1
|
Zhao W, Zhang Z, Xie M, Ding F, Zheng X, Sun S, Du J. Exploring tumor-associated macrophages in glioblastoma: from diversity to therapy. NPJ Precis Oncol 2025; 9:126. [PMID: 40316746 PMCID: PMC12048723 DOI: 10.1038/s41698-025-00920-x] [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: 11/21/2024] [Accepted: 04/22/2025] [Indexed: 05/04/2025] Open
Abstract
Glioblastoma is the most aggressive and lethal cancer of the central nervous system, presenting substantial treatment challenges. The current standard treatment, which includes surgical resection followed by temozolomide and radiation, offers limited success. While immunotherapies, such as immune checkpoint inhibitors, have proven effective in other cancers, they have not demonstrated significant efficacy in GBM. Emerging research highlights the pivotal role of tumor-associated macrophages (TAMs) in supporting tumor growth, fostering treatment resistance, and shaping an immunosuppressive microenvironment. Preclinical studies show promising results for therapies targeting TAMs, suggesting potential in overcoming these barriers. TAMs consist of brain-resident microglia and bone marrow-derived macrophages, both exhibiting diverse phenotypes and functions within the tumor microenvironment. This review delves into the origin, heterogeneity, and functional roles of TAMs in GBM, underscoring their dual roles in tumor promotion and suppression. It also summarizes recent progress in TAM-targeted therapies, which may, in combination with other treatments like immunotherapy, pave the way for more effective and personalized strategies against this aggressive malignancy.
Collapse
Affiliation(s)
- Wenwen Zhao
- Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Zhi Zhang
- Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Mingyuan Xie
- Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Feng Ding
- Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Xiangrong Zheng
- Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Shicheng Sun
- Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Jianyang Du
- Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China.
| |
Collapse
|
2
|
Branco F, Cunha J, Mendes M, Sousa JJ, Vitorino C. 3D Bioprinting Models for Glioblastoma: From Scaffold Design to Therapeutic Application. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2025; 37:e2501994. [PMID: 40116532 DOI: 10.1002/adma.202501994] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/28/2025] [Indexed: 03/23/2025]
Abstract
Conventional in vitro models fail to accurately mimic the tumor in vivo characteristics, being appointed as one of the causes of clinical attrition rate. Recent advances in 3D culture techniques, replicating essential physical and biochemical cues such as cell-cell and cell-extracellular matrix interactions, have led to the development of more realistic tumor models. Bioprinting has emerged to advance the creation of 3D in vitro models, providing enhanced flexibility, scalability, and reproducibility. This is crucial for the development of more effective drug treatments, and glioblastoma (GBM) is no exception. GBM, the most common and deadly brain cancer, remains a major challenge, with a median survival of only 15 months post-diagnosis. This review highlights the key components needed for 3D bioprinted GBM models. It encompasses an analysis of natural and synthetic biomaterials, along with crosslinking methods to improve structural integrity. Also, it critically evaluates current 3D bioprinted GBM models and their integration into GBM-on-a-chip platforms, which hold noteworthy potential for drug screening and personalized therapies. A versatile development framework grounded on Quality-by-Design principles is proposed to guide the design of bioprinting models. Future perspectives, including 4D bioprinting and machine learning approaches, are discussed, along with the current gaps to advance the field further.
Collapse
Affiliation(s)
- Francisco Branco
- Faculty of Pharmacy, University of Coimbra, Pólo das Ciências da Saúde, Azinhaga de Santa Comba, Coimbra, 3000-548, Portugal
| | - Joana Cunha
- Faculty of Pharmacy, University of Coimbra, Pólo das Ciências da Saúde, Azinhaga de Santa Comba, Coimbra, 3000-548, Portugal
| | - Maria Mendes
- Faculty of Pharmacy, University of Coimbra, Pólo das Ciências da Saúde, Azinhaga de Santa Comba, Coimbra, 3000-548, Portugal
- Coimbra Chemistry Centre, Institute of Molecular Sciences - IMS, Faculty of Sciences and Technology, University of Coimbra, Coimbra, 3004-535, Portugal
| | - João J Sousa
- Faculty of Pharmacy, University of Coimbra, Pólo das Ciências da Saúde, Azinhaga de Santa Comba, Coimbra, 3000-548, Portugal
- Coimbra Chemistry Centre, Institute of Molecular Sciences - IMS, Faculty of Sciences and Technology, University of Coimbra, Coimbra, 3004-535, Portugal
| | - Carla Vitorino
- Faculty of Pharmacy, University of Coimbra, Pólo das Ciências da Saúde, Azinhaga de Santa Comba, Coimbra, 3000-548, Portugal
- Coimbra Chemistry Centre, Institute of Molecular Sciences - IMS, Faculty of Sciences and Technology, University of Coimbra, Coimbra, 3004-535, Portugal
| |
Collapse
|
3
|
Zhang Y, He H, Fu X, Liu G, Wang H, Zhong W, Xu X, Chen B, Mei L. Glioblastoma-associated macrophages in glioblastoma: from their function and mechanism to therapeutic advances. Cancer Gene Ther 2025:10.1038/s41417-025-00905-9. [PMID: 40307579 DOI: 10.1038/s41417-025-00905-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2024] [Revised: 04/01/2025] [Accepted: 04/07/2025] [Indexed: 05/02/2025]
Abstract
Glioblastoma multiforme (GBM) is the most aggressive primary brain tumor in adults and has high mortality rates worldwide. GBM progression, treatment, and prognosis are influenced by the tumor microenvironment (TME), which includes immune, stromal, and tumor cells. Among them, glioblastoma-associated macrophages (GAMs) act as key regulators of GBM pathobiology. GAMs exhibit remarkable plasticity, as they can exhibit both protumor and antitumor effects. However, their function is determined by polarization and the TME. In this review, we provide a comprehensive overview of the current understanding of the biology of GAMs in GBM, including their origins, phenotypic diversity, and functional roles. We discuss the intricate crosstalk between GAMs and tumor cells, as well as other immune and stromal components, and highlight the mechanisms underlying GAM-mediated tumor progression, invasion, angiogenesis, and immune system evasion. Furthermore, we explore the therapeutic implications of targeting GAMs in GBM and discuss emerging strategies aimed at reprogramming GAMs toward an antitumorigenic phenotype or selectively depleting protumorigenic subsets. The final aim is to develop innovative therapeutic approaches that disrupt GBMs. By leveraging our increased understanding of GAM biology, we lay the foundation for transformative advances in GBM treatment to improve patient prognosis.
Collapse
Affiliation(s)
- Yuqin Zhang
- Department of General Practice, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Hanxing He
- Department of Orthopedics and Traumatology, School of Clinical Medicine, LKS Faculty of Medicine, The University of Hong Kong, Queen Mary Hospital, Hong Kong SAR, China
| | - Xin Fu
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou, China
| | - Ganzhi Liu
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Huiying Wang
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Wen Zhong
- Department of General Practice, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Xia Xu
- Department of General Practice, Xiangya Hospital, Central South University, Changsha, Hunan, China.
| | - Bo Chen
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, Hunan, China.
- Department of Surgery, School of Clinical Medicine, LKS Faculty of Medicine, The University of Hong Kong, Queen Mary Hospital, Hong Kong SAR, China.
| | - Lin Mei
- Chinese Academy of Medical Sciences & Peking Union Medical College Institute of Biomedical Engineering, Tianjin, China.
| |
Collapse
|
4
|
Fuse H, Zheng Y, Alzoubi I, Graeber MB. TAMing Gliomas: Unraveling the Roles of Iba1 and CD163 in Glioblastoma. Cancers (Basel) 2025; 17:1457. [PMID: 40361384 PMCID: PMC12070867 DOI: 10.3390/cancers17091457] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2025] [Revised: 04/19/2025] [Accepted: 04/23/2025] [Indexed: 05/15/2025] Open
Abstract
Gliomas, the most common type of primary brain tumor, are a significant cause of morbidity and mortality worldwide. Glioblastoma, a highly malignant subtype, is particularly common, aggressive, and resistant to treatment. The tumor microenvironment (TME) of gliomas, especially glioblastomas, is characterized by a distinct presence of tumor-associated macrophages (TAMs), which densely infiltrate glioblastomas, a hallmark of these tumors. This macrophage population comprises both tissue-resident microglia as well as macrophages derived from the walls of blood vessels and the blood stream. Ionized calcium-binding adapter molecule 1 (Iba1) and CD163 are established cellular markers that enable the identification and functional characterization of these cells within the TME. This review provides an in-depth examination of the roles of Iba1 and CD163 in malignant gliomas, with a focus on TAM activation, migration, and immunomodulatory functions. Additionally, we will discuss how recent advances in AI-enhanced cell identification and visualization techniques have begun to transform the analysis of TAMs, promising unprecedented precision in their characterization and providing new insights into their roles within the TME. Iba1 and CD163 appear to have both unique and shared roles in glioma pathobiology, and both have the potential to be targeted through different molecular and cellular mechanisms. We discuss the therapeutic potential of Iba1 and CD163 based on available preclinical (experimental) and clinical (human tissue-based) evidence.
Collapse
Affiliation(s)
- Haneya Fuse
- School of Medicine, Sydney Campus, University of Notre Dame, 160 Oxford Street, Sydney, NSW 2010, Australia;
| | - Yuqi Zheng
- Ken Parker Brain Tumor Research Laboratories, Brain and Mind Centre, Faculty of Medicine and Health, University of Sydney, Sydney, NSW 2050, Australia;
| | - Islam Alzoubi
- School of Computer Science, The University of Sydney, J12/1 Cleveland St, Sydney, NSW 2008, Australia;
| | - Manuel B. Graeber
- Ken Parker Brain Tumor Research Laboratories, Brain and Mind Centre, Faculty of Medicine and Health, University of Sydney, Sydney, NSW 2050, Australia;
- University of Sydney Association of Professors (USAP), University of Sydney, Sydney, NSW 2006, Australia
| |
Collapse
|
5
|
Wang X, Gu J, Tang H, Gu L, Bi Y, Kong Y, Shan Q, Yin J, Lou M, Li S, Liu Y. Single-Cell Profiling and Proteomics-Based Insights Into mTORC1-Mediated Angio+TAMs Polarization in Recurrent IDH-Mutant Gliomas. CNS Neurosci Ther 2025; 31:e70371. [PMID: 40202138 PMCID: PMC11979715 DOI: 10.1111/cns.70371] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2024] [Revised: 01/26/2025] [Accepted: 03/23/2025] [Indexed: 04/10/2025] Open
Abstract
BACKGROUND IDH mutant gliomas often exhibit recurrence and progression, with the mTORC1 pathway and tumor-associated macrophages potentially contributing to these processes. However, the precise mechanisms are not fully understood. This study seeks to investigate these relationships using proteomic, phosphoproteomic, and multi-dimensional transcriptomic approaches. METHODS This study established a matched transcriptomic, proteomic, and phosphoproteomic cohort of IDH-mutant gliomas with recurrence and progression, incorporating multiple glioma-related datasets. We first identified the genomic landscape of recurrent IDH-mutant gliomas through multi-dimensional differential enrichment, GSVA, and deconvolution analyses. Next, we explored tumor-associated macrophage subpopulations using single-cell sequencing in mouse models of IDH-mutant and wild-type gliomas, analyzing transcriptional changes via AddmodelScore and pseudotime analysis. We then identified these subpopulations in matched primary and recurrent IDH-mutant datasets, investigating their interactions with the tumor microenvironment and performing deconvolution to explore their contribution to glioma progression. Finally, spatial transcriptomics was used to map these subpopulations to glioma tissue sections, revealing spatial co-localization with mTORC1 and angiogenesis-related pathways. RESULTS Multi-dimensional differential enrichment, GSVA, and deconvolution analyses indicated that the mTORC1 pathway and the proportion of M2 macrophages are upregulated during the recurrence and progression of IDH-mutant gliomas. CGGA database analysis showed that mTORC1 activity is significantly higher in recurrent IDH-mutant gliomas compared to IDH-wildtype, with a correlation to M2 macrophage infiltration. KSEA revealed that AURKA is enriched during progression, and its inhibition reduces mTORC1 pathway activity. Single-cell sequencing in mouse models identified a distinct glioma subpopulation with upregulated mTORC1, exhibiting both M2 macrophage and angiogenesis transcriptional features, which increased after implantation of IDH-mutant tumor cells. Similarly, human glioma single-cell data revealed the same subpopulation, with cell-cell communication analysis showing active VEGF signaling. Finally, spatial transcriptomics deconvolution confirmed the co-localization of this subpopulation with mTORC1 and VEGFA in high-grade IDH-mutant gliomas. CONCLUSIONS Our findings suggest mTORC1 activation and Angio-TAMs play key roles in the recurrence and progression of IDH-mutant gliomas.
Collapse
Affiliation(s)
- Xu Wang
- Department of NeurosurgeryShanghai General HospitalShanghaiChina
| | - Jingyan Gu
- Department of NeurosurgeryShanghai General HospitalShanghaiChina
- Shanghai Jiao Tong University School of MedicineShanghaiChina
| | - Hongyu Tang
- Department of NeurosurgeryShanghai General HospitalShanghaiChina
- Shanghai Jiao Tong University School of MedicineShanghaiChina
| | - Lianping Gu
- Department of NeurosurgeryShanghai General HospitalShanghaiChina
- Nanjing Medical UniversityJiangsuChina
| | - Yunke Bi
- Department of NeurosurgeryShanghai General HospitalShanghaiChina
| | - Yue Kong
- Sun Yat‐Sen University Guanghua School of StomatologyGuangzhouChina
| | - Qiao Shan
- Department of NeurosurgeryShanghai General HospitalShanghaiChina
| | - Jian Yin
- Department of NeurosurgeryShanghai General HospitalShanghaiChina
| | - Meiqing Lou
- Department of NeurosurgeryShanghai General HospitalShanghaiChina
- Shanghai Jiao Tong University School of MedicineShanghaiChina
- Nanjing Medical UniversityJiangsuChina
| | - Shouwei Li
- Department of NeurosurgeryCapital Medical University Sanbo Brain HospitalBeijingChina
| | - Yaohua Liu
- Department of NeurosurgeryShanghai General HospitalShanghaiChina
- Shanghai Jiao Tong University School of MedicineShanghaiChina
| |
Collapse
|
6
|
Ramachandran R, Jeans AF. Breaking Down Glioma-Microenvironment Crosstalk. Neuroscientist 2025; 31:177-194. [PMID: 39066464 PMCID: PMC11909767 DOI: 10.1177/10738584241259773] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/28/2024]
Abstract
High-grade gliomas (HGGs) are the commonest primary brain cancers. They are characterized by a pattern of aggressive growth and diffuse infiltration of the host brain that severely limits the efficacy of conventional treatments and patient outcomes, which remain generally poor. Recent work has described a suite of mechanisms via which HGGs interact, predominantly bidirectionally, with various cell types in the host brain including neurons, glial cells, immune cells, and vascular elements to drive tumor growth and invasion. These insights have the potential to inspire novel approaches to HGG therapy that are critically needed. This review explores HGG-host brain interactions and considers whether and how they might be exploited for therapeutic gain.
Collapse
|
7
|
Diep YN, Park HJ, Zhu X, Nam JH, Shim JK, Chang JH, Nguyen DD, Kim I, Jo DG, Lee LP, Yun M, Cho H. A Neuroimmune-Oncology Microphysiological Analysis Platform (NEO-MAP) for Evaluating Astrocytic Scar Formation and Microgliosis in Glioblastoma Microenvironment. Adv Healthc Mater 2025; 14:e2404821. [PMID: 40072333 DOI: 10.1002/adhm.202404821] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2025] [Revised: 02/28/2025] [Indexed: 04/26/2025]
Abstract
Glioblastoma multiforme (GBM) is the most aggressive type of brain tumor, characterized by its heterogeneity in cellular components, including reactive astrocytes and microglia. Since neuroimmune responses like astrogliosis and microgliosis gain recognition as vital factors in brain tumor progression, there is a growing need for clinically relevant models that assess the interactions between astrocytes, microglia, and GBM. Here, a NEuroimmune-Oncology Microphysiological Analysis Platform (NEO-MAP) is presented as a "new map" to observe astrocytic scar formation and microgliosis in response to GBM. NEO-MAP based on pathophysiological principles is designed to replicate the GBM-glia interactions, multi-phenotypic microglia activities, scar-forming astrocytes with chondroitin sulfate proteoglycans (CSPGs) in the extracellular matrix, and the biophysical characteristics of the astrocytic scar barrier. The NEO-MAP reveals that inhibiting mTORC2 in GBM promotes the proinflammatory transformation of astrocytes and enhanced astrocytic scar formation. Astrocytes that form scars prompted microglia to change from the M2 to M1 phenotype, enhancing chemotherapy sensitivity. Tissues from GBM patients show a significant correlation between reduced mTORC2 activity and increased astrogliosis, alongside a decrease in M2-polarized microgliosis, aligning with the NEO-MAP findings. Overall, the NEO-MAP is foreseen as a clinically significant tool for exploring tumor-glia interactions, opening avenues for drug development aimed at the tumor microenvironment.
Collapse
Affiliation(s)
- Yen N Diep
- Institute of Quantum Biophysics, Sungkyunkwan University, Suwon, 16419, Republic of Korea
- Department of Biophysics, Sungkyunkwan University, Suwon, 16419, Republic of Korea
- Department of Intelligent Precision Healthcare Convergence, Sungkyunkwan University, Suwon, 16419, Republic of Korea
| | - Hee Jung Park
- Department of Nuclear Medicine, Yonsei University College of Medicine, Seoul, 03722, Republic of Korea
| | - Xiaohui Zhu
- Institute of Quantum Biophysics, Sungkyunkwan University, Suwon, 16419, Republic of Korea
- Department of Biophysics, Sungkyunkwan University, Suwon, 16419, Republic of Korea
| | - Jin Ho Nam
- Department of Nuclear Medicine, Yonsei University College of Medicine, Seoul, 03722, Republic of Korea
| | - Jin-Kyoung Shim
- Department of Neurosurgery, Brain Tumor Center, Severance Hospital, Yonsei University College of Medicine, Seoul, 03722, Republic of Korea
- Brain Tumor Translational Research Laboratory, Severance Biomedical Research Institute, Yonsei University College of Medicine, Seoul, 03722, Republic of Korea
| | - Jong Hee Chang
- Department of Neurosurgery, Brain Tumor Center, Severance Hospital, Yonsei University College of Medicine, Seoul, 03722, Republic of Korea
| | - Dang Du Nguyen
- Institute of Quantum Biophysics, Sungkyunkwan University, Suwon, 16419, Republic of Korea
- Department of Biophysics, Sungkyunkwan University, Suwon, 16419, Republic of Korea
- Department of Intelligent Precision Healthcare Convergence, Sungkyunkwan University, Suwon, 16419, Republic of Korea
| | - Inki Kim
- Institute of Quantum Biophysics, Sungkyunkwan University, Suwon, 16419, Republic of Korea
- Department of Biophysics, Sungkyunkwan University, Suwon, 16419, Republic of Korea
- Department of Intelligent Precision Healthcare Convergence, Sungkyunkwan University, Suwon, 16419, Republic of Korea
| | - Dong-Gyu Jo
- Institute of Quantum Biophysics, Sungkyunkwan University, Suwon, 16419, Republic of Korea
- School of Pharmacy, Sungkyunkwan University, Suwon, 16419, Republic of Korea
- Biomedical Institute for Convergence, Sungkyunkwan University, Suwon, 16419, Republic of Korea
- Samsung Advanced Institute for Health Sciences and Technology, Sungkyunkwan University, Seoul, 06351, Republic of Korea
| | - Luke P Lee
- Institute of Quantum Biophysics, Sungkyunkwan University, Suwon, 16419, Republic of Korea
- Department of Biophysics, Sungkyunkwan University, Suwon, 16419, Republic of Korea
- Harvard Medical School, Division of Engineering in Medicine and Renal Division, Department of Medicine, Brigham and Women's Hospital, Boston, MA, 02115, USA
- Department of Bioengineering, Department of Electrical Engineering and Computer Science, University of California, Berkeley, CA, 94720, USA
| | - Mijin Yun
- Department of Nuclear Medicine, Yonsei University College of Medicine, Seoul, 03722, Republic of Korea
| | - Hansang Cho
- Institute of Quantum Biophysics, Sungkyunkwan University, Suwon, 16419, Republic of Korea
- Department of Biophysics, Sungkyunkwan University, Suwon, 16419, Republic of Korea
- Department of Intelligent Precision Healthcare Convergence, Sungkyunkwan University, Suwon, 16419, Republic of Korea
| |
Collapse
|
8
|
Fang X, Jia H, Pan S, Liu Q, Wang Q, Feng Y, Ding W, Luo T. Matrix Stiffness Regulates Interleukin-10 Secretion in Human Microglia (HMC3) via YAP-Mediated Mechanotransduction. Cell Biochem Funct 2025; 43:e70061. [PMID: 40011226 DOI: 10.1002/cbf.70061] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2024] [Revised: 01/28/2025] [Accepted: 02/14/2025] [Indexed: 02/28/2025]
Abstract
Microglia, as resident immune cells in the brain, adhere to the extracellular matrix and typically exhibit anti-inflammatory polarization under normal physiological conditions. Despite their pivotal roles, the regulatory effects of extracellular matrix properties on microglial function and the associated molecular mechanisms remain inadequately understood. Here, we elucidate how matrix stiffness modulates interleukin-10 (IL-10) secretion in human microglia (HMC3) via yes-associated protein (YAP)-mediated mechanotransduction. Using soft collagen Ⅰ-coated hydrogels, we observed a substantial reduction in IL-10 secretion, accompanied by a decrease in the expression and nuclear localization of YAP compared to cells adhered to glass substrates. With increasing hydrogel substrate stiffness, the expression and nuclear localization of YAP were enhanced, leading to an elevated secretion of IL-10. Subsequently, to further investigate the relationship between YAP and IL-10, we performed YAP depletion experiments, which revealed that nuclear exclusion of YAP suppressed IL-10 secretion. Interestingly, overexpression of YAP in microglia did not markedly affect IL-10 levels. We seeded YAP-knockdown microglia onto hydrogels of varying stiffness, and no significant differences were observed in IL-10 secretion. Our findings suggested that cytoskeletal polymerization was crucial for the regulation of IL-10 secretion mediated by YAP. Given the crucial role of IL-10 in the tumor microenvironment, we further found shYAP-microglia attenuated the pro-proliferative effect of microglia on gliomas. Besides, when YAP was silenced, actin of human microglia decreased, and their contractility was weakened. In summary, this study identifies YAP as a pivotal molecule in controlling cytokine secretion and sensing matrix stiffness in microglia. These insights offer potential therapeutic avenues for glioma treatment by targeting YAP-mediated pathways in microglial cells.
Collapse
Affiliation(s)
- Xue Fang
- CAS Key Laboratory of Mechanical Behavior and Design of Materials, Department of Modern Mechanics, University of Science and Technology of China, Hefei, China
| | - Haiying Jia
- Chinese People's Liberation Army Strategic Support Force Special Medical Center, Beijing, China
| | - Shaoshan Pan
- CAS Key Laboratory of Mechanical Behavior and Design of Materials, Department of Modern Mechanics, University of Science and Technology of China, Hefei, China
| | - Qian Liu
- CAS Key Laboratory of Mechanical Behavior and Design of Materials, Department of Modern Mechanics, University of Science and Technology of China, Hefei, China
| | - Qian Wang
- CAS Key Laboratory of Mechanical Behavior and Design of Materials, Department of Modern Mechanics, University of Science and Technology of China, Hefei, China
| | - Ye Feng
- CAS Key Laboratory of Mechanical Behavior and Design of Materials, Department of Modern Mechanics, University of Science and Technology of China, Hefei, China
| | - Weiping Ding
- School of Information Science and Technology, University of Science and Technology of China, Hefei, China
| | - Tianzhi Luo
- CAS Key Laboratory of Mechanical Behavior and Design of Materials, Department of Modern Mechanics, University of Science and Technology of China, Hefei, China
- The First Affiliated Hospital, University of Science and Technology of China, Hefei, China
| |
Collapse
|
9
|
Mo Y, Fan D, Wang W, Wang S, Yan Y, Zhao Z. Identification of inflammation-related genes signature to establish a prognostic model in MGMT unmethylated glioblastoma patients. Discov Oncol 2025; 16:154. [PMID: 39932605 DOI: 10.1007/s12672-025-01894-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/22/2024] [Accepted: 02/03/2025] [Indexed: 02/14/2025] Open
Abstract
BACKGROUND Patients with unmethylated O6-methylguanine-DNA methyltransferase promoter (uMGMT) glioblastoma (GBM) have a poor prognosis. Inflammatory response can affect the prognosis, for it may have a significant impact on the tumor microenvironment (TME). This study aims to identify a prognostic signature of inflammation-related genes, which can predict the prognosis of uMGMT GBM patients. METHODS We examined the gene expression, somatic mutations, and overall survival of 159 GBM patients with uMGMT using the TCGA and CGGA databases. We identified molecular subtypes of uMGMT GBM patients based on the expression of inflammation-related genes. Furthermore, we determined principal component analysis (PCA), gene ontology (GO) analysis, pathway analysis and immune infiltration analysis between high and low-inflammation subtypes. We also examined the spatial and longitudinal heterogeneity of these two subtypes. The LASSO-Cox analyses were used to develop an inflammation-related prognostic model. RESULTS Our findings indicate that patients with uMGMT GBM can be divided into high-inflammation and low-inflammation subtypes. Patients with high levels of inflammation are more likely to develop an immunosuppressive microenvironment, which stimulates the production of immunosuppressive cytokines, immune checkpoints, and immunosuppressive cells. Nine inflammation-related genes (EREG, BDKRB1, DCBLD2, CD14, AHR, CLEC5A, LTA, SLC4A4, and LY6E) were found to have excellent predictive potential for patient survival in the prognostic model. CONCLUSIONS In conclusion, we created a new prognostic model including 9 inflammation-related genes. This model has produced meaningful results in evaluating patient prognosis, which may help with future therapeutic strategies for patients with uMGMT GBM.
Collapse
Affiliation(s)
- Yunzhao Mo
- Department of Neurosurgery, General Hospital of Southern Theater Command, Guangzhou, 510010, China
| | - Dandan Fan
- Department of Neurosurgery, General Hospital of Southern Theater Command, Guangzhou, 510010, China
| | - Wei Wang
- Department of Pathology, General Hospital of Southern Theater Command, Guangzhou, 510010, China
| | - Shenchuan Wang
- Department of Neurosurgery, General Hospital of Southern Theater Command, Guangzhou, 510010, China
| | - Yingyu Yan
- Quality Management Department, The 921st Hospital of the Joint Logistics Support Force, Changsha, 410008, China
| | - Zhenyu Zhao
- Department of Neurosurgery, General Hospital of Southern Theater Command, Guangzhou, 510010, China.
| |
Collapse
|
10
|
Pancholi S, Shah R, Bose U, Yadav A, Murukan K, Pillai P. Hypoxic Secretome and Exosomes Derived From Human Glioblastoma Cells (U87MG) Promote Protumorigenic Phenotype of Microglia in Vitro. J Cell Biochem 2025; 126:e70002. [PMID: 39905831 DOI: 10.1002/jcb.70002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2024] [Revised: 01/08/2025] [Accepted: 01/25/2025] [Indexed: 02/06/2025]
Abstract
Glioblastoma multiforme (GBM), a highly heterogeneous CNS tumor known for its highest incidence rates and poor prognosis has shown limited success in the therapies due to hypoxia-driving immune-suppression in the tumor microenvironment (TME). Emerging evidence highlights the involvement of tumor cell-derived exosomes in tumor-associated microglia polarization via transfer of exosomal onco-proteins and miRNAs. Although the regulatory role of long noncoding RNAs (lncRNAs) in immune signaling are known, its mechanism in microglial polarization via exosomes in GBM still remains poorly understood. In our study, we found that in comparison to the normoxic GBM-derived exosomes lncRNA H19 was significantly upregulated in hypoxic GBM-derived exosomes. Hypoxic GBM-derived exosomes and secretome (conditioned media) caused the reduction in the % phagocytosis of microglia as compared with the control group. Moreover, GBM secretome caused increase in the M2-specific genes (IL10, STAT-3, CD163, CD206) in microglia indicating its polarization to the protumorigenic (M2) phenotype. LncRNA H19 knocked down GBM-secretome treatment in microglia further reduced the STAT-3 expression indicating H19 mediated signaling. Overall, our results suggest the involvement of hypoxic exosomes and lncRNA H19 in microglial polarization and H19 as a potential target.
Collapse
Affiliation(s)
- Sangati Pancholi
- Department of Zoology, Division of Neurobiology, Faculty of Science, The Maharaja Sayajirao University of Baroda, Vadodara, Gujarat, India
| | - Ritvi Shah
- Department of Zoology, Division of Neurobiology, Faculty of Science, The Maharaja Sayajirao University of Baroda, Vadodara, Gujarat, India
| | - Utsav Bose
- Department of Zoology, Division of Neurobiology, Faculty of Science, The Maharaja Sayajirao University of Baroda, Vadodara, Gujarat, India
| | - Ankit Yadav
- Department of Zoology, Division of Neurobiology, Faculty of Science, The Maharaja Sayajirao University of Baroda, Vadodara, Gujarat, India
| | - Karthik Murukan
- Department of Zoology, Division of Neurobiology, Faculty of Science, The Maharaja Sayajirao University of Baroda, Vadodara, Gujarat, India
| | - Prakash Pillai
- Department of Zoology, Division of Neurobiology, Faculty of Science, The Maharaja Sayajirao University of Baroda, Vadodara, Gujarat, India
| |
Collapse
|
11
|
Zannikou MZ, Duffy JT, Procissi D, Najem H, Levine RN, Hambardzumyan D, Lee-Chang C, Leoni L, Zhang B, Heimberger AB, Miska J, Balyasnikova IV. A Bi-Specific T Cell-Engaging Antibody Shows Potent Activity, Specificity, and Tumor Microenvironment Remodeling in Experimental Syngeneic and Genetically Engineered Models of GBM. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2024.12.18.628714. [PMID: 39763755 PMCID: PMC11702671 DOI: 10.1101/2024.12.18.628714] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/01/2025]
Abstract
BACKGROUND Bispecific T cell-engagers (BTEs) are engineered antibodies that redirect T cells to target antigen-expressing tumors. BTEs targeting tumor-specific antigens such as interleukin 13 receptor alpha 2 (IL13Rα2) and EGFRvIII have been developed for glioblastoma (GBM). However, there is limited mechanistic understanding of the action of BTE since prior studies were mostly conducted in immunocompromised animal models. To close this gap, the function of BTEs was assessed in the immunosuppressive glioma microenvironment (TME) of orthotopic and genetically engineered mouse models (GEMM) with intact immune systems. METHODS A BTE that bridges CD3 epsilon on murine T cells to IL13Rα2-positive GBM cells was developed and the therapeutic mechanism investigated in immunocompetent mouse models of GBM. Multi-color flow cytometry, single-cell RNA sequencing (scRNA-Seq), multiplex immunofluorescence, and multiparametric magnetic resonance imaging (MRI) across multiple pre-clinical models of GBM were used to evaluate the mechanism and action and response. RESULTS BTE-mediated interactions between murine T cells and GBM cells triggered T cell activation and antigen-dependent killing of GBM cells. BTE treatment significantly extended the survival of mice bearing IL13Rα2-expressing orthotopic glioma and de novo forming GBM in the GEMM. Quantified parametric MR imaging validated the survival data showing a reduction in glioma volume and decreased glioma viability. Flow cytometric and scRNA-seq analyses of the TME revealed robust increases in activated and memory T cells and decreases in immunosuppressive myeloid cells in the brains of mice following BTE treatment. CONCLUSIONS Our data demonstrate that the survival benefits of BTEs in preclinical models of glioma are due to the ability to engage the host immune system in direct killing, induction of immunological memory, and modulation of the TME. These findings provide a deeper insight into the mechanism of BTE actions in GBM.
Collapse
|
12
|
Yang L, Li S, Yu L, Leng J, Li N. Targeting glycolysis: exploring a new frontier in glioblastoma therapy. Front Immunol 2025; 15:1522392. [PMID: 39877360 PMCID: PMC11772265 DOI: 10.3389/fimmu.2024.1522392] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2024] [Accepted: 12/23/2024] [Indexed: 01/31/2025] Open
Abstract
Glioblastoma(GBM) is a highly malignant primary central nervous system tumor that poses a significant threat to patient survival due to its treatment resistance and rapid recurrence.Current treatment options, including maximal safe surgical resection, radiotherapy, and temozolomide (TMZ) chemotherapy, have limited efficacy.In recent years, the role of glycolytic metabolic reprogramming in GBM has garnered increasing attention. This review delves into the pivotal role of glycolytic metabolic reprogramming in GBM, with a particular focus on the multifaceted roles of lactate, a key metabolic product, within the tumor microenvironment (TME). Lactate has been implicated in promoting tumor cell proliferation, invasion, and immune evasion. Additionally, this review systematically analyzes potential therapeutic strategies targeting key molecules within the glycolytic pathway, such as Glucose Transporters (GLUTs), Monocarboxylate Transporters(MCTs), Hexokinase 2 (HK2), 6-Phosphofructo-2-Kinase/Fructose-2,6-Biphosphatase 3 (PFKFB3), Pyruvate Kinase Isozyme Type M2 (PKM2), and the Lactate Dehydrogenase A (LDHA). These studies provide a novel perspective for GBM treatment. Despite progress made in existing research, challenges remain, including drug penetration across the blood-brain barrier, side effects, and resistance. Future research will aim to address these challenges by improving drug delivery, minimizing side effects, and exploring combination therapies with radiotherapy, chemotherapy, and immunotherapy to develop more precise and effective personalized treatment strategies for GBM.
Collapse
Affiliation(s)
| | | | | | | | - Na Li
- Department of Oncology, Suining Central Hospital, Suining, Sichuan, China
| |
Collapse
|
13
|
Shojaee P, Weinholtz E, Schaadt NS, Feuerhake F, Hatzikirou H. Biopsy location and tumor-associated macrophages in predicting malignant glioma recurrence using an in-silico model. NPJ Syst Biol Appl 2025; 11:3. [PMID: 39779740 PMCID: PMC11711667 DOI: 10.1038/s41540-024-00478-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2024] [Accepted: 12/15/2024] [Indexed: 01/11/2025] Open
Abstract
Predicting the biological behavior and time to recurrence (TTR) of high-grade diffuse gliomas (HGG) after maximum safe neurosurgical resection and combined radiation and chemotherapy plays a pivotal role in planning clinical follow-up, selecting potentially necessary second-line treatment and improving the quality of life for patients diagnosed with a malignant brain tumor. The current standard-of-care (SoC) for HGG includes follow-up neuroradiological imaging to detect recurrence as early as possible and relies on several clinical, neuropathological, and radiological prognostic factors, which have limited accuracy in predicting TTR. In this study, using an in-silico analysis, we aim to improve predictive power for TTR by considering the role of (i) prognostically relevant information available through diagnostics used in the current SoC, (ii) advanced image-based information not currently part of the standard diagnostic workup, such as tumor-normal tissue interface (edge) features and quantitative data specific to biopsy positions within the tumor, and (iii) information on tumor-associated macrophages. In particular, we introduced a state-of-the-art spatio-temporal model of tumor-immune interactions, emphasizing the interplay between macrophages and glioma cells. This model serves as a synthetic reality for assessing the predictive value of various features. We generated a cohort of virtual patients based on our mathematical model. Each patient's dataset includes simulated T1Gd and Fluid-attenuated inversion recovery (FLAIR) MRI volumes. T1-weighted imaging highlights anatomical structures with high contrast, providing clear detail on brain morphology, whereas FLAIR suppresses fluid signals, improving the visualization of lesions near fluid-filled spaces, which is particularly helpful for identifying peritumoral edema. Additionally, we simulated results on macrophage density and proliferative activity, either in a specified part of the tumor, namely the tumor core or edge ("localized"), or unspecified ("non-localized"). To enhance the realism of our synthetic data, we imposed different levels of noise. Our findings reveal that macrophage density at the tumor edge contributed to a high predictive value of feature importance for the selected regression model. Moreover, there are lower MSE values for the "localized" biopsy in prediction accuracy toward recurrence post-resection compared with "non-localized" specimens in the noisy data. In conclusion, the results show that localized biopsies provided more information about tumor behavior, especially at the interface of tumor and normal tissue (Edge).
Collapse
Affiliation(s)
- Pejman Shojaee
- Center for Interdisciplinary Digital Sciences (CIDS), Department Information Services and High-Performance Computing (ZIH), Dresden University of Technology, 01062, Dresden, Germany
| | - Edwin Weinholtz
- Center for Interdisciplinary Digital Sciences (CIDS), Department Information Services and High-Performance Computing (ZIH), Dresden University of Technology, 01062, Dresden, Germany
| | - Nadine S Schaadt
- Department of Neuropathology, Institute for Pathology, Hannover Medical School, Hannover, Germany
| | - Friedrich Feuerhake
- Department of Neuropathology, Institute for Pathology, Hannover Medical School, Hannover, Germany
- Institute for Neuropathology, University Clinic Freiburg, Freiburg, Germany
| | - Haralampos Hatzikirou
- Center for Interdisciplinary Digital Sciences (CIDS), Department Information Services and High-Performance Computing (ZIH), Dresden University of Technology, 01062, Dresden, Germany.
- Mathematics Department, Khalifa University, Abu Dhabi, UAE.
| |
Collapse
|
14
|
Ćwiklińska A, Przewodowska D, Koziorowski D, Szlufik S. Innovative Approaches to Brain Cancer: The Use of Magnetic Resonance-guided Focused Ultrasound in Glioma Therapy. Cancers (Basel) 2024; 16:4235. [PMID: 39766134 PMCID: PMC11674718 DOI: 10.3390/cancers16244235] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2024] [Revised: 12/15/2024] [Accepted: 12/17/2024] [Indexed: 01/11/2025] Open
Abstract
Gliomas are a wide group of common brain tumors, with the most aggressive type being glioblastoma multiforme (GBM), with a 5-year survival rate of less than 5% and a median survival time of approximately 12-14 months. The standard treatment of GBM includes surgical excision, radiotherapy, and chemotherapy with temozolomide (TMZ). However, tumor recurrence and progression are common. Therefore, more effective treatment for GBM should be found. One of the main obstacles to the treatment of GBM and other gliomas is the blood-brain barrier (BBB), which impedes the penetration of antitumor chemotherapeutic agents into glioblastoma cells. Nowadays, one of the most promising novel methods for glioma treatment is Magnetic Resonance-guided Focused Ultrasound (MRgFUS). Low-intensity FUS causes the BBB to open transiently, which allows better drug delivery to the brain tissue. Under magnetic resonance guidance, ultrasound waves can be precisely directed to the tumor area to prevent side effects in healthy tissues. Through the open BBB, we can deliver targeted chemotherapeutics, anti-tumor agents, immunotherapy, and gene therapy directly to gliomas. Other strategies for MRgFUS include radiosensitization, sonodynamic therapy, histotripsy, and thermal ablation. FUS can also be used to monitor the treatment and progression of gliomas using blood-based liquid biopsy. All these methods are still under preclinical or clinical trials and are described in this review to summarize current knowledge and ongoing trials.
Collapse
Affiliation(s)
| | | | | | - Stanisław Szlufik
- Department of Neurology, Faculty of Health Sciences, Medical University of Warsaw, 03-242 Warsaw, Poland
| |
Collapse
|
15
|
Zhang Y, Kang T, Wang Y, Song C, Li H, Mi H, Li Y, Dong M, Ma X, Zhu H, Cheng L, Zhang P, Chen Z, Zhou L, Wu Q, Mao F, Wang B, Zhang S, Shu K, Wan F, Zhou W, Rich JN, Shen J, Xiao Q, Yu X. A low level of tumor necrosis factor α in tumor microenvironment maintains the self-renewal of glioma stem cells by Vasorin-mediated glycolysis. Neuro Oncol 2024; 26:2256-2271. [PMID: 39093693 PMCID: PMC11630517 DOI: 10.1093/neuonc/noae147] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2023] [Indexed: 08/04/2024] Open
Abstract
BACKGROUND Self-renewal of glioma stem cells (GSCs) is responsible for glioblastoma (GBM) therapy resistance and recurrence. Tumor necrosis factor α (TNFα) and TNF signaling pathway display an antitumor activity in preclinical models and in tumor patients. However, TNFα exhibits no significance for glioma clinical prognosis based on the Glioma Genome Atlas database. This study aimed to explore whether TNFα of tumor microenvironment maintains self-renewal of GSCs and promotes worse prognosis in glioma patients. METHODS Spatial transcriptomics, immunoblotting, sphere formation assay, extreme limiting dilution, and gene expression analysis were used to determine the role of TNFα on GSC's self-renewal. Mass spectrometry, RNA-sequencing detection, bioinformatic analyses, qRT-RNA, immunofluorescence, immunohistochemistry, single-cell RNA sequencing, in vitro and in vivo models were used to uncover the mechanism of TNFα-induced GSC self-renewal. RESULTS A low level of TNFα displays a promoting effect on GSC self-renewal and worse glioma prognosis. Mechanistically, Vasorin (VASN) mediated TNFα-induced self-renewal by potentiating glycolysis. Lactate produced by glycolysis inhibits the TNFα secretion of tumor-associated macrophages (TAMs) and maintains TNFα at a low level. CONCLUSIONS TNFα-induced GSC self-renewal mediated by VASN provides a possible explanation for the failures of endogenous TNFα effect on GBM. A combination of targeting VASN and TNFα antitumor effect may be an effective approach for treating GBM.
Collapse
Affiliation(s)
- Yang Zhang
- Department of Histology and Embryology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Tianxu Kang
- School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yuxi Wang
- School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Chao Song
- Department of Histology and Embryology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Huan Li
- Department of Histology and Embryology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Hailong Mi
- Department of Histology and Embryology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yachao Li
- Department of Histology and Embryology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Minhai Dong
- Department of Neurosurgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xiaoyu Ma
- Department of Neurosurgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Hongtao Zhu
- Department of Neurosurgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Lidong Cheng
- Department of Neurosurgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Po Zhang
- Department of Medicine, UPMC Hillman Cancer Center, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania, USA
| | - Zhiye Chen
- Department of Neurosurgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Lin Zhou
- Department of Histology and Embryology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Qiulian Wu
- Department of Medicine, UPMC Hillman Cancer Center, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania, USA
| | - Feng Mao
- Department of Neurosurgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Baofeng Wang
- Department of Neurosurgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Suojun Zhang
- Department of Neurosurgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Kai Shu
- Department of Neurosurgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Feng Wan
- Department of Neurosurgery, Guangdong Provincial People’s Hospital, Guangdong Academy of Medical Sciences, Southern Medical University, Guangzhou, China
| | - Wenchao Zhou
- Division of Life Sciences and Medicine, Intelligent Pathology Institute, The First Affiliated Hospital of USTC, University of Science and Technology of China, Hefei, China
| | - Jeremy N Rich
- Department of Neurology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
- Department of Medicine, UPMC Hillman Cancer Center, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania, USA
| | - Jianying Shen
- Department of Histology and Embryology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Qungen Xiao
- Department of Neurosurgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xingjiang Yu
- Hubei Key Laboratory of Drug Target Research and Pharmacodynamic Evaluation, Wuhan, China
- Department of Histology and Embryology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
16
|
Cómitre-Mariano B, Vellila-Alonso G, Segura-Collar B, Mondéjar-Ruescas L, Sepulveda JM, Gargini R. Sentinels of neuroinflammation: the crucial role of myeloid cells in the pathogenesis of gliomas and neurodegenerative diseases. J Neuroinflammation 2024; 21:304. [PMID: 39578808 PMCID: PMC11583668 DOI: 10.1186/s12974-024-03298-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2024] [Accepted: 11/13/2024] [Indexed: 11/24/2024] Open
Abstract
The inflammatory processes that drive pathologies of the central nervous system (CNS) are complex and involve significant contributions from the immune system, particularly myeloid cells. Understanding the shared and distinct pathways of myeloid cell regulation in different CNS diseases may offer critical insights into therapeutic development. This review aims to elucidate the mechanisms underlying myeloid cell dysfunction and neuroinflammation in two groups of neurological pathologies with significant social impact and a limited efficacy of their treatments: the most common primary brain tumors -gliomas-, and the most prevalent neurodegenerative disorders -Alzheimer's and Parkinson's disease. Despite their distinct clinical manifestations, these diseases share key pathological features, including chronic inflammation and immune dysregulation. The role of myeloid cells in neuroinflammation has garnered special interest in recent years in both groups, as evidenced by the growing focus on therapeutic research centred on myeloid cells. By examining the cellular and molecular dynamics that govern these conditions, we hope to identify common and unique therapeutic targets that can inform the development of more effective treatments. Recent advances in single-cell technologies have revolutionized our understanding of myeloid cell heterogeneity, revealing diverse phenotypes and molecular profiles across different disease stages and microenvironments. Here, we present a comprehensive analysis of myeloid cell involvement in gliomas, Alzheimer's and Parkinson's disease, with a focus on phenotypic acquisition, molecular alterations, and therapeutic strategies targeting myeloid cells. This integrated approach not only addresses the limitations of current treatments but also suggests new avenues for therapeutic intervention, aimed at modulating the immune landscape to improve patient outcomes.
Collapse
Affiliation(s)
- Blanca Cómitre-Mariano
- Instituto de Investigación Biomédicas I+12, Hospital Universitario 12 de Octubre, Madrid, 28041, Spain
- Pathology and Neurooncology Unit, Hospital Universitario 12 de Octubre, Av. de Córdoba, S/N, Madrid, 28041, Spain
| | - Gabriel Vellila-Alonso
- Pathology and Neurooncology Unit, Hospital Universitario 12 de Octubre, Av. de Córdoba, S/N, Madrid, 28041, Spain
- Medical Oncology, Hospital Universitario 12 de Octubre, Madrid, 28041, Spain
| | - Berta Segura-Collar
- Instituto de Investigación Biomédicas I+12, Hospital Universitario 12 de Octubre, Madrid, 28041, Spain
- Pathology and Neurooncology Unit, Hospital Universitario 12 de Octubre, Av. de Córdoba, S/N, Madrid, 28041, Spain
| | - Lucía Mondéjar-Ruescas
- Instituto de Investigación Biomédicas I+12, Hospital Universitario 12 de Octubre, Madrid, 28041, Spain
- Pathology and Neurooncology Unit, Hospital Universitario 12 de Octubre, Av. de Córdoba, S/N, Madrid, 28041, Spain
| | - Juan M Sepulveda
- Instituto de Investigación Biomédicas I+12, Hospital Universitario 12 de Octubre, Madrid, 28041, Spain.
- Medical Oncology, Hospital Universitario 12 de Octubre, Madrid, 28041, Spain.
| | - Ricardo Gargini
- Instituto de Investigación Biomédicas I+12, Hospital Universitario 12 de Octubre, Madrid, 28041, Spain.
- Pathology and Neurooncology Unit, Hospital Universitario 12 de Octubre, Av. de Córdoba, S/N, Madrid, 28041, Spain.
| |
Collapse
|
17
|
Chang CW, Bale A, Bhargava R, Harley BAC. Glioblastoma Drives Protease-Independent Extracellular Matrix Invasion of Microglia. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.11.08.622715. [PMID: 39605524 PMCID: PMC11601220 DOI: 10.1101/2024.11.08.622715] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/29/2024]
Abstract
Glioblastoma (GBM) is the most common and lethal form of primary brain cancer. Microglia infiltration into the tumor microenvironment is associated with immunosuppression and poor prognosis. Improved physicochemical understanding of microglia activation and invasion may provide novel GBM therapeutic strategies essential for improving long-term treatment efficacy. Here, we combine microfluidic systems with 3-D collagen hydrogels to systematically investigate microglia activation, invasion, contractility and cytokine secretion in response of GBM-microglia crosstalk. GBM inflammatory biomolecules significantly promote activation and 3D invasion of microglia. Interestingly, microglia invasion is not significantly affected by inhibitors of MMP activity or cellular glycolysis. In contrast, ROCK-pathway inhibition significantly impedes microglia invasion. Infrared microscopy analyses show that GBM co-culture does not significantly alter microglia lipid content. Further, GBM conditioned media resulted in significantly increased collagen hydrogel contraction, suggesting the importance of microglia contractility to physically remodel the local extracellular matrix (ECM). We also identify a panel of soluble proteins that may contribute to microglia chemotaxis, such as TIMP-1 and CXCL12. Taken together, this study suggests that the presence of GBM cells can enhance microglia invasion via increased cellular contractility, independent of MMP activity and cellular glycolysis.
Collapse
|
18
|
Bertola L, Pellizzoni B, Giudice C, Grieco V, Ferrari R, Chiti LE, Stefanello D, Manfredi M, De Zani D, Recordati C. Tumor-associated macrophages and tumor-infiltrating lymphocytes in canine cutaneous and subcutaneous mast cell tumors. Vet Pathol 2024; 61:882-895. [PMID: 38647163 DOI: 10.1177/03009858241244851] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/25/2024]
Abstract
Cutaneous and subcutaneous mast cell tumors (MCTs) are common canine neoplasms characterized by variable biological behavior. Tumor-associated macrophages (TAMs) and tumor-infiltrating lymphocytes (TILs) can be effective prognostic markers in numerous human neoplasms and are increasingly investigated in dogs. The aim of this study was to characterize immune cells in canine MCTs and their relationship with histological location (cutaneous, subcutaneous) and histologic nodal metastatic status (HN0-3). Thirty-eight MCTs (26 cutaneous, 12 subcutaneous) from 33 dogs with known sentinel lymph node (SLN) metastatic status were immunolabeled for Iba1 (macrophages), CD20 (B cells), CD3 (T cells), and Foxp3 (regulatory T cells). Semiquantitative scoring of interstitial and perivascular CD3+, CD20+, and Foxp3+ cells and morphological evaluation of Iba1+ cells were performed. For each marker, the percent immunopositive area was evaluated by image analysis. All MCTs were diffusely infiltrated by Iba1+ cells and variably infiltrated by CD20+, CD3+, and rare Foxp3+ cells. Stellate/spindle Iba1+ cells were associated with HN2 and HN3 SLNs. Perivascular Foxp3+ cells, CD3+ cells, and percent CD3+ areas were increased in subcutaneous MCTs. Interstitial CD3+ cells were increased in cutaneous MCTs with HN0 SLNs. No differences in CD20+ cells were identified between cutaneous and subcutaneous MCTs and among SLN classes. MCTs were markedly infiltrated by TAMs and variably infiltrated by TILs. Stellate/spindle morphology of TAMs associated with HN2 and HN3 SLNs is suggestive of a pro-tumoral (M2) phenotype. Cutaneous and subcutaneous MCTs have different tumor-immune microenvironments, and T-cell infiltration might contribute to prevention of nodal metastatic spread of cutaneous MCTs.
Collapse
Affiliation(s)
- Luca Bertola
- University of Milan, Lodi, Italy
- Fondazione UNIMI, Milano, Italy
| | | | | | | | | | | | | | | | | | | |
Collapse
|
19
|
Han X, Zhou H, Ge X, Hou L, Li H, Zhang D, Wang Y, Xue X. Identification of an Immune signature assisted prognosis, and immunotherapy prediction for IDH wildtype glioblastoma. J Cancer 2024; 15:6452-6467. [PMID: 39513108 PMCID: PMC11540507 DOI: 10.7150/jca.100144] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2024] [Accepted: 10/01/2024] [Indexed: 11/15/2024] Open
Abstract
IDH-wildtype glioblastoma (GBM) is the most common and malignant primary brain tumor. The purpose of this study is to establish a prognostic gene signature for IDH-wildtype GBM. RNA sequencing data of normal brain tissue and GBM patients were obtained from TCGA, CGGA, GEO and the GTEx databases. Identification of prognostic differentially expressed genes (DEGs) with | log2 fold change | > 0.5 and adjust p < 0.05 in TCGA and CGGA databases by "limma" method. By LASSO regression analysis and multivariate Cox analysis, a 3-gene prognostic signature composed of FMOD, MXRA5 and RAB36 was established. The 3-gene prognostic risk model is validated by TCGA and GSE43378 datasets. The expression of FMOD, MXRA5 and RAB36 in GBM patients was significantly higher than that in normal brain tissues in CCGA, TCGA and GSE29796 data sets. In order to further verify this result, total RNA was extracted from tumors and paracancerous tissues of 9 GBM patients. RT-PCR results showed that the expression of FMOD, MXRA5 and RAB36 in tumor tissues of most patients was higher than that in paracancerous tissues. The results of GSEA showed that the pathway enrichment of the 3-gene signature was mainly related to tumor immunity. Immune cell infiltration analyzed by ssGSEA showed that there were significant differences in macrophages between high- and low-risk groups. Immune checkpoint genes correlation analysis showed that PD-L1 gene expression is closely related to risk score. Our study identifies a prognostic-associated risk model and provides a potential effective immunotherapy target for IDH-wildtype GBM patients.
Collapse
Affiliation(s)
- Xuetao Han
- Department of Radiotherapy, The Second Hospital of Hebei Medical University, Shijiazhuang 050000, Hebei Province, China
| | - Huandi Zhou
- Department of Radiotherapy, The Second Hospital of Hebei Medical University, Shijiazhuang 050000, Hebei Province, China
- Department of Central Laboratory, The Second Hospital of Hebei Medical University, Shijiazhuang 050000, Hebei Province, China
| | - Xiaohui Ge
- Department of Radiotherapy, The Second Hospital of Hebei Medical University, Shijiazhuang 050000, Hebei Province, China
| | - Liubing Hou
- Department of Radiotherapy, The Second Hospital of Hebei Medical University, Shijiazhuang 050000, Hebei Province, China
- Department of Central Laboratory, The Second Hospital of Hebei Medical University, Shijiazhuang 050000, Hebei Province, China
| | - Haonan Li
- Department of Radiotherapy, The Second Hospital of Hebei Medical University, Shijiazhuang 050000, Hebei Province, China
| | - Dongdong Zhang
- Department of Radiotherapy, The Second Hospital of Hebei Medical University, Shijiazhuang 050000, Hebei Province, China
| | - Yu Wang
- Department of Radiotherapy, The Second Hospital of Hebei Medical University, Shijiazhuang 050000, Hebei Province, China
| | - Xiaoying Xue
- Department of Radiotherapy, The Second Hospital of Hebei Medical University, Shijiazhuang 050000, Hebei Province, China
| |
Collapse
|
20
|
Ansari A, Bhattacharyya T, Das P, Chandra Y, Kundu TK, Banerjee R. Lipid-Conjugated Reduced Haloperidol in Association with Glucose-Based Nanospheres: A Strategy for Glioma Treatment. Mol Pharm 2024; 21:5053-5070. [PMID: 39302161 DOI: 10.1021/acs.molpharmaceut.4c00468] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/22/2024]
Abstract
Aggressive glioma exhibits a poor survival rate. Increased tumor aggression is linked to both tumor cells and tumor-associated macrophages (TAMs), which induce pro-aggression, invasion, and metastasis. Imperatively, for effective treatment, it is important to target both glioma cells and TAMs. Haloperidol, a neuropsychotic drug, avidly targets the sigma receptor (SR), which is expressed in higher levels in both the cell types. Herein, we present the development of a novel cationic lipid-conjugated reduced haloperidol (±RHPC8), which aims to mediate the SR-targeted antiglioma effect. Hypothetically, ±RHPC8 would act simultaneously as an SR-targeting ligand and anticancer agent. As the blood-brain barrier (BBB) obstructs direct targeting of in situ glioma, we used BBB-crossing glucose-based carbon nanospheres (CSPs) to deliver ±RHPC8 within the glioma tumor-bearing mouse brain. The resultant ±RHPC8-CSP nanoconjugate targeted SR-expressing glioma cells. In both orthotopic and subcutaneous mouse tumor models, ±RHPC8-CSP prolonged survival and regressed tumors compared to other treated groups. Notably, ±RHPC8-CSP was significantly taken up by SR-expressing TAMs thus resulting in macrophage polarization from M2 to M1, as exhibited by markedly reduced expression of immunosuppressive cytokines released by TAMs, including TGF-β, IL-10, and VEGF. In conclusion, the designed ±RHPC8-CSP nanoconjugate presented an effective nanodrug delivery system for brain cancer treatment.
Collapse
Affiliation(s)
- Aasia Ansari
- Department of Oils, Lipid, Science & Technology, CSIR-Indian Institute of Chemical Technology, Hyderabad 500 007, India
- Academy of Scientific and Innovative Research (AcSIR), CSIR-HRDC Campus, Ghaziabad 201002, India
| | - Tithi Bhattacharyya
- Department of Oils, Lipid, Science & Technology, CSIR-Indian Institute of Chemical Technology, Hyderabad 500 007, India
- Academy of Scientific and Innovative Research (AcSIR), CSIR-HRDC Campus, Ghaziabad 201002, India
| | - Pritam Das
- Department of Oils, Lipid, Science & Technology, CSIR-Indian Institute of Chemical Technology, Hyderabad 500 007, India
- Academy of Scientific and Innovative Research (AcSIR), CSIR-HRDC Campus, Ghaziabad 201002, India
| | - Yogesh Chandra
- Department of Applied Biology, CSIR-Indian Institute of Chemical Technology, Hyderabad 500 007, India
| | - Tapas K Kundu
- Transcription and Disease Laboratory, Molecular Biology and Genetics Unit, Jawaharlal Nehru Centre for Advanced Scientific Research, Jakkur P.O., Bangalore 560 064, India
| | - Rajkumar Banerjee
- Department of Oils, Lipid, Science & Technology, CSIR-Indian Institute of Chemical Technology, Hyderabad 500 007, India
- Academy of Scientific and Innovative Research (AcSIR), CSIR-HRDC Campus, Ghaziabad 201002, India
| |
Collapse
|
21
|
Niu X, Zhang Y, Wang Y. Co-culture models for investigating cellular crosstalk in the glioma microenvironment. CANCER PATHOGENESIS AND THERAPY 2024; 2:219-230. [PMID: 39371093 PMCID: PMC11447344 DOI: 10.1016/j.cpt.2023.11.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/27/2023] [Revised: 10/30/2023] [Accepted: 11/03/2023] [Indexed: 10/08/2024]
Abstract
Glioma is the most prevalent primary malignant tumor in the central nervous system (CNS). It represents a diverse group of brain malignancies characterized by the presence of various cancer cell types as well as an array of noncancerous cells, which together form the intricate glioma tumor microenvironment (TME). Understanding the interactions between glioma cells/glioma stem cells (GSCs) and these noncancerous cells is crucial for exploring the pathogenesis and development of glioma. To invesigate these interactions requires in vitro co-culture models that closely mirror the actual TME in vivo. In this review, we summarize the two- and three-dimensional in vitro co-culture model systems for glioma-TME interactions currently available. Furthermore, we explore common glioma-TME cell interactions based on these models, including interactions of glioma cells/GSCs with endothelial cells/pericytes, microglia/macrophages, T cells, astrocytes, neurons, or other multi-cellular interactions. Together, this review provides an update on the glioma-TME interactions, offering insights into glioma pathogenesis.
Collapse
Affiliation(s)
- Xiaodong Niu
- Department of Neurosurgery, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Yan Zhang
- National Clinical Research Center for Geriatrics, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Yuan Wang
- Department of Neurosurgery, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| |
Collapse
|
22
|
Orozco-Mera J, Montoya-Gómez A, Lopes DS, Jiménez-Charris E. Snake venom bioprospecting as an approach to finding potential anti-glioblastoma molecules. J Venom Anim Toxins Incl Trop Dis 2024; 30:e20240015. [PMID: 39285908 PMCID: PMC11404105 DOI: 10.1590/1678-9199-jvatitd-2024-0015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2024] [Accepted: 07/30/2024] [Indexed: 09/19/2024] Open
Abstract
Glioblastoma (GB) is the most common type of malignant tumor of the central nervous system, responsible for significant morbidity and with a 5-year overall relative survival of only 6.8%. Without advances in treatment in the last twenty years, the standard of care continues to be maximum safe resection, Temozolomide (TMZ), and radiotherapy. Many new trials are ongoing, and despite showing increased progression-free survival, these trials did not improve overall survival. They did not consider the adverse effects of these therapies. Therefore, an increasing number of bioprospecting studies have used snake venom molecules to search for new strategies to attack GB selectively without producing side effects. The present review aims to describe GB characteristics and current and new approaches for treatment considering their side effects. Besides, we focused on the antitumoral activity of snake venom proteins from the Viperidae family against GB, exploring the potential for drug design based on in vitro and in vivo studies. This review followed the Preferred Reporting Items for Systematic Reviews and Meta-Analysis (PRISMA) guidelines. In January 2024, a systematic search was performed in the PubMed, EMBASE, and Web of Science databases from January 2000 to December 2023. Search terms were selected based on the population/exposure/outcome (PEO) framework and combined using Boolean operators ("AND", "OR"). The search strategy used these terms: glioblastoma, glioma, high-grade glioma, WHO IV glioma, brain cancer, snake venom, Viperidae, and bioprospection. We identified 10 in vivo and in vitro studies with whole and isolated proteins from Viperidae venom that could have antitumor activity against glioblastoma. Studies in bioprospecting exploring the advantage of snake venom proteins against GB deserve to be investigated due to their high specificity, small size, inherent bioactivity, and few side effects to cross the blood-brain barrier (BBB) to reach the tumor microenvironment.
Collapse
Affiliation(s)
- Javier Orozco-Mera
- Grupo de Nutrición, Facultad de Salud, Universidad del Valle, Cali, Colombia
- Department of Neurosurgery, Clínica Imbanaco, Cali, Colombia
| | | | - Daiana Silva Lopes
- Multidisciplinary Institute in Health, Federal University of Bahia (UFBA), Vitória da Conquista, BA, Brazil
| | | |
Collapse
|
23
|
Wang X, Zhang H, XinZhang, Liu Y. Abscopal effect: from a rare phenomenon to a new frontier in cancer therapy. Biomark Res 2024; 12:98. [PMID: 39228005 PMCID: PMC11373306 DOI: 10.1186/s40364-024-00628-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2024] [Accepted: 07/30/2024] [Indexed: 09/05/2024] Open
Abstract
Radiotherapy (RT) controls local lesions, meantime it has the capability to induce systemic response to inhibit distant, metastatic, non-radiated tumors, which is referred to as the "abscopal effect". It is widely recognized that radiotherapy can stimulate systemic immune response. This provides a compelling theoretical basis for the combination of immune therapy combined with radiotherapy(iRT). Indeed, this phenomenon has also been observed in clinical treatment, bringing significant clinical benefits to patients, and a series of basic studies are underway to amplify this effect. However, the molecular mechanisms of immune response induced by RT, determination of the optimal treatment regimen for iRT, and how to amplify the abscopal effect. In order to amplify and utilize this effect in clinical management, these key issues require to be well addressed; In this review, we comprehensively summarize the growing consensus and emphasize the emerging limitations of enhancing the abscopal effect with radiotherapy or immunotherapy. Finally, we discuss the prospects and barriers to the current clinical translational applications.
Collapse
Affiliation(s)
- Xueying Wang
- Department of Otolaryngology Head and Neck Surgery, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, 410008, Hunan, People's Republic of China
- Otolaryngology Major Disease Research Key Laboratory of Hunan Province, 87 Xiangya Road, Changsha, 410008, Hunan, People's Republic of China
- Clinical Research Center for Laryngopharyngeal and Voice Disorders in Hunan Province, 87 Xiangya Road, Changsha, 410008, Hunan, People's Republic of China
- National Clinical Research Center for Geriatric Disorders (Xiangya Hospital), Changsha, 410008, Hunan, China
| | - Haoyu Zhang
- Department of Otolaryngology Head and Neck Surgery, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, 410008, Hunan, People's Republic of China
- Otolaryngology Major Disease Research Key Laboratory of Hunan Province, 87 Xiangya Road, Changsha, 410008, Hunan, People's Republic of China
- Clinical Research Center for Laryngopharyngeal and Voice Disorders in Hunan Province, 87 Xiangya Road, Changsha, 410008, Hunan, People's Republic of China
- National Clinical Research Center for Geriatric Disorders (Xiangya Hospital), Changsha, 410008, Hunan, China
| | - XinZhang
- Department of Otolaryngology Head and Neck Surgery, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, 410008, Hunan, People's Republic of China.
- Otolaryngology Major Disease Research Key Laboratory of Hunan Province, 87 Xiangya Road, Changsha, 410008, Hunan, People's Republic of China.
- Clinical Research Center for Laryngopharyngeal and Voice Disorders in Hunan Province, 87 Xiangya Road, Changsha, 410008, Hunan, People's Republic of China.
- National Clinical Research Center for Geriatric Disorders (Xiangya Hospital), Changsha, 410008, Hunan, China.
| | - Yong Liu
- Department of Otolaryngology Head and Neck Surgery, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, 410008, Hunan, People's Republic of China.
- Otolaryngology Major Disease Research Key Laboratory of Hunan Province, 87 Xiangya Road, Changsha, 410008, Hunan, People's Republic of China.
- Clinical Research Center for Laryngopharyngeal and Voice Disorders in Hunan Province, 87 Xiangya Road, Changsha, 410008, Hunan, People's Republic of China.
- National Clinical Research Center for Geriatric Disorders (Xiangya Hospital), Changsha, 410008, Hunan, China.
| |
Collapse
|
24
|
Gu J, Tong W, Wang X, Gu L, Wang W, Zang T, Lou M, Liu Y. Multi-omics Analysis Revealed that the CCN Family Regulates Cell Crosstalk, Extracellular Matrix, and Immune Escape, Leading to a Poor Prognosis of Glioma. Cell Biochem Biophys 2024; 82:2157-2170. [PMID: 38837011 DOI: 10.1007/s12013-024-01323-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/17/2024] [Indexed: 06/06/2024]
Abstract
The CCN family is a group of matricellular proteins associated with the extracellular matrix. This study aims to explore the role of the CCN family in glioma development and its implications in the tumor microenvironment. Through analysis of bulk RNA-seq cohorts, correlations between CCN family expression and glioma subtypes, patient survival, and bioactive pathway enrichment were investigated. Additionally, single-cell datasets were employed to identify novel cell subgroups, followed by analyses of cell communication and transcription factors. Spatial transcriptomic analysis was utilized to validate the CCN family's involvement in glioma. Results indicate overexpression of CYR61,CTGF, and WISP1 in glioma, associated with unfavorable subtypes and reduced survival. Enrichment analyses revealed associations with oncogenic pathways, while CTGF and WISP1 expression correlated with increased infiltration of regulatory T cells and M2 macrophages. Single-cell analysis identified MES-like cells as the highest CCN expression. Moreover, intercellular signal transduction analysis demonstrated active pathways, including SPP1-CD44, in cell subgroups with elevated CYR61 and CTGF expression. Spatial transcriptomic analysis confirmed co-localization of CYR61,CTGF and SPP1-CD44 with high oncogenic pathway activity. These findings suggest that CCN family members may serve as potential prognostic biomarkers and therapeutic targets for glioma.
Collapse
Affiliation(s)
- Jingyan Gu
- Department of Neurosurgery, Shanghai General Hospital affiliated to Nanjing Medical University, Shanghai, China
- Department of Neurosurgery, Shanghai General Hospital affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Wenjie Tong
- Department of Neurosurgery, Shanghai General Hospital affiliated to Nanjing Medical University, Shanghai, China
- Department of Neurosurgery, Songjiang Hospital Affiliated to Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Xu Wang
- Department of Neurosurgery, Shanghai General Hospital affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Lianping Gu
- Department of Neurosurgery, Shanghai General Hospital affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Wei Wang
- Department of Neurosurgery, Shanghai General Hospital affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Tingting Zang
- Department of Neurosurgery, Shanghai General Hospital affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Meiqing Lou
- Department of Neurosurgery, Shanghai General Hospital affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| | - Yaohua Liu
- Department of Neurosurgery, Shanghai General Hospital affiliated to Nanjing Medical University, Shanghai, China.
- Department of Neurosurgery, Shanghai General Hospital affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| |
Collapse
|
25
|
Song P, Deng H, Liu Y, Zhang M. Integrated bioinformatics analysis and experimental validation reveal the relationship between ALOX5AP and the prognosis and immune microenvironment in glioma. BMC Med Genomics 2024; 17:218. [PMID: 39169376 PMCID: PMC11337642 DOI: 10.1186/s12920-024-01991-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2024] [Accepted: 08/13/2024] [Indexed: 08/23/2024] Open
Abstract
BACKGROUND Treatment of gliomas, the most prevalent primary malignant neoplasm of the central nervous system, is challenging. Arachidonate 5-lipoxygenase activating protein (ALOX5AP) is crucial for converting arachidonic acid into leukotrienes and is associated with poor prognosis in multiple cancers. Nevertheless, its relationship with the prognosis and the immune microenvironment of gliomas remains incompletely understood. METHODS The differential expression of ALOX5AP was evaluated based on public Databases. Kaplan-Meier, multivariate Cox proportional hazards regression analysis, time-dependent receiver operating characteristic, and nomogram were used to estimate the prognostic value of ALOX5AP. The relationship between ALOX5AP and immune infiltration was calculated using ESTIMATE and CIBERSORT algorithms. Relationships between ALOX5AP and human leukocyte antigen molecules, immune checkpoints, tumor mutation burden, TIDE score, and immunophenoscore were calculated to evaluate glioma immunotherapy response. Single gene GSEA and co-expression network-based GO and KEGG enrichment analysis were performed to explore the potential function of ALOX5AP. ALOX5AP expression was verified using multiplex immunofluorescence staining and its prognostic effects were confirmed using a glioma tissue microarray. RESULT ALOX5AP was highly expressed in gliomas, and the expression level was related to World Health Organization (WHO) grade, age, sex, IDH mutation status, 1p19q co-deletion status, MGMTp methylation status, and poor prognosis. Single-cell RNA sequencing showed that ALOX5AP was expressed in macrophages, monocytes, and T cells but not in tumor cells. ALOX5AP expression positively correlated with M2 macrophage infiltration and poor immunotherapy response. Immunofluorescence staining demonstrated that ALOX5AP was upregulated in WHO higher-grade gliomas, localizing to M2 macrophages. Glioma tissue microarray confirmed the adverse effect of ALOX5AP in the prognosis of glioma. CONCLUSION ALOX5AP is highly expressed in M2 macrophages and may act as a potential biomarker for predicting prognosis and immunotherapy response in patients with glioma.
Collapse
Affiliation(s)
- Ping Song
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Hubei, Wuhan, 430030, P.R. China
| | - Hui Deng
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Hubei, Wuhan, 430030, P.R. China
| | - Yushu Liu
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Hubei, Wuhan, 430030, P.R. China
| | - Mengxian Zhang
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Hubei, Wuhan, 430030, P.R. China.
| |
Collapse
|
26
|
Mongeon B, Hébert-Doutreloux J, Surendran A, Karimi E, Fiset B, Quail DF, Walsh LA, Jenner AL, Craig M. Spatial computational modelling illuminates the role of the tumour microenvironment for treating glioblastoma with immunotherapies. NPJ Syst Biol Appl 2024; 10:91. [PMID: 39155294 PMCID: PMC11330976 DOI: 10.1038/s41540-024-00419-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2023] [Accepted: 08/07/2024] [Indexed: 08/20/2024] Open
Abstract
Glioblastoma is the most common and deadliest brain tumour in adults, with a median survival of 15 months under the current standard of care. Immunotherapies like immune checkpoint inhibitors and oncolytic viruses have been extensively studied to improve this endpoint. However, most thus far have failed. To improve the efficacy of immunotherapies to treat glioblastoma, new single-cell imaging modalities like imaging mass cytometry can be leveraged and integrated with computational models. This enables a better understanding of the tumour microenvironment and its role in treatment success or failure in this hard-to-treat tumour. Here, we implemented an agent-based model that allows for spatial predictions of combination chemotherapy, oncolytic virus, and immune checkpoint inhibitors against glioblastoma. We initialised our model with patient imaging mass cytometry data to predict patient-specific responses and found that oncolytic viruses drive combination treatment responses determined by intratumoral cell density. We found that tumours with higher tumour cell density responded better to treatment. When fixing the number of cancer cells, treatment efficacy was shown to be a function of CD4 + T cell and, to a lesser extent, of macrophage counts. Critically, our simulations show that care must be put into the integration of spatial data and agent-based models to effectively capture intratumoral dynamics. Together, this study emphasizes the use of predictive spatial modelling to better understand cancer immunotherapy treatment dynamics, while highlighting key factors to consider during model design and implementation.
Collapse
Affiliation(s)
- Blanche Mongeon
- Sainte-Justine University Hospital Azrieli Research Centre, Montréal, QC, Canada
- Department of Mathematics and Statistics, Université de Montréal, Montréal, QC, Canada
| | | | - Anudeep Surendran
- Center for Advanced Systems Understanding, Görlitz, Germany
- Helmholtz-Zentrum Dresden-Rossendorf, Dresden, Germany
| | - Elham Karimi
- Rosalind and Morris Goodman Cancer Institute, McGill University, Montréal, QC, Canada
| | - Benoit Fiset
- Rosalind and Morris Goodman Cancer Institute, McGill University, Montréal, QC, Canada
| | - Daniela F Quail
- Rosalind and Morris Goodman Cancer Institute, McGill University, Montréal, QC, Canada
- Department of Physiology, Faculty of Medicine, McGill University, Montréal, QC, Canada
- Department of Medicine, Division of Experimental Medicine, McGill University, Montréal, QC, Canada
| | - Logan A Walsh
- Rosalind and Morris Goodman Cancer Institute, McGill University, Montréal, QC, Canada
- Department of Human Genetics, McGill University, Montréal, QC, Canada
| | - Adrianne L Jenner
- School of Mathematical Sciences, Queensland University of Technology, Brisbane, QLD, Australia
| | - Morgan Craig
- Sainte-Justine University Hospital Azrieli Research Centre, Montréal, QC, Canada.
- Department of Mathematics and Statistics, Université de Montréal, Montréal, QC, Canada.
| |
Collapse
|
27
|
Yuan Z, Zhang Y, Wang X, Wang X, Ren S, He X, Su J, Zheng A, Guo S, Chen Y, Deng S, Wu X, Li M, Du F, Zhao Y, Shen J, Wang Z, Xiao Z. The investigation of oncolytic viruses in the field of cancer therapy. Front Oncol 2024; 14:1423143. [PMID: 39055561 PMCID: PMC11270537 DOI: 10.3389/fonc.2024.1423143] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2024] [Accepted: 06/26/2024] [Indexed: 07/27/2024] Open
Abstract
Oncolytic viruses (OVs) have emerged as a potential strategy for tumor treatment due to their ability to selectively replicate in tumor cells, induce apoptosis, and stimulate immune responses. However, the therapeutic efficacy of single OVs is limited by the complexity and immunosuppressive nature of the tumor microenvironment (TME). To overcome these challenges, engineering OVs has become an important research direction. This review focuses on engineering methods and multi-modal combination therapies for OVs aimed at addressing delivery barriers, viral phagocytosis, and antiviral immunity in tumor therapy. The engineering approaches discussed include enhancing in vivo immune response, improving replication efficiency within the tumor cells, enhancing safety profiles, and improving targeting capabilities. In addition, this review describes the potential mechanisms of OVs combined with radiotherapy, chemotherapy, cell therapy and immune checkpoint inhibitors (ICIs), and summarizes the data of ongoing clinical trials. By continuously optimizing engineering strategies and combination therapy programs, we can achieve improved treatment outcomes and quality of life for cancer patients.
Collapse
Affiliation(s)
- Zijun Yuan
- Gulin Traditional Chinese Medicine Hospital, Luzhou, China
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, China
| | - Yinping Zhang
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, China
| | - Xiang Wang
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, China
| | - Xingyue Wang
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, China
| | - Siqi Ren
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, China
| | - Xinyu He
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, China
| | - Jiahong Su
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, China
| | - Anfu Zheng
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, China
| | - Sipeng Guo
- Research And Experiment Center, Sichuan College of Traditional Chinese Medicine, Mianyang, China
| | - Yu Chen
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, China
- Cell Therapy & Cell Drugs of Luzhou Key Laboratory, Luzhou, China
- South Sichuan Institute of Translational Medicine, Luzhou, China
| | - Shuai Deng
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, China
- Cell Therapy & Cell Drugs of Luzhou Key Laboratory, Luzhou, China
- South Sichuan Institute of Translational Medicine, Luzhou, China
| | - Xu Wu
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, China
- Cell Therapy & Cell Drugs of Luzhou Key Laboratory, Luzhou, China
- South Sichuan Institute of Translational Medicine, Luzhou, China
| | - Mingxing Li
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, China
- Cell Therapy & Cell Drugs of Luzhou Key Laboratory, Luzhou, China
- South Sichuan Institute of Translational Medicine, Luzhou, China
| | - Fukuan Du
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, China
- Cell Therapy & Cell Drugs of Luzhou Key Laboratory, Luzhou, China
- South Sichuan Institute of Translational Medicine, Luzhou, China
| | - Yueshui Zhao
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, China
- Cell Therapy & Cell Drugs of Luzhou Key Laboratory, Luzhou, China
- South Sichuan Institute of Translational Medicine, Luzhou, China
| | - Jing Shen
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, China
- Cell Therapy & Cell Drugs of Luzhou Key Laboratory, Luzhou, China
- South Sichuan Institute of Translational Medicine, Luzhou, China
| | - Zechen Wang
- Gulin Traditional Chinese Medicine Hospital, Luzhou, China
| | - Zhangang Xiao
- Gulin Traditional Chinese Medicine Hospital, Luzhou, China
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, China
- Cell Therapy & Cell Drugs of Luzhou Key Laboratory, Luzhou, China
- South Sichuan Institute of Translational Medicine, Luzhou, China
- Department of Pharmacology, School of Pharmacy, Sichuan College of Traditional Chinese Medicine, Mianyang, China
| |
Collapse
|
28
|
Loginova N, Aniskin D, Timashev P, Ulasov I, Kharwar RK. GBM Immunotherapy: Macrophage Impacts. Immunol Invest 2024; 53:730-751. [PMID: 38634572 DOI: 10.1080/08820139.2024.2337022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/19/2024]
Abstract
BACKGROUND Glioblastoma (GBM) is an extremely aggressive form of brain tumor with low survival rates. Current treatments such as chemotherapy, radiation, and surgery are problematic due to tumor growth, invasion, and tumor microenvironment. GBM cells are resistant to these standard treatments, and the heterogeneity of the tumor makes it difficult to find a universal approach. Progression of GBM and acquisition of resistance to therapy are due to the complex interplay between tumor cells and the TME. A significant portion of the TME consists of an inflammatory infiltrate, with microglia and macrophages being the predominant cells. METHODS Analysis of the literature data over a course of 5 years suggest that the tumor-associated macrophages (TAMs) are capable of releasing cytokines and growth factors that promote tumor proliferation, survival, and metastasis while inhibiting immune cell function at the same time. RESULTS Thus, immunosuppressive state, provided with this intensively studied kind of TME cells, is supposed to promote GBM development through TAMs modulation of tumor treatment-resistance and aggressiveness. Therefore, TAMs are an attractive therapeutic target in the treatment of glioblastoma. CONCLUSION This review provides a comprehensive overview of the latest research on the nature of TAMs and the development of therapeutic strategies targeting TAMs, focusing on the variety of macrophage properties, being modulated, as well as molecular targets.
Collapse
Affiliation(s)
- Nina Loginova
- Group of Experimental Biotherapy and Diagnostics, Institute for Regenerative Medicine, World-Class Research Centre "Digital Biodesign and Personalized Healthcare", I.M. Sechenov First Moscow State Medical University, Moscow, Russia
| | - Denis Aniskin
- Group of Experimental Biotherapy and Diagnostics, Institute for Regenerative Medicine, World-Class Research Centre "Digital Biodesign and Personalized Healthcare", I.M. Sechenov First Moscow State Medical University, Moscow, Russia
| | - Peter Timashev
- World-Class Research Centre "Digital Biodesign and Personalized Healthcare", Sechenov First Moscow State Medical University, Moscow, Russia
| | - Ilya Ulasov
- Group of Experimental Biotherapy and Diagnostics, Institute for Regenerative Medicine, World-Class Research Centre "Digital Biodesign and Personalized Healthcare", I.M. Sechenov First Moscow State Medical University, Moscow, Russia
| | - Rajesh Kumar Kharwar
- Endocrine Research Laboratory, Department of Zoology, University of Lucknow, Lucknow, India
| |
Collapse
|
29
|
Aghajani M, Jalilzadeh N, Aghebati-Maleki A, Yari A, Tabnak P, Mardi A, Saeedi H, Aghebati-Maleki L, Baradaran B. Current approaches in glioblastoma multiforme immunotherapy. Clin Transl Oncol 2024; 26:1584-1612. [PMID: 38512448 DOI: 10.1007/s12094-024-03395-7] [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: 07/26/2023] [Accepted: 01/08/2024] [Indexed: 03/23/2024]
Abstract
Glioblastoma multiform (GBM) is the most prevalent CNS (central nervous system) tumor in adults, with an average survival length shorter than 2 years and rare metastasis to organs other than CNS. Despite extensive attempts at surgical resecting, the inherently permeable nature of this disease has rendered relapse nearly unavoidable. Thus, immunotherapy is a feasible alternative, as stimulated immune cells can enter into the remote and inaccessible tumor cells. Immunotherapy has revolutionized patient upshots in various malignancies and might introduce different effective ways for GBM patients. Currently, researchers are exploring various immunotherapeutic strategies in patients with GBM to target both the innate and acquired immune responses. These approaches include reprogrammed tumor-associated macrophages, the use of specific antibodies to inhibit tumor progression and metastasis, modifying tumor-associated macrophages with antibodies, vaccines that utilize tumor-specific dendritic cells to activate anti-tumor T cells, immune checkpoint inhibitors, and enhanced T cells that function against tumor cells. Despite these findings, there is still room for improving the response faults of the many currently tested immunotherapies. This study aims to review the currently used immunotherapy approaches with their molecular mechanisms and clinical application in GBM.
Collapse
Affiliation(s)
- Marjan Aghajani
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Nazila Jalilzadeh
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Ali Aghebati-Maleki
- Molecular Medicine Department, Faculty of Modern Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Amirhossein Yari
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
- Department of Biology, Islamic Azad University, Tabriz Branch, Tabriz, Iran
| | - Peyman Tabnak
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
- Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Amirhossein Mardi
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
- Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran
- Department of Immunology, School of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Hossein Saeedi
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Leili Aghebati-Maleki
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.
- Department of Immunology, School of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran.
| | - Behzad Baradaran
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.
- Department of Immunology, School of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran.
| |
Collapse
|
30
|
Koula G, Yakati V, Rachamalla HK, Bhamidipati K, Kathirvel M, Banerjee R, Puvvada N. Integrin receptor-targeted, doxorubicin-loaded cerium oxide nanoparticles delivery to combat glioblastoma. Nanomedicine (Lond) 2024; 19:1389-1406. [PMID: 38912661 PMCID: PMC11318704 DOI: 10.1080/17435889.2024.2350357] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2023] [Accepted: 04/29/2024] [Indexed: 06/25/2024] Open
Abstract
Aim: To assess the chemo-immunomodulatory effects of doxorubicin-loaded cerium oxide nanoparticles coated with oleyl amine-linked cyclic RGDfK peptide (CeNP+Dox+RGD) to target both gliomas and its tumor microenvironment (TME) via integrin receptors. Materials & methods: CeNP+Dox+RGD nanoparticles are synthesized by the sequential addition of cerium III chloride heptahydrate, beta-cyclodextrin, oleic acid, and F127 micelle (CeNP). Doxorubicin was then loaded into CeNPs and coated with oleyl amine-linked cyclic RGDfK peptide to form stable CeNP+Dox+RGD nanoparticles. Results: CeNP+Dox+RGD nanoparticles crossed blood-brain barrier (BBB) effectively and demonstrated threefold enhanced survivability in glioma-bearing mice. The IHC profiling of glial tumor cross-sections showed increased CD80 expression (M1 TAMs) and decreased arginase-1 expression (M2 TAMs). Conclusion: CeNP+Dox+RGD can be an immunotherapeutic treatment option to combat glioblastoma.
Collapse
Affiliation(s)
- Gayathri Koula
- Department of Oils, Lipids Sciences & Technology, CSIR-Indian Institute of Chemical Technology, Hyderabad, 500007, India
- Academy of Scientific & Innovative Research (AcSIR), Ghaziabad, 201002, Uttar Pradesh (U.P), India
| | - Venu Yakati
- Department of Oils, Lipids Sciences & Technology, CSIR-Indian Institute of Chemical Technology, Hyderabad, 500007, India
- Academy of Scientific & Innovative Research (AcSIR), Ghaziabad, 201002, Uttar Pradesh (U.P), India
| | - Hari Krishnareddy Rachamalla
- Department of Oils, Lipids Sciences & Technology, CSIR-Indian Institute of Chemical Technology, Hyderabad, 500007, India
- Academy of Scientific & Innovative Research (AcSIR), Ghaziabad, 201002, Uttar Pradesh (U.P), India
| | - Keerti Bhamidipati
- Applied Biology Division, CSIR-Indian Institute of Chemical Technology, Hyderabad, 500007, India
- Academy of Scientific & Innovative Research (AcSIR), Ghaziabad, 201002, Uttar Pradesh (U.P), India
| | - Muralidharan Kathirvel
- Applied Biology Division, CSIR-Indian Institute of Chemical Technology, Hyderabad, 500007, India
| | - Rajkumar Banerjee
- Department of Oils, Lipids Sciences & Technology, CSIR-Indian Institute of Chemical Technology, Hyderabad, 500007, India
| | - Nagaprasad Puvvada
- Department of Oils, Lipids Sciences & Technology, CSIR-Indian Institute of Chemical Technology, Hyderabad, 500007, India
| |
Collapse
|
31
|
Si J, Guo J, Zhang X, Li W, Zhang S, Shang S, Zhang Q. Hypoxia-induced activation of HIF-1alpha/IL-1beta axis in microglia promotes glioma progression via NF-κB-mediated upregulation of heparanase expression. Biol Direct 2024; 19:45. [PMID: 38863009 PMCID: PMC11165725 DOI: 10.1186/s13062-024-00487-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2024] [Accepted: 05/31/2024] [Indexed: 06/13/2024] Open
Abstract
BACKGROUND Glioma is a common tumor that occurs in the brain and spinal cord. Hypoxia is a crucial feature of the tumor microenvironment. Tumor-associated macrophages/microglia play a crucial role in the advancement of glioma. This study aims to illuminate the detailed mechanisms by which hypoxia regulates microglia and, consequently, influences the progression of glioma. METHODS The glioma cell viability and proliferation were analyzed by cell counting kit-8 assay and 5-ethynyl-2'-deoxyuridine assay. Wound healing assay and transwell assay were implemented to detect glioma cell migration and invasion, respectively. Enzyme-linked immunosorbent assay was conducted to detect protein levels in cell culture medium. The protein levels in glioma cells and tumor tissues were evaluated using western blot analysis. The histological morphology of tumor tissue was determined by hematoxylin-eosin staining. The protein expression in tumor tissues was determined using immunohistochemistry. Human glioma xenograft in nude mice was employed to test the influence of hypoxic microglia-derived interleukin-1beta (IL-1β) and heparanase (HPSE) on glioma growth in vivo. RESULTS Hypoxic HMC3 cells promoted proliferation, migration, and invasion abilities of U251 and U87 cells by secreting IL-1β, which was upregulated by hypoxia-induced activation of hypoxia inducible factor-1alpha (HIF-1α). Besides, IL-1β from HMC3 cells promoted glioma progression and caused activation of nuclear factor-κB (NF-κB) and upregulation of HPSE in vivo. We also confirmed that IL-1β facilitated HPSE expression in U251 and U87 cells by activating NF-κB. Hypoxic HMC3 cells-secreted IL-1β facilitated the proliferation, migration, and invasion of U251 and U87 cells via NF-κB-mediated upregulation of HPSE expression. Finally, we revealed that silencing HPSE curbed the proliferation and metastasis of glioma in mice. CONCLUSION Hypoxia-induced activation of HIF-1α/IL-1β axis in microglia promoted glioma progression via NF-κB-mediated upregulation of HPSE expression.
Collapse
Affiliation(s)
- Jinchao Si
- Department of Neurology, the Second Affiliated Hospital of Zhengzhou University, Zhengzhou, 450014, China
| | - Jingya Guo
- Department of Neuroelectrophysiology, Zhengzhou Central Hospital Affiliated to Zhengzhou University, Zhengzhou, 450007, China
| | - Xu Zhang
- Department of General Practice, the Second Affiliated Hospital of Zhengzhou University, Zhengzhou, 450014, China
| | - Wei Li
- Department of Physiology, School of Basic Medicine, Zhengzhou University, Zhengzhou, 450066, China
| | - Shen Zhang
- Department of Neuroelectrophysiology, Zhengzhou Central Hospital Affiliated to Zhengzhou University, Zhengzhou, 450007, China
| | - Shuyu Shang
- Department of Physiology, Medical College, HuangHe Science and Technology University, Zhengzhou, 450064, China
| | - Quanwu Zhang
- Department of Pathology, Zhengzhou Central Hospital Affiliated to Zhengzhou University, No. 16 Tongbai North Road, Zhengzhou, Henan Province, 450007, China.
| |
Collapse
|
32
|
Tharp ME, Han CZ, Balak CD, Fitzpatrick C, O'Connor C, Preissl S, Buchanan J, Nott A, Escoubet L, Mavrommatis K, Gupta M, Schwartz MS, Sang UH, Jones PS, Levy ML, Gonda DD, Ben-Haim S, Ciacci J, Barba D, Khalessi A, Coufal NG, Chen CC, Glass CK, Page DC. The inactive X chromosome drives sex differences in microglial inflammatory activity in human glioblastoma. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.06.06.597433. [PMID: 38895459 PMCID: PMC11185629 DOI: 10.1101/2024.06.06.597433] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/21/2024]
Abstract
Biological sex is an important risk factor in cancer, but the underlying cell types and mechanisms remain obscure. Since tumor development is regulated by the immune system, we hypothesize that sex-biased immune interactions underpin sex differences in cancer. The male-biased glioblastoma multiforme (GBM) is an aggressive and treatment-refractory tumor in urgent need of more innovative approaches, such as considering sex differences, to improve outcomes. GBM arises in the specialized brain immune environment dominated by microglia, so we explored sex differences in this immune cell type. We isolated adult human TAM-MGs (tumor-associated macrophages enriched for microglia) and control microglia and found sex-biased inflammatory signatures in GBM and lower-grade tumors associated with pro-tumorigenic activity in males and anti-tumorigenic activity in females. We demonstrated that genes expressed or modulated by the inactive X chromosome facilitate this bias. Together, our results implicate TAM-MGs, specifically their sex chromosomes, as drivers of male bias in GBM.
Collapse
Affiliation(s)
- Marla E Tharp
- Whitehead Institute, Cambridge, MA 02142, USA
- These authors contributed equally
| | - Claudia Z Han
- Department of Cellular and Molecular Medicine, University of California, San Diego, La Jolla, CA 92093, USA
- These authors contributed equally
| | - Chris D Balak
- Department of Cellular and Molecular Medicine, University of California, San Diego, La Jolla, CA 92093, USA
| | - Conor Fitzpatrick
- Flow Cytometry Core Facility, The Salk Institute for Biological Studies, La Jolla, CA 92037, USA
| | - Carolyn O'Connor
- Flow Cytometry Core Facility, The Salk Institute for Biological Studies, La Jolla, CA 92037, USA
| | - Sebastian Preissl
- Department of Cellular and Molecular Medicine, University of California, San Diego, La Jolla, CA 92093, USA
- Center for Epigenomics, University of California, San Diego, La Jolla, CA 92093, USA
- Present address: Institute of Experimental and Clinical Pharmacology and Toxicology, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Justin Buchanan
- Department of Cellular and Molecular Medicine, University of California, San Diego, La Jolla, CA 92093, USA
- Center for Epigenomics, University of California, San Diego, La Jolla, CA 92093, USA
| | - Alexi Nott
- Department of Cellular and Molecular Medicine, University of California, San Diego, La Jolla, CA 92093, USA
- Department of Brain Sciences, Imperial College London, London, United Kingdom
- UK Dementia Research Institute, Imperial College London, London, United Kingdom
| | | | | | - Mihir Gupta
- Department of Neurosurgery, University of California, San Diego, La Jolla, CA 92037, USA
- Present address: Department of Neurosurgery, Yale University, New Haven, CT 06520, USA
| | - Marc S Schwartz
- Department of Neurosurgery, University of California, San Diego, La Jolla, CA 92037, USA
| | - U Hoi Sang
- Department of Neurosurgery, University of California, San Diego, La Jolla, CA 92037, USA
| | - Pamela S Jones
- Department of Neurosurgery, University of California, San Diego, La Jolla, CA 92037, USA
- Present address: Department of Neurosurgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
| | - Michael L Levy
- Department of Neurosurgery, University of California, San Diego-Rady Children's Hospital, San Diego, CA 92123, USA
| | - David D Gonda
- Department of Neurosurgery, University of California, San Diego-Rady Children's Hospital, San Diego, CA 92123, USA
| | - Sharona Ben-Haim
- Department of Neurosurgery, University of California, San Diego, La Jolla, CA 92037, USA
| | - Joseph Ciacci
- Department of Neurosurgery, University of California, San Diego, La Jolla, CA 92037, USA
| | - David Barba
- Department of Neurosurgery, University of California, San Diego, La Jolla, CA 92037, USA
| | - Alexander Khalessi
- Department of Neurosurgery, University of California, San Diego, La Jolla, CA 92037, USA
| | - Nicole G Coufal
- Department of Pediatrics University of California, San Diego, La Jolla, CA 92093, USA
- Sanford Consortium for Regenerative Medicine, La Jolla, CA 92037, USA
| | - Clark C Chen
- Department of Neurosurgery, University of California, San Diego, La Jolla, CA 92037, USA
- Present address: Department of Neurosurgery, University of Minnesota, Minneapolis, MN 55455, USA
| | - Christopher K Glass
- Department of Cellular and Molecular Medicine, University of California, San Diego, La Jolla, CA 92093, USA
| | - David C Page
- Whitehead Institute, Cambridge, MA 02142, USA
- Department of Biology, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
- Howard Hughes Medical Institute, Whitehead Institute, Cambridge, MA 02142, USA
| |
Collapse
|
33
|
Niu X, Zhang Z, Zhou Q, Wuethrich A, Lobb R, Trau M. Analysis of secreted small extracellular vesicles from activated human microglial cell lines reveals distinct pro- and anti-inflammatory proteomic profiles. Proteomics 2024; 24:e2300094. [PMID: 38343172 DOI: 10.1002/pmic.202300094] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2023] [Revised: 12/12/2023] [Accepted: 01/26/2024] [Indexed: 06/04/2024]
Abstract
Microglia are a specialized population of innate immune cells located in the central nervous system. In response to physiological and pathological changes in their microenvironment, microglia can polarize into pro-inflammatory or anti-inflammatory phenotypes. A dysregulation in the pro-/anti-inflammatory balance is associated with many pathophysiological changes in the brain and nervous system. Therefore, the balance between microglia pro-/anti-inflammatory polarization can be a potential biomarker for the various brain pathologies. A non-invasive method of detecting microglia polarization in patients would have promising clinical applications. Here, we perform proteomic analysis of small extracellular vesicles (sEVs) derived from microglia cells to identify sEVs biomarkers indicative of pro-inflammatory and anti-inflammatory phenotypic changes. sEVs were isolated from microglia cell lines under different inflammatory conditions and analyzed by proteomics by liquid chromatography with mass spectrometry. Our findings provide the potential roles of sEVs that could be related to the pathogenesis of various brain diseases.
Collapse
Affiliation(s)
- Xueming Niu
- Centre for Personalised Nanomedicine, Australian Institute for Bioengineering and Nanotechnology (AIBN), The University of Queensland, Brisbane, QLD, Australia
| | - Zhen Zhang
- Centre for Personalised Nanomedicine, Australian Institute for Bioengineering and Nanotechnology (AIBN), The University of Queensland, Brisbane, QLD, Australia
| | - Quan Zhou
- Centre for Personalised Nanomedicine, Australian Institute for Bioengineering and Nanotechnology (AIBN), The University of Queensland, Brisbane, QLD, Australia
| | - Alain Wuethrich
- Centre for Personalised Nanomedicine, Australian Institute for Bioengineering and Nanotechnology (AIBN), The University of Queensland, Brisbane, QLD, Australia
| | - Richard Lobb
- Centre for Personalised Nanomedicine, Australian Institute for Bioengineering and Nanotechnology (AIBN), The University of Queensland, Brisbane, QLD, Australia
| | - Matt Trau
- Centre for Personalised Nanomedicine, Australian Institute for Bioengineering and Nanotechnology (AIBN), The University of Queensland, Brisbane, QLD, Australia
- School of Chemistry and Molecular Biosciences, The University of Queensland, Brisbane, QLD, Australia
| |
Collapse
|
34
|
Smerdi D, Moutafi M, Kotsantis I, Stavrinou LC, Psyrri A. Overcoming Resistance to Temozolomide in Glioblastoma: A Scoping Review of Preclinical and Clinical Data. Life (Basel) 2024; 14:673. [PMID: 38929657 PMCID: PMC11204771 DOI: 10.3390/life14060673] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2024] [Revised: 05/15/2024] [Accepted: 05/17/2024] [Indexed: 06/28/2024] Open
Abstract
Glioblastoma (GB) is the most common and most aggressive primary brain tumor in adults, with an overall survival almost 14.6 months. Optimal resection followed by combined temozolomide chemotherapy and radiotherapy, also known as Stupp protocol, remains the standard of treatment; nevertheless, resistance to temozolomide, which can be obtained throughout many molecular pathways, is still an unsurpassed obstacle. Several factors influence the efficacy of temozolomide, including the involvement of other DNA repair systems, aberrant signaling pathways, autophagy, epigenetic modifications, microRNAs, and extracellular vesicle production. The blood-brain barrier, which serves as both a physical and biochemical obstacle, the tumor microenvironment's pro-cancerogenic and immunosuppressive nature, and tumor-specific characteristics such as volume and antigen expression, are the subject of ongoing investigation. In this review, preclinical and clinical data about temozolomide resistance acquisition and possible ways to overcome chemoresistance, or to treat gliomas without restoration of chemosensitinity, are evaluated and presented. The objective is to offer a thorough examination of the clinically significant molecular mechanisms and their intricate interrelationships, with the aim of enhancing understanding to combat resistance to TMZ more effectively.
Collapse
Affiliation(s)
- Dimitra Smerdi
- Department of Medical Oncology, Second Department of Internal Medicine, “Attikon” University General Hospital, Athens Medical School, National and Kapodistrian University of Athens, 11528 Athens, Greece
| | - Myrto Moutafi
- Department of Medical Oncology, Second Department of Internal Medicine, “Attikon” University General Hospital, Athens Medical School, National and Kapodistrian University of Athens, 11528 Athens, Greece
| | - Ioannis Kotsantis
- Department of Medical Oncology, Second Department of Internal Medicine, “Attikon” University General Hospital, Athens Medical School, National and Kapodistrian University of Athens, 11528 Athens, Greece
| | - Lampis C. Stavrinou
- Department of Neurosurgery and Neurotraumatology, “Attikon” University General Hospital, Athens Medical School, National and Kapodistrian University of Athens, 12462 Athens, Greece
| | - Amanda Psyrri
- Department of Medical Oncology, Second Department of Internal Medicine, “Attikon” University General Hospital, Athens Medical School, National and Kapodistrian University of Athens, 11528 Athens, Greece
| |
Collapse
|
35
|
Nusraty S, Boddeti U, Zaghloul KA, Brown DA. Microglia in Glioblastomas: Molecular Insight and Immunotherapeutic Potential. Cancers (Basel) 2024; 16:1972. [PMID: 38893093 PMCID: PMC11171200 DOI: 10.3390/cancers16111972] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2024] [Revised: 05/18/2024] [Accepted: 05/20/2024] [Indexed: 06/21/2024] Open
Abstract
Glioblastoma (GBM) is one of the most aggressive and devastating primary brain tumors, with a median survival of 15 months following diagnosis. Despite the intense treatment regimen which routinely includes maximal safe neurosurgical resection followed by adjuvant radio- and chemotherapy, the disease remains uniformly fatal. The poor prognosis associated with GBM is multifactorial owing to factors such as increased proliferation, angiogenesis, and metabolic switching to glycolytic pathways. Critically, GBM-mediated local and systemic immunosuppression result in inadequate immune surveillance and ultimately, tumor-immune escape. Microglia-the resident macrophages of the central nervous system (CNS)-play crucial roles in mediating the local immune response in the brain. Depending on the specific pathological cues, microglia are activated into either a pro-inflammatory, neurotoxic phenotype, known as M1, or an anti-inflammatory, regenerative phenotype, known as M2. In either case, microglia secrete corresponding pro- or anti-inflammatory cytokines and chemokines that either promote or hinder tumor growth. Herein, we review the interplay between GBM cells and resident microglia with a focus on contemporary studies highlighting the effect of GBM on the subtypes of microglia expressed, the associated cytokines/chemokines secreted, and ultimately, their impact on tumor pathogenesis. Finally, we explore how understanding the intricacies of the tumor-immune landscape can inform novel immunotherapeutic strategies against this devastating disease.
Collapse
Affiliation(s)
| | | | | | - Desmond A. Brown
- Surgical Neurology Branch, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD 20892, USA; (S.N.); (U.B.); (K.A.Z.)
| |
Collapse
|
36
|
Bugakova AS, Chudakova DA, Myzina MS, Yanysheva EP, Ozerskaya IV, Soboleva AV, Baklaushev VP, Yusubalieva GM. Non-Tumor Cells within the Tumor Microenvironment-The "Eminence Grise" of the Glioblastoma Pathogenesis and Potential Targets for Therapy. Cells 2024; 13:808. [PMID: 38786032 PMCID: PMC11119139 DOI: 10.3390/cells13100808] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2024] [Revised: 04/26/2024] [Accepted: 04/30/2024] [Indexed: 05/25/2024] Open
Abstract
Glioblastoma (GBM) is the most common malignancy of the central nervous system in adults. GBM has high levels of therapy failure and its prognosis is usually dismal. The phenotypic heterogeneity of the tumor cells, dynamic complexity of non-tumor cell populations within the GBM tumor microenvironment (TME), and their bi-directional cross-talk contribute to the challenges of current therapeutic approaches. Herein, we discuss the etiology of GBM, and describe several major types of non-tumor cells within its TME, their impact on GBM pathogenesis, and molecular mechanisms of such an impact. We also discuss their value as potential therapeutic targets or prognostic biomarkers, with reference to the most recent works on this subject. We conclude that unless all "key player" populations of non-tumor cells within the TME are considered, no breakthrough in developing treatment for GBM can be achieved.
Collapse
Affiliation(s)
- Aleksandra S. Bugakova
- Federal Center for Brain and Neurotechnologies, Federal Medical and Biological Agency of Russia, 117513 Moscow, Russia
| | - Daria A. Chudakova
- Federal Center for Brain and Neurotechnologies, Federal Medical and Biological Agency of Russia, 117513 Moscow, Russia
| | - Maria S. Myzina
- Federal Center for Brain and Neurotechnologies, Federal Medical and Biological Agency of Russia, 117513 Moscow, Russia
| | - Elvira P. Yanysheva
- Federal Research and Clinical Center of Specialized Medical Care and Medical Technologies Federal Medical and Biological Agency of Russia, 115682 Moscow, Russia
| | - Iuliia V. Ozerskaya
- Pulmonology Research Institute, Federal Medical and Biological Agency of Russia, 115682 Moscow, Russia
| | - Alesya V. Soboleva
- Federal Center for Brain and Neurotechnologies, Federal Medical and Biological Agency of Russia, 117513 Moscow, Russia
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, 119991 Moscow, Russia
| | - Vladimir P. Baklaushev
- Federal Center for Brain and Neurotechnologies, Federal Medical and Biological Agency of Russia, 117513 Moscow, Russia
- Federal Research and Clinical Center of Specialized Medical Care and Medical Technologies Federal Medical and Biological Agency of Russia, 115682 Moscow, Russia
- Pulmonology Research Institute, Federal Medical and Biological Agency of Russia, 115682 Moscow, Russia
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, 119991 Moscow, Russia
- Department of Medical Nanobiotechnology of Medical and Biological Faculty, Pirogov Russian National Research Medical University, Ministry of Health of the Russian Federation, 117997 Moscow, Russia
| | - Gaukhar M. Yusubalieva
- Federal Center for Brain and Neurotechnologies, Federal Medical and Biological Agency of Russia, 117513 Moscow, Russia
- Federal Research and Clinical Center of Specialized Medical Care and Medical Technologies Federal Medical and Biological Agency of Russia, 115682 Moscow, Russia
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, 119991 Moscow, Russia
| |
Collapse
|
37
|
Martinez-Morga M, Garrigos D, Rodriguez-Montero E, Pombero A, Garcia-Lopez R, Martinez S. Pericytes Are Immunoregulatory Cells in Glioma Genesis and Progression. Int J Mol Sci 2024; 25:5072. [PMID: 38791110 PMCID: PMC11120873 DOI: 10.3390/ijms25105072] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2024] [Revised: 05/03/2024] [Accepted: 05/03/2024] [Indexed: 05/26/2024] Open
Abstract
Vascular co-option is a consequence of the direct interaction between perivascular cells, known as pericytes (PCs), and glioblastoma multiforme (GBM) cells (GBMcs). This process is essential for inducing changes in the pericytes' anti-tumoral and immunoreactive phenotypes. Starting from the initial stages of carcinogenesis in GBM, PCs conditioned by GBMcs undergo proliferation, acquire a pro-tumoral and immunosuppressive phenotype by expressing and secreting immunosuppressive molecules, and significantly hinder the activation of T cells, thereby facilitating tumor growth. Inhibiting the pericyte (PC) conditioning mechanisms in the GBM tumor microenvironment (TME) results in immunological activation and tumor disappearance. This underscores the pivotal role of PCs as a key cell in the TME, responsible for tumor-induced immunosuppression and enabling GBM cells to evade the immune system. Other cells within the TME, such as tumor-associated macrophages (TAMs) and microglia, have also been identified as contributors to this immunomodulation. In this paper, we will review the role of these three cell types in the immunosuppressive properties of the TME. Our conclusion is that the cellular heterogeneity of immunocompetent cells within the TME may lead to the misinterpretation of cellular lineage identification due to different reactive stages and the identification of PCs as TAMs. Consequently, novel therapies could be developed to disrupt GBM-PC interactions and/or PC conditioning through vascular co-option, thereby exposing GBMcs to the immune system.
Collapse
Affiliation(s)
- Marta Martinez-Morga
- Instituto de Neurociencias, Universidad Miguel Hernández–CSIC, Excellence Center Severo Ochoa, Campus de San Juan, Avda. Ramón y Cajal sn, 03550 Alicante, Spain; (M.M.-M.); (D.G.); (E.R.-M.); (A.P.); (R.G.-L.)
| | - Daniel Garrigos
- Instituto de Neurociencias, Universidad Miguel Hernández–CSIC, Excellence Center Severo Ochoa, Campus de San Juan, Avda. Ramón y Cajal sn, 03550 Alicante, Spain; (M.M.-M.); (D.G.); (E.R.-M.); (A.P.); (R.G.-L.)
| | - Elena Rodriguez-Montero
- Instituto de Neurociencias, Universidad Miguel Hernández–CSIC, Excellence Center Severo Ochoa, Campus de San Juan, Avda. Ramón y Cajal sn, 03550 Alicante, Spain; (M.M.-M.); (D.G.); (E.R.-M.); (A.P.); (R.G.-L.)
| | - Ana Pombero
- Instituto de Neurociencias, Universidad Miguel Hernández–CSIC, Excellence Center Severo Ochoa, Campus de San Juan, Avda. Ramón y Cajal sn, 03550 Alicante, Spain; (M.M.-M.); (D.G.); (E.R.-M.); (A.P.); (R.G.-L.)
| | - Raquel Garcia-Lopez
- Instituto de Neurociencias, Universidad Miguel Hernández–CSIC, Excellence Center Severo Ochoa, Campus de San Juan, Avda. Ramón y Cajal sn, 03550 Alicante, Spain; (M.M.-M.); (D.G.); (E.R.-M.); (A.P.); (R.G.-L.)
| | - Salvador Martinez
- Instituto de Neurociencias, Universidad Miguel Hernández–CSIC, Excellence Center Severo Ochoa, Campus de San Juan, Avda. Ramón y Cajal sn, 03550 Alicante, Spain; (M.M.-M.); (D.G.); (E.R.-M.); (A.P.); (R.G.-L.)
- Centro de Investigación Biomédica en Red en Salud Mental, CIBERSAM-ISCIII, 46010 Valencia, Spain
| |
Collapse
|
38
|
Yan C, Yang Z, Chen P, Yeh Y, Sun C, Xie T, Huang W, Zhang X. GPR65 sensing tumor-derived lactate induces HMGB1 release from TAM via the cAMP/PKA/CREB pathway to promote glioma progression. J Exp Clin Cancer Res 2024; 43:105. [PMID: 38576043 PMCID: PMC10993467 DOI: 10.1186/s13046-024-03025-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2023] [Accepted: 03/23/2024] [Indexed: 04/06/2024] Open
Abstract
BACKGROUND Lactate has emerged as a critical regulator within the tumor microenvironment, including glioma. However, the precise mechanisms underlying how lactate influences the communication between tumor cells and tumor-associated macrophages (TAMs), the most abundant immune cells in glioma, remain poorly understood. This study aims to elucidate the impact of tumor-derived lactate on TAMs and investigate the regulatory pathways governing TAM-mediated tumor-promotion in glioma. METHODS Bioinformatic analysis was conducted using datasets from TCGA and CGGA. Single-cell RNA-seq datasets were analyzed by using UCSC Cell Browser and Single Cell Portal. Cell proliferation and mobility were evaluated through CCK8, colony formation, wound healing, and transwell assays. Western blot and immunofluorescence staining were applied to assess protein expression and cell distribution. RT-PCR and ELISA were employed to identify the potential secretory factors. Mechanistic pathways were explored by western blotting, ELISA, shRNA knockdown, and specific inhibitors and activators. The effects of pathway blockades were further assessed using subcutaneous and intracranial xenograft tumor models in vivo. RESULTS Elevated expressions of LDHA and MCT1 were observed in glioma and exhibited a positive correlation with M2-type TAM infiltration. Lactate derived from glioma cells induced TAMs towards M2-subtype polarization, subsequently promoting glioma cells proliferation, migration, invasion, and mesenchymal transition. GPR65, highly expressed on TAMs, sensed lactate-stimulation in the TME, fueling glioma cells malignant progression through the secretion of HMGB1. GPR65 on TAMs triggered HMGB1 release in response to lactate stimulation via the cAMP/PKA/CREB signaling pathway. Disrupting this feedback loop by GPR65-knockdown or HMGB1 inhibition mitigated glioma progression in vivo. CONCLUSION These findings unveil the intricate interplay between TAMs and tumor cells mediated by lactate and HMGB1, driving tumor progression in glioma. GPR65, selectively highly expressed on TAMs in glioma, sensed lactate stimulation and fostered HMGB1 secretion via the cAMP/PKA/CREB signaling pathway. Blocking this feedback loop presents a promising therapeutic strategy for GBM.
Collapse
Affiliation(s)
- Chaolong Yan
- Department of Neurosurgery, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Zijiang Yang
- Department of Neurosurgery, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Pin Chen
- Department of Neurosurgery, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Yuyang Yeh
- Department of Neurosurgery, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Chongjing Sun
- Department of Neurosurgery, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Tao Xie
- Department of Neurosurgery, Zhongshan Hospital, Fudan University, Shanghai, China.
| | - Wei Huang
- State Key Laboratory of Neuroscience, Institute of Neuroscience, Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, China.
| | - Xiaobiao Zhang
- Department of Neurosurgery, Zhongshan Hospital, Fudan University, Shanghai, China.
| |
Collapse
|
39
|
Redmer T, Schumann E, Peters K, Weidemeier ME, Nowak S, Schroeder HWS, Vidal A, Radbruch H, Lehmann A, Kreuzer-Redmer S, Jürchott K, Radke J. MET receptor serves as a promising target in melanoma brain metastases. Acta Neuropathol 2024; 147:44. [PMID: 38386085 PMCID: PMC10884227 DOI: 10.1007/s00401-024-02694-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2023] [Revised: 01/22/2024] [Accepted: 01/23/2024] [Indexed: 02/23/2024]
Abstract
The development of brain metastases hallmarks disease progression in 20-40% of melanoma patients and is a serious obstacle to therapy. Understanding the processes involved in the development and maintenance of melanoma brain metastases (MBM) is critical for the discovery of novel therapeutic strategies. Here, we generated transcriptome and methylome profiles of MBM showing high or low abundance of infiltrated Iba1high tumor-associated microglia and macrophages (TAMs). Our survey identified potential prognostic markers of favorable disease course and response to immune checkpoint inhibitor (ICi) therapy, among them APBB1IP and the interferon-responsive gene ITGB7. In MBM with high ITGB7/APBB1IP levels, the accumulation of TAMs correlated significantly with the immune score. Signature-based deconvolution of MBM via single sample GSEA revealed enrichment of interferon-response and immune signatures and revealed inflammation, stress and MET receptor signaling. MET receptor phosphorylation/activation maybe elicited by inflammatory processes in brain metastatic melanoma cells via stroma cell-released HGF. We found phospho-METY1234/1235 in a subset of MBM and observed a marked response of brain metastasis-derived cell lines (BMCs) that lacked druggable BRAF mutations or developed resistance to BRAF inhibitors (BRAFi) in vivo to MET inhibitors PHA-665752 and ARQ197 (tivantinib). In summary, the activation of MET receptor in brain colonizing melanoma cells by stromal cell-released HGF may promote tumor self-maintenance and expansion and might counteract ICi therapy. Therefore, therapeutic targeting of MET possibly serves as a promising strategy to control intracranial progressive disease and improve patient survival.
Collapse
Affiliation(s)
- Torben Redmer
- Institute for Medical Biochemistry, University of Veterinary Medicine Vienna, Vienna, Austria.
- Institute of Pathology, Unit of Laboratory Animal Pathology, University of Veterinary Medicine Vienna, Vienna, Austria.
| | - Elisa Schumann
- Department of Neuropathology, Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin and Berlin Institute of Health, Berlin, Germany
- German Cancer Consortium (DKTK), Partner Site Berlin, CCCC (Campus Mitte), Berlin, Germany
| | - Kristin Peters
- Institute of Pathology, University Medicine Greifswald, Greifswald, Germany
| | - Martin E Weidemeier
- Department of Neurosurgery, University Medicine Greifswald, Greifswald, Germany
| | - Stephan Nowak
- Department of Neurosurgery, University Medicine Greifswald, Greifswald, Germany
| | - Henry W S Schroeder
- Department of Neurosurgery, University Medicine Greifswald, Greifswald, Germany
| | - Anna Vidal
- Institute for Medical Biochemistry, University of Veterinary Medicine Vienna, Vienna, Austria
| | - Helena Radbruch
- Department of Neuropathology, Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin and Berlin Institute of Health, Berlin, Germany
| | - Annika Lehmann
- Institute of Pathology, Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin and Berlin Institute of Health, Berlin, Germany
| | - Susanne Kreuzer-Redmer
- Nutrigenomics Unit, Institute of Animal Nutrition and Functional Plant Compounds, University of Veterinary Medicine Vienna, Vienna, Austria
| | - Karsten Jürchott
- Center for Regenerative Therapies (BCRT), Berlin Institute of Health at Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Josefine Radke
- Institute of Pathology, University Medicine Greifswald, Greifswald, Germany.
| |
Collapse
|
40
|
Obrador E, Moreno-Murciano P, Oriol-Caballo M, López-Blanch R, Pineda B, Gutiérrez-Arroyo JL, Loras A, Gonzalez-Bonet LG, Martinez-Cadenas C, Estrela JM, Marqués-Torrejón MÁ. Glioblastoma Therapy: Past, Present and Future. Int J Mol Sci 2024; 25:2529. [PMID: 38473776 PMCID: PMC10931797 DOI: 10.3390/ijms25052529] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2023] [Revised: 02/10/2024] [Accepted: 02/16/2024] [Indexed: 03/14/2024] Open
Abstract
Glioblastoma (GB) stands out as the most prevalent and lethal form of brain cancer. Although great efforts have been made by clinicians and researchers, no significant improvement in survival has been achieved since the Stupp protocol became the standard of care (SOC) in 2005. Despite multimodality treatments, recurrence is almost universal with survival rates under 2 years after diagnosis. Here, we discuss the recent progress in our understanding of GB pathophysiology, in particular, the importance of glioma stem cells (GSCs), the tumor microenvironment conditions, and epigenetic mechanisms involved in GB growth, aggressiveness and recurrence. The discussion on therapeutic strategies first covers the SOC treatment and targeted therapies that have been shown to interfere with different signaling pathways (pRB/CDK4/RB1/P16ink4, TP53/MDM2/P14arf, PI3k/Akt-PTEN, RAS/RAF/MEK, PARP) involved in GB tumorigenesis, pathophysiology, and treatment resistance acquisition. Below, we analyze several immunotherapeutic approaches (i.e., checkpoint inhibitors, vaccines, CAR-modified NK or T cells, oncolytic virotherapy) that have been used in an attempt to enhance the immune response against GB, and thereby avoid recidivism or increase survival of GB patients. Finally, we present treatment attempts made using nanotherapies (nanometric structures having active anti-GB agents such as antibodies, chemotherapeutic/anti-angiogenic drugs or sensitizers, radionuclides, and molecules that target GB cellular receptors or open the blood-brain barrier) and non-ionizing energies (laser interstitial thermal therapy, high/low intensity focused ultrasounds, photodynamic/sonodynamic therapies and electroporation). The aim of this review is to discuss the advances and limitations of the current therapies and to present novel approaches that are under development or following clinical trials.
Collapse
Affiliation(s)
- Elena Obrador
- Scientia BioTech S.L., 46002 Valencia, Spain; (P.M.-M.); (M.O.-C.); (R.L.-B.); (J.M.E.)
- Department of Physiology, Faculty of Medicine and Odontology, University of Valencia, 46010 Valencia, Spain;
| | - Paz Moreno-Murciano
- Scientia BioTech S.L., 46002 Valencia, Spain; (P.M.-M.); (M.O.-C.); (R.L.-B.); (J.M.E.)
| | - María Oriol-Caballo
- Scientia BioTech S.L., 46002 Valencia, Spain; (P.M.-M.); (M.O.-C.); (R.L.-B.); (J.M.E.)
- Department of Physiology, Faculty of Medicine and Odontology, University of Valencia, 46010 Valencia, Spain;
| | - Rafael López-Blanch
- Scientia BioTech S.L., 46002 Valencia, Spain; (P.M.-M.); (M.O.-C.); (R.L.-B.); (J.M.E.)
- Department of Physiology, Faculty of Medicine and Odontology, University of Valencia, 46010 Valencia, Spain;
| | - Begoña Pineda
- Department of Physiology, Faculty of Medicine and Odontology, University of Valencia, 46010 Valencia, Spain;
| | - Julia Lara Gutiérrez-Arroyo
- Department of Medicine, Jaume I University of Castellon, 12071 Castellon, Spain; (J.L.G.-A.); (A.L.); (C.M.-C.)
| | - Alba Loras
- Department of Medicine, Jaume I University of Castellon, 12071 Castellon, Spain; (J.L.G.-A.); (A.L.); (C.M.-C.)
| | - Luis G. Gonzalez-Bonet
- Department of Neurosurgery, Castellon General University Hospital, 12004 Castellon, Spain;
| | - Conrado Martinez-Cadenas
- Department of Medicine, Jaume I University of Castellon, 12071 Castellon, Spain; (J.L.G.-A.); (A.L.); (C.M.-C.)
| | - José M. Estrela
- Scientia BioTech S.L., 46002 Valencia, Spain; (P.M.-M.); (M.O.-C.); (R.L.-B.); (J.M.E.)
- Department of Physiology, Faculty of Medicine and Odontology, University of Valencia, 46010 Valencia, Spain;
- Department of Physiology, Faculty of Pharmacy, University of Valencia, 46100 Burjassot, Spain
| | | |
Collapse
|
41
|
Kosianova А, Pak O, Bryukhovetskiy I. Regulation of cancer stem cells and immunotherapy of glioblastoma (Review). Biomed Rep 2024; 20:24. [PMID: 38170016 PMCID: PMC10758921 DOI: 10.3892/br.2023.1712] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2023] [Accepted: 11/24/2023] [Indexed: 01/05/2024] Open
Abstract
Glioblastoma (GB) is one of the most adverse diagnoses in oncology. Complex current treatment results in a median survival of 15 months. Resistance to treatment is associated with the presence of cancer stem cells (CSCs). The present review aimed to analyze the mechanisms of CSC plasticity, showing the particular role of β-catenin in regulating vital functions of CSCs, and to describe the molecular mechanisms of Wnt-independent increase of β-catenin levels, which is influenced by the local microenvironment of CSCs. The present review also analyzed the reasons for the low effectiveness of using medication in the regulation of CSCs, and proposed the development of immunotherapy scenarios with tumor cell vaccines, containing heterogenous cancer cells able of producing a multidirectional antineoplastic immune response. Additionally, the possibility of managing lymphopenia by transplanting hematopoietic stem cells from a healthy sibling and using clofazimine or other repurposed drugs that reduce β-catenin concentration in CSCs was discussed in the present study.
Collapse
Affiliation(s)
- Аleksandra Kosianova
- Medical Center, School of Medicine and Life Science, Far Eastern Federal University, Vladivostok 690091, Russian Federation
| | - Oleg Pak
- Medical Center, School of Medicine and Life Science, Far Eastern Federal University, Vladivostok 690091, Russian Federation
| | - Igor Bryukhovetskiy
- Medical Center, School of Medicine and Life Science, Far Eastern Federal University, Vladivostok 690091, Russian Federation
| |
Collapse
|
42
|
Lombardi F, Augello FR, Artone S, Ciafarone A, Topi S, Cifone MG, Cinque B, Palumbo P. Involvement of Cyclooxygenase-2 in Establishing an Immunosuppressive Microenvironment in Tumorspheres Derived from TMZ-Resistant Glioblastoma Cell Lines and Primary Cultures. Cells 2024; 13:258. [PMID: 38334650 PMCID: PMC10854914 DOI: 10.3390/cells13030258] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2023] [Revised: 01/26/2024] [Accepted: 01/28/2024] [Indexed: 02/10/2024] Open
Abstract
Glioblastoma (GBM) is characterized by an immunosuppressive tumor microenvironment (TME) strictly associated with therapy resistance. Cyclooxygenase-2 (COX-2) fuels GBM proliferation, stemness, and chemoresistance. We previously reported that COX-2 upregulation induced by temozolomide (TMZ) supported chemoresistance. Also, COX-2 transfer by extracellular vesicles released by T98G promoted M2 polarization in macrophages, whereas COX-2 inhibition counteracted these effects. Here, we investigated the COX-2 role in the stemness potential and modulation of the GBM immunosuppressive microenvironment. The presence of macrophages U937 within tumorspheres derived from GBM cell lines and primary cultures exposed to celecoxib (COX-2 inhibitor) with or without TMZ was studied by confocal microscopy. M2 polarization was analyzed by TGFβ-1 and CD206 levels. Osteopontin (OPN), a crucial player within the TME by driving the macrophages' infiltration, and CD44 expression was assessed by Western blot. TMZ strongly enhanced tumorsphere size and induced the M2 polarization of infiltrating macrophages. In macrophage-infiltrated tumorspheres, TMZ upregulated OPN and CD44 expression. These TMZ effects were counteracted by the concurrent addition of CXB. Remarkably, exogenous prostaglandin-E2 restored OPN and CD44, highlighting the COX-2 pivotal role in the protumor macrophages' state promotion. COX-2 inhibition interfered with TMZ's ability to induce M2-polarization and counteracted the development of an immunosuppressive TME.
Collapse
Affiliation(s)
- Francesca Lombardi
- Department of Life, Health and Environmental Sciences, University of L’Aquila, 67100 L’Aquila, Italy; (F.L.); (F.R.A.); (M.G.C.); (B.C.)
| | - Francesca Rosaria Augello
- Department of Life, Health and Environmental Sciences, University of L’Aquila, 67100 L’Aquila, Italy; (F.L.); (F.R.A.); (M.G.C.); (B.C.)
| | - Serena Artone
- PhD School in Medicine and Public Health, Department of Life, Health and Environmental Sciences, University of L’Aquila, 67100 L’Aquila, Italy;
| | - Alessia Ciafarone
- PhD School in Health & Environmental Sciences, Department of Life, Health and Environmental Sciences, University of L’Aquila, 67100 L’Aquila, Italy;
| | - Skender Topi
- Department of Clinical Disciplines, Aleksandër Xhuvani University, 3001 Elbasan, Albania;
| | - Maria Grazia Cifone
- Department of Life, Health and Environmental Sciences, University of L’Aquila, 67100 L’Aquila, Italy; (F.L.); (F.R.A.); (M.G.C.); (B.C.)
| | - Benedetta Cinque
- Department of Life, Health and Environmental Sciences, University of L’Aquila, 67100 L’Aquila, Italy; (F.L.); (F.R.A.); (M.G.C.); (B.C.)
| | - Paola Palumbo
- Department of Life, Health and Environmental Sciences, University of L’Aquila, 67100 L’Aquila, Italy; (F.L.); (F.R.A.); (M.G.C.); (B.C.)
| |
Collapse
|
43
|
Mishchenko TA, Turubanova VD, Gorshkova EN, Krysko O, Vedunova MV, Krysko DV. Glioma: bridging the tumor microenvironment, patient immune profiles and novel personalized immunotherapy. Front Immunol 2024; 14:1299064. [PMID: 38274827 PMCID: PMC10809268 DOI: 10.3389/fimmu.2023.1299064] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2023] [Accepted: 12/11/2023] [Indexed: 01/27/2024] Open
Abstract
Glioma is the most common primary brain tumor, characterized by a consistently high patient mortality rate and a dismal prognosis affecting both survival and quality of life. Substantial evidence underscores the vital role of the immune system in eradicating tumors effectively and preventing metastasis, underscoring the importance of cancer immunotherapy which could potentially address the challenges in glioma therapy. Although glioma immunotherapies have shown promise in preclinical and early-phase clinical trials, they face specific limitations and challenges that have hindered their success in further phase III trials. Resistance to therapy has been a major challenge across many experimental approaches, and as of now, no immunotherapies have been approved. In addition, there are several other limitations facing glioma immunotherapy in clinical trials, such as high intra- and inter-tumoral heterogeneity, an inherently immunosuppressive microenvironment, the unique tissue-specific interactions between the central nervous system and the peripheral immune system, the existence of the blood-brain barrier, which is a physical barrier to drug delivery, and the immunosuppressive effects of standard therapy. Therefore, in this review, we delve into several challenges that need to be addressed to achieve boosted immunotherapy against gliomas. First, we discuss the hurdles posed by the glioma microenvironment, particularly its primary cellular inhabitants, in particular tumor-associated microglia and macrophages (TAMs), and myeloid cells, which represent a significant barrier to effective immunotherapy. Here we emphasize the impact of inducing immunogenic cell death (ICD) on the migration of Th17 cells into the tumor microenvironment, converting it into an immunologically "hot" environment and enhancing the effectiveness of ongoing immunotherapy. Next, we address the challenge associated with the accurate identification and characterization of the primary immune profiles of gliomas, and their implications for patient prognosis, which can facilitate the selection of personalized treatment regimens and predict the patient's response to immunotherapy. Finally, we explore a prospective approach to developing highly personalized vaccination strategies against gliomas, based on the search for patient-specific neoantigens. All the pertinent challenges discussed in this review will serve as a compass for future developments in immunotherapeutic strategies against gliomas, paving the way for upcoming preclinical and clinical research endeavors.
Collapse
Affiliation(s)
- Tatiana A. Mishchenko
- Institute of Biology and Biomedicine, National Research Lobachevsky State University of Nizhny Novgorod, Nizhny Novgorod, Russia
| | - Victoria D. Turubanova
- Institute of Biology and Biomedicine, National Research Lobachevsky State University of Nizhny Novgorod, Nizhny Novgorod, Russia
- Neuroscience Research Institute, National Research Lobachevsky State University of Nizhny Novgorod, Nizhny Novgorod, Russia
| | - Ekaterina N. Gorshkova
- Institute of Biology and Biomedicine, National Research Lobachevsky State University of Nizhny Novgorod, Nizhny Novgorod, Russia
| | - Olga Krysko
- Cell Death Investigation and Therapy Laboratory, Anatomy and Embryology Unit, Department of Human Structure and Repair, Faculty of Medicine and Health Sciences, Ghent University, Ghent, Belgium
| | - Maria V. Vedunova
- Institute of Biology and Biomedicine, National Research Lobachevsky State University of Nizhny Novgorod, Nizhny Novgorod, Russia
- Faculty of Biology and Biotechnologies, National Research University Higher School of Economics, Moscow, Russia
| | - Dmitri V. Krysko
- Institute of Biology and Biomedicine, National Research Lobachevsky State University of Nizhny Novgorod, Nizhny Novgorod, Russia
- Cell Death Investigation and Therapy Laboratory, Anatomy and Embryology Unit, Department of Human Structure and Repair, Faculty of Medicine and Health Sciences, Ghent University, Ghent, Belgium
- Department of Pathophysiology, Sechenov First Moscow State Medical University (Sechenov University), Moscow, Russia
- Cancer Research Institute Ghent, Ghent, Belgium
| |
Collapse
|
44
|
McKean WB, Yang J, Boucher K, Shrieve DC, Suneja G, Salzman K, Jensen R, Colman H, Cohen AL. D-TERMINED, a phase 1 trial in newly diagnosed high-grade glioma with temozolomide, radiation, and minocycline followed by adjuvant minocycline/temozolomide. Neurooncol Adv 2024; 6:vdae063. [PMID: 38800698 PMCID: PMC11125402 DOI: 10.1093/noajnl/vdae063] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/29/2024] Open
Abstract
Background Standard treatment for newly diagnosed high-grade gliomas remains suboptimal. Preclinical data indicate that mesenchymal transition and radiation resistance in glioblastoma are driven by NF-κB and microglia activation, which can be inhibited by minocycline. We assessed the safety and efficacy of minocycline combined with standard radiation and temozolomide in newly diagnosed high-grade gliomas. Methods Adults with newly diagnosed high-grade glioma were eligible. Minocycline was given with concurrent and adjuvant temozolomide. Minocycline doses were escalated using a 3 + 3 design and expanded to identify the maximum tolerated dose (MTD) and adverse event profile. Individual progression-free survival (PFS) was compared to predicted PFS based on RTOG RPA class using a binomial test. The relationships between mesenchymal and microglial biomarkers were analyzed with immunohistochemistry. Results The MTD of minocycline was 150 mg twice per day (N = 20); 1 patient (5%) experienced CTCAE grade 3 + nausea and dizziness, and 2 patients (10%) demonstrated thrombocytopenia requiring temozolomide interruptions. Twelve patients exceeded their predicted PFS (60%), which did not meet the predefined efficacy endpoint of 70%. Symptoms increased during post-radiation treatment but remained mild. No significant correlation was seen between biomarkers and PFS. Expression levels of P-p65, a marker of NF-κB activation, were correlated with the microglia marker IBA-1. Conclusions Minocycline at 150 mg twice per day is well tolerated with standard chemoradiation in patients with newly diagnosed high-grade gliomas. PFS was not significantly increased with the addition of minocycline when compared to historical controls. NF-κB activation correlates with microglia levels in high-grade glioma.
Collapse
Affiliation(s)
- William B McKean
- Huntsman Cancer Institute, University of Utah, Salt Lake City, Utah, USA
- Division of Medical Oncology, University of Utah School of Medicine, Salt Lake City, Utah, USA
| | - Jingye Yang
- Huntsman Cancer Institute, University of Utah, Salt Lake City, Utah, USA
- Department of Neurosurgery, University of Utah School of Medicine, Salt Lake City, Utah, USA
| | - Kenneth Boucher
- Huntsman Cancer Institute, University of Utah, Salt Lake City, Utah, USA
- Division of Medical Oncology, University of Utah School of Medicine, Salt Lake City, Utah, USA
- Department of Epidemiology, University of Utah School of Medicine, Salt Lake City, Utah, USA
| | - Dennis C Shrieve
- Huntsman Cancer Institute, University of Utah, Salt Lake City, Utah, USA
- Department of Radiation Oncology, University of Utah School of Medicine, Salt Lake City, Utah, USA
| | - Gita Suneja
- Huntsman Cancer Institute, University of Utah, Salt Lake City, Utah, USA
- Department of Radiation Oncology, University of Utah School of Medicine, Salt Lake City, Utah, USA
| | - Karen Salzman
- Huntsman Cancer Institute, University of Utah, Salt Lake City, Utah, USA
- Department of Radiology, University of Utah School of Medicine, Salt Lake City, Utah, USA
| | - Randy Jensen
- Huntsman Cancer Institute, University of Utah, Salt Lake City, Utah, USA
- Department of Neurosurgery, University of Utah School of Medicine, Salt Lake City, Utah, USA
| | - Howard Colman
- Huntsman Cancer Institute, University of Utah, Salt Lake City, Utah, USA
- Department of Neurosurgery, University of Utah School of Medicine, Salt Lake City, Utah, USA
| | - Adam L Cohen
- Huntsman Cancer Institute, University of Utah, Salt Lake City, Utah, USA
- Division of Medical Oncology, University of Utah School of Medicine, Salt Lake City, Utah, USA
- Division of Oncology, Inova Schar Cancer Institute, Fairfax, Virginia, USA
| |
Collapse
|
45
|
Laureano RS, Vanmeerbeek I, Sprooten J, Govaerts J, Naulaerts S, Garg AD. The cell stress and immunity cycle in cancer: Toward next generation of cancer immunotherapy. Immunol Rev 2024; 321:71-93. [PMID: 37937803 DOI: 10.1111/imr.13287] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2023] [Revised: 10/05/2023] [Accepted: 10/20/2023] [Indexed: 11/09/2023]
Abstract
The cellular stress and immunity cycle is a cornerstone of organismal homeostasis. Stress activates intracellular and intercellular communications within a tissue or organ to initiate adaptive responses aiming to resolve the origin of this stress. If such local measures are unable to ameliorate this stress, then intercellular communications expand toward immune activation with the aim of recruiting immune cells to effectively resolve the situation while executing tissue repair to ameliorate any damage and facilitate homeostasis. This cellular stress-immunity cycle is severely dysregulated in diseased contexts like cancer. On one hand, cancer cells dysregulate the normal cellular stress responses to reorient them toward upholding growth at all costs, even at the expense of organismal integrity and homeostasis. On the other hand, the tumors severely dysregulate or inhibit various components of organismal immunity, for example, by facilitating immunosuppressive tumor landscape, lowering antigenicity, and increasing T-cell dysfunction. In this review we aim to comprehensively discuss the basis behind tumoral dysregulation of cellular stress-immunity cycle. We also offer insights into current understanding of the regulators and deregulators of this cycle and how they can be targeted for conceptualizing successful cancer immunotherapy regimen.
Collapse
Affiliation(s)
- Raquel S Laureano
- Cell Stress & Immunity (CSI) Lab, Department of Cellular and Molecular Medicine, KU Leuven, Leuven, Belgium
| | - Isaure Vanmeerbeek
- Cell Stress & Immunity (CSI) Lab, Department of Cellular and Molecular Medicine, KU Leuven, Leuven, Belgium
| | - Jenny Sprooten
- Cell Stress & Immunity (CSI) Lab, Department of Cellular and Molecular Medicine, KU Leuven, Leuven, Belgium
| | - Jannes Govaerts
- Cell Stress & Immunity (CSI) Lab, Department of Cellular and Molecular Medicine, KU Leuven, Leuven, Belgium
| | - Stefan Naulaerts
- Cell Stress & Immunity (CSI) Lab, Department of Cellular and Molecular Medicine, KU Leuven, Leuven, Belgium
| | - Abhishek D Garg
- Cell Stress & Immunity (CSI) Lab, Department of Cellular and Molecular Medicine, KU Leuven, Leuven, Belgium
| |
Collapse
|
46
|
von Roemeling C, Yegorov O, Yang C, Klippel K, Russell R, Trivedi V, Bhatia A, Doonan B, Carpenter S, Ryu D, Grippen A, Futch H, Ran Y, Hoang-Minh L, Weidert F, Golde T, Mitchell D. CXCL9 recombinant adeno-associated virus (AAV) virotherapy sensitizes glioblastoma (GBM) to anti-PD-1 immune checkpoint blockade. RESEARCH SQUARE 2023:rs.3.rs-3463730. [PMID: 38014191 PMCID: PMC10680939 DOI: 10.21203/rs.3.rs-3463730/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/29/2023]
Abstract
The promise of immunotherapy to induce long-term durable responses in conventionally treatment resistant tumors like glioblastoma (GBM) has given hope for patients with a dismal prognosis. Yet, few patients have demonstrated a significant survival benefit despite multiple clinical trials designed to invigorate immune recognition and tumor eradication. Insights gathered over the last two decades have revealed numerous mechanisms by which glioma cells resist conventional therapy and evade immunological detection, underscoring the need for strategic combinatorial treatments as necessary to achieve appreciable therapeutic effects. However, new combination therapies are inherently difficult to develop as a result of dose-limiting toxicities, the constraints of the blood-brain barrier, and the suppressive nature of the GBM tumor microenvironment (TME). GBM is notoriously devoid of lymphocytes driven in part by a paucity of lymphocyte trafficking factors necessary to prompt their recruitment, infiltration, and activation. We have developed a novel recombinant adeno-associated virus (AAV) gene therapy strategy that enables focal and stable reconstitution of the GBM TME with C-X-C motif ligand 9 (CXCL9), a powerful call-and-receive chemokine for cytotoxic T lymphocytes (CTLs). By precisely manipulating local chemokine directional guidance, AAV-CXCL9 increases tumor infiltration by CD8-postive cytotoxic lymphocytes, sensitizing GBM to anti-PD-1 immune checkpoint blockade (ICB). These effects are accompanied by immunologic signatures evocative of an inflamed and responsive TME. These findings support targeted AAV gene therapy as a promising adjuvant strategy for reconditioning GBM immunogenicity given its excellent safety profile, TME-tropism, modularity, and off-the-shelf capability, where focal delivery bypasses the constrains of the blood-brain barrier, further mitigating risks observed with high-dose systemic therapy.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | | | | | | | | | - Frances Weidert
- Department of Neurosurgery, Preston A. Wells, Jr. Center for Brain Tumor Therapy, University of Florida
| | | | | |
Collapse
|
47
|
Finotto L, Cole B, Giese W, Baumann E, Claeys A, Vanmechelen M, Decraene B, Derweduwe M, Dubroja Lakic N, Shankar G, Nagathihalli Kantharaju M, Albrecht JP, Geudens I, Stanchi F, Ligon KL, Boeckx B, Lambrechts D, Harrington K, Van Den Bosch L, De Vleeschouwer S, De Smet F, Gerhardt H. Single-cell profiling and zebrafish avatars reveal LGALS1 as immunomodulating target in glioblastoma. EMBO Mol Med 2023; 15:e18144. [PMID: 37791581 PMCID: PMC10630887 DOI: 10.15252/emmm.202318144] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2023] [Revised: 08/29/2023] [Accepted: 09/04/2023] [Indexed: 10/05/2023] Open
Abstract
Glioblastoma (GBM) remains the most malignant primary brain tumor, with a median survival rarely exceeding 2 years. Tumor heterogeneity and an immunosuppressive microenvironment are key factors contributing to the poor response rates of current therapeutic approaches. GBM-associated macrophages (GAMs) often exhibit immunosuppressive features that promote tumor progression. However, their dynamic interactions with GBM tumor cells remain poorly understood. Here, we used patient-derived GBM stem cell cultures and combined single-cell RNA sequencing of GAM-GBM co-cultures and real-time in vivo monitoring of GAM-GBM interactions in orthotopic zebrafish xenograft models to provide insight into the cellular, molecular, and spatial heterogeneity. Our analyses revealed substantial heterogeneity across GBM patients in GBM-induced GAM polarization and the ability to attract and activate GAMs-features that correlated with patient survival. Differential gene expression analysis, immunohistochemistry on original tumor samples, and knock-out experiments in zebrafish subsequently identified LGALS1 as a primary regulator of immunosuppression. Overall, our work highlights that GAM-GBM interactions can be studied in a clinically relevant way using co-cultures and avatar models, while offering new opportunities to identify promising immune-modulating targets.
Collapse
Affiliation(s)
- Lise Finotto
- Max Delbrück Center for Molecular Medicine in the Helmholtz AssociationBerlinGermany
- VIB ‐ KU Leuven Center for Cancer BiologyVIB ‐ KU LeuvenLeuvenBelgium
- The Laboratory for Precision Cancer Medicine, Translational Cell and Tissue Research Unit, Department of Imaging & PathologyKU LeuvenLeuvenBelgium
- KU Leuven Institute for Single Cell Omics (LISCO)KU LeuvenLeuvenBelgium
| | - Basiel Cole
- The Laboratory for Precision Cancer Medicine, Translational Cell and Tissue Research Unit, Department of Imaging & PathologyKU LeuvenLeuvenBelgium
- KU Leuven Institute for Single Cell Omics (LISCO)KU LeuvenLeuvenBelgium
| | - Wolfgang Giese
- Max Delbrück Center for Molecular Medicine in the Helmholtz AssociationBerlinGermany
- DZHK (German Center for Cardiovascular Research), Partner Site BerlinBerlinGermany
| | - Elisabeth Baumann
- Max Delbrück Center for Molecular Medicine in the Helmholtz AssociationBerlinGermany
- Charité ‐ Universitätsmedizin BerlinBerlinGermany
| | - Annelies Claeys
- The Laboratory for Precision Cancer Medicine, Translational Cell and Tissue Research Unit, Department of Imaging & PathologyKU LeuvenLeuvenBelgium
- KU Leuven Institute for Single Cell Omics (LISCO)KU LeuvenLeuvenBelgium
| | - Maxime Vanmechelen
- The Laboratory for Precision Cancer Medicine, Translational Cell and Tissue Research Unit, Department of Imaging & PathologyKU LeuvenLeuvenBelgium
- KU Leuven Institute for Single Cell Omics (LISCO)KU LeuvenLeuvenBelgium
- Department of Medical OncologyUniversity Hospitals LeuvenLeuvenBelgium
| | - Brecht Decraene
- The Laboratory for Precision Cancer Medicine, Translational Cell and Tissue Research Unit, Department of Imaging & PathologyKU LeuvenLeuvenBelgium
- KU Leuven Institute for Single Cell Omics (LISCO)KU LeuvenLeuvenBelgium
- Laboratory of Experimental Neurosurgery and Neuroanatomy, Department of Neurosciences, KU Leuven & Leuven Brain Institute (LBI)KU LeuvenLeuvenBelgium
- Department of NeurosurgeryUniversity Hospitals LeuvenLeuvenBelgium
| | - Marleen Derweduwe
- The Laboratory for Precision Cancer Medicine, Translational Cell and Tissue Research Unit, Department of Imaging & PathologyKU LeuvenLeuvenBelgium
- KU Leuven Institute for Single Cell Omics (LISCO)KU LeuvenLeuvenBelgium
| | - Nikolina Dubroja Lakic
- The Laboratory for Precision Cancer Medicine, Translational Cell and Tissue Research Unit, Department of Imaging & PathologyKU LeuvenLeuvenBelgium
- KU Leuven Institute for Single Cell Omics (LISCO)KU LeuvenLeuvenBelgium
| | - Gautam Shankar
- The Laboratory for Precision Cancer Medicine, Translational Cell and Tissue Research Unit, Department of Imaging & PathologyKU LeuvenLeuvenBelgium
- KU Leuven Institute for Single Cell Omics (LISCO)KU LeuvenLeuvenBelgium
| | - Madhu Nagathihalli Kantharaju
- Max Delbrück Center for Molecular Medicine in the Helmholtz AssociationBerlinGermany
- Humboldt University of BerlinBerlinGermany
| | - Jan Philipp Albrecht
- Max Delbrück Center for Molecular Medicine in the Helmholtz AssociationBerlinGermany
- Humboldt University of BerlinBerlinGermany
| | - Ilse Geudens
- VIB ‐ KU Leuven Center for Cancer BiologyVIB ‐ KU LeuvenLeuvenBelgium
| | - Fabio Stanchi
- VIB ‐ KU Leuven Center for Cancer BiologyVIB ‐ KU LeuvenLeuvenBelgium
| | - Keith L Ligon
- Center for Neuro‐oncologyDana‐Farber Cancer InstituteBostonMAUSA
- Department of PathologyBrigham and Women's HospitalBostonMAUSA
- Department of PathologyHarvard Medical SchoolBostonMAUSA
| | - Bram Boeckx
- VIB ‐ KU Leuven Center for Cancer BiologyVIB ‐ KU LeuvenLeuvenBelgium
- KU Leuven Institute for Single Cell Omics (LISCO)KU LeuvenLeuvenBelgium
- Laboratory of Translational Genetics, Department of Human GeneticsKU LeuvenLeuvenBelgium
| | - Diether Lambrechts
- VIB ‐ KU Leuven Center for Cancer BiologyVIB ‐ KU LeuvenLeuvenBelgium
- KU Leuven Institute for Single Cell Omics (LISCO)KU LeuvenLeuvenBelgium
- Laboratory of Translational Genetics, Department of Human GeneticsKU LeuvenLeuvenBelgium
| | - Kyle Harrington
- Max Delbrück Center for Molecular Medicine in the Helmholtz AssociationBerlinGermany
- Chan Zuckerberg InitiativeRedwood CityCAUSA
| | - Ludo Van Den Bosch
- Laboratory of Neurobiology, Department of Neurosciences, Experimental Neurology & Leuven Brain Institute (LBI)KU LeuvenLeuvenBelgium
- VIB ‐ KU Leuven Center for Brain & Disease Research, Laboratory of NeurobiologyVIB ‐ KU LeuvenLeuvenBelgium
| | - Steven De Vleeschouwer
- KU Leuven Institute for Single Cell Omics (LISCO)KU LeuvenLeuvenBelgium
- Laboratory of Experimental Neurosurgery and Neuroanatomy, Department of Neurosciences, KU Leuven & Leuven Brain Institute (LBI)KU LeuvenLeuvenBelgium
- Department of NeurosurgeryUniversity Hospitals LeuvenLeuvenBelgium
| | - Frederik De Smet
- The Laboratory for Precision Cancer Medicine, Translational Cell and Tissue Research Unit, Department of Imaging & PathologyKU LeuvenLeuvenBelgium
- KU Leuven Institute for Single Cell Omics (LISCO)KU LeuvenLeuvenBelgium
| | - Holger Gerhardt
- Max Delbrück Center for Molecular Medicine in the Helmholtz AssociationBerlinGermany
- DZHK (German Center for Cardiovascular Research), Partner Site BerlinBerlinGermany
- Charité ‐ Universitätsmedizin BerlinBerlinGermany
- Berlin Institute of HealthBerlinGermany
| |
Collapse
|
48
|
Marangon D, Lecca D. Exosomal non-coding RNAs in glioma progression: insights into tumor microenvironment dynamics and therapeutic implications. Front Cell Dev Biol 2023; 11:1275755. [PMID: 38020906 PMCID: PMC10646304 DOI: 10.3389/fcell.2023.1275755] [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: 08/10/2023] [Accepted: 10/16/2023] [Indexed: 12/01/2023] Open
Abstract
Gliomas are the most common and deadly types of brain tumors, known for their extensive genetic and epigenetic variability, which poses considerable challenges for pharmacological treatment. Glioma heterogeneity is also related to their intricate and dynamic tumor microenvironment (TME), which comprises a diverse array of cell types, including immune cells, vascular cells, glial cells, and neural precursors, collectively influencing tumor behavior and progression. A pivotal aspect of this intercellular communication relies on the exchange of extracellular vesicles (EVs), which contain and transfer complex molecular cargoes typical of their cells of origin, such as proteins, lipids, carbohydrates, metabolites, and non-coding RNAs (ncRNAs), that encompass microRNAs (miRNAs), long non-coding RNAs (lncRNAs), and circular RNAs (circRNAs). Glioma cells actively release EVs loaded with specific ncRNAs that can target genes and other ncRNAs in recipient cells residing within the TME. Among these recipient cells, prominent players include tumor-associated macrophages and microglia (TAMs), non-neoplastic astrocytes and endothelial cells. The intricate interplay between EVs derived from glioma cells and these recipient cells significantly contributes to the establishment of a tumor-permissive microenvironment, promoting tumor cell proliferation, migration, angiogenesis, and invasion, by targeting various downstream pathways. This review critically examines the current understanding of the intricate interplay between glioma, exosomal ncRNAs, and various components of the glioma TME. By shedding light on the roles of ncRNAs in mediating intercellular communication, this review underscores their significance in orchestrating TME transformation and highlights their potential as novel therapeutic targets for effectively tackling glioma progression.
Collapse
Affiliation(s)
- Davide Marangon
- Laboratory of Molecular and Cellular Pharmacology of Purinergic Transmission, Department of Pharmaceutical Sciences, Università degli Studi di Milano, Milan, Italy
| | | |
Collapse
|
49
|
Irshad K, Huang YK, Rodriguez P, Lo J, Aghoghovwia BE, Pan Y, Chang KC. The Neuroimmune Regulation and Potential Therapeutic Strategies of Optic Pathway Glioma. Brain Sci 2023; 13:1424. [PMID: 37891793 PMCID: PMC10605541 DOI: 10.3390/brainsci13101424] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2023] [Revised: 09/28/2023] [Accepted: 10/03/2023] [Indexed: 10/29/2023] Open
Abstract
Optic pathway glioma (OPG) is one of the causes of pediatric visual impairment. Unfortunately, there is as yet no cure for such a disease. Understanding the underlying mechanisms and the potential therapeutic strategies may help to delay the progression of OPG and rescue the visual morbidities. Here, we provide an overview of preclinical OPG studies and the regulatory pathways controlling OPG pathophysiology. We next discuss the role of microenvironmental cells (neurons, T cells, and tumor-associated microglia and macrophages) in OPG development. Last, we provide insight into potential therapeutic strategies for treating OPG and promoting axon regeneration.
Collapse
Affiliation(s)
- Khushboo Irshad
- Department of Symptom Research, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA; (K.I.); (B.E.A.)
| | - Yu-Kai Huang
- Division of Neurosurgery, Department of Surgery, Kaohsiung Medical University Hospital, Kaohsiung 80708, Taiwan;
- Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 80708, Taiwan
| | - Paul Rodriguez
- Department of Ophthalmology, Louis J. Fox Center for Vision Restoration, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA;
| | - Jung Lo
- Department of Ophthalmology, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung 83301, Taiwan;
| | - Benjamin E. Aghoghovwia
- Department of Symptom Research, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA; (K.I.); (B.E.A.)
| | - Yuan Pan
- Department of Symptom Research, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA; (K.I.); (B.E.A.)
- Department of Neuro-Oncology, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Kun-Che Chang
- Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 80708, Taiwan
- Department of Ophthalmology, Louis J. Fox Center for Vision Restoration, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA;
- Department of Neurobiology, Center of Neuroscience, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA
| |
Collapse
|
50
|
Vincent CA, Nissen I, Dakhel S, Hörnblad A, Remeseiro S. Epigenomic perturbation of novel EGFR enhancers reduces the proliferative and invasive capacity of glioblastoma and increases sensitivity to temozolomide. BMC Cancer 2023; 23:945. [PMID: 37803333 PMCID: PMC10557167 DOI: 10.1186/s12885-023-11418-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2023] [Accepted: 09/18/2023] [Indexed: 10/08/2023] Open
Abstract
BACKGROUND Glioblastoma (GB) is the most aggressive of all primary brain tumours and due to its highly invasive nature, surgical resection is nearly impossible. Patients typically rely on radiotherapy with concurrent temozolomide (TMZ) treatment and face a median survival of ~ 14 months. Alterations in the Epidermal Growth Factor Receptor gene (EGFR) are common in GB tumours, but therapies targeting EGFR have not shown significant clinical efficacy. METHODS Here, we investigated the influence of the EGFR regulatory genome on GB cells and identified novel EGFR enhancers located near the GB-associated SNP rs723527. We used CRISPR/Cas9-based approaches to target the EGFR enhancer regions, generating multiple modified GB cell lines, which enabled us to study the functional response to enhancer perturbation. RESULTS Epigenomic perturbation of the EGFR regulatory region decreases EGFR expression and reduces the proliferative and invasive capacity of glioblastoma cells, which also undergo a metabolic reprogramming in favour of mitochondrial respiration and present increased apoptosis. Moreover, EGFR enhancer-perturbation increases the sensitivity of GB cells to TMZ, the first-choice chemotherapeutic agent to treat glioblastoma. CONCLUSIONS Our findings demonstrate how epigenomic perturbation of EGFR enhancers can ameliorate the aggressiveness of glioblastoma cells and enhance the efficacy of TMZ treatment. This study demonstrates how CRISPR/Cas9-based perturbation of enhancers can be used to modulate the expression of key cancer genes, which can help improve the effectiveness of existing cancer treatments and potentially the prognosis of difficult-to-treat cancers such as glioblastoma.
Collapse
Affiliation(s)
- Craig A Vincent
- Umeå Centre for Molecular Medicine (UCMM), Umeå University, Umeå, Sweden
- Wallenberg Centre for Molecular Medicine (WCMM), Umeå University, Umeå, Sweden
| | - Itzel Nissen
- Umeå Centre for Molecular Medicine (UCMM), Umeå University, Umeå, Sweden
- Wallenberg Centre for Molecular Medicine (WCMM), Umeå University, Umeå, Sweden
| | - Soran Dakhel
- Umeå Centre for Molecular Medicine (UCMM), Umeå University, Umeå, Sweden
- Wallenberg Centre for Molecular Medicine (WCMM), Umeå University, Umeå, Sweden
| | - Andreas Hörnblad
- Umeå Centre for Molecular Medicine (UCMM), Umeå University, Umeå, Sweden
| | - Silvia Remeseiro
- Umeå Centre for Molecular Medicine (UCMM), Umeå University, Umeå, Sweden.
- Wallenberg Centre for Molecular Medicine (WCMM), Umeå University, Umeå, Sweden.
| |
Collapse
|