBM tissues and TMZ-resistant GBM cells. RUNX1 depletion inhibited the viability, proliferation, migration, invasion and TMZ resistance of GBM cells, which could be rescued by RUNX1 overexpression. We further identified miR-128-3p as a tumor-suppressor whose overexpression restored the sensitivity of TMZ in GBM cells. miR-128-3p negatively regulated RUNX1 and subsequently downregulated multidrug resistance-associated protein 1 (MRP1). Together, the present study indicates that RUNX1 confers TMZ resistance in GBM by upregulating MRP1, which is negatively regulated by miR-128-3p. Targeting miR-128-3p/RUNX1/MRP1 axis provides a potential strategy to overcome TMZ resistance in GBM.
Collapse
Affiliation(s)
- Jianglong Xu
- Department of Neurosurgery, Affiliated Hospital of Hebei University, Baoding, China
| | - Jia Song
- School of Basic Medicine, Hebei University, Baoding, China
| | - Menglin Xiao
- Department of Neurosurgery, Affiliated Hospital of Hebei University, Baoding, China
| | - Changsheng Wang
- Department of Neurosurgery, Affiliated Hospital of Hebei University, Baoding, China
| | - Qisong Zhang
- Department of Neurosurgery, Affiliated Hospital of Hebei University, Baoding, China
| | - Xiaoye Yuan
- School of Basic Medicine, Hebei University, Baoding, China
| | - Shaohui Tian
- Department of Neurosurgery, Affiliated Hospital of Hebei University, Baoding, China
| |
Collapse
|
12
|
Saha A, Dalal Y. A glitch in the snitch: the role of linker histone H1 in shaping the epigenome in normal and diseased cells. Open Biol 2021; 11:210124. [PMID: 34343462 PMCID: PMC8331230 DOI: 10.1098/rsob.210124] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
Histone H1s or the linker histones are a family of dynamic chromatin compacting proteins that are essential for higher-order chromatin organization. These highly positively charged proteins were previously thought to function solely as repressors of transcription. However, over the last decade, there is a growing interest in understanding this multi-protein family, finding that not all variants act as repressors. Indeed, the H1 family members appear to have distinct affinities for chromatin and may potentially affect distinct functions. This would suggest a more nuanced contribution of H1 to chromatin organization. The advent of new technologies to probe H1 dynamics in vivo, combined with powerful computational biology, and in vitro imaging tools have greatly enhanced our knowledge of the mechanisms by which H1 interacts with chromatin. This family of proteins can be metaphorically compared to the Golden Snitch from the Harry Potter series, buzzing on and off several regions of the chromatin, in combat with competing transcription factors and chromatin remodellers, thereby critical to the epigenetic endgame on short and long temporal scales in the life of the nucleus. Here, we summarize recent efforts spanning structural, computational, genomic and genetic experiments which examine the linker histone as an unseen architect of chromatin fibre in normal and diseased cells and explore unanswered fundamental questions in the field.
Collapse
Affiliation(s)
- Ankita Saha
- Center for Cancer Research, National Cancer Institute, NIH, Bethesda, MD 20892, USA
| | - Yamini Dalal
- Center for Cancer Research, National Cancer Institute, NIH, Bethesda, MD 20892, USA
| |
Collapse
|
13
|
Tumor-associated hematopoietic stem and progenitor cells positively linked to glioblastoma progression. Nat Commun 2021; 12:3895. [PMID: 34162860 PMCID: PMC8222381 DOI: 10.1038/s41467-021-23995-z] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2020] [Accepted: 05/28/2021] [Indexed: 12/13/2022] Open
Abstract
Brain tumors are typically immunosuppressive and refractory to immunotherapies for reasons that remain poorly understood. The unbiased profiling of immune cell types in the tumor microenvironment may reveal immunologic networks affecting therapy and course of disease. Here we identify and validate the presence of hematopoietic stem and progenitor cells (HSPCs) within glioblastoma tissues. Furthermore, we demonstrate a positive link of tumor-associated HSPCs with malignant and immunosuppressive phenotypes. Compared to the medullary hematopoietic compartment, tumor-associated HSPCs contain a higher fraction of immunophenotypically and transcriptomically immature, CD38- cells, such as hematopoietic stem cells and multipotent progenitors, express genes related to glioblastoma progression and display signatures of active cell cycle phases. When cultured ex vivo, tumor-associated HSPCs form myeloid colonies, suggesting potential in situ myelopoiesis. In experimental models, HSPCs promote tumor cell proliferation, expression of the immune checkpoint PD-L1 and secretion of tumor promoting cytokines such as IL-6, IL-8 and CCL2, indicating concomitant support of both malignancy and immunosuppression. In patients, the amount of tumor-associated HSPCs in tumor tissues is prognostic for patient survival and correlates with immunosuppressive phenotypes. These findings identify an important element in the complex landscape of glioblastoma that may serve as a target for brain tumor immunotherapies. A deeper knowledge of the immune cell profile within the brain cancer tumor microenvironment (TM) could identify targets to improve immunotherapy efficacy. Here, in glioblastoma, the authors find haematopoietic stem and progenitor cells in the TM, which are associated with poor prognosis and increased immunosuppression.
Collapse
|
14
|
Min TL, Allen JW, Velazquez Vega JE, Neill SG, Weinberg BD. MRI Imaging Characteristics of Glioblastoma with Concurrent Gain of Chromosomes 19 and 20. ACTA ACUST UNITED AC 2021; 7:228-237. [PMID: 34199376 PMCID: PMC8293438 DOI: 10.3390/tomography7020021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2021] [Revised: 05/17/2021] [Accepted: 05/27/2021] [Indexed: 11/16/2022]
Abstract
Glioblastoma (GBM) is the most common and deadly primary brain tumor in adults. Some of the genetic variations identified thus far, such as IDH mutation and MGMT promotor methylation, have implications for survival and response to therapy. A recent analysis of long-term GBM survivors showed that concurrent gain of chromosomes 19 and 20 (19/20 co-gain) is a positive prognostic factor that is independent of IDH mutation status. In this study, we retrospectively identified 18 patients with 19/20 co-gain and compared their imaging features to a control cohort without 19/20 co-gain. Imaging features such as tumor location, size, pial invasion, and ependymal extension were examined manually. When compared without further genetic subclassification, both groups showed similar imaging features except for rates of pial invasion. When each group was subclassified by MGMT promotor methylation status however, the two groups showed different imaging features in a number of additional ways including tumor location, size, and ependymal extension. Our results indicate that different permutations of various genetic mutations that coexist in GBM may interact in unpredictable ways to affect imaging appearance, and that imaging prognostication may be better approached in the context of the global genomic profile rather than individual genetic alterations.
Collapse
Affiliation(s)
- Taejin L. Min
- Department of Radiology and Imaging Sciences, Emory University School of Medicine, Emory University Hospital, Suite D112, 1364 Clifton Road NE, Atlanta, GA 30322, USA; (T.L.M.); (J.W.A.)
| | - Jason W. Allen
- Department of Radiology and Imaging Sciences, Emory University School of Medicine, Emory University Hospital, Suite D112, 1364 Clifton Road NE, Atlanta, GA 30322, USA; (T.L.M.); (J.W.A.)
| | - Jose E. Velazquez Vega
- Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Emory University Hospital, Room H184, 1364 Clifton Road NE, Atlanta, GA 30322, USA; (J.E.V.V.); (S.G.N.)
| | - Stewart G. Neill
- Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Emory University Hospital, Room H184, 1364 Clifton Road NE, Atlanta, GA 30322, USA; (J.E.V.V.); (S.G.N.)
| | - Brent D. Weinberg
- Department of Radiology and Imaging Sciences, Emory University School of Medicine, Emory University Hospital, Suite D112, 1364 Clifton Road NE, Atlanta, GA 30322, USA; (T.L.M.); (J.W.A.)
- Correspondence:
| |
Collapse
|
15
|
Fisher JP, Adamson DC. Current FDA-Approved Therapies for High-Grade Malignant Gliomas. Biomedicines 2021; 9:biomedicines9030324. [PMID: 33810154 PMCID: PMC8004675 DOI: 10.3390/biomedicines9030324] [Citation(s) in RCA: 178] [Impact Index Per Article: 44.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2021] [Revised: 03/16/2021] [Accepted: 03/17/2021] [Indexed: 12/26/2022] Open
Abstract
The standard of care (SOC) for high-grade gliomas (HGG) is maximally safe surgical resection, followed by concurrent radiation therapy (RT) and temozolomide (TMZ) for 6 weeks, then adjuvant TMZ for 6 months. Before this SOC was established, glioblastoma (GBM) patients typically lived for less than one year after diagnosis, and no adjuvant chemotherapy had demonstrated significant survival benefits compared with radiation alone. In 2005, the Stupp et al. randomized controlled trial (RCT) on newly diagnosed GBM patients concluded that RT plus TMZ compared to RT alone significantly improved overall survival (OS) (14.6 vs. 12.1 months) and progression-free survival (PFS) at 6 months (PFS6) (53.9% vs. 36.4%). Outside of TMZ, there are four drugs and one device FDA-approved for the treatment of HGGs: lomustine, intravenous carmustine, carmustine wafer implants, bevacizumab (BVZ), and tumor treatment fields (TTFields). These treatments are now mainly used to treat recurrent HGGs and symptoms. TTFields is the only treatment that has been shown to improve OS (20.5 vs. 15.6 months) and PFS6 (56% vs. 37%) in comparison to the current SOC. TTFields is the newest addition to this list of FDA-approved treatments, but has not been universally accepted yet as part of SOC.
Collapse
Affiliation(s)
- Jacob P. Fisher
- Division of Biochemistry, Southern Virginia University, Buena Vista, VA 24416, USA
- Correspondence:
| | - David C. Adamson
- Department of Neurosurgery, School of Medicine, Emory University, Atlanta, GA 30322, USA;
- Atlanta VA Healthcare System, Decatur, GA 30033, USA
| |
Collapse
|
16
|
Nguyen HM, Guz-Montgomery K, Lowe DB, Saha D. Pathogenetic Features and Current Management of Glioblastoma. Cancers (Basel) 2021; 13:cancers13040856. [PMID: 33670551 PMCID: PMC7922739 DOI: 10.3390/cancers13040856] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2020] [Revised: 02/09/2021] [Accepted: 02/16/2021] [Indexed: 02/06/2023] Open
Abstract
Glioblastoma (GBM) is the most common form of primary malignant brain tumor with a devastatingly poor prognosis. The disease does not discriminate, affecting adults and children of both sexes, and has an average overall survival of 12-15 months, despite advances in diagnosis and rigorous treatment with chemotherapy, radiation therapy, and surgical resection. In addition, most survivors will eventually experience tumor recurrence that only imparts survival of a few months. GBM is highly heterogenous, invasive, vascularized, and almost always inaccessible for treatment. Based on all these outstanding obstacles, there have been tremendous efforts to develop alternative treatment options that allow for more efficient targeting of the tumor including small molecule drugs and immunotherapies. A number of other strategies in development include therapies based on nanoparticles, light, extracellular vesicles, and micro-RNA, and vessel co-option. Advances in these potential approaches shed a promising outlook on the future of GBM treatment. In this review, we briefly discuss the current understanding of adult GBM's pathogenetic features that promote treatment resistance. We also outline novel and promising targeted agents currently under development for GBM patients during the last few years with their current clinical status.
Collapse
|
17
|
Duggan MR, Weaver M, Khalili K. PAM (PIK3/AKT/mTOR) signaling in glia: potential contributions to brain tumors in aging. Aging (Albany NY) 2021; 13:1510-1527. [PMID: 33472174 PMCID: PMC7835031 DOI: 10.18632/aging.202459] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2020] [Accepted: 12/10/2020] [Indexed: 02/07/2023]
Abstract
Despite a growing proportion of aged individuals at risk for developing cancer in the brain, the prognosis for these conditions remains abnormally poor due to limited knowledge of underlying mechanisms and minimal treatment options. While cancer metabolism in other organs is commonly associated with upregulated glycolysis (i.e. Warburg effect) and hyperactivation of PIK3/AKT/mTOR (PAM) pathways, the unique bioenergetic demands of the central nervous system may interact with these oncogenic processes to promote tumor progression in aging. Specifically, constitutive glycolysis and PIK3/AKT/mTOR signaling in glia may be dysregulated by age-dependent alterations in neurometabolic demands, ultimately contributing to pathological processes otherwise associated with PIK3/AKT/mTOR induction (e.g. cell cycle entry, impaired autophagy, dysregulated inflammation). Although several limitations to this theoretical model exist, the consideration of aberrant PIK3/AKT/mTOR signaling in glia during aging elucidates several therapeutic opportunities for brain tumors, including non-pharmacological interventions.
Collapse
Affiliation(s)
- Michael R. Duggan
- Department of Neuroscience Lewis Katz School of Medicine at Temple University Philadelphia, PA 19140, USA
| | - Michael Weaver
- Department of Neurosurgery Temple University Hospital Philadelphia, PA 19140, USA
| | - Kamel Khalili
- Department of Neuroscience Lewis Katz School of Medicine at Temple University Philadelphia, PA 19140, USA
| |
Collapse
|
18
|
White K, Connor K, Clerkin J, Murphy BM, Salvucci M, O'Farrell AC, Rehm M, O'Brien D, Prehn JHM, Niclou SP, Lamfers MLM, Verreault M, Idbaih A, Verhaak R, Golebiewska A, Byrne AT. New hints towards a precision medicine strategy for IDH wild-type glioblastoma. Ann Oncol 2020; 31:1679-1692. [PMID: 32918998 DOI: 10.1016/j.annonc.2020.08.2336] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2020] [Revised: 08/18/2020] [Accepted: 08/31/2020] [Indexed: 12/11/2022] Open
Abstract
Glioblastoma represents the most common primary malignancy of the central nervous system in adults and remains a largely incurable disease. The elucidation of disease subtypes based on mutational profiling, gene expression and DNA methylation has so far failed to translate into improved clinical outcomes. However, new knowledge emerging from the subtyping effort in the IDH-wild-type setting may provide directions for future precision therapies. Here, we review recent learnings in the field, and further consider how tumour microenvironment differences across subtypes may reveal novel contexts of vulnerability. We discuss recent treatment approaches and ongoing trials in the IDH-wild-type glioblastoma setting, and propose an integrated discovery stratagem incorporating multi-omics, single-cell technologies and computational approaches.
Collapse
Affiliation(s)
- K White
- Precision Cancer Medicine Group, Department of Physiology and Medical Physics, Royal College of Surgeons in Ireland, Dublin, Ireland
| | - K Connor
- Precision Cancer Medicine Group, Department of Physiology and Medical Physics, Royal College of Surgeons in Ireland, Dublin, Ireland
| | - J Clerkin
- Precision Cancer Medicine Group, Department of Physiology and Medical Physics, Royal College of Surgeons in Ireland, Dublin, Ireland; National Neurosurgical Department, Beaumont Hospital, Dublin, Ireland
| | - B M Murphy
- Centre for Systems Medicine, Department of Physiology and Medical Physics, Royal College of Surgeons in Ireland, Dublin, Ireland
| | - M Salvucci
- Centre for Systems Medicine, Department of Physiology and Medical Physics, Royal College of Surgeons in Ireland, Dublin, Ireland
| | - A C O'Farrell
- Precision Cancer Medicine Group, Department of Physiology and Medical Physics, Royal College of Surgeons in Ireland, Dublin, Ireland
| | - M Rehm
- Institute of Cell Biology and Immunology and Stuttgart Research Center Systems Biology, University of Stuttgart, Stuttgart, Germany
| | - D O'Brien
- National Neurosurgical Department, Beaumont Hospital, Dublin, Ireland
| | - J H M Prehn
- Centre for Systems Medicine, Department of Physiology and Medical Physics, Royal College of Surgeons in Ireland, Dublin, Ireland
| | - S P Niclou
- NORLUX Neuro-Oncology Laboratory, Department of Oncology, Luxembourg Institute of Health, Luxembourg
| | - M L M Lamfers
- Department of Neurosurgery, Brain Tumor Center, Erasmus MC, University Medical Center Rotterdam, Rotterdam, Netherlands
| | - M Verreault
- Sorbonne Université, Institut du Cerveau et de la Moelle Épinière, Hôpitaux Universitaires La Pitié Salpêtrière - Charles Foix, Service de Neurologie, Paris, France
| | - A Idbaih
- Sorbonne Université, Institut du Cerveau et de la Moelle Épinière, Hôpitaux Universitaires La Pitié Salpêtrière - Charles Foix, Service de Neurologie, Paris, France
| | - R Verhaak
- Jackson Laboratory for Genomic Medicine, Farmington, USA
| | - A Golebiewska
- NORLUX Neuro-Oncology Laboratory, Department of Oncology, Luxembourg Institute of Health, Luxembourg
| | - A T Byrne
- Precision Cancer Medicine Group, Department of Physiology and Medical Physics, Royal College of Surgeons in Ireland, Dublin, Ireland.
| |
Collapse
|
19
|
Zhang P, Xia Q, Liu L, Li S, Dong L. Current Opinion on Molecular Characterization for GBM Classification in Guiding Clinical Diagnosis, Prognosis, and Therapy. Front Mol Biosci 2020; 7:562798. [PMID: 33102518 PMCID: PMC7506064 DOI: 10.3389/fmolb.2020.562798] [Citation(s) in RCA: 105] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2020] [Accepted: 08/18/2020] [Indexed: 12/11/2022] Open
Abstract
Glioblastoma (GBM) is highly invasive and the deadliest brain tumor in adults. It is characterized by inter-tumor and intra-tumor heterogeneity, short patient survival, and lack of effective treatment. Prognosis and therapy selection is driven by molecular data from gene transcription, genetic alterations and DNA methylation. The four GBM molecular subtypes are proneural, neural, classical, and mesenchymal. More effective personalized therapy heavily depends on higher resolution molecular subtype signatures, combined with gene therapy, immunotherapy and organoid technology. In this review, we summarize the principal GBM molecular classifications that guide diagnosis, prognosis, and therapeutic recommendations.
Collapse
Affiliation(s)
- Pei Zhang
- School of Life Sciences, Beijing Institute of Technology, Beijing, China
| | - Qin Xia
- School of Life Sciences, Beijing Institute of Technology, Beijing, China
| | - Liqun Liu
- School of Life Sciences, Beijing Institute of Technology, Beijing, China
| | - Shouwei Li
- Department of Neurosurgery, Sanbo Brain Hospital, Capital Medical University, Beijing, China
| | - Lei Dong
- School of Life Sciences, Beijing Institute of Technology, Beijing, China
| |
Collapse
|
20
|
Dumas AA, Pomella N, Rosser G, Guglielmi L, Vinel C, Millner TO, Rees J, Aley N, Sheer D, Wei J, Marisetty A, Heimberger AB, Bowman RL, Brandner S, Joyce JA, Marino S. Microglia promote glioblastoma via mTOR-mediated immunosuppression of the tumour microenvironment. EMBO J 2020; 39:e103790. [PMID: 32567735 PMCID: PMC7396846 DOI: 10.15252/embj.2019103790] [Citation(s) in RCA: 101] [Impact Index Per Article: 20.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2019] [Revised: 05/03/2020] [Accepted: 05/08/2020] [Indexed: 12/31/2022] Open
Abstract
Tumour-associated microglia/macrophages (TAM) are the most numerous non-neoplastic populations in the tumour microenvironment in glioblastoma multiforme (GBM), the most common malignant brain tumour in adulthood. The mTOR pathway, an important regulator of cell survival/proliferation, is upregulated in GBM, but little is known about the potential role of this pathway in TAM. Here, we show that GBM-initiating cells induce mTOR signalling in the microglia but not bone marrow-derived macrophages in both in vitro and in vivo GBM mouse models. mTOR-dependent regulation of STAT3 and NF-κB activity promotes an immunosuppressive microglial phenotype. This hinders effector T-cell infiltration, proliferation and immune reactivity, thereby contributing to tumour immune evasion and promoting tumour growth in mouse models. The translational value of our results is demonstrated in whole transcriptome datasets of human GBM and in a novel in vitro model, whereby expanded-potential stem cells (EPSC)-derived microglia-like cells are conditioned by syngeneic patient-derived GBM-initiating cells. These results raise the possibility that microglia could be the primary target of mTOR inhibition, rather than the intrinsic tumour cells in GBM.
Collapse
Affiliation(s)
- Anaelle A Dumas
- Blizard InstituteBarts and The London School of Medicine and DentistryQueen Mary University LondonLondonUK
| | - Nicola Pomella
- Blizard InstituteBarts and The London School of Medicine and DentistryQueen Mary University LondonLondonUK
| | - Gabriel Rosser
- Blizard InstituteBarts and The London School of Medicine and DentistryQueen Mary University LondonLondonUK
| | - Loredana Guglielmi
- Blizard InstituteBarts and The London School of Medicine and DentistryQueen Mary University LondonLondonUK
| | - Claire Vinel
- Blizard InstituteBarts and The London School of Medicine and DentistryQueen Mary University LondonLondonUK
| | - Thomas O Millner
- Blizard InstituteBarts and The London School of Medicine and DentistryQueen Mary University LondonLondonUK
| | - Jeremy Rees
- National Hospital for Neurology and NeurosurgeryUniversity College London Hospitals NHS Foundation TrustLondonUK
| | - Natasha Aley
- Division of NeuropathologyDepartment of Neurodegenerative DiseaseUCL Queen Square Institute of NeurologyLondonUK
| | - Denise Sheer
- Blizard InstituteBarts and The London School of Medicine and DentistryQueen Mary University LondonLondonUK
| | - Jun Wei
- Department of NeurosurgeryThe University of Texas MD Anderson Cancer CenterHoustonTXUSA
| | - Anantha Marisetty
- Department of NeurosurgeryThe University of Texas MD Anderson Cancer CenterHoustonTXUSA
| | - Amy B Heimberger
- Department of NeurosurgeryThe University of Texas MD Anderson Cancer CenterHoustonTXUSA
| | - Robert L Bowman
- Human Oncology and Pathogenesis ProgramMemorial Sloan Kettering Cancer CenterNew YorkNYUSA
| | - Sebastian Brandner
- National Hospital for Neurology and NeurosurgeryUniversity College London Hospitals NHS Foundation TrustLondonUK
| | - Johanna A Joyce
- Department of OncologyLudwig Institute for Cancer ResearchUniversity of LausanneLausanneSwitzerland
| | - Silvia Marino
- Blizard InstituteBarts and The London School of Medicine and DentistryQueen Mary University LondonLondonUK
| |
Collapse
|
21
|
Alharbi M, Mobark NA, Balbaid AAO, Alanazi FA, Aljabarat WAR, Bakhsh EA, Ramkissoon SH, Abedalthagafi M. Regression of ETV6-NTRK3 Infantile Glioblastoma After First-Line Treatment With Larotrectinib. JCO Precis Oncol 2020; 4:2000017. [PMID: 32923892 PMCID: PMC7446437 DOI: 10.1200/po.20.00017] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/19/2020] [Indexed: 02/07/2023] Open
Affiliation(s)
- Musa Alharbi
- Department of Paediatric Oncology, Comprehensive Cancer Centre, King Fahad Medical, Riyadh, Saudi Arabia
| | - Nahla Ali Mobark
- Department of Paediatric Oncology, Comprehensive Cancer Centre, King Fahad Medical, Riyadh, Saudi Arabia
| | - Ali Abdullah O Balbaid
- Radiation Oncology Department, Comprehensive Cancer Centre, King Fahad Medical City, Riyadh, Saudi Arabia
| | - Fatmah A Alanazi
- Department of Clinical Pharmacy, King Fahad Medical City, Riyadh, Saudi Arabia
| | | | - Eman A Bakhsh
- Radiology Department, King Fahad Medical City, Riyadh, Saudi Arabia
| | - Shakti H Ramkissoon
- Foundation Medicine, Morrisville, NC.,Wake Forest Comprehensive Cancer Center and Department of Pathology, Wake Forest School of Medicine, Winston-Salem, NC
| | - Malak Abedalthagafi
- Genomics Research Department, Saudi Human Genome Project, King Fahad Medical City and King Abdulaziz City for Science and Technology, Riyadh, Saudi Arabia
| |
Collapse
|
22
|
Newcastle Disease Virus (NDV) Oncolytic Activity in Human Glioma Tumors Is Dependent on CDKN2A-Type I IFN Gene Cluster Codeletion. Cells 2020; 9:cells9061405. [PMID: 32516884 PMCID: PMC7349162 DOI: 10.3390/cells9061405] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2020] [Revised: 05/29/2020] [Accepted: 05/31/2020] [Indexed: 12/16/2022] Open
Abstract
Glioblastoma (GBM) is the most aggressive and frequent primary brain tumor in adults with a median overall survival of 15 months. Tumor recurrence and poor prognosis are related to cancer stem cells (CSCs), which drive resistance to therapies. A common characteristic in GBM is CDKN2A gene loss, located close to the cluster of type I IFN genes at Ch9p21. Newcastle disease virus (NDV) is an avian paramyxovirus with oncolytic and immunostimulatory properties that has been proposed for the treatment of GBM. We have analyzed the CDKN2A-IFN I gene cluster in 1018 glioma tumors and evaluated the NDV oncolytic effect in six GBM CSCs ex vivo and in a mouse model. Our results indicate that more than 50% of GBM patients have some IFN deletion. Moreover, GBM susceptibility to NDV is dependent on the loss of the type I IFN. Infection of GBM with an NDV-expressing influenza virus NS1 protein can overcome the resistance to oncolysis by NDV of type I-competent cells. These results highlight the potential of using NDV vectors in antitumor therapies.
Collapse
|
23
|
von der Brelie C, Schatlo B, Bettag C, Rohde V. Safety aspects of opioid-naïve patients with high-grade glioma treated with D,L-Methadone: an observational case series. Neurosurg Rev 2020; 44:579-586. [PMID: 32060760 DOI: 10.1007/s10143-020-01250-1] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2019] [Revised: 12/22/2019] [Accepted: 01/20/2020] [Indexed: 12/31/2022]
Abstract
It was suggested that D, L-Methadone might improve the clinical course of glioma patients. Owing to massive press coverage, patients demand the prescription of D, L-Methadone, but regarding its adjunctive use in glioma therapy there is no standard medication plan. Furthermore, it is not known which side effects the administration of D, L-Methadone might harbor, especially if the patients are opioid-naïve and if D, L-Methadone therapy was managed by the patients themselves or their general practitioners. Opioid-naïve patients with high-grade glioma (new diagnosis or recurrent) receiving D, L-Methadone were included in this retrospective observational analysis. Side effects were assigned if the condition deteriorated in conjunction with the initiation of D, L-Methadone and resolved/ ameliorated after cessation of the intake/reduction of the dosage. Side effects were categorized according to the common toxicity criteria (CTC). Twenty-four patients were included. All patients were opioid-naïve and received D, L-Methadone from their general practitioners. Sixteen patients experienced side effects. The median dosage when side effects began to occur was 15.8 mg/ 24 h. Fatigue and mood changes were reported most frequently (14 of 24 patients). Five patients had severe side effects related to relatively high doses. In all cases, symptoms resolved after cessation or dose reduction. Our results show that D/L M intake lead to frequent occurrence of side effects in opioid-naïve patients especially when not handled with caution and close supervision. Patients, their relatives, their GPs and neuro-oncologists need to be informed about the broad spectrum of side effects in order to thoroughly counsel glioma patients.
Collapse
Affiliation(s)
- Christian von der Brelie
- Department of Neurosurgery, Georg August Universität of Göttingen, Robert-Koch-Strasse 40, 37075, Göttingen, Germany.
| | - Bawarjan Schatlo
- Department of Neurosurgery, Georg August Universität of Göttingen, Robert-Koch-Strasse 40, 37075, Göttingen, Germany
| | - Chistoph Bettag
- Department of Neurosurgery, Georg August Universität of Göttingen, Robert-Koch-Strasse 40, 37075, Göttingen, Germany
| | - Veit Rohde
- Department of Neurosurgery, Georg August Universität of Göttingen, Robert-Koch-Strasse 40, 37075, Göttingen, Germany
| |
Collapse
|