1
|
Zhou Q, Ke X, Man J, Jiang J, Ren J, Xue C, Zhang B, Zhang P, Zhao J, Zhou J. Integrated MRI radiomics, tumor microenvironment, and clinical risk factors for improving survival prediction in patients with glioblastomas. Strahlenther Onkol 2025; 201:398-410. [PMID: 39249499 DOI: 10.1007/s00066-024-02283-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: 12/27/2023] [Accepted: 07/14/2024] [Indexed: 09/10/2024]
Abstract
PURPOSE To construct a comprehensive model for predicting the prognosis of patients with glioblastoma (GB) using a radiomics method and integrating clinical risk factors, tumor microenvironment (TME), and imaging characteristics. MATERIALS AND METHODS In this retrospective study, we included 148 patients (85 males and 63 females; median age 53 years) with isocitrate dehydrogenase-wildtype GB between January 2016 and April 2022. Patients were randomly divided into the training (n = 104) and test (n = 44) sets. The best feature combination related to GB overall survival (OS) was selected using LASSO Cox regression analyses. Clinical, radiomics, clinical-radiomics, clinical-TME, and clinical-radiomics-TME models were established. The models' concordance index (C-index) was evaluated. The survival curve was drawn using the Kaplan-Meier method, and the prognostic stratification ability of the model was tested. RESULTS LASSO Cox analyses were used to screen the factors related to OS in patients with GB, including MGMT (hazard ratio [HR] = 0.642; 95% CI 0.414-0.997; P = 0.046), TERT (HR = 1.755; 95% CI 1.095-2.813; P = 0.019), peritumoral edema (HR = 1.013; 95% CI 0.999-1.027; P = 0.049), tumor purity (TP; HR = 0.982; 95% CI 0.964-1.000; P = 0.054), CD163 + tumor-associated macrophages (TAMs; HR = 1.049; 95% CI 1.021-1.078; P < 0.001), CD68 + TAMs (HR = 1.055; 95% CI 1.018-1.093; P = 0.004), and the six radiomics features. The clinical-radiomics-TME model had the best survival prediction ability, the C‑index was 0.768 (0.717-0.819). The AUC of 1‑, 2‑, and 3‑year OS prediction in the test set was 0.842, 0.844, and 0.795, respectively. CONCLUSION The clinical-radiomics-TME model is the most effective for predicting the survival of patients with GB. Radiomics features, TP, and TAMs play important roles in the prognostic model.
Collapse
Affiliation(s)
- Qing Zhou
- Department of Radiology, Lanzhou University Second Hospital, Lanzhou, Gansu, China
- Key Laboratory of Medical Imaging of Gansu Province, Lanzhou, Gansu, China
- Gansu International Scientific and Technological Cooperation Base of Medical Imaging Artificial Intelligence, Lanzhou, Gansu, China
| | - Xiaoai Ke
- Department of Radiology, Lanzhou University Second Hospital, Lanzhou, Gansu, China
- Key Laboratory of Medical Imaging of Gansu Province, Lanzhou, Gansu, China
- Gansu International Scientific and Technological Cooperation Base of Medical Imaging Artificial Intelligence, Lanzhou, Gansu, China
| | - Jiangwei Man
- Second Clinical School, Lanzhou University, Lanzhou, Gansu, China
- Department of Surgical, Lanzhou University Second Hospital, Lanzhou, Gansu, China
| | - Jian Jiang
- Department of Radiology, Lanzhou University Second Hospital, Lanzhou, Gansu, China
- Second Clinical School, Lanzhou University, Lanzhou, Gansu, China
- Key Laboratory of Medical Imaging of Gansu Province, Lanzhou, Gansu, China
- Gansu International Scientific and Technological Cooperation Base of Medical Imaging Artificial Intelligence, Lanzhou, Gansu, China
| | - Jialiang Ren
- Department of Pharmaceuticals Diagnostics, GE HealthCare, Beijing, China
| | - Caiqiang Xue
- Department of Radiology, Lanzhou University Second Hospital, Lanzhou, Gansu, China
- Second Clinical School, Lanzhou University, Lanzhou, Gansu, China
- Key Laboratory of Medical Imaging of Gansu Province, Lanzhou, Gansu, China
- Gansu International Scientific and Technological Cooperation Base of Medical Imaging Artificial Intelligence, Lanzhou, Gansu, China
| | - Bin Zhang
- Department of Radiology, Lanzhou University Second Hospital, Lanzhou, Gansu, China
- Second Clinical School, Lanzhou University, Lanzhou, Gansu, China
- Key Laboratory of Medical Imaging of Gansu Province, Lanzhou, Gansu, China
- Gansu International Scientific and Technological Cooperation Base of Medical Imaging Artificial Intelligence, Lanzhou, Gansu, China
| | - Peng Zhang
- Second Clinical School, Lanzhou University, Lanzhou, Gansu, China
- Department of Pathology, Lanzhou University Second Hospital, Lanzhou, Gansu, China
| | - Jun Zhao
- Department of Radiology, Lanzhou University Second Hospital, Lanzhou, Gansu, China
- Key Laboratory of Medical Imaging of Gansu Province, Lanzhou, Gansu, China
- Gansu International Scientific and Technological Cooperation Base of Medical Imaging Artificial Intelligence, Lanzhou, Gansu, China
| | - Junlin Zhou
- Department of Radiology, Lanzhou University Second Hospital, Lanzhou, Gansu, China.
- Second Clinical School, Lanzhou University, Lanzhou, Gansu, China.
- Key Laboratory of Medical Imaging of Gansu Province, Lanzhou, Gansu, China.
- Gansu International Scientific and Technological Cooperation Base of Medical Imaging Artificial Intelligence, Lanzhou, Gansu, China.
- Department of Radiology, Lanzhou University Second Hospital, Cuiyingmen No. 82, 730030, Lanzhou, Gansu, China.
| |
Collapse
|
2
|
Piyadasa H, Oberlton B, Ribi M, Ranek JS, Averbukh I, Leow K, Amouzgar M, Liu CC, Greenwald NF, McCaffrey EF, Kumar R, Ferrian S, Tsai AG, Filiz F, Fullaway CC, Bosse M, Varra SR, Kong A, Sowers C, Gephart MH, Nuñez-Perez P, Yang E, Travers M, Schachter MJ, Liang S, Santi MR, Bucktrout S, Gherardini PF, Cole K, Barish ME, Brown CE, Oldridge DA, Drake RR, Phillips JJ, Okada H, Prins R, Bendall SC, Angelo M. Multi-omic landscape of human gliomas from diagnosis to treatment and recurrence. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.03.12.642624. [PMID: 40161803 PMCID: PMC11952471 DOI: 10.1101/2025.03.12.642624] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 04/02/2025]
Abstract
Gliomas are among the most lethal cancers, with limited treatment options. To uncover hallmarks of therapeutic escape and tumor microenvironment (TME) evolution, we applied spatial proteomics, transcriptomics, and glycomics to 670 lesions from 310 adult and pediatric patients. Single-cell analysis shows high B7H3+ tumor cell prevalence in glioblastoma (GBM) and pleomorphic xanthoastrocytoma (PXA), while most gliomas, including pediatric cases, express targetable tumor antigens in less than 50% of tumor cells, potentially explaining trial failures. Longitudinal samples of isocitrate dehydrogenase (IDH)-mutant gliomas reveal recurrence driven by tumor-immune spatial reorganization, shifting from T-cell and vasculature-associated myeloid cell-enriched niches to microglia and CD206+ macrophage-dominated tumors. Multi-omic integration identified N-glycosylation as the best classifier of grade, while the immune transcriptome best predicted GBM survival. Provided as a community resource, this study opens new avenues for glioma targeting, classification, outcome prediction, and a baseline of TME composition across all stages.
Collapse
Affiliation(s)
- Hadeesha Piyadasa
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, USA
| | - Benjamin Oberlton
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, USA
- Immunology Program, Stanford University School of Medicine, Stanford, CA, USA
| | - Mikaela Ribi
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, USA
- Department of Chemistry and Sarafan ChEM-H, Stanford University, Stanford, CA, USA
| | - Jolene S. Ranek
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, USA
| | - Inna Averbukh
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, USA
| | - Ke Leow
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, USA
- Cancer Biology Program, Stanford University School of Medicine, Stanford, CA, USA
| | - Meelad Amouzgar
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, USA
- Immunology Program, Stanford University School of Medicine, Stanford, CA, USA
| | - Candace C. Liu
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, USA
- Immunology Program, Stanford University School of Medicine, Stanford, CA, USA
| | - Noah F. Greenwald
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, USA
- Cancer Biology Program, Stanford University School of Medicine, Stanford, CA, USA
| | - Erin F. McCaffrey
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, USA
| | - Rashmi Kumar
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, USA
| | - Selena Ferrian
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, USA
| | - Albert G. Tsai
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, USA
| | - Ferda Filiz
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, USA
| | | | - Marc Bosse
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, USA
| | | | - Alex Kong
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, USA
| | - Cameron Sowers
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, USA
| | | | - Pablo Nuñez-Perez
- Department of Neurosurgery, Stanford University School of Medicine, Stanford, CA, USA
| | - EnJun Yang
- Parker Institute for Cancer Immunotherapy, San Francisco, CA, USA
| | - Mike Travers
- Parker Institute for Cancer Immunotherapy, San Francisco, CA, USA
| | | | - Samantha Liang
- Parker Institute for Cancer Immunotherapy, San Francisco, CA, USA
| | - Maria R. Santi
- Children’s Hospital of Philadelphia, Perelman School of Medicine, University of Pennsylvania, PA, USA
| | | | - Pier Federico Gherardini
- Parker Institute for Cancer Immunotherapy, San Francisco, CA, USA
- Department of Biology, University of Rome “Tor Vergata”, Rome, Italy
| | - Kristina Cole
- Children’s Hospital of Philadelphia, Perelman School of Medicine, University of Pennsylvania, PA, USA
| | - Michael E. Barish
- Parker Institute for Cancer Immunotherapy, San Francisco, CA, USA
- Department of Stem Cell Biology and Regenerative Medicine, Beckman Research Institute of the City of Hope, Duarte, CA, USA
| | - Christine E. Brown
- Departments of Hematology & Hematopoietic Cell Transplantation and Immuno-Oncology, Beckman Research Institute of the City of Hope, Duarte, CA, USA
| | - Derek A. Oldridge
- Parker Institute for Cancer Immunotherapy, San Francisco, CA, USA
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
- Center for Computational and Genomic Medicine, Children’s Hospital of Philadelphia, PA, USA
| | - Richard R. Drake
- Department of Pharmacology and Immunology, Medical University of South Carolina, Charleston, SC, USA
| | - Joanna J. Phillips
- Department of Pathology, University of California San Francisco, San Francisco, CA, USA
- Department of Neurological Surgery, University of California San Francisco, San Francisco, CA, USA
| | - Hideho Okada
- Parker Institute for Cancer Immunotherapy, San Francisco, CA, USA
- Department of Neurological Surgery, University of California San Francisco, San Francisco, CA, USA
| | - Robert Prins
- Parker Institute for Cancer Immunotherapy, San Francisco, CA, USA
- Department of Neurosurgery, UCLA, Los Angeles, CA, USA
| | - Sean C. Bendall
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, USA
- Immunology Program, Stanford University School of Medicine, Stanford, CA, USA
- Cancer Biology Program, Stanford University School of Medicine, Stanford, CA, USA
- Parker Institute for Cancer Immunotherapy, San Francisco, CA, USA
| | - Michael Angelo
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, USA
- Immunology Program, Stanford University School of Medicine, Stanford, CA, USA
- Cancer Biology Program, Stanford University School of Medicine, Stanford, CA, USA
- Parker Institute for Cancer Immunotherapy, San Francisco, CA, USA
| |
Collapse
|
3
|
Ispirjan M, Marx S, Freund E, Fleck SK, Baldauf J, Roessler K, Schroeder HW, Bekeschus S. Markers of tumor-associated macrophages and microglia exhibit high intratumoral heterogeneity in human glioblastoma tissue. Oncoimmunology 2024; 13:2425124. [PMID: 39523551 PMCID: PMC11556281 DOI: 10.1080/2162402x.2024.2425124] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2024] [Revised: 09/18/2024] [Accepted: 10/30/2024] [Indexed: 11/16/2024] Open
Abstract
BACKGROUND Human glioblastoma multiforme (GBM) is a highly aggressive tumor with insufficient therapies available. Especially, novel concepts of immune therapies fail due to a complex immunosuppressive microenvironment, high mutational rates, and inter-patient variations. The intratumoral heterogeneity is currently not sufficiently investigated. METHODS Biopsies from six different locations were taken in a cohort of 16 GBM patients who underwent surgery. The tissue slides were analyzed utilizing high-content imaging microscopy and algorithm-based image quantification. Several immune markers for macrophage and microglia subpopulations were investigated. Flow cytometry was used to validate key results. Besides the surface marker, cytokines were measured and categorized based on their heterogenicity and overall expression. RESULTS M2-like antigens, including CD204, CD163, Arg1, and CSF1R, showed comparatively higher expression, with GFAP displaying the least intratumoral heterogeneity. In contrast, anti-tumor-macrophage-like antigens, such as PSGL-1, CD16, CD68, and MHC-II, exhibited low overall expression and concurrent high intratumoral heterogeneity. CD16 and PSGL-1 were the most heterogeneous antigens. High expression levels were observed for cytokines IL-6, VEGF, and CCL-2. VILIP-a was revealed to differentiate most in principle component analysis. Cytokines with the lowest overall expression, such as TGF-β1, β-NGF, TNF-α, and TREM1, showed low intratumoral heterogeneity, in contrast to βNGF, TNF-α, and IL-18, which displayed high heterogeneity despite low expression. CONCLUSION The study showed high intratumoral heterogeneity in GBM, emphasizing the need for a more detailed understanding of the tumor microenvironment. The described findings could be essential for future personalized treatment strategies and the implementation of reliable diagnostics in GBM.
Collapse
Affiliation(s)
- Mikael Ispirjan
- Department of Neurosurgery, Greifswald University Medical Center, Greifswald, Germany
- ZIK plasmatis, Leibniz Institute for Plasma Science and Technology (INP), Greifswald, Germany
- Department of Cardiology, Heidelberg University Hospital, Heidelberg, Germany
| | - Sascha Marx
- Department of Neurosurgery, Greifswald University Medical Center, Greifswald, Germany
- Department of Cancer Immunology and Virology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | - Eric Freund
- ZIK plasmatis, Leibniz Institute for Plasma Science and Technology (INP), Greifswald, Germany
- Department of Neurosurgery, Medical University of Vienna, Vienna, Austria
| | - Steffen K. Fleck
- Department of Neurosurgery, Greifswald University Medical Center, Greifswald, Germany
| | - Joerg Baldauf
- Department of Neurosurgery, Greifswald University Medical Center, Greifswald, Germany
| | - Karl Roessler
- Department of Neurosurgery, Medical University of Vienna, Vienna, Austria
| | - Henry W.S. Schroeder
- Department of Neurosurgery, Greifswald University Medical Center, Greifswald, Germany
| | - Sander Bekeschus
- ZIK plasmatis, Leibniz Institute for Plasma Science and Technology (INP), Greifswald, Germany
- Department of Dermatology and Venerology, Rostock University Medical Center, Rostock, Germany
| |
Collapse
|
4
|
Salahlou R, Farajnia S, Alizadeh E, Dastmalchi S, Bargahi N, Rahbarnia L, Steyar SH. Design and in silico analysis of a novel peptide-based multiepitope vaccine against glioblastoma multiforme by targeting tumor-associated macrophage. Heliyon 2024; 10:e40774. [PMID: 39759328 PMCID: PMC11696665 DOI: 10.1016/j.heliyon.2024.e40774] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2024] [Revised: 11/23/2024] [Accepted: 11/27/2024] [Indexed: 01/07/2025] Open
Abstract
CD204 is a distinct indicator for tumor-associated macrophages (TAMs) in glioma. Evidence indicates that CD204-positive TAMs are involved in the aggressive behavior of various types of cancers. This study was conducted to develop a new and effective peptide-based vaccine for GBM, specifically targeting CD204. Epitopes of the target protein were identified using NetMHCpan 4.1a, NetMHCIIpan-4.0, and ABCpred tools. Subsequently, the predicted epitopes were evaluated using bioinformatics tools to assess their antigenicity, non-allergenicity, immunogenicity, non-toxicity, and potential to stimulate the production of IL-4 and IFN-γ in HTL epitopes. Selected T-cell epitopes demonstrated a robust binding affinity with the particular HLA alleles. Finally, four HTL epitopes, three CTL epitopes, and two B-cell epitopes, jointed via linkers and adjuvant, were used for the final vaccine construct design. Analysis disclosed that the developed vaccine demonstrated robust antigenic properties while proving soluble, stable, non-toxic, and non-allergenic. Additionally, molecular docking studies and molecular dynamics simulations confirmed a robust correlation between the designed vaccine and TLR-2 and TLR-4 immune receptors. The molecular docking results demonstrated a strong interaction between the newly developed vaccine and TLR2 (-895.1 kcal/mol) and TLR4 (-881.0 kcal/mol) receptors. During the simulation, the vaccine-TLR2 and vaccine-TLR4 complexes exhibited binding energies of -113.41 and -106.61 kcal/mol, respectively. Analysis by different bioinformatic tools indicated the potential of the designed vaccine in immune stimulation and a significant elevation in IgG and IgM antibodies, T-helper cells, T-cytotoxic cells, INF-γ, IL-2, and IL-4. Research findings show that the newly designed multi-epitope vaccine is promising in providing long-term immunity against GBM and offers a promising therapeutic alternative.
Collapse
Affiliation(s)
- Reza Salahlou
- Department of Medical Biotechnology, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
- Biotechnology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
- Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Safar Farajnia
- Biotechnology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
- Drug Applied Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Effat Alizadeh
- Department of Medical Biotechnology, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Siavoush Dastmalchi
- Biotechnology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
- Department of Medicinal Chemistry, School of Pharmacy, Tabriz University of Medical Sciences, Tabriz, Iran
- Faculty of Pharmacy, Near East University, P.O. Box 99138, Nicosia, North Cyprus, Mersin 10, Turkey
| | - Nasrin Bargahi
- Biotechnology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Leila Rahbarnia
- Infectious and Tropical Diseases Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | | |
Collapse
|
5
|
He C, Xiong P, Zeng C, Qiu X, Long T, Song H. Bibliometric analysis of the top 100 most cited articles on the immunotherapy for glioblastoma. Childs Nerv Syst 2024; 41:33. [PMID: 39633087 DOI: 10.1007/s00381-024-06693-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/25/2024] [Accepted: 11/24/2024] [Indexed: 12/07/2024]
Abstract
BACKGROUND The nature of immunotherapy has rendered it a focal point in the management of glioblastoma (GBM). This bibliometric analysis aimed to analyze the top 100 most cited articles about immunotherapy for GBM to expand the knowledge of research related to this therapeutic approach. METHODS The retrieval of the top 100 articles on "Immunotherapy AND Glioblastoma" was performed using the Web of Science Core Collection database. Relevant details were extracted for bibliometric analysis, and to gain insights, a comparison was made between older and newer articles. Categorical data underwent analysis utilizing Pearson's chi-square test, while continuous data were analyzed using the Wilcoxon rank-sum test. RESULTS The top 100 articles were distributed across the years 2000 to 2021, with the number of citations ranging from 135 to 1058. Among these articles, publications peaked in 2017 and 2018 (n = 12), and the most common country of correspondence was the USA (n = 73). Clinical Cancer Research was the most cited journal, and authors SAMPSON JH and LIM M had the highest number of papers (n = 6). Newer articles had significantly higher citation rates (p < 0.01), more authors (p < 0.05), more institutes (p < 0.01), and more collaborations between institutions (p < 0.01). Subspecialties showed a trend of more "TME" (1% vs 7%, p = 0.05945) in the before and after group comparison, although this difference was not statistically significant (p > 0.05). CONCLUSION The study of the top 100 cited articles on immunotherapy for GBM shows that researchers are actively working together to develop novel approaches to GBM immunotherapy. The high citations, leading countries, journals, and authors, along with evolving research characteristics, indicate a field with great interest and potential. Immunotherapy holds significant promise in GBM treatment.
Collapse
Affiliation(s)
- Chunming He
- Department of Neurosurgery, The First Affiliated Hospital of Gannan Medical University, West Jinling Road, Ganzhou, 341000, Jiangxi Province, China
| | - Peng Xiong
- The First Clinical Medical College of Gannan Medical University, Ganzhou City, 341000, Jiangxi Province, China
| | - Chuan Zeng
- The First Clinical Medical College of Gannan Medical University, Ganzhou City, 341000, Jiangxi Province, China
| | - Xinyu Qiu
- The First Clinical Medical College of Gannan Medical University, Ganzhou City, 341000, Jiangxi Province, China
| | - Tao Long
- The First Clinical Medical College of Gannan Medical University, Ganzhou City, 341000, Jiangxi Province, China
| | - Haimin Song
- Department of Neurosurgery, The First Affiliated Hospital of Gannan Medical University, West Jinling Road, Ganzhou, 341000, Jiangxi Province, China.
| |
Collapse
|
6
|
De Luca C, Virtuoso A, Papa M, Cirillo G, La Rocca G, Corvino S, Barbarisi M, Altieri R. The Three Pillars of Glioblastoma: A Systematic Review and Novel Analysis of Multi-Omics and Clinical Data. Cells 2024; 13:1754. [PMID: 39513861 PMCID: PMC11544881 DOI: 10.3390/cells13211754] [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: 09/14/2024] [Revised: 10/11/2024] [Accepted: 10/18/2024] [Indexed: 11/16/2024] Open
Abstract
Glioblastoma is the most fatal and common malignant brain tumor, excluding metastasis and with a median survival of approximately one year. While solid tumors benefit from newly approved drugs, immunotherapy, and prevention, none of these scenarios are opening for glioblastoma. The key to unlocking the peculiar features of glioblastoma is observing its molecular and anatomical features tightly entangled with the host's central nervous system (CNS). In June 2024, we searched the PUBMED electronic database. Data collection and analysis were conducted independently by two reviewers. Results: A total of 215 articles were identified, and 192 were excluded based on inclusion and exclusion criteria. The remaining 23 were used for collecting divergent molecular pathways and anatomical features of glioblastoma. The analysis of the selected papers revealed a multifaced tumor with extreme variability and cellular reprogramming that are observable within the same patient. All the variability of glioblastoma could be clustered into three pillars to dissect the physiology of the tumor: 1. necrotic core; 2. vascular proliferation; 3. CNS infiltration. These three pillars support glioblastoma survival, with a pivotal role of the neurovascular unit, as supported by the most recent paper published by experts in the field.
Collapse
Affiliation(s)
- Ciro De Luca
- Laboratory of Neuronal Networks Morphology and System Biology, Department of Mental and Physical Health and Preventive Medicine, University of Campania “Luigi Vanvitelli”, 80138 Naples, Italy; (A.V.); (M.P.); (G.C.)
| | - Assunta Virtuoso
- Laboratory of Neuronal Networks Morphology and System Biology, Department of Mental and Physical Health and Preventive Medicine, University of Campania “Luigi Vanvitelli”, 80138 Naples, Italy; (A.V.); (M.P.); (G.C.)
| | - Michele Papa
- Laboratory of Neuronal Networks Morphology and System Biology, Department of Mental and Physical Health and Preventive Medicine, University of Campania “Luigi Vanvitelli”, 80138 Naples, Italy; (A.V.); (M.P.); (G.C.)
- ISBE Italy, SYSBIO Centre of Systems Biology, 20126 Milan, Italy
| | - Giovanni Cirillo
- Laboratory of Neuronal Networks Morphology and System Biology, Department of Mental and Physical Health and Preventive Medicine, University of Campania “Luigi Vanvitelli”, 80138 Naples, Italy; (A.V.); (M.P.); (G.C.)
| | - Giuseppe La Rocca
- Department of Neurosurgery, Fondazione Policlinico Universitario “A. Gemelli” IRCCS, Catholic University of Rome School of Medicine, 00153 Rome, Italy;
| | - Sergio Corvino
- Department of Neurosciences and Reproductive and Odontostomatological Sciences, Neurosurgical Clinic, University “Federico II” of Naples, 80131 Naples, Italy;
| | - Manlio Barbarisi
- Multidisciplinary Department of Medical-Surgical and Dental Specialties, University of Campania “Luigi Vanvitelli”, 80131 Naples, Italy (R.A.)
| | - Roberto Altieri
- Multidisciplinary Department of Medical-Surgical and Dental Specialties, University of Campania “Luigi Vanvitelli”, 80131 Naples, Italy (R.A.)
| |
Collapse
|
7
|
Zhou Q, Zhang B, Xue C, Ren J, Zhang P, Ke X, Man J, Zhou J. Magnetic resonance imaging-based radiomics for predicting infiltration levels of CD68+ tumor-associated macrophages in glioblastomas. Strahlenther Onkol 2024:10.1007/s00066-024-02289-5. [PMID: 39269469 DOI: 10.1007/s00066-024-02289-5] [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: 12/21/2023] [Accepted: 07/29/2024] [Indexed: 09/15/2024]
Abstract
PURPOSE Tumor-associated macrophages (TAMs) are important biomarkers of tumor invasion and prognosis in patients with glioblastoma. We combined the imaging and radiomics features of preoperative MRI to predict CD68+ macrophage infiltration. METHODS Clinical, MRI image, and pathology data of 188 patients with glioblastoma were analyzed. Overall, 143 patients were included in the training (n = 101) and validation (n = 42) sets, whereas 45 patients were included in an independent test set. The optimal cut-off value (14.8%) was based on the minimum p-value formed by the Kaplan-Meier survival analysis and log-rank tests which divided patients into groups with high CD68+ TAMs (≥ 14.8%) and low CD68+ TAMs (< 14.8%). Regions of interest and radiomics features extraction were based on contrast-enhanced T1-weighted images (CE-T1WI) and T2WI. Multi-parameter stepwise regression was used to create the clinical, radiomics, and combined models, each evaluated using the receiver operating characteristic curve. Decision curve analysis was used to assess the clinical applicability of the nomogram. RESULTS A clinical model based on the minimum apparent diffusion coefficient (ADCmin) revealed an area under the curve (AUC) of 0.768, 0.764, and 0.624 for the training set, validation set, and test set, respectively. The 2D radiomics model, based on two features, revealed an AUC of 0.783, 0.724, and 0.789 for the training, validation, and test sets, respectively. The 3D radiomics model, based on three features, revealed AUCs of 0.823, 0.811, and 0.787 for the training, validation, and test sets, respectively. The combined model, with ADCmin and radiomics features, showed the best performance, with AUCs of 0.865, 0.822, and 0.776 for the training, validation, and test sets, respectively. The calibration curve of the combined model nomogram showed good agreement between the estimated and actual probabilities. CONCLUSION The combined model constructed using ADCmin, a quantitative imaging parameter, combined with five key radiomics features can be used to evaluate the extent of CD68+ macrophages before surgery.
Collapse
Affiliation(s)
- Qing Zhou
- Department of Radiology, Lanzhou University Second Hospital, Cuiyingmen No.82, Chengguan District, 730030, Lanzhou, Gansu, China
- Gansu International Scientific and Technological Cooperation Base of Medical Imaging Artificial Intelligence, Lanzhou, Gansu, China
| | - Bin Zhang
- Department of Radiology, Lanzhou University Second Hospital, Cuiyingmen No.82, Chengguan District, 730030, Lanzhou, Gansu, China
- Second Clinical School, Lanzhou University, Lanzhou, Gansu, China
- Gansu International Scientific and Technological Cooperation Base of Medical Imaging Artificial Intelligence, Lanzhou, Gansu, China
| | - Caiqiang Xue
- Department of Radiology, Lanzhou University Second Hospital, Cuiyingmen No.82, Chengguan District, 730030, Lanzhou, Gansu, China
- Second Clinical School, Lanzhou University, Lanzhou, Gansu, China
- Gansu International Scientific and Technological Cooperation Base of Medical Imaging Artificial Intelligence, Lanzhou, Gansu, China
| | | | - Peng Zhang
- Second Clinical School, Lanzhou University, Lanzhou, Gansu, China
- Department of Pathology, Lanzhou University Second Hospital, Lanzhou, Gansu, China
| | - Xiaoai Ke
- Department of Radiology, Lanzhou University Second Hospital, Cuiyingmen No.82, Chengguan District, 730030, Lanzhou, Gansu, China
- Gansu International Scientific and Technological Cooperation Base of Medical Imaging Artificial Intelligence, Lanzhou, Gansu, China
| | - Jiangwei Man
- Second Clinical School, Lanzhou University, Lanzhou, Gansu, China
- Department of Surgical, Lanzhou University Second Hospital, Lanzhou, Gansu, China
| | - Junlin Zhou
- Department of Radiology, Lanzhou University Second Hospital, Cuiyingmen No.82, Chengguan District, 730030, Lanzhou, Gansu, China.
- Second Clinical School, Lanzhou University, Lanzhou, Gansu, China.
- Gansu International Scientific and Technological Cooperation Base of Medical Imaging Artificial Intelligence, Lanzhou, Gansu, China.
| |
Collapse
|
8
|
Huang Z, Huang J, Lin Y, Deng Y, Yang L, Zhang X, Huang H, Sun Q, Liu H, Liang H, Lv Z, He B, Hu F. Construction and validation of a TAMRGs prognostic signature for gliomas by integrated analysis of scRNA and bulk RNA sequencing data. Brain Res 2024; 1846:149237. [PMID: 39270996 DOI: 10.1016/j.brainres.2024.149237] [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: 05/31/2024] [Revised: 09/06/2024] [Accepted: 09/10/2024] [Indexed: 09/15/2024]
Abstract
BACKGROUND This study aimed to construct and validate a prognostic model based on tumor associated macrophage-related genes (TAMRGs) by integrating single-cell RNA sequencing (scRNA-seq) and bulk RNA sequencing (bulk RNA-seq) data. METHODS The scRNA-seq data of three inhouse glioma tissues were used to identify the tumor-associated macrophages (TAMs) marker genes, the DEGs from the The Cancer Genome Atlas (TCGA) - Genotype-Tissue Expression (GTEx) dataset were used to further select TAMs marker genes. Subsequently, a TAMRG-score was constructed by Least absolute shrinkage and selection operator (LASSO) regression and multivariate Cox regression analysis in the TCGA dataset and validated in the Chinese Glioma Genome Atlas (CGGA) dataset. RESULTS We identified 186 TAMs marker genes, and a total of 6 optimal prognostic genes including CKS2, LITAF, CTSB, TWISTNB, PPIF and G0S2 were selected to construct a TAMRG-score. The high TAMRG-score was significantly associated with worse prognosis (log-rank test, P<0.001). Moreover, the TAMRG-score outperformed the other three models with AUC of 0.808. Immune cell infiltration, TME scores, immune checkpoints, TMB and drug susceptibility were significantly different between TAMRG-score groups. In addition, a nomogram were constructed by combing the TAMRG-score and clinical information (Age, Grade, IDH mutation and 1p19q codeletion) to predict the survival of glioma patients with AUC of 0.909 for 1-year survival. CONCLUSION The high TAMRG-score group was associated with a poor prognosis. A nomogram by incorporating TMARG-score could precisely predict glioma survival, and provide evidence for personalized treatment of glioma.
Collapse
Affiliation(s)
- Zhicong Huang
- Department of Epidemiology and Health Statistics, Fujian Provincial Key Laboratory of Environment Factors and Cancer, School of Public Health, Fujian Medical University, Fujian, PR China
| | - Jingyao Huang
- Department of Epidemiology and Health Statistics, Fujian Provincial Key Laboratory of Environment Factors and Cancer, School of Public Health, Fujian Medical University, Fujian, PR China
| | - Ying Lin
- Department of Epidemiology and Health Statistics, Fujian Provincial Key Laboratory of Environment Factors and Cancer, School of Public Health, Fujian Medical University, Fujian, PR China
| | - Ying Deng
- Department of Epidemiology and Health Statistics, Fujian Provincial Key Laboratory of Environment Factors and Cancer, School of Public Health, Fujian Medical University, Fujian, PR China
| | - Longkun Yang
- Department of Epidemiology and Health Statistics, Fujian Provincial Key Laboratory of Environment Factors and Cancer, School of Public Health, Fujian Medical University, Fujian, PR China
| | - Xing Zhang
- Department of Epidemiology and Health Statistics, Fujian Provincial Key Laboratory of Environment Factors and Cancer, School of Public Health, Fujian Medical University, Fujian, PR China
| | - Hao Huang
- Department of Epidemiology, School of Public Health, Shenzhen University Medical School, Shenzhen, Guangdong, PR China
| | - Qian Sun
- Department of Neurosurgery, The Tumor Hospital Affiliated to Harbin Medical University, Harbin, Heilongjiang, PR China
| | - Hui Liu
- Department of Neurosurgery, The Tumor Hospital Affiliated to Harbin Medical University, Harbin, Heilongjiang, PR China
| | - Hongsheng Liang
- Department of Neurosurgery, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, PR China
| | - Zhonghua Lv
- Department of Neurosurgery, The Tumor Hospital Affiliated to Harbin Medical University, Harbin, Heilongjiang, PR China.
| | - Baochang He
- Department of Epidemiology and Health Statistics, Fujian Provincial Key Laboratory of Environment Factors and Cancer, School of Public Health, Fujian Medical University, Fujian, PR China.
| | - Fulan Hu
- Department of Epidemiology, School of Public Health, Shenzhen University Medical School, Shenzhen, Guangdong, PR China.
| |
Collapse
|
9
|
Hagemeyer H, Hellwinkel OJC, Plata-Bello J. Zonulin as Gatekeeper in Gut-Brain Axis: Dysregulation in Glioblastoma. Biomedicines 2024; 12:1649. [PMID: 39200114 PMCID: PMC11352073 DOI: 10.3390/biomedicines12081649] [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: 06/24/2024] [Revised: 07/18/2024] [Accepted: 07/21/2024] [Indexed: 09/01/2024] Open
Abstract
Novel biomarkers and therapeutic strategies for glioblastoma, the most common malignant brain tumor with an extremely unfavorable prognosis, are urgently needed. Recent studies revealed a significant upregulation of the protein zonulin in glioblastoma, which correlates with patient survival. Originally identified as pre-haptoglobin-2, zonulin modulates both the intestinal barrier and the blood-brain barrier by disassembling tight junctions. An association of zonulin with various neuroinflammatory diseases has been observed. It can be suggested that zonulin links a putative impairment of the gut-brain barrier with glioblastoma carcinogenesis, leading to an interaction of the gut microbiome, the immune system, and glioblastoma. We therefore propose three interconnected hypotheses: (I) elevated levels of zonulin in glioblastoma contribute to its aggressiveness; (II) upregulated (serum-) zonulin increases the permeability of the microbiota-gut-brain barrier; and (III) this creates a carcinogenic and immunosuppressive microenvironment preventing the host from an effective antitumor response. The role of zonulin in glioblastoma highlights a promising field of research that could yield diagnostic and therapeutic options for glioblastoma patients and other diseases with a disturbed microbiota-gut-brain barrier.
Collapse
Affiliation(s)
- Hannah Hagemeyer
- Institut für Neuroimmunologie und Multiple Sklerose, University Medical Center Hamburg-Eppendorf, Falkenried 94, 20251 Hamburg, Germany;
| | - Olaf J. C. Hellwinkel
- Department of Forensic Medicine, University Medical Center Hamburg-Eppendorf, Martinistraße 52, 20251 Hamburg, Germany
| | - Julio Plata-Bello
- Department of Neurosurgery, Hospital Universitario de Canarias, S/C de Tenerife, 38320 La Laguna, Spain
| |
Collapse
|
10
|
Pan T, Xie DK, Li J, Qiang YJ, Fan SY, Wang TT, Han YY, Zang J, Yang Y, Zhao JL, Li SZ, Wu S. Glioma-Stem-Cell-Derived Exosomes Remodeled Glioma-Associated Macrophage via NEAT1/miR-125a/STAT3 Pathway. Cancers (Basel) 2024; 16:2500. [PMID: 39061140 PMCID: PMC11274466 DOI: 10.3390/cancers16142500] [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: 05/27/2024] [Accepted: 06/06/2024] [Indexed: 07/28/2024] Open
Abstract
Glioblastoma (GBM), as the most common primary brain tumor, usually results in an extremely poor prognosis, in which glioma stem cells (GSCs) and their immunosuppressive microenvironment prominently intervene in the resistance to radiotherapy and chemotherapy that directly leads to tumor recurrence and shortened survival time. The specific mechanism through which exosomes generated from GSCs support the creation of an immunosuppressive microenvironment remains unknown, while it is acknowledged to be engaged in intercellular communication and the regulation of the glioma immunosuppressive microenvironment. The elevated expression of LncRNA-NEAT1 was found in glioma cells after radiotherapy, chemotherapy, and DNA damage stimulation, and NEAT1 could promote the malignant biological activities of GSCs. Emerging evidence suggests that lncRNAs may reply to external stimuli or DNA damage by playing a role in modulating different aspects of tumor biology. Our study demonstrated a promotive role of the carried NEAT1 by GSC-derived exosomes in the polarization of M2-like macrophages. Further experiments demonstrated the mediative role of miR-125a and its target gene STAT3 in NEAT1-induced polarization of M2-like macrophages that promote glioma progression. Our findings elucidate the mechanism by which GSCs influence the polarization of M2-like macrophages through exosomes, which may contribute to the formation of immunosuppressive microenvironments. Taken together, our study reveals the miR-125a-STAT3 pathway through which exosomal NEAT1 from treatment-resistant GSCs contributes to M2-like macrophage polarization, indicating the potential of exosomal NEAT1 for treating glioma.
Collapse
Affiliation(s)
- Tong Pan
- Department of Neurosurgery, Xijing Hospital, Air Force Medical University, Xi’an 710032, China; (T.P.); (J.L.); (Y.-J.Q.); (T.-T.W.); (Y.-Y.H.)
- Department of Biochemistry and Molecular Biology, Air Force Medical University, Xi’an 710032, China
| | - Dong-Kun Xie
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, Faculty of Life Sciences, Northwest University, Xi’an 710069, China; (D.-K.X.); (Y.Y.)
- State Key Laboratory of Cancer Biology, Department of Medical Genetics and Developmental Biology, Air Force Medical University, Xi’an 710032, China;
| | - Juan Li
- Department of Neurosurgery, Xijing Hospital, Air Force Medical University, Xi’an 710032, China; (T.P.); (J.L.); (Y.-J.Q.); (T.-T.W.); (Y.-Y.H.)
| | - Yu-Jie Qiang
- Department of Neurosurgery, Xijing Hospital, Air Force Medical University, Xi’an 710032, China; (T.P.); (J.L.); (Y.-J.Q.); (T.-T.W.); (Y.-Y.H.)
| | - Song-Yuan Fan
- Department of Neurosurgery, The Air Force Hospital of Central Theater of PLA, Datong 037000, China;
| | - Ting-Ting Wang
- Department of Neurosurgery, Xijing Hospital, Air Force Medical University, Xi’an 710032, China; (T.P.); (J.L.); (Y.-J.Q.); (T.-T.W.); (Y.-Y.H.)
| | - Yuan-Yuan Han
- Department of Neurosurgery, Xijing Hospital, Air Force Medical University, Xi’an 710032, China; (T.P.); (J.L.); (Y.-J.Q.); (T.-T.W.); (Y.-Y.H.)
| | - Jian Zang
- Department of Radiotherapy, Xijing Hospital, Air Force Medical University, Xi’an 710032, China;
| | - Yang Yang
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, Faculty of Life Sciences, Northwest University, Xi’an 710069, China; (D.-K.X.); (Y.Y.)
| | - Jun-Long Zhao
- State Key Laboratory of Cancer Biology, Department of Medical Genetics and Developmental Biology, Air Force Medical University, Xi’an 710032, China;
| | - San-Zhong Li
- Department of Neurosurgery, Xijing Hospital, Air Force Medical University, Xi’an 710032, China; (T.P.); (J.L.); (Y.-J.Q.); (T.-T.W.); (Y.-Y.H.)
| | - Shuang Wu
- Department of Neurosurgery, Xijing Hospital, Air Force Medical University, Xi’an 710032, China; (T.P.); (J.L.); (Y.-J.Q.); (T.-T.W.); (Y.-Y.H.)
- Department of Neurosurgery, The Air Force Hospital of Central Theater of PLA, Datong 037000, China;
| |
Collapse
|
11
|
Coupey J, Leblond MM, Hue ES, Valable S. Flow cytometry detection and quantification of circulating leukocyte subpopulations in mice after brain irradiation. Methods Cell Biol 2024; 189:135-152. [PMID: 39393880 DOI: 10.1016/bs.mcb.2024.06.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/13/2024]
Abstract
In the context of high-grade gliomas such as glioblastoma (GBM), the immune part of the tumor microenvironment (TME) is involved in tumor growth and tumor recurrence. It is mostly represented by high amount of macrophages and low amount of lymphocytes. GBM in itself as well as x-ray-based radiotherapy, a standard treatment for brain tumors, are also associated with systemic effects like lymphopenia that correlates with a poor prognosis. This contributes to the immune-suppressive nature of the TME and may explain the lack of the anti-tumor immune response. Radiation-induced lymphopenia (RIL) is generally evaluated on CD4+ and CD8+ count or on a CBC (complete blood count), but the heterogeneity of the subtypes prompts us to explore them in detail to better understand the cellular response to brain irradiation. To facilitate and develop the evaluation of x-ray brain exposure on circulating immune cells, we developed a reproducible and reliable method to quantify the variation of lymphoid and myeloid subtypes using flow cytometry after brain irradiation in the rodent.
Collapse
Affiliation(s)
- Julie Coupey
- Université de Caen Normandie, CNRS, Normandie Université, ISTCT UMR6030, GIP Cyceron, Caen, France
| | - Marine M Leblond
- Department of Oncology UNIL CHUV, University of Lausanne, Lausanne, Switzerland; Ludwig Institute for Cancer Research, University of Lausanne, Lausanne, Switzerland
| | - Erika S Hue
- LABÉO, Saint-Contest, France; Normandie Université, UNICAEN BIOTARGEN, Saint-Contest, France
| | - Samuel Valable
- Université de Caen Normandie, CNRS, Normandie Université, ISTCT UMR6030, GIP Cyceron, Caen, France.
| |
Collapse
|
12
|
Nusraty S, Boddeti U, Zaghloul KA, Brown DA. Microglia in Glioblastomas: Molecular Insight and Immunotherapeutic Potential. Cancers (Basel) 2024; 16:1972. [PMID: 38893093 PMCID: PMC11171200 DOI: 10.3390/cancers16111972] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2024] [Revised: 05/18/2024] [Accepted: 05/20/2024] [Indexed: 06/21/2024] Open
Abstract
Glioblastoma (GBM) is one of the most aggressive and devastating primary brain tumors, with a median survival of 15 months following diagnosis. Despite the intense treatment regimen which routinely includes maximal safe neurosurgical resection followed by adjuvant radio- and chemotherapy, the disease remains uniformly fatal. The poor prognosis associated with GBM is multifactorial owing to factors such as increased proliferation, angiogenesis, and metabolic switching to glycolytic pathways. Critically, GBM-mediated local and systemic immunosuppression result in inadequate immune surveillance and ultimately, tumor-immune escape. Microglia-the resident macrophages of the central nervous system (CNS)-play crucial roles in mediating the local immune response in the brain. Depending on the specific pathological cues, microglia are activated into either a pro-inflammatory, neurotoxic phenotype, known as M1, or an anti-inflammatory, regenerative phenotype, known as M2. In either case, microglia secrete corresponding pro- or anti-inflammatory cytokines and chemokines that either promote or hinder tumor growth. Herein, we review the interplay between GBM cells and resident microglia with a focus on contemporary studies highlighting the effect of GBM on the subtypes of microglia expressed, the associated cytokines/chemokines secreted, and ultimately, their impact on tumor pathogenesis. Finally, we explore how understanding the intricacies of the tumor-immune landscape can inform novel immunotherapeutic strategies against this devastating disease.
Collapse
Affiliation(s)
| | | | | | - Desmond A. Brown
- Surgical Neurology Branch, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD 20892, USA; (S.N.); (U.B.); (K.A.Z.)
| |
Collapse
|
13
|
Haley MJ, Bere L, Minshull J, Georgaka S, Garcia-Martin N, Howell G, Coope DJ, Roncaroli F, King A, Wedge DC, Allan SM, Pathmanaban ON, Brough D, Couper KN. Hypoxia coordinates the spatial landscape of myeloid cells within glioblastoma to affect survival. SCIENCE ADVANCES 2024; 10:eadj3301. [PMID: 38758780 PMCID: PMC11100569 DOI: 10.1126/sciadv.adj3301] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/30/2023] [Accepted: 04/15/2024] [Indexed: 05/19/2024]
Abstract
Myeloid cells are highly prevalent in glioblastoma (GBM), existing in a spectrum of phenotypic and activation states. We now have limited knowledge of the tumor microenvironment (TME) determinants that influence the localization and the functions of the diverse myeloid cell populations in GBM. Here, we have utilized orthogonal imaging mass cytometry with single-cell and spatial transcriptomic approaches to identify and map the various myeloid populations in the human GBM tumor microenvironment (TME). Our results show that different myeloid populations have distinct and reproducible compartmentalization patterns in the GBM TME that is driven by tissue hypoxia, regional chemokine signaling, and varied homotypic and heterotypic cellular interactions. We subsequently identified specific tumor subregions in GBM, based on composition of identified myeloid cell populations, that were linked to patient survival. Our results provide insight into the spatial organization of myeloid cell subpopulations in GBM, and how this is predictive of clinical outcome.
Collapse
Affiliation(s)
- Michael J. Haley
- Geoffrey Jefferson Brain Research Centre, Manchester Academic Health Science Centre, Northern Care Alliance NHS Foundation Trust, University of Manchester, Manchester, UK
- Lydia Becker Institute of Inflammation and Immunology, University of Manchester, Manchester, UK
| | - Leoma Bere
- Geoffrey Jefferson Brain Research Centre, Manchester Academic Health Science Centre, Northern Care Alliance NHS Foundation Trust, University of Manchester, Manchester, UK
- Lydia Becker Institute of Inflammation and Immunology, University of Manchester, Manchester, UK
| | - James Minshull
- Geoffrey Jefferson Brain Research Centre, Manchester Academic Health Science Centre, Northern Care Alliance NHS Foundation Trust, University of Manchester, Manchester, UK
- Division of Neuroscience, University of Manchester, Manchester, UK
| | - Sokratia Georgaka
- Division of Informatics, Imaging and Data Sciences, University of Manchester, Manchester, UK
| | | | - Gareth Howell
- Flow Cytometry Core Research Facility, University of Manchester, Manchester, UK
| | - David J. Coope
- Geoffrey Jefferson Brain Research Centre, Manchester Academic Health Science Centre, Northern Care Alliance NHS Foundation Trust, University of Manchester, Manchester, UK
- Division of Neuroscience, University of Manchester, Manchester, UK
- Manchester Centre for Clinical Neurosciences, Manchester, UK
| | - Federico Roncaroli
- Geoffrey Jefferson Brain Research Centre, Manchester Academic Health Science Centre, Northern Care Alliance NHS Foundation Trust, University of Manchester, Manchester, UK
- Division of Neuroscience, University of Manchester, Manchester, UK
- Manchester Centre for Clinical Neurosciences, Manchester, UK
| | - Andrew King
- Geoffrey Jefferson Brain Research Centre, Manchester Academic Health Science Centre, Northern Care Alliance NHS Foundation Trust, University of Manchester, Manchester, UK
- Manchester Centre for Clinical Neurosciences, Manchester, UK
- Division of Cardiovascular Sciences, University of Manchester, Manchester, UK
| | - David C. Wedge
- Manchester Cancer Research Centre, University of Manchester, Manchester, UK
| | - Stuart M. Allan
- Geoffrey Jefferson Brain Research Centre, Manchester Academic Health Science Centre, Northern Care Alliance NHS Foundation Trust, University of Manchester, Manchester, UK
- Division of Neuroscience, University of Manchester, Manchester, UK
| | - Omar N. Pathmanaban
- Geoffrey Jefferson Brain Research Centre, Manchester Academic Health Science Centre, Northern Care Alliance NHS Foundation Trust, University of Manchester, Manchester, UK
- Division of Neuroscience, University of Manchester, Manchester, UK
- Manchester Centre for Clinical Neurosciences, Manchester, UK
| | - David Brough
- Geoffrey Jefferson Brain Research Centre, Manchester Academic Health Science Centre, Northern Care Alliance NHS Foundation Trust, University of Manchester, Manchester, UK
- Lydia Becker Institute of Inflammation and Immunology, University of Manchester, Manchester, UK
- Division of Neuroscience, University of Manchester, Manchester, UK
| | - Kevin N. Couper
- Geoffrey Jefferson Brain Research Centre, Manchester Academic Health Science Centre, Northern Care Alliance NHS Foundation Trust, University of Manchester, Manchester, UK
- Lydia Becker Institute of Inflammation and Immunology, University of Manchester, Manchester, UK
| |
Collapse
|
14
|
Tang W, Lo CWS, Ma W, Chu ATW, Tong AHY, Chung BHY. Revealing the role of SPP1 + macrophages in glioma prognosis and therapeutic targeting by investigating tumor-associated macrophage landscape in grade 2 and 3 gliomas. Cell Biosci 2024; 14:37. [PMID: 38515213 PMCID: PMC10956315 DOI: 10.1186/s13578-024-01218-4] [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: 10/27/2023] [Accepted: 03/13/2024] [Indexed: 03/23/2024] Open
Abstract
BACKGROUND Glioma is a highly heterogeneous brain tumor categorized into World Health Organization (WHO) grades 1-4 based on its malignancy. The suppressive immune microenvironment of glioma contributes significantly to unfavourable patient outcomes. However, the cellular composition and their complex interplays within the glioma environment remain poorly understood, and reliable prognostic markers remain elusive. Therefore, in-depth exploration of the tumor microenvironment (TME) and identification of predictive markers are crucial for improving the clinical management of glioma patients. RESULTS Our analysis of single-cell RNA-sequencing data from glioma samples unveiled the immunosuppressive role of tumor-associated macrophages (TAMs), mediated through intricate interactions with tumor cells and lymphocytes. We also discovered the heterogeneity within TAMs, among which a group of suppressive TAMs named TAM-SPP1 demonstrated a significant association with Epidermal Growth Factor Receptor (EGFR) amplification, impaired T cell response and unfavourable patient survival outcomes. Furthermore, by leveraging genomic and transcriptomic data from The Cancer Genome Atlas (TCGA) dataset, two distinct molecular subtypes with a different constitution of TAMs, EGFR status and clinical outcomes were identified. Exploiting the molecular differences between these two subtypes, we developed a four-gene-based prognostic model. This model displayed strong associations with an elevated level of suppressive TAMs and could be used to predict anti-tumor immune response and prognosis in glioma patients. CONCLUSION Our findings illuminated the molecular and cellular mechanisms that shape the immunosuppressive microenvironment in gliomas, providing novel insights into potential therapeutic targets. Furthermore, the developed prognostic model holds promise for predicting immunotherapy response and assisting in more precise risk stratification for glioma patients.
Collapse
Affiliation(s)
- Wenshu Tang
- Hong Kong Genome Institute, 2/F, Building 20E, Hong Kong Science Park, Hong Kong, China
| | - Cario W S Lo
- Hong Kong Genome Institute, 2/F, Building 20E, Hong Kong Science Park, Hong Kong, China
| | - Wei Ma
- Hong Kong Genome Institute, 2/F, Building 20E, Hong Kong Science Park, Hong Kong, China
| | - Annie T W Chu
- Hong Kong Genome Institute, 2/F, Building 20E, Hong Kong Science Park, Hong Kong, China
| | - Amy H Y Tong
- Hong Kong Genome Institute, 2/F, Building 20E, Hong Kong Science Park, Hong Kong, China
| | - Brian H Y Chung
- Hong Kong Genome Institute, 2/F, Building 20E, Hong Kong Science Park, Hong Kong, China.
- Department of Pediatrics and Adolescent Medicine, School of Clinical Medicine, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong, China.
| |
Collapse
|
15
|
Guo X, Zhang H, He C, Qin K, Lai Q, Fang Y, Chen Q, Li W, Wang Y, Wang X, Li A, Liu S, Li Q. RUNX1 promotes angiogenesis in colorectal cancer by regulating the crosstalk between tumor cells and tumor associated macrophages. Biomark Res 2024; 12:29. [PMID: 38419056 PMCID: PMC10903076 DOI: 10.1186/s40364-024-00573-1] [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: 03/19/2023] [Accepted: 02/06/2024] [Indexed: 03/02/2024] Open
Abstract
Colorectal cancer (CRC) is a common malignancy worldwide. Angiogenesis and metastasis are the critical hallmarks of malignant tumor. Runt-related transcription factor 1 (RUNX1), an efficient transcription factor, facilitates CRC proliferation, metastasis and chemotherapy resistance. We aimed to investigate the RUNX1 mediated crosstalk between tumor cells and M2 polarized tumor associated macrophages (TAMs) in CRC, as well as its relationship with neoplastic angiogenesis. We found that RUNX1 recruited macrophages and induced M2 polarized TAMs in CRC by promoting the production of chemokine 2 (CCL2) and the activation of Hedgehog pathway. In addition, we found that the M2 macrophage-specific generated cytokine, platelet-derived growth factor (PDGF)-BB, promoted vessel formation both in vitro and vivo. PDGF-BB was also found to enhance the expression of RUNX1 in CRC cell lines, and promote its migration and invasion in vitro. A positive feedback loop of RUNX1 and PDGF-BB was thus formed. In conclusion, our data suggest that RUNX1 promotes CRC angiogenesis by regulating M2 macrophages during the complex crosstalk between tumor cells and TAMs. This observation provides a potential combined therapy strategy targeting RUNX1 and TAMs-related PDGF-BB in CRC.
Collapse
Affiliation(s)
- Xuxue Guo
- Guangdong Provincial Key Laboratory of Gastroenterology, Department of Gastroenterology, Nanfang Hospital, Southern Medical University, No. 1838, Guangzhou Avenue North, Guangzhou, 510515, People's Republic of China
- Department of Gastroenterology, the Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Haonan Zhang
- Guangdong Provincial Key Laboratory of Gastroenterology, Department of Gastroenterology, Nanfang Hospital, Southern Medical University, No. 1838, Guangzhou Avenue North, Guangzhou, 510515, People's Republic of China
- Department of Gastroenterology, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Chengcheng He
- Guangdong Provincial Key Laboratory of Gastroenterology, Department of Gastroenterology, Nanfang Hospital, Southern Medical University, No. 1838, Guangzhou Avenue North, Guangzhou, 510515, People's Republic of China
- Department of Gastroenterology, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Kaiwen Qin
- Guangdong Provincial Key Laboratory of Gastroenterology, Department of Gastroenterology, Nanfang Hospital, Southern Medical University, No. 1838, Guangzhou Avenue North, Guangzhou, 510515, People's Republic of China
- The First School of Clinical Medicine), Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Qiuhua Lai
- Guangdong Provincial Key Laboratory of Gastroenterology, Department of Gastroenterology, Nanfang Hospital, Southern Medical University, No. 1838, Guangzhou Avenue North, Guangzhou, 510515, People's Republic of China
| | - Yuxin Fang
- Guangdong Provincial Key Laboratory of Gastroenterology, Department of Gastroenterology, Nanfang Hospital, Southern Medical University, No. 1838, Guangzhou Avenue North, Guangzhou, 510515, People's Republic of China
| | - Qianhui Chen
- State Key Laboratory of Organ Failure Research, Guangdong Provincial Key Laboratory of Viral Hepatitis Research, Department of Hepatology Unit and Infectious Diseases, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Weize Li
- The First School of Clinical Medicine), Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Yiqing Wang
- Department of Pathology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
- Department of Pathology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, Guangdong, China
| | - Xinke Wang
- Guangdong Provincial Key Laboratory of Gastroenterology, Department of Gastroenterology, Nanfang Hospital, Southern Medical University, No. 1838, Guangzhou Avenue North, Guangzhou, 510515, People's Republic of China
| | - Aimin Li
- Guangdong Provincial Key Laboratory of Gastroenterology, Department of Gastroenterology, Nanfang Hospital, Southern Medical University, No. 1838, Guangzhou Avenue North, Guangzhou, 510515, People's Republic of China
| | - Side Liu
- Guangdong Provincial Key Laboratory of Gastroenterology, Department of Gastroenterology, Nanfang Hospital, Southern Medical University, No. 1838, Guangzhou Avenue North, Guangzhou, 510515, People's Republic of China.
- Pazhou Lab, Guangzhou, Guangdong, China.
| | - Qingyuan Li
- Guangdong Provincial Key Laboratory of Gastroenterology, Department of Gastroenterology, Nanfang Hospital, Southern Medical University, No. 1838, Guangzhou Avenue North, Guangzhou, 510515, People's Republic of China.
| |
Collapse
|
16
|
Stepanenko AA, Sosnovtseva AO, Valikhov MP, Chernysheva AA, Abramova OV, Pavlov KA, Chekhonin VP. Systemic and local immunosuppression in glioblastoma and its prognostic significance. Front Immunol 2024; 15:1326753. [PMID: 38481999 PMCID: PMC10932993 DOI: 10.3389/fimmu.2024.1326753] [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: 10/23/2023] [Accepted: 02/06/2024] [Indexed: 04/07/2024] Open
Abstract
The effectiveness of tumor therapy, especially immunotherapy and oncolytic virotherapy, critically depends on the activity of the host immune cells. However, various local and systemic mechanisms of immunosuppression operate in cancer patients. Tumor-associated immunosuppression involves deregulation of many components of immunity, including a decrease in the number of T lymphocytes (lymphopenia), an increase in the levels or ratios of circulating and tumor-infiltrating immunosuppressive subsets [e.g., macrophages, microglia, myeloid-derived suppressor cells (MDSCs), and regulatory T cells (Tregs)], as well as defective functions of subsets of antigen-presenting, helper and effector immune cell due to altered expression of various soluble and membrane proteins (receptors, costimulatory molecules, and cytokines). In this review, we specifically focus on data from patients with glioblastoma/glioma before standard chemoradiotherapy. We discuss glioblastoma-related immunosuppression at baseline and the prognostic significance of different subsets of circulating and tumor-infiltrating immune cells (lymphocytes, CD4+ and CD8+ T cells, Tregs, natural killer (NK) cells, neutrophils, macrophages, MDSCs, and dendritic cells), including neutrophil-to-lymphocyte ratio (NLR), focus on the immune landscape and prognostic significance of isocitrate dehydrogenase (IDH)-mutant gliomas, proneural, classical and mesenchymal molecular subtypes, and highlight the features of immune surveillance in the brain. All attempts to identify a reliable prognostic immune marker in glioblastoma tissue have led to contradictory results, which can be explained, among other things, by the unprecedented level of spatial heterogeneity of the immune infiltrate and the significant phenotypic diversity and (dys)functional states of immune subpopulations. High NLR is one of the most repeatedly confirmed independent prognostic factors for shorter overall survival in patients with glioblastoma and carcinoma, and its combination with other markers of the immune response or systemic inflammation significantly improves the accuracy of prediction; however, more prospective studies are needed to confirm the prognostic/predictive power of NLR. We call for the inclusion of dynamic assessment of NLR and other blood inflammatory markers (e.g., absolute/total lymphocyte count, platelet-to-lymphocyte ratio, lymphocyte-to-monocyte ratio, systemic immune-inflammation index, and systemic immune response index) in all neuro-oncology studies for rigorous evaluation and comparison of their individual and combinatorial prognostic/predictive significance and relative superiority.
Collapse
Affiliation(s)
- Aleksei A. Stepanenko
- Department of Fundamental and Applied Neurobiology, V. P. Serbsky National Medical Research Center of Psychiatry and Narcology, the Ministry of Health of the Russian Federation, Moscow, Russia
- Department of Medical Nanobiotechnology, Institute of Translational Medicine, N. I. Pirogov Russian National Research Medical University, The Ministry of Health of the Russian Federation, Moscow, Russia
| | - Anastasiia O. Sosnovtseva
- Department of Fundamental and Applied Neurobiology, V. P. Serbsky National Medical Research Center of Psychiatry and Narcology, the Ministry of Health of the Russian Federation, Moscow, Russia
- Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Moscow, Russia
| | - Marat P. Valikhov
- Department of Fundamental and Applied Neurobiology, V. P. Serbsky National Medical Research Center of Psychiatry and Narcology, the Ministry of Health of the Russian Federation, Moscow, Russia
- Department of Medical Nanobiotechnology, Institute of Translational Medicine, N. I. Pirogov Russian National Research Medical University, The Ministry of Health of the Russian Federation, Moscow, Russia
| | - Anastasia A. Chernysheva
- Department of Fundamental and Applied Neurobiology, V. P. Serbsky National Medical Research Center of Psychiatry and Narcology, the Ministry of Health of the Russian Federation, Moscow, Russia
| | - Olga V. Abramova
- Department of Fundamental and Applied Neurobiology, V. P. Serbsky National Medical Research Center of Psychiatry and Narcology, the Ministry of Health of the Russian Federation, Moscow, Russia
| | - Konstantin A. Pavlov
- Department of Fundamental and Applied Neurobiology, V. P. Serbsky National Medical Research Center of Psychiatry and Narcology, the Ministry of Health of the Russian Federation, Moscow, Russia
| | - Vladimir P. Chekhonin
- Department of Fundamental and Applied Neurobiology, V. P. Serbsky National Medical Research Center of Psychiatry and Narcology, the Ministry of Health of the Russian Federation, Moscow, Russia
- Department of Medical Nanobiotechnology, Institute of Translational Medicine, N. I. Pirogov Russian National Research Medical University, The Ministry of Health of the Russian Federation, Moscow, Russia
| |
Collapse
|
17
|
Wang H, Yang J, Li X, Zhao H. Current state of immune checkpoints therapy for glioblastoma. Heliyon 2024; 10:e24729. [PMID: 38298707 PMCID: PMC10828821 DOI: 10.1016/j.heliyon.2024.e24729] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2023] [Revised: 01/12/2024] [Accepted: 01/12/2024] [Indexed: 02/02/2024] Open
Abstract
Glioblastoma (GBM), one of the most aggressive forms of brain cancer, has limited treatment options. Recent years have witnessed the remarkable success of checkpoint inhibitor immunotherapy across various cancer types. Against this backdrop, several clinical trials investigating checkpoint inhibitors for GBM are underway in multiple countries. Furthermore, the integration of immunotherapy with traditional treatment approaches is now emerging as a highly promising strategy. This review summarizes the latest advancements in checkpoint inhibitor immunotherapy for GBM treatment. We provide a concise yet comprehensive overview of current GBM immunotherapy options. Additionally, this review underscores combination strategies and potential biomarkers for predicting response and resistance in GBM immunotherapies.
Collapse
Affiliation(s)
- He Wang
- Department of Neurosurgery, the Affiliated Hospital of Qingdao University, No. 16 Jiangsu Road, Qingdao, Shandong, 266005, China
| | - Jing Yang
- Department of Emergency Surgery, the Affiliated Hospital of Qingdao University, No. 16 Jiangsu Road, Qingdao, Shandong, 266005, China
| | - Xiangjun Li
- School of medicine, Department of Breast surgery, the Affiliated Hospital of Qingdao University, Qingdao University, Qingdao, Shandong, 266000, China
| | - Hai Zhao
- Department of Neurosurgery, the Affiliated Hospital of Qingdao University, No. 16 Jiangsu Road, Qingdao, Shandong, 266005, China
| |
Collapse
|
18
|
Zeng C, Zhang C, He C, Song H. Investigating the causal impact of gut microbiota on glioblastoma: a bidirectional Mendelian randomization study. BMC Genomics 2023; 24:784. [PMID: 38110895 PMCID: PMC10726622 DOI: 10.1186/s12864-023-09885-2] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2023] [Accepted: 12/08/2023] [Indexed: 12/20/2023] Open
Abstract
BACKGROUND Currently, the influence of microbiota on the occurrence, progression, and treatment of cancer is a topic of considerable research interest. Therefore, based on the theory of the gut-brain axis proved by previous studies, our objective was to uncover the causal relationship between glioblastoma and the gut microbiome using Mendelian randomization analysis. METHODS We conducted a bidirectional Mendelian randomization study using summary statistics of gut microbiota derived from the MiBioGen consortium, the largest database of gut microbiota. Summary statistics for glioblastoma were obtained from IEU OpenGWAS project, which included 91 cases and 218,701 controls. We assessed the presence of heterogeneity and horizontal pleiotropy in the analyzed data. We primarily employed the inverse variance weighting method to investigate the causal relationship between gut microbiota and glioblastoma after excluding cases of horizontal pleiotropy. Four other analysis methods were employed as supplementary. Excluding abnormal results based on leave-one-out sensitivity analysis. Finally, reverse Mendelian randomization analysis was performed. RESULTS Four genus-level taxa and one family-level taxa exhibited causal associations with glioblastoma. And these results of reverse Mendelian randomization analysis shown glioblastoma exhibited causal associations with three genus-level taxa and one family-level taxa. However, the Prevotella7(Forward, P=0.006, OR=0.34, 95%CI:0.158-0.732; Reverse, P=0.004, OR=0.972, 95%CI:0.953-0.991) shown the causal associations with glioblastoma in the bidirectional Mendelian randomization. CONCLUSIONS In this bidirectional Mendelian randomization study, we identified five gut microbiota species with causal associations to glioblastoma. However, additional randomized controlled trials are required to clarify the impact of gut microbiota on glioblastoma and to reveal its precise mechanisms.
Collapse
Affiliation(s)
- Chuan Zeng
- The First Clinical Medical College of Gannan Medical University, Ganzhou City, 341000, Jiangxi Province, China
| | - Chaolong Zhang
- The First Clinical Medical College of Gannan Medical University, Ganzhou City, 341000, Jiangxi Province, China
| | - Chunming He
- Department of Neurosurgery, First Affiliated Hospital of Gannan Medical University, Qingnian Road, Ganzhou City, 341000, Jiangxi Province, China.
| | - Haimin Song
- Department of Neurosurgery, First Affiliated Hospital of Gannan Medical University, Qingnian Road, Ganzhou City, 341000, Jiangxi Province, China.
| |
Collapse
|
19
|
Eckerdt F, Platanias LC. Emerging Role of Glioma Stem Cells in Mechanisms of Therapy Resistance. Cancers (Basel) 2023; 15:3458. [PMID: 37444568 PMCID: PMC10340782 DOI: 10.3390/cancers15133458] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2023] [Revised: 06/14/2023] [Accepted: 06/29/2023] [Indexed: 07/15/2023] Open
Abstract
Since their discovery at the beginning of this millennium, glioma stem cells (GSCs) have sparked extensive research and an energetic scientific debate about their contribution to glioblastoma (GBM) initiation, progression, relapse, and resistance. Different molecular subtypes of GBM coexist within the same tumor, and they display differential sensitivity to chemotherapy. GSCs contribute to tumor heterogeneity and recapitulate pathway alterations described for the three GBM subtypes found in patients. GSCs show a high degree of plasticity, allowing for interconversion between different molecular GBM subtypes, with distinct proliferative potential, and different degrees of self-renewal and differentiation. This high degree of plasticity permits adaptation to the environmental changes introduced by chemo- and radiation therapy. Evidence from mouse models indicates that GSCs repopulate brain tumors after therapeutic intervention, and due to GSC plasticity, they reconstitute heterogeneity in recurrent tumors. GSCs are also inherently resilient to standard-of-care therapy, and mechanisms of resistance include enhanced DNA damage repair, MGMT promoter demethylation, autophagy, impaired induction of apoptosis, metabolic adaptation, chemoresistance, and immune evasion. The remarkable oncogenic properties of GSCs have inspired considerable interest in better understanding GSC biology and functions, as they might represent attractive targets to advance the currently limited therapeutic options for GBM patients. This has raised expectations for the development of novel targeted therapeutic approaches, including targeting GSC plasticity, chimeric antigen receptor T (CAR T) cells, and oncolytic viruses. In this review, we focus on the role of GSCs as drivers of GBM and therapy resistance, and we discuss how insights into GSC biology and plasticity might advance GSC-directed curative approaches.
Collapse
Affiliation(s)
- Frank Eckerdt
- Robert H. Lurie Comprehensive Cancer Center of Northwestern University, Chicago, IL 60611, USA
- Division of Hematology-Oncology, Department of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Leonidas C. Platanias
- Robert H. Lurie Comprehensive Cancer Center of Northwestern University, Chicago, IL 60611, USA
- Division of Hematology-Oncology, Department of Medicine, Northwestern University, Chicago, IL 60611, USA
- Medicine Service, Jesse Brown VA Medical Center, Chicago, IL 60612, USA
| |
Collapse
|
20
|
Lin B, Ye Z, Ye Z, Wang M, Cao Z, Gao R, Zhang Y. Gut microbiota in brain tumors: An emerging crucial player. CNS Neurosci Ther 2023; 29 Suppl 1:84-97. [PMID: 36627748 PMCID: PMC10314108 DOI: 10.1111/cns.14081] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2022] [Revised: 12/18/2022] [Accepted: 12/21/2022] [Indexed: 01/12/2023] Open
Abstract
In recent decades, various roles of the gut microbiota in physiological and pathological conditions have been uncovered. Among the many interacting pathways between the host and gut flora, the gut-brain axis has drawn increasing attention and is generally considered a promising way to understand and treat brain tumors, one of the most lethal neoplasms. In this narrative review, we aimed to unveil and dissect the sophisticated mechanisms by which the gut-brain axis exerts its influence on brain tumors. Furthermore, we summarized the latest research regarding the gastrointestinal microbial landscape and the effect of gut-brain axis malfunction on different brain tumors. Finally, we outlined the ongoing developing approaches of microbial manipulation and their corresponding research related to neuro-malignancies. Collectively, we recapitulated the advances in gut microbial alterations along with their potential interactive mechanisms in brain tumors and encouraged increased efforts in this area.
Collapse
Affiliation(s)
- Ben Lin
- Department of Neurosurgery, Huashan Hospital, Shanghai Medical CollegeFudan UniversityShanghaiChina
- National Center for Neurological DisordersShanghaiChina
- Shanghai Key Laboratory of Brain Function and Restoration and Neural RegenerationShanghaiChina
- Neurosurgical Institute of Fudan UniversityShanghaiChina
- Shanghai Clinical Medical Center of NeurosurgeryShanghaiChina
| | - Zhen Ye
- Department of Neurosurgery, Huashan Hospital, Shanghai Medical CollegeFudan UniversityShanghaiChina
- National Center for Neurological DisordersShanghaiChina
- Shanghai Key Laboratory of Brain Function and Restoration and Neural RegenerationShanghaiChina
- Neurosurgical Institute of Fudan UniversityShanghaiChina
- Shanghai Clinical Medical Center of NeurosurgeryShanghaiChina
| | - Zhao Ye
- Department of Neurosurgery, Huashan Hospital, Shanghai Medical CollegeFudan UniversityShanghaiChina
- National Center for Neurological DisordersShanghaiChina
- Shanghai Key Laboratory of Brain Function and Restoration and Neural RegenerationShanghaiChina
- Neurosurgical Institute of Fudan UniversityShanghaiChina
- Shanghai Clinical Medical Center of NeurosurgeryShanghaiChina
| | - Meng Wang
- Department of Endocrinology and Metabolism, Huashan Hospital, Shanghai Medical CollegeFudan UniversityShanghaiChina
| | - Zhan Cao
- Department of General Surgery, Shanghai Tenth People's Hospital, School of MedicineTongji UniversityShanghaiChina
| | - Renyuan Gao
- Department of General Surgery, Shanghai Tenth People's Hospital, School of MedicineTongji UniversityShanghaiChina
| | - Yichao Zhang
- Department of Neurosurgery, Huashan Hospital, Shanghai Medical CollegeFudan UniversityShanghaiChina
- National Center for Neurological DisordersShanghaiChina
- Shanghai Key Laboratory of Brain Function and Restoration and Neural RegenerationShanghaiChina
- Neurosurgical Institute of Fudan UniversityShanghaiChina
- Shanghai Clinical Medical Center of NeurosurgeryShanghaiChina
| |
Collapse
|
21
|
Li J, Wang K, Yang C, Zhu K, Jiang C, Wang M, Zhou Z, Tang N, Wang Q, Wang S, Shu P, Yuan H, Xiong Z, Li J, Liang T, Rao J, Wang X, Jiang X. Tumor-Associated Macrophage-Derived Exosomal LINC01232 Induces the Immune Escape in Glioma by Decreasing Surface MHC-I Expression. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2023; 10:e2207067. [PMID: 37097629 PMCID: PMC10265094 DOI: 10.1002/advs.202207067] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Revised: 03/12/2023] [Indexed: 06/15/2023]
Abstract
Tumor-associated macrophage (TAM) infiltration facilitates glioma malignancy, but the underlying mechanisms remain unclear. Herein, it is reported that TAMs secrete exosomal LINC01232 to induce tumor immune escape. Mechanistically, LINC01232 is found to directly bind E2F2 and promote E2F2 entry into the nucleus; the two synergistically promots the transcription of NBR1. The increase in binding between NBR1 binding and the ubiquitinating MHC-I protein through the ubiquitin domain causes an increase in the degradation of MHC-I in autophagolysosomes and a decrease in the expression of MHC-I on the surface of tumor cells, which in turn led to tumor cell escape from CD8+ CTL immune attack. Disruption of E2F2/NBR1/MHC-I signaling with shRNAs or blockade with the corresponding antibodies largely abolishes the tumor-supportive effects of LINC01232 and inhibits tumor growth driven by M2-type macrophages. Importantly, knockdown of LINC01232 enhances the expression of MHC-I on the surface of tumor cells and improves the response to reinfusion with CD8+ T cells. This study reveals the existence of critical molecular crosstalk between TAMs and glioma mediates through the LINC01232/E2F2/NBR1/MHC-I axis to support malignant tumor growth, indicating that targeting this axis may have therapeutic potential.
Collapse
Affiliation(s)
- Junjun Li
- Department of Neurosurgery, Union HospitalTongji Medical CollegeHuazhong University of Science and Technology1277 Jiefang AvenueWuhanHubei430022China
| | - Keshan Wang
- Department of UrologyUnion HospitalTongji Medical CollegeHuazhong University of Science and Technology1277 Jiefang AvenueWuhanHubei430022China
| | - Chao Yang
- Tianjin Institute of Industrial Biotechnology Chinese Academy of Sciences TianjinTianjin300308China
| | - Kai Zhu
- Department of Neurosurgery, Union HospitalTongji Medical CollegeHuazhong University of Science and Technology1277 Jiefang AvenueWuhanHubei430022China
| | - Cheng Jiang
- Department of Neurosurgery, Union HospitalTongji Medical CollegeHuazhong University of Science and Technology1277 Jiefang AvenueWuhanHubei430022China
| | - Minjie Wang
- Department of Neurosurgery, Union HospitalTongji Medical CollegeHuazhong University of Science and Technology1277 Jiefang AvenueWuhanHubei430022China
| | - Zijie Zhou
- Department of Neurosurgery, Union HospitalTongji Medical CollegeHuazhong University of Science and Technology1277 Jiefang AvenueWuhanHubei430022China
| | - Nan Tang
- Department of Neurosurgery, Union HospitalTongji Medical CollegeHuazhong University of Science and Technology1277 Jiefang AvenueWuhanHubei430022China
| | - Qiangping Wang
- Department of Neurosurgery, Union HospitalTongji Medical CollegeHuazhong University of Science and Technology1277 Jiefang AvenueWuhanHubei430022China
| | - Siqi Wang
- Department of RadiologyUnion HospitalTongji Medical CollegeHuazhong University of Science and Technology1277 Jiefang AvenueWuhanHubei430022China
| | - Pengwei Shu
- Department of RadiologyUnion HospitalTongji Medical CollegeHuazhong University of Science and Technology1277 Jiefang AvenueWuhanHubei430022China
| | - Hongliang Yuan
- Department of UltrasoundUnion HospitalTongji Medical CollegeHuazhong University of Science and Technology1277 Jiefang AvenueWuhanHubei430022China
| | - Zhiyong Xiong
- Department of Neurosurgery, Union HospitalTongji Medical CollegeHuazhong University of Science and Technology1277 Jiefang AvenueWuhanHubei430022China
| | - Jinsong Li
- Department of Thoracic SurgeryUnion HospitalTongji Medical CollegeHuazhong University of Science and Technology1277 Jiefang AvenueWuhanHubei430022China
| | - Tao Liang
- Department of Clinical LaboratoryUnion HospitalTongji Medical CollegeHuazhong University of Science and Technology1277 Jiefang AvenueWuhanHubei430022China
| | - Jin Rao
- Department of Neurosurgery, Union HospitalTongji Medical CollegeHuazhong University of Science and Technology1277 Jiefang AvenueWuhanHubei430022China
| | - Xuan Wang
- Department of Neurosurgery, Union HospitalTongji Medical CollegeHuazhong University of Science and Technology1277 Jiefang AvenueWuhanHubei430022China
| | - Xiaobing Jiang
- Department of Neurosurgery, Union HospitalTongji Medical CollegeHuazhong University of Science and Technology1277 Jiefang AvenueWuhanHubei430022China
| |
Collapse
|
22
|
Brisson L, Henrique Geraldo L, Bikfalvi A, Mathivet T. The strange Microenvironment of Glioblastoma. Rev Neurol (Paris) 2023; 179:490-501. [PMID: 36964121 PMCID: PMC11195635 DOI: 10.1016/j.neurol.2023.03.007] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2023] [Revised: 02/24/2023] [Accepted: 03/01/2023] [Indexed: 03/26/2023]
Abstract
Glioblastoma (GB) is the most common and aggressive primary brain tumor, with poor patient survival and lack of effective therapies. Late advances trying to decipher the composition of the GB tumor microenvironment (TME) emphasized its role in tumor progression and potentialized it as a therapeutic target. Many components participate critically to tumor development and expansion such as blood vessels, immune cells or components of the nervous system. Dysmorphic tumor vasculature brings challenges to optimal delivery of cytotoxic agents currently used in clinics. Also, massive infiltration of immunosuppressive myeloid cells and limited recruitment of T cells limits the success of conventional immunotherapies. Neuronal input seems also be required for tumor expansion. In this review, we provide a comprehensive report of vascular and immune component of the GB TME and their cross talk during GB progression.
Collapse
Affiliation(s)
- L Brisson
- BRIC Inserm U1312, Université de Bordeaux, 33615 Pessac, France
| | - L Henrique Geraldo
- Cardiovascular Research Center, Department of Internal Medicine, Yale University School of Medicine, New Haven, CT, USA
| | - A Bikfalvi
- BRIC Inserm U1312, Université de Bordeaux, 33615 Pessac, France.
| | - T Mathivet
- BRIC Inserm U1312, Université de Bordeaux, 33615 Pessac, France
| |
Collapse
|
23
|
Yan P, Wang J, Liu H, Liu X, Fu R, Feng J. M1 macrophage-derived exosomes containing miR-150 inhibit glioma progression by targeting MMP16. Cell Signal 2023:110731. [PMID: 37244635 DOI: 10.1016/j.cellsig.2023.110731] [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: 03/15/2023] [Revised: 05/08/2023] [Accepted: 05/22/2023] [Indexed: 05/29/2023]
Abstract
A large amount of clinical and experimental evidence indicates that M1 macrophages can inhibit tumor progression and expansion; however, the molecular mechanism by which macrophage-derived exosomes inhibit the proliferation of glioblastoma cells has not yet been elucidated. Here, we used M1 macrophage exosomes encapsulating microRNAs to inhibit the proliferation of glioma cells. Exosomes derived from M1 macrophages exhibited high expression levels of miR-150, and the inhibition of glioma cell proliferation mediated by exosomes derived from M1 macrophages was dependent on this microRNA. Mechanistically, miR-150 is transferred to glioblastoma cells through M1 macrophages and binds to MMP16, downregulating its expression and inhibiting glioma progression. Overall, these findings indicate that M1 macrophage-derived exosomes carrying miR-150 inhibit the proliferation of glioblastoma cells through targeted binding to MMP16. This dynamic mutual influence between glioblastoma cells and M1 macrophages provides new opportunities for the treatment of glioma.
Collapse
Affiliation(s)
- Pengfei Yan
- Department of Neurosurgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Avenue, Wuhan, Hubei 430022, China
| | - Jia Wang
- Department of Critical Care, People's Hospital of Dongxihu District, Wuhan, Hubei 430040, China
| | - Hongya Liu
- Wuhan Cell Learning Technology Co., Ltd., Optics Valley International Biomedical enterprise accelerator phase I project, No. 388, Gaoxin Second Road, Donghu New Technology Development Zone, Wuhan, Hubei 430000, China
| | - Xia Liu
- Wuhan Cell Learning Technology Co., Ltd., Optics Valley International Biomedical enterprise accelerator phase I project, No. 388, Gaoxin Second Road, Donghu New Technology Development Zone, Wuhan, Hubei 430000, China
| | - Rong Fu
- Department of Neurosurgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Avenue, Wuhan, Hubei 430022, China.
| | - Jun Feng
- Department of Neurosurgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Avenue, Wuhan, Hubei 430022, China.
| |
Collapse
|
24
|
Identification of Prognostic Aging-Related Genes Associated with Immune Cell Infiltration in Glioblastoma. JOURNAL OF ONCOLOGY 2023. [DOI: 10.1155/2023/9220547] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/23/2023]
Abstract
Background. Aging is recognized as a main tumor risk factor, and thus aging has become a field of interest in the tumor research field. Glioblastoma multiforme represents the most typical primary malignant intracranial tumor, particularly in the elderly. However, the association between aging-related genes (AGs) and GBM prognosis remains unknown. As a result, the primary goal of this study was to determine the association among AGs and the prognosis of GBM. Methods. A total of 307 human AGs were downloaded from the HAGR database, while the expression profiles of GSE4290 and GSE4412 were obtained from the GEO database. Furthermore, data on GBM expression profiles were obtained from the Chinese Glioma Genome Atlas (CGGA) database. The DEAGs that were differentially expressed among the AG and GBM gene expression profiles derived from GSE4290 were then identified, followed by functional analysis of the DEAGs. The survival-related AGs were then screened using univariate Cox regression analysis , which was used to build and validate a prognostic risk model. Furthermore, the ESTIMATE and CIBERSORT algorithms were utilized to explore the association between the survival-related AGs and the tumor immune microenvironment. Results. In entire, 29 DEAGs were identified in the GSE4290. This was monitored by the construction of the prognosis risk model using four DEAGs from the CGGA training set, including C1QA, CDK1, EFEMP1, and IGFBP2. Next, the risk model was confirmed in the CGGA experiment set and the GSE 4412 dataset. Results showed that C1QA, CDK1, EFEMP1, and IGFBP2 levels were remarkably higher in the high-risk score groups, and they had a good association with immune and stromal scores. Conclusion. A robust prognostic risk model was constructed and validated using four AGs, including C1QA, CDK1, EFEMP1, and IGFBP2, which had a close relationship with the immune microenvironment of GBM. This study offers a new reference to further explore the pathogenesis of GBM and recognize new and more effective GBM treatments.
Collapse
|
25
|
Zhao L, Xu DG, Hu YH. The Regulation of Microglial Cell Polarization in the Tumor Microenvironment: A New Potential Strategy for Auxiliary Treatment of Glioma-A Review. Cell Mol Neurobiol 2023; 43:193-204. [PMID: 35137327 PMCID: PMC11415204 DOI: 10.1007/s10571-022-01195-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2021] [Accepted: 01/09/2022] [Indexed: 01/07/2023]
Abstract
Glioma is the most common primary tumor of the central nervous system and normally should be treated by synthetic therapy, mainly with surgical operation assisted by radiotherapy and chemotherapy; however, the therapeutic effect has not been satisfactory, and the 5-year survival rates of anaplastic glioma and glioblastoma are 29.7% and 5.5%, respectively. To identify a more efficient strategy to treat glioma, in recent years, the influence of the inflammatory microenvironment on the progression of glioma has been studied. Various immunophenotypes exist in microglial cells, each of which has a different functional property. In this review, references about the phenotypic conversion of microglial cell polarity in the microenvironment were briefly summarized, and the differences in polarized state and function, their influences on glioma progression under different physiological and pathological conditions, and the interactive effects between the two were mainly discussed. Certain signaling molecules and regulatory pathways involved in the microglial cell polarization process were investigated, and the feasibility of targeted regulation of microglial cell conversion to an antitumor phenotype was analyzed to provide new clues for the efficient auxiliary treatment of neural glioma.
Collapse
Affiliation(s)
- Lei Zhao
- Department of Neurosurgery, The Second Hospital of Hebei Medical University, 215 Heping West Road, Shijiazhuang, 050000, Hebei, People's Republic of China
| | - Dong-Gang Xu
- Institute of Military Cognition and Brain Science, Research Academy of Military Medical Sciences, Beijing, 100850, People's Republic of China
| | - Yu-Hua Hu
- Department of Neurosurgery, The Second Hospital of Hebei Medical University, 215 Heping West Road, Shijiazhuang, 050000, Hebei, People's Republic of China.
| |
Collapse
|
26
|
Single-cell spatial immune landscapes of primary and metastatic brain tumours. Nature 2023; 614:555-563. [PMID: 36725935 PMCID: PMC9931580 DOI: 10.1038/s41586-022-05680-3] [Citation(s) in RCA: 151] [Impact Index Per Article: 75.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2022] [Accepted: 12/22/2022] [Indexed: 02/03/2023]
Abstract
Single-cell technologies have enabled the characterization of the tumour microenvironment at unprecedented depth and have revealed vast cellular diversity among tumour cells and their niche. Anti-tumour immunity relies on cell-cell relationships within the tumour microenvironment1,2, yet many single-cell studies lack spatial context and rely on dissociated tissues3. Here we applied imaging mass cytometry to characterize the immunological landscape of 139 high-grade glioma and 46 brain metastasis tumours from patients. Single-cell analysis of more than 1.1 million cells across 389 high-dimensional histopathology images enabled the spatial resolution of immune lineages and activation states, revealing differences in immune landscapes between primary tumours and brain metastases from diverse solid cancers. These analyses revealed cellular neighbourhoods associated with survival in patients with glioblastoma, which we leveraged to identify a unique population of myeloperoxidase (MPO)-positive macrophages associated with long-term survival. Our findings provide insight into the biology of primary and metastatic brain tumours, reinforcing the value of integrating spatial resolution to single-cell datasets to dissect the microenvironmental contexture of cancer.
Collapse
|
27
|
Liu YY, Yao RQ, Long LY, Liu YX, Tao BY, Liu HY, Liu JL, Li Z, Chen L, Yao YM. Worldwide productivity and research trend of publications concerning glioma-associated macrophage/microglia: A bibliometric study. Front Neurol 2022; 13:1047162. [PMID: 36570441 PMCID: PMC9772275 DOI: 10.3389/fneur.2022.1047162] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2022] [Accepted: 11/14/2022] [Indexed: 12/12/2022] Open
Abstract
Glioma-associated macrophage/microglia (GAM) represents a key player in shaping a unique glioma ecosystem to facilitate tumor progression and therapeutic resistance. Numerous studies have been published concerning GAM, but no relevant bibliometric study has been performed yet. Our bibliometric study aimed to comprehensively summarize and analyze the global scientific output, research hotspots, and trendy topics of publications on GAM over time. Data on publications on GAM were collected using the Web of Science (WoS). The search date was 16 January 2022, and the publications were collected from 2002 to 2021. Totally, 1,224 articles and reviews were incorporated and analyzed in the current study. It showed that the annual publications concerning GAM kept increasing over the past 20 years. The United States had the largest number of publications and total citations. Holland, Kettenmann, and Gutmann were the top three authors in terms of citation frequency. Neuro-oncology represented the most influential journal in GAM studies, with the highest H-index, total citations, and publication numbers. The paper published by Hambardzumyan in 2016 had the highest local citations. Additionally, the analysis of keywords implied that "prognosis," "tumor microenvironment," and "immunotherapy" might become research hotspots. Furthermore, trendy topics in GAM studies suggested that "immune infiltration," "immune microenvironment," "bioinformatics," "prognosis," and "immunotherapy" deserved additional attention. In conclusion, this bibliometric study comprehensively analyzed the publication trend of GAM studies for the past 20 years, in which the research hotspots and trendy topics were also uncovered. This information offered scholars critical references for conducting in-depth studies on GAM in the future.
Collapse
Affiliation(s)
- Yu-yang Liu
- Translational Medicine Research Center, Medical Innovation Research Division and Fourth Medical Center of the Chinese PLA General Hospital, Beijing, China,Medical School of Chinese PLA, Beijing, China,Department of Neurosurgery, Chinese PLA General Hospital, Beijing, China
| | - Ren-qi Yao
- Translational Medicine Research Center, Medical Innovation Research Division and Fourth Medical Center of the Chinese PLA General Hospital, Beijing, China,Department of Burn Surgery, The First Affiliated Hospital of Naval Medical University, Shanghai, China
| | - Li-yan Long
- Library, Medical School of Chinese PLA, Beijing, China
| | - Yu-xiao Liu
- Department of Neurosurgery, Chinese PLA General Hospital, Beijing, China
| | - Bing-Yan Tao
- Medical School of Chinese PLA, Beijing, China,Department of Neurosurgery, Chinese PLA General Hospital, Beijing, China
| | - Hong-yu Liu
- Medical School of Chinese PLA, Beijing, China,Department of Neurosurgery, Chinese PLA General Hospital, Beijing, China
| | - Jia-lin Liu
- Department of Neurosurgery, Chinese PLA General Hospital, Beijing, China
| | - Ze Li
- Department of Neurosurgery, Chinese PLA General Hospital, Beijing, China
| | - Ling Chen
- Department of Neurosurgery, Chinese PLA General Hospital, Beijing, China,Ling Chen
| | - Yong-ming Yao
- Translational Medicine Research Center, Medical Innovation Research Division and Fourth Medical Center of the Chinese PLA General Hospital, Beijing, China,*Correspondence: Yong-ming Yao
| |
Collapse
|
28
|
Tabu K, Taga T. Cancer ego-system in glioma: an iron-replenishing niche network systemically self-organized by cancer stem cells. Inflamm Regen 2022; 42:54. [PMID: 36451253 PMCID: PMC9710158 DOI: 10.1186/s41232-022-00240-8] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2022] [Accepted: 11/16/2022] [Indexed: 12/03/2022] Open
Abstract
For all living organisms, the adaptation to outside environments is an essential determinant to survive natural and artificial selections and to sustain the whole ecosystem intact with functional biodiversity. Likewise, cancer cells have similar characteristics that evade not only stresses from the host-internal innate and adaptive immune systems but also those from host-externally administered therapeutic interventions. Such selfish characteristics of cancer cells lead to the formation of cancerous ecosystem with a wide variety of phenotypic heterogeneity, which should be called cancer "egosystem" from the host point of view. Recently increasing evidence demonstrates that cancer stem cells (CSCs) are responsible for this cancer egosystem by effectively exploiting host inflammatory and hematopoietic cells and thereby reconstructing their own advantageous niches, which may well be a driving force in cancer recurrence. CSCs are further likely to render multiple niches mutually interconnected and cooperating as a network to support back CSCs themselves. Here, we summarize a recently identified iron-replenishing niche network self-organized by glioma CSCs (GSCs) through remote regulation of host myeloid and erythroid lineage cells. GSCs recruit bone marrow (BM)-derived inflammatory monocytes into tumor parenchyma, facilitate their differentiation into macrophages (Mφs) and skew their polarization into pro-tumoral phenotype, i.e., tumor-associated Mφs (TAMs). Meanwhile, GSCs distantly enhance erythropoiesis in host hematopoietic organs like BM and spleen potentially by secreting some soluble mediators that maintain continuous supply of erythrocytes within tumors. In addition, as normal red pulp Mφs (RPMs) under steady state conditions in spleen recycle iron by phagocytosing the aged or damaged erythrocytes (a/dECs) and release it in time of need, TAMs at least in gliomas phagocytose the hemorrhaged erythrocytes within tumors and potentially serve as a source of iron, an important nutrient indispensable to GSC survival and glioma progression. Taken together, these studies provide the substantial evidence that CSCs have a unique strategy to orchestrate multiple niches as an ecosystem that threatens the host living, which in this sense must be an egosystem. Targeting such an adaptive subpopulation of CSCs could achieve drastic disturbance of the CSC niches and subsequent extinction of malignant neoplasms.
Collapse
Affiliation(s)
- Kouichi Tabu
- grid.265073.50000 0001 1014 9130Department of Stem Cell Regulation, Medical Research Institute, Tokyo Medical and Dental University (TMDU), Tokyo, 113-8510 Japan
| | - Tetsuya Taga
- grid.265073.50000 0001 1014 9130Department of Stem Cell Regulation, Medical Research Institute, Tokyo Medical and Dental University (TMDU), Tokyo, 113-8510 Japan
| |
Collapse
|
29
|
Li D, Zhang Q, Li L, Chen K, Yang J, Dixit D, Gimple RC, Ci S, Lu C, Hu L, Gao J, Shan D, Li Y, Zhang J, Shi Z, Gu D, Yuan W, Wu Q, Yang K, Zhao L, Qiu Z, Lv D, Gao W, Yang H, Lin F, Wang Q, Man J, Li C, Tao W, Agnihotri S, Qian X, Shi Y, You Y, Zhang N, Rich JN, Wang X. β2-Microglobulin Maintains Glioblastoma Stem Cells and Induces M2-like Polarization of Tumor-Associated Macrophages. Cancer Res 2022; 82:3321-3334. [PMID: 35841593 DOI: 10.1158/0008-5472.can-22-0507] [Citation(s) in RCA: 52] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2022] [Revised: 06/08/2022] [Accepted: 07/13/2022] [Indexed: 11/16/2022]
Abstract
Glioblastoma (GBM) is a complex ecosystem that includes a heterogeneous tumor population and the tumor-immune microenvironment (TIME), prominently containing tumor-associated macrophages (TAM) and microglia. Here, we demonstrated that β2-microglobulin (B2M), a subunit of the class I major histocompatibility complex (MHC-I), promotes the maintenance of stem-like neoplastic populations and reprograms the TIME to an anti-inflammatory, tumor-promoting state. B2M activated PI3K/AKT/mTOR signaling by interacting with PIP5K1A in GBM stem cells (GSC) and promoting MYC-induced secretion of transforming growth factor-β1 (TGFβ1). Inhibition of B2M attenuated GSC survival, self-renewal, and tumor growth. B2M-induced TGFβ1 secretion activated paracrine SMAD and PI3K/AKT signaling in TAMs and promoted an M2-like macrophage phenotype. These findings reveal tumor-promoting functions of B2M and suggest that targeting B2M or its downstream axis may provide an effective approach for treating GBM. SIGNIFICANCE β2-microglobulin signaling in glioblastoma cells activates a PI3K/AKT/MYC/TGFβ1 axis that maintains stem cells and induces M2-like macrophage polarization, highlighting potential therapeutic strategies for targeting tumor cells and the immunosuppressive microenvironment in glioblastoma.
Collapse
Affiliation(s)
- Daqi Li
- National Health Commission Key Laboratory of Antibody Techniques, Department of Cell Biology, Jiangsu Provincial Key Laboratory of Human Functional Genomics, School of Basic Medical Sciences, Nanjing Medical University, Nanjing, Jiangsu, China
- Institute for Brain Tumors, Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Personalized Cancer Medicine, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Qian Zhang
- National Health Commission Key Laboratory of Antibody Techniques, Department of Cell Biology, Jiangsu Provincial Key Laboratory of Human Functional Genomics, School of Basic Medical Sciences, Nanjing Medical University, Nanjing, Jiangsu, China
- Institute for Brain Tumors, Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Personalized Cancer Medicine, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Lu Li
- National Health Commission Key Laboratory of Antibody Techniques, Department of Cell Biology, Jiangsu Provincial Key Laboratory of Human Functional Genomics, School of Basic Medical Sciences, Nanjing Medical University, Nanjing, Jiangsu, China
- Institute for Brain Tumors, Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Personalized Cancer Medicine, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Kexin Chen
- National Health Commission Key Laboratory of Antibody Techniques, Department of Cell Biology, Jiangsu Provincial Key Laboratory of Human Functional Genomics, School of Basic Medical Sciences, Nanjing Medical University, Nanjing, Jiangsu, China
- Institute for Brain Tumors, Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Personalized Cancer Medicine, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Junlei Yang
- National Health Commission Key Laboratory of Antibody Techniques, Department of Cell Biology, Jiangsu Provincial Key Laboratory of Human Functional Genomics, School of Basic Medical Sciences, Nanjing Medical University, Nanjing, Jiangsu, China
- Institute for Brain Tumors, Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Personalized Cancer Medicine, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Deobrat Dixit
- Department of Medicine, Division of Regenerative Medicine, University of California, San Diego, La Jolla, California
| | - Ryan C Gimple
- Department of Pathology, Case Western Reserve University, Cleveland, Ohio
| | - Shusheng Ci
- National Health Commission Key Laboratory of Antibody Techniques, Department of Cell Biology, Jiangsu Provincial Key Laboratory of Human Functional Genomics, School of Basic Medical Sciences, Nanjing Medical University, Nanjing, Jiangsu, China
- Institute for Brain Tumors, Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Personalized Cancer Medicine, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Chenfei Lu
- National Health Commission Key Laboratory of Antibody Techniques, Department of Cell Biology, Jiangsu Provincial Key Laboratory of Human Functional Genomics, School of Basic Medical Sciences, Nanjing Medical University, Nanjing, Jiangsu, China
- Institute for Brain Tumors, Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Personalized Cancer Medicine, Nanjing Medical University, Nanjing, Jiangsu, China
- Department of Neurosurgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Lang Hu
- National Health Commission Key Laboratory of Antibody Techniques, Department of Cell Biology, Jiangsu Provincial Key Laboratory of Human Functional Genomics, School of Basic Medical Sciences, Nanjing Medical University, Nanjing, Jiangsu, China
- Institute for Brain Tumors, Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Personalized Cancer Medicine, Nanjing Medical University, Nanjing, Jiangsu, China
- Department of Neurosurgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Jiancheng Gao
- National Health Commission Key Laboratory of Antibody Techniques, Department of Cell Biology, Jiangsu Provincial Key Laboratory of Human Functional Genomics, School of Basic Medical Sciences, Nanjing Medical University, Nanjing, Jiangsu, China
- Institute for Brain Tumors, Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Personalized Cancer Medicine, Nanjing Medical University, Nanjing, Jiangsu, China
- Department of Neurosurgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Danyang Shan
- National Health Commission Key Laboratory of Antibody Techniques, Department of Cell Biology, Jiangsu Provincial Key Laboratory of Human Functional Genomics, School of Basic Medical Sciences, Nanjing Medical University, Nanjing, Jiangsu, China
- Institute for Brain Tumors, Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Personalized Cancer Medicine, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Yangqing Li
- Ministry of Education Key Laboratory of Model Animals for Disease Study, Model Animal Research Center and School of Medicine, Nanjing University, National Resource Center for Mutant Mice, Nanjing, China
| | - Junxia Zhang
- Department of Neurosurgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Zhumei Shi
- Department of Neurosurgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Danling Gu
- Department of Hematology, The First Affiliated Hospital of Nanjing Medical University, Jiangsu Province Hospital, Nanjing, China
| | - Wei Yuan
- Department of Pathology, The Fourth Affiliated Hospital of Nantong University, The First people's Hospital of Yancheng, Yancheng, China
| | - Qiulian Wu
- Department of Medicine, Division of Regenerative Medicine, University of California, San Diego, La Jolla, California
- UPMC Hillman Cancer Center, Pittsburgh, Pennsylvania
| | - Kailin Yang
- Department of Radiation Oncology, Taussig Cancer Center, Cleveland Clinic, Cleveland, Ohio
| | - Linjie Zhao
- Department of Medicine, Division of Regenerative Medicine, University of California, San Diego, La Jolla, California
- UPMC Hillman Cancer Center, Pittsburgh, Pennsylvania
| | - Zhixin Qiu
- Department of Medicine, Division of Regenerative Medicine, University of California, San Diego, La Jolla, California
- UPMC Hillman Cancer Center, Pittsburgh, Pennsylvania
- Institute for Translational Brain Research, Fudan University, Shanghai, China
| | - Deguan Lv
- Department of Medicine, Division of Regenerative Medicine, University of California, San Diego, La Jolla, California
| | - Wei Gao
- National Health Commission Key Laboratory of Antibody Techniques, Department of Cell Biology, Jiangsu Provincial Key Laboratory of Human Functional Genomics, School of Basic Medical Sciences, Nanjing Medical University, Nanjing, Jiangsu, China
- Institute for Brain Tumors, Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Personalized Cancer Medicine, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Hui Yang
- Department of Neurosurgery, Huashan Hospital, Shanghai Key Laboratory of Brain Function Restoration and Neural Regeneration, Shanghai Clinical Medical Center of Neurosurgery, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institute for Translational Brain Research, Shanghai Medical College, Fudan University, Shanghai, China
| | - Fan Lin
- National Health Commission Key Laboratory of Antibody Techniques, Department of Cell Biology, Jiangsu Provincial Key Laboratory of Human Functional Genomics, School of Basic Medical Sciences, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Qianghu Wang
- Institute for Brain Tumors, Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Personalized Cancer Medicine, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Jianghong Man
- State Key Laboratory of Proteomics, National Center of Biomedical Analysis, Beijing, China
| | - Chaojun Li
- Ministry of Education Key Laboratory of Model Animals for Disease Study, Model Animal Research Center and School of Medicine, Nanjing University, National Resource Center for Mutant Mice, Nanjing, China
| | - Weiwei Tao
- College of Biomedicine and Health and College of Life Science and Technology, Huazhong Agricultural University, Wuhan, Hubei, China
| | - Sameer Agnihotri
- Department of Neurological Surgery, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Xu Qian
- Institute for Brain Tumors, Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Personalized Cancer Medicine, Nanjing Medical University, Nanjing, Jiangsu, China
- Department of Nutrition and Food Hygiene, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Yu Shi
- Institute of Pathology, Ministry of Education Key Laboratory of Tumor Immunopathology, Southwest Hospital, Chongqing, China
| | - Yongping You
- Department of Neurosurgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Nu Zhang
- Department of Neurosurgery, The First Affiliated Hospital of Sun Yat-sen University, Guangdong Provincial Key Laboratory of Brain Function and Disease, Guangdong Translational Medicine Innovation Platform, Guangzhou, Guangdong, China
| | - Jeremy N Rich
- Department of Medicine, Division of Regenerative Medicine, University of California, San Diego, La Jolla, California
- UPMC Hillman Cancer Center, Pittsburgh, Pennsylvania
- Department of Neurology, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Xiuxing Wang
- National Health Commission Key Laboratory of Antibody Techniques, Department of Cell Biology, Jiangsu Provincial Key Laboratory of Human Functional Genomics, School of Basic Medical Sciences, Nanjing Medical University, Nanjing, Jiangsu, China
- Institute for Brain Tumors, Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Personalized Cancer Medicine, Nanjing Medical University, Nanjing, Jiangsu, China
- Department of Medicine, Division of Regenerative Medicine, University of California, San Diego, La Jolla, California
- Jiangsu Cancer Hospital, Affiliated Cancer Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| |
Collapse
|
30
|
Kishk A, Pacheco MP, Heurtaux T, Sinkkonen L, Pang J, Fritah S, Niclou SP, Sauter T. Review of Current Human Genome-Scale Metabolic Models for Brain Cancer and Neurodegenerative Diseases. Cells 2022; 11:2486. [PMID: 36010563 PMCID: PMC9406599 DOI: 10.3390/cells11162486] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2022] [Revised: 07/28/2022] [Accepted: 08/08/2022] [Indexed: 11/16/2022] Open
Abstract
Brain disorders represent 32% of the global disease burden, with 169 million Europeans affected. Constraint-based metabolic modelling and other approaches have been applied to predict new treatments for these and other diseases. Many recent studies focused on enhancing, among others, drug predictions by generating generic metabolic models of brain cells and on the contextualisation of the genome-scale metabolic models with expression data. Experimental flux rates were primarily used to constrain or validate the model inputs. Bi-cellular models were reconstructed to study the interaction between different cell types. This review highlights the evolution of genome-scale models for neurodegenerative diseases and glioma. We discuss the advantages and drawbacks of each approach and propose improvements, such as building bi-cellular models, tailoring the biomass formulations for glioma and refinement of the cerebrospinal fluid composition.
Collapse
Affiliation(s)
- Ali Kishk
- Department of Life Sciences and Medicine, University of Luxembourg, L-4367 Belvaux, Luxembourg
| | - Maria Pires Pacheco
- Department of Life Sciences and Medicine, University of Luxembourg, L-4367 Belvaux, Luxembourg
| | - Tony Heurtaux
- Department of Life Sciences and Medicine, University of Luxembourg, L-4367 Belvaux, Luxembourg
- Luxembourg Center of Neuropathology, L-3555 Dudelange, Luxembourg
| | - Lasse Sinkkonen
- Department of Life Sciences and Medicine, University of Luxembourg, L-4367 Belvaux, Luxembourg
| | - Jun Pang
- Department of Computer Science, University of Luxembourg, L-4364 Esch-sur-Alzette, Luxembourg
| | - Sabrina Fritah
- NORLUX Neuro-Oncology Laboratory, Luxembourg Institute of Health, Department of Cancer Research, L-1526 Luxembourg, Luxembourg
| | - Simone P. Niclou
- NORLUX Neuro-Oncology Laboratory, Luxembourg Institute of Health, Department of Cancer Research, L-1526 Luxembourg, Luxembourg
| | - Thomas Sauter
- Department of Life Sciences and Medicine, University of Luxembourg, L-4367 Belvaux, Luxembourg
| |
Collapse
|
31
|
Shibahara I, Shibahara Y, Hagiwara H, Watanabe T, Orihashi Y, Handa H, Inukai M, Hide T, Yasui Y, Kumabe T. Ventricular opening and cerebrospinal fluid circulation accelerate the biodegradation process of carmustine wafers suggesting their immunomodulation potential in the human brain. J Neurooncol 2022; 159:425-435. [PMID: 35802230 DOI: 10.1007/s11060-022-04078-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2022] [Accepted: 06/25/2022] [Indexed: 11/28/2022]
Abstract
PURPOSE Opening the ventricular system during glioblastoma surgery is often necessary, but the consequent effect on the tumor microenvironment of glioblastoma remains unknown. Implantation of carmustine wafer enables direct drug delivery to the tumor site; however, the exact mechanism of the wafer's biodegradation process is unclear, and the available data is limited to in vivo non-human mammalian studies. We hypothesized that the ventricular opening affects the degradation process of the wafer and the glioblastoma tumor microenvironment. METHODS This study included 30 glioblastoma patients. 21 patients underwent carmustine wafer implantation during initial surgery. All patients underwent repeated surgical resection upon recurrence, allowing for pathological comparison of changes associated with wafer implantation. Immunohistochemical analyses were performed using CD68, TMEM119, CD163, IBA1, BIN1, and CD31 antibodies to highlight microglia, macrophages, and tumor vascularity, and the quantitative scoring results were correlated with clinical, molecular, and surgical variables, including the effect of the ventricular opening. RESULTS The carmustine wafer implanted group presented significantly less TMEM119-positive microglia within the tumor (P = 0.0002). Simple and multiple regression analyses revealed that the decrease in TMEM119-positive microglia was correlated with longer intervals between surgeries and opened ventricular systems. No correlation was observed between age, methylated O6-methylguanine DNA methyltransferase promoter expression, and the extent of surgical resection. CONCLUSIONS Our study findings strongly suggest that biomaterials may possess immunomodulation capacity, which is significantly impacted by the ventricular opening procedure. Furthermore, our data highlights the pathophysiological effects of the ventricular opening within the surrounding human brain, especially after the wafer implantation.
Collapse
Affiliation(s)
- Ichiyo Shibahara
- Department of Neurosurgery, Kitasato University School of Medicine, 1-15-1 Kitasato Minami-ku, Sagamihara, Kanagawa, 252-0374, Japan.
| | - Yukiko Shibahara
- Department of Pathology, Kitasato University School of Medicine, Sagamihara, Kanagawa, Japan
| | - Hiroyuki Hagiwara
- Department of Neurosurgery, Yamato Municipal Hospital, Yamato, Kanagawa, Japan
| | - Takashi Watanabe
- Department of General Internal Medicine, JCHO Sendai Hospital, Sendai, Miyagi, Japan
| | - Yasushi Orihashi
- Division of Clinical Research, Kitasato University Hospital, Sagamihara, Kanagawa, Japan
| | - Hajime Handa
- Department of Neurosurgery, Kitasato University School of Medicine, 1-15-1 Kitasato Minami-ku, Sagamihara, Kanagawa, 252-0374, Japan
| | - Madoka Inukai
- Department of Neurosurgery, Kitasato University School of Medicine, 1-15-1 Kitasato Minami-ku, Sagamihara, Kanagawa, 252-0374, Japan.,Department of Pathology, Kitasato University School of Medicine, Sagamihara, Kanagawa, Japan
| | - Takuichiro Hide
- Department of Neurosurgery, Kitasato University School of Medicine, 1-15-1 Kitasato Minami-ku, Sagamihara, Kanagawa, 252-0374, Japan
| | - Yoshie Yasui
- Department of Neurosurgery, Kitasato University School of Medicine, 1-15-1 Kitasato Minami-ku, Sagamihara, Kanagawa, 252-0374, Japan
| | - Toshihiro Kumabe
- Department of Neurosurgery, Kitasato University School of Medicine, 1-15-1 Kitasato Minami-ku, Sagamihara, Kanagawa, 252-0374, Japan
| |
Collapse
|
32
|
Liu YQ, Luo M, Shi Y, Guo Y, Zhang H, Yang KD, Li TR, Yang LQ, Liu TT, Huang B, Liu Q, He ZC, Zhang XN, Wang WY, Wang S, Zeng H, Niu Q, Zhang X, Cui YH, Zhang ZR, Bian XW, Ping YF. Dicer deficiency impairs proliferation but potentiates anti-tumoral effect of macrophages in glioblastoma. Oncogene 2022; 41:3791-3803. [PMID: 35764885 DOI: 10.1038/s41388-022-02393-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2021] [Revised: 06/05/2022] [Accepted: 06/16/2022] [Indexed: 11/09/2022]
Abstract
Glioblastoma is a lethal primary brain tumor with abundant immune-suppressive glioblastoma-associated macrophage (GAM) infiltration. Skewing immune suppressive GAMs towards an immune-activating phenotype represents a promising immunotherapeutic strategy against glioblastoma. Herein, we reported that genetic deletion of miRNA-processing enzyme Dicer in macrophages inhibited the growth of GL261 murine glioblastoma xenografts and prolonged survival of tumor-bearing mice. Single cell RNA sequencing (scRNA-seq) of the tumor-infiltrating immune cells revealed that Dicer deletion in macrophages reduced the proportion of cell-cycling GAM cluster and reprogramed the remaining GAMs towards a proinflammatory activation state (enhanced phagocytotic and IFN-producing signature). Dicer-deficient GAMs showed reduced level of cyclin-dependent kinases (CDK1 and CDK2) and increased expression of CDK inhibitor p27 Kip1, thus manifesting impaired proliferation. Dicer knockout enhanced phagocytotic activity of GAMs to eliminate GL261 tumor cells. Increased proinflammatory GAM clusters in macrophage Dicer-deficient mice actively interacted with tumor-infiltrating T cells and NK cells through TNF paracrine signaling to create a pro-inflammatory immune microenvironment for tumor cell elimination. Our work identifies the role of Dicer deletion in macrophages in generating an immune-activating microenvironment, which could be further developed as a potential immunotherapeutic strategy against glioblastoma.
Collapse
Affiliation(s)
- Yu-Qi Liu
- Institute of Pathology and Southwest Cancer Center, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, China.,Key Laboratory of Tumor Immunopathology, Ministry of Education of China, Chongqing, 400038, China.,Army 953 Hospital, Shigatse Branch of Xinqiao Hospital, Third Military Medical University (Army Medical University), Shigatse, 857000, China
| | - Min Luo
- Institute of Pathology and Southwest Cancer Center, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, China.,Key Laboratory of Tumor Immunopathology, Ministry of Education of China, Chongqing, 400038, China
| | - Yu Shi
- Institute of Pathology and Southwest Cancer Center, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, China.,Key Laboratory of Tumor Immunopathology, Ministry of Education of China, Chongqing, 400038, China
| | - Ying Guo
- Institute of Pathology and Southwest Cancer Center, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, China.,Key Laboratory of Tumor Immunopathology, Ministry of Education of China, Chongqing, 400038, China
| | - Hua Zhang
- Institute of Pathology and Southwest Cancer Center, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, China.,Key Laboratory of Tumor Immunopathology, Ministry of Education of China, Chongqing, 400038, China
| | - Kai-Di Yang
- Institute of Pathology and Southwest Cancer Center, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, China.,Key Laboratory of Tumor Immunopathology, Ministry of Education of China, Chongqing, 400038, China
| | - Tian-Ran Li
- Institute of Pathology and Southwest Cancer Center, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, China.,Key Laboratory of Tumor Immunopathology, Ministry of Education of China, Chongqing, 400038, China
| | - Liu-Qing Yang
- Institute of Pathology and Southwest Cancer Center, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, China.,Key Laboratory of Tumor Immunopathology, Ministry of Education of China, Chongqing, 400038, China
| | - Ting-Ting Liu
- Institute of Immunology, Third Military Medical University (Army Medical University), Chongqing, 400038, China
| | - Bo Huang
- Institute of Immunology, Third Military Medical University (Army Medical University), Chongqing, 400038, China
| | - Qing Liu
- Institute of Pathology and Southwest Cancer Center, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, China.,Key Laboratory of Tumor Immunopathology, Ministry of Education of China, Chongqing, 400038, China
| | - Zhi-Cheng He
- Institute of Pathology and Southwest Cancer Center, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, China.,Key Laboratory of Tumor Immunopathology, Ministry of Education of China, Chongqing, 400038, China
| | - Xiao-Ning Zhang
- Institute of Pathology and Southwest Cancer Center, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, China.,Key Laboratory of Tumor Immunopathology, Ministry of Education of China, Chongqing, 400038, China
| | - Wen-Ying Wang
- Institute of Pathology and Southwest Cancer Center, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, China.,Key Laboratory of Tumor Immunopathology, Ministry of Education of China, Chongqing, 400038, China
| | - Shuai Wang
- Institute of Pathology and Southwest Cancer Center, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, China.,Key Laboratory of Tumor Immunopathology, Ministry of Education of China, Chongqing, 400038, China
| | - Hui Zeng
- Institute of Pathology and Southwest Cancer Center, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, China.,Key Laboratory of Tumor Immunopathology, Ministry of Education of China, Chongqing, 400038, China
| | - Qin Niu
- Institute of Pathology and Southwest Cancer Center, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, China.,Key Laboratory of Tumor Immunopathology, Ministry of Education of China, Chongqing, 400038, China
| | - Xia Zhang
- Institute of Pathology and Southwest Cancer Center, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, China.,Key Laboratory of Tumor Immunopathology, Ministry of Education of China, Chongqing, 400038, China
| | - You-Hong Cui
- Institute of Pathology and Southwest Cancer Center, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, China.,Key Laboratory of Tumor Immunopathology, Ministry of Education of China, Chongqing, 400038, China
| | - Zhi-Ren Zhang
- Institute of Immunology, Third Military Medical University (Army Medical University), Chongqing, 400038, China.
| | - Xiu-Wu Bian
- Institute of Pathology and Southwest Cancer Center, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, China. .,Key Laboratory of Tumor Immunopathology, Ministry of Education of China, Chongqing, 400038, China.
| | - Yi-Fang Ping
- Institute of Pathology and Southwest Cancer Center, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, China. .,Key Laboratory of Tumor Immunopathology, Ministry of Education of China, Chongqing, 400038, China.
| |
Collapse
|
33
|
Menna G, Mattogno PP, Donzelli CM, Lisi L, Olivi A, Della Pepa GM. Glioma-Associated Microglia Characterization in the Glioblastoma Microenvironment through a 'Seed-and Soil' Approach: A Systematic Review. Brain Sci 2022; 12:718. [PMID: 35741603 PMCID: PMC9220868 DOI: 10.3390/brainsci12060718] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2022] [Revised: 05/25/2022] [Accepted: 05/28/2022] [Indexed: 12/04/2022] Open
Abstract
Background and aim: Ever since the discovery of tumor-associated immune cells, there has been growing interest in the understanding of the mechanisms underlying the crosstalk between these cells and tumor cells. A "seed and soil" approach has been recently introduced to describe the glioblastoma (GBM) landscape: tumor microenvironments act as fertile "soil" and interact with the "seed" (glial and stem cells compartment). In the following article, we provide a systematic review of the current evidence pertaining to the characterization of glioma-associated macrophages and microglia (GAMs) and microglia and macrophage cells in the glioma tumor microenvironment (TME). Methods: An online literature search was launched on PubMed Medline and Scopus using the following research string: "((Glioma associated macrophages OR GAM OR Microglia) AND (glioblastoma tumor microenvironment OR TME))". The last search for articles pertinent to the topic was conducted in February 2022. Results: The search of the literature yielded a total of 349 results. A total of 235 studies were found to be relevant to our research question and were assessed for eligibility. Upon a full-text review, 58 articles were included in the review. The reviewed papers were further divided into three categories based on their focus: (1) Microglia maintenance of immunological homeostasis and protection against autoimmunity; (2) Microglia crosstalk with dedifferentiated and stem-like glioblastoma cells; (3) Microglia migratory behavior and its activation pattern. Conclusions: Aggressive growth, inevitable recurrence, and scarce response to immunotherapies are driving the necessity to focus on the GBM TME from a different perspective to possibly disentangle its role as a fertile 'soil' for tumor progression and identify within it feasible therapeutic targets. Against this background, our systematic review confirmed microglia to play a paramount role in promoting GBM progression and relapse after treatments. The correct and extensive understanding of microglia-glioma crosstalk could help in understanding the physiopathology of this complex disease, possibly opening scenarios for improvement of treatments.
Collapse
Affiliation(s)
- Grazia Menna
- Institute of Neurosurgery, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, 00168 Rome, Italy; (G.M.); (P.P.M.); (C.M.D.); (A.O.)
| | - Pier Paolo Mattogno
- Institute of Neurosurgery, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, 00168 Rome, Italy; (G.M.); (P.P.M.); (C.M.D.); (A.O.)
| | - Carlo Maria Donzelli
- Institute of Neurosurgery, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, 00168 Rome, Italy; (G.M.); (P.P.M.); (C.M.D.); (A.O.)
| | - Lucia Lisi
- Institute of Pharmacology, Catholic University of Rome, 00168 Rome, Italy;
| | - Alessandro Olivi
- Institute of Neurosurgery, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, 00168 Rome, Italy; (G.M.); (P.P.M.); (C.M.D.); (A.O.)
| | - Giuseppe Maria Della Pepa
- Institute of Neurosurgery, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, 00168 Rome, Italy; (G.M.); (P.P.M.); (C.M.D.); (A.O.)
| |
Collapse
|
34
|
Tumor-Associated Macrophages in Gliomas—Basic Insights and Treatment Opportunities. Cancers (Basel) 2022; 14:cancers14051319. [PMID: 35267626 PMCID: PMC8909866 DOI: 10.3390/cancers14051319] [Citation(s) in RCA: 52] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2022] [Revised: 02/22/2022] [Accepted: 02/25/2022] [Indexed: 02/04/2023] Open
Abstract
Simple Summary Macrophages are a specialized immune cell type found in both invertebrates and vertebrates. Versatile in functionality, macrophages carry out important tasks such as cleaning cellular debris in healthy tissues and mounting immune responses during infection. In many cancer types, macrophages make up a significant portion of tumor tissue, and these are aptly called tumor-associated macrophages. In gliomas, a group of primary brain tumors, these macrophages are found in very high frequency. Tumor-associated macrophages can promote glioma development and influence the outcome of various therapeutic regimens. At the same time, these cells provide various potential points of intervention for therapeutic approaches in glioma patients. The significance of tumor-associated macrophages in the glioma microenvironment and potential therapeutic targets are the focus of this review. Abstract Glioma refers to a group of primary brain tumors which includes glioblastoma (GBM), astrocytoma and oligodendroglioma as major entities. Among these, GBM is the most frequent and most malignant one. The highly infiltrative nature of gliomas, and their intrinsic intra- and intertumoral heterogeneity, pose challenges towards developing effective treatments. The glioma microenvironment, in addition, is also thought to play a critical role during tumor development and treatment course. Unlike most other solid tumors, the glioma microenvironment is dominated by macrophages and microglia—collectively known as tumor-associated macrophages (TAMs). TAMs, like their homeostatic counterparts, are plastic in nature and can polarize to either pro-inflammatory or immunosuppressive states. Many lines of evidence suggest that immunosuppressive TAMs dominate the glioma microenvironment, which fosters tumor development, contributes to tumor aggressiveness and recurrence and, very importantly, impedes the therapeutic effect of various treatment regimens. However, through the development of new therapeutic strategies, TAMs can potentially be shifted towards a proinflammatory state which is of great therapeutic interest. In this review, we will discuss various aspects of TAMs in the context of glioma. The focus will be on the basic biology of TAMs in the central nervous system (CNS), potential biomarkers, critical evaluation of model systems for studying TAMs and finally, special attention will be given to the potential targeted therapeutic options that involve the TAM compartment in gliomas.
Collapse
|
35
|
Wu X, Wan Q, Wang J, Hou P, Zhang Q, Wang Q, Lu X. Epigenetic Activation of lncRNA MIR155HG Mediated by Promoter Hypomethylation and SP1 is Correlated with Immune Infiltration in Glioma. Onco Targets Ther 2022; 15:219-235. [PMID: 35299997 PMCID: PMC8922801 DOI: 10.2147/ott.s349078] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2021] [Accepted: 02/22/2022] [Indexed: 01/04/2023] Open
Affiliation(s)
- Xuechao Wu
- Department of Neurosurgery, The Affiliated Wuxi No.2 People’s Hospital of Nanjing Medical University, Wuxi, 214002, People’s Republic of China
- Department of Neurosurgery, The Affiliated Wuxi Clinical College of Nantong University, Wuxi, 214002, People’s Republic of China
| | - Quan Wan
- Department of Neurosurgery, The Affiliated Wuxi No.2 People’s Hospital of Nanjing Medical University, Wuxi, 214002, People’s Republic of China
| | - Jing Wang
- Department of Neurosurgery, The Affiliated Wuxi No.2 People’s Hospital of Nanjing Medical University, Wuxi, 214002, People’s Republic of China
| | - Peng Hou
- Department of Neurosurgery, Nantong Hospital of Traditional Chinese Medicine, Nantong, 226001, People’s Republic of China
| | - Qijian Zhang
- Department of Neurosurgery, The Affiliated Wuxi No.2 People’s Hospital of Nanjing Medical University, Wuxi, 214002, People’s Republic of China
| | - Qing Wang
- Department of Neurosurgery, The Affiliated Wuxi No.2 People’s Hospital of Nanjing Medical University, Wuxi, 214002, People’s Republic of China
- Department of Neurosurgery, The Affiliated Wuxi Clinical College of Nantong University, Wuxi, 214002, People’s Republic of China
| | - Xiaojie Lu
- Department of Neurosurgery, The Affiliated Wuxi No.2 People’s Hospital of Nanjing Medical University, Wuxi, 214002, People’s Republic of China
- Department of Neurosurgery, The Affiliated Hospital of Jiangnan University, Wuxi, 214000, People’s Republic of China
- Correspondence: Xiaojie Lu; Qing Wang, Department of Neurosurgery, The Affiliated Wuxi No.2 People’s Hospital of Nanjing Medical University, 68 Zhongshan Road, Wuxi, 214002, People’s Republic of China, Email ;
| |
Collapse
|
36
|
Challenges and Opportunities for Immunotherapeutic Intervention against Myeloid Immunosuppression in Glioblastoma. J Clin Med 2022; 11:jcm11041069. [PMID: 35207340 PMCID: PMC8880446 DOI: 10.3390/jcm11041069] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2022] [Revised: 02/05/2022] [Accepted: 02/10/2022] [Indexed: 12/25/2022] Open
Abstract
Glioblastoma multiforme (GBM), the most common and deadly brain cancer, exemplifies the paradigm that cancers grow with help from an immunosuppressive tumor microenvironment (TME). In general, TME includes a large contribution from various myeloid lineage-derived cell types, including (in the brain) altered pathogenic microglia as well as monocyte-macrophages (Macs), myeloid-derived suppressor cells (MDSC) and dendritic cell (DC) populations. Each can have protective roles, but has, by definition, been coopted by the tumor in patients with progressive disease. However, evidence demonstrates that myeloid immunosuppressive activities can be reversed in different ways, leading to enthusiasm for this therapeutic approach, both alone and in combination with potentially synergistic immunotherapeutic and other strategies. Here, we review the current understanding of myeloid cell immunosuppression of anti-tumor responses as well as potential targets, challenges, and developing means to reverse immunosuppression with various therapeutics and their status. Targets include myeloid cell colony stimulating factors (CSFs), insulin-like growth factor 1 (IGF1), several cytokines and chemokines, as well as CD40 activation and COX2 inhibition. Approaches in clinical development include antibodies, antisense RNA-based drugs, cell-based combinations, polarizing cytokines, and utilizing Macs as a platform for Chimeric Antigen Receptors (CAR)-based tumor targeting, like with CAR-T cells. To date, promising clinical results have been reported with several of these approaches.
Collapse
|
37
|
Bausart M, Préat V, Malfanti A. Immunotherapy for glioblastoma: the promise of combination strategies. J Exp Clin Cancer Res 2022; 41:35. [PMID: 35078492 PMCID: PMC8787896 DOI: 10.1186/s13046-022-02251-2] [Citation(s) in RCA: 109] [Impact Index Per Article: 36.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2021] [Accepted: 01/11/2022] [Indexed: 12/11/2022] Open
Abstract
Glioblastoma (GBM) treatment has remained almost unchanged for more than 20 years. The current standard of care involves surgical resection (if possible) followed by concomitant radiotherapy and chemotherapy. In recent years, immunotherapy strategies have revolutionized the treatment of many cancers, increasing the hope for GBM therapy. However, mostly due to the high, multifactorial immunosuppression occurring in the microenvironment, the poor knowledge of the neuroimmune system and the presence of the blood-brain barrier, the efficacy of immunotherapy in GBM is still low. Recently, new strategies for GBM treatments have employed immunotherapy combinations and have provided encouraging results in both preclinical and clinical studies. The lessons learned from clinical trials highlight the importance of tackling different arms of immunity. In this review, we aim to summarize the preclinical evidence regarding combination immunotherapy in terms of immune and survival benefits for GBM management. The outcomes of recent studies assessing the combination of different classes of immunotherapeutic agents (e.g., immune checkpoint blockade and vaccines) will be discussed. Finally, future strategies to ameliorate the efficacy of immunotherapy and facilitate clinical translation will be provided to address the unmet medical needs of GBM.
Collapse
Affiliation(s)
- Mathilde Bausart
- UCLouvain, Louvain Drug Research Institute, Advanced Drug Delivery and Biomaterials, Avenue Mounier 73 B1.73.12, 1200, Brussels, Belgium
| | - Véronique Préat
- UCLouvain, Louvain Drug Research Institute, Advanced Drug Delivery and Biomaterials, Avenue Mounier 73 B1.73.12, 1200, Brussels, Belgium.
| | - Alessio Malfanti
- UCLouvain, Louvain Drug Research Institute, Advanced Drug Delivery and Biomaterials, Avenue Mounier 73 B1.73.12, 1200, Brussels, Belgium
| |
Collapse
|
38
|
Abstract
BACKGROUND Glioblastoma stem cells (GSCs) and their interplay with tumor-associated macrophages (TAMs) are responsible for malignant growth and tumor recurrence of glioblastoma multiforme (GBM), but the underlying mechanisms are largely unknown. METHODS Cell viability, stemness, migration, and invasion were measured in GSCs after the knockdown of upstream stimulating factor 1 (USF1). Luciferase assay and chromatin immunoprecipitation qPCR were performed to determine the regulation of CD90 by USF1. Immunohistochemistry and immunofluorescent staining were used to examine the expression of USF1 and GSC markers, as well as the crosstalk between GSCs and TAMs. In addition, the interaction between GSCs and TAMs was confirmed using in vivo GBM models. RESULTS We show that USF1 promotes malignant glioblastoma phenotypes and GSCs-TAMs physical interaction by inducing CD90 expression. USF1 predicts a poor prognosis for glioma patients and is upregulated in patient-derived GSCs and glioblastoma cell lines. USF1 overexpression increases the proliferation, invasion, and neurosphere formation of GSCs and glioblastoma cell lines, while USF1 knockdown exerts an opposite effect. Further mechanistic studies reveal that USF1 promotes GSC stemness by directly regulating CD90 expression. Importantly, CD90 of GSCs functions as an anchor for physical interaction with macrophages. Additionally, the USF1/CD90 signaling axis supports the GSCs and TAMs adhesion and immunosuppressive feature of TAMs, which in turn enhance the stemness of GSCs. Moreover, the overexpression of CD90 restores the stemness property in USF1 knockdown GSCs and its immunosuppressive microenvironment. CONCLUSIONS Our findings indicate that the USF1/CD90 axis might be a potential therapeutic target for the treatment of glioblastoma.
Collapse
|
39
|
Lee AH, Sun L, Mochizuki AY, Reynoso JG, Orpilla J, Chow F, Kienzler JC, Everson RG, Nathanson DA, Bensinger SJ, Liau LM, Cloughesy T, Hugo W, Prins RM. Neoadjuvant PD-1 blockade induces T cell and cDC1 activation but fails to overcome the immunosuppressive tumor associated macrophages in recurrent glioblastoma. Nat Commun 2021; 12:6938. [PMID: 34836966 PMCID: PMC8626557 DOI: 10.1038/s41467-021-26940-2] [Citation(s) in RCA: 132] [Impact Index Per Article: 33.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2021] [Accepted: 10/25/2021] [Indexed: 12/20/2022] Open
Abstract
Primary brain tumors, such as glioblastoma (GBM), are remarkably resistant to immunotherapy, even though pre-clinical models suggest effectiveness. To understand this better in patients, here we take advantage of our recent neoadjuvant treatment paradigm to map the infiltrating immune cell landscape of GBM and how this is altered following PD-1 checkpoint blockade using high dimensional proteomics, single cell transcriptomics, and quantitative multiplex immunofluorescence. Neoadjuvant PD-1 blockade increases T cell infiltration and the proportion of a progenitor exhausted population of T cells found within the tumor. We identify an early activated and clonally expanded CD8+ T cell cluster whose TCR overlaps with a CD8+ PBMC population. Distinct changes are also observed in conventional type 1 dendritic cells that may facilitate T cell recruitment. Macrophages and monocytes still constitute the majority of infiltrating immune cells, even after anti-PD-1 therapy. Interferon-mediated changes in the myeloid population are consistently observed following PD-1 blockade; these also mediate an increase in chemotactic factors that recruit T cells. However, sustained high expression of T-cell-suppressive checkpoints in these myeloid cells continue to prevent the optimal activation of the tumor infiltrating T cells. Therefore, future immunotherapeutic strategies may need to incorporate the targeting of these cells for clinical benefit.
Collapse
Affiliation(s)
- Alexander H Lee
- Department of Neurosurgery, University of California, Los Angeles, Los Angeles, CA, 90095, USA
- Department of Molecular and Medical Pharmacology, University of California, Los Angeles, Los Angeles, CA, 90095, USA
| | - Lu Sun
- Department of Neurosurgery, University of California, Los Angeles, Los Angeles, CA, 90095, USA
| | - Aaron Y Mochizuki
- Department of Neurosurgery, University of California, Los Angeles, Los Angeles, CA, 90095, USA
| | - Jeremy G Reynoso
- Department of Neurosurgery, University of California, Los Angeles, Los Angeles, CA, 90095, USA
| | - Joey Orpilla
- Department of Neurosurgery, University of California, Los Angeles, Los Angeles, CA, 90095, USA
| | - Frances Chow
- Department of Neurology/Neuro-Oncology, University of California, Los Angeles, Los Angeles, CA, 90095, USA
| | - Jenny C Kienzler
- Department of Neurosurgery, University of California, Los Angeles, Los Angeles, CA, 90095, USA
| | - Richard G Everson
- Department of Neurosurgery, University of California, Los Angeles, Los Angeles, CA, 90095, USA
- UCLA Jonsson Comprehensive Cancer Center, University of California, Los Angeles, Los Angeles, CA, 90095, USA
| | - David A Nathanson
- Department of Molecular and Medical Pharmacology, University of California, Los Angeles, Los Angeles, CA, 90095, USA
- UCLA Jonsson Comprehensive Cancer Center, University of California, Los Angeles, Los Angeles, CA, 90095, USA
| | - Steven J Bensinger
- Department of Molecular and Medical Pharmacology, University of California, Los Angeles, Los Angeles, CA, 90095, USA
- UCLA Jonsson Comprehensive Cancer Center, University of California, Los Angeles, Los Angeles, CA, 90095, USA
- Department of Microbiology, Immunology, and Molecular Genetics, University of California, Los Angeles, Los Angeles, CA, 90095, USA
| | - Linda M Liau
- Department of Neurosurgery, University of California, Los Angeles, Los Angeles, CA, 90095, USA
- Department of Molecular and Medical Pharmacology, University of California, Los Angeles, Los Angeles, CA, 90095, USA
- UCLA Jonsson Comprehensive Cancer Center, University of California, Los Angeles, Los Angeles, CA, 90095, USA
| | - Timothy Cloughesy
- Department of Molecular and Medical Pharmacology, University of California, Los Angeles, Los Angeles, CA, 90095, USA
- Department of Neurology/Neuro-Oncology, University of California, Los Angeles, Los Angeles, CA, 90095, USA
- UCLA Jonsson Comprehensive Cancer Center, University of California, Los Angeles, Los Angeles, CA, 90095, USA
| | - Willy Hugo
- UCLA Jonsson Comprehensive Cancer Center, University of California, Los Angeles, Los Angeles, CA, 90095, USA.
- Parker Institute for Cancer Immunotherapy, 1 Letterman Drive, Suite D3500, San Francisco, CA, 94129, USA.
- Department of Medicine/Dermatology, University of California, Los Angeles, Los Angeles, CA, 90095, USA.
| | - Robert M Prins
- Department of Neurosurgery, University of California, Los Angeles, Los Angeles, CA, 90095, USA.
- Department of Molecular and Medical Pharmacology, University of California, Los Angeles, Los Angeles, CA, 90095, USA.
- UCLA Jonsson Comprehensive Cancer Center, University of California, Los Angeles, Los Angeles, CA, 90095, USA.
- Parker Institute for Cancer Immunotherapy, 1 Letterman Drive, Suite D3500, San Francisco, CA, 94129, USA.
| |
Collapse
|
40
|
Widodo SS, Dinevska M, Furst LM, Stylli SS, Mantamadiotis T. IL-10 in glioma. Br J Cancer 2021; 125:1466-1476. [PMID: 34349251 PMCID: PMC8609023 DOI: 10.1038/s41416-021-01515-6] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2021] [Revised: 07/05/2021] [Accepted: 07/22/2021] [Indexed: 02/07/2023] Open
Abstract
The prognosis for patients with glioblastoma (GBM), the most common and malignant type of primary brain tumour, is very poor, despite current standard treatments such as surgery, radiotherapy and chemotherapy. Moreover, the immunosuppressive tumour microenvironment hinders the development of effective immunotherapies for GBM. Cytokines such as interleukin-10 (IL-10) play a major role in modulating the activity of infiltrating immune cells and tumour cells in GBM, predominantly conferring an immunosuppressive action; however, in some circumstances, IL-10 can have an immunostimulatory effect. Elucidating the function of IL-10 in GBM is necessary to better strategise and improve the efficacy of immunotherapy. This review discusses the immunostimulatory and immunosuppressive roles of IL-10 in the GBM tumour microenvironment while considering IL-10-targeted treatment strategies. The molecular mechanisms that underlie the expression of IL-10 in various cell types are also outlined, and how this resulting information might provide an avenue for the improvement of immunotherapy in GBM is explored.
Collapse
Affiliation(s)
- Samuel S. Widodo
- grid.1008.90000 0001 2179 088XDepartment of Surgery, Royal Melbourne Hospital, The University of Melbourne, Parkville, VIC Australia
| | - Marija Dinevska
- grid.1008.90000 0001 2179 088XDepartment of Surgery, Royal Melbourne Hospital, The University of Melbourne, Parkville, VIC Australia
| | - Liam M. Furst
- grid.1008.90000 0001 2179 088XDepartment of Microbiology and Immunology, The University of Melbourne, Melbourne, VIC Australia
| | - Stanley S. Stylli
- grid.1008.90000 0001 2179 088XDepartment of Surgery, Royal Melbourne Hospital, The University of Melbourne, Parkville, VIC Australia ,grid.416153.40000 0004 0624 1200Department of Neurosurgery, Royal Melbourne Hospital, Parkville, VIC Australia
| | - Theo Mantamadiotis
- grid.1008.90000 0001 2179 088XDepartment of Surgery, Royal Melbourne Hospital, The University of Melbourne, Parkville, VIC Australia ,grid.1008.90000 0001 2179 088XDepartment of Microbiology and Immunology, The University of Melbourne, Melbourne, VIC Australia ,grid.418025.a0000 0004 0606 5526Florey Institute of Neuroscience and Mental Health, Parkville, VIC Australia
| |
Collapse
|
41
|
Himes BT, Geiger PA, Ayasoufi K, Bhargav AG, Brown DA, Parney IF. Immunosuppression in Glioblastoma: Current Understanding and Therapeutic Implications. Front Oncol 2021; 11:770561. [PMID: 34778089 PMCID: PMC8581618 DOI: 10.3389/fonc.2021.770561] [Citation(s) in RCA: 96] [Impact Index Per Article: 24.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2021] [Accepted: 09/24/2021] [Indexed: 12/12/2022] Open
Abstract
Glioblastoma (GBM) is the most common primary brain tumor in adults an carries and carries a terrible prognosis. The current regiment of surgical resection, radiation, and chemotherapy has remained largely unchanged in recent years as new therapeutic approaches have struggled to demonstrate benefit. One of the most challenging hurdles to overcome in developing novel treatments is the profound immune suppression found in many GBM patients. This limits the utility of all manner of immunotherapeutic agents, which have revolutionized the treatment of a number of cancers in recent years, but have failed to show similar benefit in GBM therapy. Understanding the mechanisms of tumor-mediated immune suppression in GBM is critical to the development of effective novel therapies, and reversal of this effect may prove key to effective immunotherapy for GBM. In this review, we discuss the current understanding of tumor-mediated immune suppression in GBM in both the local tumor microenvironment and systemically. We also discuss the effects of current GBM therapy on the immune system. We specifically explore some of the downstream effectors of tumor-driven immune suppression, particularly myeloid-derived suppressor cells (MDSCs) and other immunosuppressive monocytes, and the manner by which GBM induces their formation, with particular attention to the role of GBM-derived extracellular vesicles (EVs). Lastly, we briefly review the current state of immunotherapy for GBM and discuss additional hurdles to overcome identification and implementation of effective therapeutic strategies.
Collapse
Affiliation(s)
- Benjamin T Himes
- Department of Neurologic Surgery, Mayo Clinic, Rochester, MN, United States
| | - Philipp A Geiger
- Department of Neurosurgery, University Hospital Innsbruck, Tirol, Austria
| | | | - Adip G Bhargav
- Department of Neurosurgery, University of Kansas, Kansas City, KS, United States
| | - Desmond A Brown
- Surgical Neurology Branch, National Institutes of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, United States
| | - Ian F Parney
- Department of Neurologic Surgery, Mayo Clinic, Rochester, MN, United States.,Department of Immunology, Mayo Clinic, Rochester, MN, United States
| |
Collapse
|
42
|
Hatlen RR, Rajagopalan P. Environmental interplay: Stromal cells and biomaterial composition influence in the glioblastoma microenvironment. Acta Biomater 2021; 132:421-436. [PMID: 33276155 DOI: 10.1016/j.actbio.2020.11.044] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2020] [Revised: 11/24/2020] [Accepted: 11/25/2020] [Indexed: 12/12/2022]
Abstract
Glioblastoma multiforme (GBM) is the most deadly form of brain cancer. Recurrence is common, and established therapies have not been able to significantly extend overall patient survival. One platform through which GBM research can progress is to design biomimetic systems for discovery and investigation into the mechanisms of invasion, cellular properties, as well as the efficacy of therapies. In this review, 2D and 3D GBM in vitro cultures will be discussed. We focus on the effects of biomaterial properties, interactions between stromal cells, and vascular influence on cancer cell survival and progression. This review will summarize critical findings in each of these areas and how they have led to a more comprehensive scientific understanding of GBM. STATEMENT OF SIGNIFICANCE: Glioblastoma multiforme (GBM) is the most deadly form of brain cancer. Recurrence is common, and established therapies have not been able to significantly extend overall patient survival. One platform through which GBM research can progress is to design biomimetic systems for discovery and investigation into the mechanisms of invasion, cellular properties, as well as the efficacy of therapies. In this review, 2D and 3D GBM in vitro cultures will be discussed. We focus on the effects of biomaterial properties, interactions between stromal cells and vascular influence on cancer cell survival and progression. This review will summarize critical findings in each of these areas and how they have lead to a more comprehensive scientific understanding of GBM.
Collapse
Affiliation(s)
- Rosalyn R Hatlen
- Department of Chemical Engineering, Virginia Tech, Blacksburg, VA 24061, United States
| | | |
Collapse
|
43
|
Andersen RS, Anand A, Harwood DSL, Kristensen BW. Tumor-Associated Microglia and Macrophages in the Glioblastoma Microenvironment and Their Implications for Therapy. Cancers (Basel) 2021; 13:cancers13174255. [PMID: 34503065 PMCID: PMC8428223 DOI: 10.3390/cancers13174255] [Citation(s) in RCA: 65] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2021] [Revised: 08/17/2021] [Accepted: 08/18/2021] [Indexed: 12/23/2022] Open
Abstract
Glioblastoma is the most frequent and malignant primary brain tumor. Standard of care includes surgery followed by radiation and temozolomide chemotherapy. Despite treatment, patients have a poor prognosis with a median survival of less than 15 months. The poor prognosis is associated with an increased abundance of tumor-associated microglia and macrophages (TAMs), which are known to play a role in creating a pro-tumorigenic environment and aiding tumor progression. Most treatment strategies are directed against glioblastoma cells; however, accumulating evidence suggests targeting of TAMs as a promising therapeutic strategy. While TAMs are typically dichotomously classified as M1 and M2 phenotypes, recent studies utilizing single cell technologies have identified expression pattern differences, which is beginning to give a deeper understanding of the heterogeneous subpopulations of TAMs in glioblastomas. In this review, we evaluate the role of TAMs in the glioblastoma microenvironment and discuss how their interactions with cancer cells have an extensive impact on glioblastoma progression and treatment resistance. Finally, we summarize the effects and challenges of therapeutic strategies, which specifically aim to target TAMs.
Collapse
Affiliation(s)
- Rikke Sick Andersen
- Department of Pathology, Odense University Hospital, 5000 Odense, Denmark; (R.S.A.); (A.A.)
| | - Atul Anand
- Department of Pathology, Odense University Hospital, 5000 Odense, Denmark; (R.S.A.); (A.A.)
- Department of Clinical Research, University of Southern Denmark, 5000 Odense, Denmark
| | - Dylan Scott Lykke Harwood
- Department of Pathology, The Bartholin Institute, Rigshospitalet, Copenhagen University Hospital, 2100 Copenhagen, Denmark;
- Department of Clinical Medicine and Biotech Research and Innovation Center (BRIC), University of Copenhagen, 2200 Copenhagen, Denmark
| | - Bjarne Winther Kristensen
- Department of Pathology, Odense University Hospital, 5000 Odense, Denmark; (R.S.A.); (A.A.)
- Department of Clinical Research, University of Southern Denmark, 5000 Odense, Denmark
- Department of Pathology, The Bartholin Institute, Rigshospitalet, Copenhagen University Hospital, 2100 Copenhagen, Denmark;
- Department of Clinical Medicine and Biotech Research and Innovation Center (BRIC), University of Copenhagen, 2200 Copenhagen, Denmark
- Correspondence:
| |
Collapse
|
44
|
Basheer AS, Abas F, Othman I, Naidu R. Role of Inflammatory Mediators, Macrophages, and Neutrophils in Glioma Maintenance and Progression: Mechanistic Understanding and Potential Therapeutic Applications. Cancers (Basel) 2021; 13:4226. [PMID: 34439380 PMCID: PMC8393628 DOI: 10.3390/cancers13164226] [Citation(s) in RCA: 55] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2021] [Revised: 08/16/2021] [Accepted: 08/17/2021] [Indexed: 02/07/2023] Open
Abstract
Gliomas are the most common, highly malignant, and deadliest forms of brain tumors. These intra-cranial solid tumors are comprised of both cancerous and non-cancerous cells, which contribute to tumor development, progression, and resistance to the therapeutic regimen. A variety of soluble inflammatory mediators (e.g., cytokines, chemokines, and chemotactic factors) are secreted by these cells, which help in creating an inflammatory microenvironment and contribute to the various stages of cancer development, maintenance, and progression. The major tumor infiltrating immune cells of the tumor microenvironment include TAMs and TANs, which are either recruited peripherally or present as brain-resident macrophages (microglia) and support stroma for cancer cell expansion and invasion. These cells are highly plastic in nature and can be polarized into different phenotypes depending upon different types of stimuli. During neuroinflammation, glioma cells interact with TAMs and TANs, facilitating tumor cell proliferation, survival, and migration. Targeting inflammatory mediators along with the reprogramming of TAMs and TANs could be of great importance in glioma treatment and may delay disease progression. In addition, an inhibition of the key signaling pathways such as NF-κB, JAK/STAT, MAPK, PI3K/Akt/mTOR, and TLRs, which are activated during neuroinflammation and have an oncogenic role in glioblastoma (GBM), can exert more pronounced anti-glioma effects.
Collapse
Affiliation(s)
- Abdul Samad Basheer
- Jeffrey Cheah School of Medicine and Health Sciences, Monash University Malaysia, Jalan Lagoon Selatan, Bandar Sunway 47500, Malaysia; (A.S.B.); (I.O.)
| | - Faridah Abas
- Laboratory of Natural Products, Faculty of Science, University Putra Malaysia (UPM), Serdang 43400, Malaysia;
- Department of Food Science, Faculty of Food Science and Technology, University Putra Malaysia (UPM), Serdang 434000, Malaysia
| | - Iekhsan Othman
- Jeffrey Cheah School of Medicine and Health Sciences, Monash University Malaysia, Jalan Lagoon Selatan, Bandar Sunway 47500, Malaysia; (A.S.B.); (I.O.)
| | - Rakesh Naidu
- Jeffrey Cheah School of Medicine and Health Sciences, Monash University Malaysia, Jalan Lagoon Selatan, Bandar Sunway 47500, Malaysia; (A.S.B.); (I.O.)
| |
Collapse
|
45
|
Kučić N, Rački V, Šverko R, Vidović T, Grahovac I, Mršić-Pelčić J. Immunometabolic Modulatory Role of Naltrexone in BV-2 Microglia Cells. Int J Mol Sci 2021; 22:ijms22168429. [PMID: 34445130 PMCID: PMC8395119 DOI: 10.3390/ijms22168429] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2021] [Revised: 08/01/2021] [Accepted: 08/03/2021] [Indexed: 11/16/2022] Open
Abstract
Background: Naltrexone is an opioid receptor antagonist commonly used to treat opioid and alcohol dependence. The use of low dose naltrexone (LDN) was found to have anti-inflammatory properties for treatment of diseases such as fibromyalgia, Crohn’s disease, multiple sclerosis and regional pain syndromes. Related to its anti-neuroinflammatory properties, the mechanism of action is possibly mediated via Toll-like receptor 4 antagonism, which is widely expressed on microglial cells. The aim of the present study was to assess the immunometabolic effects of naltrexone on microglia cells in in vitro conditions. Methods: All experiments were performed in the BV-2 microglial cell line. The cells were treated with naltrexone at 100 μM concentrations corresponding to low dose for 24 h. Cell viability was assessed for every drug dose. To induce additional activation, the cells were pretreated with LPS and IFN-γ. Immunofluorescence was used to analyse the classical microglial activation markers iNOS and CD206, while Seahorse was used for real-time cellular metabolic assessments. mTOR activity measured over the expression of a major direct downstream target S6K was assessed using western blot. Results: LDN induced a shift from highly activated pro-inflammatory phenotype (iNOShighCD206low) to quiescent anti-inflammatory M2 phenotype (iNOSlowCD206high) in BV-2 microglia cells. Changes in the inflammatory profile were accompanied by cellular metabolic switching based on the transition from high glycolysis to mitochondrial oxidative phosphorylation (OXPHOS). LDN-treated cells were able to maintain a metabolically suppressive phenotype by supporting OXPHOS with high oxygen consumption, and also maintain a lower energetic state due to lower lactate production. The metabolic shift induced by transition from glycolysis to mitochondrial oxidative metabolism was more prominent in cells pretreated with immunometabolic modulators such as LPS and IFN-γ. In a dose-dependent manner, naltrexone also modulated mTOR/S6K expression, which underlies the cell metabolic phenotype regulating microglia immune properties and adaptation. Conclusion: By modulating the phenotypic features by metabolic switching of activated microglia, naltrexone was found to be an effective and powerful tool for immunometabolic reprogramming and could be a promising novel treatment for various neuroinflammatory conditions.
Collapse
Affiliation(s)
- Natalia Kučić
- Department of Physiology and Immunology, Faculty of Medicine, University of Rijeka, Braće Branchetta 20, 51000 Rijeka, Croatia
- Correspondence: ; Tel.: +385-51-651-192; Fax: +385-51-675-699
| | - Valentino Rački
- Department of Neurology, Clinical Hospital Center Rijeka, University of Rijeka, Krešimirova 42, 51000 Rijeka, Croatia;
| | - Roberta Šverko
- Emergency Department, Clinical Hospital Center Rijeka, University of Rijeka, Krešimirova 42, 51000 Rijeka, Croatia; (R.Š.); (T.V.)
| | - Toni Vidović
- Emergency Department, Clinical Hospital Center Rijeka, University of Rijeka, Krešimirova 42, 51000 Rijeka, Croatia; (R.Š.); (T.V.)
| | - Irena Grahovac
- Pharmacy Irena Grahovac, Trg I. Istarske brigade 5, 52100 Pula, Croatia;
| | - Jasenka Mršić-Pelčić
- Department of Pharmacology, Faculty of Medicine, University of Rijeka, Braće Branchetta 20, 51000 Rijeka, Croatia;
| |
Collapse
|
46
|
Raghavan JV, Ganesh RA, Sonpatki P, Naik D, John AE, Arunachalam P, Shah D, P S H, Lakshmikantha A, Pillai S, Chandrachari KP, Mariswamappa K, Lale S, Shah N, Jhunjhunwala S. Immuno-phenotyping of IDH-mutant grade 3 astrocytoma and IDH-wildtype glioblastoma reveals specific differences in cells of myeloid origin. Oncoimmunology 2021; 10:1957215. [PMID: 34377594 PMCID: PMC8331013 DOI: 10.1080/2162402x.2021.1957215] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Gliomas are heavily infiltrated with immune cells of myeloid origin. Past studies have shown that high-grade gliomas have a higher proportion of alternatively activated and suppressive myeloid cells when compared to low-grade gliomas, which correlate with poor prognosis. However, the differences in immune cell phenotypes within high-grade gliomas (between grade 3 and grade 4 or GBM) are relatively less explored, and a correlation of phenotypic characteristics between immune cells in the blood and high-grade tumors has not been performed. Additionally, myeloid cells of granulocytic origin present in gliomas remain poorly characterized. Herein, we address these questions through phenotypic characterizations of monocytes and neutrophils present in blood and tumors of individuals with glioblastoma (GBM, IDH-wild type) or grade 3 IDH-mutant gliomas. We observe that neutrophils are highly heterogeneous among individuals with glioma, and are different from healthy controls. We also show that CD163 expressing M2 monocytes are present in greater proportions in GBM tissue when compared to grade 3 IDH-mutant glioma tissue, and a larger proportion of granulocytic myeloid-derived suppressor cells are present in grade 3 IDH-mutant gliomas when compared to GBM. Finally, we demonstrate that the expression levels of CD86 and CD63 showed a high correlation between blood and tumor and suggest that these may be used as possible markers for prognosis.
Collapse
Affiliation(s)
- Jayashree V Raghavan
- Centre for BioSystems Science and Engineering, Indian Institute of Science, India
| | | | | | - Divya Naik
- Mazumdar Shaw Center for Translational Research, India
| | | | - Priyanka Arunachalam
- Centre for BioSystems Science and Engineering, Indian Institute of Science, India
| | - Darshat Shah
- Mazumdar Shaw Center for Translational Research, India
| | - Hari P S
- Mazumdar Shaw Center for Translational Research, India
| | | | | | | | | | | | - Nameeta Shah
- Mazumdar Shaw Center for Translational Research, India
| | | |
Collapse
|
47
|
Prosniak M, Kenyon LC, Hooper DC. Glioblastoma Contains Topologically Distinct Proliferative and Metabolically Defined Subpopulations of Nestin- and Glut1-Expressing Cells. J Neuropathol Exp Neurol 2021; 80:674-684. [PMID: 34297838 DOI: 10.1093/jnen/nlab044] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
The difficulty in treatment of glioblastoma is a consequence of its natural infiltrative growth and the existence of a population of therapy-resistant glioma cells that contribute to growth and recurrence. To identify cells more likely to have these properties, we examined the expression in tumor specimens of several protein markers important for glioma progression including the intermediate filament protein, Nestin (NES), a glucose transporter (Glut1/SLC2A1), the glial lineage marker, glial fibrillary acidic protein, and the proliferative indicator, Ki-67. We also examined the expression of von Willebrand factor, a marker for endothelial cells as well as the macrophage/myeloid markers CD163 and CD15. Using a multicolor immunofluorescence and hematoxylin and eosin staining approach with archival formalin-fixed, paraffin embedded tissue from primary, recurrent, and autopsy IDH1 wildtype specimens combined with high-resolution tissue image analysis, we have identified highly proliferative NES(+)/Glut1(-) cells that are preferentially perivascular. In contrast, Glut1(+)/NES(-) cells are distant from blood vessels, show low proliferation, and are preferentially located at the borders of pseudopalisading necrosis. We hypothesize that Glut1(+)/NES(-) cells would be naturally resistant to conventional chemotherapy and radiation due to their low proliferative capacity and may act as a reservoir for tumor recurrence.
Collapse
Affiliation(s)
| | - Lawrence C Kenyon
- Department of Pathology, Anatomy, and Cell Biology, Thomas Jefferson University, Pennsylvania, Philadelphia, USA
| | | |
Collapse
|
48
|
Kim AR, Choi KS, Kim MS, Kim KM, Kang H, Kim S, Chowdhury T, Yu HJ, Lee CE, Lee JH, Lee ST, Won JK, Kim JW, Kim YH, Kim TM, Park SH, Choi SH, Shin EC, Park CK. Absolute quantification of tumor-infiltrating immune cells in high-grade glioma identifies prognostic and radiomics values. Cancer Immunol Immunother 2021; 70:1995-2008. [PMID: 33416947 PMCID: PMC10991432 DOI: 10.1007/s00262-020-02836-w] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2020] [Accepted: 12/17/2020] [Indexed: 12/21/2022]
Abstract
PURPOSE To understand the tumor immune microenvironment precisely, it is important to secure the quantified data of tumor-infiltrating immune cells, since the immune cells are true working unit. We analyzed unit immune cell number per unit volume of core tumor tissue of high-grade gliomas (HGG) to correlate their immune microenvironment characteristics with clinical prognosis and radiomic signatures. METHODS The number of tumor-infiltrating immune cells from 64 HGG core tissue were analyzed using flow cytometry and standardized. After sorting out patient groups according to diverse immune characteristics, the groups were tested if they have any clinical prognostic relevance and specific radiomic signature relationships. Sparse partial least square with discriminant analysis using multimodal magnetic resonance images was employed for all radiomic classifications. RESULTS The median number of CD45 + cells per one gram of HGG core tissue counted 865,770 cells which was equivalent to 8.0% of total cells including tumor cells. There was heterogeneity in the distribution of immune cell subpopulations among patients. Overall survival was significantly better in T cell-deficient group than T cell-enriched group (p = 0.019), and T8 dominant group than T4 dominant group (p = 0.023). The number of tumor-associated macrophages (TAM) and M2-TAM was significantly decreased in isocitrate dehydrogenase mutated HGG. Radiomic signature classification showed good performance in predicting immune phenotypes especially with features extracted from apparent diffusion coefficient maps. CONCLUSIONS Absolute quantification of tumor-infiltrating immune cells confirmed the heterogeneity of immune microenvironment in HGG which harbors prognostic impact. This immune microenvironment could be predicted by radiomic signatures non-invasively.
Collapse
Affiliation(s)
- A Reum Kim
- Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology, KAIST, 291 Daehak-ro, Yuseong-gu, Daejeon, 34141, Korea
| | - Kyu Sung Choi
- Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology, KAIST, 291 Daehak-ro, Yuseong-gu, Daejeon, 34141, Korea
| | - Min-Sung Kim
- Department of Neurosurgery, Seoul National University Hospital, 101 Daehak-ro, Jongno-gu, Seoul, 03080, Korea
- Seoul National University College of Medicine, 101 Daehak-ro, Jongno-gu, Seoul, 03080, South Korea
| | - Kyung-Min Kim
- Department of Neurosurgery, Seoul National University Hospital, 101 Daehak-ro, Jongno-gu, Seoul, 03080, Korea
- Seoul National University College of Medicine, 101 Daehak-ro, Jongno-gu, Seoul, 03080, South Korea
| | - Ho Kang
- Department of Neurosurgery, Seoul National University Hospital, 101 Daehak-ro, Jongno-gu, Seoul, 03080, Korea
- Seoul National University College of Medicine, 101 Daehak-ro, Jongno-gu, Seoul, 03080, South Korea
| | - Sojin Kim
- Department of Neurosurgery, Seoul National University Hospital, 101 Daehak-ro, Jongno-gu, Seoul, 03080, Korea
- Seoul National University College of Medicine, 101 Daehak-ro, Jongno-gu, Seoul, 03080, South Korea
| | - Tamrin Chowdhury
- Department of Neurosurgery, Seoul National University Hospital, 101 Daehak-ro, Jongno-gu, Seoul, 03080, Korea
- Seoul National University College of Medicine, 101 Daehak-ro, Jongno-gu, Seoul, 03080, South Korea
| | - Hyeon Jong Yu
- Department of Neurosurgery, Seoul National University Hospital, 101 Daehak-ro, Jongno-gu, Seoul, 03080, Korea
- Seoul National University College of Medicine, 101 Daehak-ro, Jongno-gu, Seoul, 03080, South Korea
| | - Chae Eun Lee
- Department of Neurosurgery, Seoul National University Hospital, 101 Daehak-ro, Jongno-gu, Seoul, 03080, Korea
- Seoul National University College of Medicine, 101 Daehak-ro, Jongno-gu, Seoul, 03080, South Korea
| | - Joo Ho Lee
- Seoul National University College of Medicine, 101 Daehak-ro, Jongno-gu, Seoul, 03080, South Korea
- Department of Radiation Oncology, Seoul National University Hospital, 101 Daehak-ro, Jongno-gu, Seoul, 03080, Korea
| | - Soon-Tae Lee
- Seoul National University College of Medicine, 101 Daehak-ro, Jongno-gu, Seoul, 03080, South Korea
- Department of Neurology, Seoul National University Hospital, 101 Daehak-ro, Jongno-gu, Seoul, 03080, Korea
| | - Jae Kyung Won
- Seoul National University College of Medicine, 101 Daehak-ro, Jongno-gu, Seoul, 03080, South Korea
- Department of Pathology, Seoul National University Hospital, 101 Daehak-ro, Jongno-gu, Seoul, 03080, Korea
| | - Jin Wook Kim
- Department of Neurosurgery, Seoul National University Hospital, 101 Daehak-ro, Jongno-gu, Seoul, 03080, Korea
- Seoul National University College of Medicine, 101 Daehak-ro, Jongno-gu, Seoul, 03080, South Korea
| | - Yong-Hwy Kim
- Department of Neurosurgery, Seoul National University Hospital, 101 Daehak-ro, Jongno-gu, Seoul, 03080, Korea
- Seoul National University College of Medicine, 101 Daehak-ro, Jongno-gu, Seoul, 03080, South Korea
| | - Tae Min Kim
- Seoul National University College of Medicine, 101 Daehak-ro, Jongno-gu, Seoul, 03080, South Korea
- Department of Internal Medicine, Seoul National University Hospital, 101 Daehak-ro, Jongno-gu, Seoul, 03080, Korea
| | - Sung-Hye Park
- Seoul National University College of Medicine, 101 Daehak-ro, Jongno-gu, Seoul, 03080, South Korea
- Department of Pathology, Seoul National University Hospital, 101 Daehak-ro, Jongno-gu, Seoul, 03080, Korea
| | - Seung Hong Choi
- Seoul National University College of Medicine, 101 Daehak-ro, Jongno-gu, Seoul, 03080, South Korea.
- Department of Radiology, Seoul National University Hospital, 101 Daehak-ro, Jongno-gu, Seoul, 03080, Korea.
| | - Eui-Cheol Shin
- Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology, KAIST, 291 Daehak-ro, Yuseong-gu, Daejeon, 34141, Korea.
| | - Chul-Kee Park
- Department of Neurosurgery, Seoul National University Hospital, 101 Daehak-ro, Jongno-gu, Seoul, 03080, Korea.
- Seoul National University College of Medicine, 101 Daehak-ro, Jongno-gu, Seoul, 03080, South Korea.
| |
Collapse
|
49
|
Takacs GP, Flores-Toro JA, Harrison JK. Modulation of the chemokine/chemokine receptor axis as a novel approach for glioma therapy. Pharmacol Ther 2021; 222:107790. [PMID: 33316289 PMCID: PMC8122077 DOI: 10.1016/j.pharmthera.2020.107790] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2020] [Accepted: 12/04/2020] [Indexed: 02/06/2023]
Abstract
Chemokines are a large subfamily of cytokines known for their ability to facilitate cell migration, most notably leukocytes, throughout the body. Chemokines are necessary for a functioning immune system in both health and disease and have received considerable attention for their roles in orchestrating temporal-spatial regulation of immune cell populations in cancer. Gliomas comprise a group of common central nervous system (CNS) primary tumors that are extremely challenging to treat. Immunotherapy approaches for highly malignant brain tumors offer an exciting new avenue for therapeutic intervention but so far, have seen limited successful clinical outcomes. Herein we focus on important chemokine/chemokine receptor systems in the regulation of pro- and anti-tumor mechanisms, highlighting potential therapeutic advantages of modulating these systems in malignant gliomas and other cancers.
Collapse
Affiliation(s)
- Gregory P Takacs
- Department of Pharmacology & Therapeutics, College of Medicine, University of Florida, Gainesville, FL 32610, USA
| | - Joseph A Flores-Toro
- Department of Pharmacology & Therapeutics, College of Medicine, University of Florida, Gainesville, FL 32610, USA
| | - Jeffrey K Harrison
- Department of Pharmacology & Therapeutics, College of Medicine, University of Florida, Gainesville, FL 32610, USA.
| |
Collapse
|
50
|
Lyu Y, Yang H, Chen L. Metabolic regulation on the immune environment of glioma through gut microbiota. Semin Cancer Biol 2021; 86:990-997. [PMID: 33971263 DOI: 10.1016/j.semcancer.2021.05.005] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2021] [Revised: 04/08/2021] [Accepted: 05/04/2021] [Indexed: 12/12/2022]
Abstract
The gut-brain axis has paved our way in understanding varieties of disease. The gut microbiota especially the bacterial population plays critical roles in immune system development and function. Glioma comprises 80 percent of malignant brain cancer and glioblastoma (GBM) is the most malignant kind. GBM has a reputation for its suppressive immune environment and poor patient prognosis. Moreover, altered metabolites from gut microbiota affect both systemic immune and central nervous system (CNS) immunity. Here we will focus on the crosstalk between gut microbiota and GBM, and further explore how this communication contributes to glioma initiation and development. Finally, we highlight the latest insights on the metabolic regulation of immunity through gut microbiota, which provides a promising therapeutic strategy for GBM.
Collapse
Affiliation(s)
- Yingying Lyu
- Department of Neurosurgery, Huashan Hospital, Fudan University, Shanghai, PR China
| | - Hui Yang
- Department of Neurosurgery, Huashan Hospital, Fudan University, Shanghai, PR China; Institute for Translational Brain Research, Shanghai Medical College, Fudan University, Shanghai, PR China; Shanghai Key Laboratory of Brain Function Restoration and Neural Regeneration, Shanghai Clinical Medical Center of Neurosurgery, MOE Frontiers Center for Brain Science, Shanghai Medical College, Fudan University, Shanghai, PR China.
| | - Liang Chen
- Department of Neurosurgery, Huashan Hospital, Fudan University, Shanghai, PR China; Shanghai Key Laboratory of Brain Function Restoration and Neural Regeneration, Shanghai Clinical Medical Center of Neurosurgery, MOE Frontiers Center for Brain Science, Shanghai Medical College, Fudan University, Shanghai, PR China.
| |
Collapse
|