1
|
Vetsika EK, Katsianou MA, Sarantis P, Palamaris K, Papavassiliou AG, Piperi C. Pediatric gliomas immunity challenges and immunotherapy advances. Cancer Lett 2025; 618:217640. [PMID: 40090572 DOI: 10.1016/j.canlet.2025.217640] [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: 02/11/2025] [Revised: 03/11/2025] [Accepted: 03/12/2025] [Indexed: 03/18/2025]
Abstract
Pediatric gliomas, the most frequent brain tumors in children, are characterized by heterogeneity and a unique tumor immune microenvironment. They are categorized into different subtypes, including low-grade gliomas like pilocytic astrocytomas and high-grade gliomas such as diffuse midline gliomas and diffuse intrinsic pontine gliomas, each exhibiting distinct immunological profiles. The tumor immune microenvironment in pediatric gliomas is shaped by cellular and non-cellular components, including immune cells, cytokines, and the extracellular matrix, involved in tumor progression, immune evasion, and response to therapy. While pediatric low-grade gliomas often display an immunosuppressed microenvironment, high-grade gliomas are characterized by complex immune infiltrates and intricate immunosuppressive mechanisms. The blood-brain barrier further obscures immune cell recruitment and therapeutic delivery. Despite advances in understanding adult gliomas, the immunobiology of pediatric tumors is poorly investigated, with limited data on the interactions between glioma cells and immune populations such as T and natural killer cells, as well as tumor-associated macrophages. Herein, we provide an update of the current knowledge on tumor immune microenvironment interactions in pediatric gliomas, highlighting the immunosuppressive mechanisms and emerging immunotherapeutic strategies aiming at overcoming these barriers to improve clinical outcomes for affected children.
Collapse
Affiliation(s)
- Eleni-Kyriaki Vetsika
- Centre of New Biotechnologies and Precision Medicine (CNBPM), School of Medicine, National and Kapodistrian University of Athens, Athens, Greece
| | - Maria A Katsianou
- Department of Biological Chemistry, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | - Panagiotis Sarantis
- Department of Biological Chemistry, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | - Kostas Palamaris
- First Department of Pathology, School of Medicine, National and Kapodistrian University of Athens, 10679, Athens, Greece
| | - Athanasios G Papavassiliou
- Department of Biological Chemistry, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | - Christina Piperi
- Department of Biological Chemistry, Medical School, National and Kapodistrian University of Athens, Athens, Greece.
| |
Collapse
|
2
|
Maity S, Jewell C, Yilgor C, Kawakita S, Sharma S, Gomez A, Mecwan M, Falcone N, Ermis M, Monirizad M, Kouchehbaghi NH, Zehtabi F, Khorsandi D, Dokmeci MR, Moniz-Garcia D, Quiñones-Hinojosa A, Khademhosseini A, Jucaud V. Deciphering pericyte-induced temozolomide resistance in glioblastoma with a 3D microphysiological system mimicking the biomechanical properties of brain tissue. Acta Biomater 2025:S1742-7061(25)00363-0. [PMID: 40383349 DOI: 10.1016/j.actbio.2025.05.038] [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: 11/27/2024] [Revised: 04/29/2025] [Accepted: 05/14/2025] [Indexed: 05/20/2025]
Abstract
Glioblastoma (GBM) is a highly aggressive malignancy with a poor prognosis and frequent resistance to temozolomide (TMZ), the standard-of-care chemotherapy. The complex mechanisms underlying GBM chemoresistance, particularly the role of pericytes, remain poorly understood due to the lack of physiologically relevant in vitro models replicating the complex tumor microenvironment (TME). Here, we present a biomimetic 3D GBM microphysiological system that replicates the biomechanical properties of brain tissue (G'∼800Pa, G"∼100Pa) and enables the study of pericyte-mediated TMZ resistance. GBM spheroids (U87, LN229, PDM140) were cultured alone or co-cultured with pericytes in a composite hydrogel for 14 days and remained viable and proliferative. In response to TMZ, PDM140 was the most sensitive (IC50=73μM), followed by LN229 (IC50=278μM) and U87 (IC50=446μM). Co-culture with pericytes significantly increased GBM spheroid viability by 22.7% (PDM140), 32.5% (LN229), and 22.1% (U87), confirming pericyte-induced TMZ resistance. Notably, pericytes exhibited a 160-fold upregulation of C-C motif chemokine ligand 5 (CCL5) upon TMZ treatment, implicating the CCL5-mediated pathway in chemoresistance. This innovative brain-mimicking 3D GBM model provides a physiologically relevant platform for studying tumor-pericyte interactions and testing therapeutic strategies targeting CCL5-mediated resistance mechanisms in GBM. STATEMENT OF SIGNIFICANCE: We developed a multicellular 3D glioblastoma microphysiological system mimicking the physicochemical properties of brain tissues and pericyte-mediated temozolomide resistance that can be used to screen for standard-of-care chemotherapy. This advanced hydrogel-based platform demonstrated the critical role of the glioblastoma tumor microenvironment in modulating chemotherapy sensitivity, particularly the pericyte-induced CCL5-CCR5 paracrine axis that can lead to the identification of therapeutic targets within the CCL5-CCR5 pathway toward more effective treatments disrupting these resistance mechanisms. Overall, the proposed 3D glioblastoma microphysiological system can transform drug screening and personalized treatment for GBM by offering ethical and cost-effective alternatives to animal testing and more effective drug screening and discovery efforts, ultimately improving GBM patient outcomes.
Collapse
Affiliation(s)
- Surjendu Maity
- Terasaki Institute for Biomedical Innovation, Woodland Hills, CA, 91367, USA.; Department of Orthopedic Surgery, Duke University School of Medicine, Duke University, Durham, NC 27705, USA
| | - Christopher Jewell
- Terasaki Institute for Biomedical Innovation, Woodland Hills, CA, 91367, USA
| | - Can Yilgor
- Terasaki Institute for Biomedical Innovation, Woodland Hills, CA, 91367, USA
| | - Satoru Kawakita
- Terasaki Institute for Biomedical Innovation, Woodland Hills, CA, 91367, USA
| | - Saurabh Sharma
- Department of Surgery, Stanford School of Medicine, Stanford University Medical Center, CA 94305, USA
| | - Alejandro Gomez
- Terasaki Institute for Biomedical Innovation, Woodland Hills, CA, 91367, USA
| | - Marvin Mecwan
- Terasaki Institute for Biomedical Innovation, Woodland Hills, CA, 91367, USA
| | - Natashya Falcone
- Terasaki Institute for Biomedical Innovation, Woodland Hills, CA, 91367, USA
| | - Menekse Ermis
- Terasaki Institute for Biomedical Innovation, Woodland Hills, CA, 91367, USA
| | - Mahsa Monirizad
- Terasaki Institute for Biomedical Innovation, Woodland Hills, CA, 91367, USA
| | - Negar Hosseinzadeh Kouchehbaghi
- Terasaki Institute for Biomedical Innovation, Woodland Hills, CA, 91367, USA.; Department of Textile Engineering, Amirkabir University of Technology (Tehran Polytechnic), Hafez Avenue, 1591634311 Tehran, Iran
| | - Fatemeh Zehtabi
- Terasaki Institute for Biomedical Innovation, Woodland Hills, CA, 91367, USA
| | - Danial Khorsandi
- Terasaki Institute for Biomedical Innovation, Woodland Hills, CA, 91367, USA
| | | | - Diogo Moniz-Garcia
- Mayo Clinic Florida, Department of Neurosurgery, Jacksonville, FL, 32224, USA
| | | | - Ali Khademhosseini
- Terasaki Institute for Biomedical Innovation, Woodland Hills, CA, 91367, USA..
| | - Vadim Jucaud
- Terasaki Institute for Biomedical Innovation, Woodland Hills, CA, 91367, USA..
| |
Collapse
|
3
|
Zhang L, Yang J, Zhou Z, Ren Y, Chen B, Tang A, Zhang K, Li C, Zhou H, Fung KM, Xu C, Kang C, Battiste JD, Bronze MS, Houchen CW, Liu Z, Dunn IF, Cavenee WK, Li M. A zinc transporter drives glioblastoma progression via extracellular vesicles-reprogrammed microglial plasticity. Proc Natl Acad Sci U S A 2025; 122:e2427073122. [PMID: 40305049 PMCID: PMC12067291 DOI: 10.1073/pnas.2427073122] [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: 12/27/2024] [Accepted: 03/22/2025] [Indexed: 05/02/2025] Open
Abstract
Glioblastoma (GBM) is the most aggressive form of brain cancer, with limited therapeutic options. While microglia contribute to GBM progression, the mechanisms by which they foster a protumorigenic immune environment remain poorly understood. We identify the zinc transporter Zrt- And Irt-Like Protein 4 (ZIP4) as a pivotal regulator of the GBM immune landscape. In orthotopic mouse models, ZIP4 drives tumor growth and behavioral changes. Mechanistically, ZIP4 modulates microglial plasticity through tumor-derived extracellular vesicles carrying triggering receptor expressed on myeloid cells-1 (TREM1), a process regulated by the zinc-dependent transcription factor Zinc Finger E-box Binding Homeobox 1 in GBM cells. TREM1 enhances microglial plasticity through the spleen associated tyrosine kinase-Pyruvate dehydrogenase kinase-signal transducer and activator of transcription 3 (SYK-PDK-STAT3) signaling axis, ultimately promoting an immune environment favorable to tumor progression. ZIP4 depletion or TREM1 inhibition attenuates tumor growth and behavioral effects in vivo by disrupting the tumor-microglia interaction. These findings establish ZIP4 as a key modulator of the GBM immune landscape and suggest a promising therapeutic target to counteract microglia-mediated tumor progression.
Collapse
Affiliation(s)
- Liyang Zhang
- Department of Medicine, The University of Oklahoma Health Sciences Center, Oklahoma City, OK73104
- Department of Surgery, The University of Oklahoma Health Sciences Center, Oklahoma City, OK73104
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, Hunan410008, China
| | - Jingxuan Yang
- Department of Medicine, The University of Oklahoma Health Sciences Center, Oklahoma City, OK73104
- Department of Surgery, The University of Oklahoma Health Sciences Center, Oklahoma City, OK73104
| | - Zhijun Zhou
- Department of Medicine, The University of Oklahoma Health Sciences Center, Oklahoma City, OK73104
- Department of Surgery, The University of Oklahoma Health Sciences Center, Oklahoma City, OK73104
| | - Yu Ren
- Department of Medicine, The University of Oklahoma Health Sciences Center, Oklahoma City, OK73104
- Department of Surgery, The University of Oklahoma Health Sciences Center, Oklahoma City, OK73104
| | - Bo Chen
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, Hunan410008, China
| | - Anliu Tang
- Department of Gastroenterology, The Third Xiangya Hospital of Central South University, Changsha, Hunan410013, China
| | - Kailiang Zhang
- Department of Medicine, The University of Oklahoma Health Sciences Center, Oklahoma City, OK73104
- Department of Surgery, The University of Oklahoma Health Sciences Center, Oklahoma City, OK73104
| | - Chuntao Li
- Department of Medicine, The University of Oklahoma Health Sciences Center, Oklahoma City, OK73104
- Department of Surgery, The University of Oklahoma Health Sciences Center, Oklahoma City, OK73104
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, Hunan410008, China
| | - Hongshu Zhou
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, Hunan410008, China
| | - Kar-Ming Fung
- Department of Pathology, The University of Oklahoma Health Sciences Center, Oklahoma City, OK73104
| | - Chao Xu
- Department of Biostatistics and Epidemiology, College of Public Health, The University of Oklahoma Health Sciences Center, Oklahoma City, OK73104
| | - Chunsheng Kang
- Department of Neurosurgery, Tianjin Medical University General Hospital, Laboratory of Neuro-Oncology, Tianjin Neurological Institute, Key Laboratory of Post-Trauma Neuro-repair and Regeneration in Central Nervous System, Ministry of Education, Tianjin300052, China
| | - James D. Battiste
- Department of Neurosurgery, The University of Oklahoma Health Sciences Center, Oklahoma City, OK73104
| | - Michael S. Bronze
- Department of Medicine, The University of Oklahoma Health Sciences Center, Oklahoma City, OK73104
| | - Courtney W. Houchen
- Department of Medicine, The University of Oklahoma Health Sciences Center, Oklahoma City, OK73104
| | - Zhixiong Liu
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, Hunan410008, China
| | - Ian F. Dunn
- Department of Neurosurgery, The University of Oklahoma Health Sciences Center, Oklahoma City, OK73104
| | - Webster K. Cavenee
- Department of Medicine, University of California at San Diego, San Diego, CA92093
| | - Min Li
- Department of Medicine, The University of Oklahoma Health Sciences Center, Oklahoma City, OK73104
- Department of Surgery, The University of Oklahoma Health Sciences Center, Oklahoma City, OK73104
| |
Collapse
|
4
|
Jing L, Xiao W, Hu Z, Liu X, Yuan M. A Systematic Review of Nanoparticle-Mediated Ferroptosis in Glioma Therapy. Int J Nanomedicine 2025; 20:5779-5797. [PMID: 40351706 PMCID: PMC12065465 DOI: 10.2147/ijn.s523008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2025] [Accepted: 04/22/2025] [Indexed: 05/14/2025] Open
Abstract
Glioma, a highly malignant central nervous system tumor, exhibits aggressive invasiveness, extensive infiltration, and poor prognosis. Conventional treatments such as surgery, radiotherapy, and chemotherapy are hindered by limitations including the inability to overcome the blood-brain barrier (BBB), drug resistance, and high recurrence rates. Ferroptosis induced by nanoparticle-based systems offers an innovative strategy for glioma therapy by efficiently traversing the BBB, precisely delivering ferroptosis inducers, enhancing tumor accumulation, and enabling stimuli-responsive drug release. These features collectively improve the induction efficiency of ferroptosis in glioma cells. Various nanoplatforms, including inorganic nanoparticles, biomimetic carriers, and polymer-based systems, have demonstrated potential in crossing the BBB, inducing ferroptosis, and suppressing glioma progression. These systems enhance reactive oxygen species generation, deplete glutathione, and disrupt tumor microenvironment defense mechanisms, achieving synergistic therapeutic effects. The integration of ferroptosis with nanotechnology is emerging as a promising, non-invasive strategy for the treatment of gliomas, offering substantial therapeutic potential.
Collapse
Affiliation(s)
- Lin Jing
- Guangxi Key Laboratory of Special Biomedicine; School of Medicine, Guangxi University, Nanning, 530004, People’s Republic of China
| | - Wenguang Xiao
- Guangxi Key Laboratory of Special Biomedicine; School of Medicine, Guangxi University, Nanning, 530004, People’s Republic of China
| | - Zhouxing Hu
- Guangxi Key Laboratory of Special Biomedicine; School of Medicine, Guangxi University, Nanning, 530004, People’s Republic of China
| | - Xu Liu
- Guangxi Key Laboratory of Special Biomedicine; School of Medicine, Guangxi University, Nanning, 530004, People’s Republic of China
| | - Mingqing Yuan
- Guangxi Key Laboratory of Special Biomedicine; School of Medicine, Guangxi University, Nanning, 530004, People’s Republic of China
| |
Collapse
|
5
|
Xu H, Fan Z, Jiang S, Sun M, Chai H, Zhu R, Liu X, Wang Y, Chen J, Wei J, Mao Y, Shi Z. Integrating Multiplex Immunohistochemistry and Machine Learning for Glioma Subtyping and Prognosis Prediction. MedComm (Beijing) 2025; 6:e70138. [PMID: 40264576 PMCID: PMC12013734 DOI: 10.1002/mco2.70138] [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: 07/30/2024] [Revised: 01/24/2025] [Accepted: 02/09/2025] [Indexed: 04/24/2025] Open
Abstract
Glioma subtyping is crucial for treatment decisions, but traditional approaches often fail to capture tumor heterogeneity. This study proposes a novel framework integrating multiplex immunohistochemistry (mIHC) and machine learning for glioma subtyping and prognosis prediction. 185 patient samples from the Huashan hospital cohort were stained using a multi-label mIHC panel and analyzed with an AI-based auto-scanning system to calculate cell ratios and determine the proportion of positive tumor cells for various markers. Patients were divided into two cohorts (training: N = 111, testing: N = 74), and a machine learning model was then developed and validated for subtype classification and prognosis prediction. The framework identified two distinct glioma subtypes with significant differences in prognosis, clinical characteristics, and molecular profiles. The high-risk subtype, associated with older age, poorer outcomes, astrocytoma/glioblastoma, higher tumor grades, elevated mesenchymal scores, and an inhibitory immune microenvironment, exhibited IDH wild-type, 1p19q non-codeletion, and MGMT promoter unmethylation, suggesting chemotherapy resistance. Conversely, the low-risk subtype, characterized by younger age, better prognosis, astrocytoma/oligodendroglioma, lower tumor grades, and favorable molecular profiles (IDH mutation, 1p19q codeletion, MGMT promoter methylation), indicated chemotherapy sensitivity. The mIHC-based framework enables rapid glioma classification, facilitating tailored treatment strategies and accurate prognosis prediction, potentially improving patient management and outcomes.
Collapse
Affiliation(s)
- Houshi Xu
- Department of NeurosurgeryHuashan HospitalShanghai Medical CollegeFudan UniversityShanghaiChina
- Research Unit of New Technologies of Micro‐Endoscopy Combination in Skull Base Surgery (2018RU008)Chinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
- Department of NeurosurgeryPeking Union Medical College HospitalChinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
| | - Zhen Fan
- Department of NeurosurgeryHuashan HospitalShanghai Medical CollegeFudan UniversityShanghaiChina
- Research Unit of New Technologies of Micro‐Endoscopy Combination in Skull Base Surgery (2018RU008)Chinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
| | - Shan Jiang
- Department of NeurosurgeryHuashan HospitalShanghai Medical CollegeFudan UniversityShanghaiChina
- Research Unit of New Technologies of Micro‐Endoscopy Combination in Skull Base Surgery (2018RU008)Chinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
| | - Maoyuan Sun
- Department of NeurosurgeryHuashan HospitalShanghai Medical CollegeFudan UniversityShanghaiChina
- Research Unit of New Technologies of Micro‐Endoscopy Combination in Skull Base Surgery (2018RU008)Chinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
| | - Huihui Chai
- Department of NeurosurgeryHuashan HospitalShanghai Medical CollegeFudan UniversityShanghaiChina
- Research Unit of New Technologies of Micro‐Endoscopy Combination in Skull Base Surgery (2018RU008)Chinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
| | - Ruize Zhu
- Department of NeurosurgeryHuashan HospitalShanghai Medical CollegeFudan UniversityShanghaiChina
- Research Unit of New Technologies of Micro‐Endoscopy Combination in Skull Base Surgery (2018RU008)Chinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
| | - Xiaoyu Liu
- Department of NeurosurgeryPeking Union Medical College HospitalChinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
| | - Yue Wang
- Department of NeurosurgeryHuashan HospitalShanghai Medical CollegeFudan UniversityShanghaiChina
- Research Unit of New Technologies of Micro‐Endoscopy Combination in Skull Base Surgery (2018RU008)Chinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
- Department of NeurosurgeryPeking Union Medical College HospitalChinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
| | - Jiawen Chen
- Department of NeurosurgeryHuashan HospitalShanghai Medical CollegeFudan UniversityShanghaiChina
- Research Unit of New Technologies of Micro‐Endoscopy Combination in Skull Base Surgery (2018RU008)Chinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
| | - Junji Wei
- Research Unit of New Technologies of Micro‐Endoscopy Combination in Skull Base Surgery (2018RU008)Chinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
- Department of NeurosurgeryPeking Union Medical College HospitalChinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
| | - Ying Mao
- Department of NeurosurgeryHuashan HospitalShanghai Medical CollegeFudan UniversityShanghaiChina
- Research Unit of New Technologies of Micro‐Endoscopy Combination in Skull Base Surgery (2018RU008)Chinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
| | - Zhifeng Shi
- Department of NeurosurgeryHuashan HospitalShanghai Medical CollegeFudan UniversityShanghaiChina
- Research Unit of New Technologies of Micro‐Endoscopy Combination in Skull Base Surgery (2018RU008)Chinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
| |
Collapse
|
6
|
Saha S, Zhang Y, Gibert MK, Dube C, Hanif F, Mulcahy E, Bednarek S, Marcinkiewicz P, Wang X, Kwak G, Hudson K, Sun Y, Dinda M, Saha T, Guessous F, Cruickshanks N, Colon RR, Dell'Olio LG, Anbu R, Kefas B, Kumar P, Klibanov AL, Schiff D, Suk JS, Hanes J, Mata J, Hafner M, Abounader R. Discovery and therapeutic exploitation of Master Regulatory miRNAs in Glioblastoma. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.04.01.646663. [PMID: 40236125 PMCID: PMC11996502 DOI: 10.1101/2025.04.01.646663] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 04/17/2025]
Abstract
Glioblastoma is a fatal primary malignant brain tumor. Despite therapies involving surgical resection, chemotherapy, and radiation therapy, the average survival for glioblastoma patients remains at approximately 15 months. MicroRNAs (miRNAs) are short noncoding RNA molecules that regulate the expression of the majority of human genes. Numerous genes are concurrently deregulated in glioblastoma. Consequently, molecular monotherapies have failed to achieve improvements in clinical outcomes. Several lines of evidence suggest that simultaneous targeting of several deregulated molecules is required to achieve better therapies. However, the simultaneous targeting of several deregulated oncogenic drivers is severely limited by the fact that the drugs needed to target many deregulated molecules do not currently exist, and because combining several drugs in a clinical setting leads to an exponential increase in toxicity. We hypothesized that we can develop and use miRNA to simultaneously inhibit multiple deregulated genes for more efficacious glioblastoma therapies. The goal of this study was therefore to identify master regulatory microRNAs (miRNAs) and use them to simultaneously target multiple deregulated molecules for GBM therapy. We defined master regulatory miRNAs as those that target several deregulated genes in glioblastoma. To find master regulatory miRNAs, we first used PAR-CLIP screenings to identify all targets of all miRNAs in glioblastoma cells. We then analyzed TCGA tumor data to determine which of these targets are deregulated in human tumors. We developed and used an algorithm to rank these targets for significance in glioblastoma malignancy based on their magnitude of deregulation, frequency of deregulation, and correlation with patient survival. We then ranked the miRNAs for their capacity of targeting multiple glioblastoma-deregulated genes and therefore the potential to exhibit strong anti-tumor effects when delivered as therapy. Using this strategy, we selected two tumor suppressor master regulatory miRNAs, miR-340, miR-382 and an oncogenic master regulatory miRNA, miR-17. We validated the target genes of the miRNAs and showed that they form part of important glioblastoma regulatory pathways. We then showed that the miRNAs (miR-340 and miR-582) or the miR-17 inhibitor have strong inhibitory effects on glioblastoma cell growth, survival, invasion, stemness and in vivo tumor growth. Ultimately, we developed and successfully tested a new therapeutic approach to delivery miR-340 using MRI guided focused ultrasound and microbubbles (FUS-MB) and special brain penetrating nanoparticles (BPN). This approach resulted in a substantial reduction in tumor volume and prolongation of the survival of glioblastoma-bearing mice and can be translated into clinical trials. We therefore developed and successfully tested a novel strategy to discover and deliver miRNAs for glioblastoma and cancer therapy.
Collapse
|
7
|
Wei H, Li X, Feng P, He Z. SERPINH1 and CTSZ are Key Markers of Glioma Angiogenesis. J Mol Neurosci 2025; 75:51. [PMID: 40257572 DOI: 10.1007/s12031-025-02349-0] [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: 02/07/2025] [Accepted: 04/14/2025] [Indexed: 04/22/2025]
Abstract
Glioma, as one of the most complex and prognostically variable malignant tumors of the central nervous system, poses a significant challenge to clinical decision-making due to its molecular heterogeneity. This study aims to deepen our understanding of glioma molecular subtypes and explore key gene markers with prognostic and diagnostic value. We utilized an angiogenesis-related gene set and employed the Non-negative Matrix Factorization (NMF) algorithm to successfully identify two distinct prognostic subtypes, with subtype one exhibiting more unfavorable prognostic characteristics. To further elucidate the biological functional differences between these two subtypes, we conducted Gene Ontology (GO) functional annotation, Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway analysis, and Gene Set Enrichment Analysis (GSEA). Building on this, we integrated differentially expressed genes between subtypes with core genes revealed by Weighted Gene Co-expression Network Analysis (WGCNA) through intersection analysis to pinpoint a series of key candidate genes. Subsequently, we constructed a Protein-Protein Interaction (PPI) network to identify genes occupying central nodes within the network. To screen markers with high specificity and sensitivity for prognosis and diagnosis, we adopted a dual-track strategy: on the one hand, we utilized machine learning algorithms such as Lasso regression, Support Vector Machine (SVM), and Random Forest (RF) to select core genes, identifying markers that can accurately predict the subtype with a poor prognosis; on the other hand, we employed a comprehensive evaluation system incorporating 101 machine learning ensemble algorithms to further validate and screen prognosis-related genes. Through cross-validation using these two strategies, we ultimately determined SERPINH1 and CTSZ as dual prognostic and diagnostic markers for glioma. This study not only provides a new perspective and tool for the molecular subclassification of glioma but also, through a rigorous multi-algorithm, multi-dimensional screening process, uncovers SERPINH1 and CTSZ as markers with potential clinical translational value. These findings are expected to offer more precise biomarker support for the early diagnosis and prognostic assessment of glioma, potentially paving new avenues for the development of personalized treatment strategies and improving patient outcomes. This has far-reaching implications for the clinical management of glioma in the field of neurosurgery.
Collapse
Affiliation(s)
- Haotian Wei
- Department of Neurosurgery, First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Xinlong Li
- Lanzhou University Second Hospital, The Second Medical College of Lanzhou University, Lanzhou, 730030, China
| | - Peng Feng
- Lanzhou University Second Hospital, The Second Medical College of Lanzhou University, Lanzhou, 730030, China
| | - Zhaohui He
- Department of Neurosurgery, First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China.
| |
Collapse
|
8
|
Zhang W, Wang J, Ji J, Wang P, Yuan G, Fang S, Liu F, Jin G, Zhang J. Glioblastoma cells secrete ICAM1 via FASN signaling to promote glioma-associated macrophage infiltration. Cell Signal 2025; 132:111823. [PMID: 40252818 DOI: 10.1016/j.cellsig.2025.111823] [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: 01/30/2025] [Revised: 04/14/2025] [Accepted: 04/16/2025] [Indexed: 04/21/2025]
Abstract
Glioma-associated macrophages (GAMs) constitute the most abundant subset of immune cells in the glioblastoma (GBM) microenvironment, but the underlying mechanism of intense infiltration needs to be elucidated. In this study, we found that GBM cells secrete ICAM1 via FASN signaling to promote GAM infiltration. FASN expression is correlated with GAM density in GBM patients. In vitro experiments revealed that FASN regulates the type-I interferon pathway, particularly STAT1 expression. Moreover, disrupting FASN-STAT1 signaling through the overexpression or inhibition of FASN or STAT1 in GBM cells strongly influences microglial recruitment. Additionally, ICAM1 acts as a direct transcriptional candidate of FASN-STAT1 and a paracrine soluble factor, recruiting microglia to GBM tumors. This study revealed crosstalk between GBM cells and GAMs through FASN-STAT1-ICAM1 signaling to promote microglial infiltration, suggesting potential strategies for treating GBM patients.
Collapse
Affiliation(s)
- Wenxin Zhang
- Brain Tumor Research Center, Beijing Neurosurgical Institute, Capital Medical University, Beijing, China
| | - Jialin Wang
- Brain Tumor Research Center, Beijing Neurosurgical Institute, Capital Medical University, Beijing, China
| | - Jiayu Ji
- Brain Tumor Research Center, Beijing Neurosurgical Institute, Capital Medical University, Beijing, China; China Rehabilitation Science Institute, China Rehabilitation Research Center, School of Rehabilitation, Capital Medical University, Beijing, PR China
| | - Peiwen Wang
- Brain Tumor Research Center, Beijing Neurosurgical Institute, Capital Medical University, Beijing, China
| | - Guiqiang Yuan
- Brain Tumor Research Center, Beijing Neurosurgical Institute, Capital Medical University, Beijing, China
| | - Sheng Fang
- Brain Tumor Research Center, Beijing Neurosurgical Institute, Capital Medical University, Beijing, China
| | - Fusheng Liu
- Brain Tumor Research Center, Beijing Neurosurgical Institute, Capital Medical University, Beijing, China; Department of Neurosurgery, Beijing Tiantan, Capital Medical University, Beijing, China
| | - Guishan Jin
- Brain Tumor Research Center, Beijing Neurosurgical Institute, Capital Medical University, Beijing, China.
| | - Junwen Zhang
- Brain Tumor Research Center, Beijing Neurosurgical Institute, Capital Medical University, Beijing, China.
| |
Collapse
|
9
|
Stone TJ, Pickles JC, Ogunbiyi O, Yasin SA, Taylor CA, Ahmed SW, Chalker J, Dryden C, Slodkowska I, Pang E, Kristiansen M, Williams R, Tutill H, Williams CA, Madhan GK, Forrest L, Brooks T, Hubank M, Hughes D, Proszek P, Pietka G, Peat E, Hargrave D, Jacques TS. The tumour microenvironment of pilocytic astrocytoma evolves over time via enrichment for microglia. Acta Neuropathol Commun 2025; 13:30. [PMID: 39948623 PMCID: PMC11823165 DOI: 10.1186/s40478-024-01922-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2024] [Accepted: 12/23/2024] [Indexed: 02/16/2025] Open
Abstract
Pilocytic astrocytoma (PA) is the commonest low-grade tumour affecting children and is frequently experienced as a chronic disease associated with extended treatment, periods of regrowth, and long-term disability. This contrasts with the view of PA as a benign tumour with positive clinical outcomes and raises the fundamental question of biologically driven change over time within these tumours, which will impact diagnosis, stratification, and management. To investigate the molecular, cellular, and pathological stability of PA we performed RNA sequencing, methylation array profiling, immunohistochemistry, and targeted panel DNA sequencing on a cohort of 15 PA patients with matched primary/longitudinal samples at a mean sampling interval of 2.7 years. Through pairwise analysis of primary versus longitudinal tumour samples we identified changes to immune-related pathways within the expression and methylation profiles of longitudinal PA. Further interrogation of these changes revealed an enrichment over time for microglial cell populations, which was validated by immunohistochemistry against common monocyte/microglial markers. Moreover, immunohistochemical characterisation revealed concurrent increases in the expression of M2-like and anti-inflammatory markers. Microglial enrichments were consistent across the cohort and were not adequately explained by a range of potential confounders, including receipt of adjuvant therapy. Taken together, these data challenge the idea of pilocytic astrocytoma as a static entity and indicate that they consistently accumulate microglia over time, potentially co-opting the immune microenvironment towards an anti-inflammatory phenotype that may affect the natural course and treatment response of the tumours.
Collapse
Affiliation(s)
- Thomas J Stone
- Developmental Biology and Cancer Research and Teaching Department, UCL Great Ormond Street Institute of Child Health, 30 Guilford Street, London, WC1N 1EH, UK.
- Department of Histopathology, Great Ormond Street Hospital for Children NHS Foundation Trust, Great Ormond Street, London, WC1N 3JH, UK.
| | - Jessica C Pickles
- Developmental Biology and Cancer Research and Teaching Department, UCL Great Ormond Street Institute of Child Health, 30 Guilford Street, London, WC1N 1EH, UK
- Department of Histopathology, Great Ormond Street Hospital for Children NHS Foundation Trust, Great Ormond Street, London, WC1N 3JH, UK
| | - Olumide Ogunbiyi
- Department of Histopathology, Great Ormond Street Hospital for Children NHS Foundation Trust, Great Ormond Street, London, WC1N 3JH, UK
| | - Shireena A Yasin
- Department of Histopathology, Great Ormond Street Hospital for Children NHS Foundation Trust, Great Ormond Street, London, WC1N 3JH, UK
| | - Catherine A Taylor
- Developmental Biology and Cancer Research and Teaching Department, UCL Great Ormond Street Institute of Child Health, 30 Guilford Street, London, WC1N 1EH, UK
- Department of Histopathology, Great Ormond Street Hospital for Children NHS Foundation Trust, Great Ormond Street, London, WC1N 3JH, UK
| | - Saira W Ahmed
- Developmental Biology and Cancer Research and Teaching Department, UCL Great Ormond Street Institute of Child Health, 30 Guilford Street, London, WC1N 1EH, UK
- Department of Histopathology, Great Ormond Street Hospital for Children NHS Foundation Trust, Great Ormond Street, London, WC1N 3JH, UK
| | - Jane Chalker
- Specialist Integrated Haematology and Malignancy Diagnostic Service, Great Ormond Street Hospital for Children NHS Foundation Trust, Great Ormond Street, London, WC1N 3JH, UK
| | - Carryl Dryden
- Specialist Integrated Haematology and Malignancy Diagnostic Service, Great Ormond Street Hospital for Children NHS Foundation Trust, Great Ormond Street, London, WC1N 3JH, UK
| | - Iwona Slodkowska
- Specialist Integrated Haematology and Malignancy Diagnostic Service, Great Ormond Street Hospital for Children NHS Foundation Trust, Great Ormond Street, London, WC1N 3JH, UK
| | - Emily Pang
- Specialist Integrated Haematology and Malignancy Diagnostic Service, Great Ormond Street Hospital for Children NHS Foundation Trust, Great Ormond Street, London, WC1N 3JH, UK
| | - Mark Kristiansen
- UCL Genomics, Zayed Centre for Research into Rare Disease in Children, 20 Guilford Street, London, WC1N 1DZ, UK
| | - Rachel Williams
- UCL Genomics, Zayed Centre for Research into Rare Disease in Children, 20 Guilford Street, London, WC1N 1DZ, UK
| | - Helena Tutill
- UCL Genomics, Zayed Centre for Research into Rare Disease in Children, 20 Guilford Street, London, WC1N 1DZ, UK
| | - Charlotte A Williams
- UCL Genomics, Zayed Centre for Research into Rare Disease in Children, 20 Guilford Street, London, WC1N 1DZ, UK
| | - Gaganjit K Madhan
- UCL Genomics, Zayed Centre for Research into Rare Disease in Children, 20 Guilford Street, London, WC1N 1DZ, UK
| | - Leysa Forrest
- UCL Genomics, Zayed Centre for Research into Rare Disease in Children, 20 Guilford Street, London, WC1N 1DZ, UK
| | - Tony Brooks
- UCL Genomics, Zayed Centre for Research into Rare Disease in Children, 20 Guilford Street, London, WC1N 1DZ, UK
| | - Mike Hubank
- Clinical Genomics, Centre for Molecular Pathology, Royal Marsden Hospital, London, SM2 5NG, UK
| | - Debbie Hughes
- Clinical Genomics, Centre for Molecular Pathology, Royal Marsden Hospital, London, SM2 5NG, UK
| | - Paula Proszek
- Clinical Genomics, Centre for Molecular Pathology, Royal Marsden Hospital, London, SM2 5NG, UK
| | - Grzegorz Pietka
- Clinical Genomics, Centre for Molecular Pathology, Royal Marsden Hospital, London, SM2 5NG, UK
| | - Erin Peat
- Clinical Genomics, Centre for Molecular Pathology, Royal Marsden Hospital, London, SM2 5NG, UK
| | - Darren Hargrave
- Developmental Biology and Cancer Research and Teaching Department, UCL Great Ormond Street Institute of Child Health, 30 Guilford Street, London, WC1N 1EH, UK
- Department of Haematology and Oncology, Great Ormond Street Hospital for Children NHS Foundation Trust, Great Ormond Street, London, WC1N 3JH, UK
| | - Thomas S Jacques
- Developmental Biology and Cancer Research and Teaching Department, UCL Great Ormond Street Institute of Child Health, 30 Guilford Street, London, WC1N 1EH, UK
- Department of Histopathology, Great Ormond Street Hospital for Children NHS Foundation Trust, Great Ormond Street, London, WC1N 3JH, UK
| |
Collapse
|
10
|
Perelroizen R, Gaoni-Yogev A, Mayo L. Stereotaxic Genetic Perturbation of the Tumor Microenvironment in the Rodent Brain. Methods Mol Biol 2025; 2926:73-89. [PMID: 40266519 DOI: 10.1007/978-1-0716-4542-0_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] [Indexed: 04/24/2025]
Abstract
Brain cancers such as glioblastoma (GBM) cause a disproportionate level of morbidity and mortality among cancer patients. Therapeutic advances over the past decade have done little to change this bleak prognosis, helping only a minority of patients. The brain tumor microenvironment (TME) is highly supportive of the tumors. It differs from other peripheral malignancies due to its unique composition, which includes the glial, neural, and immune cell populations. Until recently, the study of the brain TME was limited by the lack of methods to target the different cells in TME. This protocol describes stereotaxic surgery optimized for gene delivery by recombinant adeno-associated viruses or lentiviruses in mice and rats. This method allows the manipulation of gene expression in the TME with excellent spatiotemporal control in specific cells (or a subpopulation of cells). Many aspects of the technique, including its versatility, ease of application, and high reproducibility, make it an attractive approach for studying cellular and circuit functions in the TME.
Collapse
Affiliation(s)
- Rita Perelroizen
- Sagol School of Neuroscience, Tel Aviv University, Tel Aviv, Israel
| | - Anat Gaoni-Yogev
- Shmunis School of Biomedicine and Cancer Research, Life Sciences Faculty, Tel Aviv University, Tel Aviv, Israel
| | - Lior Mayo
- Sagol School of Neuroscience, Tel Aviv University, Tel Aviv, Israel.
- Shmunis School of Biomedicine and Cancer Research, Life Sciences Faculty, Tel Aviv University, Tel Aviv, Israel.
| |
Collapse
|
11
|
Maity S, Bhuyan T, Jewell C, Kawakita S, Sharma S, Nguyen HT, Najafabadi AH, Ermis M, Falcone N, Chen J, Mandal K, Khorsandi D, Yilgor C, Choroomi A, Torres E, Mecwan M, John JV, Akbari M, Wang Z, Moniz-Garcia D, Quiñones-Hinojosa A, Jucaud V, Dokmeci MR, Khademhosseini A. Recent Developments in Glioblastoma-On-A-Chip for Advanced Drug Screening Applications. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2025; 21:e2405511. [PMID: 39535474 PMCID: PMC11719323 DOI: 10.1002/smll.202405511] [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: 07/03/2024] [Revised: 09/08/2024] [Indexed: 11/16/2024]
Abstract
Glioblastoma (GBM) is an aggressive form of cancer, comprising ≈80% of malignant brain tumors. However, there are no effective treatments for GBM due to its heterogeneity and the presence of the blood-brain barrier (BBB), which restricts the delivery of therapeutics to the brain. Despite in vitro models contributing to the understanding of GBM, conventional 2D models oversimplify the complex tumor microenvironment. Organ-on-a-chip (OoC) models have emerged as promising platforms that recapitulate human tissue physiology, enabling disease modeling, drug screening, and personalized medicine. There is a sudden increase in GBM-on-a-chip models that can significantly advance the knowledge of GBM etiology and revolutionize drug development by reducing animal testing and enhancing translation to the clinic. In this review, an overview of GBM-on-a-chip models and their applications is reported for drug screening and discussed current challenges and potential future directions for GBM-on-a-chip models.
Collapse
Affiliation(s)
- Surjendu Maity
- Terasaki Institute for Biomedical Innovation, Los Angeles,
CA, 90064 USA
- Department of Orthopedic Surgery, Duke University School of
Medicine, Duke University, Durham, NC 27705
| | - Tamanna Bhuyan
- Department of Applied Biology, School of Biological
Sciences, University of Science & Technology Meghalaya, Meghalaya, 793101,
India
| | - Christopher Jewell
- Terasaki Institute for Biomedical Innovation, Los Angeles,
CA, 90064 USA
| | - Satoru Kawakita
- Terasaki Institute for Biomedical Innovation, Los Angeles,
CA, 90064 USA
| | - Saurabh Sharma
- Terasaki Institute for Biomedical Innovation, Los Angeles,
CA, 90064 USA
| | - Huu Tuan Nguyen
- Terasaki Institute for Biomedical Innovation, Los Angeles,
CA, 90064 USA
| | | | - Menekse Ermis
- Terasaki Institute for Biomedical Innovation, Los Angeles,
CA, 90064 USA
- Center of Excellence in Biomaterials and Tissue
Engineering, Middle East Technical University, Ankara, Turkey
| | - Natashya Falcone
- Terasaki Institute for Biomedical Innovation, Los Angeles,
CA, 90064 USA
| | - Junjie Chen
- Terasaki Institute for Biomedical Innovation, Los Angeles,
CA, 90064 USA
| | - Kalpana Mandal
- Terasaki Institute for Biomedical Innovation, Los Angeles,
CA, 90064 USA
| | - Danial Khorsandi
- Terasaki Institute for Biomedical Innovation, Los Angeles,
CA, 90064 USA
| | - Can Yilgor
- Terasaki Institute for Biomedical Innovation, Los Angeles,
CA, 90064 USA
| | - Auveen Choroomi
- Terasaki Institute for Biomedical Innovation, Los Angeles,
CA, 90064 USA
| | - Emily Torres
- Terasaki Institute for Biomedical Innovation, Los Angeles,
CA, 90064 USA
| | - Marvin Mecwan
- Terasaki Institute for Biomedical Innovation, Los Angeles,
CA, 90064 USA
| | - Johnson V. John
- Terasaki Institute for Biomedical Innovation, Los Angeles,
CA, 90064 USA
| | - Mohsen Akbari
- Terasaki Institute for Biomedical Innovation, Los Angeles,
CA, 90064 USA
- Laboratoryfor Innovations in Micro Engineering (LiME),
Department of Mechanical Engineering, University of Victoria, Victoria, BC V8P 5C2,
Canada
- Biotechnology Center, Silesian University of Technology,
Akademicka 2A, 44-100 Gliwice, Poland
| | - Zhaohui Wang
- Terasaki Institute for Biomedical Innovation, Los Angeles,
CA, 90064 USA
| | | | | | - Vadim Jucaud
- Terasaki Institute for Biomedical Innovation, Los Angeles,
CA, 90064 USA
| | | | - Ali Khademhosseini
- Terasaki Institute for Biomedical Innovation, Los Angeles,
CA, 90064 USA
| |
Collapse
|
12
|
Kricha A, Bouchmaa N, Ben Mkaddem S, Abbaoui A, Ben Mrid R, El Fatimy R. Glioblastoma-associated macrophages: A key target in overcoming glioblastoma therapeutic resistance. Cytokine Growth Factor Rev 2024; 80:97-108. [PMID: 39510901 DOI: 10.1016/j.cytogfr.2024.10.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2024] [Revised: 10/24/2024] [Accepted: 10/24/2024] [Indexed: 11/15/2024]
Abstract
Glioblastoma multiforme (GBM) is recognized as the most aggressive and malignant form of brain cancer, characterized by a highly heterogeneous phenotype, poor prognosis, and a median survival time of less than 16 months. Recent studies have highlighted the critical role of glioblastoma-associated macrophages (GAMs) in promoting tumor progression and resistance not only to immunotherapy but also to radiotherapy and chemotherapy. GAMs actively suppress immune responses, promote angiogenesis, facilitate tumor metastasis, and induce therapy resistance, through various mechanisms such as cytokines production, signaling pathways regulation, and angiogenesis. In this context, understanding these regulatory mechanisms is essential for developing efficient therapies. Preclinical studies have investigated diverse approaches to target these cells, both as monotherapies or in combination with other treatments. While these approaches have shown promise in strengthening antitumor immune responses in animal models, their clinical success remains to be fully determined. Herein, we provide a comprehensive overview of GAMs's role in GBM therapeutic resistance and summarizes existing approaches to target GAMs in overcoming tumor resistance.
Collapse
Affiliation(s)
- Aymane Kricha
- Institute of Biological Sciences (IBS), Faculty of Medical Sciences, Mohammed VI Polytechnic University (FMS-UM6P), Benguerir, Morocco.
| | - Najat Bouchmaa
- Institute of Biological Sciences (IBS), Faculty of Medical Sciences, Mohammed VI Polytechnic University (FMS-UM6P), Benguerir, Morocco.
| | - Sanae Ben Mkaddem
- Institute of Biological Sciences (IBS), Faculty of Medical Sciences, Mohammed VI Polytechnic University (FMS-UM6P), Benguerir, Morocco.
| | - Abdellatif Abbaoui
- Institute of Biological Sciences (IBS), Faculty of Medical Sciences, Mohammed VI Polytechnic University (FMS-UM6P), Benguerir, Morocco.
| | - Reda Ben Mrid
- Institute of Biological Sciences (IBS), Faculty of Medical Sciences, Mohammed VI Polytechnic University (FMS-UM6P), Benguerir, Morocco.
| | - Rachid El Fatimy
- Institute of Biological Sciences (IBS), Faculty of Medical Sciences, Mohammed VI Polytechnic University (FMS-UM6P), Benguerir, Morocco.
| |
Collapse
|
13
|
Asioli S, Gatto L, Vardy U, Agostinelli C, Di Nunno V, Righi S, Tosoni A, Ambrosi F, Bartolini S, Giannini C, Franceschi E. Immunophenotypic Profile of Adult Glioblastoma IDH-Wildtype Microenvironment: A Cohort Study. Cancers (Basel) 2024; 16:3859. [PMID: 39594814 PMCID: PMC11592556 DOI: 10.3390/cancers16223859] [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: 07/21/2024] [Revised: 10/15/2024] [Accepted: 11/13/2024] [Indexed: 11/28/2024] Open
Abstract
Background: Glioblastoma IDH-wildtype (GBM IDH-wt) is the most aggressive brain tumor in adults and is characterized by an immunosuppressive microenvironment. Different factors shaping its tumor microenvironment (TME) regulate tumor progression and treatment response. The aim of this study was to characterize the main immunosuppressive elements of the GBM IDH-wt TME. Methods: Immunohistochemistry for CD3, CD4, CD8, CD163, programmed death ligand 1 (PD-L1) and programmed death 1 (PD1) was performed on surgical tumor specimens from patients diagnosed with GBM IDH-wt, according to the CNS WHO 2021 criteria. The impact of categorical variables on time-dependent outcomes such as overall survival (OS) and progression-free survival (PFS) has been estimated through the Kaplan-Meier method. Results: We included 30 patients (19 males and 11 females), median age of 59.8 years (range 40.2-69.1 years). All patients underwent surgery followed by temozolomide concurrent with and adjuvant to radiotherapy. MGMT was methylated in 14 patients (47%) and unmethylated in 16 patients (53%). The overall absolute percentages of CD4+ lymphocytes, both intratumoral and perivascular, were significantly more represented than CD8+ lymphocytes in the TME (p = 0.02). A low density of CD4+ lymphocytes (≤10%) was found to be a favorable prognostic factor for GBM outcome (p = 0.02). Patients with MGMT methylated and unmethylated tumors exhibited a distinct TME composition, with a significant higher number of perivascular CD8+ lymphocytes (p = 0.002), intratumoral CD8+ lymphocytes (p = 0.0024) and perivascular CD4+ lymphocytes (p = 0.014) in MGMT unmethylated tumors. PD-L1 expression in tumor cell surface was observed in four tumors (13.3%), and PD1 expression in infiltrating T lymphocytes was observed in nine (30%) tumors, with predominantly perivascular distribution. Conclusions: MGMT methylated and unmethylated tumors exhibit different immune profiles, likely reflecting the different biology of these tumors. The expression of PD-L1 in GBM IDH-wt patients is confined to a small subpopulation. While we found a significant association between low CD4+ lymphocyte density (≤10%) and survival, given the small numbers of our cohort, the prognostic value of CD4+ lymphocyte density will need to be validated in large-scale studies.
Collapse
Affiliation(s)
- Sofia Asioli
- Department of Biomedical and Neuromotor Sciences (DIBINEM), University of Bologna, 40127 Bologna, Italy; (S.A.); (C.G.)
- IRCCS Istituto delle Scienze Neurologiche di Bologna, 40139 Bologna, Italy
| | - Lidia Gatto
- Nervous System Medical Oncology Department, IRCCS Istituto delle Scienze Neurologiche di Bologna, 40139 Bologna, Italy; (V.D.N.); (A.T.); (S.B.); (E.F.)
| | - Uri Vardy
- School of Medicine and Surgery, University of Bologna, 40138 Bologna, Italy;
| | - Claudio Agostinelli
- Haematopathology Unit, IRCCS Azienda Ospedaliero-Universitaria of Bologna, Via Massarenti 9, 40138 Bologna, Italy;
- Department of Medical and Surgical Sciences, University of Bologna, 40138 Bologna, Italy
| | - Vincenzo Di Nunno
- Nervous System Medical Oncology Department, IRCCS Istituto delle Scienze Neurologiche di Bologna, 40139 Bologna, Italy; (V.D.N.); (A.T.); (S.B.); (E.F.)
| | - Simona Righi
- Pathology Unit, Maggiore Hospital-AUSL Bologna, 40133 Bologna, Italy; (S.R.); (F.A.)
| | - Alicia Tosoni
- Nervous System Medical Oncology Department, IRCCS Istituto delle Scienze Neurologiche di Bologna, 40139 Bologna, Italy; (V.D.N.); (A.T.); (S.B.); (E.F.)
| | - Francesca Ambrosi
- Pathology Unit, Maggiore Hospital-AUSL Bologna, 40133 Bologna, Italy; (S.R.); (F.A.)
- Department of Medical and Surgical Sciences (DIMEC), University of Bologna, 40126 Bologna, Italy
| | - Stefania Bartolini
- Nervous System Medical Oncology Department, IRCCS Istituto delle Scienze Neurologiche di Bologna, 40139 Bologna, Italy; (V.D.N.); (A.T.); (S.B.); (E.F.)
| | - Caterina Giannini
- Department of Biomedical and Neuromotor Sciences (DIBINEM), University of Bologna, 40127 Bologna, Italy; (S.A.); (C.G.)
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN 55905, USA
| | - Enrico Franceschi
- Nervous System Medical Oncology Department, IRCCS Istituto delle Scienze Neurologiche di Bologna, 40139 Bologna, Italy; (V.D.N.); (A.T.); (S.B.); (E.F.)
| |
Collapse
|
14
|
Pacal I, Celik O, Bayram B, Cunha A. Enhancing EfficientNetv2 with global and efficient channel attention mechanisms for accurate MRI-Based brain tumor classification. CLUSTER COMPUTING 2024; 27:11187-11212. [DOI: 10.1007/s10586-024-04532-1] [Citation(s) in RCA: 15] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/14/2024] [Revised: 04/08/2024] [Accepted: 04/22/2024] [Indexed: 05/14/2025]
Abstract
AbstractThe early and accurate diagnosis of brain tumors is critical for effective treatment planning, with Magnetic Resonance Imaging (MRI) serving as a key tool in the non-invasive examination of such conditions. Despite the advancements in Computer-Aided Diagnosis (CADx) systems powered by deep learning, the challenge of accurately classifying brain tumors from MRI scans persists due to the high variability of tumor appearances and the subtlety of early-stage manifestations. This work introduces a novel adaptation of the EfficientNetv2 architecture, enhanced with Global Attention Mechanism (GAM) and Efficient Channel Attention (ECA), aimed at overcoming these hurdles. This enhancement not only amplifies the model’s ability to focus on salient features within complex MRI images but also significantly improves the classification accuracy of brain tumors. Our approach distinguishes itself by meticulously integrating attention mechanisms that systematically enhance feature extraction, thereby achieving superior performance in detecting a broad spectrum of brain tumors. Demonstrated through extensive experiments on a large public dataset, our model achieves an exceptional high-test accuracy of 99.76%, setting a new benchmark in MRI-based brain tumor classification. Moreover, the incorporation of Grad-CAM visualization techniques sheds light on the model’s decision-making process, offering transparent and interpretable insights that are invaluable for clinical assessment. By addressing the limitations inherent in previous models, this study not only advances the field of medical imaging analysis but also highlights the pivotal role of attention mechanisms in enhancing the interpretability and accuracy of deep learning models for brain tumor diagnosis. This research sets the stage for advanced CADx systems, enhancing patient care and treatment outcomes.
Collapse
|
15
|
Noor L, Upadhyay A, Joshi V. Role of T Lymphocytes in Glioma Immune Microenvironment: Two Sides of a Coin. BIOLOGY 2024; 13:846. [PMID: 39452154 PMCID: PMC11505600 DOI: 10.3390/biology13100846] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/11/2024] [Revised: 10/16/2024] [Accepted: 10/18/2024] [Indexed: 10/26/2024]
Abstract
Glioma is known for its immunosuppressive microenvironment, which makes it challenging to target through immunotherapies. Immune cells like macrophages, microglia, myeloid-derived suppressor cells, and T lymphocytes are known to infiltrate the glioma tumor microenvironment and regulate immune response distinctively. Among the variety of immune cells, T lymphocytes have highly complex and multifaceted roles in the glioma immune landscape. T lymphocytes, which include CD4+ helper and CD8+ cytotoxic T cells, are known for their pivotal roles in anti-tumor responses. However, these cells may behave differently in the highly dynamic glioma microenvironment, for example, via an immune invasion mechanism enforced by tumor cells. Therefore, T lymphocytes play dual roles in glioma immunity, firstly by their anti-tumor responses, and secondly by exploiting gliomas to promote immune invasion. As an immunosuppression strategy, glioma induces T-cell exhaustion and suppression of effector T cells by regulatory T cells (Tregs) or by altering their signaling pathways. Further, the expression of immune checkpoint inhibitors on the glioma cell surface leads to T cell anergy and dysfunction. Overall, this dynamic interplay between T lymphocytes and glioma is crucial for designing more effective immunotherapies. The current review provides detailed knowledge on the roles of T lymphocytes in the glioma immune microenvironment and helps to explore novel therapeutic approaches to reinvigorate T lymphocytes.
Collapse
Affiliation(s)
- Laiba Noor
- Department of Biotechnology, Bennett University, Greater Noida 201310, Uttar Pradesh, India
| | - Arun Upadhyay
- Department of Bioscience and Biomedical Engineering, Indian Institute of Technology Bhilai, Durg 491002, Chhattisgarh, India
| | - Vibhuti Joshi
- Department of Biotechnology, Bennett University, Greater Noida 201310, Uttar Pradesh, India
| |
Collapse
|
16
|
Curry RN, Ma Q, McDonald MF, Ko Y, Srivastava S, Chin PS, He P, Lozzi B, Athukuri P, Jing J, Wang S, Harmanci AO, Arenkiel B, Jiang X, Deneen B, Rao G, Serin Harmanci A. Integrated electrophysiological and genomic profiles of single cells reveal spiking tumor cells in human glioma. Cancer Cell 2024; 42:1713-1728.e6. [PMID: 39241781 PMCID: PMC11479845 DOI: 10.1016/j.ccell.2024.08.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/04/2023] [Revised: 04/15/2024] [Accepted: 08/08/2024] [Indexed: 09/09/2024]
Abstract
Prior studies have described the complex interplay that exists between glioma cells and neurons; however, the electrophysiological properties endogenous to glioma cells remain obscure. To address this, we employed Patch-sequencing (Patch-seq) on human glioma specimens and found that one-third of patched cells in IDH mutant (IDHmut) tumors demonstrate properties of both neurons and glia. To define these hybrid cells (HCs), which fire single, short action potentials, and discern if they are of tumoral origin, we developed the single cell rule association mining (SCRAM) computational tool to annotate each cell individually. SCRAM revealed that HCs possess select features of GABAergic neurons and oligodendrocyte precursor cells, and include both tumor and non-tumor cells. These studies characterize the combined electrophysiological and molecular properties of human glioma cells and describe a cell type in human glioma with unique electrophysiological and transcriptomic properties that may also exist in the non-tumor brain.
Collapse
Affiliation(s)
- Rachel N Curry
- The Integrative Molecular and Biomedical Sciences Graduate Program, Baylor College of Medicine, Houston, TX, USA; Center for Cell and Gene Therapy, Baylor College of Medicine, Houston, TX, USA
| | - Qianqian Ma
- Jan and Dan Duncan Neurological Research Institute at Texas Children's Hospital, Houston, TX, USA; Department of Neuroscience, Baylor College of Medicine, Houston, TX, USA
| | - Malcolm F McDonald
- Center for Cell and Gene Therapy, Baylor College of Medicine, Houston, TX, USA; Medical Scientist Training Program, Baylor College of Medicine, Houston, TX, USA; Program in Development, Disease Models, and Therapeutics, Baylor College of Medicine, Houston, TX, USA
| | - Yeunjung Ko
- Center for Cell and Gene Therapy, Baylor College of Medicine, Houston, TX, USA; Center for Cancer Neuroscience, Baylor College of Medicine, Houston, TX, USA; Department of Neurosurgery, Baylor College of Medicine, Houston, TX, USA
| | - Snigdha Srivastava
- Jan and Dan Duncan Neurological Research Institute at Texas Children's Hospital, Houston, TX, USA; Medical Scientist Training Program, Baylor College of Medicine, Houston, TX, USA; Department of Human and Molecular Genetics, Baylor College of Medicine, Houston, TX, USA
| | - Pey-Shyuan Chin
- Department of Neuroscience, Baylor College of Medicine, Houston, TX, USA
| | - Peihao He
- Center for Cell and Gene Therapy, Baylor College of Medicine, Houston, TX, USA; Center for Cancer Neuroscience, Baylor College of Medicine, Houston, TX, USA; Program in Cancer Cell Biology, Baylor College of Medicine, Houston, TX, USA
| | - Brittney Lozzi
- Program in Genetics and Genomics, Baylor College of Medicine, Houston, TX, USA
| | - Prazwal Athukuri
- Department of Neurosurgery, Baylor College of Medicine, Houston, TX, USA
| | - Junzhan Jing
- Jan and Dan Duncan Neurological Research Institute at Texas Children's Hospital, Houston, TX, USA; Department of Neuroscience, Baylor College of Medicine, Houston, TX, USA
| | - Su Wang
- Department of Neurosurgery, Baylor College of Medicine, Houston, TX, USA
| | - Arif O Harmanci
- McWilliams School of Biomedical Informatics, University of Texas Health Science Center, Houston, TX, USA
| | - Benjamin Arenkiel
- Department of Neuroscience, Baylor College of Medicine, Houston, TX, USA; Department of Human and Molecular Genetics, Baylor College of Medicine, Houston, TX, USA
| | - Xiaolong Jiang
- Jan and Dan Duncan Neurological Research Institute at Texas Children's Hospital, Houston, TX, USA; Department of Neuroscience, Baylor College of Medicine, Houston, TX, USA; Department of Ophthalmology, Baylor College of Medicine, Houston, TX, USA.
| | - Benjamin Deneen
- The Integrative Molecular and Biomedical Sciences Graduate Program, Baylor College of Medicine, Houston, TX, USA; Center for Cell and Gene Therapy, Baylor College of Medicine, Houston, TX, USA; Program in Development, Disease Models, and Therapeutics, Baylor College of Medicine, Houston, TX, USA; Center for Cancer Neuroscience, Baylor College of Medicine, Houston, TX, USA; Department of Neurosurgery, Baylor College of Medicine, Houston, TX, USA; Program in Cancer Cell Biology, Baylor College of Medicine, Houston, TX, USA.
| | - Ganesh Rao
- Center for Cancer Neuroscience, Baylor College of Medicine, Houston, TX, USA; Department of Neurosurgery, Baylor College of Medicine, Houston, TX, USA.
| | - Akdes Serin Harmanci
- Center for Cancer Neuroscience, Baylor College of Medicine, Houston, TX, USA; Department of Neurosurgery, Baylor College of Medicine, Houston, TX, USA.
| |
Collapse
|
17
|
Lv W, Wang Y. Neural Influences on Tumor Progression Within the Central Nervous System. CNS Neurosci Ther 2024; 30:e70097. [PMID: 39469896 PMCID: PMC11519750 DOI: 10.1111/cns.70097] [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: 07/08/2024] [Revised: 09/21/2024] [Accepted: 10/13/2024] [Indexed: 10/30/2024] Open
Abstract
For decades, researchers have studied how brain tumors, the immune system, and drugs interact. With the advances in cancer neuroscience, which centers on defining and therapeutically targeting nervous system-cancer interactions, both within the local tumor microenvironment (TME) and on a systemic level, the subtle relationship between neurons and tumors in the central nervous system (CNS) has been deeply studied. Neurons, as the executors of brain functional activities, have been shown to significantly influence the emergence and development of brain tumors, including both primary and metastatic tumors. They engage with tumor cells via chemical or electrical synapses, directly regulating tumors or via intricate coupling networks, and also contribute to the TME through paracrine signaling, secreting proteins that exert regulatory effects. For instance, in a study involving a mouse model of glioblastoma, the authors observed a 42% increase in tumor volume when neuronal activity was stimulated, compared to controls (p < 0.01), indicating a direct correlation between neural activity and tumor growth. These thought-provoking results offer promising new strategies for brain tumor therapies, highlighting the potential of neuronal modulation to curb tumor progression. Future strategies may focus on developing drugs to inhibit or neutralize proteins and other bioactive substances secreted by neurons, break synaptic connections and interactions between infiltrating cells and tumor cells, as well as disrupt electrical coupling within glioma cell networks. By harnessing the insights gained from this research, we aspire to usher in a new era of brain tumor therapies that are both more potent and precise.
Collapse
Affiliation(s)
- Wenhao Lv
- Affiliated Hospital of Hangzhou Normal UniversityHangzhou Normal UniversityHangzhouZhejiangChina
- School of PharmacyHangzhou Normal UniversityHangzhouZhejiangChina
| | - Yongjie Wang
- School of PharmacyHangzhou Normal UniversityHangzhouZhejiangChina
| |
Collapse
|
18
|
Pacal I. A novel Swin transformer approach utilizing residual multi-layer perceptron for diagnosing brain tumors in MRI images. INT J MACH LEARN CYB 2024; 15:3579-3597. [DOI: 10.1007/s13042-024-02110-w] [Citation(s) in RCA: 24] [Impact Index Per Article: 24.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2023] [Accepted: 01/24/2024] [Indexed: 05/14/2025]
Abstract
AbstractSerious consequences due to brain tumors necessitate a timely and accurate diagnosis. However, obstacles such as suboptimal imaging quality, issues with data integrity, varying tumor types and stages, and potential errors in interpretation hinder the achievement of precise and prompt diagnoses. The rapid identification of brain tumors plays a pivotal role in ensuring patient safety. Deep learning-based systems hold promise in aiding radiologists to make diagnoses swiftly and accurately. In this study, we present an advanced deep learning approach based on the Swin Transformer. The proposed method introduces a novel Hybrid Shifted Windows Multi-Head Self-Attention module (HSW-MSA) along with a rescaled model. This enhancement aims to improve classification accuracy, reduce memory usage, and simplify training complexity. The Residual-based MLP (ResMLP) replaces the traditional MLP in the Swin Transformer, thereby improving accuracy, training speed, and parameter efficiency. We evaluate the Proposed-Swin model on a publicly available brain MRI dataset with four classes, using only test data. Model performance is enhanced through the application of transfer learning and data augmentation techniques for efficient and robust training. The Proposed-Swin model achieves a remarkable accuracy of 99.92%, surpassing previous research and deep learning models. This underscores the effectiveness of the Swin Transformer with HSW-MSA and ResMLP improvements in brain tumor diagnosis. This method introduces an innovative diagnostic approach using HSW-MSA and ResMLP in the Swin Transformer, offering potential support to radiologists in timely and accurate brain tumor diagnosis, ultimately improving patient outcomes and reducing risks.
Collapse
|
19
|
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
|
20
|
Wang Y, Suo J, Wang Z, Ran K, Tian Y, Han W, Liu Y, Peng X. The PTPRZ1-MET/STAT3/ISG20 axis in glioma stem-like cells modulates tumor-associated macrophage polarization. Cell Signal 2024; 120:111191. [PMID: 38685521 DOI: 10.1016/j.cellsig.2024.111191] [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: 03/13/2024] [Revised: 04/19/2024] [Accepted: 04/24/2024] [Indexed: 05/02/2024]
Abstract
Recent studies have revealed that PTPRZ1-MET (ZM) fusion plays a pivotal role in the progression of glioma to glioblastoma multiforme (GBM), thus serving as a biomarker to distinguish between primary GBM and secondary GBM (sGBM). However, the mechanisms through which ZM fusion influences this progression remain to be elucidated. GBMs with ZM showed poorer prognoses and greater infiltration of tumor-associated macrophages (TAMs) than those without ZM. Glioma stem-like cells (GSCs) and TAMs play complex roles in glioma recurrence, glioma progression and therapy resistance. In this study, we analyzed RNA-seq data from sGBM patients' glioma tissues with or without ZM fusion, and found that stemness and macrophage markers were more highly expressed in sGBM patients harboring ZM than in those without ZM fusion. ZM enhanced the self-renewal and proliferation of GSCs, thereby accelerating glioma progression. In addition, ZM-positive GSCs facilitated the infiltration of TAMs and drove their polarization toward an immunosuppressive phenotype, which was primarily accomplished through the extracellular secretion of ISG20. Our research identified the MET-STAT3-ISG20 axis within GSCs, thus demonstrating the critical role of ZM in GBM initiation and progression. Our study demonstrated that, in contrast to ZM-positive differentiated glioma cells, ZM-positive GSCs upregulated ISG20 expression through the MET-STAT3-ISG20 axis. The extracellular secretion of ISG20 recruited and induced M2-like polarization in macrophages, thereby promoting tumor progression. Our results reveal a novel mechanism involved in ZM-positive GBM pathogenesis and identify potential therapeutic targets.
Collapse
Affiliation(s)
- Yuxin Wang
- Department of Molecular Biology and Biochemistry, Medical Primate Research Center, Neuroscience Center, Institute of Basic Medical Sciences Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical College, Beijing 100005, China; State Key Laboratory of Common Mechanism Research for Major Diseases, Institute of Basic Medical Sciences Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical College, Beijing 100005, China
| | - Jinghao Suo
- Department of Molecular Biology and Biochemistry, Medical Primate Research Center, Neuroscience Center, Institute of Basic Medical Sciences Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical College, Beijing 100005, China; State Key Laboratory of Common Mechanism Research for Major Diseases, Institute of Basic Medical Sciences Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical College, Beijing 100005, China
| | - Zhixing Wang
- Department of Molecular Biology and Biochemistry, Medical Primate Research Center, Neuroscience Center, Institute of Basic Medical Sciences Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical College, Beijing 100005, China; State Key Laboratory of Common Mechanism Research for Major Diseases, Institute of Basic Medical Sciences Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical College, Beijing 100005, China
| | - Kunnian Ran
- Department of Molecular Biology and Biochemistry, Medical Primate Research Center, Neuroscience Center, Institute of Basic Medical Sciences Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical College, Beijing 100005, China; State Key Laboratory of Common Mechanism Research for Major Diseases, Institute of Basic Medical Sciences Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical College, Beijing 100005, China
| | - Yuan Tian
- Department of Molecular Biology and Biochemistry, Medical Primate Research Center, Neuroscience Center, Institute of Basic Medical Sciences Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical College, Beijing 100005, China; State Key Laboratory of Common Mechanism Research for Major Diseases, Institute of Basic Medical Sciences Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical College, Beijing 100005, China
| | - Wei Han
- Department of Molecular Biology and Biochemistry, Medical Primate Research Center, Neuroscience Center, Institute of Basic Medical Sciences Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical College, Beijing 100005, China; State Key Laboratory of Common Mechanism Research for Major Diseases, Institute of Basic Medical Sciences Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical College, Beijing 100005, China.
| | - Yanwei Liu
- Department of Radiation Oncology, Beijing Tiantan Hospital, Capital Medical University, Beijing 100070, China.
| | - Xiaozhong Peng
- Department of Molecular Biology and Biochemistry, Medical Primate Research Center, Neuroscience Center, Institute of Basic Medical Sciences Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical College, Beijing 100005, China; State Key Laboratory of Respiratory Health and Multimorbidity, Institute of Laboratory Animal Science, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100021, China.
| |
Collapse
|
21
|
Abidin ZU, Naqvi RA, Haider A, Kim HS, Jeong D, Lee SW. Recent deep learning-based brain tumor segmentation models using multi-modality magnetic resonance imaging: a prospective survey. Front Bioeng Biotechnol 2024; 12:1392807. [PMID: 39104626 PMCID: PMC11298476 DOI: 10.3389/fbioe.2024.1392807] [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: 02/28/2024] [Accepted: 06/14/2024] [Indexed: 08/07/2024] Open
Abstract
Radiologists encounter significant challenges when segmenting and determining brain tumors in patients because this information assists in treatment planning. The utilization of artificial intelligence (AI), especially deep learning (DL), has emerged as a useful tool in healthcare, aiding radiologists in their diagnostic processes. This empowers radiologists to understand the biology of tumors better and provide personalized care to patients with brain tumors. The segmentation of brain tumors using multi-modal magnetic resonance imaging (MRI) images has received considerable attention. In this survey, we first discuss multi-modal and available magnetic resonance imaging modalities and their properties. Subsequently, we discuss the most recent DL-based models for brain tumor segmentation using multi-modal MRI. We divide this section into three parts based on the architecture: the first is for models that use the backbone of convolutional neural networks (CNN), the second is for vision transformer-based models, and the third is for hybrid models that use both convolutional neural networks and transformer in the architecture. In addition, in-depth statistical analysis is performed of the recent publication, frequently used datasets, and evaluation metrics for segmentation tasks. Finally, open research challenges are identified and suggested promising future directions for brain tumor segmentation to improve diagnostic accuracy and treatment outcomes for patients with brain tumors. This aligns with public health goals to use health technologies for better healthcare delivery and population health management.
Collapse
Affiliation(s)
- Zain Ul Abidin
- Department of Intelligent Mechatronics Engineering, Sejong University, Seoul, Republic of Korea
| | - Rizwan Ali Naqvi
- Department of Intelligent Mechatronics Engineering, Sejong University, Seoul, Republic of Korea
| | - Amir Haider
- Department of Intelligent Mechatronics Engineering, Sejong University, Seoul, Republic of Korea
| | - Hyung Seok Kim
- Department of Intelligent Mechatronics Engineering, Sejong University, Seoul, Republic of Korea
| | - Daesik Jeong
- College of Convergence Engineering, Sangmyung University, Seoul, Republic of Korea
| | - Seung Won Lee
- School of Medicine, Sungkyunkwan University, Suwon, Republic of Korea
| |
Collapse
|
22
|
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
|
23
|
White J, White MPJ, Wickremesekera A, Peng L, Gray C. The tumour microenvironment, treatment resistance and recurrence in glioblastoma. J Transl Med 2024; 22:540. [PMID: 38844944 PMCID: PMC11155041 DOI: 10.1186/s12967-024-05301-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2024] [Accepted: 05/13/2024] [Indexed: 06/10/2024] Open
Abstract
The adaptability of glioblastoma (GBM) cells, encouraged by complex interactions with the tumour microenvironment (TME), currently renders GBM an incurable cancer. Despite intensive research, with many clinical trials, GBM patients rely on standard treatments including surgery followed by radiation and chemotherapy, which have been observed to induce a more aggressive phenotype in recurrent tumours. This failure to improve treatments is undoubtedly a result of insufficient models which fail to incorporate components of the human brain TME. Research has increasingly uncovered mechanisms of tumour-TME interactions that correlate to worsened patient prognoses, including tumour-associated astrocyte mitochondrial transfer, neuronal circuit remodelling and immunosuppression. This tumour hijacked TME is highly implicated in driving therapy resistance, with further alterations within the TME and tumour resulting from therapy exposure inducing increased tumour growth and invasion. Recent developments improving organoid models, including aspects of the TME, are paving an exciting future for the research and drug development for GBM, with the hopes of improving patient survival growing closer. This review focuses on GBMs interactions with the TME and their effect on tumour pathology and treatment efficiency, with a look at challenges GBM models face in sufficiently recapitulating this complex and highly adaptive cancer.
Collapse
Affiliation(s)
- Jasmine White
- Gillies McIndoe Research Institute, Newtown, Wellington, 6021, New Zealand
- Centre for Biodiscovery and School of Biological Sciences, Victoria University of Wellington, Kelburn, Wellington, 6021, New Zealand
| | | | - Agadha Wickremesekera
- Gillies McIndoe Research Institute, Newtown, Wellington, 6021, New Zealand
- Department of Neurosurgery, Wellington Regional Hospital, Wellington, New Zealand
| | - Lifeng Peng
- Centre for Biodiscovery and School of Biological Sciences, Victoria University of Wellington, Kelburn, Wellington, 6021, New Zealand.
| | - Clint Gray
- Gillies McIndoe Research Institute, Newtown, Wellington, 6021, New Zealand.
- Centre for Biodiscovery and School of Biological Sciences, Victoria University of Wellington, Kelburn, Wellington, 6021, New Zealand.
| |
Collapse
|
24
|
Peddinti V, Rout B, Agnihotri TG, Gomte SS, Jain A. Functionalized liposomes: an enticing nanocarrier for management of glioma. J Liposome Res 2024; 34:349-367. [PMID: 37855432 DOI: 10.1080/08982104.2023.2270060] [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: 06/06/2023] [Accepted: 10/07/2023] [Indexed: 10/20/2023]
Abstract
Glioma is one of the most severe central nervous systems (CNS)-specific tumors, with rapidly growing malignant glial cells accounting for roughly half of all brain tumors and having a poor survival rate ranging from 12 to 15 months. Despite being the most often used technique for glioma therapy, conventional chemotherapy suffers from low permeability of the blood-brain barrier (BBB) and blood-brain tumor barrier (BBTB) to anticancer drugs. When it comes to nanocarriers, liposomes are thought of as one of the most promising nanocarrier systems for glioma treatment. However, owing to BBB tight junctions, non-targeted liposomes, which passively accumulate in most cancer cells primarily via the increased permeability and retention effect (EPR), would not be suitable for glioma treatment. The surface modification of liposomes with various active targeting ligands has shown encouraging outcomes in the recent times by allowing various chemotherapy drugs to pass across the BBB and BBTB and enter glioma cells. This review article introduces by briefly outlining the landscape of glioma, its classification, and some of the pathogenic causes. Further, it discusses major barriers for delivering drugs to glioma such as the BBB, BBTB, and tumor microenvironment. It further discusses modified liposomes such as long-acting circulating liposomes, actively targeted liposomes, stimuli responsive liposomes. Finally, it highlighted the limitations of liposomes in the treatment of glioma and the various actively targeted liposomes undergoing clinical trials for the treatment of glioma.
Collapse
Affiliation(s)
- Vasu Peddinti
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER)-Ahmedabad, Gandhinagar, Gujarat, India
| | - Biswajit Rout
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER)-Ahmedabad, Gandhinagar, Gujarat, India
| | - Tejas Girish Agnihotri
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER)-Ahmedabad, Gandhinagar, Gujarat, India
| | - Shyam Sudhakar Gomte
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER)-Ahmedabad, Gandhinagar, Gujarat, India
| | - Aakanchha Jain
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER)-Ahmedabad, Gandhinagar, Gujarat, India
| |
Collapse
|
25
|
Chakraborty A, Yang C, Kresak JL, Silver AJ, Feier D, Tian G, Andrews M, Sobanjo OO, Hodge ED, Engelbart MK, Huang J, Harrison JK, Sarkisian MR, Mitchell DA, Deleyrolle LP. KR158 Spheres Harboring Slow-Cycling Cells Recapitulate High-Grade Glioma Features in an Immunocompetent System. Cells 2024; 13:938. [PMID: 38891070 PMCID: PMC11171638 DOI: 10.3390/cells13110938] [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] [Revised: 05/20/2024] [Accepted: 05/24/2024] [Indexed: 06/21/2024] Open
Abstract
Glioblastoma (GBM) poses a significant challenge in clinical oncology due to its aggressive nature, heterogeneity, and resistance to therapies. Cancer stem cells (CSCs) play a critical role in GBM, particularly in treatment resistance and tumor relapse, emphasizing the need to comprehend the mechanisms regulating these cells. Also, their multifaceted contributions to the tumor microenvironment (TME) underline their significance, driven by their unique properties. This study aimed to characterize glioblastoma stem cells (GSCs), specifically slow-cycling cells (SCCs), in an immunocompetent murine GBM model to explore their similarities with their human counterparts. Using the KR158 mouse model, we confirmed that SCCs isolated from this model exhibited key traits and functional properties akin to human SCCs. KR158 murine SCCs, expanded in the gliomasphere assay, demonstrated sphere forming ability, self-renewing capacity, positive tumorigenicity, enhanced stemness and resistance to chemotherapy. Together, our findings validate the KR158 murine model as a framework to investigate GSCs and SCCs in GBM pathology, and explore specifically the SCC-immune system communications, understand their role in disease progression, and evaluate the effect of therapeutic strategies targeting these specific connections.
Collapse
Affiliation(s)
- Avirup Chakraborty
- Adam Michael Rosen Neuro-Oncology Laboratories, Department of Neurosurgery, University of Florida, Gainesville, FL 32608, USA (A.J.S.)
- Preston A. Wells Jr. Center for Brain Tumor Therapy, University of Florida, Gainesville, FL 32608, USA
| | - Changlin Yang
- Adam Michael Rosen Neuro-Oncology Laboratories, Department of Neurosurgery, University of Florida, Gainesville, FL 32608, USA (A.J.S.)
- Preston A. Wells Jr. Center for Brain Tumor Therapy, University of Florida, Gainesville, FL 32608, USA
| | - Jesse L. Kresak
- Department of Pathology, Immunology and Laboratory Medicine, University of Florida, Gainesville, FL 32610, USA
| | - Aryeh J. Silver
- Adam Michael Rosen Neuro-Oncology Laboratories, Department of Neurosurgery, University of Florida, Gainesville, FL 32608, USA (A.J.S.)
| | - Diana Feier
- Adam Michael Rosen Neuro-Oncology Laboratories, Department of Neurosurgery, University of Florida, Gainesville, FL 32608, USA (A.J.S.)
| | - Guimei Tian
- Department of Surgery, University of Florida, Gainesville, FL 32610, USA
| | - Michael Andrews
- College of Dental Medicine, Nova Southeastern University, Fort Lauderdale, FL 33314, USA
| | - Olusegun O. Sobanjo
- Adam Michael Rosen Neuro-Oncology Laboratories, Department of Neurosurgery, University of Florida, Gainesville, FL 32608, USA (A.J.S.)
| | - Ethan D. Hodge
- Adam Michael Rosen Neuro-Oncology Laboratories, Department of Neurosurgery, University of Florida, Gainesville, FL 32608, USA (A.J.S.)
| | - Mia K. Engelbart
- Adam Michael Rosen Neuro-Oncology Laboratories, Department of Neurosurgery, University of Florida, Gainesville, FL 32608, USA (A.J.S.)
| | - Jianping Huang
- Adam Michael Rosen Neuro-Oncology Laboratories, Department of Neurosurgery, University of Florida, Gainesville, FL 32608, USA (A.J.S.)
- Preston A. Wells Jr. Center for Brain Tumor Therapy, University of Florida, Gainesville, FL 32608, USA
| | - Jeffrey K. Harrison
- Preston A. Wells Jr. Center for Brain Tumor Therapy, University of Florida, Gainesville, FL 32608, USA
- Department of Pharmacology and Therapeutics, University of Florida, Gainesville, FL 32603, USA
| | - Matthew R. Sarkisian
- Preston A. Wells Jr. Center for Brain Tumor Therapy, University of Florida, Gainesville, FL 32608, USA
- Department of Neuroscience, McKnight Brain Institute, University of Florida, Gainesville, FL 32610, USA
| | - Duane A. Mitchell
- Adam Michael Rosen Neuro-Oncology Laboratories, Department of Neurosurgery, University of Florida, Gainesville, FL 32608, USA (A.J.S.)
- Preston A. Wells Jr. Center for Brain Tumor Therapy, University of Florida, Gainesville, FL 32608, USA
| | - Loic P. Deleyrolle
- Adam Michael Rosen Neuro-Oncology Laboratories, Department of Neurosurgery, University of Florida, Gainesville, FL 32608, USA (A.J.S.)
- Preston A. Wells Jr. Center for Brain Tumor Therapy, University of Florida, Gainesville, FL 32608, USA
- Department of Neuroscience, McKnight Brain Institute, University of Florida, Gainesville, FL 32610, USA
| |
Collapse
|
26
|
Ruan X, Yan W, Cao M, Daza RAM, Fong MY, Yang K, Wu J, Liu X, Palomares M, Wu X, Li A, Chen Y, Jandial R, Spitzer NC, Hevner RF, Wang SE. Breast cancer cell-secreted miR-199b-5p hijacks neurometabolic coupling to promote brain metastasis. Nat Commun 2024; 15:4549. [PMID: 38811525 PMCID: PMC11137082 DOI: 10.1038/s41467-024-48740-0] [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: 12/12/2022] [Accepted: 05/09/2024] [Indexed: 05/31/2024] Open
Abstract
Breast cancer metastasis to the brain is a clinical challenge rising in prevalence. However, the underlying mechanisms, especially how cancer cells adapt a distant brain niche to facilitate colonization, remain poorly understood. A unique metabolic feature of the brain is the coupling between neurons and astrocytes through glutamate, glutamine, and lactate. Here we show that extracellular vesicles from breast cancer cells with a high potential to develop brain metastases carry high levels of miR-199b-5p, which shows higher levels in the blood of breast cancer patients with brain metastases comparing to those with metastatic cancer in other organs. miR-199b-5p targets solute carrier transporters (SLC1A2/EAAT2 in astrocytes and SLC38A2/SNAT2 and SLC16A7/MCT2 in neurons) to hijack the neuron-astrocyte metabolic coupling, leading to extracellular retention of these metabolites and promoting cancer cell growth. Our findings reveal a mechanism through which cancer cells of a non-brain origin reprogram neural metabolism to fuel brain metastases.
Collapse
Affiliation(s)
- Xianhui Ruan
- Department of Pathology, University of California San Diego, La Jolla, CA, USA
| | - Wei Yan
- Department of Pathology, University of California San Diego, La Jolla, CA, USA
| | - Minghui Cao
- Department of Pathology, University of California San Diego, La Jolla, CA, USA
| | - Ray Anthony M Daza
- Department of Pathology, University of California San Diego, La Jolla, CA, USA
| | - Miranda Y Fong
- Department of Pathology, University of California San Diego, La Jolla, CA, USA
- Department of Cancer Biology, City of Hope Beckman Research Institute, Duarte, CA, USA
| | - Kaifu Yang
- School of Biological Sciences, University of California San Diego, La Jolla, CA, USA
| | - Jun Wu
- Center for Comparative Medicine, City of Hope Beckman Research Institute, Duarte, CA, USA
| | - Xuxiang Liu
- Department of Cancer Biology, City of Hope Beckman Research Institute, Duarte, CA, USA
| | | | - Xiwei Wu
- Department of Computational and Quantitative Medicine, City of Hope Beckman Research Institute, Duarte, CA, USA
| | - Arthur Li
- Division of Biostatistics, City of Hope Beckman Research Institute, Duarte, CA, USA
| | - Yuan Chen
- Department of Surgery, University of California San Diego, La Jolla, CA, USA
- Moores Cancer Center, University of California San Diego, La Jolla, CA, USA
| | - Rahul Jandial
- Department of Surgery; City of Hope, Duarte, CA, USA
| | - Nicholas C Spitzer
- Neurobiology Department, School of Biological Sciences and Center for Neural Circuits and Behavior, University of California San Diego, La Jolla, CA, USA
- Kavli Institute for Brain and Mind, University of California San Diego, La Jolla, CA, USA
| | - Robert F Hevner
- Department of Pathology, University of California San Diego, La Jolla, CA, USA
| | - Shizhen Emily Wang
- Department of Pathology, University of California San Diego, La Jolla, CA, USA.
- Moores Cancer Center, University of California San Diego, La Jolla, CA, USA.
| |
Collapse
|
27
|
Peres N, Lepski GA, Fogolin CS, Evangelista GCM, Flatow EA, de Oliveira JV, Pinho MP, Bergami-Santos PC, Barbuto JAM. Profiling of Tumor-Infiltrating Immune Cells and Their Impact on Survival in Glioblastoma Patients Undergoing Immunotherapy with Dendritic Cells. Int J Mol Sci 2024; 25:5275. [PMID: 38791312 PMCID: PMC11121326 DOI: 10.3390/ijms25105275] [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] [Revised: 05/07/2024] [Accepted: 05/08/2024] [Indexed: 05/26/2024] Open
Abstract
Glioblastomas (GBM) are the most common primary malignant brain tumors, comprising 2% of all cancers in adults. Their location and cellular and molecular heterogeneity, along with their highly infiltrative nature, make their treatment challenging. Recently, our research group reported promising results from a prospective phase II clinical trial involving allogeneic vaccination with dendritic cells (DCs). To date, six out of the thirty-seven reported cases remain alive without tumor recurrence. In this study, we focused on the characterization of infiltrating immune cells observed at the time of surgical resection. An analytical model employing a neural network-based predictive algorithm was used to ascertain the potential prognostic implications of immunological variables on patients' overall survival. Counterintuitively, immune phenotyping of tumor-associated macrophages (TAMs) has revealed the extracellular marker PD-L1 to be a positive predictor of overall survival. In contrast, the elevated expression of CD86 within this cellular subset emerged as a negative prognostic indicator. Fundamentally, the neural network algorithm outlined here allows a prediction of the responsiveness of patients undergoing dendritic cell vaccination in terms of overall survival based on clinical parameters and the profile of infiltrated TAMs observed at the time of tumor excision.
Collapse
Affiliation(s)
- Nataly Peres
- Department of Psychiatry, Medical School, Universidade de Sao Paulo, Sao Paulo 05403-010, Brazil;
| | - Guilherme A. Lepski
- LIM 26, Hospital das Clínicas HCFMUSP, Faculdade de Medicina, Universidade de Sao Paulo, Sao Paulo 05403-000, Brazil
- Department of Neurosurgery, Eberhard-Karls University, 72074 Tuebingen, Germany
| | - Carla S. Fogolin
- Department of Immunology, Instituto de Ciencias Biomedicas, Universidade de Sao Paulo, Sao Paulo 05508-000, Brazil; (C.S.F.); (G.C.M.E.); (E.A.F.); (J.V.d.O.); (M.P.P.); (P.C.B.-S.); (J.A.M.B.)
- Laboratory of Medical Investigation in Pathogenesis and Targeted Therapy in Onco-Immuno-Hematology (LIM-31), Department of Hematology, Hospital das Clínicas HCFMUSP, Faculdade de Medicina, Universidade de Sao Paulo, Sao Paulo 05403-000, Brazil
| | - Gabriela C. M. Evangelista
- Department of Immunology, Instituto de Ciencias Biomedicas, Universidade de Sao Paulo, Sao Paulo 05508-000, Brazil; (C.S.F.); (G.C.M.E.); (E.A.F.); (J.V.d.O.); (M.P.P.); (P.C.B.-S.); (J.A.M.B.)
- Laboratory of Medical Investigation in Pathogenesis and Targeted Therapy in Onco-Immuno-Hematology (LIM-31), Department of Hematology, Hospital das Clínicas HCFMUSP, Faculdade de Medicina, Universidade de Sao Paulo, Sao Paulo 05403-000, Brazil
| | - Elizabeth A. Flatow
- Department of Immunology, Instituto de Ciencias Biomedicas, Universidade de Sao Paulo, Sao Paulo 05508-000, Brazil; (C.S.F.); (G.C.M.E.); (E.A.F.); (J.V.d.O.); (M.P.P.); (P.C.B.-S.); (J.A.M.B.)
- Laboratory of Medical Investigation in Pathogenesis and Targeted Therapy in Onco-Immuno-Hematology (LIM-31), Department of Hematology, Hospital das Clínicas HCFMUSP, Faculdade de Medicina, Universidade de Sao Paulo, Sao Paulo 05403-000, Brazil
| | - Jaqueline V. de Oliveira
- Department of Immunology, Instituto de Ciencias Biomedicas, Universidade de Sao Paulo, Sao Paulo 05508-000, Brazil; (C.S.F.); (G.C.M.E.); (E.A.F.); (J.V.d.O.); (M.P.P.); (P.C.B.-S.); (J.A.M.B.)
- Laboratory of Medical Investigation in Pathogenesis and Targeted Therapy in Onco-Immuno-Hematology (LIM-31), Department of Hematology, Hospital das Clínicas HCFMUSP, Faculdade de Medicina, Universidade de Sao Paulo, Sao Paulo 05403-000, Brazil
| | - Mariana P. Pinho
- Department of Immunology, Instituto de Ciencias Biomedicas, Universidade de Sao Paulo, Sao Paulo 05508-000, Brazil; (C.S.F.); (G.C.M.E.); (E.A.F.); (J.V.d.O.); (M.P.P.); (P.C.B.-S.); (J.A.M.B.)
| | - Patricia C. Bergami-Santos
- Department of Immunology, Instituto de Ciencias Biomedicas, Universidade de Sao Paulo, Sao Paulo 05508-000, Brazil; (C.S.F.); (G.C.M.E.); (E.A.F.); (J.V.d.O.); (M.P.P.); (P.C.B.-S.); (J.A.M.B.)
| | - José A. M. Barbuto
- Department of Immunology, Instituto de Ciencias Biomedicas, Universidade de Sao Paulo, Sao Paulo 05508-000, Brazil; (C.S.F.); (G.C.M.E.); (E.A.F.); (J.V.d.O.); (M.P.P.); (P.C.B.-S.); (J.A.M.B.)
- Laboratory of Medical Investigation in Pathogenesis and Targeted Therapy in Onco-Immuno-Hematology (LIM-31), Department of Hematology, Hospital das Clínicas HCFMUSP, Faculdade de Medicina, Universidade de Sao Paulo, Sao Paulo 05403-000, Brazil
| |
Collapse
|
28
|
Curry RN, Ma Q, McDonald MF, Ko Y, Srivastava S, Chin PS, He P, Lozzi B, Athukuri P, Jing J, Wang S, Harmanci AO, Arenkiel B, Jiang X, Deneen B, Rao G, Harmanci AS. Integrated electrophysiological and genomic profiles of single cells reveal spiking tumor cells in human glioma. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.03.02.583026. [PMID: 38496434 PMCID: PMC10942290 DOI: 10.1101/2024.03.02.583026] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/19/2024]
Abstract
Prior studies have described the complex interplay that exists between glioma cells and neurons, however, the electrophysiological properties endogenous to tumor cells remain obscure. To address this, we employed Patch-sequencing on human glioma specimens and found that one third of patched cells in IDH mutant (IDH mut ) tumors demonstrate properties of both neurons and glia by firing single, short action potentials. To define these hybrid cells (HCs) and discern if they are tumor in origin, we developed a computational tool, Single Cell Rule Association Mining (SCRAM), to annotate each cell individually. SCRAM revealed that HCs represent tumor and non-tumor cells that feature GABAergic neuron and oligodendrocyte precursor cell signatures. These studies are the first to characterize the combined electrophysiological and molecular properties of human glioma cells and describe a new cell type in human glioma with unique electrophysiological and transcriptomic properties that are likely also present in the non-tumor mammalian brain.
Collapse
|
29
|
Kim D, Ko HY, Chung JI, Park YM, Lee S, Kim SY, Kim J, Chun JH, Han KS, Lee M, Ju YH, Park SJ, Park KD, Nam MH, Kim SH, Shim JK, Park Y, Lim H, Park J, Lee GH, Kim H, Kim S, Park U, Ryu H, Lee SY, Park S, Kang SG, Chang JH, Lee CJ, Yun M. Visualizing cancer-originating acetate uptake through monocarboxylate transporter 1 in reactive astrocytes in the glioblastoma tumor microenvironment. Neuro Oncol 2024; 26:843-857. [PMID: 38085571 PMCID: PMC11066945 DOI: 10.1093/neuonc/noad243] [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] [Indexed: 05/04/2024] Open
Abstract
BACKGROUND Reactive astrogliosis is a hallmark of various brain pathologies, including neurodegenerative diseases and glioblastomas. However, the specific intermediate metabolites contributing to reactive astrogliosis remain unknown. This study investigated how glioblastomas induce reactive astrogliosis in the neighboring microenvironment and explore 11C-acetate PET as an imaging technique for detecting reactive astrogliosis. METHODS Through in vitro, mouse models, and human tissue experiments, we examined the association between elevated 11C-acetate uptake and reactive astrogliosis in gliomas. We explored acetate from glioblastoma cells, which triggers reactive astrogliosis in neighboring astrocytes by upregulating MAO-B and monocarboxylate transporter 1 (MCT1) expression. We evaluated the presence of cancer stem cells in the reactive astrogliosis region of glioblastomas and assessed the correlation between the volume of 11C-acetate uptake beyond MRI and prognosis. RESULTS Elevated 11C-acetate uptake is associated with reactive astrogliosis and astrocytic MCT1 in the periphery of glioblastomas in human tissues and mouse models. Glioblastoma cells exhibit increased acetate production as a result of glucose metabolism, with subsequent secretion of acetate. Acetate derived from glioblastoma cells induces reactive astrogliosis in neighboring astrocytes by increasing the expression of MAO-B and MCT1. We found cancer stem cells within the reactive astrogliosis at the tumor periphery. Consequently, a larger volume of 11C-acetate uptake beyond contrast-enhanced MRI was associated with a worse prognosis. CONCLUSIONS Our results highlight the role of acetate derived from glioblastoma cells in inducing reactive astrogliosis and underscore the potential value of 11C-acetate PET as an imaging technique for detecting reactive astrogliosis, offering important implications for the diagnosis and treatment of glioblastomas.
Collapse
Affiliation(s)
- Dongwoo Kim
- Department of Nuclear Medicine, Severance Hospital, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Hae Young Ko
- Department of Nuclear Medicine, Severance Hospital, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Jee-In Chung
- Department of Nuclear Medicine, Severance Hospital, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Yongmin Mason Park
- Center for Cognition and Sociality, Institute for Basic Science, Daejeon, Republic of Korea
- IBS School, University of Science and Technology, Daejeon, Republic of Korea
| | - Sangwon Lee
- Department of Nuclear Medicine, Severance Hospital, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Seon Yoo Kim
- Department of Nuclear Medicine, Severance Hospital, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Jisu Kim
- Department of Nuclear Medicine, Severance Hospital, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Joong-Hyun Chun
- Department of Nuclear Medicine, Severance Hospital, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Kyung-Seok Han
- Department of Biological Sciences, Chungnam National University, Daejeon, Republic of Korea
| | - Misu Lee
- Division of Life Science, College of Life Science and Bioengineering, Incheon National University, Incheon, Republic of Korea
| | - Yeon Ha Ju
- Center for Cognition and Sociality, Institute for Basic Science, Daejeon, Republic of Korea
- IBS School, University of Science and Technology, Daejeon, Republic of Korea
- Brain Science Institute, Korea Institute of Science and Technology (KIST), Seoul, Republic of Korea
| | - Sun Jun Park
- Brain Science Institute, Korea Institute of Science and Technology (KIST), Seoul, Republic of Korea
- Division of Bio-Med Science & Technology, KIST School, Korea University of Science and Technology, Seoul, Republic of Korea
| | - Ki Duk Park
- Brain Science Institute, Korea Institute of Science and Technology (KIST), Seoul, Republic of Korea
- Division of Bio-Med Science & Technology, KIST School, Korea University of Science and Technology, Seoul, Republic of Korea
| | - Min-Ho Nam
- Brain Science Institute, Korea Institute of Science and Technology (KIST), Seoul, Republic of Korea
- Division of Bio-Med Science & Technology, KIST School, Korea University of Science and Technology, Seoul, Republic of Korea
- Department of KHU-KIST Convergence Science and Technology, Kyung Hee University, Seoul, Republic of Korea
| | - Se Hoon Kim
- Department of Pathology, Severance Hospital, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Jin-Kyoung Shim
- Department of Neurosurgery, Severance Hospital, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Youngjoo Park
- Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Hyunkeong Lim
- Department of Nuclear Medicine, Severance Hospital, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Jaekyung Park
- Department of Nuclear Medicine, Severance Hospital, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Gwan-Ho Lee
- Research Resources Division, Korea Institute of Science and Technology (KIST), Seoul, Republic of Korea
| | - Hyunjin Kim
- Brain Science Institute, Korea Institute of Science and Technology (KIST), Seoul, Republic of Korea
| | - Suhyun Kim
- K-Laboratory, Brain Science Institute, Korea Institute of Science and Technology (KIST), Seoul, Republic of Korea
| | - Uiyeol Park
- K-Laboratory, Brain Science Institute, Korea Institute of Science and Technology (KIST), Seoul, Republic of Korea
| | - Hoon Ryu
- K-Laboratory, Brain Science Institute, Korea Institute of Science and Technology (KIST), Seoul, Republic of Korea
| | - So Yun Lee
- Natural Products Research Institute, College of Pharmacy, Seoul National University, Seoul, Republic of Korea
| | - Sunghyouk Park
- Natural Products Research Institute, College of Pharmacy, Seoul National University, Seoul, Republic of Korea
| | - Seok-Gu Kang
- Department of Neurosurgery, Severance Hospital, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Jong Hee Chang
- Department of Neurosurgery, Severance Hospital, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - C Justin Lee
- IBS School, University of Science and Technology, Daejeon, Republic of Korea
| | - Mijin Yun
- Department of Nuclear Medicine, Severance Hospital, Yonsei University College of Medicine, Seoul, Republic of Korea
| |
Collapse
|
30
|
Scuderi SA, Ardizzone A, Salako AE, Pantò G, De Luca F, Esposito E, Capra AP. Pentraxin 3: A Main Driver of Inflammation and Immune System Dysfunction in the Tumor Microenvironment of Glioblastoma. Cancers (Basel) 2024; 16:1637. [PMID: 38730589 PMCID: PMC11083335 DOI: 10.3390/cancers16091637] [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: 03/21/2024] [Revised: 04/12/2024] [Accepted: 04/22/2024] [Indexed: 05/13/2024] Open
Abstract
Brain tumors are a heterogeneous group of brain neoplasms that are highly prevalent in individuals of all ages worldwide. Within this pathological framework, the most prevalent and aggressive type of primary brain tumor is glioblastoma (GB), a subtype of glioma that falls within the IV-grade astrocytoma group. The death rate for patients with GB remains high, occurring within a few months after diagnosis, even with the gold-standard therapies now available, such as surgery, radiation, or a pharmaceutical approach with Temozolomide. For this reason, it is crucial to continue looking for cutting-edge therapeutic options to raise patients' survival chances. Pentraxin 3 (PTX3) is a multifunctional protein that has a variety of regulatory roles in inflammatory processes related to extracellular matrix (ECM). An increase in PTX3 blood levels is considered a trustworthy factor associated with the beginning of inflammation. Moreover, scientific evidence suggested that PTX3 is a sensitive and earlier inflammation-related marker compared to the short pentraxin C-reactive protein (CRP). In several tumoral subtypes, via regulating complement-dependent and macrophage-associated tumor-promoting inflammation, it has been demonstrated that PTX3 may function as a promoter of cancer metastasis, invasion, and stemness. Our review aims to deeply evaluate the function of PTX3 in the pathological context of GB, considering its pivotal biological activities and its possible role as a molecular target for future therapies.
Collapse
Affiliation(s)
- Sarah Adriana Scuderi
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Viale Ferdinando Stagno D’Alcontres, 31, 98166 Messina, Italy; (S.A.S.); (A.A.); (A.E.S.); (F.D.L.); (A.P.C.)
| | - Alessio Ardizzone
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Viale Ferdinando Stagno D’Alcontres, 31, 98166 Messina, Italy; (S.A.S.); (A.A.); (A.E.S.); (F.D.L.); (A.P.C.)
| | - Ayomide Eniola Salako
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Viale Ferdinando Stagno D’Alcontres, 31, 98166 Messina, Italy; (S.A.S.); (A.A.); (A.E.S.); (F.D.L.); (A.P.C.)
- University of Florence, 50121 Florence, Italy
| | - Giuseppe Pantò
- Department of Biomedical and Dental Sciences and Morphofunctional Imaging, University of Messina, Via Consolare Valeria 1, 98125 Messina, Italy;
| | - Fabiola De Luca
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Viale Ferdinando Stagno D’Alcontres, 31, 98166 Messina, Italy; (S.A.S.); (A.A.); (A.E.S.); (F.D.L.); (A.P.C.)
| | - Emanuela Esposito
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Viale Ferdinando Stagno D’Alcontres, 31, 98166 Messina, Italy; (S.A.S.); (A.A.); (A.E.S.); (F.D.L.); (A.P.C.)
| | - Anna Paola Capra
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Viale Ferdinando Stagno D’Alcontres, 31, 98166 Messina, Italy; (S.A.S.); (A.A.); (A.E.S.); (F.D.L.); (A.P.C.)
| |
Collapse
|
31
|
Ten A, Kumeiko V, Farniev V, Gao H, Shevtsov M. Tumor Microenvironment Modulation by Cancer-Derived Extracellular Vesicles. Cells 2024; 13:682. [PMID: 38667297 PMCID: PMC11049026 DOI: 10.3390/cells13080682] [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: 02/11/2024] [Revised: 04/06/2024] [Accepted: 04/11/2024] [Indexed: 04/28/2024] Open
Abstract
The tumor microenvironment (TME) plays an important role in the process of tumorigenesis, regulating the growth, metabolism, proliferation, and invasion of cancer cells, as well as contributing to tumor resistance to the conventional chemoradiotherapies. Several types of cells with relatively stable phenotypes have been identified within the TME, including cancer-associated fibroblasts (CAFs), tumor-associated macrophages (TAMs), neutrophils, and natural killer (NK) cells, which have been shown to modulate cancer cell proliferation, metastasis, and interaction with the immune system, thus promoting tumor heterogeneity. Growing evidence suggests that tumor-cell-derived extracellular vesicles (EVs), via the transfer of various molecules (e.g., RNA, proteins, peptides, and lipids), play a pivotal role in the transformation of normal cells in the TME into their tumor-associated protumorigenic counterparts. This review article focuses on the functions of EVs in the modulation of the TME with a view to how exosomes contribute to the transformation of normal cells, as well as their importance for cancer diagnosis and therapy.
Collapse
Affiliation(s)
- Artem Ten
- School of Medicine and Life Sciences, Far Eastern Federal University, 690922 Vladivostok, Russia; (A.T.); (V.K.); (V.F.)
| | - Vadim Kumeiko
- School of Medicine and Life Sciences, Far Eastern Federal University, 690922 Vladivostok, Russia; (A.T.); (V.K.); (V.F.)
| | - Vladislav Farniev
- School of Medicine and Life Sciences, Far Eastern Federal University, 690922 Vladivostok, Russia; (A.T.); (V.K.); (V.F.)
| | - Huile Gao
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu 610064, China;
| | - Maxim Shevtsov
- School of Medicine and Life Sciences, Far Eastern Federal University, 690922 Vladivostok, Russia; (A.T.); (V.K.); (V.F.)
- Laboratory of Biomedical Nanotechnologies, Institute of Cytology of the Russian Academy of Sciences, Tikhoretsky Ave., 4, 194064 St. Petersburg, Russia
- Personalized Medicine Centre, Almazov National Medical Research Centre, Akkuratova Str., 2, 197341 St. Petersburg, Russia
- Department of Radiation Oncology, Technishe Universität München (TUM), Klinikum Rechts der Isar, Ismaninger Str., 22, 81675 Munich, Germany
| |
Collapse
|
32
|
Pallarés-Moratalla C, Bergers G. The ins and outs of microglial cells in brain health and disease. Front Immunol 2024; 15:1305087. [PMID: 38665919 PMCID: PMC11043497 DOI: 10.3389/fimmu.2024.1305087] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2023] [Accepted: 03/19/2024] [Indexed: 04/28/2024] Open
Abstract
Microglia are the brain's resident macrophages that play pivotal roles in immune surveillance and maintaining homeostasis of the Central Nervous System (CNS). Microglia are functionally implicated in various cerebrovascular diseases, including stroke, aneurysm, and tumorigenesis as they regulate neuroinflammatory responses and tissue repair processes. Here, we review the manifold functions of microglia in the brain under physiological and pathological conditions, primarily focusing on the implication of microglia in glioma propagation and progression. We further review the current status of therapies targeting microglial cells, including their re-education, depletion, and re-population approaches as therapeutic options to improve patient outcomes for various neurological and neuroinflammatory disorders, including cancer.
Collapse
|
33
|
Chakraborty A, Yang C, Kresak JL, Silver A, Feier D, Tian G, Andrews M, Sobanjo OO, Hodge ED, Engelbart MK, Huang J, Harrison JK, Sarkisian MR, Mitchell DA, Deleyrolle LP. KR158 spheres harboring slow-cycling cells recapitulate GBM features in an immunocompetent system. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.01.26.577279. [PMID: 38501121 PMCID: PMC10945590 DOI: 10.1101/2024.01.26.577279] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/20/2024]
Abstract
Glioblastoma (GBM) poses a significant challenge in clinical oncology due to its aggressive nature, heterogeneity, and resistance to therapies. Cancer stem cells (CSCs) play a critical role in GBM, particularly in treatment-resistance and tumor relapse, emphasizing the need to comprehend the mechanisms regulating these cells. Also, their multifaceted contributions to the tumor-microenvironment (TME) underline their significance, driven by their unique properties. This study aimed to characterize glioblastoma stem cells (GSCs), specifically slow-cycling cells (SCCs), in an immunocompetent murine GBM model to explore their similarities with their human counterparts. Using the KR158 mouse model, we confirmed that SCCs isolated from this model exhibited key traits and functional properties akin to human SCCs. KR158 murine SCCs, expanded in the gliomasphere assay, demonstrated sphere forming ability, self-renewing capacity, positive tumorigenicity, enhanced stemness and resistance to chemotherapy. Together, our findings validate the KR158 murine model as a framework to investigate GSCs and SCCs in GBM-pathology, and explore specifically the SCC-immune system communications, understand their role in disease progression, and evaluate the effect of therapeutic strategies targeting these specific connections.
Collapse
|
34
|
Nóbrega AHL, Pimentel RS, Prado AP, Garcia J, Frozza RL, Bernardi A. Neuroinflammation in Glioblastoma: The Role of the Microenvironment in Tumour Progression. Curr Cancer Drug Targets 2024; 24:579-594. [PMID: 38310461 DOI: 10.2174/0115680096265849231031101449] [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: 06/30/2023] [Revised: 08/25/2023] [Accepted: 09/08/2023] [Indexed: 02/05/2024]
Abstract
Glioblastoma (GBM) stands as the most aggressive and lethal among the main types of primary brain tumors. It exhibits malignant growth, infiltrating the brain tissue, and displaying resistance toward treatment. GBM is a complex disease characterized by high degrees of heterogeneity. During tumour growth, microglia and astrocytes, among other cells, infiltrate the tumour microenvironment and contribute extensively to gliomagenesis. Tumour-associated macrophages (TAMs), either of peripheral origin or representing brain-intrinsic microglia, are the most numerous nonneoplastic populations in the tumour microenvironment in GBM. The complex heterogeneous nature of GBM cells is facilitated by the local inflammatory tumour microenvironment, which mostly induces tumour aggressiveness and drug resistance. The immunosuppressive tumour microenvironment of GBM provides multiple pathways for tumour immune evasion, contributing to tumour progression. Additionally, TAMs and astrocytes can contribute to tumour progression through the release of cytokines and activation of signalling pathways. In this review, we summarize the role of the microenvironment in GBM progression, focusing on neuroinflammation. These recent advancements in research of the microenvironment hold the potential to offer a promising approach to the treatment of GBM in the coming times.
Collapse
Affiliation(s)
| | - Rafael Sampaio Pimentel
- Laboratory of Inflammation, Oswaldo Cruz Institute, Oswaldo Cruz Foundation (FIOCRUZ), Rio de Janeiro/RJ, Brazil
| | - Ana Paula Prado
- Laboratory of Inflammation, Oswaldo Cruz Institute, Oswaldo Cruz Foundation (FIOCRUZ), Rio de Janeiro/RJ, Brazil
| | - Jenifer Garcia
- Laboratory of Inflammation, Oswaldo Cruz Institute, Oswaldo Cruz Foundation (FIOCRUZ), Rio de Janeiro/RJ, Brazil
| | - Rudimar Luiz Frozza
- Laboratory on Thymus Research, Oswaldo Cruz Institute, Oswaldo Cruz Foundation (FIOCRUZ), Rio de Janeiro/RJ, Brazil
| | - Andressa Bernardi
- Laboratory of Inflammation, Oswaldo Cruz Institute, Oswaldo Cruz Foundation (FIOCRUZ), Rio de Janeiro/RJ, Brazil
| |
Collapse
|
35
|
Martinez PJ, Green AL, Borden MA. Targeting diffuse midline gliomas: The promise of focused ultrasound-mediated blood-brain barrier opening. J Control Release 2024; 365:412-421. [PMID: 38000663 PMCID: PMC10842695 DOI: 10.1016/j.jconrel.2023.11.037] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2023] [Revised: 11/13/2023] [Accepted: 11/18/2023] [Indexed: 11/26/2023]
Abstract
Diffuse midline gliomas (DMGs), including diffuse intrinsic pontine glioma, have among the highest mortality rates of all childhood cancers, despite recent advancements in cancer therapeutics. This is partly because, unlike some CNS tumors, the blood-brain barrier (BBB) of DMG tumor vessels remains intact. The BBB prevents the permeation of many molecular therapies into the brain parenchyma, where the cancer cells reside. Focused ultrasound (FUS) with microbubbles has recently emerged as an innovative and exciting technology that non-invasively permeabilizes the BBB in a small focal region with millimeter precision. In this review, current treatment methods and biological barriers to treating DMGs are discussed. State-of-the-art FUS-mediated BBB opening is then examined, with a focus on the effects of various ultrasound parameters and the treatment of DMGs.
Collapse
Affiliation(s)
- Payton J Martinez
- Biomedical Engineering Program, University of Colorado Boulder, Boulder, CO 80303, United States; Department of Mechanical Engineering, University of Colorado Boulder, Boulder, CO 80303, United States.
| | - Adam L Green
- Morgan Adams Foundation Pediatric Brain Tumor Research Program, Department of Pediatrics, University of Colorado School of Medicine, Aurora, CO 80045, United States
| | - Mark A Borden
- Biomedical Engineering Program, University of Colorado Boulder, Boulder, CO 80303, United States; Department of Mechanical Engineering, University of Colorado Boulder, Boulder, CO 80303, United States
| |
Collapse
|
36
|
Al-Hawary SIS, Alhajlah S, Olegovich BD, Hjazi A, Rajput P, Ali SHJ, Abosoda M, Ihsan A, Oudah SK, Mustafa YF. Effective extracellular vesicles in glioma: Focusing on effective ncRNA exosomes and immunotherapy methods for treatment. Cell Biochem Funct 2024; 42:e3921. [PMID: 38269511 DOI: 10.1002/cbf.3921] [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: 11/02/2023] [Revised: 12/26/2023] [Accepted: 12/27/2023] [Indexed: 01/26/2024]
Abstract
This comprehensive article explores the complex field of glioma treatment, with a focus on the important roles of non-coding RNAsRNAs (ncRNAs) and exosomes, as well as the potential synergies of immunotherapy. The investigation begins by examining the various functions of ncRNAs and their involvement in glioma pathogenesis, progression, and as potential diagnostic biomarkers. Special attention is given to exosomes as carriers of ncRNAs and their intricate dynamics within the tumor microenvironment. The exploration extends to immunotherapy methods, analyzing their mechanisms and clinical implications in the treatment of glioma. By synthesizing these components, the article aims to provide a comprehensive understanding of how ncRNAs, exosomes, and immunotherapy interact, offering valuable insights into the evolving landscape of glioma research and therapeutic strategies.
Collapse
Affiliation(s)
| | - Sharif Alhajlah
- Department of Medical Laboratories, College of Applied Medical Sciences, Shaqra University, Shaqraa, Saudi Arabia
| | - Bokov Dmitry Olegovich
- Institute of Pharmacy, Sechenov First Moscow State Medical University, Moscow, Russian Federation
- Laboratory of Food Chemistry, Federal Research Center of Nutrition, Biotechnology and Food Safety, Moscow, Russian Federation
| | - Ahmed Hjazi
- Department of Medical Laboratory Sciences, College of Applied Medical Sciences, Prince Sattam bin Abdulaziz University, Al-Kharj, Saudi Arabia
| | - Pranchal Rajput
- School of Applied and Life Sciences, Uttaranchal University, Dehradun, India
| | - Saad Hayif Jasim Ali
- Department of Medical Laboratory, College of Health and Medical Technology, Al-Ayen University, Thi-Qar, Iraq
| | - Munther Abosoda
- College of Pharmacy, The Islamic University, Najaf, Iraq
- College of Pharmacy, The Islamic University of Al Diwaniyah, Al Diwaniyah, Iraq
- College of Pharmacy, The Islamic University of Babylon, Babylon, Iraq
| | - Ali Ihsan
- Department of Medical Laboratories Techniques, Imam Ja'afar Al-Sadiq University, Iraq
| | - Shamam Kareem Oudah
- College of Pharmacy, National University of Science and Technology, Dhi Qar, Iraq
| | - Yasser Fakri Mustafa
- Department of Pharmaceutical Chemistry, College of Pharmacy, University of Mosul, Mosul, Iraq
| |
Collapse
|
37
|
Sun L, Jiang Y, Tan H, Liang R. Collagen and derivatives-based materials as substrates for the establishment of glioblastoma organoids. Int J Biol Macromol 2024; 254:128018. [PMID: 37967599 DOI: 10.1016/j.ijbiomac.2023.128018] [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: 10/09/2023] [Revised: 10/31/2023] [Accepted: 11/09/2023] [Indexed: 11/17/2023]
Abstract
Glioblastoma (GBM) is a common primary brain malignancy known for its ability to invade the brain, resistance to chemotherapy and radiotherapy, tendency to recur frequently, and unfavorable prognosis. Attempts have been undertaken to create 2D and 3D models, such as glioblastoma organoids (GBOs), to recapitulate the glioma microenvironment, explore tumor biology, and develop efficient therapies. However, these models have limitations and are unable to fully recapitulate the complex networks formed by the glioma microenvironment that promote tumor cell growth, invasion, treatment resistance, and immune escape. Therefore, it is necessary to develop advanced experimental models that could better simulate clinical physiology. Here, we review recent advances in natural biomaterials (mainly focus on collagen and its derivatives)-based GBO models, as in vitro experimental platforms to simulate GBM tumor biology and response to tested drugs. Special attention will be given to 3D models that use collagen, gelatin, further modified derivatives, and composite biomaterials (e.g., with other natural or synthetic polymers) as substrates. Application of these collagen/derivatives-constructed GBOs incorporate the physical as well as chemical characteristics of the GBM microenvironment. A perspective on future research is given in terms of current issues. Generally, natural materials based on collagen/derivatives (monomers or composites) are expected to enrich the toolbox of GBO modeling substrates and potentially help to overcome the limitations of existing models.
Collapse
Affiliation(s)
- Lu Sun
- Department of Targeting Therapy & Immunology; Department of Radiation Oncology, Cancer Center, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Yuelin Jiang
- West China Hospital, Sichuan University, Chengdu 610041, China.
| | - Hong Tan
- College of Polymer Science and Engineering, State Key Laboratory of Polymer Materials Engineering, Sichuan University, Chengdu 610065, China.
| | - Ruichao Liang
- Department of Neurosurgery, West China Hospital, Sichuan University, Chengdu 610041, China.
| |
Collapse
|
38
|
Panada J, Klopava V, Kulahava T, Koran S, Faletrov Y, Frolova N, Fomina E, Shkumatov V. Differential induction of C6 glioma apoptosis and autophagy by 3β-hydroxysteroid-indolamine conjugates. Steroids 2023; 200:109326. [PMID: 37827441 DOI: 10.1016/j.steroids.2023.109326] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/17/2023] [Revised: 09/28/2023] [Accepted: 10/09/2023] [Indexed: 10/14/2023]
Abstract
In a previous work, we reported the synthesis of four novel indole steroids and their effect on rat C6 glioma proliferation in vitro. The steroid derived from dehydroepiandrosterone and tryptamine (IS-1) was the most active (52 % inhibition at 10 µM), followed by one of the epimers derived from pregnenolone and tryptamine (IS-3, 36 % inhibition at 10 µM). By contrast, the steroid derived from estrone and tryptamine (IS-2) showed negligible activity at 10 µM. No necrosis, increase in intracellular calcium or ROS levels was observed. In this work, the effect of compounds on C6 glioma apoptosis and autophagy is examined by fluorimetry and fluorescent microscopy. The IS-3 epimers disrupt the mitochondrial membrane potential and induce apoptosis in vitro moderately whereas IS-1 and IS-2 do not. However, IS-1 produces a large increase in monodansylcadaverine-positive autophagic vesicles over 24 h. The antiproliferative effect of indole steroids is ameliorated by autophagy inhibitor hydroxychloroquine, suggesting an autophagy-dependent mechanism of cell death.
Collapse
Affiliation(s)
- Jan Panada
- Research Institute for Physical Chemical Problems of the Belarusian State University, 220006, 14 Lieninhradskaja str., Minsk, Belarus
| | - Valeriya Klopava
- Research Institute for Physical Chemical Problems of the Belarusian State University, 220006, 14 Lieninhradskaja str., Minsk, Belarus
| | - Tatsiana Kulahava
- Institute for Nuclear Problems of the Belarusian State University, 220006, 11 Babrujskaja str., Minsk, Belarus
| | - Siarhei Koran
- Republican Research and Practical Center for Epidemiology and Microbiology, 220114, 23 Filimonava str., Minsk, Belarus
| | - Yaroslav Faletrov
- Research Institute for Physical Chemical Problems of the Belarusian State University, 220006, 14 Lieninhradskaja str., Minsk, Belarus; Department of Chemistry, Belarusian State University, 220050, 4 Independence ave., Minsk, Belarus
| | - Nina Frolova
- Research Institute for Physical Chemical Problems of the Belarusian State University, 220006, 14 Lieninhradskaja str., Minsk, Belarus
| | - Elena Fomina
- Republican Research and Practical Center for Epidemiology and Microbiology, 220114, 23 Filimonava str., Minsk, Belarus
| | - Vladimir Shkumatov
- Research Institute for Physical Chemical Problems of the Belarusian State University, 220006, 14 Lieninhradskaja str., Minsk, Belarus; Department of Chemistry, Belarusian State University, 220050, 4 Independence ave., Minsk, Belarus.
| |
Collapse
|
39
|
Akins EA, Wilkins D, Aghi MK, Kumar S. An engineered glioblastoma model yields novel macrophage-secreted drivers of invasion. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.11.18.567683. [PMID: 38014161 PMCID: PMC10680873 DOI: 10.1101/2023.11.18.567683] [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
Glioblastomas (GBMs) are highly invasive brain tumors replete with brain- and blood-derived macrophages, collectively known as tumor-associated macrophages (TAMs). Targeting TAMs has been proposed as a therapeutic strategy but has thus far yielded limited clinical success in slowing GBM progression, due in part to an incomplete understanding of TAM function in GBM. Here, by using an engineered hyaluronic acid-based 3D invasion platform, patient-derived GBM cells, and multi-omics analysis of GBM tumor microenvironments, we show that M2-polarized macrophages stimulate GBM stem cell (GSC) mesenchymal transition and invasion. We identify TAM-derived transforming growth factor beta induced (TGFβI/BIGH3) as a pro-tumorigenic factor in the GBM microenvironment. In GBM patients, BIGH3 mRNA expression correlates with poor patient prognosis and is highest in the most aggressive GBM molecular subtype. Inhibiting TAM-derived BIGH3 signaling with a blocking antibody or small molecule inhibitor suppresses GSC invasion. Our work highlights the utility of 3D in vitro tumor microenvironment platforms to investigate TAM-cancer cell crosstalk and offers new insights into TAM function to guide novel TAM-targeting therapies.
Collapse
Affiliation(s)
- Erin A. Akins
- University of California, Berkeley – University of California, San Francisco Graduate Program in Bioengineering, Berkeley, CA, USA
- Department of Bioengineering, University of California, Berkeley, Berkeley, CA 94720, USA
| | - Dana Wilkins
- University of California, Berkeley – University of California, San Francisco Graduate Program in Bioengineering, Berkeley, CA, USA
- Department of Bioengineering, University of California, Berkeley, Berkeley, CA 94720, USA
| | - Manish K. Aghi
- Department of Neurosurgery; University of California San Francisco (UCSF)
| | - Sanjay Kumar
- University of California, Berkeley – University of California, San Francisco Graduate Program in Bioengineering, Berkeley, CA, USA
- Department of Bioengineering, University of California, Berkeley, Berkeley, CA 94720, USA
- Department of Chemical and Biomolecular Engineering, University of California, Berkeley, Berkeley, CA 94720, USA
| |
Collapse
|
40
|
Trevisi G, Mangiola A. Current Knowledge about the Peritumoral Microenvironment in Glioblastoma. Cancers (Basel) 2023; 15:5460. [PMID: 38001721 PMCID: PMC10670229 DOI: 10.3390/cancers15225460] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2023] [Revised: 10/31/2023] [Accepted: 11/15/2023] [Indexed: 11/26/2023] Open
Abstract
Glioblastoma is a deadly disease, with a mean overall survival of less than 2 years from diagnosis. Recurrence after gross total surgical resection and adjuvant chemo-radiotherapy almost invariably occurs within the so-called peritumoral brain zone (PBZ). The aim of this narrative review is to summarize the most relevant findings about the biological characteristics of the PBZ currently available in the medical literature. The PBZ presents several peculiar biological characteristics. The cellular landscape of this area is different from that of healthy brain tissue and is characterized by a mixture of cell types, including tumor cells (seen in about 30% of cases), angiogenesis-related endothelial cells, reactive astrocytes, glioma-associated microglia/macrophages (GAMs) with anti-inflammatory polarization, tumor-infiltrating lymphocytes (TILs) with an "exhausted" phenotype, and glioma-associated stromal cells (GASCs). From a genomic and transcriptomic point of view, compared with the tumor core and healthy brain tissue, the PBZ presents a "half-way" pattern with upregulation of genes related to angiogenesis, the extracellular matrix, and cellular senescence and with stemness features and downregulation in tumor suppressor genes. This review illustrates that the PBZ is a transition zone with a pre-malignant microenvironment that constitutes the base for GBM progression/recurrence. Understanding of the PBZ could be relevant to developing more effective treatments to prevent GBM development and recurrence.
Collapse
Affiliation(s)
- Gianluca Trevisi
- Department of Neurosciences, Imaging and Clinical Sciences, G. D’Annunzio University Chieti-Pescara, 66100 Chieti, Italy;
- Neurosurgical Unit, Ospedale Spirito Santo, 65122 Pescara, Italy
| | - Annunziato Mangiola
- Department of Neurosciences, Imaging and Clinical Sciences, G. D’Annunzio University Chieti-Pescara, 66100 Chieti, Italy;
| |
Collapse
|
41
|
Shi B, Ge F, Cai L, Yang Y, Guo X, Wu R, Fan Z, Cao B, Wang N, Si Y, Lin X, Dong W, Sun H. Significance of NotchScore and JAG1 in predicting prognosis and immune response of low-grade glioma. Front Immunol 2023; 14:1247288. [PMID: 38022677 PMCID: PMC10679421 DOI: 10.3389/fimmu.2023.1247288] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2023] [Accepted: 10/30/2023] [Indexed: 12/01/2023] Open
Abstract
Introduction Low-grade glioma (LGG) is a prevalent malignant tumor in the intracranial region. Despite the advancements in treatment methods for this malignancy over the past decade, significant challenges still persist in the form of drug resistance and tumor recurrence. The Notch signaling pathway plays essential roles in many physiological processes as well as in cancer development. However, the significance of the pathway and family genes in LGG are poorly understood. Methods We conducted gene expression profiling analysis using the TCGA dataset to investigate the gene set associated with the Notch signaling pathway. we have proposed a metric called "NotchScore" that quantifies the strength of the Notch signaling pathway and enables us to assess its significance in predicting prognosis and immune response in LGG. We downregulated JAG1 in low-grade gliomas to assess its influence on the proliferation and migration of these tumors. Ultimately, we determined the impact of the transcription factor VDR on the transcription of PDL1 through chip-seq data analysis. Results Our findings indicate that tumors with a higher NotchScore, exhibit poorer prognosis, potentially due to their ability to evade the anti-tumor effects of immune cells by expressing immune checkpoints. Among the genes involved in the Notch signaling pathway, JAG1 has emerged as the most representative in terms of capturing the characteristics of both NotchScore and Notch pathways. The experimental results demonstrate that silencing JAG1 yielded a significant decrease in tumor cell proliferation in LGG cell lines. Our study revealed mechanisms by which tumors evade the immune system through the modulation of PDL1 transcription levels via the PI3K-Akt signaling pathway. Additionally, JAG1 potentially influences PDL1 in LGG by regulating the PI3K-Akt signaling pathway and the expression of the transcription factor VDR. Discussion These findings contribute to our understanding of immune evasion by tumors in LGG. The insights gained from this research may have implications for the development of therapeutic interventions for LGG.
Collapse
Affiliation(s)
- Bo Shi
- Institute of Translational Medicine, Medical College, Yangzhou University, Yangzhou, China
- School of Life Science, Liaoning Normal University, Dalian, Liaoning, China
- Jiangsu Key Laboratory of Experimental & Translational Non-Coding RNA Research, Yangzhou, Jiangsu, China
| | - Fei Ge
- Department of Gastroenterology, Haian Hospital of Traditional Chinese Medicine Affiliated to Nanjing University of Chinese Medicine, Nantong, Jiangsu, China
| | - Liangliang Cai
- Institute of Translational Medicine, Medical College, Yangzhou University, Yangzhou, China
- Jiangsu Key Laboratory of Experimental & Translational Non-Coding RNA Research, Yangzhou, Jiangsu, China
| | - Yi Yang
- Institute of Translational Medicine, Medical College, Yangzhou University, Yangzhou, China
- Jiangsu Key Laboratory of Experimental & Translational Non-Coding RNA Research, Yangzhou, Jiangsu, China
| | - Xiaohui Guo
- School of Life Science, Liaoning Normal University, Dalian, Liaoning, China
| | - Rui Wu
- School of Life Science, Liaoning Normal University, Dalian, Liaoning, China
| | - Zhehao Fan
- Institute of Translational Medicine, Medical College, Yangzhou University, Yangzhou, China
- Jiangsu Key Laboratory of Experimental & Translational Non-Coding RNA Research, Yangzhou, Jiangsu, China
| | - Binjie Cao
- Institute of Translational Medicine, Medical College, Yangzhou University, Yangzhou, China
- Jiangsu Key Laboratory of Experimental & Translational Non-Coding RNA Research, Yangzhou, Jiangsu, China
| | - Ning Wang
- Institute of Translational Medicine, Medical College, Yangzhou University, Yangzhou, China
- Jiangsu Key Laboratory of Experimental & Translational Non-Coding RNA Research, Yangzhou, Jiangsu, China
| | - Yue Si
- Institute of Translational Medicine, Medical College, Yangzhou University, Yangzhou, China
- Jiangsu Key Laboratory of Experimental & Translational Non-Coding RNA Research, Yangzhou, Jiangsu, China
| | - Xinyue Lin
- Institute of Translational Medicine, Medical College, Yangzhou University, Yangzhou, China
- Jiangsu Key Laboratory of Experimental & Translational Non-Coding RNA Research, Yangzhou, Jiangsu, China
| | - Weibing Dong
- School of Life Science, Liaoning Normal University, Dalian, Liaoning, China
| | - Haibo Sun
- Institute of Translational Medicine, Medical College, Yangzhou University, Yangzhou, China
- Jiangsu Key Laboratory of Experimental & Translational Non-Coding RNA Research, Yangzhou, Jiangsu, China
| |
Collapse
|
42
|
Messiaen J, Jacobs SA, De Smet F. The tumor micro-environment in pediatric glioma: friend or foe? Front Immunol 2023; 14:1227126. [PMID: 37901250 PMCID: PMC10611473 DOI: 10.3389/fimmu.2023.1227126] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2023] [Accepted: 09/27/2023] [Indexed: 10/31/2023] Open
Abstract
Brain tumors are the leading cause of morbidity and mortality related to cancer in children, where high-grade glioma harbor the worst prognosis. It has become obvious that pediatric glioma differs significantly from their adult counterparts, rendering extrapolations difficult. Curative options for several types of glioma are lacking, albeit ongoing research efforts and clinical trials. As already proven in the past, inter- and intratumoral heterogeneity plays an important role in the resistance to therapy and thus implicates morbidity and mortality for these patients. However, while less studied, the tumor micro-environment (TME) adds another level of heterogeneity. Knowledge gaps exist on how the TME interacts with the tumor cells and how the location of the various cell types in the TME influences tumor growth and the response to treatment. Some studies identified the presence of several (immune) cell types as prognostic factors, but often lack a deeper understanding of the underlying mechanisms, possibly leading to contradictory findings. Although the TME in pediatric glioma is regarded as "cold", several treatment options are emerging, with the TME being the primary target of treatment. Therefore, it is crucial to study the TME of pediatric glioma, so that the interactions between TME, tumoral cells and therapeutics can be better understood before, during and after treatment. In this review, we provide an overview of the available insights into the composition and role of the TME across different types of pediatric glioma. Moreover, where possible, we provide a framework on how a particular TME may influence responses to conventional- and/or immunotherapy.
Collapse
Affiliation(s)
- Julie Messiaen
- Department of Pediatrics, University Hospitals Leuven, Leuven, Belgium
- Laboratory for Precision Cancer Medicine, Translational Cell and Tissue Research, Department of Imaging and Pathology, KU Leuven, Leuven, Belgium
| | - Sandra A. Jacobs
- Department of Pediatrics, University Hospitals Leuven, Leuven, Belgium
- Pediatric Oncology, Department of Oncology, KU Leuven, Leuven, Belgium
| | - Frederik De Smet
- Laboratory for Precision Cancer Medicine, Translational Cell and Tissue Research, Department of Imaging and Pathology, KU Leuven, Leuven, Belgium
| |
Collapse
|
43
|
Zeng J, Zeng XX. Systems Medicine for Precise Targeting of Glioblastoma. Mol Biotechnol 2023; 65:1565-1584. [PMID: 36859639 PMCID: PMC9977103 DOI: 10.1007/s12033-023-00699-x] [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: 10/03/2022] [Accepted: 02/14/2023] [Indexed: 03/03/2023]
Abstract
Glioblastoma (GBM) is a malignant cancer that is fatal even after standard therapy and the effects of current available therapeutics are not promising due its complex and evolving epigenetic and genetic profile. The mysteries that lead to GBM intratumoral heterogeneity and subtype transitions are not entirely clear. Systems medicine is an approach to view the patient in a whole picture integrating systems biology and synthetic biology along with computational techniques. Since the GBM oncogenesis involves genetic mutations, various therapies including gene therapeutics based on CRISPR-Cas technique, MicroRNAs, and implanted synthetic cells endowed with synthetic circuits against GBM with neural stem cells and mesenchymal stem cells acting as potential vehicles carrying therapeutics via the intranasal route, avoiding the risks of invasive methods in order to reach the GBM cells in the brain are discussed and proposed in this review. Systems medicine approach is a rather novel strategy, and since the GBM of a patient is complex and unique, thus to devise an individualized treatment strategy to tailor personalized multimodal treatments for the individual patient taking into account the phenotype of the GBM, the unique body health profile of the patient and individual responses according to the systems medicine concept might show potential to achieve optimum effects.
Collapse
Affiliation(s)
- Jie Zeng
- Benjoe Institute of Systems Bio-Engineering, High Technology Park, Xinbei District, Changzhou, 213022 Jiangsu People’s Republic of China
| | - Xiao Xue Zeng
- Department of Health Management, Centre of General Practice, The Seventh Affiliated Hospital, Southern Medical University, No. 28, Desheng Road Section, Liguan Road, Lishui Town, Nanhai District, Foshan, 528000 Guangdong People’s Republic of China
| |
Collapse
|
44
|
Mirarchi A, Albi E, Beccari T, Arcuri C. Microglia and Brain Disorders: The Role of Vitamin D and Its Receptor. Int J Mol Sci 2023; 24:11892. [PMID: 37569267 PMCID: PMC10419106 DOI: 10.3390/ijms241511892] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2023] [Revised: 07/17/2023] [Accepted: 07/23/2023] [Indexed: 08/13/2023] Open
Abstract
Accounting for 5-20% of the total glial cells present in the adult brain, microglia are involved in several functions: maintenance of the neural environment, response to injury and repair, immunesurveillance, cytokine secretion, regulation of phagocytosis, synaptic pruning, and sculpting postnatal neural circuits. Microglia contribute to some neurodevelopmental disorders, such as Nasu-Hakola disease (NHD), Tourette syndrome (TS), autism spectrum disorder (ASD), and schizophrenia. Moreover, microglial involvement in neurodegenerative diseases, such as Alzheimer's (AD) and Parkinson's (PD) diseases, has also been well established. During the last two decades, epidemiological and research studies have demonstrated the involvement of vitamin D3 (VD3) in the brain's pathophysiology. VD3 is a fat-soluble metabolite that is required for the proper regulation of many of the body's systems, as well as for normal human growth and development, and shows neurotrophic and neuroprotective actions and influences on neurotransmission and synaptic plasticity, playing a role in various neurological diseases. In order to better understand the exact mechanisms behind the diverse actions of VD3 in the brain, a large number of studies have been performed on isolated cells or tissues of the central nervous system (CNS). Here, we discuss the involvement of VD3 and microglia on neurodegeneration- and aging-related diseases.
Collapse
Affiliation(s)
- Alessandra Mirarchi
- Department of Medicine and Surgery, University of Perugia, 06123 Perugia, Italy;
| | - Elisabetta Albi
- Department of Pharmaceutical Sciences, University of Perugia, 06123 Perugia, Italy; (E.A.); (T.B.)
| | - Tommaso Beccari
- Department of Pharmaceutical Sciences, University of Perugia, 06123 Perugia, Italy; (E.A.); (T.B.)
| | - Cataldo Arcuri
- Department of Medicine and Surgery, University of Perugia, 06123 Perugia, Italy;
| |
Collapse
|
45
|
Cheng F, Su T, Zhou S, Liu X, Yang S, Lin S, Guo W, Zhu G. Single-dose injectable nanovaccine-in-hydrogel for robust immunotherapy of large tumors with abscopal effect. SCIENCE ADVANCES 2023; 9:eade6257. [PMID: 37450588 PMCID: PMC10348685 DOI: 10.1126/sciadv.ade6257] [Citation(s) in RCA: 30] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/28/2022] [Accepted: 06/09/2023] [Indexed: 07/18/2023]
Abstract
Current cancer immunotherapy [e.g., immune checkpoint blockade (ICB)] only benefits small subsets of patients, largely due to immunosuppressive tumor microenvironment (TME). In situ tumor vaccination can reduce TME immunosuppression and thereby improve cancer immunotherapy. Here, we present single-dose injectable (nanovaccines + ICBs)-in-hydrogel (NvIH) for robust immunotherapy of large tumors with abscopal effect. NvIH is thermo-responsive hydrogel co-encapsulated with ICB antibodies and novel polymeric nanoparticles loaded with three immunostimulatory agonists for Toll-like receptors 7/8/9 (TLR7/8/9) and stimulator of interferon genes (STING). Upon in situ tumor vaccination, NvIH undergoes rapid sol-to-gel transformation, prolongs tumor retention, sustains the release of immunotherapeutics, and reduces acute systemic inflammation. In multiple poorly immunogenic tumor models, single-dose NvIH reduces multitier TME immunosuppression, elicits potent TME and systemic innate and adaptive antitumor immunity with memory, and regresses both local (vaccinated) and distant large tumors with abscopal effect, including distant orthotopic glioblastoma. Overall, NvIH holds great potential for tumor immunotherapy.
Collapse
Affiliation(s)
- Furong Cheng
- Translational Medicine Center, The Second Affiliated Hospital, Guangzhou Medical University, Guangzhou 510260, China
- Department of Pharmaceutics and Center for Pharmaceutical Engineering and Sciences, Institute for Structural Biology and Drug Discovery, School of Pharmacy, The Developmental Therapeutics Program, Massey Cancer Center, Virginia Commonwealth University, Richmond, VA 23298, USA
| | - Ting Su
- Department of Pharmaceutics and Center for Pharmaceutical Engineering and Sciences, Institute for Structural Biology and Drug Discovery, School of Pharmacy, The Developmental Therapeutics Program, Massey Cancer Center, Virginia Commonwealth University, Richmond, VA 23298, USA
- Center for Translational Medicine, Precision Medicine Institute, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou 510080, China
| | - Shurong Zhou
- Department of Pharmaceutics and Center for Pharmaceutical Engineering and Sciences, Institute for Structural Biology and Drug Discovery, School of Pharmacy, The Developmental Therapeutics Program, Massey Cancer Center, Virginia Commonwealth University, Richmond, VA 23298, USA
- Department of Pharmaceutical Sciences, College of Pharmacy, Biointerfaces Institute, University of Michigan, Ann Arbor, MI 48109, USA
| | - Xiang Liu
- Department of Pharmaceutics and Center for Pharmaceutical Engineering and Sciences, Institute for Structural Biology and Drug Discovery, School of Pharmacy, The Developmental Therapeutics Program, Massey Cancer Center, Virginia Commonwealth University, Richmond, VA 23298, USA
- Department of Pharmaceutical Sciences, College of Pharmacy, Biointerfaces Institute, University of Michigan, Ann Arbor, MI 48109, USA
| | - Suling Yang
- Department of Pharmaceutical Sciences, College of Pharmacy, Biointerfaces Institute, University of Michigan, Ann Arbor, MI 48109, USA
| | - Shuibin Lin
- Center for Translational Medicine, Precision Medicine Institute, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou 510080, China
| | - Weisheng Guo
- Translational Medicine Center, The Second Affiliated Hospital, Guangzhou Medical University, Guangzhou 510260, China
| | - Guizhi Zhu
- Department of Pharmaceutical Sciences, College of Pharmacy, Biointerfaces Institute, University of Michigan, Ann Arbor, MI 48109, USA
| |
Collapse
|
46
|
Xu X, Zhou S, Tao Y, Zhong Z, Shao Y, Yi Y. Development and validation of a two glycolysis-related LncRNAs prognostic signature for glioma and in vitro analyses. Cell Div 2023; 18:10. [PMID: 37355624 DOI: 10.1186/s13008-023-00092-9] [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: 02/28/2023] [Accepted: 06/11/2023] [Indexed: 06/26/2023] Open
Abstract
BACKGROUND Mounting evidence suggests that there is a complex regulatory relationship between long non-coding RNAs (lncRNAs) and the glycolytic process during glioma development. This study aimed to investigate the prognostic role of glycolysis-related lncRNAs in glioma and their impact on the tumor microenvironment. METHODS This study utilized glioma transcriptome data from public databases to construct, evaluate, and validate a prognostic signature based on differentially expressed (DE)-glycolysis-associated lncRNAs through consensus clustering, DE-lncRNA analysis, Cox regression analysis, and receiver operating characteristic (ROC) curves. The clusterProfiler package was applied to reveal the potential functions of the risk score-related differentially expressed genes (DEGs). ESTIMATE and Gene Set Enrichment Analysis (GSEA) were utilized to evaluate the relationship between prognostic signature and the immune landscape of gliomas. Furthermore, the sensitivity of patients to immune checkpoint inhibitor (ICI) treatment based on the prognostic feature was predicted with the assistance of the Tumor Immune Dysfunction and Exclusion (TIDE) algorithm. Finally, qRT-PCR was used to verify the difference in the expression of the lncRNAs in glioma cells and normal cell. RESULTS By consensus clustering based on glycolytic gene expression profiles, glioma patients were divided into two clusters with significantly different overall survival (OS), from which 2 DE-lncRNAs, AL390755.1 and FLJ16779, were obtained. Subsequently, Cox regression analysis demonstrated that all of these lncRNAs were associated with OS in glioma patients and constructed a prognostic signature with a robust prognostic predictive efficacy. Functional enrichment analysis revealed that DEGs associated with risk scores were involved in immune responses, neurons, neurotransmitters, synapses and other terms. Immune landscape analysis suggested an extreme enrichment of immune cells in the high-risk group. Moreover, patients in the low-risk group were likely to benefit more from ICI treatment. qRT-PCR results showed that the expression of AL390755.1 and FLJ16779 was significantly different in glioma and normal cells. CONCLUSION We constructed a novel prognostic signature for glioma patients based on glycolysis-related lncRNAs. Besides, this project had provided a theoretical basis for the exploration of new ICI therapeutic targets for glioma patients.
Collapse
Affiliation(s)
- Xiaoping Xu
- Department of Neurosurgery, The Second People's Hospital of Yibin, Yibin, 644000, Sichuan Province, China.
| | - Shijun Zhou
- Department of Neurosurgery, The Second People's Hospital of Yibin, Yibin, 644000, Sichuan Province, China
| | - Yuchuan Tao
- Department of Neurosurgery, The Second People's Hospital of Yibin, Yibin, 644000, Sichuan Province, China
| | - Zhenglan Zhong
- Department of Health Examination, The Second People's Hospital of Yibin, Yibin, 644000, Sichuan Province, China
| | - Yongxiang Shao
- Department of Neurosurgery, The Second People's Hospital of Yibin, Yibin, 644000, Sichuan Province, China
| | - Yong Yi
- Department of Neurosurgery, The Second People's Hospital of Yibin, Yibin, 644000, Sichuan Province, China
| |
Collapse
|
47
|
Brisson L, Henrique Geraldo L, Bikfalvi A, Mathivet T. The strange Microenvironment of Glioblastoma. Rev Neurol (Paris) 2023; 179:490-501. [PMID: 36964121 PMCID: PMC11195635 DOI: 10.1016/j.neurol.2023.03.007] [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: 01/27/2023] [Revised: 02/24/2023] [Accepted: 03/01/2023] [Indexed: 03/26/2023]
Abstract
Glioblastoma (GB) is the most common and aggressive primary brain tumor, with poor patient survival and lack of effective therapies. Late advances trying to decipher the composition of the GB tumor microenvironment (TME) emphasized its role in tumor progression and potentialized it as a therapeutic target. Many components participate critically to tumor development and expansion such as blood vessels, immune cells or components of the nervous system. Dysmorphic tumor vasculature brings challenges to optimal delivery of cytotoxic agents currently used in clinics. Also, massive infiltration of immunosuppressive myeloid cells and limited recruitment of T cells limits the success of conventional immunotherapies. Neuronal input seems also be required for tumor expansion. In this review, we provide a comprehensive report of vascular and immune component of the GB TME and their cross talk during GB progression.
Collapse
Affiliation(s)
- L Brisson
- BRIC Inserm U1312, Université de Bordeaux, 33615 Pessac, France
| | - L Henrique Geraldo
- Cardiovascular Research Center, Department of Internal Medicine, Yale University School of Medicine, New Haven, CT, USA
| | - A Bikfalvi
- BRIC Inserm U1312, Université de Bordeaux, 33615 Pessac, France.
| | - T Mathivet
- BRIC Inserm U1312, Université de Bordeaux, 33615 Pessac, France
| |
Collapse
|
48
|
Kopper TJ, Yu X, Graner MW. Immunopathology of Extracellular Vesicles in Macrophage and Glioma Cross-Talk. J Clin Med 2023; 12:3430. [PMID: 37240536 PMCID: PMC10219523 DOI: 10.3390/jcm12103430] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2023] [Revised: 04/25/2023] [Accepted: 05/11/2023] [Indexed: 05/28/2023] Open
Abstract
Glioblastomas (GBM) are a devastating disease with extremely poor clinical outcomes. Resident (microglia) and infiltrating macrophages are a substantial component of the tumor environment. In GBM and other cancers, tumor-derived extracellular vesicles (EVs) suppress macrophage inflammatory responses, impairing their ability to identify and phagocytose cancerous tissues. Furthermore, these macrophages then begin to produce EVs that support tumor growth and migration. This cross-talk between macrophages/microglia and gliomas is a significant contributor to GBM pathophysiology. Here, we review the mechanisms through which GBM-derived EVs impair macrophage function, how subsequent macrophage-derived EVs support tumor growth, and the current therapeutic approaches to target GBM/macrophage EV crosstalk.
Collapse
Affiliation(s)
| | | | - Michael W. Graner
- Department of Neurosurgery, University of Colorado Anschutz Medical Campus, 12700 E 19th Ave., Aurora, CO 80045, USA; (T.J.K.); (X.Y.)
| |
Collapse
|
49
|
Dain L, Zhu G. Nucleic acid immunotherapeutics and vaccines: A promising approach to glioblastoma multiforme treatment. Int J Pharm 2023; 638:122924. [PMID: 37037396 PMCID: PMC10194422 DOI: 10.1016/j.ijpharm.2023.122924] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2022] [Revised: 03/28/2023] [Accepted: 03/31/2023] [Indexed: 04/12/2023]
Abstract
Glioblastoma multiforme (GBM) is a deadly and difficult to treat primary brain tumor for which satisfactory therapeutics have yet to be discovered. While cancer immunotherapeutics, such as immune checkpoint inhibitors, have successfully improved the treatment of some other types of cancer, the poorly immunogenic GBM tumor cells and the immunosuppressive GBM tumor microenvironment have made it difficult to develop GBM immunotherapeutics. Nucleic acids therapeutics and vaccines, particularly those of mRNA, have become a popular field of research in recent years. This review presents the progress of nucleic acid therapeutics and vaccines for GBM and briefly covers some representative delivery methods of nucleic acids to the central nervous system (CNS) for GBM therapy.
Collapse
Affiliation(s)
- Lauren Dain
- Department of Pharmaceutics and Center for Pharmaceutical Engineering and Sciences, Institute for Structural Biology and Drug Discovery, School of Pharmacy; The Developmental Therapeutics Program, Massey Cancer Center, Virginia Commonwealth University, Richmond, VA 23298, USA
| | - Guizhi Zhu
- Department of Pharmaceutics and Center for Pharmaceutical Engineering and Sciences, Institute for Structural Biology and Drug Discovery, School of Pharmacy; The Developmental Therapeutics Program, Massey Cancer Center, Virginia Commonwealth University, Richmond, VA 23298, USA; Department of Pharmaceutical Sciences, College of Pharmacy, University of Michigan, Ann Arbor, MI 48109, USA.
| |
Collapse
|
50
|
Montemurro N, Pahwa B, Tayal A, Shukla A, De Jesus Encarnacion M, Ramirez I, Nurmukhametov R, Chavda V, De Carlo A. Macrophages in Recurrent Glioblastoma as a Prognostic Factor in the Synergistic System of the Tumor Microenvironment. Neurol Int 2023; 15:595-608. [PMID: 37218976 PMCID: PMC10204554 DOI: 10.3390/neurolint15020037] [Citation(s) in RCA: 32] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2023] [Revised: 04/05/2023] [Accepted: 04/21/2023] [Indexed: 05/24/2023] Open
Abstract
Glioblastoma (GBM) is a common and highly malignant primary tumor of the central nervous system in adults. Ever more recent papers are focusing on understanding the role of the tumor microenvironment (TME) in affecting tumorigenesis and the subsequent prognosis. We assessed the impact of macrophages in the TME on the prognosis in patients with recurrent GBM. A PubMed, MEDLINE and Scopus review was conducted to identify all studies dealing with macrophages in the GBM microenvironment from January 2016 to December 2022. Glioma-associated macrophages (GAMs) act critically in enhancing tumor progression and can alter drug resistance, promoting resistance to radiotherapy and establishing an immunosuppressive environment. M1 macrophages are characterized by increased secretion of proinflammatory cytokines, such as IL-1ß, tumor necrosis factor (TNF), IL-27, matrix metalloproteinase (MMPs), CCL2, and VEGF (vascular endothelial growth factor), IGF1, that can lead to the destruction of the tissue. In contrast, M2 is supposed to participate in immunosuppression and tumor progression, which is formed after being exposed to the macrophage M-CSF, IL-10, IL-35 and the transforming growth factor-ß (TGF-β). Because there is currently no standard of care in recurrent GBM, novel identified targeted therapies based on the complex signaling and interactions between the glioma stem cells (GSCs) and the TME, especially resident microglia and bone-marrow-derived macrophages, may be helpful in improving the overall survival of these patients in the near future.
Collapse
Affiliation(s)
- Nicola Montemurro
- Department of Neurosurgery, Azienda Ospedaliero Universitaria Pisana (AOUP), University of Pisa, 56100 Pisa, Italy
| | - Bhavya Pahwa
- University College of Medical Sciences and GTB Hospital, New Delhi 110095, India
| | - Anish Tayal
- University College of Medical Sciences and GTB Hospital, New Delhi 110095, India
| | - Anushruti Shukla
- University College of Medical Sciences and GTB Hospital, New Delhi 110095, India
| | | | - Issael Ramirez
- Royal Melbourne Hospital, Melbourne, VIC 3000, Australia
| | - Renat Nurmukhametov
- Department of Spinal Surgery, Central Clinical Hospital of the Russian Academy of Sciences, 121359 Moscow, Russia
| | - Vishal Chavda
- Department of Pathology, Stanford of School of Medicine, Stanford University Medical Centre, Palo Alto, CA 94305, USA
| | - Antonella De Carlo
- Department of Neurosurgery, Azienda Ospedaliero Universitaria Pisana (AOUP), University of Pisa, 56100 Pisa, Italy
| |
Collapse
|