1
|
Saadh MJ, Allela OQB, Kareem RA, Sanghvi G, Menon SV, Sharma P, Tomar BS, Sharma A, Sameer HN, Hamad AK, Athab ZH, Adil M. From Gut to Brain: The Impact of Short-Chain Fatty Acids on Brain Cancer. Neuromolecular Med 2025; 27:10. [PMID: 39821841 DOI: 10.1007/s12017-025-08830-8] [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: 12/17/2024] [Accepted: 01/06/2025] [Indexed: 01/19/2025]
Abstract
The primary source of short-chain fatty acids (SCFAs), now recognized as critical mediators of host health, particularly in the context of neurobiology and cancer development, is the gut microbiota's fermentation of dietary fibers. Recent research highlights the complex influence of SCFAs, such as acetate, propionate, and butyrate, on brain cancer progression. These SCFAs impact immune modulation and the tumor microenvironment, particularly in brain tumors like glioma. They play a critical role in regulating cellular processes, including apoptosis, cell differentiation, and inflammation. Moreover, studies have linked SCFAs to maintaining the integrity of the blood-brain barrier (BBB), suggesting a protective role in preventing tumor infiltration and enhancing anti-tumor immunity. As our understanding of the gut-brain axis deepens, it becomes increasingly important to investigate SCFAs' therapeutic potential in brain cancer management. Looking into how SCFAs affect brain tumor cells and the environment around them could lead to new ways to prevent and treat these diseases, which could lead to better outcomes for people who are dealing with these challenging cancers.
Collapse
Affiliation(s)
- Mohamed J Saadh
- Faculty of Pharmacy, Middle East University, Amman, 11831, Jordan.
| | | | | | - Gaurav Sanghvi
- Department of Microbiology, Faculty of Science, Marwadi University Research Center, Marwadi University, Rajkot, Gujarat, 360003, India
| | - Soumya V Menon
- Department of Chemistry and Biochemistry, School of Sciences, JAIN (Deemed to Be University), Bangalore, Karnataka, India
| | - Pawan Sharma
- Department of Sciences, Vivekananda Global University, Jaipur, Rajasthan, 303012, India
| | - Balvir S Tomar
- Institute of Pediatric Gastroenterology and Hepatology, National Institute of Medical Sciences, NIMS University Rajasthan, Jaipur, India
| | - Aanchal Sharma
- Department of Medical Lab Sciences, Chandigarh Group of Colleges-Jhanjeri, Mohali, Punjab, 140307, India
| | - Hayder Naji Sameer
- Collage of Pharmacy, National University of Science and Technology, Dhi Qar, 64001, Iraq
| | | | - Zainab H Athab
- Department of Pharmacy, Al-Zahrawi University College, Karbala, Iraq
| | - Mohaned Adil
- Pharmacy College, Al-Farahidi University, Baghdad, Iraq
| |
Collapse
|
2
|
Wang Z, Fang Z, Gui Y, Xi B, Xie Z. Elevated HSPB1 Expression Is Associated with a Poor Prognosis in Glioblastoma Multiforme Patients. J Neurol Surg A Cent Eur Neurosurg 2025; 86:17-29. [PMID: 38959943 DOI: 10.1055/s-0043-1777761] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/05/2024]
Abstract
BACKGROUND Glioblastoma multiforme (GBM) is a highly aggressive form of brain cancer. This study investigated the clinical predictive value of heat shock protein β1 (HSPB1) in patients with GBM. METHODS A correlation was established between HSPB1 expression and GBM progression using data from The Cancer Genome Atlas (TCGA) dataset, Chinese Glioma Genome Atlas dataset, Gene Expression Omnibus dataset, and Human Protein Atlas database. A survival analysis was conducted and an HSPB1-based nomogram was constructed to evaluate the prognostic value of HSPB1 in patients with GBM. RESULTS Based on TCGA data mining, we discovered that HSPB1 was significantly elevated in patients with GBM and may reflect their response to immunotherapy. In survival analysis, it appeared to have a predictive role in the prognosis of patients with GBM. Five signaling pathways were significantly enriched in the high HSPB1 expression phenotype according to the gene set enrichment analysis. In addition, a significant association was found between HSPB1 expression and immune checkpoints, tumor immune infiltration, tumor immune microenvironment, and immune cell markers in glioma. Overall, our results suggest that HSPB1 may regulate the function of immune cells, serve as a new immunotherapy target, and predict the response to immunotherapy in patients with GBM. CONCLUSION HSPB1 appears to serve as a potential predictor of the clinical prognosis and response to immunotherapy in patients with GBM. It may be possible to identify patients who are likely to benefit from immunotherapy by assessing the expression level of HSPB1.
Collapse
Affiliation(s)
- Zhihua Wang
- Department of Neurosurgery, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
| | - Zhaohua Fang
- Department of Neurosurgery, Chongren County People's Hospital, Fuzhou, Jiangxi, China
| | - Yongping Gui
- Department of Neurosurgery, Jiangxi Provincial People's Hospital, The First Affiliated Hospital of Nanchang Medical College, Nanchang, Jiangxi, China
- Department of Neurosurgery, Xiangya Hospital Jiangxi Hospital, Central South University, Nanchang, Jiangxi, China
| | - Bin Xi
- Department of Neurosurgery, Jiangxi Provincial People's Hospital, The First Affiliated Hospital of Nanchang Medical College, Nanchang, Jiangxi, China
- Department of Neurosurgery, Xiangya Hospital Jiangxi Hospital, Central South University, Nanchang, Jiangxi, China
| | - Zhiping Xie
- Department of Neurosurgery, Jiangxi Provincial People's Hospital, The First Affiliated Hospital of Nanchang Medical College, Nanchang, Jiangxi, China
- Department of Neurosurgery, Xiangya Hospital Jiangxi Hospital, Central South University, Nanchang, Jiangxi, China
| |
Collapse
|
3
|
Gupta P, Dang M, Oberai S, Migliozzi S, Trivedi R, Kumar G, Peshoff M, Milam N, Ahmed A, Bojja K, Tran TM, Gumin J, Kamiya-Matsuoka C, Huse J, Cox K, Li J, Shehwana H, Sheth SA, Saxon R, Baohua S, Parker Kerrigan B, Maheshwari A, Parra Cuentas ER, Navin NE, Heimberger AB, Lang FF, Iavarone A, Clise-Dwyer K, Wang L, Bhat KP. Immune landscape of isocitrate dehydrogenase-stratified primary and recurrent human gliomas. Neuro Oncol 2024; 26:2239-2255. [PMID: 39126294 PMCID: PMC11630528 DOI: 10.1093/neuonc/noae139] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2023] [Indexed: 08/12/2024] Open
Abstract
BACKGROUND Human gliomas are classified using isocitrate dehydrogenase (IDH) status as a prognosticator; however, the influence of genetic differences and treatment effects on ensuing immunity remains unclear. METHODS In this study, we used sequential single-cell transcriptomics on 144 678 and spectral cytometry on over 2 million immune cells encompassing 48 human gliomas to decipher their immune landscape. RESULTS We identified 22 distinct immune cell types that contribute to glioma immunity. Specifically, brain-resident microglia (MG) were reduced with a concomitant increase in CD8+ T lymphocytes during glioma recurrence independent of IDH status. In contrast, IDH-wild type-associated patterns, such as an abundance of antigen-presenting cell-like MG and cytotoxic CD8+ T cells, were observed. Beyond elucidating the differences in IDH, relapse, and treatment-associated immunity, we discovered novel inflammatory MG subpopulations expressing granulysin, a cytotoxic peptide that is otherwise expressed in lymphocytes only. Furthermore, we provide a robust genomic framework for defining macrophage polarization beyond M1/M2 paradigm and reference signatures of glioma-specific tumor immune microenvironment (termed GlioTIME-36) for deconvoluting transcriptomic datasets. CONCLUSIONS This study provides advanced optics of the human pan-glioma immune contexture as a valuable guide for translational and clinical applications.
Collapse
Affiliation(s)
- Pravesh Gupta
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Minghao Dang
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Shivangi Oberai
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Simona Migliozzi
- Department of Neurological Surgery, Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Miami, Florida, USA
| | - Rakesh Trivedi
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Gayatri Kumar
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Mekenzie Peshoff
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
- MD Anderson UTHealth Graduate School of Biomedical Sciences, Houston, Texas, USA
| | - Nancy Milam
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Aml Ahmed
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Krishna Bojja
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Tuan M Tran
- Department of Systems Biology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Joy Gumin
- Department of Neurosurgery, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Carlos Kamiya-Matsuoka
- Department of Neuro-Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Jason Huse
- Department of Pathology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Kathryn Cox
- Department of Hematopoietic Biology and Malignancy, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Jianzhuo Li
- Department of Systems Biology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Huma Shehwana
- Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Sameer A Sheth
- Department of Neurosurgery, Baylor College of Medicine, Houston, Texas, USA
| | - Rodriguez Saxon
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Sun Baohua
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Brittany Parker Kerrigan
- Department of Neurosurgery, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Atul Maheshwari
- Department of Neurology and Neuroscience, Baylor College of Medicine, Houston, Texas, USA
| | - Edwin Roger Parra Cuentas
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Nicholas E Navin
- Department of Systems Biology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
- MD Anderson UTHealth Graduate School of Biomedical Sciences, Houston, Texas, USA
| | - Amy B Heimberger
- Department of Neurosurgery, Northwestern University, Evanston, Illinois, USA
| | - Frederick F Lang
- Department of Neurosurgery, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Antonio Iavarone
- Department of Neurological Surgery, Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Miami, Florida, USA
| | - Karen Clise-Dwyer
- Department of Hematopoietic Biology and Malignancy, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Linghua Wang
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
- MD Anderson UTHealth Graduate School of Biomedical Sciences, Houston, Texas, USA
| | - Krishna P Bhat
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
- Department of Neurosurgery, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
- MD Anderson UTHealth Graduate School of Biomedical Sciences, Houston, Texas, USA
| |
Collapse
|
4
|
Wang L, He Z, Fan S, Mo L, Li Y, Yuan X, Xu B, Mou Y, Yin Y. Quantitative analysis of immune cells within the tumor microenvironment of glioblastoma and their relevance for prognosis. Int Immunopharmacol 2024; 142:113109. [PMID: 39255678 DOI: 10.1016/j.intimp.2024.113109] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2024] [Revised: 08/09/2024] [Accepted: 09/04/2024] [Indexed: 09/12/2024]
Abstract
Glioblastoma (GBM) is a high malignant tumor with no effective treatment. To comprehensively characterize the landscape of immune cells in GBM and evaluate their correlation with prognosis, we developed a multispectral fluorescent imaging pipeline that included tumor-infiltrating lymphocytic markers (CD3, CD4, CD8, FOXP3, NKP46), immune checkpoint markers (PD-1, PD-L1), and markers to characterize myeloid cells (CD68, CD66b, CD163, HLA-DR), to spatially quantify 18 immune cell subsets in 21 GBM cases. We found that macrophages are the most abundant in GBM microenvironment, followed by T cells and neutrophils, while NK and NKT cells are the least. Previously unreported CD8+ Treg, PD-L1+ neutrophils, and high proportion of PD-1+ NK and PD-1+ T cells were also detected. Single high densities of PD-1+CD8+ T cells, neutrophils, and PD-L1-expressing CD68+ cells were associated with longer survival. Moreover, closer proximity of T cells to PD-L1+ macrophages or PD-L1+ neutrophils were associated with poor prognosis. Correlative analysis revealed circulating PMN-MDSC and e-MDSC were positively correlated with intratumoral M2 macrophages, while circulating NK cells were inversely associated with infiltrating CD4+ Treg cells in GBM patients. Our findings highlighted the potential roles of infiltrating immune cells in prognosis prediction and developing novel immunotherapeutic strategies for GBM patients.
Collapse
Affiliation(s)
- Lu Wang
- Department of Immunology, School of Basic Medical Sciences, NHC Key Laboratory of Medical Immunology, Medicine Innovation Center for Fundamental Research on Major Immunology-related Diseases, Peking University, Beijing 100191, China
| | - Zhenqiang He
- Department of Neurosurgery/Neuro-oncology, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou 510060, China
| | - Shuning Fan
- Department of Immunology, School of Basic Medical Sciences, NHC Key Laboratory of Medical Immunology, Medicine Innovation Center for Fundamental Research on Major Immunology-related Diseases, Peking University, Beijing 100191, China
| | - Li Mo
- Department of Immunology, School of Basic Medical Sciences, NHC Key Laboratory of Medical Immunology, Medicine Innovation Center for Fundamental Research on Major Immunology-related Diseases, Peking University, Beijing 100191, China
| | - Yan Li
- Department of Immunology, School of Basic Medical Sciences, NHC Key Laboratory of Medical Immunology, Medicine Innovation Center for Fundamental Research on Major Immunology-related Diseases, Peking University, Beijing 100191, China
| | - Xia Yuan
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China
| | - Bo Xu
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China
| | - Yonggao Mou
- Department of Neurosurgery/Neuro-oncology, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou 510060, China.
| | - Yanhui Yin
- Department of Immunology, School of Basic Medical Sciences, NHC Key Laboratory of Medical Immunology, Medicine Innovation Center for Fundamental Research on Major Immunology-related Diseases, Peking University, Beijing 100191, China.
| |
Collapse
|
5
|
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
|
6
|
Mikolajewicz N, Tatari N, Wei J, Savage N, Granda Farias A, Dimitrov V, Chen D, Zador Z, Dasgupta K, Aguilera-Uribe M, Xiao YX, Lee SY, Mero P, McKenna D, Venugopal C, Brown KR, Han H, Singh S, Moffat J. Functional profiling of murine glioma models highlights targetable immune evasion phenotypes. Acta Neuropathol 2024; 148:74. [PMID: 39592459 PMCID: PMC11599368 DOI: 10.1007/s00401-024-02831-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2024] [Revised: 11/10/2024] [Accepted: 11/11/2024] [Indexed: 11/28/2024]
Abstract
Cancer-intrinsic immune evasion mechanisms and pleiotropy are a barrier to cancer immunotherapy. This is apparent in certain highly fatal cancers, including high-grade gliomas and glioblastomas (GBM). In this study, we evaluated two murine syngeneic glioma models (GL261 and CT2A) as preclinical models for human GBM using functional genetic screens, single-cell transcriptomics and machine learning approaches. Through CRISPR genome-wide co-culture killing screens with various immune cells (cytotoxic T cells, natural killer cells, and macrophages), we identified three key cancer-intrinsic evasion mechanisms: NFκB signaling, autophagy/endosome machinery, and chromatin remodeling. Additional fitness screens identified dependencies in murine gliomas that partially recapitulated those seen in human GBM (e.g., UFMylation). Our single-cell analyses showed that different glioma models exhibited distinct immune infiltration patterns and recapitulated key immune gene programs observed in human GBM, including hypoxia, interferon, and TNF signaling. Moreover, in vivo orthotopic tumor engraftment was associated with phenotypic shifts and changes in proliferative capacity, with murine tumors recapitulating the intratumoral heterogeneity observed in human GBM, exhibiting propensities for developmental- and mesenchymal-like phenotypes. Notably, we observed common transcription factors and cofactors shared with human GBM, including developmental (Nfia and Tcf4), mesenchymal (Prrx1 and Wwtr1), as well as cycling-associated genes (Bub3, Cenpa, Bard1, Brca1, and Mis18bp1). Perturbation of these genes led to reciprocal phenotypic shifts suggesting intrinsic feedback mechanisms that balance in vivo cellular states. Finally, we used a machine-learning approach to identify two distinct immune evasion gene programs, one of which represents a clinically-relevant phenotype and delineates a subpopulation of stem-like glioma cells that predict response to immune checkpoint inhibition in human patients. This comprehensive characterization helps bridge the gap between murine glioma models and human GBM, providing valuable insights for future therapeutic development.
Collapse
Affiliation(s)
- Nicholas Mikolajewicz
- Program in Genetics and Genome Biology, The Hospital for Sick Children, Toronto, Canada
| | - Nazanin Tatari
- Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, Canada
- Centre for Discovery in Cancer Research (CDCR), McMaster University, Hamilton, Canada
- Department of Biomedicine, University Hospital Basel, Basel, Switzerland
| | - Jiarun Wei
- Program in Genetics and Genome Biology, The Hospital for Sick Children, Toronto, Canada
- Department of Molecular Genetics, University of Toronto, Toronto, Canada
| | - Neil Savage
- Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, Canada
- Centre for Discovery in Cancer Research (CDCR), McMaster University, Hamilton, Canada
| | - Adrian Granda Farias
- Program in Genetics and Genome Biology, The Hospital for Sick Children, Toronto, Canada
- Department of Molecular Genetics, University of Toronto, Toronto, Canada
| | - Vassil Dimitrov
- Program in Genetics and Genome Biology, The Hospital for Sick Children, Toronto, Canada
| | - David Chen
- Program in Genetics and Genome Biology, The Hospital for Sick Children, Toronto, Canada
| | - Zsolt Zador
- Department of Surgery, Faculty of Health Sciences, McMaster University, Hamilton, Canada
| | - Kuheli Dasgupta
- Program in Genetics and Genome Biology, The Hospital for Sick Children, Toronto, Canada
- Department of Molecular Genetics, University of Toronto, Toronto, Canada
| | - Magali Aguilera-Uribe
- Program in Genetics and Genome Biology, The Hospital for Sick Children, Toronto, Canada
- Department of Molecular Genetics, University of Toronto, Toronto, Canada
| | - Yu-Xi Xiao
- Program in Genetics and Genome Biology, The Hospital for Sick Children, Toronto, Canada
- Department of Molecular Genetics, University of Toronto, Toronto, Canada
| | - Seon Yong Lee
- Program in Genetics and Genome Biology, The Hospital for Sick Children, Toronto, Canada
| | - Patricia Mero
- Program in Genetics and Genome Biology, The Hospital for Sick Children, Toronto, Canada
| | - Dillon McKenna
- Centre for Discovery in Cancer Research (CDCR), McMaster University, Hamilton, Canada
- Department of Surgery, Faculty of Health Sciences, McMaster University, Hamilton, Canada
| | - Chitra Venugopal
- Centre for Discovery in Cancer Research (CDCR), McMaster University, Hamilton, Canada
- Department of Surgery, Faculty of Health Sciences, McMaster University, Hamilton, Canada
| | - Kevin R Brown
- Program in Genetics and Genome Biology, The Hospital for Sick Children, Toronto, Canada
| | - Hong Han
- Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, Canada
- Centre for Discovery in Cancer Research (CDCR), McMaster University, Hamilton, Canada
| | - Sheila Singh
- Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, Canada.
- Centre for Discovery in Cancer Research (CDCR), McMaster University, Hamilton, Canada.
- Department of Surgery, Faculty of Health Sciences, McMaster University, Hamilton, Canada.
| | - Jason Moffat
- Program in Genetics and Genome Biology, The Hospital for Sick Children, Toronto, Canada.
- Department of Molecular Genetics, University of Toronto, Toronto, Canada.
- Institute for Biomedical Engineering, University of Toronto, Toronto, Canada.
| |
Collapse
|
7
|
Liu R, Wang X, Zhou M, Zhai J, Sun J. PSF-lncRNA interaction as a target for novel targeted anticancer therapies. Biomed Pharmacother 2024; 180:117491. [PMID: 39332189 DOI: 10.1016/j.biopha.2024.117491] [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: 05/19/2024] [Revised: 09/15/2024] [Accepted: 09/20/2024] [Indexed: 09/29/2024] Open
Abstract
The Polypyrimidine Tract-Binding Protein-Associated Splicing Factor (PSF), a component of the Drosophila Behavior/Human Splicing (DBHS) complex, plays a pivotal role in cancer pathogenesis. The epigenetic regulation mediated by PSF and long noncoding RNA (lncRNA), along with PSF's alternative splicing activity, has been implicated in promoting cancer cell proliferation, migration, invasion, metastasis, and drug resistance in various human cancers. Recent research highlights the therapeutic promise of targeting the PSF-lncRNA interaction to combat aggressive malignancies, making it a compelling target for cancer therapy. This review offers a detailed synthesis of the current understanding of PSF's role in oncogenic pathways and recent progress in identifying inhibitors of PSF-lncRNA interactions. Furthermore, it discusses the potential of using these inhibitors in cancer treatment strategies, especially as adjuncts to immune checkpoint blockade therapies to improve the efficacy of anti-PD-(L)1 treatments in Glioblastoma Multiforme (GBM). By outlining the interaction patterns of existing PSF-lncRNA inhibitors, this article aims to guide the development and refinement of future pharmacological interventions.
Collapse
Affiliation(s)
- Ren Liu
- School of Pharmacy and Pharmaceutical Sciences & Institute of Materia Medica, Shandong First Medical University & Shandong Academy of Medical Sciences, NHC Key Laboratory of Biotechnology Drugs (Shandong Academy of Medical Sciences), Key Lab for Rare & Uncommon Diseases of Shandong Province, Jinan, Shandong 250117, China
| | - Xiaojing Wang
- School of Pharmacy and Pharmaceutical Sciences & Institute of Materia Medica, Shandong First Medical University & Shandong Academy of Medical Sciences, NHC Key Laboratory of Biotechnology Drugs (Shandong Academy of Medical Sciences), Key Lab for Rare & Uncommon Diseases of Shandong Province, Jinan, Shandong 250117, China
| | - Min Zhou
- School of Pharmacy and Pharmaceutical Sciences & Institute of Materia Medica, Shandong First Medical University & Shandong Academy of Medical Sciences, NHC Key Laboratory of Biotechnology Drugs (Shandong Academy of Medical Sciences), Key Lab for Rare & Uncommon Diseases of Shandong Province, Jinan, Shandong 250117, China
| | - Jingfang Zhai
- School of Pharmacy and Pharmaceutical Sciences & Institute of Materia Medica, Shandong First Medical University & Shandong Academy of Medical Sciences, NHC Key Laboratory of Biotechnology Drugs (Shandong Academy of Medical Sciences), Key Lab for Rare & Uncommon Diseases of Shandong Province, Jinan, Shandong 250117, China
| | - Jie Sun
- School of Pharmacy and Pharmaceutical Sciences & Institute of Materia Medica, Shandong First Medical University & Shandong Academy of Medical Sciences, NHC Key Laboratory of Biotechnology Drugs (Shandong Academy of Medical Sciences), Key Lab for Rare & Uncommon Diseases of Shandong Province, Jinan, Shandong 250117, China.
| |
Collapse
|
8
|
Ahmady F, Curpen P, Perriman L, Fonseca Teixeira A, Wu S, Zhu HJ, Poddar A, Jayachandran A, Kannourakis G, Luwor RB. Reduced T and NK Cell Activity in Glioblastoma Patients Correlates with TIM-3 and BAT3 Dysregulation. Cells 2024; 13:1777. [PMID: 39513882 PMCID: PMC11545661 DOI: 10.3390/cells13211777] [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: 08/22/2024] [Revised: 10/17/2024] [Accepted: 10/22/2024] [Indexed: 11/16/2024] Open
Abstract
Inhibitory receptors are critical for regulating immune cell function. In cancer, these receptors are often over-expressed on the cell surface of T and NK cells, leading to reduced anti-tumor activity. Here, through the analysis of 11 commonly studied checkpoint and inhibitory receptors, we discern that only HAVCR2 (TIM3) and ENTPD1 (CD39) display significantly greater gene expression in glioblastoma compared to normal brain and lower grade glioma. Cell surface TIM-3, but not ENTPD1, was also elevated on activated CD4+ and CD8+ T cells, as well as on NK cells from glioblastoma patients compared to healthy donor T and NK cells. A subsequent analysis of molecules known to co-ordinate TIM-3 function and regulation was performed, which revealed that BAT3 expression was significantly reduced in CD4+ and CD8+ T cells, as well as NK cells from glioblastoma patients compared to counterparts from healthy donors. These pro-inhibitory changes are also correlated with reduced levels of the activation marker CD69 and the pro-inflammatory cytokine IFNγ in CD4+ and CD8+ T cells, as well as NK cells from glioblastoma patients. Collectively, these data reveal that glioblastoma-mediated CD4+ and CD8+ T cell and NK cell suppression is due, at least in part, to dysregulated TIM-3 and BAT3 expression and the associated downstream immunoregulatory and dysfunctional effects.
Collapse
Affiliation(s)
- Farah Ahmady
- Fiona Elsey Cancer Research Institute, Ballarat, VIC 3350, Australia; (F.A.); (L.P.); (A.P.); (A.J.); (G.K.)
- Federation University, Ballarat, VIC 3350, Australia
| | - Peter Curpen
- Townsville Hospital and Health Service, James Cook University, Townsville, QLD 4814, Australia;
| | - Louis Perriman
- Fiona Elsey Cancer Research Institute, Ballarat, VIC 3350, Australia; (F.A.); (L.P.); (A.P.); (A.J.); (G.K.)
- Federation University, Ballarat, VIC 3350, Australia
- Murdoch Children’s Research Institute, Parkville, VIC 3052, Australia
| | - Adilson Fonseca Teixeira
- Department of Surgery, The Royal Melbourne Hospital, The University of Melbourne, Parkville, VIC 3050, Australia; (A.F.T.); (S.W.); (H.-J.Z.)
- Huagene Institute, Kecheng Science and Technology Park, Pukou District, Nanjing 211806, China
| | - Siqi Wu
- Department of Surgery, The Royal Melbourne Hospital, The University of Melbourne, Parkville, VIC 3050, Australia; (A.F.T.); (S.W.); (H.-J.Z.)
- Huagene Institute, Kecheng Science and Technology Park, Pukou District, Nanjing 211806, China
| | - Hong-Jian Zhu
- Department of Surgery, The Royal Melbourne Hospital, The University of Melbourne, Parkville, VIC 3050, Australia; (A.F.T.); (S.W.); (H.-J.Z.)
- Huagene Institute, Kecheng Science and Technology Park, Pukou District, Nanjing 211806, China
| | - Arpita Poddar
- Fiona Elsey Cancer Research Institute, Ballarat, VIC 3350, Australia; (F.A.); (L.P.); (A.P.); (A.J.); (G.K.)
- Federation University, Ballarat, VIC 3350, Australia
| | - Aparna Jayachandran
- Fiona Elsey Cancer Research Institute, Ballarat, VIC 3350, Australia; (F.A.); (L.P.); (A.P.); (A.J.); (G.K.)
- Federation University, Ballarat, VIC 3350, Australia
| | - George Kannourakis
- Fiona Elsey Cancer Research Institute, Ballarat, VIC 3350, Australia; (F.A.); (L.P.); (A.P.); (A.J.); (G.K.)
- Federation University, Ballarat, VIC 3350, Australia
| | - Rodney B. Luwor
- Fiona Elsey Cancer Research Institute, Ballarat, VIC 3350, Australia; (F.A.); (L.P.); (A.P.); (A.J.); (G.K.)
- Federation University, Ballarat, VIC 3350, Australia
- Department of Surgery, The Royal Melbourne Hospital, The University of Melbourne, Parkville, VIC 3050, Australia; (A.F.T.); (S.W.); (H.-J.Z.)
- Huagene Institute, Kecheng Science and Technology Park, Pukou District, Nanjing 211806, China
| |
Collapse
|
9
|
Ji H, Lan Y, Xing P, Wang Z, Zhong X, Tang W, Wei Q, Chen H, Liu B, Guo H. IL-18, a therapeutic target for immunotherapy boosting, promotes temozolomide chemoresistance via the PI3K/AKT pathway in glioma. J Transl Med 2024; 22:951. [PMID: 39434175 PMCID: PMC11492732 DOI: 10.1186/s12967-024-05755-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: 04/04/2024] [Accepted: 10/08/2024] [Indexed: 10/23/2024] Open
Abstract
Interleukin-18, a member of the interleukin - 1 family of cytokines, is upregulated in glioma. However, its effects on glioma remain unclear. This study aimed to explore the role and underlying mechanisms of interleukin-18 expression in glioma. Here, we demonstrated that interleukin-18 enhanced resistance to temozolomide by increasing proliferation and inhibiting apoptosis in cultured glioma cells. Further in vivo studies revealed that interleukin-18 promoted temozolomide resistance in BALB/c nude mice bearing tumor. Mechanical exploration indicated that interleukin-18 stimulation could activate the PI3K/AKT signaling pathway in glioma cells, and PI3K inhibition could reduce the temozolomide resistance promoted by interleukin-18. We found that interleukin-18 upregulated CD274 expression in glioma, revealing its potential effects on the microenvironment. Furthermore, we established a tumor xenograft model and explored the therapeutic efficacy of anti-interleukin-18 monoclonal antibody. Targeting interleukin-18 prolonged survival and attenuated CD274 expression in the mice bearing tumor. Combined treatment with anti-interleukin-18 and anti-PD-1 monoclonal antibody showed better efficacy in suppressing tumor growth than either treatment alone in mice bearing tumor. Collectively, these data present that interleukin-18 promotes temozolomide chemoresistance in glioma cells via PI3K/Akt activation and establishes an immunosuppressive milieu by modulating CD274. This study highlights the therapeutic value of interleukin-18 in glioma.
Collapse
Affiliation(s)
- Huangyi Ji
- Department of Neurosurgery, The National Key Clinical Specialty, The Engineering Technology Research Center of Education Ministry of China on Diagnosis and Treatment of Cerebrovascular Disease, Guangdong Provincial Key Laboratory on Brain Function Repair and Regeneration, The Neurosurgery Institute of Guangdong Province, Zhujiang Hospital, Southern Medical University, Guangzhou, 510282, China
- Department of Neurosurgery, The First Affiliated Hospital of Shantou University Medical College, Shantou, 515041, China
| | - Yufei Lan
- Department of Neurosurgery, The National Key Clinical Specialty, The Engineering Technology Research Center of Education Ministry of China on Diagnosis and Treatment of Cerebrovascular Disease, Guangdong Provincial Key Laboratory on Brain Function Repair and Regeneration, The Neurosurgery Institute of Guangdong Province, Zhujiang Hospital, Southern Medical University, Guangzhou, 510282, China
| | - Pengpeng Xing
- ZhiXin High School, No. 152, ZhiXin South Road, Yuexiu District, Guangzhou, 510080, China
| | - Zhao Wang
- Department of Neurosurgery, The National Key Clinical Specialty, The Engineering Technology Research Center of Education Ministry of China on Diagnosis and Treatment of Cerebrovascular Disease, Guangdong Provincial Key Laboratory on Brain Function Repair and Regeneration, The Neurosurgery Institute of Guangdong Province, Zhujiang Hospital, Southern Medical University, Guangzhou, 510282, China
| | - Xiangyang Zhong
- Department of Neurosurgery, The National Key Clinical Specialty, The Engineering Technology Research Center of Education Ministry of China on Diagnosis and Treatment of Cerebrovascular Disease, Guangdong Provincial Key Laboratory on Brain Function Repair and Regeneration, The Neurosurgery Institute of Guangdong Province, Zhujiang Hospital, Southern Medical University, Guangzhou, 510282, China
| | - Wenhui Tang
- Department of Neurosurgery, The National Key Clinical Specialty, The Engineering Technology Research Center of Education Ministry of China on Diagnosis and Treatment of Cerebrovascular Disease, Guangdong Provincial Key Laboratory on Brain Function Repair and Regeneration, The Neurosurgery Institute of Guangdong Province, Zhujiang Hospital, Southern Medical University, Guangzhou, 510282, China
| | - Quantang Wei
- Department of Neurosurgery, The First Affiliated Hospital of Shantou University Medical College, Shantou, 515041, China
| | - Hongbin Chen
- The Second Clinical School, Southern Medical University, Guangzhou, 510515, China
| | - Boyang Liu
- Department of Neurosurgery, Department of Neuro-Oncological Surgery, The National Key Clinical Specialty, The Engineering Technology Research Center of Education Ministry of China on Diagnosis and Treatment of Cerebrovascular Disease, Guangdong Provincial Key Laboratory on Brain Function Repair and Regeneration, The Neurosurgery Institute of Guangdong Province, Zhujiang Hospital, Southern Medical University, Guangzhou, 510282, China.
| | - Hongbo Guo
- Department of Neurosurgery, The National Key Clinical Specialty, The Engineering Technology Research Center of Education Ministry of China on Diagnosis and Treatment of Cerebrovascular Disease, Guangdong Provincial Key Laboratory on Brain Function Repair and Regeneration, The Neurosurgery Institute of Guangdong Province, Zhujiang Hospital, Southern Medical University, Guangzhou, 510282, China.
| |
Collapse
|
10
|
Haseeb M, Khan I, Kartal Z, Mahfooz S, Hatiboglu MA. Status Quo in the Liposome-Based Therapeutic Strategies Against Glioblastoma: "Targeting the Tumor and Tumor Microenvironment". Int J Mol Sci 2024; 25:11271. [PMID: 39457052 PMCID: PMC11509082 DOI: 10.3390/ijms252011271] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2024] [Revised: 10/06/2024] [Accepted: 10/16/2024] [Indexed: 10/28/2024] Open
Abstract
Glioblastoma is the most aggressive and fatal brain cancer, characterized by a high growth rate, invasiveness, and treatment resistance. The presence of the blood-brain barrier (BBB) and blood-brain tumor barrier (BBTB) poses a challenging task for chemotherapeutics, resulting in low efficacy, bioavailability, and increased dose-associated side effects. Despite the rigorous treatment strategies, including surgical resection, radiotherapy, and adjuvant chemotherapy with temozolomide, overall survival remains poor. The failure of current chemotherapeutics and other treatment regimens in glioblastoma necessitates the development of new drug delivery methodologies to precisely and efficiently target glioblastoma. Nanoparticle-based drug delivery systems offer a better therapeutic option in glioblastoma, considering their small size, ease of diffusion, and ability to cross the BBB. Liposomes are a specific category of nanoparticles made up of fatty acids. Furthermore, liposomes can be surface-modified to target a particular receptor and are nontoxic. This review discusses various methods of liposome modification for active/directed targeting and various liposome-based therapeutic approaches in the delivery of current chemotherapeutic drugs and nucleic acids in targeting the glioblastoma and tumor microenvironment.
Collapse
Affiliation(s)
- Mohd Haseeb
- Department of Molecular Biology, Beykoz Institute of Life Sciences and Biotechnology, Bezmialem Vakif University, Yalıköy St., Beykoz, 34820 Istanbul, Turkey; (M.H.); (S.M.)
| | - Imran Khan
- Department of Molecular Biology, Beykoz Institute of Life Sciences and Biotechnology, Bezmialem Vakif University, Yalıköy St., Beykoz, 34820 Istanbul, Turkey; (M.H.); (S.M.)
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Zeynep Kartal
- Department of Molecular Biology, Beykoz Institute of Life Sciences and Biotechnology, Bezmialem Vakif University, Yalıköy St., Beykoz, 34820 Istanbul, Turkey; (M.H.); (S.M.)
| | - Sadaf Mahfooz
- Department of Molecular Biology, Beykoz Institute of Life Sciences and Biotechnology, Bezmialem Vakif University, Yalıköy St., Beykoz, 34820 Istanbul, Turkey; (M.H.); (S.M.)
- Department of Radiation Oncology, College of Medicine, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Mustafa Aziz Hatiboglu
- Department of Molecular Biology, Beykoz Institute of Life Sciences and Biotechnology, Bezmialem Vakif University, Yalıköy St., Beykoz, 34820 Istanbul, Turkey; (M.H.); (S.M.)
- Department of Neurosurgery, Bezmialem Vakif University Medical School, Vatan Street, Fatih, 34093 Istanbul, Turkey
| |
Collapse
|
11
|
Ainslie K. Modifying Post-Surgical Immunity: Controlled Release of TLR7/8 Agonist for Immune Mediated Clearance of Glioblastoma. RESEARCH SQUARE 2024:rs.3.rs-5024510. [PMID: 39399681 PMCID: PMC11469459 DOI: 10.21203/rs.3.rs-5024510/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/15/2024]
Abstract
Glioblastoma is an aggressive brain cancer with a dismal prognosis despite current therapeutic interventions. Tumor resection is standard-of-care for glioblastoma and has profound immunostimulatory effects. Resulting in a nadir in tumor burden, resection offers a unique opportunity to break local immune tolerance and mount an effective anti-tumor immune response. Here, we explore the effect of local and controlled release of TLR7/8 agonist from a polymer scaffold implanted at the time of tumor resection. We find that sustained release of TLR7/8 agonist leads to clearance of residual post-resection tumor, improved survival, and subsequent protection from tumor challenge in mice bearing orthotopic GL261 or CT2A gliomas. We show that scaffold therapy boosts resection-mediated disruption to the tumor microenvironment, leading to an early inflammatory innate immune response both in the brain and cervical lymph node. This is followed by an influx of activated NK cells in the brain and effector T cells in the lymph node and brain. In sum, sustained local TLR7/8 agonism within the context of tumor resection is a promising approach for glioblastoma.
Collapse
|
12
|
Wang S, Wang Z, Liu Z, Wu J. Prognostic value of four immune-related genes in lower-grade gliomas: a biomarker discovery study. Front Genet 2024; 15:1403587. [PMID: 39192888 PMCID: PMC11347950 DOI: 10.3389/fgene.2024.1403587] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2024] [Accepted: 07/30/2024] [Indexed: 08/29/2024] Open
Abstract
Introduction The tumor microenvironment and IRGs are highly correlated with tumor occurrence, progression, and prognosis. However, their roles in grade II and III gliomas, termed LGGs in this study, remain to be fully elucidated. Our research aims to develop immune-related features for risk stratification and prognosis prediction in LGG. Methods Using the ssGSEA method, we assessed the immune characteristics of the LGG population. We conducted differential analysis using LGG samples from the TCGA database and normal samples from GTEx, identifying 412 differentially expressed immune-related genes (DEIRGs). Subsequently, we utilized univariate Cox, LASSO, and multivariate Cox regression analyses to establish both a gene predictive model and a nomogram predictive model. Results Here, we found that the ESTIMATE score, immune score and stromal score of high-immunity, high-grade and isocitrate dehydrogenase (IDH) wild-type glioma were higher than those of the corresponding group, and the tumor purity was lower. Higher ESTIMATE scores, stromal scores and immune scores indicated a poor prognosis in patients with LGG. Our four-gene prognostic model demonstrated superior accuracy compared to other molecular features. Validation using the CGGA as a testing set and the combined TCGA and CGGA cohort confirmed its robust prognostic value. Additionally, a nomogram integrating the prognostic model and clinical variables showed enhanced predictive capability. Discussion Our study highlights the prognostic significance of the identified four DEIRGs (KLRC3, MR1, PDIA2, and RFXAP) in LGG patients. The predictive model and nomogram developed herein offer valuable tools for personalized treatment strategies in LGG. Future research should focus on further validating these findings and exploring the functional roles of these DEIRGs within the LGG tumor microenvironment.
Collapse
Affiliation(s)
- Shuowen Wang
- Capital Institute of Pediatrics, Beijing, China
- Beijing Tongren Hospital, Capital Medical University, Beijing, China
| | - Zijun Wang
- Beijing Shijitan Hospital, Capital Medical University, Beijing, China
| | - Zhuo Liu
- Beijing Tongren Hospital, Capital Medical University, Beijing, China
| | - Jianxin Wu
- Capital Institute of Pediatrics, Beijing, China
- Beijing Tongren Hospital, Capital Medical University, Beijing, China
| |
Collapse
|
13
|
Sherman JH, Bobak A, Arsiwala T, Lockman P, Aulakh S. Targeting drug resistance in glioblastoma (Review). Int J Oncol 2024; 65:80. [PMID: 38994761 PMCID: PMC11251740 DOI: 10.3892/ijo.2024.5668] [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: 05/21/2022] [Accepted: 05/16/2024] [Indexed: 07/13/2024] Open
Abstract
Glioblastoma (GBM) is the most common malignancy of the central nervous system in adults. The current standard of care includes surgery, radiation therapy, temozolomide; and tumor‑treating fields leads to dismal overall survival. There are far limited treatments upon recurrence. Therapies to date are ineffective as a result of several factors, including the presence of the blood‑brain barrier, blood tumor barrier, glioma stem‑like cells and genetic heterogeneity in GBM. In the present review, the potential mechanisms that lead to treatment resistance in GBM and the measures which have been taken so far to attempt to overcome the resistance were discussed. The complex biology of GBM and lack of comprehensive understanding of the development of therapeutic resistance in GBM demands discovery of novel antigens that are targetable and provide effective therapeutic strategies.
Collapse
Affiliation(s)
- Jonathan H. Sherman
- Department of Neurosurgery, Rockefeller Neuroscience Institute, West Virginia University, Martinsburg, WV 25401, USA
| | - Adam Bobak
- Department of Biology, Seton Hill University, Greensburg, PA 15601, USA
| | - Tasneem Arsiwala
- Department of Basic Pharmaceutical Sciences, School of Pharmacy, West Virginia University, Morgantown, WV 26506, USA
| | - Paul Lockman
- Department of Basic Pharmaceutical Sciences, School of Pharmacy, West Virginia University, Morgantown, WV 26506, USA
| | - Sonikpreet Aulakh
- Section of Hematology/Oncology, Department of Internal Medicine, West Virginia University, Morgantown, WV 26506, USA
- Department of Neuroscience, West Virginia University, Morgantown, WV 26505, USA
| |
Collapse
|
14
|
Weiss A, D'Amata C, Pearson BJ, Hayes MN. A syngeneic spontaneous zebrafish model of tp53-deficient, EGFR vIII, and PI3KCA H1047R-driven glioblastoma reveals inhibitory roles for inflammation during tumor initiation and relapse in vivo. eLife 2024; 13:RP93077. [PMID: 39052000 PMCID: PMC11272161 DOI: 10.7554/elife.93077] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/27/2024] Open
Abstract
High-throughput vertebrate animal model systems for the study of patient-specific biology and new therapeutic approaches for aggressive brain tumors are currently lacking, and new approaches are urgently needed. Therefore, to build a patient-relevant in vivo model of human glioblastoma, we expressed common oncogenic variants including activated human EGFRvIII and PI3KCAH1047R under the control of the radial glial-specific promoter her4.1 in syngeneic tp53 loss-of-function mutant zebrafish. Robust tumor formation was observed prior to 45 days of life, and tumors had a gene expression signature similar to human glioblastoma of the mesenchymal subtype, with a strong inflammatory component. Within early stage tumor lesions, and in an in vivo and endogenous tumor microenvironment, we visualized infiltration of phagocytic cells, as well as internalization of tumor cells by mpeg1.1:EGFP+ microglia/macrophages, suggesting negative regulatory pressure by pro-inflammatory cell types on tumor growth at early stages of glioblastoma initiation. Furthermore, CRISPR/Cas9-mediated gene targeting of master inflammatory transcription factors irf7 or irf8 led to increased tumor formation in the primary context, while suppression of phagocyte activity led to enhanced tumor cell engraftment following transplantation into otherwise immune-competent zebrafish hosts. Altogether, we developed a genetically relevant model of aggressive human glioblastoma and harnessed the unique advantages of zebrafish including live imaging, high-throughput genetic and chemical manipulations to highlight important tumor-suppressive roles for the innate immune system on glioblastoma initiation, with important future opportunities for therapeutic discovery and optimizations.
Collapse
Affiliation(s)
- Alex Weiss
- Developmental and Stem Cell Biology Program, The Hospital for Sick ChildrenTorontoCanada
| | - Cassandra D'Amata
- Developmental and Stem Cell Biology Program, The Hospital for Sick ChildrenTorontoCanada
| | - Bret J Pearson
- Department of Molecular Genetics, University of TorontoTorontoCanada
- Knight Cancer Institute, Oregon Health & Science UniversityPortlandUnited States
- Department of Pediatrics, Papé Research Institute, Oregon Health & Science UniversityPortlandUnited States
| | - Madeline N Hayes
- Developmental and Stem Cell Biology Program, The Hospital for Sick ChildrenTorontoCanada
- Department of Molecular Genetics, University of TorontoTorontoCanada
| |
Collapse
|
15
|
Squalli Houssaini A, Lamrabet S, Nshizirungu JP, Senhaji N, Sekal M, Karkouri M, Bennis S. Glioblastoma Vaccines as Promising Immune-Therapeutics: Challenges and Current Status. Vaccines (Basel) 2024; 12:655. [PMID: 38932383 PMCID: PMC11209492 DOI: 10.3390/vaccines12060655] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2024] [Revised: 05/02/2024] [Accepted: 05/08/2024] [Indexed: 06/28/2024] Open
Abstract
Glioblastoma (GBM) is the most common and aggressive malignant brain tumor. Standard treatments including surgical resection, radiotherapy, and chemotherapy, have failed to significantly improve the prognosis of glioblastoma patients. Currently, immunotherapeutic approaches based on vaccines, chimeric antigen-receptor T-cells, checkpoint inhibitors, and oncolytic virotherapy are showing promising results in clinical trials. The combination of different immunotherapeutic approaches is proving satisfactory and promising. In view of the challenges of immunotherapy and the resistance of glioblastomas, the treatment of these tumors requires further efforts. In this review, we explore the obstacles that potentially influence the efficacy of the response to immunotherapy and that should be taken into account in clinical trials. This article provides a comprehensive review of vaccine therapy for glioblastoma. In addition, we identify the main biomarkers, including isocitrate dehydrogenase, epidermal growth factor receptor, and telomerase reverse transcriptase, known as potential immunotherapeutic targets in glioblastoma, as well as the current status of clinical trials. This paper also lists proposed solutions to overcome the obstacles facing immunotherapy in glioblastomas.
Collapse
Affiliation(s)
- Asmae Squalli Houssaini
- Laboratory of Biomedical and Translational Research, Faculty of Medicine, Pharmacy and Dental Medicine of Fez, Sidi Mohamed Ben Abdellah University, Fez 30070, Morocco;
| | - Salma Lamrabet
- Laboratory of Biomedical and Translational Research, Faculty of Medicine, Pharmacy and Dental Medicine of Fez, Sidi Mohamed Ben Abdellah University, Fez 30070, Morocco;
| | - Jean Paul Nshizirungu
- Biology Department, School of Science, College of Science and Technology, University of Rwanda, Kigali P.O. Box 3900, Rwanda;
| | - Nadia Senhaji
- Department of Biology, Faculty of Sciences, Moulay Ismail University, Meknes 50000, Morocco;
| | - Mohammed Sekal
- Laboratory of Epidemiology and Research in Health Sciences, Faculty of Medicine, Pharmacy and Dental Medicine of Fez, Sidi Mohamed Ben Abdellah University, Fez 30070, Morocco;
| | - Mehdi Karkouri
- Department of Pathological Anatomy, Ibn Rochd University Hospital of Casablanca, Casablanca 20250, Morocco;
- Laboratory of Cellular and molecular Pathology, Faculty of Medicine and Pharmacy of Casablanca, Hassan II University, Casablanca 20360, Morocco
| | - Sanae Bennis
- Laboratory of Biomedical and Translational Research, Faculty of Medicine, Pharmacy and Dental Medicine of Fez, Sidi Mohamed Ben Abdellah University, Fez 30070, Morocco;
| |
Collapse
|
16
|
Wu Q, Berglund AE, Macaulay RJ, Etame AB. The Role of Mesenchymal Reprogramming in Malignant Clonal Evolution and Intra-Tumoral Heterogeneity in Glioblastoma. Cells 2024; 13:942. [PMID: 38891074 PMCID: PMC11171993 DOI: 10.3390/cells13110942] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2024] [Revised: 05/26/2024] [Accepted: 05/29/2024] [Indexed: 06/21/2024] Open
Abstract
Glioblastoma (GBM) is the most common yet uniformly fatal adult brain cancer. Intra-tumoral molecular and cellular heterogeneities are major contributory factors to therapeutic refractoriness and futility in GBM. Molecular heterogeneity is represented through molecular subtype clusters whereby the proneural (PN) subtype is associated with significantly increased long-term survival compared to the highly resistant mesenchymal (MES) subtype. Furthermore, it is universally recognized that a small subset of GBM cells known as GBM stem cells (GSCs) serve as reservoirs for tumor recurrence and progression. The clonal evolution of GSC molecular subtypes in response to therapy drives intra-tumoral heterogeneity and remains a critical determinant of GBM outcomes. In particular, the intra-tumoral MES reprogramming of GSCs using current GBM therapies has emerged as a leading hypothesis for therapeutic refractoriness. Preventing the intra-tumoral divergent evolution of GBM toward the MES subtype via new treatments would dramatically improve long-term survival for GBM patients and have a significant impact on GBM outcomes. In this review, we examine the challenges of the role of MES reprogramming in the malignant clonal evolution of glioblastoma and provide future perspectives for addressing the unmet therapeutic need to overcome resistance in GBM.
Collapse
Affiliation(s)
- Qiong Wu
- Department of Neuro-Oncology, H. Lee Moffitt Cancer Center and Research Institute, 12902 Magnolia Drive, Tampa, FL 33612, USA
| | - Anders E. Berglund
- Department of Biostatistics and Bioinformatics, H. Lee Moffitt Cancer Center and Research Institute, 12902 Magnolia Drive, Tampa, FL 33612, USA
| | - Robert J. Macaulay
- Departments of Anatomic Pathology, H. Lee Moffitt Cancer Center and Research Institute, 12902 Magnolia Drive, Tampa, FL 33612, USA
| | - Arnold B. Etame
- Department of Neuro-Oncology, H. Lee Moffitt Cancer Center and Research Institute, 12902 Magnolia Drive, Tampa, FL 33612, USA
| |
Collapse
|
17
|
Zhao S, Li Y, Xu J, Shen L. APOBEC3C is a novel target for the immune treatment of lower-grade gliomas. Neurol Res 2024; 46:227-242. [PMID: 38007705 DOI: 10.1080/01616412.2023.2287340] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2023] [Accepted: 11/21/2023] [Indexed: 11/28/2023]
Abstract
BACKGROUND Apolipoprotein B mRNA editing catalytic polypeptide-like (APOBEC) type 3C (A3C) has been identified as a cancer molecular biomarker in the past decade. However, the practical role of A3C in lower-grade gliomas (LGGs) in improving the clinical outcome remains unclear. This study aims to discuss the function of A3C in immunotherapy in LGGs. METHODS The RNA-Sequencing (RNA-seq) and corresponding clinical data were extracted from UCSC Xena and the results were verified in the Chinese Glioma Genome Atlas (CGGA). Weighted gene co-expression network analysis (WGCNA) was used for screening A3C-related genes. Comprehensive bioinformation analyses were performed and multiple levels of expression, survival rate, and biological functions were assessed to explore the functions of A3C. RESULTS A3C expression was significantly higher in LGGs than in normal tissues but lower than in glioblastoma (GBM), indicating its role as an independent prognosis predictor for LGGs. Twenty-eight A3C-related genes were found with WGCNA for unsupervised clustering analysis and three modification patterns with different outcomes and immune cell infiltration were identified. A3C and the A3C score were also correlated with immune cell infiltration and the expression of immune checkpoints. In addition, the A3C score was correlated with increased sensitivity to chemotherapy. Single-cell RNA (scRNA) analysis indicated that A3C most probably expresses on immune cells, such as T cells, B cells and macrophage. CONCLUSIONS A3C is an immune-related prognostic biomarker in LGGs. Developing drugs to block A3C could enhance the efficiency of immunotherapy and improve disease survival.Abbreviation: A3C: Apolipoprotein B mRNA editing catalytic polypeptide-like (APOBEC) type 3C; LGGs: lower-grade gliomas; CGGA: Chinese Glioma Genome Atlas; WGCNA: Weighted gene co-expression network analysis; scRNA: Single-cell RNA; HGG: higher-grade glioma; OS: overall survival; TME: tumor microenvironment; KM: Kaplan-Meier; PFI: progression-free interval; IDH: isocitrate dehydrogenase; ROC: receiver operating characteristic; GS: gene significance; MM: module membership; TIMER: Tumor IMmune Estimation Resource; GSVA: gene set variation analysis; ssGSEA: single-sample gene-set enrichment analysis; PCA: principal component analysis; AUC: area under ROC curve; HAVCR2: hepatitis A virus cellular receptor 2; PDCD1: programmed cell death 1; PDCD1LG2: PDCD1 ligand 2; PTPRC: protein tyrosine phosphatase receptor type C; ACC: Adrenocortical carcinoma; BLCA: Bladder Urothelial Carcinoma;BRCA: Breast invasive carcinoma; CESC: Cervical squamous cell carcinoma and endocervical adenocarcinoma; CHOLCholangiocarcinoma; COADColon adenocarcinoma; DLBC: Lymphoid Neoplasm Diffuse Large B-cell Lymphoma; ESCA: Esophageal carcinoma; GBM: Glioblastoma multiforme; HNSC: Head and Neck squamous cell carcinoma; KICH: Kidney Chromophobe; KIRC: Kidney renal clear cell carcinoma; KIRP: Kidney renal papillary cell carcinoma; LAML: Acute Myeloid Leukemia; LGG: Brain Lower Grade Glioma; LIHC: Liver hepatocellular carcinoma; LUAD: Lung adenocarcinoma; LUSC: Lung squamous cell carcinoma; MESO: Mesothelioma; OV: Ovarian serous cystadenocarcinoma; PAAD: Pancreatic adenocarcinoma; PCPG: Pheochromocytoma and Paraganglioma; PRAD: Prostate adenocarcinoma; READ: Rectum adenocarcinoma; SARC: Sarcoma; SKCM: Skin Cutaneous Melanoma; STAD: Stomach adenocarcinoma; TGCT: Testicular Germ Cell Tumors; THCA: Thyroid carcinoma; THYM: Thymoma; UCEC: Uterine Corpus Endometrial Carcinoma; UCS: Uterine Carcinosarcoma; UVM: Uveal Melanoma.
Collapse
Affiliation(s)
- Shufa Zhao
- Department of Neurosurgery, Huzhou Cent Hospital, Affiliated Cent Hospital Huzhou University, Huzhou, Zhejiang, China
| | - Yuntao Li
- Department of Neurosurgery, Huzhou Cent Hospital, Affiliated Cent Hospital Huzhou University, Huzhou, Zhejiang, China
| | - Jie Xu
- Department of Neurosurgery, Huzhou Cent Hospital, Affiliated Cent Hospital Huzhou University, Huzhou, Zhejiang, China
| | - Liang Shen
- Department of Neurosurgery, The affiliated Changzhou Second People's Hospital of Nanjing Medical University, Changzhou, Jiangsu, China
| |
Collapse
|
18
|
Singh RR, Mondal I, Janjua T, Popat A, Kulshreshtha R. Engineered smart materials for RNA based molecular therapy to treat Glioblastoma. Bioact Mater 2024; 33:396-423. [PMID: 38059120 PMCID: PMC10696434 DOI: 10.1016/j.bioactmat.2023.11.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2023] [Revised: 10/19/2023] [Accepted: 11/14/2023] [Indexed: 12/08/2023] Open
Abstract
Glioblastoma (GBM) is an aggressive malignancy of the central nervous system (CNS) that remains incurable despite the multitude of improvements in cancer therapeutics. The conventional chemo and radiotherapy post-surgery have only been able to improve the prognosis slightly; however, the development of resistance and/or tumor recurrence is almost inevitable. There is a pressing need for adjuvant molecular therapies that can successfully and efficiently block tumor progression. During the last few decades, non-coding RNAs (ncRNAs) have emerged as key players in regulating various hallmarks of cancer including that of GBM. The levels of many ncRNAs are dysregulated in cancer, and ectopic modulation of their levels by delivering antagonists or overexpression constructs could serve as an attractive option for cancer therapy. The therapeutic potential of several types of ncRNAs, including miRNAs, lncRNAs, and circRNAs, has been validated in both in vitro and in vivo models of GBM. However, the delivery of these RNA-based therapeutics is highly challenging, especially to the tumors of the brain as the blood-brain barrier (BBB) poses as a major obstacle, among others. Also, since RNA is extremely fragile in nature, careful considerations must be met while designing a delivery agent. In this review we have shed light on how ncRNA therapy can overcome the limitations of its predecessor conventional therapy with an emphasis on smart nanomaterials that can aide in the safe and targeted delivery of nucleic acids to treat GBM. Additionally, critical gaps that currently exist for successful transition from viral to non-viral vector delivery systems have been identified. Finally, we have provided a perspective on the future directions, potential pathways, and target areas for achieving rapid clinical translation of, RNA-based macromolecular therapy to advance the effective treatment of GBM and other related diseases.
Collapse
Affiliation(s)
- Ravi Raj Singh
- Department of Biochemical Engineering and Biotechnology, Indian Institute of Technology Delhi, New Delhi, India
- School of Pharmacy, The University of Queensland, Brisbane, QLD, 4072, Australia
- University of Queensland –IIT Delhi Academy of Research (UQIDAR)
| | - Indranil Mondal
- Department of Biochemical Engineering and Biotechnology, Indian Institute of Technology Delhi, New Delhi, India
| | - Taskeen Janjua
- School of Pharmacy, The University of Queensland, Brisbane, QLD, 4072, Australia
| | - Amirali Popat
- School of Pharmacy, The University of Queensland, Brisbane, QLD, 4072, Australia
- Department of Functional Materials and Catalysis, Faculty of Chemistry, University of Vienna, Währinger Straße 42, 1090 Vienna, Austria
| | - Ritu Kulshreshtha
- Department of Biochemical Engineering and Biotechnology, Indian Institute of Technology Delhi, New Delhi, India
| |
Collapse
|
19
|
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
|
20
|
Liguori GL. Challenges and Promise for Glioblastoma Treatment through Extracellular Vesicle Inquiry. Cells 2024; 13:336. [PMID: 38391949 PMCID: PMC10886570 DOI: 10.3390/cells13040336] [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: 12/15/2023] [Revised: 01/31/2024] [Accepted: 02/08/2024] [Indexed: 02/24/2024] Open
Abstract
Glioblastoma (GB) is a rare but extremely aggressive brain tumor that significantly impacts patient outcomes, affecting both duration and quality of life. The protocol established by Stupp and colleagues in 2005, based on radiotherapy and chemotherapy with Temozolomide, following maximum safe surgical resection remains the gold standard for GB treatment; however, it is evident nowadays that the extreme intratumoral and intertumoral heterogeneity, as well as the invasiveness and tendency to recur, of GB are not compatible with a routine and unfortunately ineffective treatment. This review article summarizes the main challenges in the search for new valuable therapies for GB and focuses on the impact that extracellular vesicle (EV) research and exploitation may have in the field. EVs are natural particles delimited by a lipidic bilayer and filled with functional cellular content that are released and uptaken by cells as key means of cell communication. Furthermore, EVs are stable in body fluids and well tolerated by the immune system, and are able to cross physiological, interspecies, and interkingdom barriers and to target specific cells, releasing inherent or externally loaded functionally active molecules. Therefore, EVs have the potential to be ideal allies in the fight against GB and to improve the prognosis for GB patients. The present work describes the main preclinical results obtained so far on the use of EVs for GB treatment, focusing on both the EV sources and molecular cargo used in the various functional studies, primarily in vivo. Finally, a SWOT analysis is performed, highlighting the main advantages and pitfalls of developing EV-based GB therapeutic strategies. The analysis also suggests the main directions to explore to realize the possibility of exploiting EVs for the treatment of GB.
Collapse
Affiliation(s)
- Giovanna L Liguori
- Institute of Genetics and Biophysics (IGB) "Adriano Buzzati-Traverso", National Research Council (CNR) of Italy, 80131 Naples, Italy
| |
Collapse
|
21
|
Dusoswa SA, Verhoeff J, van Asten S, Lübbers J, van den Braber M, Peters S, Abeln S, Crommentuijn MH, Wesseling P, Vandertop WP, Twisk JWR, Würdinger T, Noske D, van Kooyk Y, Garcia-Vallejo JJ. The immunological landscape of peripheral blood in glioblastoma patients and immunological consequences of age and dexamethasone treatment. Front Immunol 2024; 15:1343484. [PMID: 38318180 PMCID: PMC10839779 DOI: 10.3389/fimmu.2024.1343484] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2023] [Accepted: 01/02/2024] [Indexed: 02/07/2024] Open
Abstract
Background Glioblastomas manipulate the immune system both locally and systemically, yet, glioblastoma-associated changes in peripheral blood immune composition are poorly studied. Age and dexamethasone administration in glioblastoma patients have been hypothesized to limit the effectiveness of immunotherapy, but their effects remain unclear. We compared peripheral blood immune composition in patients with different types of brain tumor to determine the influence of age, dexamethasone treatment, and tumor volume. Methods High-dimensional mass cytometry was used to characterise peripheral blood mononuclear cells of 169 patients with glioblastoma, lower grade astrocytoma, metastases and meningioma. We used blood from medically-refractory epilepsy patients and healthy controls as control groups. Immune phenotyping was performed using FlowSOM and t-SNE analysis in R followed by supervised annotation of the resulting clusters. We conducted multiple linear regression analysis between intracranial pathology and cell type abundance, corrected for clinical variables. We tested correlations between cell type abundance and survival with Cox-regression analyses. Results Glioblastoma patients had significantly fewer naive CD4+ T cells, but higher percentages of mature NK cells than controls. Decreases of naive CD8+ T cells and alternative monocytes and an increase of memory B cells in glioblastoma patients were influenced by age and dexamethasone treatment, and only memory B cells by tumor volume. Progression free survival was associated with percentages of CD4+ regulatory T cells and double negative T cells. Conclusion High-dimensional mass cytometry of peripheral blood in patients with different types of intracranial tumor provides insight into the relation between intracranial pathology and peripheral immune status. Wide immunosuppression associated with age and pre-operative dexamethasone treatment provide further evidence for their deleterious effects on treatment with immunotherapy.
Collapse
Affiliation(s)
- Sophie A. Dusoswa
- Department of Molecular Cell Biology and Immunology, Amsterdam Infection and Immunity Institute, Cancer Center Amsterdam, Amsterdam UMC, VU Amsterdam, Amsterdam, Netherlands
- Department of Neurosurgery, Amsterdam UMC, VU Amsterdam, Amsterdam, Netherlands
| | - Jan Verhoeff
- Department of Molecular Cell Biology and Immunology, Amsterdam Infection and Immunity Institute, Cancer Center Amsterdam, Amsterdam UMC, VU Amsterdam, Amsterdam, Netherlands
| | - Saskia van Asten
- Department of Molecular Cell Biology and Immunology, Amsterdam Infection and Immunity Institute, Cancer Center Amsterdam, Amsterdam UMC, VU Amsterdam, Amsterdam, Netherlands
| | - Joyce Lübbers
- Department of Molecular Cell Biology and Immunology, Amsterdam Infection and Immunity Institute, Cancer Center Amsterdam, Amsterdam UMC, VU Amsterdam, Amsterdam, Netherlands
| | - Marlous van den Braber
- Department of Molecular Cell Biology and Immunology, Amsterdam Infection and Immunity Institute, Cancer Center Amsterdam, Amsterdam UMC, VU Amsterdam, Amsterdam, Netherlands
| | - Sophie Peters
- Department of Molecular Cell Biology and Immunology, Amsterdam Infection and Immunity Institute, Cancer Center Amsterdam, Amsterdam UMC, VU Amsterdam, Amsterdam, Netherlands
| | - Sanne Abeln
- Department of Computer Science, Free University, Amsterdam, Netherlands
| | - Matheus H.W. Crommentuijn
- Department of Molecular Cell Biology and Immunology, Amsterdam Infection and Immunity Institute, Cancer Center Amsterdam, Amsterdam UMC, VU Amsterdam, Amsterdam, Netherlands
| | - Pieter Wesseling
- Department of Pathology, Cancer Center Amsterdam, Brain Tumor Center Amsterdam, Amsterdam and Princes Máxima Center for Pediatric Oncology, Amsterdam UMC, VU Amsterdam, Utrecht, Netherlands
| | | | - Jos W. R. Twisk
- Department of Epidemiology and Biostatistics and Biostatistics, Amsterdam Public Health Research Institute, Amsterdam UMC, VU Amsterdam, Amsterdam, Netherlands
| | - Thomas Würdinger
- Department of Neurosurgery, Amsterdam UMC, VU Amsterdam, Amsterdam, Netherlands
| | - David Noske
- Department of Neurosurgery, Amsterdam UMC, VU Amsterdam, Amsterdam, Netherlands
| | - Yvette van Kooyk
- Department of Molecular Cell Biology and Immunology, Amsterdam Infection and Immunity Institute, Cancer Center Amsterdam, Amsterdam UMC, VU Amsterdam, Amsterdam, Netherlands
| | - Juan J. Garcia-Vallejo
- Department of Molecular Cell Biology and Immunology, Amsterdam Infection and Immunity Institute, Cancer Center Amsterdam, Amsterdam UMC, VU Amsterdam, Amsterdam, Netherlands
| |
Collapse
|
22
|
Low JT, Brown MC, Reitman ZJ, Bernstock JD, Markert JM, Friedman GK, Waitkus MS, Bowie ML, Ashley DM. Understanding and therapeutically exploiting cGAS/STING signaling in glioblastoma. J Clin Invest 2024; 134:e163452. [PMID: 38226619 PMCID: PMC10786687 DOI: 10.1172/jci163452] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2024] Open
Abstract
Since the discovery that cGAS/STING recognizes endogenous DNA released from dying cancer cells and induces type I interferon and antitumor T cell responses, efforts to understand and therapeutically target the STING pathway in cancer have ensued. Relative to other cancer types, the glioma immune microenvironment harbors few infiltrating T cells, but abundant tumor-associated myeloid cells, possibly explaining disappointing responses to immune checkpoint blockade therapies in cohorts of patients with glioblastoma. Notably, unlike most extracranial tumors, STING expression is absent in the malignant compartment of gliomas, likely due to methylation of the STING promoter. Nonetheless, several preclinical studies suggest that inducing cGAS/STING signaling in the glioma immune microenvironment could be therapeutically beneficial, and cGAS/STING signaling has been shown to mediate inflammatory and antitumor effects of other modalities either in use or being developed for glioblastoma therapy, including radiation, tumor-treating fields, and oncolytic virotherapy. In this Review, we discuss cGAS/STING signaling in gliomas, its implications for glioma immunobiology, compartment-specific roles for STING signaling in influencing immune surveillance, and efforts to target STING signaling - either directly or indirectly - for antiglioma therapy.
Collapse
Affiliation(s)
| | | | - Zachary J. Reitman
- Department of Radiation Oncology, Duke University, Durham, North Carolina, USA
| | - Joshua D. Bernstock
- Department of Neurosurgery, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts, USA
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA
| | - James M. Markert
- Department of Neurosurgery, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Gregory K. Friedman
- Department of Pediatrics, Division of Pediatric Hematology and Oncology, University of Alabama at Birmingham, Birmingham, Alabama, USA
- Division of Pediatrics, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | | | | | | |
Collapse
|
23
|
Fei X, Wu J, Tian H, Jiang D, Chen H, Yan K, Wang Y, Zhao Y, Chen H, Xie X, Wang Z, Zhu W, Huang Q. Glioma stem cells remodel immunotolerant microenvironment in GBM and are associated with therapeutic advancements. Cancer Biomark 2024; 41:1-24. [PMID: 39240627 PMCID: PMC11492047 DOI: 10.3233/cbm-230486] [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: 11/15/2023] [Accepted: 07/19/2024] [Indexed: 09/07/2024]
Abstract
Glioma is the most common primary tumor of the central nervous system (CNS). Glioblastoma (GBM) is incurable with current treatment strategies. Additionally, the treatment of recurrent GBM (rGBM) is often referred to as terminal treatment, necessitating hospice-level care and management. The presence of the blood-brain barrier (BBB) gives GBM a more challenging or "cold" tumor microenvironment (TME) than that of other cancers and gloma stem cells (GSCs) play an important role in the TME remodeling, occurrence, development and recurrence of giloma. In this review, our primary focus will be on discussing the following topics: niche-associated GSCs and macrophages, new theories regarding GSC and TME involving pyroptosis and ferroptosis in GBM, metabolic adaptations of GSCs, the influence of the cold environment in GBM on immunotherapy, potential strategies to transform the cold GBM TME into a hot one, and the advancement of GBM immunotherapy and GBM models.
Collapse
Affiliation(s)
- Xifeng Fei
- Department of Neurosurgery, Suzhou Kowloon Hospital, Shanghai Jiaotong University School of Medicine, Suzhou, Jiangsu, China
| | - Jie Wu
- Department of Neurosurgery, Suzhou Kowloon Hospital, Shanghai Jiaotong University School of Medicine, Suzhou, Jiangsu, China
- Department of Neurosurgery, The Affiliated Suzhou Science and Technology Town Hospital of Nanjing University Medical School, Suzhou, Jiangsu, China
| | - Haiyan Tian
- Department of Neurosurgery, Suzhou Kowloon Hospital, Shanghai Jiaotong University School of Medicine, Suzhou, Jiangsu, China
- Department of GCP, Suzhou Kowloon Hospital, Shanghai Jiaotong University School of Medicine, Suzhou, Jiangsu, China
| | - Dongyi Jiang
- Department of Neurosurgery, Suzhou Kowloon Hospital, Shanghai Jiaotong University School of Medicine, Suzhou, Jiangsu, China
| | - Hanchun Chen
- Department of Neurosurgery, Suzhou Kowloon Hospital, Shanghai Jiaotong University School of Medicine, Suzhou, Jiangsu, China
| | - Ke Yan
- Department of Neurosurgery, The Affiliated Suzhou Science and Technology Town Hospital of Nanjing University Medical School, Suzhou, Jiangsu, China
| | - Yuan Wang
- Pediatric Cancer Center, College of Pharmaceutical Sciences, Soochow University, Suzhou, Jiangsu, China
| | - Yaodong Zhao
- Department of Neurosurgery, Shanghai General Hospital, Shanghai, China
| | - Hua Chen
- Department of Neurosurgery, The First Affiliated Hospital with Nanjing Medical University, Nanjing, Jiangsu, China
| | - Xiangtong Xie
- Department of Neurosurgery, Suzhou Kowloon Hospital, Shanghai Jiaotong University School of Medicine, Suzhou, Jiangsu, China
| | - Zhimin Wang
- Department of Neurosurgery, Suzhou Kowloon Hospital, Shanghai Jiaotong University School of Medicine, Suzhou, Jiangsu, China
- Department of Neurosurgery, Dushu Lake Hospital Affiliated to Soochow University, Suzhou, Jiangsu, China
| | - Wenyu Zhu
- Department of Neurosurgery, The Affiliated Suzhou Science and Technology Town Hospital of Nanjing University Medical School, Suzhou, Jiangsu, China
| | - Qiang Huang
- Department of Neurosurgery, Second Affiliated Hospital of Suzhou Medical College of Soochow University, Suzhou, Jiangsu, China
| |
Collapse
|
24
|
Rynda AY, Rostovthev DM, Zabrodskaya YM, Olyushin VE. Immunotherapy with autologous dendritic cells in the complex treatment of malignant gliomas - results. J Neurooncol 2024; 166:309-319. [PMID: 38227144 DOI: 10.1007/s11060-023-04559-1] [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: 11/30/2023] [Accepted: 12/27/2023] [Indexed: 01/17/2024]
Abstract
ANNOTATION Malignant gliomas are the most common primary brain tumor. Despite the variety of modern treatments, it is still a fatal disease with an extremely poor prognosis. The use of immunotherapy as a technique for the treatment of malignant tumors has great promise, retraining and exploiting the patient's immune response against tumors. OBJECTIVE Evaluation of the effectiveness of dendritic cell vaccine in patients with malignant brain gliomas in the structure of complex treatment in comparison with the control group of patients without immunotherapy in the structure of treatment. MATERIALS AND METHODS In a single-center, prospective, cohort study, taking place on the basis of the RNSI named after prof. A.L. Polenov, 91 patients with morphologically established malignant glial tumor (glioblastoma) took part. The main group of 41 patients who, in addition to standard treatment (surgical, radiation and chemotherapy), underwent specific antitumor immunotherapy. 50 patients received only standard treatment, without immunotherapy. RESULTS Median survival was 21.7 months in the immunotherapy group (95% CI 4-37 months) and 15.8 months (95% CI 3-22 months) in the non-immunotherapy group (p = 0.002). The median relapse-free period in the group with immunotherapy was 13.8 months (95% CI 1-20 months), and in the group without immunotherapy 7.9 months (95% CI 1-12 months) (p = 0.003). CONCLUSION In general, the use of immunotherapy in the structure of complex treatment of patients with malignant gliomas demonstrates a clear positive trend in terms of overall survival and median relapse-free period. But nevertheless, immunotherapy requires further development as a therapeutic tool, study and improvement, which will take into account immunosuppression in malignant gliomas and means of overcoming it, optimization in terms of target antigen selection, cell preparation and integration of dendritic vaccines into other treatment regimens.
Collapse
Affiliation(s)
- Artemii Yuryevich Rynda
- Russian Neurosurgical Institute (RNSI) named after prof. A.L. Polenov- a branch of the National Medical Research Center named after V.A. Almazov »Ministry of Health of Russia, St. Petersburg, Russia.
| | - Dmitrii M Rostovthev
- Russian Neurosurgical Institute (RNSI) named after prof. A.L. Polenov- a branch of the National Medical Research Center named after V.A. Almazov »Ministry of Health of Russia, St. Petersburg, Russia
| | - Yulia M Zabrodskaya
- Russian Neurosurgical Institute (RNSI) named after prof. A.L. Polenov- a branch of the National Medical Research Center named after V.A. Almazov »Ministry of Health of Russia, St. Petersburg, Russia
| | - Victor E Olyushin
- Russian Neurosurgical Institute (RNSI) named after prof. A.L. Polenov- a branch of the National Medical Research Center named after V.A. Almazov »Ministry of Health of Russia, St. Petersburg, Russia
| |
Collapse
|
25
|
Amin T, Hossain A, Jerin N, Mahmud I, Rahman MA, Rafiqul Islam SM, Islam SMBUL. Immunoediting Dynamics in Glioblastoma: Implications for Immunotherapy Approaches. Cancer Control 2024; 31:10732748241290067. [PMID: 39353594 PMCID: PMC11459535 DOI: 10.1177/10732748241290067] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2024] [Revised: 09/14/2024] [Accepted: 09/23/2024] [Indexed: 10/04/2024] Open
Abstract
Glioblastoma is an aggressive primary brain tumor that poses many therapeutic difficulties because of the high rate of proliferation, genetic variability, and its immunosuppressive microenvironment. The theory of cancer immunoediting, which includes the phases of elimination, equilibrium, and escape, offers a paradigm for comprehending interactions between the immune system and glioblastoma. Immunoediting indicates the process by which immune cells initially suppress tumor development, but thereafter select for immune-resistant versions leading to tumor escape and progression. The tumor microenvironment (TME) in glioblastoma is particularly immunosuppressive, with regulatory T cells and myeloid-derived suppressor cells being involved in immune escape. To achieve an efficient immunotherapy for glioblastoma, it is crucial to understand these mechanisms within the TME. Existing immunotherapeutic modalities such as chimeric antigen receptor T cells and immune checkpoint inhibitors have been met with some level of resistance because of the heterogeneous nature of the immune response to glioblastoma. Solving these issues is critical to develop novel strategies capable of modulating the TME and re-establishing normal immune monitoring. Further studies should be conducted to identify the molecular and cellular events that underlie the immunosuppressive tumor microenvironment in glioblastoma. Comprehending and modifying the stages of immunoediting in glioblastoma could facilitate the development of more potent and long-lasting therapies.
Collapse
Affiliation(s)
- Tasbir Amin
- Department of Biochemistry & Microbiology, North South University, Dhaka, Bangladesh
| | - Amana Hossain
- Department of Biochemistry & Microbiology, North South University, Dhaka, Bangladesh
| | - Nusrat Jerin
- Department of Biochemistry & Microbiology, North South University, Dhaka, Bangladesh
| | - Imteaz Mahmud
- Department of Public Health, North South University, Dhaka, Bangladesh
| | - Md Ahasanur Rahman
- Department of Physiology and Biophysics, Howard University, College of Medicine, Washington, DC, USA
| | - SM Rafiqul Islam
- Surgery Branch, National Cancer Institute, National Institute of Health, Bethesda, USA
| | - S M Bakhtiar UL Islam
- Department of Biochemistry & Microbiology, North South University, Dhaka, Bangladesh
| |
Collapse
|
26
|
Guo Z, Su Z, Wei Y, Zhang X, Hong X. Pyroptosis in glioma: Current management and future application. Immunol Rev 2024; 321:152-168. [PMID: 38063042 DOI: 10.1111/imr.13294] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2024]
Abstract
Glioma, the predominant form of central nervous system (CNS) malignancies, presents a significant challenge due to its high prevalence and low 5-year survival rate. The efficacy of current treatment methods is limited by the presence of the blood-brain barrier, the immunosuppressive microenvironment, and other factors. Immunotherapy has emerged as a promising approach, as it can overcome the blood-brain barrier. A tumor's immune privilege, which is induced by an immunosuppressive environment, constricts immunotherapy's clinical impact in glioma. Pyroptosis, a programmed cell death mechanism facilitated by gasdermins, plays a significant role in the management of glioma. Its ability to initiate and regulate tumor occurrence, progression, and metastasis is well-established. However, it is crucial to note that uncontrolled or excessive cell death can result in tissue damage, acute inflammation, and cytokine release syndrome, thereby potentially promoting tumor advancement or recurrence. This paper aims to elucidate the molecular pathways involved in pyroptosis and subsequently discuss its induction in cancer therapy. In addition, the current treatment methods of glioma and the use of pyroptosis in these treatments are introduced. It is hoped to provide more ideas for the treatment of glioma.
Collapse
Affiliation(s)
- Zeshang Guo
- Department of Neurosurgery, The First Bethune Hospital of Jilin University, Changchun, China
| | - Zhenjin Su
- Department of Neurosurgery, The First Bethune Hospital of Jilin University, Changchun, China
| | - Ying Wei
- Department of Radiology, The First Bethune Hospital of Jilin University, Changchun, China
| | - Xingmei Zhang
- College of Medical Technology, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Xinyu Hong
- Department of Neurosurgery, The First Bethune Hospital of Jilin University, Changchun, China
| |
Collapse
|
27
|
Mundt D, Melguizo-Gavilanes I, Tumu AY, Dubner S, Walters MK, McFarlane L. Somatic POLE Mutation and Ultra-Hypermutated Genotype in a De Novo High-Grade, Isocitrate Dehydrogenase Wild-Type Glioma: Treatment Implications. JCO Precis Oncol 2024; 8:e2300324. [PMID: 38237101 DOI: 10.1200/po.23.00324] [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: 06/21/2023] [Revised: 09/29/2023] [Accepted: 10/26/2023] [Indexed: 01/23/2024] Open
Abstract
Pembrolizumab leads to a durable response in ultra-hypermutated, high-grade, glioma.
Collapse
|
28
|
Ramsoomair CK, Ceccarelli M, Heiss JD, Shah AH. The epitranscriptome of high-grade gliomas: a promising therapeutic target with implications from the tumor microenvironment to endogenous retroviruses. J Transl Med 2023; 21:893. [PMID: 38071304 PMCID: PMC10709919 DOI: 10.1186/s12967-023-04725-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2023] [Accepted: 11/13/2023] [Indexed: 12/18/2023] Open
Abstract
Glioblastoma (GBM) comprises 45.6% of all primary malignant brain cancers and is one of the most common and aggressive intracranial tumors in adults. Intratumoral heterogeneity with a wide range of proteomic, genetic, and epigenetic dysregulation contributes to treatment resistance and poor prognosis, thus demanding novel therapeutic approaches. To date, numerous clinical trials have been developed to target the proteome and epigenome of high-grade gliomas with promising results. However, studying RNA modifications, or RNA epitranscriptomics, is a new frontier within neuro-oncology. RNA epitranscriptomics was discovered in the 1970s, but in the last decade, the extent of modification of mRNA and various non-coding RNAs has emerged and been implicated in transposable element activation and many other oncogenic processes within the tumor microenvironment. This review provides background information and discusses the therapeutic potential of agents modulating epitranscriptomics in high-grade gliomas. A particular emphasis will be placed on how combination therapies that include immune agents targeting hERV-mediated viral mimicry could improve the treatment of GBM.
Collapse
Affiliation(s)
- Christian K Ramsoomair
- Section of Virology and Immunotherapy, Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, 1095 NW 14Th Terrace, Miami, FL, 33136, USA.
- Medical Scientist Training Program, University of Miami Miller School of Medicine, 1095 NW 14Th Terrace, Miami, FL, 33136, USA.
| | - Michele Ceccarelli
- Sylvester Comprehensive Cancer Center, Miller School of Medicine, University of Miami, 1550 N.W. 10Th Avenue, Miami, FL, 33136, USA
| | - John D Heiss
- Surgical Neurology Branch, Disorders and Stroke, National Institute of Neurological, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Ashish H Shah
- Section of Virology and Immunotherapy, Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, 1095 NW 14Th Terrace, Miami, FL, 33136, USA.
| |
Collapse
|
29
|
Mao J, Li J, Chen J, Wen Q, Cao M, Zhang F, Li B, Zhang Q, Wang Z, Zhang J, Shen J. CXCL10 and Nrf2-upregulated mesenchymal stem cells reinvigorate T lymphocytes for combating glioblastoma. J Immunother Cancer 2023; 11:e007481. [PMID: 38056897 PMCID: PMC10711923 DOI: 10.1136/jitc-2023-007481] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/19/2023] [Indexed: 12/08/2023] Open
Abstract
BACKGROUND Lack of tumor-infiltrating T lymphocytes and concurrent T-cell dysfunction have been identified as major contributors to glioblastoma (GBM) immunotherapy resistance. Upregulating CXCL10 in the tumor microenvironment (TME) is a promising immunotherapeutic approach that potentially increases tumor-infiltrating T cells and boosts T-cell activity but is lacking effective delivery methods. METHODS In this study, mesenchymal stem cells (MSCs) were transduced with a recombinant lentivirus encoding Cxcl10, Nrf2 (an anti-apoptosis gene), and a ferritin heavy chain (Fth) reporter gene in order to increase their CXCL10 secretion, TME survival, and MRI visibility. Using FTH-MRI guidance, these cells were injected into the tumor periphery of orthotopic GL261 and CT2A GBMs in mice. Combination therapy consisting of CXCL10-Nrf2-FTH-MSC transplantation together with immune checkpoint blockade (ICB) was also performed for CT2A GBMs. Thereafter, in vivo and serial MRI, survival analysis, and histology examinations were conducted to assess the treatments' efficacy and mechanism. RESULTS CXCL10-Nrf2-FTH-MSCs exhibit enhanced T lymphocyte recruitment, oxidative stress tolerance, and iron accumulation. Under in vivo FTH-MRI guidance and monitoring, peritumoral transplantation of CXCL10-Nrf2-FTH-MSCs remarkably inhibited orthotopic GL261 and CT2A tumor growth in C57BL6 mice and prolonged animal survival. While ICB alone demonstrated no therapeutic impact, CXCL10-Nrf2-FTH-MSC transplantation combined with ICB demonstrated an enhanced anticancer effect for CT2A GBMs compared with transplanting it alone. Histology revealed that peritumorally injected CXCL10-Nrf2-FTH-MSCs survived longer in the TME, increased CXCL10 production, and ultimately remodeled the TME by increasing CD8+ T cells, interferon-γ+ cytotoxic T lymphocytes (CTLs), GzmB+ CTLs, and Th1 cells while reducing regulatory T cells (Tregs), exhausted CD8+ and exhausted CD4+ T cells. CONCLUSIONS MRI-guided peritumoral administration of CXCL10 and Nrf2-overexpressed MSCs can significantly limit GBM growth by revitalizing T lymphocytes within TME. The combination application of CXCL10-Nrf2-FTH-MSC transplantation and ICB therapy presents a potentially effective approach to treating GBM.
Collapse
Affiliation(s)
- Jiaji Mao
- Department of Radiology, Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, Guangdong, China
| | - Jianing Li
- Department of Radiology, Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, Guangdong, China
| | - Junwei Chen
- Department of Radiology, Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, Guangdong, China
| | - Qin Wen
- Department of Radiology, Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, Guangdong, China
| | - Minghui Cao
- Department of Radiology, Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, Guangdong, China
| | - Fang Zhang
- Department of Radiology, Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, Guangdong, China
| | - Baoxun Li
- Department of Radiology, Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, Guangdong, China
| | - Qinyuan Zhang
- Department of Radiology, Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, Guangdong, China
| | - Zhe Wang
- Department of Radiology, Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, Guangdong, China
| | - Jingzhong Zhang
- The Key Laboratory of Bio-Medical Diagnostics, Suzhou Institute of Biomedical Engineering and Technology, Suzhou, Jiangsu, China
| | - Jun Shen
- Department of Radiology, Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, Guangdong, China
| |
Collapse
|
30
|
Tian Z, Yang Z, Jin M, Ding R, Wang Y, Chai Y, Wu J, Yang M, Zhao W. Identification of cytokine-predominant immunosuppressive class and prognostic risk signatures in glioma. J Cancer Res Clin Oncol 2023; 149:13185-13200. [PMID: 37479756 DOI: 10.1007/s00432-023-05173-4] [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: 06/11/2023] [Accepted: 07/09/2023] [Indexed: 07/23/2023]
Abstract
PURPOSE The advent of immune checkpoint blockade (ICB) therapies this year has changed the way glioblastoma (GBM) is treated. Meanwhile, some patients with strong PD-L1 expression remain immune checkpoint resistant. To better understand the molecular processes that influence the immune environment, there is an urgent need to characterize the immunosuppressive tumor microenvironment and identify biomarkers to predict patient survival outcomes. PATIENTS AND METHODS Our study analyzed RNA-sequencing data from 178 GBM samples. Their unique gene expression patterns in the tumor microenvironment were analyzed by an unsupervised clustering algorithm. Through these expression patterns, a panel of T-cell exhaustion signatures, immunosuppressive cells, and clinical features correlates with immunotherapy response. The presence or absence of immune status and prognostic signatures was then validated with the test dataset. RESULTS 38.2% of GBM patients showed increased expression of anti-inflammatory cytokines, significant enrichment of T cell exhaustion signals, higher proportion of immunosuppressive cells (macrophages and CD4 regulatory T cells) and nine inhibitory checkpoints (CTLA4, PDCD1, LAG3, BTLA, TIGIT, HAVCR2, IDO1, SIGLEC7, and VISTA). The immunodepleted class (IDC) was used to classify these immunocompromised individuals. Despite the high density of tumor-infiltrating lymphocytes shown by IDC, such patients have a poor prognosis. Although PD-L1 was highly expressed in IDC, it suggested that there might be ICB resistance. There are many IDC predictive signatures to discover. CONCLUSION PD-1 is strongly expressed in a novel immunosuppressive class of GBM, but this cluster may be resistant to ICB therapy. A comprehensive description of this drug-resistant tumor microenvironment could provide new insights into drug resistance mechanisms and improved immunotherapy techniques.
Collapse
Affiliation(s)
- Ziyue Tian
- The Affiliated Hospital of Changchun University of Chinese Medicine, Changchun, 130117, China
| | - Zhongyi Yang
- The Affiliated Hospital of Changchun University of Chinese Medicine, Changchun, 130117, China
| | - Meng Jin
- The Third Affiliated Hospital of Changchun University of Chinese Medicine, Changchun, 130117, China
| | - Ran Ding
- School of Biomedical Sciences and Engineering, South China University of Technology, Guangzhou International Campus, Guangzhou, 511442, Guangdong, China
| | - Yuhan Wang
- School of Medical Informatics Engineering, Changchun University of Chinese Medicine, Changchun, 130118, Jilin, China
| | - Yuying Chai
- The Affiliated Hospital of Changchun University of Chinese Medicine, Changchun, 130117, China
| | - Jinpu Wu
- The Affiliated Hospital of Changchun University of Chinese Medicine, Changchun, 130117, China
| | - Miao Yang
- The Affiliated Hospital of Changchun University of Chinese Medicine, Changchun, 130117, China
| | - Weimin Zhao
- The Affiliated Hospital of Changchun University of Chinese Medicine, Changchun, 130117, China.
| |
Collapse
|
31
|
Angom RS, Nakka NMR, Bhattacharya S. Advances in Glioblastoma Therapy: An Update on Current Approaches. Brain Sci 2023; 13:1536. [PMID: 38002496 PMCID: PMC10669378 DOI: 10.3390/brainsci13111536] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2023] [Revised: 10/16/2023] [Accepted: 10/26/2023] [Indexed: 11/26/2023] Open
Abstract
Glioblastoma multiforme (GBM) is a primary malignant brain tumor characterized by a high grade of malignancy and an extremely unfavorable prognosis. The current efficacy of established treatments for GBM is insufficient, necessitating the prompt development of novel therapeutic approaches. The progress made in the fundamental scientific understanding of GBM is swiftly translated into more advanced stages of therapeutic studies. Despite extensive efforts to identify new therapeutic approaches, GBM exhibits a high mortality rate. The current efficacy of treatments for GBM patients is insufficient due to factors such as tumor heterogeneity, the blood-brain barrier, glioma stem cells, drug efflux pumps, and DNA damage repair mechanisms. Considering this, pharmacological cocktail therapy has demonstrated a growing efficacy in addressing these challenges. Towards this, various forms of immunotherapy, including the immune checkpoint blockade, chimeric antigen receptor T (CAR T) cell therapy, oncolytic virotherapy, and vaccine therapy have emerged as potential strategies for enhancing the prognosis of GBM. Current investigations are focused on exploring combination therapies to mitigate undesirable side effects and enhance immune responses against tumors. Furthermore, clinical trials are underway to evaluate the efficacy of several strategies to circumvent the blood-brain barrier (BBB) to achieve targeted delivery in patients suffering from recurrent GBM. In this review, we have described the biological and molecular targets for GBM therapy, pharmacologic therapy status, prominent resistance mechanisms, and new treatment approaches. We also discuss these promising therapeutic approaches to assess prospective innovative therapeutic agents and evaluated the present state of preclinical and clinical studies in GBM treatment. Overall, this review attempts to provide comprehensive information on the current status of GBM therapy.
Collapse
Affiliation(s)
- Ramcharan Singh Angom
- Department of Biochemistry and Molecular Biology, Mayo Clinic College of Medicine and Science, 4500 San Pablo Road South, Jacksonville, FL 32224, USA; (R.S.A.); (N.M.R.N.)
| | - Naga Malleswara Rao Nakka
- Department of Biochemistry and Molecular Biology, Mayo Clinic College of Medicine and Science, 4500 San Pablo Road South, Jacksonville, FL 32224, USA; (R.S.A.); (N.M.R.N.)
| | - Santanu Bhattacharya
- Department of Biochemistry and Molecular Biology, Mayo Clinic College of Medicine and Science, 4500 San Pablo Road South, Jacksonville, FL 32224, USA; (R.S.A.); (N.M.R.N.)
- Department of Physiology and Biomedical Engineering, Mayo Clinic College of Medicine and Science, 4500 San Pablo Road South, Jacksonville, FL 32224, USA
| |
Collapse
|
32
|
Miller TE, El Farran CA, Couturier CP, Chen Z, D’Antonio JP, Verga J, Villanueva MA, Castro LNG, Tong YE, Saadi TA, Chiocca AN, Fischer DS, Heiland DH, Guerriero JL, Petrecca K, Suva ML, Shalek AK, Bernstein BE. Programs, Origins, and Niches of Immunomodulatory Myeloid Cells in Gliomas. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.10.24.563466. [PMID: 37961527 PMCID: PMC10634776 DOI: 10.1101/2023.10.24.563466] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/15/2023]
Abstract
Gliomas are incurable malignancies notable for an immunosuppressive microenvironment with abundant myeloid cells whose immunomodulatory properties remain poorly defined. Here, utilizing scRNA-seq data for 183,062 myeloid cells from 85 human tumors, we discover that nearly all glioma-associated myeloid cells express at least one of four immunomodulatory activity programs: Scavenger Immunosuppressive, C1Q Immunosuppressive, CXCR4 Inflammatory, and IL1B Inflammatory. All four programs are present in IDH1 mutant and wild-type gliomas and are expressed in macrophages, monocytes, and microglia whether of blood or resident myeloid cell origins. Integrating our scRNA-seq data with mitochondrial DNA-based lineage tracing, spatial transcriptomics, and organoid explant systems that model peripheral monocyte infiltration, we show that these programs are driven by microenvironmental cues and therapies rather than myeloid cell type, origin, or mutation status. The C1Q Immunosuppressive program is driven by routinely administered dexamethasone. The Scavenger Immunosuppressive program includes ligands with established roles in T-cell suppression, is induced in hypoxic regions, and is associated with immunotherapy resistance. Both immunosuppressive programs are less prevalent in lower-grade gliomas, which are instead enriched for the CXCR4 Inflammatory program. Our study provides a framework to understand immunomodulatory myeloid cells in glioma, and a foundation to develop more effective immunotherapies.
Collapse
Affiliation(s)
- Tyler E. Miller
- Department of Pathology and Center for Cancer Research, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA
- Broad Institute of Harvard and MIT, Cambridge, MA 02142, USA
- Department of Cancer Biology, Dana Farber Cancer Institute, Boston, MA 02215, USA
- Department of Cell Biology and Pathology, Harvard Medical School, Boston, MA 02215, USA
- Ludwig Center at Harvard Medical School, Boston, MA, USA
| | - Chadi A. El Farran
- Broad Institute of Harvard and MIT, Cambridge, MA 02142, USA
- Department of Cancer Biology, Dana Farber Cancer Institute, Boston, MA 02215, USA
- Department of Cell Biology and Pathology, Harvard Medical School, Boston, MA 02215, USA
- Ludwig Center at Harvard Medical School, Boston, MA, USA
| | - Charles P. Couturier
- Broad Institute of Harvard and MIT, Cambridge, MA 02142, USA
- Department of Cancer Biology, Dana Farber Cancer Institute, Boston, MA 02215, USA
- Institute for Medical Engineering and Sciences and Department of Chemistry, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
- Department of Neurosurgery, Brigham and Women’s Hospital, Boston, MA 02115 USA
- Ragon Institute of MGH, MIT and Harvard, Cambridge, MA 02139, USA
| | - Zeyu Chen
- Broad Institute of Harvard and MIT, Cambridge, MA 02142, USA
- Department of Cancer Biology, Dana Farber Cancer Institute, Boston, MA 02215, USA
- Department of Cell Biology and Pathology, Harvard Medical School, Boston, MA 02215, USA
| | - Joshua P. D’Antonio
- Department of Cancer Biology, Dana Farber Cancer Institute, Boston, MA 02215, USA
- Department of Cell Biology and Pathology, Harvard Medical School, Boston, MA 02215, USA
| | - Julia Verga
- Department of Cancer Biology, Dana Farber Cancer Institute, Boston, MA 02215, USA
| | - Martin A. Villanueva
- Broad Institute of Harvard and MIT, Cambridge, MA 02142, USA
- Institute for Medical Engineering and Sciences and Department of Chemistry, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - L. Nicolas Gonzalez Castro
- Broad Institute of Harvard and MIT, Cambridge, MA 02142, USA
- Center for Neuro-Oncology, Dana-Farber Cancer Institute; Department of Neurology, Brigham and Women’s Hospital, Boston, MA 02115 USA
| | - Yuzhou Evelyn Tong
- Broad Institute of Harvard and MIT, Cambridge, MA 02142, USA
- Institute for Medical Engineering and Sciences and Department of Chemistry, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
- Ragon Institute of MGH, MIT and Harvard, Cambridge, MA 02139, USA
| | - Tariq Al Saadi
- Department of Neurology and Neurosurgery, Montreal Neurological Institute-Hospital, McGill University, Montreal, Quebec, Canada
| | - Andrew N. Chiocca
- Department of Cancer Biology, Dana Farber Cancer Institute, Boston, MA 02215, USA
| | | | - Dieter Henrik Heiland
- Microenvironment and Immunology Research Laboratory, Medical Center - University of Freiburg, Freiburg, Germany. Department of Neurological Surgery, Northwestern University Feinberg School of Medicine, Chicago, USA
| | - Jennifer L. Guerriero
- Ludwig Center at Harvard Medical School, Boston, MA, USA
- Breast Oncology Program, Dana-Farber Cancer Institute; Division of Breast Surgery, Department of Surgery, Brigham and Women’s Hospital, Boston, MA, USA
| | - Kevin Petrecca
- Department of Neurology and Neurosurgery, Montreal Neurological Institute-Hospital, McGill University, Montreal, Quebec, Canada
| | - Mario L. Suva
- Department of Pathology and Center for Cancer Research, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA
- Broad Institute of Harvard and MIT, Cambridge, MA 02142, USA
| | - Alex K. Shalek
- Broad Institute of Harvard and MIT, Cambridge, MA 02142, USA
- Institute for Medical Engineering and Sciences and Department of Chemistry, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
- Ragon Institute of MGH, MIT and Harvard, Cambridge, MA 02139, USA
| | - Bradley E. Bernstein
- Broad Institute of Harvard and MIT, Cambridge, MA 02142, USA
- Department of Cancer Biology, Dana Farber Cancer Institute, Boston, MA 02215, USA
- Department of Cell Biology and Pathology, Harvard Medical School, Boston, MA 02215, USA
- Ludwig Center at Harvard Medical School, Boston, MA, USA
| |
Collapse
|
33
|
Surendran A, Jenner AL, Karimi E, Fiset B, Quail DF, Walsh LA, Craig M. Agent-Based Modelling Reveals the Role of the Tumor Microenvironment on the Short-Term Success of Combination Temozolomide/Immune Checkpoint Blockade to Treat Glioblastoma. J Pharmacol Exp Ther 2023; 387:66-77. [PMID: 37442619 DOI: 10.1124/jpet.122.001571] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2022] [Revised: 06/08/2023] [Accepted: 06/13/2023] [Indexed: 07/15/2023] Open
Abstract
Glioblastoma is the most common and deadly primary brain tumor in adults. All glioblastoma patients receiving standard-of-care surgery-radiotherapy-chemotherapy (i.e., temozolomide (TMZ)) recur, with an average survival time of only 15 months. New approaches to the treatment of glioblastoma, including immune checkpoint blockade and oncolytic viruses, offer the possibility of improving glioblastoma outcomes and have as such been under intense study. Unfortunately, these treatment modalities have thus far failed to achieve approval. Recently, in an attempt to bolster efficacy and improve patient outcomes, regimens combining chemotherapy and immune checkpoint inhibitors have been tested in trials. Unfortunately, these efforts have not resulted in significant increases to patient survival. To better understand the various factors impacting treatment outcomes of combined TMZ and immune checkpoint blockade, we developed a systems-level, computational model that describes the interplay between glioblastoma, immune, and stromal cells with this combination treatment. Initializing our model to spatial resection patient samples labeled using imaging mass cytometry, our model's predictions show how the localization of glioblastoma cells, influence therapeutic success. We further validated these predictions in samples of brain metastases from patients given they generally respond better to checkpoint blockade compared with primary glioblastoma. Ultimately, our model provides novel insights into the mechanisms of therapeutic success of immune checkpoint inhibitors in brain tumors and delineates strategies to translate combination immunotherapy regimens more effectively into the clinic. SIGNIFICANCE STATEMENT: Extending survival times for glioblastoma patients remains a critical challenge. Although immunotherapies in combination with chemotherapy hold promise, clinical trials have not shown much success. Here, systems models calibrated to and validated against patient samples can improve preclinical and clinical studies by shedding light on the factors distinguishing responses/failures. By initializing our model with imaging mass cytometry visualization of patient samples, we elucidate how factors such as localization of glioblastoma cells and CD8+ T cell infiltration impact treatment outcomes.
Collapse
Affiliation(s)
- Anudeep Surendran
- Department of Mathematics and Statistics, Université de Montréal, Montréal, Canada (A.S., M.C.); Centre de recherches mathématiques, Montréal, Canada (A.S.); School of Mathematical Sciences, Queensland University of Technology, Brisbane, Australia (A.L.J.); Rosalind and Morris Goodman Cancer Institute, McGill University, Montréal, Canada (E.K., B.F., D.F.Q., L.A.W.); Department of Physiology, Faculty of Medicine, McGill University, Montréal, Canada (D.F.Q.); Department of Medicine, Division of Experimental Medicine, McGill University, Montréal, Canada (D.F.Q.); Department of Human Genetics, McGill University, Montréal, Canada (L.A.W.); and Sainte-Justine University Hospital Research Centre, Montréal, Canada (M.C.)
| | - Adrianne L Jenner
- Department of Mathematics and Statistics, Université de Montréal, Montréal, Canada (A.S., M.C.); Centre de recherches mathématiques, Montréal, Canada (A.S.); School of Mathematical Sciences, Queensland University of Technology, Brisbane, Australia (A.L.J.); Rosalind and Morris Goodman Cancer Institute, McGill University, Montréal, Canada (E.K., B.F., D.F.Q., L.A.W.); Department of Physiology, Faculty of Medicine, McGill University, Montréal, Canada (D.F.Q.); Department of Medicine, Division of Experimental Medicine, McGill University, Montréal, Canada (D.F.Q.); Department of Human Genetics, McGill University, Montréal, Canada (L.A.W.); and Sainte-Justine University Hospital Research Centre, Montréal, Canada (M.C.)
| | - Elham Karimi
- Department of Mathematics and Statistics, Université de Montréal, Montréal, Canada (A.S., M.C.); Centre de recherches mathématiques, Montréal, Canada (A.S.); School of Mathematical Sciences, Queensland University of Technology, Brisbane, Australia (A.L.J.); Rosalind and Morris Goodman Cancer Institute, McGill University, Montréal, Canada (E.K., B.F., D.F.Q., L.A.W.); Department of Physiology, Faculty of Medicine, McGill University, Montréal, Canada (D.F.Q.); Department of Medicine, Division of Experimental Medicine, McGill University, Montréal, Canada (D.F.Q.); Department of Human Genetics, McGill University, Montréal, Canada (L.A.W.); and Sainte-Justine University Hospital Research Centre, Montréal, Canada (M.C.)
| | - Benoit Fiset
- Department of Mathematics and Statistics, Université de Montréal, Montréal, Canada (A.S., M.C.); Centre de recherches mathématiques, Montréal, Canada (A.S.); School of Mathematical Sciences, Queensland University of Technology, Brisbane, Australia (A.L.J.); Rosalind and Morris Goodman Cancer Institute, McGill University, Montréal, Canada (E.K., B.F., D.F.Q., L.A.W.); Department of Physiology, Faculty of Medicine, McGill University, Montréal, Canada (D.F.Q.); Department of Medicine, Division of Experimental Medicine, McGill University, Montréal, Canada (D.F.Q.); Department of Human Genetics, McGill University, Montréal, Canada (L.A.W.); and Sainte-Justine University Hospital Research Centre, Montréal, Canada (M.C.)
| | - Daniela F Quail
- Department of Mathematics and Statistics, Université de Montréal, Montréal, Canada (A.S., M.C.); Centre de recherches mathématiques, Montréal, Canada (A.S.); School of Mathematical Sciences, Queensland University of Technology, Brisbane, Australia (A.L.J.); Rosalind and Morris Goodman Cancer Institute, McGill University, Montréal, Canada (E.K., B.F., D.F.Q., L.A.W.); Department of Physiology, Faculty of Medicine, McGill University, Montréal, Canada (D.F.Q.); Department of Medicine, Division of Experimental Medicine, McGill University, Montréal, Canada (D.F.Q.); Department of Human Genetics, McGill University, Montréal, Canada (L.A.W.); and Sainte-Justine University Hospital Research Centre, Montréal, Canada (M.C.)
| | - Logan A Walsh
- Department of Mathematics and Statistics, Université de Montréal, Montréal, Canada (A.S., M.C.); Centre de recherches mathématiques, Montréal, Canada (A.S.); School of Mathematical Sciences, Queensland University of Technology, Brisbane, Australia (A.L.J.); Rosalind and Morris Goodman Cancer Institute, McGill University, Montréal, Canada (E.K., B.F., D.F.Q., L.A.W.); Department of Physiology, Faculty of Medicine, McGill University, Montréal, Canada (D.F.Q.); Department of Medicine, Division of Experimental Medicine, McGill University, Montréal, Canada (D.F.Q.); Department of Human Genetics, McGill University, Montréal, Canada (L.A.W.); and Sainte-Justine University Hospital Research Centre, Montréal, Canada (M.C.)
| | - Morgan Craig
- Department of Mathematics and Statistics, Université de Montréal, Montréal, Canada (A.S., M.C.); Centre de recherches mathématiques, Montréal, Canada (A.S.); School of Mathematical Sciences, Queensland University of Technology, Brisbane, Australia (A.L.J.); Rosalind and Morris Goodman Cancer Institute, McGill University, Montréal, Canada (E.K., B.F., D.F.Q., L.A.W.); Department of Physiology, Faculty of Medicine, McGill University, Montréal, Canada (D.F.Q.); Department of Medicine, Division of Experimental Medicine, McGill University, Montréal, Canada (D.F.Q.); Department of Human Genetics, McGill University, Montréal, Canada (L.A.W.); and Sainte-Justine University Hospital Research Centre, Montréal, Canada (M.C.)
| |
Collapse
|
34
|
Srivastava R, Labani-Motlagh A, Chen A, Bohorquez JA, Qin B, Dodda M, Yang F, Ansari D, Patel S, Ji H, Trasti S, Chao Y, Patel Y, Zou H, Hu B, Yi G. Development of a human glioblastoma model using humanized DRAG mice for immunotherapy. Antib Ther 2023; 6:253-264. [PMID: 38075240 PMCID: PMC10702851 DOI: 10.1093/abt/tbad021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2023] [Revised: 09/27/2023] [Accepted: 09/28/2023] [Indexed: 12/20/2023] Open
Abstract
Glioblastoma (GBM) is the most common and lethal primary brain tumor. The development of alternative humanized mouse models with fully functional human immune cells will potentially accelerate the progress of GBM immunotherapy. We successfully generated humanized DRAG (NOD.Rag1KO.IL2RγcKO) mouse model by transplantation of human DR4+ hematopoietic stem cells (hHSCs), and effectively grafted GBM patient-derived tumorsphere cells to form xenografted tumors intracranially. The engrafted tumors recapitulated the pathological features and the immune cell composition of human GBM. Administration of anti-human PD-1 antibodies in these tumor-bearing humanized DRAG mice decreased the major tumor-infiltrating immunosuppressive cell populations, including CD4+PD-1+ and CD8+PD-1+ T cells, CD11b+CD14+HLA-DR+ macrophages, CD11b+CD14+HLA-DR-CD15- and CD11b+CD14-CD15+ myeloid-derived suppressor cells, indicating the humanized DRAG mice as a useful model to test the efficacy of GBM immunotherapy. Taken together, these results suggest that the humanized DRAG mouse model is a reliable preclinical platform for studying brain cancer immunotherapy and beyond.
Collapse
Affiliation(s)
- Rashmi Srivastava
- Department of Neurological Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA
- John G. Rangos Sr. Research Center, UPMC Children's Hospital of Pittsburgh, Pittsburgh, PA 15224, USA
| | - Alireza Labani-Motlagh
- Department of Medicine, The University of Texas at Tyler School of Medicine, Tyler, TX 75708, USA
- Center for Biomedical Research, The University of Texas Health Science Center at Tyler, Tyler, TX 75708, USA
- Department of Cellular and Molecular Biology, The University of Texas Health Science Center at Tyler, Tyler, TX 75708, USA
| | - Apeng Chen
- State Key Laboratory of Veterinary Etiological Biology, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou 730046, China
| | - Jose Alejandro Bohorquez
- Department of Medicine, The University of Texas at Tyler School of Medicine, Tyler, TX 75708, USA
- Center for Biomedical Research, The University of Texas Health Science Center at Tyler, Tyler, TX 75708, USA
- Department of Cellular and Molecular Biology, The University of Texas Health Science Center at Tyler, Tyler, TX 75708, USA
| | - Bin Qin
- Department of Neurological Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA
- John G. Rangos Sr. Research Center, UPMC Children's Hospital of Pittsburgh, Pittsburgh, PA 15224, USA
- National Centre for International Research in Cell and Gene Therapy, Academy of Medical Sciences, Zhengzhou University, Zhengzhou, Henan Province 450001, People’s Republic of China
| | - Meghana Dodda
- Department of Neurological Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA
- John G. Rangos Sr. Research Center, UPMC Children's Hospital of Pittsburgh, Pittsburgh, PA 15224, USA
| | - Fan Yang
- Department of Medicine, The University of Texas at Tyler School of Medicine, Tyler, TX 75708, USA
- Center for Biomedical Research, The University of Texas Health Science Center at Tyler, Tyler, TX 75708, USA
- Department of Cellular and Molecular Biology, The University of Texas Health Science Center at Tyler, Tyler, TX 75708, USA
| | - Danish Ansari
- Department of Medicine, The University of Texas at Tyler School of Medicine, Tyler, TX 75708, USA
- Center for Biomedical Research, The University of Texas Health Science Center at Tyler, Tyler, TX 75708, USA
- Department of Cellular and Molecular Biology, The University of Texas Health Science Center at Tyler, Tyler, TX 75708, USA
| | - Sahil Patel
- Department of Medicine, The University of Texas at Tyler School of Medicine, Tyler, TX 75708, USA
- Center for Biomedical Research, The University of Texas Health Science Center at Tyler, Tyler, TX 75708, USA
- Department of Cellular and Molecular Biology, The University of Texas Health Science Center at Tyler, Tyler, TX 75708, USA
| | - Honglong Ji
- Center for Biomedical Research, The University of Texas Health Science Center at Tyler, Tyler, TX 75708, USA
- Department of Cellular and Molecular Biology, The University of Texas Health Science Center at Tyler, Tyler, TX 75708, USA
| | - Scott Trasti
- Laboratory Animal Resource Center, Texas Tech University Health Sciences Center, Lubbock, TX 79410, USA
| | - Yapeng Chao
- Department of Neurological Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA
- John G. Rangos Sr. Research Center, UPMC Children's Hospital of Pittsburgh, Pittsburgh, PA 15224, USA
| | - Yash Patel
- Department of Neurological Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA
- John G. Rangos Sr. Research Center, UPMC Children's Hospital of Pittsburgh, Pittsburgh, PA 15224, USA
| | - Han Zou
- Department of Neurological Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA
- John G. Rangos Sr. Research Center, UPMC Children's Hospital of Pittsburgh, Pittsburgh, PA 15224, USA
| | - Baoli Hu
- Department of Neurological Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA
- John G. Rangos Sr. Research Center, UPMC Children's Hospital of Pittsburgh, Pittsburgh, PA 15224, USA
- Cancer Biology Program, UPMC Hillman Cancer Center, Pittsburgh, PA 15232, USA
| | - Guohua Yi
- Department of Medicine, The University of Texas at Tyler School of Medicine, Tyler, TX 75708, USA
- Center for Biomedical Research, The University of Texas Health Science Center at Tyler, Tyler, TX 75708, USA
- Department of Cellular and Molecular Biology, The University of Texas Health Science Center at Tyler, Tyler, TX 75708, USA
| |
Collapse
|
35
|
Shen Y, Zheng D, Hu D, Ma B, Cai C, Chen W, Zeng J, Luo J, Xiao D, Zhao Y, Wu Z, Jing G, Xie Y. The prognostic value of tumor-associated macrophages in glioma patients. Medicine (Baltimore) 2023; 102:e35298. [PMID: 37747032 PMCID: PMC10519474 DOI: 10.1097/md.0000000000035298] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/12/2023] [Revised: 08/03/2023] [Accepted: 08/29/2023] [Indexed: 09/26/2023] Open
Abstract
Glioma is a complex tumor composed of both neoplastic and non-neoplastic cells, including tumor-infiltrating leukocytes (TILs), and each cell type contributes to tumor formation and malignant progression. Among TILs, tumor-associated macrophages (TAMs) are of great importance and play a key role in the immune response to cancer. In this study, 22 types of adaptive and innate TILs were evaluated in gliomas. TAMs, which account for 38.7% of all these cells, are the most abundant immune infiltrates in the tumor microenvironment. In addition, we observed different immune cell patterns in low-grade glioma and glioblastoma. Our research indicated that there was a connection between TILs, and 13 of 22 TILs were significantly associated with patient outcomes. Finally, the prognosis and diagnostic value of TAMs were revealed using Kaplan-Meier analysis. We identified the optimal cutoff point of TAMs at an infiltrating level of 0.47 to predict patient prognosis, with a median overall survival of 448 days in patients with higher TAM infiltration levels and 2660 days in patients with lower TAM infiltration levels. These findings provide a new idea for glioma to regulate tumor-specific immunity, clarify the potential effects of TAMs on disease pathology, and provide a theoretical basis for immune intervention treatment of gliomas.
Collapse
Affiliation(s)
- Yang Shen
- Department of Cerebrovascular Disease, Huizhou First People’s Hospital, Huizhou, Guangdong, China
| | - Dingke Zheng
- Department of Cerebrovascular Disease, Huizhou First People’s Hospital, Huizhou, Guangdong, China
| | - Dong Hu
- Department of Cerebrovascular Disease, Huizhou First People’s Hospital, Huizhou, Guangdong, China
| | - Baoxin Ma
- Department of Cerebrovascular Disease, Huizhou First People’s Hospital, Huizhou, Guangdong, China
| | - Chunsheng Cai
- Department of Cerebrovascular Disease, Huizhou First People’s Hospital, Huizhou, Guangdong, China
| | - Wei Chen
- Department of Cerebrovascular Disease, Huizhou First People’s Hospital, Huizhou, Guangdong, China
| | - Jiahao Zeng
- Department of Cerebrovascular Disease, Huizhou First People’s Hospital, Huizhou, Guangdong, China
| | - Junran Luo
- Department of Cerebrovascular Disease, Huizhou First People’s Hospital, Huizhou, Guangdong, China
| | - Dan Xiao
- Department of Cerebrovascular Disease, Huizhou First People’s Hospital, Huizhou, Guangdong, China
| | - Yao Zhao
- Department of Cerebrovascular Disease, Huizhou First People’s Hospital, Huizhou, Guangdong, China
| | - Zhiyan Wu
- Department of Cerebrovascular Disease, Huizhou First People’s Hospital, Huizhou, Guangdong, China
| | - Guojie Jing
- Department of Cerebrovascular Disease, Huizhou First People’s Hospital, Huizhou, Guangdong, China
| | - Yituan Xie
- Department of Cerebrovascular Disease, Huizhou First People’s Hospital, Huizhou, Guangdong, China
| |
Collapse
|
36
|
Chen Y, Huo R, Kang W, Liu Y, Zhao Z, Fu W, Ma R, Zhang X, Tang J, Zhu Z, Lyu Q, Huang Y, Yan M, Jiang B, Chai R, Bao Z, Hu Z, Wang W, Jiang T, Cao Y, Wang J. Tumor-associated monocytes promote mesenchymal transformation through EGFR signaling in glioma. Cell Rep Med 2023; 4:101177. [PMID: 37652019 PMCID: PMC10518634 DOI: 10.1016/j.xcrm.2023.101177] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2022] [Revised: 03/12/2023] [Accepted: 08/09/2023] [Indexed: 09/02/2023]
Abstract
The role of brain immune compartments in glioma evolution remains elusive. We profile immune cells in glioma microenvironment and the matched peripheral blood from 11 patients. Glioblastoma exhibits specific infiltration of blood-originated monocytes expressing epidermal growth factor receptor (EGFR) ligands EREG and AREG, coined as tumor-associated monocytes (TAMo). TAMo infiltration is mutually exclusive with EGFR alterations (p = 0.019), while co-occurring with mesenchymal subtype (p = 4.7 × 10-7) and marking worse prognosis (p = 0.004 and 0.032 in two cohorts). Evolutionary analysis of initial-recurrent glioma pairs and single-cell study of a multi-centric glioblastoma reveal association between elevated TAMo and glioma mesenchymal transformation. Further analyses identify FOSL2 as a TAMo master regulator and demonstrates that FOSL2-EREG/AREG-EGFR signaling axis promotes glioma invasion in vitro. Collectively, we identify TAMo in tumor microenvironment and reveal its driving role in activating EGFR signaling to shape glioma evolution.
Collapse
Affiliation(s)
- Yiyun Chen
- Division of Life Science, Department of Chemical and Biological Engineering, and State Key Laboratory of Molecular Neuroscience, The Hong Kong University of Science and Technology, Hong Kong SAR, China; SIAT-HKUST Joint Laboratory of Cell Evolution and Digital Health, HKUST Shenzhen-Hong Kong Collaborative Innovation Research Institute, Futian, Shenzhen, China
| | - Ran Huo
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China; China National Clinical Research Center for Neurological Diseases, Beijing, China
| | - Weirong Kang
- Department of Pharmacology and Pharmacy, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China; Laboratory of Molecular Engineering and Nanomedicine, Dr. Li Dak-Sum Research Centre, The University of Hong Kong, Hong Kong SAR, China
| | - Yuwei Liu
- Department of Pharmacology and Pharmacy, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China; Laboratory of Molecular Engineering and Nanomedicine, Dr. Li Dak-Sum Research Centre, The University of Hong Kong, Hong Kong SAR, China
| | - Zheng Zhao
- Division of Life Science, Department of Chemical and Biological Engineering, and State Key Laboratory of Molecular Neuroscience, The Hong Kong University of Science and Technology, Hong Kong SAR, China; Beijing Neurosurgical Institute, Capital Medical University, Beijing, China
| | - Weilun Fu
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China; China National Clinical Research Center for Neurological Diseases, Beijing, China
| | - Ruochen Ma
- Division of Life Science, Department of Chemical and Biological Engineering, and State Key Laboratory of Molecular Neuroscience, The Hong Kong University of Science and Technology, Hong Kong SAR, China
| | - Xiaomeng Zhang
- Division of Life Science, Department of Chemical and Biological Engineering, and State Key Laboratory of Molecular Neuroscience, The Hong Kong University of Science and Technology, Hong Kong SAR, China
| | - Jihong Tang
- Division of Life Science, Department of Chemical and Biological Engineering, and State Key Laboratory of Molecular Neuroscience, The Hong Kong University of Science and Technology, Hong Kong SAR, China
| | - Zhihan Zhu
- Division of Life Science, Department of Chemical and Biological Engineering, and State Key Laboratory of Molecular Neuroscience, The Hong Kong University of Science and Technology, Hong Kong SAR, China
| | - Qingyang Lyu
- Department of Pharmacology and Pharmacy, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China; Laboratory of Molecular Engineering and Nanomedicine, Dr. Li Dak-Sum Research Centre, The University of Hong Kong, Hong Kong SAR, China
| | - Yi Huang
- Department of Pharmacology and Pharmacy, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China; Laboratory of Molecular Engineering and Nanomedicine, Dr. Li Dak-Sum Research Centre, The University of Hong Kong, Hong Kong SAR, China
| | - Mengli Yan
- Division of Life Science, Department of Chemical and Biological Engineering, and State Key Laboratory of Molecular Neuroscience, The Hong Kong University of Science and Technology, Hong Kong SAR, China
| | - Biaobin Jiang
- Division of Life Science, Department of Chemical and Biological Engineering, and State Key Laboratory of Molecular Neuroscience, The Hong Kong University of Science and Technology, Hong Kong SAR, China
| | - Ruichao Chai
- Division of Life Science, Department of Chemical and Biological Engineering, and State Key Laboratory of Molecular Neuroscience, The Hong Kong University of Science and Technology, Hong Kong SAR, China; SIAT-HKUST Joint Laboratory of Cell Evolution and Digital Health, HKUST Shenzhen-Hong Kong Collaborative Innovation Research Institute, Futian, Shenzhen, China; Beijing Neurosurgical Institute, Capital Medical University, Beijing, China
| | - Zhaoshi Bao
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Zheng Hu
- SIAT-HKUST Joint Laboratory of Cell Evolution and Digital Health, HKUST Shenzhen-Hong Kong Collaborative Innovation Research Institute, Futian, Shenzhen, China; CAS Key Laboratory of Quantitative Engineering Biology, Shenzhen Institute of Synthetic Biology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Weiping Wang
- Department of Pharmacology and Pharmacy, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China; Laboratory of Molecular Engineering and Nanomedicine, Dr. Li Dak-Sum Research Centre, The University of Hong Kong, Hong Kong SAR, China.
| | - Tao Jiang
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China; China National Clinical Research Center for Neurological Diseases, Beijing, China; Beijing Neurosurgical Institute, Capital Medical University, Beijing, China.
| | - Yong Cao
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China; China National Clinical Research Center for Neurological Diseases, Beijing, China.
| | - Jiguang Wang
- Division of Life Science, Department of Chemical and Biological Engineering, and State Key Laboratory of Molecular Neuroscience, The Hong Kong University of Science and Technology, Hong Kong SAR, China; SIAT-HKUST Joint Laboratory of Cell Evolution and Digital Health, HKUST Shenzhen-Hong Kong Collaborative Innovation Research Institute, Futian, Shenzhen, China; Hong Kong Center for Neurodegenerative Diseases, InnoHK, Hong Kong SAR, China.
| |
Collapse
|
37
|
Jain P, Vashist S, Panjiyar BK. Navigating the Immune Challenge in Glioblastoma: Exploring Immunotherapeutic Avenues for Overcoming Immune Suppression. Cureus 2023; 15:e46089. [PMID: 37900496 PMCID: PMC10611557 DOI: 10.7759/cureus.46089] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/27/2023] [Indexed: 10/31/2023] Open
Abstract
Glioblastoma multiforme (GBM) is a primary brain tumor known for its short survival time, typically 14-18 months from diagnosis to fatality. Managing GBM poses significant challenges due to factors like the formidable blood-brain barrier, the immunosuppressive conditions within GBM, and the intricacies of surgical procedures. Currently, the typical treatment for GBM combines surgical procedures, radiation therapy, and chemotherapy using temozolomide. Unfortunately, this conventional approach has not proven effective in substantially extending the lives of GBM patients. Consequently, researchers are exploring alternative methods for GBM management. One promising avenue receiving attention in recent years is immunotherapy. This approach has successfully treated cancer types like non-small cell lung cancer and blood-related malignancies. Various immunotherapeutic strategies are currently under investigation for GBM treatment, including checkpoint inhibitors, vaccines, chimeric antigen receptor (CAR) T-cell therapy, and oncolytic viruses. A comprehensive review of 26 high-quality studies conducted over the past decade, involving thorough searches of databases such as PubMed and Google Scholar, has been conducted. The findings from this review suggest that while immunotherapeutic strategies show promise, they face significant limitations and challenges in practical application for GBM treatment. The study emphasizes the importance of combining diverse approaches, customizing treatments for individual patients, and ongoing research efforts to improve GBM patients' outlook.
Collapse
Affiliation(s)
- Prateek Jain
- Internal Medicine, Maulana Azad Medical College, Delhi, IND
| | | | - Binay K Panjiyar
- Medicine, Harvard Medical School, Boston, USA
- Internal Medicine, California Institute of Behavioral Neurosciences & Psychology, Fairfield, USA
| |
Collapse
|
38
|
Yuan Y, Su Y, Wu Y, Xue Y, Zhang Y, Zhang Y, Zheng M, Chang T, Qu Y, Zhao T. Knowledge structure and hotspots research of glioma immunotherapy: a bibliometric analysis. Front Oncol 2023; 13:1229905. [PMID: 37671057 PMCID: PMC10476340 DOI: 10.3389/fonc.2023.1229905] [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: 05/27/2023] [Accepted: 08/03/2023] [Indexed: 09/07/2023] Open
Abstract
Background Glioma is the most common primary brain tumor. Traditional treatments for glioma include surgical resection, radiotherapy, chemotherapy, and bevacizumab therapy, but their efficacies are limited. Immunotherapy provides a new direction for glioma treatment. This study aimed to summarize the knowledge structure and research hotspots of glioma immunotherapy through a bibliometric analysis. Method Publications pertaining to glioma immunotherapy published during the period from 1st January 1990 to 27th March 2023 were downloaded from the Web of Science Core Collection (WoSCC). Bibliometric analysis and visualization were performed using the CiteSpace, VOSviewer, Online Analysis Platform of Literature Metrology, and R software. The hotspots and prospects of glioma immunotherapy research were illustrated via analyzing the countries, institutions, journals, authors, citations and keywords of eligible publications. Results A total of 1,929 publications pertaining to glioma immunotherapy in 502 journals were identified as of 27th March 2023, involving 9,505 authors from 1,988 institutions in 62 countries. Among them were 1,285 articles and 644 reviews. Most of publications were produced by the United States. JOURNAL OF NEURO-ONCOLOGY published the majority of publications pertaining to glioma immunotherapy. Among the authors, Lim M contributed the largest number of publications. Through analyzing keyword bursts and co-cited references, immune-checkpoint inhibitors (ICIs) were identified as the research focus and hotspot. Conclusion Using a bibliometric analysis, this study provided the knowledge structure and research hotspots in glioma immunotherapy research during the past 33 years, with ICIs staying in the current and future hotspot. Our findings may direct the research of glioma immunotherapy in the future.
Collapse
Affiliation(s)
- Yexin Yuan
- Department of Neurosurgery, Tangdu Hospital, Air Force Medical University, Xi’an, Shaanxi, China
| | - Yue Su
- Department of Neurology, Tangdu Hospital, Air Force Medical University, Xi’an, Shaanxi, China
| | - Yingxi Wu
- Department of Neurosurgery, Tangdu Hospital, Air Force Medical University, Xi’an, Shaanxi, China
| | - Yafei Xue
- Department of Neurosurgery, Tangdu Hospital, Air Force Medical University, Xi’an, Shaanxi, China
| | - Yunze Zhang
- Department of Neurosurgery, Tangdu Hospital, Air Force Medical University, Xi’an, Shaanxi, China
| | - Yangyang Zhang
- Department of Neurosurgery, Tangdu Hospital, Air Force Medical University, Xi’an, Shaanxi, China
| | - Min Zheng
- Department of Neurosurgery, Tangdu Hospital, Air Force Medical University, Xi’an, Shaanxi, China
| | - Ting Chang
- Department of Neurology, Tangdu Hospital, Air Force Medical University, Xi’an, Shaanxi, China
| | - Yan Qu
- Department of Neurosurgery, Tangdu Hospital, Air Force Medical University, Xi’an, Shaanxi, China
| | - Tianzhi Zhao
- Department of Neurosurgery, Tangdu Hospital, Air Force Medical University, Xi’an, Shaanxi, China
| |
Collapse
|
39
|
Sun H, Zhang H, Jing L, Zhao H, Chen B, Song W. FBP1 is a potential prognostic biomarker and correlated with tumor immunosuppressive microenvironment in glioblastoma. Neurosurg Rev 2023; 46:187. [PMID: 37507483 DOI: 10.1007/s10143-023-02097-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2023] [Revised: 07/10/2023] [Accepted: 07/22/2023] [Indexed: 07/30/2023]
Abstract
Hypoxia has been shown to contribute to tumor immunosuppressive microenvironment and is an effective prognostic indicator. This study aimed to screen prognostic hypoxia-related genes (HRGs) in glioblastoma and investigate the association between HRGs and tumor immunosuppressive microenvironment. The glioblastoma-related mRNA data were collected from TCGA, GEO, and CGGA databases. Totally 200 HRGs were obtained from the GSEA website. The prognostic HRGs were screened by univariate Cox regression analysis. Somatic mutation data of glioblastoma from TCGA was visualized using the "maftools" of R package. Immune cell infiltration proportions were calculated by CIBERSORT. The TISIDB online tool was applied to analyze the relationship between HRGs and immunoinhibitors as well as the HRG expression in different glioblastoma immune and molecular subtypes. Hub gene's mRNA and protein levels in cell lines were determined by qRT-PCR and western blot, respectively. The effects of hub gene knockdown on cell viability and migration ability were evaluated employing CCK8 and wound healing assays. The univariate Cox regression showed that high level of FBP1 (fructose-1,6-bisphosphatase 1) was a poor prognostic biomarker, and FBP1 was mainly expressed in lymphocyte depleted immune subtype of glioblastoma. High FBP1 mRNA and protein levels have been successfully validated in vitro. The somatic mutation analysis suggested that TP53 mutation rate was the highest in the high FBP1 glioblastoma group, while EGFR mutation rate was the highest in the low FBP1 glioblastoma group. In the high FBP1 group, the infiltration proportions and types of immune cells were less, dominated by macrophages M2, and the expression of CTLA4, LAG3, TIGIT, PDL1, and PDL2 was significantly upregulated. The expression of FBP1 was positively correlated with several immunoinhibitors, such as IL-10 and TGFβ-1. In conclusion, we demonstrated that FBP1 could serve as a prognostic biomarker for glioblastoma. The immune microenvironment in the high FBP1 group might be suppressed by up-regulating immune checkpoints and immunoinhibitors.
Collapse
Affiliation(s)
- Hu Sun
- Department of Neurosurgery, Zibo Central Hospital, 255000 Zibo, Shandong, China
| | - Hui Zhang
- Department of Cardiology, Zibo Central Hospital, 255000 Zibo, Shandong, China
| | - Lijie Jing
- Department of Neurosurgery, Zibo Central Hospital, 255000 Zibo, Shandong, China
| | - Hao Zhao
- Department of Neurosurgery, Zibo Central Hospital, 255000 Zibo, Shandong, China
| | - Bing Chen
- Department of Neurosurgery, The Affiliated Hospital of Qingdao University, No. 1677 Wutaishan Road, Huangdao District, Qingdao, 266000, Shandong, China.
| | - Wei Song
- Department of Breast and Thyroid Surgery, Zibo Central Hospital, No. 54 Gongqingtuan West Road, 255000 Zibo, Qingdao, Shandong, China.
| |
Collapse
|
40
|
Sun F, Lv H, Feng B, Sun J, Zhang L, Dong B. Identification of natural killer cell-related characteristics to predict the clinical prognosis and immune microenvironment of patients with low-grade glioma. Aging (Albany NY) 2023; 15:6264-6291. [PMID: 37405952 PMCID: PMC10373982 DOI: 10.18632/aging.204850] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2023] [Accepted: 06/15/2023] [Indexed: 07/07/2023]
Abstract
BACKGROUND Individuals with low-grade glioma (LGG) have a dismal prognosis, and most patients will eventually progress to high-grade disease. Therefore, it is crucial to accurately determine their prognoses. METHODS Seventy-nine NK cell genes were downloaded from the LM22 database and univariate Cox regression analysis was utilized to detect NK cell-related genes affecting prognosis. Molecular types were established for LGG using the "ConsensusClusterPlus" R package. The results from a functional enrichment analysis and the immune microenvironment were intensively explored to determine molecular heterogeneity and immune characteristics across distinct subtypes. Furthermore, a RiskScore model was developed and verified using expression profiles of NK cells, and a nomogram consisting of the RiskScore model and clinical traits was constructed. Moreover, pan-cancer traits of NK cells were also investigated. RESULTS The C1 subtype included the greatest amount of immune infiltration and the poorest prognosis among well-established subtypes. The majority of enriched pathways were those involved in tumor progression, including epithelial-mesenchymal transition and cell cycle pathways. Differentially expressed genes among distinct subtypes were determined and used to develop a novel RiskScore model. This model was able to distinguish low-risk patients with LGG from those with high-risk disease. An accurate nomogram including the RiskScore, disease grade and patient's age was constructed to predict clinical outcomes of LGG patients. Finally, a pan-cancer analysis further highlighted the crucial roles of NK cell-related genes in the tumor microenvironment. CONCLUSIONS An NK cell-related RiskScore model can accurately predict the prognoses of patients with LGG and provide valuable insights into personalized medicine.
Collapse
Affiliation(s)
- Fei Sun
- Department of Neurosurgery, The First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, China
- Department of Neurosurgery, Xinhua Hospital Affiliated to Dalian University, Dalian, Liaoning, China
| | - Hongtao Lv
- Department of Neurosurgery, The First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, China
| | - Baozhi Feng
- Department of Neurosurgery, The First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, China
| | - Jiaao Sun
- Department of Urology, The First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, China
| | - Linyun Zhang
- Department of Neurosurgery, The First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, China
| | - Bin Dong
- Department of Neurosurgery, The First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, China
| |
Collapse
|
41
|
Nisnboym M, Vincze SR, Xiong Z, Sneiderman CT, Raphael RA, Li B, Jaswal AP, Sever RE, Day KE, LaToche JD, Foley LM, Karimi H, Hitchens TK, Agnihotri S, Hu B, Rajasundaram D, Anderson CJ, Blumenthal DT, Pearce TM, Uttam S, Nedrow JR, Panigrahy A, Pollack IF, Lieberman FS, Drappatz J, Raphael I, Edwards WB, Kohanbash G. Immuno-PET Imaging of CD69 Visualizes T-Cell Activation and Predicts Survival Following Immunotherapy in Murine Glioblastoma. CANCER RESEARCH COMMUNICATIONS 2023; 3:1173-1188. [PMID: 37426447 PMCID: PMC10324623 DOI: 10.1158/2767-9764.crc-22-0434] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/25/2022] [Revised: 03/19/2023] [Accepted: 06/06/2023] [Indexed: 07/11/2023]
Abstract
Glioblastoma (GBM) is the most common and malignant primary brain tumor in adults. Immunotherapy may be promising for the treatment of some patients with GBM; however, there is a need for noninvasive neuroimaging techniques to predict immunotherapeutic responses. The effectiveness of most immunotherapeutic strategies requires T-cell activation. Therefore, we aimed to evaluate an early marker of T-cell activation, CD69, for its use as an imaging biomarker of response to immunotherapy for GBM. Herein, we performed CD69 immunostaining on human and mouse T cells following in vitro activation and post immune checkpoint inhibitors (ICI) in an orthotopic syngeneic mouse glioma model. CD69 expression on tumor-infiltrating leukocytes was assessed using single-cell RNA sequencing (scRNA-seq) data from patients with recurrent GBM receiving ICI. Radiolabeled CD69 Ab PET/CT imaging (CD69 immuno-PET) was performed on GBM-bearing mice longitudinally to quantify CD69 and its association with survival following immunotherapy. We show CD69 expression is upregulated upon T-cell activation and on tumor-infiltrating lymphocytes (TIL) in response to immunotherapy. Similarly, scRNA-seq data demonstrated elevated CD69 on TILs from patients with ICI-treated recurrent GBM as compared with TILs from control cohorts. CD69 immuno-PET studies showed a significantly higher tracer uptake in the tumors of ICI-treated mice compared with controls. Importantly, we observed a positive correlation between survival and CD69 immuno-PET signals in immunotherapy-treated animals and established a trajectory of T-cell activation by virtue of CD69-immuno-PET measurements. Our study supports the potential use of CD69 immuno-PET as an immunotherapy response assessment imaging tool for patients with GBM. Significance Immunotherapy may hold promise for the treatment of some patients with GBM. There is a need to assess therapy responsiveness to allow the continuation of effective treatment in responders and to avoid ineffective treatment with potential adverse effects in the nonresponders. We demonstrate that noninvasive PET/CT imaging of CD69 may allow early detection of immunotherapy responsiveness in patients with GBM.
Collapse
Affiliation(s)
- Michal Nisnboym
- Department of Neurological Surgery, UPMC Children's Hospital of Pittsburgh, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
- Department of Neurology, Tel-Aviv Sourasky Medical Center, Tel-Aviv University, Tel-Aviv, Israel
| | - Sarah R. Vincze
- Department of Neurological Surgery, UPMC Children's Hospital of Pittsburgh, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Zujian Xiong
- Department of Neurological Surgery, UPMC Children's Hospital of Pittsburgh, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Chaim T. Sneiderman
- Department of Neurological Surgery, UPMC Children's Hospital of Pittsburgh, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Rebecca A. Raphael
- Department of Computational and Systems Biology, UPMC Hillman Cancer Center, Cancer Biology Program, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Bo Li
- Department of Neurological Surgery, UPMC Children's Hospital of Pittsburgh, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Ambika P. Jaswal
- Department of Neurological Surgery, UPMC Children's Hospital of Pittsburgh, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - ReidAnn E. Sever
- Department of Neurological Surgery, UPMC Children's Hospital of Pittsburgh, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Kathryn E. Day
- In Vivo Imaging Facility, University of Pittsburgh Medical Center, UPMC Hillman Cancer Center, Pittsburgh, Pennsylvania
| | - Joseph D. LaToche
- In Vivo Imaging Facility, University of Pittsburgh Medical Center, UPMC Hillman Cancer Center, Pittsburgh, Pennsylvania
| | - Lesley M. Foley
- In Vivo Imaging Facility, University of Pittsburgh Medical Center, UPMC Hillman Cancer Center, Pittsburgh, Pennsylvania
| | - Hanieh Karimi
- Department of Biochemistry, University of Missouri, Columbia, Missouri
| | - T. Kevin Hitchens
- In Vivo Imaging Facility, University of Pittsburgh Medical Center, UPMC Hillman Cancer Center, Pittsburgh, Pennsylvania
- Department of Neurobiology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Sameer Agnihotri
- Department of Neurological Surgery, UPMC Children's Hospital of Pittsburgh, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Baoli Hu
- Department of Neurological Surgery, UPMC Children's Hospital of Pittsburgh, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Dhivyaa Rajasundaram
- Division of Health Informatics, Department of Pediatrics, UPMC Children's Hospital of Pittsburgh, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | | | - Deborah T. Blumenthal
- Neuro-oncology Division, Tel-Aviv Sourasky Medical Center, Tel-Aviv University, Tel-Aviv, Israel
| | - Thomas M. Pearce
- Division of Neuropathology, Department of Pathology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Shikhar Uttam
- Department of Computational and Systems Biology, UPMC Hillman Cancer Center, Cancer Biology Program, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Jessie R. Nedrow
- In Vivo Imaging Facility, University of Pittsburgh Medical Center, UPMC Hillman Cancer Center, Pittsburgh, Pennsylvania
| | - Ashok Panigrahy
- Department of Radiology, UPMC Children's Hospital of Pittsburgh, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania
| | - Ian F. Pollack
- Department of Neurological Surgery, UPMC Children's Hospital of Pittsburgh, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Frank S. Lieberman
- Neuro-oncology Program, Division of Hematology/Oncology, UPMC Hillman Cancer Center, Pittsburgh, Pennsylvania
| | - Jan Drappatz
- Neuro-oncology Program, Division of Hematology/Oncology, UPMC Hillman Cancer Center, Pittsburgh, Pennsylvania
| | - Itay Raphael
- Department of Neurological Surgery, UPMC Children's Hospital of Pittsburgh, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Wilson B. Edwards
- Department of Biochemistry, University of Missouri, Columbia, Missouri
| | - Gary Kohanbash
- Department of Neurological Surgery, UPMC Children's Hospital of Pittsburgh, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
- Department of Immunology, University of Pittsburgh, Pittsburgh, Pennsylvania
| |
Collapse
|
42
|
Zhang G, Shen L, Li Z, Zhao Y. FDX1 serves as a prognostic biomarker and promotes glioma progression by regulating the immune response. Aging (Albany NY) 2023; 15:204772. [PMID: 37301546 PMCID: PMC10292899 DOI: 10.18632/aging.204772] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2022] [Accepted: 05/23/2023] [Indexed: 06/12/2023]
Abstract
The present study investigates the prognostic value of the FDX1 gene and its association with immune infiltration in gliomas. Gene expression profiles and corresponding clinical parameters of glioma patients were obtained from the Cancer Genome Atlas and Chinese Glioma Genome Atlas databases. In vitro experiments were also performed to validate its impact on malignant phenotypes of glioma cells. Kaplan-Meier analysis demonstrated that high FDX1 expression was associated with poor prognosis in glioma. Function and pathway enrichment for FDX1 predominantly demonstrated immunomodulatory function. In addition, the high-FDX1 expression group had higher Estimation of Stromal and Immune cells in malignant tumor tissues using Expression data, stromal, and immune scores (p<0.001). On evaluation of immunotherapy response, TIDE and dysfunction scores were higher in the low-FDX1 group, while the exclusion score demonstrated an opposite trend. In vitro tests showed that FDX1 silencing-induced inhibition of cell invasion and migration inactivated the nucleotide oligomerization domain (NOD)-like receptor signaling pathway by regulating PD-L1 expression. Notably, NOD1 expression was reversed in FDX1-knockdown cells after treatment with NOD1 agonists. In conclusion, FDX1 may play an important role in the diagnosis and treatment of gliomas. Regulating its expression may therefore help improve immunotherapy for these tumors.
Collapse
Affiliation(s)
- Guangying Zhang
- Department of Infectious Diseases, Hunan Key Laboratory of Viral Hepatitis, Xiangya Hospital, Central South University, Changsha 410008, Hunan Province, P.R. China
| | - Liangfang Shen
- Department of Oncology, Xiangya Hospital, Central South University, Changsha 410008, Hunan Province, P.R. China
| | - Zhanzhan Li
- Department of Oncology, Xiangya Hospital, Central South University, Changsha 410008, Hunan Province, P.R. China
| | - Yajie Zhao
- Department of Nuclear Medicine, Xiangya Hospital, Central South University, Changsha 410008, Hunan Province, P.R. China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha 410008, Hunan Province, P.R. China
| |
Collapse
|
43
|
Padmakumar S, Amiji MM. Long-Acting Therapeutic Delivery Systems for the Treatment of Gliomas. Adv Drug Deliv Rev 2023; 197:114853. [PMID: 37149040 DOI: 10.1016/j.addr.2023.114853] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2023] [Revised: 04/13/2023] [Accepted: 04/23/2023] [Indexed: 05/08/2023]
Abstract
Despite the emergence of cutting-edge therapeutic strategies and tremendous progress in research, a complete cure of glioma remains elusive. The heterogenous nature of tumor, immunosuppressive state and presence of blood brain barrier are few of the major obstacles in this regard. Long-acting depot formulations such as injectables and implantables are gaining attention for drug delivery to brain owing to their ease in administration and ability to elute drug locally for extended durations in a controlled manner with minimal toxicity. Hybrid matrices fabricated by incorporating nanoparticulates within such systems help to enhance pharmaceutical advantages. Utilization of long-acting depots as monotherapy or in conjunction with existing strategies rendered significant survival benefits in many preclinical studies and some clinical trials. The discovery of novel targets, immunotherapeutic strategies and alternative drug administration routes are now coupled with several long-acting systems with an ultimate aim to enhance patient survival and prevent glioma recurrences.
Collapse
Affiliation(s)
- Smrithi Padmakumar
- Department of Pharmaceutical Sciences, School of Pharmacy, Northeastern University, Boston, MA, 02115
| | - Mansoor M Amiji
- Department of Pharmaceutical Sciences, School of Pharmacy, Northeastern University, Boston, MA, 02115; Department of Chemical Engineering, College of Engineering, Northeastern University, Boston, MA, 02115.
| |
Collapse
|
44
|
Wang Z, Dai Z, Zhang H, Liang X, Zhang X, Wen Z, Luo P, Zhang J, Liu Z, Zhang M, Cheng Q. Tumor-secreted lactate contributes to an immunosuppressive microenvironment and affects CD8 T-cell infiltration in glioblastoma. Front Immunol 2023; 14:894853. [PMID: 37122693 PMCID: PMC10130393 DOI: 10.3389/fimmu.2023.894853] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2022] [Accepted: 01/05/2023] [Indexed: 05/02/2023] Open
Abstract
Introduction Glioblastoma is a malignant brain tumor with poor prognosis. Lactate is the main product of tumor cells, and its secretion may relate to immunocytes' activation. However, its role in glioblastoma is poorly understood. Methods This work performed bulk RNA-seq analysis and single cell RNA-seq analysis to explore the role of lactate in glioblastoma progression. Over 1400 glioblastoma samples were grouped into different clusters according to their expression and the results were validated with our own data, the xiangya cohort. Immunocytes infiltration analysis, immunogram and the map of immune checkpoint genes' expression were applied to analyze the potential connection between the lactate level with tumor immune microenvironment. Furthermore, machine learning algorithms and cell-cell interaction algorithm were introduced to reveal the connection of tumor cells with immunocytes. By co-culturing CD8 T cells with tumor cells, and performing immunohistochemistry on Xiangya cohort samples further validated results from previous analysis. Discussion In this work, lactate is proved that contributes to glioblastoma immune suppressive microenvironment. High level of lactate in tumor microenvironment can affect CD8 T cells' migration and infiltration ratio in glioblastoma. To step further, potential compounds that targets to samples from different groups were also predicted for future exploration.
Collapse
Affiliation(s)
- Zeyu Wang
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, Hunan, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, China
- MRC Centre for Regenerative Medicine, Institute for Regeneration and Repair, University of Edinburgh, Edinburgh, United Kingdom
| | - Ziyu Dai
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, Hunan, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Hao Zhang
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, Hunan, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Xisong Liang
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, Hunan, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Xun Zhang
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, Hunan, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Zhipeng Wen
- Department of Pharmacy, The Affiliated Hospital of Guizhou Medical University, Guizhou Medical University, Guiyang, Guizhou, China
| | - Peng Luo
- Department of Oncology, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Jian Zhang
- Department of Oncology, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Zaoqu Liu
- Department of Interventional Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Mingyu Zhang
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, Hunan, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, China
- Clinical Diagnosis and Therapy Center for Gliomas of Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Quan Cheng
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, Hunan, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, China
- Clinical Diagnosis and Therapy Center for Gliomas of Xiangya Hospital, Central South University, Changsha, Hunan, China
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha, Hunan, China
| |
Collapse
|
45
|
Cruz N, Herculano-Carvalho M, Roque D, Faria CC, Cascão R, Ferreira HA, Reis CP, Matela N. Highlighted Advances in Therapies for Difficult-To-Treat Brain Tumours Such as Glioblastoma. Pharmaceutics 2023; 15:pharmaceutics15030928. [PMID: 36986790 PMCID: PMC10054750 DOI: 10.3390/pharmaceutics15030928] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2023] [Revised: 02/25/2023] [Accepted: 03/10/2023] [Indexed: 03/15/2023] Open
Abstract
Glioblastoma multiforme (GBM) remains a challenging disease, as it is the most common and deadly brain tumour in adults and has no curative solution and an overall short survival time. This incurability and short survival time means that, despite its rarity (average incidence of 3.2 per 100,000 persons), there has been an increased effort to try to treat this disease. Standard of care in newly diagnosed glioblastoma is maximal tumour resection followed by initial concomitant radiotherapy and temozolomide (TMZ) and then further chemotherapy with TMZ. Imaging techniques are key not only to diagnose the extent of the affected tissue but also for surgery planning and even for intraoperative use. Eligible patients may combine TMZ with tumour treating fields (TTF) therapy, which delivers low-intensity and intermediate-frequency electric fields to arrest tumour growth. Nonetheless, the blood–brain barrier (BBB) and systemic side effects are obstacles to successful chemotherapy in GBM; thus, more targeted, custom therapies such as immunotherapy and nanotechnological drug delivery systems have been undergoing research with varying degrees of success. This review proposes an overview of the pathophysiology, possible treatments, and the most (not all) representative examples of the latest advancements.
Collapse
Affiliation(s)
- Nuno Cruz
- Instituto de Biofísica e Engenharia Biomédica, Faculdade de Ciências, Universidade de Lisboa, 1749-016 Lisboa, Portugal
- iMED.ULisboa, Research Institute for Medicines, Faculdade de Farmácia, Universidade de Lisboa, Av. Prof. Gama Pinto, 1649-003 Lisboa, Portugal
| | - Manuel Herculano-Carvalho
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, 1649-028 Lisboa, Portugal
- Department of Neurosurgery, Hospital de Santa Maria, Centro Hospitalar Universitário Lisboa Norte (CHULN), 1649-028 Lisboa, Portugal
| | - Diogo Roque
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, 1649-028 Lisboa, Portugal
- Department of Neurosurgery, Hospital de Santa Maria, Centro Hospitalar Universitário Lisboa Norte (CHULN), 1649-028 Lisboa, Portugal
| | - Cláudia C. Faria
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, 1649-028 Lisboa, Portugal
- Department of Neurosurgery, Hospital de Santa Maria, Centro Hospitalar Universitário Lisboa Norte (CHULN), 1649-028 Lisboa, Portugal
| | - Rita Cascão
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, 1649-028 Lisboa, Portugal
| | - Hugo Alexandre Ferreira
- Instituto de Biofísica e Engenharia Biomédica, Faculdade de Ciências, Universidade de Lisboa, 1749-016 Lisboa, Portugal
| | - Catarina Pinto Reis
- Instituto de Biofísica e Engenharia Biomédica, Faculdade de Ciências, Universidade de Lisboa, 1749-016 Lisboa, Portugal
- iMED.ULisboa, Research Institute for Medicines, Faculdade de Farmácia, Universidade de Lisboa, Av. Prof. Gama Pinto, 1649-003 Lisboa, Portugal
- Correspondence: (C.P.R.); (N.M.); Tel.: +351-217-946-400 (ext. 14244) (C.P.R.); Fax: +351-217-946-470 (C.P.R.)
| | - Nuno Matela
- Instituto de Biofísica e Engenharia Biomédica, Faculdade de Ciências, Universidade de Lisboa, 1749-016 Lisboa, Portugal
- Correspondence: (C.P.R.); (N.M.); Tel.: +351-217-946-400 (ext. 14244) (C.P.R.); Fax: +351-217-946-470 (C.P.R.)
| |
Collapse
|
46
|
Kerezoudis P, Kerezoudi EN, Choudhry A, Himes BT, Parney IF. Complementary and Alternative Medicine for Gliomas: Systematic Review and Critical Appraisal of Current Literature. Neurosurgery 2023; 92:464-471. [PMID: 36650046 DOI: 10.1227/neu.0000000000002236] [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: 05/18/2022] [Accepted: 09/14/2022] [Indexed: 01/19/2023] Open
Abstract
BACKGROUND Many patients with glioma and their caregivers seek complementary and alternative medicine (CAM) methods to comfort themselves, cope with cancer medication side effects, and feel they are taking control of their disease. OBJECTIVE To summarize existing evidence on safety and efficacy of CAM treatments for gliomas. METHODS We performed an exhaustive electronic literature search for in vitro, animal, and clinical studies (English language, all years available) on CAM modalities for gliomas. RESULTS A total of 378 studies (315 unique articles) were analyzed. Distribution was as follows: in vitro-274 (73%), animal-77 (20%), and clinical-26 (7%, 2491 patients). Most studies were conducted in China (n = 135, 43%), followed by the United States (n = 62, 20%) and Spain (n = 17, 5%-6%). Resveratrol was the most commonly investigated CAM therapy in the in vitro (n = 62) and in vivo (n = 17) setting. Safety/toxicity was examined in 21% of in vitro (cytotoxic at same dose in 48%), 39% of in vivo (no evidence of organ toxicity), and 50% of clinical studies (adverse events reported in 6). Cytotoxicity was the most frequent end point among in vitro (60%) and animal studies (56%), followed by synergistic action with chemotherapy and inhibition of invasiveness and migration. Finally, 7 of 26 studies found no clinical effect, whereas 5 reported possible impact on progression-free or overall survival, 3 demonstrated decrease or arrest of tumor progression, and 2 showed positive impact on symptoms and quality of life. CONCLUSION These findings will hopefully educate providers and patients and stimulate further research in the field of CAM therapy for gliomas.
Collapse
Affiliation(s)
- Panagiotis Kerezoudis
- Department of Neurologic Surgery, Albert Einstein College of Medicine, Montefiore Medical Center, Bronx, NY, USA
| | - Evangelia N Kerezoudi
- School of Medical Sciences, Faculty of Medicine and Health, Örebro University, Örebro, Sweden
| | | | - Benjamin T Himes
- Department of Neurologic Surgery, Albert Einstein College of Medicine, Montefiore Medical Center, Bronx, NY, USA
| | - Ian F Parney
- Department of Neurologic Surgery, Albert Einstein College of Medicine, Montefiore Medical Center, Bronx, NY, USA
| |
Collapse
|
47
|
Wu H, Guo C, Wang C, Xu J, Zheng S, Duan J, Li Y, Bai H, Xu Q, Ning F, Wang F, Yang Q. Single-cell RNA sequencing reveals tumor heterogeneity, microenvironment, and drug-resistance mechanisms of recurrent glioblastoma. Cancer Sci 2023. [PMID: 36853018 DOI: 10.1111/cas.15773] [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: 10/08/2022] [Revised: 02/02/2023] [Accepted: 02/20/2023] [Indexed: 03/01/2023] Open
Abstract
Glioblastomas are highly heterogeneous brain tumors. Despite the availability of standard treatment for glioblastoma multiforme (GBM), i.e., Stupp protocol, which involves surgical resection followed by radiotherapy and chemotherapy, glioblastoma remains refractory to treatment and recurrence is inevitable. Moreover, the biology of recurrent glioblastoma remains unclear. Increasing evidence has shown that intratumoral heterogeneity and the tumor microenvironment contribute to therapeutic resistance. However, the interaction between intracellular heterogeneity and drug resistance in recurrent GBMs remains controversial. The aim of this study was to map the transcriptome landscape of cancer cells and the tumor heterogeneity and tumor microenvironment in recurrent and drug-resistant GBMs at a single-cell resolution and further explore the mechanism of drug resistance of GBMs. We analyzed six tumor tissue samples from three patients with primary GBM and three patients with recurrent GBM in which recurrence and drug resistance developed after treatment with the standard Stupp protocol using single-cell RNA sequencing. Using unbiased clustering, nine major cell clusters were identified. Upregulation of the expression of stemness-related and cell-cycle-related genes was observed in recurrent GBM cells. Compared with the initial GBM tissues, recurrent GBM tissues showed a decreased proportion of microglia, consistent with previous reports. Finally, vascular endothelial growth factor A expression and the blood-brain barrier permeability were high, and the O6 -methylguanine DNA methyltransferase-related signaling pathway was activated in recurrent GBM. Our results delineate the single-cell map of recurrent glioblastoma, tumor heterogeneity, tumor microenvironment, and drug-resistance mechanisms, providing new insights into treatment strategies for recurrent glioblastomas.
Collapse
Affiliation(s)
- Haibin Wu
- Department of Neurosurgery, The First Affiliated Hospital of Nanchang University, Nanchang, China
| | - Chengcheng Guo
- Department of Neurosurgery/Neuro-oncology, Sun Yat-sen University Cancer Center, Guangzhou, China.,State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Chaoye Wang
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, China.,Department of Biometric Information, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Jiang Xu
- Department of Neurosurgery, The First Affiliated Hospital of Nanchang University, Nanchang, China
| | - Suyue Zheng
- Department of Neurosurgery, The First Affiliated Hospital of Nanchang University, Nanchang, China
| | - Jian Duan
- Department of Neurosurgery, The First Affiliated Hospital of Nanchang University, Nanchang, China
| | - Yiyun Li
- Department of Neurosurgery, The First Affiliated Hospital of Nanchang University, Nanchang, China
| | - Hongming Bai
- Department of Neurosurgery, General Hospital of Southern Theatre Command of PLA, Guangzhou, China
| | - Qiuyan Xu
- Department of Neurosurgery/Neuro-oncology, Sun Yat-sen University Cancer Center, Guangzhou, China.,State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Fangling Ning
- Department of Medical Oncology, Binzhou Medical University Hospital, Binzhou, China
| | - Feng Wang
- Department of Medical Oncology, Binzhou Medical University Hospital, Binzhou, China
| | - Qunying Yang
- Department of Neurosurgery/Neuro-oncology, Sun Yat-sen University Cancer Center, Guangzhou, China.,State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, China
| |
Collapse
|
48
|
Hamad A, Yusubalieva GM, Baklaushev VP, Chumakov PM, Lipatova AV. Recent Developments in Glioblastoma Therapy: Oncolytic Viruses and Emerging Future Strategies. Viruses 2023; 15:547. [PMID: 36851761 PMCID: PMC9958853 DOI: 10.3390/v15020547] [Citation(s) in RCA: 38] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2022] [Revised: 01/24/2023] [Accepted: 02/10/2023] [Indexed: 02/18/2023] Open
Abstract
Glioblastoma is the most aggressive form of malignant brain tumor. Standard treatment protocols and traditional immunotherapy are poorly effective as they do not significantly increase the long-term survival of glioblastoma patients. Oncolytic viruses (OVs) may be an effective alternative approach. Combining OVs with some modern treatment options may also provide significant benefits for glioblastoma patients. Here we review virotherapy for glioblastomas and describe several OVs and their combination with other therapies. The personalized use of OVs and their combination with other treatment options would become a significant area of research aiming to develop the most effective treatment regimens for glioblastomas.
Collapse
Affiliation(s)
- Azzam Hamad
- Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, 119991 Moscow, Russia
| | - Gaukhar M. Yusubalieva
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, 119991 Moscow, Russia
- Federal Research and Clinical Center of Specialized Medical Care and Medical Technologies, Federal Medical and Biological Agency of Russia, 115682 Moscow, Russia
| | - Vladimir P. Baklaushev
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, 119991 Moscow, Russia
- Federal Research and Clinical Center of Specialized Medical Care and Medical Technologies, Federal Medical and Biological Agency of Russia, 115682 Moscow, Russia
| | - Peter M. Chumakov
- Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, 119991 Moscow, Russia
| | - Anastasiya V. Lipatova
- Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, 119991 Moscow, Russia
| |
Collapse
|
49
|
Srivastava R, Labani-Motlagh A, Chen A, Yang F, Ansari D, Patel S, Ji H, Trasti S, Dodda M, Patel Y, Zou H, Hu B, Yi G. Development of a human glioblastoma model using humanized DRAG mice for immunotherapy. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.02.15.528743. [PMID: 36824969 PMCID: PMC9948970 DOI: 10.1101/2023.02.15.528743] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/18/2023]
Abstract
Glioblastoma (GBM) is the most common and lethal primary brain tumor with high mortality rates and a short median survival rate of about 15 months despite intensive multimodal treatment of maximal surgical resection, radiotherapy, and chemotherapy. Although immunotherapies have been successful in the treatment of various cancers, disappointing results from clinical trials for GBM immunotherapy represent our incomplete understanding. The development of alternative humanized mouse models with fully functional human immune cells will potentially accelerate the progress of GBM immunotherapy. In this study, we developed a humanized DRAG (NOD.Rag1KO.IL2RγcKO) mouse model, in which the human hematopoietic stem cells (HSCs) were well-engrafted and subsequently differentiated into a full lineage of immune cells. Using this humanized DRAG mouse model, GBM patient-derived tumorsphere lines were successfully engrafted to form xenografted tumors, which can recapitulate the pathological features and the immune cell composition of human GBM. Importantly, the administration of anti-human PD-1 antibodies in these DRAG mice bearing a GBM patient-derived tumorsphere line resulted in decreasing the major tumor-infiltrating immunosuppressive cell populations, including CD4 + PD-1 + and CD8 + PD-1 + T cells, CD11b + CD14 + HLA-DR + macrophages, CD11b + CD14 + HLA-DR - CD15 - and CD11b + CD14 - CD15 + myeloid-derived suppressor cells, indicating the humanized DRAG mouse model as a useful model to test the efficacy of immune checkpoint inhibitors in GBM immunotherapy. Together, these results suggest that humanized DRAG mouse models are a reliable preclinical platform for brain cancer immunotherapy and beyond.
Collapse
|
50
|
Individualized Multimodal Immunotherapy for Adults with IDH1 Wild-Type GBM: A Single Institute Experience. Cancers (Basel) 2023; 15:cancers15041194. [PMID: 36831536 PMCID: PMC9954396 DOI: 10.3390/cancers15041194] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2022] [Revised: 02/08/2023] [Accepted: 02/10/2023] [Indexed: 02/16/2023] Open
Abstract
Synergistic activity between maintenance temozolomide (TMZm) and individualized multimodal immunotherapy (IMI) during/after first-line treatment has been suggested to improve the overall survival (OS) of adults with IDH1 wild-type MGMT promoter-unmethylated (unmeth) GBM. We expand the data and include the OS of MGMT promoter-methylated (meth) adults with GBM. Unmeth (10 f, 18 m) and meth (12 f, 10 m) patients treated between 27 May 2015 and 1 January 2022 were analyzed retrospectively. There were no differences in age (median: 48 y) or Karnofsky performance index (median: 80). The IMI consisted of 5-day immunogenic cell death (ICD) therapies during TMZm: Newcastle disease virus (NDV) bolus injections and sessions of modulated electrohyperthermia (mEHT); subsequent active specific immunotherapy: dendritic cell (DC) vaccines plus modulatory immunotherapy; and maintenance ICD therapy. There were no differences in the number of vaccines (median: 2), total number of DCs (median: 25.6 × 106), number of NDV injections (median: 31), and number of mEHT sessions (median: 28) between both groups. The median OS of 28 unmeth patients was 22 m (2y-OS: 39%), confirming previous results. OS of 22 meth patients was significantly better (p = 0.0414) with 38 m (2y-OS: 81%). There were no major treatment-related adverse reactions. The addition of IMI during/after standard of care should be prospectively explored.
Collapse
|