1
|
Önner H, Özer H, Gezer B, Körez KM, Cebeci H, Eren OÖ, Köktekir E, Karabağlı H. Comparing the diagnostic performance of DSC-MRI and FAPI PET in differentiating tumor progression from treatment-related changes in IDH-Wildtype Glioblastoma: A pilot study. Eur J Radiol 2025; 187:112075. [PMID: 40188635 DOI: 10.1016/j.ejrad.2025.112075] [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: 11/29/2024] [Revised: 02/19/2025] [Accepted: 03/26/2025] [Indexed: 04/08/2025]
Abstract
OBJECTIVES This pilot study compared the diagnostic performances of DSC-MRI and FAPI PET in differentiating tumor progression (TP) and treatment-related change (TRC) in isocitrate dehydrogenase (IDH) wild-type glioblastoma during follow-up. METHODS IDH wild-type glioblastoma patients who underwent DSC-MRI and FAPI PET were analyzed retrospectively. TP and TRC lesions were confirmed through radiological and clinical follow-up, with a median follow-up period of 8 months (2-12 months). The differences in DSC-MRI (CBVmax, CBVmean, and rCBVmean) and FAPI PET (SUVmax, SUVmean, and TBR SUVmean) parameters between TP and TRC were compared. ROC curve analyses were performed to assess the diagnostic performance. DeLong's test evaluated the differences in AUCs. RESULTS Twelve patients (6 men and 6 women, aged 33-70) with IDH wild-type glioblastoma were enrolled. Totally 18 lesions (8 TRC and 10 TP) were detected. All DSC-MRI and FAPI PET parameters were significantly higher in the TP than in the TRC. CBVmean showed the highest diagnostic performance among all parameters. However, the DeLong test revealed no significant difference in diagnostic performance between DSC-MRI and FAPI PET parameters. CONCLUSIONS Although the CBVmean has excellent diagnostic performance in differentiating TP from TRC, FAPI PET parameters were statistically found to have similar diagnostic performance. FAPI PET may be an alternative modality for patients with IDH wild-type glioblastoma who are unable to undergo DSC-MRI. However, further prospective large cohort studies and clinical validation are necessary.
Collapse
Affiliation(s)
- Hasan Önner
- Selcuk University, Faculty of Medicine, Department of Nuclear Medicine, Konya, Turkey.
| | - Halil Özer
- Selcuk University, Faculty of Medicine, Department of Radiology, Konya, Turkey
| | - Burak Gezer
- Selcuk University, Faculty of Medicine, Department of Neurosurgery, Konya, Turkey
| | - Kazım Muslu Körez
- Selcuk University, Faculty of Medicine, Department of Biostatistics, Konya, Turkey
| | - Hakan Cebeci
- Selcuk University, Faculty of Medicine, Department of Radiology, Konya, Turkey
| | - Orhan Önder Eren
- Selcuk University, Faculty of Medicine, Department of Medical Oncology, Konya, Turkey
| | - Ender Köktekir
- Selcuk University, Faculty of Medicine, Department of Neurosurgery, Konya, Turkey
| | - Hakan Karabağlı
- Selcuk University, Faculty of Medicine, Department of Neurosurgery, Konya, Turkey
| |
Collapse
|
2
|
Ismailov A, Spallone A, Belogurov A, Herbert A, Poptsova M. Molecular biology of the deadliest cancer - glioblastoma: what do we know? Front Immunol 2025; 16:1530305. [PMID: 40191211 PMCID: PMC11968700 DOI: 10.3389/fimmu.2025.1530305] [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: 11/18/2024] [Accepted: 03/07/2025] [Indexed: 04/09/2025] Open
Abstract
Glioblastomas are the most prevalent primary brain tumors and are associated with a dramatically poor prognosis. Despite an intensive treatment approach, including maximal surgical tumor removal followed by radio- and chemotherapy, the median survival for glioblastoma patients has remained around 18 months for decades. Glioblastoma is distinguished by its highly complex mechanisms of immune evasion and pronounced heterogeneity. This variability is apparent both within the tumor itself, which can exhibit multiple phenotypes simultaneously, and in its surrounding microenvironment. Another key feature of glioblastoma is its "cold" microenvironment, characterized by robust immunosuppression. Recent advances in single-cell RNA sequencing have uncovered new promising insights, revealing previously unrecognized aspects of this tumor. In this review, we consolidate current knowledge on glioblastoma cells and its microenvironment, with an emphasis on their biological properties and unique patterns of molecular communication through signaling pathways. The evidence underscores the critical need for personalized poly-immunotherapy and other approaches to overcome the plasticity of glioblastoma stem cells. Analyzing the tumor microenvironment of individual patients using single-cell transcriptomics and implementing a customized immunotherapeutic strategy could potentially improve survival outcomes for those facing this formidable disease.
Collapse
Affiliation(s)
- Aly Ismailov
- International Laboratory of Bioinformatics, Institute of Artificial Intelligence and Digital Sciences, Faculty of Computer Science, National Research University Higher School of Economics, Moscow, Russia
| | - Aldo Spallone
- International Laboratory of Bioinformatics, Institute of Artificial Intelligence and Digital Sciences, Faculty of Computer Science, National Research University Higher School of Economics, Moscow, Russia
- Laboratory of Hormonal Regulation Proteins, Shemyakin and Ovchinnikov Institute of Bioorganic Chemistry Russian Academy of Sciences (RAS), Moscow, Russia
| | - Alexey Belogurov
- Laboratory of Hormonal Regulation Proteins, Shemyakin and Ovchinnikov Institute of Bioorganic Chemistry Russian Academy of Sciences (RAS), Moscow, Russia
- Scientific and Educational Institute of Fundamental Medicine named after V.I. Pokrovsky, Department of Biological Chemistry, Russian University of Medicine, Moscow, Russia
| | - Alan Herbert
- International Laboratory of Bioinformatics, Institute of Artificial Intelligence and Digital Sciences, Faculty of Computer Science, National Research University Higher School of Economics, Moscow, Russia
- Discovery Department, InsideOutBio, Boston, MA, United States
| | - Maria Poptsova
- International Laboratory of Bioinformatics, Institute of Artificial Intelligence and Digital Sciences, Faculty of Computer Science, National Research University Higher School of Economics, Moscow, Russia
| |
Collapse
|
3
|
You H, Geng S, Li S, Imani M, Brambilla D, Sun T, Jiang C. Recent advances in biomimetic strategies for the immunotherapy of glioblastoma. Biomaterials 2024; 311:122694. [PMID: 38959533 DOI: 10.1016/j.biomaterials.2024.122694] [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: 04/08/2024] [Revised: 06/22/2024] [Accepted: 06/26/2024] [Indexed: 07/05/2024]
Abstract
Immunotherapy is regarded as one of the most promising approaches for treating tumors, with a multitude of immunotherapeutic thoughts currently under consideration for the lethal glioblastoma (GBM). However, issues with immunotherapeutic agents, such as limited in vivo stability, poor blood-brain barrier (BBB) penetration, insufficient GBM targeting, and represented monotherapy, have hindered the success of immunotherapeutic interventions. Moreover, even with the aid of conventional drug delivery systems, outcomes remain suboptimal. Biomimetic strategies seek to overcome these formidable drug delivery challenges by emulating nature's intelligent structures and functions. Leveraging the variety of biological structures and functions, biomimetic drug delivery systems afford a versatile platform with enhanced biocompatibility for the co-delivery of diverse immunotherapeutic agents. Moreover, their inherent capacity to traverse the BBB and home in on GBM holds promise for augmenting the efficacy of GBM immunotherapy. Thus, this review begins by revisiting the various thoughts and agents on immunotherapy for GBM. Then, the barriers to successful GBM immunotherapy are analyzed, and the corresponding biomimetic strategies are explored from the perspective of function and structure. Finally, the clinical translation's current state and prospects of biomimetic strategy are addressed. This review aspires to provide fresh perspectives on the advancement of immunotherapy for GBM.
Collapse
Affiliation(s)
- Haoyu You
- Key Laboratory of Smart Drug Delivery/Innovative Center for New Drug Development of Immune Inflammatory Diseases (Ministry of Education), Minhang Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Department of Pharmaceutics, School of Pharmacy, Fudan University, Shanghai 201203, China
| | - Shuo Geng
- Key Laboratory of Smart Drug Delivery/Innovative Center for New Drug Development of Immune Inflammatory Diseases (Ministry of Education), Minhang Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Department of Pharmaceutics, School of Pharmacy, Fudan University, Shanghai 201203, China
| | - Shangkuo Li
- Key Laboratory of Smart Drug Delivery/Innovative Center for New Drug Development of Immune Inflammatory Diseases (Ministry of Education), Minhang Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Department of Pharmaceutics, School of Pharmacy, Fudan University, Shanghai 201203, China
| | - Mohammad Imani
- Department of Science, Iran Polymer and Petrochemical Institute, Tehran 14977-13115, Iran; Center for Nanoscience and Nanotechnology, Institute for Convergence Science & Technology, Tehran 14588-89694, Iran
| | - Davide Brambilla
- Faculty of Pharmacy, University of Montreal, Montreal Quebec H3T 1J4, Canada
| | - Tao Sun
- Key Laboratory of Smart Drug Delivery/Innovative Center for New Drug Development of Immune Inflammatory Diseases (Ministry of Education), Minhang Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Department of Pharmaceutics, School of Pharmacy, Fudan University, Shanghai 201203, China.
| | - Chen Jiang
- Key Laboratory of Smart Drug Delivery/Innovative Center for New Drug Development of Immune Inflammatory Diseases (Ministry of Education), Minhang Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Department of Pharmaceutics, School of Pharmacy, Fudan University, Shanghai 201203, China
| |
Collapse
|
4
|
Zhang G, Tai P, Fang J, Wang Z, Yu R, Yin Z, Cao K. Multi-omics reveals the impact of cancer-associated fibroblasts on the prognosis and treatment response of adult diffuse highest-grade gliomas. Heliyon 2024; 10:e34526. [PMID: 39157370 PMCID: PMC11327523 DOI: 10.1016/j.heliyon.2024.e34526] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2024] [Revised: 07/09/2024] [Accepted: 07/10/2024] [Indexed: 08/20/2024] Open
Abstract
Background Cancer associated fibroblasts (CAF), an important cancer-promoting and immunosuppressive component of the tumor immune microenvironment (TIME), have recently been found to infiltrate adult diffuse highest-grade gliomas (ADHGG) (gliomas of grade IV). Methods Gene expression and clinical data of ADHGG patients were obtained from the CGGA and TCGA databases. Consensus clustering was used to identify CAF subtypes based on CAF key genes acquired from single-cell omics and spatial transcriptomomics. CIBERSORT, ssGSEA, MCPcounter, and ESTIMATE analyses were used to assess the TIME of GBM. Survival analysis, drug sensitivity analysis, TCIA database, TIDE and cMap algorithms were used to compare the prognosis and treatment response between patients with different CAF subtypes. An artificial neural network (ANN) model based on random forest was constructed to exactly identify CAF subtypes, which was validated in a real-world patient cohort of ADHGG. Results Consensus clustering classified ADHGG into two CAF subtypes. Compared with subtype B, patients with ADHGG subtype A had a poorer prognosis, worse responsiveness to immunotherapy and radiotherapy, higher CAF infiltration in TIME, but higher sensitivity to temozolomide. Furthermore, patients with subtype A had a much lower proportion of IDH mutations. Finally, the ANN model based on five genes (COL3A1, COL1A2, CD248, FN1, and COL1A1) could exactly discriminate CAF subtypes, and the validation of the real-world cohort indicated consistent results with the bioinformatics analyses. Conclusion This study revealed a novel CAF subtype to distinguish ADHGG patients with different prognosis and treatment responsiveness, which may be helpful for accurate clinical decision-making of ADHGG.
Collapse
Affiliation(s)
| | | | - Jianing Fang
- Department of Oncology, Third Xiangya Hospital, Central South University, Changsha, China
| | - Zhanwang Wang
- Department of Oncology, Third Xiangya Hospital, Central South University, Changsha, China
| | - Rui Yu
- Department of Oncology, Third Xiangya Hospital, Central South University, Changsha, China
| | - Zhijing Yin
- Department of Oncology, Third Xiangya Hospital, Central South University, Changsha, China
| | - Ke Cao
- Department of Oncology, Third Xiangya Hospital, Central South University, Changsha, China
| |
Collapse
|
5
|
Tang J, Chen Y, Wang C, Xia Y, Yu T, Tang M, Meng K, Yin L, Yang Y, Shen L, Xing H, Mao X. The role of mesenchymal stem cells in cancer and prospects for their use in cancer therapeutics. MedComm (Beijing) 2024; 5:e663. [PMID: 39070181 PMCID: PMC11283587 DOI: 10.1002/mco2.663] [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: 01/28/2024] [Revised: 06/26/2024] [Accepted: 07/01/2024] [Indexed: 07/30/2024] Open
Abstract
Mesenchymal stem cells (MSCs) are recruited by malignant tumor cells to the tumor microenvironment (TME) and play a crucial role in the initiation and progression of malignant tumors. This role encompasses immune evasion, promotion of angiogenesis, stimulation of cancer cell proliferation, correlation with cancer stem cells, multilineage differentiation within the TME, and development of treatment resistance. Simultaneously, extensive research is exploring the homing effect of MSCs and MSC-derived extracellular vesicles (MSCs-EVs) in tumors, aiming to design them as carriers for antitumor substances. These substances are targeted to deliver antitumor drugs to enhance drug efficacy while reducing drug toxicity. This paper provides a review of the supportive role of MSCs in tumor progression and the associated molecular mechanisms. Additionally, we summarize the latest therapeutic strategies involving engineered MSCs and MSCs-EVs in cancer treatment, including their utilization as carriers for gene therapeutic agents, chemotherapeutics, and oncolytic viruses. We also discuss the distribution and clearance of MSCs and MSCs-EVs upon entry into the body to elucidate the potential of targeted therapies based on MSCs and MSCs-EVs in cancer treatment, along with the challenges they face.
Collapse
Affiliation(s)
- Jian Tang
- Central LaboratoryXiangyang Central HospitalAffiliated Hospital of Hubei University of Arts and ScienceXiangyangChina
| | - Yu Chen
- Central LaboratoryXiangyang Central HospitalAffiliated Hospital of Hubei University of Arts and ScienceXiangyangChina
- Medical Affairs, Xiangyang Central HospitalAffiliated Hospital of Hubei University of Arts and ScienceXiangyangChina
| | - Chunhua Wang
- Department of Clinical LaboratoryXiangyang No. 1 People's HospitalHubei University of MedicineXiangyangHubei ProvinceChina
| | - Ying Xia
- Central LaboratoryXiangyang Central HospitalAffiliated Hospital of Hubei University of Arts and ScienceXiangyangChina
| | - Tingyu Yu
- Central LaboratoryXiangyang Central HospitalAffiliated Hospital of Hubei University of Arts and ScienceXiangyangChina
| | - Mengjun Tang
- Central LaboratoryXiangyang Central HospitalAffiliated Hospital of Hubei University of Arts and ScienceXiangyangChina
| | - Kun Meng
- Central LaboratoryXiangyang Central HospitalAffiliated Hospital of Hubei University of Arts and ScienceXiangyangChina
| | - Lijuan Yin
- State Key Laboratory of Food Nutrition and SafetyKey Laboratory of Industrial MicrobiologyMinistry of EducationTianjin Key Laboratory of Industry MicrobiologyNational and Local United Engineering Lab of Metabolic Control Fermentation TechnologyChina International Science and Technology Cooperation Base of Food Nutrition/Safety and Medicinal ChemistryCollege of BiotechnologyTianjin University of Science & TechnologyTianjinChina
| | - Yang Yang
- Shenzhen Key Laboratory of Pathogen and ImmunityNational Clinical Research Center for Infectious DiseaseState Key Discipline of Infectious DiseaseShenzhen Third People's HospitalSecond Hospital Affiliated to Southern University of Science and TechnologyShenzhenChina
| | - Liang Shen
- Central LaboratoryXiangyang Central HospitalAffiliated Hospital of Hubei University of Arts and ScienceXiangyangChina
| | - Hui Xing
- Central LaboratoryXiangyang Central HospitalAffiliated Hospital of Hubei University of Arts and ScienceXiangyangChina
- Department of Obstetrics and GynecologyXiangyang Central HospitalAffiliated Hospital of Hubei University of Arts and SciencesXiangyangChina
| | - Xiaogang Mao
- Central LaboratoryXiangyang Central HospitalAffiliated Hospital of Hubei University of Arts and ScienceXiangyangChina
- Department of Obstetrics and GynecologyXiangyang Central HospitalAffiliated Hospital of Hubei University of Arts and SciencesXiangyangChina
| |
Collapse
|
6
|
Sajjadi SF, Salehi N, Sadeghi M. Comprehensive integrated single-cell RNA sequencing analysis of brain metastasis and glioma microenvironment: Contrasting heterogeneity landscapes. PLoS One 2024; 19:e0306220. [PMID: 39058687 PMCID: PMC11280140 DOI: 10.1371/journal.pone.0306220] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2024] [Accepted: 06/12/2024] [Indexed: 07/28/2024] Open
Abstract
Understanding the specific type of brain malignancy, source of brain metastasis, and underlying transformation mechanisms can help provide better treatment and less harm to patients. The tumor microenvironment plays a fundamental role in cancer progression and affects both primary and metastatic cancers. The use of single-cell RNA sequencing to gain insights into the heterogeneity profiles in the microenvironment of brain malignancies is useful for guiding treatment decisions. To comprehensively investigate the heterogeneity in gliomas and brain metastasis originating from different sources (lung and breast), we integrated data from three groups of single-cell RNA-sequencing datasets obtained from GEO. We gathered and processed single-cell RNA sequencing data from 90,168 cells obtained from 17 patients. We then employed the R package Seurat for dataset integration. Next, we clustered the data within the UMAP space and acquired differentially expressed genes for cell categorization. Our results underscore the significance of macrophages as abundant and pivotal constituents of gliomas. In contrast, lung-to-brain metastases exhibit elevated numbers of AT2, cytotoxic CD4+ T, and exhausted CD8+ T cells. Conversely, breast-to-brain metastases are characterized by an abundance of epithelial and myCAF cells. Our study not only illuminates the variation in the TME between brain metastasis with different origins but also opens the door to utilizing established markers for these cell types to differentiate primary brain metastatic cancers.
Collapse
Affiliation(s)
- Seyedeh Fatemeh Sajjadi
- School of Biological Science, Institute for Research in Fundamental Sciences (IPM), Tehran, Iran
| | - Najmeh Salehi
- School of Biological Science, Institute for Research in Fundamental Sciences (IPM), Tehran, Iran
| | - Mehdi Sadeghi
- School of Biological Science, Institute for Research in Fundamental Sciences (IPM), Tehran, Iran
- National Institute of Genetic Engineering and Biotechnology (NIGEB), Tehran, Iran
| |
Collapse
|
7
|
Kang QM, Wang J, Chen SM, Song SR, Yu SC. Glioma-associated mesenchymal stem cells. Brain 2024; 147:755-765. [PMID: 37850820 DOI: 10.1093/brain/awad360] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2023] [Revised: 09/06/2023] [Accepted: 10/05/2023] [Indexed: 10/19/2023] Open
Abstract
Recent studies have revealed that glioma-associated mesenchymal stem cells play instrumental roles in tumorigenesis and tumour progression and cannot be ignored as a cellular component of the glioma microenvironment. Nevertheless, the origin of these cells and their roles are poorly understood. The only relevant studies have shown that glioma-associated mesenchymal stem cells play a large role in promoting tumour proliferation, invasion and angiogenesis. This review provides a comprehensive summary of their discovery and definition, origin, differences from other tissue-derived mesenchymal stem cells, spatial distribution, functions and prognostic and therapeutic opportunities to deepen the understanding of these cells and provide new insight into the treatment of glioma.
Collapse
Affiliation(s)
- Qing-Mei Kang
- Department of Stem Cell and Regenerative Medicine, Institute of Pathology and Southwest Cancer Center, Southwest Hospital, Chongqing, 400038, China
- Department of Pathology, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, 646000, China
- International Joint Research Center for Precision Biotherapy, Ministry of Science and Technology, Chongqing, 400038, China
- Key Laboratory of Cancer Immunopathology, Ministry of Education, Chongqing, 400038, China
- Jin-feng Laboratory, Chongqing, 401329, China
| | - Jun Wang
- Department of Stem Cell and Regenerative Medicine, Institute of Pathology and Southwest Cancer Center, Southwest Hospital, Chongqing, 400038, China
- International Joint Research Center for Precision Biotherapy, Ministry of Science and Technology, Chongqing, 400038, China
- Key Laboratory of Cancer Immunopathology, Ministry of Education, Chongqing, 400038, China
- Jin-feng Laboratory, Chongqing, 401329, China
| | - Shi-Man Chen
- Department of Stem Cell and Regenerative Medicine, Institute of Pathology and Southwest Cancer Center, Southwest Hospital, Chongqing, 400038, China
- International Joint Research Center for Precision Biotherapy, Ministry of Science and Technology, Chongqing, 400038, China
- Key Laboratory of Cancer Immunopathology, Ministry of Education, Chongqing, 400038, China
- Jin-feng Laboratory, Chongqing, 401329, China
| | - Si-Rong Song
- Department of Stem Cell and Regenerative Medicine, Institute of Pathology and Southwest Cancer Center, Southwest Hospital, Chongqing, 400038, China
- International Joint Research Center for Precision Biotherapy, Ministry of Science and Technology, Chongqing, 400038, China
- Key Laboratory of Cancer Immunopathology, Ministry of Education, Chongqing, 400038, China
- Jin-feng Laboratory, Chongqing, 401329, China
| | - Shi-Cang Yu
- Department of Stem Cell and Regenerative Medicine, Institute of Pathology and Southwest Cancer Center, Southwest Hospital, Chongqing, 400038, China
- International Joint Research Center for Precision Biotherapy, Ministry of Science and Technology, Chongqing, 400038, China
- Key Laboratory of Cancer Immunopathology, Ministry of Education, Chongqing, 400038, China
- Jin-feng Laboratory, Chongqing, 401329, China
| |
Collapse
|
8
|
Galbo PM, Madsen AT, Liu Y, Peng M, Wei Y, Ciesielski MJ, Fenstermaker RA, Graff S, Montagna C, Segall JE, Sidoli S, Zang X, Zheng D. Functional Contribution and Clinical Implication of Cancer-Associated Fibroblasts in Glioblastoma. Clin Cancer Res 2024; 30:865-876. [PMID: 38060213 PMCID: PMC10922678 DOI: 10.1158/1078-0432.ccr-23-0493] [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: 02/16/2023] [Revised: 05/11/2023] [Accepted: 12/05/2023] [Indexed: 12/08/2023]
Abstract
PURPOSE The abundance and biological contribution of cancer-associated fibroblasts (CAF) in glioblastoma (GBM) are poorly understood. Here, we aim to uncover its molecular signature, cellular roles, and potential tumorigenesis implications. EXPERIMENTAL DESIGN We first applied single-cell RNA sequencing (RNA-seq) and bioinformatics analysis to identify and characterize stromal cells with CAF transcriptomic features in human GBM tumors. Then, we performed functional enrichment analysis and in vitro assays to investigate their interactions with malignant GBM cells. RESULTS We found that CAF abundance was low but significantly correlated with tumor grade, poor clinical outcome, and activation of extracellular matrix remodeling using three large cohorts containing bulk RNA-seq data and clinical information. Proteomic analysis of a GBM-derived CAF line and its secretome revealed fibronectin (FN1) as a critical candidate factor mediating CAF functions. This was validated using in vitro cellular models, which demonstrated that CAF-conditioned media and recombinant FN1 could facilitate the migration and invasion of GBM cells. In addition, we showed that CAFs were more abundant in the mesenchymal-like state (or subtype) than in other states of GBMs. Interestingly, cell lines resembling the proneural state responded to the CAF signaling better for the migratory and invasive phenotypes. CONCLUSIONS Overall, this study characterized the molecular features and functional impacts of CAFs in GBM, alluding to novel cell interactions mediated by CAFs in the GBM microenvironment.
Collapse
Affiliation(s)
- Phillip M. Galbo
- Department of Genetics, Albert Einstein College of Medicine, Bronx, New York
- Department of Microbiology and Immunology, Albert Einstein College of Medicine, Bronx, New York
| | - Anne Tranberg Madsen
- Department of Microbiology and Immunology, Albert Einstein College of Medicine, Bronx, New York
| | - Yang Liu
- Department of Genetics, Albert Einstein College of Medicine, Bronx, New York
| | - Mou Peng
- Department of Microbiology and Immunology, Albert Einstein College of Medicine, Bronx, New York
| | - Yao Wei
- Department of Microbiology and Immunology, Albert Einstein College of Medicine, Bronx, New York
| | - Michael J Ciesielski
- Department of Neurosurgery, Roswell Park Comprehensive Cancer Center, Buffalo, NY
| | | | - Sarah Graff
- Department of Biochemistry, Albert Einstein College of Medicine, Bronx, New York
| | - Cristina Montagna
- Department of Radiation Oncology, Rutgers University, New Brunswick, New Jersey
| | - Jeffrey E. Segall
- Department of Pathology, Albert Einstein College of Medicine, Bronx, New York
| | - Simone Sidoli
- Department of Biochemistry, Albert Einstein College of Medicine, Bronx, New York
| | - Xingxing Zang
- Department of Microbiology and Immunology, Albert Einstein College of Medicine, Bronx, New York
- Department of Medicine, Albert Einstein College of Medicine, Bronx, New York
- Department of Urology, Albert Einstein College of Medicine, Bronx, New York
| | - Deyou Zheng
- Department of Genetics, Albert Einstein College of Medicine, Bronx, New York
- Departments of Neurology and Neuroscience, Albert Einstein College of Medicine, Bronx, New York
| |
Collapse
|
9
|
Lootens T, Roman BI, Stevens CV, De Wever O, Raedt R. Glioblastoma-Associated Mesenchymal Stem/Stromal Cells and Cancer-Associated Fibroblasts: Partners in Crime? Int J Mol Sci 2024; 25:2285. [PMID: 38396962 PMCID: PMC10889514 DOI: 10.3390/ijms25042285] [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: 12/22/2023] [Revised: 02/08/2024] [Accepted: 02/12/2024] [Indexed: 02/25/2024] Open
Abstract
Tumor-associated mesenchymal stem/stromal cells (TA-MSCs) have been recognized as attractive therapeutic targets in several cancer types, due to their ability to enhance tumor growth and angiogenesis and their contribution to an immunosuppressive tumor microenvironment (TME). In glioblastoma (GB), mesenchymal stem cells (MSCs) seem to be recruited to the tumor site, where they differentiate into glioblastoma-associated mesenchymal stem/stromal cells (GA-MSCs) under the influence of tumor cells and the TME. GA-MSCs are reported to exert important protumoral functions, such as promoting tumor growth and invasion, increasing angiogenesis, stimulating glioblastoma stem cell (GSC) proliferation and stemness, mediating resistance to therapy and contributing to an immunosuppressive TME. Moreover, they could act as precursor cells for cancer-associated fibroblasts (CAFs), which have recently been identified in GB. In this review, we provide an overview of the different functions exerted by GA-MSCs and CAFs and the current knowledge on the relationship between these cell types. Increasing our understanding of the interactions and signaling pathways in relevant models might contribute to future regimens targeting GA-MSCs and GB-associated CAFs to inhibit tumor growth and render the TME less immunosuppressive.
Collapse
Affiliation(s)
- Thibault Lootens
- 4Brain, Department of Head and Skin, Ghent University, 9000 Ghent, Belgium;
- Laboratory of Experimental Cancer Research, Department of Human Structure and Repair, Ghent University, 9000 Ghent, Belgium;
- Cancer Research Institute Ghent (CRIG), 9000 Ghent, Belgium; (B.I.R.); (C.V.S.)
| | - Bart I. Roman
- Cancer Research Institute Ghent (CRIG), 9000 Ghent, Belgium; (B.I.R.); (C.V.S.)
- SynBioC, Department of Green Chemistry and Technology, Faculty of Bioscience Engineering, Ghent University, 9000 Ghent, Belgium
| | - Christian V. Stevens
- Cancer Research Institute Ghent (CRIG), 9000 Ghent, Belgium; (B.I.R.); (C.V.S.)
- SynBioC, Department of Green Chemistry and Technology, Faculty of Bioscience Engineering, Ghent University, 9000 Ghent, Belgium
| | - Olivier De Wever
- Laboratory of Experimental Cancer Research, Department of Human Structure and Repair, Ghent University, 9000 Ghent, Belgium;
- Cancer Research Institute Ghent (CRIG), 9000 Ghent, Belgium; (B.I.R.); (C.V.S.)
| | - Robrecht Raedt
- 4Brain, Department of Head and Skin, Ghent University, 9000 Ghent, Belgium;
- Cancer Research Institute Ghent (CRIG), 9000 Ghent, Belgium; (B.I.R.); (C.V.S.)
| |
Collapse
|
10
|
Bahcheli AT, Min HK, Bayati M, Zhao H, Fortuna A, Dong W, Dzneladze I, Chan J, Chen X, Guevara-Hoyer K, Dirks PB, Huang X, Reimand J. Pan-cancer ion transport signature reveals functional regulators of glioblastoma aggression. EMBO J 2024; 43:196-224. [PMID: 38177502 PMCID: PMC10897389 DOI: 10.1038/s44318-023-00016-x] [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: 05/05/2023] [Revised: 11/30/2023] [Accepted: 11/30/2023] [Indexed: 01/06/2024] Open
Abstract
Ion channels, transporters, and other ion-flux controlling proteins, collectively comprising the "ion permeome", are common drug targets, however, their roles in cancer remain understudied. Our integrative pan-cancer transcriptome analysis shows that genes encoding the ion permeome are significantly more often highly expressed in specific subsets of cancer samples, compared to pan-transcriptome expectations. To enable target selection, we identified 410 survival-associated IP genes in 33 cancer types using a machine-learning approach. Notably, GJB2 and SCN9A show prominent expression in neoplastic cells and are associated with poor prognosis in glioblastoma, the most common and aggressive brain cancer. GJB2 or SCN9A knockdown in patient-derived glioblastoma cells induces transcriptome-wide changes involving neuron projection and proliferation pathways, impairs cell viability and tumor sphere formation in vitro, perturbs tunneling nanotube dynamics, and extends the survival of glioblastoma-bearing mice. Thus, aberrant activation of genes encoding ion transport proteins appears as a pan-cancer feature defining tumor heterogeneity, which can be exploited for mechanistic insights and therapy development.
Collapse
Affiliation(s)
- Alexander T Bahcheli
- Computational Biology Program, Ontario Institute for Cancer Research, Toronto, ON, Canada
- Department of Molecular Genetics, University of Toronto, Toronto, ON, Canada
| | - Hyun-Kee Min
- Department of Molecular Genetics, University of Toronto, Toronto, ON, Canada
- Developmental and Stem Cell Biology Program, The Hospital for Sick Children, Toronto, ON, Canada
- Arthur and Sonia Labatt Brain Tumour Research Centre, The Hospital for Sick Children, Toronto, ON, Canada
| | - Masroor Bayati
- Computational Biology Program, Ontario Institute for Cancer Research, Toronto, ON, Canada
- Department of Medical Biophysics, University of Toronto, Toronto, ON, Canada
| | - Hongyu Zhao
- Developmental and Stem Cell Biology Program, The Hospital for Sick Children, Toronto, ON, Canada
- Arthur and Sonia Labatt Brain Tumour Research Centre, The Hospital for Sick Children, Toronto, ON, Canada
- Department of Neurosurgery and Hunan International Scientific and Technological Cooperation Base of Brain Tumor Research, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Alexander Fortuna
- Computational Biology Program, Ontario Institute for Cancer Research, Toronto, ON, Canada
| | - Weifan Dong
- Department of Molecular Genetics, University of Toronto, Toronto, ON, Canada
- Developmental and Stem Cell Biology Program, The Hospital for Sick Children, Toronto, ON, Canada
- Arthur and Sonia Labatt Brain Tumour Research Centre, The Hospital for Sick Children, Toronto, ON, Canada
| | - Irakli Dzneladze
- Computational Biology Program, Ontario Institute for Cancer Research, Toronto, ON, Canada
| | - Jade Chan
- Department of Molecular Genetics, University of Toronto, Toronto, ON, Canada
- Developmental and Stem Cell Biology Program, The Hospital for Sick Children, Toronto, ON, Canada
- Arthur and Sonia Labatt Brain Tumour Research Centre, The Hospital for Sick Children, Toronto, ON, Canada
| | - Xin Chen
- Developmental and Stem Cell Biology Program, The Hospital for Sick Children, Toronto, ON, Canada
- Arthur and Sonia Labatt Brain Tumour Research Centre, The Hospital for Sick Children, Toronto, ON, Canada
- Songjiang Research Institute, Songjiang Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Kissy Guevara-Hoyer
- Computational Biology Program, Ontario Institute for Cancer Research, Toronto, ON, Canada
- Cancer Immunomonitoring and Immuno-Mediated Pathologies Support Unit, Department of Clinical Immunology, Institute of Laboratory Medicine (IML) and Biomedical Research Foundation (IdiSCC), San Carlos Clinical Hospital, Madrid, Spain
| | - Peter B Dirks
- Department of Molecular Genetics, University of Toronto, Toronto, ON, Canada
- Developmental and Stem Cell Biology Program, The Hospital for Sick Children, Toronto, ON, Canada
- Arthur and Sonia Labatt Brain Tumour Research Centre, The Hospital for Sick Children, Toronto, ON, Canada
| | - Xi Huang
- Department of Molecular Genetics, University of Toronto, Toronto, ON, Canada.
- Developmental and Stem Cell Biology Program, The Hospital for Sick Children, Toronto, ON, Canada.
- Arthur and Sonia Labatt Brain Tumour Research Centre, The Hospital for Sick Children, Toronto, ON, Canada.
| | - Jüri Reimand
- Computational Biology Program, Ontario Institute for Cancer Research, Toronto, ON, Canada.
- Department of Molecular Genetics, University of Toronto, Toronto, ON, Canada.
- Department of Medical Biophysics, University of Toronto, Toronto, ON, Canada.
| |
Collapse
|
11
|
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
|
12
|
Blanchard R, Adjei I. Engineering the glioblastoma microenvironment with bioactive nanoparticles for effective immunotherapy. RSC Adv 2023; 13:31411-31425. [PMID: 37901257 PMCID: PMC10603567 DOI: 10.1039/d3ra01153d] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2023] [Accepted: 09/27/2023] [Indexed: 10/31/2023] Open
Abstract
While immunotherapies have revolutionized treatment for other cancers, glioblastoma multiforme (GBM) patients have not shown similar positive responses. The limited response to immunotherapies is partly due to the unique challenges associated with the GBM tumor microenvironment (TME), which promotes resistance to immunotherapies, causing many promising therapies to fail. There is, therefore, an urgent need to develop strategies that make the TME immune permissive to promote treatment efficacy. Bioactive nano-delivery systems, in which the nanoparticle, due to its chemical composition, provides the pharmacological function, have recently emerged as an encouraging option for enhancing the efficacy of immunotherapeutics. These systems are designed to overcome immunosuppressive mechanisms in the TME to improve the efficacy of a therapy. This review will discuss different aspects of the TME and how they impede therapy success. Then, we will summarize recent developments in TME-modifying nanotherapeutics and the in vitro models utilized to facilitate these advances.
Collapse
Affiliation(s)
- Ryan Blanchard
- Department of Biomedical Engineering, Texas A&M University TX USA
| | - Isaac Adjei
- Department of Biomedical Engineering, Texas A&M University TX USA
| |
Collapse
|
13
|
Agrawal K, Asthana S, Kumar D. Role of Oxidative Stress in Metabolic Reprogramming of Brain Cancer. Cancers (Basel) 2023; 15:4920. [PMID: 37894287 PMCID: PMC10605619 DOI: 10.3390/cancers15204920] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2023] [Revised: 09/29/2023] [Accepted: 10/04/2023] [Indexed: 10/29/2023] Open
Abstract
Brain cancer is known as one of the deadliest cancers globally. One of the causative factors is the imbalance between oxidative and antioxidant activities in the body, which is referred to as oxidative stress (OS). As part of regular metabolism, oxygen is reduced by electrons, resulting in the creation of numerous reactive oxygen species (ROS). Inflammation is intricately associated with the generation of OS, leading to the increased production and accumulation of reactive oxygen and nitrogen species (RONS). Glioma stands out as one of the most common malignant tumors affecting the central nervous system (CNS), characterized by changes in the redox balance. Brain cancer cells exhibit inherent resistance to most conventional treatments, primarily due to the distinctive tumor microenvironment. Oxidative stress (OS) plays a crucial role in the development of various brain-related malignancies, such as glioblastoma multiforme (GBM) and medulloblastoma, where OS significantly disrupts the normal homeostasis of the brain. In this review, we provide in-depth descriptions of prospective targets and therapeutics, along with an assessment of OS and its impact on brain cancer metabolism. We also discuss targeted therapies.
Collapse
Affiliation(s)
- Kirti Agrawal
- School of Health Sciences and Technology (SoHST), UPES, Dehradun 248007, India
| | - Shailendra Asthana
- Translational Health Science and Technology Institute (THSTI), Faridabad 121001, India
| | - Dhruv Kumar
- School of Health Sciences and Technology (SoHST), UPES, Dehradun 248007, India
| |
Collapse
|
14
|
Ah-Pine F, Khettab M, Bedoui Y, Slama Y, Daniel M, Doray B, Gasque P. On the origin and development of glioblastoma: multifaceted role of perivascular mesenchymal stromal cells. Acta Neuropathol Commun 2023; 11:104. [PMID: 37355636 PMCID: PMC10290416 DOI: 10.1186/s40478-023-01605-x] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2023] [Accepted: 06/18/2023] [Indexed: 06/26/2023] Open
Abstract
Glioblastoma, IDH wild-type is the most common and aggressive form of glial tumors. The exact mechanisms of glioblastoma oncogenesis, including the identification of the glioma-initiating cell, are yet to be discovered. Recent studies have led to the hypothesis that glioblastoma arises from neural stem cells and glial precursor cells and that cell lineage constitutes a key determinant of the glioblastoma molecular subtype. These findings brought significant advancement to the comprehension of gliomagenesis. However, the cellular origin of glioblastoma with mesenchymal molecular features remains elusive. Mesenchymal stromal cells emerge as potential glioblastoma-initiating cells, especially with regard to the mesenchymal molecular subtype. These fibroblast-like cells, which derive from the neural crest and reside in the perivascular niche, may underlie gliomagenesis and exert pro-tumoral effects within the tumor microenvironment. This review synthesizes the potential roles of mesenchymal stromal cells in the context of glioblastoma and provides novel research avenues to better understand this lethal disease.
Collapse
Affiliation(s)
- F. Ah-Pine
- Unité de Recherche en Pharmaco-Immunologie (UR-EPI), Université et CHU de La Réunion, 97400 Saint-Denis, France
- Service d’Anatomie et Cytologie Pathologiques, CHU de La Réunion sites SUD – Saint-Pierre, BP 350, 97448 Saint-Pierre Cedex, France
| | - M. Khettab
- Unité de Recherche en Pharmaco-Immunologie (UR-EPI), Université et CHU de La Réunion, 97400 Saint-Denis, France
- Service d’Oncologie Médicale, CHU de La Réunion sites SUD – Saint-Pierre, BP 350, 97448 Saint-Pierre Cedex, France
| | - Y. Bedoui
- Unité de Recherche en Pharmaco-Immunologie (UR-EPI), Université et CHU de La Réunion, 97400 Saint-Denis, France
- Service d’Anatomie et Cytologie Pathologiques, CHU de La Réunion sites SUD – Saint-Pierre, BP 350, 97448 Saint-Pierre Cedex, France
| | - Y. Slama
- Unité de Recherche en Pharmaco-Immunologie (UR-EPI), Université et CHU de La Réunion, 97400 Saint-Denis, France
| | - M. Daniel
- Unité de Recherche en Pharmaco-Immunologie (UR-EPI), Université et CHU de La Réunion, 97400 Saint-Denis, France
- Service de Médecine d’Urgences-SAMU-SMUR, CHU de La Réunion - Site Félix Guyon, Allée Des Topazes CS 11 021, 97400 Saint-Denis, France
| | - B. Doray
- Unité de Recherche en Pharmaco-Immunologie (UR-EPI), Université et CHU de La Réunion, 97400 Saint-Denis, France
- Service de Génétique, CHU de La Réunion - Site Félix Guyon, Allée Des Topazes CS 11 021, 97400 Saint-Denis, France
| | - P. Gasque
- Unité de Recherche en Pharmaco-Immunologie (UR-EPI), Université et CHU de La Réunion, 97400 Saint-Denis, France
| |
Collapse
|
15
|
Han YP, Lin HW, Li H. Cancer Stem Cells in Tumours of the Central Nervous System in Children: A Comprehensive Review. Cancers (Basel) 2023; 15:3154. [PMID: 37370764 DOI: 10.3390/cancers15123154] [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: 04/14/2023] [Revised: 05/30/2023] [Accepted: 06/09/2023] [Indexed: 06/29/2023] Open
Abstract
Cancer stem cells (CSCs) are a subgroup of cells found in various kinds of tumours with stem cell characteristics, such as self-renewal, induced differentiation, and tumourigenicity. The existence of CSCs is regarded as a major source of tumour recurrence, metastasis, and resistance to conventional chemotherapy and radiation treatment. Tumours of the central nervous system (CNS) are the most common solid tumours in children, which have many different types including highly malignant embryonal tumours and midline gliomas, and low-grade gliomas with favourable prognoses. Stem cells from the CNS tumours have been largely found and reported by researchers in the last decade and their roles in tumour biology have been deeply studied. However, the cross-talk of CSCs among different CNS tumour types and their clinical impacts have been rarely discussed. This article comprehensively reviews the achievements in research on CSCs in paediatric CNS tumours. Biological functions, diagnostic values, and therapeutic perspectives are reviewed in detail. Further investigations into CSCs are warranted to improve the clinical practice in treating children with CNS tumours.
Collapse
Affiliation(s)
- Yi-Peng Han
- Department of Neurosurgery, Children's Hospital of Fudan University, National Children's Medical Center, Shanghai 201102, China
| | - Hou-Wei Lin
- Department of Paediatric Urology, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200092, China
- Department of Paediatric Surgery, Jiaxing Women and Children Hospital Affiliated to Jiaxing University, Jiaxing 314001, China
| | - Hao Li
- Department of Neurosurgery, Children's Hospital of Fudan University, National Children's Medical Center, Shanghai 201102, China
| |
Collapse
|
16
|
Nakhle J, Khattar K, Özkan T, Boughlita A, Abba Moussa D, Darlix A, Lorcy F, Rigau V, Bauchet L, Gerbal-Chaloin S, Daujat-Chavanieu M, Bellvert F, Turchi L, Virolle T, Hugnot JP, Buisine N, Galloni M, Dardalhon V, Rodriguez AM, Vignais ML. Mitochondria Transfer from Mesenchymal Stem Cells Confers Chemoresistance to Glioblastoma Stem Cells through Metabolic Rewiring. CANCER RESEARCH COMMUNICATIONS 2023; 3:1041-1056. [PMID: 37377608 PMCID: PMC10266428 DOI: 10.1158/2767-9764.crc-23-0144] [Citation(s) in RCA: 25] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 04/02/2023] [Revised: 05/12/2023] [Accepted: 05/19/2023] [Indexed: 06/29/2023]
Abstract
Glioblastomas (GBM) are heterogeneous tumors with high metabolic plasticity. Their poor prognosis is linked to the presence of glioblastoma stem cells (GSC), which support resistance to therapy, notably to temozolomide (TMZ). Mesenchymal stem cells (MSC) recruitment to GBM contributes to GSC chemoresistance, by mechanisms still poorly understood. Here, we provide evidence that MSCs transfer mitochondria to GSCs through tunneling nanotubes, which enhances GSCs resistance to TMZ. More precisely, our metabolomics analyses reveal that MSC mitochondria induce GSCs metabolic reprograming, with a nutrient shift from glucose to glutamine, a rewiring of the tricarboxylic acid cycle from glutaminolysis to reductive carboxylation and increase in orotate turnover as well as in pyrimidine and purine synthesis. Metabolomics analysis of GBM patient tissues at relapse after TMZ treatment documents increased concentrations of AMP, CMP, GMP, and UMP nucleotides and thus corroborate our in vitro analyses. Finally, we provide a mechanism whereby mitochondrial transfer from MSCs to GSCs contributes to GBM resistance to TMZ therapy, by demonstrating that inhibition of orotate production by Brequinar (BRQ) restores TMZ sensitivity in GSCs with acquired mitochondria. Altogether, these results identify a mechanism for GBM resistance to TMZ and reveal a metabolic dependency of chemoresistant GBM following the acquisition of exogenous mitochondria, which opens therapeutic perspectives based on synthetic lethality between TMZ and BRQ. Significance Mitochondria acquired from MSCs enhance the chemoresistance of GBMs. The discovery that they also generate metabolic vulnerability in GSCs paves the way for novel therapeutic approaches.
Collapse
Affiliation(s)
- Jean Nakhle
- Institute of Functional Genomics, University of Montpellier, CNRS, INSERM, Montpellier, France
- Institute for Regenerative Medicine and Biotherapy, University of Montpellier, INSERM, CHU Montpellier, Montpellier, France
- Institute of Molecular Genetics of Montpellier, University of Montpellier, CNRS, Montpellier, France
- RESTORE Research Center, University of Toulouse, INSERM 1301, CNRS 5070, EFS, ENVT, Toulouse, France
| | - Khattar Khattar
- Institute of Functional Genomics, University of Montpellier, CNRS, INSERM, Montpellier, France
| | - Tülin Özkan
- Institute for Regenerative Medicine and Biotherapy, University of Montpellier, INSERM, CHU Montpellier, Montpellier, France
- Faculty of Medicine, Department of Medical Biology, University of Ankara, Ankara, Turkey
| | - Adel Boughlita
- Institute for Regenerative Medicine and Biotherapy, University of Montpellier, INSERM, CHU Montpellier, Montpellier, France
| | - Daouda Abba Moussa
- Institute for Regenerative Medicine and Biotherapy, University of Montpellier, INSERM, CHU Montpellier, Montpellier, France
| | - Amélie Darlix
- Institute of Functional Genomics, University of Montpellier, CNRS, INSERM, Montpellier, France
- Department of Medical Oncology, Institut Régional du Cancer de Montpellier (ICM), University of Montpellier, Montpellier, France
| | - Frédérique Lorcy
- Department of Pathology and Oncobiology, Hôpital Gui de Chauliac, Montpellier, France
- The Center of the Biological Resource Center of University Hospital Center of Montpellier (BRC), Montpellier, France
| | - Valérie Rigau
- Institute of Functional Genomics, University of Montpellier, CNRS, INSERM, Montpellier, France
- Department of Pathology and Oncobiology, Hôpital Gui de Chauliac, Montpellier, France
- The Center of the Biological Resource Center of University Hospital Center of Montpellier (BRC), Montpellier, France
| | - Luc Bauchet
- Institute of Functional Genomics, University of Montpellier, CNRS, INSERM, Montpellier, France
- Department of Neurosurgery, Hopital Gui de Chauliac, Montpellier, France
| | - Sabine Gerbal-Chaloin
- Institute for Regenerative Medicine and Biotherapy, University of Montpellier, INSERM, CHU Montpellier, Montpellier, France
| | - Martine Daujat-Chavanieu
- Institute for Regenerative Medicine and Biotherapy, University of Montpellier, INSERM, CHU Montpellier, Montpellier, France
| | - Floriant Bellvert
- Toulouse Biotechnology Institute, University of Toulouse, CNRS, INRA, INSA, Toulouse, France
- MetaboHUB-MetaToul, National Infrastructure of Metabolomics and Fluxomics, Toulouse, France
| | - Laurent Turchi
- Université Côte D'Azur, CNRS, INSERM, Institut de Biologie Valrose, Team INSERM, “Cancer Stem Cell Plasticity and Functional Intra-tumor Heterogeneity”, Nice, France
| | - Thierry Virolle
- Université Côte D'Azur, CNRS, INSERM, Institut de Biologie Valrose, Team INSERM, “Cancer Stem Cell Plasticity and Functional Intra-tumor Heterogeneity”, Nice, France
| | - Jean-Philippe Hugnot
- Institute of Functional Genomics, University of Montpellier, CNRS, INSERM, Montpellier, France
| | - Nicolas Buisine
- UMR7221 Physiologie Moléculaire et Adaptation, CNRS, Muséum National d'Histoire Naturelle, Paris, France
| | - Mireille Galloni
- Institute for Regenerative Medicine and Biotherapy, University of Montpellier, INSERM, CHU Montpellier, Montpellier, France
| | - Valérie Dardalhon
- Institute of Molecular Genetics of Montpellier, University of Montpellier, CNRS, Montpellier, France
| | - Anne-Marie Rodriguez
- Sorbonne Université, Institut de Biologie Paris-Seine (IBPS), CNRS UMR 8256, INSERM ERL U1164, Biological Adaptation and Ageing, Paris, France
| | - Marie-Luce Vignais
- Institute of Functional Genomics, University of Montpellier, CNRS, INSERM, Montpellier, France
- Institute for Regenerative Medicine and Biotherapy, University of Montpellier, INSERM, CHU Montpellier, Montpellier, France
| |
Collapse
|
17
|
Ma J, Dai L, Yu J, Cao H, Bao Y, Hu J, Zhou L, Yang J, Sofia A, Chen H, Wu F, Xie Z, Qian W, Zhan R. Tumor microenvironment targeting system for glioma treatment via fusion cell membrane coating nanotechnology. Biomaterials 2023; 295:122026. [PMID: 36731366 DOI: 10.1016/j.biomaterials.2023.122026] [Citation(s) in RCA: 26] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2021] [Revised: 12/31/2022] [Accepted: 01/24/2023] [Indexed: 01/27/2023]
Abstract
The tumor microenvironment (TME), comprising cancer cells and stroma, plays a significant role in determining clinical outcomes, which makes targeting cancer cells in the TME an important area of research. One way in which cancer cells in the TME can be specifically targeted is by coating drug-encapsulated nanoparticles (NPs) with homotypic cancer cell membranes. However, incomplete targeting is inevitable for biomimetic nanoformulations coated with only cancer cell membranes because of the inherent heterogeneity of the TME. After observing the structural connection between glioma-associated stromal cells (GASCs) and glioma cells from a clinic, we designed a novel drug delivery system that targets the TME by coating polylactic-co-glycolic acid (PLGA) NPs with GASC-glioma cell fusion cell (SG cell) membranes. The resulting SGNPs inherited membrane proteins from both the glioma membrane and GASC membrane, significantly enhancing the tumor targeting efficiency compared to nanoformulations coated with cancer cell membranes alone. We further demonstrated that encapsulation of temozolomide (TMZ) improved the therapeutic efficacy of TMZ in both heterotopic and orthotopic glioma mouse models. Owing to its significant efficacy, our TME-targeting nanoplatform has potential for clinical applications in the treatment of various cancers.
Collapse
Affiliation(s)
- Junning Ma
- Department of Neurosurgery of First Affiliated Hospital, Zhejiang University School of Medicine, China; School of Medicine Zhejiang University, China.
| | - Lisi Dai
- Department of Pathology& Pathophysiology, and Department of Surgical Oncology of Second Affiliated Hospital, Zhejiang University School of Medicine, China; School of Basic Medical Sciences Zhejiang University, China.
| | - Jianbo Yu
- Department of Neurosurgery of First Affiliated Hospital, Zhejiang University School of Medicine, China
| | - Hui Cao
- Department of Neurosurgery of First Affiliated Hospital, Zhejiang University School of Medicine, China
| | - Youmei Bao
- Department of Neurosurgery, School of Medicine, Yale University, USA
| | - JiaJia Hu
- Department of Nuclear Medicine, Shanghai Ruijin Hospital, School of Medicine, Shanghai Jiaotong University, China
| | - Lihui Zhou
- Department of Neurosurgery of First Affiliated Hospital, Zhejiang University School of Medicine, China
| | - Jiqi Yang
- Department of Neurosurgery of First Affiliated Hospital, Zhejiang University School of Medicine, China
| | - Adame Sofia
- School of Medicine Zhejiang University, China
| | - Hongwei Chen
- Department of Neurosurgery of First Affiliated Hospital, Zhejiang University School of Medicine, China
| | - Fan Wu
- Department of Neurosurgery of First Affiliated Hospital, Zhejiang University School of Medicine, China
| | - Zhikai Xie
- Department of Neurosurgery of First Affiliated Hospital, Zhejiang University School of Medicine, China
| | - Wenqi Qian
- Department of Neurosurgery of First Affiliated Hospital, Zhejiang University School of Medicine, China
| | - Renya Zhan
- Department of Neurosurgery of First Affiliated Hospital, Zhejiang University School of Medicine, China.
| |
Collapse
|
18
|
Jain S, Rick JW, Joshi RS, Beniwal A, Spatz J, Gill S, Chang ACC, Choudhary N, Nguyen AT, Sudhir S, Chalif EJ, Chen JS, Chandra A, Haddad AF, Wadhwa H, Shah SS, Choi S, Hayes JL, Wang L, Yagnik G, Costello JF, Diaz A, Heiland DH, Aghi MK. Single-cell RNA sequencing and spatial transcriptomics reveal cancer-associated fibroblasts in glioblastoma with protumoral effects. J Clin Invest 2023; 133:e147087. [PMID: 36856115 PMCID: PMC9974099 DOI: 10.1172/jci147087] [Citation(s) in RCA: 76] [Impact Index Per Article: 38.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2021] [Accepted: 01/12/2023] [Indexed: 03/02/2023] Open
Abstract
Cancer-associated fibroblasts (CAFs) were presumed absent in glioblastoma given the lack of brain fibroblasts. Serial trypsinization of glioblastoma specimens yielded cells with CAF morphology and single-cell transcriptomic profiles based on their lack of copy number variations (CNVs) and elevated individual cell CAF probability scores derived from the expression of 9 CAF markers and absence of 5 markers from non-CAF stromal cells sharing features with CAFs. Cells without CNVs and with high CAF probability scores were identified in single-cell RNA-Seq of 12 patient glioblastomas. Pseudotime reconstruction revealed that immature CAFs evolved into subtypes, with mature CAFs expressing actin alpha 2, smooth muscle (ACTA2). Spatial transcriptomics from 16 patient glioblastomas confirmed CAF proximity to mesenchymal glioblastoma stem cells (GSCs), endothelial cells, and M2 macrophages. CAFs were chemotactically attracted to GSCs, and CAFs enriched GSCs. We created a resource of inferred crosstalk by mapping expression of receptors to their cognate ligands, identifying PDGF and TGF-β as mediators of GSC effects on CAFs and osteopontin and HGF as mediators of CAF-induced GSC enrichment. CAFs induced M2 macrophage polarization by producing the extra domain A (EDA) fibronectin variant that binds macrophage TLR4. Supplementing GSC-derived xenografts with CAFs enhanced in vivo tumor growth. These findings are among the first to identify glioblastoma CAFs and their GSC interactions, making them an intriguing target.
Collapse
Affiliation(s)
- Saket Jain
- Department of Neurosurgery, UCSF, San Francisco, California, USA
| | - Jonathan W. Rick
- Department of Neurosurgery, UCSF, San Francisco, California, USA
| | | | - Angad Beniwal
- Department of Neurosurgery, UCSF, San Francisco, California, USA
| | - Jordan Spatz
- Department of Neurosurgery, UCSF, San Francisco, California, USA
| | - Sabraj Gill
- Department of Neurosurgery, UCSF, San Francisco, California, USA
| | | | - Nikita Choudhary
- Department of Neurosurgery, UCSF, San Francisco, California, USA
| | - Alan T. Nguyen
- Department of Neurosurgery, UCSF, San Francisco, California, USA
| | - Sweta Sudhir
- Department of Neurosurgery, UCSF, San Francisco, California, USA
| | - Eric J. Chalif
- Department of Neurosurgery, UCSF, San Francisco, California, USA
| | - Jia-Shu Chen
- Department of Neurosurgery, UCSF, San Francisco, California, USA
| | - Ankush Chandra
- Department of Neurosurgery, UCSF, San Francisco, California, USA
| | | | - Harsh Wadhwa
- Department of Neurosurgery, UCSF, San Francisco, California, USA
| | - Sumedh S. Shah
- Department of Neurosurgery, UCSF, San Francisco, California, USA
| | - Serah Choi
- Department of Neurosurgery, UCSF, San Francisco, California, USA
| | - Josie L. Hayes
- Department of Neurosurgery, UCSF, San Francisco, California, USA
| | - Lin Wang
- Department of Neurosurgery, UCSF, San Francisco, California, USA
| | - Garima Yagnik
- Department of Neurosurgery, UCSF, San Francisco, California, USA
| | | | - Aaron Diaz
- Department of Neurosurgery, UCSF, San Francisco, California, USA
| | | | - Manish K. Aghi
- Department of Neurosurgery, UCSF, San Francisco, California, USA
| |
Collapse
|
19
|
Giambra M, Di Cristofori A, Valtorta S, Manfrellotti R, Bigiogera V, Basso G, Moresco RM, Giussani C, Bentivegna A. The peritumoral brain zone in glioblastoma: where we are and where we are going. J Neurosci Res 2023; 101:199-216. [PMID: 36300592 PMCID: PMC10091804 DOI: 10.1002/jnr.25134] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2022] [Revised: 09/01/2022] [Accepted: 10/01/2022] [Indexed: 12/13/2022]
Abstract
Glioblastoma (GBM) is the most aggressive and invasive primary brain tumor. Current therapies are not curative, and patients' outcomes remain poor with an overall survival of 20.9 months after surgery. The typical growing pattern of GBM develops by infiltrating the surrounding apparent normal brain tissue within which the recurrence is expected to appear in the majority of cases. Thus, in the last decades, an increased interest has developed to investigate the cellular and molecular interactions between GBM and the peritumoral brain zone (PBZ) bordering the tumor tissue. The aim of this review is to provide up-to-date knowledge about the oncogenic properties of the PBZ to highlight possible druggable targets for more effective treatment of GBM by limiting the formation of recurrence, which is almost inevitable in the majority of patients. Starting from the description of the cellular components, passing through the illustration of the molecular profiles, we finally focused on more clinical aspects, represented by imaging and radiological details. The complete picture that emerges from this review could provide new input for future investigations aimed at identifying new effective strategies to eradicate this still incurable tumor.
Collapse
Affiliation(s)
- Martina Giambra
- School of Medicine and Surgery, University of Milano-Bicocca, Monza, Italy.,PhD Program in Neuroscience, University of Milano-Bicocca, Monza, Italy
| | - Andrea Di Cristofori
- PhD Program in Neuroscience, University of Milano-Bicocca, Monza, Italy.,Division of Neurosurgery, Azienda Socio Sanitaria Territoriale - Monza, Ospedale San Gerardo, Monza, Italy
| | - Silvia Valtorta
- Department of Nuclear Medicine, San Raffaele Scientific Institute, IRCCS, Milan, Italy.,Institute of Molecular Bioimaging and Physiology, National Research Council (IBFM-CNR), Segrate, Italy.,NBFC, National Biodiversity Future Center, 90133, Palermo, Italy
| | - Roberto Manfrellotti
- School of Medicine and Surgery, University of Milano-Bicocca, Monza, Italy.,Division of Neurosurgery, Azienda Socio Sanitaria Territoriale - Monza, Ospedale San Gerardo, Monza, Italy
| | - Vittorio Bigiogera
- School of Medicine and Surgery, University of Milano-Bicocca, Monza, Italy
| | - Gianpaolo Basso
- School of Medicine and Surgery, University of Milano-Bicocca, Monza, Italy
| | - Rosa Maria Moresco
- School of Medicine and Surgery, University of Milano-Bicocca, Monza, Italy.,Department of Nuclear Medicine, San Raffaele Scientific Institute, IRCCS, Milan, Italy.,Institute of Molecular Bioimaging and Physiology, National Research Council (IBFM-CNR), Segrate, Italy
| | - Carlo Giussani
- School of Medicine and Surgery, University of Milano-Bicocca, Monza, Italy.,Division of Neurosurgery, Azienda Socio Sanitaria Territoriale - Monza, Ospedale San Gerardo, Monza, Italy
| | - Angela Bentivegna
- School of Medicine and Surgery, University of Milano-Bicocca, Monza, Italy
| |
Collapse
|
20
|
Long H, Zhang P, Bi Y, Yang C, Wu M, He D, Huang S, Yang K, Qi S, Wang J. MRI radiomic features of peritumoral edema may predict the recurrence sites of glioblastoma multiforme. Front Oncol 2023; 12:1042498. [PMID: 36686829 PMCID: PMC9845721 DOI: 10.3389/fonc.2022.1042498] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2022] [Accepted: 12/02/2022] [Indexed: 01/05/2023] Open
Abstract
Background and purpose As one of the most aggressive malignant tumor in the central nervous system, the main cause of poor outcome of glioblastoma (GBM) is recurrence, a non-invasive method which can predict the area of recurrence pre-operation is necessary.To investigate whether there is radiological heterogeneity within peritumoral edema and identify the reproducible radiomic features predictive of the sites of recurrence of glioblastoma(GBM), which may be of value to optimize patients' management. Materials and methods The clinical information and MR images (contrast-enhanced T1 weighted and FLAIR sequences) of 22 patients who have been histologically proven glioblastoma, were retrospectively evaluated. Kaplan-Meier methods was used for survival analysis. Oedematous regions were manually segmented by an expert into recurrence region, non-recurrence region. A set of 94 radiomic features were obtained from each region using the function of analyzing MR image of 3D slicer. Paired t test was performed to identify the features existing significant difference. Subsequently, the data of two patients from TCGA database was used to evaluate whether these features have clinical value. Results Ten features with significant differences between the recurrence and non-recurrence subregions were identified and verified on two individual patients from the TCGA database with pathologically confirmed diagnosis of GBM. Conclusions Our results suggested that heterogeneity does exist in peritumoral edema, indicating that the radiomic features of peritumoral edema from routine MR images can be utilized to predict the sites of GBM recurrence. Our findings may further guide the surgical treatment strategy for GBM.
Collapse
Affiliation(s)
- Hao Long
- Department of Neurosurgery, Nanfang Hospital, Southern Medical University, Guangzhou, China,The First Clinical Medicine College, Southern Medical University, Guangzhou, China
| | - Ping Zhang
- Department of oncology, Guangdong 999 Brain Hospital, Guangzhou, China
| | - Yuewei Bi
- The First Clinical Medicine College, Southern Medical University, Guangzhou, China,Neural Networks Surgery Team, Southern Medical University, Guangzhou, China
| | - Chen Yang
- The First Clinical Medicine College, Southern Medical University, Guangzhou, China,Neural Networks Surgery Team, Southern Medical University, Guangzhou, China
| | - Manfeng Wu
- The First Clinical Medicine College, Southern Medical University, Guangzhou, China,Neural Networks Surgery Team, Southern Medical University, Guangzhou, China
| | - Dian He
- The First Clinical Medicine College, Southern Medical University, Guangzhou, China,Neural Networks Surgery Team, Southern Medical University, Guangzhou, China
| | - Shaozhuo Huang
- The First Clinical Medicine College, Southern Medical University, Guangzhou, China,Neural Networks Surgery Team, Southern Medical University, Guangzhou, China
| | - Kaijun Yang
- Department of Neurosurgery, Nanfang Hospital, Southern Medical University, Guangzhou, China,The First Clinical Medicine College, Southern Medical University, Guangzhou, China
| | - Songtao Qi
- Department of Neurosurgery, Nanfang Hospital, Southern Medical University, Guangzhou, China,The First Clinical Medicine College, Southern Medical University, Guangzhou, China
| | - Jun Wang
- Department of Neurosurgery, Nanfang Hospital, Southern Medical University, Guangzhou, China,The First Clinical Medicine College, Southern Medical University, Guangzhou, China,Neural Networks Surgery Team, Southern Medical University, Guangzhou, China,*Correspondence: Jun Wang,
| |
Collapse
|
21
|
Yao Y, Tan X, Yin W, Kou Y, Wang X, Jiang X, Chen S, Liu Y, Dang J, Yin J, Cheng Z. Performance of 18 F-FAPI PET/CT in assessing glioblastoma before radiotherapy: a pilot study. BMC Med Imaging 2022; 22:226. [PMID: 36566187 PMCID: PMC9789562 DOI: 10.1186/s12880-022-00952-w] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2022] [Accepted: 12/14/2022] [Indexed: 12/25/2022] Open
Abstract
BACKGROUND We aimed to determine the performance of 18 F-FAPI PET/CT used for preprocedural assessment of glioblastoma before radiotherapy. METHODS Twelve glioblastoma patients having undergone incomplete surgical resection or biopsy were examined with 18 F-FAPI PET/CT and MRI scanning before radiotherapy. All patients had confirmed tumor residues according to findings of histopathological and/or long-term clinical and radiological follow-ups. Lesion characterization data, including SUVmax and tumor-to-background ratio (TBR) on PET/CT were attained. PET/CT and MRI findings were compared in terms of number of lesions. The correlation between immunohistochemistry, molecular expression, and PET/CT parameters was also evaluated. RESULTS 18 F-FAPI PET/CT detected 16 FAPI-avid out of 23 lesions in 12 patients described on MRI. MRI was statistically different from 18 F-FAPI PET/CT for lesion detection according to the exact McNemar statistical test (P = 0.0156). The SUVmax and TBR of the glioblastomas was 7.08 ± 3.55 and 19.95 ± 13.22, respectively. The sensitivity and positive predictive value (PPV) of 18 F-FAPI PET were 69.6% and 100%, respectively. Neither the Ki-67 index nor the molecular expression was correlated with the FAPI-PET/CT parameters. CONCLUSION 18 F-FAPI PET/CT detects glioblastomas at a lower rate than MRI. However, the 100% PPV of the examination may make it useful for differentiating controversial lesions detected on MRI. The 18 F-FAPI-avid lesions are displayed more clearly probably due to a higher TBR. 18 F-FAPI PET/CT imaging might find application in glioblastoma biopsy and radiotherapy planning.
Collapse
Affiliation(s)
- Yutang Yao
- grid.54549.390000 0004 0369 4060Department of Nuclear Medicine, Sichuan Cancer Hospital&Institute, Sichuan Cancer Center, University of Electronic Science and Technology of China, No.55, Section 4, South People’s Road, Sichuan 610041 Chengdu, China
| | - Xiaofei Tan
- grid.54549.390000 0004 0369 4060Department of Nuclear Medicine, Sichuan Cancer Hospital&Institute, Sichuan Cancer Center, University of Electronic Science and Technology of China, No.55, Section 4, South People’s Road, Sichuan 610041 Chengdu, China
| | - Wenya Yin
- grid.54549.390000 0004 0369 4060Department of Radiation Oncology, Sichuan Cancer Hospital&Institute, Sichuan Cancer Center, University of Electronic Science and Technology of China, No.55, Section 4, South People’s Road, 610041 Chengdu, China
| | - Ying Kou
- grid.54549.390000 0004 0369 4060Department of Nuclear Medicine, Sichuan Cancer Hospital&Institute, Sichuan Cancer Center, University of Electronic Science and Technology of China, No.55, Section 4, South People’s Road, Sichuan 610041 Chengdu, China
| | - Xiaoxiong Wang
- grid.54549.390000 0004 0369 4060Department of Nuclear Medicine, Sichuan Cancer Hospital&Institute, Sichuan Cancer Center, University of Electronic Science and Technology of China, No.55, Section 4, South People’s Road, Sichuan 610041 Chengdu, China
| | - Xiao Jiang
- grid.54549.390000 0004 0369 4060Department of Nuclear Medicine, Sichuan Cancer Hospital&Institute, Sichuan Cancer Center, University of Electronic Science and Technology of China, No.55, Section 4, South People’s Road, Sichuan 610041 Chengdu, China ,grid.410655.30000 0001 0157 8259Institute of Isotope, China Institute of Atomic Energy, 102413 Beijing, China
| | - Shirong Chen
- grid.54549.390000 0004 0369 4060Department of Nuclear Medicine, Sichuan Cancer Hospital&Institute, Sichuan Cancer Center, University of Electronic Science and Technology of China, No.55, Section 4, South People’s Road, Sichuan 610041 Chengdu, China
| | - Yongli Liu
- grid.54549.390000 0004 0369 4060Department of Nuclear Medicine, Sichuan Cancer Hospital&Institute, Sichuan Cancer Center, University of Electronic Science and Technology of China, No.55, Section 4, South People’s Road, Sichuan 610041 Chengdu, China
| | - Jun Dang
- grid.54549.390000 0004 0369 4060Department of Nuclear Medicine, Sichuan Cancer Hospital&Institute, Sichuan Cancer Center, University of Electronic Science and Technology of China, No.55, Section 4, South People’s Road, Sichuan 610041 Chengdu, China
| | - Jun Yin
- grid.54549.390000 0004 0369 4060Department of Radiation Oncology, Sichuan Cancer Hospital&Institute, Sichuan Cancer Center, University of Electronic Science and Technology of China, No.55, Section 4, South People’s Road, 610041 Chengdu, China
| | - Zhuzhong Cheng
- grid.54549.390000 0004 0369 4060Department of Nuclear Medicine, Sichuan Cancer Hospital&Institute, Sichuan Cancer Center, University of Electronic Science and Technology of China, No.55, Section 4, South People’s Road, Sichuan 610041 Chengdu, China
| |
Collapse
|
22
|
Jenner AL, Smalley M, Goldman D, Goins WF, Cobbs CS, Puchalski RB, Chiocca EA, Lawler S, Macklin P, Goldman A, Craig M. Agent-based computational modeling of glioblastoma predicts that stromal density is central to oncolytic virus efficacy. iScience 2022; 25:104395. [PMID: 35637733 PMCID: PMC9142563 DOI: 10.1016/j.isci.2022.104395] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2021] [Revised: 03/18/2022] [Accepted: 04/08/2022] [Indexed: 11/26/2022] Open
Abstract
Oncolytic viruses (OVs) are emerging cancer immunotherapy. Despite notable successes in the treatment of some tumors, OV therapy for central nervous system cancers has failed to show efficacy. We used an ex vivo tumor model developed from human glioblastoma tissue to evaluate the infiltration of herpes simplex OV rQNestin (oHSV-1) into glioblastoma tumors. We next leveraged our data to develop a computational, model of glioblastoma dynamics that accounts for cellular interactions within the tumor. Using our computational model, we found that low stromal density was highly predictive of oHSV-1 therapeutic success, suggesting that the efficacy of oHSV-1 in glioblastoma may be determined by stromal-to-tumor cell regional density. We validated these findings in heterogenous patient samples from brain metastatic adenocarcinoma. Our integrated modeling strategy can be applied to suggest mechanisms of therapeutic responses for central nervous system cancers and to facilitate the successful translation of OVs into the clinic.
Collapse
Affiliation(s)
- Adrianne L. Jenner
- Department of Mathematics and Statistics, Université de Montréal, Montréal, QC, Canada
- Sainte-Justine University Hospital Research Centre, Montréal, QC, Canada
| | - Munisha Smalley
- Division of Engineering in Medicine, Brigham and Women’s Hospital, Boston, MA, USA
| | | | - William F. Goins
- Department of Microbiology and Molecular Genetics, University of Pittsburgh, Pittsburgh, PA, USA
| | - Charles S. Cobbs
- Ben and Catherine Ivy Center for Advanced Brain Tumor Treatment, Swedish Neuroscience Institute, Seattle, WA, USA
| | - Ralph B. Puchalski
- Ben and Catherine Ivy Center for Advanced Brain Tumor Treatment, Swedish Neuroscience Institute, Seattle, WA, USA
| | - E. Antonio Chiocca
- Department of Neurosurgery, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, USA
| | - Sean Lawler
- Department of Neurosurgery, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, USA
| | - Paul Macklin
- Department of Intelligent Systems Engineering, Indiana University, Bloomington, IN, USA
| | - Aaron Goldman
- Division of Engineering in Medicine, Brigham and Women’s Hospital, Boston, MA, USA
- Department of Medicine, Harvard Medical School, Boston, MA, USA
| | - Morgan Craig
- Department of Mathematics and Statistics, Université de Montréal, Montréal, QC, Canada
- Sainte-Justine University Hospital Research Centre, Montréal, QC, Canada
| |
Collapse
|
23
|
Cai Y, Li K, Lin J, Liang X, Xu W, Zhan Z, Xue S, Zeng Y, Chai P, Mao Y, Song Z, Han L, Song Y, Zhang X, Wang H. Lighting a Fire: Gasdermin-Mediated Pyroptosis Remodels the Glioma Microenvironment and Promotes Immune Checkpoint Blockade Response. Front Immunol 2022; 13:910490. [PMID: 35784306 PMCID: PMC9249059 DOI: 10.3389/fimmu.2022.910490] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2022] [Accepted: 05/11/2022] [Indexed: 12/02/2022] Open
Abstract
Pyroptosis is a proinflammatory programmed cell death pathway mediated by gasdermins. Exploring the role of pyroptosis can provide new insights into tumor malignancy. The most recent studies on pyroptosis have focused on tumor cells. However, the effects of pyroptosis on the tumor microenvironment (TME), immunotherapeutic responses, and efficacy have been neglected, especially in case of glioma. In this study, four independent glioma cohorts comprising 1,339 samples and a pan-cancer cohort comprising 10,535 tumor samples were analyzed. The relationships among pyroptosis status, prognosis, microenvironment cellular components, and clinical and biological phenotypes were investigated through the identification of pyroptosis subtypes, construction of a gasdermin-related prognostic index (GPI), and evaluation of immunological characteristics in glioma. The Genomics of Drug Sensitivity in Cancer database and “pRRophetic” package in R were used to estimate temozolomide (TMZ) sensitivity. The “Submap” package and external immunotherapy cohorts were used to investigate and confirm the role of GPI in response to and efficacy of immunotherapy in glioma. Finally, potential small-molecule compounds related to GPI were identified using the connectivity map database and mode-of-action analysis. We identified three different pyroptosis subtypes: cluster 1 (C1) characterized by a higher GPI, while cluster 2 (C2) and cluster 3 (C3) characterized by a lower GPI. The high GPI of C1 was associated with glioma progression and worse prognoses, whereas the low GPI of subtype C2 and C3 was associated with better prognoses. However, patients with high GPIs were found to be more sensitive to TMZ and immune checkpoint blockade than those with low GPIs. Furthermore, gasdermin D may be a principal potential biomarker and play key roles in pyroptosis-inducible therapy combined with immunotherapy in glioma. This study provides a clinical, biological, and molecular landscape of pyroptosis and suggests that pyroptosis of glioma cells may perform the dual function of promoting both tumorigenesis and antitumor immunity.
Collapse
Affiliation(s)
- Yonghua Cai
- Department of Neurosurgery, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Ke Li
- Department of Neurosurgery, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Jie Lin
- Department of Neurosurgery, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Xianqiu Liang
- Department of Neurosurgery, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Wei Xu
- Department of Neurosurgery, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Zhengming Zhan
- Department of Neurosurgery, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Shuaishuai Xue
- Department of Neurosurgery, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Yu Zeng
- Department of Neurosurgery, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Peng Chai
- Department of Neurosurgery, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Yangqi Mao
- Department of Neurosurgery, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Zibin Song
- Department of Neurosurgery, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Lei Han
- Department of Neurosurgery, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Ye Song
- Department of Neurosurgery, Nanfang Hospital, Southern Medical University, Guangzhou, China
- Department of Neurosurgery, Ganzhou People’s Hospital, Ganzhou, China
- *Correspondence: Hai Wang, ; Xian Zhang, ; Ye Song,
| | - Xian Zhang
- Department of Neurosurgery, Nanfang Hospital, Southern Medical University, Guangzhou, China
- *Correspondence: Hai Wang, ; Xian Zhang, ; Ye Song,
| | - Hai Wang
- Department of Neurosurgery, Nanfang Hospital, Southern Medical University, Guangzhou, China
- *Correspondence: Hai Wang, ; Xian Zhang, ; Ye Song,
| |
Collapse
|
24
|
LeBlanc VG, Trinh DL, Aslanpour S, Hughes M, Livingstone D, Jin D, Ahn BY, Blough MD, Cairncross JG, Chan JA, Kelly JJP, Marra MA. Single-cell landscapes of primary glioblastomas and matched explants and cell lines show variable retention of inter- and intratumor heterogeneity. Cancer Cell 2022; 40:379-392.e9. [PMID: 35303420 DOI: 10.1016/j.ccell.2022.02.016] [Citation(s) in RCA: 80] [Impact Index Per Article: 26.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/26/2021] [Revised: 10/01/2021] [Accepted: 02/23/2022] [Indexed: 12/12/2022]
Abstract
Glioblastomas (GBMs) are aggressive brain tumors characterized by extensive inter- and intratumor heterogeneity. Patient-derived models, such as organoids and explants, have recently emerged as useful models to study such heterogeneity, although the extent to which they can recapitulate GBM genomic features remains unclear. Here, we analyze bulk exome and single-cell genome and transcriptome profiles of 12 IDH wild-type GBMs, including two recurrent tumors, and of patient-derived explants (PDEs) and gliomasphere (GS) lines derived from these tumors. We find that PDEs are genetically similar to, and variably retain gene expression characteristics of, their parent tumors. Notably, PDEs appear to exhibit similar levels of transcriptional heterogeneity compared with their parent tumors, whereas GS lines tend to be enriched for cells in a more uniform transcriptional state. The approaches and datasets introduced here will provide a valuable resource to help guide experiments using GBM-derived models, especially in the context of studying cellular heterogeneity.
Collapse
Affiliation(s)
- Véronique G LeBlanc
- Canada's Michael Smith Genome Sciences Centre, BC Cancer, Vancouver, V5Z 4S6 BC, Canada
| | - Diane L Trinh
- Canada's Michael Smith Genome Sciences Centre, BC Cancer, Vancouver, V5Z 4S6 BC, Canada
| | - Shaghayegh Aslanpour
- Department of Clinical Neurosciences, University of Calgary, Calgary, T2N 2T9 AB, Canada; Arnie Charbonneau Cancer Institute, University of Calgary, Calgary, T2N 4Z6 AB, Canada
| | - Martha Hughes
- Department of Clinical Neurosciences, University of Calgary, Calgary, T2N 2T9 AB, Canada; Arnie Charbonneau Cancer Institute, University of Calgary, Calgary, T2N 4Z6 AB, Canada
| | - Dorothea Livingstone
- Department of Clinical Neurosciences, University of Calgary, Calgary, T2N 2T9 AB, Canada; Arnie Charbonneau Cancer Institute, University of Calgary, Calgary, T2N 4Z6 AB, Canada
| | - Dan Jin
- Canada's Michael Smith Genome Sciences Centre, BC Cancer, Vancouver, V5Z 4S6 BC, Canada
| | - Bo Young Ahn
- Arnie Charbonneau Cancer Institute, University of Calgary, Calgary, T2N 4Z6 AB, Canada
| | - Michael D Blough
- Arnie Charbonneau Cancer Institute, University of Calgary, Calgary, T2N 4Z6 AB, Canada
| | - J Gregory Cairncross
- Department of Clinical Neurosciences, University of Calgary, Calgary, T2N 2T9 AB, Canada; Arnie Charbonneau Cancer Institute, University of Calgary, Calgary, T2N 4Z6 AB, Canada
| | - Jennifer A Chan
- Arnie Charbonneau Cancer Institute, University of Calgary, Calgary, T2N 4Z6 AB, Canada; Department of Pathology & Laboratory Medicine, University of Calgary, Calgary, T2L 2K8 AB, Canada
| | - John J P Kelly
- Department of Clinical Neurosciences, University of Calgary, Calgary, T2N 2T9 AB, Canada; Arnie Charbonneau Cancer Institute, University of Calgary, Calgary, T2N 4Z6 AB, Canada
| | - Marco A Marra
- Canada's Michael Smith Genome Sciences Centre, BC Cancer, Vancouver, V5Z 4S6 BC, Canada; Department of Medical Genetics, University of British Columbia, Vancouver, V6H 3N1 BC, Canada.
| |
Collapse
|
25
|
Li SY, Johnson R, Smyth LC, Dragunow M. Platelet-derived growth factor signalling in neurovascular function and disease. Int J Biochem Cell Biol 2022; 145:106187. [PMID: 35217189 DOI: 10.1016/j.biocel.2022.106187] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2021] [Revised: 02/08/2022] [Accepted: 02/21/2022] [Indexed: 11/25/2022]
Abstract
Platelet-derived growth factors are critical for cerebrovascular development and homeostasis. Abnormalities in this signalling pathway are implicated in neurological diseases, especially those where neurovascular dysfunction and neuroinflammation plays a prominent role in disease pathologies, such as stroke and Alzheimer's disease; the angiogenic nature of this pathway also draws its significance in brain malignancies such as glioblastoma where tumour angiogenesis is profuse. In this review, we provide an updated overview of the actions of the platelet-derived growth factors on neurovascular function, their role in the regulation of perivascular cell types expressing the cognate receptors, neurological diseases associated with aberrance in signalling, and highlight the clinical relevance and therapeutic potentials of this pathway for central nervous system diseases.
Collapse
Affiliation(s)
- Susan Ys Li
- Department of Pharmacology and Centre for Brain Research, Faculty of Medical and Health Sciences, The University of Auckland, Auckland, New Zealand.
| | - Rebecca Johnson
- Department of Pharmacology and Centre for Brain Research, Faculty of Medical and Health Sciences, The University of Auckland, Auckland, New Zealand.
| | - Leon Cd Smyth
- Center for Brain Immunology and Glia, Department of Pathology and Immunology, Washington University in St Louis, MO, USA.
| | - Mike Dragunow
- Department of Pharmacology and Centre for Brain Research, Faculty of Medical and Health Sciences, The University of Auckland, Auckland, New Zealand.
| |
Collapse
|
26
|
Chen JWE, Leary S, Barnhouse V, Sarkaria JN, Harley BA. Matrix Hyaluronic Acid and Hypoxia Influence a CD133 + Subset of Patient-Derived Glioblastoma Cells. Tissue Eng Part A 2022; 28:330-340. [PMID: 34435883 PMCID: PMC9057908 DOI: 10.1089/ten.tea.2021.0117] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2021] [Accepted: 08/24/2021] [Indexed: 11/12/2022] Open
Abstract
Glioblastoma (GBM) displays diffusive invasion throughout the brain microenvironment, which is partially responsible for its short median survival rate (<15 months). Stem-like subpopulations (GBM stem-like cells, GSCs) are believed to play a central role in therapeutic resistance and poor patient prognosis. Given the extensive tissue remodeling and processes such as vessel co-option and regression that occur in the tumor microenvironment, it is essential to understand the role of metabolic constraint such as hypoxia on GBM cell populations. This work describes the use of a multidimensional gelatin hydrogel to culture patient-derived GBM cells, to evaluate the influence of hypoxia and the inclusion brain-mimetic hyaluronic acid on the relative activity of GSCs versus overall GBM cells. Notably, CD133+ GBM cell fraction is crucial for robust formation of tumor spheroids in multidimensional cultures. In addition, while the relative size of the CD133+ GBM subpopulation increased in response to both hypoxia and matrix-bound hyaluronan, we did not observe cell subtype-specific changes in invasion signaling pathway activation. Taken together, this study highlights the potential of biomimetic culture systems for resolving changes in the population dynamics and behavior of subsets of GBM specimens for the future development of precision medicine applications. Impact Statement This study describes a gelatin hydrogel platform to investigate the role of extracellular hyaluronic acid and hypoxia on the behavior of a CD133+ subset of cells within patient-derived glioblastoma (GBM) specimens. We report that the relative expansion of the CD133+ GBM stem cell-like population is strongly responsive to extracellular cues, highlighting the significance of biomimetic hydrogel models of the tumor microenvironment to investigate invasion and therapeutic response.
Collapse
Affiliation(s)
- Jee-Wei Emily Chen
- Department of Chemical and Biomolecular Engineering, University of Illinois at Urbana-Champaign, Urbana, Illinois, USA
- Carl R. Woese Institute for Genomic Biology, University of Illinois at Urbana-Champaign, Urbana, Illinois, USA
| | - Sarah Leary
- Department of Chemistry, and University of Illinois at Urbana-Champaign, Urbana, Illinois, USA
| | - Victoria Barnhouse
- Department of Bioengineering, University of Illinois at Urbana-Champaign, Urbana, Illinois, USA
| | - Jann N. Sarkaria
- Department of Radiation Oncology, Mayo Clinic, Rochester, Minnesota, USA
| | - Brendan A.C. Harley
- Department of Chemical and Biomolecular Engineering, University of Illinois at Urbana-Champaign, Urbana, Illinois, USA
- Carl R. Woese Institute for Genomic Biology, University of Illinois at Urbana-Champaign, Urbana, Illinois, USA
- Cancer Center at Illinois, University of Illinois at Urbana-Champaign, Urbana, Illinois, USA
| |
Collapse
|
27
|
Hatlen RR, Rajagopalan P. Environmental interplay: Stromal cells and biomaterial composition influence in the glioblastoma microenvironment. Acta Biomater 2021; 132:421-436. [PMID: 33276155 DOI: 10.1016/j.actbio.2020.11.044] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2020] [Revised: 11/24/2020] [Accepted: 11/25/2020] [Indexed: 12/12/2022]
Abstract
Glioblastoma multiforme (GBM) is the most deadly form of brain cancer. Recurrence is common, and established therapies have not been able to significantly extend overall patient survival. One platform through which GBM research can progress is to design biomimetic systems for discovery and investigation into the mechanisms of invasion, cellular properties, as well as the efficacy of therapies. In this review, 2D and 3D GBM in vitro cultures will be discussed. We focus on the effects of biomaterial properties, interactions between stromal cells, and vascular influence on cancer cell survival and progression. This review will summarize critical findings in each of these areas and how they have led to a more comprehensive scientific understanding of GBM. STATEMENT OF SIGNIFICANCE: Glioblastoma multiforme (GBM) is the most deadly form of brain cancer. Recurrence is common, and established therapies have not been able to significantly extend overall patient survival. One platform through which GBM research can progress is to design biomimetic systems for discovery and investigation into the mechanisms of invasion, cellular properties, as well as the efficacy of therapies. In this review, 2D and 3D GBM in vitro cultures will be discussed. We focus on the effects of biomaterial properties, interactions between stromal cells and vascular influence on cancer cell survival and progression. This review will summarize critical findings in each of these areas and how they have lead to a more comprehensive scientific understanding of GBM.
Collapse
Affiliation(s)
- Rosalyn R Hatlen
- Department of Chemical Engineering, Virginia Tech, Blacksburg, VA 24061, United States
| | | |
Collapse
|
28
|
Chen J, Lee H, Schmitt P, Choy CJ, Miller DM, Williams BJ, Bearer EL, Frieboes HB. Bioengineered Models to Study Microenvironmental Regulation of Glioblastoma Metabolism. J Neuropathol Exp Neurol 2021; 80:1012–1023. [PMID: 34524448 DOI: 10.1093/jnen/nlab092] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Despite extensive research and aggressive therapies, glioblastoma (GBM) remains a central nervous system malignancy with poor prognosis. The varied histopathology of GBM suggests a landscape of differing microenvironments and clonal expansions, which may influence metabolism, driving tumor progression. Indeed, GBM metabolic plasticity in response to differing nutrient supply within these microenvironments has emerged as a key driver of aggressiveness. Additionally, emergent biophysical and biochemical interactions in the tumor microenvironment (TME) are offering new perspectives on GBM metabolism. Perivascular and hypoxic niches exert crucial roles in tumor maintenance and progression, facilitating metabolic relationships between stromal and tumor cells. Alterations in extracellular matrix and its biophysical characteristics, such as rigidity and topography, regulate GBM metabolism through mechanotransductive mechanisms. This review highlights insights gained from deployment of bioengineering models, including engineered cell culture and mathematical models, to study the microenvironmental regulation of GBM metabolism. Bioengineered approaches building upon histopathology measurements may uncover potential therapeutic strategies that target both TME-dependent mechanotransductive and biomolecular drivers of metabolism to tackle this challenging disease. Longer term, a concerted effort integrating in vitro and in silico models predictive of patient therapy response may offer a powerful advance toward tailoring of treatment to patient-specific GBM characteristics.
Collapse
Affiliation(s)
- Joseph Chen
- From the Department of Bioengineering, University of Louisville, Louisville, Kentucky, USA (JC, CJC, HBF); Department of Pharmacology and Toxicology, University of Louisville, Louisville, Kentucky, USA (JC, DMM, HBF); Department of Neurological Surgery, University of Louisville, Louisville, Kentucky, USA (HL, BJW); Department of Medicine, University of Louisville, Louisville, Kentucky, USA (PS, DMM); Department of Radiation Oncology, James Graham Brown Cancer Center, University of Louisville, Louisville, Kentucky, USA (DMM, BJW, HBF); Center for Predictive Medicine, University of Louisville, Louisville, Kentucky, USA (HBF); Department of Pathology, University of New Mexico Health Sciences Center, Albuquerque, New Mexico, USA (ELB)
| | - Hyunchul Lee
- From the Department of Bioengineering, University of Louisville, Louisville, Kentucky, USA (JC, CJC, HBF); Department of Pharmacology and Toxicology, University of Louisville, Louisville, Kentucky, USA (JC, DMM, HBF); Department of Neurological Surgery, University of Louisville, Louisville, Kentucky, USA (HL, BJW); Department of Medicine, University of Louisville, Louisville, Kentucky, USA (PS, DMM); Department of Radiation Oncology, James Graham Brown Cancer Center, University of Louisville, Louisville, Kentucky, USA (DMM, BJW, HBF); Center for Predictive Medicine, University of Louisville, Louisville, Kentucky, USA (HBF); Department of Pathology, University of New Mexico Health Sciences Center, Albuquerque, New Mexico, USA (ELB)
| | - Philipp Schmitt
- From the Department of Bioengineering, University of Louisville, Louisville, Kentucky, USA (JC, CJC, HBF); Department of Pharmacology and Toxicology, University of Louisville, Louisville, Kentucky, USA (JC, DMM, HBF); Department of Neurological Surgery, University of Louisville, Louisville, Kentucky, USA (HL, BJW); Department of Medicine, University of Louisville, Louisville, Kentucky, USA (PS, DMM); Department of Radiation Oncology, James Graham Brown Cancer Center, University of Louisville, Louisville, Kentucky, USA (DMM, BJW, HBF); Center for Predictive Medicine, University of Louisville, Louisville, Kentucky, USA (HBF); Department of Pathology, University of New Mexico Health Sciences Center, Albuquerque, New Mexico, USA (ELB)
| | - Caleb J Choy
- From the Department of Bioengineering, University of Louisville, Louisville, Kentucky, USA (JC, CJC, HBF); Department of Pharmacology and Toxicology, University of Louisville, Louisville, Kentucky, USA (JC, DMM, HBF); Department of Neurological Surgery, University of Louisville, Louisville, Kentucky, USA (HL, BJW); Department of Medicine, University of Louisville, Louisville, Kentucky, USA (PS, DMM); Department of Radiation Oncology, James Graham Brown Cancer Center, University of Louisville, Louisville, Kentucky, USA (DMM, BJW, HBF); Center for Predictive Medicine, University of Louisville, Louisville, Kentucky, USA (HBF); Department of Pathology, University of New Mexico Health Sciences Center, Albuquerque, New Mexico, USA (ELB)
| | - Donald M Miller
- From the Department of Bioengineering, University of Louisville, Louisville, Kentucky, USA (JC, CJC, HBF); Department of Pharmacology and Toxicology, University of Louisville, Louisville, Kentucky, USA (JC, DMM, HBF); Department of Neurological Surgery, University of Louisville, Louisville, Kentucky, USA (HL, BJW); Department of Medicine, University of Louisville, Louisville, Kentucky, USA (PS, DMM); Department of Radiation Oncology, James Graham Brown Cancer Center, University of Louisville, Louisville, Kentucky, USA (DMM, BJW, HBF); Center for Predictive Medicine, University of Louisville, Louisville, Kentucky, USA (HBF); Department of Pathology, University of New Mexico Health Sciences Center, Albuquerque, New Mexico, USA (ELB)
| | - Brian J Williams
- From the Department of Bioengineering, University of Louisville, Louisville, Kentucky, USA (JC, CJC, HBF); Department of Pharmacology and Toxicology, University of Louisville, Louisville, Kentucky, USA (JC, DMM, HBF); Department of Neurological Surgery, University of Louisville, Louisville, Kentucky, USA (HL, BJW); Department of Medicine, University of Louisville, Louisville, Kentucky, USA (PS, DMM); Department of Radiation Oncology, James Graham Brown Cancer Center, University of Louisville, Louisville, Kentucky, USA (DMM, BJW, HBF); Center for Predictive Medicine, University of Louisville, Louisville, Kentucky, USA (HBF); Department of Pathology, University of New Mexico Health Sciences Center, Albuquerque, New Mexico, USA (ELB)
| | - Elaine L Bearer
- From the Department of Bioengineering, University of Louisville, Louisville, Kentucky, USA (JC, CJC, HBF); Department of Pharmacology and Toxicology, University of Louisville, Louisville, Kentucky, USA (JC, DMM, HBF); Department of Neurological Surgery, University of Louisville, Louisville, Kentucky, USA (HL, BJW); Department of Medicine, University of Louisville, Louisville, Kentucky, USA (PS, DMM); Department of Radiation Oncology, James Graham Brown Cancer Center, University of Louisville, Louisville, Kentucky, USA (DMM, BJW, HBF); Center for Predictive Medicine, University of Louisville, Louisville, Kentucky, USA (HBF); Department of Pathology, University of New Mexico Health Sciences Center, Albuquerque, New Mexico, USA (ELB)
| | - Hermann B Frieboes
- From the Department of Bioengineering, University of Louisville, Louisville, Kentucky, USA (JC, CJC, HBF); Department of Pharmacology and Toxicology, University of Louisville, Louisville, Kentucky, USA (JC, DMM, HBF); Department of Neurological Surgery, University of Louisville, Louisville, Kentucky, USA (HL, BJW); Department of Medicine, University of Louisville, Louisville, Kentucky, USA (PS, DMM); Department of Radiation Oncology, James Graham Brown Cancer Center, University of Louisville, Louisville, Kentucky, USA (DMM, BJW, HBF); Center for Predictive Medicine, University of Louisville, Louisville, Kentucky, USA (HBF); Department of Pathology, University of New Mexico Health Sciences Center, Albuquerque, New Mexico, USA (ELB)
| |
Collapse
|
29
|
Balaziova E, Vymola P, Hrabal P, Mateu R, Zubal M, Tomas R, Netuka D, Kramar F, Zemanova Z, Svobodova K, Brabec M, Sedo A, Busek P. Fibroblast Activation Protein Expressing Mesenchymal Cells Promote Glioblastoma Angiogenesis. Cancers (Basel) 2021; 13:cancers13133304. [PMID: 34282761 PMCID: PMC8267680 DOI: 10.3390/cancers13133304] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2021] [Revised: 06/25/2021] [Accepted: 06/26/2021] [Indexed: 12/27/2022] Open
Abstract
Simple Summary The perivascular niche in glioblastoma is crucial for maintaining a tumour- permissive microenvironment. In various extracranial cancers, mesenchymal cells that express fibroblast activation protein (FAP) are an important stromal component and a potential therapeutic target. In this study, we examine their functions in the glioblastoma microenvironment where their role is so far largely unexplored. Glioblastoma-associated FAP+ mesenchymal cells are localised around activated endothelial cells and their presence positively correlates with vascular density. They represent a subpopulation of stromal, non-tumorigenic cells which mostly lack the chromosomal aberrations characteristic of glioma cells. By soluble factors they induce angiogenic sprouting, chemotaxis of endothelial cells, contribute to destabilisation of blood vessels, and increase the migration and growth of glioma cells. Taken together, we identified a subpopulation of FAP+ mesenchymal cells in the perivascular niche in glioblastoma that may contribute to tumour progression by promoting angiogenesis and supporting dissemination of transformed cells into the surrounding tissue. Abstract Fibroblast activation protein (FAP) is a membrane-bound protease that is upregulated in a wide range of tumours and viewed as a marker of tumour-promoting stroma. Previously, we demonstrated increased FAP expression in glioblastomas and described its localisation in cancer and stromal cells. In this study, we show that FAP+ stromal cells are mostly localised in the vicinity of activated CD105+ endothelial cells and their quantity positively correlates with glioblastoma vascularisation. FAP+ mesenchymal cells derived from human glioblastomas are non-tumorigenic and mostly lack the cytogenetic aberrations characteristic of glioblastomas. Conditioned media from these cells induce angiogenic sprouting and chemotaxis of endothelial cells and promote migration and growth of glioma cells. In a chorioallantoic membrane assay, co-application of FAP+ mesenchymal cells with glioma cells was associated with enhanced abnormal angiogenesis, as evidenced by an increased number of erythrocytes in vessel-like structures and higher occurrence of haemorrhages. FAP+ mesenchymal cells express proangiogenic factors, but in comparison to normal pericytes exhibit decreased levels of antiangiogenic molecules and an increased Angiopoietin 2/1 ratio. Our results show that FAP+ mesenchymal cells promote angiogenesis and glioma cell migration and growth by paracrine communication and in this manner, they may thus contribute to glioblastoma progression.
Collapse
Affiliation(s)
- Eva Balaziova
- Laboratory of Cancer Cell Biology, Institute of Biochemistry and Experimental Oncology, First Faculty of Medicine, Charles University, 128 00 Prague, Czech Republic; (E.B.); (P.V.); (R.M.); (M.Z.)
| | - Petr Vymola
- Laboratory of Cancer Cell Biology, Institute of Biochemistry and Experimental Oncology, First Faculty of Medicine, Charles University, 128 00 Prague, Czech Republic; (E.B.); (P.V.); (R.M.); (M.Z.)
| | - Petr Hrabal
- Department of Pathology, Military University Hospital, 169 02 Prague, Czech Republic;
| | - Rosana Mateu
- Laboratory of Cancer Cell Biology, Institute of Biochemistry and Experimental Oncology, First Faculty of Medicine, Charles University, 128 00 Prague, Czech Republic; (E.B.); (P.V.); (R.M.); (M.Z.)
| | - Michal Zubal
- Laboratory of Cancer Cell Biology, Institute of Biochemistry and Experimental Oncology, First Faculty of Medicine, Charles University, 128 00 Prague, Czech Republic; (E.B.); (P.V.); (R.M.); (M.Z.)
| | - Robert Tomas
- Departments of Neurosurgery, Na Homolce Hospital, 150 00 Prague, Czech Republic;
| | - David Netuka
- Department of Neurosurgery and Neurooncology, First Faculty of Medicine, Charles University and Military University Hospital, 168 02 Prague, Czech Republic; (D.N.); (F.K.)
| | - Filip Kramar
- Department of Neurosurgery and Neurooncology, First Faculty of Medicine, Charles University and Military University Hospital, 168 02 Prague, Czech Republic; (D.N.); (F.K.)
| | - Zuzana Zemanova
- Center of Oncocytogenomics, Institute of Clinical Biochemistry and Laboratory Diagnostics, General University Hospital and First Faculty of Medicine, Charles University, 128 00 Prague, Czech Republic; (Z.Z.); (K.S.)
| | - Karla Svobodova
- Center of Oncocytogenomics, Institute of Clinical Biochemistry and Laboratory Diagnostics, General University Hospital and First Faculty of Medicine, Charles University, 128 00 Prague, Czech Republic; (Z.Z.); (K.S.)
| | - Marek Brabec
- Institute of Computer Science, The Czech Academy of Sciences, 128 00 Prague, Czech Republic;
| | - Aleksi Sedo
- Laboratory of Cancer Cell Biology, Institute of Biochemistry and Experimental Oncology, First Faculty of Medicine, Charles University, 128 00 Prague, Czech Republic; (E.B.); (P.V.); (R.M.); (M.Z.)
- Correspondence: (A.S.); (P.B.)
| | - Petr Busek
- Laboratory of Cancer Cell Biology, Institute of Biochemistry and Experimental Oncology, First Faculty of Medicine, Charles University, 128 00 Prague, Czech Republic; (E.B.); (P.V.); (R.M.); (M.Z.)
- Correspondence: (A.S.); (P.B.)
| |
Collapse
|
30
|
Cai X, Yuan F, Zhu J, Yang J, Tang C, Cong Z, Ma C. Glioma-Associated Stromal Cells Stimulate Glioma Malignancy by Regulating the Tumor Immune Microenvironment. Front Oncol 2021; 11:672928. [PMID: 33996602 PMCID: PMC8117153 DOI: 10.3389/fonc.2021.672928] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2021] [Accepted: 04/12/2021] [Indexed: 12/21/2022] Open
Abstract
Background The glioma-associated stromal cell (GASC) is a recently identified type of cell in the glioma microenvironment and may be a prognostic marker for glioma. However, the potential mechanisms of GASCs in the glioma microenvironment remain largely unknown. In this work, we aimed to explore the mechanisms of GASCs in gliomas, particularly in high-grade gliomas (HGG). Methods We used glioma datasets from The Cancer Genome Atlas (TCGA) and the Chinese Glioma Genome Atlas (CGGA). We utilized the Single-sample Gene Set Enrichment Analysis (ssGSEA) algorithm to discriminate between patients with high or low GASC composition. The xCELL and CIBERSORT algorithms were used to analyze the composition of stromal cells and immune cells. Risk score and a nomogram model were constructed for prognostic prediction of glioma. Results We observed for the first time that the levels of M2 macrophages and immune checkpoints (PD-1, PD-L1, PD-L2, TIM3, Galectin-9, CTLA-4, CD80, CD86, CD155, and CIITA) were significantly higher in the high GASC group and showed positive correlation with the GASC score in all glioma population and the HGG population. Copy number variations of DR3 and CIITA were higher in the high-GASC group. THY1, one of the GASC markers, exhibited lower methylation in the high GASC group. The constructed risk score was an independent predictor of glioma prognostics. Finally, a credible nomogram based on the risk score was established. Conclusions GASCs stimulate glioma malignancy through the M2 macrophage, and are associated with the level of immune checkpoints in the glioma microenvironment. The methylation of THY1 could be used as prognostic indicator and treatment target for glioma. However, further studies are required to verify these findings.
Collapse
Affiliation(s)
- Xiangming Cai
- School of Medicine, Southeast University, Nanjing, China
| | - Feng Yuan
- School of Medicine, Nanjing University, Nanjing, China.,Department of Neurosurgery, Jinling Hospital, Nanjing, China
| | - Junhao Zhu
- School of Medicine, Nanjing Medical University, Nanjing, China
| | - Jin Yang
- Department of Neurosurgery, Jinling Hospital, Nanjing, China
| | - Chao Tang
- Department of Neurosurgery, Jinling Hospital, Nanjing, China
| | - Zixiang Cong
- School of Medicine, Nanjing University, Nanjing, China.,Department of Neurosurgery, Jinling Hospital, Nanjing, China
| | - Chiyuan Ma
- School of Medicine, Southeast University, Nanjing, China.,School of Medicine, Nanjing University, Nanjing, China.,Department of Neurosurgery, Jinling Hospital, Nanjing, China.,School of Medicine, Nanjing Medical University, Nanjing, China
| |
Collapse
|
31
|
Virtuoso A, Giovannoni R, De Luca C, Gargano F, Cerasuolo M, Maggio N, Lavitrano M, Papa M. The Glioblastoma Microenvironment: Morphology, Metabolism, and Molecular Signature of Glial Dynamics to Discover Metabolic Rewiring Sequence. Int J Mol Sci 2021; 22:3301. [PMID: 33804873 PMCID: PMC8036663 DOI: 10.3390/ijms22073301] [Citation(s) in RCA: 45] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2021] [Revised: 03/17/2021] [Accepted: 03/22/2021] [Indexed: 02/07/2023] Open
Abstract
Different functional states determine glioblastoma (GBM) heterogeneity. Brain cancer cells coexist with the glial cells in a functional syncytium based on a continuous metabolic rewiring. However, standard glioma therapies do not account for the effects of the glial cells within the tumor microenvironment. This may be a possible reason for the lack of improvements in patients with high-grade gliomas therapies. Cell metabolism and bioenergetic fitness depend on the availability of nutrients and interactions in the microenvironment. It is strictly related to the cell location in the tumor mass, proximity to blood vessels, biochemical gradients, and tumor evolution, underlying the influence of the context and the timeline in anti-tumor therapeutic approaches. Besides the cancer metabolic strategies, here we review the modifications found in the GBM-associated glia, focusing on morphological, molecular, and metabolic features. We propose to analyze the GBM metabolic rewiring processes from a systems biology perspective. We aim at defining the crosstalk between GBM and the glial cells as modules. The complex networking may be expressed by metabolic modules corresponding to the GBM growth and spreading phases. Variation in the oxidative phosphorylation (OXPHOS) rate and regulation appears to be the most important part of the metabolic and functional heterogeneity, correlating with glycolysis and response to hypoxia. Integrated metabolic modules along with molecular and morphological features could allow the identification of key factors for controlling the GBM-stroma metabolism in multi-targeted, time-dependent therapies.
Collapse
Affiliation(s)
- Assunta Virtuoso
- Laboratory of Neuronal Networks, Department of Mental and Physical Health and Preventive Medicine, University of Campania ‘‘Luigi Vanvitelli”, 80138 Naples, Italy; (A.V.); (F.G.); (M.C.); (M.P.)
- School of Medicine and Surgery, University of Milano-Bicocca, 20900 Monza, Italy;
| | | | - Ciro De Luca
- Laboratory of Neuronal Networks, Department of Mental and Physical Health and Preventive Medicine, University of Campania ‘‘Luigi Vanvitelli”, 80138 Naples, Italy; (A.V.); (F.G.); (M.C.); (M.P.)
| | - Francesca Gargano
- Laboratory of Neuronal Networks, Department of Mental and Physical Health and Preventive Medicine, University of Campania ‘‘Luigi Vanvitelli”, 80138 Naples, Italy; (A.V.); (F.G.); (M.C.); (M.P.)
| | - Michele Cerasuolo
- Laboratory of Neuronal Networks, Department of Mental and Physical Health and Preventive Medicine, University of Campania ‘‘Luigi Vanvitelli”, 80138 Naples, Italy; (A.V.); (F.G.); (M.C.); (M.P.)
| | - Nicola Maggio
- Department of Neurology, Sackler Faculty of Medicine, Sagol School of Neuroscience, Tel Aviv University, Tel Aviv 6997801, Israel;
- Department of Neurology, The Chaim Sheba Medical Center, Ramat Gan 5211401, Israel
| | - Marialuisa Lavitrano
- School of Medicine and Surgery, University of Milano-Bicocca, 20900 Monza, Italy;
| | - Michele Papa
- Laboratory of Neuronal Networks, Department of Mental and Physical Health and Preventive Medicine, University of Campania ‘‘Luigi Vanvitelli”, 80138 Naples, Italy; (A.V.); (F.G.); (M.C.); (M.P.)
- SYSBIO Centre of Systems Biology ISBE-IT, University of Milano-Bicocca, 20126 Milan, Italy
| |
Collapse
|
32
|
Sharma P, Singh SS, Gayana S. Fibroblast Activation Protein Inhibitor PET/CT: A Promising Molecular Imaging Tool. Clin Nucl Med 2021; 46:e141-e150. [PMID: 33351507 DOI: 10.1097/rlu.0000000000003489] [Citation(s) in RCA: 77] [Impact Index Per Article: 19.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
PURPOSE Fibroblast activation protein (FAP) is a cell membrane-bound serine peptidase, overexpressed in cancer-associated fibroblasts and activated fibroblasts at wound healing/inflammatory sites. Recently, molecular PET/CT imaging with radiolabeled FAP inhibitor (FAPI) has been evaluated in different diseases. We aimed to assess its potential role based on the available literature. PATIENTS AND METHODS We conducted a comprehensive review of the available preclinical and clinical data on FAPI PET/CT in an attempt to summarize its current status and potential future role. Based on that, we have discussed the pathophysiology behind FAP-based imaging, followed by a discussion of FAPI radiopharmaceuticals including their synthesis, biodistribution, and dosimetry. Next, we have discussed studies evaluating FAPI PET/CT in different oncological and nononcological pathologies. The potential of FAPI PET/CT in theranostics has also been addressed. RESULTS Based on the early scientific evidence available, including preclinical and clinical studies, FAPI PET/CT seems to be a promising molecular imaging tool, especially in oncology. It can be used for imaging different types of cancers and outperforms 18F-FDG PET/CT in some of these. Its potential as a theranostic tool warrants special attention. CONCLUSIONS Fibroblast activation protein inhibitor PET/CT has the potential to emerge as a powerful molecular imaging tool in the future. However, as of yet, the available evidence is limited, warranting further research and trials in this field.
Collapse
Affiliation(s)
- Punit Sharma
- From the Department of Nuclear Medicine and PET/CT, Apollo Gleneagles Hospital, Kolkata
| | | | | |
Collapse
|
33
|
Liu HL, Wang YN, Feng SY. Brain tumors: Cancer stem-like cells interact with tumor microenvironment. World J Stem Cells 2020; 12:1439-1454. [PMID: 33505594 PMCID: PMC7789119 DOI: 10.4252/wjsc.v12.i12.1439] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/29/2020] [Revised: 10/07/2020] [Accepted: 10/27/2020] [Indexed: 02/06/2023] Open
Abstract
Cancer stem-like cells (CSCs) with potential of self-renewal drive tumorigenesis. Brain tumor microenvironment (TME) has been identified as a critical regulator of malignancy progression. Many researchers are searching new ways to characterize tumors with the goal of predicting how they respond to treatment. Here, we describe the striking parallels between normal stem cells and CSCs. We review the microenvironmental aspects of brain tumors, in particular composition and vital roles of immune cells infiltrating glioma and medulloblastoma. By highlighting that CSCs cooperate with TME via various cellular communication approaches, we discuss the recent advances in therapeutic strategies targeting the components of TME. Identification of the complex and interconnected factors can facilitate the development of promising treatments for these deadly malignancies.
Collapse
Affiliation(s)
- Hai-Long Liu
- Department of Neurosurgery, Chinese PLA General Hospital, Beijing 100853, China
| | - Ya-Nan Wang
- Department of Pathology, Affiliated Hospital of Hebei University, Baoding 071000, Hebei Province, China
| | - Shi-Yu Feng
- Department of Neurosurgery, Chinese PLA General Hospital, Beijing 100853, China
| |
Collapse
|
34
|
Li M, Li G, Kiyokawa J, Tirmizi Z, Richardson LG, Ning J, Das S, Martuza RL, Stemmer-Rachamimov A, Rabkin SD, Wakimoto H. Characterization and oncolytic virus targeting of FAP-expressing tumor-associated pericytes in glioblastoma. Acta Neuropathol Commun 2020; 8:221. [PMID: 33308315 PMCID: PMC7730751 DOI: 10.1186/s40478-020-01096-0] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2020] [Accepted: 11/26/2020] [Indexed: 12/16/2022] Open
Abstract
Cancer-associated fibroblasts (CAFs) are activated fibroblasts constituting the major stromal components in many types of cancer. CAFs contribute to hallmarks of cancer such as proliferation, invasion and immunosuppressive tumor microenvironment, and are associated with poor prognosis of patients with cancer. However, in glioblastoma (GBM), the most common and aggressive primary malignant brain tumor, our knowledge about CAFs or CAF-like stromal cells is limited. Here, using commonly accepted CAF markers, we characterized CAF-like cell populations in clinical glioma specimens and datasets along with mouse models of GBM. We found that tumor-associated pericytes marked by co-expression of fibroblast activation protein α (FAP) and PDGFRβ represent major stromal cell subsets in both human GBM and mouse GBM models, while a fraction of mesenchymal neoplastic cells also express FAP in patient tumors. Since oncolytic viruses can kill cancer cells and simultaneously modulate the tumor microenvironment by impacting non-neoplastic populations such as immune cells and tumor vasculature, we further investigated the ability of oncolytic viruses to target GBM-associated stromal cells. An oncolytic adenovirus, ICOVIR15, carrying ∆24-E1A and an RGD-fiber, infects and depletes FAP+ pericytes as well as GBM cells in murine GBM. Our study thus identifies FAP+/PDGFRβ+ pericytes as a major CAF-like stromal cell population in GBM, and highlights the unique property of this oncolytic adenovirus to target both GBM cells and GBM-associated stromal FAP+ cells.
Collapse
|
35
|
Chen JWE, Lumibao J, Leary S, Sarkaria JN, Steelman AJ, Gaskins HR, Harley BAC. Crosstalk between microglia and patient-derived glioblastoma cells inhibit invasion in a three-dimensional gelatin hydrogel model. J Neuroinflammation 2020; 17:346. [PMID: 33208156 PMCID: PMC7677841 DOI: 10.1186/s12974-020-02026-6] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2020] [Accepted: 11/05/2020] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Glioblastoma is the most common and deadly form of primary brain cancer, accounting for more than 13,000 new diagnoses annually in the USA alone. Microglia are the innate immune cells within the central nervous system, acting as a front-line defense against injuries and inflammation via a process that involves transformation from a quiescent to an activated phenotype. Crosstalk between GBM cells and microglia represents an important axis to consider in the development of tissue engineering platforms to examine pathophysiological processes underlying GBM progression and therapy. METHODS This work used a brain-mimetic hydrogel system to study patient-derived glioblastoma specimens and their interactions with microglia. Here, glioblastoma cells were either cultured alone in 3D hydrogels or in co-culture with microglia in a manner that allowed secretome-based signaling but prevented direct GBM-microglia contact. Patterns of GBM cell invasion were quantified using a three-dimensional spheroid assay. Secretome and transcriptome (via RNAseq) were used to profile the consequences of GBM-microglia interactions. RESULTS Microglia displayed an activated phenotype as a result of GBM crosstalk. Three-dimensional migration patterns of patient-derived glioblastoma cells showed invasion was significantly decreased in response to microglia paracrine signaling. Potential molecular mechanisms underlying with this phenotype were identified from bioinformatic analysis of secretome and RNAseq data. CONCLUSION The data demonstrate a tissue engineered hydrogel platform can be used to investigate crosstalk between immune cells of the tumor microenvironment related to GBM progression. Such multi-dimensional models may provide valuable insight to inform therapeutic innovations to improve GBM treatment.
Collapse
Affiliation(s)
- Jee-Wei Emily Chen
- Department of Chemical & Biomolecular Engineering, University of Illinois at Urbana-Champaign, Urbana, IL, 61801, USA
- Carl R. Woese Institute for Genomic Biology, University of Illinois at Urbana-Champaign, Urbana, IL, 61801, USA
- Department of Materials Science and Engineering, University of Illinois at Urbana-Champaign, Urbana, IL, 61801, USA
| | - Jan Lumibao
- Carl R. Woese Institute for Genomic Biology, University of Illinois at Urbana-Champaign, Urbana, IL, 61801, USA
- Division of Nutritional Sciences, University of Illinois at Urbana-Champaign, Urbana, IL, 61801, USA
- Current Address: Salk Institute for Biological Studies, La Jolla, CA, USA
| | - Sarah Leary
- Department of Chemistry, University of Illinois at Urbana-Champaign, Urbana, IL, 61801, USA
| | - Jann N Sarkaria
- Department of Radiation Oncology, Mayo Clinic, Rochester, MN, USA
| | - Andrew J Steelman
- Department of Animal Sciences, University of Illinois at Urbana-Champaign, 110 Roger Adams Laboratory, 600 S. Mathews Ave, Urbana, IL, 61801, USA
| | - H Rex Gaskins
- Carl R. Woese Institute for Genomic Biology, University of Illinois at Urbana-Champaign, Urbana, IL, 61801, USA
- Division of Nutritional Sciences, University of Illinois at Urbana-Champaign, Urbana, IL, 61801, USA
- Department of Animal Sciences, University of Illinois at Urbana-Champaign, 110 Roger Adams Laboratory, 600 S. Mathews Ave, Urbana, IL, 61801, USA
- Cancer Center at Illinois, University of Illinois at Urbana-Champaign, 110 Roger Adams Laboratory, 600 S. Mathews Ave, Urbana, IL, 61801, USA
| | - Brendan A C Harley
- Department of Chemical & Biomolecular Engineering, University of Illinois at Urbana-Champaign, Urbana, IL, 61801, USA.
- Carl R. Woese Institute for Genomic Biology, University of Illinois at Urbana-Champaign, Urbana, IL, 61801, USA.
- Department of Materials Science and Engineering, University of Illinois at Urbana-Champaign, Urbana, IL, 61801, USA.
- Cancer Center at Illinois, University of Illinois at Urbana-Champaign, 110 Roger Adams Laboratory, 600 S. Mathews Ave, Urbana, IL, 61801, USA.
| |
Collapse
|
36
|
Clavreul A, Menei P. Mesenchymal Stromal-Like Cells in the Glioma Microenvironment: What Are These Cells? Cancers (Basel) 2020; 12:E2628. [PMID: 32942567 PMCID: PMC7565954 DOI: 10.3390/cancers12092628] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2020] [Revised: 09/11/2020] [Accepted: 09/12/2020] [Indexed: 12/13/2022] Open
Abstract
The glioma microenvironment is a critical regulator of tumor progression. It contains different cellular components such as blood vessels, immune cells, and neuroglial cells. It also contains non-cellular components, such as the extracellular matrix, extracellular vesicles, and cytokines, and has certain physicochemical properties, such as low pH, hypoxia, elevated interstitial pressure, and impaired perfusion. This review focuses on a particular type of cells recently identified in the glioma microenvironment: glioma-associated stromal cells (GASCs). This is just one of a number of names given to these mesenchymal stromal-like cells, which have phenotypic and functional properties similar to those of mesenchymal stem cells and cancer-associated fibroblasts. Their close proximity to blood vessels may provide a permissive environment, facilitating angiogenesis, invasion, and tumor growth. Additional studies are required to characterize these cells further and to analyze their role in tumor resistance and recurrence.
Collapse
Affiliation(s)
- Anne Clavreul
- Département de Neurochirurgie, CHU, 49933 Angers, France;
- Université d’Angers, CHU d’Angers, CRCINA, F-49000 Angers, France
| | - Philippe Menei
- Département de Neurochirurgie, CHU, 49933 Angers, France;
- Université d’Angers, CHU d’Angers, CRCINA, F-49000 Angers, France
| |
Collapse
|
37
|
FAP-specific PET signaling shows a moderately positive correlation with relative CBV and no correlation with ADC in 13 IDH wildtype glioblastomas. Eur J Radiol 2020; 127:109021. [PMID: 32344293 DOI: 10.1016/j.ejrad.2020.109021] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2019] [Revised: 02/14/2020] [Accepted: 04/13/2020] [Indexed: 01/27/2023]
Abstract
OBJECTIVES Targeting Fibroblast Activation Protein (FAP) is a new approach for glioblastoma imaging. In a recent pilot study glioblastomas showed elevated tracer uptake with high intratumoral heterogeneity in projection on the corresponding T2w/FLAIR and contrast enhanced MRI lesions. In this study, we correlated FAP-specific signaling with apparent diffusion coefficient (ADC) and relative cerebral blood volume (rCBV) signals in MRI to further characterize the significance of FAP uptake. METHODS Clinical PET/CT scans of 13 glioblastoma patients were performed post i. v. administration of 68Ga-labelled-FAP-specific tracer molecules. PET- and corresponding MRI-scans were co-registrated. 3d volumetric segmentations were performed of T2w/FLAIR lesions and contrast enhancing lesions within co-registrated MRI slides. Signal intensity values of FAP-specific PET signaling, ADC and rCBV were analyzed for their pixel wise correlation in each patient. Pooled estimates of the correlation coefficients were calculated by using the Fisher z-transformation. RESULTS FAP-specific PET signals showed a moderately positive correlation with rCBV values which is more pronounced within the T2w/FLAIR lesion (pooled correlation 0,229) than in the contrast enhancing tumor region (pooled correlation 0.09). FAP-specific PET signals showed no correlation with ADC values. CONCLUSIONS The moderately positive correlation of FAP-specific signals with rCBV values in MRI indicates that FAP-signaling is not independent from perfusion, but also does not only reflect intratumoral perfusion differences. The missing correlation of FAP-specific signals with ADC indicates that FAP-specific imaging does not reflect cell density, but the spot-like expression of FAP in glioblastomas. The clinical value of FAP-specific imaging needs further investigation.
Collapse
|
38
|
Hermida MA, Kumar JD, Schwarz D, Laverty KG, Di Bartolo A, Ardron M, Bogomolnijs M, Clavreul A, Brennan PM, Wiegand UK, Melchels FP, Shu W, Leslie NR. Three dimensional in vitro models of cancer: Bioprinting multilineage glioblastoma models. Adv Biol Regul 2019; 75:100658. [PMID: 31727590 DOI: 10.1016/j.jbior.2019.100658] [Citation(s) in RCA: 52] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2019] [Revised: 09/19/2019] [Accepted: 09/30/2019] [Indexed: 12/18/2022]
Abstract
Three dimensional (3D) bioprinting of multiple cell types within optimised extracellular matrices has the potential to more closely model the 3D environment of human physiology and disease than current alternatives. In this study, we used a multi-nozzle extrusion bioprinter to establish models of glioblastoma made up of cancer and stromal cells printed within matrices comprised of alginate modified with RGDS cell adhesion peptides, hyaluronic acid and collagen-1. Methods were developed using U87MG glioblastoma cells and MM6 monocyte/macrophages, whilst more disease relevant constructs contained glioblastoma stem cells (GSCs), co-printed with glioma associated stromal cells (GASCs) and microglia. Printing parameters were optimised to promote cell-cell interaction, avoiding the 'caging in' of cells due to overly dense cross-linking. Such printing had a negligible effect on cell viability, and cells retained robust metabolic activity and proliferation. Alginate gels allowed the rapid recovery of printed cell protein and RNA, and fluorescent reporters provided analysis of protein kinase activation at the single cell level within printed constructs. GSCs showed more resistance to chemotherapeutic drugs in 3D printed tumour constructs compared to 2D monolayer cultures, reflecting the clinical situation. In summary, a novel 3D bioprinting strategy is developed which allows control over the spatial organisation of tumour constructs for pre-clinical drug sensitivity testing and studies of the tumour microenvironment.
Collapse
Affiliation(s)
- Miguel A Hermida
- Institute of Biological Chemistry, Biophysics & Bioengineering, Heriot Watt University, Edinburgh, UK
| | - Jothi Dinesh Kumar
- Institute of Biological Chemistry, Biophysics & Bioengineering, Heriot Watt University, Edinburgh, UK
| | - Daniela Schwarz
- Institute of Biological Chemistry, Biophysics & Bioengineering, Heriot Watt University, Edinburgh, UK
| | - Keith G Laverty
- Institute of Biological Chemistry, Biophysics & Bioengineering, Heriot Watt University, Edinburgh, UK
| | - Alberto Di Bartolo
- Institute of Biological Chemistry, Biophysics & Bioengineering, Heriot Watt University, Edinburgh, UK
| | - Marcus Ardron
- Renishaw PLC, Research Avenue North, Riccarton, Edinburgh, UK
| | | | - Anne Clavreul
- Département de Neurochirurgie, CHU, Angers, France; CRCINA, INSERM, Université de Nantes, Université D'Angers, France
| | - Paul M Brennan
- Translational Neurosurgery, Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh, UK
| | - Ulrich K Wiegand
- Queens' Medical Research Institute, University of Edinburgh, Edinburgh, UK
| | - Ferry Pw Melchels
- Institute of Biological Chemistry, Biophysics & Bioengineering, Heriot Watt University, Edinburgh, UK
| | - Will Shu
- Biomedical Engineering, University of Strathclyde, Glasgow, UK
| | - Nicholas R Leslie
- Institute of Biological Chemistry, Biophysics & Bioengineering, Heriot Watt University, Edinburgh, UK.
| |
Collapse
|
39
|
Röhrich M, Loktev A, Wefers AK, Altmann A, Paech D, Adeberg S, Windisch P, Hielscher T, Flechsig P, Floca R, Leitz D, Schuster JP, Huber PE, Debus J, von Deimling A, Lindner T, Haberkorn U. IDH-wildtype glioblastomas and grade III/IV IDH-mutant gliomas show elevated tracer uptake in fibroblast activation protein-specific PET/CT. Eur J Nucl Med Mol Imaging 2019; 46:2569-2580. [PMID: 31388723 DOI: 10.1007/s00259-019-04444-y] [Citation(s) in RCA: 97] [Impact Index Per Article: 16.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2019] [Accepted: 07/16/2019] [Indexed: 12/13/2022]
Abstract
PURPOSE Targeting fibroblast activation protein (FAP) is a new diagnostic approach allowing the visualization of tumor stroma. Here, we applied FAP-specific PET imaging to gliomas. We analyzed the target affinity and specificity of two FAP ligands (FAPI-02 and FAPI-04) in vitro, and the pharmacokinetics and biodistribution in mice in vivo. Clinically, we used 68Ga-labeled FAPI-02/04 for PET imaging in 18 glioma patients (five IDH-mutant gliomas, 13 IDH-wildtype glioblastomas). METHODS For binding studies with 177Lu-radiolabeled FAPI-02/04, we used the glioblastoma cell line U87MG, FAP-transfected fibrosarcoma cells, and CD26-transfected human embryonic kidney cells. For pharmacokinetic and biodistribution studies, U87MG-xenografted mice were injected with 68Ga-labeled compounds followed by small-animal PET imaging and 177Lu-labeled FAPI-02/04, respectively. Clinical PET/CT scans were performed 30 min post intravenous administration of 68Ga-FAPI-02/04. PET and MRI scans were co-registrated. Immunohistochemistry was done on 14 gliomas using a FAP-specific antibody. RESULTS FAPI-02 and FAPI-04 showed high binding specificity to FAP. FAPI-04 demonstrated higher tumor accumulation and delayed elimination compared with FAPI-02 in preclinical studies. IDH-wildtype glioblastomas and grade III/IV, but not grade II, IDH-mutant gliomas showed elevated tracer uptake. In glioblastomas, we observed spots with increased uptake in projection on contrast-enhancing areas. Immunohistochemistry showed FAP-positive cells with mainly elongated cell bodies and perivascular FAP-positive cells in glioblastomas and an anaplastic IDH-mutant astrocytoma. CONCLUSIONS Using FAP-specific PET imaging, increased tracer uptake in IDH-wildtype glioblastomas and high-grade IDH-mutant astrocytomas, but not in diffuse astrocytomas, may allow non-invasive distinction between low-grade IDH-mutant and high-grade gliomas. Therefore, FAP-specific imaging in gliomas may be useful for follow-up studies although further clinical evaluation is required.
Collapse
Affiliation(s)
- Manuel Röhrich
- Clinical Cooperation Unit Nuclear Medicine, German Cancer Research Center (DKFZ), Im Neuenheimer Feld 400, 69120, Heidelberg, Germany.
- Department of Nuclear Medicine, University Hospital Heidelberg, Heidelberg, Germany.
| | - Anastasia Loktev
- Clinical Cooperation Unit Nuclear Medicine, German Cancer Research Center (DKFZ), Im Neuenheimer Feld 400, 69120, Heidelberg, Germany
| | - Annika K Wefers
- Department of Neuropathology, Institute of Pathology, University Hospital Heidelberg, Heidelberg, Germany
- Clinical Cooperation Unit Neuropathology, German Consortium for Translational Cancer Research (DKTK), German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Annette Altmann
- Clinical Cooperation Unit Nuclear Medicine, German Cancer Research Center (DKFZ), Im Neuenheimer Feld 400, 69120, Heidelberg, Germany
- Department of Nuclear Medicine, University Hospital Heidelberg, Heidelberg, Germany
| | - Daniel Paech
- Division of Radiology, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Sebastian Adeberg
- Department of Radiation Oncology, University Hospital Heidelberg, Heidelberg, Germany
| | - Paul Windisch
- Department of Radiation Oncology, University Hospital Heidelberg, Heidelberg, Germany
| | - Thomas Hielscher
- Department of Biostatistics, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Paul Flechsig
- Clinical Cooperation Unit Nuclear Medicine, German Cancer Research Center (DKFZ), Im Neuenheimer Feld 400, 69120, Heidelberg, Germany
| | - Ralf Floca
- Division of Medical Image Computing, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Dominik Leitz
- Translational Lung Research Center Heidelberg (TLRC), German Center for Lung Research (DZL), Heidelberg, Germany
| | - Julius P Schuster
- Department of Radiation Oncology, University Hospital Heidelberg, Heidelberg, Germany
- Clinical Cooperation Unit Molecular Radiooncology, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Peter E Huber
- Department of Radiation Oncology, University Hospital Heidelberg, Heidelberg, Germany
- Clinical Cooperation Unit Molecular Radiooncology, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Jürgen Debus
- Department of Radiation Oncology, University Hospital Heidelberg, Heidelberg, Germany
| | - Andreas von Deimling
- Department of Neuropathology, Institute of Pathology, University Hospital Heidelberg, Heidelberg, Germany
- Clinical Cooperation Unit Neuropathology, German Consortium for Translational Cancer Research (DKTK), German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Thomas Lindner
- Clinical Cooperation Unit Nuclear Medicine, German Cancer Research Center (DKFZ), Im Neuenheimer Feld 400, 69120, Heidelberg, Germany
| | - Uwe Haberkorn
- Clinical Cooperation Unit Nuclear Medicine, German Cancer Research Center (DKFZ), Im Neuenheimer Feld 400, 69120, Heidelberg, Germany
- Department of Nuclear Medicine, University Hospital Heidelberg, Heidelberg, Germany
- Translational Lung Research Center Heidelberg (TLRC), German Center for Lung Research (DZL), Heidelberg, Germany
| |
Collapse
|
40
|
Hyaluronic acid-functionalized gelatin hydrogels reveal extracellular matrix signals temper the efficacy of erlotinib against patient-derived glioblastoma specimens. Biomaterials 2019; 219:119371. [PMID: 31352310 DOI: 10.1016/j.biomaterials.2019.119371] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2019] [Revised: 07/11/2019] [Accepted: 07/17/2019] [Indexed: 01/08/2023]
Abstract
Therapeutic options to treat primary glioblastoma (GBM) tumors are scarce. GBM tumors with epidermal growth factor receptor (EGFR) mutations, in particular a constitutively active EGFRvIII mutant, have extremely poor clinical outcomes. GBM tumors with concurrent EGFR amplification and active phosphatase and tensin homolog (PTEN) are sensitive to the tyrosine kinase inhibitor erlotinib, but the effect is not durable. A persistent challenge to improved treatment is the poorly understood role of cellular, metabolic, and biophysical signals from the GBM tumor microenvironment on therapeutic efficacy and acquired resistance. The intractable nature of studying GBM cell in vivo motivates tissue engineering approaches to replicate aspects of the complex GBM tumor microenvironment. Here, we profile the effect of erlotinib on two patient-derived GBM specimens: EGFR + GBM12 and EGFRvIII GBM6. We use a three-dimensional gelatin hydrogel to present brain-mimetic hyaluronic acid (HA) and evaluate the coordinated influence of extracellular matrix signals and EGFR mutation status on GBM cell migration, survival and proliferation, as well as signaling pathway activation in response to cyclic erlotinib exposure. Comparable to results observed in vivo for xenograft tumors, erlotinib exposure is not cytotoxic for GBM6 EGFRvIII specimens. We also identify a role of extracellular HA (via CD44) in altering the effect of erlotinib in GBM EGFR + cells by modifying STAT3 phosphorylation status. Taken together, we report an in vitro tissue engineered platform to monitor signaling associated with poor response to targeted inhibitors in GBM.
Collapse
|
41
|
Clavreul A, Soulard G, Lemée JM, Rigot M, Fabbro-Peray P, Bauchet L, Figarella-Branger D, Menei P. The French glioblastoma biobank (FGB): a national clinicobiological database. J Transl Med 2019; 17:133. [PMID: 31014363 PMCID: PMC6480741 DOI: 10.1186/s12967-019-1859-6] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2019] [Accepted: 03/27/2019] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND Glioblastomas (GB) are the most common and lethal primary brain tumors. Significant progress has been made toward identifying potential risk factors for GB and diagnostic and prognostic biomarkers. However, the current standard of care for newly diagnosed GB, the Stupp protocol, has remained unchanged for over a decade. Large-scale translational programs based on a large clinicobiological database are required to improve our understanding of GB biology, potentially facilitating the development of personalized and specifically targeted therapies. With this goal in mind, a well-annotated clinicobiological database housing data and samples from GB patients has been set up in France: the French GB biobank (FGB). METHODS The biobank contains data and samples from adult GB patients from 24 centers in France providing written informed consent. Clinical and biomaterial data are stored in anonymized certified electronic case report forms. Biological samples (including frozen and formalin-fixed paraffin-embedded tumor tissues, blood samples, and hair) are conserved in certified biological resource centers or tumor tissue banks at each participating center. RESULTS Clinical data and biological materials have been collected for 1087 GB patients. A complete set of samples (tumor, blood and hair) is available for 66%, and at least one frozen tumor sample is available for 88% of the GB patients. CONCLUSIONS This large biobank is unique in Europe and can support the large-scale translational projects required to improve GB care. Additional biological materials, such as peritumoral brain zone and fecal samples, will be collected in the future, to respond to research needs.
Collapse
Affiliation(s)
- Anne Clavreul
- Département de Neurochirurgie, CHU, 4 rue Larrey, 49 933, Angers Cedex 9, France.,CRCINA, INSERM, Université de Nantes, Université d'Angers, Angers, France
| | - Gwénaëlle Soulard
- Département de Neurochirurgie, CHU, 4 rue Larrey, 49 933, Angers Cedex 9, France.,CRCINA, INSERM, Université de Nantes, Université d'Angers, Angers, France
| | - Jean-Michel Lemée
- Département de Neurochirurgie, CHU, 4 rue Larrey, 49 933, Angers Cedex 9, France.,CRCINA, INSERM, Université de Nantes, Université d'Angers, Angers, France
| | - Marion Rigot
- Département Promotion, Direction de la Recherche, CHU Nantes, Nantes, France
| | - Pascale Fabbro-Peray
- Département de Biostatistique, Epidémiologie, Santé Publique, CHU Nîmes, Nîmes, France.,Unité de recherche EA2415, Université de Montpellier, Montpellier, France
| | - Luc Bauchet
- Département de Neurochirurgie, Hôpital Gui de Chauliac, CHU Montpellier, Université de Montpellier, Montpellier, France.,Institut des Neurosciences de Montpellier INSERM U1051, Montpellier, France
| | - Dominique Figarella-Branger
- APHM, Hôpital de la Timone, Service d'Anatomie Pathologique et de Neuropathologie, Marseille, France.,Aix-Marseille Univ, CNRS, INP, Inst Neurophysiopathol, Marseille, France
| | - Philippe Menei
- Département de Neurochirurgie, CHU, 4 rue Larrey, 49 933, Angers Cedex 9, France. .,CRCINA, INSERM, Université de Nantes, Université d'Angers, Angers, France.
| | | |
Collapse
|
42
|
Clavreul A, Pourbaghi-Masouleh M, Roger E, Menei P. Nanocarriers and nonviral methods for delivering antiangiogenic factors for glioblastoma therapy: the story so far. Int J Nanomedicine 2019; 14:2497-2513. [PMID: 31040671 PMCID: PMC6461002 DOI: 10.2147/ijn.s194858] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Angiogenesis, the formation of new blood vessels, is an essential component of glioblastoma (GB) progression. The development of angiogenesis inhibitor therapy, including treatments targeting vascular endothelial growth factor (VEGF) in particular, raised new hopes for the treatment of GB, but no Phase III clinical trial to date has reported survival benefits relative to standard treatment. There are several possible reasons for this limited efficacy, including VEGF-independent angiogenesis, induction of tumor invasion, and inefficient antiangiogenic factor delivery to the tumor. Efforts have been made to overcome these limitations by identifying new angiogenesis inhibitors that target angiogenesis through different mechanisms of action without inducing tumor invasion, and through the development of viral and nonviral delivery methods to improve antiangiogenic activity. Herein, we describe the nonviral methods, including convection-enhanced delivery devices, implantable polymer devices, nanocarriers, and cellular vehicles, to deliver antiangiogenic factors. We focus on those evaluated in intracranial (orthotopic) animal models of GB, the most relevant models of this disease, as they reproduce the clinical scenario of tumor progression and therapy response encountered in GB patients.
Collapse
Affiliation(s)
- Anne Clavreul
- Department of Neurosurgery, CHU, Angers, France, .,CRCINA, INSERM, University of Nantes, University of Angers, Angers, France,
| | - Milad Pourbaghi-Masouleh
- CRCINA, INSERM, University of Nantes, University of Angers, Angers, France, .,Division of Drug Delivery and Tissue Engineering, School of Pharmacy, University of Nottingham, Nottingham, UK
| | - Emilie Roger
- MINT, INSERM 1066, CNRS 6021, University of Angers, Angers, France
| | - Philippe Menei
- Department of Neurosurgery, CHU, Angers, France, .,CRCINA, INSERM, University of Nantes, University of Angers, Angers, France,
| |
Collapse
|
43
|
Ngo MT, Harley BAC. Perivascular signals alter global gene expression profile of glioblastoma and response to temozolomide in a gelatin hydrogel. Biomaterials 2019; 198:122-134. [PMID: 29941152 DOI: 10.1101/273763] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2018] [Revised: 05/30/2018] [Accepted: 06/10/2018] [Indexed: 05/25/2023]
Abstract
Glioblastoma (GBM) is the most common primary malignant brain tumor, with patients exhibiting poor survival (median survival time: 15 months). Difficulties in treating GBM include not only the inability to resect the diffusively-invading tumor cells, but also therapeutic resistance. The perivascular niche (PVN) within the GBM tumor microenvironment contributes significantly to tumor cell invasion, cancer stem cell maintenance, and has been shown to protect tumor cells from radiation and chemotherapy. In this study, we examine how the inclusion of non-tumor cells in culture with tumor cells within a hydrogel impacts the overall gene expression profile of an in vitro artificial perivascular niche (PVN) comprised of endothelial and stromal cells directly cultured with GBM tumor cells within a methacrylamide-functionalized gelatin hydrogel. Using RNA-seq, we demonstrate that genes related to angiogenesis and extracellular matrix remodeling are upregulated in the PVN model compared to hydrogels containing only tumor or perivascular niche cells, while downregulated genes are related to cell cycle and DNA damage repair. Signaling pathways and genes commonly implicated in GBM malignancy, such as MGMT, EGFR, PI3K-Akt signaling, and Ras/MAPK signaling are also upregulated in the PVN model. We describe the kinetics of gene expression within the PVN hydrogels over a course of 14 days, observing the patterns associated with tumor cell-mediated endothelial network co-option and regression. We finally examine the effect of temozolomide, a frontline chemotherapy used clinically against GBM, on the PVN culture. Notably, the PVN model is less responsive to TMZ compared to hydrogels containing only tumor cells. Overall, these results demonstrate that inclusion of cellular and matrix-associated elements of the PVN within an in vitro model of GBM allows for the development of gene expression patterns and therapeutic response relevant to GBM.
Collapse
Affiliation(s)
- Mai T Ngo
- Dept. Chemical and Biomolecular Engineering, University of Illinois at Urbana-Champaign, Urbana, IL, 61801, USA
| | - Brendan A C Harley
- Dept. Chemical and Biomolecular Engineering, University of Illinois at Urbana-Champaign, Urbana, IL, 61801, USA; Carl R. Woese Institute for Genomic Biology, University of Illinois at Urbana-Champaign, Urbana, IL, 61801, USA.
| |
Collapse
|
44
|
Autier L, Clavreul A, Cacicedo ML, Franconi F, Sindji L, Rousseau A, Perrot R, Montero-Menei CN, Castro GR, Menei P. A new glioblastoma cell trap for implantation after surgical resection. Acta Biomater 2019; 84:268-279. [PMID: 30465922 DOI: 10.1016/j.actbio.2018.11.027] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2018] [Revised: 11/09/2018] [Accepted: 11/18/2018] [Indexed: 12/11/2022]
Abstract
Glioblastoma (GB) is a highly infiltrative tumor, recurring, in 90% of cases, within a few centimeters of the surgical resection cavity, even with adjuvant chemo/radiotherapy. Residual GB cells left in the margins or infiltrating the brain parenchyma shelter behind the extremely fragile and sensitive brain tissue and may favor recurrence. Tools for eliminating these cells without damaging the brain microenvironment are urgently required. We propose a strategy involving the implantation, into the tumor bed after resection, of a scaffold to concentrate and trap these cells, to facilitate their destruction by targeted therapies, such as stereotactic radiosurgery. We used bacterial cellulose (BC), an easily synthesized and modifiable random nanofibrous biomaterial, to make the trap. We showed that the structure of BC membranes was ideal for trapping tumor cells and that BC implants were biocompatible with brain parenchyma. We also demonstrated the visibility of BC on magnetic resonance imaging, making it possible to follow its fate in clinical situations and to define the target volume for stereotactic radiosurgery more precisely. Furthermore, BC membranes can be loaded with chemoattractants, which were released and attracted tumor cells in vitro. This is of particular interest for trapping GB cells infiltrating tissues within a few centimeters of the resection cavity. Our data suggest that BC membranes could be a scaffold of choice for implantation after surgical resection to trap residual GB cells. STATEMENT OF SIGNIFICANCE: Glioblastoma is a highly infiltrative tumor, recurring, in 90% of cases, within a few centimeters of the surgical resection cavity, even with adjuvant chemo/radiotherapy. Residual tumor cells left in the margins or infiltrating the brain parenchyma shelter behind the extremely fragile and sensitive brain tissue and contribute to the risk of recurrence. Finding tools to eliminate these cells without damaging the brain microenvironment is a real challenge. We propose a strategy involving the implantation, into the walls of the surgical resection cavity, of a scaffold to concentrate and trap the residual tumor cells, to facilitate their destruction by targeted therapies, such as stereotactic radiosurgery.
Collapse
Affiliation(s)
- Lila Autier
- Département de Neurochirurgie, CHU, Angers, France; CRCINA, INSERM, Université de Nantes, Université d'Angers, Angers, France; Département de Neurologie, CHU, Angers, France
| | - Anne Clavreul
- Département de Neurochirurgie, CHU, Angers, France; CRCINA, INSERM, Université de Nantes, Université d'Angers, Angers, France.
| | - Maximiliano L Cacicedo
- Nanobiomaterials Lab, CINDEFI, School of Sciences, National University of La Plata-CONICET (CCT La Plata), Buenos Aires, Argentina
| | - Florence Franconi
- PRISM, Plate-forme de recherche en imagerie et spectroscopie multi-modales, PRISM-Icat, UNIV Angers, Angers, France; MINT, Micro & Nanomedecines Translationnelles, UNIV Angers, INSERM U1066, CNRS UMR 6021, Angers, France
| | - Laurence Sindji
- CRCINA, INSERM, Université de Nantes, Université d'Angers, Angers, France
| | - Audrey Rousseau
- CRCINA, INSERM, Université de Nantes, Université d'Angers, Angers, France; Laboratoire Pathologie Cellulaire et Tissulaire, CHU, Angers, France
| | - Rodolphe Perrot
- SCIAM, Service Commun d'Imageries et d'Analyses Microscopiques, UNIV Angers, Angers, France
| | | | - Guillermo R Castro
- Nanobiomaterials Lab, CINDEFI, School of Sciences, National University of La Plata-CONICET (CCT La Plata), Buenos Aires, Argentina
| | - Philippe Menei
- Département de Neurochirurgie, CHU, Angers, France; CRCINA, INSERM, Université de Nantes, Université d'Angers, Angers, France
| |
Collapse
|
45
|
Lemée JM, Clavreul A, Aubry M, Com E, de Tayrac M, Mosser J, Menei P. Integration of transcriptome and proteome profiles in glioblastoma: looking for the missing link. BMC Mol Biol 2018; 19:13. [PMID: 30463513 PMCID: PMC6249855 DOI: 10.1186/s12867-018-0115-6] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2018] [Accepted: 11/09/2018] [Indexed: 12/21/2022] Open
Abstract
Background Glioblastoma (GB) is the most common and aggressive tumor of the brain. Genotype-based approaches and independent analyses of the transcriptome or the proteome have led to progress in understanding the underlying biology of GB. Joint transcriptome and proteome profiling may reveal new biological insights, and identify pathogenic mechanisms or therapeutic targets for GB therapy. We present a comparison of transcriptome and proteome data from five GB biopsies (TZ) vs their corresponding peritumoral brain zone (PBZ). Omic analyses were performed using RNA microarray chips and the isotope-coded protein label method (ICPL). Results As described in other cancers, we found a poor correlation between transcriptome and proteome data in GB. We observed only two commonly deregulated mRNAs/proteins (neurofilament light polypeptide and synapsin 1) and 12 altered biological processes; they are related to cell communication, synaptic transmission and nervous system processes. This poor correlation may be a consequence of the techniques used to produce the omic profiles, the intrinsic properties of mRNA and proteins and/or of cancer- or GB-specific phenomena. Of interest, the analysis of the transcription factor binding sites present upstream from the open reading frames of all altered proteins identified by ICPL method shows a common binding site for the topoisomerase I and p53-binding protein TOPORS. Its expression was observed in 7/11 TZ samples and not in PBZ. Some findings suggest that TOPORS may function as a tumor suppressor; its implication in gliomagenesis should be examined in future studies. Conclusions In this study, we showed a low correlation between transcriptome and proteome data for GB samples as described in other cancer tissues. We observed that NEFL, SYN1 and 12 biological processes were deregulated in both the transcriptome and proteome data. It will be important to analyze more specifically these processes and these two proteins to allow the identification of new theranostic markers or potential therapeutic targets for GB. Electronic supplementary material The online version of this article (10.1186/s12867-018-0115-6) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Jean-Michel Lemée
- Department of Neurosurgery, CHU Angers, University Hospital of Angers, 4, Rue Larrey, 49933, Angers Cedex 09, France. .,CRCINA, INSERM, Université de Nantes, Université d'Angers, Angers, France.
| | - Anne Clavreul
- Department of Neurosurgery, CHU Angers, University Hospital of Angers, 4, Rue Larrey, 49933, Angers Cedex 09, France.,CRCINA, INSERM, Université de Nantes, Université d'Angers, Angers, France
| | - Marc Aubry
- UEB, UMS 3480 Biosit, Faculté de Médecine, Université Rennes 1, Rennes, France.,Plate-forme Génomique Santé Biosit, Université Rennes 1, Rennes, France
| | - Emmanuelle Com
- Inserm U1085 IRSET, Université de Rennes 1, Rennes, France.,Protim, Université de Rennes 1, Rennes, France
| | - Marie de Tayrac
- UEB, UMS 3480 Biosit, Faculté de Médecine, Université Rennes 1, Rennes, France.,Service de Génétique Moléculaire et Génomique, CHU Rennes, Rennes, France.,CNRS, UMR 6290, Institut de Génétique et Développement de Rennes (IGdR), Rennes, France
| | - Jean Mosser
- UEB, UMS 3480 Biosit, Faculté de Médecine, Université Rennes 1, Rennes, France.,Plate-forme Génomique Santé Biosit, Université Rennes 1, Rennes, France.,Service de Génétique Moléculaire et Génomique, CHU Rennes, Rennes, France.,CNRS, UMR 6290, Institut de Génétique et Développement de Rennes (IGdR), Rennes, France
| | - Philippe Menei
- Department of Neurosurgery, CHU Angers, University Hospital of Angers, 4, Rue Larrey, 49933, Angers Cedex 09, France.,CRCINA, INSERM, Université de Nantes, Université d'Angers, Angers, France
| |
Collapse
|
46
|
Ngo MT, Harley BAC. Perivascular signals alter global gene expression profile of glioblastoma and response to temozolomide in a gelatin hydrogel. Biomaterials 2018; 198:122-134. [PMID: 29941152 DOI: 10.1016/j.biomaterials.2018.06.013] [Citation(s) in RCA: 46] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2018] [Revised: 05/30/2018] [Accepted: 06/10/2018] [Indexed: 12/22/2022]
Abstract
Glioblastoma (GBM) is the most common primary malignant brain tumor, with patients exhibiting poor survival (median survival time: 15 months). Difficulties in treating GBM include not only the inability to resect the diffusively-invading tumor cells, but also therapeutic resistance. The perivascular niche (PVN) within the GBM tumor microenvironment contributes significantly to tumor cell invasion, cancer stem cell maintenance, and has been shown to protect tumor cells from radiation and chemotherapy. In this study, we examine how the inclusion of non-tumor cells in culture with tumor cells within a hydrogel impacts the overall gene expression profile of an in vitro artificial perivascular niche (PVN) comprised of endothelial and stromal cells directly cultured with GBM tumor cells within a methacrylamide-functionalized gelatin hydrogel. Using RNA-seq, we demonstrate that genes related to angiogenesis and extracellular matrix remodeling are upregulated in the PVN model compared to hydrogels containing only tumor or perivascular niche cells, while downregulated genes are related to cell cycle and DNA damage repair. Signaling pathways and genes commonly implicated in GBM malignancy, such as MGMT, EGFR, PI3K-Akt signaling, and Ras/MAPK signaling are also upregulated in the PVN model. We describe the kinetics of gene expression within the PVN hydrogels over a course of 14 days, observing the patterns associated with tumor cell-mediated endothelial network co-option and regression. We finally examine the effect of temozolomide, a frontline chemotherapy used clinically against GBM, on the PVN culture. Notably, the PVN model is less responsive to TMZ compared to hydrogels containing only tumor cells. Overall, these results demonstrate that inclusion of cellular and matrix-associated elements of the PVN within an in vitro model of GBM allows for the development of gene expression patterns and therapeutic response relevant to GBM.
Collapse
Affiliation(s)
- Mai T Ngo
- Dept. Chemical and Biomolecular Engineering, University of Illinois at Urbana-Champaign, Urbana, IL, 61801, USA
| | - Brendan A C Harley
- Dept. Chemical and Biomolecular Engineering, University of Illinois at Urbana-Champaign, Urbana, IL, 61801, USA; Carl R. Woese Institute for Genomic Biology, University of Illinois at Urbana-Champaign, Urbana, IL, 61801, USA.
| |
Collapse
|
47
|
He Z, You C, Zhao D. Long non-coding RNA UCA1/miR-182/PFKFB2 axis modulates glioblastoma-associated stromal cells-mediated glycolysis and invasion of glioma cells. Biochem Biophys Res Commun 2018; 500:569-576. [DOI: 10.1016/j.bbrc.2018.04.091] [Citation(s) in RCA: 49] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2018] [Accepted: 04/12/2018] [Indexed: 02/09/2023]
|
48
|
Chen JWE, Lumibao J, Blazek A, Gaskins HR, Harley B. Hypoxia activates enhanced invasive potential and endogenous hyaluronic acid production by glioblastoma cells. Biomater Sci 2018; 6:854-862. [PMID: 29485655 PMCID: PMC5869158 DOI: 10.1039/c7bm01195d] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
Glioblastoma (GBM) is the most common, aggressive, and deadly form of adult brain cancer, and is associated with a short survival rate (median 12-15 months, 5+ year less than 5%). The complex tumor microenvironment includes matrix transitions at the tumor margin, such as gradations in hyaluronic acid (HA). In addition, metabolic stress induced by decreased oxygen content across the tumor may contribute to tumor progression. However, cross-talk between matrix composition and metabolic stress remains unclear. In this study, we fabricated an in vitro brain memetic HA-decorated gelatin hydrogel platform incorporating variable oxygen concentrations to mimic intra-tumoral hypoxia. We observed that EGFR status (wildtype vs. a constitutively active EGFRvIII mutant) of U87 GBM cells affected proliferation and metabolic activity in response to hypoxia and matrix-bound HA. The use of an invasion assay revealed that invasion was significantly enhanced in both cell types under hypoxia. Moreover, we observed compensatory secretion of soluble HA in cases of enhanced GBM cell invasion, consistent with our previous findings using other GBM cell lines. Interestingly, U87 GBM cells adapted to hypoxia by shifting toward a more anaerobic metabolic state, a mechanism that may contribute to GBM cell invasion. Collectively, these data demonstrate that the use of a three-dimensional hydrogel provides a robust method to study the impact of matrix composition and metabolic challenges on GBM cell invasion, a key factor contributing to the most common, aggressive, and deadly form of adult brain cancer.
Collapse
Affiliation(s)
- Jee-Wei Emily Chen
- Dept. of Chemical and Biomolecular Engineering, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA.
| | | | | | | | | |
Collapse
|
49
|
Vieira de Castro J, Gomes ED, Granja S, Anjo SI, Baltazar F, Manadas B, Salgado AJ, Costa BM. Impact of mesenchymal stem cells' secretome on glioblastoma pathophysiology. J Transl Med 2017; 15:200. [PMID: 28969635 PMCID: PMC5625623 DOI: 10.1186/s12967-017-1303-8] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2017] [Accepted: 09/19/2017] [Indexed: 12/19/2022] Open
Abstract
Background Glioblastoma (GBM) is a highly aggressive primary brain cancer, for which curative therapies are not available. An emerging therapeutic approach suggested to have potential to target malignant gliomas has been based on the use of multipotent mesenchymal stem cells (MSCs), either unmodified or engineered to deliver anticancer therapeutic agents, as these cells present an intrinsic capacity to migrate towards malignant tumors. Nevertheless, it is still controversial whether this innate tropism of MSCs towards the tumor area is associated with cancer promotion or suppression. Considering that one of the major mechanisms by which MSCs interact with and modulate tumor cells is via secreted factors, we studied how the secretome of MSCs modulates critical hallmark features of GBM cells. Methods The effect of conditioned media (CM) from human umbilical cord perivascular cells (HUCPVCs, a MSC population present in the Wharton’s jelly of the umbilical cord) on GBM cell viability, migration, proliferation and sensitivity to temozolomide treatment of U251 and SNB-19 GBM cells was evaluated. The in vivo chicken chorioallantoic membrane (CAM) assay was used to evaluate the effect of HUCPVCs CM on tumor growth and angiogenesis. The secretome of HUCPVCs was characterized by proteomic analyses. Results We found that both tested GBM cell lines exposed to HUCPVCs CM presented significantly higher cellular viability, proliferation and migration. In contrast, resistance of GBM cells to temozolomide chemotherapy was not significantly affected by HUCPVCs CM. In the in vivo CAM assay, CM from HUCPVCs promoted U251 and SNB-19 tumor cells growth. Proteomic analysis to characterize the secretome of HUCPVCs identified several proteins involved in promotion of cell survival, proliferation and migration, revealing novel putative molecular mediators for the effects observed in GBM cells exposed to HUCPVCs CM. Conclusions These findings provide novel insights to better understand the interplay between GBM cells and MSCs, raising awareness to potential safety issues regarding the use of MSCs as stem-cell based therapies for GBM. Electronic supplementary material The online version of this article (doi:10.1186/s12967-017-1303-8) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Joana Vieira de Castro
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Campus de Gualtar, 4710-057, Braga, Portugal.,ICVS/3B's-PT Government Associate Laboratory, University of Minho, Braga/Guimarães, Campus de Gualtar, 4710-057, Braga, Portugal
| | - Eduardo D Gomes
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Campus de Gualtar, 4710-057, Braga, Portugal.,ICVS/3B's-PT Government Associate Laboratory, University of Minho, Braga/Guimarães, Campus de Gualtar, 4710-057, Braga, Portugal
| | - Sara Granja
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Campus de Gualtar, 4710-057, Braga, Portugal.,ICVS/3B's-PT Government Associate Laboratory, University of Minho, Braga/Guimarães, Campus de Gualtar, 4710-057, Braga, Portugal
| | - Sandra I Anjo
- CNC-Center for Neuroscience and Cell Biology, University of Coimbra, 3004-517, Coimbra, Portugal.,Department of Life Sciences, Faculty of Sciences and Technology, University of Coimbra, 3004-517, Coimbra, Portugal
| | - Fátima Baltazar
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Campus de Gualtar, 4710-057, Braga, Portugal.,ICVS/3B's-PT Government Associate Laboratory, University of Minho, Braga/Guimarães, Campus de Gualtar, 4710-057, Braga, Portugal
| | - Bruno Manadas
- CNC-Center for Neuroscience and Cell Biology, University of Coimbra, 3004-517, Coimbra, Portugal
| | - António J Salgado
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Campus de Gualtar, 4710-057, Braga, Portugal.,ICVS/3B's-PT Government Associate Laboratory, University of Minho, Braga/Guimarães, Campus de Gualtar, 4710-057, Braga, Portugal
| | - Bruno M Costa
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Campus de Gualtar, 4710-057, Braga, Portugal. .,ICVS/3B's-PT Government Associate Laboratory, University of Minho, Braga/Guimarães, Campus de Gualtar, 4710-057, Braga, Portugal.
| |
Collapse
|
50
|
Clavreul A, Pourbaghi-Masouleh M, Roger E, Lautram N, Montero-Menei CN, Menei P. Human mesenchymal stromal cells as cellular drug-delivery vectors for glioblastoma therapy: a good deal? JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2017; 36:135. [PMID: 28962658 PMCID: PMC5622550 DOI: 10.1186/s13046-017-0605-2] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/20/2017] [Accepted: 09/21/2017] [Indexed: 02/06/2023]
Abstract
Background Glioblastoma (GB) is the most malignant brain tumor in adults. It is characterized by angiogenesis and a high proliferative and invasive capacity. Standard therapy (surgery, radiotherapy and chemotherapy with temozolomide) is of limited efficacy. Innovative anticancer drugs targeting both tumor cells and angiogenesis are urgently required, together with effective systems for their delivery to the brain. We assessed the ability of human mesenchymal stromal cells (MSCs) to uptake the multikinase inhibitor, sorafenib (SFN), and to carry this drug to a brain tumor following intranasal administration. Method MSCs were primed with SFN and drug content and release were quantified by analytical chemistry techniques. The ability of SFN-primed MSCs to inhibit the survival of the human U87MG GB cell line and endothelial cells was assessed in in vitro assays. These cells were then administered intranasally to nude mice bearing intracerebral U87MG xenografts. Their effect on tumor growth and angiogenesis was evaluated by magnetic resonance imaging and immunofluorescence analyses, and was compared with the intranasal administration of unprimed MSCs or SFN alone. Results MSCs took up about 9 pg SFN per cell, with no effect on viability, and were able to release 60% of the primed drug. The cytostatic activity of the released SFN was entirely conserved, resulting in a significant inhibition of U87MG and endothelial cell survival in vitro. Two intranasal administrations of SFN-primed MSCs in U87MG-bearing mice resulted in lower levels of tumor angiogenesis than the injection of unprimed MSCs or SFN alone, but had no effect on tumor volume. We also observed an increase in the proportion of small intratumoral vessels in animals treated with unprimed MSCs; this effect being abolished if the MSCs were primed with SFN. Conclusion We show the potential of MSCs to carry SFN to brain tumors following an intranasal administration. However, the therapeutic effect is modest probably due to the pro-tumorigenic properties of MSCs, which may limit the action of the released SFN. This calls into question the suitability of MSCs for use in GB therapy and renders it necessary to find methods guaranteeing the safety of this cellular vector after drug delivery.
Collapse
Affiliation(s)
- Anne Clavreul
- Département de Neurochirurgie, CHU, Angers, France. .,CRCINA, INSERM, Université de Nantes, Université d'Angers, Angers, France.
| | - Milad Pourbaghi-Masouleh
- CRCINA, INSERM, Université de Nantes, Université d'Angers, Angers, France.,Division of Drug Delivery and Tissue Engineering, School of Pharmacy, University of Nottingham, Nottingham, UK
| | - Emilie Roger
- MINT, UNIV Angers, INSERM 1066, CNRS 6021, Université Bretagne Loire, Angers, France
| | - Nolwenn Lautram
- MINT, UNIV Angers, INSERM 1066, CNRS 6021, Université Bretagne Loire, Angers, France
| | | | - Philippe Menei
- Département de Neurochirurgie, CHU, Angers, France.,CRCINA, INSERM, Université de Nantes, Université d'Angers, Angers, France
| |
Collapse
|